1
|
Weaver D, Gago S, Bassetti M, Giacobbe DR, Prattes J, Hoenigl M, Reizine F, Guegan H, Gangneux JP, Bromley MJ, Bowyer P. Mycobiome analyses of critically ill COVID-19 patients. Microbiol Spectr 2024:e0411023. [PMID: 39699254 DOI: 10.1128/spectrum.04110-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2023] [Accepted: 07/07/2024] [Indexed: 12/20/2024] Open
Abstract
Coronavirus disease 2019 (COVID-19)-associated pulmonary aspergillosis (CAPA) is a life-threatening complication in patients with severe COVID-19. Previously, acute respiratory distress syndrome in patients with COVID-19 has been associated with lung fungal dysbiosis, evidenced by reduced microbial diversity and Candida colonization. Increased fungal burden in the lungs of critically ill COVID-19 patients is linked to prolonged mechanical ventilation and increased mortality. However, specific mycobiome signatures associated with severe COVID-19 in the context of survival and antifungal drug prophylaxis have not yet been determined, and such knowledge could have an important impact on treatment. To understand the composition of the respiratory mycobiome in critically ill COVID-19 patients with and without CAPA and the impact of antifungal use in patient outcome, we performed a multinational study of 39 COVID-19 patients in intensive care units (ICUs). Respiratory mycobiome was profiled using internal transcribed spacer 1 sequencing, and Aspergillus fumigatus burden was further validated using quantitative PCR. Fungal communities were investigated using alpha diversity, beta diversity, taxa predominance, and taxa abundances. Respiratory mycobiomes of COVID-19 patients were dominated by Candida and Aspergillus. There was no significant association with corticosteroid use or CAPA diagnosis and respiratory fungal communities. Increased A. fumigatus burden was associated with mortality and, the use of azoles at ICU admission was linked with an absence of A. fumigatus. Our findings suggest that mold-active antifungal treatment at ICU admission may be linked with reduced A. fumigatus-associated mortality in severe COVID-19. However, further studies are warranted on this topic.IMPORTANCEInvasive fungal infections are a serious complication affecting up to a third of patients with severe COVID-19. Nevertheless, our understanding of the fungal communities in the lungs during critically ill COVID-19 remains limited. Evidence suggests a higher fungal burden is associated with prolonged ventilation and higher mortality, although the particular organisms responsible for this link are unclear. Antifungal prophylaxis may be beneficial for reducing the burden of fungal co-infections in COVID-19 intensive care. However, the composition of the fungal microbiome in severe COVID-19 in relation to prophylactic antifungals, as well as how this is associated with survival outcomes, is yet to be studied. Our study provides insights into the lung fungal microbiome in severe COVID-19 and has found antifungal treatment to be associated with lower Aspergillus fumigatus burden and that higher levels of this pathogen are associated with mortality. Therefore, our study suggests mold-active antifungal prophylaxis may be beneficial in severe COVID-19.
Collapse
Affiliation(s)
| | - Sara Gago
- University of Manchester, Manchester, United Kingdom
| | - Matteo Bassetti
- Department of Health Sciences (DISSAL), University of Genoa, Genoa, Italy
- Infectious Diseases Unit, IRCCS San Martino Polyclinic Hospital, Genoa, Italy
| | - Daniele Roberto Giacobbe
- Department of Health Sciences (DISSAL), University of Genoa, Genoa, Italy
- Infectious Diseases Unit, IRCCS San Martino Polyclinic Hospital, Genoa, Italy
| | - Juergen Prattes
- Division of Infectious Diseases, Medical University of Graz, Graz, Austria
| | - Martin Hoenigl
- Division of Infectious Diseases, Medical University of Graz, Graz, Austria
- Biotech Med, Graz, Austria
- Translational Medical Mycology Research Unit, ECMM Excellence Center for Medical Mycology, Medical University of Graz, Graz, Austria
| | - Florian Reizine
- Medical Intensive Care Unit, Centre Hospitalier Universitaire de Rennes, Rennes, France
| | - Hélène Guegan
- CHU Rennes, Inserm, EHESP, Institut de Recherche en Santé, Environnement et Travail (IRSET), Université de Rennes, Rennes, France
- Centre Hospitalier Universitaire de Rennes, Laboratoire de Parasitologie-Mycologie, Centre National de Référence Mycoses et Antifongiques-Laboratoire Associé Asp-C, European Excellence Center for Medical Mycology (ECMM), Rennes, France
| | - Jean-Pierre Gangneux
- CHU Rennes, Inserm, EHESP, Institut de Recherche en Santé, Environnement et Travail (IRSET), Université de Rennes, Rennes, France
- Centre Hospitalier Universitaire de Rennes, Laboratoire de Parasitologie-Mycologie, Centre National de Référence Mycoses et Antifongiques-Laboratoire Associé Asp-C, European Excellence Center for Medical Mycology (ECMM), Rennes, France
| | | | - Paul Bowyer
- University of Manchester, Manchester, United Kingdom
| |
Collapse
|
2
|
Hoffmann T, Michel J, Nitsche A, Mache C, Schulze J, Wolff T, Laue M. Electron microscopy images and morphometric data of SARS-CoV-2 variants in ultrathin plastic sections. Sci Data 2024; 11:1322. [PMID: 39632915 PMCID: PMC11618623 DOI: 10.1038/s41597-024-04182-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2024] [Accepted: 11/28/2024] [Indexed: 12/07/2024] Open
Abstract
Conventional thin section electron microscopy of viral pathogens, such as the pandemic SARS-CoV-2, can provide structural information on the virus particle phenotype and its evolution. We recorded about 900 transmission electron microscopy images of different SARS-CoV-2 variants, including Alpha (B.1.1.7), Beta (B.1.351), Delta (B.1.617.2) and Omicron BA.2 (B.1.1.529) and determined various morphometric parameters, such as maximal diameter and spike number, using a previously published measurement method. The datasets of the evolved virus variants were supplemented with images and measurements of the early SARS-CoV-2 isolates Munich929 and Italy-INMI1 to allow direct comparison. Infected Vero cell cultures were cultivated under comparable conditions to produce the viruses for imaging and morphometric analysis. The images and measurements can be used as a basis to analyse the morphometric changes of further evolving viruses at the particle level or for developing automated image processing workflows and analysis.
Collapse
Affiliation(s)
- Tobias Hoffmann
- Advanced Light and Electron Microscopy, Centre for Biological Threats and Special Pathogens 4 (ZBS 4), Robert Koch Institute, Berlin, Germany
| | - Janine Michel
- Highly Pathogenic Viruses, Centre for Biological Threats and Special Pathogens 1 (ZBS 1), Robert Koch Institute, Berlin, Germany
| | - Andreas Nitsche
- Highly Pathogenic Viruses, Centre for Biological Threats and Special Pathogens 1 (ZBS 1), Robert Koch Institute, Berlin, Germany
| | - Christin Mache
- Influenza and Other Respiratory Viruses (Unit 17), Robert Koch Institute, Berlin, Germany
| | - Jessica Schulze
- Influenza and Other Respiratory Viruses (Unit 17), Robert Koch Institute, Berlin, Germany
| | - Thorsten Wolff
- Influenza and Other Respiratory Viruses (Unit 17), Robert Koch Institute, Berlin, Germany
| | - Michael Laue
- Advanced Light and Electron Microscopy, Centre for Biological Threats and Special Pathogens 4 (ZBS 4), Robert Koch Institute, Berlin, Germany.
| |
Collapse
|
3
|
Pedroso RB, Torres L, Ventura LA, Camatta GC, Mota C, Mendes AC, Ribeiro F, Guimarães HC, Barbuto RC, Caixeta F, Nascimento LS, Oliveira MA, Martins VD, Silveira-Nunes G, Tupinambás U, Teixeira-Carvalho A, Graça L, Faria AMC. Rapid progression of CD8 and CD4 T cells to cellular exhaustion and senescence during SARS-CoV2 infection. J Leukoc Biol 2024; 116:1385-1397. [PMID: 39298288 DOI: 10.1093/jleuko/qiae180] [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: 11/24/2023] [Revised: 05/19/2024] [Indexed: 09/21/2024] Open
Abstract
Risk factors for the development of severe COVID-19 include several comorbidities, but age was the most striking one since elderly people were disproportionately affected by SARS-CoV-2 infection. Among the reasons for this markedly unfavorable response in the elderly, immunosenescence and inflammaging appear as major drivers of this outcome. A finding that was also notable was that hospitalized patients with severe COVID-19 have an accumulation of senescent T cells, suggesting that immunosenescence may be aggravated by SARS-CoV-2 infection. The present work was designed to examine whether these immunosenescence changes are characteristic of COVID-19 and whether it is dependent on disease severity using cross-sectional and longitudinal studies. Our cross-sectional data show that COVID-19, but not other respiratory infections, rapidly increased cellular senescence and exhaustion in CD4 and CD8 T cells during early infection. In addition, longitudinal analyses with patients from Brazil and Portugal provided evidence of increased frequencies of senescent and exhausted T cells over a 7-d period in patients with mild/moderate and severe COVID-19. Altogether, the study suggests that accelerated immunosenescence in CD4 and especially CD8 T-cell compartments may represent a common and unique outcome of SARS-CoV2 infection.
Collapse
Affiliation(s)
- Rodrigo Balsinha Pedroso
- Faculdade de Medicina, Instituto de Medicina Molecular, Universidade de Lisboa, Av. Prof. Egas Moniz, 1649, 1649-028, Lisboa, Portugal
| | - Lícia Torres
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas (ICB), Universidade Federal de Minas Gerais (UFMG), Av. Antonio Carlos, 6627, 31270-901, Belo Horizonte, MG, Brazil
| | - Lucas Araújo Ventura
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas (ICB), Universidade Federal de Minas Gerais (UFMG), Av. Antonio Carlos, 6627, 31270-901, Belo Horizonte, MG, Brazil
| | - Giovanna Caliman Camatta
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas (ICB), Universidade Federal de Minas Gerais (UFMG), Av. Antonio Carlos, 6627, 31270-901, Belo Horizonte, MG, Brazil
| | - Catarina Mota
- Faculdade de Medicina, Instituto de Medicina Molecular, Universidade de Lisboa, Av. Prof. Egas Moniz, 1649, 1649-028, Lisboa, Portugal
| | - Ana Catarina Mendes
- Faculdade de Medicina, Instituto de Medicina Molecular, Universidade de Lisboa, Av. Prof. Egas Moniz, 1649, 1649-028, Lisboa, Portugal
| | - Filipa Ribeiro
- Faculdade de Medicina, Instituto de Medicina Molecular, Universidade de Lisboa, Av. Prof. Egas Moniz, 1649, 1649-028, Lisboa, Portugal
| | | | - Rafael Calvão Barbuto
- Hospital Risoleta Tolentino Neves, R. das Gabirobas, 1, 31744-012, Belo Horizonte, MG, Brazil
| | - Felipe Caixeta
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas (ICB), Universidade Federal de Minas Gerais (UFMG), Av. Antonio Carlos, 6627, 31270-901, Belo Horizonte, MG, Brazil
| | - Leandro Souza Nascimento
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas (ICB), Universidade Federal de Minas Gerais (UFMG), Av. Antonio Carlos, 6627, 31270-901, Belo Horizonte, MG, Brazil
| | - Mariana Almeida Oliveira
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas (ICB), Universidade Federal de Minas Gerais (UFMG), Av. Antonio Carlos, 6627, 31270-901, Belo Horizonte, MG, Brazil
| | - Vinícius Dantas Martins
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas (ICB), Universidade Federal de Minas Gerais (UFMG), Av. Antonio Carlos, 6627, 31270-901, Belo Horizonte, MG, Brazil
| | - Gabriela Silveira-Nunes
- Departamento de Medicina, Universidade Federal de Juiz de Fora (UFJF), Av. Doutor Raimundo Monteiro Resende, Governador Valadares, 35010-177, MG, Brazil
| | - Unaí Tupinambás
- Departamento de Clínica Médica, Faculdade de Medicina, Universidade Federal de Minas Gerais, Av. Alfredo Balena, 190, Belo Horizonte, 30130-100, MG, Brazil
| | - Andrea Teixeira-Carvalho
- Laboratório de Biomarcadores, Instituto de Pesquisa René Rachou, Fundação Oswaldo Cruz, FIOCRUZ-MG, Av. Augusto de Lima, 1715, Belo Horizonte, 30190-002, MG, Brazil
| | - Luis Graça
- Faculdade de Medicina, Instituto de Medicina Molecular, Universidade de Lisboa, Av. Prof. Egas Moniz, 1649, 1649-028, Lisboa, Portugal
| | - Ana Maria Caetano Faria
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas (ICB), Universidade Federal de Minas Gerais (UFMG), Av. Antonio Carlos, 6627, 31270-901, Belo Horizonte, MG, Brazil
| |
Collapse
|
4
|
Scala R, Renda T, Bambina S, Guidelli L, Arniani S, Carrassa L, Oczkowski S. Oxygenation indices and early prediction of outcome in hypoxemic patients with COVID-19 pneumonia requiring noninvasive respiratory support in pulmonary intermediate care unit. Pneumonia (Nathan) 2024; 16:22. [PMID: 39582005 PMCID: PMC11587655 DOI: 10.1186/s41479-024-00145-9] [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: 05/29/2024] [Accepted: 08/26/2024] [Indexed: 11/26/2024] Open
Abstract
BACKGROUND Early prediction of non-invasive respiratory therapy (NIRT) failure is crucial to avoid needless prolongation of respiratory support and delayed endotracheal intubation. Data comparing the predictive value of oxygenation indices (OI) in COVID-19 receiving NIRT are scant. The aim of this monocentric retrospective study of prospectively collected data was to assess the effectiveness of different OI in predicting NIRT outcome at baseline (t0), 12 h (t12) and 24 h (t24) of treatment in hypoxemic patients with COVID-19-related pneumonia, managed in a Pulmonary Intermediate Care Unit (October 2020-June 2021). METHODS We assessed the predictive value of SpO2/FiO2, PaO2/FiO2, standardised PaO2/FiO2 ratio (s-PaO2/FiO2), respiratory index (RI), arterial-alveolar oxygen gradient (a-ADO2), age adjusted arterial-alveolar oxygen ratio (adj-a-ADO2D). Receiver operating characteristics (ROC), AUC and best sensitivity-specificity cut-off values were calculated at t0, t12, t24. NIRT failure risk was adjusted for non-oxygenation predictors. RESULTS Among 590 patients with COVID-19 infection, 368 met the eligibility criteria for inclusion in the study [mean (CI95%): PaO2/FiO2 214(206,8-221,9); PaCO2 mean 32,9 mmHg,(32,4-33,4)]. NIRT failure and hospital mortality rate were 23,4% and 19,6%, respectively. Older age, male gender, agitation/confusion, need for sedation, inability to tolerate prone positioning were independent predictors of NIRT failure. SpO2/FiO2, a-ADO2 and adj-aADO2 at t12 and t24, PaO2/FiO2 and RI at t24 were associated with NIRT failure. Prognostic predictivity of OI increased from t0 to t24. Greater ROC-AUC values were obtained with SpO2/FiO2 0,662 (0,60-0,72) (t0), PaO2/FiO2 0,697 (0,63-0,76) (t12) and s-PaO2/FiO2 0,769 (0,71-0,83) (t24). NIRT failure was independently predicted by PaO2/FiO2, s-PaO2/FiO2 and RI at any observation time and by SpO2/FiO2 and O2 gradients respectively at t0 and t24. SaO2/FiO2 ≤ 300 (t0), PaO2/FiO2 ≤ 151,7 (t12) and s-PaO2/FiO2 ≤ 160,4 (t24) turned out to be the best predictors of NIRT outcome. CONCLUSIONS OI showed different effectiveness in predicting NIRT failure within 24 h of treatment in COVID-19 related pneumonia. This may be due to the multi-factorial pathophysiology of hypoxemia. Our study empathises furthermore the role of non-oxygenation-related parameters in contributing to the outcome. These findings may be useful to build a predictive model also in no COVID-19 related hypoxemic pneumonia.
Collapse
Affiliation(s)
- Raffaele Scala
- Pulmonology and PIMCU, Cardio-Toraco-Neurovascular Department, S. Donato Hospital, Arezzo Usl Toscana Sudest, Via Nenni, 20, Arezzo, 52100, Italy.
| | - Teresa Renda
- Pulmonology and PIMCU, Cardio-Toraco-Neurovascular Department, S. Donato Hospital, Arezzo Usl Toscana Sudest, Via Nenni, 20, Arezzo, 52100, Italy
| | - Sonia Bambina
- Pulmonology and PIMCU, Cardio-Toraco-Neurovascular Department, S. Donato Hospital, Arezzo Usl Toscana Sudest, Via Nenni, 20, Arezzo, 52100, Italy
| | - Luca Guidelli
- Pulmonology and PIMCU, Cardio-Toraco-Neurovascular Department, S. Donato Hospital, Arezzo Usl Toscana Sudest, Via Nenni, 20, Arezzo, 52100, Italy
| | - Stefania Arniani
- Demographic and Epidemiologic Section, Prevention Department, S. Donato Hospital, Arezzo Usl Toscana Sudest, Arezzo, Italy
| | - Laura Carrassa
- Department of Oncology, S. Donato Hospital, Arezzo Usl Toscana Sudest, Arezzo, Italy
| | - Simon Oczkowski
- Department of Medicine, Division of Critical Care, McMaster University, Hamilton, ON, Canada
- Department of Health Research Methods, Evidence, and Impact, McMaster University, Hamilton, ON, Canada
| |
Collapse
|
5
|
Lyne T, Camporota L, Montgomery H. Contribution of intrapulmonary shunt to the pathogenesis of profound hypoxaemia in viral infection: a mechanistic discussion with an illustrative case. J Intensive Care Soc 2024; 25:427-431. [PMID: 39524067 PMCID: PMC11549710 DOI: 10.1177/17511437241267745] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2024] Open
Abstract
Background The formation of anastomoses between the pulmonary arteries and pulmonary veins, or the pulmonary and the bronchial circulation, is part of normal foetal lung development. They persist in approximately 30% of adults at rest, and open in almost all adults during exertion. Blood flowing through these anastomoses bypasses the alveolar surface and increases in such shunting can thus cause hypoxaemia. This is now known to contribute to the pathogenesis of hypoxaemia in COVID-19 disease. We here provide evidence to support a similar role in influenza A infection. Illustrative case presentation We describe a case of influenza A infection associated with severe hypoxaemia, poorly responsive to supplemental oxygen and which worsened following the application of continuous positive airway pressure (CPAP), despite the presence of a normal physical examination, chest radiograph and echocardiogram. This combination suggests a significant intrapulmonary (extra-alveolar) shunt as a cause of the severe hypoxaemia. The shunt fraction was estimated to be approximately 57%. Discussion and conclusion Intrapulmonary vascular shunts can contribute substantially to hypoxaemia in viral infection. Seeking to understand the pathogenesis of observed hypoxaemia can help guide respiratory therapy. Mechanistic research may suggest novel therapeutic targets which could assist in avoiding intubation and mechanical ventilatory support.
Collapse
Affiliation(s)
- Tom Lyne
- Whittington Hospital Intensive Care Unit, Department of Intensive Care, London, UK
| | - Luigi Camporota
- Department of Adult Critical Care, Guy’s and St Thomas’ NHS Foundation Trust, London, UK
- Centre for Human and Applied Physiological Sciences, King’s College London, London, UK
| | - Hugh Montgomery
- Whittington Hospital Intensive Care Unit, Department of Intensive Care, London, UK
- Department of Medicine, University College London, London, UK
| |
Collapse
|
6
|
Wendt R, Macholz M, Kalbitz S, Herrmann N, Herbst V, Hammes T, Kai M, Ankersmit HJ, Beige J, Lübbert C, Graf A, Scherberich J. Serum uromodulin associates with kidney function and outcome in a cohort of hospitalised COVID-19 patients. Sci Rep 2024; 14:25420. [PMID: 39455668 PMCID: PMC11512065 DOI: 10.1038/s41598-024-76372-3] [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: 03/19/2024] [Accepted: 10/14/2024] [Indexed: 10/28/2024] Open
Abstract
This study investigates the prevalence and evaluates the prognostic implications of acute kidney injury (AKI) in COVID-19 patients, with a novel emphasis on the evaluation of serum uromodulin (sUmod) as a potential kidney-specific biomarker. A cohort of hospitalised COVID-19 patients (n = 378) was examined for AKI using standard criteria. In addition to traditional urinary biomarkers, sUmod levels were analysed. Univariable and multivariable regression models were employed to evaluate the association of sUmod and AKI and in-hospital mortality. Levels of sUmod were significantly lower in patients with CKD (91.8 ± 60.7 ng/ml) compared to patients with normal kidney function (204.7 ± 91.7 ng/ml; p < 0.001). 151 patients (40.0%) presented with AKI at the time of hospital admission or developed an AKI during hospitalization. 116 patients (76.8%) had an AKI already at the time of hospital admission. COVID-19 patients with AKI had significantly lower levels of sUmod compared to patients without AKI during hospitalisation (124.8 ± 79.5 ng/ml) vs 214.6 ± 92.3 ng/ml; p < 0.001). The in-hospital mortality rate in this cohort of COVID-19 patients was 15.3%. Patients with AKI had a higher probability for in-hospital death (OR 5.6, CI 1.76 to 17.881, p = 0.004). Patients who died during hospital stay, had significantly lower sUmod levels (129.14 ± 89.56 ng/ml) compared to patients surviving hospitalisation (187.71 ± 96,64 ng/ml; p < 0.001). AKI is frequently associated with COVID-19 in hospitalized patients. Serum uromodulin may emerge as a promising biomarker for AKI in COVID-19 patients. Further research is warranted to explore its clinical application and refine risk stratification in this patient population.
Collapse
Affiliation(s)
- Ralph Wendt
- Department of Nephrology, St. Georg Hospital, Delitzscher Str. 141, 04129, Leipzig, Germany.
| | - Martin Macholz
- Department of Infectious Diseases and Tropical Medicine, St. Georg Hospital, Delitzscher Str. 141, 04129, Leipzig, Germany
| | - Sven Kalbitz
- Department of Infectious Diseases and Tropical Medicine, St. Georg Hospital, Delitzscher Str. 141, 04129, Leipzig, Germany
| | - Nadja Herrmann
- Department of Finance and Controlling, St. Georg Hospital, Delitzscher Str. 141, 04129, Leipzig, Germany
| | - Victor Herbst
- Institute for Experimental Immunology, Affiliated to EUROIMMUN Medizinische Labordiagnostika AG, Seekamp 31, 23560, Lübeck, Germany
| | - Tabea Hammes
- Institute for Experimental Immunology, Affiliated to EUROIMMUN Medizinische Labordiagnostika AG, Seekamp 31, 23560, Lübeck, Germany
| | - Marco Kai
- Institute for Experimental Immunology, Affiliated to EUROIMMUN Medizinische Labordiagnostika AG, Seekamp 31, 23560, Lübeck, Germany
| | - Hendrik Jan Ankersmit
- Clinic of Thoracic Surgery, Medical University of Vienna, Spitalgasse 23, 1090, Vienna, Austria
| | - Joachim Beige
- Kuratorium for Dialysis and Transplantation (KfH) Renal Unit, Hospital St. Georg, Delitzscher Str. 141, 04129, Leipzig, Germany
- Martin-Luther University Halle/Wittenberg, Halle, Germany
| | - Christoph Lübbert
- Department of Infectious Diseases and Tropical Medicine, St. Georg Hospital, Delitzscher Str. 141, 04129, Leipzig, Germany
- Division of Infectious Diseases and Tropical Medicine, Leipzig University Hospital, Liebigstr. 20, 04103, Leipzig, Germany
| | - Alexandra Graf
- Institute of Medical Statistics, Center for Medical Data Science, Medical University of Vienna, Spitalgasse 23, 1090, Vienna, Austria
| | - Jürgen Scherberich
- Department of Nephrology and Clinical Immunology, Klinikum München, 81545, Munich, Germany
| |
Collapse
|
7
|
Margarita V, Pau MC, Carboni G, Mannu F, Turrini F, Rappelli P, Pantaleo A, Dessì D, Zinellu E, Piras B, Fois AG, Rubino S, Pirina P, Fiori PL. Comparison of microbiological and molecular diagnosis for identification of respiratory secondary infections in COVID-19 patients and their antimicrobial resistance patterns. Diagn Microbiol Infect Dis 2024; 110:116479. [PMID: 39116653 DOI: 10.1016/j.diagmicrobio.2024.116479] [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: 07/16/2024] [Revised: 07/30/2024] [Accepted: 08/01/2024] [Indexed: 08/10/2024]
Abstract
We report the use of a new multiplex Real-Time PCR platform to simultaneously identify 24 pathogens and 3 antimicrobial-resistance genes directly from respiratory samples of COVID-19 patients. Results were compared to culture-based diagnosis. Secondary infections were detected in 60% of COVID-19 patients by molecular analysis and 73% by microbiological assays, with no significant differences in accuracy, indicating Gram-negative bacteria as the predominant species. Among fungal superinfections, Aspergillus spp. were detected by both methods in more than 7% of COVID-19 patients. Oxacillin-resistant S. aureus and carbapenem-resistant K. pneumoniae were highlighted by both methods. Secondary microbial infections in SARS-CoV-2 patients are associated with poor outcomes and an increased risk of death. Since PCR-based tests significantly reduce the turnaround time to 4 hours and 30 minutes (compared to 48 hours for microbial culture), we strongly support the routine use of molecular techniques, in conjunction with microbiological analysis, to identify co/secondary infections.
Collapse
Affiliation(s)
- Valentina Margarita
- Department of Biomedical Sciences, University of Sassari, 07100 Sassari, Italy
| | - Maria Carmina Pau
- Department of Medicine, Surgery and Pharmacy, University of Sassari, 07100 Sassari, Italy
| | - Gavino Carboni
- Department of Biomedical Sciences, University of Sassari, 07100 Sassari, Italy
| | | | | | - Paola Rappelli
- Department of Biomedical Sciences, University of Sassari, 07100 Sassari, Italy; Azienda Ospedaliero Università (AOU), Sassari, 07100 Sassari, Italy
| | - Antonella Pantaleo
- Department of Biomedical Sciences, University of Sassari, 07100 Sassari, Italy
| | - Daniele Dessì
- Department of Biomedical Sciences, University of Sassari, 07100 Sassari, Italy
| | | | - Barbara Piras
- Azienda Ospedaliero Università (AOU), Sassari, 07100 Sassari, Italy
| | - Alessandro G Fois
- Department of Medicine, Surgery and Pharmacy, University of Sassari, 07100 Sassari, Italy; Azienda Ospedaliero Università (AOU), Sassari, 07100 Sassari, Italy
| | - Salvatore Rubino
- Department of Biomedical Sciences, University of Sassari, 07100 Sassari, Italy; Azienda Ospedaliero Università (AOU), Sassari, 07100 Sassari, Italy
| | - Pietro Pirina
- Department of Medicine, Surgery and Pharmacy, University of Sassari, 07100 Sassari, Italy; Azienda Ospedaliero Università (AOU), Sassari, 07100 Sassari, Italy
| | - Pier Luigi Fiori
- Department of Biomedical Sciences, University of Sassari, 07100 Sassari, Italy; Azienda Ospedaliero Università (AOU), Sassari, 07100 Sassari, Italy
| |
Collapse
|
8
|
Soni S, Antonescu L, Ro K, Horowitz JC, Mebratu YA, Nho RS. Influenza, SARS-CoV-2, and Their Impact on Chronic Lung Diseases and Fibrosis: Exploring Therapeutic Options. THE AMERICAN JOURNAL OF PATHOLOGY 2024; 194:1807-1822. [PMID: 39032604 PMCID: PMC11423761 DOI: 10.1016/j.ajpath.2024.06.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/15/2024] [Revised: 06/11/2024] [Accepted: 06/26/2024] [Indexed: 07/23/2024]
Abstract
Respiratory tract infections represent a significant global public health concern, disproportionately affecting vulnerable populations such as children, the elderly, and immunocompromised individuals. RNA viruses, particularly influenza viruses and coronaviruses, significantly contribute to respiratory illnesses, especially in immunosuppressed and elderly individuals. Influenza A viruses (IAVs) and severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) continue to pose global health threats due to their capacity to cause annual epidemics, with profound implications for public health. In addition, the increase in global life expectancy is influencing the dynamics and outcomes of respiratory viral infections. Understanding the molecular mechanisms by which IAVs and SARS-CoV-2 contribute to lung disease progression is therefore crucial. The aim of this review is to comprehensively explore the impact of IAVs and SARS-CoV-2 on chronic lung diseases, with a specific focus on pulmonary fibrosis in the elderly. It also outlines potential preventive and therapeutic strategies and suggests directions for future research.
Collapse
Affiliation(s)
- Sourabh Soni
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Internal Medicine and The Davis Heart and Lung Research Institute, The Ohio State University, Columbus, Ohio
| | - Laura Antonescu
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Internal Medicine and The Davis Heart and Lung Research Institute, The Ohio State University, Columbus, Ohio
| | - Kaylin Ro
- Scripps Research Institute, San Diego, California
| | - Jeffrey C Horowitz
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Internal Medicine and The Davis Heart and Lung Research Institute, The Ohio State University, Columbus, Ohio
| | - Yohannes A Mebratu
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Internal Medicine and The Davis Heart and Lung Research Institute, The Ohio State University, Columbus, Ohio.
| | - Richard S Nho
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Internal Medicine and The Davis Heart and Lung Research Institute, The Ohio State University, Columbus, Ohio.
| |
Collapse
|
9
|
Casitas R, Galera R, Torres-Vargas M, Garcia-Tovar S, Alfaro E, Díaz-Garcia E, Martinez-Cerón E, Garcia-Garcia M, Torres I, Núñez-Fernández M, Fernández-Villar A, Fernández-Velilla M, Añón JM, Cubillos-Zapata C, García-Río F. Medium-Term Disability and Long-Term Functional Impairment Persistence in Survivors of Severe COVID-19 ARDS: Clinical and Physiological Insights. Arch Bronconeumol 2024; 60:619-626. [PMID: 38853119 DOI: 10.1016/j.arbres.2024.05.021] [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: 03/11/2024] [Revised: 05/09/2024] [Accepted: 05/14/2024] [Indexed: 06/11/2024]
Abstract
BACKGROUND Although the medium- and long-term sequelae of survivor of acute respiratory distress syndrome (ARDS) of any cause have been documented, little is known about the way in which COVID-19-induced ARDS affects functional disability and exercise components. Our aims were to examine the medium-term disability in severe COVID-19-associated ARDS survivors, delineate pathophysiological changes contributing to their exercise intolerance, and explore its utility in predicting long-term functional impairment persistence. METHODS We studied 108 consecutive subjects with severe COVID-19 ARDS who remained alive 6 months after intensive care unit (ICU) discharge. Lung morphology was assessed with chest non-contrast CT scans and CT angiography. Functional evaluation included spirometry, plethysmography, muscle strength, and diffusion capacity, with assessment of gas exchange components through diffusing capacity of nitric oxide. Disability was assessed through an incremental exercise test, and measurements were repeated 12 and 24 months later in patients with functional impairments. RESULTS At 6 months after ICU discharge, a notable dissociation between morphological and clinical-functional sequelae was identified. Moderate-severe disability was present in 47% of patients and these subjects had greater limitation of ventilatory mechanics and gas exchange, as well as greater symptomatic perception during exercise and a probable associated cardiac limitation. Female sex, hypothyroidism, reduced membrane diffusion component, lower functional residual capacity, and high-attenuation lung volume were independently associated with the presence of moderate-severe functional disability, which in turn was related to higher frequency and greater intensity of dyspnea and worse quality of life. Out of the 71 patients with reduced lung volumes or diffusion capacity at 6 months post-ICU discharge, only 19 maintained a restrictive disorder associated with gas exchange impairment at 24 months post-discharge. In these patients, 6-month values for diffusion membrane component, maximal oxygen uptake, ventilatory equivalent for CO2, and dead space to tidal volume ratio were identified as independent risk factors for persistence of long-term functional sequelae. CONCLUSIONS Less than half of survivors of COVID-19 ARDS have moderate-severe disability in the medium term, identifying several risk factors. In turn, diffusion membrane component and exercise tolerance at 6-month ICU discharge are independently associated with the persistence of long-term functional sequelae.
Collapse
Affiliation(s)
- Raquel Casitas
- Servicio de Neumología, Hospital Universitario La Paz, IdiPAZ, Madrid, Spain; Centro de Investigación Biomédica en Red en Enfermedades Respiratorias (CIBERES), Madrid, Spain; Departamento de Medicina, Universidad Autónoma de Madrid, Spain
| | - Raúl Galera
- Servicio de Neumología, Hospital Universitario La Paz, IdiPAZ, Madrid, Spain; Centro de Investigación Biomédica en Red en Enfermedades Respiratorias (CIBERES), Madrid, Spain; Grupo de Enfermedades Respiratorias, IdiPAZ, Madrid, Spain
| | - María Torres-Vargas
- Servicio de Neumología, Hospital Universitario La Paz, IdiPAZ, Madrid, Spain
| | - Sara Garcia-Tovar
- Centro de Investigación Biomédica en Red en Enfermedades Respiratorias (CIBERES), Madrid, Spain; Grupo de Enfermedades Respiratorias, IdiPAZ, Madrid, Spain
| | - Enrique Alfaro
- Centro de Investigación Biomédica en Red en Enfermedades Respiratorias (CIBERES), Madrid, Spain; Grupo de Enfermedades Respiratorias, IdiPAZ, Madrid, Spain
| | - Elena Díaz-Garcia
- Centro de Investigación Biomédica en Red en Enfermedades Respiratorias (CIBERES), Madrid, Spain; Grupo de Enfermedades Respiratorias, IdiPAZ, Madrid, Spain
| | - Elisabet Martinez-Cerón
- Servicio de Neumología, Hospital Universitario La Paz, IdiPAZ, Madrid, Spain; Centro de Investigación Biomédica en Red en Enfermedades Respiratorias (CIBERES), Madrid, Spain; Grupo de Enfermedades Respiratorias, IdiPAZ, Madrid, Spain
| | - Miguel Garcia-Garcia
- Departamento de Análisis Económico y Economía Cuantitativa, Universidad Complutense de Madrid, Spain
| | - Isabel Torres
- Servicio de Radiodiagnóstico, Hospital Universitario La Paz, IdiPAZ, Madrid, Spain
| | - Marta Núñez-Fernández
- Servicio de Neumología, Complejo Hospitalario Universitario de Vigo, Spain; NeumoVigo I+i, Galicia Sur Health Research Institute (IIGS), Vigo, Spain
| | - Alberto Fernández-Villar
- Servicio de Neumología, Complejo Hospitalario Universitario de Vigo, Spain; NeumoVigo I+i, Galicia Sur Health Research Institute (IIGS), Vigo, Spain
| | | | - José Manuel Añón
- Centro de Investigación Biomédica en Red en Enfermedades Respiratorias (CIBERES), Madrid, Spain; Servicio de Medicina Intensiva, Hospital Universitario La Paz, IdiPAZ, Madrid, Spain
| | - Carolina Cubillos-Zapata
- Centro de Investigación Biomédica en Red en Enfermedades Respiratorias (CIBERES), Madrid, Spain; Grupo de Enfermedades Respiratorias, IdiPAZ, Madrid, Spain
| | - Francisco García-Río
- Servicio de Neumología, Hospital Universitario La Paz, IdiPAZ, Madrid, Spain; Centro de Investigación Biomédica en Red en Enfermedades Respiratorias (CIBERES), Madrid, Spain; Departamento de Medicina, Universidad Autónoma de Madrid, Spain; Grupo de Enfermedades Respiratorias, IdiPAZ, Madrid, Spain.
| |
Collapse
|
10
|
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
|
11
|
Nielsen FM, Klitgaard TL, Granholm A, Lange T, Perner A, Schjørring OL, Rasmussen BS. Lower or Higher Oxygenation Targets in Patients With COVID-19 in the ICU: A Secondary Bayesian Analysis of the Handling Oxygenation Targets in COVID-19 Trial. Chest 2024:S0012-3692(24)05164-X. [PMID: 39303806 DOI: 10.1016/j.chest.2024.08.055] [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: 03/23/2024] [Revised: 08/23/2024] [Accepted: 08/27/2024] [Indexed: 09/22/2024] Open
Abstract
BACKGROUND In the Handling Oxygenation Targets in COVID-19 (HOT-COVID) trial, a Pao2 target of 60 mm Hg compared with 90 mm Hg resulted in more days alive without life support at 90 days in adults in the ICU with COVID-19 and hypoxemia. The trial was stopped after enrolling 726 of 780 planned patients because of slow recruitment. Herein, we present the preplanned Bayesian analysis of the HOT-COVID trial. RESEARCH QUESTION What are the probabilities of any benefits and of clinically relevant benefits resulting from a Pao2 target of 60 mm Hg vs 90 mm Hg in adult patients with COVID-19 and hypoxemia in the ICU and does heterogeneity of treatment effects (HTE) exist according to selected baseline characteristics?. STUDY DESIGN AND METHODS We analyzed days alive without life support and 90-day mortality in the HOT-COVID intention-to-treat population (n = 697) using Bayesian general linear models to assess probabilities for benefit or harm, including clinically relevant benefits defined as > 1 day alive without life support and > 2 percentage points lower 90-day mortality. HTE was evaluated based on baseline Sequential Organ Failure Assessment scores, Pao2 to Fio2 ratio, norepinephrine doses, and lactate concentrations. RESULTS The mean difference in days alive without life support was 5.7 days (95% credible interval [CrI], 0.2-11.2), with a 95.2% probability of clinically relevant benefit and a 98.0% probability of any benefit from the lower Pao2 target. The risk difference in 90-day mortality was -4.6 percentage points (95% CrI, -11.8 to 2.6 percentage points), with a 76.5% probability of a clinically relevant benefit from the lower target. HTE analyses revealed potential interaction with baseline norepinephrine dose and lactate concentrations for both outcomes. INTERPRETATION In patients with COVID-19 and hypoxemia in the ICU, we found a high probability for a clinically relevant benefit of targeting a Pao2 of 60 mm Hg vs 90 mm Hg on number of days alive without life support. CLINICAL TRIAL REGISTRY ClinicalTrials.gov; No.: NCT04425031; URL: www. CLINICALTRIALS gov.
Collapse
Affiliation(s)
- Frederik Mølgaard Nielsen
- Department of Anesthesia and Intensive Care, Aalborg University Hospital, Aalborg, Denmark; Collaboration for Research in Intensive Care, Copenhagen, Denmark.
| | - Thomas Lass Klitgaard
- Department of Anesthesia and Intensive Care, Aalborg University Hospital, Aalborg, Denmark; Collaboration for Research in Intensive Care, Copenhagen, Denmark
| | - Anders Granholm
- Collaboration for Research in Intensive Care, Copenhagen, Denmark; Department of Intensive Care, Copenhagen University Hospital - Rigshospitalet, Copenhagen, Denmark
| | - Theis Lange
- Collaboration for Research in Intensive Care, Copenhagen, Denmark; Section of Biostatistics, Department of Public Health, University of Copenhagen, Copenhagen, Denmark
| | - Anders Perner
- Collaboration for Research in Intensive Care, Copenhagen, Denmark; Department of Intensive Care, Copenhagen University Hospital - Rigshospitalet, Copenhagen, Denmark; Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Olav Lilleholt Schjørring
- Department of Anesthesia and Intensive Care, Aalborg University Hospital, Aalborg, Denmark; Collaboration for Research in Intensive Care, Copenhagen, Denmark
| | - Bodil Steen Rasmussen
- Department of Anesthesia and Intensive Care, Aalborg University Hospital, Aalborg, Denmark; Collaboration for Research in Intensive Care, Copenhagen, Denmark
| |
Collapse
|
12
|
Molfino A, Anastasi E, Assanto E, Toccini L, Imbimbo G, Gigante A, Viggiani V, Farina A, Picconi O, Angeloni A, Muscaritoli M. Association between serum levels of GDF-15, suPAR, PIVKA-II, sdLDL and clinical outcomes in hospitalized COVID-19 patients. Intern Emerg Med 2024; 19:1557-1566. [PMID: 38700782 PMCID: PMC11405538 DOI: 10.1007/s11739-024-03630-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/20/2024] [Accepted: 04/24/2024] [Indexed: 09/17/2024]
Abstract
To quantify the circulating levels of novel serum biomarkers including GDF-15, PIVKA-II, sdLDL, suPAR, and of CRP in hospitalized COVID-19 patients compared with healthy subjects, and to evaluate their association(s) with outcomes in COVID-19. We considered patients with confirmed COVID-19, hospitalized in an Internal Medicine ward. The clinical characteristics were collected, including the number and type of comorbidities. Serum levels of GDF-15, PIVKA-II, suPAR, sdLDL, as well as CRP were measured. As outcomes, we considered Intensive Care Unit (ICU) transfer or death, as well as the length of stay (days) and in-hospital complications. Data were statistically analyzed, as appropriate, and a p value < 0.05 was considered significant. Ninety-three patients and 20 healthy controls were enrolled. COVID-19 patients vs. controls showed higher median levels of GDF-15 (p < 0.0001), PIVKA-II (p < 0.0001) and sdLDL (p = 0.0002), whereas no difference was observed for suPAR. In COVID-19 patients, the most frequent comorbidities were arterial hypertension (62.4%) and cardiovascular disease (30.1%). GDF-15 levels positively correlated with age (r = 0.433, p < 0.0001), and this correlation was confirmed for suPAR (r = 0.308, p = 0.003) and CRP (Rho = 0.40 p < 0.0001), but not for PIVKA-II and sdLDL. Higher GDF-15 levels were associated with a higher number of comorbidities (p = 0.021). The median length of stay was 22 (15; 30) days. During hospitalization, 15 patients (16%) were ICU transferred, and 6 (6.45%) died. GDF-15 serum levels correlated with the length of stay (rho = 0.27 p = 0.010), and were associated with ICU transfer or death (p = 0.003), as well as PIVKA-II (p = 0.038) and CRP (p < 0.001). Moreover, higher GDF-15 and PIVKA-II serum levels were associated with infectious complications (p = 0.008 and p = 0.017, respectively). In this cohort of hospitalized COVID-19 patients, novel inflammatory biomarkers, including GDF-15, suPAR and PIVKA II were associated with some patient's clinical characteristics, complications, and poor outcomes.
Collapse
Affiliation(s)
- Alessio Molfino
- Department of Translational and Precision Medicine, Sapienza University of Rome, Rome, Italy.
| | - Emanuela Anastasi
- Department of Experimental Medicine, Sapienza University of Rome, Rome, Italy.
- Department of Molecular Medicine, Sapienza University of Rome, Rome, Italy.
| | - Eleonora Assanto
- Department of Translational and Precision Medicine, Sapienza University of Rome, Rome, Italy
| | - Ludovica Toccini
- Department of Translational and Precision Medicine, Sapienza University of Rome, Rome, Italy
| | - Giovanni Imbimbo
- Department of Translational and Precision Medicine, Sapienza University of Rome, Rome, Italy
| | - Antonietta Gigante
- Department of Translational and Precision Medicine, Sapienza University of Rome, Rome, Italy
| | - Valentina Viggiani
- Department of Experimental Medicine, Sapienza University of Rome, Rome, Italy
| | - Antonella Farina
- Department of Experimental Medicine, Sapienza University of Rome, Rome, Italy
| | - Orietta Picconi
- National HIV/AIDS Center, Istituto Superiore Di Sanità, Rome, Italy
| | - Antonio Angeloni
- Department of Experimental Medicine, Sapienza University of Rome, Rome, Italy
| | - Maurizio Muscaritoli
- Department of Translational and Precision Medicine, Sapienza University of Rome, Rome, Italy
| |
Collapse
|
13
|
Michailides C, Papantoniou K, Paraskevas T, Lagadinou M, Marangos M, Kavvousanos M, Michailidou P, Velissaris D. Multisystem Inflammatory Syndrome of the Adults (MIS-A) - The undercover threat for young adults. A systematic review and meta-analysis of medical cases. LE INFEZIONI IN MEDICINA 2024; 32:272-279. [PMID: 39282538 PMCID: PMC11392540 DOI: 10.53854/liim-3203-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/21/2024] [Accepted: 07/30/2024] [Indexed: 09/19/2024]
Abstract
Background COVID-19 related syndromes are not yet well described and understood. Multisystem inflammatory syndrome of the adults (MIS-A) is a recently characterized syndrome affecting multiple organs of young adults, causing serious complications, even shock and death. Objectives To determine the clinical characteristics, course, and complications of MIS-A in a systematic way and summarize currently used treatments. Methods Literature search in March 2023 in PubMed and Scopus databases. Case reports and case-series that fulfilled the CDC criteria for MIS-A were eligible for inclusion. Results A total of 71 patients from 60 reports were included. 66% of the patients were male and the mean age of the synthetic cohort was 32.9 years old. The majority (70.4%) of the enrolled cases had no significant medical history. MIS-A was diagnosed after a median of 4 weeks period. All but two patients presented with cardiac symptoms, while the most common secondary diagnostic criterion was abdominal pain, vomiting or diarrhea followed by shock or hypotension. Heart failure therapy and immunomodulation were used as therapeutic options. Although more than half of the cohort was admitted to the Intensive Care Unit (ICU) (n=39) only 4 deaths were reported. Conclusion MIS-A can affect patients independently of age, sex, and co-morbidity status, resulting in serious complications, often including severe cardiac disease, shock, acute kidney injury and sometimes death. It can occur immediately after SARS-CoV-2 acute infection until two months later, usually manifesting four weeks after acute disease.
Collapse
Affiliation(s)
| | | | | | - Maria Lagadinou
- Department of Internal Medicine, Patras University Hospital, Patras, Greece
| | - Markos Marangos
- Department of Internal Medicine, Patras University Hospital, Patras, Greece
| | | | | | | |
Collapse
|
14
|
Arévalo-Cortés A, Rodriguez-Pinto D, Aguilar-Ayala L. Evidence for Molecular Mimicry between SARS-CoV-2 and Human Antigens: Implications for Autoimmunity in COVID-19. Autoimmune Dis 2024; 2024:8359683. [PMID: 39247752 PMCID: PMC11380714 DOI: 10.1155/2024/8359683] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2024] [Accepted: 08/26/2024] [Indexed: 09/10/2024] Open
Abstract
As for other viral diseases, the mechanisms behind the apparent relationship between COVID-19 and autoimmunity are yet to be clearly defined. Molecular mimicry, the existence of sequence and/or conformational homology between viral and human antigens, could be an important contributing factor. Here, we review the accumulated evidence supporting the occurrence of mimicry between SARS-CoV-2 and human proteins. Both bioinformatic approaches and antibody cross-reactions have yielded a significant magnitude of mimicry events, far more common than expected to happen by chance. The clinical implication of this phenomenon is ample since many of the identified antigens may participate in COVID-19 pathophysiology or are targets of autoimmune diseases. Thus, autoimmunity related to COVID-19 may be partially explained by molecular mimicry and further research designed specifically to address this possibility is needed.
Collapse
Affiliation(s)
| | - Daniel Rodriguez-Pinto
- Department of Health Sciences Faculty of Health Sciences Universidad Técnica Particular de Loja, Loja 110108, Ecuador
| | - Leonardo Aguilar-Ayala
- Department of Health Sciences Faculty of Health Sciences Universidad Técnica Particular de Loja, Loja 110108, Ecuador
| |
Collapse
|
15
|
Yang Z, Gao Y, Zhao L, Lv X, Du Y. Molecular mechanisms of Sepsis attacking the immune system and solid organs. Front Med (Lausanne) 2024; 11:1429370. [PMID: 39267971 PMCID: PMC11390691 DOI: 10.3389/fmed.2024.1429370] [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: 05/13/2024] [Accepted: 08/15/2024] [Indexed: 09/15/2024] Open
Abstract
Remarkable progress has been achieved in sepsis treatment in recent times, the mortality rate of sepsis has experienced a gradual decline as a result of the prompt administration of antibiotics, fluid resuscitation, and the implementation of various therapies aimed at supporting multiple organ functions. However, there is still significant mortality and room for improvement. The mortality rate for septic patients, 22.5%, is still unacceptably high, accounting for 19.7% of all global deaths. Therefore, it is crucial to thoroughly comprehend the pathogenesis of sepsis in order to enhance clinical diagnosis and treatment methods. Here, we summarized classic mechanisms of sepsis progression, activation of signal pathways, mitochondrial quality control, imbalance of pro-and anti- inflammation response, diseminated intravascular coagulation (DIC), cell death, presented the latest research findings for each mechanism and identify potential therapeutic targets within each mechanism.
Collapse
Affiliation(s)
- Zhaoyun Yang
- Department of Respiratory and Critical Care Medicine, The Second Hospital of Jilin University, Changchun, China
- Department of Rehabilitation, School of Nursing, Jilin University, Changchun, China
| | - Yan Gao
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Biomedical Sciences, College of Chemistry and Chemical Engineering, Hunan University, Changsha, China
| | - Lijing Zhao
- Department of Rehabilitation, School of Nursing, Jilin University, Changchun, China
| | - Xuejiao Lv
- Department of Respiratory and Critical Care Medicine, The Second Hospital of Jilin University, Changchun, China
| | - Yanwei Du
- Department of Rehabilitation, School of Nursing, Jilin University, Changchun, China
| |
Collapse
|
16
|
Wiebe M, Mackay M, Krishnan R, Tian J, Larsson J, Modanloo S, Job McIntosh C, Sztym M, Elton-Smith G, Rose A, Ho C, Greenshaw A, Cao B, Chan A, Hayward J. Feasibility characteristics of wrist-worn fitness trackers in health status monitoring for post-COVID patients in remote and rural areas. PLOS DIGITAL HEALTH 2024; 3:e0000571. [PMID: 39172978 PMCID: PMC11340956 DOI: 10.1371/journal.pdig.0000571] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/29/2023] [Accepted: 07/04/2024] [Indexed: 08/24/2024]
Abstract
INTRODUCTION Common, consumer-grade biosensors mounted on fitness trackers and smartwatches can measure an array of biometrics that have potential utility in post-discharge medical monitoring, especially in remote/rural communities. The feasibility characteristics for wrist-worn biosensors are poorly described for post-COVID conditions and rural populations. METHODS We prospectively recruited patients in rural communities who were enrolled in an at-home rehabilitation program for post-COVID conditions. They were asked to wear a FitBit Charge 2 device and biosensor parameters were analyzed [e.g. heart rate, sleep, and activity]. Electronic patient reported outcome measures [E-PROMS] for mental [bi-weekly] and physical [daily] symptoms were collected using SMS text or email [per patient preference]. Exit surveys and interviews evaluated the patient experience. RESULTS Ten patients were observed for an average of 58 days and half [N = 5] were monitored for 8 weeks or more. Five patients [50%] had been hospitalized with COVID [mean stay = 41 days] and 4 [36%] had required mechanical ventilation. As baseline, patients had moderate to severe levels of anxiety, depression, and stress; fatigue and shortness of breath were the most prevalent physical symptoms. Four patients [40%] already owned a smartwatch. In total, 575 patient days of patient monitoring occurred across 10 patients. Biosensor data was usable for 91.3% of study hours and surveys were completed 82.1% and 78.7% of the time for physical and mental symptoms, respectively. Positive correlations were observed between stress and resting heart rate [r = 0.360, p<0.01], stress and daily steps [r = 0.335, p<0.01], and anxiety and daily steps [r = 0.289, p<0.01]. There was a trend toward negative correlation between sleep time and physical symptom burden [r = -0.211, p = 0.05]. Patients reported an overall positive experience and identified the potential for wearable devices to improve medical safety and access to care. Concerns around data privacy/security were infrequent. CONCLUSIONS We report excellent feasibility characteristics for wrist-worn biosensors and e-PROMS as a possible substrate for multi-modal disease tracking in post-COVID conditions. Adapting consumer-grade wearables for medical use and scalable remote patient monitoring holds great potential.
Collapse
Affiliation(s)
- Madeleine Wiebe
- Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Canada
| | - Marnie Mackay
- Department of Psychiatry, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Canada
| | - Ragur Krishnan
- Department of Electrical and Computer Engineering, Faculty of Engineering University of Alberta, Edmonton, Canada
| | - Julie Tian
- Department of Psychiatry, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Canada
| | - Jakob Larsson
- Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Canada
| | - Setayesh Modanloo
- Department of Psychiatry, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Canada
| | - Christiane Job McIntosh
- Neurosciences, Rehabilitation and Vision Strategic Clinical Network, Alberta Health Services, Edmonton, Canada
| | - Melissa Sztym
- Covenant Health Rural Health Services, Edmonton, Canada
| | | | - Alyssa Rose
- Covenant Health Rural Health Services, Edmonton, Canada
| | - Chester Ho
- Glenrose Rehabilitation Hospital, Edmonton, Canada
- Department of Physical Medicine and Rehabilitation, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Canada
| | - Andrew Greenshaw
- Department of Psychiatry, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Canada
| | - Bo Cao
- Department of Psychiatry, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Canada
| | - Andrew Chan
- Glenrose Rehabilitation Hospital, Edmonton, Canada
| | - Jake Hayward
- Department of Emergency Medicine, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Canada
| |
Collapse
|
17
|
Miner D, Smith K, Wu PT, Price JH, Piscitelli D, Chui K. Pragmatic approach to mobilizing individuals with critical illness due to COVID-19: clinical perspective. Disabil Rehabil 2024; 46:4040-4048. [PMID: 37752855 DOI: 10.1080/09638288.2023.2263370] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2023] [Revised: 09/19/2023] [Accepted: 09/21/2023] [Indexed: 09/28/2023]
Abstract
PURPOSE To provide pragmatic guidance for acute rehabilitation management and implementation of early mobility for individuals with critical illness due to COVID-19. METHODS Clinical perspective developed through reflective clinical practice and narrative review of best available evidence. RESULTS Current clinical practice guidelines do not provide guidance for implementation of early mobility interventions for individuals with critical illness due to COVID-19 who require enhanced ventilatory support or support of inhaled pulmonary artery vasodilators. Many individuals who may benefit from implementation of early mobility interventions are excluded by strict interpretation of current guidelines. CONCLUSIONS Risk vs benefit of implementing early mobility interventions in individuals with critical illness due to COVID-19 can be mitigated through coordinated efforts of interdisciplinary teams to promote shared decision-making through therapeutic alliances with patients and their families. Clinicians must clearly define the goals of care, understand the limitations of monitoring equipment in the intensive care unit, prepare to titrate levels of oxygen based on an individual's physiologic response to mobility interventions, and help individuals maintain external goal-directed focus of attention to optimize outcomes of early mobility interventions.
Collapse
Affiliation(s)
- Daniel Miner
- Department of Physical Therapy, Carilion Clinic, Radford University Carilion, Radford, VA, USA
| | - Kellen Smith
- Department of Physical Therapy, Carilion Clinic, Radford University, Roanoke, VA, USA
| | - Pei-Tzu Wu
- Department of Physical Therapy, Pacific University, Hillsboro, OR, USA
| | - Justin H Price
- Carilion Clinic, VA Tech Carilion School of Medicine, Roanoke, VA, USA
| | - Daniele Piscitelli
- Department of Physical Therapy, University of Connecticut, Storrs, CT, USA
| | - Kevin Chui
- Department of Physical Therapy, Radford University, Radford, VA, USA
| |
Collapse
|
18
|
Nielsen FM, Klitgaard TL, Bruun NH, Møller MH, Schjørring OL, Rasmussen BS. Lower or higher oxygenation targets in the intensive care unit: an individual patient data meta-analysis. Intensive Care Med 2024; 50:1275-1286. [PMID: 38990335 PMCID: PMC11306534 DOI: 10.1007/s00134-024-07523-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2024] [Accepted: 06/11/2024] [Indexed: 07/12/2024]
Abstract
PURPOSE Optimal oxygenation targets for patients with acute hypoxemic respiratory failure in the intensive care unit (ICU) are not clearly defined due to substantial variability in design of previous trials. This study aimed to perform a pre-specified individual patient data meta-analysis of the Handling Oxygenation Targets in the ICU (HOT-ICU) and the Handling Oxygenation Targets in coronavirus disease 2019 (COVID-19) (HOT-COVID) trials to compare targeting a partial pressure of arterial oxygen (PaO2) of 8-12 kPa in adult ICU patients, assessing both benefits and harms. METHODS We assessed 90-day all-cause mortality and days alive without life support in 90 days using a generalised mixed model. Heterogeneity of treatment effects (HTE) was evaluated in 14 subgroups, and results graded using the Instrument to assess the Credibility of Effect Modification Analyses (ICEMAN). RESULTS At 90 days, mortality was 40.4% (724/1792) in the 8 kPa group and 40.9% (733/1793) in the 12 kPa group (risk ratio, 0.99; 95% confidence interval [CI] 0.92-1.07; P = 0.80). No difference was observed in number of days alive without life support. Subgroup analyses indicated more days alive without life support in COVID-19 patients targeting 8 kPa (P = 0.04) (moderate credibility), and lower mortality (P = 0.03) and more days alive without life support (P = 0.02) in cancer-patients targeting 12 kPa (low credibility). CONCLUSION This study reported no overall differences comparing a PaO2 target of 8-12 kPa on mortality or days alive without life support in 90 days. Subgroup analyses suggested HTE in patients with COVID-19 (moderate credibility) and cancer (low credibility).
Collapse
Affiliation(s)
- Frederik Mølgaard Nielsen
- Department of Anaesthesia and Intensive Care, Aalborg University Hospital, Hobrovej 18-21, 9000, Aalborg, Denmark.
| | - Thomas L Klitgaard
- Department of Anaesthesia and Intensive Care, Aalborg University Hospital, Hobrovej 18-21, 9000, Aalborg, Denmark
| | - Niels Henrik Bruun
- Unit of Clinical Biostatistics, Aalborg University Hospital, Aalborg, Denmark
| | - Morten H Møller
- Department of Intensive Care, Rigshospitalet, Copenhagen, Denmark
| | - Olav L Schjørring
- Department of Anaesthesia and Intensive Care, Aalborg University Hospital, Hobrovej 18-21, 9000, Aalborg, Denmark
| | - Bodil S Rasmussen
- Department of Anaesthesia and Intensive Care, Aalborg University Hospital, Hobrovej 18-21, 9000, Aalborg, Denmark
| |
Collapse
|
19
|
Guarienti FA, Xavier FAC, Ferraz MD, Wagner F, Marinowic DR, da Costa JC, Machado DC. Are COVID-19 Polymorphisms in ACE and ACE2 Prognosis Predictors? Curr Issues Mol Biol 2024; 46:8111-8117. [PMID: 39194697 DOI: 10.3390/cimb46080480] [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: 05/23/2024] [Revised: 06/21/2024] [Accepted: 06/25/2024] [Indexed: 08/29/2024] Open
Abstract
Regardless of the containment of the SARS-CoV-2 pandemic, it remains paramount to comprehensively understand its underlying mechanisms to mitigate potential future health and economic impacts, comparable to those experienced throughout the course of the pandemic. The angiotensin-converting enzyme 2 (ACE2) provides anchorage for SARS-CoV-2 binding, thus implicating that ACE and ACE2 might contribute to the variability in infection severity. This study aimed to elucidate predisposing factors influencing the disease course among people infected by SARS-CoV-2, focusing on angiotensin-converting enzyme (ACE) and ACE2 polymorphisms. Notably, despite similar demographics and comorbidities, COVID-19 patients exhibit substantial differences in prognosis. Genetic polymorphisms in ACE and ACE2 have been implicated in disease progression, prompting our investigation into their role in COVID-19 evolution. Using next-generation sequencing (NGS), we analyzed ACE and ACE2 genes in a sample group comprising six subjects infected by SARS-CoV-2. Our findings revealed a correlation between specific polymorphisms and COVID-19 outcomes. Specifically, ACE and ACE2 intronic deletions were observed in all deceased patients, suggesting a potential association with mortality. These results highlight the significance of genetic factors in shaping the clinical course of COVID-19, emphasizing the importance of further research into the impact of genetic variations on COVID-19 severity.
Collapse
Affiliation(s)
- Fabiana Amaral Guarienti
- Post Graduation Program of Geriatrics and Gerontology, Pontifical Catholic University of Rio Grande do Sul, Porto Alegre 90619, Brazil
- Laboratory of Molecular Biology and Immunology, Brain Institute of Rio Grande do Sul, Porto Alegre 90610, Brazil
| | - Fernando Antônio Costa Xavier
- Laboratory of Molecular Biology and Immunology, Brain Institute of Rio Grande do Sul, Porto Alegre 90610, Brazil
- Post Graduation Program of Medicine and Health Sciences, Pontifical Catholic University of Rio Grande do Sul, Porto Alegre 90619, Brazil
- Laboratory of Precision Medicine and Biotechnology, Brain Institute of Rio Grande do Sul, Porto Alegre 90610, Brazil
| | - Mateus Duarte Ferraz
- Laboratory of Molecular Biology and Immunology, Brain Institute of Rio Grande do Sul, Porto Alegre 90610, Brazil
- Post Graduation Program of Medicine and Health Sciences, Pontifical Catholic University of Rio Grande do Sul, Porto Alegre 90619, Brazil
| | - Fernanda Wagner
- Post Graduation Program of Medicine and Health Sciences, Pontifical Catholic University of Rio Grande do Sul, Porto Alegre 90619, Brazil
- Laboratory of Precision Medicine and Biotechnology, Brain Institute of Rio Grande do Sul, Porto Alegre 90610, Brazil
- School of Medicine, Pontifical Catholic University of Rio Grande do Sul, Porto Alegre 90619, Brazil
| | - Daniel Rodrigo Marinowic
- Post Graduation Program of Geriatrics and Gerontology, Pontifical Catholic University of Rio Grande do Sul, Porto Alegre 90619, Brazil
- Laboratory of Molecular Biology and Immunology, Brain Institute of Rio Grande do Sul, Porto Alegre 90610, Brazil
- Post Graduation Program of Medicine and Health Sciences, Pontifical Catholic University of Rio Grande do Sul, Porto Alegre 90619, Brazil
- Laboratory of Precision Medicine and Biotechnology, Brain Institute of Rio Grande do Sul, Porto Alegre 90610, Brazil
| | - Jaderson Costa da Costa
- Post Graduation Program of Medicine and Health Sciences, Pontifical Catholic University of Rio Grande do Sul, Porto Alegre 90619, Brazil
- Laboratory of Neurosciences and Electrophisiology, Brain Institute of Rio Grande do Sul, Porto Alegre 90610, Brazil
| | - Denise Cantarelli Machado
- Post Graduation Program of Geriatrics and Gerontology, Pontifical Catholic University of Rio Grande do Sul, Porto Alegre 90619, Brazil
- Laboratory of Molecular Biology and Immunology, Brain Institute of Rio Grande do Sul, Porto Alegre 90610, Brazil
- Post Graduation Program of Medicine and Health Sciences, Pontifical Catholic University of Rio Grande do Sul, Porto Alegre 90619, Brazil
| |
Collapse
|
20
|
López-Izquierdo R, Del Pozo Vegas C, Sanz-García A, Mayo Íscar A, Castro Villamor MA, Silva Alvarado E, Gracia Villar S, Dzul López LA, Aparicio Obregón S, Calderon Iglesias R, Soriano JB, Martín-Rodríguez F. Clinical phenotypes and short-term outcomes based on prehospital point-of-care testing and on-scene vital signs. NPJ Digit Med 2024; 7:197. [PMID: 39048671 PMCID: PMC11269726 DOI: 10.1038/s41746-024-01194-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2023] [Accepted: 07/12/2024] [Indexed: 07/27/2024] Open
Abstract
Emergency medical services (EMSs) face critical situations that require patient risk classification based on analytical and vital signs. We aimed to establish clustering-derived phenotypes based on prehospital analytical and vital signs that allow risk stratification. This was a prospective, multicenter, EMS-delivered, ambulance-based cohort study considering six advanced life support units, 38 basic life support units, and four tertiary hospitals in Spain. Adults with unselected acute diseases managed by the EMS and evacuated with discharge priority to emergency departments were considered between January 1, 2020, and June 30, 2023. Prehospital point-of-care testing and on-scene vital signs were used for the unsupervised machine learning method (clustering) to determine the phenotypes. Then phenotypes were compared with the primary outcome (cumulative mortality (all-cause) at 2, 7, and 30 days). A total of 7909 patients were included. The median (IQR) age was 64 (51-80) years, 41% were women, and 26% were living in rural areas. Three clusters were identified: alpha 16.2% (1281 patients), beta 28.8% (2279), and gamma 55% (4349). The mortality rates for alpha, beta and gamma at 2 days were 18.6%, 4.1%, and 0.8%, respectively; at 7 days, were 24.7%, 6.2%, and 1.7%; and at 30 days, were 33%, 10.2%, and 3.2%, respectively. Based on standard vital signs and blood test biomarkers in the prehospital scenario, three clusters were identified: alpha (high-risk), beta and gamma (medium- and low-risk, respectively). This permits the EMS system to quickly identify patients who are potentially compromised and to proactively implement the necessary interventions.
Collapse
Affiliation(s)
- Raúl López-Izquierdo
- Faculty of Medicine. Universidad de Valladolid, Valladolid, Spain
- Emergency Department. Hospital Universitario Rio Hortega, Valladolid, Spain
- CIBER of Respiratory Diseases (CIBERES), Institute of Health Carlos III, Madrid, Spain
| | - Carlos Del Pozo Vegas
- Faculty of Medicine. Universidad de Valladolid, Valladolid, Spain
- Emergency Department. Hospital Clínico Universitario, Valladolid, Spain
| | - Ancor Sanz-García
- Faculty of Health Sciences, University of Castilla la Mancha, Talavera de la Reina, Spain.
- Technological Innovation Applied to Health Research Group (ITAS Group), Faculty of Health Sciences, University of de Castilla-La Mancha, Talavera de la Reina, Spain.
- Evaluación de Cuidados de Salud (ECUSAL), Instituto de Investigación Sanitaria de Castilla-La Mancha (IDISCAM), Talavera de la Reina, Spain.
| | - Agustín Mayo Íscar
- Department of Statistics and Operative Research. Faculty of Medicine, University of Valladolid, Valladolid, Spain
| | | | - Eduardo Silva Alvarado
- Universidad Europea del Atlántico, Santander, Spain
- Universidad Internacional Iberoamericana, Campeche, México
- Universidad de La Romana, La Romana, República Dominicana
| | - Santos Gracia Villar
- Universidad Europea del Atlántico, Santander, Spain
- Universidad Internacional Iberoamericana, Campeche, México
- Universidad Internacional Iberoamericana Arecibo, Puerto Rico, USA
| | - Luis Alonso Dzul López
- Universidad Europea del Atlántico, Santander, Spain
- Universidad Internacional Iberoamericana, Campeche, México
- Universidad Internacional Iberoamericana Arecibo, Puerto Rico, USA
| | - Silvia Aparicio Obregón
- Universidad Europea del Atlántico, Santander, Spain
- Universidad de La Romana, La Romana, República Dominicana
- Fundación Universitaria Internacional de Colombia, Bogotá, Colombia
| | - Rubén Calderon Iglesias
- Universidad Europea del Atlántico, Santander, Spain
- Universidad de La Romana, La Romana, República Dominicana
- Universidade Internacional do Cuanza. Cuito, Bié, Angola
| | - Joan B Soriano
- CIBER of Respiratory Diseases (CIBERES), Institute of Health Carlos III, Madrid, Spain
- Facultad de Medicina, Universidad Autónoma de Madrid, Madrid, Spain
- Servicio de Neumología; Hospital Universitario de La Princesa, Madrid, Spain
| | - Francisco Martín-Rodríguez
- Faculty of Medicine. Universidad de Valladolid, Valladolid, Spain
- Advanced Life Support, Emergency Medical Services (SACYL), Valladolid, Spain
| |
Collapse
|
21
|
Wynberg E, Han AX, van Willigen HDG, Verveen A, van Pul L, Maurer I, van Leeuwen EM, van den Aardweg JG, de Jong MD, Nieuwkerk P, Prins M, Kootstra NA, de Bree GJ. Inflammatory profiles are associated with long COVID up to 6 months after COVID-19 onset: A prospective cohort study of individuals with mild to critical COVID-19. PLoS One 2024; 19:e0304990. [PMID: 39008486 PMCID: PMC11249251 DOI: 10.1371/journal.pone.0304990] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2023] [Accepted: 05/17/2024] [Indexed: 07/17/2024] Open
Abstract
BACKGROUND After initial COVID-19, immune dysregulation may persist and drive post-acute sequelae of COVID-19 (PASC). We described longitudinal trajectories of cytokines in adults up to 6 months following SARS-CoV-2 infection and explored early predictors of PASC. METHODS RECoVERED is a prospective cohort of individuals with laboratory-confirmed SARS-CoV-2 infection between May 2020 and June 2021 in Amsterdam, the Netherlands. Serum was collected at weeks 4, 12 and 24 of follow-up. Monthly symptom questionnaires were completed from month 2 after COVID-19 onset onwards; lung diffusion capacity (DLCO) was tested at 6 months. Cytokine concentrations were analysed by human magnetic Luminex screening assay. We used a linear mixed-effects model to study log-concentrations of cytokines over time, assessing their association with socio-demographic and clinical characteristics that were included in the model as fixed effects. RESULTS 186/349 (53%) participants had ≥2 serum samples and were included in current analyses. Of these, 101/186 (54%: 45/101[45%] female, median age 55 years [IQR = 45-64]) reported PASC at 12 and 24 weeks after COVID-19 onset. We included 37 reference samples (17/37[46%] female, median age 49 years [IQR = 40-56]). In a multivariate model, PASC was associated with raised CRP and abnormal diffusion capacity with raised IL10, IL17, IL6, IP10 and TNFα at 24 weeks. Early (0-4 week) IL-1β and BMI at COVID-19 onset were predictive of PASC at 24 weeks. CONCLUSIONS Our findings indicate that immune dysregulation plays an important role in PASC pathogenesis, especially among individuals with reduced pulmonary function. Early IL-1β shows promise as a predictor of PASC.
Collapse
Affiliation(s)
- Elke Wynberg
- Department of Medical Microbiology & Infection Prevention, Amsterdam UMC, University of Amsterdam, Amsterdam Institute for Infection and Immunity, Amsterdam, the Netherlands
- Department of Infectious Diseases, Public Health Service of Amsterdam, Amsterdam, the Netherlands
| | - Alvin X Han
- Department of Medical Microbiology & Infection Prevention, Amsterdam UMC, University of Amsterdam, Amsterdam Institute for Infection and Immunity, Amsterdam, the Netherlands
| | - Hugo D G van Willigen
- Department of Medical Microbiology & Infection Prevention, Amsterdam UMC, University of Amsterdam, Amsterdam Institute for Infection and Immunity, Amsterdam, the Netherlands
| | - Anouk Verveen
- Department of Medical Psychology, Amsterdam UMC, Amsterdam Public Health Research Institute, University of Amsterdam, Amsterdam, the Netherlands
| | - Lisa van Pul
- Department of Experimental Immunology, Amsterdam UMC, University of Amsterdam, Amsterdam Institute for Infection and Immunity, Amsterdam, the Netherlands
| | - Irma Maurer
- Department of Experimental Immunology, Amsterdam UMC, University of Amsterdam, Amsterdam Institute for Infection and Immunity, Amsterdam, the Netherlands
| | - Ester M van Leeuwen
- Department of Experimental Immunology, Amsterdam UMC, University of Amsterdam, Amsterdam Institute for Infection and Immunity, Amsterdam, the Netherlands
| | - Joost G van den Aardweg
- Department of Pulmonology, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands
| | - Menno D de Jong
- Department of Medical Microbiology & Infection Prevention, Amsterdam UMC, University of Amsterdam, Amsterdam Institute for Infection and Immunity, Amsterdam, the Netherlands
| | - Pythia Nieuwkerk
- Department of Medical Psychology, Amsterdam UMC, Amsterdam Public Health Research Institute, University of Amsterdam, Amsterdam, the Netherlands
| | - Maria Prins
- Department of Infectious Diseases, Public Health Service of Amsterdam, Amsterdam, the Netherlands
- Department of Infectious Diseases, Internal Medicine, Amsterdam UMC, University of Amsterdam, Amsterdam Institute for Infection and Immunity, Amsterdam, the Netherlands
| | - Neeltje A Kootstra
- Department of Medical Psychology, Amsterdam UMC, Amsterdam Public Health Research Institute, University of Amsterdam, Amsterdam, the Netherlands
| | - Godelieve J de Bree
- Department of Infectious Diseases, Internal Medicine, Amsterdam UMC, University of Amsterdam, Amsterdam Institute for Infection and Immunity, Amsterdam, the Netherlands
| |
Collapse
|
22
|
Karasu M, Cevik M, Biberoglu S, Kaplanoglu ES, Cetinkaya N, Konukoglu D, Kucur M. The relationship between Nuclear Factor-Kappa B and Inhibitor-Kappa B parameters with clinical course in COVID-19 patients. Mol Biol Rep 2024; 51:813. [PMID: 39008220 DOI: 10.1007/s11033-024-09729-6] [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: 04/13/2024] [Accepted: 06/14/2024] [Indexed: 07/16/2024]
Abstract
BACKGROUND We aimed to investigate the serum Nuclear Factor Kappa B (NF-κB) p105, NF-κB p65 and Inhibitor Kappa B Alpha (IκBα) levels in patients with mild/moderate Coronavirus Disease 2019 (COVID-19) and their association with the course of the disease. MATERIALS AND METHODS Blood was drawn from 35 COVID-19 patients who applied to the Department of Emergency Medicine of Istanbul University-Cerrahpasa at the time of diagnosis and from 35 healthy individuals. The patients were evaluated to have mild/moderate degree of disease according to National Early Warning Score 2 (NEWS2) scoring and computed tomography (CT) findings. The markers were studied in the obtained serum samples, using enzyme-linked immunoassay (ELISA). Receiver Operating Characteristic (ROC) analysis was performed. Statistical significance was evaluated to be p < 0.05. RESULTS NF-κB p105 levels were significantly higher in the COVID-19 group compared to the control group. C-reactive protein (CRP), D-dimer, ferritin levels of the patients were significantly higher (p < 0.001) compared to the control group, while the lymphocyte count was found lower (p = 0.001). IκBα and NF-κB p65 levels are similar in both groups. Threshold value for NF-κB p105 was above 0.78 ng/mL, sensitivity was 71.4% and specificity was 97.1% (p < 0.05). NF-κB p105 levels at the time of diagnosis of the patients who required supplemental oxygen (O2), were significantly higher (p < 0.01). CONCLUSIONS The rise in serum NF-κB p105 levels during the early stages of infection holds diagnostic value. Besides its relation with severity might have a prognostic feature to foresee the requirement for supplemental O2 that occurs during hospitalization.
Collapse
Affiliation(s)
- Melek Karasu
- Cerrahpasa Faculty of Medicine, Department of Medical Biochemistry, Istanbul University-Cerrahpasa, Istanbul, Turkey.
| | - Muhdi Cevik
- Cerrahpasa Faculty of Medicine, Department of Emergency Medicine, Istanbul University-Cerrahpasa, Istanbul, Turkey
| | - Serap Biberoglu
- Cerrahpasa Faculty of Medicine, Department of Emergency Medicine, Istanbul University-Cerrahpasa, Istanbul, Turkey
| | - Emine Selva Kaplanoglu
- Cerrahpasa Faculty of Medicine Hospital, Fikret Biyal Biochemistry Laboratory, Istanbul University-Cerrahpasa, Istanbul, Turkey
| | - Nilgun Cetinkaya
- Cerrahpasa Faculty of Medicine Hospital, Fikret Biyal Biochemistry Laboratory, Istanbul University-Cerrahpasa, Istanbul, Turkey
| | - Dildar Konukoglu
- Cerrahpasa Faculty of Medicine, Department of Medical Biochemistry, Istanbul University-Cerrahpasa, Istanbul, Turkey
- Cerrahpasa Faculty of Medicine Hospital, Fikret Biyal Biochemistry Laboratory, Istanbul University-Cerrahpasa, Istanbul, Turkey
| | - Mine Kucur
- Cerrahpasa Faculty of Medicine, Department of Medical Biochemistry, Istanbul University-Cerrahpasa, Istanbul, Turkey
- Cerrahpasa Faculty of Medicine Hospital, Fikret Biyal Biochemistry Laboratory, Istanbul University-Cerrahpasa, Istanbul, Turkey
| |
Collapse
|
23
|
Llitjos JF, Carrol ED, Osuchowski MF, Bonneville M, Scicluna BP, Payen D, Randolph AG, Witte S, Rodriguez-Manzano J, François B. Enhancing sepsis biomarker development: key considerations from public and private perspectives. Crit Care 2024; 28:238. [PMID: 39003476 PMCID: PMC11246589 DOI: 10.1186/s13054-024-05032-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2024] [Accepted: 07/10/2024] [Indexed: 07/15/2024] Open
Abstract
Implementation of biomarkers in sepsis and septic shock in emergency situations, remains highly challenging. This viewpoint arose from a public-private 3-day workshop aiming to facilitate the transition of sepsis biomarkers into clinical practice. The authors consist of international academic researchers and clinician-scientists and industry experts who gathered (i) to identify current obstacles impeding biomarker research in sepsis, (ii) to outline the important milestones of the critical path of biomarker development and (iii) to discuss novel avenues in biomarker discovery and implementation. To define more appropriately the potential place of biomarkers in sepsis, a better understanding of sepsis pathophysiology is mandatory, in particular the sepsis patient's trajectory from the early inflammatory onset to the late persisting immunosuppression phase. This time-varying host response urges to develop time-resolved test to characterize persistence of immunological dysfunctions. Furthermore, age-related difference has to be considered between adult and paediatric septic patients. In this context, numerous barriers to biomarker adoption in practice, such as lack of consensus about diagnostic performances, the absence of strict recommendations for sepsis biomarker development, cost and resources implications, methodological validation challenges or limited awareness and education have been identified. Biomarker-guided interventions for sepsis to identify patients that would benefit more from therapy, such as sTREM-1-guided Nangibotide treatment or Adrenomedullin-guided Enibarcimab treatment, appear promising but require further evaluation. Artificial intelligence also has great potential in the sepsis biomarker discovery field through capability to analyse high volume complex data and identify complex multiparametric patient endotypes or trajectories. To conclude, biomarker development in sepsis requires (i) a comprehensive and multidisciplinary approach employing the most advanced analytical tools, (ii) the creation of a platform that collaboratively merges scientific and commercial needs and (iii) the support of an expedited regulatory approval process.
Collapse
Affiliation(s)
- Jean-Francois Llitjos
- Open Innovation and Partnerships (OI&P), bioMérieux S.A., Marcy l'Etoile, France.
- Anesthesiology and Critical Care Medicine, Hospices Civils de Lyon, Edouard Herriot Hospital, Lyon, France.
| | - Enitan D Carrol
- Department of Clinical Infection, Microbiology and Immunology, University of Liverpool Institute of Infection Veterinary and Ecological Sciences, Liverpool, UK
- Department of Paediatric Infectious Diseases and Immunology, Alder Hey Children's NHS Foundation Trust, Liverpool, UK
| | - Marcin F Osuchowski
- Ludwig Boltzmann Institute for Traumatology, The Research Center in Cooperation with AUVA, Vienna, Austria
| | - Marc Bonneville
- Medical and Scientific Affairs, Institut Mérieux, Lyon, France
| | - Brendon P Scicluna
- Department of Applied Biomedical Science, Faculty of Health Sciences, Mater Dei Hospital, University of Malta, Msida, Malta
- Centre for Molecular Medicine and Biobanking, University of Malta, Msida, Malta
| | - Didier Payen
- Paris 7 University Denis Diderot, Paris Sorbonne, Cité, France
| | - Adrienne G Randolph
- Departments of Anaesthesia and Pediatrics, Harvard Medical School, Boston, MA, USA
- Department of Anesthesiology, Critical Care and Pain Medicine, Boston Children's Hospital, Boston, MA, USA
| | | | | | - Bruno François
- Medical-Surgical Intensive Care Unit, Réanimation Polyvalente, Dupuytren University Hospital, CHU de Limoges, 2 Avenue Martin Luther King, 87042, Limoges Cedex, France.
- Inserm CIC 1435, Dupuytren University Hospital, Limoges, France.
- Inserm UMR 1092, Medicine Faculty, University of Limoges, Limoges, France.
| |
Collapse
|
24
|
Corrao S, Raspanti M, Agugliaro F, Gervasi F, Di Bernardo F, Natoli G, Argano C. Safety of High-Dose Vitamin C in Non-Intensive Care Hospitalized Patients with COVID-19: An Open-Label Clinical Study. J Clin Med 2024; 13:3987. [PMID: 38999551 PMCID: PMC11242388 DOI: 10.3390/jcm13133987] [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: 05/11/2024] [Revised: 06/30/2024] [Accepted: 07/02/2024] [Indexed: 07/14/2024] Open
Abstract
Background: Vitamin C has been used as an antioxidant and has been proven effective in boosting immunity in different diseases, including coronavirus disease (COVID-19). An increasing awareness was directed to the role of intravenous vitamin C in COVID-19. Methods: In this study, we aimed to assess the safety of high-dose intravenous vitamin C added to the conventional regimens for patients with different stages of COVID-19. An open-label clinical trial was conducted on patients with COVID-19. One hundred four patients underwent high-dose intravenous administration of vitamin C (in addition to conventional therapy), precisely 10 g in 250 cc of saline solution in slow infusion (60 drops/min) for three consecutive days. At the same time, 42 patients took the standard-of-care therapy. Results: This study showed the safety of high-dose intravenous administration of vitamin C. No adverse reactions were found. When we evaluated the renal function indices and estimated the glomerular filtration rate (eGRF, calculated with the CKD-EPI Creatinine Equation) as the main side effect and contraindication related to chronic renal failure, no statistically significant differences between the two groups were found. High-dose vitamin C treatment was not associated with a statistically significant reduction in mortality and admission to the intensive care unit, even if the result was bound to the statistical significance. On the contrary, age was independently associated with admission to the intensive care unit and in-hospital mortality as well as noninvasive ventilation (N.I.V.) and continuous positive airway pressure (CPAP) (OR 2.17, 95% CI 1.41-3.35; OR 7.50, 95% CI 1.97-28.54; OR 8.84, 95% CI 2.62-29.88, respectively). When considering the length of hospital stay, treatment with high-dose vitamin C predicts shorter hospitalization (OR -4.95 CI -0.21--9.69). Conclusions: Our findings showed that an intravenous high dose of vitamin C is configured as a safe and promising therapy for patients with moderate to severe COVID-19.
Collapse
Affiliation(s)
- Salvatore Corrao
- Department of Internal Medicine, National Relevance and High Specialization Hospital Trust ARNAS Civico, Di Cristina, Benfratelli, 90127 Palermo, Italy; (F.A.); (G.N.); (C.A.)
- Department of Health Promotion Sciences, Maternal and Infant Care, Internal Medicine and Medical Specialties (PROMISE), University of Palermo, 90127 Palermo, Italy
| | - Massimo Raspanti
- Cardiology and Intensive Care Unit, A. Aiello Hospital, 91026 Mazzara del Vallo, Italy;
| | - Federica Agugliaro
- Department of Internal Medicine, National Relevance and High Specialization Hospital Trust ARNAS Civico, Di Cristina, Benfratelli, 90127 Palermo, Italy; (F.A.); (G.N.); (C.A.)
| | - Francesco Gervasi
- Specialized Laboratory of Oncology, National Relevance and High Specialization Hospital Trust ARNAS Civico, Di Cristina, Benfratelli, 90127 Palermo, Italy;
| | - Francesca Di Bernardo
- Department of Microbiology and Virology, National Relevance and High Specialization Hospital Trust ARNAS Civico, Di Cristina, Benfratelli, 90127 Palermo, Italy;
| | - Giuseppe Natoli
- Department of Internal Medicine, National Relevance and High Specialization Hospital Trust ARNAS Civico, Di Cristina, Benfratelli, 90127 Palermo, Italy; (F.A.); (G.N.); (C.A.)
| | - Christiano Argano
- Department of Internal Medicine, National Relevance and High Specialization Hospital Trust ARNAS Civico, Di Cristina, Benfratelli, 90127 Palermo, Italy; (F.A.); (G.N.); (C.A.)
| |
Collapse
|
25
|
Lin M, Xu F, Sun J, Song J, Shen Y, Lu S, Ding H, Lan L, Chen C, Ma W, Wu X, Song Z, Wang W. Integrative multi-omics analysis unravels the host response landscape and reveals a serum protein panel for early prognosis prediction for ARDS. Crit Care 2024; 28:213. [PMID: 38956604 PMCID: PMC11218270 DOI: 10.1186/s13054-024-05000-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2024] [Accepted: 06/24/2024] [Indexed: 07/04/2024] Open
Abstract
BACKGROUND The multidimensional biological mechanisms underpinning acute respiratory distress syndrome (ARDS) continue to be elucidated, and early biomarkers for predicting ARDS prognosis are yet to be identified. METHODS We conducted a multicenter observational study, profiling the 4D-DIA proteomics and global metabolomics of serum samples collected from patients at the initial stage of ARDS, alongside samples from both disease control and healthy control groups. We identified 28-day prognosis biomarkers of ARDS in the discovery cohort using the LASSO method, fold change analysis, and the Boruta algorithm. The candidate biomarkers were validated through parallel reaction monitoring (PRM) targeted mass spectrometry in an external validation cohort. Machine learning models were applied to explore the biomarkers of ARDS prognosis. RESULTS In the discovery cohort, comprising 130 adult ARDS patients (mean age 72.5, 74.6% male), 33 disease controls, and 33 healthy controls, distinct proteomic and metabolic signatures were identified to differentiate ARDS from both control groups. Pathway analysis highlighted the upregulated sphingolipid signaling pathway as a key contributor to the pathological mechanisms underlying ARDS. MAP2K1 emerged as the hub protein, facilitating interactions with various biological functions within this pathway. Additionally, the metabolite sphingosine 1-phosphate (S1P) was closely associated with ARDS and its prognosis. Our research further highlights essential pathways contributing to the deceased ARDS, such as the downregulation of hematopoietic cell lineage and calcium signaling pathways, contrasted with the upregulation of the unfolded protein response and glycolysis. In particular, GAPDH and ENO1, critical enzymes in glycolysis, showed the highest interaction degree in the protein-protein interaction network of ARDS. In the discovery cohort, a panel of 36 proteins was identified as candidate biomarkers, with 8 proteins (VCAM1, LDHB, MSN, FLG2, TAGLN2, LMNA, MBL2, and LBP) demonstrating significant consistency in an independent validation cohort of 183 patients (mean age 72.6 years, 73.2% male), confirmed by PRM assay. The protein-based model exhibited superior predictive accuracy compared to the clinical model in both the discovery cohort (AUC: 0.893 vs. 0.784; Delong test, P < 0.001) and the validation cohort (AUC: 0.802 vs. 0.738; Delong test, P = 0.008). INTERPRETATION Our multi-omics study demonstrated the potential biological mechanism and therapy targets in ARDS. This study unveiled several novel predictive biomarkers and established a validated prediction model for the poor prognosis of ARDS, offering valuable insights into the prognosis of individuals with ARDS.
Collapse
Affiliation(s)
- Mengna Lin
- Shanghai Institute of Infectious Disease and Biosecurity, School of Public Health, Fudan University, Shanghai, China
- Department of Emergency Medicine, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Feixiang Xu
- Department of Emergency Medicine, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Jian Sun
- Department of Emergency Medicine, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Jianfeng Song
- Department of Emergency Medicine, Minhang Hospital, Fudan University, Shanghai, China
| | - Yao Shen
- Department of Respiratory Medicine, Pudong Hospital, Fudan University, Shanghai, China
| | - Su Lu
- Department of Emergency Medicine, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Hailin Ding
- Department of Emergency Medicine, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Lulu Lan
- Department of Emergency Medicine, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Chen Chen
- Department of Emergency Medicine, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Wen Ma
- School of Public Health, Fudan University, Shanghai, China
| | - Xueling Wu
- Department of Respiratory Medicine, Renji Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China.
| | - Zhenju Song
- Shanghai Institute of Infectious Disease and Biosecurity, School of Public Health, Fudan University, Shanghai, China.
- Department of Emergency Medicine, Zhongshan Hospital, Fudan University, Shanghai, China.
- Institute of Emergency Rescue and Critical Care, Fudan University, Shanghai, China.
| | - Weibing Wang
- Shanghai Institute of Infectious Disease and Biosecurity, School of Public Health, Fudan University, Shanghai, China.
- School of Public Health, Fudan University, Shanghai, China.
| |
Collapse
|
26
|
Cavaillon JM, Chousterman BG, Skirecki T. Compartmentalization of the inflammatory response during bacterial sepsis and severe COVID-19. JOURNAL OF INTENSIVE MEDICINE 2024; 4:326-340. [PMID: 39035623 PMCID: PMC11258514 DOI: 10.1016/j.jointm.2024.01.001] [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: 08/27/2023] [Revised: 01/04/2024] [Accepted: 01/06/2024] [Indexed: 07/23/2024]
Abstract
Acute infections cause local and systemic disorders which can lead in the most severe forms to multi-organ failure and eventually to death. The host response to infection encompasses a large spectrum of reactions with a concomitant activation of the so-called inflammatory response aimed at fighting the infectious agent and removing damaged tissues or cells, and the anti-inflammatory response aimed at controlling inflammation and initiating the healing process. Fine-tuning at the local and systemic levels is key to preventing local and remote injury due to immune system activation. Thus, during bacterial sepsis and Coronavirus disease 2019 (COVID-19), concomitant systemic and compartmentalized pro-inflammatory and compensatory anti-inflammatory responses are occurring. Immune cells (e.g., macrophages, neutrophils, natural killer cells, and T-lymphocytes), as well as endothelial cells, differ from one compartment to another and contribute to specific organ responses to sterile and microbial insult. Furthermore, tissue-specific microbiota influences the local and systemic response. A better understanding of the tissue-specific immune status, the organ immunity crosstalk, and the role of specific mediators during sepsis and COVID-19 can foster the development of more accurate biomarkers for better diagnosis and prognosis and help to define appropriate host-targeted treatments and vaccines in the context of precision medicine.
Collapse
Affiliation(s)
| | - Benjamin G. Chousterman
- Department of Anesthesia and Critical Care, Lariboisière University Hospital, DMU Parabol, APHP Nord, Paris, France
- Inserm U942, University of Paris, Paris, France
| | - Tomasz Skirecki
- Department of Translational Immunology and Experimental Intensive Care, Centre of Postgraduate Medical Education, Warsaw, Poland
| |
Collapse
|
27
|
Alfaro E, Díaz-García E, García-Tovar S, Galera R, Casitas R, Torres-Vargas M, López-Fernández C, Añón JM, García-Río F, Cubillos-Zapata C. Endothelial dysfunction and persistent inflammation in severe post-COVID-19 patients: implications for gas exchange. BMC Med 2024; 22:242. [PMID: 38867241 PMCID: PMC11170912 DOI: 10.1186/s12916-024-03461-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/25/2024] [Accepted: 05/30/2024] [Indexed: 06/14/2024] Open
Abstract
BACKGROUND Understanding the enduring respiratory consequences of severe COVID-19 is crucial for comprehensive patient care. This study aims to evaluate the impact of post-COVID conditions on respiratory sequelae of severe acute respiratory distress syndrome (ARDS). METHODS We examined 88 survivors of COVID-19-associated severe ARDS six months post-intensive care unit (ICU) discharge. Assessments included clinical and functional evaluation as well as plasma biomarkers of endothelial dysfunction, inflammation, and viral response. Additionally, an in vitro model using human umbilical vein endothelial cells (HUVECs) explored the direct impact of post-COVID plasma on endothelial function. RESULTS Post-COVID patients with impaired gas exchange demonstrated persistent endothelial inflammation marked by elevated ICAM-1, IL-8, CCL-2, and ET-1 plasma levels. Concurrently, systemic inflammation, evidenced by NLRP3 overexpression and elevated levels of IL-6, sCD40-L, and C-reactive protein, was associated with endothelial dysfunction biomarkers and increased in post-COVID patients with impaired gas exchange. T-cell activation, reflected in CD69 expression, and persistently elevated levels of interferon-β (IFN-β) further contributed to sustained inflammation. The in vitro model confirmed that patient plasma, with altered levels of sCD40-L and IFN-β proteins, has the capacity to alter endothelial function. CONCLUSIONS Six months post-ICU discharge, survivors of COVID-19-associated ARDS exhibited sustained elevation in endothelial dysfunction biomarkers, correlating with the severity of impaired gas exchange. NLRP3 inflammasome activity and persistent T-cell activation indicate on going inflammation contributing to persistent endothelial dysfunction, potentially intensified by sustained viral immune response.
Collapse
Affiliation(s)
- Enrique Alfaro
- Respiratory Diseases Group, Respiratory Service, La Paz University Hospital, IdiPAZ, Paseo de La Castellana 261, 28046, Madrid, Spain
- Biomedical Research Networking Centre On Respiratory Diseases (CIBERES), Madrid, Spain
| | - Elena Díaz-García
- Respiratory Diseases Group, Respiratory Service, La Paz University Hospital, IdiPAZ, Paseo de La Castellana 261, 28046, Madrid, Spain
- Biomedical Research Networking Centre On Respiratory Diseases (CIBERES), Madrid, Spain
| | - Sara García-Tovar
- Respiratory Diseases Group, Respiratory Service, La Paz University Hospital, IdiPAZ, Paseo de La Castellana 261, 28046, Madrid, Spain
| | - Raúl Galera
- Respiratory Diseases Group, Respiratory Service, La Paz University Hospital, IdiPAZ, Paseo de La Castellana 261, 28046, Madrid, Spain
- Biomedical Research Networking Centre On Respiratory Diseases (CIBERES), Madrid, Spain
| | - Raquel Casitas
- Respiratory Diseases Group, Respiratory Service, La Paz University Hospital, IdiPAZ, Paseo de La Castellana 261, 28046, Madrid, Spain
- Biomedical Research Networking Centre On Respiratory Diseases (CIBERES), Madrid, Spain
| | - María Torres-Vargas
- Respiratory Diseases Group, Respiratory Service, La Paz University Hospital, IdiPAZ, Paseo de La Castellana 261, 28046, Madrid, Spain
- Biomedical Research Networking Centre On Respiratory Diseases (CIBERES), Madrid, Spain
| | - Cristina López-Fernández
- Respiratory Diseases Group, Respiratory Service, La Paz University Hospital, IdiPAZ, Paseo de La Castellana 261, 28046, Madrid, Spain
- Biomedical Research Networking Centre On Respiratory Diseases (CIBERES), Madrid, Spain
| | - José M Añón
- Department of Intensive Medicine, La Paz University Hospital, Madrid, Spain
| | - Francisco García-Río
- Respiratory Diseases Group, Respiratory Service, La Paz University Hospital, IdiPAZ, Paseo de La Castellana 261, 28046, Madrid, Spain.
- Biomedical Research Networking Centre On Respiratory Diseases (CIBERES), Madrid, Spain.
- Faculty of Medicine, Autonomous University of Madrid, Madrid, Spain.
| | - Carolina Cubillos-Zapata
- Respiratory Diseases Group, Respiratory Service, La Paz University Hospital, IdiPAZ, Paseo de La Castellana 261, 28046, Madrid, Spain.
- Biomedical Research Networking Centre On Respiratory Diseases (CIBERES), Madrid, Spain.
| |
Collapse
|
28
|
Carvajal JJ, García-Castillo V, Cuellar SV, Campillay-Véliz CP, Salazar-Ardiles C, Avellaneda AM, Muñoz CA, Retamal-Díaz A, Bueno SM, González PA, Kalergis AM, Lay MK. New insights into the pathogenesis of SARS-CoV-2 during and after the COVID-19 pandemic. Front Immunol 2024; 15:1363572. [PMID: 38911850 PMCID: PMC11190347 DOI: 10.3389/fimmu.2024.1363572] [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: 12/31/2023] [Accepted: 04/24/2024] [Indexed: 06/25/2024] Open
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is responsible for the respiratory distress condition known as COVID-19. This disease broadly affects several physiological systems, including the gastrointestinal, renal, and central nervous (CNS) systems, significantly influencing the patient's overall quality of life. Additionally, numerous risk factors have been suggested, including gender, body weight, age, metabolic status, renal health, preexisting cardiomyopathies, and inflammatory conditions. Despite advances in understanding the genome and pathophysiological ramifications of COVID-19, its precise origins remain elusive. SARS-CoV-2 interacts with a receptor-binding domain within angiotensin-converting enzyme 2 (ACE2). This receptor is expressed in various organs of different species, including humans, with different abundance. Although COVID-19 has multiorgan manifestations, the main pathologies occur in the lung, including pulmonary fibrosis, respiratory failure, pulmonary embolism, and secondary bacterial pneumonia. In the post-COVID-19 period, different sequelae may occur, which may have various causes, including the direct action of the virus, alteration of the immune response, and metabolic alterations during infection, among others. Recognizing the serious adverse health effects associated with COVID-19, it becomes imperative to comprehensively elucidate and discuss the existing evidence surrounding this viral infection, including those related to the pathophysiological effects of the disease and the subsequent consequences. This review aims to contribute to a comprehensive understanding of the impact of COVID-19 and its long-term effects on human health.
Collapse
Affiliation(s)
- Jonatan J. Carvajal
- Department of Biotechnology, Faculty of Marine Sciences and Biological Resources, University of Antofagasta, Antofagasta, Chile
| | - Valeria García-Castillo
- Department of Biotechnology, Faculty of Marine Sciences and Biological Resources, University of Antofagasta, Antofagasta, Chile
| | - Shelsy V. Cuellar
- Department of Biotechnology, Faculty of Marine Sciences and Biological Resources, University of Antofagasta, Antofagasta, Chile
| | | | - Camila Salazar-Ardiles
- Center for Research in Physiology and Altitude Medicine (FIMEDALT), Biomedical Department, Faculty of Health Sciences, University of Antofagasta, Antofagasta, Chile
| | - Andrea M. Avellaneda
- Department of Biotechnology, Faculty of Marine Sciences and Biological Resources, University of Antofagasta, Antofagasta, Chile
- Department of Basic Sciences, Faculty of Sciences, Universidad Santo Tomás, Antofagasta, Chile
| | - Christian A. Muñoz
- Research Center in Immunology and Biomedical Biotechnology of Antofagasta (CIIBBA), University of Antofagasta, Antofagasta, Chile
- Department of Medical Technology, Faculty of Health Sciences, University of Antofagasta, Antofagasta, Chile
- Millennium Institute on Immunology and Immunotherapy, Department of Biotechnology, Faculty of Marine Sciences and Biological Resources, Department of Medical Technology, Faculty of Health Sciences, University of Antofagasta, Antofagasta, Chile
| | - Angello Retamal-Díaz
- Department of Biotechnology, Faculty of Marine Sciences and Biological Resources, University of Antofagasta, Antofagasta, Chile
- Research Center in Immunology and Biomedical Biotechnology of Antofagasta (CIIBBA), University of Antofagasta, Antofagasta, Chile
- Millennium Institute on Immunology and Immunotherapy, Department of Biotechnology, Faculty of Marine Sciences and Biological Resources, Department of Medical Technology, Faculty of Health Sciences, University of Antofagasta, Antofagasta, Chile
| | - Susan M. Bueno
- Millennium Institute on Immunology and Immunotherapy, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Pablo A. González
- Millennium Institute on Immunology and Immunotherapy, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Alexis M. Kalergis
- Millennium Institute on Immunology and Immunotherapy, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
- Departamento de Endocrinología, Facultad de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Margarita K. Lay
- Department of Biotechnology, Faculty of Marine Sciences and Biological Resources, University of Antofagasta, Antofagasta, Chile
- Research Center in Immunology and Biomedical Biotechnology of Antofagasta (CIIBBA), University of Antofagasta, Antofagasta, Chile
- Millennium Institute on Immunology and Immunotherapy, Department of Biotechnology, Faculty of Marine Sciences and Biological Resources, Department of Medical Technology, Faculty of Health Sciences, University of Antofagasta, Antofagasta, Chile
| |
Collapse
|
29
|
Suryawanshi P, Patil‐Takbhate B, Athavale P, Mirza S, Tripathy A, Kanitkar S, Shivnitwar S, Barthwal MS, Dole S, Chavan H, Jali P, Pawale S, Kakad D, Kakrani AL, Bhawalkar J, Gandhi M, Chaturvedi S, Karandikar M, Tripathy S. T-cell responses in COVID-19 survivors 6-8 months after infection: A longitudinal cohort study in Pune. Immun Inflamm Dis 2024; 12:e1238. [PMID: 38860770 PMCID: PMC11165687 DOI: 10.1002/iid3.1238] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2023] [Revised: 02/26/2024] [Accepted: 03/20/2024] [Indexed: 06/12/2024] Open
Abstract
BACKGROUND The severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) immune response is crucial for disease management, although diminishing immunity raises the possibility of reinfection. METHODS We examined the immunological response to SARS-CoV-2 in a cohort of convalescent COVID-19 patients in matched samples collected at 1 and 6-8 months after infection. The peripheral blood mononuclear cells were isolated from enrolled study participants and flow cytometry analysis was done to assess the lymphocyte subsets of naive, effector, central memory, and effector memory CD4+ or CD8+ T cells in COVID-19 patients at 1 and 6-8 months after infection. Immunophenotypic characterization of immune cell subsets was performed on individuals who were followed longitudinally for 1 month (n = 44) and 6-8 months (n = 25) after recovery from COVID infection. RESULTS We observed that CD4 +T cells in hospitalized SARS-CoV-2 patients tended to decrease, whereas CD8+ T cells steadily recovered after 1 month, while there was a sustained increase in the population of effector T cells and effector memory T cells. Furthermore, COVID-19 patients showed persistently low B cells and a small increase in the NK cell population. CONCLUSION Our findings show that T cell responses were maintained at 6-8 months after infection. This opens new pathways for further research into the long-term effects in COVID-19 immunopathogenesis.
Collapse
Affiliation(s)
- Poonam Suryawanshi
- Central Research Facility, Dr D. Y. Patil Medical College, Hospital and Research CentreDr D. Y. Patil Vidyapeeth, (deemed to be University)PimpriPuneIndia
| | - Bhagyashri Patil‐Takbhate
- Central Research Facility, Dr D. Y. Patil Medical College, Hospital and Research CentreDr D. Y. Patil Vidyapeeth, (deemed to be University)PimpriPuneIndia
| | - Prachi Athavale
- Department of Microbiology, Dr D. Y. Patil Medical College, Hospital and Research CentreDr D. Y. Patil Vidyapeeth, (deemed to be University)PimpriPuneIndia
| | - Shahzad Mirza
- Department of Microbiology, Dr D. Y. Patil Medical College, Hospital and Research CentreDr D. Y. Patil Vidyapeeth, (deemed to be University)PimpriPuneIndia
| | | | - Shubhangi Kanitkar
- Department of General Medicine, Dr D. Y. Patil Medical College, Hospital and Research CentreDr D. Y. Patil Vidyapeeth, (deemed to be University)PimpriPuneIndia
| | - Sachin Shivnitwar
- Department of General Medicine, Dr D. Y. Patil Medical College, Hospital and Research CentreDr D. Y. Patil Vidyapeeth, (deemed to be University)PimpriPuneIndia
| | - Madhusudan S. Barthwal
- Department of Respiratory Medicine, Dr D. Y. Patil Medical College, Hospital and Research CentreDr D. Y. Patil Vidyapeeth, Pimpri, (deemed to be University)PuneIndia
| | - Sachin Dole
- Department of Respiratory Medicine, Dr D. Y. Patil Medical College, Hospital and Research CentreDr D. Y. Patil Vidyapeeth, Pimpri, (deemed to be University)PuneIndia
| | - Hanumant Chavan
- Dr D. Y. Patil Medical College, Hospital and Research CentreDr D. Y. Patil Vidyapeeth, (deemed to be University)PimpriPuneIndia
| | - Priyanka Jali
- Central Research Facility, Dr D. Y. Patil Medical College, Hospital and Research CentreDr D. Y. Patil Vidyapeeth, (deemed to be University)PimpriPuneIndia
| | - Sujata Pawale
- Central Research Facility, Dr D. Y. Patil Medical College, Hospital and Research CentreDr D. Y. Patil Vidyapeeth, (deemed to be University)PimpriPuneIndia
| | - Dhanashree Kakad
- Central Research Facility, Dr D. Y. Patil Medical College, Hospital and Research CentreDr D. Y. Patil Vidyapeeth, (deemed to be University)PimpriPuneIndia
| | - Arjun Lal Kakrani
- Department of General Medicine, Dr D. Y. Patil Medical College, Hospital and Research CentreDr D. Y. Patil Vidyapeeth, (deemed to be University)PimpriPuneIndia
| | - Jitendra Bhawalkar
- Department of Community Medicine, Dr D. Y. Patil Medical College, Hospital and Research CentreDr D. Y. Patil Vidyapeeth, (deemed to be University)PimpriPuneIndia
| | - Madhura Gandhi
- Central Research Facility, Dr D. Y. Patil Medical College, Hospital and Research CentreDr D. Y. Patil Vidyapeeth, (deemed to be University)PimpriPuneIndia
| | | | - Mahesh Karandikar
- Central Research Facility, Dr D. Y. Patil Medical College, Hospital and Research CentreDr D. Y. Patil Vidyapeeth, (deemed to be University)PimpriPuneIndia
| | - Srikanth Tripathy
- Central Research Facility, Dr D. Y. Patil Medical College, Hospital and Research CentreDr D. Y. Patil Vidyapeeth, (deemed to be University)PimpriPuneIndia
| |
Collapse
|
30
|
Lafon T, Chapuis N, Guerin E, Daix T, Otranto M, Boumediene A, Jeannet R, Fontenay M, Henri Hani K, Vignon P, Monneret G, François B, Jean-Philippe J, Feuillard J. Along with PaO2/FiO2 ratio and lymphopenia, low HLA-DR monocytes are the only additional parameter that independently predicts the clinical course of undifferentiated SARS-CoV-2 patients in emergency departments. J Leukoc Biol 2024; 115:1131-1142. [PMID: 38366559 DOI: 10.1093/jleuko/qiae022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2023] [Revised: 12/28/2023] [Accepted: 01/01/2024] [Indexed: 02/18/2024] Open
Abstract
Because one-third of patients deteriorate after their admission to the emergency department, assessing the prognosis of COVID-19 patients is of great importance. However, to date, only lymphopenia and the partial pressure of oxygen/fraction of inspired oxygen (PaO2/FiO2) ratio have been reported as partly predictive of COVID-19-related further deterioration, and their association has not been evaluated. We asked whether other key biomarkers of SARS-CoV-2 immunologic defects-increase in circulating immature granulocytes, loss of monocyte HLA-DR (mHLA-DR) expression, and monocyte differentiation blockade-could also predict further COVID-19 deterioration. A series of 284 consecutive COVID-19 patients, with the sole inclusion criterion of being an adult, were prospectively enrolled at emergency department admission (day 0) of 2 different hospitals: 1 for the exploratory cohort (180 patients) and 1 for the confirmatory cohort (104 patients). Deterioration was assessed over the next 7 days. Neither increased immature granulocyte levels nor monocyte differentiation blockade predicted patient worsening. Among more than 30 clinical, biological, and radiological parameters, the value of decreased P/F ratio and lymphopenia for prediction of further COVID-19 deterioration was strongly confirmed, and the loss of mHLA-DR was the only additional independent marker. Combined together in a simple OxyLymphoMono score, the 3 variables perfectly predicted patients who did not worsen and correctly predicted worsening in 59% of cases. By highlighting lymphocyte and monocyte defects as preceding COVID-19 deterioration, these results point on early immunosuppression in COVID-19 deterioration. Combining P/F ratio, lymphopenia, and loss of mHLA-DR together in a simple and robust score could offer a pragmatic method for COVID-19 patient stratification.
Collapse
Affiliation(s)
- Thomas Lafon
- Emergency Department, University Hospital Center of Limoges, 2 avenue Martin Luther King, 87042 Limoges, France
- Institut National de la Santé et de la Recherche Médicale Centre d'Investigation Clinique 1435, Centre Hospitalier Universitaire Dupuytren, 2 avenue Martin Luther King, 87042 Limoges, France
| | - Nicolas Chapuis
- Institut National de la Santé et de la Recherche Médicale U1016, Centre National de la Recherche Scientifique Unité Mixte de Recherche 8104, Institut Cochin, Université Paris Cité, 27 Rue du Faubourg Saint-Jacques, 75014 Paris, France
- Service d'Hématologie Biologique, Hôpital Cochin, Assistance Publique-Hôpitaux de Paris, Centre-Université Paris Cité, 27 Rue du Faubourg Saint-Jacques, 75014 Paris, France
| | - Estelle Guerin
- Laboratoire d'Hématologie Biologique, Centre Hospitalier Universitaire de Limoges, Centre de Biologie et de Recherche en Santé, rue Bernard Descottes, 87042 Limoges, France
| | - Thomas Daix
- Institut National de la Santé et de la Recherche Médicale Centre d'Investigation Clinique 1435, Centre Hospitalier Universitaire Dupuytren, 2 avenue Martin Luther King, 87042 Limoges, France
- Réanimation Polyvalente, Centre Hospitalier Universitaire de Limoges, 2 avenue Martin Luther King, 87042 Limoges, France
- Institut National de la Santé et de la Recherche Médicale Unité Mixte de Recherche 1092, Centre Hospitalier Universitaire Dupuytren, 2 avenue Martin Luther King, 87042 Limoges, France
| | - Marcela Otranto
- Emergency Department, University Hospital Center of Limoges, 2 avenue Martin Luther King, 87042 Limoges, France
| | - Ahmed Boumediene
- Laboratoire Immunologie, Centre Hospitalier Universitaire de Limoges, Centre de Biologie et de Recherche en Santé, rue Bernard Descottes, 87042 Limoges, France
| | - Robin Jeannet
- Institut National de la Santé et de la Recherche Médicale Centre d'Investigation Clinique 1435, Centre Hospitalier Universitaire Dupuytren, 2 avenue Martin Luther King, 87042 Limoges, France
- Unité Mixte de Recherche Centre National de Recherche Scientifique 7276/Institut National de la Santé et de la Recherche Médicale U1262, Contrôle de la Réponse Immune B et Lymphoproliférations, Centre de Biologie et de Recherche en Santé, rue Bernard Descottes, 87025 Limoges, France
| | - Michaela Fontenay
- Institut National de la Santé et de la Recherche Médicale U1016, Centre National de la Recherche Scientifique Unité Mixte de Recherche 8104, Institut Cochin, Université Paris Cité, 27 Rue du Faubourg Saint-Jacques, 75014 Paris, France
- Service d'Hématologie Biologique, Hôpital Cochin, Assistance Publique-Hôpitaux de Paris, Centre-Université Paris Cité, 27 Rue du Faubourg Saint-Jacques, 75014 Paris, France
| | - Karam Henri Hani
- Emergency Department, University Hospital Center of Limoges, 2 avenue Martin Luther King, 87042 Limoges, France
| | - Philippe Vignon
- Institut National de la Santé et de la Recherche Médicale Centre d'Investigation Clinique 1435, Centre Hospitalier Universitaire Dupuytren, 2 avenue Martin Luther King, 87042 Limoges, France
- Réanimation Polyvalente, Centre Hospitalier Universitaire de Limoges, 2 avenue Martin Luther King, 87042 Limoges, France
- Institut National de la Santé et de la Recherche Médicale Unité Mixte de Recherche 1092, Centre Hospitalier Universitaire Dupuytren, 2 avenue Martin Luther King, 87042 Limoges, France
| | - Guillaume Monneret
- Laboratoire d'Immunologie, Hôpital E. Herriot, Hospices Civils de Lyon, 5 Pl. d'Arsonval, 69003 Lyon, France
| | - Bruno François
- Institut National de la Santé et de la Recherche Médicale Centre d'Investigation Clinique 1435, Centre Hospitalier Universitaire Dupuytren, 2 avenue Martin Luther King, 87042 Limoges, France
- Réanimation Polyvalente, Centre Hospitalier Universitaire de Limoges, 2 avenue Martin Luther King, 87042 Limoges, France
- Institut National de la Santé et de la Recherche Médicale Unité Mixte de Recherche 1092, Centre Hospitalier Universitaire Dupuytren, 2 avenue Martin Luther King, 87042 Limoges, France
| | - Jais Jean-Philippe
- Imagine Institute, Université Paris Cité, 24 Bd du Montparnasse, 75015 Paris, France
- Biostatistic Unit, Necker University Hospital, Assistance Publique-Hôpitaux de Paris, 149 rue de Sèvre, 75015 Paris, France
- Human Genetics of Infectious Diseases: Complex Predisposition, Institut National de la Santé et de la Recherche Médicale Unité Mixte de Recherche 1163, 24 Bd du Montparnasse, 75015 Paris, France
| | - Jean Feuillard
- Laboratoire d'Hématologie Biologique, Centre Hospitalier Universitaire de Limoges, Centre de Biologie et de Recherche en Santé, rue Bernard Descottes, 87042 Limoges, France
- Unité Mixte de Recherche Centre National de Recherche Scientifique 7276/Institut National de la Santé et de la Recherche Médicale U1262, Contrôle de la Réponse Immune B et Lymphoproliférations, Centre de Biologie et de Recherche en Santé, rue Bernard Descottes, 87025 Limoges, France
| |
Collapse
|
31
|
Armignacco R, Carlier N, Jouinot A, Birtolo MF, de Murat D, Tubach F, Hausfater P, Simon T, Gorochov G, Pourcher V, Beurton A, Goulet H, Manivet P, Bertherat J, Assié G. Whole blood transcriptome signature predicts severe forms of COVID-19: Results from the COVIDeF cohort study. Funct Integr Genomics 2024; 24:107. [PMID: 38772950 PMCID: PMC11108918 DOI: 10.1007/s10142-024-01359-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2024] [Revised: 04/11/2024] [Accepted: 04/12/2024] [Indexed: 05/23/2024]
Abstract
COVID-19 is associated with heterogeneous outcome. Early identification of a severe progression of the disease is essential to properly manage the patients and improve their outcome. Biomarkers reflecting an increased inflammatory response, as well as individual features including advanced age, male gender, and pre-existing comorbidities, are risk factors of severe COVID-19. Yet, these features show limited accuracy for outcome prediction. The aim was to evaluate the prognostic value of whole blood transcriptome at an early stage of the disease. Blood transcriptome of patients with mild pneumonia was profiled. Patients with subsequent severe COVID-19 were compared to those with favourable outcome, and a molecular predictor based on gene expression was built. Unsupervised classification discriminated patients who would later develop a COVID-19-related severe pneumonia. The corresponding gene expression signature reflected the immune response to the viral infection dominated by a prominent type I interferon, with IFI27 among the most over-expressed genes. A 48-genes transcriptome signature predicting the risk of severe COVID-19 was built on a training cohort, then validated on an external independent cohort, showing an accuracy of 81% for predicting severe outcome. These results identify an early transcriptome signature of severe COVID-19 pneumonia, with a possible relevance to improve COVID-19 patient management.
Collapse
Affiliation(s)
- Roberta Armignacco
- Université Paris Cité, CNRS UMR8104, INSERM U1016, Institut Cochin, F-75014, Paris, France.
| | - Nicolas Carlier
- Service de Pneumologie, AP-HP, Hôpital Cochin, 75014, Paris, France
| | - Anne Jouinot
- Université Paris Cité, CNRS UMR8104, INSERM U1016, Institut Cochin, F-75014, Paris, France
- Service d'Endocrinologie, Center for Rare Adrenal Diseases, AP-HP, Hôpital Cochin, 75014, Paris, France
| | | | - Daniel de Murat
- Université Paris Cité, CNRS UMR8104, INSERM U1016, Institut Cochin, F-75014, Paris, France
| | - Florence Tubach
- Sorbonne Université, INSERM, Institut Pierre Louis d'Epidémiologie Et de Santé Publique, AP-HP, 1901, F-75013, Paris, France
| | - Pierre Hausfater
- Emergency Department, APHP-Sorbonne Université, Hôpital Pitié-Salpêtrière, GRC-14 BIOSFAST, CIMI, UMR 1135, Sorbonne Université, Paris, France
| | - Tabassome Simon
- Service de Pharmacologie, Plateforme de Recherche Clinique URC-CRC-CRB de L'Est Parisien, Assistance Publique-Hôpitaux de Paris, Hôpital Saint Antoine, Sorbonne Université, Paris, France
| | - Guy Gorochov
- Centre d'Immunologie Et Des Maladies Infectieuses (CIMI), Department of Immunology, Sorbonne Université, Inserm, Hôpital Pitié Salpêtrière, Groupe Hospitalo-Universitaire Assistance Publique - Hôpitaux de Paris, Paris, France
| | - Valérie Pourcher
- Department of Infectious Diseases, Hôpital Pitié Salpêtrière, Groupe Hospitalo-Universitaire Assistance Publique - Hôpitaux de Paris, Sorbonne Université, Paris, France
| | - Alexandra Beurton
- Service de Médecine Intensive Réanimation EOLE - Département R3S - Sorbonne, Université - Hôpital Universitaire Pitié - Salpêtrière - Assistance Publique Hôpitaux de Paris - 83 Boulevard de L'Hôpital, 75013, Paris, France
- UMRS 1158 Inserm-Sorbonne Université "Neurophysiologie Respiratoire Expérimentale Et Clinique'' Intensive Care Unit, Hôpital Tenon, Assistance Publique-Hôpitaux de Paris, Paris, France
| | - Hélène Goulet
- Emergency Department, Hôpital Tenon, Assistance Publique-Hôpitaux de Paris, Paris, France
| | - Philippe Manivet
- INSERM UMR 1141 "NeuroDiderot", Université Paris Cité, FHU I2-D2, Paris, France
- AP-HP, DMU BioGem, Centre de Ressources Biologiques Biobank Lariboisière/Saint Louis (BB-0033-00064), Hôpital Lariboisière, Paris, France
| | - Jérôme Bertherat
- Université Paris Cité, CNRS UMR8104, INSERM U1016, Institut Cochin, F-75014, Paris, France
- Service d'Endocrinologie, Center for Rare Adrenal Diseases, AP-HP, Hôpital Cochin, 75014, Paris, France
| | - Guillaume Assié
- Université Paris Cité, CNRS UMR8104, INSERM U1016, Institut Cochin, F-75014, Paris, France.
- Service d'Endocrinologie, Center for Rare Adrenal Diseases, AP-HP, Hôpital Cochin, 75014, Paris, France.
| |
Collapse
|
32
|
Monneret G, Voirin N, Richard JC, Cour M, Rimmelé T, Garnier L, Yonis H, Coudereau R, Gossez M, Malcus C, Wallet F, Delignette MC, Dailler F, Buisson M, Argaud L, Lukaszewicz AC, Venet F. Monitoring monocyte HLA-DR expression and CD4 + T lymphocyte count in dexamethasone-treated severe COVID-19 patients. Ann Intensive Care 2024; 14:76. [PMID: 38762684 PMCID: PMC11102415 DOI: 10.1186/s13613-024-01310-5] [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: 12/18/2023] [Accepted: 05/12/2024] [Indexed: 05/20/2024] Open
Abstract
BACKGROUND A 10-day dexamethasone regimen has emerged as the internationally adopted standard-of-care for severe COVID-19 patients. However, the immune response triggered by SARS-CoV-2 infection remains a complex and dynamic phenomenon, leading to various immune profiles and trajectories. The immune status of severe COVID-19 patients following complete dexamethasone treatment has yet to be thoroughly documented. RESULTS To analyze monocyte HLA-DR expression (mHLA-DR) and CD4 + T lymphocyte count (CD4) in critically ill COVID-19 patients after a dexamethasone course and evaluate their association with 28-day ICU mortality, adult COVID-19 patients (n = 176) with an ICU length of stay of at least 10 days and under dexamethasone treatment were included. Associations between each biomarker value (or in combination) measured at day 10 after ICU admission and 28-day mortality in ICU were evaluated. At day 10, the majority of patients presented decreased values of both parameters. A significant association between low mHLA-DR and 28-day mortality was observed. This association remained significant in a multivariate analysis including age, comorbidities or pre-existing immunosuppression (adjusted Hazard ratio (aHR) = 2.86 [1.30-6.32], p = 0.009). Similar results were obtained with decreased CD4 + T cell count (aHR = 2.10 [1.09-4.04], p = 0.027). When combining these biomarkers, patients with both decreased mHLA-DR and low CD4 presented with an independent and significant elevated risk of 28-day mortality (i.e., 60%, aHR = 4.83 (1.72-13.57), p = 0.001). CONCLUSIONS By using standardized immunomonitoring tools available in clinical practice, it is possible to identify a subgroup of patients at high risk of mortality at the end of a 10-day dexamethasone treatment. This emphasizes the significance of integrating immune monitoring into the surveillance of intensive care patients in order to guide further immumodulation approaches.
Collapse
Affiliation(s)
- Guillaume Monneret
- Immunology Laboratory, Hospices Civils de Lyon, Edouard Herriot Hospital, 5 Place d'Arsonval, 69437, Lyon, France.
- EA 7426 "Pathophysiology of Injury-Induced Immunosuppression", Joint Research Unit HCL-bioMérieux, (Université Claude Bernard Lyon 1 - Hospices Civils de Lyon - bioMérieux), 69003, Lyon, France.
| | | | - Jean-Christophe Richard
- Medical Intensive Care Department, Hospices Civils de Lyon, Croix-Rousse University Hospital, 69004, Lyon, France
| | - Martin Cour
- Medical Intensive Care Department, Hospices Civils de Lyon, Edouard Herriot Hospital, 69437, Lyon, France
| | - Thomas Rimmelé
- EA 7426 "Pathophysiology of Injury-Induced Immunosuppression", Joint Research Unit HCL-bioMérieux, (Université Claude Bernard Lyon 1 - Hospices Civils de Lyon - bioMérieux), 69003, Lyon, France
- Anesthesia and Critical Care Medicine Department, Hospices Civils de Lyon, Edouard Herriot Hospital, 69437, Lyon, France
| | - Lorna Garnier
- Immunology Laboratory, Hospices Civils de Lyon, Lyon-Sud University Hospital, 69495, Pierre Bénite, France
| | - Hodane Yonis
- Medical Intensive Care Department, Hospices Civils de Lyon, Croix-Rousse University Hospital, 69004, Lyon, France
| | - Remy Coudereau
- Immunology Laboratory, Hospices Civils de Lyon, Edouard Herriot Hospital, 5 Place d'Arsonval, 69437, Lyon, France
- EA 7426 "Pathophysiology of Injury-Induced Immunosuppression", Joint Research Unit HCL-bioMérieux, (Université Claude Bernard Lyon 1 - Hospices Civils de Lyon - bioMérieux), 69003, Lyon, France
| | - Morgane Gossez
- Immunology Laboratory, Hospices Civils de Lyon, Edouard Herriot Hospital, 5 Place d'Arsonval, 69437, Lyon, France
- Centre International de Recherche en Infectiologie (CIRI), Inserm U1111, CNRS, UMR5308, Ecole Normale Supérieure de Lyon, Université Claude, Bernard-Lyon 1, Lyon, France
| | - Christophe Malcus
- Immunology Laboratory, Hospices Civils de Lyon, Edouard Herriot Hospital, 5 Place d'Arsonval, 69437, Lyon, France
- EA 7426 "Pathophysiology of Injury-Induced Immunosuppression", Joint Research Unit HCL-bioMérieux, (Université Claude Bernard Lyon 1 - Hospices Civils de Lyon - bioMérieux), 69003, Lyon, France
| | - Florent Wallet
- Intensive Care Department, Hospices Civils de Lyon, Lyon-Sud University Hospital, 69495, Pierre-Bénite, France
| | - Marie-Charlotte Delignette
- Anesthesia and Critical Care Medicine Department, Hospices Civils de Lyon, Croix-Rousse University Hospital, 69004, Lyon, France
| | - Frederic Dailler
- Neurological Anesthesiology and Intensive Care Department, Hospices Civils de Lyon, Pierre Wertheimer Hospital, Lyon, France
| | - Marielle Buisson
- Centre d'Investigation Clinique de Lyon (CIC 1407 Inserm), Hospices Civils de Lyon, Lyon, France
| | - Laurent Argaud
- Medical Intensive Care Department, Hospices Civils de Lyon, Croix-Rousse University Hospital, 69004, Lyon, France
| | - Anne-Claire Lukaszewicz
- EA 7426 "Pathophysiology of Injury-Induced Immunosuppression", Joint Research Unit HCL-bioMérieux, (Université Claude Bernard Lyon 1 - Hospices Civils de Lyon - bioMérieux), 69003, Lyon, France
- Medical Intensive Care Department, Hospices Civils de Lyon, Edouard Herriot Hospital, 69437, Lyon, France
| | - Fabienne Venet
- Immunology Laboratory, Hospices Civils de Lyon, Edouard Herriot Hospital, 5 Place d'Arsonval, 69437, Lyon, France
- Centre International de Recherche en Infectiologie (CIRI), Inserm U1111, CNRS, UMR5308, Ecole Normale Supérieure de Lyon, Université Claude, Bernard-Lyon 1, Lyon, France
| |
Collapse
|
33
|
Paukner S, Kimber S, Cumper C, Rea-Davies T, Sueiro Ballesteros L, Kirkham C, Hargreaves A, Gelone SP, Richards C, Wicha WW. In Vivo Immune-Modulatory Activity of Lefamulin in an Influenza Virus A (H1N1) Infection Model in Mice. Int J Mol Sci 2024; 25:5401. [PMID: 38791439 PMCID: PMC11121702 DOI: 10.3390/ijms25105401] [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: 04/08/2024] [Revised: 05/07/2024] [Accepted: 05/13/2024] [Indexed: 05/26/2024] Open
Abstract
Lefamulin is a first-in-class systemic pleuromutilin antimicrobial and potent inhibitor of bacterial translation, and the most recent novel antimicrobial approved for the treatment of community-acquired pneumonia (CAP). It exhibits potent antibacterial activity against the most prevalent bacterial pathogens that cause typical and atypical pneumonia and other infectious diseases. Early studies indicate additional anti-inflammatory activity. In this study, we further investigated the immune-modulatory activity of lefamulin in the influenza A/H1N1 acute respiratory distress syndrome (ARDS) model in BALB/c mice. Comparators included azithromycin, an anti-inflammatory antimicrobial, and the antiviral oseltamivir. Lefamulin significantly decreased the total immune cell infiltration, specifically the neutrophils, inflammatory monocytes, CD4+ and CD8+ T-cells, NK cells, and B-cells into the lung by Day 6 at both doses tested compared to the untreated vehicle control group (placebo), whereas azithromycin and oseltamivir did not significantly affect the total immune cell counts at the tested dosing regimens. Bronchioalveolar lavage fluid concentrations of pro-inflammatory cytokines and chemokines including TNF-α, IL-6, IL-12p70, IL-17A, IFN-γ, and GM-CSF were significantly reduced, and MCP-1 concentrations were lowered (not significantly) by lefamulin at the clinically relevant 'low' dose on Day 3 when the viral load peaked. Similar effects were also observed for oseltamivir and azithromycin. Lefamulin also decreased the viral load (TCID50) by half a log10 by Day 6 and showed positive effects on the gross lung pathology and survival. Oseltamivir and lefamulin were efficacious in the suppression of the development of influenza-induced bronchi-interstitial pneumonia, whereas azithromycin did not show reduced pathology at the tested treatment regimen. The observed anti-inflammatory and immune-modulatory activity of lefamulin at the tested treatment regimens highlights a promising secondary pharmacological property of lefamulin. While these results require confirmation in a clinical trial, they indicate that lefamulin may provide an immune-modulatory activity beyond its proven potent antibacterial activity. This additional activity may benefit CAP patients and potentially prevent acute lung injury (ALI) and ARDS.
Collapse
Affiliation(s)
- Susanne Paukner
- Nabriva Therapeutics GmbH, Leberstrasse 20, 1110 Vienna, Austria;
| | - Sandra Kimber
- Charles River Discovery, Portishead BS20 7AW, UK; (S.K.); (C.C.); (T.R.-D.); (L.S.B.); (C.K.); (C.R.)
| | - Charlotte Cumper
- Charles River Discovery, Portishead BS20 7AW, UK; (S.K.); (C.C.); (T.R.-D.); (L.S.B.); (C.K.); (C.R.)
| | - Tina Rea-Davies
- Charles River Discovery, Portishead BS20 7AW, UK; (S.K.); (C.C.); (T.R.-D.); (L.S.B.); (C.K.); (C.R.)
| | - Lorena Sueiro Ballesteros
- Charles River Discovery, Portishead BS20 7AW, UK; (S.K.); (C.C.); (T.R.-D.); (L.S.B.); (C.K.); (C.R.)
| | - Christopher Kirkham
- Charles River Discovery, Portishead BS20 7AW, UK; (S.K.); (C.C.); (T.R.-D.); (L.S.B.); (C.K.); (C.R.)
| | | | | | - Claire Richards
- Charles River Discovery, Portishead BS20 7AW, UK; (S.K.); (C.C.); (T.R.-D.); (L.S.B.); (C.K.); (C.R.)
| | | |
Collapse
|
34
|
Qin J, Wang G, Han D. Mesenchymal Stem Cells on Mortality in COVID-19 Patients: A Systematic Review and Meta-Analysis of Randomized Controlled Trials. Stem Cell Rev Rep 2024; 20:931-937. [PMID: 38427315 DOI: 10.1007/s12015-024-10705-7] [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] [Accepted: 02/26/2024] [Indexed: 03/02/2024]
Abstract
BACKGROUND The Coronavirus disease-2019 (COVID-19) pandemic continues, and the death toll continues to surge. This meta-analysis aimed to determine the efficacy of mesenchymal stem cells (MSCs) on mortality in patients with COVID-19. METHODS A systematic search was made of PubMed, Embase, Cochrane Library, and clinicaltrials.gov, without language restrictions. Randomized controlled trials (RCTs) on treatment of COVID-19 with MSCs, compared with placebo or blank, were reviewed. Studies were pooled to risk ratios (RRs), with 95% confidence intervals (CIs). RESULTS Seventeen RCTs (enrolling 1019 participants) met the inclusion criteria. MSCs showed significant effect on 28-day mortality (RR 0.76, 95% CI 0.62 to 0.93; P = 0.008). There was no statistically significant difference in 60-day mortality (RR 0.87, 95% CI 0.70 to 1.09; P = 0.22), and 90-day mortality (RR 0.91, 95% CI 0.72 to 1.15; P = 0.44) between the two groups. CONCLUSIONS MSCs significantly reduced 28-day mortality in patients with COVID-19. The long-term effect of MSCs on mortality require further study.
Collapse
Affiliation(s)
- Jinlv Qin
- Radioimmunoassay Center, Shaanxi Provincial People's Hospital, Xi'an, 710068, Shaanxi, China
| | - Guizuo Wang
- Department of Respiratory and Critical Care Medicine, Shaanxi Provincial People's Hospital, No. 256, West Youyi Road, Xi'an, 710068, Shaanxi, China
| | - Dong Han
- Department of Respiratory and Critical Care Medicine, Shaanxi Provincial People's Hospital, No. 256, West Youyi Road, Xi'an, 710068, Shaanxi, China.
| |
Collapse
|
35
|
Ambrożek-Latecka M, Kozlowski P, Hoser G, Bandyszewska M, Hanusek K, Nowis D, Gołąb J, Grzanka M, Piekiełko-Witkowska A, Schulz L, Hornung F, Deinhardt-Emmer S, Kozlowska E, Skirecki T. SARS-CoV-2 and its ORF3a, E and M viroporins activate inflammasome in human macrophages and induce of IL-1α in pulmonary epithelial and endothelial cells. Cell Death Discov 2024; 10:191. [PMID: 38664396 PMCID: PMC11045860 DOI: 10.1038/s41420-024-01966-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2024] [Revised: 04/09/2024] [Accepted: 04/15/2024] [Indexed: 04/28/2024] Open
Abstract
Inflammasome assembly is a potent mechanism responsible for the host protection against pathogens, including viruses. When compromised, it can allow viral replication, while when disrupted, it can perpetuate pathological responses by IL-1 signaling and pyroptotic cell death. SARS-CoV-2 infection was shown to activate inflammasome in the lungs of COVID-19 patients, however, potential mechanisms responsible for this response are not fully elucidated. In this study, we investigated the effects of ORF3a, E and M SARS-CoV-2 viroporins in the inflammasome activation in major populations of alveolar sentinel cells: macrophages, epithelial and endothelial cells. We demonstrated that each viroporin is capable of activation of the inflammasome in macrophages to trigger pyroptosis-like cell death and IL-1α release from epithelial and endothelial cells. Small molecule NLRP3 inflammasome inhibitors reduced IL-1 release but weakly affected the pyroptosis. Importantly, we discovered that while SARS-CoV-2 could not infect the pulmonary microvascular endothelial cells it induced IL-1α and IL-33 release. Together, these findings highlight the essential role of macrophages as the major inflammasome-activating cell population in the lungs and point to endothelial cell expressed IL-1α as a potential novel component driving the pulmonary immunothromobosis in COVID-19.
Collapse
Affiliation(s)
- Magdalena Ambrożek-Latecka
- Department of Translational Immunology and Experimental Intensive Care, Centre of Translational Research, Centre of Postgraduate Medical Education, Warsaw, Poland
| | - Piotr Kozlowski
- Department of Molecular Biology, Institute of Biochemistry, Faculty of Biology, University of Warsaw, Warsaw, Poland
| | - Grażyna Hoser
- Department of Translational Immunology and Experimental Intensive Care, Centre of Translational Research, Centre of Postgraduate Medical Education, Warsaw, Poland
| | - Magdalena Bandyszewska
- Department of Translational Immunology and Experimental Intensive Care, Centre of Translational Research, Centre of Postgraduate Medical Education, Warsaw, Poland
| | - Karolina Hanusek
- Department of Biochemistry and Molecular Biology, Centre of Translational Research, Centre of Postgraduate Medical Education, Warsaw, Poland
| | - Dominika Nowis
- Laboratory of Experimental Medicine, Faculty of Medicine, Medial University of Warsaw, Warsaw, Poland
| | - Jakub Gołąb
- Department of Immunology, Medical University of Warsaw, Warsaw, Poland
| | - Małgorzata Grzanka
- Department of Biochemistry and Molecular Biology, Centre of Translational Research, Centre of Postgraduate Medical Education, Warsaw, Poland
| | - Agnieszka Piekiełko-Witkowska
- Department of Biochemistry and Molecular Biology, Centre of Translational Research, Centre of Postgraduate Medical Education, Warsaw, Poland
| | - Luise Schulz
- Institute of Medical Microbiology, Jena University Hospital, Jena, Germany
| | - Franziska Hornung
- Institute of Medical Microbiology, Jena University Hospital, Jena, Germany
| | | | - Ewa Kozlowska
- Department of Immunology, Institute of Functional Biology and Ecology, Faculty of Biology, University of Warsaw, Warsaw, Poland
| | - Tomasz Skirecki
- Department of Translational Immunology and Experimental Intensive Care, Centre of Translational Research, Centre of Postgraduate Medical Education, Warsaw, Poland.
| |
Collapse
|
36
|
Galati D, Mallardo D, Nicastro C, Zanotta S, Capitelli L, Lombardi C, Baino B, Cavalcanti E, Sale S, Labonia F, Boenzi R, Atripaldi L, Ascierto PA, Bocchino M. The Dysregulation of the Monocyte-Dendritic Cell Interplay Is Associated with In-Hospital Mortality in COVID-19 Pneumonia. J Clin Med 2024; 13:2481. [PMID: 38731010 PMCID: PMC11084469 DOI: 10.3390/jcm13092481] [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: 03/15/2024] [Revised: 04/19/2024] [Accepted: 04/21/2024] [Indexed: 05/13/2024] Open
Abstract
Background: The monocyte-phagocyte system (MPS), including monocytes/macrophages and dendritic cells (DCs), plays a key role in anti-viral immunity. We aimed to analyze the prognostic value of the MPS components on in-hospital mortality in a cohort of 58 patients (M/F; mean age ± SD years) with COVID-19 pneumonia and 22 age- and sex-matched healthy controls. Methods: We measured frequencies and absolute numbers of peripheral blood CD169+ monocytes, conventional CD1c+ and CD141+ (namely cDC2 and cDC1), and plasmacytoid CD303+ DCs by means of multi-parametric flow cytometry. A gene profile analysis of 770 immune-inflammatory-related human genes and 20 SARS-CoV-2 genes was also performed. Results: Median frequencies and absolute counts of CD169-expressing monocytes were significantly higher in COVID-19 patients than in controls (p 0.04 and p 0.01, respectively). Conversely, percentages and absolute numbers of all DC subsets were markedly depleted in patients (p < 0.0001). COVID-19 cases with absolute counts of CD169+ monocytes above the median value of 114.68/μL had significantly higher in-hospital mortality (HR 4.96; 95% CI: 1.42-17.27; p = 0.02). Interleukin (IL)-6 concentrations were significantly increased in COVID-19 patients (p < 0.0001 vs. controls), and negatively correlated with the absolute counts of circulating CD1c+ cDC2 (r = -0.29, p = 0.034) and CD303+ pDC (r = -0.29, p = 0.036) subsets. Viral genes were upregulated in patients with worse outcomes along with inflammatory mediators such as interleukin (IL)-1 beta, tumor necrosis-α (TNF-α) and the anticoagulant protein (PROS1). Conversely, surviving patients had upregulated genes related to inflammatory and anti-viral-related pathways along with the T cell membrane molecule CD4. Conclusions: Our results suggest that the dysregulated interplay between the different components of the MPS along with the imbalance between viral gene expression and host anti-viral immunity negatively impacts COVID-19 outcomes. Although the clinical scenario of COVID-19 has changed over time, a deepening of its pathogenesis remains a priority in clinical and experimental research.
Collapse
Affiliation(s)
- Domenico Galati
- Hematology-Oncology and Stem Cell Transplantation Unit, Department of Hematology and Innovative Diagnostics, Istituto Nazionale Tumori–IRCCS-Fondazione G. Pascale, 80131 Naples, Italy; (D.G.); (S.Z.)
| | - Domenico Mallardo
- Unit of Melanoma and Innovative Therapy, Department of Melanoma, Cancer Immunotherapy and Development Therapeutics, Istituto Nazionale Tumori–IRCCS-Fondazione G. Pascale, 80131 Naples, Italy; (D.M.); (P.A.A.)
| | - Carmine Nicastro
- Clinical Biochemistry Unit, AORN dei Colli, Ospedale Monaldi, 80131 Naples, Italy; (C.N.); (S.S.); (R.B.); (L.A.)
| | - Serena Zanotta
- Hematology-Oncology and Stem Cell Transplantation Unit, Department of Hematology and Innovative Diagnostics, Istituto Nazionale Tumori–IRCCS-Fondazione G. Pascale, 80131 Naples, Italy; (D.G.); (S.Z.)
| | - Ludovica Capitelli
- Respiratory Medicine Division, Department of Clinical Medicine and Surgery, Federico II University, Monaldi Hospital, 80131 Naples, Italy; (L.C.); (C.L.); (B.B.)
| | - Carmen Lombardi
- Respiratory Medicine Division, Department of Clinical Medicine and Surgery, Federico II University, Monaldi Hospital, 80131 Naples, Italy; (L.C.); (C.L.); (B.B.)
| | - Bianca Baino
- Respiratory Medicine Division, Department of Clinical Medicine and Surgery, Federico II University, Monaldi Hospital, 80131 Naples, Italy; (L.C.); (C.L.); (B.B.)
| | - Ernesta Cavalcanti
- Laboratory Medicine Unit, Istituto Nazionale Tumori–IRCCS-Fondazione G. Pascale, 80131 Naples, Italy; (E.C.)
| | - Silvia Sale
- Clinical Biochemistry Unit, AORN dei Colli, Ospedale Monaldi, 80131 Naples, Italy; (C.N.); (S.S.); (R.B.); (L.A.)
| | - Francesco Labonia
- Laboratory Medicine Unit, Istituto Nazionale Tumori–IRCCS-Fondazione G. Pascale, 80131 Naples, Italy; (E.C.)
| | - Rita Boenzi
- Clinical Biochemistry Unit, AORN dei Colli, Ospedale Monaldi, 80131 Naples, Italy; (C.N.); (S.S.); (R.B.); (L.A.)
| | - Luigi Atripaldi
- Clinical Biochemistry Unit, AORN dei Colli, Ospedale Monaldi, 80131 Naples, Italy; (C.N.); (S.S.); (R.B.); (L.A.)
| | - Paolo Antonio Ascierto
- Unit of Melanoma and Innovative Therapy, Department of Melanoma, Cancer Immunotherapy and Development Therapeutics, Istituto Nazionale Tumori–IRCCS-Fondazione G. Pascale, 80131 Naples, Italy; (D.M.); (P.A.A.)
| | - Marialuisa Bocchino
- Respiratory Medicine Division, Department of Clinical Medicine and Surgery, Federico II University, Monaldi Hospital, 80131 Naples, Italy; (L.C.); (C.L.); (B.B.)
| |
Collapse
|
37
|
Nielsen FM, Klitgaard TL, Siegemund M, Laake JH, Thormar KM, Cole JM, Aagaard SR, Bunzel AMG, Vestergaard SR, Langhoff PK, Pedersen CH, Hejlesen JØ, Abdelhamid S, Dietz A, Gebhard CE, Zellweger N, Hollinger A, Poulsen LM, Weihe S, Andersen-Ranberg NC, Pedersen UG, Mathiesen O, Andreasen AS, Brix H, Thomsen JJ, Petersen CH, Bestle MH, Wichmann S, Lund MS, Mortensen KM, Brand BA, Haase N, Iversen SA, Marcussen KV, Brøchner AC, Borup M, Grøfte T, Hildebrandt T, Kjær MBN, Engstrøm J, Lange T, Perner A, Schjørring OL, Rasmussen BS. Lower vs Higher Oxygenation Target and Days Alive Without Life Support in COVID-19: The HOT-COVID Randomized Clinical Trial. JAMA 2024; 331:1185-1194. [PMID: 38501214 PMCID: PMC10951852 DOI: 10.1001/jama.2024.2934] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/23/2023] [Accepted: 02/28/2024] [Indexed: 03/20/2024]
Abstract
Importance Supplemental oxygen is ubiquitously used in patients with COVID-19 and severe hypoxemia, but a lower dose may be beneficial. Objective To assess the effects of targeting a Pao2 of 60 mm Hg vs 90 mm Hg in patients with COVID-19 and severe hypoxemia in the intensive care unit (ICU). Design, Setting, and Participants Multicenter randomized clinical trial including 726 adults with COVID-19 receiving at least 10 L/min of oxygen or mechanical ventilation in 11 ICUs in Europe from August 2020 to March 2023. The trial was prematurely stopped prior to outcome assessment due to slow enrollment. End of 90-day follow-up was June 1, 2023. Interventions Patients were randomized 1:1 to a Pao2 of 60 mm Hg (lower oxygenation group; n = 365) or 90 mm Hg (higher oxygenation group; n = 361) for up to 90 days in the ICU. Main Outcomes and Measures The primary outcome was the number of days alive without life support (mechanical ventilation, circulatory support, or kidney replacement therapy) at 90 days. Secondary outcomes included mortality, proportion of patients with serious adverse events, and number of days alive and out of hospital, all at 90 days. Results Of 726 randomized patients, primary outcome data were available for 697 (351 in the lower oxygenation group and 346 in the higher oxygenation group). Median age was 66 years, and 495 patients (68%) were male. At 90 days, the median number of days alive without life support was 80.0 days (IQR, 9.0-89.0 days) in the lower oxygenation group and 72.0 days (IQR, 2.0-88.0 days) in the higher oxygenation group (P = .009 by van Elteren test; supplemental bootstrapped adjusted mean difference, 5.8 days [95% CI, 0.2-11.5 days]; P = .04). Mortality at 90 days was 30.2% in the lower oxygenation group and 34.7% in the higher oxygenation group (risk ratio, 0.86 [98.6% CI, 0.66-1.13]; P = .18). There were no statistically significant differences in proportion of patients with serious adverse events or in number of days alive and out of hospital. Conclusion and Relevance In adult ICU patients with COVID-19 and severe hypoxemia, targeting a Pao2 of 60 mm Hg resulted in more days alive without life support in 90 days than targeting a Pao2 of 90 mm Hg. Trial Registration ClinicalTrials.gov Identifier: NCT04425031.
Collapse
Affiliation(s)
- Frederik M. Nielsen
- Department of Anaesthesia and Intensive Care, Aalborg University Hospital, Aalborg, Denmark
- Department of Clinical Medicine, Aalborg University, Aalborg, Denmark
| | - Thomas L. Klitgaard
- Department of Anaesthesia and Intensive Care, Aalborg University Hospital, Aalborg, Denmark
| | - Martin Siegemund
- Department of Intensive Care and Department of Clinical Research, Basel University Hospital, Basel, Switzerland
| | - Jon H. Laake
- Department of Anaesthesia and Intensive Care, Division of Emergencies and Critical Care, Rikshospitalet Medical Centre, Oslo University Hospital, Oslo, Norway
| | - Katrin M. Thormar
- Department of Anesthesia and Intensive Care, Landspitali, University Hospital of Reykjavik, Reykjavik, Iceland
| | - Jade M. Cole
- Department of Intensive Care, Cardiff University Hospital of Wales, Cardiff, Wales
| | - Søren R. Aagaard
- Department of Anaesthesia and Intensive Care, Aalborg University Hospital, Aalborg, Denmark
- Department of Clinical Medicine, Aalborg University, Aalborg, Denmark
| | - Anne-Marie G. Bunzel
- Department of Anaesthesia and Intensive Care, Aalborg University Hospital, Aalborg, Denmark
| | - Stine R. Vestergaard
- Department of Anaesthesia and Intensive Care, Aalborg University Hospital, Aalborg, Denmark
| | - Peter K. Langhoff
- Department of Anaesthesia and Intensive Care, Aalborg University Hospital, Aalborg, Denmark
| | - Caroline H. Pedersen
- Department of Anaesthesia and Intensive Care, Aalborg University Hospital, Aalborg, Denmark
| | - Josefine Ø. Hejlesen
- Department of Anaesthesia and Intensive Care, Aalborg University Hospital, Aalborg, Denmark
| | - Salim Abdelhamid
- Department of Intensive Care and Department of Clinical Research, Basel University Hospital, Basel, Switzerland
| | - Anna Dietz
- Department of Intensive Care and Department of Clinical Research, Basel University Hospital, Basel, Switzerland
| | - Caroline E. Gebhard
- Department of Intensive Care and Department of Clinical Research, Basel University Hospital, Basel, Switzerland
| | - Nuria Zellweger
- Department of Intensive Care and Department of Clinical Research, Basel University Hospital, Basel, Switzerland
| | - Alexa Hollinger
- Department of Intensive Care and Department of Clinical Research, Basel University Hospital, Basel, Switzerland
| | - Lone M. Poulsen
- Department of Anaesthesia and Intensive Care, Zealand University Hospital, Køge, Denmark
| | - Sarah Weihe
- Department of Anaesthesia and Intensive Care, Zealand University Hospital, Køge, Denmark
| | | | - Ulf G. Pedersen
- Department of Anaesthesia and Intensive Care, Zealand University Hospital, Køge, Denmark
| | - Ole Mathiesen
- Department of Anaesthesia and Intensive Care, Zealand University Hospital, Køge, Denmark
| | - Anne Sofie Andreasen
- Department of Anaesthesia and Intensive Care, Copenhagen University Hospital, Herlev, Denmark
| | - Helene Brix
- Department of Anaesthesia and Intensive Care, Copenhagen University Hospital, Herlev, Denmark
| | - Jonas J. Thomsen
- Department of Anaesthesia and Intensive Care, Copenhagen University Hospital, Herlev, Denmark
| | - Christina H. Petersen
- Department of Anaesthesia and Intensive Care, Copenhagen University Hospital, Herlev, Denmark
| | - Morten H. Bestle
- Department of Anaesthesia and Intensive Care, Copenhagen University Hospital, North Zealand, Hillerød, Denmark
| | - Sine Wichmann
- Department of Anaesthesia and Intensive Care, Copenhagen University Hospital, North Zealand, Hillerød, Denmark
| | - Martin S. Lund
- Department of Anaesthesia and Intensive Care, Copenhagen University Hospital, North Zealand, Hillerød, Denmark
| | - Karoline M. Mortensen
- Department of Anaesthesia and Intensive Care, Copenhagen University Hospital, North Zealand, Hillerød, Denmark
| | - Björn A. Brand
- Department of Intensive Care, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Nicolai Haase
- Department of Intensive Care, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Susanne A. Iversen
- Department of Anaesthesia and Intensive Care, Slagelse Hospital, Slagelse, Denmark
| | - Klaus V. Marcussen
- Department of Anaesthesia and Intensive Care, Slagelse Hospital, Slagelse, Denmark
| | - Anne C. Brøchner
- Department of Anaesthesia and Intensive Care, Kolding Hospital, Kolding, Denmark
| | - Morten Borup
- Department of Anaesthesia and Intensive Care, Kolding Hospital, Kolding, Denmark
| | - Thorbjørn Grøfte
- Department of Anaesthesia and Intensive Care, Randers Hospital, Randers, Denmark
| | - Thomas Hildebrandt
- Department of Anaesthesia and Intensive Care, Zealand University Hospital, Roskilde, Denmark
| | - Maj-Brit N. Kjær
- Department of Intensive Care, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Janus Engstrøm
- Copenhagen Trial Unit, Centre for Clinical Intervention, Capital Region, Copenhagen University Hospital, Rigshospitalet, Denmark
| | - Theis Lange
- Section of Biostatistics, University of Copenhagen, Copenhagen, Denmark
| | - Anders Perner
- Department of Intensive Care, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
- Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Olav L. Schjørring
- Department of Anaesthesia and Intensive Care, Aalborg University Hospital, Aalborg, Denmark
- Department of Clinical Medicine, Aalborg University, Aalborg, Denmark
| | - Bodil S. Rasmussen
- Department of Anaesthesia and Intensive Care, Aalborg University Hospital, Aalborg, Denmark
- Department of Clinical Medicine, Aalborg University, Aalborg, Denmark
| |
Collapse
|
38
|
Vajdi M, Karimi A, Hassanizadeh S, Farhangi MA, Bagherniya M, Askari G, Roufogalis BD, Davies NM, Sahebkar A. Effect of polyphenols against complications of COVID-19: current evidence and potential efficacy. Pharmacol Rep 2024; 76:307-327. [PMID: 38498260 DOI: 10.1007/s43440-024-00585-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2023] [Revised: 03/01/2024] [Accepted: 03/03/2024] [Indexed: 03/20/2024]
Abstract
The COVID-19 pandemic that started in 2019 and resulted in significant morbidity and mortality continues to be a significant global health challenge, characterized by inflammation, oxidative stress, and immune system dysfunction.. Developing therapies for preventing or treating COVID-19 remains an important goal for pharmacology and drug development research. Polyphenols are effective against various viral infections and can be extracted and isolated from plants without losing their therapeutic potential. Researchers have developed methods for separating and isolating polyphenols from complex matrices. Polyphenols are effective in treating common viral infections, including COVID-19, and can also boost immunity. Polyphenolic-based antiviral medications can mitigate SARS-CoV-2 enzymes vital to virus replication and infection. Individual polyphenolic triterpenoids, flavonoids, anthraquinonoids, and tannins may also inhibit the SARS-CoV-2 protease. Polyphenol pharmacophore structures identified to date can explain their action and lead to the design of novel anti-COVID-19 compounds. Polyphenol-containing mixtures offer the advantages of a well-recognized safety profile with few known severe side effects. However, studies to date are limited, and further animal studies and randomized controlled trials are needed in future studies. The purpose of this study was to review and present the latest findings on the therapeutic impact of plant-derived polyphenols on COVID-19 infection and its complications. Exploring alternative approaches to traditional therapies could aid in developing novel drugs and remedies against coronavirus infection.
Collapse
Affiliation(s)
- Mahdi Vajdi
- Department of Community Nutrition, School of Nutrition and Food Science, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Arash Karimi
- Traditional Medicine and Hydrotherapy Research Center, Ardabil University of Medical Sciences, Ardabil, Iran
| | - Shirin Hassanizadeh
- Department of Community Nutrition, School of Nutrition and Food Science, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Mahdieh Abbasalizad Farhangi
- Department of Community Nutrition, Faculty of Nutrition and Food Science, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mohammad Bagherniya
- Department of Community Nutrition, Food Security Research Center, School of Nutrition and Food Science, Isfahan University of Medical Sciences, Isfahan, Iran
- Anesthesia and Critical Care Research Center, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Gholamreza Askari
- Department of Community Nutrition, Food Security Research Center, School of Nutrition and Food Science, Isfahan University of Medical Sciences, Isfahan, Iran
- Anesthesia and Critical Care Research Center, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Basil D Roufogalis
- Discipline of Pharmacology, School of Medical Sciences, University of Sydney, Sydney, NSW, Australia
- NICM Health Research Institute, Western Sydney University, Penrith, NSW, Australia
| | - Neal M Davies
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, AB, T6G 2P5, Canada
| | - Amirhossein Sahebkar
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran.
- Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
39
|
Murley C, Pettersson E, Hillert J, Machado A, Friberg E. Coronavirus disease 2019 infection among working-aged people with multiple sclerosis and the impact of disease-modifying therapies. Mult Scler J Exp Transl Clin 2024; 10:20552173241248293. [PMID: 38680165 PMCID: PMC11055478 DOI: 10.1177/20552173241248293] [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: 10/16/2023] [Accepted: 04/03/2024] [Indexed: 05/01/2024] Open
Abstract
Background The risk of coronavirus disease 2019 among people with multiple sclerosis with different disease-modifying therapies is not well established. Objective To investigate the occurrence of coronavirus disease 2019 and the remaining symptoms among people with multiple sclerosis and the associations with different disease-modifying therapies. Methods Individuals aged 20-50 listed in the Swedish Multiple Sclerosis Registry were invited to participate in a survey in 2021. Information on reported coronavirus disease 2019 infection and remaining symptoms were linked to individual-level register data. The risks by disease-modifying therapy of having coronavirus disease 2019 or having remaining symptoms were estimated with logistic regression. Results Of the 4393 participants, 1030 (23.4%) self-reported coronavirus disease 2019 (749 confirmed and 281 suspected). The observed odds for coronavirus disease 2019 did not differ by disease-modifying therapy (p-values <0.05). The majority reporting coronavirus disease 2019 had fully recovered (68.5%), 4.2% were currently/recently sick, and 27.0% had symptoms remaining after 2 months. The most frequently reported remaining symptoms involved one's sense of smell or taste (37.0%), fatigue (20.0%), and breathing (12.0%). No statistically significant associations were observed between having remaining symptoms and the disease-modifying therapy. Conclusion Despite the initial concerns of differing infection risks by MS treatments, we observed no differences in coronavirus disease 2019 occurrence or remaining symptoms among those who had coronavirus disease 2019. Nonetheless, exercising caution in interpreting our findings, it remains implicit that people with multiple sclerosis are particularly susceptible to infection and that lingering symptoms may persist beyond the initial infection.
Collapse
Affiliation(s)
- Chantelle Murley
- Division of Insurance Medicine, Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Emma Pettersson
- Division of Insurance Medicine, Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Jan Hillert
- Division of Neurology, Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Alejandra Machado
- Division of Insurance Medicine, Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Emilie Friberg
- Division of Insurance Medicine, Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
40
|
Ramón A, Bas A, Herrero S, Blasco P, Suárez M, Mateo J. Personalized Assessment of Mortality Risk and Hospital Stay Duration in Hospitalized Patients with COVID-19 Treated with Remdesivir: A Machine Learning Approach. J Clin Med 2024; 13:1837. [PMID: 38610602 PMCID: PMC11013017 DOI: 10.3390/jcm13071837] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2024] [Revised: 03/15/2024] [Accepted: 03/20/2024] [Indexed: 04/14/2024] Open
Abstract
Background: Despite advancements in vaccination, early treatments, and understanding of SARS-CoV-2, its impact remains significant worldwide. Many patients require intensive care due to severe COVID-19. Remdesivir, a key treatment option among viral RNA polymerase inhibitors, lacks comprehensive studies on factors associated with its effectiveness. Methods: We conducted a retrospective study in 2022, analyzing data from 252 hospitalized COVID-19 patients treated with remdesivir. Six machine learning algorithms were compared to predict factors influencing remdesivir's clinical benefits regarding mortality and hospital stay. Results: The extreme gradient boost (XGB) method showed the highest accuracy for both mortality (95.45%) and hospital stay (94.24%). Factors associated with worse outcomes in terms of mortality included limitations in life support, ventilatory support needs, lymphopenia, low albumin and hemoglobin levels, flu and/or coinfection, and cough. For hospital stay, factors included vaccine doses, lung density, pulmonary radiological status, comorbidities, oxygen therapy, troponin, lactate dehydrogenase levels, and asthenia. Conclusions: These findings underscore XGB's effectiveness in accurately categorizing COVID-19 patients undergoing remdesivir treatment.
Collapse
Affiliation(s)
- Antonio Ramón
- Department of Pharmacy, University General Hospital, 46014 Valencia, Spain; (A.R.); (A.B.); (S.H.); (P.B.)
- Medical Analysis Expert Group, Institute of Technology, University of Castilla-La Mancha, 16002 Cuenca, Spain
| | - Andrés Bas
- Department of Pharmacy, University General Hospital, 46014 Valencia, Spain; (A.R.); (A.B.); (S.H.); (P.B.)
| | - Santiago Herrero
- Department of Pharmacy, University General Hospital, 46014 Valencia, Spain; (A.R.); (A.B.); (S.H.); (P.B.)
| | - Pilar Blasco
- Department of Pharmacy, University General Hospital, 46014 Valencia, Spain; (A.R.); (A.B.); (S.H.); (P.B.)
- Medical Analysis Expert Group, Institute of Technology, University of Castilla-La Mancha, 16002 Cuenca, Spain
| | - Miguel Suárez
- Medical Analysis Expert Group, Institute of Technology, University of Castilla-La Mancha, 16002 Cuenca, Spain
- Department of Gastroenterology, Virgen de la Luz Hospital, 16002 Cuenca, Spain
| | - Jorge Mateo
- Medical Analysis Expert Group, Institute of Technology, University of Castilla-La Mancha, 16002 Cuenca, Spain
- Medical Analysis Expert Group, Instituto de Investigación Sanitaria de Castilla-La Mancha (IDISCAM), 45071 Toledo, Spain
| |
Collapse
|
41
|
Conte C, Cipponeri E, Roden M. Diabetes Mellitus, Energy Metabolism, and COVID-19. Endocr Rev 2024; 45:281-308. [PMID: 37934800 PMCID: PMC10911957 DOI: 10.1210/endrev/bnad032] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/10/2023] [Revised: 08/30/2023] [Accepted: 11/01/2023] [Indexed: 11/09/2023]
Abstract
Obesity, diabetes mellitus (mostly type 2), and COVID-19 show mutual interactions because they are not only risk factors for both acute and chronic COVID-19 manifestations, but also because COVID-19 alters energy metabolism. Such metabolic alterations can lead to dysglycemia and long-lasting effects. Thus, the COVID-19 pandemic has the potential for a further rise of the diabetes pandemic. This review outlines how preexisting metabolic alterations spanning from excess visceral adipose tissue to hyperglycemia and overt diabetes may exacerbate COVID-19 severity. We also summarize the different effects of SARS-CoV-2 infection on the key organs and tissues orchestrating energy metabolism, including adipose tissue, liver, skeletal muscle, and pancreas. Last, we provide an integrative view of the metabolic derangements that occur during COVID-19. Altogether, this review allows for better understanding of the metabolic derangements occurring when a fire starts from a small flame, and thereby help reducing the impact of the COVID-19 pandemic.
Collapse
Affiliation(s)
- Caterina Conte
- Department of Human Sciences and Promotion of the Quality of Life, San Raffaele Roma Open University, Rome 00166, Italy
- Department of Endocrinology, Nutrition and Metabolic Diseases, IRCCS MultiMedica, Milan 20099, Italy
| | - Elisa Cipponeri
- Department of Endocrinology, Nutrition and Metabolic Diseases, IRCCS MultiMedica, Milan 20099, Italy
| | - Michael Roden
- Department of Endocrinology and Diabetology, Medical Faculty and University Hospital Düsseldorf, Heinrich-Heine-University Düsseldorf, Düsseldorf 40225, Germany
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich-Heine-University Düsseldorf, Düsseldorf 40225, Germany
- German Center for Diabetes Research, Partner Düsseldorf, Neuherberg 85764, Germany
| |
Collapse
|
42
|
Santacroce E, D’Angerio M, Ciobanu AL, Masini L, Lo Tartaro D, Coloretti I, Busani S, Rubio I, Meschiari M, Franceschini E, Mussini C, Girardis M, Gibellini L, Cossarizza A, De Biasi S. Advances and Challenges in Sepsis Management: Modern Tools and Future Directions. Cells 2024; 13:439. [PMID: 38474403 PMCID: PMC10931424 DOI: 10.3390/cells13050439] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2024] [Revised: 02/27/2024] [Accepted: 02/29/2024] [Indexed: 03/14/2024] Open
Abstract
Sepsis, a critical condition marked by systemic inflammation, profoundly impacts both innate and adaptive immunity, often resulting in lymphopenia. This immune alteration can spare regulatory T cells (Tregs) but significantly affects other lymphocyte subsets, leading to diminished effector functions, altered cytokine profiles, and metabolic changes. The complexity of sepsis stems not only from its pathophysiology but also from the heterogeneity of patient responses, posing significant challenges in developing universally effective therapies. This review emphasizes the importance of phenotyping in sepsis to enhance patient-specific diagnostic and therapeutic strategies. Phenotyping immune cells, which categorizes patients based on clinical and immunological characteristics, is pivotal for tailoring treatment approaches. Flow cytometry emerges as a crucial tool in this endeavor, offering rapid, low cost and detailed analysis of immune cell populations and their functional states. Indeed, this technology facilitates the understanding of immune dysfunctions in sepsis and contributes to the identification of novel biomarkers. Our review underscores the potential of integrating flow cytometry with omics data, machine learning and clinical observations to refine sepsis management, highlighting the shift towards personalized medicine in critical care. This approach could lead to more precise interventions, improving outcomes in this heterogeneously affected patient population.
Collapse
Affiliation(s)
- Elena Santacroce
- Department of Medical and Surgical Sciences for Children & Adults, University of Modena and Reggio Emilia, 41125 Modena, Italy; (E.S.); (M.D.); (A.L.C.); (L.M.); (D.L.T.); (L.G.); (A.C.)
| | - Miriam D’Angerio
- Department of Medical and Surgical Sciences for Children & Adults, University of Modena and Reggio Emilia, 41125 Modena, Italy; (E.S.); (M.D.); (A.L.C.); (L.M.); (D.L.T.); (L.G.); (A.C.)
| | - Alin Liviu Ciobanu
- Department of Medical and Surgical Sciences for Children & Adults, University of Modena and Reggio Emilia, 41125 Modena, Italy; (E.S.); (M.D.); (A.L.C.); (L.M.); (D.L.T.); (L.G.); (A.C.)
| | - Linda Masini
- Department of Medical and Surgical Sciences for Children & Adults, University of Modena and Reggio Emilia, 41125 Modena, Italy; (E.S.); (M.D.); (A.L.C.); (L.M.); (D.L.T.); (L.G.); (A.C.)
| | - Domenico Lo Tartaro
- Department of Medical and Surgical Sciences for Children & Adults, University of Modena and Reggio Emilia, 41125 Modena, Italy; (E.S.); (M.D.); (A.L.C.); (L.M.); (D.L.T.); (L.G.); (A.C.)
| | - Irene Coloretti
- Department of Surgery, Medicine, Dentistry and Morphological Sciences, University of Modena and Reggio Emilia, 41121 Modena, Italy; (I.C.); (S.B.); (M.M.); (E.F.); (C.M.); (M.G.)
| | - Stefano Busani
- Department of Surgery, Medicine, Dentistry and Morphological Sciences, University of Modena and Reggio Emilia, 41121 Modena, Italy; (I.C.); (S.B.); (M.M.); (E.F.); (C.M.); (M.G.)
| | - Ignacio Rubio
- Department of Anesthesiology and Intensive Care Medicine, Center for Sepsis Control and Care, Jena University Hospital, 07747 Jena, Germany;
| | - Marianna Meschiari
- Department of Surgery, Medicine, Dentistry and Morphological Sciences, University of Modena and Reggio Emilia, 41121 Modena, Italy; (I.C.); (S.B.); (M.M.); (E.F.); (C.M.); (M.G.)
| | - Erica Franceschini
- Department of Surgery, Medicine, Dentistry and Morphological Sciences, University of Modena and Reggio Emilia, 41121 Modena, Italy; (I.C.); (S.B.); (M.M.); (E.F.); (C.M.); (M.G.)
| | - Cristina Mussini
- Department of Surgery, Medicine, Dentistry and Morphological Sciences, University of Modena and Reggio Emilia, 41121 Modena, Italy; (I.C.); (S.B.); (M.M.); (E.F.); (C.M.); (M.G.)
| | - Massimo Girardis
- Department of Surgery, Medicine, Dentistry and Morphological Sciences, University of Modena and Reggio Emilia, 41121 Modena, Italy; (I.C.); (S.B.); (M.M.); (E.F.); (C.M.); (M.G.)
| | - Lara Gibellini
- Department of Medical and Surgical Sciences for Children & Adults, University of Modena and Reggio Emilia, 41125 Modena, Italy; (E.S.); (M.D.); (A.L.C.); (L.M.); (D.L.T.); (L.G.); (A.C.)
| | - Andrea Cossarizza
- Department of Medical and Surgical Sciences for Children & Adults, University of Modena and Reggio Emilia, 41125 Modena, Italy; (E.S.); (M.D.); (A.L.C.); (L.M.); (D.L.T.); (L.G.); (A.C.)
| | - Sara De Biasi
- Department of Medical and Surgical Sciences for Children & Adults, University of Modena and Reggio Emilia, 41125 Modena, Italy; (E.S.); (M.D.); (A.L.C.); (L.M.); (D.L.T.); (L.G.); (A.C.)
| |
Collapse
|
43
|
Asiaee M, Nourbakhsh M, Vahedian-Azimi A, Zare M, Jafari R, Atashi SS, Keramatfar A. The feasibility of using acoustic measures for predicting the Total Opacity Scores of chest computed tomography scans in patients with COVID-19. CLINICAL LINGUISTICS & PHONETICS 2024; 38:97-115. [PMID: 36592050 DOI: 10.1080/02699206.2022.2160659] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Revised: 12/10/2022] [Accepted: 12/13/2022] [Indexed: 06/17/2023]
Abstract
To study the possibility of using acoustic parameters, i.e., Acoustic Voice Quality Index (AVQI) and Maximum Phonation Time (MPT) for predicting the degree of lung involvement in COVID-19 patients. This cross-sectional case-control study was conducted on the voice samples collected from 163 healthy individuals and 181 patients with COVID-19. Each participant produced a sustained vowel/a/, and a phonetically balanced Persian text containing 36 syllables. AVQI and MPT were measured using Praat scripts. Each patient underwent a non-enhanced chest computed tomographic scan and the Total Opacity score was rated to assess the degree of lung involvement. The results revealed significant differences between patients with COVID-19 and healthy individuals in terms of AVQI and MPT. A significant difference was also observed between male and female participants in AVQI and MPT. The results from the receiver operating characteristic curve analysis and area under the curve indicated that MPT (0.909) had higher diagnostic accuracy than AVQI (0.771). A significant relationship was observed between AVQI and TO scores. In the case of MPT, however, no such relationship was observed. The findings indicated that MPT was a better classifier in differentiating patients from healthy individuals, in comparison with AVQI. The results also showed that AVQI can be used as a predictor of the degree of patients' and recovered individuals' lung involvement. A formula is suggested for calculating the degree of lung involvement using AVQI.
Collapse
Affiliation(s)
- Maral Asiaee
- Department of Linguistics, Faculty of Literature, Alzahra University, Tehran, Iran
| | - Mandana Nourbakhsh
- Department of Linguistics, Faculty of Literature, Alzahra University, Tehran, Iran
| | - Amir Vahedian-Azimi
- Trauma Research Center, Nursing faculty, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Mohammad Zare
- Department of Statistics, Faculty of Mathematics and Computer Science, Amirkabir University of Technology (Tehran Polytechnic), Tehran, Iran
| | - Ramezan Jafari
- Department of Radiology, Health Research Center, Lifestyle Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Seyed Shahab Atashi
- Department of Food and Drug control, Jundishapour University of Medical Sciences, Ahvaz, Iran
| | | |
Collapse
|
44
|
Teixeira Alves LG, Baumgardt M, Langner C, Fischer M, Maria Adler J, Bushe J, Firsching TC, Mastrobuoni G, Grobe J, Hoenzke K, Kempa S, Gruber AD, Hocke AC, Trimpert J, Wyler E, Landthaler M. Protective role of the HSP90 inhibitor, STA-9090, in lungs of SARS-CoV-2-infected Syrian golden hamsters. BMJ Open Respir Res 2024; 11:e001762. [PMID: 38423952 PMCID: PMC10910676 DOI: 10.1136/bmjresp-2023-001762] [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: 04/13/2023] [Accepted: 01/26/2024] [Indexed: 03/02/2024] Open
Abstract
INTRODUCTION The emergence of new SARS-CoV-2 variants, capable of escaping the humoral immunity acquired by the available vaccines, together with waning immunity and vaccine hesitancy, challenges the efficacy of the vaccination strategy in fighting COVID-19. Improved therapeutic strategies are urgently needed to better intervene particularly in severe cases of the disease. They should aim at controlling the hyperinflammatory state generated on infection, reducing lung tissue pathology and inhibiting viral replication. Previous research has pointed to a possible role for the chaperone HSP90 in SARS-CoV-2 replication and COVID-19 pathogenesis. Pharmacological intervention through HSP90 inhibitors was shown to be beneficial in the treatment of inflammatory diseases, infections and reducing replication of diverse viruses. METHODS In this study, we investigated the effects of the potent HSP90 inhibitor Ganetespib (STA-9090) in vitro on alveolar epithelial cells and alveolar macrophages to characterise its effects on cell activation and viral replication. Additionally, the Syrian hamster animal model was used to evaluate its efficacy in controlling systemic inflammation and viral burden after infection. RESULTS In vitro, STA-9090 reduced viral replication on alveolar epithelial cells in a dose-dependent manner and lowered significantly the expression of proinflammatory genes, in both alveolar epithelial cells and alveolar macrophages. In vivo, although no reduction in viral load was observed, administration of STA-9090 led to an overall improvement of the clinical condition of infected animals, with reduced oedema formation and lung tissue pathology. CONCLUSION Altogether, we show that HSP90 inhibition could serve as a potential treatment option for moderate and severe cases of COVID-19.
Collapse
Affiliation(s)
- Luiz Gustavo Teixeira Alves
- RNA Biology and Posttranscriptional Regulation, Max Delbruck Centre for Molecular Medicine in the Helmholtz Association, Berlin, Germany
| | - Morris Baumgardt
- Corporate Member of Freie Universität Berlin und Humboldt-Universität zu Berlin, Department of Infectious Diseases, Respiratory Medicine and Critical Care, Charité Universitätsmedizin Berlin, Berlin, Germany
| | | | - Mara Fischer
- Corporate Member of Freie Universität Berlin und Humboldt-Universität zu Berlin, Department of Infectious Diseases, Respiratory Medicine and Critical Care, Charité Universitätsmedizin Berlin, Berlin, Germany
| | | | - Judith Bushe
- Research Unit Analytical Pathology, Helmholtz Zentrum Munchen Deutsches Forschungszentrum fur Gesundheit und Umwelt, Neuherberg, Germany
| | | | - Guido Mastrobuoni
- Proteomics and Metabolomics, Max Delbruck Centre for Molecular Medicine in the Helmholtz Association, Berlin, Germany
| | - Jenny Grobe
- Proteomics and Metabolomics, Max Delbruck Centre for Molecular Medicine in the Helmholtz Association, Berlin, Germany
| | - Katja Hoenzke
- Corporate Member of Freie Universität Berlin und Humboldt-Universität zu Berlin, Department of Infectious Diseases, Respiratory Medicine and Critical Care, Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Stefan Kempa
- Proteomics and Metabolomics, Max Delbruck Centre for Molecular Medicine in the Helmholtz Association, Berlin, Germany
| | - Achim Dieter Gruber
- Department of Veterinary Pathology, Free University of Berlin, Berlin, Germany
| | - Andreas Christian Hocke
- Corporate Member of Freie Universität Berlin und Humboldt-Universität zu Berlin, Department of Infectious Diseases, Respiratory Medicine and Critical Care, Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Jakob Trimpert
- Institute of Virology, Free University of Berlin, Berlin, Germany
| | - Emanuel Wyler
- RNA Biology and Posttranscriptional Regulation, Max Delbruck Centre for Molecular Medicine in the Helmholtz Association, Berlin, Germany
| | - Markus Landthaler
- RNA Biology and Posttranscriptional Regulation, Max Delbruck Centre for Molecular Medicine in the Helmholtz Association, Berlin, Germany
- Institute for Biology, Humboldt-Universitat zu Berlin, Berlin, Germany
| |
Collapse
|
45
|
Doubravská L, Htoutou Sedláková M, Fišerová K, Klementová O, Turek R, Langová K, Kolář M. Bacterial Community- and Hospital-Acquired Pneumonia in Patients with Critical COVID-19-A Prospective Monocentric Cohort Study. Antibiotics (Basel) 2024; 13:192. [PMID: 38391578 PMCID: PMC10886267 DOI: 10.3390/antibiotics13020192] [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: 11/24/2023] [Revised: 02/12/2024] [Accepted: 02/13/2024] [Indexed: 02/24/2024] Open
Abstract
The impact of bacterial pneumonia on patients with COVID-19 infection remains unclear. This prospective observational monocentric cohort study aims to determine the incidence of bacterial community- and hospital-acquired pneumonia (CAP and HAP) and its effect on mortality in critically ill COVID-19 patients admitted to the intensive care unit (ICU) at University Hospital Olomouc between 1 November 2020 and 31 December 2022. The secondary objectives of this study include identifying the bacterial etiology of CAP and HAP and exploring the capabilities of diagnostic tools, with a focus on inflammatory biomarkers. Data were collected from the electronic information hospital system, encompassing biomarkers, microbiological findings, and daily visit records, and subsequently evaluated by ICU physicians and clinical microbiologists. Out of 171 patients suffering from critical COVID-19, 46 (27%) had CAP, while 78 (46%) developed HAP. Critically ill COVID-19 patients who experienced bacterial CAP and HAP exhibited higher mortality compared to COVID-19 patients without any bacterial infection, with rates of 38% and 56% versus 11%, respectively. In CAP, the most frequent causative agents were chlamydophila and mycoplasma; Enterobacterales, which were multidrug-resistant in 71% of cases; Gram-negative non-fermenting rods; and Staphylococcus aureus. Notably, no strains of Streptococcus pneumoniae were detected, and only a single strain each of Haemophilus influenzae and Moraxella catarrhalis was isolated. The most frequent etiologic agents causing HAP were Enterobacterales and Gram-negative non-fermenting rods. Based on the presented results, commonly used biochemical markers demonstrated poor predictive and diagnostic accuracy. To confirm the diagnosis of bacterial CAP in our patient cohort, it was necessary to assess the initial values of inflammatory markers (particularly procalcitonin), consider clinical signs indicative of bacterial infection, and/or rely on positive microbiological findings. For HAP diagnostics, it was appropriate to conduct regular detailed clinical examinations (with a focus on evaluating respiratory functions) and closely monitor the dynamics of inflammatory markers (preferably Interleukin-6).
Collapse
Affiliation(s)
- Lenka Doubravská
- Department of Anaesthesiology, Resuscitation and Intensive Care, University Hospital Olomouc, Zdravotniku 248/7, 779 00 Olomouc, Czech Republic
- Department of Anaesthesiology, Resuscitation and Intensive Care, Faculty of Medicine and Dentistry, Palacky University Olomouc, Hnevotinska 3, 779 00 Olomouc, Czech Republic
| | - Miroslava Htoutou Sedláková
- Department of Microbiology, University Hospital Olomouc, Zdravotniku 248/7, 779 00 Olomouc, Czech Republic
- Department of Microbiology, Faculty of Medicine and Dentistry, Palacky University Olomouc, Hnevotinska 3, 779 00 Olomouc, Czech Republic
| | - Kateřina Fišerová
- Department of Microbiology, University Hospital Olomouc, Zdravotniku 248/7, 779 00 Olomouc, Czech Republic
- Department of Microbiology, Faculty of Medicine and Dentistry, Palacky University Olomouc, Hnevotinska 3, 779 00 Olomouc, Czech Republic
| | - Olga Klementová
- Department of Anaesthesiology, Resuscitation and Intensive Care, University Hospital Olomouc, Zdravotniku 248/7, 779 00 Olomouc, Czech Republic
- Department of Anaesthesiology, Resuscitation and Intensive Care, Faculty of Medicine and Dentistry, Palacky University Olomouc, Hnevotinska 3, 779 00 Olomouc, Czech Republic
| | - Radovan Turek
- Department of Anaesthesiology, Resuscitation and Intensive Care, Faculty of Medicine and Dentistry, Palacky University Olomouc, Hnevotinska 3, 779 00 Olomouc, Czech Republic
| | - Kateřina Langová
- Department of Medical Biophysics, Faculty of Medicine and Dentistry, Palacky University Olomouc, Hnevotinska 3, 779 00 Olomouc, Czech Republic
| | - Milan Kolář
- Department of Microbiology, University Hospital Olomouc, Zdravotniku 248/7, 779 00 Olomouc, Czech Republic
- Department of Microbiology, Faculty of Medicine and Dentistry, Palacky University Olomouc, Hnevotinska 3, 779 00 Olomouc, Czech Republic
| |
Collapse
|
46
|
Michels EHA, Appelman B, de Brabander J, van Amstel RBE, van Linge CCA, Chouchane O, Reijnders TDY, Schuurman AR, Sulzer TAL, Klarenbeek AM, Douma RA, Bos LDJ, Wiersinga WJ, Peters-Sengers H, van der Poll T. Host Response Changes and Their Association with Mortality in COVID-19 Patients with Lymphopenia. Am J Respir Crit Care Med 2024; 209:402-416. [PMID: 37948687 PMCID: PMC10878379 DOI: 10.1164/rccm.202305-0890oc] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2023] [Accepted: 11/09/2023] [Indexed: 11/12/2023] Open
Abstract
Rationale: Lymphopenia in coronavirus disease (COVID-19) is associated with increased mortality. Objectives: To explore the association between lymphopenia, host response aberrations, and mortality in patients with lymphopenic COVID-19. Methods: We determined 43 plasma biomarkers reflective of four pathophysiological domains: endothelial cell and coagulation activation, inflammation and organ damage, cytokine release, and chemokine release. We explored if decreased concentrations of lymphocyte-derived proteins in patients with lymphopenia were associated with an increase in mortality. We sought to identify host response phenotypes in patients with lymphopenia by cluster analysis of plasma biomarkers. Measurements and Main Results: A total of 439 general ward patients with COVID-19 were stratified by baseline lymphocyte counts: normal (>1.0 × 109/L; n = 167), mild lymphopenia (>0.5 to ⩽1.0 × 109/L; n = 194), and severe lymphopenia (⩽0.5 × 109/L; n = 78). Lymphopenia was associated with alterations in each host response domain. Lymphopenia was associated with increased mortality. Moreover, in patients with lymphopenia (n = 272), decreased concentrations of several lymphocyte-derived proteins (e.g., CCL5, IL-4, IL-13, IL-17A) were associated with an increase in mortality (at P < 0.01 or stronger significance levels). A cluster analysis revealed three host response phenotypes in patients with lymphopenia: "hyporesponsive" (23.2%), "hypercytokinemic" (36.4%), and "inflammatory-injurious" (40.4%), with substantially differing mortality rates of 9.5%, 5.1%, and 26.4%, respectively. A 10-biomarker model accurately predicted these host response phenotypes in an external cohort with similar mortality distribution. The inflammatory-injurious phenotype showed a remarkable combination of relatively high inflammation and organ damage markers with high antiinflammatory cytokine levels yet low proinflammatory cytokine levels. Conclusions: Lymphopenia in COVID-19 signifies a heterogenous group of patients with distinct host response features. Specific host responses contribute to lymphopenia-associated mortality in COVID-19, including reduced CCL5 levels.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | - Renée A. Douma
- Department of Internal Medicine, Flevo Hospital, Almere, the Netherlands; and
| | | | - W. Joost Wiersinga
- Center for Experimental and Molecular Medicine
- Division of Infectious Diseases, Amsterdam UMC location University of Amsterdam, Amsterdam, the Netherlands
| | - Hessel Peters-Sengers
- Center for Experimental and Molecular Medicine
- Department of Epidemiology and Data Science, Amsterdam UMC location Vrije Universiteit Amsterdam, Amsterdam, the Netherlands
| | - Tom van der Poll
- Center for Experimental and Molecular Medicine
- Division of Infectious Diseases, Amsterdam UMC location University of Amsterdam, Amsterdam, the Netherlands
| |
Collapse
|
47
|
Fischer C, Willscher E, Paschold L, Gottschick C, Klee B, Diexer S, Bosurgi L, Dutzmann J, Sedding D, Frese T, Girndt M, Hoell JI, Gekle M, Addo MM, Schulze Zur Wiesch J, Mikolajczyk R, Binder M, Schultheiß C. SARS-CoV-2 vaccination may mitigate dysregulation of IL-1/IL-18 and gastrointestinal symptoms of the post-COVID-19 condition. NPJ Vaccines 2024; 9:23. [PMID: 38316833 PMCID: PMC10844289 DOI: 10.1038/s41541-024-00815-1] [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: 09/07/2023] [Accepted: 01/24/2024] [Indexed: 02/07/2024] Open
Abstract
The rapid development of safe and effective vaccines helped to prevent severe disease courses after SARS-CoV-2 infection and to mitigate the progression of the COVID-19 pandemic. While there is evidence that vaccination may reduce the risk of developing post-COVID-19 conditions (PCC), this effect may depend on the viral variant. Therapeutic effects of post-infection vaccination have been discussed but the data for individuals with PCC remains inconclusive. In addition, extremely rare side effects after SARS-CoV-2 vaccination may resemble the heterogeneous PCC phenotype. Here, we analyze the plasma levels of 25 cytokines and SARS-CoV-2 directed antibodies in 540 individuals with or without PCC relative to one or two mRNA-based COVID-19 vaccinations as well as in 20 uninfected individuals one month after their initial mRNA-based COVID-19 vaccination. While none of the SARS-CoV-2 naïve individuals reported any persisting sequelae or exhibited PCC-like dysregulation of plasma cytokines, we detected lower levels of IL-1β and IL-18 in patients with ongoing PCC who received one or two vaccinations at a median of six months after infection as compared to unvaccinated PCC patients. This reduction correlated with less frequent reporting of persisting gastrointestinal symptoms. These data suggest that post-infection vaccination in patients with PCC might be beneficial in a subgroup of individuals displaying gastrointestinal symptoms.
Collapse
Affiliation(s)
- Claudia Fischer
- Division of Medical Oncology, University Hospital Basel, Basel, Switzerland
- Laboratory of Translational Immuno-Oncology, Department of Biomedicine, University, and University Hospital Basel, Basel, Switzerland
| | - Edith Willscher
- Department of Internal Medicine IV, Oncology/Hematology, Martin Luther University Halle-Wittenberg, Halle, (Saale), Germany
| | - Lisa Paschold
- Department of Internal Medicine IV, Oncology/Hematology, Martin Luther University Halle-Wittenberg, Halle, (Saale), Germany
| | - Cornelia Gottschick
- Institute for Medical Epidemiology, Biometrics, and Informatics (IMEBI), Interdisciplinary Center for Health Sciences, Medical School of the Martin Luther University Halle-Wittenberg, Halle, (Saale), Germany
| | - Bianca Klee
- Institute for Medical Epidemiology, Biometrics, and Informatics (IMEBI), Interdisciplinary Center for Health Sciences, Medical School of the Martin Luther University Halle-Wittenberg, Halle, (Saale), Germany
| | - Sophie Diexer
- Institute for Medical Epidemiology, Biometrics, and Informatics (IMEBI), Interdisciplinary Center for Health Sciences, Medical School of the Martin Luther University Halle-Wittenberg, Halle, (Saale), Germany
| | - Lidia Bosurgi
- I. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Protozoa Immunology, Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany
| | - Jochen Dutzmann
- Mid-German Heart Center, Department of Cardiology and Intensive Care Medicine, University Hospital, Martin Luther University Halle-Wittenberg, Halle, (Saale), Germany
| | - Daniel Sedding
- Mid-German Heart Center, Department of Cardiology and Intensive Care Medicine, University Hospital, Martin Luther University Halle-Wittenberg, Halle, (Saale), Germany
| | - Thomas Frese
- Institute of General Practice and Family Medicine, Martin-Luther-University Halle-Wittenberg, Halle, (Saale), Germany
| | - Matthias Girndt
- Department of Internal Medicine II, Martin Luther University Halle-Wittenberg, Halle, (Saale), Germany
| | - Jessica I Hoell
- Pediatric Hematology and Oncology, Martin Luther University Halle-Wittenberg, Halle, (Saale), Germany
| | - Michael Gekle
- Julius Bernstein-Institute of Physiology, Faculty of Medicine, Martin Luther University Halle-Wittenberg, Halle, (Saale), Germany
| | - Marylyn M Addo
- I. Department of Internal Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- German Center for Infection Research (DZIF), Partner Site Hamburg-Lübeck-Borstel-Riems, Braunschweig, Germany
- University Medical Center Hamburg-Eppendorf, Institute for Infection Research and Vaccine Development (IIRVD), Hamburg, Germany
| | | | - Rafael Mikolajczyk
- Institute for Medical Epidemiology, Biometrics, and Informatics (IMEBI), Interdisciplinary Center for Health Sciences, Medical School of the Martin Luther University Halle-Wittenberg, Halle, (Saale), Germany
| | - Mascha Binder
- Division of Medical Oncology, University Hospital Basel, Basel, Switzerland
- Laboratory of Translational Immuno-Oncology, Department of Biomedicine, University, and University Hospital Basel, Basel, Switzerland
| | - Christoph Schultheiß
- Division of Medical Oncology, University Hospital Basel, Basel, Switzerland.
- Laboratory of Translational Immuno-Oncology, Department of Biomedicine, University, and University Hospital Basel, Basel, Switzerland.
| |
Collapse
|
48
|
Camps-Vilaró A, Pinsach-Abuin ML, Degano IR, Ramos R, Martí-Lluch R, Elosua R, Subirana I, Solà-Richarte C, Puigmulé M, Pérez A, Vilaró I, Cruz R, Diz-de Almeida S, Nogues X, Masclans JR, Güerri-Fernández R, Marin J, Tizon-Marcos H, Vaquerizo B, Brugada R, Marrugat J. Genetic characteristics involved in COVID-19 severity. The CARGENCORS case-control study and meta-analysis. J Med Virol 2024; 96:e29404. [PMID: 38293834 DOI: 10.1002/jmv.29404] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2023] [Revised: 11/30/2023] [Accepted: 01/04/2024] [Indexed: 02/01/2024]
Abstract
Pre-existing coronary artery disease (CAD), and thrombotic, inflammatory, or virus infectivity response phenomena have been associated with COVID-19 disease severity. However, the association of candidate single nucleotide variants (SNVs) related to mechanisms of COVID-19 complications has been seldom analysed. Our aim was to test and validate the effect of candidate SNVs on COVID-19 severity. CARGENCORS (CARdiovascular GENetic risk score for Risk Stratification of patients positive for SARS-CoV-2 [COVID-19] virus) is an age- and sex-matched case-control study with 818 COVID-19 cases hospitalized with hypoxemia, and 1636 controls with COVID-19 treated at home. The association between severity and SNVs related to CAD (n = 32), inflammation (n = 19), thrombosis (n = 14), virus infectivity (n = 11), and two published to be related to COVID-19 severity was tested with adjusted logistic regression models. Two external independent cohorts were used for meta-analysis (SCOURGE and UK Biobank). After adjustment for potential confounders, 14 new SNVs were associated with COVID-19 severity in the CARGENCORS Study. These SNVs were related to CAD (n = 10), thrombosis (n = 2), and inflammation (n = 2). We also confirmed eight SNVs previously related to severe COVID-19 and virus infectivity. The meta-analysis showed five SNVs associated with severe COVID-19 in adjusted analyses (rs11385942, rs1561198, rs6632704, rs6629110, and rs12329760). We identified 14 novel SNVs and confirmed eight previously related to COVID-19 severity in the CARGENCORS data. In the meta-analysis, five SNVs were significantly associated to COVID-19 severity, one of them previously related to CAD.
Collapse
Affiliation(s)
- Anna Camps-Vilaró
- Registre Gironí del Cor (REGICOR) Study Group, Hospital del Mar Research Institute (IMIM), Barcelona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares, Instituto de Salud Carlos III, Madrid, Spain
- Doctoral College, University of Vic-Central University of Catalonia (UVic-UCC), Vic, Spain
| | - Mel Lina Pinsach-Abuin
- Cardiovascular Genetics Center, Institut d'Investigació Biomèdica de Girona Dr. Josep Trueta (IdIBGi), Salt, Spain
| | - Irene R Degano
- Registre Gironí del Cor (REGICOR) Study Group, Hospital del Mar Research Institute (IMIM), Barcelona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares, Instituto de Salud Carlos III, Madrid, Spain
- Faculty of Medicine, University of Vic-Central University of Catalonia (UVic-UCC), Vic, Spain
- Institute for Research and Innovation in Life Sciences and Health in Central Catalonia (IRIS-CC), Vic, Spain
| | - Rafel Ramos
- Medical Science Department, School of Medicine, University of Girona, Girona, Spain
- Vascular Health Research Group, Institut Universitari per a la Recerca en Atenció Primària Jordi Gol i Gurina, Girona, Spain
- Girona Biomedical Research Institute, Girona, Spain
- Primary Care Services, Catalan Institute of Health, Girona, Spain
| | - Ruth Martí-Lluch
- Vascular Health Research Group, Institut Universitari per a la Recerca en Atenció Primària Jordi Gol i Gurina, Girona, Spain
- Girona Biomedical Research Institute, Girona, Spain
| | - Roberto Elosua
- Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares, Instituto de Salud Carlos III, Madrid, Spain
- Faculty of Medicine, University of Vic-Central University of Catalonia (UVic-UCC), Vic, Spain
- Cardiovascular Epidemiology and Genetics Group, Hospital del Mar Research Institute (IMIM), Barcelona, Spain
| | - Isaac Subirana
- Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares, Instituto de Salud Carlos III, Madrid, Spain
- Cardiovascular Epidemiology and Genetics Group, Hospital del Mar Research Institute (IMIM), Barcelona, Spain
| | - Clàudia Solà-Richarte
- Registre Gironí del Cor (REGICOR) Study Group, Hospital del Mar Research Institute (IMIM), Barcelona, Spain
| | - Marta Puigmulé
- Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares, Instituto de Salud Carlos III, Madrid, Spain
| | - Alexandra Pérez
- Cardiovascular Genetics Center, Institut d'Investigació Biomèdica de Girona Dr. Josep Trueta (IdIBGi), Salt, Spain
| | | | - Raquel Cruz
- Centro Singular de Investigación en Medicina Molecular y Enfermedades Crónicas (CIMUS), Universidade de Santiago de Compostela, Santiago de Compostela, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras, Instituto de Salud Carlos III, Madrid, Spain
| | - Silvia Diz-de Almeida
- Centro Singular de Investigación en Medicina Molecular y Enfermedades Crónicas (CIMUS), Universidade de Santiago de Compostela, Santiago de Compostela, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras, Instituto de Salud Carlos III, Madrid, Spain
| | - Xavier Nogues
- Musculoskeletal Research Unit, Hospital del Mar Research Institute (IMIM), Barcelona, Spain
- Department of Internal Medicine, Hospital del Mar, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Fragilidad y Envejecimiento Saludable, Instituto de Salud Carlos III, Madrid, Spain
- Department of Medicine, Universitat Autònoma de Barcelona (UAB), Bellaterra, Spain
| | - Joan R Masclans
- Critical Illness Research Group (GREPAC), Hospital del Mar Research Institute (IMIM), Barcelona, Spain
- Department of Critical Care, Hospital del Mar, Barcelona, Spain
- Medicine and Life Sciences department, Universitat Pompeu Fabra (UPF), Barcelona, Spain
| | - Roberto Güerri-Fernández
- Department of Medicine, Universitat Autònoma de Barcelona (UAB), Bellaterra, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Infecciosas, Instituto de Salud Carlos III, Madrid, Spain
- Department of Infectious Diseases, Hospital del Mar Research Institute, Barcelona, Spain
| | - Judith Marin
- Critical Illness Research Group (GREPAC), Hospital del Mar Research Institute (IMIM), Barcelona, Spain
- Department of Critical Care, Hospital del Mar, Barcelona, Spain
| | - Helena Tizon-Marcos
- Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares, Instituto de Salud Carlos III, Madrid, Spain
- Biomedical Research in Heart Diseases Group, Hospital del Mar Research Institute (IMIM), Barcelona, Spain
- Department of Cardiology, Hospital del Mar, Barcelona, Spain
| | - Beatriz Vaquerizo
- Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares, Instituto de Salud Carlos III, Madrid, Spain
- Department of Medicine, Universitat Autònoma de Barcelona (UAB), Bellaterra, Spain
- Biomedical Research in Heart Diseases Group, Hospital del Mar Research Institute (IMIM), Barcelona, Spain
- Department of Cardiology, Hospital del Mar, Barcelona, Spain
| | - Ramon Brugada
- Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares, Instituto de Salud Carlos III, Madrid, Spain
- Cardiovascular Genetics Center, Institut d'Investigació Biomèdica de Girona Dr. Josep Trueta (IdIBGi), Salt, Spain
- Medical Science Department, School of Medicine, University of Girona, Girona, Spain
- Department of Cardiology, Hospital Josep Trueta & University of Girona, Girona, Spain
| | - Jaume Marrugat
- Registre Gironí del Cor (REGICOR) Study Group, Hospital del Mar Research Institute (IMIM), Barcelona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares, Instituto de Salud Carlos III, Madrid, Spain
| |
Collapse
|
49
|
Chaytee J, Dinh A, D’Anglejan E, Bouchand F, Jaffal K, Duran C, Le Gall C. Digital health for remote home monitoring of patients with COVID-19 requiring oxygen: a cohort study and literature review. Front Med (Lausanne) 2024; 10:1255798. [PMID: 38356737 PMCID: PMC10864516 DOI: 10.3389/fmed.2023.1255798] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2023] [Accepted: 12/31/2023] [Indexed: 02/16/2024] Open
Abstract
Background The clinical course and outcome of COVID-19 vary widely, from asymptomatic and mild to critical. Elderly patients and patients with comorbidities are at increased risk of respiratory failure and oxygen requirements. Due to the massive surge, the pandemic has created challenges for overwhelmed hospitals. Thus, the original home management of COVID-19 patients requiring oxygen and remote monitoring by a web app and a nurse at home were implemented in our center. We aimed to evaluate the outcome of patients with COVID-19 requiring oxygen who benefited from home remote monitoring management. Methods We performed a retrospective cohort study on all COVID-19 patients requiring oxygen (< 5 L/min) who consulted from October 2020 to April 2021 at our emergency department and were managed with home remote monitoring by a web app and an in-home nurse. We also carried out a literature review of studies on COVID-19 patients requiring oxygen with remote monitoring. Results We included 300 patients [184 (61.3%) male patients, median age 51 years]. The main comorbidities were cardiovascular disease (n = 117; 39.0%), diabetes mellitus (n = 72; 24.0%), and chronic respiratory disease (n = 32; 10.7%). Among the 28 (9.3%) patients readmitted to the hospital, 6 (1.9%) were hospitalized in the intensive care unit, and 3 (0.9%) died. In the multivariable analysis, risk factors for unplanned hospitalization were chronic respiratory failure (odds ratio (OR) =4.476, 95%CI 1.565-12.80), immunosuppression (OR = 3.736, 95%CI 1.208-11.552), and short delay between symptoms onset and start of telemonitoring (OR = 0.744, 95%CI 0.653-0.847). In the literature review, we identified seven other experiences of remote monitoring management. Mortality rate and unplanned hospitalization were low (maximum 1.9 and 12%, respectively). Conclusion Our study confirms the safety of home remote monitoring of patients with COVID-19 who require oxygen, as well as our literature review. However, patients with chronic respiratory failure and immunosuppression should be closely monitored.
Collapse
Affiliation(s)
- Johann Chaytee
- Emergency Department, Victor Dupouy Hospital, Argenteuil, France
| | - Aurélien Dinh
- Infectious Disease Department, Raymond-Poincaré Hospital, AP-HP Paris Saclay University, Garches, France
| | - Emma D’Anglejan
- Infectious Disease Department, Raymond-Poincaré Hospital, AP-HP Paris Saclay University, Garches, France
| | - Frédérique Bouchand
- Pharmacy Department, Raymond-Poincaré Hospital, AP-HP Paris Saclay University, Garches, France
| | - Karim Jaffal
- Infectious Disease Department, Raymond-Poincaré Hospital, AP-HP Paris Saclay University, Garches, France
| | - Clara Duran
- Infectious Disease Department, Raymond-Poincaré Hospital, AP-HP Paris Saclay University, Garches, France
| | | |
Collapse
|
50
|
Duijvelaar E, Gisby J, Peters JE, Bogaard HJ, Aman J. Longitudinal plasma proteomics reveals biomarkers of alveolar-capillary barrier disruption in critically ill COVID-19 patients. Nat Commun 2024; 15:744. [PMID: 38272877 PMCID: PMC10811341 DOI: 10.1038/s41467-024-44986-w] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2023] [Accepted: 01/11/2024] [Indexed: 01/27/2024] Open
Abstract
The pathobiology of respiratory failure in COVID-19 consists of a complex interplay between viral cytopathic effects and a dysregulated host immune response. In critically ill patients, imatinib treatment demonstrated potential for reducing invasive ventilation duration and mortality. Here, we perform longitudinal profiling of 6385 plasma proteins in 318 hospitalised patients to investigate the biological processes involved in critical COVID-19, and assess the effects of imatinib treatment. Nine proteins measured at hospital admission accurately predict critical illness development. Next to dysregulation of inflammation, critical illness is characterised by pathways involving cellular adhesion, extracellular matrix turnover and tissue remodelling. Imatinib treatment attenuates protein perturbations associated with inflammation and extracellular matrix turnover. These proteomic alterations are contextualised using external pulmonary RNA-sequencing data of deceased COVID-19 patients and imatinib-treated Syrian hamsters. Together, we show that alveolar capillary barrier disruption in critical COVID-19 is reflected in the plasma proteome, and is attenuated with imatinib treatment. This study comprises a secondary analysis of both clinical data and plasma samples derived from a clinical trial that was registered with the EU Clinical Trials Register (EudraCT 2020-001236-10, https://www.clinicaltrialsregister.eu/ctr-search/trial/2020-001236-10/NL ) and Netherlands Trial Register (NL8491, https://www.trialregister.nl/trial/8491 ).
Collapse
Affiliation(s)
- Erik Duijvelaar
- Department of Pulmonary Medicine, Amsterdam University Medical Centers, Amsterdam Cardiovascular Sciences, Amsterdam, The Netherlands.
| | - Jack Gisby
- Department of Immunology and Inflammation, Centre for Inflammatory Disease, Imperial College London, London, UK
| | - James E Peters
- Department of Immunology and Inflammation, Centre for Inflammatory Disease, Imperial College London, London, UK
| | - Harm Jan Bogaard
- Department of Pulmonary Medicine, Amsterdam University Medical Centers, Amsterdam Cardiovascular Sciences, Amsterdam, The Netherlands
| | - Jurjan Aman
- Department of Pulmonary Medicine, Amsterdam University Medical Centers, Amsterdam Cardiovascular Sciences, Amsterdam, The Netherlands.
| |
Collapse
|