1
|
Di Febo R, Saeed Z, Serafini F, Brocco D, D'Ascanio F, Pizzi AD, Tinari N, Crescitelli R, Lanuti P, Renda G. Diagnostic and prognostic roles of endothelial- and platelet-derived extracellular vesicles in cardiovascular diseases. J Transl Med 2025; 23:553. [PMID: 40380176 PMCID: PMC12085008 DOI: 10.1186/s12967-025-06522-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2025] [Accepted: 04/22/2025] [Indexed: 05/19/2025] Open
Abstract
Extracellular vesicles (EVs) are membrane-bound structures released by all cell types. They play a critical role in intercellular communication by transferring their cargo, comprising proteins, lipids, metabolites, RNAs, miRNAs, and DNA fragments, to recipient cells. This transfer influences gene expression, signaling pathways, and cellular behavior. Due to their ability to alter the physiology of recipient cells, EVs hold significant therapeutic potential. Additionally, EVs are implicated in various physiological and pathological processes, including immune regulation, cancer progression, and cardiovascular diseases. EVs have been detected in many biological fluids, such as peripheral blood, saliva, urine, cerebrospinal fluid, and breast milk. The cargo of EVs dynamically reflects the physiological and pathological state of their parent cells, making them promising candidates for liquid biopsies in various clinical conditions. Specifically, different EV subtypes in cardiovascular diseases have been studied, with both endothelial and platelet-derived EVs playing significant roles in cardiovascular pathologies. This review focuses on the diagnostic and prognostic potential of endothelial and platelet-derived EVs in cardiovascular diseases, highlighting the role of EV subpopulations.
Collapse
Affiliation(s)
- Riccardo Di Febo
- Department of Health Sciences, University of Milan, Milan, Italy
| | - Zeeba Saeed
- Department of Medical, Oral and Biotechnological Sciences, G. d'Annunzio University of Chieti-Pescara, 66100, Chieti, Italy
- Center for Advanced Studies and Technology, G. d'Annunzio University of Chieti-Pescara, 66100, Chieti, Italy
| | - Francesco Serafini
- Department of Neuroscience, Imaging and Clinical Sciences, G. d'Annunzio University of Chieti-Pescara, 66100, Chieti, Italy
| | - Davide Brocco
- Department of Medical, Oral and Biotechnological Sciences, G. d'Annunzio University of Chieti-Pescara, 66100, Chieti, Italy
- Center for Advanced Studies and Technology, G. d'Annunzio University of Chieti-Pescara, 66100, Chieti, Italy
| | - Francesca D'Ascanio
- Center for Advanced Studies and Technology, G. d'Annunzio University of Chieti-Pescara, 66100, Chieti, Italy
- Department of Humanities, Law and Economics, Leonardo da Vinci University, 66010, Torrevecchia Teatina (CH), Italy
| | - Andrea Delli Pizzi
- Department of Innovative Technologies in Medicine & Dentistry, G. d'Annunzio University of Chieti-Pescara, 66100, Chieti, Italy
- Institute for Advanced Biomedical Technologies, G. d'Annunzio University of Chieti-Pescara, Chieti, Italy
| | - Nicola Tinari
- Department of Medical, Oral and Biotechnological Sciences, G. d'Annunzio University of Chieti-Pescara, 66100, Chieti, Italy
- Center for Advanced Studies and Technology, G. d'Annunzio University of Chieti-Pescara, 66100, Chieti, Italy
| | - Rossella Crescitelli
- Sahlgrenska Center for Cancer Research, Department of Surgery, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Paola Lanuti
- Center for Advanced Studies and Technology, G. d'Annunzio University of Chieti-Pescara, 66100, Chieti, Italy
- Department of Medicine and Aging Science, G. d'Annunzio University of Chieti-Pescara, 66100, Chieti, Italy
| | - Giulia Renda
- Center for Advanced Studies and Technology, G. d'Annunzio University of Chieti-Pescara, 66100, Chieti, Italy.
- Department of Neuroscience, Imaging and Clinical Sciences, G. d'Annunzio University of Chieti-Pescara, 66100, Chieti, Italy.
| |
Collapse
|
2
|
Protopapas AA, Takardaki A, Protopapa N, Papagiouvanni I, Protopapas AN, Skoura L, Savopoulos C, Goulis I. Microvesicle Tissue Factor Procoagulant Activity Is Elevated and Correlated With Disease Severity in Patients With Cirrhosis. Liver Int 2025; 45:e16192. [PMID: 39601330 PMCID: PMC11907220 DOI: 10.1111/liv.16192] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/02/2024] [Revised: 11/12/2024] [Accepted: 11/17/2024] [Indexed: 11/29/2024]
Abstract
BACKGROUND AND AIMS Tissue factor-expressing microvesicles (MV-TF) have been found to correlate with thrombotic complications in various diseases. Simultaneously, there is expanding research regarding the effect of the coagulation cascade on liver fibrosis progression. The aim of our manuscript was to evaluate MV-TF activity in patients with cirrhosis and its correlation with disease severity. METHODS We prospectively enrolled 82 patients [11 with cirrhosis and hepatocellular cancer (Group 1), 50 with cirrhosis (Group 2) and 21 controls (Group 3)]. Extensive workup for disease staging and exclusion criteria was undertaken. Exclusion criteria included thrombophilia, history of thrombosis, recent hospitalisation, ongoing infection, alcohol dependence, cancer, haematological diseases and use of anticoagulant, antiplatelet or contraceptive drugs. Plasma tissue factor antigen concentration and MV-TF activity were assessed. RESULTS MV-TF showed median values of 4.03 [1.57], 3.17 [1.59] and 2.26 [1.23] pg/mL in Groups 1, 2 and 3, respectively. There was a statistically significant difference between Groups 1 and 3 (p < 0.001) and Groups 2 and 3 (p = 0.003), while Group 1 had higher values than Group 2 without statistical significance (p = 0.088). In Group 2, the patients' Child-Pugh (CP) stage was A in 56%, B in 26% and C in 18% of cases. MV-TF activity significantly correlated with decompensated cirrhosis (p = 0.005) and higher CP stage (p = 0.011). Finally, MV-TF activity significantly correlated with 12-month mortality (p = 0.021). CONCLUSIONS MV-TF activity is elevated in patients with cirrhosis, showing a significant correlation with disease severity. MV-TF may play a role in the procoagulant imbalance of liver cirrhosis and their contribution in disease progression should be studied further.
Collapse
Affiliation(s)
- Adonis A. Protopapas
- First Propaedeutic Department of Internal MedicineAristotle University of Thessaloniki, AHEPA University HospitalThessalonikiGreece
- Fourth Department of Internal MedicineAristotle University of Thessaloniki, Hippokration General HospitalThessalonikiGreece
| | - Anna Takardaki
- Department of MicrobiologyAristotle University of Thessaloniki, AHEPA University HospitalThessalonikiGreece
| | - Nefeli Protopapa
- First Propaedeutic Department of Internal MedicineAristotle University of Thessaloniki, AHEPA University HospitalThessalonikiGreece
| | - Ioanna Papagiouvanni
- Fourth Department of Internal MedicineAristotle University of Thessaloniki, Hippokration General HospitalThessalonikiGreece
| | - Andreas N. Protopapas
- First Propaedeutic Department of Internal MedicineAristotle University of Thessaloniki, AHEPA University HospitalThessalonikiGreece
| | - Lemonia Skoura
- Department of MicrobiologyAristotle University of Thessaloniki, AHEPA University HospitalThessalonikiGreece
| | - Christos Savopoulos
- First Propaedeutic Department of Internal MedicineAristotle University of Thessaloniki, AHEPA University HospitalThessalonikiGreece
| | - Ioannis Goulis
- Fourth Department of Internal MedicineAristotle University of Thessaloniki, Hippokration General HospitalThessalonikiGreece
| |
Collapse
|
3
|
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. J Extracell Vesicles 2025; 14:e70051. [PMID: 40205960 PMCID: PMC11982706 DOI: 10.1002/jev2.70051] [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: 06/07/2024] [Accepted: 02/05/2025] [Indexed: 04/11/2025] Open
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection and resulting coronavirus disease (COVID-19) cause 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. We hypothesized that maternal circulating extracellular vesicles (EVs), altered by COVID-19 during pregnancy, contribute to placental dysfunction. To examine this hypothesis, we characterized circulating EVs from pregnancies complicated by COVID-19 and tested their effects on trophoblast cell physiology in vitro. Trophoblast exposure to EVs isolated from patients with an active infection (AI), but not controls, altered key trophoblast functions including hormone production and invasion. Thus, circulating EVs from participants with an AI, both symptomatic and asymptomatic cases, can disrupt vital trophoblast functions. EV cargo differed between participants with COVID-19, depending on the gestational timing of infection, 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
Affiliation(s)
- Thea N. Golden
- Department of Obstetrics and GynecologyPerelman School of Medicine at the University of PennsylvaniaPhiladelphiaPennsylvaniaUSA
- Center for Women's Health and Reproductive MedicineUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
- Center for Excellence in Environmental ToxicologyUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
| | - Sneha Mani
- Department of Obstetrics and GynecologyPerelman School of Medicine at the University of PennsylvaniaPhiladelphiaPennsylvaniaUSA
- Center for Women's Health and Reproductive MedicineUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
| | - Rebecca L. Linn
- Department of Pathology and Laboratory MedicineChildren's Hospital of PhiladelphiaPhiladelphiaPennsylvaniaUSA
| | - Rita Leite
- Department of Obstetrics and GynecologyPerelman School of Medicine at the University of PennsylvaniaPhiladelphiaPennsylvaniaUSA
- Center for Women's Health and Reproductive MedicineUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
| | - Natalie A. Trigg
- Epigenetics InstitutePerelman School of Medicine at the University of PennsylvaniaPhiladelphiaPennsylvaniaUSA
| | - Annette Wilson
- Department of MedicineUniversity of PittsburghPittsburghPennsylvaniaUSA
| | - Lauren Anton
- Department of Obstetrics and GynecologyPerelman School of Medicine at the University of PennsylvaniaPhiladelphiaPennsylvaniaUSA
- Center for Women's Health and Reproductive MedicineUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
| | - Monica Mainigi
- Department of Obstetrics and GynecologyPerelman School of Medicine at the University of PennsylvaniaPhiladelphiaPennsylvaniaUSA
- Center for Women's Health and Reproductive MedicineUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
| | - Colin C. Conine
- Center for Women's Health and Reproductive MedicineUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
- Epigenetics InstitutePerelman School of Medicine at the University of PennsylvaniaPhiladelphiaPennsylvaniaUSA
- Institute for Regenerative MedicinePerelman School of Medicine at the University of PennsylvaniaPhiladelphiaPennsylvaniaUSA
- Department of GeneticsPerelman School of Medicine at the University of PennsylvaniaPhiladelphiaPennsylvaniaUSA
- Department of PediatricsPerelman School of Medicine at the University of PennsylvaniaPhiladelphiaPennsylvaniaUSA
- Division of NeonatologyChildren's Hospital of PhiladelphiaPhiladelphiaPennsylvaniaUSA
| | - Brett A. Kaufman
- Department of MedicineUniversity of PittsburghPittsburghPennsylvaniaUSA
| | - Jerome F. Strauss
- Department of Obstetrics and GynecologyPerelman School of Medicine at the University of PennsylvaniaPhiladelphiaPennsylvaniaUSA
- Center for Women's Health and Reproductive MedicineUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
| | - Samuel Parry
- Department of Obstetrics and GynecologyPerelman School of Medicine at the University of PennsylvaniaPhiladelphiaPennsylvaniaUSA
- Center for Women's Health and Reproductive MedicineUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
| | - Rebecca A. Simmons
- Center for Women's Health and Reproductive MedicineUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
- Center for Excellence in Environmental ToxicologyUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
- Department of PediatricsPerelman School of Medicine at the University of PennsylvaniaPhiladelphiaPennsylvaniaUSA
- Division of NeonatologyChildren's Hospital of PhiladelphiaPhiladelphiaPennsylvaniaUSA
| |
Collapse
|
4
|
Cui J, Li H, Ye D, Zhang G, Zhang Y, Yang L, Sim MMS, Wood JP, Wei Y, Li Z, Wu C. Inhibiting NINJ1-dependent plasma membrane rupture protects against inflammasome-induced blood coagulation and inflammation. eLife 2025; 12:RP91329. [PMID: 40094828 PMCID: PMC11913443 DOI: 10.7554/elife.91329] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/19/2025] Open
Abstract
Systemic blood coagulation accompanies inflammation during severe infections like sepsis and COVID. We previously established a link between coagulopathy and pyroptosis, a vital defense mechanism against infection. During pyroptosis, the formation of gasdermin-D (GSDMD) pores on the plasma membrane leads to the release of tissue factor (TF)-positive microvesicles (MVs) that are procoagulant. Mice lacking GSDMD release fewer of these procoagulant MVs. However, the specific mechanisms coupling the activation of GSDMD to MV release remain unclear. Plasma membrane rupture (PMR) in pyroptosis was recently reported to be actively mediated by the transmembrane protein Ninjurin-1 (NINJ1). Here, we show that NINJ1 promotes procoagulant MV release during pyroptosis. Haploinsufficiency or glycine inhibition of NINJ1 limited the release of procoagulant MVs and inflammatory cytokines, and partially protected against blood coagulation and lethality triggered by bacterial flagellin. Our findings suggest a crucial role for NINJ1-dependent PMR in inflammasome-induced blood coagulation and inflammation.
Collapse
Affiliation(s)
- Jian Cui
- Saha Cardiovascular Research Center, College of Medicine, University of KentuckyLexingtonUnited States
| | - Hua Li
- Saha Cardiovascular Research Center, College of Medicine, University of KentuckyLexingtonUnited States
| | - Dien Ye
- Saha Cardiovascular Research Center, College of Medicine, University of KentuckyLexingtonUnited States
| | - Guoying Zhang
- Department of Pharmaceutical Sciences, Irma Lerma Rangel School of Pharmacy, Texas A&M UniversityCollege StationUnited States
| | - Yan Zhang
- Department of Pharmaceutical Sciences, Irma Lerma Rangel School of Pharmacy, Texas A&M UniversityCollege StationUnited States
| | - Ling Yang
- Department of Pharmaceutical Sciences, Irma Lerma Rangel School of Pharmacy, Texas A&M UniversityCollege StationUnited States
| | - Martha MS Sim
- Department of Molecular and Cellular Biochemistry, University of KentuckyLexingtonUnited States
| | - Jeremy P Wood
- Saha Cardiovascular Research Center, College of Medicine, University of KentuckyLexingtonUnited States
- Department of Molecular and Cellular Biochemistry, University of KentuckyLexingtonUnited States
- The Gill Heart and Vascular Institute, College of Medicine, University of KentuckyLexingtonUnited States
| | - Yinan Wei
- Department of Pharmaceutical Sciences, Irma Lerma Rangel School of Pharmacy, Texas A&M UniversityCollege StationUnited States
| | - Zhenyu Li
- Department of Pharmaceutical Sciences, Irma Lerma Rangel School of Pharmacy, Texas A&M UniversityCollege StationUnited States
| | - Congqing Wu
- Saha Cardiovascular Research Center, College of Medicine, University of KentuckyLexingtonUnited States
- Department of Microbiology, Immunology, and Molecular Genetics, College of Medicine, University of KentuckyLexingtonUnited States
- Department of Surgery, College of Medicine, University of KentuckyLexingtonUnited States
| |
Collapse
|
5
|
Shakhidzhanov S, Filippova A, Bovt E, Gubkin A, Sukhikh G, Tsarenko S, Spiridonov I, Protsenko D, Zateyshchikov D, Vasilieva E, Kalinskaya A, Dukhin O, Novichkova G, Karamzin S, Serebriyskiy I, Lipets E, Kopnenkova D, Morozova D, Melnikova E, Rumyantsev A, Ataullakhanov F. Severely Ill COVID-19 Patients May Exhibit Hypercoagulability Despite Escalated Anticoagulation. J Clin Med 2025; 14:1966. [PMID: 40142778 PMCID: PMC11943368 DOI: 10.3390/jcm14061966] [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: 02/13/2025] [Revised: 03/07/2025] [Accepted: 03/09/2025] [Indexed: 03/28/2025] Open
Abstract
Introduction: Severely ill COVID-19 patients receiving prophylactic-dose anticoagulation exhibit high rates of thrombosis and mortality. The escalation of anticoagulation also does not reduce mortality and has an uncertain impact on thrombosis rates. The reasons why escalated doses fail to outperform prophylactic doses in reducing risks of thrombosis and death in severely ill COVID-19 patients remain unclear. We hypothesized that escalated anticoagulation would not effectively prevent hypercoagulability and, consequently, would not reduce the risk of thrombosis and death in some severely ill patients. Methods: We conducted a prospective multicenter study that enrolled 3860 COVID-19 patients, including 1654 severely ill. They received different doses of low-molecular-weight or unfractionated heparin, and their blood coagulation was monitored with activated partial thromboplastin time, D-dimer, and Thrombodynamics. A primary outcome was hypercoagulability detected by Thrombodynamics. Blood samples were collected at the trough level of anticoagulation. Results: We found that escalated anticoagulation did not prevent hypercoagulability in 28.3% of severely ill patients at the trough level of the pharmacological activity. Severely ill patients with such hypercoagulability had higher levels of inflammation markers and better creatinine clearance compared to severely ill patients without it. Hypercoagulability detected by Thrombodynamics was associated with a 1.68-fold higher hazard rate for death and a 3.19-fold higher hazard rate for thrombosis. Elevated D-dimer levels were also associated with higher hazard rates for thrombosis and death, while shortened APTTs were not. The simultaneous use of Thrombodynamics and D-dimer data enhanced the accuracy for predicting thrombotic events and fatal outcomes in severely ill patients. Conclusions: Thrombodynamics reliably detects hypercoagulability in COVID-19 patients and can be used in conjunction with D-dimer to assess the risk of thrombosis and death in severely ill patients. The pharmacological effect of LMWH at the trough level might be too low to prevent thrombosis in some severely ill patients with severe inflammation and better creatinine clearance, even if escalated doses are used.
Collapse
Affiliation(s)
- Soslan Shakhidzhanov
- Dmitriy Rogachev National Medical Research Center of Pediatric Hematology, Oncology, and Immunology, 117997 Moscow, Russia; (A.F.); (E.B.); (G.N.); (D.M.); (A.R.)
- Center for Theoretical Problems of Physicochemical Pharmacology, 109029 Moscow, Russia; (I.S.); (S.K.); (I.S.); (E.L.); (E.M.)
| | - Anna Filippova
- Dmitriy Rogachev National Medical Research Center of Pediatric Hematology, Oncology, and Immunology, 117997 Moscow, Russia; (A.F.); (E.B.); (G.N.); (D.M.); (A.R.)
- Center for Theoretical Problems of Physicochemical Pharmacology, 109029 Moscow, Russia; (I.S.); (S.K.); (I.S.); (E.L.); (E.M.)
| | - Elizaveta Bovt
- Dmitriy Rogachev National Medical Research Center of Pediatric Hematology, Oncology, and Immunology, 117997 Moscow, Russia; (A.F.); (E.B.); (G.N.); (D.M.); (A.R.)
- Center for Theoretical Problems of Physicochemical Pharmacology, 109029 Moscow, Russia; (I.S.); (S.K.); (I.S.); (E.L.); (E.M.)
| | - Andrew Gubkin
- Central Clinical Hospital No. 2 Named After N.A.Semashko “RZD-Medicine”, 121359 Moscow, Russia;
| | - Gennady Sukhikh
- National Medical Research Center for Obstetrics, Gynecology and Perinatology Named After Academician V.I.Kulakov, 117997 Moscow, Russia;
| | - Sergey Tsarenko
- City Clinical Hospital No. 52 of Moscow Health Care Department, 123182 Moscow, Russia;
| | - Ilya Spiridonov
- Center for Theoretical Problems of Physicochemical Pharmacology, 109029 Moscow, Russia; (I.S.); (S.K.); (I.S.); (E.L.); (E.M.)
| | - Denis Protsenko
- Moscow Multiprofile Clinical Center “Kommunarka” of Moscow Healthcare Department, 142770 Moscow, Russia; (D.P.); (D.K.)
| | - Dmitriy Zateyshchikov
- City Clinical Hospital No. 51 of Moscow Health Care Department, 121309 Moscow, Russia;
| | - Elena Vasilieva
- City Clinical Hospital No. 23 of Moscow Health Care Department, 109004 Moscow, Russia; (E.V.); (A.K.); (O.D.)
| | - Anna Kalinskaya
- City Clinical Hospital No. 23 of Moscow Health Care Department, 109004 Moscow, Russia; (E.V.); (A.K.); (O.D.)
| | - Oleg Dukhin
- City Clinical Hospital No. 23 of Moscow Health Care Department, 109004 Moscow, Russia; (E.V.); (A.K.); (O.D.)
| | - Galina Novichkova
- Dmitriy Rogachev National Medical Research Center of Pediatric Hematology, Oncology, and Immunology, 117997 Moscow, Russia; (A.F.); (E.B.); (G.N.); (D.M.); (A.R.)
| | - Sergey Karamzin
- Center for Theoretical Problems of Physicochemical Pharmacology, 109029 Moscow, Russia; (I.S.); (S.K.); (I.S.); (E.L.); (E.M.)
| | - Ilya Serebriyskiy
- Center for Theoretical Problems of Physicochemical Pharmacology, 109029 Moscow, Russia; (I.S.); (S.K.); (I.S.); (E.L.); (E.M.)
| | - Elena Lipets
- Center for Theoretical Problems of Physicochemical Pharmacology, 109029 Moscow, Russia; (I.S.); (S.K.); (I.S.); (E.L.); (E.M.)
| | - Daria Kopnenkova
- Moscow Multiprofile Clinical Center “Kommunarka” of Moscow Healthcare Department, 142770 Moscow, Russia; (D.P.); (D.K.)
| | - Daria Morozova
- Dmitriy Rogachev National Medical Research Center of Pediatric Hematology, Oncology, and Immunology, 117997 Moscow, Russia; (A.F.); (E.B.); (G.N.); (D.M.); (A.R.)
- Center for Theoretical Problems of Physicochemical Pharmacology, 109029 Moscow, Russia; (I.S.); (S.K.); (I.S.); (E.L.); (E.M.)
| | - Evgeniya Melnikova
- Center for Theoretical Problems of Physicochemical Pharmacology, 109029 Moscow, Russia; (I.S.); (S.K.); (I.S.); (E.L.); (E.M.)
| | - Alexander Rumyantsev
- Dmitriy Rogachev National Medical Research Center of Pediatric Hematology, Oncology, and Immunology, 117997 Moscow, Russia; (A.F.); (E.B.); (G.N.); (D.M.); (A.R.)
| | - Fazoil Ataullakhanov
- Dmitriy Rogachev National Medical Research Center of Pediatric Hematology, Oncology, and Immunology, 117997 Moscow, Russia; (A.F.); (E.B.); (G.N.); (D.M.); (A.R.)
- Center for Theoretical Problems of Physicochemical Pharmacology, 109029 Moscow, Russia; (I.S.); (S.K.); (I.S.); (E.L.); (E.M.)
| |
Collapse
|
6
|
Liao Y, Liu Y, Li D, Luo S, Huang Y, Wu J, Su J, Yang Y, Wu J, Zhu Z, Yanglan M, Deng H, Wu X, Xu J, Cao F, Cai C, Li Z, Yang R, Deng X, Wei J, Wang L. COVID-19 patient serum-derived extracellular vesicles deliver miR-20b-5p induces neutrophil extracellular traps. Cell Commun Signal 2025; 23:93. [PMID: 39962581 PMCID: PMC11834185 DOI: 10.1186/s12964-025-02095-1] [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: 06/07/2024] [Accepted: 02/08/2025] [Indexed: 02/20/2025] Open
Abstract
BACKGROUND Severe cases of COVID-19 are characterized by an excessive presence of neutrophils. Neutrophil extracellular traps (NETs), released by activated neutrophils due to SARS-CoV-2 infection, contribute to lung epithelial cell death and are key drivers in COVID-19-associated immunothrombosis. However, the mechanism underlying NET formation in COVID-19 remain unclear. METHODS Extracellular vesicles (EVs) were isolated from the serum of COVID-19 patients and healthy volunteers, while neutrophils were isolated from blood samples of healthy volunteers. Neutrophils were treated with EVs, and the formation of NETs was observed. To identify the components responsible for the COVID-19-EVs-induced NET formation, we analyzed the expression profiles of microRNA (miRNAs) in COVID-19-EVs. We identified eight highly expressed miRNAs in COVID-19-EVs and explored their potential roles in COVID-19-EVs-mediated NET formation. Additionally, we explored the role of miR-20b-5p in COVID-19-EVs-induced NET formation. RESULTS In this study, we demonstrate that patients with COVID-19 have a higher concentration of serum EVs (COVID-19-EVs) than healthy controls (Normal-EVs). We also found that COVID-19-EVs are internalized by neutrophils to induced NET formation. Through comprehensive miRNA profiling of COVID-19-EVs versus Normal-EVs, we identified 78 differentially expressed miRNAs, with 27 of these being upregulated and 51 being downregulated. Subsequently, we discovered that COVID-19-EVs that were highly abundant with certain miRNAs promote NET formation. Specifically, miR-20b-5p was found to be the strongest inducer of NET formation of the identified miRNAs. Inhibition of miR-20b-5p resulted in a significant decrease in COVID-19-EVs-mediated induction of NET formation. CONCLUSION Herein, we reveal a previously unknown role of COVID-19-EVs in NET formation, which contributes to COVID-19 progression. This study suggests that miR-20b-5p may serve as a potential therapeutic target for COVID-19 treatment.
Collapse
Affiliation(s)
- Yao Liao
- KingMed School of Laboratory Medicine, The Second Affiliated Hospital, The Affiliated Traditional Chinese Medicine Hospital, Guangzhou Medical University, Guangzhou, 511436, China
| | - Yuheng Liu
- KingMed School of Laboratory Medicine, The Second Affiliated Hospital, The Affiliated Traditional Chinese Medicine Hospital, Guangzhou Medical University, Guangzhou, 511436, China
| | - Dinghao Li
- Department of Parasitology of Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080, China
| | - Shiqi Luo
- Institute of Virology, Helmholtz Centre Munich - German Research Centre for Environmental Health, 85764, Neuherberg, Germany
- Chair for Preventions of Infectious Microbial Diseases, School of Life Sciences, Central Institute of Disease Prevention, Technical University of Munich, 85354, Freising, Germany
| | - Yun Huang
- Department of Parasitology of Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080, China
| | - Junwei Wu
- KingMed School of Laboratory Medicine, The Second Affiliated Hospital, The Affiliated Traditional Chinese Medicine Hospital, Guangzhou Medical University, Guangzhou, 511436, China
| | - Jin Su
- KingMed School of Laboratory Medicine, The Second Affiliated Hospital, The Affiliated Traditional Chinese Medicine Hospital, Guangzhou Medical University, Guangzhou, 511436, China
| | - Yi Yang
- KingMed School of Laboratory Medicine, The Second Affiliated Hospital, The Affiliated Traditional Chinese Medicine Hospital, Guangzhou Medical University, Guangzhou, 511436, China
| | - Ji Wu
- Department of Parasitology of Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080, China
| | - Zifeng Zhu
- Department of Parasitology of Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080, China
| | - Mengxi Yanglan
- KingMed School of Laboratory Medicine, The Second Affiliated Hospital, The Affiliated Traditional Chinese Medicine Hospital, Guangzhou Medical University, Guangzhou, 511436, China
| | - Haiyi Deng
- KingMed School of Laboratory Medicine, The Second Affiliated Hospital, The Affiliated Traditional Chinese Medicine Hospital, Guangzhou Medical University, Guangzhou, 511436, China
| | - Xinyi Wu
- KingMed School of Laboratory Medicine, The Second Affiliated Hospital, The Affiliated Traditional Chinese Medicine Hospital, Guangzhou Medical University, Guangzhou, 511436, China
| | - Junhao Xu
- KingMed School of Laboratory Medicine, The Second Affiliated Hospital, The Affiliated Traditional Chinese Medicine Hospital, Guangzhou Medical University, Guangzhou, 511436, China
| | - Feiyang Cao
- KingMed School of Laboratory Medicine, The Second Affiliated Hospital, The Affiliated Traditional Chinese Medicine Hospital, Guangzhou Medical University, Guangzhou, 511436, China
| | - Chunmei Cai
- KingMed School of Laboratory Medicine, The Second Affiliated Hospital, The Affiliated Traditional Chinese Medicine Hospital, Guangzhou Medical University, Guangzhou, 511436, China
| | - Zhen Li
- KingMed School of Laboratory Medicine, The Second Affiliated Hospital, The Affiliated Traditional Chinese Medicine Hospital, Guangzhou Medical University, Guangzhou, 511436, China
| | - Ruibing Yang
- KingMed School of Laboratory Medicine, The Second Affiliated Hospital, The Affiliated Traditional Chinese Medicine Hospital, Guangzhou Medical University, Guangzhou, 511436, China.
| | - Xiaoyan Deng
- KingMed School of Laboratory Medicine, The Second Affiliated Hospital, The Affiliated Traditional Chinese Medicine Hospital, Guangzhou Medical University, Guangzhou, 511436, China.
| | - Jie Wei
- KingMed School of Laboratory Medicine, The Second Affiliated Hospital, The Affiliated Traditional Chinese Medicine Hospital, Guangzhou Medical University, Guangzhou, 511436, China.
| | - Lifu Wang
- KingMed School of Laboratory Medicine, The Second Affiliated Hospital, The Affiliated Traditional Chinese Medicine Hospital, Guangzhou Medical University, Guangzhou, 511436, China.
| |
Collapse
|
7
|
Suades R, Greco MF, Padró T, de Santisteban V, Domingo P, Benincasa G, Napoli C, Greco S, Madè A, Ranucci M, Devaux Y, Martelli F, Badimon L. Blood CD45 +/CD3 + lymphocyte-released extracellular vesicles and mortality in hospitalized patients with coronavirus disease 2019. Eur J Clin Invest 2025; 55:e14354. [PMID: 39548690 DOI: 10.1111/eci.14354] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/14/2024] [Accepted: 11/05/2024] [Indexed: 11/18/2024]
Abstract
BACKGROUND The global pandemic of coronavirus disease 2019 (COVID-19) represented a major public health concern. Growing evidence shows that plasma of COVID-19 patients contains large numbers of circulating extracellular vesicles (cEVs) that correlate with disease severity and recovery. In this study, we sought to characterize the longitudinal cEV signature in critically ill COVID-19 patients during hospitalization and its relation to mortality risk. METHODS cEVs were quantitatively and phenotypically analysed in hospitalized non-surviving COVID-19 patients at baseline (n = 42) and before exitus (n = 40) and in 40 healthy volunteers as a reference group by high sensitivity nano flow cytometry using specific markers for parental cell sources and activation. RESULTS Levels of cEV subtypes differed between patients with severe COVID-19 and healthy subjects, specifically those from platelets and endothelial, inflammatory and viral infected cells, which associate to high mortality risk. In the longitudinal analysis from baseline to the time point immediately preceding death, no changes were found for platelet, pan-leukocyte, and lung epithelial cell-shed cEVs, while endothelial cell releases of EVs (eEVs) significantly differed. Vascular endothelial growth factor receptor 2-positive eEVs were significantly increased before death compared to admission whereas endoglin and E-selectin-containing eEVs did not change. Conversely, lymphocyte (ℓEV), monocyte, macrophage, pericyte and progenitor cell-derived cEVs displayed significant reductions before exitus. Noteworthy, levels of CD45+/CD3+-ℓEVs were significantly associated to the patient's survival time. CONCLUSIONS An evolving cEV profile able to discriminate prompt risk of death during hospitalization has been defined suggesting a role for circulating and vascular cell-derived EVs in COVID-19 pathogenesis.
Collapse
Affiliation(s)
- Rosa Suades
- Institut de Recerca Sant Pau (IR SANT PAU), Barcelona, Spain
- Centro de Investigación Biomédica en Red Enfermedades Cardiovasculares (CIBERCV), Institute of Health Carlos III (ISCIII), Madrid, Spain
| | - Maria F Greco
- Institut de Recerca Sant Pau (IR SANT PAU), Barcelona, Spain
- Department of Clinical Sciences and Community Health, Università Degli Studi di Milano, Milan, Italy
| | - Teresa Padró
- Institut de Recerca Sant Pau (IR SANT PAU), Barcelona, Spain
- Centro de Investigación Biomédica en Red Enfermedades Cardiovasculares (CIBERCV), Institute of Health Carlos III (ISCIII), Madrid, Spain
| | | | - Pere Domingo
- Infectious Diseases Unit, Department of Internal Medicine, Hospital de la Santa Creu i Sant Pau-IR SANT PAU, Universitat Autònoma de Barcelona (UAB), Barcelona, Spain
| | - Giuditta Benincasa
- Institut de Recerca Sant Pau (IR SANT PAU), Barcelona, Spain
- Department of Advanced Medical and Surgical Sciences (DAMSS), University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Claudio Napoli
- Department of Advanced Medical and Surgical Sciences (DAMSS), University of Campania "Luigi Vanvitelli", Naples, Italy
- Department of Internal Medicine and Specialistics, Division of Clinical Immunology, Immunohematology, Transfusion Medicine and Transplant Immunology (SIMT), Azienda Universitaria Policlinico (AOU), Naples, Italy
| | - Simona Greco
- Molecular Cardiology Laboratory, IRCCS Policlinico San Donato, Milan, Italy
| | - Alisia Madè
- Molecular Cardiology Laboratory, IRCCS Policlinico San Donato, Milan, Italy
| | - Marco Ranucci
- Department of Cardiovascular Anesthesia and Intensive Care, IRCCS Policlinico San Donato, Milan, Italy
| | - Yvan Devaux
- Cardiovascular Research Unit, Department of Precision Health, Luxembourg Institute of Health, Strassen, Luxembourg
| | - Fabio Martelli
- Molecular Cardiology Laboratory, IRCCS Policlinico San Donato, Milan, Italy
| | - Lina Badimon
- Institut de Recerca Sant Pau (IR SANT PAU), Barcelona, Spain
- Centro de Investigación Biomédica en Red Enfermedades Cardiovasculares (CIBERCV), Institute of Health Carlos III (ISCIII), Madrid, Spain
- Cardiovascular Research Chair, UAB, Barcelona, Spain
| |
Collapse
|
8
|
Ahmed O, King NE, Qureshi MA, Choudhry AA, Osama M, Zehner C, Ali A, Hamzeh IR, Palaskas NL, Thompson KA, Koutroumpakis E, Deswal A, Yusuf SW. Non-bacterial thrombotic endocarditis: a clinical and pathophysiological reappraisal. Eur Heart J 2025; 46:236-249. [PMID: 39565324 DOI: 10.1093/eurheartj/ehae788] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/06/2024] [Revised: 08/11/2024] [Accepted: 10/30/2024] [Indexed: 11/21/2024] Open
Abstract
Non-bacterial thrombotic endocarditis (NBTE), formerly recognized as marantic endocarditis, represents a rare cardiovascular pathology intricately linked with hypercoagulable states, notably malignancy and autoimmune disorders. Characterized by the development of sterile vegetations comprised of fibrin and platelets on cardiac valves, NBTE poses a diagnostic challenge due to its resemblance to infective endocarditis. Therapeutic endeavours primarily revolve around addressing the underlying aetiology and instituting anticoagulant regimens to forestall embolic events, with surgical intervention seldom warranted. Non-bacterial thrombotic endocarditis frequently coexists with malignancies and autoimmune conditions, such as lupus and antiphospholipid antibody syndrome, and, more recently, has been associated with COVID-19. Its pathogenesis is underpinned by a complex interplay of endothelial dysfunction, hypercoagulability, hypoxia, and immune complex deposition. Clinical manifestations typically manifest as embolic phenomena, particularly cerebrovascular accidents, bearing substantial mortality rates. Diagnosis necessitates a high index of suspicion and meticulous exclusion of infective endocarditis, often facilitated by advanced cardiac imaging modalities. Anticoagulation, typically employing low molecular weight heparin or warfarin, constitutes the cornerstone of pharmacological intervention. Surgical recourse may be warranted in instances of refractory heart failure or recurrent embolic events. Given its multifaceted nature, the management of NBTE mandates a multidisciplinary approach, with prognosis contingent upon individual clinical intricacies. Future endeavours should prioritize further research to refine therapeutic strategies and enhance patient outcomes.
Collapse
Affiliation(s)
- Omair Ahmed
- Department of Internal Medicine, Henry Ford Jackson Hospital, Jackson, MI 49201, USA
| | - Nicholas E King
- Division of Cardiovascular Medicine, McGovern Medical School, University of Texas Health Sciences Center at Houston, Houston, TX 77030, USA
| | | | - Abira Afzal Choudhry
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Muhammad Osama
- Department of Internal Medicine, Rochester Regional Health, Rochester, NY 14617, USA
| | - Carl Zehner
- Department of Cardiology, University of Texas Health Sciences Center, MD Anderson Cancer Center, 1515 Holcombe Blvd, Houston, TX 77030, USA
| | - Abdelrahman Ali
- Department of Cardiology, University of Texas Health Sciences Center, MD Anderson Cancer Center, 1515 Holcombe Blvd, Houston, TX 77030, USA
| | - Ihab R Hamzeh
- Department of Cardiology, University of Texas Health Sciences Center, MD Anderson Cancer Center, 1515 Holcombe Blvd, Houston, TX 77030, USA
| | - Nicolas L Palaskas
- Department of Cardiology, University of Texas Health Sciences Center, MD Anderson Cancer Center, 1515 Holcombe Blvd, Houston, TX 77030, USA
| | - Kara A Thompson
- Department of Cardiology, University of Texas Health Sciences Center, MD Anderson Cancer Center, 1515 Holcombe Blvd, Houston, TX 77030, USA
| | - Efstratios Koutroumpakis
- Department of Cardiology, University of Texas Health Sciences Center, MD Anderson Cancer Center, 1515 Holcombe Blvd, Houston, TX 77030, USA
| | - Anita Deswal
- Department of Cardiology, University of Texas Health Sciences Center, MD Anderson Cancer Center, 1515 Holcombe Blvd, Houston, TX 77030, USA
| | - Syed Wamique Yusuf
- Department of Cardiology, University of Texas Health Sciences Center, MD Anderson Cancer Center, 1515 Holcombe Blvd, Houston, TX 77030, USA
| |
Collapse
|
9
|
Cui J, Li H, Ye D, Zhang G, Zhang Y, Yang L, Sim MM, Wood JP, Wei Y, Li Z, Wu C. Inhibiting NINJ1-dependent plasma membrane rupture protects against inflammasome-induced blood coagulation and inflammation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.08.30.555561. [PMID: 37693519 PMCID: PMC10491273 DOI: 10.1101/2023.08.30.555561] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/12/2023]
Abstract
Systemic blood coagulation accompanies inflammation during severe infection like sepsis and COVID. We've previously established a link between pyroptosis, a vital defense mechanism against infection, and coagulopathy. During pyroptosis, the formation of gasdermin-D (GSDMD) pores on the plasma membrane leads to the release of tissue factor (TF)-positive microvesicles (MVs) that are procoagulant. Mice lacking GSDMD release fewer TF MVs. However, the specific mechanisms leading from activation of GSDMD to MV release remain unclear. Plasma membrane rupture (PMR) in pyroptosis was recently reported to be actively mediated by the transmembrane protein Ninjurin-1 (NINJ1). Here we show that NINJ1 promotes procoagulant MV release during pyroptosis. Haploinsuffciency or glycine inhibition of NINJ1 limited the release of procoagulant MVs and inflammatory cytokines and partially protected against blood coagulation and lethality triggered by bacterial flagellin. Our findings suggest a crucial role for NINJ1-dependent PMR in inflammasome-induced blood coagulation and inflammation.
Collapse
Affiliation(s)
- Jian Cui
- Saha Cardiovascular Research Center, College of Medicine, University of Kentucky, Lexington, KY
| | - Hua Li
- Saha Cardiovascular Research Center, College of Medicine, University of Kentucky, Lexington, KY
| | - Dien Ye
- Saha Cardiovascular Research Center, College of Medicine, University of Kentucky, Lexington, KY
| | - Guoying Zhang
- Department of Pharmaceutical Sciences, Irma Lerma Rangel School of Pharmacy, Texas A&M University, College Station, TX
| | - Yan Zhang
- Department of Pharmaceutical Sciences, Irma Lerma Rangel School of Pharmacy, Texas A&M University, College Station, TX
| | - Ling Yang
- Department of Pharmaceutical Sciences, Irma Lerma Rangel School of Pharmacy, Texas A&M University, College Station, TX
| | - Martha M.S. Sim
- Department of Molecular and Cellular Biochemistry, University of Kentucky, Lexington, KY
| | - Jeremy P. Wood
- Department of Molecular and Cellular Biochemistry, University of Kentucky, Lexington, KY
- Department of Internal Medicine, College of Medicine, University of Kentucky, Lexington, KY
| | - Yinan Wei
- Department of Pharmaceutical Sciences, Irma Lerma Rangel School of Pharmacy, Texas A&M University, College Station, TX
| | - Zhenyu Li
- Department of Pharmaceutical Sciences, Irma Lerma Rangel School of Pharmacy, Texas A&M University, College Station, TX
| | - Congqing Wu
- Saha Cardiovascular Research Center, College of Medicine, University of Kentucky, Lexington, KY
- Department of Surgery, College of Medicine, University of Kentucky, Lexington, KY
- Department of Microbiology, Immunology, and Molecular Genetics, College of Medicine, University of Kentucky, Lexington, KY
| |
Collapse
|
10
|
Roytenberg R, Yue H, DeHart A, Kim E, Bai F, Kim Y, Denning K, Kwei A, Zhang Q, Liu J, Zheng XL, Li W. Thymidine phosphorylase mediates SARS-CoV-2 spike protein enhanced thrombosis in K18-hACE2 TG mice. Thromb Res 2024; 244:109195. [PMID: 39442286 PMCID: PMC11585440 DOI: 10.1016/j.thromres.2024.109195] [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: 07/06/2024] [Revised: 09/09/2024] [Accepted: 10/17/2024] [Indexed: 10/25/2024]
Abstract
INTRODUCTION Thymidine phosphorylase (TYMP), which facilitates platelet activation and thrombosis, is significantly increased in COVID-19 patients. We hypothesize that TYMP mediates SARS-CoV-2 spike protein (SP)-induced thrombosis. MATERIALS AND METHODS Plasmids encoding wildtype SP or empty vector (p3.1) were transfected into COS-7 cells, and cell lysates were prepared as a reservoir for SP or p3.1 (control), respectively. K18-hACE2TG and K18-hACE2TG/Tymp-/- mice were treated with a single dose of SP or p3.1 by intraperitoneal injection and then subjected to thrombosis studies three days later. The role of SP on inflammatory signaling activation was assessed in BEAS-2B cells. RESULTS SARS-CoV-2 SP increased the expression of TYMP, resulting in the activation of STAT3 and NF-κB in BEAS-2B cells. A siRNA-mediated knockdown of TYMP attenuated SP-enhanced activation of STAT3. SP significantly promoted arterial thrombosis in K18-hACE2TG mice. SP-accelerated thrombosis was attenuated by inhibition or genetic ablation of TYMP. SP treatment did not influence ADP- or collagen-induced platelet aggregation but significantly increased platelet adhesion to fibrinogen. SP treatment also significantly shortened activated partial thromboplastin time, which was reversed and even prolonged by TYMP deficiency. Additionally, SP binds to platelet factor 4 (PF4) and TYMP. TYMP does not bind PF4 but enhances the formation of the SP/PF4 complex, which may augment the procoagulant and prothrombotic effect of PF4. CONCLUSIONS We conclude that SP is prothrombotic and upregulates TYMP expression, and TYMP inhibition or knockout mitigates SP-enhanced thrombosis. These findings suggest that inhibition of TYMP may be a novel therapeutic strategy for COVID-19-associated thrombosis.
Collapse
Affiliation(s)
- Renat Roytenberg
- Department of Biomedical Sciences, Joan C. Edwards School of Medicine at Marshall University, Huntington, WV 25755, USA
| | - Hong Yue
- Department of Biomedical Sciences, Joan C. Edwards School of Medicine at Marshall University, Huntington, WV 25755, USA
| | - Autumn DeHart
- Department of Biomedical Sciences, Joan C. Edwards School of Medicine at Marshall University, Huntington, WV 25755, USA
| | - Eugene Kim
- Department of Chemistry, College of Sciences, Marshall University, Huntington, WV 25755, USA
| | - Fang Bai
- Department of Biomedical Sciences, Joan C. Edwards School of Medicine at Marshall University, Huntington, WV 25755, USA
| | - Yongick Kim
- Department of Chemistry, College of Sciences, Marshall University, Huntington, WV 25755, USA
| | - Krista Denning
- Department of Pathology, Joan C. Edwards School of Medicine at Marshall University, Huntington, WV 25755, USA
| | - Alec Kwei
- Department of Biomedical Sciences, Joan C. Edwards School of Medicine at Marshall University, Huntington, WV 25755, USA
| | - Quan Zhang
- Department of Pathology and Laboratory Medicine, The University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Jiang Liu
- Department of Biomedical Sciences, Joan C. Edwards School of Medicine at Marshall University, Huntington, WV 25755, USA
| | - X Long Zheng
- Department of Pathology and Laboratory Medicine, The University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Wei Li
- Department of Biomedical Sciences, Joan C. Edwards School of Medicine at Marshall University, Huntington, WV 25755, USA.
| |
Collapse
|
11
|
Qian H, Zang R, Zhang R, Zheng G, Qiu G, Meng J, Wang J, Xia J, Huang R, Le Z, Shu Q, Xu J. Circulating extracellular vesicles from severe COVID-19 patients induce lung inflammation. mSphere 2024; 9:e0076424. [PMID: 39475319 PMCID: PMC11580465 DOI: 10.1128/msphere.00764-24] [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: 09/09/2024] [Accepted: 10/11/2024] [Indexed: 11/22/2024] Open
Abstract
Circulating extracellular vesicles (EVs) have been associated with the development of COVID-19 due to their roles in viral infection, inflammatory response, and thrombosis. However, the direct induction of lung inflammation by circulating EVs from severe COVID-19 patients remains unknown. EVs were extracted from the plasma of severe COVID-19 patients admitted to intensive care and healthy controls. To study the effect of COVID-19 EVs on lung inflammation, mice were intratracheally instilled with EVs. To examine the proinflammatory effects of EVs in vitro, bone marrow-derived macrophages were treated with EVs. COVID-19 but not control EVs triggered lung inflammation, as assessed by total protein level, total cell count, neutrophil count, and levels of proinflammatory cytokines in the bronchoalveolar lavage. COVID-19 EVs also promoted M1 polarization of alveolar macrophages in vivo. Treatment of bone marrow-derived macrophages with COVID-19 EVs enhanced the M1 phenotype and augmented the production of IL-1β, IL-6, and TNF-α. In summary, circulating EVs from severe COVID-19 patients induce lung inflammation in mice. EVs could become a potential therapeutic target for alleviating lung injury in COVID-19. IMPORTANCE Extracellular vesicles (EVs) have been reported to facilitate cytokine storm, coagulation, vascular dysfunction, and the spread of the virus in COVID-19. The direct role of circulating EVs from severe COVID-19 patients in lung injury remains unrecognized. Our study demonstrated that plasma EVs obtained from severe COVID-19 patients induced lung inflammation and polarization of alveolar macrophages in vivo. In vitro experiments also revealed the proinflammatory effects of COVID-19 EVs. The present study sheds fresh insight into the mechanisms of COVID-19-induced lung injury, highlighting EVs as a potential therapeutic target in combating the disease.
Collapse
Affiliation(s)
- Huifeng Qian
- Shaoxing Second Hospital, Shaoxing, Zhejiang, China
| | - Ruoxi Zang
- National Clinical Research Center for Child Health, The Children’s Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Ruoyang Zhang
- National Clinical Research Center for Child Health, The Children’s Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | | | - Guanguan Qiu
- Shaoxing Second Hospital, Shaoxing, Zhejiang, China
| | - Jianbiao Meng
- Tongde Hospital of Zhejiang, Hangzhou, Zhejiang, China
| | - Jiangmei Wang
- National Clinical Research Center for Child Health, The Children’s Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Jie Xia
- National Clinical Research Center for Child Health, The Children’s Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Ruoqiong Huang
- National Clinical Research Center for Child Health, The Children’s Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Zhenkai Le
- National Clinical Research Center for Child Health, The Children’s Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Qiang Shu
- National Clinical Research Center for Child Health, The Children’s Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Jianguo Xu
- Shaoxing Second Hospital, Shaoxing, Zhejiang, China
- National Clinical Research Center for Child Health, The Children’s Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| |
Collapse
|
12
|
Bonifay A, Cointe S, Plantureux L, Lacroix R, Dignat-George F. Update on Tissue Factor Detection in Blood in 2024: A Narrative Review. Hamostaseologie 2024; 44:368-376. [PMID: 39442509 DOI: 10.1055/a-2381-6854] [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: 10/25/2024] Open
Abstract
Tissue factor (TF) is a transmembrane protein essential for hemostasis. Different forms of active TF circulate in the blood, either as a component of blood cells and extracellular vesicles (EVs) or as a soluble plasma protein. Accumulating experimental and clinical evidence suggests that TF plays an important role in thrombosis. Many in-house and commercially available assays have been developed to measure TF-dependent procoagulant activity or antigen in blood and have shown promising results for the prediction of disease outcomes or the occurrence of thrombosis events in diseases such as cancer or infectious coagulopathies. This review addresses the different assays that have been published for measuring circulating TF antigen and/or activity in whole blood, cell-free plasma, and EVs and discusses the main preanalytical and analytical parameters that impact results and their interpretation, highlighting their strengths and limitations. In the recent decade, EVTF assays have been significantly developed. Among them, functional assays that use a blocking anti-TF antibody or immunocapture to measure EVTF activity have higher specificity and sensitivity than antigen assays. However, there is still a high variability between assays. Standardization and automatization are prerequisites for the measurement of EVTF in clinical laboratories.
Collapse
Affiliation(s)
- Amandine Bonifay
- Aix-Marseille University, C2VN, INSERM 1263, INRAE 1260, Marseille, France
- Department of Hematology, Biogénopôle, CHU La Timone, APHM, Marseille, France
| | - Sylvie Cointe
- Aix-Marseille University, C2VN, INSERM 1263, INRAE 1260, Marseille, France
- Department of Hematology, Biogénopôle, CHU La Timone, APHM, Marseille, France
| | - Léa Plantureux
- Aix-Marseille University, C2VN, INSERM 1263, INRAE 1260, Marseille, France
| | - Romaric Lacroix
- Aix-Marseille University, C2VN, INSERM 1263, INRAE 1260, Marseille, France
- Department of Hematology, Biogénopôle, CHU La Timone, APHM, Marseille, France
| | - Françoise Dignat-George
- Aix-Marseille University, C2VN, INSERM 1263, INRAE 1260, Marseille, France
- Department of Hematology, Biogénopôle, CHU La Timone, APHM, Marseille, France
| |
Collapse
|
13
|
Bonifay A, Mackman N, Hisada Y, Sachetto ATA, Hau C, Gray E, Hogwood J, Aharon A, Badimon L, Barile L, Baudar J, Beckmann L, Benedikter B, Bolis S, Bouriche T, Brambilla M, Burrello J, Camera M, Campello E, Ettelaie C, Faille D, Featherby S, Franco C, Guldenpfennig M, Hansen JB, Judicone C, Kim Y, Kristensen SR, Laakmann K, Langer F, Latysheva N, Lucien F, de Menezes EM, Mullier F, Norris P, Nybo J, Orbe J, Osterud B, Paramo JA, Radu CM, Roncal C, Samadi N, Snir O, Suades R, Wahlund C, Chareyre C, Abdili E, Martinod K, Thaler J, Dignat-George F, Nieuwland R, Lacroix R. Comparison of assays measuring extracellular vesicle tissue factor in plasma samples: communication from the ISTH SSC Subcommittee on Vascular Biology. J Thromb Haemost 2024; 22:2910-2921. [PMID: 38925490 DOI: 10.1016/j.jtha.2024.05.037] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2023] [Revised: 04/23/2024] [Accepted: 05/23/2024] [Indexed: 06/28/2024]
Abstract
BACKGROUND Scientific and clinical interest in extracellular vesicles (EVs) is growing. EVs that expose tissue factor (TF) bind factor VII/VIIa and can trigger coagulation. Highly procoagulant TF-exposing EVs are detectable in the circulation in various diseases, such as sepsis, COVID-19, or cancer. Many in-house and commercially available assays have been developed to measure EV-TF activity and antigen, but only a few studies have compared some of these assays. OBJECTIVES The International Society on Thrombosis and Haemostasis Scientific and Standardization Committee Subcommittee on Vascular Biology initiated a multicenter study to compare the sensitivity, specificity, and reproducibility of these assays. METHODS Platelet-depleted plasma samples were prepared from blood of healthy donors. The plasma samples were spiked either with EVs from human milk or EVs from TF-positive and TF-negative cell lines. Plasma was also prepared from whole human blood with or without lipopolysaccharide stimulation. Twenty-one laboratories measured EV-TF activity and antigen in the prepared samples using their own assays representing 18 functional and 9 antigenic assays. RESULTS There was a large variability in the absolute values for the different EV-TF activity and antigen assays. Activity assays had higher specificity and sensitivity compared with antigen assays. In addition, there was a large intra-assay and interassay variability. Functional assays that used a blocking anti-TF antibody or immunocapture were the most specific and sensitive. Activity assays that used immunocapture had a lower coefficient of variation compared with assays that isolated EVs by high-speed centrifugation. CONCLUSION Based on this multicenter study, we recommend measuring EV-TF using a functional assay in the presence of an anti-TF antibody.
Collapse
Affiliation(s)
- Amandine Bonifay
- Aix-Marseille University, C2VN, INSERM 1263, INRAE1260, Marseille, France; Department of Hematology and Vascular Biology, CHU La Conception, APHM, Marseille, France
| | - Nigel Mackman
- UNC Blood Research Center, Division of Hematology, Department of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Yohei Hisada
- UNC Blood Research Center, Division of Hematology, Department of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Ana Teresa Azevedo Sachetto
- UNC Blood Research Center, Division of Hematology, Department of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Chi Hau
- Laboratory of Experimental Clinical Chemistry, and Amsterdam Vesicle Center, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands
| | - Elaine Gray
- National Institute for Biological Standards and Control, Potter's Bar, Hertfordshire, United Kingdom
| | - John Hogwood
- National Institute for Biological Standards and Control, Potter's Bar, Hertfordshire, United Kingdom
| | - Anat Aharon
- Hematology Research Laboratory, Department of Hematology, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel; The Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Lina Badimon
- Cardiovascular ICCC Program, Research Institute Hospital de la Santa Creu i Sant Pau, IIB-Sant Pau, Barcelona, Spain; Centro de Investigación Biomédica en Red Cardiovascular (CIBERCV), Instituto de Salud Carlos III, Madrid, Spain
| | - Lucio Barile
- Cardiovascular Theranostics, Istituto Cardiocentro Ticino, Laboratories for Translational Research, Ente Ospedaliero Cantonale, Bellinzona, Switzerland; Faculty of Biomedical Sciences, Università della Svizzera Italiana, Lugano, Switzerland
| | - Justine Baudar
- Université Catholique de Louvain, CHU UCL NAMUR, Namur Thrombosis and Hemostasis Center (NTHC), Yvoir, Belgium
| | - Lennart Beckmann
- Department of Hematology and Oncology, University Cancer Center Hamburg (UCCH), University Medical Center Eppendorf, Hamburg, Germany
| | - Birke Benedikter
- Institute for Lung Research, Universities of Giessen and Marburg Lung Centre, Philipps-University Marburg, German Center for Lung Research (DZL), Marburg, Germany; University Eye Clinic Maastricht, MHeNs School for Mental Health and Neuroscience, Maastricht University Medical Center + (MUMC+), Maastricht, the Netherlands
| | - Sara Bolis
- Cardiovascular Theranostics, Istituto Cardiocentro Ticino, Laboratories for Translational Research, Ente Ospedaliero Cantonale, Bellinzona, Switzerland
| | - Tarik Bouriche
- Research and Technology Department, BioCytex, Marseille, France
| | | | - Jacopo Burrello
- Cardiovascular Theranostics, Istituto Cardiocentro Ticino, Laboratories for Translational Research, Ente Ospedaliero Cantonale, Bellinzona, Switzerland
| | - Marina Camera
- Centro Cardiologico Monzino IRCCS, Milan, Italy; Department of Pharmaceutical Sciences, Università degli Studi di Milano, Milan, Italy
| | - Elena Campello
- Department of Medicine, University of Padova, Padua, Italy
| | - Camille Ettelaie
- Biomedical Science, University of Hull/HYMS, Cottingham Road, Hull, United Kingdom
| | - Dorothée Faille
- Université Paris Cité and Université Sorbonne Paris Nord, INSERM, LVTS, Paris, France; Laboratoire d'Hématologie, AP-HP, Hôpital Bichat, Paris, France
| | - Sophie Featherby
- Biomedical Science, University of Hull/HYMS, Cottingham Road, Hull, United Kingdom
| | - Corentin Franco
- Research and Technology Department, BioCytex, Marseille, France
| | - Maite Guldenpfennig
- Université Catholique de Louvain, CHU UCL NAMUR, Namur Thrombosis and Hemostasis Center (NTHC), Yvoir, Belgium
| | - John-Bjarne Hansen
- Thrombosis Research Group (TREC), Institute of Clinical Medicine, UiT - The Arctic University of Norway, Tromsø, Norway; Thrombosis Research Center (TREC), Division of Internal Medicine, University Hospital of North Norway, Tromsø, Norway
| | | | - Yohan Kim
- epartment of Urology, Department of Immunology, Mayo Clinic, Rochester, Minnesota, USA
| | - Soren Risom Kristensen
- Department of Clinical Biochemistry, Aalborg University Hospital, Aalborg, Denmark; Department of Clinical Medicine, Aalborg University, Aalborg, Denmark
| | - Katrin Laakmann
- Institute for Lung Research, Universities of Giessen and Marburg Lung Centre, Philipps-University Marburg, German Center for Lung Research (DZL), Marburg, Germany
| | - Florian Langer
- Department of Hematology and Oncology, University Cancer Center Hamburg (UCCH), University Medical Center Eppendorf, Hamburg, Germany
| | - Nadezhda Latysheva
- Thrombosis Research Group (TREC), Institute of Clinical Medicine, UiT - The Arctic University of Norway, Tromsø, Norway
| | - Fabrice Lucien
- epartment of Urology, Department of Immunology, Mayo Clinic, Rochester, Minnesota, USA
| | - Erika Marques de Menezes
- Vitalant Research Institute, San Francisco, California, USA; Department of Laboratory Medicine, University of California, San Francisco, California, USA
| | - François Mullier
- Université Catholique de Louvain, CHU UCL NAMUR, Namur Thrombosis and Hemostasis Center (NTHC), Yvoir, Belgium
| | - Philip Norris
- Vitalant Research Institute, San Francisco, California, USA; Department of Laboratory Medicine, University of California, San Francisco, California, USA; Department of Medicine, UCSF, San Francisco, California, USA
| | - Jette Nybo
- Department of Clinical Biochemistry, Aalborg University Hospital, Aalborg, Denmark
| | - Josune Orbe
- Laboratory of Atherothrombosis, Program of Cardiovascular Diseases, Cima Universidad de Navarra, Pamplona, Spain; IdiSNA, Instituto de Investigación Sanitaria de Navarra, Pamplona, Spain; RICORS-Cerebrovascular Diseases, Instituto de Salud Carlos III, Madrid, Spain
| | - Bjarne Osterud
- Thrombosis Research Group (TREC), Institute of Clinical Medicine, UiT - The Arctic University of Norway, Tromsø, Norway
| | - Jose A Paramo
- Centro de Investigación Biomédica en Red Cardiovascular (CIBERCV), Instituto de Salud Carlos III, Madrid, Spain; Laboratory of Atherothrombosis, Program of Cardiovascular Diseases, Cima Universidad de Navarra, Pamplona, Spain; IdiSNA, Instituto de Investigación Sanitaria de Navarra, Pamplona, Spain
| | - Claudia M Radu
- Department of Medicine, University of Padova, Padua, Italy
| | - Carmen Roncal
- Centro de Investigación Biomédica en Red Cardiovascular (CIBERCV), Instituto de Salud Carlos III, Madrid, Spain; Laboratory of Atherothrombosis, Program of Cardiovascular Diseases, Cima Universidad de Navarra, Pamplona, Spain; IdiSNA, Instituto de Investigación Sanitaria de Navarra, Pamplona, Spain; RICORS-Cerebrovascular Diseases, Instituto de Salud Carlos III, Madrid, Spain
| | - Nazanin Samadi
- Clinical Division of Hematology and Hemostaseology, Department of Medicine I, Medical University of Vienna, Vienna, Austria
| | - Omri Snir
- Thrombosis Research Group (TREC), Institute of Clinical Medicine, UiT - The Arctic University of Norway, Tromsø, Norway
| | - Rosa Suades
- Cardiovascular ICCC Program, Research Institute Hospital de la Santa Creu i Sant Pau, IIB-Sant Pau, Barcelona, Spain; Centro de Investigación Biomédica en Red Cardiovascular (CIBERCV), Instituto de Salud Carlos III, Madrid, Spain
| | - Casper Wahlund
- Thrombosis Research Group (TREC), Institute of Clinical Medicine, UiT - The Arctic University of Norway, Tromsø, Norway
| | - Corinne Chareyre
- Aix-Marseille University, C2VN, INSERM 1263, INRAE1260, Marseille, France
| | - Evelyne Abdili
- Department of Hematology and Vascular Biology, CHU La Conception, APHM, Marseille, France
| | - Kimberly Martinod
- Center for Molecular and Vascular Biology, Department of Cardiovascular Sciences, KU Leuven, Leuven, Belgium
| | - Johannes Thaler
- Clinical Division of Hematology and Hemostaseology, Department of Medicine I, Medical University of Vienna, Vienna, Austria
| | - Françoise Dignat-George
- Aix-Marseille University, C2VN, INSERM 1263, INRAE1260, Marseille, France; Department of Hematology and Vascular Biology, CHU La Conception, APHM, Marseille, France.
| | - Rienk Nieuwland
- Laboratory of Experimental Clinical Chemistry, and Amsterdam Vesicle Center, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands
| | - Romaric Lacroix
- Aix-Marseille University, C2VN, INSERM 1263, INRAE1260, Marseille, France; Department of Hematology and Vascular Biology, CHU La Conception, APHM, Marseille, France
| |
Collapse
|
14
|
Wallensten J, Havervall S, Power Y, Åsberg M, Borg K, Nager A, Thålin C, Mobarrez F. Oneyear longitudinal study on biomarkers of blood-brain barrier permeability in COVID-19 patients. Sci Rep 2024; 14:22735. [PMID: 39349618 PMCID: PMC11442946 DOI: 10.1038/s41598-024-73321-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2023] [Accepted: 09/16/2024] [Indexed: 10/04/2024] Open
Abstract
The pathophysiology behind neurological and cognitive sequelae of COVID-19 may be related to dysfunction of the blood-brain barrier (BBB) and previous research indicate transient neuronal injury and glial activation. The aim of this study was to investigate if COVID-19 is related to increased BBB permeability by analyzing leakage of biomarkers such as astrocyte-derived extracellular vesicles (EVs) and S100B. We also investigated whether levels of these biomarkers correlated with self-reported symptoms that persisted > 2 months. The samples in this 1-year follow-up study came from an ongoing longitudinal study of unvaccinated patients hospitalized for COVID-19 at Danderyd University Hospital, Stockholm, Sweden, between April and June 2020. Blood samples were collected at baseline and 4, 8, and 12 months after hospitalization. Information on self-reported clinical symptoms was collected at follow-up visits. A total of 102 patients were enrolled, and 47 completed all follow-up measurements. Peak levels of both biomarkers were observed at 4 months in the subset of 55 patients who were measured at this timepoint. At 12 months, the biomarkers had returned to baseline levels. The biomarkers were not correlated with any of the long-term self-reported symptoms. COVID-19 is associated with transient increased BBB permeability, shown by elevated levels of astrocyte biomarkers in plasma. However, these levels return to baseline 12 months post-infection and do not correlate with long-term symptoms. Further research is needed to unravel the underlying mechanisms causing long-term symptoms in COVID-19 patients.
Collapse
Affiliation(s)
- Johanna Wallensten
- Academic Primary Health Care Centre, Region Stockholm, Solnavägen 1E, Box 45436, 104 31, Stockholm, Sweden.
- Department of Clinical Sciences, Karolinska Institute, Danderyd University Hospital, 18288, Stockholm, Sweden.
| | - Sebastian Havervall
- Department of Clinical Sciences, Karolinska Institute, Danderyd University Hospital, 18288, Stockholm, Sweden
| | - Yvonne Power
- Department of Medical Sciences, Uppsala University, 75185, Uppsala, Sweden
| | - Marie Åsberg
- Department of Clinical Sciences, Karolinska Institute, Danderyd University Hospital, 18288, Stockholm, Sweden
| | - Kristian Borg
- Department of Clinical Sciences, Karolinska Institute, Danderyd University Hospital, 18288, Stockholm, Sweden
| | - Anna Nager
- Division of Family Medicine and Primary Health Care, Department of Neurobiology, Care Sciences and Society, Karolinska Institute, 17177, Stockholm, Sweden
| | - Charlotte Thålin
- Department of Clinical Sciences, Karolinska Institute, Danderyd University Hospital, 18288, Stockholm, Sweden
| | - Fariborz Mobarrez
- Department of Medical Sciences, Uppsala University, 75185, Uppsala, Sweden
| |
Collapse
|
15
|
Eustes AS, Ahmed A, Swamy J, Patil G, Jensen M, Wilson KM, Kudchadkar S, Wahab A, Perepu U, Miller FJ, Lentz SR, Dayal S. Extracellular histones: a unifying mechanism driving platelet-dependent extracellular vesicle release and thrombus formation in COVID-19. J Thromb Haemost 2024; 22:2514-2530. [PMID: 38815756 PMCID: PMC11343660 DOI: 10.1016/j.jtha.2024.05.019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2023] [Revised: 05/12/2024] [Accepted: 05/18/2024] [Indexed: 06/01/2024]
Abstract
BACKGROUND COVID-19 can cause profound inflammation and coagulopathy, and while many mechanisms have been proposed, there is no known common pathway leading to a prothrombotic state. OBJECTIVES From the beginning of the COVID-19 pandemic, elevated levels of extracellular histones have been found in plasma of patients infected with SARS-CoV-2. We hypothesized that platelet activation triggered by extracellular histones might represent a unifying mechanism leading to increased thrombin generation and thrombosis. METHODS We utilized blood samples collected from an early clinical trial of hospitalized COVID-19 patients (NCT04360824) and recruited healthy subjects as controls. Using plasma samples, we measured the procoagulant and prothrombotic potential of circulating extracellular histones and extracellular vesicles (EVs). Platelet prothrombotic activity was assessed via thrombin generation potential and platelet thrombus growth. Circulating EVs were assessed for thrombin generation potential in vitro in plasma and enhancement of thrombotic susceptibility in vivo in mice. RESULTS Compared with controls, COVID-19 patients had elevated plasma levels of citrullinated histone H3, cell-free DNA, nucleosomes, and EVs. Plasma from COVID-19 patients promoted platelet activation, platelet-dependent thrombin generation, thrombus growth under venous shear stress, and release of platelet-derived EVs. These prothrombotic effects of COVID-19 plasma were inhibited by an RNA aptamer that neutralizes both free and DNA-bound histones. EVs isolated from COVID-19 plasma enhanced thrombin generation in vitro and potentiated venous thrombosis in mice in vivo. CONCLUSION We conclude that extracellular histones and procoagulant EVs drive the prothrombotic state in COVID-19 and that histone-targeted therapy may prove beneficial.
Collapse
Affiliation(s)
- Alicia S Eustes
- Department of Internal Medicine, University of Iowa, Iowa City, Iowa, USA
| | - Azaj Ahmed
- Department of Internal Medicine, University of Iowa, Iowa City, Iowa, USA
| | - Jagadish Swamy
- Department of Internal Medicine, University of Iowa, Iowa City, Iowa, USA
| | - Gokul Patil
- Department of Internal Medicine, University of Iowa, Iowa City, Iowa, USA
| | - Melissa Jensen
- Department of Internal Medicine, University of Iowa, Iowa City, Iowa, USA
| | - Katina M Wilson
- Department of Internal Medicine, University of Iowa, Iowa City, Iowa, USA
| | - Shibani Kudchadkar
- Department of Internal Medicine, University of Iowa, Iowa City, Iowa, USA
| | - Abdul Wahab
- Department of Internal Medicine, University of Iowa, Iowa City, Iowa, USA
| | - Usha Perepu
- Department of Internal Medicine, University of Iowa, Iowa City, Iowa, USA
| | - Francis J Miller
- Department of Internal Medicine, Vanderbilt University Medical Center and VA Medical Center, Nashville, Tennessee, USA
| | - Steven R Lentz
- Department of Internal Medicine, University of Iowa, Iowa City, Iowa, USA
| | - Sanjana Dayal
- Department of Internal Medicine, University of Iowa, Iowa City, Iowa, USA; Iowa City VA Healthcare System, Iowa City, Iowa, USA.
| |
Collapse
|
16
|
Fanelli M, Petrone V, Chirico R, Radu CM, Minutolo A, Matteucci C. Flow cytometry for extracellular vesicle characterization in COVID-19 and post-acute sequelae of SARS-CoV-2 infection. EXTRACELLULAR VESICLES AND CIRCULATING NUCLEIC ACIDS 2024; 5:417-437. [PMID: 39697632 PMCID: PMC11648478 DOI: 10.20517/evcna.2024.20] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/15/2024] [Revised: 07/19/2024] [Accepted: 08/05/2024] [Indexed: 12/20/2024]
Abstract
Infection with SARS-CoV-2, the virus responsible for COVID-19 diseases, can impact different tissues and induce significant cellular alterations. The production of extracellular vesicles (EVs), which are physiologically involved in cell communication, is also altered during COVID-19, along with the dysfunction of cytoplasmic organelles. Since circulating EVs reflect the state of their cells of origin, they represent valuable tools for monitoring pathological conditions. Despite challenges in detecting EVs due to their size and specific cellular compartment origin using different methodologies, flow cytometry has proven to be an effective method for assessing the role of EVs in COVID-19. This review summarizes the involvement of plasmatic EVs in COVID-19 patients and individuals with Long COVID (LC) affected by post-acute sequelae of SARS-CoV-2 infection (PASC), highlighting their dual role in exerting both pro- and antiviral effects. We also emphasize how flow cytometry, with its multiparametric approach, can be employed to characterize circulating EVs, particularly in infectious diseases such as COVID-19, and suggest their potential role in chronic impairments during post-infection.
Collapse
Affiliation(s)
- Marialaura Fanelli
- Department of Experimental Medicine, University of Rome Tor Vergata, Rome 00133, Italy
| | - Vita Petrone
- Department of Experimental Medicine, University of Rome Tor Vergata, Rome 00133, Italy
| | - Rossella Chirico
- Department of Experimental Medicine, University of Rome Tor Vergata, Rome 00133, Italy
| | - Claudia Maria Radu
- Department of Medicine - DIMED, Thrombotic and Hemorrhagic Diseases Unit, University of Padua, Padua 35128 Italy
| | - Antonella Minutolo
- Department of Experimental Medicine, University of Rome Tor Vergata, Rome 00133, Italy
- Authors contributed equally
| | - Claudia Matteucci
- Department of Experimental Medicine, University of Rome Tor Vergata, Rome 00133, Italy
- Authors contributed equally
| |
Collapse
|
17
|
Rahmani A, Soleymani A, Almukhtar M, Behzad Moghadam K, Vaziri Z, Hosein Tabar Kashi A, Adabi Firoozjah R, Jafari Tadi M, Zolfaghari Dehkharghani M, Valadi H, Moghadamnia AA, Gasser RB, Rostami A. Exosomes, and the potential for exosome-based interventions against COVID-19. Rev Med Virol 2024; 34:e2562. [PMID: 38924213 DOI: 10.1002/rmv.2562] [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: 09/08/2023] [Revised: 05/17/2024] [Accepted: 06/08/2024] [Indexed: 06/28/2024]
Abstract
Since late 2019, the world has been devastated by the coronavirus disease 2019 (COVID-19) induced by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), with more than 760 million people affected and ∼seven million deaths reported. Although effective treatments for COVID-19 are currently limited, there has been a strong focus on developing new therapeutic approaches to address the morbidity and mortality linked to this disease. An approach that is currently being investigated is the use of exosome-based therapies. Exosomes are small, extracellular vesicles that play a role in many clinical diseases, including viral infections, infected cells release exosomes that can transmit viral components, such as miRNAs and proteins, and can also include receptors for viruses that facilitate viral entry into recipient cells. SARS-CoV-2 has the ability to impact the formation, secretion, and release of exosomes, thereby potentially facilitating or intensifying the transmission of the virus among cells, tissues and individuals. Therefore, designing synthetic exosomes that carry immunomodulatory cargo and antiviral compounds are proposed to be a promising strategy for the treatment of COVID-19 and other viral diseases. Moreover, exosomes generated from mesenchymal stem cells (MSC) might be employed as cell-free therapeutic agents, as MSC-derived exosomes can diminish the cytokine storm and reverse the suppression of host anti-viral defences associated with COVID-19, and boost the repair of lung damage linked to mitochondrial activity. The present article discusses the significance and roles of exosomes in COVID-19, and explores potential future applications of exosomes in combating this disease. Despite the challenges posed by COVID-19, exosome-based therapies could represent a promising avenue for improving patient outcomes and reducing the impact of this disease.
Collapse
Affiliation(s)
- Abolfazl Rahmani
- Infectious Diseases and Tropical Medicine Research Center, Health Research Institute, Babol University of Medical Sciences, Babol, Iran
| | - Ali Soleymani
- Infectious Diseases and Tropical Medicine Research Center, Health Research Institute, Babol University of Medical Sciences, Babol, Iran
| | | | - Kimia Behzad Moghadam
- Independent Researcher, Former University of California, San Francisco (UCSF), San Francisco, California, USA
| | - Zahra Vaziri
- Infectious Diseases and Tropical Medicine Research Center, Health Research Institute, Babol University of Medical Sciences, Babol, Iran
| | - Ali Hosein Tabar Kashi
- Infectious Diseases and Tropical Medicine Research Center, Health Research Institute, Babol University of Medical Sciences, Babol, Iran
| | - Reza Adabi Firoozjah
- Infectious Diseases and Tropical Medicine Research Center, Health Research Institute, Babol University of Medical Sciences, Babol, Iran
| | - Mehrdad Jafari Tadi
- Department of Cell and Molecular Medicine, Rush University Medical Center, Chicago, Illinois, USA
| | - Maryam Zolfaghari Dehkharghani
- Department of Healthcare Administration and Policy, School of Public Health, University of Nevada Las Vegas (UNLV), Las Vegas, Nevada, USA
| | - Hadi Valadi
- Department of Rheumatology and Inflammation Research Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Ali Akbar Moghadamnia
- Department of Pharmacology and Toxicology, Babol University of Medical Sciences, Babol, Iran
- Pharmaceutical Sciences Research Center, Health Research Institute, Babol University of Medical Sciences, Babol, Iran
| | - Robin B Gasser
- Department of Veterinary Biosciences, Faculty of Science, Melbourne Veterinary School, The University of Melbourne, Parkville, Victoria, Australia
| | - Ali Rostami
- Infectious Diseases and Tropical Medicine Research Center, Health Research Institute, Babol University of Medical Sciences, Babol, Iran
| |
Collapse
|
18
|
Ebrahimi R, Nasri F, Kalantari T. Coagulation and Inflammation in COVID-19: Reciprocal Relationship between Inflammatory and Coagulation Markers. Ann Hematol 2024; 103:1819-1831. [PMID: 38349409 DOI: 10.1007/s00277-024-05630-1] [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: 09/17/2023] [Accepted: 01/16/2024] [Indexed: 05/14/2024]
Abstract
The coronavirus disease 2019 (COVID-19) is caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), formerly known as 2019-nCoV. Numerous cellular and biochemical issues arise after COVID-19 infection. The severe inflammation that is caused by a number of cytokines appears to be one of the key hallmarks of COVID-19. Additionally, people with severe COVID-19 have coagulopathy and fulminant thrombotic events. We briefly reviewed the COVID-19 disease at the beginning of this paper. The inflammation and coagulation markers and their alterations in COVID-19 illness are briefly discussed in the parts that follow. Next, we talked about NETosis, which is a crucial relationship between coagulation and inflammation. In the end, we mentioned the two-way relationship between inflammation and coagulation, as well as the factors involved in it. We suggest that inflammation and coagulation are integrated systems in COVID-19 that act on each other in such a way that not only inflammation can activate coagulation but also coagulation can activate inflammation.
Collapse
Affiliation(s)
- Rasoul Ebrahimi
- Division of Laboratory Hematology and Blood Banking, Department of Medical Laboratory Sciences, School of Paramedical Sciences, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Fatemeh Nasri
- Division of Laboratory Hematology and Blood Banking, Department of Medical Laboratory Sciences, School of Paramedical Sciences, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Tahereh Kalantari
- Division of Laboratory Hematology and Blood Banking, Department of Medical Laboratory Sciences, School of Paramedical Sciences, Shiraz University of Medical Sciences, Shiraz, Iran.
| |
Collapse
|
19
|
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
|
20
|
Barbosa MS, de Lima F, Peachazepi Moraes CR, Borba-Junior IT, Huber SC, Santos I, Bombassaro B, Dertkigil SSJ, Ilich A, Key NS, Annichino-Bizzacchi JM, Orsi FA, Mansour E, Velloso LA, De Paula EV. Angiopoietin2 is associated with coagulation activation and tissue factor expression in extracellular vesicles in COVID-19. Front Med (Lausanne) 2024; 11:1367544. [PMID: 38803346 PMCID: PMC11128612 DOI: 10.3389/fmed.2024.1367544] [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: 01/08/2024] [Accepted: 04/12/2024] [Indexed: 05/29/2024] Open
Abstract
Coagulation activation in immunothrombosis involves various pathways distinct from classical hemostasis, offering potential therapeutic targets to control inflammation-induced hypercoagulability while potentially sparing hemostasis. The Angiopoietin/Tie2 pathway, previously linked to embryonic angiogenesis and sepsis-related endothelial barrier regulation, was recently associated with coagulation activation in sepsis and COVID-19. This study explores the connection between key mediators of the Angiopoietin/Tie2 pathway and coagulation activation. The study included COVID-19 patients with hypoxia and healthy controls. Blood samples were processed to obtain platelet-free plasma, and frozen until analysis. Extracellular vesicles (EVs) in plasma were characterized and quantified using flow cytometry, and their tissue factor (TF) procoagulant activity was measured using a kinetic chromogenic method. Several markers of hemostasis were assessed. Levels of ANGPT1, ANGPT2, and soluble Tie2 correlated with markers of coagulation and platelet activation. EVs from platelets and endothelial cells were increased in COVID-19 patients, and a significant increase in TF+ EVs derived from endothelial cells was observed. In addition, ANGPT2 levels were associated with TF expression and activity in EVs. In conclusion, we provide further evidence for the involvement of the Angiopoietin/Tie2 pathway in the coagulopathy of COVID-19 mediated in part by release of EVs as a potential source of TF activity.
Collapse
Affiliation(s)
- Mayck Silva Barbosa
- School of Medical Sciences, Universidade Estadual de Campinas, Campinas, Brazil
| | - Franciele de Lima
- School of Medical Sciences, Universidade Estadual de Campinas, Campinas, Brazil
| | | | | | - Stephany Cares Huber
- Hematology and Hemotherapy Center, Universidade Estadual de Campinas, Campinas, Brazil
| | - Irene Santos
- Hematology and Hemotherapy Center, Universidade Estadual de Campinas, Campinas, Brazil
| | - Bruna Bombassaro
- Obesity and Comorbidities Research Center, Universidade Estadual de Campinas, Campinas, Brazil
| | | | - Anton Ilich
- Blood Research Center, University of North Carolina, Chapel Hill, NC, United States
- Division of Hematology, Department of Medicine, University of North Carolina, Chapel Hill, NC, United States
| | - Nigel S. Key
- Blood Research Center, University of North Carolina, Chapel Hill, NC, United States
- Division of Hematology, Department of Medicine, University of North Carolina, Chapel Hill, NC, United States
- Department of Pathology and Laboratory Medicine, University of North Carolina, Chapel Hill, NC, United States
| | - Joyce M. Annichino-Bizzacchi
- School of Medical Sciences, Universidade Estadual de Campinas, Campinas, Brazil
- Hematology and Hemotherapy Center, Universidade Estadual de Campinas, Campinas, Brazil
| | - Fernanda Andrade Orsi
- Hematology and Hemotherapy Center, Universidade Estadual de Campinas, Campinas, Brazil
| | - Eli Mansour
- School of Medical Sciences, Universidade Estadual de Campinas, Campinas, Brazil
| | - Licio A. Velloso
- School of Medical Sciences, Universidade Estadual de Campinas, Campinas, Brazil
- Obesity and Comorbidities Research Center, Universidade Estadual de Campinas, Campinas, Brazil
| | - Erich Vinicius De Paula
- School of Medical Sciences, Universidade Estadual de Campinas, Campinas, Brazil
- Hematology and Hemotherapy Center, Universidade Estadual de Campinas, Campinas, Brazil
| |
Collapse
|
21
|
Goonewardena SN, Chen Q, Tate AM, Grushko OG, Damodaran D, Blakely P, Hayek SS, Pinsky DJ, Rosenson RS. Monocyte-Mediated Thrombosis Linked to Circulating Tissue Factor and Immune Paralysis in COVID-19. Arterioscler Thromb Vasc Biol 2024; 44:1124-1134. [PMID: 38511328 PMCID: PMC11043007 DOI: 10.1161/atvbaha.122.318721] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2022] [Accepted: 02/29/2024] [Indexed: 03/22/2024]
Abstract
BACKGROUND SARS-CoV-2 infections cause COVID-19 and are associated with inflammation, coagulopathy, and high incidence of thrombosis. Myeloid cells help coordinate the initial immune response in COVID-19. Although we appreciate that myeloid cells lie at the nexus of inflammation and thrombosis, the mechanisms that unite the two in COVID-19 remain largely unknown. METHODS In this study, we used systems biology approaches including proteomics, transcriptomics, and mass cytometry to define the circulating proteome and circulating immune cell phenotypes in subjects with COVID-19. RESULTS In a cohort of subjects with COVID-19 (n=35), circulating markers of inflammation (CCL23 [C-C motif chemokine ligand 23] and IL [interleukin]-6) and vascular dysfunction (ACE2 [angiotensin-converting enzyme 2] and TF [tissue factor]) were elevated in subjects with severe compared with mild COVID-19. Additionally, although the total white blood cell counts were similar between COVID-19 groups, CD14+ (cluster of differentiation) monocytes from subjects with severe COVID-19 expressed more TF. At baseline, transcriptomics demonstrated increased IL-6, CCL3, ACOD1 (aconitate decarboxylase 1), C5AR1 (complement component 5a receptor), C5AR2, and TF in subjects with severe COVID-19 compared with controls. Using stress transcriptomics, we found that circulating immune cells from subjects with severe COVID-19 had evidence of profound immune paralysis with greatly reduced transcriptional activation and release of inflammatory markers in response to TLR (Toll-like receptor) activation. Finally, sera from subjects with severe (but not mild) COVID-19 activated human monocytes and induced TF expression. CONCLUSIONS Taken together, these observations further elucidate the pathological mechanisms that underlie immune dysfunction and coagulation abnormalities in COVID-19, contributing to our growing understanding of SARS-CoV-2 infections that could also be leveraged to develop novel diagnostic and therapeutic strategies.
Collapse
Affiliation(s)
- Sascha N. Goonewardena
- Division of Cardiovascular Medicine, Department of Internal Medicine, University of MichiganAnn Arbor, Michigan, USA
| | - Qinzhong Chen
- Metabolism and Lipids Unit, Cardiovascular Institute, Marie-Josee and Henry R Kravis Center for Cardiovascular Health, Icahn School of Medicine at Mount Sinai, New York, USA
| | - Ashley M. Tate
- Division of Cardiovascular Medicine, Department of Internal Medicine, University of MichiganAnn Arbor, Michigan, USA
| | - Olga G Grushko
- Division of Cardiovascular Medicine, Department of Internal Medicine, University of MichiganAnn Arbor, Michigan, USA
| | - Dilna Damodaran
- Division of Cardiovascular Medicine, Department of Internal Medicine, University of MichiganAnn Arbor, Michigan, USA
| | - Pennelope Blakely
- Division of Cardiovascular Medicine, Department of Internal Medicine, University of MichiganAnn Arbor, Michigan, USA
| | - Salim S. Hayek
- Division of Cardiovascular Medicine, Department of Internal Medicine, University of MichiganAnn Arbor, Michigan, USA
| | - David J. Pinsky
- Division of Cardiovascular Medicine, Department of Internal Medicine, University of MichiganAnn Arbor, Michigan, USA
| | - Robert S. Rosenson
- Metabolism and Lipids Unit, Cardiovascular Institute, Marie-Josee and Henry R Kravis Center for Cardiovascular Health, Icahn School of Medicine at Mount Sinai, New York, USA
| |
Collapse
|
22
|
Hillung J, Olmo-Uceda MJ, Muñoz-Sánchez JC, Elena SF. Accumulation Dynamics of Defective Genomes during Experimental Evolution of Two Betacoronaviruses. Viruses 2024; 16:644. [PMID: 38675984 PMCID: PMC11053736 DOI: 10.3390/v16040644] [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: 03/21/2024] [Revised: 04/18/2024] [Accepted: 04/19/2024] [Indexed: 04/28/2024] Open
Abstract
Virus-encoded replicases often generate aberrant RNA genomes, known as defective viral genomes (DVGs). When co-infected with a helper virus providing necessary proteins, DVGs can multiply and spread. While DVGs depend on the helper virus for propagation, they can in some cases disrupt infectious virus replication, impact immune responses, and affect viral persistence or evolution. Understanding the dynamics of DVGs alongside standard viral genomes during infection remains unclear. To address this, we conducted a long-term experimental evolution of two betacoronaviruses, the human coronavirus OC43 (HCoV-OC43) and the murine hepatitis virus (MHV), in cell culture at both high and low multiplicities of infection (MOI). We then performed RNA-seq at regular time intervals, reconstructed DVGs, and analyzed their accumulation dynamics. Our findings indicate that DVGs evolved to exhibit greater diversity and abundance, with deletions and insertions being the most common types. Notably, some high MOI deletions showed very limited temporary existence, while others became prevalent over time. We observed differences in DVG abundance between high and low MOI conditions in HCoV-OC43 samples. The size distribution of HCoV-OC43 genomes with deletions differed between high and low MOI passages. In low MOI lineages, short and long DVGs were the most common, with an additional cluster in high MOI lineages which became more prevalent along evolutionary time. MHV also showed variations in DVG size distribution at different MOI conditions, though they were less pronounced compared to HCoV-OC43, suggesting a more random distribution of DVG sizes. We identified hotspot regions for deletions that evolved at a high MOI, primarily within cistrons encoding structural and accessory proteins. In conclusion, our study illustrates the widespread formation of DVGs during betacoronavirus evolution, influenced by MOI and cell- and virus-specific factors.
Collapse
Affiliation(s)
- Julia Hillung
- Instituto de Biología Integrativa de Sistemas (I2SysBio), CSIC-UV, Catedrático Agustín Escardino Benlloch 9, 46980 Paterna, Valencia, Spain; (J.H.); (M.J.O.-U.); (J.C.M.-S.)
| | - María J. Olmo-Uceda
- Instituto de Biología Integrativa de Sistemas (I2SysBio), CSIC-UV, Catedrático Agustín Escardino Benlloch 9, 46980 Paterna, Valencia, Spain; (J.H.); (M.J.O.-U.); (J.C.M.-S.)
| | - Juan C. Muñoz-Sánchez
- Instituto de Biología Integrativa de Sistemas (I2SysBio), CSIC-UV, Catedrático Agustín Escardino Benlloch 9, 46980 Paterna, Valencia, Spain; (J.H.); (M.J.O.-U.); (J.C.M.-S.)
| | - Santiago F. Elena
- Instituto de Biología Integrativa de Sistemas (I2SysBio), CSIC-UV, Catedrático Agustín Escardino Benlloch 9, 46980 Paterna, Valencia, Spain; (J.H.); (M.J.O.-U.); (J.C.M.-S.)
- Santa Fe Institute, 1399 Hyde Park Road, Santa Fe, NM 87501, USA
| |
Collapse
|
23
|
Barion BG, Rocha TRFD, Ho YL, Mazetto Fonseca BDM, Okazaki E, Rothschild C, Stefanello B, Rocha VG, Villaça PR, Orsi FA. Extracellular vesicles are a late marker of inflammation, hypercoagulability and COVID-19 severity. Hematol Transfus Cell Ther 2024; 46:176-185. [PMID: 38341321 DOI: 10.1016/j.htct.2023.12.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2023] [Revised: 09/27/2023] [Accepted: 12/08/2023] [Indexed: 02/12/2024] Open
Abstract
Exacerbated inflammation and coagulation are a hallmark of COVID-19 severity. Extracellular vesicles (EVs) are intercellular transmitters involved in inflammatory conditions, which are capable of triggering prothrombotic mechanisms. Since the release of EVs is potentially associated with COVID-19-induced coagulopathy, the aim of this study was to evaluate changes in inflammation- and hypercoagulability-related EVs during the first month after symptom onset and to determine whether they are associated with disease severity. Blood samples of patients with mild or severe forms of the disease were collected on three occasions: in the second, third and fourth weeks after symptom onset for the quantification by flow cytometry of CD41A (platelet glycoprotein IIb/IIIa), CD162 (PSGL-1), CD31 (PECAM-1) and CD142 cells (tissue factor). Analysis of variance (ANOVA) with repeated measures, Kruskal-Wallis and correlation tests were used. Eighty-five patients were enrolled, 71% of whom had mild disease. Seventeen uninfected individuals served as controls. Compared to controls, both mild and severe COVID-19 were associated with higher EV-CD31+, EV-CD41+ and EV-CD142+ levels. All EV levels were higher in severe than in mild COVID-19 only after the third week from symptom onset, as opposed to C-reactive protein and D-dimer levels, which were higher in severe than in mild COVID-19 earlier during disease progression. EV levels were also associated with C-reactive protein and D-dimer levels only after the third week of symptoms. In conclusion, EVs expressing CD41A, CD31, TF, and CD162 appear as late markers of COVID-19 severity. This finding may contribute to the understanding of the pathogenesis of acute and possibly long COVID-19.
Collapse
Affiliation(s)
| | | | - Yeh-Li Ho
- Hospital das Clínicas da Faculdade de Medicina da Universidade de São (HCFMUSP), Sao Paulo, Brazil
| | | | - Erica Okazaki
- Hospital das Clínicas da Faculdade de Medicina da Universidade de São (HCFMUSP), Sao Paulo, Brazil
| | - Cynthia Rothschild
- Hospital das Clínicas da Faculdade de Medicina da Universidade de São (HCFMUSP), Sao Paulo, Brazil
| | - Bianca Stefanello
- Hospital das Clínicas da Faculdade de Medicina da Universidade de São (HCFMUSP), Sao Paulo, Brazil
| | - Vanderson Geraldo Rocha
- Hospital das Clínicas da Faculdade de Medicina da Universidade de São (HCFMUSP), Sao Paulo, Brazil
| | - Paula Ribeiro Villaça
- Hospital das Clínicas da Faculdade de Medicina da Universidade de São (HCFMUSP), Sao Paulo, Brazil
| | - Fernanda A Orsi
- Hospital das Clínicas da Faculdade de Medicina da Universidade de São (HCFMUSP), Sao Paulo, Brazil; Department of Pathology, School of Medical Sciences, Universidade de Campinas (UNICAMP), Campinas, Brazil.
| |
Collapse
|
24
|
Müller-Calleja N, Grunz K, Nguyen TS, Posma J, Pedrosa D, Meineck M, Hollerbach A, Braun J, Muth S, Schild H, Saar K, Hübner N, Krishnaswamy S, Royce J, Teyton L, Lemmermann N, Weinmann-Menke J, Lackner KJ, Ruf W. Targeting the tissue factor coagulation initiation complex prevents antiphospholipid antibody development. Blood 2024; 143:1167-1180. [PMID: 38142429 PMCID: PMC10972716 DOI: 10.1182/blood.2023022276] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2023] [Revised: 12/08/2023] [Accepted: 12/13/2023] [Indexed: 12/26/2023] Open
Abstract
ABSTRACT Antiphospholipid antibodies (aPL) in primary or secondary antiphospholipid syndrome (APS) are a major cause for acquired thrombophilia, but specific interventions preventing autoimmune aPL development are an unmet clinical need. Although autoimmune aPL cross react with various coagulation regulatory proteins, lipid-reactive aPL, including those derived from patients with COVID-19, recognize the endolysosomal phospholipid lysobisphosphatidic acid presented by the cell surface-expressed endothelial protein C receptor. This specific recognition leads to complement-mediated activation of tissue factor (TF)-dependent proinflammatory signaling and thrombosis. Here, we show that specific inhibition of the TF coagulation initiation complex with nematode anticoagulant protein c2 (NAPc2) prevents the prothrombotic effects of aPL derived from patients with COVID-19 in mice and the aPL-induced proinflammatory and prothrombotic activation of monocytes. The induction of experimental APS is dependent on the nicotinamide adenine dinucleotide phosphate (NADPH) oxidase complex, and NAPc2 suppresses monocyte endosomal reactive oxygen species production requiring the TF cytoplasmic domain and interferon-α secretion from dendritic cells. Latent infection with murine cytomegalovirus causes TF cytoplasmic domain-dependent development of persistent aPL and circulating phospholipid-reactive B1 cells, which is prevented by short-term intervention with NAPc2 during acute viral infection. In addition, treatment of lupus prone MRL-lpr mice with NAPc2, but not with heparin, suppresses dendritic-cell activation in the spleen, aPL production and circulating phospholipid-reactive B1 cells, and attenuates lupus pathology. These data demonstrate a convergent TF-dependent mechanism of aPL development in latent viral infection and autoimmune disease and provide initial evidence that specific targeting of the TF initiation complex has therapeutic benefits beyond currently used clinical anticoagulant strategies.
Collapse
Affiliation(s)
- Nadine Müller-Calleja
- Center for Thrombosis and Hemostasis, Johannes Gutenberg University Medical Center, Mainz, Germany
- Institute of Clinical Chemistry and Laboratory Medicine, Johannes Gutenberg University Medical Center, Mainz, Germany
| | - Kristin Grunz
- Center for Thrombosis and Hemostasis, Johannes Gutenberg University Medical Center, Mainz, Germany
| | - T. Son Nguyen
- Center for Thrombosis and Hemostasis, Johannes Gutenberg University Medical Center, Mainz, Germany
| | - Jens Posma
- Center for Thrombosis and Hemostasis, Johannes Gutenberg University Medical Center, Mainz, Germany
| | - Denise Pedrosa
- Center for Thrombosis and Hemostasis, Johannes Gutenberg University Medical Center, Mainz, Germany
| | - Myriam Meineck
- Department of Medicine I, Johannes Gutenberg University Medical Center, Mainz, Germany
| | - Anne Hollerbach
- Institute of Clinical Chemistry and Laboratory Medicine, Johannes Gutenberg University Medical Center, Mainz, Germany
| | - Johannes Braun
- Center for Thrombosis and Hemostasis, Johannes Gutenberg University Medical Center, Mainz, Germany
| | - Sabine Muth
- Institute for Immunology, Johannes Gutenberg University Medical Center, Mainz, Germany
| | - Hansjörg Schild
- Institute for Immunology, Johannes Gutenberg University Medical Center, Mainz, Germany
| | - Kathrin Saar
- Max Delbrück Center for Molecular Medicine, Berlin, Germany
- Charite-Universitätsmedizin Berlin, Berlin, Germany
- German Center for Cardiovascular Research (DZHK), Partner site Berlin, Berlin, Germany
| | - Norbert Hübner
- Max Delbrück Center for Molecular Medicine, Berlin, Germany
- Charite-Universitätsmedizin Berlin, Berlin, Germany
- German Center for Cardiovascular Research (DZHK), Partner site Berlin, Berlin, Germany
| | - Sriram Krishnaswamy
- Department of Pediatrics, Children’s Hospital of Philadelphia, University of Pennsylvania, Philadelphia, PA
| | - Jennifer Royce
- Department of Immunology and Microbiology, Scripps Research, La Jolla, CA
| | - Luc Teyton
- Department of Immunology and Microbiology, Scripps Research, La Jolla, CA
| | - Niels Lemmermann
- Institute for Virology, Johannes Gutenberg University Medical Center, Mainz, Germany
- Institute of Virology, University Hospital Bonn, Bonn, Germany
| | - Julia Weinmann-Menke
- Department of Medicine I, Johannes Gutenberg University Medical Center, Mainz, Germany
| | - Karl J. Lackner
- Institute of Clinical Chemistry and Laboratory Medicine, Johannes Gutenberg University Medical Center, Mainz, Germany
| | - Wolfram Ruf
- Center for Thrombosis and Hemostasis, Johannes Gutenberg University Medical Center, Mainz, Germany
- Department of Immunology and Microbiology, Scripps Research, La Jolla, CA
- German Center for Cardiovascular Research (DZHK), Partner site Rhein-Main, Mainz, Germany
| |
Collapse
|
25
|
Mackman N. Tissue Factor and COVID-19 Associated Thrombosis. Arterioscler Thromb Vasc Biol 2024; 44:523-529. [PMID: 38381854 PMCID: PMC10883617 DOI: 10.1161/atvbaha.123.320144] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2023] [Accepted: 01/17/2024] [Indexed: 02/23/2024]
Abstract
Microbial infections activate the innate and adaptive immune systems.1 Pathogen-associated molecular patterns produced by microbes, such as double-stranded RNA, are detected by PRRs (pattern-recognition receptors), such as toll-like receptor 3, and this leads to the expression of interferons and cytokines.1,2.
Collapse
Affiliation(s)
- Nigel Mackman
- Department of Medicine, UNC Blood Research Center, University of North Carolina at Chapel Hill
| |
Collapse
|
26
|
Chang H, Chen E, Hu Y, Wu L, Deng L, Ye‐Lehmann S, Mao X, Zhu T, Liu J, Chen C. Extracellular Vesicles: The Invisible Heroes and Villains of COVID-19 Central Neuropathology. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2305554. [PMID: 38143270 PMCID: PMC10933635 DOI: 10.1002/advs.202305554] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/09/2023] [Revised: 11/18/2023] [Indexed: 12/26/2023]
Abstract
Acknowledging the neurological symptoms of COVID-19 and the long-lasting neurological damage even after the epidemic ends are common, necessitating ongoing vigilance. Initial investigations suggest that extracellular vesicles (EVs), which assist in the evasion of the host's immune response and achieve immune evasion in SARS-CoV-2 systemic spreading, contribute to the virus's attack on the central nervous system (CNS). The pro-inflammatory, pro-coagulant, and immunomodulatory properties of EVs contents may directly drive neuroinflammation and cerebral thrombosis in COVID-19. Additionally, EVs have attracted attention as potential candidates for targeted therapy in COVID-19 due to their innate homing properties, low immunogenicity, and ability to cross the blood-brain barrier (BBB) freely. Mesenchymal stromal/stem cell (MSCs) secreted EVs are widely applied and evaluated in patients with COVID-19 for their therapeutic effect, considering the limited antiviral treatment. This review summarizes the involvement of EVs in COVID-19 neuropathology as carriers of SARS-CoV-2 or other pathogenic contents, as predictors of COVID-19 neuropathology by transporting brain-derived substances, and as therapeutic agents by delivering biotherapeutic substances or drugs. Understanding the diverse roles of EVs in the neuropathological aspects of COVID-19 provides a comprehensive framework for developing, treating, and preventing central neuropathology and the severe consequences associated with the disease.
Collapse
Affiliation(s)
- Haiqing Chang
- Department of AnesthesiologyWest China HospitalSichuan UniversityLaboratory of Anesthesia and Critical Care MedicineNational‐Local Joint Engineering Research Centre of Translational Medicine of AnesthesiologyWest China HospitalSichuan UniversityChengduSichuan610041China
| | - Erya Chen
- Department of AnesthesiologyWest China HospitalSichuan UniversityLaboratory of Anesthesia and Critical Care MedicineNational‐Local Joint Engineering Research Centre of Translational Medicine of AnesthesiologyWest China HospitalSichuan UniversityChengduSichuan610041China
| | - Yi Hu
- Department of Cardiology, Honghui hospitalXi'an Jiaotong UniversityXi'an710049China
| | - Lining Wu
- Department of AnesthesiologyWest China HospitalSichuan UniversityLaboratory of Anesthesia and Critical Care MedicineNational‐Local Joint Engineering Research Centre of Translational Medicine of AnesthesiologyWest China HospitalSichuan UniversityChengduSichuan610041China
| | - Liyun Deng
- Department of AnesthesiologyWest China HospitalSichuan UniversityLaboratory of Anesthesia and Critical Care MedicineNational‐Local Joint Engineering Research Centre of Translational Medicine of AnesthesiologyWest China HospitalSichuan UniversityChengduSichuan610041China
| | - Shixin Ye‐Lehmann
- Diseases and Hormones of the Nervous System University of Paris‐Scalay Bicêtre Hosptial BâtGrégory Pincus 80 Rue du Gal Leclerc, CedexLe Kremlin Bicêtre94276France
| | - Xiaobo Mao
- Department of NeurologyInstitute of Cell EngineeringSchool of MedicineJohns Hopkins UniversityBaltimoreMD21218USA
| | - Tao Zhu
- Department of AnesthesiologyWest China HospitalSichuan UniversityLaboratory of Anesthesia and Critical Care MedicineNational‐Local Joint Engineering Research Centre of Translational Medicine of AnesthesiologyWest China HospitalSichuan UniversityChengduSichuan610041China
| | - Jin Liu
- Department of AnesthesiologyWest China HospitalSichuan UniversityLaboratory of Anesthesia and Critical Care MedicineNational‐Local Joint Engineering Research Centre of Translational Medicine of AnesthesiologyWest China HospitalSichuan UniversityChengduSichuan610041China
| | - Chan Chen
- Department of AnesthesiologyWest China HospitalSichuan UniversityLaboratory of Anesthesia and Critical Care MedicineNational‐Local Joint Engineering Research Centre of Translational Medicine of AnesthesiologyWest China HospitalSichuan UniversityChengduSichuan610041China
| |
Collapse
|
27
|
Hillung J, Lázaro JT, Muñoz-Sánchez JC, Olmo-Uceda MJ, Sardanyés J, Elena SF. Decay of HCoV-OC43 infectivity is lower in cell debris-containing media than in fresh culture media. MICROPUBLICATION BIOLOGY 2024; 2024:10.17912/micropub.biology.001092. [PMID: 38440329 PMCID: PMC10910279 DOI: 10.17912/micropub.biology.001092] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Figures] [Subscribe] [Scholar Register] [Received: 12/15/2023] [Revised: 02/13/2024] [Accepted: 02/15/2024] [Indexed: 03/06/2024]
Abstract
In the quantitative description of viral dynamics within cell cultures and, more broadly, in modeling within-host viral infections, a question that commonly arises is whether the degradation of a fraction of the virus could be disregarded in comparison with the massive synthesis of new viral particles. Surprisingly, quantitative data on the synthesis and degradation rates of RNA viruses in cell cultures are scarce. In this study, we investigated the decay of the human betacoronavirus OC43 (HCoV-OC43) infectivity in cell culture lysates and in fresh media. Our findings revealed a significantly slower viral decay rate in the medium containing lysate cells compared to the fresh medium. This observation suggests that the presence of cellular debris from lysed cells may offer protection or stabilize virions, slowing down their degradation. Moreover, the growth rate of HCoV-OC43 infectivity is significantly higher than degradation as long as there are productive cells in the medium, suggesting that, as a first approximation, degradation can be neglected during early infection.
Collapse
Affiliation(s)
- Julia Hillung
- Evolutionary Systems Virology, Instituto de Biología Integrativa de Sistemas (I2SysBio), CSIC - Universitat de València, Paterna, 46980 València, Spain
| | - J. Tomás Lázaro
- Dynamical Systems and Computational Virology, CSIC Associated Unit CRM - I2SysBio, Spain
- Departament de Matemàtiques, Universitat Politècnica de Catalunya (UPC), 08028 Barcelona, Spain
- Institute of Mathematics, UPC - BarcelonaTech (IMTech), 08028 Barcelona, Spain
- Centre de Recerca Matemàtica (CRM), Campus de Bellaterra, Cerdanyola del Vallès, 08193 Barcelona, Spain
| | - Juan-Carlos Muñoz-Sánchez
- Evolutionary Systems Virology, Instituto de Biología Integrativa de Sistemas (I2SysBio), CSIC - Universitat de València, Paterna, 46980 València, Spain
| | - María-José Olmo-Uceda
- Evolutionary Systems Virology, Instituto de Biología Integrativa de Sistemas (I2SysBio), CSIC - Universitat de València, Paterna, 46980 València, Spain
| | - Josep Sardanyés
- Centre de Recerca Matemàtica (CRM), Campus de Bellaterra, Cerdanyola del Vallès, 08193 Barcelona, Spain
- Dynamical Systems and Computational Virology, CSIC Associated Unit CRM - I2SysBio, Spain
| | - Santiago F. Elena
- Evolutionary Systems Virology, Instituto de Biología Integrativa de Sistemas (I2SysBio), CSIC - Universitat de València, Paterna, 46980 València, Spain
- Santa Fe Institute, Santa Fe, New Mexico, United States
| |
Collapse
|
28
|
Papareddy P, Tapken I, Kroh K, Varma Bhongir RK, Rahman M, Baumgarten M, Cim EI, Györffy L, Smeds E, Neumann A, Veerla S, Olinder J, Thorlacus H, Ryden C, Bartakova E, Holub M, Herwald H. The role of extracellular vesicle fusion with target cells in triggering systemic inflammation. Nat Commun 2024; 15:1150. [PMID: 38326335 PMCID: PMC10850166 DOI: 10.1038/s41467-024-45125-1] [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: 07/17/2023] [Accepted: 01/16/2024] [Indexed: 02/09/2024] Open
Abstract
Extracellular vesicles (EVs) play a crucial role in intercellular communication by transferring bioactive molecules from donor to recipient cells. As a result, EV fusion leads to the modulation of cellular functions and has an impact on both physiological and pathological processes in the recipient cell. This study explores the impact of EV fusion on cellular responses to inflammatory signaling. Our findings reveal that fusion renders non-responsive cells susceptible to inflammatory signaling, as evidenced by increased NF-κB activation and the release of inflammatory mediators. Syntaxin-binding protein 1 is essential for the merge and activation of intracellular signaling. Subsequent analysis show that EVs transfer their functionally active receptors to target cells, making them prone to an otherwise unresponsive state. EVs in complex with their agonist, require no further stimulation of the target cells to trigger mobilization of NF-κB. While receptor antagonists were unable to inhibit NF-κB activation, blocking of the fusion between EVs and their target cells with heparin mitigated inflammation in mice challenged with EVs.
Collapse
Affiliation(s)
- Praveen Papareddy
- Division of Infection Medicine, Department of Clinical Sciences, Lund University, Lund, Sweden.
| | - Ines Tapken
- Division of Infection Medicine, Department of Clinical Sciences, Lund University, Lund, Sweden
- SMATHERIA gGmbH - Non-Profit Biomedical Research Institute, Hannover, Germany
- Center for Systems Neuroscience (ZSN), Hannover, Germany
| | - Keshia Kroh
- Division of Infection Medicine, Department of Clinical Sciences, Lund University, Lund, Sweden
- Department of Viroscience, Erasmus Medical Center, Rotterdam, the Netherlands
| | | | - Milladur Rahman
- Section of Surgery, Department of Clinical Sciences, Lund University, Malmö, Sweden
| | - Maria Baumgarten
- Division of Infection Medicine, Department of Clinical Sciences, Lund University, Lund, Sweden
| | - Eda Irem Cim
- Division of Infection Medicine, Department of Clinical Sciences, Lund University, Lund, Sweden
| | - Lilla Györffy
- Division of Infection Medicine, Department of Clinical Sciences, Lund University, Lund, Sweden
| | - Emanuel Smeds
- Division of Infection Medicine, Department of Clinical Sciences, Lund University, Lund, Sweden
| | - Ariane Neumann
- Division of Infection Medicine, Department of Clinical Sciences, Lund University, Lund, Sweden
| | - Srinivas Veerla
- Division of Oncology and Pathology, Lund, Department of Clinical Sciences, Lund University, Lund, Sweden
| | - Jon Olinder
- Division of Infection Medicine, Helsingborg Hospital and Department of Clinical Sciences Helsingborg, Lund University, Lund, Sweden
| | - Henrik Thorlacus
- Section of Surgery, Department of Clinical Sciences, Lund University, Malmö, Sweden
| | - Cecilia Ryden
- Division of Infection Medicine, Helsingborg Hospital and Department of Clinical Sciences Helsingborg, Lund University, Lund, Sweden
| | - Eva Bartakova
- Department of Infectious Diseases, First Faculty of Medicine, Charles University and Military University Hospital Prague, Praha, Czech Republic
| | - Michal Holub
- Department of Infectious Diseases, First Faculty of Medicine, Charles University and Military University Hospital Prague, Praha, Czech Republic
| | - Heiko Herwald
- Division of Infection Medicine, Department of Clinical Sciences, Lund University, Lund, Sweden.
| |
Collapse
|
29
|
Raadsen M, Langerak T, Du Toit J, Kruip MJHA, Aynekulu Mersha D, De Maat MPM, Vermin B, Van den Akker JPC, Schmitz KS, Bakhtiari K, Meijers JCM, van Gorp ECM, Short KR, Haagmans B, de Vries RD, Gommers DAMPJ, Endeman H, Goeijenbier M. Presence of procoagulant peripheral blood mononuclear cells in severe COVID-19 patients relate to ventilation perfusion mismatch and precede pulmonary embolism. J Crit Care 2024; 79:154463. [PMID: 37976997 DOI: 10.1016/j.jcrc.2023.154463] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Revised: 10/07/2023] [Accepted: 10/31/2023] [Indexed: 11/19/2023]
Abstract
PURPOSE Pulmonary emboli (PE) contribute substantially to coronavirus disease 2019 (COVID-19) related mortality and morbidity. Immune cell-mediated hyperinflammation drives the procoagulant state in COVID-19 patients, resulting in immunothrombosis. To study the role of peripheral blood mononuclear cells (PBMC) in the procoagulant state of COVID-19 patients, we performed a functional bioassay and related outcomes to the occurrence of PE. Secondary aims were to relate this functional assay to plasma D-dimer levels, ventilation perfusion mismatch and TF expression on monocyte subsets. METHODS PBMC from an ICU biobank were obtained from 20 patients with a computed tomography angiograph (CTA) proven PE and compared to 15 COVID-19 controls without a proven PE. Functional procoagulant properties of PBMC were measured using a modified fibrin generation time (MC-FGT) assay. Tissue factor (TF) expression on monocyte subsets were measured by flow cytometry. Additional clinical data were obtained from patient records including end-tidal to arterial carbon dioxide gradient. RESULTS MC-FGT levels were highest in the samples taken closest to the PE detection, similar to the end-tidal to arterial carbon dioxide gradient (ETCO2 - PaCO2), a measurement to quantify ventilation-perfusion mismatch. In patients without proven PE, peak MC-FGT relates to an increase in end-tidal to arterial carbon dioxide gradient. We identified non-classical, CD16 positive monocytes as the subset with increased TF expression. CONCLUSION We show that the procoagulant state of PBMC could aid in early detection of PE in COVID-19 ICU patients. Combined with end-tidal to ETCO2 - PaCO2 gradient, these tests could improve early detection of PE on the ICU.
Collapse
Affiliation(s)
- M Raadsen
- Department of Viroscience, Erasmus Medical Center, Rotterdam, the Netherlands
| | - T Langerak
- Department of Viroscience, Erasmus Medical Center, Rotterdam, the Netherlands
| | - J Du Toit
- Department of Viroscience, Erasmus Medical Center, Rotterdam, the Netherlands; Department of Hematology, Wits Donal Gordon Medical Center, Johannesburg, South Africa
| | - M J H A Kruip
- Department of Hematology, Erasmus Medical Center, Rotterdam, the Netherlands
| | - D Aynekulu Mersha
- Department of Viroscience, Erasmus Medical Center, Rotterdam, the Netherlands; Department of Intensive Care, Erasmus MC, Rotterdam, the Netherlands
| | - M P M De Maat
- Department of Hematology, Erasmus Medical Center, Rotterdam, the Netherlands
| | - B Vermin
- Department of Intensive care, Spaarne Gasthuis, Haarlem, Hoofddorp, the Netherlands
| | | | - K S Schmitz
- Department of Viroscience, Erasmus Medical Center, Rotterdam, the Netherlands
| | - K Bakhtiari
- Department of Molecular Hematology, Sanquin Research, Amsterdam, the Netherlands
| | - J C M Meijers
- Department of Molecular Hematology, Sanquin Research, Amsterdam, the Netherlands
| | - E C M van Gorp
- Department of Viroscience, Erasmus Medical Center, Rotterdam, the Netherlands
| | - K R Short
- School of Chemistry and Molecular Biosciences, The University of Queensland, St Lucia, Australia
| | - B Haagmans
- Department of Viroscience, Erasmus Medical Center, Rotterdam, the Netherlands
| | - R D de Vries
- Department of Viroscience, Erasmus Medical Center, Rotterdam, the Netherlands
| | - D A M P J Gommers
- Department of Intensive Care, Erasmus MC, Rotterdam, the Netherlands
| | - H Endeman
- Department of Intensive Care, Erasmus MC, Rotterdam, the Netherlands
| | - M Goeijenbier
- Department of Intensive care, Spaarne Gasthuis, Haarlem, Hoofddorp, the Netherlands; Department of Intensive Care, Erasmus MC, Rotterdam, the Netherlands.
| |
Collapse
|
30
|
Gonjilashvili A, Tatishvili S. The interplay between Sars-Cov-2 infection related cardiovascular diseases and depression. Common mechanisms, shared symptoms. AMERICAN HEART JOURNAL PLUS : CARDIOLOGY RESEARCH AND PRACTICE 2024; 38:100364. [PMID: 38510743 PMCID: PMC10945907 DOI: 10.1016/j.ahjo.2024.100364] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/18/2023] [Revised: 01/05/2024] [Accepted: 01/06/2024] [Indexed: 03/22/2024]
Abstract
In 2020 the World Health organization announced a pandemic due to the outbreak of the Coronavirus disease 19. Pneumonia was the most common manifestation of the Sars-Cov-2 infection, however, clinical papers describe Sars-Cov-2 associated cardiovascular pathologies, such as ACS, myopericarditis, cardiomyopathies, dysrhythmias, as leading causes of increased morbidity and mortality. The short and long term prognosis of Sars-Cov-2-related cardiovascular diseases was defined not only by the disease severity itself but also by associated conditions and complications, among which mental health issues (stress, depression and anxiety) have a negative impact. The interplay between Sars-Cov-2 infection, cardiovascular disease and depression may be explained by hyperinflammation, unhealthy lifestyle and inter-organ communication, mediated by extracellular vesicles (EV) and non-coding MicroRNA (miRNA). The long Covid syndrome is characterized with orthostatic hypotension, impaired cardiac and cerebral perfusion, postural orthostatic tachycardia syndrome (POTS), syncope, chest pain, dyspnea, palpitation, chronic fatigue syndrome, 'brain fog', memory, cognitive and sleep difficulties, depression and anxiety. From a clinical point of view these symptoms may be considered as common symptoms representing not only a cardiac but also a neurological/psychiatric problem. Consequently assessment of these symptoms are of paramount importance. Due to their complexity, management of these patients requires multidisciplinary care.
Collapse
|
31
|
Gheitasi H, Sabbaghian M, Shekarchi AA, Mirmazhary AA, Poortahmasebi V. Exosome-mediated regulation of inflammatory pathway during respiratory viral disease. Virol J 2024; 21:30. [PMID: 38273382 PMCID: PMC10811852 DOI: 10.1186/s12985-024-02297-y] [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: 10/06/2023] [Accepted: 01/13/2024] [Indexed: 01/27/2024] Open
Abstract
Viruses have developed many mechanisms by which they can stimulate or inhibit inflammation and cause various diseases, including viral respiratory diseases that kill many people every year. One of the mechanisms that viruses use to induce or inhibit inflammation is exosomes. Exosomes are small membrane nanovesicles (30-150 nm) released from cells that contain proteins, DNA, and coding and non-coding RNA species. They are a group of extracellular vesicles that cells can take up to produce and mediate communication. Intercellular effect exosomes can deliver a broad confine of biological molecules, containing nucleic acids, proteins, and lipids, to the target cell, where they can convey therapeutic or pathogenic consequences through the modulation of inflammation and immune processes. Recent research has shown that exosomes can deliver entire virus genomes or virions to distant target cells, then the delivered viruses can escape the immune system and infect cells. Adenoviruses, orthomyxoviruses, paramyxoviruses, respiratory syncytial viruses, picornaviruses, coronaviruses, and rhinoviruses are mostly related to respiratory diseases. In this article, we will first discuss the current knowledge of exosomes. We will learn about the relationship between exosomes and viral infections, and We mention the inflammations caused by viruses in the airways, the role of exosomes in them, and finally, we examine the relationship between the viruses as mentioned earlier, and the regulation of inflammatory pathways that play a role in causing the disease.
Collapse
Affiliation(s)
- Hamidreza Gheitasi
- Department of Bacteriology and Virology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mohammad Sabbaghian
- Department of Bacteriology and Virology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Ali Akbar Shekarchi
- Department of Pathology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Amir Ali Mirmazhary
- Department of Bacteriology and Virology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Vahdat Poortahmasebi
- Department of Bacteriology and Virology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran.
- Research Center for Clinical Virology, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
32
|
He S, Blombäck M, Wallén H. COVID-19: Not a thrombotic disease but a thromboinflammatory disease. Ups J Med Sci 2024; 129:9863. [PMID: 38327640 PMCID: PMC10845889 DOI: 10.48101/ujms.v129.9863] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/02/2023] [Revised: 09/17/2023] [Accepted: 10/21/2023] [Indexed: 02/09/2024] Open
Abstract
While Coronavirus Disease in 2019 (COVID-19) may no longer be classified as a global public health emergency, it still poses a significant risk at least due to its association with thrombotic events. This study aims to reaffirm our previous hypothesis that COVID-19 is fundamentally a thrombotic disease. To accomplish this, we have undertaken an extensive literature review focused on assessing the comprehensive impact of COVID-19 on the entire hemostatic system. Our analysis revealed that severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection significantly enhances the initiation of thrombin generation. However, it is noteworthy that the thrombin generation may be modulated by specific anticoagulants present in patients' plasma. Consequently, higher levels of fibrinogen appear to play a more pivotal role in promoting coagulation in COVID-19, as opposed to thrombin generation. Furthermore, the viral infection can stimulate platelet activation either through widespread dissemination from the lungs to other organs or localized effects on platelets themselves. An imbalance between Von Willebrand Factor (VWF) and ADAMTS-13 also contributes to an exaggerated platelet response in this disease, in addition to elevated D-dimer levels, coupled with a significant increase in fibrin viscoelasticity. This paradoxical phenotype has been identified as 'fibrinolysis shutdown'. To clarify the pathogenesis underlying these hemostatic disorders in COVID-19, we also examined published data, tracing the reaction process of relevant proteins and cells, from ACE2-dependent viral invasion, through induced tissue inflammation, endothelial injury, and innate immune responses, to occurrence of thrombotic events. We therefrom understand that COVID-19 should no longer be viewed as a thrombotic disease solely based on abnormalities in fibrin clot formation and proteolysis. Instead, it should be regarded as a thromboinflammatory disorder, incorporating both classical elements of cellular inflammation and their intricate interactions with the specific coagulopathy.
Collapse
Affiliation(s)
- Shu He
- Department of Clinical Sciences, Danderyd Hospital, Karolinska Institutet, Stockholm, Sweden
- Division of Coagulation Research, Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden
| | - Margareta Blombäck
- Department of Clinical Sciences, Danderyd Hospital, Karolinska Institutet, Stockholm, Sweden
- Division of Coagulation Research, Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden
| | - Håkan Wallén
- Department of Clinical Sciences, Danderyd Hospital, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
33
|
D’Avila H, Lima CNR, Rampinelli PG, Mateus LCO, de Sousa Silva RV, Correa JR, de Almeida PE. Lipid Metabolism Modulation during SARS-CoV-2 Infection: A Spotlight on Extracellular Vesicles and Therapeutic Prospects. Int J Mol Sci 2024; 25:640. [PMID: 38203811 PMCID: PMC10778989 DOI: 10.3390/ijms25010640] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2023] [Revised: 12/05/2023] [Accepted: 12/12/2023] [Indexed: 01/12/2024] Open
Abstract
Extracellular vesicles (EVs) have a significant impact on the pathophysiological processes associated with various diseases such as tumors, inflammation, and infection. They exhibit molecular, biochemical, and entry control characteristics similar to viral infections. Viruses, on the other hand, depend on host metabolic machineries to fulfill their biosynthetic requirements. Due to potential advantages such as biocompatibility, biodegradation, and efficient immune activation, EVs have emerged as potential therapeutic targets against the SARS-CoV-2 infection. Studies on COVID-19 patients have shown that they frequently have dysregulated lipid profiles, which are associated with an increased risk of severe repercussions. Lipid droplets (LDs) serve as organelles with significant roles in lipid metabolism and energy homeostasis as well as having a wide range of functions in infections. The down-modulation of lipids, such as sphingolipid ceramide and eicosanoids, or of the transcriptional factors involved in lipogenesis seem to inhibit the viral multiplication, suggesting their involvement in the virus replication and pathogenesis as well as highlighting their potential as targets for drug development. Hence, this review focuses on the role of modulation of lipid metabolism and EVs in the mechanism of immune system evasion during SARS-CoV-2 infection and explores the therapeutic potential of EVs as well as application for delivering therapeutic substances to mitigate viral infections.
Collapse
Affiliation(s)
- Heloisa D’Avila
- Cell Biology Laboratory, Department of Biology, Federal University of Juiz de Fora, Juiz de Fora 36036-900, Brazil; (H.D.); (P.G.R.); (L.C.O.M.); (R.V.d.S.S.)
| | | | - Pollianne Garbero Rampinelli
- Cell Biology Laboratory, Department of Biology, Federal University of Juiz de Fora, Juiz de Fora 36036-900, Brazil; (H.D.); (P.G.R.); (L.C.O.M.); (R.V.d.S.S.)
| | - Laiza Camila Oliveira Mateus
- Cell Biology Laboratory, Department of Biology, Federal University of Juiz de Fora, Juiz de Fora 36036-900, Brazil; (H.D.); (P.G.R.); (L.C.O.M.); (R.V.d.S.S.)
| | - Renata Vieira de Sousa Silva
- Cell Biology Laboratory, Department of Biology, Federal University of Juiz de Fora, Juiz de Fora 36036-900, Brazil; (H.D.); (P.G.R.); (L.C.O.M.); (R.V.d.S.S.)
| | - José Raimundo Correa
- Laboratory of Microscopy and Microanalysis, University of Brasília, Brasília 70910-900, Brazil;
| | - Patrícia Elaine de Almeida
- Cell Biology Laboratory, Department of Biology, Federal University of Juiz de Fora, Juiz de Fora 36036-900, Brazil; (H.D.); (P.G.R.); (L.C.O.M.); (R.V.d.S.S.)
| |
Collapse
|
34
|
Heidari Z, Naeimzadeh Y, Fallahi J, Savardashtaki A, Razban V, Khajeh S. The Role of Tissue Factor In Signaling Pathways of Pathological Conditions and Angiogenesis. Curr Mol Med 2024; 24:1135-1151. [PMID: 37817529 DOI: 10.2174/0115665240258746230919165935] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2023] [Revised: 07/10/2023] [Accepted: 07/27/2023] [Indexed: 10/12/2023]
Abstract
Tissue factor (TF) is an integral transmembrane protein associated with the extrinsic coagulation pathway. TF gene expression is regulated in response to inflammatory cytokines, bacterial lipopolysaccharides, and mechanical injuries. TF activity may be affected by phosphorylation of its cytoplasmic domain and alternative splicing. TF acts as the primary initiator of physiological hemostasis, which prevents local bleeding at the injury site. However, aberrant expression of TF, accompanied by the severity of diseases and infections under various pathological conditions, triggers multiple signaling pathways that support thrombosis, angiogenesis, inflammation, and metastasis. Protease-activated receptors (PARs) are central in the downstream signaling pathways of TF. In this study, we have reviewed the TF signaling pathways in different pathological conditions, such as wound injury, asthma, cardiovascular diseases (CVDs), viral infections, cancer and pathological angiogenesis. Angiogenic activities of TF are critical in the repair of wound injuries and aggressive behavior of tumors, which are mainly performed by the actions of vascular endothelial growth factor (VEGF) and hypoxia-inducible factor-1 (HIF1-α). Pro-inflammatory effects of TF have been reported in asthma, CVDs and viral infections, including COVID-19, which result in tissue hypertrophy, inflammation, and thrombosis. TF-FVII induces angiogenesis via clotting-dependent and -independent mechanisms. Clottingdependent angiogenesis is induced via the generation of thrombin and cross-linked fibrin network, which facilitate vessel infiltration and also act as a reservoir for endothelial cells (ECs) growth factors. Expression of TF in tumor cells and ECs triggers clotting-independent angiogenesis through induction of VEGF, urokinase-type plasminogen activator (uPAR), early growth response 1 (EGR1), IL8, and cysteine-rich angiogenic inducer 61 (Cyr61).
Collapse
Affiliation(s)
- Zahra Heidari
- Department of Molecular Medicine, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Yasaman Naeimzadeh
- Department of Molecular Medicine, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Jafar Fallahi
- Department of Molecular Medicine, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Amir Savardashtaki
- Department of Medical Biotechnology, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran
- Infertility Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Vahid Razban
- Department of Molecular Medicine, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Sahar Khajeh
- Bone and Joint Diseases Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| |
Collapse
|
35
|
Eichhorn T, Weiss R, Huber S, Ebeyer-Masotta M, Mostageer M, Emprechtinger R, Knabl L, Knabl L, Würzner R, Weber V. Expression of Tissue Factor and Platelet/Leukocyte Markers on Extracellular Vesicles Reflect Platelet-Leukocyte Interaction in Severe COVID-19. Int J Mol Sci 2023; 24:16886. [PMID: 38069209 PMCID: PMC10707108 DOI: 10.3390/ijms242316886] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2023] [Revised: 11/21/2023] [Accepted: 11/24/2023] [Indexed: 12/18/2023] Open
Abstract
Severe COVID-19 is frequently associated with thromboembolic complications. Increased platelet activation and platelet-leukocyte aggregate formation can amplify thrombotic responses by inducing tissue factor (TF) expression on leukocytes. Here, we characterized TF-positive extracellular vesicles (EVs) and their cellular origin in 12 patients suffering from severe COVID-19 (time course, 134 samples overall) and 25 healthy controls. EVs exposing phosphatidylserine (PS) were characterized by flow cytometry. Their cellular origin was determined by staining with anti-CD41, anti-CD45, anti-CD235a, and anti-CD105 as platelet, leukocyte, red blood cell, and endothelial markers. We further investigated the association of EVs with TF, platelet factor 4 (PF4), C-reactive protein (CRP), and high mobility group box-1 protein (HMGB-1). COVID-19 patients showed higher levels of PS-exposing EVs compared to controls. The majority of these EVs originated from platelets. A higher amount of EVs in patient samples was associated with CRP, HMGB-1, PF4, and TF as compared to EVs from healthy donors. In COVID-19 samples, 16.5% of all CD41+ EVs displayed the leukocyte marker CD45, and 55.5% of all EV aggregates (CD41+CD45+) co-expressed TF, which reflects the interaction of platelets and leukocytes in COVID-19 on an EV level.
Collapse
Affiliation(s)
- Tanja Eichhorn
- Center for Biomedical Technology, Department for Biomedical Research, University for Continuing Education Krems, 3500 Krems, Austria; (R.W.); (M.E.-M.); (M.M.); (V.W.)
| | - René Weiss
- Center for Biomedical Technology, Department for Biomedical Research, University for Continuing Education Krems, 3500 Krems, Austria; (R.W.); (M.E.-M.); (M.M.); (V.W.)
| | - Silke Huber
- Institute of Hygiene and Medical Microbiology, Medical University of Innsbruck, 6020 Innsbruck, Austria; (S.H.); (R.W.)
| | - Marie Ebeyer-Masotta
- Center for Biomedical Technology, Department for Biomedical Research, University for Continuing Education Krems, 3500 Krems, Austria; (R.W.); (M.E.-M.); (M.M.); (V.W.)
| | - Marwa Mostageer
- Center for Biomedical Technology, Department for Biomedical Research, University for Continuing Education Krems, 3500 Krems, Austria; (R.W.); (M.E.-M.); (M.M.); (V.W.)
| | - Robert Emprechtinger
- Faculty of Health and Medicine, University for Continuing Education Krems, 3500 Krems, Austria;
| | - Ludwig Knabl
- Department of Internal Medicine, Hospital St. Vinzenz, 6511 Zams, Austria;
| | | | - Reinhard Würzner
- Institute of Hygiene and Medical Microbiology, Medical University of Innsbruck, 6020 Innsbruck, Austria; (S.H.); (R.W.)
| | - Viktoria Weber
- Center for Biomedical Technology, Department for Biomedical Research, University for Continuing Education Krems, 3500 Krems, Austria; (R.W.); (M.E.-M.); (M.M.); (V.W.)
| |
Collapse
|
36
|
Wan N, Shi J, Xu J, Huang J, Gan D, Tang M, Li X, Huang Y, Li P. Gasdermin D: A Potential New Auxiliary Pan-Biomarker for the Detection and Diagnosis of Diseases. Biomolecules 2023; 13:1664. [PMID: 38002346 PMCID: PMC10669528 DOI: 10.3390/biom13111664] [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: 10/08/2023] [Revised: 11/10/2023] [Accepted: 11/14/2023] [Indexed: 11/26/2023] Open
Abstract
Pyroptosis is a form of programmed cell death mediated by gasdermins, particularly gasdermin D (GSDMD), which is widely expressed in tissues throughout the body. GSDMD belongs to the gasdermin family, which is expressed in a variety of cell types including epithelial cells and immune cells. It is involved in the regulation of anti-inflammatory responses, leading to its differential expression in a wide range of diseases. In this review, we provide an overview of the current understanding of the major activation mechanisms and effector pathways of GSDMD. Subsequently, we examine the importance and role of GSDMD in different diseases, highlighting its potential as a pan-biomarker. We specifically focus on the biological characteristics of GSDMD in several diseases and its promising role in diagnosis, early detection, and differential diagnosis. Furthermore, we discuss the application of GSDMD in predicting prognosis and monitoring treatment efficacy in cancer. This review proposes a new strategy to guide therapeutic decision-making and suggests potential directions for further research into GSDMD.
Collapse
Affiliation(s)
- Ningyi Wan
- Department of Clinical Laboratory, The Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, China
| | - Jing Shi
- Department of Clinical Laboratory, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Jianguo Xu
- Department of Clinical Laboratory, The Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, China
| | - Juan Huang
- Department of Information Center, The Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, China
| | - Delu Gan
- Department of Clinical Laboratory, The Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, China
| | - Min Tang
- Key Laboratory of Medical Diagnostics Designated by Chinese Ministry of Education, Chongqing Medical University, Chongqing 400016, China
| | - Xiaohan Li
- Department of Clinical Laboratory, The Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, China
| | - Ying Huang
- Department of Clinical Laboratory, The Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, China
| | - Pu Li
- Department of Clinical Laboratory, The Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, China
| |
Collapse
|
37
|
Sachetto ATA, Mackman N. Monocyte Tissue Factor Expression: Lipopolysaccharide Induction and Roles in Pathological Activation of Coagulation. Thromb Haemost 2023; 123:1017-1033. [PMID: 37168007 PMCID: PMC10615589 DOI: 10.1055/a-2091-7006] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2021] [Accepted: 05/08/2023] [Indexed: 05/13/2023]
Abstract
The coagulation system is a part of the mammalian host defense system. Pathogens and pathogen components, such as bacterial lipopolysaccharide (LPS), induce tissue factor (TF) expression in circulating monocytes that then activates the coagulation protease cascade. Formation of a clot limits dissemination of pathogens, enhances the recruitment of immune cells, and facilitates killing of pathogens. However, excessive activation of coagulation can lead to thrombosis. Here, we review studies on the mechanism of LPS induction of TF expression in monocytes and its contribution to thrombosis and disseminated intravascular coagulation. Binding of LPS to Toll-like receptor 4 on monocytes induces a transient expression of TF that involves activation of intracellular signaling pathways and binding of various transcription factors, such as c-rel/p65 and c-Fos/c-Jun, to the TF promoter. Inhibition of TF in endotoxemia and sepsis models reduces activation of coagulation and improves survival. Studies with endotoxemic mice showed that hematopoietic cells and myeloid cells play major roles in the activation of coagulation. Monocyte TF expression is also increased after surgery. Activated monocytes release TF-positive extracellular vesicles (EVs) and levels of circulating TF-positive EVs are increased in endotoxemic mice and in patients with sepsis. More recently, it was shown that inflammasomes contribute to the induction of TF expression and activation of coagulation in endotoxemic mice. Taken together, these studies indicate that monocyte TF plays a major role in activation of coagulation. Selective inhibition of monocyte TF expression may reduce pathologic activation of coagulation in sepsis and other diseases without affecting hemostasis.
Collapse
Affiliation(s)
- Ana T. A. Sachetto
- Division of Hematology, Department of Medicine, UNC Blood Research Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States
| | - Nigel Mackman
- Division of Hematology, Department of Medicine, UNC Blood Research Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States
| |
Collapse
|
38
|
Min L, Bao H, Bu F, Li X, Guo Q, Liu M, Zhu S, Meng J, Zhang S, Wang S. Machine-Learning-Assisted Procoagulant Extracellular Vesicle Barcode Assay toward High-Performance Evaluation of Thrombosis-Induced Death Risk in Cancer Patients. ACS NANO 2023; 17:19914-19924. [PMID: 37791763 DOI: 10.1021/acsnano.3c04615] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/05/2023]
Abstract
Venous thromboembolism (VTE) is the most fatal complication in cancer patients. Unfortunately, the frequent misdiagnosis of VTE owing to the lack of accurate and efficient evaluation approaches may cause belated medical intervention and even sudden death. Herein, we present a rapid, easily operable, highly specific, and highly sensitive procoagulant extracellular vesicle barcode (PEVB) assay composed of TiO2 nanoflower (TiNFs) for visually evaluating VTE risk in cancer patients. TiNFs demonstrate rapid label-free EV capture capability by the synergetic effect of TiO2-phospholipids molecular interactions and topological interactions between TiNFs and EVs. From ordinary plasma samples, the PEVB assay can evaluate potential VTE risk by integrating TiNFs-based EV capture and in situ EV procoagulant ability test with machine-learning-assisted clinical data analysis. We demonstrate the feasibility of this PEVB assay in VTE risk evaluation by screening 167 cancer patients, as well as the high specificity (97.1%) and high sensitivity (96.8%), fully exceeding the nonspecific and posterior traditional VTE test. Together, we proposed a TiNFs platform allowing for highly accurate and timely diagnosis of VTE in cancer patients.
Collapse
Affiliation(s)
- Li Min
- Department of Gastroenterology, Beijing Friendship Hospital, Capital Medical University, National Key Laboratory of Digestive Health, National Clinical Research Center for Digestive Disease, Beijing Key Laboratory for Precancerous Lesion of Digestive Disease, Beijing, 100050, P. R. China
| | - Han Bao
- CAS Key Laboratory of Bio-inspired Materials and Interfacial Science, Technical Institute of Physics and Chemistry, Chinese Academy of Sciences, Beijing, 100190, P. R. China
| | - Fanqin Bu
- Department of Gastroenterology, Beijing Friendship Hospital, Capital Medical University, National Key Laboratory of Digestive Health, National Clinical Research Center for Digestive Disease, Beijing Key Laboratory for Precancerous Lesion of Digestive Disease, Beijing, 100050, P. R. China
| | - Xueqing Li
- Department of Gastroenterology, The Second Hospital of Shanxi Medical University, Taiyuan, Shanxi Province 030001, P. R. China
| | - Qingdong Guo
- Department of Gastroenterology, Beijing Friendship Hospital, Capital Medical University, National Key Laboratory of Digestive Health, National Clinical Research Center for Digestive Disease, Beijing Key Laboratory for Precancerous Lesion of Digestive Disease, Beijing, 100050, P. R. China
| | - Mingyuan Liu
- Department of Vascular Surgery, Beijing Friendship Hospital, Capital Medical University, Beijing, 100050, P. R. China
| | - Shengtao Zhu
- Department of Gastroenterology, Beijing Friendship Hospital, Capital Medical University, National Key Laboratory of Digestive Health, National Clinical Research Center for Digestive Disease, Beijing Key Laboratory for Precancerous Lesion of Digestive Disease, Beijing, 100050, P. R. China
| | - Jingxin Meng
- CAS Key Laboratory of Bio-inspired Materials and Interfacial Science, Technical Institute of Physics and Chemistry, Chinese Academy of Sciences, Beijing, 100190, P. R. China
- Weiqiao-UCAS Science and Technology Park, Binzhou Institute of Technology, Binzhou City, Shandong Province 256606, P. R. China
- University of Chinese Academy of Sciences (UCAS), Beijing, 100049, P. R. China
| | - Shutian Zhang
- Department of Gastroenterology, Beijing Friendship Hospital, Capital Medical University, National Key Laboratory of Digestive Health, National Clinical Research Center for Digestive Disease, Beijing Key Laboratory for Precancerous Lesion of Digestive Disease, Beijing, 100050, P. R. China
| | - Shutao Wang
- CAS Key Laboratory of Bio-inspired Materials and Interfacial Science, Technical Institute of Physics and Chemistry, Chinese Academy of Sciences, Beijing, 100190, P. R. China
- Weiqiao-UCAS Science and Technology Park, Binzhou Institute of Technology, Binzhou City, Shandong Province 256606, P. R. China
- University of Chinese Academy of Sciences (UCAS), Beijing, 100049, P. R. China
| |
Collapse
|
39
|
Nair S, Nova-Lamperti E, Labarca G, Kulasinghe A, Short KR, Carrión F, Salomon C. Genomic communication via circulating extracellular vesicles and long-term health consequences of COVID-19. J Transl Med 2023; 21:709. [PMID: 37817137 PMCID: PMC10563316 DOI: 10.1186/s12967-023-04552-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2023] [Accepted: 09/22/2023] [Indexed: 10/12/2023] Open
Abstract
COVID-19 continues to affect an unprecedented number of people with the emergence of new variants posing a serious challenge to global health. There is an expansion of knowledge in understanding the pathogenesis of Coronavirus disease 2019 (COVID-19), caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), and the impact of the acute disease on multiple organs. In addition, growing evidence reports that the impact of COVID-19 on different organs persists long after the recovery phase of the disease, leading to long-term consequences of COVID-19. These long-term consequences involve pulmonary as well as extra-pulmonary sequelae of the disease. Noteably, recent research has shown a potential association between COVID-19 and change in the molecular cargo of extracellular vesicles (EVs). EVs are vesicles released by cells and play an important role in cell communication by transfer of bioactive molecules between cells. Emerging evidence shows a strong link between EVs and their molecular cargo, and regulation of metabolism in health and disease. This review focuses on current knowledge about EVs and their potential role in COVID-19 pathogenesis, their current and future implications as tools for biomarker and therapeutic development and their possible effects on long-term impact of COVID-19.
Collapse
Affiliation(s)
- Soumyalekshmi Nair
- Translational Extracellular Vesicles in Obstetrics and Gynae-Oncology Group, UQ Centre for Clinical Research, Royal Brisbane and Women's Hospital, Faculty of Medicine, The University of Queensland, Brisbane, Qld, 4072, Australia
| | - Estefania Nova-Lamperti
- Molecular and Translational Immunology Laboratory, Clinical Biochemistry and Immunology Department, Pharmacy Faculty, Universidad de Concepción, Concepción, Chile
| | - Gonzalo Labarca
- Molecular and Translational Immunology Laboratory, Clinical Biochemistry and Immunology Department, Pharmacy Faculty, Universidad de Concepción, Concepción, Chile
| | - Arutha Kulasinghe
- Frazer Institute, Faculty of Medicine, The University of Queensland, Brisbane, Qld, 4102, Australia
| | - Kirsty R Short
- School of Chemistry and Molecular Biosciences, The University of Queensland, Brisbane, Australia
| | - Flavio Carrión
- Departamento de Investigación, Postgrado y Educación Continua (DIPEC), Facultad de Ciencias de la Salud, Universidad del Alba, Santiago, Chile.
| | - Carlos Salomon
- Translational Extracellular Vesicles in Obstetrics and Gynae-Oncology Group, UQ Centre for Clinical Research, Royal Brisbane and Women's Hospital, Faculty of Medicine, The University of Queensland, Brisbane, Qld, 4072, Australia.
- Departamento de Investigación, Postgrado y Educación Continua (DIPEC), Facultad de Ciencias de la Salud, Universidad del Alba, Santiago, Chile.
| |
Collapse
|
40
|
Potere N, Garrad E, Kanthi Y, Di Nisio M, Kaplanski G, Bonaventura A, Connors JM, De Caterina R, Abbate A. NLRP3 inflammasome and interleukin-1 contributions to COVID-19-associated coagulopathy and immunothrombosis. Cardiovasc Res 2023; 119:2046-2060. [PMID: 37253117 PMCID: PMC10893977 DOI: 10.1093/cvr/cvad084] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/28/2022] [Revised: 01/30/2023] [Accepted: 02/21/2023] [Indexed: 06/01/2023] Open
Abstract
Immunothrombosis-immune-mediated activation of coagulation-is protective against pathogens, but excessive immunothrombosis can result in pathological thrombosis and multiorgan damage, as in severe coronavirus disease 2019 (COVID-19). The NACHT-, LRR-, and pyrin domain-containing protein 3 (NLRP3) inflammasome produces major proinflammatory cytokines of the interleukin (IL)-1 family, IL-1β and IL-18, and induces pyroptotic cell death. Activation of the NLRP3 inflammasome pathway also promotes immunothrombotic programs including release of neutrophil extracellular traps and tissue factor by leukocytes, and prothrombotic responses by platelets and the vascular endothelium. NLRP3 inflammasome activation occurs in patients with COVID-19 pneumonia. In preclinical models, NLRP3 inflammasome pathway blockade restrains COVID-19-like hyperinflammation and pathology. Anakinra, recombinant human IL-1 receptor antagonist, showed safety and efficacy and is approved for the treatment of hypoxaemic COVID-19 patients with early signs of hyperinflammation. The non-selective NLRP3 inhibitor colchicine reduced hospitalization and death in a subgroup of COVID-19 outpatients but is not approved for the treatment of COVID-19. Additional COVID-19 trials testing NLRP3 inflammasome pathway blockers are inconclusive or ongoing. We herein outline the contribution of immunothrombosis to COVID-19-associated coagulopathy, and review preclinical and clinical evidence suggesting an engagement of the NLRP3 inflammasome pathway in the immunothrombotic pathogenesis of COVID-19. We also summarize current efforts to target the NLRP3 inflammasome pathway in COVID-19, and discuss challenges, unmet gaps, and the therapeutic potential that inflammasome-targeted strategies may provide for inflammation-driven thrombotic disorders including COVID-19.
Collapse
Affiliation(s)
- Nicola Potere
- Department of Medicine and Ageing Sciences, ‘G. d’Annunzio’ University, Via Luigi Polacchi 11, Chieti 66100, Italy
| | - Evan Garrad
- Laboratory of Vascular Thrombosis and Inflammation, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA
- University of Missouri School of Medicine, Columbia, MO, USA
| | - Yogendra Kanthi
- Laboratory of Vascular Thrombosis and Inflammation, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Marcello Di Nisio
- Department of Medicine and Ageing Sciences, ‘G. d’Annunzio’ University, Via Luigi Polacchi 11, Chieti 66100, Italy
| | - Gilles Kaplanski
- Aix-Marseille Université, INSERM, INRAE, Marseille, France
- Division of Internal Medicine and Clinical Immunology, Assistance Publique - Hôpitaux de Marseille, Hôpital Conception, Aix-Marseille Université, Marseille, France
| | - Aldo Bonaventura
- Department of Internal Medicine, Medicina Generale 1, Medical Center, Ospedale di Circolo e Fondazione Macchi, ASST Sette Laghi, Varese, Italy
| | - Jean Marie Connors
- Division of Hematology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
| | - Raffaele De Caterina
- University Cardiology Division, Pisa University Hospital, Pisa, Italy
- Chair and Postgraduate School of Cardiology, University of Pisa, Pisa, Italy
- Fondazione Villa Serena per la Ricerca, Città Sant’Angelo, Pescara, Italy
| | - Antonio Abbate
- Robert M. Berne Cardiovascular Research Center, Department of Medicine, Division of Cardiovascular Medicine, University of Virginia, 415 Lane Rd (MR5), PO Box 801394, Charlottesville, VA 22903, USA
| |
Collapse
|
41
|
George MS, Sanchez J, Rollings C, Fear D, Irving P, Sinclair LV, Schurich A. Extracellular vesicles in COVID-19 convalescence can regulate T cell metabolism and function. iScience 2023; 26:107280. [PMID: 37520724 PMCID: PMC10371842 DOI: 10.1016/j.isci.2023.107280] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2023] [Revised: 05/11/2023] [Accepted: 06/30/2023] [Indexed: 08/01/2023] Open
Abstract
Long-term T cell dysregulation has been reported following COVID-19 disease. Prolonged T cell activation is associated with disease severity and may be implicated in producing long-covid symptoms. Here, we assess the role of extracellular vesicles (EV) in regulating T cell function over several weeks post COVID-19 disease. We find that alterations in cellular origin and protein content of EV in COVID-19 convalescence are linked to initial disease severity. We demonstrate that convalescent donor-derived EV can alter the function and metabolic rewiring of CD4 and CD8 T cells. Of note, EV following mild, but not severe disease, show distinctly immune-suppressive properties, reducing T cell effector cytokine production and glucose metabolism. Mechanistically our data indicate the involvement of EV-surface ICAM-1 in facilitating EV-T cell interaction. Our data demonstrate that circulatory EV are phenotypically and functionally altered several weeks following acute infection, suggesting a role for EV as long-term immune modulators.
Collapse
Affiliation(s)
- Molly S. George
- Department of Infectious Diseases, School of Immunology and Microbial Sciences, King’s College London, London SE1 9RT, UK
| | - Jenifer Sanchez
- Department of Infectious Diseases, School of Immunology and Microbial Sciences, King’s College London, London SE1 9RT, UK
| | - Christina Rollings
- Cell Signalling and Immunology, School of Life Sciences, University of Dundee, Scotland DD1 5EH, UK
| | - David Fear
- Peter Gorer Department of Immunobiology, School of Immunology and Microbial Sciences, King’s College London, London SE1 9RT, UK
| | - Peter Irving
- Peter Gorer Department of Immunobiology, School of Immunology and Microbial Sciences, King’s College London, London SE1 9RT, UK
- Department of Gastroenterology, Guy’s and St Thomas’ Hospital, London SE1 9RT, UK
| | - Linda V. Sinclair
- Cell Signalling and Immunology, School of Life Sciences, University of Dundee, Scotland DD1 5EH, UK
| | - Anna Schurich
- Department of Infectious Diseases, School of Immunology and Microbial Sciences, King’s College London, London SE1 9RT, UK
| |
Collapse
|
42
|
Santoro L, Zaccone V, Falsetti L, Ruggieri V, Danese M, Miro C, Di Giorgio A, Nesci A, D’Alessandro A, Moroncini G, Santoliquido A. Role of Endothelium in Cardiovascular Sequelae of Long COVID. Biomedicines 2023; 11:2239. [PMID: 37626735 PMCID: PMC10452509 DOI: 10.3390/biomedicines11082239] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2023] [Revised: 07/26/2023] [Accepted: 08/03/2023] [Indexed: 08/27/2023] Open
Abstract
The global action against coronavirus disease 2019 (COVID-19), caused by SARS-CoV-2 infection, shed light on endothelial dysfunction. Although SARS-CoV-2 primarily affects the pulmonary system, multiple studies have documented pan-vascular involvement in COVID-19. The virus is able to penetrate the endothelial barrier, damaging it directly or indirectly and causing endotheliitis and multi-organ injury. Several mechanisms cooperate to development of endothelial dysfunction, including endothelial cell injury and pyroptosis, hyperinflammation and cytokine storm syndrome, oxidative stress and reduced nitric oxide bioavailability, glycocalyx disruption, hypercoagulability, and thrombosis. After acute-phase infection, some patients reported signs and symptoms of a systemic disorder known as long COVID, in which a broad range of cardiovascular (CV) disorders emerged. To date, the exact pathophysiology of long COVID remains unclear: in addition to the persistence of acute-phase infection mechanisms, specific pathways of CV damage have been postulated, such as persistent viral reservoirs in the heart or an autoimmune response to cardiac antigens through molecular mimicry. The aim of this review is to provide an overview of the main molecular patterns of enduring endothelial activation following SARS-CoV-2 infection and to offer the latest summary of CV complications in long COVID.
Collapse
Affiliation(s)
- Luca Santoro
- Department of Cardiovascular and Thoracic Sciences, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00168 Rome, Italy; (L.S.); (A.D.G.); (A.N.); (A.D.); (A.S.)
| | - Vincenzo Zaccone
- Department of Emergency Medicine, Internal and Sub-Intensive Medicine, Azienda Ospedaliero-Universitaria delle Marche, 60126 Ancona, Italy
| | - Lorenzo Falsetti
- Clinica Medica, Department of Clinical and Molecular Sciences, Università Politecnica delle Marche, 60126 Ancona, Italy; (L.F.); (G.M.)
| | - Vittorio Ruggieri
- Department of Internal Medicine, Università Cattolica del Sacro Cuore, 00168 Rome, Italy; (V.R.); (M.D.); (C.M.)
| | - Martina Danese
- Department of Internal Medicine, Università Cattolica del Sacro Cuore, 00168 Rome, Italy; (V.R.); (M.D.); (C.M.)
| | - Chiara Miro
- Department of Internal Medicine, Università Cattolica del Sacro Cuore, 00168 Rome, Italy; (V.R.); (M.D.); (C.M.)
| | - Angela Di Giorgio
- Department of Cardiovascular and Thoracic Sciences, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00168 Rome, Italy; (L.S.); (A.D.G.); (A.N.); (A.D.); (A.S.)
| | - Antonio Nesci
- Department of Cardiovascular and Thoracic Sciences, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00168 Rome, Italy; (L.S.); (A.D.G.); (A.N.); (A.D.); (A.S.)
| | - Alessia D’Alessandro
- Department of Cardiovascular and Thoracic Sciences, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00168 Rome, Italy; (L.S.); (A.D.G.); (A.N.); (A.D.); (A.S.)
| | - Gianluca Moroncini
- Clinica Medica, Department of Clinical and Molecular Sciences, Università Politecnica delle Marche, 60126 Ancona, Italy; (L.F.); (G.M.)
| | - Angelo Santoliquido
- Department of Cardiovascular and Thoracic Sciences, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00168 Rome, Italy; (L.S.); (A.D.G.); (A.N.); (A.D.); (A.S.)
- Università Cattolica del Sacro Cuore, 00168 Rome, Italy
| |
Collapse
|
43
|
Conti M, Minniti M, Tiné M, De Francesco M, Gaeta R, Nieri D, Semenzato U, Biondini D, Camera M, Cosio MG, Saetta M, Celi A, Bazzan E, Neri T. Extracellular Vesicles in Pulmonary Hypertension: A Dangerous Liaison? BIOLOGY 2023; 12:1099. [PMID: 37626985 PMCID: PMC10451884 DOI: 10.3390/biology12081099] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Revised: 08/04/2023] [Accepted: 08/05/2023] [Indexed: 08/27/2023]
Abstract
The term pulmonary hypertension (PH) refers to different conditions, all characterized by increased pressure and resistance in the pulmonary arterial bed. PH has a wide range of causes (essentially, cardiovascular, pulmonary, or connective tissue disorders); however, idiopathic (i.e., without a clear cause) PH exists. This chronic, progressive, and sometimes devastating disease can finally lead to right heart failure and eventually death, through pulmonary vascular remodeling and dysfunction. The exact nature of PH pathophysiology is sometimes still unclear. Extracellular vesicles (EVs), previously known as apoptotic bodies, microvesicles, and exosomes, are small membrane-bound vesicles that are generated by almost all cell types and can be detected in a variety of physiological fluids. EVs are involved in intercellular communication, thus influencing immunological response, inflammation, embryogenesis, aging, and regenerative processes. Indeed, they transport chemokines, cytokines, lipids, RNA and miRNA, and other biologically active molecules. Although the precise functions of EVs are still not fully known, there is mounting evidence that they can play a significant role in the pathophysiology of PH. In this review, after briefly recapping the key stages of PH pathogenesis, we discuss the current evidence on the functions of EVs both as PH biomarkers and potential participants in the distinct pathways of disease progression.
Collapse
Affiliation(s)
- Maria Conti
- Department of Cardiac, Thoracic, Vascular Sciences and Public Health, University of Padova, 35128 Padova, Italy; (M.C.); (M.T.); (U.S.); (D.B.); (M.G.C.); (M.S.); (E.B.)
- Centro Cardiologico Monzino IRCCS, 20138 Milan, Italy;
| | - Marianna Minniti
- Centro Dipartimentale di Biologia Cellulare Cardiorespiratoria, Dipartimento di Patologia Chirurgica, Medica, Molecolare e dell’Area Critica, Università Degli Studi di Pisa, 56124 Pisa, Italy; (M.M.); (M.D.F.); (R.G.); (D.N.); (A.C.)
| | - Mariaenrica Tiné
- Department of Cardiac, Thoracic, Vascular Sciences and Public Health, University of Padova, 35128 Padova, Italy; (M.C.); (M.T.); (U.S.); (D.B.); (M.G.C.); (M.S.); (E.B.)
| | - Miriam De Francesco
- Centro Dipartimentale di Biologia Cellulare Cardiorespiratoria, Dipartimento di Patologia Chirurgica, Medica, Molecolare e dell’Area Critica, Università Degli Studi di Pisa, 56124 Pisa, Italy; (M.M.); (M.D.F.); (R.G.); (D.N.); (A.C.)
| | - Roberta Gaeta
- Centro Dipartimentale di Biologia Cellulare Cardiorespiratoria, Dipartimento di Patologia Chirurgica, Medica, Molecolare e dell’Area Critica, Università Degli Studi di Pisa, 56124 Pisa, Italy; (M.M.); (M.D.F.); (R.G.); (D.N.); (A.C.)
| | - Dario Nieri
- Centro Dipartimentale di Biologia Cellulare Cardiorespiratoria, Dipartimento di Patologia Chirurgica, Medica, Molecolare e dell’Area Critica, Università Degli Studi di Pisa, 56124 Pisa, Italy; (M.M.); (M.D.F.); (R.G.); (D.N.); (A.C.)
| | - Umberto Semenzato
- Department of Cardiac, Thoracic, Vascular Sciences and Public Health, University of Padova, 35128 Padova, Italy; (M.C.); (M.T.); (U.S.); (D.B.); (M.G.C.); (M.S.); (E.B.)
| | - Davide Biondini
- Department of Cardiac, Thoracic, Vascular Sciences and Public Health, University of Padova, 35128 Padova, Italy; (M.C.); (M.T.); (U.S.); (D.B.); (M.G.C.); (M.S.); (E.B.)
- Department of Medicine, University of Padova, 35128 Padova, Italy
| | - Marina Camera
- Centro Cardiologico Monzino IRCCS, 20138 Milan, Italy;
- Department of Pharmaceutical Sciences, Università Degli Studi di Milano, 20138 Milan, Italy
| | - Manuel G. Cosio
- Department of Cardiac, Thoracic, Vascular Sciences and Public Health, University of Padova, 35128 Padova, Italy; (M.C.); (M.T.); (U.S.); (D.B.); (M.G.C.); (M.S.); (E.B.)
- Meakins-Christie Laboratories, Respiratory Division, McGill University, Montreal, QC H3A 0G4, Canada
| | - Marina Saetta
- Department of Cardiac, Thoracic, Vascular Sciences and Public Health, University of Padova, 35128 Padova, Italy; (M.C.); (M.T.); (U.S.); (D.B.); (M.G.C.); (M.S.); (E.B.)
| | - Alessandro Celi
- Centro Dipartimentale di Biologia Cellulare Cardiorespiratoria, Dipartimento di Patologia Chirurgica, Medica, Molecolare e dell’Area Critica, Università Degli Studi di Pisa, 56124 Pisa, Italy; (M.M.); (M.D.F.); (R.G.); (D.N.); (A.C.)
| | - Erica Bazzan
- Department of Cardiac, Thoracic, Vascular Sciences and Public Health, University of Padova, 35128 Padova, Italy; (M.C.); (M.T.); (U.S.); (D.B.); (M.G.C.); (M.S.); (E.B.)
| | - Tommaso Neri
- Centro Dipartimentale di Biologia Cellulare Cardiorespiratoria, Dipartimento di Patologia Chirurgica, Medica, Molecolare e dell’Area Critica, Università Degli Studi di Pisa, 56124 Pisa, Italy; (M.M.); (M.D.F.); (R.G.); (D.N.); (A.C.)
| |
Collapse
|
44
|
Su P, Wu Y, Xie F, Zheng Q, Chen L, Liu Z, Meng X, Zhou F, Zhang L. A Review of Extracellular Vesicles in COVID-19 Diagnosis, Treatment, and Prevention. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2206095. [PMID: 37144543 PMCID: PMC10323633 DOI: 10.1002/advs.202206095] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/19/2022] [Revised: 04/15/2023] [Indexed: 05/06/2023]
Abstract
The 2019 novel coronavirus disease (COVID-19) pandemic caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is ongoing, and has necessitated scientific efforts in disease diagnosis, treatment, and prevention. Interestingly, extracellular vesicles (EVs) have been crucial in these developments. EVs are a collection of various nanovesicles which are delimited by a lipid bilayer. They are enriched in proteins, nucleic acids, lipids, and metabolites, and naturally released from different cells. Their natural material transport properties, inherent long-term recycling ability, excellent biocompatibility, editable targeting, and inheritance of parental cell properties make EVs one of the most promising next-generation drug delivery nanocarriers and active biologics. During the COVID-19 pandemic, many efforts have been made to exploit the payload of natural EVs for the treatment of COVID-19. Furthermore, strategies that use engineered EVs to manufacture vaccines and neutralization traps have produced excellent efficacy in animal experiments and clinical trials. Here, the recent literature on the application of EVs in COVID-19 diagnosis, treatment, damage repair, and prevention is reviewed. And the therapeutic value, application strategies, safety, and biotoxicity in the production and clinical applications of EV agents for COVID-19 treatment, as well as inspiration for using EVs to block and eliminate novel viruses are discussed.
Collapse
Affiliation(s)
- Peng Su
- Department of Breast SurgeryZhejiang Provincial People's HospitalHangzhou310014P. R. China
- Institutes of Biology and Medical ScienceSoochow UniversitySuzhou215123P. R. China
| | - Yuchen Wu
- Department of Clinical MedicineThe First School of MedicineWenzhou Medical UniversityWenzhouZhejiang325035P. R. China
| | - Feng Xie
- Institutes of Biology and Medical ScienceSoochow UniversitySuzhou215123P. R. China
| | - Qinghui Zheng
- Department of Breast SurgeryZhejiang Provincial People's HospitalHangzhou310014P. R. China
| | - Long Chen
- Center for Translational MedicineThe Affiliated Zhangjiagang Hospital of Soochow UniversityZhangjiagangJiangsu215600China
| | - Zhuang Liu
- Institute of Functional Nano and Soft Materials (FUNSOM)Jiangsu Key Laboratory for Carbon‐Based Functional Materials and DevicesSoochow UniversitySuzhouJiangsu215123China
| | - Xuli Meng
- Department of Breast SurgeryZhejiang Provincial People's HospitalHangzhou310014P. R. China
| | - Fangfang Zhou
- Institutes of Biology and Medical ScienceSoochow UniversitySuzhou215123P. R. China
| | - Long Zhang
- Department of Breast SurgeryZhejiang Provincial People's HospitalHangzhou310014P. R. China
- MOE Laboratory of Biosystems Homeostasis & Protection and Innovation Center for Cell Signaling NetworkLife Sciences InstituteZhejiang UniversityHangzhou310058P. R. China
- Cancer CenterZhejiang UniversityHangzhouZhejiang310058P. R. China
| |
Collapse
|
45
|
Boccatonda A, Campello E, Simion C, Simioni P. Long-term hypercoagulability, endotheliopathy and inflammation following acute SARS-CoV-2 infection. Expert Rev Hematol 2023; 16:1035-1048. [PMID: 38018136 DOI: 10.1080/17474086.2023.2288154] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2023] [Accepted: 11/22/2023] [Indexed: 11/30/2023]
Abstract
INTRODUCTION both symptomatic and asymptomatic SARS-CoV-2 infections - coined Coronavirus disease 2019 (COVID-19) - have been linked to a higher risk of cardiovascular events after recovery. AREAS COVERED our review aims to summarize the latest evidence on the increased thrombotic and cardiovascular risk in recovered COVID-19 patients and to examine the pathophysiological mechanisms underlying the interplay among endothelial dysfunction, inflammatory response and coagulation in long-COVID. We performed a systematic search of studies on hypercoagulability, endothelial dysfunction and inflammation after SARS-CoV-2 infection. EXPERT OPINION endothelial dysfunction is a major pathophysiological mechanism responsible for most clinical manifestations in COVID-19. The pathological activation of endothelial cells by a virus infection results in a pro-adhesive and chemokine-secreting phenotype, which in turn promotes the recruitment of circulating leukocytes. Cardiovascular events after COVID-19 appear to be related to persistent immune dysregulation. Patients with long-lasting symptoms display higher amounts of proinflammatory molecules such as tumor necrosis factor-α, interferon γ and interleukins 2 and 6. Immune dysregulation can trigger the activation of the coagulation pathway. The formation of extensive microclots in vivo, both during acute COVID-19 and in long-COVID-19, appears to be a relevant mechanism responsible for persistent symptoms and cardiovascular events.
Collapse
Affiliation(s)
- Andrea Boccatonda
- Internal Medicine, Bentivoglio Hospital, AUSL Bologna, Bentivoglio, Italy
| | - Elena Campello
- General Medicine and Thrombotic and Hemorrhagic Diseases Unit, Department of Medicine, University Hospital of Padova, Padova, Italy
| | - Chiara Simion
- General Medicine and Thrombotic and Hemorrhagic Diseases Unit, Department of Medicine, University Hospital of Padova, Padova, Italy
| | - Paolo Simioni
- General Medicine and Thrombotic and Hemorrhagic Diseases Unit, Department of Medicine, University Hospital of Padova, Padova, Italy
| |
Collapse
|
46
|
Ryan TAJ, Hooftman A, Rehill AM, Johansen MD, Brien ECO, Toller-Kawahisa JE, Wilk MM, Day EA, Weiss HJ, Sarvari P, Vozza EG, Schramm F, Peace CG, Zotta A, Miemczyk S, Nalkurthi C, Hansbro NG, McManus G, O'Doherty L, Gargan S, Long A, Dunne J, Cheallaigh CN, Conlon N, Carty M, Fallon PG, Mills KHG, Creagh EM, Donnell JSO, Hertzog PJ, Hansbro PM, McLoughlin RM, Wygrecka M, Preston RJS, Zasłona Z, O'Neill LAJ. Dimethyl fumarate and 4-octyl itaconate are anticoagulants that suppress Tissue Factor in macrophages via inhibition of Type I Interferon. Nat Commun 2023; 14:3513. [PMID: 37316487 PMCID: PMC10265568 DOI: 10.1038/s41467-023-39174-1] [Citation(s) in RCA: 30] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2023] [Accepted: 06/01/2023] [Indexed: 06/16/2023] Open
Abstract
Excessive inflammation-associated coagulation is a feature of infectious diseases, occurring in such conditions as bacterial sepsis and COVID-19. It can lead to disseminated intravascular coagulation, one of the leading causes of mortality worldwide. Recently, type I interferon (IFN) signaling has been shown to be required for tissue factor (TF; gene name F3) release from macrophages, a critical initiator of coagulation, providing an important mechanistic link between innate immunity and coagulation. The mechanism of release involves type I IFN-induced caspase-11 which promotes macrophage pyroptosis. Here we find that F3 is a type I IFN-stimulated gene. Furthermore, F3 induction by lipopolysaccharide (LPS) is inhibited by the anti-inflammatory agents dimethyl fumarate (DMF) and 4-octyl itaconate (4-OI). Mechanistically, inhibition of F3 by DMF and 4-OI involves suppression of Ifnb1 expression. Additionally, they block type I IFN- and caspase-11-mediated macrophage pyroptosis, and subsequent TF release. Thereby, DMF and 4-OI inhibit TF-dependent thrombin generation. In vivo, DMF and 4-OI suppress TF-dependent thrombin generation, pulmonary thromboinflammation, and lethality induced by LPS, E. coli, and S. aureus, with 4-OI additionally attenuating inflammation-associated coagulation in a model of SARS-CoV-2 infection. Our results identify the clinically approved drug DMF and the pre-clinical tool compound 4-OI as anticoagulants that inhibit TF-mediated coagulopathy via inhibition of the macrophage type I IFN-TF axis.
Collapse
Affiliation(s)
- Tristram A J Ryan
- School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin 2, Ireland
| | - Alexander Hooftman
- School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin 2, Ireland
| | - Aisling M Rehill
- Irish Centre for Vascular Biology, School of Pharmacy and Biomolecular Sciences, RCSI University of Medicine and Health Sciences, Dublin 2, Ireland
| | - Matt D Johansen
- Centre for Inflammation, Centenary Institute and University of Technology Sydney, Faculty of Science, Sydney, NSW, Australia
| | - Eóin C O' Brien
- Host Pathogen Interactions Group, School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin 2, Ireland
| | - Juliana E Toller-Kawahisa
- School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin 2, Ireland
| | - Mieszko M Wilk
- School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin 2, Ireland
- Department of Immunology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Kraków, Poland
| | - Emily A Day
- School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin 2, Ireland
| | - Hauke J Weiss
- School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin 2, Ireland
| | - Pourya Sarvari
- Center for Infection and Genomics of the Lung, German Center for Lung Research (DZL), Faculty of Medicine, Justus Liebig University, Giessen, Germany
| | - Emilio G Vozza
- Host Pathogen Interactions Group, School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin 2, Ireland
| | - Fabian Schramm
- Center for Infection and Genomics of the Lung, German Center for Lung Research (DZL), Faculty of Medicine, Justus Liebig University, Giessen, Germany
| | - Christian G Peace
- School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin 2, Ireland
| | - Alessia Zotta
- School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin 2, Ireland
| | - Stefan Miemczyk
- Centre for Inflammation, Centenary Institute and University of Technology Sydney, Faculty of Science, Sydney, NSW, Australia
| | - Christina Nalkurthi
- Centre for Inflammation, Centenary Institute and University of Technology Sydney, Faculty of Science, Sydney, NSW, Australia
| | - Nicole G Hansbro
- Centre for Inflammation, Centenary Institute and University of Technology Sydney, Faculty of Science, Sydney, NSW, Australia
| | - Gavin McManus
- School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin 2, Ireland
| | - Laura O'Doherty
- Department of Infectious Diseases, St. James's Hospital, Dublin, Ireland
- Clinical Research Facility, St. James's Hospital, Dublin, Ireland
- Department of Clinical Medicine, School of Medicine, Trinity Translational Medicine Institute, Trinity College Dublin, Dublin, Ireland
| | - Siobhan Gargan
- Department of Clinical Medicine, School of Medicine, Trinity Translational Medicine Institute, Trinity College Dublin, Dublin, Ireland
| | - Aideen Long
- Department of Clinical Medicine, School of Medicine, Trinity Translational Medicine Institute, Trinity College Dublin, Dublin, Ireland
| | - Jean Dunne
- Department of Immunology, St James's Hospital, Dublin, Ireland
| | - Clíona Ní Cheallaigh
- Department of Infectious Diseases, St. James's Hospital, Dublin, Ireland
- Department of Clinical Medicine, School of Medicine, Trinity Translational Medicine Institute, Trinity College Dublin, Dublin, Ireland
| | - Niall Conlon
- Clinical Research Facility, St. James's Hospital, Dublin, Ireland
- Department of Clinical Medicine, School of Medicine, Trinity Translational Medicine Institute, Trinity College Dublin, Dublin, Ireland
- Department of Immunology, St James's Hospital, Dublin, Ireland
| | - Michael Carty
- School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin 2, Ireland
| | - Padraic G Fallon
- Department of Clinical Medicine, School of Medicine, Trinity Translational Medicine Institute, Trinity College Dublin, Dublin, Ireland
- School of Medicine, Trinity Biomedical Sciences Institute, Trinity College, Dublin 2, Ireland
| | - Kingston H G Mills
- School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin 2, Ireland
| | - Emma M Creagh
- School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin 2, Ireland
| | - James S O' Donnell
- Irish Centre for Vascular Biology, School of Pharmacy and Biomolecular Sciences, RCSI University of Medicine and Health Sciences, Dublin 2, Ireland
| | - Paul J Hertzog
- Centre for Innate Immunity and Infectious Diseases, Hudson Institute of Medical Research, Clayton, VIC, Australia
- Department of Molecular and Translational Science, Monash University, Clayton, VIC, Australia
| | - Philip M Hansbro
- Centre for Inflammation, Centenary Institute and University of Technology Sydney, Faculty of Science, Sydney, NSW, Australia
| | - Rachel M McLoughlin
- Host Pathogen Interactions Group, School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin 2, Ireland
| | - Małgorzata Wygrecka
- Center for Infection and Genomics of the Lung, German Center for Lung Research (DZL), Faculty of Medicine, Justus Liebig University, Giessen, Germany
| | - Roger J S Preston
- Irish Centre for Vascular Biology, School of Pharmacy and Biomolecular Sciences, RCSI University of Medicine and Health Sciences, Dublin 2, Ireland
| | - Zbigniew Zasłona
- School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin 2, Ireland
| | - Luke A J O'Neill
- School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin 2, Ireland.
| |
Collapse
|
47
|
Deng Y, Zou Y, Song X, Jiang A, Wang M, Qin Q, Song Y, Yue C, Yang D, Yu B, Lu H, Zheng Y. Potential of extracellular vesicles for early prediction of severity and potential risk stratification in critical inflammatory diseases. J Cell Commun Signal 2023:10.1007/s12079-023-00763-w. [PMID: 37195382 DOI: 10.1007/s12079-023-00763-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2022] [Accepted: 05/02/2023] [Indexed: 05/18/2023] Open
Abstract
Some acute inflammatory diseases are often exacerbated during or after hospitalization, leading to some severe manifestations like systemic inflammatory response syndrome, multiple organ failure, and high mortality. Early clinical predictors of disease severity are urgently needed to optimize patient management for better prognosis. The existing clinical scoring system and laboratory tests cannot circumvent the problems of low sensitivity and limited specificity. Extracellular vesicles (EVs) are heterogeneous nanosecretory vesicles containing various biomolecules related to immune regulation, inflammation activation, and inflammation-related complications. This review provides an overview of EVs as inflammatory mediators, inflammatory signaling pathway regulators, promoters of inflammatory exacerbation, and markers of severity and prognosis. Currently, although relevant biomarkers are clinically available or are in the preclinical research stage, searching for new markers and detection methods is still warranted, as the problems of low sensitivity/specificity, cumbersome laboratory operation and high cost still plague clinicians. In-depth study of EVs might open a door in the search for novel predictors.
Collapse
Affiliation(s)
- Yuchuan Deng
- Department of Biotherapy,Cancer Center and State Key Laboratory of Biotherapy,West China Hospital, Sichuan University, Chengdu, 6110041, Sichuan, China
| | - Yu Zou
- Department of Biotherapy,Cancer Center and State Key Laboratory of Biotherapy,West China Hospital, Sichuan University, Chengdu, 6110041, Sichuan, China
| | - Xiaoshuang Song
- Department of Biotherapy,Cancer Center and State Key Laboratory of Biotherapy,West China Hospital, Sichuan University, Chengdu, 6110041, Sichuan, China
| | - Ailing Jiang
- Department of Biotherapy,Cancer Center and State Key Laboratory of Biotherapy,West China Hospital, Sichuan University, Chengdu, 6110041, Sichuan, China
| | - Mao Wang
- Department of Biotherapy,Cancer Center and State Key Laboratory of Biotherapy,West China Hospital, Sichuan University, Chengdu, 6110041, Sichuan, China
| | - Qin Qin
- Department of Biotherapy,Cancer Center and State Key Laboratory of Biotherapy,West China Hospital, Sichuan University, Chengdu, 6110041, Sichuan, China
| | - Yiran Song
- Department of Biotherapy,Cancer Center and State Key Laboratory of Biotherapy,West China Hospital, Sichuan University, Chengdu, 6110041, Sichuan, China
| | - Chao Yue
- Center of Excellence for Pancreatitis, Institute of Integrated Traditional Chinese and Western Medicine, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan Province, China
| | - Dujiang Yang
- Center of Excellence for Pancreatitis, Institute of Integrated Traditional Chinese and Western Medicine, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan Province, China
| | - Bo Yu
- Zhejiang Pushkang Biotechnology Co., Ltd, Shaoxing, Zhejiang Province, China
| | - Huimin Lu
- Center of Excellence for Pancreatitis, Institute of Integrated Traditional Chinese and Western Medicine, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan Province, China.
| | - Yu Zheng
- Department of Biotherapy,Cancer Center and State Key Laboratory of Biotherapy,West China Hospital, Sichuan University, Chengdu, 6110041, Sichuan, China.
| |
Collapse
|
48
|
Wang M, Yu F, Chang W, Zhang Y, Zhang L, Li P. Inflammasomes: a rising star on the horizon of COVID-19 pathophysiology. Front Immunol 2023; 14:1185233. [PMID: 37251383 PMCID: PMC10213254 DOI: 10.3389/fimmu.2023.1185233] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Accepted: 05/02/2023] [Indexed: 05/31/2023] Open
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is a contagious respiratory virus that is the cause of the coronavirus disease 2019 (COVID-19) pandemic which has posed a serious threat to public health. COVID-19 is characterized by a wide spectrum of clinical manifestations, ranging from asymptomatic infection to mild cold-like symptoms, severe pneumonia or even death. Inflammasomes are supramolecular signaling platforms that assemble in response to danger or microbial signals. Upon activation, inflammasomes mediate innate immune defense by favoring the release of proinflammatory cytokines and triggering pyroptotic cell death. Nevertheless, abnormalities in inflammasome functioning can result in a variety of human diseases such as autoimmune disorders and cancer. A growing body of evidence has showed that SARS-CoV-2 infection can induce inflammasome assembly. Dysregulated inflammasome activation and consequent cytokine burst have been associated with COVID-19 severity, alluding to the implication of inflammasomes in COVID-19 pathophysiology. Accordingly, an improved understanding of inflammasome-mediated inflammatory cascades in COVID-19 is essential to uncover the immunological mechanisms of COVID-19 pathology and identify effective therapeutic approaches for this devastating disease. In this review, we summarize the most recent findings on the interplay between SARS-CoV-2 and inflammasomes and the contribution of activated inflammasomes to COVID-19 progression. We dissect the mechanisms involving the inflammasome machinery in COVID-19 immunopathogenesis. In addition, we provide an overview of inflammasome-targeted therapies or antagonists that have potential clinical utility in COVID-19 treatment.
Collapse
Affiliation(s)
- Man Wang
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao, China
| | | | | | | | | | - Peifeng Li
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao, China
| |
Collapse
|
49
|
Sayyadi M, Hassani S, Shams M, Dorgalaleh A. Status of major hemostatic components in the setting of COVID-19: the effect on endothelium, platelets, coagulation factors, fibrinolytic system, and complement. Ann Hematol 2023; 102:1307-1322. [PMID: 37074380 PMCID: PMC10115391 DOI: 10.1007/s00277-023-05234-1] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2023] [Accepted: 04/14/2023] [Indexed: 04/20/2023]
Abstract
The coagulation, fibrinolytic, anticoagulation, and complement systems are in delicate balance with the vessel wall endothelium ensuring appropriate hemostasis. Coagulopathy in coronavirus disease 2019 (COVID-19) is not a simple disorder of one hemostatic component but a complicated process affecting most of the hemostasis system. COVID-19 disturbs the balance between the procoagulant systems and the regulatory mechanisms. Here, we investigate the effect of COVID-19 on key hemostatic components, including platelets, endothelial cells, coagulation factors, fibrinolytic system, anticoagulant protein system, and complement system, to improve our understanding of the pathophysiological processes underlying COVID-19 coagulopathy based on evidence.
Collapse
Affiliation(s)
- Mohammad Sayyadi
- Department of Medical Laboratory Sciences, School of Allied Medical Sciences, Arak University of Medical Sciences, Arak, Iran
| | - Saeed Hassani
- Department of Medical Laboratory Sciences, School of Allied Medical Sciences, Arak University of Medical Sciences, Arak, Iran.
| | - Mahmood Shams
- Cellular and Molecular Biology Research Center, Health Research Institute, Babol University of Medical Sciences, Babol, Iran
| | | |
Collapse
|
50
|
Turner S, Khan MA, Putrino D, Woodcock A, Kell DB, Pretorius E. Long COVID: pathophysiological factors and abnormalities of coagulation. Trends Endocrinol Metab 2023; 34:321-344. [PMID: 37080828 PMCID: PMC10113134 DOI: 10.1016/j.tem.2023.03.002] [Citation(s) in RCA: 101] [Impact Index Per Article: 50.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/27/2023] [Revised: 03/10/2023] [Accepted: 03/14/2023] [Indexed: 04/22/2023]
Abstract
Acute COVID-19 infection is followed by prolonged symptoms in approximately one in ten cases: known as Long COVID. The disease affects ~65 million individuals worldwide. Many pathophysiological processes appear to underlie Long COVID, including viral factors (persistence, reactivation, and bacteriophagic action of SARS CoV-2); host factors (chronic inflammation, metabolic and endocrine dysregulation, immune dysregulation, and autoimmunity); and downstream impacts (tissue damage from the initial infection, tissue hypoxia, host dysbiosis, and autonomic nervous system dysfunction). These mechanisms culminate in the long-term persistence of the disorder characterized by a thrombotic endothelialitis, endothelial inflammation, hyperactivated platelets, and fibrinaloid microclots. These abnormalities of blood vessels and coagulation affect every organ system and represent a unifying pathway for the various symptoms of Long COVID.
Collapse
Affiliation(s)
- Simone Turner
- Department of Physiological Sciences, Faculty of Science, Stellenbosch University, Stellenbosch, Private Bag X1, Matieland, 7602, South Africa
| | - M Asad Khan
- North West Lung Centre, Manchester University Hospitals, Manchester, M23 9LT, UK
| | - David Putrino
- Department of Rehabilitation and Human Performance, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Ashley Woodcock
- The University of Manchester, Oxford Road, Manchester, M13 9PL, UK; Manchester Academic Health Science Centre, CityLabs, Manchester, M13 9NQ, UK
| | - 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, Kemitorvet, 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
|