1
|
Harker JA, Thwaites RS. Unravelling the interplay between respiratory disease and the immune landscape in long COVID. Nat Immunol 2025; 26:640-641. [PMID: 40307448 DOI: 10.1038/s41590-025-02140-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/02/2025]
Affiliation(s)
- James A Harker
- National Heart and Lung Institute, Imperial College London, London, UK.
| | - Ryan S Thwaites
- National Heart and Lung Institute, Imperial College London, London, UK.
| |
Collapse
|
2
|
Levi R, Birberg Thornberg U, Blystad I, Divanoglou A, Engblom D, Leon F, Morberg Jämterud S, Zeiler K. Reconceptualizing rehabilitation research via an enactive framework and a radically interdisciplinary cross-analysis: a study protocol on fatigue in post COVID-19 condition (PCC). J Rehabil Med 2025; 57:jrm42254. [PMID: 40189912 PMCID: PMC11995433 DOI: 10.2340/jrm.v57.42254] [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: 10/15/2024] [Accepted: 02/04/2025] [Indexed: 04/16/2025] Open
Abstract
OBJECTIVE To present a radically interdisciplinary research approach to ill-defined symptoms, with a focus on fatigue as a major symptom of post COVID-19 condition, where multiple and, to date, rarely combined approaches may yield a fuller understanding of these symptoms. DESIGN Protocol for a mixed-methods study comprising an interdisciplinary cross-analysis. PATIENTS 35 persons with post COVID-19 condition and severe fatigue were included, and 35 age-, sex-, and educationally matched controls who recovered from COVID-19 without post COVID-19 condition. METHODS Participants were assessed by a multidisciplinary research team as follows: physician assessment; blood and urinalysis; spirometry and physical performance tests; neuropsychological tests; structural and functional magnetic resonance imaging; extended immunological tests (cytokines); and qualitative phenomenological analysis of interviews. Data will be analysed in accordance with established methods in each of these research fields and by a cross-analysis methodology developed from within an enactive framework. This framework encompasses a focus on neuroscientific, physiological, and experiential aspects of the person as a living being in their sociocultural world. CONCLUSION The biopsychosocial model needs to be implemented in research according to methods that allow radically different research paradigms, typically seen as incommensurable, to inform each other in a non-reductionist manner. One application of such an approach is therefore described.
Collapse
Affiliation(s)
- Richard Levi
- Department of Rehabilitation Medicine and Department of Medical and Health Sciences, Linköping University, Linköping, Sweden
| | - Ulrika Birberg Thornberg
- Department of Rehabilitation Medicine and Department of Medical and Health Sciences, Linköping University, Linköping, Sweden
| | - Ida Blystad
- Department of Radiology Linköping, and Department of Health, Medicine and Caring Sciences, Linköping University, Linköping, Sweden; Centre for Medical Image Science and Visualization (CMIV), Linköping University, Linköping, Sweden
| | - Anestis Divanoglou
- Department of Rehabilitation Medicine and Department of Medical and Health Sciences, Linköping University, Linköping, Sweden
| | - David Engblom
- Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
| | - Felipe Leon
- Centre of Philosophy, School of Arts and Humanities, University of Lisbon, Lisboa, Portugal
| | - Sofia Morberg Jämterud
- Department of Thematic Studies: Technology and Social Change and the Centre for Medical Humanities and Bioethics, Linköping University, Linköping, Sweden
| | - Kristin Zeiler
- Centre for Medical Image Science and Visualization (CMIV), Linköping University, Linköping, Sweden; Department of Thematic Studies: Technology and Social Change and the Centre for Medical Humanities and Bioethics, Linköping University, Linköping, Sweden.
| |
Collapse
|
3
|
Scott JM, Qiu Z, Rahman J, Moskowitz CS, Michalski MG, Lehman S, Lee CP, Harrison J, Yu AF, Marouf A, Vardhana S, Boutros PC, Jones LW. Case Report: Decentralized trial of tolerability-adapted exercise therapy after severe Covid-19. Front Immunol 2025; 16:1529385. [PMID: 40248705 PMCID: PMC12003135 DOI: 10.3389/fimmu.2025.1529385] [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: 11/16/2024] [Accepted: 03/17/2025] [Indexed: 04/19/2025] Open
Abstract
We assessed the safety, tolerability, and effects of exercise therapy in three patients with cancer and hospitalization for SARS-CoV-2 infection in an early-phase prospective trial. All study assessments and exercise sessions were conducted remotely (decentralized) in patient's homes. Patients received five escalated doses of aerobic exercise therapy (range, 90 to 375 minutes per week) following a tolerability-based adapted schedule over 30 consecutive weeks. Exercise therapy was safe (i.e., no serious adverse events), tolerable (i.e., all exercise therapy doses were completed, with an overall average relative exercise dose intensity of 89%), and associated with improvements in patient physiology (e.g., exercise capacity) and patient-reported outcomes (e.g., quality of life). Correlative proteomic and single-cell immune sequencing of peripheral blood samples revealed marked alterations in protein and immune phenotypes implicated in post COVID-19 condition. (ClinicalTrials.gov number, NCT04824443).
Collapse
Affiliation(s)
- Jessica M. Scott
- Department of Medicine, Memorial Sloan Kettering Cancer Center (MSK), New York, NY, United States
- Department of Medicine, Weill Cornell Medical College, New York, NY, United States
| | - Zhuyu Qiu
- Jonsson Comprehensive Cancer Center, University of California, Los Angeles, Los Angeles, CA, United States
- Department of Human Genetics, University of California, Los Angeles, Los Angeles, CA, United States
| | - Jahan Rahman
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY, United States
| | - Chaya S. Moskowitz
- Department of Medicine, Memorial Sloan Kettering Cancer Center (MSK), New York, NY, United States
| | - Meghan G. Michalski
- Department of Medicine, Memorial Sloan Kettering Cancer Center (MSK), New York, NY, United States
| | - Sarah Lehman
- Department of Medicine, Memorial Sloan Kettering Cancer Center (MSK), New York, NY, United States
| | - Catherine P. Lee
- Department of Medicine, Memorial Sloan Kettering Cancer Center (MSK), New York, NY, United States
| | - Jenna Harrison
- Department of Medicine, Memorial Sloan Kettering Cancer Center (MSK), New York, NY, United States
| | - Anthony F. Yu
- Department of Medicine, Memorial Sloan Kettering Cancer Center (MSK), New York, NY, United States
- Department of Medicine, Weill Cornell Medical College, New York, NY, United States
| | - Amira Marouf
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY, United States
- Department of Medicine, Lymphoma Service, Memorial Sloan Kettering Cancer Center, New York, NY, United States
| | - Santosha Vardhana
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY, United States
- Department of Medicine, Lymphoma Service, Memorial Sloan Kettering Cancer Center, New York, NY, United States
| | - Paul C. Boutros
- Jonsson Comprehensive Cancer Center, University of California, Los Angeles, Los Angeles, CA, United States
- Department of Human Genetics, University of California, Los Angeles, Los Angeles, CA, United States
- Department of Urology, University of California, Los Angeles, CA, United States
- Institute for Precision Health, University of California, Los Angeles, CA, United States
| | - Lee W. Jones
- Department of Medicine, Memorial Sloan Kettering Cancer Center (MSK), New York, NY, United States
- Department of Medicine, Weill Cornell Medical College, New York, NY, United States
| |
Collapse
|
4
|
Liu J, Guo L, Zhong J, Wu Y, Wang X, Tang X, Min K, Yang Y, Peng W, Wang Q, Ding T, Gu X, Zhang H, Liu Y, Huang C, Cao B, Wang J, Ren L, Yang J. Proteomic Analysis of 442 Clinical Plasma Samples From Individuals With Symptom Records Revealed Subtypes of Convalescent Patients Who Had COVID-19. J Med Virol 2025; 97:e70203. [PMID: 40207927 PMCID: PMC11984345 DOI: 10.1002/jmv.70203] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Revised: 01/11/2025] [Accepted: 01/21/2025] [Indexed: 04/11/2025]
Abstract
After the coronavirus disease 2019 (COVID-19) pandemic, the postacute effects of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection have gradually attracted attention. To precisely evaluate the health status of convalescent patients with COVID-19, we analyzed symptom and proteome data of 442 plasma samples from healthy controls, hospitalized patients, and convalescent patients 6 or 12 months after SARS-CoV-2 infection. Symptoms analysis revealed distinct relationships in convalescent patients. Results of plasma protein expression levels showed that C1QA, C1QB, C2, CFH, CFHR1, and F10, which regulate the complement system and coagulation, remained highly expressed even at the 12-month follow-up compared with their levels in healthy individuals. By combining symptom and proteome data, 442 plasma samples were categorized into three subtypes: S1 (metabolism-healthy), S2 (COVID-19 retention), and S3 (long COVID). We speculated that convalescent patients reporting hair loss could have a better health status than those experiencing headaches and dyspnea. Compared to other convalescent patients, those reporting sleep disorders, appetite decrease, and muscle weakness may need more attention because they were classified into the S2 subtype, which had the most samples from hospitalized patients with COVID-19. Subtyping convalescent patients with COVID-19 may enable personalized treatments tailored to individual needs. This study provides valuable plasma proteomic datasets for further studies associated with long COVID.
Collapse
Grants
- This work was supported by grants from the National Key R&D Program of China (2023YFC2507102), the Chinese Academy of Medical Sciences Innovation Fund for Medical Sciences, China (CIFMS2022-I2M-1-011, CIFMS2022-I2M-2-001, CIFMS2021-I2M-1-057, CIFMS2021-I2M-1-049, CIFMS2021-I2M-1-044, CIFMS2021-I2M-1-016, CIFMS2021-I2M-1-001, 2022-I2M-CoV19-003, and CIFMS2022-I2M-JB-003), the National Natural Science Foundation of China (82341064), the Haihe Laboratory of Cell Ecosystem Innovation Fund (22HHXBSS00008 and 22HHKYZX0034), and State Key Laboratory Special Fund 2060204.
Collapse
Affiliation(s)
- Jiangfeng Liu
- Haihe Laboratory of Cell EcosystemTianjinChina
- State Key Laboratory of Common Mechanism Research for Major Diseases, Department of Biochemistry and Molecular BiologySchool of Basic Medicine, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, Peking Union Medical CollegeBeijingChina
- Plastic Surgery Hospital, Chinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Li Guo
- NHC Key Laboratory of Systems Biology of Pathogens and Christophe Merieux LaboratoryInstitute of Pathogen Biology, Chinese Academy of Medical SciencesBeijingChina
| | - Jingchuan Zhong
- NHC Key Laboratory of Systems Biology of Pathogens and Christophe Merieux LaboratoryInstitute of Pathogen Biology, Chinese Academy of Medical SciencesBeijingChina
| | - Yue Wu
- State Key Laboratory of Common Mechanism Research for Major Diseases, Department of Biochemistry and Molecular BiologySchool of Basic Medicine, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, Peking Union Medical CollegeBeijingChina
| | - Xinming Wang
- NHC Key Laboratory of Systems Biology of Pathogens and Christophe Merieux LaboratoryInstitute of Pathogen Biology, Chinese Academy of Medical SciencesBeijingChina
| | - Xiaoyue Tang
- State Key Laboratory of Common Mechanism Research for Major Diseases, Department of Biochemistry and Molecular BiologySchool of Basic Medicine, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, Peking Union Medical CollegeBeijingChina
| | - Kaiyuan Min
- State Key Laboratory of Common Mechanism Research for Major Diseases, Department of Biochemistry and Molecular BiologySchool of Basic Medicine, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, Peking Union Medical CollegeBeijingChina
| | - Yehong Yang
- State Key Laboratory of Common Mechanism Research for Major Diseases, Department of Biochemistry and Molecular BiologySchool of Basic Medicine, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, Peking Union Medical CollegeBeijingChina
| | - Wanjun Peng
- State Key Laboratory of Common Mechanism Research for Major Diseases, Department of Biochemistry and Molecular BiologySchool of Basic Medicine, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, Peking Union Medical CollegeBeijingChina
| | - Qiaochu Wang
- State Key Laboratory of Common Mechanism Research for Major Diseases, Department of Biochemistry and Molecular BiologySchool of Basic Medicine, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, Peking Union Medical CollegeBeijingChina
| | - Tao Ding
- State Key Laboratory of Common Mechanism Research for Major Diseases, Department of Biochemistry and Molecular BiologySchool of Basic Medicine, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, Peking Union Medical CollegeBeijingChina
| | - Xiaoying Gu
- Tsinghua University‐Peking University Joint Center for Life SciencesBeijingChina
- Department of Pulmonary and Critical Care MedicineNational Center for Respiratory Medicine, Center of Respiratory Medicine, National Clinical Research Center for Respiratory DiseasesBeijingChina
- Institute of Respiratory Medicine, Chinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
- Department of Pulmonary and Critical Care MedicineCapital Medical UniversityBeijingChina
| | - Hui Zhang
- Tsinghua University‐Peking University Joint Center for Life SciencesBeijingChina
- Department of Pulmonary and Critical Care MedicineNational Center for Respiratory Medicine, Center of Respiratory Medicine, National Clinical Research Center for Respiratory DiseasesBeijingChina
- Institute of Respiratory Medicine, Chinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
- Department of Pulmonary and Critical Care MedicineCapital Medical UniversityBeijingChina
| | - Ying Liu
- Medical DepartmentJin Yin‐Tan HospitalWuhanHubeiChina
- Wuhan Research Center for Communicable Disease Diagnosis and Treatment, Chinese Academy of Medical SciencesWuhanHubeiChina
| | - Chaolin Huang
- Medical DepartmentJin Yin‐Tan HospitalWuhanHubeiChina
- Wuhan Research Center for Communicable Disease Diagnosis and Treatment, Chinese Academy of Medical SciencesWuhanHubeiChina
| | - Bin Cao
- Tsinghua University‐Peking University Joint Center for Life SciencesBeijingChina
- Department of Pulmonary and Critical Care MedicineNational Center for Respiratory Medicine, Center of Respiratory Medicine, National Clinical Research Center for Respiratory DiseasesBeijingChina
- Institute of Respiratory Medicine, Chinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
- Department of Pulmonary and Critical Care MedicineCapital Medical UniversityBeijingChina
| | - Jianwei Wang
- NHC Key Laboratory of Systems Biology of Pathogens and Christophe Merieux LaboratoryInstitute of Pathogen Biology, Chinese Academy of Medical SciencesBeijingChina
- Key Laboratory of Respiratory Disease PathogenomicsChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Lili Ren
- NHC Key Laboratory of Systems Biology of Pathogens and Christophe Merieux LaboratoryInstitute of Pathogen Biology, Chinese Academy of Medical SciencesBeijingChina
- Key Laboratory of Respiratory Disease PathogenomicsChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Juntao Yang
- Haihe Laboratory of Cell EcosystemTianjinChina
- State Key Laboratory of Common Mechanism Research for Major Diseases, Department of Biochemistry and Molecular BiologySchool of Basic Medicine, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, Peking Union Medical CollegeBeijingChina
- Plastic Surgery Hospital, Chinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| |
Collapse
|
5
|
Canderan G, Muehling LM, Kadl A, Ladd S, Bonham C, Cross CE, Lima SM, Yin X, Sturek JM, Wilson JM, Keshavarz B, Enfield KB, Ramani C, Bryant N, Murphy DD, Cheon IS, Solga M, Pramoonjago P, McNamara CA, Sun J, Utz PJ, Dolatshahi S, Irish JM, Woodfolk JA. Distinct type 1 immune networks underlie the severity of restrictive lung disease after COVID-19. Nat Immunol 2025; 26:595-606. [PMID: 40140496 DOI: 10.1038/s41590-025-02110-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2024] [Accepted: 02/14/2025] [Indexed: 03/28/2025]
Abstract
The variable origins of persistent breathlessness after coronavirus disease 2019 (COVID-19) have hindered efforts to decipher the immunopathology of lung sequelae. Here we analyzed hundreds of cellular and molecular features in the context of discrete pulmonary phenotypes to define the systemic immune landscape of post-COVID lung disease. Cluster analysis of lung physiology measures highlighted two phenotypes of restrictive lung disease that differed according to their impaired diffusion and severity of fibrosis. Machine learning revealed marked CCR5+CD95+CD8+ T cell perturbations in milder lung disease but attenuated T cell responses hallmarked by elevated CXCL13 in more severe disease. Distinct sets of cells, mediators and autoantibodies distinguished each restrictive phenotype and differed from those of patients without substantial lung involvement. These differences were reflected in divergent T cell-based type 1 networks according to the severity of lung disease. Our findings, which provide an immunological basis for active lung injury versus advanced disease after COVID-19, might offer new targets for treatment.
Collapse
Affiliation(s)
- Glenda Canderan
- Department of Medicine, University of Virginia School of Medicine, Charlottesville, VA, USA
| | - Lyndsey M Muehling
- Department of Medicine, University of Virginia School of Medicine, Charlottesville, VA, USA
| | - Alexandra Kadl
- Department of Medicine, University of Virginia School of Medicine, Charlottesville, VA, USA
- Department of Pharmacology, University of Virginia School of Medicine, Charlottesville, VA, USA
| | - Shay Ladd
- Department of Biomedical Engineering, University of Virginia School of Engineering and Applied Science, Charlottesville, VA, USA
| | - Catherine Bonham
- Department of Medicine, University of Virginia School of Medicine, Charlottesville, VA, USA
| | - Claire E Cross
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, TN, USA
- Vanderbilt Center for Immunobiology, Vanderbilt University Medical Center, Nashville, TN, USA
- Department of Pediatrics, University of Colorado Anschutz Medical Center, Aurora, CO, USA
| | - Sierra M Lima
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, TN, USA
- Vanderbilt Center for Immunobiology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Xihui Yin
- Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA
- Institute for Immunity, Transplantation and Infection, Stanford University School of Medicine, Stanford, CA, USA
| | - Jeffrey M Sturek
- Department of Medicine, University of Virginia School of Medicine, Charlottesville, VA, USA
- Beirne B. Carter Center for Immunology Research, University of Virginia School of Medicine, Charlottesville, VA, USA
| | - Jeffrey M Wilson
- Department of Medicine, University of Virginia School of Medicine, Charlottesville, VA, USA
- Beirne B. Carter Center for Immunology Research, University of Virginia School of Medicine, Charlottesville, VA, USA
| | - Behnam Keshavarz
- Department of Medicine, University of Virginia School of Medicine, Charlottesville, VA, USA
| | - Kyle B Enfield
- Department of Medicine, University of Virginia School of Medicine, Charlottesville, VA, USA
| | - Chintan Ramani
- Department of Medicine, University of Virginia School of Medicine, Charlottesville, VA, USA
| | - Naomi Bryant
- Department of Medicine, University of Virginia School of Medicine, Charlottesville, VA, USA
- Department of Microbiology, Immunology, and Cancer Biology, University of Virginia School of Medicine, Charlottesville, VA, USA
| | - Deborah D Murphy
- Department of Medicine, University of Virginia School of Medicine, Charlottesville, VA, USA
| | - In Su Cheon
- Department of Medicine, University of Virginia School of Medicine, Charlottesville, VA, USA
| | - Michael Solga
- Department of Microbiology, Immunology, and Cancer Biology, University of Virginia School of Medicine, Charlottesville, VA, USA
| | - Patcharin Pramoonjago
- Department of Pathology, University of Virginia School of Medicine, Charlottesville, VA, USA
| | - Coleen A McNamara
- Department of Medicine, University of Virginia School of Medicine, Charlottesville, VA, USA
- Beirne B. Carter Center for Immunology Research, University of Virginia School of Medicine, Charlottesville, VA, USA
| | - Jie Sun
- Department of Medicine, University of Virginia School of Medicine, Charlottesville, VA, USA
- Beirne B. Carter Center for Immunology Research, University of Virginia School of Medicine, Charlottesville, VA, USA
| | - Paul J Utz
- Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA
- Institute for Immunity, Transplantation and Infection, Stanford University School of Medicine, Stanford, CA, USA
| | - Sepideh Dolatshahi
- Beirne B. Carter Center for Immunology Research, University of Virginia School of Medicine, Charlottesville, VA, USA
- Department of Biomedical Engineering, University of Virginia School of Medicine, Charlottesville, VA, USA
| | - Jonathan M Irish
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, TN, USA
- Vanderbilt Center for Immunobiology, Vanderbilt University Medical Center, Nashville, TN, USA
- Department of Pediatrics, University of Colorado Anschutz Medical Center, Aurora, CO, USA
| | - Judith A Woodfolk
- Department of Medicine, University of Virginia School of Medicine, Charlottesville, VA, USA.
- Beirne B. Carter Center for Immunology Research, University of Virginia School of Medicine, Charlottesville, VA, USA.
- Department of Microbiology, Immunology, and Cancer Biology, University of Virginia School of Medicine, Charlottesville, VA, USA.
| |
Collapse
|
6
|
Talwar S, Harker JA, Openshaw PJM, Thwaites RS. Autoimmunity in long COVID. J Allergy Clin Immunol 2025; 155:1082-1094. [PMID: 39956285 DOI: 10.1016/j.jaci.2025.02.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2024] [Revised: 01/24/2025] [Accepted: 02/07/2025] [Indexed: 02/18/2025]
Abstract
Long COVID (also termed postacute sequelae of SARS-CoV-2, or PASC) affects up to 10% of people recovering from infection with severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). Diagnosis is hampered by diffuse symptomatology, lack of biomarkers, incomplete understanding of pathogenesis, and lack of validated treatments. In terms of pathogenesis, hypothesized causes include virus persistence, the legacy of endotheliitis and thrombosis, low-grade tissue-based inflammation and/or scarring, perturbation of the host virome/microbiome, or triggering of autoimmunity. Several studies show preexisting and/or de novo production of autoantibodies after infection with SARS-CoV-2, but the persistence of these antibodies and their role in causing long COVID is debated. Here, we review the mechanisms through which autoimmune responses can arise during and after viral infection, focusing on the evidence for B-cell dysregulation and autoantibody production in acute and long COVID.
Collapse
Affiliation(s)
- Shubha Talwar
- National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - James A Harker
- National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - Peter J M Openshaw
- National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - Ryan S Thwaites
- National Heart and Lung Institute, Imperial College London, London, United Kingdom.
| |
Collapse
|
7
|
Yang H, Guan L, Xue Y, Li X, Gao L, Zhang Z, Zhang H, Ma H, Liu F, Huang X, Tong Z, Li J. Longitudinal multi-omics analysis of convalescent individuals with respiratory sequelae 6-36 months after COVID-19. BMC Med 2025; 23:134. [PMID: 40038650 DOI: 10.1186/s12916-025-03971-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/18/2024] [Accepted: 02/26/2025] [Indexed: 03/06/2025] Open
Abstract
BACKGROUND Approximately 10-30% of individuals continue to experience symptoms classified as post-acute sequelae of coronavirus disease 2019 (COVID-19 (PASC)). PASC is a multisystem condition primarily characterized by respiratory symptoms, such as reduced diffusing capacity for carbon monoxide (DLco). Although many studies have investigated the pathogenesis of acute COVID-19, the long-term molecular changes in COVID-19 convalescents with PASC remain poorly understood. METHODS We prospectively recruited 70 individuals who had been diagnosed with COVID-19 from 7 January 2020 to 29 May 2020 (i.e., COVID-19 convalescents); we performed follow-up visits at 6 months, 1 year, 2 years, and 3 years after hospital discharge. Thirty-five healthy controls (CONs), recruited from a physical examination center before the COVID-19 pandemic, served as a comparison group. We explored the proteomic and metabolomic profiles of 174 plasma samples from the 70 COVID-19 convalescents and 35 CONs. RESULTS We performed a comprehensive molecular analysis of COVID-19 convalescents to investigate host changes up to 3 years after hospital discharge. Our multi-omics analysis revealed activation of cytoskeletal organization and glycolysis/gluconeogenesis, as well as suppression of gas transport and adaptive immune responses, in COVID-19 convalescents. Additionally, metabolites involved in glutathione metabolism; alanine, aspartate, and glutamate metabolism; and ascorbate and aldarate metabolism were significantly upregulated in COVID-19 convalescents. Pulmonary and molecular abnormalities persisted for 3 years in COVID-19 convalescents; impaired diffusing capacity for carbon monoxide (DLco) was the most prominent feature. We used this multi-omics profile to develop a model involving one protein (heterogeneous nuclear ribonucleoprotein K (HNRNPK)) and two metabolites (arachidonoyl-EA and 1-O-(2r-hydroxy-pentadecyl)-sn-glycerol)) for identification of COVID-19 convalescents with abnormal DLco. CONCLUSIONS These data provide insights concerning molecular sequelae among COVID-19 convalescents up to 3 years after hospital discharge, clarify mechanisms driving respiratory sequelae, and support the development of a novel model to predict reduced DLco. This longitudinal multi-omics analysis may illuminate the trajectory of altered lung function in COVID-19 convalescents.
Collapse
Affiliation(s)
- Huqin Yang
- Department of Respiratory and Critical Care Medicine, Beijing Institute of Respiratory Medicine and Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
| | - Lujia Guan
- Department of Respiratory and Critical Care Medicine, Beijing Institute of Respiratory Medicine and Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
| | - Yi Xue
- Department of Respiratory and Critical Care Medicine, Beijing Institute of Respiratory Medicine and Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
| | - Xuyan Li
- Department of Respiratory and Critical Care Medicine, Beijing Institute of Respiratory Medicine and Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
| | - Leyi Gao
- Department of Respiratory and Critical Care Medicine, Beijing Institute of Respiratory Medicine and Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
| | - Zhijin Zhang
- Department of Respiratory and Critical Care Medicine, Beijing Institute of Respiratory Medicine and Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
| | - Haifan Zhang
- Department of Respiratory and Critical Care Medicine, Beijing Institute of Respiratory Medicine and Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
| | - Haomiao Ma
- Department of Respiratory and Critical Care Medicine, Beijing Institute of Respiratory Medicine and Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
| | - Fengjiao Liu
- Medical Research Center, Beijing Institute of Respiratory Medicine and Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
| | - Xuan Huang
- Department of Respiratory and Critical Care Medicine, Beijing Institute of Respiratory Medicine and Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
- Medical Research Center, Beijing Institute of Respiratory Medicine and Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
| | - Zhaohui Tong
- Department of Respiratory and Critical Care Medicine, Beijing Institute of Respiratory Medicine and Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China.
- Beijing Research Center for Respiratory Infectious Diseases, Beijing, China.
| | - Jieqiong Li
- Department of Respiratory and Critical Care Medicine, Beijing Institute of Respiratory Medicine and Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China.
- Medical Research Center, Beijing Institute of Respiratory Medicine and Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China.
| |
Collapse
|
8
|
Chen Y, Jan J, Yang C, Yen T, Linh TTD, Annavajjula S, Satapathy MK, Tsao S, Hsieh C. Cognitive Sequelae of COVID-19: Mechanistic Insights and Therapeutic Approaches. CNS Neurosci Ther 2025; 31:e70348. [PMID: 40152069 PMCID: PMC11950837 DOI: 10.1111/cns.70348] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2024] [Revised: 03/10/2025] [Accepted: 03/13/2025] [Indexed: 03/29/2025] Open
Abstract
BACKGROUND The COVID-19 pandemic has left an indelible mark on the world, with mounting evidence suggesting that it not only posed acute challenges to global healthcare systems but has also unveiled a complex array of long-term consequences, particularly cognitive impairment (CI). As the persistence of post-COVID-19 neurological syndrome could evolve into the next public health crisis, it is imperative to gain a better understanding of the intricate pathophysiology of CI in COVID-19 patients and viable treatment strategies. METHODS This comprehensive review explores the pathophysiology and management of cognitive impairment across the phases of COVID-19, from acute infection to Long-COVID, by synthesizing findings from clinical, preclinical, and mechanistic studies to identify key contributors to CI, as well as current therapeutic approaches. RESULTS Key mechanisms contributing to CI include persistent neuroinflammation, cerebrovascular complications, direct neuronal injury, activation of the kynurenine pathway, and psychological distress. Both pharmacological interventions, such as anti-inflammatory therapies and agents targeting neuroinflammatory pathways, and non-pharmacological strategies, including cognitive rehabilitation, show promise in addressing these challenges. Although much of the current evidence is derived from preclinical and animal studies, these findings provide foundational insights into potential treatment approaches. CONCLUSION By synthesizing current knowledge, this review highlights the importance of addressing COVID-19-related cognitive impairment and offers actionable insights for mitigation and recovery as the global community continues to grapple with the pandemic's long-term impact.
Collapse
Affiliation(s)
- Yu‐Hao Chen
- Section of Neurosurgery, Department of SurgeryDitmanson Medical Foundation, Chia‐Yi Christian HospitalChia‐Yi CityTaiwan
- Chung‐Jen Junior College of Nursing, Health Sciences and ManagementChia‐Yi CountryTaiwan
- Department of PharmacologySchool of Medicine, College of Medicine, Taipei Medical UniversityTaipeiTaiwan
| | - Jing‐Shiun Jan
- Department of PharmacologySchool of Medicine, College of Medicine, Taipei Medical UniversityTaipeiTaiwan
| | - Chih‐Hao Yang
- Department of PharmacologySchool of Medicine, College of Medicine, Taipei Medical UniversityTaipeiTaiwan
| | - Ting‐Lin Yen
- Department of PharmacologySchool of Medicine, College of Medicine, Taipei Medical UniversityTaipeiTaiwan
- Department of Medical ResearchCathay General HospitalTaipeiTaiwan
| | - Tran Thanh Duy Linh
- Department of PharmacologySchool of Medicine, College of Medicine, Taipei Medical UniversityTaipeiTaiwan
- Family Medicine Training Center, University of Medicine and Pharmacy at Ho Chi Minh CityHo Chi Minh CityVietnam
| | - Saileela Annavajjula
- Department of PharmacologySchool of Medicine, College of Medicine, Taipei Medical UniversityTaipeiTaiwan
| | - Mantosh Kumar Satapathy
- Department of PharmacologySchool of Medicine, College of Medicine, Taipei Medical UniversityTaipeiTaiwan
| | - Shin‐Yi Tsao
- Department of PharmacologySchool of Medicine, College of Medicine, Taipei Medical UniversityTaipeiTaiwan
- Division of Endocrinology and Metabolism, Department of Internal MedicineTaipeiTaiwan
| | - Cheng‐Ying Hsieh
- Department of PharmacologySchool of Medicine, College of Medicine, Taipei Medical UniversityTaipeiTaiwan
| |
Collapse
|
9
|
Gabernet G, Maciuch J, Gygi JP, Moore JF, Hoch A, Syphurs C, Chu T, Jayavelu ND, Corry DB, Kheradmand F, Baden LR, Sekaly RP, McComsey GA, Haddad EK, Cairns CB, Rouphael N, Fernandez-Sesma A, Simon V, Metcalf JP, Agudelo Higuita NI, Hough CL, Messer WB, Davis MM, Nadeau KC, Pulendran B, Kraft M, Bime C, Reed EF, Schaenman J, Erle DJ, Calfee CS, Atkinson MA, Brackenridge SC, Melamed E, Shaw AC, Hafler DA, Ozonoff A, Bosinger SE, Eckalbar W, Maecker HT, Kim-Schulze S, Steen H, Krammer F, Westendorf K, Network I, Peters B, Fourati S, Altman MC, Levy O, Smolen KK, Montgomery RR, Diray-Arce J, Kleinstein SH, Guan L, Ehrlich LIR. Identification of a multi-omics factor predictive of long COVID in the IMPACC study. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.12.637926. [PMID: 39990442 PMCID: PMC11844572 DOI: 10.1101/2025.02.12.637926] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 02/25/2025]
Abstract
Following SARS-CoV-2 infection, ∼10-35% of COVID-19 patients experience long COVID (LC), in which often debilitating symptoms persist for at least three months. Elucidating the biologic underpinnings of LC could identify therapeutic opportunities. We utilized machine learning methods on biologic analytes and patient reported outcome surveys provided over 12 months after hospital discharge from >500 hospitalized COVID-19 patients in the IMPACC cohort to identify a multi-omics "recovery factor". IMPACC participants who experienced LC had lower recovery factor scores compared to participants without LC. Biologic characterization revealed increased levels of plasma proteins associated with inflammation, elevated transcriptional signatures of heme metabolism, and decreased androgenic steroids in LC patients. The recovery factor was also associated with altered circulating immune cell frequencies. Notably, recovery factor scores were predictive of LC occurrence in patients as early as hospital admission, irrespective of acute disease severity. Thus, the recovery factor identifies patients at risk of LC early after SARS-CoV-2 infection and reveals LC biomarkers and potential treatment targets.
Collapse
|
10
|
Dal-Pizzol F, Kluwe-Schiavon B, Dal-Pizzol HR, da Silveira Prestes G, Dominguini D, Girardi CS, Santos L, Moreira JCF, Gelain DP, Walz R, Barichello T, Ritter C. Association of systemic inflammation and long-term dysfunction in COVID-19 patients: A prospective cohort. Psychoneuroendocrinology 2025; 172:107269. [PMID: 39778322 PMCID: PMC12045274 DOI: 10.1016/j.psyneuen.2024.107269] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/25/2024] [Revised: 12/19/2024] [Accepted: 12/22/2024] [Indexed: 01/11/2025]
Abstract
COVID-19 has significant long-term impacts, including a chronic syndrome known as long-COVID, characterized by persistent symptoms post-recovery. The inflammatory response during acute infection is hypothesized to influence long-term outcomes. This study aimed to identify inflammatory biomarkers predictive of functional outcomes one year after hospital discharge. A prospective cohort study was conducted with 213 COVID-19 patients admitted to ICUs in Southern Brazil between June and November 2020. After exclusions and follow-ups, 109 patients were evaluated for one-year post-discharge. Plasma levels of Th1 (TNF-α, INF-γ, IL-12), Th2 (IL-4, IL-5, IL-6, IL-10, IL-13), and Th17 (IL-17, IL-22) cytokines were measured. Functional outcomes in psychiatric, cognitive, general health, and health perception domains were assessed. Statistical analyses included multivariate regression, regularized partial correlation network analysis, and K-means clustering. We demonstrate that plasma levels of various cytokines, along with demographic and clinical characteristics, can predict four distinct domains of functional outcomes one year following hospital discharge due to COVID-19 and that an hyperinflammatory phenotype was associated with the occurrence of a worse in psychiatric, general health, and health perception domains. The network analysis highlighted complex interconnections among immune markers and clinical variables, elucidating their roles in long-term health. These findings support using biomarkers for patient stratification and indicate potential targets for therapeutic interventions.
Collapse
Affiliation(s)
- Felipe Dal-Pizzol
- Laboratory of Experimental Physiopathology, Graduate Program in Health Sciences, Health Sciences Unit, University of Southern Santa Catarina, Criciuma, SC, Brazil; Hospital São José Research Center, Criciúma, SC, Brazil.
| | - Bruno Kluwe-Schiavon
- Translational Psychiatry Program, Faillace Department of Psychiatry and Behavioral Sciences, McGovern Medical School, University of Texas Health Science Center at Houston (UTHealth), Houston, TX 77054, USA
| | - Henrique Ritter Dal-Pizzol
- Center of Oxidative Stress Studies, Department of Biochemistry, Institute of Basic Health Sciences, Federal University of Rio Grande do Sul - UFRGS, Porto Alegre 90035-003, Brazil
| | - Gabriele da Silveira Prestes
- Laboratory of Experimental Physiopathology, Graduate Program in Health Sciences, Health Sciences Unit, University of Southern Santa Catarina, Criciuma, SC, Brazil
| | - Diogo Dominguini
- Laboratory of Experimental Physiopathology, Graduate Program in Health Sciences, Health Sciences Unit, University of Southern Santa Catarina, Criciuma, SC, Brazil
| | - Carolina Saibro Girardi
- Center of Oxidative Stress Studies, Department of Biochemistry, Institute of Basic Health Sciences, Federal University of Rio Grande do Sul - UFRGS, Porto Alegre 90035-003, Brazil
| | - Lucas Santos
- Center of Oxidative Stress Studies, Department of Biochemistry, Institute of Basic Health Sciences, Federal University of Rio Grande do Sul - UFRGS, Porto Alegre 90035-003, Brazil
| | - José Cláudio Fonseca Moreira
- Center of Oxidative Stress Studies, Department of Biochemistry, Institute of Basic Health Sciences, Federal University of Rio Grande do Sul - UFRGS, Porto Alegre 90035-003, Brazil
| | - Daniel Pens Gelain
- Center of Oxidative Stress Studies, Department of Biochemistry, Institute of Basic Health Sciences, Federal University of Rio Grande do Sul - UFRGS, Porto Alegre 90035-003, Brazil
| | - Roger Walz
- Center for Applied Neuroscience, University Hospital (HU), Federal University of Santa Catarina (UFSC), Florianópolis, SC, Brazil; Neurology Division, Department of Internal Medicine, University Hospital, Federal University of Santa Catarina (UFSC), Florianópolis, SC, Brazil
| | - Tatiana Barichello
- Translational Psychiatry Program, Faillace Department of Psychiatry and Behavioral Sciences, McGovern Medical School, University of Texas Health Science Center at Houston (UTHealth), Houston, TX 77054, USA
| | - Cristiane Ritter
- Laboratory of Experimental Physiopathology, Graduate Program in Health Sciences, Health Sciences Unit, University of Southern Santa Catarina, Criciuma, SC, Brazil; Hospital São José Research Center, Criciúma, SC, Brazil
| |
Collapse
|
11
|
Walsh JML, Miao VN, Owings AH, Tang Y, Bromley JD, Kazer SW, Kimler K, Asare C, Ziegler CGK, Ibrahim S, Jivanjee T, George M, Navia AW, Drake RS, Parker A, Billingsley BC, Dotherow P, Tarugu S, Kota SK, Laird H, Wichman TG, Davis YT, Dhaliwal NS, Pride Y, Guo Y, Senitko M, Harvey J, Bates JT, Diamond G, Garrett MR, Robinson DA, Frame IJ, Lyons JJ, Robinson TO, Shalek AK, Horwitz BH, Glover SC, Ordovas-Montanes J. Variants and vaccines impact nasal immunity over three waves of SARS-CoV-2. Nat Immunol 2025; 26:294-307. [PMID: 39833605 DOI: 10.1038/s41590-024-02052-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2024] [Accepted: 12/05/2024] [Indexed: 01/22/2025]
Abstract
Viral variant and host vaccination status impact infection with severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), yet how these factors shift cellular responses in the human nasal mucosa remains uncharacterized. We performed single-cell RNA sequencing (scRNA-seq) on nasopharyngeal swabs from vaccinated and unvaccinated adults with acute Delta and Omicron SARS-CoV-2 infections and integrated with data from acute infections with ancestral SARS-CoV-2. Patients with Delta and Omicron exhibited greater similarity in nasal cell composition driven by myeloid, T cell and SARS-CoV-2hi cell subsets, which was distinct from that of ancestral cases. Delta-infected samples had a marked increase in viral RNA, and a subset of PER2+EGR1+GDF15+ epithelial cells was enriched in SARS-CoV-2 RNA+ cells in all variants. Prior vaccination was associated with increased frequency and activation of nasal macrophages. Expression of interferon-stimulated genes negatively correlated with coronavirus disease 2019 (COVID-19) severity in patients with ancestral and Delta but not Omicron variants. Our study defines nasal cell responses and signatures of disease severity across SARS-CoV-2 variants and vaccination.
Collapse
Affiliation(s)
- Jaclyn M L Walsh
- Division of Gastroenterology, Hepatology and Nutrition, Boston Children's Hospital, Boston, MA, USA
- Department of Immunology, Harvard Medical School, Boston, MA, USA
- Program in Immunology, Harvard Medical School, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Vincent N Miao
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA, USA
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA, USA
- Program in Health Sciences and Technology, Harvard Medical School and MIT, Boston, MA, USA
| | - Anna H Owings
- Department of Medicine, University of Mississippi Medical Center, Jackson, MS, USA
- Division of Digestive Diseases, University of Mississippi Medical Center, Jackson, MS, USA
| | - Ying Tang
- Division of Gastroenterology, Hepatology and Nutrition, Boston Children's Hospital, Boston, MA, USA
| | - Joshua D Bromley
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA, USA
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA, USA
- Graduate Program in Microbiology, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Samuel W Kazer
- Division of Gastroenterology, Hepatology and Nutrition, Boston Children's Hospital, Boston, MA, USA
- Department of Immunology, Harvard Medical School, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA, USA
| | - Kyle Kimler
- Division of Gastroenterology, Hepatology and Nutrition, Boston Children's Hospital, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA, USA
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Chemistry, Massachusetts Institute of Technology, Cambridge, MA, USA
- Division of Pediatric Hematology/Oncology, Boston Children's Hospital, Boston, MA, USA
| | - Chelsea Asare
- Division of Gastroenterology, Hepatology and Nutrition, Boston Children's Hospital, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Carly G K Ziegler
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA, USA
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA, USA
- Program in Health Sciences and Technology, Harvard Medical School and MIT, Boston, MA, USA
- Harvard Graduate Program in Biophysics, Cambridge, MA, USA
| | - Samira Ibrahim
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA, USA
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Tasneem Jivanjee
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA, USA
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Micayla George
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA, USA
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Andrew W Navia
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA, USA
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Chemistry, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Riley S Drake
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA, USA
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Adam Parker
- Division of Digestive Diseases, University of Mississippi Medical Center, Jackson, MS, USA
| | | | - Paul Dotherow
- Division of Digestive Diseases, University of Mississippi Medical Center, Jackson, MS, USA
| | - Spurthi Tarugu
- Department of Medicine, University of Mississippi Medical Center, Jackson, MS, USA
| | - Sai K Kota
- Division of Digestive Diseases, University of Mississippi Medical Center, Jackson, MS, USA
| | - Hannah Laird
- Department of Medicine, University of Mississippi Medical Center, Jackson, MS, USA
- Division of Digestive Diseases, University of Mississippi Medical Center, Jackson, MS, USA
| | - T Grant Wichman
- Department of Medicine, University of Mississippi Medical Center, Jackson, MS, USA
| | - Yesenia T Davis
- Division of Digestive Diseases, University of Mississippi Medical Center, Jackson, MS, USA
| | - Neha S Dhaliwal
- Department of Medicine, University of Mississippi Medical Center, Jackson, MS, USA
- Division of Digestive Diseases, University of Mississippi Medical Center, Jackson, MS, USA
| | - Yilianys Pride
- Division of Digestive Diseases, University of Mississippi Medical Center, Jackson, MS, USA
| | - Yanglin Guo
- Division of Pulmonary, Critical Care and Sleep Medicine, University of Mississippi Medical Center, Jackson, MS, USA
| | - Michal Senitko
- Division of Pulmonary, Critical Care and Sleep Medicine, University of Mississippi Medical Center, Jackson, MS, USA
| | - Jessie Harvey
- Department of Medicine, University of Mississippi Medical Center, Jackson, MS, USA
| | - John T Bates
- Department of Cell and Molecular Biology, University of Mississippi Medical Center, Jackson, MS, USA
| | - Gill Diamond
- Department of Oral Immunology and Infectious Diseases, University of Louisville School of Dentistry, Louisville, KY, USA
| | - Michael R Garrett
- Department of Medicine, University of Mississippi Medical Center, Jackson, MS, USA
- Department of Cell and Molecular Biology, University of Mississippi Medical Center, Jackson, MS, USA
| | - D Ashley Robinson
- Department of Cell and Molecular Biology, University of Mississippi Medical Center, Jackson, MS, USA
| | - I J Frame
- Department of Pathology, University of Mississippi Medical Center, Jackson, MS, USA
| | - Jonathan J Lyons
- Division of Allergy and Immunology, Department of Medicine, University of California San Diego, La Jolla, CA, USA
- Veterans Affairs San Diego Healthcare System, La Jolla, CA, USA
| | - Tanya O Robinson
- Division of Digestive Diseases, University of Mississippi Medical Center, Jackson, MS, USA
| | - Alex K Shalek
- Program in Immunology, Harvard Medical School, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA, USA
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA, USA
- Program in Health Sciences and Technology, Harvard Medical School and MIT, Boston, MA, USA
- Department of Chemistry, Massachusetts Institute of Technology, Cambridge, MA, USA
- Harvard Graduate Program in Biophysics, Cambridge, MA, USA
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Bruce H Horwitz
- Division of Gastroenterology, Hepatology and Nutrition, Boston Children's Hospital, Boston, MA, USA
- Program in Immunology, Harvard Medical School, Boston, MA, USA
- Division of Emergency Medicine, Boston Children's Hospital, Boston, MA, USA
| | - Sarah C Glover
- Division of Digestive Diseases, University of Mississippi Medical Center, Jackson, MS, USA
- Department of Cell and Molecular Biology, University of Mississippi Medical Center, Jackson, MS, USA
- Department of Medicine, Section of Gastroenterology and Hepatology, Tulane University, New Orleans, LA, USA
| | - Jose Ordovas-Montanes
- Division of Gastroenterology, Hepatology and Nutrition, Boston Children's Hospital, Boston, MA, USA.
- Program in Immunology, Harvard Medical School, Boston, MA, USA.
- Broad Institute of MIT and Harvard, Cambridge, MA, USA.
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA, USA.
- Harvard Stem Cell Institute, Cambridge, MA, USA.
| |
Collapse
|
12
|
Adilović M, Hromić-Jahjefendić A, Mahmutović L, Šutković J, Rubio-Casillas A, Redwan EM, Uversky VN. Intrinsic Factors Behind the Long-COVID: V. Immunometabolic Disorders. J Cell Biochem 2025; 126:e30683. [PMID: 39639607 DOI: 10.1002/jcb.30683] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2024] [Revised: 11/02/2024] [Accepted: 11/15/2024] [Indexed: 12/07/2024]
Abstract
The complex link between COVID-19 and immunometabolic diseases demonstrates the important interaction between metabolic dysfunction and immunological response during viral infections. Severe COVID-19, defined by a hyperinflammatory state, is greatly impacted by underlying chronic illnesses aggravating the cytokine storm caused by increased levels of Pro-inflammatory cytokines. Metabolic reprogramming, including increased glycolysis and altered mitochondrial function, promotes viral replication and stimulates inflammatory cytokine production, contributing to illness severity. Mitochondrial metabolism abnormalities, strongly linked to various systemic illnesses, worsen metabolic dysfunction during and after the pandemic, increasing cardiovascular consequences. Long COVID-19, defined by chronic inflammation and immune dysregulation, poses continuous problems, highlighting the need for comprehensive therapy solutions that address both immunological and metabolic aspects. Understanding these relationships shows promise for effectively managing COVID-19 and its long-term repercussions, which is the focus of this review paper.
Collapse
Affiliation(s)
- Muhamed Adilović
- Department of Genetics and Bioengineering, Faculty of Engineering and Natural Sciences, International University of Sarajevo, Sarajevo, Bosnia and Herzegovina
| | - Altijana Hromić-Jahjefendić
- Department of Genetics and Bioengineering, Faculty of Engineering and Natural Sciences, International University of Sarajevo, Sarajevo, Bosnia and Herzegovina
| | - Lejla Mahmutović
- Department of Genetics and Bioengineering, Faculty of Engineering and Natural Sciences, International University of Sarajevo, Sarajevo, Bosnia and Herzegovina
| | - Jasmin Šutković
- Department of Genetics and Bioengineering, Faculty of Engineering and Natural Sciences, International University of Sarajevo, Sarajevo, Bosnia and Herzegovina
| | - Alberto Rubio-Casillas
- Autlan Regional Hospital, Health Secretariat, Autlan, Mexico
- Biology Laboratory, Autlan Regional Preparatory School, University of Guadalajara, Autlan, Mexico
| | - Elrashdy M Redwan
- Department of Biological Sciences, Faculty of Science, King Abdulaziz University, Jeddah, Saudi Arabia
- Centre of Excellence in Bionanoscience Research, King Abdulaziz University, Jeddah, Saudi Arabia
- Therapeutic and Protective Proteins Laboratory, Protein Research Department, Genetic Engineering and Biotechnology Research Institute, City of Scientific Research and Technological Applications (SRTA-City), New Borg EL-Arab, Alexandria, Egypt
| | - Vladimir N Uversky
- Department of Molecular Medicine and USF Health Byrd Alzheimer's Research Institute, Morsani College of Medicine, University of South Florida, Tampa, Florida, USA
| |
Collapse
|
13
|
de Bruijn S, Tulen AD, Rodenburg J, Boshuizen H, Schipper M, Mutubuki EN, Knoop H, Franz E, van der Maaden T, van den Hof S, van Hoek AJ, van den Wijngaard CC. Post-acute sequelae of COVID-19 3 to 12 months after infection: Delta vs Omicron. Int J Infect Dis 2025; 150:107302. [PMID: 39549783 DOI: 10.1016/j.ijid.2024.107302] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2024] [Revised: 11/04/2024] [Accepted: 11/11/2024] [Indexed: 11/18/2024] Open
Abstract
OBJECTIVES Studies have shown temporal changes in post-acute sequelae of COVID-19 (PASC) prevalence for early SARS-CoV-2 variants, although often lacking controls. This prospective study assesses the prevalence of symptoms in Delta- and Omicron-infected cases up to 12 months compared with population controls. METHODS Adult participants filled out surveys every 3 months (T0-T12) between July 2021 and August 2023. Cases were recruited with a positive SARS-CoV-2 test during the Delta or Omicron domination. Population controls were randomly invited from the Dutch Personal Records Database. Participants indicated the presence of 13 PASC-associated symptoms, and severity scores of fatigue, cognitive impairment, dyspnea, and pain. PASC prevalence was defined as the excess prevalence of havingat least one PASC-associated symptom in cases compared with population controls. RESULTS PASC prevalence was 34.3% at T3 and decreased to 21.7% at T12 for Delta and decreased from 18.7% at T3 to 16.7% at T12 for Omicron. At T12, the difference between Delta and Omicron was not significant. Delta cases generally had higher excess symptom scores for fatigue, dyspnea, and cognitive impairment than Omicron. CONCLUSIONS In the first 9 months after infection, PASC prevalence was higher for Delta than Omicron, but the difference reduced over time and approximated after 12 months.
Collapse
Affiliation(s)
- Siméon de Bruijn
- Center for Infectious Disease Control, National Institute for Public Health and the Environment (RIVM), Bilthoven, the Netherlands.
| | - Anna D Tulen
- Center for Infectious Disease Control, National Institute for Public Health and the Environment (RIVM), Bilthoven, the Netherlands
| | - Jeroen Rodenburg
- Department of Statistics, Data Science and Mathematical Modelling, National Institute for Public Health and the Environment (RIVM), Bilthoven, the Netherlands
| | - Hendriek Boshuizen
- Department of Statistics, Data Science and Mathematical Modelling, National Institute for Public Health and the Environment (RIVM), Bilthoven, the Netherlands
| | - Maarten Schipper
- Department of Statistics, Data Science and Mathematical Modelling, National Institute for Public Health and the Environment (RIVM), Bilthoven, the Netherlands
| | - Elizabeth N Mutubuki
- Center for Infectious Disease Control, National Institute for Public Health and the Environment (RIVM), Bilthoven, the Netherlands
| | - Hans Knoop
- Department of Medical Psychology and Amsterdam Public Health from the Amsterdam UMC, location University of Amsterdam, Amsterdam, the Netherlands
| | - Eelco Franz
- Center for Infectious Disease Control, National Institute for Public Health and the Environment (RIVM), Bilthoven, the Netherlands
| | - Tessa van der Maaden
- Center for Infectious Disease Control, National Institute for Public Health and the Environment (RIVM), Bilthoven, the Netherlands
| | - Susan van den Hof
- Center for Infectious Disease Control, National Institute for Public Health and the Environment (RIVM), Bilthoven, the Netherlands
| | - Albert Jan van Hoek
- Center for Infectious Disease Control, National Institute for Public Health and the Environment (RIVM), Bilthoven, the Netherlands
| | - Cees C van den Wijngaard
- Center for Infectious Disease Control, National Institute for Public Health and the Environment (RIVM), Bilthoven, the Netherlands
| |
Collapse
|
14
|
Wang Y, Alcalde-Herraiz M, Güell KL, Chen L, Mateu L, Li C, Ali R, Wareham N, Paredes R, Prieto-Alhambra D, Xie J. Refinement of post-COVID condition core symptoms, subtypes, determinants, and health impacts: a cohort study integrating real-world data and patient-reported outcomes. EBioMedicine 2025; 111:105493. [PMID: 39662181 PMCID: PMC11697707 DOI: 10.1016/j.ebiom.2024.105493] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2024] [Revised: 11/25/2024] [Accepted: 11/26/2024] [Indexed: 12/13/2024] Open
Abstract
BACKGROUND Post-COVID-19 condition (PCC) affects millions of people, and is an essential component of the long-term impact of COVID-19 during the post-pandemic era. Yet, consensus on clinical case definition and core components of PCC remains lacking, affecting our ability to inform research and evidence-based management. Our study aims 1) to identify the most specific symptoms for PCC, and identify clinical subtypes; 2) to evaluate both virus- and host-related determinants of PCC, and 3) assess the impact of PCC on physical and mental health. METHODS We studied participants from UK Biobank who completed a health and wellbeing survey between June and September 2022. Participants reported the current conditions of the presence, duration, and functional limitations of 45 symptoms, using an online questionnaire designed specifically for COVID-19 research. SARS-CoV-2 infection status and disease history were obtained through linkage to surveillance data and electronic medical records, respectively. Participants reporting symptoms within 30 days after infection (acute phase) were excluded. The most specific PCC symptoms were defined using two criteria: statistical significance (P < 0.05 after Bonferroni correction) and clinical relevance (absolute risk increase > 5%). Propensity score weighting was used to control for confounding. Subtypes of PCC were then defined based on the specific symptoms among the COVID-19 infected individuals. A multivariable regression was used to study pathogen- and host-related risk factors for PCC, and its impact on 13 physical and 4 mental health patient-reported functional outcomes. FINDINGS 172,303 participants (mean age 68.9, 57.4% female) were included in the analysis, of whom 43,395 had PCR-confirmed COVID-19. We identified 10 most specific symptoms and classified four PCC subtypes: ENT subtype (30.1%), characterized by alterations in smell, taste, and hearing loss; cardiopulmonary subtype (10.4%), characterized by shortness of breath, postural tachycardia, chest tightness, and chest pressure; neurological subtype (23.5%), characterized by brain fog and difficulty speaking; and general fatigue subtype (38.0%), characterized by mild fatigue. A higher PCC risk was observed for patients with Wild-type variant, multiple infections, and severe acute COVID-19 illness, consistently across the four PCC subtypes. In addition, a range of factors, including socioeconomic deprivation, higher BMI, unhealthy lifestyle, and multiple chronic health conditions, were associated with increased PCC risk, except for age and sex. Conversely, vaccination was associated with a largely reduced PCC risk, particularly for the cardiopulmonary subtypes. Individuals with PCC experienced a much worse physical and mental health. Specifically, the cardiopulmonary subtype had the most pronounced adverse impact on function impairments, followed by neurological, mild fatigue, and ENT subtype. The most affected functions included the ability to concentrate, participate in day-to-day work, and emotional vulnerability to health problems. INTERPRETATION PCC can be categorized into four distinct subtypes based on ten core symptoms. These subtypes appeared to share a majority of pathogen and host-related risk factors, but their impact on health varied markedly by subtype. Our findings could help refine current guidelines for precise PCC diagnosis and progression, enhance the identification of PCC subgroups for targeted research, and inform evidence-based policy making to tackle this new and debilitating condition. FUNDING NIHR Senior Research Fellowship (grant SRF-2018-11-ST2-004).
Collapse
Affiliation(s)
- Yunhe Wang
- Nuffield Department of Population Health, University of Oxford, Oxford, UK
| | - Marta Alcalde-Herraiz
- Centre for Statistics in Medicine and NIHR Biomedical Research Centre Oxford, NDORMS, University of Oxford, Oxford, UK
| | - Kim López Güell
- Centre for Statistics in Medicine and NIHR Biomedical Research Centre Oxford, NDORMS, University of Oxford, Oxford, UK
| | - Li Chen
- Centre for Statistics in Medicine and NIHR Biomedical Research Centre Oxford, NDORMS, University of Oxford, Oxford, UK; Institute of Child and Adolescent Health, School of Public Health, Peking University, China
| | - Lourdes Mateu
- Department of Infectious Diseases & irsiCaixa AIDS Research Institute, Hospital Universitari Germans Trias i Pujol, Badalona, Catalonia, Spain; Chair in Infectious Diseases and Immunity, Center for Health and Social Care Research (CEESS), Faculty of Medicine, University of Vic- Central University of Catalonia (UVic-UCC), Spain; Universitat Autònoma de Barcelona, Catalonia, Spain; REICOP (Red de Investigación Covid Persistente), Madrid, Spain
| | - Chunxiao Li
- Medical Research Council Epidemiology Unit, University of Cambridge, Cambridge, UK
| | - Raghib Ali
- Medical Research Council Epidemiology Unit, University of Cambridge, Cambridge, UK
| | - Nicholas Wareham
- Medical Research Council Epidemiology Unit, University of Cambridge, Cambridge, UK
| | - Roger Paredes
- Chair in Infectious Diseases and Immunity, Center for Health and Social Care Research (CEESS), Faculty of Medicine, University of Vic- Central University of Catalonia (UVic-UCC), Spain; Center for Global Health and Diseases, Department of Pathology, Case Western Reserve University School of Medicine, Cleveland, OH, USA
| | - Daniel Prieto-Alhambra
- Centre for Statistics in Medicine and NIHR Biomedical Research Centre Oxford, NDORMS, University of Oxford, Oxford, UK; Department of Medical Informatics, Erasmus University Medical Center, Rotterdam, the Netherlands.
| | - Junqing Xie
- Centre for Statistics in Medicine and NIHR Biomedical Research Centre Oxford, NDORMS, University of Oxford, Oxford, UK
| |
Collapse
|
15
|
Wei Y, Gu H, Ma J, Mao X, Wang B, Wu W, Yu S, Wang J, Zhao H, He Y. Proteomic and metabolomic profiling of plasma uncovers immune responses in patients with Long COVID-19. Front Microbiol 2024; 15:1470193. [PMID: 39802657 PMCID: PMC11718655 DOI: 10.3389/fmicb.2024.1470193] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2024] [Accepted: 11/19/2024] [Indexed: 01/16/2025] Open
Abstract
Long COVID is an often-debilitating condition with severe, multisystem symptoms that can persist for weeks or months and increase the risk of various diseases. Currently, there is a lack of diagnostic tools for Long COVID in clinical practice. Therefore, this study utilizes plasma proteomics and metabolomics technologies to understand the molecular profile and pathophysiological mechanisms of Long COVID, providing clinical evidence for the development of potential biomarkers. This study included three age- and gender-matched cohorts: healthy controls (n = 18), COVID-19 recovered patients (n = 17), and Long COVID patients (n = 15). The proteomics results revealed significant differences in proteins between Long COVID-19 patients and COVID-19 recovered patients, with dysregulation mainly focused on pathways such as coagulation, platelets, complement cascade reactions, GPCR cell signal transduction, and substance transport, which can participate in regulating immune responses, inflammation, and tissue vascular repair. Metabolomics results showed that Long COVID patients and COVID-19 recovered patients have similar metabolic disorders, mainly involving dysregulation in lipid metabolites and fatty acid metabolism, such as glycerophospholipids, sphingolipid metabolism, and arachidonic acid metabolism processes. In summary, our study results indicate significant protein dysregulation and metabolic abnormalities in the plasma of Long COVID patients, leading to coagulation dysfunction, impaired energy metabolism, and chronic immune dysregulation, which are more pronounced than in COVID-19 recovered patients.
Collapse
Affiliation(s)
- Yulin Wei
- Department of Pulmonary and Critical Care Medicine, Affiliated Nantong Hospital of Shanghai University (The Sixth People’s Hospital of Nantong), Nantong, Jiangsu, China
| | - Hongyan Gu
- Department of Pulmonary and Critical Care Medicine, Affiliated Nantong Hospital of Shanghai University (The Sixth People’s Hospital of Nantong), Nantong, Jiangsu, China
| | - Jun Ma
- Department of Pulmonary and Critical Care Medicine, Affiliated Nantong Hospital of Shanghai University (The Sixth People’s Hospital of Nantong), Nantong, Jiangsu, China
| | - Xiaojuan Mao
- Department of Pulmonary and Critical Care Medicine, Affiliated Nantong Hospital of Shanghai University (The Sixth People’s Hospital of Nantong), Nantong, Jiangsu, China
| | - Bing Wang
- Key Laboratory of Digital Technology in Medical Diagnostics of Zhejiang Province, Hangzhou, China
- Dian Diagnostics Group Co., Ltd., Hangzhou, Zhejiang, China
| | - Weiyan Wu
- Key Laboratory of Digital Technology in Medical Diagnostics of Zhejiang Province, Hangzhou, China
- Dian Diagnostics Group Co., Ltd., Hangzhou, Zhejiang, China
| | - Shiming Yu
- Key Laboratory of Digital Technology in Medical Diagnostics of Zhejiang Province, Hangzhou, China
- Dian Diagnostics Group Co., Ltd., Hangzhou, Zhejiang, China
| | - Jinyuan Wang
- Key Laboratory of Digital Technology in Medical Diagnostics of Zhejiang Province, Hangzhou, China
- Dian Diagnostics Group Co., Ltd., Hangzhou, Zhejiang, China
| | - Huan Zhao
- Department of Pulmonary and Critical Care Medicine, Affiliated Nantong Hospital of Shanghai University (The Sixth People’s Hospital of Nantong), Nantong, Jiangsu, China
| | - Yanbin He
- Key Laboratory of Digital Technology in Medical Diagnostics of Zhejiang Province, Hangzhou, China
- Dian Diagnostics Group Co., Ltd., Hangzhou, Zhejiang, China
| |
Collapse
|
16
|
An Y, He L, Xu X, Piao M, Wang B, Liu T, Cao H. Gut microbiota in post-acute COVID-19 syndrome: not the end of the story. Front Microbiol 2024; 15:1500890. [PMID: 39777148 PMCID: PMC11703812 DOI: 10.3389/fmicb.2024.1500890] [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: 09/24/2024] [Accepted: 12/09/2024] [Indexed: 01/11/2025] Open
Abstract
The coronavirus disease 2019 (COVID-19), caused by severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2), has led to major global health concern. However, the focus on immediate effects was assumed as the tip of iceberg due to the symptoms following acute infection, which was defined as post-acute COVID-19 syndrome (PACS). Gut microbiota alterations even after disease resolution and the gastrointestinal symptoms are the key features of PACS. Gut microbiota and derived metabolites disorders may play a crucial role in inflammatory and immune response after SARS-CoV-2 infection through the gut-lung axis. Diet is one of the modifiable factors closely related to gut microbiota and COVID-19. In this review, we described the reciprocal crosstalk between gut and lung, highlighting the participation of diet and gut microbiota in and after COVID-19 by destroying the gut barrier, perturbing the metabolism and regulating the immune system. Therefore, bolstering beneficial species by dietary supplements, probiotics or prebiotics and fecal microbiota transplantation (FMT) may be a novel avenue for COVID-19 and PACS prevention. This review provides a better understanding of the association between gut microbiota and the long-term consequences of COVID-19, which indicates modulating gut dysbiosis may be a potentiality for addressing this multifaceted condition.
Collapse
Affiliation(s)
| | | | | | | | | | - Tianyu Liu
- Tianjin Key Laboratory of Digestive Diseases, Department of Gastroenterology and Hepatology, Tianjin Institute of Digestive Diseases, National Key Clinical Specialty, General Hospital, Tianjin Medical University, Tianjin, China
| | - Hailong Cao
- Tianjin Key Laboratory of Digestive Diseases, Department of Gastroenterology and Hepatology, Tianjin Institute of Digestive Diseases, National Key Clinical Specialty, General Hospital, Tianjin Medical University, Tianjin, China
| |
Collapse
|
17
|
Sinclair JE, Vedelago C, Ryan FJ, Carney M, Redd MA, Lynn MA, Grubor-Bauk B, Cao Y, Henders AK, Chew KY, Gilroy D, Greaves K, Labzin L, Ziser L, Ronacher K, Wallace LM, Zhang Y, Macauslane K, Ellis DJ, Rao S, Burr L, Bain A, Karawita A, Schulz BL, Li J, Lynn DJ, Palpant N, Wuethrich A, Trau M, Short KR. Post-acute sequelae of SARS-CoV-2 cardiovascular symptoms are associated with trace-level cytokines that affect cardiomyocyte function. Nat Microbiol 2024; 9:3135-3147. [PMID: 39478108 PMCID: PMC11602718 DOI: 10.1038/s41564-024-01838-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2024] [Accepted: 09/25/2024] [Indexed: 11/06/2024]
Abstract
An estimated 65 million people globally suffer from post-acute sequelae of COVID-19 (PASC), with many experiencing cardiovascular symptoms (PASC-CVS) like chest pain and heart palpitations. This study examines the role of chronic inflammation in PASC-CVS, particularly in individuals with symptoms persisting over a year after infection. Blood samples from three groups-recovered individuals, those with prolonged PASC-CVS and SARS-CoV-2-negative individuals-revealed that those with PASC-CVS had a blood signature linked to inflammation. Trace-level pro-inflammatory cytokines were detected in the plasma from donors with PASC-CVS 18 months post infection using nanotechnology. Importantly, these trace-level cytokines affected the function of primary human cardiomyocytes. Plasma proteomics also demonstrated higher levels of complement and coagulation proteins in the plasma from patients with PASC-CVS. This study highlights chronic inflammation's role in the symptoms of PASC-CVS.
Collapse
Affiliation(s)
- Jane E Sinclair
- School of Chemistry and Molecular Biosciences, The University of Queensland, St Lucia, Queensland, Australia
| | - Courtney Vedelago
- Centre for Personalised Nanomedicine, Australian Institute for Bioengineering and Nanotechnology (AIBN), The University of Queensland, Brisbane, Queensland, Australia
| | - Feargal J Ryan
- Precision Medicine Theme, South Australian Health and Medical Research Institute, Adelaide, South Australia, Australia
- College of Medicine and Public Health and Flinders Health and Medical Research Institute, Flinders University, Bedford Park, South Australia, Australia
| | - Meagan Carney
- School of Mathematics and Physics, University of Queensland, Brisbane, Queensland, Australia
| | - Meredith A Redd
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland, Australia
| | - Miriam A Lynn
- Precision Medicine Theme, South Australian Health and Medical Research Institute, Adelaide, South Australia, Australia
- College of Medicine and Public Health and Flinders Health and Medical Research Institute, Flinders University, Bedford Park, South Australia, Australia
| | - Branka Grubor-Bauk
- Viral Immunology Group, Adelaide Medical School, University of Adelaide, Adelaide, South Australia, Australia
| | - Yuanzhao Cao
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland, Australia
| | - Anjali K Henders
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland, Australia
| | - Keng Yih Chew
- School of Chemistry and Molecular Biosciences, The University of Queensland, St Lucia, Queensland, Australia
| | - Deborah Gilroy
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland, Australia
| | - Kim Greaves
- Sunshine Coast University Hospital, Queensland Health, Birtinya, Queensland, Australia
- National Centre for Epidemiology and Population Health, ANU College of Health and Medicine, Australian National University, Canberra, Australian Capital Territory, Australia
| | - Larisa Labzin
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland, Australia
| | - Laura Ziser
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland, Australia
| | - Katharina Ronacher
- Australian Infectious Diseases Research Centre, The University of Queensland, St Lucia, Queensland, Australia
- Mater Research Institute - The University of Queensland, Translational Research Institute, Brisbane, Queensland, Australia
| | - Leanne M Wallace
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland, Australia
| | - Yiwen Zhang
- Centre for Personalised Nanomedicine, Australian Institute for Bioengineering and Nanotechnology (AIBN), The University of Queensland, Brisbane, Queensland, Australia
| | - Kyle Macauslane
- School of Chemistry and Molecular Biosciences, The University of Queensland, St Lucia, Queensland, Australia
- Australian Infectious Diseases Research Centre, The University of Queensland, St Lucia, Queensland, Australia
| | - Daniel J Ellis
- School of Chemistry and Molecular Biosciences, The University of Queensland, St Lucia, Queensland, Australia
- Australian Infectious Diseases Research Centre, The University of Queensland, St Lucia, Queensland, Australia
| | - Sudha Rao
- Gene Regulation and Translational Medicine Laboratory, Department of Infection and Inflammation, QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
| | - Lucy Burr
- Mater Research Institute, The University of Queensland, South Brisbane, Queensland, Australia
- Department of Respiratory Medicine, Mater Adult Hospital, South Brisbane, Queensland, Australia
- Faculty of Medicine, University of Queensland, Brisbane, Queensland, Australia
| | - Amanda Bain
- Gene Regulation and Translational Medicine Laboratory, Department of Infection and Inflammation, QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
| | - Anjana Karawita
- Australian Centre for Disease Preparedness, Commonwealth Scientific and Industrial Research Organisation, Geelong, Victoria, Australia
| | - Benjamin L Schulz
- School of Chemistry and Molecular Biosciences, The University of Queensland, St Lucia, Queensland, Australia
- Australian Infectious Diseases Research Centre, The University of Queensland, St Lucia, Queensland, Australia
| | - Junrong Li
- Centre for Personalised Nanomedicine, Australian Institute for Bioengineering and Nanotechnology (AIBN), The University of Queensland, Brisbane, Queensland, Australia
| | - David J Lynn
- Precision Medicine Theme, South Australian Health and Medical Research Institute, Adelaide, South Australia, Australia
- College of Medicine and Public Health and Flinders Health and Medical Research Institute, Flinders University, Bedford Park, South Australia, Australia
| | - Nathan Palpant
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland, Australia
| | - Alain Wuethrich
- Centre for Personalised Nanomedicine, Australian Institute for Bioengineering and Nanotechnology (AIBN), The University of Queensland, Brisbane, Queensland, Australia
| | - Matt Trau
- School of Chemistry and Molecular Biosciences, The University of Queensland, St Lucia, Queensland, Australia
- Centre for Personalised Nanomedicine, Australian Institute for Bioengineering and Nanotechnology (AIBN), The University of Queensland, Brisbane, Queensland, Australia
| | - Kirsty R Short
- School of Chemistry and Molecular Biosciences, The University of Queensland, St Lucia, Queensland, Australia.
- Australian Infectious Diseases Research Centre, The University of Queensland, St Lucia, Queensland, Australia.
- Queensland Immunology Research Centre, The University of Queensland, St Lucia, Queensland, Australia.
| |
Collapse
|
18
|
Narasimhan H, Sun J. Nanotech unveils cytokine traces in post-COVID cardiovascular complications. Nat Immunol 2024; 25:2178-2179. [PMID: 39516646 DOI: 10.1038/s41590-024-02017-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2024]
Affiliation(s)
- Harish Narasimhan
- Beirne B. Carter Center for Immunology Research, University of Virginia, Charlottesville, VA, USA
- Division of Infectious Disease and International Health, Department of Medicine, University of Virginia, Charlottesville, VA, USA
- Department of Microbiology, Immunology and Cancer Biology, University of Virginia, Charlottesville, VA, USA
| | - Jie Sun
- Beirne B. Carter Center for Immunology Research, University of Virginia, Charlottesville, VA, USA.
- Division of Infectious Disease and International Health, Department of Medicine, University of Virginia, Charlottesville, VA, USA.
| |
Collapse
|
19
|
Antar AAR, Cox AL. Translating insights into therapies for Long Covid. Sci Transl Med 2024; 16:eado2106. [PMID: 39536116 DOI: 10.1126/scitranslmed.ado2106] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2024] [Accepted: 10/18/2024] [Indexed: 11/16/2024]
Abstract
Long Covid is defined by a wide range of symptoms that persist after the acute phase of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection. Commonly reported symptoms include fatigue, weakness, postexertional malaise, and cognitive dysfunction, with many other symptoms reported. Symptom range, duration, and severity are highly variable and partially overlap with symptoms of myalgic encephalomyelitis/chronic fatigue syndrome and other post-acute infectious syndromes, highlighting opportunities to define shared mechanisms of pathogenesis. Potential mechanisms of Long Covid are diverse, including persistence of viral reservoirs, dysregulated immune responses, direct viral damage of tissues targeted by SARS-CoV-2, inflammation driven by reactivation of latent viral infections, vascular endothelium activation or dysfunction, and subsequent thromboinflammation, autoimmunity, metabolic derangements, microglial activation, and microbiota dysbiosis. The heterogeneity of symptoms and baseline characteristics of people with Long Covid, as well as the varying states of immunity and therapies given at the time of acute infection, have made etiologies of Long Covid difficult to determine. Here, we examine progress on preclinical models for Long Covid and review progress being made in clinical trials, highlighting the need for large human studies and further development of models to better understand Long Covid. Such studies will inform clinical trials that will define treatments to benefit those living with this condition.
Collapse
Affiliation(s)
- Annukka A R Antar
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Andrea L Cox
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| |
Collapse
|
20
|
Callery EL, Morais CLM, Taylor JV, Challen K, Rowbottom AW. Investigation of Long-Term CD4+ T Cell Receptor Repertoire Changes Following SARS-CoV-2 Infection in Patients with Different Severities of Disease. Diagnostics (Basel) 2024; 14:2330. [PMID: 39451653 PMCID: PMC11507081 DOI: 10.3390/diagnostics14202330] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2024] [Revised: 10/04/2024] [Accepted: 10/16/2024] [Indexed: 10/26/2024] Open
Abstract
BACKGROUND The difference in the immune response to severe acute respiratory syndrome coro-navirus 2 (SARS-CoV-2) in patients with mild versus severe disease remains poorly understood. Recent scientific advances have recognised the vital role of both B cells and T cells; however, many questions remain unanswered, particularly for T cell responses. T cells are essential for helping the generation of SARS-CoV-2 antibody responses but have also been recognised in their own right as a major factor influencing COVID-19 disease outcomes. The examination of T cell receptor (TCR) family differences over a 12-month period in patients with varying COVID-19 disease severity is crucial for understanding T cell responses to SARS-CoV-2. METHODS We applied a machine learning approach to analyse TCR vb family responses in COVID-19 patients (n = 151) across multiple timepoints and disease severities alongside SARS-CoV-2 infection-naïve (healthy control) individ-uals (n = 62). RESULTS Blood samples from hospital in-patients with moderate, severe, or critical disease could be classified with an accuracy of 94%. Furthermore, we identified significant variances in TCR vb family specificities between disease and control subgroups. CONCLUSIONS Our findings suggest advantageous and disadvantageous TCR repertoire patterns in relation to disease severity. Following validation in larger cohorts, our methodology may be useful in detecting protective immunity and the assessment of long-term outcomes, particularly as we begin to unravel the immunological mechanisms leading to post-COVID complications.
Collapse
Affiliation(s)
- Emma L. Callery
- Department of Immunology, Lancashire Teaching Hospitals NHS Foundation, Preston PR2 9HT, UK;
| | - Camilo L. M. Morais
- Institute of Chemistry, Federal University of Rio Grande do Norte, Natal 59072-970, Brazil;
| | - Jemma V. Taylor
- Department of Immunology, Lancashire Teaching Hospitals NHS Foundation, Preston PR2 9HT, UK;
| | - Kirsty Challen
- Department of Emergency Medicine, Lancashire Teaching Hospitals NHS Foundation, Preston PR2 9HT, UK;
| | - Anthony W. Rowbottom
- Department of Immunology, Lancashire Teaching Hospitals NHS Foundation, Preston PR2 9HT, UK;
- School of Medicine, University of Central Lancashire, Preston PR1 2HE, UK
| |
Collapse
|
21
|
Brunel J, Paganini J, Galloux M, Charvet B, Perron H. HERV-W ENV transcription in B cells predicting symptomatic COVID-19 and risk for long COVID can express a full-length protein despite stop codon in mRNA from chromosome X via a ribosome readthrough. Microbes Infect 2024:105431. [PMID: 39419470 DOI: 10.1016/j.micinf.2024.105431] [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: 06/21/2024] [Revised: 10/02/2024] [Accepted: 10/15/2024] [Indexed: 10/19/2024]
Abstract
The human genome comprises 8 % of endogenous retroviruses (HERVs). Though HERVS contribute to physiological functions, copies retained pathogenic potential. The HERV-W ENV protein was shown expressed in patients with worse COVID-19 symptoms and post-COVID syndrome. A significant detection of the mRNA encoding HERV-W ENV from patients with COVID-19 in B cells from RNAseq reads obtained from peripheral blond mononuclear cells. This data stratified with increased COVID-19 symptoms or with post-acute sequelae of COVID-19 (long COVID) after 3 months. The HERV-W ENV-U3R RNA was confirmed to display the best alignment with chromosome X ERVWE2 locus. However, a stop codon precluding its translation was re-addressed after recent understandings of ribosome readthrough mechanisms. Experimental results evidenced that this HERV gene can effectively express a full-length protein in the presence of molecules allowing translation via a readthrough mechanism at the ribosome level. Results not only confirm HERV-W ENV RNA origin in these patients but show for the first time how a defective HERV copy can be translated into a complete protein when specific factors make it possible at the ribosome level. The present proof of concept now requires further studies to identify the factors involved in this newly understood mechanism, following SARS-CoV-2 exposure.
Collapse
Affiliation(s)
- Joanna Brunel
- GeNeuro Innovation, 60A Avenue Rockefeller, 69008, Lyon, France
| | | | | | | | - Hervé Perron
- GeNeuro Innovation, 60A Avenue Rockefeller, 69008, Lyon, France.
| |
Collapse
|
22
|
Dempsey B, Blake HA, Madan I, Stevelink SAM, Greenberg N, Raine R, Rafferty AM, Bhundia R, Wessely S, Lamb D. Post COVID-19 syndrome among 5248 healthcare workers in England: longitudinal findings from NHS CHECK. Occup Environ Med 2024; 81:471-479. [PMID: 39358009 PMCID: PMC11503206 DOI: 10.1136/oemed-2024-109621] [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: 05/23/2024] [Accepted: 09/08/2024] [Indexed: 10/04/2024]
Abstract
OBJECTIVES The objectives of this study were to examine post COVID-19 syndrome (PCS) among healthcare workers (HCWs) in England and explore risk factors for the condition. METHODS Data were collected by National Health Service (NHS) CHECK, a longitudinal study exploring HCWs' mental and physical well-being during and after the COVID-19 pandemic. NHS CHECK collected data at four timepoints: the baseline survey between April 2020 and January 2021, and then three follow-up surveys at approximately 6, 12 and 32 months post baseline. PCS data were collected at 12 and 32 months, while risk factor data were from baseline. HCWs were asked what COVID-19 symptoms they experienced and for how long and were classified as having PCS if they had any symptom for ≥12 weeks. Multilevel regressions were used to examine risk factors for PCS. RESULTS This study included 5248 HCWs. While 33.6% (n=1730) reported prolonged COVID-19 symptoms consistent with PCS, only 7.4% (n=385) reported a formal diagnosis of PCS. Fatigue, difficult concentrating, insomnia and anxiety or depression were the most common PCS symptoms. Baseline risk factors for reporting PCS included screening for common mental disorders, direct contact with COVID-19 patients, pre-existing respiratory illnesses, female sex and older age. CONCLUSIONS While a third of HCWs reported prolonged COVID-19 symptoms consistent with PCS, a smaller percentage reported a formal diagnosis of the condition. We replicate findings that direct contact with COVID-19 patients, older age, female sex, pre-existing respiratory illness and symptoms of common mental disorders are associated with increased risk of PCS.
Collapse
Affiliation(s)
- Brendan Dempsey
- Department of Primary Care and Population Health, University College London, London, UK
| | - Helen A Blake
- Department of Primary Care and Population Health, University College London, London, UK
| | - Ira Madan
- Guys and St Thomas NHS Foundation Trust, London, UK
| | - Sharon A M Stevelink
- Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, UK
| | - Neil Greenberg
- Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, UK
| | - Rosalind Raine
- Department of Primary Care and Population Health, University College London, London, UK
| | - Anne-Marie Rafferty
- Florence Nightingale Faculty of Nursing, Midwifery and Palliative Care, King's College London, London, UK
| | - Rupa Bhundia
- Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, UK
| | - Simon Wessely
- Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, UK
| | - Danielle Lamb
- Department of Primary Care and Population Health, University College London, London, UK
| |
Collapse
|
23
|
Peluso MJ, Deeks SG. Mechanisms of long COVID and the path toward therapeutics. Cell 2024; 187:5500-5529. [PMID: 39326415 PMCID: PMC11455603 DOI: 10.1016/j.cell.2024.07.054] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Revised: 07/30/2024] [Accepted: 07/30/2024] [Indexed: 09/28/2024]
Abstract
Long COVID, a type of post-acute sequelae of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) (PASC) defined by medically unexplained symptoms following infection with SARS-CoV-2, is a newly recognized infection-associated chronic condition that causes disability in some people. Substantial progress has been made in defining its epidemiology, biology, and pathophysiology. However, there is no cure for the tens of millions of people believed to be experiencing long COVID, and industry engagement in developing therapeutics has been limited. Here, we review the current state of knowledge regarding the biology and pathophysiology of long COVID, focusing on how the proposed mechanisms explain the physiology of the syndrome and how they provide a rationale for the implementation of a broad experimental medicine and clinical trials agenda. Progress toward preventing and curing long COVID and other infection-associated chronic conditions will require deep and sustained investment by funders and industry.
Collapse
Affiliation(s)
- Michael J Peluso
- Division of HIV, Infectious Diseases, and Global Medicine, 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.
| |
Collapse
|
24
|
Grant A, Stage E, Blane D, Goss H, Ormerod J, McIver S, Duncan E, Patel G, Campbell A, Manson P, Subramanian G, Cooper K. Four Years in, What Are the Research Priorities for Long COVID? A Research Priority-Setting Partnership Between People With Lived Experience, Carers, Clinicians and Researchers. Health Expect 2024; 27:e70072. [PMID: 39445814 PMCID: PMC11500202 DOI: 10.1111/hex.70072] [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: 06/21/2024] [Revised: 09/26/2024] [Accepted: 10/05/2024] [Indexed: 10/25/2024] Open
Abstract
INTRODUCTION Long COVID is a life-limiting condition that affects 65 million people worldwide. It devastates lives with uncertain illness trajectories, and yet, there are many research uncertainties as there is a lack of understanding of its causes, effective treatments and management plans. We set out to identify current research priorities for people with Long COVID, carers, healthcare professionals and researchers. METHODS A systematic literature review and previous Long COVID priority-setting exercises identified three broad under-researched areas of Long COVID research within the fields of Public Health and Health Services Research: symptoms; managing day-to-day life; and the emotional impact of Long COVID. We disseminated an elicitation survey that asked for research questions in these areas; responses were analysed and summarised into 42 research questions. A survey was then disseminated, asking respondents to prioritise these 42 questions. Workshops were held with people with Long COVID, carers, healthcare professionals and researchers to analyse responses and agree the top 10 priorities. RESULTS The top priorities in order were pharmacological treatment of Long COVID; understanding the pathophysiology; nonpharmacological symptom management; improving public and professional understanding of Long COVID; understanding of the long-term risks of Long COVID; improving financial and social supports; improving understanding of postviral syndromes; diagnostics; service redesign/pathways; and the well-being of children with Long COVID. CONCLUSION Four years into the pandemic, there is an emphasis on the need for research on treatment, understanding and support for people living with Long COVID. PATIENT AND PUBLIC CONTRIBUTION People with Long COVID and carers were involved in the study design, survey design, dissemination, data analysis, interpretation and reviewing and editing the manuscript.
Collapse
Affiliation(s)
- Aileen Grant
- School of HealthRobert Gordon UniversityAberdeenUK
| | - Emma Stage
- School of HealthRobert Gordon UniversityAberdeenUK
| | - David Blane
- General Practice & Primary Care, School of Health & WellbeingUniversity of GlasgowGlasgowUK
| | - Helen Goss
- Long Covid KidsEdinburghUK
- Long Covid ScotlandEdinburghUK
| | | | | | - Edward Duncan
- Nursing Midwifery and Allied Health Professions Research Unit, Faculty of Health Sciences and SportUniversity of StirlingStirlingUK
| | - Gail Patel
- Perth & Kinross long COVID SupportNHS TaysidePerthUK
| | | | - Paul Manson
- Health Services Research Unit, Institute of Applied Health SciencesUniversity of AberdeenForesterhillAberdeenUK
| | | | - Kay Cooper
- School of HealthRobert Gordon UniversityAberdeenUK
| |
Collapse
|
25
|
Rowntree LC, Audsley J, Allen LF, McQuilten HA, Hagen RR, Chaurasia P, Petersen J, Littler DR, Tan HX, Murdiyarso L, Habel JR, Foo IJH, Zhang W, Ten Berge ERV, Ganesh H, Kaewpreedee P, Lee KWK, Cheng SMS, Kwok JSY, Jayasinghe D, Gras S, Juno JA, Wheatley AK, Kent SJ, Rossjohn J, Cheng AC, Kotsimbos TC, Trubiano JA, Holmes NE, Pang Chan KK, Hui DSC, Peiris M, Poon LLM, Lewin SR, Doherty PC, Thevarajan I, Valkenburg SA, Kedzierska K, Nguyen THO. SARS-CoV-2-specific CD8 + T cells from people with long COVID establish and maintain effector phenotype and key TCR signatures over 2 years. Proc Natl Acad Sci U S A 2024; 121:e2411428121. [PMID: 39284068 PMCID: PMC11441481 DOI: 10.1073/pnas.2411428121] [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: 06/13/2024] [Accepted: 07/23/2024] [Indexed: 10/02/2024] Open
Abstract
Long COVID occurs in a small but important minority of patients following COVID-19, reducing quality of life and contributing to healthcare burden. Although research into underlying mechanisms is evolving, immunity is understudied. SARS-CoV-2-specific T cell responses are of key importance for viral clearance and COVID-19 recovery. However, in long COVID, the establishment and persistence of SARS-CoV-2-specific T cells are far from clear, especially beyond 12 mo postinfection and postvaccination. We defined ex vivo antigen-specific B cell and T cell responses and their T cell receptors (TCR) repertoires across 2 y postinfection in people with long COVID. Using 13 SARS-CoV-2 peptide-HLA tetramers, spanning 11 HLA allotypes, as well as spike and nucleocapsid probes, we tracked SARS-CoV-2-specific CD8+ and CD4+ T cells and B-cells in individuals from their first SARS-CoV-2 infection through primary vaccination over 24 mo. The frequencies of ORF1a- and nucleocapsid-specific T cells and B cells remained stable over 24 mo. Spike-specific CD8+ and CD4+ T cells and B cells were boosted by SARS-CoV-2 vaccination, indicating immunization, in fully recovered and people with long COVID, altered the immunodominance hierarchy of SARS-CoV-2 T cell epitopes. Meanwhile, influenza-specific CD8+ T cells were stable across 24 mo, suggesting no bystander-activation. Compared to total T cell populations, SARS-CoV-2-specific T cells were enriched for central memory phenotype, although the proportion of central memory T cells decreased following acute illness. Importantly, TCR repertoire composition was maintained throughout long COVID, including postvaccination, to 2 y postinfection. Overall, we defined ex vivo SARS-CoV-2-specific B cells and T cells to understand primary and recall responses, providing key insights into antigen-specific responses in people with long COVID.
Collapse
Affiliation(s)
- Louise C Rowntree
- Department of Microbiology and Immunology, University of Melbourne, at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC 3000, Australia
| | - Jennifer Audsley
- Department of Infectious Diseases, University of Melbourne, at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC 3000, Australia
| | - Lilith F Allen
- Department of Microbiology and Immunology, University of Melbourne, at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC 3000, Australia
| | - Hayley A McQuilten
- Department of Microbiology and Immunology, University of Melbourne, at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC 3000, Australia
| | - Ruth R Hagen
- Department of Microbiology and Immunology, University of Melbourne, at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC 3000, Australia
| | - Priyanka Chaurasia
- Infection and Immunity Program and Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton, VIC 3800, Australia
| | - Jan Petersen
- Infection and Immunity Program and Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton, VIC 3800, Australia
| | - Dene R Littler
- Infection and Immunity Program and Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton, VIC 3800, Australia
| | - Hyon-Xhi Tan
- Department of Microbiology and Immunology, University of Melbourne, at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC 3000, Australia
| | - Lydia Murdiyarso
- Department of Microbiology and Immunology, University of Melbourne, at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC 3000, Australia
| | - Jennifer R Habel
- Department of Microbiology and Immunology, University of Melbourne, at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC 3000, Australia
| | - Isabelle J H Foo
- Department of Microbiology and Immunology, University of Melbourne, at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC 3000, Australia
| | - Wuji Zhang
- Department of Microbiology and Immunology, University of Melbourne, at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC 3000, Australia
| | - Elizabeth R V Ten Berge
- Department of Microbiology and Immunology, University of Melbourne, at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC 3000, Australia
| | - Hanujah Ganesh
- Department of Infectious Diseases, University of Melbourne, at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC 3000, Australia
| | - Prathanporn Kaewpreedee
- HKU-Pasteur Research Pole, School of Public Health, The University of Hong Kong, Hong Kong Special Administrative Region, China
| | - Kelly W K Lee
- HKU-Pasteur Research Pole, School of Public Health, The University of Hong Kong, Hong Kong Special Administrative Region, China
| | - Samuel M S Cheng
- Division of Public Health Laboratory Sciences, School of Public Health, The University of Hong Kong, Hong Kong Special Administrative Region, China
| | - Janette S Y Kwok
- Division of Transplantation and Immunogenetics, Department of Pathology, Queen Mary Hospital, Hong Kong Special Administrative Region, China
| | - Dhilshan Jayasinghe
- Infection & Immunity Program, La Trobe Institute for Molecular Science, La Trobe University, Bundoora, VIC 3083, Australia
- Department of Biochemistry and Chemistry, School of Agriculture, Biomedicine and Environment, La Trobe University, Bundoora, VIC 3083, Australia
| | - Stephanie Gras
- Infection and Immunity Program and Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton, VIC 3800, Australia
- Infection & Immunity Program, La Trobe Institute for Molecular Science, La Trobe University, Bundoora, VIC 3083, Australia
- Department of Biochemistry and Chemistry, School of Agriculture, Biomedicine and Environment, La Trobe University, Bundoora, VIC 3083, Australia
| | - Jennifer A Juno
- Department of Microbiology and Immunology, University of Melbourne, at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC 3000, Australia
| | - Adam K Wheatley
- Department of Microbiology and Immunology, University of Melbourne, at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC 3000, Australia
| | - Stephen J Kent
- Department of Microbiology and Immunology, University of Melbourne, at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC 3000, Australia
| | - Jamie Rossjohn
- Infection and Immunity Program and Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton, VIC 3800, Australia
- Institute of Infection and Immunity, Cardiff University School of Medicine, Cardiff CF14 4XN, United Kingdom
| | - Allen C Cheng
- School of Public Health and Preventive Medicine, Monash University, Melbourne, VIC 3004, Australia
- Monash Infectious Diseases, Monash Health and School of Clinical Sciences, Monash University, Clayton, VIC 3168, Australia
| | - Tom C Kotsimbos
- Department of Respiratory Medicine, The Alfred Hospital, Melbourne, VIC 3004, Australia
- Department of Medicine, Central Clinical School, The Alfred Hospital, Monash University, Melbourne, VIC 3004, Australia
| | - Jason A Trubiano
- Department of Infectious Diseases, University of Melbourne, at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC 3000, Australia
- Department of Infectious Diseases, Peter MacCallum Cancer Centre, Melbourne, VIC 3000, Australia
- National Centre for Infections in Cancer, Peter McCallum Cancer Centre, Melbourne, VIC 3000, Australia
- Department of Medicine (Austin Health), University of Melbourne, Heidelberg, VIC 3084, Australia
- Centre for Antibiotic Allergy and Research, Department of Infectious Diseases, Austin Health, Heidelberg, VIC 3084, Australia
| | - Natasha E Holmes
- Centre for Antibiotic Allergy and Research, Department of Infectious Diseases, Austin Health, Heidelberg, VIC 3084, Australia
- Department of Critical Care, University of Melbourne, Parkville, VIC 3000, Australia
- Data Analytics Research and Evaluation Centre, Austin Health and University of Melbourne, Heidelberg, VIC 3084, Australia
| | - Ken Ka Pang Chan
- Department of Medicine and Therapeutics, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong Special Administrative Region, China
- Li Ka Shing Institute of Health Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong Special Administrative Region, China
| | - David S C Hui
- Department of Medicine and Therapeutics, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong Special Administrative Region, China
- Li Ka Shing Institute of Health Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong Special Administrative Region, China
| | - Malik Peiris
- HKU-Pasteur Research Pole, School of Public Health, The University of Hong Kong, Hong Kong Special Administrative Region, China
- Division of Public Health Laboratory Sciences, School of Public Health, The University of Hong Kong, Hong Kong Special Administrative Region, China
- Centre for Immunology and Infection, Hong Kong Science and Technology Park, New Territories, Hong Kong Special Administrative Region, China
| | - Leo L M Poon
- HKU-Pasteur Research Pole, School of Public Health, The University of Hong Kong, Hong Kong Special Administrative Region, China
- Division of Public Health Laboratory Sciences, School of Public Health, The University of Hong Kong, Hong Kong Special Administrative Region, China
- Centre for Immunology and Infection, Hong Kong Science and Technology Park, New Territories, Hong Kong Special Administrative Region, China
| | - Sharon R Lewin
- Department of Infectious Diseases, University of Melbourne, at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC 3000, Australia
- Victorian Infectious Diseases Service, Royal Melbourne Hospital, at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC 3000, Australia
- Department of Infectious Disease, Alfred Hospital and Monash University, Melbourne, VIC 3000, Australia
| | - Peter C Doherty
- Department of Microbiology and Immunology, University of Melbourne, at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC 3000, Australia
| | - Irani Thevarajan
- Department of Infectious Diseases, University of Melbourne, at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC 3000, Australia
- Victorian Infectious Diseases Service, Royal Melbourne Hospital, at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC 3000, Australia
| | - Sophie A Valkenburg
- Department of Microbiology and Immunology, University of Melbourne, at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC 3000, Australia
- HKU-Pasteur Research Pole, School of Public Health, The University of Hong Kong, Hong Kong Special Administrative Region, China
| | - Katherine Kedzierska
- Department of Microbiology and Immunology, University of Melbourne, at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC 3000, Australia
| | - Thi H O Nguyen
- Department of Microbiology and Immunology, University of Melbourne, at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC 3000, Australia
| |
Collapse
|
26
|
Avelino-Silva VI, Bruhn R, Zurita KG, Deng X, Yu EA, Grebe E, Stone M, Lanteri MC, Spencer BR, Busch MP, Custer B. SARS-CoV-2 antibody levels and long COVID occurrence in blood donors. Transfusion 2024; 64:1719-1731. [PMID: 38984497 DOI: 10.1111/trf.17952] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2024] [Revised: 06/21/2024] [Accepted: 06/23/2024] [Indexed: 07/11/2024]
Abstract
BACKGROUND Long COVID is a common condition lacking consensus definition; determinants remain incompletely understood. Characterizing immune profiles associated with long COVID could support the development of preventive and therapeutic strategies. METHODS We used a survey to investigate blood donors' infection/vaccination history and acute/persistent symptoms following COVID-19. The prevalence of long COVID was evaluated using self-report and an adapted definition from the RECOVER study. We evaluated factors associated with long COVID, focusing on anti-spike and anti-nucleocapsid SARS-CoV-2 antibodies. Lastly, we investigated long COVID clinical subphenotypes using hierarchical clustering. RESULTS Of 33,610 participants, 16,003 (48%) reported having had COVID-19; 1853 (12%) had self-reported long COVID, 685 (4%) met an adapted RECOVER definition, and 2050 (13%) met at least one definition. Higher anti-nucleocapsid levels measured 12-24 weeks post-infection were associated with higher risk of self-reported and RECOVER long COVID. Higher anti-spike IgG levels measured 12-24 weeks post-infection were associated with lower risk of self-reported long COVID. Higher total anti-spike measured 24-48 weeks post-infection was associated with lower risk of RECOVER long COVID. Cluster analysis identified four clinical subphenotypes; patterns included neurological and psychiatric for cluster 1; neurological and respiratory for cluster 2; multi-systemic for cluster 3; and neurological for cluster 4. DISCUSSION Long COVID prevalence in blood donors varies depending on the adopted definition. Anti-SARS-CoV-2 antibodies were time-dependently associated with long COVID; higher anti-nucleocapsid levels were associated with higher risk; and higher anti-spike levels were associated with lower risk of long COVID. Different underlying pathophysiologic mechanisms may be associated with distinct clinical subphenotypes.
Collapse
Affiliation(s)
- Vivian I Avelino-Silva
- Vitalant Research Institute, California, San Francisco, USA
- Department of Epidemiology and Biostatistics, University of California San Francisco, California, San Francisco, USA
| | - Roberta Bruhn
- Vitalant Research Institute, California, San Francisco, USA
- Department of Laboratory Medicine, University of California San Francisco, California, San Francisco, USA
| | - Karla G Zurita
- Vitalant Research Institute, California, San Francisco, USA
- Department of Laboratory Medicine, University of California San Francisco, California, San Francisco, USA
| | - Xutao Deng
- Vitalant Research Institute, California, San Francisco, USA
- Department of Laboratory Medicine, University of California San Francisco, California, San Francisco, USA
| | - Elaine A Yu
- Vitalant Research Institute, California, San Francisco, USA
- Department of Laboratory Medicine, University of California San Francisco, California, San Francisco, USA
| | - Eduard Grebe
- Vitalant Research Institute, California, San Francisco, USA
- Department of Laboratory Medicine, University of California San Francisco, California, San Francisco, USA
- South African Centre for Epidemiological Modeling and Analysis (SACEMA), Stellenbosch University, Stellenbosch, South Africa
| | - Mars Stone
- Vitalant Research Institute, California, San Francisco, USA
- Department of Laboratory Medicine, University of California San Francisco, California, San Francisco, USA
| | - Marion C Lanteri
- Department of Laboratory Medicine, University of California San Francisco, California, San Francisco, USA
- Creative Testing Solutions, Tempe, Arizona, USA
| | - Bryan R Spencer
- Scientific Affairs, American Red Cross, Rockville, Maryland, USA
| | - Michael P Busch
- Vitalant Research Institute, California, San Francisco, USA
- Department of Laboratory Medicine, University of California San Francisco, California, San Francisco, USA
| | - Brian Custer
- Vitalant Research Institute, California, San Francisco, USA
- Department of Laboratory Medicine, University of California San Francisco, California, San Francisco, USA
| |
Collapse
|
27
|
Xu D, Qin X. Type I Interferonopathy among Non-Elderly Female Patients with Post-Acute Sequelae of COVID-19. Viruses 2024; 16:1369. [PMID: 39339845 PMCID: PMC11435747 DOI: 10.3390/v16091369] [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: 07/29/2024] [Revised: 08/19/2024] [Accepted: 08/26/2024] [Indexed: 09/30/2024] Open
Abstract
The pathophysiological mechanisms of the post-acute sequelae of COVID-19 (PASC) remain unclear. Sex differences not only exist in the disease severity of acute SARS-CoV-2 infection but also in the risk of suffering from PASC. Women have a higher risk of suffering from PASC and a longer time to resolution than men. To explore the possible immune mechanisms of PASC among non-elderly females, we mined single-cell transcriptome data from peripheral blood samples of non-elderly female patients with PASC and acute SARS-CoV-2 infection, together with age- and gender-matched non-PASC and healthy controls available from the Gene Expression Omnibus database. By comparing the differences, we found that a CD14+ monocyte subset characterized by higher expression of signal transducers and activators of transcription 2 (STAT2) (CD14+STAT2high) was notably increased in the PASC patients compared with the non-PASC individuals. The transcriptional factor (TF) activity analysis revealed that STAT2 and IRF9 were the key TFs determining the function of CD14+STAT2high monocytes. STAT2 and IRF9 are TFs exclusively involving type I and III interferon (IFN) signaling pathways, resulting in uncontrolled IFN-I signaling activation and type I interferonopathy. Furthermore, increased expression of common interferon-stimulated genes (ISGs) has also been identified in most monocyte subsets among the non-elderly female PASC patients, including IFI6, IFITM3, IFI44L, IFI44, EPSTI1, ISG15, and MX1. This study reveals a featured CD14+STAT2high monocyte associated with uncontrolled IFN-I signaling activation, which is indicative of a possible type I interferonopathy in the non-elderly female patients with PASC.
Collapse
Affiliation(s)
- Donghua Xu
- Division of Comparative Pathology, Tulane National Primate Research Center, Tulane University School of Medicine, Tulane University, 18703 Three Rivers Road, Covington, LA 70433, USA;
- Department of Microbiology and Immunology, School of Medicine, Tulane University, New Orleans, LA 70112, USA
| | - Xuebin Qin
- Division of Comparative Pathology, Tulane National Primate Research Center, Tulane University School of Medicine, Tulane University, 18703 Three Rivers Road, Covington, LA 70433, USA;
- Department of Microbiology and Immunology, School of Medicine, Tulane University, New Orleans, LA 70112, USA
| |
Collapse
|
28
|
Compeer B, Neijzen TR, van Lelyveld SFL, Martina BEE, Russell CA, Goeijenbier M. Uncovering the Contrasts and Connections in PASC: Viral Load and Cytokine Signatures in Acute COVID-19 versus Post-Acute Sequelae of SARS-CoV-2 (PASC). Biomedicines 2024; 12:1941. [PMID: 39335455 PMCID: PMC11428903 DOI: 10.3390/biomedicines12091941] [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: 07/09/2024] [Revised: 08/13/2024] [Accepted: 08/20/2024] [Indexed: 09/30/2024] Open
Abstract
The recent global COVID-19 pandemic has had a profound and enduring impact, resulting in substantial loss of life. The scientific community has responded unprecedentedly by investigating various aspects of the crisis, particularly focusing on the acute phase of COVID-19. The roles of the viral load, cytokines, and chemokines during the acute phase and in the context of patients who experienced enduring symptoms upon infection, so called Post-Acute Sequelae of COVID-19 or PASC, have been studied extensively. Here, in this review, we offer a virologist's perspective on PASC, highlighting the dynamics of SARS-CoV-2 viral loads, cytokines, and chemokines in different organs of patients across the full clinical spectrum of acute-phase disease. We underline that the probability of severe or critical disease progression correlates with increased viral load levels detected in the upper respiratory tract (URT), lower respiratory tract (LRT), and plasma. Acute-phase viremia is a clear, although not unambiguous, predictor of PASC development. Moreover, both the quantity and diversity of functions of cytokines and chemokines increase with acute-phase disease severity. Specific cytokines remain or become elevated in the PASC phase, although the driving factor of ongoing inflammation found in patients with PASC remains to be investigated. The key findings highlighted in this review contribute to a further understanding of PASC and their differences and overlap with acute disease.
Collapse
Affiliation(s)
- Brandon Compeer
- Artemis Bioservices B.V., 2629 JD Delft, The Netherlands
- Department of Medical Microbiology, University Medical Center Amsterdam (UMC, Amsterdam), 1105 AZ Amsterdam, The Netherlands
| | - Tobias R Neijzen
- Department of Intensive Care Medicine, Spaarne Gasthuis, 2035 RC Haarlem, The Netherlands
| | | | | | - Colin A Russell
- Department of Medical Microbiology, University Medical Center Amsterdam (UMC, Amsterdam), 1105 AZ Amsterdam, The Netherlands
| | - Marco Goeijenbier
- Department of Medical Microbiology, University Medical Center Amsterdam (UMC, Amsterdam), 1105 AZ Amsterdam, The Netherlands
- Department of Intensive Care, Erasmus MC University Medical Centre, 3015 GD Rotterdam, The Netherlands
| |
Collapse
|
29
|
Greenhalgh T, Sivan M, Perlowski A, Nikolich JŽ. Long COVID: a clinical update. Lancet 2024; 404:707-724. [PMID: 39096925 DOI: 10.1016/s0140-6736(24)01136-x] [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: 01/27/2024] [Revised: 05/07/2024] [Accepted: 05/30/2024] [Indexed: 08/05/2024]
Abstract
Post-COVID-19 condition (also known as long COVID) is generally defined as symptoms persisting for 3 months or more after acute COVID-19. Long COVID can affect multiple organ systems and lead to severe and protracted impairment of function as a result of organ damage. The burden of this disease, both on the individual and on health systems and national economies, is high. In this interdisciplinary Review, with a coauthor with lived experience of severe long COVID, we sought to bring together multiple streams of literature on the epidemiology, pathophysiology (including the hypothesised mechanisms of organ damage), lived experience and clinical manifestations, and clinical investigation and management of long COVID. Although current approaches to long COVID care are largely symptomatic and supportive, recent advances in clinical phenotyping, deep molecular profiling, and biomarker identification might herald a more mechanism-informed and personally tailored approach to clinical care. We also cover the organisation of services for long COVID, approaches to preventing long COVID, and suggestions for future research.
Collapse
Affiliation(s)
- Trisha Greenhalgh
- Nuffield Department of Primary Care Health Sciences, Radcliffe Observatory Quarter, Oxford, UK.
| | - Manoj Sivan
- Academic Department of Rehabilitation Medicine, Leeds Institute of Rheumatic and Musculoskeletal Medicine University of Leeds, Leeds General Infirmary, Leeds, UK
| | | | - Janko Ž Nikolich
- Department of Immunobiology and University of Arizona Center on Aging, University of Arizona College of Medicine-Tucson, Tucson, AZ, USA; The Aegis Consortium for Pandemic-Free Future, University of Arizona Health Sciences, Tucson, AZ, USA
| |
Collapse
|
30
|
Shafqat A, Masters MC, Tripathi U, Tchkonia T, Kirkland JL, Hashmi SK. Long COVID as a disease of accelerated biological aging: An opportunity to translate geroscience interventions. Ageing Res Rev 2024; 99:102400. [PMID: 38945306 DOI: 10.1016/j.arr.2024.102400] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2024] [Revised: 06/12/2024] [Accepted: 06/27/2024] [Indexed: 07/02/2024]
Abstract
It has been four years since long COVID-the protracted consequences that survivors of COVID-19 face-was first described. Yet, this entity continues to devastate the quality of life of an increasing number of COVID-19 survivors without any approved therapy and a paucity of clinical trials addressing its biological root causes. Notably, many of the symptoms of long COVID are typically seen with advancing age. Leveraging this similarity, we posit that Geroscience-which aims to target the biological drivers of aging to prevent age-associated conditions as a group-could offer promising therapeutic avenues for long COVID. Bearing this in mind, this review presents a translational framework for studying long COVID as a state of effectively accelerated biological aging, identifying research gaps and offering recommendations for future preclinical and clinical studies.
Collapse
Affiliation(s)
- Areez Shafqat
- College of Medicine, Alfaisal University, Riyadh, Saudi Arabia.
| | - Mary Clare Masters
- Division of Infectious Diseases, Northwestern University, Chicago, IL, USA
| | - Utkarsh Tripathi
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, MN, USA
| | - Tamara Tchkonia
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, MN, USA; Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN, USA
| | - James L Kirkland
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, MN, USA; Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN, USA; Department of Internal Medicine, Mayo Clinic, Rochester, MN, USA
| | - Shahrukh K Hashmi
- Department of Internal Medicine, Mayo Clinic, Rochester, MN, USA; Research and Innovation Center, Department of Health, Abu Dhabi, UAE; College of Medicine and Health Sciences, Khalifa University, Abu Dhabi, United Arab Emirates
| |
Collapse
|
31
|
Al-Aly Z, Davis H, McCorkell L, Soares L, Wulf-Hanson S, Iwasaki A, Topol EJ. Long COVID science, research and policy. Nat Med 2024; 30:2148-2164. [PMID: 39122965 DOI: 10.1038/s41591-024-03173-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2024] [Accepted: 07/02/2024] [Indexed: 08/12/2024]
Abstract
Long COVID represents the constellation of post-acute and long-term health effects caused by SARS-CoV-2 infection; it is a complex, multisystem disorder that can affect nearly every organ system and can be severely disabling. The cumulative global incidence of long COVID is around 400 million individuals, which is estimated to have an annual economic impact of approximately $1 trillion-equivalent to about 1% of the global economy. Several mechanistic pathways are implicated in long COVID, including viral persistence, immune dysregulation, mitochondrial dysfunction, complement dysregulation, endothelial inflammation and microbiome dysbiosis. Long COVID can have devastating impacts on individual lives and, due to its complexity and prevalence, it also has major ramifications for health systems and economies, even threatening progress toward achieving the Sustainable Development Goals. Addressing the challenge of long COVID requires an ambitious and coordinated-but so far absent-global research and policy response strategy. In this interdisciplinary review, we provide a synthesis of the state of scientific evidence on long COVID, assess the impacts of long COVID on human health, health systems, the economy and global health metrics, and provide a forward-looking research and policy roadmap.
Collapse
Affiliation(s)
- Ziyad Al-Aly
- VA St. Louis Health Care System, Saint Louis, MO, USA.
- Washington University in St. Louis, Saint Louis, MO, USA.
| | - Hannah Davis
- Patient-led Research Collaborative, Calabasas, CA, USA
| | | | | | | | - Akiko Iwasaki
- Yale University, New Haven, CT, USA
- Howard Hughes Medical Institute, Chevy Chase, MD, USA
| | - Eric J Topol
- Scripps Institute, San Diego, California, CA, USA
| |
Collapse
|
32
|
Kulkarni DH, Starick M, Aponte Alburquerque R, Kulkarni HS. Local complement activation and modulation in mucosal immunity. Mucosal Immunol 2024; 17:739-751. [PMID: 38838816 PMCID: PMC11929374 DOI: 10.1016/j.mucimm.2024.05.006] [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: 01/01/2024] [Revised: 05/24/2024] [Accepted: 05/29/2024] [Indexed: 06/07/2024]
Abstract
The complement system is an evolutionarily conserved arm of innate immunity, which forms one of the first lines of host response to pathogens and assists in the clearance of debris. A deficiency in key activators/amplifiers of the cascade results in recurrent infection, whereas a deficiency in regulating the cascade predisposes to accelerated organ failure, as observed in colitis and transplant rejection. Given that there are over 60 proteins in this system, it has become an attractive target for immunotherapeutics, many of which are United States Food and Drug Administration-approved or in multiple phase 2/3 clinical trials. Moreover, there have been key advances in the last few years in the understanding of how the complement system operates locally in tissues, independent of its activities in circulation. In this review, we will put into perspective the abovementioned discoveries to optimally modulate the spatiotemporal nature of complement activation and regulation at mucosal surfaces.
Collapse
Affiliation(s)
- Devesha H Kulkarni
- Division of Gastroenterology, Washington University School of Medicine, St. Louis, MO, USA
| | - Marick Starick
- Division of Pulmonary and Critical Care Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - Rafael Aponte Alburquerque
- Division of Pulmonary and Critical Care Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - Hrishikesh S Kulkarni
- Division of Pulmonary and Critical Care Medicine, Washington University School of Medicine, St. Louis, MO, USA.
| |
Collapse
|
33
|
Haddad NS, Morrison-Porter A, Quehl H, Capric V, Lamothe PA, Anam F, Runnstrom MC, Truong AD, Dixit AN, Woodruff MC, Chen A, Park J, Nguyen DC, Hentenaar I, Kim CY, Kyu S, Stewart B, Wagman E, Geoffroy H, Sanz D, Cashman KS, Ramonell RP, Cabrera-Mora M, Alter DN, Roback JD, Horwath MC, O’Keefe JB, Dretler AW, Gripaldo R, Yeligar SM, Natoli T, Betin V, Patel R, Vela K, Hernandez MR, Usman S, Varghese J, Jalal A, Lee S, Le SN, Amoss RT, Daiss JL, Sanz I, Lee FEH. MENSA, a Media Enriched with Newly Synthesized Antibodies, to Identify SARS-CoV-2 Persistence and Latent Viral Reactivation in Long-COVID. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.07.05.24310017. [PMID: 39006446 PMCID: PMC11245097 DOI: 10.1101/2024.07.05.24310017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/16/2024]
Abstract
Post-acute sequelae of SARS-CoV-2 (SARS2) infection (PASC) is a heterogeneous condition, but the main viral drivers are unknown. Here, we use MENSA, Media Enriched with Newly Synthesized Antibodies, secreted exclusively from circulating human plasmablasts, to provide an immune snapshot that defines the underlying viral triggers. We provide proof-of-concept testing that the MENSA technology can capture the new host immune response to accurately diagnose acute primary and breakthrough infections when known SARS2 virus or proteins are present. It is also positive after vaccination when spike proteins elicit an acute immune response. Applying the same principles for long-COVID patients, MENSA is positive for SARS2 in 40% of PASC vs none of the COVID recovered (CR) patients without any sequelae demonstrating ongoing SARS2 viral inflammation only in PASC. Additionally, in PASC patients, MENSAs are also positive for Epstein-Barr Virus (EBV) in 37%, Human Cytomegalovirus (CMV) in 23%, and herpes simplex virus 2 (HSV2) in 15% compared to 17%, 4%, and 4% in CR controls respectively. Combined, a total of 60% of PASC patients have a positive MENSA for SARS2, EBV, CMV, and/or HSV2. MENSA offers a unique antibody snapshot to reveal the underlying viral drivers in long-COVID thus demonstrating the persistence of SARS2 and reactivation of viral herpes in 60% of PASC patients.
Collapse
Affiliation(s)
- Natalie S. Haddad
- Division of Pulmonary, Allergy, Critical Care & Sleep Medicine, Department of Medicine, Emory University, Atlanta, GA, 30322, USA
- MicroB-plex Inc, Atlanta, GA, 30332, USA
| | - Andrea Morrison-Porter
- Division of Pulmonary, Allergy, Critical Care & Sleep Medicine, Department of Medicine, Emory University, Atlanta, GA, 30322, USA
- MicroB-plex Inc, Atlanta, GA, 30332, USA
| | - Hannah Quehl
- Division of Pulmonary, Allergy, Critical Care & Sleep Medicine, Department of Medicine, Emory University, Atlanta, GA, 30322, USA
| | - Violeta Capric
- Division of Pulmonary, Allergy, Critical Care & Sleep Medicine, Department of Medicine, Emory University, Atlanta, GA, 30322, USA
| | - Pedro A. Lamothe
- Division of Pulmonary, Allergy, Critical Care & Sleep Medicine, Department of Medicine, Emory University, Atlanta, GA, 30322, USA
| | - Fabliha Anam
- Division of Rheumatology, Department of Medicine, Emory University, Atlanta, GA, 30322, USA
| | - Martin C. Runnstrom
- Division of Pulmonary, Allergy, Critical Care & Sleep Medicine, Department of Medicine, Emory University, Atlanta, GA, 30322, USA
- Department of Medicine, Atlanta Veterans Affairs Health Care System, Decatur, Georgia, 30033, USA
| | - Alex D. Truong
- Division of Pulmonary, Allergy, Critical Care & Sleep Medicine, Department of Medicine, Emory University, Atlanta, GA, 30322, USA
| | - Adviteeya N. Dixit
- Division of Pulmonary, Allergy, Critical Care & Sleep Medicine, Department of Medicine, Emory University, Atlanta, GA, 30322, USA
| | - Matthew C. Woodruff
- Division of Rheumatology, Department of Medicine, Emory University, Atlanta, GA, 30322, USA
- Lowance Center for Human Immunology, Emory University, Atlanta, GA, 30322, USA
| | - Anting Chen
- Division of Pulmonary, Allergy, Critical Care & Sleep Medicine, Department of Medicine, Emory University, Atlanta, GA, 30322, USA
| | - Jiwon Park
- Division of Pulmonary, Allergy, Critical Care & Sleep Medicine, Department of Medicine, Emory University, Atlanta, GA, 30322, USA
| | - Doan C. Nguyen
- Division of Pulmonary, Allergy, Critical Care & Sleep Medicine, Department of Medicine, Emory University, Atlanta, GA, 30322, USA
| | - Ian Hentenaar
- Division of Pulmonary, Allergy, Critical Care & Sleep Medicine, Department of Medicine, Emory University, Atlanta, GA, 30322, USA
| | - Caroline Y. Kim
- Division of Pulmonary, Allergy, Critical Care & Sleep Medicine, Department of Medicine, Emory University, Atlanta, GA, 30322, USA
| | - Shuya Kyu
- Division of Pulmonary, Allergy, Critical Care & Sleep Medicine, Department of Medicine, Emory University, Atlanta, GA, 30322, USA
- Department of Pathology and Laboratory Medicine, Emory University, Atlanta, GA, 30322, USA
| | - Brandon Stewart
- Division of Pulmonary, Allergy, Critical Care & Sleep Medicine, Department of Medicine, Emory University, Atlanta, GA, 30322, USA
| | - Elizabeth Wagman
- Division of Pulmonary, Allergy, Critical Care & Sleep Medicine, Department of Medicine, Emory University, Atlanta, GA, 30322, USA
| | - Hannah Geoffroy
- Division of Pulmonary, Allergy, Critical Care & Sleep Medicine, Department of Medicine, Emory University, Atlanta, GA, 30322, USA
| | | | - Kevin S. Cashman
- Division of Rheumatology, Department of Medicine, Emory University, Atlanta, GA, 30322, USA
- Lowance Center for Human Immunology, Emory University, Atlanta, GA, 30322, USA
| | - Richard P. Ramonell
- Division of Pulmonary, Allergy, Critical Care & Sleep Medicine, Department of Medicine, Emory University, Atlanta, GA, 30322, USA
- Division of Pulmonary, Allergy, Critical Care, and Sleep Medicine, Department of Medicine, University of Pittsburgh, Pittsburgh, PA, 15213, USA
- Asthma and Environmental Lung Health Institute, University of Pittsburgh Medical Center, Pittsburgh, PA, 15213, USA
| | - Monica Cabrera-Mora
- Division of Pulmonary, Allergy, Critical Care & Sleep Medicine, Department of Medicine, Emory University, Atlanta, GA, 30322, USA
| | - David N. Alter
- Department of Pathology and Laboratory Medicine, Emory University, Atlanta, GA, 30322, USA
| | - John D. Roback
- Department of Pathology and Laboratory Medicine, Emory University, Atlanta, GA, 30322, USA
| | - Michael C. Horwath
- Department of Pathology and Laboratory Medicine, Emory University, Atlanta, GA, 30322, USA
| | - James B. O’Keefe
- Division of General Internal Medicine, Department of Medicine, Emory University School of Medicine, Atlanta, GA, 30322, USA
| | | | - Ria Gripaldo
- Division of Pulmonary, Allergy, Critical Care & Sleep Medicine, Department of Medicine, Emory University, Atlanta, GA, 30322, USA
| | - Samantha M. Yeligar
- Division of Pulmonary, Allergy, Critical Care & Sleep Medicine, Department of Medicine, Emory University, Atlanta, GA, 30322, USA
- Department of Medicine, Atlanta Veterans Affairs Health Care System, Decatur, Georgia, 30033, USA
| | - Ted Natoli
- ImmuneID, Inc Biotechnology Research, Waltham, MA, 02451, USA
| | - Viktoria Betin
- ImmuneID, Inc Biotechnology Research, Waltham, MA, 02451, USA
| | - Rahulkumar Patel
- Division of Pulmonary, Allergy, Critical Care & Sleep Medicine, Department of Medicine, Emory University, Atlanta, GA, 30322, USA
- Division of Rheumatology, Department of Medicine, Emory University, Atlanta, GA, 30322, USA
| | - Kennedy Vela
- Division of Pulmonary, Allergy, Critical Care & Sleep Medicine, Department of Medicine, Emory University, Atlanta, GA, 30322, USA
- Division of Rheumatology, Department of Medicine, Emory University, Atlanta, GA, 30322, USA
| | - Mindy Rodriguez Hernandez
- Division of Pulmonary, Allergy, Critical Care & Sleep Medicine, Department of Medicine, Emory University, Atlanta, GA, 30322, USA
- Division of Rheumatology, Department of Medicine, Emory University, Atlanta, GA, 30322, USA
| | - Sabeena Usman
- Division of Pulmonary, Allergy, Critical Care & Sleep Medicine, Department of Medicine, Emory University, Atlanta, GA, 30322, USA
- Division of Rheumatology, Department of Medicine, Emory University, Atlanta, GA, 30322, USA
| | - John Varghese
- Division of Pulmonary, Allergy, Critical Care & Sleep Medicine, Department of Medicine, Emory University, Atlanta, GA, 30322, USA
- Division of Rheumatology, Department of Medicine, Emory University, Atlanta, GA, 30322, USA
| | - Anum Jalal
- Division of Pulmonary, Allergy, Critical Care & Sleep Medicine, Department of Medicine, Emory University, Atlanta, GA, 30322, USA
- Division of Rheumatology, Department of Medicine, Emory University, Atlanta, GA, 30322, USA
| | - Saeyun Lee
- Division of Pulmonary, Allergy, Critical Care & Sleep Medicine, Department of Medicine, Emory University, Atlanta, GA, 30322, USA
- Division of Rheumatology, Department of Medicine, Emory University, Atlanta, GA, 30322, USA
| | - Sang N. Le
- Division of Pulmonary, Allergy, Critical Care & Sleep Medicine, Department of Medicine, Emory University, Atlanta, GA, 30322, USA
| | - R. Toby Amoss
- Division of Pulmonary, Allergy, Critical Care & Sleep Medicine, Department of Medicine, Emory University, Atlanta, GA, 30322, USA
- Division of Rheumatology, Department of Medicine, Emory University, Atlanta, GA, 30322, USA
| | | | - Ignacio Sanz
- Division of Rheumatology, Department of Medicine, Emory University, Atlanta, GA, 30322, USA
- Lowance Center for Human Immunology, Emory University, Atlanta, GA, 30322, USA
| | - F. Eun-Hyung Lee
- Division of Pulmonary, Allergy, Critical Care & Sleep Medicine, Department of Medicine, Emory University, Atlanta, GA, 30322, USA
- Lowance Center for Human Immunology, Emory University, Atlanta, GA, 30322, USA
| |
Collapse
|
34
|
Ceglarek L, Boyman O. Immune dysregulation in long COVID. Nat Immunol 2024; 25:587-589. [PMID: 38589620 DOI: 10.1038/s41590-024-01795-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/10/2024]
Affiliation(s)
- Laura Ceglarek
- Department of Immunology, University of Zurich, University Hospital Zurich, Zurich, Switzerland
| | - Onur Boyman
- Department of Immunology, University of Zurich, University Hospital Zurich, Zurich, Switzerland.
- Faculty of Medicine and Faculty of Science, University of Zurich, Zurich, Switzerland.
| |
Collapse
|
35
|
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
|