151
|
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
|
152
|
Murphy WJ, Collins CP, Ashwood P. Potential role of anti-Idiotype responses on the neurological effects of post-acute sequelae of COVID-19 (PASC). Brain Behav Immun 2024; 116:317-320. [PMID: 38123027 DOI: 10.1016/j.bbi.2023.12.017] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/21/2023] [Revised: 12/06/2023] [Accepted: 12/08/2023] [Indexed: 12/23/2023] Open
Affiliation(s)
- William J Murphy
- Departments of Dermatology and Internal Medicine (Hematology/Oncology), UC Davis School of Medicine, Sacramento, CA, United States.
| | - Craig P Collins
- Graduate Program in Immunology, UC Davis, Davis, CA, United States
| | - Paul Ashwood
- Department of Medical Microbiology and Immunology, MIND Institute, UC Davis, Sacramento, CA, United States
| |
Collapse
|
153
|
Ghafari M, Hall M, Golubchik T, Ayoubkhani D, House T, MacIntyre-Cockett G, Fryer HR, Thomson L, Nurtay A, Kemp SA, Ferretti L, Buck D, Green A, Trebes A, Piazza P, Lonie LJ, Studley R, Rourke E, Smith DL, Bashton M, Nelson A, Crown M, McCann C, Young GR, Santos RAND, Richards Z, Tariq MA, Cahuantzi R, Barrett J, Fraser C, Bonsall D, Walker AS, Lythgoe K. Prevalence of persistent SARS-CoV-2 in a large community surveillance study. Nature 2024; 626:1094-1101. [PMID: 38383783 PMCID: PMC10901734 DOI: 10.1038/s41586-024-07029-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2023] [Accepted: 01/04/2024] [Indexed: 02/23/2024]
Abstract
Persistent SARS-CoV-2 infections may act as viral reservoirs that could seed future outbreaks1-5, give rise to highly divergent lineages6-8 and contribute to cases with post-acute COVID-19 sequelae (long COVID)9,10. However, the population prevalence of persistent infections, their viral load kinetics and evolutionary dynamics over the course of infections remain largely unknown. Here, using viral sequence data collected as part of a national infection survey, we identified 381 individuals with SARS-CoV-2 RNA at high titre persisting for at least 30 days, of which 54 had viral RNA persisting at least 60 days. We refer to these as 'persistent infections' as available evidence suggests that they represent ongoing viral replication, although the persistence of non-replicating RNA cannot be ruled out in all. Individuals with persistent infection had more than 50% higher odds of self-reporting long COVID than individuals with non-persistent infection. We estimate that 0.1-0.5% of infections may become persistent with typically rebounding high viral loads and last for at least 60 days. In some individuals, we identified many viral amino acid substitutions, indicating periods of strong positive selection, whereas others had no consensus change in the sequences for prolonged periods, consistent with weak selection. Substitutions included mutations that are lineage defining for SARS-CoV-2 variants, at target sites for monoclonal antibodies and/or are commonly found in immunocompromised people11-14. This work has profound implications for understanding and characterizing SARS-CoV-2 infection, epidemiology and evolution.
Collapse
Affiliation(s)
- Mahan Ghafari
- Big Data Institute, Nuffield Department of Medicine, University of Oxford, Oxford, UK.
- Department of Biology, University of Oxford, Oxford, UK.
- Pandemic Science Institute, University of Oxford, Oxford, UK.
| | - Matthew Hall
- Big Data Institute, Nuffield Department of Medicine, University of Oxford, Oxford, UK
- Pandemic Science Institute, University of Oxford, Oxford, UK
| | - Tanya Golubchik
- Big Data Institute, Nuffield Department of Medicine, University of Oxford, Oxford, UK
- Sydney Infectious Diseases Institute (Sydney ID), School of Medical Sciences, Faculty of Medicine and Health, University of Sydney, Sydney, New South Wales, Australia
| | - Daniel Ayoubkhani
- Office for National Statistics, Newport, UK
- Leicester Real World Evidence Unit, Diabetes Research Centre, University of Leicester, Leicester, UK
| | - Thomas House
- Department of Mathematics, University of Manchester, Manchester, UK
| | - George MacIntyre-Cockett
- Big Data Institute, Nuffield Department of Medicine, University of Oxford, Oxford, UK
- Wellcome Centre for Human Genetics, Nuffield Department of Medicine, NIHR Biomedical Research Centre, University of Oxford, Oxford, UK
| | - Helen R Fryer
- Big Data Institute, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Laura Thomson
- Big Data Institute, Nuffield Department of Medicine, University of Oxford, Oxford, UK
- Pandemic Science Institute, University of Oxford, Oxford, UK
| | - Anel Nurtay
- Big Data Institute, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Steven A Kemp
- Big Data Institute, Nuffield Department of Medicine, University of Oxford, Oxford, UK
- Department of Biology, University of Oxford, Oxford, UK
- Pandemic Science Institute, University of Oxford, Oxford, UK
| | - Luca Ferretti
- Big Data Institute, Nuffield Department of Medicine, University of Oxford, Oxford, UK
- Pandemic Science Institute, University of Oxford, Oxford, UK
| | - David Buck
- Wellcome Centre for Human Genetics, Nuffield Department of Medicine, NIHR Biomedical Research Centre, University of Oxford, Oxford, UK
| | - Angie Green
- Wellcome Centre for Human Genetics, Nuffield Department of Medicine, NIHR Biomedical Research Centre, University of Oxford, Oxford, UK
| | - Amy Trebes
- Wellcome Centre for Human Genetics, Nuffield Department of Medicine, NIHR Biomedical Research Centre, University of Oxford, Oxford, UK
| | - Paolo Piazza
- Wellcome Centre for Human Genetics, Nuffield Department of Medicine, NIHR Biomedical Research Centre, University of Oxford, Oxford, UK
| | - Lorne J Lonie
- Wellcome Centre for Human Genetics, Nuffield Department of Medicine, NIHR Biomedical Research Centre, University of Oxford, Oxford, UK
| | | | | | - Darren L Smith
- The Hub for Biotechnology in the Built Environment, Department of Applied Sciences, Faculty of Health and Life Sciences, Northumbria University, Newcastle upon Tyne, UK
- Department of Applied Sciences, Faculty of Health and Life Sciences, Northumbria University, Newcastle upon Tyne, UK
| | - Matthew Bashton
- The Hub for Biotechnology in the Built Environment, Department of Applied Sciences, Faculty of Health and Life Sciences, Northumbria University, Newcastle upon Tyne, UK
- Department of Applied Sciences, Faculty of Health and Life Sciences, Northumbria University, Newcastle upon Tyne, UK
| | - Andrew Nelson
- Department of Applied Sciences, Faculty of Health and Life Sciences, Northumbria University, Newcastle upon Tyne, UK
| | - Matthew Crown
- The Hub for Biotechnology in the Built Environment, Department of Applied Sciences, Faculty of Health and Life Sciences, Northumbria University, Newcastle upon Tyne, UK
- Department of Applied Sciences, Faculty of Health and Life Sciences, Northumbria University, Newcastle upon Tyne, UK
| | - Clare McCann
- Department of Applied Sciences, Faculty of Health and Life Sciences, Northumbria University, Newcastle upon Tyne, UK
| | - Gregory R Young
- The Hub for Biotechnology in the Built Environment, Department of Applied Sciences, Faculty of Health and Life Sciences, Northumbria University, Newcastle upon Tyne, UK
- Department of Applied Sciences, Faculty of Health and Life Sciences, Northumbria University, Newcastle upon Tyne, UK
| | - Rui Andre Nunes Dos Santos
- Department of Applied Sciences, Faculty of Health and Life Sciences, Northumbria University, Newcastle upon Tyne, UK
| | - Zack Richards
- Department of Applied Sciences, Faculty of Health and Life Sciences, Northumbria University, Newcastle upon Tyne, UK
| | - Mohammad Adnan Tariq
- Department of Applied Sciences, Faculty of Health and Life Sciences, Northumbria University, Newcastle upon Tyne, UK
| | | | | | - Christophe Fraser
- Big Data Institute, Nuffield Department of Medicine, University of Oxford, Oxford, UK
- Pandemic Science Institute, University of Oxford, Oxford, UK
- Wellcome Centre for Human Genetics, Nuffield Department of Medicine, NIHR Biomedical Research Centre, University of Oxford, Oxford, UK
- Wellcome Sanger Institute, Cambridge, UK
| | - David Bonsall
- Big Data Institute, Nuffield Department of Medicine, University of Oxford, Oxford, UK
- Pandemic Science Institute, University of Oxford, Oxford, UK
- Wellcome Centre for Human Genetics, Nuffield Department of Medicine, NIHR Biomedical Research Centre, University of Oxford, Oxford, UK
- Oxford University Hospitals NHS Foundation Trust, John Radcliffe Hospital, Headington, Oxford, UK
| | - Ann Sarah Walker
- Nuffield Department of Medicine, University of Oxford, Oxford, UK
- The National Institute for Health Research Health Protection Research Unit in Healthcare Associated Infections and Antimicrobial Resistance at the University of Oxford, Oxford, UK
- The National Institute for Health Research Oxford Biomedical Research Centre, University of Oxford, Oxford, UK
- MRC Clinical Trials Unit at UCL, UCL, London, UK
| | - Katrina Lythgoe
- Big Data Institute, Nuffield Department of Medicine, University of Oxford, Oxford, UK.
- Department of Biology, University of Oxford, Oxford, UK.
- Pandemic Science Institute, University of Oxford, Oxford, UK.
| |
Collapse
|
154
|
Titze-de-Almeida R, Araújo Lacerda PH, de Oliveira EP, de Oliveira MEF, Vianna YSS, Costa AM, Pereira Dos Santos E, Guérard LMC, Ferreira MADM, Rodrigues Dos Santos IC, Gonçalves JDDS, Ginani Ferreira G, Souza Titze-de-Almeida S, Brandão PRDP, Eri Shimizu H, Silva APB, Delgado-Rodrigues RN. Sleep and memory complaints in long COVID: an insight into clustered psychological phenotypes. PeerJ 2024; 12:e16669. [PMID: 38313024 PMCID: PMC10836207 DOI: 10.7717/peerj.16669] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2023] [Accepted: 11/22/2023] [Indexed: 02/06/2024] Open
Abstract
This study evaluated clinical features of individuals with long COVID (5-8 months after diagnosis) who reported sleep and memory problems (62 cases) compared to those without (52 controls). Both groups had a similar mean age (41 vs. 39 years). Around 86% of the participants were non-hospitalized at the time of infection, and none of them were vaccinated at that point. Subsequently, both cases and controls received the vaccine; however, the vaccination rates differed significantly between the groups (30.7% vs. 51.0%). Cases and controls had similar rates of symptoms at acute COVID phase. However, cases were more likely to experience coryza, dyspnea, headache, and nausea/vomiting during long COVID. Regarding new-onset symptoms in long COVID, 12.9% of cases had dyspnea, and 14.5% experienced nausea/vomiting, whereas in the control group there were only 1.9% and 0.0%, respectively. Cases also had a significantly higher prevalence of persistent headache (22.6% vs. 7.7%), and dyspnea (12.9% vs. 0.0). In addition, cases also showed an increased rate of mental health complaints: disability in daily activities (45.2% vs. 9.6%; P < 0.001); concentration/sustained attention difficulties (74.2% vs. 9.6%; P < 0.001); anxiety-Generalized Anxiety Disorder 2-item scale (GAD-2) ≥ 3 (66.1% vs. 34.6%; P = 0.0013); and "post-COVID sadness" (82.3% vs. 40.4%; P < 0.001). We observed a significant correlation between sadness and anxiety in cases, which was not observed in controls (P=0.0212; Spearman correlation test). Furthermore, the frequency of concomitant sadness and anxiety was markedly higher in cases compared to controls (59.7% vs. 19.2%) (P < 0.0001; Mann-Whitney test). These findings highlight a noteworthy association between sadness and anxiety specifically in cases. In conclusion, our data identified concurrent psychological phenotypes in individuals experiencing sleep and memory disturbances during long COVID. This strengthens the existing evidence that SARS-CoV-2 causes widespread brain pathology with interconnected phenotypic clusters. This finding highlights the need for comprehensive medical attention to address these complex issues, as well as major investments in testing strategies capable of preventing the development of long COVID sequelae, such as vaccination.
Collapse
Affiliation(s)
- Ricardo Titze-de-Almeida
- Central Institute of Sciences, Research Center for Major Themes, University of Brasília, Brasília, DF, Brazil
- University of Brasília/FAV, Central Institute of Sciences, Technology for Gene Therapy Laboratory, Brasília, DF, Brazil
| | | | - Edson Pereira de Oliveira
- Central Institute of Sciences, Research Center for Major Themes, University of Brasília, Brasília, DF, Brazil
| | | | | | - Amanda Machado Costa
- Central Institute of Sciences, Research Center for Major Themes, University of Brasília, Brasília, DF, Brazil
| | - Eloísa Pereira Dos Santos
- Central Institute of Sciences, Research Center for Major Themes, University of Brasília, Brasília, DF, Brazil
| | - Louise Marie Coelho Guérard
- Central Institute of Sciences, Research Center for Major Themes, University of Brasília, Brasília, DF, Brazil
| | | | | | | | - Gabriel Ginani Ferreira
- Central Institute of Sciences, Research Center for Major Themes, University of Brasília, Brasília, DF, Brazil
- University of Brasília/FAV, Central Institute of Sciences, Technology for Gene Therapy Laboratory, Brasília, DF, Brazil
| | - Simoneide Souza Titze-de-Almeida
- Central Institute of Sciences, Research Center for Major Themes, University of Brasília, Brasília, DF, Brazil
- University of Brasília/FAV, Central Institute of Sciences, Technology for Gene Therapy Laboratory, Brasília, DF, Brazil
| | - Pedro Renato de Paula Brandão
- Central Institute of Sciences, Research Center for Major Themes, University of Brasília, Brasília, DF, Brazil
- Sírio-Libanês Hospital, Brasília, Brazil., Brasília, DF, Brazil
| | - Helena Eri Shimizu
- Department of Collective Health, Research Center for Major Themes, University of Brasília, Brasília, DF, Brazil
| | - Andrezza Paula Brito Silva
- Central Institute of Sciences, Research Center for Major Themes, University of Brasília, Brasília, DF, Brazil
| | | |
Collapse
|
155
|
Rose J, Hulscher N, McCullough PA. Determinants of COVID-19 vaccine-induced myocarditis. Ther Adv Drug Saf 2024; 15:20420986241226566. [PMID: 38293564 PMCID: PMC10823859 DOI: 10.1177/20420986241226566] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Accepted: 01/01/2024] [Indexed: 02/01/2024] Open
Abstract
BACKGROUND Following the roll-out of the Pfizer-BioNTech BNT162b2, Moderna mRNA-1273, and Janssen Ad26.COV2.S coronavirus disease 2019 (COVID-19) injections in the United States, millions of individuals have reported adverse events (AEs) using the vaccine adverse events reports system (VAERS). The objective of this analysis is to describe the myocarditis data in VAERS and the COVID-19 vaccines as potential determinants of myocarditis. METHODS We used VAERS data to examine the frequency of reporting myocarditis since the beginning of the mass vaccination campaign and compared this with historical values in VAERS and COVID-19 vaccine administration data from the Our World in Data database. We examined myocarditis reports in VAERS in the context of sex, age, and dose. Statistical analysis was done using the Student's t-test to determine statistically significant differences between ages among myocarditis adverse events (AEs) and the chi-square test to determine relationships between categorical variables with statistical significance. RESULTS We found the number of myocarditis reports in VAERS after COVID-19 vaccination in 2021 was 223 times higher than the average of all vaccines combined for the past 30 years. This represented a 2500% increase in the absolute number of reports in the first year of the campaign when comparing historical values prior to 2021. Demographic data revealed that myocarditis occurred most in youths (50%) and males (69%). A total of 76% of cases resulted in emergency care and hospitalization. Of the total myocarditis reports, 92 individuals died (3%). Myocarditis was more likely after dose 2 (p < 0.00001) and individuals less than 30 years of age were more likely than individuals older than 30 to acquire myocarditis (p < 0.00001). CONCLUSION COVID-19 vaccination is strongly associated with a serious adverse safety signal of myocarditis, particularly in children and young adults resulting in hospitalization and death. Further investigation into the underlying mechanisms of COVID-19 vaccine-induced myocarditis is imperative to create effective mitigation strategies and ensure the safety of COVID-19 vaccination programs across populations.
Collapse
Affiliation(s)
| | - Nicolas Hulscher
- University of Michigan School of Public Health, 1415 Washington Heights, Ann Arbor, MI 48109, USA
| | | |
Collapse
|
156
|
LaVergne SM, Dutt TS, McFann K, Baxter BA, Webb TL, Berry K, Tipton M, Stromberg S, Sullivan BM, Dunn J, Henao-Tamayo M, Ryan EP. Persistent CD8 + T cell proliferation and activation in COVID-19 adult survivors with post-acute sequelae: a longitudinal, observational cohort study of persistent symptoms and T cell markers. Front Immunol 2024; 14:1303971. [PMID: 38327763 PMCID: PMC10848319 DOI: 10.3389/fimmu.2023.1303971] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2023] [Accepted: 12/12/2023] [Indexed: 02/09/2024] Open
Abstract
Introduction Post-acute sequelae of COVID-19 affects the quality of life of many COVID-19 survivors, yet the etiology of post-acute sequelae of COVID-19 remains unknown. We aimed to determine if persistent inflammation and ongoing T-cell activation during convalescence were a contributing factor to the pathogenesis of post-acute sequelae of COVID-19. Methods We evaluated 67 individuals diagnosed with COVID-19 by nasopharyngeal polymerase chain reaction for persistent symptoms during convalescence at separate time points occurring up to 180 days post-diagnosis. Fifty-two of these individuals were evaluated longitudinally. We obtained whole blood samples at each study visit, isolated peripheral blood mononuclear cells, and stained for multiple T cell activation markers for flow cytometry analysis. The activation states of participants' CD4+ and CD8+ T-cells were next analyzed for each of the persistent symptoms. Results Overall, we found that participants with persistent symptoms had significantly higher levels of inflammation at multiple time points during convalescence when compared to those who fully recovered from COVID-19. Participants with persistent dyspnea, forgetfulness, confusion, and chest pain had significantly higher levels of proliferating effector T-cells (CD8+Ki67+), and those with chest pain, joint pain, difficulty concentrating, and forgetfulness had higher levels of regulatory T-cells (CD4+CD25+). Additionally, those with dyspnea had significantly higher levels of CD8+CD38+, CD8+ Granzyme B+, and CD8+IL10+ cells. A retrospective comparison of acute phase inflammatory markers in adults with and without post-acute sequelae of COVID-19 showed that CD8+Ki67+ cells were significantly higher at the time of acute illness (up to 14 days post-diagnosis) in those who developed persistent dyspnea. Discussion These findings suggest continued CD8+ T-cell activation following SARS-CoV-2 infection in adults experiencing post-acute sequelae of COVID-19 and that the increase in T regulatory cells for a subset of these patients represents the ongoing attempt by the host to reduce inflammation.
Collapse
Affiliation(s)
- Stephanie M. LaVergne
- Department of Environmental Radiological and Health Sciences, Colorado State University, Fort Collins, CO, United States
| | - Taru S. Dutt
- Department of Microbiology, Immunology and Pathology, Colorado State University, Fort Collins, CO, United States
| | - Kim McFann
- University of Colorado Health, Medical Center of the Rockies, Loveland, CO, United States
| | - Bridget A. Baxter
- Department of Environmental Radiological and Health Sciences, Colorado State University, Fort Collins, CO, United States
| | - Tracy L. Webb
- Department of Clinical Sciences, Colorado State University, Fort Collins, CO, United States
| | - Kailey Berry
- Department of Molecular, Cellular, and Integrative Neurosciences, Colorado State University, Fort Collins, CO, United States
| | - Maddy Tipton
- Department of Environmental Radiological and Health Sciences, Colorado State University, Fort Collins, CO, United States
| | - Sophia Stromberg
- Department of Food Science and Human Nutrition, Colorado State University, Fort Collins, CO, United States
| | - Brian M. Sullivan
- Department of Immunology and Microbiology, Scripps Research, La Jolla, CA, United States
| | - Julie Dunn
- University of Colorado Health, Medical Center of the Rockies, Loveland, CO, United States
| | - Marcela Henao-Tamayo
- Department of Microbiology, Immunology and Pathology, Colorado State University, Fort Collins, CO, United States
| | - Elizabeth P. Ryan
- Department of Environmental Radiological and Health Sciences, Colorado State University, Fort Collins, CO, United States
| |
Collapse
|
157
|
Álvarez-Santacruz C, Tyrkalska SD, Candel S. The Microbiota in Long COVID. Int J Mol Sci 2024; 25:1330. [PMID: 38279329 PMCID: PMC10816132 DOI: 10.3390/ijms25021330] [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] [Subscribe] [Scholar Register] [Received: 12/11/2023] [Revised: 01/19/2024] [Accepted: 01/20/2024] [Indexed: 01/28/2024] Open
Abstract
Interest in the coronavirus disease 2019 (COVID-19) has progressively decreased lately, mainly due to the great effectivity of vaccines. Furthermore, no new severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) variants able to circumvent the protection of these vaccines, while presenting high transmissibility and/or lethality, have appeared. However, long COVID has emerged as a huge threat to human health and economy globally. The human microbiota plays an important role in health and disease, participating in the modulation of innate and adaptive immune responses. Thus, multiple studies have found that the nasopharyngeal microbiota is altered in COVID-19 patients, with these changes associated with the onset and/or severity of the disease. Nevertheless, although dysbiosis has also been reported in long COVID patients, mainly in the gut, little is known about the possible involvement of the microbiota in the development of this disease. Therefore, in this work, we aim to fill this gap in the knowledge by discussing and comparing the most relevant studies that have been published in this field up to this point. Hence, we discuss that the relevance of long COVID has probably been underestimated, and that the available data suggest that the microbiota could be playing a pivotal role on the pathogenesis of the disease. Further research to elucidate the involvement of the microbiota in long COVID will be essential to explore new therapeutic strategies based on manipulation of the microbiota.
Collapse
Affiliation(s)
| | - Sylwia D. Tyrkalska
- Departamento de Biología Celular e Histología, Facultad de Biología, Universidad de Murcia, 30100 Murcia, Spain;
- Instituto Murciano de Investigación Biosanitaria (IMIB)-Pascual Parrilla, 30120 Murcia, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Sergio Candel
- Departamento de Biología Celular e Histología, Facultad de Biología, Universidad de Murcia, 30100 Murcia, Spain;
- Instituto Murciano de Investigación Biosanitaria (IMIB)-Pascual Parrilla, 30120 Murcia, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Instituto de Salud Carlos III, 28029 Madrid, Spain
| |
Collapse
|
158
|
Villacampa A, Alfaro E, Morales C, Díaz-García E, López-Fernández C, Bartha JL, López-Sánchez F, Lorenzo Ó, Moncada S, Sánchez-Ferrer CF, García-Río F, Cubillos-Zapata C, Peiró C. SARS-CoV-2 S protein activates NLRP3 inflammasome and deregulates coagulation factors in endothelial and immune cells. Cell Commun Signal 2024; 22:38. [PMID: 38225643 PMCID: PMC10788971 DOI: 10.1186/s12964-023-01397-6] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2023] [Accepted: 11/12/2023] [Indexed: 01/17/2024] Open
Abstract
BACKGROUND Hyperinflammation, hypercoagulation and endothelial injury are major findings in acute and post-COVID-19. The SARS-CoV-2 S protein has been detected as an isolated element in human tissues reservoirs and is the main product of mRNA COVID-19 vaccines. We investigated whether the S protein alone triggers pro-inflammatory and pro-coagulant responses in primary cultures of two cell types deeply affected by SARS-CoV-2, such are monocytes and endothelial cells. METHODS In human umbilical vein endothelial cells (HUVEC) and monocytes, the components of NF-κB and the NLRP3 inflammasome system, as well as coagulation regulators, were assessed by qRT-PCR, Western blot, flow cytometry, or indirect immunofluorescence. RESULTS S protein activated NF-κB, promoted pro-inflammatory cytokines release, and triggered the priming and activation of the NLRP3 inflammasome system resulting in mature IL-1β formation in both cell types. This was paralleled by enhanced production of coagulation factors such as von Willebrand factor (vWF), factor VIII or tissue factor, that was mediated, at least in part, by IL-1β. Additionally, S protein failed to enhance ADAMTS-13 levels to counteract the pro-coagulant activity of vWF multimers. Monocytes and HUVEC barely expressed angiotensin-converting enzyme-2. Pharmacological approaches and gene silencing showed that TLR4 receptors mediated the effects of S protein in monocytes, but not in HUVEC. CONCLUSION S protein behaves both as a pro-inflammatory and pro-coagulant stimulus in human monocytes and endothelial cells. Interfering with the receptors or signaling pathways evoked by the S protein may help preventing immune and vascular complications driven by such an isolated viral element. Video Abstract.
Collapse
Affiliation(s)
- Alicia Villacampa
- Department of Pharmacology, School of Medicine, Universidad Autónoma de Madrid, Madrid, Spain
| | - Enrique Alfaro
- Respiratory Diseases Group, Respiratory Service, La Paz University Hospital, IdiPAZ, Madrid, Spain
- Biomedical Research Networking Center on Respiratory Diseases (CIBERES), Madrid, Spain
| | - Cristina Morales
- Department of Pharmacology, School of Medicine, Universidad Autónoma de Madrid, Madrid, Spain
| | - Elena Díaz-García
- Respiratory Diseases Group, Respiratory Service, La Paz University Hospital, IdiPAZ, Madrid, Spain
- Biomedical Research Networking Center on Respiratory Diseases (CIBERES), Madrid, Spain
| | - Cristina López-Fernández
- Respiratory Diseases Group, Respiratory Service, La Paz University Hospital, IdiPAZ, Madrid, Spain
| | - José Luis Bartha
- Department of Obstetrics and Gynecology, School of Medicine, Universidad Autónoma de Madrid, Madrid, Spain
- Gynecology and Obstetrics Service, La Paz University Hospital, Madrid, Spain
| | | | - Óscar Lorenzo
- Laboratory of Diabetes and Vascular pathology, IIS-Fundación Jiménez Díaz, Madrid, Spain
- Biomedical Research Networking Centre on Diabetes and Associated Metabolic Disorders (CIBERDEM), Madrid, Spain
- Department of Medicine, School of Medicine, Universidad Autónoma de Madrid, Madrid, Spain
| | - Salvador Moncada
- Department of Pharmacology, School of Medicine, Universidad Autónoma de Madrid, Madrid, Spain
| | - Carlos F Sánchez-Ferrer
- Department of Pharmacology, School of Medicine, Universidad Autónoma de Madrid, Madrid, Spain
- Vascular Pharmacology and Metabolism (FARMAVASM) group, IdiPAZ, Madrid, Spain
| | - Francisco García-Río
- Respiratory Diseases Group, Respiratory Service, La Paz University Hospital, IdiPAZ, Madrid, Spain
- Biomedical Research Networking Center on Respiratory Diseases (CIBERES), Madrid, Spain
- Department of Medicine, School of Medicine, Universidad Autónoma de Madrid, Madrid, Spain
| | - Carolina Cubillos-Zapata
- Respiratory Diseases Group, Respiratory Service, La Paz University Hospital, IdiPAZ, Madrid, Spain.
- Biomedical Research Networking Center on Respiratory Diseases (CIBERES), Madrid, Spain.
| | - Concepción Peiró
- Department of Pharmacology, School of Medicine, Universidad Autónoma de Madrid, Madrid, Spain.
- Vascular Pharmacology and Metabolism (FARMAVASM) group, IdiPAZ, Madrid, Spain.
| |
Collapse
|
159
|
Grady CB, Bhattacharjee B, Silva J, Jaycox J, Lee LW, Monteiro VS, Sawano M, Massey D, Caraballo C, Gehlhausen JR, Tabachnikova A, Mao T, Lucas C, Peña-Hernandez MA, Xu L, Tzeng TJ, Takahashi T, Herrin J, Güthe DB, Akrami A, Assaf G, Davis H, Harris K, McCorkell L, Schulz WL, Grffin D, Wei H, Ring AM, Guan L, Cruz CD, Iwasaki A, Krumholz HM. Impact of COVID-19 vaccination on symptoms and immune phenotypes in vaccine-naïve individuals with Long COVID. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.01.11.24300929. [PMID: 38260484 PMCID: PMC10802754 DOI: 10.1101/2024.01.11.24300929] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/24/2024]
Abstract
Background Long COVID contributes to the global burden of disease. Proposed root cause hypotheses include the persistence of SARS-CoV-2 viral reservoir, autoimmunity, and reactivation of latent herpesviruses. Patients have reported various changes in Long COVID symptoms after COVID-19 vaccinations, leaving uncertainty about whether vaccine-induced immune responses may alleviate or worsen disease pathology. Methods In this prospective study, we evaluated changes in symptoms and immune responses after COVID-19 vaccination in 16 vaccine-naïve individuals with Long COVID. Surveys were administered before vaccination and then at 2, 6, and 12 weeks after receiving the first vaccine dose of the primary series. Simultaneously, SARS-CoV-2-reactive TCR enrichment, SARS-CoV-2-specific antibody responses, antibody responses to other viral and self-antigens, and circulating cytokines were quantified before vaccination and at 6 and 12 weeks after vaccination. Results Self-report at 12 weeks post-vaccination indicated 10 out of 16 participants had improved health, 3 had no change, 1 had worse health, and 2 reported marginal changes. Significant elevation in SARS-CoV-2-specific TCRs and Spike protein-specific IgG were observed 6 and 12 weeks after vaccination. No changes in reactivities were observed against herpes viruses and self-antigens. Within this dataset, higher baseline sIL-6R was associated with symptom improvement, and the two top features associated with non-improvement were high IFN-β and CNTF, among soluble analytes. Conclusions Our study showed that in this small sample, vaccination improved the health or resulted in no change to the health of most participants, though few experienced worsening. Vaccination was associated with increased SARS-CoV-2 Spike protein-specific IgG and T cell expansion in most individuals with Long COVID. Symptom improvement was observed in those with baseline elevated sIL-6R, while elevated interferon and neuropeptide levels were associated with a lack of improvement.
Collapse
Affiliation(s)
- Connor B Grady
- Department of Biostatistics, Epidemiology, and Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, Massachusetts
| | - Bornali Bhattacharjee
- Department of Immunobiology, Yale University School of Medicine, New Haven, Connecticut
- Department of Microbial Pathogenesis, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Julio Silva
- Department of Immunobiology, Yale University School of Medicine, New Haven, Connecticut
| | - Jillian Jaycox
- Department of Immunobiology, Yale University School of Medicine, New Haven, Connecticut
| | | | - Valter Silva Monteiro
- Department of Immunobiology, Yale University School of Medicine, New Haven, Connecticut
| | - Mitsuaki Sawano
- Center for Outcomes Research and Evaluation, Yale New Haven Hospital, New Haven, Connecticut
| | - Daisy Massey
- Center for Outcomes Research and Evaluation, Yale New Haven Hospital, New Haven, Connecticut
| | - César Caraballo
- Center for Outcomes Research and Evaluation, Yale New Haven Hospital, New Haven, Connecticut
- Department of Internal Medicine, Yale School of Medicine, New Haven, Connecticut
| | - Jeff R Gehlhausen
- Department of Immunobiology, Yale University School of Medicine, New Haven, Connecticut
| | | | - Tianyang Mao
- Department of Immunobiology, Yale University School of Medicine, New Haven, Connecticut
| | - Carolina Lucas
- Department of Immunobiology, Yale University School of Medicine, New Haven, Connecticut
| | - Mario A Peña-Hernandez
- Department of Immunobiology, Yale University School of Medicine, New Haven, Connecticut
- Department of Microbial Pathogenesis, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Lan Xu
- Department of Immunobiology, Yale University School of Medicine, New Haven, Connecticut
| | - Tiffany J Tzeng
- Department of Immunobiology, Yale University School of Medicine, New Haven, Connecticut
| | - Takehiro Takahashi
- Department of Immunobiology, Yale University School of Medicine, New Haven, Connecticut
| | - Jeph Herrin
- Department of Internal Medicine, Yale School of Medicine, New Haven, Connecticut
| | | | - Athena Akrami
- Sainsbury Wellcome Centre, University College London, London, UK
- Patient-Led Research Collaborative
| | | | | | | | | | - Wade L Schulz
- Center for Infection and Immunity, Yale School of Medicine, New Haven, Connecticut
- Center for Outcomes Research and Evaluation, Yale New Haven Hospital, New Haven, Connecticut
- Department of Laboratory Medicine, Yale University School of Medicine, New Haven, Connecticut
| | - Daniel Grffin
- Department of Medicine, Columbia University Vagelos College of Physicians and Surgeons, New York City, New York
| | | | - Aaron M Ring
- Department of Immunobiology, Yale University School of Medicine, New Haven, Connecticut
| | - Leying Guan
- Center for Infection and Immunity, Yale School of Medicine, New Haven, Connecticut
- Department of Biostatistics, Yale School of Public Health, New Haven, Connecticut
| | - Charles Dela Cruz
- Department of Microbial Pathogenesis, Yale University School of Medicine, New Haven, Connecticut, USA
- Center for Infection and Immunity, Yale School of Medicine, New Haven, Connecticut
- Department of Medicine, Section of Pulmonary and Critical Care Medicine, Yale University School of Medicine, New Haven, Connecticut
| | - Akiko Iwasaki
- Department of Immunobiology, Yale University School of Medicine, New Haven, Connecticut
- Center for Infection and Immunity, Yale School of Medicine, New Haven, Connecticut
- Howard Hughes Medical Institute, Chevy Chase, Maryland
| | - Harlan M Krumholz
- Center for Infection and Immunity, Yale School of Medicine, New Haven, Connecticut
- Center for Outcomes Research and Evaluation, Yale New Haven Hospital, New Haven, Connecticut
- Department of Health Policy and Management, Yale School of Public Health, New Haven, Connecticut
- Section of Cardiovascular Medicine, Department of Internal Medicine, Yale School of Medicine, New Haven, Connecticut
| |
Collapse
|
160
|
Matías-Pérez D, Antonio-Estrada C, Guerra-Martínez A, García-Melo KS, Hernández-Bautista E, García-Montalvo IA. Relationship of quercetin intake and oxidative stress in persistent COVID. Front Nutr 2024; 10:1278039. [PMID: 38260057 PMCID: PMC10800910 DOI: 10.3389/fnut.2023.1278039] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2023] [Accepted: 12/26/2023] [Indexed: 01/24/2024] Open
Affiliation(s)
- Diana Matías-Pérez
- Division of Graduate Studies and Research, Tecnológico Nacional de México/Instituto Tecnológico de Oaxaca, Oaxaca, Mexico
| | - Carolina Antonio-Estrada
- Division of Graduate Studies and Research, Tecnológico Nacional de México/Instituto Tecnológico de Oaxaca, Oaxaca, Mexico
| | - Araceli Guerra-Martínez
- Division of Graduate Studies and Research, Tecnológico Nacional de México/Instituto Tecnológico de Oaxaca, Oaxaca, Mexico
| | - Karen Seydel García-Melo
- Division of Graduate Studies and Research, Tecnológico Nacional de México/Instituto Tecnológico de Oaxaca, Oaxaca, Mexico
| | - Emilio Hernández-Bautista
- Department of Chemical Engineering, Tecnológico Nacional de México/Instituto Tecnológico de Oaxaca, Oaxaca, Mexico
| | - Iván Antonio García-Montalvo
- Division of Graduate Studies and Research, Tecnológico Nacional de México/Instituto Tecnológico de Oaxaca, Oaxaca, Mexico
| |
Collapse
|
161
|
Appelman B, Charlton BT, Goulding RP, Kerkhoff TJ, Breedveld EA, Noort W, Offringa C, Bloemers FW, van Weeghel M, Schomakers BV, Coelho P, Posthuma JJ, Aronica E, Joost Wiersinga W, van Vugt M, Wüst RCI. Muscle abnormalities worsen after post-exertional malaise in long COVID. Nat Commun 2024; 15:17. [PMID: 38177128 PMCID: PMC10766651 DOI: 10.1038/s41467-023-44432-3] [Citation(s) in RCA: 126] [Impact Index Per Article: 126.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2023] [Accepted: 12/13/2023] [Indexed: 01/06/2024] Open
Abstract
A subgroup of patients infected with SARS-CoV-2 remain symptomatic over three months after infection. A distinctive symptom of patients with long COVID is post-exertional malaise, which is associated with a worsening of fatigue- and pain-related symptoms after acute mental or physical exercise, but its underlying pathophysiology is unclear. With this longitudinal case-control study (NCT05225688), we provide new insights into the pathophysiology of post-exertional malaise in patients with long COVID. We show that skeletal muscle structure is associated with a lower exercise capacity in patients, and local and systemic metabolic disturbances, severe exercise-induced myopathy and tissue infiltration of amyloid-containing deposits in skeletal muscles of patients with long COVID worsen after induction of post-exertional malaise. This study highlights novel pathways that help to understand the pathophysiology of post-exertional malaise in patients suffering from long COVID and other post-infectious diseases.
Collapse
Affiliation(s)
- Brent Appelman
- Amsterdam UMC location University of Amsterdam, Center for Experimental and Molecular Medicine, Meibergdreef 9, Amsterdam, the Netherlands
- Amsterdam Institute for Infection and Immunity, Infectious diseases, Amsterdam, the Netherlands
| | - Braeden T Charlton
- Department of Human Movement Sciences, Faculty of Behavioral and Movement Sciences, Vrije Universiteit Amsterdam, Amsterdam, the Netherlands
- Amsterdam Movement Sciences, Amsterdam, the Netherlands
| | - Richie P Goulding
- Department of Human Movement Sciences, Faculty of Behavioral and Movement Sciences, Vrije Universiteit Amsterdam, Amsterdam, the Netherlands
- Amsterdam Movement Sciences, Amsterdam, the Netherlands
| | - Tom J Kerkhoff
- Department of Human Movement Sciences, Faculty of Behavioral and Movement Sciences, Vrije Universiteit Amsterdam, Amsterdam, the Netherlands
- Amsterdam Movement Sciences, Amsterdam, the Netherlands
- Department of Physiology, Amsterdam UMC location Vrije Universiteit Amsterdam, De Boelelaan 1117, Amsterdam, the Netherlands
- Amsterdam Cardiovascular Sciences, Amsterdam, the Netherlands
| | - Ellen A Breedveld
- Department of Human Movement Sciences, Faculty of Behavioral and Movement Sciences, Vrije Universiteit Amsterdam, Amsterdam, the Netherlands
- Amsterdam Movement Sciences, Amsterdam, the Netherlands
| | - Wendy Noort
- Department of Human Movement Sciences, Faculty of Behavioral and Movement Sciences, Vrije Universiteit Amsterdam, Amsterdam, the Netherlands
- Amsterdam Movement Sciences, Amsterdam, the Netherlands
| | - Carla Offringa
- Department of Human Movement Sciences, Faculty of Behavioral and Movement Sciences, Vrije Universiteit Amsterdam, Amsterdam, the Netherlands
- Amsterdam Movement Sciences, Amsterdam, the Netherlands
| | - Frank W Bloemers
- Amsterdam Movement Sciences, Amsterdam, the Netherlands
- Department of Trauma Surgery, Amsterdam UMC location University of Amsterdam, Meibergdreef 9, Amsterdam, the Netherlands
| | - Michel van Weeghel
- Laboratory Genetic Metabolic Diseases, Core Facility Metabolomics, Amsterdam UMC location University of Amsterdam, Meibergdreef 9, Amsterdam, the Netherlands
| | - Bauke V Schomakers
- Laboratory Genetic Metabolic Diseases, Core Facility Metabolomics, Amsterdam UMC location University of Amsterdam, Meibergdreef 9, Amsterdam, the Netherlands
| | - Pedro Coelho
- Serviço de Neurologia, Departamento de Neurociências e Saúde Mental, Hospital de Santa Maria, CHULN, Lisbon, Portugal
- Faculdade de Medicina, Centro de Estudos Egas Moniz, University of Lisbon, Lisbon, Portugal
- Department of (Neuro)pathology, Amsterdam Neuroscience, Amsterdam UMC location University of Amsterdam, Meibergdreef 9, Amsterdam, the Netherlands
| | - Jelle J Posthuma
- Department of Trauma Surgery, Amsterdam UMC location University of Amsterdam, Meibergdreef 9, Amsterdam, the Netherlands
- Flevoziekenhuis, Division of Surgery, Hospitaalweg 1, Almere, the Netherlands
| | - Eleonora Aronica
- Department of (Neuro)pathology, Amsterdam Neuroscience, Amsterdam UMC location University of Amsterdam, Meibergdreef 9, Amsterdam, the Netherlands
| | - W Joost Wiersinga
- Amsterdam UMC location University of Amsterdam, Center for Experimental and Molecular Medicine, Meibergdreef 9, Amsterdam, the Netherlands
- Amsterdam Institute for Infection and Immunity, Infectious diseases, Amsterdam, the Netherlands
- Division of Infectious Diseases, Department of Internal Medicine, Amsterdam UMC location University of Amsterdam, Meibergdreef 9, Amsterdam, the Netherlands
| | - Michèle van Vugt
- Amsterdam Institute for Infection and Immunity, Infectious diseases, Amsterdam, the Netherlands.
- Division of Infectious Diseases, Tropical Medicine, Department of Medicine, Amsterdam UMC location University of Amsterdam, Meibergdreef 9, Amsterdam, the Netherlands.
| | - Rob C I Wüst
- Department of Human Movement Sciences, Faculty of Behavioral and Movement Sciences, Vrije Universiteit Amsterdam, Amsterdam, the Netherlands.
- Amsterdam Movement Sciences, Amsterdam, the Netherlands.
| |
Collapse
|
162
|
Howard J, Cloeren M, Vanichkachorn G. Long COVID and Occupational Medicine Practice. J Occup Environ Med 2024; 66:1-5. [PMID: 37696788 DOI: 10.1097/jom.0000000000002961] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/13/2023]
Affiliation(s)
- John Howard
- From the National Institute for Occupational Safety and Health, Centers for Disease Control and Prevention, US Department of Health and Human Services, Washington, District of Columbia (J.H.); Division of Occupational and Environmental Medicine, University of Maryland School of Medicine, Baltimore, Maryland (M.C.); and Occupational Medicine, Mayo Clinic, Rochester, Minnesota (G.V.)
| | | | | |
Collapse
|
163
|
El-Baky NA, Amara AA, Uversky VN, Redwan EM. Intrinsic factors behind long COVID: III. Persistence of SARS-CoV-2 and its components. J Cell Biochem 2024; 125:22-44. [PMID: 38098317 DOI: 10.1002/jcb.30514] [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: 09/04/2023] [Revised: 11/27/2023] [Accepted: 12/07/2023] [Indexed: 01/16/2024]
Abstract
Considerable research has been done in investigating SARS-CoV-2 infection, its characteristics, and host immune response. However, debate is still ongoing over the emergence of post-acute sequelae of SARS-CoV-2 infection (PASC). A multitude of long-lasting symptoms have been reported several weeks after the primary acute SARS-CoV-2 infection that resemble several other viral infections. Thousands of research articles have described various post-COVID-19 conditions. Yet, the evidence around these ongoing health problems, the reasons behind them, and their molecular underpinnings are scarce. These persistent symptoms are also known as long COVID-19. The persistence of SARS-CoV-2 and/or its components in host tissues can lead to long COVID. For example, the presence of viral nucleocapsid protein and RNA was detected in the skin, appendix, and breast tissues of some long COVID patients. The persistence of viral RNA was reported in multiple anatomic sites, including non-respiratory tissues such as the adrenal gland, ocular tissue, small intestine, lymph nodes, myocardium, and sciatic nerve. Distinctive viral spike sequence variants were also found in non-respiratory tissues. Interestingly, prolonged detection of viral subgenomic RNA was observed across all tissues, sometimes in multiple tissues of the same patient, which likely reflects recent but defective viral replication. Moreover, the persistence of SARS-CoV-2 RNA was noticed throughout the brain at autopsy, as late as 230 days following symptom onset among unvaccinated patients who died of severe infection. Here, we review the persistence of SARS-CoV-2 and its components as an intrinsic factor behind long COVID. We also highlight the immunological consequences of this viral persistence.
Collapse
Affiliation(s)
- Nawal Abd El-Baky
- Protein Research Department, Genetic Engineering and Biotechnology Research Institute (GEBRI), City of Scientific Research and Technological Applications (SRTA-City), New Borg El-Arab City, Egypt
| | - Amro A Amara
- Protein Research Department, Genetic Engineering and Biotechnology Research Institute (GEBRI), City of Scientific Research and Technological Applications (SRTA-City), New Borg El-Arab City, Egypt
| | - Vladimir N Uversky
- Department of Molecular Medicine, USF Health Byrd Alzheimer's Research Institute, Morsani College of Medicine, University of South Florida, Tampa, Florida, USA
| | - Elrashdy M Redwan
- Biological Sciences Department, Faculty of Science, King Abdulaziz University, Jeddah, Saudi Arabia
| |
Collapse
|
164
|
Yoon H, Li Y, Goldfeld KS, Cobb GF, Sturm-Reganato CL, Ostrosky-Zeichner L, Jayaweera DT, Philley JV, Desruisseaux MS, Keller MJ, Hochman JS, Pirofski LA, Ortigoza MB. COVID-19 Convalescent Plasma Therapy: Long-term Implications. Open Forum Infect Dis 2024; 11:ofad686. [PMID: 38269049 PMCID: PMC10807994 DOI: 10.1093/ofid/ofad686] [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: 10/27/2023] [Accepted: 12/27/2023] [Indexed: 01/26/2024] Open
Abstract
Background The long-term effect of coronavirus disease 2019 (COVID-19) acute treatments on postacute sequelae of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection (PASC) is unknown. The CONTAIN-Extend study explores the long-term impact of COVID-19 convalescent plasma (CCP) therapy on postacute sequelae of SARS-CoV-2 infection (PASC) symptoms and general health 18 months following hospitalization. Methods The CONTAIN-Extend study examined 281 participants from the original CONTAIN COVID-19 trial (CONTAIN-RCT, NCT04364737) at 18 months post-hospitalization for acute COVID-19. Symptom surveys, global health assessments, and biospecimen collection were performed from November 2021 to October 2022. Multivariable logistic and linear regression estimated associations between the randomization arms and self-reported symptoms and Patient-Reported Outcomes Measurement Information System (PROMIS) scores and adjusted for covariables, including age, sex, race/ethnicity, disease severity, and CONTAIN enrollment quarter and sites. Results There were no differences in symptoms or PROMIS scores between CCP and placebo (adjusted odds ratio [aOR] of general symptoms, 0.95; 95% CI, 0.54-1.67). However, females (aOR, 3.01; 95% CI, 1.73-5.34), those 45-64 years (aOR, 2.55; 95% CI, 1.14-6.23), and April-June 2020 enrollees (aOR, 2.39; 95% CI, 1.10-5.19) were more likely to report general symptoms and have poorer PROMIS physical health scores than their respective reference groups. Hispanic participants (difference, -3.05; 95% CI, -5.82 to -0.27) and Black participants (-4.48; 95% CI, -7.94 to -1.02) had poorer PROMIS physical health than White participants. Conclusions CCP demonstrated no lasting effect on PASC symptoms or overall health in comparison to the placebo. This study underscores the significance of demographic factors, including sex, age, and timing of acute infection, in influencing symptom reporting 18 months after acute hypoxic COVID-19 hospitalization.
Collapse
Affiliation(s)
- Hyunah Yoon
- Division of Infectious Diseases, Department of Medicine, Albert Einstein College of Medicine, Montefiore Medical Center, Bronx, New York, USA
| | - Yi Li
- Division of Biostatistics, Department of Population Health, NYU Grossman School of Medicine, New York, New York, USA
| | - Keith S Goldfeld
- Division of Biostatistics, Department of Population Health, NYU Grossman School of Medicine, New York, New York, USA
| | - Gia F Cobb
- Department of Medicine, NYU Grossman School of Medicine, New York, New York, USA
| | | | - Luis Ostrosky-Zeichner
- Division of Infectious Diseases, Department of Internal Medicine, The University of Texas Health Science Center at Houston, McGovern Medical School, Houston, Texas, USA
| | - Dushyantha T Jayaweera
- Division of Infectious Diseases, Department of Medicine, University of Miami Miller School of Medicine, Miami, Florida, USA
- Miami Clinical and Translational Science Institute, University of Miami Miller School of Medicine, Miami, Florida, USA
| | - Julie V Philley
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, The University of Texas Health Science Center at Tyler, UTHealth East Texas, Tyler, Texas, USA
| | - Mahalia S Desruisseaux
- Section of Infectious Diseases, Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Marla J Keller
- Division of Infectious Diseases, Department of Medicine, Albert Einstein College of Medicine, Montefiore Medical Center, Bronx, New York, USA
- Harold and Muriel Block Institute for Clinical and Translational Research, Albert Einstein College of Medicine and Montefiore Medical Center, Bronx, New York, USA
| | - Judith S Hochman
- Leon H. Charney Division of Cardiology, Department of Medicine, NYU Grossman School of Medicine, New York, New York, USA
| | - Liise-anne Pirofski
- Division of Infectious Diseases, Department of Medicine, Albert Einstein College of Medicine, Montefiore Medical Center, Bronx, New York, USA
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, New York, USA
| | - Mila B Ortigoza
- Division of Infectious Diseases, Department of Medicine, NYU Grossman School of Medicine, New York, New York, USA
- Department of Microbiology, NYU Grossman School of Medicine, New York, New York, USA
| |
Collapse
|
165
|
Zhang C, Hung CY, Hsu CG. Epidemiology, Symptoms and Pathophysiology of Long Covid Complications. JOURNAL OF CELLULAR IMMUNOLOGY 2024; 6:219-230. [PMID: 40276305 PMCID: PMC12021439 DOI: 10.33696/immunology.6.209] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 04/26/2025]
Abstract
Long COVID, or post-acute sequelae of SARS-CoV-2 infection, reports to affect a significant proportion of COVID-19 survivors, leading to persistent and multi-organ complications. This review examines the epidemiology, symptoms of long COVID complications, including cardiac, hematological, vascular, pulmonary, neuropsychiatric, renal, gastrointestinal, musculoskeletal, immune dysregulation, and dermatological issues. By synthesizing the latest research, this article provides a comprehensive overview of the prevalence and detailed pathophysiological mechanisms underlying these complications. The purpose of this review is to enhance the understanding of diverse and complex nature of long COVID and emphasize the need for ongoing research, seeking to support future studies for better management of long COVID.
Collapse
Affiliation(s)
- Chongyang Zhang
- Department of Pharmacology and Physiology, University of Rochester, School of Medicine and Dentistry, Rochester, NY 14642, USA
| | - Chiung-Yu Hung
- Department of Molecular Microbiology and Immunology, South Texas Center for Emerging Infectious Diseases, The University of Texas at San Antonio, San Antonio, TX 78249, USA
| | - Chia George Hsu
- Department of Kinesiology, The University of Texas at San Antonio, San Antonio, TX 78249, USA
| |
Collapse
|
166
|
Durstenfeld MS, Peluso MJ, Lin F, Peyser ND, Isasi C, Carton TW, Henrich TJ, Deeks SG, Olgin JE, Pletcher MJ, Beatty AL, Marcus GM, Hsue PY. Association of nirmatrelvir for acute SARS-CoV-2 infection with subsequent Long COVID symptoms in an observational cohort study. J Med Virol 2024; 96:e29333. [PMID: 38175151 PMCID: PMC10786003 DOI: 10.1002/jmv.29333] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2023] [Revised: 12/07/2023] [Accepted: 12/08/2023] [Indexed: 01/05/2024]
Abstract
Oral nirmatrelvir/ritonavir is approved as treatment for acute COVID-19, but the effect of treatment during acute infection on risk of Long COVID is unknown. We hypothesized that nirmatrelvir treatment during acute SARS-CoV-2 infection reduces risk of developing Long COVID and rebound after treatment is associated with Long COVID. We conducted an observational cohort study within the Covid Citizen Science (CCS) study, an online cohort study with over 100 000 participants. We included vaccinated, nonhospitalized, nonpregnant individuals who reported their first SARS-CoV-2 positive test March-August 2022. Oral nirmatrelvir/ritonavir treatment was ascertained during acute SARS-CoV-2 infection. Patient-reported Long COVID symptoms, symptom rebound and test-positivity rebound were asked on subsequent surveys at least 3 months after SARS-CoV-2 infection. A total of 4684 individuals met the eligibility criteria, of whom 988 (21.1%) were treated and 3696 (78.9%) were untreated; 353/988 (35.7%) treated and 1258/3696 (34.0%) untreated responded to the Long COVID survey (n = 1611). Among 1611 participants, median age was 55 years and 66% were female. At 5.4 ± 1.3 months after infection, nirmatrelvir treatment was not associated with subsequent Long COVID symptoms (odds ratio [OR]: 1.15; 95% confidence interval [CI]: 0.80-1.64; p = 0.45). Among 666 treated who answered rebound questions, rebound symptoms or test positivity were not associated with Long COVID symptoms (OR: 1.34; 95% CI: 0.74-2.41; p = 0.33). Within this cohort of vaccinated, nonhospitalized individuals, oral nirmatrelvir treatment during acute SARS-CoV-2 infection and rebound after nirmatrelvir treatment were not associated with Long COVID symptoms more than 90 days after infection.
Collapse
Affiliation(s)
- Matthew S. Durstenfeld
- Division of Cardiology at ZSFG and Department of Medicine, University of California, San Francisco (UCSF), USA
| | | | - Feng Lin
- Department of Epidemiology and Biostatistics, UCSF, USA
| | | | - Carmen Isasi
- Department of Epidemiology & Population Health, Albert Einstein College of Medicine
| | | | | | - Steven G. Deeks
- Division of HIV, Infectious Disease, & Global Medicine, UCSF, USA
| | | | | | - Alexis L. Beatty
- Department of Epidemiology and Biostatistics and Division of Cardiology, Department of Medicine, UCSF, USA
| | | | - Priscilla Y. Hsue
- Division of Cardiology at ZSFG and Department of Medicine, University of California, San Francisco (UCSF), USA
| |
Collapse
|
167
|
Scheppke KA, Pepe PE, Jui J, Crowe RP, Scheppke EK, Klimas NG, Marty AM. Remission of severe forms of long COVID following monoclonal antibody (MCA) infusions: A report of signal index cases and call for targeted research. Am J Emerg Med 2024; 75:122-127. [PMID: 37944296 DOI: 10.1016/j.ajem.2023.09.051] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Accepted: 09/29/2023] [Indexed: 11/12/2023] Open
Abstract
OBJECTIVE Long COVID has afflicted tens of millions globally leaving many previously-healthy persons severely and indefinitely debilitated. The objective here was to report cases of complete, rapid remission of severe forms of long COVID following certain monoclonal antibody (MCA) infusions and review the corresponding pathophysiological implications. DESIGN Case histories of the first three index events (among others) are presented. Unaware of others with similar remissions, each subject independently completed personal narratives and standardized surveys regarding demographics/occupation, past history, and the presence and respective severity grading of 33 signs/symptoms associated with long COVID, comparing the presence/severity of those symptoms during the pre-COVID, long-COVID, post-vaccination, and post-MCA phases. SETTING Patient interviews, e-mails and telephone conversations. SUBJECTS Three previously healthy, middle-aged, highly-functioning persons, two women and one man (ages 60, 43, and 63 years respectively) who, post-acute COVID-19 infection, developed chronic, unrelenting fatigue and cognitive impairment along with other severe, disabling symptoms. Each then independently reported incidental and unanticipated complete remissions within days of MCA treatment. INTERVENTIONS The casirivimab/imdevimab cocktail. MEASUREMENTS AND MAIN RESULTS Irrespective of sex, age, medical history, vaccination status, or illness duration (18, 8 and 5 months, respectively), each subject experienced the same complete remission of their persistent disabling disease within a week of MCA infusion. Each rapidly returned to normal health and previous lifestyles/occupations with normalized exercise tolerance, still sustained to date over two years later. CONCLUSIONS These index cases provide compelling clinical signals that MCA infusions may be capable of treating long COVID in certain cases, including those with severe debilitation. While the complete and sustained remissions observed here may only apply to long COVID resulting from pre-Delta variants and the specific MCA infused, the striking rapid and complete remissions observed in these cases also provide mechanistic implications for treating/managing other post-viral chronic conditions and long COVID from other variants. KEY POINTS
Collapse
Affiliation(s)
- Kenneth A Scheppke
- Florida Department of Health, 4052 Bald Cypress Way, Tallahassee, FL 32399, USA; Palm Beach County Fire Rescue, 405 Pike Road, West Palm Beach, FL 33411, USA; Broward Sheriff's Office, 2601 West Broward Boulevard, Ft Lauderdale, FL 33312, USA
| | - Paul E Pepe
- Palm Beach County Fire Rescue, 405 Pike Road, West Palm Beach, FL 33411, USA; Broward Sheriff's Office, 2601 West Broward Boulevard, Ft Lauderdale, FL 33312, USA; Broward Health Medical Center, 1600 S Andrews Ave, Fort Lauderdale, FL 33316, USA; Department of Management, Policy and Community Health, School of Public Health, University of Texas Health Sciences Center, 1200 Pressler St, Houston, TX 77030, USA; Dallas County EMS/Public Safety Agencies, Suite 500, 500 Elm St, Dallas, TX 75202, USA.
| | - Jonathan Jui
- Department of Emergency Medicine, Oregon Health & Sciences University, CDW-EM, 3181 SW Sam Jackson Park Road, Portland, OR 97239, USA
| | | | - Eric K Scheppke
- Edward Via College of Osteopathic Medicine-Auburn, 910 S Donahue Dr, Auburn, AL 36832, USA
| | - Nancy G Klimas
- Institute for Neuro-Immune Medicine, Nova Southeastern University, 7595 SW 33rd Street, Fourth Floor, Ft Lauderdale, FL 33314, USA; Miami Veterans Administration Medical Center, 1201 NW 16th St, Miami, FL 33125, USA
| | - Aileen M Marty
- Department of Translational Medicine, Florida International University, 885 SW 109th Ave, PG-5, Suite 1313, Miami, FL, 33199, USA
| |
Collapse
|
168
|
Ganesh R, Yadav S, Hurt RT, Mueller MR, Aakre CA, Gilman EA, Grach SL, Overgaard J, Snyder MR, Collins NM, Croghan IT, Badley AD, Razonable RR, Salonen BR. Pro Inflammatory Cytokines Profiles of Patients With Long COVID Differ Between Variant Epochs. J Prim Care Community Health 2024; 15:21501319241254751. [PMID: 38808863 PMCID: PMC11138192 DOI: 10.1177/21501319241254751] [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: 02/20/2024] [Revised: 04/22/2024] [Accepted: 04/24/2024] [Indexed: 05/30/2024] Open
Abstract
BACKGROUND Over 30% of patients with COVID-19 have persistent symptoms that last beyond 30 days and referred to as Long COVID. Long COVID has been associated with a persistent elevation in peripheral cytokines including interleukin-6, interleukin-1β, and tumor necrosis factor-α. This study reports cytokine profiles of patients in our clinic across SARS-COV-2 variant epochs. METHODS The clinical cytokine panel was analyzed in patients with Long COVID during periods that were stratified according to variant epoch. The 4 variant epochs were defined as: (1) wild-type through alpha, (2) alpha/beta/gamma, (3) delta, and (4) omicron variants. RESULTS A total of 390 patients had the clinical cytokine panel performed; the median age was 48 years (IQR 38-59) and 62% were female. Distribution by variant was wild-type and alpha, 50% (n = 196); alpha/beta/gamma, 7.9% (n = 31); delta, 18% (n = 72); and omicron, 23% (n = 91). Time to cytokine panel testing was significantly longer for the earlier epochs. Tumor necrosis factor-α (P < .001) and interleukin 1β (P < .001) were significantly more elevated in the earlier epochs (median of 558 days in wild-type through Alpha epoch vs 263 days in omicron epoch, P < .001)). Nucleocapsid antibodies were consistently detected across epochs. DISCUSSION When stratified by variant epoch, patients with early epoch Long COVID had persistently elevated peripheral pro-inflammatory cytokine levels when compared to later epoch Long COVID. Patients with Long COVID have similar clusters of symptoms across epochs, suggesting that the underlying pathology is independent of the peripheral cytokine signature.
Collapse
|
169
|
Perico L, Benigni A, Remuzzi G. SARS-CoV-2 and the spike protein in endotheliopathy. Trends Microbiol 2024; 32:53-67. [PMID: 37393180 PMCID: PMC10258582 DOI: 10.1016/j.tim.2023.06.004] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2023] [Revised: 06/05/2023] [Accepted: 06/08/2023] [Indexed: 07/03/2023]
Abstract
SARS-CoV-2, the causative agent of COVID-19, primarily affects the epithelial compartment in the upper and lower airways. There is evidence that the microvasculature in both the pulmonary and extrapulmonary systems is a major target of SARS-CoV-2. Consistent with this, vascular dysfunction and thrombosis are the most severe complications in COVID-19. The proinflammatory milieu triggered by the hyperactivation of the immune system by SARS-CoV-2 has been suggested to be the main trigger for endothelial dysfunction during COVID-19. More recently, a rapidly growing number of reports have indicated that SARS-CoV-2 can interact directly with endothelial cells through the spike protein, leading to multiple instances of endothelial dysfunction. Here, we describe all the available findings showing the direct effect of the SARS-CoV-2 spike protein on endothelial cells and offer mechanistic insights into the molecular basis of vascular dysfunction in severe COVID-19.
Collapse
Affiliation(s)
- Luca Perico
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Centro Anna Maria Astori, Bergamo, Italy.
| | - Ariela Benigni
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Centro Anna Maria Astori, Bergamo, Italy
| | - Giuseppe Remuzzi
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Centro Anna Maria Astori, Bergamo, Italy
| |
Collapse
|
170
|
Peluso MJ, Swank ZN, Goldberg SA, Lu S, Dalhuisen T, Borberg E, Senussi Y, Luna MA, Song CC, Clark A, Zamora A, Lew M, Viswanathan B, Huang B, Anglin K, Hoh R, Hsue PY, Durstenfeld MS, Spinelli MA, Glidden DV, Henrich TJ, Daniel Kelly J, Deeks SG, Walt DR, Martin JN. Plasma-based antigen persistence in the post-acute phase of SARS-CoV-2 infection. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2023:2023.10.24.23297114. [PMID: 37961239 PMCID: PMC10635183 DOI: 10.1101/2023.10.24.23297114] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/15/2023]
Abstract
BACKGROUND Persistent symptoms among some persons who develop COVID-19 has led to the hypothesis that SARS-CoV-2 may, in some form or location, persist for long periods following acute infection. Several studies have shown data in this regard but are limited by non-representative and small study populations, short duration since acute infection, and lack of a true-negative comparator group to assess assay specificity. METHODS We evaluated adults with RNA-confirmed COVID-19 at multiple time points following acute infection (pandemic-era participants) and adults with specimens collected prior to 2020 (pre-pandemic era). Using once-thawed plasma, we employed the Simoa® (Quanterix) single molecule array detection platform to measure SARS-CoV-2 spike, S1, and nucleocapsid antigens. RESULTS Compared to 250 pre-pandemic participants who had 2% assay positivity, detection of any SARS-CoV-2 antigen was significantly more frequent among 171 pandemic-era participants at three different time periods in the post-acute phase of infection. The absolute difference in SARS-CoV-2 plasma antigen prevalence was +11% (95% CI: +5.0% to +16%) at 3.0-6.0 months post-onset of COVID-19; +8.7% (95% CI: +3.1% to +14%) at 6.1 to 10.0 months; and +5.4% (95% CI: +0.42% to +10%) at 10.1-14.1 months. Hospitalization for acute COVID-19 and, among the non-hospitalized, worse self-reported health during acute COVID-19 were associated with greater post-acute phase antigen detection. CONCLUSIONS Compared to uninfected persons, there is an excess prevalence of SARS-CoV-2 antigenemia in SARS-CoV-2-infected individuals up to 14 months after acute COVID-19. These findings motivate an urgent research agenda regarding the short-term and long-term clinical manifestations of this viral persistence.
Collapse
Affiliation(s)
- Michael J. Peluso
- Division of HIV, Infectious Diseases, and Global Medicine, University of California, San Francisco, San Francisco, CA, USA
| | - Zoe N. Swank
- Harvard Medical School, Boston, MA, USA
- Department of Pathology, Brigham & Women’s Hospital, Boston, MA, USA
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, USA
| | - Sarah A. Goldberg
- Department of Epidemiology and Biostatistics, University of California, San Francisco, San Francisco, CA, USA
| | - Scott Lu
- Department of Epidemiology and Biostatistics, University of California, San Francisco, San Francisco, CA, USA
| | - Thomas Dalhuisen
- Department of Epidemiology and Biostatistics, University of California, San Francisco, San Francisco, CA, USA
| | - Ella Borberg
- Harvard Medical School, Boston, MA, USA
- Department of Pathology, Brigham & Women’s Hospital, Boston, MA, USA
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, USA
| | - Yasmeen Senussi
- Harvard Medical School, Boston, MA, USA
- Department of Pathology, Brigham & Women’s Hospital, Boston, MA, USA
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, USA
| | - Michael A. Luna
- Division of HIV, Infectious Diseases, and Global Medicine, University of California, San Francisco, San Francisco, CA, USA
| | - Celina Chang Song
- Division of HIV, Infectious Diseases, and Global Medicine, University of California, San Francisco, San Francisco, CA, USA
| | - Alexus Clark
- Division of HIV, Infectious Diseases, and Global Medicine, University of California, San Francisco, San Francisco, CA, USA
| | - Andhy Zamora
- Division of HIV, Infectious Diseases, and Global Medicine, University of California, San Francisco, San Francisco, CA, USA
| | - Megan Lew
- Division of HIV, Infectious Diseases, and Global Medicine, University of California, San Francisco, San Francisco, CA, USA
| | - Badri Viswanathan
- Department of Epidemiology and Biostatistics, University of California, San Francisco, San Francisco, CA, USA
| | - Beatrice Huang
- Division of HIV, Infectious Diseases, and Global Medicine, University of California, San Francisco, San Francisco, CA, USA
| | - Khamal Anglin
- Department of Epidemiology and Biostatistics, University of California, San Francisco, San Francisco, CA, USA
| | - Rebecca Hoh
- Division of HIV, Infectious Diseases, and Global Medicine, University of California, San Francisco, San Francisco, CA, USA
| | - Priscila Y. Hsue
- Division of Cardiology, University of California, San Francisco, San Francisco, CA, USA
| | | | - Matthew A. Spinelli
- Division of HIV, Infectious Diseases, and Global Medicine, University of California, San Francisco, San Francisco, CA, USA
| | - David V. Glidden
- Department of Epidemiology and Biostatistics, University of California, San Francisco, San Francisco, CA, USA
| | - Timothy J. Henrich
- Division of Experimental Medicine, University of California, San Francisco, San Francisco, CA, USA
| | - J. Daniel Kelly
- Department of Epidemiology and Biostatistics, University of California, San Francisco, San Francisco, CA, USA
| | - Steven G. Deeks
- Division of HIV, Infectious Diseases, and Global Medicine, University of California, San Francisco, San Francisco, CA, USA
| | - David R. Walt
- Harvard Medical School, Boston, MA, USA
- Department of Pathology, Brigham & Women’s Hospital, Boston, MA, USA
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, USA
| | - Jeffrey N. Martin
- Department of Epidemiology and Biostatistics, University of California, San Francisco, San Francisco, CA, USA
| |
Collapse
|
171
|
Zurlo M, Gasparello J, Verona M, Papi C, Cosenza LC, Finotti A, Marzaro G, Gambari R. The anti-SARS-CoV-2 BNT162b2 vaccine suppresses mithramycin-induced erythroid differentiation and expression of embryo-fetal globin genes in human erythroleukemia K562 cells. Exp Cell Res 2023; 433:113853. [PMID: 37944576 DOI: 10.1016/j.yexcr.2023.113853] [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: 09/16/2023] [Revised: 11/04/2023] [Accepted: 11/05/2023] [Indexed: 11/12/2023]
Abstract
The severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is causative of the ongoing coronavirus disease 2019 (COVID-19) pandemic. The SARS-CoV-2 Spike protein (S-protein) plays an important role in the early phase of SARS-CoV-2 infection through efficient interaction with ACE2. The S-protein is produced by RNA-based COVID-19 vaccines, that were fundamental for the reduction of the viral spread within the population and the clinical severity of COVID-19. However, the S-protein has been hypothesized to be responsible for damaging cells of several tissues and for some important side effects of RNA-based COVID-19 vaccines. Considering the impact of COVID-19 and SARS-CoV-2 infection on the hematopoietic system, the aim of this study was to verify the effect of the BNT162b2 vaccine on erythroid differentiation of the human K562 cell line, that has been in the past intensively studied as a model system mimicking some steps of erythropoiesis. In this context, we focused on hemoglobin production and induced expression of embryo-fetal globin genes, that are among the most important features of K562 erythroid differentiation. We found that the BNT162b2 vaccine suppresses mithramycin-induced erythroid differentiation of K562 cells. Reverse-transcription-qPCR and Western blotting assays demonstrated that suppression of erythroid differentiation was associated with sharp inhibition of the expression of α-globin and γ-globin mRNA accumulation. Inhibition of accumulation of ζ-globin and ε-globin mRNAs was also observed. In addition, we provide in silico studies suggesting a direct interaction between SARS-CoV-2 Spike protein and Hb Portland, that is the major hemoglobin produced by K562 cells. This study thus provides information suggesting the need of great attention on possible alteration of hematopoietic parameters following SARS-CoV-2 infection and/or COVID-19 vaccination.
Collapse
Affiliation(s)
- Matteo Zurlo
- Department of Life Sciences and Biotechnology, Section of Biochemistry and Molecular Biology, University of Ferrara, 44121 Ferrara, Italy.
| | - Jessica Gasparello
- Department of Life Sciences and Biotechnology, Section of Biochemistry and Molecular Biology, University of Ferrara, 44121 Ferrara, Italy
| | - Marco Verona
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, 35131 Padova, Italy
| | - Chiara Papi
- Department of Life Sciences and Biotechnology, Section of Biochemistry and Molecular Biology, University of Ferrara, 44121 Ferrara, Italy
| | - Lucia Carmela Cosenza
- Department of Life Sciences and Biotechnology, Section of Biochemistry and Molecular Biology, University of Ferrara, 44121 Ferrara, Italy
| | - Alessia Finotti
- Department of Life Sciences and Biotechnology, Section of Biochemistry and Molecular Biology, University of Ferrara, 44121 Ferrara, Italy; Center 'Chiara Gemmo and Elio Zago' for the Research on Thalassemia, University of Ferrara, 44121 Ferrara, Italy
| | - Giovanni Marzaro
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, 35131 Padova, Italy
| | - Roberto Gambari
- Department of Life Sciences and Biotechnology, Section of Biochemistry and Molecular Biology, University of Ferrara, 44121 Ferrara, Italy; Center 'Chiara Gemmo and Elio Zago' for the Research on Thalassemia, University of Ferrara, 44121 Ferrara, Italy.
| |
Collapse
|
172
|
Fernández-de-las-Peñas C, Raveendran AV, Giordano R, Arendt-Nielsen L. Long COVID or Post-COVID-19 Condition: Past, Present and Future Research Directions. Microorganisms 2023; 11:2959. [PMID: 38138102 PMCID: PMC10745830 DOI: 10.3390/microorganisms11122959] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2023] [Revised: 12/02/2023] [Accepted: 12/09/2023] [Indexed: 12/24/2023] Open
Abstract
The presence of symptoms after an acute SARS-CoV-2 infection (long-COVID) has become a worldwide healthcare emergency but remains underestimated and undertreated due to a lack of recognition of the condition and knowledge of the underlying mechanisms. In fact, the prevalence of post-COVID symptoms ranges from 50% during the first months after the infection up to 20% two-years after. This perspective review aimed to map the existing literature on post-COVID symptoms and to identify gaps in the literature to guide the global effort toward an improved understanding of long-COVID and suggest future research directions. There is a plethora of symptomatology that can be due to COVID-19; however, today, there is no clear classification and definition of this condition, termed long-COVID or post-COVID-19 condition. The heterogeneity in the symptomatology has led to the presence of groups/clusters of patients, which could exhibit different risk factors and different mechanisms. Viral persistence, long-lasting inflammation, immune dysregulation, autoimmune reactions, reactivation of latent infections, endothelial dysfunction and alteration in gut microbiota have been proposed as potential mechanisms explaining the complexity of long-COVID. In such an equation, viral biology (e.g., re-infections, SARS-CoV-2 variants), host biology (e.g., genetics, epigenetics) and external factors (e.g., vaccination) should be also considered. These various factors will be discussed in the current perspective review and future directions suggested.
Collapse
Affiliation(s)
- César Fernández-de-las-Peñas
- Department of Physical Therapy, Occupational Therapy, Physical Medicine and Rehabilitation, Universidad Rey Juan Carlos, 28922 Madrid, Spain
- Center for Neuroplasticity and Pain (CNAP), Center for Sensory-Motor Interaction (SMI), Department of Health Science and Technology, Faculty of Medicine, Aalborg University, DK-9220 Aalborg, Denmark; (R.G.); (L.A.-N.)
| | | | - Rocco Giordano
- Center for Neuroplasticity and Pain (CNAP), Center for Sensory-Motor Interaction (SMI), Department of Health Science and Technology, Faculty of Medicine, Aalborg University, DK-9220 Aalborg, Denmark; (R.G.); (L.A.-N.)
| | - Lars Arendt-Nielsen
- Center for Neuroplasticity and Pain (CNAP), Center for Sensory-Motor Interaction (SMI), Department of Health Science and Technology, Faculty of Medicine, Aalborg University, DK-9220 Aalborg, Denmark; (R.G.); (L.A.-N.)
- Department of Medical Gastroenterology, Mech-Sense, Aalborg University Hospital, DK-9000 Aalborg, Denmark
- Steno Diabetes Center North Denmark, Clinical Institute, Aalborg University Hospital, DK-9000 Aalborg, Denmark
| |
Collapse
|
173
|
Liu Y, Zhou Y, Xu W, Li J, Wang S, Shen X, Wen X, Liu L. Aptamer-based kinetically controlled DNA reactions coupled with metal-organic framework nanoprobes for sensitive detection of SARS-CoV-2 spike protein. ANALYTICAL METHODS : ADVANCING METHODS AND APPLICATIONS 2023; 15:6583-6589. [PMID: 38014562 DOI: 10.1039/d3ay01585h] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/29/2023]
Abstract
Since the outbreak in 2019, COVID-19, caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), has become the deadliest infectious disease worldwide for people of all ages, from children to older adults. As a main structural protein of SARS-CoV-2, spike protein is reported to play a key role in the entry of the virus into host cells and is considered as an effective antigenic marker for COVID-19 diagnosis. Herein, we develop a new aptamer-based fluorescence method for SARS-CoV-2 spike protein detection based on using kinetically controlled DNA reactions and metal-organic framework nanoprobes. Specifically, the binding of SARS-CoV-2 spike protein to its aptamer is designed to precisely control the kinetics of a DNA displacement reaction, leading to the release of free signaling probes. By reasonable integration of magnetic enrichment and exonuclease-fuelled recycling, the released probes efficiently disrupt the interaction within metal-organic framework nanoprobes, thereby generating a remarkable fluorescent response. Experimental results show that the method not only exhibits a wide linear range and a low detection limit of 7.8 fg mL-1 for SARS-CoV-2 spike protein detection but also demonstrates desirable specificity and utility in complex samples. Therefore, the method may provide a valuable tool for the detection of SARS-CoV-2 spike protein, and has bright prospects in the rapid diagnosis of COVID-19, which is of great significance for guiding rational treatment during a pandemic of respiratory infectious diseases and reducing the occurrence of severe disease in children.
Collapse
Affiliation(s)
- Yan Liu
- Department of Pediatrics, Chengdu Second People's Hospital, Chengdu 610021, Sichuan, China.
| | - Yuanlin Zhou
- Department of Pediatrics, Chengdu Second People's Hospital, Chengdu 610021, Sichuan, China.
| | - Wanting Xu
- Department of Pediatrics, Chengdu Second People's Hospital, Chengdu 610021, Sichuan, China.
| | - Jiarong Li
- College of Clinical Medicine, Southwest Medical University, Luzhou 646000, Sichuan, China
| | - Shuning Wang
- School of Life Sciences, Shanghai University, Shanghai 200444, China
| | - Xiaojia Shen
- Department of Pediatrics, Chengdu Second People's Hospital, Chengdu 610021, Sichuan, China.
| | - Xiaobin Wen
- Department of Pediatrics, Chengdu Second People's Hospital, Chengdu 610021, Sichuan, China.
| | - Li Liu
- Department of Pediatrics, Chengdu Second People's Hospital, Chengdu 610021, Sichuan, China.
| |
Collapse
|
174
|
Ailioaie LM, Ailioaie C, Litscher G. Gut Microbiota and Mitochondria: Health and Pathophysiological Aspects of Long COVID. Int J Mol Sci 2023; 24:17198. [PMID: 38139027 PMCID: PMC10743487 DOI: 10.3390/ijms242417198] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Revised: 11/30/2023] [Accepted: 12/03/2023] [Indexed: 12/24/2023] Open
Abstract
The current understanding of long COVID (LC) is still limited. This review highlights key findings regarding the role of gut microbiota, mitochondria, and the main pathophysiological aspects of LC revealed by clinical studies, related to the complex interplay between infection, intestinal dysbiosis, dysfunctional mitochondria, and systemic inflammation generated in a vicious circle, reflecting the molecular and cellular processes from the "leaky gut" to the "leaky electron transport chain (ETC)" into a quantum leap. The heterogeneity of LC has hindered progress in deciphering all the pathophysiological mechanisms, and therefore, the approach must be multidisciplinary, with a special focus not only on symptomatic management but also on addressing the underlying health problems of the patients. It is imperative to further assess and validate the effects of COVID-19 and LC on the gut microbiome and their relationship to infections with other viral agents or pathogens. Further studies are needed to better understand LC and expand the interdisciplinary points of view that are required to accurately diagnose and effectively treat this heterogeneous condition. Given the ability of SARS-CoV-2 to induce autoimmunity in susceptible patients, they should be monitored for symptoms of autoimmune disease after contracting the viral infection. One question remains open, namely, whether the various vaccines developed to end the pandemic will also induce autoimmunity. Recent data highlighted in this review have revealed that the persistence of SARS-CoV-2 and dysfunctional mitochondria in organs such as the heart and, to a lesser extent, the kidneys, liver, and lymph nodes, long after the organism has been able to clear the virus from the lungs, could be an explanation for LC.
Collapse
Affiliation(s)
- Laura Marinela Ailioaie
- Department of Medical Physics, Alexandru Ioan Cuza University, 11 Carol I Boulevard, 700506 Iasi, Romania; (L.M.A.); (C.A.)
| | - Constantin Ailioaie
- Department of Medical Physics, Alexandru Ioan Cuza University, 11 Carol I Boulevard, 700506 Iasi, Romania; (L.M.A.); (C.A.)
| | - Gerhard Litscher
- President of the International Society for Medical Laser Applications (ISLA Transcontinental), German Vice President of the German-Chinese Research Foundation (DCFG) for TCM, Honorary President of the European Federation of Acupuncture and Moxibustion Societies, Honorary Professor of China Beijing International Acupuncture Training Center, China Academy of Chinese Medical Sciences, Former Head of Two Research Units and the TCM Research Center at the Medical University of Graz, Auenbruggerplatz, 8036 Graz, Austria
| |
Collapse
|
175
|
Wu J, Yang H, Yu D, Yang X. Blood-derived product therapies for SARS-CoV-2 infection and long COVID. MedComm (Beijing) 2023; 4:e426. [PMID: 38020714 PMCID: PMC10651828 DOI: 10.1002/mco2.426] [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: 06/28/2023] [Revised: 10/15/2023] [Accepted: 10/25/2023] [Indexed: 12/01/2023] Open
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is capable of large-scale transmission and has caused the coronavirus disease 2019 (COVID-19) pandemic. Patients with COVID-19 may experience persistent long-term health issues, known as long COVID. Both acute SARS-CoV-2 infection and long COVID have resulted in persistent negative impacts on global public health. The effective application and development of blood-derived products are important strategies to combat the serious damage caused by COVID-19. Since the emergence of COVID-19, various blood-derived products that target or do not target SARS-CoV-2 have been investigated for therapeutic applications. SARS-CoV-2-targeting blood-derived products, including COVID-19 convalescent plasma, COVID-19 hyperimmune globulin, and recombinant anti-SARS-CoV-2 neutralizing immunoglobulin G, are virus-targeting and can provide immediate control of viral infection in the short term. Non-SARS-CoV-2-targeting blood-derived products, including intravenous immunoglobulin and human serum albumin exhibit anti-inflammatory, immunomodulatory, antioxidant, and anticoagulatory properties. Rational use of these products can be beneficial to patients with SARS-CoV-2 infection or long COVID. With evidence accumulated since the pandemic began, we here summarize the progress of blood-derived product therapies for COVID-19, discuss the effective methods and scenarios regarding these therapies, and provide guidance and suggestions for clinical treatment.
Collapse
Affiliation(s)
- Junzheng Wu
- Chengdu Rongsheng Pharmaceuticals Co., Ltd.ChengduChina
| | | | - Ding Yu
- Chengdu Rongsheng Pharmaceuticals Co., Ltd.ChengduChina
- Beijing Tiantan Biological Products Co., Ltd.BeijingChina
| | | |
Collapse
|
176
|
Salazar-Ardiles C, Asserella-Rebollo L, Cornejo C, Arias D, Vasquez-Muñoz M, Toledo C, Andrade DC. Molecular diagnostic approaches for SARS-CoV-2 detection and pathophysiological consequences. Mol Biol Rep 2023; 50:10367-10382. [PMID: 37817022 DOI: 10.1007/s11033-023-08844-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2023] [Accepted: 09/25/2023] [Indexed: 10/12/2023]
Abstract
SARS-CoV-2, a novel coronavirus within the Coronaviridae family, is the causative agent behind the respiratory ailment referred to as COVID-19. Operating on a global scale, COVID-19 has led to a substantial number of fatalities, exerting profound effects on both public health and the global economy. The most frequently reported symptoms encompass fever, cough, muscle or body aches, loss of taste or smell, headaches, and fatigue. Furthermore, a subset of individuals may manifest more severe symptoms, including those consistent with viral pneumonitis, which can be so profound as to result in fatalities. Consequently, this situation has spurred the rapid advancement of disease diagnostic technologies worldwide. Predominantly employed in diagnosing COVID-19, the real-time quantitative reverse transcription PCR has been the foremost diagnostic method, effectively detecting SARS-CoV-2 viral RNA. As the pandemic has evolved, antigen and serological tests have emerged as valuable diagnostic tools. Antigen tests pinpoint specific viral proteins of SARS-CoV-2, offering swift results, while serological tests identify the presence of antibodies in blood samples. Additionally, there have been notable strides in sample collection methods, notably with the introduction of saliva-based tests, presenting a non-invasive substitute to nasopharyngeal swabs. Given the ongoing mutations in SARS-CoV-2, there has been a continuous need for genomic surveillance, encompassing full genome sequencing and the identification of new variants through Illumina technology and, more recently, nanopore metagenomic sequencing (SMTN). Consequently, while diagnostic testing methods for COVID-19 have experienced remarkable progress, no test is flawless, and there exist limitations with each technique, including sensitivity, specificity, sample collection, and the minimum viral load necessary for accurate detection. These aspects are comprehensively addressed within this current review.
Collapse
Affiliation(s)
- Camila Salazar-Ardiles
- Exercise Applied Physiology Laboratory, Centro de Investigación en Fisiología y Medicina de Altura (FIMEDALT), Biomedical Department, Faculty of Health Sciences, Universidad de Antofagasta, Av. Universidad de Antofagasta #02800, Antofagasta, Chile
| | | | - Carlos Cornejo
- Exercise Applied Physiology Laboratory, Centro de Investigación en Fisiología y Medicina de Altura (FIMEDALT), Biomedical Department, Faculty of Health Sciences, Universidad de Antofagasta, Av. Universidad de Antofagasta #02800, Antofagasta, Chile
| | - Dayana Arias
- Exercise Applied Physiology Laboratory, Centro de Investigación en Fisiología y Medicina de Altura (FIMEDALT), Biomedical Department, Faculty of Health Sciences, Universidad de Antofagasta, Av. Universidad de Antofagasta #02800, Antofagasta, Chile
| | - Manuel Vasquez-Muñoz
- Dirección de Docencia de Especialidades Médicas, Dirección de Postgrado, Facultad de Medicina y Ciencias de la Salud, Universidad Mayor, Santiago, Chile
| | - Camilo Toledo
- Laboratory of Cardiorespiratory and Sleep Physiology, Institute of Physiology, Faculty of Medicine, Universidad Austral de Chile, Valdivia, Chile
| | - David C Andrade
- Exercise Applied Physiology Laboratory, Centro de Investigación en Fisiología y Medicina de Altura (FIMEDALT), Biomedical Department, Faculty of Health Sciences, Universidad de Antofagasta, Av. Universidad de Antofagasta #02800, Antofagasta, Chile.
| |
Collapse
|
177
|
Scheim DE, Vottero P, Santin AD, Hirsh AG. Sialylated Glycan Bindings from SARS-CoV-2 Spike Protein to Blood and Endothelial Cells Govern the Severe Morbidities of COVID-19. Int J Mol Sci 2023; 24:17039. [PMID: 38069362 PMCID: PMC10871123 DOI: 10.3390/ijms242317039] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Revised: 11/24/2023] [Accepted: 11/27/2023] [Indexed: 12/18/2023] Open
Abstract
Consistent with well-established biochemical properties of coronaviruses, sialylated glycan attachments between SARS-CoV-2 spike protein (SP) and host cells are key to the virus's pathology. SARS-CoV-2 SP attaches to and aggregates red blood cells (RBCs), as shown in many pre-clinical and clinical studies, causing pulmonary and extrapulmonary microthrombi and hypoxia in severe COVID-19 patients. SARS-CoV-2 SP attachments to the heavily sialylated surfaces of platelets (which, like RBCs, have no ACE2) and endothelial cells (having minimal ACE2) compound this vascular damage. Notably, experimentally induced RBC aggregation in vivo causes the same key morbidities as for severe COVID-19, including microvascular occlusion, blood clots, hypoxia and myocarditis. Key risk factors for COVID-19 morbidity, including older age, diabetes and obesity, are all characterized by markedly increased propensity to RBC clumping. For mammalian species, the degree of clinical susceptibility to COVID-19 correlates to RBC aggregability with p = 0.033. Notably, of the five human betacoronaviruses, the two common cold strains express an enzyme that releases glycan attachments, while the deadly SARS, SARS-CoV-2 and MERS do not, although viral loads for COVID-19 and the two common cold infections are similar. These biochemical insights also explain the previously puzzling clinical efficacy of certain generics against COVID-19 and may support the development of future therapeutic strategies for COVID-19 and long COVID patients.
Collapse
Affiliation(s)
- David E Scheim
- US Public Health Service, Commissioned Corps, Inactive Reserve, Blacksburg, VA 24060, USA
| | - Paola Vottero
- Department of Biomedical Engineering, University of Alberta, Edmonton, AB T6G 1Z2, Canada
| | - Alessandro D Santin
- Department of Obstetrics, Gynecology & Reproductive Sciences, Yale School of Medicine, P.O. Box 208063, New Haven, CT 06520, USA
| | | |
Collapse
|
178
|
Paniskaki K, Goretzki S, Anft M, Konik MJ, Meister TL, Pfaender S, Lechtenberg K, Vogl M, Dogan B, Dolff S, Westhoff TH, Rohn H, Felderhoff-Mueser U, Stervbo U, Witzke O, Dohna-Schwake C, Babel N. Increased SARS-CoV-2 reactive low avidity T cells producing inflammatory cytokines in pediatric post-acute COVID-19 sequelae (PASC). Pediatr Allergy Immunol 2023; 34:e14060. [PMID: 38146118 DOI: 10.1111/pai.14060] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/22/2023] [Revised: 11/13/2023] [Accepted: 11/29/2023] [Indexed: 12/27/2023]
Abstract
BACKGROUND A proportion of the convalescent SARS-CoV-2 pediatric population presents nonspecific symptoms, mental health problems, and a reduction in quality of life similar to myalgic encephalomyelitis/chronic fatigue syndrome (ME/CFS) and long COVID-19 symptomatic. However, data regarding its clinical manifestation and immune mechanisms are currently scarce. METHODS In this study, we perform a comprehensive clinical and immunological profiling of 17 convalescent COVID-19 children with post-acute COVID-19 sequelae (PASC) manifestation and 13 convalescent children without PASC manifestation. A detailed medical history, blood and instrumental tests, and physical examination were obtained from all patients. SARS-CoV-2 reactive T-cell response was analyzed via multiparametric flow cytometry and the humoral immunity was addressed via pseudovirus neutralization and ELISA assay. RESULTS The most common PASC symptoms were shortness of breath/exercise intolerance, paresthesia, smell/taste disturbance, chest pain, dyspnea, headache, and lack of concentration. Blood count and clinical chemistry showed no statistical differences among the study groups. We detected higher frequencies of spike (S) reactive CD4+ and CD8+ T cells among the PASC study group, characterized by TNFα and IFNγ production and low functional avidity. CRP levels are positively correlated with IFNγ producing reactive CD8+ T cells. CONCLUSIONS Our data might indicate a possible involvement of a persistent cellular inflammatory response triggered by SARS-CoV-2 in the development of the observed sequelae in pediatric PASC. These results may have implications on future therapeutic and prevention strategies.
Collapse
Affiliation(s)
- Krystallenia Paniskaki
- Department of Infectious Diseases, West German Centre of Infectious Diseases, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
- Center for Translational Medicine and Immune Diagnostics Laboratory, Medical Department I, Marien Hospital Herne, University Hospital of the Ruhr-University Bochum, Bochum, Germany
| | - Sarah Goretzki
- Department of Infectious Diseases, West German Centre of Infectious Diseases, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
- Department of Pediatrics I, Children's Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Moritz Anft
- Center for Translational Medicine and Immune Diagnostics Laboratory, Medical Department I, Marien Hospital Herne, University Hospital of the Ruhr-University Bochum, Bochum, Germany
| | - Margarethe J Konik
- Department of Infectious Diseases, West German Centre of Infectious Diseases, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Toni L Meister
- Department of Molecular and Medical Virology, Ruhr-University Bochum, Bochum, Germany
| | - Stephanie Pfaender
- Department of Molecular and Medical Virology, Ruhr-University Bochum, Bochum, Germany
| | - Klara Lechtenberg
- Department of Pediatrics I, Children's Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Melanie Vogl
- Department of Pediatrics III, Pediatric Pulmonology and Sleep Medicine, Children's Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Burcin Dogan
- Department of Pediatrics I, Children's Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Sebastian Dolff
- Department of Infectious Diseases, West German Centre of Infectious Diseases, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Timm H Westhoff
- Medical Department I, Marien Hospital Herne, University Hospital of the Ruhr-University Bochum, Herne, Germany
| | - Hana Rohn
- Department of Infectious Diseases, West German Centre of Infectious Diseases, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Ursula Felderhoff-Mueser
- Department of Pediatrics I, Children's Hospital Essen, University of Duisburg-Essen, Essen, Germany
- Center for Translational Neuro- and Behavioral Sciences C-TNBS, University of Duisburg-Essen, Essen, Germany
| | - Ulrik Stervbo
- Center for Translational Medicine and Immune Diagnostics Laboratory, Medical Department I, Marien Hospital Herne, University Hospital of the Ruhr-University Bochum, Bochum, Germany
| | - Oliver Witzke
- Department of Infectious Diseases, West German Centre of Infectious Diseases, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Christian Dohna-Schwake
- Department of Infectious Diseases, West German Centre of Infectious Diseases, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
- Department of Pediatrics I, Children's Hospital Essen, University of Duisburg-Essen, Essen, Germany
- Center for Translational Neuro- and Behavioral Sciences C-TNBS, University of Duisburg-Essen, Essen, Germany
| | - Nina Babel
- Center for Translational Medicine and Immune Diagnostics Laboratory, Medical Department I, Marien Hospital Herne, University Hospital of the Ruhr-University Bochum, Bochum, Germany
- Berlin Institute of Health at Charité - University Clinic Berlin, BIH Center for Regenerative Therapies (BCRT) Berlin, Berlin, Germany
| |
Collapse
|
179
|
Liu Y, Gu X, Li H, Zhang H, Xu J. Mechanisms of long COVID: An updated review. CHINESE MEDICAL JOURNAL PULMONARY AND CRITICAL CARE MEDICINE 2023; 1:231-240. [PMID: 39171285 PMCID: PMC11332859 DOI: 10.1016/j.pccm.2023.10.003] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/29/2023] [Indexed: 08/23/2024]
Abstract
The coronavirus disease 2019 (COVID-19) pandemic has been ongoing for more than 3 years, with an enormous impact on global health and economies. In some patients, symptoms and signs may remain after recovery from severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection, which cannot be explained by an alternate diagnosis; this condition has been defined as long COVID. Long COVID may exist in patients with both mild and severe disease and is prevalent after infection with different SARS-CoV-2 variants. The most common symptoms include fatigue, dyspnea, and other symptoms involving multiple organs. Vaccination results in lower rates of long COVID. To date, the mechanisms of long COVID remain unclear. In this narrative review, we summarized the clinical presentations and current evidence regarding the pathogenesis of long COVID.
Collapse
Affiliation(s)
- Yan Liu
- National Center for Respiratory Medicine; State Key Laboratory of Respiratory Health and Multimorbidity; National Clinical Research Center for Respiratory Diseases; Institute of Respiratory Medicine, Chinese Academy of Medical Sciences; Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, China–Japan Friendship Hospital, Beijing 100029, China
- Department of Infectious Disease, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, Shandong 264000, China
| | - Xiaoying Gu
- Department of Clinical Research and Data Management, Center of Respiratory Medicine, China-Japan Friendship Hospital, Beijing 100029, China
| | - Haibo Li
- National Center for Respiratory Medicine; State Key Laboratory of Respiratory Health and Multimorbidity; National Clinical Research Center for Respiratory Diseases; Institute of Respiratory Medicine, Chinese Academy of Medical Sciences; Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, China–Japan Friendship Hospital, Beijing 100029, China
| | - Hui Zhang
- National Center for Respiratory Medicine; State Key Laboratory of Respiratory Health and Multimorbidity; National Clinical Research Center for Respiratory Diseases; Institute of Respiratory Medicine, Chinese Academy of Medical Sciences; Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, China–Japan Friendship Hospital, Beijing 100029, China
- Department of Pulmonary and Critical Care Medicine, China–Japan Friendship Hospital, Capital Medical University, Beijing 100029, China
| | - Jiuyang Xu
- National Center for Respiratory Medicine; State Key Laboratory of Respiratory Health and Multimorbidity; National Clinical Research Center for Respiratory Diseases; Institute of Respiratory Medicine, Chinese Academy of Medical Sciences; Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, China–Japan Friendship Hospital, Beijing 100029, China
| |
Collapse
|
180
|
Shimohata T. [Cognitive Impairment as a Sequela of COVID-19: Pathophysiology and Prospects for Treatment]. Rinsho Shinkeigaku 2023; 63:725-731. [PMID: 37880112 DOI: 10.5692/clinicalneurol.cn-001902] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2023]
Abstract
Cognitive impairment has been reported as a potential sequela of COVID-19. Risk factors associated with this impairment include advanced age, severe infection, and prolonged duration of anosmia (loss of smell). Furthermore, COVID-19 has been reported as a risk factor for Alzheimer disease, and even mild infections have been associated with visuospatial cognitive impairments. While multiple pathophysiological mechanisms have been implicated in COVID-19-related cognitive impairment, persistent infection by the SARS-CoV-2 virus is of particular interest due to its potential implications for treatment. Persistent infection could lead to cognitive impairments through mechanisms such as neurotoxicity mediated by spike proteins, neuroinflammation induced by cytokines, and neuronal cell fusion (syncytia). In terms of treatment, the effectiveness of vaccination has been demonstrated in meta-analyses, and drugs like metformin and antiviral agents hold promise.
Collapse
|
181
|
Hikmet RG, Wejse C, Agergaard J. Effect of Vitamin D in Long COVID Patients. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2023; 20:7058. [PMID: 37998290 PMCID: PMC10671780 DOI: 10.3390/ijerph20227058] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/27/2023] [Revised: 11/09/2023] [Accepted: 11/11/2023] [Indexed: 11/25/2023]
Abstract
Vitamin D deficiency has been studied in the context of acute coronavirus disease 2019 (COVID-19), revealing associations with increased severity and mortality. Yet, the influence of vitamin D on long COVID symptoms remains unknown. The purpose of this study is to examine the effect of vitamin D on long COVID symptoms. Over the study period, 50,432 individuals within the catchment area of the outpatient COVID-19 clinic tested positive for SARS-CoV-2 via PCR, with 547 patients subsequently referred to a specialized Post-COVID Clinic, and 447 of them enrolled in the study. Patient-reported symptoms and paraclinical measures including vitamin D were evaluated in 442 patients. The majority of participants were female (72%, n = 320/442). The consumption of alcohol and number of current smokers were low. Low vitamin D was observed in 26% (n = 115/442) of the patients, most commonly in male participants (odds ratio (OR) = 1.77, 95% confidence interval (CI) (1.12, 2.79), p = 0.014). Additionally, low vitamin D was correlated with a younger mean age of 41 years (standard deviation (SD) = 12) as opposed to 48 years (SD = 13) in patients with normal vitamin D levels (OR = 0.96, 95% CI (0.94, 0.97), p < 0.001). While our study population indicated a potentially higher prevalence of vitamin D insufficiency in this population compared to the general population, no significant differences in prevalence of symptom or symptom severity scores were observed between the low and normal vitamin D groups. In patients in a Post-COVID Clinic, we found no association between vitamin D levels and long COVID symptoms.
Collapse
Affiliation(s)
- Ramsen Ghasan Hikmet
- Department of Infectious Diseases, Aarhus University Hospital, 8200 Aarhus, Denmark (J.A.)
| | - Christian Wejse
- Department of Infectious Diseases, Aarhus University Hospital, 8200 Aarhus, Denmark (J.A.)
- Center for Global Health, Aarhus University (GloHAU), 8000 Aarhus, Denmark
| | - Jane Agergaard
- Department of Infectious Diseases, Aarhus University Hospital, 8200 Aarhus, Denmark (J.A.)
- Department of Clinical Medicine, Aarhus University, 8000 Aarhus, Denmark
| |
Collapse
|
182
|
Luchian ML, Higny J, Benoit M, Robaye B, Berners Y, Henry JP, Colle B, Xhaët O, Blommaert D, Droogmans S, Motoc AI, Cosyns B, Gabriel L, Guedes A, Demeure F. Unmasking Pandemic Echoes: An In-Depth Review of Long COVID's Unabated Cardiovascular Consequences beyond 2020. Diagnostics (Basel) 2023; 13:3368. [PMID: 37958264 PMCID: PMC10647305 DOI: 10.3390/diagnostics13213368] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2023] [Revised: 10/23/2023] [Accepted: 10/30/2023] [Indexed: 11/15/2023] Open
Abstract
At the beginning of 2020, coronavirus disease 2019 (COVID-19) emerged as a new pandemic, leading to a worldwide health crisis and overwhelming healthcare systems due to high numbers of hospital admissions, insufficient resources, and a lack of standardized therapeutic protocols. Multiple genetic variants of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) have been detected since its first public declaration in 2020, some of them being considered variants of concern (VOCs) corresponding to several pandemic waves. Nevertheless, a growing number of COVID-19 patients are continuously discharged from hospitals, remaining symptomatic even months after their first episode of COVID-19 infection. Long COVID-19 or 'post-acute COVID-19 syndrome' emerged as the new pandemic, being characterized by a high variability of clinical manifestations ranging from cardiorespiratory and neurological symptoms such as chest pain, exertional dyspnoea or cognitive disturbance to psychological disturbances, e.g., depression, anxiety or sleep disturbance with a crucial impact on patients' quality of life. Moreover, Long COVID is viewed as a new cardiovascular risk factor capable of modifying the trajectory of current and future cardiovascular diseases, altering the patients' prognosis. Therefore, in this review we address the current definitions of Long COVID and its pathophysiology, with a focus on cardiovascular manifestations. Furthermore, we aim to review the mechanisms of acute and chronic cardiac injury and the variety of cardiovascular sequelae observed in recovered COVID-19 patients, in addition to the potential role of Long COVID clinics in the medical management of this new condition. We will further address the role of future research for a better understanding of the actual impact of Long COVID and future therapeutic directions.
Collapse
Affiliation(s)
- Maria-Luiza Luchian
- Department of Cardiology, Université Catholique de Louvain, CHU UCL Namur Site Godinne, Av. Dr. G. Thérasse, 1, 5530 Yvoir, Belgium (A.G.); (F.D.)
| | - Julien Higny
- Department of Cardiology, Université Catholique de Louvain, CHU UCL Namur Site Godinne, Av. Dr. G. Thérasse, 1, 5530 Yvoir, Belgium (A.G.); (F.D.)
| | - Martin Benoit
- Department of Cardiology, Université Catholique de Louvain, CHU UCL Namur Site Godinne, Av. Dr. G. Thérasse, 1, 5530 Yvoir, Belgium (A.G.); (F.D.)
| | - Benoit Robaye
- Department of Cardiology, Université Catholique de Louvain, CHU UCL Namur Site Godinne, Av. Dr. G. Thérasse, 1, 5530 Yvoir, Belgium (A.G.); (F.D.)
| | - Yannick Berners
- Department of Cardiology, Université Catholique de Louvain, CHU UCL Namur Site Godinne, Av. Dr. G. Thérasse, 1, 5530 Yvoir, Belgium (A.G.); (F.D.)
| | - Jean-Philippe Henry
- Department of Cardiology, Université Catholique de Louvain, CHU UCL Namur Site Godinne, Av. Dr. G. Thérasse, 1, 5530 Yvoir, Belgium (A.G.); (F.D.)
| | - Benjamin Colle
- Department of Cardiology, Université Catholique de Louvain, CHU UCL Namur Site Godinne, Av. Dr. G. Thérasse, 1, 5530 Yvoir, Belgium (A.G.); (F.D.)
| | - Olivier Xhaët
- Department of Cardiology, Université Catholique de Louvain, CHU UCL Namur Site Godinne, Av. Dr. G. Thérasse, 1, 5530 Yvoir, Belgium (A.G.); (F.D.)
| | - Dominique Blommaert
- Department of Cardiology, Université Catholique de Louvain, CHU UCL Namur Site Godinne, Av. Dr. G. Thérasse, 1, 5530 Yvoir, Belgium (A.G.); (F.D.)
| | - Steven Droogmans
- Department of Cardiology, Centrum voor Hart-en Vaatziekten, Universitair Ziekenhuis Brussel, Vrije Universiteit Brussel (VUB), Laarbeeklaan 101, 1090 Brussels, Belgium
| | - Andreea Iulia Motoc
- Department of Cardiology, Centrum voor Hart-en Vaatziekten, Universitair Ziekenhuis Brussel, Vrije Universiteit Brussel (VUB), Laarbeeklaan 101, 1090 Brussels, Belgium
| | - Bernard Cosyns
- Department of Cardiology, Centrum voor Hart-en Vaatziekten, Universitair Ziekenhuis Brussel, Vrije Universiteit Brussel (VUB), Laarbeeklaan 101, 1090 Brussels, Belgium
| | - Laurence Gabriel
- Department of Cardiology, Université Catholique de Louvain, CHU UCL Namur Site Godinne, Av. Dr. G. Thérasse, 1, 5530 Yvoir, Belgium (A.G.); (F.D.)
| | - Antoine Guedes
- Department of Cardiology, Université Catholique de Louvain, CHU UCL Namur Site Godinne, Av. Dr. G. Thérasse, 1, 5530 Yvoir, Belgium (A.G.); (F.D.)
| | - Fabian Demeure
- Department of Cardiology, Université Catholique de Louvain, CHU UCL Namur Site Godinne, Av. Dr. G. Thérasse, 1, 5530 Yvoir, Belgium (A.G.); (F.D.)
| |
Collapse
|
183
|
Klein J, Wood J, Jaycox JR, Dhodapkar RM, Lu P, Gehlhausen JR, Tabachnikova A, Greene K, Tabacof L, Malik AA, Silva Monteiro V, Silva J, Kamath K, Zhang M, Dhal A, Ott IM, Valle G, Peña-Hernández M, Mao T, Bhattacharjee B, Takahashi T, Lucas C, Song E, McCarthy D, Breyman E, Tosto-Mancuso J, Dai Y, Perotti E, Akduman K, Tzeng TJ, Xu L, Geraghty AC, Monje M, Yildirim I, Shon J, Medzhitov R, Lutchmansingh D, Possick JD, Kaminski N, Omer SB, Krumholz HM, Guan L, Dela Cruz CS, van Dijk D, Ring AM, Putrino D, Iwasaki A. Distinguishing features of long COVID identified through immune profiling. Nature 2023; 623:139-148. [PMID: 37748514 PMCID: PMC10620090 DOI: 10.1038/s41586-023-06651-y] [Citation(s) in RCA: 261] [Impact Index Per Article: 130.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2022] [Accepted: 09/18/2023] [Indexed: 09/27/2023]
Abstract
Post-acute infection syndromes may develop after acute viral disease1. Infection with SARS-CoV-2 can result in the development of a post-acute infection syndrome known as long COVID. Individuals with long COVID frequently report unremitting fatigue, post-exertional malaise, and a variety of cognitive and autonomic dysfunctions2-4. However, the biological processes that are associated with the development and persistence of these symptoms are unclear. Here 275 individuals with or without long COVID were enrolled in a cross-sectional study that included multidimensional immune phenotyping and unbiased machine learning methods to identify biological features associated with long COVID. Marked differences were noted in circulating myeloid and lymphocyte populations relative to the matched controls, as well as evidence of exaggerated humoral responses directed against SARS-CoV-2 among participants with long COVID. Furthermore, higher antibody responses directed against non-SARS-CoV-2 viral pathogens were observed among individuals with long COVID, particularly Epstein-Barr virus. Levels of soluble immune mediators and hormones varied among groups, with cortisol levels being lower among participants with long COVID. Integration of immune phenotyping data into unbiased machine learning models identified the key features that are most strongly associated with long COVID status. Collectively, these findings may help to guide future studies into the pathobiology of long COVID and help with developing relevant biomarkers.
Collapse
Affiliation(s)
- Jon Klein
- Department of Immunobiology, Yale School of Medicine, New Haven, CT, USA
| | - Jamie Wood
- Abilities Research Center, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Jillian R Jaycox
- Department of Immunobiology, Yale School of Medicine, New Haven, CT, USA
| | - Rahul M Dhodapkar
- Department of Immunobiology, Yale School of Medicine, New Haven, CT, USA
- Department of Ophthalmology, USC Keck School of Medicine, Los Angeles, CA, USA
| | - Peiwen Lu
- Department of Immunobiology, Yale School of Medicine, New Haven, CT, USA
| | - Jeff R Gehlhausen
- Department of Immunobiology, Yale School of Medicine, New Haven, CT, USA
- Department of Dermatology, Yale School of Medicine, New Haven, CT, USA
| | | | - Kerrie Greene
- Department of Immunobiology, Yale School of Medicine, New Haven, CT, USA
| | - Laura Tabacof
- Abilities Research Center, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Amyn A Malik
- Yale Institute for Global Health, Yale School of Public Health, New Haven, CT, USA
| | | | - Julio Silva
- Department of Immunobiology, Yale School of Medicine, New Haven, CT, USA
| | | | | | | | - Isabel M Ott
- Department of Immunobiology, Yale School of Medicine, New Haven, CT, USA
| | - Gabrielee Valle
- Department of Internal Medicine (Pulmonary, Critical Care and Sleep Medicine), Yale School of Medicine, New Haven, CT, USA
| | - Mario Peña-Hernández
- Department of Immunobiology, Yale School of Medicine, New Haven, CT, USA
- Department of Microbiology, Yale School of Medicine, New Haven, CT, USA
| | - Tianyang Mao
- Department of Immunobiology, Yale School of Medicine, New Haven, CT, USA
| | | | - Takehiro Takahashi
- Department of Immunobiology, Yale School of Medicine, New Haven, CT, USA
| | - Carolina Lucas
- Department of Immunobiology, Yale School of Medicine, New Haven, CT, USA
- Center for Infection and Immunity, Yale School of Medicine, New Haven, CT, USA
| | - Eric Song
- Department of Immunobiology, Yale School of Medicine, New Haven, CT, USA
| | - Dayna McCarthy
- Abilities Research Center, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Erica Breyman
- Abilities Research Center, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Jenna Tosto-Mancuso
- Abilities Research Center, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Yile Dai
- Department of Immunobiology, Yale School of Medicine, New Haven, CT, USA
| | - Emily Perotti
- Department of Immunobiology, Yale School of Medicine, New Haven, CT, USA
| | - Koray Akduman
- Department of Immunobiology, Yale School of Medicine, New Haven, CT, USA
| | - Tiffany J Tzeng
- Department of Immunobiology, Yale School of Medicine, New Haven, CT, USA
| | - Lan Xu
- Department of Immunobiology, Yale School of Medicine, New Haven, CT, USA
| | - Anna C Geraghty
- Department of Neurology and Neurological Sciences, Stanford University, Palo Alto, CA, USA
| | - Michelle Monje
- Department of Neurology and Neurological Sciences, Stanford University, Palo Alto, CA, USA
- Howard Hughes Medical Institute, Chevy Chase, MD, USA
| | - Inci Yildirim
- Yale Institute for Global Health, Yale School of Public Health, New Haven, CT, USA
- Center for Infection and Immunity, Yale School of Medicine, New Haven, CT, USA
- Department of Pediatrics (Infectious Diseases), Yale New Haven Hospital, New Haven, CT, USA
- Department of Epidemiology of Microbial Diseases, Yale School of Public Health, New Haven, CT, USA
| | | | - Ruslan Medzhitov
- Department of Immunobiology, Yale School of Medicine, New Haven, CT, USA
- Center for Infection and Immunity, Yale School of Medicine, New Haven, CT, USA
- Howard Hughes Medical Institute, Chevy Chase, MD, USA
| | - Denyse Lutchmansingh
- Department of Internal Medicine (Pulmonary, Critical Care and Sleep Medicine), Yale School of Medicine, New Haven, CT, USA
| | - Jennifer D Possick
- Department of Internal Medicine (Pulmonary, Critical Care and Sleep Medicine), Yale School of Medicine, New Haven, CT, USA
| | - Naftali Kaminski
- Department of Internal Medicine (Pulmonary, Critical Care and Sleep Medicine), Yale School of Medicine, New Haven, CT, USA
| | - Saad B Omer
- Yale Institute for Global Health, Yale School of Public Health, New Haven, CT, USA
- Center for Infection and Immunity, Yale School of Medicine, New Haven, CT, USA
- Department of Epidemiology of Microbial Diseases, Yale School of Public Health, New Haven, CT, USA
- Department of Internal Medicine (Infectious Diseases), Yale School of Medicine, New Haven, CT, USA
| | - Harlan M Krumholz
- Center for Infection and Immunity, Yale School of Medicine, New Haven, CT, USA
- Center for Outcomes Research and Evaluation, Yale New Haven Hospital, New Haven, CT, USA
- Section of Cardiovascular Medicine, Department of Internal Medicine, Yale School of Medicine, New Haven, CT, USA
- Department of Health Policy and Management, Yale School of Public Health, New Haven, CT, USA
| | - Leying Guan
- Center for Infection and Immunity, Yale School of Medicine, New Haven, CT, USA
- Department of Biostatistics, Yale School of Public Health, New Haven, CT, USA
| | - Charles S Dela Cruz
- Department of Internal Medicine (Pulmonary, Critical Care and Sleep Medicine), Yale School of Medicine, New Haven, CT, USA
- Center for Infection and Immunity, Yale School of Medicine, New Haven, CT, USA
| | - David van Dijk
- Center for Infection and Immunity, Yale School of Medicine, New Haven, CT, USA.
- Department of Computer Science, Yale University, New Haven, CT, USA.
- Department of Internal Medicine (Cardiology), Yale School of Medicine, New Haven, CT, USA.
| | - Aaron M Ring
- Department of Immunobiology, Yale School of Medicine, New Haven, CT, USA.
- Center for Infection and Immunity, Yale School of Medicine, New Haven, CT, USA.
| | - David Putrino
- Abilities Research Center, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- Department of Rehabilitation and Human Performance, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| | - Akiko Iwasaki
- Department of Immunobiology, Yale School of Medicine, New Haven, CT, USA.
- Center for Infection and Immunity, Yale School of Medicine, New Haven, CT, USA.
- Howard Hughes Medical Institute, Chevy Chase, MD, USA.
| |
Collapse
|
184
|
Steiner S, Fehrer A, Hoheisel F, Schoening S, Aschenbrenner A, Babel N, Bellmann-Strobl J, Finke C, Fluge Ø, Froehlich L, Goebel A, Grande B, Haas JP, Hohberger B, Jason LA, Komaroff AL, Lacerda E, Liebl M, Maier A, Mella O, Nacul L, Paul F, Prusty BK, Puta C, Riemekasten G, Ries W, Rowe PC, Sawitzki B, Shoenfeld Y, Schultze JL, Seifert M, Sepúlveda N, Sotzny F, Stein E, Stingl M, Ufer F, Veauthier C, Westermeier F, Wirth K, Wolfarth B, Zalewski P, Behrends U, Scheibenbogen C. Understanding, diagnosing, and treating Myalgic encephalomyelitis/chronic fatigue syndrome - State of the art: Report of the 2nd international meeting at the Charité Fatigue Center. Autoimmun Rev 2023; 22:103452. [PMID: 37742748 DOI: 10.1016/j.autrev.2023.103452] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2023] [Accepted: 09/20/2023] [Indexed: 09/26/2023]
Abstract
Myalgic Encephalomyelitis/Chronic Fatigue Syndrome (ME/CFS) is a devastating disease affecting millions of people worldwide. Due to the 2019 pandemic of coronavirus disease (COVID-19), we are facing a significant increase of ME/CFS prevalence. On May 11th to 12th, 2023, the second international ME/CFS conference of the Charité Fatigue Center was held in Berlin, Germany, focusing on pathomechanisms, diagnosis, and treatment. During the two-day conference, more than 100 researchers from various research fields met on-site and over 700 attendees participated online to discuss the state of the art and novel findings in this field. Key topics from the conference included: the role of the immune system, dysfunction of endothelial and autonomic nervous system, and viral reactivation. Furthermore, there were presentations on innovative diagnostic measures and assessments for this complex disease, cutting-edge treatment approaches, and clinical studies. Despite the increased public attention due to the COVID-19 pandemic, the subsequent rise of Long COVID-19 cases, and the rise of funding opportunities to unravel the pathomechanisms underlying ME/CFS, this severe disease remains highly underresearched. Future adequately funded research efforts are needed to further explore the disease etiology and to identify diagnostic markers and targeted therapies.
Collapse
Affiliation(s)
- Sophie Steiner
- Institute of Medical Immunology, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt Universität zu Berlin and Berlin Institute of Health (BIH), Berlin, Germany
| | - Annick Fehrer
- Institute of Medical Immunology, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt Universität zu Berlin and Berlin Institute of Health (BIH), Berlin, Germany
| | - Friederike Hoheisel
- Institute of Medical Immunology, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt Universität zu Berlin and Berlin Institute of Health (BIH), Berlin, Germany.
| | | | - Anna Aschenbrenner
- Systems Medicine, Deutsches Zentrum für Neurodegenerative Erkrankungen (DZNE), Bonn, Germany
| | - Nina Babel
- Institute of Medical Immunology, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt Universität zu Berlin and Berlin Institute of Health (BIH), Berlin, Germany; Center for Translational Medicine and Immune Diagnostics Laboratory, Medical Department I, Ruhr-University Bochum, Marien Hospital Herne, University Hospital of the Ruhr-University Bochum, Herne, Germany; Berlin Institute of Health at Charité-Universitätsmedizin Berlin, BIH Center for Regenerative Therapies (BCRT), Charitéplatz 1, Berlin, Germany
| | - Judith Bellmann-Strobl
- Department of Neurology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany; Experimental and Clinical Research Center, A Cooperation Between the Max Delbrück Center for Molecular Medicine in the Helmholtz Association and Charité-Universitätsmedizin Berlin, Berlin, Germany; Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany; NeuroCure Clinical Research Center, Charité Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin Institute of Health, and Max Delbrück Center for Molecular Medicine, Berlin, Germany
| | - Carsten Finke
- Department of Neurology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Øystein Fluge
- Department of Oncology and Medical Physics, Haukeland University Hospital, Bergen, Norway; Department of Clinical Sciences, University of Bergen, Bergen, Norway
| | - Laura Froehlich
- Center of Advanced Technology for Assisted Learning and Predictive Analytics (CATALPA), FernUniversität in Hagen, Germany
| | - Andreas Goebel
- Pain Research Institute, Institute of Life Course and Medical Sciences, University of Liverpool, and Walton Centre NHS Foundation Trust, Liverpool, UK
| | | | - Johannes-Peter Haas
- Deutsches Zentrum für Kinder- und Jugendrheumatologie, Zentrum für Schmerztherapie junger Menschen, Garmisch-Partenkirchen, Germany
| | - Bettina Hohberger
- Department of Ophthalmology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Leonard A Jason
- Center for Community Research, DePaul University, Chicago, IL, USA
| | - Anthony L Komaroff
- Division of General Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Eliana Lacerda
- Department of Clinical Research, Faculty of Infectious and Tropical Diseases, London School of Hygiene and Tropical Medicine, London, UK
| | - Max Liebl
- Department of Physical Medicine, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt Universität zu Berlin, Berlin, Germany
| | - Andrea Maier
- Department of Neurology, Medical Faculty RWTH Aachen University, Aachen, Germany
| | - Olav Mella
- Department of Oncology and Medical Physics, Haukeland University Hospital, Bergen, Norway; Department of Clinical Sciences, University of Bergen, Bergen, Norway
| | - Luis Nacul
- Department of Clinical Research, Faculty of Infectious and Tropical Diseases, London School of Hygiene and Tropical Medicine, London, UK; Women's Health Research Institute, British Columbia Women's Hospital, Vancouver, BC, Canada; Department of Family Practice, Faculty of Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Friedemann Paul
- Experimental and Clinical Research Center, A Cooperation Between the Max Delbrück Center for Molecular Medicine in the Helmholtz Association and Charité-Universitätsmedizin Berlin, Berlin, Germany; Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany; NeuroCure Clinical Research Center, Charité Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin Institute of Health, and Max Delbrück Center for Molecular Medicine, Berlin, Germany
| | - Bhupesh K Prusty
- Institute for Virology and Immunobiology, Julius-Maximilians University Würzburg, Würzburg, Germany
| | - Christian Puta
- Department of Sports Medicine and Health Promotion, Friedrich-Schiller-University Jena, Jena, Germany; Center for Interdisciplinary Prevention of Diseases Related to Professional Activities, Friedrich-Schiller-University Jena, Jena, Germany; Center for Sepsis Control and Care (CSCC), Jena University Hospital/Friedrich-Schiller-University Jena, Jena, Germany
| | - Gabriela Riemekasten
- Department of Rheumatology and Clinical Immunology, University of Lübeck, Lübeck, Germany
| | - Wolfgang Ries
- Internal Medicine, Department of Nephrology, Diakonissenkrankenhaus, Flensburg, Germany
| | - Peter C Rowe
- Department of Pediatrics, Division of Adolescent and Young Adult Medicine, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Birgit Sawitzki
- Translational Immunology, Berlin Institute of Health (BIH) & Charité University Medicine, Berlin, Germany
| | - Yehuda Shoenfeld
- Zabludowicz Center for Autoimmune Diseases, Sheba Medical Center, Reichman University Herzelia, Israel
| | - Joachim L Schultze
- Systems Medicine, Deutsches Zentrum für Neurodegenerative Erkrankungen (DZNE), Bonn, Germany; Department for Genomics & Immunoregulation, Life and Medical Sciences Institute (LIMES), University of Bonn, Bonn, Germany; PRECISE Platform for Single Cell Genomics and Epigenomics, DZNE und Universität Bonn, Bonn, Germany
| | - Martina Seifert
- Institute of Medical Immunology, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt Universität zu Berlin and Berlin Institute of Health (BIH), Berlin, Germany; Berlin Institute of Health at Charité-Universitätsmedizin Berlin, BIH Center for Regenerative Therapies (BCRT), Charitéplatz 1, Berlin, Germany; DZHK (German Center for Cardiovascular Research), Partner Site, 10785 Berlin, Germany
| | - Nuno Sepúlveda
- Department of Mathematics & Information Science, Warsaw University of Technology, Warsaw, Poland.; CEAUL - Centro de Estatística e Aplicações da Universidade de Lisboa, Lisbon, Portugal
| | - Franziska Sotzny
- Institute of Medical Immunology, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt Universität zu Berlin and Berlin Institute of Health (BIH), Berlin, Germany
| | - Elisa Stein
- Institute of Medical Immunology, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt Universität zu Berlin and Berlin Institute of Health (BIH), Berlin, Germany
| | - Michael Stingl
- Department of Neurology, Zentrum Votivpark, Vienna, Austria
| | - Friederike Ufer
- Institute of Neuroimmunology and Multiple Sclerosis, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Christian Veauthier
- Interdisciplinary Center of Sleep Medicine, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Francisco Westermeier
- Institute of Biomedical Science, Department of Health Studies, FH, Joanneum University of Applied Sciences, Graz, Austria; Centro Integrativo de Biología y Química Aplicada (CIBQA), Universidad Bernardo O'Higgins, Santiago, Chile
| | - Klaus Wirth
- Institute of General Pharmacology and Toxicology, University Hospital Frankfurt, Goethe-University, Frankfurt, Germany
| | - Bernd Wolfarth
- Department of Sports Medicine, Charité-Universitätsmedizin Berlin, Humboldt University of Berlin, Germany
| | - Pawel Zalewski
- Department of Exercise Physiology and Functional Anatomy, Ludwik Rydygier Collegium Medicum in Bydgoszcz Nicolaus Copernicus University Toruń, Toruń, Poland; Department of Experimental and Clinical Physiology, Warsaw Medical University, Stefana Banacha 2a, Warszawa 02-097, Poland
| | - Uta Behrends
- Children's Hospital, School of Medicine, Technical University of Munich, Munich, Germany; German Center for Infection Research (DZIF), Berlin, Germany; AGV Research Unit Gene Vectors, Helmholtz Munich (HMGU), Munich, Germany
| | - Carmen Scheibenbogen
- Institute of Medical Immunology, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt Universität zu Berlin and Berlin Institute of Health (BIH), Berlin, Germany
| |
Collapse
|
185
|
Buck AM, Deitchman AN, Takahashi S, Lu S, Goldberg SA, Bodansky A, Kung A, Hoh R, Williams MC, Kerbleski M, Maison DP, Deveau TM, Munter SE, Lombardo J, Wrin T, Petropoulos CJ, Durstenfeld MS, Hsue PY, Daniel Kelly J, Greenhouse B, Martin JN, Deeks SG, Peluso MJ, Henrich TJ. The breadth of the neutralizing antibody response to original SARS-CoV-2 infection is linked to the presence of Long COVID symptoms. J Med Virol 2023; 95:e29216. [PMID: 37988251 PMCID: PMC10754238 DOI: 10.1002/jmv.29216] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2023] [Revised: 10/20/2023] [Accepted: 10/21/2023] [Indexed: 11/23/2023]
Abstract
The associations between longitudinal dynamics and the breadth of SARS-CoV-2 neutralizing antibody (nAb) response with various Long COVID phenotypes before vaccination are not known. The capacity of antibodies to cross-neutralize a variety of viral variants may be associated with ongoing pathology and persistent symptoms. We measured longitudinal neutralizing and cross-neutralizing antibody responses to pre- and post-SARS-CoV-2 Omicron variants in participants infected early in the COVID-19 pandemic, before widespread rollout of SARS-CoV-2 vaccines. Cross-sectional regression models adjusted for clinical covariates and longitudinal mixed-effects models were used to determine the impact of the breadth and rate of decay of neutralizing responses on the development of Long COVID symptoms, as well as Long COVID phenotypes. We identified several novel relationships between SARS-CoV-2 antibody neutralization and the presence of Long COVID symptoms. Specifically, we show that, although nAb responses to the original, infecting strain of SARS-CoV-2 were not associated with Long COVID in cross-sectional analyses, cross-neutralization ID50 levels to the Omicron BA.5 variant approximately 4 months following acute infection was independently and significantly associated with greater odds of Long COVID and with persistent gastrointestinal and neurological symptoms. Longitudinal modeling demonstrated significant associations in the overall levels and rates of decay of neutralization capacity with Long COVID phenotypes. A higher proportion of participants had antibodies capable of neutralizing Omicron BA.5 compared with BA.1 or XBB.1.5 variants. Our findings suggest that relationships between various immune responses and Long COVID are likely complex but may involve the breadth of antibody neutralization responses.
Collapse
Affiliation(s)
- Amanda M. Buck
- Division of Experimental Medicine, University of California San Francisco, San Francisco, CA, United States
| | - Amelia N. Deitchman
- Department of Clinical Pharmacy, University of California San Francisco, San Francisco, CA, United States
| | - Saki Takahashi
- Division of HIV, ID and Global Medicine, University of California San Francisco, San Francisco, CA, United States
| | - Scott Lu
- Department of Epidemiology and Biostatistics, University of California San Francisco, San Francisco, CA, United States
| | - Sarah A. Goldberg
- Department of Epidemiology and Biostatistics, University of California San Francisco, San Francisco, CA, United States
| | - Aaron Bodansky
- Division of Pediatric Critical Care Medicine, University of California San Francisco, San Francisco, CA, United States
| | - Andrew Kung
- Department of Biochemistry and Biophysics, University of California San Francisco, San Francisco, CA, United States
| | - Rebecca Hoh
- Division of HIV, ID and Global Medicine, University of California San Francisco, San Francisco, CA, United States
| | - Meghann C. Williams
- Division of HIV, ID and Global Medicine, University of California San Francisco, San Francisco, CA, United States
| | - Marian Kerbleski
- Division of HIV, ID and Global Medicine, University of California San Francisco, San Francisco, CA, United States
| | - David P. Maison
- Division of Experimental Medicine, University of California San Francisco, San Francisco, CA, United States
| | - Tyler-Marie Deveau
- Division of Experimental Medicine, University of California San Francisco, San Francisco, CA, United States
| | - Sadie E. Munter
- Division of Experimental Medicine, University of California San Francisco, San Francisco, CA, United States
| | - James Lombardo
- Division of HIV, ID and Global Medicine, University of California San Francisco, San Francisco, CA, United States
| | - Terri Wrin
- Monogram Biosciences, South San Francisco, CA, United States
| | | | - Matthew S. Durstenfeld
- Division of Cardiology, University of California San Francisco, San Francisco, CA, United States
| | - Priscilla Y. Hsue
- Division of Cardiology, University of California San Francisco, San Francisco, CA, United States
| | - J. Daniel Kelly
- Department of Epidemiology and Biostatistics, University of California San Francisco, San Francisco, CA, United States
| | - Bryan Greenhouse
- Division of HIV, ID and Global Medicine, University of California San Francisco, San Francisco, CA, United States
| | - Jeffrey N. Martin
- Department of Epidemiology and Biostatistics, University of California San Francisco, San Francisco, CA, United States
| | - Steven G. Deeks
- Division of HIV, ID and Global Medicine, University of California San Francisco, San Francisco, CA, United States
| | - Michael J. Peluso
- Division of HIV, ID and Global Medicine, University of California San Francisco, San Francisco, CA, United States
| | - Timothy J. Henrich
- Division of Experimental Medicine, University of California San Francisco, San Francisco, CA, United States
| |
Collapse
|
186
|
Li J, Zhou Y, Ma J, Zhang Q, Shao J, Liang S, Yu Y, Li W, Wang C. The long-term health outcomes, pathophysiological mechanisms and multidisciplinary management of long COVID. Signal Transduct Target Ther 2023; 8:416. [PMID: 37907497 PMCID: PMC10618229 DOI: 10.1038/s41392-023-01640-z] [Citation(s) in RCA: 38] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Revised: 08/04/2023] [Accepted: 09/04/2023] [Indexed: 11/02/2023] Open
Abstract
There have been hundreds of millions of cases of coronavirus disease 2019 (COVID-19), which is caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). With the growing population of recovered patients, it is crucial to understand the long-term consequences of the disease and management strategies. Although COVID-19 was initially considered an acute respiratory illness, recent evidence suggests that manifestations including but not limited to those of the cardiovascular, respiratory, neuropsychiatric, gastrointestinal, reproductive, and musculoskeletal systems may persist long after the acute phase. These persistent manifestations, also referred to as long COVID, could impact all patients with COVID-19 across the full spectrum of illness severity. Herein, we comprehensively review the current literature on long COVID, highlighting its epidemiological understanding, the impact of vaccinations, organ-specific sequelae, pathophysiological mechanisms, and multidisciplinary management strategies. In addition, the impact of psychological and psychosomatic factors is also underscored. Despite these crucial findings on long COVID, the current diagnostic and therapeutic strategies based on previous experience and pilot studies remain inadequate, and well-designed clinical trials should be prioritized to validate existing hypotheses. Thus, we propose the primary challenges concerning biological knowledge gaps and efficient remedies as well as discuss the corresponding recommendations.
Collapse
Affiliation(s)
- Jingwei Li
- Department of Pulmonary and Critical Care Medicine, State Key Laboratory of Respiratory Health and Multimorbidity, Med-X Center for Manufacturing, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, China
| | - Yun Zhou
- Department of Pulmonary and Critical Care Medicine, State Key Laboratory of Respiratory Health and Multimorbidity, Med-X Center for Manufacturing, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, China
| | - Jiechao Ma
- AI Lab, Deepwise Healthcare, Beijing, China
| | - Qin Zhang
- Department of Pulmonary and Critical Care Medicine, State Key Laboratory of Respiratory Health and Multimorbidity, Med-X Center for Manufacturing, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, China
- Department of Postgraduate Student, West China Hospital, West China School of Medicine, Sichuan University, Chengdu, China
| | - Jun Shao
- Department of Pulmonary and Critical Care Medicine, State Key Laboratory of Respiratory Health and Multimorbidity, Med-X Center for Manufacturing, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, China
| | - Shufan Liang
- Department of Pulmonary and Critical Care Medicine, State Key Laboratory of Respiratory Health and Multimorbidity, Med-X Center for Manufacturing, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, China
| | - Yizhou Yu
- Department of Computer Science, The University of Hong Kong, Hong Kong, China.
| | - Weimin Li
- Department of Pulmonary and Critical Care Medicine, State Key Laboratory of Respiratory Health and Multimorbidity, Med-X Center for Manufacturing, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, China.
| | - Chengdi Wang
- Department of Pulmonary and Critical Care Medicine, State Key Laboratory of Respiratory Health and Multimorbidity, Med-X Center for Manufacturing, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, China.
| |
Collapse
|
187
|
Nayyerabadi M, Fourcade L, Joshi SA, Chandrasekaran P, Chakravarti A, Massé C, Paul ML, Houle J, Boubekeur AM, DuSablon C, Boudreau V, Bovan D, Darbinian E, Coleman EA, Vinci S, Routy JP, Hétu PO, Poudrier J, Falcone EL. Vaccination after developing long COVID: Impact on clinical presentation, viral persistence, and immune responses. Int J Infect Dis 2023; 136:136-145. [PMID: 37717649 DOI: 10.1016/j.ijid.2023.09.006] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2023] [Revised: 09/01/2023] [Accepted: 09/11/2023] [Indexed: 09/19/2023] Open
Abstract
BACKGROUND Vaccination protects against severe COVID-19 manifestations. For those with post-COVID-19 conditions (PCC) or long COVID, the impact of COVID-19 vaccination on the evolution of symptoms, immune responses, and viral persistence is unclear. METHODS In this prospective observational cohort study, we evaluated the number of PCC symptoms, affected organ systems, and psychological well-being scores before and after patients with PCC received COVID-19 vaccination. We simultaneously evaluated biomarkers of systemic inflammation and levels of plasma cytokines/chemokines. We measured plasma and intracellular levels of SARS-CoV-2 antigens, and immunoreactivity to SARS-CoV-2 antigens in blood. RESULTS COVID-19 vaccination was associated with decreases in number of PCC symptoms (pre-vaccination: 6.56 ± 3.1 vs post-vaccination: 3.92 ± 4.02; P <0.001) and affected organ systems (pre-vaccination: 3.19 ± 1.04 vs post-vaccination: 1.89 ± 1.12; P <0.001), and increases in World Health Organization (WHO)-5 Well-Being Index Scores (pre-vaccination: 42.67 ± 22.76 vs post-vaccination: 56.15 ± 22.83; P <0.001). Patients with PCC also had significantly decreased levels of several pro-inflammatory plasma cytokines/chemokines after COVID-19 vaccination including sCD40L, GRO-⍺, macrophage inflammatory protein (MIP)-1⍺, interleukin (IL)-12p40, G-colony stimulating factor (CSF), M-CSF, IL-1β, and stem cell factor (SCF). PCC participants presented a certain level of immunoreactivity toward SARS-CoV-2, that was boosted with vaccination. SARS-CoV-2 S1 antigen persisted in the blood of PCC participants, mostly in non-classical monocytes, regardless of participants receiving vaccination. CONCLUSIONS Our study shows higher pro-inflammatory responses associated with PCC symptoms and brings forward a possible role for vaccination in mitigating PCC symptoms by decreasing systemic inflammation. We also observed persistence of viral products independent of vaccination that could be involved in perpetuating inflammation through non-classical monocytes.
Collapse
Affiliation(s)
- Maryam Nayyerabadi
- Center for Inflammation, Immunity and Infectious Diseases, Montreal Clinical Research Institute (IRCM), Montreal, QC, Canada
| | - Lyvia Fourcade
- Center for Inflammation, Immunity and Infectious Diseases, Montreal Clinical Research Institute (IRCM), Montreal, QC, Canada
| | - Swarali A Joshi
- Center for Inflammation, Immunity and Infectious Diseases, Montreal Clinical Research Institute (IRCM), Montreal, QC, Canada; Center for Commercialization of Regenerative Medicine (CCRM), Toronto, ON, Canada
| | | | - Arpita Chakravarti
- Department of Microbiology, Infectious Diseases and Immunology, Université de Montréal, Montreal, QC, Canada; Department of Infectious Diseases and Medical Microbiology, Centre Hospitalier de l'Université de Montréal (CHUM), Montreal, QC, Canada
| | - Chantal Massé
- Center for Inflammation, Immunity and Infectious Diseases, Montreal Clinical Research Institute (IRCM), Montreal, QC, Canada
| | - Marie-Lorna Paul
- Center for Inflammation, Immunity and Infectious Diseases, Montreal Clinical Research Institute (IRCM), Montreal, QC, Canada; Morphocell Technologies Inc., Montreal, QC, Canada
| | - Joanie Houle
- Center for Inflammation, Immunity and Infectious Diseases, Montreal Clinical Research Institute (IRCM), Montreal, QC, Canada
| | - Amina M Boubekeur
- Center for Inflammation, Immunity and Infectious Diseases, Montreal Clinical Research Institute (IRCM), Montreal, QC, Canada
| | - Charlotte DuSablon
- Center for Inflammation, Immunity and Infectious Diseases, Montreal Clinical Research Institute (IRCM), Montreal, QC, Canada
| | - Valérie Boudreau
- Center for Cardiometabolic Health, Montreal Clinical Research Institute (IRCM), Montreal, QC, Canada
| | - Danijela Bovan
- Center for Cardiometabolic Health, Montreal Clinical Research Institute (IRCM), Montreal, QC, Canada
| | - Emma Darbinian
- Center for Inflammation, Immunity and Infectious Diseases, Montreal Clinical Research Institute (IRCM), Montreal, QC, Canada
| | - Emilia Aïsha Coleman
- Center for Inflammation, Immunity and Infectious Diseases, Montreal Clinical Research Institute (IRCM), Montreal, QC, Canada
| | - Sandra Vinci
- Center for Inflammation, Immunity and Infectious Diseases, Montreal Clinical Research Institute (IRCM), Montreal, QC, Canada
| | - Jean-Pierre Routy
- Department of Medicine, McGill University Health Center (MUHC), Montreal, QC, Canada
| | - Pierre-Olivier Hétu
- Department of Laboratory Medicine, Centre Hospitalier de l'Université de Montréal (CHUM), Montreal, QC, Canada; Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Montreal, QC, Canada
| | - Johanne Poudrier
- Center for Inflammation, Immunity and Infectious Diseases, Montreal Clinical Research Institute (IRCM), Montreal, QC, Canada; Department of Microbiology, Infectious Diseases and Immunology, Université de Montréal, Montreal, QC, Canada
| | - Emilia Liana Falcone
- Center for Inflammation, Immunity and Infectious Diseases, Montreal Clinical Research Institute (IRCM), Montreal, QC, Canada; Department of Microbiology, Infectious Diseases and Immunology, Université de Montréal, Montreal, QC, Canada; Department of Infectious Diseases and Medical Microbiology, Centre Hospitalier de l'Université de Montréal (CHUM), Montreal, QC, Canada; Department of Medicine, Université de Montréal, Montreal, QC, Canada.
| |
Collapse
|
188
|
VanElzakker MB, Bues HF, Brusaferri L, Kim M, Saadi D, Ratai EM, Dougherty DD, Loggia ML. Neuroinflammation in post-acute sequelae of COVID-19 (PASC) as assessed by [ 11C]PBR28 PET correlates with vascular disease measures. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.10.19.563117. [PMID: 37905031 PMCID: PMC10614970 DOI: 10.1101/2023.10.19.563117] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/02/2023]
Abstract
The COVID-19 pandemic caused by SARS-CoV-2 has triggered a consequential public health crisis of post-acute sequelae of COVID-19 (PASC), sometimes referred to as long COVID. The mechanisms of the heterogeneous persistent symptoms and signs that comprise PASC are under investigation, and several studies have pointed to the central nervous and vascular systems as being potential sites of dysfunction. In the current study, we recruited individuals with PASC with diverse symptoms, and examined the relationship between neuroinflammation and circulating markers of vascular dysfunction. We used [11C]PBR28 PET neuroimaging, a marker of neuroinflammation, to compare 12 PASC individuals versus 43 normative healthy controls. We found significantly increased neuroinflammation in PASC versus controls across a wide swath of brain regions including midcingulate and anterior cingulate cortex, corpus callosum, thalamus, basal ganglia, and at the boundaries of ventricles. We also collected and analyzed peripheral blood plasma from the PASC individuals and found significant positive correlations between neuroinflammation and several circulating analytes related to vascular dysfunction. These results suggest that an interaction between neuroinflammation and vascular health may contribute to common symptoms of PASC.
Collapse
Affiliation(s)
- Michael B VanElzakker
- Division of Neurotherapeutics, Department of Psychiatry, Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- PolyBio Research Foundation, Medford, MA, USA
| | - Hannah F Bues
- Division of Neurotherapeutics, Department of Psychiatry, Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Ludovica Brusaferri
- Department of Radiology, Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Department of Computer Science And Informatics, School of Engineering, London South Bank University, London, UK
| | - Minhae Kim
- Department of Radiology, Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Deena Saadi
- Division of Neurotherapeutics, Department of Psychiatry, Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Eva-Maria Ratai
- Department of Radiology, Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Darin D Dougherty
- Division of Neurotherapeutics, Department of Psychiatry, Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Marco L Loggia
- Department of Radiology, Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
189
|
Nair S, Nova-Lamperti E, Labarca G, Kulasinghe A, Short KR, Carrión F, Salomon C. Genomic communication via circulating extracellular vesicles and long-term health consequences of COVID-19. J Transl Med 2023; 21:709. [PMID: 37817137 PMCID: PMC10563316 DOI: 10.1186/s12967-023-04552-2] [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: 05/06/2023] [Accepted: 09/22/2023] [Indexed: 10/12/2023] Open
Abstract
COVID-19 continues to affect an unprecedented number of people with the emergence of new variants posing a serious challenge to global health. There is an expansion of knowledge in understanding the pathogenesis of Coronavirus disease 2019 (COVID-19), caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), and the impact of the acute disease on multiple organs. In addition, growing evidence reports that the impact of COVID-19 on different organs persists long after the recovery phase of the disease, leading to long-term consequences of COVID-19. These long-term consequences involve pulmonary as well as extra-pulmonary sequelae of the disease. Noteably, recent research has shown a potential association between COVID-19 and change in the molecular cargo of extracellular vesicles (EVs). EVs are vesicles released by cells and play an important role in cell communication by transfer of bioactive molecules between cells. Emerging evidence shows a strong link between EVs and their molecular cargo, and regulation of metabolism in health and disease. This review focuses on current knowledge about EVs and their potential role in COVID-19 pathogenesis, their current and future implications as tools for biomarker and therapeutic development and their possible effects on long-term impact of COVID-19.
Collapse
Affiliation(s)
- Soumyalekshmi Nair
- Translational Extracellular Vesicles in Obstetrics and Gynae-Oncology Group, UQ Centre for Clinical Research, Royal Brisbane and Women's Hospital, Faculty of Medicine, The University of Queensland, Brisbane, Qld, 4072, Australia
| | - Estefania Nova-Lamperti
- Molecular and Translational Immunology Laboratory, Clinical Biochemistry and Immunology Department, Pharmacy Faculty, Universidad de Concepción, Concepción, Chile
| | - Gonzalo Labarca
- Molecular and Translational Immunology Laboratory, Clinical Biochemistry and Immunology Department, Pharmacy Faculty, Universidad de Concepción, Concepción, Chile
| | - Arutha Kulasinghe
- Frazer Institute, Faculty of Medicine, The University of Queensland, Brisbane, Qld, 4102, Australia
| | - Kirsty R Short
- School of Chemistry and Molecular Biosciences, The University of Queensland, Brisbane, Australia
| | - Flavio Carrión
- Departamento de Investigación, Postgrado y Educación Continua (DIPEC), Facultad de Ciencias de la Salud, Universidad del Alba, Santiago, Chile.
| | - Carlos Salomon
- Translational Extracellular Vesicles in Obstetrics and Gynae-Oncology Group, UQ Centre for Clinical Research, Royal Brisbane and Women's Hospital, Faculty of Medicine, The University of Queensland, Brisbane, Qld, 4072, Australia.
- Departamento de Investigación, Postgrado y Educación Continua (DIPEC), Facultad de Ciencias de la Salud, Universidad del Alba, Santiago, Chile.
| |
Collapse
|
190
|
Pileggi CA, Parmar G, Elkhatib H, Stewart CM, Alecu I, Côté M, Bennett SA, Sandhu JK, Cuperlovic-Culf M, Harper ME. The SARS-CoV-2 spike glycoprotein interacts with MAO-B and impairs mitochondrial energetics. CURRENT RESEARCH IN NEUROBIOLOGY 2023; 5:100112. [PMID: 38020812 PMCID: PMC10663135 DOI: 10.1016/j.crneur.2023.100112] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2023] [Revised: 08/21/2023] [Accepted: 09/25/2023] [Indexed: 12/01/2023] Open
Abstract
SARS-CoV-2 infection is associated with both acute and post-acute neurological symptoms. Emerging evidence suggests that SARS-CoV-2 can alter mitochondrial metabolism, suggesting that changes in brain metabolism may contribute to the development of acute and post-acute neurological complications. Monoamine oxidase B (MAO-B) is a flavoenzyme located on the outer mitochondrial membrane that catalyzes the oxidative deamination of monoamine neurotransmitters. Computational analyses have revealed high similarity between the SARS-CoV-2 spike glycoprotein receptor binding domain on the ACE2 receptor and MAO-B, leading to the hypothesis that SARS-CoV-2 spike glycoprotein may alter neurotransmitter metabolism by interacting with MAO-B. Our results empirically establish that the SARS-CoV-2 spike glycoprotein interacts with MAO-B, leading to increased MAO-B activity in SH-SY5Y neuron-like cells. Common to neurodegenerative disease pathophysiological mechanisms, we also demonstrate that the spike glycoprotein impairs mitochondrial bioenergetics, induces oxidative stress, and perturbs the degradation of depolarized aberrant mitochondria through mitophagy. Our findings also demonstrate that SH-SY5Y neuron-like cells expressing the SARS-CoV-2 spike protein were more susceptible to MPTP-induced necrosis, likely necroptosis. Together, these results reveal novel mechanisms that may contribute to SARS-CoV-2-induced neurodegeneration.
Collapse
Affiliation(s)
- Chantal A. Pileggi
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, 451 Smyth Road, Ottawa, ON, K1H 8M5, Canada
- Ottawa Institute of Systems Biology, University of Ottawa, ON, K1H 8M5, Canada
- National Research Council of Canada, Digital Technologies Research Centre, 1200 Montreal Road, Ottawa, ON, K1A 0R6, Canada
| | - Gaganvir Parmar
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, 451 Smyth Road, Ottawa, ON, K1H 8M5, Canada
- Ottawa Institute of Systems Biology, University of Ottawa, ON, K1H 8M5, Canada
| | - Hussein Elkhatib
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, 451 Smyth Road, Ottawa, ON, K1H 8M5, Canada
- Ottawa Institute of Systems Biology, University of Ottawa, ON, K1H 8M5, Canada
| | - Corina M. Stewart
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, 451 Smyth Road, Ottawa, ON, K1H 8M5, Canada
- Ottawa Institute of Systems Biology, University of Ottawa, ON, K1H 8M5, Canada
- Current Address: Faculty of Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Irina Alecu
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, 451 Smyth Road, Ottawa, ON, K1H 8M5, Canada
- Ottawa Institute of Systems Biology, University of Ottawa, ON, K1H 8M5, Canada
- Neural Regeneration Laboratory, Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, 451 Smyth Road, Ottawa, ON, K1H 8M5, Canada
| | - Marceline Côté
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, 451 Smyth Road, Ottawa, ON, K1H 8M5, Canada
- Ottawa Institute of Systems Biology, University of Ottawa, ON, K1H 8M5, Canada
- Centre for Infection, Immunity and Inflammation, University of Ottawa, ON, K1H 8M5, Canada
| | - Steffany A.L. Bennett
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, 451 Smyth Road, Ottawa, ON, K1H 8M5, Canada
- Ottawa Institute of Systems Biology, University of Ottawa, ON, K1H 8M5, Canada
- Neural Regeneration Laboratory, Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, 451 Smyth Road, Ottawa, ON, K1H 8M5, Canada
| | - Jagdeep K. Sandhu
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, 451 Smyth Road, Ottawa, ON, K1H 8M5, Canada
- Ottawa Institute of Systems Biology, University of Ottawa, ON, K1H 8M5, Canada
- Centre for Infection, Immunity and Inflammation, University of Ottawa, ON, K1H 8M5, Canada
- Human Health Therapeutics Research Centre, National Research Council Canada, 1200 Montreal Road, Ottawa, ON, K1A 0R6, Canada
| | - Miroslava Cuperlovic-Culf
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, 451 Smyth Road, Ottawa, ON, K1H 8M5, Canada
- Ottawa Institute of Systems Biology, University of Ottawa, ON, K1H 8M5, Canada
- National Research Council of Canada, Digital Technologies Research Centre, 1200 Montreal Road, Ottawa, ON, K1A 0R6, Canada
| | - Mary-Ellen Harper
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, 451 Smyth Road, Ottawa, ON, K1H 8M5, Canada
- Ottawa Institute of Systems Biology, University of Ottawa, ON, K1H 8M5, Canada
- Centre for Infection, Immunity and Inflammation, University of Ottawa, ON, K1H 8M5, Canada
| |
Collapse
|
191
|
Manfredelli D, Pariano M, Costantini C, Graziani A, Bozza S, Romani L, Puccetti P, Talesa VN, Antognelli C. Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) Spike Protein S1 Induces Methylglyoxal-Derived Hydroimidazolone/Receptor for Advanced Glycation End Products (MG-H1/RAGE) Activation to Promote Inflammation in Human Bronchial BEAS-2B Cells. Int J Mol Sci 2023; 24:14868. [PMID: 37834316 PMCID: PMC10573269 DOI: 10.3390/ijms241914868] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2023] [Revised: 09/28/2023] [Accepted: 10/02/2023] [Indexed: 10/15/2023] Open
Abstract
The pathogenesis of coronavirus disease 2019 (COVID-19) is associated with a hyperinflammatory response. The mechanisms of SARS-CoV-2-induced inflammation are scantly known. Methylglyoxal (MG) is a glycolysis-derived byproduct endowed with a potent glycating action, leading to the formation of advanced glycation end products (AGEs), the main one being MG-H1. MG-H1 exerts strong pro-inflammatory effects, frequently mediated by the receptor for AGEs (RAGE). Here, we investigated the involvement of the MG-H1/RAGE axis as a potential novel mechanism in SARS-CoV-2-induced inflammation by resorting to human bronchial BEAS-2B and alveolar A549 epithelial cells, expressing different levels of the ACE2 receptor (R), exposed to SARS-CoV-2 spike protein 1 (S1). Interestingly, we found in BEAS-2B cells that do not express ACE2-R that S1 exerted a pro-inflammatory action through a novel MG-H1/RAGE-based pathway. MG-H1 levels, RAGE and IL-1β expression levels in nasopharyngeal swabs from SARS-CoV-2-positive and -negative individuals, as well as glyoxalase 1 expression, the major scavenging enzyme of MG, seem to support the results obtained in vitro. Altogether, our findings reveal a novel mechanism involved in the inflammation triggered by S1, paving the way for the study of the MG-H1/RAGE inflammatory axis in SARS-CoV-2 infection as a potential therapeutic target to mitigate COVID-19-associated pathogenic inflammation.
Collapse
Affiliation(s)
- Dominga Manfredelli
- Department of Medicine and Surgery, Bioscience and Medical Embryology Division, University of Perugia, L. Severi Square, 06129 Perugia, Italy; (D.M.); (M.P.); (V.N.T.)
| | - Marilena Pariano
- Department of Medicine and Surgery, Bioscience and Medical Embryology Division, University of Perugia, L. Severi Square, 06129 Perugia, Italy; (D.M.); (M.P.); (V.N.T.)
| | - Claudio Costantini
- Department of Medicine and Surgery, Pathology Division, University of Perugia, L. Severi Square, 06129 Perugia, Italy; (C.C.); (L.R.)
| | - Alessandro Graziani
- Department of Medicine and Surgery, Microbiology and Clinical Microbiology Division, University of Perugia, L. Severi Square, 06129 Perugia, Italy; (A.G.); (S.B.)
| | - Silvia Bozza
- Department of Medicine and Surgery, Microbiology and Clinical Microbiology Division, University of Perugia, L. Severi Square, 06129 Perugia, Italy; (A.G.); (S.B.)
| | - Luigina Romani
- Department of Medicine and Surgery, Pathology Division, University of Perugia, L. Severi Square, 06129 Perugia, Italy; (C.C.); (L.R.)
| | - Paolo Puccetti
- Department of Medicine and Surgery, Pharmacology Division, University of Perugia, L. Severi Square, 06129 Perugia, Italy;
| | - Vincenzo Nicola Talesa
- Department of Medicine and Surgery, Bioscience and Medical Embryology Division, University of Perugia, L. Severi Square, 06129 Perugia, Italy; (D.M.); (M.P.); (V.N.T.)
| | - Cinzia Antognelli
- Department of Medicine and Surgery, Bioscience and Medical Embryology Division, University of Perugia, L. Severi Square, 06129 Perugia, Italy; (D.M.); (M.P.); (V.N.T.)
| |
Collapse
|
192
|
Allan-Blitz LT, Akbari O, Kojima N, Saavedra E, Chellamuthu P, Denny N, MacMullan MA, Hess V, Shacreaw M, Brobeck M, Turner F, Slepnev VI, Ibrayeva A, Klausner JD. Unique immune and inflammatory cytokine profiles may define long COVID syndrome. Clin Exp Med 2023; 23:2925-2930. [PMID: 37061998 PMCID: PMC10105906 DOI: 10.1007/s10238-023-01065-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2022] [Accepted: 04/02/2023] [Indexed: 04/17/2023]
Abstract
PURPOSE Long COVID is estimated to occur in 5-10% of individuals after acute SARS-CoV-2 infection. However, the pathophysiology driving the disease process is poorly understood. METHODS We evaluated urine and plasma inflammatory and immune cytokine profiles in 33 individuals with long COVID compared to 33 who were asymptomatic and recovered, and 34 without prior infection. RESULTS Mean urinary leukotriene E4 was significantly elevated among individuals with long COVID compared to asymptomatic and recovered individuals (mean difference 774.2 pg/mL; SD 335.7) and individuals without prior SARS-CoV-2 infection (mean difference 503.1 pg/ml; SD 467.7). Plasma chemokine ligand 6 levels were elevated among individuals with long COVID compared to individuals with no prior SARS-CoV-2 infection (mean difference 0.59 units; SD 0.42). We found no significant difference in angiotensin-converting enzyme 2 antibody levels. Plasma tumor necrosis factor receptor-associated factor 2 (TRAF2) levels were reduced among individuals with long COVID compared to individuals who were asymptomatic and recovered (mean difference = 0.6 units, SD 0.46). Similarly, the mean level of Sarcoma Homology 2-B adapter protein 3 was 3.3 units (SD 1.24) among individuals with long COVID, lower than 4.2 units (SD 1.1) among individuals with recovered, asymptomatic COVID. CONCLUSION Our findings suggest that further studies should be conducted to evaluate the role of leukotriene E4 as a potential biomarker for a diagnostic test. Furthermore, based on reductions in TRAF2, long COVID may be driven in part by impaired TRAF2-dependent immune-mediated inflammation and potentially immune exhaustion.
Collapse
Affiliation(s)
- Lao-Tzu Allan-Blitz
- Division of Global Health Equity: Department of Medicine, Brigham and Women’s Hospital, 75 Francis Street, Boston, MA 02115 USA
| | - Omid Akbari
- Department of Population and Public Health Sciences, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033 USA
| | - Noah Kojima
- Department of Medicine, University of California Los Angeles, Los Angeles, CA USA
| | | | | | | | | | | | | | | | | | | | - Albina Ibrayeva
- Department of Medicine, University of California Los Angeles, Los Angeles, CA USA
| | - Jeffrey D. Klausner
- Department of Population and Public Health Sciences, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033 USA
| |
Collapse
|
193
|
Connors JM, Ariëns RAS. Uncertainties about the roles of anticoagulation and microclots in postacute sequelae of severe acute respiratory syndrome coronavirus 2 infection. J Thromb Haemost 2023; 21:2697-2701. [PMID: 37495081 DOI: 10.1016/j.jtha.2023.07.012] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2023] [Revised: 07/13/2023] [Accepted: 07/14/2023] [Indexed: 07/28/2023]
Abstract
Three years after the start of the pandemic, approaches to the prevention and treatment of acute COVID-19 have been established. However, postacute sequelae of SARS-CoV-2 infection (PASC) is now the focus of current investigations searching for the etiology of the symptoms and developing diagnostic and treatment strategies, as up to 10% of those with acute COVID-19 will go on to develop PASC, a significant public health burden. Some have suggested that ongoing microvascular thrombosis and microclots may play a role in the persistent sequelae of COVID-19 infection. Treatments are being given to address the presumed role of thrombosis in PASC, and some suggest that randomized controlled trials of anticoagulants in patients with PASC should be performed. In this Forum article, we focus on findings from patients with PASC that have led to the suggestion of using anticoagulants and discuss alternative considerations.
Collapse
Affiliation(s)
- Jean M Connors
- Hematology Division, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA.
| | - Robert A S Ariëns
- Discovery and Translational Science Department, Leeds Institute of Cardiovascular and Metabolic Medicine, University of Leeds, UK
| |
Collapse
|
194
|
Altmann DM, Whettlock EM, Liu S, Arachchillage DJ, Boyton RJ. The immunology of long COVID. Nat Rev Immunol 2023; 23:618-634. [PMID: 37433988 DOI: 10.1038/s41577-023-00904-7] [Citation(s) in RCA: 178] [Impact Index Per Article: 89.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/02/2023] [Indexed: 07/13/2023]
Abstract
Long COVID is the patient-coined term for the disease entity whereby persistent symptoms ensue in a significant proportion of those who have had COVID-19, whether asymptomatic, mild or severe. Estimated numbers vary but the assumption is that, of all those who had COVID-19 globally, at least 10% have long COVID. The disease burden spans from mild symptoms to profound disability, the scale making this a huge, new health-care challenge. Long COVID will likely be stratified into several more or less discrete entities with potentially distinct pathogenic pathways. The evolving symptom list is extensive, multi-organ, multisystem and relapsing-remitting, including fatigue, breathlessness, neurocognitive effects and dysautonomia. A range of radiological abnormalities in the olfactory bulb, brain, heart, lung and other sites have been observed in individuals with long COVID. Some body sites indicate the presence of microclots; these and other blood markers of hypercoagulation implicate a likely role of endothelial activation and clotting abnormalities. Diverse auto-antibody (AAB) specificities have been found, as yet without a clear consensus or correlation with symptom clusters. There is support for a role of persistent SARS-CoV-2 reservoirs and/or an effect of Epstein-Barr virus reactivation, and evidence from immune subset changes for broad immune perturbation. Thus, the current picture is one of convergence towards a map of an immunopathogenic aetiology of long COVID, though as yet with insufficient data for a mechanistic synthesis or to fully inform therapeutic pathways.
Collapse
Affiliation(s)
- Daniel M Altmann
- Department of Immunology and Inflammation, Imperial College London, Hammersmith Hospital, London, UK.
| | - Emily M Whettlock
- Department of Infectious Disease, Imperial College London, Hammersmith Hospital, London, UK
| | - Siyi Liu
- Department of Immunology and Inflammation, Imperial College London, Hammersmith Hospital, London, UK
- Department of Infectious Disease, Imperial College London, Hammersmith Hospital, London, UK
| | - Deepa J Arachchillage
- Centre for Haematology, Department of Immunology and Inflammation, Imperial College London, Hammersmith Hospital, London, UK
- Department of Haematology, Imperial College Healthcare NHS Trust, London, UK
| | - Rosemary J Boyton
- Department of Infectious Disease, Imperial College London, Hammersmith Hospital, London, UK
- Lung Division, Royal Brompton and Harefield Hospitals, Guy's and St Thomas' NHS Foundation Trust, London, UK
| |
Collapse
|
195
|
Allan-Blitz LT, Hu H, Klausner JD. What Long COVID Prevention Strategies Suggest About Its Pathophysiology. Open Forum Infect Dis 2023; 10:ofad466. [PMID: 37822557 PMCID: PMC10563786 DOI: 10.1093/ofid/ofad466] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2023] [Accepted: 09/06/2023] [Indexed: 10/13/2023] Open
Affiliation(s)
- Lao-Tzu Allan-Blitz
- Division of Global Health Equity, Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts, USA
| | - Howard Hu
- Department of Population and Public Health Sciences, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| | - Jeffrey D Klausner
- Department of Population and Public Health Sciences, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| |
Collapse
|
196
|
Burns MD, Bartsch YC, Davis JP, Boribong BP, Loiselle M, Kang J, Kane AS, Edlow AG, Fasano A, Alter G, Yonker LM. Long-term humoral signatures following acute pediatric COVID-19 and Multisystem Inflammatory Syndrome in Children. Pediatr Res 2023; 94:1327-1334. [PMID: 37173406 PMCID: PMC10176275 DOI: 10.1038/s41390-023-02627-w] [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: 11/11/2022] [Revised: 03/21/2023] [Accepted: 03/30/2023] [Indexed: 05/15/2023]
Abstract
BACKGROUND Although most children experience mild symptoms during acute SARS-CoV-2 infection, some develop the severe post-COVID-19 complication, Multisystem Inflammatory Syndrome in Children (MIS-C). While acute presentations of COVID-19 and MIS-C have been well immunophenotyped, little is known about the lasting immune profile in children after acute illness. METHODS Children 2 months-20 years of age presenting with either acute COVID-19 (n = 9) or MIS-C (n = 12) were enrolled in a Pediatric COVID-19 Biorepository at a single medical center. We deeply profiled humoral immune responses and circulating cytokines following pediatric COVID-19 and MIS-C. RESULTS Twenty-one children and young adults provided blood samples at both acute presentation and 6-month follow-up (mean: 6.5 months; standard deviation: 1.77 months). Pro-inflammatory cytokine elevations resolved after both acute COVID-19 and MIS-C. Humoral profiles continue to mature after acute COVID-19, displaying decreasing IgM and increasing IgG over time, as well as stronger effector functions, including antibody-dependent monocyte activation. In contrast, MIS-C immune signatures, especially anti-Spike IgG1, diminished over time. CONCLUSIONS Here, we show the mature immune signature after pediatric COVID-19 and MIS-C, displaying resolving inflammation with recalibration of the humoral responses. These humoral profiles highlight immune activation and vulnerabilities over time in these pediatric post-infectious cohorts. IMPACT The pediatric immune profile matures after both COVID-19 and MIS-C, suggesting a diversified anti-SARS-CoV-2 antibody response after resolution of acute illness. While pro-inflammatory cytokine responses resolve in the months following acute infection in both conditions, antibody-activated responses remain relatively heightened in convalescent COVID-19. These data may inform long-term immunoprotection from reinfection in children with past SARS-CoV-2 infections or MIS-C.
Collapse
Affiliation(s)
- Madeleine D Burns
- Mucosal Immunology and Biology Research Center, Massachusetts General Hospital, Boston, MA, USA
- Department of Pediatrics, Massachusetts General Hospital for Children, Boston, MA, USA
| | - Yannic C Bartsch
- Harvard Medical School, Boston, MA, USA
- Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology and Harvard, Cambridge, MA, USA
| | - Jameson P Davis
- Mucosal Immunology and Biology Research Center, Massachusetts General Hospital, Boston, MA, USA
- Department of Pediatrics, Massachusetts General Hospital for Children, Boston, MA, USA
| | - Brittany P Boribong
- Mucosal Immunology and Biology Research Center, Massachusetts General Hospital, Boston, MA, USA
- Department of Pediatrics, Massachusetts General Hospital for Children, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Maggie Loiselle
- Mucosal Immunology and Biology Research Center, Massachusetts General Hospital, Boston, MA, USA
- Department of Pediatrics, Massachusetts General Hospital for Children, Boston, MA, USA
| | - Jaewon Kang
- Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology and Harvard, Cambridge, MA, USA
| | - Abigail S Kane
- Mucosal Immunology and Biology Research Center, Massachusetts General Hospital, Boston, MA, USA
- Department of Pediatrics, Massachusetts General Hospital for Children, Boston, MA, USA
| | - Andrea G Edlow
- Harvard Medical School, Boston, MA, USA
- Division of Maternal-Fetal Medicine, Department of Obstetrics and Gynecology, Massachusetts General Hospital, Boston, MA, USA
- Vincent Center for Reproductive Biology, Massachusetts General Hospital, Boston, MA, USA
| | - Alessio Fasano
- Mucosal Immunology and Biology Research Center, Massachusetts General Hospital, Boston, MA, USA
- Department of Pediatrics, Massachusetts General Hospital for Children, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Galit Alter
- Harvard Medical School, Boston, MA, USA
- Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology and Harvard, Cambridge, MA, USA
| | - Lael M Yonker
- Mucosal Immunology and Biology Research Center, Massachusetts General Hospital, Boston, MA, USA.
- Department of Pediatrics, Massachusetts General Hospital for Children, Boston, MA, USA.
- Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
197
|
Pérez Catalán I, Gascón Buj A, García Muñoz S, Gómez Alfaro I, Roig Martí C, Torres García M, Reig Valero R, Ferrando Piqueres R, Mateu Campos L, Ramos Rincón JM, Usó Blasco J. Nirmatrelvir/ritonavir as a possible treatment for Long-COVID. REVISTA ESPANOLA DE QUIMIOTERAPIA : PUBLICACION OFICIAL DE LA SOCIEDAD ESPANOLA DE QUIMIOTERAPIA 2023; 36:545-546. [PMID: 37357766 PMCID: PMC10586732 DOI: 10.37201/req/028.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/21/2023] [Revised: 04/24/2023] [Accepted: 05/08/2023] [Indexed: 06/27/2023]
Affiliation(s)
| | | | | | | | - C Roig Martí
- Celia Roig Martí, Internal Medicine, General University Hospital of Castellón, Castellón de la Plana, Spain.
| | | | | | | | | | | | | |
Collapse
|
198
|
Proal AD, VanElzakker MB, Aleman S, Bach K, Boribong BP, Buggert M, Cherry S, Chertow DS, Davies HE, Dupont CL, Deeks SG, Eimer W, Ely EW, Fasano A, Freire M, Geng LN, Griffin DE, Henrich TJ, Iwasaki A, Izquierdo-Garcia D, Locci M, Mehandru S, Painter MM, Peluso MJ, Pretorius E, Price DA, Putrino D, Scheuermann RH, Tan GS, Tanzi RE, VanBrocklin HF, Yonker LM, Wherry EJ. SARS-CoV-2 reservoir in post-acute sequelae of COVID-19 (PASC). Nat Immunol 2023; 24:1616-1627. [PMID: 37667052 DOI: 10.1038/s41590-023-01601-2] [Citation(s) in RCA: 123] [Impact Index Per Article: 61.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Accepted: 07/18/2023] [Indexed: 09/06/2023]
Abstract
Millions of people are suffering from Long COVID or post-acute sequelae of COVID-19 (PASC). Several biological factors have emerged as potential drivers of PASC pathology. Some individuals with PASC may not fully clear the coronavirus SARS-CoV-2 after acute infection. Instead, replicating virus and/or viral RNA-potentially capable of being translated to produce viral proteins-persist in tissue as a 'reservoir'. This reservoir could modulate host immune responses or release viral proteins into the circulation. Here we review studies that have identified SARS-CoV-2 RNA/protein or immune responses indicative of a SARS-CoV-2 reservoir in PASC samples. Mechanisms by which a SARS-CoV-2 reservoir may contribute to PASC pathology, including coagulation, microbiome and neuroimmune abnormalities, are delineated. We identify research priorities to guide the further study of a SARS-CoV-2 reservoir in PASC, with the goal that clinical trials of antivirals or other therapeutics with potential to clear a SARS-CoV-2 reservoir are accelerated.
Collapse
Affiliation(s)
- Amy D Proal
- PolyBio Research Foundation, Medford, MA, USA.
| | - Michael B VanElzakker
- PolyBio Research Foundation, Medford, MA, USA
- Division of Neurotherapeutics, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Soo Aleman
- Dept of Infectious Diseases and Unit of Post-Covid Huddinge, Karolinska University Hospital, Stockholm, Sweden
| | - Katie Bach
- PolyBio Research Foundation, Medford, MA, USA
- Nonresident Senior Fellow, Brookings Institution, Washington, DC, USA
| | - Brittany P Boribong
- Department of Pediatrics, Massachusetts General Hospital, Boston, MA, USA
- Mucosal Immunology and Biology Research Center, Massachusetts General Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Marcus Buggert
- Center for Infectious Medicine, Department of Medicine Huddinge, Karolinska Institutet, Huddinge, Sweden
| | - Sara Cherry
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, UPENN, Philadelphia, PA, USA
| | - Daniel S Chertow
- Emerging Pathogens Section, Critical Care Medicine Department, Clinical Center, National Institutes of Health, Bethesda, MD, USA
- Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Helen E Davies
- Department of Respiratory Medicine, University Hospital Llandough, Cardiff University School of Medicine, University Hospital of Wales, Cardiff, UK
| | | | - Steven G Deeks
- Division of HIV, Infectious Diseases, and Global Medicine, University of California, San Francisco, San Francisco, CA, USA
| | - William Eimer
- Harvard Medical School, Boston, MA, USA
- Genetics and Aging Research Unit, Mass General Institute for Neurodegenerative Disease, Charlestown, MA, USA
- Department of Neurology, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, USA
- McCance Center for Brain Health, Massachusetts General Hospital, Boston, MA, USA
| | - E Wesley Ely
- The Critical Illness, Brain Dysfunction, Survivorship (CIBS) Center at Vanderbilt University Medical Center and the Veteran's Affairs Tennessee Valley Geriatric Research Education Clinical Center (GRECC), Nashville, TN, USA
| | - Alessio Fasano
- Department of Pediatrics, Massachusetts General Hospital, Boston, MA, USA
- Mucosal Immunology and Biology Research Center, Massachusetts General Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Marcelo Freire
- J. Craig Venter Institute Department of Infectious Diseases, University of California, San Diego, La Jolla, CA, USA
| | - Linda N Geng
- Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Diane E Griffin
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Timothy J Henrich
- Division of Experimental Medicine, University of California, San Francisco, San Francisco, CA, USA
| | - Akiko Iwasaki
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT, USA
- Center for Infection and Immunity, Yale University School of Medicine, New Haven, CT, USA
- Howard Hughes Medical Institute, Chevy Chase, MD, USA
| | - David Izquierdo-Garcia
- Department of Radiology, Harvard Medical School, Charlestown, MA, USA
- Department of Health Sciences and Technology, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Michela Locci
- Institute for Immunology and Immune Health, and Department of Microbiology, University of Pennsylvania Perelman School Medicine, Philadelphia, PA, USA
| | - Saurabh Mehandru
- Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Henry D. Janowitz Division of Gastroenterology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Mark M Painter
- Institute for Immunology and Immune Health, and Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania Perelman School Medicine, Philadelphia, PA, USA
| | - Michael J Peluso
- Division of HIV, Infectious Diseases, and Global Medicine, University of California, San Francisco, San Francisco, CA, USA
| | - Etheresia Pretorius
- Department of Physiological Sciences, Faculty of Science, Stellenbosch University, Stellenbosch, South Africa
- Department of Biochemistry and Systems Biology, Institute of Systems, Molecular and Integrative Biology, Faculty of Health and Life Sciences, University of Liverpool, Liverpool, UK
| | - David A Price
- Division of Infection and Immunity, Cardiff University School of Medicine, University Hospital of Wales, Cardiff, UK
- Systems Immunity Research Institute, Cardiff University School of Medicine, University Hospital of Wales, Cardiff, UK
| | - David Putrino
- Department of Rehabilitation and Human Performance, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Richard H Scheuermann
- Department of Informatics, J. Craig Venter Institute, La Jolla, CA, USA
- Department of Pathology, University of California, San Diego, San Diego, CA, USA
- La Jolla Institute for Immunology, San Diego, CA, USA
| | - Gene S Tan
- J. Craig Venter Institute, La Jolla, CA, USA
- Division of Infectious Diseases, Department of Medicine, University of California San Diego, La Jolla, CA, USA
| | - Rudolph E Tanzi
- Harvard Medical School, Boston, MA, USA
- Genetics and Aging Research Unit, Mass General Institute for Neurodegenerative Disease, Charlestown, MA, USA
- Department of Neurology, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, USA
- McCance Center for Brain Health, Massachusetts General Hospital, Boston, MA, USA
| | - Henry F VanBrocklin
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, San Francisco, CA, USA
| | - Lael M Yonker
- Department of Pediatrics, Massachusetts General Hospital, Boston, MA, USA
- Mucosal Immunology and Biology Research Center, Massachusetts General Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - E John Wherry
- Institute for Immunology and Immune Health, and Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania Perelman School Medicine, Philadelphia, PA, USA
| |
Collapse
|
199
|
Caldeira D, Brito J, Gregório C, Plácido R, Pinto FJ. Short- and long-term effects of the COVID-19 pandemic on patients with cardiovascular diseases: A mini-review. HEART AND MIND 2023; 7:217-223. [DOI: 10.4103/hm.hm-d-23-00043] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2023] [Accepted: 10/23/2023] [Indexed: 01/03/2025] Open
Abstract
The COVID-19 pandemic had profound implications for patients with cardiovascular diseases (CVDs), both in the short- and long-term. In this article, we provide an overview of the effects of the pandemic on individuals with preexisting cardiovascular conditions. In the short term, the severe acute respiratory syndrome coronavirus 2 infection increased the risk of many cardiovascular events. Furthermore, the pandemic has disrupted health-care systems worldwide, leading to constraints in routine care, and limited access to specialized cardiovascular services and procedure. This has resulted in increased morbidity and mortality rates among patients with CVD (coronary artery disease, hypertrophic cardiomyopathy, heart failure (HF), heart transplant recipients, atrial fibrillation, atrial flutter, previous stroke, or previous peripheral artery disease). In the long term, the COVID-19 impact on patients with CVD extends beyond the acute phase of the disease. Studies have highlighted the development of long-term cardiovascular complications in COVID-19 survivors, such as acute coronary syndrome myocarditis, HF, stroke, venous thromboembolism, and arrhythmias, which may lead to a surge of new cases associated with CVD in the postpandemic era. Health-care systems must prioritize cardiovascular care, developing strategies to identify the patients at higher risk and provide the care to minimize the impact of the pandemic on patients with CVD.
Collapse
Affiliation(s)
- Daniel Caldeira
- Department of Cardiology, Santa Maria Hospital, CHULN, CAML, CCUL@RISE, Faculty of Medicine, University of Lisbon, Lisbon, Portugal
- Evidence-Based Medicine Center (CEMBE), Faculty of Medicine, University of Lisbon (Faculdade de Medicina, Universidade de Lisboa), Lisbon, Portugal
- Laboratory of Clinical Pharmacology and Therapeutics, Faculty of Medicine, University of Lisbon, Lisbon, Portugal
| | - Joana Brito
- Department of Cardiology, Santa Maria Hospital, CHULN, CAML, CCUL@RISE, Faculty of Medicine, University of Lisbon, Lisbon, Portugal
| | - Catarina Gregório
- Department of Cardiology, Santa Maria Hospital, CHULN, CAML, CCUL@RISE, Faculty of Medicine, University of Lisbon, Lisbon, Portugal
| | - Rui Plácido
- Department of Cardiology, Santa Maria Hospital, CHULN, CAML, CCUL@RISE, Faculty of Medicine, University of Lisbon, Lisbon, Portugal
| | - Fausto J. Pinto
- Department of Cardiology, Santa Maria Hospital, CHULN, CAML, CCUL@RISE, Faculty of Medicine, University of Lisbon, Lisbon, Portugal
| |
Collapse
|
200
|
Mateu L, Tebe C, Loste C, Santos JR, Lladós G, López C, España-Cueto S, Toledo R, Font M, Chamorro A, Muñoz-López F, Nevot M, Vallejo N, Teis A, Puig J, Fumaz CR, Muñoz-Moreno JA, Prats A, Estany-Quera C, Coll-Fernández R, Herrero C, Casares P, Garcia A, Clotet B, Paredes R, Massanella M. Determinants of the onset and prognosis of the post-COVID-19 condition: a 2-year prospective observational cohort study. THE LANCET REGIONAL HEALTH. EUROPE 2023; 33:100724. [PMID: 37954002 PMCID: PMC10636281 DOI: 10.1016/j.lanepe.2023.100724] [Citation(s) in RCA: 29] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/07/2023] [Revised: 08/09/2023] [Accepted: 08/16/2023] [Indexed: 11/14/2023]
Abstract
Background At least 5-10% of subjects surviving COVID-19 develop the post-COVID-19 condition (PCC) or "Long COVID". The clinical presentation of PCC is heterogeneous, its pathogenesis is being deciphered, and objective, validated biomarkers are lacking. It is unknown if PCC is a single entity or a heterogeneous syndrome with overlapping pathophysiological basis. The large US RECOVER study identified four clusters of subjects with PCC according to their presenting symptoms. However, the long-term clinical implications of PCC remain unknown. Methods We conducted a 2-year prospective cohort study of subjects surviving COVID-19, including individuals fulfilling the WHO PCC definition and subjects with full clinical recovery. We systematically collected post-COVID-19 symptoms using prespecified questionnaires and performed additional diagnostic imaging tests when needed. Factors associated with PCC were identified and modelled using logistic regression. Unsupervised clustering analysis was used to group subjects with PCC according to their presenting symptoms. Factors associated with PCC recovery were modelled using a direct acyclic graph approach. Findings The study included 548 individuals, 341 with PCC, followed for a median of 23 months (IQR 16.5-23.5), and 207 subjects fully recovered. In the model with the best fit, subjects who were male and had tertiary studies were less likely to develop PCC, whereas a history of headache, or presence of tachycardia, fatigue, neurocognitive and neurosensitive complaints and dyspnea at COVID-19 diagnosis predicted the development of PCC. The cluster analysis revealed the presence of three symptom clusters with an additive number of symptoms. Only 26 subjects (7.6%) recovered from PCC during follow-up; almost all of them (n = 24) belonged to the less symptomatic cluster A, dominated mainly by fatigue. Recovery from PCC was more likely in subjects who were male, required ICU admission, or had cardiovascular comorbidities, hyporexia and/or smell/taste alterations during acute COVID-19. Subjects presenting with muscle pain, impaired attention, dyspnea, or tachycardia, conversely, were less likely to recover from PCC. Interpretation Preexisting medical and socioeconomic factors, as well as acute COVID-19 symptoms, are associated with the development of and recovery from the PCC. Recovery is extremely rare during the first 2 years, posing a major challenge to healthcare systems. Funding Fundació Lluita contra les Infeccions.
Collapse
Affiliation(s)
- Lourdes Mateu
- Department of Infectious Diseases, Hospital Germans Trias i Pujol, Badalona, Catalonia, Spain
- Fundació Lluita Contra les Infeccions, Badalona, Catalonia, Spain
- Universitat Autònoma de Barcelona, Catalonia, Spain
- Universitat de Vic – UCC, Vic, Catalonia, Spain
- REICOP, Spain
| | - Cristian Tebe
- Biostatistics Unit, Hospital Germans Trias i Pujol, Institut de Recerca Germans Trias i Pujol, Can Ruti Campus, Badalona, Catalonia, Spain
| | - Cora Loste
- Department of Infectious Diseases, Hospital Germans Trias i Pujol, Badalona, Catalonia, Spain
- Fundació Lluita Contra les Infeccions, Badalona, Catalonia, Spain
- Universitat de Vic – UCC, Vic, Catalonia, Spain
- REICOP, Spain
| | - José Ramón Santos
- Department of Infectious Diseases, Hospital Germans Trias i Pujol, Badalona, Catalonia, Spain
- Fundació Lluita Contra les Infeccions, Badalona, Catalonia, Spain
| | - Gemma Lladós
- Department of Infectious Diseases, Hospital Germans Trias i Pujol, Badalona, Catalonia, Spain
- Fundació Lluita Contra les Infeccions, Badalona, Catalonia, Spain
- Universitat Autònoma de Barcelona, Catalonia, Spain
- REICOP, Spain
| | - Cristina López
- Department of Infectious Diseases, Hospital Germans Trias i Pujol, Badalona, Catalonia, Spain
- Fundació Lluita Contra les Infeccions, Badalona, Catalonia, Spain
| | - Sergio España-Cueto
- Department of Infectious Diseases, Hospital Germans Trias i Pujol, Badalona, Catalonia, Spain
- Fundació Lluita Contra les Infeccions, Badalona, Catalonia, Spain
| | - Ruth Toledo
- Department of Infectious Diseases, Hospital Germans Trias i Pujol, Badalona, Catalonia, Spain
- Fundació Lluita Contra les Infeccions, Badalona, Catalonia, Spain
| | - Marta Font
- Department of Infectious Diseases, Hospital Germans Trias i Pujol, Badalona, Catalonia, Spain
- Fundació Lluita Contra les Infeccions, Badalona, Catalonia, Spain
| | - Anna Chamorro
- Department of Infectious Diseases, Hospital Germans Trias i Pujol, Badalona, Catalonia, Spain
- Fundació Lluita Contra les Infeccions, Badalona, Catalonia, Spain
| | - Francisco Muñoz-López
- IrsiCaixa AIDS Research Institute, Germans Trias i Pujol Research Institute (IGTP), Can Ruti Campus, Badalona, Catalonia, Spain
| | - Maria Nevot
- IrsiCaixa AIDS Research Institute, Germans Trias i Pujol Research Institute (IGTP), Can Ruti Campus, Badalona, Catalonia, Spain
| | - Nuria Vallejo
- Cardiology Department, Hospital Germans Trias i Pujol, Badalona, Catalonia, Spain
| | - Albert Teis
- Cardiology Department, Hospital Germans Trias i Pujol, Badalona, Catalonia, Spain
| | - Jordi Puig
- Department of Infectious Diseases, Hospital Germans Trias i Pujol, Badalona, Catalonia, Spain
- Fundació Lluita Contra les Infeccions, Badalona, Catalonia, Spain
| | - Carmina R. Fumaz
- Department of Infectious Diseases, Hospital Germans Trias i Pujol, Badalona, Catalonia, Spain
- Fundació Lluita Contra les Infeccions, Badalona, Catalonia, Spain
- REICOP, Spain
| | - José A. Muñoz-Moreno
- Department of Infectious Diseases, Hospital Germans Trias i Pujol, Badalona, Catalonia, Spain
- Fundació Lluita Contra les Infeccions, Badalona, Catalonia, Spain
| | - Anna Prats
- Department of Infectious Diseases, Hospital Germans Trias i Pujol, Badalona, Catalonia, Spain
- Fundació Lluita Contra les Infeccions, Badalona, Catalonia, Spain
- REICOP, Spain
| | - Carla Estany-Quera
- Department of Infectious Diseases, Hospital Germans Trias i Pujol, Badalona, Catalonia, Spain
- Fundació Lluita Contra les Infeccions, Badalona, Catalonia, Spain
| | - Roser Coll-Fernández
- REICOP, Spain
- Department of Rehabilitation, Hospital Germans Trias i Pujol, Can Ruti Campus, Badalona, Catalonia, Spain
| | - Cristina Herrero
- Department of Infectious Diseases, Hospital Germans Trias i Pujol, Badalona, Catalonia, Spain
- Fundació Lluita Contra les Infeccions, Badalona, Catalonia, Spain
| | - Patricia Casares
- Department of Infectious Diseases, Hospital Germans Trias i Pujol, Badalona, Catalonia, Spain
- Fundació Lluita Contra les Infeccions, Badalona, Catalonia, Spain
| | - Ana Garcia
- Department of Infectious Diseases, Hospital Germans Trias i Pujol, Badalona, Catalonia, Spain
- Fundació Lluita Contra les Infeccions, Badalona, Catalonia, Spain
| | - Bonaventura Clotet
- Department of Infectious Diseases, Hospital Germans Trias i Pujol, Badalona, Catalonia, Spain
- Fundació Lluita Contra les Infeccions, Badalona, Catalonia, Spain
- Universitat Autònoma de Barcelona, Catalonia, Spain
- Universitat de Vic – UCC, Vic, Catalonia, Spain
- IrsiCaixa AIDS Research Institute, Germans Trias i Pujol Research Institute (IGTP), Can Ruti Campus, Badalona, Catalonia, Spain
- CIBER Infectious Diseases (CIBERINFEC), Institute of Health Carlos III (ISCIII), Madrid, Spain
| | - Roger Paredes
- Department of Infectious Diseases, Hospital Germans Trias i Pujol, Badalona, Catalonia, Spain
- Fundació Lluita Contra les Infeccions, Badalona, Catalonia, Spain
- Universitat Autònoma de Barcelona, Catalonia, Spain
- Universitat de Vic – UCC, Vic, Catalonia, Spain
- IrsiCaixa AIDS Research Institute, Germans Trias i Pujol Research Institute (IGTP), Can Ruti Campus, Badalona, Catalonia, Spain
- CIBER Infectious Diseases (CIBERINFEC), Institute of Health Carlos III (ISCIII), Madrid, Spain
- Center for Global Health and Diseases, Department of Pathology, Case Western Reserve University School of Medicine, Cleveland, OH, USA
| | - Marta Massanella
- REICOP, Spain
- IrsiCaixa AIDS Research Institute, Germans Trias i Pujol Research Institute (IGTP), Can Ruti Campus, Badalona, Catalonia, Spain
- CIBER Infectious Diseases (CIBERINFEC), Institute of Health Carlos III (ISCIII), Madrid, Spain
| |
Collapse
|