1
|
Bagoly Z, Tóth EL, Orbán-Kálmándi R, Lóczi L, Deli T, Török O, Kozma B, Baráth S, Singh P, Hevessy Z, Tóth J, Katona É, Molnár S, Krasznai ZT. Complex evaluation of coagulation, fibrinolysis, and inflammatory cytokines in SARS-CoV-2 infected pregnant women: a prospective, case-control study. Front Immunol 2025; 16:1556878. [PMID: 40303405 PMCID: PMC12037393 DOI: 10.3389/fimmu.2025.1556878] [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: 01/07/2025] [Accepted: 03/04/2025] [Indexed: 05/02/2025] Open
Abstract
Background Given the physiological hemostasis changes during pregnancy, limited data exists on COVID-19-induced inflammatory response and hemostasis alterations in pregnant women. Objectives To test a comprehensive set of hemostasis and inflammatory cytokines in pregnancies with/without COVID-19 and correlate results with maternal and perinatal outcomes. Patients/methods In this observational case-control study, 100 women with acute COVID-19 at 24-40 gestational weeks (COVID-19+ group), and 100 healthy, age- and gestational week-matched, SARS-CoV-2 negative pregnant women (32 with proven recovery of COVID-19) were enrolled. All women were outpatients with mild/no symptoms at admission. Detailed hemostasis (fibrinogen, FVIII, FXIII, VWF, plasminogen, α2-plasmin inhibitor, PAI-1, thrombin generation, clot lysis, D-dimer) and inflammatory cytokine/chemokine panels were performed. Clinical parameters of pregnancy, labor and postpartum period were registered. Results COVID-19+ women exhibited significantly lower FVIII, FXIII, plasminogen, higher VWF levels, decreased peak thrombin and enhanced clot lysis vs. controls. Despite mild/no symptoms, significantly elevated cytokine levels, including IL-6, INF-γ, MCP-1, and IL-18 were observed in COVID-19+ pregnancies, associated with distinct hemostasis alterations. Admission IL-1β, and IL-33 were significantly lower, while IL-18 was significantly higher in cases when COVID-19 became more severe, along with significantly decreased FVIII, FXIII and plasminogen. In the COVID-19+ group, postpartum hemorrhage (PPH) developed in 4 cases, associated with significantly reduced plasminogen, α2-plasmin inhibitor, and increased IL-8, IL-17A, IL-23 levels. Conclusion In third trimester mild/asymptomatic COVID-19+ pregnancies, marked inflammatory cytokine changes, hemostasis alterations and enhanced fibrinolysis were found. A potential link between inflammation and PPH in the context of COVID-19 warrants further research.
Collapse
Affiliation(s)
- Zsuzsa Bagoly
- Division of Clinical Laboratory Sciences, Department of Laboratory Medicine, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Eszter Lilla Tóth
- Department of Obstetrics and Gynecology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
- Doctoral School of Molecular Medicine, University of Debrecen, Debrecen, Hungary
| | - Rita Orbán-Kálmándi
- Healthcare Industry Institute, Faculty of Pharmacy, University of Debrecen, Debrecen, Hungary
| | - Linda Lóczi
- Division of Clinical Laboratory Sciences, Department of Laboratory Medicine, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
- Kálmán Laki Doctoral School, University of Debrecen, Debrecen, Hungary
| | - Tamás Deli
- Department of Obstetrics and Gynecology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Olga Török
- Department of Obstetrics and Gynecology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Bence Kozma
- Department of Obstetrics and Gynecology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Sándor Baráth
- Department of Laboratory Medicine, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Parvind Singh
- Kálmán Laki Doctoral School, University of Debrecen, Debrecen, Hungary
- Department of Laboratory Medicine, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Zsuzsanna Hevessy
- Department of Laboratory Medicine, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Judit Tóth
- Department of Laboratory Medicine, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Éva Katona
- Division of Clinical Laboratory Sciences, Department of Laboratory Medicine, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Szabolcs Molnár
- Department of Obstetrics and Gynecology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Zoárd Tibor Krasznai
- Department of Obstetrics and Gynecology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| |
Collapse
|
2
|
Saha I, Banerjee O, Sarkar Biswas S, Mukherjee S. COVID-19 beyond the lungs: Unraveling its vascular impact and cardiovascular complications-mechanisms and therapeutic implications. Sci Prog 2025; 108:368504251322069. [PMID: 40091392 PMCID: PMC11912160 DOI: 10.1177/00368504251322069] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/19/2025]
Abstract
COVID-19, caused by severe acute respiratory syndrome corona virus 2 (SARS-CoV-2), is primarily a respiratory illness but significantly affects the cardiovascular system as well. After entering the body through the respiratory tract, the virus directly and indirectly disrupts the vascular system. Vascular endothelial cells (ECs), which express ACE2 and TMPRSS2, are targets for viral invasion. However, the predominant cause of widespread vascular damage is the "cytokine storm" induced by the immune response. This leads to EC activation, inflammation, neutrophil activation, and neutrophil-platelet aggregation, causing endothelial injury. Additionally, increased expression of plasminogen activator inhibitor-1 disrupts the balance between prothrombotic and fibrinolytic processes, while activation of the renin-angiotensin-aldosterone system adds oxidative stress to the vascular endothelium. In the heart, SARS-CoV-2 invades ECs, leading to apoptosis and pyroptosis, exacerbated by inflammation and elevated catecholamines. These factors contribute to arrhythmias, strokes, and myocardial infarction in severe cases of COVID-19. This narrative review aims to explore the mechanisms by which SARS-CoV-2 affects the cardiovascular system and to highlight the resulting complications. It also identifies research gaps and discusses potential therapeutic strategies to mitigate the cardiovascular impacts of COVID-19.
Collapse
Affiliation(s)
- Ishita Saha
- Department of Physiology, Medical College & Hospital, Kolkata, West Bengal, India
| | - Oly Banerjee
- Department of Medical Laboratory Technology, School of Allied Health Sciences, Swami Vivekananda University, Bara Kanthalia, West Bengal, India
| | | | - Sandip Mukherjee
- Department of Physiology, Serampore College, Hooghly, West Bengal, India
| |
Collapse
|
3
|
Obeagu EI, Tukur M, Akaba K. Impacts of COVID-19 on hemostasis: coagulation abnormalities and management perspectives. Ann Med Surg (Lond) 2024; 86:5844-5850. [PMID: 39359765 PMCID: PMC11444586 DOI: 10.1097/ms9.0000000000002237] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2024] [Accepted: 05/22/2024] [Indexed: 10/04/2024] Open
Abstract
The COVID-19 pandemic caused by SARS-CoV-2 has transcended its initial characterization as a respiratory illness, revealing substantial implications for hemostasis and coagulation pathways. COVID-19-associated coagulopathies have emerged as critical determinants of disease severity and prognosis, presenting a multifaceted challenge in clinical management. This paper aims to elucidate the intricate interplay between COVID-19 and hemostasis, delving into the underlying mechanisms of coagulation abnormalities, exploring the spectrum of thrombotic complications, and discussing evolving management strategies. Therapeutic interventions and anticoagulation strategies tailored for managing COVID-19-related coagulopathies form a significant focus, encompassing prophylactic and therapeutic approaches, heparin-based therapies, and individualized treatment paradigms. This paper underscores the imperative for ongoing research endeavors to refine diagnostic modalities, identify novel therapeutic targets, and ascertain long-term sequelae of COVID-19-induced coagulation abnormalities. Ultimately, a comprehensive understanding of the intricate relationship between COVID-19 and hemostasis is pivotal in devising effective management strategies to mitigate thrombotic risks, improve clinical outcomes, and pave the way for tailored interventions in affected individuals.
Collapse
Affiliation(s)
| | - Muhammad Tukur
- Department of Science Education, Faculty of Education, Kampala International University, Kampala, Uganda
| | - Kingsley Akaba
- Department of Haematology, University of Calabar, Calabar, Cross-River State, Nigeria
| |
Collapse
|
4
|
Tan X, Gao X, Zheng H, Yuan H, Liu H, Ran Q, Luo M. Platelet dysfunction caused by differentially expressed genes as key pathogenic mechanisms in COVID-19. Minerva Cardiol Angiol 2024; 72:517-534. [PMID: 38804627 DOI: 10.23736/s2724-5683.24.06501-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/29/2024]
Abstract
At the end of 2019, the novel coronavirus disease 2019 (COVID-19) caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) became prevalent worldwide, which brought a heavy medical burden and tremendous economic losses to the world population. In addition to the common clinical respiratory symptoms such as fever, cough and headache, patients with COVID-19 often have hematological diseases, especially platelet dysfunction. Platelet dysfunction usually leads to multiple organ dysfunction, which is closely related to patient severity or mortality. In addition, studies have confirmed significant changes in the gene expression profile of circulating platelets under SARS-CoV-2 infection, which will further lead to changes in platelet function. At the same time, studies have shown that platelets may absorb SARS-COV-2 mRNA independently of ACE2, which further emphasizes the importance of the stability of platelet function in defense against SARS-CoV-2 infection. This study reviewed the relationship between COVID-19 and platelet and SARS-CoV-2 damage to the circulatory system, and further analyzed the significantly differentially expressed mRNA in platelets after infection with SARS-CoV-2 on the basis of previous studies. The top eight hub genes were identified as NLRP3, MT-CO1, CD86, ICAM1, MT-CYB, CASP8, CXCL8 and CXCR4. Subsequently, the effects of SARS-CoV-2 infection on platelet transcript abnormalities and platelet dysfunction were further explored on the basis of 8 hub genes. Finally, the treatment measures of complications caused by platelet dysfunction in patients with COVID-19 were discussed in detail, so as to provide reference for the prevention, diagnosis and treatment of COVID-19.
Collapse
Affiliation(s)
- Xiaoyong Tan
- Department of Pharmacy, Xuanhan County People's Hospital, Dazhou, China
| | - Xiaojun Gao
- Basic Medicine Research Innovation Center for Cardiometabolic Diseases, Ministry of Education, Southwest Medical University, Luzhou, China
- Key Laboratory of Medical Electrophysiology, Ministry of Education, Drug Discovery Research Center, Southwest Medical University, Luzhou, China
- Laboratory for Cardiovascular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, China
| | - Huanhuan Zheng
- School of Public Health, Southwest Medical University, Luzhou, China
| | - Hui Yuan
- Department of Clinical Medicine, the Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Hong Liu
- Department of Pharmacy, Xuanhan County People's Hospital, Dazhou, China
| | - Qijun Ran
- Department of Pharmacy, Xuanhan County People's Hospital, Dazhou, China
| | - Mao Luo
- Basic Medicine Research Innovation Center for Cardiometabolic Diseases, Ministry of Education, Southwest Medical University, Luzhou, China -
- Key Laboratory of Medical Electrophysiology, Ministry of Education, Drug Discovery Research Center, Southwest Medical University, Luzhou, China
- Laboratory for Cardiovascular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, China
| |
Collapse
|
5
|
Rossio R, Tettamanti M, Galbussera AA, Gualtierotti R, Giachi A, Torri A, Montano N, Fracanzani AL, Bandera A, Nobili A, Peyvandi F. Bleeding and thrombotic events and intensity of heparin therapy in the two first waves of COVID-19. Intern Emerg Med 2024; 19:1577-1583. [PMID: 38761332 DOI: 10.1007/s11739-024-03635-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Accepted: 05/05/2024] [Indexed: 05/20/2024]
Abstract
A systemic inflammatory response occurs during SARS-CoV2 infection and is associated with hypercoagulability and thrombotic events. From March 2020 in our hospital different dosages of low-molecular weight heparin (LMWH) were introduced according to the level of patient care intensity. Because bleeding episodes occurred in hospitalized COVID-19 patients on heparin, the dosage of LMWH at the end of first wave was tailored on the severity of COVID-19. The aim of this study is to describe bleeding and thrombotic events in patients hospitalized with SARS-CoV2 infection on LMWH therapy in the two waves of COVID-19 and analyze the factors associated with these events. Among 1143 patients enrolled in the COVID-19 Network registry, 912 were included in the analysis, 537 of them admitted during the first wave and 375 during the second. Bleeding events were 30 (3.3%): 22 (2.4%) major and 8 (0.9%) non-major. Arterial and venous thrombotic events were 6 (0.7%) and 24 (2.6%). The incidence of venous thrombotic events was higher in the first than in the second wave (0.29% [95% CI 0.20-0.45] events/day vs. 0.05% [95% CI 0.02-0.16]), with a 71% risk reduction (95% CI 22%-94%). The incidence of bleeding was 0.33% (95% CI 0.22-0.50) vs 0.14% events/day (95% CI 0.07-0.28), with no statistical between-wave difference (HR 0.41 95% CI 0.16-1.08). After adjusting for the competing risks of death and comorbidities, patients in the second wave had lower odds to have thrombotic events than in the first wave (0.24 HR [95% C.I. 0.07-0.89]). In this retrospective study on COVID-19 we found a low rate of hemorrhagic and thrombotic events, that may be explained by the absence in the case material of patients admitted to intensive care unit.
Collapse
Affiliation(s)
- Raffaella Rossio
- Angelo Bianchi Bonomi Hemophilia and Thrombosis Center, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Via Sforza 35, Milan, Italy.
| | - Mauro Tettamanti
- Laboratory of Geriatric Epidemiology, Istituto Di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
| | | | - Roberta Gualtierotti
- Angelo Bianchi Bonomi Hemophilia and Thrombosis Center, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Via Sforza 35, Milan, Italy
- Department of Pathophysiology and Transplantation, Università Degli Studi Di Milano, Milan, Italy
| | - Andrea Giachi
- Angelo Bianchi Bonomi Hemophilia and Thrombosis Center, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Via Sforza 35, Milan, Italy
| | - Adriana Torri
- Department of Pathophysiology and Transplantation, Università Degli Studi Di Milano, Milan, Italy
| | - Nicola Montano
- Department of Clinical Sciences and Community Health, Università Degli Studi Di Milano, 20122, Milan, Italy
| | - Anna Ludovica Fracanzani
- Department of Pathophysiology and Transplantation, Unit of Metabolic and Internal Medicine, University of Milan, Milan, Italy
| | - Alessandra Bandera
- Infectious Diseases Unit, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Alessandro Nobili
- Laboratory of Geriatric Epidemiology, Istituto Di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
| | - Flora Peyvandi
- Angelo Bianchi Bonomi Hemophilia and Thrombosis Center, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Via Sforza 35, Milan, Italy
- Department of Pathophysiology and Transplantation, Università Degli Studi Di Milano, Milan, Italy
| |
Collapse
|
6
|
Theresa C, Katebe B, Shibao CA, Kirabo A. Arterial stiffness in adults with Long COVID in sub-Saharan Africa. Physiol Rep 2024; 12:e70029. [PMID: 39251404 PMCID: PMC11386233 DOI: 10.14814/phy2.70029] [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: 06/05/2024] [Revised: 08/22/2024] [Accepted: 08/22/2024] [Indexed: 09/11/2024] Open
Abstract
Severe acute coronavirus-2 (SARS-CoV-2) infection has been associated with endothelial damage, and impaired nitric oxide production, which results in arterial stiffness and increased risk of cardiovascular disease. Long COVID is a term used to describe the persistence or the development of new symptoms that can occur after an acute infection. Little is known about the association between arterial stiffness and Long COVID. An observational, cross-sectional study in which arterial stiffness was measured with pulse wave velocity (PWV) was carried out in 74 participants between 19 and 40 years old (53 with Long COVID, 21 age and gender-matched controls). Data was collected from participants between 1 and 9 months after acute COVID-19 infection using the Complior analyze unit protocol. The Long COVID group had higher carotid-radial-PWV (crPWV) than controls (10 m/s interquartile range [IQR] 8.5-11.2 m/s) versus 8.8 m/s (IQR 7.7-9.2 m/s) as was their carotid-radial-arterial stiffness index (crASI) (2.26 cm/ms (IQR 1.9-2.56 cm/ms) vs. 2.01 cm/ms (IQR 1.82-2.27 cm/ms); p < 0.05) in both. They also had more type-A waveforms, indicating increased arterial stiffening. Peripheral arterial stiffness was higher in adults with Long COVID than in controls who were never infected with SARS-CoV-2 as noted by the elevated levels of crPWV and crASI among adults with Long COVID.
Collapse
Affiliation(s)
- Chikopela Theresa
- Department of Human Physiology, Faculty of MedicineLusaka Apex Medical UniversityLusakaZambia
| | - Bwalya Katebe
- Department of Human Physiology, Faculty of MedicineLusaka Apex Medical UniversityLusakaZambia
| | - Cyndya A. Shibao
- Department of MedicineVanderbilt University Medical CenterNashvilleTennesseeUSA
| | - Annet Kirabo
- Department of MedicineVanderbilt University Medical CenterNashvilleTennesseeUSA
| |
Collapse
|
7
|
Dumitrescu G, Antovic J, Soutari N, Gran C, Antovic A, Al-Abani K, Grip J, Rooyackers O, Taxiarchis A. The role of complement and extracellular vesicles in the development of pulmonary embolism in severe COVID-19 cases. PLoS One 2024; 19:e0309112. [PMID: 39178205 PMCID: PMC11343408 DOI: 10.1371/journal.pone.0309112] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2024] [Accepted: 08/05/2024] [Indexed: 08/25/2024] Open
Abstract
Complement and extracellular vesicles (EVs) association with thrombogenic tendencies is acknowledged, but limited evidence exists for their link to COVID-19 venous thromboembolism. This study aims to examine the relationship between pulmonary embolism and the expression of complement and other proteins related to thrombogenesis in severe Covid-19 patients. We included prospectively 207 severe COVID-19 patients and retrospectively screened for pulmonary embolism (PE). This analysis comprises 20 confirmed PE cases and 20 matched patients without PE. Blood samples taken at the admission in the intensive care unit were analyzed for complement using ELISA. EVs derived from neutrophils, endothelium, or platelets, as well carrying complement or tissue factor were analyzed using flow cytometry. Complement levels were markedly elevated, with a notable increase in C3a and Terminal Complement Complex. The most prevalent EV population was identified as tissue factor (TF)-carrying EVs which peaked in patients with PE during ICU days 4-9. However, for both the complement and analyzed EV populations, no statistically significant differences were found between the patients who developed pulmonary embolism and those who did not. In conclusion, complement factors and EVs expressing tissue factor, along with EVs derived from endothelial cells and platelets, are elevated in severe COVID-19 patients, regardless of the presence of pulmonary embolism. However, the involvement of complement and procoagulant EVs in peripheral plasma in the development of pulmonary embolism is still unclear and requires further investigation.
Collapse
Affiliation(s)
- Gabriel Dumitrescu
- Division of Anaesthesia and Intensive Care, Department of Clinical Science, Intervention and Technology, Karolinska Institutet, and Perioperative and Intensive Care Medicine, Karolinska University Hospital, Stockholm, Sweden
| | - Jovan Antovic
- Department of Molecular Medicine and Surgery, Karolinska Institutet, and Clinical Chemistry, Karolinska University Laboratory, Karolinska University Hospital, Stockholm, Sweden
| | - Nida Soutari
- Department of Molecular Medicine and Surgery, Karolinska Institutet, and Clinical Chemistry, Karolinska University Laboratory, Karolinska University Hospital, Stockholm, Sweden
| | - Charlotte Gran
- Department of Molecular Medicine and Surgery, Karolinska Institutet, and Clinical Chemistry, Karolinska University Laboratory, Karolinska University Hospital, Stockholm, Sweden
| | - Aleksandra Antovic
- Division of Rheumatology, Department of Medicine, Karolinska Institutet, and Unit of Rheumatology, Karolinska University Hospital, Stockholm, Sweden
| | - Kais Al-Abani
- Department of Clinical Physiology, Karolinska University Hospital, Stockholm, Sweden
| | - Jonathan Grip
- Division of Anaesthesia and Intensive Care, Department of Clinical Science, Intervention and Technology, Karolinska Institutet, and Perioperative and Intensive Care Medicine, Karolinska University Hospital, Stockholm, Sweden
| | - Olav Rooyackers
- Division of Anaesthesia and Intensive Care, Department of Clinical Science, Intervention and Technology, Karolinska Institutet, and Perioperative and Intensive Care Medicine, Karolinska University Hospital, Stockholm, Sweden
| | - Apostolos Taxiarchis
- Department of Molecular Medicine and Surgery, Karolinska Institutet, and Clinical Chemistry, Karolinska University Laboratory, Karolinska University Hospital, Stockholm, Sweden
| |
Collapse
|
8
|
Burmeister L. COVID-19 and Type 1 Diabetes. Pediatr Ann 2024; 53:e244-e248. [PMID: 38949873 DOI: 10.3928/19382359-20240502-02] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 07/03/2024]
Abstract
The coronavirus disease 2019 (COVID-19) pandemic brought about several challenges for pediatric patients that were considerably different than those for adult patients. While adult patients had severe illness with associated respiratory and multiorgan failure and subsequent death, pediatric patients typically had milder disease. There were increases in postinfection complications, including an increased incidence of new-onset type 1 diabetes (T1D) following COVID-19 infection, particularly in adolescent patients. Currently, there is increasing concern that COVID-19 infection may be contributing to the development of T1D. This review will provide an overview of COVID-19, T1D, and the increased incidence noted during the pandemic, and the proposed mechanism of development of T1D in this specific patient demographic. Future studies will be needed to understand the long-term impact of the COVID-19 pandemic on T1D in children and adolescents. [Pediatr Ann. 2024;53(7):e244-e248.].
Collapse
|
9
|
Prieto-Campo Á, Zapata-Cachafeiro M, Portela-Romero M, Piñeiro-Lamas M, Figueiras A, Salgado-Barreira Á. Impact of prior use of antiplatelets on COVID-19 susceptibility, progression, and severity: a population-based study. REVISTA ESPANOLA DE CARDIOLOGIA (ENGLISH ED.) 2024; 77:539-546. [PMID: 38191054 DOI: 10.1016/j.rec.2023.12.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/11/2023] [Accepted: 12/19/2023] [Indexed: 01/10/2024]
Abstract
INTRODUCTION AND OBJECTIVES Hypercoagulability and thromboembolism are processes that arise from severe acute respiratory syndrome coronavirus 2 infection and are responsible for a high degree of coronavirus disease 2019 (COVID-19)-related morbidity and mortality. This study sought to assess the effect of antiplatelet drugs on COVID-19 severity (risk of hospitalization and mortality), susceptibility to severe acute respiratory syndrome coronavirus 2 infection, and progression to severe COVID-19. METHODS We conducted a population-based case-control study in a northwestern region of Spain in 2020. The study involved 3060 participants with a positive polymerase chain reaction test who were hospitalized, 26 757 participants with a positive polymerase chain reaction test who were not hospitalized, and 56 785 healthy controls. RESULTS Triflusal seemed to be associated with a significant increase in risk of hospitalization (aOR, 1.97; 95%CI, 1.27-3.04) and susceptibility to infection (OR, 1.45; 95%CI, 1.07-1.96). It also appeared to lead to a nonsignificant increase in the risk of mortality (OR, 2.23; 95%CI, 0.89-5.55) and/or progression to more severe disease stages (OR, 1.42; 95%CI, 0.8-2.51). Aspirin seemed to be associated with a statistically significant decrease in susceptibility to severe acute respiratory syndrome coronavirus 2 infection (OR, 0.92; 95%CI, 0.86-0.98). CONCLUSIONS Triflusal use appears to increase the risk of susceptibility to COVID-19 infection and an even higher risk of hospitalization, whereas the other antiplatelets could be associated with a reduction in the risk of the various outcomes or have no effect on risk. These findings could support reconsideration of triflusal prescription in COVID-19 pandemic situations.
Collapse
Affiliation(s)
- Ángela Prieto-Campo
- Departamento de Salud Pública, Facultad de Farmacia, Universidad de Santiago de Compostela, Santiago de Compostela, A Coruña, Spain
| | - Maruxa Zapata-Cachafeiro
- Departamento de Salud Pública, Facultad de Farmacia, Universidad de Santiago de Compostela, Santiago de Compostela, A Coruña, Spain; Consorcio de Investigación Biomédica en Red de Epidemiología y Salud Pública (CIBERESP), Spain; Institute of Health Research of Santiago de Compostela (IDIS), Santiago de Compostela, A Coruña, Spain.
| | - Manuel Portela-Romero
- Centro de Salud Concepción Arenal, Santiago de Compostela, A Coruña, Spain; Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), Spain
| | - María Piñeiro-Lamas
- Consorcio de Investigación Biomédica en Red de Epidemiología y Salud Pública (CIBERESP), Spain; Institute of Health Research of Santiago de Compostela (IDIS), Santiago de Compostela, A Coruña, Spain
| | - Adolfo Figueiras
- Departamento de Salud Pública, Facultad de Farmacia, Universidad de Santiago de Compostela, Santiago de Compostela, A Coruña, Spain; Consorcio de Investigación Biomédica en Red de Epidemiología y Salud Pública (CIBERESP), Spain; Institute of Health Research of Santiago de Compostela (IDIS), Santiago de Compostela, A Coruña, Spain
| | - Ángel Salgado-Barreira
- Departamento de Salud Pública, Facultad de Farmacia, Universidad de Santiago de Compostela, Santiago de Compostela, A Coruña, Spain; Consorcio de Investigación Biomédica en Red de Epidemiología y Salud Pública (CIBERESP), Spain; Institute of Health Research of Santiago de Compostela (IDIS), Santiago de Compostela, A Coruña, Spain
| |
Collapse
|
10
|
Padín JF, Pérez-Ortiz JM, Redondo-Calvo FJ. Aprotinin (II): Inhalational Administration for the Treatment of COVID-19 and Other Viral Conditions. Int J Mol Sci 2024; 25:7209. [PMID: 39000315 PMCID: PMC11241800 DOI: 10.3390/ijms25137209] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2024] [Revised: 06/25/2024] [Accepted: 06/26/2024] [Indexed: 07/16/2024] Open
Abstract
Aprotinin is a broad-spectrum inhibitor of human proteases that has been approved for the treatment of bleeding in single coronary artery bypass surgery because of its potent antifibrinolytic actions. Following the outbreak of the COVID-19 pandemic, there was an urgent need to find new antiviral drugs. Aprotinin is a good candidate for therapeutic repositioning as a broad-spectrum antiviral drug and for treating the symptomatic processes that characterise viral respiratory diseases, including COVID-19. This is due to its strong pharmacological ability to inhibit a plethora of host proteases used by respiratory viruses in their infective mechanisms. The proteases allow the cleavage and conformational change of proteins that make up their viral capsid, and thus enable them to anchor themselves by recognition of their target in the epithelial cell. In addition, the activation of these proteases initiates the inflammatory process that triggers the infection. The attraction of the drug is not only its pharmacodynamic characteristics but also the possibility of administration by the inhalation route, avoiding unwanted systemic effects. This, together with the low cost of treatment (≈2 Euro/dose), makes it a good candidate to reach countries with lower economic means. In this article, we will discuss the pharmacodynamic, pharmacokinetic, and toxicological characteristics of aprotinin administered by the inhalation route; analyse the main advances in our knowledge of this medication; and the future directions that should be taken in research in order to reposition this medication in therapeutics.
Collapse
Affiliation(s)
- Juan-Fernando Padín
- Department of Medical Sciences, School of Medicine at Ciudad Real, University of Castilla-La Mancha, 13971 Ciudad Real, Spain
| | - José Manuel Pérez-Ortiz
- Facultad HM de Ciencias de la Salud, Universidad Camilo José Cela, 28692 Madrid, Spain
- Instituto de Investigación Sanitaria HM Hospitales, 28015 Madrid, Spain
| | - Francisco Javier Redondo-Calvo
- Department of Medical Sciences, School of Medicine at Ciudad Real, University of Castilla-La Mancha, 13971 Ciudad Real, Spain
- Department of Anaesthesiology and Critical Care Medicine, University General Hospital, 13005 Ciudad Real, Spain
- Translational Research Unit, University General Hospital and Research Institute of Castilla-La Mancha (IDISCAM), 13005 Ciudad Real, Spain
| |
Collapse
|
11
|
Safdari H, Bagheri S, Talkhi N, Saberi Teymourian E, Hosseini Bafghi M, Ahmadi MH. Cq values as an indicator for COVID-19 outcomes: A study of the correlation between laboratory parameters. Immun Inflamm Dis 2024; 12:e1326. [PMID: 38923849 PMCID: PMC11194972 DOI: 10.1002/iid3.1326] [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: 02/13/2024] [Revised: 05/26/2024] [Accepted: 06/10/2024] [Indexed: 06/28/2024] Open
Abstract
OBJECTIVE The ongoing outbreak of the respiratory disease coronavirus disease 2019 (COVID-19) is currently presenting a major global health threat. This pandemic is unprecedented in recent human history. The objective of this study was to examine the relationship between cycle quantitation (Cq) and laboratory parameters in COVID-19 patients, aiming to determine if Cq levels can provide valuable insights into the COVID-19 disease. METHODS This study involved 234 participants who were divided into case and control groups. Real-time PCR tests were used to diagnose COVID-19 cases in the study participants. Blood tests, including complete blood count, C-reactive protein (CRP), erythrocyte sedimentation rate (ESR), lactate dehydrogenase (LDH), D-dimer, IgG, and IgM, were also conducted. Statistical analysis was performed using SPSS 22 software. RESULTS The findings showed that COVID-19-positive cases had significantly higher levels of the neutrophil-to-lymphocyte ratio (NLR), platelet-to-lymphocyte ratio (PLR), D-dimer, ESR, CRP, and LDH compared to normal cases. Additionally, the case group had significantly lower lymphocyte and platelet counts. There was a statistically significant positive correlation between Cq levels and lymphocyte count (r = .124, p = .014). Conversely, there was a statistically significant inverse correlation between Cq levels and NLR (r = -.208, p = .017). Furthermore, the evaluation of hematological, inflammatory, and biochemical indexes in COVID-19 patients using the receiver-operating characteristics curve demonstrated statistically appropriate sensitivity and specificity. CONCLUSION Our outcomes indicated a significant association between Cq levels and PLR, NLR, D-dimer, CRP, and ESR in COVID-19 patients. Consequently, including the report of laboratory parameters alongside Cq values offers a promising prognosis.
Collapse
Affiliation(s)
- Hadi Safdari
- Department of Laboratory Sciences, School of Paramedical Sciences, Faculty of Paramedical and Rehabilitation SciencesMashhad University of Medical SciencesMashhadIran
| | - Saeede Bagheri
- Department of Laboratory Sciences, School of Paramedical Sciences, Faculty of Paramedical and Rehabilitation SciencesMashhad University of Medical SciencesMashhadIran
| | - Nasrin Talkhi
- Department of Biostatistics, School of Allied Medical SciencesShahid Beheshti University of Medical SciencesTehranIran
| | - Elahe Saberi Teymourian
- Department of Laboratory Sciences, School of Paramedical Sciences, Faculty of Paramedical and Rehabilitation SciencesMashhad University of Medical SciencesMashhadIran
| | - Mahdi Hosseini Bafghi
- Department of Laboratory Sciences, School of Paramedical Sciences, Faculty of Paramedical and Rehabilitation SciencesMashhad University of Medical SciencesMashhadIran
| | - Mohammad Hossein Ahmadi
- Department of Laboratory Sciences, School of Paramedical Sciences, Faculty of Paramedical and Rehabilitation SciencesMashhad University of Medical SciencesMashhadIran
| |
Collapse
|
12
|
Chen JY, Huang TR, Hsu SY, Huang CC, Wang HS, Chang JS. Effect and mechanism of quercetin or quercetin-containing formulas against COVID-19: From bench to bedside. Phytother Res 2024; 38:2597-2618. [PMID: 38479376 DOI: 10.1002/ptr.8175] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2023] [Revised: 02/13/2024] [Accepted: 02/14/2024] [Indexed: 06/13/2024]
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has caused the global coronavirus disease 2019 (COVID-19) pandemic since 2019. Immunopathogenesis and thromboembolic events are central to its pathogenesis. Quercetin exhibits several beneficial activities against COVID-19, including antiviral, anti-inflammatory, immunomodulatory, antioxidative, and antithrombotic effects. Although several reviews have been published, these reviews are incomplete from the viewpoint of translational medicine. The authors comprehensively evaluated the evidence of quercetin against COVID-19, both basically and clinically, to apply quercetin and/or its derivatives in the future. The authors searched the PubMed, Embase, and the Cochrane Library databases without any restrictions. The search terms included COVID-19, SARS-CoV-2, quercetin, antiviral, anti-inflammatory, immunomodulatory, thrombosis, embolism, oxidative, and microbiota. The references of relevant articles were also reviewed. All authors independently screened and reviewed the quality of each included manuscript. The Cochrane Risk of Bias Tool, version 2 (RoB 2) was used to assess the quality of the included randomized controlled trials (RCTs). All selected studies were discussed monthly. The effectiveness of quercetin against COVID-19 is not solid due to methodological flaws in the clinical trials. High-quality studies are also required for quercetin-containing traditional Chinese medicines. The low bioavailability and highly variable pharmacokinetics of quercetin hinder its clinical applications. Its positive impact on immunomodulation through reverting dysbiosis of gut microbiota still lacks robust evidence. Quercetin against COVID-19 does not have tough clinical evidence. Strategies to improve its bioavailability and/or to develop its effective derivatives are needed. Well-designed RCTs are also crucial to confirm their effectiveness in the future.
Collapse
Affiliation(s)
- Jhong Yuan Chen
- Department of Traditional Chinese Medicine, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Tsung Rung Huang
- Department of Traditional Chinese Medicine, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Shih Yun Hsu
- Department of Traditional Chinese Medicine, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Ching Chun Huang
- Department of Traditional Chinese Medicine, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Huei Syun Wang
- Department of Traditional Chinese Medicine, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Jung San Chang
- Division of Gastroenterology, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan
- Department of Medical Research, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan
- PhD Program in Toxicology, College of Pharmacy, Kaohsiung Medical University, Kaohsiung, Taiwan
| |
Collapse
|
13
|
Zhang Y, Chen S, Tian Y, Fu X. Host factors of SARS-CoV-2 in infection, pathogenesis, and long-term effects. Front Cell Infect Microbiol 2024; 14:1407261. [PMID: 38846354 PMCID: PMC11155306 DOI: 10.3389/fcimb.2024.1407261] [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: 03/26/2024] [Accepted: 05/08/2024] [Indexed: 06/09/2024] Open
Abstract
SARS-CoV-2 is the causative virus of the devastating COVID-19 pandemic that results in an unparalleled global health and economic crisis. Despite unprecedented scientific efforts and therapeutic interventions, the fight against COVID-19 continues as the rapid emergence of different SARS-CoV-2 variants of concern and the increasing challenge of long COVID-19, raising a vast demand to understand the pathomechanisms of COVID-19 and its long-term sequelae and develop therapeutic strategies beyond the virus per se. Notably, in addition to the virus itself, the replication cycle of SARS-CoV-2 and clinical severity of COVID-19 is also governed by host factors. In this review, we therefore comprehensively overview the replication cycle and pathogenesis of SARS-CoV-2 from the perspective of host factors and host-virus interactions. We sequentially outline the pathological implications of molecular interactions between host factors and SARS-CoV-2 in multi-organ and multi-system long COVID-19, and summarize current therapeutic strategies and agents targeting host factors for treating these diseases. This knowledge would be key for the identification of new pathophysiological aspects and mechanisms, and the development of actionable therapeutic targets and strategies for tackling COVID-19 and its sequelae.
Collapse
Affiliation(s)
| | | | - Yan Tian
- Department of Endocrinology and Metabolism, Department of Biotherapy, State Key Laboratory of Biotherapy and Cancer Center, West China Medical School, West China Hospital and Cancer Center, Sichuan University and Collaborative Innovation Center of Biotherapy, Sichuan, Chengdu, China
| | - Xianghui Fu
- Department of Endocrinology and Metabolism, Department of Biotherapy, State Key Laboratory of Biotherapy and Cancer Center, West China Medical School, West China Hospital and Cancer Center, Sichuan University and Collaborative Innovation Center of Biotherapy, Sichuan, Chengdu, China
| |
Collapse
|
14
|
Mohamed Hassan AS, Abo Gaziah SSA, Ezzelregal Awad HG, Hegab Abdelhady SM, Talaat Elkhafif NA, Hassan Mostafa NB. "Ultrastructural changes of platelets in COVID-19 and chronic viral hepatitis patients ". Ultrastruct Pathol 2024; 48:234-245. [PMID: 38619195 DOI: 10.1080/01913123.2024.2342437] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2024] [Accepted: 04/09/2024] [Indexed: 04/16/2024]
Abstract
Platelet-viral interactions are evolving as a new concern. Coagulation disorder is a major consequence of the COVID-19 infection. In chronic hepatitis virus infections, defect in coagulation factors, thrombocytopenia and platelet function abnormalities are common. A SARS-CoV-2 infection on top of chronic viral hepatitis infection can be common in areas where viral hepatitis is endemic. Here, we investigate the platelet ultrastructural changes and estimate the serum platelet factor-4 (PF-4), ferritin, CRP, and D-dimer in COVID-19 patients (n = 60), COVID-19 patients with associated chronic viral hepatitis (n = 20), and healthy subjects (n = 20). Ultrastructural changes were demonstrated in all test groups, denoting platelet activation. In chronic viral hepatitis patients, Platelet ultrastrustural apoptotic changes were also seen. Significantly high levels of PF-4 were confirmed in moderate and severe COVID-19 patients (P.value <0.001), with a cut off value of 17 ng/ml for predicting disease severity. A positive correlation of PF-4 with the level of serum ferritin, CRP, and D-dimer (p value < 0.001) was noted, while negatively correlated with platelet count and platelet granule count (p value < 0.001). In our study, chronic viral hepatitis patients presented mild COVID-19 signs, and their PF-4 level was comparable with the subgroup of mild COVID-19 infection. The platelet's critical role in COVID-19 coagulopathy and chronic viral hepatitis is evidenced by the ultrastructural changes and the high levels of PF4. Moreover, a dual viral infection poses a substantial burden on the platelets, necessitating close monitoring of the patient's coagulation profile.
Collapse
|
15
|
Ferreira G, Taylor A, Mensah SA. Deciphering the triad of endothelial glycocalyx, von Willebrand Factor, and P-selectin in inflammation-induced coagulation. Front Cell Dev Biol 2024; 12:1372355. [PMID: 38745860 PMCID: PMC11091309 DOI: 10.3389/fcell.2024.1372355] [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: 01/17/2024] [Accepted: 04/11/2024] [Indexed: 05/16/2024] Open
Abstract
This review examines the endothelial glycocalyx's role in inflammation and explores its involvement in coagulation. The glycocalyx, composed of proteins and glycosaminoglycans, interacts with von Willebrand Factor and could play a crucial role in anchoring it to the endothelium. In inflammatory conditions, glycocalyx degradation may leave P-selectin as the only attachment point for von Willebrand Factor, potentially leading to uncontrolled release of ultralong von Willebrand Factor in the bulk flow in a shear stress-dependent manner. Identifying specific glycocalyx glycosaminoglycan interactions with von Willebrand Factor and P-selectin can offer insights into unexplored coagulation mechanisms.
Collapse
Affiliation(s)
- Guinevere Ferreira
- Biomedical Engineering Department, Worcester Polytechnic Institute, Worcester, MA, United States
- Mechanical Engineering Department, Worcester Polytechnic Institute, Worcester, MA, United States
| | - Alexandra Taylor
- Biomedical Engineering Department, Worcester Polytechnic Institute, Worcester, MA, United States
| | - Solomon A. Mensah
- Biomedical Engineering Department, Worcester Polytechnic Institute, Worcester, MA, United States
- Mechanical Engineering Department, Worcester Polytechnic Institute, Worcester, MA, United States
| |
Collapse
|
16
|
Nana P, Dakis K, Spanos K, Tsolaki V, Karavidas N, Zakynthinos G, Kouvelos G, Giannoukas A, Matsagkas M. COVID-19 related peripheral arterial thrombotic events in intensive care unit and non-intensive care unit patients: A retrospective case series. Vascular 2024; 32:366-373. [PMID: 36395575 PMCID: PMC9679328 DOI: 10.1177/17085381221140159] [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] [Indexed: 08/27/2023]
Abstract
OBJECTIVES COVID-19 associated arterial thrombosis has been attributed to multiple inflammation and coagulation mechanisms. The aim of this study was to report the experience of a tertiary center on COVID-19 patients managed for acute peripheral arterial thrombosis. METHODS A single-center case series was conducted, including retrospectively collected data from all COVID-19 patients presenting arterial thrombosis, from March 2020 to February 2022. Intensive care unit (ICU) and non-ICU cases were included. The primary outcomes were mortality, successful revascularization, and amputation at 30 days. RESULTS Twenty patients presented peripheral arterial thrombosis (90% males, mean age 65 years (16-82 years)). Eighteen were diagnosed with the Delta variant and none was previously vaccinated. All cases presented acute lower limb ischemia; in 20% with bilateral involvement. Nine patients were hospitalized in the ward while 11 in the ICU. From the non-ICU cases, five presented Rutherford IIb and four cases, Rutherford's IIa ischemia. Six cases underwent revascularization (67%). Two of them were finally amputated (33%) and two died during hospitalization (33%). Two revascularizations were considered successful (33%). The ICU group presented mainly with Rutherford's III ischemia (54.5%). The mortality in the ICU cohort was 72.7%. Only one patient underwent successful revascularization and two were amputated in this subgroup. Early mortality was 50% for the total cohort while the type of management was not related to mortality. CONCLUSIONS Covid-19 related arterial thrombosis in non-vaccinated population is associated with 50% early mortality; increased up to 72% in the ICU patients. The amputation rate was 20% while only 40% of the revascularizations were considered successful.
Collapse
Affiliation(s)
- Petroula Nana
- Department of Vascular Surgery, University Hospital of Larissa, Faculty of Medicine, School of Health Sciences, University of Thessaly, Larissa, Greece
| | - Konstantinos Dakis
- Department of Vascular Surgery, University Hospital of Larissa, Faculty of Medicine, School of Health Sciences, University of Thessaly, Larissa, Greece
| | - Konstantinos Spanos
- Department of Vascular Surgery, University Hospital of Larissa, Faculty of Medicine, School of Health Sciences, University of Thessaly, Larissa, Greece
| | - Vasiliki Tsolaki
- Intensive Care Unit, University Hospital of Larissa, Faculty of Medicine, School of Health Sciences, University of Thessaly, Larissa, Greece
| | - Nikitas Karavidas
- Intensive Care Unit, University Hospital of Larissa, Faculty of Medicine, School of Health Sciences, University of Thessaly, Larissa, Greece
| | - George Zakynthinos
- Intensive Care Unit, University Hospital of Larissa, Faculty of Medicine, School of Health Sciences, University of Thessaly, Larissa, Greece
| | - George Kouvelos
- Department of Vascular Surgery, University Hospital of Larissa, Faculty of Medicine, School of Health Sciences, University of Thessaly, Larissa, Greece
| | - Athanasios Giannoukas
- Department of Vascular Surgery, University Hospital of Larissa, Faculty of Medicine, School of Health Sciences, University of Thessaly, Larissa, Greece
| | - Miltiadis Matsagkas
- Department of Vascular Surgery, University Hospital of Larissa, Faculty of Medicine, School of Health Sciences, University of Thessaly, Larissa, Greece
| |
Collapse
|
17
|
Xi Y, Mao Y, Zhu W, Xi P, Huang F, Tan H, Liao X, Zhou L. IL-6 is a predictor and potential therapeutic target for coronavirus disease 2019-related heart failure: A single-center retrospective study. Cytokine 2024; 176:156514. [PMID: 38277928 DOI: 10.1016/j.cyto.2024.156514] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2023] [Revised: 01/13/2024] [Accepted: 01/17/2024] [Indexed: 01/28/2024]
Abstract
BACKGROUND Inflammation is linked to coronavirus disease 2019 (COVID-19)-related heart failure (HF), but the specific mechanisms are unclear. This study aimed to assess the relationship between specific inflammatory factors, such as interleukin (IL)-1β, IL-2, IL-4, IL-5, IL-6, IL-8, IL-10, IL-12, IL-17, interferon (IFN)-α, and IFN-γ, and COVID-19-related HF. METHODS We retrospectively identified 212 adult patients with COVID-19 who were hospitalized at Shanghai Public Health Center from March 1 to May 30, 2022 (including 80 patients with HF and 132 without HF). High-sensitivity C-reactive protein (hs-CRP), procalcitonin (PCT), and inflammatory factors, including IL-1β, IL-2, IL-4, IL-5, IL-6, IL-8, IL-10, IL-12, IL-17, IFN-α, and IFN-γ, were compared between patients with COVID-19 with and without HF. RESULTS Patients with COVID-19 having and not having HF differed with regard to sex, age, hs-CRP, PCT, and IL-6 levels (p < 0.05). Logistic regression analysis indicated a significant positive association between IL and 6 and HF (odds ratio = 1.055; 95 % confidence interval: 1.019-1.093, p < 0.005). Sex, age, and hs-CRP were also associated with HF. Women had a greater risk of HF than men. Older age, higher levels of hs-CRP, and IL-6 were associated with a greater risk of HF. CONCLUSIONS In patients with COVID-19, increased IL-6 levels are significantly associated with COVID-19-related HF.
Collapse
Affiliation(s)
- Yan Xi
- Department of General Practice, Tongji Hospital, School of Medicine, Tongji University, Shanghai 20065, China
| | - Yu Mao
- Department of Cardiology, Tongji Hospital, Tongji University, Shanghai 20065, China
| | - Wei Zhu
- General Hospital of Eastern Theater Command, Nanjing City, Jiangsu Province 210000, China
| | - Peng Xi
- Department of Cardiology, Tongji Hospital, Tongji University, Shanghai 20065, China
| | - Feifei Huang
- Department of Cardiology, Tongji Hospital, Tongji University, Shanghai 20065, China
| | - Hongwei Tan
- Department of Cardiology, Tongji Hospital, Tongji University, Shanghai 20065, China
| | - Xudong Liao
- Department of Cardiology, Tongji Hospital, Tongji University, Shanghai 20065, China
| | - Lin Zhou
- Department of Cardiology, Tongji Hospital, Tongji University, Shanghai 20065, China.
| |
Collapse
|
18
|
Heo KY, Bonsu JM, Muffly BT, Rieger E, Song J, Ayeni AM, Guild GN, Premkumar A. Complications Rates Among Revision Total Knee Arthroplasty Patients Diagnosed With COVID-19 Postoperatively. J Arthroplasty 2024; 39:766-771.e2. [PMID: 37757979 DOI: 10.1016/j.arth.2023.09.028] [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: 06/06/2023] [Revised: 09/13/2023] [Accepted: 09/16/2023] [Indexed: 09/29/2023] Open
Abstract
BACKGROUND The COVID-19 pandemic introduced a new set of challenges for the arthroplasty community, including the management of patients diagnosed with COVID-19 following revision total knee arthroplasty (rTKA) and its potential impact on postoperative recovery. This study sought to characterize the risks of postoperative COVID-19 infection among rTKA patients. METHODS A large national database was utilized to query 8,022 total patients who underwent rTKA between 2018 and 2021, of which 60 had a COVID diagnosis within 90 days after surgery (rTKA/COVID positive). These patients were 1:10 propensity-score matched to 600 rTKA patients who did not have a 90-day postoperative COVID diagnosis (rTKA/COVID negative) and 600 COVID positive patients who did not undergo rTKA. Controlling for potential confounders, multivariate logistic regressions were utilized to compare 90-day postoperative complications between groups. RESULTS Compared to rTKA/COVID negativepatients, the rTKA/COVID positive cohort had significantly higher rates of pneumonia (odds ratio [OR] = 6.1, P < .001), pulmonary embolism (PE) (OR = 32.4, P < .001), deep venous thrombosis (DVT) (OR = 32.4, P < .001), and 90-day readmissions (OR = 2.1, P = .02). Similarly, the rTKA/COVID positive cohort had significantly higher rates of pneumonia (OR = 4.3, P = .001), PE (OR = 36.8, P < .001), and DVT (OR = 36.8, P < .001) compared to COVID positive patients who did not undergo rTKA. CONCLUSIONS Revision total knee arthroplasty patients diagnosed with COVID-19 postoperatively had increased rates of thromboembolic events, pneumoniae, and 90-day readmissions. Risk mitigation efforts would suggest extending the prophylactic anticoagulation period for rTKA patients diagnosed with postoperative COVID-19.
Collapse
Affiliation(s)
- Kevin Y Heo
- Department of Orthopaedic Surgery, Emory University School of Medicine, Atlanta, Georgia
| | - Janice M Bonsu
- Department of Orthopaedic Surgery, Emory University School of Medicine, Atlanta, Georgia
| | - Brian T Muffly
- Department of Orthopaedic Surgery, Emory University School of Medicine, Atlanta, Georgia
| | - Elizabeth Rieger
- Department of Orthopaedic Surgery, Emory University School of Medicine, Atlanta, Georgia
| | - Joseph Song
- Department of Orthopaedic Surgery, Emory University School of Medicine, Atlanta, Georgia
| | - Ayomide M Ayeni
- Department of Orthopaedic Surgery, Emory University School of Medicine, Atlanta, Georgia
| | - George N Guild
- Department of Orthopaedic Surgery, Emory University School of Medicine, Atlanta, Georgia
| | - Ajay Premkumar
- Department of Orthopaedic Surgery, Emory University School of Medicine, Atlanta, Georgia
| |
Collapse
|
19
|
Wang Q, Qin Y, Ma J, Zhou K, Xia G, Li Y, Xie L, Afful RG, Lan Q, Huo X, Zou J, Yang H. An early warning indicator of mortality risk in patients with COVID-19: the neutrophil extracellular traps/neutrophilic segmented granulocyte ratio. Front Immunol 2024; 15:1287132. [PMID: 38348024 PMCID: PMC10859410 DOI: 10.3389/fimmu.2024.1287132] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2023] [Accepted: 01/15/2024] [Indexed: 02/15/2024] Open
Abstract
Background Neutrophil extracellular traps (NETs) play a key role in thrombus formation in patients with coronavirus disease 2019 (COVID-19). However, the existing detection and observation methods for NETs are limited in their ability to provide quantitative, convenient, and accurate descriptions of in situ NETs. Therefore, establishing a quantitative description of the relationship between NETs and thrombosis remains a challenge. Objective We employed morphological observations of blood cells and statistical analyses to investigate the correlation between the NETs/neutrophilic segmented granulocyte ratio and mortality risk in patients with COVID-19. Methods Peripheral blood samples were collected from 117 hospitalized patients with COVID-19 between November 2022 and February 2023, and various blood cell parameters were measured. Two types of smudge cells were observed in the blood and counted: lymphatic and neutral smudge cells. Statistical data analysis was used to establish COVID-19 mortality risk assessment indicators. Results Morphological observations of neutrophilic smudge cells revealed swelling, eruption, and NETs formation in the neutrophil nuclei. Subsequently, the NETs/neutrophilic segmented granulocyte ratio (NNSR) was calculated. A high concentration of NETs poses a fatal risk for thrombus formation in patients. Statistical analysis indicated that a high NNSR was more suitable for evaluating the risk of death in patients with COVID-19 compared to elevated fibrinogen (FIB) and D-dimer (DD) levels. Conclusion Observing blood cell morphology is an effective method for the detection of NETs, NNSR are important markers for revealing the mortality risk of patients with COVID-19.
Collapse
Affiliation(s)
- Qiong Wang
- The Affiliated Wuxi People’s Hospital of Nanjing Medical University, Wuxi People’s Hospital, Wuxi Medical Center, Nanjing Medical University, Wuxi, China
| | - Yu Qin
- Key Laboratory of Industrial Biotechnology, Ministry of Education, School of Biotechnology, Jiangnan University, Wuxi, China
| | - Jingyun Ma
- The Affiliated Wuxi People’s Hospital of Nanjing Medical University, Wuxi People’s Hospital, Wuxi Medical Center, Nanjing Medical University, Wuxi, China
| | - Kehao Zhou
- Key Laboratory of Industrial Biotechnology, Ministry of Education, School of Biotechnology, Jiangnan University, Wuxi, China
| | - Guiping Xia
- The Affiliated Wuxi People’s Hospital of Nanjing Medical University, Wuxi People’s Hospital, Wuxi Medical Center, Nanjing Medical University, Wuxi, China
| | - Ya Li
- Key Laboratory of Industrial Biotechnology, Ministry of Education, School of Biotechnology, Jiangnan University, Wuxi, China
| | - Li Xie
- School of Internet of Things Engineering, Jiangnan University, Wuxi, China
| | - Richmond Godwin Afful
- Key Laboratory of Industrial Biotechnology, Ministry of Education, School of Biotechnology, Jiangnan University, Wuxi, China
| | - Qian Lan
- School of Internet of Things Engineering, Jiangnan University, Wuxi, China
| | - Xingyu Huo
- Key Laboratory of Industrial Biotechnology, Ministry of Education, School of Biotechnology, Jiangnan University, Wuxi, China
| | - Jian Zou
- The Affiliated Wuxi People’s Hospital of Nanjing Medical University, Wuxi People’s Hospital, Wuxi Medical Center, Nanjing Medical University, Wuxi, China
| | - Hailin Yang
- Key Laboratory of Industrial Biotechnology, Ministry of Education, School of Biotechnology, Jiangnan University, Wuxi, China
| |
Collapse
|
20
|
Josuttis D, Schwedler C, Heymann G, Gümbel D, Schmittner MD, Kruse M, Hoppe B. Vascular Endothelial Growth Factor as Potential Biomarker for COVID-19 Severity. J Intensive Care Med 2023; 38:1165-1173. [PMID: 37448220 PMCID: PMC10345830 DOI: 10.1177/08850666231186787] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Revised: 06/20/2023] [Accepted: 06/26/2023] [Indexed: 07/15/2023]
Abstract
INTRODUCTION COVID-19 is characterized by immunological responses to viral replication and coherent with endothelitis, microvascular disturbance of lung vasculature and coagulopathy. Vascular Endothelial Growth Factor (VEGF) is a proangiogenic mediator regulating endothelial changes. It is induced by proinflammatory signaling and hypoxia. We sought to determine whether VEGF levels differ between SARS-CoV-2-positive patients of different disease severity and whether VEGF might be useful in risk stratification. METHODS After retrospective screening of all SARS-CoV-2-positive patients treated in Unfallkrankenhaus Berlin in 2020, we included those with documented VEGF measurement. We extracted laboratory values and clinical parameters. An exploratory data analysis was performed to detect possible relations between VEGF level and clinical disease features. RESULTS We included 167 SARS-CoV-2-positive patients of which 139 suffered from COVID-19. Seventy-one of the COVID-19 patients had to be treated in the intensive care unit (ICU), those patients exhibited higher VEGF levels than those being admitted to normal wards (535 vs 279 pg/L, P < .001). APACHE-2 (Acute Physiology And Chronic Health Evaluation Score) correlated with mortality and patients with high values showed higher VEGF concentrations on admission (456 vs 875 pg/L, p = 0.006). Receiver operating characteristic analytic revealed that the occurrence of organ dysfunctions like acute respiratory distress syndrome (ARDS), shock, or acute kidney injury could be predicted by VEGF. It was significantly higher in patients who later died compared to survivors (637 vs 389 pg/mL, P = 0.041) and predicted mortality with same accuracy as established markers. In our cohort, association of VEGF above 277 pg/L on admission with risk of ARDS could be confirmed in logistic regression adjusting for possible confounding factors (odds ratio 3.1, 95% confidence interval: 1.34-7.7). DISCUSSION Even though there are several limitations to this retrospective study it revealed that in COVID-19 patients VEGF can contribute to the prediction of necessity of ICU, mortality and the prediction of ARDS, kidney injury or shock. Its use in risk stratification and potential pathogenetic involvement should be further investigated.
Collapse
Affiliation(s)
- David Josuttis
- Department of Anesthesiology, Intensive Care and Pain Medicine, BG-Klinikum Unfallkrankenhaus Berlin, Berlin, Germany
| | | | - Guido Heymann
- Department of Laboratory Medicine, BG-Klinikum Unfallkrankenhaus Berlin, Berlin, Germany
| | - Denis Gümbel
- Department of Trauma, Reconstructive Surgery and Rehabilitation Medicine, University Medicine Greifswald, Greifswald, Germany
- Department of Trauma and Orthopaedic Surgery, BG-Klinikum Unfallkrankenhaus Berlin, Berlin, Germany
| | - Marc Dominik Schmittner
- Department of Anesthesiology, Intensive Care and Pain Medicine, BG-Klinikum Unfallkrankenhaus Berlin, Berlin, Germany
| | - Marianne Kruse
- Department of Anesthesiology, Intensive Care and Pain Medicine, BG-Klinikum Unfallkrankenhaus Berlin, Berlin, Germany
| | - Berthold Hoppe
- Health and Medical University Potsdam, Potsdam, Germany
- Department of Laboratory Medicine, BG-Klinikum Unfallkrankenhaus Berlin, Berlin, Germany
| |
Collapse
|
21
|
Li LZ, Zhou C, Wang P, Ke QH, Zhang J, Lei SS, Li ZQ. Molecular mechanism of the effect of Gegen Qinlian decoction on COVID-19 comorbid with diabetes mellitus based on network pharmacology and molecular docking: A review. Medicine (Baltimore) 2023; 102:e34683. [PMID: 37933071 PMCID: PMC10627614 DOI: 10.1097/md.0000000000034683] [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: 05/13/2023] [Accepted: 07/20/2023] [Indexed: 11/08/2023] Open
Abstract
To explore the potential mechanism of Gegen Qinlian decoction (GGQL) in the treatment of COVID-19 comorbid with diabetes mellitus (DM) through network pharmacology and molecular docking, and to provide theoretical guidance for clinical transformation research. Traditional Chinese Medicine Systems Pharmacology Database and Analysis Platform was used to screen the active compounds and targets of GGQL, the targets of COVID-19 comorbid with DM were searched based on Genecards database. Protein-protein interaction network was constructed using String data platform for the intersection of compounds and disease targets, the Gene Ontology and Kyoto Encyclopedia of Genes and Genomes analysis of the intersection targets was performed using DAVID database. Cytoscape software was used to construct the "compound target-pathway (C-T-P)" of GGQL in the treatment of COVID-19 comorbid with DM, the molecular docking platform was used to complete the simulated docking of key compounds and targets. We obtained 141 compounds from GGQL, revealed 127 bioactive compounds and 283 potential targets of GGQL. Quercetin, kaempferol and formononetin in GGQL play a role by modulating the targets (including AR, GSK3B, DPP4, F2, and NOS3). GGQL might affect diverse signaling pathways related to the pathogenesis of coronavirus disease - COVID-19, AGE-RAGE signaling pathway in diabetic complications, IL-17 signaling pathway, human cytomegalovirus infection and Th17 cell differentiation. Meanwhile, molecular docking showed that the selected GGQL core active components had strong binding activity with the key targets. This study revealed that GGQL play a role in the treatment of COVID-19 comorbid with DM through multi-component, multi-target and multi-pathway mode of action, which provided good theoretical basis for further verification research.
Collapse
Affiliation(s)
- Lin-zi Li
- Jingmen Central Hospital, Jingmen, China
| | - Cong Zhou
- AnKang University, School of Medicine, AnKang, China
| | - Pei Wang
- Jingmen Central Hospital, Jingmen, China
| | | | - Jie Zhang
- Jingmen Central Hospital, Jingmen, China
| | - Shan-shan Lei
- Department of Medicine, Zhejiang Academy of Traditional Chinese Medicine, Hangzhou, China
| | | |
Collapse
|
22
|
Fallahi P, Elia G, Ragusa F, Paparo SR, Patrizio A, Balestri E, Mazzi V, Benvenga S, Varricchi G, Gragnani L, Botrini C, Baldini E, Centanni M, Ferri C, Antonelli A, Ferrari SM. Thyroid Autoimmunity and SARS-CoV-2 Infection. J Clin Med 2023; 12:6365. [PMID: 37835009 PMCID: PMC10573843 DOI: 10.3390/jcm12196365] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2023] [Revised: 10/01/2023] [Accepted: 10/03/2023] [Indexed: 10/15/2023] Open
Abstract
The severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), the etiological culprit of COronaVIrus Disease 19 (COVID-19), can enter the cells via the angiotensin-converting enzyme 2 (ACE2) receptor, which has been found in several tissues including in endocrine organs, such as the ovaries, testes, pancreas, and thyroid. Several thyroid disorders have been associated with SARS-CoV-2 infection [subacute thyroiditis (SAT), thyrotoxicosis, and non-thyroidal illness syndrome (NTIS)] and, in part, they are believed to be secondary to the local virus replication within the gland cells. However, as documented for other viruses, SARS-CoV-2 seems to interfere with several aspects of the immune system, inducing the synthesis of autoantibodies and triggering latent or new onset autoimmune disease (AID), including autoimmune thyroid disease (AITD), such as Hashimoto Thyroiditis (HT) and Graves' disease (GD). Several mechanisms have been hypothesized to explain this induction of autoimmunity by SARS-CoV-2 infection: the immune system hyper-stimulation, the molecular mimicry between the self-antigens of the host and the virus, neutrophils extracellular traps, and finally, the virus induced transcriptional changes in the immune genes; nonetheless, more evidence is needed especially from large, long-term cohort studies involving COVID-19 patients, to establish or reject this pathogenetic relationship.
Collapse
Affiliation(s)
- Poupak Fallahi
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, 56126 Pisa, Italy; (P.F.); (S.R.P.); (L.G.)
| | - Giusy Elia
- Department of Surgery, Medical and Molecular Pathology and of Critical Area, University of Pisa, 56126 Pisa, Italy; (G.E.); (F.R.); (E.B.); (V.M.); (C.B.)
| | - Francesca Ragusa
- Department of Surgery, Medical and Molecular Pathology and of Critical Area, University of Pisa, 56126 Pisa, Italy; (G.E.); (F.R.); (E.B.); (V.M.); (C.B.)
| | - Sabrina Rosaria Paparo
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, 56126 Pisa, Italy; (P.F.); (S.R.P.); (L.G.)
| | - Armando Patrizio
- Department of Emergency Medicine, Azienda Ospedaliero-Universitaria Pisana, 56126 Pisa, Italy;
| | - Eugenia Balestri
- Department of Surgery, Medical and Molecular Pathology and of Critical Area, University of Pisa, 56126 Pisa, Italy; (G.E.); (F.R.); (E.B.); (V.M.); (C.B.)
| | - Valeria Mazzi
- Department of Surgery, Medical and Molecular Pathology and of Critical Area, University of Pisa, 56126 Pisa, Italy; (G.E.); (F.R.); (E.B.); (V.M.); (C.B.)
| | - Salvatore Benvenga
- Department of Clinical and Experimental Medicine—Endocrinology, University of Messina, 98122 Messina, Italy;
- Master Program on Childhood, Adolescent and Women’s Endocrine Health, University of Messina, 98122 Messina, Italy
- Interdepartmental Program of Molecular & Clinical Endocrinology and Women’s Endocrine Health, University Hospital Policlinico “G. Martino”, 98124 Messina, Italy
| | - Gilda Varricchi
- Department of Translational Medical Sciences, University of Naples Federico II, 80131 Naples, Italy;
- Center for Basic and Clinical Immunology Research, University of Naples Federico II, 80131 Naples, Italy
- World Allergy Organization Center of Excellence, University of Naples Federico II, 80131 Naples, Italy
- Institute of Experimental Endocrinology and Oncology “Gaetano Salvatore”, National Research Council, 80131 Naples, Italy
| | - Laura Gragnani
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, 56126 Pisa, Italy; (P.F.); (S.R.P.); (L.G.)
| | - Chiara Botrini
- Department of Surgery, Medical and Molecular Pathology and of Critical Area, University of Pisa, 56126 Pisa, Italy; (G.E.); (F.R.); (E.B.); (V.M.); (C.B.)
| | - Enke Baldini
- Department of Experimental Medicine, “Sapienza” University of Rome, 00185 Rome, Italy;
| | - Marco Centanni
- Department of Medico-Surgical Sciences and Biotechnologies, Endocrinology Section, ‘‘Sapienza’’ University of Rome, 00185 Rome, Italy;
- Endocrine Unit, Azienda Unità Sanitaria Locale (AUSL) Latina, 04100 Latina, Italy
| | - Clodoveo Ferri
- Rheumatology Unit, School of Medicine, University of Modena and Reggio Emilia, 41100 Modena, Italy;
- Rheumatology Clinic ‘Madonna Dello Scoglio’ Cotronei, 88836 Crotone, Italy
| | - Alessandro Antonelli
- Department of Surgery, Medical and Molecular Pathology and of Critical Area, University of Pisa, 56126 Pisa, Italy; (G.E.); (F.R.); (E.B.); (V.M.); (C.B.)
| | - Silvia Martina Ferrari
- Department of Clinical and Experimental Medicine, University of Pisa, 56126 Pisa, Italy;
| |
Collapse
|
23
|
Dutsch A, Uhlig C, Bock M, Graesser C, Schuchardt S, Uhlig S, Schunkert H, Joner M, Holdenrieder S, Lechner K. Multi-Omic Candidate Screening for Markers of Severe Clinical Courses of COVID-19. J Clin Med 2023; 12:6225. [PMID: 37834869 PMCID: PMC10573369 DOI: 10.3390/jcm12196225] [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: 08/18/2023] [Revised: 09/17/2023] [Accepted: 09/22/2023] [Indexed: 10/15/2023] Open
Abstract
BACKGROUND Severe coronavirus disease 2019 (COVID-19) disease courses are characterized by immuno-inflammatory, thrombotic, and parenchymal alterations. Prediction of individual COVID-19 disease courses to guide targeted prevention remains challenging. We hypothesized that a distinct serologic signature precedes surges of IL-6/D-dimers in severely affected COVID-19 patients. METHODS We performed longitudinal plasma profiling, including proteome, metabolome, and routine biochemistry, on seven seropositive, well-phenotyped patients with severe COVID-19 referred to the Intensive Care Unit at the German Heart Center. Patient characteristics were: 65 ± 8 years, 29% female, median CRP 285 ± 127 mg/dL, IL-6 367 ± 231 ng/L, D-dimers 7 ± 10 mg/L, and NT-proBNP 2616 ± 3465 ng/L. RESULTS Based on time-series analyses of patient sera, a prediction model employing feature selection and dimensionality reduction through least absolute shrinkage and selection operator (LASSO) revealed a number of candidate proteins preceding hyperinflammatory immune response (denoted ΔIL-6) and COVID-19 coagulopathy (denoted ΔD-dimers) by 24-48 h. These candidates are involved in biological pathways such as oxidative stress/inflammation (e.g., IL-1alpha, IL-13, MMP9, C-C motif chemokine 23), coagulation/thrombosis/immunoadhesion (e.g., P- and E-selectin), tissue repair (e.g., hepatocyte growth factor), and growth factor response/regulatory pathways (e.g., tyrosine-protein kinase receptor UFO and low-density lipoprotein receptor (LDLR)). The latter are host- or co-receptors that promote SARS-CoV-2 entry into cells in the absence of ACE2. CONCLUSIONS Our novel prediction model identified biological and regulatory candidate networks preceding hyperinflammation and coagulopathy, with the most promising group being the proteins that explain changes in D-dimers. These biomarkers need validation. If causal, our work may help predict disease courses and guide personalized treatment for COVID-19.
Collapse
Affiliation(s)
- Alexander Dutsch
- Department of Cardiology, German Heart Centre Munich, Technical University of Munich, Lazarettstraße 36, 80636 Munich, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Munich, Munich Heart Alliance, 80336 Munich, Germany
| | - Carsten Uhlig
- Institute for Laboratory Medicine, German Heart Centre Munich, Technical University of Munich, Lazarettstraße 36, 80636 Munich, Germany
| | - Matthias Bock
- Department of Cardiology, German Heart Centre Munich, Technical University of Munich, Lazarettstraße 36, 80636 Munich, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Munich, Munich Heart Alliance, 80336 Munich, Germany
| | - Christian Graesser
- Department of Cardiology, German Heart Centre Munich, Technical University of Munich, Lazarettstraße 36, 80636 Munich, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Munich, Munich Heart Alliance, 80336 Munich, Germany
| | - Sven Schuchardt
- Fraunhofer Institute for Toxicology and Experimental Medicine ITEM, 30625 Hannover, Germany
| | - Steffen Uhlig
- QuoData Gesellschaft für Qualitätsmanagement und Statistik, Fabeckstr. 43, 14195 Berlin, Germany
| | - Heribert Schunkert
- Department of Cardiology, German Heart Centre Munich, Technical University of Munich, Lazarettstraße 36, 80636 Munich, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Munich, Munich Heart Alliance, 80336 Munich, Germany
| | - Michael Joner
- Department of Cardiology, German Heart Centre Munich, Technical University of Munich, Lazarettstraße 36, 80636 Munich, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Munich, Munich Heart Alliance, 80336 Munich, Germany
| | - Stefan Holdenrieder
- Institute for Laboratory Medicine, German Heart Centre Munich, Technical University of Munich, Lazarettstraße 36, 80636 Munich, Germany
| | - Katharina Lechner
- Department of Cardiology, German Heart Centre Munich, Technical University of Munich, Lazarettstraße 36, 80636 Munich, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Munich, Munich Heart Alliance, 80336 Munich, Germany
| |
Collapse
|
24
|
Cai X, Deng J, Shi W, Cai Y, Ma Z. Mining the potential therapeutic targets for COVID-19 infection in patients with severe burn injuries via bioinformatics analysis. Int Wound J 2023; 20:2742-2752. [PMID: 36924127 PMCID: PMC10410338 DOI: 10.1111/iwj.14151] [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: 12/27/2022] [Revised: 02/24/2023] [Accepted: 02/27/2023] [Indexed: 03/18/2023] Open
Abstract
The Coronavirus Disease-19 (COVID-19) pandemic is posing a serious challenge to human health. Burn victims are susceptible to severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection leading to delayed recovery and even profound debilitation. Nevertheless, the molecular mechanisms underlying COVID-19 and severe burn are yet to be elucidated. In our work, the differentially expressed genes (DEGs) were identified from GSE157852 and GSE19743, and the common DEGs between COVID-19 and severe burn were extracted. Gene Ontology (GO), Kyoto Encyclopedia of Genes and Genomes (KEGG), protein-protein interactions (PPI), gene coexpression network, and multifactor regulatory network analysis of hub genes were carried out. A total of 44 common DEGs were found between COVID-19 and severe burn. Functional analyses indicated that the pathways of immune regulation and cytokine response participated collectively in the development of severe burn and progression of COVID-19. Ten significant hub genes were identified, including MERTK, SIRPA, TLR3, ITGB1, DPP4, PTPRC, LY75, IFIT1, IL4R, and CD2. In addition, the gene coexpression network and regulatory network were constructed containing 42 microRNAs (miRNAs) and 2 transcription factors (TFs). Our study showed the shared pathogenic link between COVID-19 and severe burn. The identified common genes and pivotal pathways pave a new road for future mechanistic researches in severe burn injuries complicated with COVID-19.
Collapse
Affiliation(s)
- Xueyao Cai
- Department of Burn and Plastic SurgeryDongguan Tungwah HospitalDongguanChina
| | - Jing Deng
- Department of Burn and Plastic SurgeryDongguan Tungwah HospitalDongguanChina
| | - Wenjun Shi
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Yuchen Cai
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Zhengzheng Ma
- Department of Burn and Plastic SurgeryDongguan Tungwah HospitalDongguanChina
| |
Collapse
|
25
|
Ali A, Liang W, Abdelhafiz AS, Saleh MM, Salem H, Moazen EM, Elmazny MI, Rakha MA, Elfeky SEF. Elevation of D-dimer levels are associated with early need for mechanical ventilation support in patients with COVID-19. BMC Pulm Med 2023; 23:283. [PMID: 37537520 PMCID: PMC10401731 DOI: 10.1186/s12890-023-02551-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Accepted: 07/05/2023] [Indexed: 08/05/2023] Open
Abstract
BACKGROUND Severe COVID-19 disease is typically associated with an urgent need for supplemental oxygen therapy that may be successfully delivered through conventional methods or require invasive mechanical ventilation. Early prediction of the need for invasive mechanical ventilation could significantly improve outcomes of COVID-19 patients. Plasma levels of D-dimer and a number of inflammatory markers as well as values of complete blood counts, all measured in the first two days of hospital admission of COVID-19 patients, were evaluated for their significance as predictors of the eventual need for invasive mechanical ventilation support as well as their values as predictors of post-ventilation morbidly and mortality. METHODS This retrospective cohort study was conducted at a single center and included data pertaining to 200 patients with previously confirmed moderate to severe COVID-19 disease in the period between May 2021 and the end of December 2022. Data were retrieved from medical records for further analysis. RESULTS The mean (SD) age of patients stood at 59 (14) years of age, and with a majority of patients being male (77%). About 18% of cases, all of significantly older age, had been connected to invasive mechanical ventilation (IMV). Total leucocytic count (TLC), as well as levels of urea, creatinine, D-dimer, ferritin, and CRP in IMV patients were significantly higher than non-ventilated patients (p < 0.01 for all). In contrast, lymphocytic count, hemoglobin level, and platelet count were significantly lower in IMV patients (p < 0.001, 0.04, and 0.002, respectively). The mortality rate was significantly higher in IMV patients (p < 0.001). D-dimer independently predicted IMV demand (OR = 1, p = 0.001 in adjusted and unadjusted models). The utility of D-dimer was excellent; and the cutoff level of above 1415 µ/L showed sensitivity and specificity of about 92% and 76%, respectively. Also, the D-dimer level was very effective in predicting post-IMV survival; the AUC = 0.86, p = 0.02, and a cutoff value below 4558 µ/L was associated with 100% and 66% sensitivity and specificity, respectively. CONCLUSIONS High D-dimer levels independently correlated with the need for invasive mechanical ventilation. Low levels of this marker could evidently predict post-IMV survival of mechanically ventilated COVID-19 patients. Measuring D-dimer levels during routine follow up of those patients would thus be useful in predicting patient outcomes.
Collapse
Affiliation(s)
- Asmaa Ali
- Department of laboratory medicine, School of Medicine, Jiangsu University, Zhenjiang, 212013, China
- Department of pulmonary medicine, Abbassia Chest Hospital, MOH, Cairo, Egypt
- Department of respiratory allergy, Al-Rashed Allergy Centre, MOH, Kuwait, Kuwait
| | - Wu Liang
- Department of laboratory medicine, School of Medicine, Jiangsu University, Zhenjiang, 212013, China
| | - Ahmed Samir Abdelhafiz
- Department of Clinical Pathology, National Cancer Institute, Cairo University, Kasr Al-Aini Street, from El-Khalig Square, Cairo, 11796, Egypt.
| | - Mai M Saleh
- Department of chest disease, Faculty of Medicine, Tanta University, Tanta, Egypt
| | - Heba Salem
- Department of chest disease, Faculty of Medicine, Tanta University, Tanta, Egypt
| | - Eman M Moazen
- Department of chest disease, Faculty of Medicine, Al-Azhar University, Cairo, Egypt
| | - Maram I Elmazny
- Department of anesthesia, intensive care and pain, Faculty of Medicine, Tanta University, Tanta, Egypt
| | | | | |
Collapse
|
26
|
Li J, Zhang K, Zhang Y, Gu Z, Huang C. Neutrophils in COVID-19: recent insights and advances. Virol J 2023; 20:169. [PMID: 37533131 PMCID: PMC10398943 DOI: 10.1186/s12985-023-02116-w] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2023] [Accepted: 07/06/2023] [Indexed: 08/04/2023] Open
Abstract
Coronavirus disease 2019 (COVID-19) is an acute respiratory disease caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), which can lead to acute respiratory distress syndrome (ARDS), multi-organ failure and death, posing significant threat to human health. Studies have found that pathological mechanisms, such as cytokine storms caused by uncontrolled innate immune system activation, release of damage-associated molecular patterns during tissue injury and a high incidence of thrombotic events, are associated with the function and dysfunction of neutrophils. Specifically, the increased formation of low-density neutrophils (LDNs) and neutrophil extracellular traps (NETs) has been shown to be closely linked with the severity and poor prognosis in patients with COVID-19. Our work focuses on understanding the increased number, abnormal activation, lung tissue infiltration, and elevated neutrophil-to-lymphocyte ratio in the pathogenesis of COVID-19. We also explore the involvement of NETs and LDNs in disease progression and thrombosis formation, along with potential therapeutic strategies targeting neutrophil and NETs formation.
Collapse
Affiliation(s)
- Jiayu Li
- Department of Infectious Diseases, Second Affiliated Hospital of Air Force Military Medical University, Xi'an, 710038, China
| | - Kegong Zhang
- Department of Infectious Diseases, Second Affiliated Hospital of Air Force Military Medical University, Xi'an, 710038, China
| | - Ye Zhang
- Department of Infectious Diseases, Second Affiliated Hospital of Air Force Military Medical University, Xi'an, 710038, China
| | - Ziyang Gu
- Department of Infectious Diseases, Second Affiliated Hospital of Air Force Military Medical University, Xi'an, 710038, China
| | - Changxing Huang
- Department of Infectious Diseases, Second Affiliated Hospital of Air Force Military Medical University, Xi'an, 710038, China.
| |
Collapse
|
27
|
Li S, Wang H, Shao Q. The central role of neutrophil extracellular traps (NETs) and by-products in COVID-19 related pulmonary thrombosis. Immun Inflamm Dis 2023; 11:e949. [PMID: 37647446 PMCID: PMC10461423 DOI: 10.1002/iid3.949] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2023] [Revised: 06/26/2023] [Accepted: 07/08/2023] [Indexed: 09/01/2023] Open
Abstract
Extracellular trap networks (neutrophil extracellular traps [NETs]) of polymorphonuclear neutrophils are mesh-like substances that prevent the spread of pathogens. They primarily consist of DNA skeletons, histones, granule components, and cytoplasmic proteins. NETs formation requires a certain environment and there are different pathways for NETs production. However, it is still not clear how severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) promotes NETs. NETs exert antiinflammatory effects through immune response, while they can also lead to certain adverse outcomes, such as the development of immunothrombosis. Coronavirus disease 2019 (COVID-19) is an inflammatory reaction affecting various organs caused by SARS-CoV-2, especially the lungs. NETs production and disease severity are linked with unique neutrophil clusters by single-cell RNA sequencing. NETs might exert an anti-inflammatory role in the initial stage of lung tissue inflammation. Nevertheless, numerous studies and cases have shown that they can also result in pulmonary thrombosis. There is mounting evidence that NETs are tightly related with COVID-19 pulmonary thrombosis, and many studies on the mechanisms are involved. The role and mechanism of NETs in the development of pulmonary thrombosis will be the main topics of this manuscript. Additionally, we address the potential targeting of NETs in COVID-19 patients.
Collapse
Affiliation(s)
- Shi Li
- Department of ImmunologySchool of Medicine, Jiangsu UniversityZhenjiangJiangsuChina
| | - Hui Wang
- Department of ImmunologySchool of Medicine, Jiangsu UniversityZhenjiangJiangsuChina
| | - Qixiang Shao
- Department of ImmunologySchool of Medicine, Jiangsu UniversityZhenjiangJiangsuChina
- Department of Medical Microbiology and Immunology, Institute of Medical Genetics and Reproductive Immunity, School of Medical Science and Laboratory MedicineJiangsu College of NursingHuai'anJiangsuChina
| |
Collapse
|
28
|
Teixeira-Vaz A, Rocha JA, Oliveira M, Almeida e Reis D, Simões Moreira T, Silva AI, Paiva JA. The PRINCOVID Retrospective Study: A Predictive Model of Pressure Injuries for Critical COVID-19 Patients. Am J Phys Med Rehabil 2023; 102:707-714. [PMID: 36722899 PMCID: PMC10368159 DOI: 10.1097/phm.0000000000002195] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
OBJECTIVE The aim of the study is to characterize pressure injuries, identify risk factors, and develop a predictive model for pressure injuries at intensive care unit admission for critical COVID-19 patients. DESIGN This study was a retrospective analysis of a consecutive sample of patients admitted to intensive care unit between May 2020 and September 2021. Inclusion criteria encompassed the diagnosis of acute respiratory distress syndrome due to SARS-CoV-2, requiring invasive mechanical ventilation more than 48 hrs. The following predictors were evaluated: sociodemographic characteristics, comorbidities, as well as clinical and laboratory findings at intensive care unit admission. The primary outcome was the presence of pressure injuries. RESULTS Two hundred five patients were included, mostly males (73%) with a mean age of 62 yrs. Pressure injury prevalence was 58%. On multivariable analysis, male sex, hypertension, hemoglobin, and albumin at intensive care unit admission were independently associated with pressure injuries, constituting the PRINCOVID model. The model reached an area under the receiver operating characteristic curve of 0.71, surpassing the Braden scale ( P = 0.0015). The PRINCOVID score ranges from 0 to 15, with two risk groups: "at risk"(≤7 points) and "high risk"(>7 points). CONCLUSIONS This study proposes PRINCOVID as a multivariable model for developing pressure injuries in critical COVID-19 patients. Based on four parameters (sex, hypertension, hemoglobin, and albumin at intensive care unit admission), this model fairly predicts the development of pressure injuries. The PRINCOVID score allows patients' classification into two groups, facilitating early identification of high-risk patients.
Collapse
|
29
|
Affiliation(s)
- Scott J Cameron
- Department of Cardiovascular Medicine,
Section of Vascular Medicine, Heart, Vascular and Thoracic Institute, Cleveland Clinic
Foundation, Cleveland, OH, USA
- Department of Hematology, Taussig Cancer
Institute, Cleveland Clinic Foundation, Cleveland, OH, USA
| | - Teresa L Carman
- Harrington Heart & Vascular Institute,
University Hospitals Cleveland Medical Center, Cleveland, OH, USA
| |
Collapse
|
30
|
Ball EE, Weiss CM, Liu H, Jackson K, Keel MK, Miller CJ, Van Rompay KKA, Coffey LL, Pesavento PA. Severe Acute Respiratory Syndrome Coronavirus 2 Vasculopathy in a Syrian Golden Hamster Model. THE AMERICAN JOURNAL OF PATHOLOGY 2023; 193:690-701. [PMID: 36906263 PMCID: PMC9998130 DOI: 10.1016/j.ajpath.2023.02.013] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/22/2022] [Revised: 02/01/2023] [Accepted: 02/13/2023] [Indexed: 03/11/2023]
Abstract
Clinical evidence of vascular dysfunction and hypercoagulability as well as pulmonary vascular damage and microthrombosis are frequently reported in severe cases of human coronavirus disease 2019 (COVID-19). Syrian golden hamsters recapitulate histopathologic pulmonary vascular lesions reported in patients with COVID-19. Herein, special staining techniques and transmission electron microscopy further define vascular pathologies in a Syrian golden hamster model of human COVID-19. The results show that regions of active pulmonary inflammation in severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection are characterized by ultrastructural evidence of endothelial damage with platelet marginalization and both perivascular and subendothelial macrophage infiltration. SARS-CoV-2 antigen/RNA was not detectable within affected blood vessels. Taken together, these findings suggest that the prominent microscopic vascular lesions in SARS-CoV-2-inoculated hamsters likely occur due to endothelial damage followed by platelet and macrophage infiltration.
Collapse
Affiliation(s)
- Erin E Ball
- Department of Pathology, Microbiology, and Immunology, University of California, Davis, California; US Army Veterinary Corps, Washington, District of Columbia
| | - Christopher M Weiss
- Department of Pathology, Microbiology, and Immunology, University of California, Davis, California
| | - Hongwei Liu
- Department of Pathology, Microbiology, and Immunology, University of California, Davis, California
| | - Kenneth Jackson
- Department of Pathology, Microbiology, and Immunology, University of California, Davis, California
| | - M Kevin Keel
- Department of Pathology, Microbiology, and Immunology, University of California, Davis, California
| | - Christopher J Miller
- California National Primate Center, University of California, Davis, California; Center for Immunology and Infectious Diseases, University of California, Davis, California
| | - Koen K A Van Rompay
- Department of Pathology, Microbiology, and Immunology, University of California, Davis, California; California National Primate Center, University of California, Davis, California
| | - Lark L Coffey
- Department of Pathology, Microbiology, and Immunology, University of California, Davis, California.
| | - Patricia A Pesavento
- Department of Pathology, Microbiology, and Immunology, University of California, Davis, California
| |
Collapse
|
31
|
Bonfim LCMG, Guerini IS, Zambon MG, Pires GB, Silva ACF, Gobatto ALN, Lopes MA, Brosnahan SB. Optimal dosing of heparin for prophylactic anticoagulation in critically ill COVID-19 patients a systematic review and meta-analysis of randomized controlled trials. J Crit Care 2023; 77:154344. [PMID: 37244209 PMCID: PMC10211463 DOI: 10.1016/j.jcrc.2023.154344] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2023] [Revised: 04/10/2023] [Accepted: 04/11/2023] [Indexed: 05/29/2023]
Abstract
PURPOSE The optimal amount of anticoagulation for critically ill COVID-19 patients is controversial. Therefore, we aimed to evaluate the efficacy and safety of escalated doses of anticoagulation in critically ill patients with severe COVID-19. MATERIALS AND METHODS We conducted a systematic search of three major databases, including PubMed, Cochrane Library, and Embase, from inception to May 2022. Randomized controlled trials (RCTs) were included comparing therapeutic or intermediate doses to standard prophylactic doses of anticoagulants in critically ill COVID-19 patients, with heparins as the only anticoagulation therapy considered. RESULTS Out of the six RCTs, 2130 patients were administered escalated dose anticoagulation (50.2%) and standard thromboprophylaxis therapy (49.8%). The escalated dose showed no significant impact on mortality (RR, 1.01; 95% CI, 0.90-1.13). Although there was no significant difference in DVT (RR, 0.81; 95% CI, 0.61-1.08), the risk of PE was significantly reduced in patients receiving escalated dose anticoagulation (RR, 0.35; 95% CI, 0.21-0.60), with an increased risk of bleeding events (RR, 1.65; 95% CI, 1.08-2.53). CONCLUSION This systematic review and meta-analysis fail to support escalated anticoagulation doses to reduce mortality in critically ill COVID-19 patients. However, higher doses of anticoagulants appear to reduce thrombotic events while increasing the risk of bleeding effectively.
Collapse
Affiliation(s)
- Luana C M G Bonfim
- Department of Medicine, University of South Santa Catarina, Florianópolis, SC, Brazil.
| | - Isadora S Guerini
- Department of Medicine, State University of Western Paraná, Francisco Beltrão, PR, Brazil
| | - Marjorie G Zambon
- Department of Medicine, Mackenzie Evangelical University Hospital, Curitiba, PR, Brazil
| | - Gabriela B Pires
- Department of Medicine, School of Medicine Souza Marques, Rio de Janeiro, RJ, Brazil
| | - Amanda C F Silva
- Department of Medicine, Federal University of Santa Catarina, Florianópolis, SC, Brazil
| | - André L N Gobatto
- Department of Critical Care, São Rafael Hospital, Salvador, Bahia, Brazil; Department of Critical Care, Hospital da Cidade, Salvador, Bahia, Brazil
| | - Marcela A Lopes
- Department of Critical Care, Hospital da Cidade, Salvador, Bahia, Brazil
| | - Shari B Brosnahan
- Department of Pulmonary and Critical Care, New York University School of Medicine, New York, NY, USA
| |
Collapse
|
32
|
Wang T, Zhai Y, Xue H, Zhou W, Ding Y, Nie H. Regulation of Epithelial Sodium Transport by SARS-CoV-2 Is Closely Related with Fibrinolytic System-Associated Proteins. Biomolecules 2023; 13:biom13040578. [PMID: 37189326 DOI: 10.3390/biom13040578] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Revised: 03/08/2023] [Accepted: 03/21/2023] [Indexed: 05/17/2023] Open
Abstract
Dyspnea and progressive hypoxemia are the main clinical features of patients with coronavirus disease 2019 (COVID-19), which is caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). Pulmonary pathology shows diffuse alveolar damage with edema, hemorrhage, and the deposition of fibrinogens in the alveolar space, which are consistent with the Berlin Acute Respiratory Distress Syndrome Criteria. The epithelial sodium channel (ENaC) is a key channel protein in alveolar ion transport and the rate-limiting step for pulmonary edema fluid clearance, the dysregulation of which is associated with acute lung injury/acute respiratory distress syndrome. The main protein of the fibrinolysis system, plasmin, can bind to the furin site of γ-ENaC and induce it to an activation state, facilitating pulmonary fluid reabsorption. Intriguingly, the unique feature of SARS-CoV-2 from other β-coronaviruses is that the spike protein of the former has the same furin site (RRAR) with ENaC, suggesting that a potential competition exists between SARS-CoV-2 and ENaC for the cleavage by plasmin. Extensive pulmonary microthrombosis caused by disorders of the coagulation and fibrinolysis system has also been seen in COVID-19 patients. To some extent, high plasmin (ogen) is a common risk factor for SARS-CoV-2 infection since an increased cleavage by plasmin accelerates virus invasion. This review elaborates on the closely related relationship between SARS-CoV-2 and ENaC for fibrinolysis system-related proteins, aiming to clarify the regulation of ENaC under SARS-CoV-2 infection and provide a novel reference for the treatment of COVID-19 from the view of sodium transport regulation in the lung epithelium.
Collapse
Affiliation(s)
- Tingyu Wang
- Department of Stem Cells and Regenerative Medicine, College of Basic Medical Science, China Medical University, Shenyang 110122, China
| | - Yiman Zhai
- Department of Stem Cells and Regenerative Medicine, College of Basic Medical Science, China Medical University, Shenyang 110122, China
| | - Hao Xue
- Department of Stem Cells and Regenerative Medicine, College of Basic Medical Science, China Medical University, Shenyang 110122, China
| | - Wei Zhou
- Department of Stem Cells and Regenerative Medicine, College of Basic Medical Science, China Medical University, Shenyang 110122, China
| | - Yan Ding
- Department of Stem Cells and Regenerative Medicine, College of Basic Medical Science, China Medical University, Shenyang 110122, China
| | - Hongguang Nie
- Department of Stem Cells and Regenerative Medicine, College of Basic Medical Science, China Medical University, Shenyang 110122, China
| |
Collapse
|
33
|
Ishikura H, Maruyama J, Nakashio M, Hoshino K, Morimoto S, Izutani Y, Noake J, Yamagaito T, Yoshida M, Kitamura T, Nakamura Y. Daily combined measurement of platelet count and presepsin concentration can predict in-hospital death of patients with severe coronavirus disease 2019 (COVID-19). Int J Hematol 2023; 117:845-855. [PMID: 36920687 PMCID: PMC10016182 DOI: 10.1007/s12185-023-03555-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Revised: 01/31/2023] [Accepted: 02/01/2023] [Indexed: 03/16/2023]
Abstract
The purpose of this study was to classify patients with severe COVID-19 into more detailed risk groups using coagulation/fibrinolysis, inflammation/immune response, and alveolar/myocardial damage biomarkers, as well as to identify prognostic markers for these patients. These biomarkers were measured every day for eight intensive care unit days in 54 adult patients with severe COVID-19. The patients were classified into survivor (n = 40) and non-survivor (n = 14) groups. Univariate and multivariate analyses showed that the combined measurement of platelet count and presepsin concentrations may be the most valuable for predicting in-hospital death, and receiver operating characteristic curve analysis further confirmed this result (area under the curve = 0.832). Patients were consequently classified into three groups (high-, medium-, and low-risk) on the basis of their cutoff values (platelet count 53 × 103/µL, presepsin 714 pg/mL). The Kaplan-Meier curve for 90-day survival by each group showed that the 90-day mortality rate significantly increased as risk level increased (P < 0.01 by the log-rank test). Daily combined measurement of platelet count and presepsin concentration may be useful for predicting in-hospital death and classifying patients with severe COVID-19 into more detailed risk groups.
Collapse
Affiliation(s)
- Hiroyasu Ishikura
- Department of Emergency and Critical Care Medicine, Faculty of Medicine, Fukuoka University, 7-45-1 Nanakuma, Jonan-ku, Fukuoka, 814-0180, Japan.
| | - Junichi Maruyama
- Department of Emergency and Critical Care Medicine, Faculty of Medicine, Fukuoka University, 7-45-1 Nanakuma, Jonan-ku, Fukuoka, 814-0180, Japan
| | - Maiko Nakashio
- Department of Emergency and Critical Care Medicine, Faculty of Medicine, Fukuoka University, 7-45-1 Nanakuma, Jonan-ku, Fukuoka, 814-0180, Japan
| | - Kota Hoshino
- Department of Emergency and Critical Care Medicine, Faculty of Medicine, Fukuoka University, 7-45-1 Nanakuma, Jonan-ku, Fukuoka, 814-0180, Japan
| | - Shinichi Morimoto
- Department of Emergency and Critical Care Medicine, Faculty of Medicine, Fukuoka University, 7-45-1 Nanakuma, Jonan-ku, Fukuoka, 814-0180, Japan
| | - Yoshito Izutani
- Department of Emergency and Critical Care Medicine, Faculty of Medicine, Fukuoka University, 7-45-1 Nanakuma, Jonan-ku, Fukuoka, 814-0180, Japan
| | - Junta Noake
- Department of Emergency and Critical Care Medicine, Faculty of Medicine, Fukuoka University, 7-45-1 Nanakuma, Jonan-ku, Fukuoka, 814-0180, Japan
| | | | - Maho Yoshida
- Sysmex Scientific Affairs, 1-3-2 Murotani, Nishi-ku, Kobe, Japan
| | - Taisuke Kitamura
- Department of Emergency and Critical Care Medicine, Faculty of Medicine, Fukuoka University, 7-45-1 Nanakuma, Jonan-ku, Fukuoka, 814-0180, Japan
| | - Yoshihiko Nakamura
- Department of Emergency and Critical Care Medicine, Faculty of Medicine, Fukuoka University, 7-45-1 Nanakuma, Jonan-ku, Fukuoka, 814-0180, Japan
| |
Collapse
|
34
|
Bashir H, Yildiz M, Cafardi J, Bhatia A, Garcia S, Henry TD, Chung ES. A Review of Heart Failure in patients with COVID-19. Heart Fail Clin 2023; 19:e1-e8. [PMID: 37169437 PMCID: PMC9988711 DOI: 10.1016/j.hfc.2023.03.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 03/09/2023]
Abstract
The interplay of COVID-19 and heart failure is complex and involves direct and indirect effects. Patients with existing heart failure develop more severe COVID-19 symptoms and have worse clinical outcomes. Pandemic-related policies and protocols have negatively affected care for cardiovascular conditions and established hospital protocols, which is particularly important for patients with heart failure.
Collapse
Affiliation(s)
- Hanad Bashir
- The Carl and Edyth Lindner Center for Research and Education at The Christ Hospital, The Christ Hospital Health Network, 2139 Auburn Avenue, Suite 424, Cincinnati, OH 45219, USA; The Christ Hospital Heart and Vascular Institute, The Christ Hospital, Cincinnati, OH, USA. https://twitter.com/HanadBashirMD
| | - Mehmet Yildiz
- The Carl and Edyth Lindner Center for Research and Education at The Christ Hospital, The Christ Hospital Health Network, 2139 Auburn Avenue, Suite 424, Cincinnati, OH 45219, USA; The Christ Hospital Heart and Vascular Institute, The Christ Hospital, Cincinnati, OH, USA
| | - John Cafardi
- Infectious Disease Department, The Christ Hospital, The Christ Hospital Health Network, 2139 Auburn Avenue, Suite 424, Cincinnati, OH 45219, USA
| | - Ankit Bhatia
- The Carl and Edyth Lindner Center for Research and Education at The Christ Hospital, The Christ Hospital Health Network, 2139 Auburn Avenue, Suite 424, Cincinnati, OH 45219, USA; The Christ Hospital Heart and Vascular Institute, The Christ Hospital, Cincinnati, OH, USA. https://twitter.com/AKBhatiaMD
| | - Santiago Garcia
- The Carl and Edyth Lindner Center for Research and Education at The Christ Hospital, The Christ Hospital Health Network, 2139 Auburn Avenue, Suite 424, Cincinnati, OH 45219, USA; The Christ Hospital Heart and Vascular Institute, The Christ Hospital, Cincinnati, OH, USA
| | - Timothy D Henry
- The Carl and Edyth Lindner Center for Research and Education at The Christ Hospital, The Christ Hospital Health Network, 2139 Auburn Avenue, Suite 424, Cincinnati, OH 45219, USA; The Christ Hospital Heart and Vascular Institute, The Christ Hospital, Cincinnati, OH, USA. https://twitter.com/HenrytTimothy
| | - Eugene S Chung
- The Carl and Edyth Lindner Center for Research and Education at The Christ Hospital, The Christ Hospital Health Network, 2139 Auburn Avenue, Suite 424, Cincinnati, OH 45219, USA; The Christ Hospital Heart and Vascular Institute, The Christ Hospital, Cincinnati, OH, USA.
| |
Collapse
|
35
|
Hogwood J, Mulloy B, Lever R, Gray E, Page CP. Pharmacology of Heparin and Related Drugs: An Update. Pharmacol Rev 2023; 75:328-379. [PMID: 36792365 DOI: 10.1124/pharmrev.122.000684] [Citation(s) in RCA: 30] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2022] [Revised: 11/04/2022] [Accepted: 11/08/2022] [Indexed: 02/17/2023] Open
Abstract
Heparin has been used extensively as an antithrombotic and anticoagulant for close to 100 years. This anticoagulant activity is attributed mainly to the pentasaccharide sequence, which potentiates the inhibitory action of antithrombin, a major inhibitor of the coagulation cascade. More recently it has been elucidated that heparin exhibits anti-inflammatory effect via interference of the formation of neutrophil extracellular traps and this may also contribute to heparin's antithrombotic activity. This illustrates that heparin interacts with a broad range of biomolecules, exerting both anticoagulant and nonanticoagulant actions. Since our previous review, there has been an increased interest in these nonanticoagulant effects of heparin, with the beneficial role in patients infected with SARS2-coronavirus a highly topical example. This article provides an update on our previous review with more recent developments and observations made for these novel uses of heparin and an overview of the development status of heparin-based drugs. SIGNIFICANCE STATEMENT: This state-of-the-art review covers recent developments in the use of heparin and heparin-like materials as anticoagulant, now including immunothrombosis observations, and as nonanticoagulant including a role in the treatment of SARS-coronavirus and inflammatory conditions.
Collapse
Affiliation(s)
- John Hogwood
- Sackler Institute of Pulmonary Pharmacology, Institute of Pharmaceutical Science, King's College London, London, United Kingdom (B.M., E.G., C.P.P.); National Institute for Biological Standards and Control, South Mimms, Hertfordshire, United Kingdom (J.H., E.G.) and School of Pharmacy, University College London, London, United Kingdom (R.L.)
| | - Barbara Mulloy
- Sackler Institute of Pulmonary Pharmacology, Institute of Pharmaceutical Science, King's College London, London, United Kingdom (B.M., E.G., C.P.P.); National Institute for Biological Standards and Control, South Mimms, Hertfordshire, United Kingdom (J.H., E.G.) and School of Pharmacy, University College London, London, United Kingdom (R.L.)
| | - Rebeca Lever
- Sackler Institute of Pulmonary Pharmacology, Institute of Pharmaceutical Science, King's College London, London, United Kingdom (B.M., E.G., C.P.P.); National Institute for Biological Standards and Control, South Mimms, Hertfordshire, United Kingdom (J.H., E.G.) and School of Pharmacy, University College London, London, United Kingdom (R.L.)
| | - Elaine Gray
- Sackler Institute of Pulmonary Pharmacology, Institute of Pharmaceutical Science, King's College London, London, United Kingdom (B.M., E.G., C.P.P.); National Institute for Biological Standards and Control, South Mimms, Hertfordshire, United Kingdom (J.H., E.G.) and School of Pharmacy, University College London, London, United Kingdom (R.L.)
| | - Clive P Page
- Sackler Institute of Pulmonary Pharmacology, Institute of Pharmaceutical Science, King's College London, London, United Kingdom (B.M., E.G., C.P.P.); National Institute for Biological Standards and Control, South Mimms, Hertfordshire, United Kingdom (J.H., E.G.) and School of Pharmacy, University College London, London, United Kingdom (R.L.)
| |
Collapse
|
36
|
Zerangian N, Erabi G, Poudineh M, Monajjem K, Diyanati M, Khanlari M, Khalaji A, Allafi D, Faridzadeh A, Amali A, Alizadeh N, Salimi Y, Ghane Ezabadi S, Abdi A, Hasanabadi Z, ShojaeiBaghini M, Deravi N. Venous thromboembolism in viral diseases: A comprehensive literature review. Health Sci Rep 2023; 6:e1085. [PMID: 36778773 PMCID: PMC9900357 DOI: 10.1002/hsr2.1085] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Revised: 12/25/2022] [Accepted: 01/19/2023] [Indexed: 02/09/2023] Open
Abstract
Venous thromboembolism (VTE) is known to be a common respiratory and/or cardiovascular complication in hospitalized patients with viral infections. Numerous studies have proven human immunodeficiency virus infection to be a prothrombotic condition. An elevated VTE risk has been observed in critically ill H1N1 influenza patients. VTE risk is remarkably higher in patients infected with the Hepatitis C virus in contrast to uninfected subjects. The elevation of D-dimer levels supported the association between Chikungunya and the Zika virus and the rise of clinical VTE risk. Varicella-zoster virus is a risk factor for both cellulitis and the consequent invasive bacterial disease which may take part in thrombotic initiation. Eventually, hospitalized patients infected with the coronavirus disease of 2019 (COVID-19), the cause of the ongoing worldwide pandemic, could mainly suffer from an anomalous risk of coagulation activation with enhanced venous thrombosis events and poor quality clinical course. Although the risk of VTE in nonhospitalized COVID-19 patients is not known yet, there are a large number of guidelines and studies on thromboprophylaxis administration for COVID-19 cases. This study aims to take a detailed look at the effect of viral diseases on VTE, the epidemiology of VTE in viral diseases, and the diagnosis and treatment of VTE.
Collapse
Affiliation(s)
- Nasibeh Zerangian
- Health Education and Health Promotion, Department of Health Education and Health Promotion, School of HealthMashhad University of Medical SciencesMashhadIran
| | - Gisou Erabi
- Student Research CommitteeUrmia University of Medical SciencesUrmiaIran
| | | | - Kosar Monajjem
- Student Research CommitteeTabriz University of Medical SciencesTabrizIran
| | - Maryam Diyanati
- Student Research CommitteeRafsanjan University of Medical SciencesRafsanjanIran
| | - Maryam Khanlari
- Student Research CommitteeTabriz University of Medical SciencesTabrizIran
| | | | - Diba Allafi
- Student Research CommitteeUrmia University of Medical SciencesUrmiaIran
| | - Arezoo Faridzadeh
- Department of Immunology and Allergy, School of MedicineMashhad University of Medical SciencesMashhadIran
- Immunology Research CenterMashhad University of Medical SciencesMashhadIran
| | - Arian Amali
- Student Research Committee, Paramedical DepartmentIslamic Azad University, Mashhad BranchMashhadIran
| | - Nilufar Alizadeh
- Doctor of Medicine (MD), School of MedicineIran University of Medical SciencesTehranIran
| | - Yasaman Salimi
- Student Research CommitteeKermanshah University of Medical SciencesKermanshahIran
| | - Sajjad Ghane Ezabadi
- Student's Scientific Research Center, School of MedicineTehran University of Medical SciencesTehranIran
| | - Amir Abdi
- Student Research Committee, School of Medicine, Tehran Medical SciencesIslamic Azad UniversityTehranIran
| | - Zahra Hasanabadi
- Doctor of Medicine (MD), School of MedicineQazvin University of Medical ScienceQazvinIran
| | - Mahdie ShojaeiBaghini
- Medical Informatics Research Center, Institute for Futures Studies in HealthKerman University of Medical SciencesKermanIran
| | - Niloofar Deravi
- Student Research Committee, School of MedicineShahid Beheshti University of Medical SciencesTehranIran
| |
Collapse
|
37
|
Volod O, Bunch CM, Miller J, Moore EE, Moore HB, Kwaan HC, Patel SS, Wiarda G, Aboukhaled M, Thomas SG, Fulkerson D, Erdman L, Tincher A, Walsh MM. Reply to Bareille et al. Are Viscoelastometric Assays of Old Generation Ready for Disposal? Comment on "Volod et al. Viscoelastic Hemostatic Assays: A Primer on Legacy and New Generation Devices. J. Clin. Med. 2022, 11, 860". J Clin Med 2023; 12:jcm12020478. [PMID: 36675408 PMCID: PMC9862366 DOI: 10.3390/jcm12020478] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2022] [Revised: 12/16/2022] [Accepted: 12/27/2022] [Indexed: 01/11/2023] Open
Abstract
We are pleased to see that Bareille et al. have written a Commentary: "Are viscoelastometric assays of old generation ready for disposal?" [...].
Collapse
Affiliation(s)
- Oksana Volod
- Department of Pathology and Laboratory Medicine, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
- Correspondence:
| | - Connor M. Bunch
- Department of Emergency Medicine, Henry Ford Hospital, Detroit, MI 48402, USA
- Department of Internal Medicine, Saint Joseph Regional Medical Center, Mishawaka, IN 46545, USA
| | - Joseph Miller
- Department of Emergency Medicine, Henry Ford Hospital, Detroit, MI 48402, USA
| | - Ernest E. Moore
- Department of Surgery, Ernest E. Moore Shock Trauma Center at Denver Health, University of Colorado Health Sciences Center, Denver, CO 80204, USA
| | - Hunter B. Moore
- Department of Surgery, Ernest E. Moore Shock Trauma Center at Denver Health, University of Colorado Health Sciences Center, Denver, CO 80204, USA
| | - Hau C. Kwaan
- Division of Hematology and Oncology, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Shivani S. Patel
- Department of Internal Medicine, Saint Joseph Regional Medical Center, Mishawaka, IN 46545, USA
| | - Grant Wiarda
- Department of Internal Medicine, Saint Joseph Regional Medical Center, Mishawaka, IN 46545, USA
| | - Michael Aboukhaled
- Department of Internal Medicine, Saint Joseph Regional Medical Center, Mishawaka, IN 46545, USA
| | - Scott G. Thomas
- Department of Trauma Surgery, Memorial Leighton Trauma Center, Beacon Health System, South Bend, IN 46601, USA
| | - Daniel Fulkerson
- Department of Trauma Surgery, Memorial Leighton Trauma Center, Beacon Health System, South Bend, IN 46601, USA
| | - Lee Erdman
- Department of Internal Medicine, Saint Joseph Regional Medical Center, Mishawaka, IN 46545, USA
| | - Anna Tincher
- Department of Internal Medicine, Saint Joseph Regional Medical Center, Mishawaka, IN 46545, USA
| | - Mark M. Walsh
- Department of Internal Medicine, Saint Joseph Regional Medical Center, Mishawaka, IN 46545, USA
| |
Collapse
|
38
|
Yachi S, Takeyama M, Nishimoto Y, Tsujino I, Nakamura J, Yamamoto N, Nakata H, Ikeda S, Umetsu M, Aikawa S, Hayashi H, Satokawa H, Okuno Y, Iwata E, Ogihara Y, Ikeda N, Kondo A, Iwai T, Yamada N, Ogawa T, Kobayashi T, Mo M, Yamashita Y. Risk Factors and Impact on Outcomes of Thrombosis in Patients with COVID-19 in Japan: From the CLOT-COVID Study. Ann Vasc Dis 2023; 16:31-37. [PMID: 37006864 PMCID: PMC10064299 DOI: 10.3400/avd.oa.22-00071] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2022] [Accepted: 11/06/2022] [Indexed: 01/15/2023] Open
Abstract
Objectives: The relationship between the thrombotic event and prognosis in patients with coronavirus disease 2019 (COVID-19) has not yet been fully investigated in Japan. Our study aimed to investigate the clinical outcomes and risk factors for thrombosis in hospitalized patients with COVID-19 in Japan. Materials and Methods: We compared the patient characteristics and clinical outcomes among patients with thrombosis (N=55) and those without thrombosis (N=2839) by using a large-scale data of CLOT-COVID study (thrombosis and antiCoaguLatiOn Therapy in patients with COVID-19 in Japan Study: UMIN000045800). Thrombosis included venous thromboembolism, ischemic stroke, myocardial infarction, and systemic arterial thromboembolism. Results: Higher rates of mortality and bleeding events were shown in hospitalized patients with COVID-19 with thrombosis compared to those without thrombosis (all-cause mortality, 23.6% vs. 5.1%, P<0.001; major bleeding, 23.6% vs. 1.6%, P<0.001). Multivariable analysis revealed that the independent risk factors of thrombosis were male sex, D-dimer level on admission>1.0 µg/mL, and moderate and severe COVID-19 status on admission. Conclusions: The development of thrombosis in hospitalized patients with COVID-19 was related to higher mortality and major bleeding, and several independent risk factors for thrombosis could help determine the patient-appropriate treatment for COVID-19.
Collapse
Affiliation(s)
- Sen Yachi
- Japan Community Health care Organization Tokyo Shinjuku Medical Center
| | - Makoto Takeyama
- Japan Community Health care Organization Tokyo Shinjuku Medical Center
| | | | | | | | | | | | - Satoshi Ikeda
- Nagasaki University Graduate School of Biomedical Sciences
| | | | | | - Hiroya Hayashi
- Osaka Metropolitan University Graduate School of Medicine
| | | | | | - Eriko Iwata
- Japan Community Health care Organization Nankai Medical Center
| | | | | | - Akane Kondo
- Shikoku Medical Center for Children and Adults
| | | | | | | | | | | | | |
Collapse
|
39
|
Moafi M, Ebrahimi MJ, Hatami F, Javandoust Gharehbagh F, Ahmadzadeh A, Emam MM, Rajaei A, Mansouri D, Alavi Darazam I. Lupus Anticoagulant Is Associated with Critical Cases and High Mortality in COVID-19: A Literature Review. TANAFFOS 2023; 22:53-60. [PMID: 37920325 PMCID: PMC10618584] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Subscribe] [Scholar Register] [Received: 09/13/2022] [Accepted: 01/04/2023] [Indexed: 11/04/2023]
Abstract
Background In severe COVID-19 cases, a hypercoagulable state may occur. Antiphospholipid syndrome-related auto-antibodies (APSRAs) contribute to coagulopathy, but their role in COVID- 19 remains unclear. We aimed to investigate the prevalence of positive APSRAs and their effect on clinical outcomes in confirmed COVID-19 patients. Materials and Methods In this cross-sectional study, severe hospitalized COVID-19 cases were enrolled. Demographic and clinical data were obtained from the day of admission. APSRAs including IgG and/or IgM anticardiolipin (aCL) and anti-β2-glycoprotein1 (anti-β2GP1) as well as lupus anticoagulant (LAC) were measured. Results In this study, 54 severe COVID-19 cases with positive RT-PCR and chest CT scans were recruited. Positive APSRAs were found in 7 (12.9%) patients. Positive LAC was a more prevalent marker as compared to other tests (11.1%). The prevalence of positive aCL (IgM or IgG) and anti-ß2 GPI (IgM or IgG) was 1.8% (in an elderly woman). Lower oxygen saturation was found in the positive APSRAs group as opposed to the negative APSRAs group (70.3±9 vs. 84.8±9.7%). The mortality rate in the positive APSRAs group was significantly higher relative to the negative APSRAs group (83.3% vs. 27.1%; P-value: 0.01). Likewise, the mechanical ventilation requirement in the positive group was also higher (50% vs. 27.1%, P-value: 0.28). Conclusion This study indicated that LAC might be associated with critical cases and high mortality of COVID-19. Nonetheless, the mortality was not related to macrothrombotic incidence.
Collapse
Affiliation(s)
- Maral Moafi
- Department of Infectious Diseases and Tropical Medicine, Loghman Hakim Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Infectious Diseases and Tropical Medicine Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mohammad Javad Ebrahimi
- Department of Infectious Diseases and Tropical Medicine, Loghman Hakim Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Infectious Diseases and Tropical Medicine Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Firouze Hatami
- Department of Infectious Diseases and Tropical Medicine, Loghman Hakim Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Infectious Diseases and Tropical Medicine Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Farid Javandoust Gharehbagh
- Infectious Diseases and Tropical Medicine Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Arman Ahmadzadeh
- Rheumatology Department, Loghman Hakim Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mohammad Mehdi Emam
- Rheumatology Department, Loghman Hakim Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Alireza Rajaei
- Rheumatology Department, Loghman Hakim Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Davood Mansouri
- Division of Infectious Diseases and Clinical Immunology, National Research Institute of Tuberculosis and Lung Diseases, Shahid Beheshti University of Medical Sciences, Teheran, Iran
| | - Ilad Alavi Darazam
- Department of Infectious Diseases and Tropical Medicine, Loghman Hakim Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Infectious Diseases and Tropical Medicine Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
40
|
Sun H, Du Y, Kumar R, Buchkovich N, He P. Increased circulating microparticles contribute to severe infection and adverse outcomes of COVID-19 in patients with diabetes. Am J Physiol Heart Circ Physiol 2022; 323:H1176-H1193. [PMID: 36269646 PMCID: PMC9678425 DOI: 10.1152/ajpheart.00409.2022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/08/2022] [Revised: 10/13/2022] [Accepted: 10/15/2022] [Indexed: 12/14/2022]
Abstract
Patients with diabetes infected with COVID-19 have greater mortality than those without comorbidities, but the underlying mechanisms remain unknown. This study aims to identify the mechanistic interactions between diabetes and severe COVID-19. Microparticles (MPs), the cell membrane-derived vesicles released on cell activation, are largely increased in patients with diabetes. To date, many mechanisms have been postulated for increased severity of COVID-19 in patients with underlying conditions, but the contributions of excessive MPs in patients with diabetes have been overlooked. This study characterizes plasma MPs from normal human subjects and patients with type 2 diabetes in terms of amount, cell origins, surface adhesive properties, ACE2 expression, spike protein binding capacity, and their roles in SARS-CoV-2 infection. Results showed that over 90% of plasma MPs express ACE2 that binds the spike protein of SARS-CoV-2. MPs in patients with diabetes increase 13-fold in quantity and 11-fold in adhesiveness when compared with normal subjects. Perfusion of human plasma with pseudo-typed SARS-CoV-2 virus or spike protein-bound MPs into human endothelial cell-formed microvessels-on-a chip demonstrated that MPs from patients with diabetes, not normal subjects, interact with endothelium and carry SARS-CoV-2 into cells through endocytosis, providing additional virus entry pathways and enhanced infection. Results also showed a large percentage of platelet-derived tissue factor-bearing MPs in diabetic plasma, which could contribute to thrombotic complications with SARS-CoV-2 infection. This study reveals a dual role of diabetic MPs in promoting SARS-CoV-2 entry and propagating vascular inflammation. These findings provide novel mechanistic insight into the high prevalence of COVID-19 in patients with diabetes and their propensity to develop severe vascular complications.NEW & NOTEWORTHY This study provides the first evidence that over 90% of human plasma microparticles express ACE2 that binds SARS-CoV-2 S protein with high affinity. Thus, the highly elevated adhesive circulating microparticles identified in patients with diabetes not only have greater SARS-CoV-2 binding capacity but also enable additional viral entry through virus-bound microparticle-endothelium interactions and enhanced infection. These findings reveal a novel mechanistic insight into the adverse outcomes of COVID-19 in patients with diabetes.
Collapse
Affiliation(s)
- Haoyu Sun
- Department of Cellular and Molecular Physiology, College of Medicine, Pennsylvania State University, Hershey, Pennsylvania
| | - Yong Du
- Department of Cellular and Molecular Physiology, College of Medicine, Pennsylvania State University, Hershey, Pennsylvania
| | - Rinki Kumar
- Department of Microbiology and Immunology, College of Medicine, Pennsylvania State University, Hershey, Pennsylvania
| | - Nicholas Buchkovich
- Department of Microbiology and Immunology, College of Medicine, Pennsylvania State University, Hershey, Pennsylvania
| | - Pingnian He
- Department of Cellular and Molecular Physiology, College of Medicine, Pennsylvania State University, Hershey, Pennsylvania
| |
Collapse
|
41
|
Cabrera-Garcia D, Miltiades A, Yim P, Parsons S, Elisman K, Mansouri MT, Wagener G, Harrison NL. Plasma biomarkers associated with survival and thrombosis in hospitalized COVID-19 patients. Int J Hematol 2022; 116:937-946. [PMID: 35994163 PMCID: PMC9395834 DOI: 10.1007/s12185-022-03437-2] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2022] [Revised: 07/25/2022] [Accepted: 08/01/2022] [Indexed: 10/26/2022]
Abstract
Severe coronavirus disease-19 (COVID-19) has been associated with fibrin-mediated hypercoagulability and thromboembolic complications. To evaluate potential biomarkers of coagulopathy and disease severity in COVID-19, we measured plasma levels of eight biomarkers potentially associated with coagulation, fibrinolysis, and platelet function in 43 controls and 63 COVID-19 patients, including 47 patients admitted to the intensive care unit (ICU) and 16 non-ICU patients. COVID-19 patients showed significantly elevated levels of fibrinogen, tissue plasminogen activator (t-PA), and its inhibitor plasminogen activation inhibitor 1 (PAI-1), as well as ST2 (the receptor for interleukin-33) and von Willebrand factor (vWF) compared to the control group. We found that higher levels of t-PA, ST2, and vWF at the time of admission were associated with lower survival rates, and that thrombotic events were more frequent in patients with initial higher levels of vWF. These results support a predictive role of specific biomarkers such as t-PA and vWF in the pathophysiology of COVID-19. The data provide support for the case that hypercoagulability in COVID-19 is fibrin-mediated, but also highlights the important role that vWF may play in the genesis of thromboses in the pathophysiology of COVID-19. Interventions designed to enhance fibrinolysis might prove to be useful adjuncts in the treatment of coagulopathy in a subset of COVID-19 patients.
Collapse
Affiliation(s)
- David Cabrera-Garcia
- Department of Anesthesiology, Columbia University Irving Medical Center, 630 West 168th Street, New York, NY, 10032, USA
| | - Andrea Miltiades
- Department of Anesthesiology, Columbia University Irving Medical Center, 630 West 168th Street, New York, NY, 10032, USA.
| | - Peter Yim
- Department of Anesthesiology, Columbia University Irving Medical Center, 630 West 168th Street, New York, NY, 10032, USA
| | - Samantha Parsons
- Department of Anesthesiology, Columbia University Irving Medical Center, 630 West 168th Street, New York, NY, 10032, USA
| | - Katerina Elisman
- Department of Anesthesiology, Columbia University Irving Medical Center, 630 West 168th Street, New York, NY, 10032, USA
| | - Mohammad Taghi Mansouri
- Department of Anesthesiology, Columbia University Irving Medical Center, 630 West 168th Street, New York, NY, 10032, USA
| | - Gebhard Wagener
- Department of Anesthesiology, Columbia University Irving Medical Center, 630 West 168th Street, New York, NY, 10032, USA.
| | - Neil L Harrison
- Department of Anesthesiology, Columbia University Irving Medical Center, 630 West 168th Street, New York, NY, 10032, USA.
- Department of Molecular Pharmacology and Therapeutics, Columbia University Irving Medical Center, 630 West 168th Street, New York, NY, 10032, USA.
| |
Collapse
|
42
|
COVID-19 and Pulmonary Thrombosis-An Unresolved Clinical Puzzle: A Single-Center Cohort Study. J Clin Med 2022; 11:jcm11237049. [PMID: 36498623 PMCID: PMC9740696 DOI: 10.3390/jcm11237049] [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: 10/31/2022] [Revised: 11/11/2022] [Accepted: 11/26/2022] [Indexed: 12/03/2022] Open
Abstract
Pulmonary thrombosis (PT) is a frequent complication of COVID-19. However, the risk factors, predictive scores, and precise diagnostic guidelines on indications for CT pulmonary angiography (CTPA) are still lacking. This study aimed to analyze the clinical and laboratory characteristics associated with PT in patients with COVID-19. We conducted a cohort study of consecutively hospitalized adult patients with COVID-19 who underwent CTPA at the University Hospital for Infectious Diseases in Zagreb, Croatia between 1 April and 31 December 2021. Of 2078 hospitalized patients, 575 (27.6%) underwent CTPA. PT was diagnosed in 178 (30.9%) patients (69.6% males, median age of 61, IQR 50-69 years). The PT group had a higher CRP, LDH, D-dimer, platelets, and CHOD score. PT was more frequent in patients requiring ≥15 L O2/min (25.0% vs. 39.7%). In multivariable analysis, only D-dimer ≥ 1.0 mg/L (OR 1.78, 95%CI 1.12-2.75) and O2 ≥ 15 L (OR 1.89, 95%CI 1.26-2.84) were associated with PT. PT was not associated with in-hospital mortality. In conclusion, our data confirmed a high incidence of PT in hospitalized patients with COVID-19, however, no correlation with traditional risk factors and mortality was found. CTPA should be performed in patients requiring high-flow supplemental oxygen or those with increased D-dimer levels.
Collapse
|
43
|
Cooper N, Ghanima W, Hill QA, Nicolson PLR, Markovtsov V, Kessler C. Recent advances in understanding spleen tyrosine kinase (SYK) in human biology and disease, with a focus on fostamatinib. Platelets 2022; 34:2131751. [DOI: 10.1080/09537104.2022.2131751] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Affiliation(s)
- Nichola Cooper
- Clinical Reader in Immune Haematology and Honorary Consultant, Faculty of Medicine, Department of Immunology and Inflammation, Imperial College London, London, UK
| | - Waleed Ghanima
- Head of Research and Consultant Haematologist, Department of Hemato-oncology, Østfold Hospital, and Department of Hematology, Institute of Clinical Medicine, Oslo University, Oslo, Norway
| | - Quentin A Hill
- Consultant Haematologist, Department of Haematology, Leeds Teaching Hospitals, Leeds, UK
| | - Phillip LR Nicolson
- Clinical Lecturer in Haematology, Institute of Cardiovascular Sciences, University of Birmingham, and Department of Haematology, University Hospitals Birmingham NHS Foundation Trust, Birmingham, UK
| | - Vadim Markovtsov
- Translational Biology, Rigel Pharmaceuticals, South San Francisco, CA, USA
| | - Craig Kessler
- Medicine and Pathology, Director, Division of Coagulation, Director, Cellular and Therapeutic Apheresis and Cellular Collection, Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC, USA
| |
Collapse
|
44
|
Axelerad A, Stuparu AZ, Muja LF, Docu Axelerad S, Petrov SG, Gogu AE, Jianu DC. Narrative Review of New Insight into the Influence of the COVID-19 Pandemic on Cardiovascular Care. MEDICINA (KAUNAS, LITHUANIA) 2022; 58:1554. [PMID: 36363511 PMCID: PMC9694465 DOI: 10.3390/medicina58111554] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/25/2022] [Revised: 10/21/2022] [Accepted: 10/26/2022] [Indexed: 09/10/2024]
Abstract
Background and Objectives: The purpose of this paper was to perform a literature review on the effects of the COVID-19 pandemic on cardiothoracic and vascular surgery care and departments. Materials and Methods: To conduct this evaluation, an electronic search of many databases was conducted, and the resulting papers were chosen and evaluated. Results: Firstly, we have addressed the impact of COVID-19 infection on the cardiovascular system from the pathophysiological and treatment points of view. Afterwards, we analyzed every cardiovascular disease that seemed to appear after a COVID-19 infection, emphasizing the treatment. In addition, we have analyzed the impact of the pandemic on the cardiothoracic and vascular departments in different countries and the transitions that appeared. Finally, we discussed the implications of the cardiothoracic and vascular specialists' and residents' work and studies on the pandemic. Conclusions: The global pandemic caused by SARS-CoV-2 compelled the vascular profession to review the treatment of certain vascular illnesses and find solutions to address the vascular consequences of COVID-19 infection. The collaboration between vascular surgeons, public health specialists, and epidemiologists must continue to investigate the impact of the pandemic and the response to the public health issue.
Collapse
Affiliation(s)
- Any Axelerad
- Department of Neurology, General Medicine Faculty, ‘Ovidius’ University, 900470 Constanta, Romania
- Department of Neurology, ‘Sf. Ap. Andrei’ County Clinical Emergency Hospital of Constanta, 900591 Constanta, Romania
| | - Alina Zorina Stuparu
- Department of Neurology, General Medicine Faculty, ‘Ovidius’ University, 900470 Constanta, Romania
- Department of Neurology, ‘Sf. Ap. Andrei’ County Clinical Emergency Hospital of Constanta, 900591 Constanta, Romania
| | - Lavinia Florenta Muja
- Department of Neurology, General Medicine Faculty, ‘Ovidius’ University, 900470 Constanta, Romania
- Department of Neurology, ‘Sf. Ap. Andrei’ County Clinical Emergency Hospital of Constanta, 900591 Constanta, Romania
| | | | - Silvia Georgeta Petrov
- Doctoral School of the Faculty of Psychology and Educational Sciences within the University of Bucharest, 050663 Bucharest, Romania
| | - Anca Elena Gogu
- Department of Neurology, “Victor Babeș” University of Medicine and Pharmacy, 300041 Timișoara, Romania
- Centre for Cognitive Research in Neuropsychiatric Pathology (Neuropsy-Cog), Faculty of Medicine, “Victor Babeș” University of Medicine and Pharmacy, 300041 Timișoara, Romania
| | - Dragos Catalin Jianu
- Department of Neurology, “Victor Babeș” University of Medicine and Pharmacy, 300041 Timișoara, Romania
- Centre for Cognitive Research in Neuropsychiatric Pathology (Neuropsy-Cog), Faculty of Medicine, “Victor Babeș” University of Medicine and Pharmacy, 300041 Timișoara, Romania
| |
Collapse
|
45
|
Xanthopoulos A, Bourazana A, Giamouzis G, Skoularigki E, Dimos A, Zagouras A, Papamichalis M, Leventis I, Magouliotis DE, Triposkiadis F, Skoularigis J. COVID-19 and the heart. World J Clin Cases 2022; 10:9970-9984. [PMID: 36246800 PMCID: PMC9561576 DOI: 10.12998/wjcc.v10.i28.9970] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/23/2022] [Revised: 07/27/2022] [Accepted: 08/24/2022] [Indexed: 02/05/2023] Open
Abstract
An outbreak of coronavirus disease 2019 (COVID-19) occurred in December 2019 due to severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), which is a strain of SARS-CoV. Patients infected with the virus present a wide spectrum of manifestations ranging from mild flu-like symptoms, cough, fever and fatigue to severe lung injury, appearing as bilateral interstitial pneumonia or acute respiratory failure. Although SARS-CoV-2 infection predominantly offends the respiratory system, it has been associated with several cardiovascular complications as well. For example, patients with COVID-19 may either develop type 2 myocardial infarction due to myocardial oxygen demand and supply imbalance or acute coronary syndrome resulting from excessive inflammatory response to the primary infection. The incidence of COVID-19 related myocarditis is estimated to be accountable for an average of 7% of all COVID-19 related fatal cases, whereas heart failure (HF) may develop due to infiltration of the heart by inflammatory cells, destructive action of pro-inflammatory cytokines, micro-thrombosis and new onset or aggravated endothelial and respiratory failure. Lastly, SARS-CoV-2 can engender arrhythmias through direct myocardial damage causing acute myocarditis or through HF decompensation or secondary, through respiratory failure or severe respiratory distress syndrome. In this comprehensive review we summarize the COVID-19 related cardiovascular complications (acute coronary syndromes, myocarditis, HF, arrhythmias) and discuss the main underlying pathophysiological mechanisms.
Collapse
Affiliation(s)
- Andrew Xanthopoulos
- Department of Cardiology, University Hospital of Larissa, Larissa 41110, Greece
| | - Angeliki Bourazana
- Department of Cardiology, University Hospital of Larissa, Larissa 41110, Greece
| | - Grigorios Giamouzis
- Department of Cardiology, University Hospital of Larissa, Larissa 41110, Greece
| | | | - Apostolos Dimos
- Department of Cardiology, University Hospital of Larissa, Larissa 41110, Greece
| | - Alexandros Zagouras
- Department of Cardiology, University Hospital of Larissa, Larissa 41110, Greece
| | | | - Ioannis Leventis
- Department of Cardiology, University Hospital of Larissa, Larissa 41110, Greece
| | - Dimitrios E Magouliotis
- Department of Cardiothoracic Surgery, University of Thessaly, Larissa Biopolis, Larissa 41110, Greece
| | | | - John Skoularigis
- Department of Cardiology, University Hospital of Larissa, Larissa 41110, Greece
| |
Collapse
|
46
|
Coscas R, Dubosq M, Charton J, El Batti S, Gaudric J, Koskas F, Chiche L, Couture T, Davaine JM, Castier Y, Cerceau P, EL Hajjam M, Samb P, Beauchet A, Grimaldi L, Javerliat I, Goeau-Brissonniere O, Alsac JM, Onorati I, Martinod E, Desgranges P, Touma J, Cochennec F, Pellenc Q, Julia P, Coggia M. Thrombosis of Medium-Sized and Large Arteries During Covid-19 Infection: Results of the COVIVASC Study. Ann Vasc Surg 2022; 86:35-42. [PMID: 35780947 PMCID: PMC9242891 DOI: 10.1016/j.avsg.2022.04.055] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2022] [Revised: 03/15/2022] [Accepted: 04/15/2022] [Indexed: 01/31/2023]
Abstract
BACKGROUND COVID-19 infection is associated not only with venous thromboses but also with arterial thromboses (COV-ATs) in relation with an endothelial dysfunction, a coagulopathy and rhythm disorders. The incidence, the topography, and the prognosis of COV-ATs remain poorly known. The objective of this study was to report the overall experience of the Greater Paris University Hospitals (Assistance Publique - Hopitaux de Paris, AP-HP) during the first pandemic wave of COVID-19 infection. METHODS After approval by the ethics committee, a study using the AP-HP clinical data warehouse was carried out between March and May 2020. Overall, 124,609 patients had a polymerase chain reaction for COVID-19 in our hospitals, of which 25,345 were positive. From 20,710 exploitable stays, patients tested positive for COVID who presented an episode of acute COV-AT (except coronary and intracranial arteries) were selected on the basis of the French medical classification for clinical procedures codes. The data are presented as absolute values with percentages and/or means with standard deviation. RESULTS Over the studied period, 60 patients (aged 71±14 years, 42 men) presented a COV-AT at the time of their hospitalization, an incidence of 0.2%. The arterial complication occurred 3±7 days after the COVID infection and was inaugural in 30% of the cases (n = 18). The sites of COV-AT were the lower extremities (n = 35%, 58%), the abdominal aorta (n = 10%, 17%), the thoracic aorta (n = 7%, 12%), the upper limbs (n = 7%, 12%), the cerebral arteries (n = 7%, 12%), the digestive arteries (n = 6%, 10%), the renal arteries (n = 2%, 3%), and the ophthalmic artery (n = 1%, 2%). Multiple COV-ATs were observed in 13 patients (22%). At the time of diagnosis, 20 (33%) patients were in intensive care, including six (10%) patients who were intubated. On computed tomography angiography, COVID lesions were classified as moderate and severe in 25 (42%) and 21 (35%) cases, respectively. Revascularization was attempted in 27 patients (45%), by open surgery in 16 cases, using endovascular techniques in 8 cases and with a hybrid approach in three cases. Six patients (22%) required reinterventions. The duration of hospitalization was 12±9 days. Early mortality (in-hospital or at 30 days) was 30% (n = 18). Nine (15%) patients presented severe nonlethal ischemic complications. CONCLUSIONS Arterial involvement is rare during COVID-19 infection. The aorta and the arteries of the limbs are the privileged sites. The morbi-mortality of these patients is high. Future studies will have to determine if the systematization of anticoagulation therapy decreases the incidence and the severity of the condition.
Collapse
Affiliation(s)
- Raphael Coscas
- Department of Vascular Surgery, Ambroise Paré University Hospital, Assistance Publique-Hôpitaux de Paris, 92104 Boulogne-Billancourt, France,UMR 1018, Inserm-Paris11 - CESP, Versailles Saint-Quentin-en-Yvelines University, Paris-Saclay University, Paul Brousse Hospital, 94807 Villejuif, France,Correspondence to: Raphaël Coscas, Department of Vascular Surgery, Ambroise Paré University Hospital, 9 avenue Charles de Gaulle, 92104 Boulogne Cedex, France
| | - Maxime Dubosq
- Department of Vascular Surgery, Ambroise Paré University Hospital, Assistance Publique-Hôpitaux de Paris, 92104 Boulogne-Billancourt, France,UMR 1018, Inserm-Paris11 - CESP, Versailles Saint-Quentin-en-Yvelines University, Paris-Saclay University, Paul Brousse Hospital, 94807 Villejuif, France
| | - Johanna Charton
- Department of Vascular Surgery, Ambroise Paré University Hospital, Assistance Publique-Hôpitaux de Paris, 92104 Boulogne-Billancourt, France,UMR 1018, Inserm-Paris11 - CESP, Versailles Saint-Quentin-en-Yvelines University, Paris-Saclay University, Paul Brousse Hospital, 94807 Villejuif, France
| | - Salma El Batti
- Department of Cardiovascular Surgery, Hôpital Européen Georges Pompidou, Assistance Publique-Hôpitaux de Paris, 75015 Paris, France
| | - Julien Gaudric
- Department of Vascular and Endovascular Surgery–Tertiary Aortic Center, Pitié-Salpêtrière University Hospital, Assistance Publique-Hôpitaux de Paris and Sorbonne Médecine Universités, 75013 Paris, France
| | - Fabien Koskas
- Department of Vascular and Endovascular Surgery–Tertiary Aortic Center, Pitié-Salpêtrière University Hospital, Assistance Publique-Hôpitaux de Paris and Sorbonne Médecine Universités, 75013 Paris, France
| | - Laurent Chiche
- Department of Vascular and Endovascular Surgery–Tertiary Aortic Center, Pitié-Salpêtrière University Hospital, Assistance Publique-Hôpitaux de Paris and Sorbonne Médecine Universités, 75013 Paris, France
| | - Thibault Couture
- Department of Vascular and Endovascular Surgery–Tertiary Aortic Center, Pitié-Salpêtrière University Hospital, Assistance Publique-Hôpitaux de Paris and Sorbonne Médecine Universités, 75013 Paris, France
| | - Jean-Michel Davaine
- Department of Vascular and Endovascular Surgery–Tertiary Aortic Center, Pitié-Salpêtrière University Hospital, Assistance Publique-Hôpitaux de Paris and Sorbonne Médecine Universités, 75013 Paris, France
| | - Yves Castier
- Department of Vascular Surgery, Bichat Hospital, Assistance Publique-Hôpitaux de Paris, Paris, France and Paris-Diderot University, Paris, France
| | - Pierre Cerceau
- Department of Vascular Surgery, Bichat Hospital, Assistance Publique-Hôpitaux de Paris, Paris, France and Paris-Diderot University, Paris, France
| | - Mostafa EL Hajjam
- Department of Radiology, Ambroise Paré University Hospital, Assistance Publique-Hôpitaux de Paris, 92104 Boulogne-Billancourt, France
| | - Patricia Samb
- Clinical Research Unit, Ambroise Paré University Hospital, Assistance Publique-Hôpitaux de Paris, Boulogne-Billancourt, France,University of Versailles Saint-Quentin en Yvelines, Paris-Saclay University, Paris, France
| | - Alain Beauchet
- Clinical Research Unit, Ambroise Paré University Hospital, Assistance Publique-Hôpitaux de Paris, Boulogne-Billancourt, France,University of Versailles Saint-Quentin en Yvelines, Paris-Saclay University, Paris, France
| | - Lamiae Grimaldi
- Clinical Research Unit, Ambroise Paré University Hospital, Assistance Publique-Hôpitaux de Paris, Boulogne-Billancourt, France,University of Versailles Saint-Quentin en Yvelines, Paris-Saclay University, Paris, France
| | - Isabelle Javerliat
- Department of Vascular Surgery, Ambroise Paré University Hospital, Assistance Publique-Hôpitaux de Paris, 92104 Boulogne-Billancourt, France,UMR 1018, Inserm-Paris11 - CESP, Versailles Saint-Quentin-en-Yvelines University, Paris-Saclay University, Paul Brousse Hospital, 94807 Villejuif, France
| | - Olivier Goeau-Brissonniere
- Department of Vascular Surgery, Ambroise Paré University Hospital, Assistance Publique-Hôpitaux de Paris, 92104 Boulogne-Billancourt, France,UMR 1018, Inserm-Paris11 - CESP, Versailles Saint-Quentin-en-Yvelines University, Paris-Saclay University, Paul Brousse Hospital, 94807 Villejuif, France
| | - Jean-Marc Alsac
- Department of Cardiovascular Surgery, Hôpital Européen Georges Pompidou, Assistance Publique-Hôpitaux de Paris, 75015 Paris, France
| | - Ilaria Onorati
- Department of Thoracic and Vascular Surgery, Hôpitaux Universitaires Paris Seine-Saint-Denis, Hôpital Avicenne, Assistance Publique-Hôpitaux de Paris, and Université Paris 13, Sorbonne Paris Cité, UFR Santé, Médecine et Biologie Humaine, Bobigny, France
| | - Emmanuel Martinod
- Department of Thoracic and Vascular Surgery, Hôpitaux Universitaires Paris Seine-Saint-Denis, Hôpital Avicenne, Assistance Publique-Hôpitaux de Paris, and Université Paris 13, Sorbonne Paris Cité, UFR Santé, Médecine et Biologie Humaine, Bobigny, France
| | - Pascal Desgranges
- Department of Vascular Surgery, Henri Mondor University Hospital, Assistance Publique-Hôpitaux de Paris, Créteil, France,Paris-Est Créteil Faculty of Medicine, Créteil, France
| | - Joseph Touma
- Department of Vascular Surgery, Henri Mondor University Hospital, Assistance Publique-Hôpitaux de Paris, Créteil, France,Paris-Est Créteil Faculty of Medicine, Créteil, France
| | - Frédéric Cochennec
- Department of Vascular Surgery, Henri Mondor University Hospital, Assistance Publique-Hôpitaux de Paris, Créteil, France,Paris-Est Créteil Faculty of Medicine, Créteil, France
| | - Quentin Pellenc
- Department of Vascular Surgery, Bichat Hospital, Assistance Publique-Hôpitaux de Paris, Paris, France and Paris-Diderot University, Paris, France
| | - Pierre Julia
- Department of Cardiovascular Surgery, Hôpital Européen Georges Pompidou, Assistance Publique-Hôpitaux de Paris, 75015 Paris, France
| | - Marc Coggia
- Department of Vascular Surgery, Ambroise Paré University Hospital, Assistance Publique-Hôpitaux de Paris, 92104 Boulogne-Billancourt, France,UMR 1018, Inserm-Paris11 - CESP, Versailles Saint-Quentin-en-Yvelines University, Paris-Saclay University, Paul Brousse Hospital, 94807 Villejuif, France
| |
Collapse
|
47
|
Anwar MM, Sah R, Shrestha S, Ozaki A, Roy N, Fathah Z, Rodriguez-Morales AJ. Disengaging the COVID-19 Clutch as a Discerning Eye Over the Inflammatory Circuit During SARS-CoV-2 Infection. Inflammation 2022; 45:1875-1894. [PMID: 35639261 PMCID: PMC9153229 DOI: 10.1007/s10753-022-01674-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2022] [Revised: 03/29/2022] [Accepted: 04/18/2022] [Indexed: 01/08/2023]
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) causes the cytokine release syndrome (CRS) and leads to multiorgan dysfunction. Mitochondrial dynamics are fundamental to protect against environmental insults, but they are highly susceptible to viral infections. Defective mitochondria are potential sources of reactive oxygen species (ROS). Infection with SARS-CoV-2 damages mitochondria, alters autophagy, reduces nitric oxide (NO), and increases both nicotinamide adenine dinucleotide phosphate oxidases (NOX) and ROS. Patients with coronavirus disease 2019 (COVID-19) exhibited activated toll-like receptors (TLRs) and the Nucleotide-binding and oligomerization domain (NOD-), leucine-rich repeat (LRR-), pyrin domain-containing protein 3 (NLRP3) inflammasome. The activation of TLRs and NLRP3 by SARS-CoV-2 induces interleukin 6 (IL-6), IL-1β, IL-18, and lactate dehydrogenase (LDH). Herein, we outline the inflammatory circuit of COVID-19 and what occurs behind the scene, the interplay of NOX/ROS and their role in hypoxia and thrombosis, and the important role of ROS scavengers to reduce COVID-19-related inflammation.
Collapse
Affiliation(s)
- Mohammed Moustapha Anwar
- Department of Biotechnology, Institute of Graduate Studies and Research (IGSR), Alexandria University, Alexandria, Egypt.
| | - Ranjit Sah
- Tribhuvan University Institute of Medicine, Kathmandu, Nepal
| | - Sunil Shrestha
- Department of Pharmaceutical and Health Service Research, Nepal Health Research and Innovation Foundation, Lalitpur, Nepal
| | - Akihiko Ozaki
- Department of Breast Surgery, Jyoban Hospital of Tokiwa Foundation, Iwaki, Japan
- Medical Governance Research Institute, Tokyo, Japan
| | - Namrata Roy
- SRM University, SRM Nagar, Kattankulathur, Chengalpattu, Tamil Nadu, 603203, India
| | - Zareena Fathah
- Kings College London, London, UK
- College of Medicine and Health Sciences, United Arab University, Abu Dhabi, United Arab Emirates
| | - Alfonso J Rodriguez-Morales
- Grupo de Investigación Biomedicina, Faculty of Medicine, Fundacion Universitaria Autonoma de Las Americas, Pereira, Risaralda, Colombia.
- Institución Universitaria Visión de Las Americas, Pereira, Risaralda, Colombia.
- Faculty of Health Sciences, Universidad Cientifica del Sur, Lima, Peru.
- School of Medicine, Universidad Privada Franz Tamayo (UNIFRANZ), Cochabamba, Bolivia.
| |
Collapse
|
48
|
Bolla AM, Loretelli C, Montefusco L, Finzi G, Abdi R, Ben Nasr M, Lunati ME, Pastore I, Bonventre JV, Nebuloni M, Rusconi S, Santus P, Zuccotti G, Galli M, D’Addio F, Fiorina P. Inflammation and vascular dysfunction: The negative synergistic combination of diabetes and COVID-19. Diabetes Metab Res Rev 2022; 38:e3565. [PMID: 35830597 PMCID: PMC9349661 DOI: 10.1002/dmrr.3565] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/06/2021] [Revised: 05/31/2022] [Accepted: 06/03/2022] [Indexed: 01/08/2023]
Abstract
AIMS Several reports indicate that diabetes determines an increased mortality risk in patients with coronavirus disease 19 (COVID-19) and a good glycaemic control appears to be associated with more favourable outcomes. Evidence also supports that COVID-19 pneumonia only accounts for a part of COVID-19 related deaths. This disease is indeed characterised by abnormal inflammatory response and vascular dysfunction, leading to the involvement and failure of different systems, including severe acute respiratory distress syndrome, coagulopathy, myocardial damage and renal failure. Inflammation and vascular dysfunction are also well-known features of hyperglycemia and diabetes, making up the ground for a detrimental synergistic combination that could explain the increased mortality observed in hyperglycaemic patients. MATERIALS AND METHODS In this work, we conduct a narrative review on this intriguing connection. Together with this, we also present the clinical characteristics, outcomes, laboratory and histopathological findings related to this topic of a cohort of nearly 1000 subjects with COVID-19 admitted to a third-level Hospital in Milan. RESULTS We found an increased mortality in subjects with COVID-19 and diabetes, together with an altered inflammatory profile. CONCLUSIONS This may support the hypothesis that diabetes and COVID-19 meet at the crossroads of inflammation and vascular dysfunction. (ClinicalTrials.gov NCT04463849 and NCT04382794).
Collapse
Affiliation(s)
| | - Cristian Loretelli
- International Center for T1DPediatric Clinical Research Center Romeo ed Enrica InvernizziDepartment of Biomedical and Clinical Science L. SaccoUniversity of MilanMilanItaly
| | | | | | - Reza Abdi
- Nephrology DivisionBrigham and Women's HospitalHarvard Medical SchoolBostonMassachusettsUSA
| | - Moufida Ben Nasr
- International Center for T1DPediatric Clinical Research Center Romeo ed Enrica InvernizziDepartment of Biomedical and Clinical Science L. SaccoUniversity of MilanMilanItaly
- Nephrology DivisionBoston Children's HospitalHarvard Medical SchoolBostonMassachusettsUSA
| | | | - Ida Pastore
- Division of EndocrinologyASST Fatebenefratelli‐SaccoMilanItaly
| | - Joseph V. Bonventre
- Nephrology DivisionBrigham and Women's HospitalHarvard Medical SchoolBostonMassachusettsUSA
| | - Manuela Nebuloni
- Pathology UnitASST Fatebenefratelli‐SaccoMilanItaly
- Department of Biomedical and Clinical Sciences L. SaccoUniversity of MilanMilanItaly
| | - Stefano Rusconi
- Department of Biomedical and Clinical Sciences L. SaccoUniversity of MilanMilanItaly
| | - Pierachille Santus
- Department of Biomedical and Clinical Sciences L. SaccoUniversity of MilanMilanItaly
- Division of Respiratory DiseasesASST Fatebenefratelli‐SaccoMilanItaly
| | - Gianvincenzo Zuccotti
- Pediatric Clinical Research Center Romeo ed Enrica InvernizziDepartment of Biomedical and Clinical Science L. SaccoUniversity of MilanMilanItaly
- Department of Pediatrics“V. Buzzi” Children's HospitalMilanItaly
| | - Massimo Galli
- Department of Biomedical and Clinical Sciences L. SaccoUniversity of MilanMilanItaly
- III Division of Infectious DiseasesLuigi Sacco HospitalASST Fatebenefratelli‐SaccoMilanItaly
| | - Francesca D’Addio
- Division of EndocrinologyASST Fatebenefratelli‐SaccoMilanItaly
- International Center for T1DPediatric Clinical Research Center Romeo ed Enrica InvernizziDepartment of Biomedical and Clinical Science L. SaccoUniversity of MilanMilanItaly
| | - Paolo Fiorina
- Division of EndocrinologyASST Fatebenefratelli‐SaccoMilanItaly
- International Center for T1DPediatric Clinical Research Center Romeo ed Enrica InvernizziDepartment of Biomedical and Clinical Science L. SaccoUniversity of MilanMilanItaly
- Nephrology DivisionBoston Children's HospitalHarvard Medical SchoolBostonMassachusettsUSA
| |
Collapse
|
49
|
Ramakrishnan N, Ramasubban S, Hegde A, Govil D. Approach to Thromboprophylaxis for Prevention of Venous Thromboembolism in COVID-19: Global Updates and Clinical Insights from India. Clin Pract 2022; 12:766-781. [PMID: 36286066 PMCID: PMC9601217 DOI: 10.3390/clinpract12050080] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2022] [Revised: 09/08/2022] [Accepted: 09/13/2022] [Indexed: 11/30/2022] Open
Abstract
Venous thromboembolism (VTE) frequently occurs in patients with coronavirus disease-19 (COVID-19) and is associated with increased mortality. Several global guidelines recommended prophylactic-intensity anticoagulation rather than intermediate-intensity or therapeutic-intensity anticoagulation for patients with COVID-19-related acute or critical illness without suspected or confirmed VTE. Even though standard doses of thromboprophylaxis are received, many cases of thrombotic complications are reported; hence, appropriate and adequate thromboprophylaxis is critical for the prevention of VTE in COVID-19. In spite of an increased prevalence of VTE in Indian patients, sufficient data on patient characteristics, diagnosis, and therapeutic approach for VTE in COVID is lacking. In this article, we review the available global literature (search conducted up to 31 May 2021) and provide clinical insights into our approach towards managing VTE in patients with COVID-19. Furthermore, in this review, we summarize the incidence and risk factors for VTE with emphasis on the thromboprophylaxis approach in hospitalized patients and special populations with COVID-19 and assess clinical implications in the Indian context.
Collapse
Affiliation(s)
- Nagarajan Ramakrishnan
- Department of Critical Care Medicine, Apollo Hospitals, Chennai 600006, Tamil Nadu, India
| | - Suresh Ramasubban
- Department of Critical Care, Apollo Gleneagles Hospital, Kolkata 700054, West Bengal, India
| | - Ashit Hegde
- Department of Critical Care and Medicine, PD Hinduja National Hospital and Medical Research Centre, Mumbai 400016, Maharashtra, India
| | - Deepak Govil
- Institute of Critical Care and Anesthesiology, Medanta The Medicity, Gurgaon 122006, Haryana, India
| |
Collapse
|
50
|
Coagulopathy during COVID-19 infection: a brief review. Clin Exp Med 2022:10.1007/s10238-022-00891-4. [PMID: 36121504 PMCID: PMC9483403 DOI: 10.1007/s10238-022-00891-4] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2022] [Accepted: 09/08/2022] [Indexed: 11/03/2022]
Abstract
The COVID-19 pandemic caused by SARS-CoV-2 continues to spread rapidly due to its virulence and ability to be transmitted by asymptomatic infected persons. If they are present, the symptoms of COVID-19 may include rhinorrhea (runny nose), headache, cough, and fever. Up to 5% of affected persons may experience more severe COVID-19 illness, including severe coagulopathy, acute respiratory distress syndrome (ARDS) characterized by respiratory failure that requires supplementary oxygen and mechanical ventilation, and multi-organ failure. Interestingly, clinical evidence has highlighted the distinction between COVID-19-associated coagulopathy (CAC) and disseminated intravascular coagulation (DIC). Patients with CAC exhibit different laboratory values than DIC patients for activated partial thromboplastin time (aPTT) and prothrombin time (PT) which may be normal or shortened, varying platelet counts, altered red blood cell morphology, unique bleeding complications, a lack of schistocytes in the peripheral blood, and no decrease in fibrinogen levels. In this review, we consider the search for 1) laboratory results that can diagnose or predict development of CAC, including serum levels of D-dimers, fibrinogen, interleukin-6 (IL-6) and the growth factor angiopoietin-2 (Ang-2), 2) mechanisms of CAC induction, and 3) novel therapeutic regimens that will successfully treat COVID-19 before development of CAC.
Collapse
|