51
|
Fixed-Dose Ultrasound-Assisted Catheter-Directed Thrombolysis for Acute Pulmonary Embolism Associated with COVID-19. Viruses 2022; 14:v14081606. [PMID: 35893672 PMCID: PMC9394471 DOI: 10.3390/v14081606] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2022] [Accepted: 07/20/2022] [Indexed: 12/07/2022] Open
Abstract
Background. Fixed-dose ultrasound-assisted catheter-directed thrombolysis (USAT) rapidly improves hemodynamic parameters and reverses right ventricular dysfunction caused by acute pulmonary embolism (PE). The effectiveness of USAT for acute PE associated with coronavirus disease 2019 (COVID-19) is unknown. Methods and results. The study population of this cohort study consisted of 36 patients with an intermediate-high- or high-risk acute PE treated with a fixed low-dose USAT protocol (r-tPA 10–20 mg/15 h). Of these, 9 patients tested positive for COVID-19 and were age–sex-matched to 27 patients without COVID-19. The USAT protocol included, beyond the infusion of recombinant tissue plasminogen activator, anti-Xa-activity-adjusted unfractionated heparin therapy (target 0.3–0.7 U/mL). The study outcomes were the invasively measured mean pulmonary arterial pressure (mPAP) before and at completion of USAT, and the National Early Warning Score (NEWS), according to which more points indicate more severe hemodynamic impairment. Twenty-four (66.7%) patients were men; the mean age was 67 ± 14 years. Mean ± standard deviation mPAP decreased from 32.3 ± 8.3 to 22.4 ± 7.0 mmHg among COVID-19 patients and from 35.4 ± 9.7 to 24.6 ± 7.0 mmHg among unexposed, with no difference in the relative improvement between groups (p = 0.84). Within 12 h of USAT start, the median NEWS decreased from six (Q1–Q3: 4–8) to three (Q1–Q3: 2–4) points among COVID-19 patients and from four (Q1–Q3: 2–6) to two (Q1–Q3: 2–3) points among unexposed (p = 0.29). One COVID-19 patient died due to COVID-19-related complications 14 days after acute PE. No major bleeding events occurred. Conclusions. Among patients with COVID-19-associated acute PE, mPAP rapidly decreased during USAT with a concomitant progressive improvement of the NEWS. The magnitude of mPAP reduction was similar in patients with and without COVID-19.
Collapse
|
52
|
Dmytriw AA, Ghozy S, Sweid A, Piotin M, Bekelis K, Sourour N, Raz E, Vela-Duarte D, Linfante I, Dabus G, Kole M, Martínez-Galdámez M, Nimjee SM, Lopes DK, Hassan AE, Kan P, Ghorbani M, Levitt MR, Escalard S, Missios S, Shapiro M, Clarençon F, Elhorany M, Tahir RA, Youssef PP, Pandey AS, Starke RM, El Naamani K, Abbas R, Mansour OY, Galvan J, Billingsley JT, Mortazavi A, Walker M, Dibas M, Settecase F, Heran MKS, Kuhn AL, Puri AS, Menon BK, Sivakumar S, Mowla A, D'Amato S, Zha AM, Cooke D, Vranic JE, Regenhardt RW, Rabinov JD, Stapleton CJ, Goyal M, Wu H, Cohen J, Turkel-Parella D, Xavier A, Waqas M, Tutino V, Siddiqui A, Gupta G, Nanda A, Khandelwal P, Tiu C, Portela PC, Perez de la Ossa N, Urra X, de Lera M, Arenillas JF, Ribo M, Requena M, Piano M, Pero G, De Sousa K, Al-Mufti F, Hashim Z, Nayak S, Renieri L, Du R, Aziz-Sultan MA, Liebeskind D, Nogueira RG, Abdalkader M, Nguyen TN, Vigilante N, Siegler JE, Grossberg JA, Saad H, Gooch MR, Herial NA, Rosenwasser RH, Tjoumakaris S, Patel AB, Tiwari A, Jabbour P. International Controlled Study of Revascularization and Outcomes Following COVID-Positive Mechanical Thrombectomy. Eur J Neurol 2022; 29:3273-3287. [PMID: 35818781 PMCID: PMC9349405 DOI: 10.1111/ene.15493] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2022] [Revised: 06/24/2022] [Accepted: 06/29/2022] [Indexed: 11/29/2022]
Abstract
Background and purpose Previous studies suggest that mechanisms and outcomes in patients with COVID‐19‐associated stroke differ from those in patients with non‐COVID‐19‐associated strokes, but there is limited comparative evidence focusing on these populations. The aim of this study, therefore, was to determine if a significant association exists between COVID‐19 status with revascularization and functional outcomes following thrombectomy for large vessel occlusion (LVO), after adjustment for potential confounding factors. Methods A cross‐sectional, international multicenter retrospective study was conducted in consecutively admitted COVID‐19 patients with concomitant acute LVO, compared to a control group without COVID‐19. Data collected included age, gender, comorbidities, clinical characteristics, details of the involved vessels, procedural technique, and various outcomes. A multivariable‐adjusted analysis was conducted. Results In this cohort of 697 patients with acute LVO, 302 had COVID‐19 while 395 patients did not. There was a significant difference (p < 0.001) in the mean age (in years) and gender of patients, with younger patients and more males in the COVID‐19 group. In terms of favorable revascularization (modified Thrombolysis in Cerebral Infarction [mTICI] grade 3), COVID‐19 was associated with lower odds of complete revascularization (odds ratio 0.33, 95% confidence interval [CI] 0.23–0.48; p < 0.001), which persisted on multivariable modeling with adjustment for other predictors (adjusted odds ratio 0.30, 95% CI 0.12–0.77; p = 0.012). Moreover, endovascular complications, in‐hospital mortality, and length of hospital stay were significantly higher among COVID‐19 patients (p < 0.001). Conclusion COVID‐19 was an independent predictor of incomplete revascularization and poor functional outcome in patients with stroke due to LVO. Furthermore, COVID‐19 patients with LVO were more often younger and had higher morbidity/mortality rates.
Collapse
Affiliation(s)
- Adam A Dmytriw
- Neuroendovascular Program, Mass General Brigham Partners, Harvard Medical School, Boston, MA, USA.,Neuroradiology & Neurosurgery Services, Brigham & Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Sherief Ghozy
- Neuroradiology & Neurosurgery Services, Brigham & Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Ahmad Sweid
- Department of Neurological Surgery, Thomas Jefferson University Hospital, Philadelphia, Pennsylvania, USA
| | - Michel Piotin
- Department of Interventional Neuroradiology, Rothschild Foundation Hospital, Paris, France
| | - Kimon Bekelis
- Department of Neurosurgery, Good Samaritan Hospital Medical Center, West Islip, New York, USA
| | - Nader Sourour
- Department of Interventional Neuroradiology, Pitié-Salpêtrière Hospital, Paris, France
| | - Eytan Raz
- Department of Radiology, New York University Langone Medical Center, New York, New York, USA
| | - Daniel Vela-Duarte
- Department of Interventional Neuroradiology & Neuroendovascular Surgery, Miami Cardiac and Vascular Institute, Baptist Hospital of Miami, Florida, USA
| | - Italo Linfante
- Department of Interventional Neuroradiology & Neuroendovascular Surgery, Miami Cardiac and Vascular Institute, Baptist Hospital of Miami, Florida, USA
| | - Guilherme Dabus
- Department of Interventional Neuroradiology & Neuroendovascular Surgery, Miami Cardiac and Vascular Institute, Baptist Hospital of Miami, Florida, USA
| | - Max Kole
- Department of Neurosurgery, Henry Ford Hospital, Michigan, USA
| | - Mario Martínez-Galdámez
- Department of Interventional Neuroradiology, Hospital Clinico Universitario de Valladolid, Spain
| | - Shahid M Nimjee
- Department of Neurosurgery, The Ohio State University Wexner Medical Center, Columbus, Ohio, USA
| | - Demetrius K Lopes
- Department of Neurosurgery, Advocate Aurora Health, Chicago, Illinois, USA
| | - Ameer E Hassan
- Department of Neuroscience, Valley Baptist Medical Center/University of Texas Rio Grande Valley, Harlingen, Texas, USA
| | - Peter Kan
- Department of Neurosurgery, UTMB, Houston, Texas, USA
| | | | - Michael R Levitt
- Departments of Neurological Surgery, Radiology, Mechanical Engineering, and Stroke & Applied Neuroscience Center, University of Washington, Seattle, Washington, USA
| | - Simon Escalard
- Department of Interventional Neuroradiology, Rothschild Foundation Hospital, Paris, France
| | - Symeon Missios
- Department of Neurosurgery, Good Samaritan Hospital Medical Center, West Islip, New York, USA
| | - Maksim Shapiro
- Department of Radiology, New York University Langone Medical Center, New York, New York, USA
| | - Fréderic Clarençon
- Department of Interventional Neuroradiology, Pitié-Salpêtrière Hospital, Paris, France
| | - Mahmoud Elhorany
- Department of Interventional Neuroradiology, Pitié-Salpêtrière Hospital, Paris, France
| | - Rizwan A Tahir
- Department of Neurosurgery, Henry Ford Hospital, Michigan, USA
| | - Patrick P Youssef
- Department of Neurosurgery, The Ohio State University Wexner Medical Center, Columbus, Ohio, USA
| | - Aditya S Pandey
- Department of Neurosurgery, University of Michigan, Ann Arbor, Michigan, USA
| | - Robert M Starke
- Department of Neurosurgery & Neuroradiology, University of Miami & Jackson Memorial Hospital, Miami, Florida, USA
| | - Kareem El Naamani
- Department of Neurological Surgery, Thomas Jefferson University Hospital, Philadelphia, Pennsylvania, USA
| | - Rawad Abbas
- Department of Neurological Surgery, Thomas Jefferson University Hospital, Philadelphia, Pennsylvania, USA
| | | | - Jorge Galvan
- Department of Interventional Neuroradiology, Hospital Clinico Universitario de Valladolid, Spain
| | | | - Abolghasem Mortazavi
- Department of Neuroscience, Valley Baptist Medical Center/University of Texas Rio Grande Valley, Harlingen, Texas, USA
| | - Melanie Walker
- Departments of Neurological Surgery, Radiology, Mechanical Engineering, and Stroke & Applied Neuroscience Center, University of Washington, Seattle, Washington, USA
| | - Mahmoud Dibas
- Neuroradiology & Neurosurgery Services, Brigham & Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Fabio Settecase
- Division of Neuroradiology, Vancouver General Hospital, University of British Columbia, Vancouver, British Columbia, Canada
| | - Manraj K S Heran
- Division of Neuroradiology, Vancouver General Hospital, University of British Columbia, Vancouver, British Columbia, Canada
| | - Anna L Kuhn
- Division of Neurointerventional Radiology, Department of Radiology, UMass Memorial Medical Center, Worcester, Massachusetts, USA
| | - Ajit S Puri
- Division of Neurointerventional Radiology, Department of Radiology, UMass Memorial Medical Center, Worcester, Massachusetts, USA
| | - Bijoy K Menon
- Calgary Stroke Program, Cumming School of Medicine, Calgary, Alberta, Canada
| | - Sanjeev Sivakumar
- Department of Medicine (Neurology), Prisma Health Upstate, USC, Greenville, South Carolina, USA
| | - Ashkan Mowla
- Department of Neurological Surgery, University of Southern California, Los Angeles, California, USA
| | - Salvatore D'Amato
- Neuroendovascular Program, Mass General Brigham Partners, Harvard Medical School, Boston, MA, USA
| | - Alicia M Zha
- Department of Neurology, UT Health Science Center, Houston, Texas, USA
| | - Daniel Cooke
- Department of Neurointerventional Radiology, San Francisco General Hospital, San Francisco, California, USA
| | - Justin E Vranic
- Neuroendovascular Program, Mass General Brigham Partners, Harvard Medical School, Boston, MA, USA
| | - Robert W Regenhardt
- Neuroendovascular Program, Mass General Brigham Partners, Harvard Medical School, Boston, MA, USA
| | - James D Rabinov
- Neuroendovascular Program, Mass General Brigham Partners, Harvard Medical School, Boston, MA, USA
| | - Christopher J Stapleton
- Neuroendovascular Program, Mass General Brigham Partners, Harvard Medical School, Boston, MA, USA
| | - Mayank Goyal
- Calgary Stroke Program, Cumming School of Medicine, Calgary, Alberta, Canada
| | - Hannah Wu
- Department of Neurology, Brookdale University Hospital, Brooklyn, New York, USA.,Department of Neurology, Jamaica Medical Center, Richmond Hill, New York, USA.,Department of Neurology, NYU Lutheran Hospital, Brooklyn, New York, USA
| | - Jake Cohen
- Department of Neurology, Brookdale University Hospital, Brooklyn, New York, USA.,Department of Neurology, Jamaica Medical Center, Richmond Hill, New York, USA.,Department of Neurology, NYU Lutheran Hospital, Brooklyn, New York, USA
| | - David Turkel-Parella
- Department of Neurology, Brookdale University Hospital, Brooklyn, New York, USA.,Department of Neurology, Jamaica Medical Center, Richmond Hill, New York, USA.,Department of Neurology, NYU Lutheran Hospital, Brooklyn, New York, USA
| | - Andrew Xavier
- Department of Neurology, Sinai Grace Hospital, Detroit, Michigan, USA.,Department of Neurology, St. Joseph Mercy Health, Ann Arbor, Michigan, USA
| | - Muhammad Waqas
- Department of Neurosurgery, University of New York at Buffalo, Buffalo, New York, USA
| | - Vincent Tutino
- Department of Neurosurgery, University of New York at Buffalo, Buffalo, New York, USA
| | - Adnan Siddiqui
- Department of Neurosurgery, University of New York at Buffalo, Buffalo, New York, USA
| | - Gaurav Gupta
- Department of Neurology, Robert Wood Johnson University Hospital, New Brunswick, New Jersey, USA
| | - Anil Nanda
- Department of Neurology, Robert Wood Johnson University Hospital, New Brunswick, New Jersey, USA
| | - Priyank Khandelwal
- Department of Neurology, Robert Wood Johnson University Hospital, New Brunswick, New Jersey, USA
| | - Cristina Tiu
- Department of Neurology, University Emergency Hospital Bucharest, Bucharest, Romania; "Carol Davila" University of Medicine and Pharmacy, Bucharest, Romania
| | - Pere C Portela
- Department of Neurology, Hospital Universitari, Bellvitge, Barcelona, Spain
| | - Natalia Perez de la Ossa
- Stroke Unit, Neuroscience Department, Hospital Universitari Germans Trias i Pujol, Badalona, Barcelona, Spain
| | - Xabier Urra
- Department of Neurology, Hospital Clínic, Barcelona, Spain
| | - Mercedes de Lera
- Department of Neurology, Hospital Clínico Universitario, Valladolid, Spain
| | - Juan F Arenillas
- Department of Neurology, Hospital Clínico Universitario, Valladolid, Spain
| | - Marc Ribo
- Stroke Unit, Department of Neurology, Vall d'Hebron Research Institute, Barcelona, Spain; Departament de Medicina, Universitat Autónoma de Barcelona, Barcelona, Spain
| | - Manuel Requena
- Stroke Unit, Department of Neurology, Vall d'Hebron Research Institute, Barcelona, Spain; Departament de Medicina, Universitat Autónoma de Barcelona, Barcelona, Spain
| | - Mariangela Piano
- Department of Neuroradiology, ASST Grande Ospedale Metropolitano Niguarda, Milan, Italy
| | - Guglielmo Pero
- Department of Neuroradiology, ASST Grande Ospedale Metropolitano Niguarda, Milan, Italy
| | - Keith De Sousa
- Department of Neurology, Eastern Region, Northwell Health, Long Island, New York, New York, USA
| | - Fawaz Al-Mufti
- Department of Neurology, Radiology, and Neurosurgery, Westchester Medical Center at NY Medical College, Valhalla, New York, USA
| | - Zafar Hashim
- Department of Radiology, University Hospital of North Midlands, Stoke-on-Trent, United Kingdom
| | - Sanjeev Nayak
- Department of Radiology, University Hospital of North Midlands, Stoke-on-Trent, United Kingdom
| | - Leonardo Renieri
- Department of Radiology, Neurovascular Unit, Careggi University Hospital, Florence, Italy
| | - Rose Du
- Neuroradiology & Neurosurgery Services, Brigham & Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Mohamed A Aziz-Sultan
- Neuroradiology & Neurosurgery Services, Brigham & Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - David Liebeskind
- Department of Neurology, Ronald Reagan UCLA Medical Center, Los Angeles, USA
| | - Raul G Nogueira
- Department of Neurology, Marcus Stroke and Neuroscience Center, Grady Memorial Hospital, Emory University School of Medicine, Atlanta, GA, USA
| | - Mohamad Abdalkader
- Departments of Neurology and Radiology, Boston Medical Center, Boston University School of Medicine, Massachusetts, USA
| | - Thanh N Nguyen
- Departments of Neurology and Radiology, Boston Medical Center, Boston University School of Medicine, Massachusetts, USA
| | - Nicholas Vigilante
- Cooper Neurological Institute, Cooper University Hospital, Camden, New Jersey, USA
| | - James E Siegler
- Cooper Neurological Institute, Cooper University Hospital, Camden, New Jersey, USA
| | | | - Hassan Saad
- Department of Neurosurgery, Emory University, Atlanta, Georgia, USA
| | - Michael R Gooch
- Department of Neurological Surgery, Thomas Jefferson University Hospital, Philadelphia, Pennsylvania, USA
| | - Nabeel A Herial
- Department of Neurological Surgery, Thomas Jefferson University Hospital, Philadelphia, Pennsylvania, USA
| | - Robert H Rosenwasser
- Department of Neurological Surgery, Thomas Jefferson University Hospital, Philadelphia, Pennsylvania, USA
| | - Stavropoula Tjoumakaris
- Department of Neurological Surgery, Thomas Jefferson University Hospital, Philadelphia, Pennsylvania, USA
| | - Aman B Patel
- Neuroendovascular Program, Mass General Brigham Partners, Harvard Medical School, Boston, MA, USA
| | - Ambooj Tiwari
- Department of Neurology, Brookdale University Hospital, Brooklyn, New York, USA.,Department of Neurology, Jamaica Medical Center, Richmond Hill, New York, USA.,Department of Neurology, NYU Lutheran Hospital, Brooklyn, New York, USA
| | - Pascal Jabbour
- Department of Neurological Surgery, Thomas Jefferson University Hospital, Philadelphia, Pennsylvania, USA
| | | |
Collapse
|
53
|
Abdel Moneim A, Radwan MA, Yousef AI. COVID-19 and cardiovascular disease: manifestations, pathophysiology, vaccination, and long-term implication. Curr Med Res Opin 2022; 38:1071-1079. [PMID: 35575011 DOI: 10.1080/03007995.2022.2078081] [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] [Indexed: 12/15/2022]
Abstract
The coronavirus disease 2019 (COVID-19) pandemic is caused by a new coronavirus family member, severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) and is linked with many disease manifestations in multiple organ systems on top of pulmonary manifestations. COVID-19 is also accompanied by several cardiovascular pathologies including myocarditis, acute myocardial infarction, stress cardiomyopathy, arterial and venous thromboembolism, pericarditis, and arrhythmias. The pathophysiological mechanisms explaining these clinical symptoms are multifactorial including systemic inflammation (cytokine storm), coagulopathy, direct viral invasion through angiotensin-converting enzyme 2, hypoxemia, electrolyte imbalance, and fever. Several case reports have shown the development of an unusual cardiovascular event after receiving SARS-CoV-2 vaccines. The current article aimed to review cardiovascular involvement in the COVID-19 pandemic with respect to clinical features, pathogenesis, long-term effects, and the adverse effects of treatments and vaccines based on the latest evidence.
Collapse
Affiliation(s)
- Adel Abdel Moneim
- Molecular Physiology Division, Faculty of Science, Beni-Suef University, Egypt
| | - Marwa A Radwan
- Molecular Physiology Division, Faculty of Science, Beni-Suef University, Egypt
| | - Ahmed I Yousef
- Molecular Physiology Division, Faculty of Science, Beni-Suef University, Egypt
| |
Collapse
|
54
|
Corey KM, Olson LB, Naqvi IA, Morrison SA, Davis C, Nimjee SM, Que LG, Bachelder RE, Kraft BD, Chen L, Nair SK, Levy JH, Sullenger BA. Suppression of Fibrinolysis and Hypercoagulability, Severity of Hypoxemia, and Mortality in COVID-19 Patients: A Retrospective Cohort Study. Anesthesiology 2022; 137:67-78. [PMID: 35412597 PMCID: PMC9250792 DOI: 10.1097/aln.0000000000004239] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
BACKGROUND COVID-19 causes hypercoagulability, but the association between coagulopathy and hypoxemia in critically ill patients has not been thoroughly explored. This study hypothesized that severity of coagulopathy would be associated with acute respiratory distress syndrome severity, major thrombotic events, and mortality in patients requiring intensive care unit-level care. METHODS Viscoelastic testing by rotational thromboelastometry and coagulation factor biomarker analyses were performed in this prospective observational cohort study of critically ill COVID-19 patients from April 2020 to October 2020. Statistical analyses were performed to identify significant coagulopathic biomarkers such as fibrinolysis-inhibiting plasminogen activator inhibitor 1 and their associations with clinical outcomes such as mortality, extracorporeal membrane oxygenation requirement, occurrence of major thrombotic events, and severity of hypoxemia (arterial partial pressure of oxygen/fraction of inspired oxygen categorized into mild, moderate, and severe per the Berlin criteria). RESULTS In total, 53 of 55 (96%) of the cohort required mechanical ventilation and 9 of 55 (16%) required extracorporeal membrane oxygenation. Extracorporeal membrane oxygenation-naïve patients demonstrated lysis indices at 30 min indicative of fibrinolytic suppression on rotational thromboelastometry. Survivors demonstrated fewer procoagulate acute phase reactants, such as microparticle-bound tissue factor levels (odds ratio, 0.14 [0.02, 0.99]; P = 0.049). Those who did not experience significant bleeding events had smaller changes in ADAMTS13 levels compared to those who did (odds ratio, 0.05 [0, 0.7]; P = 0.026). Elevations in plasminogen activator inhibitor 1 (odds ratio, 1.95 [1.21, 3.14]; P = 0.006), d-dimer (odds ratio, 3.52 [0.99, 12.48]; P = 0.05), and factor VIII (no clot, 1.15 ± 0.28 vs. clot, 1.42 ± 0.31; P = 0.003) were also demonstrated in extracorporeal membrane oxygenation-naïve patients who experienced major thrombotic events. Plasminogen activator inhibitor 1 levels were significantly elevated during periods of severe compared to mild and moderate acute respiratory distress syndrome (severe, 44.2 ± 14.9 ng/ml vs. mild, 31.8 ± 14.7 ng/ml and moderate, 33.1 ± 15.9 ng/ml; P = 0.029 and 0.039, respectively). CONCLUSIONS Increased inflammatory and procoagulant markers such as plasminogen activator inhibitor 1, microparticle-bound tissue factor, and von Willebrand factor levels are associated with severe hypoxemia and major thrombotic events, implicating fibrinolytic suppression in the microcirculatory system and subsequent micro- and macrovascular thrombosis in severe COVID-19. EDITOR’S PERSPECTIVE
Collapse
Affiliation(s)
- Kristin M. Corey
- Department of Medicine, Duke University School of Medicine, Durham, NC 27710
| | - Lyra B. Olson
- Duke Medical Scientist Training Program, Department of Pharmacology and Cancer Biology, Duke University School of Medicine, Durham, NC 27710
| | - Ibtehaj A. Naqvi
- Department of Surgery, Duke University School of Medicine, Durham, NC 27710
| | - Sarah A Morrison
- Department of Surgery, Duke University School of Medicine, Durham, NC 27710
| | - Connor Davis
- Duke Institute for Health Innovation, Duke University School of Medicine, Durham, NC 27710
| | - Shahid M. Nimjee
- Department of Neurosurgery, The Ohio State University Medical Center, Columbus, OH 43203
| | - Loretta G. Que
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, Duke University Medical Center, Durham, NC 27710
| | - Robin E. Bachelder
- Department of Surgery, Duke University School of Medicine, Durham, NC 27710
| | - Bryan D. Kraft
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, Duke University Medical Center, Durham, NC 27710
| | - Lingye Chen
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, Duke University Medical Center, Durham, NC 27710
| | - Smita K. Nair
- Department of Surgery, Duke University School of Medicine, Durham, NC 27710
| | - Jerrold H. Levy
- Departments of Anesthesiology, Critical Care, and Surgery, Duke University School of Medicine, Durham, NC 27710
| | - Bruce A. Sullenger
- Department of Surgery, Duke University School of Medicine, Durham, NC 27710
| |
Collapse
|
55
|
Chowdary P. COVID-19 coagulopathy - what should we treat? Exp Physiol 2022; 107:749-758. [PMID: 35733235 PMCID: PMC9328279 DOI: 10.1113/ep089404] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Accepted: 05/31/2022] [Indexed: 12/15/2022]
Abstract
NEW FINDINGS What is the topic of this review? Overview of the coagulation abnormalities, including elevated D-dimers widely reported with COVID-19, often labelled as COVID coagulopathy. What advances does it highlight? The review highlights the changes in bronchoalveolar haemostasis due to apoptosis of alveolar cells, which contributes to acute lung injury and acute respiratory distress syndrome; the pathophysiological mechanisms, including endothelial dysfunction and damage responsible for thrombosis of pulmonary microcirculation and potential contribution to the hypoxaemia of COVID-19 acute lung injury; and changes in coagulation proteins responsible for the hypercoagulability and increased risk of thrombosis in other venous and arterial beds. The rationale for anticoagulation and fibrinolytic therapies is detailed, and potential confounders that might have led to less than expected improvement in the various randomised controlled trials are considered. ABSTRACT Coronavirus disease 19 (COVID-19) causes acute lung injury with diffuse alveolar damage, alveolar-capillary barrier disruption, thrombin generation and alveolar fibrin deposition. Clinically, hypoxaemia is associated with preserved lung compliance early in the disease, suggesting the lack of excessive fluid accumulation typical of other lung injuries. Notably, autopsy studies demonstrate infection of the endothelium with extensive capillary thrombosis distinct from the embolic thrombi in pulmonary arteries. The inflammatory thrombosis in pulmonary vasculature secondary to endothelial infection and dysfunction appears to contribute to hypoxaemia. This is associated with elevated D-dimers and acquired hypercoagulability with an increased risk of deep vein thrombosis. Hypercoagulability is secondary to elevated plasma tissue factor levels, von Willebrand factor, fibrinogen, reduced ADAMTS-13 with platelet activation and inhibition of fibrinolysis. Multi-platform randomised controlled studies of systemic therapeutic anticoagulation with unfractionated and low molecular mass heparins demonstrated a survival benefit over standard care with full-dose anticoagulation in patients with non-severe disease who require supplemental oxygen, but not in severe disease requiring ventilatory support. Late intervention and the heterogeneous nature of enrolled patients can potentially explain the apparent lack of benefit in severe disease. Improvement in oxygenation has been demonstrated with intravenous fibrinolytics in small studies. Inhaled anticoagulants, thrombolytic agents and non-specific proteolytic drugs in clinical trials for decreasing alveolar fibrin deposition might benefit early disease. Essentially, COVID-19 is a multi-system disorder with pulmonary vascular inflammatory thrombosis that requires an interdisciplinary approach to combination therapies addressing both inflammation and intravascular thrombosis or alveolar fibrin deposits to improve outcomes.
Collapse
Affiliation(s)
- Pratima Chowdary
- Katharine Dormandy Haemophilia and Thrombosis Centre, Royal Free Hospital, London, UK.,Cancer Institute, University College London, London, UK
| |
Collapse
|
56
|
Jabbour P, Dmytriw AA, Sweid A, Piotin M, Bekelis K, Sourour N, Raz E, Linfante I, Dabus G, Kole M, Martínez-Galdámez M, Nimjee SM, Lopes DK, Hassan AE, Kan P, Ghorbani M, Levitt MR, Escalard S, Missios S, Shapiro M, Clarençon F, Elhorany M, Vela-Duarte D, Tahir RA, Youssef PP, Pandey AS, Starke RM, El Naamani K, Abbas R, Hammoud B, Mansour OY, Galvan J, Billingsley JT, Mortazavi A, Walker M, Dibas M, Settecase F, Heran MKS, Kuhn AL, Puri AS, Menon BK, Sivakumar S, Mowla A, D'Amato S, Zha AM, Cooke D, Goyal M, Wu H, Cohen J, Turkel-Parrella D, Xavier A, Waqas M, Tutino VM, Siddiqui A, Gupta G, Nanda A, Khandelwal P, Tiu C, Portela PC, Perez de la Ossa N, Urra X, de Lera M, Arenillas JF, Ribo M, Requena M, Piano M, Pero G, De Sousa K, Al-Mufti F, Hashim Z, Nayak S, Renieri L, Aziz-Sultan MA, Nguyen TN, Feineigle P, Patel AB, Siegler JE, Badih K, Grossberg JA, Saad H, Gooch MR, Herial NA, Rosenwasser RH, Tjoumakaris S, Tiwari A. Characteristics of a COVID-19 Cohort With Large Vessel Occlusion: A Multicenter International Study. Neurosurgery 2022; 90:725-733. [PMID: 35238817 PMCID: PMC9514728 DOI: 10.1227/neu.0000000000001902] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2021] [Accepted: 12/06/2021] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND The mechanisms and outcomes in coronavirus disease (COVID-19)-associated stroke are unique from those of non-COVID-19 stroke. OBJECTIVE To describe the efficacy and outcomes of acute revascularization of large vessel occlusion (LVO) in the setting of COVID-19 in an international cohort. METHODS We conducted an international multicenter retrospective study of consecutively admitted patients with COVID-19 with concomitant acute LVO across 50 comprehensive stroke centers. Our control group constituted historical controls of patients presenting with LVO and receiving a mechanical thrombectomy between January 2018 and December 2020. RESULTS The total cohort was 575 patients with acute LVO; 194 patients had COVID-19 while 381 patients did not. Patients in the COVID-19 group were younger (62.5 vs 71.2; P < .001) and lacked vascular risk factors (49, 25.3% vs 54, 14.2%; P = .001). Modified thrombolysis in cerebral infarction 3 revascularization was less common in the COVID-19 group (74, 39.2% vs 252, 67.2%; P < .001). Poor functional outcome at discharge (defined as modified Ranklin Scale 3-6) was more common in the COVID-19 group (150, 79.8% vs 132, 66.7%; P = .004). COVID-19 was independently associated with a lower likelihood of achieving modified thrombolysis in cerebral infarction 3 (odds ratio [OR]: 0.4, 95% CI: 0.2-0.7; P < .001) and unfavorable outcomes (OR: 2.5, 95% CI: 1.4-4.5; P = .002). CONCLUSION COVID-19 was an independent predictor of incomplete revascularization and poor outcomes in patients with stroke due to LVO. Patients with COVID-19 with LVO were younger, had fewer cerebrovascular risk factors, and suffered from higher morbidity/mortality rates.
Collapse
Affiliation(s)
- Pascal Jabbour
- Department of Neurological Surgery, Thomas Jefferson University Hospital, Philadelphia, Pennsylvania, USA;
| | - Adam A. Dmytriw
- Interventional Neuroradiology & Endovascular Neurosurgery Service, Mass General Brigham Partners, Harvard Medical School, Boston, Massachusetts, USA
| | - Ahmad Sweid
- Department of Neurological Surgery, Thomas Jefferson University Hospital, Philadelphia, Pennsylvania, USA;
| | - Michel Piotin
- Department of Interventional Neuroradiology, Rothschild Foundation Hospital, Paris, France;
| | - Kimon Bekelis
- Department of Neurosurgery, Good Samaritan Hospital Medical Center, West Islip, New York, USA
| | - Nader Sourour
- Department of Interventional Neuroradiology, Pitié-Salpêtrière Hospital, Paris, France
| | - Eytan Raz
- Department of Radiology, New York University Langone Medical Center, New York, New York, USA
| | - Italo Linfante
- Department of Interventional Neuroradiology & Neuroendovascular Surgery, Miami Cardiac and Vascular Institute, Baptist Hospital of Miami, Miami, Florida, USA
| | - Guilherme Dabus
- Department of Interventional Neuroradiology & Neuroendovascular Surgery, Miami Cardiac and Vascular Institute, Baptist Hospital of Miami, Miami, Florida, USA
| | - Max Kole
- Department of Neurosurgery, Henry Ford Hospital, Detroit, Michigan, USA;
| | - Mario Martínez-Galdámez
- Department of Interventional Neuroradiology, Hospital Clinico Universitario de Valladolid, Valladolid, Spain;
| | - Shahid M. Nimjee
- Department of Neurosurgery, The Ohio State University Wexner Medical Center, Columbus, Ohio, USA;
| | - Demetrius K. Lopes
- Department of Neurosurgery, Advocate Aurora Health, Chicago, Illinois, USA
| | - Ameer E. Hassan
- Department of Neuroscience, Valley Baptist Medical Center/University of Texas Rio Grande Valley, Harlingen, Texas, USA
| | - Peter Kan
- Department of Neurosurgery, Baylor College of Medicine, Houston, Texas, USA
- Department of Neurosurgery, University of Texas Medical Branch, Galveston, Texas, USA
| | | | - Michael R. Levitt
- Departments of Neurological Surgery, Radiology, Mechanical Engineering, and Stroke & Applied Neuroscience Center, University of Washington, Seattle, Washington, USA;
| | - Simon Escalard
- Department of Interventional Neuroradiology, Rothschild Foundation Hospital, Paris, France;
| | - Symeon Missios
- Department of Neurosurgery, Good Samaritan Hospital Medical Center, West Islip, New York, USA
| | - Maksim Shapiro
- Department of Radiology, New York University Langone Medical Center, New York, New York, USA
| | - Frédéric Clarençon
- Department of Interventional Neuroradiology, Pitié-Salpêtrière Hospital, Paris, France
| | - Mahmoud Elhorany
- Department of Interventional Neuroradiology, Pitié-Salpêtrière Hospital, Paris, France
| | - Daniel Vela-Duarte
- Department of Interventional Neuroradiology & Neuroendovascular Surgery, Miami Cardiac and Vascular Institute, Baptist Hospital of Miami, Miami, Florida, USA
| | - Rizwan A. Tahir
- Department of Neurosurgery, Henry Ford Hospital, Detroit, Michigan, USA;
| | - Patrick P. Youssef
- Department of Neurosurgery, The Ohio State University Wexner Medical Center, Columbus, Ohio, USA;
| | - Aditya S. Pandey
- Department of Neurosurgery, University of Michigan, Ann Arbor, Michigan, USA
| | - Robert M. Starke
- Department of Neurosurgery & Neuroradiology, University of Miami & Jackson Memorial Hospital, Miami, Florida, USA;
| | - Kareem El Naamani
- Department of Neurological Surgery, Thomas Jefferson University Hospital, Philadelphia, Pennsylvania, USA;
| | - Rawad Abbas
- Department of Neurological Surgery, Thomas Jefferson University Hospital, Philadelphia, Pennsylvania, USA;
| | - Bassel Hammoud
- Department of Biomedical Engineering, American University of Beirut, Beirut, Lebanon;
| | - Ossama Y. Mansour
- Department of Neurology and Neuroradiology, Alexandria University Hospital, Al Attarin, Egypt;
| | - Jorge Galvan
- Department of Interventional Neuroradiology, Hospital Clinico Universitario de Valladolid, Valladolid, Spain;
| | | | - Abolghasem Mortazavi
- Department of Neuroscience, Valley Baptist Medical Center/University of Texas Rio Grande Valley, Harlingen, Texas, USA
| | - Melanie Walker
- Departments of Neurological Surgery and Stroke & Applied Neuroscience Center, University of Washington, Seattle, Washington, USA;
| | - Mahmoud Dibas
- Interventional Neuroradiology & Endovascular Neurosurgery Service, Mass General Brigham Partners, Harvard Medical School, Boston, Massachusetts, USA
| | - Fabio Settecase
- Division of Neuroradiology, Vancouver General Hospital, University of British Columbia, Vancouver, British Columbia, USA
| | - Manraj K. S. Heran
- Division of Neuroradiology, Vancouver General Hospital, University of British Columbia, Vancouver, British Columbia, USA
| | - Anna L. Kuhn
- Division of Neurointerventional Radiology, Department of Radiology, UMass Memorial Medical Center, Worcester, Massachusetts, USA
| | - Ajit S. Puri
- Division of Neurointerventional Radiology, Department of Radiology, UMass Memorial Medical Center, Worcester, Massachusetts, USA
| | - Bijoy K. Menon
- Calgary Stroke Program, Cumming School of Medicine, Calgary, Alberta, Canada
| | - Sanjeev Sivakumar
- Department of Neurosurgery & Neuroradiology, University of Miami & Jackson Memorial Hospital, Miami, Florida, USA;
| | - Ashkan Mowla
- Department of Neurological Surgery, University of Southern California, Los Angeles, California, USA
| | - Salvatore D'Amato
- Interventional Neuroradiology & Endovascular Neurosurgery Service, Mass General Brigham Partners, Harvard Medical School, Boston, Massachusetts, USA
| | - Alicia M. Zha
- Department of Neurology, UT Health Science Center, Houston, Texas, USA
| | - Daniel Cooke
- Department of Neurointerventional Radiology, San Francisco General Hospital, San Francisco, California, USA
| | - Mayank Goyal
- Calgary Stroke Program, Cumming School of Medicine, Calgary, Alberta, Canada
| | - Hannah Wu
- Department of Neurology, Brookdale University Hospital, Brooklyn, New York, USA
- Department of Neurology, Jamaica Medical Center, Richmond Hill, New York, USA
- Department of Neurology, NYU Lutheran Hospital, Brooklyn, New York, USA
| | - Jake Cohen
- Department of Neurology, Brookdale University Hospital, Brooklyn, New York, USA
- Department of Neurology, Jamaica Medical Center, Richmond Hill, New York, USA
- Department of Neurology, NYU Lutheran Hospital, Brooklyn, New York, USA
| | - David Turkel-Parrella
- Department of Neurology, Brookdale University Hospital, Brooklyn, New York, USA
- Department of Neurology, Jamaica Medical Center, Richmond Hill, New York, USA
- Department of Neurology, NYU Lutheran Hospital, Brooklyn, New York, USA
| | - Andrew Xavier
- Department of Neurology, Sinai Grace Hospital, Detroit, Michigan, USA
- Department of Neurology, St. Joseph Mercy Health, Ann Arbor, Michigan, USA
| | - Muhammad Waqas
- Department of Neurosurgery, University of New York at Buffalo, Buffalo, New York, USA
| | - Vincent M. Tutino
- Department of Neurosurgery, University of New York at Buffalo, Buffalo, New York, USA
| | - Adnan Siddiqui
- Department of Neurosurgery, University of New York at Buffalo, Buffalo, New York, USA
| | - Gaurav Gupta
- Department of Neurology, Robert Wood Johnson University Hospital, New Brunswick, New Jersey, USA
| | - Anil Nanda
- Department of Neurology, Robert Wood Johnson University Hospital, New Brunswick, New Jersey, USA
| | - Priyank Khandelwal
- Department of Neurology, Robert Wood Johnson University Hospital, New Brunswick, New Jersey, USA
| | - Cristina Tiu
- Department of Physics, University of Toronto, Toronto, Ontario, Canada;
| | - Pere C. Portela
- Department of Neurosurgery, Emory University, Atlanta, Georgia, USA
| | - Natalia Perez de la Ossa
- Stroke Unit, Neuroscience Department, Hospital Universitari Germans Trias i Pujol, Badalona, Barcelona, Spain;
| | - Xabier Urra
- Department of Neurology, Hospital Clínic, Barcelona, Spain;
| | - Mercedes de Lera
- Department of Neurology, Hospital Clínico Universitario, Valladolid, Spain;
| | - Juan F. Arenillas
- Department of Neurology, Hospital Clínico Universitario, Valladolid, Spain;
| | - Marc Ribo
- Stroke Unit, Department of Neurology, Vall d'Hebron Research Institute, Barcelona, Spain;
- Cooper Neurological Institute, Cooper University Hospital, Camden, New Jersey, USA;
| | - Manuel Requena
- Stroke Unit, Department of Neurology, Vall d'Hebron Research Institute, Barcelona, Spain;
- Cooper Neurological Institute, Cooper University Hospital, Camden, New Jersey, USA;
| | - Mariangela Piano
- Department of Physics, University of Toronto, Toronto, Ontario, Canada;
| | - Guglielmo Pero
- Department of Physics, University of Toronto, Toronto, Ontario, Canada;
| | - Keith De Sousa
- Department of Neurosurgery, Emory University, Atlanta, Georgia, USA
| | - Fawaz Al-Mufti
- Stroke Unit, Neuroscience Department, Hospital Universitari Germans Trias i Pujol, Badalona, Barcelona, Spain;
| | - Zafar Hashim
- Department of Radiology, University Hospital of North Midlands, Stoke-on-Trent, UK
| | - Sanjeev Nayak
- Department of Neurology, Hospital Clínic, Barcelona, Spain;
| | - Leonardo Renieri
- Department of Radiology, Neurovascular Unit, Careggi University Hospital, Florence, Italy
| | - Mohamed A. Aziz-Sultan
- Interventional Neuroradiology & Endovascular Neurosurgery Service, Mass General Brigham Partners, Harvard Medical School, Boston, Massachusetts, USA
| | - Thanh N. Nguyen
- Departments of Neurology and Radiology, Boston Medical Center, Boston University School of Medicine, Boston, Massachusetts, USA;
| | - Patricia Feineigle
- Cooper Neurological Institute, Cooper University Hospital, Camden, New Jersey, USA;
| | - Aman B. Patel
- Interventional Neuroradiology & Endovascular Neurosurgery Service, Mass General Brigham Partners, Harvard Medical School, Boston, Massachusetts, USA
| | - James E. Siegler
- Cooper Neurological Institute, Cooper University Hospital, Camden, New Jersey, USA;
| | - Khodr Badih
- Department of Physics, University of Toronto, Toronto, Ontario, Canada;
| | | | - Hassan Saad
- Department of Neurosurgery, Emory University, Atlanta, Georgia, USA
| | - M. Reid Gooch
- Department of Neurological Surgery, Thomas Jefferson University Hospital, Philadelphia, Pennsylvania, USA;
| | - Nabeel A. Herial
- Department of Neurological Surgery, Thomas Jefferson University Hospital, Philadelphia, Pennsylvania, USA;
| | - Robert H. Rosenwasser
- Department of Neurological Surgery, Thomas Jefferson University Hospital, Philadelphia, Pennsylvania, USA;
| | - Stavropoula Tjoumakaris
- Department of Neurological Surgery, Thomas Jefferson University Hospital, Philadelphia, Pennsylvania, USA;
| | - Ambooj Tiwari
- Department of Neurology, Brookdale University Hospital, Brooklyn, New York, USA
- Department of Neurology, Jamaica Medical Center, Richmond Hill, New York, USA
- Department of Neurology, NYU Lutheran Hospital, Brooklyn, New York, USA
| |
Collapse
|
57
|
Duman M, Şık N, Tekşam Ö, Akça H, Kurt F, Çağlar AA, Yıldız LA, Taşar MA, Fidancı İ, Yayla BCC, Yılmaz D, Güngör E, Demir Ş, Çokuğraş H, Cebeci SO, Önal P, Saz EU, Yurtseven A, Uysalol M, Yıldız R, Gümüş S, Bal A, Bayturan SŞ, Zengin N, Atik S, Çiftdoğan DY, Berksoy E, Çiçek A, Şahin S, Kızıl MC, Kara Y, Apa H, Ulusoy E, Kara AA, Yesil E, Erdem M, Turan C, Arslanoglu S, Duyu M, Besli GE, Arslan G, Oflu AT, Çeleğen M, Buldu E, Pişkin İE, Kardeş H, Yılmaz HL, Yıldızdaş D, Gökulu G, Çay P, Özer U, Güleryüz OD, Çolak Ö, Güneysu ST. COVID-19 disease in children presenting to the pediatric emergency department: A multicenter study with 8886 cases from Turkey. Am J Emerg Med 2022; 59:133-140. [PMID: 35849960 PMCID: PMC9181308 DOI: 10.1016/j.ajem.2022.06.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Revised: 06/03/2022] [Accepted: 06/04/2022] [Indexed: 11/16/2022] Open
Abstract
Background The aim was to evaluate the epidemiological, clinical, laboratory, and radiologic data of children with SARS-CoV-2 positivity by polymerase chain reaction (PCR) together with treatment strategies and clinical outcomes and to evaluate cases of multisystem inflammatory syndrome in children (MIS-C) in this population. Methods This was a multicenter retrospective observational cohort study performed in the pediatric emergency departments of 19 tertiary hospitals. From March 11, 2020, to May 31, 2021, children who were diagnosed with confirmed nasopharyngeal/tracheal specimen SARS-CoV-2 PCR positivity or positivity for serum-specific antibodies against SARS-CoV-2 were included. Demographics, presence of chronic illness, symptoms, history of contact with SARS-CoV-2 PCR-positive individuals, laboratory and radiologic investigations, clinical severity, hospital admissions, and prognosis were recorded. Results A total of 8886 cases were included. While 8799 (99.0%) cases resulted in a diagnosis of SARS-CoV-2 with PCR positivity, 87 (1.0%) patients were diagnosed with MIS-C. Among SARS-CoV-2 PCR-positive patients, 51.0% were male and 8.5% had chronic illnesses. The median age was 11.6 years (IQR: 5.0–15.4) and 737 (8.4%) patients were aged <1 year. Of the patients, 15.5% were asymptomatic. The most common symptoms were fever (48.5%) and cough (30.7%) for all age groups. There was a decrease in the rate of fever as age increased (p < 0.001); the most common age group for this symptom was <1 year with the rate of 69.6%. There was known contact with a SARS-CoV-2 PCR-positive individual in 67.3% of the cases, with household contacts in 71.3% of those cases. In terms of clinical severity, 83 (0.9%) patients were in the severe-critical group. There was hospital admission in 1269 (14.4%) cases, with 106 (1.2%) of those patients being admitted to the pediatric intensive care unit (PICU). Among patients with MIS-C, 60.9% were male and the median age was 6.4 years (IQR: 3.9–10.4). Twelve (13.7%) patients presented with shock. There was hospital admission in 89.7% of these cases, with 29.9% of the patients with MIS-C being admitted to the PICU. Conclusion Most SARS-CoV-2 PCR-positive patients presented with a mild clinical course. Although rare, MIS-C emerges as a serious consequence with frequent PICU admission. Further understanding of the characteristics of COVID-19 disease could provide insights and guide the development of therapeutic strategies for target groups.
Collapse
Affiliation(s)
- Murat Duman
- Division of Pediatric Emergency Care, Department of Pediatrics, Dokuz Eylul University Faculty of Medicine, Izmir, Turkey.
| | - Nihan Şık
- Division of Pediatric Emergency Care, Department of Pediatrics, Dokuz Eylul University Faculty of Medicine, Izmir, Turkey
| | - Özlem Tekşam
- Hacettepe University Ihsan Dogramaci Children's Hospital, Ankara, Turkey
| | - Halise Akça
- Ankara City Hospital, Department of Pediatric Emergency Medicine, Ankara, Turkey
| | - Funda Kurt
- Ankara City Hospital, Department of Pediatric Emergency Medicine, Ankara, Turkey
| | - Ayla Akca Çağlar
- Ankara City Hospital, Department of Pediatric Emergency Medicine, Ankara, Turkey
| | - Leman Akcan Yıldız
- Ankara City Hospital, Department of Pediatric Emergency Medicine, Ankara, Turkey
| | - Medine Ayşin Taşar
- University of Health Sciences, Ankara Training and Research Hospital, Pediatric Emergency Department, Ankara, Turkey
| | - İlknur Fidancı
- University of Health Sciences, Ankara Training and Research Hospital, Pediatric Emergency Department, Ankara, Turkey
| | - Burcu Ceylan Cura Yayla
- University of Health Sciences, Ankara Training and Research Hospital, Pediatric Emergency Department, Ankara, Turkey
| | - Durgül Yılmaz
- Division of Pediatric Emergency Care, Department of Pediatrics, Dokuz Eylul University Faculty of Medicine, Izmir, Turkey
| | - Emre Güngör
- Hacettepe University Ihsan Dogramaci Children's Hospital, Ankara, Turkey
| | - Şule Demir
- Aydın Gynecology and Childhood Hospital, Pediatric Emergency Department, Aydın, Turkey
| | - Haluk Çokuğraş
- Cerrahpasa Faculty of Medicine, İstanbul University-Cerrahpasa, Istanbul, Turkey
| | - Sinem Oral Cebeci
- Cerrahpasa Faculty of Medicine, İstanbul University-Cerrahpasa, Istanbul, Turkey
| | - Pınar Önal
- Cerrahpasa Faculty of Medicine, İstanbul University-Cerrahpasa, Istanbul, Turkey
| | | | | | - Metin Uysalol
- Istanbul University, Istanbul Faculty of Medicine, Department of Pediatrics, Division of Pediatric Emergency, Istanbul, Turkey
| | - Raif Yıldız
- Istanbul University, Istanbul Faculty of Medicine, Department of Pediatrics, Division of Pediatric Emergency, Istanbul, Turkey
| | - Süheyla Gümüş
- Istanbul University, Istanbul Faculty of Medicine, Department of Pediatrics, Division of Pediatric Emergency, Istanbul, Turkey
| | - Alkan Bal
- Manisa Celal Bayar University Faculty of Medicine, Hafsa Sultan Hospital, Manisa, Turkey
| | - Semra Şen Bayturan
- Manisa Celal Bayar University Faculty of Medicine, Hafsa Sultan Hospital, Manisa, Turkey
| | - Neslihan Zengin
- Manisa Celal Bayar University Faculty of Medicine, Hafsa Sultan Hospital, Manisa, Turkey
| | - Sinem Atik
- Manisa Celal Bayar University Faculty of Medicine, Hafsa Sultan Hospital, Manisa, Turkey
| | - Dilek Yılmaz Çiftdoğan
- Pediatric Emergency Medicine Clinic, University of Health Sciences, Tepecik Education and Research Hospital, Izmir, Turkey
| | - Emel Berksoy
- Pediatric Emergency Medicine Clinic, University of Health Sciences, Tepecik Education and Research Hospital, Izmir, Turkey
| | - Alper Çiçek
- Pediatric Emergency Medicine Clinic, University of Health Sciences, Tepecik Education and Research Hospital, Izmir, Turkey
| | - Sabiha Şahin
- Eskisehir Osmangazi University Faculty of Medicine, Eskisehir, Turkey
| | - Mahmut Can Kızıl
- Eskisehir Osmangazi University Faculty of Medicine, Eskisehir, Turkey
| | - Yalçın Kara
- Eskisehir Osmangazi University Faculty of Medicine, Eskisehir, Turkey
| | - Hurşit Apa
- Dr. Behçet Uz Child Disease and Pediatric Surgery Training and Research Hospital, Izmir, Turkey
| | - Emel Ulusoy
- Dr. Behçet Uz Child Disease and Pediatric Surgery Training and Research Hospital, Izmir, Turkey
| | - Aybüke Akaslan Kara
- Dr. Behçet Uz Child Disease and Pediatric Surgery Training and Research Hospital, Izmir, Turkey
| | - Edanur Yesil
- Mersin City Training and Research Hospital, Mersin, Turkey
| | - Meltem Erdem
- Mersin City Training and Research Hospital, Mersin, Turkey
| | - Caner Turan
- Mersin City Training and Research Hospital, Mersin, Turkey
| | - Sertac Arslanoglu
- Istanbul Medeniyet University Faculty of Medicine, Goztepe Prof Dr Suleyman Yalcin City Hospital, Istanbul, Turkey
| | - Muhterem Duyu
- Istanbul Medeniyet University Faculty of Medicine, Goztepe Prof Dr Suleyman Yalcin City Hospital, Istanbul, Turkey
| | - Gulser Esen Besli
- Istanbul Medeniyet University Faculty of Medicine, Goztepe Prof Dr Suleyman Yalcin City Hospital, Istanbul, Turkey
| | - Gazi Arslan
- Derince Training and Research Hospital, Kocaeli, Turkey
| | - Ayşe Tolunay Oflu
- Afyonkarahisar Health Sciences University Faculty of Medicine, Department of Pediatrics, Afyon, Turkey
| | - Mehmet Çeleğen
- Afyonkarahisar Health Sciences University Faculty of Medicine, Department of Pediatrics, Afyon, Turkey
| | - Ebru Buldu
- Afyonkarahisar Health Sciences University Faculty of Medicine, Department of Pediatrics, Afyon, Turkey
| | - İbrahim Etem Pişkin
- Zonguldak Bulent Ecevit University Faculty of Medicine, Department of Pediatrics, Zonguldak, Turkey
| | - Hakan Kardeş
- Zonguldak Bulent Ecevit University Faculty of Medicine, Department of Pediatrics, Zonguldak, Turkey
| | | | | | - Gamze Gökulu
- Cukurova University, Faculty of Medicine, Adana, Turkey
| | - Pınar Çay
- Cukurova University, Faculty of Medicine, Adana, Turkey
| | - Utku Özer
- Cukurova University, Faculty of Medicine, Adana, Turkey
| | | | - Özlem Çolak
- Gazi University, Faculty of Medicine, Department of Pediatric Emergency, Ankara, Turkey
| | - Songül Tomar Güneysu
- Gazi University, Faculty of Medicine, Department of Pediatric Emergency, Ankara, Turkey
| |
Collapse
|
58
|
Chaudhary PK, Kim S, Kim S. An Insight into Recent Advances on Platelet Function in Health and Disease. Int J Mol Sci 2022; 23:ijms23116022. [PMID: 35682700 PMCID: PMC9181192 DOI: 10.3390/ijms23116022] [Citation(s) in RCA: 31] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2022] [Revised: 05/22/2022] [Accepted: 05/24/2022] [Indexed: 12/04/2022] Open
Abstract
Platelets play a variety of roles in vascular biology and are best recognized as primary hemostasis and thrombosis mediators. Platelets have a large number of receptors and secretory molecules that are required for platelet functionality. Upon activation, platelets release multiple substances that have the ability to influence both physiological and pathophysiological processes including inflammation, tissue regeneration and repair, cancer progression, and spreading. The involvement of platelets in the progression and seriousness of a variety of disorders other than thrombosis is still being discovered, especially in the areas of inflammation and the immunological response. This review represents an integrated summary of recent advances on the function of platelets in pathophysiology that connects hemostasis, inflammation, and immunological response in health and disease and suggests that antiplatelet treatment might be used for more than only thrombosis.
Collapse
|
59
|
Zhang L, Xu J, Qi X, Tao Z, Yang Z, Chen W, Wang X, Pan T, Dai Y, Tian R, Chen Y, Tang B, Liu Z, Tan R, Qu H, Yu Y, Liu J. Development and Validation of a Nomogram for Predicting the Risk of Coronavirus-Associated Acute Respiratory Distress Syndrome: A Retrospective Cohort Study. Infect Drug Resist 2022; 15:2371-2381. [PMID: 35528184 PMCID: PMC9075028 DOI: 10.2147/idr.s348278] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2021] [Accepted: 02/12/2022] [Indexed: 12/15/2022] Open
Abstract
Background Since the outbreak of coronavirus disease (COVID-19) in December 2019 in Wuhan, it has spread rapidly worldwide. We aimed to establish and validate a nomogram that predicts the probability of coronavirus-associated acute respiratory distress syndrome (CARDS). Methods In this single-centre, retrospective study, 261 patients with COVID-19 were recruited using positive reverse transcription–polymerase chain reaction tests for severe acute respiratory syndrome coronavirus 2 in Tongji Hospital at Huazhong University of Science and Technology (Wuhan, China). These patients were randomly distributed into the training cohort (75%) and the validation cohort (25%). The factors included in the nomogram were determined using univariate and multivariate logistic regression analyses based on the training cohort. The area under the receiver operating characteristic curve (AUC), consistency index (C-index), calibration curve, and decision curve analysis (DCA) were used to evaluate the efficiency of the nomogram in the training and validation cohorts. Results Independent predictive factors, including fasting plasma glucose, platelet, D-dimer, and cTnI, were determined using the nomogram. In the training cohort, the AUC and concordance index were 0.93. Similarly, in the validation cohort, the nomogram still showed great distinction (AUC: 0.92) and better calibration. The calibration plot also showed a high degree of agreement between the predicted and actual probabilities of CARDS. In addition, the DCA proved that the nomogram was clinically beneficial. Conclusion Based on the results of laboratory tests, we established a predictive model for acute respiratory distress syndrome in patients with COVID-19. This model shows good performance and can be used clinically to identify CARDS early. Trial Registration Ethics committee of Ruijin Hospital, Shanghai Jiao Tong University School of Medicine (No.:(2020) Linlun-34th).
Collapse
Affiliation(s)
- Li Zhang
- Department of Critical Care Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People’s Republic of China
| | - Jing Xu
- Department of Critical Care Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People’s Republic of China
| | - Xiaoling Qi
- Department of Critical Care Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People’s Republic of China
| | - Zheying Tao
- Department of Critical Care Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People’s Republic of China
| | - Zhitao Yang
- Emergency Department, Ruijin Hospital affiliate to Shanghai Jiaotong University School of Medicine, Shanghai, People’s Republic of China
| | - Wei Chen
- Department of Pulmonary and Critical Care Medicine, Ruijin Hospital Affiliate to Shanghai Jiaotong University School of Medicine, Shanghai, People’s Republic of China
| | - Xiaoli Wang
- Department of Critical Care Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People’s Republic of China
| | - Tingting Pan
- Department of Critical Care Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People’s Republic of China
| | - Yunqi Dai
- Department of Critical Care Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People’s Republic of China
| | - Rui Tian
- Department of Critical Care Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People’s Republic of China
| | - Yang Chen
- Department of Critical Care Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People’s Republic of China
| | - Bin Tang
- Department of Critical Care Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People’s Republic of China
| | - Zhaojun Liu
- Department of Critical Care Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People’s Republic of China
| | - Ruoming Tan
- Department of Critical Care Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People’s Republic of China
| | - Hongping Qu
- Department of Critical Care Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People’s Republic of China
| | - Yue Yu
- Department of Critical Care Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People’s Republic of China
| | - Jialin Liu
- Department of Critical Care Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People’s Republic of China
- Correspondence: Jialin Liu; Yue Yu, Department of Critical Care Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200020, People’s Republic of China, Email ;
| |
Collapse
|
60
|
Fricke-Galindo I, Buendia-Roldan I, Chavez-Galan L, Pérez-Rubio G, Hernández-Zenteno RDJ, Ramos-Martinez E, Zazueta-Márquez A, Reyes-Melendres F, Alarcón-Dionet A, Guzmán-Vargas J, Bravo-Gutiérrez OA, Quintero-Puerta T, Gutiérrez-Pérez IA, Ortega-Martínez A, Ambrocio-Ortiz E, Nava-Quiroz KJ, Bañuelos-Flores JL, Jaime-Capetillo ME, Mejía M, Rojas-Serrano J, Falfán-Valencia R. SERPINE1 rs6092 Variant Is Related to Plasma Coagulation Proteins in Patients with Severe COVID-19 from a Tertiary Care Hospital. BIOLOGY 2022; 11:biology11040595. [PMID: 35453794 PMCID: PMC9029072 DOI: 10.3390/biology11040595] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Revised: 03/17/2022] [Accepted: 04/12/2022] [Indexed: 12/24/2022]
Abstract
An impaired coagulation process has been described in patients with severe or critical coronavirus disease (COVID-19). Nevertheless, the implication of coagulation-related genes has not been explored. We aimed to evaluate the impact of F5 rs6025 and SERPINE1 rs6092 on invasive mechanical ventilation (IMV) requirement and the levels of coagulation proteins among patients with severe COVID-19. Four-hundred fifty-five patients with severe COVID-19 were genotyped using TaqMan assays. Coagulation-related proteins (P-Selectin, D-dimer, P-selectin glycoprotein ligand-1, tissue plasminogen activator [tPA], plasminogen activator inhibitor-1, and Factor IX) were assessed by cytometric bead arrays in one- and two-time determinations. Accordingly, SERPINE1 rs6092, P-Selectin (GG 385 pg/mL vs. AG+AA 632 pg/mL, p = 0.0037), and tPA (GG 1858 pg/mL vs. AG+AA 2546 pg/mL, p = 0.0284) levels were different. Patients carrying the CT F5-rs6025 genotype exhibited lower levels of factor IX (CC 17,136 pg/mL vs. CT 10,247 pg/mL, p = 0.0355). Coagulation proteins were also different among IMV patients than non-IMV. PSGL-1 levels were significantly increased in the late stage of COVID-19 (>10 days). The frequencies of F5 rs6025 and SERPINE1 rs6092 variants were not different among IMV and non-IMV. The SERPINE1 rs6092 variant is related to the impaired coagulation process in patients with COVID-19 severe.
Collapse
Affiliation(s)
- Ingrid Fricke-Galindo
- HLA Laboratory, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, Mexico City 14080, Mexico; (I.F.-G.); (G.P.-R.); (A.Z.-M.); (F.R.-M.); (J.G.-V.); (O.A.B.-G.); (T.Q.-P.); (I.A.G.-P.); (A.O.-M.); (E.A.-O.); (K.J.N.-Q.)
| | - Ivette Buendia-Roldan
- Translational Research Laboratory on Aging and Pulmonary Fibrosis, Instituto Nacional de Enfermedades Respiratorias Ismael Cosio Villegas, Mexico City 14080, Mexico; (I.B.-R.); (A.A.-D.)
| | - Leslie Chavez-Galan
- Laboratory of Integrative Immunology, Instituto Nacional de Enfermedades Respiratorias Ismael Cosio Villegas, Mexico City 14080, Mexico;
| | - Gloria Pérez-Rubio
- HLA Laboratory, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, Mexico City 14080, Mexico; (I.F.-G.); (G.P.-R.); (A.Z.-M.); (F.R.-M.); (J.G.-V.); (O.A.B.-G.); (T.Q.-P.); (I.A.G.-P.); (A.O.-M.); (E.A.-O.); (K.J.N.-Q.)
| | | | - Espiridión Ramos-Martinez
- Unidad de Investigación en Medicina Experimental, Facultad de Medicina, Universidad Nacional Autónoma de México, Mexico City 06720, Mexico;
| | - Armando Zazueta-Márquez
- HLA Laboratory, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, Mexico City 14080, Mexico; (I.F.-G.); (G.P.-R.); (A.Z.-M.); (F.R.-M.); (J.G.-V.); (O.A.B.-G.); (T.Q.-P.); (I.A.G.-P.); (A.O.-M.); (E.A.-O.); (K.J.N.-Q.)
| | - Felipe Reyes-Melendres
- HLA Laboratory, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, Mexico City 14080, Mexico; (I.F.-G.); (G.P.-R.); (A.Z.-M.); (F.R.-M.); (J.G.-V.); (O.A.B.-G.); (T.Q.-P.); (I.A.G.-P.); (A.O.-M.); (E.A.-O.); (K.J.N.-Q.)
| | - Aimé Alarcón-Dionet
- Translational Research Laboratory on Aging and Pulmonary Fibrosis, Instituto Nacional de Enfermedades Respiratorias Ismael Cosio Villegas, Mexico City 14080, Mexico; (I.B.-R.); (A.A.-D.)
| | - Javier Guzmán-Vargas
- HLA Laboratory, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, Mexico City 14080, Mexico; (I.F.-G.); (G.P.-R.); (A.Z.-M.); (F.R.-M.); (J.G.-V.); (O.A.B.-G.); (T.Q.-P.); (I.A.G.-P.); (A.O.-M.); (E.A.-O.); (K.J.N.-Q.)
| | - Omar Andrés Bravo-Gutiérrez
- HLA Laboratory, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, Mexico City 14080, Mexico; (I.F.-G.); (G.P.-R.); (A.Z.-M.); (F.R.-M.); (J.G.-V.); (O.A.B.-G.); (T.Q.-P.); (I.A.G.-P.); (A.O.-M.); (E.A.-O.); (K.J.N.-Q.)
| | - Teresa Quintero-Puerta
- HLA Laboratory, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, Mexico City 14080, Mexico; (I.F.-G.); (G.P.-R.); (A.Z.-M.); (F.R.-M.); (J.G.-V.); (O.A.B.-G.); (T.Q.-P.); (I.A.G.-P.); (A.O.-M.); (E.A.-O.); (K.J.N.-Q.)
| | - Ilse Adriana Gutiérrez-Pérez
- HLA Laboratory, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, Mexico City 14080, Mexico; (I.F.-G.); (G.P.-R.); (A.Z.-M.); (F.R.-M.); (J.G.-V.); (O.A.B.-G.); (T.Q.-P.); (I.A.G.-P.); (A.O.-M.); (E.A.-O.); (K.J.N.-Q.)
| | - Alejandro Ortega-Martínez
- HLA Laboratory, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, Mexico City 14080, Mexico; (I.F.-G.); (G.P.-R.); (A.Z.-M.); (F.R.-M.); (J.G.-V.); (O.A.B.-G.); (T.Q.-P.); (I.A.G.-P.); (A.O.-M.); (E.A.-O.); (K.J.N.-Q.)
| | - Enrique Ambrocio-Ortiz
- HLA Laboratory, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, Mexico City 14080, Mexico; (I.F.-G.); (G.P.-R.); (A.Z.-M.); (F.R.-M.); (J.G.-V.); (O.A.B.-G.); (T.Q.-P.); (I.A.G.-P.); (A.O.-M.); (E.A.-O.); (K.J.N.-Q.)
| | - Karol J. Nava-Quiroz
- HLA Laboratory, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, Mexico City 14080, Mexico; (I.F.-G.); (G.P.-R.); (A.Z.-M.); (F.R.-M.); (J.G.-V.); (O.A.B.-G.); (T.Q.-P.); (I.A.G.-P.); (A.O.-M.); (E.A.-O.); (K.J.N.-Q.)
| | - José Luis Bañuelos-Flores
- Clinical Laboratory Service, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, Mexico City 14080, Mexico; (J.L.B.-F.); (M.E.J.-C.)
| | - María Esther Jaime-Capetillo
- Clinical Laboratory Service, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, Mexico City 14080, Mexico; (J.L.B.-F.); (M.E.J.-C.)
| | - Mayra Mejía
- Interstitial Pulmonary Diseases and Rheumatology Unit, Instituto Nacional de Enfermedades Respiratorias Ismael Cosio Villegas, Mexico City 06720, Mexico; (M.M.); (J.R.-S.)
| | - Jorge Rojas-Serrano
- Interstitial Pulmonary Diseases and Rheumatology Unit, Instituto Nacional de Enfermedades Respiratorias Ismael Cosio Villegas, Mexico City 06720, Mexico; (M.M.); (J.R.-S.)
| | - Ramcés Falfán-Valencia
- HLA Laboratory, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, Mexico City 14080, Mexico; (I.F.-G.); (G.P.-R.); (A.Z.-M.); (F.R.-M.); (J.G.-V.); (O.A.B.-G.); (T.Q.-P.); (I.A.G.-P.); (A.O.-M.); (E.A.-O.); (K.J.N.-Q.)
- Correspondence: ; Tel.: +52-55-5487-1700 (ext. 5152)
| |
Collapse
|
61
|
SARS-CoV-2 Spike Protein Binding of Glycated Serum Albumin-Its Potential Role in the Pathogenesis of the COVID-19 Clinical Syndromes and Bias towards Individuals with Pre-Diabetes/Type 2 Diabetes and Metabolic Diseases. Int J Mol Sci 2022; 23:ijms23084126. [PMID: 35456942 PMCID: PMC9030890 DOI: 10.3390/ijms23084126] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2022] [Revised: 04/04/2022] [Accepted: 04/06/2022] [Indexed: 01/08/2023] Open
Abstract
The immune response to SARS-CoV-2 infection requires antibody recognition of the spike protein. In a study designed to examine the molecular features of anti-spike and anti-nucleocapsid antibodies, patient plasma proteins binding to pre-fusion stabilised complete spike and nucleocapsid proteins were isolated and analysed by matrix-assisted laser desorption ionisation–time of flight (MALDI-ToF) mass spectrometry. Amongst the immunoglobulins, a high affinity for human serum albumin was evident in the anti-spike preparations. Careful mass comparison revealed the preferential capture of advanced glycation end product (AGE) forms of glycated human serum albumin by the pre-fusion spike protein. The ability of bacteria and viruses to surround themselves with serum proteins is a recognised immune evasion and pathogenic process. The preference of SARS-CoV-2 for AGE forms of glycated serum albumin may in part explain the severity and pathology of acute respiratory distress and the bias towards the elderly and those with (pre)diabetic and atherosclerotic/metabolic disease.
Collapse
|
62
|
Coagulopathy and Fibrinolytic Pathophysiology in COVID-19 and SARS-CoV-2 Vaccination. Int J Mol Sci 2022; 23:ijms23063338. [PMID: 35328761 PMCID: PMC8955234 DOI: 10.3390/ijms23063338] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2022] [Revised: 03/03/2022] [Accepted: 03/15/2022] [Indexed: 02/06/2023] Open
Abstract
Coronavirus Disease 2019 (COVID-19) is caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) and is frequently complicated by thrombosis. In some cases of severe COVID-19, fibrinolysis may be markedly enhanced within a few days, resulting in fatal bleeding. In the treatment of COVID-19, attention should be paid to both coagulation activation and fibrinolytic activation. Various thromboses are known to occur after vaccination with SARS-CoV-2 vaccines. Vaccine-induced immune thrombotic thrombocytopenia (VITT) can occur after adenovirus-vectored vaccination, and is characterized by the detection of anti-platelet factor 4 antibodies by enzyme-linked immunosorbent assay and thrombosis in unusual locations such as cerebral venous sinuses and visceral veins. Treatment comprises high-dose immunoglobulin, argatroban, and fondaparinux. Some VITT cases show marked decreases in fibrinogen and platelets and marked increases in D-dimer, suggesting the presence of enhanced-fibrinolytic-type disseminated intravascular coagulation with a high risk of bleeding. In the treatment of VITT, evaluation of both coagulation activation and fibrinolytic activation is important, adjusting treatments accordingly to improve outcomes.
Collapse
|
63
|
COVID-19 Induced Coagulopathy (CIC): Thrombotic Manifestations of Viral Infection. TH OPEN : COMPANION JOURNAL TO THROMBOSIS AND HAEMOSTASIS 2022; 6:e70-e79. [PMID: 35280973 PMCID: PMC8913175 DOI: 10.1055/s-0042-1744185] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/29/2021] [Accepted: 11/18/2021] [Indexed: 01/08/2023]
Abstract
Coronavirus disease 2019 (COVID-19) is caused by the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) and may result in an overactive coagulative system, thereby resulting in serious cardiovascular consequences in critically affected patients. The respiratory tract is a primary target for COVID-19 infection, which is manifested as acute lung injury in the most severe form of the viral infection, leading to respiratory failure. A proportion of infected patients may progress to serious systemic disease including dysfunction of multiple organs, acute respiratory distress syndrome (ARDS), and coagulation abnormalities, all of which are associated with increased mortality, additionally depending on age and compromised immunity. Coagulation abnormalities associated with COVID-19 mimic other systemic coagulopathies otherwise involved in other severe infections, such as disseminated intravascular coagulation (DIC) and may be termed COVID-19 induced coagulopathy (CIC). There is substantial evidence that patients with severe COVID-19 exhibiting CIC can develop venous and arterial thromboembolic complications. In the initial stages of CIC, significant elevation of D-dimer and fibrin/fibrinogen degradation products is observed. Alteration in prothrombin time, activated partial thromboplastin time, and platelet counts are less common in the early phase of the disease. In patients admitted to intensive care units (ICUs), coagulation test screening involving the measurement of D-dimer and fibrinogen levels, has been recommended. Prior established protocols for thromboembolic prophylaxis are also followed for CIC, including the use of heparin and other standard supportive care measures. In the present review, we summarize the characteristics of CIC and its implications for thrombosis, clinical findings of coagulation parameters in SARS-CoV-2 infected patients with incidences of thromboembolic events and plausible therapeutic measures.
Collapse
|
64
|
Juang LJ, Hur WS, Silva LM, Strilchuk AW, Francisco B, Leung J, Robertson MK, Groeneveld DJ, La Prairie B, Chun EM, Cap AP, Luyendyk JP, Palumbo JS, Cullis PR, Bugge TH, Flick MJ, Kastrup CJ. Suppression of fibrin(ogen)-driven pathologies in disease models through controlled knockdown by lipid nanoparticle delivery of siRNA. Blood 2022; 139:1302-1311. [PMID: 34958662 PMCID: PMC8900269 DOI: 10.1182/blood.2021014559] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2021] [Accepted: 12/14/2021] [Indexed: 11/20/2022] Open
Abstract
Fibrinogen plays a pathologic role in multiple diseases. It contributes to thrombosis and modifies inflammatory and immune responses, supported by studies in mice expressing fibrinogen variants with altered function or with a germline fibrinogen deficiency. However, therapeutic strategies to safely and effectively tailor plasma fibrinogen concentration are lacking. Here, we developed a strategy to tune fibrinogen expression by administering lipid nanoparticle (LNP)-encapsulated small interfering RNA (siRNA) targeting the fibrinogen α chain (siFga). Three distinct LNP-siFga reagents reduced both hepatic Fga messenger RNA and fibrinogen levels in platelets and plasma, with plasma levels decreased to 42%, 16%, and 4% of normal within 1 week of administration. Using the most potent siFga, circulating fibrinogen was controllably decreased to 32%, 14%, and 5% of baseline with 0.5, 1.0, and 2.0 mg/kg doses, respectively. Whole blood from mice treated with siFga formed clots with significantly decreased clot strength ex vivo, but siFga treatment did not compromise hemostasis following saphenous vein puncture or tail transection. In an endotoxemia model, siFga suppressed the acute phase response and decreased plasma fibrinogen, D-dimer, and proinflammatory cytokine levels. In a sterile peritonitis model, siFga restored normal macrophage migration in plasminogen-deficient mice. Finally, treatment of mice with siFga decreased the metastatic potential of tumor cells in a manner comparable to that observed in fibrinogen-deficient mice. The results indicate that siFga causes robust and controllable depletion of fibrinogen and provides the proof-of-concept that this strategy can modulate the pleiotropic effects of fibrinogen in relevant disease models.
Collapse
Affiliation(s)
- Lih Jiin Juang
- Michael Smith Laboratories
- Centre for Blood Research, and
- Department of Biochemistry and Molecular Biology, University of British Columbia, Vancouver, BC, Canada
| | - Woosuk S Hur
- Department of Pathology and Laboratory Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC
| | - Lakmali M Silva
- Proteases and Tissue Remodeling Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD
| | - Amy W Strilchuk
- Michael Smith Laboratories
- Centre for Blood Research, and
- Department of Biochemistry and Molecular Biology, University of British Columbia, Vancouver, BC, Canada
| | - Brenton Francisco
- Cancer and Blood Diseases Institute, Cincinnati Children's Hospital Medical Center and the University of Cincinnati College of Medicine, Cincinnati, OH
| | - Jerry Leung
- Michael Smith Laboratories
- Centre for Blood Research, and
- Department of Biochemistry and Molecular Biology, University of British Columbia, Vancouver, BC, Canada
| | - Madelaine K Robertson
- Michael Smith Laboratories
- Centre for Blood Research, and
- Department of Biochemistry and Molecular Biology, University of British Columbia, Vancouver, BC, Canada
| | - Dafna J Groeneveld
- Department of Pathobiology and Diagnostic Investigation, Institute for Integrative Toxicology, Michigan State University, East Lansing, MI
| | - Bridget La Prairie
- Michael Smith Laboratories
- Centre for Blood Research, and
- Department of Biochemistry and Molecular Biology, University of British Columbia, Vancouver, BC, Canada
| | - Elizabeth M Chun
- Proteases and Tissue Remodeling Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD
| | - Andrew P Cap
- The United States Army Institute of Surgical Research, JBSA-Fort Sam Houston, TX
- Department of Medicine, Uniformed Services University, Bethesda, MD
| | - James P Luyendyk
- Department of Pathobiology and Diagnostic Investigation, Institute for Integrative Toxicology, Michigan State University, East Lansing, MI
| | - Joseph S Palumbo
- Cancer and Blood Diseases Institute, Cincinnati Children's Hospital Medical Center and the University of Cincinnati College of Medicine, Cincinnati, OH
| | - Pieter R Cullis
- Department of Biochemistry and Molecular Biology, University of British Columbia, Vancouver, BC, Canada
| | - Thomas H Bugge
- Proteases and Tissue Remodeling Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD
| | - Matthew J Flick
- Department of Pathology and Laboratory Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC
| | - Christian J Kastrup
- Michael Smith Laboratories
- Centre for Blood Research, and
- Department of Biochemistry and Molecular Biology, University of British Columbia, Vancouver, BC, Canada
- Blood Research Institute, Versiti, Milwaukee, WI; and
- Department of Surgery, Department of Biochemistry, Department of Biomedical Engineering, and Department of Pharmacology and Toxicology, Medical College of Wisconsin, Milwaukee, WI
| |
Collapse
|
65
|
Barrett CD, Moore HB, Moore EE, Wang J, Hajizadeh N, Biffl WL, Lottenberg L, Patel PR, Truitt MS, McIntyre RC, Bull TM, Ammons LA, Ghasabyan A, Chandler J, Douglas IS, Schmidt EP, Moore PK, Wright FL, Ramdeo R, Borrego R, Rueda M, Dhupa A, McCaul DS, Dandan T, Sarkar PK, Khan B, Sreevidya C, McDaniel C, Grossman Verner HM, Pearcy C, Anez-Bustillos L, Baedorf-Kassis EN, Jhunjhunwala R, Shaefi S, Capers K, Banner-Goodspeed V, Talmor DS, Sauaia A, Yaffe MB. Study of Alteplase for Respiratory Failure in SARS-CoV-2 COVID-19: A Vanguard Multicenter, Rapidly Adaptive, Pragmatic, Randomized Controlled Trial. Chest 2022; 161:710-727. [PMID: 34592318 PMCID: PMC8474873 DOI: 10.1016/j.chest.2021.09.024] [Citation(s) in RCA: 34] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2021] [Revised: 08/31/2021] [Accepted: 09/20/2021] [Indexed: 12/30/2022] Open
Abstract
BACKGROUND Pulmonary vascular microthrombi are a proposed mechanism of COVID-19 respiratory failure. We hypothesized that early administration of tissue plasminogen activator (tPA) followed by therapeutic heparin would improve pulmonary function in these patients. RESEARCH QUESTION Does tPA improve pulmonary function in severe COVID-19 respiratory failure, and is it safe? STUDY DESIGN AND METHODS Adults with COVID-19-induced respiratory failure were randomized from May14, 2020 through March 3, 2021, in two phases. Phase 1 (n = 36) comprised a control group (standard-of-care treatment) vs a tPA bolus (50-mg tPA IV bolus followed by 7 days of heparin; goal activated partial thromboplastin time [aPTT], 60-80 s) group. Phase 2 (n = 14) comprised a control group vs a tPA drip (50-mg tPA IV bolus, followed by tPA drip 2 mg/h plus heparin 500 units/h over 24 h, then heparin to maintain aPTT of 60-80 s for 7 days) group. Patients were excluded from enrollment if they had not undergone a neurologic examination or cross-sectional brain imaging within the previous 4.5 h to rule out stroke and potential for hemorrhagic conversion. The primary outcome was Pao2 to Fio2 ratio improvement from baseline at 48 h after randomization. Secondary outcomes included Pao2 to Fio2 ratio improvement of > 50% or Pao2 to Fio2 ratio of ≥ 200 at 48 h (composite outcome), ventilator-free days (VFD), and mortality. RESULTS Fifty patients were randomized: 17 in the control group and 19 in the tPA bolus group in phase 1 and eight in the control group and six in the tPA drip group in phase 2. No severe bleeding events occurred. In the tPA bolus group, the Pao2 to Fio2 ratio values were significantly (P < .017) higher than baseline at 6 through 168 h after randomization; the control group showed no significant improvements. Among patients receiving a tPA bolus, the percent change of Pao2 to Fio2 ratio at 48 h (16.9% control [interquartile range (IQR), -8.3% to 36.8%] vs 29.8% tPA bolus [IQR, 4.5%-88.7%]; P = .11), the composite outcome (11.8% vs 47.4%; P = .03), VFD (0.0 [IQR, 0.0-9.0] vs 12.0 [IQR, 0.0-19.0]; P = .11), and in-hospital mortality (41.2% vs 21.1%; P = .19) did not reach statistically significant differences when compared with those of control participants. The patients who received a tPA drip did not experience benefit. INTERPRETATION The combination of tPA bolus plus heparin is safe in severe COVID-19 respiratory failure. A phase 3 study is warranted given the improvements in oxygenation and promising observations in VFD and mortality. TRIAL REGISTRY ClinicalTrials.gov; No.: NCT04357730; URL: www. CLINICALTRIALS gov.
Collapse
Affiliation(s)
- Christopher D Barrett
- Department of Surgery, Boston University School of Medicine, Boston, MA; Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA; Koch Institute for Integrative Cancer Research, Center for Precision Cancer Medicine, Departments of Biological Engineering and Biology, Massachusetts Institute of Technology, Cambridge, MA
| | - Hunter B Moore
- Department of Surgery, University of Colorado Denver, Aurora, CO
| | - Ernest E Moore
- Department of Surgery, University of Colorado Denver, Aurora, CO; Ernest E. Moore Shock Trauma Center at Denver Health, Department of Surgery, Denver, CO.
| | - Janice Wang
- Feinstein Institutes for Medical Research, Northwell Health, Manhasset, NY
| | - Negin Hajizadeh
- Feinstein Institutes for Medical Research, Northwell Health, Manhasset, NY
| | - Walter L Biffl
- Division of Trauma/Acute Care Surgery, Department of Surgery, Scripps Memorial Hospital La Jolla, La Jolla, CA
| | - Lawrence Lottenberg
- Department of Surgery, St. Mary's Medical Center, Florida Atlantic University, West Palm Beach, FL
| | - Purvesh R Patel
- Department of Medicine, Baylor College of Medicine, Houston, Dallas, TX
| | - Michael S Truitt
- Department of Surgery, Methodist Dallas Medical Center, Dallas, TX
| | | | - Todd M Bull
- Division of Pulmonary Sciences and Critical Care Medicine, Department of Medicine, University of Colorado Denver, Aurora, CO
| | - Lee Anne Ammons
- Ernest E. Moore Shock Trauma Center at Denver Health, Department of Surgery, Denver, CO
| | - Arsen Ghasabyan
- Ernest E. Moore Shock Trauma Center at Denver Health, Department of Surgery, Denver, CO
| | - James Chandler
- Ernest E. Moore Shock Trauma Center at Denver Health, Department of Surgery, Denver, CO
| | - Ivor S Douglas
- Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Medicine, Denver Health Medical Center, Denver, CO
| | - Eric P Schmidt
- Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Medicine, Denver Health Medical Center, Denver, CO
| | - Peter K Moore
- Division of Pulmonary Sciences and Critical Care Medicine, Department of Medicine, University of Colorado Denver, Aurora, CO
| | | | - Ramona Ramdeo
- Feinstein Institutes for Medical Research, Northwell Health, Manhasset, NY
| | - Robert Borrego
- Division of Pulmonary/Critical Care Medicine, Department of Medicine, Scripps Memorial Hospital La Jolla, La Jolla, CA
| | - Mario Rueda
- Division of Pulmonary/Critical Care Medicine, Department of Medicine, Scripps Memorial Hospital La Jolla, La Jolla, CA
| | - Achal Dhupa
- Division of Trauma/Acute Care Surgery, Department of Surgery, Scripps Memorial Hospital La Jolla, La Jolla, CA
| | - D Scott McCaul
- Division of Trauma/Acute Care Surgery, Department of Surgery, Scripps Memorial Hospital La Jolla, La Jolla, CA
| | - Tala Dandan
- Division of Trauma/Acute Care Surgery, Department of Surgery, Scripps Memorial Hospital La Jolla, La Jolla, CA
| | - Pralay K Sarkar
- Department of Medicine, Baylor College of Medicine, Houston, Dallas, TX
| | - Benazir Khan
- Department of Medicine, Baylor College of Medicine, Houston, Dallas, TX
| | | | - Conner McDaniel
- Department of Surgery, Methodist Dallas Medical Center, Dallas, TX
| | | | | | - Lorenzo Anez-Bustillos
- Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA
| | - Elias N Baedorf-Kassis
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA
| | - Rashi Jhunjhunwala
- Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA
| | - Shahzad Shaefi
- Department of Anesthesia, Critical Care and Pain Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston
| | - Krystal Capers
- Department of Anesthesia, Critical Care and Pain Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston
| | - Valerie Banner-Goodspeed
- Department of Anesthesia, Critical Care and Pain Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston
| | - Daniel S Talmor
- Department of Anesthesia, Critical Care and Pain Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston
| | - Angela Sauaia
- Ernest E. Moore Shock Trauma Center at Denver Health, Department of Surgery, Denver, CO; Colorado School of Public Health and Department of Surgery, University of Colorado Denver, Denver, CO
| | - Michael B Yaffe
- Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA; Koch Institute for Integrative Cancer Research, Center for Precision Cancer Medicine, Departments of Biological Engineering and Biology, Massachusetts Institute of Technology, Cambridge, MA
| |
Collapse
|
66
|
Menter DG, Afshar-Kharghan V, Shen JP, Martch SL, Maitra A, Kopetz S, Honn KV, Sood AK. Of vascular defense, hemostasis, cancer, and platelet biology: an evolutionary perspective. Cancer Metastasis Rev 2022; 41:147-172. [PMID: 35022962 PMCID: PMC8754476 DOI: 10.1007/s10555-022-10019-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/12/2021] [Accepted: 01/04/2022] [Indexed: 01/08/2023]
Abstract
We have established considerable expertise in studying the role of platelets in cancer biology. From this expertise, we were keen to recognize the numerous venous-, arterial-, microvascular-, and macrovascular thrombotic events and immunologic disorders are caused by severe, acute-respiratory-syndrome coronavirus 2 (SARS-CoV-2) infections. With this offering, we explore the evolutionary connections that place platelets at the center of hemostasis, immunity, and adaptive phylogeny. Coevolutionary changes have also occurred in vertebrate viruses and their vertebrate hosts that reflect their respective evolutionary interactions. As mammals adapted from aquatic to terrestrial life and the heavy blood loss associated with placentalization-based live birth, platelets evolved phylogenetically from thrombocytes toward higher megakaryocyte-blebbing-based production rates and the lack of nuclei. With no nuclei and robust RNA synthesis, this adaptation may have influenced viral replication to become less efficient after virus particles are engulfed. Human platelets express numerous receptors that bind viral particles, which developed from archetypal origins to initiate aggregation and exocytic-release of thrombo-, immuno-, angiogenic-, growth-, and repair-stimulatory granule contents. Whether by direct, evolutionary, selective pressure, or not, these responses may help to contain virus spread, attract immune cells for eradication, and stimulate angiogenesis, growth, and wound repair after viral damage. Because mammalian and marsupial platelets became smaller and more plate-like their biophysical properties improved in function, which facilitated distribution near vessel walls in fluid-shear fields. This adaptation increased the probability that platelets could then interact with and engulf shedding virus particles. Platelets also generate circulating microvesicles that increase membrane surface-area encounters and mark viral targets. In order to match virus-production rates, billions of platelets are generated and turned over per day to continually provide active defenses and adaptation to suppress the spectrum of evolving threats like SARS-CoV-2.
Collapse
Affiliation(s)
- David G Menter
- Department of GI Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| | - Vahid Afshar-Kharghan
- Division of Internal Medicine, Benign Hematology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - John Paul Shen
- Department of GI Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Stephanie L Martch
- Department of GI Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Anirban Maitra
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Scott Kopetz
- Department of GI Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Kenneth V Honn
- Department of Pathology, Bioactive Lipids Research Program, Wayne State University, 5101 Cass Ave. 430 Chemistry, Detroit, MI, 48202, USA
- Department of Pathology, Wayne State University School of Medicine, 431 Chemistry Bldg, Detroit, MI, 48202, USA
- Cancer Biology Division, Wayne State University School of Medicine, 431 Chemistry Bldg, Detroit, MI, 48202, USA
| | - Anil K Sood
- Department of Gynecologic Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Center for RNA Interference and Non-Coding RNA, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
- Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| |
Collapse
|
67
|
Significance of Immune Status of SARS-CoV-2 Infected Patients in Determining the Efficacy of Therapeutic Interventions. J Pers Med 2022; 12:jpm12030349. [PMID: 35330349 PMCID: PMC8955701 DOI: 10.3390/jpm12030349] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2021] [Revised: 02/12/2022] [Accepted: 02/14/2022] [Indexed: 12/15/2022] Open
Abstract
Coronavirus disease 2019 (COVID-19) is now being investigated for its distinctive patterns in the course of disease development which can be indicated with miscellaneous immune responses in infected individuals. Besides this series of investigations on the pathophysiology of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), significant fundamental immunological and physiological processes are indispensable to address clinical markers of COVID-19 disease and essential to identify or design effective therapeutics. Recent developments in the literature suggest that deficiency of type I interferon (IFN) in serum samples can be used to represent a severe progression of COVID-19 disease and can be used as the basis to develop combined immunotherapeutic strategies. Precise control over inflammatory response is a significant aspect of targeting viral infections. This account presents a brief review of the pathophysiological characteristics of the SARS-CoV-2 virus and the understanding of the immune status of infected patients. We further discuss the immune system’s interaction with the SARS-CoV-2 virus and their subsequent involvement of dysfunctional immune responses during the progression of the disease. Finally, we highlight some of the implications of the different approaches applicable in developing promising therapeutic interventions that redirect immunoregulation and viral infection.
Collapse
|
68
|
Kell DB, Laubscher GJ, Pretorius E. A central role for amyloid fibrin microclots in long COVID/PASC: origins and therapeutic implications. Biochem J 2022; 479:537-559. [PMID: 35195253 PMCID: PMC8883497 DOI: 10.1042/bcj20220016] [Citation(s) in RCA: 115] [Impact Index Per Article: 57.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2022] [Revised: 02/08/2022] [Accepted: 02/09/2022] [Indexed: 12/15/2022]
Abstract
Post-acute sequelae of COVID (PASC), usually referred to as 'Long COVID' (a phenotype of COVID-19), is a relatively frequent consequence of SARS-CoV-2 infection, in which symptoms such as breathlessness, fatigue, 'brain fog', tissue damage, inflammation, and coagulopathies (dysfunctions of the blood coagulation system) persist long after the initial infection. It bears similarities to other post-viral syndromes, and to myalgic encephalomyelitis/chronic fatigue syndrome (ME/CFS). Many regulatory health bodies still do not recognize this syndrome as a separate disease entity, and refer to it under the broad terminology of 'COVID', although its demographics are quite different from those of acute COVID-19. A few years ago, we discovered that fibrinogen in blood can clot into an anomalous 'amyloid' form of fibrin that (like other β-rich amyloids and prions) is relatively resistant to proteolysis (fibrinolysis). The result, as is strongly manifested in platelet-poor plasma (PPP) of individuals with Long COVID, is extensive fibrin amyloid microclots that can persist, can entrap other proteins, and that may lead to the production of various autoantibodies. These microclots are more-or-less easily measured in PPP with the stain thioflavin T and a simple fluorescence microscope. Although the symptoms of Long COVID are multifarious, we here argue that the ability of these fibrin amyloid microclots (fibrinaloids) to block up capillaries, and thus to limit the passage of red blood cells and hence O2 exchange, can actually underpin the majority of these symptoms. Consistent with this, in a preliminary report, it has been shown that suitable and closely monitored 'triple' anticoagulant therapy that leads to the removal of the microclots also removes the other symptoms. Fibrin amyloid microclots represent a novel and potentially important target for both the understanding and treatment of Long COVID and related disorders.
Collapse
Affiliation(s)
- Douglas B. Kell
- Department of Biochemistry and Systems Biology, Institute of Systems, Molecular and Integrative Biology, Faculty of Health and Life Sciences, University of Liverpool, Liverpool L69 7ZB, U.K
- The Novo Nordisk Foundation Centre for Biosustainability, Technical University of Denmark, Kemitorvet 200, 2800 Kgs Lyngby, Denmark
- Department of Physiological Sciences, Faculty of Science, Stellenbosch University, Stellenbosch Private Bag X1 Matieland, 7602, South Africa
| | | | - Etheresia Pretorius
- Department of Physiological Sciences, Faculty of Science, Stellenbosch University, Stellenbosch Private Bag X1 Matieland, 7602, South Africa
| |
Collapse
|
69
|
Martín LH, Sainz-Gil M, Navarro-García E, Salado-Valdivieso I, Sanz-Fadrique R. Thromboembolism and Oral Contraceptives During the COVID-19 Pandemic: A Disproportionality Analysis Within the Spanish Pharmacovigilance Database. Drugs Real World Outcomes 2022; 9:211-218. [PMID: 35146697 PMCID: PMC8831020 DOI: 10.1007/s40801-021-00282-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/29/2021] [Indexed: 01/19/2023] Open
Abstract
Background Thromboembolic events (TEs) are known to be a severe complication for COVID-19. They are associated with a systemic inflammatory response syndrome with coagulation cascade activation. Objective The aim of this study was to determine a potential association between the COVID-19
pandemic and the increment of the risk of suspected TEs in women on systemic hormonal contraceptives (SHCs). Patients and Methods This study utilised a case/non-case approach in the Spanish Pharmacovigilance Database, which includes more than 290,000 cases of suspected adverse drug reactions (ADRs). The reporting odds ratio (ROR) was calculated during an initial pandemic period in 2020 compared with a pre-pandemic period in 2019 and an additional control period in 2018. Results While there was a decreased number of ADR notifications for any medications and for any type of ADR in patients on SHCs during the pandemic period, the TE ROR for all SHCs was higher in the 2020 pandemic period [ROR = 11.8 (5.6–24.7)] relative to the pre-pandemic period in 2019 [ROR = 6.3 (3.2–12.5)] and the additional control period in 2018 [ROR = 4.6. (2.1–9.9)]. In contrast, ROR for progestogen-only contraceptives was lower during the pandemic as compared with the two control periods. Conclusion The reported disproportionality of TEs in women on SHCs rose during the pandemic period. This suggests a potential interaction of the drug (SHC) with COVID-19, which led to an increased risk of TEs in women exposed to both factors. This should be taken into consideration in the context of the COVID-19 pandemic.
Collapse
Affiliation(s)
- Luis H Martín
- Centre for Drug Safety (CESME), Faculty of Medicine, Valladolid University, Valladolid, Spain.,Centre for Pharmacovigilance of Castilla y León, Valladolid, Spain.,Department of Cellular Biology, Histology and Pharmacology, Valladolid University, Valladolid, Spain
| | - María Sainz-Gil
- Centre for Drug Safety (CESME), Faculty of Medicine, Valladolid University, Valladolid, Spain.,Centre for Pharmacovigilance of Castilla y León, Valladolid, Spain.,Department of Cellular Biology, Histology and Pharmacology, Valladolid University, Valladolid, Spain
| | - Ester Navarro-García
- Centre for Drug Safety (CESME), Faculty of Medicine, Valladolid University, Valladolid, Spain. .,De La Plana University Hospital, Castellón, Spain.
| | - Inés Salado-Valdivieso
- Centre for Drug Safety (CESME), Faculty of Medicine, Valladolid University, Valladolid, Spain.,Department of Cellular Biology, Histology and Pharmacology, Valladolid University, Valladolid, Spain
| | - Rosario Sanz-Fadrique
- Centre for Drug Safety (CESME), Faculty of Medicine, Valladolid University, Valladolid, Spain.,Centre for Pharmacovigilance of Castilla y León, Valladolid, Spain
| |
Collapse
|
70
|
Ullah MF, Ali Y, Khan MR, Khan IU, Yan B, Ijaz Khan M, Malik M. A review of COVID-19: Treatment strategies and CRISPR/Cas9 gene editing technology approaches to the coronavirus disease. Saudi J Biol Sci 2022; 29:860-871. [PMID: 34658640 PMCID: PMC8511869 DOI: 10.1016/j.sjbs.2021.10.020] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2021] [Revised: 09/12/2021] [Accepted: 10/05/2021] [Indexed: 12/12/2022] Open
Abstract
The new coronavirus SARS-CoV-2 pandemic has put the world on lockdown for the first time in decades. This has wreaked havoc on the global economy, put additional burden on local and global public health resources, and, most importantly, jeopardised human health. CRISPR stands for Clustered Regularly Interspaced Short Palindromic Repeats, and the CRISPR associated (Cas) protein (CRISPR/Cas) was identified to have structures in E. coli. The most modern of these systems is CRISPR/Cas. Editing the genomes of plants and animals took several years and cost hundreds of thousands of dollars until the CRISPR approach was discovered in 2012. As a result, CRISPR/Cas has piqued the scientific community's attention, particularly for disease diagnosis and treatment, because it is faster, less expensive, and more precise than previous genome editing technologies. Data from gene mutations in specific patients gathered using CRISPR/Cas can aid in the identification of the best treatment strategy for each patient, as well as other research domains such as coronavirus replication in cell culture, such as SARS-CoV2. The implications of the most prevalent driver mutations, on the other hand, are often unknown, making treatment interpretation difficult. For detecting a wide range of target genes, the CRISPR/Cas categories provide highly sensitive and selective tools. Genome-wide association studies are a relatively new strategy to discovering genes involved in human disease when it comes to the next steps in genomic research. Furthermore, CRISPR/Cas provides a method for modifying non-coding portions of the genome, which will help advance whole genome libraries by speeding up the analysis of these poorly defined parts of the genome.
Collapse
Affiliation(s)
- Muhammad Farhat Ullah
- Genome Editing & Sequencing Lab, National Centre for Bioinformatics, Quaid-i-Azam University Islamabad, Pakistan
| | - Yasir Ali
- Genome Editing & Sequencing Lab, National Centre for Bioinformatics, Quaid-i-Azam University Islamabad, Pakistan
| | - Muhammad Ramzan Khan
- Genome Editing & Sequencing Lab, National Centre for Bioinformatics, Quaid-i-Azam University Islamabad, Pakistan
| | - Inam Ullah Khan
- University of Sheffield, Department of Chemical and Biological Engineering, Arts Tower Western Bank, Sheffield, S102TN, The University of Sheffield, Manchester, UK
| | - Bing Yan
- Department of Pharmacy, The First Affiliated Hospital of Huzhou University, Huzhou 313000, PR China
| | - M. Ijaz Khan
- Department of Mathematics and Statistics, Riphah International University, I-14, Islamabad 44000, Pakistan
| | - M.Y. Malik
- Department of Mathematics, College of Sciences, King Khalid University, Abha 61413, Saudi Arabia
| |
Collapse
|
71
|
El-Qutob D, Alvarez-Arroyo L, Barreda I, Nieto M, Pin M, Poveda-Andrés JL, Carrera-Hueso FJ. High incidence of pulmonary thromboembolism in hospitalized SARS-CoV-2 infected patients despite thrombo-prophylaxis. Heart Lung 2022; 53:77-82. [PMID: 35180507 PMCID: PMC8823955 DOI: 10.1016/j.hrtlng.2022.02.003] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2021] [Revised: 02/02/2022] [Accepted: 02/04/2022] [Indexed: 01/02/2023]
Abstract
Background Objectives Methods Results Conclusion
Collapse
Affiliation(s)
- D El-Qutob
- Unit of Allergy, Universitary Hospital of La Plana in Vila-Real, Carretera Vila-Real-Burriana Km. 0.5 Vila-Real Castellon 12540, Spain.
| | - L Alvarez-Arroyo
- Service of Pharmacy, Universitary Hospital of La Plana in Vila-Real, Spain; Doctoral Program of Pharmacy, Universitary of Granada, Spain
| | - I Barreda
- Section of Neurophysiology, Universitary Hospital of La Plana in Vila-Real, Spain
| | - M Nieto
- Unit of Allergy, Universitary Hospital of La Plana in Vila-Real, Carretera Vila-Real-Burriana Km. 0.5 Vila-Real Castellon 12540, Spain
| | - M Pin
- Unit of Nephrology, Universitary Hospital of La Plana in Vila-Real, Spain
| | | | - F J Carrera-Hueso
- Service of Pharmacy, Universitary Hospital of La Plana in Vila-Real, Spain
| |
Collapse
|
72
|
Barrett CD, Moore HB, Moore EE, Benjamin Christie D, Orfanos S, Anez‐Bustillos L, Jhunjhunwala R, Hussain S, Shaefi S, Wang J, Hajizadeh N, Baedorf‐Kassis EN, Al‐Shammaa A, Capers K, Banner‐Goodspeed V, Wright FL, Bull T, Moore PK, Nemec H, Thomas Buchanan J, Nonnemacher C, Rajcooar N, Ramdeo R, Yacoub M, Guevara A, Espinal A, Hattar L, Moraco A, McIntyre R, Talmor DS, Sauaia A, Yaffe MB. MUlticenter STudy of tissue plasminogen activator (alteplase) use in COVID-19 severe respiratory failure (MUST COVID): A retrospective cohort study. Res Pract Thromb Haemost 2022; 6:e12669. [PMID: 35341072 PMCID: PMC8935535 DOI: 10.1002/rth2.12669] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2021] [Revised: 12/28/2021] [Accepted: 01/16/2022] [Indexed: 01/08/2023] Open
Abstract
Background Few therapies exist to treat severe COVID-19 respiratory failure once it develops. Given known diffuse pulmonary microthrombi on autopsy studies of COVID-19 patients, we hypothesized that tissue plasminogen activator (tPA) may improve pulmonary function in COVID-19 respiratory failure. Methods A multicenter, retrospective, observational study of patients with confirmed COVID-19 and severe respiratory failure who received systemic tPA (alteplase) was performed. Seventy-nine adults from seven medical centers were included in the final analysis after institutional review boards' approval; 23 were excluded from analysis because tPA was administered for pulmonary macroembolism or deep venous thrombosis. The primary outcome was improvement in the PaO2/FiO2 ratio from baseline to 48 h after tPA. Linear mixed modeling was used for analysis. Results tPA was associated with significant PaO2/FiO2 improvement at 48 h (estimated paired difference = 23.1 ± 6.7), which was sustained at 72 h (interaction term p < 0.00). tPA administration was also associated with improved National Early Warning Score 2 scores at 24, 48, and 72 h after receiving tPA (interaction term p = 0.00). D-dimer was significantly elevated immediately after tPA, consistent with lysis of formed clot. Patients with declining respiratory status preceding tPA administration had more marked improvement in PaO2/FiO2 ratios than those who had poor but stable (not declining) respiratory status. There was one intracranial hemorrhage, which occurred within 24 h following tPA administration. Conclusions These data suggest tPA is associated with significant improvement in pulmonary function in severe COVID-19 respiratory failure, especially in patients whose pulmonary function is in decline, and has an acceptable safety profile in this patient population.
Collapse
|
73
|
Tian F, Wang J, Xi X, Sun X, He M, Zhao C, Feng F, Wang H, Sun W, Mao L, Hu X, Yuan H. Efficacy and safety of short-wave diathermy treatment for moderate COVID-19 patients: a prospective, double-blind, randomized controlled clinical study. Eur J Phys Rehabil Med 2022; 58:137-143. [PMID: 34042412 PMCID: PMC9980486 DOI: 10.23736/s1973-9087.21.06892-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
BACKGROUND Millions of human beings have suffered in the epidemic of Coronavirus disease 2019 (COVID-19), but until now the effective treatment methods have been limited. AIM This study aimed to evaluate the efficacy and safety of short-wave diathermy (SWD) treatment for moderate COVID-19 patients. DESIGN A prospective, double-blind, randomized controlled clinical study. SETTING Inpatients Unit of a COVID-19 specialized hospital. POPULATION Forty-two patients with moderate COVID-19 were randomly allocated at a 2:1 ratio to two groups: the SWD group and the control group. METHODS Participants of the SWD group received SWD treatment, and participants of the control group received placebo SWD treatment for one session per day, 10 minutes per session, for no more than 14 days. Both groups were given standard care treatment. Primary outcome was the rate of clinical improvement according to a seven-category ordinal scale. Secondary outcomes included the rate of computed tomography (CT) improvement and the rate of potential adverse events. RESULTS Clinical improvement occurred in 92.6% of patients in the SWD group by day 14 compared with 69.2% of patients in the control group (P=0.001). The Cox model indicated that the SWD group had a higher clinical improvement probability than the control group (hazard ratio: 3.045; 95% CI: 1.391-6.666; P=0.005). Similarly, CT improvement occurred in 85.2% of patients in the SWD group and 46.2% of patients in the control group respectively by day 14 (P=0.001). The Cox model indicated SWD group had a higher CT improvement probability than control group (hazard ratio: 3.720; 95% CI: 1.486-9.311; P=0.005). There was no significant difference in adverse events between the SWD group and the control group (2 of 27 [7.4%] SWD vs. 1 of 13 [7.7%] control, P=1.000), the most frequent of which were headache (1 of 27 [3.7%] SWD vs. 1 of 13 [7.7%] control patients) and dizziness (1 of 27 [3.7%] SWD vs. 0 of 13 [0%] control patients). CONCLUSIONS SWD is a valid and reliable adjuvant therapy with a favorable safety profile for moderate COVID-19 patients. CLINICAL REHABILITATION IMPACT Clinically relevant information is lacking regarding the efficacy and safety of SWD for patients with COVID-19. This study provides the first evidence that SWD is a promising adjuvant therapy for COVID-19.
Collapse
Affiliation(s)
- Fei Tian
- Department of Rehabilitation Medicine, Xijing Hospital, the Fourth Military Medical University, Xi'an, China
| | - Jin Wang
- Department of Traditional Chinese Medicine.,Hospital of Joint Logistic Support Force, Lanzhou, China
| | - Xiao Xi
- Department of Rehabilitation Medicine, Xijing Hospital, the Fourth Military Medical University, Xi'an, China
| | - Xiaolong Sun
- Department of Rehabilitation Medicine, Xijing Hospital, the Fourth Military Medical University, Xi'an, China
| | - Miao He
- Department of Traditional Chinese Medicine.,Hospital of Joint Logistic Support Force, Lanzhou, China
| | - Chenguang Zhao
- Department of Rehabilitation Medicine, Xijing Hospital, the Fourth Military Medical University, Xi'an, China
| | - Feng Feng
- Department of Rehabilitation Medicine, Xijing Hospital, the Fourth Military Medical University, Xi'an, China
| | - Hongbin Wang
- Department of Rehabilitation Medicine, Xijing Hospital, the Fourth Military Medical University, Xi'an, China
| | - Wei Sun
- Department of Rehabilitation Medicine, Xijing Hospital, the Fourth Military Medical University, Xi'an, China
| | - Li Mao
- Department of Rehabilitation Medicine, Xijing Hospital, the Fourth Military Medical University, Xi'an, China
| | - Xu Hu
- Department of Rehabilitation Medicine, Xijing Hospital, the Fourth Military Medical University, Xi'an, China
| | - Hua Yuan
- Department of Rehabilitation Medicine, Xijing Hospital, the Fourth Military Medical University, Xi'an, China -
| |
Collapse
|
74
|
Magoon R, Choudhary N, Jose J. Fibrinolysis and D-dimer in COVID-19: A Twisted Plot! Indian J Crit Care Med 2022; 26:164-166. [PMID: 35712729 PMCID: PMC8857708 DOI: 10.5005/jp-journals-10071-24105] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/02/2022] Open
Abstract
Coronavirus disease-2019 (COVID-19) and the associated pathophysiological perturbations continue to bewilder the fraternity at large. In this context, the thromboembolic predisposition in COVID-19 has particularly emerged as a matter of an ardent debate. The index commentary aims to present an account of the recent developments in the understanding of the immunothrombosis in the enigmatical setting of COVID-19. How to cite this article Magoon R, Choudhary N, Jose J. Fibrinolysis and D-dimer in COVID-19: A Twisted Plot! Indian J Crit Care Med 2022;26(2):164-166.
Collapse
Affiliation(s)
- Rohan Magoon
- Department of Anaesthesia, Atal Bihari Vajpayee Institute of Medical Sciences and Dr Ram Manohar Lohia Hospital, New Delhi, India
| | - Nitin Choudhary
- Department of Anaesthesia, Atal Bihari Vajpayee Institute of Medical Sciences and Dr Ram Manohar Lohia Hospital, New Delhi, India
| | - Jes Jose
- Department of Cardiac Anesthesiology, Sri Jayadeva Institute of Cardiovascular Sciences and Research, Bengaluru, Karnataka, India
| |
Collapse
|
75
|
Angelini DE, Kaatz S, Rosovsky R, Zon RL, Pillai S, Robertson WE, Elavalakanar P, Patell R, Khorana A. COVID-19 and venous thromboembolism: A narrative review. Res Pract Thromb Haemost 2022; 6:e12666. [PMID: 35224417 PMCID: PMC8847419 DOI: 10.1002/rth2.12666] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2021] [Revised: 12/27/2021] [Accepted: 01/02/2022] [Indexed: 12/15/2022] Open
Abstract
COVID-19 (severe acute respiratory syndrome coronavirus 2 [SARS-CoV-2]) is associated with coagulopathy through numerous mechanisms. The reported incidence of venous thromboembolism (VTE) in hospitalized patients with COVID-19 has varied widely, and several meta-analyses have been performed to assess the overall prevalence of VTE. The novelty of this coronavirus strain along with its unique mechanisms for microvascular and macrovascular thrombosis has led to uncertainty as to how to diagnose, prevent, and treat thrombosis in patients affected by this virus. This review discusses the epidemiology and pathophysiology of thrombosis in the setting of SARS-CoV-2 infection along with an updated review on the preventative and treatment strategies for VTE associated with SARS-CoV-2 infection.
Collapse
Affiliation(s)
- Dana E. Angelini
- Department of Hematology and Medical OncologyTaussig Cancer InstituteCleveland Clinic FoundationClevelandOhioUSA
| | - Scott Kaatz
- Division of Hospital MedicineHenry Ford HospitalDetroitMichiganUSA
| | - Rachel P. Rosovsky
- Department of MedicineMassachusetts General HospitalBostonMassachusettsUSA
| | - Rebecca L. Zon
- Dana Farber Cancer Institute and Massachusetts General BrighamBostonMassachusettsUSA
| | - Shreejith Pillai
- Division of Hospital MedicineHenry Ford HospitalDetroitMichiganUSA
| | - William E. Robertson
- National Blood Clot AllianceDepartment of Emergency HealthcareDumke College of Health ProfessionsWeber State UniversityOgdenUtahUSA
| | - Pavania Elavalakanar
- Division of Hematology and Hematologic MalignanciesBeth Israel Deaconess Medical CenterBostonMassachusettsUSA
| | - Rushad Patell
- Division of Hematology and Hematologic MalignanciesBeth Israel Deaconess Medical CenterBostonMassachusettsUSA
| | - Alok Khorana
- Department of Hematology and Medical OncologyTaussig Cancer InstituteCleveland Clinic FoundationClevelandOhioUSA
| |
Collapse
|
76
|
Chu X, Zhang GF, Zheng YK, Zhong YG, Wen L, Zeng P, Fu CY, Tong XL, Long YF, Li J, Liu YL, Chang ZG, Xi H. Clinical features and risk factors of severely and critically ill patients with COVID-19. World J Clin Cases 2022; 10:840-855. [PMID: 35127900 PMCID: PMC8790448 DOI: 10.12998/wjcc.v10.i3.840] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/02/2021] [Revised: 10/20/2021] [Accepted: 12/23/2021] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND As of June 1, 2020, over 370000 coronavirus disease 2019 (COVID-19) deaths have been reported to the World Health Organization. However, the risk factors for patients with moderate-to-severe or severe-to-critical COVID-19 remain unclear.
AIM To explore the characteristics and predictive markers of severely and critically ill patients with COVID-19.
METHODS A retrospective study was conducted at the B11 Zhongfaxincheng campus and E1-3 Guanggu campus of Tongji Hospital affiliated with Huazhong University of Science and Technology in Wuhan. Patients with COVID-19 admitted from 1st February 2020 to 8th March 2020 were enrolled and categorized into 3 groups: The moderate group, severe group and critically ill group. Epidemiological data, demographic data, clinical symptoms and outcomes, complications, laboratory tests and radiographic examinations were collected retrospectively from the hospital information system and then compared between groups.
RESULTS A total of 126 patients were enrolled. There were 59 in the moderate group, 49 in the severe group, and 18 in the critically ill group. Multivariate logistic regression analysis showed that age [odd ratio (OR) = 1.055, 95% (confidence interval) CI: 1.099-1.104], elevated neutrophil-to-lymphocyte ratios (OR = 4.019, 95%CI: 1.045-15.467) and elevated high-sensitivity cardiac troponin I (OR = 10.126, 95%CI: 1.088 -94.247) were high-risk factors.
CONCLUSION The following indicators can help clinicians identify patients with severe COVID-19 at an early stage: age, an elevated neutrophil-to-lymphocyte ratio and high sensitivity cardiac troponin I.
Collapse
Affiliation(s)
- Xin Chu
- Department of Surgical Intensive Care Medicine, Beijing Hospital, National Center of Gerontology; Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing 100730, China
| | - Gui-Fang Zhang
- The Key Laboratory of Geriatrics, Beijing Institute of Geriatrics, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing Hospital /National Center of Gerontology of National Health Commission, Beijing 100730, China
| | - Yong-Ke Zheng
- Department of Intensive Care Unit, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou 310006, Zhejiang Province, China
| | - Yi-Gang Zhong
- Department of Cardiology, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou 310006, Zhejiang Province, China
| | - Li Wen
- Department of Emergency, Beijing Hospital, National Center of Gerontology; Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing 100730, China
| | - Ping Zeng
- The Key Laboratory of Geriatrics, Beijing Institute of Geriatrics, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing Hospital /National Center of Gerontology of National Health Commission, Beijing 100730, China
| | - Chun-Yi Fu
- Department of Emergency, Beijing Hospital, National Center of Gerontology; Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing 100730, China
| | - Xun-Liang Tong
- Department of Respiratory and Critical Care Medicine, Beijing Hospital, National Center of Gerontology; Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing 100730, China
| | - Yun-Fei Long
- Department of Neurology, Beijing Hospital, National Center of Gerontology; Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing 100730, China
| | - Jing Li
- Department of Cardiology, Beijing Hospital, National Center of Gerontology; Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing 100730, China
| | - Ya-Lin Liu
- Department of Surgical Intensive Care Medicine, Beijing Hospital, National Center of Gerontology; Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing 100730, China
| | - Zhi-Gang Chang
- Department of Surgical Intensive Care Medicine, Beijing Hospital, National Center of Gerontology; Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing 100730, China
| | - Huan Xi
- Department of Geriatrics, Beijing Hospital, National Center of Gerontology, National Health Commission; Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing 100730, China
| |
Collapse
|
77
|
Direct comparison of different therapeutic cell types susceptibility to inflammatory cytokines associated with COVID-19 acute lung injury. Stem Cell Res Ther 2022; 13:20. [PMID: 35033181 PMCID: PMC8760881 DOI: 10.1186/s13287-021-02699-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2021] [Accepted: 12/29/2021] [Indexed: 12/13/2022] Open
Abstract
Background Although 90% of infections with the novel coronavirus 2 (COVID-19) are mild, many patients progress to acute respiratory distress syndrome (ARDS) which carries a high risk of mortality. Given that this dysregulated immune response plays a key role in the pathology of COVID-19, several clinical trials are underway to evaluate the effect of immunomodulatory cell therapy on disease progression. However, little is known about the effect of ARDS associated pro-inflammatory mediators on transplanted stem cell function and survival, and any deleterious effects could undermine therapeutic efficacy. As such, we assessed the impact of inflammatory cytokines on the viability, and paracrine profile (extracellular vesicles) of bone marrow-derived mesenchymal stromal cells, heart-derived cells, and umbilical cord-derived mesenchymal stromal cells. Methods All cell products were manufactured and characterized to established clinical release standards by an accredited clinical cell manufacturing facility. Cytokines and Extracellular vesicles in the cell conditioned media were profiled using proteomic array and nanoparticle tracking analysis. Using a survey of the clinical literature, 6 cytotoxic cytokines implicated in the progression of COVID-19 ARDS. Flow cytometry was employed to determine receptor expression of these 6 cytokines in three cell products. Based on clinical survey and flow cytometry data, a cytokine cocktail that mimics cytokine storm seen in COVID-19 ARDS patients was designed and the impact on cytokine cocktail on viability and paracrine secretory ability of cell products were assessed using cell viability and nanoparticle tracking analysis. Results Flow cytometry revealed the presence of receptors for all cytokines but IL-6, which was subsequently excluded from further experimentation. Despite this widespread expression, exposure of each cell type to individual cytokines at doses tenfold greater than observed clinically or in combination at doses associated with severe ARDS did not alter cell viability or extracellular vesicle character/production in any of the 3 cell products. Conclusions The paracrine production and viability of the three leading cell products under clinical evaluation for the treatment of severe COVID-19 ARDS are not altered by inflammatory mediators implicated in disease progression. Supplementary Information The online version contains supplementary material available at 10.1186/s13287-021-02699-7.
Collapse
|
78
|
Kurushina OV, Barulin AE. Central Nervous System Lesions in COVID-19. NEUROSCIENCE AND BEHAVIORAL PHYSIOLOGY 2022; 51:1222-1227. [PMID: 35001988 PMCID: PMC8720549 DOI: 10.1007/s11055-021-01183-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 12/02/2020] [Accepted: 01/12/2021] [Indexed: 01/08/2023]
Abstract
This review discusses current data on CNS lesions in infections with the new coronavirus that causes COVID-19. The pathogenetic mechanisms leading to infection of the brain and spinal cord are presented. Published studies and meta-analyses published by autumn 2020 are assessed. The authors present results from their own clinical observations of various types of CNS infection in COVID-19 virus disease. Particular attention is paid to certain common forms of CNS lesion such as encephalitis, cerebrovascular pathology, and headache. A form of acute hemorrhagic necrotic encephalopathy is identified, which is a quite rare but fatal pathology comorbid with COVID-19. A description of our own clinical observation of acute necrotic encephalopathy is presented. The importance of further studies of the effects of the coronavirus on the CNS and possible approaches to treatment and rehabilitation of this category of patients is emphasized.
Collapse
Affiliation(s)
- O. V. Kurushina
- Volgograd State Medical University, Russian Ministry of Health, Volgograd, Russia
| | - A. E. Barulin
- Volgograd State Medical University, Russian Ministry of Health, Volgograd, Russia
| |
Collapse
|
79
|
Veterano C, Antunes I, Coelho A, Silva I, Almeida R, Pinto PS. Endovascular Treatment of Renal Artery Thrombosis in Living-Donor Kidney Transplant Recipient With Severe COVID-19 Disease. J Endovasc Ther 2022; 29:966-970. [PMID: 34994219 DOI: 10.1177/15266028211068756] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PURPOSE Coronavirus disease 2019 (COVID-19) patients have a higher prevalence of micro-and macrovascular thrombotic events. However, the underlying mechanism for the increased thrombotic risk is not completely understood. Solid organ transplant recipients infected with SARS-CoV-2 may have an exponential increase in thrombotic risk and the best management strategy is unknown. CASE REPORT A female kidney transplant recipient presented with allograft's renal artery thrombosis after a recent COVID-19 infection. Due to the risk of kidney failure or exclusion, catheter directed thrombolysis was performed. Residual thrombus was excluded using an endoprosthesis with an excellent result. There were no adverse events and kidney function improved. CONCLUSION This paper reports the endovascular treatment of renal artery thrombosis in a living-donor kidney transplant recipient with severe COVID-19 disease.
Collapse
Affiliation(s)
- Carlos Veterano
- Department of Angiology and Vascular Surgery, Centro Hospitalar Universitário do Porto, Porto, Portugal
| | - Inês Antunes
- Department of Angiology and Vascular Surgery, Centro Hospitalar Universitário do Porto, Porto, Portugal
| | - Andreia Coelho
- Department of Angiology and Vascular Surgery, Centro Hospitalar Universitário do Porto, Porto, Portugal
| | - Ivone Silva
- Department of Angiology and Vascular Surgery, Centro Hospitalar Universitário do Porto, Porto, Portugal.,Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, Porto, Portugal.,Kidney Transplant Program, Department of Transplantation, Centro Hospitalar Universitário do Porto, Porto, Portugal
| | - Rui Almeida
- Department of Angiology and Vascular Surgery, Centro Hospitalar Universitário do Porto, Porto, Portugal.,Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, Porto, Portugal.,Kidney Transplant Program, Department of Transplantation, Centro Hospitalar Universitário do Porto, Porto, Portugal
| | - Pedro Sá Pinto
- Department of Angiology and Vascular Surgery, Centro Hospitalar Universitário do Porto, Porto, Portugal.,Kidney Transplant Program, Department of Transplantation, Centro Hospitalar Universitário do Porto, Porto, Portugal
| |
Collapse
|
80
|
Strategy for Locating People to Reduce the Transmission of COVID-19 Using Different Interference Measures. SUSTAINABILITY 2022. [DOI: 10.3390/su14010529] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
COVID-19 is generally transmitted from person to person through small droplets of saliva emitted when talking, sneezing, coughing, or breathing. For this reason, social distancing and ventilation have been widely emphasized to control the pandemic. The spread of the virus has brought with it many challenges in locating people under distance constraints. The effects of wakes between turbines have been studied extensively in the literature on wind energy, and there are well-established interference models. Does this apply to the propagation functions of the virus? In this work, a parallel relationship between the two problems is proposed. A mixed-integer linear programming (MIP) model and a mixed-integer quadratic programming model (MIQP) are formulated to locate people to avoid the spread of COVID-19. Both models were constructed according to the distance constraints proposed by the World Health Organization and the interference functions representing the effects of wake between turbines. Extensive computational tests show that people should not be less than two meters apart, in agreement with the adapted Wells–Riley model, which indicates that 1.6 to 3.0 m (5.2 to 9.8 ft) is the safe social distance when considering the aerosol transmission of large droplets exhaled when speaking, while the distance can be up to 8.2 m (26 ft) if all the droplets in a calm air environment are taken into account.
Collapse
|
81
|
Hajra A, Torrado J, Alviar CL, Bangalore S, Keller N, Faillace R, Sokol S. COVID-19-induced latent relapsing hypercoagulable state in the absence of persistent viral infection. SAGE Open Med Case Rep 2022; 10:2050313X221113934. [PMID: 35899245 PMCID: PMC9310331 DOI: 10.1177/2050313x221113934] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2022] [Accepted: 06/29/2022] [Indexed: 11/21/2022] Open
Abstract
Hypercoagulability in coronavirus disease 2019 infection is already a known fact. But in this article, we have discussed a unique case where the patient had suffered from relapsing thrombus formation. This report describes the case of a patient who presented with chronic coronavirus disease 2019-induced recurrent thrombi refractory to multiple antithrombotic regimens because of multiple recurrent inflammatory flares without any evidence of chronic persistent viral infection. The patient was treated with anticoagulation and anti-inflammatory medications. Still, he had repeated episodes of right ventricular thrombus. Coronavirus disease 2019 can provoke a severe relapsing hypercoagulable state without evidence of persisting viral infection. Rebound inflammatory flares rather than viral recurrence may play a trigger.
Collapse
Affiliation(s)
- Adrija Hajra
- Department of Internal Medicine, New York City Health and Hospitals-Jacobi Medical Center, New York, NY, USA
| | - Juan Torrado
- Department of Internal Medicine, New York City Health and Hospitals-Jacobi Medical Center, New York, NY, USA
| | - Carlos L Alviar
- Department of Cardiology, New York City Health and Hospitals-Bellevue, New York, NY, USA
| | - Sripal Bangalore
- Department of Cardiology, New York City Health and Hospitals-Bellevue, New York, NY, USA
| | - Norma Keller
- Department of Cardiology, New York City Health and Hospitals-Bellevue, New York, NY, USA
| | - Robert Faillace
- Department of Internal Medicine, New York City Health and Hospitals-Jacobi Medical Center, New York, NY, USA
| | - Seth Sokol
- Division of Cardiology, Department of Medicine, New York City Health and Hospitals-Jacobi Medical Center, New York, NY, USA
| |
Collapse
|
82
|
Castro P, Palomo M, Moreno-Castaño AB, Fernández S, Torramadé-Moix S, Pascual G, Martinez-Sanchez J, Richardson E, Téllez A, Nicolas JM, Carreras E, Richardson PG, Badimon JJ, Escolar G, Diaz-Ricart M. Is the Endothelium the Missing Link in the Pathophysiology and Treatment of COVID-19 Complications? Cardiovasc Drugs Ther 2022; 36:547-560. [PMID: 34097193 PMCID: PMC8181544 DOI: 10.1007/s10557-021-07207-w] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 05/24/2021] [Indexed: 02/08/2023]
Abstract
Patients with COVID-19 present a wide spectrum of disease severity, from asymptomatic cases in the majority to serious disease leading to critical care and even death. Clinically, four different scenarios occur within the typical disease timeline: first, an incubation and asymptomatic period; second, a stage with mild symptoms due mainly to the virus itself; third, in up to 20% of the patients, a stage with severe symptoms where a hyperinflammatory response with a cytokine storm driven by host immunity induces acute respiratory distress syndrome; and finally, a post-acute sequelae (PASC) phase, which present symptoms that can range from mild or annoying to actually quite incapacitating. Although the most common manifestation is acute respiratory failure of the lungs, other organs are also frequently involved. The clinical manifestations of the COVID-19 infection support a key role for endothelial dysfunction in the pathobiology of this condition. The virus enters into the organism via its interaction with angiotensin-converting enzyme 2-receptor that is present prominently in the alveoli, but also in endothelial cells, which can be directly infected by the virus. Cytokine release syndrome can also drive endothelial damage independently. Consequently, a distinctive feature of SARS-CoV-2 infection is vascular harm, with severe endothelial injury, widespread thrombosis, microangiopathy, and neo-angiogenesis in response to endothelial damage. Therefore, endothelial dysfunction seems to be the pathophysiological substrate for severe COVID-19 complications. Biomarkers of endothelial injury could constitute strong indicators of disease progression and severity. In addition, the endothelium could represent a very attractive target to both prevent and treat these complications. To establish an adequate therapy, the underlying pathophysiology and corresponding clinical stage should be clearly identified. In this review, the clinical features of COVID-19, the central role of the endothelium in COVID-19 and in other pathologies, and the potential of specific therapies aimed at protecting the endothelium in COVID-19 patients are addressed.
Collapse
Affiliation(s)
- Pedro Castro
- Medical Intensive Care Unit, Hospital Clinic, Barcelona, Spain
- School of Medicine, University of Barcelona, Barcelona, Spain
- IDIBAPS, Barcelona, Spain
| | - Marta Palomo
- Josep Carreras Leukaemia Research Institute, Hospital Clinic, University of Barcelona, Barcelona, Spain
- Barcelona Endothelium Team, Barcelona, Spain
| | - Ana Belen Moreno-Castaño
- Barcelona Endothelium Team, Barcelona, Spain
- Hematopathology, Pathology Department, CDB, Hospital Clinic, Villarroel 170, 08036, Barcelona, Spain
| | - Sara Fernández
- Medical Intensive Care Unit, Hospital Clinic, Barcelona, Spain
- Barcelona Endothelium Team, Barcelona, Spain
| | - Sergi Torramadé-Moix
- IDIBAPS, Barcelona, Spain
- Hematopathology, Pathology Department, CDB, Hospital Clinic, Villarroel 170, 08036, Barcelona, Spain
| | | | - Julia Martinez-Sanchez
- Josep Carreras Leukaemia Research Institute, Hospital Clinic, University of Barcelona, Barcelona, Spain
- Barcelona Endothelium Team, Barcelona, Spain
| | - Edward Richardson
- Frank H. Netter M.D. School of Medicine At, Quinnipiac University, North Haven, CT, USA
- Department of Surgery, Yale University School of Medicine, New Haven, CT, USA
| | - Adrián Téllez
- Medical Intensive Care Unit, Hospital Clinic, Barcelona, Spain
| | - Josep M Nicolas
- Medical Intensive Care Unit, Hospital Clinic, Barcelona, Spain
- School of Medicine, University of Barcelona, Barcelona, Spain
- IDIBAPS, Barcelona, Spain
| | - Enric Carreras
- Josep Carreras Leukaemia Research Institute, Hospital Clinic, University of Barcelona, Barcelona, Spain
- Barcelona Endothelium Team, Barcelona, Spain
| | - Paul G Richardson
- Jerome Lipper Multiple Myeloma Center, Division of Hematologic Malignancy, Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Juan José Badimon
- Cardiology Department, Cardiovascular Institute, Mount Sinai Hospital, New York, NY, USA
- AtheroThrombosis Research Unit, Cardiovascular Institute, Icahn School of Medicine At Mount Sinai, New York, NY, USA
| | - Gines Escolar
- School of Medicine, University of Barcelona, Barcelona, Spain
- IDIBAPS, Barcelona, Spain
- Barcelona Endothelium Team, Barcelona, Spain
- Hematopathology, Pathology Department, CDB, Hospital Clinic, Villarroel 170, 08036, Barcelona, Spain
| | - Maribel Diaz-Ricart
- School of Medicine, University of Barcelona, Barcelona, Spain.
- IDIBAPS, Barcelona, Spain.
- Barcelona Endothelium Team, Barcelona, Spain.
- Hematopathology, Pathology Department, CDB, Hospital Clinic, Villarroel 170, 08036, Barcelona, Spain.
| |
Collapse
|
83
|
Fernández S, Moreno-Castaño AB, Palomo M, Martinez-Sanchez J, Torramadé-Moix S, Téllez A, Ventosa H, Seguí F, Escolar G, Carreras E, Nicolás JM, Richardson E, García-Bernal D, Carlo-Stella C, Moraleda JM, Richardson PG, Díaz-Ricart M, Castro P. Distinctive Biomarker Features in the Endotheliopathy of COVID-19 and Septic Syndromes. Shock 2022; 57:95-105. [PMID: 34172614 PMCID: PMC8662948 DOI: 10.1097/shk.0000000000001823] [Citation(s) in RCA: 33] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2021] [Accepted: 06/07/2021] [Indexed: 12/20/2022]
Abstract
BACKGROUND Endotheliopathy is a key element in COVID-19 pathophysiology, contributing to both morbidity and mortality. Biomarkers distinguishing different COVID-19 phenotypes from sepsis syndrome remain poorly understood. OBJECTIVE To characterize circulating biomarkers of endothelial damage in different COVID-19 clinical disease stages compared with sepsis syndrome and normal volunteers. METHODS Patients with COVID-19 pneumonia (n = 49) were classified into moderate, severe, or critical (life-threatening) disease. Plasma samples were collected within 48 to 72 h of hospitalization to analyze endothelial activation markers, including soluble Vascular Cell Adhesion Molecule-1 (sVCAM-1), von Willebrand Factor (VWF), A disintegrin-like and metalloprotease with thrombospondin type 1 motif no. 13 (ADAMTS-13) activity, thrombomodulin (TM), and soluble TNF receptor I (sTNFRI); heparan sulfate (HS) for endothelial glycocalyx degradation; C5b9 deposits on endothelial cells in culture and soluble C5b9 for complement activation; circulating dsDNA for neutrophil extracellular traps (NETs) presence, and α2-antiplasmin and PAI-1 as parameters of fibrinolysis. We compared the level of each biomarker in all three COVID-19 groups and healthy donors as controls (n = 45). Results in critically ill COVID-19 patients were compared with other intensive care unit (ICU) patients with septic shock (SS, n = 14), sepsis (S, n = 7), and noninfectious systemic inflammatory response syndrome (NI-SIRS, n = 7). RESULTS All analyzed biomarkers were increased in COVID-19 patients versus controls (P < 0.001), except for ADAMTS-13 activity that was normal in both groups. The increased expression of sVCAM-1, VWF, sTNFRI, and HS was related to COVID-19 disease severity (P < 0.05). Several differences in these parameters were found between ICU groups: SS patients showed significantly higher levels of VWF, TM, sTNFRI, and NETS compared with critical COVID-19 patients and ADAMTS-13 activity was significantly lover in SS, S, and NI-SIRS versus critical COVID-19 (P < 0.001). Furthermore, α2-antiplasmin activity was higher in critical COVID-19 versus NI-SIRS (P < 0.01) and SS (P < 0.001), whereas PAI-1 levels were significantly lower in COVID-19 patients compared with NI-SIRS, S, and SS patients (P < 0.01). CONCLUSIONS COVID-19 patients present with increased circulating endothelial stress products, complement activation, and fibrinolytic dysregulation, associated with disease severity. COVID-19 endotheliopathy differs from SS, in which endothelial damage is also a critical feature of pathobiology. These biomarkers could help to stratify the severity of COVID-19 disease and may also provide information to guide specific therapeutic strategies to mitigate endotheliopathy progression.
Collapse
Affiliation(s)
- Sara Fernández
- Medical Intensive Care Unit, Hospital Clinic, Barcelona, Spain
| | - Ana B. Moreno-Castaño
- Hematopathology, Pathology Department, CDB, Hospital Clinic, Barcelona, Spain
- Barcelona Endothelium Team, Barcelona, Spain
- IDIBAPS, Barcelona, Spain
| | - Marta Palomo
- Barcelona Endothelium Team, Barcelona, Spain
- Josep Carreras Leukaemia Research Institute, Hospital Clinic, University of Barcelona, Barcelona, Spain
| | - Julia Martinez-Sanchez
- Barcelona Endothelium Team, Barcelona, Spain
- Josep Carreras Leukaemia Research Institute, Hospital Clinic, University of Barcelona, Barcelona, Spain
| | - Sergi Torramadé-Moix
- Hematopathology, Pathology Department, CDB, Hospital Clinic, Barcelona, Spain
- Barcelona Endothelium Team, Barcelona, Spain
- IDIBAPS, Barcelona, Spain
| | - Adrián Téllez
- Medical Intensive Care Unit, Hospital Clinic, Barcelona, Spain
| | - Helena Ventosa
- Medical Intensive Care Unit, Hospital Clinic, Barcelona, Spain
| | - Ferran Seguí
- Medical Intensive Care Unit, Hospital Clinic, Barcelona, Spain
| | - Ginés Escolar
- Hematopathology, Pathology Department, CDB, Hospital Clinic, Barcelona, Spain
- Barcelona Endothelium Team, Barcelona, Spain
- IDIBAPS, Barcelona, Spain
| | - Enric Carreras
- Barcelona Endothelium Team, Barcelona, Spain
- Josep Carreras Leukaemia Research Institute, Hospital Clinic, University of Barcelona, Barcelona, Spain
| | - Josep M. Nicolás
- Medical Intensive Care Unit, Hospital Clinic, Barcelona, Spain
- IDIBAPS, Barcelona, Spain
- School of Medicine, University of Barcelona, Barcelona, Spain
| | - Edward Richardson
- Frank H. Netter M.D. School of Medicine at Quinnipiac University, North Haven, Connecticut
- Department of Surgery, Yale University School of Medicine, New Haven, Connecticut
| | - David García-Bernal
- Hematopoietic Transplant and Cellular Therapy Unit, Instituto Murciano de Investigación Biosanitaria IMIB-Arrixaca, Virgen de la Arrixaca University Hospital, University of Murcia, Murcia, Spain
| | - Carmelo Carlo-Stella
- Department of Oncology and Hematology, Humanitas Clinical and Research Center-IRCCS, Rozzano-Milano, Italy
- Department of Biomedical Sciences, Humanitas University, Rozzano-Milano, Italy
| | - José M. Moraleda
- Hematopoietic Transplant and Cellular Therapy Unit, Instituto Murciano de Investigación Biosanitaria IMIB-Arrixaca, Virgen de la Arrixaca University Hospital, University of Murcia, Murcia, Spain
| | - Paul G. Richardson
- Division of Hematologic Malignancy, Department of Medical Oncology, Dana-Farber Cancer Institute, Jerome Lipper Multiple Myeloma Center, Harvard Medical School, Boston, Massachusetts
| | - Maribel Díaz-Ricart
- Hematopathology, Pathology Department, CDB, Hospital Clinic, Barcelona, Spain
- Barcelona Endothelium Team, Barcelona, Spain
- IDIBAPS, Barcelona, Spain
| | - Pedro Castro
- Medical Intensive Care Unit, Hospital Clinic, Barcelona, Spain
- IDIBAPS, Barcelona, Spain
- School of Medicine, University of Barcelona, Barcelona, Spain
| |
Collapse
|
84
|
Amiri-Dashatan N, Koushki M, Rezaei-Tavirani M. Mass Spectrometry-Based Proteomics Research to Fight COVID-19: An Expert Review on Hopes and Challenges. OMICS : A JOURNAL OF INTEGRATIVE BIOLOGY 2022; 26:19-34. [PMID: 35005991 DOI: 10.1089/omi.2021.0182] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
The COVID-19 pandemic caused by the severe acute respiratory syndrome (SARS)-CoV-2 infection is a systemic disease and a major planetary health burden. While SARS-CoV-2 impacts host biology extensively, our knowledge of these alterations from a systems perspective remains incomplete. Moreover, there is currently only a limited description of this systemic disease. For precision diagnosis and treatment of SARS-CoV-2, multiomics technologies and systems science research offer significant prospects. This expert review offers a critical analysis of the prospects and challenges of the emerging mass spectrometry-based proteomics approaches to the study of COVID-19 as seen through a systems medicine lens. We also discuss the ways in which proteomics is poised to offer hope for diagnostics and therapeutics innovation on SARS-CoV-2 infection as the disease transitions from a pandemic to an endemic disease, and thus further challenging the health systems and services worldwide in the coming decade. Proteomics is an important high-throughput technology platform to achieve a functional overview of the ways in which COVID-19 changes host biology, and hence, can help identify possible points of entry for innovation in medicines and vaccines, among others.
Collapse
Affiliation(s)
- Nasrin Amiri-Dashatan
- Proteomics Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran.,Zanjan Metabolic Diseases Research Center, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Mehdi Koushki
- Department of Clinical Biochemistry, School of Medicine, Zanjan University of Medical Sciences, Zanjan, Iran
| | | |
Collapse
|
85
|
Khandelwal G, Ray A, Sethi S, Harikrishnan HK, Khandelwal C, Sadasivam B. COVID-19 and thrombotic complications-the role of anticoagulants, antiplatelets and thrombolytics. J Family Med Prim Care 2021; 10:3561-3567. [PMID: 34934647 PMCID: PMC8653484 DOI: 10.4103/jfmpc.jfmpc_1297_20] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Revised: 09/13/2020] [Accepted: 07/09/2021] [Indexed: 01/08/2023] Open
Abstract
Coronavirus disease 2019 (COVID-19) is a global pandemic the world is dealing with currently. Clinical evidences suggest that the patients are predisposed to both venous and arterial thrombotic complications. This is because of severe inflammatory responses, injury to endothelium and activation of platelets leading to increased coagulation. Additionally, individuals who are already receiving antithrombotic drug therapy for various cardiovascular diseases and complications might contract the disease in which case, attention should be given to the choice and duration of the therapy besides close monitoring of biochemical blood parameters. Herein, we review the incidences of thrombotic complications and their outcomes in COVID-19 patients as reported till date, while understanding the prophylactic and therapeutic roles of anticoagulants, antiplatelets and thrombolytics in the management of this severe viral respiratory illness.
Collapse
Affiliation(s)
- Gaurav Khandelwal
- Department of Cardiology, All India Institute of Medical Sciences, Bhopal, Madhya Pradesh, India
| | - Avik Ray
- Department of Pharmacology, All India Institute of Medical Sciences, Bhopal, Madhya Pradesh, India
| | - Samdish Sethi
- Department of Medicine, All India Institute of Medical Sciences, Raipur, Chhattisgarh, India
| | - H K Harikrishnan
- Department of Medicine, All India Institute of Medical Sciences, Bhopal, Madhya Pradesh, India
| | - Chaitanya Khandelwal
- Department of Medicine, All India Institute of Medical Sciences, Raipur, Chhattisgarh, India
| | - Balakrishnan Sadasivam
- Department of Pharmacology, All India Institute of Medical Sciences, Bhopal, Madhya Pradesh, India
| |
Collapse
|
86
|
Karacan A, Aksoy YE, Öztürk MH. The radiological findings of COVID-19. Turk J Med Sci 2021; 51:3328-3339. [PMID: 34365783 PMCID: PMC8771018 DOI: 10.3906/sag-2106-203] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2021] [Accepted: 08/07/2021] [Indexed: 02/05/2023] Open
Abstract
Background/aim Available information on the radiological findings of the 2019 novel coronavirus disease (COVID-19) is constantly updated. Ground glass opacities (GGOs) and consolidation with bilateral and peripheral distribution have been reported as the most common CT findings, but less typical features can also be identified. According to the reported studies, SARS-CoV-2 infection is not limited to the respiratory system, and it can also affect other organs. Renal dysfunction, gastrointestinal complications, liver dysfunction, cardiac manifestations, and neurological abnormalities are among the reported extrapulmonary features. This review aims to provide updated information for radiologists and all clinicians to better understand the radiological manifestations of COVID-19. Materials and methods Radiological findings observed in SARS-CoV-2 virus infections were explored in detail in PubMed and Google Scholar databases. Results The typical pulmonary manifestations of COVID-19 pneumonia were determined as GGOs and accompanying consolidations that primarily involve the periphery of the bilateral lower lobes. The most common extrapulmonary findings were increased resistance to flow in the kidneys, thickening of vascular walls, fatty liver, pancreas, and heart inflammation findings. However, these findings were not specific and significantly overlapped those caused by other viral diseases, and therefore alternative diagnoses should be considered in patients with negative diagnostic tests. Conclusion Radiological imaging plays a supportive role in the care of patients with COVID-19. Both clinicians and radiologists need to know associated pulmonary and extrapulmonary findings and imaging features to help diagnose and manage the possible complications of the disease at an early stage. They should also be familiar with CT findings in patients with COVID-19 since the disease can be incidentally detected during imaging performed with other indications.
Collapse
Affiliation(s)
- Alper Karacan
- Department of Radiology, Faculty of Medicine, Sakarya University, Sakarya, Turkey
| | - Yakup Ersel Aksoy
- Department of Radiology, Faculty of Medicine, Sakarya University, Sakarya, Turkey
| | - Mehmet Halil Öztürk
- Department of Radiology, Faculty of Medicine, Sakarya University, Sakarya, Turkey
| |
Collapse
|
87
|
Castillo-Perez M, Jerjes-Sanchez C, Castro-Varela A, Paredes-Vazquez JG, Vazquez-Garza E, Ramos-Cazares RE, Salinas-Casanova JA, Molina-Rodriguez AM, Martinez-Ibarra AA, Fabiani MA, Flores-Sayavedra YZ, Guajardo-Lozano JA, Lopez-de la Garza H, Betancourt-Del Campo H, Martinez-Magallanes D, Panneflek J. Differences between surviving and non-surviving venous thromboembolism COVID-19 patients: a systematic review. Thromb J 2021; 19:101. [PMID: 34911551 PMCID: PMC8672331 DOI: 10.1186/s12959-021-00346-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2021] [Accepted: 11/14/2021] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND To our knowledge, the treatment, outcome, clinical presentation, risk stratification of patients with venous thromboembolism and COVID-19 have not been well characterized. METHODS We searched for systematic reviews, cohorts, case series, case reports, editor letters, and venous thromboembolism COVID-19 patients' abstracts following PRISMA and PROSPERO statements. We analyzed therapeutic approaches and clinical outcomes of venous thromboembolism COVID-19 patients. Inclusion: COVID-19 patients with venous thromboembolism confirmed by an imaging method (venous doppler ultrasound, ventilation-perfusion lung scan, computed tomography pulmonary angiogram, pulmonary angiography). We assessed and reported the original Pulmonary Embolism Severity Index for each pulmonary embolism patient. In addition, we defined major bleedings according to the International Society of Thrombosis and Haemostasis criteria. RESULTS We performed a systematic review from August 9 to August 30, 2020. We collected 1,535 papers from PubMed, Scopus, Web of Science, Wiley, and Opengrey. We extracted data from 89 studies that describe 143 patients. Unfractionated and low-molecular-weight heparin was used as parenteral anticoagulation in 85/143 (59%) cases. The Food and Drug Administration-approved alteplase regimen guided the advanced treatment in 39/143 (27%) patients. The mortality was high (21.6%, CI 95% 15.2-29.3). The incidence of major bleeding complications was 1 (0.9%) in the survival group and 1 (3.2%) in the death group. Pulmonary Embolism Severity Index was class I in 11.6% and II in 22.3% in survivors compared to 0% and 6.5% in non-survivors, respectively. Patients who experienced venous thromboembolism events at home were more likely to live than in-hospital events. CONCLUSIONS We determined a high mortality incidence of pulmonary embolism and a low rate of bleeding. Unfractionated and low-molecular-weight heparin drove parenteral anticoagulation and alteplase the advanced treatment in both groups. The original Pulmonary Embolism Severity Index could be helpful in the risk stratification.
Collapse
Affiliation(s)
- Mauricio Castillo-Perez
- Tecnologico de Monterrey. Escuela de Medicina y Ciencias de la Salud., Nuevo Leon, San Pedro Garza Garcia, Mexico
| | - Carlos Jerjes-Sanchez
- Tecnologico de Monterrey. Escuela de Medicina y Ciencias de la Salud., Nuevo Leon, San Pedro Garza Garcia, Mexico.
- Centro de Investigacion Biomedica del Hospital Zambrano Hellion, TecSalud, Escuela de Medicina y Ciencias de la Salud, Tecnologico de Monterrey, Nuevo Leon, San Pedro Garza Garcia, Mexico.
- Instituto de Cardiologia y Medicina Vascular, TecSalud, Escuela de Medicina y Ciencias de la Salud, Tecnologico de Monterrey, Batallón San Patricio 112, Real de San Agustin, Nuevo Leon, 66278, San Pedro Garza Garcia, Mexico.
- Tecnologico de Monterrey, Escuela de Medicina y Ciencias de la Salud, Av. Ignacio Morones Prieto 3000, N.L., CP, 64718, Monterrey, Mexico.
| | - Alejandra Castro-Varela
- Tecnologico de Monterrey. Escuela de Medicina y Ciencias de la Salud., Nuevo Leon, San Pedro Garza Garcia, Mexico
| | - Jose Gildardo Paredes-Vazquez
- Tecnologico de Monterrey. Escuela de Medicina y Ciencias de la Salud., Nuevo Leon, San Pedro Garza Garcia, Mexico
- Instituto de Cardiologia y Medicina Vascular, TecSalud, Escuela de Medicina y Ciencias de la Salud, Tecnologico de Monterrey, Batallón San Patricio 112, Real de San Agustin, Nuevo Leon, 66278, San Pedro Garza Garcia, Mexico
| | - Eduardo Vazquez-Garza
- Tecnologico de Monterrey. Escuela de Medicina y Ciencias de la Salud., Nuevo Leon, San Pedro Garza Garcia, Mexico
- Centro de Investigacion Biomedica del Hospital Zambrano Hellion, TecSalud, Escuela de Medicina y Ciencias de la Salud, Tecnologico de Monterrey, Nuevo Leon, San Pedro Garza Garcia, Mexico
| | - Ray Erick Ramos-Cazares
- Tecnologico de Monterrey. Escuela de Medicina y Ciencias de la Salud., Nuevo Leon, San Pedro Garza Garcia, Mexico
| | | | | | | | - Mario Alejandro Fabiani
- Instituto de Cardiologia y Medicina Vascular, TecSalud, Escuela de Medicina y Ciencias de la Salud, Tecnologico de Monterrey, Batallón San Patricio 112, Real de San Agustin, Nuevo Leon, 66278, San Pedro Garza Garcia, Mexico
| | - Yoezer Z Flores-Sayavedra
- Tecnologico de Monterrey. Escuela de Medicina y Ciencias de la Salud., Nuevo Leon, San Pedro Garza Garcia, Mexico
| | | | - Hector Lopez-de la Garza
- Tecnologico de Monterrey. Escuela de Medicina y Ciencias de la Salud., Nuevo Leon, San Pedro Garza Garcia, Mexico
| | - Hector Betancourt-Del Campo
- Tecnologico de Monterrey. Escuela de Medicina y Ciencias de la Salud., Nuevo Leon, San Pedro Garza Garcia, Mexico
| | - Daniela Martinez-Magallanes
- Tecnologico de Monterrey. Escuela de Medicina y Ciencias de la Salud., Nuevo Leon, San Pedro Garza Garcia, Mexico
| | - Jathniel Panneflek
- Centro de Investigacion Biomedica del Hospital Zambrano Hellion, TecSalud, Escuela de Medicina y Ciencias de la Salud, Tecnologico de Monterrey, Nuevo Leon, San Pedro Garza Garcia, Mexico
| |
Collapse
|
88
|
Infanger L, Dibiasi C, Schaden E, Ulbing S, Wiegele M, Lacom C, Gratz J. Comparison of the New Viscoelastic Coagulation Analyzer ClotPro® With ROTEM® Delta and Conventional Coagulation Tests in Critically Ill Patients With COVID-19. Front Med (Lausanne) 2021; 8:777145. [PMID: 34869496 PMCID: PMC8635118 DOI: 10.3389/fmed.2021.777145] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Accepted: 10/27/2021] [Indexed: 12/12/2022] Open
Abstract
Background: Viscoelastic coagulation testing has been suggested to help manage coagulopathy in critically ill patients with COVID-19. However, results from different viscoelastic devices are not readily comparable. ClotPro® is a novel thromboelastometry analyzer offering a wider range of commercially available assays. Methods: We compared the results from ClotPro with results from the well-established ROTEM® Delta device and conventional coagulation tests in critically ill patients with COVID-19. Results: Viscoelastic parameters indicated the presence of a potentially hypercoagulable state in the majority of patients. In up to 95 paired measurements, we found strong correlations between several parameters routinely used in clinical practice: (i) EX test vs. EXTEM CT, A5, A10, MCF, (ii) IN test vs. INTEM A5, A10, MCF, and (iii) FIB test vs. FIBTEM A5, A10, MCF (all R > 0.7 and p < 0.001). In contrast, IN test CT vs. INTEM CT showed only a moderate correlation (R = 0.53 and p < 0.001). Clot strength parameters of both devices exhibited strong correlations with platelet counts and fibrinogen levels (all R > 0.7 and p < 0.001). Divergent correlations of intrinsically activated assays with aPTT and anti-factor Xa activity were visible. Regarding absolute differences of test results, considerable delta occurred in CT, CFT, and clot strength parameters (all p < 0.001) between both devices. Conclusions: Several parameters obtained by ClotPro show strong correlations with ROTEM Delta. Due to weak correlations of intrinsically activated clotting times and considerable absolute differences in a number of parameters, our findings underline the need for device-specific algorithms in this patient cohort.
Collapse
Affiliation(s)
- Lukas Infanger
- Department of Anaesthesia, Intensive Care Medicine and Pain Medicine, Medical University of Vienna, Vienna, Austria
| | - Christoph Dibiasi
- Department of Anaesthesia, Intensive Care Medicine and Pain Medicine, Medical University of Vienna, Vienna, Austria
| | - Eva Schaden
- Department of Anaesthesia, Intensive Care Medicine and Pain Medicine, Medical University of Vienna, Vienna, Austria
| | - Stefan Ulbing
- Department of Anaesthesia, Intensive Care Medicine and Pain Medicine, Medical University of Vienna, Vienna, Austria
| | - Marion Wiegele
- Department of Anaesthesia, Intensive Care Medicine and Pain Medicine, Medical University of Vienna, Vienna, Austria
| | - Conrad Lacom
- Department of Anaesthesia, Intensive Care Medicine and Pain Medicine, Medical University of Vienna, Vienna, Austria
| | - Johannes Gratz
- Department of Anaesthesia, Intensive Care Medicine and Pain Medicine, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
89
|
Rashidi F, Barco S, Rezaeifar P, Sadeghipour P, Ghodrati S, Bakhshandeh H, Mousavi-Aghdas SA, Sadeghi A, Sharifi A, Valizadeh H, Mikaeili H, Rafiee F, Navarbaf Z, Farajian G, Bikdeli B, Ansarin K, Mahmoodpoor A. Tissue plasminogen activator for the treatment of adults with critical COVID-19: A pilot randomized clinical trial. Thromb Res 2021; 216:125-128. [PMID: 34969528 PMCID: PMC8648570 DOI: 10.1016/j.thromres.2021.12.003] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2021] [Revised: 09/28/2021] [Accepted: 12/02/2021] [Indexed: 12/15/2022]
Affiliation(s)
- Farid Rashidi
- Tuberculosis and lung disease research center, Tabriz University of Medical Sciences, Tabriz, Iran.
| | - Stefano Barco
- Center for Thrombosis and Hemostasis, Johannes Gutenberg University Mainz, Mainz, Germany; Clinic of Angiology, University Hospital Zurich, Zurich, Switzerland
| | - Parisa Rezaeifar
- Tuberculosis and lung disease research center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Parham Sadeghipour
- Cardiovascular Intervention Research Center, Rajaie Cardiovascular, Medical, and Research Center, Iran University of Medical Sciences, Tehran, Iran; Clinical Trial Center, Rajaie Cardiovascular, Medical, and Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Samad Ghodrati
- Department of Pulmonology, Vali-e-Asr Hospital, School of Medicine, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Hooman Bakhshandeh
- Clinical Trial Center, Rajaie Cardiovascular, Medical, and Research Center, Iran University of Medical Sciences, Tehran, Iran; Rajaie Cardiovascular, Medical, and Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Seyed Ali Mousavi-Aghdas
- Tuberculosis and lung disease research center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Armin Sadeghi
- Tuberculosis and lung disease research center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Akbar Sharifi
- Tuberculosis and lung disease research center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Hamed Valizadeh
- Tuberculosis and lung disease research center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Haleh Mikaeili
- Tuberculosis and lung disease research center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Farnaz Rafiee
- Rajaie Cardiovascular, Medical, and Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Zahar Navarbaf
- Tuberculosis and lung disease research center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Gita Farajian
- Department of Pulmonology, Vali-e-Asr Hospital, School of Medicine, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Behnood Bikdeli
- Cardiovascular Medicine Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA; Yale Center for Outcomes Research and Evaluation (CORE), New Haven, CT, USA; Cardiovascular Research Foundation (CRF), New York, NY, USA
| | - Khalil Ansarin
- Tuberculosis and lung disease research center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Ata Mahmoodpoor
- Department of Anesthesiology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
90
|
Aslan A, Aslan C, Zolbanin NM, Jafari R. Acute respiratory distress syndrome in COVID-19: possible mechanisms and therapeutic management. Pneumonia (Nathan) 2021; 13:14. [PMID: 34872623 PMCID: PMC8647516 DOI: 10.1186/s41479-021-00092-9] [Citation(s) in RCA: 65] [Impact Index Per Article: 21.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Accepted: 11/20/2021] [Indexed: 02/07/2023] Open
Abstract
COVID-19 pandemic is a serious concern in the new era. Acute respiratory distress syndrome (ARDS), and lung failure are the main lung diseases in COVID-19 patients. Even though COVID-19 vaccinations are available now, there is still an urgent need to find potential treatments to ease the effects of COVID-19 on already sick patients. Multiple experimental drugs have been approved by the FDA with unknown efficacy and possible adverse effects. Probably the increasing number of studies worldwide examining the potential COVID-19 related therapies will help to identification of effective ARDS treatment. In this review article, we first provide a summary on immunopathology of ARDS next we will give an overview of management of patients with COVID-19 requiring intensive care unit (ICU), while focusing on the current treatment strategies being evaluated in the clinical trials in COVID-19-induced ARDS patients.
Collapse
Affiliation(s)
- Anolin Aslan
- Department of Critical Care Nursing, School of Nursing and Midwifery, Tehran University of Medical Science, Tehran, Iran
| | - Cynthia Aslan
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.,Department of Immunology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Naime Majidi Zolbanin
- Experimental and Applied Pharmaceutical Research Center, Urmia University of Medical Sciences, Urmia, Iran.,Department of Pharmacology and Toxicology, School of Pharmacy, Urmia University of Medical Sciences, Urmia, Iran
| | - Reza Jafari
- Nephrology and Kidney Transplant Research Center, Clinical Research Institute, Urmia University of Medical Sciences, Shafa St., Ershad Blvd., P.O. Box: 1138, Urmia, 57147, Iran. .,Hematology, Immune Cell Therapy, and Stem Cell Transplantation Research Center, Clinical Research Institute, Urmia University of Medical Sciences, Urmia, Iran.
| |
Collapse
|
91
|
Immunotherapy of multisystem inflammatory syndrome in children (MIS-C) following COVID-19 through mesenchymal stem cells. Int Immunopharmacol 2021. [DOI: oi.org/10.1016/j.intimp.2021.108217] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
|
92
|
Kalita B, Saviola AJ, Samuel SP, Mukherjee AK. State-of-the-art review - A review on snake venom-derived antithrombotics: Potential therapeutics for COVID-19-associated thrombosis? Int J Biol Macromol 2021; 192:1040-1057. [PMID: 34656540 PMCID: PMC8514616 DOI: 10.1016/j.ijbiomac.2021.10.015] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2021] [Revised: 10/03/2021] [Accepted: 10/04/2021] [Indexed: 12/30/2022]
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), the causative agent responsible for the Coronavirus Disease-2019 (COVID-19) pandemic, has infected over 185 million individuals across 200 countries since December 2019 resulting in 4.0 million deaths. While COVID-19 is primarily associated with respiratory illnesses, an increasing number of clinical reports indicate that severely ill patients often develop thrombotic complications that are associated with increased mortality. As a consequence, treatment strategies that target COVID-associated thrombosis are of utmost clinical importance. An array of pharmacologically active compounds from natural products exhibit effects on blood coagulation pathways, and have generated interest for their potential therapeutic applications towards thrombotic diseases. In particular, a number of snake venom compounds exhibit high specificity on different blood coagulation factors and represent excellent tools that could be utilized to treat thrombosis. The aim of this review is to provide a brief summary of the current understanding of COVID-19 associated thrombosis, and highlight several snake venom compounds that could be utilized as antithrombotic agents to target this disease.
Collapse
Affiliation(s)
- Bhargab Kalita
- Department of Molecular Biology and Biotechnology, Tezpur University, Tezpur 784028, Assam, India; National Centre for Cell Science, Pune 411007, Maharashtra, India
| | - Anthony J Saviola
- Department of Biochemistry and Molecular Genetics, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Stephen P Samuel
- Queen Elizabeth Hospital King's Lynn NHS Foundation Trust, King's Lynn, Norfolk PE30 4ET, UK
| | - Ashis K Mukherjee
- Department of Molecular Biology and Biotechnology, Tezpur University, Tezpur 784028, Assam, India; Institute of Advanced Study in Science and Technology, Guwahati 781035, Assam, India.
| |
Collapse
|
93
|
Merino JJ, Macho-González A, Benedi J, González MP. Neurological manifestations of COVID-19 in patients: from path physiology to therapy. Neurol Sci 2021; 42:4867-4879. [PMID: 34417704 PMCID: PMC8379040 DOI: 10.1007/s10072-021-05505-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2021] [Accepted: 07/19/2021] [Indexed: 01/10/2023]
Abstract
Coronavirus is a family of ARN positive single-stranded belonging to the family of Coronaviridae. There are several families of coronavirus that transmit more or less serious diseases. However, the so-called coronavirus-19 (SARS-CoV2) is the one that is currently causing most of the problems; in fact, biological dysfunctions that this virus causes provoke damage in various organs, from the lung to the heart, the kidney, the circulatory system, and even the brain. The neurological manifestations caused by viral infection, as well as the hypercoagulopathy and systemic inflammation, have been reported in several studies. In this review, we update the neurological mechanisms by which coronavirus-19 causes neurological manifestation in patients such as encephalomyelitis, Guillain-Barré syndrome, lacunars infarcts, neuropsychiatry disorders such as anxiety and depression, and vascular alterations. This review explains (a) the possible pathways by which coronavirus-19 can induce the different neurological manifestations, (b) the strategies used by the virus to cross the barrier system, (c) how the immune system responds to the infection, and (d) the treatment than can be administered to the COVID-19 patients.
Collapse
Affiliation(s)
- J J Merino
- Departamento de Farmacología, Farmacognosia y Botánica, Facultad de Farmacia, Universidad Complutense de Madrid (UCM), Madrid, Spain
| | - A Macho-González
- Departamento de Farmacología, Farmacognosia y Botánica, Facultad de Farmacia, Universidad Complutense de Madrid (UCM), Madrid, Spain
| | - J Benedi
- Departamento de Farmacología, Farmacognosia y Botánica, Facultad de Farmacia, Universidad Complutense de Madrid (UCM), Madrid, Spain
| | - M P González
- Departamento de Farmacología, Farmacognosia y Botánica, Facultad de Farmacia, Universidad Complutense de Madrid (UCM), Madrid, Spain.
| |
Collapse
|
94
|
Suksatan W, Chupradit S, Yumashev AV, Ravali S, Shalaby MN, Mustafa YF, Kurochkin A, Siahmansouri H. Immunotherapy of multisystem inflammatory syndrome in children (MIS-C) following COVID-19 through mesenchymal stem cells. Int Immunopharmacol 2021; 101:108217. [PMID: 34627083 PMCID: PMC8487784 DOI: 10.1016/j.intimp.2021.108217] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2021] [Revised: 09/28/2021] [Accepted: 09/30/2021] [Indexed: 02/06/2023]
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is a new type of coronavirus causing coronavirus 2019 (COVID-19) that was first observed in Wuhan, China, in Dec. 2019. An inflammatory immune response targeting children appeared during the pandemic, which was associated with COVID-19 named multisystem inflammatory syndrome in children (MIS-C). Characteristics of MIS-C include the classic inflammation findings, multi-organ dysfunction, and fever as the cardinal feature. Up to now, no specific therapy has been identified for MIS-C. Currently, considerable progress has been obtained in the MIS-C treatment by cell therapy, specially Mesenchymal stem cells (MSCs). Unique properties have been reported for MSCs, such as various resources for purification of cell, high proliferation, self-renewal, non-invasive procedure, tissue regenerator, multidirectional differentiation, and immunosuppression. As indicated by a recent clinical research, MSCs have the ability of reducing disease inflammation and severity in children with MIS-C. In the present review study, the benefits and characteristics of MSCs and exosomes are discussed for treating patients with MIS-C.
Collapse
Affiliation(s)
- Wanich Suksatan
- Faculty of Nursing, HRH Princess Chulabhorn College of Medical Science, Chulabhorn Royal Academy, Bangkok, Thailand
| | - Supat Chupradit
- Department of Occupational Therapy, Faculty of Associated Medical Sciences, Chiang Mai University, Chiang Mai 50200, Thailand
| | | | - Sahithya Ravali
- Department of Pharmacy Practice, SRM College of Pharmacy, SRM Institute of Science and Technology, Chennai, India
| | | | - Yasser Fakri Mustafa
- Department of Pharmaceutical Chemistry, College of Pharmacy, University of Mosul, Mosul 41001, Iraq
| | | | - Homayoon Siahmansouri
- Department of Immunology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
95
|
Bagheri SM, Tabrizi Z. Deep dorsal penile vein thrombosis in a patient with COVID-19 infection: A rare complication and the first reported case. Clin Case Rep 2021; 9:e05117. [PMID: 34925832 PMCID: PMC8647807 DOI: 10.1002/ccr3.5117] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2021] [Revised: 11/05/2021] [Accepted: 11/06/2021] [Indexed: 01/19/2023] Open
Abstract
COVID-19 infection may have extrapulmunary manifestations such as blood hypercoagulability that may cause thrombosis in both arterial and venous system. Deep dorsal penile vein thrombosis is very rare, and the most common reason is coagulation disorders. The common observed symptom is penile pain especially during erection and it is diagnosed by ultrasound evaluation of the vein. It is necessary to distinguish deep dorsal penile vein thrombosis from superficial dorsal penile vein thrombosis as it needs anti-coagulant treatment. In present study, we describe a unique case of the deep dorsal penile vein thrombosis following COVID-19 infection.
Collapse
Affiliation(s)
- Seyed Morteza Bagheri
- Department of RadiologyHasheminejad Kidney Center (HKC)Iran University of Medical SciencesTehranIran
| | - Zhale Tabrizi
- Radiology DepartmentIran University of Medical ScienceTehranIran
| |
Collapse
|
96
|
Sarkar M, Madabhavi IV, Quy PN, Govindagoudar MB. COVID-19 and coagulopathy. THE CLINICAL RESPIRATORY JOURNAL 2021; 15:1259-1274. [PMID: 34399021 PMCID: PMC8444678 DOI: 10.1111/crj.13438] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/29/2020] [Revised: 06/22/2021] [Accepted: 08/10/2021] [Indexed: 02/07/2023]
Abstract
The SARS-CoV-2 is a new coronavirus responsible for the COVID-19 disease and has caused the pandemic worldwide. A large number of cases have overwhelmed the healthcare system worldwide. The COVID-19 infection has been associated with a heightened risk of thromboembolic complications. Various mechanisms are leading to the high thrombotic risk in COVID-19 patients such as inflammation, endotheliitis, hyperviscosity, and hypercoagulability. We searched PubMed, EMBASE, and CINAHL from January 2020 to December 2020. We used the following search terms: COVID-19, coagulopathy, and thrombosis. We reviewed the epidemiology, clinical features, mechanisms, and treatment of COVID-19-associated coagulopathy.
Collapse
Affiliation(s)
- Malay Sarkar
- Department of pulmonary medicineIndira Gandhi Medical CollegeShimlaHimachal PradeshIndia
| | - Irappa V. Madabhavi
- Department of Medical and Pediatric OncologyKerudi Cancer HospitalBagalkotKarnatakaIndia
- Department of Medical OncologyJ N Medical CollegeBelagaviKarnatakaIndia
- Department of Medical OncologyNanjappa HospitalShimogaKarnatakaIndia
| | - Pham Nguyen Quy
- Department of Medical OncologyKyoto Miniren Central HospitalKyotoJapan
| | - Manjunath B. Govindagoudar
- Department of Pulmonary and Critical CarePt B D Sharma Postgraduate Institute of Medical SciencesRohtakHaryanaIndia
| |
Collapse
|
97
|
Suksatan W, Chupradit S, Yumashev AV, Ravali S, Shalaby MN, Mustafa YF, Kurochkin A, Siahmansouri H. Immunotherapy of multisystem inflammatory syndrome in children (MIS-C) following COVID-19 through mesenchymal stem cells. Int Immunopharmacol 2021. [DOI: https://doi.org/10.1016/j.intimp.2021.108217] [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]
|
98
|
Wadaa-Allah A, Emhamed MS, Sadeq MA, Ben Hadj Dahman N, Ullah I, Farrag NS, Negida A. Efficacy of the current investigational drugs for the treatment of COVID-19: a scoping review. Ann Med 2021; 53:318-334. [PMID: 33706639 PMCID: PMC7971293 DOI: 10.1080/07853890.2021.1875500] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Accepted: 01/08/2021] [Indexed: 01/08/2023] Open
Abstract
To date, there is no final FDA-approved treatment for COVID-19. There are thousands of studies published on the available treatments for COVID-19 virus in the past year. Therefore, it is crucial to synthesize and summarize the evidence from published studies on the safety and efficacy of experimental treatments of COVID-19. We conducted a systematic literature search of MEDLINE, PubMed, Cochrane Library, GHL, OpenGrey, ICTRP, and ClinicalTrials.gov databases through April 2020. We obtained 2699 studies from the initial literature search. Of them, we included 28 eligible studies that met our eligibility criteria. The sample size of the included studies is 2079 individuals. We extracted and pooled the available data and conducted a quality assessment for the eligible studies. From the 28 studies, only 13 studies provide strong evidence. Our results showed that Favipiravir and Hydroxycholoroquine shorten viral clearance and clinical recovery time and promote pneumonia absorption. On the other hand, Lopinavir-ritonavir either alone or combined with arbidol or interferons has no significant difference superior to the standard care. Corticosteroids, Convalescent plasma transfusion, and anticoagulant therapies provide a better prognosis. Remedsivir, Tocilizumab, Immunoglobulin, Mesenchymal stem cell transplantation showed effective treatment results, but further confirmatory studies are needed. In conclusion, Favipiravir and Remedsivir might be promising drugs in the treatment of COVID-19 patients. .
Collapse
Affiliation(s)
- Ahmed Wadaa-Allah
- Biochemistry Department, Faculty of Science, Ain Shams University, Cairo, Egypt
| | | | | | | | - Irfan Ullah
- Kabir Medical College, Gandhara University, Peshawar, Pakistan
| | - Nesrine S. Farrag
- Community Medicine Department, Faculty of Medicine, Mansoura University, Mansoura, Egypt
| | - Ahmed Negida
- Zagazig University Hospitals, Zagazig University, El-Sharkia, Egypt
| |
Collapse
|
99
|
Lim MS, Mcrae S. COVID-19 and immunothrombosis: Pathophysiology and therapeutic implications. Crit Rev Oncol Hematol 2021; 168:103529. [PMID: 34800652 PMCID: PMC8596655 DOI: 10.1016/j.critrevonc.2021.103529] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2021] [Revised: 09/11/2021] [Accepted: 11/15/2021] [Indexed: 12/11/2022] Open
Abstract
The coagulopathy of COVID-19 is characterised by significantly elevated D Dimer and fibrinogen, mild thrombocytopenia and a mildly prolonged PT/APTT. A high incidence of thrombotic complications occurs despite standard thromboprophylaxis. The evidence to date supports immunothrombosis as the underlying mechanism for this coagulopathy which is triggered by a hyperinflammatory response and endotheliopathy. A hypercoagulable state results from endothelial damage/activation, complement activation, platelet hyperactivity, release of Extracellular Neutrophil Traps, activation of the coagulation system and a "hypofibrinolytic" state. Significant cross-talk occurs between the innate/adaptive immune system, endothelium and the coagulation system. D dimer has been shown to be the most reliable predictor of disease severity, thrombosis, and overall survival. In this context, targeting pathways upstream of coagulation using novel or repurposed drugs alone or in combination with other anti-thrombotic agents may be a rational approach to prevent the mortality/morbidity due to COVID-19 associated coagulopathy.
Collapse
Affiliation(s)
- Ming Sheng Lim
- Department of Hematology, Launceston General Hospital, WP Holman Clinic, Level 1. PO Box 1963, Launceston, Tasmania, Australia.
| | - Simon Mcrae
- Department of Hematology, Launceston General Hospital, WP Holman Clinic, Level 1. PO Box 1963, Launceston, Tasmania, Australia.
| |
Collapse
|
100
|
Shah S, Switzer S, Shippee ND, Wogensen P, Kosednar K, Jones E, Pestka DL, Badlani S, Butler M, Wagner B, White K, Rhein J, Benson B, Reding M, Usher M, Melton GB, Tignanelli CJ. Implementation of an Anticoagulation Practice Guideline for COVID-19 via a Clinical Decision Support System in a Large Academic Health System and Its Evaluation: Observational Study. JMIR Med Inform 2021; 9:e30743. [PMID: 34550900 PMCID: PMC8604256 DOI: 10.2196/30743] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2021] [Revised: 08/17/2021] [Accepted: 09/17/2021] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND Studies evaluating strategies for the rapid development, implementation, and evaluation of clinical decision support (CDS) systems supporting guidelines for diseases with a poor knowledge base, such as COVID-19, are limited. OBJECTIVE We developed an anticoagulation clinical practice guideline (CPG) for COVID-19, which was delivered and scaled via CDS across a 12-hospital Midwest health care system. This study represents a preplanned 6-month postimplementation evaluation guided by the RE-AIM (Reach, Effectiveness, Adoption, Implementation, and Maintenance) framework. METHODS The implementation outcomes evaluated were reach, adoption, implementation, and maintenance. To evaluate effectiveness, the association of CPG adherence on hospital admission with clinical outcomes was assessed via multivariable logistic regression and nearest neighbor propensity score matching. A time-to-event analysis was conducted. Sensitivity analyses were also conducted to evaluate the competing risk of death prior to intensive care unit (ICU) admission. The models were risk adjusted to account for age, gender, race/ethnicity, non-English speaking status, area deprivation index, month of admission, remdesivir treatment, tocilizumab treatment, steroid treatment, BMI, Elixhauser comorbidity index, oxygen saturation/fraction of inspired oxygen ratio, systolic blood pressure, respiratory rate, treating hospital, and source of admission. A preplanned subgroup analysis was also conducted in patients who had laboratory values (D-dimer, C-reactive protein, creatinine, and absolute neutrophil to absolute lymphocyte ratio) present. The primary effectiveness endpoint was the need for ICU admission within 48 hours of hospital admission. RESULTS A total of 2503 patients were included in this study. CDS reach approached 95% during implementation. Adherence achieved a peak of 72% during implementation. Variation was noted in adoption across sites and nursing units. Adoption was the highest at hospitals that were specifically transformed to only provide care to patients with COVID-19 (COVID-19 cohorted hospitals; 74%-82%) and the lowest in academic settings (47%-55%). CPG delivery via the CDS system was associated with improved adherence (odds ratio [OR] 1.43, 95% CI 1.2-1.7; P<.001). Adherence with the anticoagulation CPG was associated with a significant reduction in the need for ICU admission within 48 hours (OR 0.39, 95% CI 0.30-0.51; P<.001) on multivariable logistic regression analysis. Similar findings were noted following 1:1 propensity score matching for patients who received adherent versus nonadherent care (21.5% vs 34.3% incidence of ICU admission within 48 hours; log-rank test P<.001). CONCLUSIONS Our institutional experience demonstrated that adherence with the institutional CPG delivered via the CDS system resulted in improved clinical outcomes for patients with COVID-19. CDS systems are an effective means to rapidly scale a CPG across a heterogeneous health care system. Further research is needed to investigate factors associated with adherence at low and high adopting sites and nursing units.
Collapse
Affiliation(s)
- Surbhi Shah
- University of Minnesota, Minneapolis, MN, United States
| | - Sean Switzer
- University of Minnesota, Minneapolis, MN, United States
| | | | - Pamela Wogensen
- Information Technology, Fairview Health Services, Minneapolis, MN, United States
| | - Kathryn Kosednar
- Information Technology, Fairview Health Services, Minneapolis, MN, United States
| | - Emma Jones
- Department of Surgery, University of Minnesota, Minneapolis, MN, United States
| | - Deborah L Pestka
- College of Pharmacy, University of Minnesota, Minneapolis, MN, United States
| | - Sameer Badlani
- Information Technology, Fairview Health Services, Minneapolis, MN, United States
| | - Mary Butler
- School of Public Health, University of Minnesota, Minneapolis, MN, United States
| | - Brittin Wagner
- School of Public Health, University of Minnesota, Minneapolis, MN, United States
| | - Katie White
- School of Public Health, University of Minnesota, Minneapolis, MN, United States
| | - Joshua Rhein
- Department of Medicine, University of Minnesota, Minneapolis, MN, United States
| | - Bradley Benson
- Department of Medicine, University of Minnesota, Minneapolis, MN, United States
| | - Mark Reding
- Department of Medicine, University of Minnesota, Minneapolis, MN, United States
| | - Michael Usher
- Department of Medicine, University of Minnesota, Minneapolis, MN, United States
| | - Genevieve B Melton
- Department of Surgery, University of Minnesota, Minneapolis, MN, United States
| | | |
Collapse
|