1
|
Graham EL, D’Isabel S, Lofrano-Porto A, Smith DL. Musculoskeletal, Pulmonary, and Cardiovascular COVID-19 Sequelae in the Context of Firefighter Occupational Health: A Narrative Review. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2024; 21:1383. [PMID: 39457356 PMCID: PMC11508007 DOI: 10.3390/ijerph21101383] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/29/2024] [Revised: 10/13/2024] [Accepted: 10/16/2024] [Indexed: 10/28/2024]
Abstract
For most individuals infected with SARS-CoV-2, the acute illness resolves completely. However, for millions of people, symptoms or sequelae from COVID-19 recur or persist for months to years after infection. Post-COVID-19 sequelae are wide-ranging, often affecting the musculoskeletal, pulmonary, and cardiovascular systems. All who experience post-COVID-19 sequelae face significant challenges navigating home and work life. Occupations such as firefighting, however, are of particular concern given the strenuous nature of a job that relies on a healthy musculoskeletal, pulmonary, and cardiovascular system. Research has documented significant musculoskeletal impairment (including muscle weakness, pain, and fatigue), respiratory dysfunction (including reduced lung function, interstitial disease, and diffusion abnormalities), cardiovascular conditions (including cardiac events, ischemic disease, dysrhythmias, and infectious diseases), and diminished cardiorespiratory fitness that continues for months to years in some individuals. These persistent post-COVID-19 conditions may affect a firefighter's ability to return to work, function at full capacity while at work, and potentially compromise firefighter health and public safety. This review, therefore, explores musculoskeletal, pulmonary, and cardiovascular sequelae post-COVID-19 and the impact of these sequelae on firefighter health and occupational readiness.
Collapse
Affiliation(s)
- Elliot L. Graham
- Integrative Cardiovascular Physiology Laboratory, Colorado State University, Fort Collins, CO 80526, USA
- Intestinal Health Laboratory, Colorado State University, Fort Collins, CO 80526, USA
| | - Susanne D’Isabel
- First Responder Health and Safety Laboratory, Department of Health and Human Physiological Sciences, Skidmore College, Saratoga Springs, NY 12866, USA;
| | - Adriana Lofrano-Porto
- Molecular Pharmacology Laboratory, Health Sciences School, University of Brasilia, Brasilia 70910-900, DF, Brazil
- Endocrine Diseases Clinics, University Hospital of Brasilia, Brasilia 70840-901, DF, Brazil
| | - Denise L. Smith
- First Responder Health and Safety Laboratory, Department of Health and Human Physiological Sciences, Skidmore College, Saratoga Springs, NY 12866, USA;
| |
Collapse
|
2
|
Aljadah M, Khan N, Beyer AM, Chen Y, Blanker A, Widlansky ME. Clinical Implications of COVID-19-Related Endothelial Dysfunction. JACC. ADVANCES 2024; 3:101070. [PMID: 39055276 PMCID: PMC11269277 DOI: 10.1016/j.jacadv.2024.101070] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/10/2024] [Revised: 04/10/2024] [Accepted: 05/24/2024] [Indexed: 07/27/2024]
Abstract
Endothelial dysfunction represents a measurable and early manifestation of vascular disease. Emerging evidence suggests cardiovascular risk remains elevated after COVID-19 infection for at least 12 months, regardless of cardiovascular disease status prior to infection. We review the relationship between the severity of endothelial dysfunction and the severity of acute COVID-19 illness, the degree of impairment following recovery in both those with and without postacute sequalae SARS-CoV-2 infection, and current therapeutic efforts targeting endothelial function in patients following COVID-19 infection. We identify gaps in the literature to highlight specific areas where clinical research efforts hold promise for progress in understanding the connections between endothelial function, COVID-19, and clinical outcomes that will lead to beneficial therapeutics.
Collapse
Affiliation(s)
- Michael Aljadah
- Division of Cardiovascular Medicine, Department of Medicine, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| | - Nabeel Khan
- Division of Cardiovascular Medicine, Department of Medicine, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| | - Andreas M. Beyer
- Division of Cardiovascular Medicine, Department of Medicine, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| | - Yiliang Chen
- Division of Endocrinology and Molecular Medicine, Department of Medicine, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
- Versiti Blood Center of Wisconsin, Milwaukee, Wisconsin, USA
| | - Andrew Blanker
- Division of Cardiovascular Medicine, Department of Medicine, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| | - Michael E. Widlansky
- Division of Cardiovascular Medicine, Department of Medicine, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| |
Collapse
|
3
|
Chidambaram V, Kumar A, Sadaf MI, Lu E, Al’Aref SJ, Tarun T, Galiatsatos P, Gulati M, Blumenthal RS, Leucker TM, Karakousis PC, Mehta JL. COVID-19 in the Initiation and Progression of Atherosclerosis: Pathophysiology During and Beyond the Acute Phase. JACC. ADVANCES 2024; 3:101107. [PMID: 39113913 PMCID: PMC11304887 DOI: 10.1016/j.jacadv.2024.101107] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/28/2023] [Revised: 04/24/2024] [Accepted: 06/01/2024] [Indexed: 08/10/2024]
Abstract
The incidence of atherosclerotic cardiovascular disease is increasing globally, especially in low- and middle-income countries, despite significant efforts to reduce traditional risk factors. Premature subclinical atherosclerosis has been documented in association with several viral infections. The magnitude of the recent COVID-19 pandemic has highlighted the need to understand the association between SARS-CoV-2 and atherosclerosis. This review examines various pathophysiological mechanisms, including endothelial dysfunction, platelet activation, and inflammatory and immune hyperactivation triggered by SARS-CoV-2 infection, with specific attention on their roles in initiating and promoting the progression of atherosclerotic lesions. Additionally, it addresses the various pathogenic mechanisms by which COVID-19 in the post-acute phase may contribute to the development of vascular disease. Understanding the overlap of these syndromes may enable novel therapeutic strategies. We further explore the need for guidelines for closer follow-up for the often-overlooked evidence of atherosclerotic cardiovascular disease among patients with recent COVID-19, particularly those with cardiometabolic risk factors.
Collapse
Affiliation(s)
- Vignesh Chidambaram
- Department of Internal Medicine, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA
| | - Amudha Kumar
- Division of Cardiology, Department of Medicine, Loyola University Medical Center, Maywood, Illinois, USA
| | - Murrium I. Sadaf
- Division of Cardiovascular Medicine, Department of Medicine, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA
| | - Emily Lu
- Division of Infectious Diseases, Department of Medicine, Johns Hopkins School of Medicine, Baltimore, Maryland, USA
| | - Subhi J. Al’Aref
- Division of Cardiovascular Medicine, Department of Medicine, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA
| | - Tushar Tarun
- Division of Cardiovascular Medicine, Department of Medicine, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA
| | - Panagis Galiatsatos
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Johns Hopkins School of Medicine, Baltimore, Maryland, USA
| | - Martha Gulati
- Barbra Streisand Women's Heart Center, Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Roger S. Blumenthal
- Ciccarone Center for the Prevention of Cardiovascular Disease, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Thorsten M. Leucker
- Ciccarone Center for the Prevention of Cardiovascular Disease, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Petros C. Karakousis
- Division of Infectious Diseases, Department of Medicine, Johns Hopkins School of Medicine, Baltimore, Maryland, USA
- Department of International Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
- Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
| | - Jawahar L. Mehta
- Division of Cardiovascular Medicine, Department of Medicine, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA
- Division of Cardiovascular Medicine, Central Arkansas Veterans Healthcare System, Little Rock, Arkansas, USA
| |
Collapse
|
4
|
Falsetti L, Guerrieri E, Zaccone V, Viticchi G, Santini S, Giovenali L, Lagonigro G, Carletti S, Gialluca Palma LE, Tarquinio N, Moroncini G. Cutting-Edge Techniques and Drugs for the Treatment of Pulmonary Embolism: Current Knowledge and Future Perspectives. J Clin Med 2024; 13:1952. [PMID: 38610717 PMCID: PMC11012374 DOI: 10.3390/jcm13071952] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2024] [Revised: 03/11/2024] [Accepted: 03/25/2024] [Indexed: 04/14/2024] Open
Abstract
Pulmonary embolism (PE) is a potentially life-threatening condition requiring prompt diagnosis and treatment. Recent advances have led to the development of newer techniques and drugs aimed at improving PE management, reducing its associated morbidity and mortality and the complications related to anticoagulation. This review provides an overview of the current knowledge and future perspectives on PE treatment. Anticoagulation represents the first-line treatment of hemodynamically stable PE, direct oral anticoagulants being a safe and effective alternative to traditional anticoagulation: these drugs have a rapid onset of action, predictable pharmacokinetics, and low bleeding risk. Systemic fibrinolysis is suggested in patients with cardiac arrest, refractory hypotension, or shock due to PE. With this narrative review, we aim to assess the state of the art of newer techniques and drugs that could radically improve PE management in the near future: (i) mechanical thrombectomy and pulmonary embolectomy are promising techniques reserved to patients with massive PE and contraindications or failure to systemic thrombolysis; (ii) catheter-directed thrombolysis is a minimally invasive approach that can be suggested for the treatment of massive or submassive PE, but the lack of large, randomized controlled trials represents a limitation to widespread use; (iii) novel pharmacological approaches, by agents inhibiting thrombin-activatable fibrinolysis inhibitor, factor Xia, and the complement cascade, are currently under investigation to improve PE-related outcomes in specific settings.
Collapse
Affiliation(s)
- Lorenzo Falsetti
- Clinica Medica, Dipartimento di Scienze Cliniche e Molecolari, Università Politecnica delle Marche, 60126 Ancona, Italy; (L.F.)
| | - Emanuele Guerrieri
- Emergency Medicine Residency Program, Università Politecnica delle Marche, 60126 Ancona, Italy; (E.G.)
| | - Vincenzo Zaccone
- Internal and Subintensive Medicine, Azienda Ospedaliero-Universitaria delle Marche, 60126 Ancona, Italy
| | - Giovanna Viticchi
- Clinica di Neurologia, Dipartimento Scienze Cliniche e Molecolare, Università Politecnica delle Marche, 60126 Ancona, Italy
| | - Silvia Santini
- Emergency Medicine Residency Program, Università Politecnica delle Marche, 60126 Ancona, Italy; (E.G.)
| | - Laura Giovenali
- Emergency Medicine Residency Program, Università Politecnica delle Marche, 60126 Ancona, Italy; (E.G.)
| | - Graziana Lagonigro
- Emergency Medicine Residency Program, Università Politecnica delle Marche, 60126 Ancona, Italy; (E.G.)
| | - Stella Carletti
- Emergency Medicine Residency Program, Università Politecnica delle Marche, 60126 Ancona, Italy; (E.G.)
| | | | - Nicola Tarquinio
- Internal Medicine Department, INRCA-IRCCS Osimo-Ancona, 60027 Ancona, Italy
| | - Gianluca Moroncini
- Clinica Medica, Dipartimento di Scienze Cliniche e Molecolari, Università Politecnica delle Marche, 60126 Ancona, Italy; (L.F.)
| |
Collapse
|
5
|
Mone P, Jankauskas SS, Manzi MV, Gambardella J, Coppola A, Kansakar U, Izzo R, Fiorentino G, Lombardi A, Varzideh F, Sorriento D, Trimarco B, Santulli G. Endothelial Extracellular Vesicles Enriched in microRNA-34a Predict New-Onset Diabetes in Coronavirus Disease 2019 (COVID-19) Patients: Novel Insights for Long COVID Metabolic Sequelae. J Pharmacol Exp Ther 2024; 389:34-39. [PMID: 38336381 PMCID: PMC10949163 DOI: 10.1124/jpet.122.001253] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2022] [Revised: 01/15/2024] [Accepted: 01/23/2024] [Indexed: 02/12/2024] Open
Abstract
Emerging evidence indicates that the relationship between coronavirus disease 2019 (COVID-19) and diabetes is 2-fold: 1) it is known that the presence of diabetes and other metabolic alterations poses a considerably high risk to develop a severe COVID-19; 2) patients who survived a severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection have an increased risk of developing new-onset diabetes. However, the mechanisms underlying this association are mostly unknown, and there are no reliable biomarkers to predict the development of new-onset diabetes. In the present study, we demonstrate that a specific microRNA (miR-34a) contained in circulating extracellular vesicles released by endothelial cells reliably predicts the risk of developing new-onset diabetes in COVID-19. This association was independent of age, sex, body mass index (BMI), hypertension, dyslipidemia, smoking status, and D-dimer. SIGNIFICANCE STATEMENT: We demonstrate for the first time that a specific microRNA (miR-34a) contained in circulating extracellular vesicles released by endothelial cells is able to reliably predict the risk of developing diabetes after having contracted coronavirus disease 2019 (COVID-19). This association was independent of age, sex, body mass index (BMI), hypertension, dyslipidemia, smoking status, and D-dimer. Our findings are also relevant when considering the emerging importance of post-acute sequelae of COVID-19, with systemic manifestations observed even months after viral negativization (long COVID).
Collapse
Affiliation(s)
- Pasquale Mone
- Department of Medicine, Einstein-Sinai Diabetes Research Center, Fleischer Institute for Diabetes and Metabolism, Einstein Institute for Aging Research (P.M., S.S.J., J.G., U.K., A.L., F.V., G.S.) and Department of Molecular Pharmacology, Wilf Family Cardiovascular Research Institute, Einstein Institute for Neuroimmunology and Inflammation (G.S.), Albert Einstein College of Medicine, New York, New York; Department of Advanced Biomedical Sciences, International Translational Research and Medical Education, "Federico II" University, Naples, Italy (M.V.M., J.G., R.I., D.S., B.T., G.S.); Clinica Montevergine, Mercogliano, Avellino (P.M.); and COVID-19 Division, A.O.R.N. Ospedali dei Colli, Naples, Italy (A.C., G.F.)
| | - Stanislovas S Jankauskas
- Department of Medicine, Einstein-Sinai Diabetes Research Center, Fleischer Institute for Diabetes and Metabolism, Einstein Institute for Aging Research (P.M., S.S.J., J.G., U.K., A.L., F.V., G.S.) and Department of Molecular Pharmacology, Wilf Family Cardiovascular Research Institute, Einstein Institute for Neuroimmunology and Inflammation (G.S.), Albert Einstein College of Medicine, New York, New York; Department of Advanced Biomedical Sciences, International Translational Research and Medical Education, "Federico II" University, Naples, Italy (M.V.M., J.G., R.I., D.S., B.T., G.S.); Clinica Montevergine, Mercogliano, Avellino (P.M.); and COVID-19 Division, A.O.R.N. Ospedali dei Colli, Naples, Italy (A.C., G.F.)
| | - Maria Virginia Manzi
- Department of Medicine, Einstein-Sinai Diabetes Research Center, Fleischer Institute for Diabetes and Metabolism, Einstein Institute for Aging Research (P.M., S.S.J., J.G., U.K., A.L., F.V., G.S.) and Department of Molecular Pharmacology, Wilf Family Cardiovascular Research Institute, Einstein Institute for Neuroimmunology and Inflammation (G.S.), Albert Einstein College of Medicine, New York, New York; Department of Advanced Biomedical Sciences, International Translational Research and Medical Education, "Federico II" University, Naples, Italy (M.V.M., J.G., R.I., D.S., B.T., G.S.); Clinica Montevergine, Mercogliano, Avellino (P.M.); and COVID-19 Division, A.O.R.N. Ospedali dei Colli, Naples, Italy (A.C., G.F.)
| | - Jessica Gambardella
- Department of Medicine, Einstein-Sinai Diabetes Research Center, Fleischer Institute for Diabetes and Metabolism, Einstein Institute for Aging Research (P.M., S.S.J., J.G., U.K., A.L., F.V., G.S.) and Department of Molecular Pharmacology, Wilf Family Cardiovascular Research Institute, Einstein Institute for Neuroimmunology and Inflammation (G.S.), Albert Einstein College of Medicine, New York, New York; Department of Advanced Biomedical Sciences, International Translational Research and Medical Education, "Federico II" University, Naples, Italy (M.V.M., J.G., R.I., D.S., B.T., G.S.); Clinica Montevergine, Mercogliano, Avellino (P.M.); and COVID-19 Division, A.O.R.N. Ospedali dei Colli, Naples, Italy (A.C., G.F.)
| | - Antonietta Coppola
- Department of Medicine, Einstein-Sinai Diabetes Research Center, Fleischer Institute for Diabetes and Metabolism, Einstein Institute for Aging Research (P.M., S.S.J., J.G., U.K., A.L., F.V., G.S.) and Department of Molecular Pharmacology, Wilf Family Cardiovascular Research Institute, Einstein Institute for Neuroimmunology and Inflammation (G.S.), Albert Einstein College of Medicine, New York, New York; Department of Advanced Biomedical Sciences, International Translational Research and Medical Education, "Federico II" University, Naples, Italy (M.V.M., J.G., R.I., D.S., B.T., G.S.); Clinica Montevergine, Mercogliano, Avellino (P.M.); and COVID-19 Division, A.O.R.N. Ospedali dei Colli, Naples, Italy (A.C., G.F.)
| | - Urna Kansakar
- Department of Medicine, Einstein-Sinai Diabetes Research Center, Fleischer Institute for Diabetes and Metabolism, Einstein Institute for Aging Research (P.M., S.S.J., J.G., U.K., A.L., F.V., G.S.) and Department of Molecular Pharmacology, Wilf Family Cardiovascular Research Institute, Einstein Institute for Neuroimmunology and Inflammation (G.S.), Albert Einstein College of Medicine, New York, New York; Department of Advanced Biomedical Sciences, International Translational Research and Medical Education, "Federico II" University, Naples, Italy (M.V.M., J.G., R.I., D.S., B.T., G.S.); Clinica Montevergine, Mercogliano, Avellino (P.M.); and COVID-19 Division, A.O.R.N. Ospedali dei Colli, Naples, Italy (A.C., G.F.)
| | - Raffaele Izzo
- Department of Medicine, Einstein-Sinai Diabetes Research Center, Fleischer Institute for Diabetes and Metabolism, Einstein Institute for Aging Research (P.M., S.S.J., J.G., U.K., A.L., F.V., G.S.) and Department of Molecular Pharmacology, Wilf Family Cardiovascular Research Institute, Einstein Institute for Neuroimmunology and Inflammation (G.S.), Albert Einstein College of Medicine, New York, New York; Department of Advanced Biomedical Sciences, International Translational Research and Medical Education, "Federico II" University, Naples, Italy (M.V.M., J.G., R.I., D.S., B.T., G.S.); Clinica Montevergine, Mercogliano, Avellino (P.M.); and COVID-19 Division, A.O.R.N. Ospedali dei Colli, Naples, Italy (A.C., G.F.)
| | - Giuseppe Fiorentino
- Department of Medicine, Einstein-Sinai Diabetes Research Center, Fleischer Institute for Diabetes and Metabolism, Einstein Institute for Aging Research (P.M., S.S.J., J.G., U.K., A.L., F.V., G.S.) and Department of Molecular Pharmacology, Wilf Family Cardiovascular Research Institute, Einstein Institute for Neuroimmunology and Inflammation (G.S.), Albert Einstein College of Medicine, New York, New York; Department of Advanced Biomedical Sciences, International Translational Research and Medical Education, "Federico II" University, Naples, Italy (M.V.M., J.G., R.I., D.S., B.T., G.S.); Clinica Montevergine, Mercogliano, Avellino (P.M.); and COVID-19 Division, A.O.R.N. Ospedali dei Colli, Naples, Italy (A.C., G.F.)
| | - Angela Lombardi
- Department of Medicine, Einstein-Sinai Diabetes Research Center, Fleischer Institute for Diabetes and Metabolism, Einstein Institute for Aging Research (P.M., S.S.J., J.G., U.K., A.L., F.V., G.S.) and Department of Molecular Pharmacology, Wilf Family Cardiovascular Research Institute, Einstein Institute for Neuroimmunology and Inflammation (G.S.), Albert Einstein College of Medicine, New York, New York; Department of Advanced Biomedical Sciences, International Translational Research and Medical Education, "Federico II" University, Naples, Italy (M.V.M., J.G., R.I., D.S., B.T., G.S.); Clinica Montevergine, Mercogliano, Avellino (P.M.); and COVID-19 Division, A.O.R.N. Ospedali dei Colli, Naples, Italy (A.C., G.F.)
| | - Fahimeh Varzideh
- Department of Medicine, Einstein-Sinai Diabetes Research Center, Fleischer Institute for Diabetes and Metabolism, Einstein Institute for Aging Research (P.M., S.S.J., J.G., U.K., A.L., F.V., G.S.) and Department of Molecular Pharmacology, Wilf Family Cardiovascular Research Institute, Einstein Institute for Neuroimmunology and Inflammation (G.S.), Albert Einstein College of Medicine, New York, New York; Department of Advanced Biomedical Sciences, International Translational Research and Medical Education, "Federico II" University, Naples, Italy (M.V.M., J.G., R.I., D.S., B.T., G.S.); Clinica Montevergine, Mercogliano, Avellino (P.M.); and COVID-19 Division, A.O.R.N. Ospedali dei Colli, Naples, Italy (A.C., G.F.)
| | - Daniela Sorriento
- Department of Medicine, Einstein-Sinai Diabetes Research Center, Fleischer Institute for Diabetes and Metabolism, Einstein Institute for Aging Research (P.M., S.S.J., J.G., U.K., A.L., F.V., G.S.) and Department of Molecular Pharmacology, Wilf Family Cardiovascular Research Institute, Einstein Institute for Neuroimmunology and Inflammation (G.S.), Albert Einstein College of Medicine, New York, New York; Department of Advanced Biomedical Sciences, International Translational Research and Medical Education, "Federico II" University, Naples, Italy (M.V.M., J.G., R.I., D.S., B.T., G.S.); Clinica Montevergine, Mercogliano, Avellino (P.M.); and COVID-19 Division, A.O.R.N. Ospedali dei Colli, Naples, Italy (A.C., G.F.)
| | - Bruno Trimarco
- Department of Medicine, Einstein-Sinai Diabetes Research Center, Fleischer Institute for Diabetes and Metabolism, Einstein Institute for Aging Research (P.M., S.S.J., J.G., U.K., A.L., F.V., G.S.) and Department of Molecular Pharmacology, Wilf Family Cardiovascular Research Institute, Einstein Institute for Neuroimmunology and Inflammation (G.S.), Albert Einstein College of Medicine, New York, New York; Department of Advanced Biomedical Sciences, International Translational Research and Medical Education, "Federico II" University, Naples, Italy (M.V.M., J.G., R.I., D.S., B.T., G.S.); Clinica Montevergine, Mercogliano, Avellino (P.M.); and COVID-19 Division, A.O.R.N. Ospedali dei Colli, Naples, Italy (A.C., G.F.)
| | - Gaetano Santulli
- Department of Medicine, Einstein-Sinai Diabetes Research Center, Fleischer Institute for Diabetes and Metabolism, Einstein Institute for Aging Research (P.M., S.S.J., J.G., U.K., A.L., F.V., G.S.) and Department of Molecular Pharmacology, Wilf Family Cardiovascular Research Institute, Einstein Institute for Neuroimmunology and Inflammation (G.S.), Albert Einstein College of Medicine, New York, New York; Department of Advanced Biomedical Sciences, International Translational Research and Medical Education, "Federico II" University, Naples, Italy (M.V.M., J.G., R.I., D.S., B.T., G.S.); Clinica Montevergine, Mercogliano, Avellino (P.M.); and COVID-19 Division, A.O.R.N. Ospedali dei Colli, Naples, Italy (A.C., G.F.)
| |
Collapse
|
6
|
Lui KO, Ma Z, Dimmeler S. SARS-CoV-2 induced vascular endothelial dysfunction: direct or indirect effects? Cardiovasc Res 2024; 120:34-43. [PMID: 38159046 DOI: 10.1093/cvr/cvad191] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/22/2023] [Revised: 10/18/2023] [Accepted: 11/03/2023] [Indexed: 01/03/2024] Open
Abstract
Clinical evidence reveals that manifestations of endothelial dysfunction are widely observed in COVID-19 and long-COVID patients. However, whether these detrimental effects are caused by direct infection of the endothelium or are indirectly mediated by systemic inflammation has been a matter of debate. It has been well acknowledged that endothelial cells (ECs) of the cardiovascular system ubiquitously express the SARS-CoV-2 entry receptor angiotensin-converting enzyme 2 (ACE2), yet accumulating evidence suggests that it is more predominantly expressed by pericytes and vascular smooth muscle cells of the mammalian blood vessel. Besides, replicative infection of ECs by SARS-CoV-2 has yet to be demonstrated both in vitro and in vivo. In this study, we review latest research on endothelial ACE2 expression in different vascular beds, and the heterogeneity in various EC subsets with differential ACE2 expression in response to SARS-CoV-2. We also discuss ACE2-independent alternative mechanisms underlying endothelial activation in COVID-19, and the clinical manifestations of SARS-CoV-2-induced endothelial dysfunction. Altogether, understanding ACE2-dependent and ACE2-independent mechanisms driving SARS-CoV-2-induced vascular dysfunction would shed light on strategies of more effective therapies targeting cardiovascular complications associated with COVID-19.
Collapse
Affiliation(s)
- Kathy O Lui
- Department of Chemical Pathology, and Li Ka Shing Institute of Health Science, Prince of Wales Hospital, The Chinese University of Hong Kong, 30-32 Ngan Shing Street, Sha Tin, New Territories, 999077 Hong Kong, China
| | - Zhangjing Ma
- Department of Chemical Pathology, and Li Ka Shing Institute of Health Science, Prince of Wales Hospital, The Chinese University of Hong Kong, 30-32 Ngan Shing Street, Sha Tin, New Territories, 999077 Hong Kong, China
| | - Stefanie Dimmeler
- Institute for Cardiovascular Regeneration, and Faculty of Biological Sciences, Goethe University Frankfurt, Theodor Stern Kai 7, 60590 Frankfurt, Germany
| |
Collapse
|
7
|
Zimmermann P, Sourij H, Aberer F, Rilstone S, Schierbauer J, Moser O. SGLT2 Inhibitors in Long COVID Syndrome: Is There a Potential Role? J Cardiovasc Dev Dis 2023; 10:478. [PMID: 38132646 PMCID: PMC10744331 DOI: 10.3390/jcdd10120478] [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: 10/16/2023] [Revised: 11/25/2023] [Accepted: 11/28/2023] [Indexed: 12/23/2023] Open
Abstract
The coronavirus disease (COVID)-19 has turned into a pandemic causing a global public health crisis. While acute COVID-19 mainly affects the respiratory system and can cause acute respiratory distress syndrome, an association with persistent inflammatory stress affecting different organ systems has been elucidated in long COVID syndrome (LCS). Increased severity and mortality rates have been reported due to cardiophysiological and metabolic systemic disorders as well as multiorgan failure in COVID-19, additionally accompanied by chronic dyspnea and fatigue in LCS. Hence, novel therapies have been tested to improve the outcomes of LCS of which one potential candidate might be sodium-glucose cotransporter 2 (SGLT2) inhibitors. The aim of this narrative review was to discuss rationales for investigating SGLT2 inhibitor therapy in people suffering from LCS. In this regard, we discuss their potential positive effects-next to the well described "cardio-renal-metabolic" conditions-with a focus on potential anti-inflammatory and beneficial systemic effects in LCS. However, potential beneficial as well as potential disadvantageous effects of SGLT2 inhibitors on the prevalence and long-term outcomes of COVID-19 will need to be established in ongoing research.
Collapse
Affiliation(s)
- Paul Zimmermann
- Division of Exercise Physiology and Metabolism, BaySpo—Bayreuth Center of Sport Science, University of Bayreuth, 95440 Bayreuth, Germany; (P.Z.); (S.R.); (J.S.)
- Interdisciplinary Center of Sportsmedicine Bamberg, Klinikum Bamberg, 96049 Bamberg, Germany
- Department of Cardiology, Klinikum Bamberg, 96049 Bamberg, Germany
| | - Harald Sourij
- Interdisciplinary Metabolic Medicine Research Group, Division of Endocrinology and Diabetology, Medical University of Graz, 8036 Graz, Austria; (H.S.); (F.A.)
| | - Felix Aberer
- Interdisciplinary Metabolic Medicine Research Group, Division of Endocrinology and Diabetology, Medical University of Graz, 8036 Graz, Austria; (H.S.); (F.A.)
| | - Sian Rilstone
- Division of Exercise Physiology and Metabolism, BaySpo—Bayreuth Center of Sport Science, University of Bayreuth, 95440 Bayreuth, Germany; (P.Z.); (S.R.); (J.S.)
- Faculty of Medicine, Department of Metabolism, Digestion and Reproduction, Imperial College London, London SW7 2AZ, UK
| | - Janis Schierbauer
- Division of Exercise Physiology and Metabolism, BaySpo—Bayreuth Center of Sport Science, University of Bayreuth, 95440 Bayreuth, Germany; (P.Z.); (S.R.); (J.S.)
| | - Othmar Moser
- Division of Exercise Physiology and Metabolism, BaySpo—Bayreuth Center of Sport Science, University of Bayreuth, 95440 Bayreuth, Germany; (P.Z.); (S.R.); (J.S.)
- Interdisciplinary Metabolic Medicine Research Group, Division of Endocrinology and Diabetology, Medical University of Graz, 8036 Graz, Austria; (H.S.); (F.A.)
| |
Collapse
|
8
|
Falsetti L, Zaccone V, Santoro L, Santini S, Guerrieri E, Giuliani L, Viticchi G, Cataldi S, Gasbarrini A, Landi F, Santoliquido A, Moroncini G. The Relationship between Post-COVID Syndrome and the Burden of Comorbidities Assessed Using the Charlson Comorbidity Index. MEDICINA (KAUNAS, LITHUANIA) 2023; 59:1583. [PMID: 37763702 PMCID: PMC10533175 DOI: 10.3390/medicina59091583] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/01/2023] [Revised: 08/22/2023] [Accepted: 08/30/2023] [Indexed: 09/29/2023]
Abstract
Introduction: The post-COVID-19 syndrome is a clinical entity characterized by the manifestation of signs and symptoms that develop after the acute phase of COVID-19, which persist for a duration of more than 12 weeks and are not explained by any alternative diagnosis. It has been observed that individuals with pre-existing chronic diseases, including cardiovascular and pulmonary diseases, are at a greater risk of developing post-COVID-19 syndrome. The Charlson Comorbidity Index (CCI) is a useful tool employed to evaluate the burden of comorbidities and predict the prognosis of patients with post-COVID-19 syndrome. The present study aims to assess whether the burden of comorbidities, evaluated using the CCI, correlates with post-COVID-19 syndrome. Materials and Methods: Between 21 April 2020 and 15 May 2023, we enrolled all consecutive outpatients with previous COVID-19 admissions to a post-acute day-hospital service three months after a negative SARS-CoV-2 molecular test. We assessed age, sex, BMI, acute COVID-19 and post-COVID-19 signs, and symptoms and calculated CCI according to its current definition. Post-COVID-19 syndrome was defined as the persistence of at least one sign or symptom lasting more than 12 weeks after COVID-19 resolution and not explained by an alternative diagnosis. The relationship between post-COVID-19 and CCI was explored first with the chi-squared test, then with different binary logistic regression models. We considered significant values of p lower than 0.05. Results: We obtained a cohort of 3636 patients and observed a significant association between the number of post-COVID-19 symptoms and CCI. Patients developing post-COVID-19 were more commonly affected by a greater burden of comorbidities. Patients with at least one CCI point had an increased risk of post-COVID-19 syndrome (OR:2.961; 95%CI: 2.269-3.863; p < 0.0001), which increased further for CCI ≥ 4 (OR:6.062; 95%CI: 3.163-11.618; p < 0.0001). Conclusions: Patients affected by post-COVID-19 show a greater clinical complexity and a larger burden of comorbidities, synthesized by a higher CCI; moreover, a higher CCI seems to correlate with an increasing post-COVID-19 risk, being the presence of ≥1 or ≥4 CCI points associated with a 3-fold and 6-fold increased risk of post-COVID-19 syndrome, respectively.
Collapse
Affiliation(s)
- Lorenzo Falsetti
- Clinica Medica, Department of Clinical and Molecular Sciences, Marche Polytechnic University, 60126 Ancona, Italy; (L.F.); (G.M.)
| | - Vincenzo Zaccone
- Internal and Subintensive Medicine, Azienda Ospedaliero-Universitaria delle Marche, 60126 Ancona, Italy;
| | - Luca Santoro
- Department of Cardiovascular and Thoracic Sciences, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00168 Rome, Italy;
| | - Silvia Santini
- Emergency Medicine Residency Program, Marche Polytechnic University, 60126 Ancona, Italy; (S.S.); (E.G.); (L.G.)
| | - Emanuele Guerrieri
- Emergency Medicine Residency Program, Marche Polytechnic University, 60126 Ancona, Italy; (S.S.); (E.G.); (L.G.)
| | - Luca Giuliani
- Emergency Medicine Residency Program, Marche Polytechnic University, 60126 Ancona, Italy; (S.S.); (E.G.); (L.G.)
| | - Giovanna Viticchi
- Clinica Neurologica, Department of Medical and Surgical Sciences, Marche Polytechnic University, 60126 Ancona, Italy;
| | - Serena Cataldi
- Department of Pediatrics, Marche Polytechnic University, 60126 Ancona, Italy;
| | - Antonio Gasbarrini
- Department of Medical and Surgical Sciences, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00168 Rome, Italy;
- Università Cattolica del Sacro Cuore, 00168 Rome, Italy;
| | - Francesco Landi
- Università Cattolica del Sacro Cuore, 00168 Rome, Italy;
- Geriatrics Department, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00168 Rome, Italy
| | - Angelo Santoliquido
- Department of Cardiovascular and Thoracic Sciences, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00168 Rome, Italy;
- Università Cattolica del Sacro Cuore, 00168 Rome, Italy;
| | - Gianluca Moroncini
- Clinica Medica, Department of Clinical and Molecular Sciences, Marche Polytechnic University, 60126 Ancona, Italy; (L.F.); (G.M.)
| | | |
Collapse
|
9
|
Santoro L, Zaccone V, Falsetti L, Ruggieri V, Danese M, Miro C, Di Giorgio A, Nesci A, D’Alessandro A, Moroncini G, Santoliquido A. Role of Endothelium in Cardiovascular Sequelae of Long COVID. Biomedicines 2023; 11:2239. [PMID: 37626735 PMCID: PMC10452509 DOI: 10.3390/biomedicines11082239] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2023] [Revised: 07/26/2023] [Accepted: 08/03/2023] [Indexed: 08/27/2023] Open
Abstract
The global action against coronavirus disease 2019 (COVID-19), caused by SARS-CoV-2 infection, shed light on endothelial dysfunction. Although SARS-CoV-2 primarily affects the pulmonary system, multiple studies have documented pan-vascular involvement in COVID-19. The virus is able to penetrate the endothelial barrier, damaging it directly or indirectly and causing endotheliitis and multi-organ injury. Several mechanisms cooperate to development of endothelial dysfunction, including endothelial cell injury and pyroptosis, hyperinflammation and cytokine storm syndrome, oxidative stress and reduced nitric oxide bioavailability, glycocalyx disruption, hypercoagulability, and thrombosis. After acute-phase infection, some patients reported signs and symptoms of a systemic disorder known as long COVID, in which a broad range of cardiovascular (CV) disorders emerged. To date, the exact pathophysiology of long COVID remains unclear: in addition to the persistence of acute-phase infection mechanisms, specific pathways of CV damage have been postulated, such as persistent viral reservoirs in the heart or an autoimmune response to cardiac antigens through molecular mimicry. The aim of this review is to provide an overview of the main molecular patterns of enduring endothelial activation following SARS-CoV-2 infection and to offer the latest summary of CV complications in long COVID.
Collapse
Affiliation(s)
- Luca Santoro
- Department of Cardiovascular and Thoracic Sciences, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00168 Rome, Italy; (L.S.); (A.D.G.); (A.N.); (A.D.); (A.S.)
| | - Vincenzo Zaccone
- Department of Emergency Medicine, Internal and Sub-Intensive Medicine, Azienda Ospedaliero-Universitaria delle Marche, 60126 Ancona, Italy
| | - Lorenzo Falsetti
- Clinica Medica, Department of Clinical and Molecular Sciences, Università Politecnica delle Marche, 60126 Ancona, Italy; (L.F.); (G.M.)
| | - Vittorio Ruggieri
- Department of Internal Medicine, Università Cattolica del Sacro Cuore, 00168 Rome, Italy; (V.R.); (M.D.); (C.M.)
| | - Martina Danese
- Department of Internal Medicine, Università Cattolica del Sacro Cuore, 00168 Rome, Italy; (V.R.); (M.D.); (C.M.)
| | - Chiara Miro
- Department of Internal Medicine, Università Cattolica del Sacro Cuore, 00168 Rome, Italy; (V.R.); (M.D.); (C.M.)
| | - Angela Di Giorgio
- Department of Cardiovascular and Thoracic Sciences, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00168 Rome, Italy; (L.S.); (A.D.G.); (A.N.); (A.D.); (A.S.)
| | - Antonio Nesci
- Department of Cardiovascular and Thoracic Sciences, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00168 Rome, Italy; (L.S.); (A.D.G.); (A.N.); (A.D.); (A.S.)
| | - Alessia D’Alessandro
- Department of Cardiovascular and Thoracic Sciences, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00168 Rome, Italy; (L.S.); (A.D.G.); (A.N.); (A.D.); (A.S.)
| | - Gianluca Moroncini
- Clinica Medica, Department of Clinical and Molecular Sciences, Università Politecnica delle Marche, 60126 Ancona, Italy; (L.F.); (G.M.)
| | - Angelo Santoliquido
- Department of Cardiovascular and Thoracic Sciences, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00168 Rome, Italy; (L.S.); (A.D.G.); (A.N.); (A.D.); (A.S.)
- Università Cattolica del Sacro Cuore, 00168 Rome, Italy
| |
Collapse
|
10
|
Boccatonda A, Campello E, Simion C, Simioni P. Long-term hypercoagulability, endotheliopathy and inflammation following acute SARS-CoV-2 infection. Expert Rev Hematol 2023; 16:1035-1048. [PMID: 38018136 DOI: 10.1080/17474086.2023.2288154] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2023] [Accepted: 11/22/2023] [Indexed: 11/30/2023]
Abstract
INTRODUCTION both symptomatic and asymptomatic SARS-CoV-2 infections - coined Coronavirus disease 2019 (COVID-19) - have been linked to a higher risk of cardiovascular events after recovery. AREAS COVERED our review aims to summarize the latest evidence on the increased thrombotic and cardiovascular risk in recovered COVID-19 patients and to examine the pathophysiological mechanisms underlying the interplay among endothelial dysfunction, inflammatory response and coagulation in long-COVID. We performed a systematic search of studies on hypercoagulability, endothelial dysfunction and inflammation after SARS-CoV-2 infection. EXPERT OPINION endothelial dysfunction is a major pathophysiological mechanism responsible for most clinical manifestations in COVID-19. The pathological activation of endothelial cells by a virus infection results in a pro-adhesive and chemokine-secreting phenotype, which in turn promotes the recruitment of circulating leukocytes. Cardiovascular events after COVID-19 appear to be related to persistent immune dysregulation. Patients with long-lasting symptoms display higher amounts of proinflammatory molecules such as tumor necrosis factor-α, interferon γ and interleukins 2 and 6. Immune dysregulation can trigger the activation of the coagulation pathway. The formation of extensive microclots in vivo, both during acute COVID-19 and in long-COVID-19, appears to be a relevant mechanism responsible for persistent symptoms and cardiovascular events.
Collapse
Affiliation(s)
- Andrea Boccatonda
- Internal Medicine, Bentivoglio Hospital, AUSL Bologna, Bentivoglio, Italy
| | - Elena Campello
- General Medicine and Thrombotic and Hemorrhagic Diseases Unit, Department of Medicine, University Hospital of Padova, Padova, Italy
| | - Chiara Simion
- General Medicine and Thrombotic and Hemorrhagic Diseases Unit, Department of Medicine, University Hospital of Padova, Padova, Italy
| | - Paolo Simioni
- General Medicine and Thrombotic and Hemorrhagic Diseases Unit, Department of Medicine, University Hospital of Padova, Padova, Italy
| |
Collapse
|
11
|
Komaroff AL, Lipkin WI. ME/CFS and Long COVID share similar symptoms and biological abnormalities: road map to the literature. Front Med (Lausanne) 2023; 10:1187163. [PMID: 37342500 PMCID: PMC10278546 DOI: 10.3389/fmed.2023.1187163] [Citation(s) in RCA: 74] [Impact Index Per Article: 74.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2023] [Accepted: 05/09/2023] [Indexed: 06/23/2023] Open
Abstract
Some patients remain unwell for months after "recovering" from acute COVID-19. They develop persistent fatigue, cognitive problems, headaches, disrupted sleep, myalgias and arthralgias, post-exertional malaise, orthostatic intolerance and other symptoms that greatly interfere with their ability to function and that can leave some people housebound and disabled. The illness (Long COVID) is similar to myalgic encephalomyelitis/chronic fatigue syndrome (ME/CFS) as well as to persisting illnesses that can follow a wide variety of other infectious agents and following major traumatic injury. Together, these illnesses are projected to cost the U.S. trillions of dollars. In this review, we first compare the symptoms of ME/CFS and Long COVID, noting the considerable similarities and the few differences. We then compare in extensive detail the underlying pathophysiology of these two conditions, focusing on abnormalities of the central and autonomic nervous system, lungs, heart, vasculature, immune system, gut microbiome, energy metabolism and redox balance. This comparison highlights how strong the evidence is for each abnormality, in each illness, and helps to set priorities for future investigation. The review provides a current road map to the extensive literature on the underlying biology of both illnesses.
Collapse
Affiliation(s)
- Anthony L. Komaroff
- Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, United States
| | - W. Ian Lipkin
- Center for Infection and Immunity, Mailman School of Public Health, Vagelos College of Physicians and Surgeons of Columbia University, New York, NY, United States
| |
Collapse
|
12
|
Sykes RA, Neves KB, Alves-Lopes R, Caputo I, Fallon K, Jamieson NB, Kamdar A, Legrini A, Leslie H, McIntosh A, McConnachie A, Morrow A, McFarlane RW, Mangion K, McAbney J, Montezano AC, Touyz RM, Wood C, Berry C. Vascular mechanisms of post-COVID-19 conditions: Rho-kinase is a novel target for therapy. EUROPEAN HEART JOURNAL. CARDIOVASCULAR PHARMACOTHERAPY 2023; 9:371-386. [PMID: 37019821 PMCID: PMC10236521 DOI: 10.1093/ehjcvp/pvad025] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/09/2023] [Revised: 02/24/2023] [Accepted: 04/04/2023] [Indexed: 04/07/2023]
Abstract
BACKGROUND In post-coronavirus disease-19 (post-COVID-19) conditions (long COVID), systemic vascular dysfunction is implicated, but the mechanisms are uncertain, and the treatment is imprecise. METHODS AND RESULTS Patients convalescing after hospitalization for COVID-19 and risk factor matched controls underwent multisystem phenotyping using blood biomarkers, cardiorenal and pulmonary imaging, and gluteal subcutaneous biopsy (NCT04403607). Small resistance arteries were isolated and examined using wire myography, histopathology, immunohistochemistry, and spatial transcriptomics. Endothelium-independent (sodium nitroprusside) and -dependent (acetylcholine) vasorelaxation and vasoconstriction to the thromboxane A2 receptor agonist, U46619, and endothelin-1 (ET-1) in the presence or absence of a RhoA/Rho-kinase inhibitor (fasudil), were investigated. Thirty-seven patients, including 27 (mean age 57 years, 48% women, 41% cardiovascular disease) 3 months post-COVID-19 and 10 controls (mean age 57 years, 20% women, 30% cardiovascular disease), were included. Compared with control responses, U46619-induced constriction was increased (P = 0.002) and endothelium-independent vasorelaxation was reduced in arteries from COVID-19 patients (P < 0.001). This difference was abolished by fasudil. Histopathology revealed greater collagen abundance in COVID-19 arteries {Masson's trichrome (MT) 69.7% [95% confidence interval (CI): 67.8-71.7]; picrosirius red 68.6% [95% CI: 64.4-72.8]} vs. controls [MT 64.9% (95% CI: 59.4-70.3) (P = 0.028); picrosirius red 60.1% (95% CI: 55.4-64.8), (P = 0.029)]. Greater phosphorylated myosin light chain antibody-positive staining in vascular smooth muscle cells was observed in COVID-19 arteries (40.1%; 95% CI: 30.9-49.3) vs. controls (10.0%; 95% CI: 4.4-15.6) (P < 0.001). In proof-of-concept studies, gene pathways associated with extracellular matrix alteration, proteoglycan synthesis, and viral mRNA replication appeared to be upregulated. CONCLUSION Patients with post-COVID-19 conditions have enhanced vascular fibrosis and myosin light change phosphorylation. Rho-kinase activation represents a novel therapeutic target for clinical trials.
Collapse
Affiliation(s)
- Robert A Sykes
- School of Cardiovascular and Metabolic Health, University of Glasgow, UK
- West of Scotland Heart and Lung Centre, Golden Jubilee National Hospital, Glasgow, UK
| | - Karla B Neves
- School of Cardiovascular and Metabolic Health, University of Glasgow, UK
- Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow, UK
| | - Rhéure Alves-Lopes
- School of Cardiovascular and Metabolic Health, University of Glasgow, UK
| | - Ilaria Caputo
- Università degli Studi di Padova, 35122 Padova, Italy
| | - Kirsty Fallon
- Clinical Research Facility, Queen Elizabeth University Hospital, NHS Greater Glasgow & Clyde Health Board, Glasgow, UK
| | - Nigel B Jamieson
- Wolfson Wohl Cancer Research Centre, School of Cancer Sciences, University of Glasgow, Glasgow, UK
| | - Anna Kamdar
- School of Cardiovascular and Metabolic Health, University of Glasgow, UK
| | - Assya Legrini
- Wolfson Wohl Cancer Research Centre, School of Cancer Sciences, University of Glasgow, Glasgow, UK
| | - Holly Leslie
- Wolfson Wohl Cancer Research Centre, School of Cancer Sciences, University of Glasgow, Glasgow, UK
| | - Alasdair McIntosh
- Robertson Centre for Biostatistics, School of Health and Wellbeing, University of Glasgow, Glasgow, UK
| | - Alex McConnachie
- Robertson Centre for Biostatistics, School of Health and Wellbeing, University of Glasgow, Glasgow, UK
| | - Andrew Morrow
- School of Cardiovascular and Metabolic Health, University of Glasgow, UK
- West of Scotland Heart and Lung Centre, Golden Jubilee National Hospital, Glasgow, UK
| | | | - Kenneth Mangion
- School of Cardiovascular and Metabolic Health, University of Glasgow, UK
- Department of Cardiology, Queen Elizabeth University Hospital, NHS Greater Glasgow and Clyde Health Board, Glasgow, UK
| | - John McAbney
- Institute of Biomedical and Life Sciences (FBLS), University of Glasgow, Glasgow G12 8QQ, UK
| | - Augusto C Montezano
- School of Cardiovascular and Metabolic Health, University of Glasgow, UK
- Research Institute of the McGill University Health Centre (RI-MUHC), Montreal, QC H4A 3J1, Canada
| | - Rhian M Touyz
- School of Cardiovascular and Metabolic Health, University of Glasgow, UK
- Research Institute of the McGill University Health Centre (RI-MUHC), Montreal, QC H4A 3J1, Canada
| | - Colin Wood
- Wolfson Wohl Cancer Research Centre, School of Cancer Sciences, University of Glasgow, Glasgow, UK
| | - Colin Berry
- School of Cardiovascular and Metabolic Health, University of Glasgow, UK
- West of Scotland Heart and Lung Centre, Golden Jubilee National Hospital, Glasgow, UK
- Department of Cardiology, Queen Elizabeth University Hospital, NHS Greater Glasgow and Clyde Health Board, Glasgow, UK
| |
Collapse
|
13
|
Neves PFMD, Quaresma JAS, Queiroz MAF, Silva CC, Maia EV, Oliveira JSDS, Neves CMAD, Mendonça SDS, Falcão ASC, Melo GS, Santos IBF, Sousa JRD, Santos EJMD, Vasconcelos PFDC, Vallinoto ACR, Falcão LFM. Imbalance of Peripheral Temperature, Sympathovagal, and Cytokine Profile in Long COVID. BIOLOGY 2023; 12:biology12050749. [PMID: 37237560 DOI: 10.3390/biology12050749] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Revised: 03/17/2023] [Accepted: 03/21/2023] [Indexed: 05/28/2023]
Abstract
A persistent state of inflammation has been reported during the COVID-19 pandemic. This study aimed to assess short-term heart rate variability (HRV), peripheral body temperature, and serum cytokine levels in patients with long COVID. We evaluated 202 patients with long COVID symptoms categorized them according to the duration of their COVID symptoms (≤120 days, n = 81; >120 days, n = 121), in addition to 95 healthy individuals selected as controls. All HRV variables differed significantly between the control group and patients with long COVID in the ≤120 days group (p < 0.05), and participants in the long COVID ≤120 days group had higher temperatures than those in the long COVID >120 days group in all regions analysed (p < 0.05). Cytokine analysis showed higher levels of interleukin 17 (IL-17) and interleukin 2 (IL-2), and lower levels of interleukin 4 (IL-4) (p < 0.05). Our results suggest a reduction in parasympathetic activation during long COVID and an increase in body temperature due to possible endothelial damage caused by the maintenance of elevated levels of inflammatory mediators. Furthermore, high serum levels of IL-17 and IL-2 and low levels of IL-4 appear to constitute a long-term profile of COVID-19 cytokines, and these markers are potential targets for long COVID-treatment and prevention strategies.
Collapse
Affiliation(s)
| | - Juarez Antônio Simões Quaresma
- Center for Biological Health Sciences, State University of Pará, Belém 66087-670, Brazil
- Tropical Medicine Center, Federal University of Pará, Belém 66055-240, Brazil
- School of Medicine, São Paulo University, São Paulo 01246-903, Brazil
| | - Maria Alice Freitas Queiroz
- Laboratory of Virology, Institute of Biological Sciences, Federal University of Pará, Belém 66075-110, Brazil
| | - Camilla Costa Silva
- Center for Biological Health Sciences, State University of Pará, Belém 66087-670, Brazil
| | - Enzo Varela Maia
- Center for Biological Health Sciences, State University of Pará, Belém 66087-670, Brazil
| | | | | | | | | | - Giovana Salomão Melo
- Institute of Health Sciences, Federal University of Pará, Belém 66075-110, Brazil
| | | | - Jorge Rodrigues de Sousa
- Center for Biological Health Sciences, State University of Pará, Belém 66087-670, Brazil
- Institute of Health Sciences, Federal University of Pará, Belém 66075-110, Brazil
- Instituto Evandro Chagas, Secretaria de Vigilância em Saúde, Ministério da Saúde, Ananindeua 67030-000, Brazil
| | - Eduardo José Melo Dos Santos
- Laboratory of Genetic of Complex Discasse, Institute of Biological Sciences, Federal University of Pará, Belém 66075-110, Brazil
- Graduate Program in Biology of Infectious and Parasitic Agents, Federal University of Pará, Belém 66075-110, Brazil
| | - Pedro Fernando da Costa Vasconcelos
- Center for Biological Health Sciences, State University of Pará, Belém 66087-670, Brazil
- Departamento de Arbovirologia e Febres Hemorrágicas, Instituto Evandro Chagas, Ananindeua 67030-000, Brazil
| | - Antonio Carlos Rosário Vallinoto
- Laboratory of Virology, Institute of Biological Sciences, Federal University of Pará, Belém 66075-110, Brazil
- Graduate Program in Biology of Infectious and Parasitic Agents, Federal University of Pará, Belém 66075-110, Brazil
| | - Luiz Fábio Magno Falcão
- Center for Biological Health Sciences, State University of Pará, Belém 66087-670, Brazil
- School of Medicine, São Paulo University, São Paulo 01246-903, Brazil
| |
Collapse
|
14
|
Kalinskaya A, Vorobyeva D, Rusakovich G, Maryukhnich E, Anisimova A, Dukhin O, Elizarova A, Ivanova O, Bugrova A, Brzhozovskiy A, Kononikhin A, Nikolaev E, Vasilieva E. Targeted Blood Plasma Proteomics and Hemostasis Assessment of Post COVID-19 Patients with Acute Myocardial Infarction. Int J Mol Sci 2023; 24:ijms24076523. [PMID: 37047497 PMCID: PMC10094800 DOI: 10.3390/ijms24076523] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Revised: 03/26/2023] [Accepted: 03/28/2023] [Indexed: 04/03/2023] Open
Abstract
The molecular mechanisms underlying cardiovascular complications after the SARS-CoV-2 infection remain unknown. The goal of our study was to analyze the features of blood coagulation, platelet aggregation, and plasma proteomics in COVID-19 convalescents with AMI. The study included 66 AMI patients and 58 healthy volunteers. The groups were divided according to the anti-N IgG levels (AMI post-COVID (n = 44), AMI control (n = 22), control post-COVID (n = 31), and control (n = 27)). All participants underwent rotational thromboelastometry, thrombodynamics, impedance aggregometry, and blood plasma proteomics analysis. Both AMI groups of patients demonstrated higher values of clot growth rates, thrombus size and density, as well as the elevated levels of components of the complement system, proteins modifying the state of endothelium, acute-phase and procoagulant proteins. In comparison with AMI control, AMI post-COVID patients demonstrated decreased levels of proteins connected to inflammation and hemostasis (lipopolysaccharide-binding protein, C4b-binding protein alpha-chain, plasma protease C1 inhibitor, fibrinogen beta-chain, vitamin K-dependent protein S), and altered correlations between inflammation and fibrinolysis. A new finding is that AMI post-COVID patients opposite the AMI control group, are characterized by a less noticeable growth of acute-phase proteins and hemostatic markers that could be explained by prolonged immune system alteration after COVID-19.
Collapse
|
15
|
Gounaridi MI, Vontetsianos A, Oikonomou E, Theofilis P, Chynkiamis N, Lampsas S, Anastasiou A, Papamikroulis GA, Katsianos E, Kalogeras K, Pesiridis T, Tsatsaragkou A, Vavuranakis M, Koulouris N, Siasos G. The Role of Rehabilitation in Arterial Function Properties of Convalescent COVID-19 Patients. J Clin Med 2023; 12:2233. [PMID: 36983234 PMCID: PMC10056228 DOI: 10.3390/jcm12062233] [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: 12/27/2022] [Revised: 02/25/2023] [Accepted: 03/12/2023] [Indexed: 03/15/2023] Open
Abstract
Coronavirus disease (COVID-19) is a respiratory disease, although arterial function involvement has been documented. We assess the impact of a post-acute COVID-19 rehabilitation program on endothelium-dependent vasodilation and arterial wall properties. We enrolled 60 convalescent patients from COVID-19 and one-month post-acute disease, who were randomized at a 1:1 ratio in a 3-month cardiopulmonary rehabilitation program (study group) or not (control group). Endothelium-dependent vasodilation was evaluated by flow-mediated dilation (FMD), and arterial wall properties were evaluated by carotid-femoral pulse wave velocity (cf-PWV) and augmentation index (AIx) at 1 month and at 4 months post-acute disease. FMD was significantly improved in both the study (6.2 ± 1.8% vs. 8.6 ± 2.4%, p < 0.001) and control groups (5.9 ± 2.2% vs. 6.6 ± 1.8%, p = 0.009), but the improvement was significantly higher in the study group (rehabilitation) (p < 0.001). PWV was improved in the study group (8.2 ± 1.3 m/s vs. 6.6 ± 1.0 m/s, p < 0.001) but not in the control group (8.9 ± 1.8 m/s vs. 8.8 ± 1.9 m/s, p = 0.74). Similarly, AIx was improved in the study group (25.9 ± 9.8% vs. 21.1 ± 9.3%, p < 0.001) but not in the control group (27.6 ± 9.2% vs. 26.2 ± 9.8 m/s, p = 0.15). Convalescent COVID-19 subjects of the study group (rehabilitation) with increased serum levels of circulating IL-6 had a greater reduction in FMD. Conclusively, a 3-month cardiopulmonary post-acute COVID-19 rehabilitation program improves recovery of endothelium-dependent vasodilation and arteriosclerosis.
Collapse
Affiliation(s)
- Maria Ioanna Gounaridi
- 3rd Department of Cardiology, Sotiria Chest Disease Hospital, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece
| | - Angelos Vontetsianos
- Rehabilitation Unit, 1st Respiratory Medicine Department, Sotiria Hospital, National and Kapodistrian University of Athens, 11527 Athens, Greece
| | - Evangelos Oikonomou
- 3rd Department of Cardiology, Sotiria Chest Disease Hospital, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece
| | - Panagiotis Theofilis
- 1st Department of Cardiology, Hippokration General Hospital, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece
| | - Nikolaos Chynkiamis
- Rehabilitation Unit, 1st Respiratory Medicine Department, Sotiria Hospital, National and Kapodistrian University of Athens, 11527 Athens, Greece
| | - Stamatios Lampsas
- 3rd Department of Cardiology, Sotiria Chest Disease Hospital, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece
| | - Artemis Anastasiou
- 3rd Department of Cardiology, Sotiria Chest Disease Hospital, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece
| | - Georgios Angelos Papamikroulis
- 3rd Department of Cardiology, Sotiria Chest Disease Hospital, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece
| | - Efstratios Katsianos
- 3rd Department of Cardiology, Sotiria Chest Disease Hospital, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece
| | - Konstantinos Kalogeras
- 3rd Department of Cardiology, Sotiria Chest Disease Hospital, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece
| | - Theodoros Pesiridis
- 3rd Department of Cardiology, Sotiria Chest Disease Hospital, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece
| | - Aikaterini Tsatsaragkou
- 3rd Department of Cardiology, Sotiria Chest Disease Hospital, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece
| | - Manolis Vavuranakis
- 3rd Department of Cardiology, Sotiria Chest Disease Hospital, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece
| | - Nikolaos Koulouris
- Rehabilitation Unit, 1st Respiratory Medicine Department, Sotiria Hospital, National and Kapodistrian University of Athens, 11527 Athens, Greece
| | - Gerasimos Siasos
- 3rd Department of Cardiology, Sotiria Chest Disease Hospital, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece
- Cardiovascular Division, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA
| |
Collapse
|
16
|
Tosato M, Calvani R, Picca A, Ciciarello F, Galluzzo V, Coelho-Júnior HJ, Di Giorgio A, Di Mario C, Gervasoni J, Gremese E, Leone PM, Nesci A, Paglionico AM, Santoliquido A, Santoro L, Santucci L, Tolusso B, Urbani A, Marini F, Marzetti E, Landi F. Effects of l-Arginine Plus Vitamin C Supplementation on Physical Performance, Endothelial Function, and Persistent Fatigue in Adults with Long COVID: A Single-Blind Randomized Controlled Trial. Nutrients 2022; 14:4984. [PMID: 36501014 PMCID: PMC9738241 DOI: 10.3390/nu14234984] [Citation(s) in RCA: 33] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2022] [Revised: 11/19/2022] [Accepted: 11/20/2022] [Indexed: 11/25/2022] Open
Abstract
Long COVID, a condition characterized by symptom and/or sign persistence following an acute COVID-19 episode, is associated with reduced physical performance and endothelial dysfunction. Supplementation of l-arginine may improve endothelial and muscle function by stimulating nitric oxide synthesis. A single-blind randomized, placebo-controlled trial was conducted in adults aged between 20 and 60 years with persistent fatigue attending a post-acute COVID-19 outpatient clinic. Participants were randomized 1:1 to receive twice-daily orally either a combination of 1.66 g l-arginine plus 500 mg liposomal vitamin C or a placebo for 28 days. The primary outcome was the distance walked on the 6 min walk test. Secondary outcomes were handgrip strength, flow-mediated dilation, and fatigue persistence. Fifty participants were randomized to receive either l-arginine plus vitamin C or a placebo. Forty-six participants (median (interquartile range) age 51 (14), 30 [65%] women), 23 per group, received the intervention to which they were allocated and completed the study. At 28 days, l-arginine plus vitamin C increased the 6 min walk distance (+30 (40.5) m; placebo: +0 (75) m, p = 0.001) and induced a greater improvement in handgrip strength (+3.4 (7.5) kg) compared with the placebo (+1 (6.6) kg, p = 0.03). The flow-mediated dilation was greater in the active group than in the placebo (14.3% (7.3) vs. 9.4% (5.8), p = 0.03). At 28 days, fatigue was reported by two participants in the active group (8.7%) and 21 in the placebo group (80.1%; p < 0.0001). l-arginine plus vitamin C supplementation improved walking performance, muscle strength, endothelial function, and fatigue in adults with long COVID. This supplement may, therefore, be considered to restore physical performance and relieve persistent symptoms in this patient population.
Collapse
Affiliation(s)
- Matteo Tosato
- Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy
| | - Riccardo Calvani
- Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy
| | - Anna Picca
- Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy
- Department of Medicine and Surgery, LUM University, 70010 Casamassima, Italy
| | | | - Vincenzo Galluzzo
- Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy
| | - Hélio José Coelho-Júnior
- Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy
- Department of Geriatrics and Orthopedics, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
| | - Angela Di Giorgio
- Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy
| | - Clara Di Mario
- Immunology Core Facility, Gemelli Science Technological Park (GSTeP), Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy
| | - Jacopo Gervasoni
- Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy
| | - Elisa Gremese
- Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy
- Department of Geriatrics and Orthopedics, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
- Immunology Core Facility, Gemelli Science Technological Park (GSTeP), Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy
| | - Paolo Maria Leone
- Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy
| | - Antonio Nesci
- Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy
| | | | - Angelo Santoliquido
- Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy
- Department of Cardiovascular Sciences, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
| | - Luca Santoro
- Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy
| | - Lavinia Santucci
- Metabolomics Research Core Facility, Gemelli Science and Technology Park (GSTeP), Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy
| | - Barbara Tolusso
- Immunology Core Facility, Gemelli Science Technological Park (GSTeP), Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy
| | - Andrea Urbani
- Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy
- Department of Biochemistry and Clinical Biochemistry, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
| | - Federico Marini
- Department of Chemistry, Sapienza University of Rome, 00185 Rome, Italy
| | - Emanuele Marzetti
- Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy
- Department of Geriatrics and Orthopedics, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
| | - Francesco Landi
- Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy
- Department of Geriatrics and Orthopedics, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
| |
Collapse
|
17
|
Abstract
Hintergrund Pulmonale Manifestationen sind sehr häufige Folgeerscheinungen nach einer Severe-acute-respiratory-syndrome-coronavirus-type-2(SARS-CoV-2)-Infektion, die unter dem Begriff Long-COVID-Syndrom (COVID „coronavirus disease“) zusammengefasst werden. Ziel und Methoden Zusammenfassung der aktuellen Literatur zu den pulmonalen Manifestationen mit einem Fokus auf Expertenempfehlungen. Ergebnisse Dyspnoe ist nach der chronischen Fatigue das häufigste Symptom bei Patienten mit Long-COVID-Syndrom. Auffällige Befunde finden sich vor allem nach schwerem akutem COVID-19-Verlauf und beinhalten radiologische Veränderungen im Sinne interstitieller Lungenerkrankungen, restriktive lungenfunktionelle Befunde und Einschränkungen der Diffusionskapazität als häufigsten pathologischen Befund. Obwohl sich sowohl Beschwerden als auch pathologische pulmonale Befunde im Verlauf bessern, können einige Patienten noch Monate nach der akuten Infektion unter Auffälligkeiten leiden. Dabei ist die Relevanz der pathologischen Befunde sowie eine Beteiligung funktioneller respiratorischer Einschränkungen, einer kardiopulmonalen Dekonditionierung, nichtsomatischer Ursachen und vorbestehender Erkrankungen aktuell unklar. Die diagnostische Abklärung fokussiert entsprechend auf Risikopatienten und schließt neben einer bildgebenden und lungenfunktionellen Abklärung eine Belastungsuntersuchung und bei unklaren Befunden eine Echokardiographie zur Diagnose einer pulmonalvaskulären Komponente ein. Die therapeutischen Möglichkeiten beinhalten aktuell die leitliniengerechte Therapie von Ursachen der Beschwerden (beispielsweise interstitielle Lungenerkrankungen, Husten) und Rehabilitationsmaßnahmen. Schlussfolgerung Das aktuelle Wissen zum Krankheitsbild wird ständig erweitert, allerdings existieren aufgrund mangelnder Studienlage noch keine evidenzbasierten Leitlinien zur Diagnostik und Therapie pulmonaler Manifestationen beim Long-COVID-Syndrom.
Collapse
Affiliation(s)
- Natascha Sommer
- Excellence Cluster Cardio-Pulmonary Institute (CPI), Universities of Giessen and Marburg Lung Center (UGMLC), Justus-Liebig-Universität, Klinikstr. 33, 35390 Gießen, Deutschland
| | - Bernd Schmeck
- Klinik für Innere Medizin mit Schwerpunkt Pneumologie, Schlaf- und Intensivmedizin, Sektion für Atemwegsinfektionen, Philipps-Universität Marburg, Marburg, Deutschland
- Institut für Lungenforschung, Universities of Giessen and Marburg Lung Center (UGMLC), Mitglied des Deutschen Zentrums für Lungenforschung (DZL) und des Deutschen Zentrums für Infektionsforschung (DZIF), Philipps-Universität Marburg, Marburg, Deutschland
| |
Collapse
|
18
|
Radial Peripapillary Capillary Plexus Perfusion and Endothelial Dysfunction in Early Post-SARS-CoV-2 Infection. Vision (Basel) 2022; 6:vision6020026. [PMID: 35645380 PMCID: PMC9149887 DOI: 10.3390/vision6020026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Revised: 04/27/2022] [Accepted: 05/02/2022] [Indexed: 11/24/2022] Open
Abstract
Background: Endothelial cells damage and thromboinflammation are considered key elements in the generation of organ impairment in patients with COVID-19 disease. The endothelial function is evaluated by measuring flow-mediated dilation (FMD). We aimed to analyze the association between FMD impairment and retinal vascular parameters in early post-COVID-19 patients. 00118-00199Tomography (OCT), OCT Angiography (OCTA) and slit lamp examination were performed. FMD ≤ 7% was considered as pathological. Our primary outcome was to assess potential differences in the radial peripapillary capillary plexus flow index (RPCP-FI) and RPCP density (RPCP-D) values between post-COVID-19 patients with and without FMD impairment. The associations of other retinal vascular parameters with FMD impairment were assessed as secondary endpoints. Results: FMD impairment was detected in 31 patients (37.8%). RPCP-FI (p = 0.047), age (p = 0.048) and prevalence of diabetes (p = 0.046) significantly differed in patients with FMD ≤ 7% in regression analysis. RPCP-FI was linearly correlated with FMD values (R = 0.244, p =0.027). SCT was found to be lower in patients with impaired FMD (p = 0.004), although this difference was only a trend in binary logistic regression output (p = 0.07). Conclusions: Early post-COVID-19 patients showed a higher prevalence of FMD impairment compared to the general population. Age, diabetes and RPCP-FI were independently correlated with the presence of endothelial impairment in the early post-infective period.
Collapse
|