1
|
Belvederi F, Leggeri S, Urbani A, Baroni S. suPAR as a biomarker of support in different clinical settings. Clin Chim Acta 2025; 573:120303. [PMID: 40222544 DOI: 10.1016/j.cca.2025.120303] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2025] [Revised: 04/09/2025] [Accepted: 04/09/2025] [Indexed: 04/15/2025]
Abstract
The urokinase-type plasminogen activator receptor (uPAR) system, which includes protease, receptor and inhibitors, is essential for key cellular functions like immune activation, cell migration, and tissue remodeling. Soluble uPAR (suPAR), released into circulation, serves as a valuable biomarker for systemic inflammation and immune activation. Elevated suPAR levels are associated with disease severity in conditions such as infections, sepsis, cardiovascular diseases, renal injury, cancer, and autoimmune diseases providing prognostic value especially in acute settings. Recent advancements in diagnostic methods, have enhanced the accuracy of suPAR measurement in serum and plasma. New rapid tests, such as suPARnostic Quick Triage, as well as turbidimetric assays, further expand its clinical applicability. In this review, we discuss the suPAR biomarker, focusing on its biochemical structure, biological functions, measurement methods and areas of clinical interest in different fields of medicine.
Collapse
Affiliation(s)
- Fabio Belvederi
- Department of Basic Biotechnological Sciences, Intensive Care and Perioperative Clinics, Catholic University of Sacred Heart, 00168 Rome, Italy
| | - Simone Leggeri
- Department of Basic Biotechnological Sciences, Intensive Care and Perioperative Clinics, Catholic University of Sacred Heart, 00168 Rome, Italy
| | - Andrea Urbani
- Department of Basic Biotechnological Sciences, Intensive Care and Perioperative Clinics, Catholic University of Sacred Heart, 00168 Rome, Italy; Unit of Chemistry, Biochemistry and Molecular Biology, "A. Gemelli" Hospital Foundation IRCCS, Largo A. Gemelli, 8, 00168 Rome, Italy
| | - Silvia Baroni
- Department of Basic Biotechnological Sciences, Intensive Care and Perioperative Clinics, Catholic University of Sacred Heart, 00168 Rome, Italy; Unit of Chemistry, Biochemistry and Molecular Biology, "A. Gemelli" Hospital Foundation IRCCS, Largo A. Gemelli, 8, 00168 Rome, Italy.
| |
Collapse
|
2
|
Murphy J, Healy C, Mongan D, R Susai S, Cannon M, Cotter DR. Developmental stage of childhood trauma exposure and markers of inflammation at age 24. Brain Behav Immun 2025; 126:225-234. [PMID: 39978695 DOI: 10.1016/j.bbi.2025.02.020] [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: 10/23/2024] [Revised: 01/21/2025] [Accepted: 02/17/2025] [Indexed: 02/22/2025] Open
Abstract
BACKGROUND It is largely unknown whether the specific developmental stage at which childhood trauma occurs is related to inflammatory dysregulation in adulthood. We aimed to explore if trauma exposure at distinct developmental stages in childhood is differentially associated with the novel marker of chronic inflammation - soluble urokinase plasminogen activator receptor (suPAR), as well as with C-Reactive Protein (CRP) and Interleukin-6 (IL-6) levels in early adulthood. METHODS Participants were drawn from the Avon Longitudinal Study of Parents and Children (n = 3272). The trauma variables represent any trauma exposure within early (0-4.9 years), middle (5-10.9 years), or late (11-17 years) childhood, and were derived from the responses to 121 questions collected via standardised questionnaires regarding traumatic experiences including physical abuse, sexual abuse, emotional abuse, emotional neglect, domestic violence, and bullying. Plasma suPAR, CRP and IL-6 samples were collected at age 24. Linear regression models assessed the relationship between trauma exposure at different developmental stages and the inflammatory markers, adjusting for sex, socio-economic status (SES) and child ethnicity. Latent profile analysis (LPA) identified age 24 inflammatory profiles and multinomial logistic regressions identified associations between childhood trauma and these latent groups. RESULTS After adjustment for confounders, late childhood trauma was significantly associated with age 24 suPAR (β = 0.06, 95 % CI [.03, 0.1], p = 0.001), CRP (β = 0.09, 95 % CI [.01, 0.17], p = 0.04) and IL-6 (β = 0.1, 95 % CI [.02, 0.19], p = 0.02). The relationship between late trauma and suPAR survived additional adjustment for prior trauma (β = 0.06, 95 % CI [.01, 0.11], p = 0.03). Middle childhood trauma was significantly associated with IL-6 (β = 0.1, 95 % CI [.02, 0.18], p = 0.02). This attenuated after additionally adjusting for prior trauma (β = 0.11, 95 % CI [-0.09, 0.3], p = 0.29). There was little evidence of an association between early trauma and any inflammatory marker. Exposure to any trauma from 0-17 years was associated with elevated suPAR (β = 0.04, 95 % CI [.005, 0.07], p = 0.025) and IL-6 (β = 0.1, 95 % CI [.02, 0.18], p = 0.02) after adjustment for confounders. Additionally, LPA identified three distinct inflammatory profiles: 1. no inflammatory dysregulation; 2. elevated CRP and IL-6 levels; and 3. a high inflammatory group characterised by elevated levels of suPAR, CRP and IL-6. After adjustment for confounders, individuals with trauma either in early (RR = 2.31, 95 % CI [1.16, 4.6], p = 0.017), middle (RR = 2.72, 95 % CI [1.4, 5.29], p = 0.003) or late (RR = 3.37, 95 % CI [1.7, 6.64], p < 0.001) childhood had an increased risk of being in the high inflammatory group. The association between late childhood trauma and this high inflammatory group survived adjustment for prior trauma (RR = 3.69, 95 % CI [1.44, 9.47], p = 0.007). DISCUSSION When the inflammatory markers were analysed independently, late childhood trauma showed a strong association with age 24 suPAR levels after adjusting for confounders and prior trauma. When the inflammatory markers were analysed in combination, those with late childhood trauma also were likely to have an elevated suPAR, CRP and IL-6 inflammatory profile. Collectively, the findings highlight the propensity of late childhood trauma (rather than early or mid-childhood trauma) for the dysregulation of suPAR in early adulthood and support the measurement of suPAR in combination with other markers to better characterise the effects of childhood trauma on adult inflammation. Future studies should use suPAR in combination with CRP and IL-6 to further explore the inflammatory contribution in the relationship between trauma and adverse health outcomes in adulthood.
Collapse
Affiliation(s)
- Jennifer Murphy
- Department of Psychiatry, RCSI University of Medicine and Health Sciences, Dublin, Ireland.
| | - Colm Healy
- Department of Child and Adolescent Psychiatry, School of Medicine, University College Dublin, Ireland; Centre for Clinical Brain Sciences, Division of Psychiatry, University of Edinburgh, Edinburgh, United Kingdom
| | - David Mongan
- Department of Psychiatry, RCSI University of Medicine and Health Sciences, Dublin, Ireland; Centre for Public Health, Queen's University Belfast, United Kingdom; Belfast Health and Social Care Trust, Belfast, United Kingdom
| | - Subash R Susai
- Department of Psychiatry, RCSI University of Medicine and Health Sciences, Dublin, Ireland; FutureNeuro Research Ireland Centre, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Mary Cannon
- Department of Psychiatry, RCSI University of Medicine and Health Sciences, Dublin, Ireland; FutureNeuro Research Ireland Centre, Royal College of Surgeons in Ireland, Dublin, Ireland; Department of Psychiatry, Beaumont Hospital, Dublin 9, Ireland
| | - David R Cotter
- Department of Psychiatry, RCSI University of Medicine and Health Sciences, Dublin, Ireland; FutureNeuro Research Ireland Centre, Royal College of Surgeons in Ireland, Dublin, Ireland; Department of Psychiatry, Beaumont Hospital, Dublin 9, Ireland
| |
Collapse
|
3
|
Sarwar S, Ahmed F, Kadoya Y, Hakimjavadi R, Boczar KE. Assessment of Coronary Microvascular Dysfunction in Patients with Systemic Vasculitis. Curr Cardiol Rep 2025; 27:81. [PMID: 40198429 DOI: 10.1007/s11886-025-02231-w] [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] [Accepted: 03/24/2025] [Indexed: 04/10/2025]
Abstract
INTRODUCTION Coronary microvascular dysfunction (CMD) is characterized by impaired coronary blood flow in the absence of obstructive coronary artery disease. CMD primarily involves the microvasculature, leading to myocardial ischemia, angina, and increased cardiovascular risk. Systemic vasculitides (e.g., giant cell arteritis, antineutrophil cytoplasmic antibody-associated vasculitis, and Takayasu arteritis) are a group of autoimmune conditions known to affect the vasculature through inflammation of the blood vessels that have been associated with more prevalent and severe CMD. Although systemic inflammation likely plays a role in the increased risk of cardiovascular events, the underlying pathogenesis is not well understood. PURPOSE OF REVIEW Invasive and non-invasive techniques for assessing coronary microvascular function have been developed to assess for blood flow and coronary flow reserve (CFR), defined as the ratio of the maximum achievable blood flow during stress to the resting blood flow. The purpose of this review is to further explore the relationship between vasculitis and CMD as well as the techniques available for assessing this association. RECENT FINDINGS Studies have shown that CMD is significantly more prevalent in patients with systemic vasculitis compared to the general population. Moreover, in the absence of significant atherosclerotic burden, patients with vasculitis have a lower CFR than controls, indicating more severely impaired coronary vasomotor function. This suggests that systemic inflammation itself is a factor in driving coronary vasomotor abnormalities and CMD development. CMD contributes to cardiovascular morbidity in patients with systemic vasculitis, underscoring the need for early recognition and management. Further studies are needed to determine whether therapies targeting the reduction of systemic inflammation can lead to improved coronary microvascular function and cardiovascular outcomes.
Collapse
Affiliation(s)
- Shihab Sarwar
- Division of Cardiology, Department of Medicine, University of Ottawa Heart Institute, Ottawa, Canada
| | - Faisal Ahmed
- Division of Cardiology, Department of Medicine, University of Ottawa Heart Institute, Ottawa, Canada
| | - Yoshito Kadoya
- Division of Cardiology, Department of Medicine, University of Ottawa Heart Institute, Ottawa, Canada
| | - Ramtin Hakimjavadi
- Division of Cardiology, Department of Medicine, University of Ottawa Heart Institute, Ottawa, Canada
| | - Kevin Emery Boczar
- Division of Cardiology, Department of Medicine, University of Ottawa Heart Institute, Ottawa, Canada.
- School of Epidemiology and Public Health, Department of Medicine, University of Ottawa, Ottawa, Canada.
| |
Collapse
|
4
|
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
|
5
|
Bahrami HSZ, Jørgensen PG, Hove JD, Dixen U, Rasmussen LJH, Eugen-Olsen J, Rossing P, Jensen MT. Association between interleukin-6, suPAR, and hsCRP with subclinical left ventricular dysfunction in type 1 diabetes: The Thousand & 1 study. Diabetes Res Clin Pract 2025; 222:112071. [PMID: 40043809 DOI: 10.1016/j.diabres.2025.112071] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/06/2025] [Revised: 02/05/2025] [Accepted: 02/24/2025] [Indexed: 03/18/2025]
Abstract
AIMS To investigate the association between chronic inflammation and subclinical left ventricular dysfunction in type 1 diabetes (T1D). METHODS In a cross-sectional study of individuals with T1D without known heart disease, interleukin-6 (IL-6), soluble-urokinase-plasminogen-activator-receptor (suPAR), and high-sensitivity C-reactive-protein (hsCRP) were examined for associations with echocardiographic E/e' (primary outcome) and global longitudinal strain (GLS) (secondary outcome). We adjusted for several clinical variables in linear regression analysis, including N-terminal pro-B-type natriuretic peptide (NT-proBNP). The biomarkers were categorized as elevated/non-elevated based on their upper quartiles. RESULTS Of 962 individuals (52 % male, mean age 49 ± 14 years), mean E/e' was 7 ± 3 and GLS 18 ± 3. In fully adjusted models, all biomarkers were each associated with increased E/e': beta coefficients for IL-6 0.2 (95 % confidence intervals: 0.1-0.3, P = 0.001), suPAR 0.5 (0.1-0.7, P = 0.011), and hsCRP 0.1 (0.0-0.2, P = 0.023). Combining biomarkers showed stronger associations: elevated IL-6 and suPAR 1.3 (0.7-2.0, P < 0.001), elevated all three 1.9 (1.1-2.7, P < 0.001). Results were similar for decreased GLS with IL-6-0.4 (-0.7 to 0.0, P = 0.039), IL-6 and hsCRP -1.0 (-1.7 to -0.4, P = 0.007), all three -1.1 (-2.0 to -0.3, P = 0.009). CONCLUSIONS Inflammatory biomarkers are independently associated with subclinical left ventricular dysfunction. Chronic inflammation may contribute to the development of myocardial dysfunction in T1D.
Collapse
Affiliation(s)
- Hashmat Sayed Zohori Bahrami
- Department of Clinical and Translational Research, Steno Diabetes Center Copenhagen, Borgmester Ib Juuls Vej 83, 2730 Herlev, Denmark; Department of Cardiology, Copenhagen University Hospital, Amager & Hvidovre, Kettegård Alle 30, 2650 Hvidovre, Denmark; Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3b, 2200 Copenhagen, Denmark.
| | - Peter Godsk Jørgensen
- Department of Cardiology, Copenhagen University Hospital, Herlev & Gentofte, Borgmester Ib Juuls Vej 1, 2730 Herlev, Denmark
| | - Jens Dahlgaard Hove
- Department of Cardiology, Copenhagen University Hospital, Amager & Hvidovre, Kettegård Alle 30, 2650 Hvidovre, Denmark; Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3b, 2200 Copenhagen, Denmark
| | - Ulrik Dixen
- Department of Cardiology, Copenhagen University Hospital, Amager & Hvidovre, Kettegård Alle 30, 2650 Hvidovre, Denmark; Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3b, 2200 Copenhagen, Denmark
| | - Line Jee Hartmann Rasmussen
- Department of Clinical Research, Copenhagen University Hospital, Amager & Hvidovre, Kettegård Alle 30, 2650 Hvidovre, Denmark; Department of Psychology & Neuroscience, Duke University, 2020 W Main St, Durham, NC 27708, USA
| | - Jesper Eugen-Olsen
- Department of Cardiology, Copenhagen University Hospital, Herlev & Gentofte, Borgmester Ib Juuls Vej 1, 2730 Herlev, Denmark; Department of Clinical Research, Copenhagen University Hospital, Amager & Hvidovre, Kettegård Alle 30, 2650 Hvidovre, Denmark
| | - Peter Rossing
- Department of Clinical and Translational Research, Steno Diabetes Center Copenhagen, Borgmester Ib Juuls Vej 83, 2730 Herlev, Denmark; Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3b, 2200 Copenhagen, Denmark
| | - Magnus T Jensen
- Department of Clinical and Translational Research, Steno Diabetes Center Copenhagen, Borgmester Ib Juuls Vej 83, 2730 Herlev, Denmark; Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3b, 2200 Copenhagen, Denmark; William Harvey Research Institute, NIHR Barts Biomedical Centre, Queen Mary University London, Charterhouse Square, London EC1M 6BQ, UK
| |
Collapse
|
6
|
Wisborg FD, El Caidi NO, Taraldsen IA, Tonning S, Kandiah A, El‐Sheikh M, Bahrami HSZ, Andersen O, Rasmussen LJH, Hove J, Dixen U, Grand J. Soluble urokinase plasminogen activator receptor (suPAR) as a prognostic biomarker in acutely admitted patients with atrial fibrillation. J Arrhythm 2025; 41:e70077. [PMID: 40271386 PMCID: PMC12017082 DOI: 10.1002/joa3.70077] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2024] [Revised: 03/12/2025] [Accepted: 04/04/2025] [Indexed: 04/25/2025] Open
Abstract
Background Atrial fibrillation (AF) is associated with a higher incidence of stroke, heart failure, and mortality. Risk assessment of clinical outcomes in patients hospitalized acutely with AF remains a challenge. Purpose To investigate if soluble urokinase plasminogen activator receptor (suPAR) levels at admission to the Emergency Department (ED) are associated with 1-year all-cause mortality in patients admitted with AF. Methods A prospective cohort study of patients consecutively admitted to the medical ED of a university hospital in Copenhagen, Denmark, between 2020 and 2022 with symptoms of COVID-19. Patients were included if they were admitted with AF as the primary or secondary diagnosis. All patients had suPAR measured at the index admission, and follow-up was up to 1 year. The association between suPAR and 1-year mortality was investigated with multivariate Cox regression. We adjusted for age, sex, smoking, C-reactive protein, creatinine, hemoglobin, albumin, and comorbidities. Results Of the 7,258 patients included during the period, 362 (5.0%) patients were admitted with AF as the primary or secondary diagnosis. Due to missing data, 23 (6.4%) patients were excluded. Among the remaining 339 patients, 68 (20.1%) patients were dead at follow-up. The multivariate Cox regression showed that elevated suPAR was independently associated with an increased risk of 1-year mortality, with a hazard ratio of 1.12 (95% confidence interval: 1.05-1.20, p < 0.001). Conclusion Elevated suPAR levels were significantly associated with 1-year all-cause mortality in patients acutely admitted with AF to the ED.
Collapse
Affiliation(s)
| | - Nora Olsen El Caidi
- Department of CardiologyCopenhagen University Hospital, Amager and HvidovreHvidovreDenmark
| | - Ida Arentz Taraldsen
- Department of CardiologyCopenhagen University Hospital, Amager and HvidovreHvidovreDenmark
| | - Sandra Tonning
- Department of CardiologyCopenhagen University Hospital, Amager and HvidovreHvidovreDenmark
| | - Aginsha Kandiah
- Department of CardiologyCopenhagen University Hospital, Amager and HvidovreHvidovreDenmark
| | - Mohammed El‐Sheikh
- Department of CardiologyCopenhagen University Hospital, Amager and HvidovreHvidovreDenmark
| | - Hashmat S. Z. Bahrami
- Department of CardiologyCopenhagen University Hospital, Amager and HvidovreHvidovreDenmark
- Department of Clinical and Translational ResearchSteno Diabetes Center CopenhagenHerlevDenmark
| | - Ove Andersen
- Department of Clinical ResearchCopenhagen University Hospital, Amager and HvidovreHvidovreDenmark
- Department of Emergency MedicineCopenhagen University Hospital, Amager and HvidovreHvidovreDenmark
- Faculty of Health and Medical SciencesUniversity of CopenhagenCopenhagenDenmark
| | - Line Jee Hartmann Rasmussen
- Department of Clinical ResearchCopenhagen University Hospital, Amager and HvidovreHvidovreDenmark
- Department of Emergency MedicineCopenhagen University Hospital, Amager and HvidovreHvidovreDenmark
- Faculty of Health and Medical SciencesUniversity of CopenhagenCopenhagenDenmark
| | - Jens Hove
- Department of CardiologyCopenhagen University Hospital, Amager and HvidovreHvidovreDenmark
| | - Ulrik Dixen
- Department of CardiologyCopenhagen University Hospital, Amager and HvidovreHvidovreDenmark
| | - Johannes Grand
- Department of CardiologyCopenhagen University Hospital, Amager and HvidovreHvidovreDenmark
| |
Collapse
|
7
|
Tan B, Tang W, Zeng Y, Liu J, Du X, Su H, Pang X, Liao L, Hu Q. Development of animal models with chronic kidney disease-mineral and bone disorder based on clinical characteristics and pathogenesis. Front Endocrinol (Lausanne) 2025; 16:1549562. [PMID: 40201764 PMCID: PMC11975589 DOI: 10.3389/fendo.2025.1549562] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/21/2024] [Accepted: 03/10/2025] [Indexed: 04/10/2025] Open
Abstract
Chronic kidney disease-mineral and bone disorder (CKD-MBD) is a systemic complication of chronic kidney disease (CKD), resulting in high morbidity and mortality. However, effective treatment strategies are lacking. The pathogenesis of CKD-MBD is unclear but involves feedback mechanisms between calcium, phosphorus, parathyroid hormone, vitamin D and other factors, in addition to FGF23, Klotho, Wnt inhibitors, and activin A. Construction of a perfect animal model of CKD-MBD with clinical characteristics is important for in-depth study of disease development, pathological changes, targeted drug screening, and management of patients. Currently, the modeling methods of CKD-MBD include surgery, feeding and radiation. Additionally, the method of CKD-MBD modeling by surgical combined feeding is worth promoting because of short time, simplicity, and low mortality. Therefore, this review based on the pathogenesis and clinical features of CKD-MBD, combined with the current status of animal models, outlines the advantages and disadvantages of modeling methods, and provides a reference for further CKD-MBD research.
Collapse
Affiliation(s)
- Biyu Tan
- Department of Nephrology, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Sichuan, China
| | - Weili Tang
- Department of Orthopedics, The Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Sichuan, China
| | - Yan Zeng
- Department of Nephrology, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Sichuan, China
| | - Jian Liu
- Department of Nephrology, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Sichuan, China
| | - Xiaomei Du
- Department of Nephrology, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Sichuan, China
| | - Hongwei Su
- Department of Urology, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Sichuan, China
| | - Xianlun Pang
- Department of Orthopedics, The Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Sichuan, China
| | - Lishang Liao
- Department of Neurosurgery, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Sichuan, China
| | - Qiongdan Hu
- Department of Nephrology, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Sichuan, China
| |
Collapse
|
8
|
Mascolo R, Wu MA, Berra S, Vidali M, Pancrazi M, Trotta L, Ceriani E, Negro E, Serati L, Carrozzo L, Calabrò E, Ceriotti F, Andreis A, Collini V, Imazio M, Brucato A. Recurrent pericarditis with normal C-reactive protein: Clinical and laboratory features, biomarkers and outcome in a non-inflammatory phenotype. Eur J Intern Med 2025:S0953-6205(25)00097-4. [PMID: 40113467 DOI: 10.1016/j.ejim.2025.03.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/30/2024] [Revised: 02/23/2025] [Accepted: 03/12/2025] [Indexed: 03/22/2025]
Abstract
BACKGROUND Differences in recurrent pericarditis with normal vs. elevated C-reactive protein (CRP) are unknown. MATERIALS AND METHODS We studied 448 patients with recurrent or incessant pericarditis. CRP levels <10 mg/L were considered normal. Forty-one patients with normal CRP were tested for interleukin 6 (IL-6), serum amyloid A (SAA) and soluble urokinase plasminogen activator receptor (suPAR). RESULTS Among the cohort (median age 43 years, 48.4% females), 336 patients (75%) had elevated CRP, while 112 (25%) had normal levels. CRP-negative patients were younger (38.6 vs. 43.5 years, p=0.007) and predominantly female (67.9% vs. 42.0%, p<0.001). They presented less often with fever (23.2% vs. 74.1%, p<0.001), pleural involvement (7.1% vs. 61.6% p<0.001), and neutrophilic leucocytosis (WBC 6760 vs. 12315/µL, p<0.001). Pericardiocentesis was performed rarely in CRP-negative patients (2.7% vs. 13.4%, p=0.001). Recurrence rates were similar (5.4 vs. 5.5/100 months-patient, p=0.918). Among CRP-negative patients, 53 (47.3%) had an incessant course characterized by persistent symptoms (pain and tachycardia); abnormal instrumental findings included ECG changes (28.6%), mild pericardial effusion (86.6%), and cardiac magnetic resonance evidence of effusion/oedema/late gadolinium enhancement (43.7%). Anakinra was administered to 48 CRP-positive (14.3%) and 10 CRP-negative patients (8.9%), leading to good responses with discontinuation of NSAIDs and corticosteroids in 39/48 (81.2%) and 5/10 (50.0%), respectively. Among the 41 CRP-negative patients tested, IL-6 and suPAR levels were always normal, while SAA was elevated in 17.1%. CONCLUSIONS Pericarditis with normal CRP exhibits distinct clinical and laboratory features, often presenting with an incessant course. Although rarely elevated, SAA may help to identify inflammation beyond CRP.
Collapse
Affiliation(s)
- Ruggiero Mascolo
- Division of Internal Medicine, ASST Fatebenefratelli Sacco, Fatebenefratelli Hospital, University of Milan, Milan, Italy.
| | - Maddalena Alessandra Wu
- Division of Internal Medicine, ASST Fatebenefratelli Sacco, Luigi Sacco Hospital, University of Milan, Milan, Italy; Department of Biomedical and Clinical Sciences, University of Milan, Milan, Italy
| | - Silvia Berra
- Division of Internal Medicine, ASST Fatebenefratelli Sacco, Fatebenefratelli Hospital, University of Milan, Milan, Italy
| | - Matteo Vidali
- Clinical Pathology Unit, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milano, Italy
| | - Massimo Pancrazi
- Division of Internal Medicine, ASST Fatebenefratelli Sacco, Fatebenefratelli Hospital, University of Milan, Milan, Italy
| | - Lucia Trotta
- Division of Internal Medicine, ASST Fatebenefratelli Sacco, Fatebenefratelli Hospital, University of Milan, Milan, Italy
| | - Elisa Ceriani
- Division of Internal Medicine, ASST Fatebenefratelli Sacco, Luigi Sacco Hospital, University of Milan, Milan, Italy; Department of Biomedical and Clinical Sciences, University of Milan, Milan, Italy
| | - Enrica Negro
- Division of Internal Medicine, ASST Fatebenefratelli Sacco, Fatebenefratelli Hospital, University of Milan, Milan, Italy
| | - Lisa Serati
- Division of Internal Medicine, ASST Fatebenefratelli Sacco, Fatebenefratelli Hospital, University of Milan, Milan, Italy
| | - Luisa Carrozzo
- Division of Internal Medicine, ASST Fatebenefratelli Sacco, Fatebenefratelli Hospital, University of Milan, Milan, Italy
| | - Elisa Calabrò
- Division of Internal Medicine, ASST Fatebenefratelli Sacco, Fatebenefratelli Hospital, University of Milan, Milan, Italy
| | - Ferruccio Ceriotti
- Clinical Pathology Unit, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milano, Italy
| | - Alessandro Andreis
- Department of Cardiology, University Hospital Città della Salute e della Scienza, Torino, Italy
| | - Valentino Collini
- Cardiothoracic Department, University Hospital Santa Maria della Misericordia, ASUFC, Udine, Italy
| | - Massimo Imazio
- Cardiothoracic Department, University Hospital Santa Maria della Misericordia, ASUFC, Udine, Italy
| | - Antonio Brucato
- Division of Internal Medicine, ASST Fatebenefratelli Sacco, Fatebenefratelli Hospital, University of Milan, Milan, Italy; Department of Biomedical and Clinical Sciences, University of Milan, Milan, Italy
| |
Collapse
|
9
|
Nègre P, Tayac D, Jamme T, Combis MS, Maupas-Schwalm F. Early suPAR levels as a predictor of COVID-19 severity: A new tool for efficient patient triage. Infect Dis Now 2025; 55:105058. [PMID: 40101896 DOI: 10.1016/j.idnow.2025.105058] [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: 11/07/2024] [Revised: 02/18/2025] [Accepted: 03/14/2025] [Indexed: 03/20/2025]
Abstract
BACKGROUND Following several waves of the COVID-19 pandemic, we are now facing a lower but persistent rate of SARS-CoV-2 infections, with seasonal resurgences often coinciding with other respiratory tract infections. OBJECTIVE We aimed to identify early clinico-biological variables predictive of an unfavorable outcome in patients with primary SARS-CoV-2 infection. We also evaluated the role of suPAR, an innovative biomarker, in predicting disease severity. METHODS We included 255 patients with PCR-confirmed primary SARS-CoV-2 infection and with a 30-day follow-up minimum. Blood samples were collected within the first 24 h of hospitalization to measure suPAR levels. Comprehensive data from medical records were analyzed to assess their predictive value in stratifying patients into seven severity groups, with groups 1 to 3 representing severe COVID-19 (death, intubation, ECMO, or non-invasive ventilation). RESULTS Early plasma suPAR levels were significantly associated with severe disease progression, as evidenced by ANOVA and logistic regression models, highlighting suPAR as a persistent predictive factor for unfavorable outcomes. CONCLUSION Our findings suggest that a single suPAR measurement, performed early after a positive PCR test for SARS-CoV-2, holds strong predictive value for patient outcomes. This biomarker, alongside pulse oximetry and CT scan results, could be instrumental in early patient triage during seasonal COVID-19 resurgences.
Collapse
Affiliation(s)
- Pauline Nègre
- Faculty of Pharmacy, Toulouse III University, France; Medical Biochemistry Laboratory, CHU Toulouse, France
| | - Didier Tayac
- Medical Biochemistry Laboratory, CHU Toulouse, France
| | - Thibaut Jamme
- Medical Biochemistry Laboratory, CHU Toulouse, France
| | | | - Françoise Maupas-Schwalm
- Medical Biochemistry Laboratory, CHU Toulouse, France; Faculty of Medicine, Toulouse III University, France.
| |
Collapse
|
10
|
Houlind M, Nielsen A, Walls A, Christensen L, Nielsen R, Andersen A, Jawad B, Andersen O, Damgaard M, Iversen E, Tavenier J, Juul‐Larsen H. Plasma NGAL, suPAR, KIM-1 and GDF-15 for Improving Glomerular Filtration Rate Estimation in Older Hospitalized Patients. Basic Clin Pharmacol Toxicol 2025; 136:e70002. [PMID: 39865371 PMCID: PMC11771596 DOI: 10.1111/bcpt.70002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2024] [Revised: 01/13/2025] [Accepted: 01/15/2025] [Indexed: 01/28/2025]
Affiliation(s)
- Morten Baltzer Houlind
- Department of Clinical ResearchCopenhagen University Hospital, Amager and HvidovreHvidovreDenmark
- The Capital Region PharmacyHerlevDenmark
- Department of Drug Design and PharmacologyUniversity of CopenhagenCopenhagenDenmark
- Department of NephrologyCopenhagen University Hospital, RigshospitaletCopenhagenDenmark
| | - Alberte Linnet Nielsen
- Department of Clinical ResearchCopenhagen University Hospital, Amager and HvidovreHvidovreDenmark
- Department of Drug Design and PharmacologyUniversity of CopenhagenCopenhagenDenmark
| | - Anne Byriel Walls
- The Capital Region PharmacyHerlevDenmark
- Department of Drug Design and PharmacologyUniversity of CopenhagenCopenhagenDenmark
| | - Louise Westberg Strejby Christensen
- Department of Clinical ResearchCopenhagen University Hospital, Amager and HvidovreHvidovreDenmark
- The Capital Region PharmacyHerlevDenmark
- Department of Clinical MedicineUniversity of CopenhagenCopenhagenDenmark
| | - Rikke Lundsgaard Nielsen
- Department of Clinical ResearchCopenhagen University Hospital, Amager and HvidovreHvidovreDenmark
- Department of Clinical MedicineUniversity of CopenhagenCopenhagenDenmark
| | - Aino Andersen
- Department of Clinical ResearchCopenhagen University Hospital, Amager and HvidovreHvidovreDenmark
| | - Baker Nawfal Jawad
- Department of Clinical ResearchCopenhagen University Hospital, Amager and HvidovreHvidovreDenmark
| | - Ove Andersen
- Department of Clinical ResearchCopenhagen University Hospital, Amager and HvidovreHvidovreDenmark
- Department of Clinical MedicineUniversity of CopenhagenCopenhagenDenmark
- Emergency DepartmentCopenhagen University Hospital, Amager and HvidovreHvidovreDenmark
| | - Morten Damgaard
- Department of Clinical Physiology and Nuclear Medicine, Centre for Functional and Diagnostic Imaging and ResearchCopenhagen University Hospital, Amager & HvidovreHvidovreDenmark
| | - Esben Iversen
- Department of Clinical ResearchCopenhagen University Hospital, Amager and HvidovreHvidovreDenmark
| | - Juliette Tavenier
- Department of Clinical ResearchCopenhagen University Hospital, Amager and HvidovreHvidovreDenmark
| | - Helle Gybel Juul‐Larsen
- Department of Clinical ResearchCopenhagen University Hospital, Amager and HvidovreHvidovreDenmark
| |
Collapse
|
11
|
Popa AD, Gherasim A, Mihalache L, Arhire LI, Graur M, Niță O. Fasting Mimicking Diet for Metabolic Syndrome: A Narrative Review of Human Studies. Metabolites 2025; 15:150. [PMID: 40137116 PMCID: PMC11943686 DOI: 10.3390/metabo15030150] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2025] [Revised: 02/19/2025] [Accepted: 02/21/2025] [Indexed: 03/27/2025] Open
Abstract
Metabolic syndrome (MetS) is an association of risk factors that share insulin resistance (IR), exerting a super cumulative effect on the risk of developing cardiometabolic diseases. Lifestyle optimization is a key element in the prevention and non-pharmacological therapy of MetS. Certain studies have concluded that some dietary patterns could be more beneficial as an adjunctive treatment for MetS. Fasting mimicking diet (FMD) is a form of periodic fasting in which caloric intake is restricted for 5 days each month. It has been studied for its beneficial effects not only in patients with neoplasia and neurodegenerative diseases but also for its effects on IR and metabolism. In this narrative review, the effects of FMD in patients with MetS were analyzed, focusing on its impact on key metabolic components and summarizing findings from human studies. FMD has demonstrated beneficial effects on MetS by reducing BMI and waist circumference, preserving lean mass, and improving the metabolic profile. Moreover, individuals with a higher BMI or a greater number of MetS components appear to derive greater benefits from this intervention. However, limitations such as high dropout rates, small sample sizes, and methodological constraints restrict the generalizability of current findings. Further large-scale studies are needed to confirm these effects and establish FMD as a viable non-pharmacological strategy for managing MetS.
Collapse
Affiliation(s)
- Alina Delia Popa
- Internal Medicine II Department, Faculty of Medicine, University of Medicine and Pharmacy “Grigore T Popa”, 700115 Iasi, Romania; (A.D.P.); (L.M.); (L.I.A.); (O.N.)
| | - Andreea Gherasim
- Internal Medicine II Department, Faculty of Medicine, University of Medicine and Pharmacy “Grigore T Popa”, 700115 Iasi, Romania; (A.D.P.); (L.M.); (L.I.A.); (O.N.)
| | - Laura Mihalache
- Internal Medicine II Department, Faculty of Medicine, University of Medicine and Pharmacy “Grigore T Popa”, 700115 Iasi, Romania; (A.D.P.); (L.M.); (L.I.A.); (O.N.)
| | - Lidia Iuliana Arhire
- Internal Medicine II Department, Faculty of Medicine, University of Medicine and Pharmacy “Grigore T Popa”, 700115 Iasi, Romania; (A.D.P.); (L.M.); (L.I.A.); (O.N.)
| | - Mariana Graur
- Faculty of Medicine and Biological Sciences, University “Ștefan cel Mare” of Suceava, 720229 Suceava, Romania;
| | - Otilia Niță
- Internal Medicine II Department, Faculty of Medicine, University of Medicine and Pharmacy “Grigore T Popa”, 700115 Iasi, Romania; (A.D.P.); (L.M.); (L.I.A.); (O.N.)
| |
Collapse
|
12
|
Tesfay B, Ashina H, Christensen RH, Al-Khazali HM, Karlsson WK, Amin FM, Jawad BN, Andersen O, Ashina M. Association of plasma soluble urokinase plasminogen activator receptor concentrations and migraine with aura: a REFORM study. Brain Commun 2025; 7:fcae475. [PMID: 39963289 PMCID: PMC11831075 DOI: 10.1093/braincomms/fcae475] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2024] [Revised: 10/22/2024] [Accepted: 02/12/2025] [Indexed: 02/20/2025] Open
Abstract
Soluble urokinase plasminogen activator receptor (suPAR) has garnered attention as a potential blood-based biomarker for low-grade chronic inflammation. However, its specific association with migraine, including its subtypes, remains to be elucidated. We sought to examine the association of plasma suPAR levels with migraine and its subtypes. In this single-centre, cross-sectional study, plasma was collected at a single time point in adults with migraine and sex-matched healthy controls from October 2020 to June 2022. The quantification of plasma suPAR levels was performed in a blinded fashion using a validated enzyme-linked immunosorbent assay. Plasma suPAR levels were compared between participants with migraine (including subgroups) and healthy controls. Plasma samples were analysed from 634 eligible participants with migraine [mean (SD) age, 44.0 (12.2) years; 568 (89.6%) females] and 154 healthy controls [mean (SD), 41.3 (11.8%) years; 132 (86%) females]. Plasma suPAR levels were 6.7% higher (95% CI: 0.1-13.6%; P = 0.045, adjusted for age, sex, body mass index and smoking) in participants with migraine with aura, when compared with healthy controls. Further analysis revealed no difference in plasma suPAR levels between the overall migraine group and healthy controls (3.7%; 95% CI: -0.7-8.2%; P = 0.097), as well as between participants with migraine without aura and healthy controls (2.5%; 95% CI: -2.9-8.3%; P = 0.81). Similarly, plasma suPAR levels did not differ across participants with episodic migraine, chronic migraine and healthy controls. Finally, we found no difference when comparing participants with migraine at time of blood sampling with participants with non-migraine headache (1.0%; 95% CI: -5.7-8.2; P > 0.99), participants without headache (1.2%; 95% CI: -4.2-7.0%; P > 0.99) or healthy controls (4.5%; 95% CI: -1.9-11.3%; P = 0.39). Elevated plasma suPAR levels in migraine with aura indicate the presence of low-grade chronic inflammation. Future research should explore the role of suPAR in the neurobiologic underpinnings of migraine with aura.
Collapse
Affiliation(s)
- Betel Tesfay
- Department of Neurology, Danish Headache Center, Copenhagen University Hospital—Rigshospitalet, Copenhagen 2600, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen 2200, Denmark
| | - Håkan Ashina
- Department of Neurology, Danish Headache Center, Copenhagen University Hospital—Rigshospitalet, Copenhagen 2600, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen 2200, Denmark
- Translational Research Center, Copenhagen University Hospital—Rigshospitalet, Copenhagen 2600, Denmark
| | - Rune Häckert Christensen
- Department of Neurology, Danish Headache Center, Copenhagen University Hospital—Rigshospitalet, Copenhagen 2600, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen 2200, Denmark
- Translational Research Center, Copenhagen University Hospital—Rigshospitalet, Copenhagen 2600, Denmark
| | - Haidar M Al-Khazali
- Department of Neurology, Danish Headache Center, Copenhagen University Hospital—Rigshospitalet, Copenhagen 2600, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen 2200, Denmark
- Translational Research Center, Copenhagen University Hospital—Rigshospitalet, Copenhagen 2600, Denmark
| | - William Kristian Karlsson
- Department of Neurology, Danish Headache Center, Copenhagen University Hospital—Rigshospitalet, Copenhagen 2600, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen 2200, Denmark
| | - Faisal Mohammad Amin
- Department of Neurology, Danish Headache Center, Copenhagen University Hospital—Rigshospitalet, Copenhagen 2600, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen 2200, Denmark
| | - Baker Nawfal Jawad
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen 2200, Denmark
- Department of Clinical Research, Copenhagen University Hospital—Amager and Hvidovre, Hvidovre 2650, Denmark
| | - Ove Andersen
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen 2200, Denmark
- Department of Clinical Research, Copenhagen University Hospital—Amager and Hvidovre, Hvidovre 2650, Denmark
- Department of Emergency Medicine, Copenhagen University Hospital—Amager and Hvidovre, Hvidovre 2650, Denmark
| | - Messoud Ashina
- Department of Neurology, Danish Headache Center, Copenhagen University Hospital—Rigshospitalet, Copenhagen 2600, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen 2200, Denmark
- Danish Knowledge Center on Headache Disorders, Copenhagen University Hospital—Rigshospitalet, Copenhagen 2600, Denmark
| |
Collapse
|
13
|
May CJ, Ford NP, Welsh GI, Saleem MA. Biomarkers to predict or measure steroid resistance in idiopathic nephrotic syndrome: A systematic review. PLoS One 2025; 20:e0312232. [PMID: 39946431 PMCID: PMC11824968 DOI: 10.1371/journal.pone.0312232] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2023] [Accepted: 10/02/2024] [Indexed: 02/16/2025] Open
Abstract
In this systematic review we have sought to summarise the current knowledge concerning biomarkers that can distinguish between steroid-resistant nephrotic syndrome and steroid-sensitive nephrotic syndrome. Additionally, we aim to select biomarkers that have the best evidence-base and should be prioritised for further research. Pub med and web of science databases were searched using "steroid resistant nephrotic syndrome AND biomarker". Papers published between 01/01/2012 and 10/05/2022 were included. Papers that did not compare steroid resistant and steroid sensitive nephrotic syndrome, did not report sensitivity/specificity or area under curve and reviews/letters were excluded. The selected papers were then assessed for bias using the QUADAS-2 tool. The source of the biomarker, cut off, sensitivity/specificity, area under curve and sample size were all extracted. Quality assessment was performed using the BIOCROSS tool. 17 studies were included, comprising 15 case-control studies and 2 cross-sectional studies. Given the rarity of nephrotic syndrome and difficulty in recruiting large cohorts, case-control studies were accepted despite their limitations. We present a range of candidate biomarkers along with scores relating to the quality of the original publications and the risk of bias to inform future investigations. None of the selected papers stated whether the authors were blinded to the patient's disease when assessing the index test in the cohort. Highlighting a key problem in the field that needs to be addressed. These candidate biomarkers must now be tested with much larger sample sizes. Using new biobanks such as the one built by the NURTuRE-INS team will be very helpful in this regard.
Collapse
Affiliation(s)
- Carl J. May
- Bristol Renal, University of Bristol, Bristol, United Kingdom
| | | | - Gavin I. Welsh
- Bristol Renal, University of Bristol, Bristol, United Kingdom
| | - Moin A. Saleem
- Bristol Renal, University of Bristol, Bristol, United Kingdom
- Bristol Royal Hospital for Children, Bristol, United Kingdom
| |
Collapse
|
14
|
Galliera E, Massaccesi L, Mangiavini L, De Vecchi E, Villa F, Corsi Romanelli MM, Peretti GM. The Evaluation of New-Generation Biomarker sCD14ST Provides New Insight into COVID-19's Effect on Bone Remodeling. J Clin Med 2025; 14:979. [PMID: 39941649 PMCID: PMC11818815 DOI: 10.3390/jcm14030979] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2024] [Revised: 01/24/2025] [Accepted: 01/29/2025] [Indexed: 02/16/2025] Open
Abstract
Background/Objectives: The COVID-19 pandemic has increased interest in osteoimmunology because of the impact of SARS-CoV-2 on both the immune system and the bone microenvironment. Soluble CD14ST could influence the production of the osteoimmunological regulators of osteoclast differentiation. The aim of this study is to evaluate the role of sCD14ST in COVID-19's effects on bone remodeling-evaluating, in particular, the correlation with new-generation osteoimmunological biomarkers-and to acquire comprehensive knowledge of the effects of the disease on the immune and skeletal system. Methods: The serum level of sCD14ST was measured in COVID-19-positive and COVID-19-negative patients undergoing orthopedic surgery and correlated with the inflammatory and osteoimmunological biomarkers RANKL/OPG, FGF23, IL-6, C-reactive protein (CRP), procalcitonin (PCT), sRAGE, and SuPAR. Results: In our patients, sCD14ST showed a strong increase in COVID-19-positive patients, and a significant decrease in tandem with the infection resolution, confirming its diagnostic and prognostic value. sCD14ST was more clinically relevant than the two canonically inflammatory makers used in the clinical protocols, CRP and PCT, and displayed a good positive correlation with FGF23, RANKL/OPG, IL-6, and SuPAR and a negative correlation with sRAGE. Conclusions: Monitoring sCD14ST along with SuPAR may offer valuable insights into immune system dysregulation and bone-related complications in conditions characterized by inflammation. These soluble receptors represent important links between immune activation and bone metabolism, especially in the context of diseases like COVID-19, where the inflammatory response may impact bone fragility.
Collapse
Affiliation(s)
- Emanuela Galliera
- IRCCS Ospedale Galeazzi-Sant’Ambrogio, 20157 Milan, Italy; (L.M.); (L.M.); (E.D.V.); (F.V.); (G.M.P.)
- Department of Biomedical Sciences for Health, Università degli Studi di Milano, 20133 Milan, Italy;
| | - Luca Massaccesi
- IRCCS Ospedale Galeazzi-Sant’Ambrogio, 20157 Milan, Italy; (L.M.); (L.M.); (E.D.V.); (F.V.); (G.M.P.)
- Department of Biomedical Sciences for Health, Università degli Studi di Milano, 20133 Milan, Italy;
| | - Laura Mangiavini
- IRCCS Ospedale Galeazzi-Sant’Ambrogio, 20157 Milan, Italy; (L.M.); (L.M.); (E.D.V.); (F.V.); (G.M.P.)
- Department of Biomedical Sciences for Health, Università degli Studi di Milano, 20133 Milan, Italy;
| | - Elena De Vecchi
- IRCCS Ospedale Galeazzi-Sant’Ambrogio, 20157 Milan, Italy; (L.M.); (L.M.); (E.D.V.); (F.V.); (G.M.P.)
| | - Francesca Villa
- IRCCS Ospedale Galeazzi-Sant’Ambrogio, 20157 Milan, Italy; (L.M.); (L.M.); (E.D.V.); (F.V.); (G.M.P.)
| | - Massimiliano Marco Corsi Romanelli
- Department of Biomedical Sciences for Health, Università degli Studi di Milano, 20133 Milan, Italy;
- Department of Experimental and Clinical Pathology, IRCCS Istituto Auxologico Italiano, 20095 Cusano Milanino, Italy
| | - Giuseppe Maria Peretti
- IRCCS Ospedale Galeazzi-Sant’Ambrogio, 20157 Milan, Italy; (L.M.); (L.M.); (E.D.V.); (F.V.); (G.M.P.)
- Department of Biomedical Sciences for Health, Università degli Studi di Milano, 20133 Milan, Italy;
| |
Collapse
|
15
|
Sotelo-Ramírez CE, Valdés-Tovar M, Zaragoza-Hoyos JU, Ortiz-López L, Argueta J, Rosel-Vales M, Miranda-Labra RU, Camarena B. Molecular and Functional Analysis of TLR 1, 2 and 6 in Peripheral Blood Monocytes of Patients with Schizophrenia: A Pilot Study. Int J Mol Sci 2025; 26:926. [PMID: 39940697 PMCID: PMC11817014 DOI: 10.3390/ijms26030926] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2024] [Revised: 12/05/2024] [Accepted: 12/13/2024] [Indexed: 02/16/2025] Open
Abstract
Schizophrenia (SZ) is a chronic disabling mental disorder with high heritability, and several immune-regulating genes have been implicated in its pathophysiology In this study, we investigated the expression of Toll-like receptors (TLRs) 1, 2, and 6 in peripheral blood monocytes from SZ patients and healthy control subjects (HCSs) in the Mexican population, focusing on specific SZ-associated gene variants. Gene expressions were assessed by qPCR, and protein expression was measured using flow cytometry. The secretory profiles of MALP2-stimulated monocytes were evaluated through immunoproteomic arrays. Our results indicate that patients with SZ carrying the rs4833093/TLR1 GG genotype exhibited significantly lower TLR1 gene expression compared to TT carriers. Notably, HCSs with the TT genotype showed markedly higher TLR1 protein expression, while all patients with SZ exhibited significantly reduced protein levels regardless of genotype. Furthermore, monocytes from patients with SZ displayed altered secretion profiles upon TLR stimulation, with significant elevations in IL-18, uPAR, angiopoietin-2, and serpin E1, alongside reductions in MCP-1, IL-17A, IL-24, MIF, and myeloperoxidase compared to HCSs. These findings suggest a dysfunctional TLR-mediated innate immune response in SZ.
Collapse
Affiliation(s)
- Carlo E. Sotelo-Ramírez
- Doctorado en Biología Experimental, Universidad Autónoma Metropolitana (UAM)-Iztapalapa, Mexico City 09340, Mexico;
- Departamento de Farmacogenética, Subdirección de Investigaciones Clínicas, Instituto Nacional de Psiquiatría Ramón de la Fuente Muñiz, Mexico City 14370, Mexico;
| | - Marcela Valdés-Tovar
- Subdirección de Investigaciones Clínicas, Instituto Nacional de Psiquiatría Ramón de la Fuente Muñiz, Mexico City 14370, Mexico; (M.V.-T.); (L.O.-L.)
| | - Julio Uriel Zaragoza-Hoyos
- Departamento de Farmacogenética, Subdirección de Investigaciones Clínicas, Instituto Nacional de Psiquiatría Ramón de la Fuente Muñiz, Mexico City 14370, Mexico;
| | - Leonardo Ortiz-López
- Subdirección de Investigaciones Clínicas, Instituto Nacional de Psiquiatría Ramón de la Fuente Muñiz, Mexico City 14370, Mexico; (M.V.-T.); (L.O.-L.)
| | - Jesús Argueta
- Laboratorio de Neurofarmacología, Subdirección de Investigaciones Clínicas, Instituto Nacional de Psiquiatría Ramón de la Fuente Muñiz, Mexico City 14370, Mexico;
| | - Mauricio Rosel-Vales
- Dirección de Servicios Clínicos, Instituto Nacional de Psiquiatría Ramón de la Fuente Muñiz, Mexico City 14370, Mexico
| | - Roxana U. Miranda-Labra
- Departamento de Ciencias de la Salud, Universidad Autónoma Metropolitana (UAM)-Iztapalapa, Mexico City 09340, Mexico
| | - Beatriz Camarena
- Departamento de Farmacogenética, Subdirección de Investigaciones Clínicas, Instituto Nacional de Psiquiatría Ramón de la Fuente Muñiz, Mexico City 14370, Mexico;
| |
Collapse
|
16
|
Zeng Y, Buonfiglio F, Li J, Pfeiffer N, Gericke A. Mechanisms Underlying Vascular Inflammaging: Current Insights and Potential Treatment Approaches. Aging Dis 2025:AD.2024.0922. [PMID: 39812546 DOI: 10.14336/ad.2024.0922] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Accepted: 10/18/2024] [Indexed: 01/16/2025] Open
Abstract
Inflammaging refers to chronic, low-grade inflammation that becomes more common with age and plays a central role in the pathophysiology of various vascular diseases. Key inflammatory mediators involved in inflammaging contribute to endothelial dysfunction and accelerate the progression of atherosclerosis. In addition, specific pathological mechanisms and the role of inflammasomes have emerged as critical drivers of immune responses within the vasculature. A comprehensive understanding of these processes may lead to innovative treatment strategies that could significantly improve the management of age-related vascular diseases. Emerging therapeutic approaches, including cytokine inhibitors, senolytics, and specialized pro-resolving mediators, aim to counteract inflammaging and restore vascular health. This review seeks to provide an in-depth exploration of the molecular pathways underlying vascular inflammaging and highlight potential therapeutic interventions.
Collapse
|
17
|
Yadalam AK, Gold ME, Patel KJ, Liu C, Razavi AC, Jain V, Vatsa N, Gold D, Owais M, Haroun N, Sun YV, Quyyumi AA. Proteomics-Based Soluble Urokinase Plasminogen Activator Receptor Levels Are Associated With Incident Heart Failure Risk. JACC. ADVANCES 2025; 4:101442. [PMID: 39737138 PMCID: PMC11683231 DOI: 10.1016/j.jacadv.2024.101442] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/07/2024] [Revised: 10/28/2024] [Accepted: 11/06/2024] [Indexed: 01/01/2025]
Abstract
Background Higher soluble urokinase plasminogen activator receptor (suPAR) levels are associated with adverse outcomes in chronic heart failure (HF). Objectives The authors assessed the association between proteomics-based suPAR levels and incident HF risk in the general population. Methods In 40,418 UK Biobank participants without HF or coronary artery disease at enrollment, the association between Olink-based suPAR levels measured as relative protein expression levels and incident all-cause, ischemic, and nonischemic HF was analyzed by competing-risk regression, while accounting for all-cause death as a competing risk. The additional variability in incident HF risk attributable to suPAR levels beyond demographics, traditional risk factors, N-terminal pro B-type natriuretic peptide (NT-proBNP), and C-reactive protein (CRP) levels was assessed with nested Cox modeling and likelihood ratio testing. Results The mean age was 56 years; 45% were male, and 94% were White. During a median follow-up of 13.7 (IQR: 1.5) years, 1,428 (3.5%) incident HF events occurred. Proteomics-based suPAR levels (per 1-SD) were independently associated with incident HF (subdistribution HR (sHR): 1.37, 95% CI: 1.29-1.46), ischemic HF (sHR: 1.40, 95% CI: 1.28-1.54), and nonischemic HF (sHR: 1.32, 95% CI: 1.21-1.44) risk, after adjustment for demographics, traditional cardiovascular risk factors, NT-proBNP, and CRP levels. The addition of suPAR levels to a base risk factor model significantly improved the explained variability of incident HF risk (R 2 = 0.76 vs 0.73, P < 0.001). Conclusions Independent of demographics, traditional risk factors, NT-proBNP, and CRP levels, proteomics-based suPAR levels were significantly associated with incident all-cause, ischemic, and nonischemic HF risk. Proteomics-based measurement of suPAR levels may underestimate the effect size of this relationship.
Collapse
Affiliation(s)
- Adithya K. Yadalam
- Emory University School of Medicine, Division of Cardiology, Department of Medicine, Atlanta, Georgia, USA
| | - Matthew E. Gold
- Emory University School of Medicine, Division of Cardiology, Department of Medicine, Atlanta, Georgia, USA
| | - Krishan J. Patel
- Emory University School of Medicine, Division of Cardiology, Department of Medicine, Atlanta, Georgia, USA
| | - Chang Liu
- Emory University, Rollins School of Public Health, Department of Epidemiology, Atlanta, Georgia, USA
| | - Alexander C. Razavi
- Emory University School of Medicine, Division of Cardiology, Department of Medicine, Atlanta, Georgia, USA
| | - Vardhmaan Jain
- Emory University School of Medicine, Division of Cardiology, Department of Medicine, Atlanta, Georgia, USA
| | - Nishant Vatsa
- Emory University School of Medicine, Division of Cardiology, Department of Medicine, Atlanta, Georgia, USA
| | - Daniel Gold
- Emory University School of Medicine, Division of Cardiology, Department of Medicine, Atlanta, Georgia, USA
| | - Muhammad Owais
- Emory University School of Medicine, Division of Cardiology, Department of Medicine, Atlanta, Georgia, USA
| | - Nisreen Haroun
- Emory University School of Medicine, Division of Cardiology, Department of Medicine, Atlanta, Georgia, USA
| | - Yan V. Sun
- Emory University, Rollins School of Public Health, Department of Epidemiology, Atlanta, Georgia, USA
- Atlanta Veterans Affairs Healthcare System, Decatur, Georgia, USA
| | - Arshed A. Quyyumi
- Emory University School of Medicine, Division of Cardiology, Department of Medicine, Atlanta, Georgia, USA
| |
Collapse
|
18
|
Power E, Mongan D, Healy C, Susai SR, Föcking M, Zammit S, Cannon M, Cotter D. Cannabis use in youth is associated with chronic inflammation. Psychol Med 2024; 54:1-11. [PMID: 39648682 PMCID: PMC11779551 DOI: 10.1017/s0033291724002848] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/17/2022] [Revised: 10/15/2024] [Accepted: 10/22/2024] [Indexed: 12/10/2024]
Abstract
BACKGROUND Markers of inflammation and cannabis exposure are associated with an increased risk of mental disorders. In the current study, we investigated associations between cannabis use and biomarkers of inflammation. METHODS Utilizing a sample of 914 participants from the Avon Longitudinal Study of Parents and Children, we investigated whether interleukin-6 (IL-6), tumor necrosis factor α (TNFα), C-reactive protein (CRP), and soluble urokinase plasminogen activator receptor (suPAR) measured at age 24 were associated with past year daily cannabis use, less frequent cannabis use, and no past year cannabis use. We adjusted for a number of covariates including sociodemographic measures, body mass index, childhood trauma, and tobacco smoking. We found evidence of a strong association between daily or near daily cannabis use and suPAR. RESULTS We did not find any associations between less frequent cannabis use and suPAR. We did not find evidence of an association between IL-6, TNFα or CRP, and cannabis use. CONCLUSIONS Our finding that frequent cannabis use is strongly associated with suPAR, a biomarker of systemic chronic inflammation implicated in neurodevelopmental and neurodegenerative processes is novel. These findings may provide valuable insights into biological mechanisms by which cannabis affects the brain and impacts the risk of serious mental disorders.
Collapse
Affiliation(s)
- Emmet Power
- Department of Psychiatry, Royal College of Surgeons in Ireland, Smurfit Building, Beaumont Hospital, Dublin 9, Ireland
- Department of Liaison Psychiatry, Children's Health Ireland, Dublin 1, Ireland
| | - David Mongan
- Department of Psychiatry, Royal College of Surgeons in Ireland, Smurfit Building, Beaumont Hospital, Dublin 9, Ireland
- Centre for Public Health, Queens University Belfast, Belfast, UK
| | - Colm Healy
- Department of Psychiatry, Royal College of Surgeons in Ireland, Smurfit Building, Beaumont Hospital, Dublin 9, Ireland
- Centre for Clinical Brain Sciences, Division of Psychiatry, University of Edinburgh, Edinburgh, UK
- Department of Child and Adolescent Psychiatry, School of Medicine, University College Dublin, Dublin, Ireland
| | - Subash Raj Susai
- Department of Psychiatry, Royal College of Surgeons in Ireland, Smurfit Building, Beaumont Hospital, Dublin 9, Ireland
| | - Melanie Föcking
- Department of Psychiatry, Royal College of Surgeons in Ireland, Smurfit Building, Beaumont Hospital, Dublin 9, Ireland
| | - Stanley Zammit
- Centre for Academic Mental Health, Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK
- Division of Psychological Medicine and Clinical Neurosciences, MRC Centre for Neuropsychiatric Genetics and Genomics, Cardiff University School of Medicine, Cardiff, UK
| | - Mary Cannon
- Department of Psychiatry, Royal College of Surgeons in Ireland, Smurfit Building, Beaumont Hospital, Dublin 9, Ireland
- Department of Liaison Psychiatry, Beaumont Hospital, Dublin 9, Ireland
- FutureNeuro Research Ireland Centre, Royal College of Surgeons in Ireland, Smurfit Building, Beaumont Hospital, Dublin 9, Ireland
| | - David Cotter
- Department of Psychiatry, Royal College of Surgeons in Ireland, Smurfit Building, Beaumont Hospital, Dublin 9, Ireland
- Department of Liaison Psychiatry, Beaumont Hospital, Dublin 9, Ireland
- FutureNeuro Research Ireland Centre, Royal College of Surgeons in Ireland, Smurfit Building, Beaumont Hospital, Dublin 9, Ireland
| |
Collapse
|
19
|
Chen TK, Estrella MM, Katz R, Sarnak MJ, Grams ME, Cushman M, Levitan EB, Parikh CR, Kimmel PL, Bonventre JV, Coca SG, Gutiérrez OM, Ix JH, Shlipak MG. Plasma Biomarkers of Kidney Health and Mortality in Diabetes and Chronic Kidney Disease in the REGARDS Study. Clin J Am Soc Nephrol 2024; 19:1585-1593. [PMID: 39652331 PMCID: PMC11637710 DOI: 10.2215/cjn.0000000000000544] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2024] [Accepted: 09/17/2024] [Indexed: 12/14/2024]
Abstract
Key Points In diabetes and CKD, creatinine- and cystatin C–based eGFR has a strong inverse correlation with plasma TNF receptor 1, TNF receptor 2, and soluble urokinase-type plasminogen activator receptor. Higher plasma soluble TNF receptors 1 and 2 and soluble urokinase-type plasminogen activator receptor were each individually associated with mortality, independent of baseline kidney measures. Background Several plasma biomarkers of kidney health have been associated with CKD progression in persons with diabetes, but their associations with mortality risk have been largely unexplored. Methods In a random sample of 594 participants with diabetes and creatinine-based eGFR <60 ml/min per 1.73 m2 from the REGARDS cohort study, Cox proportional hazards regression was used to determine hazard ratios of mortality by plasma concentrations of soluble TNF receptors 1 and 2 (TNFR1 and TNFR2), soluble urokinase-type plasminogen activator receptor (suPAR), kidney injury molecule 1 (KIM-1), chitinase 3–like 1 (YKL-40), and monocyte chemotactic protein 1 (MCP-1). Covariates included sociodemographic and clinical factors, urine albumin-to-creatinine ratio (UACR), and creatinine- and cystatin C–based eGFR (eGFRcr-cys). Results At baseline, the mean age was 70 years, 47% were male, 53% self-identified as Black, mean±SD eGFRcr-cys was 41±13 ml/min per 1.73 m2, and median (interquartile range) UACR was 32 (9–224) mg/g. Correlations with eGFRcr-cys were stronger for TNFR1, TNFR2, and suPAR (r =−0.72 to −0.76) than for KIM-1, YKL-40, and MCP-1 (r =−0.10 to −0.40). With a median follow-up of 7 years, 332 participants died. In models adjusted for sociodemographic and clinical factors, each SD higher baseline concentration of plasma TNFR1 (hazard ratio [HR], 1.28; 95% confidence interval [CI], 1.20 to 1.38), TNFR2 (HR, 1.61; 95% CI, 1.42 to 1.82), suPAR (HR, 1.33; 95% CI, 1.22 to 1.44), KIM-1 (HR, 1.20; 95% CI, 1.08 to 1.33), and YKL-40 (HR, 1.23; 95% CI, 1.11 to 1.38) was associated with higher risk of all-cause mortality, whereas MCP-1 was not. Upon further adjustment for baseline eGFRcr-cys and UACR, only the associations for TNFR1 (HR, 1.16; 95% CI, 1.04 to 1.29), TNFR2 (HR, 1.34; 95% CI, 1.12 to 1.60), and suPAR (HR, 1.23; 95% CI, 1.11 to 1.36) persisted. Conclusions Among adults with diabetes and CKD, higher plasma TNFR1, TNFR2, and suPAR were associated with all-cause mortality, independent of baseline kidney function.
Collapse
Affiliation(s)
- Teresa K. Chen
- Kidney Health Research Collaborative and Department of Medicine, University of California, San Francisco and San Francisco VA Health Care System, San Francisco, California
| | - Michelle M. Estrella
- Kidney Health Research Collaborative and Department of Medicine, University of California, San Francisco and San Francisco VA Health Care System, San Francisco, California
| | - Ronit Katz
- Department of Obstetrics and Gynecology, University of Washington, Seattle, Washington
| | - Mark J. Sarnak
- Division of Nephrology, Department of Medicine, Tufts University School of Medicine, Boston, Massachusetts
| | - Morgan E. Grams
- Department of Medicine, New York University Langone School of Medicine, New York, New York
| | - Mary Cushman
- Departments of Medicine and Pathology and Laboratory Medicine, University of Vermont Larner College of Medicine, Burlington, Vermont
| | - Emily B. Levitan
- Department of Epidemiology, University of Alabama at Birmingham School of Public Health, Birmingham, Alabama
| | - Chirag R. Parikh
- Division of Nephrology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Paul L. Kimmel
- Division of Kidney, Urologic, and Hematologic Diseases, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland
| | - Joseph V. Bonventre
- Division of Renal Medicine and Engineering in Medicine Division, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Steven G. Coca
- Division of Nephrology, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Orlando M. Gutiérrez
- Division of Nephrology, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama
| | - Joachim H. Ix
- Division of Nephrology-Hypertension, Department of Medicine, University of California San Diego, and Veterans Affairs San Diego Healthcare System, San Diego, California
| | - Michael G. Shlipak
- Kidney Health Research Collaborative and Department of Medicine, University of California, San Francisco and San Francisco VA Health Care System, San Francisco, California
| |
Collapse
|
20
|
Almalki WH, Almujri SS. Aging, ROS, and cellular senescence: a trilogy in the progression of liver fibrosis. Biogerontology 2024; 26:10. [PMID: 39546058 DOI: 10.1007/s10522-024-10153-3] [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: 09/01/2024] [Accepted: 10/30/2024] [Indexed: 11/17/2024]
Abstract
Ageing is an inevitable and multifaceted biological process that impacts a wide range of cellular and molecular mechanisms, leading to the development of various diseases, such as liver fibrosis. Liver fibrosis progresses to cirrhosis, which is an advanced form due to high amounts of extracellular matrix and restoration of normal liver structure with failure to repair damaged tissue and cells, marking the end of liver function and total liver failure, ultimately death. The most important factors are reactive oxygen species (ROS) and cellular senescence. Oxidative stress is defined as an impairment by ROS, which are by-products of the mitochondrial electron transport chain and other key molecular pathways that induce cell damage and can activate cellular senescence pathways. Cellular senescence is characterized by pro-inflammatory cytokines, growth factors, and proteases secreted by senescent cells, collectively known as the senescence-associated secretory phenotype (SASP). The presence of senescent cells, which disrupt tissue architecture and function and increase senescent cell production in liver tissues, contributes to fibrogenesis. Hepatic stellate cells (HSCs) are activated in response to chronic liver injury, oxidative stress, and senescence signals that drive excessive production and deposition of extracellular matrix. This review article aims to provide a comprehensive overview of the pathogenic role of ROS and cellular senescence in the aging liver and their contribution to fibrosis.
Collapse
Affiliation(s)
- Waleed Hassan Almalki
- Department of Pharmacology, College of Pharmacy, Umm Al-Qura University, Makkah, Saudi Arabia
| | - Salem Salman Almujri
- Department of Pharmacology, College of Pharmacy, King Khalid University, 61421, Abha, Aseer, Saudi Arabia.
| |
Collapse
|
21
|
Wang X, Han SJ, Wang XL, Xu YF, Wang HC, Peng JY, Pan GM, Chen YH, Wang C. Soluble ST2 Is a Biomarker Associated With Left Ventricular Hypertrophy and Concentric Hypertrophy in Patients With Essential Hypertension. Am J Hypertens 2024; 37:987-994. [PMID: 39136164 PMCID: PMC11565189 DOI: 10.1093/ajh/hpae105] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2024] [Revised: 07/03/2024] [Accepted: 08/05/2024] [Indexed: 11/16/2024] Open
Abstract
BACKGROUND Elevated soluble stimulating factor 2 (sST2) level is observed in cardiovascular diseases, such as heart failure and acute coronary syndrome, which reflects myocardial fibrosis and hypertrophy, indicating adverse clinical outcomes. However, the association between sST2 and hypertensive heart disease are less understood. This study aimed to determine the relationship of sST2 with left ventricular hypertrophy (LVH) and geometric remodeling in essential hypertension (EH). METHODS We enrolled 483 patients (aged 18-80 years; 51.35% female). sST2 measurements and echocardiographic analyses were performed. RESULTS Stepwise multiple linear regression analysis showed significant associations among sST2, left ventricular (LV) mass, and LV mass index. The prevalence of LVH and concentric hypertrophy (CH) increased with higher sST2 grade levels (P for trend < 0.05). Logistic regression analysis suggested that the highest tertile of sST2 was significantly associated with increased LVH risk, compared with the lowest tertile (multivariate-adjusted odds ratio [OR] of highest group: 6.61; P < 0.001). Similar results were observed in the left ventricular geometric remodeling; the highest tertile of sST2 was significantly associated with increased CH risk (multivariate-adjusted OR of highest group: 5.80; P < 0.001). The receiver operating characteristic analysis results revealed that sST2 had potential predictive value for LVH (area under the curve [AUC]: 0.752, 95% confidence interval [CI]: 0.704-0.800) and CH (AUC: 0.750, 95% CI: 0.699-0.802) in patients with EH. CONCLUSIONS High sST2 level is strongly related to LVH and CH in patients with EH and can be used as a biomarker for the diagnosis and risk assessment of hypertensive heart disease. CLINICAL TRIALS REGISTRATION Trial Number ChiCTR2400082764.
Collapse
Affiliation(s)
- Xia Wang
- Department of Cardiovascular, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
- The Second Clinical College, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Shu-Jie Han
- Department of Cardiovascular, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
- The Second Clinical College, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Xiao-Li Wang
- Department of Cardiovascular, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
- The Second Clinical College, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Yun-Feng Xu
- The Second Clinical College, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Hui-Cheng Wang
- Department of Cardiovascular, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
- The Second Clinical College, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Jiang-Yang Peng
- Department of Cardiovascular, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
- The Second Clinical College, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Guang-Ming Pan
- Department of Cardiovascular, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
- The Second Clinical College, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Ya-Hui Chen
- Department of Cardiovascular, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
- The Second Clinical College, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Chuangchang Wang
- Department of Cardiovascular, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
- The Second Clinical College, Guangzhou University of Chinese Medicine, Guangzhou, China
| |
Collapse
|
22
|
Sajanti A, Hellström S, Bennett C, Srinath A, Jhaveri A, Cao Y, Takala R, Frantzén J, Koskimäki F, Falter J, Lyne SB, Rantamäki T, Posti JP, Roine S, Jänkälä M, Puolitaival J, Kolehmainen S, Girard R, Rahi M, Rinne J, Castrén E, Koskimäki J. Soluble Urokinase-Type Plasminogen Activator Receptor and Inflammatory Biomarker Response with Prognostic Significance after Acute Neuronal Injury - a Prospective Cohort Study. Inflammation 2024:10.1007/s10753-024-02185-1. [PMID: 39540961 DOI: 10.1007/s10753-024-02185-1] [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: 07/04/2024] [Revised: 10/30/2024] [Accepted: 11/05/2024] [Indexed: 11/16/2024]
Abstract
Aneurysmal subarachnoid hemorrhage (aSAH), ischemic stroke (IS), and traumatic brain injury (TBI) are severe conditions impacting individuals and society. Identifying reliable prognostic biomarkers for predicting survival or recovery remains a challenge. Soluble urokinase type plasminogen activator receptor (suPAR) has gained attention as a potential prognostic biomarker in acute sepsis. This study evaluates suPAR and related neuroinflammatory biomarkers in serum for brain injury prognosis. This prospective study included 31 aSAH, 30 IS, 13 TBI, and three healthy controls (n = 77). Serum samples were collected on average 5.9 days post-injury, analyzing suPAR, IL-1β, cyclophilin A, and TNFα levels using ELISA. Outcomes were assessed 90 days post-injury with the modified Rankin Scale (mRS), categorized as favorable (mRS 0-2) or unfavorable (mRS 3-6). Statistical analyses included 2-tailed t-tests, Pearson's correlations, and machine learning linear discriminant analysis (LDA) for biomarker combinations. Elevated suPAR levels were found in brain injury patients compared to controls (p = 0.017). Increased suPAR correlated with unfavorable outcomes (p = 0.0018) and showed prognostic value (AUC = 0.66, p = 0.03). IL-1β levels were higher in the unfavorable group (p = 0.0015). LDA combinatory analysis resulted a fair prognostic accuracy with canonical equation = 0.775[suPAR] + 0.667[IL1-β] (AUC = 0.77, OR 0.296, sensitivity 93.1%, specificity 53.1%, p = 0.0007). No correlation was found between suPAR and CRP or infection status. Elevated suPAR levels in acute brain injury patients were associated with poorer outcomes, highlighting suPAR's potential as a prognostic biomarker across different brain injury types. Combining IL-1β with suPAR improved prognostic accuracy, supporting a multimodal biomarker approach for predicting outcomes.
Collapse
Affiliation(s)
- Antti Sajanti
- Neurocenter, Department of Neurosurgery, Turku University Hospital and University of Turku, P.O. Box 52, Hämeentie 11, FI-20521, Turku, Finland
| | - Santtu Hellström
- Neurocenter, Department of Neurosurgery, Turku University Hospital and University of Turku, P.O. Box 52, Hämeentie 11, FI-20521, Turku, Finland
| | - Carolyn Bennett
- Neurovascular Surgery Program, Section of Neurosurgery, The University of Chicago Medicine and Biological Sciences, 5841 S. Maryland, Chicago, IL, 60637, USA
| | - Abhinav Srinath
- Neurovascular Surgery Program, Section of Neurosurgery, The University of Chicago Medicine and Biological Sciences, 5841 S. Maryland, Chicago, IL, 60637, USA
| | - Aditya Jhaveri
- Neurovascular Surgery Program, Section of Neurosurgery, The University of Chicago Medicine and Biological Sciences, 5841 S. Maryland, Chicago, IL, 60637, USA
| | - Ying Cao
- Department of Radiation Oncology, Kansas University Medical Center, Kansas City, KS, 66160, USA
| | - Riikka Takala
- Perioperative Services, Intensive Care and Pain Medicine, Turku University Hospital and University of Turku, POB 52, 20521, Turku, Finland
| | - Janek Frantzén
- Neurocenter, Department of Neurosurgery, Turku University Hospital and University of Turku, P.O. Box 52, Hämeentie 11, FI-20521, Turku, Finland
| | - Fredrika Koskimäki
- Neurocenter, Acute Stroke Unit, Turku University Hospital, P.O. Box 52, FI-20521, Turku, Finland
| | - Johannes Falter
- Department of Neurosurgery, University Medical Center of Regensburg, Regensburg, Germany
| | - Seán B Lyne
- Department of Neurosurgery, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Tomi Rantamäki
- Laboratory of Neurotherapeutics, Molecular and Integrative Biosciences Research Programme, Faculty of Biological and Environmental Sciences and Drug Research Program, Division of Pharmacology and Pharmacotherapy, Faculty of Pharmacy, University of Helsinki, Helsinki, Finland
| | - Jussi P Posti
- Neurocenter, Department of Neurosurgery, Turku University Hospital and University of Turku, P.O. Box 52, Hämeentie 11, FI-20521, Turku, Finland
| | - Susanna Roine
- Neurocenter, Acute Stroke Unit, Turku University Hospital, P.O. Box 52, FI-20521, Turku, Finland
| | - Miro Jänkälä
- Department of Neurosurgery, Oulu University Hospital, Box 25, 90029 OYS, Oulu, Finland
| | - Jukka Puolitaival
- Department of Neurosurgery, Oulu University Hospital, Box 25, 90029 OYS, Oulu, Finland
| | - Sulo Kolehmainen
- Neuroscience Center, HiLIFE, University of Helsinki, Box 63, 00014, Helsinki, Finland
| | - Romuald Girard
- Neurovascular Surgery Program, Section of Neurosurgery, The University of Chicago Medicine and Biological Sciences, 5841 S. Maryland, Chicago, IL, 60637, USA
| | - Melissa Rahi
- Neurocenter, Department of Neurosurgery, Turku University Hospital and University of Turku, P.O. Box 52, Hämeentie 11, FI-20521, Turku, Finland
| | - Jaakko Rinne
- Neurocenter, Department of Neurosurgery, Turku University Hospital and University of Turku, P.O. Box 52, Hämeentie 11, FI-20521, Turku, Finland
| | - Eero Castrén
- Neuroscience Center, HiLIFE, University of Helsinki, Box 63, 00014, Helsinki, Finland
| | - Janne Koskimäki
- Neurocenter, Department of Neurosurgery, Turku University Hospital and University of Turku, P.O. Box 52, Hämeentie 11, FI-20521, Turku, Finland.
- Department of Neurosurgery, Oulu University Hospital, Box 25, 90029 OYS, Oulu, Finland.
- Neuroscience Center, HiLIFE, University of Helsinki, Box 63, 00014, Helsinki, Finland.
| |
Collapse
|
23
|
Ruzanovic A, Saric-Matutinovic M, Milinkovic N, Jovicic S, Dimic A, Matejevic D, Kostic O, Koncar I, Ignjatovic S. Significance of myeloperoxidase, pentraxin-3 and soluble urokinase plasminogen activator receptor determination in patients with moderate carotid artery stenosis. Scand J Clin Lab Invest 2024; 84:486-492. [PMID: 39508179 DOI: 10.1080/00365513.2024.2422404] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2024] [Revised: 05/30/2024] [Accepted: 10/20/2024] [Indexed: 11/08/2024]
Abstract
We investigated serum concentrations of specific inflammatory parameters in patients with significant carotid artery stenosis (CAS) of 50-99%, with an additional focus on patients with moderate stenosis (50-69%), in terms of both symptomatic status and plaque morphology, to determine whether there are certain parameters that can be associated with plaque instability before the progression of CAS to a high degree. The study included 119 CAS patients, 29 of whom had moderate stenosis, and 46 controls. Ultrasonography of the carotid arteries was performed using color flow Doppler and B-mode duplex ultrasound, and serum inflammatory parameters were measured using commercially available enzyme immunoassays. When comparing patients with 50-99% stenosis, only serum amyloid A (SAA) was higher in symptomatic patients, while in the group of patients with 50-69% stenosis, myeloperoxidase (MPO) was higher and pentraxin-3 (PTX-3) was lower in symptomatic compared to asymptomatic patients, and soluble urokinase plasminogen activator receptor (suPAR) was higher in patients with carotid plaque of unstable compared to stable morphology. Our results suggest that the importance of different inflammatory parameters in patients with moderate CAS is not the same as in CAS patients in general, and therefore their separate investigation in patients with high and moderate stenosis may be beneficial. SAA has the potential to be further considered in research to predict CAS symptom risk. There is a possibility that MPO and PTX-3 play a role in the development of CAS symptoms originating from less stenotic plaques and that suPAR is involved in the destabilisation of such plaques.
Collapse
Affiliation(s)
- Ana Ruzanovic
- Faculty of Pharmacy, University of Belgrade, Belgrade, Serbia
| | | | - Neda Milinkovic
- Faculty of Pharmacy, University of Belgrade, Belgrade, Serbia
| | - Snezana Jovicic
- Faculty of Pharmacy, University of Belgrade, Belgrade, Serbia
| | - Andreja Dimic
- University Clinical Center of Serbia, Clinic for Vascular and Endovascular surgery, Belgrade, Serbia
- Faculty of Medicine, University of Belgrade, Belgrade, Serbia
| | - David Matejevic
- University Clinical Center of Serbia, Clinic for Vascular and Endovascular surgery, Belgrade, Serbia
| | - Ognjen Kostic
- University Clinical Center of Serbia, Clinic for Vascular and Endovascular surgery, Belgrade, Serbia
| | - Igor Koncar
- University Clinical Center of Serbia, Clinic for Vascular and Endovascular surgery, Belgrade, Serbia
- Faculty of Medicine, University of Belgrade, Belgrade, Serbia
| | | |
Collapse
|
24
|
Gavriilaki E, Demosthenous C, Evangelidis P, Bousiou Z, Batsis I, Vardi A, Mallouri D, Koravou EE, Spyridis N, Panteliadou A, Karavalakis G, Masmanidou M, Touloumenidou T, Papalexandri A, Poziopoulos C, Yannaki E, Sakellari I, Politou M, Papassotiriou I. Soluble Urokinase-Type Plasminogen Activator Receptor (suPAR), Growth Differentiation Factor-15 (GDF-15), and Soluble C5b-9 (sC5b-9) Levels Are Significantly Associated with Endothelial Injury Indices in CAR-T Cell Recipients. Int J Mol Sci 2024; 25:11028. [PMID: 39456810 PMCID: PMC11507105 DOI: 10.3390/ijms252011028] [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: 08/26/2024] [Revised: 10/09/2024] [Accepted: 10/13/2024] [Indexed: 10/28/2024] Open
Abstract
Endothelial injury indices, such as Endothelial Activation and Stress Index (EASIX), modified EASIX (m-EASIX), and simplified EASIX (s-EASIX) scores, have been previously associated with chimeric antigen receptor-T (CAR-T) cell immunotherapy complications. Soluble urokinase-type plasminogen activator receptor (suPAR), growth differentiation factor-15 (GDF-15), and soluble C5b-9 (sC5b-9) have been described as markers of endothelial injury post-hematopoietic stem cell transplantation. In the current study, we examined whether suPAR, GDF-15, and sC5b-9 levels were associated with endothelial injury indices in adult CAR-T cell recipients. The levels of these markers were measured in patients before CAR-T cell infusion and in healthy individuals with immunoenzymatic methods. We studied 45 CAR-T cell recipients and 20 healthy individuals as the control group. SuPAR, GDF-15, and sC5b-9 levels were significantly higher in the patients' group compared to the healthy control group (p < 0.001, in all comparisons). SuPAR levels at baseline were associated with the m-EASIX scores calculated at the same time point (p = 0.020), while suPAR and GDF-15 concentrations were correlated with EASIX scores at day 14 post-infusion (p < 0.001 in both comparisons). Moreover, sC5b-9 levels were correlated with the s-EASIX scores at infusion (p = 0.008) and the EASIX scores at day 14 (p = 0.005). In our study, sC5b9, suPAR, and GDF-15 levels were found to reflect endothelial injury in CAR-T cell recipients.
Collapse
Affiliation(s)
- Eleni Gavriilaki
- BMT Unit, Hematology Department, George Papanicolaou General Hospital, 57010 Thessaloniki, Greece; (C.D.); (Z.B.); (I.B.); (A.V.); (D.M.); (E.-E.K.); (N.S.); (A.P.); (G.K.); (M.M.); (T.T.); (A.P.); (E.Y.); (I.S.)
- Second Propedeutic Department of Internal Medicine, Hippocration Hospital, Aristotle University of Thessaloniki, 54642 Thessaloniki, Greece;
| | - Christos Demosthenous
- BMT Unit, Hematology Department, George Papanicolaou General Hospital, 57010 Thessaloniki, Greece; (C.D.); (Z.B.); (I.B.); (A.V.); (D.M.); (E.-E.K.); (N.S.); (A.P.); (G.K.); (M.M.); (T.T.); (A.P.); (E.Y.); (I.S.)
| | - Paschalis Evangelidis
- Second Propedeutic Department of Internal Medicine, Hippocration Hospital, Aristotle University of Thessaloniki, 54642 Thessaloniki, Greece;
| | - Zoi Bousiou
- BMT Unit, Hematology Department, George Papanicolaou General Hospital, 57010 Thessaloniki, Greece; (C.D.); (Z.B.); (I.B.); (A.V.); (D.M.); (E.-E.K.); (N.S.); (A.P.); (G.K.); (M.M.); (T.T.); (A.P.); (E.Y.); (I.S.)
| | - Ioannis Batsis
- BMT Unit, Hematology Department, George Papanicolaou General Hospital, 57010 Thessaloniki, Greece; (C.D.); (Z.B.); (I.B.); (A.V.); (D.M.); (E.-E.K.); (N.S.); (A.P.); (G.K.); (M.M.); (T.T.); (A.P.); (E.Y.); (I.S.)
| | - Anna Vardi
- BMT Unit, Hematology Department, George Papanicolaou General Hospital, 57010 Thessaloniki, Greece; (C.D.); (Z.B.); (I.B.); (A.V.); (D.M.); (E.-E.K.); (N.S.); (A.P.); (G.K.); (M.M.); (T.T.); (A.P.); (E.Y.); (I.S.)
| | - Despina Mallouri
- BMT Unit, Hematology Department, George Papanicolaou General Hospital, 57010 Thessaloniki, Greece; (C.D.); (Z.B.); (I.B.); (A.V.); (D.M.); (E.-E.K.); (N.S.); (A.P.); (G.K.); (M.M.); (T.T.); (A.P.); (E.Y.); (I.S.)
| | - Eudoxia-Evaggelia Koravou
- BMT Unit, Hematology Department, George Papanicolaou General Hospital, 57010 Thessaloniki, Greece; (C.D.); (Z.B.); (I.B.); (A.V.); (D.M.); (E.-E.K.); (N.S.); (A.P.); (G.K.); (M.M.); (T.T.); (A.P.); (E.Y.); (I.S.)
| | - Nikolaos Spyridis
- BMT Unit, Hematology Department, George Papanicolaou General Hospital, 57010 Thessaloniki, Greece; (C.D.); (Z.B.); (I.B.); (A.V.); (D.M.); (E.-E.K.); (N.S.); (A.P.); (G.K.); (M.M.); (T.T.); (A.P.); (E.Y.); (I.S.)
| | - Alkistis Panteliadou
- BMT Unit, Hematology Department, George Papanicolaou General Hospital, 57010 Thessaloniki, Greece; (C.D.); (Z.B.); (I.B.); (A.V.); (D.M.); (E.-E.K.); (N.S.); (A.P.); (G.K.); (M.M.); (T.T.); (A.P.); (E.Y.); (I.S.)
| | - Georgios Karavalakis
- BMT Unit, Hematology Department, George Papanicolaou General Hospital, 57010 Thessaloniki, Greece; (C.D.); (Z.B.); (I.B.); (A.V.); (D.M.); (E.-E.K.); (N.S.); (A.P.); (G.K.); (M.M.); (T.T.); (A.P.); (E.Y.); (I.S.)
| | - Marianna Masmanidou
- BMT Unit, Hematology Department, George Papanicolaou General Hospital, 57010 Thessaloniki, Greece; (C.D.); (Z.B.); (I.B.); (A.V.); (D.M.); (E.-E.K.); (N.S.); (A.P.); (G.K.); (M.M.); (T.T.); (A.P.); (E.Y.); (I.S.)
| | - Tasoula Touloumenidou
- BMT Unit, Hematology Department, George Papanicolaou General Hospital, 57010 Thessaloniki, Greece; (C.D.); (Z.B.); (I.B.); (A.V.); (D.M.); (E.-E.K.); (N.S.); (A.P.); (G.K.); (M.M.); (T.T.); (A.P.); (E.Y.); (I.S.)
| | - Apostolia Papalexandri
- BMT Unit, Hematology Department, George Papanicolaou General Hospital, 57010 Thessaloniki, Greece; (C.D.); (Z.B.); (I.B.); (A.V.); (D.M.); (E.-E.K.); (N.S.); (A.P.); (G.K.); (M.M.); (T.T.); (A.P.); (E.Y.); (I.S.)
| | | | - Evangelia Yannaki
- BMT Unit, Hematology Department, George Papanicolaou General Hospital, 57010 Thessaloniki, Greece; (C.D.); (Z.B.); (I.B.); (A.V.); (D.M.); (E.-E.K.); (N.S.); (A.P.); (G.K.); (M.M.); (T.T.); (A.P.); (E.Y.); (I.S.)
| | - Ioanna Sakellari
- BMT Unit, Hematology Department, George Papanicolaou General Hospital, 57010 Thessaloniki, Greece; (C.D.); (Z.B.); (I.B.); (A.V.); (D.M.); (E.-E.K.); (N.S.); (A.P.); (G.K.); (M.M.); (T.T.); (A.P.); (E.Y.); (I.S.)
| | - Marianna Politou
- Thrombosis–Bleeding–Transfusion Medicine Postgraduate Studies, School of Medicine, National and Kapodistrian University of Athens, 11527 Athens, Greece;
- Hematology Laboratory-Blood Bank, Aretaieion Hospital, School of Medicine, National and Kapodistrian University of Athens, 11527 Athens, Greece
| | - Ioannis Papassotiriou
- First Department of Pediatrics, School of Medicine, National and Kapodistrian University of Athens, 11527 Athens, Greece;
| |
Collapse
|
25
|
Vastrad B, Vastrad C. Screening and identification of key biomarkers associated with endometriosis using bioinformatics and next-generation sequencing data analysis. EGYPTIAN JOURNAL OF MEDICAL HUMAN GENETICS 2024; 25:116. [DOI: 10.1186/s43042-024-00572-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2024] [Accepted: 08/23/2024] [Indexed: 01/04/2025] Open
Abstract
Abstract
Background
Endometriosis is a common cause of endometrial-type mucosa outside the uterine cavity with symptoms such as painful periods, chronic pelvic pain, pain with intercourse and infertility. However, the early diagnosis of endometriosis is still restricted. The purpose of this investigation is to identify and validate the key biomarkers of endometriosis.
Methods
Next-generation sequencing dataset GSE243039 was obtained from the Gene Expression Omnibus database, and differentially expressed genes (DEGs) between endometriosis and normal control samples were identified. After screening of DEGs, gene ontology (GO) and REACTOME pathway enrichment analyses were performed. Furthermore, a protein–protein interaction (PPI) network was constructed and modules were analyzed using the Human Integrated Protein–Protein Interaction rEference database and Cytoscape software, and hub genes were identified. Subsequently, a network between miRNAs and hub genes, and network between TFs and hub genes were constructed using the miRNet and NetworkAnalyst tool, and possible key miRNAs and TFs were predicted. Finally, receiver operating characteristic curve analysis was used to validate the hub genes.
Results
A total of 958 DEGs, including 479 upregulated genes and 479 downregulated genes, were screened between endometriosis and normal control samples. GO and REACTOME pathway enrichment analyses of the 958 DEGs showed that they were mainly involved in multicellular organismal process, developmental process, signaling by GPCR and muscle contraction. Further analysis of the PPI network and modules identified 10 hub genes, including vcam1, snca, prkcb, adrb2, foxq1, mdfi, actbl2, prkd1, dapk1 and actc1. Possible target miRNAs, including hsa-mir-3143 and hsa-mir-2110, and target TFs, including tcf3 (transcription factor 3) and clock (clock circadian regulator), were predicted by constructing a miRNA-hub gene regulatory network and TF-hub gene regulatory network.
Conclusions
This investigation used bioinformatics techniques to explore the potential and novel biomarkers. These biomarkers might provide new ideas and methods for the early diagnosis, treatment and monitoring of endometriosis.
Collapse
|
26
|
Tavenier J, Nehlin JO, Houlind MB, Rasmussen LJ, Tchkonia T, Kirkland JL, Andersen O, Rasmussen LJH. Fisetin as a senotherapeutic agent: Evidence and perspectives for age-related diseases. Mech Ageing Dev 2024; 222:111995. [PMID: 39384074 DOI: 10.1016/j.mad.2024.111995] [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: 08/07/2024] [Revised: 09/25/2024] [Accepted: 09/30/2024] [Indexed: 10/11/2024]
Abstract
Fisetin, a flavonoid naturally occurring in plants, fruits, and vegetables, has recently gained attention for its potential role as a senotherapeutic agent for the treatment of age-related chronic diseases. Senotherapeutics target senescent cells, which accumulate with age and disease, in both circulating immune cell populations and solid organs and tissues. Senescent cells contribute to development of many chronic diseases, primarily by eliciting systemic chronic inflammation through their senescence-associated secretory phenotype. Here, we explore whether fisetin as a senotherapeutic can eliminate senescent cells, and thereby alleviate chronic diseases, by examining current evidence from in vitro studies and animal models that investigate fisetin's impact on age-related diseases, as well as from phase I/II trials in various patient populations. We discuss the application of fisetin in humans, including challenges and future directions. Our review of available data suggests that targeting senescent cells with fisetin offers a promising strategy for managing multiple chronic diseases, potentially transforming future healthcare for older and multimorbid patients. However, further studies are needed to establish the safety, pharmacokinetics, and efficacy of fisetin as a senotherapeutic, identify relevant and reliable outcome measures in human trials, optimize dosing, and better understand the possible limitations of fisetin as a senotherapeutic agent.
Collapse
Affiliation(s)
- Juliette Tavenier
- Department of Clinical Research, Copenhagen University Hospital Amager and Hvidovre, Kettegaard Allé 30, Hvidovre 2650, Denmark.
| | - Jan O Nehlin
- Department of Clinical Research, Copenhagen University Hospital Amager and Hvidovre, Kettegaard Allé 30, Hvidovre 2650, Denmark.
| | - Morten Baltzer Houlind
- Department of Clinical Research, Copenhagen University Hospital Amager and Hvidovre, Kettegaard Allé 30, Hvidovre 2650, Denmark; The Hospital Pharmacy, Marielundsvej 25, Herlev 2730, Denmark; Department of Drug Design and Pharmacology, University of Copenhagen, Universitetsparken 2, Copenhagen 2100, Denmark.
| | - Lene Juel Rasmussen
- Center for Healthy Aging, Department of Cellular and Molecular Medicine, University of Copenhagen, Blegdamsvej 3B, Copenhagen 2200, Denmark.
| | - Tamara Tchkonia
- Center for Advanced Gerotherapeutics, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA; Division of Endocrinology, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA; Division of General Internal Medicine, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA.
| | - James L Kirkland
- Center for Advanced Gerotherapeutics, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA; Division of Endocrinology, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA; Division of General Internal Medicine, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA.
| | - Ove Andersen
- Department of Clinical Research, Copenhagen University Hospital Amager and Hvidovre, Kettegaard Allé 30, Hvidovre 2650, Denmark; Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3B, Copenhagen N 2200, Denmark; The Emergency Department, Copenhagen University Hospital Amager and Hvidovre, Kettegaard Allé 30, Hvidovre 2650, Denmark.
| | - Line Jee Hartmann Rasmussen
- Department of Clinical Research, Copenhagen University Hospital Amager and Hvidovre, Kettegaard Allé 30, Hvidovre 2650, Denmark; Department of Psychology & Neuroscience, Duke University, 2020 West Main Street Suite 201, Durham, NC 27708, USA.
| |
Collapse
|
27
|
Christaki M, Samanidou V, Liontos A, Konstantopoulou R, Milionis H. Post-COVID-19 Multisystem Inflammatory Syndrome in Adults (MIS-A) With Elevated Levels of Soluble Urokinase Plasminogen Activator Receptor (suPAR) Treated With Anakinra: A Case Report. Cureus 2024; 16:e70848. [PMID: 39493146 PMCID: PMC11531791 DOI: 10.7759/cureus.70848] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/04/2024] [Indexed: 11/05/2024] Open
Abstract
The COVID-19 pandemic has brought attention to a newly identified syndrome of multisystem inflammation. This potentially fatal complication of the disease was initially observed in children and later in adults. It affects primarily unvaccinated patients and may manifest within a timeframe of 2-12 weeks following infection. Soluble urokinase plasminogen activator receptor (suPAR), a novel biomarker, highlights the severity of inflammation and the degree of immune system activation. Herein, we report a case of a patient with multisystem inflammatory syndrome in adults (MIS-A) and markedly elevated suPAR levels, successfully treated with interleukin-1 (IL-1) receptor antagonist. A 59-year-old female was admitted to our hospital due to febrile illness (up to 40°C) with chills, vomiting, non-bloody diarrhea, and abdominal pain for four days prior to her admission. She tested positive for SARS-CoV-2 12 weeks before her presentation. During hospitalization, the patient deteriorated clinically with multiorgan involvement and hemodynamically instability, with concomitant markedly elevated inflammatory markers. Extensive workup with high suPAR levels led to post-COVID-19 MIS-A diagnosis, and treatment with dexamethasone and an interleukin-1 receptor antagonist (IL-1ra), anakinra, was administered. The subcutaneous injection of anakinra effectively and safely deterred MIS-A. Further research is needed to investigate the role of interleukin-1 inhibitors for the management of this potentially life-threatening condition.
Collapse
Affiliation(s)
- Maria Christaki
- 1st Division of Internal Medicine and Infectious Diseases Unit, University Hospital of Ioannina, Faculty of Medicine, University of Ioannina, Ioannina, GRC
| | - Valentini Samanidou
- 1st Division of Internal Medicine and Infectious Diseases Unit, University Hospital of Ioannina, Faculty of Medicine, University of Ioannina, Ioannina, GRC
| | - Angelos Liontos
- 1st Division of Internal Medicine and Infectious Diseases Unit, University Hospital of Ioannina, Faculty of Medicine, University of Ioannina, Ioannina, GRC
| | - Revekka Konstantopoulou
- 1st Division of Internal Medicine and Infectious Diseases Unit, University Hospital of Ioannina, Faculty of Medicine, University of Ioannina, Ioannina, GRC
| | - Haralampos Milionis
- 1st Division of Internal Medicine and Infectious Diseases Unit, University Hospital of Ioannina, Faculty of Medicine, University of Ioannina, Ioannina, GRC
| |
Collapse
|
28
|
Urban BC, Gonçalves ANA, Loukov D, Passos FM, Reiné J, Gonzalez-Dias P, Solórzano C, Mitsi E, Nikolaou E, O'Connor D, Collins AM, Adler H, Pollard A, Rylance J, Gordon SB, Jochems SP, Nakaya HI, Ferreira DM. Inflammation of the nasal mucosa is associated with susceptibility to experimental pneumococcal challenge in older adults. Mucosal Immunol 2024; 17:973-989. [PMID: 38950826 PMCID: PMC11464406 DOI: 10.1016/j.mucimm.2024.06.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2024] [Revised: 06/25/2024] [Accepted: 06/26/2024] [Indexed: 07/03/2024]
Abstract
Streptococcus pneumoniae colonization in the upper respiratory tract is linked to pneumococcal disease development, predominantly affecting young children and older adults. As the global population ages and comorbidities increase, there is a heightened concern about this infection. We investigated the immunological responses of older adults to pneumococcal-controlled human infection by analyzing the cellular composition and gene expression in the nasal mucosa. Our comparative analysis with data from a concurrent study in younger adults revealed distinct gene expression patterns in older individuals susceptible to colonization, highlighted by neutrophil activation and elevated levels of CXCL9 and CXCL10. Unlike younger adults challenged with pneumococcus, older adults did not show recruitment of monocytes into the nasal mucosa following nasal colonization. However, older adults who were protected from colonization showed increased degranulation of cluster of differentiation 8+ T cells, both before and after pneumococcal challenge. These findings suggest age-associated cellular changes, in particular enhanced mucosal inflammation, that may predispose older adults to pneumococcal colonization.
Collapse
Affiliation(s)
- Britta C Urban
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford, Oxford, UK; Clinical Sciences, Liverpool School of Tropical Medicine, Liverpool, UK; NIHR Oxford Biomedical Research Centre, Oxford, UK.
| | - André N A Gonçalves
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford, Oxford, UK; Clinical Sciences, Liverpool School of Tropical Medicine, Liverpool, UK; NIHR Oxford Biomedical Research Centre, Oxford, UK
| | - Dessi Loukov
- Clinical Sciences, Liverpool School of Tropical Medicine, Liverpool, UK
| | - Fernando M Passos
- Clinical Sciences, Liverpool School of Tropical Medicine, Liverpool, UK
| | - Jesús Reiné
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford, Oxford, UK; Clinical Sciences, Liverpool School of Tropical Medicine, Liverpool, UK; NIHR Oxford Biomedical Research Centre, Oxford, UK
| | - Patrícia Gonzalez-Dias
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford, Oxford, UK; Clinical Sciences, Liverpool School of Tropical Medicine, Liverpool, UK; NIHR Oxford Biomedical Research Centre, Oxford, UK
| | - Carla Solórzano
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford, Oxford, UK; Clinical Sciences, Liverpool School of Tropical Medicine, Liverpool, UK; NIHR Oxford Biomedical Research Centre, Oxford, UK
| | - Elena Mitsi
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford, Oxford, UK; Clinical Sciences, Liverpool School of Tropical Medicine, Liverpool, UK; NIHR Oxford Biomedical Research Centre, Oxford, UK
| | - Elissavet Nikolaou
- Clinical Sciences, Liverpool School of Tropical Medicine, Liverpool, UK; Infection, Immunity and Global Health, Murdoch Children's Research Institute, Parkville, Victoria, Australia; Department of Microbiology and Immunology at the Peter Doherty Institute for Infection and Immunity, The University of Melbourne, Parkville, Victoria, Australia
| | - Daniel O'Connor
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford, Oxford, UK; NIHR Oxford Biomedical Research Centre, Oxford, UK
| | - Andrea M Collins
- Clinical Sciences, Liverpool School of Tropical Medicine, Liverpool, UK; University Hospital Aintree, Liverpool University Hospitals Trust, Liverpool, UK
| | - Hugh Adler
- Clinical Sciences, Liverpool School of Tropical Medicine, Liverpool, UK
| | - Andrew Pollard
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford, Oxford, UK; NIHR Oxford Biomedical Research Centre, Oxford, UK
| | - Jamie Rylance
- Clinical Sciences, Liverpool School of Tropical Medicine, Liverpool, UK
| | - Stephen B Gordon
- Clinical Sciences, Liverpool School of Tropical Medicine, Liverpool, UK; Malawi-Liverpool-Wellcome Clinical Research Programme, Blantyre, Malawi
| | - Simon P Jochems
- Leiden University Centre for Infectious Diseases, Leiden University Medical Centre, Leiden, The Netherlands
| | - Helder I Nakaya
- Hospital Israelita Albert Einstein, São Paulo, Brazil; Department of Clinical and Toxicological Analysis, School of Pharmaceutical Sciences, University of São Paulo, São Paulo, Brazil
| | - Daniela M Ferreira
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford, Oxford, UK; Clinical Sciences, Liverpool School of Tropical Medicine, Liverpool, UK; NIHR Oxford Biomedical Research Centre, Oxford, UK.
| |
Collapse
|
29
|
Christensen LWS, Iversen E, Andersen AL, Walls AB, Rasmussen LJH, Andersen O, Kallemose T, Houlind MB. A characterization of patients with low soluble urokinase plasminogen activator receptor who died within 90 days of hospital discharge. Basic Clin Pharmacol Toxicol 2024; 135:364-371. [PMID: 38988231 DOI: 10.1111/bcpt.14050] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2024] [Revised: 05/29/2024] [Accepted: 06/27/2024] [Indexed: 07/12/2024]
Abstract
Soluble urokinase plasminogen activator receptor (suPAR) is a marker of systemic chronic inflammation. Elevated suPAR levels are associated with adverse clinical outcomes, but a small subset of patients with low suPAR also experience poor outcomes. Therefore, we aimed to characterize patients presenting to the emergency department with low suPAR (<3 ng/mL) who died within 90 days after discharge in a registry-based study. Compared to patients with low suPAR who survived (n = 15 122), those who died within 90 days (n = 87) had higher age (75.4 years), higher medication use (7.0; 71.3% with polypharmacy) and more blood tests outside reference intervals (5.0) (including C-reactive protein, neutrophils and albumin), and the most common diagnoses were chronic pulmonary disease (27.6%), cerebrovascular disease (18.4%) and dementia (11.5%). Patients with low suPAR were more morbid than what was reflected by suPAR alone. Future studies must determine which factors that contribute the most to potential algorithms when stratifying patients based on their risk of adverse clinical outcomes. These data indicate that inclusion of medication data could be relevant.
Collapse
Affiliation(s)
- Louise Westberg Strejby Christensen
- Department of Clinical Research, Copenhagen University Hospital Amager and Hvidovre, Hvidovre, Denmark
- The Capital Region Pharmacy, Herlev, Denmark
- Department of Drug Design and Pharmacology, University of Copenhagen, Copenhagen, Denmark
- Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Esben Iversen
- Department of Clinical Research, Copenhagen University Hospital Amager and Hvidovre, Hvidovre, Denmark
| | - Aino Leegaard Andersen
- Department of Clinical Research, Copenhagen University Hospital Amager and Hvidovre, Hvidovre, Denmark
| | - Anne Byriel Walls
- The Capital Region Pharmacy, Herlev, Denmark
- Department of Drug Design and Pharmacology, University of Copenhagen, Copenhagen, Denmark
- Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Line Jee Hartmann Rasmussen
- Department of Clinical Research, Copenhagen University Hospital Amager and Hvidovre, Hvidovre, Denmark
- Department of Psychology and Neuroscience, Duke University, Durham, North Carolina, USA
| | - Ove Andersen
- Department of Clinical Research, Copenhagen University Hospital Amager and Hvidovre, Hvidovre, Denmark
- Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
- Emergency Department, Copenhagen University Hospital Amager and Hvidovre, Hvidovre, Denmark
| | - Thomas Kallemose
- Department of Clinical Research, Copenhagen University Hospital Amager and Hvidovre, Hvidovre, Denmark
| | - Morten Baltzer Houlind
- Department of Clinical Research, Copenhagen University Hospital Amager and Hvidovre, Hvidovre, Denmark
- The Capital Region Pharmacy, Herlev, Denmark
- Department of Drug Design and Pharmacology, University of Copenhagen, Copenhagen, Denmark
- Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
30
|
Zhang B, Liao R. Early Serum Biomarkers of Cardiovascular Disease in Elderly Patients with Chronic Kidney Disease. Cardiorenal Med 2024; 14:508-520. [PMID: 39217975 DOI: 10.1159/000541014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/07/2024] [Accepted: 08/12/2024] [Indexed: 09/04/2024] Open
Abstract
BACKGROUND The global population is aging. It is estimated that by 2050, the proportion of the elderly population will reach 16%. Various studies have suggested that elderly people have a greater incidence of CKD. These elderly patients are also susceptible to cardiovascular disease (CVD), which is the leading cause of death, resulting in poor prognosis in this population. However, CVD in such patients is often insidious and lacks early markers for effective evaluation. Fortunately, several studies have recently proposed biomarkers associated with this process. SUMMARY This study aimed to summarize the early biomarkers of CVD in elderly patients with CKD to provide a basis for its prevention and treatment. KEY MESSAGES This review outlines four categories of potential early biomarkers. All of them have been shown to have some clinical value for these patients, but more research is still needed.
Collapse
Affiliation(s)
- Bohua Zhang
- Department of Nephrology, West China Hospital of Sichuan University, Chengdu, China
| | - Ruoxi Liao
- Department of Nephrology, West China Hospital of Sichuan University, Chengdu, China
| |
Collapse
|
31
|
Yatsenko T, Rios R, Nogueira T, Salama Y, Takahashi S, Adachi E, Tabe Y, Hattori N, Osada T, Naito T, Takahashi K, Hattori K, Heissig B. The influence of 4G/5G polymorphism in the plasminogen-activator-inhibitor-1 promoter on COVID-19 severity and endothelial dysfunction. Front Immunol 2024; 15:1445294. [PMID: 39281671 PMCID: PMC11392769 DOI: 10.3389/fimmu.2024.1445294] [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: 06/07/2024] [Accepted: 08/08/2024] [Indexed: 09/18/2024] Open
Abstract
Introduction Plasminogen activator inhibitor-1 (PAI-1) is linked to thrombosis and endothelial dysfunction in severe COVID-19. The +43 G>A PAI-1 and 4G/5G promoter polymorphism can influence PAI-1 expression. The 4G5G PAI-1 promoter gene polymorphism constitutes the 4G4G, 4G5G, and 5G5G genotypes. However, the impact of PAI-1 polymorphisms on disease severity or endothelial dysfunction remains unclear. Methods Clinical data, sera, and peripheral blood mononuclear cells (PBMCs) of COVID-19 patients were studied. Results Comorbidities and clinical biomarkers did not correlate with genotypes in either polymorphism. However, differences between fibrinolytic factors and interleukin-1β (IL-1β) were identified in genotypes of the 4G/5G but not the 43 G>A PAI polymorphism. Patients with the 4G4G genotype of the 4G/5G polymorphism showed high circulating PAI-1, mainly complexed with plasminogen activators, and low IL-1β and plasmin levels, indicating suppressed fibrinolysis. NFκB was upregulated in PBMCs of COVID-19 patients with the 4G4G genotype. Discussion Mechanistically, IL-1β enhanced PAI-1 expression in 4G4G endothelial cells, preventing the generation of plasmin and cleavage products like angiostatin, soluble uPAR, and VCAM1. We identified inflammation-induced endothelial dysfunction coupled with fibrinolytic system overactivation as a risk factor for patients with the 5G5G genotype.
Collapse
Affiliation(s)
- Tetiana Yatsenko
- Department of Research Support Utilizing Bioresource Bank, Graduate School of Medicine, Juntendo University, Tokyo, Japan
- Department of Enzymes Chemistry and Biochemistry, Palladin Institute of Biochemistry of National Academy of Sciences of Ukraine, Kyiv, Ukraine
| | - Ricardo Rios
- Institute of Computing, Federal University of Bahia, Salvador, Bahia, Brazil
| | - Tatiane Nogueira
- Institute of Computing, Federal University of Bahia, Salvador, Bahia, Brazil
| | - Yousef Salama
- An-Najah Center for Cancer and Stem Cell Research, Faculty of Medicine and Health Sciences, An-Najah National University, Nablus, Palestine
| | - Satoshi Takahashi
- Division of Clinical Precision Research Platform, the Institute of Medical Science, the University of Tokyo, Tokyo, Japan
| | - Eisuke Adachi
- Department of Infectious Diseases and Applied Immunology, the Institute of Medical Science, the University of Tokyo, Tokyo, Japan
| | - Yoko Tabe
- Department of Research Support Utilizing Bioresource Bank, Graduate School of Medicine, Juntendo University, Tokyo, Japan
| | - Nobutaka Hattori
- Center for Genome and Regenerative Medicine, Juntendo University, Graduate School of Medicine, Tokyo, Japan
| | - Taro Osada
- Department of Gastroenterology, Juntendo University, Urayasu Hospital, Urayasu-shi, Japan
| | - Toshio Naito
- Department of Research Support Utilizing Bioresource Bank, Graduate School of Medicine, Juntendo University, Tokyo, Japan
| | - Kazuhisa Takahashi
- Department of Research Support Utilizing Bioresource Bank, Graduate School of Medicine, Juntendo University, Tokyo, Japan
| | - Koichi Hattori
- Center for Genome and Regenerative Medicine, Juntendo University, Graduate School of Medicine, Tokyo, Japan
- Department of Hematology/Oncology, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Beate Heissig
- Department of Research Support Utilizing Bioresource Bank, Graduate School of Medicine, Juntendo University, Tokyo, Japan
| |
Collapse
|
32
|
Hou L, Su K, He T, Zhao J, Li Y. Using XGBoost for Predicting In-Stent Restenosis Post-DES Implantation: Role of Lymphocyte-to-Monocyte Ratio and Residual Cholesterol. Int J Gen Med 2024; 17:3443-3452. [PMID: 39139709 PMCID: PMC11321347 DOI: 10.2147/ijgm.s477053] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2024] [Accepted: 08/02/2024] [Indexed: 08/15/2024] Open
Abstract
Objective This study aims to investigate their correlation and predictive utility for in-stent restenosis (ISR) in patients with acute coronary syndrome (ACS) following percutaneous coronary intervention (PCI). Methods We collected medical records of 668 patients who underwent PCI treatment from January 2022 to December 2022. Based on follow-up results (ISR defined as luminal narrowing ≥ 50% on angiography), all participants were divided into ISR and non-ISR groups. The XGBoost machine learning (ML) model was employed to identify the optimal predictive variables from a set of 31 variables. Discriminatory ability was evaluated using the area under the receiver operating characteristic (ROC) curve (AUC), while calibration and performance of the prediction models were assessed using the Hosmer-Lemeshow (HL) test and calibration plots. Clinical utility of each model was evaluated using decision curve analysis (DCA). Results In the XGBoost importance ranking of predictive factors, LMR and RC ranked first and fourth, respectively. The AUC of the entire XGBoost ML model was 0.8098, whereas the model using traditional stepwise backward regression, comprising five predictive factors, had an AUC of 0.706. The XGBoost model showed superior predictive performance with a higher AUC, indicating better discrimination and predictive accuracy for ISR compared to traditional methods. Conclusion LMR and RC are identified as cost-effective and reliable biomarkers for predicting ISR risk in ACS patients following drug-eluting stent (DES) implantation. LMR and RC represent cost-effective and reliable biomarkers for predicting ISR risk in ACS patients following drug-eluting stent implantation. Enhances the accuracy and clinical utility of ISR prediction models, offering clinicians a robust tool for risk stratification and personalized patient management.
Collapse
Affiliation(s)
- Ling Hou
- Central Hospital of Tujia and Miao Autonomous Prefecture, Hubei University of Medicine, Shiyan, Hubei Province, People’s Republic of China
| | - Ke Su
- Cardiovascular Disease Center, Central Hospital of Tujia and Miao Autonomous Prefecture, Hubei University of Medicine, Enshi, Hubei Province, People’s Republic of China
| | - Ting He
- Central Hospital of Tujia and Miao Autonomous Prefecture, Hubei University of Medicine, Shiyan, Hubei Province, People’s Republic of China
| | - Jinbo Zhao
- Cardiovascular Disease Center, Central Hospital of Tujia and Miao Autonomous Prefecture, Hubei University of Medicine, Enshi, Hubei Province, People’s Republic of China
| | - Yuanhong Li
- Cardiovascular Disease Center, Central Hospital of Tujia and Miao Autonomous Prefecture, Hubei University of Medicine, Enshi, Hubei Province, People’s Republic of China
| |
Collapse
|
33
|
Murphy J, Zierotin A, Mongan D, Healy C, Susai SR, O'Donoghue B, Clarke M, O'Connor K, Cannon M, Cotter DR. Associations between soluble urokinase plasminogen activator receptor (suPAR) concentration and psychiatric disorders - A systematic review and meta-analysis. Brain Behav Immun 2024; 120:327-338. [PMID: 38857636 DOI: 10.1016/j.bbi.2024.06.003] [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: 11/30/2023] [Revised: 04/29/2024] [Accepted: 06/05/2024] [Indexed: 06/12/2024] Open
Abstract
BACKGROUND There is some evidence of an association between inflammation in the pathogenesis of mental disorders. Soluble urokinase plasminogen activator receptor (suPAR) is a biomarker of chronic inflammation, which provides a more stable index of systemic inflammation than more widely used biomarkers. This review aims to synthesise studies that measured suPAR concentrations in individuals with a psychiatric disorder, to determine if these concentrations are altered in comparison to healthy participants. METHOD Comprehensive literature searches from inception to October 2023 were conducted of five relevant databases (PubMed, Web of Science, Embase, Scopus, APA PsychInfo). Random-effects meta-analyses were performed to compare the standardised mean difference of blood suPAR levels (i.e. plasma or serum) for individuals with any psychiatric disorder relative to controls. Separate meta-analyses of suPAR levels were conducted for individuals with schizophrenia or other psychotic disorder and depressive disorder. Risk of bias was assessed using the Newcastle Ottawa Scale. Post-hoc sensitivity analyses included excluding studies at high risk of bias, and analyses of studies that measured suPAR concentrations either in serum or in plasma separately. RESULTS The literature search identified 149 records. Ten full-text studies were screened for eligibility and 9 studies were included for review. Primary analyses revealed no significant difference in suPAR levels between individuals with any psychiatric disorder compared to controls (k = 7, SMD = 0.42, 95 % CI [-0.20, 1.04]). However, those with depressive disorder had elevated suPAR levels relative to controls (k = 3, SMD = 0.61, 95 % CI [0.34, 0.87]). Similarly, secondary analyses showed no evidence of a significant difference in suPAR levels in individuals with any psychiatric disorder when studies at high risk of bias were excluded (k = 6, SMD = 0.54, 95 % CI [-0.14, 1.22]), but elevated suPAR concentrations for those with schizophrenia or other psychotic disorder were found (k = 3, SMD = 0.98, 95 % CI [0.39, 1.58]). Furthermore, studies that analysed plasma suPAR concentrations found elevated plasma suPAR levels in individuals with any psychiatric disorder relative to controls (k = 5, SMD = 0.84, 95 % CI [0.38, 1.29]), while studies measuring serum suPAR levels in any psychiatric disorder did not find a difference (k = 2, SMD = -0.61, 95 % CI [-1.27, 0.04]). For plasma, elevated suPAR concentrations were also identified for those with schizophrenia or other psychotic disorder (k = 3, SMD = 0.98, 95 % CI [0.39, 1.58]). DISCUSSION When studies measuring either only serum or only plasma suPAR were considered, no significant difference in suPAR levels were observed between psychiatric disorder groups, although significantly elevated suPAR levels were detected in those with moderate to severe depressive disorder. However, plasma suPAR levels were significantly elevated in those with any psychiatric disorder relative to controls, while no difference in serum samples was found. A similar finding was reported for schizophrenia or other psychotic disorder. The plasma findings suggest that chronic inflammatory dysregulation may contribute to the pathology of schizophrenia and depressive disorder. Future longitudinal studies are required to fully elucidate the role of this marker in the psychopathology of these disorders.
Collapse
Affiliation(s)
- Jennifer Murphy
- Department of Psychiatry, Royal College of Surgeons in Ireland, Dublin, Ireland.
| | - Anna Zierotin
- Department of Psychiatry, University College Dublin, Ireland
| | - David Mongan
- Department of Psychiatry, Royal College of Surgeons in Ireland, Dublin, Ireland; Centre for Public Health, Queen's University Belfast, United Kingdom
| | - Colm Healy
- Department of Psychiatry, Royal College of Surgeons in Ireland, Dublin, Ireland; School of Medicine, University College Dublin, Dublin, Ireland
| | - Subash R Susai
- Department of Psychiatry, Royal College of Surgeons in Ireland, Dublin, Ireland; SFI FutureNeuro Research Centre, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Brian O'Donoghue
- Department of Psychiatry, University College Dublin, Ireland; Department of Psychiatry, St Vincent's University Hospital, Elm Park, Dublin 4, Ireland
| | - Mary Clarke
- Department of Psychiatry, University College Dublin, Ireland; DETECT Early Intervention for Psychosis Service, Blackrock, Co. Dublin, Ireland
| | - Karen O'Connor
- RISE, Early Intervention in Psychosis Team, South Lee Mental Health Services, Cork, Ireland; Department of Psychiatry and Neurobehavioral Science, University College Cork, Cork, Ireland
| | - Mary Cannon
- Department of Psychiatry, Royal College of Surgeons in Ireland, Dublin, Ireland; SFI FutureNeuro Research Centre, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - David R Cotter
- Department of Psychiatry, Royal College of Surgeons in Ireland, Dublin, Ireland; SFI FutureNeuro Research Centre, Royal College of Surgeons in Ireland, Dublin, Ireland
| |
Collapse
|
34
|
Campanelli R, Carolei A, Catarsi P, Abbà C, Boveri E, Paulli M, Gentile R, Morosini M, Albertini R, Mantovani S, Massa M, Barosi G, Rosti V. Circulating Polymorphonuclear Myeloid-Derived Suppressor Cells (PMN-MDSCs) Have a Biological Role in Patients with Primary Myelofibrosis. Cancers (Basel) 2024; 16:2556. [PMID: 39061196 PMCID: PMC11275082 DOI: 10.3390/cancers16142556] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2024] [Revised: 06/18/2024] [Accepted: 07/10/2024] [Indexed: 07/28/2024] Open
Abstract
Primary myelofibrosis (PMF) is a myeloproliferative neoplasm characterized by a chronic inflammatory state that plays a relevant role in the disease pathogenesis (as proven by high levels of inflammatory cytokines with prognostic significance and by a persistent oxidative stress) and by extensive neoangiogenesis in bone marrow (BM) and spleen. Myeloid-derived suppressor cells (MDSCs) are immature cells that expand in patients with cancer, sepsis or chronic inflammation, favoring tumor onset and progression mainly through the decrease in immune surveillance and the promotion of neoangiogenesis. In this paper, we evaluated the presence of circulating MDSCs in PMF patients, the plasmatic factors involved in their mobilization/expansion and the correlations with laboratory, genetic and clinical parameters. The data indicated that MDSCs could have a relevant role in PMF as a new pathogenic mechanism contributing to explaining the phenotypic diversity observed during the clinical course of the disease, or a potential new target for personalized treatment.
Collapse
Affiliation(s)
- Rita Campanelli
- Center for the Study of Myelofibrosis, Fondazione IRCCS Policlinico San Matteo, 27100 Pavia, Italy; (A.C.)
| | - Adriana Carolei
- Center for the Study of Myelofibrosis, Fondazione IRCCS Policlinico San Matteo, 27100 Pavia, Italy; (A.C.)
| | - Paolo Catarsi
- Center for the Study of Myelofibrosis, Fondazione IRCCS Policlinico San Matteo, 27100 Pavia, Italy; (A.C.)
| | - Carlotta Abbà
- General Medicine 2-Center for Systemic Amyloidosis and High-Complexity Diseases, Fondazione IRCCS Policlinico San Matteo, 27100 Pavia, Italy
| | - Emanuela Boveri
- Unit of Anatomic Pathology, Fondazione IRCCS Policlinico San Matteo, 27100 Pavia, Italy
| | - Marco Paulli
- Unit of Anatomic Pathology, Fondazione IRCCS Policlinico San Matteo, 27100 Pavia, Italy
- Department of Molecular Medicine, Unit of Anatomic Pathology, University of Pavia, 27100 Pavia, Italy
| | - Raffaele Gentile
- Chemical and Clinics Laboratory, Fondazione IRCCS Policlinico San Matteo, 27100 Pavia, Italy
| | - Monica Morosini
- Chemical and Clinics Laboratory, Fondazione IRCCS Policlinico San Matteo, 27100 Pavia, Italy
| | - Riccardo Albertini
- Chemical and Clinics Laboratory, Fondazione IRCCS Policlinico San Matteo, 27100 Pavia, Italy
| | - Stefania Mantovani
- Research Department, Division of Clinical Immunology—Infectious Diseases, Fondazione IRCCS Policlinico San Matteo, 27100 Pavia, Italy
| | - Margherita Massa
- General Medicine 2-Center for Systemic Amyloidosis and High-Complexity Diseases, Fondazione IRCCS Policlinico San Matteo, 27100 Pavia, Italy
| | - Giovanni Barosi
- Center for the Study of Myelofibrosis, Fondazione IRCCS Policlinico San Matteo, 27100 Pavia, Italy; (A.C.)
| | - Vittorio Rosti
- Center for the Study of Myelofibrosis, Fondazione IRCCS Policlinico San Matteo, 27100 Pavia, Italy; (A.C.)
| |
Collapse
|
35
|
Akdeniz YS, Özkan S. New markers in chronic obstructive pulmonary disease. Adv Clin Chem 2024; 123:1-63. [PMID: 39181619 DOI: 10.1016/bs.acc.2024.06.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/27/2024]
Abstract
Chronic obstructive pulmonary disease (COPD), a global healthcare and socioeconomic burden, is a multifaceted respiratory disorder that results in substantial decline in health status and life quality. Acute exacerbations of the disease contribute significantly to increased morbidity and mortality. Consequently, the identification of reliable and effective biomarkers for rapid diagnosis, prediction, and prognosis of exacerbations is imperative. In addition, biomarkers play a crucial role in monitoring responses to therapeutic interventions and exploring innovative treatment strategies. Although established markers such as CRP, fibrinogen and neutrophil count are routinely used, a universal marker is lacking. Fortunately, an increasing number of studies based on next generation analytics have explored potential biomarkers in COPD. Here we review those advances and the need for standardized validation studies in the appropriate clinical setting.
Collapse
Affiliation(s)
- Yonca Senem Akdeniz
- Department of Emergency Medicine, Cerrahpaşa Faculty of Medicine, İstanbul University-Cerrahpaşa, İstanbul, Türkiye.
| | - Seda Özkan
- Department of Emergency Medicine, Cerrahpaşa Faculty of Medicine, İstanbul University-Cerrahpaşa, İstanbul, Türkiye
| |
Collapse
|
36
|
Nusshag C, Theobald V, Wortmann M, Kaimann P, Dietrich M, Gruneberg D, Tourelle K, von der Forst M, Weigand MA, Bischoff MS, Böckler D, Schmitt FCF. Biomarker-guided detection of acute kidney injury in abdominal aortic surgery: the new and the old. Front Med (Lausanne) 2024; 11:1386018. [PMID: 39021823 PMCID: PMC11251966 DOI: 10.3389/fmed.2024.1386018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2024] [Accepted: 06/17/2024] [Indexed: 07/20/2024] Open
Abstract
Introduction Acute kidney injury (AKI) is a common complication in patients undergoing major vascular surgery. Despite significant research efforts in this area, the incidence of AKI remains high, posing a significant challenge to healthcare systems, especially in situations where resources are limited. Early prediction of AKI severity and individualized postoperative care is therefore essential. Methods The primary objective of this exploratory study was to assess the diagnostic value of urine cell-cycle arrest biomarkers [(TIMP-2) × (IGFBP7)] and soluble urokinase plasminogen activator receptor (suPAR) for predicting moderate or severe AKI within 24 h after open aortic surgery, and compared to routine kidney biomarkers. Seventy-five patients undergoing elective aortic surgery were included. Clinical parameters, urine and blood samples were collected preoperatively, immediately postoperatively, and 24 h later. AKI was defined using KDIGO criteria. Individual and combined diagnostic performance of biomarkers were evaluated. Results Of the 75 patients, 61% developed AKI, of which 28% developed moderate or severe AKI within 24 h of surgery. Baseline demographics, comorbidities and kidney parameters did not differ between patients with moderate or severe AKI (AKI II/III) and none or mild AKI (AKI 0/I), except for higher preoperative suPAR levels in later AKI II/III patients. Urine osmolality, Cystatin C and serum creatinine had the highest predictive power for AKI II/III with AUCs of 0.75-0.72. (TIMP-2) × (IGFBP7), and neither (TIMP-2) × (IGFBP7) nor suPAR individually showed superior diagnostic value. Combining CysC or SCr with urine osmolality and 6 h urine output gave the best performance with AUCs of 0.86 (95% CI, 0.74-0.96) and 0.85 (95% CI, 0.75-0.95) respectively. Conclusion Our study suggests that routine parameters like urine osmolality, CysC, SCr and 6 h urine output perform best in predicting postoperative AKI after aortic surgery compared to the new biomarkers (TIMP-2) × (IGFBP7) and suPAR. Combining biomarkers, particularly CysC or SCr with urine output, urine osmolality, may enhance diagnostic accuracy. Further validation in larger cohorts and clinical settings is warranted to establish their clinical utility.
Collapse
Affiliation(s)
- Christian Nusshag
- Department of Nephrology, Medical Faculty Heidelberg, Heidelberg University, Heidelberg, Germany
| | - Vivienne Theobald
- Department of Anesthesiology, Medical Faculty Heidelberg, Heidelberg University, Heidelberg, Germany
| | - Markus Wortmann
- Department of Vascular, Endovascular Surgery and Transplant Surgery, Stuttgart Hospital, Stuttgart, Germany
| | - Philipp Kaimann
- Department of Anesthesiology, Medical Faculty Heidelberg, Heidelberg University, Heidelberg, Germany
| | - Maximilian Dietrich
- Department of Anesthesiology, Medical Faculty Heidelberg, Heidelberg University, Heidelberg, Germany
| | - Daniel Gruneberg
- Department of Anesthesiology, Medical Faculty Heidelberg, Heidelberg University, Heidelberg, Germany
| | - Kevin Tourelle
- Department of Anesthesiology, Medical Faculty Heidelberg, Heidelberg University, Heidelberg, Germany
| | - Maik von der Forst
- Department of Anesthesiology, Medical Faculty Heidelberg, Heidelberg University, Heidelberg, Germany
| | - Markus A. Weigand
- Department of Anesthesiology, Medical Faculty Heidelberg, Heidelberg University, Heidelberg, Germany
| | - Moritz S. Bischoff
- Department of Vascular and Endovascular Surgery, University Hospital Heidelberg, Heidelberg, Germany
| | - Dittmar Böckler
- Department of Vascular and Endovascular Surgery, University Hospital Heidelberg, Heidelberg, Germany
| | - Felix C. F. Schmitt
- Department of Anesthesiology, Medical Faculty Heidelberg, Heidelberg University, Heidelberg, Germany
| |
Collapse
|
37
|
Rehan ST, Imran L, Eqbal F, Khan Z, Nashwan AJ, Asghar MS. Prognostic role of suPAR in acute pancreatitis: A protocol for systematic review. Medicine (Baltimore) 2024; 103:e37064. [PMID: 38941433 PMCID: PMC11466201 DOI: 10.1097/md.0000000000037064] [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: 09/01/2023] [Accepted: 01/04/2024] [Indexed: 06/30/2024] Open
Abstract
BACKGROUND Acute pancreatitis (AP) is a common emergency condition with high morbidity, mortality, and socio-economic impact. Soluble urokinase plasminogen activator receptor (suPAR) is a potential biomarker for AP prognosis. This study systematically reviews the literature on suPAR's prognostic roles in assessing AP severity, organ failure, mortality, and other pathological markers. METHODS A comprehensive search of 5 databases up to March 19, 2023, was conducted, selecting cohort studies that examined suPAR's relationship with AP outcomes. Outcome variables included AP severity, organ failure, mortality, hospital stay length, and suPAR's association with other inflammatory markers. Our paper has been registered on Prospero (ID: CRD42023410628). RESULTS Nine prospective observational studies with 1033 AP patients were included. Seven of eight studies found suPAR significantly elevated in severe acute pancreatitis (P < .05). Four studies showed suPAR effectively predicted organ failure risk, and 4 studies concluded suPAR significantly predicted mortality (P < .05). The review had no high-risk studies, enhancing credibility. CONCLUSION suPAR is a valuable prognostic marker in AP, significantly predicting severity, organ failure, hospital stay length, and mortality. Further large-scale studies are needed to explore suPAR's role in other clinical outcomes related to AP disease course, to establish it as a mainstay of AP prognosis.
Collapse
Affiliation(s)
| | - Laiba Imran
- Department of Medicine, Dow University of Health Science, Karachi, Pakistan
| | - Farea Eqbal
- Department of Medicine, Dow University of Health Science, Karachi, Pakistan
| | - Zayeema Khan
- Department of Medicine, Dow University of Health Science, Karachi, Pakistan
| | | | | |
Collapse
|
38
|
Khan H, Abu-Raisi M, Feasson M, Shaikh F, Saposnik G, Mamdani M, Qadura M. Current Prognostic Biomarkers for Abdominal Aortic Aneurysm: A Comprehensive Scoping Review of the Literature. Biomolecules 2024; 14:661. [PMID: 38927064 PMCID: PMC11201473 DOI: 10.3390/biom14060661] [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: 05/02/2024] [Revised: 05/29/2024] [Accepted: 06/03/2024] [Indexed: 06/28/2024] Open
Abstract
Abdominal aortic aneurysm (AAA) is a progressive dilatation of the aorta that can lead to aortic rupture. The pathophysiology of the disease is not well characterized but is known to be caused by the general breakdown of the extracellular matrix within the aortic wall. In this comprehensive literature review, all current research on proteins that have been investigated for their potential prognostic capabilities in patients with AAA was included. A total of 45 proteins were found to be potential prognostic biomarkers for AAA, predicting incidence of AAA, AAA rupture, AAA growth, endoleak, and post-surgical mortality. The 45 proteins fell into the following seven general categories based on their primary function: (1) cardiovascular health, (2) hemostasis, (3) transport proteins, (4) inflammation and immunity, (5) kidney function, (6) cellular structure, (7) and hormones and growth factors. This is the most up-to-date literature review on current prognostic markers for AAA and their functions. This review outlines the wide pathophysiological processes that are implicated in AAA disease progression.
Collapse
Affiliation(s)
- Hamzah Khan
- Division of Vascular Surgery, St. Michael’s Hospital, Toronto, ON M5B 1W8, Canada
- Li Ka Shing Knowledge Institute, St Michael’s Hospital, Unity Health Toronto, Toronto, ON M5B 1W8, Canada
| | - Mohamed Abu-Raisi
- Division of Vascular Surgery, St. Michael’s Hospital, Toronto, ON M5B 1W8, Canada
- Li Ka Shing Knowledge Institute, St Michael’s Hospital, Unity Health Toronto, Toronto, ON M5B 1W8, Canada
| | - Manon Feasson
- Li Ka Shing Knowledge Institute, St Michael’s Hospital, Unity Health Toronto, Toronto, ON M5B 1W8, Canada
| | - Farah Shaikh
- Division of Vascular Surgery, St. Michael’s Hospital, Toronto, ON M5B 1W8, Canada
- Li Ka Shing Knowledge Institute, St Michael’s Hospital, Unity Health Toronto, Toronto, ON M5B 1W8, Canada
| | - Gustavo Saposnik
- Li Ka Shing Knowledge Institute, St Michael’s Hospital, Unity Health Toronto, Toronto, ON M5B 1W8, Canada
- Division of Neurology, Department of Medicine, University of Toronto, Toronto, ON M5S 1A1, Canada
| | - Muhammad Mamdani
- Li Ka Shing Knowledge Institute, St Michael’s Hospital, Unity Health Toronto, Toronto, ON M5B 1W8, Canada
| | - Mohammad Qadura
- Division of Vascular Surgery, St. Michael’s Hospital, Toronto, ON M5B 1W8, Canada
- Li Ka Shing Knowledge Institute, St Michael’s Hospital, Unity Health Toronto, Toronto, ON M5B 1W8, Canada
- Department of Surgery, University of Toronto, Toronto, ON M5T 1P5, Canada
| |
Collapse
|
39
|
Houlind MB, Andersen O, Iversen E. Cystatin C to Creatinine Ratio and Measured GFR in Hospitalized Older Adults. Am J Kidney Dis 2024; 83:834-835. [PMID: 38224732 DOI: 10.1053/j.ajkd.2023.11.010] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2023] [Revised: 11/06/2023] [Accepted: 11/20/2023] [Indexed: 01/17/2024]
Affiliation(s)
- Morten Baltzer Houlind
- Department of Clinical Research, Copenhagen University Hospital Amager and Hvidovre, Hvidovre, Denmark; The Capital Region Pharmacy, Herlev, Denmark; Department of Drug Design and Pharmacology, University of Copenhagen, Copenhagen, Denmark
| | - Ove Andersen
- Department of Clinical Research, Copenhagen University Hospital Amager and Hvidovre, Hvidovre, Denmark; Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark; Emergency Department, Copenhagen University Hospital Amager and Hvidovre, Hvidovre, Denmark
| | - Esben Iversen
- Department of Clinical Research, Copenhagen University Hospital Amager and Hvidovre, Hvidovre, Denmark
| |
Collapse
|
40
|
Hamada M, Varkoly KS, Riyadh O, Beladi R, Munuswamy-Ramanujam G, Rawls A, Wilson-Rawls J, Chen H, McFadden G, Lucas AR. Urokinase-Type Plasminogen Activator Receptor (uPAR) in Inflammation and Disease: A Unique Inflammatory Pathway Activator. Biomedicines 2024; 12:1167. [PMID: 38927374 PMCID: PMC11201033 DOI: 10.3390/biomedicines12061167] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Revised: 04/24/2024] [Accepted: 05/10/2024] [Indexed: 06/28/2024] Open
Abstract
The urokinase-type plasminogen activator receptor (uPAR) is a unique protease binding receptor, now recognized as a key regulator of inflammation. Initially, uPA/uPAR was considered thrombolytic (clot-dissolving); however, recent studies have demonstrated its predominant immunomodulatory functions in inflammation and cancer. The uPA/uPAR complex has a multifaceted central role in both normal physiological and also pathological responses. uPAR is expressed as a glycophosphatidylinositol (GPI)-linked receptor interacting with vitronectin, integrins, G protein-coupled receptors, and growth factor receptors within a large lipid raft. Through protein-to-protein interactions, cell surface uPAR modulates intracellular signaling, altering cellular adhesion and migration. The uPA/uPAR also modifies extracellular activity, activating plasminogen to form plasmin, which breaks down fibrin, dissolving clots and activating matrix metalloproteinases that lyse connective tissue, allowing immune and cancer cell invasion and releasing growth factors. uPAR is now recognized as a biomarker for inflammatory diseases and cancer; uPAR and soluble uPAR fragments (suPAR) are increased in viral sepsis (COVID-19), inflammatory bowel disease, and metastasis. Here, we provide a comprehensive overview of the structure, function, and current studies examining uPAR and suPAR as diagnostic markers and therapeutic targets. Understanding uPAR is central to developing diagnostic markers and the ongoing development of antibody, small-molecule, nanogel, and virus-derived immune-modulating treatments that target uPAR.
Collapse
Affiliation(s)
- Mostafa Hamada
- College of Medicine, Kansas City University, 1750 Independence Ave, Kansas City, MO 64106, USA; (M.H.); (O.R.)
| | - Kyle Steven Varkoly
- Department of Internal Medicine, McLaren Macomb Hospital, Michigan State University College of Human Medicine, 1000 Harrington St., Mt Clemens, MI 48043, USA
| | - Omer Riyadh
- College of Medicine, Kansas City University, 1750 Independence Ave, Kansas City, MO 64106, USA; (M.H.); (O.R.)
| | - Roxana Beladi
- Department of Neurosurgery, Ascension Providence Hospital, Michigan State University College of Human Medicine, 16001 W Nine Mile Rd, Southfield, MI 48075, USA;
| | - Ganesh Munuswamy-Ramanujam
- Molecular Biology and Immunobiology Division, Interdisciplinary Institute of Indian System of Medicine, SRM Institute of Science and Technology, Kattankulathur 603203, India;
| | - Alan Rawls
- School of Life Sciences, Arizona State University, 427 E Tyler Mall, Tempe, AZ 85281, USA; (A.R.); (J.W.-R.)
| | - Jeanne Wilson-Rawls
- School of Life Sciences, Arizona State University, 427 E Tyler Mall, Tempe, AZ 85281, USA; (A.R.); (J.W.-R.)
| | - Hao Chen
- Department of Tumor Center, Lanzhou University Second Hospital, Lanzhou 730030, China;
| | - Grant McFadden
- Center for Personalized Diagnostics, Biodesign Institute, Arizona State University, 727 E Tyler St., Tempe, AZ 85287, USA;
| | - Alexandra R. Lucas
- Center for Personalized Diagnostics, Biodesign Institute, Arizona State University, 727 E Tyler St., Tempe, AZ 85287, USA;
| |
Collapse
|
41
|
Murthy VL, Mosley JD, Perry AS, Jacobs DR, Tanriverdi K, Zhao S, Sawicki KT, Carnethon M, Wilkins JT, Nayor M, Das S, Abel ED, Freedman JE, Clish CB, Shah RV. Metabolic liability for weight gain in early adulthood. Cell Rep Med 2024; 5:101548. [PMID: 38703763 PMCID: PMC11148768 DOI: 10.1016/j.xcrm.2024.101548] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2022] [Revised: 03/27/2023] [Accepted: 04/10/2024] [Indexed: 05/06/2024]
Abstract
While weight gain is associated with a host of chronic illnesses, efforts in obesity have relied on single "snapshots" of body mass index (BMI) to guide genetic and molecular discovery. Here, we study >2,000 young adults with metabolomics and proteomics to identify a metabolic liability to weight gain in early adulthood. Using longitudinal regression and penalized regression, we identify a metabolic signature for weight liability, associated with a 2.6% (2.0%-3.2%, p = 7.5 × 10-19) gain in BMI over ≈20 years per SD higher score, after comprehensive adjustment. Identified molecules specified mechanisms of weight gain, including hunger and appetite regulation, energy expenditure, gut microbial metabolism, and host interaction with external exposure. Integration of longitudinal and concurrent measures in regression with Mendelian randomization highlights the complexity of metabolic regulation of weight gain, suggesting caution in interpretation of epidemiologic or genetic effect estimates traditionally used in metabolic research.
Collapse
Affiliation(s)
- Venkatesh L Murthy
- Division of Cardiovascular Medicine, Department of Medicine, University of Michigan, Ann Arbor, MI, USA.
| | - Jonathan D Mosley
- Vanderbilt Translational and Clinical Cardiovascular Research Center, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Andrew S Perry
- Vanderbilt Translational and Clinical Cardiovascular Research Center, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - David R Jacobs
- Division of Epidemiology and Community Health, School of Public Health, University of Minnesota, Minneapolis, MN, USA
| | - Kahraman Tanriverdi
- Vanderbilt Translational and Clinical Cardiovascular Research Center, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Shilin Zhao
- Vanderbilt Translational and Clinical Cardiovascular Research Center, Vanderbilt University School of Medicine, Nashville, TN, USA
| | | | | | | | - Matthew Nayor
- Section of Cardiovascular Medicine, Department of Medicine, Boston University School of Medicine, Boston, MA, USA
| | - Saumya Das
- Cardiology Division, Massachusetts General Hospital, Boston, MA, USA
| | - E Dale Abel
- Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
| | - Jane E Freedman
- Vanderbilt Translational and Clinical Cardiovascular Research Center, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Clary B Clish
- Broad Institute of Harvard and MIT, Cambridge, MA, USA
| | - Ravi V Shah
- Vanderbilt Translational and Clinical Cardiovascular Research Center, Vanderbilt University School of Medicine, Nashville, TN, USA.
| |
Collapse
|
42
|
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
|
43
|
Montecillo J, Pirker T, Pemberton C, Chew-Harris J. suPAR in cardiovascular disease. Adv Clin Chem 2024; 121:89-131. [PMID: 38797545 DOI: 10.1016/bs.acc.2024.04.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/29/2024]
Abstract
Soluble urokinase plasminogen activator receptor (suPAR), the soluble counterpart of urokinase plasminogen activator receptor, is found in the circulation at various levels. suPAR and its parent molecule, cell surface uPAR, exhibit similar structure and extracellular functional roles facilitating fibrinolysis, cellular adhesion, and migration. Studies have assessed the correlation between suPAR in cardiovascular disease (CVD). It is postulated that suPAR may serve as an indicator of inflammatory activation and burden during CVD progression. Increased suPAR independently predicts poorer outcomes in acute coronary syndromes, in heart failure, as well as in coronary artery disease and atherosclerosis. To guide translation into clinical utization, suPAR has been assessed in numerous CVD settings for improved risk discrimination independently or in association with established traditional risk factors. Whilst the involvement of suPAR has been explored in other diseases such as kidney diseases and cancer, there is only emerging evidence of suPAR's mechanistic involvement in cardiovascular disease. In this review, we provide a background into suPAR and its potential role as a biomarker in CVD.
Collapse
Affiliation(s)
- Jaya Montecillo
- Christchurch Heart Institute, University of Otago, Christchurch, New Zealand
| | - Thomas Pirker
- Christchurch Heart Institute, University of Otago, Christchurch, New Zealand
| | | | - Janice Chew-Harris
- Christchurch Heart Institute, University of Otago, Christchurch, New Zealand.
| |
Collapse
|
44
|
Kanno Y. The Roles of Fibrinolytic Factors in Bone Destruction Caused by Inflammation. Cells 2024; 13:516. [PMID: 38534360 PMCID: PMC10968824 DOI: 10.3390/cells13060516] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2024] [Revised: 03/14/2024] [Accepted: 03/15/2024] [Indexed: 03/28/2024] Open
Abstract
Chronic inflammatory diseases, such as rheumatoid arthritis, spondyloarthritis, systemic lupus erythematosus, Crohn's disease, periodontitis, and carcinoma metastasis frequently result in bone destruction. Pro-inflammatory cytokines such as tumor necrosis factor-α (TNF-α), interleukin-1β (IL-1β), IL-6, and IL-17 are known to influence bone loss by promoting the differentiation and activation of osteoclasts. Fibrinolytic factors, such as plasminogen (Plg), plasmin, urokinase-type plasminogen activator (uPA), its receptor (uPAR), tissue-type plasminogen activator (tPA), α2-antiplasmin (α2AP), and plasminogen activator inhibitor-1 (PAI-1) are expressed in osteoclasts and osteoblasts and are considered essential in maintaining bone homeostasis by regulating the functions of both osteoclasts and osteoblasts. Additionally, fibrinolytic factors are associated with the regulation of inflammation and the immune system. This review explores the roles of fibrinolytic factors in bone destruction caused by inflammation.
Collapse
Affiliation(s)
- Yosuke Kanno
- Department of Molecular Pathology, Faculty of Pharmaceutical Science, Doshisha Women's College of Liberal Arts, 97-1 Kodo Kyotanabe, Kyoto 610-0395, Japan
| |
Collapse
|
45
|
Ehrlich KB, Celia-Sanchez ML, Yu T, Heard-Garris N, Chen E, Miller GE, Brody GH. Exposure to parental depression in adolescence and proinflammatory phenotypes 20 years later. Brain Behav Immun 2024; 117:196-203. [PMID: 38242368 PMCID: PMC10932843 DOI: 10.1016/j.bbi.2024.01.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/19/2023] [Revised: 01/11/2024] [Accepted: 01/14/2024] [Indexed: 01/21/2024] Open
Abstract
Although the biological embedding model of adversity proposes that stressful experiences in childhood create a durable proinflammatory phenotype in immune cells, research to date has relied on study designs that limit our ability to make conclusions about whether the phenotype is long-lasting. The present study leverages an ongoing 20-year investigation of African American youth to test research questions about the extent to which stressors measured in childhood forecast a proinflammatory phenotype in adulthood, as indicated by exaggerated cytokine responses to bacterial stimuli, monocyte insensitivity to inhibitory signals from hydrocortisone, and low-grade inflammation. Parents reported on their depressive symptoms and unsupportive parenting tendencies across youths' adolescence. At age 31, youth participants (now adults) completed a fasting blood draw. Samples were incubated with lipopolysaccharide and doses of hydrocortisone to evaluate proinflammatory processes. Additionally, blood samples were tested for indicators of low-grade inflammation, including IL-6, IL-8, IL-10, and TNF-α, and soluble urokinase plasminogen activator receptor. Analyses revealed that parental depression across youths' adolescence prospectively predicted indicators of proinflammatory phenotypes at age 31. Follow-up analyses suggested that unsupportive parenting mediated these associations. These findings suggest that exposure to parental depression in adolescence leaves an imprint on inflammatory activity that can be observed 20 years later.
Collapse
Affiliation(s)
- Katherine B Ehrlich
- Department of Psychology, University of Georgia, Athens, GA, USA; Center for Family Research, University of Georgia, Athens, GA, USA.
| | | | - Tianyi Yu
- Center for Family Research, University of Georgia, Athens, GA, USA
| | - Nia Heard-Garris
- Department of Pediatrics, Northwestern University Feinberg School of Medicine, Chicago, IL, USA; Institute for Policy Research, Northwestern University, Evanston, IL, USA; Department of Psychology, Northwestern University, Evanston, IL, USA
| | - Edith Chen
- Institute for Policy Research, Northwestern University, Evanston, IL, USA; Department of Psychology, Northwestern University, Evanston, IL, USA
| | - Gregory E Miller
- Institute for Policy Research, Northwestern University, Evanston, IL, USA; Department of Psychology, Northwestern University, Evanston, IL, USA
| | - Gene H Brody
- Center for Family Research, University of Georgia, Athens, GA, USA
| |
Collapse
|
46
|
Cano I, Blaker E, Hartnell D, Farbos A, Moore KA, Cobb A, Santos EM, van Aerle R. Transcriptomic Responses to Koi Herpesvirus in Isolated Blood Leukocytes from Infected Common Carp. Viruses 2024; 16:380. [PMID: 38543746 PMCID: PMC10974277 DOI: 10.3390/v16030380] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Revised: 02/08/2024] [Accepted: 02/23/2024] [Indexed: 04/01/2024] Open
Abstract
Koi herpesvirus (KHV, CyHV-3) causes severe economic losses in carp farms. Its eradication is challenging due to the establishment of latency in blood leukocytes and other tissues. To understand the molecular mechanisms leading to KHV infection in leukocytes, common carp were bath-exposed to KHV at 17 °C. After confirming the presence of viral transcripts in blood leukocytes at ten days post infection, RNA-Seq was performed on peripheral blood leukocytes on the Illumina NovaSeq. KHV infection triggered a robust immune response mediated by pattern recognition receptors, mainly toll-like receptors (tlr2, tlr5, tlr7, and tlr13), urokinase plasminogen activator surface receptor-like, galectin proteins, and lipid mediators such as leukotriene B4 receptor 1. Enriched pathways showed increased mitochondria oxidative phosphorylation and the activation of signalling pathways such as mitogen-activated protein kinases (MAPKs) and vascular endothelial growth factor (VEGF). KHV-infected leukocytes showed low production of reactive oxygen species (ROS) and glutathione metabolism, high iron export and phagocytosis activity, and low autophagy. Macrophage polarization was deduced from the up-regulation of genes such as arginase non-hepatic 1-like, macrophage mannose receptor-1, crem, il-10, and il-13 receptors, while markers for cytotoxic T cells were observed to be down-regulated. Further work is required to characterise these leukocyte subsets and the molecular events leading to KHV latency in blood leukocytes.
Collapse
Affiliation(s)
- Irene Cano
- International Centre of Excellence for Aquatic Animal Health, Cefas Laboratory, Dorset DT4 8UB, UK; (E.B.); (D.H.); (A.C.); (R.v.A.)
- Centre for Sustainable Aquaculture Futures, University of Exeter, Exeter EX2 4TH, UK;
| | - Ellen Blaker
- International Centre of Excellence for Aquatic Animal Health, Cefas Laboratory, Dorset DT4 8UB, UK; (E.B.); (D.H.); (A.C.); (R.v.A.)
| | - David Hartnell
- International Centre of Excellence for Aquatic Animal Health, Cefas Laboratory, Dorset DT4 8UB, UK; (E.B.); (D.H.); (A.C.); (R.v.A.)
| | - Audrey Farbos
- Biosciences, Faculty of Life and Health Sciences, University of Exeter, Exeter EX2 4TH, UK; (A.F.); (K.A.M.)
| | - Karen A. Moore
- Biosciences, Faculty of Life and Health Sciences, University of Exeter, Exeter EX2 4TH, UK; (A.F.); (K.A.M.)
| | - Adele Cobb
- International Centre of Excellence for Aquatic Animal Health, Cefas Laboratory, Dorset DT4 8UB, UK; (E.B.); (D.H.); (A.C.); (R.v.A.)
| | - Eduarda M. Santos
- Centre for Sustainable Aquaculture Futures, University of Exeter, Exeter EX2 4TH, UK;
- Biosciences, Faculty of Life and Health Sciences, University of Exeter, Exeter EX2 4TH, UK; (A.F.); (K.A.M.)
| | - Ronny van Aerle
- International Centre of Excellence for Aquatic Animal Health, Cefas Laboratory, Dorset DT4 8UB, UK; (E.B.); (D.H.); (A.C.); (R.v.A.)
- Centre for Sustainable Aquaculture Futures, University of Exeter, Exeter EX2 4TH, UK;
| |
Collapse
|
47
|
Ritter A, Lötterle L, Han J, Kalbitz M, Henrich D, Marzi I, Leppik L, Weber B. Evaluation of New Cardiac Damage Biomarkers in Polytrauma: GDF-15, HFABP and uPAR for Predicting Patient Outcomes. J Clin Med 2024; 13:961. [PMID: 38398274 PMCID: PMC10888743 DOI: 10.3390/jcm13040961] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2023] [Revised: 01/29/2024] [Accepted: 02/05/2024] [Indexed: 02/25/2024] Open
Abstract
Background: Polytrauma is one of the leading mortality factors in younger patients, and in particular, the presence of cardiac damage correlates with a poor prognosis. Currently, troponin T is the gold standard, although troponin is limited as a biomarker. Therefore, there is a need for new biomarkers of cardiac damage early after trauma. Methods: Polytraumatized patients (ISS ≥ 16) were divided into two groups: those with cardiac damage (troponin T > 50 pg/mL, n = 37) and those without cardiac damage (troponin T < 12 pg/mL, n = 32) on admission to the hospital. Patients' plasma was collected in the emergency room 24 h after trauma, and plasma from healthy volunteers (n = 10) was sampled. The plasma was analyzed for the expression of HFABP, GDF-15 and uPAR proteins, as well as miR-21, miR-29, miR-34, miR-122, miR-125b, miR-133, miR-194, miR-204, and miR-155. Results were correlated with patients' outcomes. Results: HFABP, uPAR, and GDF-15 were increased in polytraumatized patients with cardiac damage (p < 0.001) with a need for catecholamines. HFABP was increased in non-survivors. Analysis of systemic miRNA concentrations showed a significant increase in miR-133 (p < 0.01) and miR-21 (p < 0.05) in patients with cardiac damage. Conclusion: All tested plasma proteins, miR-133, and miR-21 were found to reflect the cardiac damage in polytrauma patients. GDF-15 and HFABP were shown to strongly correlate with patients' outcomes.
Collapse
Affiliation(s)
- Aileen Ritter
- Department of Trauma-, Hand- and Reconstructive Surgery, University Hospital Frankfurt, Goethe-University, 60596 Frankfurt am Main, Germany; (L.L.); (J.H.); (D.H.); (I.M.); (L.L.); (B.W.)
| | - Lorenz Lötterle
- Department of Trauma-, Hand- and Reconstructive Surgery, University Hospital Frankfurt, Goethe-University, 60596 Frankfurt am Main, Germany; (L.L.); (J.H.); (D.H.); (I.M.); (L.L.); (B.W.)
| | - Jiaoyan Han
- Department of Trauma-, Hand- and Reconstructive Surgery, University Hospital Frankfurt, Goethe-University, 60596 Frankfurt am Main, Germany; (L.L.); (J.H.); (D.H.); (I.M.); (L.L.); (B.W.)
| | - Miriam Kalbitz
- Department of Trauma and Orthopedic Surgery, University Hospital Erlangen, Friedrich-Alexander-University Erlangen-Nuremberg, 91054 Erlangen, Germany;
| | - Dirk Henrich
- Department of Trauma-, Hand- and Reconstructive Surgery, University Hospital Frankfurt, Goethe-University, 60596 Frankfurt am Main, Germany; (L.L.); (J.H.); (D.H.); (I.M.); (L.L.); (B.W.)
| | - Ingo Marzi
- Department of Trauma-, Hand- and Reconstructive Surgery, University Hospital Frankfurt, Goethe-University, 60596 Frankfurt am Main, Germany; (L.L.); (J.H.); (D.H.); (I.M.); (L.L.); (B.W.)
| | - Liudmila Leppik
- Department of Trauma-, Hand- and Reconstructive Surgery, University Hospital Frankfurt, Goethe-University, 60596 Frankfurt am Main, Germany; (L.L.); (J.H.); (D.H.); (I.M.); (L.L.); (B.W.)
| | - Birte Weber
- Department of Trauma-, Hand- and Reconstructive Surgery, University Hospital Frankfurt, Goethe-University, 60596 Frankfurt am Main, Germany; (L.L.); (J.H.); (D.H.); (I.M.); (L.L.); (B.W.)
| |
Collapse
|
48
|
Adami ME, Kotsaki A, Antonakos N, Giannitsioti E, Chalvatzis S, Saridaki M, Avgoustou C, Akinosoglou K, Dakou K, Damoraki G, Katrini K, Koufargyris P, Lekakis V, Panagaki A, Safarika A, Eugen-Olsen J, Giamarellos-Bourboulis EJ. qSOFA combined with suPAR for early risk detection and guidance of antibiotic treatment in the emergency department: a randomized controlled trial. Crit Care 2024; 28:42. [PMID: 38321472 PMCID: PMC10848347 DOI: 10.1186/s13054-024-04825-2] [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: 12/16/2023] [Accepted: 02/01/2024] [Indexed: 02/08/2024] Open
Abstract
BACKGROUND Sepsis guidelines suggest immediate start of resuscitation for patients with quick Sequential Organ Failure Assessment (qSOFA) 2 or 3. However, the interpretation of qSOFA 1 remains controversial. We investigated whether measurements of soluble urokinase plasminogen activator receptor (suPAR) may improve risk detection when qSOFA is 1. METHODS The study had two parts. At the first part, the combination of suPAR with qSOFA was analyzed in a prospective cohort for early risk detection. At the second part, the double-blind, randomized controlled trial (RCT) SUPERIOR evaluated the efficacy of the suPAR-guided medical intervention. SUPERIOR took place between November 2018 and December 2020. Multivariate stepwise Cox regression was used for the prospective cohort, while univariate and multivariate logistic regression was used for the RCT. Consecutive admissions at the emergency department (ED) with suspected infection, qSOFA 1 and suPAR ≥ 12 ng/mL were allocated to single infusion of placebo or meropenem. The primary endpoint was early deterioration, defined as at least one-point increase of admission Sequential Organ Failure Assessment (SOFA) score the first 24 h. RESULTS Most of the mortality risk was for patients with qSOFA 2 and 3. Taking the hazard ratio (HR) for death of patients with qSOFA = 1 and suPAR < 12 ng/mL as reference, the HR of qSOFA = 1 and suPAR ≥ 12 ng/mL for 28-day mortality was 2.98 (95% CI 2.11-3.96). The prospective RCT was prematurely ended due to pandemia-related ED re-allocations, with 91 patients enrolled: 47 in the placebo and 44 in the meropenem arm. The primary endpoint was met in 40.4% (n = 19) and 15.9% (n = 7), respectively (difference 24.5% [5.9-40.8]; odds ratio 0.14 [0.04-0.50]). One post hoc analysis showed significant median changes of SOFA score after 72 and 96 h equal to 0 and - 1, respectively. CONCLUSIONS Combining qSOFA 1 with the biomarker suPAR improves its prognostic performance for unfavorable outcome and can help decision for earlier treatment. Trial registration EU Clinical Trials Register (EudraCT, 2018-001008-13) and Clinical-Trials.gov (NCT03717350). Registered 24 October 2018.
Collapse
Affiliation(s)
- Maria Evangelia Adami
- Fourth Department of Internal Medicine, Medical School, National and Kapodistrian University of Athens, ATTIKON University General Hospital, 1 Rimini Str, 124 62, Athens, Greece
| | - Antigone Kotsaki
- Fourth Department of Internal Medicine, Medical School, National and Kapodistrian University of Athens, ATTIKON University General Hospital, 1 Rimini Str, 124 62, Athens, Greece
| | - Nikolaos Antonakos
- Fourth Department of Internal Medicine, Medical School, National and Kapodistrian University of Athens, ATTIKON University General Hospital, 1 Rimini Str, 124 62, Athens, Greece
| | - Efthymia Giannitsioti
- Fourth Department of Internal Medicine, Medical School, National and Kapodistrian University of Athens, ATTIKON University General Hospital, 1 Rimini Str, 124 62, Athens, Greece
| | - Stamatios Chalvatzis
- Fourth Department of Internal Medicine, Medical School, National and Kapodistrian University of Athens, ATTIKON University General Hospital, 1 Rimini Str, 124 62, Athens, Greece
| | - Maria Saridaki
- Fourth Department of Internal Medicine, Medical School, National and Kapodistrian University of Athens, ATTIKON University General Hospital, 1 Rimini Str, 124 62, Athens, Greece
| | - Christina Avgoustou
- Fourth Department of Internal Medicine, Medical School, National and Kapodistrian University of Athens, ATTIKON University General Hospital, 1 Rimini Str, 124 62, Athens, Greece
| | | | | | - Georgia Damoraki
- Fourth Department of Internal Medicine, Medical School, National and Kapodistrian University of Athens, ATTIKON University General Hospital, 1 Rimini Str, 124 62, Athens, Greece
| | - Konstantina Katrini
- Fourth Department of Internal Medicine, Medical School, National and Kapodistrian University of Athens, ATTIKON University General Hospital, 1 Rimini Str, 124 62, Athens, Greece
| | - Panagiotis Koufargyris
- Fourth Department of Internal Medicine, Medical School, National and Kapodistrian University of Athens, ATTIKON University General Hospital, 1 Rimini Str, 124 62, Athens, Greece
| | - Vasileios Lekakis
- Fourth Department of Internal Medicine, Medical School, National and Kapodistrian University of Athens, ATTIKON University General Hospital, 1 Rimini Str, 124 62, Athens, Greece
| | - Antonia Panagaki
- Fourth Department of Internal Medicine, Medical School, National and Kapodistrian University of Athens, ATTIKON University General Hospital, 1 Rimini Str, 124 62, Athens, Greece
| | - Asimina Safarika
- Fourth Department of Internal Medicine, Medical School, National and Kapodistrian University of Athens, ATTIKON University General Hospital, 1 Rimini Str, 124 62, Athens, Greece
| | - Jesper Eugen-Olsen
- Department of Clinical Research, Copenhagen University Hospital Amager and Hvidovre, Hvidovre, Denmark
| | - Evangelos J Giamarellos-Bourboulis
- Fourth Department of Internal Medicine, Medical School, National and Kapodistrian University of Athens, ATTIKON University General Hospital, 1 Rimini Str, 124 62, Athens, Greece.
- Hellenic Institute for the Study of Sepsis, Athens, Greece.
| |
Collapse
|
49
|
Kim EY, Dryer SE. Role of Formyl Peptide Receptors and β-Arrestin-1 in suPAR Signal Transduction in Mouse Podocytes: Interactions with αVβ3-Integrin. Cells 2024; 13:172. [PMID: 38247863 PMCID: PMC10814688 DOI: 10.3390/cells13020172] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2023] [Revised: 01/02/2024] [Accepted: 01/11/2024] [Indexed: 01/23/2024] Open
Abstract
The soluble urokinase plasminogen activator receptor (suPAR) has been implicated in a wide range of pathological conditions including primary nephrotic syndromes and acute kidney injuries. suPAR can trigger transduction cascades in podocytes by outside-in activation of αVβ3-integrin, but there is evidence that the functional cell surface response element is actually a complex of different types of receptors, which may also include the receptor for advanced glycation end-products (RAGE) and formyl peptide receptors (FPRs). Here we observed that ROS accumulation and Src activation could be evoked by continuous 24 h exposure to either suPAR or the FPR agonist fMLF. Responses to suPAR and fMLF were completely blocked by either the FPR antagonist WRW4 or by the αV-integrin inhibitor cilengitide. Moreover, endogenous podocyte mouse Fpr1 co-immunoprecipitates with β3-integrin, suggesting that these receptors occur as a complex on the cell surface. suPAR- and fMLF-evoked activation of Src and ROS differed in time course. Thus, robust pertussis toxin (PTX)-sensitive responses were evoked by 60 min exposures to fMLF but not to suPAR. By contrast, responses to 24 h exposures to either suPAR or fMLF were PTX-resistant and were instead abolished by knockdown of β-arrestin-1 (BAR1). FPRs, integrins, and RAGE (along with various Toll-like receptors) can all function as pattern-recognition receptors that respond to "danger signals" associated with infections and tissue injury. The fact that podocytes express such a wide array of pattern-recognition receptors suggests that the glomerular filter is designed to change its function under certain conditions, possibly to facilitate clearance of toxic macromolecules.
Collapse
Affiliation(s)
- Eun Young Kim
- Department of Biology and Biochemistry, University of Houston, Houston, TX 77204, USA;
| | - Stuart E. Dryer
- Department of Biology and Biochemistry, University of Houston, Houston, TX 77204, USA;
- Department of Biomedical Sciences, Tilman J. Fertitta Family College of Medicine, University of Houston, Houston, TX 77204, USA
| |
Collapse
|
50
|
Gulbiniene V, Dumalakiene I, Balciuniene G, Pilypiene I, Narkeviciute I, Novickij V, Vysniauskis G, Ramasauskaite D. Soluble urokinase plasminogen activator receptor in vaginally collected amniotic fluid predicting fetal inflammatory response syndrome: a prospective cohort study. BMC Pregnancy Childbirth 2024; 24:54. [PMID: 38200448 PMCID: PMC10782524 DOI: 10.1186/s12884-023-06221-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2023] [Accepted: 12/21/2023] [Indexed: 01/12/2024] Open
Abstract
BACKGROUND Improving noninvasive antenatal diagnosis of fetal inflammatory response syndrome (FIRS) can assist in the evaluation of prenatal risk and reduce perinatal outcomes. This study aimed to determine whether soluble urokinase-type plasminogen activator receptor (suPAR) in vaginally collected amniotic fluid is significant in identifying FIRS after preterm premature rupture of membranes before 34 weeks of gestation. METHODS This was a prospective cohort study of 114 pregnant women and their newborns after preterm premature rupture of membranes at 22-34+6 weeks of gestation. SuPAR was evaluated using an enzyme-linked immunosorbent assay in vaginally collected amniotic fluid. Patients were classified according to the presence or absence of FIRS. FIRS was defined by umbilical cord blood interleukin-6 level > 11 pg/mL or histological funisitis. The data were analyzed using the R package (R-4.0.5). RESULTS SuPAR was detected in all amniotic fluid samples with a median of 26.23 ng/mL (interquartile range (IQR), 15.19-51.14). The median level of suPAR was higher in the FIRS group than in the non-FIRS group, 32.36 ng/mL (IQR, 17.27-84.16) vs. 20.46 ng/mL (IQR, 11.49-36.63) (P = 0.01), respectively. The presence of histological chorioamnionitis significantly increased the suPAR concentration in the FIRS group (P < 0.001). The areas under the curve for FIRS and FIRS with histological chorioamnionitis were 0.65 and 0.74, respectively, with an optimum cutoff value of 27.60 ng/mL. Controlling for gestational age, the cutoff of suPAR more than 27.60 ng/mL predicted threefold higher odds for FIRS and sixfold higher odds for FIRS with histologic chorioamnionitis. CONCLUSION Soluble urokinase-type plasminogen activator receptor in vaginally obtained amniotic fluid may assist in evaluating prenatal risk of FIRS in patients after preterm premature rupture of membranes before 34 weeks of gestation.
Collapse
Affiliation(s)
| | - Irena Dumalakiene
- Department of Immunology, State Research Institute Center of Innovative Medicine, Vilnius, Lithuania
| | | | | | - Ieva Narkeviciute
- Department of Immunology, State Research Institute Center of Innovative Medicine, Vilnius, Lithuania
| | - Vitalij Novickij
- Department of Immunology, State Research Institute Center of Innovative Medicine, Vilnius, Lithuania
| | - Gintautas Vysniauskis
- Department of Immunology, State Research Institute Center of Innovative Medicine, Vilnius, Lithuania
| | | |
Collapse
|