1
|
Marrero Eligio De La Puente CE, Flota Ruiz D, Besalduch LS, Capó XF, Sala DG, Ibars CP, Mindiolaza IB, Chiscano Camon LS, Sanmartin AR, Ruiz-Rodríguez JC, Ferrer R, Montoya SB. Systematic Ultrasound Screening for Lower Extremity Deep Vein Thrombosis in ICU Patients with Severe COVID-19: A Randomized Clinical Trial. J Intensive Care Med 2025; 40:739-748. [PMID: 39838947 DOI: 10.1177/08850666251313774] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2025]
Abstract
BackgroundVenous thromboembolism (VTE), whether pulmonary embolism (PE) or deep vein thrombosis (DVT), is common in patients with COVID-19. Recommendations on systematic screening in the intensive care unit (ICU) are lacking.Research questionIs there any clinical benefit of systematic screening for DVT in critically ill patients with severe COVID-19?Study design and methodsSingle-center randomized clinical trial (RCT) of COVID-19 cases admitted to the ICU. Patients were randomized into two groups: a study group that underwent ultrasound (US) screening for DVT Mondays and Thursdays, and a control group that was treated according to the unit protocol. The primary outcome was the presence of DVT. Secondary outcomes were ICU total stay, death within 21-day follow-up and bleeding complications (minor or major). A composite outcome of poor prognosis variables was analyzed. We tested a superiority hypothesis with a confidence level of 95% and an equivalence limit of 20%.Results163 patients (84 screening group, 79 control group) were enrolled between April and July 2021. There were 90 men (55.2%) with a mean ± SD age of 49.8 ± 13.58 years. In screening group 16.7% developed DVT versus 3.8% in control group (p = .007), and 3.6% versus 5.1% developed PE, respectively (p = 0.7). Poor outcome variables were male sex, age, COVID-19 vaccination status, Fibrinogen, Urea, Creatinine and Interleukin 6 (IL6) levels; Acute Physiology and Chronic Health Evaluation II (APACHE II) and Sequential Organ Failure Assessment (SOFA) scales. The superiority comparison, with a power of 95%, showed no statistically significant differences for a composite endpoint (p = .123). After adjusting by group, the OR for poor outcome is 1.966 (0.761-5.081) p = 0.163InterpretationAmong these patients, a strategy of systematic US screening for DVT was not associated with any significant improvements to clinical outcomes compared with usual care.Clinical Trial RegistrationClinicaltrials.org registration number: NCT05028244.
Collapse
Affiliation(s)
- Carlos Ernesto Marrero Eligio De La Puente
- Servicio de Angiología, cirugía vascular y endovascular. Hospital Universitario Vall d'Hebron, Barcelona, Spain
- Departamento de Cirugía y Ciencias Morfológicas, Universidad Autónoma de Barcelona, Barcelona, Spain
| | - David Flota Ruiz
- Servicio de Angiología, cirugía vascular y endovascular. Hospital Universitario Vall d'Hebron, Barcelona, Spain
| | - Lluis Sánchez Besalduch
- Servicio de Angiología, cirugía vascular y endovascular. Hospital Universitario Vall d'Hebron, Barcelona, Spain
| | - Xavier Faner Capó
- Servicio de Angiología, cirugía vascular y endovascular. Hospital Universitario Vall d'Hebron, Barcelona, Spain
| | - Daniel Gil Sala
- Servicio de Angiología, cirugía vascular y endovascular. Hospital Universitario Vall d'Hebron, Barcelona, Spain
| | - Clara Palmada Ibars
- Servicio de Medicina Intensiva. Hospital Universitario Vall d'Hebron, Barcelona, Spain
| | - Ivan Bajaña Mindiolaza
- Servicio de Medicina Intensiva. Hospital Universitario Vall d'Hebron, Barcelona, Spain
- Grupo de investigación Shock, Disfunción Orgánica y Resucitación. Vall d'Hebron Institut de Recerca, Barcelona, Spain
| | - Luis Silvestre Chiscano Camon
- Servicio de Medicina Intensiva. Hospital Universitario Vall d'Hebron, Barcelona, Spain
- Grupo de investigación Shock, Disfunción Orgánica y Resucitación. Vall d'Hebron Institut de Recerca, Barcelona, Spain
| | - Adolfo Ruiz Sanmartin
- Servicio de Medicina Intensiva. Hospital Universitario Vall d'Hebron, Barcelona, Spain
- Grupo de investigación Shock, Disfunción Orgánica y Resucitación. Vall d'Hebron Institut de Recerca, Barcelona, Spain
| | - Juan Carlos Ruiz-Rodríguez
- Servicio de Medicina Intensiva. Hospital Universitario Vall d'Hebron, Barcelona, Spain
- Grupo de investigación Shock, Disfunción Orgánica y Resucitación. Vall d'Hebron Institut de Recerca, Barcelona, Spain
- Departamento de Medicina, Universidad Autónoma de Barcelona, Barcelona, Spain
| | - Ricard Ferrer
- Servicio de Medicina Intensiva. Hospital Universitario Vall d'Hebron, Barcelona, Spain
- Grupo de investigación Shock, Disfunción Orgánica y Resucitación. Vall d'Hebron Institut de Recerca, Barcelona, Spain
- Departamento de Medicina, Universidad Autónoma de Barcelona, Barcelona, Spain
| | - Sergi Bellmunt Montoya
- Servicio de Angiología, cirugía vascular y endovascular. Hospital Universitario Vall d'Hebron, Barcelona, Spain
- Departamento de Cirugía y Ciencias Morfológicas, Universidad Autónoma de Barcelona, Barcelona, Spain
| |
Collapse
|
2
|
Xue L, Qi Y, Zou Y. Short-term safety and efficacy of aspirin in patients with COVID-19: a systematic review and meta-analysis of randomized controlled trials. PeerJ 2025; 13:e19466. [PMID: 40416616 PMCID: PMC12103164 DOI: 10.7717/peerj.19466] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2025] [Accepted: 04/23/2025] [Indexed: 05/27/2025] Open
Abstract
Objective Coagulation activation and inflammatory derangements are key characteristics of coronavirus disease 2019 (COVID-19). Aspirin therapy in patients with COVID-19 remains uncertain due to conflicting evidence regarding its ability to balance anti-inflammatory and antithrombotic benefits against potential bleeding risks in the context of COVID-19-associated coagulopathy. This study aimed to compare the clinical safety and efficacy of aspirin in patients with COVID-19 in randomized controlled trials (RCTs). Methods In the present systematic review and meta-analysis, the Medline, Embase, and Cochrane Library databases were searched for RCTs from database inception to January 13, 2023. Data were independently extracted and screened by two authors using structured data collection forms based on published reports. Results were calculated using odds ratios (ORs) and 95% confidence intervals (CIs) with the Mantel-Haenszel method. Quality was assessed using the Cochrane Risk of Bias tool. The main outcomes were short-term all-cause mortality, bleeding events and any thrombosis events. This meta-analysis was registered on PROSPERO. Results A total of 922 studies were identified. Finally, six RCTs with low risk of bias were pooled in the analysis. The results showed that aspirin use was not associated with a reduction in all-cause mortality (OR = 0.95, 95% CI [0.88-1.03], I2 = 0%) or the risk of any thrombosis (RR 0.88, 95% CI [0.77-1.01], I2 = 0%), but aspirin use was associated with a higher risk of bleeding (OR 1.72, 95% CI [1.32-2.24], I2 = 0%). No obvious risk of bias was found among the included RCTs for the primary outcome. Conclusion Routine low-dose aspirin use does not reduce the risk of short-term mortality and risk of any thrombosis but increases the risk of bleeding. The data does not support the use of low-dose aspirin in patients with COVID-19.
Collapse
Affiliation(s)
- Liwen Xue
- Department of Pathology and Pathophysiology, School of Basic Medicine, Kunming Medical University, Kunming, China
| | - Yuhan Qi
- West China School of Medicine, West China Hospital, Sichuan University, Chengdu, China
- Division of Vascular Surgery Department of General Surgery, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Yingying Zou
- Department of Pathology and Pathophysiology, School of Basic Medicine, Kunming Medical University, Kunming, China
- Frontiers Medical Center, Tianfu Jincheng Laboratory, Chengdu, Sichuan Province, China
| |
Collapse
|
3
|
Victorin D, Bergquist H, Hafsten L, Nihlén Å, Lindell E. Low Molecular Weight Heparin Dosing in Relation to Postoperative Bleeding After Tracheotomy in Patients Infected With SARS-CoV-2-A Descriptive Study. Laryngoscope Investig Otolaryngol 2025; 10:e70122. [PMID: 40177253 PMCID: PMC11963079 DOI: 10.1002/lio2.70122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2024] [Revised: 01/02/2025] [Accepted: 02/02/2025] [Indexed: 04/05/2025] Open
Abstract
Objective The aim of this study was to analyze whether patients with SARS-CoV-2 who received surgical tracheotomy had a lower incidence of postoperative bleeding if their LMWH was postponed or canceled on the day of surgery. Methods Patients with SARS-CoV-2 admitted to the intensive care units who underwent surgical tracheotomy were assessed retrospectively through their medical records. Data on comorbidity, LMWH dose, and timing were collected. Bleedings < 72 h post surgery were noted as stomal or airway bleedings. Results All 101 patients included were on LMWH. Twenty-two patients had no change of dose of LMWH, 24 patients had their dose of LMWH postponed to post surgery, and 50 patients had their dose reduced to only the evening dose on the day of surgery. Twenty-six patients had a stomal bleeding, one patient had an airway bleeding, and four patients had both stomal and airway bleedings. No significant difference in the incidence of bleeding was identified between various groups of different LMWH doses or timing, reduced dose versus no change of dose, OR 1.29 (95% CI 0.42-3.92). Postponed dose versus no change of dose of LMWH, OR 1.03 (95% CI 0.28-3.75). Increasing age was correlated to a higher risk of bleeding post-surgery by an OR of 1.64 (95% CI 1.06-2.54, p = 0.026 for every 10 years added). No fatal bleeding related to surgical tracheotomy was observed. Conclusion Decreased doses of LMWH on the day of surgery were not associated with a risk reduction for post-surgical bleeding in patients with SARS-CoV-2 who received tracheotomy. Increasing age was a risk factor for post-surgical bleeding. Level of Evidence Retrospective, level 3.
Collapse
Affiliation(s)
- David Victorin
- Department of OtorhinolaryngologyRegion Västra Götaland, Södra Älvsborg HospitalBoråsSweden
- Department of Research, Education and InnovationRegion Västra Götaland, Södra Älvsborg HospitalBoråsSweden
| | - Henrik Bergquist
- Department of Otorhinolaryngology, Head and Neck Surgery, Institute of Clinical SciencesSahlgrenska Academy, University of GothenburgGothenburgSweden
- Department of OtorhinolaryngologyRegion Västra Götaland, Sahlgrenska University HospitalGothenburgSweden
| | - Louise Hafsten
- Department of Otorhinolaryngology, Head and Neck Surgery, Institute of Clinical SciencesSahlgrenska Academy, University of GothenburgGothenburgSweden
| | - Åsa Nihlén
- Department of OtorhinolaryngologyRegion Västra Götaland, Södra Älvsborg HospitalBoråsSweden
| | - Ellen Lindell
- Department of OtorhinolaryngologyRegion Västra Götaland, Södra Älvsborg HospitalBoråsSweden
- Department of Otorhinolaryngology, Head and Neck Surgery, Institute of Clinical SciencesSahlgrenska Academy, University of GothenburgGothenburgSweden
| |
Collapse
|
4
|
Alonso Fernández MÁ, Bledig C, Manso Álvarez M, Gómez Guardiola R, Blancas García M, Bartolomé I, Quintana Díaz M, Marcos Neira P, Silva Obregón JA, Serrano Lázaro A, Campillo Morales S, López Matamala B, Martín Parra C, Algaba Calderón Á, Blancas Gómez-Casero R, Martínez González Ó. SARS-CoV-2 vaccination reduces the risk of thrombotic complications in severe COVID-19. Med Intensiva 2025:502167. [PMID: 40121176 DOI: 10.1016/j.medine.2025.502167] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2024] [Accepted: 01/31/2025] [Indexed: 03/25/2025]
Abstract
OBJECTIVES The aim of this study was to evaluate the association between SARS-CoV-2 vaccination and the occurrence of thrombotic complications in patients admitted to intensive care for severe COVID-19 pneumonia. DESIGN Observational, descriptive, prospective, multicentre study. SETTING Intensive care units of five university hospitals. PATIENTS A total of 255 patients admitted to the intensive care unit (ICU) with SARS-CoV-2 pneumonia, confirmed by RT-PCR in throat swab or tracheal aspirate, starting the date the first vaccinated patient against SARS-CoV-2 was admitted in one of the participating ICUs, were included in the analysis. MAIN VARIABLES OF INTEREST Vaccination status against SARS-CoV-2 and thrombotic events. RESULTS 18.8% of patients had received some form of vaccination. Thrombotic events occurred in 21.2% of patients. Lack of vaccination was associated with thrombotic events (OR 5.024; 95% CI: 1.104-23.123; p = 0.0037) and death (OR 5.161; 95% CI: 1.075-24.787; p = 0.04). ICU mortality was not associated with the occurrence of thrombotic complications. CONCLUSIONS In this series of patients, vaccination against SARS-CoV-2 reduced the risk of thrombotic events and mortality in patients with severe COVID-19 admitted to the ICU. Thrombotic complications did not alter ICU mortality.
Collapse
Affiliation(s)
| | - Carola Bledig
- Ospedale Michele e Pietro Ferrero, Servicio de Anestesia e Rianimazione, Italy
| | - Madian Manso Álvarez
- Hospital Universitario del Tajo, Critical Care Department, Universidad Alfonso X El Sabio, Spain
| | | | | | | | - Manuel Quintana Díaz
- Hospital Universitario La Paz, Critical Care Department, Universidad Autónoma de Madrid, Spain
| | | | | | | | | | - Blanca López Matamala
- Hospital Universitario del Tajo, Critical Care Department, Universidad Alfonso X El Sabio, Spain
| | - Carmen Martín Parra
- Hospital Universitario del Tajo, Critical Care Department, Universidad Alfonso X El Sabio, Spain
| | - Ángela Algaba Calderón
- Hospital Universitario del Tajo, Critical Care Department, Universidad Alfonso X El Sabio, Spain
| | | | - Óscar Martínez González
- Hospital Universitario del Tajo, Critical Care Department, Universidad Alfonso X El Sabio, Spain
| |
Collapse
|
5
|
Jimenez Tejero E, Lopez-Alcalde J, Correa-Pérez A, Stallings E, Gaetano Gil A, Del Campo Albendea L, Mateos-Haro M, Fernandez-Felix BM, Stallings R, Alvarez-Diaz N, García Laredo E, Solier A, Fernández-Martínez E, Morillo Guerrero R, de Miguel M, Perez R, Antequera A, Muriel A, Jimenez D, Zamora J. Sex as a prognostic factor for mortality in adults with acute symptomatic pulmonary embolism. Cochrane Database Syst Rev 2025; 3:CD013835. [PMID: 40110896 PMCID: PMC12043200 DOI: 10.1002/14651858.cd013835.pub2] [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: 03/22/2025]
Abstract
BACKGROUND Pulmonary embolism (PE) is relatively common worldwide. It is a serious condition that can be life-threatening. Studies on the relationship between adverse outcomes of this condition and whether a patient is male or female have yielded inconsistent results. Determining whether there is an association between sex and short-term mortality in patients with acute PE is important as this information may help guide different approaches to PE monitoring and treatment. OBJECTIVES To determine whether sex (i.e. being a male or a female patient) is an independent prognostic factor for predicting mortality in adults with acute symptomatic pulmonary embolism. SEARCH METHODS The Cochrane Vascular Information Specialist searched the Cochrane Vascular Specialised Register, CENTRAL, MEDLINE, Embase, and CINAHL databases, and the World Health Organization International Clinical Trials Registry Platform and ClinicalTrials.gov trials register up to 17 February 2023. We scanned conference abstracts and reference lists of included studies and systematic reviews. We also contacted experts to identify additional studies. There were no restrictions with respect to language or date of publication. SELECTION CRITERIA We included phase 2-confirmatory prognostic studies, that is, any longitudinal study (prospective or retrospective) evaluating the independent association between sex (male or female) and mortality in adults with acute PE. DATA COLLECTION AND ANALYSIS We followed the Checklist for Critical Appraisal and Data Extraction for Systematic Reviews of prognostic factor studies (CHARMS-PF) and the Cochrane Prognosis Methods Group template for prognosis reviews. Two review authors independently screened the studies, extracted data, assessed the risk of bias according to the Quality in Prognosis Studies (QUIPS) tool, and assessed the certainty of the evidence (GRADE). Meta-analyses were performed by pooling adjusted estimates. When meta-analysis was not possible, we reported the main results narratively. MAIN RESULTS We included seven studies (726,293 participants), all of which were retrospective cohort studies with participants recruited and managed in hospitals between 2000 and 2018. Studies took place in the USA, Spain, and Japan. Most studies were multicentre. None were conducted in low- or middle-income countries. The participants' mean age ranged from 62 to 69 years, and the proportion of females was higher in six of the seven studies, ranging from 46% to 60%. Sex and gender terms were used inconsistently. Participants received different PE treatments: reperfusion, inferior vena cava filter, anticoagulation, and haemodynamic/respiratory support. The prognostication time (the point from which the outcome was predicted) was frequently omitted. The included studies provided data for three of our outcomes of interest. We did not consider any of the studies to be at an overall low risk of bias for any of the outcomes analysed. We judged the certainty of the evidence as moderate to low due to imprecision and risk of bias. We found moderate-certainty evidence (due to imprecision) that for female patients there is likely a small but clinically important reduction in all-cause mortality at 30 days (odds ratio (OR) 0.81, 95% confidence interval (CI) 0.72 to 0.92; I2 = 0%; absolute risk difference (ARD) 24 fewer deaths in women per 1000 participants, 95% CI 35 to 10 fewer; 2 studies, 17,627 participants). However, the remaining review outcomes do not indicate lower mortality in female patients. There is low-certainty evidence (due to serious risk of bias and imprecision) indicating that for females with PE, there may be a small but clinically important increase in all-cause hospital mortality (OR 1.11, 95% CI 1.00 to 1.22; I2 = 21.7%; 95% prediction interval (PI) 0.76 to 1.61; ARD 13 more deaths in women per 1000 participants, 95% CI 0 to 26 more; 3 studies, 611,210 participants). There is also low-certainty evidence (due to very serious imprecision) indicating that there may be little to no difference between males and females in PE-related mortality at 30 days (OR 1.08, 95% CI 0.55 to 2.12; I2 = 0%; ARD 4 more deaths in women per 1000 participants, 95% CI 22 fewer to 50 more; 2 studies, 3524 participants). No study data was found for the other outcomes, including sex-specific mortality data at one year. Moreover, due to insufficient studies, many of our planned methods were not implemented. In particular, we were unable to conduct assessments of heterogeneity or publication bias or subgroup and sensitivity analyses. AUTHORS' CONCLUSIONS The evidence is uncertain about sex (being male or female) as an independent prognostic factor for predicting mortality in adults with PE. We found that, for female patients with PE, there is likely a small but clinically important reduction in all-cause mortality at 30 days relative to male patients. However, this result should be interpreted cautiously, as the remaining review outcomes do not point to an association between being female and having a lower risk of death. In fact, the evidence in the review also suggested that, in female patients, there may be a small but clinically important increase in all-cause hospital mortality. It also showed that there may be little to no difference in PE-related mortality at 30 days between male and female patients. There is currently no study evidence from longitudinal studies for our other review outcomes. Although the available evidence is conflicting and therefore cannot support a recommendation for or against routinely considering sex to quantify prognosis or to guide personalised therapeutic approaches for patients with PE, this Cochrane review offers information to guide future primary research and systematic reviews.
Collapse
Affiliation(s)
- Elena Jimenez Tejero
- Clinical Biostatistics Unit, Hospital Universitario Ramón y Cajal (IRYCIS), Madrid, Spain
- Faculty of Medicine, Universidad Francisco de Vitoria, Pozuelo de Alarcón, Spain
- Cochrane Associate Centre of Madrid, Madrid, Spain
| | - Jesús Lopez-Alcalde
- Faculty of Medicine, Universidad Francisco de Vitoria, Pozuelo de Alarcón, Spain
- Cochrane Associate Centre of Madrid, Madrid, Spain
- Clinical Biostatistics Unit, Hospital Universitario Ramón y Cajal (IRYCIS); CIBER Epidemiology and Public Health (CIBERESP), Madrid, Spain
- Institute for Complementary and Integrative Medicine, University Hospital Zurich; University of Zurich, Zurich, Switzerland
| | - Andrea Correa-Pérez
- Hospital Pharmacy and Medical Devices Department, Hospital Central de la Defensa "Gomez Ulla", Madrid, Spain
| | - Elena Stallings
- Clinical Biostatistics Unit, Hospital Universitario Ramón y Cajal (IRYCIS); CIBER Epidemiology and Public Health (CIBERESP), Madrid, Spain
| | - Andrea Gaetano Gil
- Clinical Biostatistics Unit, Hospital Universitario Ramón y Cajal (IRYCIS), Madrid, Spain
| | - Laura Del Campo Albendea
- Clinical Biostatistics Unit, Hospital Universitario Ramón y Cajal (IRYCIS); CIBER Epidemiology and Public Health (CIBERESP), Madrid, Spain
| | - Miriam Mateos-Haro
- Clinical Biostatistics Unit, Hospital Universitario Ramón y Cajal (IRYCIS), Madrid, Spain
| | - Borja Manuel Fernandez-Felix
- Clinical Biostatistics Unit, Hospital Universitario Ramón y Cajal (IRYCIS); CIBER Epidemiology and Public Health (CIBERESP), Madrid, Spain
| | - Raymond Stallings
- School of Pharmacy and Biomolecular Sciences, RCSI University of Medicine and Health Sciences, Dublin, Ireland
| | | | - Eduardo García Laredo
- Faculty of Health Sciences, Universidad Internacional de La Rioja (UNIR), Logroño, Spain
- Comet Global Innovation SL, Barcelona, Spain
| | - Aurora Solier
- Respiratory Department, Hospital Universitario Ramón y Cajal (IRYCIS), Madrid, Spain
| | | | - Raquel Morillo Guerrero
- Department of Pneumology, Hospital Universitario Ramón y Cajal (IRYCIS); CIBER Centro de Investigación Biomédica en Red de Enfermedades Respiratorias (CIBERES), Madrid, Spain
| | - Marcos de Miguel
- Department of Anesthesiology and Intensive Care, Hospital Universitari Vall d'Hebron, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Raquel Perez
- Respiratory Department, Hospital Universitario 12 de Octubre, Universidad Complutense Madrid, Madrid, Spain
| | - Alba Antequera
- International Health Department, ISGlobal, Hospital Clínic - Universitat de Barcelona, Barcelona, Spain
| | - Alfonso Muriel
- Clinical Biostatistics Unit, Hospital Universitario Ramón y Cajal (IRYCIS); CIBER Epidemiology and Public Health (CIBERESP), Madrid, Spain
- Department of Nursing and Physiotherapy, Universidad de Alcalá, Alcalá De Henares, Spain
| | - David Jimenez
- Respiratory Department, Hospital Universitario Ramón y Cajal (IRYCIS), Madrid, Spain
- Medicine Department, Universidad de Alcalá (IRYCIS), Madrid, Spain
- CIBER Enfermedades Respiratorias, CIBERES, Madrid, Spain
| | - Javier Zamora
- Clinical Biostatistics Unit, Hospital Universitario Ramón y Cajal (IRYCIS); CIBER Epidemiology and Public Health (CIBERESP), Madrid, Spain
| |
Collapse
|
6
|
Hager DN, Zhu Y, Sohn I, Stubblefield WB, Streiff MB, Gaglani M, Steingrub JS, Duggal A, Felzer JR, O'Rourke M, Peltan ID, Mohamed A, Stiller R, Wilson JG, Qadir N, Ginde AA, Zepeski AE, Mallow C, Lauring AS, Johnson NJ, Gibbs KW, Kwon JH, Self WH. Effectiveness of the Original Monovalent Messenger RNA Coronavirus Disease 2019 (COVID-19) Vaccination Series Against Hospitalization for COVID-19-Associated Venous Thromboembolism. J Infect Dis 2025; 231:378-385. [PMID: 39405261 PMCID: PMC12063076 DOI: 10.1093/infdis/jiae502] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2024] [Revised: 10/04/2024] [Accepted: 10/11/2024] [Indexed: 02/06/2025] Open
Abstract
BACKGROUND Coronavirus disease 2019 (COVID-19) is a strong risk factor for venous thromboembolism (VTE). Few studies have evaluated the effectiveness of COVID-19 vaccination in preventing hospitalization for COVID-19 with VTE. METHODS Adults hospitalized at 21 sites between March 2021 and October 2022 with symptoms of acute respiratory illness were assessed for COVID-19, completion of the original monovalent messenger RNA (mRNA) COVID-19 vaccination series, and VTE. Prevalence of VTE was compared between unvaccinated and vaccinated patients with COVID-19. The vaccine effectiveness (VE) in preventing COVID-19 hospitalization with VTE was calculated using a test-negative design. The VE was also stratified by predominant circulating severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) variant. RESULTS Among 18 811 patients (median age [interquartile range], 63 [50-73] years; 49% women; 59% non-Hispanic white, 20% non-Hispanic black, and 14% Hispanic; and median of 2 comorbid conditions [interquartile range, 1-3]), 9792 were admitted with COVID-19 (44% vaccinated), and 9019 were test-negative controls (73% vaccinated). Among patients with COVID-19, 601 had VTE diagnosed by hospital day 28, of whom 170 were vaccinated. VTE was more common among unvaccinated than vaccinated patients with COVID-19 (7.8% vs 4.0%; P = .001). The VE against COVID-19 hospitalization with VTE was 84% overall (95% confidence interval, 80%-87%), and VE stratified by predominant circulating variant was 88% (73%-95%) for Alpha, 93% (90%-95%) for Delta, and 68% (58%-76%) for Omicron variants. CONCLUSIONS Vaccination with the original monovalent mRNA series was associated with a decrease in COVID-19 hospitalization with VTE, though data detailing prior history of VTE and use of anticoagulation were not available. These findings will inform risk-benefit considerations for those considering vaccination.
Collapse
Affiliation(s)
- David N Hager
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Yuwei Zhu
- Department of Biostatistics, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Ine Sohn
- Department of Biostatistics, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - William B Stubblefield
- Department of Emergency Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Michael B Streiff
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Manjusha Gaglani
- Baylor Scott and White Health, Baylor College of Medicine—Temple, Texas A&M University College of Medicine, Temple, Texas, USA
| | - Jay S Steingrub
- Department of Medicine, Baystate Medical Center, Springfield, Massachusetts, USA
| | - Abhijit Duggal
- Department of Medicine, Cleveland Clinic, Cleveland, Ohio, USA
| | - Jamie R Felzer
- Department of Medicine, Emory University, Atlanta, Georgia, USA
| | - Mary O'Rourke
- Department of Emergency Medicine and Medicine, Hennepin County Medical Center, Minneapolis, Minnesota, USA
| | - Ithan D Peltan
- Department of Medicine, Intermountain Medical Center, Murray, Utah, USA
- Department of Medicine, University of Utah, Salt Lake City, Utah, USA
| | - Amira Mohamed
- Department of Medicine, Montefiore Medical Center, Albert Einstein College of Medicine, Bronx, New York, USA
| | - Robin Stiller
- Division of Pulmonary, Allergy and Critical Care Medicine, Oregon Health and Sciences University, Portland, Oregon, USA
| | - Jennifer G Wilson
- Department of Emergency Medicine, Stanford University School of Medicine, Stanford, California, USA
| | - Nida Qadir
- Department of Medicine, University of California-Los Angeles, Los Angeles, California, USA
| | - Adit A Ginde
- Department of Emergency Medicine, University of Colorado School of Medicine, Aurora, Colorado, USA
| | - Anne E Zepeski
- Department of Emergency Medicine, University of Iowa, Iowa City, Iowa, USA
| | | | - Adam S Lauring
- Departments of Internal Medicine and Microbiology and Immunology, University of Michigan, Ann Arbor, Michigan, USA
| | - Nicholas J Johnson
- Department of Emergency Medicine and Division of Pulmonary, Critical Care and Sleep Medicine, University of Washington, Seattle, Washington, USA
| | - Kevin W Gibbs
- Department of Medicine, Wake Forest School of Medicine, Winston-Salem, North Carolina, USA
| | - Jennie H Kwon
- Department of Medicine, Washington University, St. Louis, Missouri, USA
| | - Wesley H Self
- Department of Emergency Medicine and Vanderbilt Institute for Clinical and Translational Research, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| |
Collapse
|
7
|
Subramaniam S, Jose A, Kenney D, O’Connell AK, Bosmann M, Douam F, Crossland N. Challenging the notion of endothelial infection by SARS-CoV-2: insights from the current scientific evidence. Front Immunol 2025; 16:1443932. [PMID: 39967675 PMCID: PMC11832389 DOI: 10.3389/fimmu.2025.1443932] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Accepted: 01/14/2025] [Indexed: 02/20/2025] Open
Affiliation(s)
- Saravanan Subramaniam
- Department of Pharmacology and Toxicology, Massachusetts College of Pharmacy and Health Sciences, Boston, MA, United States
- Renal Section, Department of Medicine, Chobanian & Avedisian School of Medicine, Boston University, Boston, MA, United States
| | - Asha Jose
- Renal Section, Department of Medicine, Chobanian & Avedisian School of Medicine, Boston University, Boston, MA, United States
| | - Devin Kenney
- Department of Virology, Immunology and Microbiology, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, United States
- National Emerging Infectious Diseases Laboratories (NEIDL), Boston University, Boston, MA, United States
| | - Aoife K. O’Connell
- National Emerging Infectious Diseases Laboratories (NEIDL), Boston University, Boston, MA, United States
| | - Markus Bosmann
- Department of Medicine, Pulmonary Center, Chobanian & Avedisian School of Medicine, Boston University, Boston, MA, United States
- Department of Pathology and Laboratory Medicine, Chobanian & Avedisian School of Medicine, Boston University, Boston, MA, United States
| | - Florian Douam
- Department of Virology, Immunology and Microbiology, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, United States
- National Emerging Infectious Diseases Laboratories (NEIDL), Boston University, Boston, MA, United States
| | - Nicholas Crossland
- Department of Virology, Immunology and Microbiology, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, United States
- National Emerging Infectious Diseases Laboratories (NEIDL), Boston University, Boston, MA, United States
- Department of Pathology and Laboratory Medicine, Chobanian & Avedisian School of Medicine, Boston University, Boston, MA, United States
| |
Collapse
|
8
|
Zaki DP, Zeng E, Duet ML, Stone CE, Giglio RS, Tapp MW, Llull R, Calder BW, Robinson JM. Impact of COVID-19 on Thrombotic Complications in Microsurgery: Deep Inferior Epigastric Perforator Flap Outcomes Amid Pandemic. PLASTIC AND RECONSTRUCTIVE SURGERY-GLOBAL OPEN 2025; 13:e6544. [PMID: 39958714 PMCID: PMC11828035 DOI: 10.1097/gox.0000000000006544] [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: 06/09/2024] [Accepted: 12/13/2024] [Indexed: 02/18/2025]
Abstract
Background Emerging research underscores the heightened risk of vasculitis and microvascular thrombosis in COVID-19 patients, alongside concerns about prothrombotic events post-severe acute respiratory syndrome coronavirus 2 vaccination. Following the pandemic's end, we sought a comprehensive analysis to elucidate its impact on microsurgical thrombosis rates, informed by empirical and anecdotal evidence. Methods An institutional review board-approved retrospective review analyzed autologous breast reconstruction cases in women from January 2019 to March 2022. Data on patient history, COVID-19 infection, vaccination status, and postoperative complications were collected. Patients were categorized as prepandemic and pandemic, and based on COVID-19 influence (infection or vaccination) for statistical evaluation. Results Among 527 patients, 216 underwent surgery prepandemic and 311 during the pandemic, revealing thrombotic event rates of 3.2% and 5.4%, respectively. Further comparative analysis showed no significant difference in thrombotic events among patients affected by COVID-19 through infection or vaccination during the pandemic. Conclusions Contrary to concerns, COVID-19 infection or vaccination status does not significantly increase thrombotic event rates in deep inferior epigastric perforator flap breast reconstructions. This study offers vital insights, affirming the safety and efficacy of microsurgical procedures amid the pandemic, thereby guiding microsurgeons in optimizing patient care in the post-COVID-19 era.
Collapse
Affiliation(s)
- Daniel P. Zaki
- From the Department of Plastic and Reconstructive Surgery, Atrium Health Wake Forest Baptist, Winston-Salem, NC
| | - Eric Zeng
- Wake Forest University School of Medicine, Winston-Salem, NC
| | - Mary L. Duet
- From the Department of Plastic and Reconstructive Surgery, Atrium Health Wake Forest Baptist, Winston-Salem, NC
| | - Courtney E. Stone
- From the Department of Plastic and Reconstructive Surgery, Atrium Health Wake Forest Baptist, Winston-Salem, NC
| | | | - Marion W. Tapp
- From the Department of Plastic and Reconstructive Surgery, Atrium Health Wake Forest Baptist, Winston-Salem, NC
| | - Ramon Llull
- From the Department of Plastic and Reconstructive Surgery, Atrium Health Wake Forest Baptist, Winston-Salem, NC
| | - Bennett W. Calder
- From the Department of Plastic and Reconstructive Surgery, Atrium Health Wake Forest Baptist, Winston-Salem, NC
| | - John M. Robinson
- From the Department of Plastic and Reconstructive Surgery, Atrium Health Wake Forest Baptist, Winston-Salem, NC
| |
Collapse
|
9
|
Xie R, Tan D, Liu B, Xiao G, Gong F, Zhang Q, Qi L, Zheng S, Yuan Y, Yang Z, Chen Y, Fei J, Xu D. Acute respiratory distress syndrome (ARDS): from mechanistic insights to therapeutic strategies. MedComm (Beijing) 2025; 6:e70074. [PMID: 39866839 PMCID: PMC11769712 DOI: 10.1002/mco2.70074] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2024] [Revised: 12/22/2024] [Accepted: 01/01/2025] [Indexed: 01/28/2025] Open
Abstract
Acute respiratory distress syndrome (ARDS) is a clinical syndrome of acute hypoxic respiratory failure caused by diffuse lung inflammation and edema. ARDS can be precipitated by intrapulmonary factors or extrapulmonary factors, which can lead to severe hypoxemia. Patients suffering from ARDS have high mortality rates, including a 28-day mortality rate of 34.8% and an overall in-hospital mortality rate of 40.0%. The pathophysiology of ARDS is complex and involves the activation and dysregulation of multiple overlapping and interacting pathways of systemic inflammation and coagulation, including the respiratory system, circulatory system, and immune system. In general, the treatment of inflammatory injuries is a coordinated process that involves the downregulation of proinflammatory pathways and the upregulation of anti-inflammatory pathways. Given the complexity of the underlying disease, treatment needs to be tailored to the problem. Hence, we discuss the pathogenesis and treatment methods of affected organs, including 2019 coronavirus disease (COVID-19)-related pneumonia, drowning, trauma, blood transfusion, severe acute pancreatitis, and sepsis. This review is intended to provide a new perspective concerning ARDS and offer novel insight into future therapeutic interventions.
Collapse
Affiliation(s)
- Rongli Xie
- Department of General SurgeryRuijin Hospital Lu Wan Branch, Shanghai Jiaotong University School of MedicineShanghaiChina
| | - Dan Tan
- Department of General SurgeryRuijin Hospital Lu Wan Branch, Shanghai Jiaotong University School of MedicineShanghaiChina
| | - Boke Liu
- Department of UrologyRuijin Hospital, Shanghai Jiaotong University School of MedicineShanghaiChina
| | - Guohui Xiao
- Department of General Surgery, Pancreatic Disease CenterRuijin Hospital, Shanghai Jiaotong University School of MedicineShanghaiChina
| | - Fangchen Gong
- Department of EmergencyRuijin Hospital, Shanghai Jiaotong University School of MedicineShanghaiChina
| | - Qiyao Zhang
- Department of RadiologySödersjukhuset (Southern Hospital)StockholmSweden
| | - Lei Qi
- Department of Molecular and Human GeneticsBaylor College of MedicineHoustonTexasUSA
| | - Sisi Zheng
- Department of RadiologyThe First Affiliated Hospital of Zhejiang Chinese Medical UniversityHangzhouZhejiangChina
| | - Yuanyang Yuan
- Department of Immunology and MicrobiologyShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Zhitao Yang
- Department of EmergencyRuijin Hospital, Shanghai Jiaotong University School of MedicineShanghaiChina
| | - Ying Chen
- Department of EmergencyRuijin Hospital, Shanghai Jiaotong University School of MedicineShanghaiChina
| | - Jian Fei
- Department of General Surgery, Pancreatic Disease CenterRuijin Hospital, Shanghai Jiaotong University School of MedicineShanghaiChina
| | - Dan Xu
- Department of EmergencyRuijin Hospital, Shanghai Jiaotong University School of MedicineShanghaiChina
| |
Collapse
|
10
|
Fekete M, Lehoczki A, Szappanos Á, Toth A, Mahdi M, Sótonyi P, Benyó Z, Yabluchanskiy A, Tarantini S, Ungvari Z. Cerebromicrovascular mechanisms contributing to long COVID: implications for neurocognitive health. GeroScience 2025; 47:745-779. [PMID: 39777702 PMCID: PMC11872997 DOI: 10.1007/s11357-024-01487-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2024] [Accepted: 12/17/2024] [Indexed: 01/11/2025] Open
Abstract
Long COVID (also known as post-acute sequelae of SARS-CoV-2 infection [PASC] or post-COVID syndrome) is characterized by persistent symptoms that extend beyond the acute phase of SARS-CoV-2 infection, affecting approximately 10% to over 30% of those infected. It presents a significant clinical challenge, notably due to pronounced neurocognitive symptoms such as brain fog. The mechanisms underlying these effects are multifactorial, with mounting evidence pointing to a central role of cerebromicrovascular dysfunction. This review investigates key pathophysiological mechanisms contributing to cerebrovascular dysfunction in long COVID and their impacts on brain health. We discuss how endothelial tropism of SARS-CoV-2 and direct vascular infection trigger endothelial dysfunction, impaired neurovascular coupling, and blood-brain barrier disruption, resulting in compromised cerebral perfusion. Furthermore, the infection appears to induce mitochondrial dysfunction, enhancing oxidative stress and inflammation within cerebral endothelial cells. Autoantibody formation following infection also potentially exacerbates neurovascular injury, contributing to chronic vascular inflammation and ongoing blood-brain barrier compromise. These factors collectively contribute to the emergence of white matter hyperintensities, promote amyloid pathology, and may accelerate neurodegenerative processes, including Alzheimer's disease. This review also emphasizes the critical role of advanced imaging techniques in assessing cerebromicrovascular health and the need for targeted interventions to address these cerebrovascular complications. A deeper understanding of the cerebrovascular mechanisms of long COVID is essential to advance targeted treatments and mitigate its long-term neurocognitive consequences.
Collapse
Affiliation(s)
- Monika Fekete
- Institute of Preventive Medicine and Public Health, Semmelweis University, Budapest, Hungary
| | - Andrea Lehoczki
- Institute of Preventive Medicine and Public Health, Semmelweis University, Budapest, Hungary.
- Doctoral College, Health Sciences Program, Semmelweis University, Budapest, Hungary.
| | - Ágnes Szappanos
- Heart and Vascular Center, Semmelweis University, Budapest, Hungary
- Department of Rheumatology and Clinical Immunology, Semmelweis University, Budapest, Hungary
| | - Attila Toth
- Division of Clinical Physiology, Department of Cardiology, Faculty of Medicine, University of Debrecen, Debrecen, 4032, Hungary
- Research Centre for Molecular Medicine, University of Debrecen, Debrecen, 4032, Hungary
| | - Mohamed Mahdi
- Laboratory of Retroviral Biochemistry, Department of Biochemistry and Molecular Biology, University of Debrecen, 4032, Debrecen, Hungary
- Infectology Clinic, University of Debrecen Clinical Centre, 4031, Debrecen, Hungary
| | - Péter Sótonyi
- Department of Vascular and Endovascular Surgery, Heart and Vascular Centre, Semmelweis University, 1122, Budapest, Hungary
| | - Zoltán Benyó
- Institute of Translational Medicine, Semmelweis University, 1094, Budapest, Hungary
- Cerebrovascular and Neurocognitive Disorders Research Group, HUN-REN , Semmelweis University, 1094, Budapest, Hungary
| | - Andriy Yabluchanskiy
- Vascular Cognitive Impairment, Neurodegeneration and Healthy Brain Aging Program, Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- International Training Program in Geroscience, Doctoral College/Institute of Preventive Medicine and Public Health, Semmelweis University, Budapest, Hungary
- Stephenson Cancer Center, University of Oklahoma, Oklahoma City, OK, USA
- Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Department of Health Promotion Sciences, College of Public Health, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Stefano Tarantini
- Vascular Cognitive Impairment, Neurodegeneration and Healthy Brain Aging Program, Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- International Training Program in Geroscience, Doctoral College/Institute of Preventive Medicine and Public Health, Semmelweis University, Budapest, Hungary
- Stephenson Cancer Center, University of Oklahoma, Oklahoma City, OK, USA
- Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Department of Health Promotion Sciences, College of Public Health, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Zoltan Ungvari
- Vascular Cognitive Impairment, Neurodegeneration and Healthy Brain Aging Program, Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- International Training Program in Geroscience, Doctoral College/Institute of Preventive Medicine and Public Health, Semmelweis University, Budapest, Hungary
- Stephenson Cancer Center, University of Oklahoma, Oklahoma City, OK, USA
- Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Department of Health Promotion Sciences, College of Public Health, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| |
Collapse
|
11
|
López-Jiménez C, Gutiérrez A, Juliao Caamaño DS, Soto Alsar J, Catoya Villa JL, Blanco Abad C, Morón B, Ortega Morán L, Martín M, Muñoz Martín AJ. Impact of COVID-19 in the incidence of venous thromboembolism (VTE) and clinical outcomes in cancer patients: a cohort study. Clin Transl Oncol 2025; 27:756-769. [PMID: 39090424 DOI: 10.1007/s12094-024-03635-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2024] [Accepted: 07/20/2024] [Indexed: 08/04/2024]
Abstract
PURPOSE To determine the incidence of VTE and clinical outcomes in a cohort of cancer patients and COVID-19 infection, and to establish possible predictive factors of VTE. METHODS/PATIENTS A single-center retrospective cohort study was performed to determine the incidence of VTE and mortality in 118 cancer patients with SARS-CoV-2 infection from March to August 2020. We calculated individual Khorana Risk and CATS-MICA scores in order to evaluate their utility to identify risk of VTE or death. Continuous variables were compared using Wilcoxon or Student's T test, and categorical variables were compared using the Chi-Square or Fisher's exact text among patients with and without VTE. A Log-Rank test was performed to detect mortality differences between the groups. RESULTS A total of 118 patients were included. VTE global incidence was 4.2% (n = 5), and mortality 25.4% (n = 30). Obesity (p = 0.05), recent chemotherapy (p = 0.049) and use of steroids (p = 0.006) were related to higher risk of VTE in the univariate analysis, although they were not confirmed in the multivariate analysis as independent risk factors. Statistically significant differences in all-cause, COVID-19-related and cancer-related mortality according to the Khorana risk score (KRS) were observed. CATS-MICA score (CMS) also showed statistically significant differences in mortality between low- and high-risk patients. Prediction of risk of VTE development with these scores showed a tendency towards significance. CONCLUSIONS In this cohort, VTE incidence was similar to previously reported in the general population with SARS-CoV-2 infection. KRS was associated with overall and specific-cause mortality, and might be a useful prognostic tool in this setting.
Collapse
Affiliation(s)
- Carlos López-Jiménez
- Medical Oncology Department, Hospital General Universitario Gregorio Marañón, Calle del Dr. Esquerdo, 46, 28007, Madrid, Spain.
| | - Ana Gutiérrez
- Medical Oncology Department, Hospital General Universitario Gregorio Marañón, Calle del Dr. Esquerdo, 46, 28007, Madrid, Spain
| | - David Salomón Juliao Caamaño
- Medical Oncology Department, Hospital General Universitario Gregorio Marañón, Calle del Dr. Esquerdo, 46, 28007, Madrid, Spain
| | - Javier Soto Alsar
- Medical Oncology Department, Hospital General Universitario Gregorio Marañón, Calle del Dr. Esquerdo, 46, 28007, Madrid, Spain
| | | | - Carmen Blanco Abad
- Medical Oncology Department, Hospital Universitario Miguel Servet, Zaragoza, Spain
| | - Blanca Morón
- Medical Oncology Department, Hospital Universitario Ramón y Cajal, Madrid, Spain
| | - Laura Ortega Morán
- Medical Oncology Department, Hospital General Universitario Gregorio Marañón, Calle del Dr. Esquerdo, 46, 28007, Madrid, Spain
| | - Miguel Martín
- Medical Oncology Department, Hospital General Universitario Gregorio Marañón, Calle del Dr. Esquerdo, 46, 28007, Madrid, Spain
| | - Andrés Jesús Muñoz Martín
- Medical Oncology Department, Hospital General Universitario Gregorio Marañón, Calle del Dr. Esquerdo, 46, 28007, Madrid, Spain
| |
Collapse
|
12
|
Licata A, Seidita A, Como S, de Carlo G, Cammilleri M, Bonica R, Soresi M, Veronese N, Chianetta R, Citarrella R, Giannitrapani L, Barbagallo M. Herbal and Dietary Supplements as Adjunctive Treatment for Mild SARS-CoV-2 Infection in Italy. Nutrients 2025; 17:230. [PMID: 39861359 PMCID: PMC11767322 DOI: 10.3390/nu17020230] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2024] [Revised: 01/02/2025] [Accepted: 01/03/2025] [Indexed: 01/27/2025] Open
Abstract
During the COVID-19 pandemic, several observational studies proved a certain efficacy of nutraceuticals, herbal products, and other dietary supplements as adjuvant therapies used alongside antiviral drugs. Although their use has not been widespread in Italy, according to preliminary evidence, many supplements with demonstrated immunomodulatory effects, such as vitamins C and D, herbal medicines and essential oils, might relieve the respiratory symptoms of COVID-19, since SARS-CoV-2 can activate inflammasome-mediated inflammatory signaling pathways. Other observational studies have shown that herbal treatments, such as Echinacea purpurea and ginseng, help alleviate respiratory symptoms and reduce serum levels of inflammatory cytokines, which are typically overexpressed in both adult and pediatric SARS-CoV-2 patients. Further, vitamins C and D can attenuate the immune response thanks to their cytokine suppression ability and to their known antimicrobial activity and potential to modulate T helper cell response. The strong immune response triggered by SARS-CoV-2 infection is responsible for the severity of the disease. Preliminary data have also shown that L-arginine, an endothelial-derived relaxing factor, is able to modulate endothelial damage, which appears to be one of the main targets of this systemic disease. Finally, some essential oils and their isolated compounds, such as eucalyptol, may be helpful in reducing many of the respiratory symptoms of COVID-19, although others, such as menthol, are not recommended, since it can lead to an undervaluation of the clinical status of a patient. In this narrative review, despite the lack of strong evidence in this field, we aimed to give an overview of the current available literature (mainly observational and cross-sectional studies) regarding herbal products and dietary supplements and their use in the treatment of mild disease from SARS-CoV-2 infection. Obviously, dietary supplements and herbal products do not constitute a standardized treatment for COVID-19 disease, but they could represent an adjunctive and useful treatment when used together with antivirals.
Collapse
Affiliation(s)
- Anna Licata
- Unit of Internal Medicine, AOU Policlinico “P. Giaccone”, Department of Health Promotion Sciences, Maternal and Infant Care, Internal Medicine and Medical Specialties (PROMISE), University of Palermo, 90134 Palermo, Italy; (S.C.); (M.C.); (N.V.)
| | - Aurelio Seidita
- Unit of Internal Medicine, “V. Cervello” Hospital, Ospedali Riuniti “Villa Sofia-Cervello”, 90146 Palermo, Italy; (A.S.)
- Department of Health Promotion Sciences, Maternal and Infant Care, Internal Medicine and Medical Specialties (PROMISE), University of Palermo, 90146 Palermo, Italy
- Institute for Biomedical Research and Innovation (IRIB), National Research Council (CNR), 90146 Palermo, Italy
| | - Silvia Como
- Unit of Internal Medicine, AOU Policlinico “P. Giaccone”, Department of Health Promotion Sciences, Maternal and Infant Care, Internal Medicine and Medical Specialties (PROMISE), University of Palermo, 90134 Palermo, Italy; (S.C.); (M.C.); (N.V.)
| | - Gabriele de Carlo
- Unit of Internal Medicine, “V. Cervello” Hospital, Ospedali Riuniti “Villa Sofia-Cervello”, 90146 Palermo, Italy; (A.S.)
| | - Marcella Cammilleri
- Unit of Internal Medicine, AOU Policlinico “P. Giaccone”, Department of Health Promotion Sciences, Maternal and Infant Care, Internal Medicine and Medical Specialties (PROMISE), University of Palermo, 90134 Palermo, Italy; (S.C.); (M.C.); (N.V.)
| | - Roberta Bonica
- Unit of Internal Medicine, AOU Policlinico “P. Giaccone”, Department of Health Promotion Sciences, Maternal and Infant Care, Internal Medicine and Medical Specialties (PROMISE), University of Palermo, 90134 Palermo, Italy; (S.C.); (M.C.); (N.V.)
| | - Maurizio Soresi
- Unit of Internal Medicine, AOU Policlinico “P. Giaccone”, Department of Health Promotion Sciences, Maternal and Infant Care, Internal Medicine and Medical Specialties (PROMISE), University of Palermo, 90134 Palermo, Italy; (S.C.); (M.C.); (N.V.)
| | - Nicola Veronese
- Unit of Internal Medicine, AOU Policlinico “P. Giaccone”, Department of Health Promotion Sciences, Maternal and Infant Care, Internal Medicine and Medical Specialties (PROMISE), University of Palermo, 90134 Palermo, Italy; (S.C.); (M.C.); (N.V.)
| | - Roberta Chianetta
- Unit of Internal Medicine, AOU Policlinico “P. Giaccone”, Department of Health Promotion Sciences, Maternal and Infant Care, Internal Medicine and Medical Specialties (PROMISE), University of Palermo, 90134 Palermo, Italy; (S.C.); (M.C.); (N.V.)
| | - Roberto Citarrella
- Unit of Internal Medicine, AOU Policlinico “P. Giaccone”, Department of Health Promotion Sciences, Maternal and Infant Care, Internal Medicine and Medical Specialties (PROMISE), University of Palermo, 90134 Palermo, Italy; (S.C.); (M.C.); (N.V.)
| | - Lydia Giannitrapani
- Unit of Internal Medicine, AOU Policlinico “P. Giaccone”, Department of Health Promotion Sciences, Maternal and Infant Care, Internal Medicine and Medical Specialties (PROMISE), University of Palermo, 90134 Palermo, Italy; (S.C.); (M.C.); (N.V.)
- Institute for Biomedical Research and Innovation (IRIB), National Research Council (CNR), 90146 Palermo, Italy
| | - Mario Barbagallo
- Unit of Internal Medicine, AOU Policlinico “P. Giaccone”, Department of Health Promotion Sciences, Maternal and Infant Care, Internal Medicine and Medical Specialties (PROMISE), University of Palermo, 90134 Palermo, Italy; (S.C.); (M.C.); (N.V.)
| |
Collapse
|
13
|
Wang KC, Wang S, Serna J, Su F, Halvorson RT, Lansdown DA, Ma CB, Zhang AL. COVID-19 Diagnosis Within 6 Weeks of Arthroscopic Knee Surgery Leads to Higher Risk for Postoperative Venous Thromboembolism. Arthroscopy 2024:S0749-8063(24)01076-4. [PMID: 39709106 DOI: 10.1016/j.arthro.2024.12.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/15/2024] [Revised: 12/05/2024] [Accepted: 12/06/2024] [Indexed: 12/23/2024]
Abstract
PURPOSE To determine if preoperative infection with COVID-19 increased risk for postoperative venous thromboembolism (VTE) in patients undergoing arthroscopic knee surgery. METHODS PearlDiver Mariner 165 database was queried for patients undergoing knee arthroscopy between 2010 and October 2022. Patients were categorized by history of COVID-19 diagnosis and timing in relation to surgery. Multivariate logistic regression was performed to isolate the effect of COVID-19 diagnosis on postoperative VTE rates. Covariates included age, obesity, smoking, oral contraceptive pill use, hypertension, coronary artery disease, congestive heart failure, malignancy, renal disease, and diabetes. RESULTS A total of 954,294 patients met inclusion criteria, and 7,637 patients experienced VTE, including deep vein thrombosis (n = 5,830, 0.61%) and pulmonary embolism (n = 2,790, 0.29%). Patients with a COVID-19 diagnosis before surgery (7,858) had an overall higher incidence of VTE (1.72%) compared to patients without a preoperative COVID-19 diagnosis (0.81%) (P < .001). There was no difference in VTE incidence among patients with a preoperative COVID-19 diagnosis undergoing knee arthroscopy in the pre- and post-COVID-19 vaccination eras (odds ratio, 0.82; 95% CI, 0.51-1.31; P = .48). Multivariate regression accounting for covariates showed that patients with a preoperative COVID-19 diagnosis within 6 weeks before arthroscopy had significantly increased odds of experiencing postoperative VTE (odds ratio, 1.68; 95% CI, 1.09-2.45; P = .012). There was no significant difference in VTE risk among patients with a preoperative COVID-19 diagnosis between 6 and 12 weeks before arthroscopy and more than 12 weeks before arthroscopy compared to those with no prior COVID-19 diagnosis. CONCLUSIONS COVID-19 diagnosis within 6 weeks preceding arthroscopic knee surgery leads to a significantly higher risk for postoperative VTE. LEVEL OF EVIDENCE Level III, retrospective case control study.
Collapse
Affiliation(s)
- Kevin C Wang
- Department of Orthopedic Surgery, University of California San Francisco, San Francisco, California, USA
| | - Sophia Wang
- Department of Orthopedic Surgery, University of California San Francisco, San Francisco, California, USA
| | - Juan Serna
- Department of Orthopedic Surgery, University of California San Francisco, San Francisco, California, USA
| | - Favian Su
- Department of Orthopedic Surgery, University of California San Francisco, San Francisco, California, USA
| | - Ryan T Halvorson
- Department of Orthopedic Surgery, University of California San Francisco, San Francisco, California, USA
| | - Drew A Lansdown
- Department of Orthopedic Surgery, University of California San Francisco, San Francisco, California, USA
| | - C Benjamin Ma
- Department of Orthopedic Surgery, University of California San Francisco, San Francisco, California, USA
| | - Alan L Zhang
- Department of Orthopedic Surgery, University of California San Francisco, San Francisco, California, USA.
| |
Collapse
|
14
|
El-Menyar A, Ramzee AF, Asim M, Shahid F, Ata YM, El Baba H, Fino A, Nair AP, Peralta R, Almaslamani MA, Al Suwaidi J, Al-Thani H, Rizoli S. COVID-19 Increases the Risk of New Myocardial Infarction in Patients with Old Myocardial Infarction: A Retrospective Observational Study. CLINICAL MEDICINE INSIGHTS-CARDIOLOGY 2024; 18:11795468241301133. [PMID: 39697349 PMCID: PMC11653445 DOI: 10.1177/11795468241301133] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/25/2024] [Accepted: 10/07/2024] [Indexed: 12/20/2024]
Abstract
Background We aimed to investigate the incidence of new acute myocardial infarction (AMI), in patients with Coronavirus disease (COVID-19) who had old MI. We hypothesized that COVID-19 increases the rate of repeated AMI in this population regardless of age and gender. Methods A retrospective analysis was conducted for adult patients admitted with COVID-19 and developed thromboembolic event (TEE) in 2020. Patients were categorized based on the history of old MI, new MI, age, and gender. Results Among 16,903 patients with COVID-19 who were admitted, 210 (1.2%) developed TEE (89% were males, 55% were <55 years old, and 80.5% had an old MI). COVID-19 was severe in 32% of cases. AMI occurred in 160 patients (42.5% STEMI and 57.5% NSTEMI). In patients with prior MI, 92.5% developed another AMI. NSTEMI was higher in patients with severe COVID-19 than STEMI (33% vs 21%). Patients with severe COVID-19 had higher mortality (39.4% vs 5.6%), fewer rates of prior MI (74% vs 83%), hypertension (40% vs 60%), and STEMI (31.8% vs 46.5%) than mild COVID-19 patients. On multivariable analysis, COVID-19 severity was an independent predictor of mortality (OR10; 95%CI 1.62-67.19) after adjustment for age, gender, diabetes mellitus, C-reactive protein, serum Ferritin, Procalcitonin, and Fibrinogen values, and prior or new MI. Conclusions Patients with old MI could develop a new AMI in 80% of COVID-19. However, the mortality was higher in patients without a history of MI due to the severity of COVID-19. Attention should be given to patients who possess thrombotic risk factors in pandemics.
Collapse
Affiliation(s)
- Ayman El-Menyar
- Clinical Research, Trauma and Vascular Surgery, Hamad Medical Corporation, Doha, Qatar
- Clinical Medicine, Weill Cornell Medical College, Doha, Qatar
| | | | - Mohammad Asim
- Clinical Research, Trauma and Vascular Surgery, Hamad Medical Corporation, Doha, Qatar
| | - Fakhar Shahid
- Department of Surgery, Hamad Medical Corporation, Doha, Qatar
| | - Yaser M Ata
- Department of Surgery, Hamad Medical Corporation, Doha, Qatar
| | - Hamzah El Baba
- Department of Surgery, Hamad Medical Corporation, Doha, Qatar
| | - Areen Fino
- Department of Family Medicine, Hamad Medical Corporation, Doha, Qatar
| | - Arun P Nair
- Communicable Disease Center (CDC), Hamad Medical Corporation, Doha, Qatar
| | - Ruben Peralta
- Trauma Surgery Section, Hamad Medical Corporation, Doha, Qatar
- Department of Surgery, Universidad Nacional Pedro Henriquez Urena, Santo Domingo, Dominican Republic
| | - Muna A Almaslamani
- Communicable Disease Center (CDC), Hamad Medical Corporation, Doha, Qatar
| | - Jassim Al Suwaidi
- Department of Cardiology, Heart Hospital, Hamad Medical Corporation, Doha, Qatar
| | - Hassan Al-Thani
- Trauma Surgery Section, Hamad Medical Corporation, Doha, Qatar
| | - Sandro Rizoli
- Trauma Surgery Section, Hamad Medical Corporation, Doha, Qatar
| |
Collapse
|
15
|
Xu J, Ye R, Zhao J, Fan X, Xue K, Li X, Zhu X, Gao Y, Wang Y. Hospitalization costs in patients with stroke in southeastern China: a retrospective population-based cohort study, 2019-2022. Front Public Health 2024; 12:1442171. [PMID: 39583074 PMCID: PMC11582024 DOI: 10.3389/fpubh.2024.1442171] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2024] [Accepted: 10/23/2024] [Indexed: 11/26/2024] Open
Abstract
Introduction Stroke remains a predominant cause of mortality and accounts for one-third of all stroke-related fatalities worldwide. Increasing expenses associated with stroke are a matter of significant concern; however, this aspect has been insufficiently examined. Methods The purpose of this study was to analyze in-hospital stroke costs and explore potential factors influencing them across stroke subtypes. The records of stroke patients from 50 hospitals in southeastern China between 2019 and 2022 were reviewed using multistage stratified cluster random sampling. We focused on the cost patterns of four stroke types and used multivariate linear regression to identify cost determinants. Results A total of 417 (1.1%) patients had subarachnoid hemorrhage (SAH), 9309 (25.9%) had intracerebral hemorrhage (ICH), 22,248 (61.8%) had ischemic stroke (IS), and 4025 had transient ischemic attack (TIA). The number of stroke patients has sharply increased since the onset of COVID-19, with a majority of them being male (72.2%). Despite the fact that hospitalization costs are highest in tertiary hospitals (Chinese yuan [CNY] 30610.8/United States dollar [USD] 4551.0, interquartile range [IQR] 9944.9, 29668.4/1478.6, 4410.9), the majority of patients are admitted to tertiary hospitals (74.6%) or public hospitals (90.2%). Across all stroke subtypes, patients with SAH had the highest costs (CNY 93,454.9/USD13894.4, IQR 12273.2, 169920.0/1824.7, 25262.8), followed by those with ICH (CNY 48,724.2/USD 7244.0, IQR 16789.6, 57540.7/2496.2, 8554.8), IS (CNY 26,550.3/USD3947.4, IQR 8684.2, 28697.7/1291.1, 4266.6), and TIA (CNY 11,170.1/USD1660.7, IQR 6823.7, 12965.2/1014.5, 1927.6). Therapy fees comprised a significant portion of costs in ICH and IS cases (47.9% and 42.7%, respectively). Materials accounted for the highest proportion of expenses for SAH (56.1%), whereas patients with TIA spent more time on examinations (34.1%). Linear regression analysis revealed that length of stay (LOS), stroke subtype, hospital level, and stroke type were key factors influencing hospitalization costs. Discussion The visiting rate and charges were highest in tertiary public hospitals, and hospitalization costs were higher in hemorrhagic types of stroke than in ischemic types of stroke; the proportion of hospitalization cost categories varied among different types of stroke, with LOS, hospital type, and level substantially affecting hospitalization costs. Enhancing medical insurance reimbursement rates for hemorrhagic strokes, implementing a hierarchical medical system, tailoring cost categories to accommodate varying stroke subtypes, and shortening LOS may help alleviate the economic burden of stroke.
Collapse
Affiliation(s)
- Jing Xu
- Department of Rehabilitation, Shenzhen Second People’s Hospital, The First Affiliated Hospital, Shenzhen University School of Medicine, Shenzhen, Guangdong, China
| | - Ruixue Ye
- Department of Rehabilitation, Shenzhen Second People’s Hospital, The First Affiliated Hospital, Shenzhen University School of Medicine, Shenzhen, Guangdong, China
| | - Jingpu Zhao
- Department of Rehabilitation, Shenzhen Second People’s Hospital, The First Affiliated Hospital, Shenzhen University School of Medicine, Shenzhen, Guangdong, China
| | - Xuehui Fan
- Department of Rehabilitation, Shenzhen Second People’s Hospital, The First Affiliated Hospital, Shenzhen University School of Medicine, Shenzhen, Guangdong, China
| | - Kaiwen Xue
- Department of Rehabilitation, Shenzhen Second People’s Hospital, The First Affiliated Hospital, Shenzhen University School of Medicine, Shenzhen, Guangdong, China
| | - Xiaoxuan Li
- Rehabilitation Medical College, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Xiaolong Zhu
- Department of Rehabilitation, Shenzhen Second People’s Hospital, The First Affiliated Hospital, Shenzhen University School of Medicine, Shenzhen, Guangdong, China
| | - Yan Gao
- Department of Rehabilitation, Shenzhen Second People’s Hospital, The First Affiliated Hospital, Shenzhen University School of Medicine, Shenzhen, Guangdong, China
| | - Yulong Wang
- Department of Rehabilitation, Shenzhen Second People’s Hospital, The First Affiliated Hospital, Shenzhen University School of Medicine, Shenzhen, Guangdong, China
| |
Collapse
|
16
|
Morán LO, Mateo FJP, Balanyà RP, Revuelta JR, Martínez SR, Fombella JPB, Vázquez EMB, Caro NL, Langa JM, Fernández MS. SEOM clinical guidelines on venous thromboembolism (VTE) and cancer (2023). Clin Transl Oncol 2024; 26:2877-2901. [PMID: 39110395 PMCID: PMC11467034 DOI: 10.1007/s12094-024-03605-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/10/2024] [Indexed: 10/11/2024]
Abstract
The Spanish Society of Medical Oncology (SEOM) last published clinical guidelines on venous thromboembolism (VTE) and cancer in 2019, with a partial update in 2020. In this new update to the guidelines, SEOM seeks to incorporate recent evidence, based on a critical review of the literature, to provide practical current recommendations for the prophylactic and therapeutic management of VTE in patients with cancer. Special clinical situations whose management and/or choice of currently recommended therapeutic options (low-molecular-weight heparins [LMWHs] or direct-acting oral anticoagulants [DOACs]) is controversial are included.
Collapse
Affiliation(s)
- Laura Ortega Morán
- Hospital General Universitario Gregorio Marañón, Instituto de Investigación Sanitaria Gregorio Marañón, Madrid, Spain.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
17
|
Aliyeva N, Çalişkaner Öztürk B, Kiliçkiran Avci B, Atahan E. Treatment and long term follow-up results in patients with pulmonary vascular thrombosis related to COVID-19. Medicine (Baltimore) 2024; 103:e40319. [PMID: 39495981 PMCID: PMC11537611 DOI: 10.1097/md.0000000000040319] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/19/2024] [Accepted: 10/11/2024] [Indexed: 11/06/2024] Open
Abstract
Pulmonary embolism is a complication of COVID-19 infection. The aim of this study is to assess prognosis and treatment response, including incidences of chronicity, relapse, and mortality among outpatients diagnosed with COVID-19-related pulmonary embolism between 2020 and 2022. A total of 101 patients with pulmonary embolism, started on anticoagulation during or within a month of COVID-19 infection, were included after testing positive by PCR. Data about comorbidities, Pulmonary Embolism Severity Index scores, PE diagnostic modalities, biochemical parameters, and transthoracic echocardiographic findings at diagnosis and at 24-month follow-up were collected. Cardiac catheterization parameters were recorded and compared between groups at diagnosis and at the 24-month follow-up. Groups were comparable with respect to gender, age, body mass index, and comorbidity score. Use of Q-SPECT for diagnosis was found significantly higher in patients with COVID-19-related pulmonary embolism (P < .001). The incidence of deep vein thrombosis was similar. In the study group, 43.6% of patients received anticoagulants for 3 months, with 49.1% using low molecular weight heparin and 50.9% using direct oral anticoagulants. At 24 months, rate of patients continuing treatment was comparable between groups. Specific pulmonary artery blockage value was found to be higher in patients with chronic thromboembolic pulmonary hypertension compared to those who demonstrated a response to pulmonary embolism treatment (P = .009). No adverse effects of anticoagulant therapy were observed during course of treatment. Over 24-month follow-up period, mortality, relapse, chronic thromboembolic hypertension and thromboembolic disease was observed in 2%, 2.2%, 4.9%, and 9.9% of patients, respectively.
Collapse
Affiliation(s)
- Nigar Aliyeva
- Department of Chest Diseases, Istanbul University-Cerrahpaşa, Cerrahpaşa Faculty of Medicine, Istanbul, Turkey
| | - Buket Çalişkaner Öztürk
- Department of Chest Diseases, Istanbul University-Cerrahpaşa, Cerrahpaşa Faculty of Medicine, Istanbul, Turkey
| | - Burçak Kiliçkiran Avci
- Department of Cardiology, Istanbul University-Cerrahpaşa, Cerrahpaşa Faculty of Medicine, Istanbul, Turkey
| | - Ersan Atahan
- Department of Chest Diseases, Istanbul University-Cerrahpaşa, Cerrahpaşa Faculty of Medicine, Istanbul, Turkey
| |
Collapse
|
18
|
Agrawal A, Bajaj S, Bhagat U, Chandna S, Arockiam AD, El Dahdah J, Haroun E, Gupta R, Shekhar S, Raj K, Nayar D, Bajaj D, Chaudhury P, Griffin BP, Wang TKM. Incidence, Predictors, and Outcomes of Venous and Arterial Thrombosis in COVID-19: A Nationwide Inpatient Analysis. Heart Lung Circ 2024; 33:1563-1573. [PMID: 38942623 DOI: 10.1016/j.hlc.2024.04.167] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2023] [Revised: 03/30/2024] [Accepted: 04/08/2024] [Indexed: 06/30/2024]
Abstract
BACKGROUND Coronavirus disease 2019 (COVID-19) is known to increase the risk of venous thromboembolism (VTE) and arterial thromboembolism (ATE). However, the incidence, predictors, and outcomes of clinical thrombosis for inpatients with COVID-19 are not well known. This study aimed to enhance our understanding of clinical thrombosis in COVID-19, its associated factors, and mortality outcomes. METHOD Hospitalised adult (≥18 years of age) patients with COVID-19 in 2020 were retrospectively identified from the US National Inpatient Sample database. Clinical characteristics, incident VTE, ATE, and in-hospital mortality outcomes were recorded. Multivariable logistic regression was performed to identify clinical factors associated with thrombosis and in-hospital mortality in COVID-19 inpatients. RESULTS A total of 1,583,135 adult patients with COVID-19 in the year 2020 were identified from the National Inpatient Sample database; patients with thrombosis were 41% females with a mean age of 65.4 (65.1-65.6) years. The incidence of thrombosis was 6.1% (97,185), including VTE at 4.8% (76,125), ATE at 3.0% (47,790), and the in-hospital mortality rate was 13.4% (212,785). Patients with thrombosis were more likely to have respiratory symptoms of COVID-19 (76.7% vs 75%, p<0.001) compared with patients without thrombosis. The main factors associated with overall thrombosis, VTE, and ATE were paralysis, ventilation, solid tumours without metastasis, metastatic cancer, and acute liver failure. Although all thrombosis categories were associated with higher in-hospital mortality for COVID-19 inpatients in univariable analyses (p<0.001), they were not in multivariable analyses-thrombosis (odds ratio [OR] 1.24; 95% confidence interval [CI] 0.90-1.70; p=0.19), VTE (OR 0.70; 95% CI 0.52-1.00; p=0.05), and ATE (OR 1.07; 95% CI 0.92-1.25; p=0.36). CONCLUSIONS The association of COVID-19 with thrombosis and VTE increases with increasing severity of the COVID-19 disease. Risk stratification of thrombosis is crucial in COVID-19 patients to determine the necessity of thromboprophylaxis.
Collapse
Affiliation(s)
- Ankit Agrawal
- Department of Cardiovascular Medicine, Heart, Vascular, and Thoracic Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Suryansh Bajaj
- Department of Radiology, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Umesh Bhagat
- Department of Hospital Medicine, Cleveland Clinic, Cleveland, OH, USA
| | - Sanya Chandna
- Department of Hospital Medicine, Cleveland Clinic, Cleveland, OH, USA
| | - Aro Daniela Arockiam
- Department of Cardiovascular Medicine, Heart, Vascular, and Thoracic Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Joseph El Dahdah
- Department of Cardiovascular Medicine, Heart, Vascular, and Thoracic Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Elio Haroun
- Department of Cardiovascular Medicine, Heart, Vascular, and Thoracic Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Rahul Gupta
- Lehigh Valley Heart Institute, Lehigh Valley Health Network, Allentown, PA, USA
| | - Shashank Shekhar
- Department of Cardiovascular Medicine, Heart, Vascular, and Thoracic Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Kavin Raj
- Division of Cardiology, Department of Medicine, University of California Riverside School of Medicine, Riverside, CA, USA
| | - Divya Nayar
- Department of Neurology, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Divyansh Bajaj
- Department of Pulmonary, Critical Care, and Sleep Medicine, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Pulkit Chaudhury
- Department of Cardiovascular Medicine, Heart, Vascular, and Thoracic Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Brian P Griffin
- Department of Cardiovascular Medicine, Heart, Vascular, and Thoracic Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Tom Kai Ming Wang
- Department of Cardiovascular Medicine, Heart, Vascular, and Thoracic Institute, Cleveland Clinic, Cleveland, OH, USA.
| |
Collapse
|
19
|
Almskog LM, Sjöström A, Sundén-Cullberg J, Taxiarchis A, Ågren A, Freyland S, Börjesson M, Wikman A, Wahlgren CM, Wanecek M, van der Linden J, Antovic J, Lampa J, Magnusson M. Tocilizumab reduces hypercoagulation in COVID-19 - Perspectives from the coagulation and immunomodulation Covid assessment (Coag-ImmCovA) clinical trial. Thromb Res 2024; 243:109135. [PMID: 39226747 DOI: 10.1016/j.thromres.2024.109135] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2024] [Revised: 08/25/2024] [Accepted: 08/27/2024] [Indexed: 09/05/2024]
Abstract
BACKGROUND Despite medical interventions, COVID-19 continues to persist at pandemic proportions. A hypercoagulation state was rapidly observed in the severely ill, and the incidence of thromboembolic events remains elevated. Interleukin inhibitors have demonstrated positive effects on the hyperactivation of the immune system in COVID-19, with the interleukin-6 inhibitor tocilizumab showing promising results in reducing mortality. Nevertheless, the impact of interleukin inhibitors on the coagulation system remains incompletely understood. METHODS In this clinical trial conducted in Stockholm, Sweden, interleukin inhibitors, namely anakinra (ANA) or tocilizumab (TOCI), were randomly administered in addition to standard care (SC) to hospitalized patients with COVID-19. A control group received only SC. The primary outcome sought to measure effects on global hemostasis, as indicated by changes in functional coagulation tests, specifically Rotational Thromboelastometry (ROTEM) or Overall Hemostatic Potential (OHP), visualized through scanning electron microscopy images. Secondary outcomes included effects on conventional coagulation laboratory tests. RESULTS The study enrolled 74 patients who were randomized to receive either ANA or TOCI in addition to SC, or SC alone. In the TOCI group, ROTEM variables exhibited less hypercoagulation after 29 days compared with ANA or SC treatment groups, characterized by prolonged clot formation time and decreased clot firmness. OHP decreased, but there were no significant differences among the three treatment groups. Plasma fibrinogen levels, initially elevated, decreased significantly in TOCI recipients over time. CONCLUSION Tocilizumab treatment demonstrated a significant reduction of hypercoagulation in hospitalized COVID-19 patients, by improvements in both global coagulation tests and conventional laboratory tests, in comparison with anakinra or SC alone. This finding underscores the significance of tocilizumab as a viable treatment option in severe COVID-19 cases, with the potential to decrease thrombosis incidence.
Collapse
Affiliation(s)
- Lou M Almskog
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden; Department of Perioperative Medicine and Intensive Care, Karolinska University Hospital, Stockholm, Sweden.
| | - Anna Sjöström
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden; Department of Clinical Chemistry, Karolinska University Hospital, Stockholm, Sweden
| | - Jonas Sundén-Cullberg
- Department of Infectious Diseases, Karolinska University Hospital, Stockholm, Sweden; Division of Infectious Diseases, Department of Medicine Huddinge, Karolinska Institute, Stockholm, Sweden
| | - Apostolos Taxiarchis
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden; Department of Clinical Chemistry, Karolinska University Hospital, Stockholm, Sweden
| | - Anna Ågren
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden; Department of Clinical Sciences, Danderyd Hospital, Division of Cardiovascular Medicine, Karolinska Institutet, Stockholm, Sweden; Coagulation Unit, Department of Hematology, Karolinska University Hospital, Stockholm, Sweden
| | - Sara Freyland
- Division of Biostatistics, Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Madeleine Börjesson
- Department of Perioperative Medicine and Intensive Care, Karolinska University Hospital, Stockholm, Sweden
| | - Agneta Wikman
- Department of Clinical Immunology and Transfusion Medicine, Karolinska University Hospital, Stockholm, Sweden; Department of Center for Hematology and Regenerative Medicine (HERM), Karolinska Institutet, Stockholm, Sweden
| | - Carl Magnus Wahlgren
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden; Department of Vascular Surgery, Karolinska University Hospital, Stockholm, Sweden
| | - Michael Wanecek
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden; Intensive Care Unit, Capio St Göran's Hospital, Stockholm, Sweden
| | - Jan van der Linden
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | - Jovan Antovic
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden; Department of Clinical Chemistry, Karolinska University Hospital, Stockholm, Sweden
| | - Jon Lampa
- Rheumatology Division, Department of Medicine Solna, Center for Molecular Medicine (CMM), Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Maria Magnusson
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden; Coagulation Unit, Department of Hematology, Karolinska University Hospital, Stockholm, Sweden; Department of Clinical Science, Intervention and Technology (CLINTEC), Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
20
|
McElroy S, Cramer E, Amos L. COVID-19 venous thromboembolism prophylaxis guidelines in pediatrics. Thromb Res 2024; 243:109169. [PMID: 39366112 DOI: 10.1016/j.thromres.2024.109169] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2024] [Revised: 09/17/2024] [Accepted: 09/19/2024] [Indexed: 10/06/2024]
Abstract
INTRODUCTION SARS-CoV-2 (COVID-19) infection and multisystem inflammatory syndrome in children (MIS-C) are risk factors for venous thromboembolism (VTE). Guidelines for VTE prophylaxis were established at our institution at the beginning of the pandemic. Patients who had any VTE risk factors in addition to COVID-19 met criteria for anticoagulation prophylaxis. Patients who were diagnosed with MIS-C met criteria regardless of additional risk factors. MATERIALS AND METHODS We conducted a retrospective review of patients admitted with COVID-19 or MIS-C to determine compliance with VTE prophylaxis guidelines and to evaluate the incidence of VTE and bleeding events in our population. RESULTS AND CONCLUSIONS Among a total of 678 patients admitted with COVID-19 or MIS-C, 519 (76 %) patients met criteria for VTE prophylaxis and 348 (65.6 %) started prophylaxis. Logistic regression analysis identified a personal or family history of thrombosis or thrombophilia, diagnosis of MIS-C, admission to the intensive care unit, and presence of a central venous catheter as significantly associated with starting VTE prophylaxis. There were 18 patients who developed VTE. Minor bleeding events occurred in 19 patients (5 %), patient important bleeding, no intervention occurred in 8 patients (2 %), clinically relevant nonmajor bleeding in 8 patients (2 %), and major bleeding in 10 patients (3 %). The incidence of VTE in our patients with COVID-19 and MIS-C is similar to VTE rates at other institutions. We found that universally recognized VTE risk factors were appropriate to include as risk factors for thrombosis in hospitalized children with COVID-19 and MIS-C.
Collapse
Affiliation(s)
- Sara McElroy
- Pediatric Hematology and Oncology, Children's Mercy Hospital, University of Missouri-Kansas City School of Medicine, 2401 Gillham Rd., Kansas City, MO 64108, United States of America.
| | - Emily Cramer
- Health Services and Outcomes Research, Children's Mercy Hospital, University of Missouri-Kansas City School of Medicine, 2401 Gillham Rd., Kansas City, MO 64108, United States of America.
| | - Lauren Amos
- Pediatric Hematology and Oncology, Children's Mercy Hospital, University of Missouri-Kansas City School of Medicine, 2401 Gillham Rd., Kansas City, MO 64108, United States of America.
| |
Collapse
|
21
|
Zhou Y, Yang Y, Wang Y, Hou D, Song Y. Anticoagulant therapy in adult with COVID-19: a systematic review and meta-analysis of randomized controlled trial. J Thorac Dis 2024; 16:6391-6405. [PMID: 39552875 PMCID: PMC11565368 DOI: 10.21037/jtd-24-744] [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: 05/07/2024] [Accepted: 09/06/2024] [Indexed: 11/19/2024]
Abstract
Background Blood coagulation dysfunction is a risk factor for adverse outcomes in patients with coronavirus disease 2019 (COVID-19), especially in severe cases. The evidence for the effects of anticoagulation therapy on prognosis of COVID-19 patients and its risk of causing bleeding events is accumulating. Here we conducted a meta-analysis to assess the efficacy and safety of anticoagulants in COVID-19 patients of different severity. Methods We searched PubMed, Embase databases, Cochrane Trials, OVID MEDLINE from December 2019 to April 2023. We included randomized controlled trials (RCTs) involving COVID-19 patients over 18 years of age, which explored the effect of anticoagulant and its dose on outcomes including all-cause mortality, bleeding events or thrombotic events. We calculated the risk ratio (RR) and its 95% confidence interval (CI) for each outcome. We also performed subgroup analyses to assess the impact of disease severity, using a fixed-effect model to test for heterogeneity. The risk of bias, publication bias, and the quality of evidence were also evaluated. Results A total of 20 RCTs were included for final analysis. When compared with standard care, anticoagulation treatment reduced all-cause mortality (RR 0.47, 95% CI: 0.29-0.76) and thrombotic events (RR 0.35, 95% CI: 0.15-0.83) in the whole population with COVID-19 (n=2,365), without increase in bleeding events (total: RR 1.47, 95% CI: 0.54-4.00). Most of the studies only enrolled non-severe patients (n=2,329), while the number of severe patients (n=36) was scarce. In RCTs compared therapeutical and prophylactic doses of anticoagulants, no significant difference in on all-cause mortality was found in the whole population and non-severe and severe subgroups (total: RR 1.01, 95% CI: 0.92-1.10; non-severe: RR 1.03, 95% CI: 0.81-1.32; severe: RR 1.00, 95% CI: 0.91-1.11). Therapeutical dose reduced risk of thrombotic events (total: RR 0.59, 95% CI: 0.48-0.73; subtotal of non-severe: RR 0.57, 95% CI: 0.39-0.84; Subtotal of severe: RR 0.61, 95% CI: 0.47-0.78), while risk of bleeding was increased (total: RR 1.98, 95% CI: 1.47-2.66; non-severe: RR 2.38, 95% CI: 1.56-3.62; severe: RR 1.63, 95% CI: 1.07-2.47). Study heterogeneity was found only in the analysis of effects of anticoagulants on risk of thrombotic events. Conclusions Anticoagulant therapy reduces all-cause mortality and risk of thrombosis in non-severe COVID-19 patients. Therapeutic dose of anticoagulant therapy can be considered in both non-severe and severe COVID-19 patients to reduce thrombosis, but may be associated with increased bleeding events.
Collapse
Affiliation(s)
- Yanan Zhou
- Shanghai Institute of Infectious Disease and Biosecurity, Fudan University, Shanghai, China
| | - Yanping Yang
- Shanghai Institute of Infectious Disease and Biosecurity, Fudan University, Shanghai, China
| | - Ying Wang
- Department of Critical Care Medicine, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Dongni Hou
- Shanghai Key Laboratory of Lung Inflammation and Injury, Department of Pulmonary Medicine, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Yuanlin Song
- Shanghai Institute of Infectious Disease and Biosecurity, Fudan University, Shanghai, China
- Shanghai Key Laboratory of Lung Inflammation and Injury, Department of Pulmonary Medicine, Zhongshan Hospital, Fudan University, Shanghai, China
- Shanghai Respiratory Research Institute, Shanghai, China
- National and Shanghai Clinical Research Center for Aging and Medicine, Huashan Hospital, Fudan University, Shanghai, China
- Key Laboratory of Chemical Injury, Emergency and Critical Medicine of Shanghai Municipal Health Commission, Center of Emergency and Critical Medicine, Jinshan Hospital of Fudan University, Shanghai, China
| |
Collapse
|
22
|
Maurizot A, Chabay S, Roger G, Tapiero S, Georges JL, Flaujac C, Paul M, Roche A, Bruneel F, Ferré A. Incidence of deep venous thrombosis in COVID-19 critically ill patients treated with intermediate-dose of heparin for thromboprophylaxis: The COVIDOP-DVT observational study. Vascular 2024; 32:1099-1106. [PMID: 37009990 PMCID: PMC10071186 DOI: 10.1177/17085381231165083] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/04/2023]
Abstract
INTRODUCTION The high prevalence of deep vein thrombosis (DVT) in patients admitted to intensive care unit (ICU) for COVID-19-related acute respiratory distress syndrome (ARDS) would justify systematic screening of these patients or higher therapeutic dose of heparin for thromboprophylaxis. MATERIAL AND METHOD We performed a systematic echo-Doppler of the lower limb proximal veins during the first 48 h (visit 1) and from 7 to 9 days after visit 1 (visit 2) in consecutive patients admitted to the ICU of a university-affiliated tertiary hospital for severe proven COVID-19 during the second wave. All patients received intermediate-dose heparin (IDH). The primary objective was to determine DVT incidence on venous Doppler ultrasound. Secondary objectives were to determine whether the presence of DVT modifies the anticoagulation regimen, the incidence of major bleeding according to International Society on Thrombosis and Haemostasis (ISTH) criteria, and the mortality rate of patients with and without DVT. RESULTS We included 48 patients (30 [62.5%] men) with a median age of 63 years [IQR, 54-70]. The prevalence of proximal deep vein thrombosis was 4.2% (2/48). In these two patients, after DVT diagnosis, anticoagulation was changed from intermediate to curative dose. Two patients (4.2%) had a major bleeding complication according to ISTH criteria. Among the 48 patients, 9 (18.8%) died before hospital discharge. No DVT or pulmonary embolism was diagnosed in these deceased patients during their hospital stay. CONCLUSION In critically ill patients with COVID-19, management with IDH results in a low incidence of DVT. Although our study is not designed to demonstrate any difference in outcome, our results do not suggest any signal of harm when using intermediate-dose heparin (IDH) COVID-19 with a frequency of major bleeding complications less than 5%.
Collapse
Affiliation(s)
- Aurélien Maurizot
- Vascular Medicine Unit, Cardiology Department, Versailles Hospital, Le Chesnay, France
| | - Simon Chabay
- Vascular Medicine Unit, Cardiology Department, Versailles Hospital, Le Chesnay, France
| | - Guillaume Roger
- Vascular Medicine Unit, Cardiology Department, Versailles Hospital, Le Chesnay, France
| | - Stéphanie Tapiero
- Vascular Medicine Unit, Cardiology Department, Versailles Hospital, Le Chesnay, France
| | | | - Claire Flaujac
- Haemostasis Unit, Medical Biology Department, Versailles Hospital, Le Chesnay, France
| | - Marine Paul
- Intensive Care Unit, Versailles Hospital, Le Chesnay, France
| | - Anne Roche
- Intensive Care Unit, Versailles Hospital, Le Chesnay, France
| | - Fabrice Bruneel
- Intensive Care Unit, Versailles Hospital, Le Chesnay, France
| | - Alexis Ferré
- Intensive Care Unit, Versailles Hospital, Le Chesnay, France
| |
Collapse
|
23
|
Iqbal K, Banga A, Arif TB, Rathore SS, Bhurwal A, Naqvi SKB, Mehdi M, Kumar P, Salklan MM, Iqbal A, Ahmed J, Sharma N, Lal A, Kashyap R, Bansal V, Domecq JP. Anticoagulant use before COVID-19 diagnosis prevent COVID-19 associated acute venous thromboembolism or not: A systematic review and meta-analysis. World J Methodol 2024; 14:92983. [PMID: 39310244 PMCID: PMC11230074 DOI: 10.5662/wjm.v14.i3.92983] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/15/2024] [Revised: 04/24/2024] [Accepted: 05/11/2024] [Indexed: 06/25/2024] Open
Abstract
BACKGROUND Coagulopathy and thromboembolic events are associated with poor outcomes in coronavirus disease 2019 (COVID-19) patients. There is conflicting evidence on the effects of chronic anticoagulation on mortality and severity of COVID-19 disease. AIM To summarize the body of evidence on the effects of pre-hospital anticoagulation on outcomes in COVID-19 patients. METHODS A Literature search was performed on LitCovid PubMed, WHO, and Scopus databases from inception (December 2019) till June 2023 for original studies reporting an association between prior use of anticoagulants and patient outcomes in adults with COVID-19. The primary outcome was the risk of thromboembolic events in COVID-19 patients taking anticoagulants. Secondary outcomes included COVID-19 disease severity, in terms of intensive care unit admission or invasive mechanical ventilation/intubation requirement in patients hospitalized with COVID-19 infection, and mortality. The random effects models were used to calculate crude and adjusted odds ratios (aORs) with 95% confidence intervals (95%CIs). RESULTS Forty-six observational studies met our inclusion criteria. The unadjusted analysis found no association between prior anticoagulation and thromboembolic event risk [n = 43851, 9 studies, odds ratio (OR)= 0.67 (0.22, 2.07); P = 0.49; I 2 = 95%]. The association between prior anticoagulation and disease severity was non-significant [n = 186782; 22 studies, OR = 1.08 (0.78, 1.49); P = 0.64; I 2 = 89%]. However, pre-hospital anticoagulation significantly increased all-cause mortality risk [n = 207292; 35 studies, OR = 1.72 (1.37, 2.17); P < 0.00001; I 2 = 93%]. Pooling adjusted estimates revealed a statistically non-significant association between pre-hospital anticoagulation and thromboembolic event risk [aOR = 0.87 (0.42, 1.80); P = 0.71], mortality [aOR = 0.94 (0.84, 1.05); P = 0.31], and disease severity [aOR = 0.96 (0.72, 1.26); P = 0.76]. CONCLUSION Prehospital anticoagulation was not significantly associated with reduced risk of thromboembolic events, improved survival, and lower disease severity in COVID-19 patients.
Collapse
Affiliation(s)
- Kinza Iqbal
- Department of Internal Medicine, Dow Medical College, Karachi 74200, Pakistan
| | - Akshat Banga
- Department of Internal Medicine, Sawai Man Singh Medical College, Jaipur 302004, India
| | - Taha Bin Arif
- Department of Internal Medicine, Dow Medical College, Karachi 74200, Pakistan
| | - Sawai Singh Rathore
- Department of Internal Medicine, Dr. Sampurnanand Medical College, Jodhpur 342003, Rajasthan, India
| | - Abhishek Bhurwal
- Department of Gastroenterology and Hepatology, Rutgers Robert Wood Johnson School of Medicine, New Brunswick, NJ 08901, United States
| | | | - Muhammad Mehdi
- Department of Internal Medicine, Dow Medical College, Karachi 74200, Pakistan
| | - Pankaj Kumar
- Department of Internal Medicine, Dow Medical College, Karachi 74200, Pakistan
| | - Mitali Madhu Salklan
- Department of Internal Medicine, Pandit Bhagwat Dayal Sharma Post Graduate Institute of Medical Sciences, Rohtak 124001, Haryana, India
| | - Ayman Iqbal
- Department of Internal Medicine, Dow Medical College, Karachi 74200, Pakistan
| | - Jawad Ahmed
- Department of Internal Medicine, Dow Medical College, Karachi 74200, Pakistan
| | - Nikhil Sharma
- Department of Nephrology and Hypertension, Mayo Clinic, Rochester, MN 55905, United States
| | - Amos Lal
- Department of Medicine, Division of Pulmonary and Critical Care Medicine, Mayo Clinic, Rochester, MN 55905, United States
| | - Rahul Kashyap
- Department of Research, Wellspan Health, York, PA 17403, United States
| | - Vikas Bansal
- Department of Nephrology and Hypertension, Mayo Clinic, Rochester, MN 55905, United States
| | - Juan Pablo Domecq
- Department of Nephrology and Hypertension, Mayo Clinic, Rochester, MN 55905, United States
| |
Collapse
|
24
|
Attiq A, Afzal S, Wahab HA, Ahmad W, Kandeel M, Almofti YA, Alameen AO, Wu YS. Cytokine Storm-Induced Thyroid Dysfunction in COVID-19: Insights into Pathogenesis and Therapeutic Approaches. Drug Des Devel Ther 2024; 18:4215-4240. [PMID: 39319193 PMCID: PMC11421457 DOI: 10.2147/dddt.s475005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2024] [Accepted: 08/26/2024] [Indexed: 09/26/2024] Open
Abstract
Angiotensin-converting enzyme 2 receptors (ACE2R) are requisite to enter the host cells for severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2). ACE2R is constitutive and functions as a type I transmembrane metallo-carboxypeptidase in the renin-angiotensin system (RAS). On thyroid follicular cells, ACE2R allows SARS-CoV-2 to invade the thyroid gland, impose cytopathic effects and produce endocrine abnormalities, including stiff back, neck pain, muscle ache, lethargy, and enlarged, inflamed thyroid gland in COVID-19 patients. Further damage is perpetuated by the sudden bursts of pro-inflammatory cytokines, which is suggestive of a life-threatening syndrome known as a "cytokine storm". IL-1β, IL-6, IFN-γ, and TNF-α are identified as the key orchestrators of the cytokine storm. These inflammatory mediators upregulate transcriptional turnover of nuclear factor-kappa B (NF-κB), Janus kinase/signal transducer and activator of transcription (JAK/STAT), and mitogen-activated protein kinase (MAPK), paving the pathway for cytokine storm-induced thyroid dysfunctions including euthyroid sick syndrome, autoimmune thyroid diseases, and thyrotoxicosis in COVID-19 patients. Targeted therapies with corticosteroids (dexamethasone), JAK inhibitor (baricitinib), nucleotide analogue (remdesivir) and N-acetyl-cysteine have demonstrated effectiveness in terms of attenuating the severity and frequency of cytokine storm-induced thyroid dysfunctions, morbidity and mortality in severe COVID-19 patients. Here, we review the pathogenesis of cytokine storms and the mechanisms and pathways that establish the connection between thyroid disorder and COVID-19. Moreover, cross-talk interactions of signalling pathways and therapeutic strategies to address COVID-19-associated thyroid diseases are also discussed herein.
Collapse
Affiliation(s)
- Ali Attiq
- School of Pharmaceutical Sciences, Universiti Sains Malaysia, Gelugor, Penang, 11800, Malaysia
| | - Sheryar Afzal
- Department of Biomedical Sciences, College of Veterinary Medicine, King Faisal University, Al Ahsa, 31982, Saudi Arabia
| | - Habibah A Wahab
- School of Pharmaceutical Sciences, Universiti Sains Malaysia, Gelugor, Penang, 11800, Malaysia
| | - Waqas Ahmad
- School of Pharmaceutical Sciences, Universiti Sains Malaysia, Gelugor, Penang, 11800, Malaysia
| | - Mahmoud Kandeel
- Department of Biomedical Sciences, College of Veterinary Medicine, King Faisal University, Al Ahsa, 31982, Saudi Arabia
- Department of Pharmacology, Faculty of Veterinary Medicine, Kafrelsheikh University, Kafrel Sheikh, 6860404, Egypt
| | - Yassir A Almofti
- Department of Biomedical Sciences, College of Veterinary Medicine, King Faisal University, Al Ahsa, 31982, Saudi Arabia
- Department of Biochemistry, Molecular Biology and Bioinformatics, College of Veterinary Medicine, University of Bahri, Khartoum, 12217, Sudan
| | - Ahmed O Alameen
- Department of Biomedical Sciences, College of Veterinary Medicine, King Faisal University, Al Ahsa, 31982, Saudi Arabia
- Department of Physiology, Faculty of Veterinary Medicine, University of Khartoum, Shambat, 13314, Sudan
| | - Yuan Seng Wu
- Sunway Microbiome Centre, School of Medical and Life Sciences, Sunway University, Subang Jaya, Selangor, 47500, Malaysia
- Department of Biological Sciences, School of Medical and Life Sciences, Sunway University, Subang Jaya, Selangor, 47500, Malaysia
| |
Collapse
|
25
|
Tran HNQ, Risk M, Nair GB, Zhao L. Risk benefit analysis to evaluate risk of thromboembolic events after mRNA COVID-19 vaccination and COVID-19. NPJ Vaccines 2024; 9:166. [PMID: 39271677 PMCID: PMC11399239 DOI: 10.1038/s41541-024-00960-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2024] [Accepted: 08/29/2024] [Indexed: 09/15/2024] Open
Abstract
We compared the risks and benefits of COVID-19 vaccines using a causal pathway analysis to weigh up possible risk factors of thromboembolic events post-vaccination. The self-controlled case series (SCCS) method examined the association between thromboembolic events and vaccination while a case-control study assessed the association between thromboembolic events and COVID-19, addressing under-reported infection data issues. The net vaccine effect was estimated using results from SCCS and case-control studies. We used electronic health record data from Corewell Health (16,640 subjects in SCCS and 106,143 in case-control). We found increased risks of thromboembolic events post-vaccination (incidence rate ratio: 1.19, 95% CI: [1.08, 1.31] after the first dose; 1.22, 95% CI: [1.11, 1.34] after the second dose). Vaccination attenuated infection-associated thromboembolic risks (odds ratio: 4.65, 95% CI: [4.18, 5.17] in unvaccinated vs 2.77, 95% CI: [2.40, 3.24] in vaccinated). After accounting for vaccine efficacy and protection against infection-associated thromboembolic events, vaccination decreases thromboembolic event risk, especially during high infection rate periods.
Collapse
Affiliation(s)
- Huong N Q Tran
- Division of Biostatistics & Health Informatics, Corewell Health Research Institute, Royal Oak, MI, USA
| | - Malcolm Risk
- Department of Biostatistics, University of Michigan, Ann Arbor, MI, USA
| | - Girish B Nair
- William Beaumont University Hospital, Corewell Health East, Royal Oak, MI, USA
| | - Lili Zhao
- Division of Biostatistics, Department of Preventive Medicine, Northwestern University, Chicago, IL, USA.
| |
Collapse
|
26
|
Stark K, Kilani B, Stockhausen S, Busse J, Schubert I, Tran TD, Gaertner F, Leunig A, Pekayvaz K, Nicolai L, Fumagalli V, Stermann J, Stephan F, David C, Müller MB, Heyman B, Lux A, da Palma Guerreiro A, Frenzel LP, Schmidt CQ, Dopler A, Moser M, Chandraratne S, von Brühl ML, Lorenz M, Korff T, Rudelius M, Popp O, Kirchner M, Mertins P, Nimmerjahn F, Iannacone M, Sperandio M, Engelmann B, Verschoor A, Massberg S. Antibodies and complement are key drivers of thrombosis. Immunity 2024; 57:2140-2156.e10. [PMID: 39226900 DOI: 10.1016/j.immuni.2024.08.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Revised: 05/17/2024] [Accepted: 08/07/2024] [Indexed: 09/05/2024]
Abstract
Venous thromboembolism (VTE) is a common, deadly disease with an increasing incidence despite preventive efforts. Clinical observations have associated elevated antibody concentrations or antibody-based therapies with thrombotic events. However, how antibodies contribute to thrombosis is unknown. Here, we show that reduced blood flow enabled immunoglobulin M (IgM) to bind to FcμR and the polymeric immunoglobulin receptor (pIgR), initiating endothelial activation and platelet recruitment. Subsequently, the procoagulant surface of activated platelets accommodated antigen- and FcγR-independent IgG deposition. This leads to classical complement activation, setting in motion a prothrombotic vicious circle. Key elements of this mechanism were present in humans in the setting of venous stasis as well as in the dysregulated immunothrombosis of COVID-19. This antibody-driven thrombosis can be prevented by pharmacologically targeting complement. Hence, our results uncover antibodies as previously unrecognized central regulators of thrombosis. These findings carry relevance for therapeutic application of antibodies and open innovative avenues to target thrombosis without compromising hemostasis.
Collapse
Affiliation(s)
- Konstantin Stark
- Medizinische Klinik und Poliklinik I, University Hospital, LMU Munich, Munich, Germany; German Center for Cardiovascular Research (DZHK), partner site Munich Heart Alliance, Munich, Germany; Walter-Brendel Center of Experimental Medicine, Faculty of Medicine, LMU Munich, Munich, Germany.
| | - Badr Kilani
- Medizinische Klinik und Poliklinik I, University Hospital, LMU Munich, Munich, Germany; German Center for Cardiovascular Research (DZHK), partner site Munich Heart Alliance, Munich, Germany; Walter-Brendel Center of Experimental Medicine, Faculty of Medicine, LMU Munich, Munich, Germany
| | - Sven Stockhausen
- Medizinische Klinik und Poliklinik I, University Hospital, LMU Munich, Munich, Germany; German Center for Cardiovascular Research (DZHK), partner site Munich Heart Alliance, Munich, Germany; Walter-Brendel Center of Experimental Medicine, Faculty of Medicine, LMU Munich, Munich, Germany
| | - Johanna Busse
- Medizinische Klinik und Poliklinik I, University Hospital, LMU Munich, Munich, Germany; Walter-Brendel Center of Experimental Medicine, Faculty of Medicine, LMU Munich, Munich, Germany
| | - Irene Schubert
- Medizinische Klinik und Poliklinik I, University Hospital, LMU Munich, Munich, Germany; Walter-Brendel Center of Experimental Medicine, Faculty of Medicine, LMU Munich, Munich, Germany
| | - Thuy-Duong Tran
- Medizinische Klinik und Poliklinik I, University Hospital, LMU Munich, Munich, Germany; Walter-Brendel Center of Experimental Medicine, Faculty of Medicine, LMU Munich, Munich, Germany
| | - Florian Gaertner
- Medizinische Klinik und Poliklinik I, University Hospital, LMU Munich, Munich, Germany; German Center for Cardiovascular Research (DZHK), partner site Munich Heart Alliance, Munich, Germany; Walter-Brendel Center of Experimental Medicine, Faculty of Medicine, LMU Munich, Munich, Germany; Institute of Science and Technology Austria, Klosterneuburg, Austria
| | - Alexander Leunig
- Medizinische Klinik und Poliklinik I, University Hospital, LMU Munich, Munich, Germany; German Center for Cardiovascular Research (DZHK), partner site Munich Heart Alliance, Munich, Germany; Walter-Brendel Center of Experimental Medicine, Faculty of Medicine, LMU Munich, Munich, Germany
| | - Kami Pekayvaz
- Medizinische Klinik und Poliklinik I, University Hospital, LMU Munich, Munich, Germany; German Center for Cardiovascular Research (DZHK), partner site Munich Heart Alliance, Munich, Germany; Walter-Brendel Center of Experimental Medicine, Faculty of Medicine, LMU Munich, Munich, Germany
| | - Leo Nicolai
- Medizinische Klinik und Poliklinik I, University Hospital, LMU Munich, Munich, Germany; German Center for Cardiovascular Research (DZHK), partner site Munich Heart Alliance, Munich, Germany; Walter-Brendel Center of Experimental Medicine, Faculty of Medicine, LMU Munich, Munich, Germany
| | - Valeria Fumagalli
- Division of Immunology, Transplantation, and Infectious Diseases, IRCCS San Raffaele Scientific Institute, Milan, Italy; Vita-Salute San Raffaele University, Milan, Italy
| | - Julia Stermann
- Medizinische Klinik und Poliklinik I, University Hospital, LMU Munich, Munich, Germany; German Center for Cardiovascular Research (DZHK), partner site Munich Heart Alliance, Munich, Germany; Walter-Brendel Center of Experimental Medicine, Faculty of Medicine, LMU Munich, Munich, Germany
| | - Felix Stephan
- Medizinische Klinik und Poliklinik I, University Hospital, LMU Munich, Munich, Germany; German Center for Cardiovascular Research (DZHK), partner site Munich Heart Alliance, Munich, Germany; Walter-Brendel Center of Experimental Medicine, Faculty of Medicine, LMU Munich, Munich, Germany
| | - Christian David
- Institute for Cardiovascular Physiology and Pathophysiology, Walter Brendel Center for Experimental Medicine, Biomedical Center (BMC) LMU Munich, Munich, Germany
| | - Martin B Müller
- Walter-Brendel Center of Experimental Medicine, Faculty of Medicine, LMU Munich, Munich, Germany; Department of Anaesthesiology, University Hospital, LMU Munich, Munich, Germany
| | - Birgitta Heyman
- Department of Medical Biochemistry and Microbiology, Uppsala University, Uppsala, Sweden
| | - Anja Lux
- Department of Biology, Institute of Genetics, Friedrich-Alexander University Erlangen-Nürnberg, Erlangen, Germany; Medical Immunology Campus Erlangen (MICE), Friedrich-Alexander University Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Alexandra da Palma Guerreiro
- Department I of Internal Medicine, University Hospital Cologne, Cologne 50937, Germany; Center of Integrated Oncology ABCD, University Hospital of Cologne, Cologne, Germany; Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, Cologne 50937, Germany
| | - Lukas P Frenzel
- Department I of Internal Medicine, University Hospital Cologne, Cologne 50937, Germany; Center of Integrated Oncology ABCD, University Hospital of Cologne, Cologne, Germany; Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, Cologne 50937, Germany
| | - Christoph Q Schmidt
- Institute of Experimental and Clinical Pharmacology, Toxicology and Pharmacology of Natural Products, University of Ulm Medical Center, Ulm, Germany
| | - Arthur Dopler
- Institute of Experimental and Clinical Pharmacology, Toxicology and Pharmacology of Natural Products, University of Ulm Medical Center, Ulm, Germany
| | - Markus Moser
- Department of Molecular Medicine, Max-Planck-Institute of Biochemistry, Martinsried, Germany; Institute of Experimental Hematology, TranslaTUM, Klinikum rechts der Isar der Technischen Universität München, Munich, Germany
| | - Sue Chandraratne
- Medizinische Klinik und Poliklinik I, University Hospital, LMU Munich, Munich, Germany; Walter-Brendel Center of Experimental Medicine, Faculty of Medicine, LMU Munich, Munich, Germany
| | - Marie-Luise von Brühl
- Medizinische Klinik und Poliklinik I, University Hospital, LMU Munich, Munich, Germany; Walter-Brendel Center of Experimental Medicine, Faculty of Medicine, LMU Munich, Munich, Germany
| | - Michael Lorenz
- Medizinische Klinik und Poliklinik I, University Hospital, LMU Munich, Munich, Germany; Walter-Brendel Center of Experimental Medicine, Faculty of Medicine, LMU Munich, Munich, Germany
| | - Thomas Korff
- Division of Cardiovascular Physiology, Institute of Physiology and Pathophysiology, Heidelberg University, Heidelberg, Germany
| | - Martina Rudelius
- Institute of Pathology, Ludwig-Maximilian University, Munich, Germany
| | - Oliver Popp
- Max Delbrück Center for Molecular Medicine (MDC) and Berlin Institute of Health (BIH), Berlin, Germany; German Center for Cardiovascular Research (DZHK), partner site Berlin, Berlin, Germany
| | - Marieluise Kirchner
- Max Delbrück Center for Molecular Medicine (MDC) and Berlin Institute of Health (BIH), Berlin, Germany; German Center for Cardiovascular Research (DZHK), partner site Berlin, Berlin, Germany
| | - Philipp Mertins
- Max Delbrück Center for Molecular Medicine (MDC) and Berlin Institute of Health (BIH), Berlin, Germany; German Center for Cardiovascular Research (DZHK), partner site Berlin, Berlin, Germany
| | - Falk Nimmerjahn
- Department of Biology, Institute of Genetics, Friedrich-Alexander University Erlangen-Nürnberg, Erlangen, Germany; Medical Immunology Campus Erlangen (MICE), Friedrich-Alexander University Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Matteo Iannacone
- Division of Immunology, Transplantation, and Infectious Diseases, IRCCS San Raffaele Scientific Institute, Milan, Italy; Vita-Salute San Raffaele University, Milan, Italy
| | - Markus Sperandio
- Institute for Cardiovascular Physiology and Pathophysiology, Walter Brendel Center for Experimental Medicine, Biomedical Center (BMC) LMU Munich, Munich, Germany
| | - Bernd Engelmann
- Institut für Laboratoriumsmedizin, University Hospital, LMU Munich, Munich, Germany
| | - Admar Verschoor
- Department of Dermatology, Allergy, and Venereology, University of Lübeck, Lübeck, Germany; Department of Otorhinolaryngology, Technische Universität München and Klinikum Rechts der Isar, Munich, Germany.
| | - Steffen Massberg
- Medizinische Klinik und Poliklinik I, University Hospital, LMU Munich, Munich, Germany; German Center for Cardiovascular Research (DZHK), partner site Munich Heart Alliance, Munich, Germany; Walter-Brendel Center of Experimental Medicine, Faculty of Medicine, LMU Munich, Munich, Germany
| |
Collapse
|
27
|
Eustes AS, Ahmed A, Swamy J, Patil G, Jensen M, Wilson KM, Kudchadkar S, Wahab A, Perepu U, Miller FJ, Lentz SR, Dayal S. Extracellular histones: a unifying mechanism driving platelet-dependent extracellular vesicle release and thrombus formation in COVID-19. J Thromb Haemost 2024; 22:2514-2530. [PMID: 38815756 PMCID: PMC11343660 DOI: 10.1016/j.jtha.2024.05.019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2023] [Revised: 05/12/2024] [Accepted: 05/18/2024] [Indexed: 06/01/2024]
Abstract
BACKGROUND COVID-19 can cause profound inflammation and coagulopathy, and while many mechanisms have been proposed, there is no known common pathway leading to a prothrombotic state. OBJECTIVES From the beginning of the COVID-19 pandemic, elevated levels of extracellular histones have been found in plasma of patients infected with SARS-CoV-2. We hypothesized that platelet activation triggered by extracellular histones might represent a unifying mechanism leading to increased thrombin generation and thrombosis. METHODS We utilized blood samples collected from an early clinical trial of hospitalized COVID-19 patients (NCT04360824) and recruited healthy subjects as controls. Using plasma samples, we measured the procoagulant and prothrombotic potential of circulating extracellular histones and extracellular vesicles (EVs). Platelet prothrombotic activity was assessed via thrombin generation potential and platelet thrombus growth. Circulating EVs were assessed for thrombin generation potential in vitro in plasma and enhancement of thrombotic susceptibility in vivo in mice. RESULTS Compared with controls, COVID-19 patients had elevated plasma levels of citrullinated histone H3, cell-free DNA, nucleosomes, and EVs. Plasma from COVID-19 patients promoted platelet activation, platelet-dependent thrombin generation, thrombus growth under venous shear stress, and release of platelet-derived EVs. These prothrombotic effects of COVID-19 plasma were inhibited by an RNA aptamer that neutralizes both free and DNA-bound histones. EVs isolated from COVID-19 plasma enhanced thrombin generation in vitro and potentiated venous thrombosis in mice in vivo. CONCLUSION We conclude that extracellular histones and procoagulant EVs drive the prothrombotic state in COVID-19 and that histone-targeted therapy may prove beneficial.
Collapse
Affiliation(s)
- Alicia S Eustes
- Department of Internal Medicine, University of Iowa, Iowa City, Iowa, USA
| | - Azaj Ahmed
- Department of Internal Medicine, University of Iowa, Iowa City, Iowa, USA
| | - Jagadish Swamy
- Department of Internal Medicine, University of Iowa, Iowa City, Iowa, USA
| | - Gokul Patil
- Department of Internal Medicine, University of Iowa, Iowa City, Iowa, USA
| | - Melissa Jensen
- Department of Internal Medicine, University of Iowa, Iowa City, Iowa, USA
| | - Katina M Wilson
- Department of Internal Medicine, University of Iowa, Iowa City, Iowa, USA
| | - Shibani Kudchadkar
- Department of Internal Medicine, University of Iowa, Iowa City, Iowa, USA
| | - Abdul Wahab
- Department of Internal Medicine, University of Iowa, Iowa City, Iowa, USA
| | - Usha Perepu
- Department of Internal Medicine, University of Iowa, Iowa City, Iowa, USA
| | - Francis J Miller
- Department of Internal Medicine, Vanderbilt University Medical Center and VA Medical Center, Nashville, Tennessee, USA
| | - Steven R Lentz
- Department of Internal Medicine, University of Iowa, Iowa City, Iowa, USA
| | - Sanjana Dayal
- Department of Internal Medicine, University of Iowa, Iowa City, Iowa, USA; Iowa City VA Healthcare System, Iowa City, Iowa, USA.
| |
Collapse
|
28
|
van Lith TJ, Sluis WM, Wijers NT, Meijer FJA, Ulzen KKV, de Bresser J, Dankbaar JW, de Mast Q, Klok FA, Cannegieter SC, Wermer MJH, Huisman MV, Tuladhar AM, van der Worp HB, de Leeuw FE. Prevalence and 3-month follow-up of cerebrovascular MRI markers in hospitalized COVID-19 patients: the CORONIS study. Neuroradiology 2024; 66:1565-1575. [PMID: 38953988 PMCID: PMC11322373 DOI: 10.1007/s00234-024-03411-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2024] [Accepted: 06/21/2024] [Indexed: 07/04/2024]
Abstract
PURPOSE To investigate the prevalence of cerebrovascular MRI markers in unselected patients hospitalized for COVID-19 (Coronavirus disease 2019), we compared these with healthy controls without previous SARS-CoV-2 infection or hospitalization and subsequently, investigated longitudinal (incidental) lesions in patients after three months. METHODS CORONIS (CORONavirus and Ischemic Stroke) was an observational cohort study in adult hospitalized patients for COVID-19 and controls without COVID-19, conducted between April 2021 and September 2022. Brain MRI was performed shortly after discharge and after 3 months. Outcomes included recent ischemic (DWI-positive) lesions, previous infarction, microbleeds, white matter hyperintensities (WMH) and intracerebral hemorrhage and were analysed with logistic regression to adjust for confounders. RESULTS 125 patients with COVID-19 and 47 controls underwent brain MRI a median of 41.5 days after symptom onset. DWI-positive lesions were found in one patient (1%) and in one (2%) control, both clinically silent. WMH were more prevalent in patients (78%) than in controls (62%) (adjusted OR: 2.95 [95% CI: 1.07-8.57]), other cerebrovascular MRI markers did not differ. Prevalence of markers in ICU vs. non-ICU patients was similar. After three months, five patients (5%) had new cerebrovascular lesions, including DWI-positive lesions (1 patient, 1.0%), cerebral infarction (2 patients, 2.0%) and microbleeds (3 patients, 3.1%). CONCLUSION Overall, we found no higher prevalence of cerebrovascular markers in unselected hospitalized COVID-19 patients compared to controls. The few incident DWI-lesions were most likely to be explained by risk-factors of small vessel disease. In the general hospitalized COVID-19 population, COVID-19 shows limited impact on cerebrovascular MRI markers shortly after hospitalization.
Collapse
Affiliation(s)
- Theresa J van Lith
- Department of Neurology, Donders Center for Medical Neuroscience, Radboud University Medical Center, PO Box 9101, Nijmegen, 6500 HB, the Netherlands
| | - Wouter M Sluis
- Department of Neurology and Neurosurgery, Brain Center, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Naomi T Wijers
- Department of Neurology, Leiden University Medical Center, Leiden, The Netherlands
| | - Frederick J A Meijer
- Department of Medical Imaging, Radboud University Medical Center, Nijmegen, The Netherlands
| | | | - Jeroen de Bresser
- Department of Radiology, Leiden University Medical Center, Leiden, the Netherlands
| | - Jan Willem Dankbaar
- Department of Radiology and Nuclear Medicine, University Medical Center, Utrecht University, Utrecht, The Netherlands
| | - Quirijn de Mast
- Department of Internal Medicine, Radboud Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Frederikus A Klok
- Department of Medicine - Thrombosis and Hemostasis, Leiden University Medical Centre, Leiden, The Netherlands
| | - Suzanne C Cannegieter
- Department of Medicine - Thrombosis and Hemostasis, Leiden University Medical Centre, Leiden, The Netherlands
- Department of Clinical Epidemiology, Leiden University Medical Centre, Leiden, The Netherlands
| | - Marieke J H Wermer
- Department of Neurology, University Medical Center, Groningen, The Netherlands
| | - Menno V Huisman
- Department of Medicine - Thrombosis and Hemostasis, Leiden University Medical Centre, Leiden, The Netherlands
| | - Anil M Tuladhar
- Department of Neurology, Donders Center for Medical Neuroscience, Radboud University Medical Center, PO Box 9101, Nijmegen, 6500 HB, the Netherlands
| | - H Bart van der Worp
- Department of Neurology and Neurosurgery, Brain Center, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Frank-Erik de Leeuw
- Department of Neurology, Donders Center for Medical Neuroscience, Radboud University Medical Center, PO Box 9101, Nijmegen, 6500 HB, the Netherlands.
| |
Collapse
|
29
|
Li HN, Xu XN, Qin YH, Liu R, Guo WY, Huang XY, Fan ML, Zhang LJ, Qi Y, Zhang C, Yang L, Shi FD, Yang CS. Clinical features of COVID-19 infection in patients with myasthenia gravis: a real-world retrospective study. Front Public Health 2024; 12:1421211. [PMID: 39257951 PMCID: PMC11384569 DOI: 10.3389/fpubh.2024.1421211] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2024] [Accepted: 08/13/2024] [Indexed: 09/12/2024] Open
Abstract
Objective We investigated the risk factors associated with severe or critical Coronavirus disease 2019 (COVID-19) infection due to the Omicron variant in patients with myasthenia gravis (MG) and determined the potential effect of COVID-19 on myasthenic exacerbation during the Omicron pandemic. Methods This retrospective study included 287 patients with MG in Tianjin, China. Clinical data of the patients were collected using electronic questionnaires, databases, and clinical records. Results The overall infection rate was 84.7%. Advanced age, comorbidities, generalized phenotype, and MG instability were drivers of COVID-19 severity, and post-COVID-19 myasthenic exacerbation. The concurrent use of a steroid-sparing agent did not affect COVID-19 susceptibility or severity. It did lower the risk of myasthenic exacerbation after COVID-19 infection. Patients with severe COVID-19 experienced myasthenic exacerbation earlier than patients with non-severe infection (p < 0.001). The severity of COVID-19 (Hazards Ratio = 3.04, 95% CI: 1.41-6.54, p = 0.004) and the clinical phenotype (Hazards Ratio = 3.29, 95% CI: 1.63-6.63, p < 0.001) emerged as independent risk factors for early MG exacerbation. Conclusion Generally, patients with MG appear to be susceptible to the Omicron strains. Immunotherapy for MG did not increase COVID-19 susceptibility or severity. We do not advocate an immediate cessation of ongoing immunosuppressive treatments once a COVID-19 infection is diagnosed. Instead, a judicious evaluation of the risks and benefits, tailored to each individual, is recommended.
Collapse
Affiliation(s)
- Hui-Ning Li
- Department of Neurology, Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin, China
| | - Xiao-Na Xu
- Department of Neurology, Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin, China
| | - Ying-Hui Qin
- Department of Neurology, Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin, China
| | - Rui Liu
- Department of Neurology, Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin, China
| | - Wen-Yue Guo
- Department of Neurology, Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin, China
| | - Xiao-Yu Huang
- Department of Neurology, Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin, China
| | - Mo-Li Fan
- Department of Neurology, Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin, China
| | - Lin-Jie Zhang
- Department of Neurology, Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin, China
| | - Yuan Qi
- Department of Neurology, Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin, China
| | - Chao Zhang
- Department of Neurology, Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin, China
- China National Clinical Research Center for Neurological Diseases, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Li Yang
- Department of Neurology, Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin, China
- China National Clinical Research Center for Neurological Diseases, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Fu-Dong Shi
- Department of Neurology, Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin, China
- China National Clinical Research Center for Neurological Diseases, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Chun-Sheng Yang
- Department of Neurology, Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin, China
- China National Clinical Research Center for Neurological Diseases, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
30
|
Qing L, Wu W. The mechanism of geniposide in patients with COVID-19 and atherosclerosis: A pharmacological and bioinformatics analysis. Medicine (Baltimore) 2024; 103:e39065. [PMID: 39093733 PMCID: PMC11296471 DOI: 10.1097/md.0000000000039065] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/01/2024] [Accepted: 07/03/2024] [Indexed: 08/04/2024] Open
Abstract
In patients with severe acute respiratory syndrome coronavirus 2 (which causes coronavirus disease 2019 [COVID-19]), oxidative stress (OS) is associated with disease severity and death. OS is also involved in the pathogenesis of atherosclerosis (AS). Previous studies have shown that geniposide has anti-inflammatory and anti-viral properties, and can protect cells against OS. However, the potential target(s) of geniposide in patients with COVID-19 and AS, as well as the mechanism it uses, are unclear. We combined pharmacology and bioinformatics analysis to obtain geniposide against COVID-19/AS targets, and build protein-protein interaction network to filter hub genes. The hub genes were performed an enrichment analysis by ClueGO, including Gene Ontology and KEGG. The Enrichr database and the target microRNAs (miRNAs) of hub genes were predicted through the MiRTarBase via Enrichr. The common miRNAs were used to construct the miRNAs-mRNAs regulated network, and the miRNAs' function was evaluated by mirPath v3.0 software. Two hundred forty-seven targets of geniposide were identified in patients with COVID-19/AS comorbidity by observing the overlap between the genes modulated by geniposide, COVID-19, and AS. A protein-protein interaction network of geniposide in patients with COVID-19/AS was constructed, and 27 hub genes were identified. The results of enrichment analysis suggested that geniposide may be involved in regulating the OS via the FoxO signaling pathway. MiRNA-mRNA network revealed that hsa-miR-34a-5p may play an important role in the therapeutic mechanism of geniposide in COVID-19/AS patients. Our study found that geniposide represents a promising therapy for patients with COVID-19 and AS comorbidity. Furthermore, the target genes and miRNAs that we identified may aid the development of new treatment strategies against COVID-19/AS.
Collapse
Affiliation(s)
- Lijin Qing
- First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510405, China
| | - Wei Wu
- First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510405, China
| |
Collapse
|
31
|
Kato PJ, Kato J, Napier JA, Khorfan R, Aziz A. Unstable Carotid Artery Thrombus in a Patient With COVID-19 Infection. Vasc Endovascular Surg 2024; 58:659-662. [PMID: 38498941 DOI: 10.1177/15385744241240543] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/20/2024]
Abstract
We present a case of an unvaccinated, 43-year-old African American female patient with COVID-19 infection and clinical evidence of a left hemispheric stroke. A non-occlusive thrombus with a radiographic target lesion was identified on computed tomography angiography (CTA). A multi-disciplinary discussion regarding concern for embolization was provided due to its unstable nature, as well as evidence of recent stroke. Given her acute COVID-19 infection, symptomatology, and radiographic findings, it was concluded that the etiology of her stroke appeared most consistent with a hypercoagulable-related embolism rather than an atheroembolic event. The patient underwent left carotid artery thrombectomy with bovine patch angioplasty. Operative findings included: left carotid thrombus, minimal plaque after evacuation of the thrombus, and a small proximal internal carotid artery diameter. Given concern for stenosis with primary repair a bovine pericardial patch angioplasty was performed. We present a paradigm for extracranial carotid thrombectomy with therapeutic anticoagulation for COVID-related spontaneous arterial thrombosis.
Collapse
Affiliation(s)
- Patrick J Kato
- Department of Surgery, Section of Vascular and Endovascular Surgery, Trinity Health Ann Arbor, Ypsilanti, MI, USA
| | - Jesse Kato
- Department of Surgery, Section of Vascular and Endovascular Surgery, Trinity Health Ann Arbor, Ypsilanti, MI, USA
| | - Jarred A Napier
- Department of Surgery, Section of Vascular and Endovascular Surgery, Trinity Health Ann Arbor, Ypsilanti, MI, USA
| | - Rhami Khorfan
- Department of Surgery, Section of Vascular and Endovascular Surgery, Trinity Health Ann Arbor, Ypsilanti, MI, USA
| | - Abdulhameed Aziz
- Department of Surgery, Section of Vascular and Endovascular Surgery, Trinity Health Ann Arbor, Ypsilanti, MI, USA
| |
Collapse
|
32
|
Rettew A, Garrahy I, Rahimian S, Brown R, Sangha N. COVID-19 Coagulopathy. Life (Basel) 2024; 14:953. [PMID: 39202695 PMCID: PMC11355811 DOI: 10.3390/life14080953] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2024] [Revised: 07/25/2024] [Accepted: 07/26/2024] [Indexed: 09/03/2024] Open
Abstract
Coronavirus disease of 2019 (COVID-19) is the respiratory viral infection caused by the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). Despite being a primary respiratory illness, it is commonly complicated by systemic involvement of the vasculature leading to arterial and venous thrombosis. In this review, we will focus on the association between COVID-19 and thrombosis. We will highlight the pathophysiology of COVID-19 coagulopathy. The clinical manifestations of COVID-19 vasculopathy will be discussed with a focus on venous and arterial thromboembolic events. COVID-19 vasculopathy and disseminated intravascular coagulation (DIC) are distinguished within, as well as areas of controversy, such as "long COVID". Finally, the current professional guidelines on prevention and treatment of thrombosis associated with SARS-CoV-2 infection will be discussed.
Collapse
Affiliation(s)
| | - Ian Garrahy
- Tower Health System, Reading Hospital, West Reading, PA 19611, USA; (A.R.); (S.R.); (R.B.); (N.S.)
| | | | | | | |
Collapse
|
33
|
Wang H, Wu S, Pan D, Meng W, Hu L, Zhang H, Ning Y, Guo J, Gu Y. Causal relationships between COVID-19 and venous thromboembolism: A mendelian randomization analysis. Phlebology 2024:2683555241266659. [PMID: 39033375 DOI: 10.1177/02683555241266659] [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: 07/23/2024]
Abstract
Objective: Observational studies show the correlation between COVID-19 and venous thromboembolism (VTE) risk. However, the causal effects remain uncertain. We aimed to explore the potential causal association between COVID-19 and VTE using Mendelian randomization (MR) design. Methods: Two-sample MR was used to evaluate the potential causality between COVID-19 and VTE by selecting single-nucleotide polymorphisms (SNPs) as instrumental variables (IVs) from genome-wide association studies (GWAS). The weighted median, MR-Egger, simple mode, and weighted mode were employed as supplementary methods for MR estimations, with the inverse-variance weighted (IVW) method serving as the principal analysis. In addition, we took sensitivity analyses, including Cochran's test, MR-Pleiotropy Residual Sum and Outlier (MR-PRESSO), and leave-one-out analysis to ensure that we obtained stable and reliable results. Results: Our study selected 26 COVID-19 severity, 31 COVID-19 hospitalization, and 13 COVID-19 susceptibility SNPs as instrumental variables. The IVW analysis results revealed that there was no causal relationship between COVID-19 severity, hospitalization, or susceptibility and VTE, with odds ratios of 0.974 (95%CI: 0.936-1.013, p = 0.19), 0.976 (95%CI: 0.918-1.039, p = 0.447), and 0.908 (95%CI: 0.775-1.065, p = 0.235), respectively. The IVW approach yielded consistent results with MR-Egger, Weighted Median simple mode, and weighted mode. MR-PRESSO and sensitivity analysis further confirmed the stability and consistency of the MR results. Conclusions: This study did not find evidence to support a causal relationship between COVID-19 and VTE at the genetic level. Further investigation is warranted to determine if the significant association reported in previous observational studies between the two is due to confounding factors.
Collapse
Affiliation(s)
- Hui Wang
- Department of Vascular Surgery, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Sensen Wu
- Department of Vascular Surgery, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Dikang Pan
- Department of Vascular Surgery, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Wenzhuo Meng
- Department of Vascular Surgery, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Lefan Hu
- Department of Vascular Surgery, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Hanyu Zhang
- Department of Vascular Surgery, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Yachan Ning
- Department of Vascular Surgery, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Jianming Guo
- Department of Vascular Surgery, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Yongquan Gu
- Department of Vascular Surgery, Xuanwu Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
34
|
Abrishami M, Hassanpour K, Bayat K, Hosseini SM, Amini N, Abrishami M, Sayyah Ensan P, Heidarzadeh HR. Optic nerve head alterations after COVID-19: an optical coherence tomography angiography-based longitudinal study. J Int Med Res 2024; 52:3000605241263236. [PMID: 39082309 PMCID: PMC11295231 DOI: 10.1177/03000605241263236] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Accepted: 05/31/2024] [Indexed: 08/04/2024] Open
Abstract
OBJECTIVE To evaluate longitudinal changes in peripapillary vessel density (VD) following coronavirus disease 2019 (COVID-19) using optical coherence tomography angiography. METHODS As part of a prospective longitudinal observational study, we studied healthy individuals with a history of mild COVID-19 confirmed via real-time polymerase chain reaction. After recovery, we used the Optovue RTVue XR Avanti machine to perform optic nerve head (ONH) imaging. We also assessed the VD of all vessels and of small vessels in the disc and the radial peripapillary capillary (RPC) network at 1 and 3 months post-recovery. RESULTS We included 17 patients (34 eyes; mean age: 36.9 ± 10.2 years, range: 24-62 years) who had recovered from COVID-19. No changes were observed in the ONH parameters. However, there was a noticeable trend of increased small vessel VD values in the RPC. These increases were significant for the peripapillary whole, superior hemifield, inferior-temporal, temporal-superior, and superior-temporal small vessels. Moreover, the evaluation of all vessel VD values in the RPC revealed a significant decrease in the inside disc and a significant increase in a grid-based inferior region. CONCLUSION COVID-19 may affect VD of the RPC in the ONH, and should be considered in ONH evaluations.
Collapse
Affiliation(s)
- Mojtaba Abrishami
- Eye Research Center, Khatam-al-Anbia Eye Hospital, Mashhad University of Medical Sciences, Mashhad, Iran
- Ocular Oncology Service, Department of Ophthalmology and Visual Sciences, University of Toronto, Toronto, Canada
| | - Kiana Hassanpour
- Ophthalmic Research Center, Research Institute for Ophthalmology and Vision Science, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Kia Bayat
- Ophthalmic Research Center, Research Institute for Ophthalmology and Vision Science, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Seyedeh Maryam Hosseini
- Eye Research Center, Khatam-al-Anbia Eye Hospital, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Nasibeh Amini
- Eye Research Center, Khatam-al-Anbia Eye Hospital, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Majid Abrishami
- Eye Research Center, Khatam-al-Anbia Eye Hospital, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Parham Sayyah Ensan
- School of Medicine, UFR Simone Veil, Université Versailles Saint-Quentin-en-Yvelines (Paris Saclay University), Paris, France
| | - Hamid Reza Heidarzadeh
- Eye Research Center, Khatam-al-Anbia Eye Hospital, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
35
|
Li Y, Gao Z, Zheng X, Pan Y, Xu J, Li Y, Chen H. Interventional Removal of Travelling Microthrombi Using Targeted Magnetic Microbubble. Adv Healthc Mater 2024:e2401631. [PMID: 38938195 DOI: 10.1002/adhm.202401631] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2024] [Revised: 06/17/2024] [Indexed: 06/29/2024]
Abstract
Microthrombus is one of the major causes of the sequelae of Corona Virus Disease 2019 (COVID-19 and leads to subsequent embolism and necrosis. Due to their small size and irregular movements, the early detection and efficient removal of microthrombi in vivo remain a great challenge. In this work, an interventional method is developed to identify and remove the traveling microthrombi using targeted-magnetic-microbubbles (TMMBs) and an interventional magnetic catheter. The thrombus-targeted drugs are coated on the TMMBs and magnetic nanoparticles are shelled inside, which allow not only targeted adhesion onto the traveling microthrombi, but also the effective capture by the magnetic catheter in the vessel. In the proof-of-concept experiments in the rat models, the concentration of microthrombus is reduced by more than 60% in 3 min, without damaging the organs. It is a promising method for treating microthrombus issues.
Collapse
Affiliation(s)
- Yongjian Li
- The State Key Laboratory of Tribology, Department of Mechanical Engineering, Tsinghua University, Beijing, 100084, P. R. China
| | - Zujie Gao
- The State Key Laboratory of Tribology, Department of Mechanical Engineering, Tsinghua University, Beijing, 100084, P. R. China
| | - Xiaobing Zheng
- The State Key Laboratory of Tribology, Department of Mechanical Engineering, Tsinghua University, Beijing, 100084, P. R. China
| | - Yunfan Pan
- The State Key Laboratory of Tribology, Department of Mechanical Engineering, Tsinghua University, Beijing, 100084, P. R. China
| | - Jinlong Xu
- The State Key Laboratory of Tribology, Department of Mechanical Engineering, Tsinghua University, Beijing, 100084, P. R. China
| | - Yan Li
- The State Key Laboratory of Tribology, Department of Mechanical Engineering, Tsinghua University, Beijing, 100084, P. R. China
| | - Haosheng Chen
- The State Key Laboratory of Tribology, Department of Mechanical Engineering, Tsinghua University, Beijing, 100084, P. R. China
| |
Collapse
|
36
|
Subramaniam S, Kenney D, Jayaraman A, O’Connell AK, Walachowski S, Montanaro P, Reinhardt C, Colucci G, Crossland NA, Douam F, Bosmann M. Aging is associated with an insufficient early inflammatory response of lung endothelial cells in SARS-CoV-2 infection. Front Immunol 2024; 15:1397990. [PMID: 38911865 PMCID: PMC11190167 DOI: 10.3389/fimmu.2024.1397990] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2024] [Accepted: 05/20/2024] [Indexed: 06/25/2024] Open
Abstract
Advanced age is associated with an increased susceptibility to Coronavirus Disease (COVID)-19 and more severe outcomes, although the underlying mechanisms are understudied. The lung endothelium is located next to infected epithelial cells and bystander inflammation may contribute to thromboinflammation and COVID-19-associated coagulopathy. Here, we investigated age-associated SARS-CoV-2 pathogenesis and endothelial inflammatory responses using humanized K18-hACE2 mice. Survival was reduced to 20% in aged mice (85-112 weeks) versus 50% in young mice (12-15 weeks) at 10 days post infection (dpi). Bulk RNA-sequencing of endothelial cells from mock and infected mice at 2dpi of both age groups (aged: 72-85 weeks; young: 15 weeks) showed substantially lower significant differentially regulated genes in infected aged mice than in young mice (712 versus 2294 genes). Viral recognition and anti-viral pathways such as RIG-I-like receptor signaling, NOD-like receptor signaling and interferon signaling were regulated in response to SARS-CoV-2. Young mice showed several fold higher interferon responses (Ifitm3, Ifit1, Isg15, Stat1) and interferon-induced chemokines (Cxcl10 and Cxcl11) than aged mice. Endothelial cells from infected young mice displayed elevated expression of chemokines (Cxcl9, Ccl2) and leukocyte adhesion markers (Icam1) underscoring that inflammation of lung endothelium during infection could facilitate leukocyte adhesion and thromboinflammation. TREM1 and acute phase response signaling were particularly prominent in endothelial cells from infected young mice. Immunohistochemistry was unable to detect viral protein in pulmonary endothelium. In conclusion, our data demonstrate that the early host response of the endothelium to SARS-CoV-2 infection declines with aging, which could be a potential contributor to disease severity.
Collapse
Affiliation(s)
- Saravanan Subramaniam
- Pulmonary Center, Department of Medicine, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, United States
| | - Devin Kenney
- National Emerging Infectious Diseases Laboratories (NEIDL), Boston University, Boston, MA, United States
- Department of Virology, Immunology and Microbiology, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, United States
| | - Archana Jayaraman
- Pulmonary Center, Department of Medicine, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, United States
| | - Aoife Kateri O’Connell
- National Emerging Infectious Diseases Laboratories (NEIDL), Boston University, Boston, MA, United States
- Department of Pathology and Laboratory Medicine, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, United States
| | - Sarah Walachowski
- Pulmonary Center, Department of Medicine, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, United States
- Center for Thrombosis and Hemostasis, University Medical Center of the Johannes Gutenberg-University, Mainz, Germany
| | - Paige Montanaro
- Department of Virology, Immunology and Microbiology, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, United States
- Department of Pathology and Laboratory Medicine, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, United States
| | - Christoph Reinhardt
- Center for Thrombosis and Hemostasis, University Medical Center of the Johannes Gutenberg-University, Mainz, Germany
- German Center for Cardiovascular Research (DZHK), Partner Site Rhine-Main, Mainz, Germany
| | - Giuseppe Colucci
- Outer Corelab, Viollier AG, Allschwil, Switzerland
- Department of Hematology, University of Basel, Basel, Switzerland
| | - Nicholas A. Crossland
- National Emerging Infectious Diseases Laboratories (NEIDL), Boston University, Boston, MA, United States
- Department of Virology, Immunology and Microbiology, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, United States
- Department of Pathology and Laboratory Medicine, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, United States
| | - Florian Douam
- National Emerging Infectious Diseases Laboratories (NEIDL), Boston University, Boston, MA, United States
- Department of Virology, Immunology and Microbiology, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, United States
| | - Markus Bosmann
- Pulmonary Center, Department of Medicine, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, United States
- National Emerging Infectious Diseases Laboratories (NEIDL), Boston University, Boston, MA, United States
- Center for Thrombosis and Hemostasis, University Medical Center of the Johannes Gutenberg-University, Mainz, Germany
| |
Collapse
|
37
|
Vinod P, Krishnappa V, Rathell W, Amir S, Sundil S, Dogbey G, Patel H, Herzog W. Effect of Aspirin Use on the Adverse Outcomes in Patients Hospitalized for COVID-19. Cardiol Res 2024; 15:179-188. [PMID: 38994222 PMCID: PMC11236346 DOI: 10.14740/cr1645] [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: 03/31/2024] [Accepted: 05/02/2024] [Indexed: 07/13/2024] Open
Abstract
Background Coronavirus disease 2019 (COVID-19) triggers multiple components of the immune system and causes inflammation of endothelial walls across vascular beds, resulting in respiratory failure, arterial and venous thrombosis, myocardial injury, and multi-organ failure leading to death. Early in the COVID-19 pandemic, aspirin was suggested for the treatment of symptomatic individuals, given its analgesic, antipyretic, anti-inflammatory, anti-thrombotic, and antiviral effects. This study aimed to evaluate the association of aspirin use with various clinical outcomes in patients hospitalized for COVID-19. Methods This was a retrospective study involving patients aged ≥ 18 years and hospitalized for COVID-19 from March 2020 to October 2020. Primary outcomes were acute cardiovascular events (ST elevation myocardial infarction (STEMI), type 1 non-ST elevation myocardial infarction (NSTEMI), acute congestive heart failure (CHF), and acute stroke) and death. Secondary outcomes were respiratory failure, need for mechanical ventilation, and acute deep vein thrombosis (DVT)/pulmonary embolism (PE). Results Of 376 patients hospitalized for COVID-19, 128 were taking aspirin. Significant proportions of native Americans were hospitalized for COVID-19 in both aspirin (22.7%) and non-aspirin (24.6%) groups. Between aspirin and non-aspirin groups, no significant differences were found with regard to mechanical ventilator support (21.1% vs. 15.3%, P = 0.16), acute cardiovascular events (7.8% vs. 5.2%, P = 0.32), acute DVT/PE (3.9% vs. 5.2%, P = 0.9), readmission rate (13.3% vs. 12.9%, P = 0.91) and mortality (23.4% vs. 20.2%, P = 0.5); however, the median duration of mechanical ventilation was significantly shorter (7 vs. 9 days, P = 0.04) and median length of hospitalization was significantly longer (5.5 vs. 4 days, P = 0.01) in aspirin group compared to non-aspirin group. Conclusion No significant differences were found in acute cardiovascular events, acute DVT/PE, mechanical ventilator support, and mortality rate between hospitalized COVID-19 patients who were taking aspirin compared to those not taking aspirin. However, larger studies are required to confirm our findings.
Collapse
Affiliation(s)
- Poornima Vinod
- Department of Internal Medicine, University of North Carolina Health Southeastern, Lumberton, NC, USA
- Department of Medicine, Campbell University, Buies Creek, NC, USA
| | - Vinod Krishnappa
- Department of Internal Medicine, University of North Carolina Health Southeastern, Lumberton, NC, USA
| | - William Rathell
- Department of Internal Medicine, University of North Carolina Health Southeastern, Lumberton, NC, USA
| | - Saira Amir
- Department of Nephrology, University of Maryland, Baltimore, MD, USA
| | - Subrina Sundil
- Department of Nephrology, East Carolina University, Greenville, NC, USA
| | - Godwin Dogbey
- Campbell University School of Osteopathic Medicine, Buies Creek, NC, USA
| | - Hiten Patel
- Department of Cardiology, University of North Carolina Health Southeastern, Lumberton, NC, USA
| | - William Herzog
- Department of Cardiology, University of North Carolina Health Southeastern, Lumberton, NC, USA
- Department of Cardiology, Duke University, Durham, NC, USA
| |
Collapse
|
38
|
Zhong L, Lin Q, He L, Liu D, Zhu L, Zeng Q, Song J. Time to maximum amplitude of thromboelastography can predict mortality in patients with severe COVID-19: a retrospective observational study. Front Med (Lausanne) 2024; 11:1356283. [PMID: 38756947 PMCID: PMC11097111 DOI: 10.3389/fmed.2024.1356283] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2023] [Accepted: 04/19/2024] [Indexed: 05/18/2024] Open
Abstract
Objective To predict mortality in severe patients with COVID-19 at admission to the intensive care unit (ICU) using thromboelastography (TEG). Methods This retrospective, two-center, observational study involved 87 patients with PCR-and chest CT-confirmed severe COVID-19 who were admitted to at Wuhan Huoshenshan Hospital and the 908th Hospital of Chinese PLA Logistic Support Force between February 2020 and February 2023. Clinic demographics, laboratory results, and outcomes were compared between those who survived and those who died during hospitalization. Results Thromboelastography showed that of the 87 patients, 14 were in a hypercoagulable state, 25 were in a hypocoagulable state, and 48 were normal, based on the time to maximum amplitude (TMA). Patients who died showed significantly lower α angle, but significantly longer R-time, K-time and TMA than patients who survived. Random forest selection showed that K-time, TMA, prothrombin time (PT), international normalized ratio (INR), D-dimer, C-reactive protein (CRP), aspartate aminotransferase (AST), and total bilirubin (Tbil) were significant predictors. Multivariate logistic regression identified that TMA and CRP were independently associated with mortality. TMA had a greater predictive power than CRP levels based on time-dependent AUCs. Patients with TMA ≥ 26.4 min were at significantly higher risk of mortality (hazard ratio 3.99, 95% Confidence Interval, 1.92-8.27, p < 0.01). Conclusion TMA ≥26.4 min at admission to ICU may be an independent predictor of in-hospital mortality for patients with severe COVID-19.
Collapse
Affiliation(s)
- Lincui Zhong
- Intensive Care Unit, The 908th Hospital of Chinese PLA Logistic Support Force, Nanchang, Jiangxi, China
| | - Qingwei Lin
- Intensive Care Unit, The 908th Hospital of Chinese PLA Logistic Support Force, Nanchang, Jiangxi, China
| | - Longping He
- Intensive Care Unit, The 908th Hospital of Chinese PLA Logistic Support Force, Nanchang, Jiangxi, China
| | - Dongmei Liu
- Intensive Care Unit, Huoshenshan Hospital, Wuhan, Hubei, China
- Intensive Care Unit, The 940th Hospital of Chinese PLA Logistic Support Force, Lanzhou, Gansu, China
| | - Lin Zhu
- Intensive Care Unit, Huoshenshan Hospital, Wuhan, Hubei, China
- Department of Critical Care Medicine, The 944th Hospital of Chinese PLA Logistic Support Force, Jiuquan, Gansu, China
| | - Qingbo Zeng
- Intensive Care Unit, The 908th Hospital of Chinese PLA Logistic Support Force, Nanchang, Jiangxi, China
| | - Jingchun Song
- Intensive Care Unit, The 908th Hospital of Chinese PLA Logistic Support Force, Nanchang, Jiangxi, China
- Intensive Care Unit, Huoshenshan Hospital, Wuhan, Hubei, China
| |
Collapse
|
39
|
Shukla E, Narain U, Gupta A. Hemodialysis: The Life Boat for AKI Patients in the COVID Cytokine Storm. Indian J Nephrol 2024; 34:237-240. [PMID: 39114395 PMCID: PMC11302509 DOI: 10.4103/ijn.ijn_96_23] [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: 03/14/2023] [Accepted: 07/01/2023] [Indexed: 08/10/2024] Open
Abstract
Background Interleukin-6 (IL-6), a biomarker of hyperinflammatory immune response, can be used to determine the severity of coronavirus disease 2019 (COVID-19) in patients with multi-organ involvement requiring critical care. The aim of our study is to understand the utility of hemodialysis, not only in terms of reducing renal burden, but also improving the outcome by tackling the COVID cytokine storm syndrome. Materials and Methods In this prospective, observational study, 126 patients admitted to the COVID intensive care unit (ICU) wards were treated with hemodialysis for acute kidney injury (AKI). Patients' routine baseline blood parameters were evaluated. IL-6 was measured predialysis in all patients and on the day of discharge in the patients who survived. Results Out of total 126 patients, 79 were survivors and 47 were nonsurvivors. Among nonsurvivors, majority were older (P = 0.009). Both the groups had a higher percentage of males (78.72% and 55.69% in survivors and nonsurvivors, respectively). Mean neutrophil lymphocyte ratio (NLR) and D-dimer level were significantly higher in nonsurvivors compared to survivors (P < 0.001). Mean serum urea, creatinine, and IL-6 levels were significantly greater in nonsurvivors (P < 0.001). Mean number of hemodialysis sessions received by survivors was higher. The curve between delta IL-6 and delta serum creatinine for survivors showed a significant positive association (r = 0.819, P < 0.001). Conclusion Our study establishes IL-6 as a poor outcome predictor in COVID ICU patients with AKI. It also emphasizes the use of hemodialysis as a cost-effective lifesaving therapeutic interventional modality to not only improve the renal outcome, but also curb the cytokine storm by reducing IL-6 levels.
Collapse
Affiliation(s)
- Eesha Shukla
- Department of Medicine, MLN Medical College, Prayagraj, Uttar Pradesh, India
| | - Upma Narain
- Department of Microbiology, TEJAS Micro Diagnostic Centre, Tagore Town, Prayagraj, Uttar Pradesh, India
| | - Arvind Gupta
- Department of Nephrology, SRN Hospital, MLN Medical College, Prayagraj, Uttar Pradesh, India
| |
Collapse
|
40
|
Riou M, Coste F, Meyer A, Enache I, Talha S, Charloux A, Reboul C, Geny B. Mechanisms of Pulmonary Vasculopathy in Acute and Long-Term COVID-19: A Review. Int J Mol Sci 2024; 25:4941. [PMID: 38732160 PMCID: PMC11084496 DOI: 10.3390/ijms25094941] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Revised: 04/26/2024] [Accepted: 04/26/2024] [Indexed: 05/13/2024] Open
Abstract
Despite the end of the pandemic, coronavirus disease 2019 (COVID-19) remains a major public health concern. The first waves of the virus led to a better understanding of its pathogenesis, highlighting the fact that there is a specific pulmonary vascular disorder. Indeed, COVID-19 may predispose patients to thrombotic disease in both venous and arterial circulation, and many cases of severe acute pulmonary embolism have been reported. The demonstrated presence of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) within the endothelial cells suggests that direct viral effects, in addition to indirect effects of perivascular inflammation and coagulopathy, may contribute to pulmonary vasculopathy in COVID-19. In this review, we discuss the pathological mechanisms leading to pulmonary vascular damage during acute infection, which appear to be mainly related to thromboembolic events, an impaired coagulation cascade, micro- and macrovascular thrombosis, endotheliitis and hypoxic pulmonary vasoconstriction. As many patients develop post-COVID symptoms, including dyspnea, we also discuss the hypothesis of pulmonary vascular damage and pulmonary hypertension as a sequela of the infection, which may be involved in the pathophysiology of long COVID.
Collapse
Affiliation(s)
- Marianne Riou
- Translational Medicine Federation of Strasbourg (FMTS), University of Strasbourg, CRBS, Team 3072 “Mitochondria, Oxidative Stress and Muscle Protection”, 1 rue Eugène Boeckel, CS 60026, 67084 Strasbourg, France; (M.R.); (A.M.); (I.E.); (S.T.); (A.C.)
- Physiology and Functional Exploration Service, University Hospital of Strasbourg, 1 Place de l’hôpital, 67091 Strasbourg, France
| | - Florence Coste
- EA4278, Laboratoire de Pharm-Ecologie Cardiovasculaire, UFR Sciences Technologies Santé, Pôle Sport et Recherche, 74 rue Louis Pasteur, 84000 Avignon, France; (F.C.); (C.R.)
| | - Alain Meyer
- Translational Medicine Federation of Strasbourg (FMTS), University of Strasbourg, CRBS, Team 3072 “Mitochondria, Oxidative Stress and Muscle Protection”, 1 rue Eugène Boeckel, CS 60026, 67084 Strasbourg, France; (M.R.); (A.M.); (I.E.); (S.T.); (A.C.)
- Physiology and Functional Exploration Service, University Hospital of Strasbourg, 1 Place de l’hôpital, 67091 Strasbourg, France
| | - Irina Enache
- Translational Medicine Federation of Strasbourg (FMTS), University of Strasbourg, CRBS, Team 3072 “Mitochondria, Oxidative Stress and Muscle Protection”, 1 rue Eugène Boeckel, CS 60026, 67084 Strasbourg, France; (M.R.); (A.M.); (I.E.); (S.T.); (A.C.)
- Physiology and Functional Exploration Service, University Hospital of Strasbourg, 1 Place de l’hôpital, 67091 Strasbourg, France
| | - Samy Talha
- Translational Medicine Federation of Strasbourg (FMTS), University of Strasbourg, CRBS, Team 3072 “Mitochondria, Oxidative Stress and Muscle Protection”, 1 rue Eugène Boeckel, CS 60026, 67084 Strasbourg, France; (M.R.); (A.M.); (I.E.); (S.T.); (A.C.)
- Physiology and Functional Exploration Service, University Hospital of Strasbourg, 1 Place de l’hôpital, 67091 Strasbourg, France
| | - Anne Charloux
- Translational Medicine Federation of Strasbourg (FMTS), University of Strasbourg, CRBS, Team 3072 “Mitochondria, Oxidative Stress and Muscle Protection”, 1 rue Eugène Boeckel, CS 60026, 67084 Strasbourg, France; (M.R.); (A.M.); (I.E.); (S.T.); (A.C.)
- Physiology and Functional Exploration Service, University Hospital of Strasbourg, 1 Place de l’hôpital, 67091 Strasbourg, France
| | - Cyril Reboul
- EA4278, Laboratoire de Pharm-Ecologie Cardiovasculaire, UFR Sciences Technologies Santé, Pôle Sport et Recherche, 74 rue Louis Pasteur, 84000 Avignon, France; (F.C.); (C.R.)
| | - Bernard Geny
- Translational Medicine Federation of Strasbourg (FMTS), University of Strasbourg, CRBS, Team 3072 “Mitochondria, Oxidative Stress and Muscle Protection”, 1 rue Eugène Boeckel, CS 60026, 67084 Strasbourg, France; (M.R.); (A.M.); (I.E.); (S.T.); (A.C.)
- Physiology and Functional Exploration Service, University Hospital of Strasbourg, 1 Place de l’hôpital, 67091 Strasbourg, France
| |
Collapse
|
41
|
Naeimzadeh F, Sadeghi A, Saghaleini S, Sarbakhsh P, Mahmoodpoor A, Gharekhani A. Effect of parenteral L-carnitine in hospitalized patients with moderate to severe COVID-19: A randomized double-blind clinical trial. BIOIMPACTS : BI 2024; 15:30261. [PMID: 39963575 PMCID: PMC11830133 DOI: 10.34172/bi.2024.30261] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/08/2024] [Revised: 02/26/2024] [Accepted: 03/05/2024] [Indexed: 02/20/2025]
Abstract
Introduction Pro-inflammatory responses have an important role in developing coronavirus disease 2019 (COVID-19). L-carnitine (LC) has been known to possess anti-inflammatory, anticoagulant, and antiviral effects. So, we aimed to evaluate the efficacy of LC in hospitalized patients with moderate-to-severe COVID-19. Methods This double-blind, placebo-controlled, randomized clinical trial was conducted on hospitalized patients with moderate to severe COVID-19. The patients were randomized (1:1) to receive LC (n = 50) at a dose of 20 mg/kg or matching placebo (n = 51) from normal saline once daily for 14 days or until hospitalization and standard care. The primary outcome was hospital mortality and disease severity according to the World Health Organization's clinical progression scale. We also assessed the free carnitine level at baseline and the end of the study. C-reactive protein (CRP), ferritin, D-dimer, lactate dehydrogenase (LDH), and improvement of respiratory conditions were chosen as secondary outcomes. Results From 104 patients who met the inclusion criteria, 101 individuals' data were analyzed. The LC group showed a significant reduction in LDH levels (P = 0.003), although CRP, ferritin, and D-dimer levels did not significantly differ from the placebo group. Also, no significant difference was observed in disease severity, oxygenation status, hospital mortality, or hospital stay between the two groups. Additionally, there was no increase in serum-free carnitine levels in the LC group (P > 0.05 for all). Conclusion The results of the current study did not support the superiority of LC over placebo in improving oxygenation, decreasing mortality, and hospital stay, as well as CRP, ferritin, and D-dimer in moderate to severe COVID-19 patients. Trial Registration IRCT20170609034406N10; https://en.irct.ir/trial/60306.
Collapse
Affiliation(s)
- Farnaz Naeimzadeh
- Student Research Committee, Tabriz University of Medical Sciences, Iran
- Department of Clinical Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Armin Sadeghi
- Tuberculosis and Lung Disease Research Center of Tabriz University of Medical Sciences, Tabriz, Iran
| | - Seiedhadi Saghaleini
- Department of Anesthesiology, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Parvin Sarbakhsh
- Department of Statistics and Epidemiology, Faculty of Public Health, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Ata Mahmoodpoor
- Department of Anesthesiology, Tabriz University of Medical Sciences, Tabriz, Iran
- Research Center for Integrative Medicine in Aging, Aging Research Institute, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Afshin Gharekhani
- Department of Clinical Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran
- Liver and Gastrointestinal Diseases Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
42
|
Zabeeda W, Cohen JB, Reiner Benaim A, Zarour S, Lichter Y, Matot I, Goren O. Utility of NICaS Non-Invasive Hemodynamic Monitoring in Critically Ill Patients with COVID-19. J Clin Med 2024; 13:2072. [PMID: 38610837 PMCID: PMC11012855 DOI: 10.3390/jcm13072072] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2024] [Revised: 03/21/2024] [Accepted: 03/27/2024] [Indexed: 04/14/2024] Open
Abstract
(1) Background: COVID-19 presented many challenges to effective treatments, such as managing cardiovascular insufficiency while mitigating risks to healthcare providers. This study utilized NICaS, a non-invasive hemodynamic monitor that provides advanced data via whole-body impedance analysis. We investigated the associated trends in hemodynamic parameters obtained by the NICaS device and their correlation with in-hospital all-cause mortality during COVID-19 hospitalization in the intensive care unit. (2) Methods: Data from 29 patients with COVID-19 admitted to the intensive care unit and monitored with NICaS between April 2020 and February 2021 were analyzed retrospectively. (3) Results: Decreasing cardiac output and cardiac power were significantly associated with death. Total peripheral resistance was significantly increasing in non-survivors as was total body water percentage. Those admitted with a heart rate above 90 beats per minute had a significantly reduced survival. (4) Conclusions: Non-invasive hemodynamic monitoring via the NICaS device is simple and effective in evaluating critically ill patients with COVID-19 and may help guide clinical management via remote monitoring. Controlling tachycardia may help ensure adequate oxygen supply-demand ratio. A hint toward a beneficiary effect of a restrictive fluid balance may be observed.
Collapse
Affiliation(s)
- Wisam Zabeeda
- Department of Anesthesiology, Pain and Intensive Care, Tel Aviv Medical Center, Tel Aviv University, Tel Aviv 6997801, Israel; (W.Z.); (S.Z.); (Y.L.); (I.M.); (O.G.)
| | - Jonah Benjamin Cohen
- Department of Anesthesiology, Pain and Intensive Care, Tel Aviv Medical Center, Tel Aviv University, Tel Aviv 6997801, Israel; (W.Z.); (S.Z.); (Y.L.); (I.M.); (O.G.)
| | - Anat Reiner Benaim
- Department of Epidemiology, Biostatistics and Community Health Sciences, School of Public Health, Faculty of Health Sciences, Ben Gurion University of the Negev, Beer Sheva 8410501, Israel;
| | - Shiri Zarour
- Department of Anesthesiology, Pain and Intensive Care, Tel Aviv Medical Center, Tel Aviv University, Tel Aviv 6997801, Israel; (W.Z.); (S.Z.); (Y.L.); (I.M.); (O.G.)
| | - Yael Lichter
- Department of Anesthesiology, Pain and Intensive Care, Tel Aviv Medical Center, Tel Aviv University, Tel Aviv 6997801, Israel; (W.Z.); (S.Z.); (Y.L.); (I.M.); (O.G.)
| | - Idit Matot
- Department of Anesthesiology, Pain and Intensive Care, Tel Aviv Medical Center, Tel Aviv University, Tel Aviv 6997801, Israel; (W.Z.); (S.Z.); (Y.L.); (I.M.); (O.G.)
| | - Or Goren
- Department of Anesthesiology, Pain and Intensive Care, Tel Aviv Medical Center, Tel Aviv University, Tel Aviv 6997801, Israel; (W.Z.); (S.Z.); (Y.L.); (I.M.); (O.G.)
| |
Collapse
|
43
|
Nana P, Dakis K, Spanos K, Tsolaki V, Karavidas N, Zakynthinos G, Kouvelos G, Giannoukas A, Matsagkas M. COVID-19 related peripheral arterial thrombotic events in intensive care unit and non-intensive care unit patients: A retrospective case series. Vascular 2024; 32:366-373. [PMID: 36395575 PMCID: PMC9679328 DOI: 10.1177/17085381221140159] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/27/2023]
Abstract
OBJECTIVES COVID-19 associated arterial thrombosis has been attributed to multiple inflammation and coagulation mechanisms. The aim of this study was to report the experience of a tertiary center on COVID-19 patients managed for acute peripheral arterial thrombosis. METHODS A single-center case series was conducted, including retrospectively collected data from all COVID-19 patients presenting arterial thrombosis, from March 2020 to February 2022. Intensive care unit (ICU) and non-ICU cases were included. The primary outcomes were mortality, successful revascularization, and amputation at 30 days. RESULTS Twenty patients presented peripheral arterial thrombosis (90% males, mean age 65 years (16-82 years)). Eighteen were diagnosed with the Delta variant and none was previously vaccinated. All cases presented acute lower limb ischemia; in 20% with bilateral involvement. Nine patients were hospitalized in the ward while 11 in the ICU. From the non-ICU cases, five presented Rutherford IIb and four cases, Rutherford's IIa ischemia. Six cases underwent revascularization (67%). Two of them were finally amputated (33%) and two died during hospitalization (33%). Two revascularizations were considered successful (33%). The ICU group presented mainly with Rutherford's III ischemia (54.5%). The mortality in the ICU cohort was 72.7%. Only one patient underwent successful revascularization and two were amputated in this subgroup. Early mortality was 50% for the total cohort while the type of management was not related to mortality. CONCLUSIONS Covid-19 related arterial thrombosis in non-vaccinated population is associated with 50% early mortality; increased up to 72% in the ICU patients. The amputation rate was 20% while only 40% of the revascularizations were considered successful.
Collapse
Affiliation(s)
- Petroula Nana
- Department of Vascular Surgery, University Hospital of Larissa, Faculty of Medicine, School of Health Sciences, University of Thessaly, Larissa, Greece
| | - Konstantinos Dakis
- Department of Vascular Surgery, University Hospital of Larissa, Faculty of Medicine, School of Health Sciences, University of Thessaly, Larissa, Greece
| | - Konstantinos Spanos
- Department of Vascular Surgery, University Hospital of Larissa, Faculty of Medicine, School of Health Sciences, University of Thessaly, Larissa, Greece
| | - Vasiliki Tsolaki
- Intensive Care Unit, University Hospital of Larissa, Faculty of Medicine, School of Health Sciences, University of Thessaly, Larissa, Greece
| | - Nikitas Karavidas
- Intensive Care Unit, University Hospital of Larissa, Faculty of Medicine, School of Health Sciences, University of Thessaly, Larissa, Greece
| | - George Zakynthinos
- Intensive Care Unit, University Hospital of Larissa, Faculty of Medicine, School of Health Sciences, University of Thessaly, Larissa, Greece
| | - George Kouvelos
- Department of Vascular Surgery, University Hospital of Larissa, Faculty of Medicine, School of Health Sciences, University of Thessaly, Larissa, Greece
| | - Athanasios Giannoukas
- Department of Vascular Surgery, University Hospital of Larissa, Faculty of Medicine, School of Health Sciences, University of Thessaly, Larissa, Greece
| | - Miltiadis Matsagkas
- Department of Vascular Surgery, University Hospital of Larissa, Faculty of Medicine, School of Health Sciences, University of Thessaly, Larissa, Greece
| |
Collapse
|
44
|
Kumar H, Gupta R. Neuroinvasion of severe acute respiratory syndrome corona virus-2 (SARS-CoV-2): future risk of epilepsy. Int J Neurosci 2024; 134:323-332. [PMID: 35815479 DOI: 10.1080/00207454.2022.2100784] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Revised: 06/24/2022] [Accepted: 06/30/2022] [Indexed: 10/17/2022]
Abstract
Central nervous system (CNS) infection is one of the important risk factors for epilepsy. COVID-19 pandemic, which is caused by severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) has primarily been considered to involve respiratory system only, but it can also affect the CNS. A wide range of neurological manifestations have been reported in SARS-CoV-2 infected patients including seizures, status epilepticus, stroke, which are considered as important risk factors for the development of epilepsy. In post-mortem, brain tissue samples of COVID-19 patients have shown neuropathological changes and presence of SARS-CoV-2 RNA and viral proteins. In this review, mechanisms of SARS-CoV-2 neuroinvasion like neuronal retrograde trans-synaptic route and vascular route are described along with important neurological manifestations in COVID-19 patients such as seizures and cerebrovascular diseases, which have been found to be associated with the development of epilepsy. Hence, an increased risk of future burden of epilepsy in susceptible COVID-19 survivors has been proposed and preventive measures are suggested. The present review highlights about the possible association between neurological manifestations and future risk of epilepsy in COVID-19 patients.
Collapse
Affiliation(s)
- Hemant Kumar
- Department of Pharmacology, University College of Medical Sciences, University of Delhi, Delhi, India
| | - Rachna Gupta
- Department of Pharmacology, University College of Medical Sciences, University of Delhi, Delhi, India
| |
Collapse
|
45
|
Yamashita Y, Yachi S, Takeyama M, Nishimoto Y, Tsujino I, Nakamura J, Yamamoto N, Nakata H, Ikeda S, Umetsu M, Aikawa S, Hayashi H, Satokawa H, Okuno Y, Iwata E, Ogihara Y, Ikeda N, Kondo A, Iwai T, Yamada N, Ogawa T, Kobayashi T, Mo M, on behalf of the Clot-COVID Study Investigators. Prophylactic Anticoagulation and Thrombosis in Hospitalized Patients with Clinically Stable COVID-19 at Admission: From the Practice-Based Observational Study. Ann Vasc Dis 2024; 17:1-8. [PMID: 38628927 PMCID: PMC11018098 DOI: 10.3400/avd.oa.23-00031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2023] [Accepted: 10/29/2023] [Indexed: 04/19/2024] Open
Abstract
Objectives: The potential benefit of routine prophylactic anticoagulation for all hospitalized patients with clinically stable coronavirus disease 2019 (COVID-19) is still controversial. Method: The CLOT-COVID Study was a multicenter observational study enrolling 2894 consecutive hospitalized patients with COVID-19. The current study population consisted of 1738 hospitalized patients with mild COVID-19 at admission not requiring oxygen administration, who were divided into 2 groups: patients with prophylactic anticoagulation (n = 326) and those without (n = 1412). Results: Patients with prophylactic anticoagulation had more severe status of the worst severity of COVID-19 during hospitalization compared with those without (mild: 38% versus 82%, moderate: 55% versus 17%, and severe or death at discharge: 6.4% versus 0.7%, P <0.001). During hospitalization, 8 patients (0.5%) developed thrombosis, and the incidences of thrombosis were numerically higher in patients with more severe status of worst severity of COVID-19 during hospitalization (mild: 0.2%, moderate: 1.2%, and severe or death at discharge: 3.2%). Conclusions: Among hospitalized patients with clinically stable COVID-19 at admission, patients who did not worsen in COVID-19 severity after admission rarely developed thrombosis, although patients with worsening of COVID-19 severity after admission more often received prophylactic anticoagulation and might have a higher risk of thrombosis.
Collapse
Affiliation(s)
- Yugo Yamashita
- Department of Cardiovascular Medicine, Kyoto University Hospital, Kyoto, Kyoto, Japan
| | - Sen Yachi
- Japan Community Health Care Organization Tokyo Shinjuku Medical Center, Tokyo, Japan
| | - Makoto Takeyama
- Japan Community Health Care Organization Tokyo Shinjuku Medical Center, Tokyo, Japan
| | - Yuji Nishimoto
- Hyogo Prefectural Amagasaki General Medical Center, Amagasaki, Hyogo, Japan
| | | | | | | | - Hiroko Nakata
- Yokosuka General Hospital Uwamachi, Yokosuka, Kanagawa, Japan
| | - Satoshi Ikeda
- Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Nagasaki, Japan
| | | | - Shizu Aikawa
- Tsukuba Medical Center Hospital, Tsukuba, Ibaraki, Japan
| | - Hiroya Hayashi
- Osaka Metropolitan University Graduate School of Medicine, Suita, Osaka, Japan
| | - Hirono Satokawa
- Fukushima Medical University, School of Medicine, Fukushima, Fukushima, Japan
| | - Yoshinori Okuno
- Department of Cardiovascular Medicine, Kyoto University Hospital, Kyoto, Kyoto, Japan
| | - Eriko Iwata
- Nankai Medical Center Japan Community Health Care Organization, Saiki, Oita, Japan
| | | | | | - Akane Kondo
- Shikoku Medical Center for Children and Adults, Zentsuji, Kagawa, Japan
| | | | | | | | | | - Makoto Mo
- Yokohama Minami Kyosai Hospital, Yokohama, Kanagawa, Japan
| | - on behalf of the Clot-COVID Study Investigators
- Department of Cardiovascular Medicine, Kyoto University Hospital, Kyoto, Kyoto, Japan
- Japan Community Health Care Organization Tokyo Shinjuku Medical Center, Tokyo, Japan
- Hyogo Prefectural Amagasaki General Medical Center, Amagasaki, Hyogo, Japan
- Hokkaido University Hospital, Sapporo, Hokkaido, Japan
- Hamamatsu Medical Center, Hamamatsu, Shizuoka, Japan
- Yokosuka General Hospital Uwamachi, Yokosuka, Kanagawa, Japan
- Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Nagasaki, Japan
- Tohoku University Hospital, Sendai, Miyagi, Japan
- Tsukuba Medical Center Hospital, Tsukuba, Ibaraki, Japan
- Osaka Metropolitan University Graduate School of Medicine, Suita, Osaka, Japan
- Fukushima Medical University, School of Medicine, Fukushima, Fukushima, Japan
- Nankai Medical Center Japan Community Health Care Organization, Saiki, Oita, Japan
- Mie University Hospital, Tsu, Mie, Japan
- Toho University Ohashi Medical Center, Tokyo, Japan
- Shikoku Medical Center for Children and Adults, Zentsuji, Kagawa, Japan
- Tsukuba Vascular Center, Moriya, Ibaraki, Japan
- Kuwana City Medical Center, Kuwana, Mie, Japan
- Fukushima Daiich Hospital, Fukushima, Fukushima, Japan
- Yokohama Minami Kyosai Hospital, Yokohama, Kanagawa, Japan
| |
Collapse
|
46
|
Franco-Moreno A, Palma-Huerta E, Fernández-Vidal E, Madroñal-Cerezo E, Marco-Martínez J, Romero-Pareja R, Izquierdo-Martínez A, Carpintero-García L, Ruiz-Giardín JM, Torres-Macho J, de Ancos-Aracil CL. External validation of the CHEDDAR score for suspected pulmonary embolism in patients with SARS-CoV-2 infection in an independent cohort. J Thromb Thrombolysis 2024; 57:352-357. [PMID: 38095742 DOI: 10.1007/s11239-023-02918-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 10/30/2023] [Indexed: 03/26/2024]
Abstract
The accuracy of the classic scores that help stratify the pretest clinical probability of pulmonary embolism (PE) in SARS-CoV-2 infection (COVID-19) is low. Therefore, to estimate the risk of PE in these patients, a new set of guidelines must be established. The recently published CHEDDAR score proposes a new diagnostic strategy to reduce the use of computed tomography pulmonary angiography (CTPA) in non-critically ill SARS-COV-2 patients with suspected PE. According to the nomogram, patients are segregated into low-risk (< 182 points) or high-risk (≥ 182 points) based on the best cut-off value to discard PE in the original cohort. We aimed to externally validate this diagnostic strategy in an independent cohort. We analyzed data from two retrospective cohorts of hospitalized non-critically ill COVID-19 patients who underwent a CTPA due to suspicion for PE. CHEDDAR score was applied. As per the CHEDDAR nomogram, patients were classified as having a low or high clinical pre-test probability. Of the 270 patients included, 69 (25.5%) had PE. Applying the CHEDDAR score, 182 (67.4%) patients could have had PE excluded without imaging. Among 58 patients classified as having high clinical pre-test probability, 39 (67.2%) had PE. Sensitivity, specificity, positive and negative predictive values, and AUC were 56%, 90%, 67%, 85%, and 0.783 (95% CI 0.71-0.85), respectively. We provide external validation of the CHEDDAR score in an independent cohort. Even though the CHEDDAR score showed good discrimination capacity, caution is required in patients classified as having low clinical pre-test probability with a D-dimer value > 3000 ng/mL, and a RALE score ≥ 4.
Collapse
Affiliation(s)
- Anabel Franco-Moreno
- Internal Medicine Department, Hospital Universitario Infanta Leonor-Virgen de la Torre, Gran Via del Este Avenue, 80, 28031, Madrid, Spain.
| | - Elena Palma-Huerta
- Internal Medicine Department, Hospital Universitario Infanta Leonor-Virgen de la Torre, Gran Via del Este Avenue, 80, 28031, Madrid, Spain
| | - Elisa Fernández-Vidal
- Internal Medicine Department, Hospital Universitario Infanta Leonor-Virgen de la Torre, Gran Via del Este Avenue, 80, 28031, Madrid, Spain
| | | | | | | | | | | | - José Manuel Ruiz-Giardín
- Internal Medicine Department, Hospital Universitario de Fuenlabrada, Madrid, Spain
- Internal Medicine Department, Hospital Universitario de Fuenlabrada, CiberInfect, Madrid, Spain
| | - Juan Torres-Macho
- Internal Medicine Department, Hospital Universitario Infanta Leonor-Virgen de la Torre, Gran Via del Este Avenue, 80, 28031, Madrid, Spain
| | | |
Collapse
|
47
|
Yan AP, Parsons C, Caplan G, Kelly DP, Duzan J, Drake E, Kumar R. Clinical decision support to enhance venous thromboembolism pharmacoprophylaxis prescribing for pediatric inpatients with COVID-19. Pediatr Blood Cancer 2024; 71:e30843. [PMID: 38173090 DOI: 10.1002/pbc.30843] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/19/2023] [Revised: 11/20/2023] [Accepted: 12/18/2023] [Indexed: 01/05/2024]
Abstract
OBJECTIVE To design and evaluate a clinical decision support (CDS) module to improve guideline concordant venous thromboembolism (VTE) pharmacoprophylaxis prescribing for pediatric inpatients with COVID-19. MATERIALS AND METHODS The proportion of patients who met our institutional clinical practice guideline's (CPG) criteria for VTE prophylaxis was compared to those who triggered a CDS alert, indicating the patient needed VTE prophylaxis, and to those who were prescribed prophylaxis pre and post the launch of a new VTE CDS module to support VTE pharmacoprophylaxis prescribing. The sensitivity, specificity, positive predictive value (PPV), negative predictive value, F1-score and accuracy of the tool were calculated for the pre- and post-intervention periods using the CPG recommendation as the gold standard. Accuracy was defined as the sum of the true positives and true negatives over the sum of the true positives, false positives, true negatives, and false negatives. Logistic regression was used to identify variables associated with correct thromboprophylaxis prescribing. RESULTS A significant increase in the proportion of patients triggering a CDS alert occurred in the post-intervention period (44.3% vs. 6.9%, p < .001); however, no reciprocal increase in VTE prophylaxis prescribing was achieved (36.6% vs. 40.9%, p = .53). The updated CDS module had an improved sensitivity (55.0% vs. 13.3%), NPV (44.9% vs. 36.3%), F1-score (66.7% vs. 23.5%), and accuracy (62.5% vs. 42.0%), but an inferior specificity (78.6% vs. 100%) and PPV (84.6% vs. 100%). DISCUSSION The updated CDS model had an improved accuracy and overall performance in correctly identifying patients requiring VTE prophylaxis. Despite an increase in correct patient identification by the CDS module, the proportion of patients receiving appropriate pharmacologic prophylaxis did not change. CONCLUSION CDS tools to support correct VTE prophylaxis prescribing need ongoing refinement and validation to maximize clinical utility.
Collapse
Affiliation(s)
- Adam Paul Yan
- Division of Hematology and Oncology, Boston Children's Hospital and Harvard Medical School, Boston, Massachusetts, USA
- Division of Hematology and Oncology, The Hospital for Sick Children and University of Toronto, Toronto, Ontario, Canada
| | - Chase Parsons
- Division of General Pediatrics, Boston Children's Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Gregory Caplan
- Boston Children's Hospital Program for Patient Safety and Quality, Boston, Massachusetts, USA
| | - Daniel P Kelly
- Division of Medical Critical Care, Boston Children's Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Julie Duzan
- Division of Hematology and Oncology, Boston Children's Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Emily Drake
- Division of Hematology and Oncology, Boston Children's Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Riten Kumar
- Division of Hematology and Oncology, Boston Children's Hospital and Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
48
|
Maliha ST, Fatemi R, Araf Y. COVID-19 and the brain: understanding the pathogenesis and consequences of neurological damage. Mol Biol Rep 2024; 51:318. [PMID: 38386201 DOI: 10.1007/s11033-024-09279-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2023] [Accepted: 01/22/2024] [Indexed: 02/23/2024]
Abstract
SARS-CoV-2 has been known remarkably since December 2019 as a strain of pathogenic coronavirus. Starting from the earlier stages of the COVID-19 pandemic until now, we have witnessed many cases of neurological damage caused by SARS-CoV-2. There are many studies and research conducted on COVID-19-positive-patients that have found brain-related abnormalities with clear neurological symptoms, ranging from simple headaches to life-threatening strokes. For treating neurological damage, knowing the actual pathway or mechanism of causing brain damage via SARS-CoV-2 is very important. For this reason, we have tried to explain the possible pathways of brain damage due to SARS-CoV-2 with mechanisms and illustrations. The SARS-CoV-2 virus enters the human body by binding to specific ACE2 receptors in the targeted cells, which are present in the glial cells and CNS neurons of the human brain. It is found that direct and indirect infections with SARS-CoV-2 in the brain result in endothelial cell death, which alters the BBB tight junctions. These probable alterations can be the reason for the excessive transmission and pathogenicity of SARS-CoV-2 in the human brain. In this precise review, we have tried to demonstrate the neurological symptoms in the case of COVID-19-positive-patients and the possible mechanisms of neurological damage, along with the treatment options for brain-related abnormalities. Knowing the transmission mechanism of SARS-CoV-2 in the human brain can assist us in generating novel treatments associated with neuroinflammation in other brain diseases.
Collapse
Affiliation(s)
- Sumaiya Tasnim Maliha
- Biotechnology Program, Department of Mathematics and Natural Sciences, School of Data and Sciences, BRAC University, Dhaka, Bangladesh
| | - Rabeya Fatemi
- Department of Genetic Engineering and Biotechnology, East West University, Dhaka, 1212, Bangladesh
| | - Yusha Araf
- Department of Biotechnology, Bangladesh Agricultural University, Mymensingh, Bangladesh.
| |
Collapse
|
49
|
Nguyen TH, Chen LY, Khan NZ, Lindenbauer A, Bui VC, Zipfel PF, Heinrich D. The Binding of the SARS-CoV-2 Spike Protein to Platelet Factor 4: A Proposed Mechanism for the Generation of Pathogenic Antibodies. Biomolecules 2024; 14:245. [PMID: 38540666 PMCID: PMC10967930 DOI: 10.3390/biom14030245] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2023] [Revised: 01/25/2024] [Accepted: 01/31/2024] [Indexed: 04/02/2024] Open
Abstract
Pathogenic platelet factor 4 (PF4) antibodies contributed to the abnormal coagulation profiles in COVID-19 and vaccinated patients. However, the mechanism of what triggers the body to produce these antibodies has not yet been clarified. Similar patterns and many comparable features between the COVID-19 virus and heparin-induced thrombocytopenia (HIT) have been reported. Previously, we identified a new mechanism of autoimmunity in HIT in which PF4-antibodies self-clustered PF4 and exposed binding epitopes for other pathogenic PF4/eparin antibodies. Here, we first proved that the SARS-CoV-2 spike protein (SP) also binds to PF4. The binding was evidenced by the increase in mass and optical intensity as observed through quartz crystal microbalance and immunosorbent assay, while the switching of the surface zeta potential caused by protein interactions and binding affinity of PF4-SP were evaluated by dynamic light scattering and isothermal spectral shift analysis. Based on our results, we proposed a mechanism for the generation of PF4 antibodies in COVID-19 patients. We further validated the changes in zeta potential and interaction affinity between PF4 and SP and found that their binding mechanism differs from ACE2-SP binding. Importantly, the PF4/SP complexes facilitate the binding of anti-PF4/Heparin antibodies. Our findings offer a fresh perspective on PF4 engagement with the SARS-CoV-2 SP, illuminating the role of PF4/SP complexes in severe thrombotic events.
Collapse
Affiliation(s)
- Thi-Huong Nguyen
- Institute for Bioprocessing and Analytical Measurement Techniques (iba), 37308 Heilbad Heiligenstadt, Germany
- Faculty of Mathematics and Natural Sciences, Technische Universität Ilmenau, 98694 Ilmenau, Germany
| | - Li-Yu Chen
- Institute for Bioprocessing and Analytical Measurement Techniques (iba), 37308 Heilbad Heiligenstadt, Germany
- Institute of Miccrobiology, Friedrich-Schiller-University, 07745 Jena, Germany
| | - Nida Zaman Khan
- Institute for Bioprocessing and Analytical Measurement Techniques (iba), 37308 Heilbad Heiligenstadt, Germany
- Faculty of Mathematics and Natural Sciences, Technische Universität Ilmenau, 98694 Ilmenau, Germany
| | - Annerose Lindenbauer
- Institute for Bioprocessing and Analytical Measurement Techniques (iba), 37308 Heilbad Heiligenstadt, Germany
| | - Van-Chien Bui
- Department of Water Supply and Wastewater Treatment, Eichsfeldwerke GmbH, 37308 Heilbad Heiligenstadt, Germany
| | - Peter F. Zipfel
- Institute of Miccrobiology, Friedrich-Schiller-University, 07745 Jena, Germany
| | - Doris Heinrich
- Institute for Bioprocessing and Analytical Measurement Techniques (iba), 37308 Heilbad Heiligenstadt, Germany
- Faculty of Mathematics and Natural Sciences, Technische Universität Ilmenau, 98694 Ilmenau, Germany
- Fraunhofer Institut für Silicatforschung, Neunerplatz, 97082 Würzburg, Germany
| |
Collapse
|
50
|
Kutlutürk I, Tokuç EÖ, Karabaş L, Rückert R, Kaya M, Karagöz A, Munk MR. How the immune response to the structural proteins of SARS-CoV-2 affects the retinal vascular endothelial cells: an immune thrombotic and/or endotheliopathy process with in silico modeling. Immunol Res 2024; 72:50-71. [PMID: 37642808 DOI: 10.1007/s12026-023-09412-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2023] [Accepted: 07/28/2023] [Indexed: 08/31/2023]
Abstract
Thrombotic events associated with SARS-CoV-2 at the vascular endothelium still remains unclear. The aim of the current study is to determine the relationship between cellular proteins on the (ocular) vascular endothelial surface and the immune thrombotic and/or endotheliopathy process elicited by SARS-CoV-2 using an in-silico modeling. The structural S (spike glycoprotein), N (nucleocapsid protein), M (membrane protein), and E (envelope protein) proteins, an accessory protein (ORF1ab) of SARS-CoV-2 and 158 cellular proteins associated with retinal vascular endothelial cell surface or structure were included in this study for comparison of three-dimensional (3D) structure and sequence. Sixty-nine of the retinal proteins were obtained from the Uniprot database. Remaining proteins not included in the database were included in the study after they were converted into 3D structures using the RaptorX web tool. Sequence and three-dimensional structure of SARS-COV-2 S, N, M, E, ORF1ab proteins and retinal vascular endothelial proteins were compared with mTM-align server. Proteins with significant similarity (score above 0.5) were validated with the TM-align web server. Immune and thrombosis-related protein-receptor interactions of similar proteins was checked with CABS-dock. We detected a high level of structural similarity between E protein and ACE, ACE2, LAT1, and TM9SF4 endothelial proteins. In addition, PECAM-1 was found to be structurally similar to ORF1ab and S protein. When we evaluated the likelihood/potential to stimulate an immune responses/a cytokine release, TLR-2 and TLR-3, which are highly susceptible to SARS-CoV2, showed a potential receptor-protein interaction with retinal vascular endothelial proteins. Our study demonstrates that SARS-CoV-2 proteins may have structural similarities with vascular endothelial proteins, and therefore, as immunological target sites, the counterpart proteins on the endothelial surface of many organs may also be secondarily affected by any immune response against SARS-CoV-2 structural proteins.
Collapse
Affiliation(s)
- Işıl Kutlutürk
- Division of Ophthalmology, Ümraniye Trn. And Rch. Hospital, Istanbul, Turkey.
| | - Ecem Önder Tokuç
- Ophthalmology Department, University of Health Science, Derince Training and Research Hospital, Izmit-Kocaeli, Turkey
| | - Levent Karabaş
- Ophthalmology Department, Kocaeli University School of Medicine, Izmit-Kocaeli, Turkey
| | | | | | - Ali Karagöz
- Koşuyolu High Specialization Education and Research Hospital, Istanbul, Turkey
| | - Marion R Munk
- Inselspital, University Hospital Bern, Bern, Switzerland
- Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
- Augenarzt-Praxisgemeinschaft Gutblick AG, Bern, Switzerland
| |
Collapse
|