1
|
Tintore C, Cuartero J, Camps-Vilaró A, Subirana, Elosua R, Marrugat J, Degano IR. Increased risk of arrhythmias, heart failure, and thrombosis in SARS-CoV-2 positive individuals persists at one year post-infection. Comput Struct Biotechnol J 2024; 24:476-483. [PMID: 39050244 PMCID: PMC11266869 DOI: 10.1016/j.csbj.2024.06.024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Revised: 06/14/2024] [Accepted: 06/18/2024] [Indexed: 07/27/2024] Open
Abstract
Risk of cardiovascular events is increased after COVID-19. However, information on cardiovascular risk trends after COVID-19 infection is lacking and estimates by sex are inconsistent. Our aim was to examine cardiovascular outcomes and mortality in a large cohort (164,346 participants) of SARS-CoV-2 positive individuals compared to non-positive individuals, stratified by sex. Data were obtained from the Spanish Health System's electronic medical records. Selected individuals were ≥ 45 years old with/without a positive SARS-CoV-2 test in the period March-May 2020. Follow-up was obtained until January 31, 2021, for cardiovascular events (angina/myocardial infarction, arrhythmias, bypass/revascularization, heart failure, peripheral artery disease, stroke/transient ischemic attack, and thrombosis), and until March 31, 2021, for mortality. Individuals were matched by propensity score. Incidence of cardiovascular events and mortality was compared with accelerated failure time models. The effect of matching and of COVID-19 severity was assessed with sensitivity analyses. In the first 3 months of follow-up, SARS-CoV-2 positive individuals had a higher risk of mortality and of all cardiovascular events. From 4-12 months, there was increased risk of mortality in SARS-CoV-2 positive individuals overall, of heart failure in SARS-CoV-2 positive females (HR= 1.26 [1.11-1.42]), and of arrhythmias and thrombosis in SARS-CoV-2 positive males (HR= 1.29 [1.14-1.47] and HR= 1.35 [1.03-1.77], respectively). When COVID-19 patients admitted to the ICU were excluded, incidence of thrombosis was similar in males regardless of positive/non-positive SARS-CoV-2 status. In the full year of follow-up, increased incidence of heart failure and of arrhythmias and thrombosis was observed in SARS-CoV-2 positive females and males, respectively.
Collapse
Affiliation(s)
- C. Tintore
- Faculty of Medicine, University of Vic-Central University of Catalonia, 08500 Vic, Spain
| | - J. Cuartero
- Department of Medicine, Universitat Autònoma de Barcelona, 08193 Barcelona, Spain
- Department of Oncology, Hospital de la Santa Creu i Sant Pau, 08041 Barcelona, Spain
| | - A. Camps-Vilaró
- Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), Instituto de Salud Carlos III, 28029 Madrid, Spain
- Registre Gironí del Cor (REGICOR) Study Group, Hospital del Mar Medical Research Institute (IMIM), 08003 Barcelona, Spain
| | - Subirana
- Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), Instituto de Salud Carlos III, 28029 Madrid, Spain
- Registre Gironí del Cor (REGICOR) Study Group, Hospital del Mar Medical Research Institute (IMIM), 08003 Barcelona, Spain
| | - R. Elosua
- Faculty of Medicine, University of Vic-Central University of Catalonia, 08500 Vic, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), Instituto de Salud Carlos III, 28029 Madrid, Spain
- Cardiovascular Epidemiology and Genetics Research Group, IMIM, 08003 Barcelona, Spain
| | - J. Marrugat
- Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), Instituto de Salud Carlos III, 28029 Madrid, Spain
- Registre Gironí del Cor (REGICOR) Study Group, Hospital del Mar Medical Research Institute (IMIM), 08003 Barcelona, Spain
| | - IR Degano
- Faculty of Medicine, University of Vic-Central University of Catalonia, 08500 Vic, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), Instituto de Salud Carlos III, 28029 Madrid, Spain
- Registre Gironí del Cor (REGICOR) Study Group, Hospital del Mar Medical Research Institute (IMIM), 08003 Barcelona, Spain
- Institute for Research and Innovation in Life Sciences and Health in Central Catalonia (IRIS-CC), 08500 Vic, Spain
| |
Collapse
|
2
|
O’Carroll A, Richard SA, Byrne C, Rusiecki J, Wier B, Berjohn CM, Fries AC, Lalani T, Smith AG, Mody RM, Ganesan A, Huprikar N, Colombo RE, Schofield C, Lindholm DA, Mende K, Jones MU, Flanagan R, Larson DT, Ewers EC, Saunders D, Maves RC, Maldonado CJ, Sanchez Edwards M, O’Connell RJ, Simons MP, Tribble DR, Agan BK, Burgess TH, Pollett SD. Estimating the Effect of Coronavirus Disease 2019 (COVID-19) Vaccination and Infection Variant on Post-COVID-19 Venous Thrombosis or Embolism Risk. Open Forum Infect Dis 2024; 11:ofae557. [PMID: 39494453 PMCID: PMC11530954 DOI: 10.1093/ofid/ofae557] [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: 04/01/2024] [Accepted: 09/19/2024] [Indexed: 11/05/2024] Open
Abstract
Background Previous research has shown that vaccination reduces risk of post-coronavirus disease 2019 (COVID-19) venous thrombosis or embolism (VTE), but the effect of vaccine boosting on post-COVID-19 VTE risk reduction is unclear. We sought to estimate the effect of COVID-19 vaccination on the risk of post-COVID-19 VTE and to examine if the magnitude of this association differed among variant eras. Methods We performed a case-control study of Military Health System (MHS) beneficiaries who tested positive for severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) in 2020-2022. Cases were defined as those with medically attended VTE within 90 days after their first positive SARS-CoV-2 test; controls were defined as SARS-CoV-2 infections without incident VTE by 90 days. Multivariate logistic regression estimated the odds of post-SARS-CoV-2 VTE based on pre-COVID-19 vaccine status, adjusting for other VTE risk factors. Results A total of 4646 MHS beneficiaries were included in this analysis; 1370 received a primary vaccine series and a further 790 received at least 1 booster at time of infection; 71 had VTE within 90 days of SARS-CoV-2 infection. Those who were vaccinated had lower odds of VTE (adjusted odds ratio [95% confidence interval]) compared to the unvaccinated following infection (primary series: 0.28 [.13-.62]; booster dose: 0.06 [.01-.46]). Post-COVID-19 VTE risk was lowest during the Omicron era, but VTEs were too rare to examine for an interaction of variant era and vaccine effect. Conclusions Among MHS beneficiaries, COVID-19 vaccination was associated with a reduced risk of post-COVID-19 VTE diagnosis; estimated risk reduction was larger among those who received a booster.
Collapse
Affiliation(s)
- Andrew O’Carroll
- Department of Preventive Medicine, Uniformed Services University of the Health Sciences, Bethesda, Maryland, USA
| | - Stephanie A Richard
- Infectious Disease Clinical Research Program, Department of Preventive Medicine and Biostatistics, Uniformed Services University of the Health Sciences, Bethesda, Maryland, USA
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc, Bethesda, Maryland, USA
| | - Celia Byrne
- Department of Preventive Medicine, Uniformed Services University of the Health Sciences, Bethesda, Maryland, USA
| | - Jennifer Rusiecki
- Department of Preventive Medicine, Uniformed Services University of the Health Sciences, Bethesda, Maryland, USA
| | - Ben Wier
- Department of Preventive Medicine, Uniformed Services University of the Health Sciences, Bethesda, Maryland, USA
| | - Catherine M Berjohn
- Infectious Disease Clinical Research Program, Department of Preventive Medicine and Biostatistics, Uniformed Services University of the Health Sciences, Bethesda, Maryland, USA
- Naval Medical Center San Diego, San Diego, California, USA
- Department of Medicine, Uniformed Services University of the Health Sciences, Bethesda, Maryland, USA
| | - Anthony C Fries
- US Air Force School of Aerospace Medicine, Wright-Patterson, Ohio, USA
| | - Tahaniyat Lalani
- Infectious Disease Clinical Research Program, Department of Preventive Medicine and Biostatistics, Uniformed Services University of the Health Sciences, Bethesda, Maryland, USA
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc, Bethesda, Maryland, USA
- Naval Medical Center Portsmouth, Portsmouth, Virginia, USA
| | - Alfred G Smith
- Naval Medical Center Portsmouth, Portsmouth, Virginia, USA
| | - Rupal M Mody
- William Beaumont Army Medical Center, El Paso, Texas, USA
| | - Anuradha Ganesan
- Infectious Disease Clinical Research Program, Department of Preventive Medicine and Biostatistics, Uniformed Services University of the Health Sciences, Bethesda, Maryland, USA
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc, Bethesda, Maryland, USA
- Walter Reed National Military Medical Center, Bethesda, Maryland, USA
| | - Nikhil Huprikar
- Walter Reed National Military Medical Center, Bethesda, Maryland, USA
| | - Rhonda E Colombo
- Infectious Disease Clinical Research Program, Department of Preventive Medicine and Biostatistics, Uniformed Services University of the Health Sciences, Bethesda, Maryland, USA
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc, Bethesda, Maryland, USA
- Department of Medicine, Uniformed Services University of the Health Sciences, Bethesda, Maryland, USA
- Madigan Army Medical Center, Joint Base Lewis McChord, Washington, USA
| | | | - David A Lindholm
- Department of Medicine, Uniformed Services University of the Health Sciences, Bethesda, Maryland, USA
- Brooke Army Medical Center, Joint Base San Antonio–Fort Sam Houston, Houston, Texas, USA
| | - Katrin Mende
- Infectious Disease Clinical Research Program, Department of Preventive Medicine and Biostatistics, Uniformed Services University of the Health Sciences, Bethesda, Maryland, USA
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc, Bethesda, Maryland, USA
- Brooke Army Medical Center, Joint Base San Antonio–Fort Sam Houston, Houston, Texas, USA
| | - Milissa U Jones
- Department of Pediatrics, Uniformed Services University of the Health Sciences, Bethesda, Maryland, USA
| | - Ryan Flanagan
- Department of Pediatrics, Uniformed Services University of the Health Sciences, Bethesda, Maryland, USA
- Tripler Army Medical Center, Honolulu, Hawaii, USA
| | - Derek T Larson
- Department of Medicine, Uniformed Services University of the Health Sciences, Bethesda, Maryland, USA
- Alexander T. Augusta Military Medical Center, Virginia, USA
| | - Evan C Ewers
- Department of Medicine, Uniformed Services University of the Health Sciences, Bethesda, Maryland, USA
- Alexander T. Augusta Military Medical Center, Virginia, USA
| | - David Saunders
- Department of Medicine, Uniformed Services University of the Health Sciences, Bethesda, Maryland, USA
| | - Ryan C Maves
- Infectious Disease Clinical Research Program, Department of Preventive Medicine and Biostatistics, Uniformed Services University of the Health Sciences, Bethesda, Maryland, USA
- Department of Medicine, Uniformed Services University of the Health Sciences, Bethesda, Maryland, USA
- Section for Infectious Diseases, Wake Forest University School of Medicine, Winston-Salem, North Carolina, USA
| | | | - Margaret Sanchez Edwards
- Infectious Disease Clinical Research Program, Department of Preventive Medicine and Biostatistics, Uniformed Services University of the Health Sciences, Bethesda, Maryland, USA
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc, Bethesda, Maryland, USA
| | - Robert J O’Connell
- Infectious Disease Clinical Research Program, Department of Preventive Medicine and Biostatistics, Uniformed Services University of the Health Sciences, Bethesda, Maryland, USA
| | - Mark P Simons
- Infectious Disease Clinical Research Program, Department of Preventive Medicine and Biostatistics, Uniformed Services University of the Health Sciences, Bethesda, Maryland, USA
| | - David R Tribble
- Infectious Disease Clinical Research Program, Department of Preventive Medicine and Biostatistics, Uniformed Services University of the Health Sciences, Bethesda, Maryland, USA
| | - Brian K Agan
- Infectious Disease Clinical Research Program, Department of Preventive Medicine and Biostatistics, Uniformed Services University of the Health Sciences, Bethesda, Maryland, USA
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc, Bethesda, Maryland, USA
| | - Timothy H Burgess
- Infectious Disease Clinical Research Program, Department of Preventive Medicine and Biostatistics, Uniformed Services University of the Health Sciences, Bethesda, Maryland, USA
| | - Simon D Pollett
- Infectious Disease Clinical Research Program, Department of Preventive Medicine and Biostatistics, Uniformed Services University of the Health Sciences, Bethesda, Maryland, USA
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc, Bethesda, Maryland, USA
| |
Collapse
|
3
|
Wick KD, Siegel L, Oldmixon C, Lundgren JD, Thompson BT, Jones C, Leroux C, Matthay MA. Longitudinal importance of the soluble receptor for advanced glycation end-products in nonintubated hospitalized patients with COVID-19 pneumonia. Am J Physiol Lung Cell Mol Physiol 2024; 327:L607-L614. [PMID: 39076084 PMCID: PMC11563646 DOI: 10.1152/ajplung.00350.2023] [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: 11/14/2023] [Revised: 07/10/2024] [Accepted: 07/11/2024] [Indexed: 07/31/2024] Open
Abstract
The soluble receptor for advanced glycation end-products (sRAGE) is a marker of alveolar type I cell injury associated with outcomes in COVID-19 pneumonia. How plasma sRAGE changes over time and whether it remains associated with long-term clinical outcomes beyond a single measurement in COVID-19 have not been well studied. We studied two cohorts in randomized clinical trials of monoclonal antibody treatment for COVID-19 (bamlanivimab and tixagevimab/cilgavimab). We first studied the association between baseline plasma sRAGE and 90-day clinical outcomes, which had been previously demonstrated in the bamlanivimab cohort, among hospitalized patients with COVID-19 supported with high-flow nasal oxygen (HFNO) or noninvasive ventilation (NIV) in the tixagevimab/cilgavimab study. Next, we investigated the relationship between day 3 sRAGE and 90-day outcomes and how plasma sRAGE changes over the first 3 days of hospitalization in both clinical trial cohorts. We found that plasma sRAGE in the highest quartile in the HFNO/NIV participants in the tixagevimab/cilgavimab trial was associated with a significantly lower rate of 90-day sustained recovery [recovery rate ratio = 0.31, 95% confidence interval (CI) = 0.14-0.71, P = 0.005] and with a significantly higher rate of 90-day mortality (hazard ratio = 2.49, 95% CI = 1.15-5.43, P = 0.021) compared with the lower three quartiles. Day 3 plasma sRAGE in both clinical trial cohorts remained associated with 90-day clinical outcomes. The trajectory of sRAGE was not influenced by treatment assignment. Our results indicate that plasma sRAGE is a valuable prognostic marker in COVID-19 up to 3 days after initial hospital presentation.NEW & NOTEWORTHY The soluble receptor for advanced glycation end-products (sRAGE) is a marker of alveolar type I epithelial cell injury associated with clinical outcomes in acute respiratory distress syndrome and, more recently, in hospitalized subjects with COVID-19. How plasma sRAGE changes over time and whether plasma sRAGE remains associated with long-term clinical outcomes beyond a single baseline measurement in patients with COVID-19 have not been well studied.
Collapse
Affiliation(s)
- Katherine D Wick
- Cardiovascular Research Institute, University of California, San Francisco, California, United States
| | - Lianne Siegel
- Division of Biostatistics and Health Data Science, University of Minnesota, Minneapolis, Minnesota, United States
| | - Cathryn Oldmixon
- Biostatistics Center, Massachusetts General Hospital, Boston, Massachusetts, United States
| | - Jens D Lundgren
- CHIP (Centre of Excellence for Health, Immunity and Infections), Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - B Taylor Thompson
- Division of Pulmonary and Critical Care Medicine, Massachusetts General Hospital, Boston, Massachusetts, United States
| | - Chayse Jones
- Cardiovascular Research Institute, University of California, San Francisco, California, United States
| | - Carolyn Leroux
- Cardiovascular Research Institute, University of California, San Francisco, California, United States
| | - Michael A Matthay
- Cardiovascular Research Institute, University of California, San Francisco, California, United States
- Department of Medicine and Anesthesia, University of California, San Francisco, California, United States
| |
Collapse
|
4
|
de Almeida IM, Tosta BR, Pena LDC, Silva HDS, Reis-Goes FS, Silva NN, Cruz JVA, Silva MDA, de Araújo JF, Rodrigues JL, Oliveira G, Figueiredo RG, Vaz SN, Montaño-Castellón I, Santana D, Torres A, Beltrão FEDL, Carneiro VL, Campos GS, Brites C, Fortuna V, Figueiredo CA, Trindade SC, Ramos HE, Costa RDS. Genetic signatures of AKT1 variants associated with worse COVID-19 outcomes - a multicentric observational study. Front Immunol 2024; 15:1422349. [PMID: 39439795 PMCID: PMC11493623 DOI: 10.3389/fimmu.2024.1422349] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Accepted: 09/18/2024] [Indexed: 10/25/2024] Open
Abstract
Introduction The COVID-19, triggered by the SARS-CoV-2 virus, has varied clinical manifestations, ranging from mild cases to severe forms such as fatal pneumonia and acute respiratory distress syndrome (ARDS). Disease severity is influenced by an exacerbated immune response, characterized by high pro-inflammatory cytokine levels. Inhibition of AKT can potentially suppress pathological inflammation, cytokine storm and platelet activation associated with COVID-19. In this study, we aimed to investigate the rs2494746 and rs1130214 variants in the AKT1 gene associated with severe COVID-19 outcomes. Methods Peripheral blood samples and sociodemographic data from 508 individuals with COVID-19, measuring plasma cytokine concentrations using ELISA and genotyped the AKT1 variants. Results The rs2494746-C allele was associated with severity, ICU admission, and death from COVID-19. The C allele at rs1130214 was linked to increased TNF and D-dimer levels. Moreover, both variants exhibited an increased cumulative risk of disease severity, ICU admission, and mortality caused by COVID-19. In the predictive analysis, the rs2494746 obtained an accuracy of 71%, suggesting a high probability of the test determining the severity of the disease. Discussion Our findings contribute to understanding the influence of the AKT1 gene variants on the immunological damage in individuals infected with SARS-CoV-2.
Collapse
Affiliation(s)
- Ingrid Marins de Almeida
- Laboratório de Imunofarmacologia e Biologia Molecular, Instituto de Ciências da Saúde, Universidade Federal da Bahia, Salvador, Brazil
| | - Bruna Ramos Tosta
- Laboratório de Imunofarmacologia e Biologia Molecular, Instituto de Ciências da Saúde, Universidade Federal da Bahia, Salvador, Brazil
| | - Laiane da Cruz Pena
- Laboratório de Imunofarmacologia e Biologia Molecular, Instituto de Ciências da Saúde, Universidade Federal da Bahia, Salvador, Brazil
| | - Hatilla dos Santos Silva
- Laboratório de Imunofarmacologia e Biologia Molecular, Instituto de Ciências da Saúde, Universidade Federal da Bahia, Salvador, Brazil
| | - Fabiane S. Reis-Goes
- Laboratório de Imunologia e Biologia Molecular, Instituto de Ciências da Saúde, Universidade Federal da Bahia, Salvador, Brazil
| | - Nívia N. Silva
- Laboratório de Imunologia e Biologia Molecular, Instituto de Ciências da Saúde, Universidade Federal da Bahia, Salvador, Brazil
| | - João Victor Andrade Cruz
- Laboratório de Imunofarmacologia e Biologia Molecular, Instituto de Ciências da Saúde, Universidade Federal da Bahia, Salvador, Brazil
| | - Mailane dos Anjos Silva
- Laboratório de Imunofarmacologia e Biologia Molecular, Instituto de Ciências da Saúde, Universidade Federal da Bahia, Salvador, Brazil
| | - Jéssica Francisco de Araújo
- Laboratório de Imunofarmacologia e Biologia Molecular, Instituto de Ciências da Saúde, Universidade Federal da Bahia, Salvador, Brazil
| | - Juliana Lopes Rodrigues
- Laboratório de Imunofarmacologia e Biologia Molecular, Instituto de Ciências da Saúde, Universidade Federal da Bahia, Salvador, Brazil
| | - Gabriella Oliveira
- Laboratório de Análises Clínicas, Instituto Couto Maia, Salvador, Brazil
| | | | - Sara Nunes Vaz
- Hospital Universitário Professor Edgard Santos, Universidade Federal da Bahia, Salvador, Brazil
| | - Iris Montaño-Castellón
- Hospital Universitário Professor Edgard Santos, Universidade Federal da Bahia, Salvador, Brazil
| | - Daniele Santana
- Hospital Universitário Professor Edgard Santos, Universidade Federal da Bahia, Salvador, Brazil
| | - Alex Torres
- Laboratório de Imunologia e Biologia Molecular, Instituto de Ciências da Saúde, Universidade Federal da Bahia, Salvador, Brazil
| | | | | | - Gubio Soares Campos
- Laboratório de Virologia, Instituto de Ciências da Saúde, Universidade Federal da Bahia, Salvador, Brazil
| | - Carlos Brites
- Hospital Universitário Professor Edgard Santos, Universidade Federal da Bahia, Salvador, Brazil
| | - Vitor Fortuna
- Laboratório de Imunologia e Biologia Molecular, Instituto de Ciências da Saúde, Universidade Federal da Bahia, Salvador, Brazil
| | - Camila Alexandrina Figueiredo
- Laboratório de Imunofarmacologia e Biologia Molecular, Instituto de Ciências da Saúde, Universidade Federal da Bahia, Salvador, Brazil
| | - Soraya Castro Trindade
- Laboratório de Imunologia e Biologia Molecular, Instituto de Ciências da Saúde, Universidade Federal da Bahia, Salvador, Brazil
- Universidade Estadual de Feira de Santana, Feira de Santana, Brazil
| | - Helton Estrela Ramos
- Programa de Pós-Graduação em Processos Interativos de Órgãos e Sistema, Instituto de Saúde e Ciência, Universidade Federal da Bahia, Salvador, Brazil
| | - Ryan dos Santos Costa
- Laboratório de Imunofarmacologia e Biologia Molecular, Instituto de Ciências da Saúde, Universidade Federal da Bahia, Salvador, Brazil
| |
Collapse
|
5
|
Oliveira SD, Almodóvar S, Butrous G, De Jesus Perez V, Fabro A, Graham BB, Mocumbi A, Nyasulu PS, Tura‐Ceide O, Oliveira RKF, Dhillon NK. Infection and pulmonary vascular diseases consortium: United against a global health challenge. Pulm Circ 2024; 14:e70003. [PMID: 39534510 PMCID: PMC11555293 DOI: 10.1002/pul2.70003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Revised: 09/16/2024] [Accepted: 10/06/2024] [Indexed: 11/16/2024] Open
Abstract
Leveraging the potential of virtual platforms in the post-COVID-19 era, the Infection and Pulmonary Vascular Diseases Consortium (iPVDc), with the support of the Pulmonary Vascular Research Institute (PVRI), launched a globally accessible educational program to highlight top-notch research on inflammation and infectious diseases affecting the lung vasculature. This innovative virtual series has already successfully brought together distinguished investigators across five continents - Asia, Europe, South and North America, and Africa. Moreover, these open global forums have contributed to a comprehensive understanding of the complex interplay among immunology, inflammation, infection, and cardiopulmonary health, especially concerning pulmonary hypertension and related pulmonary disorders. These enlightening discussions have not only heightened awareness about the impact of various pathogenic microorganisms, including fungi, parasites, and viruses, on the onset and development of pulmonary vascular diseases but have also cast a spotlight on co-infections and neglected illnesses like schistosomiasis - a disease that continues to impose a heavy socioeconomic burden in numerous regions worldwide. Thus, the overall goal of this review article is to present the most recent breakthroughs from infectious PVDs as well as bring to light the scientific and educational insights from the 2023 iPVDc/PVRI virtual symposium series, shaping our understanding of these crucial health issues in this more than ever interconnected world.
Collapse
Affiliation(s)
- S. D. Oliveira
- Vascular Immunobiology Lab, Department of Anesthesiology, Department of Physiology and Biophysics, College of MedicineUniversity of Illinois ChicagoChicagoIllinoisUSA
| | - S. Almodóvar
- Department of Immunology & Molecular MicrobiologyTexas Tech University Health Sciences Center, School of MedicineLubbockTexasUSA
| | - G. Butrous
- Medway School of PharmacyUniversity of KentMedwayKentUnited Kingdom
| | - V De Jesus Perez
- Division of Pulmonary and Critical CareStanford UniversityPalo AltoCaliforniaUSA
| | - A. Fabro
- Division of Respiratory DiseasesFederal University of São PauloSao PauloBrazil
- Department of Pathology and Forensic Medicine, Ribeirão Preto Medical SchoolUniversidade de São PauloRibeirão PretoBrazil
| | - B. B. Graham
- Department of Medicine, Zuckerberg San Francisco General HospitalUniversity of California San FranciscoSan FranciscoCaliforniaUSA
| | - A. Mocumbi
- Department of MedicineUniversidade Eduardo MondlaneMaputoMozambique
- Division of Determinants of Chronic Diseases, Instituto Nacional de SaúdeVila de MarracueneMozambique
| | - P. S. Nyasulu
- Department of Global Health, Faculty of Medicine & Health SciencesStellenbosch UniversityCape TownSouth Africa
- School of Public Health, Faculty of Health SciencesUniversity of the WitwatersrandJohannesburgSouth Africa
| | - O. Tura‐Ceide
- Biomedical Research Institute‐IDIBGIGironaSpain
- Biomedical Research Networking Centre on Respiratory Diseases (CIBERES)MadridSpain
| | - R. K. F. Oliveira
- Division of Respiratory Diseases, Department of MedicineFederal University of São Paulo (Unifesp)São PauloBrazil
| | - N. K. Dhillon
- Division of Pulmonary and Critical Care Medicine, Department of Internal MedicineUniversity of Kansas Medical CenterKansas CityKansasUSA
| | | |
Collapse
|
6
|
Ahmed NJ, Amin ZA, Kheder RK, Pirot RQ, Mutalib GA, Jabbar SN. Immuno-inflammatory and organ dysfunction markers in severe COVID-19 patients. Cytokine 2024; 182:156715. [PMID: 39067395 DOI: 10.1016/j.cyto.2024.156715] [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: 04/05/2024] [Revised: 07/16/2024] [Accepted: 07/23/2024] [Indexed: 07/30/2024]
Abstract
Infection with the SARS-CoV-2 virus may induce some complications among people who experience mild to moderate respiratory illness and some of them recover without requiring special treatment. Albeit, some individuals become seriously reached risk points and require special medical attention especially older people and people who suffer from chronic diseases. Serum and whole blood samples were collected from confirmed infected persons with SARS CoV-2 by real-time PCR and the control group. All lab. Investigations were performed using Cobas 6000. Significant differences were noted between patients compared to the control group in the Mean ± SD of IL-6 (76.06 ± 7.60 vs 3.61 ± 0.296 pg/ml), Procalcitonin (0.947 ± 0.117 vs 0.061 ± 0.007 ng/ml), CRP (125.3 ± 7.560 vs 4.027 ± 0.251 mg/dl), ALT (154.8 ± 30.47 vs 49.75 ± 2.977 IU/L) and AST (70.83 ± 9.215 vs 27.23 ± 1.767) respectively. While other parameters were also showed significant differences were noted between patients compared to the control group for D-Dimmer, PT, PTT, LDH, Ferritin, WBC, Lymphocyte and Creatinine. The results reached that the effect of SARS CoV-2 and cytokine storm was clear on the body's organs through vital biomarker investigations that were performed in this study.
Collapse
Affiliation(s)
- Najat Jabbar Ahmed
- Department of Medical Laboratory Technology, Erbil Technical Health and Medical College, Erbil Polytechnic University, Erbil 44001, Iraq
| | - Zahra A Amin
- Department of Clinical Analysis, College of Pharmacy, Hawler Medical University Erbil 44001, Iraq
| | - Ramiar Kamal Kheder
- Medical Laboratory Science Department, College of Science, University of Raparin, Rania 46012, Sulaymaniyah, Iraq; Department of Medical Analysis, Faculty of Applied Science, Tishk International University, Erbil, Iraq.
| | - Rzgar Qadir Pirot
- Biology Department, College of Science, University of Raparin, Rania 46012, Sulaymaniyah, Iraq
| | - Gulstan A Mutalib
- Department of Medical Laboratory Technology, Erbil Technical Health and Medical College, Erbil Polytechnic University, Erbil 44001, Iraq
| | - Sana Najat Jabbar
- Department of Medical Laboratory Technology, Erbil Technical Health and Medical College, Erbil Polytechnic University, Erbil 44001, Iraq
| |
Collapse
|
7
|
Schultz IC, Dos Santos Pereira Andrade AC, Dubuc I, Laroche A, Allaeys I, Doré E, Bertrand N, Vallières L, Fradette J, Flamand L, Wink MR, Boilard E. Targeting Cytokines: Evaluating the Potential of Mesenchymal Stem Cell Derived Extracellular Vesicles in the Management of COVID-19. Stem Cell Rev Rep 2024:10.1007/s12015-024-10794-4. [PMID: 39340739 DOI: 10.1007/s12015-024-10794-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/24/2024] [Indexed: 09/30/2024]
Abstract
The Coronavirus Disease 2019 (COVID-19), caused by virus SARS-CoV-2, is characterized by massive inflammation and immune system imbalance. Despite the implementation of vaccination protocols, the accessibility of treatment remains uneven. Furthermore, the persistent threat of new variants underscores the urgent need for expanded research into therapeutic options for SARS-CoV-2. Mesenchymal stem cells (MSCs) are known for their immunomodulatory potential through the release of molecules into the extracellular space, either as soluble elements or carried by extracellular vesicles (EVs). The aim of this study was to evaluate the anti-inflammatory potential of EVs obtained from human adipose tissue (ASC-EVs) against SARS-CoV-2 infection. ASC-EVs were purified by size-exclusion chromatography, and co-culture assays confirmed that ASC-EVs were internalized by human lung cells and could colocalize with SARS-CoV-2 into early and late endosomes. To determine the functionality of ASC-EVs, lung cells were infected with SARS-CoV-2 in the presence of increasing concentrations of ASC-EVs, and the release of cytokines, chemokines and viruses were measured. While SARS-CoV-2 replication was significantly reduced only at the highest concentrations tested, multiplex analysis highlighted that lower concentrations of ASC-EV sufficed to prevent the production of immune modulators. Importantly, ASC-EVs did not contain detectable inflammatory cytokines, nor did they trigger inflammatory mediators, nor affect cellular viability. In conclusion, this work suggests that ASC-EVs have the potential to attenuate inflammation by decreasing the production of pro-inflammatory cytokines in lung cells following SARS-CoV-2 infection.
Collapse
Affiliation(s)
- Iago Carvalho Schultz
- Axe Maladies Infectieuses et Immunitaires, Centre de Recherche du Centre Hospitalier Universitaire de Québec-Université Laval, Québec, QC, Canada
- Departamento de Ciências Básicas da Saúde, Universidade Federal de Ciências da Saúde de Porto Alegre (UFCSPA), Porto Alegre, RS, Brazil
| | - Ana Claudia Dos Santos Pereira Andrade
- Axe Maladies Infectieuses et Immunitaires, Centre de Recherche du Centre Hospitalier Universitaire de Québec-Université Laval, Québec, QC, Canada
- Département de Microbiologie-Infectiologie et d'Immunologie, Faculté de Médecine de l'Université Laval, Québec, QC, Canada
| | - Isabelle Dubuc
- Axe Maladies Infectieuses et Immunitaires, Centre de Recherche du Centre Hospitalier Universitaire de Québec-Université Laval, Québec, QC, Canada
- Département de Microbiologie-Infectiologie et d'Immunologie, Faculté de Médecine de l'Université Laval, Québec, QC, Canada
| | - Audrée Laroche
- Axe Maladies Infectieuses et Immunitaires, Centre de Recherche du Centre Hospitalier Universitaire de Québec-Université Laval, Québec, QC, Canada
- Département de Microbiologie-Infectiologie et d'Immunologie, Faculté de Médecine de l'Université Laval, Québec, QC, Canada
| | - Isabelle Allaeys
- Axe Maladies Infectieuses et Immunitaires, Centre de Recherche du Centre Hospitalier Universitaire de Québec-Université Laval, Québec, QC, Canada
- Département de Microbiologie-Infectiologie et d'Immunologie, Faculté de Médecine de l'Université Laval, Québec, QC, Canada
| | - Etienne Doré
- Axe Maladies Infectieuses et Immunitaires, Centre de Recherche du Centre Hospitalier Universitaire de Québec-Université Laval, Québec, QC, Canada
- Département de Microbiologie-Infectiologie et d'Immunologie, Faculté de Médecine de l'Université Laval, Québec, QC, Canada
| | - Nicolas Bertrand
- Axe Endocrinologie et Néphrologie, Centre de Recherche du Centre Hospitalier Universitaire de Québec-Université Laval, Québec, QC, Canada
- Faculté de Pharmacie, Université Laval, Québec, QC, Canada
| | - Luc Vallières
- Axe Neurosciences, Centre de Recherche du Centre Hospitalier Universitaire de Québec-Université Laval, Québec, QC, Canada
| | - Julie Fradette
- Centre de recherche en organogénèse expérimentale de l'Université Laval/LOEX, Québec, QC, Canada
- Département de Chirurgie, Faculté de Médecine de l'Université Laval, Québec, QC, Canada
- Division of Regenerative Medicine, Centre de Recherche du Centre Hospitalier Universitaire de Québec-Université Laval, Québec, QC, Canada
| | - Louis Flamand
- Axe Maladies Infectieuses et Immunitaires, Centre de Recherche du Centre Hospitalier Universitaire de Québec-Université Laval, Québec, QC, Canada
- Département de Microbiologie-Infectiologie et d'Immunologie, Faculté de Médecine de l'Université Laval, Québec, QC, Canada
| | - Marcia Rosangela Wink
- Departamento de Ciências Básicas da Saúde, Universidade Federal de Ciências da Saúde de Porto Alegre (UFCSPA), Porto Alegre, RS, Brazil
| | - Eric Boilard
- Axe Maladies Infectieuses et Immunitaires, Centre de Recherche du Centre Hospitalier Universitaire de Québec-Université Laval, Québec, QC, Canada.
- Département de Microbiologie-Infectiologie et d'Immunologie, Faculté de Médecine de l'Université Laval, Québec, QC, Canada.
| |
Collapse
|
8
|
Schuermans A, Verstraete A, Lammi V, Nakanishi T, Ardissino M, Van den Eynde J, Sun BB, Georgakis MK, Van Weyenbergh J, Lewandowski AJ, Raman B, Ollila HM, Burgess S, Natarajan P, Honigberg MC, Freson K, Vanassche T, Verhamme P. WITHDRAWN: Proteogenomic analyses identify coagulation factor XI as a thromboinflammatory mediator of long COVID. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.05.17.24307553. [PMID: 38798608 PMCID: PMC11118620 DOI: 10.1101/2024.05.17.24307553] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/29/2024]
Abstract
The authors have withdrawn their manuscript due to analytical errors invalidating the main study findings. The authors of this work discovered the errors after submitting the initial version of the preprint. Therefore, the authors do not wish this work to be cited as reference for the project. If you have any questions, please contact the corresponding author.
Collapse
|
9
|
Becker RC, Tantry US, Khan M, Gurbel PA. The COVID-19 thrombus: distinguishing pathological, mechanistic, and phenotypic features and management. J Thromb Thrombolysis 2024:10.1007/s11239-024-03028-4. [PMID: 39179952 DOI: 10.1007/s11239-024-03028-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 08/01/2024] [Indexed: 08/26/2024]
Abstract
A heightened risk for thrombosis is a hallmark of COVID-19. Expansive clinical experience and medical literature have characterized small (micro) and large (macro) vessel involvement of the venous and arterial circulatory systems. Most events occur in patients with serious or critical illness in the hyperacute (first 1-2 weeks) or acute phases (2-4 weeks) of SARS-CoV-2 infection. However, thrombosis involving the venous, arterial, and microcirculatory systems has been reported in the subacute (4-8 weeks), convalescent (> 8-12 weeks) and chronic phases (> 12 weeks) among patients with mild-to-moderate illness. The purpose of the current focused review is to highlight the distinguishing clinical features, pathological components, and potential mechanisms of venous, arterial, and microvascular thrombosis in patients with COVID-19. The overarching objective is to better understand the proclivity for thrombosis, laying a solid foundation for screening and surveillance modalities, preventive strategies, and optimal patient management.
Collapse
Affiliation(s)
- Richard C Becker
- Cardiovascular Center, University of Cincinnati College of Medicine, 231 Albert Sabin Way, Cincinnati, OH, 45267, USA.
| | - Udaya S Tantry
- Sinai Center for Thrombosis Research and Drug Development, Baltimore, USA
| | - Muhammad Khan
- Division of General Internal Medicine, University of Cincinnati College of Medicine, Cincinnati, USA
| | - Paul A Gurbel
- Sinai Center for Thrombosis Research and Drug Development, Baltimore, USA
| |
Collapse
|
10
|
Cuevas-Sierra A, Chero-Sandoval L, Higuera-Gómez A, Vargas JA, Martínez-Urbistondo M, Castejón R, Martínez JA. Modulatory role of Faecalibacterium on insulin resistance and coagulation in patients with post-viral long haulers depending on adiposity. iScience 2024; 27:110450. [PMID: 39081294 PMCID: PMC11284562 DOI: 10.1016/j.isci.2024.110450] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2024] [Revised: 05/05/2024] [Accepted: 07/01/2024] [Indexed: 08/02/2024] Open
Abstract
Patients with Post-viral long hauler encompass lasting symptoms and comorbid complexities, often exacerbated in individuals with excessive body weight. The aim was to study gut microbiota in 130 patients with post-viral long hauler stratified by body mass index (BMI) and the relationship between inflammation and microbiota. Significant higher values were found for anthropometric variables and markers of glucose and dyslipidemia in individuals with higher BMI, as well as elevated levels of C-reactive protein, fibrinogen, IL-6, uric acid, and D-dimer. An interactive association showed an interplay between Faecalibacterium, D-dimer levels, and insulin resistance. This investigation showed that anthropometric, biochemical, and inflammatory variables were impaired in patients with post-viral long haulers with higher BMI. In addition, gut microbiota differences were found between groups and a modification effect on Faecalibacterium abundance regarding insulin resistance and D-dimer. These findings suggest that considering adiposity and gut microbiota structure and composition may improve personalized clinical interventions in patients with chronic inflammation.
Collapse
Affiliation(s)
- Amanda Cuevas-Sierra
- Precision Nutrition and Cardiometabolic Health, IMDEA-Food Institute (Madrid Institute for Advanced Studies), Campus of International Excellence (CEI) UAM+CSIC, 28049 Madrid, Spain
| | - Lourdes Chero-Sandoval
- Precision Nutrition and Cardiometabolic Health, IMDEA-Food Institute (Madrid Institute for Advanced Studies), Campus of International Excellence (CEI) UAM+CSIC, 28049 Madrid, Spain
- Department of Endocrinology and Nutrition of the University Clinical Hospital, University of Valladolid, 47002 Valladolid, Spain
| | - Andrea Higuera-Gómez
- Precision Nutrition and Cardiometabolic Health, IMDEA-Food Institute (Madrid Institute for Advanced Studies), Campus of International Excellence (CEI) UAM+CSIC, 28049 Madrid, Spain
| | - J. Antonio Vargas
- Internal Medicine Service of Puerta de Hierro Majadahonda University Hospital, 2822 Madrid, Spain
| | | | - Raquel Castejón
- Internal Medicine Service of Puerta de Hierro Majadahonda University Hospital, 2822 Madrid, Spain
| | - J. Alfredo Martínez
- Precision Nutrition and Cardiometabolic Health, IMDEA-Food Institute (Madrid Institute for Advanced Studies), Campus of International Excellence (CEI) UAM+CSIC, 28049 Madrid, Spain
- Centro de Medicina y Endocrinología, Universidad de Valladolid, Valladolid, Spain
- Centro de Investigación Biomédica en Red de la Fisiopatología de la Obesidad y Nutrición (CIBERobn), Instituto de Salud Carlos III, Madrid, Spain
| |
Collapse
|
11
|
Illibauer J, Clodi-Seitz T, Zoufaly A, Aberle JH, Weninger WJ, Foedinger M, Elsayad K. Diagnostic potential of blood plasma longitudinal viscosity measured using Brillouin light scattering. Proc Natl Acad Sci U S A 2024; 121:e2323016121. [PMID: 39088388 PMCID: PMC11331083 DOI: 10.1073/pnas.2323016121] [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: 12/31/2023] [Accepted: 07/01/2024] [Indexed: 08/03/2024] Open
Abstract
Blood plasma viscosity (PV) is an established biomarker for numerous diseases. Measurement of the shear PV using conventional rheological techniques is, however, time consuming and requires significant plasma volumes. Here, we show that Brillouin light scattering (BLS) and angle-resolved spectroscopy measurements of the longitudinal PV from microliter-sized plasma volumes can serve as a proxy for the shear PV measured using conventional viscometers. This is not trivial given the distinct frequency regime probed and the longitudinal viscosity, a combination of the shear and bulk viscosity, representing a unique material property on account of the latter. We demonstrate this for plasma from healthy persons and patients suffering from different severities of COVID-19 (CoV), which has been associated with an increased shear PV. We further show that the additional information contained in the BLS-measured effective longitudinal PV and its temperature scaling can provide unique insight into the chemical constituents and physical properties of plasma that can be of diagnostic value. In particular, we find that changes in the effective longitudinal viscosity are consistent with an increased suspension concentration in CoV patient samples at elevated temperatures that is correlated with disease severity and progression. This is supported by results from rapid BLS spatial-mapping, angle-resolved BLS measurements, changes in the elastic scattering, and anomalies in the temperature scaling of the shear viscosity. Finally, we introduce a compact BLS probe to rapidly perform measurements in plastic transport tubes. Our results open a broad avenue for PV diagnostics based on the high-frequency effective longitudinal PV and show that BLS can provide a means for its implementation.
Collapse
Affiliation(s)
- Jennifer Illibauer
- Division of Anatomy, Center for Anatomy & Cell Biology, Medical University of Vienna, ViennaA-1090, Austria
- Medical Imaging Cluster, Medical University of Vienna, ViennaA-1090, Austria
| | | | - Alexander Zoufaly
- Department of Medicine, Klinik Favoriten, ViennaA-1100, Austria
- Sigmund Freud Private University, ViennaA-1020, Austria
| | - Judith H. Aberle
- Center for Virology, Medical University of Vienna, ViennaA-1090, Austria
| | - Wolfgang J. Weninger
- Division of Anatomy, Center for Anatomy & Cell Biology, Medical University of Vienna, ViennaA-1090, Austria
- Medical Imaging Cluster, Medical University of Vienna, ViennaA-1090, Austria
| | - Manuela Foedinger
- Sigmund Freud Private University, ViennaA-1020, Austria
- Institute of Laboratory Diagnostics, Klinik Favoriten, ViennaA-1100, Austria
| | - Kareem Elsayad
- Division of Anatomy, Center for Anatomy & Cell Biology, Medical University of Vienna, ViennaA-1090, Austria
- Medical Imaging Cluster, Medical University of Vienna, ViennaA-1090, Austria
- Advanced Microscopy, Vienna Biocenter Core Facilities, ViennaA-1030, Austria
| |
Collapse
|
12
|
Gorog DA, Patel B. Tissue Factor Pathway Inhibitor and Interleukin-1 Receptor Levels in COVID-19. Thromb Haemost 2024; 124:721-724. [PMID: 38677277 DOI: 10.1055/a-2315-8278] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/29/2024]
Affiliation(s)
- Diana A Gorog
- Royal Brompton Campus, Faculty of Medicine, National Heart and Lung Institute, Imperial College, London, United Kingdom
- Centre for Health Services Research, Postgraduate Medical School, University of Hertfordshire, Hertfordshire, United Kingdom
| | - Brijesh Patel
- Adult Critical Care, Royal Brompton Hospital, Guy's and St Thomas' NHS Foundation Trust, London, United Kingdom
- Division of Anaesthetics, Pain Medicine and Intensive Care, Department of Surgery and Cancer, Faculty of Medicine, Imperial College London, London, United Kingdom
| |
Collapse
|
13
|
Li Z, Zeng M, Wu T, Wang Z, Sun Y, Zhang Z, Wu F, Chen Z, Fu M, Meng F. Causal Effects of COVID-19 on the Risk of Thrombosis: A Two-Sample Mendel Randomization Study. Thromb Haemost 2024; 124:709-720. [PMID: 38325400 PMCID: PMC11259497 DOI: 10.1055/a-2263-8514] [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: 07/19/2023] [Accepted: 11/17/2023] [Indexed: 02/09/2024]
Abstract
BACKGROUND Coronavirus disease 2019 (COVID-19) and thrombosis are linked, but the biomolecular mechanism is unclear. We aimed to investigate the causal relationship between COVID-19 and thrombotic biomarkers. METHODS We used two-sample Mendelian randomization (MR) to assess the effect of COVID-19 on 20 thrombotic biomarkers. We estimated causality using inverse variance weighting with multiplicative random effect, and performed sensitivity analysis using weighted median, MR-Egger regression and MR Pleiotropy Residual Sum and Outlier (MR-PRESSO) methods. All the results were examined by false discovery rate (FDR) with the Benjamin and Hochberg method for this correction to minimize false positives. We used R language for the analysis. RESULTS All COVID-19 classes showed lower levels of tissue factor pathway inhibitor (TFPI) and interleukin-1 receptor type 1 (IL-1R1). COVID-19 significantly reduced TFPI (odds ratio [OR] = 0.639, 95% confidence interval [CI]: 0.435-0.938) and IL-1R1 (OR = 0.603, 95% CI = 0.417-0.872), nearly doubling the odds. We also found that COVID-19 lowered multiple coagulation factor deficiency protein 2 and increased C-C motif chemokine 3. Hospitalized COVID-19 cases had less plasminogen activator, tissue type (tPA) and P-selectin glycoprotein ligand 1 (PSGL-1), while severe cases had higher mean platelet volume (MPV) and lower platelet count. These changes in TFPI, tPA, IL-1R1, MPV, and platelet count suggested a higher risk of thrombosis. Decreased PSGL-1 indicated a lower risk of thrombosis. CONCLUSION TFPI, IL-1R, and seven other indicators provide causal clues of the pathogenesis of COVID-19 and thrombosis. This study demonstrated that COVID-19 causally influences thrombosis at the biomolecular level.
Collapse
Affiliation(s)
- Zhengran Li
- The Second Clinical Medicine School, Southern Medical University, Guangzhou, Guangdong, China
- Department of Ophthalmology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Minghui Zeng
- Institute of Scientific Research, Southern Medical University, Guangzhou, China
| | - Tong Wu
- The First Clinical Medicine School, Southern Medical University, Guangzhou, Guangdong, China
| | - Zijin Wang
- The Second Clinical Medicine School, Southern Medical University, Guangzhou, Guangdong, China
| | - Yuxin Sun
- The Second Clinical Medicine School, Southern Medical University, Guangzhou, Guangdong, China
| | - Ziran Zhang
- The Second Clinical Medicine School, Southern Medical University, Guangzhou, Guangdong, China
| | - Fanye Wu
- The Second Clinical Medicine School, Southern Medical University, Guangzhou, Guangdong, China
| | - Zejun Chen
- The Second Clinical Medicine School, Southern Medical University, Guangzhou, Guangdong, China
| | - Min Fu
- Department of Ophthalmology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Fanke Meng
- Emergency Department, Zhujiang Hospital of Southern Medical University, Guangzhou, China
| |
Collapse
|
14
|
Farmakis IT, Konstantinides SV. COVID-19 changed the world - without changing CTEPH. Eur Respir J 2024; 64:2401467. [PMID: 39209467 DOI: 10.1183/13993003.01467-2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2024] [Accepted: 07/26/2024] [Indexed: 09/04/2024]
Affiliation(s)
- Ioannis T Farmakis
- Center for Thrombosis and Hemostasis, University Medical Center of the Johannes Gutenberg University, Mainz, Germany
- Department of Cardiology, University Medical Center of the Johannes Gutenberg University, Mainz, Germany
| | - Stavros V Konstantinides
- Center for Thrombosis and Hemostasis, University Medical Center of the Johannes Gutenberg University, Mainz, Germany
- Department of Cardiology, Democritus University of Thrace, Alexandroupolis, Greece
| |
Collapse
|
15
|
Suades R, Greco MF, Prieto P, Padró T, Devaux Y, Domingo P, Badimon L. CD66b +/CD68 + circulating extracellular vesicles, lactate dehydrogenase and neutrophil-to-lymphocyte ratio can differentiate coronavirus disease 2019 severity during and after infection. J Extracell Vesicles 2024; 13:e12456. [PMID: 39007437 PMCID: PMC11247396 DOI: 10.1002/jev2.12456] [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: 01/24/2024] [Revised: 05/07/2024] [Accepted: 05/09/2024] [Indexed: 07/16/2024] Open
Abstract
Coronavirus disease 2019 (COVID-19) has been a major public health burden. We hypothesised that circulating extracellular vesicles (cEVs), key players in health and disease, could trace the cell changes during COVID-19 infection and recovery. Therefore, we studied the temporal trend of cEV and inflammatory marker levels in plasma samples of COVID-19 patients that were collected within 24 h of patient admission (baseline, n = 80) and after hospital discharge at day-90 post-admission (n = 59). Inflammatory markers were measured by standard biochemical methods. cEVs were quantitatively and phenotypically characterized by high-sensitivity nano flow cytometry. In patients recovered from COVID-19 lower levels of inflammatory markers were detected. cEVs from vascular (endothelial cells) and blood (platelets, distinct immune subsets) cells were significantly reduced at day-90 compared to admission levels, a pattern also observed for cEVs from progenitor, perivascular and epithelial cells. The best discriminatory power for COVID-19 severity was found for inflammatory markers lactate dehydrogenase and neutrophil-to-lymphocyte ratio and for granulocyte/macrophage-released CD66b+/CD68+-cEVs. Albeit inflammatory markers were good indicators of systemic inflammatory response and discriminators of COVID-19 remission, they do not completely reveal cell stress and organ damage states. cEVs reaching baseline pre-infection levels at 90 days post-infection in recovered patients discriminate parental cells affected by disease.
Collapse
Affiliation(s)
- Rosa Suades
- Cardiovascular Program ICCCInstitut de Recerca Sant Pau (IR SANT PAU)BarcelonaSpain
- Centro de Investigación Biomédica en Red Enfermedades Cardiovasculares (CIBERCV), Institute of Health Carlos IIIMadridSpain
| | | | - Paula Prieto
- Infectious Diseases Unit, Department of Internal MedicineHospital de la Santa Creu i Sant Pau – IR SANT PAUBarcelonaSpain
| | - Teresa Padró
- Cardiovascular Program ICCCInstitut de Recerca Sant Pau (IR SANT PAU)BarcelonaSpain
- Centro de Investigación Biomédica en Red Enfermedades Cardiovasculares (CIBERCV), Institute of Health Carlos IIIMadridSpain
| | - Yvan Devaux
- Cardiovascular Research Unit, Department of Precision HealthLuxembourg Institute of HealthStrassenLuxembourg
| | - Pere Domingo
- Infectious Diseases Unit, Department of Internal MedicineHospital de la Santa Creu i Sant Pau – IR SANT PAUBarcelonaSpain
- Universitat Autònoma de Barcelona (UAB)BarcelonaSpain
| | - Lina Badimon
- Cardiovascular Program ICCCInstitut de Recerca Sant Pau (IR SANT PAU)BarcelonaSpain
- Centro de Investigación Biomédica en Red Enfermedades Cardiovasculares (CIBERCV), Institute of Health Carlos IIIMadridSpain
- Cardiovascular Research Chair, UABBarcelonaSpain
| |
Collapse
|
16
|
Shen B, Yang L, Jia X, Kong D, Jing L, Gao Y, Gao S, Chen R, Chen F, Zhao C, Li Y, Tan R, Zhao X. Contribution of platelets to disruption of the blood-brain barrier during arterial baroreflex dysfunction. Microvasc Res 2024; 154:104681. [PMID: 38493885 DOI: 10.1016/j.mvr.2024.104681] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2023] [Revised: 03/07/2024] [Accepted: 03/14/2024] [Indexed: 03/19/2024]
Abstract
BACKGROUND Arterial baroreflex dysfunction, like many other central nervous system disorders, involves disruption of the blood-brain barrier, but what causes such disruption in ABR dysfunction is unclear. Here we explored the potential role of platelets in this disruption. METHODS ABR dysfunction was induced in rats using sinoaortic denervation, and the effects on integrity of the blood-brain barrier were explored based on leakage of Evans blue or FITC-dextran, while the effects on expression of CD40L in platelets and of key proteins in microvascular endothelial cells were explored using immunohistochemistry, western blotting and enzyme-linked immunosorbent assay. Similar experiments were carried out in rat brain microvascular endothelial cell line, which we exposed to platelets taken from rats with ABR dysfunction. RESULTS Sinoaortic denervation permeabilized the blood-brain barrier and downregulated zonula occludens-1 and occludin in rat brain, while upregulating expression of CD40L on the surface of platelets and stimulating platelet aggregation. Similar effects of permeabilization and downregulation were observed in healthy rats that received platelets from animals with ABR dysfunction, and in rat brain microvascular endothelial cells, but only in the presence of lipopolysaccharide. These effects were associated with activation of NF-κB signaling and upregulation of matrix metalloprotease-9. These effects of platelets from animals with ABR dysfunction were partially blocked by neutralizing antibody against CD40L or the platelet inhibitor clopidogrel. CONCLUSION During ABR dysfunction, platelets may disrupt the blood-brain barrier when CD40L on their surface activates NF-kB signaling within cerebral microvascular endothelial cells, leading to upregulation of matrix metalloprotease-9. Our findings imply that targeting CD40L may be effective against cerebral diseases involving ABR dysfunction.
Collapse
Affiliation(s)
- Bowen Shen
- Institute of Pharmacology, Shandong First Medical University & Shandong Academy of Medical Sciences, Tai'an 271016, China
| | - Lili Yang
- Institute of Pharmacology, Shandong First Medical University & Shandong Academy of Medical Sciences, Tai'an 271016, China
| | - Xiaoli Jia
- Department of Pharmacy, Liaocheng People's Hospital Affiliated to Shandong First Medical University & Shandong Academy of Medical Sciences, Liao'cheng 252000, China
| | - Deping Kong
- Institute of Pharmacology, Shandong First Medical University & Shandong Academy of Medical Sciences, Tai'an 271016, China
| | - Lei Jing
- Department of Pharmacy, Dongping People's Hospital, Tai'an 271500, China
| | - Yongfeng Gao
- Institute of Pharmacology, Shandong First Medical University & Shandong Academy of Medical Sciences, Tai'an 271016, China
| | - Shan Gao
- Institute of Pharmacology, Shandong First Medical University & Shandong Academy of Medical Sciences, Tai'an 271016, China
| | - Ruimin Chen
- Institute of Pharmacology, Shandong First Medical University & Shandong Academy of Medical Sciences, Tai'an 271016, China
| | - Fengbao Chen
- Institute of Pharmacology, Shandong First Medical University & Shandong Academy of Medical Sciences, Tai'an 271016, China
| | - Chunyu Zhao
- Institute of Pharmacology, Shandong First Medical University & Shandong Academy of Medical Sciences, Tai'an 271016, China
| | - Yue Li
- Institute of Pharmacology, Shandong First Medical University & Shandong Academy of Medical Sciences, Tai'an 271016, China
| | - Rui Tan
- Institute of Pharmacology, Shandong First Medical University & Shandong Academy of Medical Sciences, Tai'an 271016, China.
| | - Xiaomin Zhao
- Institute of Pharmacology, Shandong First Medical University & Shandong Academy of Medical Sciences, Tai'an 271016, China.
| |
Collapse
|
17
|
Letelier P, Delgado H, Garrido F, Quiñones F, San MA, Hernández L, Garcés P, Guzmán-Oyarzo D, Boguen R, Hernandez A, Medina G, Schwerter P, Guzmán N. Dynamic changes of hematological and hemostatic parameters in COVID-19 hospitalized patients: Potential role as severity biomarkers for the Chilean population. J Med Biochem 2024; 43:556-564. [PMID: 39139154 PMCID: PMC11318854 DOI: 10.5937/jomb0-47588] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2023] [Accepted: 12/29/2023] [Indexed: 08/15/2024] Open
Abstract
Background COVID-19 is still a global health issue, there is limited evidence in South America regarding laboratory biomarkers associated with severe disease. The objective of our study was to identify hematological and hemostatic changes associated with severe COVID-19. Methods A total of 170 hospitalized patients with COVID19 were included in the study, defining their severity according to established criteria. Demographic, clinical, and laboratory (days 1, 3, 7, 15) data were obtained. We performed a statistical analysis, assuming significance with a value of p < 0.05. We analyzed the correlation between severity and biomarkers and established cut-off values for severe patients through ROC curves, estimating Odds Ratio associated with severe disease. Results Day 1 was observed significant differences between moderate vs severe patients for leukocytes (WBC), Neutrophil-lymphocyte ratio (NLR), platelet-lymphocyte ratio (PLR) and D-dimer, establishing cut-off points for each of them. The markers we found associated to risk of severe disease were WBC (OR=3.2396; p = 0.0003), NLR (OR=5.7084; p < 0.0001), PLR (OR=4.4094; p < 0.0001), Neutrophil (OR=4.1193; p < 0.0001), D-dimer (OR=2.7827; p = 0.0124). Conclusions The results allow to establish basic laboratory biomarkers associated to severe disease, which could be used as prognostic markers.
Collapse
Affiliation(s)
- Pablo Letelier
- Universidad Católica de Temuco, Facultad de Ciencias de la Salud, Departamento de Procesos Diagnósticos y Evaluación, Precision Health Research Laboratory, Temuco, Chile
| | - Hugo Delgado
- Dr. Hernán Henríquez Aravena Hospital, Clinical Laboratory, Temuco, Chile
| | - Felipe Garrido
- Dr. Hernán Henríquez Aravena Hospital, Clinical Laboratory, Temuco, Chile
| | - Francisco Quiñones
- Dr. Hernán Henríquez Aravena Hospital, Clinical Laboratory, Temuco, Chile
| | - Martín Andrés San
- Dr. Hernán Henríquez Aravena Hospital, Clinical Laboratory, Temuco, Chile
| | - Loreto Hernández
- Complejo Asistencial Padre Las Casas, Padre Las Casas, Araucanía, Chile
| | | | - Dina Guzmán-Oyarzo
- Universidad San Sebastián, Facultad de Medicina y Ciencias, School of Medical Technology, Campus Concepción, Concepción, Chile
| | - Rodrigo Boguen
- Universidad Católica de Temuco, Facultad de Ciencias de la Salud, Departamento de Procesos Diagnósticos y Evaluación, Precision Health Research Laboratory, Temuco, Chile
| | - Alfonso Hernandez
- Universidad Católica de Temuco, Facultad de Ciencias de la Salud, Departamento de Procesos Diagnósticos y Evaluación, Precision Health Research Laboratory, Temuco, Chile
| | - Gustavo Medina
- Universidad Católica de Temuco, Facultad de Ciencias de la Salud, Departamento de Procesos Diagnósticos y Evaluación, Precision Health Research Laboratory, Temuco, Chile
| | - Patricia Schwerter
- Universidad Católica de Temuco, Facultad de Ingeniería, Department of Mathematical and Physics Sciences, Temuco, Chile
| | - Neftalí Guzmán
- Universidad Católica de Temuco, Facultad de Ciencias de la Salud, Departamento de Procesos Diagnósticos y Evaluación, Precision Health Research Laboratory, Temuco, Chile
| |
Collapse
|
18
|
Zhu J, Bouzid R, Travert B, Géri G, Cohen Y, Picod A, Heming N, Rottman M, Joly-Laffargue B, Veyradier A, Capron C, Coppo P. Combined coagulation and inflammation markers as predictors of venous thrombo-embolism and death in COVID-19. Front Med (Lausanne) 2024; 11:1399335. [PMID: 38915768 PMCID: PMC11194426 DOI: 10.3389/fmed.2024.1399335] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2024] [Accepted: 05/03/2024] [Indexed: 06/26/2024] Open
Abstract
Background The COVID-19 pandemic related to SARS-CoV-2 virus was responsible for global pandemic. The severe form of the disease was linked to excessive activation of immune pathways together with a systemic cytokine storm response and thrombotic venous or arterial complications. Factors predicting severe outcomes including venous and/or pulmonary thrombosis (VT) and death were identified, but the prognostic role of their combination was not addressed extensively. Objectives We investigated the role of prognostic factors from the coagulation or inflammatory pathways to better understand the outcome of the disease. Methods For this, we prospectively studied 167 SARS-CoV-2-positive patients from admission in intensive care units (ICU) or emergency departments from four academic hospitals over a 14-month period. Besides standard biology, we assessed serum concentrations of inflammatory markers, coagulation factors and peripheral blood cells immunophenotyping. Results Thirty-nine patients (23.3%) developed VT and 30 patients (18%) died. By univariate analysis, C-reactive protein (CRP) level > 150 mg/L, interleukin-6 (IL-6) ≥ 20 pg/mL, D-dimers > 1,500 μg/L, ADAMTS13 activity ≤ 50%, Von. Conclusion A combination of coagulation and inflammatory markers can refine the prognostication of severe outcome in COVID-19, and could be useful for the initial evaluation of other types of viral infection.
Collapse
Affiliation(s)
- Jaja Zhu
- Service d’Hématologie-Immunologie-Transfusion, AP-HP Paris Saclay, CHU Ambroise Paré, Université de Versailles Saint Quentin-Université Paris Saclay, Montigny-le-Bretonneux, France
- Laboratoire Cellules Souches et Applications Thérapeutiques, UMR INSERM 1184, Commissariat à l’Energie Atomique et aux Energies Alternatives, Fontenay-aux-Roses, France
| | - Raïda Bouzid
- Centre de Référence des Microangiopathies Thrombotiques (CNR-MAT), AP-HP, Paris, France
| | - Benoît Travert
- Service de Médecine Interne, Hôpital Ambroise-Paré, AP-HP, Boulogne-Billancourt, France
| | - Guillaume Géri
- Service de Médecine Intensive et Réanimation, Hôpital Ambroise-Paré, AP-HP, Boulogne-Billancourt, France
| | - Yves Cohen
- Service de Médecine Intensive et Réanimation, Hôpital Avicenne, AP-HP, Paris, France
| | - Adrien Picod
- Service de Médecine Intensive et Réanimation, Hôpital Avicenne, AP-HP, Paris, France
| | - Nicholas Heming
- Department of Intensive Care, Raymond Poincaré Hospital, APHP University Versailles Saint Quentin-University Paris Saclay, Garches, France
- Institut Hospitalo Universitaire PROMETHEUS, Garches, France
- Innovative Biomarkers Plateform, Laboratory of Infection & Inflammation-U1173, School of Medicine, INSERM, University Versailles Saint Quentin-University Paris Saclay, Garches, France
- FHU SEPSIS, Garches, France
| | - Martin Rottman
- Innovative Biomarkers Plateform, Laboratory of Infection & Inflammation-U1173, School of Medicine, INSERM, University Versailles Saint Quentin-University Paris Saclay, Garches, France; FHU SEPSIS, Garches, France
- General Intensive Care Unit, Raymond Poincaré Hospital (AP-HP), FHU SEPSIS, Laboratory of Infection and Inflammation-U1173, School of Medicine Simone Veil, Université Versailles Saint Quentin, University Paris Saclay, INSERM, Garches, France
| | - Bérangère Joly-Laffargue
- Centre de Référence des Microangiopathies Thrombotiques (CNR-MAT), AP-HP, Paris, France
- EA3518, Institut de Recherche Saint Louis, Université de Paris, Paris, France
- Service D’hématologie Biologique, Laboratoire ADAMTS13, Hôpital Lariboisière, AP-HP Nord, Université de Paris, Paris, France
| | - Agnès Veyradier
- Centre de Référence des Microangiopathies Thrombotiques (CNR-MAT), AP-HP, Paris, France
- EA3518, Institut de Recherche Saint Louis, Université de Paris, Paris, France
- Service D’hématologie Biologique, Laboratoire ADAMTS13, Hôpital Lariboisière, AP-HP Nord, Université de Paris, Paris, France
| | - Claude Capron
- Service d’Hématologie-Immunologie-Transfusion, AP-HP Paris Saclay, CHU Ambroise Paré, Université de Versailles Saint Quentin-Université Paris Saclay, Montigny-le-Bretonneux, France
- Université Paris-Saclay, Université de Versailles Saint Quentin en Yvelines (UVSQ), Biomarqueurs en cancérologie et onco-hématologie (BECCOH), Boulogne-Billancourt, France
| | - Paul Coppo
- Centre de Référence des Microangiopathies Thrombotiques (CNR-MAT), AP-HP, Paris, France
- Service d’Hématologie, Hôpital Saint-Antoine, AP-HP-Sorbonne Université, Paris, France
- NSERM UMRS 1138, Centre de Recherche des Cordeliers, Paris, France
| |
Collapse
|
19
|
Baykara N. Clinical Characteristics, Outcomes, and Risk Factors for Mortality in Pregnant/Puerperal Women with COVID-19 Admitted to ICU in Turkey: A Multicenter, Retrospective Study from a Middle-Income Country. J Intensive Care Med 2024; 39:577-594. [PMID: 38320979 DOI: 10.1177/08850666231222838] [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: 02/08/2024]
Abstract
BACKGROUND Coronaviruses have been the cause of 3 major outbreaks during the last 2 decades. Information on coronavirus diseases in pregnant women is limited, and even less is known about seriously ill pregnant women. Data are also lacking regarding the real burden of coronavirus disease 2019 (COVID-19) infection in pregnant women from low/middle-income countries. The aim of this study was to determine the characteristics and clinical course of COVID-19 in pregnant/puerperal women admitted to ICUs in Turkey. METHODS This was a national, multicenter, retrospective study. The study population comprised all SARS-CoV-2-infected pregnant/puerperal women admitted to participating ICUs between 1 March 2020 and 1 January 2022. Data regarding demographics, comorbidities, illness severity, therapies, extrapulmonary organ injuries, non-COVID-19 infections, and maternal and fetal/neonatal outcomes were recorded. LASSO logistic regression and multiple logistic regression analyses were used to identify predictive variables in terms of ICU mortality. RESULTS A total of 597 patients (341 pregnant women, 255 puerperal women) from 59 ICUs in 44 hospitals were included and of these patients, 87.1% were unvaccinated. The primary reason for ICU admission was acute hypoxemic respiratory failure in 522 (87.4%), acute hypoxemic respiratory failure plus shock in 14 (2.3%), ischemic cerebrovascular accident (CVA) in 5 (0.8%), preeclampsia/eclampsia/HELLP syndrome in 6 (1.0%), and post-caesarean follow-up in 36 (6.0%). Nonsurvivors were sicker than survivors upon ICU admission, with higher APACHE II (p < 0.001) and SOFA scores (p < 0.001). A total of 181 (30.3%) women died and 280 (46.6%) had received invasive mechanical ventilation (IMV). Myocardial injury, the highest SOFA score during ICU stay, LDH levels on admission, the highest levels of AST during ICU stay, average daily dose of corticosteroids, IMV, prophylactic dose anticoagulation (compared with therapeutic dose anticoagulation), PaO2/FiO2 ratio <100, pulmonary embolism, and shock were identified as predictors of mortality. Rates of premature birth (46.4%), cesarean section (53.7%), fetal distress (15.3%), stillbirth (6.5%), and low birth weight (19.4%) were high. Rates of neonatal death (8%) and respiratory distress syndrome (21%) were also high among live-born infants. CONCLUSIONS Severe/critical COVID-19 infection during the pregnancy/puerperal period was associated with high maternal mortality and fetal/neonatal complication rates in Turkey.
Collapse
Affiliation(s)
- Nur Baykara
- Department of Anesthesiology, Division of Critical Care, School of Medicine, Kocaeli University, Kocaeli, Turkey
| |
Collapse
|
20
|
Qiu X, Nair MG, Jaroszewski L, Godzik A. Deciphering Abnormal Platelet Subpopulations in COVID-19, Sepsis and Systemic Lupus Erythematosus through Machine Learning and Single-Cell Transcriptomics. Int J Mol Sci 2024; 25:5941. [PMID: 38892129 PMCID: PMC11173046 DOI: 10.3390/ijms25115941] [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: 04/16/2024] [Revised: 05/22/2024] [Accepted: 05/27/2024] [Indexed: 06/21/2024] Open
Abstract
This study focuses on understanding the transcriptional heterogeneity of activated platelets and its impact on diseases such as sepsis, COVID-19, and systemic lupus erythematosus (SLE). Recognizing the limited knowledge in this area, our research aims to dissect the complex transcriptional profiles of activated platelets to aid in developing targeted therapies for abnormal and pathogenic platelet subtypes. We analyzed single-cell transcriptional profiles from 47,977 platelets derived from 413 samples of patients with these diseases, utilizing Deep Neural Network (DNN) and eXtreme Gradient Boosting (XGB) to distinguish transcriptomic signatures predictive of fatal or survival outcomes. Our approach included source data annotations and platelet markers, along with SingleR and Seurat for comprehensive profiling. Additionally, we employed Uniform Manifold Approximation and Projection (UMAP) for effective dimensionality reduction and visualization, aiding in the identification of various platelet subtypes and their relation to disease severity and patient outcomes. Our results highlighted distinct platelet subpopulations that correlate with disease severity, revealing that changes in platelet transcription patterns can intensify endotheliopathy, increasing the risk of coagulation in fatal cases. Moreover, these changes may impact lymphocyte function, indicating a more extensive role for platelets in inflammatory and immune responses. This study identifies crucial biomarkers of platelet heterogeneity in serious health conditions, paving the way for innovative therapeutic approaches targeting platelet activation, which could improve patient outcomes in diseases characterized by altered platelet function.
Collapse
Affiliation(s)
| | | | | | - Adam Godzik
- Division of Biomedical Sciences, University of California Riverside School of Medicine, Riverside, CA 92521, USA; (X.Q.); (M.G.N.); (L.J.)
| |
Collapse
|
21
|
Cernera G, Gelzo M, De Placido P, Pietroluongo E, Raia M, Scalia G, Tortora M, Formisano P, Palmieri G, Giuliano M, Castaldo G. Serum biomarkers of inflammation and vascular damage upon SARS-Cov-2 mRNA vaccine in patients with thymic epithelial tumors. Clin Chem Lab Med 2024; 62:1198-1205. [PMID: 38232092 DOI: 10.1515/cclm-2023-1283] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2023] [Accepted: 01/03/2024] [Indexed: 01/19/2024]
Abstract
OBJECTIVES Thymic epithelial tumors (TET) patients are at high risk of autoimmune and hypoimmune complications. Limited evidence is available on the potential risk of immune-related and inflammatory reactions induced by SARS-Cov-2 vaccine in this patient population. METHODS In order to identify subjects at higher risk for vaccine complications, we prospectively evaluated a panel of serum biomarkers related to inflammation (TNF-α, IL-1β, -6, -10, -12, and -17A, IFN-α, β and γ, MPO, MMP-9), and vascular damage (E- and P-selectin, VEGF-A, P-ANCA and MCP-1) in 44 TET patients and in 30 healthy controls along the whole SARS-Cov-2 vaccine cycle. RESULTS About 50 % of subjects (either TET and controls) showed an increase of serum biochemical markers of inflammation and endothelial damage with a large heterogeneity of values. Such increase appeared early, after the first dose in control subjects and later, after the second dose in TET patients (in which we observed mainly an increase of inflammatory biomarkers). The values normalized after about 3 months and did not increase after the third, booster dose. No autoimmune or vascular complications were observed in the study subjects and no difference was observed in terms of vaccine response among subjects showing serum biomarkers increase and those who experienced no changes. CONCLUSIONS Our data highlight the relevance of Sars-Cov-2 vaccine in TET patients, as it resulted safe and prevented severe COVID-19. However, further studies are awaited to explore the mechanisms and the potential consequences of the observed increase of serum inflammatory and vascular damage biomarkers.
Collapse
Affiliation(s)
- Gustavo Cernera
- Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università di Napoli Federico II, Naples, Italy
- CEINGE-Biotecnologie Avanzate, scarl, Naples, Italy
| | - Monica Gelzo
- Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università di Napoli Federico II, Naples, Italy
- CEINGE-Biotecnologie Avanzate, scarl, Naples, Italy
| | - Pietro De Placido
- Dipartimento di Medicina Clinica e Chirurgia, Università di Napoli Federico II, Naples, Italy
| | - Erica Pietroluongo
- Dipartimento di Medicina Clinica e Chirurgia, Università di Napoli Federico II, Naples, Italy
| | | | | | - Marianna Tortora
- Rare Tumors Coordinating Center of Campania Region (CRCTR), Naples, Italy
| | - Pietro Formisano
- Dipartimento di Scienze Mediche Traslazionali, Università di Napoli Federico II, Naples, Italy
| | | | - Mario Giuliano
- Dipartimento di Medicina Clinica e Chirurgia, Università di Napoli Federico II, Naples, Italy
- Rare Tumors Coordinating Center of Campania Region (CRCTR), Naples, Italy
| | - Giuseppe Castaldo
- Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università di Napoli Federico II, Naples, Italy
- CEINGE-Biotecnologie Avanzate, scarl, Naples, Italy
| |
Collapse
|
22
|
Viode A, Smolen KK, van Zalm P, Stevenson D, Jha M, Parker K, Levy O, Steen JA, Steen H. Longitudinal plasma proteomic analysis of 1117 hospitalized patients with COVID-19 identifies features associated with severity and outcomes. SCIENCE ADVANCES 2024; 10:eadl5762. [PMID: 38787940 PMCID: PMC11122669 DOI: 10.1126/sciadv.adl5762] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Accepted: 04/18/2024] [Indexed: 05/26/2024]
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection is characterized by highly heterogeneous manifestations ranging from asymptomatic cases to death for still incompletely understood reasons. As part of the IMmunoPhenotyping Assessment in a COVID-19 Cohort study, we mapped the plasma proteomes of 1117 hospitalized patients with COVID-19 from 15 hospitals across the United States. Up to six samples were collected within ~28 days of hospitalization resulting in one of the largest COVID-19 plasma proteomics cohorts with 2934 samples. Using perchloric acid to deplete the most abundant plasma proteins allowed for detecting 2910 proteins. Our findings show that increased levels of neutrophil extracellular trap and heart damage markers are associated with fatal outcomes. Our analysis also identified prognostic biomarkers for worsening severity and death. Our comprehensive longitudinal plasma proteomics study, involving 1117 participants and 2934 samples, allowed for testing the generalizability of the findings of many previous COVID-19 plasma proteomics studies using much smaller cohorts.
Collapse
Affiliation(s)
- Arthur Viode
- Department of Pathology, Boston Children’s Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Kinga K. Smolen
- Harvard Medical School, Boston, MA, USA
- Precision Vaccines Program, Boston Children’s Hospital, Boston, MA, USA
| | - Patrick van Zalm
- Department of Pathology, Boston Children’s Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
- Department of Neuropsychology and Psychopharmacology, EURON, Faculty of Psychology and Neuroscience, Maastricht University, Maastricht, Netherlands
| | - David Stevenson
- Department of Pathology, Boston Children’s Hospital, Boston, MA, USA
| | - Meenakshi Jha
- Department of Pathology, Boston Children’s Hospital, Boston, MA, USA
| | - Kenneth Parker
- Department of Pathology, Boston Children’s Hospital, Boston, MA, USA
| | - IMPACC Network‡
- Department of Pathology, Boston Children’s Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
- Precision Vaccines Program, Boston Children’s Hospital, Boston, MA, USA
- Department of Neuropsychology and Psychopharmacology, EURON, Faculty of Psychology and Neuroscience, Maastricht University, Maastricht, Netherlands
- Broad Institute of MIT & Harvard, Cambridge, MA, USA
- F. M. Kirby Neurobiology Center, Boston Children's Hospital, Boston, MA, USA
- Neurobiology Program, Boston Children's Hospital, Boston, MA, USA
| | - Ofer Levy
- Harvard Medical School, Boston, MA, USA
- Precision Vaccines Program, Boston Children’s Hospital, Boston, MA, USA
- Broad Institute of MIT & Harvard, Cambridge, MA, USA
| | - Judith A. Steen
- Harvard Medical School, Boston, MA, USA
- F. M. Kirby Neurobiology Center, Boston Children's Hospital, Boston, MA, USA
- Neurobiology Program, Boston Children's Hospital, Boston, MA, USA
| | - Hanno Steen
- Department of Pathology, Boston Children’s Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
- Precision Vaccines Program, Boston Children’s Hospital, Boston, MA, USA
- Neurobiology Program, Boston Children's Hospital, Boston, MA, USA
| |
Collapse
|
23
|
Patel MA, Daley M, Van Nynatten LR, Slessarev M, Cepinskas G, Fraser DD. A reduced proteomic signature in critically ill Covid-19 patients determined with plasma antibody micro-array and machine learning. Clin Proteomics 2024; 21:33. [PMID: 38760690 PMCID: PMC11100131 DOI: 10.1186/s12014-024-09488-3] [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: 11/09/2023] [Accepted: 05/06/2024] [Indexed: 05/19/2024] Open
Abstract
BACKGROUND COVID-19 is a complex, multi-system disease with varying severity and symptoms. Identifying changes in critically ill COVID-19 patients' proteomes enables a better understanding of markers associated with susceptibility, symptoms, and treatment. We performed plasma antibody microarray and machine learning analyses to identify novel proteins of COVID-19. METHODS A case-control study comparing the concentration of 2000 plasma proteins in age- and sex-matched COVID-19 inpatients, non-COVID-19 sepsis controls, and healthy control subjects. Machine learning was used to identify a unique proteome signature in COVID-19 patients. Protein expression was correlated with clinically relevant variables and analyzed for temporal changes over hospitalization days 1, 3, 7, and 10. Expert-curated protein expression information was analyzed with Natural language processing (NLP) to determine organ- and cell-specific expression. RESULTS Machine learning identified a 28-protein model that accurately differentiated COVID-19 patients from ICU non-COVID-19 patients (accuracy = 0.89, AUC = 1.00, F1 = 0.89) and healthy controls (accuracy = 0.89, AUC = 1.00, F1 = 0.88). An optimal nine-protein model (PF4V1, NUCB1, CrkL, SerpinD1, Fen1, GATA-4, ProSAAS, PARK7, and NET1) maintained high classification ability. Specific proteins correlated with hemoglobin, coagulation factors, hypertension, and high-flow nasal cannula intervention (P < 0.01). Time-course analysis of the 28 leading proteins demonstrated no significant temporal changes within the COVID-19 cohort. NLP analysis identified multi-system expression of the key proteins, with the digestive and nervous systems being the leading systems. CONCLUSIONS The plasma proteome of critically ill COVID-19 patients was distinguishable from that of non-COVID-19 sepsis controls and healthy control subjects. The leading 28 proteins and their subset of 9 proteins yielded accurate classification models and are expressed in multiple organ systems. The identified COVID-19 proteomic signature helps elucidate COVID-19 pathophysiology and may guide future COVID-19 treatment development.
Collapse
Affiliation(s)
- Maitray A Patel
- Epidemiology and Biostatistics, Western University, London, ON, N6A 3K7, Canada
| | - Mark Daley
- Epidemiology and Biostatistics, Western University, London, ON, N6A 3K7, Canada
- Computer Science, Western University, London, ON, N6A 3K7, Canada
| | | | - Marat Slessarev
- Medicine, Western University, London, ON, N6A 3K7, Canada
- Lawson Health Research Institute, London, ON, N6C 2R5, Canada
| | - Gediminas Cepinskas
- Lawson Health Research Institute, London, ON, N6C 2R5, Canada
- Medical Biophysics, Western University, London, ON, N6A 3K7, Canada
| | - Douglas D Fraser
- Lawson Health Research Institute, London, ON, N6C 2R5, Canada.
- Children's Health Research Institute, London, ON, N6C 4V3, Canada.
- Pediatrics, Western University, London, ON, N6A 3K7, Canada.
- Clinical Neurological Sciences, Western University, London, ON, N6A 3K7, Canada.
- Physiology & Pharmacology, Western University, London, ON, N6A 3K7, Canada.
- London Health Sciences Centre, 800 Commissioners Road East, London, ON, N6A 5W9, Canada.
| |
Collapse
|
24
|
Sharma H, Mossman K, Austin RC. Fatal attractions that trigger inflammation and drive atherosclerotic disease. Eur J Clin Invest 2024; 54:e14169. [PMID: 38287209 DOI: 10.1111/eci.14169] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/18/2023] [Revised: 12/14/2023] [Accepted: 01/09/2024] [Indexed: 01/31/2024]
Abstract
BACKGROUND Atherosclerosis is the salient, underlying cause of cardiovascular diseases, such as arrhythmia, coronary artery disease, cardiomyopathy, pulmonary embolism and myocardial infarction. In recent years, atherosclerosis pathophysiology has evolved from a lipid-based to an inflammation-centric ideology. METHODS This narrative review is comprised of review and original articles that were found through the PubMed search engine. The following search terms or amalgamation of terms were used: "cardiovascular disease," "atherosclerosis," "inflammation," "GRP78," "Hsp60," "oxidative low-density lipoproteins," "aldehyde dehydrogenase," "β2-glycoprotein," "lipoprotein lipase A," "human cytomegalovirus." "SARS-CoV-2," "chlamydia pneumonia," "autophagy," "thrombosis" and "therapeutics." RESULTS Emerging evidence supports the concept that atherosclerosis is associated with the interaction between cell surface expression of stress response chaperones, including GRP78 and Hsp60, and their respective autoantibodies. Moreover, various other autoantigens and their autoantibodies have displayed a compelling connection with the development of atherosclerosis, including oxidative low-density lipoproteins, aldehyde dehydrogenase, β2-glycoprotein and lipoprotein lipase A. Atherosclerosis progression is also concurrent with viral and bacterial activators of various diseases. This narrative review will focus on the contributions of human cytomegalovirus as well as SARS-CoV-2 and chlamydia pneumonia in atherosclerosis development. Notably, the interaction of an autoantigen with their respective autoantibodies or the presence of a foreign antigen can enhance inflammation development, which leads to atherosclerotic lesion progression. CONCLUSION We will highlight and discuss the complex role of the interaction between autoantigens and autoantibodies, and the presence of foreign antigens in the development of atherosclerotic lesions in relationship to pro-inflammatory responses.
Collapse
Affiliation(s)
- Hitesh Sharma
- Division of Nephrology, Department of Medicine, McMaster University, The Research Institute of St. Joe's Hamilton and the Hamilton Centre for Kidney Research, Hamilton, Ontario, Canada
| | - Karen Mossman
- Department of Medicine, Michael DeGroote Institute for Infectious Disease Research and the McMaster Immunology Research Centre, McMaster University, Hamilton, Ontario, Canada
| | - Richard C Austin
- Division of Nephrology, Department of Medicine, McMaster University, The Research Institute of St. Joe's Hamilton and the Hamilton Centre for Kidney Research, Hamilton, Ontario, Canada
| |
Collapse
|
25
|
Lopuhaä BV, Guzel C, van der Lee A, van den Bosch TPP, van Kemenade FJ, Huisman MV, Kruip MJHA, Luider TM, von der Thüsen JH. Increase in venous thromboembolism in SARS-CoV-2 infected lung tissue: proteome analysis of lung parenchyma, isolated endothelium, and thrombi. Histopathology 2024; 84:967-982. [PMID: 38253958 DOI: 10.1111/his.15143] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2023] [Revised: 12/22/2023] [Accepted: 01/04/2024] [Indexed: 01/24/2024]
Abstract
AIMS COVID-19 pneumonia is characterized by an increased rate of deep venous thrombosis and pulmonary embolism. To better understand the pathophysiology behind thrombosis in COVID-19, we performed proteomics analysis on SARS-CoV-2 infected lung tissue. METHODS Liquid chromatography mass spectrometry was performed on SARS-CoV-2 infected postmortem lung tissue samples. Five protein profiling analyses were performed: whole slide lung parenchyma analysis, followed by analysis of isolated thrombi and endothelium, both stratified by disease (COVID-19 versus influenza) and thrombus morphology (embolism versus in situ). Influenza autopsy cases with pulmonary thrombi were used as controls. RESULTS Compared to influenza controls, both analyses of COVID-19 whole-tissue and isolated endothelium showed upregulation of proteins and pathways related to liver metabolism including urea cycle activation, with arginase being among the top upregulated proteins in COVID-19 lung tissue. Analysis of isolated COVID-19 thrombi showed significant downregulation of pathways related to platelet activation compared to influenza thrombi. Analysis of isolated thrombi based on histomorphology shows that in situ thrombi have significant upregulation of coronavirus pathogenesis proteins. CONCLUSIONS The decrease in platelet activation pathways in severe COVID-19 thrombi suggests a relative increase in venous thromboembolism, as thrombi from venous origin tend to contain fewer platelets than arterial thrombi. Based on histomorphology, in situ thrombi show upregulation of various proteins related to SARS-CoV-2 pathogenesis compared to thromboemboli, which may indicate increased in situ pulmonary thrombosis in COVID-19. Therefore, this study supports the increase of venous thromboembolism without undercutting the involvement of in situ thrombosis in severe COVID-19.
Collapse
Affiliation(s)
- Boaz V Lopuhaä
- Department of Pathology, Erasmus University Medical Centre, Rotterdam, the Netherlands
| | - Coşkun Guzel
- Laboratory of Neuro-Oncology, Clinical and Cancer Proteomics, Department of Neurology, Erasmus University Medical Centre, Rotterdam, the Netherlands
| | | | | | | | - Menno V Huisman
- Department of Thrombosis and Hemostasis, Leiden University Medical Center, Leiden, the Netherlands
| | - Marieke J H A Kruip
- Department of Haematology, Erasmus University Medical Centre, Rotterdam, the Netherlands
| | - Theo M Luider
- Laboratory of Neuro-Oncology, Clinical and Cancer Proteomics, Department of Neurology, Erasmus University Medical Centre, Rotterdam, the Netherlands
| | - Jan H von der Thüsen
- Department of Pathology, Erasmus University Medical Centre, Rotterdam, the Netherlands
| |
Collapse
|
26
|
Zekić T, Belančić A. Antiphospholipid syndrome, thrombosis, and vaccination in the COVID-19 pandemic. Rheumatol Int 2024; 44:749-755. [PMID: 38393386 DOI: 10.1007/s00296-023-05531-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2023] [Accepted: 12/29/2023] [Indexed: 02/25/2024]
Abstract
Thrombosis is one of the many signs of antiphospholipid syndrome (APS) and COVID-19 infection. Although the mechanisms contributing to thrombosis in APS and COVID-19 are relatively similar, this remains an open subject. Even now (when the COVID-19 pandemic has subsided), there is no conclusive solution to APS and COVID-19 co-occurrence. The presence of newly generated antiphospholipid antibodies (aPLs) in COVID-19 infection may or may not be connected to the diagnosis of APS. The prevalence of aPLs is substantial in severe COVID-19 but not related to thrombosis or a worse outcome. Adequate monitoring of antibody positivity over time is recommended for APL diagnosis. On the other hand, thrombosis and thrombocytopenia can rarely occur with vaccination with mRNA vaccines. Some studies have shown that COVID-19 immunization is well tolerated among APS patients who are triple-positive for aPL, which may comfort patients and referring physicians and lessen hesitation in unvaccinated APS/aPL-positive patients. In this narrative review, we will give an overview of the interaction between aPL-APS-COVID-19-thrombosis and related diagnostic insights learned during the pandemic.
Collapse
Affiliation(s)
- Tatjana Zekić
- Clinical Hospital Center Rijeka, Department of Rheumatology and Clinical Immunology, University of Rijeka, Faculty of Medicine, Rijeka, Croatia.
- University of Rijeka, Faculty of Medicine, 51000, Rijeka, Croatia.
| | - Andrej Belančić
- Department of Clinical Pharmacology, Clinical Hospital Centre Rijeka, Krešimirova 42, 51000, Rijeka, Croatia
- Department of Basic and Clinical Pharmacology with Toxicology, University of Rijeka, Faculty of Medicine, Braće Branchetta 20, 51000, Rijeka, Croatia
| |
Collapse
|
27
|
Bilehjani E, Fakhari S, Farzin H, Tajlil A, Nader ND. Diagnosis and treatment of cardiovascular manifestations of COVID-19: a narrative review. Acta Cardiol 2024; 79:267-273. [PMID: 37606350 DOI: 10.1080/00015385.2023.2246200] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2023] [Accepted: 08/03/2023] [Indexed: 08/23/2023]
Abstract
Severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) was the main pathogen in the COVID-19 pandemic. This viral infection has been associated with several respiratory and non-respiratory complications contributing to a higher mortality rate, especially in patients with underlying heart diseases worldwide. Once considered a respiratory tract disease, it is now well-known that COVID-19 patients may experience a wide range of cardiac manifestations. Because of its remarkable direct and indirect effects on the cardiovascular system, herein, we examined the published literature that studied the hypothetical mechanisms of injury, manifestations, and diagnostic modalities, including changes in molecular biomarkers with a predictive value in the prognostication of the disease, as well as emerging evidence regarding the long-term cardiac complications of the disease.
Collapse
Affiliation(s)
- Eissa Bilehjani
- Department of Anesthesiology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Solmaz Fakhari
- Department of Anesthesiology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Haleh Farzin
- Department of Anesthesiology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Arezou Tajlil
- Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY, USA
| | - Nader D Nader
- Departments of Anesthesiology and Surgery, University at Buffalo Jacobs School of Medicine and Biomedical Sciences, Buffalo, NY, USA
| |
Collapse
|
28
|
Rasyid A, Harris S, Kurniawan M, Mesiano T, Hidayat R, Wiyarta E. Predictive value of admission D-dimer levels in patient with acute ischaemic stroke and COVID-19: a second-wave prospective cohort study. BMJ Open 2024; 14:e077500. [PMID: 38580372 PMCID: PMC11002405 DOI: 10.1136/bmjopen-2023-077500] [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: 07/08/2023] [Accepted: 03/21/2024] [Indexed: 04/07/2024] Open
Abstract
OBJECTIVES This study aimed to evaluate the predictive value of admission D-dimer levels for in-hospital mortality in patients with COVID-19 and acute ischaemic stroke. DESIGN Cohort (prospective). SETTING Tertiary referral hospital in the capital city of Indonesia conducted from June to December 2021. PARTICIPANTS 60 patients with acute ischaemic stroke and COVID-19 were included. Patients were classified into D-dimer groups (low and high) according to a 2 110 ng/mL cut-off value, determined via receiver operating characteristic analysis. PRIMARY AND SECONDARY OUTCOME MEASURES The primary outcome was in-hospital mortality, with admission D-dimer levels as the major predictor. Secondary outcomes included associations between other demographic and clinical variables and the admission D-dimer value. Kaplan-Meier method was used to carry out survival analysis, with univariable and multivariable Cox regression performed to assess the association of D-dimer levels and other confounding variables (including demographic, clinical and laboratory parameters) with in-hospital mortality. RESULTS The findings demonstrated an association between elevated admission D-dimer levels (≥2 110 ng/mL) and an increased likelihood of death during hospitalisation. The adjusted HR was 14.054 (95% CI 1.710 to 115.519; p=0.014), demonstrating an increase in mortality risk after accounting for confounders such as age and diabetes history. Other significant predictors of mortality included a history of diabetes and increased white blood cell count. CONCLUSIONS Admission D-dimer levels may be a useful predictive indicator for the likelihood of death during hospitalisation in individuals with COVID-19 and acute ischaemic stroke.
Collapse
Affiliation(s)
- Al Rasyid
- Department of Neurology, Faculty of Medicine, Universitas Indonesia-Dr Cipto Mangunkusumo National Hospital, Jakarta, Indonesia
| | - Salim Harris
- Department of Neurology, Faculty of Medicine, Universitas Indonesia-Dr Cipto Mangunkusumo National Hospital, Jakarta, Indonesia
| | - Mohammad Kurniawan
- Department of Neurology, Faculty of Medicine, Universitas Indonesia-Dr Cipto Mangunkusumo National Hospital, Jakarta, Indonesia
| | - Taufik Mesiano
- Department of Neurology, Faculty of Medicine, Universitas Indonesia-Dr Cipto Mangunkusumo National Hospital, Jakarta, Indonesia
| | - Rakhmad Hidayat
- Department of Neurology, Faculty of Medicine, Universitas Indonesia-Dr Cipto Mangunkusumo National Hospital, Jakarta, Indonesia
| | - Elvan Wiyarta
- Department of Neurology, Faculty of Medicine, Universitas Indonesia-Dr Cipto Mangunkusumo National Hospital, Jakarta, Indonesia
| |
Collapse
|
29
|
Herlo A, Marinescu AR, Cut TG, Laza R, Oancea CI, Manolescu D, Hogea E, Porosnicu TM, Sincaru SV, Dumache R, Ispas S, Nelson Twakor A, Nicolae M, Lazureanu VE. Risk Factors for Pulmonary Embolism in Individuals Infected with SARS-CoV2-A Single-Centre Retrospective Study. Biomedicines 2024; 12:774. [PMID: 38672130 PMCID: PMC11048050 DOI: 10.3390/biomedicines12040774] [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: 03/01/2024] [Revised: 03/24/2024] [Accepted: 03/27/2024] [Indexed: 04/28/2024] Open
Abstract
The emergence of SARS-CoV2 has presented itself as a significant global health crisis. The prevalence of thrombotic events is known to be high in these patients, affecting various organ systems, sometimes leading to cutaneous thrombosis, pulmonary embolism (PE), stroke, or coronary thrombosis. The available evidence suggests that thromboembolism, hypercoagulability, and the excessive production of proinflammatory cytokines play a significant role in the development of multiorgan failure. Methodology: This retrospective single-centre study was conducted at "Victor Babes" University of Medicine and Pharmacy from Timisoara, Romania, involving a total of 420 patients diagnosed with COVID-19. We separated them into a CONTROL group that included 319 patients, and an intervention group (PE) with 101 patients that, subsequent to infection with the virus, developed pulmonary embolism. The study included the reporting of demographic data, laboratory findings, and comorbidities. Results: Out of a total of 420 patients, 24% experienced pulmonary embolism, while 21.42% died. Arterial thrombotic events were found to be associated with factors such as age, cardiovascular disease, levels of white blood cells, D-dimers, and albumin in the blood. The findings of the study indicate that there is an independent association between pulmonary thrombosis and hypertension (odds ratio (OR): 1.1; 95% confidence interval (CI): 0.7 to 1.7; p = 0.6463), cancer (OR: 1.1; 95% CI: 0.6 to 2.3; p = 0.6014), and COPD (OR: 1.2; 95% CI: 0.6 to 2.3; p = 0.4927). On the other hand, there is a stronger correlation between PE and obesity (OR: 2.8; 95% CI: 1.7 to 4.6; p < 0.0001), diabetes (OR: 3.3; 95% CI: 2 to 5.3; p < 0.0001), and dyslipidemia (OR: 3.6; 95% CI: 2.3 to 5.8; p < 0.0001) in a multivariable regression logistic model. Conclusions: Patients diagnosed with severe forms of COVID-19 display a comparable incidence of arterial thrombotic events, which have been linked to poor survival rates.
Collapse
Affiliation(s)
- Alexandra Herlo
- Department XIII, Discipline of Infectious Diseases, Victor Babes University of Medicine and Pharmacy Timisoara, 2 Eftimie Murgu Square, 300041 Timisoara, Romania; (A.H.); (A.R.M.); (T.G.C.); (R.L.); (V.E.L.)
| | - Adelina Raluca Marinescu
- Department XIII, Discipline of Infectious Diseases, Victor Babes University of Medicine and Pharmacy Timisoara, 2 Eftimie Murgu Square, 300041 Timisoara, Romania; (A.H.); (A.R.M.); (T.G.C.); (R.L.); (V.E.L.)
| | - Talida Georgiana Cut
- Department XIII, Discipline of Infectious Diseases, Victor Babes University of Medicine and Pharmacy Timisoara, 2 Eftimie Murgu Square, 300041 Timisoara, Romania; (A.H.); (A.R.M.); (T.G.C.); (R.L.); (V.E.L.)
| | - Ruxandra Laza
- Department XIII, Discipline of Infectious Diseases, Victor Babes University of Medicine and Pharmacy Timisoara, 2 Eftimie Murgu Square, 300041 Timisoara, Romania; (A.H.); (A.R.M.); (T.G.C.); (R.L.); (V.E.L.)
| | - Cristian Iulian Oancea
- Department XIII, Discipline of Pneumology, Victor Babes University of Medicine and Pharmacy Timisoara, E. Murgu Square, Nr. 2, 300041 Timisoara, Romania;
- Center for Research and Innovation in Precision Medicine of Respiratory Diseases (CRIPMRD), Victor Babes University of Medicine and Pharmacy Timisoara, E. Murgu Square, Nr. 2, 300041 Timisoara, Romania
| | - Diana Manolescu
- Department XV, Discipline of Radiology, Victor Babes University of Medicine and Pharmacy Timisoara, E. Murgu Square, Nr. 2, 300041 Timisoara, Romania;
| | - Elena Hogea
- Department XIV, Discipline of Microbiology, Victor Babes University of Medicine and Pharmacy Timisoara, E. Murgu Square, Nr. 2, 300041 Timisoara, Romania;
| | - Tamara Mirela Porosnicu
- Doctoral School, Victor Babes University of Medicine and Pharmacy Timisoara, 300041 Timisoara, Romania;
- Intensive Care Unit, Victor Babes Clinical Hospital for Infectious Diseases and Pneumology, 300041 Timisoara, Romania
| | - Suzana Vasilica Sincaru
- Emergency Institute for Cardiovascular Diseases and Transplant, Strada Gheorghe Maricescu, 540327 Targu Mures, Romania
| | - Raluca Dumache
- Department of Forensic Medicine, Bioethics, Medical Ethics and Medical Law, “Victor Babes” University of Medicine and Pharmacy, 300041 Timisoara, Romania;
| | - Sorina Ispas
- Department of Anatomy, Faculty of General Medicine, “Ovidius” University, 900470 Constanta, Romania;
| | - Andreea Nelson Twakor
- Department of Internal Medicine, County Clinical Emergency Hospital of Constanta, 900591 Constanta, Romania;
| | - Maria Nicolae
- Department of Pediatrics, County Clinical Emergency Hospital of Constanta, 900591 Constanta, Romania;
| | - Voichita Elena Lazureanu
- Department XIII, Discipline of Infectious Diseases, Victor Babes University of Medicine and Pharmacy Timisoara, 2 Eftimie Murgu Square, 300041 Timisoara, Romania; (A.H.); (A.R.M.); (T.G.C.); (R.L.); (V.E.L.)
| |
Collapse
|
30
|
Ramphul K, Singh Dhaliwal J, Sombans S, Passi JK, Aggarwal S, Kumar N, Sakthivel H, Ahmed R, Verma R. Trends in admissions for COVID-19 in the United States between April 2020 and December 2021 and cardiovascular events. Arch Med Sci Atheroscler Dis 2024; 9:e60-e65. [PMID: 38846059 PMCID: PMC11155464 DOI: 10.5114/amsad/185410] [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: 02/26/2024] [Accepted: 02/27/2024] [Indexed: 06/09/2024] Open
Abstract
Introduction Coronavirus disease 2019 (COVID-19) can lead to cardiovascular complications. We aimed to understand the trends in admission for COVID-19 and the incidence of various cardiovascular events. Material and methods The 2020 and 2021 National Inpatient Sample (NIS) was studied for cases of COVID-19 between April 2020 and December 2021 in the United States. Linear-by-linear association helped us understand the trends of various events. Results The number of cases of COVID-19 was highest in January 2021 (261,469 patients). The incidence of acute pulmonary embolism rose from 2.08% in April 2020 to 4.82% in November 2021, while deep vein thrombosis cases rose from 1.74% in April 2020 to 2.63% in December 2021. The incidence of cardiac arrest varied, with a maximum of 3.00% in August 2021. Similarly, acute ischemic stroke cases experienced their highest incidence in January 2021 (0.91%). The incidence of myocarditis was highest in April and May 2020 (0.42% each). Peak takotsubo cases were seen between October and December 2021. The highest overall all-cause mortality among COVID-19 cases was seen in April 2020 (16.74%). Conclusions Throughout the 21 months of our analysis, various trends in COVID-19 cases and incidence of cardiac events were noticed. This could relate to the different variants of COVID-19, their direct and indirect impact on coagulation pathways and the myocardial tissues, and the protective roles of the vaccines.
Collapse
Affiliation(s)
| | | | | | - Jatin Kumar Passi
- Department of Internal Medicine, Guru Gobind Singh Medical College, Faridkot, India
| | - Shruti Aggarwal
- Department of Internal Medicine, Guru Gobind Singh Medical College, Faridkot, India
| | - Nomesh Kumar
- Department of Internal Medicine, Detroit Medical Center Sinai Grace-Wayne State University, Michigan, US
| | | | - Raheel Ahmed
- Royal Brompton Hospital, part of Guy’s and St. Thomas’ NHS Foundation Trust, London, United Kingdom
| | - Renuka Verma
- Department of Internal Medicine, University of Nevada, Las Vegas(UNLV), Las Vegas, US
| |
Collapse
|
31
|
Chen JX, Xu LL, Cheng JP, Xu XH. Challenging anticoagulation therapy for multiple primary malignant tumors combined with thrombosis: A case report and review of literature. World J Clin Cases 2024; 12:1704-1711. [PMID: 38576733 PMCID: PMC10989423 DOI: 10.12998/wjcc.v12.i9.1704] [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: 01/24/2024] [Revised: 02/21/2024] [Accepted: 03/01/2024] [Indexed: 03/25/2024] Open
Abstract
BACKGROUND Venous thromboembolism significantly contributes to patient deterioration and mortality. Management of its etiology and anticoagulation treatment is intricate, necessitating a comprehensive consideration of various factors, including the bleeding risk, dosage, specific anticoagulant medications, and duration of therapy. Herein, a case of lower extremity thrombosis with multiple primary malignant tumors and high risk of bleeding was reviewed to summarize the shortcomings of treatment and prudent anticoagulation experience. CASE SUMMARY An 83-year-old female patient was admitted to the hospital due to a 2-wk history of left lower extremity edema that had worsened over 2 d. Considering her medical history and relevant post-admission investigations, it was determined that the development of left lower extremity venous thrombosis and pulmonary embolism in this case could be attributed to a combination of factors, including multiple primary malignant tumors, iliac venous compression syndrome, previous novel coronavirus infection, and inadequate treatment for prior thrombotic events. However, the selection of appropriate anticoagulant medications, determination of optimal drug dosages, and establishment of an appropriate duration of anticoagulation therapy were important because of concurrent thrombocytopenia, decreased quantitative fibrinogen levels, and renal insufficiency. CONCLUSION Anticoagulant prophylaxis should be promptly initiated in cases of high-risk thrombosis. Individualized anticoagulation therapy is required for complex thrombosis.
Collapse
Affiliation(s)
- Jia-Xin Chen
- Department of Gerontology, China Resources and Wisco General Hospital, Wuhan University of Science and Technology, Wuhan 430080, Hubei Province, China
- Medical College, Wuhan University of Science and Technology, Wuhan 430065, Hubei Province, China
| | - Ling-Ling Xu
- Department of Gerontology, China Resources and Wisco General Hospital, Wuhan University of Science and Technology, Wuhan 430080, Hubei Province, China
- Medical College, Wuhan University of Science and Technology, Wuhan 430065, Hubei Province, China
| | - Jing-Ping Cheng
- Department of Gerontology, China Resources and Wisco General Hospital, Wuhan 430080, Hubei Province, China
| | - Xun-Hua Xu
- Department of Radiology, China Resources and Wisco General Hospital, Wuhan 430080, Hubei Province, China
| |
Collapse
|
32
|
Minutti-Zanella C, Gallardo-Pérez MM, Ruiz-Argüelles GJ. D-dimer in Coronavirus 2019: An Acute Phase Reactant? Semin Thromb Hemost 2024; 50:295-297. [PMID: 37353044 DOI: 10.1055/s-0043-1770365] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/25/2023]
Affiliation(s)
- Claudia Minutti-Zanella
- Universidad Popular Autónoma del Estado de Puebla, Puebla, México
- Laboratorios Ruiz/SYNLAB, Puebla, México
| | - Moisés M Gallardo-Pérez
- Universidad Popular Autónoma del Estado de Puebla, Puebla, México
- Centro de Hematología y Medicina Interna, Clínica Ruiz, Puebla, México
| | - Guillermo J Ruiz-Argüelles
- Universidad Popular Autónoma del Estado de Puebla, Puebla, México
- Centro de Hematología y Medicina Interna, Clínica Ruiz, Puebla, México
| |
Collapse
|
33
|
Zhang T, Zhang QF, Yang HM, Liu P, Sun P, Li YM, Zhang Z, Huang YZ, Yu XY, Chao-Lu-Men QQG, Su Q, Liu CF. Children with severe neurological symptoms associated with SARS-CoV-2 infection during Omicron pandemic in China. Pediatr Res 2024; 95:1088-1094. [PMID: 37990079 DOI: 10.1038/s41390-023-02904-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/15/2023] [Revised: 10/16/2023] [Accepted: 11/02/2023] [Indexed: 11/23/2023]
Abstract
BACKGROUND To analyze the clinical characteristics and outcomes of children with severe neurological symptoms associated with the severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) infection during the Omicron pandemic in China. METHODS This study used a questionnaire to obtain data from pediatric intensive care unit (PICU) centers in seven tertiary hospitals in Northeast China from December 1, 2022, to January 31, 2023. RESULTS A total of 255 patients were confirmed to have SARS-CoV-2 infection, and 45 patients (17.65 %) were included in this study. Of these, seven (15.6%) patients died, and the median time from admission to death was 35 h (IQR, 14-120 h). Twenty (52.6%) survivors experienced neurological sequelae. Patients with platelet counts lower than 100 × 109/L had a higher incidence of complications such as multiple organ dysfunction, mechanical ventilation rate, and mortality. Cranial magnetic resonance imaging (MRI) always reveals cerebral tissue edema, with some severe lesions forming a softening site. CONCLUSION Children infected with SARS-CoV-2 often exhibit severe neurological symptoms, and in some cases, they may rapidly develop malignant cerebral edema or herniation, leading to a fatal outcome. An early decrease in platelet count may associated with an unfavorable prognosis. IMPACT Since early December 2022, China has gradually adjusted its prevention and control policy of SARS-CoV-2; Omicron outbreaks have occurred in some areas for a relatively short period. Due to the differences in ethnicity, endemic strains and vaccination status, there was a little difference from what has been reported about children with SARS-CoV-2 infection with severe neurological symptoms in abroad. This is the first multicenter clinical study in children with nervous system involvement after acute SARS-CoV-2 infection in China, and helpful for pediatricians to have a more comprehensive understanding of the clinical symptoms and prognosis of such disease.
Collapse
Affiliation(s)
- Tao Zhang
- Department of Pediatrics, Shengjing Hospital of China Medical University, Shenyang, China
| | - Qiao-Feng Zhang
- Department of Pediatric Intensive Care Unit, Dalian Women and Children's Medical Group, Dalian, China
| | - Hong-Mei Yang
- Department of Pediatric Intensive Care Unit, Dalian Women and Children's Medical Group, Dalian, China
| | - Pin Liu
- Department of Pediatric Intensive Care Unit, Shenyang Children's Hospital, Shenyang, China
| | - Peng Sun
- Department of Pediatric Intensive Care Unit, Shenyang Children's Hospital, Shenyang, China
| | - Yu-Mei Li
- Department of Pediatric Intensive Care Unit, The First Hospital of Jilin University, Changchun, China
| | - Zhen Zhang
- Department of Pediatric Intensive Care Unit, The First Hospital of Jilin University, Changchun, China
| | | | - Xin-Yan Yu
- Department of Critical Medicine, Jiangnan Hospital of the Sixth Affiliated Hospital of Harbin Medical University (Harbin Children's Hospital), Harbin, China
| | - Qi-Qi-Ge Chao-Lu-Men
- Department of Intensive Care Unit, Inner Mongolia Medical University Affiliated Hospital, Hohhot, China
| | - Qin Su
- Department of Intensive Care Unit, Inner Mongolia Medical University Affiliated Hospital, Hohhot, China
| | - Chun-Feng Liu
- Department of Pediatrics, Shengjing Hospital of China Medical University, Shenyang, China.
| |
Collapse
|
34
|
Vettori M, Carpenè G, Salvagno GL, Gelati M, Dima F, Celegon G, Favaloro EJ, Lippi G. Effects of Recombinant SARS-CoV-2 Spike Protein Variants on Platelet Morphology and Activation. Semin Thromb Hemost 2024; 50:275-283. [PMID: 37327884 DOI: 10.1055/s-0043-1769939] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/18/2023]
Abstract
Platelets are central elements of hemostasis and also play a pivotal role in the pathogenesis of thrombosis in coronavirus disease 2019. This study was planned to investigate the effects of different severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) recombinant spike protein variants on platelet morphology and activation. Citrated whole blood collected from ostensibly healthy subjects was challenged with saline (control sample) and with 2 and 20 ng/mL final concentration of SARS-CoV-2 recombinant spike protein of Ancestral, Alpha, Delta, and Omicron variants. Platelet count was found to be decreased with all SARS-CoV-2 recombinant spike protein variants and concentrations tested, achieving the lowest values with 20 ng/mL Delta recombinant spike protein. The mean platelet volume increased in all samples irrespective of SARS-CoV-2 recombinant spike protein variants and concentrations tested, but especially using Delta and Alpha recombinant spike proteins. The values of both platelet function analyzer-200 collagen-adenosine diphosphate and collagen-epinephrine increased in all samples irrespective of SARS-CoV-2 recombinant spike protein variants and concentrations tested, and thus reflecting platelet exhaustion, and displaying again higher increases with Delta and Alpha recombinant spike proteins. Most samples where SARS-CoV-2 recombinant spike proteins were added were flagged as containing platelet clumps. Morphological analysis revealed the presence of a considerable number of activated platelets, platelet clumps, platelet-monocyte, and platelet-neutrophils aggregates, especially in samples spiked with Alpha and Delta recombinant spike proteins at 20 ng/mL. These results provide support to the evidence that SARS-CoV-2 is capable of activating platelets through its spike protein, though such effect varies depending on different spike protein variants.
Collapse
Affiliation(s)
- Marco Vettori
- Section of Clinical Biochemistry and School of Medicine, University of Verona, Verona, Italy
| | - Giovanni Carpenè
- Section of Clinical Biochemistry and School of Medicine, University of Verona, Verona, Italy
| | - Gian Luca Salvagno
- Section of Clinical Biochemistry and School of Medicine, University of Verona, Verona, Italy
| | - Matteo Gelati
- Section of Clinical Biochemistry and School of Medicine, University of Verona, Verona, Italy
| | - Francesco Dima
- Section of Clinical Biochemistry and School of Medicine, University of Verona, Verona, Italy
| | - Giovanni Celegon
- Section of Clinical Biochemistry and School of Medicine, University of Verona, Verona, Italy
| | - Emmanuel J Favaloro
- Department of Haematology, Sydney Centers for Thrombosis and Haemostasis, Institute of Clinical Pathology and Medical Research (ICPMR), NSW Health Pathology, Westmead Hospital, Westmead, New South Wales, Australia
- School of Medical Sciences, Faculty of Medicine and Health, University of Sydney, Westmead Hospital, Westmead, New South Wales, Australia
- Faculty of Science and Health, Charles Sturt University, Wagga Wagga, New South Wales, Australia
| | - Giuseppe Lippi
- Section of Clinical Biochemistry and School of Medicine, University of Verona, Verona, Italy
| |
Collapse
|
35
|
Chu YJ, Wong LC, Weng WC, Fan PC, Wang HP, Kuo YT, Yen TY, Lu CY, Lee PI, Chang LY, Wang CC, Wu ET, Lu FL, Peng SSF, Lee WT. High incidence of cerebrovascular lesions on magnetic resonance imaging in pediatric COVID-19 during omicron outbreak - A retrospective case series. J Formos Med Assoc 2024:S0929-6646(24)00141-4. [PMID: 38431481 DOI: 10.1016/j.jfma.2024.02.015] [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: 09/23/2023] [Revised: 01/21/2024] [Accepted: 02/26/2024] [Indexed: 03/05/2024] Open
Abstract
BACKGROUND The incidence of pediatric hospitalizations has significantly increased since the spread of the omicron variant of COVID-19. Changes of characteristics in respiratory and neurological symptoms have been reported. We performed a retrospective, cross-sectional study to characterize the MRI change in children with an emphasis on the change of cerebral vasculatures. METHODS We retrospectively collected clinical and MRI data of 31 pediatric patients with neurological symptoms during the acute infection and abnormalities on MRI during the outbreak of omicron variant from April 2022 to June 2022 in Taiwan. The clinical manifestations and MRI abnormalities were collected and proportion of patients with vascular abnormalities was calculated. RESULTS Among 31 pediatric patients with post-COVID-19 neurological symptoms, MRI abnormalities were observed in 15 (48.4%), predominantly encephalitis/encephalopathy (73.3%). Notable MRI findings included focal diffusion-weighted imaging (DWI) hyperintensity in cerebral cortex and thalamus, diffuse cortical T2/DWI hyperintensity, and lesions in the medulla, pons, cerebellum, and splenium of corpus callosum. Vascular abnormalities were seen in 12 (80%) patients with MRI abnormalities, mainly affecting the middle cerebral arteries. The spectrum of neurological manifestations ranged from seizures to Alice in Wonderland syndrome, underscoring the diverse impact of COVID-19 on pediatric patients. CONCLUSION A high proportion of vascular abnormalities was observed in pediatric patients with neurological involvements, suggesting that vascular involvement is an important mechanism of neurological manifestations in omicron variant infection.
Collapse
Affiliation(s)
- Yen-Ju Chu
- Department of Emergency Medicine, National Taiwan University Hospital, Taipei, Taiwan
| | - Lee-Chin Wong
- Department of Pediatric Neurology, National Taiwan University Children's Hospital, Taipei, Taiwan
| | - Wen-Chin Weng
- Department of Pediatric Neurology, National Taiwan University Children's Hospital, Taipei, Taiwan; Department of Pediatrics, National Taiwan University College of Medicine, Taipei, Taiwan
| | - Pi-Chuan Fan
- Department of Pediatric Neurology, National Taiwan University Children's Hospital, Taipei, Taiwan; Department of Pediatrics, National Taiwan University College of Medicine, Taipei, Taiwan
| | - Hsin-Pei Wang
- Department of Pediatrics, National Taiwan University Hospital YunLin Branch, Yun-Lin, Taiwan
| | - Yung-Ting Kuo
- Department of Pediatrics, Shuang Ho Hospital, Ministry of Health and Welfare, Taipei Medical University, New Taipei, Taiwan; Department of Pediatrics, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Ting-Yu Yen
- Department of Pediatrics, National Taiwan University College of Medicine, Taipei, Taiwan; Department of Pediatric, Division of Pediatric Infectious Diseases, National Taiwan University Children's Hospital, Taipei, Taiwan
| | - Chun-Yi Lu
- Department of Pediatrics, National Taiwan University College of Medicine, Taipei, Taiwan; Department of Pediatric, Division of Pediatric Infectious Diseases, National Taiwan University Children's Hospital, Taipei, Taiwan
| | - Ping-Ing Lee
- Department of Pediatrics, National Taiwan University College of Medicine, Taipei, Taiwan; Department of Pediatric, Division of Pediatric Infectious Diseases, National Taiwan University Children's Hospital, Taipei, Taiwan
| | - Luan-Yin Chang
- Department of Pediatrics, National Taiwan University College of Medicine, Taipei, Taiwan; Department of Pediatric, Division of Pediatric Infectious Diseases, National Taiwan University Children's Hospital, Taipei, Taiwan
| | - Ching-Chia Wang
- Department of Pediatrics, National Taiwan University College of Medicine, Taipei, Taiwan; Department of Pediatric, Division of Pediatric Pulmonary & Critical Care Medicine, National Taiwan University Children's Hospital, Taipei, Taiwan
| | - En-Ting Wu
- Department of Pediatrics, National Taiwan University College of Medicine, Taipei, Taiwan; Department of Pediatric, Division of Pediatric Pulmonary & Critical Care Medicine, National Taiwan University Children's Hospital, Taipei, Taiwan
| | - Frank Leigh Lu
- Department of Pediatrics, National Taiwan University College of Medicine, Taipei, Taiwan; Department of Pediatric, Division of Pediatric Pulmonary & Critical Care Medicine, National Taiwan University Children's Hospital, Taipei, Taiwan
| | - Steven Shinn-Forng Peng
- Department of Medical Imaging, National Taiwan University Hospital, Taiwan; Department of Nuclear Medicine, National Taiwan University Hospital, Taiwan; Department of Radiology, National Taiwan University College of Medicine, Taipei, Taiwan.
| | - Wang-Tso Lee
- Department of Pediatric Neurology, National Taiwan University Children's Hospital, Taipei, Taiwan; Department of Pediatrics, National Taiwan University College of Medicine, Taipei, Taiwan; Graduate Institute of Brain and Mind Sciences, National Taiwan University College of Medicine, Taipei, Taiwan.
| |
Collapse
|
36
|
de Azambuja Pias Weber A, Viero FT, Pillat MM, de Lima Gonçalves T. Changes in markers of inflammation and their correlation with death in patients with COVID-19 in the intensive care unit. Cytokine 2024; 175:156509. [PMID: 38241964 DOI: 10.1016/j.cyto.2024.156509] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2023] [Revised: 12/07/2023] [Accepted: 01/16/2024] [Indexed: 01/21/2024]
Abstract
This study aimed to characterize the changes in serum inflammatory mediators in hospitalized patients with COVID-19 correlating with death. The study includes 58 participants: i) inpatients (n = 37): patients suffering from severe acute respiratory syndrome due to COVID-19, who were admitted at Intensive Care Unit (ICU) recovered and who died and ii) control group (n = 21): community volunteers. Inflammatory mediators evaluated interleukin-2 (IL-2), interleukin-4 (IL-4), interleukin-6 (IL-6), interleukin-10 (IL-10), interleukin-17 (IL-17), interferon-gamma (IFN-γ) and interferon-gamma protein levels (IFN-γ), as well as, Urea, LDH, D-dimer, TAP/INR, AST, ALT and lymphocytes. Our results suggest that high levels of inflammatory markers, such as pro-inflammatory cytokines, and laboratory parameters, such as low levels of lymphocytes and high levels of IL-6, are associated with disease severity, especially in individuals who died. Constant measurement and monitoring of these inflammatory parameters is an effective tool in clinical practice, and it can help choosing appropriate therapies during the course of the disease.
Collapse
Affiliation(s)
- Andressa de Azambuja Pias Weber
- Postgraduate Program in Pharmaceutical Sciences, Department of Clinical and Toxicology Analysis, Center of Healthy Sciences, Federal University of Santa Maria (UFSM), Santa Maria, Brazil.
| | - Fernanda Tibolla Viero
- Postgraduate Program in Pharmacology, Department of Microbiology and Parasitology, Center of Healthy Sciences, Federal University of Santa Maria (UFSM), Santa Maria, Brazil.
| | - Micheli Mainardi Pillat
- Postgraduate Program in Pharmacology, Department of Microbiology and Parasitology, Center of Healthy Sciences, Federal University of Santa Maria (UFSM), Santa Maria, Brazil.
| | - Thissiane de Lima Gonçalves
- Postgraduate Program in Pharmaceutical Sciences, Department of Clinical and Toxicology Analysis, Center of Healthy Sciences, Federal University of Santa Maria (UFSM), Santa Maria, Brazil.
| |
Collapse
|
37
|
Tandon P, Abrams ND, Avula LR, Carrick DM, Chander P, Divi RL, Dwyer JT, Gannot G, Gordiyenko N, Liu Q, Moon K, PrabhuDas M, Singh A, Tilahun ME, Satyamitra MM, Wang C, Warren R, Liu CH. Unraveling Links between Chronic Inflammation and Long COVID: Workshop Report. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2024; 212:505-512. [PMID: 38315950 DOI: 10.4049/jimmunol.2300804] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/27/2023] [Accepted: 12/12/2023] [Indexed: 02/07/2024]
Abstract
As COVID-19 continues, an increasing number of patients develop long COVID symptoms varying in severity that last for weeks, months, or longer. Symptoms commonly include lingering loss of smell and taste, hearing loss, extreme fatigue, and "brain fog." Still, persistent cardiovascular and respiratory problems, muscle weakness, and neurologic issues have also been documented. A major problem is the lack of clear guidelines for diagnosing long COVID. Although some studies suggest that long COVID is due to prolonged inflammation after SARS-CoV-2 infection, the underlying mechanisms remain unclear. The broad range of COVID-19's bodily effects and responses after initial viral infection are also poorly understood. This workshop brought together multidisciplinary experts to showcase and discuss the latest research on long COVID and chronic inflammation that might be associated with the persistent sequelae following COVID-19 infection.
Collapse
Affiliation(s)
- Pushpa Tandon
- National Cancer Institute, National Institutes of Health, Rockville, MD
| | - Natalie D Abrams
- National Cancer Institute, National Institutes of Health, Rockville, MD
| | - Leela Rani Avula
- National Cancer Institute, National Institutes of Health, Rockville, MD
| | | | - Preethi Chander
- National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD
| | - Rao L Divi
- National Cancer Institute, National Institutes of Health, Rockville, MD
| | - Johanna T Dwyer
- Office of Dietary Supplements, National Institutes of Health, Bethesda, MD
| | - Gallya Gannot
- National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, MD
| | | | - Qian Liu
- National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD
| | - Kyung Moon
- National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD
| | - Mercy PrabhuDas
- National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD
| | - Anju Singh
- National Cancer Institute, National Institutes of Health, Rockville, MD
| | - Mulualem E Tilahun
- National Institute on Aging, National Institutes of Health, Bethesda, MD
| | - Merriline M Satyamitra
- National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD
| | - Chiayeng Wang
- National Cancer Institute, National Institutes of Health, Rockville, MD
| | - Ronald Warren
- National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD
| | - Christina H Liu
- National Institute of General Medical Sciences, National Institutes of Health, Bethesda, MD
| |
Collapse
|
38
|
Coelho SVA, Augusto FM, de Arruda LB. Potential Pathways and Pathophysiological Implications of Viral Infection-Driven Activation of Kallikrein-Kinin System (KKS). Viruses 2024; 16:245. [PMID: 38400022 PMCID: PMC10892958 DOI: 10.3390/v16020245] [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: 12/09/2023] [Revised: 01/30/2024] [Accepted: 01/31/2024] [Indexed: 02/25/2024] Open
Abstract
Microcirculatory and coagulation disturbances commonly occur as pathological manifestations of systemic viral infections. Research exploring the role of the kallikrein-kinin system (KKS) in flavivirus infections has recently linked microvascular dysfunctions to bradykinin (BK)-induced signaling of B2R, a G protein-coupled receptor (GPCR) constitutively expressed by endothelial cells. The relevance of KKS activation as an innate response to viral infections has gained increasing attention, particularly after the reports regarding thrombogenic events during COVID-19. BK receptor (B2R and B1R) signal transduction results in vascular permeability, edema formation, angiogenesis, and pain. Recent findings unveiling the role of KKS in viral pathogenesis include evidence of increased activation of KKS with elevated levels of BK and its metabolites in both intravascular and tissue milieu, as well as reports demonstrating that virus replication stimulates BKR expression. In this review, we will discuss the mechanisms triggered by virus replication and by virus-induced inflammatory responses that may stimulate KKS. We also explore how KKS activation and BK signaling may impact virus pathogenesis and further discuss the potential therapeutic application of BKR antagonists in the treatment of hemorrhagic and respiratory diseases.
Collapse
Affiliation(s)
- Sharton Vinícius Antunes Coelho
- Departamento de Virologia, Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-902, Brazil;
| | | | - Luciana Barros de Arruda
- Departamento de Virologia, Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-902, Brazil;
| |
Collapse
|
39
|
Hulshof AM, Nab L, van Rosmalen F, de Kok J, Mulder MMG, Hellenbrand D, Sels JWEM, Ten Cate H, Cannegieter SC, Henskens YMC, van Bussel BCT. Rotational thromboelastometry as a biomarker for mortality - The Maastricht Intensive Care COVID cohort. Thromb Res 2024; 234:51-58. [PMID: 38159324 DOI: 10.1016/j.thromres.2023.12.010] [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: 07/18/2023] [Revised: 12/18/2023] [Accepted: 12/18/2023] [Indexed: 01/03/2024]
Abstract
BACKGROUND Patients with severe coronavirus disease 2019 (COVID-19) present with persisting hypercoagulability, hypofibrinolysis and prolonged clot initiation as measured with viscoelastic assays. The objective of this study was to investigate the trajectories of traditional assays of hemostasis, routine and tissue plasminogen activator (tPA) rotational thromboelastometry (ROTEM) in COVID-19 patients and to study their association with mortality. METHODS Patients enrolled within the Maastricht Intensive Care COVID (MaastrICCht) cohort were included. Traditional assays of hemostasis (prothrombin time; PT, fibrinogen and D-dimer) were measured daily and ROTEM EXTEM, FIBTEM and tPA assays were performed weekly. Trajectories of these biomarkers were analyzed over time for survivors and non-survivors using linear mixed-effects models. Additional Fine and Gray competing risk survival analysis was performed for the first available measurement after intubation. RESULTS Of the 138 included patients, 57 (41 %) died in the intensive care unit (ICU). Over 450, 400 and 1900 individual measurements were available for analysis of routine, tPA ROTEM and traditional assays of hemostasis, respectively, with a median [IQR] follow-up of 15 [8-24] days. Non-survivors on average had prolonged CT (clotting time) and increased fibrinogen compared to survivors. MCF (maximum clot firmness), LOT (lysis onset time), LT (lysis time) and PT measurements increased more over time in non-survivors compared to survivors. Associations persisted after adjustment for demographics and disease severity. EXTEM and FIBTEM CT at intubation were associated with increased 45-day ICU mortality. CONCLUSIONS ROTEM measurements demonstrate a further increase of hypercoagulability and (hypo)fibrinolysis parameters in non-survivors throughout ICU admission. Furthermore, prolonged CT at intubation was associated with higher 45-day ICU mortality.
Collapse
Affiliation(s)
- Anne-Marije Hulshof
- Central Diagnostic Laboratory, Maastricht University Medical Centre+, Maastricht, the Netherlands; Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Maastricht, the Netherlands.
| | - Linda Nab
- Department of Clinical Epidemiology, Leiden University Medical Center, Leiden, the Netherlands
| | - Frank van Rosmalen
- Department of Intensive Care, Maastricht University Medical Centre+, Maastricht, the Netherlands
| | - Jip de Kok
- Department of Intensive Care, Maastricht University Medical Centre+, Maastricht, the Netherlands
| | - Mark M G Mulder
- Department of Intensive Care, Maastricht University Medical Centre+, Maastricht, the Netherlands; Department of Anesthesiology, Maastricht University Medical Centre+, Maastricht, the Netherlands
| | - Dave Hellenbrand
- Central Diagnostic Laboratory, Maastricht University Medical Centre+, Maastricht, the Netherlands
| | - Jan Willem E M Sels
- Department of Intensive Care, Maastricht University Medical Centre+, Maastricht, the Netherlands
| | - Hugo Ten Cate
- Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Maastricht, the Netherlands; Department of Internal Medicine, Maastricht University Medical Centre+, Maastricht, the Netherlands; Thrombosis Expert Centre Maastricht, Maastricht University Medical Centre+, Maastricht, the Netherlands
| | - Suzanne C Cannegieter
- Department of Clinical Epidemiology, Leiden University Medical Center, Leiden, the Netherlands; Department of Medicine - Thrombosis and Haemostasis, Leiden University Medical Center, Leiden, the Netherlands
| | - Yvonne M C Henskens
- Central Diagnostic Laboratory, Maastricht University Medical Centre+, Maastricht, the Netherlands; Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Maastricht, the Netherlands
| | - Bas C T van Bussel
- Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Maastricht, the Netherlands; Department of Intensive Care, Maastricht University Medical Centre+, Maastricht, the Netherlands; Care and Public Health Research Institute (CAPHRI), Maastricht University, Maastricht, the Netherlands
| |
Collapse
|
40
|
Parker WAE, Storey RF. The role of platelet P2Y 12 receptors in inflammation. Br J Pharmacol 2024; 181:515-531. [PMID: 37771103 DOI: 10.1111/bph.16256] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2023] [Revised: 08/15/2023] [Accepted: 09/15/2023] [Indexed: 09/30/2023] Open
Abstract
Inflammation is a complex pathophysiological process underlying many clinical conditions. Platelets contribute to the thrombo-inflammatory response. Platelet P2Y12 receptors amplify platelet activation, potentiating platelet aggregation, degranulation and shape change. The contents of platelet alpha granules, in particular, act directly on leucocytes, including mediating platelet-leucocyte aggregation and activation via platelet P-selectin. Much evidence for the role of platelet P2Y12 receptors in inflammation comes from studies using antagonists of these receptors, such as the thienopyridines clopidogrel and prasugrel, and the cyclopentyltriazolopyrimidine ticagrelor, in animal and human experimental models. These suggest that antagonism of P2Y12 receptors decreases markers of inflammation with some evidence that this reduces incidence of adverse clinical sequelae during inflammatory conditions. Interpretation is complicated by pleiotropic effects such as those of the thienopyridines on circulating leucocyte numbers and of ticagrelor on adenosine reuptake. The available evidence suggests that P2Y12 receptors are prominent mediators of inflammation and P2Y12 receptor antagonism as a potentially powerful strategy in a broad range of inflammatory conditions. LINKED ARTICLES: This article is part of a themed issue on Platelet purinergic receptor and non-thrombotic disease. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v181.4/issuetoc.
Collapse
Affiliation(s)
- William A E Parker
- Cardiovascular Research Unit, Division of Clinical Medicine, University of Sheffield, Sheffield, UK
- NIHR Sheffield Biomedical Research Centre, Sheffield Teaching Hospitals NHS Foundation Trust, Sheffield, UK
| | - Robert F Storey
- Cardiovascular Research Unit, Division of Clinical Medicine, University of Sheffield, Sheffield, UK
- NIHR Sheffield Biomedical Research Centre, Sheffield Teaching Hospitals NHS Foundation Trust, Sheffield, UK
| |
Collapse
|
41
|
S B MJ, Chacko B, Selvarajan S, Peter JV, Geevar T, Dave RG, Georgy JT, Zachariah A, George T, Sathyendra S, Hansdak SG, Krishnaswami RK, Thangakunam B, Gupta R, Karuppusami R, Nair SC, Srivastava A. Biomarkers of coagulation, endothelial, platelet function, and fibrinolysis in patients with COVID-19: a prospective study. Sci Rep 2024; 14:2011. [PMID: 38263377 PMCID: PMC10805716 DOI: 10.1038/s41598-024-51908-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2023] [Accepted: 01/11/2024] [Indexed: 01/25/2024] Open
Abstract
Prospective and sequential evaluation of homeostatic changes leading to thrombosis across COVID 19 disease severity spectrum are limited. In this prospective observational study, haemostasis was evaluated in patients with mild, moderate-severe, and critical COVID-19 infection. Markers of endothelial activation [Soluble thrombomodulin (sTM), von Willebrand Factor (VWF)], platelet activation [Soluble P-selectin, beta-thromboglobulin (BTG)] and global haemostasis [Rotational thromboelastometry (ROTEM)] were evaluated on days 1 and 5 after admission. The study cohort comprised of 100 adult patients (mild = 20, moderate-severe = 22, critical = 58). Sixty-five patients received anticoagulation for 10 (7-14) days. Thrombotic events were seen in 9 patients. In-hospital mortality was 21%. Endothelial activation markers were elevated at baseline in all subgroups, with levels in moderate-severe (sTM = 4.92 ng/ml, VWF = 295 U/dl) [reference-ranges: sTM = 2.26-4.55 ng/ml; Soluble P-selectin = 13.5-31.5 ng/ml; BTG = 0.034-1.99 ng/ml] and critical patients (sTM = 6.07 ng/ml, VWF = 294 U/dl) being significantly higher than in the mild group (sTM = 4.18 ng/ml, VWF = 206 U/dl). In contrast, platelet activation markers were elevated only in critically ill patients at baseline (Soluble P-selectin = 37.3 ng/ml, BTG = 2.51 ng/ml). The critical group had significantly lower fibrinolysis on days 1 and 5 when compared with the moderate-severe arm. COVID-19 infection was associated with graded endothelial activation and lower fibrinolysis that correlated with illness severity.
Collapse
Affiliation(s)
- Manoj Job S B
- Department of Critical Care, Christian Medical College, Vellore, 632004, India.
| | - Binila Chacko
- Department of Critical Care, Christian Medical College, Vellore, 632004, India
| | - Sushil Selvarajan
- Department of Clinical Hematology, Christian Medical College, Vellore, India
| | - John Victor Peter
- Department of Critical Care, Christian Medical College, Vellore, 632004, India
| | - Tulasi Geevar
- Department of Transfusion Medicine, Christian Medical College, Vellore, India
| | - Rutvi Gautam Dave
- Department of Transfusion Medicine, Christian Medical College, Vellore, India
| | - Josh Thomas Georgy
- Department of General Medicine, Christian Medical College, Vellore, India
| | - Anand Zachariah
- Department of General Medicine, Christian Medical College, Vellore, India
| | - Tina George
- Department of General Medicine, Christian Medical College, Vellore, India
| | - Sowmya Sathyendra
- Department of General Medicine, Christian Medical College, Vellore, India
| | | | | | | | - Richa Gupta
- Department of Respiratory Medicine, Christian Medical College, Vellore, India
| | - Reka Karuppusami
- Department of Biostatistics, Christian Medical College, Vellore, India
| | | | - Alok Srivastava
- Department of Clinical Hematology, Christian Medical College, Vellore, India
| |
Collapse
|
42
|
Che K, Zeng Z, Hong C, Peng D, Liu A, He Y. Association between serum C-reactive protein (CRP) and Omicron variant COVID-19 pneumonia in cancer patients: A multicenter cross-sectional study at the end of 2022 in China. Medicine (Baltimore) 2024; 103:e36965. [PMID: 38215120 PMCID: PMC10783274 DOI: 10.1097/md.0000000000036965] [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: 07/20/2023] [Accepted: 12/21/2023] [Indexed: 01/14/2024] Open
Abstract
Cancer patients with COVID-19 have a higher infection rate and mortality rate than non-cancer patients. However, there are few studies on the correlation between the serum C-reactive protein (CRP) and cancer patients with COVID-19. This study aims to investigate the association between serum CRP and the incidence of COVID-19 pneumonia in cancer patients at the end of 2022 in China. This cross-sectional study with a retrospective cohort between December 2022 and February 2023 assessed cancer patients complicated with COVID-19 infection in 2 Chinese institutions. Logistic regression analyses were used to compute Odds ratio (OR) and 95%CIs for the association between serum CRP and the incidence of COVID-19 pneumonia in cancer patients. A total of 213 cancer patients with COVID-19 were enrolled. Eighty-six patients (40.4%) developed COVID-19 pneumonia, among which 23 patients (10.8%) progressed to severe cases. Univariate Logistic regression showed that high CRP levels were found to be an unfavorable predictor of COVID-19 outcomes (OR = 17.9, 95%CI: 7.3, 43.6; P < .001). In the multivariate analysis, high CRP levels were associated with a higher incidence rate of COVID-19 pneumonia (OR = 9.8, 95%CI: 2.2, 43.8; P = .003). In the multivariate logistic regression model and smooth curve fitting, we found a correlation between CRP and COVID-19 pneumonia. The serum CRP was associated with the incidence of Omicron variant COVID- 19 pneumonia in cancer patients. Hence, cancer patients with high CRP level maybe need for timely computer tomography examination and more aggressive treatment.
Collapse
Affiliation(s)
- Kaijun Che
- Department of Oncology, The First People’s Hospital of Fuzhou, Fuzhou, Jiangxi Province, PR China
- Department of Oncology, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi Province, PR China
| | - Zhimin Zeng
- Department of Oncology, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi Province, PR China
- Jiangxi Key Laboratory of Clinical Translational Cancer Research, Nanchang, Jiangxi Province, PR China
- Radiation Induced Heart Damage Institute of Nanchang University, Nanchang, Jiangxi Province, PR China
| | - Chen Hong
- Department of Oncology, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi Province, PR China
| | - Duanyang Peng
- Department of Oncology, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi Province, PR China
| | - Anwen Liu
- Department of Oncology, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi Province, PR China
- Jiangxi Key Laboratory of Clinical Translational Cancer Research, Nanchang, Jiangxi Province, PR China
- Radiation Induced Heart Damage Institute of Nanchang University, Nanchang, Jiangxi Province, PR China
| | - Yanqing He
- Department of Nosocomial Infection Control, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi Province, PR China
| |
Collapse
|
43
|
Sultan MA, Kong Y, Story C, Caterson H, Dix C, Gad F, Dhaliwal JS, Dunkley S, Jo H, van Hal S, Passam F. Thrombo-inflammatory response in hospitalised patients with COVID-19: a single institution experience. Intern Med J 2024; 54:43-53. [PMID: 37926861 DOI: 10.1111/imj.16285] [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: 05/17/2023] [Accepted: 10/29/2023] [Indexed: 11/07/2023]
Abstract
BACKGROUND Severe COVID-19 causes acute inflammation, which is complicated by venous thromboembolism events (VTE). However, it is unclear if VTE risk has evolved over time since the COVID-19 outbreak. AIMS To determine markers of thrombo-inflammation and rates of symptomatic VTE in patients hospitalised for COVID-19 in a metropolitan hospital in Sydney, Australia. METHODS A retrospective, single-centre, cohort study was performed by reviewing electronic medical records of consecutive patients admitted to Royal Prince Alfred Hospital between March 2020 and September 2021. This period included three waves of COVID-19 outbreaks in Australia with the ancestral, alpha and delta variants. Standard coagulation assays and inflammatory markers were recorded over 4 weeks. RESULTS A total of 205 patients were consecutively admitted during the study period. Activated partial thromboplastin time, neutrophil count and C-reactive protein (CRP) were significantly increased in patients hospitalised in the intensive care unit (ICU) compared with non-ICU patients. The use of anti-inflammatory medication increased in 2021 compared with 2020. The mortality rate was 7.3% in our cohort. Ninety-four per cent of patients received anticoagulation with 6.3% of patients developing VTE. CONCLUSION We observed lower rates of VTE compared to the internationally reported rate for the same period. We conclude that in the setting of controlled hospital admission rate and standard anticoagulation guidelines, COVID-19 resulted in similar thrombo-inflammatory response and VTE rates over the first 1.5 years of the pandemic.
Collapse
Affiliation(s)
- Muhammad Ahmed Sultan
- Department of Haematology, Royal Prince Alfred Hospital, Sydney, New South Wales, Australia
- Central Clinical School, Faculty of Medicine Health, University of Sydney, Sydney, New South Wales, Australia
| | - Yvonne Kong
- Department of Haematology, Royal Prince Alfred Hospital, Sydney, New South Wales, Australia
| | - Chloe Story
- Department of Infectious Diseases, Royal Prince Alfred Hospital, Sydney, New South Wales, Australia
| | - Harriet Caterson
- Department of Respiratory Medicine, Royal Prince Alfred Hospital, Sydney, New South Wales, Australia
| | - Caroline Dix
- Department of Haematology, Royal Prince Alfred Hospital, Sydney, New South Wales, Australia
| | - Fady Gad
- Department of Pharmacy, Royal Prince Alfred Hospital, Sydney, New South Wales, Australia
| | - Jagpreet Singh Dhaliwal
- Department of Haematology, Royal Prince Alfred Hospital, Sydney, New South Wales, Australia
- Central Clinical School, Faculty of Medicine Health, University of Sydney, Sydney, New South Wales, Australia
| | - Scott Dunkley
- Department of Haematology, Royal Prince Alfred Hospital, Sydney, New South Wales, Australia
| | - Helen Jo
- Department of Respiratory Medicine, Royal Prince Alfred Hospital, Sydney, New South Wales, Australia
| | - Sebastian van Hal
- Department of Infectious Diseases, Royal Prince Alfred Hospital, Sydney, New South Wales, Australia
| | - Freda Passam
- Department of Haematology, Royal Prince Alfred Hospital, Sydney, New South Wales, Australia
- Central Clinical School, Faculty of Medicine Health, University of Sydney, Sydney, New South Wales, Australia
| |
Collapse
|
44
|
Emadi E, Hamidi Alamdari D, Attaran D, Attaran S. Application of methylene blue for the prevention and treatment of COVID-19: A narrative review. IRANIAN JOURNAL OF BASIC MEDICAL SCIENCES 2024; 27:780-792. [PMID: 38800024 PMCID: PMC11127079 DOI: 10.22038/ijbms.2024.71871.15617] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Figures] [Subscribe] [Scholar Register] [Received: 04/19/2023] [Accepted: 01/06/2024] [Indexed: 05/29/2024]
Abstract
The newest virus from the SARS family of viruses called acute syndrome-coronavirus-2 (SARS-CoV-2), which causes COVID-19 disease, was identified in China at the end of 2019. In March 2020, after it spread to 29 additional countries, it was declared a pandemic by the World Health Organization (WHO). SARS-CoV-2 infection mainly starts through the respiratory tract and causes a wide spectrum of symptoms from asymptomatic infections to acute respiratory distress syndrome with multi-organ failure and vasoplegic shock. Among the many immunomodulatory and antiviral drugs that have been studied for the treatment of COVID-19, methylene blue (MB) may play an influential role. This article reviews the history of MB applications, the antiviral effects of MB against SARS-CoV-2, and the results of in vivo and in vitro studies of the use of MB in COVID-19. Based on studies, MB can simultaneously affect most of the host's harmful responses caused by SARS-CoV-2 infection due to its multiple properties, including anti-hypoxemia, anti-oxidant, immune system modulator, and antiviral. The use of MB is associated with a reduction in the possibility of getting infection, and mortality, and can be used as a safe, effective, cheap, and available treatment option with minimal side effects for the clinical management of COVID-19.
Collapse
Affiliation(s)
- Elaheh Emadi
- Vascular and Endovascular Surgery Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Daryoush Hamidi Alamdari
- Surgical Oncology Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
- Department of Biochemistry, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Davood Attaran
- Lung Diseases Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Soroush Attaran
- Lung Diseases Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
45
|
Garcia-Larragoiti N, Cano-Mendez A, Jimenez-Vega Y, Trujillo M, Guzman-Cancino P, Ambriz-Murillo Y, Viveros-Sandoval ME. Inflammatory and Prothrombotic Biomarkers Contribute to the Persistence of Sequelae in Recovered COVID-19 Patients. Int J Mol Sci 2023; 24:17468. [PMID: 38139298 PMCID: PMC10744310 DOI: 10.3390/ijms242417468] [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: 11/18/2023] [Revised: 12/07/2023] [Accepted: 12/12/2023] [Indexed: 12/24/2023] Open
Abstract
The presence of long COVID (LC) following SARS-CoV-2 infection is a common condition that affects the quality of life of patients and represents a diagnostic challenge due to the diversity of symptoms that may coexist. We still do not have accurate information regarding the pathophysiological pathways that generate the presence of LC, and so it is important to know the inflammatory and immunothrombotic biomarker profiles and their implications in order to characterize risk subgroups and establish early therapeutic strategies. We performed the determination of inflammatory and immunothrombotic biomarkers in volunteers with previous diagnoses of SARS-CoV-2. The inflammatory biomarkers were analyzed in plasma by flow cytometry, and we analyzed the von Willebrand factor (vWF) in the plasma samples using ELISA. The clinical variables and the presence or absence of long COVID symptoms were then analyzed. IL-6, sCD40L, p-Selectin, PSGL-1, PAI-1, tPA, D-Dimer, TF, and Factor IX levels were elevated in the groups with LC, especially in the subgroup of patients with metabolic syndrome (MetS). VWF levels were found to be increased in patients with sequelae and MetS. Our results confirmed the persistence of an active immunothrombotic state, and so it is important to identify the population at risk in order to provide adequate clinical follow-up.
Collapse
Affiliation(s)
- Nallely Garcia-Larragoiti
- División de Estudios de Posgrado, Facultad de Ciencias Médicas y Biológicas “Dr. Ignacio Chávez”, Universidad Michoacana de San Nicolás de Hidalgo, Morelia 58060, Michoacán, Mexico
| | - Alan Cano-Mendez
- División de Estudios de Posgrado, Facultad de Ciencias Médicas y Biológicas “Dr. Ignacio Chávez”, Universidad Michoacana de San Nicolás de Hidalgo, Morelia 58060, Michoacán, Mexico
- Centro Multidisciplinario de Estudios en Biotecnología, Facultad de Medicina Veterinaria y Zootecnia, Universidad Michoacana de San Nicolás de Hidalgo, Morelia 58060, Michoacán, Mexico
| | - Yeny Jimenez-Vega
- División de Estudios de Posgrado, Facultad de Ciencias Médicas y Biológicas “Dr. Ignacio Chávez”, Universidad Michoacana de San Nicolás de Hidalgo, Morelia 58060, Michoacán, Mexico
| | - Mercedes Trujillo
- División de Estudios de Posgrado, Facultad de Ciencias Médicas y Biológicas “Dr. Ignacio Chávez”, Universidad Michoacana de San Nicolás de Hidalgo, Morelia 58060, Michoacán, Mexico
| | - Patricia Guzman-Cancino
- División de Estudios de Posgrado, Facultad de Ciencias Médicas y Biológicas “Dr. Ignacio Chávez”, Universidad Michoacana de San Nicolás de Hidalgo, Morelia 58060, Michoacán, Mexico
| | - Yesenia Ambriz-Murillo
- División de Estudios de Posgrado, Facultad de Ciencias Médicas y Biológicas “Dr. Ignacio Chávez”, Universidad Michoacana de San Nicolás de Hidalgo, Morelia 58060, Michoacán, Mexico
- Hospital Regional de Morelia ISSSTE, Morelia 58300, Michoacán, Mexico
| | - Martha Eva Viveros-Sandoval
- División de Estudios de Posgrado, Facultad de Ciencias Médicas y Biológicas “Dr. Ignacio Chávez”, Universidad Michoacana de San Nicolás de Hidalgo, Morelia 58060, Michoacán, Mexico
| |
Collapse
|
46
|
Yamazaki A, Nukui Y, Kameda T, Saito R, Koda Y, Ichimura N, Tohda S, Ohkawa R. Variation in presepsin and thrombomodulin levels for predicting COVID-19 mortality. Sci Rep 2023; 13:21493. [PMID: 38057335 PMCID: PMC10700539 DOI: 10.1038/s41598-023-48633-0] [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: 12/27/2022] [Accepted: 11/28/2023] [Indexed: 12/08/2023] Open
Abstract
Coronavirus disease (COVID-19) has caused extensive mortality globally; therefore, biomarkers predicting the severity and prognosis of COVID-19 are essential. This study aimed to evaluate the application of presepsin (P-SEP) and thrombomodulin (TM), which are biomarkers of sepsis and endothelial dysfunction, respectively, in the prognosis of COVID-19. Serum P-SEP and TM levels from COVID-19 patients (n = 183) were measured. Disease severity was classified as mild, moderate I, moderate II, or severe based on hemoglobin oxygen saturation and the history of intensive care unit transfer or use of ventilation at admission. Patients in the severe group were further divided into survivors and non-survivors. P-SEP and TM levels were significantly higher in the severe group than those in the mild group, even after adjusting for creatinine values. In addition, TM levels were significantly higher in non-survivors than in survivors. Changes in the P-SEP levels at two time points with an interval of 4.1 ± 2.2 days were significantly different between the survivors and non-survivors. In conclusion, TM and continuous P-SEP measurements may be useful for predicting mortality in patients with COVID-19. Moreover, our data indicate that P-SEP and TM values after creatinine adjustment could be independent predictive markers, apart from renal function.
Collapse
Affiliation(s)
- Azusa Yamazaki
- Department of Clinical Bioanalysis and Molecular Biology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), 1-5-45 Yushima, Bunkyo-ku, Tokyo, 113-8510, Japan
- Clinical Laboratory, Tokyo Medical and Dental University (TMDU) Hospital, 1-5-45 Yushima, Bunkyo-ku, Tokyo, 113-8510, Japan
| | - Yoko Nukui
- Department of Infection Control and Prevention, Tokyo Medical and Dental University (TMDU) Hospital, 1-5-45 Yushima, Bunkyo-ku, Tokyo, 113-8510, Japan
- Department of Infection Control and Laboratory Medicine, Kyoto Prefectural University of Medicine, 465 Kajii-cho, Kawaramachi-Hirokoji, Kamigyo-ku, Kyoto, 602-8566, Japan
| | - Takahiro Kameda
- Department of Clinical Bioanalysis and Molecular Biology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), 1-5-45 Yushima, Bunkyo-ku, Tokyo, 113-8510, Japan
| | - Ryoichi Saito
- Department of Molecular Microbiology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), 1-5-45 Yushima, Bunkyo-ku, Tokyo, 113-8510, Japan
| | - Yuki Koda
- Clinical Laboratory, Tokyo Medical and Dental University (TMDU) Hospital, 1-5-45 Yushima, Bunkyo-ku, Tokyo, 113-8510, Japan
| | - Naoya Ichimura
- Clinical Laboratory, Tokyo Medical and Dental University (TMDU) Hospital, 1-5-45 Yushima, Bunkyo-ku, Tokyo, 113-8510, Japan
| | - Shuji Tohda
- Clinical Laboratory, Tokyo Medical and Dental University (TMDU) Hospital, 1-5-45 Yushima, Bunkyo-ku, Tokyo, 113-8510, Japan
| | - Ryunosuke Ohkawa
- Department of Clinical Bioanalysis and Molecular Biology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), 1-5-45 Yushima, Bunkyo-ku, Tokyo, 113-8510, Japan.
| |
Collapse
|
47
|
Fusco F, Krasuski RA, Sadeghi S, Rosenbaum MS, Lewis MJ, Carazo MR, Rodriguez FH, Halpern DG, Feinberg JL, Galilea FA, Baraona F, Cedars AM, Ko JM, Porayette P, Maldonado JR, Frogoudaki AA, Nir A, Chaudhry A, John AS, Karbassi A, Ganame J, Hoskoppal A, Frischhertz BP, Hendrickson B, Rodriguez-Monserrate CP, Broda CR, Tobler D, Gregg D, Martinez-Quintana E, Yeung E, Krieger EV, Ruperti-Repilado FJ, Giannakoulas G, Lui GK, Ephrem G, Singh HS, Hasan A, Bartlett HL, Lindsay I, Grewal J, Nicolarsen J, Araujo JJ, Cramer JW, Bouchardy J, Al Najashi K, Ryan K, Alshawabkeh L, Andrade L, Ladouceur M, Schwerzmann M, Greutmann M, Merás P, Ferrero P, Dehghani P, Tung PP, Garcia-Orta R, Tompkins R, Gendi SM, Cohen S, Klewer SE, Hascoet S, Upadhyay S, Fisher SD, Cook S, Cotts TB, Kovacs AH, Aboulhosn JA, Scognamiglio G, Broberg CS, Sarubbi B. COVID-19-Related Thrombotic and Bleeding Events in Adults With Congenital Heart Disease. JACC. ADVANCES 2023; 2:100701. [PMID: 38938489 PMCID: PMC11198511 DOI: 10.1016/j.jacadv.2023.100701] [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: 11/07/2022] [Revised: 08/07/2023] [Accepted: 09/05/2023] [Indexed: 06/29/2024]
Abstract
Background Altered coagulation is a striking feature of COVID-19. Adult patients with congenital heart disease (ACHD) are prone to thromboembolic (TE) and bleeding complications. Objectives The purpose of this study was to investigate the prevalence and risk factors for COVID-19 TE/bleeding complications in ACHD patients. Methods COVID-19-positive ACHD patients were included between May 2020 and November 2021. TE events included ischemic cerebrovascular accident, systemic and pulmonary embolism, deep venous thrombosis, myocardial infarction, and intracardiac thrombosis. Major bleeding included cases with hemoglobin drop >2 g/dl, involvement of critical sites, or fatal bleeding. Severe infection was defined as need for intensive care unit, endotracheal intubation, renal replacement therapy, extracorporeal membrane oxygenation, or death. Patients with TE/bleeding were compared to those without events. Factors associated with TE/bleeding were determined using logistic regression. Results Of 1,988 patients (age 32 [IQR: 25-42] years, 47% male, 59 ACHD centers), 30 (1.5%) had significant TE/bleeding: 12 TE events, 12 major bleeds, and 6 with both TE and bleeding. Patients with TE/bleeding had higher in-hospital mortality compared to the remainder cohort (33% vs 1.7%; P < 0.0001) and were in more advanced physiological stage (P = 0.032) and NYHA functional class (P = 0.01), had lower baseline oxygen saturation (P = 0.0001), and more frequently had a history of atrial arrhythmia (P < 0.0001), previous hospitalization for heart failure (P < 0.0007), and were more likely hospitalized for COVID-19 (P < 0.0001). By multivariable logistic regression, prior anticoagulation (OR: 4.92; 95% CI: 2-11.76; P = 0.0003), cardiac injury (OR: 5.34; 95% CI: 1.98-14.76; P = 0.0009), and severe COVID-19 (OR: 17.39; 95% CI: 6.67-45.32; P < 0.0001) were independently associated with increased risk of TE/bleeding complications. Conclusions ACHD patients with TE/bleeding during COVID-19 infection have a higher in-hospital mortality from the illness. Risk of coagulation disorders is related to severe COVID-19, cardiac injury during infection, and use of anticoagulants.
Collapse
Affiliation(s)
- Flavia Fusco
- Adult Congenital Heart Disease Unit, Monaldi Hospital, Naples, Italy
| | - Richard A. Krasuski
- Department of Cardiovascular Medicine, Duke University Health System in Durham, Durham, North Carolina, USA
| | - Soraya Sadeghi
- Ahmanson/UCLA Adult Congenital Heart Center, Los Angeles, California, USA
| | - Marlon S. Rosenbaum
- Division of Cardiology, Columbia University Medical Center, New York, New York, USA
| | - Matthew J. Lewis
- Division of Cardiology, Columbia University Medical Center, New York, New York, USA
| | - Matthew R. Carazo
- Division of Cardiology, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Fred H. Rodriguez
- Division of Cardiology, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Dan G. Halpern
- Division of Cardiology, New York University Langone Health, New York, New York, USA
| | - Jodi L. Feinberg
- Division of Cardiology, New York University Langone Health, New York, New York, USA
| | - Francisca A. Galilea
- Instituto Nacional del Tórax - Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Fernando Baraona
- Instituto Nacional del Tórax - Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Ari M. Cedars
- Division of Cardiology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Jong M. Ko
- Division of Cardiology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Prashob Porayette
- Division of Cardiology, University of Iowa Stead Family Children’s Hospital, Iowa City, Iowa, USA
| | - Jennifer R. Maldonado
- Division of Cardiology, University of Iowa Stead Family Children’s Hospital, Iowa City, Iowa, USA
| | | | - Amiram Nir
- Pediatric Cardiology and Adult Congenital Heart Disease Unit, Shaare Zedek Medical Center, Jerusalem, Israel
| | - Anisa Chaudhry
- Penn State Hershey Heart and Vascular Institute, State College, Pennsylvania, USA
| | - Anitha S. John
- Division of Cardiology, Children’s National Hospital, Washington, District of Columbia, USA
| | | | | | - Arvind Hoskoppal
- UPMC Adult Congenital Heart Disease Program, Pittsburgh, Pennsylvania, USA
| | - Benjamin P. Frischhertz
- Division of Cardiovascular Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Benjamin Hendrickson
- Le Bonheur Heart Institute, University of Tennessee Health Science Center, Memphis, Tennessee, USA
| | | | | | - Daniel Tobler
- Division of Cardiology, University Hospital of Basel, Basel, Switzerland
| | - David Gregg
- Division of Cardiology, Medical University of South Carolina, Charleston, South Carolina, USA
| | - Efrén Martinez-Quintana
- Cardiology Service, Universitario Insular-Materno Infantil, Las Palmas de Gran Canaria, Spain
| | - Elizabeth Yeung
- Colorado’s Adult and Teen Congenital Heart Program, Colorado University School of Medicine, Aurora, Colorado, USA
| | - Eric V. Krieger
- Division of Cardiology, University of Washington School of Medicine, Seattle, Washington, USA
| | | | | | - George K. Lui
- Division of Cardiovascular Medicine and Pediatric Cardiology, Stanford University School of Medicine, Stanford, California, USA
| | - Georges Ephrem
- Krannert Cardiovascular Research Center, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Harsimran S. Singh
- Department of Medicine & Pediatrics, Weill Cornell Medicine, New York Presbyterian Hospital, New York, New York, USA
| | - Almeneisi Hasan
- The Heart Institute, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, USA
| | - Heather L. Bartlett
- Department of Pediatrics and Medicine, University of Wisconsin, Madison, Wisconsin, USA
| | - Ian Lindsay
- Division of Pediatric Cardiology, University of Utah, Salt Lake City, Utah, USA
| | - Jasmine Grewal
- St. Paul’s Hospital, University of British Columbia, Vancouver, British Columbia, Canada
| | - Jeremy Nicolarsen
- Department of Pediatric and Adult Cardiology, Providence Adult and Teen Congenital Heart Program, Spokane, Washington, USA
| | - John J. Araujo
- Department of Pediatric and Adult Congenital Heart Disease, Somer Incare Cardiovascular Center, Rionegro, Colombia
| | - Jonathan W. Cramer
- Division of Cardiovascular Medicine, University of Nebraska Medical Center, Omaha, Nebraska, USA
| | - Judith Bouchardy
- Department of Cardiology and Cardiac Surgery, University Hospital Lausanne, Lausanne, Switzerland
| | - Khalid Al Najashi
- Pediatric Cardiology Department, Prince Sultan Cardiac Center, Riyadh, Saudi Arabia
| | - Kristi Ryan
- Adult Congenital Heart Program, OSF Healthcare Children’s Hospital of Illinois, Peoria, Illinois, USA
| | - Laith Alshawabkeh
- Department of Cardiovascular Medicine, University of California-San Diego, La Jolla, California, USA
| | - Lauren Andrade
- Division of Cardiology, Hospital of University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Magalie Ladouceur
- Adult Congenital Heart Disease Unit, Hôpital Européen Georges Pompidou, AP-HP, Université de Paris Cité, Paris, France
| | - Markus Schwerzmann
- Center for Congenital Heart Disease, University Hospital Inselspital, Bern, Switzerland
| | | | - Pablo Merás
- Cardiology Department, University Hospital La Paz, Madrid, Spain
| | - Paolo Ferrero
- Cardiovascular Department, ASST Papa Giovanni XXIII, University of Milano, Bergamo, Italy
| | - Payam Dehghani
- Prairie Vascular Research Network, University of Saskatchewan, Regina, Saskatchewan, Canada
| | - Poyee P. Tung
- Division of Adult Congenital Heart Disease, University of Texas at Houston, Houston, Texas, USA
| | - Rocio Garcia-Orta
- Cardiology Department, Hospital Universitario Virgen de las Nieves, Granada, Spain
| | - Rose Tompkins
- The Geurin Family Congenital Heart Program, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Salwa M. Gendi
- Adult Congenital Heart Disease Program, West Virginia University, Morgantown, West Virginia, USA
| | - Scott Cohen
- Adult Congenital Heart Disease Program, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| | - Scott E. Klewer
- Division of Cardiology, University of Arizona, Tucson, Arizona, USA
| | - Sebastien Hascoet
- Department of Pediatric Cardiology and Congenital Heart Disease, Hôpital Marie Lannelongue, Le Plessis Robinson, France
| | - Shailendra Upadhyay
- Division of Pediatric Cardiology, Connecticut Children’s Medical Center, Hartford, Connecticut, USA
| | - Stacy D. Fisher
- Department of Medicine and Pediatrics, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Stephen Cook
- Adult Congenital Heart Center, Helen DeVos Children’s Hospital, Grand Rapids, Michigan, USA
| | - Timothy B. Cotts
- Department of Medicine and Pediatrics, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Adrienne H. Kovacs
- Knight Cardiovascular Institute, Oregon Health & Science University, Portland, Oregon, USA
| | - Jamil A. Aboulhosn
- Ahmanson/UCLA Adult Congenital Heart Center, Los Angeles, California, USA
| | | | - Craig S. Broberg
- Knight Cardiovascular Institute, Oregon Health & Science University, Portland, Oregon, USA
| | - Berardo Sarubbi
- Adult Congenital Heart Disease Unit, Monaldi Hospital, Naples, Italy
| |
Collapse
|
48
|
Goracci L, Petito E, Di Veroli A, Falcinelli E, Bencivenga C, Giglio E, Becattini C, De Robertis E, Vaudo G, Gresele P. A platelet lipidomics signature in patients with COVID-19. Platelets 2023; 34:2200847. [PMID: 37114418 DOI: 10.1080/09537104.2023.2200847] [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: 04/29/2023]
Abstract
Ischemic cardiovascular and venous thromboembolic events are a frequent cause of death in severe COVID-19 patients. Platelet activation plays a key role in these complications, however platelet lipidomics have not been studied yet. The aim of our pilot investigation was to perform a preliminary study of platelet lipidomics in COVID-19 patients compared to healthy subjects. Lipid extraction and identification of ultrapurified platelets from eight hospitalized COVID-19 patients and eight age- and sex-matched healthy controls showed a lipidomic pattern almost completely separating COVID-19 patients from healthy controls. In particular, a significant decrease of ether phospholipids and increased levels of ganglioside GM3 were observed in platelets from COVID-19 patients. In conclusion, our study shows for the first time that platelets from COVID-19 patients display a different lipidomics signature distinguishing them from healthy controls, and suggests that altered platelet lipid metabolism may play a role in viral spreading and in the thrombotic complications of COVID-19.
Collapse
Affiliation(s)
- Laura Goracci
- Department of Chemistry, Biology and Biotechnology, University of Perugia, Perugia, Italy
| | - Eleonora Petito
- Department of Medicine and Surgery, Section of Internal and Cardiovascular Medicine, University of Perugia, Perugia, Italy
| | - Alessandra Di Veroli
- Department of Chemistry, Biology and Biotechnology, University of Perugia, Perugia, Italy
| | - Emanuela Falcinelli
- Department of Medicine and Surgery, Section of Internal and Cardiovascular Medicine, University of Perugia, Perugia, Italy
| | - Caterina Bencivenga
- Department of Chemistry, Biology and Biotechnology, University of Perugia, Perugia, Italy
| | - Elisa Giglio
- Department of Medicine and Surgery, Section of Internal and Cardiovascular Medicine, University of Perugia, Perugia, Italy
| | - Cecilia Becattini
- Department of Medicine and Surgery, Section of Internal and Cardiovascular Medicine, University of Perugia, Perugia, Italy
| | - Edoardo De Robertis
- Division of Anaesthesia, Analgesia, and Intensive Care, University of Perugia, Perugia, Italy
| | - Gaetano Vaudo
- Unit of Internal Medicine, Terni University Hospital, Terni, Italy
| | - Paolo Gresele
- Department of Medicine and Surgery, Section of Internal and Cardiovascular Medicine, University of Perugia, Perugia, Italy
| |
Collapse
|
49
|
Emmenegger M, Emmenegger V, Shambat SM, Scheier TC, Gomez-Mejia A, Chang CC, Wendel-Garcia PD, Buehler PK, Buettner T, Roggenbuck D, Brugger SD, Frauenknecht KBM. Antiphospholipid antibodies are enriched post-acute COVID-19 but do not modulate the thrombotic risk. Clin Immunol 2023; 257:109845. [PMID: 37995947 DOI: 10.1016/j.clim.2023.109845] [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: 07/25/2023] [Revised: 10/29/2023] [Accepted: 11/08/2023] [Indexed: 11/25/2023]
Abstract
BACKGROUND AND OBJECTIVES COVID-19-associated coagulopathy, shown to increase the risk for the occurrence of thromboses and microthromboses, displays phenotypic features of the antiphospholipid syndrome (APS), a prototype antibody-mediated autoimmune disease. Several groups have reported elevated levels of criteria and non-criteria antiphospholipid antibodies (aPL), assumed to cause APS, during acute or post-acute COVID-19. However, disease heterogeneity of COVID-19 is accompanied by heterogeneity in molecular signatures, including aberrant cytokine profiles and an increased occurrence of autoantibodies. Moreover, little is known about the association between autoantibodies and the clinical events. Here, we first aim to characterise the antiphospholipid antibody, anti-SARS-CoV-2 antibody, and the cytokine profiles in a diverse collective of COVID-19 patients (disease severity: asymptomatic to intensive care), using vaccinated individuals and influenza patients as comparisons. We then aim to assess whether the presence of aPL in COVID-19 is associated with an increased incidence of thrombotic events in COVID-19. METHODS AND RESULTS We conducted anti-SARS-CoV-2 IgG and IgA microELISA and IgG, IgA, and IgM antiphospholipid line immunoassay (LIA) against 10 criteria and non-criteria antigens in 155 plasma samples of 124 individuals, and we measured 16 cytokines and chemokines in 112 plasma samples. We additionally employed clinical and demographic parameters to conduct multivariable regression analyses within multiple paradigms. In line with recent results, we find that IgM autoantibodies against annexin V (AnV), β2-glycoprotein I (β2GPI), and prothrombin (PT) are enriched upon infection with SARS-CoV-2. There was no evidence for seroconversion from IgM to IgG or IgA. PT, β2GPI, and AnV IgM as well as cardiolipin (CL) IgG antiphospholipid levels were significantly elevated in the COVID-19 but not in the influenza or control groups. They were associated predominantly with the strength of the anti-SARS-CoV-2 antibody titres and the major correlate for thromboses was SARS-CoV-2 disease severity. CONCLUSION While we have recapitulated previous findings, we conclude that the presence of the aPL, most notably PT, β2GPI, AnV IgM, and CL IgG in COVID-19 are not associated with a higher incidence of thrombotic events.
Collapse
Affiliation(s)
- Marc Emmenegger
- Institute of Neuropathology, University of Zurich, 8091 Zurich, Switzerland; Division of Medical Immunology, Department of Laboratory Medicine, University Hospital Basel, 4031 Basel, Switzerland.
| | - Vishalini Emmenegger
- Department of Biosystems Science and Engineering, ETH Zürich, Basel, Switzerland
| | - Srikanth Mairpady Shambat
- Department of Infectious Diseases and Hospital Epidemiology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Thomas C Scheier
- Department of Infectious Diseases and Hospital Epidemiology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Alejandro Gomez-Mejia
- Department of Infectious Diseases and Hospital Epidemiology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Chun-Chi Chang
- Department of Infectious Diseases and Hospital Epidemiology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Pedro D Wendel-Garcia
- Institute of Intensive Care Medicine, University and University Hospital Zurich, Zurich, Switzerland
| | - Philipp K Buehler
- Institute of Intensive Care Medicine, University and University Hospital Zurich, Zurich, Switzerland
| | | | - Dirk Roggenbuck
- GA Generic Assays GmbH, Dahlewitz, Germany; Institute of Biotechnology, Faculty Environment and Natural Sciences, Brandenburg University of Technology Cottbus-Senftenberg, Senftenberg, Germany; Faculty of Health Sciences Brandenburg, University of Technology Cottbus-Senftenberg, Senftenberg, Germany
| | - Silvio D Brugger
- Department of Infectious Diseases and Hospital Epidemiology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Katrin B M Frauenknecht
- Institute of Neuropathology, University Medical Center of the Johannes Gutenberg-University, 55131 Mainz, Germany; National Center of Pathology (NCP), Laboratoire National de Santé (LNS), Luxembourg Center of Neuropathology (LCNP), 3555 Dudelange, Luxembourg
| |
Collapse
|
50
|
Harte JV, Coleman-Vaughan C, Crowley MP, Mykytiv V. It's in the blood: a review of the hematological system in SARS-CoV-2-associated COVID-19. Crit Rev Clin Lab Sci 2023; 60:595-624. [PMID: 37439130 DOI: 10.1080/10408363.2023.2232010] [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: 04/10/2023] [Accepted: 06/27/2023] [Indexed: 07/14/2023]
Abstract
The emergence of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has led to an unprecedented global healthcare crisis. While SARS-CoV-2-associated COVID-19 affects primarily the respiratory system, patients with COVID-19 frequently develop extrapulmonary manifestations. Notably, changes in the hematological system, including lymphocytopenia, neutrophilia and significant abnormalities of hemostatic markers, were observed early in the pandemic. Hematological manifestations have since been recognized as important parameters in the pathophysiology of SARS-CoV-2 and in the management of patients with COVID-19. In this narrative review, we summarize the state-of-the-art regarding the hematological and hemostatic abnormalities observed in patients with SARS-CoV-2-associated COVID-19, as well as the current understanding of the hematological system in the pathophysiology of acute and chronic SARS-CoV-2-associated COVID-19.
Collapse
Affiliation(s)
- James V Harte
- Department of Haematology, Cork University Hospital, Wilton, Cork, Ireland
- School of Biochemistry & Cell Biology, University College Cork, Cork, Ireland
| | | | - Maeve P Crowley
- Department of Haematology, Cork University Hospital, Wilton, Cork, Ireland
- Irish Network for Venous Thromboembolism Research (INViTE), Ireland
| | - Vitaliy Mykytiv
- Department of Haematology, Cork University Hospital, Wilton, Cork, Ireland
| |
Collapse
|