1
|
Bello F, Fagni F, Bagni G, Hill CL, Mohammad AJ, Moiseev S, Olivotto I, Seyahi E, Emmi G. Arterial and venous thrombosis in systemic and monogenic vasculitis. Nat Rev Rheumatol 2025:10.1038/s41584-025-01252-7. [PMID: 40329108 DOI: 10.1038/s41584-025-01252-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/03/2025] [Indexed: 05/08/2025]
Abstract
Systemic vasculitis, common forms of which include anti-neutrophil cytoplasmic antibody-associated small-vessel vasculitis, large-vessel vasculitis and Behçet syndrome, are frequently complicated by arterial or venous thrombotic events (AVTEs). Newly identified entities such as DADA2 (deficiency of adenosine deaminase 2) and VEXAS (vacuoles, E1 enzyme, X-linked, autoinflammatory, somatic) syndrome, which are driven by genetic mutations, also exhibit vasculitic features and are associated with a high risk of AVTEs. AVTEs in systemic vasculitis, including monogenic forms of vasculitis, are due to the complex interaction of inflammation and coagulation. New insights into the pathogenetic mechanisms implicate endothelial dysfunction, immune complex deposition and the interplay of pro-inflammatory cytokines with prothrombotic factors, which collectively promote thrombus formation. AVTEs impose a substantial disease burden, complicate diagnosis and negatively affect prognosis by increasing the risk of morbidity and mortality. Early diagnosis and treatment are crucial to prevent lasting damage. Management strategies should target both thrombosis and underlying inflammation. Antithrombotic therapies, including low-dose aspirin, or oral anticoagulants should be used on the basis of individual thrombotic risk assessment. Immunosuppressive therapy is the cornerstone of treatment for arterial and venous thrombosis, particularly in Behçet syndrome, in which vascular inflammation has a crucial role in thrombotic complications.
Collapse
Affiliation(s)
- Federica Bello
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Filippo Fagni
- Department of Internal Medicine 3, Friedrich-Alexander University Erlangen-Nuremberg and Universitätsklinikum Erlangen, Erlangen, Germany
- Deutsches Zentrum für Immuntherapie, Friedrich-Alexander University Erlangen-Nuremberg and Universitätsklinikum Erlangen, Erlangen, Germany
| | - Giacomo Bagni
- Department of Clinical and Biological Sciences, University of Torino, Torino, Italy
| | - Catherine L Hill
- Rheumatology Unit, The Queen Elizabeth Hospital, Woodville, South Australia, Australia
- Department of Medicine, University of Adelaide, Adelaide, South Australia, Australia
| | - Aladdin J Mohammad
- Department of Clinical Sciences, Rheumatology, Lund University, Lund, Sweden
- Department of Medicine, University of Cambridge, Cambridge, UK
| | - Sergey Moiseev
- Tareev Clinic of Internal Disease, Sechenov First Moscow State Medical University, Moscow, Russia
| | - Iacopo Olivotto
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
- Meyer Children's Hospital IRCCS, Florence, Italy
| | - Emire Seyahi
- Division of Rheumatology, Department of Internal Medicine and Behçet's Disease Research Centre, Istanbul University-Cerrahpasa, School of Medicine, Istanbul, Turkey
| | - Giacomo Emmi
- Department of Medical, Surgical and Health Sciences, University of Trieste, Trieste, Italy.
- Clinical Medicine and Rheumatology Unit, Cattinara University Hospital, Trieste, Italy.
- Centre for Inflammatory Diseases, Monash University Department of Medicine, Monash Medical Centre, Melbourne, Victoria, Australia.
| |
Collapse
|
2
|
Mishra V, Agrawal S, Malik D, Mishra D, Bhavya B, Pathak E, Mishra R. Targeting Matrix Metalloproteinase-1, Matrix Metalloproteinase-7, and Serine Protease Inhibitor E1: Implications in preserving lung vascular endothelial integrity and immune modulation in COVID-19. Int J Biol Macromol 2025; 306:141602. [PMID: 40024412 DOI: 10.1016/j.ijbiomac.2025.141602] [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: 01/03/2025] [Revised: 02/25/2025] [Accepted: 02/27/2025] [Indexed: 03/04/2025]
Abstract
BACKGROUND SARS-CoV-2 disrupts lung vascular endothelial integrity, contributing to severe COVID-19 complications. However, the molecular mechanisms driving endothelial dysfunction remain underexplored, and targeted therapeutic strategies are lacking. OBJECTIVE This study investigates Naringenin-7-O-glucoside (N7G) as a multi-target therapeutic candidate for modulating vascular integrity and immune response by inhibiting MMP1, MMP7, and SERPINE1-key regulators of extracellular matrix (ECM) remodeling and inflammation. METHODS & RESULTS RNA-seq analysis of COVID-19 lung tissues identified 17 upregulated N7G targets, including MMP1, MMP7, and SERPINE1, with the latter exhibiting the highest expression. PPI network analysis linked these targets to ECM degradation, IL-17, HIF-1, and AGE-RAGE signaling pathways, and endothelial dysfunction. Disease enrichment associated these genes with idiopathic pulmonary fibrosis and asthma. Molecular docking, 200 ns MD simulations (triplicate), and MMGBSA calculations confirmed N7G's stable binding affinity to MMP1, MMP7, and SERPINE1. Immune profiling revealed increased neutrophils and activated CD4+ T cells, alongside reduced mast cells, NK cells, and naïve B cells, indicating immune dysregulation. Correlation analysis linked MMP1, MMP7, and SERPINE1 to distinct immune cell populations, supporting N7G's immunomodulatory role. CONCLUSION These findings suggest that N7G exhibits multi-target therapeutic potential by modulating vascular integrity, ECM remodeling, and immune dysregulation, positioning it as a promising candidate for mitigating COVID-19-associated endothelial dysfunction.
Collapse
Affiliation(s)
- Vibha Mishra
- Bioinformatics Department, MMV, Institute of Science, Banaras Hindu University, India
| | - Shivangi Agrawal
- Bioinformatics Department, MMV, Institute of Science, Banaras Hindu University, India
| | - Divya Malik
- Bioinformatics Department, MMV, Institute of Science, Banaras Hindu University, India
| | - Divya Mishra
- Bioinformatics Department, MMV, Institute of Science, Banaras Hindu University, India
| | - Bhavya Bhavya
- Bioinformatics Department, MMV, Institute of Science, Banaras Hindu University, India
| | - Ekta Pathak
- Institute of Diabetes and Obesity, Helmholtz Zentrum München, Neuherberg, Germany.
| | - Rajeev Mishra
- Bioinformatics Department, MMV, Institute of Science, Banaras Hindu University, India.
| |
Collapse
|
3
|
Subramaniam S, Jose A, Kenney D, O’Connell AK, Bosmann M, Douam F, Crossland N. Challenging the notion of endothelial infection by SARS-CoV-2: insights from the current scientific evidence. Front Immunol 2025; 16:1443932. [PMID: 39967675 PMCID: PMC11832389 DOI: 10.3389/fimmu.2025.1443932] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Accepted: 01/14/2025] [Indexed: 02/20/2025] Open
Affiliation(s)
- Saravanan Subramaniam
- Department of Pharmacology and Toxicology, Massachusetts College of Pharmacy and Health Sciences, Boston, MA, United States
- Renal Section, Department of Medicine, Chobanian & Avedisian School of Medicine, Boston University, Boston, MA, United States
| | - Asha Jose
- Renal Section, Department of Medicine, Chobanian & Avedisian School of Medicine, Boston University, Boston, MA, United States
| | - Devin Kenney
- Department of Virology, Immunology and Microbiology, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, United States
- National Emerging Infectious Diseases Laboratories (NEIDL), Boston University, Boston, MA, United States
| | - Aoife K. O’Connell
- National Emerging Infectious Diseases Laboratories (NEIDL), Boston University, Boston, MA, United States
| | - Markus Bosmann
- Department of Medicine, Pulmonary Center, Chobanian & Avedisian School of Medicine, Boston University, Boston, MA, United States
- Department of Pathology and Laboratory Medicine, Chobanian & Avedisian School of Medicine, Boston University, Boston, MA, United States
| | - Florian Douam
- Department of Virology, Immunology and Microbiology, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, United States
- National Emerging Infectious Diseases Laboratories (NEIDL), Boston University, Boston, MA, United States
| | - Nicholas Crossland
- Department of Virology, Immunology and Microbiology, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, United States
- National Emerging Infectious Diseases Laboratories (NEIDL), Boston University, Boston, MA, United States
- Department of Pathology and Laboratory Medicine, Chobanian & Avedisian School of Medicine, Boston University, Boston, MA, United States
| |
Collapse
|
4
|
Motyl JA, Gromadzka G, Czapski GA, Adamczyk A. SARS-CoV-2 Infection and Alpha-Synucleinopathies: Potential Links and Underlying Mechanisms. Int J Mol Sci 2024; 25:12079. [PMID: 39596147 PMCID: PMC11593367 DOI: 10.3390/ijms252212079] [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: 09/23/2024] [Revised: 10/30/2024] [Accepted: 11/08/2024] [Indexed: 11/28/2024] Open
Abstract
Alpha-synuclein (α-syn) is a 140-amino-acid, intrinsically disordered, soluble protein that is abundantly present in the brain. It plays a crucial role in maintaining cellular structures and organelle functions, particularly in supporting synaptic plasticity and regulating neurotransmitter turnover. However, for reasons not yet fully understood, α-syn can lose its physiological role and begin to aggregate. This altered α-syn disrupts dopaminergic transmission and causes both presynaptic and postsynaptic dysfunction, ultimately leading to cell death. A group of neurodegenerative diseases known as α-synucleinopathies is characterized by the intracellular accumulation of α-syn deposits in specific neuronal and glial cells within certain brain regions. In addition to Parkinson's disease (PD), these conditions include dementia with Lewy bodies (DLBs), multiple system atrophy (MSA), pure autonomic failure (PAF), and REM sleep behavior disorder (RBD). Given that these disorders are associated with α-syn-related neuroinflammation-and considering that SARS-CoV-2 infection has been shown to affect the nervous system, with COVID-19 patients experiencing neurological symptoms-it has been proposed that COVID-19 may contribute to neurodegeneration in PD and other α-synucleinopathies by promoting α-syn misfolding and aggregation. In this review, we focus on whether SARS-CoV-2 could act as an environmental trigger that facilitates the onset or progression of α-synucleinopathies. Specifically, we present new evidence on the potential role of SARS-CoV-2 in modulating α-syn function and discuss the causal relationship between SARS-CoV-2 infection and the development of parkinsonism-like symptoms.
Collapse
Affiliation(s)
- Joanna Agata Motyl
- Department of Hybrid Microbiosystems Engineering, Nalecz Institute of Biocybernetics and Biomedical Engineering, Polish Academy of Sciences, Ks. Trojdena 4 St., 02-109 Warsaw, Poland;
| | - Grażyna Gromadzka
- Department of Biomedical Sciences, Faculty of Medicine, Collegium Medicum, Cardinal Stefan Wyszynski University, Wóycickiego 1/3, 01-938 Warsaw, Poland;
| | - Grzegorz Arkadiusz Czapski
- Department of Cellular Signalling, Mossakowski Medical Research Institute, Polish Academy of Sciences, 02-106 Warsaw, Poland;
| | - Agata Adamczyk
- Department of Cellular Signalling, Mossakowski Medical Research Institute, Polish Academy of Sciences, 02-106 Warsaw, Poland;
| |
Collapse
|
5
|
Schramm T, Rast J, Mehic D, Reitsma SE, de Moreuil C, Fillitz M, Quehenberger P, de Laat B, Wolberg AS, Ay C, Pabinger I, Gebhart J. Fibrinolysis is impaired in patients with primary immune thrombocytopenia. J Thromb Haemost 2024; 22:3209-3220. [PMID: 39214313 DOI: 10.1016/j.jtha.2024.07.034] [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: 04/23/2024] [Revised: 06/27/2024] [Accepted: 07/25/2024] [Indexed: 09/04/2024]
Abstract
BACKGROUND Patients with primary immune thrombocytopenia (ITP) have an increased risk of thrombosis, which may be due to altered fibrinolysis. OBJECTIVES To elucidate the clinical impact of delayed fibrinolysis in ITP patients. METHODS A turbidimetric clot formation and lysis assay and a fluorometric plasmin generation (PG) assay were performed, and levels of plasminogen activator inhibitor-1 (PAI-1), tissue plasminogen activator (tPA), tPA-PAI-1 complexes, α2-antiplasmin, thrombin activatable fibrinolysis inhibitor, and D-dimer were assessed in 86 adult primary ITP patients and 78 healthy controls (HCs). RESULTS ITP patients showed significantly delayed clot formation, increased clot density, and prolonged clot lysis time (CLT) compared with HCs, with a median (IQR) CLT of 28.0 (13.7-34.7) minutes in patients and 17.3 (12.0-28.0) minutes in HCs, while in the PG assay, only the lag time was prolonged. In ITP patients compared with controls, PAI-1 was higher (1.2 [0.8-2.6] vs 1.1 [0.6-2.1] U/mL) and tPA antigen and activity were lower (tPA antigen: 2.6 [1.1-4.4] vs 3.7 [3.2-4.7] ng/mL; tPA activity ≤ 0 U/mL: 26% vs 7%). TPA-PAI-1 complex levels were positively associated with CLT in multiple linear regression analysis (β = 0.241; P = .019), whereas PG parameters were not associated with CLT. Six patients who developed thrombosis during follow-up had higher levels of tPA-PAI-1 complexes. CONCLUSION Prolonged CLT and delayed onset of PG may indicate a hypofibrinolytic tendency in ITP patients, as also indicated by high PAI-1 and low tPA levels. No association was found between fibrinolytic potential and the bleeding phenotype, whereas higher tPA-PAI-1 complex levels were associated with prolonged CLT and increased in patients with future thrombosis.
Collapse
Affiliation(s)
- Theresa Schramm
- Department of Medicine I, Division of Hematology and Hemostaseology, Medical University of Vienna, Vienna, Austria
| | - Jasmin Rast
- Department of Medicine I, Division of Hematology and Hemostaseology, Medical University of Vienna, Vienna, Austria
| | - Dino Mehic
- Department of Medicine I, Division of Hematology and Hemostaseology, Medical University of Vienna, Vienna, Austria
| | - Stéphanie E Reitsma
- Department of Pathology and Laboratory Medicine and University of North Carolina at Chapel Hill (UNC) Blood Research Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Claire de Moreuil
- Internal Medicine, Vascular Medicine and Pneumology Department, Brest University Hospital, Brest, France; Unité Mixte de Recherche (UMR) 1304, Groupe d'Etude de Thrombose de Bretagne Occidentale (GETBO), Institut National de la Santé et de la Recherche Médicale (INSERM), University of Brest, Brest, France
| | - Michael Fillitz
- Department of Internal Medicine, Hanusch Krankenhaus, Vienna, Austria
| | - Peter Quehenberger
- Department of Laboratory Medicine, Medical University of Vienna, Vienna, Austria
| | - Bas de Laat
- Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Maastricht, the Netherlands; Department of Functional Coagulation, Synapse Research Institute, Maastricht, The Netherlands; Department of Data Analysis and Artificial Intelligence, Synapse Research Institute, Maastricht, The Netherlands
| | - Alisa S Wolberg
- Department of Pathology and Laboratory Medicine and University of North Carolina at Chapel Hill (UNC) Blood Research Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Cihan Ay
- Department of Medicine I, Division of Hematology and Hemostaseology, Medical University of Vienna, Vienna, Austria
| | - Ingrid Pabinger
- Department of Medicine I, Division of Hematology and Hemostaseology, Medical University of Vienna, Vienna, Austria
| | - Johanna Gebhart
- Department of Medicine I, Division of Hematology and Hemostaseology, Medical University of Vienna, Vienna, Austria.
| |
Collapse
|
6
|
Pardo K, Harnof O, Barnea R, Naftali J, Kenan G, Auriel E, Peretz S. Arterial floating mural thrombi are a characteristic imaging pattern in SARS-CoV-2-related ischemic stroke. PLoS One 2024; 19:e0311622. [PMID: 39453913 PMCID: PMC11508162 DOI: 10.1371/journal.pone.0311622] [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: 05/30/2024] [Accepted: 09/17/2024] [Indexed: 10/27/2024] Open
Abstract
BACKGROUND Acute ischemic stroke (AIS) is a complication of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection. We aimed to explore neurovascular imaging patterns in patients with SARS-CoV-2-related AIS. METHODS We retrospectively analyzed clinical and radiological data of patients hospitalized with AIS and a positive PCR test for SARS-CoV-2 prior to AIS onset. The control group comprised of AIS patients from a pre-COVID-19 pandemic period matched for gender and age. RESULTS Thirty-five SARS-CoV-2-related stroke patients, and 35 controls were included. Fifty-seven percent of SARS-CoV-2 patients had either mild or asymptomatic disease. A distinctive imaging pattern of floating arterial mural thrombus was detected in 5 patients of the SARS-CoV-2 group. In 4 patients thrombus was attached to a stenotic atherosclerotic plaque in the proximal internal carotid artery. In the 5th patient a cardiac CTA showed multiple floating thrombi in the descending aorta. In the control group, floating thrombus was only detected in one patient. Treatment with dual antiplatelet therapy was associated with thrombus dissolution and good clinical outcome. Patients with floating thrombi had a longer time from SARS-CoV-2 diagnosis to stroke onset (mean 7.4 versus 3.4 days). CONCLUSIONS Floating arterial mural thrombi attached to atherosclerotic plaques are unique characteristic source of AIS in SARS-CoV-2 patients. They may lead to ischemic stroke in patients with mild or asymptomatic infection up to 1-2 weeks from SARS-CoV-2 diagnosis. Patients with embolic AIS and SARS-CoV-2 diagnosis should perform high resolution cranio-cervical vascular imaging to evaluate floating thrombi as a potential embolic source.
Collapse
Affiliation(s)
- Keshet Pardo
- Department of Neurology, Rabin Medical Center – Beilinson Hospital, Petach Tikva, Israel
- Sackler School of Medicine, Tel-Aviv University, Tel-Aviv, Israel
| | - Omer Harnof
- Sackler School of Medicine, Tel-Aviv University, Tel-Aviv, Israel
| | - Rani Barnea
- Department of Neurology, Rabin Medical Center – Beilinson Hospital, Petach Tikva, Israel
- Sackler School of Medicine, Tel-Aviv University, Tel-Aviv, Israel
| | - Jonathan Naftali
- Department of Neurology, Rabin Medical Center – Beilinson Hospital, Petach Tikva, Israel
- Sackler School of Medicine, Tel-Aviv University, Tel-Aviv, Israel
| | - Gilad Kenan
- Sackler School of Medicine, Tel-Aviv University, Tel-Aviv, Israel
- Department of Neurology, Shamir Medical Center, Be’er Ya’akov, Israel
| | - Eithan Auriel
- Department of Neurology, Rabin Medical Center – Beilinson Hospital, Petach Tikva, Israel
- Sackler School of Medicine, Tel-Aviv University, Tel-Aviv, Israel
| | - Shlomi Peretz
- Sackler School of Medicine, Tel-Aviv University, Tel-Aviv, Israel
- Department of Neurology, Shamir Medical Center, Be’er Ya’akov, Israel
| |
Collapse
|
7
|
Zhang W, Liu F, Liang E, Zhang L. Evolution of Treatment Modalities for Disseminated HAdV Infection in Neonates. Pediatrics 2024; 154:e2024066677. [PMID: 39238444 DOI: 10.1542/peds.2024-066677] [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] [Received: 03/21/2024] [Revised: 07/24/2024] [Accepted: 07/24/2024] [Indexed: 09/07/2024] Open
Abstract
Human adenovirus (HAdV) infection in newborns is a rare condition that typically affects multiple organ systems and has a high mortality rate. We report a case of neonatal HAdV-D37 infection that presented with fever and respiratory distress that was confirmed by metagenomic next-generation sequencing using blood and bronchoalveolar lavage fluid. We treated the patient with intravenous immunoglobulin, methylprednisolone, and anticoagulants, and the patient recovered. Our review of 41 cases of HAdV found that treatment with intravenous immunoglobin might have improved the outcome of HAdV-D infection. We further suggest that glucocorticoid therapy may have additional therapeutic validity in the setting of severe or disseminated disease and that monitoring coagulation function and timely anticoagulation treatment should be considered to prevent complications associated with disseminated intravascular coagulation.
Collapse
Affiliation(s)
- Wenjing Zhang
- Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu, China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Sichuan University, Ministry of Education, Chengdu, China
| | - Fang Liu
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Sichuan University, Ministry of Education, Chengdu, China
- Department of Laboratory Medicine, West China Second University Hospital, Sichuan University, Chengdu, China
| | - Enlin Liang
- Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu, China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Sichuan University, Ministry of Education, Chengdu, China
| | - Li Zhang
- Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu, China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Sichuan University, Ministry of Education, Chengdu, China
| |
Collapse
|
8
|
Rettew A, Garrahy I, Rahimian S, Brown R, Sangha N. COVID-19 Coagulopathy. Life (Basel) 2024; 14:953. [PMID: 39202695 PMCID: PMC11355811 DOI: 10.3390/life14080953] [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: 07/03/2024] [Revised: 07/25/2024] [Accepted: 07/26/2024] [Indexed: 09/03/2024] Open
Abstract
Coronavirus disease of 2019 (COVID-19) is the respiratory viral infection caused by the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). Despite being a primary respiratory illness, it is commonly complicated by systemic involvement of the vasculature leading to arterial and venous thrombosis. In this review, we will focus on the association between COVID-19 and thrombosis. We will highlight the pathophysiology of COVID-19 coagulopathy. The clinical manifestations of COVID-19 vasculopathy will be discussed with a focus on venous and arterial thromboembolic events. COVID-19 vasculopathy and disseminated intravascular coagulation (DIC) are distinguished within, as well as areas of controversy, such as "long COVID". Finally, the current professional guidelines on prevention and treatment of thrombosis associated with SARS-CoV-2 infection will be discussed.
Collapse
Affiliation(s)
| | - Ian Garrahy
- Tower Health System, Reading Hospital, West Reading, PA 19611, USA; (A.R.); (S.R.); (R.B.); (N.S.)
| | | | | | | |
Collapse
|
9
|
Sharma S, Antoniak S. Microbiota-driven coagulation activation during SARS-CoV-2 infection. J Thromb Haemost 2024; 22:1835-1837. [PMID: 38945665 DOI: 10.1016/j.jtha.2024.03.023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2024] [Revised: 03/23/2024] [Accepted: 03/25/2024] [Indexed: 07/02/2024]
Affiliation(s)
- Swati Sharma
- UNC Blood Research Center, Department of Pathology and Laboratory Medicine, School of Medicine, University of North Carolina, Chapel Hill, North Carolina, USA
| | - Silvio Antoniak
- UNC Blood Research Center, Department of Pathology and Laboratory Medicine, School of Medicine, University of North Carolina, Chapel Hill, North Carolina, USA.
| |
Collapse
|
10
|
Tsuruga T, Fujimoto H, Yasuma T, D'Alessandro-Gabazza CN, Toda M, Ito T, Tomaru A, Saiki H, Okano T, Alhawsawi MAB, Takeshita A, Nishihama K, Takei R, Kondoh Y, Cann I, Gabazza EC, Kobayashi T. Role of microbiota-derived corisin in coagulation activation during SARS-CoV-2 infection. J Thromb Haemost 2024; 22:1919-1935. [PMID: 38453025 DOI: 10.1016/j.jtha.2024.02.014] [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: 09/27/2023] [Revised: 01/23/2024] [Accepted: 02/13/2024] [Indexed: 03/09/2024]
Abstract
BACKGROUND Coagulopathy is a major cause of morbidity and mortality in COVID-19 patients. Hypercoagulability in COVID-19 results in deep vein thrombosis, thromboembolic complications, and diffuse intravascular coagulation. Microbiome dysbiosis influences the clinical course of COVID-19. However, the role of dysbiosis in COVID-19-associated coagulopathy is not fully understood. OBJECTIVES The present study tested the hypothesis that the microbiota-derived proapoptotic corisin is involved in the coagulation system activation during SARS-CoV-2 infection. METHODS This cross-sectional study included 47 consecutive patients who consulted for symptoms of COVID-19. A mouse acute lung injury model was used to recapitulate the clinical findings. A549 alveolar epithelial, THP-1, and human umbilical vein endothelial cells were used to evaluate procoagulant and anticoagulant activity of corisin. RESULTS COVID-19 patients showed significantly high circulating levels of corisin, thrombin-antithrombin complex, D-dimer, tumor necrosis factor-α, and monocyte-chemoattractant protein-1 with reduced levels of free protein S compared with healthy subjects. The levels of thrombin-antithrombin complex, D-dimer, and corisin were significantly correlated. A monoclonal anticorisin-neutralizing antibody significantly inhibited the inflammatory response and coagulation system activation in a SARS-CoV-2 spike protein-associated acute lung injury mouse model, and the levels of corisin and thrombin-antithrombin complex were significantly correlated. In an in vitro experiment, corisin increased the tissue factor activity and decreased the anticoagulant activity of thrombomodulin in epithelial, endothelial, and monocytic cells. CONCLUSION The microbiota-derived corisin is significantly increased and correlated with activation of the coagulation system during SARS-CoV-2 infection, and corisin may directly increase the procoagulant activity in epithelial, endothelial, and monocytic cells.
Collapse
Affiliation(s)
- Tatsuki Tsuruga
- Department of Pulmonary and Critical Care Medicine, Faculty and Graduate School of Medicine, Mie University, Tsu, Mie, Japan
| | - Hajime Fujimoto
- Department of Pulmonary and Critical Care Medicine, Faculty and Graduate School of Medicine, Mie University, Tsu, Mie, Japan
| | - Taro Yasuma
- Department of Pulmonary and Critical Care Medicine, Faculty and Graduate School of Medicine, Mie University, Tsu, Mie, Japan; Department of Immunology, Faculty and Graduate School of Medicine, Mie University, Tsu, Mie, Japan; Microbiome Research Center, Mie University, Tsu, Mie, Japan; Department of Diabetes, Endocrinology and Metabolism, Faculty and Graduate School of Medicine, Mie University, Tsu, Mie, Japan
| | - Corina N D'Alessandro-Gabazza
- Department of Immunology, Faculty and Graduate School of Medicine, Mie University, Tsu, Mie, Japan; Microbiome Research Center, Mie University, Tsu, Mie, Japan; Carl R. Woese Institute for Genomic Biology, University of Illinois at Urbana-Champaign, Urbana, Illinois, USA
| | - Masaaki Toda
- Department of Immunology, Faculty and Graduate School of Medicine, Mie University, Tsu, Mie, Japan
| | - Toshiyuki Ito
- Department of Pulmonary and Critical Care Medicine, Faculty and Graduate School of Medicine, Mie University, Tsu, Mie, Japan
| | - Atsushi Tomaru
- Department of Pulmonary and Critical Care Medicine, Faculty and Graduate School of Medicine, Mie University, Tsu, Mie, Japan
| | - Haruko Saiki
- Department of Pulmonary and Critical Care Medicine, Faculty and Graduate School of Medicine, Mie University, Tsu, Mie, Japan
| | - Tomohito Okano
- Department of Pulmonary and Critical Care Medicine, Faculty and Graduate School of Medicine, Mie University, Tsu, Mie, Japan
| | - Manal A B Alhawsawi
- Carl R. Woese Institute for Genomic Biology, University of Illinois at Urbana-Champaign, Urbana, Illinois, USA; Division of Nutritional Sciences, University of Illinois Urbana-Champaign, Urbana, Illinois, USA
| | - Atsuro Takeshita
- Department of Pulmonary and Critical Care Medicine, Faculty and Graduate School of Medicine, Mie University, Tsu, Mie, Japan; Department of Diabetes, Endocrinology and Metabolism, Faculty and Graduate School of Medicine, Mie University, Tsu, Mie, Japan
| | - Kota Nishihama
- Department of Diabetes, Endocrinology and Metabolism, Faculty and Graduate School of Medicine, Mie University, Tsu, Mie, Japan
| | - Reoto Takei
- Department of Respiratory Medicine and Allergy, Tosei General Hospital, Seto, Aichi, Japan
| | - Yasuhiro Kondoh
- Department of Respiratory Medicine and Allergy, Tosei General Hospital, Seto, Aichi, Japan
| | - Isaac Cann
- Carl R. Woese Institute for Genomic Biology, University of Illinois at Urbana-Champaign, Urbana, Illinois, USA; Division of Nutritional Sciences, University of Illinois Urbana-Champaign, Urbana, Illinois, USA; Department of Animal Science, University of Illinois Urbana-Champaign, Urbana, Illinois, USA; Department of Microbiology, University of Illinois at Urbana-Champaign, Urbana, Illinois, USA
| | - Esteban C Gabazza
- Department of Immunology, Faculty and Graduate School of Medicine, Mie University, Tsu, Mie, Japan; Microbiome Research Center, Mie University, Tsu, Mie, Japan; Carl R. Woese Institute for Genomic Biology, University of Illinois at Urbana-Champaign, Urbana, Illinois, USA.
| | - Tetsu Kobayashi
- Department of Pulmonary and Critical Care Medicine, Faculty and Graduate School of Medicine, Mie University, Tsu, Mie, Japan; Microbiome Research Center, Mie University, Tsu, Mie, Japan
| |
Collapse
|
11
|
Zhu J, Bouzid R, Travert B, Géri G, Cohen Y, Picod A, Heming N, Rottman M, Joly-Laffargue B, Veyradier A, Capron C, Coppo P. Combined coagulation and inflammation markers as predictors of venous thrombo-embolism and death in COVID-19. Front Med (Lausanne) 2024; 11:1399335. [PMID: 38915768 PMCID: PMC11194426 DOI: 10.3389/fmed.2024.1399335] [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: 03/11/2024] [Accepted: 05/03/2024] [Indexed: 06/26/2024] Open
Abstract
Background The COVID-19 pandemic related to SARS-CoV-2 virus was responsible for global pandemic. The severe form of the disease was linked to excessive activation of immune pathways together with a systemic cytokine storm response and thrombotic venous or arterial complications. Factors predicting severe outcomes including venous and/or pulmonary thrombosis (VT) and death were identified, but the prognostic role of their combination was not addressed extensively. Objectives We investigated the role of prognostic factors from the coagulation or inflammatory pathways to better understand the outcome of the disease. Methods For this, we prospectively studied 167 SARS-CoV-2-positive patients from admission in intensive care units (ICU) or emergency departments from four academic hospitals over a 14-month period. Besides standard biology, we assessed serum concentrations of inflammatory markers, coagulation factors and peripheral blood cells immunophenotyping. Results Thirty-nine patients (23.3%) developed VT and 30 patients (18%) died. By univariate analysis, C-reactive protein (CRP) level > 150 mg/L, interleukin-6 (IL-6) ≥ 20 pg/mL, D-dimers > 1,500 μg/L, ADAMTS13 activity ≤ 50%, Von. Conclusion A combination of coagulation and inflammatory markers can refine the prognostication of severe outcome in COVID-19, and could be useful for the initial evaluation of other types of viral infection.
Collapse
Affiliation(s)
- Jaja Zhu
- Service d’Hématologie-Immunologie-Transfusion, AP-HP Paris Saclay, CHU Ambroise Paré, Université de Versailles Saint Quentin-Université Paris Saclay, Montigny-le-Bretonneux, France
- Laboratoire Cellules Souches et Applications Thérapeutiques, UMR INSERM 1184, Commissariat à l’Energie Atomique et aux Energies Alternatives, Fontenay-aux-Roses, France
| | - Raïda Bouzid
- Centre de Référence des Microangiopathies Thrombotiques (CNR-MAT), AP-HP, Paris, France
| | - Benoît Travert
- Service de Médecine Interne, Hôpital Ambroise-Paré, AP-HP, Boulogne-Billancourt, France
| | - Guillaume Géri
- Service de Médecine Intensive et Réanimation, Hôpital Ambroise-Paré, AP-HP, Boulogne-Billancourt, France
| | - Yves Cohen
- Service de Médecine Intensive et Réanimation, Hôpital Avicenne, AP-HP, Paris, France
| | - Adrien Picod
- Service de Médecine Intensive et Réanimation, Hôpital Avicenne, AP-HP, Paris, France
| | - Nicholas Heming
- Department of Intensive Care, Raymond Poincaré Hospital, APHP University Versailles Saint Quentin-University Paris Saclay, Garches, France
- Institut Hospitalo Universitaire PROMETHEUS, Garches, France
- Innovative Biomarkers Plateform, Laboratory of Infection & Inflammation-U1173, School of Medicine, INSERM, University Versailles Saint Quentin-University Paris Saclay, Garches, France
- FHU SEPSIS, Garches, France
| | - Martin Rottman
- Innovative Biomarkers Plateform, Laboratory of Infection & Inflammation-U1173, School of Medicine, INSERM, University Versailles Saint Quentin-University Paris Saclay, Garches, France; FHU SEPSIS, Garches, France
- General Intensive Care Unit, Raymond Poincaré Hospital (AP-HP), FHU SEPSIS, Laboratory of Infection and Inflammation-U1173, School of Medicine Simone Veil, Université Versailles Saint Quentin, University Paris Saclay, INSERM, Garches, France
| | - Bérangère Joly-Laffargue
- Centre de Référence des Microangiopathies Thrombotiques (CNR-MAT), AP-HP, Paris, France
- EA3518, Institut de Recherche Saint Louis, Université de Paris, Paris, France
- Service D’hématologie Biologique, Laboratoire ADAMTS13, Hôpital Lariboisière, AP-HP Nord, Université de Paris, Paris, France
| | - Agnès Veyradier
- Centre de Référence des Microangiopathies Thrombotiques (CNR-MAT), AP-HP, Paris, France
- EA3518, Institut de Recherche Saint Louis, Université de Paris, Paris, France
- Service D’hématologie Biologique, Laboratoire ADAMTS13, Hôpital Lariboisière, AP-HP Nord, Université de Paris, Paris, France
| | - Claude Capron
- Service d’Hématologie-Immunologie-Transfusion, AP-HP Paris Saclay, CHU Ambroise Paré, Université de Versailles Saint Quentin-Université Paris Saclay, Montigny-le-Bretonneux, France
- Université Paris-Saclay, Université de Versailles Saint Quentin en Yvelines (UVSQ), Biomarqueurs en cancérologie et onco-hématologie (BECCOH), Boulogne-Billancourt, France
| | - Paul Coppo
- Centre de Référence des Microangiopathies Thrombotiques (CNR-MAT), AP-HP, Paris, France
- Service d’Hématologie, Hôpital Saint-Antoine, AP-HP-Sorbonne Université, Paris, France
- NSERM UMRS 1138, Centre de Recherche des Cordeliers, Paris, France
| |
Collapse
|
12
|
Zhou P, Guo QQ, Wang FX, Zhou L, Hu HF, Deng Z. Nonlinear relationship between platelet count and 30-day in-hospital mortality in ICU acute respiratory failure patients: a multicenter retrospective cohort study. Eur J Med Res 2024; 29:312. [PMID: 38849948 PMCID: PMC11161993 DOI: 10.1186/s40001-024-01909-1] [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: 02/05/2024] [Accepted: 05/29/2024] [Indexed: 06/09/2024] Open
Abstract
BACKGROUND Limited evidence exists regarding the link between platelet count and 30-day in-hospital mortality in acute respiratory failure (ARF) patients. Thus, this study aims to investigate this association among ICU patients experiencing acute respiratory failure. METHODS We conducted a retrospective cohort study across multiple centers, utilizing data from the US eICU-CRD v2.0 database covering 22,262 patients with ARF in the ICU from 2014 to 2015. Our aim was to investigate the correlation between platelet count and 30-day in-hospital mortality using binary logistic regression, subgroup analyses, and smooth curve fitting. RESULTS The 30-day in-hospital mortality rate was 19.73% (4393 out of 22,262), with a median platelet count of 213 × 109/L. After adjusting for covariates, our analysis revealed an inverse association between platelet count and 30-day in-hospital mortality (OR = 0.99, 95% CI 0.99, 0.99). Subgroup analyses supported the robustness of these findings. Furthermore, a nonlinear relationship was identified between platelet count and 30-day in-hospital mortality, with the inflection point at 120 × 109/L. Below the inflection point, the effect size (OR) was 0.89 (0.87, 0.91), indicating a significant association. However, beyond this point, the relationship was not statistically significant. CONCLUSION This study establishes a clear negative association between platelet count and 30-day in-hospital mortality among ICU patients with ARF. Furthermore, we have identified a nonlinear relationship with saturation effects, indicating that among ICU patients with acute respiratory failure, the lowest 30-day in-hospital mortality rate occurs when the baseline platelet count is approximately 120 × 109/L.
Collapse
Affiliation(s)
- Pan Zhou
- Department of Emergency Medicine, Shenzhen Second People's Hospital/The First Affiliated Hospital of Shenzhen University Health Science Center, Shenzhen, 518035, China
| | - Qin-Qin Guo
- Department of Emergency Medicine, Shenzhen Second People's Hospital/The First Affiliated Hospital of Shenzhen University Health Science Center, Shenzhen, 518035, China
| | - Fang-Xi Wang
- Department of Emergency Medicine, Shenzhen Second People's Hospital/The First Affiliated Hospital of Shenzhen University Health Science Center, Shenzhen, 518035, China
| | - Li Zhou
- Department of Emergency Medicine, Shenzhen Second People's Hospital/The First Affiliated Hospital of Shenzhen University Health Science Center, Shenzhen, 518035, China
| | - Hao-Fei Hu
- Department of Nephrology, Shenzhen Second People's Hospital/The First Affiliated Hospital of Shenzhen University Health Science Center, Shenzhen, 518035, China.
| | - Zhe Deng
- Department of Emergency Medicine, Shenzhen Second People's Hospital/The First Affiliated Hospital of Shenzhen University Health Science Center, Shenzhen, 518035, China.
| |
Collapse
|
13
|
Wang J, Ho P, Nandurkar H, Lim HY. Overall haemostatic potential assay for prediction of outcomes in venous and arterial thrombosis and thrombo-inflammatory diseases. J Thromb Thrombolysis 2024; 57:852-864. [PMID: 38649560 DOI: 10.1007/s11239-024-02975-2] [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: 03/28/2024] [Indexed: 04/25/2024]
Abstract
Thromboembolic diseases including arterial and venous thrombosis are common causes of morbidity and mortality globally. Thrombosis frequently recurs and can also complicate many inflammatory conditions through the process of 'thrombo-inflammation,' as evidenced during the COVID-19 pandemic. Current candidate biomarkers for thrombosis prediction, such as D-dimer, have poor predictive efficacy. This limits our capacity to tailor anticoagulation duration individually and may expose lower risk individuals to undue bleeding risk. Global coagulation assays, such as the Overall Haemostatic Potential (OHP) assay, that investigate fibrin generation and fibrinolysis, may provide a more accurate and functional assessment of hypercoagulability. We present a review of fibrin's critical role as a central modulator of thrombotic risk. The results of our studies demonstrating the OHP assay as a predictive biomarker in venous thromboembolism, chronic renal disease, diabetes mellitus, post-thrombotic syndrome, and COVID-19 are discussed. As a comprehensive and global measurement of fibrin generation and fibrinolytic capacity, the OHP assay may be a valuable addition to future multi-modal predictive tools in thrombosis.
Collapse
Affiliation(s)
- Julie Wang
- Northern Health, 185 Cooper St, Epping, VIC, 3076, Australia.
| | - Prahlad Ho
- Northern Health, 185 Cooper St, Epping, VIC, 3076, Australia
| | - Harshal Nandurkar
- Australian Centre for Blood Diseases, Monash Health, Melbourne, Australia
| | - Hui Yin Lim
- Northern Health, 185 Cooper St, Epping, VIC, 3076, Australia
| |
Collapse
|
14
|
Golden TN, Mani S, Linn RL, Leite R, Trigg NA, Wilson A, Anton L, Mainigi M, Conine CC, Kaufman BA, Strauss JF, Parry S, Simmons RA. Extracellular vesicles alter trophoblast function in pregnancies complicated by COVID-19. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.02.17.580824. [PMID: 38464046 PMCID: PMC10925147 DOI: 10.1101/2024.02.17.580824] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/12/2024]
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection and resulting coronavirus disease (COVID-19) causes placental dysfunction, which increases the risk of adverse pregnancy outcomes. While abnormal placental pathology resulting from COVID-19 is common, direct infection of the placenta is rare. This suggests that pathophysiology associated with maternal COVID-19, rather than direct placental infection, is responsible for placental dysfunction and alteration of the placental transcriptome. We hypothesized that maternal circulating extracellular vesicles (EVs), altered by COVID-19 during pregnancy, contribute to placental dysfunction. To examine this hypothesis, we characterized maternal circulating EVs from pregnancies complicated by COVID-19 and tested their effects on trophoblast cell physiology in vitro . We found that the gestational timing of COVID-19 is a major determinant of circulating EV function and cargo. In vitro trophoblast exposure to EVs isolated from patients with an active infection at the time of delivery, but not EVs isolated from Controls, altered key trophoblast functions including hormone production and invasion. Thus, circulating EVs from participants with an active infection, both symptomatic and asymptomatic cases, can disrupt vital trophoblast functions. EV cargo differed between participants with COVID-19 and Controls, which may contribute to the disruption of the placental transcriptome and morphology. Our findings show that COVID-19 can have effects throughout pregnancy on circulating EVs and circulating EVs are likely to participate in placental dysfunction induced by COVID-19.
Collapse
|
15
|
Becker AP, Mang S, Rixecker T, Lepper PM. [COVID-19 in the intensive care unit]. Pneumologie 2024; 78:330-345. [PMID: 38759701 DOI: 10.1055/a-1854-2693] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/19/2024]
Abstract
The acute respiratory failure as well as ARDS (acute respiratory distress syndrome) have challenged clinicians since the initial description over 50 years ago. Various causes can lead to ARDS and therapeutic approaches for ARDS/ARF are limited to the support or replacement of organ functions and the prevention of therapy-induced consequences. In recent years, triggered by the SARS-CoV-2 pathogen, numerous cases of acute lung failure (C-ARDS) have emerged. The pathophysiological processes of classical ARDS and C-ARDS are essentially similar. In their final stages of inflammation, both lead to a disruption of the blood-air barrier. Treatment strategies for C-ARDS, like classical ARDS, focus on supporting or replacing organ functions and preventing consequential damage. This article summarizes the treatment strategies in the intensive care unit.
Collapse
|
16
|
Hejenkowska ED, Yavuz H, Swiatecka-Urban A. Beyond Borders of the Cell: How Extracellular Vesicles Shape COVID-19 for People with Cystic Fibrosis. Int J Mol Sci 2024; 25:3713. [PMID: 38612524 PMCID: PMC11012075 DOI: 10.3390/ijms25073713] [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: 02/21/2024] [Revised: 03/23/2024] [Accepted: 03/25/2024] [Indexed: 04/14/2024] Open
Abstract
The interaction between extracellular vesicles (EVs) and SARS-CoV-2, the virus causing COVID-19, especially in people with cystic fibrosis (PwCF) is insufficiently studied. EVs are small membrane-bound particles involved in cell-cell communications in different physiological and pathological conditions, including inflammation and infection. The CF airway cells release EVs that differ from those released by healthy cells and may play an intriguing role in regulating the inflammatory response to SARS-CoV-2. On the one hand, EVs may activate neutrophils and exacerbate inflammation. On the other hand, EVs may block IL-6, a pro-inflammatory cytokine associated with severe COVID-19, and protect PwCF from adverse outcomes. EVs are regulated by TGF-β signaling, essential in different disease states, including COVID-19. Here, we review the knowledge, identify the gaps in understanding, and suggest future research directions to elucidate the role of EVs in PwCF during COVID-19.
Collapse
|
17
|
Li M, Song J, Tang X, Bi J, Li Y, Chen C, Feng N, Song Y, Wang L. Critical roles of PAI-1 in lipopolysaccharide-induced acute lung injury. Adv Med Sci 2024; 69:90-102. [PMID: 38387409 DOI: 10.1016/j.advms.2024.01.004] [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: 07/02/2023] [Revised: 10/08/2023] [Accepted: 01/30/2024] [Indexed: 02/24/2024]
Abstract
PURPOSE Plasminogen activator inhibitor-1 (PAI-1) is the main inhibitor of fibrinolytic systems. The effect of PAI-1 on inflammatory response is still inconsistent. Our study was conducted to investigate its effects on inflammation to clarify the role of PAI-1 in acute lung injury (ALI) induced by lipopolysaccharide (LPS). MATERIAL AND METHODS ALI models were established in wild-type (WT) and PAI-1 knockout (KO) mice by LPS intervention for 48 h. Lung histopathology, wet-dry ratio, total cell count and TNF-α concentration in bronchoalveolar lavage fluid (BALF), and inflammation related proteins were detected. Flow cytometry was used to sort neutrophils, macrophages, regulatory T cells (Treg) and T helper cell 17 (Th17). RNA sequencing was performed to find differentially expressed genes. Masson staining and immunohistochemistry were used to analyze pulmonary fiber deposition and proliferation. RESULTS Compared with ALI (WT) group, the wet-dry ratio, the total number of BALF cells, the concentration of TNF-α in BALF, and the expression of pp65 in the lung tissue was increased in ALI (PAI-1 KO) group, with increased proportion of neutrophils, decreased proportion of macrophages and decreased proportion of Treg/Th17 in the lung tissue. Collagen fiber deposition and PCNA expression were lighter in ALI (PAI-1 KO) group than ALI (WT) group. PPI analysis showed that PAI-1 was closely related to TNF, IL-6, IL-1β, Smad2/3 and mainly concentrated in the complement and coagulation system, TNF-α and IL-17 signaling pathways. CONCLUSIONS PAI-1 KO could aggravate ALI induced by LPS at 48 h. PAI-1 may be an important target to improve the prognosis of ALI.
Collapse
Affiliation(s)
- Miao Li
- Shanghai Key Laboratory of Lung Inflammation and Injury, Department of Pulmonary and Critical Care Medicine, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Juan Song
- Shanghai Key Laboratory of Lung Inflammation and Injury, Department of Pulmonary and Critical Care Medicine, Zhongshan Hospital, Fudan University, Shanghai, China; Shanghai Institute of Infectious Diseases and Biosafety, Shanghai, China
| | - Xinjun Tang
- Shanghai Key Laboratory of Lung Inflammation and Injury, Department of Pulmonary and Critical Care Medicine, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Jing Bi
- Shanghai Key Laboratory of Lung Inflammation and Injury, Department of Pulmonary and Critical Care Medicine, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Yufan Li
- Shanghai Key Laboratory of Lung Inflammation and Injury, Department of Pulmonary and Critical Care Medicine, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Cuicui Chen
- Shanghai Key Laboratory of Lung Inflammation and Injury, Department of Pulmonary and Critical Care Medicine, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Nana Feng
- Department of Pulmonary and Critical Care Medicine, Shanghai Eighth People's Hospital, Jiangsu University, Shanghai, China.
| | - Yuanlin Song
- Shanghai Key Laboratory of Lung Inflammation and Injury, Department of Pulmonary and Critical Care Medicine, Zhongshan Hospital, Fudan University, Shanghai, China; Shanghai Institute of Infectious Diseases and Biosafety, Shanghai, China; Shanghai Respiratory Research Institute, Shanghai, China; National Clinical Research Center for Aging and Medicine, Huashan Hospital, Fudan University, Shanghai, China; Key Laboratory of Chemical Injury, Emergency and Critical Medicine of Shanghai Municipal Health Commission, Centre of Emergency and Critical Medicine, Jinshan Hospital of Fudan University, Shanghai, China.
| | - Linlin Wang
- Shanghai Key Laboratory of Lung Inflammation and Injury, Department of Pulmonary and Critical Care Medicine, Zhongshan Hospital, Fudan University, Shanghai, China.
| |
Collapse
|
18
|
Qiu X, Liu M, Wang Q, Zhang Y, Kong L, Zhou L. Thrombosis in Critically Ill Influenza Patients: Incidence and Risk Factors. Clin Appl Thromb Hemost 2024; 30:10760296241278615. [PMID: 39183536 PMCID: PMC11348485 DOI: 10.1177/10760296241278615] [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: 06/25/2024] [Revised: 07/24/2024] [Accepted: 08/12/2024] [Indexed: 08/27/2024] Open
Abstract
Influenza infection is associated with a risk of thrombosis. Whether factors associated with reduced thrombosis might also be associated with reduced risk in patients with severe influenza is unknown. To investigate risk factors associated with thrombosis in patients with severe influenza. We used a cohort data set to identify adults diagnosed with severe influenza. Univariable and multivariable logistic regression models explored potential risk factors for thrombosis events in patients with severe influenza. Cox regression analysis was used to examine the risk factors for mortality in patients with severe influenza. A total of 854 patients with severe influenza were included in the analysis. The incidence of VTE was 9.37% (80/854). Multivariable regression analysis showed that previous aspirin medication (OR: 0.37; 95%CI: 0.14-0.84; P = .029) could reduce the risk factor of thrombosis in patients with severe influenza. Compared with patients in the non-thrombosis group, patients in the thrombosis group required more mechanical ventilation (P < .001), tracheostomy (P < .001), ECMO (P = .046), and high-frequency ventilation (P = .004). The incidence of co-infection was higher in the thrombosis group compared to the non-thrombosis group (P = .025). Univariable Cox regression analysis showed that previous aspirin medication (HR 0.52, 95%CI: 0.33-0.82, P = .005) and previous statin medication (HR 0.54, 95%CI: 0.34-0.87, P = .011) were risk factors for 60-day mortality in patients with severe influenza. Patients with severe influenza are at high risk for thrombosis. The effect of aspirin on thrombosis in patients with severe influenza needs further investigation.
Collapse
Affiliation(s)
- Xianming Qiu
- Department of Critical Care Medicine, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, China
- Shandong Institute of Respiratory Diseases, Jinan, China
- Shandong Institute of Anesthesia and Respiratory Critical Medicine, Jinan, China
| | - Mingjie Liu
- Department of Pulmonary and Critical Care Medicine, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Quanzhen Wang
- Department of Critical Care Medicine, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, China
- Shandong Institute of Respiratory Diseases, Jinan, China
- Shandong Institute of Anesthesia and Respiratory Critical Medicine, Jinan, China
| | - Yuke Zhang
- Department of Critical Care Medicine, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, China
- Shandong Institute of Respiratory Diseases, Jinan, China
- Shandong Institute of Anesthesia and Respiratory Critical Medicine, Jinan, China
| | - Li Kong
- Department of Emergency Center, Shandong University of Traditional Chinese Medicine Affiliated Hospital, Jinan, China
| | - Lei Zhou
- Department of Critical Care Medicine, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, China
- Shandong Institute of Respiratory Diseases, Jinan, China
- Shandong Institute of Anesthesia and Respiratory Critical Medicine, Jinan, China
| |
Collapse
|
19
|
Ludhiadch A, Paul SR, Khan R, Munshi A. COVID-19 induced ischemic stroke and mechanisms of viral entry in brain and clot formation: a systematic review and current update. Int J Neurosci 2023; 133:1153-1166. [PMID: 35412938 DOI: 10.1080/00207454.2022.2056460] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Accepted: 03/16/2022] [Indexed: 10/18/2022]
Abstract
Background: Coronavirus disease 2019, caused by SARS-CoV-2 (SCV-2) was stated as a pandemic on March 11 2020 by World Health Organization (WHO), and since then, it has become a major health issue worldwide. It mainly attacks the respiratory system with various accompanying complications, including cardiac injury, renal failure, encephalitis and Stroke.Materials and Methods: The current systematic review has been compiled to summarize the available literature on SCV-2 induced ischemic Stroke and its subtypes. Further, the mechanisms by which the virus crosses the blood-brain barrier (BBB) to enter the brain have also been explored. The role of CRP and D-dimer as potent prognostic markers was also explored. The literature search was carried out comprehensively on Google scholar, PubMed, SCOP US, Embase and Cochrane databases by following guidelines.Results: All the studies were reviewed thoroughly by authors and disagreements were resolved by consensus and help of the senior authors. The most common subtype of the IS was found to be large artery atherosclerosis in SCV-2 induced IS. Hypertension emerged as the most significant risk factor. The mechanism resulting in elevated levels of CRP and D-dimer have also been discussed. However, there is a scarcity of definitive evidence on how SCV-2 enters the human brain. The available literature based on various studies demonstrated that SCV-2 enters through the nasopharyngeal tract via olfactory cells to olfactory neurons, astrocytes and via choroid plexus through endothelial cells. Further, disruption of gut-brain axis has been also discussed.Conclusion: Data available in the literature is not adequate to come to a conclusion. Therefore, there is a need to carry out further studies to delineate the possible association between SCV-2 induced IS.
Collapse
Affiliation(s)
- Abhilash Ludhiadch
- Department of Human Genetics and Molecular Medicine Central, University of Punjab Bathinda, Bathinda, Punjab, India
| | - Swaraj Ranjan Paul
- Department of Human Genetics and Molecular Medicine Central, University of Punjab Bathinda, Bathinda, Punjab, India
| | - Rahul Khan
- Department of Human Genetics and Molecular Medicine Central, University of Punjab Bathinda, Bathinda, Punjab, India
| | - Anjana Munshi
- Department of Human Genetics and Molecular Medicine Central, University of Punjab Bathinda, Bathinda, Punjab, India
| |
Collapse
|
20
|
Liu L, Zhou C, Jiang H, Wei H, Zhou Y, Zhou C, Ji X. Epidemiology, pathogenesis, and management of Coronavirus disease 2019-associated stroke. Front Med 2023; 17:1047-1067. [PMID: 38165535 DOI: 10.1007/s11684-023-1041-7] [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: 05/05/2023] [Accepted: 10/15/2023] [Indexed: 01/03/2024]
Abstract
The Coronavirus disease 2019 (COVID-19) epidemic has triggered a huge impact on healthcare, socioeconomics, and other aspects of the world over the past three years. An increasing number of studies have identified a complex relationship between COVID-19 and stroke, although active measures are being implemented to prevent disease transmission. Severe COVID-19 may be associated with an increased risk of stroke and increase the rates of disability and mortality, posing a serious challenge to acute stroke diagnosis, treatment, and care. This review aims to provide an update on the influence of COVID-19 itself or vaccines on stroke, including arterial stroke (ischemic stroke and hemorrhagic stroke) and venous stroke (cerebral venous thrombosis). Additionally, the neurovascular mechanisms involved in SARS-CoV-2 infection and the clinical characteristics of stroke in the COVID-19 setting are presented. Evidence on vaccinations, potential therapeutic approaches, and effective strategies for stroke management has been highlighted.
Collapse
Affiliation(s)
- Lu Liu
- Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, 100032, China
- Neurology and Intracranial Hypertension and Cerebral Venous Disease Center, National Health Commission of China, Xuanwu Hospital, Capital Medical University, Beijing, 100032, China
| | - Chenxia Zhou
- Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, 100032, China
- Neurology and Intracranial Hypertension and Cerebral Venous Disease Center, National Health Commission of China, Xuanwu Hospital, Capital Medical University, Beijing, 100032, China
| | - Huimin Jiang
- Beijing Institute of Brain Disorders, Capital Medical University, Beijing, 100069, China
| | - Huimin Wei
- Beijing Advanced Innovation Center for Big Data-Based Precision Medicine, School of Biological Science and Medical Engineering, Beihang University, Beijing, 100191, China
| | - Yifan Zhou
- Beijing Institute of Brain Disorders, Capital Medical University, Beijing, 100069, China
| | - Chen Zhou
- Beijing Institute of Brain Disorders, Capital Medical University, Beijing, 100069, China.
- Department of Neurology, Beijing Shijitan Hospital, Capital Medical University, Beijing, 100038, China.
| | - Xunming Ji
- Neurology and Intracranial Hypertension and Cerebral Venous Disease Center, National Health Commission of China, Xuanwu Hospital, Capital Medical University, Beijing, 100032, China.
- Beijing Institute of Brain Disorders, Capital Medical University, Beijing, 100069, China.
- Department of Neurosurgery, Xuanwu Hospital, Capital Medical University, Beijing, 100032, China.
| |
Collapse
|
21
|
Wang G, Zhuo N, Liu Z. The risk of venous thromboembolism increases within one month of gout flare: comment on the article by Cipolletta et al. Arthritis Rheumatol 2023; 75:1867. [PMID: 37134128 DOI: 10.1002/art.42554] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2023] [Accepted: 04/26/2023] [Indexed: 05/04/2023]
Affiliation(s)
- Gang Wang
- Department of Rheumatology and Immunology, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Ning Zhuo
- Department of Nephrology, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Zhichun Liu
- Department of Rheumatology and Immunology, The Second Affiliated Hospital of Soochow University, Suzhou, China
| |
Collapse
|
22
|
Taquet M, Skorniewska Z, Hampshire A, Chalmers JD, Ho LP, Horsley A, Marks M, Poinasamy K, Raman B, Leavy OC, Richardson M, Elneima O, McAuley HJC, Shikotra A, Singapuri A, Sereno M, Saunders RM, Harris VC, Houchen-Wolloff L, Greening NJ, Mansoori P, Harrison EM, Docherty AB, Lone NI, Quint J, Sattar N, Brightling CE, Wain LV, Evans RE, Geddes JR, Harrison PJ. Acute blood biomarker profiles predict cognitive deficits 6 and 12 months after COVID-19 hospitalization. Nat Med 2023; 29:2498-2508. [PMID: 37653345 PMCID: PMC10579097 DOI: 10.1038/s41591-023-02525-y] [Citation(s) in RCA: 49] [Impact Index Per Article: 24.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2023] [Accepted: 07/31/2023] [Indexed: 09/02/2023]
Abstract
Post-COVID cognitive deficits, including 'brain fog', are clinically complex, with both objective and subjective components. They are common and debilitating, and can affect the ability to work, yet their biological underpinnings remain unknown. In this prospective cohort study of 1,837 adults hospitalized with COVID-19, we identified two distinct biomarker profiles measured during the acute admission, which predict cognitive outcomes 6 and 12 months after COVID-19. A first profile links elevated fibrinogen relative to C-reactive protein with both objective and subjective cognitive deficits. A second profile links elevated D-dimer relative to C-reactive protein with subjective cognitive deficits and occupational impact. This second profile was mediated by fatigue and shortness of breath. Neither profile was significantly mediated by depression or anxiety. Results were robust across secondary analyses. They were replicated, and their specificity to COVID-19 tested, in a large-scale electronic health records dataset. These findings provide insights into the heterogeneous biology of post-COVID cognitive deficits.
Collapse
Affiliation(s)
- Maxime Taquet
- Department of Psychiatry, University of Oxford, Oxford, UK.
- Oxford Health NHS Foundation Trust, Oxford, UK.
| | | | - Adam Hampshire
- Department of Brain Sciences, Imperial College London, London, UK
| | - James D Chalmers
- University of Dundee, Ninewells Hospital and Medical School, Dundee, UK
| | - Ling-Pei Ho
- MRC Human Immunology Unit, University of Oxford, Oxford, UK
| | - Alex Horsley
- Division of Infection, Immunity & Respiratory Medicine, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
- Manchester University NHS Foundation Trust, Manchester, UK
| | - Michael Marks
- Department of Clinical Research, London School of Hygiene & Tropical Medicine, London, UK
- Hospital for Tropical Diseases, University College London Hospital, London, UK
- Division of Infection and Immunity, University College London, London, UK
| | | | - Betty Raman
- Radcliffe Department of Medicine, University of Oxford, Oxford, UK
- Oxford University Hospitals NHS Foundation Trust, Oxford, UK
| | - Olivia C Leavy
- Department of Population Health Sciences, University of Leicester, Leicester, UK
- The institute for Lung Health, NIHR Leicester Biomedical Research Centre, University of Leicester, Leicester, UK
| | - Matthew Richardson
- The institute for Lung Health, NIHR Leicester Biomedical Research Centre, University of Leicester, Leicester, UK
| | - Omer Elneima
- The institute for Lung Health, NIHR Leicester Biomedical Research Centre, University of Leicester, Leicester, UK
| | - Hamish J C McAuley
- The institute for Lung Health, NIHR Leicester Biomedical Research Centre, University of Leicester, Leicester, UK
| | - Aarti Shikotra
- NIHR Leicester Biomedical Research Centre, University of Leicester, Leicester, UK
| | - Amisha Singapuri
- The institute for Lung Health, NIHR Leicester Biomedical Research Centre, University of Leicester, Leicester, UK
| | - Marco Sereno
- The institute for Lung Health, NIHR Leicester Biomedical Research Centre, University of Leicester, Leicester, UK
| | - Ruth M Saunders
- The institute for Lung Health, NIHR Leicester Biomedical Research Centre, University of Leicester, Leicester, UK
| | - Victoria C Harris
- The institute for Lung Health, NIHR Leicester Biomedical Research Centre, University of Leicester, Leicester, UK
- University Hospitals of Leicester NHS Trust, Leicester, UK
| | - Linzy Houchen-Wolloff
- Centre for Exercise and Rehabilitation Science, NIHR Leicester Biomedical Research Centre-Respiratory, University of Leicester, Leicester, UK
- Department of Respiratory Sciences, University of Leicester, Leicester, UK
- Therapy Department, University Hospitals of Leicester, NHS Trust, Leicester, UK
| | - Neil J Greening
- The institute for Lung Health, NIHR Leicester Biomedical Research Centre, University of Leicester, Leicester, UK
| | | | - Ewen M Harrison
- Centre for Medical Informatics, The Usher Institute, University of Edinburgh, Edinburgh, UK
| | - Annemarie B Docherty
- Centre for Medical Informatics, The Usher Institute, University of Edinburgh, Edinburgh, UK
| | - Nazir I Lone
- Usher Institute, University of Edinburgh, Edinburgh, UK
- Royal Infirmary of Edinburgh, NHS Lothian, Edinburgh, UK
| | | | - Naveed Sattar
- School of Cardiovascular and Metabolic Health, University of Glasgow, Glasgow, UK
| | - Christopher E Brightling
- The institute for Lung Health, NIHR Leicester Biomedical Research Centre, University of Leicester, Leicester, UK
| | - Louise V Wain
- Department of Population Health Sciences, University of Leicester, Leicester, UK
- The institute for Lung Health, NIHR Leicester Biomedical Research Centre, University of Leicester, Leicester, UK
| | - Rachael E Evans
- The institute for Lung Health, NIHR Leicester Biomedical Research Centre, University of Leicester, Leicester, UK
- University Hospitals of Leicester NHS Trust, Leicester, UK
| | - John R Geddes
- Department of Psychiatry, University of Oxford, Oxford, UK
- Oxford Health NHS Foundation Trust, Oxford, UK
| | - Paul J Harrison
- Department of Psychiatry, University of Oxford, Oxford, UK.
- Oxford Health NHS Foundation Trust, Oxford, UK.
| |
Collapse
|
23
|
Brinjikji W, Kallmes DF, Virmani R, de Meyer SF, Yoo AJ, Humphries W, Zaidat OO, Teleb MS, Jones JG, Siddiqui AH, Andersson T, Nogueira RG, Gil SM, Douglas A, Rossi R, Rentzos A, Ceder E, Carlqvist J, Dunker D, Jood K, Tatlisumak T, Doyle KM. Endotheliitis and cytokine storm as a mechanism of clot formation in COVID-19 ischemic stroke patients: A histopathologic study of retrieved clots. Interv Neuroradiol 2023:15910199231185804. [PMID: 37769315 DOI: 10.1177/15910199231185804] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/30/2023] Open
Abstract
BACKGROUND Studies during the COVID-19 pandemic have demonstrated an association between COVID-19 virus infection and the development of acute ischemic stroke, particularly large vessel occlusion (LVO). Studying the characteristics and immunohistochemistry of retrieved stroke emboli during mechanical thrombectomy for LVO may offer insights into the pathogenesis of LVO in COVID-19 patients. We examined retrieved COVID-19 emboli from the STRIP, EXCELLENT, and RESTORE registries and compared their characteristics to a control group. METHODS We identified COVID-positive LVO patients from the STRIP, RESTORE, and EXCELLENT studies who underwent mechanical thrombectomy. These patients were matched to a control group controlling for stroke etiology based on Trial of Org 10172 in Acute Stroke Treatment criteria. All clots were stained with Martius Scarlet Blue (MSB) along with immunohistochemistry for interleukin-6 (IL-6), C-reactive protein (CRP), von Willebrand factor (vWF), CD66b, fibrinogen, and citrullinated Histone H3. Clot composition was compared between groups. RESULTS Nineteen COVID-19-positive patients and 38 controls were included. COVID-19-positive patients had a significantly higher percentage of CRP and vWF. There was no difference in IL-6, fibrin, CD66b, or citrullinated Histone H3 between groups. Based on MSB staining, there was no statistically significant difference regarding the percentage of red blood cells, white blood cells, fibrin, and platelets. CONCLUSIONS Our study found higher concentrations of CRP and vWF in retrieved clots of COVID-19-positive stroke patients compared to COVID-19-negative controls. These findings support the potential role of systemic inflammation as indicated by elevated CRP and endothelial injury as indicated by elevated vWF as precipitating factors in thrombus development in these patients.
Collapse
Affiliation(s)
| | | | - Renu Virmani
- Department of Pathology, CVPath Institute, Gaithersburg, MD, USA
| | - Simon F de Meyer
- Laboratory for Thrombosis Research, KU Leuven Campus Kulak Kortrijk, Kortrijk, Belgium
| | - Albert J Yoo
- Neurointervention, Texas Stroke Institute, Plano, TX, USA
| | | | - Osama O Zaidat
- Department of Neuroscience, Mercy Health St Vincent Medical Center, Toledo, OH, USA
| | - Mohamed S Teleb
- Neurointerventional Surgery, Stroke, and Neurocritical Care, Banner Health, Mesa, AZ, USA
| | - Jesse G Jones
- Department of Neurosurgery, University of Alabama, Birmingham, AL, USA
| | - Adnan H Siddiqui
- Departments of Neurosurgery and Radiology, State University of New York at Buffalo, New York, NY, USA
| | - Tommy Andersson
- Department of Neuroradiology, Karolinska University Hospital and Karolinska Institutet, Stockholm, Sweden
| | - Raul G Nogueira
- Department of Neurology and Neurosurgery, University of Pittsburgh Medical Center, UPMC Stroke Institute, Pittsburgh, PA, USA
| | - Sara Molina Gil
- Department of Physiology and Galway Neuroscience Centre, School of Medicine, National University of Ireland, Galway, Ireland
- CÚRAM-SFI Research Centre for Medical Devices, National University of Ireland Galway, Galway, Ireland
| | - Andrew Douglas
- Department of Physiology and Galway Neuroscience Centre, School of Medicine, National University of Ireland, Galway, Ireland
- CÚRAM-SFI Research Centre for Medical Devices, National University of Ireland Galway, Galway, Ireland
| | - Rosanna Rossi
- Department of Physiology and Galway Neuroscience Centre, School of Medicine, National University of Ireland, Galway, Ireland
- CÚRAM-SFI Research Centre for Medical Devices, National University of Ireland Galway, Galway, Ireland
| | - Alexander Rentzos
- Department of Interventional and Diagnostic Neuroradiology, Sahlgrenska University Hospital, University of Gothenburg, Gothenburg, Sweden
| | - Erik Ceder
- Department of Interventional and Diagnostic Neuroradiology, Sahlgrenska University Hospital, University of Gothenburg, Gothenburg, Sweden
| | - Jeanette Carlqvist
- Department of Interventional and Diagnostic Neuroradiology, Sahlgrenska University Hospital, University of Gothenburg, Gothenburg, Sweden
| | - Dennis Dunker
- Department of Interventional and Diagnostic Neuroradiology, Sahlgrenska University Hospital, University of Gothenburg, Gothenburg, Sweden
| | - Katarina Jood
- Department of Clinical Neurosciences, Institute of Neuroscience and Physiology, Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden
- Department of Neurology, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Turgut Tatlisumak
- Department of Clinical Neurosciences, Institute of Neuroscience and Physiology, Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden
- Department of Neurology, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Karen M Doyle
- Department of Physiology and Galway Neuroscience Centre, School of Medicine, National University of Ireland, Galway, Ireland
- CÚRAM-SFI Research Centre for Medical Devices, National University of Ireland Galway, Galway, Ireland
| |
Collapse
|
24
|
Abudouleh E, Alhamlan F, Al-Qahtani AA, Bohol MF, Al Hazzani A, Khorfan K, Alkaff M, Owaidah T, Al-Qahtani AA. Changes in the Fibrinolytic System of Patients Infected with Severe Acute Respiratory Syndrome Coronavirus 2. J Clin Med 2023; 12:5223. [PMID: 37629265 PMCID: PMC10455675 DOI: 10.3390/jcm12165223] [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: 05/21/2023] [Revised: 06/15/2023] [Accepted: 06/27/2023] [Indexed: 08/27/2023] Open
Abstract
INTRODUCTION In this study, coagulation and fibrinolysis parameters and their association with disease severity were investigated in coronavirus disease (COVID-19) patients. MATERIALS AND METHODS COVID-19 patients (n = 446) admitted to our institute between 21 February 2021 and 17 March 2022, were recruited. Clinical data and staging were collected from all patients. Blood samples were collected and analyzed for several parameters of fibrinolysis and coagulation, including alpha-2-antiplasmin(α2AP) and plasminogen, thrombin activatable fibrinolysis inhibitor (TAFI), tissue plasminogen activator (tPA), plasminogen activator inhibitor-1 (PAI-1), D-dimer, and fibrinogen levels. RESULTS The TAFI, fibrinogen, and tPA levels were significantly higher in participants who died compared to that of patients who recovered (p < 0.001). However, PAI-1, tPA, and TAFI were significantly higher in patients admitted to the ICU than those of the healthy controls (p < 0.001 for PAI-1 and tPA; p = 0.0331 for TAFI). Our results showed that stage C and D COVID-19 patients had significantly higher levels of PAI-1 (p = 0.003). Furthermore, stage D COVID-19 patients had significantly higher tPA and TAFI values (p = 0.003). CONCLUSIONS Hypofibrinolysis was the most prevalent condition among patients with severe COVID-19. In this study, several coagulation markers were elevated, making them suitable prognostic markers for hypofibrinolysis.
Collapse
Affiliation(s)
- Esra’a Abudouleh
- Department of Botany and Microbiology, College of Science, King Saud University, Riyadh 11451, Saudi Arabia; (E.A.); (A.A.H.)
| | - Fatimah Alhamlan
- Department of Infection and Immunity, Research Centre, King Faisal Specialist Hospital & Research Centre, Riyadh 11211, Saudi Arabia; (F.A.); (M.F.B.)
- Department of Microbiology and Immunology, College of Medicine, Alfaisal University, Riyadh 11533, Saudi Arabia
| | - Arwa A. Al-Qahtani
- Department of Family Medicine, College of Medicine, Al-Imam Mohammad Ibn Saud Islamic University, Riyadh 13317, Saudi Arabia;
| | - Marie Fe Bohol
- Department of Infection and Immunity, Research Centre, King Faisal Specialist Hospital & Research Centre, Riyadh 11211, Saudi Arabia; (F.A.); (M.F.B.)
| | - Amal Al Hazzani
- Department of Botany and Microbiology, College of Science, King Saud University, Riyadh 11451, Saudi Arabia; (E.A.); (A.A.H.)
| | - Khadija Khorfan
- Department of Pathology and Laboratory Medicine, King Faisal Specialist Hospital and Research Center, Riyadh 11211, Saudi Arabia; (K.K.); (M.A.)
| | - Morad Alkaff
- Department of Pathology and Laboratory Medicine, King Faisal Specialist Hospital and Research Center, Riyadh 11211, Saudi Arabia; (K.K.); (M.A.)
| | - Tarek Owaidah
- Department of Pathology and Laboratory Medicine, King Faisal Specialist Hospital and Research Center, Riyadh 11211, Saudi Arabia; (K.K.); (M.A.)
- Department of Pathology, College of Medicine, Alfaisal University, Riyadh 11533, Saudi Arabia
| | - Ahmed A. Al-Qahtani
- Department of Infection and Immunity, Research Centre, King Faisal Specialist Hospital & Research Centre, Riyadh 11211, Saudi Arabia; (F.A.); (M.F.B.)
- Department of Microbiology and Immunology, College of Medicine, Alfaisal University, Riyadh 11533, Saudi Arabia
| |
Collapse
|
25
|
Grubišić B, Švitek L, Ormanac K, Sabo D, Mihaljević I, Bilić-Ćurčić I, Omanović Kolarić T. Molecular Mechanisms Responsible for Diabetogenic Effects of COVID-19 Infection-Induction of Autoimmune Dysregulation and Metabolic Disturbances. Int J Mol Sci 2023; 24:11576. [PMID: 37511334 PMCID: PMC10380525 DOI: 10.3390/ijms241411576] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2023] [Revised: 07/16/2023] [Accepted: 07/16/2023] [Indexed: 07/30/2023] Open
Abstract
The COVID-19 pandemic has revealed a significant association between SARS-CoV-2 infection and diabetes, whereby individuals with diabetes are more susceptible to severe disease and higher mortality rates. Interestingly, recent findings suggest a reciprocal relationship between COVID-19 and diabetes, wherein COVID-19 may contribute to developing new-onset diabetes and worsen existing metabolic abnormalities. This narrative review aims to shed light on the intricate molecular mechanisms underlying the diabetogenic effects of COVID-19. Specifically, the review explores the potential role of various factors, including direct damage to β-cells, insulin resistance triggered by systemic inflammation, and disturbances in hormonal regulation, aiming to enhance our understanding of the COVID-19 impact on the development and progression of diabetes. By analysing these mechanisms, the aim is to enhance our understanding of the impact of COVID-19 on the development and progression of diabetes. The binding of SARS-CoV-2 to angiotensin-converting enzyme 2 (ACE2) receptors, which are present in key metabolic organs and tissues, may interfere with glucometabolic pathways, leading to hyperglycaemia, and potentially contribute to the development of new disease mechanisms. The virus's impact on β-cells through direct invasion or systemic inflammation may induce insulin resistance and disrupt glucose homeostasis. Furthermore, glucocorticoids, commonly used to treat COVID-19, may exacerbate hyperglycaemia and insulin resistance, potentially contributing to new-onset diabetes. The long-term effects of COVID-19 on glucose metabolism are still unknown, necessitating further research into the possibility of developing a novel type of diabetes. This article provides a comprehensive overview of the current understanding of the interaction between COVID-19 and diabetes, highlighting potential areas for future research and therapeutic interventions.
Collapse
Affiliation(s)
- Barbara Grubišić
- Department of Infectious Diseases, University Hospital Centre Osijek, 4 Josip Huttler Street, HR-31000 Osijek, Croatia
- Faculty of Medicine Osijek, J. J. Strossmayer University of Osijek, 4 Josip Huttler Street, HR-31000 Osijek, Croatia
| | - Luka Švitek
- Department of Infectious Diseases, University Hospital Centre Osijek, 4 Josip Huttler Street, HR-31000 Osijek, Croatia
- Faculty of Medicine Osijek, J. J. Strossmayer University of Osijek, 4 Josip Huttler Street, HR-31000 Osijek, Croatia
| | - Klara Ormanac
- Department of Pharmacology, Faculty of Medicine Osijek, J. J. Strossmayer University of Osijek, 4 Josip Huttler Street, HR-31000 Osijek, Croatia
| | - Dea Sabo
- Department of Pharmacology, Faculty of Medicine Osijek, J. J. Strossmayer University of Osijek, 4 Josip Huttler Street, HR-31000 Osijek, Croatia
| | - Ivica Mihaljević
- Clinical Institute of Nuclear Medicine and Radiation Protection, University Hospital Centre Osijek, 4 Josip Huttler Street, HR-31000 Osijek, Croatia
- Department for Nuclear Medicine and Oncology, Faculty of Medicine, J. J. Strossmayer University of Osijek, 4 Josip Huttler Street, HR-31000 Osijek, Croatia
- Academy of Medical Sciences of Croatia, 15 Kaptol Street, HR-10000 Zagreb, Croatia
| | - Ines Bilić-Ćurčić
- Department of Pharmacology, Faculty of Medicine Osijek, J. J. Strossmayer University of Osijek, 4 Josip Huttler Street, HR-31000 Osijek, Croatia
- Department of Endocrinology and Metabolism Disorders, Internal Medicine Clinic, University Hospital Centre Osijek, 4 Josip Huttler Street, HR-31000 Osijek, Croatia
| | - Tea Omanović Kolarić
- Department of Pharmacology, Faculty of Medicine Osijek, J. J. Strossmayer University of Osijek, 4 Josip Huttler Street, HR-31000 Osijek, Croatia
- Faculty of Dental Medicine and Health Osijek, University of Osijek, 21 Crkvena Street, HR-31000 Osijek, Croatia
| |
Collapse
|
26
|
Nunes JM, Kell DB, Pretorius E. Cardiovascular and haematological pathology in myalgic encephalomyelitis/chronic fatigue syndrome (ME/CFS): A role for viruses. Blood Rev 2023; 60:101075. [PMID: 36963989 PMCID: PMC10027292 DOI: 10.1016/j.blre.2023.101075] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2023] [Revised: 03/15/2023] [Accepted: 03/16/2023] [Indexed: 03/25/2023]
Abstract
ME/CFS is a debilitating chronic condition that often develops after viral or bacterial infection. Insight from the study of Long COVID/Post Acute Sequelae of COVID-19 (PASC), the post-viral syndrome associated with SARS-CoV-2 infection, might prove to be useful for understanding pathophysiological mechanisms of ME/CFS. Disease presentation is similar between the two conditions, and a subset of Long COVID patients meet the diagnostic criteria for ME/CFS. Since Long COVID is characterized by significant vascular pathology - including endothelial dysfunction, coagulopathy, and vascular dysregulation - the question of whether or not the same biological abnormalities are of significance in ME/CFS arises. Cardiac abnormalities have for a while now been documented in ME/CFS cohorts, with recent studies demonstrating major deficits in cerebral blood flow, and hence vascular dysregulation. A growing body of research is demonstrating that ME/CFS is accompanied by platelet hyperactivation, anomalous clotting, a procoagulant phenotype, and endothelial dysfunction. Endothelial damage and dysregulated clotting can impair substance exchange between blood and tissues, and result in hypoperfusion, which may contribute to the manifestation of certain ME/CFS symptoms. Here we review the ME/CFS literature to summarize cardiovascular and haematological findings documented in patients with the condition, and, in this context, briefly discuss the potential role of previously-implicated pathogens. Overall, cardiac and haematological abnormalities are present within ME/CFS cohorts. While atherosclerotic heart disease is not significantly associated with ME/CFS, suboptimal cardiovascular function defined by reduced cardiac output, impaired cerebral blood flow, and vascular dysregulation are, and these abnormalities do not appear to be influenced by deconditioning. Rather, these cardiac abnormalities may result from dysfunction in the (autonomic) nervous system. Plenty of recently published studies are demonstrating significant platelet hyperactivity and endothelial dysfunction in ME/CFS, as well as anomalous clotting processes. It is of particular importance to determine to what extent these cardiovascular and haematological abnormalities contribute to symptom severity, and if these two systems can be targeted for therapeutic purposes. Viral reservoirs of herpesviruses exist in ME/CFS, and most likely contribute to cardiovascular and haematological dysfunction directly or indirectly. This review highlights the potential of studying cardiac functioning, the vasculature, and coagulation system in ME/CFS.
Collapse
Affiliation(s)
- Jean M Nunes
- Department of Physiological Sciences, Faculty of Science, Stellenbosch University, Stellenbosch, Private Bag X1, Matieland 7602, South Africa.
| | - Douglas B Kell
- Department of Physiological Sciences, Faculty of Science, Stellenbosch University, Stellenbosch, Private Bag X1, Matieland 7602, South Africa; Department of Biochemistry and Systems Biology, Institute of Systems, Molecular and Integrative Biology, Faculty of Health and Life Sciences, University of Liverpool, Crown St, Liverpool L69 7ZB, UK; The Novo Nordisk Foundation Centre for Biosustainability, Building 220, Chemitorvet 200, Technical University of Denmark, 2800 Kongens Lyngby, Denmark.
| | - Etheresia Pretorius
- Department of Physiological Sciences, Faculty of Science, Stellenbosch University, Stellenbosch, Private Bag X1, Matieland 7602, South Africa; Department of Biochemistry and Systems Biology, Institute of Systems, Molecular and Integrative Biology, Faculty of Health and Life Sciences, University of Liverpool, Crown St, Liverpool L69 7ZB, UK.
| |
Collapse
|
27
|
McManus D, Davis MW, Ortiz A, Britto-Leon C, Dela Cruz CS, Topal JE. Immunomodulatory Agents for Coronavirus Disease-2019 Pneumonia. Clin Chest Med 2023; 44:299-319. [PMID: 37085221 PMCID: PMC9678826 DOI: 10.1016/j.ccm.2022.11.009] [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] [Indexed: 11/23/2022]
Abstract
Morbidity and mortality from COVID-19 is due to severe inflammation and end-organ damage caused by a hyperinflammatory response. Multiple immunomodulatory agents to attenuate this response have been studied. Corticosteroids, specifically dexamethasone, have been shown to reduce mortality in hospitalized patients who require supplemental oxygen. Interleukin-6 antagonist, tocilizimab, and Janus kinase inhibitors have also been shown to reduce mortality. However, patients who have severe pulmonary end-organ damage requiring mechanical ventilation or extracorporeal membrane oxygenation appear not to benefit from immunomodulatory therapies. This highlights the importance of appropriate timing to initiate immunomodulatory therapies in the management of severe COVID-19 disease.
Collapse
Affiliation(s)
- Dayna McManus
- Department of Pharmacy Services, Yale New Haven Hospital, 20 York Street, New Haven, CT 06510, USA.
| | - Matthew W Davis
- Department of Pharmacy Services, Yale New Haven Hospital, 20 York Street, New Haven, CT 06510, USA
| | - Alex Ortiz
- Pulmonary, Critical Care & Sleep Medicine, 300 Cedar Street, P.O. Box 208057, New Haven, CT 06520-8057, USA
| | - Clemente Britto-Leon
- Pulmonary, Critical Care & Sleep Medicine, 300 Cedar Street, P.O. Box 208057, New Haven, CT 06520-8057, USA
| | - Charles S Dela Cruz
- Pulmonary, Critical Care & Sleep Medicine, 300 Cedar Street, P.O. Box 208057, New Haven, CT 06520-8057, USA
| | - Jeffrey E Topal
- Department of Pharmacy Services, Yale New Haven Hospital, 20 York Street, New Haven, CT 06510, USA.
| |
Collapse
|
28
|
Hassan AHE, El-Sayed SM, Yamamoto M, Gohda J, Matsumoto T, Shirouzu M, Inoue JI, Kawaguchi Y, Mansour RMA, Anvari A, Farahat AA. In Silico and In Vitro Evaluation of Some Amidine Derivatives as Hit Compounds towards Development of Inhibitors against Coronavirus Diseases. Viruses 2023; 15:1171. [PMID: 37243257 PMCID: PMC10223987 DOI: 10.3390/v15051171] [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: 03/09/2023] [Revised: 05/04/2023] [Accepted: 05/12/2023] [Indexed: 05/28/2023] Open
Abstract
Coronaviruses, including SARS-CoV-2, SARS-CoV, MERS-CoV and influenza A virus, require the host proteases to mediate viral entry into cells. Rather than targeting the continuously mutating viral proteins, targeting the conserved host-based entry mechanism could offer advantages. Nafamostat and camostat were discovered as covalent inhibitors of TMPRSS2 protease involved in viral entry. To circumvent their limitations, a reversible inhibitor might be required. Considering nafamostat structure and using pentamidine as a starting point, a small set of structurally diverse rigid analogues were designed and evaluated in silico to guide selection of compounds to be prepared for biological evaluation. Based on the results of in silico study, six compounds were prepared and evaluated in vitro. At the enzyme level, compounds 10-12 triggered potential TMPRSS2 inhibition with low micromolar IC50 concentrations, but they were less effective in cellular assays. Meanwhile, compound 14 did not trigger potential TMPRSS2 inhibition at the enzyme level, but it showed potential cellular activity regarding inhibition of membrane fusion with a low micromolar IC50 value of 10.87 µM, suggesting its action could be mediated by another molecular target. Furthermore, in vitro evaluation showed that compound 14 inhibited pseudovirus entry as well as thrombin and factor Xa. Together, this study presents compound 14 as a hit compound that might serve as a starting point for developing potential viral entry inhibitors with possible application against coronaviruses.
Collapse
Affiliation(s)
- Ahmed H E Hassan
- Department of Medicinal Chemistry, Faculty of Pharmacy, Mansoura University, Mansoura 35516, Egypt
| | - Selwan M El-Sayed
- Department of Medicinal Chemistry, Faculty of Pharmacy, Mansoura University, Mansoura 35516, Egypt
| | - Mizuki Yamamoto
- Research Center for Asian Infectious Diseases, Institute of Medical Science, The University of Tokyo, Tokyo 108-8639, Japan
| | - Jin Gohda
- Research Center for Asian Infectious Diseases, Institute of Medical Science, The University of Tokyo, Tokyo 108-8639, Japan
| | - Takehisa Matsumoto
- Drug Discovery Structural Biology Platform Unit, RIKEN Center for Biosystems Dynamics Research, Kanagawa 230-0045, Japan
| | - Mikako Shirouzu
- Drug Discovery Structural Biology Platform Unit, RIKEN Center for Biosystems Dynamics Research, Kanagawa 230-0045, Japan
| | - Jun-Ichiro Inoue
- Infection and Advanced Research Center (UTOPIA), The University of Tokyo Pandemic Preparedness, Tokyo 108-8639, Japan
| | - Yasushi Kawaguchi
- Research Center for Asian Infectious Diseases, Institute of Medical Science, The University of Tokyo, Tokyo 108-8639, Japan
- Division of Molecular Virology, Department of Microbiology and Immunology, The Institute of Medical Science, The University of Tokyo, Tokyo 108-8639, Japan
| | - Reem M A Mansour
- Department of Medicinal Chemistry, Faculty of Pharmacy, Mansoura University, Mansoura 35516, Egypt
| | - Abtin Anvari
- Master of Pharmaceutical Sciences Program, California Northstate University, 9700 W Taron Dr., Elk Grove, CA 95757, USA
| | - Abdelbasset A Farahat
- Master of Pharmaceutical Sciences Program, California Northstate University, 9700 W Taron Dr., Elk Grove, CA 95757, USA
- Department of Pharmaceutical Organic Chemistry, Faculty of Pharmacy, Mansoura University, Mansoura 35516, Egypt
| |
Collapse
|
29
|
Vassiliou AG, Vrettou CS, Keskinidou C, Dimopoulou I, Kotanidou A, Orfanos SE. Endotheliopathy in Acute COVID-19 and Long COVID. Int J Mol Sci 2023; 24:8237. [PMID: 37175942 PMCID: PMC10179170 DOI: 10.3390/ijms24098237] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2023] [Revised: 04/28/2023] [Accepted: 04/30/2023] [Indexed: 05/15/2023] Open
Abstract
The pulmonary endothelium is a highly regulated organ that performs a wide range of functions under physiological and pathological conditions. Since endothelial dysfunction has been demonstrated to play a direct role in sepsis and acute respiratory distress syndrome, its role in COVID-19 has also been extensively investigated. Indeed, apart from the COVID-19-associated coagulopathy biomarkers, new biomarkers were recognised early during the pandemic, including markers of endothelial cell activation or injury. We systematically searched the literature up to 10 March 2023 for studies examining the association between acute and long COVID-19 severity and outcomes and endothelial biomarkers.
Collapse
Affiliation(s)
- Alice G. Vassiliou
- First Department of Critical Care Medicine & Pulmonary Services, School of Medicine, National and Kapodistrian University of Athens, Evangelismos Hospital, 106 76 Athens, Greece; (C.S.V.); (C.K.); (I.D.); (A.K.)
| | | | | | | | | | - Stylianos E. Orfanos
- First Department of Critical Care Medicine & Pulmonary Services, School of Medicine, National and Kapodistrian University of Athens, Evangelismos Hospital, 106 76 Athens, Greece; (C.S.V.); (C.K.); (I.D.); (A.K.)
| |
Collapse
|
30
|
Hernández-Díazcouder A, Díaz-Godínez C, Carrero JC. Extracellular vesicles in COVID-19 prognosis, treatment, and vaccination: an update. Appl Microbiol Biotechnol 2023; 107:2131-2141. [PMID: 36917275 PMCID: PMC10012322 DOI: 10.1007/s00253-023-12468-6] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2022] [Revised: 02/27/2023] [Accepted: 03/02/2023] [Indexed: 03/16/2023]
Abstract
The lethality of the COVID 19 pandemic became the trigger for one of the most meteoric races on record in the search for strategies of disease control. Those include development of rapid and sensitive diagnostic methods, therapies to treat severe cases, and development of anti-SARS-CoV-2 vaccines, the latter responsible for the current relative control of the disease. However, the commercially available vaccines are still far from conferring protection against acquiring the infection, so the development of more efficient vaccines that can cut the transmission of the variants of concerns that currently predominate and those that will emerge is a prevailing need. On the other hand, considering that COVID 19 is here to stay, the development of new diagnosis and treatment strategies is also desirable. In this sense, there has recently been a great interest in taking advantage of the benefits offered by extracellular vesicles (EVs), membrane structures of nanoscale size that carry information between cells participating in this manner in many physiological homeostatic and pathological processes. The interest has been focused on the fact that EVs are relatively easy to obtain and manipulate, allowing the design of natural nanocarriers that deliver molecules of interest, as well as the information about the pathogens, which can be exploited for the aforementioned purposes. Studies have shown that infection with SARS-CoV-2 induces the release of EVs from different sources, including platelets, and that their increase in blood, as well as some of their markers, could be used as a prognosis of disease severity. Likewise, EVs from different sources are being used as the ideal carriers for delivering active molecules and drugs to treat the disease, as well as vaccine antigens. In this review, we describe the progress that has been made in these three years of pandemic regarding the use of EVs for diagnosis, treatment, and vaccination against SARS-CoV-2 infection. KEY POINTS: • Covid-19 still requires more effective and specific treatments and vaccines. • The use of extracellular vesicles is emerging as an option with multiple advantages. • Association of EVs with COVID 19 and engineered EVs for its control are presented.
Collapse
Affiliation(s)
- Adrián Hernández-Díazcouder
- Departamento de Inmunología, Instituto Nacional de Cardiología Ignacio Chávez, Ciudad de México, México
- Departamento de Ciencias de La Salud, Universidad Tecnológica de México (UNITEC), Estado de México, Los Reyes, México
| | - César Díaz-Godínez
- Departamento de Ciencias de La Salud, Universidad Tecnológica de México (UNITEC), Estado de México, Los Reyes, México
- Departamento de Inmunología, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México (UNAM), 04510, Ciudad de México, México
| | - Julio César Carrero
- Departamento de Inmunología, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México (UNAM), 04510, Ciudad de México, México.
| |
Collapse
|
31
|
Qazi M, Amin M, Khatri M. Letter to the Editor: Comment On: High incidence of pulmonary thromboembolism in hospitalized SARS-CoV-2 infected patients despite thrombo-prophylaxis. Heart Lung 2023:S0147-9563(23)00087-0. [PMID: 37012126 PMCID: PMC10036303 DOI: 10.1016/j.hrtlng.2023.03.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2023] [Accepted: 03/22/2023] [Indexed: 03/26/2023]
Affiliation(s)
- Maida Qazi
- Dow University of Health Sciences, Bihar Colony, Karachi, Pakistan.
| | - Mahnoor Amin
- Dow University of Health Sciences Karachi, Mosalane, Karachi, Pakistan
| | - Mahima Khatri
- Dow University of Health Sciences Karachi, Mosalane, Karachi, Pakistan
| |
Collapse
|
32
|
Girard TJ, Antunes L, Zhang N, Amrute JM, Subramanian R, Eldem I, Remy KE, Mazer M, Erlich EC, Cruchaga C, Steed AL, Randolph GJ, Di Paola J. Peripheral blood mononuclear cell tissue factor (F3 gene) transcript levels and circulating extracellular vesicles are elevated in severe coronavirus 2019 (COVID-19) disease. J Thromb Haemost 2023; 21:629-638. [PMID: 36696180 PMCID: PMC9773443 DOI: 10.1016/j.jtha.2022.11.033] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2022] [Revised: 10/20/2022] [Accepted: 11/01/2022] [Indexed: 01/26/2023]
Abstract
BACKGROUND Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection is associated with excessive coagulation, thrombosis, and mortality. OBJECTIVE To provide insight into mechanisms that contribute to excessive coagulation in coronavirus 2019 (COVID-19) disease. PATIENTS/METHODS Blood from COVID-19 patients was investigated for coagulation-related gene expression and functional activities. RESULTS Single-cell RNA sequencing (scRNA-seq) of peripheral blood mononuclear cells from severe COVID-19 patients revealed a 5.2-fold increase in tissue factor (TF [F3 gene]) transcript expression levels (P < .05), the trigger of extrinsic coagulation; a 7.7-fold increase in C1-inhibitor (SERPING1 gene; P < .01) transcript expression levels, an inhibitor of intrinsic coagulation; and a 4.4-fold increase in anticoagulant thrombomodulin (TM [THBD gene]) transcript expression levels (P < .001). Bulk RNA-seq analysis of sorted CD14+ monocytes on an independent cohort of COVID-19 patients confirmed these findings (P < .05). Indicative of excessive coagulation, 41% of COVID-19 patients' plasma samples contained high D-dimer levels (P < .0001); of these, 19% demonstrated extracellular vesicle TF activity (P = .109). COVID-19 patients' ex vivo plasma-based thrombin generation correlated positively with D-dimer levels (P < .01). Plasma procoagulant extracellular vesicles were elevated ∼9-fold in COVID-19 patients (P < .01). Public scRNA-seq data sets from bronchoalveolar lung fluid and our peripheral blood mononuclear cell scRNA-seq data show CD14+ monocytes/macrophages TF transcript expression levels are elevated in severe but not mild or moderate COVID-19 patients. CONCLUSIONS Beyond local lung injury, SARS-CoV-2 infection increases systemic TF (F3) transcript levels and elevates circulating extracellular vesicles that likely contribute to disease-associated coagulation, thrombosis, and related mortality.
Collapse
Affiliation(s)
- Thomas J Girard
- Department of Pediatrics, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Lilian Antunes
- Department of Pediatrics, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Nan Zhang
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Junedh M Amrute
- Department of Internal Medicine, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Renumathi Subramanian
- Department of Pediatrics, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Irem Eldem
- Department of Pediatrics, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Kenneth E Remy
- Department of Pediatrics, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Monty Mazer
- Department of Internal Medicine, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Emma C Erlich
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Carlos Cruchaga
- Department of Psychiatry, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Ashley L Steed
- Department of Pediatrics, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Gwendalyn J Randolph
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Jorge Di Paola
- Department of Pediatrics, Washington University School of Medicine, St. Louis, Missouri, USA.
| |
Collapse
|
33
|
Kumar SVRB, Elango I, Balasubramanian K, Nair SV, Ramasamy A, Kathir C. Renal Allograft Cortical Necrosis in a COVID-19 Positive Patient. Indian J Nephrol 2023; 33:125-127. [PMID: 37234436 PMCID: PMC10208533 DOI: 10.4103/ijn.ijn_3_22] [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: 01/05/2022] [Accepted: 03/15/2022] [Indexed: 11/04/2022] Open
Abstract
The incidence of acute kidney injury (AKI) has been reported to be higher in kidney transplant recipients infected with SARS-CoV-2 compared with the general population. Here, we report a case of cortical necrosis in the graft kidney due to COVID infection in a patient with stable graft function over the years. The patient was started on hemodialysis and treated with steroids, and anticoagulants for COVID infection. Later, he had gradual improvement in his graft function and became dialysis independent on follow up.
Collapse
Affiliation(s)
- SVR Bipin Kumar
- Department of Nephrology, Saveetha Medical College Hospital, Tamil Nadu, India
| | - Indumathi Elango
- Department of Nephrology, Saveetha Medical College Hospital, Tamil Nadu, India
| | | | - Sanjeev V. Nair
- Department of Nephrology, Saveetha Medical College Hospital, Tamil Nadu, India
| | - Ashok Ramasamy
- Department of Nephrology, Saveetha Medical College Hospital, Tamil Nadu, India
| | | |
Collapse
|
34
|
Xiang M, Wu X, Jing H, Novakovic VA, Shi J. The intersection of obesity and (long) COVID-19: Hypoxia, thrombotic inflammation, and vascular endothelial injury. Front Cardiovasc Med 2023; 10:1062491. [PMID: 36824451 PMCID: PMC9941162 DOI: 10.3389/fcvm.2023.1062491] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2022] [Accepted: 01/23/2023] [Indexed: 02/10/2023] Open
Abstract
The role of hypoxia, vascular endothelial injury, and thrombotic inflammation in worsening COVID-19 symptoms has been generally recognized. Damaged vascular endothelium plays a crucial role in forming in situ thrombosis, pulmonary dysfunction, and hypoxemia. Thrombotic inflammation can further aggravate local vascular endothelial injury and affect ventilation and blood flow ratio. According to the results of many studies, obesity is an independent risk factor for a variety of severe respiratory diseases and contributes to high mechanical ventilation rate, high mortality, and slow recovery in COVID-19 patients. This review will explore the mechanisms by which obesity may aggravate the acute phase of COVID-19 and delay long COVID recovery by affecting hypoxia, vascular endothelial injury, and thrombotic inflammation. A systematic search of PubMed database was conducted for papers published since January 2020, using the medical subject headings of "COVID-19" and "long COVID" combined with the following keywords: "obesity," "thrombosis," "endothelial injury," "inflammation," "hypoxia," "treatment," and "anticoagulation." In patients with obesity, the accumulation of central fat restricts the expansion of alveoli, exacerbating the pulmonary dysfunction caused by SARS-CoV-2 invasion, inflammatory damage, and lung edema. Abnormal fat secretion and immune impairment further aggravate the original tissue damage and inflammation diffusion. Obesity weakens baseline vascular endothelium function leading to an early injury and pre-thrombotic state after infection. Enhanced procoagulant activity and microthrombi promote early obstruction of the vascular. Obesity also prolongs the duration of symptoms and increases the risk of sequelae after hospital discharge. Persistent viral presence, long-term inflammation, microclots, and hypoxia may contribute to the development of persistent symptoms, suggesting that patients with obesity are uniquely susceptible to long COVID. Early interventions, including supplemental oxygen, comprehensive antithrombotic therapy, and anti-inflammatory drugs, show effectiveness in many studies in the prevention of serious hypoxia, thromboembolic events, and systemic inflammation, and are therefore recommended to reduce intensive care unit admission, mortality, and sequelae.
Collapse
Affiliation(s)
- Mengqi Xiang
- Department of Hematology, The First Affiliated Hospital of Harbin Medical University, Harbin Medical University, Harbin, China
| | - Xiaoming Wu
- Department of Hematology, The First Affiliated Hospital of Harbin Medical University, Harbin Medical University, Harbin, China
| | - Haijiao Jing
- Department of Hematology, The First Affiliated Hospital of Harbin Medical University, Harbin Medical University, Harbin, China
| | - Valerie A. Novakovic
- Department of Research, Veterans Affairs Boston Healthcare System and Harvard Medical School, Boston, MA, United States
| | - Jialan Shi
- Department of Hematology, The First Affiliated Hospital of Harbin Medical University, Harbin Medical University, Harbin, China
- Department of Research, Veterans Affairs Boston Healthcare System and Harvard Medical School, Boston, MA, United States
- Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA, United States
| |
Collapse
|
35
|
de Lima F, Moraes CRP, Barbosa MS, Bombassaro B, Palma AC, Dertkigil SSJ, Moretti ML, Orsi FA, Annichino-Bizzacchi JM, Mansour E, Velloso LA, De Paula EV. Association of heme-oxygenase 1, hemopexin, and heme levels with markers of disease severity in COVID-19. Exp Biol Med (Maywood) 2023; 248:309-316. [PMID: 36740756 PMCID: PMC9902789 DOI: 10.1177/15353702221139185] [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] [Indexed: 02/07/2023] Open
Abstract
Heme-oxygenase 1 (HO-1) is an enzyme with well-known anti-inflammatory and antioxidant properties, whose levels have been previously associated with disease severity in the context of sterile and infectious diseases. Moreover, the heme/HO-1 pathway has been associated with prothrombotic changes in other diseases. Accordingly, the potential of modulating HO-1 levels for the treatment of COVID-19 was extensively speculated during the COVID-19 pandemic, but very few actual data were generated. The aim of our study was to explore the association of HO-1, heme, and hemopexin (HPX) levels with COVID-19 severity and with markers of inflammation and coagulation activation. The study was conducted in 30 consecutive patients with COVID-19 admitted due to hypoxemia, and 30 healthy volunteers matched by sex, age, and geographic region. HO-1 and HPX levels were measured by enzyme immunoassay (ELISA) and heme levels were measured by a colorimetric method. A comprehensive panel of coagulation and fibrinolysis activation was also used. Patients with COVID-19 presented increased levels of HO-1 when compared to controls (5741 ± 2696 vs 1953 ± 612 pg/mL, respectively, P < 0.0001), as well as a trend toward increased levels of HPX (3.724 ± 0.880 vs 3.254 ± 1.022 mg/mL, respectively; P = 0.06). In addition, HO-1 and HPX levels reduced from admission to day + 4. HO-1 levels were associated with duration of intensive care unit stay and with several markers of coagulation activation. In conclusion, modulation of HO-1 could be associated with the prothrombotic state observed in COVID-19, and HO-1 could also represent a relevant biomarker for COVID-19. New independent studies are warranted to explore and expand these findings.
Collapse
Affiliation(s)
- Franciele de Lima
- School of Medical Sciences, University of Campinas, Campinas 13083-887, Brazil,Franciele de Lima.
| | | | - Mayck Silva Barbosa
- School of Medical Sciences, University of Campinas, Campinas 13083-887, Brazil
| | - Bruna Bombassaro
- Obesity and Comorbidities Research Center, University of Campinas, Campinas 13083-864, Brazil
| | - André C Palma
- School of Medical Sciences, University of Campinas, Campinas 13083-887, Brazil
| | | | - Maria Luiza Moretti
- School of Medical Sciences, University of Campinas, Campinas 13083-887, Brazil
| | | | - Joyce M Annichino-Bizzacchi
- School of Medical Sciences, University of Campinas, Campinas 13083-887, Brazil,Hematology and Hemotherapy Center, University of Campinas, Campinas 13083-878, Brazil
| | - Eli Mansour
- School of Medical Sciences, University of Campinas, Campinas 13083-887, Brazil
| | - Licio A Velloso
- School of Medical Sciences, University of Campinas, Campinas 13083-887, Brazil,Obesity and Comorbidities Research Center, University of Campinas, Campinas 13083-864, Brazil
| | - Erich Vinicius De Paula
- School of Medical Sciences, University of Campinas, Campinas 13083-887, Brazil,Hematology and Hemotherapy Center, University of Campinas, Campinas 13083-878, Brazil
| |
Collapse
|
36
|
Salihi S, Perçin B, Erkengel HI, Özalp B, Saçlı H, Kara I. Does COVID-19 infection increase the risk of pulmonary embolism in ambulatory patients with deep vein thrombosis. Vascular 2023; 31:182-188. [PMID: 34866519 DOI: 10.1177/17085381211052207] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
OBJECTIVES Coronavirus disease 2019 (COVID-19) can lead to systemic coagulation activation and thrombotic complications including venous thromboembolism. This study compares the development of pulmonary embolism, post-thrombotic syndrome, and clinical outcomes of COVID-19 and non-COVID-19 patients with deep vein thrombosis (DVT). METHODS One hundred and eight patients diagnosed with acute deep vein thrombosis (DVT) between June 2020 and February 2021 in our institution were included in this retrospective study. Thirty-nine patients had been previously diagnosed with COVID-19 and specified as the COVID-19 group. Sixty-nine patients did not have COVID-19 and specified as the non-COVID-19 group. Mean ages of both groups were 64.3 ± 15.8 and 60.1 ± 19.7 years, respectively (p = .37). RESULTS The median duration from the onset of the COVID-19 to diagnosis of DVT was 22 (2-120) days in the COVID-19 group. The patients of two groups were mostly treated outpatient at rates of 94.9% vs 94.2%, respectively (p = .88). Pulmonary embolism was seen in six patients (15.4%) in the COVID-19 group and in three patients (4.3%) in the non-COVID-19 group (p = .04). Kaplan-Meir curves showed that patients with COVİD-19 had significantly higher pulmonary embolism than those without COVID-19 (p = .015). The recurrence rate of DVT was 2.6% in the COVID-19 group (n = 1), and 4.3% in the non-COVID-19 group (n = 3), indicating no statistically significant difference (p = .63). Mortality was seen in six patients (15.4%) in the COVİD-19 group, and in seven patients (10.1%) in the non-COVID-19 group. According to the Kaplan-Meir method, 10 months survival rates were 73.9 ± 10% in the COVID-19 group, and 66.3 ± 12.8% in the non-COVID-19 group with no statistical significance (p = .218). CONCLUSIONS Our data draw attention to the fact that deep vein thrombosis should not be considered a safe and self-limited condition. Efficient preventive measures such as mobilization and prophylactic drug use should be considered to prevent DVT during the management of COVID-19.
Collapse
Affiliation(s)
- Salih Salihi
- Medicine Faculty, Department of Cardiovascular Surgery, 175678Sakarya University, Sakarya, Turkey
| | - Bilal Perçin
- Department of Cardiovascular Surgery, 175679Sakarya University Training and Research Hospital, Sakarya, Turkey
| | - Halil Ibrahim Erkengel
- Department of Cardiovascular Surgery, 175679Sakarya University Training and Research Hospital, Sakarya, Turkey
| | - Bilhan Özalp
- Department of Cardiovascular Surgery, 175679Sakarya University Training and Research Hospital, Sakarya, Turkey
| | - Hakan Saçlı
- Medicine Faculty, Department of Cardiovascular Surgery, 175678Sakarya University, Sakarya, Turkey
| | - Ibrahim Kara
- Medicine Faculty, Department of Cardiovascular Surgery, 175678Sakarya University, Sakarya, Turkey
| |
Collapse
|
37
|
Dissecting Platelet's Role in Viral Infection: A Double-Edged Effector of the Immune System. Int J Mol Sci 2023; 24:ijms24032009. [PMID: 36768333 PMCID: PMC9916939 DOI: 10.3390/ijms24032009] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2022] [Revised: 12/11/2022] [Accepted: 12/23/2022] [Indexed: 01/20/2023] Open
Abstract
Platelets play a major role in the processes of primary hemostasis and pathological inflammation-induced thrombosis. In the mid-2000s, several studies expanded the role of these particular cells, placing them in the "immune continuum" and thus changing the understanding of their function in both innate and adaptive immune responses. Among the many receptors they express on their surface, platelets express Toll-Like Receptors (TLRs), key receptors in the inflammatory cell-cell reaction and in the interaction between innate and adaptive immunity. In response to an infectious stimulus, platelets will become differentially activated. Platelet activation is variable depending on whether platelets are activated by a hemostatic or pathogen stimulus. This review highlights the role that platelets play in platelet modulation count and adaptative immune response during viral infection.
Collapse
|
38
|
Bai Y, Guo Y, Gu L. Additional risk factors improve mortality prediction for patients hospitalized with influenza pneumonia: a retrospective, single-center case-control study. BMC Pulm Med 2023; 23:19. [PMID: 36647106 PMCID: PMC9841622 DOI: 10.1186/s12890-022-02283-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2022] [Accepted: 12/12/2022] [Indexed: 01/18/2023] Open
Abstract
BACKGROUND Severe influenza, especially influenza pneumonia, causes large numbers of deaths each year. Some patients who develop severe influenza have no known risk factors. In this study we investigated risk factors for mortality of patients with influenza A-related pneumonia who have different basic conditions. We also evaluated the power of pneumonia severity assessment tools in Chinese patients hospitalized with influenza A-related pneumonia. Together, these results could provide a basis for a screening method that has improved ability for the early identification of critical patients who will have poor prognoses in clinical practice. METHODS This single-center, retrospective case-control study included 152 adult patients with severe influenza over six influenza seasons. Data for diagnoses and demographics, as well clinical data, laboratory findings, treatment methods, 30-day and 60-day outcomes of the patients were collected. Patients who had any of the risk factors for severe influenza were included in the high-risk group, and those that had no known risk factors were included in the low-risk group. RESULTS The PSI, CURB-65 and PIRO-CAP tools all underestimated the mortality rate of patients hospitalized with influenza A-related pneumonia, and this underestimate was more pronounced for low-risk patients. D-dimer (Odds ratio [OR] = 1.052, 95% confidence interval [CI] 1.001-1.106, p = 0.045) and direct bilirubin (OR = 1.143, 95%CI 1.049-1.246, p = 0.002) were independent risk factors for mortality of patients with influenza A-related pneumonia. When used in combination with ferritin and D-dimer, the area under receiver operator characteristic curve (AUCROC) was 0.851 (95%CI 0.780-0.922, p < 0.001), 0.840 (95%CI 0.763-0.916, p < 0.001) and 0.829 (95%CI 0.748-0.911, p < 0.001) for PSI, CURB-65 and PIRO-CAP, respectively, which was higher than that obtained using PSI, CURB-65 and PIRO-CAP alone. CONCLUSIONS The findings demonstrate that currently used community-acquired pneumonia (CAP) scoring systems could underestimate the risk of influenza A-related pneumonia mortality. D-dimer was shown to be an independent risk factor of mortality for influenza A-related pneumonia in hospitalized patients, and a combination of D-dimer with ferritin could improve the predictive value of PSI, CURB-65 and PIRO-CAP for adverse prognoses of patients with influenza A-related pneumonia.
Collapse
Affiliation(s)
- Yu Bai
- grid.24696.3f0000 0004 0369 153XDepartment of Infectious Diseases and Clinical Microbiology, Beijing Institute of Respiratory Medicine and Beijing Chao-Yang Hospital, Capital Medical University, No.8 Worker’s Stadium South Road, Chaoyang District, Beijing, China
| | - Yiqun Guo
- grid.24696.3f0000 0004 0369 153XDepartment of Infectious Diseases and Clinical Microbiology, Beijing Institute of Respiratory Medicine and Beijing Chao-Yang Hospital, Capital Medical University, No.8 Worker’s Stadium South Road, Chaoyang District, Beijing, China
| | - Li Gu
- grid.24696.3f0000 0004 0369 153XDepartment of Infectious Diseases and Clinical Microbiology, Beijing Institute of Respiratory Medicine and Beijing Chao-Yang Hospital, Capital Medical University, No.8 Worker’s Stadium South Road, Chaoyang District, Beijing, China
| |
Collapse
|
39
|
Cappelletto A, Allan HE, Crescente M, Schneider E, Bussani R, Ali H, Secco I, Vodret S, Simeone R, Mascaretti L, Zacchigna S, Warner TD, Giacca M. SARS-CoV-2 Spike protein activates TMEM16F-mediated platelet procoagulant activity. Front Cardiovasc Med 2023; 9:1013262. [PMID: 36684586 PMCID: PMC9845929 DOI: 10.3389/fcvm.2022.1013262] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2022] [Accepted: 11/29/2022] [Indexed: 01/05/2023] Open
Abstract
Thrombosis of the lung microvasculature is a characteristic of COVID-19 disease, which is observed in large excess compared to other forms of acute respiratory distress syndrome and thus suggests a trigger for thrombosis that is endogenous to the lung. Our recent work has shown that the SARS-CoV-2 Spike protein activates the cellular TMEM16F chloride channel and scramblase. Through a screening on >3,000 FDA/EMA approved drugs, we identified Niclosamide and Clofazimine as the most effective molecules at inhibiting Spike-induced TMEM16 activation. As TMEM16F plays an important role in stimulating the procoagulant activity of platelets, we investigated whether Spike directly affects platelet activation and pro-thrombotic function and tested the effect of Niclosamide and Clofazimine on these processes. Here we show that Spike, present either on the virion envelope or on the cell plasma membrane, promotes platelet activation, adhesion and spreading. Spike was active as a sole agonist or, even more effectively, by enhancing the function of known platelet activators. In particular, Spike-induced a marked procoagulant phenotype in platelets, by enhancing Ca2+ flux, phosphatidylserine externalization on the platelet outer cell membrane, and thrombin generation. Eventually, this increased thrombin-induced clot formation and retraction. Both Niclosamide and Clofazimine blocked this Spike-induced procoagulant response. These findings provide a pathogenic mechanism to explain lung thrombosis-associated with severe COVID-19 infection. We propose that Spike, present in SARS-CoV-2 virions or exposed on the surface of infected cells in the lungs, enhances the effects of inflammation and leads to local platelet stimulation and subsequent activation of the coagulation cascade. As platelet TMEM16F is central in this process, these findings reinforce the rationale of repurposing Niclosamide for COVID-19 therapy.
Collapse
Affiliation(s)
- Ambra Cappelletto
- British Heart Foundation Centre of Research Excellence, School of Cardiovascular Medicine & Sciences, King’s College London, London, United Kingdom
| | - Harriet E. Allan
- Barts and the London School of Medicine and Dentistry, Blizard Institute, Queen Mary University of London, London, United Kingdom
| | - Marilena Crescente
- Barts and the London School of Medicine and Dentistry, Blizard Institute, Queen Mary University of London, London, United Kingdom
| | - Edoardo Schneider
- British Heart Foundation Centre of Research Excellence, School of Cardiovascular Medicine & Sciences, King’s College London, London, United Kingdom
| | - Rossana Bussani
- Department of Medical, Surgical and Health Sciences, University of Trieste, Trieste, Italy
| | - Hashim Ali
- British Heart Foundation Centre of Research Excellence, School of Cardiovascular Medicine & Sciences, King’s College London, London, United Kingdom
| | - Ilaria Secco
- British Heart Foundation Centre of Research Excellence, School of Cardiovascular Medicine & Sciences, King’s College London, London, United Kingdom
| | - Simone Vodret
- International Centre for Genetic Engineering and Biotechnology (ICGEB), Trieste, Italy
| | - Roberto Simeone
- Dipartimento di Medicina Trasfusionale Giuliano-Isontino, Azienda Sanitaria Universitaria Giuliano Isontina (ASUGI), Trieste, Italy
| | - Luca Mascaretti
- Dipartimento di Medicina Trasfusionale Giuliano-Isontino, Azienda Sanitaria Universitaria Giuliano Isontina (ASUGI), Trieste, Italy
| | - Serena Zacchigna
- Department of Medical, Surgical and Health Sciences, University of Trieste, Trieste, Italy,International Centre for Genetic Engineering and Biotechnology (ICGEB), Trieste, Italy
| | - Timothy D. Warner
- Barts and the London School of Medicine and Dentistry, Blizard Institute, Queen Mary University of London, London, United Kingdom,*Correspondence: Timothy D. Warner,
| | - Mauro Giacca
- British Heart Foundation Centre of Research Excellence, School of Cardiovascular Medicine & Sciences, King’s College London, London, United Kingdom,Mauro Giacca,
| |
Collapse
|
40
|
Li L, Wu Y, Wang J, Yan H, Lu J, Wang Y, Zhang B, Zhang J, Yang J, Wang X, Zhang M, Li Y, Miao L, Zhang H. Potential Treatment of COVID-19 with Traditional Chinese Medicine: What Herbs Can Help Win the Battle with SARS-CoV-2? ENGINEERING (BEIJING, CHINA) 2022; 19:139-152. [PMID: 34729244 PMCID: PMC8552808 DOI: 10.1016/j.eng.2021.08.020] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/06/2021] [Revised: 06/28/2021] [Accepted: 08/03/2021] [Indexed: 05/05/2023]
Abstract
Traditional Chinese medicine (TCM) has been successfully applied worldwide in the treatment of coronavirus disease 2019 (COVID-19), which is caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). However, the pharmacological mechanisms underlying this success remain unclear. Hence, the aim of this review is to combine pharmacological assays based on the theory of TCM in order to elucidate the potential signaling pathways, targets, active compounds, and formulas of herbs that are involved in the TCM treatment of COVID-19, which exhibits combatting viral infections, immune regulation, and amelioration of lung injury and fibrosis. Extensive reports on target screening are elucidated using virtual prediction via docking analysis or network pharmacology based on existing data. The results of these reports indicate that an intricate regulatory mechanism is involved in the pathogenesis of COVID-19. Therefore, more pharmacological research on the natural herbs used in TCM should be conducted in order to determine the association between TCM and COVID-19 and account for the observed therapeutic effects of TCM against COVID-19.
Collapse
Affiliation(s)
- Lin Li
- State Key Laboratory of Component-Based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
- Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
- Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Yuzheng Wu
- State Key Laboratory of Component-Based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
- Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
- Laboratory of Pharmacology of TCM Formulae Co-Constructed by the Province-Ministry, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Jiabao Wang
- State Key Laboratory of Component-Based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
- Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
- Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Huimin Yan
- State Key Laboratory of Component-Based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
- Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
- Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Jia Lu
- State Key Laboratory of Component-Based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
- Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
- Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Yu Wang
- State Key Laboratory of Component-Based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Boli Zhang
- State Key Laboratory of Component-Based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
- Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
- Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Junhua Zhang
- State Key Laboratory of Component-Based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
- Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
- Evidence-Based Medicine Center, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Jian Yang
- State Key Laboratory of Component-Based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
- Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
- Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Xiaoying Wang
- State Key Laboratory of Component-Based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
- College of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Min Zhang
- State Key Laboratory of Component-Based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
- Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
- Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Yue Li
- State Key Laboratory of Component-Based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
- Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
- Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Lin Miao
- State Key Laboratory of Component-Based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
- Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
- Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Han Zhang
- State Key Laboratory of Component-Based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
- Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
- Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| |
Collapse
|
41
|
Subramaniam S, Kothari H, Bosmann M. Tissue factor in COVID-19-associated coagulopathy. Thromb Res 2022; 220:35-47. [PMID: 36265412 PMCID: PMC9525243 DOI: 10.1016/j.thromres.2022.09.025] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2022] [Revised: 09/24/2022] [Accepted: 09/28/2022] [Indexed: 11/17/2022]
Abstract
Evidence of micro- and macro-thrombi in the arteries and veins of critically ill COVID-19 patients and in autopsies highlight the occurrence of COVID-19-associated coagulopathy (CAC). Clinical findings of critically ill COVID-19 patients point to various mechanisms for CAC; however, the definitive underlying cause is unclear. Multiple factors may contribute to the prothrombotic state in patients with COVID-19. Aberrant expression of tissue factor (TF), an initiator of the extrinsic coagulation pathway, leads to thrombotic complications during injury, inflammation, and infections. Clinical evidence suggests that TF-dependent coagulation activation likely plays a role in CAC. Multiple factors could trigger abnormal TF expression and coagulation activation in patients with severe COVID-19 infection. Proinflammatory cytokines that are highly elevated in COVID-19 (IL-1β, IL-6 and TNF-α) are known induce TF expression on leukocytes (e.g. monocytes, macrophages) and non-immune cells (e.g. endothelium, epithelium) in other conditions. Antiphospholipid antibodies, TF-positive extracellular vesicles, pattern recognition receptor (PRR) pathways and complement activation are all candidate factors that could trigger TF-dependent procoagulant activity. In addition, coagulation factors, such as thrombin, may further potentiate the induction of TF via protease-activated receptors on cells. In this systematic review, with other viral infections, we discuss potential mechanisms and cell-type-specific expressions of TF during SARS-CoV-2 infection and its role in the development of CAC.
Collapse
Affiliation(s)
- Saravanan Subramaniam
- Pulmonary Center, Department of Medicine, Boston University School of Medicine, Boston, MA 02118, USA.
| | - Hema Kothari
- Carter Immunology Center, University of Virginia, Charlottesville, VA 22908, USA; Cardiovascular Division, Department of Medicine, University of Virginia, Charlottesville, VA 22908, USA
| | - Markus Bosmann
- Pulmonary Center, Department of Medicine, Boston University School of Medicine, Boston, MA 02118, USA; Center for Thrombosis and Hemostasis, University Medical Center of the Johannes Gutenberg-University, Mainz, Germany
| |
Collapse
|
42
|
Zekri-Nechar K, Zamorano-León JJ, Reche C, Giner M, López-de-Andrés A, Jiménez-García R, López-Farré AJ, Martínez-Martínez CH. Spike Protein Subunits of SARS-CoV-2 Alter Mitochondrial Metabolism in Human Pulmonary Microvascular Endothelial Cells: Involvement of Factor Xa. DISEASE MARKERS 2022; 2022:1118195. [PMID: 36438904 PMCID: PMC9699787 DOI: 10.1155/2022/1118195] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/09/2022] [Revised: 10/07/2022] [Accepted: 11/01/2022] [Indexed: 09/12/2023]
Abstract
BACKGROUND Mitochondria have been involved in host defense upon viral infections. Factor Xa (FXa), a coagulating factor, may also have influence on mitochondrial functionalities. The aim was to analyze if in human pulmonary microvascular endothelial cells (HPMEC), the SARS-CoV-2 (COVID-19) spike protein subunits, S1 and S2 (S1+S2), could alter mitochondrial metabolism and what is the role of FXA. METHODS HPMEC were incubated with and without recombinants S1+S2 (10 nmol/L each). RESULTS In control conditions, S1+S2 failed to modify FXa expression. However, in LPS (1 μg/mL)-incubated HPMEC, S1+S2 significantly increased FXa production. LPS tended to reduce mitochondrial membrane potential with respect to control, but in higher and significant degree, it was reduced when S1+S2 were present. LPS did not significantly modify cytochrome c oxidase activity as compared with control. Addition of S1+S2 spike subunits to LPS-incubated HPMEC significantly increased cytochrome c oxidase activity with respect to control. Lactate dehydrogenase activity was also increased by S1+S2 with respect to control and LPS alone. Protein expression level of uncoupled protein-2 (UCP-2) was markedly expressed when S1+S2 were added together to LPS. Rivaroxaban (50 nmol/L), a specific FXa inhibitor, significantly reduced all the above-mentioned alterations induced by S1+S2 including UCP-2 expression. CONCLUSIONS In HPMEC undergoing to preinflammatory condition, COVID-19 S1+S2 spike subunits promoted alterations in mitochondria metabolism suggesting a shift from aerobic towards anaerobic metabolism that was accompanied of high FXa production. Rivaroxaban prevented all the mitochondrial metabolic changes mediated by the present COVID-19 S1 and S2 spike subunits suggesting the involvement of endogenous FXa.
Collapse
Affiliation(s)
| | - José J. Zamorano-León
- Public Health and Maternal, Child Health Department, School of Medicine, Universidad Complutense, Madrid, Spain
- IdISSC, Madrid, Spain
| | - Carmen Reche
- Gomez Ulla Central Defense Hospital, Madrid, Spain
| | - Manel Giner
- Surgical Departments, School of Medicine, Universidad Complutense, Madrid, Spain
| | - Ana López-de-Andrés
- Public Health and Maternal, Child Health Department, School of Medicine, Universidad Complutense, Madrid, Spain
- IdISSC, Madrid, Spain
| | - Rodrigo Jiménez-García
- Public Health and Maternal, Child Health Department, School of Medicine, Universidad Complutense, Madrid, Spain
- IdISSC, Madrid, Spain
| | | | | |
Collapse
|
43
|
Sachetto ATA, Mackman N. Tissue Factor and COVID-19: An Update. Curr Drug Targets 2022; 23:1573-1577. [PMID: 36165519 DOI: 10.2174/1389450123666220926144432] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2022] [Revised: 08/21/2022] [Accepted: 08/29/2022] [Indexed: 01/25/2023]
Abstract
The coronavirus 2019 (COVID-19) pandemic is caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) virus. Infection with SARS-CoV-2 is associated with acute respiratory distress syndrome, thrombosis and a high rate of mortality. Thrombotic events increase with severity. Tissue factor (TF) expression is increased during viral and bacterial infections. This review summarizes studies that have examined TF expression in response to SARS-CoV-2 infection. SARS-CoV-2 virus and its proteins upregulate TF mRNA, protein and activity in a variety of cells, including bronchial epithelial cells, neutrophils, monocytes, macrophages, endothelial cells and adventitial fibroblasts. COVID-19 patients have increased TF expression in lungs, bronchoalveolar lavage fluid and circulating extracellular vesicles. The increase in TF was associated with coagulation activation markers, thrombosis, inflammatory markers, severity of disease and mortality. Taken together, the studies suggest that TF plays a central role in thrombosis in COVID- 19. TF may be a useful prognostic marker and therapeutic target to reduce thrombosis and inflammation.
Collapse
Affiliation(s)
- Ana Teresa Azevedo Sachetto
- Laboratory of Pathophysiology, Butantan Institute, São Paulo, São Paulo, Brazil.,University of São Paulo Medical School, São Paulo, São Paulo, Brazil.,Department of Medicine, UNC Blood Research Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Nigel Mackman
- Department of Medicine, UNC Blood Research Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| |
Collapse
|
44
|
Schieffer SM, Pelland SC, Hagopian C, Cline DM. SARS-CoV-2 in a Southeastern US Hospital System: Interplay of Complications and Code Status Downgrades. Respir Care 2022; 67:1452-1455. [PMID: 35672138 PMCID: PMC9993969 DOI: 10.4187/respcare.09748] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Affiliation(s)
- Sydney M Schieffer
- Department of Emergency Medicine, Atrium Health Wake Forest Baptist Medical Center, Winston Salem, North Carolina
| | - Scott C Pelland
- Department of Emergency Medicine, Atrium Health Wake Forest Baptist Medical Center, Winston Salem, North Carolina
| | - Colleen Hagopian
- Department of Emergency Medicine, Atrium Health Wake Forest Baptist Medical Center, Winston Salem, North Carolina
| | - David M Cline
- Department of Emergency Medicine, Atrium Health Wake Forest Baptist Medical Center, Winston Salem, North Carolina.
| |
Collapse
|
45
|
Coscas R, Dubosq M, Charton J, El Batti S, Gaudric J, Koskas F, Chiche L, Couture T, Davaine JM, Castier Y, Cerceau P, EL Hajjam M, Samb P, Beauchet A, Grimaldi L, Javerliat I, Goeau-Brissonniere O, Alsac JM, Onorati I, Martinod E, Desgranges P, Touma J, Cochennec F, Pellenc Q, Julia P, Coggia M. Thrombosis of Medium-Sized and Large Arteries During Covid-19 Infection: Results of the COVIVASC Study. Ann Vasc Surg 2022; 86:35-42. [PMID: 35780947 PMCID: PMC9242891 DOI: 10.1016/j.avsg.2022.04.055] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2022] [Revised: 03/15/2022] [Accepted: 04/15/2022] [Indexed: 01/31/2023]
Abstract
BACKGROUND COVID-19 infection is associated not only with venous thromboses but also with arterial thromboses (COV-ATs) in relation with an endothelial dysfunction, a coagulopathy and rhythm disorders. The incidence, the topography, and the prognosis of COV-ATs remain poorly known. The objective of this study was to report the overall experience of the Greater Paris University Hospitals (Assistance Publique - Hopitaux de Paris, AP-HP) during the first pandemic wave of COVID-19 infection. METHODS After approval by the ethics committee, a study using the AP-HP clinical data warehouse was carried out between March and May 2020. Overall, 124,609 patients had a polymerase chain reaction for COVID-19 in our hospitals, of which 25,345 were positive. From 20,710 exploitable stays, patients tested positive for COVID who presented an episode of acute COV-AT (except coronary and intracranial arteries) were selected on the basis of the French medical classification for clinical procedures codes. The data are presented as absolute values with percentages and/or means with standard deviation. RESULTS Over the studied period, 60 patients (aged 71±14 years, 42 men) presented a COV-AT at the time of their hospitalization, an incidence of 0.2%. The arterial complication occurred 3±7 days after the COVID infection and was inaugural in 30% of the cases (n = 18). The sites of COV-AT were the lower extremities (n = 35%, 58%), the abdominal aorta (n = 10%, 17%), the thoracic aorta (n = 7%, 12%), the upper limbs (n = 7%, 12%), the cerebral arteries (n = 7%, 12%), the digestive arteries (n = 6%, 10%), the renal arteries (n = 2%, 3%), and the ophthalmic artery (n = 1%, 2%). Multiple COV-ATs were observed in 13 patients (22%). At the time of diagnosis, 20 (33%) patients were in intensive care, including six (10%) patients who were intubated. On computed tomography angiography, COVID lesions were classified as moderate and severe in 25 (42%) and 21 (35%) cases, respectively. Revascularization was attempted in 27 patients (45%), by open surgery in 16 cases, using endovascular techniques in 8 cases and with a hybrid approach in three cases. Six patients (22%) required reinterventions. The duration of hospitalization was 12±9 days. Early mortality (in-hospital or at 30 days) was 30% (n = 18). Nine (15%) patients presented severe nonlethal ischemic complications. CONCLUSIONS Arterial involvement is rare during COVID-19 infection. The aorta and the arteries of the limbs are the privileged sites. The morbi-mortality of these patients is high. Future studies will have to determine if the systematization of anticoagulation therapy decreases the incidence and the severity of the condition.
Collapse
Affiliation(s)
- Raphael Coscas
- Department of Vascular Surgery, Ambroise Paré University Hospital, Assistance Publique-Hôpitaux de Paris, 92104 Boulogne-Billancourt, France,UMR 1018, Inserm-Paris11 - CESP, Versailles Saint-Quentin-en-Yvelines University, Paris-Saclay University, Paul Brousse Hospital, 94807 Villejuif, France,Correspondence to: Raphaël Coscas, Department of Vascular Surgery, Ambroise Paré University Hospital, 9 avenue Charles de Gaulle, 92104 Boulogne Cedex, France
| | - Maxime Dubosq
- Department of Vascular Surgery, Ambroise Paré University Hospital, Assistance Publique-Hôpitaux de Paris, 92104 Boulogne-Billancourt, France,UMR 1018, Inserm-Paris11 - CESP, Versailles Saint-Quentin-en-Yvelines University, Paris-Saclay University, Paul Brousse Hospital, 94807 Villejuif, France
| | - Johanna Charton
- Department of Vascular Surgery, Ambroise Paré University Hospital, Assistance Publique-Hôpitaux de Paris, 92104 Boulogne-Billancourt, France,UMR 1018, Inserm-Paris11 - CESP, Versailles Saint-Quentin-en-Yvelines University, Paris-Saclay University, Paul Brousse Hospital, 94807 Villejuif, France
| | - Salma El Batti
- Department of Cardiovascular Surgery, Hôpital Européen Georges Pompidou, Assistance Publique-Hôpitaux de Paris, 75015 Paris, France
| | - Julien Gaudric
- Department of Vascular and Endovascular Surgery–Tertiary Aortic Center, Pitié-Salpêtrière University Hospital, Assistance Publique-Hôpitaux de Paris and Sorbonne Médecine Universités, 75013 Paris, France
| | - Fabien Koskas
- Department of Vascular and Endovascular Surgery–Tertiary Aortic Center, Pitié-Salpêtrière University Hospital, Assistance Publique-Hôpitaux de Paris and Sorbonne Médecine Universités, 75013 Paris, France
| | - Laurent Chiche
- Department of Vascular and Endovascular Surgery–Tertiary Aortic Center, Pitié-Salpêtrière University Hospital, Assistance Publique-Hôpitaux de Paris and Sorbonne Médecine Universités, 75013 Paris, France
| | - Thibault Couture
- Department of Vascular and Endovascular Surgery–Tertiary Aortic Center, Pitié-Salpêtrière University Hospital, Assistance Publique-Hôpitaux de Paris and Sorbonne Médecine Universités, 75013 Paris, France
| | - Jean-Michel Davaine
- Department of Vascular and Endovascular Surgery–Tertiary Aortic Center, Pitié-Salpêtrière University Hospital, Assistance Publique-Hôpitaux de Paris and Sorbonne Médecine Universités, 75013 Paris, France
| | - Yves Castier
- Department of Vascular Surgery, Bichat Hospital, Assistance Publique-Hôpitaux de Paris, Paris, France and Paris-Diderot University, Paris, France
| | - Pierre Cerceau
- Department of Vascular Surgery, Bichat Hospital, Assistance Publique-Hôpitaux de Paris, Paris, France and Paris-Diderot University, Paris, France
| | - Mostafa EL Hajjam
- Department of Radiology, Ambroise Paré University Hospital, Assistance Publique-Hôpitaux de Paris, 92104 Boulogne-Billancourt, France
| | - Patricia Samb
- Clinical Research Unit, Ambroise Paré University Hospital, Assistance Publique-Hôpitaux de Paris, Boulogne-Billancourt, France,University of Versailles Saint-Quentin en Yvelines, Paris-Saclay University, Paris, France
| | - Alain Beauchet
- Clinical Research Unit, Ambroise Paré University Hospital, Assistance Publique-Hôpitaux de Paris, Boulogne-Billancourt, France,University of Versailles Saint-Quentin en Yvelines, Paris-Saclay University, Paris, France
| | - Lamiae Grimaldi
- Clinical Research Unit, Ambroise Paré University Hospital, Assistance Publique-Hôpitaux de Paris, Boulogne-Billancourt, France,University of Versailles Saint-Quentin en Yvelines, Paris-Saclay University, Paris, France
| | - Isabelle Javerliat
- Department of Vascular Surgery, Ambroise Paré University Hospital, Assistance Publique-Hôpitaux de Paris, 92104 Boulogne-Billancourt, France,UMR 1018, Inserm-Paris11 - CESP, Versailles Saint-Quentin-en-Yvelines University, Paris-Saclay University, Paul Brousse Hospital, 94807 Villejuif, France
| | - Olivier Goeau-Brissonniere
- Department of Vascular Surgery, Ambroise Paré University Hospital, Assistance Publique-Hôpitaux de Paris, 92104 Boulogne-Billancourt, France,UMR 1018, Inserm-Paris11 - CESP, Versailles Saint-Quentin-en-Yvelines University, Paris-Saclay University, Paul Brousse Hospital, 94807 Villejuif, France
| | - Jean-Marc Alsac
- Department of Cardiovascular Surgery, Hôpital Européen Georges Pompidou, Assistance Publique-Hôpitaux de Paris, 75015 Paris, France
| | - Ilaria Onorati
- Department of Thoracic and Vascular Surgery, Hôpitaux Universitaires Paris Seine-Saint-Denis, Hôpital Avicenne, Assistance Publique-Hôpitaux de Paris, and Université Paris 13, Sorbonne Paris Cité, UFR Santé, Médecine et Biologie Humaine, Bobigny, France
| | - Emmanuel Martinod
- Department of Thoracic and Vascular Surgery, Hôpitaux Universitaires Paris Seine-Saint-Denis, Hôpital Avicenne, Assistance Publique-Hôpitaux de Paris, and Université Paris 13, Sorbonne Paris Cité, UFR Santé, Médecine et Biologie Humaine, Bobigny, France
| | - Pascal Desgranges
- Department of Vascular Surgery, Henri Mondor University Hospital, Assistance Publique-Hôpitaux de Paris, Créteil, France,Paris-Est Créteil Faculty of Medicine, Créteil, France
| | - Joseph Touma
- Department of Vascular Surgery, Henri Mondor University Hospital, Assistance Publique-Hôpitaux de Paris, Créteil, France,Paris-Est Créteil Faculty of Medicine, Créteil, France
| | - Frédéric Cochennec
- Department of Vascular Surgery, Henri Mondor University Hospital, Assistance Publique-Hôpitaux de Paris, Créteil, France,Paris-Est Créteil Faculty of Medicine, Créteil, France
| | - Quentin Pellenc
- Department of Vascular Surgery, Bichat Hospital, Assistance Publique-Hôpitaux de Paris, Paris, France and Paris-Diderot University, Paris, France
| | - Pierre Julia
- Department of Cardiovascular Surgery, Hôpital Européen Georges Pompidou, Assistance Publique-Hôpitaux de Paris, 75015 Paris, France
| | - Marc Coggia
- Department of Vascular Surgery, Ambroise Paré University Hospital, Assistance Publique-Hôpitaux de Paris, 92104 Boulogne-Billancourt, France,UMR 1018, Inserm-Paris11 - CESP, Versailles Saint-Quentin-en-Yvelines University, Paris-Saclay University, Paul Brousse Hospital, 94807 Villejuif, France
| |
Collapse
|
46
|
Murakami Y, Okazaki S, Yamamoto M, Sakurai R, Jinno J, Ozono T, Ikenaka K, Gon Y, Todo K, Sasaki T, Hirata H, Uchiyama A, Mochizuki H. Ischemic Stroke Due to Heparin-induced Thrombocytopenia during Severe COVID-19 Infection. Intern Med 2022; 61:2797-2801. [PMID: 35793954 PMCID: PMC9556234 DOI: 10.2169/internalmedicine.9531-22] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Abstract
A 53-year-old woman with severe coronavirus disease 2019 (COVID-19) pneumonia was admitted and treated with intravenous unfractionated heparin for thromboprophylaxis under general anesthesia with mechanical ventilation. She developed right hemiparesis after hospitalization due to a large hemorrhagic infarction. Her platelet count decreased from 243,000/μL at administration to 121,000/μL. Anti-platelet factor 4-heparin antibody testing was positive according to a latex immunoturbidimetric assay. She was therefore diagnosed with heparin-induced thrombocytopenia. We immediately stopped the heparin and started argatroban; the platelet count recovered, and thrombosis did not relapse. Physicians should consider heparin-induced thrombocytopenia as a cause of ischemic stroke in patients with COVID-19 infection.
Collapse
Affiliation(s)
- Yasutaka Murakami
- Department of Neurology, Osaka University Graduate School of Medicine, Japan
| | - Shuhei Okazaki
- Department of Neurology, Osaka University Graduate School of Medicine, Japan
| | - Makoto Yamamoto
- Department of Respiratory Medicine and Clinical Immunology, Osaka University Graduate School of Medicine, Japan
| | - Rei Sakurai
- Department of Neurology, Osaka University Graduate School of Medicine, Japan
| | - Jyunki Jinno
- Department of Neurology, Osaka University Graduate School of Medicine, Japan
| | - Tatsuhiko Ozono
- Department of Neurology, Osaka University Graduate School of Medicine, Japan
| | - Kensuke Ikenaka
- Department of Neurology, Osaka University Graduate School of Medicine, Japan
| | - Yasufumi Gon
- Department of Neurology, Osaka University Graduate School of Medicine, Japan
| | - Kenichi Todo
- Department of Neurology, Osaka University Graduate School of Medicine, Japan
| | - Tsutomu Sasaki
- Department of Neurology, Osaka University Graduate School of Medicine, Japan
| | - Haruhiko Hirata
- Department of Respiratory Medicine and Clinical Immunology, Osaka University Graduate School of Medicine, Japan
| | - Akinori Uchiyama
- Department of Anesthesiology and Intensive Care Medicine, Osaka University Graduate School of Medicine, Japan
| | - Hideki Mochizuki
- Department of Neurology, Osaka University Graduate School of Medicine, Japan
| |
Collapse
|
47
|
Lecumberri R, Marcos-Jubilar M, Guillén C. Thromboprophylaxis in Patients With Cancer and COVID-19. Arch Bronconeumol 2022; 58:744-745. [PMID: 36153216 PMCID: PMC9452396 DOI: 10.1016/j.arbres.2022.08.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2022] [Accepted: 08/08/2022] [Indexed: 11/02/2022]
Affiliation(s)
- Ramón Lecumberri
- Servicio de Hematología, Clínica Universidad de Navarra, Pamplona, Spain; CIBER-CV, Instituto de Salud Carlos III, Madrid, Spain.
| | | | - Carolina Guillén
- Servicio de Hematología, Clínica Universidad de Navarra, Madrid, Spain
| |
Collapse
|
48
|
Resveratrol Downmodulates Neutrophil Extracellular Trap (NET) Generation by Neutrophils in Patients with Severe COVID-19. Antioxidants (Basel) 2022; 11:antiox11091690. [PMID: 36139764 PMCID: PMC9495554 DOI: 10.3390/antiox11091690] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2022] [Revised: 08/25/2022] [Accepted: 08/26/2022] [Indexed: 11/20/2022] Open
Abstract
The formation of microthrombi in lung autopsies indicates the involvement of NETs in the immunopathogenesis of severe COVID-19. Therefore, supplements inhibiting NET formation, in association with drugs with fewer adverse effects, should be a relevant strategy to attenuate the disease. Resveratrol (RESV) is a natural polyphenol with an important antiviral and antioxidant role. To modulate neutrophils from patients infected with SARS-CoV-2, we evaluated the in vitro effect of RESV on NET formation. Herein, we investigated 190 patients hospitalized with moderate, severe, and critical symptoms at Hospital das Clínicas, Brazil. We observed that neutrophilia in patients with severe COVID-19 infection is composed of neutrophils with activated profile able to release NET spontaneously. Notably, RESV decreased the neutrophil-activated status and the release of free DNA, inhibiting NET formation even under the specific PMA stimulus. At present, there is no evidence of the role of RESV in neutrophils from patients with COVID-19 infection. These findings suggest that adjunctive therapies with RESV may help decrease the inflammation of viral or bacterial infection, improving patient outcomes.
Collapse
|
49
|
COVID-19-Related ARDS: Key Mechanistic Features and Treatments. J Clin Med 2022; 11:jcm11164896. [PMID: 36013135 PMCID: PMC9410336 DOI: 10.3390/jcm11164896] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Revised: 08/16/2022] [Accepted: 08/17/2022] [Indexed: 12/15/2022] Open
Abstract
Acute respiratory distress syndrome (ARDS) is a heterogeneous syndrome historically characterized by the presence of severe hypoxemia, high-permeability pulmonary edema manifesting as diffuse alveolar infiltrate on chest radiograph, and reduced compliance of the integrated respiratory system as a result of widespread compressive atelectasis and fluid-filled alveoli. Coronavirus disease 19 (COVID-19)-associated ARDS (C-ARDS) is a novel etiology caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) that may present with distinct clinical features as a result of the viral pathobiology unique to SARS-CoV-2. In particular, severe injury to the pulmonary vascular endothelium, accompanied by the presence of diffuse microthrombi in the pulmonary microcirculation, can lead to a clinical presentation in which the severity of impaired gas exchange becomes uncoupled from lung capacity and respiratory mechanics. The purpose of this review is to highlight the key mechanistic features of C-ARDS and to discuss the implications these features have on its treatment. In some patients with C-ARDS, rigid adherence to guidelines derived from clinical trials in the pre-COVID era may not be appropriate.
Collapse
|
50
|
Wismüller A, DSouza AM, Abidin AZ, Ali Vosoughi M, Gange C, Cortopassi IO, Bozovic G, Bankier AA, Batra K, Chodakiewitz Y, Xi Y, Whitlow CT, Ponnatapura J, Wendt GJ, Weinberg EP, Stockmaster L, Shrier DA, Shin MC, Modi R, Lo HS, Kligerman S, Hamid A, Hahn LD, Garcia GM, Chung JH, Altes T, Abbara S, Bader AS. Early-stage COVID-19 pandemic observations on pulmonary embolism using nationwide multi-institutional data harvesting. NPJ Digit Med 2022; 5:120. [PMID: 35986059 PMCID: PMC9388980 DOI: 10.1038/s41746-022-00653-2] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Accepted: 07/06/2022] [Indexed: 11/29/2022] Open
Abstract
We introduce a multi-institutional data harvesting (MIDH) method for longitudinal observation of medical imaging utilization and reporting. By tracking both large-scale utilization and clinical imaging results data, the MIDH approach is targeted at measuring surrogates for important disease-related observational quantities over time. To quantitatively investigate its clinical applicability, we performed a retrospective multi-institutional study encompassing 13 healthcare systems throughout the United States before and after the 2020 COVID-19 pandemic. Using repurposed software infrastructure of a commercial AI-based image analysis service, we harvested data on medical imaging service requests and radiology reports for 40,037 computed tomography pulmonary angiograms (CTPA) to evaluate for pulmonary embolism (PE). Specifically, we compared two 70-day observational periods, namely (i) a pre-pandemic control period from 11/25/2019 through 2/2/2020, and (ii) a period during the early COVID-19 pandemic from 3/8/2020 through 5/16/2020. Natural language processing (NLP) on final radiology reports served as the ground truth for identifying positive PE cases, where we found an NLP accuracy of 98% for classifying radiology reports as positive or negative for PE based on a manual review of 2,400 radiology reports. Fewer CTPA exams were performed during the early COVID-19 pandemic than during the pre-pandemic period (9806 vs. 12,106). However, the PE positivity rate was significantly higher (11.6 vs. 9.9%, p < 10-4) with an excess of 92 PE cases during the early COVID-19 outbreak, i.e., ~1.3 daily PE cases more than statistically expected. Our results suggest that MIDH can contribute value as an exploratory tool, aiming at a better understanding of pandemic-related effects on healthcare.
Collapse
Affiliation(s)
- Axel Wismüller
- Department of Imaging Sciences, University of Rochester Medical Center, Rochester, NY, USA
- Department of Biomedical Engineering, University of Rochester Medical Center, Rochester, NY, USA
- Department of Electrical and Computer Engineering, University of Rochester, Rochester, NY, USA
- Faculty of Medicine, Ludwig Maximilian University of Munich, Munich, Germany
| | - Adora M DSouza
- Department of Electrical and Computer Engineering, University of Rochester, Rochester, NY, USA
| | - Anas Z Abidin
- Department of Biomedical Engineering, University of Rochester Medical Center, Rochester, NY, USA
| | - M Ali Vosoughi
- Department of Electrical and Computer Engineering, University of Rochester, Rochester, NY, USA
| | - Christopher Gange
- Department of Radiology & Biomedical Sciences, Yale University School of Medicine, New Haven, CT, USA
| | - Isabel O Cortopassi
- Department of Radiology, Mayo Clinic College of Medicine and Science, Jacksonville, FL, USA
| | - Gracijela Bozovic
- Department of Radiology, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Alexander A Bankier
- Department of Radiology, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Kiran Batra
- Department of Radiology, University of Texas, Southwestern Medical Center, Dallas, TX, USA
| | - Yosef Chodakiewitz
- Department of Imaging, S. Mark Taper Foundation Imaging Center, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Yin Xi
- Department of Radiology, University of Texas, Southwestern Medical Center, Dallas, TX, USA
| | | | | | - Gary J Wendt
- Department of Radiology, University of Wisconsin, Madison, WI, USA
| | - Eric P Weinberg
- Department of Imaging Sciences, University of Rochester Medical Center, Rochester, NY, USA
| | - Larry Stockmaster
- Department of Imaging Sciences, University of Rochester Medical Center, Rochester, NY, USA
| | - David A Shrier
- Department of Imaging Sciences, University of Rochester Medical Center, Rochester, NY, USA
| | - Min Chul Shin
- Department of Radiology, Christiana Care Health System, Newark, DE, USA
| | - Roshan Modi
- Department of Radiology, Christiana Care Health System, Newark, DE, USA
| | - Hao Steven Lo
- Department of Radiology, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Seth Kligerman
- Department of Radiology, University of California, San Diego, San Diego, CA, USA
| | - Aws Hamid
- Emory University School of Medicine, Department of Radiology and Imaging Sciences, Atlanta, GA, USA
| | - Lewis D Hahn
- Department of Radiology, University of California, San Diego, San Diego, CA, USA
| | | | - Jonathan H Chung
- Department of Radiology, University of Chicago, Chicago, IL, USA
| | | | - Suhny Abbara
- Department of Radiology, University of Texas, Southwestern Medical Center, Dallas, TX, USA
| | - Anna S Bader
- Department of Radiology & Biomedical Sciences, Yale University School of Medicine, New Haven, CT, USA.
| |
Collapse
|