1
|
Jiang Y, Xie Y, You Y, Han B. Mechanistic insights into COVID-19 mRNA vaccine-associated myocarditis: a bioinformatics analysis. Am J Transl Res 2025; 17:2339-2351. [PMID: 40226040 PMCID: PMC11982872 DOI: 10.62347/ccvm6311] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2025] [Accepted: 03/08/2025] [Indexed: 04/15/2025]
Abstract
PURPOSE While COVID-19 vaccination offers significant public health benefits, it also has potential risks, such as myocarditis. The mechanisms underlying myocarditis after COVID-19 vaccination remain poorly understood. The purpose of this study was to identify potential pathogenic genes and molecular pathways related with COVID-19 mRNA vaccine-associated myocarditis. METHODS Differentially expressed genes (DEGs) were analyzed from a fulminant myocarditis (FM) cohort and a COVID-19 mRNA vaccination dataset. Shared DEGs were intersected, followed by functional enrichment, protein-protein interaction (PPI) network construction, and hub gene identification. Transcriptional and miRNA regulatory networks, as well as therapeutic drug predictions were also performed. RESULTS Eighty shared DEGs were identified by interacting DEGs from the FM cohort and the vaccination cohort, we identified. Functional enrichment analysis revealed that DEGs are significantly involved in immune cell-mediated responses, highlighting the critical role of immune dysregulation. PPI network analysis revealed three hub genes (CXCR3, NKG7, and GZMH), which may be involved in the pathogenesis of vaccine-associated myocarditis. Furthermore, transcriptional networks highlighted TBX21 and STAT4 as key regulators of all hub genes, while hsa-mir-146a-5p targeted CXCR3 and NKG7. PhIP, a compound targeting CXCR3 and NKG7, emerged as a potential therapeutic candidate. CONCLUSION This study implicates immune dysregulation driven by CXCR3, NKG7, and GZMH in post-vaccination myocarditis, supported by regulatory networks and therapeutic insights using a bioinformatics analysis. These findings advance mechanistic understanding of this rare adverse event and propose potential treatment strategy for further investigation.
Collapse
Affiliation(s)
- Yanjie Jiang
- Department of Pediatrics, Shandong Provincial Hospital, Shandong UniversityJinan, Shandong, China
- Department of Pediatrics, The Affiliated Taian City Central Hospital of Qingdao UniversityTai’an, Shandong, China
| | - Yaxue Xie
- Department of Pediatrics, Shandong Provincial Hospital, Shandong UniversityJinan, Shandong, China
| | - Yingnan You
- Department of Pediatrics, Shandong Provincial Hospital, Shandong UniversityJinan, Shandong, China
- Department of Pediatrics, Shandong Provincial Hospital Affiliated to Shandong First Medical UniversityJinan, Shandong, China
| | - Bo Han
- Department of Pediatrics, Shandong Provincial Hospital, Shandong UniversityJinan, Shandong, China
- Department of Pediatrics, Shandong Provincial Hospital Affiliated to Shandong First Medical UniversityJinan, Shandong, China
| |
Collapse
|
2
|
Maatz H, Lindberg EL, Adami E, López-Anguita N, Perdomo-Sabogal A, Cocera Ortega L, Patone G, Reichart D, Myronova A, Schmidt S, Elsanhoury A, Klein O, Kühl U, Wyler E, Landthaler M, Yousefian S, Haas S, Kurth F, Teichmann SA, Oudit GY, Milting H, Noseda M, Seidman JG, Seidman CE, Heidecker B, Sander LE, Sawitzki B, Klingel K, Doeblin P, Kelle S, Van Linthout S, Hubner N, Tschöpe C. The cellular and molecular cardiac tissue responses in human inflammatory cardiomyopathies after SARS-CoV-2 infection and COVID-19 vaccination. NATURE CARDIOVASCULAR RESEARCH 2025; 4:330-345. [PMID: 39994453 PMCID: PMC11913730 DOI: 10.1038/s44161-025-00612-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/12/2024] [Accepted: 01/10/2025] [Indexed: 02/26/2025]
Abstract
Myocarditis, characterized by inflammatory cell infiltration, can have multiple etiologies, including severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection or, rarely, mRNA-based coronavirus disease 2019 (COVID-19) vaccination. The underlying cellular and molecular mechanisms remain poorly understood. In this study, we performed single-nucleus RNA sequencing on left ventricular endomyocardial biopsies from patients with myocarditis unrelated to COVID-19 (Non-COVID-19), after SARS-CoV-2 infection (Post-COVID-19) and after COVID-19 vaccination (Post-Vaccination). We identified distinct cytokine expression patterns, with interferon-γ playing a key role in Post-COVID-19, and upregulated IL16 and IL18 expression serving as a hallmark of Post-Vaccination myocarditis. Although myeloid responses were similar across all groups, the Post-Vaccination group showed a higher proportion of CD4+ T cells, and the Post-COVID-19 group exhibited an expansion of cytotoxic CD8+ T and natural killer cells. Endothelial cells showed gene expression changes indicative of vascular barrier dysfunction in the Post-COVID-19 group and ongoing angiogenesis across all groups. These findings highlight shared and distinct mechanisms driving myocarditis in patients with and without a history of SARS-CoV-2 infection or vaccination.
Collapse
Affiliation(s)
- Henrike Maatz
- Cardiovascular and Metabolic Sciences, Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany.
- DZHK (German Centre for Cardiovascular Research), partner site Berlin, Berlin, Germany.
| | - Eric L Lindberg
- Cardiovascular and Metabolic Sciences, Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
- Department of Medicine I, University Hospital, LMU Munich, Munich, Germany
| | - Eleonora Adami
- Cardiovascular and Metabolic Sciences, Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
| | - Natalia López-Anguita
- Cardiovascular and Metabolic Sciences, Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
| | - Alvaro Perdomo-Sabogal
- Cardiovascular and Metabolic Sciences, Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
| | - Lucía Cocera Ortega
- Cardiovascular and Metabolic Sciences, Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
| | - Giannino Patone
- Cardiovascular and Metabolic Sciences, Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
| | - Daniel Reichart
- Department of Medicine I, University Hospital, LMU Munich, Munich, Germany
- Department of Genetics, Harvard Medical School, Boston, MA, USA
- Cardiovascular Division, Brigham and Women's Hospital Boston, Boston, MA, USA
| | - Anna Myronova
- Cardiovascular and Metabolic Sciences, Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
| | - Sabine Schmidt
- Cardiovascular and Metabolic Sciences, Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
| | - Ahmed Elsanhoury
- DZHK (German Centre for Cardiovascular Research), partner site Berlin, Berlin, Germany
- Berlin Institute of Health (BIH) at Charité - Universitätsmedizin Berlin, BIH Center for Regenerative Therapies (BCRT), Berlin, Germany
| | - Oliver Klein
- DZHK (German Centre for Cardiovascular Research), partner site Berlin, Berlin, Germany
- Berlin Institute of Health (BIH) at Charité - Universitätsmedizin Berlin, BIH Center for Regenerative Therapies (BCRT), Berlin, Germany
| | - Uwe Kühl
- DZHK (German Centre for Cardiovascular Research), partner site Berlin, Berlin, Germany
- Berlin Institute of Health (BIH) at Charité - Universitätsmedizin Berlin, BIH Center for Regenerative Therapies (BCRT), Berlin, Germany
| | - Emanuel Wyler
- Berlin Institute for Medical Systems Biology (BIMSB), Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
| | - Markus Landthaler
- Berlin Institute for Medical Systems Biology (BIMSB), Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
- Institut für Biologie, Humboldt Universität zu Berlin, Berlin, Germany
| | - Schayan Yousefian
- Berlin Institute for Medical Systems Biology (BIMSB), Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
| | - Simon Haas
- Berlin Institute for Medical Systems Biology (BIMSB), Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
- Charité - Universitätsmedizin Berlin, Berlin, Germany
- German Cancer Consortium (DKTK), Partner Site Berlin, DKFZ and Charité - Universitätsmedizin Berlin, Berlin, Germany
- Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Florian Kurth
- Department of Infectious Diseases, Respiratory Medicine and Critical Care, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt - Universität zu Berlin, Berlin, Germany
| | - Sarah A Teichmann
- Cellular Genetics Programme, Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, UK
- Cavendish Laboratory, Department of Physics, University of Cambridge, Cambridge, UK
| | - Gavin Y Oudit
- Division of Cardiology, Department of Medicine, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada
- Mazankowski Alberta Heart Institute, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada
| | - Hendrik Milting
- Erich and Hanna Klessmann Institute, Heart and Diabetes Center NRW, University Hospital of the Ruhr-University Bochum, Bad Oeynhausen, Germany
| | - Michela Noseda
- National Heart and Lung Institute, Imperial College London, London, UK
- British Heart Foundation Centre for Research Excellence and Centre for Regenerative Medicine, Imperial College London, London, UK
| | | | - Christine E Seidman
- Department of Genetics, Harvard Medical School, Boston, MA, USA
- Cardiovascular Division, Brigham and Women's Hospital Boston, Boston, MA, USA
- Howard Hughes Medical Institute, Chevy Chase, MD, USA
| | - Bettina Heidecker
- Department of Cardiology, Angiology and Intensive Medicine CBF, Deutsches Herzzentrum der Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Leif E Sander
- Berlin Institute of Health (BIH) at Charité - Universitätsmedizin Berlin, BIH Center for Regenerative Therapies (BCRT), Berlin, Germany
- Department of Infectious Diseases, Respiratory Medicine and Critical Care, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt - Universität zu Berlin, Berlin, Germany
| | - Birgit Sawitzki
- Translational Immunology, Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Karin Klingel
- Cardiopathology, Institute for Pathology and Neuropathology, University Hospital Tübingen, Tübingen, Germany
| | - Patrick Doeblin
- DZHK (German Centre for Cardiovascular Research), partner site Berlin, Berlin, Germany
- Department of Cardiology, Angiology and Intensive Care, Campus Virchow, Deutsches Herzzentrum der Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Sebastian Kelle
- DZHK (German Centre for Cardiovascular Research), partner site Berlin, Berlin, Germany
- Department of Cardiology, Angiology and Intensive Care, Campus Virchow, Deutsches Herzzentrum der Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Sophie Van Linthout
- DZHK (German Centre for Cardiovascular Research), partner site Berlin, Berlin, Germany
- Berlin Institute of Health (BIH) at Charité - Universitätsmedizin Berlin, BIH Center for Regenerative Therapies (BCRT), Berlin, Germany
| | - Norbert Hubner
- Cardiovascular and Metabolic Sciences, Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany.
- DZHK (German Centre for Cardiovascular Research), partner site Berlin, Berlin, Germany.
- Charité - Universitätsmedizin Berlin, Berlin, Germany.
- Helmholtz-Institute for Translational AngioCardioScience (HI-TAC) of the Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC) at Heidelberg University, Heidelberg, Germany.
| | - Carsten Tschöpe
- DZHK (German Centre for Cardiovascular Research), partner site Berlin, Berlin, Germany.
- Berlin Institute of Health (BIH) at Charité - Universitätsmedizin Berlin, BIH Center for Regenerative Therapies (BCRT), Berlin, Germany.
- Department of Cardiology, Angiology and Intensive Care, Campus Virchow, Deutsches Herzzentrum der Charité - Universitätsmedizin Berlin, Berlin, Germany.
| |
Collapse
|
3
|
Zidar DA, McComsey GA, Anthony DD, McDaniel J, Chan TA, Al-Kindi SG. Distinct Cytokine Patterns Identify Acute and Convalescent Myocardial Involvement After Coronavirus Disease 2019: A Multicohort Biomarker Study. J Infect Dis 2025:jiaf045. [PMID: 39969260 DOI: 10.1093/infdis/jiaf045] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2024] [Accepted: 01/17/2025] [Indexed: 02/20/2025] Open
Abstract
We sought to identify the immunobiologic underpinnings of cardiac involvement as a postacute sequela of coronavirus disease 2019 (COVID-19) by comparing acute and convalescent populations. For the latter, an integrated analysis of cytokine levels, cardiac magnetic resonance imaging, and cardiopulmonary exercise capacity was performed. Unlike acute cardiac injury, which was associated with heightened tumor necrosis factor alpha (TNF-α) but not interleukin 18 (IL-18), convalescent myocardial inflammation/edema correlated with IL-18 but not TNF-α. Thus, inflammation is not a monolith in relation to cardiac involvement in the setting of COVID-19. Instead, convalescent cardiac involvement may emerge from mechanisms distinct from acute injury, and appropriately targeted therapies may prevent postacute sequalae of COVID-19.
Collapse
Affiliation(s)
- David A Zidar
- Departments of Pathology and Medicine, Case Western Reserve University School of Medicine
- Department of Medicine, University Hospitals Cleveland Medical Center
- Department of Pathology, Louis Stokes Cleveland Veterans Affairs Medical Center, Cleveland
| | - Grace A McComsey
- Departments of Pathology and Medicine, Case Western Reserve University School of Medicine
- Department of Medicine, University Hospitals Cleveland Medical Center
| | - Donald D Anthony
- Departments of Pathology and Medicine, Case Western Reserve University School of Medicine
- Department of Pathology, Louis Stokes Cleveland Veterans Affairs Medical Center, Cleveland
| | - John McDaniel
- Department of Pathology, Louis Stokes Cleveland Veterans Affairs Medical Center, Cleveland
- School of Health Sciences, Kent State University, Kent
| | - Timothy A Chan
- Global Center for Immunotherapy and Immuno-Oncology, Cleveland Clinic Lerner College of Medicine, Cleveland, Ohio
| | - Sadeer G Al-Kindi
- Departments of Pathology and Medicine, Case Western Reserve University School of Medicine
- Department of Medicine, University Hospitals Cleveland Medical Center
| |
Collapse
|
4
|
Wu Y, He L, Li R, Li J, Zhao Q, Shao B. A20 as a Potential Therapeutic Target for COVID-19. Immun Inflamm Dis 2025; 13:e70127. [PMID: 39853876 PMCID: PMC11760982 DOI: 10.1002/iid3.70127] [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: 04/12/2024] [Revised: 11/29/2024] [Accepted: 01/03/2025] [Indexed: 01/26/2025] Open
Abstract
BACKGROUND Coronavirus disease 2019 (COVID-19), caused by the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), is a major concern due to its astonishing prevalence and high fatality rate, especially among elderly people. Patients suffering from COVID-19 may exhibit immunosuppression in the initial stage of infection, while a cytokine storm can occur when the disease progresses to a severe stage. This inopportune immune rhythm not only makes patients more susceptible to the virus but also leads to numerous complications resulting from the excessive production of inflammatory factors. A20, which is widely accepted as a pivotal regulator of inflammation, has been shown to be implicated in the processes of antiviral responses and immunosuppression. Thus, A20 may participate in regulating the pathological processes of COVID-19. METHODS This narrative literature review summarizes recent evidence on the mechanisms of A20 in regulating the pathological processes of COVID-19. We also downloaded single-cell RNA-seq data sets from healthy individuals and patients with varying severities of COVID-19 from the NCBI GEO database to further dissect A20's regulatory mechanisms of these intricate cytokine pathways that are closely associated with SARS-CoV-2 infection. RESULTS A20 might be one of the most critical anti-infectious and anti-inflammatory factors involved in the pathogenesis of COVID-19. It effectively suppresses the immune damage and inflammatory storm caused by viral infection. CONCLUSIONS Understanding the relationship between A20-regulated signaling pathways and pathological processes of COVID-19 can provide insight into potential targets for intervention. Precise regulation of A20 to induce antiviral activity and an anti-inflammatory response could mediate the pathogenesis of COVID-19 and could become an effective treatment.
Collapse
Affiliation(s)
- Yongyao Wu
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, West China Hospital of StomatologySichuan UniversityChengduChina
| | - Lilan He
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, West China Hospital of StomatologySichuan UniversityChengduChina
| | - Rong Li
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, West China Hospital of StomatologySichuan UniversityChengduChina
| | - Jiuxuan Li
- Laboratory of Radiation Biology, Laboratory Medicine Centre, Department of Blood TransfusionThe Second Affiliated HospitalArmy Military Medical UniversityChongqingChina
| | - Qing Zhao
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, West China Hospital of StomatologySichuan UniversityChengduChina
| | - Bin Shao
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, West China Hospital of StomatologySichuan UniversityChengduChina
| |
Collapse
|
5
|
Hashimoto K, Yamamoto H, Ikeda Y, Isogai J, Hashimoto T. A case of biopsy-proven inflammatory dilated cardiomyopathy following heterologous mRNA-1273 third-dose immunization. ESC Heart Fail 2024; 11:4442-4449. [PMID: 38946583 PMCID: PMC11631250 DOI: 10.1002/ehf2.14924] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2024] [Revised: 05/10/2024] [Accepted: 06/15/2024] [Indexed: 07/02/2024] Open
Affiliation(s)
- Katsuya Hashimoto
- Department of Cardiovascular MedicineNarita‐Tomisato Tokushukai HospitalChibaJapan
| | - Hiroyuki Yamamoto
- Department of Cardiovascular MedicineNarita‐Tomisato Tokushukai HospitalChibaJapan
- Department of CardiologyTokyo Medical University HospitalTokyoJapan
| | - Yoshihiko Ikeda
- Department of PathologyNational Cerebral and Cardiovascular CenterSuitaJapan
| | - Jun Isogai
- Department of RadiologyAsahi General HospitalAsahiJapan
| | - Toru Hashimoto
- Department of Cardiovascular MedicineNarita‐Tomisato Tokushukai HospitalChibaJapan
| |
Collapse
|
6
|
Bernardes BG, Moura AD, Guarnieri JPDO, Silva CFMD, Costa HHMD, Silva IGMD, Cordeiro KBB, Báo SN, Prudêncio CR, Lancellotti M. Vaccination with outer membrane vesicles from Neisseria Meningitidis and SBa15, SBa16 mesoporous silica associated with SARS-CoV-2 induces protective humoral and cellular response against COVID-19 in mice. Braz J Infect Dis 2024; 28:104479. [PMID: 39547005 DOI: 10.1016/j.bjid.2024.104479] [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: 07/11/2024] [Revised: 10/13/2024] [Accepted: 11/04/2024] [Indexed: 11/17/2024] Open
Abstract
The global impact of the SARS-CoV-2 (severe acute respiratory syndrome coronavirus 2) pandemic in 2019-2020 has led to significant changes in worldwide vaccination and immune prophylactic approaches. In this study, our research delves into a new immunization strategy that does not involve the use of additional adjuvants or preservatives, focusing on the effects of virus fusion with a bacterial nanostructure. The experimental procedures outlined in this paper involved the cultivation of SARS-CoV-2, the production, extraction, and nanocharacterization of outer membrane vesicles (OMV) from Neisseria meningitidis, immunization of mice with two doses of OMV combined with SARS-CoV-2, and the use of mesoporous silica SBa15 and SBa16 adsorbed to the same virus. The immune response was assessed through an indirect elisa method, analysis of cytokine expression profiles, and seroneutralization of the SARS-CoV-2 strain. The characterizations of associated OMV - SARS-CoV-2 and adsorption SBa15 and SBa16 were performed using Nanosight Tracking Analysis (NTA), which showed a high density of particles in the formulation. mice were then immunized, resulting in an immune response that produced high levels of neutralizing antibodies in IgG and IgG1 mouse immunoglobulins. In addition, expressions of IL-2, IL-4, and IL-23 in spleen cells were reinforced after the vaccination process. The comparative study of these three vaccine formulations has shown that the development of new vaccines for SARS-CoV-2 should take into consideration the production of neutralizing antibodies and the maintenance of immunological memory.
Collapse
Affiliation(s)
- Bruno Gaia Bernardes
- Universidade de Campinas (UNICAMP), Faculdade de Ciências Farmacêuticas (FCF), Laboratório de Biotecnologia (LABIOTEC), Campinas, SP, Brazil
| | - Andrew Douglas Moura
- Universidade de Campinas (UNICAMP), Faculdade de Ciências Farmacêuticas (FCF), Laboratório de Biotecnologia (LABIOTEC), Campinas, SP, Brazil; Instituto Adolfo Lutz, Centro de Imunologia, São Paulo, SP, Brazil
| | - João Paulo de Oliveira Guarnieri
- Universidade de Campinas (UNICAMP), Faculdade de Ciências Farmacêuticas (FCF), Laboratório de Biotecnologia (LABIOTEC), Campinas, SP, Brazil
| | - Carlos Fernando Macedo da Silva
- Universidade de Campinas (UNICAMP), Faculdade de Ciências Farmacêuticas (FCF), Laboratório de Biotecnologia (LABIOTEC), Campinas, SP, Brazil
| | | | - Ingrid Gracielle Martins da Silva
- Universidade de Brasília, Campus Universitário Darcy Ribeiro, Instituto de Ciências Biológicas, Departamento de Biologia Celular, Laboratório de Microscopia e Microanálise (LMM), Brasília, DF, Brazil
| | - Karine Brenda Barros Cordeiro
- Universidade de Brasília, Campus Universitário Darcy Ribeiro, Instituto de Ciências Biológicas, Departamento de Biologia Celular, Laboratório de Microscopia e Microanálise (LMM), Brasília, DF, Brazil
| | - Sônia Nair Báo
- Universidade de Brasília, Campus Universitário Darcy Ribeiro, Instituto de Ciências Biológicas, Departamento de Biologia Celular, Laboratório de Microscopia e Microanálise (LMM), Brasília, DF, Brazil
| | | | - Marcelo Lancellotti
- Universidade de Campinas (UNICAMP), Faculdade de Ciências Farmacêuticas (FCF), Laboratório de Biotecnologia (LABIOTEC), Campinas, SP, Brazil.
| |
Collapse
|
7
|
Kyriakopoulos AM, Nigh G, McCullough PA, Seneff S. Clinical rationale for dietary lutein supplementation in long COVID and mRNA vaccine injury syndromes. F1000Res 2024; 13:191. [PMID: 39526116 PMCID: PMC11549548 DOI: 10.12688/f1000research.143517.3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 10/14/2024] [Indexed: 11/16/2024] Open
Abstract
Lutein, a plant-derived xanthophyl-carotenoid, is an exceptional antioxidant and anti-inflammatory constituent found in food. High dietary intake of lutein is beneficial against eye disease, improves cardiometabolic health, protects from neurodegenerative diseases, and is beneficial for liver, kidney, and respiratory health. Lutein protects against oxidative and nitrosative stress, both of which play a major role in long COVID and mRNA vaccination injury syndromes. Lutein is an important natural agent for therapeutic use against oxidative and nitrosative stress in chronic illnesses such as cardiovascular and neurodegenerative diseases and cancer. It can also potentially inhibit spike protein-induced inflammation. Rich dietary supplementation of lutein, naturally derived in non-biodegradable Extra Virgin Olive Oil (EVOO), can most optimally be used against oxidative and nitrosative stress during post-COVID and mRNA vaccination injury syndromes. Due to its high oleic acid (OA) content, EVOO supports optimal absorption of dietary lutein. The main molecular pathways by which the SARS-CoV-2 spike protein induces pathology, nuclear factor kappa-light-chain-enhancer activated B cells (NF-κB) and activated protein (AP)-1, can be suppressed by lutein. Synergy with other natural compounds for spike protein detoxification is likely.
Collapse
Affiliation(s)
| | - Greg Nigh
- Naturopathic Oncologist, Immersion Health, Portland, Oregon, USA
| | | | - Stephanie Seneff
- Computer Science and Artificial Intelligence Laboratory, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
| |
Collapse
|
8
|
du Preez HN, Lin J, Maguire GEM, Aldous C, Kruger HG. COVID-19 vaccine adverse events: Evaluating the pathophysiology with an emphasis on sulfur metabolism and endotheliopathy. Eur J Clin Invest 2024; 54:e14296. [PMID: 39118373 DOI: 10.1111/eci.14296] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/02/2024] [Accepted: 07/24/2024] [Indexed: 08/10/2024]
Abstract
In this narrative review, we assess the pathophysiology of severe adverse events that presented after vaccination with DNA and mRNA vaccines against COVID-19. The focus is on the perspective of an undersulfated and degraded glycocalyx, considering its impact on immunomodulation, inflammatory responses, coagulation and oxidative stress. The paper explores various factors that lead to glutathione and inorganic sulfate depletion and their subsequent effect on glycocalyx sulfation and other metabolites, including hormones. Components of COVID-19 vaccines, such as DNA and mRNA material, spike protein antigen and lipid nanoparticles, are involved in possible cytotoxic effects. The common thread connecting these adverse events is endotheliopathy or glycocalyx degradation, caused by depleted glutathione and inorganic sulfate levels, shear stress from circulating nanoparticles, aggregation and formation of protein coronas; leading to imbalanced immune responses and chronic release of pro-inflammatory cytokines, ultimately resulting in oxidative stress and systemic inflammatory response syndrome. By understanding the underlying pathophysiology of severe adverse events, better treatment options can be explored.
Collapse
Affiliation(s)
- Heidi N du Preez
- Catalysis and Peptide Research Unit, University of KwaZulu-Natal, Durban, South Africa
- College of Health Sciences, University of KwaZulu-Natal, Durban, South Africa
| | - Johnson Lin
- School of Life Sciences, University of KwaZulu-Natal, Durban, South Africa
| | - Glenn E M Maguire
- Catalysis and Peptide Research Unit, University of KwaZulu-Natal, Durban, South Africa
- School of Chemistry and Physics, University of KwaZulu-Natal, Durban, South Africa
| | - Colleen Aldous
- College of Health Sciences, University of KwaZulu-Natal, Durban, South Africa
| | - Hendrik G Kruger
- Catalysis and Peptide Research Unit, University of KwaZulu-Natal, Durban, South Africa
| |
Collapse
|
9
|
Buoninfante A, Andeweg A, Genov G, Cavaleri M. Myocarditis associated with COVID-19 vaccination. NPJ Vaccines 2024; 9:122. [PMID: 38942751 PMCID: PMC11213864 DOI: 10.1038/s41541-024-00893-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Accepted: 05/23/2024] [Indexed: 06/30/2024] Open
Abstract
Following the start of the COVID-19 vaccination campaign, the adverse events of myocarditis and pericarditis were linked mainly to mRNA COVID-19 vaccines by the regulatory authorities worldwide. COVID-19 vaccines have been administered to several million people and the risk of myocarditis post COVID-19 vaccination has been characterised in great detail. At the present time the research data available are scarce and there is still no clear understanding of the biological mechanism/s responsible for this disease. This manuscript provides a concise overview of the epidemiology of myocarditis and the most prominent mechanistic insights in the pathophysiology of the disease. Most importantly it underscores the needed next steps in the research agenda required to characterize the pathophysiology of this disease post-COVID-19 vaccination. Finally, it shares our perspectives and considerations for public health.
Collapse
Affiliation(s)
| | - Arno Andeweg
- Public Health Threats, European Medicines Agency, Amsterdam, The Netherlands
| | - Georgy Genov
- Pharmacovigilance Office, European Medicines Agency, Amsterdam, The Netherlands
| | - Marco Cavaleri
- Public Health Threats, European Medicines Agency, Amsterdam, The Netherlands.
| |
Collapse
|
10
|
Tsang HW, Kwan MYW, Chua GT, Tsao SSL, Wong JSC, Tung KTS, Chan GCF, To KKW, Wong ICK, Leung WH, Ip P. The central role of natural killer cells in mediating acute myocarditis after mRNA COVID-19 vaccination. MED 2024; 5:335-347.e3. [PMID: 38521068 DOI: 10.1016/j.medj.2024.02.008] [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: 11/03/2023] [Revised: 01/26/2024] [Accepted: 02/26/2024] [Indexed: 03/25/2024]
Abstract
BACKGROUND Vaccine-related acute myocarditis is recognized as a rare and specific vaccine complication following mRNA-based COVID-19 vaccinations. The precise mechanisms remain unclear. We hypothesized that natural killer (NK) cells play a central role in its pathogenesis. METHODS Samples from 60 adolescents with vaccine-related myocarditis were analyzed, including pro-inflammatory cytokines, cardiac troponin T, genotyping, and immunophenotyping of the corresponding activation subsets of NK cells, monocytes, and T cells. Results were compared with samples from 10 vaccinated individuals without myocarditis and 10 healthy controls. FINDINGS Phenotypically, high levels of serum cytokines pivotal for NK cells, including interleukin-1β (IL-1β), interferon α2 (IFN-α2), IL-12, and IFN-γ, were observed in post-vaccination patients with myocarditis, who also had high percentage of CD57+ NK cells in blood, which in turn correlated positively with elevated levels of cardiac troponin T. Abundance of the CD57+ NK subset was particularly prominent in males and in those after the second dose of vaccination. Genotypically, killer cell immunoglobulin-like receptor (KIR) KIR2DL5B(-)/KIR2DS3(+)/KIR2DS5(-)/KIR2DS4del(+) was a risk haplotype, in addition to single-nucleotide polymorphisms related to the NK cell-specific expression quantitative trait loci DNAM-1 and FuT11, which also correlated with cardiac troponin T levels in post-vaccination patients with myocarditis. CONCLUSION Collectively, these data suggest that NK cell activation by mRNA COVID-19 vaccine contributed to the pathogenesis of acute myocarditis in genetically and epidemiologically vulnerable subjects. FUNDING This work was funded by the Hong Kong Collaborative Research Fund (CRF) 2020/21 and the CRF Coronavirus and Novel Infectious Diseases Research Exercises (reference no. C7149-20G).
Collapse
Affiliation(s)
- Hing Wai Tsang
- Department of Paediatrics and Adolescent Medicine, Li Ka Shing Faculty of Medicine, School of Clinical Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Mike Yat Wah Kwan
- Department of Paediatrics and Adolescent Medicine, Li Ka Shing Faculty of Medicine, School of Clinical Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Gilbert T Chua
- Department of Paediatrics and Adolescent Medicine, Li Ka Shing Faculty of Medicine, School of Clinical Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Sabrina Siu Ling Tsao
- Department of Paediatrics and Adolescent Medicine, Li Ka Shing Faculty of Medicine, School of Clinical Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Joshua Sung Chih Wong
- Department of Paediatrics and Adolescent Medicine, Princess Margaret Hospital Authority, Hong Kong SAR, China
| | - Keith Tsz Suen Tung
- Department of Paediatrics and Adolescent Medicine, Li Ka Shing Faculty of Medicine, School of Clinical Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Godfrey Chi Fung Chan
- Paediatric Haematology & Oncology Centre, Hong Kong Sanatorium & Hospital, Hong Kong SAR, China
| | - Kelvin Kai Wang To
- Department of Microbiology, Li Ka Shing Faculty of Medicine, School of Clinical Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Ian Chi Kei Wong
- Department of Pharmacology and Pharmacy, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China; School of Pharmacy, Medical Sciences Division, Macau University of Science and Technology, Macau SAR, China; School of Pharmacy, Aston University, Birmingham B4 7ET, England
| | - Wing Hang Leung
- Department of Paediatrics and Adolescent Medicine, Li Ka Shing Faculty of Medicine, School of Clinical Medicine, The University of Hong Kong, Hong Kong SAR, China.
| | - Patrick Ip
- Department of Paediatrics and Adolescent Medicine, Li Ka Shing Faculty of Medicine, School of Clinical Medicine, The University of Hong Kong, Hong Kong SAR, China.
| |
Collapse
|
11
|
Muslimov A, Tereshchenko V, Shevyrev D, Rogova A, Lepik K, Reshetnikov V, Ivanov R. The Dual Role of the Innate Immune System in the Effectiveness of mRNA Therapeutics. Int J Mol Sci 2023; 24:14820. [PMID: 37834268 PMCID: PMC10573212 DOI: 10.3390/ijms241914820] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2023] [Revised: 09/24/2023] [Accepted: 09/28/2023] [Indexed: 10/15/2023] Open
Abstract
Advances in molecular biology have revolutionized the use of messenger RNA (mRNA) as a therapeutic. The concept of nucleic acid therapy with mRNA originated in 1990 when Wolff et al. reported successful expression of proteins in target organs by direct injection of either plasmid DNA or mRNA. It took decades to bring the transfection efficiency of mRNA closer to that of DNA. The next few decades were dedicated to turning in vitro-transcribed (IVT) mRNA from a promising delivery tool for gene therapy into a full-blown therapeutic modality, which changed the biotech market rapidly. Hundreds of clinical trials are currently underway using mRNA for prophylaxis and therapy of infectious diseases and cancers, in regenerative medicine, and genome editing. The potential of IVT mRNA to induce an innate immune response favors its use for vaccination and immunotherapy. Nonetheless, in non-immunotherapy applications, the intrinsic immunostimulatory activity of mRNA directly hinders the desired therapeutic effect since it can seriously impair the target protein expression. Targeting the same innate immune factors can increase the effectiveness of mRNA therapeutics for some indications and decrease it for others, and vice versa. The review aims to present the innate immunity-related 'barriers' or 'springboards' that may affect the development of immunotherapies and non-immunotherapy applications of mRNA medicines.
Collapse
Affiliation(s)
- Albert Muslimov
- Scientific Center for Translational Medicine, Sirius University of Science and Technology, Olympic Ave 1, 354340 Sirius, Russia; (V.T.); (D.S.); (V.R.); (R.I.)
- Laboratory of Nano- and Microencapsulation of Biologically Active Substances, Peter the Great St. Petersburg Polytechnic University, Polytechnicheskaya 29, 195251 St. Petersburg, Russia;
- RM Gorbacheva Research Institute, Pavlov University, L’va Tolstogo 6-8, 197022 St. Petersburg, Russia;
| | - Valeriy Tereshchenko
- Scientific Center for Translational Medicine, Sirius University of Science and Technology, Olympic Ave 1, 354340 Sirius, Russia; (V.T.); (D.S.); (V.R.); (R.I.)
| | - Daniil Shevyrev
- Scientific Center for Translational Medicine, Sirius University of Science and Technology, Olympic Ave 1, 354340 Sirius, Russia; (V.T.); (D.S.); (V.R.); (R.I.)
| | - Anna Rogova
- Laboratory of Nano- and Microencapsulation of Biologically Active Substances, Peter the Great St. Petersburg Polytechnic University, Polytechnicheskaya 29, 195251 St. Petersburg, Russia;
- Saint-Petersburg Chemical-Pharmaceutical University, Professora Popova 14, 197376 St. Petersburg, Russia
- School of Physics and Engineering, ITMO University, Lomonosova 9, 191002 St. Petersburg, Russia
| | - Kirill Lepik
- RM Gorbacheva Research Institute, Pavlov University, L’va Tolstogo 6-8, 197022 St. Petersburg, Russia;
| | - Vasiliy Reshetnikov
- Scientific Center for Translational Medicine, Sirius University of Science and Technology, Olympic Ave 1, 354340 Sirius, Russia; (V.T.); (D.S.); (V.R.); (R.I.)
- Institute of Cytology and Genetics, Siberian Branch of Russian Academy of Sciences, Prospekt Akad. Lavrentyeva 10, 630090 Novosibirsk, Russia
| | - Roman Ivanov
- Scientific Center for Translational Medicine, Sirius University of Science and Technology, Olympic Ave 1, 354340 Sirius, Russia; (V.T.); (D.S.); (V.R.); (R.I.)
| |
Collapse
|
12
|
Pari B, Babbili A, Kattubadi A, Thakre A, Thotamgari S, Gopinathannair R, Olshansky B, Dominic P. COVID-19 Vaccination and Cardiac Arrhythmias: A Review. Curr Cardiol Rep 2023; 25:925-940. [PMID: 37530946 DOI: 10.1007/s11886-023-01921-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 07/03/2023] [Indexed: 08/03/2023]
Abstract
PURPOSE OF REVIEW In this review, we aim to delve into the existing literature, seeking to uncover the mechanisms, investigate the electrocardiographic changes, and examine the treatment methods of various cardiac arrhythmias that occur after administration of the COVID-19 vaccine. RECENT FINDINGS A global survey has exposed an incidence of arrhythmia in 18.27% of hospitalized COVID-19 patients. Furthermore, any type of COVID-19 vaccine - be it mRNA, adenovirus vector, whole inactivated, or protein subunit - appears to instigate cardiac arrhythmias. Among the cardiac adverse events reported post-COVID-19 vaccination, myocarditis emerges as the most common and is thought to be a potential cause of bradyarrhythmia. When a patient post-COVID-19 vaccination presents a suspicion of cardiac involvement, clinicians should perform a comprehensive history and physical examination, measure electrolyte levels, conduct ECG, and carry out necessary imaging studies. In our extensive literature search, we uncovered various potential mechanisms that might lead to cardiac conduction abnormalities and autonomic dysfunction in patients who have received the COVID-19 vaccine. These mechanisms encompass direct viral invasion through molecular mimicry/spike (S) protein production, an escalated inflammatory response, hypoxia, myocardial cell death, and the eventual scar/fibrosis. They correspond to a range of conditions including atrial tachyarrhythmias, bradyarrhythmia, ventricular arrhythmias, sudden cardiac death, and the frequently occurring myocarditis. For treating these COVID-19 vaccination-induced arrhythmias, we should incorporate general treatment strategies, similar to those applied to arrhythmias from other causes.
Collapse
Affiliation(s)
- Bavithra Pari
- Department of Medicine, LSUHSC-S, Shreveport, LA, USA
| | | | | | - Anuj Thakre
- Department of Medicine, LSUHSC-S, Shreveport, LA, USA
| | | | - Rakesh Gopinathannair
- The Kansas City Heart Rhythm Institute (KCHRI) & Research Foundation, Overland Park Regional Medical Center, KS, Kansas City, USA
| | - Brian Olshansky
- Division of Cardiology, Department of Medicine, The University of Iowa, Carver College of Medicine, Iowa City, IA, USA
| | - Paari Dominic
- Division of Cardiology, Department of Medicine, The University of Iowa, Carver College of Medicine, Iowa City, IA, USA.
| |
Collapse
|
13
|
Palmowski J, Kohnhorst S, Bauer P, Puta C, Haunhorst S, Gebhardt K, Reichel T, Keller C, Huber M, Raifer H, Krüger K. T-cell-derived TNF-α and a cluster of immunological parameters from plasma allow a separation between SARS-CoV-2 convalescent versus vaccinated elite athletes. Front Physiol 2023; 14:1203983. [PMID: 37427401 PMCID: PMC10324374 DOI: 10.3389/fphys.2023.1203983] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2023] [Accepted: 06/09/2023] [Indexed: 07/11/2023] Open
Abstract
Guidelines for medical clearing after SARS-CoV-2 infection in elite athletes do not include T-cell immunity aspects despite its relevance in the course of COVID-19 disease. Therefore, we aimed to analyze T-cell-related cytokines before and after in-vitro activation of CD4+ T-cells. We sampled professional indoor sports athletes at medical clearing after SARS-CoV-2 infection obtaining clinical, fitness data, and serological data including CD4+ T-cell cytokines. All data were analyzed by principal component analysis and 2 × 2 repeated measures ANOVA. CD4+ T-cells were sampled for cell culture activation with anti-CD3/anti-CD28 tetramers. At medical clearing, CD4+ T-cells from convalescent athletes secreted increased levels of TNF-α 72 h after in-vitro activation compared to vaccinated athletes. IL-18 levels in plasma were elevated and a cluster of parameters differentiated convalescent from vaccinated athletes by 13 parameters at the timepoint of medical clearing. All clinical data indicate infection is resolved, while increased TNF-α may reflect altered proportions of peripheral T-cells as a hangover of infection.
Collapse
Affiliation(s)
- Jana Palmowski
- Department of Exercise Physiology and Sports Therapy, Institute of Sports Science, Justus Liebig University Giessen, Giessen, Germany
| | - Sarah Kohnhorst
- Center for Tumor and Immunology, Institute for Systems Immunology, Marburg, Germany
| | - Pascal Bauer
- Department of Cardiology and Angiology, Justus-Liebig-University Giessen, Giessen, Germany
| | - Christian Puta
- Department of Sports Medicine and Health Promotion, Friedrich Schiller University Jena, Jena, Germany
- Center for Sepsis Control and Care (CSCC), Jena University Hospital, Friedrich-Schiller-University Jena, Jena, Germany
- Center for Interdisciplinary Prevention of Diseases Related to Professional Activities, Jena, Germany
| | - Simon Haunhorst
- Department of Sports Medicine and Health Promotion, Friedrich Schiller University Jena, Jena, Germany
| | - Kristina Gebhardt
- Department of Exercise Physiology and Sports Therapy, Institute of Sports Science, Justus Liebig University Giessen, Giessen, Germany
| | - Thomas Reichel
- Department of Exercise Physiology and Sports Therapy, Institute of Sports Science, Justus Liebig University Giessen, Giessen, Germany
| | - Christian Keller
- Institute of Virology, Philipps University Marburg, Marburg, Germany
| | - Magdalena Huber
- Center for Tumor and Immunology, Institute for Systems Immunology, Marburg, Germany
| | - Hartmann Raifer
- Center for Tumor and Immunology, Institute for Systems Immunology, Marburg, Germany
| | - Karsten Krüger
- Department of Exercise Physiology and Sports Therapy, Institute of Sports Science, Justus Liebig University Giessen, Giessen, Germany
| |
Collapse
|
14
|
Carleton BC, Salmon DA, Ip P, Wong IC, Lai FT. Benefits v. risks of COVID-19 vaccination: an examination of vaccination policy impact on the occurrence of myocarditis and pericarditis. THE LANCET REGIONAL HEALTH. WESTERN PACIFIC 2023; 37:100797. [PMID: 37360870 PMCID: PMC10196680 DOI: 10.1016/j.lanwpc.2023.100797] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/26/2022] [Revised: 02/13/2023] [Accepted: 05/07/2023] [Indexed: 06/28/2023]
Abstract
Studies of myocarditis/pericarditis following mRNA COVID-19 vaccines in Hong Kong have been published. Data are consistent with data from other active surveillance or healthcare databases. The mRNA COVID-19 vaccines have been shown to rarely increase risk of myocarditis, with the highest risk among males aged 12-17 after the second dose. An increased risk of pericarditis has also been shown after the second dose, though less common than myocarditis and more evenly distributed among different sex and age groups. Because of the increased risk of post-vaccine myocarditis, Hong Kong implemented a single dose mRNA COVID-19 vaccine policy on September 15, 2021 for adolescents (age 12-17 years). Post-policy, there were no cases of carditis. 40,167 first dose patients did not receive a second dose. This policy was highly successful in the reduction of carditis, but the trade-off is the potential risk of disease and cost to population-level immunity. This commentary brings forward some important global policy considerations.
Collapse
Affiliation(s)
- Bruce C. Carleton
- Division of Translational Therapeutics, Department of Paediatrics, Faculty of Medicine, University of British Columbia, Vancouver, Canada
- Pharmaceutical Outcomes Programme, BC Children's Hospital, Vancouver, Canada
- BC Children's Hospital Research Institute, Vancouver, Canada
| | - Daniel A. Salmon
- Institute for Vaccine Safety, Johns Hopkins University Bloomberg School of Public Health, 615 N. Wolfe Street, Baltimore, MD 21205, United States
- Department of International Health, Johns Hopkins University Bloomberg School of Public Health, 615 N. Wolfe Street, Baltimore, MD 21205, United States
- Department of Health, Behavior and Society, Johns Hopkins University Bloomberg School of Public Health, 615 N. Wolfe Street, Baltimore, MD 21205, United States
| | - Patrick Ip
- Department of Paediatrics and Adolescent Medicine, School of Clinical Medicine, The University of Hong Kong, Hong Kong SAR, China
- Department of Paediatrics and Adolescent Medicine, The Hong Kong Children's Hospital, Hong Kong SAR, China
- Department of Paediatrics and Adolescent Medicine, Queen Mary Hospital, Hong Kong SAR, China
| | - Ian C.K. Wong
- The University of Hong Kong, Hong Kong SAR, China
- Aston School of Pharmacy, Aston University, Birmingham, United Kingdom
| | - Francicso T.T. Lai
- Centre for Safe Medication Practice and Research, Department of Pharmacology and Pharmacy, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| |
Collapse
|
15
|
Barmada A, Klein J, Ramaswamy A, Brodsky NN, Jaycox JR, Sheikha H, Jones KM, Habet V, Campbell M, Sumida TS, Kontorovich A, Bogunovic D, Oliveira CR, Steele J, Hall EK, Pena-Hernandez M, Monteiro V, Lucas C, Ring AM, Omer SB, Iwasaki A, Yildirim I, Lucas CL. Cytokinopathy with aberrant cytotoxic lymphocytes and profibrotic myeloid response in SARS-CoV-2 mRNA vaccine-associated myocarditis. Sci Immunol 2023; 8:eadh3455. [PMID: 37146127 PMCID: PMC10468758 DOI: 10.1126/sciimmunol.adh3455] [Citation(s) in RCA: 35] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2023] [Accepted: 04/19/2023] [Indexed: 05/07/2023]
Abstract
Rare immune-mediated cardiac tissue inflammation can occur after vaccination, including after SARS-CoV-2 mRNA vaccines. However, the underlying immune cellular and molecular mechanisms driving this pathology remain poorly understood. Here, we investigated a cohort of patients who developed myocarditis and/or pericarditis with elevated troponin, B-type natriuretic peptide, and C-reactive protein levels as well as cardiac imaging abnormalities shortly after SARS-CoV-2 mRNA vaccination. Contrary to early hypotheses, patients did not demonstrate features of hypersensitivity myocarditis, nor did they have exaggerated SARS-CoV-2-specific or neutralizing antibody responses consistent with a hyperimmune humoral mechanism. We additionally found no evidence of cardiac-targeted autoantibodies. Instead, unbiased systematic immune serum profiling revealed elevations in circulating interleukins (IL-1β, IL-1RA, and IL-15), chemokines (CCL4, CXCL1, and CXCL10), and matrix metalloproteases (MMP1, MMP8, MMP9, and TIMP1). Subsequent deep immune profiling using single-cell RNA and repertoire sequencing of peripheral blood mononuclear cells during acute disease revealed expansion of activated CXCR3+ cytotoxic T cells and NK cells, both phenotypically resembling cytokine-driven killer cells. In addition, patients displayed signatures of inflammatory and profibrotic CCR2+ CD163+ monocytes, coupled with elevated serum-soluble CD163, that may be linked to the late gadolinium enhancement on cardiac MRI, which can persist for months after vaccination. Together, our results demonstrate up-regulation in inflammatory cytokines and corresponding lymphocytes with tissue-damaging capabilities, suggesting a cytokine-dependent pathology, which may further be accompanied by myeloid cell-associated cardiac fibrosis. These findings likely rule out some previously proposed mechanisms of mRNA vaccine--associated myopericarditis and point to new ones with relevance to vaccine development and clinical care.
Collapse
Affiliation(s)
- Anis Barmada
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT, USA
| | - Jon Klein
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT, USA
| | - Anjali Ramaswamy
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT, USA
| | - Nina N. Brodsky
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT, USA
- Department of Pediatrics, Yale University School of Medicine, New Haven, CT, USA
| | - Jillian R. Jaycox
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT, USA
| | - Hassan Sheikha
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT, USA
- Department of Pediatrics, Yale University School of Medicine, New Haven, CT, USA
| | - Kate M. Jones
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT, USA
| | - Victoria Habet
- Department of Pediatrics, Yale University School of Medicine, New Haven, CT, USA
| | - Melissa Campbell
- Department of Pediatrics, Yale University School of Medicine, New Haven, CT, USA
| | - Tomokazu S. Sumida
- Department of Neurology, Yale University School of Medicine, New Haven, CT, USA
| | - Amy Kontorovich
- The Zena and Michael A. Wiener Cardiovascular Institute; Mindich Child Health and Development Institute; Institute for Genomic Health, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Dusan Bogunovic
- The Zena and Michael A. Wiener Cardiovascular Institute; Mindich Child Health and Development Institute; Institute for Genomic Health, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Center for Inborn Errors of Immunity; Precision Immunology Institute; Mindich Child Health and Development Institute; Department of Pediatrics; Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Carlos R. Oliveira
- Department of Pediatrics, Yale University School of Medicine, New Haven, CT, USA
| | - Jeremy Steele
- Department of Pediatrics, Yale University School of Medicine, New Haven, CT, USA
| | - E. Kevin Hall
- Department of Pediatrics, Yale University School of Medicine, New Haven, CT, USA
| | - Mario Pena-Hernandez
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT, USA
| | - Valter Monteiro
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT, USA
| | - Carolina Lucas
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT, USA
- Yale Center for Infection and Immunity, Yale University, New Haven, CT, USA
| | - Aaron M. Ring
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT, USA
| | - Saad B. Omer
- Department of Medicine, Yale University School of Medicine, New Haven, CT, USA
- Epidemiology of Microbial Diseases, Yale School of Public Health, New Haven, CT, USA
- Yale Institute for Global Health, Yale University, New Haven, CT, USA
| | - Akiko Iwasaki
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT, USA
- Howard Hughes Medical Institute, Chevy Chase, MD, USA
- Yale Center for Infection and Immunity, Yale University, New Haven, CT, USA
| | - Inci Yildirim
- Department of Pediatrics, Yale University School of Medicine, New Haven, CT, USA
- Epidemiology of Microbial Diseases, Yale School of Public Health, New Haven, CT, USA
- Yale Institute for Global Health, Yale University, New Haven, CT, USA
- Yale Center for Infection and Immunity, Yale University, New Haven, CT, USA
| | - Carrie L. Lucas
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT, USA
| |
Collapse
|
16
|
Giannotta G, Murrone A, Giannotta N. COVID-19 mRNA Vaccines: The Molecular Basis of Some Adverse Events. Vaccines (Basel) 2023; 11:747. [PMID: 37112659 PMCID: PMC10145134 DOI: 10.3390/vaccines11040747] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2023] [Revised: 03/21/2023] [Accepted: 03/22/2023] [Indexed: 03/30/2023] Open
Abstract
Each injection of any known vaccine results in a strong expression of pro-inflammatory cytokines. This is the result of the innate immune system activation, without which no adaptive response to the injection of vaccines is possible. Unfortunately, the degree of inflammation produced by COVID-19 mRNA vaccines is variable, probably depending on genetic background and previous immune experiences, which through epigenetic modifications could have made the innate immune system of each individual tolerant or reactive to subsequent immune stimulations.We hypothesize that we can move from a limited pro-inflammatory condition to conditions of increasing expression of pro-inflammatory cytokines that can culminate in multisystem hyperinflammatory syndromes following COVID-19 mRNA vaccines (MIS-V). We have graphically represented this idea in a hypothetical inflammatory pyramid (IP) and we have correlated the time factor to the degree of inflammation produced after the injection of vaccines. Furthermore, we have placed the clinical manifestations within this hypothetical IP, correlating them to the degree of inflammation produced. Surprisingly, excluding the possible presence of an early MIS-V, the time factor and the complexity of clinical manifestations are correlated to the increasing degree of inflammation: symptoms, heart disease and syndromes (MIS-V).
Collapse
Affiliation(s)
| | - Antonio Murrone
- Oncologia Territoriale, Hospice Cure Palliative ASUFC, 33030 Udine, Italy;
| | - Nicola Giannotta
- Medical and Surgery Sciences, Faculty of Medicine, Magna Græcia University, 88100 Catanzaro, Italy;
| |
Collapse
|
17
|
Jaycox JR, Lucas C, Yildirim I, Dai Y, Wang EY, Monteiro V, Lord S, Carlin J, Kita M, Buckner JH, Ma S, Campbell M, Ko A, Omer S, Lucas CL, Speake C, Iwasaki A, Ring AM. SARS-CoV-2 mRNA vaccines decouple anti-viral immunity from humoral autoimmunity. Nat Commun 2023; 14:1299. [PMID: 36894554 PMCID: PMC9996559 DOI: 10.1038/s41467-023-36686-8] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Accepted: 02/09/2023] [Indexed: 03/11/2023] Open
Abstract
mRNA-based vaccines dramatically reduce the occurrence and severity of COVID-19, but are associated with rare vaccine-related adverse effects. These toxicities, coupled with observations that SARS-CoV-2 infection is associated with autoantibody development, raise questions whether COVID-19 vaccines may also promote the development of autoantibodies, particularly in autoimmune patients. Here we used Rapid Extracellular Antigen Profiling to characterize self- and viral-directed humoral responses after SARS-CoV-2 mRNA vaccination in 145 healthy individuals, 38 patients with autoimmune diseases, and 8 patients with mRNA vaccine-associated myocarditis. We confirm that most individuals generated robust virus-specific antibody responses post vaccination, but that the quality of this response is impaired in autoimmune patients on certain modes of immunosuppression. Autoantibody dynamics are remarkably stable in all vaccinated patients compared to COVID-19 patients that exhibit an increased prevalence of new autoantibody reactivities. Patients with vaccine-associated myocarditis do not have increased autoantibody reactivities relative to controls. In summary, our findings indicate that mRNA vaccines decouple SARS-CoV-2 immunity from autoantibody responses observed during acute COVID-19.
Collapse
Affiliation(s)
- Jillian R Jaycox
- Department of Immunobiology, Yale School of Medicine, New Haven, CT, USA
| | - Carolina Lucas
- Department of Immunobiology, Yale School of Medicine, New Haven, CT, USA
| | - Inci Yildirim
- Department of Pediatrics, Section of Infectious Diseases and Global Health, Yale University School of Medicine, New Haven, CT, USA
- Yale Institute for Global Health, Yale University, New Haven, CT, USA
| | - Yile Dai
- Department of Immunobiology, Yale School of Medicine, New Haven, CT, USA
| | - Eric Y Wang
- Department of Immunobiology, Yale School of Medicine, New Haven, CT, USA
| | - Valter Monteiro
- Department of Immunobiology, Yale School of Medicine, New Haven, CT, USA
| | - Sandra Lord
- Center for Interventional Immunology, Benaroya Research Institute at Virginia Mason, Seattle, WA, USA
| | | | - Mariko Kita
- Virginia Mason Medical Center, Seattle, WA, USA
| | - Jane H Buckner
- Translational Research Program, Benaroya Research Institute at Virginia Mason, Seattle, WA, USA
| | - Shuangge Ma
- Department of Biostatistics, Yale School of Public Health, New Haven, CT, USA
| | - Melissa Campbell
- Department of Medicine, Section of Infectious Diseases, Yale University School of Medicine, New Haven, CT, USA
| | - Albert Ko
- Department of Medicine, Section of Infectious Diseases, Yale University School of Medicine, New Haven, CT, USA
- Department of Epidemiology of Microbial Diseases, Yale School of Public Health, New Haven, CT, USA
| | - Saad Omer
- Yale Institute for Global Health, Yale University, New Haven, CT, USA
- Department of Medicine, Section of Infectious Diseases, Yale University School of Medicine, New Haven, CT, USA
- Department of Epidemiology of Microbial Diseases, Yale School of Public Health, New Haven, CT, USA
| | - Carrie L Lucas
- Department of Immunobiology, Yale School of Medicine, New Haven, CT, USA
| | - Cate Speake
- Center for Interventional Immunology, Benaroya Research Institute at Virginia Mason, Seattle, WA, USA.
| | - Akiko Iwasaki
- Department of Immunobiology, Yale School of Medicine, New Haven, CT, USA.
- Howard Hughes Medical Institute, Chevy Chase, MD, USA.
| | - Aaron M Ring
- Department of Immunobiology, Yale School of Medicine, New Haven, CT, USA.
- Department of Pharmacology, Yale School of Medicine, New Haven, CT, USA.
| |
Collapse
|
18
|
Badaró R, Novaes G, Andrade AC, de Araujo Neto CA, Machado BA, Barbosa JDV, Soares MBP. Myocardial infarction or myocarditis? A case report and review of a myocardial adverse event associated with mRNA vaccine. Front Med (Lausanne) 2023; 10:1071239. [PMID: 36817791 PMCID: PMC9932914 DOI: 10.3389/fmed.2023.1071239] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2022] [Accepted: 01/02/2023] [Indexed: 02/05/2023] Open
Abstract
A 23-year-old man started with chest pain 8 h after his first Pfizer-BioNTech COVID-19 vaccination. ECG evaluation showed sinus tachycardia with ST-segment elevation in D1, AVL, V5, and V6, the findings compatible with acute subepicardial myocardial damage. However, cardiac MRI documented myocardial fibrosis, with cardiac late enhancement non-ischemic pattern with diffuse edema. He had no other symptoms to suggest another etiology than the vaccination. The patient was hospitalized and received corticosteroid (prednisolone) daily. Then, 2 weeks after hospitalization, all laboratory parameters and ECG were normal and the patient was discharged from the hospital. The patient had a history of Wolf-Parkinson White that was corrected with ablation when he was 11 years old. This report calls attention to myocardial adverse reaction risk for mRNA COVID-19 vaccines for people with a previous cardiac disease history.
Collapse
Affiliation(s)
- Roberto Badaró
- SENAI Institute of Innovation (ISI) in Health Advanced Systems, University Center SENAI/CIMATEC, Salvador, Brazil,Allmed Specialized Clinic, Salvador, Brazil,*Correspondence: Roberto Badaró,
| | | | | | | | - Bruna Aparecida Machado
- SENAI Institute of Innovation (ISI) in Health Advanced Systems, University Center SENAI/CIMATEC, Salvador, Brazil
| | - Josiane Dantas Viana Barbosa
- SENAI Institute of Innovation (ISI) in Health Advanced Systems, University Center SENAI/CIMATEC, Salvador, Brazil
| | - Milena Botelho Pereira Soares
- SENAI Institute of Innovation (ISI) in Health Advanced Systems, University Center SENAI/CIMATEC, Salvador, Brazil,Gonçalo Moniz Institute, Oswaldo Cruz Foundation (FIOCRUZ), Salvador, Brazil,Milena Botelho Pereira Soares,
| |
Collapse
|
19
|
Mauro AG, Mezzaroma E, Toldo S, Melendez GC, Franco RL, Lesnefsky EJ, Abbate A, Hundley WG, Salloum FN. NLRP3-mediated inflammation in cardio-oncology: sterile yet harmful. Transl Res 2023; 252:9-20. [PMID: 35948198 PMCID: PMC9839540 DOI: 10.1016/j.trsl.2022.08.004] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/02/2022] [Revised: 07/28/2022] [Accepted: 08/03/2022] [Indexed: 01/17/2023]
Abstract
Despite significant advances and the continuous development of novel, effective therapies to treat a variety of malignancies, cancer therapy-induced cardiotoxicity has been identified as a prominent cause of morbidity and mortality, closely competing with secondary malignancies. This unfortunate limitation has prompted the inception of the field of cardio-oncology with its purpose to provide the necessary knowledge and key information on mechanisms that support the use of the most efficacious cancer therapy with minimal or no interruption while paying close attention to preventing cardiovascular related morbidity and mortality. Several mechanisms that contribute to cancer therapy-induced cardiotoxicity have been proposed and studied. These mainly involve mitochondrial dysfunction and reactive oxygen species-induced oxidative stress, lysosomal damage, impaired autophagy, cell senescence, DNA damage, and sterile inflammation with the formation and activation of the NLRP3 inflammasome. In this review, we focus on describing the principal mechanisms for different classes of cancer therapies that lead to cardiotoxicity involving the NLRP3 inflammasome. We also summarize current evidence of cardio-protection with inflammasome inhibitors in the context of heart disease in general, and further highlight the potential application of this evidence for clinical translation in at risk patients for the purpose of preventing cancer therapy associated cardiovascular morbidity and mortality.
Collapse
Affiliation(s)
- Adolfo G Mauro
- Pauley Heart Center, Department of Internal Medicine, Cardiology, Virginia Commonwealth University, Richmond, VA
| | - Eleonora Mezzaroma
- Pauley Heart Center, Department of Internal Medicine, Cardiology, Virginia Commonwealth University, Richmond, VA
| | - Stefano Toldo
- Pauley Heart Center, Department of Internal Medicine, Cardiology, Virginia Commonwealth University, Richmond, VA
| | - Giselle C Melendez
- Department of Internal Medicine, Sections on Cardiovascular Medicine, Department of Pathology, Section on Comparative Medicine, Wake Forest, School of Medicine, Winston-Salem, NC
| | - R Lee Franco
- College of Humanities and Sciences, Department of Kinesiology and Health Sciences, Virginia Commonwealth University, Richmond, VA
| | - Edward J Lesnefsky
- Pauley Heart Center, Department of Internal Medicine, Cardiology, Virginia Commonwealth University, Richmond, VA; Department of the Medical Service of the McGuire Veterans Affairs Medical Center, Richmond, VA
| | - Antonio Abbate
- Pauley Heart Center, Department of Internal Medicine, Cardiology, Virginia Commonwealth University, Richmond, VA
| | - W Gregory Hundley
- Pauley Heart Center, Department of Internal Medicine, Cardiology, Virginia Commonwealth University, Richmond, VA
| | - Fadi N Salloum
- Pauley Heart Center, Department of Internal Medicine, Cardiology, Virginia Commonwealth University, Richmond, VA.
| |
Collapse
|
20
|
Lee KM, Lin SJ, Wu CJ, Kuo RL. Race with virus evolution: The development and application of mRNA vaccines against SARS-CoV-2. Biomed J 2023; 46:70-80. [PMID: 36642222 PMCID: PMC9837160 DOI: 10.1016/j.bj.2023.01.002] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2022] [Accepted: 01/10/2023] [Indexed: 01/15/2023] Open
Abstract
Since the COVID-19 pandemic was declared, vaccines against SARS-CoV-2 have been urgently developed around the world. On the basis of the mRNA vaccine technology developed previously, COVID-19 mRNA vaccines were promptly tested in animals, advanced to clinical trials, and then authorized for emergency use in humans. The administration of COVID-19 mRNA vaccines has successfully reduced the hospitalization and mortality caused by the viral infection, although the virus continuously evolves with its transmission. Therefore, the development of mRNA vaccine technology, including RNA modification and delivery systems, is well recognized for its contribution to moderating the harms caused by the COVID-19 pandemic. The scientists who developed these technologies, Katalin Karikó, Drew Weissman, and Pieter Cullis, were awarded the 2022 Tang Prize in Biopharmaceutical Science. In this review, we summarize the principles, safety and efficacy of as well as the immune response to COVID-19 mRNA vaccines. Since mRNA vaccine approaches could be practical for the prevention of infectious diseases, we also briefly describe mRNA vaccines against other human viral pathogens in clinical trials.
Collapse
Affiliation(s)
- Kuo-Ming Lee
- Research Center for Emerging Viral Infections, College of Medicine, Chang Gung University, Taoyuan, Taiwan,International Master Degree Program for Molecular Medicine in Emerging Viral Infections, College of Medicine, Chang Gung University, Taoyuan, Taiwan,Division of Infectious Diseases, Department of Pediatrics, Chang Gung Memorial Hospital at Linkou, Linkou, Taiwan
| | - Syh-Jae Lin
- Division of Allergy, Asthma, and Rheumatology, Department of Pediatrics, Chang Gung Memorial Hospital at Linkou, Linkou, Taiwan,School of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Chung-Jung Wu
- Research Center for Emerging Viral Infections, College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Rei-Lin Kuo
- Research Center for Emerging Viral Infections, College of Medicine, Chang Gung University, Taoyuan, Taiwan; International Master Degree Program for Molecular Medicine in Emerging Viral Infections, College of Medicine, Chang Gung University, Taoyuan, Taiwan; Division of Allergy, Asthma, and Rheumatology, Department of Pediatrics, Chang Gung Memorial Hospital at Linkou, Linkou, Taiwan; Department of Medical Biotechnology and Laboratory Science, College of Medicine, Chang Gung University, Taoyuan, Taiwan; Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Taoyuan, Taiwan.
| |
Collapse
|
21
|
Valore L, Junker T, Heilmann E, Zuern CS, Streif M, Drexler B, Arranto C, Halter JP, Berger CT. Case report: mRNA-1273 COVID-19 vaccine-associated myopericarditis: Successful treatment and re-exposure with colchicine. Front Cardiovasc Med 2023; 10:1135848. [PMID: 37139123 PMCID: PMC10149711 DOI: 10.3389/fcvm.2023.1135848] [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: 01/22/2023] [Accepted: 03/27/2023] [Indexed: 05/05/2023] Open
Abstract
Introduction Vaccine-induced myocarditis is a rare complication of messenger RNA (mRNA) COVID-19 vaccines. Case presentation We report a case of acute myopericarditis in a recipient of allogeneic hematopoietic cells following the first dose of the mRNA-1273 vaccine and the successful administration of a second and third dose while on prophylactic treatment with colchicine to successfully complete the vaccination. Conclusion Treatment and prevention of mRNA-vaccine-induced myopericarditis represent a clinical challenge. The use of colchicine is feasible and safe to potentially reduce the risk of this rare but severe complication and allows re-exposure to an mRNA vaccine.
Collapse
Affiliation(s)
- Luca Valore
- Hematology, University Hospital Basel, Basel, Switzerland
- Correspondence: Luca Valore
| | - Till Junker
- Hematology, University Hospital Basel, Basel, Switzerland
| | - Eva Heilmann
- Hematology, Cantonal Hospital Aarau, Aarau, Switzerland
| | - Christine S. Zuern
- Cardiology, University Hospital Basel, Basel, Switzerland
- Cardiovascular Research Institute Basel, University of Basel, Basel, Switzerland
| | | | | | | | - Jörg P. Halter
- Hematology, University Hospital Basel, Basel, Switzerland
- Cardiovascular Research Institute Basel, University of Basel, Basel, Switzerland
| | - Christoph T. Berger
- Translational Immunology, University Basel, Basel, Switzerland
- University Center for Immunology, University Hospital Basel, Basel, Switzerland
| |
Collapse
|
22
|
Ferraresi A, Isidoro C. Will Omics Biotechnologies Save Us from Future Pandemics? Lessons from COVID-19 for Vaccinomics and Adversomics. Biomedicines 2022; 11:52. [PMID: 36672560 PMCID: PMC9855897 DOI: 10.3390/biomedicines11010052] [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: 11/29/2022] [Revised: 12/19/2022] [Accepted: 12/22/2022] [Indexed: 12/28/2022] Open
Abstract
The COVID-19 pandemic had cross-cutting impacts on planetary health, quotidian life, and society. Mass vaccination with the current gene-based vaccines has helped control the pandemic but unfortunately it has not shown effectiveness in preventing the spread of the virus. In addition, not all individuals respond to these vaccines, while others develop adverse reactions that cannot be neglected. It is also a fact that some individuals are more susceptible to infection while others develop effective immunization post-infection. We note here that the person-to-person and population variations in vaccine efficacy and side effects have been studied in the field of vaccinomics long before the COVID-19 pandemic. Additionally, the field of adversomics examines the mechanisms of individual differences in the side effects of health interventions. In this review, we discuss the potential of a multi-omics approach for comprehensive profiling of the benefit/risk ratios of vaccines. Vaccinomics and adversomics stand to benefit planetary health and contribute to the prevention of future pandemics in the 21st century by offering precision guidance to clinical trials as well as promoting precision use of vaccines in ways that proactively respond to individual and population differences in their efficacy and safety. This vision of pandemic prevention based on personalized instead of mass vaccination also calls for equity in access to precision vaccines and diagnostics that support a vision and practice of vaccinomics and adversomics in planetary health.
Collapse
Affiliation(s)
| | - Ciro Isidoro
- Laboratory of Molecular Pathology, Department of Health Sciences, Università del Piemonte Orientale, Via Solaroli 17, 28100 Novara, Italy
| |
Collapse
|
23
|
Klingler J, Lambert GS, Bandres JC, Emami-Gorizi R, Nádas A, Oguntuyo KY, Amanat F, Bermúdez-González MC, Gleason C, Kleiner G, Simon V, Lee B, Zolla-Pazner S, Upadhyay C, Hioe CE. Immune profiles to distinguish hospitalized versus ambulatory COVID-19 cases in older patients. iScience 2022; 25:105608. [PMID: 36406863 PMCID: PMC9666267 DOI: 10.1016/j.isci.2022.105608] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Revised: 09/23/2022] [Accepted: 11/11/2022] [Indexed: 11/17/2022] Open
Abstract
A fraction of patients with COVID-19 develops severe disease requiring hospitalization, while the majority, including high-risk individuals, experience mild symptoms. Severe disease has been associated with higher levels of antibodies and inflammatory cytokines but often among patients with diverse demographics and comorbidity status. This study evaluated hospitalized vs. ambulatory patients with COVID-19 with demographic risk factors for severe COVID-19: median age of 63, >80% male, and >85% black and/or Hispanic. Sera were collected four to 243 days after symptom onset and evaluated for binding and functional antibodies as well as 48 cytokines and chemokines. SARS-CoV-2-specific antibody levels and functions were similar in ambulatory and hospitalized patients. However, a strong correlation between anti-S2 antibody levels and the other antibody parameters, along with higher IL-27 levels, was observed in hospitalized but not ambulatory cases. These data indicate that antibodies against the relatively conserved S2 spike subunit and immunoregulatory cytokines such as IL-27 are potential immune determinants of COVID-19.
Collapse
Affiliation(s)
- Jéromine Klingler
- Division of Infectious Diseases, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- James J. Peters VA Medical Center, Bronx, NY, USA
| | - Gregory S. Lambert
- Division of Infectious Diseases, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Juan C. Bandres
- Division of Infectious Diseases, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- James J. Peters VA Medical Center, Bronx, NY, USA
| | | | - Arthur Nádas
- Department of Environment Medicine, NYU School of Medicine, New York, NY, USA
| | | | - Fatima Amanat
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Maria C. Bermúdez-González
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Center for Vaccine Research and Pandemic Preparedness, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Charles Gleason
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Center for Vaccine Research and Pandemic Preparedness, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Giulio Kleiner
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Center for Vaccine Research and Pandemic Preparedness, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Viviana Simon
- Division of Infectious Diseases, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Pathology, Molecular and Cell Based Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Global Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Center for Vaccine Research and Pandemic Preparedness, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Benhur Lee
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Susan Zolla-Pazner
- Division of Infectious Diseases, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Chitra Upadhyay
- Division of Infectious Diseases, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Catarina E. Hioe
- Division of Infectious Diseases, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- James J. Peters VA Medical Center, Bronx, NY, USA
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| |
Collapse
|
24
|
Talotta R. Impaired VEGF-A-Mediated Neurovascular Crosstalk Induced by SARS-CoV-2 Spike Protein: A Potential Hypothesis Explaining Long COVID-19 Symptoms and COVID-19 Vaccine Side Effects? Microorganisms 2022; 10:2452. [PMID: 36557705 PMCID: PMC9784975 DOI: 10.3390/microorganisms10122452] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2022] [Revised: 12/03/2022] [Accepted: 12/10/2022] [Indexed: 12/14/2022] Open
Abstract
Long coronavirus disease-19 (COVID-19) is a newly discovered syndrome characterized by multiple organ manifestations that persist for weeks to months, following the recovery from acute disease. Occasionally, neurological and cardiovascular side effects mimicking long COVID-19 have been reported in recipients of COVID-19 vaccines. Hypothetically, the clinical similarity could be due to a shared pathogenic role of the severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) spike (S) protein produced by the virus or used for immunization. The S protein can bind to neuropilin (NRP)-1, which normally functions as a coreceptor for the vascular endothelial growth factor (VEGF)-A. By antagonizing the docking of VEGF-A to NRP-1, the S protein could disrupt physiological pathways involved in angiogenesis and nociception. One consequence could be the increase in unbound forms of VEGF-A that could bind to other receptors. SARS-CoV-2-infected individuals may exhibit increased plasma levels of VEGF-A during both acute illness and convalescence, which could be responsible for diffuse microvascular and neurological damage. A few studies suggest that serum VEGF-A may also be a potential biomarker for long COVID-19, whereas evidence for COVID-19 vaccines is lacking and merits further investigation.
Collapse
Affiliation(s)
- Rossella Talotta
- Rheumatology Unit, Department of Clinical and Experimental Medicine, University of Messina, University Hospital "G. Martino", 98124 Messina, Italy
| |
Collapse
|
25
|
Spracklen TF, Mendelsohn SC, Butters C, Facey-Thomas H, Stander R, Abrahams D, Erasmus M, Baguma R, Day J, Scott C, Zühlke LJ, Kassiotis G, Scriba TJ, Webb K. IL27 gene expression distinguishes multisystem inflammatory syndrome in children from febrile illness in a South African cohort. Front Immunol 2022; 13:992022. [PMID: 36148243 PMCID: PMC9486543 DOI: 10.3389/fimmu.2022.992022] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Accepted: 08/17/2022] [Indexed: 01/26/2023] Open
Abstract
Introduction Multisystem inflammatory syndrome in children (MIS-C) is a severe acute inflammatory reaction to SARS-CoV-2 infection in children. There is a lack of data describing differential expression of immune genes in MIS-C compared to healthy children or those with other inflammatory conditions and how expression changes over time. In this study, we investigated expression of immune-related genes in South African MIS-C patients and controls. Methods The cohort included 30 pre-treatment MIS-C cases and 54 healthy non-inflammatory paediatric controls. Other controls included 34 patients with juvenile systemic lupus erythematosus, Kawasaki disease or other inflammatory conditions. Longitudinal post-treatment MIS-C specimens were available at various timepoints. Expression of 80 immune-related genes was determined by real-time quantitative PCR. Results A total of 29 differentially expressed genes were identified in pre-treatment MIS-C compared to healthy controls. Up-regulated genes were found to be overrepresented in innate immune pathways including interleukin-1 processing and pyroptosis. Post-treatment follow-up data were available for up to 1,200 hours after first treatment. All down-regulated genes and 17/18 up-regulated genes resolved to normal levels in the timeframe, and all patients clinically recovered. When comparing MIS-C to other febrile conditions, only IL27 expression could differentiate these two groups with high sensitivity and specificity. Conclusions These data indicate a unique 29-gene signature of MIS-C in South African children. The up-regulation of interleukin-1 and pyroptosis pathway genes highlights the role of the innate immune system in MIS-C. IL-27 is a potent anti-inflammatory and antiviral cytokine that may distinguish MIS-C from other conditions in our setting.
Collapse
Affiliation(s)
- Timothy F. Spracklen
- Department of Paediatrics and Child Health, University of Cape Town, Cape Town, South Africa
- Cape Heart Institute, University of Cape Town, Cape Town, South Africa
| | - Simon C. Mendelsohn
- South African Tuberculosis Vaccine Initiative, Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Cape Town, South Africa
| | - Claire Butters
- Department of Paediatrics and Child Health, University of Cape Town, Cape Town, South Africa
- Division of Immunology, Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Cape Town, South Africa
| | - Heidi Facey-Thomas
- Department of Paediatrics and Child Health, University of Cape Town, Cape Town, South Africa
| | - Raphaella Stander
- Department of Paediatrics and Child Health, University of Cape Town, Cape Town, South Africa
| | - Debbie Abrahams
- Department of Paediatrics and Child Health, University of Cape Town, Cape Town, South Africa
| | - Mzwandile Erasmus
- South African Tuberculosis Vaccine Initiative, Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Cape Town, South Africa
| | - Richard Baguma
- South African Tuberculosis Vaccine Initiative, Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Cape Town, South Africa
| | - Jonathan Day
- Department of Paediatrics and Child Health, University of Cape Town, Cape Town, South Africa
| | - Christiaan Scott
- Department of Paediatrics and Child Health, University of Cape Town, Cape Town, South Africa
| | - Liesl J. Zühlke
- Department of Paediatrics and Child Health, University of Cape Town, Cape Town, South Africa
- Cape Heart Institute, University of Cape Town, Cape Town, South Africa
- South African Medical Research Council, Cape Town, South Africa
| | - George Kassiotis
- Retroviral Immunology Laboratory, The Francis Crick Institute, London, United Kingdom
- Department of Infectious Disease, St Mary’s Hospital, Imperial College, London, United Kingdom
| | - Thomas J. Scriba
- South African Tuberculosis Vaccine Initiative, Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Cape Town, South Africa
| | - Kate Webb
- Department of Paediatrics and Child Health, University of Cape Town, Cape Town, South Africa
- Crick African Network, The Francis Crick Institute, London, United Kingdom
| |
Collapse
|
26
|
New-onset dermatomyositis following SARS-CoV-2 infection and vaccination: a case-based review. Rheumatol Int 2022; 42:2267-2276. [PMID: 35939078 PMCID: PMC9358381 DOI: 10.1007/s00296-022-05176-3] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2022] [Accepted: 07/22/2022] [Indexed: 11/25/2022]
Abstract
Dermatomyositis is a rare, type I interferon-driven autoimmune disease, which can affect muscle, skin and internal organs (especially the pulmonary system). In 2021, we have noted an increase in new-onset dermatomyositis compared to the years before the SARS-CoV-2 pandemic in our center. We present four cases of new-onset NXP2 and/or MDA5 positive dermatomyositis shortly after SARS-CoV-2 infection or vaccination. Three cases occurred within days after vaccination with Comirnaty and one case after SARS-CoV-2 infection. All patients required intensive immunosuppressive treatment. MDA5 antibodies could be detected in three patients and NXP2 antibodies were found in two patients (one patient was positive for both antibodies). In this case-based systematic review, we further analyze and discuss the literature on SARS-CoV-2 and associated dermatomyositis. In the literature, sixteen reports (with a total of seventeen patients) of new-onset dermatomyositis in association with a SARS-CoV-2 infection or vaccination were identified. Ten cases occurred after infection and seven after vaccination. All vaccination-associated cases were seen in mRNA vaccines. The reported antibodies included for instance MDA5, NXP2, Mi-2 and TIF1γ. The reviewed literature and our cases suggest that SARS-CoV-2 infection and vaccination may be considered as a potential trigger of interferon-pathway. Consequently, this might serve as a stimulus for the production of dermatomyositis-specific autoantibodies like MDA5 and NXP2 which are closely related to viral defense or viral RNA interaction supporting the concept of infection and vaccination associated dermatomyositis.
Collapse
|
27
|
Hsieh MH, Yamaguchi Y. Immune Response in Regard to Hypersensitivity Reactions after COVID-19 Vaccination. Biomedicines 2022; 10:biomedicines10071641. [PMID: 35884946 PMCID: PMC9312871 DOI: 10.3390/biomedicines10071641] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2022] [Revised: 07/02/2022] [Accepted: 07/05/2022] [Indexed: 11/16/2022] Open
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), the virus that causes coronavirus disease 2019 (COVID-19), is a member of the genus Betacoronavirus. This virus was first detected in December 2019, and the situation quickly escalated to cause a global pandemic within a few months. COVID-19 had caused more than 5.5 million deaths as of January 2022. Hence, the urgency of effective vaccination contributed to the fastest rate of vaccine development seen to date (i.e., within 1.5 years). Despite reports of good vaccine efficacy without severe systemic reactions at the clinical trial stage, hypersensitivity reactions have been reported following worldwide vaccination campaigns. We provide a brief review regarding the structure of SARS-CoV-2. We also review the most acceptable types of vaccines in terms of safety profiles, namely the BNT162b2, mRNA-1273, and AZD1222 vaccines. This review aims to facilitate an understanding of the possible immune mechanisms regarding COVID-19-vaccination-related hypersensitivity reactions, such as thrombosis and thrombocytopenia, cutaneous adverse reactions, myocarditis, and perimyocarditis.
Collapse
|
28
|
Moosmann J, Gentles T, Occleshaw C, Mitchelson B. COVID Vaccine-Associated Myocarditis in Adolescent Siblings: Does It Run in the Family? Vaccines (Basel) 2022; 10:vaccines10040611. [PMID: 35455360 PMCID: PMC9028976 DOI: 10.3390/vaccines10040611] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Revised: 04/12/2022] [Accepted: 04/12/2022] [Indexed: 12/18/2022] Open
Abstract
The development of myocarditis after receiving messenger RNA vaccination against COVID-19 is well documented, particularly in adolescent and young adult males. We report a case of vaccine-associated myocarditis in adolescent brothers following their second dose of the BNT162b2 mRNA vaccine (Pfizer-BioNTech, Mainz, Germany). This report illustrates the need to better understand the mechanisms leading to myocarditis after mRNA vaccination.
Collapse
Affiliation(s)
- Julia Moosmann
- Green Lane Paediatric and Congenital Cardiac Service, Starship Children’s Hospital, Auckland 1023, New Zealand; (J.M.); (T.G.)
| | - Thomas Gentles
- Green Lane Paediatric and Congenital Cardiac Service, Starship Children’s Hospital, Auckland 1023, New Zealand; (J.M.); (T.G.)
- Paediatrics, Child and Youth Health, University of Auckland, Auckland 1023, New Zealand
| | | | - Bryan Mitchelson
- Green Lane Paediatric and Congenital Cardiac Service, Starship Children’s Hospital, Auckland 1023, New Zealand; (J.M.); (T.G.)
- Correspondence: ; Tel.: +64-(0)21-624-346
| |
Collapse
|
29
|
Elasomeran. REACTIONS WEEKLY 2022. [PMCID: PMC8990599 DOI: 10.1007/s40278-022-12908-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
|