1
|
Erixon C, Thelaus L, Johannesson E, Nilsson J, Teurneau-Hermansson K, Linder A, Ragnarsson S, Sterner N, Zindovic I, Dardashti A. The predictive value of postoperative soluble urokinase plasminogen activator receptor concentration for postoperative complications following valvular surgery. Scand J Clin Lab Invest 2025; 85:160-167. [PMID: 40088459 DOI: 10.1080/00365513.2025.2479042] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2024] [Revised: 03/05/2025] [Accepted: 03/10/2025] [Indexed: 03/17/2025]
Abstract
Soluble urokinase plasminogen activator receptor (suPAR) is an inflammatory marker that has been shown to predict poorer outcomes in cardiovascular disease and after cardiac surgery. The relationship between suPAR concentrations and postoperative complications after valvular surgery, however, remains unclear. This study aims to evaluate the predictive value of suPAR concentrations for infection, acute kidney injury (AKI) and prolonged mechanical ventilation after valvular surgery. This prospective, observational, single-centre study included 414 patients who underwent valvular cardiac surgery at Skåne University Hospital between 1 February 2020 and 22 September 2021. Early postoperative suPAR levels were measured, and multivariable logistic regression was used to identify significant risk factors for postoperative infection, AKI and prolonged mechanical ventilation. Left ventricular ejection fraction (LVEF) 30-50% (OR 3.57 [1.29-9.86], p = 0.014) and suPAR concentration (OR 1.41 [1.56-1.71], p <0.001) were found to be predictive risk factors for developing postoperative infection. Additionally, suPAR concentration (OR 1.23 [1.05-1.43], p = 0.008), cardiopulmonary bypass (CPB) time (OR 1.01 [1.00-1.02], p = 0.004) and age (OR 1.04 [1.01-1.08], p = 0.007) were found to be predictive risk factors for postoperative AKI. However, suPAR concentration did not predict prolonged mechanical ventilation. Plasma suPAR levels after cardiac valve surgery were found to be predictive of postoperative AKI and infection. Our results indicate that early postoperative suPAR measurements may be a valuable tool for identifying patients at higher risk for developing postoperative complications.
Collapse
Affiliation(s)
- Clara Erixon
- Department of Cardiothoracic Surgery, Institute of Clinical Sciences, Lund University, Skåne University Hospital, Lund, Sweden
| | - Louise Thelaus
- Department of Infection Medicine, Institute of Department of Clinical Sciences Lund, Faculty of Medicine, Lund University, Lund, Sweden
| | - Emilia Johannesson
- Department of Cardiothoracic Surgery, Institute of Clinical Sciences, Lund University, Skåne University Hospital, Lund, Sweden
| | - Johan Nilsson
- Thoracic Surgery and Bioinformatics Research Unit, Institute of Clinical Sciences, Department of Translational Medicine, Lund University, Lund, Sweden
- Department of Thoracic and Vascular Surgery, Skåne University Hospital, Lund, Sweden
| | - Karl Teurneau-Hermansson
- Department of Cardiothoracic Surgery, Institute of Clinical Sciences, Lund University, Skåne University Hospital, Lund, Sweden
| | - Adam Linder
- Department of Infection Medicine, Institute of Department of Clinical Sciences Lund, Faculty of Medicine, Lund University, Lund, Sweden
| | - Sigurdur Ragnarsson
- Department of Cardiothoracic Surgery, Institute of Clinical Sciences, Lund University, Skåne University Hospital, Lund, Sweden
| | - Niklas Sterner
- Department of Cardiothoracic Surgery, Institute of Clinical Sciences, Lund University, Skåne University Hospital, Lund, Sweden
| | - Igor Zindovic
- Department of Cardiothoracic Surgery, Institute of Clinical Sciences, Lund University, Skåne University Hospital, Lund, Sweden
| | - Alain Dardashti
- Department of Cardiothoracic Surgery, Institute of Clinical Sciences, Lund University, Skåne University Hospital, Lund, Sweden
| |
Collapse
|
2
|
Yang H, Chen Y, He J, Li Y, Feng Y. Advances in the diagnosis of early biomarkers for acute kidney injury: a literature review. BMC Nephrol 2025; 26:115. [PMID: 40045274 PMCID: PMC11884078 DOI: 10.1186/s12882-025-04040-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2025] [Accepted: 02/21/2025] [Indexed: 03/09/2025] Open
Abstract
Acute kidney injury (AKI) is a critical condition with diverse manifestations and variable outcomes. Its diagnosis traditionally relies on delayed indicators such as serum creatinine and urine output, making early detection challenging. Early identification is essential to improving patient outcomes, driving the need for novel biomarkers. Recent advancements have identified promising biomarkers across various biological processes. Tubular injury markers, including neutrophil gelatinase-associated lipocalin (NGAL), kidney injury molecule-1 (KIM-1), N-acetyl-β-D-glucosaminidase (NAG), and liver-type fatty acid-binding protein (L-FABP), offer insights into early tubular damage. Inflammatory and repair-associated biomarkers, such as interleukin-18 (IL-18), monocyte chemotactic protein-1 (MCP-1), osteopontin (OPN), and C-C motif chemokine ligand 14 (CCL14), reflect ongoing injury and recovery processes. Additionally, stress and repair markers like tissue inhibitor of metalloproteinase-2 (TIMP-2) and insulin-like growth factor-binding protein-7 (IGFBP-7), alongside filtration markers such as cystatin C (CysC) and proenkephalin (PenKid®) e.tal, further enhance diagnostic precision. Oxidative stress-related markers, including Superoxide Dismutase 1 (SOD1), also contribute valuable information. Emerging candidates, such as microRNAs, soluble urokinase plasminogen activator receptor (SuPAR), and chitinase-3-like protein 1 (CHI3L1), hold substantial promise for AKI detection and prognosis. This review summarizes the progress in AKI biomarker research, highlighting their clinical utility and exploring their potential to refine early diagnosis and management strategies. These findings offer a new perspective for integrating novel biomarkers into routine clinical practice, ultimately improving AKI care.
Collapse
Affiliation(s)
- Hongsha Yang
- School of Medicine, University of Electronic Science and Technology of China, Chengdu, 610054, China
| | - Yanqin Chen
- School of Medicine, University of Electronic Science and Technology of China, Chengdu, 610054, China
| | - Jiajia He
- School of Medicine, University of Electronic Science and Technology of China, Chengdu, 610054, China
| | - Yi Li
- Department of Nephrology, Institute of Nephrology, Sichuan Provincial People's Hospital, Sichuan Clinical Research Centre for Kidney Diseases, School of Medicine, University of Electronic Science and Technology of China, Chengdu, 610072, China
| | - Yunlin Feng
- Department of Nephrology, Institute of Nephrology, Sichuan Provincial People's Hospital, Sichuan Clinical Research Centre for Kidney Diseases, School of Medicine, University of Electronic Science and Technology of China, Chengdu, 610072, China.
| |
Collapse
|
3
|
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
|
4
|
Louka M, Tatsi EB, Vassiliu S, Theoharis G, Straka K, Filippatos F, Dourdouna MM, Siahanidou T, Syriopoulou V, Michos A. The Soluble Urokinase Plasminogen Activator Receptor as a Severity Biomarker in Children With Acute COVID-19 or Multisystem Inflammatory Syndrome. Pediatr Infect Dis J 2024; 43:477-482. [PMID: 38251905 DOI: 10.1097/inf.0000000000004244] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/23/2024]
Abstract
BACKGROUND Elevated soluble urokinase plasminogen activator receptor (suPAR) has been associated with a poor prognosis in serious infections. The aim of this study was to evaluate the clinical value of suPAR in children with acute coronavirus disease 2019 (COVID-19) or multisystem inflammatory syndrome (MIS-C). METHODS Serum suPAR was measured using the suPARnostic AUTO Flex enzyme-linked immunosorbent assay in hospitalized children with COVID-19, MIS-C, bacterial pneumonia, and healthy controls. RESULTS A total of 211 children with a mean (±SD) age of 6.9 ± 4.96 years were tested; with COVID-19: 59 (28%), MIS-C: 36 (17%), pneumonia: 78 (37%) and healthy controls: 38 (18%). In the acute phase, the levels of suPAR (mean ± SD) were: MIS-C: 8.11 ± 2.80 ng/mL, COVID-19: 4.91 ± 1.90 ng/mL, pneumonia: 4.25 ± 1.44 ng/mL and controls: 2.09 ± 0.47 ng/mL ( P < 0.001). Children with acute COVID-19 and a severe or moderate clinical presentation had higher values than those with mild symptoms: 5.79 ± 1.58 versus 5.40 ± 1.94 versus 3.19 ± 0.73 ng/mL, respectively ( P < 0.001). In the MIS-C group, children hospitalized in the intensive care unit and in need of mechanical ventilation had higher suPAR than those who were not admitted to an intensive care unit: 9.32 ± 3.06 versus 7.13 ± 2.19 ng/mL, respectively ( P = 0.023). In children with COVID-19 or MIS-C, a correlation was detected between suPAR values and length of hospitalization ( rs = 0.418, P < 0.001). CONCLUSIONS The findings suggest that suPAR may be a valuable biomarker of disease severity in children with COVID-19 or MIS-C. This could facilitate the identification of children in need of intensive anti-inflammatory treatment, as it has been shown in adults with severe COVID-19.
Collapse
Affiliation(s)
- Magdalini Louka
- From the First Department of Pediatrics, Infectious Diseases and Chemotherapy Research Laboratory, Medical School, National and Kapodistrian University of Athens, "Aghia Sophia" Children's Hospital, Athens Greece
| | - Elizabeth Barbara Tatsi
- From the First Department of Pediatrics, Infectious Diseases and Chemotherapy Research Laboratory, Medical School, National and Kapodistrian University of Athens, "Aghia Sophia" Children's Hospital, Athens Greece
- University Research Institute of Maternal and Child Health and Precision Medicine, Athens, Greece
| | - Sofia Vassiliu
- From the First Department of Pediatrics, Infectious Diseases and Chemotherapy Research Laboratory, Medical School, National and Kapodistrian University of Athens, "Aghia Sophia" Children's Hospital, Athens Greece
| | - George Theoharis
- From the First Department of Pediatrics, Infectious Diseases and Chemotherapy Research Laboratory, Medical School, National and Kapodistrian University of Athens, "Aghia Sophia" Children's Hospital, Athens Greece
| | - Kalliopi Straka
- Pediatric Intensive Care Unit, "Aghia Sophia" Children's Hospital, Athens, Greece
| | - Filippos Filippatos
- From the First Department of Pediatrics, Infectious Diseases and Chemotherapy Research Laboratory, Medical School, National and Kapodistrian University of Athens, "Aghia Sophia" Children's Hospital, Athens Greece
| | - Maria Myrto Dourdouna
- From the First Department of Pediatrics, Infectious Diseases and Chemotherapy Research Laboratory, Medical School, National and Kapodistrian University of Athens, "Aghia Sophia" Children's Hospital, Athens Greece
| | - Tania Siahanidou
- Neonatal Unit, First Department of Pediatrics, Medical School, National and Kapodistrian University of Athens, "Aghia Sophia" Children's Hospital, Athens, Greece
| | - Vasiliki Syriopoulou
- From the First Department of Pediatrics, Infectious Diseases and Chemotherapy Research Laboratory, Medical School, National and Kapodistrian University of Athens, "Aghia Sophia" Children's Hospital, Athens Greece
| | - Athanasios Michos
- From the First Department of Pediatrics, Infectious Diseases and Chemotherapy Research Laboratory, Medical School, National and Kapodistrian University of Athens, "Aghia Sophia" Children's Hospital, Athens Greece
| |
Collapse
|
5
|
Raina R, Jothi S, Haffner D, Somers M, Filler G, Vasistha P, Chakraborty R, Shapiro R, Randhawa PS, Parekh R, Licht C, Bunchman T, Sethi S, Mangat G, Zaritsky J, Schaefer F, Warady B, Bartosh S, McCulloch M, Alhasan K, Swiatecka-Urban A, Smoyer WE, Chandraker A, Yap HK, Jha V, Bagga A, Radhakrishnan J. Post-transplant recurrence of focal segmental glomerular sclerosis: consensus statements. Kidney Int 2024; 105:450-463. [PMID: 38142038 DOI: 10.1016/j.kint.2023.10.017] [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: 12/22/2021] [Revised: 10/03/2023] [Accepted: 10/17/2023] [Indexed: 12/25/2023]
Abstract
Focal segmental glomerular sclerosis (FSGS) is 1 of the primary causes of nephrotic syndrome in both pediatric and adult patients, which can lead to end-stage kidney disease. Recurrence of FSGS after kidney transplantation significantly increases allograft loss, leading to morbidity and mortality. Currently, there are no consensus guidelines for identifying those patients who are at risk for recurrence or for the management of recurrent FSGS. Our work group performed a literature search on PubMed/Medline, Embase, and Cochrane, and recommendations were proposed and graded for strength of evidence. Of the 614 initially identified studies, 221 were found suitable to formulate consensus guidelines for recurrent FSGS. These guidelines focus on the definition, epidemiology, risk factors, pathogenesis, and management of recurrent FSGS. We conclude that additional studies are required to strengthen the recommendations proposed in this review.
Collapse
Affiliation(s)
- Rupesh Raina
- Department of Nephrology, Akron Nephrology Associates/Cleveland Clinic Akron General Medical Center, Akron, Ohio, USA; Department of Nephrology, Akron Children's Hospital, Akron, Ohio, USA
| | - Swathi Jothi
- Department of Nephrology, Akron Nephrology Associates/Cleveland Clinic Akron General Medical Center, Akron, Ohio, USA
| | - Dieter Haffner
- Department of Pediatric Kidney, Liver and Metabolic Diseases, Hannover Medical School, Hannover, Germany
| | - Michael Somers
- Division of Nephrology, Department of Medicine, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Guido Filler
- Department of Pediatrics, Schulich School of Medicine and Dentistry, University of Western Ontario, London, Ontario, Canada; Department of Medicine, Schulich School of Medicine and Dentistry, University of Western Ontario, London, Ontario, Canada; Department of Pathology and Laboratory Medicine, Schulich School of Medicine and Dentistry, University of Western Ontario, London, Ontario, Canada
| | - Prabhav Vasistha
- Department of Nephrology, Akron Nephrology Associates/Cleveland Clinic Akron General Medical Center, Akron, Ohio, USA
| | - Ronith Chakraborty
- Department of Nephrology, Akron Nephrology Associates/Cleveland Clinic Akron General Medical Center, Akron, Ohio, USA; Department of Nephrology, Akron Children's Hospital, Akron, Ohio, USA
| | - Ron Shapiro
- Recanati/Miller Transplantation Institute, The Mount Sinai Medical Center, New York, New York, USA
| | - Parmjeet S Randhawa
- Department of Pathology, Thomas E Starzl Transplant Institute, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Rulan Parekh
- Department of Medicine, School of Medicine, Johns Hopkins University, Baltimore, Maryland, USA
| | - Christopher Licht
- Division of Pediatric Nephrology, The Hospital for Sick Children, University of Toronto, Toronto, Ontario, Canada
| | - Timothy Bunchman
- Pediatric Nephrology and Transplantation, Children's Hospital of Richmond at Virginia Commonwealth University (VCU), Richmond, Virginia, USA
| | - Sidharth Sethi
- Pediatric Nephrology, Kidney Institute, Medanta, The Medicity Hospital, Gurgaon, Haryana, India
| | - Guneive Mangat
- Department of Nephrology, Akron Nephrology Associates/Cleveland Clinic Akron General Medical Center, Akron, Ohio, USA
| | - Joshua Zaritsky
- Division of Pediatric Nephrology, Nemours, A.I. duPont Hospital for Children, Wilmington, Delaware, USA
| | - Franz Schaefer
- Department of Pediatric Nephrology, University Children's Hospital Heidelberg, Heidelberg, Germany
| | - Bradley Warady
- Division of Nephrology, University of Missouri-Kansas City School of Medicine, Children's Mercy, Kansas City, Missouri, USA
| | - Sharon Bartosh
- Department of Pediatrics, University of Wisconsin Medical School, Madison, Wisconsin, USA
| | - Mignon McCulloch
- Red Cross War Memorial Children's Hospital, University of Cape Town, Cape Town, South Africa
| | - Khalid Alhasan
- Nephrology Unit, Pediatrics Department, College of Medicine, King Saud University, Riyadh, Saudi Arabia; Pediatric Kidney Transplant Division, Organ Transplant Center, King Faisal Specialist Hospital and Research Center, Riyadh, Saudi Arabia
| | - Agnieszka Swiatecka-Urban
- University of Virginia Children's Hospital, University of Virginia School of Medicine, Charlottesville, Virginia, USA
| | - William E Smoyer
- Center for Clinical and Translational Research and Division of Nephrology, Nationwide Children's Hospital, The Ohio State University College of Medicine, Columbus, Ohio, USA
| | - Anil Chandraker
- Transplantation Research Center, Kidney and Pancreas Transplantation, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Hui Kim Yap
- Department of Pediatrics, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore; Khoo Teck Puat-National University Children's Medical Institute, National University Hospital, Singapore, Singapore
| | - Vivekanand Jha
- George Institute for Global Health, University of New South Wales (UNSW), New Delhi, India; School of Public Health, Imperial College, London, UK; Prasanna School of Public Health, Manipal Academy of Higher Education, Manipal, India
| | - Arvind Bagga
- Division of Pediatric Nephrology, All India Institute of Medical Sciences, New Delhi, India
| | - Jai Radhakrishnan
- Department of Medicine (Nephrology), Columbia University Medical Center, New York, New York, USA.
| |
Collapse
|
6
|
Rossiter A, La A, Koyner JL, Forni LG. New biomarkers in acute kidney injury. Crit Rev Clin Lab Sci 2024; 61:23-44. [PMID: 37668397 DOI: 10.1080/10408363.2023.2242481] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2023] [Revised: 05/14/2023] [Accepted: 07/26/2023] [Indexed: 09/06/2023]
Abstract
Acute kidney injury (AKI) is a commonly encountered clinical syndrome. Although it often complicates community acquired illness, it is more common in hospitalized patients, particularly those who are critically ill or who have undergone major surgery. Approximately 20% of hospitalized adult patients develop an AKI during their hospital care, and this rises to nearly 60% in the critically ill, depending on the population being considered. In general, AKI is more common in older adults, in those with preexisting chronic kidney disease and in those with known risk factors for AKI (including diabetes and hypertension). The development of AKI is associated with an increase in both mortality and morbidity, including the development of post-AKI chronic kidney disease. Currently, AKI is defined by a rise in serum creatinine from either a known or derived baseline value and/or oliguria or anuria. However, clinicians may fail to recognize the initial development of AKI because of a delay in the rise of serum creatinine or because of inaccurate urine output monitoring. This, in turn, delays any putative measures to treat AKI or to limit its degree. Consequently, efforts have focused on new biomarkers associated with AKI that may allow early recognition of this syndrome with the intent that this will translate into improved patient outcomes. Here we outline current biomarkers associated with AKI and explore their potential in aiding diagnosis, understanding the pathophysiology and directing therapy.
Collapse
Affiliation(s)
- Adam Rossiter
- Critical Care Unit, Royal Surrey Hospital, Guildford, Surry, UK
| | - Ashley La
- Department of Medicine, University of Chicago, Chicago, Illinois, USA
| | - Jay L Koyner
- Department of Medicine, University of Chicago, Chicago, Illinois, USA
| | - Lui G Forni
- Critical Care Unit, Royal Surrey Hospital, Guildford, Surry, UK
- School of Medicine, Department of Clinical & Experimental Medicine, Faculty of Health Sciences, University of Surrey, Surry, UK
| |
Collapse
|
7
|
Fioretto BS, Rosa I, Matucci-Cerinic M, Romano E, Manetti M. Current Trends in Vascular Biomarkers for Systemic Sclerosis: A Narrative Review. Int J Mol Sci 2023; 24:ijms24044097. [PMID: 36835506 PMCID: PMC9965592 DOI: 10.3390/ijms24044097] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2023] [Revised: 02/13/2023] [Accepted: 02/16/2023] [Indexed: 02/22/2023] Open
Abstract
Systemic sclerosis (SSc, scleroderma) is a multifaceted rare connective tissue disease whose pathogenesis is dominated by immune dysregulation, small vessel vasculopathy, impaired angiogenesis, and both cutaneous and visceral fibrosis. Microvascular impairment represents the initial event of the disease, preceding fibrosis by months or years and accounting for the main disabling and/or life-threatening clinical manifestations, including telangiectasias, pitting scars, periungual microvascular abnormalities (e.g., giant capillaries, hemorrhages, avascular areas, ramified/bushy capillaries) clinically detectable by nailfold videocapillaroscopy, ischemic digital ulcers, pulmonary arterial hypertension, and scleroderma renal crisis. Despite a variety of available treatment options, treatment of SSc-related vascular disease remains problematic, even considering SSc etherogenity and the quite narrow therapeutic window. In this context, plenty of studies have highlighted the great usefulness in clinical practice of vascular biomarkers allowing clinicians to assess the evolution of the pathological process affecting the vessels, as well as to predict the prognosis and the response to therapy. The current narrative review provides an up-to-date overview of the main candidate vascular biomarkers that have been proposed for SSc, focusing on their main reported associations with characteristic clinical vascular features of the disease.
Collapse
Affiliation(s)
- Bianca Saveria Fioretto
- Section of Anatomy and Histology, Department of Experimental and Clinical Medicine, University of Florence, 50134 Florence, Italy
| | - Irene Rosa
- Section of Anatomy and Histology, Department of Experimental and Clinical Medicine, University of Florence, 50134 Florence, Italy
| | - Marco Matucci-Cerinic
- Section of Internal Medicine, Department of Experimental and Clinical Medicine, University of Florence, 50134 Florence, Italy
- Unit of Immunology, Rheumatology, Allergy and Rare Diseases (UnIRAR), IRCCS San Raffaele Hospital, 20132 Milan, Italy
| | - Eloisa Romano
- Section of Internal Medicine, Department of Experimental and Clinical Medicine, University of Florence, 50134 Florence, Italy
| | - Mirko Manetti
- Section of Anatomy and Histology, Department of Experimental and Clinical Medicine, University of Florence, 50134 Florence, Italy
- Imaging Platform, Department of Experimental and Clinical Medicine, University of Florence, 50134 Florence, Italy
- Correspondence:
| |
Collapse
|
8
|
Zhan K, Wang L, Lin H, Fang X, Jia H, Ma X. Novel inflammatory biomarkers in the prognosis of COVID-19. Ther Adv Respir Dis 2023; 17:17534666231199679. [PMID: 37727063 PMCID: PMC10515606 DOI: 10.1177/17534666231199679] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2022] [Accepted: 08/18/2023] [Indexed: 09/21/2023] Open
Abstract
BACKGROUND The central role of inflammatory progression in the development of Coronavirus disease 2019 (COVID-19), especially in severe cases, is indisputable. However, the role of some novel inflammatory biomarkers in the prognosis of COVID-19 remains controversial. OBJECTIVE To assess the effect of some novel inflammatory biomarkers in the occurrence and prognosis of COVID-19. METHODS We systematically retrieved the studies related to COVID-19 and the inflammatory biomarkers of interest. The data of each biomarker in different groups were extracted, then were categorized and pooled. The standardized mean difference was chosen as an effect size measure to compare the difference between groups. RESULTS A total of 90 studies with 12,059 participants were included in this study. We found higher levels of endocan, PTX3, suPAR, sRAGE, galectin-3, and monocyte distribution width (MDW) in the COVID-19 positive groups compared to the COVID-19 negative groups. No significant differences for suPAR and galectin-3 were detected between the severe group and mild/moderate group of COVID-19. In addition, the deaths usually had higher levels of PTX3, sCD14-ST, suPAR, and MDW at admission compared to the survivors. Furthermore, patients with higher levels of endocan, galectin-3, sCD14-ST, suPAR, and MDW usually developed poorer comprehensive clinical prognoses. CONCLUSIONS In summary, this meta-analysis provides the most up-to-date and comprehensive evidence for the role of the mentioned novel inflammatory biomarkers in the prognosis of COVID-19, especially in evaluating death and other poor prognoses, with most biomarkers showing a better discriminatory ability.
Collapse
Affiliation(s)
- Kegang Zhan
- Department of Epidemiology, College of Preventive Medicine, Third Military Medical University (Army Medical University), Chongqing, China
- College of Public Health, Southwest Medical University, Luzhou, Sichuan, China
| | - Luhan Wang
- Department of Epidemiology, College of Preventive Medicine, Third Military Medical University (Army Medical University), Chongqing, China
| | - Hao Lin
- West China School of Clinical Medicine, Sichuan University, Chengdu, Sichuan, China
| | - Xiaoyu Fang
- Department of Epidemiology, College of Preventive Medicine, Third Military Medical University (Army Medical University), Chongqing, China
| | - Hong Jia
- College of Public Health, Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Xiangyu Ma
- Department of Epidemiology, College of Preventive Medicine, Third Military Medical University (Army Medical University), Chongqing 400038, China
| |
Collapse
|
9
|
Vicka V, Januskeviciute E, Bartuševiciene I, Ringaitiene D, Aleknaviciene A, Serpytis M, Rimsevicius L, Miglinas M, Jancoriene L, Sipylaite J. Kinetics of SuPAR hemoadsorption in critical COVID-19 patients on renal replacement therapy. BMC Nephrol 2022; 23:371. [PMCID: PMC9673197 DOI: 10.1186/s12882-022-03003-2] [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: 02/17/2022] [Accepted: 11/09/2022] [Indexed: 11/19/2022] Open
Abstract
Background SARS-CoV-2 viral infection is associated with a rapid and vigorous systemic inflammatory response syndrome. Soluble urokinase-type plasminogen activator receptor (suPAR) is a novel biomarker, both indicative of inflammation and propagating it. Hemoadsorption has been proposed as a potential therapy in COVID-19 patients, therefore the aim of this study is to determine suPAR kinetics during hemoadsoprtion. Methods This was a prospective observational study of critical COVID-19 patients, enrolled when hemoperfusion therapy was initiated. Hemoadsorber was integrated into the continuous renal replacement therapy circuit. The first series of suPAR measurements was performed 10 minutes after the start of the session, sampling both incoming and outgoing lines of the adsorber. A second series of the measurements was performed beforefinishing the session with the same adsorber. Statistical significance level was set < 0.05. Results This study included 18 patients. In the beginning of the session the fraction of suPAR cleared across the adsorber was 29.5% [16-41], and in the end of the session it decreased to 7.2% [4-22], 4 times lower, p = 0.003. The median length of session was 21 hours, with minimal duration of 16 hours and maximal duration of 24 hours. The median suPAR before the procedure was 8.71 [7.18-10.78] and after the session was 7.35 [6.53-11.28] ng/ml. There was no statistically significant difference in suPAR concentrations before and after the session (p = 0.831). Conclusions This study concluded that in the beginning of the hemoadsorption procedure significant amount of suPAR is removed from the circulation. However, in the end of the procedure there is a substantial drop in adsorbed capacity. Furthermore, despite a substantial amount of suPAR cleared there is no significant difference in systemic suPAR concentrations before and after the hemoadsorption procedure.
Collapse
Affiliation(s)
- Vaidas Vicka
- grid.6441.70000 0001 2243 2806Clinic of Anaesthesiology and Intensive Care, Institute of Clinical Medicine, Faculty of Medicine, Vilnius University, M.K. Ciurlionio 21, 03101 Vilnius, Lithuania
| | - Elija Januskeviciute
- grid.6441.70000 0001 2243 2806Faculty of Medicine, Vilnius University, Vilnius, Lithuania
| | - Ieva Bartuševiciene
- grid.6441.70000 0001 2243 2806Faculty of Medicine, Vilnius University, Vilnius, Lithuania
| | - Donata Ringaitiene
- grid.6441.70000 0001 2243 2806Clinic of Anaesthesiology and Intensive Care, Institute of Clinical Medicine, Faculty of Medicine, Vilnius University, M.K. Ciurlionio 21, 03101 Vilnius, Lithuania
| | - Aiste Aleknaviciene
- grid.6441.70000 0001 2243 2806Clinic of Anaesthesiology and Intensive Care, Institute of Clinical Medicine, Faculty of Medicine, Vilnius University, M.K. Ciurlionio 21, 03101 Vilnius, Lithuania
| | - Mindaugas Serpytis
- grid.6441.70000 0001 2243 2806Clinic of Anaesthesiology and Intensive Care, Institute of Clinical Medicine, Faculty of Medicine, Vilnius University, M.K. Ciurlionio 21, 03101 Vilnius, Lithuania
| | - Laurynas Rimsevicius
- grid.6441.70000 0001 2243 2806Clinic of Gastroenterology, Nephro-urology and surgery, Institute of Clinical Medicine, Faculty of Medicine, Vilnius University, Vilnius, Lithuania
| | - Marius Miglinas
- grid.6441.70000 0001 2243 2806Clinic of Gastroenterology, Nephro-urology and surgery, Institute of Clinical Medicine, Faculty of Medicine, Vilnius University, Vilnius, Lithuania
| | - Ligita Jancoriene
- grid.6441.70000 0001 2243 2806Clinic of Infectious Diseases and Dermatovenerology, Institute of Clinical Medicine, Faculty of Medicine, Vilnius University, Vilnius, Lithuania
| | - Jurate Sipylaite
- grid.6441.70000 0001 2243 2806Clinic of Anaesthesiology and Intensive Care, Institute of Clinical Medicine, Faculty of Medicine, Vilnius University, M.K. Ciurlionio 21, 03101 Vilnius, Lithuania
| |
Collapse
|
10
|
Liao TH, Wu HC, Liao MT, Hu WC, Tsai KW, Lin CC, Lu KC. The Perspective of Vitamin D on suPAR-Related AKI in COVID-19. Int J Mol Sci 2022; 23:10725. [PMID: 36142634 PMCID: PMC9500944 DOI: 10.3390/ijms231810725] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Revised: 09/08/2022] [Accepted: 09/13/2022] [Indexed: 01/08/2023] Open
Abstract
The coronavirus disease 2019 (COVID-19) pandemic has claimed the lives of millions of people around the world. Severe vitamin D deficiency can increase the risk of death in people with COVID-19. There is growing evidence that acute kidney injury (AKI) is common in COVID-19 patients and is associated with poorer clinical outcomes. The kidney effects of SARS-CoV-2 are directly mediated by angiotensin 2-converting enzyme (ACE2) receptors. AKI is also caused by indirect causes such as the hypercoagulable state and microvascular thrombosis. The increased release of soluble urokinase-type plasminogen activator receptor (suPAR) from immature myeloid cells reduces plasminogen activation by the competitive inhibition of urokinase-type plasminogen activator, which results in low plasmin levels and a fibrinolytic state in COVID-19. Frequent hypercoagulability in critically ill patients with COVID-19 may exacerbate the severity of thrombosis. Versican expression in proximal tubular cells leads to the proliferation of interstitial fibroblasts through the C3a and suPAR pathways. Vitamin D attenuates the local expression of podocyte uPAR and decreases elevated circulating suPAR levels caused by systemic inflammation. This decrease preserves the function and structure of the glomerular barrier, thereby maintaining renal function. The attenuated hyperinflammatory state reduces complement activation, resulting in lower serum C3a levels. Vitamin D can also protect against COVID-19 by modulating innate and adaptive immunity, increasing ACE2 expression, and inhibiting the renin-angiotensin-aldosterone system. We hypothesized that by reducing suPAR levels, appropriate vitamin D supplementation could prevent the progression and reduce the severity of AKI in COVID-19 patients, although the data available require further elucidation.
Collapse
Affiliation(s)
- Tzu-Hsien Liao
- Department of Chinese Medicine, Taipei Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, New Taipei City 231, Taiwan
| | - Hsien-Chang Wu
- Department of Chinese Medicine, Taipei Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, New Taipei City 231, Taiwan
- School of Post-Baccalaureate Chinese Medicine, Tzu Chi University, Hualien 970, Taiwan
| | - Min-Tser Liao
- Department of Pediatrics, Taoyuan Armed Forces General Hospital Hsinchu Branch, Hsinchu City 300, Taiwan
- Department of Pediatrics, Tri-Service General Hospital, National Defense Medical Center, Taipei 114, Taiwan
| | - Wan-Chung Hu
- Department of Clinical Pathology and Medical Research, Taipei Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, New Taipei City 231, Taiwan
| | - Kuo-Wang Tsai
- Department of Research, Taipei Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, New Taipei City 231, Taiwan
| | - Ching-Chieh Lin
- Department of Chest Medicine, Taoyuan Armed Forces General Hospital Hsinchu Branch, Hsinchu City 300, Taiwan
| | - Kuo-Cheng Lu
- Division of Nephrology, Department of Medicine, Taipei Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, New Taipei City 231, Taiwan
- Division of Nephrology, Department of Medicine, Fu-Jen Catholic University Hospital, School of Medicine, Fu-Jen Catholic University, New Taipei City 242, Taiwan
| |
Collapse
|
11
|
Azzouz M, Xu Y, Barregard L, Fagerberg B, Zöller B, Molnár P, Oudin A, Spanne M, Engström G, Stockfelt L. Air pollution and biomarkers of cardiovascular disease and inflammation in the Malmö Diet and Cancer cohort. Environ Health 2022; 21:39. [PMID: 35413834 PMCID: PMC9004064 DOI: 10.1186/s12940-022-00851-1] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2021] [Accepted: 03/31/2022] [Indexed: 05/30/2023]
Abstract
INTRODUCTION Air pollution is associated with increased risk of cardiovascular disease, possibly through chronic systemic inflammation that promotes the progression of atherosclerosis and the risk of cardiovascular events. This study aimed to investigate the associations between air pollution and established biomarkers of inflammation and cardiovascular disease. METHODS The Cardiovascular Subcohort of the Malmö Diet and Cancer cohort includes 6103 participants from the general population of Malmö, Sweden. The participants were recruited 1991-1994. Annual mean residential exposure to particulate matter < 2.5 and < 10 μm (PM2.5 and PM10), and nitrogen oxides (NOx) at year of recruitment were assigned from dispersion models. Blood samples collected at recruitment, including blood cell counts, and biomarkers (lymphocyte- and neutrophil counts, C-reactive protein (CRP), soluble urokinase-type plasminogen activator receptor (suPAR), lipoprotein-associated phospholipase A2 (Lp-PLA2), ceruloplasmin, orosomucoid, haptoglobin, complement-C3, and alpha-1-antitrypsin) were analyzed. Multiple linear regression models were used to investigate the cross-sectional associations between air pollutants and biomarkers. RESULTS The mean annual exposure levels in the cohort were only slightly or moderately above the new WHO guidelines of 5 μg/m3 PM2.5 (10.5 μg/m3 PM2.5). Residential PM2.5 exposure was associated with increased levels of ceruloplasmin, orosomucoid, C3, alpha-1-antitrypsin, haptoglobin, Lp-PLA2 and the neutrophil-lymphocyte ratio. Ceruloplasmin, orosomucoid, C3 and alpha-1-antitrypsin were also positively associated with PM10. There were no associations between air pollutants and suPAR, leukocyte counts or CRP. The associations between particles and biomarkers were still significant after removing outliers and adjustment for CRP levels. The associations were more prominent in smokers. CONCLUSION Long-term residential exposure to moderate levels of particulate air pollution was associated with several biomarkers of inflammation and cardiovascular disease. This supports inflammation as a mechanism behind the association between air pollution and cardiovascular disease.
Collapse
Affiliation(s)
- Mehjar Azzouz
- Occupational and Environmental Medicine, School of Public Health and Community Medicine, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden.
| | - Yiyi Xu
- Occupational and Environmental Medicine, School of Public Health and Community Medicine, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Lars Barregard
- Occupational and Environmental Medicine, School of Public Health and Community Medicine, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Björn Fagerberg
- Department of Molecular and Clinical Medicine, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Bengt Zöller
- Center for Primary Health Care Research, Lund University/Region Skåne, Malmö, Sweden
| | - Peter Molnár
- Occupational and Environmental Medicine, School of Public Health and Community Medicine, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Department of Occupational and Environmental Medicine, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Anna Oudin
- Occupational and Environmental Medicine, Department for Laboratory Medicine, Lund University, Lund, Sweden
- Division of Sustainable Health, Umeå University, Umeå, Sweden
| | - Mårten Spanne
- Environment Department, City of Malmö, Malmö, Sweden
| | - Gunnar Engström
- Department of Clinical Sciences in Malmö, CRC, Lund University, Lund, Sweden
| | - Leo Stockfelt
- Occupational and Environmental Medicine, School of Public Health and Community Medicine, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Department of Occupational and Environmental Medicine, Sahlgrenska University Hospital, Gothenburg, Sweden
| |
Collapse
|
12
|
Yu S, Sui Y, Wang J, Li Y, Li H, Cao Y, Chen L, Jiang L, Yuan C, Huang M. Crystal structure and cellular functions of uPAR dimer. Nat Commun 2022; 13:1665. [PMID: 35351875 PMCID: PMC8964761 DOI: 10.1038/s41467-022-29344-y] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2021] [Accepted: 01/25/2022] [Indexed: 11/09/2022] Open
Abstract
AbstractReceptor dimerization of urokinase-type plasminogen activator receptor (uPAR) was previously identified at protein level and on cell surface. Recently, a dimeric form of mouse uPAR isoform 2 was proposed to induce kidney disease. Here, we report the crystal structure of human uPAR dimer at 2.96 Å. The structure reveals enormous conformational changes of the dimer compared to the monomeric structure: D1 of uPAR opens up into a large expanded ring that captures a β-hairpin loop of a neighboring uPAR to form an expanded β-sheet, leading to an elongated, highly intertwined dimeric uPAR. Based on the structure, we identify E49P as a mutation promoting dimer formation. The mutation increases receptor binding to the amino terminal fragment of its primary ligand uPA, induces the receptor to distribute to the basal membrane, promotes cell proliferation, and alters cell morphology via β1 integrin signaling. These results reveal the structural basis for uPAR dimerization, its effect on cellular functions, and provide a basis to further study this multifunctional receptor.
Collapse
|
13
|
Padelli M, Gueye P, Guilloux D, Banydeen R, Campana V, Cabie A, Neviere R. Soluble urokinase plasminogen activator receptor levels are predictive of COVID-19 severity in Afro-Caribbean patients. Biomark Med 2022; 16:169-177. [PMID: 35081737 PMCID: PMC8809376 DOI: 10.2217/bmm-2021-0669] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Aim: To investigate association between soluble urokinase plasminogen activator receptor (suPAR) plasma levels at admission and incidence of complications in COVID-19 patients. Patients & methods: We considered Afro-Caribbean patients (n = 64) admitted to the hospital between 1 February 2020 and 28 February 2021. Primary outcome was time from the hospital admission until intensive care unit care or death. Results: Primary outcome (hazard ratio, HR [95%CI]) was associated with higher CT scan severity score (3.18 [1.15-8.78], p = 0.025), National Early Warning Score (NEWS2; 1.43 [1.02-2.02], p = 0.041) and suPAR (1.28 [1.06-2.06], p = 0.041). Kaplan-Meier analysis indicated patients with suPAR level above 8.95 ng/ml had a worse outcome (7.95 [3.33-18.97], p < 0.001). Conclusion: Our study suggests that COVID-19 patients with increased baseline suPAR levels are at a high risk of complications.
Collapse
Affiliation(s)
- Mael Padelli
- University Hospital of Martinique, Department of Biochemistry, Fort-de-France, 97261, Martinique, France
| | - Papa Gueye
- University Hospital of Martinique, Department of Emergency Medicine, Fort-de-France, 97261, Martinique, France
| | - Diane Guilloux
- University Hospital of Martinique, Department of Critical Care Medicine, Fort-de-France, 97261, Martinique, France
| | - Rishika Banydeen
- University Hospital of Martinique, Department of Epidemiology & Biostatistics, Fort de France, 97261, Martinique, France
| | - Valentine Campana
- Centre d'Investigation Clinique Antilles Guyane, Inserm CIC 1424, Fort de France, 97261, Martinique, France
| | - Andre Cabie
- University Hospital of Martinique, Department of Infectious Diseases, Fort-de-France, 97261, Martinique, France
| | - Remi Neviere
- University Hospital of Martinique, Department of Cardiology, Fort de France, 97261, Martinique, France.,Université des Antilles, Cardiovascular Research Team EA7525, Fort de France, 97261, France
| |
Collapse
|
14
|
Da Silva Lodge M, Pullen N, Pereira M, Johnson TS. Urinary levels of pro-fibrotic transglutaminase 2 (TG2) may help predict progression of chronic kidney disease. PLoS One 2022; 17:e0262104. [PMID: 35041708 PMCID: PMC8765645 DOI: 10.1371/journal.pone.0262104] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2021] [Accepted: 12/16/2021] [Indexed: 01/04/2023] Open
Abstract
Renal clinical chemistry only detects kidney dysfunction after considerable damage has occurred and is imperfect in predicting long term outcomes. Consequently, more sensitive markers of early damage and better predictors of progression are being urgently sought, to better support clinical decisions and support shorter clinical trials. Transglutaminase 2 (TG2) is strongly implicated in the fibrotic remodeling that drives chronic kidney disease (CKD). We hypothesized that urinary TG2 and its ε-(γ-glutamyl)-lysine crosslink product could be useful biomarkers of kidney fibrosis and progression. Animal models: a rat 4-month 5/6th subtotal nephrectomy model of CKD and a rat 8-month streptozotocin model of diabetic kidney disease had 24-hour collection of urine, made using a metabolic cage, at regular periods throughout disease development. Patients: Urine samples from patients with CKD (n = 290) and healthy volunteers (n = 33) were collected prospectively, and progression tracked for 3 years. An estimated glomerular filtration rate (eGFR) loss of 2-5 mL/min/year was considered progressive, with rapid progression defined as > 5 mL/min/year. Assays: TG2 was measured in human and rat urine samples by enzyme-linked immunosorbent assay (ELISA) and ε-(γ-glutamyl)-lysine by exhaustive proteolytic digestion and amino acid analysis. Urinary TG2 and ε-(γ-glutamyl)-lysine increased with the development of fibrosis in both animal model systems. Urinary TG2 was 41-fold higher in patients with CKD than HVs, with levels elevated 17-fold by CKD stage 2. The urinary TG2:creatinine ratio (UTCR) was 9 ng/mmol in HV compared with 114 ng/mmol in non-progressive CKD, 1244 ng/mmol in progressive CKD and 1898 ng/mmol in rapidly progressive CKD. Both urinary TG2 and ε-(γ-glutamyl)-lysine were significantly associated with speed of progression in univariate logistic regression models. In a multivariate model adjusted for urinary TG2, ε-(γ-glutamyl)-lysine, age, sex, urinary albumin:creatinine ratio (UACR), urinary protein:creatinine ratio (UPCR), and CKD stage, only TG2 remained statistically significant. Receiver operating characteristic (ROC) curve analysis determined an 86.4% accuracy of prediction of progression for UTCR compared with 73.5% for UACR. Urinary TG2 and ε-(γ-glutamyl)-lysine are increased in CKD. In this pilot investigation, UTCR was a better predictor of progression in patients with CKD than UACR. Larger studies are now warranted to fully evaluate UTCR value in predicting patient outcomes.
Collapse
Affiliation(s)
- Michelle Da Silva Lodge
- Academic Nephrology Unit and Sheffield Kidney Institute, University of Sheffield Medical School, Sheffield, United Kingdom
| | - Nick Pullen
- Pfizer Global Research and Development, Cambridge, MA, United States of America
| | - Miguel Pereira
- Statistical Sciences and Innovation, UCB Pharma, Slough, United Kingdom
| | - Timothy S. Johnson
- Academic Nephrology Unit and Sheffield Kidney Institute, University of Sheffield Medical School, Sheffield, United Kingdom
| |
Collapse
|
15
|
Blomberg K, Hansen TF, Brasen CL, Madsen JB, Jensen LH, Thomsen CB. The Soluble Urokinase-Type Plasminogen Activator Receptor as a Biomarker for Survival and Early Treatment Effect in Metastatic Colorectal Cancer. Cancers (Basel) 2021; 13:5100. [PMID: 34680247 PMCID: PMC8534079 DOI: 10.3390/cancers13205100] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2021] [Revised: 10/07/2021] [Accepted: 10/08/2021] [Indexed: 12/17/2022] Open
Abstract
The soluble urokinase-type plasminogen activator receptor (suPAR) is prognostic for overall survival (OS) in colorectal cancer (CRC). Our study explored the association between baseline suPAR and OS and progression-free survival (PFS) in metastatic CRC (mCRC). It is also the first study to explore the association between the initial change in suPAR level and OS, PFS and the first CT response evaluation. The study included 132 patients with mCRC treated with chemotherapy (FOLFIRI) with or without an EGFR-inhibitor. Blood samples were drawn before the first treatment cycle and in between the first and second treatment cycle. suPAR levels were determined using an ELISA assay. Using the Kaplan-Meyer method, we demonstrated a significantly shorter OS for patients with suPAR levels above the median (HR = 1.79, 95%CI = 1.10-2.92, p = 0.01). We also showed association between plasma suPAR level, gender and performance status (PS). However, we could not show any association with PFS, and analysis on the change in suPAR level provided no significant results. The results showing association between baseline suPAR and OS are in line with previous findings.
Collapse
Affiliation(s)
- Kristian Blomberg
- Department of Health Science, University of Southern Denmark, 5000 Odense, Denmark;
- Danish Colorectal Cancer Center South, Department of Oncology, University Hospital of Southern Denmark, 7100 Vejle, Denmark; (T.F.H.); (L.H.J.)
| | - Torben F. Hansen
- Danish Colorectal Cancer Center South, Department of Oncology, University Hospital of Southern Denmark, 7100 Vejle, Denmark; (T.F.H.); (L.H.J.)
- Department of Regional Health Research, University of Southern Denmark, 5000 Odense, Denmark;
| | - Claus L. Brasen
- Department of Regional Health Research, University of Southern Denmark, 5000 Odense, Denmark;
- Department of Biochemistry and Immunology, Lillebaelt Hospital, University Hospital of Southern Denmark, 7100 Vejle, Denmark;
| | - Jeppe B. Madsen
- Department of Biochemistry and Immunology, Lillebaelt Hospital, University Hospital of Southern Denmark, 7100 Vejle, Denmark;
| | - Lars H. Jensen
- Danish Colorectal Cancer Center South, Department of Oncology, University Hospital of Southern Denmark, 7100 Vejle, Denmark; (T.F.H.); (L.H.J.)
- Department of Regional Health Research, University of Southern Denmark, 5000 Odense, Denmark;
- Open Patient Data Explorative Network, The Clinical Institute, Odense University Hospital, 5000 Odense, Denmark
| | - Caroline B. Thomsen
- Danish Colorectal Cancer Center South, Department of Oncology, University Hospital of Southern Denmark, 7100 Vejle, Denmark; (T.F.H.); (L.H.J.)
| |
Collapse
|
16
|
Skjøt-Arkil H, Heltborg A, Lorentzen MH, Cartuliares MB, Hertz MA, Graumann O, Rosenvinge FS, Petersen ERB, Østergaard C, Laursen CB, Skovsted TA, Posth S, Chen M, Mogensen CB. Improved diagnostics of infectious diseases in emergency departments: a protocol of a multifaceted multicentre diagnostic study. BMJ Open 2021; 11:e049606. [PMID: 34593497 PMCID: PMC8487181 DOI: 10.1136/bmjopen-2021-049606] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/03/2022] Open
Abstract
BACKGROUND The major obstacle in prescribing an appropriate and targeted antibiotic treatment is insufficient knowledge concerning whether the patient has a bacterial infection, where the focus of infection is and which bacteria are the agents of the infection. A prerequisite for the appropriate use of antibiotics is timely access to accurate diagnostics such as point-of-care (POC) testing.The study aims to evaluate diagnostic tools and working methods that support a prompt and accurate diagnosis of hospitalised patients suspected of an acute infection. We will focus on the most common acute infections: community-acquired pneumonia (CAP) and acute pyelonephritis (APN). The objectives are to investigate (1) patient characteristics and treatment trajectory of the different acute infections, (2) diagnostic and prognostic accuracy of infection markers, (3) diagnostic accuracy of POC urine flow cytometry on diagnosing and excluding bacteriuria, (4) how effective the addition of POC analysis of sputum to the diagnostic set-up for CAP is on antibiotic prescriptions, (5) diagnostic accuracy of POC ultrasound and ultralow dose (ULD) computerized tomography (CT) on diagnosing CAP, (6) diagnostic accuracy of specialist ultrasound on diagnosing APN, (7) diagnostic accuracy of POC ultrasound in diagnosing hydronephrosis in patients suspected of APN. METHODS AND ANALYSIS It is a multifaceted multicentre diagnostic study, including 1000 adults admitted with suspicion of an acute infection. Participants will, within the first 24 hours of admission, undergo additional diagnostic tests including infection markers, POC urine flow cytometry, POC analysis of sputum, POC and specialist ultrasound, and ULDCT. The primary reference standard is an assigned diagnosis determined by a panel of experts. ETHICS, DISSEMINATION AND REGISTRATION Approved by Regional Committees on Health Research Ethics for Southern Denmark, Danish Data Protection Agency and clinicaltrials.gov. Results will be presented in peer-reviewed journals, and positive, negative and inconclusive results will be published. TRIAL REGISTRATION NUMBERS NCT04661085, NCT04681963, NCT04667195, NCT04652167, NCT04686318, NCT04686292, NCT04651712, NCT04645030, NCT04651244.
Collapse
Affiliation(s)
- Helene Skjøt-Arkil
- Emergency Department, University Hospital of Southern Denmark, Aabenraa, Denmark
- Department of Regional Health Research, University of Southern Denmark Faculty of Health Sciences, Odense, Denmark
| | - Anne Heltborg
- Department of Regional Health Research, University of Southern Denmark Faculty of Health Sciences, Odense, Denmark
- Department of Internal Medicine, University Hospital of Southern Denmark, Sønderborg, Denmark
| | - Morten Hjarnø Lorentzen
- Emergency Department, University Hospital of Southern Denmark, Aabenraa, Denmark
- Department of Regional Health Research, University of Southern Denmark Faculty of Health Sciences, Odense, Denmark
| | - Mariana Bichuette Cartuliares
- Emergency Department, University Hospital of Southern Denmark, Aabenraa, Denmark
- Department of Regional Health Research, University of Southern Denmark Faculty of Health Sciences, Odense, Denmark
| | - Mathias Amdi Hertz
- Department of Infectious Diseases, Odense University Hospital, Odense, Denmark
- Department of Clinical Research, University of Southern Denmark Faculty of Health Sciences, Odense, Denmark
| | - Ole Graumann
- Department of Clinical Research, University of Southern Denmark Faculty of Health Sciences, Odense, Denmark
- Department of Radiology, Odense University Hospital, Odense, Denmark
| | | | - Eva Rabing Brix Petersen
- Blood Samples, Biochemistry and Immunology, University Hospital of Southern Denmark, Aabenraa, Denmark
| | - Claus Østergaard
- Department of Clinical Microbiology, University Hospital of Southern Denmark, Vejle, Denmark
| | - Christian B Laursen
- Department of Clinical Research, University of Southern Denmark Faculty of Health Sciences, Odense, Denmark
- Department of Respiratory Medicine, Odense University Hospital, Odense, Denmark
| | - Thor Aage Skovsted
- Blood Samples, Biochemistry and Immunology, University Hospital of Southern Denmark, Aabenraa, Denmark
| | - Stefan Posth
- Department of Clinical Research, University of Southern Denmark Faculty of Health Sciences, Odense, Denmark
- Emergency Department, Odense University Hospital, Odense, Denmark
| | - Ming Chen
- Department of Regional Health Research, University of Southern Denmark Faculty of Health Sciences, Odense, Denmark
- Department of Microbiology, University Hospital of Southern Denmark, Aabenraa, Denmark
| | - Christian Backer Mogensen
- Emergency Department, University Hospital of Southern Denmark, Aabenraa, Denmark
- Department of Regional Health Research, University of Southern Denmark Faculty of Health Sciences, Odense, Denmark
| |
Collapse
|
17
|
Experimental and Clinical Evidence Supports the Use of Urokinase Plasminogen Activation System Components as Clinically Relevant Biomarkers in Gastroesophageal Adenocarcinoma. Cancers (Basel) 2021; 13:cancers13164097. [PMID: 34439251 PMCID: PMC8393967 DOI: 10.3390/cancers13164097] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2021] [Revised: 08/02/2021] [Accepted: 08/09/2021] [Indexed: 12/18/2022] Open
Abstract
Simple Summary Patients with gastric and oesophageal adenocarcinomas (GOCs) have short life expectancies as their tumours spread to other sites early. This is facilitated by the increased expression of the urokinase plasminogen activation system (uPAS); a feature of the majority of GOCs. There is increasing appreciation of the importance of uPAS expression in a range of cell types within the tumour microenvironment. Abundant clinical evidence indicates that altered expression of uPAS proteins is associated with worse outcomes, including time to tumour recurrence and patient survival. Emerging technologies, including liquid biopsy, suggest a role of uPAS for the detection of circulating tumour cells, which are responsible for the dissemination of cancers. We review and summarise pre-clinical and clinical data that supports the use of uPAS as a biomarker in GOC. Abstract Gastric and oesophageal cancers (GOCs) are lethal cancers which metastasise early and recur frequently, even after definitive surgery. The urokinase plasminogen activator system (uPAS) is strongly implicated in the invasion and metastasis of many aggressive tumours including GOCs. Urokinase plasminogen activator (uPA) interaction with its receptor, urokinase plasminogen activator receptor (uPAR), leads to proteolytic activation of plasminogen to plasmin, a broad-spectrum protease which enables tumour cell invasion and dissemination to distant sites. uPA, uPAR and the plasminogen activator inhibitor type 1 (PAI-1) are overexpressed in some GOCs. Accumulating evidence points to a causal role of activated receptor tyrosine kinase pathways enhancing uPAS expression in GOCs. Expression of these components are associated with poorer clinicopathological features and patient survival. Stromal cells, including tumour-associated macrophages and myofibroblasts, also express the key uPAS proteins, supporting the argument of stromal involvement in GOC progression and adverse effect on patient survival. uPAS proteins can be detected on circulating leucocytes, circulating tumour cells and within the serum; all have the potential to be developed into circulating biomarkers of GOC. Herein, we review the experimental and clinical evidence supporting uPAS expression as clinical biomarker in GOC, with the goal of developing targeted therapeutics against the uPAS.
Collapse
|
18
|
Kerget B, Kerget F, Aksakal A, Aşkın S, Uçar EY, Sağlam L. Evaluation of the relationship between KIM-1 and suPAR levels and clinical severity in COVID-19 patients: A different perspective on suPAR. J Med Virol 2021; 93:5568-5573. [PMID: 34019703 PMCID: PMC8242801 DOI: 10.1002/jmv.27099] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Revised: 05/15/2021] [Accepted: 05/18/2021] [Indexed: 12/21/2022]
Abstract
Coronavirus disease 2019 (COVID-19) is one of the most pressing health problems of this century, but our knowledge of the disease is still limited. In this study, we aimed to examine serum-soluble urokinase plasminogen activator receptor (suPAR) and kidney injury molecule 1 (KIM-1) levels based on the clinical course of COVID-19. Our study included 102 patients over the age of 18 who were diagnosed as having COVID-19 between September 2020 and December 2020 and a control group of 50 health workers over the age of 18 whose severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) PCR results were negative. KIM-1 was measured by ELISA and suPAR by suPARnostic™ assay. Analysis of previously identified variables of prognostic significance in COVID-19 revealed high neutrophil to lymphocyte ratio, lactose dehydrogenase, prothrombin time, C-reactive protein, PaO2 /FiO2 , D-dimer, ferritin, and fibrinogen levels in patients with severe disease (p < 0.05 for all). KIM-1 and suPAR levels were significantly higher in COVID-19 patients compared to the control group (p = 0.001 for all). KIM-1 level was higher in severe patients compared to moderate patients (p = 0.001), while suPAR level was lower (p = 0.001). KIM-1, which is believed to play an important role in the endocytosis of SARS-CoV-2, was elevated in patients with severe COVID-19 and may be a therapeutic target in the future. SuPAR may have a role in defense mechanism and fibrinolysis, and low levels in severe patients may be associated with poor prognosis in the early period.
Collapse
Affiliation(s)
- Buğra Kerget
- Department of Pulmonary Diseases, Ataturk University School of Medicine, Yakutiye, Erzurum, Turkey
| | - Ferhan Kerget
- Department of Infection Diseases and Clinical Microbiology, Health Sciences University Erzurum Regional Education and Research Hospital, Yakutiye, Erzurum, Turkey
| | - Alperen Aksakal
- Department of Pulmonary Diseases, Health Sciences University Erzurum Regional Education and Research Hospital, Yakutiye, Erzurum, Turkey
| | - Seda Aşkın
- Department of Biochemistry, Ataturk University School of Medicine, Yakutiye, Erzurum, Turkey
| | - Elif Yılmazel Uçar
- Department of Pulmonary Diseases, Ataturk University School of Medicine, Yakutiye, Erzurum, Turkey
| | - Leyla Sağlam
- Department of Pulmonary Diseases, Ataturk University School of Medicine, Yakutiye, Erzurum, Turkey
| |
Collapse
|
19
|
Oulhaj A, Alsuwaidi AR, Suliman A, Gasmelseed H, Khan S, Alawi S, Hukan Y, George J, Alshamsi F, Sheikh F, Babiker ZOE, Prattes J, Sourij H. Admission levels of Soluble Urokinase Plasminogen Activator Receptor (suPAR) are Associated with the Development of Severe Complications in Hospitalised COVID-19 Patients: A Prospective Cohort Study. Int J Infect Dis 2021; 107:188-194. [PMID: 33862208 PMCID: PMC8056823 DOI: 10.1016/j.ijid.2021.04.026] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2021] [Revised: 04/02/2021] [Accepted: 04/08/2021] [Indexed: 02/06/2023] Open
Abstract
Objective To examine the association between plasma levels of the soluble urokinase plasminogen activator receptor (suPAR) and the incidence of severe complications of COVID-19. Methods 403 RT-PCR-confirmed COVID-19 patients were recruited and prospectively followed-up at a major hospital in the United Arab Emirates. The primary endpoint was time from admission until the development of a composite outcome, including acute respiratory distress syndrome (ARDS), intensive care unit (ICU) admission, or death from any cause. Patients discharged alive were considered as competing events to the primary outcome. Competing risk regression was used to quantify the association between suPAR and the incidence of the primary outcome. Results 6.2% of patients experienced ARDS or ICU admission, but none died. Taking into account competing risk, the incidence of the primary outcome was 11.5% (95% confidence interval [CI], 6.7–16.3) in patients with suPAR levels >3.91 ng/mL compared to 2.9% (95% CI, 0.4–5.5) in those with suPAR ≤3.91 ng/mL. Also, an increase by 1 ng/mL in baseline suPAR resulted in a 58% rise in the hazard of developing the primary outcome (hazard ratio 1.6, 95% CI, 1.2–2.1, p = 0.003). Conclusion suPAR has an excellent prognostic utility in predicting severe complications in hospitalised COVID-19 patients.
Collapse
Affiliation(s)
- Abderrahim Oulhaj
- Institute of Public Health, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain, United Arab Emirates; Zayed Center for Health Sciences, United Arab Emirates University, United Arab Emirates.
| | - Ahmed R Alsuwaidi
- Department of Paediatrics, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain, United Arab Emirates
| | - Abubaker Suliman
- Institute of Public Health, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain, United Arab Emirates
| | | | - Shaima Khan
- Academic Affairs Department, Tawam Hospital, Al Ain, United Arab Emirates
| | - Shamma Alawi
- Academic Affairs Department, Tawam Hospital, Al Ain, United Arab Emirates
| | - Yaman Hukan
- Academic Affairs Department, Tawam Hospital, Al Ain, United Arab Emirates
| | - Junu George
- Department of Paediatrics, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain, United Arab Emirates
| | - Fayez Alshamsi
- Department of Internal Medicine, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain, United Arab Emirates
| | | | - Zahir Osman Eltahir Babiker
- Division of Infectious Diseases, Sheikh Shakhbout Medical City in Partnership with Mayo Clinic, Abu Dhabi, United Arab Emirates
| | - Juergen Prattes
- Department of Internal Medicine, Section of Infectious Diseases and Tropical Medicine, Medical University of Graz, Austria
| | - Harald Sourij
- Zayed Center for Health Sciences, United Arab Emirates University, United Arab Emirates; Department of Internal Medicine, Division of Endocrinology and Diabetology, Medical University of Graz, Austria
| |
Collapse
|
20
|
Burcsár S, Toldi G, Kovács L, Szalay B, Vásárhelyi B, Balog A. Urine soluble urokinase plasminogen activator receptor as a potential biomarker of lupus nephritis activity. Biomarkers 2021; 26:443-449. [PMID: 33825610 DOI: 10.1080/1354750x.2021.1910343] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
There is a lack of non-invasive biomarkers to identify lupus nephritis (LN). Soluble urokinase plasminogen activator receptor (suPAR) is a sensitive biomarker of ongoing inflammation and a potential marker of podocyte dysfunction. The aim of this study was to assess urine and plasma suPAR in LN. 14 systemic lupus erythematosus (SLE) patients with newly diagnosed LN, 8 active SLE patients (SLEDAI >8) without LN and 31 healthy individuals were enrolled. Urine and plasma samples were taken before the initiation of LN induction therapy, and monthly thereafter. Global and renal disease activity were defined using the SLEDAI-2K and the SLEDAI-2K renal domain score, respectively. suPAR concentrations were measured with the suPARnostic Flex ELISA assay. Urine and plasma suPAR levels were elevated in SLE patients with active LN compared with resolved LN and healthy controls. Urine suPAR levels were comparable to healthy controls in active SLE without LN. Urine and plasma suPAR levels were higher before than after the initiation of LN induction therapy. Prospective follow-up measurements also suggested that urine suPAR levels raised again in patients with a relapse of LN according to SLEDAI-2K renal domain score, whereas plasma suPAR levels did not correlate with renal disease activity. Urine suPAR is a promising LN activity biomarker, given its isolated elevation in urine in active LN and pronounced decrease with LN improvement.
Collapse
Affiliation(s)
- Szilárd Burcsár
- Department of Rheumatology and Immunology, University of Szeged, Szeged, Hungary
| | - Gergely Toldi
- Department of Laboratory Medicine, Semmelweis University, Budapest, Hungary
| | - László Kovács
- Department of Rheumatology and Immunology, University of Szeged, Szeged, Hungary
| | - Balázs Szalay
- Department of Laboratory Medicine, Semmelweis University, Budapest, Hungary
| | - Barna Vásárhelyi
- Department of Laboratory Medicine, Semmelweis University, Budapest, Hungary
| | - Attila Balog
- Department of Rheumatology and Immunology, University of Szeged, Szeged, Hungary
| |
Collapse
|
21
|
Huet F, Dupuy AM, Duflos C, Reis CA, Kuster N, Aguilhon S, Cristol JP, Roubille F. Soluble urokinase-type plasminogen activator receptor strongly predicts global mortality in acute heart failure patients: insight from the STADE-HF registry. Future Sci OA 2021; 7:FSO697. [PMID: 34046195 PMCID: PMC8147742 DOI: 10.2144/fsoa-2020-0197] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Background Whether soluble urokinase-type plasminogen activator receptor (suPAR) could be a valuable prognostic indicator remains uncertain. Materials & methods Patients from STADE-HF (Soluble Suppression of Tumorigenesis-2 as a Help for Management of Diagnosis, Evaluation and Management of Heart Failure) were included for analysis. Results 95 patients were included. The suPAR level of expression was significantly higher in the group of patients who died at one month (7.90 ± 4.35 ng/ml vs 11.94 ± 6.86 ng/ml; p < 0.05) or 1 year (7.28 ± 4.27 ng/ml vs 11.81 ± 4.88 ng/ml; p < 0.01), but there was no significant difference according to the readmission. Conclusion High suPAR levels during hospitalization for acute heart failure were highly predictive for the risk of mortality, but not the risk of readmission.
Collapse
Affiliation(s)
- Fabien Huet
- Cardiology Department, University Hospital of Montpellier, Montpellier, France.,PhyMedExp, University of Montpellier, INSERM U1046, CNRS UMR, 9214, Montpellier, 34090, France
| | - Anne-Marie Dupuy
- Department of Biochemistry, Centre Ressources Biologiques de Montpellier, University Hospital of Montpellier, Montpellier, 34090, France
| | - Claire Duflos
- Medico-Economic Research Unit, CHU Montpellier, University of Montpellier, Montpellier, 34090, France
| | - Cintia Azara Reis
- Cardiology Department, University Hospital of Montpellier, Montpellier, France
| | - Nils Kuster
- PhyMedExp, University of Montpellier, INSERM U1046, CNRS UMR, 9214, Montpellier, 34090, France.,Department of Biochemistry, Centre Ressources Biologiques de Montpellier, University Hospital of Montpellier, Montpellier, 34090, France
| | - Sylvain Aguilhon
- Cardiology Department, University Hospital of Montpellier, Montpellier, France
| | - Jean-Paul Cristol
- PhyMedExp, University of Montpellier, INSERM U1046, CNRS UMR, 9214, Montpellier, 34090, France
| | - François Roubille
- Cardiology Department, University Hospital of Montpellier, Montpellier, France.,PhyMedExp, University of Montpellier, INSERM U1046, CNRS UMR, 9214, Montpellier, 34090, France
| |
Collapse
|
22
|
Butt S, Jeppesen JL, Iversen LV, Fenger M, Eugen-Olsen J, Andersson C, Jacobsen S. Association of soluble urokinase plasminogen activator receptor levels with fibrotic and vascular manifestations in systemic sclerosis. PLoS One 2021; 16:e0247256. [PMID: 33617568 PMCID: PMC7899346 DOI: 10.1371/journal.pone.0247256] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2020] [Accepted: 02/03/2021] [Indexed: 12/28/2022] Open
Abstract
OBJECTIVE We assessed the association of suPAR (soluble urokinase plasminogen activator receptor) plasma levels with fibrotic and vascular manifestations in patients with systemic sclerosis (SSc). METHODS suPAR plasma levels were measured in 121 consecutive patients with SSc and correlated to pulmonary and vascular features of SSc, including interstitial lung disease as characterized by percentage of predicted CO diffusing capacity (DLco) and forced vital capacity (FVC), pulmonary fibrosis by computed tomography, and pulmonary arterial hypertension, telangiectasias, and digital ulcers. RESULTS Overall, 121 SSc patients (84% females; mean age, 57 ± 12 [range: 22-79] years) were enrolled; 35% had diffuse cutaneous SSc. suPAR plasma levels ranged from 1.3-10.2 [median: 2.9 (p25-p75: 2.3-3.9)] ng/mL. Log(suPAR) levels correlated with DLco (r = -0.41, p <0.0001) and FVC (r = -0.26, p = 0.004), also when adjusted for age, sex, and pulmonary hypertension. A suPAR cut-off level of >2.5 ng/mL showed a sensitivity of 91% for identifying patients with either DLco <50% or FVC < 60% of the predicted values. Similarly, 19 (90%) had a suPAR >2.5 ng/mL among those diagnosed with pulmonary fibrosis vs. 59 (60%) among those who did not (p = 0.008). suPAR values were not associated with vascular manifestations. CONCLUSION suPAR levels strongly correlated with pulmonary involvement in SSc. Future studies should test if suPAR estimation can be used for surveillance of severe pulmonary involvement in SSc.
Collapse
Affiliation(s)
- Sheraz Butt
- Department of Internal Medicine, Amager and Hvidovre University Hospital, Glostrup, Denmark
- * E-mail:
| | - Jørgen L. Jeppesen
- Department of Internal Medicine, Amager and Hvidovre University Hospital, Glostrup, Denmark
| | - Line Vinderslev Iversen
- Department of Dermatology, Bispebjerg University Hospital, Copenhagen, Denmark
- Department of Dermatology and Allergy, Odense University Hospital, Odense, Denmark
| | - Mogens Fenger
- Department of Clinical Biochemistry, Amager and Hvidovre University Hospital, Hvidovre, Denmark
| | - Jesper Eugen-Olsen
- Clinical Research Centre, Amager and Hvidovre University Hospital, Hvidovre, Denmark
| | - Charlotte Andersson
- Department of Cardiology, Herlev-Gentofte University Hospital, Hellerup, Denmark
| | - Søren Jacobsen
- Copenhagen Lupus and Vasculitis Clinic, Centre for Rheumatology and Spine Disease, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
| |
Collapse
|
23
|
Guo Q, Yaron JR, Wallen JW, Browder KF, Boyd R, Olson TL, Burgin M, Ulrich P, Aliskevich E, Schutz LN, Fromme P, Zhang L, Lucas AR. PEGylated Serp-1 Markedly Reduces Pristane-Induced Experimental Diffuse Alveolar Hemorrhage, Altering uPAR Distribution, and Macrophage Invasion. Front Cardiovasc Med 2021; 8:633212. [PMID: 33665212 PMCID: PMC7921738 DOI: 10.3389/fcvm.2021.633212] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2020] [Accepted: 01/20/2021] [Indexed: 12/22/2022] Open
Abstract
Diffuse alveolar hemorrhage (DAH) is one of the most serious clinical complications of systemic lupus erythematosus (SLE). The prevalence of DAH is reported to range from 1 to 5%, but while DAH is considered a rare complication there is a reported 50-80% mortality. There is at present no proven effective treatment for DAH and the therapeutics that have been tested have significant side effects. There is a clear necessity to discover new drugs to improve outcomes in DAH. Serine protease inhibitors, serpins, regulate thrombotic and thrombolytic protease cascades. We are investigating a Myxomavirus derived immune modulating serpin, Serp-1, as a new class of immune modulating therapeutics for vasculopathy and lung hemorrhage. Serp-1 has proven efficacy in models of herpes virus-induced arterial inflammation (vasculitis) and lung hemorrhage and has also proved safe in a clinical trial in patients with unstable coronary syndromes and stent implant. Here, we examine Serp-1, both as a native secreted protein expressed by CHO cells and as a polyethylene glycol modified (PEGylated) variant (Serp-1m5), for potential therapy in DAH. DAH was induced by intraperitoneal (IP) injection of pristane in C57BL/6J (B6) mice. Mice were treated with 100 ng/g bodyweight of either Serp-1 as native 55 kDa secreted glycoprotein, or as Serp-1m5, or saline controls after inducing DAH. Treatments were repeated daily for 14 days (6 mice/group). Serp-1 partially and Serp-1m5 significantly reduced pristane-induced DAH when compared with saline as assessed by gross pathology and H&E staining (Serp-1, p = 0.2172; Serp-1m5, p = 0.0252). Both Serp-1m5 and Serp-1 treatment reduced perivascular inflammation and reduced M1 macrophage (Serp-1, p = 0.0350; Serp-1m5, p = 0.0053), hemosiderin-laden macrophage (Serp-1, p = 0.0370; Serp-1m5, p = 0.0424) invasion, and complement C5b/9 staining. Extracellular urokinase-type plasminogen activator receptor positive (uPAR+) clusters were significantly reduced (Serp-1, p = 0.0172; Serp-1m5, p = 0.0025). Serp-1m5 also increased intact uPAR+ alveoli in the lung (p = 0.0091). In conclusion, Serp-1m5 significantly reduces lung damage and hemorrhage in a pristane model of SLE DAH, providing a new potential therapeutic approach.
Collapse
Affiliation(s)
- Qiuyun Guo
- Center for Personalized Diagnostics and Center for Immunotherapy, Vaccines and Virotherapy, The Biodesign Institute, Arizona State University, Tempe, AZ, United States.,Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jordan R Yaron
- Center for Personalized Diagnostics and Center for Immunotherapy, Vaccines and Virotherapy, The Biodesign Institute, Arizona State University, Tempe, AZ, United States
| | - John W Wallen
- Exalt Therapeutics LLC, Las Vegas, NV, United States
| | - Kyle F Browder
- Center for Personalized Diagnostics and Center for Immunotherapy, Vaccines and Virotherapy, The Biodesign Institute, Arizona State University, Tempe, AZ, United States
| | - Ryan Boyd
- Center for Applied Structural Discovery, The Biodesign Institute, Arizona State University, Tempe, AZ, United States
| | - Tien L Olson
- Center for Applied Structural Discovery, The Biodesign Institute, Arizona State University, Tempe, AZ, United States
| | - Michelle Burgin
- Center for Personalized Diagnostics and Center for Immunotherapy, Vaccines and Virotherapy, The Biodesign Institute, Arizona State University, Tempe, AZ, United States
| | - Peaches Ulrich
- Center for Personalized Diagnostics and Center for Immunotherapy, Vaccines and Virotherapy, The Biodesign Institute, Arizona State University, Tempe, AZ, United States
| | - Emily Aliskevich
- Center for Personalized Diagnostics and Center for Immunotherapy, Vaccines and Virotherapy, The Biodesign Institute, Arizona State University, Tempe, AZ, United States
| | - Lauren N Schutz
- Center for Personalized Diagnostics and Center for Immunotherapy, Vaccines and Virotherapy, The Biodesign Institute, Arizona State University, Tempe, AZ, United States
| | - Petra Fromme
- Center for Applied Structural Discovery, The Biodesign Institute, Arizona State University, Tempe, AZ, United States
| | - Liqiang Zhang
- Center for Personalized Diagnostics and Center for Immunotherapy, Vaccines and Virotherapy, The Biodesign Institute, Arizona State University, Tempe, AZ, United States
| | - Alexandra R Lucas
- Center for Personalized Diagnostics and Center for Immunotherapy, Vaccines and Virotherapy, The Biodesign Institute, Arizona State University, Tempe, AZ, United States
| |
Collapse
|
24
|
Reiner AP, Raffield LM, Franceschini N, Auer PL, Lange EM, Nickerson DA, Zakai NA, Correa A, Olson N. Effect of Sickle Cell Trait and APOL1 Genotype on the Association of Soluble uPAR with Kidney Function Measures in Black Americans. Clin J Am Soc Nephrol 2021; 16:287-289. [PMID: 33268503 PMCID: PMC7863645 DOI: 10.2215/cjn.12100720] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Affiliation(s)
- Alexander P Reiner
- Department of Epidemiology, University of Washington, Seattle, Washington
| | - Laura M Raffield
- Department of Genetics, University of North Carolina, Chapel Hill, North Carolina
| | - Nora Franceschini
- Department of Epidemiology, University of North Carolina, Chapel Hill, North Carolina
| | - Paul L Auer
- Department of Biostatistics, Joseph J. Zilber School of Public Health, University of Wisconsin-Milwaukee, Milwaukee, Wisconsin
| | - Ethan M Lange
- Division of Biomedical Informatics and Personalized Medicine, School of Medicine, University of Colorado, Anschutz Medical Campus, Aurora, Colorado
| | - Deborah A Nickerson
- Department of Genome Sciences, University of Washington, Seattle, Washington
| | - Neil A Zakai
- Department of Medicine, Larner College of Medicine at the University of Vermont, Burlington, Vermont
- Department of Pathology and Laboratory Medicine, Larner College of Medicine at the University of Vermont, Burlington, Vermont
| | - Adolfo Correa
- Department of Pediatrics and Medicine, University of Mississippi Medical Center, Jackson, Mississippi
| | - Nels Olson
- Department of Pathology and Laboratory Medicine, Larner College of Medicine at the University of Vermont, Burlington, Vermont
| |
Collapse
|
25
|
Dupuy AM, Kuster N, Bargnoux AS, Aguilhon S, Huet F, Leclercq F, Pasquié JL, Roubille F, Cristol JP. Long term pronostic value of suPAR in chronic heart failure: reclassification of patients with low MAGGIC score. Clin Chem Lab Med 2021; 59:1299-1306. [PMID: 33544524 DOI: 10.1515/cclm-2020-0903] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2020] [Accepted: 01/22/2021] [Indexed: 12/21/2022]
Abstract
OBJECTIVES Inflammation is a hallmark of heart failure (HF) and among inflammatory biomarkers, the most studied remains the C-reactive protein (CRP). In recent years several biomarkers have emerged, such as sST2 and soluble urokinase-type plasminogen activator receptor (suPAR). This study set out to examine the relative importance of long-time prognostic strength of suPAR and the potential additive information on patient risk with chronic HF in comparison with pronostic value of CRP and sST2. METHODS Demographics, clinical and biological variables were assessed in a total of 182 patients with chronic HF over median follow-up period of 80 months. Inflammatory biomarkers (i.e., CRP, sST2, and suPAR) were performed. RESULTS In univariate Cox regression analysis age, NYHA class, MAGGIC score and the five biomarkers (N-terminal pro brain natriuretic peptide [NT-proBNP], high-sensitive cardiac troponin T [hs-cTnT], CRP, sST2, and suPAR) were associated with both all-cause and cardiovascular mortality. In the multivariate model, only NT-proBNP, suPAR, and MAGGIC score remained independent predictors of all-cause mortality as well as of cardiovascular mortality. Risk classification analysis was significantly improved with the addition of suPAR particularly for all-cause short- and long-term mortality. Using a classification tree approach, the same three variables could be considered as significant classifier variables to predict all-cause or cardiovascular mortality and an algorithm were reported. We demonstrated the favorable outcome associated with patients with a low MAGGIC score and a low suPAR level by comparison to patients with low MAGGIC score but high suPAR values. CONCLUSIONS The main findings of our study are (1) that among the three inflammatory biomarkers, only suPAR levels were independently associated with 96-month mortality for patients with chronic HF and (2) that an algorithm based on clinical score, a cardiomyocyte stress biomarker and an inflammatory biomarker could help to a more reliable long term risk stratification in heart failure.
Collapse
Affiliation(s)
- Anne Marie Dupuy
- Department of Biochemistry, Centre Ressources Biologiques de Montpellier, University Hospital of Montpellier, Montpellier, France
| | - Nils Kuster
- Department of Biochemistry, Centre Ressources Biologiques de Montpellier, University Hospital of Montpellier, Montpellier, France.,PhyMedExp, University of Montpellier, INSERM U1046, CNRS UMR 9214, Montpellier, France
| | - Anne Sophie Bargnoux
- Department of Biochemistry, Centre Ressources Biologiques de Montpellier, University Hospital of Montpellier, Montpellier, France.,PhyMedExp, University of Montpellier, INSERM U1046, CNRS UMR 9214, Montpellier, France
| | - Sylvain Aguilhon
- Cardiology Department, University Hospital of Montpellier, Montpellier, France
| | - Fabien Huet
- Cardiology Department, University Hospital of Montpellier, Montpellier, France
| | - Florence Leclercq
- Cardiology Department, University Hospital of Montpellier, Montpellier, France
| | - Jean-Luc Pasquié
- Cardiology Department, University Hospital of Montpellier, Montpellier, France
| | - François Roubille
- PhyMedExp, University of Montpellier, INSERM U1046, CNRS UMR 9214, Montpellier, France.,Cardiology Department, University Hospital of Montpellier, Montpellier, France
| | - Jean Paul Cristol
- Department of Biochemistry, Centre Ressources Biologiques de Montpellier, University Hospital of Montpellier, Montpellier, France.,PhyMedExp, University of Montpellier, INSERM U1046, CNRS UMR 9214, Montpellier, France
| |
Collapse
|
26
|
Hamie L, Eid E, Abbas O, Safi R, Nammour T, Tamim H, Makki M, Stephan C, Hasbani D, Wehbe H, Ghaoui N, Hawa M, Nasser N, Eid A, Kibbi AG, Kurban M. SuPAR, a potential inflammatory mediator in psoriasis pathogenesis. Clin Exp Pharmacol Physiol 2020; 47:1705-1712. [PMID: 32558953 DOI: 10.1111/1440-1681.13365] [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: 05/09/2020] [Revised: 06/01/2020] [Accepted: 06/12/2020] [Indexed: 11/29/2022]
Abstract
Psoriasis is an inflammatory skin disorder that is strongly associated with the metabolic syndrome. The sole reliance on clinical examination to guide prognostication and treatment is insufficient at best; accurate diagnostic and prognostic psoriatic molecular biomarkers are needed. Soluble urokinase plasminogen activator receptor (suPAR) has been implicated in inflammation. The aim of this study is to determine whether suPAR plays a role in the pathogenesis of psoriasis and whether an association exists between suPAR levels, disease severity, and other variables like insulin, erythrocyte sedimentation rate (ESR) and C-reactive protein (CRP). This study also compares the pattern of uPAR staining in healthy vs psoriatic skin: 39 psoriatic and 30 control subjects were included. Two biopsies (affected and unaffected skin) and one biopsy were taken from psoriasis patients and healthy controls, respectively, with uPAR staining of all skin biopsies. Blood samples from all subjects were obtained to determine suPAR, ESR, CRP, and fasting insulin levels. uPAR staining was prominent in unaffected skin from psoriasis patients and healthy individuals vs weak/absent uPAR staining in psoriatic skin. CRP, ESR and suPAR levels were not significantly elevated in the mild psoriasis group compared to healthy controls. The loss of epidermal uPAR is suggestive of its tentative role in the pathogenesis of psoriasis. Patients with mild-moderate psoriasis possibly lack the powerful association attributed to metabolic syndrome in psoriatic patients. Further studies on larger cohorts are needed to ascertain the validity of the mentioned conclusions.
Collapse
Affiliation(s)
- Lamiaa Hamie
- Department of Dermatology, American University of Beirut Medical Center, Beirut, Lebanon
| | - Edward Eid
- Department of Dermatology, American University of Beirut Medical Center, Beirut, Lebanon
| | - Ossama Abbas
- Department of Dermatology, American University of Beirut Medical Center, Beirut, Lebanon
| | - Remi Safi
- Department of Biochemistry and Molecular Genetics, American University of Beirut, Beirut, Lebanon
| | - Tarak Nammour
- Department of Gastroenterology, American University of Beirut Medical Center, Beirut, Lebanon
| | - Hani Tamim
- Clinical Research Institute, American University of Beirut Medical Center, Beirut, Lebanon
| | - Maha Makki
- Clinical Research Institute, American University of Beirut Medical Center, Beirut, Lebanon
| | - Carla Stephan
- Department of Dermatology, American University of Beirut Medical Center, Beirut, Lebanon
| | - Divina Hasbani
- Department of Internal Medicine, American University of Beirut Medical Center, Beirut, Lebanon
| | - Hisham Wehbe
- Department of Internal Medicine, American University of Beirut Medical Center, Beirut, Lebanon
| | - Nohra Ghaoui
- Department of Internal Medicine, American University of Beirut Medical Center, Beirut, Lebanon
| | - Mariana Hawa
- Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| | - Nourhan Nasser
- Department of Dermatology, American University of Beirut Medical Center, Beirut, Lebanon
| | - Assaad Eid
- Department of Anatomy, Cell Biology and Physiology, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| | - Abdul-Ghani Kibbi
- Department of Dermatology, American University of Beirut Medical Center, Beirut, Lebanon
| | - Mazen Kurban
- Department of Dermatology, American University of Beirut Medical Center, Beirut, Lebanon
- Department of Biochemistry and Molecular Genetics, American University of Beirut, Beirut, Lebanon
| |
Collapse
|
27
|
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: 7] [Impact Index Per Article: 1.4] [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
|
28
|
Luo M, Li P, Niu W. Major Concerns Over Improving Measurement of Inflammation Remain. JAMA Pediatr 2020; 174:624. [PMID: 32338729 DOI: 10.1001/jamapediatrics.2020.0284] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/14/2022]
Affiliation(s)
- Minjing Luo
- Graduate School, Beijing University of Chinese Medicine, Beijing, China.,Institute of Clinical Medical Sciences, China-Japan Friendship Hospital, Beijing, China
| | - Ping Li
- Institute of Clinical Medical Sciences, China-Japan Friendship Hospital, Beijing, China
| | - Wenquan Niu
- Institute of Clinical Medical Sciences, China-Japan Friendship Hospital, Beijing, China
| |
Collapse
|
29
|
Jirak P, Pistulli R, Lichtenauer M, Wernly B, Paar V, Motloch LJ, Rezar R, Jung C, Hoppe UC, Schulze PC, Kretzschmar D, Braun-Dullaeus RC, Bekfani T. Expression of the Novel Cardiac Biomarkers sST2, GDF-15, suPAR, and H-FABP in HFpEF Patients Compared to ICM, DCM, and Controls. J Clin Med 2020; 9:jcm9041130. [PMID: 32326570 PMCID: PMC7230638 DOI: 10.3390/jcm9041130] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2020] [Revised: 04/03/2020] [Accepted: 04/08/2020] [Indexed: 12/11/2022] Open
Abstract
Background: Heart failure with preserved ejection fraction (HFpEF) remains an ongoing therapeutic and diagnostic challenge to date. In this study we aimed for an analysis of the diagnostic potential of four novel cardiovascular biomarkers, GDF-15, H-FABP, sST2, and suPAR in HFpEF patients compared to controls as well as ICM, and DCM. Methods: In total, we included 252 stable outpatients and controls (77 DCM, 62 ICM, 18 HFpEF, and 95 controls) in the present study. All patients were in a non-decompensated state and on a stable treatment regimen. Serum samples were obtained and analyzed for GDF-15 (inflammation, remodeling), H-FABP (ischemia and subclinical ischemia), sST2 (inflammation, remodeling) and suPAR (inflammation, remodeling) by means of ELISA. Results: A significant elevation of GDF-15 was found for all heart failure entities compared to controls (p < 0.005). Similarly, H-FABP evidenced a significant elevation in all heart failure entities compared to the control group (p < 0.0001). Levels of sST2 were significantly elevated in ICM and DCM patients compared to the control group and HFpEF patients (p < 0.0001). Regarding suPAR, a significant elevation in ICM and DCM patients compared to the control group (p < 0.0001) and HFpEF patients (p < 0.01) was observed. An AUC analysis identified H-FABP (0.792, 95% CI 0.713–0.870) and GDF-15 (0.787, 95% CI 0.696–0.878) as paramount diagnostic biomarkers for HFpEF patients. Conclusion: Based on their differences in secretion patterns, novel cardiovascular biomarkers might represent a promising diagnostic tool for HFpEF in the future.
Collapse
Affiliation(s)
- Peter Jirak
- Clinic of Internal Medicine II, Department of Cardiology, Paracelsus Medical University of Salzburg, 5020 Salzburg, Austria; (M.L.); (B.W.); (V.P.); (L.J.M.); (R.R.); (U.C.H.)
- Correspondence:
| | - Rudin Pistulli
- Division of Vascular Medicine, Department of Cardiology and Angiology, University Hospital Muenster, Albert-Schweitzer-Campus 1, Munster, North Rhine-Westphalia, 48149 Münster, Germany;
| | - Michael Lichtenauer
- Clinic of Internal Medicine II, Department of Cardiology, Paracelsus Medical University of Salzburg, 5020 Salzburg, Austria; (M.L.); (B.W.); (V.P.); (L.J.M.); (R.R.); (U.C.H.)
| | - Bernhard Wernly
- Clinic of Internal Medicine II, Department of Cardiology, Paracelsus Medical University of Salzburg, 5020 Salzburg, Austria; (M.L.); (B.W.); (V.P.); (L.J.M.); (R.R.); (U.C.H.)
| | - Vera Paar
- Clinic of Internal Medicine II, Department of Cardiology, Paracelsus Medical University of Salzburg, 5020 Salzburg, Austria; (M.L.); (B.W.); (V.P.); (L.J.M.); (R.R.); (U.C.H.)
| | - Lukas J. Motloch
- Clinic of Internal Medicine II, Department of Cardiology, Paracelsus Medical University of Salzburg, 5020 Salzburg, Austria; (M.L.); (B.W.); (V.P.); (L.J.M.); (R.R.); (U.C.H.)
| | - Richard Rezar
- Clinic of Internal Medicine II, Department of Cardiology, Paracelsus Medical University of Salzburg, 5020 Salzburg, Austria; (M.L.); (B.W.); (V.P.); (L.J.M.); (R.R.); (U.C.H.)
| | - Christian Jung
- Division of Cardiology, Pulmonology, and Vascular Medicine, Medical Faculty, University Duesseldorf, 40225 Duesseldorf, Germany;
| | - Uta C. Hoppe
- Clinic of Internal Medicine II, Department of Cardiology, Paracelsus Medical University of Salzburg, 5020 Salzburg, Austria; (M.L.); (B.W.); (V.P.); (L.J.M.); (R.R.); (U.C.H.)
| | - P. Christian Schulze
- Department of Internal Medicine I, Division of Cardiology, Angiology, Pneumology and Intensive Medical Care, University Hospital Jena, Friedrich Schiller University Jena, 07740 Jena, Germany; (P.C.S.); (D.K.)
| | - Daniel Kretzschmar
- Department of Internal Medicine I, Division of Cardiology, Angiology, Pneumology and Intensive Medical Care, University Hospital Jena, Friedrich Schiller University Jena, 07740 Jena, Germany; (P.C.S.); (D.K.)
| | - Rüdiger C. Braun-Dullaeus
- Department of Internal Medicine I, Division of Cardiology, Angiology and Intensive Medical Care, University Hospital Magdeburg, Otto von Gericke University, Magdeburg, 39120 Magdeburg, Germany; (R.C.B.-D.)
| | - Tarek Bekfani
- Department of Internal Medicine I, Division of Cardiology, Angiology and Intensive Medical Care, University Hospital Magdeburg, Otto von Gericke University, Magdeburg, 39120 Magdeburg, Germany; (R.C.B.-D.)
| |
Collapse
|
30
|
Mehta A, Desai SR, Ko YA, Liu C, Dhindsa DS, Nayak A, Hooda A, Martini MA, Ejaz K, Sperling LS, Reiser J, Hayek SS, Quyyumi AA. Sex Differences in Circulating Soluble Urokinase-Type Plasminogen Activator Receptor (suPAR) Levels and Adverse Outcomes in Coronary Artery Disease. J Am Heart Assoc 2020; 9:e015457. [PMID: 32089048 PMCID: PMC7335555 DOI: 10.1161/jaha.119.015457] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Background Women have higher circulating levels of soluble urokinase‐type plasminogen activator receptor (suPAR), and elevated suPAR is associated with cardiovascular risk. The independent association of sex with suPAR and the impact of sex on its association with cardiovascular risk are unknown. Methods and Results Plasma suPAR was measured using ELISA in 2 cohorts of 666 asymptomatic individuals (49 years, 65% women) and 4184 patients with coronary artery disease (63 years, 37% women). Independent association of sex with suPAR was studied using linear regression models adjusted for demographics, risk factors, and visceral adiposity in asymptomatic participants. Impact of sex on association of suPAR with all‐cause mortality was studied in patients with coronary artery disease using multivariable‐adjusted Cox models. Sex‐specific suPAR cutoffs for predicting all‐cause mortality were calculated. Asymptomatic women had 10% higher suPAR compared with men after adjusting for confounders, and visceral adiposity partly accounted for this association. Over a median follow‐up of 5.2 years, 795 deaths were recorded in patients with coronary artery disease. Log2‐transformed suPAR was independently associated with mortality (hazard ratio per 1‐SD 1.72, 95% CI 1.60–1.85) and an interaction with sex was noted (P=0.005). Association of suPAR with mortality was slightly weaker in women (hazard ratio 1.61, 95% CI 1.41–1.83) compared with men (hazard ratio 1.83, 95% CI 1.67–2.00). However, using sex‐specific suPAR cut‐offs (4392 pg/mL for women and 3187 pg/mL for men), a similar mortality incidence was observed for both sexes (38.5% and 35.5%, respectively, P=0.3). Conclusions Women have 10% higher plasma suPAR levels compared with men. Elevated sex‐specific plasma suPAR levels are equally predictive of risk of adverse events in both sexes.
Collapse
Affiliation(s)
- Anurag Mehta
- Division of Cardiology Department of Medicine Emory Clinical Cardiovascular Research Institute Emory University School of Medicine Atlanta GA
| | - Shivang R Desai
- Department of Medicine Emory University School of Medicine Atlanta GA
| | - Yi-An Ko
- Department of Biostatistics and Bioinformatics Emory University Atlanta GA
| | - Chang Liu
- Department of Biostatistics and Bioinformatics Emory University Atlanta GA
| | - Devinder S Dhindsa
- Division of Cardiology Department of Medicine Emory Clinical Cardiovascular Research Institute Emory University School of Medicine Atlanta GA
| | - Aditi Nayak
- Division of Cardiology Department of Medicine Emory Clinical Cardiovascular Research Institute Emory University School of Medicine Atlanta GA
| | - Ananya Hooda
- Division of Cardiology Department of Medicine Emory Clinical Cardiovascular Research Institute Emory University School of Medicine Atlanta GA
| | - Mohamed A Martini
- Division of Cardiology Department of Medicine Emory Clinical Cardiovascular Research Institute Emory University School of Medicine Atlanta GA
| | - Kiran Ejaz
- Division of Cardiology Department of Medicine Emory Clinical Cardiovascular Research Institute Emory University School of Medicine Atlanta GA
| | - Laurence S Sperling
- Division of Cardiology Department of Medicine Emory Clinical Cardiovascular Research Institute Emory University School of Medicine Atlanta GA
| | - Jochen Reiser
- Department of Internal Medicine Rush University Medical Center Chicago IL
| | - Salim S Hayek
- Frankel Cardiovascular Center University of Michigan Ann Arbor MI
| | - Arshed A Quyyumi
- Division of Cardiology Department of Medicine Emory Clinical Cardiovascular Research Institute Emory University School of Medicine Atlanta GA
| |
Collapse
|
31
|
Wei C, Zhu K, Reiser J. Soluble Urokinase Receptor and Liver Disease. Clin Liver Dis (Hoboken) 2019; 14:163-166. [PMID: 31879556 PMCID: PMC6924968 DOI: 10.1002/cld.850] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/26/2019] [Accepted: 06/07/2019] [Indexed: 02/04/2023] Open
Affiliation(s)
- Changli Wei
- Department of Internal MedicineRush University Medical CenterChicagoIL
| | - Ke Zhu
- Department of Internal MedicineRush University Medical CenterChicagoIL
| | - Jochen Reiser
- Department of Internal MedicineRush University Medical CenterChicagoIL
| |
Collapse
|
32
|
Li J, Fukase Y, Shang Y, Zou W, Muñoz-Félix JM, Buitrago L, van Agthoven J, Zhang Y, Hara R, Tanaka Y, Okamoto R, Yasui T, Nakahata T, Imaeda T, Aso K, Zhou Y, Locuson C, Nesic D, Duggan M, Takagi J, Vaughan RD, Walz T, Hodivala-Dilke K, Teitelbaum SL, Arnaout MA, Filizola M, Foley MA, Coller BS. Novel Pure αVβ3 Integrin Antagonists That Do Not Induce Receptor Extension, Prime the Receptor, or Enhance Angiogenesis at Low Concentrations. ACS Pharmacol Transl Sci 2019; 2:387-401. [PMID: 32259072 PMCID: PMC7088984 DOI: 10.1021/acsptsci.9b00041] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2019] [Indexed: 01/12/2023]
Abstract
The integrin αVβ3 receptor has been implicated in several important diseases, but no antagonists are approved for human therapy. One possible limitation of current small-molecule antagonists is their ability to induce a major conformational change in the receptor that induces it to adopt a high-affinity ligand-binding state. In response, we used structural inferences from a pure peptide antagonist to design the small-molecule pure antagonists TDI-4161 and TDI-3761. Both compounds inhibit αVβ3-mediated cell adhesion to αVβ3 ligands, but do not induce the conformational change as judged by antibody binding, electron microscopy, X-ray crystallography, and receptor priming studies. Both compounds demonstrated the favorable property of inhibiting bone resorption in vitro, supporting potential value in treating osteoporosis. Neither, however, had the unfavorable property of the αVβ3 antagonist cilengitide of paradoxically enhancing aortic sprout angiogenesis at concentrations below its IC50, which correlates with cilengitide's enhancement of tumor growth in vivo.
Collapse
Affiliation(s)
- Jihong Li
- Allen and
Frances Adler Laboratory of Blood and Vascular Biology, Rockefeller University, 1230 York Avenue, New York, New York 10065, United States
| | - Yoshiyuki Fukase
- Tri-Institutional
Therapeutics Discovery Institute, 413 East 69 Street, New York, New York 10021, United
States
| | - Yi Shang
- Department
of Pharmacological Sciences, Icahn School
of Medicine at Mount Sinai, One Gustave L. Levy Place, Box 1677, New York, New York 10029-6574, United States
| | - Wei Zou
- Washington
University School of Medicine, Campus Box 8118, 660 South Euclid Avenue, St. Louis, Missouri 63110, United States
| | - José M. Muñoz-Félix
- Adhesion
and Angiogenesis Laboratory, Centre for Tumour Biology, Barts Cancer Institute—a CR-UK Centre of Excellence,
Queen Mary University of London, John Vane Science Centre, Charterhouse Square, London, EC1M 6BQ, United Kingdom
| | - Lorena Buitrago
- Allen and
Frances Adler Laboratory of Blood and Vascular Biology, Rockefeller University, 1230 York Avenue, New York, New York 10065, United States
| | - Johannes van Agthoven
- Leukocyte
Biology and Inflammation and Structural Biology Programs, Division
of Nephrology, Massachusetts General Hospital
and Harvard Medical School, 149 13th Street, Charlestown, Massachusetts 02129, United States
| | - Yixiao Zhang
- Laboratory
of Molecular Electron Microscopy, Rockefeller
University, 1230 York Avenue, New York, New York 10065, United
States
| | - Ryoma Hara
- Tri-Institutional
Therapeutics Discovery Institute, 413 East 69 Street, New York, New York 10021, United
States
| | - Yuta Tanaka
- Tri-Institutional
Therapeutics Discovery Institute, 413 East 69 Street, New York, New York 10021, United
States
| | - Rei Okamoto
- Tri-Institutional
Therapeutics Discovery Institute, 413 East 69 Street, New York, New York 10021, United
States
| | - Takeshi Yasui
- Tri-Institutional
Therapeutics Discovery Institute, 413 East 69 Street, New York, New York 10021, United
States
| | - Takashi Nakahata
- Tri-Institutional
Therapeutics Discovery Institute, 413 East 69 Street, New York, New York 10021, United
States
| | - Toshihiro Imaeda
- Tri-Institutional
Therapeutics Discovery Institute, 413 East 69 Street, New York, New York 10021, United
States
| | - Kazuyoshi Aso
- Tri-Institutional
Therapeutics Discovery Institute, 413 East 69 Street, New York, New York 10021, United
States
| | - Yuchen Zhou
- Department
of Pharmacological Sciences, Icahn School
of Medicine at Mount Sinai, One Gustave L. Levy Place, Box 1677, New York, New York 10029-6574, United States
| | - Charles Locuson
- Agios Pharmaceuticals, 88 Sidney Street, Cambridge, Massachusetts 02139-4169, United States
| | - Dragana Nesic
- Allen and
Frances Adler Laboratory of Blood and Vascular Biology, Rockefeller University, 1230 York Avenue, New York, New York 10065, United States
| | - Mark Duggan
- LifeSci
Consulting, LLC, 18243
SE Ridgeview Drive, Tequesta, Florida 33469, United
States
| | - Junichi Takagi
- Laboratory
of Protein Synthesis and Expression, Institute for Protein Research, Osaka University, 3-2 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Roger D. Vaughan
- Rockefeller
University Center for Clinical and Translational Science, Rockefeller University, 2130 York Avenue, New York, New York 10065, United States
| | - Thomas Walz
- Laboratory
of Molecular Electron Microscopy, Rockefeller
University, 1230 York Avenue, New York, New York 10065, United
States
| | - Kairbaan Hodivala-Dilke
- Adhesion
and Angiogenesis Laboratory, Centre for Tumour Biology, Barts Cancer Institute—a CR-UK Centre of Excellence,
Queen Mary University of London, John Vane Science Centre, Charterhouse Square, London, EC1M 6BQ, United Kingdom
| | - Steven L. Teitelbaum
- Washington
University School of Medicine, Campus Box 8118, 660 South Euclid Avenue, St. Louis, Missouri 63110, United States
| | - M. Amin Arnaout
- Leukocyte
Biology and Inflammation and Structural Biology Programs, Division
of Nephrology, Massachusetts General Hospital
and Harvard Medical School, 149 13th Street, Charlestown, Massachusetts 02129, United States
| | - Marta Filizola
- Department
of Pharmacological Sciences, Icahn School
of Medicine at Mount Sinai, One Gustave L. Levy Place, Box 1677, New York, New York 10029-6574, United States
| | - Michael A. Foley
- Tri-Institutional
Therapeutics Discovery Institute, 413 East 69 Street, New York, New York 10021, United
States
| | - Barry S. Coller
- Allen and
Frances Adler Laboratory of Blood and Vascular Biology, Rockefeller University, 1230 York Avenue, New York, New York 10065, United States
| |
Collapse
|
33
|
Zhang Q, Li L, Chen H, Zhang G, Zhu S, Kong R, Chen H, Wang G, Sun B. Soluble urokinase plasminogen activator receptor associates with higher risk, advanced disease severity as well as inflammation, and might serve as a prognostic biomarker of severe acute pancreatitis. J Clin Lab Anal 2019; 34:e23097. [PMID: 31774228 PMCID: PMC7083411 DOI: 10.1002/jcla.23097] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2019] [Revised: 10/02/2019] [Accepted: 10/09/2019] [Indexed: 12/15/2022] Open
Abstract
Background This study aimed to explore the potential of soluble urokinase plasminogen activator receptor (suPAR) as a biomarker for severe acute pancreatitis (SAP) risk prediction and disease management in SAP patients. Methods Totally 225 acute pancreatitis (AP) patients (including 75 SAP, 75 moderate‐severe acute pancreatitis [MSAP], and 75 mild acute pancreatitis [MAP] patients) were recruited based on the Atlanta classification, and their serum samples were obtained within 24 hours after admission. Meanwhile, 75 health controls (HCs) were recruited with their serum samples collected at the enrollment. The serum suPAR was then detected using enzyme‐linked immunosorbent assay. Results The suPAR level was increased in SAP patients compared with MSAP patients (P = .023), MAP patients (P < .001), and HCs (P < .001). Receiver operating characteristic (ROC) curve presented that suPAR could not only differentiate SAP patients from HCs (AUC: 0.920, 95%CI: 0.875‐0.965) but also differentiate SAP patients from MSAP (AUC: 0.684, 95%CI: 0.600‐0.769) and MAP patients (AUC: 0.855, 95%CI: 0.797‐0.912). In SAP patients, suPAR was positively correlated with Ranson score (P < .001), acute physiology and chronic healthcare evaluation II score (P = .001), sequential organ failure assessment score (P < .001), and C‐reaction protein (P = .002). Further ROC curve exhibited that suPAR (AUC: 0.806, 95%CI: 0.663‐0.949) was of good value in predicting increased inhospital mortality of SAP patients. Conclusion Soluble urokinase plasminogen activator receptor is of good predictive value for SAP risk and may serve as a potential biomarker for disease severity, inflammation, and inhospital mortality in SAP patients.
Collapse
Affiliation(s)
- Qi Zhang
- Department of Pancreatic and Biliary Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, China.,Key Laboratory of Hepatosplenic Surgery, Ministry of Education, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Le Li
- Department of Pancreatic and Biliary Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, China.,Key Laboratory of Hepatosplenic Surgery, Ministry of Education, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Hongze Chen
- Department of Pancreatic and Biliary Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, China.,Key Laboratory of Hepatosplenic Surgery, Ministry of Education, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Guangquan Zhang
- Department of Pancreatic and Biliary Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, China.,Key Laboratory of Hepatosplenic Surgery, Ministry of Education, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Siqiang Zhu
- Department of Pancreatic and Biliary Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, China.,Key Laboratory of Hepatosplenic Surgery, Ministry of Education, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Rui Kong
- Department of Pancreatic and Biliary Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, China.,Key Laboratory of Hepatosplenic Surgery, Ministry of Education, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Hua Chen
- Department of Pancreatic and Biliary Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, China.,Key Laboratory of Hepatosplenic Surgery, Ministry of Education, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Gang Wang
- Department of Pancreatic and Biliary Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, China.,Key Laboratory of Hepatosplenic Surgery, Ministry of Education, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Bei Sun
- Department of Pancreatic and Biliary Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, China.,Key Laboratory of Hepatosplenic Surgery, Ministry of Education, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| |
Collapse
|
34
|
Shao SL, Cong HY, Wang MY, Liu P. The diagnostic roles of neutrophil in bloodstream infections. Immunobiology 2019; 225:151858. [PMID: 31836303 DOI: 10.1016/j.imbio.2019.10.007] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2019] [Revised: 04/15/2019] [Accepted: 10/15/2019] [Indexed: 12/21/2022]
Abstract
Bloodstream infections remain a leading cause of death worldwide, despite advances in critical care and understanding of the pathophysiology and treatment strategies. No specific biomarkers or therapy are available for these conditions. Neutrophils play a critical role in controlling infection and it is suggested that their migration and antimicrobial activity are impaired during sepsis which contribute to the dysregulation of immune responses. Recent studies further demonstrated that interruption or reversal of the impaired migration and antimicrobial function of neutrophils improves the outcome of sepsis in animal models. In this review, we provide an overview of the associated diagnostic biomarkers involved neutrophils in sepsis, and discuss the potential of neutrophils as a target to specifically predict the outcome of sepsis.
Collapse
Affiliation(s)
- Shu-Li Shao
- Department of Central Lab, Weihai Municipal Hospital Affiliated to Dalian Medical University, Weihai, Shandong, 264200, PR China
| | - Hai-Yan Cong
- Department of Central Lab, Weihai Municipal Hospital Affiliated to Dalian Medical University, Weihai, Shandong, 264200, PR China
| | - Ming-Yi Wang
- Department of Central Lab, Weihai Municipal Hospital Affiliated to Dalian Medical University, Weihai, Shandong, 264200, PR China.
| | - Peng Liu
- Department of Central Lab, Weihai Municipal Hospital Affiliated to Dalian Medical University, Weihai, Shandong, 264200, PR China.
| |
Collapse
|
35
|
Shuai T, Pei Jing Y, Huang Q, Xiong H, Liu J, Zhu L, Yang K, Jian L. Serum soluble urokinase type plasminogen activated receptor and focal segmental glomerulosclerosis: a systematic review and meta-analysis. BMJ Open 2019; 9:e031812. [PMID: 31594897 PMCID: PMC6797292 DOI: 10.1136/bmjopen-2019-031812] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/31/2019] [Revised: 09/04/2019] [Accepted: 09/10/2019] [Indexed: 01/10/2023] Open
Abstract
OBJECTIVES Soluble urokinase plasminogen activated receptor (suPAR) is a biomarker that may predict the occurrence of focal segmental glomerulosclerosis (FSGS); however, there is still controversy about whether suPAR can predict FSGS. In this study, we performed a systematic evaluation and meta-analysis to prove whether suPAR can predict FSGS, and to detect a threshold concentration of suPAR that can be used to diagnose FSGS. In addition, a threshold concentration of suPAR for the diagnosis of FSGS was proposed. DESIGN Systematic review and meta-analysis. DATA SOURCES We systematically searched PubMed, Embase, Cochrane Library, Web of Science and China Biology Medicine databases for studies published from the inception dates to 1 December 2018. ELIGIBILITY CRITERIA: (1) Data involving the suPAR level were from blood samples; (2) FSGS was diagnosed by biopsy; and (3) randomised controlled trials, cohort studies, case-control studies and cross-sectional studies. DATA EXTRACTION AND SYNTHESIS Initially, a total of 364 studies were searched, among which 29 studies were finally included. In addition, seven studies described the cut-off value of suPAR, which ranged from 2992.6 to 5500 pg/mL. RESULTS The results showed that the suPAR levels in the primary FSGS group were significantly higher when compared with that in the normal control group (p<0.001; standard mean difference (SMD): 2.56; 95% CI 1.85 to 3.28), and significant differences were observed in the secondary FSGS and in the normal control group (p<0.001; SMD: 1.68; 95% CI 1.37 to 1.98). A suPAR concentration of 3000 pg/mL may be the best threshold for the diagnosis of primary FSGS (sensitivity=0.72; specificity=0.88; area under the curve=0.85). CONCLUSION Our results suggested that suPAR might be a potential biomarker for predicting primary and secondary FSGS. In addition, our data showed that a suPAR concentration of 3000 pg/mL might be used as a threshold for the diagnosis of FSGS. TRIAL REGISTRATION NUMBER CRD42019120948.
Collapse
Affiliation(s)
- Tiankui Shuai
- Department of Intensive Care Unit, Lanzhou University First Affiliated Hospital, Lanzhou, China
- Evidence-Based Medicine Center, School of Basic Medical Sciences, Lanzhou University, Lanzhou, China
- The First Clinical Medical College of the First Hospital of Lanzhou University, Lanzhou, China
| | - Yan Pei Jing
- Evidence-Based Medicine Center, School of Basic Medical Sciences, Lanzhou University, Lanzhou, China
- Institute of Clinical Research and Evidence Based Medicine, Gansu Province People's Hospital, Lanzhou, China
| | - Qiangru Huang
- Department of Intensive Care Unit, Lanzhou University First Affiliated Hospital, Lanzhou, China
- Evidence-Based Medicine Center, School of Basic Medical Sciences, Lanzhou University, Lanzhou, China
- The First Clinical Medical College of the First Hospital of Lanzhou University, Lanzhou, China
| | - Huaiyu Xiong
- Department of Intensive Care Unit, Lanzhou University First Affiliated Hospital, Lanzhou, China
- Evidence-Based Medicine Center, School of Basic Medical Sciences, Lanzhou University, Lanzhou, China
- The First Clinical Medical College of the First Hospital of Lanzhou University, Lanzhou, China
| | - Jingjing Liu
- Department of Intensive Care Unit, Lanzhou University First Affiliated Hospital, Lanzhou, China
- Evidence-Based Medicine Center, School of Basic Medical Sciences, Lanzhou University, Lanzhou, China
- The First Clinical Medical College of the First Hospital of Lanzhou University, Lanzhou, China
| | - Lei Zhu
- Department of Intensive Care Unit, Lanzhou University First Affiliated Hospital, Lanzhou, China
- Evidence-Based Medicine Center, School of Basic Medical Sciences, Lanzhou University, Lanzhou, China
- The First Clinical Medical College of the First Hospital of Lanzhou University, Lanzhou, China
| | - Kehu Yang
- Evidence-Based Medicine Center, School of Basic Medical Sciences, Lanzhou University, Lanzhou, China
- Institute of Clinical Research and Evidence Based Medicine, Gansu Province People's Hospital, Lanzhou, China
- Evidence Based Social Science Research Center, Lanzhou University, Lanzhou, China
- Institute of Evidence Based Rehabilitation Medicine of Gansu Province, Lanzhou University, Lanzhou, China
- Key Laboratory of Evidence Based Medicine and Knowledge Translation of Gansu Province, Lanzhou, China
| | - Liu Jian
- Department of Intensive Care Unit, Lanzhou University First Affiliated Hospital, Lanzhou, China
- The First Clinical Medical College of the First Hospital of Lanzhou University, Lanzhou, China
| |
Collapse
|
36
|
Chen D, Wu X, Yang J, Yu L. Serum plasminogen activator urokinase receptor predicts elevated risk of acute respiratory distress syndrome in patients with sepsis and is positively associated with disease severity, inflammation and mortality. Exp Ther Med 2019; 18:2984-2992. [PMID: 31555383 PMCID: PMC6755407 DOI: 10.3892/etm.2019.7931] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2018] [Accepted: 06/13/2019] [Indexed: 12/21/2022] Open
Abstract
The present study aimed to evaluate the predictive value of serum soluble urokinase plasminogen activator receptor (suPAR) regarding the risk of acute respiratory distress syndrome (ARDS) in sepsis patients, and investigate its correlation/association with disease severity, inflammation and mortality in sepsis patients with ARDS. A total of 57 sepsis patients with ARDS and 58 sepsis patients without ARDS were recruited for the present case-control study. Laboratory tests, acute physiology and chronic health evaluation (APACHE) II score and sequential organ failure assessment (SOFA) score were evaluated, and mortality during hospitalization was recorded. Blood samples were collected and serum suPAR was detected by ELISA. Furthermore, tumor necrosis factor (TNF)-α, interleukin (IL)-1β, IL-6, IL-8, IL-10 and IL-17, as well as C-reactive protein (CRP) were detected. The results indicated that the serum levels of suPAR in sepsis patients with ARDS were higher than those in sepsis patients without ARDS. Receiver operating characteristics (ROC) curve analysis indicated that it was possible to distinguish sepsis patients with ARDS from sepsis patients without ARDS based on their serum suPAR levels, and multivariate logistic regression analysis suggested that serum suPAR levels were an independent predictor of the risk of ARDS in sepsis patients. In sepsis patients with ARDS, serum suPAR levels were positively correlated with the APACHE II score, SOFA score and the levels of CRP, TNF-α, IL-1β and IL-8. In addition, serum suPAR levels were lower in survivors compared with those in non-survivors, and ROC curve analysis suggested that serum suPAR was able to predict the probability of mortality. In conclusion, serum suPAR independently predicted an elevated risk of ARDS in patients with sepsis, and was correlated/associated with greater disease severity, higher inflammation and increased mortality in patients with sepsis and ARDS.
Collapse
Affiliation(s)
- Dan Chen
- Department of Intensive Care Unit, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430014, P.R. China
| | - Xiaoling Wu
- Department of Intensive Care Unit, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430014, P.R. China
| | - Junhui Yang
- Department of Intensive Care Unit, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430014, P.R. China
| | - Li Yu
- Department of Intensive Care Unit, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430014, P.R. China
| |
Collapse
|
37
|
Gussen H, Hohlstein P, Bartneck M, Warzecha KT, Buendgens L, Luedde T, Trautwein C, Koch A, Tacke F. Neutrophils are a main source of circulating suPAR predicting outcome in critical illness. J Intensive Care 2019; 7:26. [PMID: 31061709 PMCID: PMC6487050 DOI: 10.1186/s40560-019-0381-5] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2019] [Accepted: 04/12/2019] [Indexed: 11/10/2022] Open
Abstract
Background Circulating levels of soluble urokinase plasminogen activation receptor (suPAR) have been proposed as a prognostic biomarker in patients with critical illness and sepsis. However, the origin of suPAR in sepsis has remained obscure. We investigated the potential cellular sources of suPAR by analyzing membrane-bound urokinase plasminogen activator receptor (uPAR, CD87) and evaluated its clinical relevance in critically ill patients. Methods We studied 87 critically ill patients (44 with sepsis, 43 without sepsis) from the medical intensive care unit (ICU) in comparison to 48 standard care patients with infections and 27 healthy controls in a prospective single-center non-interventional cohort study. Cellular uPAR expression of different immune cell subsets (by flow cytometry from peripheral blood) and corresponding serum suPAR concentrations were determined upon ICU admission and at day 3. Furthermore, we analyzed the effects of serum from sepsis patients on the activation and uPAR cleavage of primary human neutrophils and macrophages in vitro. Results In healthy controls, uPAR (CD87) expression was detected on nearly all blood neutrophils and monocytes, but only scarcely on lymphocytes. While uPAR expression on monocytes was maintained in ICU patients, only 58% of neutrophils from critically ill patients expressed uPAR, which was significantly lower than in healthy controls or standard care patients. Concomitantly, serum suPAR levels were significantly increased in ICU patients. We noted a clear inverse correlation between low neutrophilic uPAR and high serum suPAR in standard care and ICU patients, indicating that shedding of uPAR from activated neutrophils represents a main source of suPAR in systemic inflammation. Both low uPAR and high suPAR were closely associated with mortality in critically ill patients. Furthermore, serum from sepsis patients induced uPAR protein expression and subsequent receptor shedding on isolated primary neutrophils, but not on macrophages, in vitro. Conclusions The inverse correlation between low uPAR surface expression on neutrophils and high serum suPAR in critically ill patients supports that neutrophils are a main source of shed suPAR proteins in systemic inflammation. Furthermore, high suPAR levels and low neutrophilic uPAR expression predict mortality in ICU patients. Electronic supplementary material The online version of this article (10.1186/s40560-019-0381-5) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Hendrik Gussen
- 1Department of Medicine III, RWTH-University Hospital Aachen, Aachen, 52074 Germany
| | - Philipp Hohlstein
- 1Department of Medicine III, RWTH-University Hospital Aachen, Aachen, 52074 Germany
| | - Matthias Bartneck
- 1Department of Medicine III, RWTH-University Hospital Aachen, Aachen, 52074 Germany
| | | | - Lukas Buendgens
- 1Department of Medicine III, RWTH-University Hospital Aachen, Aachen, 52074 Germany
| | - Tom Luedde
- 1Department of Medicine III, RWTH-University Hospital Aachen, Aachen, 52074 Germany
| | - Christian Trautwein
- 1Department of Medicine III, RWTH-University Hospital Aachen, Aachen, 52074 Germany
| | - Alexander Koch
- 1Department of Medicine III, RWTH-University Hospital Aachen, Aachen, 52074 Germany
| | - Frank Tacke
- 1Department of Medicine III, RWTH-University Hospital Aachen, Aachen, 52074 Germany.,2Department of Hepatology/Gastroenterology, Charité University Medical Center, Augustenburger Platz 1, 13353 Berlin, Germany
| |
Collapse
|