1
|
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 2024:1-7. [PMID: 39508179 DOI: 10.1080/00365513.2024.2422404] [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: 03/12/2024] [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
|
2
|
Tian Y, Shao S, Feng H, Zeng R, Li S, Zhang Q. Targeting senescent cells in atherosclerosis: Pathways to novel therapies. Ageing Res Rev 2024; 101:102502. [PMID: 39278272 DOI: 10.1016/j.arr.2024.102502] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2024] [Revised: 08/16/2024] [Accepted: 09/08/2024] [Indexed: 09/18/2024]
Abstract
Targeting senescent cells has recently emerged as a promising strategy for treating age-related diseases, such as atherosclerosis, which significantly contributes to global cardiovascular morbidity and mortality. This review elucidates the role of senescent cells in the development of atherosclerosis, including persistently damaging DNA, inducing oxidative stress and secreting pro-inflammatory factors known as the senescence-associated secretory phenotype. Therapeutic approaches targeting senescent cells to mitigate atherosclerosis are summarized in this review, which include the development of senotherapeutics and immunotherapies. These therapies are designed to either remove these cells or suppress their deleterious effects. These emerging therapies hold potential to decelerate or even alleviate the progression of AS, paving the way for new avenues in cardiovascular research and treatment.
Collapse
Affiliation(s)
- Yuhan Tian
- College of Pharmacy, Key Laboratory of Research and Application of Ethnic Medicine Processing and Preparation on the Qinghai-Tibet Plateau, Southwest Minzu University, Chengdu 610041, China
| | - Sihang Shao
- School of Electrical and Electronic Engineering, Nanyang Technological University, Singapore
| | - Haibo Feng
- College of Animal & Veterinary Sciences, Southwest Minzu University, Chengdu 610041, China
| | - Rui Zeng
- College of Pharmacy, Key Laboratory of Research and Application of Ethnic Medicine Processing and Preparation on the Qinghai-Tibet Plateau, Southwest Minzu University, Chengdu 610041, China
| | - Shanshan Li
- College of Pharmacy, Key Laboratory of Research and Application of Ethnic Medicine Processing and Preparation on the Qinghai-Tibet Plateau, Southwest Minzu University, Chengdu 610041, China.
| | - Qixiong Zhang
- Department of Pharmacy, Personalized Drug Therapy Key Laboratory of Sichuan Province, Sichuan Academy of Medical Science & Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu 610072, China; Department of Pharmacy, Sichuan Provincial People's Hospital East Sichuan Hospital & Dazhou First People's Hospital, Dazhou 635000, China.
| |
Collapse
|
3
|
Hamada M, Varkoly KS, Riyadh O, Beladi R, Munuswamy-Ramanujam G, Rawls A, Wilson-Rawls J, Chen H, McFadden G, Lucas AR. Urokinase-Type Plasminogen Activator Receptor (uPAR) in Inflammation and Disease: A Unique Inflammatory Pathway Activator. Biomedicines 2024; 12:1167. [PMID: 38927374 PMCID: PMC11201033 DOI: 10.3390/biomedicines12061167] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Revised: 04/24/2024] [Accepted: 05/10/2024] [Indexed: 06/28/2024] Open
Abstract
The urokinase-type plasminogen activator receptor (uPAR) is a unique protease binding receptor, now recognized as a key regulator of inflammation. Initially, uPA/uPAR was considered thrombolytic (clot-dissolving); however, recent studies have demonstrated its predominant immunomodulatory functions in inflammation and cancer. The uPA/uPAR complex has a multifaceted central role in both normal physiological and also pathological responses. uPAR is expressed as a glycophosphatidylinositol (GPI)-linked receptor interacting with vitronectin, integrins, G protein-coupled receptors, and growth factor receptors within a large lipid raft. Through protein-to-protein interactions, cell surface uPAR modulates intracellular signaling, altering cellular adhesion and migration. The uPA/uPAR also modifies extracellular activity, activating plasminogen to form plasmin, which breaks down fibrin, dissolving clots and activating matrix metalloproteinases that lyse connective tissue, allowing immune and cancer cell invasion and releasing growth factors. uPAR is now recognized as a biomarker for inflammatory diseases and cancer; uPAR and soluble uPAR fragments (suPAR) are increased in viral sepsis (COVID-19), inflammatory bowel disease, and metastasis. Here, we provide a comprehensive overview of the structure, function, and current studies examining uPAR and suPAR as diagnostic markers and therapeutic targets. Understanding uPAR is central to developing diagnostic markers and the ongoing development of antibody, small-molecule, nanogel, and virus-derived immune-modulating treatments that target uPAR.
Collapse
Affiliation(s)
- Mostafa Hamada
- College of Medicine, Kansas City University, 1750 Independence Ave, Kansas City, MO 64106, USA; (M.H.); (O.R.)
| | - Kyle Steven Varkoly
- Department of Internal Medicine, McLaren Macomb Hospital, Michigan State University College of Human Medicine, 1000 Harrington St., Mt Clemens, MI 48043, USA
| | - Omer Riyadh
- College of Medicine, Kansas City University, 1750 Independence Ave, Kansas City, MO 64106, USA; (M.H.); (O.R.)
| | - Roxana Beladi
- Department of Neurosurgery, Ascension Providence Hospital, Michigan State University College of Human Medicine, 16001 W Nine Mile Rd, Southfield, MI 48075, USA;
| | - Ganesh Munuswamy-Ramanujam
- Molecular Biology and Immunobiology Division, Interdisciplinary Institute of Indian System of Medicine, SRM Institute of Science and Technology, Kattankulathur 603203, India;
| | - Alan Rawls
- School of Life Sciences, Arizona State University, 427 E Tyler Mall, Tempe, AZ 85281, USA; (A.R.); (J.W.-R.)
| | - Jeanne Wilson-Rawls
- School of Life Sciences, Arizona State University, 427 E Tyler Mall, Tempe, AZ 85281, USA; (A.R.); (J.W.-R.)
| | - Hao Chen
- Department of Tumor Center, Lanzhou University Second Hospital, Lanzhou 730030, China;
| | - Grant McFadden
- Center for Personalized Diagnostics, Biodesign Institute, Arizona State University, 727 E Tyler St., Tempe, AZ 85287, USA;
| | - Alexandra R. Lucas
- Center for Personalized Diagnostics, Biodesign Institute, Arizona State University, 727 E Tyler St., Tempe, AZ 85287, USA;
| |
Collapse
|
4
|
Kostopoulou E, Kalavrizioti D, Davoulou P, Sinopidis X, Papachristou E, Goumenos DS, Dimitriou G, Spiliotis BE, Papasotiriou M. Soluble urokinase plasminogen activator receptor (suPAR) in children with obesity or type 1 diabetes as a marker of endothelial dysfunction: a cross-sectional study. Eur J Pediatr 2024; 183:2383-2389. [PMID: 38448612 DOI: 10.1007/s00431-024-05496-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/26/2023] [Revised: 02/20/2024] [Accepted: 02/27/2024] [Indexed: 03/08/2024]
Abstract
Pediatric obesity and type 1 diabetes mellitus (T1DM) represent two common chronic diseases associated with chronic inflammation, endothelial dysfunction and long-term complications. The aim of the present study was to assess the possible diagnostic and prognostic value of soluble urokinase plasminogen activator receptor (suPAR), a marker of inflammation and impaired endothelial function, in children with the diseases. In this cross-sectional study, children and adolescents with T1DM (N = 41) or obesity (N = 37), aged < 18 years old, and without proteinuria were included, together with children of similar age and without evident morbidity that served as controls (N = 42). Serum samples were obtained during standard outpatient follow up and the urokinase-type plasminogen activator receptor (suPAR) concentrations were measured using a commercially available sandwich ELISA kit (DUP00, R&D systems). Clinical and biochemical indices that were also assessed include body mass index (BMI) z-score, Tanner stages, glycosylated haemoglobin (HbA1c), fasting lipid profile and serum creatinine. Mean serum suPAR levels were significantly higher in patients with obesity compared to patients with T1DM and controls, while children with T1DM had similar suPAR levels to controls. Also, serum suPAR levels showed a negative correlation with age (Spearman rho -0.359, p < 0.001) and serum creatinine levels (Spearman rho -0.334, p = 0.005), and a positive correlation with BMI z-score (Spearman rho 0.354, p = 0.009) in the whole cohort. Conclusion: Serum suPAR may be a useful predictive marker of inflammation or endothelial dysfunction for children with obesity and T1DM, as well as a promising therapeutic target. Further studies are needed in order to clarify whether the reported differences in suPAR levels could reflect a greater impairment of the inflammation status and endothelial function in children with obesity compared to children with T1DM. What is Known: • Paediatric obesity and type 1 diabetes are characterised by chronic inflammation and metabolic dysregulation. • Urokinase plasminogen activator receptor (uPAR) has been proposed as a useful biomarker for chronic inflammation and cardiovascular risk in adults. What is New: • Serum suPAR levels were increased in children and adolescents with obesity compared to those with T1DM and healthy controls; thus, obesity may affect the inflammatory status and endothelial function to a higher degree than T1DM during childhood. • Serum suPAR may serve as a diagnostic and predictive marker of inflammation and endothelial dysfunction for children and adolescents with obesity and T1DM.
Collapse
Affiliation(s)
| | - Dimitra Kalavrizioti
- Department of Nephrology and Kidney Transplantation, University Hospital of Patras, Patras, 26504, Greece
| | - Panagiota Davoulou
- Department of Nephrology and Kidney Transplantation, University Hospital of Patras, Patras, 26504, Greece
| | | | - Evangelos Papachristou
- Department of Nephrology and Kidney Transplantation, University Hospital of Patras, Patras, 26504, Greece
| | - Dimitrios S Goumenos
- Department of Nephrology and Kidney Transplantation, University Hospital of Patras, Patras, 26504, Greece
| | - Gabriel Dimitriou
- Department of Pediatrics, University Hospital of Patras, Patras, Greece
| | | | - Marios Papasotiriou
- Department of Nephrology and Kidney Transplantation, University Hospital of Patras, Patras, 26504, Greece.
| |
Collapse
|
5
|
Montecillo J, Pirker T, Pemberton C, Chew-Harris J. suPAR in cardiovascular disease. Adv Clin Chem 2024; 121:89-131. [PMID: 38797545 DOI: 10.1016/bs.acc.2024.04.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/29/2024]
Abstract
Soluble urokinase plasminogen activator receptor (suPAR), the soluble counterpart of urokinase plasminogen activator receptor, is found in the circulation at various levels. suPAR and its parent molecule, cell surface uPAR, exhibit similar structure and extracellular functional roles facilitating fibrinolysis, cellular adhesion, and migration. Studies have assessed the correlation between suPAR in cardiovascular disease (CVD). It is postulated that suPAR may serve as an indicator of inflammatory activation and burden during CVD progression. Increased suPAR independently predicts poorer outcomes in acute coronary syndromes, in heart failure, as well as in coronary artery disease and atherosclerosis. To guide translation into clinical utization, suPAR has been assessed in numerous CVD settings for improved risk discrimination independently or in association with established traditional risk factors. Whilst the involvement of suPAR has been explored in other diseases such as kidney diseases and cancer, there is only emerging evidence of suPAR's mechanistic involvement in cardiovascular disease. In this review, we provide a background into suPAR and its potential role as a biomarker in CVD.
Collapse
Affiliation(s)
- Jaya Montecillo
- Christchurch Heart Institute, University of Otago, Christchurch, New Zealand
| | - Thomas Pirker
- Christchurch Heart Institute, University of Otago, Christchurch, New Zealand
| | | | - Janice Chew-Harris
- Christchurch Heart Institute, University of Otago, Christchurch, New Zealand.
| |
Collapse
|
6
|
Miceli G, Basso MG, Pintus C, Pennacchio AR, Cocciola E, Cuffaro M, Profita M, Rizzo G, Tuttolomondo A. Molecular Pathways of Vulnerable Carotid Plaques at Risk of Ischemic Stroke: A Narrative Review. Int J Mol Sci 2024; 25:4351. [PMID: 38673936 PMCID: PMC11050267 DOI: 10.3390/ijms25084351] [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: 02/26/2024] [Revised: 04/05/2024] [Accepted: 04/12/2024] [Indexed: 04/28/2024] Open
Abstract
The concept of vulnerable carotid plaques is pivotal in understanding the pathophysiology of ischemic stroke secondary to large-artery atherosclerosis. In macroscopic evaluation, vulnerable plaques are characterized by one or more of the following features: microcalcification; neovascularization; lipid-rich necrotic cores (LRNCs); intraplaque hemorrhage (IPH); thin fibrous caps; plaque surface ulceration; huge dimensions, suggesting stenosis; and plaque rupture. Recognizing these macroscopic characteristics is crucial for estimating the risk of cerebrovascular events, also in the case of non-significant (less than 50%) stenosis. Inflammatory biomarkers, such as cytokines and adhesion molecules, lipid-related markers like oxidized low-density lipoprotein (LDL), and proteolytic enzymes capable of degrading extracellular matrix components are among the key molecules that are scrutinized for their associative roles in plaque instability. Through their quantification and evaluation, these biomarkers reveal intricate molecular cross-talk governing plaque inflammation, rupture potential, and thrombogenicity. The current evidence demonstrates that plaque vulnerability phenotypes are multiple and heterogeneous and are associated with many highly complex molecular pathways that determine the activation of an immune-mediated cascade that culminates in thromboinflammation. This narrative review provides a comprehensive analysis of the current knowledge on molecular biomarkers expressed by symptomatic carotid plaques. It explores the association of these biomarkers with the structural and compositional attributes that characterize vulnerable plaques.
Collapse
Affiliation(s)
- Giuseppe Miceli
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties (ProMISE), University of Palermo, Piazza delle Cliniche 2, 90127 Palermo, Italy; (G.M.); (M.G.B.); (C.P.); (A.R.P.); (E.C.); (M.C.); (M.P.); (G.R.)
- Internal Medicine and Stroke Care Ward, University Hospital, Policlinico “P. Giaccone”, 90127 Palermo, Italy
| | - Maria Grazia Basso
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties (ProMISE), University of Palermo, Piazza delle Cliniche 2, 90127 Palermo, Italy; (G.M.); (M.G.B.); (C.P.); (A.R.P.); (E.C.); (M.C.); (M.P.); (G.R.)
- Internal Medicine and Stroke Care Ward, University Hospital, Policlinico “P. Giaccone”, 90127 Palermo, Italy
| | - Chiara Pintus
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties (ProMISE), University of Palermo, Piazza delle Cliniche 2, 90127 Palermo, Italy; (G.M.); (M.G.B.); (C.P.); (A.R.P.); (E.C.); (M.C.); (M.P.); (G.R.)
- Internal Medicine and Stroke Care Ward, University Hospital, Policlinico “P. Giaccone”, 90127 Palermo, Italy
| | - Andrea Roberta Pennacchio
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties (ProMISE), University of Palermo, Piazza delle Cliniche 2, 90127 Palermo, Italy; (G.M.); (M.G.B.); (C.P.); (A.R.P.); (E.C.); (M.C.); (M.P.); (G.R.)
- Internal Medicine and Stroke Care Ward, University Hospital, Policlinico “P. Giaccone”, 90127 Palermo, Italy
| | - Elena Cocciola
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties (ProMISE), University of Palermo, Piazza delle Cliniche 2, 90127 Palermo, Italy; (G.M.); (M.G.B.); (C.P.); (A.R.P.); (E.C.); (M.C.); (M.P.); (G.R.)
- Internal Medicine and Stroke Care Ward, University Hospital, Policlinico “P. Giaccone”, 90127 Palermo, Italy
| | - Mariagiovanna Cuffaro
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties (ProMISE), University of Palermo, Piazza delle Cliniche 2, 90127 Palermo, Italy; (G.M.); (M.G.B.); (C.P.); (A.R.P.); (E.C.); (M.C.); (M.P.); (G.R.)
- Internal Medicine and Stroke Care Ward, University Hospital, Policlinico “P. Giaccone”, 90127 Palermo, Italy
| | - Martina Profita
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties (ProMISE), University of Palermo, Piazza delle Cliniche 2, 90127 Palermo, Italy; (G.M.); (M.G.B.); (C.P.); (A.R.P.); (E.C.); (M.C.); (M.P.); (G.R.)
- Internal Medicine and Stroke Care Ward, University Hospital, Policlinico “P. Giaccone”, 90127 Palermo, Italy
| | - Giuliana Rizzo
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties (ProMISE), University of Palermo, Piazza delle Cliniche 2, 90127 Palermo, Italy; (G.M.); (M.G.B.); (C.P.); (A.R.P.); (E.C.); (M.C.); (M.P.); (G.R.)
- Internal Medicine and Stroke Care Ward, University Hospital, Policlinico “P. Giaccone”, 90127 Palermo, Italy
| | - Antonino Tuttolomondo
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties (ProMISE), University of Palermo, Piazza delle Cliniche 2, 90127 Palermo, Italy; (G.M.); (M.G.B.); (C.P.); (A.R.P.); (E.C.); (M.C.); (M.P.); (G.R.)
- Internal Medicine and Stroke Care Ward, University Hospital, Policlinico “P. Giaccone”, 90127 Palermo, Italy
| |
Collapse
|
7
|
Ali I, Zhang H, Zaidi SAA, Zhou G. Understanding the intricacies of cellular senescence in atherosclerosis: Mechanisms and therapeutic implications. Ageing Res Rev 2024; 96:102273. [PMID: 38492810 DOI: 10.1016/j.arr.2024.102273] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2023] [Revised: 01/16/2024] [Accepted: 03/13/2024] [Indexed: 03/18/2024]
Abstract
Cardiovascular disease is currently the largest cause of mortality and disability globally, surpassing communicable diseases, and atherosclerosis is the main contributor to this epidemic. Aging is intimately linked to atherosclerosis development and progression, however, the mechanism of aging in atherosclerosis is not well known. To emphasize the significant research on the involvement of senescent cells in atherosclerosis, we begin by outlining compelling evidence that indicates various types of senescent cells and SASP factors linked to atherosclerotic phenotypes. We subsequently provide a comprehensive summary of the existing knowledge, shedding light on the intricate mechanisms through which cellular senescence contributes to the pathogenesis of atherosclerosis. Further, we cover that senescence can be identified by both structural changes and several senescence-associated biomarkers. Finally, we discuss that preventing accelerated cellular senescence represents an important therapeutic potential, as permanent changes may occur in advanced atherosclerosis. Together, the review summarizes the relationship between cellular senescence and atherosclerosis, and inspects the molecular knowledge, and potential clinical significance of senescent cells in developing senescent-based therapy, thus providing crucial insights into their biology and potential therapeutic exploration.
Collapse
Affiliation(s)
- Ilyas Ali
- Department of Medical Cell Biology and Genetics, Guangdong Key Laboratory of Genomic Stability and Disease Prevention, Shenzhen Key Laboratory of Anti-Aging and Regenerative Medicine, and Shenzhen Engineering Laboratory of Regenerative Technologies for Orthopaedic Diseases, Health Sciences Center, Shenzhen University, Shenzhen 518060, PR China; Guangdong Key Laboratory for Biomedical Measurements and Ultrasound Imaging, National-Regional Key Technology Engineering Laboratory for Medical Ultrasound, School of Biomedical Engineering, Shenzhen University Medical School, Shenzhen 518060, PR China
| | - Hongliang Zhang
- Shenzhen University General Hospital, Shenzhen University, Shenzhen 518060, PR China
| | - Syed Aqib Ali Zaidi
- Department of Medical Cell Biology and Genetics, Guangdong Key Laboratory of Genomic Stability and Disease Prevention, Shenzhen Key Laboratory of Anti-Aging and Regenerative Medicine, and Shenzhen Engineering Laboratory of Regenerative Technologies for Orthopaedic Diseases, Health Sciences Center, Shenzhen University, Shenzhen 518060, PR China
| | - Guangqian Zhou
- Department of Medical Cell Biology and Genetics, Guangdong Key Laboratory of Genomic Stability and Disease Prevention, Shenzhen Key Laboratory of Anti-Aging and Regenerative Medicine, and Shenzhen Engineering Laboratory of Regenerative Technologies for Orthopaedic Diseases, Health Sciences Center, Shenzhen University, Shenzhen 518060, PR China; Guangdong Key Laboratory for Biomedical Measurements and Ultrasound Imaging, National-Regional Key Technology Engineering Laboratory for Medical Ultrasound, School of Biomedical Engineering, Shenzhen University Medical School, Shenzhen 518060, PR China.
| |
Collapse
|
8
|
Guduguntla BA, Vasbinder A, Anderson E, Azam TU, Blakely P, Webster NJ, Gonzalez R, Atonucci T, Heidebrink JL, Giordani B, Zahodne L, Hampstead BM, Ajrouch KJ, Hayek SS. Biomarkers of chronic inflammation and cognitive decline: A prospective observational study. ALZHEIMER'S & DEMENTIA (AMSTERDAM, NETHERLANDS) 2024; 16:e12568. [PMID: 38532827 PMCID: PMC10964918 DOI: 10.1002/dad2.12568] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/20/2023] [Revised: 02/09/2024] [Accepted: 02/14/2024] [Indexed: 03/28/2024]
Abstract
We sought to determine whether the biomarkers of chronic inflammation predict cognitive decline in a prospective observational study. We measured baseline serum soluble urokinase plasminogen activator receptor (suPAR) and high sensitivity C-reactive protein (hs-CRP) levels in 282 participants of the University of Michigan Memory and Aging Project. Cognitive function was measured using the Montreal Cognitive Assessment (MoCA) and the Clinical Dementia Rating (CDR) scale for up to five time points. SuPAR and hs-CRP levels were not significantly higher in participants with mild cognitive impairment (n = 97) or dementia (n = 59), compared to those with normal cognitive function (n = 126). Overall, 14% of participants experienced significant cognitive decline over the study period. The change in MoCA or CDR scores over time did not differ significantly according to baseline suPAR or hs-CRP levels. Chronic systemic inflammation, as measured by serum suPAR or hs-CRP levels, is unlikely to contribute significantly to cognitive decline.
Collapse
Affiliation(s)
| | - Alexi Vasbinder
- Division of CardiologyDepartment of Internal MedicineUniversity of MichiganAnn ArborMichiganUSA
| | - Elizabeth Anderson
- Division of CardiologyDepartment of Internal MedicineUniversity of MichiganAnn ArborMichiganUSA
| | - Tariq U. Azam
- Division of CardiologyDepartment of Internal MedicineUniversity of MichiganAnn ArborMichiganUSA
| | - Pennelope Blakely
- Division of CardiologyDepartment of Internal MedicineUniversity of MichiganAnn ArborMichiganUSA
| | - Noah J. Webster
- Institute for Social ResearchUniversity of MichiganAnn ArborMichiganUSA
| | - Richard Gonzalez
- Department of PsychologyUniversity of MichiganAnn ArborMichiganUSA
| | - Toni Atonucci
- Department of PsychologyUniversity of MichiganAnn ArborMichiganUSA
| | | | - Bruno Giordani
- Department of PsychologyUniversity of MichiganAnn ArborMichiganUSA
- Department of NeurologyUniversity of MichiganAnn ArborMichiganUSA
- Department of PsychiatryUniversity of MichiganAnn ArborMichiganUSA
| | - Laura Zahodne
- Department of PsychologyUniversity of MichiganAnn ArborMichiganUSA
| | - Benjamin M. Hampstead
- Department of PsychiatryUniversity of MichiganAnn ArborMichiganUSA
- Mental Health Service Line, Veterans Affairs Ann Arbor Healthcare SystemAnn ArborMichiganUSA
| | - Kristine J. Ajrouch
- Institute for Social ResearchUniversity of MichiganAnn ArborMichiganUSA
- Eastern Michigan UniversityYpsilantiMichiganUSA
| | - Salim S. Hayek
- Division of CardiologyDepartment of Internal MedicineUniversity of MichiganAnn ArborMichiganUSA
| |
Collapse
|
9
|
Sommerer C, Müller-Krebs S, Nadal J, Schultheiss UT, Friedrich N, Nauck M, Schmid M, Nußhag C, Reiser J, Eckardt KU, Zeier M, Hayek SS. Prospective Cohort Study of Soluble Urokinase Plasminogen Activation Receptor and Cardiovascular Events in Patients With CKD. Kidney Int Rep 2023; 8:2265-2275. [PMID: 38025216 PMCID: PMC10658273 DOI: 10.1016/j.ekir.2023.08.038] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2023] [Accepted: 08/28/2023] [Indexed: 12/01/2023] Open
Abstract
Introduction Soluble urokinase plasminogen activation receptor (suPAR) is an immune-derived pathogenic factor for kidney and atherosclerotic disease. Whether the association between suPAR and cardiovascular (CV) outcomes is dependent on the severity of underlying kidney disease is unclear. Methods We measured serum suPAR levels in 4994 participants (mean age 60 years; 60% men; 36% with diabetes mellitus; mean estimated glomerular filtration rate (eGFR) 49 ml/min per 1.73 m2, SD 18) of the German Chronic Kidney Disease (GCKD) cohort and examined its association with all-cause death, CV death, and major CV events (MACE) across the range of eGFR and urine albumin-to-creatinine ratio (UACR). Results The median suPAR level was 1771 pg/ml (interquartile range [IQR] 1447-2254 pg/ml). SuPAR levels were positively and independently correlated with age, eGFR, UACR, and parathyroid hormone levels. There were 573 deaths, including 190 CV deaths and 683 MACE events at a follow-up time of 6.5 years. In multivariable analyses, suPAR levels (log2) were associated with all-cause death (hazard ratio [HR] 1.36, 95% confidence interval [CI] 1.21-1.53), CV death (HR 1.27, 95% CI 1.03-1.57), and MACE (HR 1.13, 95% CI 1.00-1.28), and were not found to differ according to diabetes mellitus status, baseline eGFR, UACR, or parathyroid hormone levels. In mediation analysis, suPAR's direct effect on all-cause death, CV death, and MACE accounted for 77%, 67%, and 60% of the total effect, respectively; whereas the effect mediated through eGFR accounted for 23%, 34%, and 40%, respectively. Conclusion In a large cohort of individuals with chronic kidney disease (CKD), suPAR levels were associated with mortality and CV outcomes independently of indices of kidney function, consistent with its independent role in the pathogenesis of atherosclerosis.
Collapse
Affiliation(s)
- Claudia Sommerer
- Department of Nephrology, University Hospital Heidelberg, Renal Center, Heidelberg, Germany
| | - Sandra Müller-Krebs
- Department of Nephrology, University Hospital Heidelberg, Renal Center, Heidelberg, Germany
| | - Jennifer Nadal
- Department of Medical Biometry, Informatics, and Epidemiology (IMBIE), University Hospital Bonn, Bonn, Germany
| | - Ulla T. Schultheiss
- Institute of Genetic Epidemiology, Faculty of Medicine and Medical Center, University of Freiburg, Freiburg, Germany
- Department of Medicine IV, Nephrology and Primary Care, Faculty of Medicine and Medical Center - University of Freiburg, Freiburg, Germany
| | - Nele Friedrich
- Institute of Clinical Chemistry and Laboratory Medicine, University Medicine Greifswald, Greifswald, Germany
- DZHK (German Center for Cardiovascular Research), Partner Site Greifswald, University Medicine, Greifswald, Germany
| | - Matthias Nauck
- Institute of Clinical Chemistry and Laboratory Medicine, University Medicine Greifswald, Greifswald, Germany
- DZHK (German Center for Cardiovascular Research), Partner Site Greifswald, University Medicine, Greifswald, Germany
| | - Matthias Schmid
- Department of Medical Biometry, Informatics, and Epidemiology (IMBIE), University Hospital Bonn, Bonn, Germany
| | - Christian Nußhag
- Department of Nephrology, University Hospital Heidelberg, Renal Center, Heidelberg, Germany
| | - Jochen Reiser
- Department of Medicine, Rush University Medical Center, Chicago, Illinois, USA
| | - Kai-Uwe Eckardt
- Department of Nephrology and Medical Intensive Care, Charité, Universitätsmedizin Berlin, Berlin, Germany
- Department of Nephrology and Hypertension, Universitätsklinikum Erlangen, Friedrich-Alexander University Erlangen-Nürnberg, Erlangen, Germany
| | - Martin Zeier
- Department of Nephrology, University Hospital Heidelberg, Renal Center, Heidelberg, Germany
| | - Salim S. Hayek
- Department of Medicine, Division of Cardiology, University of Michigan, Michigan, USA
| |
Collapse
|
10
|
Leth JM, Newcombe EA, Grønnemose AL, Jørgensen JT, Qvist K, Clausen AS, Knudsen LBS, Kjaer A, Kragelund BB, Jørgensen TJD, Ploug M. Targeted imaging of uPAR expression in vivo with cyclic AE105 variants. Sci Rep 2023; 13:17248. [PMID: 37821532 PMCID: PMC10567728 DOI: 10.1038/s41598-023-43934-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2023] [Accepted: 09/30/2023] [Indexed: 10/13/2023] Open
Abstract
A comprehensive literature reports on the correlation between elevated levels of urokinase-type plasminogen activator receptor (uPAR) and the severity of diseases with chronic inflammation including solid cancers. Molecular imaging is widely used as a non-invasive method to locate disease dissemination via full body scans and to stratify patients for targeted treatment. To date, the only imaging probe targeting uPAR that has reached clinical phase-II testing relies on a high-affinity 9-mer peptide (AE105), and several studies by positron emission tomography (PET) scanning or near-infra red (NIR) fluorescence imaging have validated its utility and specificity in vivo. While our previous studies focused on applying various reporter groups, the current study aims to improve uPAR-targeting properties of AE105. We successfully stabilized the small uPAR-targeting core of AE105 by constraining its conformational landscape by disulfide-mediated cyclization. Importantly, this modification mitigated the penalty on uPAR-affinity typically observed after conjugation to macrocyclic chelators. Cyclization did not impair tumor targeting efficiency of AE105 in vivo as assessed by PET imaging and a trend towards increased tracer uptake was observed. In future studies, we predict that this knowledge will aid development of new fluorescent AE105 derivatives with a view to optical imaging of uPAR to assist precision guided cancer surgery.
Collapse
Affiliation(s)
- Julie Maja Leth
- Finsen Laboratory, Copenhagen University Hospital - Rigshospitalet, 2200, Copenhagen N, Denmark
- Biotech Research and Innovation Centre (BRIC), University of Copenhagen, 2200, Copenhagen N, Denmark
| | - Estella Anne Newcombe
- Structural Biology and NMR Laboratory, Copenhagen N, Denmark
- REPIN, Copenhagen N, Denmark
- The Linderstrøm Lang Centre for Protein Science, Department of Biology, University of Copenhagen, Ole Maaloes Vej 5, 2200, Copenhagen N, Denmark
| | - Anne Louise Grønnemose
- Finsen Laboratory, Copenhagen University Hospital - Rigshospitalet, 2200, Copenhagen N, Denmark
- Biotech Research and Innovation Centre (BRIC), University of Copenhagen, 2200, Copenhagen N, Denmark
- Department of Biochemistry and Molecular Biology, University of Southern Denmark, 5230, Odense M, Denmark
| | - Jesper Tranekjær Jørgensen
- Department of Clinical Physiology and Nuclear Medicine and Cluster for Molecular Imaging, Copenhagen University Hospital - Rigshospitalet, Copenhagen N, Denmark
- Department of Biomedical Sciences, University of Copenhagen, Copenhagen N, Denmark
| | - Katrine Qvist
- Department of Clinical Physiology and Nuclear Medicine and Cluster for Molecular Imaging, Copenhagen University Hospital - Rigshospitalet, Copenhagen N, Denmark
- Department of Biomedical Sciences, University of Copenhagen, Copenhagen N, Denmark
| | - Anne Skovsbo Clausen
- Department of Clinical Physiology and Nuclear Medicine and Cluster for Molecular Imaging, Copenhagen University Hospital - Rigshospitalet, Copenhagen N, Denmark
- Department of Biomedical Sciences, University of Copenhagen, Copenhagen N, Denmark
| | - Line Bruhn Schneider Knudsen
- Department of Clinical Physiology and Nuclear Medicine and Cluster for Molecular Imaging, Copenhagen University Hospital - Rigshospitalet, Copenhagen N, Denmark
- Department of Biomedical Sciences, University of Copenhagen, Copenhagen N, Denmark
| | - Andreas Kjaer
- Department of Clinical Physiology and Nuclear Medicine and Cluster for Molecular Imaging, Copenhagen University Hospital - Rigshospitalet, Copenhagen N, Denmark
- Department of Biomedical Sciences, University of Copenhagen, Copenhagen N, Denmark
| | - Birthe Brandt Kragelund
- Structural Biology and NMR Laboratory, Copenhagen N, Denmark
- REPIN, Copenhagen N, Denmark
- The Linderstrøm Lang Centre for Protein Science, Department of Biology, University of Copenhagen, Ole Maaloes Vej 5, 2200, Copenhagen N, Denmark
| | | | - Michael Ploug
- Finsen Laboratory, Copenhagen University Hospital - Rigshospitalet, 2200, Copenhagen N, Denmark.
- Biotech Research and Innovation Centre (BRIC), University of Copenhagen, 2200, Copenhagen N, Denmark.
| |
Collapse
|
11
|
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
|
12
|
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
|
13
|
Hindy G, Tyrrell DJ, Vasbinder A, Wei C, Presswalla F, Wang H, Blakely P, Ozel AB, Graham S, Holton GH, Dowsett J, Fahed AC, Amadi KM, Erne GK, Tekmulla A, Ismail A, Launius C, Sotoodehnia N, Pankow JS, Thørner LW, Erikstrup C, Pedersen OB, Banasik K, Brunak S, Ullum H, Eugen-Olsen J, Ostrowski SR, Haas ME, Nielsen JB, Lotta LA, Engström G, Melander O, Orho-Melander M, Zhao L, Murthy VL, Pinsky DJ, Willer CJ, Heckbert SR, Reiser J, Goldstein DR, Desch KC, Hayek SS. Increased soluble urokinase plasminogen activator levels modulate monocyte function to promote atherosclerosis. J Clin Invest 2022; 132:e158788. [PMID: 36194491 PMCID: PMC9754000 DOI: 10.1172/jci158788] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2022] [Accepted: 09/29/2022] [Indexed: 01/26/2023] Open
Abstract
People with kidney disease are disproportionately affected by atherosclerosis for unclear reasons. Soluble urokinase plasminogen activator receptor (suPAR) is an immune-derived mediator of kidney disease, levels of which are strongly associated with cardiovascular outcomes. We assessed suPAR's pathogenic involvement in atherosclerosis using epidemiologic, genetic, and experimental approaches. We found serum suPAR levels to be predictive of coronary artery calcification and cardiovascular events in 5,406 participants without known coronary disease. In a genome-wide association meta-analysis including over 25,000 individuals, we identified a missense variant in the plasminogen activator, urokinase receptor (PLAUR) gene (rs4760), confirmed experimentally to lead to higher suPAR levels. Mendelian randomization analysis in the UK Biobank using rs4760 indicated a causal association between genetically predicted suPAR levels and atherosclerotic phenotypes. In an experimental model of atherosclerosis, proprotein convertase subtilisin/kexin-9 (Pcsk9) transfection in mice overexpressing suPAR (suPARTg) led to substantially increased atherosclerotic plaques with necrotic cores and macrophage infiltration compared with those in WT mice, despite similar cholesterol levels. Prior to induction of atherosclerosis, aortas of suPARTg mice excreted higher levels of CCL2 and had higher monocyte counts compared with WT aortas. Aortic and circulating suPARTg monocytes exhibited a proinflammatory profile and enhanced chemotaxis. These findings characterize suPAR as a pathogenic factor for atherosclerosis acting at least partially through modulation of monocyte function.
Collapse
Affiliation(s)
- George Hindy
- Department of Clinical Sciences, Lund University, Malmö, Sweden
- Department of Population Medicine, Qatar University College of Medicine, QU Health, Doha, Qatar
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
| | - Daniel J. Tyrrell
- Division of Cardiology, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Alexi Vasbinder
- Division of Cardiology, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Changli Wei
- Department of Medicine, Rush University Medical Center, Chicago, Illinois, USA
| | - Feriel Presswalla
- Division of Cardiology, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Hui Wang
- Division of Cardiology, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Pennelope Blakely
- Division of Cardiology, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Ayse Bilge Ozel
- Department of Human Genetics, University of Michigan, Ann Arbor, Michigan, USA
| | - Sarah Graham
- Department of Human Genetics, University of Michigan, Ann Arbor, Michigan, USA
| | - Grace H. Holton
- Division of Cardiology, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Joseph Dowsett
- Department of Clinical Immunology, Copenhagen University Hospital, Copenhagen, Denmark
| | - Akl C. Fahed
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
| | - Kingsley-Michael Amadi
- Division of Cardiology, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Grace K. Erne
- Division of Cardiology, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Annika Tekmulla
- Division of Cardiology, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Anis Ismail
- Division of Cardiology, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Christopher Launius
- Division of Cardiology, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Nona Sotoodehnia
- Cardiovascular Health Research Unit, Division of Cardiology, University of Washington, Seattle, Washington, USA
| | - James S. Pankow
- Division of Epidemiology and Community Health, University of Minnesota, Minneapolis, Minnesota, USA
| | - Lise Wegner Thørner
- Department of Clinical Immunology, Copenhagen University Hospital, Copenhagen, Denmark
| | | | | | - Karina Banasik
- Novo Nordisk Foundation Center for Protein Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Søren Brunak
- Novo Nordisk Foundation Center for Protein Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | | | - Jesper Eugen-Olsen
- Department of Clinical Research, Copenhagen University Hospital Amager and Hvidovre, Hvidovre, Denmark
| | - Sisse Rye Ostrowski
- Department of Clinical Immunology, Copenhagen University Hospital, Copenhagen, Denmark
| | | | - Mary E. Haas
- Regeneron Genetics Center, Regeneron Pharmaceuticals Inc., Tarrytown, New York, USA
| | - Jonas B. Nielsen
- Regeneron Genetics Center, Regeneron Pharmaceuticals Inc., Tarrytown, New York, USA
| | - Luca A. Lotta
- Regeneron Genetics Center, Regeneron Pharmaceuticals Inc., Tarrytown, New York, USA
| | | | - Gunnar Engström
- Department of Clinical Sciences, Lund University, Malmö, Sweden
| | - Olle Melander
- Department of Clinical Sciences, Lund University, Malmö, Sweden
| | | | - Lili Zhao
- Department of Biostatistics, School of Public Health, University of Michigan, Ann Arbor, Michigan, USA
| | - Venkatesh L. Murthy
- Division of Cardiology, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - David J. Pinsky
- Division of Cardiology, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Cristen J. Willer
- Division of Cardiology, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
- Department of Human Genetics, University of Michigan, Ann Arbor, Michigan, USA
| | - Susan R. Heckbert
- Department of Epidemiology, University of Washington, Seattle, Washington, USA
| | - Jochen Reiser
- Department of Medicine, Rush University Medical Center, Chicago, Illinois, USA
| | - Daniel R. Goldstein
- Division of Cardiology, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Karl C. Desch
- Division of Neonatal-Perinatal Medicine, Department of Pediatrics, Program in Cellular and Molecular Biology, University of Michigan, Ann Arbor, Michigan, USA
| | - Salim S. Hayek
- Division of Cardiology, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| |
Collapse
|
14
|
Li Y, Ding Y, Zhao Y, Gui Y, Shen Y, Xiang Q. Prognostic value of soluble urokinase-type plasminogen activator receptor in coronary artery disease: A meta-analysis. Eur J Clin Invest 2022; 52:e13867. [PMID: 36039688 PMCID: PMC9787755 DOI: 10.1111/eci.13867] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/26/2022] [Revised: 08/19/2022] [Accepted: 08/27/2022] [Indexed: 12/30/2022]
Abstract
BACKGROUND A potential inflammatory biomarker, soluble urokinase-type plasminogen activator receptor (suPAR) has been utilized to assist the prognostic assessment of coronary artery disease (CAD) patients; however, outcomes have been inconsistent. The prognostic relevance of suPAR as a predictor of CAD patient adverse outcomes was therefore examined. METHODS Research articles published as of 1 January 2022 were retrieved from PubMed, Embase, the Web of Science and the Cochrane Library. All-cause mortality, cardiovascular mortality and other major cardiovascular events (nonfatal myocardial infarction, heart failure or stroke) were analysed as a subset of relevant studies' results. We calculated hazard ratios (HRs) and 95% confidence intervals (CIs) for each study. The broad EQUATOR guidelines were conformed. Risk of bias was assessed with ROBINS-I tool. RESULTS In total, this analysis included nine studies including 14,738 CAD patients. All included studies made a correction for certain potential confounders. However, risk of bias ranged from moderate to critical. When the ROBINS-I tool was used. Patients with CAD that exhibited increased suPAR levels had a substantially higher risk of all-cause mortality (HR = 2.24; 95% CI 1.97-2.55) or cardiovascular mortality (HR = 2.02; 95% CI 1.58-2.58), but not of developing other major cardiovascular events (HR = 1.63; 95% CI 0.86-3.11). Considerable heterogeneity across studies was observed in our meta-analyses, but no significant publication bias was detected. CONCLUSION In patients with coronary disease, suPAR may have prognostic value for both all-cause and cardiovascular mortality but not for other major cardiovascular events.
Collapse
Affiliation(s)
- Yang Li
- Emergency centerThe First Affiliated Hospital of Army Medical UniversityChongqingChina
| | - Yaqun Ding
- Emergency centerThe First Affiliated Hospital of Army Medical UniversityChongqingChina
| | - Yinjie Zhao
- Emergency centerThe First Affiliated Hospital of Army Medical UniversityChongqingChina
| | - Yongqing Gui
- Emergency centerThe First Affiliated Hospital of Army Medical UniversityChongqingChina
| | - Yajing Shen
- Emergency centerThe First Affiliated Hospital of Army Medical UniversityChongqingChina
| | - Qiang Xiang
- Emergency centerThe First Affiliated Hospital of Army Medical UniversityChongqingChina
| |
Collapse
|
15
|
Vellasamy DM, Lee SJ, Goh KW, Goh BH, Tang YQ, Ming LC, Yap WH. Targeting Immune Senescence in Atherosclerosis. Int J Mol Sci 2022; 23:13059. [PMID: 36361845 PMCID: PMC9658319 DOI: 10.3390/ijms232113059] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Revised: 10/07/2022] [Accepted: 10/10/2022] [Indexed: 10/29/2023] Open
Abstract
Atherosclerosis is one of the main underlying causes of cardiovascular diseases (CVD). It is associated with chronic inflammation and intimal thickening as well as the involvement of multiple cell types including immune cells. The engagement of innate or adaptive immune response has either athero-protective or atherogenic properties in exacerbating or alleviating atherosclerosis. In atherosclerosis, the mechanism of action of immune cells, particularly monocytes, macrophages, dendritic cells, and B- and T-lymphocytes have been discussed. Immuno-senescence is associated with aging, viral infections, genetic predispositions, and hyperlipidemia, which contribute to atherosclerosis. Immune senescent cells secrete SASP that delays or accelerates atherosclerosis plaque growth and associated pathologies such as aneurysms and coronary artery disease. Senescent cells undergo cell cycle arrest, morphological changes, and phenotypic changes in terms of their abundances and secretome profile including cytokines, chemokines, matrix metalloproteases (MMPs) and Toll-like receptors (TLRs) expressions. The senescence markers are used in therapeutics and currently, senolytics represent one of the emerging treatments where specific targets and clearance of senescent cells are being considered as therapy targets for the prevention or treatment of atherosclerosis.
Collapse
Affiliation(s)
- Danusha Michelle Vellasamy
- School of Biosciences, Faculty of Medical and Health Sciences, Taylor’s University, Subang Jaya 47500, Malaysia
| | - Sin-Jye Lee
- School of Biosciences, Faculty of Medical and Health Sciences, Taylor’s University, Subang Jaya 47500, Malaysia
| | - Khang Wen Goh
- Faculty of Data Science and Information Technology, INTI International University, Nilai 71800, Malaysia
| | - Bey-Hing Goh
- Biofunctional Molecule Exploratory (BMEX) Research Group, School of Pharmacy, Monash University Malaysia, Bandar Sunway 47500, Malaysia
- College of Pharmaceutical Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou 310058, China
| | - Yin-Quan Tang
- School of Biosciences, Faculty of Medical and Health Sciences, Taylor’s University, Subang Jaya 47500, Malaysia
- Centre for Drug Discovery and Molecular Pharmacology, Faculty of Medical and Health Sciences, Taylor’s University, Subang Jaya 47500, Malaysia
| | - Long Chiau Ming
- PAP Rashidah Sa’adatul Bolkiah Institute of Health Sciences, Universiti Brunei Darussalam, Gadong BE1410, Brunei
| | - Wei Hsum Yap
- School of Biosciences, Faculty of Medical and Health Sciences, Taylor’s University, Subang Jaya 47500, Malaysia
- Centre for Drug Discovery and Molecular Pharmacology, Faculty of Medical and Health Sciences, Taylor’s University, Subang Jaya 47500, Malaysia
| |
Collapse
|
16
|
Haring B, McGinn AP, Kamensky V, Allison M, Stefanick ML, Schnatz PF, Kuller LH, Berger JS, Johnson KC, Saquib N, Garcia L, Richey PA, Manson JE, Alderman M, Wassertheil-Smoller S. Low Diastolic Blood Pressure and Mortality in Older Women. Results From the Women's Health Initiative Long Life Study. Am J Hypertens 2022; 35:795-802. [PMID: 35522983 PMCID: PMC9434234 DOI: 10.1093/ajh/hpac056] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2022] [Revised: 03/17/2022] [Accepted: 05/02/2022] [Indexed: 02/02/2023] Open
Abstract
BACKGROUND Recommended systolic blood pressure targets often do not consider the relationship of low diastolic blood pressure (DBP) levels with cardiovascular disease (CVD) and all-cause mortality risk, which is especially relevant for older people with concurrent comorbidities. We examined the relationship of DBP levels to CVD and all-cause mortality in older women in the Women's Health Initiative Long Life Study (WHI-LLS). METHODS The study sample included 7,875 women (mean age: 79 years) who underwent a blood pressure measurement at an in-person home visit conducted in 2012-2013. CVD and all-cause mortality were centrally adjudicated. Hazard ratios (HRs) were obtained from adjusted Cox proportional hazards models. RESULTS After 5 years follow-up, all-cause mortality occurred in 18.4% of women. Compared with a DBP of 80 mm Hg, the fully adjusted HR for mortality was 1.33 (95% confidence interval [CI]: 1.04-1.71) for a DBP of 50 mm Hg and 1.67 (95% CI: 1.29-2.16) for a DBP of 100 mm Hg. The HRs for CVD were 1.14 (95% CI: 0.78-1.67) for a DBP of 50 mm Hg and HR 1.50 (95% CI: 1.03-2.17) for a DBP of 100 mm Hg. The nadir DBP associated with lowest mortality risk was 72 mm Hg overall. CONCLUSIONS In older women, consideration should be given to the potential adverse effects of low and high DBP. Low DBP may serve as a risk marker. DBP target levels between 68 and 75 mm Hg may avoid higher mortality risk.
Collapse
Affiliation(s)
- Bernhard Haring
- Department of Medicine III, Saarland University Hospital, Homburg, Germany
- Department of Medicine I, University of Würzburg, Würzburg, Germany
- Department of Epidemiology and Population Health, Albert Einstein College of Medicine, Bronx, New York, USA
| | - Aileen P McGinn
- Department of Epidemiology and Population Health, Albert Einstein College of Medicine, Bronx, New York, USA
| | - Victor Kamensky
- Department of Epidemiology and Population Health, Albert Einstein College of Medicine, Bronx, New York, USA
| | - Matthew Allison
- Department of Family Medicine, University of California San Diego, La Jolla, California, USA
| | - Marcia L Stefanick
- Department of Medicine, Stanford University Medical Center, Palo Alto, California, USA
| | - Peter F Schnatz
- Department of Obstetrics/Gynecology and Internal Medicine, Reading Hospital/Tower Health, West Reading, Pennsylvania, USA
| | - Lewis H Kuller
- Department of Epidemiology, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Jeffrey S Berger
- Department of Medicine, Center for the Prevention of Cardiovascular Disease, New York University School of Medicine, New York City, New York, USA
| | - Karen C Johnson
- Department of Preventive Medicine, University of Tennessee Health Science Center, Memphis, Tennessee, USA
| | - Nazmus Saquib
- College of Medicine, Sulaiman Al Rajhi University, Al Bukayriyah, Saudi Arabia
| | - Lorena Garcia
- Department of Public Health Sciences, UC Davis School of Medicine, Sacramento, California, USA
| | - Phyllis A Richey
- Department of Preventive Medicine, University of Tennessee Health Science Center, Memphis, Tennessee, USA
| | - JoAnn E Manson
- Department of Medicine, Division of Preventive Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Michael Alderman
- Department of Epidemiology and Population Health, Albert Einstein College of Medicine, Bronx, New York, USA
| | - Sylvia Wassertheil-Smoller
- Department of Epidemiology and Population Health, Albert Einstein College of Medicine, Bronx, New York, USA
| |
Collapse
|
17
|
Identification Markers of Carotid Vulnerable Plaques: An Update. Biomolecules 2022; 12:biom12091192. [PMID: 36139031 PMCID: PMC9496377 DOI: 10.3390/biom12091192] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2022] [Revised: 08/22/2022] [Accepted: 08/24/2022] [Indexed: 12/02/2022] Open
Abstract
Vulnerable plaques have been a hot topic in the field of stroke and carotid atherosclerosis. Currently, risk stratification and intervention of carotid plaques are guided by the degree of luminal stenosis. Recently, it has been recognized that the vulnerability of plaques may contribute to the risk of stroke. Some classical interventions, such as carotid endarterectomy, significantly reduce the risk of stroke in symptomatic patients with severe carotid stenosis, while for asymptomatic patients, clinically silent plaques with rupture tendency may expose them to the risk of cerebrovascular events. Early identification of vulnerable plaques contributes to lowering the risk of cerebrovascular events. Previously, the identification of vulnerable plaques was commonly based on imaging technologies at the macroscopic level. Recently, some microscopic molecules pertaining to vulnerable plaques have emerged, and could be potential biomarkers or therapeutic targets. This review aimed to update the previous summarization of vulnerable plaques and identify vulnerable plaques at the microscopic and macroscopic levels.
Collapse
|
18
|
Qian J, Gao Y, Lai Y, Ye Z, Yao Y, Ding K, Tong J, Lin H, Zhu G, Yu Y, Ding H, Yuan D, Chu J, Chen F, Liu X. Single-Cell RNA Sequencing of Peripheral Blood Mononuclear Cells From Acute Myocardial Infarction. Front Immunol 2022; 13:908815. [PMID: 35844519 PMCID: PMC9278132 DOI: 10.3389/fimmu.2022.908815] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Accepted: 05/26/2022] [Indexed: 11/18/2022] Open
Abstract
Background Acute myocardial infarction (AMI) can occur in patients with atherosclerotic disease, with or without plaque rupture. Previous studies have indicated a set of immune responses to plaque rupture. However, the specific circulating immune cell subsets that mediate inflammatory plaque rupture remain elusive. Methods Ten AMI patients were enrolled in our study (five with and five without plaque rupture; plaque characteristics were identified by optical coherence tomography). By single-cell RNA sequencing, we analyzed the transcriptomic profile of peripheral blood mononuclear cells. Results We identified 27 cell clusters among 82,550 cells, including monocytes, T cells, NK cells, B cells, megakaryocytes, and CD34+ cells. Classical and non-classical monocytes constitute the major inflammatory cell types, and pro-inflammatory genes such as CCL5, TLR7, and CX3CR1 were significantly upregulated in patients with plaque rupture, while the neutrophil activation and degranulation genes FPR2, MMP9, and CLEC4D were significantly expressed in the intermediate monocytes derived from patients without plaque rupture. We also found that CD4+ effector T cells may contribute to plaque rupture by producing a range of cytokines and inflammatory-related chemokines, while CD8+ effector T cells express more effector molecules in patients without plaque rupture, such as GZMB, GNLY, and PRF1, which may contribute to the progress of plaque erosion. Additionally, NK and B cells played a significant role in activating inflammatory cells and promoting chemokine production in the plaque rupture. Cell-cell communication elaborated characteristics in signaling pathways dominated by inflammatory activation of classical monocytes in patients with plaque rupture. Conclusions Our studies demonstrate that the circulating immune cells of patients with plaque rupture exhibit highly pro-inflammatory characteristics, while plaque erosion is mainly associated with intermediate monocyte amplification, neutrophil activation, and degranulation. These findings may provide novel targets for the precise treatment of patients with AMI.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | | | | | - Fei Chen
- Department of Cardiology, Tongji Hospital, Tongji University School of Medicine, Shanghai, China
| | - Xuebo Liu
- Department of Cardiology, Tongji Hospital, Tongji University School of Medicine, Shanghai, China
| |
Collapse
|
19
|
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]
|
20
|
Bicvic A, Scherrer N, Schweizer J, Fluri F, Christ-Crain M, De Marchis GM, Luft AR, Katan M. A novel biomarker panel index improves risk stratification after ischemic stroke. Eur Stroke J 2022; 7:158-165. [PMID: 35647313 PMCID: PMC9134784 DOI: 10.1177/23969873221090798] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Accepted: 03/09/2022] [Indexed: 11/15/2022] Open
Abstract
Background: We investigated 92 blood biomarkers implicated in the pathophysiological pathways of ischemic injury, inflammation, hemostasis, and regulation of vascular resistance to predict post-stroke mortality. Aim: Based on the most promising markers, we aimed to create a novel Biomarker Panel Index (BPI) for risk stratification. Methods: In this prospective study, we measured 92 biomarkers in 320 stroke patients. The primary outcome measure was mortality within 90 days. We estimated the association of each biomarker using logistic regression adjusting for multiple testing. The most significant 16 biomarkers were used to create the BPI. We fitted regression models to estimate the association and the discriminatory accuracy of the BPI with mortality and stroke etiology. Results: Adjusted for demographic and vascular covariates, the BPI remained independently associated with mortality (odds ratio (OR) 1.68, 95% confidence interval (CI): 1.29–2.18) and cardioembolic stroke etiology (OR 1.38, 95% CI: 1.10–1.74), and improved the discriminatory accuracy to predict mortality (area under the receiver operating characteristic curve (AUC) 0.93, 95% CI: 0.89–0.96) and cardioembolic stroke etiology (AUC 0.70, 95% CI: 0.64–0.77) as compared to the best clinical prediction models alone (AUC 0.89, 95% CI: 0.84–0.94 and AUC 0.66, 95% CI: 0.60-0.73, respectively). Conclusions: We identified a novel BPI improving risk stratification for mortality after ischemic stroke beyond established demographic and vascular risk factors. Furthermore, the BPI is associated with underlying cardioembolic stroke etiology. These results need external validation.
Collapse
Affiliation(s)
- Antonela Bicvic
- Department of Neurology, University Hospital of Zurich, Zurich, Switzerland
- Department of Neurology, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Natalie Scherrer
- Department of Neurology, University Hospital of Zurich, Zurich, Switzerland
| | - Juliane Schweizer
- Department of Neurology, University Hospital of Zurich, Zurich, Switzerland
| | - Felix Fluri
- Department of Neurology, SRO Gesundheitszentrum, Bad Wimpfen, Germany
- Department of Neurology, University Hospital Würzburg, Würzburg, Germany
| | - Mirjam Christ-Crain
- Department of Endocrinology, University Hospital of Basel, Basel, Switzerland
| | - Gian Marco De Marchis
- Department of Neurology, University Hospital of Basel and University of Basel, Basel, Switzerland
| | - Andreas R Luft
- Department of Neurology, University Hospital of Zurich, Zurich, Switzerland
| | - Mira Katan
- Department of Neurology, University Hospital of Zurich, Zurich, Switzerland
| |
Collapse
|
21
|
Williams SA, Ostroff R, Hinterberg MA, Coresh J, Ballantyne CM, Matsushita K, Mueller CE, Walter J, Jonasson C, Holman RR, Shah SH, Sattar N, Taylor R, Lean ME, Kato S, Shimokawa H, Sakata Y, Nochioka K, Parikh CR, Coca SG, Omland T, Chadwick J, Astling D, Hagar Y, Kureshi N, Loupy K, Paterson C, Primus J, Simpson M, Trujillo NP, Ganz P. A proteomic surrogate for cardiovascular outcomes that is sensitive to multiple mechanisms of change in risk. Sci Transl Med 2022; 14:eabj9625. [PMID: 35385337 DOI: 10.1126/scitranslmed.abj9625] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
A reliable, individualized, and dynamic surrogate of cardiovascular risk, synoptic for key biologic mechanisms, could shorten the path for drug development, enhance drug cost-effectiveness and improve patient outcomes. We used highly multiplexed proteomics to address these objectives, measuring about 5000 proteins in each of 32,130 archived plasma samples from 22,849 participants in nine clinical studies. We used machine learning to derive a 27-protein model predicting 4-year likelihood of myocardial infarction, stroke, heart failure, or death. The 27 proteins encompassed 10 biologic systems, and 12 were associated with relevant causal genetic traits. We independently validated results in 11,609 participants. Compared to a clinical model, the ratio of observed events in quintile 5 to quintile 1 was 6.7 for proteins versus 2.9 for the clinical model, AUCs (95% CI) were 0.73 (0.72 to 0.74) versus 0.64 (0.62 to 0.65), c-statistics were 0.71 (0.69 to 0.72) versus 0.62 (0.60 to 0.63), and the net reclassification index was +0.43. Adding the clinical model to the proteins only improved discrimination metrics by 0.01 to 0.02. Event rates in four predefined protein risk categories were 5.6, 11.2, 20.0, and 43.4% within 4 years; median time to event was 1.71 years. Protein predictions were directionally concordant with changed outcomes. Adverse risks were predicted for aging, approaching an event, anthracycline chemotherapy, diabetes, smoking, rheumatoid arthritis, cancer history, cardiovascular disease, high systolic blood pressure, and lipids. Reduced risks were predicted for weight loss and exenatide. The 27-protein model has potential as a "universal" surrogate end point for cardiovascular risk.
Collapse
Affiliation(s)
| | | | | | - Josef Coresh
- Johns Hopkins University, Baltimore, MD 21218, USA
| | | | | | - Christian E Mueller
- Cardiovascular Research Institute, University of Basel, Basel 4001, Switzerland
| | - Joan Walter
- Cardiovascular Research Institute, University of Basel, Basel 4001, Switzerland.,Institute of Diagnostic and Interventional Radiology, University Hospital Zürich, University of Zürich, Zürich 7491, Switzerland
| | - Christian Jonasson
- Jebsen Centre for Genetic Epidemiology, Department of Public Health and Nursing, Norwegian University of Science and Technology, Trondheim 7491, Norway
| | - Rury R Holman
- Diabetes Trials Unit, Radcliffe Department of Medicine, University of Oxford, Oxford OX3 9DU, UK
| | - Svati H Shah
- Division of Cardiology, Duke Department of Medicine, and Duke Molecular Physiology Institute, Duke University, Durham, NC 27710, USA
| | - Naveed Sattar
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow G12 8QQ, UK
| | - Roy Taylor
- Newcastle Magnetic Resonance Centre, University of Newcastle upon Tyne, Newcastle upon Tyne NE1 7RU, UK
| | - Michael E Lean
- School of Medicine, Nursing and Dentistry, University of Glasgow, Glasgow G12 8QQ, UK
| | | | - Hiroaki Shimokawa
- Tohoku University Graduate School of Medicine, Sendai 980-8576, Japan.,Graduate School, International University of Health and Welfare, Narita 286-8686, Japan
| | - Yasuhiko Sakata
- Tohoku University Graduate School of Medicine, Sendai 980-8576, Japan
| | - Kotaro Nochioka
- Tohoku University Graduate School of Medicine, Sendai 980-8576, Japan
| | | | - Steven G Coca
- Mt Sinai Clinical and Translational Science Research Unit, Icahn School of Medicine at Mount Sinai, New York, NY 11766, USA
| | - Torbjørn Omland
- Department of Cardiology, Akershus University Hospital and University of Oslo, Oslo 1478, Norway
| | | | | | | | | | | | | | | | | | | | - Peter Ganz
- Zuckerberg San Francisco General Hospital, University of California, San Francisco, San Francisco, CA 94110, USA
| |
Collapse
|
22
|
Khare HA, Døssing KBV, Ringgaard L, Christensen E, Urbak L, Sillesen H, Ripa RS, Binderup T, Pedersen SF, Kjaer A. In vivo detection of urokinase-type plasminogen activator receptor (uPAR) expression in arterial atherogenesis using [64Cu]Cu-DOTA-AE105 positron emission tomography (PET). Atherosclerosis 2022; 352:103-111. [DOI: 10.1016/j.atherosclerosis.2022.03.026] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/19/2021] [Revised: 02/23/2022] [Accepted: 03/25/2022] [Indexed: 12/21/2022]
|
23
|
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:brainsci12010039. [PMID: 35053782 PMCID: PMC8774014 DOI: 10.3390/brainsci12010039] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 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
|
24
|
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: 9.7] [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
|
25
|
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: 0.7] [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
|
26
|
Kumar S, Sur S, Perez J, Demos C, Kang DW, Kim CW, Hu S, Xu K, Yang J, Jo H. Atorvastatin and blood flow regulate expression of distinctive sets of genes in mouse carotid artery endothelium. CURRENT TOPICS IN MEMBRANES 2021; 87:97-130. [PMID: 34696890 DOI: 10.1016/bs.ctm.2021.08.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/29/2023]
Abstract
Hypercholesterolemia is a well-known pro-atherogenic risk factor and statin is the most effective anti-atherogenic drug that lowers blood cholesterol levels. However, despite systemic hypercholesterolemia, atherosclerosis preferentially occurs in arterial regions exposed to disturbed blood flow (d-flow), while the stable flow (s-flow) regions are spared. Given their predominant effects on endothelial function and atherosclerosis, we tested whether (1) statin and flow regulate the same or independent sets of genes and (2) statin can rescue d-flow-regulated genes in mouse artery endothelial cells in vivo. To test the hypotheses, C57BL/6 J mice (8-week-old male, n=5 per group) were pre-treated with atorvastatin (10mg/kg/day, Orally) or vehicle for 5 days. Thereafter, partial carotid ligation (PCL) surgery to induce d-flow in the left carotid artery (LCA) was performed, and statin or vehicle treatment was continued. The contralateral right carotid artery (RCA) remained exposed to s-flow to be used as the control. Two days or 2 weeks post-PCL surgery, endothelial-enriched RNAs from the LCAs and RCAs were collected and subjected to microarray gene expression analysis. Statin treatment in the s-flow condition (RCA+statin versus RCA+vehicle) altered the expression of 667 genes at 2-day and 187 genes at 2-week timepoint, respectively (P<0.05, fold change (FC)≥±1.5). Interestingly, statin treatment in the d-flow condition (LCA+statin versus LCA+vehicle) affected a limited number of genes: 113 and 75 differentially expressed genes at 2-day and 2-week timepoint, respectively (P<0.05, FC≥±1.5). In contrast, d-flow in the vehicle groups (LCA+vehicle versus RCA+vehicle) differentially regulated 4061 genes at 2-day and 3169 genes at 2-week timepoint, respectively (P<0.05, FC≥±1.5). Moreover, statin treatment did not reduce the number of flow-sensitive genes (LCA+statin versus RCA+statin) compared to the vehicle groups: 1825 genes at 2-day and 3788 genes at 2-week, respectively, were differentially regulated (P<0.05, FC≥±1.5). These results revealed that both statin and d-flow regulate expression of hundreds or thousands of arterial endothelial genes, respectively, in vivo. Further, statin and d-flow regulate independent sets of endothelial genes. Importantly, statin treatment did not reverse d-flow-regulated genes except for a small number of genes. These results suggest that both statin and flow play important independent roles in atherosclerosis development and highlight the need to consider their therapeutic implications for both.
Collapse
Affiliation(s)
- Sandeep Kumar
- Wallace H. Coulter Department of Biomedical Engineering at Emory University and Georgia Institute of Technology, Atlanta, GA, United States
| | - Sanjoli Sur
- Wallace H. Coulter Department of Biomedical Engineering at Emory University and Georgia Institute of Technology, Atlanta, GA, United States
| | - Julian Perez
- Wallace H. Coulter Department of Biomedical Engineering at Emory University and Georgia Institute of Technology, Atlanta, GA, United States
| | - Catherine Demos
- Wallace H. Coulter Department of Biomedical Engineering at Emory University and Georgia Institute of Technology, Atlanta, GA, United States
| | - Dong-Won Kang
- Wallace H. Coulter Department of Biomedical Engineering at Emory University and Georgia Institute of Technology, Atlanta, GA, United States
| | - Chan Woo Kim
- Wallace H. Coulter Department of Biomedical Engineering at Emory University and Georgia Institute of Technology, Atlanta, GA, United States
| | - Sarah Hu
- Thrombosis Research Unit, Bristol Myers Squibb, Lawrence, NJ, United States
| | - Ke Xu
- Thrombosis Research Unit, Bristol Myers Squibb, Lawrence, NJ, United States
| | - Jing Yang
- Thrombosis Research Unit, Bristol Myers Squibb, Lawrence, NJ, United States
| | - Hanjoong Jo
- Wallace H. Coulter Department of Biomedical Engineering at Emory University and Georgia Institute of Technology, Atlanta, GA, United States; Division of Cardiology, Emory University, Atlanta, GA, United States.
| |
Collapse
|
27
|
Siniscalchi A, Murphy S, Gray C, De Sarro G, Gallelli L. Biomarkers in unstable carotid plaque: Physiopathology and Prediction. Cardiovasc Hematol Agents Med Chem 2021; 20:13-19. [PMID: 34468303 DOI: 10.2174/1871525719666210901131509] [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: 05/26/2021] [Revised: 06/17/2021] [Accepted: 07/14/2021] [Indexed: 11/22/2022]
Abstract
AIMS To study the role of cytokines and vascular inflammatory biomarkers in unstable carotid plaque. BACKGROUND Clinical studies showed that not only the degree of stenosis but also the type of carotid plaque can be responsible for ipsilateral ischemic stroke. OBJECTIVE The objective of this study is to suggest a role for vulnerable carotid atherosclerotic disease in the occurrence of ischemic stroke. METHODS PubMed, Embase, Cochrane library, and reference lists have been used to evaluate articles published until February 15, 2021. RESULTS Several factors may be involved in unstable plaque. Clinical studies support the involvement of brain inflammatory biomarkers as well as cytokines in the unstable carotid plaque. CONCLUSIONS Biomarkers could help to stratify patients with a vulnerable carotid plaque and to personalize the drug treatment. In this review, we briefly discuss the characteristics of vulnerable plaque and the role of biomarkers in the vulnerable carotid plaque.
Collapse
Affiliation(s)
- Antonio Siniscalchi
- Department of Neurology and Stroke Unit, Annunziata Hospital of Cosenza, Cosenza, Italy
| | - Sean Murphy
- General Medicine, Stroke Unit, Mater Misericordiae University Hospital, Dublin, Ireland
| | - Cleona Gray
- Vascular and Endovascular Surgery Unit, Mater Misericordiae University Hospital, Dublin, Ireland
| | - Giovambattista De Sarro
- Chair of Pharmacology, Department of Health Science, School of Medicine, University of Catanzaro, Clinical Pharmacology Unit, Mater Domini University Hospital, Catanzaro, Italy
| | - Luca Gallelli
- Department of Health Science, School of Medicine, University of Catanzaro, Catanzaro, Italy
| |
Collapse
|
28
|
Immune-mediated entities of (primary) focal segmental glomerulosclerosis. Cell Tissue Res 2021; 385:423-434. [PMID: 33907872 PMCID: PMC8523460 DOI: 10.1007/s00441-021-03454-3] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2020] [Accepted: 03/19/2021] [Indexed: 12/21/2022]
Abstract
Focal segmental glomerulosclerosis (FSGS) represents a glomerular scar formation downstream of various different mechanisms leading to podocytopathy and podocyte loss. Recently, significant advances were made in understanding genetic factors, podocyte intrinsic mechanisms, and adaptive mechanisms causing FSGS. However, while most cases of nephrotic FSGS are being treated with immunosuppressants, the underlying immune dysregulation, involved immune cells, and soluble factors are only incompletely understood. Thus, we here summarize the current knowledge of proposed immune effector cells, secreted soluble factors, and podocyte response in immune-mediated (primary) FSGS.
Collapse
|
29
|
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: 1.0] [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
|
30
|
Lazarević M, Golubović M, Milić D, Stanojević D, Kostić T, Đorđević M, Marjanović V, Perić V. Preoperative Levels of the Soluble Urokinase-Type Plasminogen Activator Receptor as Predictor for New Episodes of Atrial Fibrillation After Vascular Surgery. Vasc Endovascular Surg 2021; 55:461-466. [PMID: 33622185 DOI: 10.1177/1538574421995321] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
INTRODUCTION The soluble urokinase-type plasminogen activator receptor (suPAR) in uPAR soluble form is produced when the uPAR is cleaved from the cell membrane during the inflammatory process. Postoperative atrial fibrillation (AF) is the most common perioperative cardiac arrhythmia. It is speculated that elevated suPAR has a role in the development of AF. The aim of our study was to investigate the predictive role of preoperative suPAR in the occurrence of AF during the first 6 months after major vascular surgery. METHODS We included 119 male and 63 female patients with an average age of 67.19 ± 6.02 years, without permanent/persistent AF. Basic predictive model (BASIC) included traditional risk factors for AF: age, gender, body mass index-BMI, smoking status, presence of arterial hypertension, diabetes mellitus and dyslipidemia. RESULTS Over the 6-month period, 19 (10.4%) patients had one new episode of atrial fibrillation and 2 patients (1.1%) had 2 episodes of paroxysmal atrial fibrillation. Paroxysms of AF were significantly more frequent in patients who had a resection of an abdominal aneurysm than in patients with other types of major vascular surgery. BASIC had good discriminatory ability in the prediction of AF paroxysms during the first 6 months after surgery (AUC = 0.715, 95%CI 0.590-0.840). Adding suPAR to the basic model significantly improved the discriminative ability of the predictive model for AF episodes (ΔAUC = 0.238, p < 0.001). The predictive performance of the model BASIC+CRP+suPAR, measured using AUC, NRI and IDI statistics, was very similar to the model BASIC+suPAR. CONCLUSION AF is a common complication in surgical patients with high mortality and morbidity. suPAR could improve the ability of traditional risk factors to predict its occurrence up to 6 months after major vascular surgery.
Collapse
Affiliation(s)
- Milan Lazarević
- Clinic of Cardiovascular and Transplant Surgery, 256420Clinical Center Niš, Niš, Serbia
| | - Mladjan Golubović
- Department for Cardiac Surgery, Clinic for Anaesthesiology and Intensive Therapy, 256420Clinical Center Niš, Niš, Serbia.,Medical School of Nis, University of Niš, Niš, Serbia
| | - Dragan Milić
- Clinic of Cardiovascular and Transplant Surgery, 256420Clinical Center Niš, Niš, Serbia.,Medical School of Nis, University of Niš, Niš, Serbia
| | | | - Tomislav Kostić
- Medical School of Nis, University of Niš, Niš, Serbia.,Clinic for Cardiology, 256420Clinical Center Niš, Niš, Serbia
| | - Miodrag Đorđević
- Medical School of Nis, University of Niš, Niš, Serbia.,Clinic for Endocrine Surgery, Clinical Center Niš, Niš, Serbia
| | - Vesna Marjanović
- Medical School of Nis, University of Niš, Niš, Serbia.,Department for Pediatric Surgery, Clinic for Anaesthesiology and Intensive Therapy, Clinical Center Niš, Niš, Serbia
| | - Velimir Perić
- Department for Cardiac Surgery, Clinic for Anaesthesiology and Intensive Therapy, 256420Clinical Center Niš, Niš, Serbia
| |
Collapse
|
31
|
Wijeratne T, Menon R, Sales C, Karimi L, Crewther S. Carotid artery stenosis and inflammatory biomarkers: the role of inflammation-induced immunological responses affecting the vascular systems. ANNALS OF TRANSLATIONAL MEDICINE 2020; 8:1276. [PMID: 33178808 PMCID: PMC7607082 DOI: 10.21037/atm-20-4388] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
The death, disability and economic cost of stroke are enormous. Indeed, among the 16 million people worldwide who suffer a stroke' annually, nearly six million die, and another five million are left permanently disabled making prevention of stroke one of the most important priorities in healthcare. Currently carotid artery stenosis (CS) or narrowing of the common carotid artery (CCA) or internal carotid artery (ICA) due to atherosclerotic plaque, accounts for 20-30% of all ischemic strokes. Atherosclerosis is now regarded as a chronic inflammatory disease in response to vascular compromise especially from hypertension. This has long been known to lead to inflammation and atherosclerotic plaque formation in the blood vessels. This mini-review aims to highlight the role of inflammation and neuro-immunological processes in carotid artery disease. Various cellular elements of inflammation and advanced imaging techniques have been identified as potential markers of plaque progression. Therapies related to decreasing and modulating immune-responsive inflammation in the carotid vessels have been shown to translate into decreased occurrence of acute neurologic events and improvement of clinical outcomes.
Collapse
Affiliation(s)
- Tissa Wijeratne
- Department of Neurology, AIMSS, WHCRE level three, Sunshine Hospital and Melbourne Medical School, St Albans, Victoria, Australia.,School of Public health and Psychology, La Trobe University, Bundoora, Victoria, Australia.,Department of Medicine, Faculty of Medicine, Rajarata University, Saliyapura, Anuradhapura, Sri Lanka
| | - Rohit Menon
- Department of Neurology and Stroke Service, Western Health, AIMSS, level 3, WHCRE, Sunshine Hospital, St Albans, Victoria, Australia
| | - Carmela Sales
- Department of Neurology and Stroke Service, Western Health, AIMSS, level 3, WHCRE, Sunshine Hospital, St Albans, Victoria, Australia
| | - Leila Karimi
- School of Psychology and Public Health, La Trobe University, Melbourne, Australia.,Ivane Javakhishvili Tbilisi State University, Tbilisi, Georgia
| | - Sheila Crewther
- School of Psychology and Public Health, La Trobe University, Melbourne, Australia
| |
Collapse
|
32
|
CD163+ macrophages are associated with a vulnerable plaque phenotype in human carotid plaques. Sci Rep 2020; 10:14362. [PMID: 32873809 PMCID: PMC7463157 DOI: 10.1038/s41598-020-71110-x] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2019] [Accepted: 07/27/2020] [Indexed: 01/15/2023] Open
Abstract
Macrophages are a functionally heterogeneous group of immune cells abundant in atherosclerotic plaques. Macrophages expressing CD163 are associated with intraplaque hemorrhage and have previously been considered atheroprotective. However, in a recent study CD163-deficient atherosclerotic ApoE-/- mice exhibited smaller and less complex plaques, suggesting a proatherogenic role of CD163. Previous smaller studies on CD163+ macrophages and plaque stability in humans have yielded diverging results. Here we assessed the association of CD163+ cells to plaque vulnerability in a large cohort of human carotid plaques. CD163 protein expression was analyzed by immunohistochemistry in 200 human carotid plaques removed by endarterectomy from 103 patients with and 93 patients without cerebrovascular symptoms. Furthermore, CD163 mRNA expression was analyzed in 66 of the plaques. Both protein and mRNA expression of CD163 was higher in plaques from symptomatic patients and in plaques with high vulnerability index. CD163+ macrophages were primarily found in shoulder regions and in the center of the plaques. The present data show that CD163 is associated with increased plaque vulnerability in human carotid plaques, supporting the notion that CD163+ macrophages could contribute to clinical events.
Collapse
|
33
|
Serum soluble urokinase plasminogen activator receptor in adolescents: interaction of chronic pain and obesity. Pain Rep 2020; 5:e836. [PMID: 32766470 PMCID: PMC7382552 DOI: 10.1097/pr9.0000000000000836] [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: 03/27/2020] [Revised: 06/05/2020] [Accepted: 06/18/2020] [Indexed: 12/24/2022] Open
Abstract
Pediatric chronic pain is exacerbated by obesity. Serum soluble urokinase plasminogen activator receptor, a novel inflammation biomarker, was increased in obese adolescents with chronic pain indicating interactive inflammatory conditions. Introduction: Obesity in adolescents is increasing in frequency and is associated with short-term and long-term negative consequences that include the exacerbation of co-occurring chronic pain. Objective: To determine whether the interaction between chronic pain and obesity would be reflected in changes in serum soluble urokinase plasminogen activator receptor (suPAR) concentrations, a novel marker of systemic inflammation associated with obesity, insulin resistance, and cardiovascular disease. Methods: We measured serum suPAR levels in 146 adolescent males and females with no pain or obesity (healthy controls; n = 40), chronic pain with healthy weight (n = 37), obesity alone (n = 41), and the combination of chronic pain and obesity (n = 28). Results: Serum suPAR (median [interquartile range]) was not increased by chronic pain alone (2.2 [1.8–2.4] ng/mL) or obesity alone (2.2 [2.0–2.4] ng/mL) but was increased significantly with the combination of chronic pain and obesity (2.4 [2.1–2.7] ng/mL; P < 0.019). This finding confirms the proposition that pain and obesity are inflammatory states that display a classic augmenting interaction. Conclusion: We propose that measurement of serum suPAR can be added to the armamentarium of serum biomarkers useful in the evaluation of mechanisms of inflammation in adolescent obesity and chronic pain.
Collapse
|
34
|
HIV-associated gut dysbiosis is independent of sexual practice and correlates with noncommunicable diseases. Nat Commun 2020; 11:2448. [PMID: 32415070 PMCID: PMC7228978 DOI: 10.1038/s41467-020-16222-8] [Citation(s) in RCA: 107] [Impact Index Per Article: 26.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2019] [Accepted: 04/15/2020] [Indexed: 02/08/2023] Open
Abstract
Loss of gut mucosal integrity and an aberrant gut microbiota are proposed mechanisms contributing to chronic inflammation and increased morbidity and mortality during antiretroviral-treated HIV disease. Sexual practice has recently been uncovered as a major source of microbiota variation, potentially confounding prior observations of gut microbiota alterations among persons with HIV (PWH). To overcome this and other confounding factors, we examine a well-powered subset of AGEhIV Cohort participants comprising antiretroviral-treated PWH and seronegative controls matched for age, body-mass index, sex, and sexual practice. We report significant gut microbiota differences in PWH regardless of sex and sexual practice including Gammaproteobacteria enrichment, Lachnospiraceae and Ruminococcaceae depletion, and decreased alpha diversity. Men who have sex with men (MSM) exhibit a distinct microbiota signature characterized by Prevotella enrichment and increased alpha diversity, which is linked with receptive anal intercourse in both males and females. Finally, the HIV-associated microbiota signature correlates with inflammatory markers including suPAR, nadir CD4 count, and prevalence of age-associated noncommunicable comorbidities.
Collapse
|
35
|
Hodges G, Lyngbæk S, Selmer C, Ahlehoff O, Theilade S, Sehestedt TB, Abildgaard U, Eugen-Olsen J, Galløe AM, Hansen PR, Jeppesen JL, Bang CN. SuPAR is associated with death and adverse cardiovascular outcomes in patients with suspected coronary artery disease. SCAND CARDIOVASC J 2020; 54:339-345. [DOI: 10.1080/14017431.2020.1762917] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Affiliation(s)
- Gethin Hodges
- Department of Cardiology, Copenhagen University Hospital Herlev and Gentofte, Copenhagen, Denmark
| | - Stig Lyngbæk
- Department of Medicine Glostrup, Amager Hvidvore Hospital Glostrup, University of Copenhagen, Copenhagen, Denmark
| | - Christian Selmer
- Department of Endocrinology, Bispebjerg and Frederiksberg Hospital, University of Copenhagen, Copenhagen, Denmark
| | - Ole Ahlehoff
- Department of Cardiology, Odense University Hospital, Odense, Denmark
| | | | - Thomas Berend Sehestedt
- Department of Cardiology, Bispebjerg and Frederiksberg Hospital, University of Copenhagen, Copenhagen, Denmark
| | - Ulrik Abildgaard
- Department of Cardiology, Copenhagen University Hospital Herlev and Gentofte, Copenhagen, Denmark
| | - Jesper Eugen-Olsen
- Clinical Research Centre, Copenhagen University Hospital Hvidovre, Hvidovre, Denmark
| | | | - Peter Riis Hansen
- Department of Cardiology, Copenhagen University Hospital Herlev and Gentofte, Copenhagen, Denmark
| | - Jørgen L. Jeppesen
- Department of Medicine Glostrup, Amager Hvidvore Hospital Glostrup, University of Copenhagen, Copenhagen, Denmark
| | - Casper N. Bang
- The Danish Heart Foundation, Copenhagen, Denmark
- Department of Cardiology, North Zealand University Hospital, Hillerød, Denmark
| |
Collapse
|
36
|
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: 1.8] [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
|
37
|
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: 2.8] [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
|
38
|
Roca N, Jatem E, Martín ML, Muñoz M, Molina M, Martínez C, Segarra A. Relationship between soluble urokinase-type plasminogen activator receptor and serum biomarkers of endothelial activation in patients with idiopathic nephrotic syndrome. Clin Kidney J 2020; 14:543-549. [PMID: 33623677 PMCID: PMC7886542 DOI: 10.1093/ckj/sfz173] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2019] [Accepted: 11/08/2019] [Indexed: 11/14/2022] Open
Abstract
Background Serum levels of soluble urokinase-type plasminogen activator receptor (suPAR) are high in some patients with idiopathic nephrotic syndrome (INS). Given that suPAR constitutes a predictor of vascular disease and has been associated with endothelial dysfunction, we hypothesized that suPAR levels are related to endothelial activation or dysfunction in INS patients. The aims of this study were to evaluate the relationship between serum concentrations of endothelial biomarkers and suPAR in patients with different histological patterns of INS and healthy controls, and to determine the demographic, clinical and biochemical characteristics of INS patients that influence suPAR serum levels. Methods This observational, cross-sectional study included patients with INS, diagnosed with minimal change disease (MCD), focal segmental glomerulosclerosis (FSGS) or membranous nephropathy (MN) by renal biopsy. Patient demographic, clinical and biochemical characteristics were recorded and blood samples were obtained at the time of diagnosis. Measurements of suPAR and endothelial molecules via serum levels were performed using Enzyme-Linked ImmunoSorbent Assay kits. Results Patients with nephrotic syndrome (n = 152) caused by FSGS, MCD or MN had increased circulating levels of endothelial markers. suPAR levels positively correlated with age and the serum levels of almost all endothelial markers. Generally, endothelial cell molecules positively correlated with each other. suPAR levels were not associated with the histopathological pattern of INS. Conclusions In patients with INS secondary to FSGS, MCD and NM, circulating levels of suPAR are independent of the primary renal disease, and significantly associated with age, glomerular filtration rate and the levels of various endothelial markers.
Collapse
Affiliation(s)
- Neus Roca
- Servicio de Nefrología Pediátrica, Consorci Hospitalari de Vic, Barcelona, Spain.,Universitat de Vic, Barcelona, Spain
| | - Elias Jatem
- Servicio de Nefrología, Hospital Universitario Arnau de Vilanova, Lleida, Spain.,Vascular & Renal Translational Research Group, IRBLleida, Spanish Research Network for Renal Diseases (RedInRen.ISCIII), Lleida, Spain
| | - María Luisa Martín
- Servicio de Nefrología, Hospital Universitario Arnau de Vilanova, Lleida, Spain.,Vascular & Renal Translational Research Group, IRBLleida, Spanish Research Network for Renal Diseases (RedInRen.ISCIII), Lleida, Spain
| | - Marina Muñoz
- Servicio de Nefrología Pediátrica, Hospital Universitario de Vall d'Hebron, Barcelona, Spain
| | - Maria Molina
- Servicio de Nefrología, Hospital Universitario Arnau de Vilanova, Lleida, Spain.,Vascular & Renal Translational Research Group, IRBLleida, Spanish Research Network for Renal Diseases (RedInRen.ISCIII), Lleida, Spain
| | - Cristina Martínez
- Servicio de Nefrología, Hospital Universitario Arnau de Vilanova, Lleida, Spain.,Vascular & Renal Translational Research Group, IRBLleida, Spanish Research Network for Renal Diseases (RedInRen.ISCIII), Lleida, Spain
| | - Alfons Segarra
- Servicio de Nefrología, Hospital Universitario Arnau de Vilanova, Lleida, Spain.,Vascular & Renal Translational Research Group, IRBLleida, Spanish Research Network for Renal Diseases (RedInRen.ISCIII), Lleida, Spain
| |
Collapse
|
39
|
Association between Soluble Urokinase-Type Plasminogen Activator Receptor Levels and Chronic Kidney Disease: A Systematic Review and Meta-Analysis. BIOMED RESEARCH INTERNATIONAL 2019; 2019:6927456. [PMID: 31886242 PMCID: PMC6899318 DOI: 10.1155/2019/6927456] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/27/2019] [Revised: 09/02/2019] [Accepted: 09/24/2019] [Indexed: 12/17/2022]
Abstract
Background Chronic kidney disease (CKD) has become a global public health problem with a high prevalence and mortality. There is no sensitive and effective markers for chronic kidney disease. Previous studies proposed suPAR as an early predict biomarker for chronic kidney disease, but the results are controversial. Therefore, the purpose of the current meta-analysis is to evaluate the association between suPAR and CKD. Methods We searched the PubMed, Embase, Cochrane Library databases, and Web of Science before May 1, 2019. The search was based on the key words including suPAR and CKD. Data are extracted independently according to standard format, and quality analysis is performed. We extracted the concentration of suPAR and hazard rate (HR) values of mortality, cardiovascular disease, and end-stage renal disease. Results There were 14 studies fulfilling the criteria. The concentration of suPAR was higher in patients with CKD than that in the control group (P < 0.001; SMD: −2.17; 95% CI: −2.71, −1.63; I2 = 67.4%). SuPAR had a higher risk of mortality (P=0.001; HR: 1.72; 95% CI: 1.24, 2.39; I2 = 68.0%). The higher suPAR level increased the risk of cardiovascular disease (P < 0.001; HR: 3.06; 95% CI: 2.21, 4.22; I2 = 0.0%) and the risk of end-stage renal disease (P < 0.001; HR: 1.40; 95% CI: 1.22, 1.60; I2 = 0.0%). Conclusions Monitoring suPAR concentrations may be used for early diagnosis and prognosis for patients with CKD, and the higher suPAR increased the risk of mortality, cardiovascular events, and end-stage renal disease.
Collapse
|
40
|
Pedersen LR, Olsen RH, Anholm C, Astrup A, Eugen-Olsen J, Fenger M, Simonsen L, Walzem RL, Haugaard SB, Prescott E. Effects of 1 year of exercise training versus combined exercise training and weight loss on body composition, low-grade inflammation and lipids in overweight patients with coronary artery disease: a randomized trial. Cardiovasc Diabetol 2019; 18:127. [PMID: 31575375 PMCID: PMC6774219 DOI: 10.1186/s12933-019-0934-x] [Citation(s) in RCA: 52] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/15/2019] [Accepted: 09/21/2019] [Indexed: 12/18/2022] Open
Abstract
Background Dyslipidaemia and low-grade inflammation are central in atherogenesis and linked to overweight and physical inactivity. Lifestyle changes are important in secondary prevention of coronary artery disease (CAD). We compared the effects of combined weight loss and interval training with interval training alone on physical fitness, body composition, dyslipidaemia and low-grade inflammation in overweight, sedentary participants with CAD. Methods Seventy CAD patients, BMI 28–40 kg/m2 and age 45–75 years were randomised to (1) 12 weeks’ aerobic interval training (AIT) at 90% of peak heart rate three times/week followed by 40 weeks’ AIT twice weekly or (2) a low energy diet (LED) (800–1000 kcal/day) for 8–10 weeks followed by 40 weeks’ weight maintenance including AIT twice weekly and a high-protein/low-glycaemic load diet. Effects of the intervention were evaluated by physical fitness, body weight and composition. Dyslipidaemia was described using both biochemical analysis of lipid concentrations and lipoprotein particle subclass distribution determined by density profiling. Low-grade inflammation was determined by C-reactive protein, soluble urokinase-type plasminogen activator receptor and tumour necrosis factor α. Effects on continuous outcomes were tested by mixed-models analysis. Results Twenty-six (74%) AIT and 29 (83%) LED + AIT participants completed the study. At baseline subject included 43 (78%) men; subjects averages were: age 63 years (6.2), body weight 95.9 kg (12.2) and VO2peak 20.7 mL O2/kg/min (4.9). Forty-six (84%) had pre-diabetes (i.e. impaired fasting glucose and/or impaired glucose tolerance). LED + AIT reduced body weight by 7.2 kg (− 8.4; − 6.1) and waist circumference by 6.6 cm (− 7.7; − 5.5) compared to 1.7 kg (− 0.7; − 2.6) and 3.3 cm (− 5.1; − 1.5) after AIT (within-group p < 0.001, between-group p < 0.001 and p = 0.018, respectively). Treatments caused similar changes in VO2peak and lowering of total cholesterol, triglycerides, non-HDL cholesterol and low-grade inflammation. A shift toward larger HDL particles was seen following LED + AIT while AIT elicited no change. Conclusions Both interventions were feasible. Both groups obtained improvements in VO2peak, serum-lipids and inflammation with superior weight loss and greater central fat loss following LED + AIT. Combined LED induced weight loss and exercise can be recommended to CAD patients. Trial registration NCT01724567, November 12, 2012, retrospectively registered (enrolment ended in April 2013).
Collapse
Affiliation(s)
- Lene Rørholm Pedersen
- Department of Cardiology, Bispebjerg University Hospital, University of Copenhagen, Building 67, 1st, Bispebjerg Bakke 23, 2400, Copenhagen, NW, Denmark. .,Department of Cardiology, University Hospital of Zealand, Roskilde, University of Copenhagen, Copenhagen, Denmark.
| | - Rasmus Huan Olsen
- Department of Cardiology, Bispebjerg University Hospital, University of Copenhagen, Building 67, 1st, Bispebjerg Bakke 23, 2400, Copenhagen, NW, Denmark
| | - Christian Anholm
- Department of Internal Medicine, Glostrup University Hospital, University of Copenhagen, Copenhagen, Denmark
| | - Arne Astrup
- Department of Nutrition, Exercise and Sports, University of Copenhagen, Copenhagen, Denmark
| | - Jesper Eugen-Olsen
- Clinical Research Centre, Hvidovre University Hospital, University of Copenhagen, Copenhagen, Denmark
| | - Mogens Fenger
- Department of Medical Biochemistry, Genetics and Molecular Biochemistry, Hvidovre University Hospital, University of Copenhagen, Copenhagen, Denmark
| | - Lene Simonsen
- Department of Clinical Physiology and Nuclear Medicine, Bispebjerg University Hospital, University of Copenhagen, Copenhagen, Denmark
| | - Rosemary L Walzem
- Faculty of Nutrition, Texas A & M University, College Station, TX, USA
| | - Steen Bendix Haugaard
- Department of Internal Medicine, Amager and Hvidovre University Hospital, University of Copenhagen, Copenhagen, Denmark
| | - Eva Prescott
- Department of Cardiology, Bispebjerg University Hospital, University of Copenhagen, Building 67, 1st, Bispebjerg Bakke 23, 2400, Copenhagen, NW, Denmark
| |
Collapse
|
41
|
Review of serum biomarkers in carotid atherosclerosis. J Vasc Surg 2019; 71:329-341. [PMID: 31327598 DOI: 10.1016/j.jvs.2019.04.488] [Citation(s) in RCA: 51] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2018] [Accepted: 04/23/2019] [Indexed: 01/23/2023]
Abstract
BACKGROUND Carotid artery atherosclerotic stenosis is a preventable major cause of stroke, but there is still a need for definition of high-risk plaque in asymptomatic patients who might benefit from interventional therapies. Several image markers are recommended to characterize unstable plaques. The measurement of serum biomarkers is a promising method to assist in decision making, but the lack of robust evidence in the carotid environment burdens their potential as a standard of care. The goal of this review was to offer an updated state-of-the-art study of available serum biomarkers with clinical implications, with focus on those that may predict carotid symptom development. METHODS The Cochrane Library and MEDLINE databases were searched (all until September 2018) for studies on carotid plaque and serum biomarkers of atherosclerosis. Nonhuman, basic science, and histology studies were excluded, focusing on clinical studies. Selected abstracts were screened to include the most relevant articles on atherosclerotic plaque presence, progression, instability or symptom development. RESULTS Some well-established biomarkers for coronary disease are not relevant to carotid atherosclerosis and other inflammatory biomarkers, lipids, interleukins, homocysteine, and adipokines may be useful in quantifying carotid disease-related risk. Some serum biomarkers combined with image features may assist vascular specialists in selecting patients at high risk for stroke and in need of intervention. CONCLUSIONS Prospective studies applying a combination of biomarkers are essential to prove clinical usefulness.
Collapse
|
42
|
Becher T, Seiler L, Rudic B, Röger S, Tülümen E, Liebe V, Kuschyk J, Trinkmann F, Michels J, Weiss C, Akin I, Kälsch T, Borggrefe M, Stach K. Cardioprotective Effects of Dronedarone Mediated by the Influence on the Expression of Urokinase-Type Plasminogen Activator Receptor. J Vasc Res 2019; 56:92-96. [PMID: 31079118 DOI: 10.1159/000499526] [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: 05/09/2018] [Accepted: 03/08/2019] [Indexed: 11/19/2022] Open
Abstract
PURPOSE Dronedarone is a multichannel-blocking antiarrhythmic drug for the treatment of atrial fibrillation. Observational data hypothesized a cardioprotective effect. In an in vitro endothelial cell-platelet model, we evaluated the molecular atheroprotective effects of dronedarone. METHODS Following a 24-h incubation of human umbilical vein endothelial cells (HUVECs) with dronedarone (concentration 50, 100, and 150 ng/mL), they were then stimulated for 1 h with lipopolysaccharide (LPS) and were subsequently incubated in direct contact with thrombin-activated platelets. After incubation, the expression of CD40L and CD62P on platelets, and the expression of ICAM-1, VCAM-1, urokinase-type plasminogen activator receptor (uPAR), and membrane type 1 matrix metalloproteinase (MT1-MMP) on endothelial cells were measured by flow cytometry. RESULTS Preincubation with 150 ng/mL of dronedarone reduced the expression of uPAR on endothelial cells after proinflammatory stimulation with LPS and also by direct endothelial contact with activated platelets (p = 0.0038). In contrast, the expression of CD40L and CD62P on platelets after proinflammatory stimulation with thrombin was significantly increased through direct preincubation with 50/100/150 ng/mL of dronedarone. However, dronedarone had no effects on the expression of MT1-MMP and ICAM-1 in HUVECs. CONCLUSION In this in vitro analysis, dronedarone directly increased platelet activation but showed significant direct effects on endothelial cells and indirect effects on platelets on selected markers of atherosclerosis.
Collapse
Affiliation(s)
- Tobias Becher
- First Department of Medicine, University Medical Centre Mannheim (UMM), Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
| | - Lea Seiler
- First Department of Medicine, University Medical Centre Mannheim (UMM), Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
| | - Boris Rudic
- First Department of Medicine, University Medical Centre Mannheim (UMM), Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany.,DZHK (German Centre of Cardiovascular Research), Partner Site Heidelberg/Mannheim, Mannheim, Germany
| | - Susanne Röger
- First Department of Medicine, University Medical Centre Mannheim (UMM), Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany.,DZHK (German Centre of Cardiovascular Research), Partner Site Heidelberg/Mannheim, Mannheim, Germany
| | - Erol Tülümen
- First Department of Medicine, University Medical Centre Mannheim (UMM), Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany.,DZHK (German Centre of Cardiovascular Research), Partner Site Heidelberg/Mannheim, Mannheim, Germany
| | - Volker Liebe
- First Department of Medicine, University Medical Centre Mannheim (UMM), Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
| | - Jürgen Kuschyk
- First Department of Medicine, University Medical Centre Mannheim (UMM), Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
| | - Frederik Trinkmann
- First Department of Medicine, University Medical Centre Mannheim (UMM), Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
| | - Julia Michels
- First Department of Medicine, University Medical Centre Mannheim (UMM), Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
| | - Christel Weiss
- Department of Medical Statistics, University Medical Centre Mannheim, Medical Faculty Mannheim, Mannheim, Germany
| | - Ibrahim Akin
- First Department of Medicine, University Medical Centre Mannheim (UMM), Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany.,DZHK (German Centre of Cardiovascular Research), Partner Site Heidelberg/Mannheim, Mannheim, Germany
| | - Thorsten Kälsch
- University Medical Centre Mannheim, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
| | - Martin Borggrefe
- First Department of Medicine, University Medical Centre Mannheim (UMM), Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany.,DZHK (German Centre of Cardiovascular Research), Partner Site Heidelberg/Mannheim, Mannheim, Germany
| | - Ksenija Stach
- First Department of Medicine, University Medical Centre Mannheim (UMM), Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany, .,DZHK (German Centre of Cardiovascular Research), Partner Site Heidelberg/Mannheim, Mannheim, Germany,
| |
Collapse
|
43
|
Sörensen NA, Nikorowitsch J, Neumann JT, Rübsamen N, Goßling A, Hartikainen TS, Blankenberg S, Westermann D, Zeller T, Karakas M. Predictive value of soluble urokinase-type plasminogen activator receptor for mortality in patients with suspected myocardial infarction. Clin Res Cardiol 2019; 108:1386-1393. [PMID: 30989318 PMCID: PMC6867986 DOI: 10.1007/s00392-019-01475-1] [Citation(s) in RCA: 10] [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: 02/04/2019] [Accepted: 04/04/2019] [Indexed: 12/14/2022]
Abstract
BACKGROUND Early risk stratification of patients with suspected acute myocardial infarction (AMI) constitutes an unmet need in current daily clinical practice. We aimed to evaluate the predictive value of soluble urokinase-type plasminogen activator receptor (suPAR) levels for 1-year mortality in patients with suspected AMI. METHODS AND RESULTS suPAR levels were determined in 1314 patients presenting to the emergency department with suspected AMI. Patients were followed up for 12 months to assess all-cause mortality. Of 1314 patients included, 308 were diagnosed with AMI. Median suPAR levels did not differ between subjects with AMI compared to non-AMI (3.5 ng/ml vs. 3.2 ng/ml, p = 0.066). suPAR levels reliably predicted all-cause mortality after 1 year. Hazard ratio for 1-year mortality was 12.6 (p < 0.001) in the quartile with the highest suPAR levels compared to the first quartile. The prognostic value for 6-month mortality was comparable to an established risk prediction model, the Global Registry of Acute Coronary Events (GRACE) score, with an AUC of 0.79 (95% CI 0.72-0.86) for the GRACE score and 0.77 (95% CI 0.69-0.84) for suPAR. Addition of suPAR improved the GRACE score, as shown by integrated discrimination improvement statistics of 0.036 (p = 0.03) suggesting a further discrimination of events from non-events by the addition of suPAR. CONCLUSIONS suPAR levels reliably predicted mortality in patients with suspected AMI. STUDY REGISTRATION http://www.clinicaltrials.gov (NCT02355457).
Collapse
Affiliation(s)
- Nils A Sörensen
- Department of General and Interventional Cardiology, University Heart Center Hamburg Eppendorf, Martinistr. 52, 20246, Hamburg, Germany.,German Center for Cardiovascular Research (DZHK), Partner Site Hamburg/Kiel/Lübeck, Hamburg, Germany
| | - Julius Nikorowitsch
- Department of General and Interventional Cardiology, University Heart Center Hamburg Eppendorf, Martinistr. 52, 20246, Hamburg, Germany
| | - Johannes T Neumann
- Department of General and Interventional Cardiology, University Heart Center Hamburg Eppendorf, Martinistr. 52, 20246, Hamburg, Germany.,German Center for Cardiovascular Research (DZHK), Partner Site Hamburg/Kiel/Lübeck, Hamburg, Germany
| | - Nicole Rübsamen
- Department of General and Interventional Cardiology, University Heart Center Hamburg Eppendorf, Martinistr. 52, 20246, Hamburg, Germany
| | - Alina Goßling
- Department of General and Interventional Cardiology, University Heart Center Hamburg Eppendorf, Martinistr. 52, 20246, Hamburg, Germany
| | - Tau S Hartikainen
- Department of General and Interventional Cardiology, University Heart Center Hamburg Eppendorf, Martinistr. 52, 20246, Hamburg, Germany
| | - Stefan Blankenberg
- Department of General and Interventional Cardiology, University Heart Center Hamburg Eppendorf, Martinistr. 52, 20246, Hamburg, Germany.,German Center for Cardiovascular Research (DZHK), Partner Site Hamburg/Kiel/Lübeck, Hamburg, Germany
| | - Dirk Westermann
- Department of General and Interventional Cardiology, University Heart Center Hamburg Eppendorf, Martinistr. 52, 20246, Hamburg, Germany.,German Center for Cardiovascular Research (DZHK), Partner Site Hamburg/Kiel/Lübeck, Hamburg, Germany
| | - Tanja Zeller
- Department of General and Interventional Cardiology, University Heart Center Hamburg Eppendorf, Martinistr. 52, 20246, Hamburg, Germany.,German Center for Cardiovascular Research (DZHK), Partner Site Hamburg/Kiel/Lübeck, Hamburg, Germany
| | - Mahir Karakas
- Department of General and Interventional Cardiology, University Heart Center Hamburg Eppendorf, Martinistr. 52, 20246, Hamburg, Germany. .,German Center for Cardiovascular Research (DZHK), Partner Site Hamburg/Kiel/Lübeck, Hamburg, Germany.
| |
Collapse
|
44
|
Topel ML, Sandesara PB, Stahl EP, Hayek SS, Tahhan AS, O'Neal WT, Ko YA, Alkhoder A, Gafeer MM, Kim JH, Wilson PWF, Shaw LJ, Epstein SE, Vaccarino V, Sperling LS, Quyyumi AA. Mechanisms underlying the J-curve for diastolic blood pressure: Subclinical myocardial injury and immune activation. Int J Cardiol 2019; 276:255-260. [PMID: 30217423 PMCID: PMC6324952 DOI: 10.1016/j.ijcard.2018.09.028] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/19/2018] [Revised: 08/28/2018] [Accepted: 09/07/2018] [Indexed: 12/18/2022]
Abstract
BACKGROUND Low diastolic blood pressure (DBP) is associated with increased risk of cardiovascular events. In patients with coronary artery disease (CAD), limitations in coronary blood flow and immune activity are implicated mechanisms, but evidence is lacking. We investigated the association between DBP, biomarkers of myocardial injury, inflammation, immune activation and incident events in patients with CAD. METHODS We studied 2448 adults (mean age 65 ± 12 years, 68% male, median follow-up 4.5 years) with CAD. DBP was categorized into 10 mm Hg increments. Biomarkers of myocardial injury (high sensitivity cardiac troponin-I [hs-cTnI]) and immune activity/inflammation (soluble urokinase plasminogen activator receptor [suPAR]) were dichotomized at their median values. DBP 70-79 mm Hg was used as the referent group, and individuals were followed prospectively for adverse outcomes. RESULTS After adjusting for demographic and clinical covariates, individuals with DBP < 60 mm Hg had increased odds of elevated levels of hs-cTnI (OR = 1.68; 95% CI = 1.07, 2.65) and suPAR (OR = 1.71; 95% CI = 1.10, 2.65) compared to the referent group. Additionally, DBP < 60 mm Hg was associated with increased adjusted risk of cardiovascular death or MI (HR = 2.04; 95% CI = 1.32, 3.16) and all-cause mortality (HR = 2.41; 95% CI = 1.69, 3.45). CONCLUSION In patients with CAD, DBP < 60 mm Hg is associated with subclinical myocardial injury, immune/inflammatory dysregulation and incident events. Aggressive BP control may be harmful in these patients, and further investigation is warranted to determine appropriate BP targets in patients with CAD.
Collapse
Affiliation(s)
- Matthew L Topel
- Emory Clinical Cardiovascular Research Institute, Division of Cardiology, Emory University School of Medicine, Atlanta, GA, United States of America.
| | - Pratik B Sandesara
- Emory Clinical Cardiovascular Research Institute, Division of Cardiology, Emory University School of Medicine, Atlanta, GA, United States of America
| | - Eric P Stahl
- Emory Clinical Cardiovascular Research Institute, Division of Cardiology, Emory University School of Medicine, Atlanta, GA, United States of America
| | - Salim S Hayek
- Emory Clinical Cardiovascular Research Institute, Division of Cardiology, Emory University School of Medicine, Atlanta, GA, United States of America
| | - Ayman Samman Tahhan
- Emory Clinical Cardiovascular Research Institute, Division of Cardiology, Emory University School of Medicine, Atlanta, GA, United States of America
| | - Wesley T O'Neal
- Emory Clinical Cardiovascular Research Institute, Division of Cardiology, Emory University School of Medicine, Atlanta, GA, United States of America
| | - Yi-An Ko
- Department of Biostatistics and Bioinformatics, Emory University, Atlanta, GA, United States of America
| | - Ayman Alkhoder
- Emory Clinical Cardiovascular Research Institute, Division of Cardiology, Emory University School of Medicine, Atlanta, GA, United States of America
| | - Mohamad Mazen Gafeer
- Emory Clinical Cardiovascular Research Institute, Division of Cardiology, Emory University School of Medicine, Atlanta, GA, United States of America
| | - Jonathan H Kim
- Emory Clinical Cardiovascular Research Institute, Division of Cardiology, Emory University School of Medicine, Atlanta, GA, United States of America
| | - Peter W F Wilson
- Emory Clinical Cardiovascular Research Institute, Division of Cardiology, Emory University School of Medicine, Atlanta, GA, United States of America
| | - Leslee J Shaw
- Emory Clinical Cardiovascular Research Institute, Division of Cardiology, Emory University School of Medicine, Atlanta, GA, United States of America
| | - Stephen E Epstein
- MedStar Heart and Vascular Institute, MedStar Washington Hospital Center, Washington, DC, United States of America
| | - Viola Vaccarino
- Rollins School of Public Health, Department of Epidemiology, Emory University School of Medicine, Atlanta, GA, United States of America
| | - Laurence S Sperling
- Emory Clinical Cardiovascular Research Institute, Division of Cardiology, Emory University School of Medicine, Atlanta, GA, United States of America
| | - Arshed A Quyyumi
- Emory Clinical Cardiovascular Research Institute, Division of Cardiology, Emory University School of Medicine, Atlanta, GA, United States of America
| |
Collapse
|
45
|
Volpe M, Battistoni A, Gallo G, Carnevale D. The "hidden side of the moon" in hypertension: When and why is dangerous low diastolic blood pressure? Int J Cardiol 2019; 276:268-270. [PMID: 30360994 DOI: 10.1016/j.ijcard.2018.10.041] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/09/2018] [Accepted: 10/10/2018] [Indexed: 11/19/2022]
Affiliation(s)
- Massimo Volpe
- Department of Clinical and Molecular Medicine, School of Medicine and Psychology, Sapienza University of Rome, Rome, Italy; IRCCS Neuromed, Pozzilli, Italy.
| | - Allegra Battistoni
- Department of Clinical and Molecular Medicine, School of Medicine and Psychology, Sapienza University of Rome, Rome, Italy
| | - Giovanna Gallo
- Department of Clinical and Molecular Medicine, School of Medicine and Psychology, Sapienza University of Rome, Rome, Italy
| | - Daniela Carnevale
- IRCCS Neuromed, Pozzilli, Italy; Department of Molecular Medicine, Sapienza University of Rome, Italy
| |
Collapse
|
46
|
Jirak P, Mirna M, Wernly B, Paar V, Thieme M, Betge S, Franz M, Hoppe U, Lauten A, Kammler J, Schulze PC, Lichtenauer M, Kretzschmar D. Analysis of novel cardiovascular biomarkers in patients with peripheral artery disease. Minerva Med 2018; 109:443-450. [PMID: 29652038 DOI: 10.23736/s0026-4806.18.05628-8] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
BACKGROUND Peripheral artery disease (PAD) is a common form of manifestation of atherosclerosis. PAD has a considerable impact on morbidity, hospitalization rates and health-care costs. Biomarkers have been introduced in many cardiovascular disease entities over the last years. However, an analysis on the correlation of biomarker levels and PAD is still lacking. METHODS A total of 106 patients were enrolled in this current study, 51 that were diagnosed with PAD and 55 with excluded coronary and peripheral artery disease as controls. During outpatient visits, plasma samples of all patients were obtained and analyzed for sST2 (hemodynamics and inflammation), galectin-3 (fibrosis and remodeling), GDF-15 (remodeling and inflammation), suPAR (inflammation), and fetuin-A (vascular calcification) by use of ELISA after informed consent. RESULTS Compared with controls, patients with PAD showed significantly higher levels of sST2 (5248 vs. 7503 pg/mL, P<0.001), suPAR (2267 vs. 2414 pg/mL, P=0.02), galectin-3 (2795 vs. 4494 pg/mL, P<0.001), and GDF-15 (549 vs. 767 pg/mL, P<0.001). Fetuin-A showed a trend towards lower levels in patients with PAD (117 vs. 100 ng/mL, P=0.119). CONCLUSIONS Circulating levels of sST2, suPAR, galectin-3, and GDF-15 were significantly elevated in PAD patients. In contrast, fetuin-A levels showed a decrease in PAD patients indicating increased vascular calcification. Thus, by incorporating different pathophysiological processes present in PAD, tested novel biomarkers facilitate a more precise diagnosis as well as a more accurate evaluation of disease severity and progression.
Collapse
Affiliation(s)
- Peter Jirak
- Department of Cardiology, Clinic of Internal Medicine II, Paracelsus Medical University, Salzburg, Austria
| | - Moritz Mirna
- Department of Cardiology, Clinic of Internal Medicine II, Paracelsus Medical University, Salzburg, Austria
| | - Bernhard Wernly
- Department of Cardiology, Clinic of Internal Medicine II, Paracelsus Medical University, Salzburg, Austria
| | - Vera Paar
- Department of Cardiology, Clinic of Internal Medicine II, Paracelsus Medical University, Salzburg, Austria
| | - Marcus Thieme
- Department of Cardiology, Clinic of Internal Medicine I, University Heart Center Thüringen, Friedrich Schiller University, Jena, Germany
| | - Stefan Betge
- Department of Cardiology, Clinic of Internal Medicine I, University Heart Center Thüringen, Friedrich Schiller University, Jena, Germany
| | - Marcus Franz
- Department of Cardiology, Clinic of Internal Medicine I, University Heart Center Thüringen, Friedrich Schiller University, Jena, Germany
| | - Uta Hoppe
- Department of Cardiology, Clinic of Internal Medicine II, Paracelsus Medical University, Salzburg, Austria
| | - Alexander Lauten
- Department of Cardiology, Charité Medical University, Berlin, Germany.,German Center for Cardiovascular Research (DZHK), Berlin, Germany
| | - Jürgen Kammler
- Department of Cardiology, Kepler University Hospital, Linz, Austria
| | - Paul C Schulze
- Department of Cardiology, Clinic of Internal Medicine I, University Heart Center Thüringen, Friedrich Schiller University, Jena, Germany
| | - Michael Lichtenauer
- Department of Cardiology, Clinic of Internal Medicine II, Paracelsus Medical University, Salzburg, Austria -
| | - Daniel Kretzschmar
- Department of Cardiology, Clinic of Internal Medicine I, University Heart Center Thüringen, Friedrich Schiller University, Jena, Germany
| |
Collapse
|
47
|
Onatsu J, Taina M, Mustonen P, Hedman M, Muuronen A, Arponen O, Korhonen M, Jäkälä P, Vanninen R, Pulkki K. Soluble Urokinase-type Plasminogen Activator Receptor Predicts All-cause 5-Year Mortality in Ischemic Stroke and TIA. ACTA ACUST UNITED AC 2018; 31:381-386. [PMID: 28438866 DOI: 10.21873/invivo.11070] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2017] [Revised: 02/26/2017] [Accepted: 03/01/2017] [Indexed: 12/26/2022]
Abstract
AIM We evaluated soluble urokinase-type plasminogen activator receptor (suPAR) levels in different stroke subtypes and assessed their prognostic value regarding 5-year outcomes. MATERIALS AND METHODS The study included 117 stroke patients (81 males; mean=age 61±11 years) with suspected cardioembolic stroke whose plasma suPAR concentration was assessed. Altogether, 20 (17.1%) patients suffered from stroke as a result of cardioembolism, 12 (10.3%) from large-artery atherosclerosis, 9 (7.7%) from small-vessel disease, 11 (9.4%) from both large-artery and cardioembolic etiology, and 65 (55.6%) had cryptogenic stroke. The mean follow-up period was 5 years. RESULTS suPAR concentration was higher in patients who suffered from stroke/transient ischemic attack due to large-artery atherosclerosis (3.2±0.9 ng/ml) compared to small-vessel disease (2.0±0.5 ng/ml, p<0.001). An elevated plasma suPAR concentration was associated with all-cause mortality during the follow-up period (p=0.003). CONCLUSION Elevated plasma suPAR concentrations predicted all-cause mortality during the 5-year follow-up after ischemic stroke. suPAR was not able to differentiate patients with cardioembolic stroke from those with other stroke types.
Collapse
Affiliation(s)
- Juha Onatsu
- Department of Neurology, NeuroCenter, Kuopio University Hospital, Kuopio, Finland
| | - Mikko Taina
- Department of Clinical Radiology, Kuopio University Hospital, Kuopio, Finland
| | - Pirjo Mustonen
- Department of Cardiology, Keski-Suomi Central Hospital, Jyväskylä, Finland
| | - Marja Hedman
- Department of Clinical Radiology, Kuopio University Hospital, Kuopio, Finland
| | - Antti Muuronen
- Department of Clinical Radiology, Kuopio University Hospital, Kuopio, Finland
| | - Otso Arponen
- Department of Clinical Radiology, Kuopio University Hospital, Kuopio, Finland
| | - Miika Korhonen
- Department of Clinical Radiology, Kuopio University Hospital, Kuopio, Finland
| | - Pekka Jäkälä
- Department of Neurology, NeuroCenter, Kuopio University Hospital, Kuopio, Finland.,Unit of Neurology, Institute of Clinical Medicine, University of Eastern Finland, Kuopio, Finland
| | - Ritva Vanninen
- Department of Clinical Radiology, Kuopio University Hospital, Kuopio, Finland.,Department of Clinical Radiology, University of Eastern Finland, Kuopio, Finland
| | - Kari Pulkki
- Department of Clinical Chemistry, University of Eastern Finland, Kuopio, Finland.,Eastern Finland Laboratory Centre, Kuopio, Finland
| |
Collapse
|
48
|
Lv JX, Kong Q, Ma X. Current advances in circulating inflammatory biomarkers in atherosclerosis and related cardio-cerebrovascular diseases. Chronic Dis Transl Med 2017; 3:207-212. [PMID: 29354803 PMCID: PMC5747494 DOI: 10.1016/j.cdtm.2017.09.002] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2017] [Indexed: 12/30/2022] Open
Abstract
Atherosclerosis (AS) is a systemic chronic disease affecting both the coronary and cerebral arteries. Inflammation plays a key role in the initiation and progression of AS, and numerous inflammatory factors have been proposed as potential biomarkers. This article reviews recent research in studies on major circulating inflammatory biomarkers to identify surrogates that may reflect processes associated with AS development and the risk of AS-related vascular events, such as Von Willebrand factor, lectin-like oxidized low-density-lipoprotein receptor-1, soluble urokinase plasminogen activator receptor, regulated upon activation, normal T-cell expressed and secreted, and microparticles, which may provide new perspectives for clinical AS evaluation and risk stratification.
Collapse
Affiliation(s)
- Jun-Xuan Lv
- Department of Neurology, Xuanwu Hospital of Capital Medical University, National Clinical Research Center for Geriatric Disorders, Clinical Center for Cardio-cerebrovascular Disease Capital Medical University, Beijing 100053, China
| | - Qi Kong
- Department of Neurology, Xuanwu Hospital of Capital Medical University, National Clinical Research Center for Geriatric Disorders, Clinical Center for Cardio-cerebrovascular Disease Capital Medical University, Beijing 100053, China
| | - Xin Ma
- Department of Neurology, Xuanwu Hospital of Capital Medical University, National Clinical Research Center for Geriatric Disorders, Clinical Center for Cardio-cerebrovascular Disease Capital Medical University, Beijing 100053, China
| |
Collapse
|
49
|
Soluble Urokinase Plasminogen Activator Receptor and the Risk of Coronary Artery Disease in Young Chinese Patients. DISEASE MARKERS 2017; 2017:4719403. [PMID: 29109596 PMCID: PMC5646332 DOI: 10.1155/2017/4719403] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 09/10/2017] [Accepted: 09/20/2017] [Indexed: 01/14/2023]
Abstract
Background Soluble urokinase plasminogen activator receptor (suPAR) is a novel marker of chronic inflammation and is considered to be a risk factor for coronary artery disease (CAD) in Caucasians. This study investigated the role of suPAR in young Chinese patients with CAD. Methods The study involved a total of 196 consecutive young (age ≤ 55 years) patients with angiographically proven CAD and 188 age-matched non-CAD individuals as controls. Traditional risk factors were evaluated using conventional assays, and levels of suPAR were measured by sandwich enzyme-linked immunosorbent assays. Results Levels of suPAR were significantly correlated with age (r = 0.20, P = 0.04), smoking (r = 0.33, P = 0.008), body mass index (r = 0.21, P = 0.03), and high-sensitivity C-reactive protein (hs-CRP; r = 0.31, P = 0.01). Multivariate logistic regression analysis showed that male sex (odds ratio (OR) = 3.12; 95% confidence interval (CI) = 1.18–8.25, P = 0.02), smoking (OR = 3.41, 95% CI = 1.55–7.50, P = 0.002), triglyceride (OR = 1.89, 95% CI = 1.10–3.25, P = 0.02), high-sensitivity C-reactive protein (OR = 1.24, 95% CI = 1.02–0.03, P = 0.03), and suPAR (OR = 1.37, 95% CI = 1.09–1.72, P = 0.007) were independently associated with CAD risk in young patients. Conclusions SuPAR is a novel independent risk factor for CAD in young Chinese patients. Further studies evaluating the effect of anti-inflammatory treatment on the suPAR levels and the risk of CAD are needed.
Collapse
|
50
|
Abstract
Several large-scale genome-wide association studies have identified single-nucleotide polymorphisms in the genomic region of A Disintegrin And Metalloproteinase with ThromboSpondin type 1 repeats (ADAMTS)-7 and associations to coronary artery disease. Experimental studies have provided evidence for a functional role of ADAMTS-7 in both injury-induced vascular neointima formation and development of atherosclerotic lesions. However, whether ADAMTS-7 is associated with a specific plaque phenotype in humans has not been investigated. Carotid plaques (n = 206) from patients with and without cerebrovascular symptoms were analyzed for expression of ADAMTS-7 by immunohistochemistry and correlated to components associated with plaque vulnerability. Plaques from symptomatic patients showed increased levels of ADAMTS-7 compared with lesions from asymptomatic patients. High levels of ADAMTS-7 correlated with high levels of CD68-staining and lipid content, but with low smooth muscle cell and collagen content, which together are characteristics of a vulnerable plaque phenotype. ADAMTS-7 levels above median were associated with increased risk for postoperative cardiovascular events. Our data show that ADAMTS-7 is associated with a vulnerable plaque phenotype in human carotid lesions. These data support previous observations of a potential proatherogenic role of ADAMTS-7.
Collapse
|