1
|
Lind A, Naredi Scherman M, Hamdan S, Agardh D. Risk of celiac disease, type 1 diabetes, and thyroid disease autoimmunity during the SARS-CoV-2 pandemic in South of Sweden: insights from the TRIAD study. Autoimmunity 2025; 58:2490491. [PMID: 40230327 DOI: 10.1080/08916934.2025.2490491] [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: 08/13/2024] [Revised: 04/02/2025] [Accepted: 04/03/2025] [Indexed: 04/16/2025]
Abstract
Recent studies have implied an increased incidence of autoimmune diseases following the SARS-CoV-2 pandemic. The objective was to determine if SARS-CoV-2 infections were associated with celiac disease (CD), type 1 diabetes (T1D), and autoimmune thyroid disease (AITD) autoantibodies in a population-based screening when the pandemic hit the South of Sweden during 2021 and 2022. Between August 2021 and June 2022 self-obtained capillary plasma samples were collected from 1088 children at 6-9 years of age and 1185 adolescents at 13-16 years of age, who were randomly invited from the general population to a screening for CD, T1D, AITD, and SARS-CoV-2 antibodies. Among children and adolescents screened for autoantibodies associated with CD, T1D and AITD, the SARS-CoV-2 infection rate was increased in tissue transglutaminase autoantibody (tTGA) positive (13/17; 76.5%) compared with tTGA negative (492/1168; 42.1%) 13-16-year-old individuals (p = 0.0057). There was no association between SARS-CoV-2 infection rate and AITD- or T1D autoantibodies. Our findings indicate a potential association between prior SARS-CoV-2 infection and screening-detected CD autoimmunity in adolescents aged 13-16 years. Further research is needed to elucidate whether ongoing CD autoimmunity increases susceptibility to infection or if SARS-CoV-2 may act as a trigger for CD autoimmunity in genetically and environmentally predisposed individuals.
Collapse
Affiliation(s)
- Alexander Lind
- Department of Clinical Sciences Malmö, Lund University, Malmö, Sweden
| | | | - Samia Hamdan
- Department of Clinical Sciences Malmö, Lund University, Malmö, Sweden
| | - Daniel Agardh
- Department of Clinical Sciences Malmö, Lund University, Malmö, Sweden
| |
Collapse
|
2
|
Saidoune F, Lee D, Di Domizio J, Le Floc’h C, Jenelten R, Le Pen J, Bondet V, Joncic A, Morren MA, Béziat V, Zhang SY, Jouanguy E, Duffy D, Rice CM, Conrad C, Fellay J, Casanova JL, Gilliet M, Yatim A. Enhanced TLR7-dependent production of type I interferon by pDCs underlies pandemic chilblains. J Exp Med 2025; 222:e20231467. [PMID: 40227192 PMCID: PMC11995862 DOI: 10.1084/jem.20231467] [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: 08/18/2023] [Revised: 01/02/2025] [Accepted: 02/28/2025] [Indexed: 04/15/2025] Open
Abstract
Outbreaks of chilblains were reported during the COVID-19 pandemic. Given the essential role of type I interferon (I-IFN) in protective immunity against SARS-CoV-2 and the association of chilblains with inherited type I interferonopathies, we hypothesized that excessive I-IFN responses to SARS-CoV-2 might underlie the occurrence of chilblains in this context. We identified a transient I-IFN signature in chilblain lesions, accompanied by an acral infiltration of activated plasmacytoid dendritic cells (pDCs). Patients with chilblains were otherwise asymptomatic or had mild disease without seroconversion. Their leukocytes produced abnormally high levels of I-IFN upon TLR7 stimulation with agonists or ssRNA viruses-particularly SARS-CoV-2-but not with DNA agonists of TLR9 or the dsDNA virus HSV-1. Moreover, the patients' pDCs displayed cell-intrinsic hyperresponsiveness to TLR7 stimulation regardless of TLR7 levels. Inherited TLR7 or I-IFN deficiency confers a predisposition to life-threatening COVID-19. Conversely, our findings suggest that enhanced TLR7 activity in predisposed individuals could confer innate, pDC-mediated, sterilizing immunity to SARS-CoV-2 infection, with I-IFN-driven chilblains as a trade-off.
Collapse
Affiliation(s)
- Fanny Saidoune
- Department of Dermatology, CHUV University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Danyel Lee
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, The Rockefeller University, New York, NY, USA
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM U1163, Paris, France
- Imagine Institute, Paris Cité University, Paris, France
| | - Jeremy Di Domizio
- Department of Dermatology, CHUV University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Corentin Le Floc’h
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, The Rockefeller University, New York, NY, USA
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM U1163, Paris, France
- Imagine Institute, Paris Cité University, Paris, France
| | - Raphael Jenelten
- Department of Dermatology, CHUV University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Jérémie Le Pen
- Laboratory of Virology and Infectious Disease, The Rockefeller University, New York, NY, USA
| | - Vincent Bondet
- Translational Immunology Unit, Institut Pasteur, Université Paris Cité, Paris, France
| | - Ana Joncic
- Department of Dermatology, CHUV University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Marie-Anne Morren
- Department of Dermatology, CHUV University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Vivien Béziat
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, The Rockefeller University, New York, NY, USA
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM U1163, Paris, France
- Imagine Institute, Paris Cité University, Paris, France
| | - Shen-Ying Zhang
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, The Rockefeller University, New York, NY, USA
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM U1163, Paris, France
- Imagine Institute, Paris Cité University, Paris, France
| | - Emmanuelle Jouanguy
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, The Rockefeller University, New York, NY, USA
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM U1163, Paris, France
- Imagine Institute, Paris Cité University, Paris, France
| | - Darragh Duffy
- Translational Immunology Unit, Institut Pasteur, Université Paris Cité, Paris, France
| | - Charles M. Rice
- Laboratory of Virology and Infectious Disease, The Rockefeller University, New York, NY, USA
| | - Curdin Conrad
- Department of Dermatology, CHUV University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Jacques Fellay
- School of Life Sciences, Ecole Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
- Precision Medicine Unit, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Jean-Laurent Casanova
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, The Rockefeller University, New York, NY, USA
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM U1163, Paris, France
- Imagine Institute, Paris Cité University, Paris, France
- Department of Pediatrics, Necker Hospital for Sick Children, Paris, France
- Howard Hughes Medical Institute, New York, NY, USA
| | - Michel Gilliet
- Department of Dermatology, CHUV University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Ahmad Yatim
- Department of Dermatology, CHUV University Hospital and University of Lausanne, Lausanne, Switzerland
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, The Rockefeller University, New York, NY, USA
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM U1163, Paris, France
- Imagine Institute, Paris Cité University, Paris, France
| |
Collapse
|
3
|
Kreslavsky T. Thymflammation: The Role of a Constitutively Active Inflammatory Network and "Ectopic" Cell Types in the Thymus in the Induction of T Cell Tolerance and Beyond. Immunol Rev 2025; 332:e70037. [PMID: 40433806 PMCID: PMC12117520 DOI: 10.1111/imr.70037] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2025] [Revised: 03/28/2025] [Accepted: 05/13/2025] [Indexed: 05/29/2025]
Abstract
The thymus exhibits constitutive activation of nearly all major inflammatory pathways, including sterile MyD88-dependent signaling and interferon production by mTECs, the presence of cellular and molecular components of type 1, type 2, and type 3 responses, as well as sustained B cell activation. The reasons for the existence of such a complex constitutively active inflammatory network at the site of T cell development-where the initial pathogen encounter is unlikely-have remained enigmatic. We propose that this inflammatory thymic 'ecosystem' has evolved to promote immunological tolerance to 'inflammatory self'-endogenous molecules absent from most peripheral tissues at steady state but upregulated during pathogen invasion. The spatial and temporal overlap with pathogen presence makes the discrimination of the inflammatory self from pathogen-derived molecules a unique challenge for the adaptive immune system. The frequent occurrence of diseases associated with autoantibodies against proinflammatory cytokines underscores the persistent risk of these molecules being misidentified as foreign. Their abundant representation in the thymus, therefore, is likely to be critical for maintaining tolerance. This review explores current insights into the thymic inflammatory network, its cellular and molecular constituents, and their role in the induction of T cell tolerance.
Collapse
Affiliation(s)
- Taras Kreslavsky
- Division of Immunology and Respiratory Medicine, Department of Medicine Solna, Karolinska InstitutetKarolinska University HospitalStockholmSweden
- Center for Molecular MedicineKarolinska University HospitalStockholmSweden
| |
Collapse
|
4
|
Antolí A, Gómez-Vázquez JL, Sierra-Fortuny A, Bermudez-Carre C, Framil M, Creus-Bachiller E, Viana-Errasti J, Rofes P, Rocamora-Blanch G, Hidalgo-Peña L, García-Serrano L, Rigo-Bonnin R, Feliubadaló L, Del Valle J, Calatayud L, Morandeira F, Lázaro C, Solanich X. Autoantibodies neutralizing type I interferons remain a significant risk factor for critical COVID-19 pneumonia in vaccinated patients. Clin Immunol 2025; 276:110491. [PMID: 40185298 DOI: 10.1016/j.clim.2025.110491] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2025] [Revised: 03/27/2025] [Accepted: 03/31/2025] [Indexed: 04/07/2025]
Abstract
BACKGROUND Neutralizing autoantibodies against type I interferons were strongly linked to severe COVID-19 in unvaccinated patients; however, this has yet to be evaluated in vaccinated individuals. OBJECTIVE To analyze how these autoantibodies influences disease variability in vaccine breakthrough COVID-19 pneumonia patients. METHODS Clinical and laboratory data; autoantibodies blocking interferon-α2 and -ω; and humoral response to SARS-CoV-2 vaccine were collected from all vaccinated COVID-19 pneumonia patients admitted from April 2021 to December 2022 at Bellvitge University Hospital, Spain. RESULTS 458 patients developed COVID-19 pneumonia despite an appropriate antibody response to SARS-CoV-2 vaccination. Autoantibodies neutralizing interferons were significantly more prevalent in patients with critical pneumonia compared to those with milder forms (8.8 % vs. 3.6 %; p = 0.037). Having these autoantibodies was an independent predictor for critical COVID-19 pneumonia (OR 2.88 [95 %CI 1.18-6.98]). CONCLUSION Vaccination considerably reduces the severity of COVID-19; however, patients with type I interferon autoantibodies remain at increased risk of severe disease.
Collapse
Affiliation(s)
- Arnau Antolí
- Internal Medicine Department, Hospital Universitari de Bellvitge. L'Hospitalet de Llobregat, Barcelona, Spain; Adult Primary Immunodeficiency Unit (UFIPA), Bellvitge University Hospital, L'Hospitalet de Llobregat, Barcelona, Spain; Bellvitge Biomedical Research Institute (IDIBELL), L'Hospitalet de Llobregat, Barcelona, Spain; Faculty of Medicine and Health Sciences, Universitat de Barcelona, Barcelona, Spain
| | - José Luis Gómez-Vázquez
- Adult Primary Immunodeficiency Unit (UFIPA), Bellvitge University Hospital, L'Hospitalet de Llobregat, Barcelona, Spain; Bellvitge Biomedical Research Institute (IDIBELL), L'Hospitalet de Llobregat, Barcelona, Spain; Faculty of Medicine and Health Sciences, Universitat de Barcelona, Barcelona, Spain
| | - Angels Sierra-Fortuny
- Adult Primary Immunodeficiency Unit (UFIPA), Bellvitge University Hospital, L'Hospitalet de Llobregat, Barcelona, Spain; Bellvitge Biomedical Research Institute (IDIBELL), L'Hospitalet de Llobregat, Barcelona, Spain
| | - Carla Bermudez-Carre
- Adult Primary Immunodeficiency Unit (UFIPA), Bellvitge University Hospital, L'Hospitalet de Llobregat, Barcelona, Spain; Bellvitge Biomedical Research Institute (IDIBELL), L'Hospitalet de Llobregat, Barcelona, Spain
| | - Mario Framil
- Adult Primary Immunodeficiency Unit (UFIPA), Bellvitge University Hospital, L'Hospitalet de Llobregat, Barcelona, Spain; Faculty of Medicine and Health Sciences, Universitat de Barcelona, Barcelona, Spain; Immunology Department, Hospital Universitari de Bellvitge, L'Hospitalet de Llobregat, Barcelona, Spain
| | - Edgar Creus-Bachiller
- Hereditary Cancer Program, Catalan Institute of Oncology, ICO, Hospitalet de Llobregat, Barcelona, Spain; Program in Molecular Mechanisms and Experimental Therapy in Oncology (Oncobell), IDIBELL, Hospitalet de Llobregat, Barcelona, Spain; Biomedical Research Centre in Cancer Network (CIBERONC), Carlos III Health Institute, Madrid, Spain
| | - Julen Viana-Errasti
- Hereditary Cancer Program, Catalan Institute of Oncology, ICO, Hospitalet de Llobregat, Barcelona, Spain; Program in Molecular Mechanisms and Experimental Therapy in Oncology (Oncobell), IDIBELL, Hospitalet de Llobregat, Barcelona, Spain
| | - Paula Rofes
- Hereditary Cancer Program, Catalan Institute of Oncology, ICO, Hospitalet de Llobregat, Barcelona, Spain; Program in Molecular Mechanisms and Experimental Therapy in Oncology (Oncobell), IDIBELL, Hospitalet de Llobregat, Barcelona, Spain; Biomedical Research Centre in Cancer Network (CIBERONC), Carlos III Health Institute, Madrid, Spain
| | - Gemma Rocamora-Blanch
- Internal Medicine Department, Hospital Universitari de Bellvitge. L'Hospitalet de Llobregat, Barcelona, Spain; Adult Primary Immunodeficiency Unit (UFIPA), Bellvitge University Hospital, L'Hospitalet de Llobregat, Barcelona, Spain; Bellvitge Biomedical Research Institute (IDIBELL), L'Hospitalet de Llobregat, Barcelona, Spain; Faculty of Medicine and Health Sciences, Universitat de Barcelona, Barcelona, Spain
| | - Lara Hidalgo-Peña
- Internal Medicine Department, Hospital Universitari de Bellvitge. L'Hospitalet de Llobregat, Barcelona, Spain; Adult Primary Immunodeficiency Unit (UFIPA), Bellvitge University Hospital, L'Hospitalet de Llobregat, Barcelona, Spain
| | - Lydia García-Serrano
- Adult Primary Immunodeficiency Unit (UFIPA), Bellvitge University Hospital, L'Hospitalet de Llobregat, Barcelona, Spain; Immunology Department, Hospital Universitari de Bellvitge, L'Hospitalet de Llobregat, Barcelona, Spain
| | - Raúl Rigo-Bonnin
- Clinical Laboratory Department, Hospital Universitari de Bellvitge, L'Hospitalet de Llobregat, Barcelona, Spain
| | - Lidia Feliubadaló
- Hereditary Cancer Program, Catalan Institute of Oncology, ICO, Hospitalet de Llobregat, Barcelona, Spain; Program in Molecular Mechanisms and Experimental Therapy in Oncology (Oncobell), IDIBELL, Hospitalet de Llobregat, Barcelona, Spain; Biomedical Research Centre in Cancer Network (CIBERONC), Carlos III Health Institute, Madrid, Spain
| | - Jesús Del Valle
- Hereditary Cancer Program, Catalan Institute of Oncology, ICO, Hospitalet de Llobregat, Barcelona, Spain; Program in Molecular Mechanisms and Experimental Therapy in Oncology (Oncobell), IDIBELL, Hospitalet de Llobregat, Barcelona, Spain; Biomedical Research Centre in Cancer Network (CIBERONC), Carlos III Health Institute, Madrid, Spain
| | - Laura Calatayud
- Microbiology Department, Hospital Universitari de Bellvitge, L'Hospitalet de Llobregat, Barcelona, Spain; Biomedical Research Centre in Respiratory Diseases Network (CIBERES), Carlos III Health Institute, Madrid, Spain
| | - Francisco Morandeira
- Adult Primary Immunodeficiency Unit (UFIPA), Bellvitge University Hospital, L'Hospitalet de Llobregat, Barcelona, Spain; Immunology Department, Hospital Universitari de Bellvitge, L'Hospitalet de Llobregat, Barcelona, Spain
| | - Conxi Lázaro
- Bellvitge Biomedical Research Institute (IDIBELL), L'Hospitalet de Llobregat, Barcelona, Spain; Faculty of Medicine and Health Sciences, Universitat de Barcelona, Barcelona, Spain; Hereditary Cancer Program, Catalan Institute of Oncology, ICO, Hospitalet de Llobregat, Barcelona, Spain; Program in Molecular Mechanisms and Experimental Therapy in Oncology (Oncobell), IDIBELL, Hospitalet de Llobregat, Barcelona, Spain; Biomedical Research Centre in Cancer Network (CIBERONC), Carlos III Health Institute, Madrid, Spain
| | - Xavier Solanich
- Internal Medicine Department, Hospital Universitari de Bellvitge. L'Hospitalet de Llobregat, Barcelona, Spain; Adult Primary Immunodeficiency Unit (UFIPA), Bellvitge University Hospital, L'Hospitalet de Llobregat, Barcelona, Spain; Bellvitge Biomedical Research Institute (IDIBELL), L'Hospitalet de Llobregat, Barcelona, Spain; Faculty of Medicine and Health Sciences, Universitat de Barcelona, Barcelona, Spain; Biomedical Research Centre in Obesity and Nutrition Physiopathology Network (CIBEROBN), Carlos III Health Institute, Madrid, Spain.
| |
Collapse
|
5
|
Böröcz K, Szinger D, Simon D, Berki T, Németh P. Regulators and Conductors of Immunity: Natural Immune System in Health and Autoimmunity. Int J Mol Sci 2025; 26:5413. [PMID: 40508221 PMCID: PMC12155508 DOI: 10.3390/ijms26115413] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2025] [Revised: 05/20/2025] [Accepted: 05/27/2025] [Indexed: 06/16/2025] Open
Abstract
Natural autoantibodies (nAAbs) recognize self-antigens and are an important component of the immune system, having evolved from invertebrates to vertebrates, and are viewed as stable byproducts of immune function and essential players in health and disease. Initially characterized by their conserved nature and multi-reactivity, primarily as IgM isotypes, nAAbs are now recognized for their adaptability in response to infections and vaccinations, bridging innate and adaptive immunity. The nAAbs and the cellular elements, such as γδ T, iNKT, and MAIT cells, of the natural immune system perform a primary defense network with moderate antigen-specificity. This comprehensive literature review was conducted to analyze the role of natural autoantibodies (nAAbs) in health and disease. The review focused on research published over the past 40 years, emphasizing studies related to infectious diseases, vaccinations, and autoimmune disorders. Recent studies suggest that nAAbs engage in complex interactions in autoimmune diseases, including systemic lupus erythematosus, rheumatoid arthritis, systemic sclerosis, and type 1 diabetes. Their roles in immunological processes, such as maternal tolerance during pregnancy, further underscore their complexity. Emerging evidence indicates that nAAbs and the cellular elements of the natural immune system may contribute to both disease pathogenesis and protective mechanisms, highlighting their dual nature. Continued research on nAAbs is vital for improving our understanding of immune responses and developing therapeutic strategies for autoimmune disorders and infectious diseases.
Collapse
Affiliation(s)
- Katalin Böröcz
- Department of Immunology and Biotechnology, Clinical Center, Medical School, University of Pécs, 7624 Pécs, Hungary
| | - Dávid Szinger
- Department of Immunology and Biotechnology, Clinical Center, Medical School, University of Pécs, 7624 Pécs, Hungary
| | - Diána Simon
- Department of Immunology and Biotechnology, Clinical Center, Medical School, University of Pécs, 7624 Pécs, Hungary
| | - Timea Berki
- Department of Immunology and Biotechnology, Clinical Center, Medical School, University of Pécs, 7624 Pécs, Hungary
| | - Péter Németh
- Medical School, University of Pécs, 7624 Pécs, Hungary
| |
Collapse
|
6
|
Vinh DC. Human immunity to fungal infections. J Exp Med 2025; 222:e20241215. [PMID: 40232283 PMCID: PMC11998751 DOI: 10.1084/jem.20241215] [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: 12/02/2024] [Revised: 01/07/2025] [Accepted: 03/31/2025] [Indexed: 04/16/2025] Open
Abstract
Fungi increasingly threaten health globally. Mycoses range from life-threatening, often iatrogenic conditions, to enigmatic syndromes occurring without apparent immunosuppression. Despite some recent advances in antifungal drug development, complementary therapeutic strategies are essential for addressing these opportunistic pathogens. One promising avenue is leveraging host immunity to combat fungal infections; this necessitates deeper understanding of the molecular immunology of human fungal susceptibility to differentiate beneficial versus harmful immunopathological responses. Investigating human models of fungal diseases in natural settings, particularly through genetic immunodeficiencies and ethnographic-specific genetic vulnerabilities, reveals crucial immune pathways essential for fighting various yeasts and molds. This review highlights the diversity in intrinsic fungal susceptibility across individuals and populations, through genetic- and autoantibody-mediated processes, complementing previous principles learned from animal studies and iatrogenic contexts. Improved understanding of human immunity to fungal diseases will facilitate the development of host-directed immunotherapies and targeted public health interventions, paving the way for precision medicine in fungal disease management.
Collapse
Affiliation(s)
- Donald C. Vinh
- Department of Medicine (Division of Infectious Diseases), McGill University Health Center, Montreal, Canada
- Department of OptiLab (Division of Medical Microbiology, Division of Molecular Genetics-Immunology), McGill University Health Center, Montreal, Canada
- Department of Human Genetics, McGill University, Montreal, Canada
- Center of Reference for Genetic Research in Infection and Immunity, McGill University Health Center Research Institute, Montreal, Canada
| |
Collapse
|
7
|
Groen K, Kuratli R, Enkelmann J, Fernbach S, Wendel-Garcia PD, Staiger WI, Lejeune M, Sauras-Colón E, Roche-Campo F, Filippidis P, Rauch A, Trkola A, Günthard HF, Kouyos RD, Brugger SD, Hale BG. Type I interferon autoantibody footprints reveal neutralizing mechanisms and allow inhibitory decoy design. J Exp Med 2025; 222:e20242039. [PMID: 40111224 PMCID: PMC11924951 DOI: 10.1084/jem.20242039] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2024] [Revised: 01/14/2025] [Accepted: 02/14/2025] [Indexed: 03/22/2025] Open
Abstract
Autoantibodies neutralizing type I interferons (IFN-Is; IFNα or IFNω) exacerbate severe viral disease, but specific treatments are unavailable. With footprint profiling, we delineate two dominant IFN-I faces commonly recognized by neutralizing IFN-I autoantibody-containing plasmas from aged individuals with HIV-1 and from individuals with severe COVID-19. These faces overlap with IFN-I regions independently essential for engaging the IFNAR1/IFNAR2 heterodimer, and neutralizing plasmas efficiently block the interaction of IFN-I with both receptor subunits in vitro. In contrast, non-neutralizing autoantibody-containing plasmas limit the interaction of IFN-I with only one receptor subunit and display relatively low IFN-I-binding avidities, thus likely hindering neutralizing function. Iterative engineering of signaling-inert mutant IFN-Is (simIFN-Is) retaining dominant autoantibody targets created potent decoys that prevent IFN-I neutralization by autoantibody-containing plasmas and that restore IFN-I-mediated antiviral activity. Additionally, microparticle-coupled simIFN-Is were effective at depleting IFN-I autoantibodies from plasmas, leaving antiviral antibodies unaffected. Our study reveals mechanisms of action for IFN-I autoantibodies and demonstrates a proof-of-concept strategy to alleviate pathogenic effects.
Collapse
Affiliation(s)
- Kevin Groen
- Institute of Medical Virology, University of Zurich, Zurich, Switzerland
| | - Roger Kuratli
- Institute of Medical Virology, University of Zurich, Zurich, Switzerland
| | - Jannik Enkelmann
- Institute of Medical Virology, University of Zurich, Zurich, Switzerland
| | - Sonja Fernbach
- Institute of Medical Virology, University of Zurich, Zurich, Switzerland
| | - Pedro D. Wendel-Garcia
- Institute of Intensive Care Medicine, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Willy I. Staiger
- Department of Infectious Diseases and Hospital Epidemiology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Marylène Lejeune
- Biobank IISPV-Node Tortosa, Hospital Verge de la Cinta, Institut d’Investigació Sanitària Pere Virgili (IISPV), Tortosa, Spain
| | - Esther Sauras-Colón
- Clinical Studies Unit, Hospital Verge de la Cinta, Institut d’Investigació Sanitària Pere Virgili (IISPV), Tortosa, Spain
| | - Ferran Roche-Campo
- Intensive Care Unit, Hospital Verge de la Cinta, Institut d’Investigació Sanitària Pere Virgili (IISPV), Tortosa, Spain
| | - Paraskevas Filippidis
- Infectious Diseases Service, Department of Medicine, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Andri Rauch
- Department of Infectious Diseases, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Alexandra Trkola
- Institute of Medical Virology, University of Zurich, Zurich, Switzerland
| | - Huldrych F. Günthard
- Institute of Medical Virology, University of Zurich, Zurich, Switzerland
- Department of Infectious Diseases and Hospital Epidemiology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Roger D. Kouyos
- Institute of Medical Virology, University of Zurich, Zurich, Switzerland
- Department of Infectious Diseases and Hospital Epidemiology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Silvio D. Brugger
- Department of Infectious Diseases and Hospital Epidemiology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Benjamin G. Hale
- Institute of Medical Virology, University of Zurich, Zurich, Switzerland
| |
Collapse
|
8
|
Shih YS, Tsai WT, Guo BC, Cheng A, Wang JT. Life-threatening COVID-19 in a thymoma patient with anti-interferon-α autoantibodies. JOURNAL OF MICROBIOLOGY, IMMUNOLOGY, AND INFECTION = WEI MIAN YU GAN RAN ZA ZHI 2025; 58:376-378. [PMID: 39929767 DOI: 10.1016/j.jmii.2025.02.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/11/2024] [Revised: 01/03/2025] [Accepted: 02/03/2025] [Indexed: 05/17/2025]
Abstract
Neutralizing anti-interferon (IFN)-α autoantibodies can lead to immune dysregulation, potentially resulting in critical coronavirus disease 2019 (COVID-19). We report a case presenting with severe COVID-19 who was subsequently diagnosed to have thymoma and neutralizing anti-IFN-α autoantibodies.
Collapse
Affiliation(s)
- Yu-Shan Shih
- Department of Internal Medicine, National Taiwan University Hospital and National Taiwan University College of Medicine, Taipei, 100225, Taiwan
| | - Wan-Ting Tsai
- Department of Internal Medicine, Taipei City Hospital, Zhongxiao Branch, Taipei, 103212, Taiwan
| | - Bei-Chia Guo
- Department of Internal Medicine, National Taiwan University Hospital and National Taiwan University College of Medicine, Taipei, 100225, Taiwan
| | - Aristine Cheng
- Department of Internal Medicine, National Taiwan University Hospital and National Taiwan University College of Medicine, Taipei, 100225, Taiwan.
| | - Jann-Tay Wang
- Department of Internal Medicine, National Taiwan University Hospital and National Taiwan University College of Medicine, Taipei, 100225, Taiwan
| |
Collapse
|
9
|
Noma K, Asano T, Taniguchi M, Ashihara K, Okada S. Anti-cytokine autoantibodies in human susceptibility to infectious diseases: insights from Inborn errors of immunity. Immunol Med 2025; 48:124-140. [PMID: 40197228 DOI: 10.1080/25785826.2025.2488553] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2024] [Accepted: 02/12/2025] [Indexed: 04/10/2025] Open
Abstract
The study of Inborn Errors of Immunity (IEIs) is critical for understanding the complex mechanisms of the human immune response to infectious diseases. Specific IEIs, characterized by selective susceptibility to certain pathogens, have enhanced our understanding of the key molecular pathways and cellular subsets involved in host defense against pathogens. These insights revealed that patients with anti-cytokine autoantibodies exhibit phenotypes similar to those with pathogenic mutations in genes encoding signaling molecules. This new disease concept is currently categorized as 'Phenocopies of IEI'. This category includes anti-cytokine autoantibodies targeting IL-17/IL-22, IFN-γ, IL-6, GM-CSF, and type I IFNs. Abundant anti-cytokine autoantibodies deplete corresponding cytokines, impair signaling pathways, and increase susceptibility to specific pathogens. We herein demonstrate the clinical and etiological significance of anti-cytokine autoantibodies in human immunity to pathogens. Insights from studies of rare IEIs underscore the pathological importance of cytokine-targeting autoantibodies. Simultaneously, the diverse clinical phenotype of patients with these autoantibodies suggests that the influences of cytokine dysfunction are broader than previously recognized. Furthermore, comprehensive studies prompted by the COVID-19 pandemic highlighted the substantial clinical impact of autoantibodies and their potential role in shaping the outcomes of infectious disease.
Collapse
Affiliation(s)
- Kosuke Noma
- Department of Pediatrics, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Takaki Asano
- Department of Pediatrics, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
- Department of Radiation Biophysics, Research Institute for Radiation Biology and Medicine, Hiroshima, Japan
| | - Maki Taniguchi
- Department of Pediatrics, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Kosuke Ashihara
- Department of Pediatrics, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Satoshi Okada
- Department of Pediatrics, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| |
Collapse
|
10
|
Moritz CP, Tholance Y, Boutahar N, Borowczyk C, Berger AE, Paul S, Antoine JC, Camdessanché JP. The antibody repertoire of autoimmune sensory neuronopathies targets pathways of the innate and adaptive immune system. An autoantigenomic approach. J Transl Autoimmun 2025; 10:100277. [PMID: 40026393 PMCID: PMC11870273 DOI: 10.1016/j.jtauto.2025.100277] [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: 08/30/2024] [Revised: 01/09/2025] [Accepted: 01/29/2025] [Indexed: 03/05/2025] Open
Abstract
Sensory neuronopathies (SNN) encompass diverse etiologies, with autoimmunity playing a major role through both cellular and humoral responses. To investigate the humoral autoantibody repertoire in autoimmune SNN, we conducted a retrospective cohort study using large Human Proteome-wide protein microarrays (HuProt 3.1, HuProt 4.0, ProtoArrays). We specifically analyzed immune system pathways targeted within the autoantigen repertoire (the autoantigenome). We included 131 participants: 44 patients with non-paraneoplastic autoimmune SNN (12 with anti-FGFR3 and/or anti-AGO antibodies), 8 with paraneoplastic SNN, and 79 controls. Findings were validated in an independent cohort of 16 SNN patients. Overrepresentation of immune-system-related proteins was assessed using the Reactome database, and serum levels of IFN-γ and IL-6 were measured with the Bio-Plex Pro™ Reagent Kit. Autoimmune SNN sera interact with significantly more immune system proteins than healthy controls (ProtoArrays: 271/863 vs. 14/863, HuProt: 112/1694 vs. 39/1694, both p < 0.0001). Overrepresentation was observed across all major immune sub-pathways, including innate and adaptive immune responses as well as cytokine signaling. Anti-FGFR3-positive SNN patients showed more frequent reactivity to immune system proteins than anti-FGFR3-negative ones. The independent SNN cohort validated the overrepresentation of targeted immune system pathways. Validation with dot blot and ELISA confirmed reactivity to TRIM21 and IL-6 and identified anti-IFN-γ-positive SNN patients. IFN-γ levels correlated weakly with levels of anti-IFN-γ antibodies (Pearson's r = 0.22, p = 0.03). We conclude that the antibody repertoire of autoimmune SNN targets pathways of the innate and adaptive immune system, potentially reflecting key disease-related immune pathways and highlighting the systemic role of immune dysregulation in SNN.
Collapse
Affiliation(s)
- Christian P. Moritz
- Synaptopathies and Autoantibodies (SynatAc) Team, Institut NeuroMyoGène, MELIS, INSERM U1314/CNRS UMR 5284, Université Claude Bernard Lyon 1, 43 Boulevard Du 11 Novembre 1918, Villeurbanne, 69622, France
- University Jean Monnet, 10, Rue de Marandière, 42270, Saint-Étienne, France
- Department of Neurology, University Hospital of Saint-Etienne, Avenue Albert Raimond, 42270, Saint-Etienne, France
| | - Yannick Tholance
- Synaptopathies and Autoantibodies (SynatAc) Team, Institut NeuroMyoGène, MELIS, INSERM U1314/CNRS UMR 5284, Université Claude Bernard Lyon 1, 43 Boulevard Du 11 Novembre 1918, Villeurbanne, 69622, France
- University Jean Monnet, 10, Rue de Marandière, 42270, Saint-Étienne, France
- Department of Biochemistry, University Hospital of Saint-Etienne, Avenue Albert Raimond, 42270, Saint-Etienne, France
| | - Nadia Boutahar
- Synaptopathies and Autoantibodies (SynatAc) Team, Institut NeuroMyoGène, MELIS, INSERM U1314/CNRS UMR 5284, Université Claude Bernard Lyon 1, 43 Boulevard Du 11 Novembre 1918, Villeurbanne, 69622, France
- University Jean Monnet, 10, Rue de Marandière, 42270, Saint-Étienne, France
- Department of Biochemistry, University Hospital of Saint-Etienne, Avenue Albert Raimond, 42270, Saint-Etienne, France
| | - Coralie Borowczyk
- Synaptopathies and Autoantibodies (SynatAc) Team, Institut NeuroMyoGène, MELIS, INSERM U1314/CNRS UMR 5284, Université Claude Bernard Lyon 1, 43 Boulevard Du 11 Novembre 1918, Villeurbanne, 69622, France
- Department of Biochemistry, University Hospital of Saint-Etienne, Avenue Albert Raimond, 42270, Saint-Etienne, France
| | - Anne-Emmanuelle Berger
- CIRI – Centre International de Recherche en Infectiologie, Team GIMAP, Univ Lyon, Université Claude Bernard Lyon 1, Inserm, U1111, CNRS, UMR530, CIC 1408 Vaccinology, Saint-Etienne, F42023, France
- Department of Immunology and Biotherapies, University Hospital of Saint-Etienne, Avenue Albert Raimond, 42270, Saint-Étienne, France
| | - Stéphane Paul
- CIRI – Centre International de Recherche en Infectiologie, Team GIMAP, Univ Lyon, Université Claude Bernard Lyon 1, Inserm, U1111, CNRS, UMR530, CIC 1408 Vaccinology, Saint-Etienne, F42023, France
- Department of Immunology and Biotherapies, University Hospital of Saint-Etienne, Avenue Albert Raimond, 42270, Saint-Étienne, France
| | - Jean-Christophe Antoine
- Synaptopathies and Autoantibodies (SynatAc) Team, Institut NeuroMyoGène, MELIS, INSERM U1314/CNRS UMR 5284, Université Claude Bernard Lyon 1, 43 Boulevard Du 11 Novembre 1918, Villeurbanne, 69622, France
- University Jean Monnet, 10, Rue de Marandière, 42270, Saint-Étienne, France
- Department of Neurology, University Hospital of Saint-Etienne, Avenue Albert Raimond, 42270, Saint-Etienne, France
| | - Jean-Philippe Camdessanché
- Synaptopathies and Autoantibodies (SynatAc) Team, Institut NeuroMyoGène, MELIS, INSERM U1314/CNRS UMR 5284, Université Claude Bernard Lyon 1, 43 Boulevard Du 11 Novembre 1918, Villeurbanne, 69622, France
- University Jean Monnet, 10, Rue de Marandière, 42270, Saint-Étienne, France
- Department of Neurology, University Hospital of Saint-Etienne, Avenue Albert Raimond, 42270, Saint-Etienne, France
| |
Collapse
|
11
|
Fekrvand S, Saleki K, Abolhassani H, Almasi-Hashiani A, Hakimelahi A, Zargarzadeh N, Yekaninejad MS, Rezaei N. COVID-19 infection in inborn errors of immunity and their phenocopies: a systematic review and meta-analysis. Infect Dis (Lond) 2025; 57:483-517. [PMID: 40178994 DOI: 10.1080/23744235.2025.2483339] [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: 10/13/2024] [Revised: 02/09/2025] [Accepted: 02/23/2025] [Indexed: 04/05/2025] Open
Abstract
BACKGROUND Inborn errors of immunity (IEI) are congenital disorders of the immune system. Due to impaired immune system, they are at a higher risk to develop a more severe COVID-19 course compared to general population. OBJECTIVES Herein, we aimed to systematically review various aspects of IEI patients infected with SARS-CoV-2. Moreover, we performed a meta-analysis to determine the frequency of COVID-19 in patients with different IEI. METHODS Embase, Web of Science, PubMed, and Scopus were searched introducing terms related to IEI and COVID-19. RESULTS 3646 IEI cases with a history of COVID-19 infection were enrolled. The majority of patients had critical infections (1013 cases, 27.8%). The highest frequency of critical and severe cases was observed in phenocopies of IEI (95.2%), defects in intrinsic and innate immunity (69.4%) and immune dysregulation (23.9%). 446 cases (12.2%) succumbed to the disease and the highest mortality was observed in IEI phenocopies (34.6%). COVID-19 frequency in immunodeficient patients was 11.9% (95% CI: 8.3 to 15.5%) with innate immunodeficiency having the highest COVID-19 frequency [34.1% (12.1 to 56.0%)]. COVID-19 case fatality rate among IEI patients was estimated as 5.4% (95% CI: 3.5-8.3%, n = 8 studies, I2 = 17.5%). CONCLUSION IEI with underlying defects in specific branches of the immune system responding to RNA virus infection experience a higher frequency and mortality of COVID-19 infection. Increasing awareness about these entities and underlying genetic defects, adherence to prophylactic strategies and allocating more clinical attention to these patients could lead to a decrease in COVID-19 frequency and mortality in these patients.
Collapse
Affiliation(s)
- Saba Fekrvand
- Research Center for Immunodeficiencies, Pediatrics Center of Excellence, Children's Medical Center, Tehran University of Medical Sciences, Tehran, Iran
- Primary Immunodeficiency Diseases Network (PIDNet), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Kiarash Saleki
- Research Center for Immunodeficiencies, Pediatrics Center of Excellence, Children's Medical Center, Tehran University of Medical Sciences, Tehran, Iran
- Primary Immunodeficiency Diseases Network (PIDNet), Universal Scientific Education and Research Network (USERN), Tehran, Iran
- Student Research Committee, Babol University of Medical Sciences, Babol, Iran
| | - Hassan Abolhassani
- Research Center for Immunodeficiencies, Pediatrics Center of Excellence, Children's Medical Center, Tehran University of Medical Sciences, Tehran, Iran
- Division of Clinical Immunology, Department of Biosciences and Nutrition, KarolinskaInstitutet, Karolinska University Hospital, Stockholm, Sweden
| | - Amir Almasi-Hashiani
- Department of Epidemiology, School of Health, Arak University of Medical Sciences, Arak, Iran
| | - Ali Hakimelahi
- Research Center for Immunodeficiencies, Pediatrics Center of Excellence, Children's Medical Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Nikan Zargarzadeh
- School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Mir Saeed Yekaninejad
- Department of Epidemiology and Biostatistics, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| | - Nima Rezaei
- Research Center for Immunodeficiencies, Pediatrics Center of Excellence, Children's Medical Center, Tehran University of Medical Sciences, Tehran, Iran
- Primary Immunodeficiency Diseases Network (PIDNet), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| |
Collapse
|
12
|
Kasem Ali Sliman R, Cohen H, Shehadeh S, Batcir R, Alter YE, Cohen K, Koren I, Halabi I, Sliman H, Saied MH. Pediatric autoimmune diseases in the light of COVID-19 pandemic, A retrospective observational big data study. J Transl Autoimmun 2025; 10:100281. [PMID: 40162434 PMCID: PMC11951201 DOI: 10.1016/j.jtauto.2025.100281] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2024] [Revised: 02/18/2025] [Accepted: 03/01/2025] [Indexed: 04/02/2025] Open
Abstract
Background The COVID-19 pandemic has raised concerns about potential links between SARS-CoV-2 infection and autoimmune diseases. This study investigated changes in the incidence rate (IR) of autoimmune diseases among children following the pandemic's onset. Methods A retrospective cross-sectional study analyzed data from Clalit Health Services, Israel's largest healthcare provider, examining the IR of different autoimmune diseases in children aged 0-18. The study compared pre-pandemic (2019) with pandemic/post-pandemic periods (2020-2023), encompassing a cohort of over 1.5 million children. Results Significant IR increases were observed across multiple autoimmune diseases. Rheumatic diseases (Juvenile Idiopathic Arthritis, Systemic Lupus Erythematosus, Henoch Schoenlein Purpura (HSP)) showed consistent increases, with HSP demonstrating the most pronounced trend. Endocrine disorders exhibited diverse patterns, with autoimmune thyroid diseases and Type 1 diabetes showing overall increases, while diabetic ketoacidosis exhibited an initial spike followed by a decline. Gastrointestinal diseases displayed heterogeneous patterns; Celiac disease and Ulcerative colitis showed general increases, Crohn's disease showed a downward trend, and autoimmune hepatitis exhibited an initial significant decrease followed by a significant increase. Dermatological conditions, including Psoriasis and Vitiligo, demonstrated consistent elevations throughout 2020-2023. Immune Thrombocytopenia Purpura showed initial decreases followed by significant increases in 2022-2023. Conclusions This comprehensive analysis reveals significant changes in pediatric autoimmune disease incidence following the COVID-19 pandemic, suggesting potential associations between SARS-CoV-2 infection and autoimmune dysregulation. The diverse patterns observed across different conditions highlight the complex interplay between viral infection and autoimmunity, emphasizing the need for continued surveillance and investigation of long-term immunological consequences of COVID-19 in pediatric populations.
Collapse
Affiliation(s)
- Rim Kasem Ali Sliman
- Technion Israel Institute of Technology, Rappaport Faculty of Medicine, Haifa 3109601, Israel
- Department of Pediatrics, Clalit Health Care Organization, Carmel Medical Center, Haifa, Israel
| | - Hilla Cohen
- Clalit Health Care Organization, Carmel Medical Center, Haifa, Israel
| | - Shereen Shehadeh
- Technion Israel Institute of Technology, Rappaport Faculty of Medicine, Haifa 3109601, Israel
- Department of Pediatrics, Clalit Health Care Organization, Carmel Medical Center, Haifa, Israel
- Infectious Disease Unit, Carmel Medical Center, Haifa, Israel
| | - Reut Batcir
- Technion Israel Institute of Technology, Rappaport Faculty of Medicine, Haifa 3109601, Israel
- Department of Pediatrics, Clalit Health Care Organization, Carmel Medical Center, Haifa, Israel
- Pediatric Gastroenterology Unit, Carmel Medical Center, Haifa, Israel
| | - Yigal Elenberg Alter
- Technion Israel Institute of Technology, Rappaport Faculty of Medicine, Haifa 3109601, Israel
- Department of Pediatrics, Clalit Health Care Organization, Carmel Medical Center, Haifa, Israel
- Pediatric Gastroenterology Unit, Carmel Medical Center, Haifa, Israel
| | - Keren Cohen
- Technion Israel Institute of Technology, Rappaport Faculty of Medicine, Haifa 3109601, Israel
- Department of Pediatrics, Clalit Health Care Organization, Carmel Medical Center, Haifa, Israel
- Pediatric Endocrine Unit, Carmel Medical Center, Haifa, Israel
| | - Ilana Koren
- Technion Israel Institute of Technology, Rappaport Faculty of Medicine, Haifa 3109601, Israel
- Department of Pediatrics, Clalit Health Care Organization, Carmel Medical Center, Haifa, Israel
- Pediatric Endocrine Unit, Carmel Medical Center, Haifa, Israel
| | - Inbal Halabi
- Technion Israel Institute of Technology, Rappaport Faculty of Medicine, Haifa 3109601, Israel
- Department of Pediatrics, Clalit Health Care Organization, Carmel Medical Center, Haifa, Israel
- Pediatric Endocrine Unit, Carmel Medical Center, Haifa, Israel
| | - Hussein Sliman
- Technion Israel Institute of Technology, Rappaport Faculty of Medicine, Haifa 3109601, Israel
- Department of Cardiology, Carmel Medical Center, Heart Center, Haifa, Israel
| | - Mohamad Hamad Saied
- Technion Israel Institute of Technology, Rappaport Faculty of Medicine, Haifa 3109601, Israel
- Department of Pediatrics, Clalit Health Care Organization, Carmel Medical Center, Haifa, Israel
- Department of Pediatric Immunology and Infectious Diseases, Wilhelmina Children's Hospital/University Medical Center, Utrecht, the Netherlands
| |
Collapse
|
13
|
Epstein-Shuman A, Zhu X, Hunt JH, Fernandez RE, Rozek GM, Redd AD, Gotthold ZA, Quiros G, Galiwango RM, Kigozi G, Caturegli P, Ssekubugu R, Grabowski MK, Chang LW, Reynolds SJ, Laeyendecker O. Autoantibodies to interferon alpha, nuclear antigens, cardiolipin, and beta 2 glycoprotein 1 in a Ugandan cohort and their relation to SARS-CoV-2 infection. J Infect Public Health 2025; 18:102722. [PMID: 40086141 DOI: 10.1016/j.jiph.2025.102722] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2024] [Revised: 02/04/2025] [Accepted: 02/20/2025] [Indexed: 03/16/2025] Open
Abstract
OBJECTIVE Autoantibodies (AAbs) to interferon alpha, nuclear antigens, cardiolipin, and beta 2 glycoprotein 1, have been associated with COVID-19 severity. Despite relatively low COVID-19 morbidity and mortality in East and Central Africa, AAb prevalence in these populations remain understudied. METHODS We evaluated AAb seroprevalence in 155 Ugandans, aged 40-50, using paired samples collected before and after the onset of the COVID-19 pandemic. Among these, 117 had serological evidence of SARS-CoV-2 infection, and 38 did not. To assess the effect of SARS-CoV-2 infection on AAb prevalence, we: 1) longitudinally compared AAb prevalence before and after evidence of infection, and 2) cross-sectionally compared AAb prevalence between those with and without infection evidence at both timepoints. Associations between AAbs and health characteristics were also explored. RESULTS There was no difference in AAb prevalence between individuals with and without evidence of infection, nor any longitudinal change after evidence of infection. However, we observed a higher-than-expected prevalence anti-beta 2 glycoprotein 1. Additionally, anti-cardiolipin was significantly associated with reported hypertension. CONCLUSIONS Our findings contribute to the limited literature on AAb prevalence in East Africa and suggest that SARS-CoV-2 does not induce these AAbs.
Collapse
Affiliation(s)
- Adam Epstein-Shuman
- Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Xianming Zhu
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Joanne H Hunt
- Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Reinaldo E Fernandez
- Department of Medicine, Division of Infectious Diseases, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Gracie M Rozek
- Department of Medicine, Division of Infectious Diseases, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Andrew D Redd
- Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA; Department of Medicine, Division of Infectious Diseases, Johns Hopkins University School of Medicine, Baltimore, MD, USA; Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Cape Town, South Africa
| | - Zoe A Gotthold
- Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Gabriel Quiros
- Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | | | | | - Patrizio Caturegli
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | | | - Mary K Grabowski
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, USA; Rakai Health Sciences Program, Kalisizo, Uganda
| | - Larry W Chang
- Department of Medicine, Division of Infectious Diseases, Johns Hopkins University School of Medicine, Baltimore, MD, USA; Rakai Health Sciences Program, Kalisizo, Uganda
| | - Steven J Reynolds
- Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA; Department of Medicine, Division of Infectious Diseases, Johns Hopkins University School of Medicine, Baltimore, MD, USA; Rakai Health Sciences Program, Kalisizo, Uganda
| | - Oliver Laeyendecker
- Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA; Department of Medicine, Division of Infectious Diseases, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| |
Collapse
|
14
|
Rosa-Baez C, Borrego-Yaniz G, Rodriguez-Martin I, Kerick M, Acosta-Herrera M, Martín J, Ortiz-Fernández L. Cross-trait GWAS in COVID-19 and systemic sclerosis reveals novel genes implicated in fibrotic and inflammation pathways. Rheumatology (Oxford) 2025; 64:4022-4031. [PMID: 39878951 DOI: 10.1093/rheumatology/keaf028] [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: 07/17/2024] [Revised: 12/17/2024] [Accepted: 01/06/2025] [Indexed: 01/31/2025] Open
Abstract
OBJECTIVES Coronavirus disease 2019 (COVID-19) and SSc share multiple similarities in their clinical manifestations, alterations in immune response and therapeutic options. These resemblances have also been identified in other immune-mediated inflammatory diseases where a common genetic component has been found. Thus, we decided to evaluate for the first time this shared genetic architecture with SSc. METHODS For this study, we retrieved genomic data from two European-ancestry cohorts: 2 597 856 individuals from The COVID-19 Host Genetics Initiative consortium, and 26 679 individuals from the largest genomic scan in SSc. We performed a cross-trait meta-analyses including >9.3 million single nucleotide polymorphisms. Finally, we conducted functional annotation to prioritize potential causal genes and performed drug repurposing analysis. RESULTS Our results revealed a total of 19 non-HLA pleiotropic loci, including 2 novel associations for both conditions (BMP1 and PPARG) and 12 emerging as new shared loci. Functional annotation of these regions underscored their potential regulatory role and identified potential causal genes, many of which are implicated in fibrotic and inflammatory pathways. Remarkably, we observed an antagonistic pleiotropy model of the IFN signalling between COVID-19 and SSc, including the well-known TYK2 P1104A missense variant, showing a protective effect for SSc while being a risk factor for COVID-19, along with two additional novel pleiotropic associations (IRF8 and SENP7). Finally, our findings provide new therapeutic options that could potentially benefit both conditions. CONCLUSION Our study confirms the genetic resemblance between susceptibility to and severity of COVID-19 and SSc, revealing a novel common genetic contribution affecting fibrotic and immune pathways.
Collapse
Affiliation(s)
- Carlos Rosa-Baez
- Department of Cell Biology and Immunology, Institute of Parasitology and Biomedicine López-Neyra, CSIC, Granada, Spain
| | - Gonzalo Borrego-Yaniz
- Department of Cell Biology and Immunology, Institute of Parasitology and Biomedicine López-Neyra, CSIC, Granada, Spain
| | - Inmaculada Rodriguez-Martin
- Department of Cell Biology and Immunology, Institute of Parasitology and Biomedicine López-Neyra, CSIC, Granada, Spain
| | - Martin Kerick
- Department of Cell Biology and Immunology, Institute of Parasitology and Biomedicine López-Neyra, CSIC, Granada, Spain
| | - Marialbert Acosta-Herrera
- Department of Cell Biology and Immunology, Institute of Parasitology and Biomedicine López-Neyra, CSIC, Granada, Spain
- Systemic Autoimmune Disease Unit, Hospital Clínico San Cecilio, Instituto de Investigación Biosanitaria Ibs. GRANADA, Granada, Spain
| | - Javier Martín
- Department of Cell Biology and Immunology, Institute of Parasitology and Biomedicine López-Neyra, CSIC, Granada, Spain
| | - Lourdes Ortiz-Fernández
- Department of Cell Biology and Immunology, Institute of Parasitology and Biomedicine López-Neyra, CSIC, Granada, Spain
| |
Collapse
|
15
|
Nehar-Belaid D, Mejías A, Xu Z, Marches R, Yerrabelli R, Chen G, Mertz S, Ye F, Sánchez PJ, Tsang JS, Aydillo T, Miorin L, Cupic A, García-Sastre A, Ucar D, Banchereau JF, Pascual V, Ramilo O. SARS-CoV-2 induced immune perturbations in infants vary with disease severity and differ from adults' responses. Nat Commun 2025; 16:4562. [PMID: 40379618 PMCID: PMC12084365 DOI: 10.1038/s41467-025-59411-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2024] [Accepted: 04/22/2025] [Indexed: 05/19/2025] Open
Abstract
Differences in immune profiles of children and adults with COVID-19 have been previously described. However, no systematic studies have been reported from infants hospitalized with severe disease. We applied a multidimensional approach to decipher the immune responses of SARS-CoV-2 infected infants (n = 26; 10 subacute, 11 moderate and 5 severe disease; median age = 1.6 months) and matched controls (n = 14; median age = 2 months). Single cell (scRNA-seq) profiling of PBMCs revealed substantial alterations in cell composition in SARS-CoV-2 infected infants; with most cell-types switching to an interferon-stimulated gene (ISGhi) state including: (i) CD14+ monocytes co-expressing ISGs and inflammasome-related molecules, (ii) ISGhi naive CD4+ T cells, (iii) ISGhi proliferating cytotoxic CD8+ T cells, and (iv) ISGhi naive and transitional B cells. We observe increased serum concentrations of both interferons and inflammatory cytokines in infected infants. Antibody responses to SARS-CoV-2 are also consistently detected in the absence of anti-IFN autoantibodies. Compared with infected adults, infants display a similar ISG signature in monocytes but a markedly enhanced ISG signature in T and B cells. These findings provide insights into the distinct immune responses to SARS-CoV-2 in the first year of life and underscore the importance of further defining the unique features of early life immunity.
Collapse
Affiliation(s)
| | - Asunción Mejías
- Department of Pediatrics, Nationwide Children's Hospital and The Ohio State University College of Medicine, Columbus, OH, USA
- Center for Vaccines and Immunity, Abigail Wexner Research Institute at Nationwide Children's Hospital, Columbus, OH, 43205, USA
- Department of Infectious Diseases, St. Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN, 38105, USA
| | - Zhaohui Xu
- Center for Vaccines and Immunity, Abigail Wexner Research Institute at Nationwide Children's Hospital, Columbus, OH, 43205, USA
- Department of Infectious Diseases, St. Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN, 38105, USA
| | - Radu Marches
- The Jackson Laboratory for Genomic Medicine, Farmington, CT, 06032, USA
| | - Rushil Yerrabelli
- The Jackson Laboratory for Genomic Medicine, Farmington, CT, 06032, USA
| | - Guo Chen
- The Jackson Laboratory for Genomic Medicine, Farmington, CT, 06032, USA
| | - Sara Mertz
- Center for Vaccines and Immunity, Abigail Wexner Research Institute at Nationwide Children's Hospital, Columbus, OH, 43205, USA
| | - Fang Ye
- Center for Vaccines and Immunity, Abigail Wexner Research Institute at Nationwide Children's Hospital, Columbus, OH, 43205, USA
| | - Pablo J Sánchez
- Department of Pediatrics, Division of Neonatology and Center for Perinatal Research, Ohio Perinatal Research Network, Abigail Wexner Research Institute at Nationwide Children's Hospital, Columbus, OH, USA
| | - John S Tsang
- Center for Systems and Engineering Immunology, Departments of Immunobiology and Biomedical Engineering, Yale University, New Haven, CT, 06520, USA
- Department of Biomedical Engineering, Yale University, New Haven, CT, USA
- Chan Zuckerberg Biohub NY, New Haven, CT, USA
| | - Teresa Aydillo
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY, 10029, USA
- Global Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY, 10029, USA
| | - Lisa Miorin
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY, 10029, USA
- Global Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY, 10029, USA
| | - Anastasija Cupic
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY, 10029, USA
- Global Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY, 10029, USA
| | - Adolfo García-Sastre
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY, 10029, USA
- Global Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY, 10029, USA
- Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Department of Medicine, Division of Infectious Diseases, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY, 10029, USA
- The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY, 10029, USA
- Department of Pathology, Molecular and Cell-Based Medicine, One Gustave L. Levy Place, New York, NY, 10029, USA
- Icahn Genomics Institute, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY, 10029, USA
| | - Duygu Ucar
- The Jackson Laboratory for Genomic Medicine, Farmington, CT, 06032, USA
| | - Jacques F Banchereau
- The Jackson Laboratory for Genomic Medicine, Farmington, CT, 06032, USA.
- Immunoledge LLC, Montclair, NJ, USA.
| | - Virginia Pascual
- Drukier Institute for Children's Health and Department of Pediatrics, Weill Cornell Medicine, New York, NY, USA.
| | - Octavio Ramilo
- Department of Pediatrics, Nationwide Children's Hospital and The Ohio State University College of Medicine, Columbus, OH, USA.
- Center for Vaccines and Immunity, Abigail Wexner Research Institute at Nationwide Children's Hospital, Columbus, OH, 43205, USA.
- Department of Infectious Diseases, St. Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN, 38105, USA.
| |
Collapse
|
16
|
Márquez-Bandala AH, Gutierrez-Xicotencatl L, Esquivel-Guadarrama F. Pathogenesis Induced by Influenza Virus Infection: Role of the Early Events of the Infection and the Innate Immune Response. Viruses 2025; 17:694. [PMID: 40431705 PMCID: PMC12115608 DOI: 10.3390/v17050694] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2025] [Revised: 04/26/2025] [Accepted: 05/08/2025] [Indexed: 05/29/2025] Open
Abstract
Infections by influenza A virus (IAV) are a significant cause of global mortality. The pathogenesis of the infection is usually studied in terms of direct viral-induced damage or the overreactive immune response that continues after the virus is cleared. However, factors such as the initial infectious dose, the early response after infection in different cell types, and the presence of autoantibodies for relevant antiviral cytokines like type I IFNs seem to influence the course of the infection and lead to fatal outcomes. In this article, we address the current knowledge about the early events during influenza virus infection, which are important for their participation in influenza-derived pathogenesis.
Collapse
Affiliation(s)
- Alicia Helena Márquez-Bandala
- Instituto de Biotecnología, Universidad Nacional Autónoma de Mexico, Cuernavaca C.P. 62209, Morelos, Mexico;
- Laboratorio de Inmunología Viral, Facultad de Medicina, Universidad Autónoma del Estado de Morelos, Cuernava C.P. 62350, Morelos, Mexico
| | - Lourdes Gutierrez-Xicotencatl
- Laboratorio de Virus y Cáncer, Centro de Investigacion Sobre Enfermedades Infecciosas, Instituto Nacional de Salud Publica, Cuernavaca C.P. 62209, Morelos, Mexico;
| | - Fernando Esquivel-Guadarrama
- Laboratorio de Inmunología Viral, Facultad de Medicina, Universidad Autónoma del Estado de Morelos, Cuernava C.P. 62350, Morelos, Mexico
| |
Collapse
|
17
|
Wang J, Ji X, Yang C, Xu J. Susceptibility from the immunological perspective of COVID-19-associated pulmonary aspergillosis: A literature review. Medicine (Baltimore) 2025; 104:e42363. [PMID: 40355215 PMCID: PMC12073940 DOI: 10.1097/md.0000000000042363] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/20/2024] [Accepted: 04/18/2025] [Indexed: 05/14/2025] Open
Abstract
The incidence rate of COVID-19-associated pulmonary aspergillosis (CAPA) is rising. However, the pathogenesis of CAPA remains unclear. Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) infection disrupts pathways related to type I interferon and Toll-like receptors, key components in innate immunity, thereby elevating the incidence of CAPA. Additionally, SARS-CoV-2 infection results in T and B cell functional deficiencies or exhaustion within adaptive immunity, weakening the defense against invasive Aspergillus. Furthermore, SARS-CoV-2 infection enhances the replication of cytomegalovirus and alters the gut microbiota, factors that may aid in diagnosing CAPA. Immunosuppressive therapy in COVID-19 patients is also believed to heighten the risk of invasive aspergillosis. Therefore, this review, examines the immune response to SARS-CoV-2 infection combined with invasive aspergillosis, and explores the pathogenesis and susceptibility factors of CAPA. We propose that variations in an individual's immune response significantly determine susceptibility to CAPA. The aim of this paper is to deepen clinical understanding of CAPA's pathogenesis, thereby aiding in mitigating susceptibility risk and advancing novel treatment approaches.
Collapse
Affiliation(s)
- Jiayin Wang
- Department of Laboratory, The First Hospital of Jilin University, Changchun, China
| | - Xufeng Ji
- Department of Laboratory, The First Hospital of Jilin University, Changchun, China
| | - Chun Yang
- Department of Laboratory, The First Hospital of Jilin University, Changchun, China
| | - Jiancheng Xu
- Department of Laboratory, The First Hospital of Jilin University, Changchun, China
- Center of Infectious Diseases and Pathogen Biology, The First Hospital of Jilin University, Changchun, China
| |
Collapse
|
18
|
Shujath J. Beyond Traditional Publishing: Social Media as a Catalyst for Biomedical Research Dissemination and Collaboration. J Interferon Cytokine Res 2025. [PMID: 40329892 DOI: 10.1089/jir.2025.0074] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/08/2025] Open
Abstract
The expansion of social media has fundamentally transformed biomedical research dissemination and collaboration, particularly within the interferon and cytokine research community. This paper explores recent trends (2024-2025) that have amplified the role of platforms such as Twitter (now "X"), LinkedIn, Mastodon, Threads, and Bluesky. These tools have facilitated rapid knowledge exchange, democratized access to scientific discourse, enabled diverse voices to participate meaningfully, and fostered cross-disciplinary and global collaborations. Additionally, the integration of preprint repositories like bioRxiv and medRxiv, along with the evolution of open access publishing, further accelerates the accessibility and immediacy of scientific communication. Despite evident benefits, the rapid dissemination facilitated by social media also poses ethical challenges, including concerns about misinformation, premature dissemination of preliminary data, and privacy considerations. Practical strategies for researchers and institutions to effectively navigate these platforms responsibly are presented, aiming to optimize the impact of social media on scientific discovery and public engagement.
Collapse
Affiliation(s)
- Jaleel Shujath
- University of the District of Columbia, Washington, District of Columbia, USA
| |
Collapse
|
19
|
Hörstke NV, Vogl T. Deciphering the autoreactome: Massively parallelized methods for autoantibody detection in humans. J Immunol Methods 2025; 541:113876. [PMID: 40339788 DOI: 10.1016/j.jim.2025.113876] [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: 11/18/2024] [Revised: 05/05/2025] [Accepted: 05/05/2025] [Indexed: 05/10/2025]
Abstract
Autoantibodies have a substantial impact on human health ranging from autoimmune diseases to cancer diagnostics. Knowledge of the antigens recognized can allow for more accurate diagnostics, a better understanding of pathogeneses and thus improved prevention, as well as laying the foundation for the development of new therapies. A critical step to acquire this knowledge is to detect the exact self-antigens targeted by autoantibodies out of the pool of 20,000 human proteins against which reactivities could be observed. Here, we review established and emerging methods for highly parallelized autoantigen detection such as human proteome microarrays, serological identification of antigens by screening of cDNA expression libraries (SEREX), serological proteome analysis (SERPA), phage display immunoprecipitation sequencing (PhIP-Seq), parallel analysis of translated ORFs (PLATO), and rapid extracellular antigen profiling (REAP). We highlight advantages and limitations of these methods, aiming to give a guideline to choose the appropriate method for a certain application.
Collapse
Affiliation(s)
- Nicolai V Hörstke
- Center for Cancer Research, Medical University of Vienna, Borschkegasse 8a, 1090 Vienna, Austria
| | - Thomas Vogl
- Center for Cancer Research, Medical University of Vienna, Borschkegasse 8a, 1090 Vienna, Austria.
| |
Collapse
|
20
|
Broomfield BJ, Tan CW, Qin RZ, Abberger H, Duckworth BC, Alvarado C, Dalit L, Lee CL, Shandre Mugan R, Mazrad ZA, Muramatsu H, Mackiewicz L, Williams BE, Chen J, Takanashi A, Fabb S, Pellegrini M, Rogers KL, Moon WJ, Pouton CW, Davis MJ, Nutt SL, Pardi N, Wimmer VC, Groom JR. Transient inhibition of type I interferon enhances CD8+ T cell stemness and vaccine protection. J Exp Med 2025; 222:e20241148. [PMID: 40062995 PMCID: PMC11893171 DOI: 10.1084/jem.20241148] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2024] [Revised: 11/25/2024] [Accepted: 02/04/2025] [Indexed: 03/14/2025] Open
Abstract
Developing vaccines that promote CD8+ T cell memory is a challenge for infectious disease and cancer immunotherapy. TCF-1+ stem cell-like memory CD8+ T (TSCM) cells are important determinants of long-lived memory. Yet, the developmental requirements for TSCM cell formation are unclear. Here, we identify the temporal window for type I interferon receptor (IFNAR) blockade to drive TSCM cell generation following viral infection and mRNA-lipid nanoparticle vaccination. We reveal a reversible developmental trajectory where transcriptionally distinct TSCM cells emerged from a transitional precursor of exhausted T cellular state concomitant with viral clearance. TSCM cell differentiation correlated with T cell retention within the lymph node paracortex due to disrupted CXCR3 chemokine gradient formation. These effects were linked to increased antigen load and a counterintuitive increase in IFNγ, which controlled cell location. Vaccination with the IFNAR blockade promoted TSCM cell differentiation and enhanced protection against chronic infection. These findings propose an approach to vaccine design whereby modulation of inflammation promotes memory formation and function.
Collapse
Affiliation(s)
- Benjamin J. Broomfield
- Walter and Eliza Hall Institute of Medical Research, Parkville, Australia
- Department of Medical Biology, The University of Melbourne, Parkville, Australia
| | - Chin Wee Tan
- Walter and Eliza Hall Institute of Medical Research, Parkville, Australia
- Department of Medical Biology, The University of Melbourne, Parkville, Australia
- Frazer Institute, Faculty of Medicine, The University of Queensland, Brisbane, Australia
| | - Raymond Z. Qin
- Walter and Eliza Hall Institute of Medical Research, Parkville, Australia
- Department of Medical Biology, The University of Melbourne, Parkville, Australia
| | - Hanna Abberger
- Walter and Eliza Hall Institute of Medical Research, Parkville, Australia
- Department of Medical Biology, The University of Melbourne, Parkville, Australia
| | - Brigette C. Duckworth
- Walter and Eliza Hall Institute of Medical Research, Parkville, Australia
- Department of Medical Biology, The University of Melbourne, Parkville, Australia
| | - Carolina Alvarado
- Walter and Eliza Hall Institute of Medical Research, Parkville, Australia
| | - Lennard Dalit
- Walter and Eliza Hall Institute of Medical Research, Parkville, Australia
- Department of Medical Biology, The University of Melbourne, Parkville, Australia
| | - Chee Leng Lee
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Australia
| | - Rekha Shandre Mugan
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Australia
| | - Zihnil A.I. Mazrad
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Australia
| | - Hiromi Muramatsu
- Department of Microbiology, Perelman School of Medicine, Philadelphia, PA, USA
| | - Liana Mackiewicz
- Walter and Eliza Hall Institute of Medical Research, Parkville, Australia
| | - Bailey E. Williams
- Walter and Eliza Hall Institute of Medical Research, Parkville, Australia
| | - Jinjin Chen
- Walter and Eliza Hall Institute of Medical Research, Parkville, Australia
- Department of Medical Biology, The University of Melbourne, Parkville, Australia
| | - Asuka Takanashi
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Australia
| | - Stewart Fabb
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Australia
| | - Marc Pellegrini
- Centenary Institute of Cancer Medicine and Cell Biology, Camperdown, Australia
| | - Kelly L. Rogers
- Walter and Eliza Hall Institute of Medical Research, Parkville, Australia
- Department of Medical Biology, The University of Melbourne, Parkville, Australia
| | | | - Colin W. Pouton
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Australia
| | - Melissa J. Davis
- Walter and Eliza Hall Institute of Medical Research, Parkville, Australia
- Frazer Institute, Faculty of Medicine, The University of Queensland, Brisbane, Australia
- School of Biomedicine, Faculty of Health and Medical Sciences, The University of Adelaide, Adelaide, Australia
| | - Stephen L. Nutt
- Walter and Eliza Hall Institute of Medical Research, Parkville, Australia
- Department of Medical Biology, The University of Melbourne, Parkville, Australia
| | - Norbert Pardi
- Department of Microbiology, Perelman School of Medicine, Philadelphia, PA, USA
| | - Verena C. Wimmer
- Walter and Eliza Hall Institute of Medical Research, Parkville, Australia
- Department of Medical Biology, The University of Melbourne, Parkville, Australia
| | - Joanna R. Groom
- Walter and Eliza Hall Institute of Medical Research, Parkville, Australia
- Department of Medical Biology, The University of Melbourne, Parkville, Australia
| |
Collapse
|
21
|
Brinkmann M, Traby L, Kussmann M, Weiss-Tessbach M, Buchtele N, Staudinger T, Gaidoschik E, Perkmann T, Haslacher H, Ratzinger F, Pickl WF, El-Gedawi K, Feichter M, Gelpi E, Höftberger R, Quehenberger P, Marculescu R, Mrak D, Kastrati K, Lechner-Radner H, Sieghart D, Aletaha D, Winkler S, Bonelli M, Göschl L. Autoantibody development is associated with clinical severity of COVID-19: A cohort study. Clin Immunol 2025; 274:110471. [PMID: 40044018 DOI: 10.1016/j.clim.2025.110471] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2024] [Revised: 02/18/2025] [Accepted: 03/01/2025] [Indexed: 03/23/2025]
Abstract
Viral infections, including respiratory diseases such as Coronavirus disease 2019 (COVID-19), are hypothesized to contribute to the onset of autoimmune disorders. Although elevated levels of autoantibodies have been observed following COVID-19, the role of specific autoantibodies linked to autoimmune diseases and their correlation with disease severity remains poorly defined. In this study, we used a comprehensive autoantibody panel to assess the autoantibody production across different cohorts of COVID-19 patients, categorized by disease severity. We also compared patients with severe COVID-19 to a control group with other severe, non-COVID-related diseases. Our findings indicate that the severity of COVID-19 corresponds to the overall production of specific autoantibodies, which are particularly associated with COVID-19. This association might predispose to an increased risk for the development of autoimmune conditions after a severe course of COVID-19.
Collapse
Affiliation(s)
- Marie Brinkmann
- Division of Rheumatology, Department of Internal Medicine III, Medical University of Vienna, Vienna, Austria; Comprehensive Center for Inflammation and Immunity (CCII), Medical University of Vienna, Vienna, Austria
| | - Ludwig Traby
- Clinical Division of Infectious Diseases and Tropical Medicine, Department of Medicine I, Medical University of Vienna, Vienna, Austria
| | - Manuel Kussmann
- Clinical Division of Infectious Diseases and Tropical Medicine, Department of Medicine I, Medical University of Vienna, Vienna, Austria
| | - Matthias Weiss-Tessbach
- Clinical Division of Infectious Diseases and Tropical Medicine, Department of Medicine I, Medical University of Vienna, Vienna, Austria; Department of Clinical Pharmacology, Medical University of Vienna, Vienna, Austria
| | - Nina Buchtele
- Intensive Care Unit 13i2, Department of Medicine I, Medical University of Vienna, Vienna, Austria
| | - Thomas Staudinger
- Intensive Care Unit 13i2, Department of Medicine I, Medical University of Vienna, Vienna, Austria
| | - Elias Gaidoschik
- Division of Rheumatology, Department of Internal Medicine III, Medical University of Vienna, Vienna, Austria; Comprehensive Center for Inflammation and Immunity (CCII), Medical University of Vienna, Vienna, Austria
| | - Thomas Perkmann
- Department of Laboratory Medicine, Medical University of Vienna, Vienna, Austria
| | - Helmuth Haslacher
- Department of Laboratory Medicine, Medical University of Vienna, Vienna, Austria
| | - Franz Ratzinger
- Department of Laboratory Medicine, Medical University of Vienna, Vienna, Austria
| | - Winfried F Pickl
- Comprehensive Center for Inflammation and Immunity (CCII), Medical University of Vienna, Vienna, Austria; Center for Pathophysiology, Infectiology and Immunology, Institute of Immunology, Medical University of Vienna, Vienna, Austria.; Karl Landsteiner University of Health Sciences, 3500 Krems, Austria
| | - Karim El-Gedawi
- Center for Pathophysiology, Infectiology and Immunology, Institute of Immunology, Medical University of Vienna, Vienna, Austria
| | - Melanie Feichter
- Center for Pathophysiology, Infectiology and Immunology, Institute of Immunology, Medical University of Vienna, Vienna, Austria
| | - Ellen Gelpi
- Division of Neuropathology and Neurochemistry, Department of Neurology, Medical University of Vienna, Vienna, Austria
| | - Romana Höftberger
- Division of Neuropathology and Neurochemistry, Department of Neurology, Medical University of Vienna, Vienna, Austria
| | - Peter Quehenberger
- Department of Laboratory Medicine, Medical University of Vienna, Vienna, Austria
| | - Rodrig Marculescu
- Department of Laboratory Medicine, Medical University of Vienna, Vienna, Austria
| | - Daniel Mrak
- Division of Rheumatology, Department of Internal Medicine III, Medical University of Vienna, Vienna, Austria
| | - Kastriot Kastrati
- Division of Rheumatology, Department of Internal Medicine III, Medical University of Vienna, Vienna, Austria
| | - Helga Lechner-Radner
- Division of Rheumatology, Department of Internal Medicine III, Medical University of Vienna, Vienna, Austria
| | - Daniela Sieghart
- Division of Rheumatology, Department of Internal Medicine III, Medical University of Vienna, Vienna, Austria
| | - Daniel Aletaha
- Division of Rheumatology, Department of Internal Medicine III, Medical University of Vienna, Vienna, Austria; Comprehensive Center for Inflammation and Immunity (CCII), Medical University of Vienna, Vienna, Austria
| | - Stefan Winkler
- Clinical Division of Infectious Diseases and Tropical Medicine, Department of Medicine I, Medical University of Vienna, Vienna, Austria
| | - Michael Bonelli
- Division of Rheumatology, Department of Internal Medicine III, Medical University of Vienna, Vienna, Austria; Comprehensive Center for Inflammation and Immunity (CCII), Medical University of Vienna, Vienna, Austria.
| | - Lisa Göschl
- Division of Rheumatology, Department of Internal Medicine III, Medical University of Vienna, Vienna, Austria; Comprehensive Center for Inflammation and Immunity (CCII), Medical University of Vienna, Vienna, Austria
| |
Collapse
|
22
|
Nguyen TH, Chandrakasan S. Biomarkers of Immune Dysregulation and What They Tell Us: Gene Sequencing Is Not the Answer to Every Question. Immunol Allergy Clin North Am 2025; 45:173-188. [PMID: 40287167 DOI: 10.1016/j.iac.2025.01.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/29/2025]
Abstract
Primary immune regulatory disorders (PIRDs) are inborn errors of immunity, with autoimmune, hyperinflammatory, and lymphoproliferative manifestations as presenting features rather than recurrent infections. Genetic testing remains the primary tool for diagnosing patients with immune defects. Not all suspected PIRDs have a known genetic cause. Many hyperinflammatory disorders require urgent intervention, limiting the usefulness of gene sequencing in some cases. Current clinically approved immunology tests can detect immune dysregulation even without apparent immune deficiency. This review presents commonly known patterns of immune dysregulation that can be detected with currently available immune testing and additional testing in the clinical immunology laboratories' pipeline.
Collapse
Affiliation(s)
- Thinh H Nguyen
- Division of Immunology, Boston Children's Hospital, Harvard Medical School, 300 Longwood Avenue, Boston, MA 02115-5737, USA
| | - Shanmuganathan Chandrakasan
- Immune Dysregulation and Immunohematology Program, Department of Pediatrics, Aflac Cancer and Blood Disorder Center, Children's Healthcare of Atlanta, Emory University School of Medicine, 1760 Haygood Drive NE, W-368, Atlanta, GA 30322, USA.
| |
Collapse
|
23
|
Fink DL, Etoori D, Hill R, Idilli O, Kartikapallil N, Payne O, Griffith S, Bradford HF, Mauri C, Kennedy PT, McCoy LE, Maini MK, Gill US. Auto-antibodies against interferons are common in people living with chronic hepatitis B virus infection and associate with PegIFNα non-response. JHEP Rep 2025; 7:101382. [PMID: 40276479 PMCID: PMC12018104 DOI: 10.1016/j.jhepr.2025.101382] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/04/2024] [Revised: 02/09/2025] [Accepted: 02/24/2025] [Indexed: 04/26/2025] Open
Abstract
Background & Aims Type one (T1) and three interferons (T3IFNs) are implicated in chronic hepatitis B (CHB) immunopathogenesis. IFN remains the only licenced immune modulating therapy for CHB. We measured the prevalence of auto-antibodies (auto-Abs) against T1 and T3IFNs to examine the hypothesis that they impact HBV control and treatment response, as highlighted by COVID-19. Methods Our multi-centre retrospective longitudinal study accessed two CHB cohorts; auto-Ab levels and neutralisation status were measured against T1IFN and T3IFN. Associations were tested against HBV clinical parameters. Results Overall, 16.7% (46/276) of patients with CHB had any detectable anti-IFN auto-Abs at any time and 6.5% (18/276) anti-T3IFN auto-Abs, with a high incidence of PegIFNα-induced de novo auto-Abs (31.4%, 11/35). However, only a minority of auto-Ab-positive sera demonstrated neutralisation in vitro (4/46, 8.7%). Auto-Ab positivity correlated with higher median HBsAg levels (p = 0.0110). All individuals with detectable anti-T1IFN auto-Abs were PegIFNα non-responders. Conclusions Non-neutralising anti-IFN auto-Abs are common in CHB and associate with higher median HBsAg levels. Further prospective study of anti-cytokine auto-Abs in CHB are required to characterise the association with long-term outcomes. Impact and implications HBV and PegIFNα individually may induce broad autoreactivity associated with dysregulated antiviral immune responses. Auto-Ab screening prior to PegIFNα treatment or other immunotherapies may play a critical role in predicting treatment responses.
Collapse
Affiliation(s)
- Douglas L. Fink
- Infection and Immunity, University College London, London, UK
- Royal Free London NHS Foundation Trust, London, UK
| | - David Etoori
- Institute for Global Health, University College London, London, UK
| | - Robert Hill
- Infection and Immunity, University College London, London, UK
| | - Orest Idilli
- Infection and Immunity, University College London, London, UK
| | | | - Olivia Payne
- Infection and Immunity, University College London, London, UK
| | - Sarah Griffith
- Infection and Immunity, University College London, London, UK
| | | | - Claudia Mauri
- Infection and Immunity, University College London, London, UK
| | - Patrick T.F. Kennedy
- Barts Liver Centre, Blizard Institute, Barts and The London, School of Medicine & Dentistry, Queen Mary University of London, London, UK
| | - Laura E. McCoy
- Infection and Immunity, University College London, London, UK
| | - Mala K. Maini
- Infection and Immunity, University College London, London, UK
| | - Upkar S. Gill
- Barts Liver Centre, Blizard Institute, Barts and The London, School of Medicine & Dentistry, Queen Mary University of London, London, UK
| |
Collapse
|
24
|
Alalwan D, Leon AAG, Saini G, Gaillard C, Negi R, Heckmann C, Kenny G, Feeney E, Cotter AG, Kelly C, Carr M, de Barra E, Yousif O, Horgan M, Sadlier C, Landay A, Gonzalez G, Mallon PWG. Associations between type III interferons, obesity and clinical severity of COVID-19. Front Immunol 2025; 16:1516756. [PMID: 40330483 PMCID: PMC12052549 DOI: 10.3389/fimmu.2025.1516756] [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: 10/24/2024] [Accepted: 04/01/2025] [Indexed: 05/08/2025] Open
Abstract
Introduction Severe COVID-19 is characterized by hyperimmune host responses contributing to airway damage and acute respiratory distress syndrome. Type III interferons (IFN), including IFN lambda 4 (IFNλ4), expressed in individuals harboring the rs368234815-ΔG allele, are implicated in host immune responses to viral infections, including SARS-CoV-2. Methods We investigated associations between IFNλ4 expression through genotyping and COVID-19 disease severity in 853 laboratory-confirmed SARS-CoV-2 cases enrolled in the All-Ireland Infectious Diseases Cohort. Additionally, we measured plasma levels of Type I, II and III IFN using quantitative immunoassays along with IFNλ4 expression and COVID-19 disease severity in a sub-group [n=321 (37.6%)] with samples available within 10 days of symptom onset. IFNλ4 was expressed in 382 (44.8%) but expression was not significantly associated with COVID-19 disease severity. Results Within the sub-group, we found no consistent associations between IFNλ4 expression and circulating IFNs. However, we observed significantly increased expression of IFNλ1 and IFNλ2 in severe COVID-19 (P<0.01), with IFNλ2 remaining significantly associated after adjustment for age, sex, ethnicity, and comorbidities, including obesity (BMI≥30 kg/m2) (P<0.001). Interestingly, although IFNλ2 levels were significantly higher in subjects with obesity, the association between higher IFNλ2 and COVID-19 disease severity was only observed in individuals without obesity (P<0.01). Conclusion These data reveal an important role for IFNλ2 as an immune correlate that predicts COVID-19 disease severity, which may be masked in those with obesity.
Collapse
Affiliation(s)
- Dana Alalwan
- Centre for Experimental Pathogen Host Research (CEPHR), University College Dublin, Dublin, Ireland
| | | | - Gurvin Saini
- Centre for Experimental Pathogen Host Research (CEPHR), University College Dublin, Dublin, Ireland
| | - Colette Gaillard
- Centre for Experimental Pathogen Host Research (CEPHR), University College Dublin, Dublin, Ireland
| | - Riya Negi
- Centre for Experimental Pathogen Host Research (CEPHR), University College Dublin, Dublin, Ireland
| | | | - Grace Kenny
- Centre for Experimental Pathogen Host Research (CEPHR), University College Dublin, Dublin, Ireland
| | - Eoin Feeney
- Centre for Experimental Pathogen Host Research (CEPHR), University College Dublin, Dublin, Ireland
- Department of Infectious Diseases, St. Vincent’s University Hospital, Dublin, Ireland
| | - Aoife G. Cotter
- Centre for Experimental Pathogen Host Research (CEPHR), University College Dublin, Dublin, Ireland
- Department of Infectious Diseases, Mater Misericordiae University Hospital, Dublin, Ireland
| | - Christine Kelly
- Centre for Experimental Pathogen Host Research (CEPHR), University College Dublin, Dublin, Ireland
- Department of Infectious Diseases, Mater Misericordiae University Hospital, Dublin, Ireland
| | - Michael Carr
- National Virus Reference Laboratory, University College Dublin, Dublin, Ireland
- International Collaboration Unit, Research Centre for Zoonosis Control, Hokkaido University, Sapporo, Japan
| | - Eoghan de Barra
- Department of Infectious Diseases, Beaumont Hospital, Dublin, Ireland
- Department of International Health and Tropical Medicine, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Obada Yousif
- Endocrinology Department, Wexford General Hospital, Wexford, Ireland
| | - Mary Horgan
- Centre for Experimental Pathogen Host Research (CEPHR), University College Dublin, Dublin, Ireland
- Department of Infectious Diseases, St. Vincent’s University Hospital, Dublin, Ireland
| | - Corinna Sadlier
- Department of Infectious Diseases, Cork University Hospital, Cork, Ireland
| | - Alan Landay
- University of Texas Medical Branch at Galveston, Texas, TX, United States
| | - Gabriel Gonzalez
- Institute for Vaccine Research and Development, Hokkaido University, Hokkaido, Japan
| | - Patrick W. G. Mallon
- Centre for Experimental Pathogen Host Research (CEPHR), University College Dublin, Dublin, Ireland
- Department of Infectious Diseases, St. Vincent’s University Hospital, Dublin, Ireland
| |
Collapse
|
25
|
Adiguzel Y, Bogdanos DP, Shoenfeld Y. Molecular/antigenic mimicry and immunological cross-reactivity explains SARS-CoV-2-induced autoimmunity. Autoimmun Rev 2025; 24:103811. [PMID: 40209971 DOI: 10.1016/j.autrev.2025.103811] [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: 01/05/2025] [Revised: 03/07/2025] [Accepted: 03/11/2025] [Indexed: 04/12/2025]
Abstract
COVID-19 pandemic is over, but its effects on chronic illnesses remain a challenging issue. Understanding the influence of SARS-COV-2-mediated autoimmunity and overt autoimmune disease is of paramount importance, as it can provide a critical mass of information regarding both infection-mediated (and vaccination-induced) autoimmune phenomena in susceptible individuals during the disease course, and short or long-term post-disease sequelae. The high prevalence of organ and non-organ specific autoantibody positivity in patients with COVID-19 led to studies attempting to delineate the origin and the underlying mechanism responsible for their induction nature, identifying novel autoantigens, and the self-epitope sequences which could be the impetus for the initiating autoreactive responses. Herein, we provide a meticulous review of the studies reporting those mimicking sequences that have been experimentally validated, based on the assumption that molecular mimicry and immunological crossreactivity may account for autoantibody development. Most reports are based on bioinformatics approaches, and only a disproportionally small number of studies currently demonstrate immunological crossreactivity. We took the opportunity to further review and searched for the linear human epitope sequences of human, through the epitopes deposited at the Immune Epitope Database. This included an analysis of autoimmune disease as the disease data to comprehensively understand the subject matter. The critical overview of the findings underscore the urgent and immense need for further research to gain a comprehensive understanding of the mechanisms involved and the anticipated appraisal that molecular mimicry and immunological crossreactivity is indeed central to the loss of immunological tolerance during SARS-COV-2 infection.
Collapse
Affiliation(s)
- Yekbun Adiguzel
- Department of Medical Biology, School of Medicine, Atilim University, Ankara, Turkey.
| | - Dimitros P Bogdanos
- Department of Rheumatology and Clinical Immunology, Faculty of Medicine, School of Health Sciences, University of Thessaly, Larissa, Greece.
| | - Yehuda Shoenfeld
- Dina Recanati School of Medicine, Reichman University, Herzliya, Israel; Zabludowicz Center for Autoimmune Diseases, Sheba Medical Center, Israel.
| |
Collapse
|
26
|
Niazi V, Ghafouri-Fard S. Effect of hypoxia on extracellular vesicles in malignant and non-malignant conditions. Cancer Treat Res Commun 2025; 43:100924. [PMID: 40209539 DOI: 10.1016/j.ctarc.2025.100924] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2025] [Revised: 03/21/2025] [Accepted: 04/04/2025] [Indexed: 04/12/2025]
Abstract
Extracellular vesicles (EVs) are produced by virtually all types of cells and can be detected in nearly all extracellular places. These particles mediate intercellular communication and transfer their cargo to the recipient cells, inducing a variety of processes in these cells through transmission of several biomolecules such as miRNAs, lncRNAs, other transcripts and a variety of proteins. It has been documented that size, quantity, and expression of biomolecules in the EVs are influenced by the level of oxygen. In fact, hypoxia can affect several cellular processes through modulation of the cargo of these vesicles. Hypoxic exosomes derived from tumor cells have several protumoral effects on the recipient cells, including enhancement of proliferation, migration, and invasion in other tumoral cells, induction of metastasis in distant organs, stimulation of angiogenesis in the endothelial cells, and modulation of macrophage polarization. Hypoxic EVs also contribute to several non-malignant diseases. This review summarizes the effect of hypoxia on EVs cargo in malignant and nonmalignant diseases of different organs.
Collapse
Affiliation(s)
- Vahid Niazi
- Stem Cell Research Center, Golestan University of Medical Science, Gorgan, Iran; School of Advanced Technologies in Medicine, Golestan University of Medical Science, Gorgan, Iran
| | - Soudeh Ghafouri-Fard
- Department of Medical Genetics, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
27
|
Martinez RJ, Hogquist KA. Sterile production of interferons in the thymus. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2025:vkaf048. [PMID: 40184034 DOI: 10.1093/jimmun/vkaf048] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/07/2025] [Accepted: 02/25/2025] [Indexed: 04/05/2025]
Abstract
T-cell central tolerance is controlled by thymocyte TCR recognition of self-peptides presented by thymic APCs. While thymic epithelial cells are essential for T-cell central tolerance, a variety of other traditional APCs also play critical roles in T-cell selection. Similar to how peripheral APCs require activation to become effective, thymic APCs also require activation to become tolerogenic. Recent studies have identified IFNs as an essential factor for the activation and generation of an optimally tolerogenic thymic environment. In this review, we focus on interferon (IFN) production within the thymus and its effects on thymic APCs and developing thymocytes. We also examine the importance of T-cell tolerance to IFN itself as well as to interferon-stimulated proteins generated during peripheral immune responses.
Collapse
Affiliation(s)
- Ryan J Martinez
- Center for Immunology, University of Minnesota Medical School, Minneapolis, MN, United States
- Department of Laboratory Medicine and Pathology, University of Minnesota Medical School, Minneapolis, MN, United States
| | - Kristin A Hogquist
- Center for Immunology, University of Minnesota Medical School, Minneapolis, MN, United States
- Department of Laboratory Medicine and Pathology, University of Minnesota Medical School, Minneapolis, MN, United States
| |
Collapse
|
28
|
Yang OO. The immunopathogenesis of SARS-CoV-2 infection: Overview of lessons learned in the first 5 years. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2025:vkaf033. [PMID: 40180332 DOI: 10.1093/jimmun/vkaf033] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/26/2024] [Accepted: 02/11/2025] [Indexed: 04/05/2025]
Abstract
This review provides a broad overview of lessons learned in the five years since COVID-19 was identified. It is a bimodal disease, starting with an initially virus-driven phase, followed by resolution or ensuing inappropriate immune activation causing severe inflammation that is no longer strictly virus dependent. Humoral immunity is beneficial for preventing or attenuating the early stage, without benefit once the later stage begins. Neutralizing antibodies elicited by natural infection or vaccination are short-lived and highly vulnerable to viral sequence variation. By contrast, cellular immunity, particularly the CD8+ T cell arm, has a role in preventing or attenuating severe disease, is far less susceptible to viral variation, and is longer-lived than antibodies. Finally, an ill-defined phenomenon of prolonged symptoms after acute infection, termed "long COVID," is poorly understood but may involve various immunologic defects that are hyperactivating or immunosuppressive. Remaining issues include needing to better understand the immune dysregulation of severe disease to allow more tailored therapeutic interventions, developing antibody strategies that cope with the viral spike sequence variability, prolonging vaccine efficacy, and unraveling the mechanisms of long COVID to design therapeutic approaches.
Collapse
Affiliation(s)
- Otto O Yang
- Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
- Department of Microbiology, Immunology and Molecular Genetics, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
| |
Collapse
|
29
|
Olivieri G, Amodio D, Manno EC, Santilli V, Cotugno N, Palma P. Shielding the immunocompromised: COVID-19 prevention strategies for patients with primary and secondary immunodeficiencies. Vaccine 2025; 51:126853. [PMID: 39946827 DOI: 10.1016/j.vaccine.2025.126853] [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: 08/07/2024] [Revised: 01/23/2025] [Accepted: 01/24/2025] [Indexed: 03/05/2025]
Abstract
The COVID-19 pandemic has significantly impacted immunocompromised patients, particularly those with inborn errors of immunity (IEI), transplant recipients, hematologic malignancies, and those undergoing treatment with immunosuppressive biologics and medications. These patients face an elevated risk of experiencing severe or even fatal consequences following SARS-CoV-2 infections. Vaccination is the primary defense against COVID-19; however, immune responses following immunization are often suboptimal in these patients, with variable specific humoral response rates. Despite the expedited regulatory approval and the widespread implementation of COVID-19 vaccines, the efficacy and safety for immunocompromised populations require thorough investigation. In future pandemics, including vulnerable populations (VPs) in vaccine and monoclonal antibody (mAb) trials is crucial to develop safe, effective immunization strategies, address gaps in vaccine efficacy and safety data, and create tailored guidelines for at-risk groups. This review provides a comprehensive examination of the efficacy of COVID-19 vaccines and mAbs in patients with primary and secondary immunodeficiency, with a specific focus on individuals with IEI, considering previous regulatory aspects and the necessity of including VPs in vaccine trials to enhance the quality of patient care and promote equitable health outcomes in future pandemics.
Collapse
Affiliation(s)
- Giulio Olivieri
- Clinical Immunology and Vaccinology Unit, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy; PhD Program in Immunology, Molecular Medicine and Applied Biotechnology, University of Rome Tor Vergata, Rome, Italy
| | - Donato Amodio
- Clinical Immunology and Vaccinology Unit, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy; Chair of Pediatrics, Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy
| | - Emma Concetta Manno
- Clinical Immunology and Vaccinology Unit, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - Veronica Santilli
- Clinical Immunology and Vaccinology Unit, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - Nicola Cotugno
- Clinical Immunology and Vaccinology Unit, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy; Chair of Pediatrics, Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy
| | - Paolo Palma
- Chair of Pediatrics, Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy; Centre for the Evaluation of Vaccination and Vaccine & Infectious Disease Institute, University of Antwerp, Antwerp, Belgium.
| |
Collapse
|
30
|
Ferrari A, Cassaniti I, Rovida F, Lilleri D, Croce S, Trespidi F, Ghirardello S, Gervais A, Zhang SY, Casanova JL, Borghesi A, Baldanti F. Human type I interferons protect Vero E6 and ARPE-19 cells against West Nile virus and are neutralized by pathogenic autoantibodies. Sci Rep 2025; 15:11271. [PMID: 40175402 PMCID: PMC11965296 DOI: 10.1038/s41598-025-89312-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2024] [Accepted: 02/04/2025] [Indexed: 04/04/2025] Open
Abstract
Auto-antibodies (auto-Abs) that neutralize type I interferons (IFNs) have been implicated in severe viral infections, including ~ 40% of cases of West Nile virus (WNV) neuroinvasive disease (WNND). Developing robust in vitro models to evaluate the protective effects of type I IFNs against viral infection, as well as the disruptive effects of auto-Abs, is essential for understanding disease pathogenesis and identifying patients at risk. In this study, we used Vero E6 and ARPE-19 cell lines to investigate the ability of type I (IFN-α, IFN-β, IFN-ω), type II (IFN-γ), and type III (IFN-λ1) IFNs to restrict WNV infection. Our results demonstrate that IFN-α, IFN-β, and IFN-ω effectively protect ARPE-19 cells from WNV infection, with IFN-β exhibiting the strongest antiviral effect. In contrast, Vero E6 cells required higher concentrations of IFN-ω to achieve comparable protection. Neither IFN-γ nor IFN-λ1 conferred protection in either cell line. We further screened serum samples from WNV-infected patients for auto-Abs neutralizing type I IFNs. Our findings confirm that the ARPE-19-based assay is consistent with other established methods for detecting neutralizing auto-Abs against type I IFNs. This simple and reliable assay offers a valuable tool for assessing the antiviral effects of type I IFNs and the neutralizing activity of auto-Abs in both research and clinical settings. Future studies should aim to validate the clinical utility of the ARPE-19-WNV infection model on a larger scale.
Collapse
Affiliation(s)
- Alessandro Ferrari
- National PhD Programme in One Health approaches to infectious diseases and life science research, Department of Public Health, Experimental and Forensic Medicine, University of Pavia, Pavia, Italy
- Microbiology and Virology Unit, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | - Irene Cassaniti
- Department of Clinical, Surgical, Diagnostic and Pediatric Sciences, University of Pavia, Pavia, Italy.
- Microbiology and Virology Unit, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy.
| | - Francesca Rovida
- Department of Clinical, Surgical, Diagnostic and Pediatric Sciences, University of Pavia, Pavia, Italy
- Microbiology and Virology Unit, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | - Daniele Lilleri
- Microbiology and Virology Unit, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | - Stefania Croce
- UOSD Cell Factory, San Matteo Research Hospital, Pavia, Italy
| | - Francesca Trespidi
- Neonatal Intensive Care Unit, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | - Stefano Ghirardello
- Neonatal Intensive Care Unit, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | - Adrian Gervais
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, Institut National de la Santé et de la Recherche Médicale (INSERM) U1163, Necker Hospital for Sick Children, Paris, France
- Paris Cité University, Imagine Institute, Paris, France
| | - Shen-Ying Zhang
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, Institut National de la Santé et de la Recherche Médicale (INSERM) U1163, Necker Hospital for Sick Children, Paris, France
- Paris Cité University, Imagine Institute, Paris, France
- St. Giles Laboratory of Human Genetics of Infectious Diseases, The Rockefeller University, Rockefeller Branch, New York, NY, USA
| | - Jean-Laurent Casanova
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, Institut National de la Santé et de la Recherche Médicale (INSERM) U1163, Necker Hospital for Sick Children, Paris, France
- Paris Cité University, Imagine Institute, Paris, France
- St. Giles Laboratory of Human Genetics of Infectious Diseases, The Rockefeller University, Rockefeller Branch, New York, NY, USA
- Howard Hughes Medical Institute, New York, NY, USA
- Department of Pediatrics, Necker Hospital for Sick Children, Paris, France
| | - Alessandro Borghesi
- Neonatal Intensive Care Unit, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
- School of Life Sciences, Swiss Federal Institute of Technology, Lausanne, Switzerland
| | - Fausto Baldanti
- Department of Clinical, Surgical, Diagnostic and Pediatric Sciences, University of Pavia, Pavia, Italy
- Microbiology and Virology Unit, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| |
Collapse
|
31
|
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
|
32
|
Anwar S, Kader A, Debnath SK, Jarin F, Sayem ASM, Miah MF. Biophysical assessments and blood profiling reveal physiological adaptations and environmental interactions of hilsa shad (Tenualosa ilisha). PLoS One 2025; 20:e0320628. [PMID: 40168310 PMCID: PMC11960910 DOI: 10.1371/journal.pone.0320628] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2024] [Accepted: 02/22/2025] [Indexed: 04/03/2025] Open
Abstract
The hilsa shad (Tenualosa ilisha) is a migratory fish of great economic and cultural importance in Bangladesh. However, its physiological adaptations to diverse environmental conditions are not well understood. This study provides a comprehensive assessment of the biophysical, hematological, and biochemical characteristics of hilsa shad from riverine, estuarine, and marine environments in Bangladesh. We sampled 180 adult fish from nine sites, revealing significant habitat-specific variations in water quality, with marine sites showing the highest levels of total dissolved solids (TDS) and specific conductance. Hematological analysis showed that fish from riverine environments had higher hematocrit values, which are crucial for oxygen transport during migration. Post-spawning fish exhibited elevated hemoglobin, RBC counts, and hematocrit levels, reflecting adaptations to the metabolic demands of migration and spawning. WBC counts were higher in females, particularly in estuarine and marine environments, suggesting a stronger immune response to environmental stressors. Serum biochemical analysis showed significant variations in liver enzyme activity, albumin, and cortisol levels, with elevated cortisol in riverine fish during spawning season, indicating heightened stress. These findings enhance our understanding of hilsa shad's physiological ecology, providing critical insights for conservation and management strategies amid environmental changes.
Collapse
Affiliation(s)
- Saeed Anwar
- Department of Genetic Engineering and Biotechnology, School of Life Sciences, Shahjalal University of Science and Technology, Sylhet, Bangladesh
| | - Abdul Kader
- Department of Genetic Engineering and Biotechnology, School of Life Sciences, Shahjalal University of Science and Technology, Sylhet, Bangladesh
| | - Smrity Kona Debnath
- Department of Genetic Engineering and Biotechnology, School of Life Sciences, Shahjalal University of Science and Technology, Sylhet, Bangladesh
| | - Faria Jarin
- Department of Genetic Engineering and Biotechnology, School of Life Sciences, Shahjalal University of Science and Technology, Sylhet, Bangladesh
| | - Abu Saleh Mohammad Sayem
- Department of Food Engineering and Tea Technology, School of Applied Sciences and Technology, Shahjalal University of Science and Technology, Sylhet, Bangladesh
| | - Md. Faruque Miah
- Department of Genetic Engineering and Biotechnology, School of Life Sciences, Shahjalal University of Science and Technology, Sylhet, Bangladesh
| |
Collapse
|
33
|
Pons S, Generenaz L, Gervais A, Puel A, Bastard P, Renard N, Guyot V, Vinit C, Zheng F, Brengel‐Pesce K, Saker K, Richard J, Mathian A, Amoura Z, Dorgham K, Gorochov G, Casanova J, Fleurie A, Trouillet‐Assant S. Rapid Detection of Anti-IFN-α2 Autoantibodies Using a New Automated VIDAS Assay Prototype. Eur J Immunol 2025; 55:e202451516. [PMID: 40223598 PMCID: PMC11995241 DOI: 10.1002/eji.202451516] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2024] [Revised: 02/20/2025] [Accepted: 02/24/2025] [Indexed: 04/15/2025]
Abstract
Autoantibodies neutralizing Type I interferons increase the risk of severe viral diseases and are linked to autoimmune conditions. The automated VIDAS assay is suitable for anti-IFN-α2 IgGs quantification, offering a swift, reliable, user-friendly, single test for clinical management.
Collapse
Affiliation(s)
- Sylvie Pons
- Joint Research Unit Civils Hospices of Lyon‐bioMérieux, Hospices Civils de LyonLyon Sud HospitalPierre‐BéniteFrance
| | - Laurence Generenaz
- Joint Research Unit Civils Hospices of Lyon‐bioMérieux, Hospices Civils de LyonLyon Sud HospitalPierre‐BéniteFrance
| | - Adrian Gervais
- St. Giles Laboratory of Human Genetics of Infectious DiseasesRockefeller BranchThe Rockefeller UniversityNew YorkNew YorkUSA
- Laboratory of Human Genetics of Infectious DiseasesNecker BranchINSERM U1163Necker Hospital for Sick ChildrenParisFrance
| | - Anne Puel
- St. Giles Laboratory of Human Genetics of Infectious DiseasesRockefeller BranchThe Rockefeller UniversityNew YorkNew YorkUSA
- Laboratory of Human Genetics of Infectious DiseasesNecker BranchINSERM U1163Necker Hospital for Sick ChildrenParisFrance
- Imagine InstituteParis Cité UniversityParisFrance
| | - Paul Bastard
- St. Giles Laboratory of Human Genetics of Infectious DiseasesRockefeller BranchThe Rockefeller UniversityNew YorkNew YorkUSA
- Laboratory of Human Genetics of Infectious DiseasesNecker BranchINSERM U1163Necker Hospital for Sick ChildrenParisFrance
- Imagine InstituteParis Cité UniversityParisFrance
- Pediatric Hematology‐Immunology and Rheumatology UnitNecker Hospital for Sick ChildrenAssistance Publique‐Hôpitaux de Paris (AP‐HP)ParisFrance
| | - Nathalie Renard
- R&D Department of ImmunoassaysbioMérieux S.A.Marcy l'EtoileFrance
| | - Valerie Guyot
- R&D Department of ImmunoassaysbioMérieux S.A.Marcy l'EtoileFrance
| | - Cecile Vinit
- R&D Department of ImmunoassaysbioMérieux S.A.Marcy l'EtoileFrance
| | - Fei Zheng
- Joint Research Unit Civils Hospices of Lyon‐bioMérieux, Hospices Civils de LyonLyon Sud HospitalPierre‐BéniteFrance
| | - Karen Brengel‐Pesce
- Joint Research Unit Civils Hospices of Lyon‐bioMérieux, Hospices Civils de LyonLyon Sud HospitalPierre‐BéniteFrance
| | - Kahina Saker
- Joint Research Unit Civils Hospices of Lyon‐bioMérieux, Hospices Civils de LyonLyon Sud HospitalPierre‐BéniteFrance
| | - Jean‐Christophe Richard
- Intensive Care UnitCroix‐Rousse HospitalHospices Civils de LyonLyonFrance
- INSA‐Lyon, CNRS, INSERM, CREATIS UMR 5220, U1294Université de Lyon, Université Claude Bernard Lyon 1VilleurbanneFrance
| | - Alexis Mathian
- Assistance Publique–Hôpitaux de ParisSorbonne Université, Groupement Hospitalier Pitié–Salpêtrière, French National Referral Center for Systemic Lupus Erythematosus, Antiphospholipid Antibody Syndrome and Other Autoimmune Disorders, Service de Médecine Interne 2, Institut E3M, Inserm UMRS, Centre d'Immunologie et des Maladies Infectieuses (CIMI‐Paris)ParisFrance
| | - Zahir Amoura
- Assistance Publique–Hôpitaux de ParisSorbonne Université, Groupement Hospitalier Pitié–Salpêtrière, French National Referral Center for Systemic Lupus Erythematosus, Antiphospholipid Antibody Syndrome and Other Autoimmune Disorders, Service de Médecine Interne 2, Institut E3M, Inserm UMRS, Centre d'Immunologie et des Maladies Infectieuses (CIMI‐Paris)ParisFrance
| | - Karim Dorgham
- Sorbonne Université, Inserm, Centre d'Immunologie et des Maladies Infectieuses (CIMI‐Paris)ParisFrance
| | - Guy Gorochov
- Sorbonne Université, Inserm, Centre d'Immunologie et des Maladies Infectieuses (CIMI‐Paris)ParisFrance
- Département d'Immunologie, AP‐HPGroupement Hospitalier Pitié‐SalpêtrièreParisFrance
| | - Jean‐Laurent Casanova
- St. Giles Laboratory of Human Genetics of Infectious DiseasesRockefeller BranchThe Rockefeller UniversityNew YorkNew YorkUSA
- Laboratory of Human Genetics of Infectious DiseasesNecker BranchINSERM U1163Necker Hospital for Sick ChildrenParisFrance
- Imagine InstituteParis Cité UniversityParisFrance
- Pediatric Hematology‐Immunology and Rheumatology UnitNecker Hospital for Sick ChildrenAssistance Publique‐Hôpitaux de Paris (AP‐HP)ParisFrance
- Howard Hughes Medical InstituteNew YorkNew YorkUSA
| | - Aurore Fleurie
- Joint Research Unit Civils Hospices of Lyon‐bioMérieux, Hospices Civils de LyonLyon Sud HospitalPierre‐BéniteFrance
| | - Sophie Trouillet‐Assant
- Joint Research Unit Civils Hospices of Lyon‐bioMérieux, Hospices Civils de LyonLyon Sud HospitalPierre‐BéniteFrance
- International Center of Research in Infectiology, Virpath teamLyon UniversityINSERM U1111, CNRS UMR 5308, ENS, UCBLLyonFrance
| |
Collapse
|
34
|
Hromić-Jahjefendić A, Mahmutović L, Sezer A, Bećirević T, Rubio-Casillas A, Redwan EM, Uversky VN. The intersection of microbiome and autoimmunity in long COVID-19: Current insights and future directions. Cytokine Growth Factor Rev 2025; 82:43-54. [PMID: 39179487 DOI: 10.1016/j.cytogfr.2024.08.002] [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: 07/15/2024] [Revised: 08/08/2024] [Accepted: 08/13/2024] [Indexed: 08/26/2024]
Abstract
Long COVID-19 affects a significant percentage of patients and is characterized by a wide range of symptoms, including weariness and mental fog as well as emotional symptoms like worry and sadness. COVID-19 is closely linked to the autoimmune disorders that are becoming more prevalent worldwide and are linked to immune system hyperactivation, neutrophil extracellular trap (NET) development, and molecular mimicry pathways. Long-term COVID-related autoimmune responses include a watchful immune system referring to the ability of immune system to constantly monitor the body for signs of infection, disease, or abnormal cells; altered innate and adaptive immune cells, autoantigens secreted by living or dead neutrophils, and high concentrations of autoantibodies directed against different proteins. The microbiome, which consists of billions of bacteria living in the human body, is essential for controlling immune responses and supporting overall health. The microbiome can affect the course of long COVID-associated autoimmunity, including the degree of illness, the rate of recovery, and the onset of autoimmune reactions. Although the precise role of the microbiome in long COVID autoimmunity is still being investigated, new studies indicate that probiotics, prebiotics, and dietary changes-interventions that target the microbiome-may be able to reduce autoimmune reactions and enhance long-term outcomes for COVID-19 survivors. More research is required to precisely understand how the microbiome affects COVID-19-related autoimmunity and to create tailored treatment plans.
Collapse
Affiliation(s)
- Altijana Hromić-Jahjefendić
- Department of Genetics and Bioengineering, Faculty of Engineering and Natural Sciences, International University of Sarajevo, Hrasnicka cesta 15, Sarajevo 71000, Bosnia and Herzegovina.
| | - Lejla Mahmutović
- Department of Genetics and Bioengineering, Faculty of Engineering and Natural Sciences, International University of Sarajevo, Hrasnicka cesta 15, Sarajevo 71000, Bosnia and Herzegovina.
| | - Abas Sezer
- Department of Genetics and Bioengineering, Faculty of Engineering and Natural Sciences, International University of Sarajevo, Hrasnicka cesta 15, Sarajevo 71000, Bosnia and Herzegovina.
| | - Tea Bećirević
- Atrijum Polyclinic, Sarajevo, Bosnia and Herzegovina
| | - Alberto Rubio-Casillas
- Autlan Regional Hospital, Health Secretariat, Autlan, Jalisco 48900, Mexico; Biology Laboratory, Autlan Regional Preparatory School, University of Guadalajara, Autlan, Jalisco 48900, Mexico.
| | - Elrashdy M Redwan
- Department of Biological Sciences, Faculty of Science, King Abdulaziz University, Jeddah 21589, Saudi Arabia; Therapeutic and Protective Proteins Laboratory, Protein Research Department, Genetic Engineering and Biotechnology Research Institute, City of Scientific Research and Technological Applications, New Borg EL-Arab 21934, 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, 12901 Bruce B. Downs Blvd., MDC07, Tampa, FL, USA.
| |
Collapse
|
35
|
Aljabali AAA, Obeid M, Gammoh O, El-Tanani M, Tambuwala MM. Guardians at the gate: Unraveling Type I interferon's role and challenges posed by anti-interferon antibodies in COVID-19. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2025; 213:135-169. [PMID: 40246343 DOI: 10.1016/bs.pmbts.2025.01.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/19/2025]
Abstract
The intricate interplay involving Type I interferon (IFN), anti-interferon antibodies, and COVID-19 elucidates a complex symphony within the immune system. This chapter thoroughly explores the dynamic landscape of Type I IFN, delineating its pivotal role as the guardian of the immune response. As SARS-CoV-2 engages the host, the delicate balance of IFN induction and signaling pathways is disrupted, resulting in a nuanced impact on the severity and pathogenesis of COVID-19. Clinical studies illuminate a critical link between impaired IFN response and severe outcomes, uncovering genetic factors contributing to susceptibility. Furthermore, the emergence of anti-interferon antibodies proves to be a disruptive force, compromising the immune arsenal and correlating with disease severity. Our chapter encompasses diagnostic and prognostic implications, highlighting the importance of assays in identifying levels of IFN and anti-interferon antibodies. This chapter examines the possible incorporation of interferon-related biomarkers in COVID-19 diagnostics, offering predictive insights into disease progression. On the therapeutic front, efforts to manipulate the IFN pathway undergo scrutiny, encountering complexities in light of anti-interferon antibodies. This chapter concludes by outlining prospective avenues for precision medicine, emphasizing the imperative need for a comprehensive comprehension of the IFN landscape and its intricate interaction with COVID-19.
Collapse
Affiliation(s)
- Alaa A A Aljabali
- Department of Pharmaceutics and Pharmaceutical Technology, Faculty of Pharmacy, Yarmouk University, Irbid, Jordan.
| | - Mohammad Obeid
- Department of Pharmaceutics and Pharmaceutical Technology, Faculty of Pharmacy, Yarmouk University, Irbid, Jordan
| | - Omar Gammoh
- Department of Clinical Pharmacy and Pharmacy Practice, Faculty of Pharmacy, Yarmouk University, Irbid, Jordan
| | - Mohamed El-Tanani
- College of Pharmacy, Ras Al Khaimah Medical and Health Sciences University, United Arab Emirates
| | - Murtaza M Tambuwala
- Lincoln Medical School, University of Lincoln, Brayford Pool Campus, Lincoln, United Kingdom.
| |
Collapse
|
36
|
Biglari S, Youssefian L, Tabatabaiefar MA, Saeidian AH, Abtahi-Naeini B, Khorram E, Sherkat R, Moghaddam AS, Mohaghegh F, Rahimi M, Rahimi H, Babaei S, Shahrooei M, Mozafari N, Zaresharifi S, Vahidnezhad F, Homayouni V, Tsoi LC, Gudjonsson JE, Hakonarson H, Casanova JL, Jouanguy E, Béziat V, Zhang Q, Cobat A, Vahidnezhad H. DOCK2 Deficiency and GATA2 Haploinsufficiency Can Underlie Critical Coronavirus Disease 2019 (COVID-19) Pneumonia. J Clin Immunol 2025; 45:85. [PMID: 40153067 PMCID: PMC11953147 DOI: 10.1007/s10875-025-01877-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2024] [Accepted: 03/16/2025] [Indexed: 03/30/2025]
Abstract
The life-threatening coronavirus disease 2019 (COVID-19) affects about 1 in 1,000 healthy people under 50 without underlying conditions. Among patients with critical COVID-19 pneumonia, rare germline variants at genes controlling type I IFN immunity have been reported in up to 5% of patients. Causal etiologies in 80-85% of cases are still unknown. We analyzed two families with hypoxemic COVID-19 pneumonia for known single-gene inborn errors of immunity. In Family 1, two siblings with critical COVID-19 were homozygous for a DOCK2 variant, c.3624+5G>A. DOCK2 deficiency is a known T-cell disorder underlying severe viral diseases. The variant resulted in skipping exon 35, which was predicted to produce a frameshift truncated protein (p.L1157Ifs*12). The proband showed markedly decreased blood CD4 T-helper cell counts, impaired T lymphocyte transformation test, and increased serum IgG, IgA, and IgE levels, as documented in other DOCK2-deficient patients. In Family 2, the proband had lethal COVID-19 and HPV-2-associated multiple recalcitrant warts. She was heterozygous for a deletion in GATA2:c.1075_1102del28, p.W360Sfs*18. GATA2 haploinsufficiency is a known cause of severe viral diseases due to a lack of plasmacytoid dendritic cell (pDC) development. The proband had monocytopenia and a lack of circulating pDCs, as reported in other patients with GATA2 haploinsufficiency. Overall, both DOCK2 deficiency and GATA2 haploinsufficiency are associated with critical and often fatal COVID-19 pneumonia.
Collapse
Affiliation(s)
- Sajjad Biglari
- Department of Genetics and Molecular Biology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Leila Youssefian
- Department of Pathology, Cytogenetics Laboratory, City of Hope National Medical Center, Irwindale, CA, USA
| | - Mohammad Amin Tabatabaiefar
- Department of Genetics and Molecular Biology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Amir Hossein Saeidian
- Center for Applied Genomics, Children'S Hospital of Philadelphia, Philadelphia, PA, USA
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA
| | - Bahareh Abtahi-Naeini
- Pediatric Dermatology Division, Department of Pediatrics, Imam Hossein Children'S Hospital, Isfahan University of Medical Sciences, Isfahan, Iran
- Skin Diseases and Leishmaniasis Research Center, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Erfan Khorram
- Applied Physiology Research Center, Cardiovascular Research Institute, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Roya Sherkat
- Immunodeficiency Diseases Research Center, Isfahan University of Medical Sciences, Isfahan, Iran
| | | | - Fatemeh Mohaghegh
- Department of Dermatology, Skin Diseases and Leishmaniasis Research Center, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Maziyar Rahimi
- Pediatrics Department, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Hamid Rahimi
- Pediatric Infectious Diseases Department, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Sharareh Babaei
- Department of Asthma, Allergy and Clinical Immunology, Child Growth and Development Research Center, Research Institute of Primordial Prevention of Non-Communicable Disease, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Mohammad Shahrooei
- Dr. Shahrooei Laboratory, Tehran, Iran
- Clinical and Diagnostic Immunology, 3000, Louvain, KU, Belgium
| | - Nikoo Mozafari
- Skin Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Shirin Zaresharifi
- Skin Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Fatemeh Vahidnezhad
- Department of Computer Science and Engineering Technology, University of Maryland Eastern Shore, Princess Anne, MD, USA
| | - Vida Homayouni
- Immunodeficiency Diseases Research Center, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Lam C Tsoi
- Department of Dermatology, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Johann E Gudjonsson
- Department of Dermatology, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Hakon Hakonarson
- Center for Applied Genomics, Children'S Hospital of Philadelphia, Philadelphia, PA, USA
- Division of Human Genetics, Children'S Hospital of Philadelphia, Philadelphia, PA, USA
- Department of Pediatrics, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA, USA
| | - Jean-Laurent Casanova
- St Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, Rockefeller University, New York, NY, USA
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, Necker Hospital for Sick Children, Inserm U1163, Paris, France
- Imagine Institute, Paris Cité University, Paris, France
- Department of Pediatrics, Necker Hospital for Sick Children, Paris, EU, France
- Howard Hughes Medical Institute, Chevy Chase, MD, 20815, USA
| | - Emmanuelle Jouanguy
- St Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, Rockefeller University, New York, NY, USA
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, Necker Hospital for Sick Children, Inserm U1163, Paris, France
- Imagine Institute, Paris Cité University, Paris, France
| | - Vivien Béziat
- St Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, Rockefeller University, New York, NY, USA
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, Necker Hospital for Sick Children, Inserm U1163, Paris, France
- Imagine Institute, Paris Cité University, Paris, France
| | - Qian Zhang
- St Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, Rockefeller University, New York, NY, USA
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, Necker Hospital for Sick Children, Inserm U1163, Paris, France
- Imagine Institute, Paris Cité University, Paris, France
| | - Aurélie Cobat
- St Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, Rockefeller University, New York, NY, USA
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, Necker Hospital for Sick Children, Inserm U1163, Paris, France
- Imagine Institute, Paris Cité University, Paris, France
| | - Hassan Vahidnezhad
- Center for Applied Genomics, Children'S Hospital of Philadelphia, Philadelphia, PA, USA.
- Division of Human Genetics, Children'S Hospital of Philadelphia, Philadelphia, PA, USA.
- Department of Pediatrics, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA, USA.
- Department of Dermatology, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA, USA.
| |
Collapse
|
37
|
Quentric P, Charuel JL, Moyon Q, Hékimian G, Dorgham K, Lifermann F, Kerneis M, Mathian A, Aacha K, Melki I, Chommeloux J, Petit M, Gautier M, Bay P, Rouvier P, Charpentier E, da Mata-Jardin O, Lefevre L, Parizot C, Saura O, Levy D, Ortuno S, Schmidt M, Luyt CE, Gorochov G, Amoura Z, Combes A, Pineton de Chambrun M. Anti-RNApol3-Associated myocarditis: an emerging disease linking autoimmunity and infection. Ann Intensive Care 2025; 15:38. [PMID: 40123018 PMCID: PMC11930900 DOI: 10.1186/s13613-025-01443-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2024] [Accepted: 01/22/2025] [Indexed: 03/25/2025] Open
Abstract
BACKGROUND Fulminant myocarditis (FM) is a severe condition primarily triggered by viruses. Anti-RNA polymerase III autoantibodies (RNApol3) which are typically found in patients with severe systemic sclerosis, have been reported in patients with influenza-related FM. Our objective is to provide additional insight into RNApol3-associated FM. METHODS We retrospectively included all patients admitted to our institution between January 2013 and June 2023 with acute myocarditis and positive serum RNApol3. We compared their characteristics, etiologies, and outcomes with those of a cohort of RNApol3 negative acute myocarditis. RESULTS Twenty-nine RNApol3-positive patients, comprising 83% females with a mean age of 39 ± 12 years, were included in this study. Each patient was admitted to the intensive care unit at least once and 11 (38%) relapsed. Triggers included influenza virus in 55% and SARS-CoV-2 virus in 48% of cases. The lowest left ventricular ejection fraction was 10 [5-10] % and the highest troponin value was 82 [22-360] times the ULN. Patients required dobutamine (94%), veno-arterial extracorporeal membrane oxygenation (85%) and pericardiocentesis (38%). At the last follow-up, 76% of patients were still alive, while 7% had undergone cardiac transplantation, and 3% required a left ventricular assist device. Compared to RNApol3-negative cases, RNApol3-positive myocarditis was associated with female gender, fulminant evolution, tamponade, a higher likelihood of being caused by a proven viral infection, and a higher rate of relapse. CONCLUSION RNApol3-associated myocarditis is an emerging disease linking autoimmunity and infection and a unique cause of acquired, pathogen-specific, organ-specific immunodeficiency. RNApol3 should be screened in all cases of FM, especially in young women infected by RNA viruses. The risk of FM in RNApol3-positive systemic sclerosis needs further investigation.
Collapse
Affiliation(s)
- Paul Quentric
- Centre d'Immunologie et des Maladies Infectieuses (CIMI-Paris), Sorbonne Université, Inserm, Paris, France
- Service de Médecine Interne 2, Centre de Référence National Lupus Systémique et Syndrome des Anticorps Anti-phospholipides, Sorbonne Université, Assistance Publique-Hôpitaux de Paris (AP-HP), Hôpital La Pitié-Salpêtrière, Institut E3M, Paris, France
| | - Jean-Luc Charuel
- Département d'Immunologie, Sorbonne Université, AP-HP, Hôpital La Pitié-Salpêtrière, Paris, France
| | - Quentin Moyon
- Service de Médecine Interne 2, Centre de Référence National Lupus Systémique et Syndrome des Anticorps Anti-phospholipides, Sorbonne Université, Assistance Publique-Hôpitaux de Paris (AP-HP), Hôpital La Pitié-Salpêtrière, Institut E3M, Paris, France
- Service de Médecine Intensive-Réanimation, Sorbonne Université, AP-HP, Hôpital La Pitié-Salpêtrière, 47-83, boulevard de l'Hôpital, Paris Cedex, 75651, France
| | - Guillaume Hékimian
- Service de Médecine Intensive-Réanimation, Sorbonne Université, AP-HP, Hôpital La Pitié-Salpêtrière, 47-83, boulevard de l'Hôpital, Paris Cedex, 75651, France
| | - Karim Dorgham
- Centre d'Immunologie et des Maladies Infectieuses (CIMI-Paris), Sorbonne Université, Inserm, Paris, France
| | | | - Mathieu Kerneis
- ACTION Study Group, Département de Cardiologie, Sorbonne Université, AP-HP, Hôpital La Pitié-Salpêtrière, Paris, France
- Institut de Cardiométabolisme et Nutrition (ICAN), Sorbonne Université, INSERM, UMRS_1166-ICAN, Paris, F-75013, France
| | - Alexis Mathian
- Centre d'Immunologie et des Maladies Infectieuses (CIMI-Paris), Sorbonne Université, Inserm, Paris, France
- Service de Médecine Interne 2, Centre de Référence National Lupus Systémique et Syndrome des Anticorps Anti-phospholipides, Sorbonne Université, Assistance Publique-Hôpitaux de Paris (AP-HP), Hôpital La Pitié-Salpêtrière, Institut E3M, Paris, France
| | - Karim Aacha
- ACTION Study Group, Département de Cardiologie, Sorbonne Université, AP-HP, Hôpital La Pitié-Salpêtrière, Paris, France
| | - Isabelle Melki
- Service de Pédiatrie Générale Paris, Université Paris Diderot, AP-HP, Hôpital Robert-Debré, Paris, France
| | - Juliette Chommeloux
- Service de Médecine Intensive-Réanimation, Sorbonne Université, AP-HP, Hôpital La Pitié-Salpêtrière, 47-83, boulevard de l'Hôpital, Paris Cedex, 75651, France
| | - Matthieu Petit
- Service de Médecine Intensive-Réanimation, Sorbonne Université, AP-HP, Hôpital La Pitié-Salpêtrière, 47-83, boulevard de l'Hôpital, Paris Cedex, 75651, France
| | - Melchior Gautier
- Service de Médecine Intensive-Réanimation, Sorbonne Université, AP-HP, Hôpital La Pitié-Salpêtrière, 47-83, boulevard de l'Hôpital, Paris Cedex, 75651, France
| | - Pierre Bay
- Service de Médecine Intensive Réanimation and UPEC, Hôpitaux universitaires Henri Mondor, AP-HP, DMU Médecine, Université Paris Est), INSERM, Unit é U955, équipe 18, Créteil, 94010, France
| | - Philippe Rouvier
- Service d'Anatomopathologie, Sorbonne Université, AP-HP, Hôpital La Pitié-Salpêtrière, Paris, France
| | - Etienne Charpentier
- Département d'Imagerie Cardiothoracique, Sorbonne Université, AP-HP, Hôpital La Pitié-Salpêtrière, Paris, France
| | - Omaira da Mata-Jardin
- Centre d'Immunologie et des Maladies Infectieuses (CIMI-Paris), Sorbonne Université, Inserm, Paris, France
| | - Lucie Lefevre
- Service de Médecine Intensive-Réanimation, Sorbonne Université, AP-HP, Hôpital La Pitié-Salpêtrière, 47-83, boulevard de l'Hôpital, Paris Cedex, 75651, France
| | - Christophe Parizot
- Département d'Immunologie, Sorbonne Université, AP-HP, Hôpital La Pitié-Salpêtrière, Paris, France
| | - Ouriel Saura
- Service de Médecine Intensive-Réanimation, Sorbonne Université, AP-HP, Hôpital La Pitié-Salpêtrière, 47-83, boulevard de l'Hôpital, Paris Cedex, 75651, France
| | - David Levy
- Service de Médecine Intensive-Réanimation, Sorbonne Université, AP-HP, Hôpital La Pitié-Salpêtrière, 47-83, boulevard de l'Hôpital, Paris Cedex, 75651, France
| | - Sofia Ortuno
- Service de Médecine Intensive-Réanimation, Sorbonne Université, AP-HP, Hôpital La Pitié-Salpêtrière, 47-83, boulevard de l'Hôpital, Paris Cedex, 75651, France
| | - Matthieu Schmidt
- Service de Médecine Intensive-Réanimation, Sorbonne Université, AP-HP, Hôpital La Pitié-Salpêtrière, 47-83, boulevard de l'Hôpital, Paris Cedex, 75651, France
- Institut de Cardiométabolisme et Nutrition (ICAN), Sorbonne Université, INSERM, UMRS_1166-ICAN, Paris, F-75013, France
| | - Charles-Edouard Luyt
- Service de Médecine Intensive-Réanimation, Sorbonne Université, AP-HP, Hôpital La Pitié-Salpêtrière, 47-83, boulevard de l'Hôpital, Paris Cedex, 75651, France
- Institut de Cardiométabolisme et Nutrition (ICAN), Sorbonne Université, INSERM, UMRS_1166-ICAN, Paris, F-75013, France
| | - Guy Gorochov
- Centre d'Immunologie et des Maladies Infectieuses (CIMI-Paris), Sorbonne Université, Inserm, Paris, France
- Département d'Immunologie, Sorbonne Université, AP-HP, Hôpital La Pitié-Salpêtrière, Paris, France
| | - Zahir Amoura
- Centre d'Immunologie et des Maladies Infectieuses (CIMI-Paris), Sorbonne Université, Inserm, Paris, France
- Service de Médecine Intensive-Réanimation, Sorbonne Université, AP-HP, Hôpital La Pitié-Salpêtrière, 47-83, boulevard de l'Hôpital, Paris Cedex, 75651, France
| | - Alain Combes
- Service de Médecine Intensive-Réanimation, Sorbonne Université, AP-HP, Hôpital La Pitié-Salpêtrière, 47-83, boulevard de l'Hôpital, Paris Cedex, 75651, France
- Institut de Cardiométabolisme et Nutrition (ICAN), Sorbonne Université, INSERM, UMRS_1166-ICAN, Paris, F-75013, France
| | - Marc Pineton de Chambrun
- Centre d'Immunologie et des Maladies Infectieuses (CIMI-Paris), Sorbonne Université, Inserm, Paris, France.
- Service de Médecine Interne 2, Centre de Référence National Lupus Systémique et Syndrome des Anticorps Anti-phospholipides, Sorbonne Université, Assistance Publique-Hôpitaux de Paris (AP-HP), Hôpital La Pitié-Salpêtrière, Institut E3M, Paris, France.
- Service de Médecine Intensive-Réanimation, Sorbonne Université, AP-HP, Hôpital La Pitié-Salpêtrière, 47-83, boulevard de l'Hôpital, Paris Cedex, 75651, France.
- Institut de Cardiométabolisme et Nutrition (ICAN), Sorbonne Université, INSERM, UMRS_1166-ICAN, Paris, F-75013, France.
| |
Collapse
|
38
|
Ma R, Zhang X, Li R, Dong X, Wang W, Jiang Q, Xiao X, Shi Y, Chen L, Zheng T, Xiang Z, Ren L, Zhou Z, Lei X, Wang J. PLSCR1 suppresses SARS-CoV-2 infection by downregulating cell surface ACE2. J Virol 2025; 99:e0208524. [PMID: 39945535 PMCID: PMC11915802 DOI: 10.1128/jvi.02085-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2024] [Accepted: 01/23/2025] [Indexed: 03/19/2025] Open
Abstract
Type I interferons exert their antiviral effects against SARS-CoV-2 by inducing the expression of interferon-stimulated genes (ISGs), including but not limited to LY6E, CH25H, IFITM2/3, and IFIH1. However, the antiviral effect and underlying mechanisms of action of most ISGs in SARS-CoV-2 infection are not yet fully understood. By screening 109 ISG-knockout cell lines, we identify that phospholipid scramblase 1 (PLSCR1), an interferon-inducible protein, acts as a crucial restriction factor against SARS-CoV-2 infection. Cells lacking PLSCR1 are highly susceptible to SARS-CoV-2 infection. Conversely, overexpression of PLSCR1 inhibits SARS-CoV-2 infection. Depletion of PLSCR1 enhances cellular entry of both pseudotyped and authentic SARS-CoV-2. Mechanistically, PLSCR1 inhibits SARS-CoV-2 entry by specifically downregulating plasma membrane expression of ACE2, the virus's receptor, without affecting the overall levels of ACE2 within the cell. As such, we unraveled previously unappreciated mechanisms by which PLSCR1 exerts its restrictive effect on SARS-CoV-2. These data provide new insights into the interplay between host innate antiviral immunity and SARS-CoV-2 and shed light on novel antiviral therapeutics. IMPORTANCE Phospholipid scramblase 1 (PLSCR1) has been identified as a critical host restriction factor against SARS-CoV-2 infection. In this study, we demonstrated that PLSCR1 inhibited SARS-CoV-2 entry by downregulating the plasma membrane expression of ACE2, the primary receptor for viral entry. Our findings elucidate a novel host-pathogen interaction that not only deepens our understanding of the innate immune response to SARS-CoV-2 but offers potential strategies for therapeutic interventions against COVID-19.
Collapse
Affiliation(s)
- Ruiyi Ma
- NHC Key Laboratory of System Biology of Pathogens, and Christophe Merieux Laboratory National Institute of Pathogen Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
- Key Laboratory of Pathogen Infection Prevention and Control (Peking Union Medical College), Ministry of Education, Beijing, China
| | - Xinyi Zhang
- Biomedical Pioneering Innovation Center, Peking-Tsinghua Center for Life Sciences, Genome Editing Research Center, State Key Laboratory of Protein and Plant Gene Research, School of Life Sciences, Peking University, Beijing, China
| | - Ruonan Li
- NHC Key Laboratory of System Biology of Pathogens, and Christophe Merieux Laboratory National Institute of Pathogen Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
- Key Laboratory of Pathogen Infection Prevention and Control (Peking Union Medical College), Ministry of Education, Beijing, China
| | - Xiaojing Dong
- NHC Key Laboratory of System Biology of Pathogens, and Christophe Merieux Laboratory National Institute of Pathogen Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
- Key Laboratory of Pathogen Infection Prevention and Control (Peking Union Medical College), Ministry of Education, Beijing, China
| | - Wenjing Wang
- NHC Key Laboratory of System Biology of Pathogens, and Christophe Merieux Laboratory National Institute of Pathogen Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
- Key Laboratory of Pathogen Infection Prevention and Control (Peking Union Medical College), Ministry of Education, Beijing, China
| | - Qi Jiang
- NHC Key Laboratory of System Biology of Pathogens, and Christophe Merieux Laboratory National Institute of Pathogen Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
- Key Laboratory of Pathogen Infection Prevention and Control (Peking Union Medical College), Ministry of Education, Beijing, China
| | - Xia Xiao
- NHC Key Laboratory of System Biology of Pathogens, and Christophe Merieux Laboratory National Institute of Pathogen Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
- Key Laboratory of Pathogen Infection Prevention and Control (Peking Union Medical College), Ministry of Education, Beijing, China
| | - Yujin Shi
- NHC Key Laboratory of System Biology of Pathogens, and Christophe Merieux Laboratory National Institute of Pathogen Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
- Key Laboratory of Pathogen Infection Prevention and Control (Peking Union Medical College), Ministry of Education, Beijing, China
| | - Lan Chen
- NHC Key Laboratory of System Biology of Pathogens, and Christophe Merieux Laboratory National Institute of Pathogen Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
- Key Laboratory of Pathogen Infection Prevention and Control (Peking Union Medical College), Ministry of Education, Beijing, China
| | - Tian Zheng
- NHC Key Laboratory of System Biology of Pathogens, and Christophe Merieux Laboratory National Institute of Pathogen Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
- Key Laboratory of Pathogen Infection Prevention and Control (Peking Union Medical College), Ministry of Education, Beijing, China
- Key Laboratory of Respiratory Disease Pathogenomics, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Zichun Xiang
- NHC Key Laboratory of System Biology of Pathogens, and Christophe Merieux Laboratory National Institute of Pathogen Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
- Key Laboratory of Pathogen Infection Prevention and Control (Peking Union Medical College), Ministry of Education, Beijing, China
- Key Laboratory of Respiratory Disease Pathogenomics, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Lili Ren
- NHC Key Laboratory of System Biology of Pathogens, and Christophe Merieux Laboratory National Institute of Pathogen Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
- Key Laboratory of Pathogen Infection Prevention and Control (Peking Union Medical College), Ministry of Education, Beijing, China
- Key Laboratory of Respiratory Disease Pathogenomics, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Zhuo Zhou
- State Key Laboratory of Common Mechanism Research for Major Diseases, Suzhou Institute of Systems Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Suzhou, Jiangsu, China
| | - Xiaobo Lei
- NHC Key Laboratory of System Biology of Pathogens, and Christophe Merieux Laboratory National Institute of Pathogen Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
- Key Laboratory of Pathogen Infection Prevention and Control (Peking Union Medical College), Ministry of Education, Beijing, China
- Key Laboratory of Respiratory Disease Pathogenomics, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jianwei Wang
- NHC Key Laboratory of System Biology of Pathogens, and Christophe Merieux Laboratory National Institute of Pathogen Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| |
Collapse
|
39
|
Framil M, García-Serrano L, Morandeira F, Luchoro JF, Antolí A, Gomez-Vazquez JL, Sierra-Fortuny À, Solanich X. Non-neutralizing anti-type I interferon autoantibodies could increase thrombotic risk in critical COVID-19 patients. Front Immunol 2025; 16:1556731. [PMID: 40165950 PMCID: PMC11955489 DOI: 10.3389/fimmu.2025.1556731] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2025] [Accepted: 02/26/2025] [Indexed: 04/02/2025] Open
Abstract
During the COVID-19 pandemic, approximately 15% of patients with severe COVID-19 pneumonia were reported to have neutralizing anti-type I interferon (IFN) autoantibodies, which impaired the antiviral response and led to a poorer prognosis. However, the physiological impact of non-neutralizing autoantibodies remains unclear. In our cohort of COVID-19 patients admitted to intensive care, the presence of non-neutralizing anti-type I IFN autoantibodies increased the risk of thrombotic complications, likely via a cytokine carrier mechanism, prolonging the half-life of cytokines and dysregulating vascular endothelial function. Previous studies have associated non-neutralizing anti-type I IFN autoantibodies with an increased risk of cardiovascular complications in autoimmune diseases like systemic lupus erythematosus, but their relevance in infectious diseases remains uncertain. Stratifying anti-type I IFN autoantibodies based on their neutralizing capacity may have clinical significance not only in terms of susceptibility to infectious diseases but also in predicting cardiovascular and thrombotic events.
Collapse
Affiliation(s)
- Mario Framil
- Immunology Department, Centre Diagnòstic Biomèdic, Hospital Clínic de Barcelona, Barcelona, Spain
- Immunology Department, Hospital Universitari de Bellvitge, L’Hospitalet de Llobregat, Spain
| | - Lydia García-Serrano
- Immunology Department, Hospital Universitari de Bellvitge, L’Hospitalet de Llobregat, Spain
| | - Francisco Morandeira
- Immunology Department, Hospital Universitari de Bellvitge, L’Hospitalet de Llobregat, Spain
| | - Juan Francisco Luchoro
- Immunology Department, Centre Diagnòstic Biomèdic, Hospital Clínic de Barcelona, Barcelona, Spain
| | - Arnau Antolí
- Bellvitge Biomedical Research Institute (IDIBELL), L’Hospitalet de Llobregat, Spain
- Internal Medicine Department, Hospital Universitari de Bellvitge, L’Hospitalet de Llobregat, Spain
| | | | | | - Xavier Solanich
- Bellvitge Biomedical Research Institute (IDIBELL), L’Hospitalet de Llobregat, Spain
- Internal Medicine Department, Hospital Universitari de Bellvitge, L’Hospitalet de Llobregat, Spain
| |
Collapse
|
40
|
Feng A, Gonzalez MV, Kalaycioglu M, Yin X, Mumau M, Shyamsundar S, Bustamante MS, Chang SE, Dhingra S, Dodig-Crnkovic T, Schwenk JM, Garg T, Yoshizaki K, van Rhee F, Fajgenbaum DC, Utz PJ. Common connective tissue disorder and anti-cytokine autoantibodies are enriched in idiopathic multicentric castleman disease patients. Front Immunol 2025; 16:1528465. [PMID: 40181993 PMCID: PMC11966032 DOI: 10.3389/fimmu.2025.1528465] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2024] [Accepted: 02/24/2025] [Indexed: 04/05/2025] Open
Abstract
Introduction Idiopathic Multicentric Castleman Disease (iMCD) is a polyclonal lymphoproliferative disorder involving cytokine storms that can lead to organ failure and death. The cause of iMCD is unknown, but some clinical evidence suggests an autoimmune etiology. For example, connective tissue disorders (CTDs) and iMCD share many clinical features, and autoantibodies have been anecdotally reported in individual iMCD patients. This study investigates whether common autoantibodies are shared across iMCD patients. Methods We assembled custom bead-based protein arrays consisting of 52 autoantigens traditionally associated with CTDs and 38 full-length cytokines and screened serum samples from 101 iMCD patients for IgG autoantibodies. We also screened samples with a 1,103-plex array of recombinant human protein fragments to identify additional autoantibody targets. Finally, we performed receptor blocking assays on select samples with anti-cytokine autoantibodies (ACAs) identified by array. Results We found that an increased proportion of iMCD patients (47%) tested positive for at least one CTD-associated autoantibody compared to healthy controls (HC) (17%). Commonly detected CTD-associated autoantibodies were associated with myositis and overlap syndromes as well as systemic lupus erythematosus (SLE) and Sjögren's Syndrome (SS). ACAs were also detected in a greater proportion of iMCD patients (38%) compared to HC (10%), while the protein fragment array identified a variety of other autoantibody targets. One iMCD sample tested positive for receptor blocking against interferon-ω (IFNω). Discussion IgG autoantibodies binding autoantigens associated with common CTDs and cytokines are elevated in iMCD patients compared to HC, suggesting that autoimmunity may be involved in iMCD pathogenesis.
Collapse
Affiliation(s)
- Allan Feng
- Department of Medicine, Division of Immunology and Rheumatology, Stanford University School of Medicine, Stanford, CA, United States
- Institute for Immunity, Transplantation and Infection, Stanford University School of Medicine, Stanford, CA, United States
| | - Michael V. Gonzalez
- Center for Cytokine Storm Treatment & Laboratory, University of Pennsylvania, Philadelphia, PA, United States
| | - Muge Kalaycioglu
- Department of Medicine, Division of Immunology and Rheumatology, Stanford University School of Medicine, Stanford, CA, United States
- Institute for Immunity, Transplantation and Infection, Stanford University School of Medicine, Stanford, CA, United States
| | - Xihui Yin
- Department of Medicine, Division of Immunology and Rheumatology, Stanford University School of Medicine, Stanford, CA, United States
- Institute for Immunity, Transplantation and Infection, Stanford University School of Medicine, Stanford, CA, United States
| | - Melanie Mumau
- Center for Cytokine Storm Treatment & Laboratory, University of Pennsylvania, Philadelphia, PA, United States
| | - Saishravan Shyamsundar
- Center for Cytokine Storm Treatment & Laboratory, University of Pennsylvania, Philadelphia, PA, United States
| | - Mateo Sarmiento Bustamante
- Center for Cytokine Storm Treatment & Laboratory, University of Pennsylvania, Philadelphia, PA, United States
| | - Sarah E. Chang
- Department of Medicine, Division of Immunology and Rheumatology, Stanford University School of Medicine, Stanford, CA, United States
- Institute for Immunity, Transplantation and Infection, Stanford University School of Medicine, Stanford, CA, United States
| | - Shaurya Dhingra
- Department of Medicine, Division of Immunology and Rheumatology, Stanford University School of Medicine, Stanford, CA, United States
- Institute for Immunity, Transplantation and Infection, Stanford University School of Medicine, Stanford, CA, United States
| | - Tea Dodig-Crnkovic
- Science for Life Laboratory, Department of Protein Science, KTH Royal Institute of Technology, Stockholm, Sweden
| | - Jochen M. Schwenk
- Science for Life Laboratory, Department of Protein Science, KTH Royal Institute of Technology, Stockholm, Sweden
| | - Tarun Garg
- Myeloma Center, University of Arkansas for Medical Sciences, Little Rock, AR, United States
| | - Kazuyuki Yoshizaki
- Department of Biomolecular Science and Regulation, The Institute of Scientific and Industrial Research, Osaka University, Osaka, Japan
| | - Frits van Rhee
- Myeloma Center, University of Arkansas for Medical Sciences, Little Rock, AR, United States
| | - David C. Fajgenbaum
- Center for Cytokine Storm Treatment & Laboratory, University of Pennsylvania, Philadelphia, PA, United States
| | - Paul J. Utz
- Department of Medicine, Division of Immunology and Rheumatology, Stanford University School of Medicine, Stanford, CA, United States
- Institute for Immunity, Transplantation and Infection, Stanford University School of Medicine, Stanford, CA, United States
| |
Collapse
|
41
|
Khatun O, Kaur S, Tripathi S. Anti-interferon armamentarium of human coronaviruses. Cell Mol Life Sci 2025; 82:116. [PMID: 40074984 PMCID: PMC11904029 DOI: 10.1007/s00018-025-05605-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2024] [Revised: 12/15/2024] [Accepted: 01/23/2025] [Indexed: 03/14/2025]
Abstract
Cellular innate immune pathways are formidable barriers against viral invasion, creating an environment unfavorable for virus replication. Interferons (IFNs) play a crucial role in driving and regulating these cell-intrinsic innate antiviral mechanisms through the action of interferon-stimulated genes (ISGs). The host IFN response obstructs viral replication at every stage, prompting viruses to evolve various strategies to counteract or evade this response. Understanding the interplay between viral proteins and cell-intrinsic IFN-mediated immune mechanisms is essential for developing antiviral and anti-inflammatory strategies. Human coronaviruses (HCoVs), including SARS-CoV-2, MERS-CoV, SARS-CoV, and seasonal coronaviruses, encode a range of proteins that, through shared and distinct mechanisms, inhibit IFN-mediated innate immune responses. Compounding the issue, a dysregulated early IFN response can lead to a hyper-inflammatory immune reaction later in the infection, resulting in severe disease. This review provides a brief overview of HCoV replication and a detailed account of its interaction with host cellular innate immune pathways regulated by IFN.
Collapse
Affiliation(s)
- Oyahida Khatun
- Emerging Viral Pathogens Laboratory, Centre for Infectious Disease Research, Indian Institute of Science, Bengaluru, India
- Microbiology & Cell Biology Department, Biological Sciences Division, Indian Institute of Science, Bengaluru, India
| | - Sumandeep Kaur
- Emerging Viral Pathogens Laboratory, Centre for Infectious Disease Research, Indian Institute of Science, Bengaluru, India
- Microbiology & Cell Biology Department, Biological Sciences Division, Indian Institute of Science, Bengaluru, India
| | - Shashank Tripathi
- Emerging Viral Pathogens Laboratory, Centre for Infectious Disease Research, Indian Institute of Science, Bengaluru, India.
- Microbiology & Cell Biology Department, Biological Sciences Division, Indian Institute of Science, Bengaluru, India.
| |
Collapse
|
42
|
Lopes da Silva VG, Schmitz GJH, Sullivan KE, Barbate J, de Haro Azinar MI, Aranda CS, de Moraes-Pinto MI. Enhanced T-cell immunity and lower humoral responses following 5-dose SARS-CoV-2 vaccination in patients with inborn errors of immunity compared with healthy controls. Front Immunol 2025; 16:1538453. [PMID: 40114918 PMCID: PMC11922935 DOI: 10.3389/fimmu.2025.1538453] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2024] [Accepted: 02/06/2025] [Indexed: 03/22/2025] Open
Abstract
Objective Patients with Inborn Errors of Immunity (IEI) are at higher risk of severe SARS-CoV-2 infection. We evaluated humoral and cellular responses to COVID-19 vaccines in Brazilian patients with IEI and healthy controls. Methods Fifty-five patients with IEI (13-61 years) and 60 controls (13-71 years) received inactivated SARS-CoV-2 (CoronaVac), non-replicating virus-vectored (ChAdOx1 nCoV-19, AstraZeneca) or monovalent mRNA (Original strain of BNT162b2, Pfizer-BioNTech) and bivalent mRNA (Original/Omicron BA.1, Pfizer-BioNTech) vaccines and were sampled five times. Diagnoses included common variable immunodeficiency (n=25), specific antibody deficiency (n=9), ataxia-telangiectasia (n=5), X-linked agammaglobulinemia (n=4), PIK3CD-related disorders (n=4), hyper-IgM syndrome (n=4), combined immunodeficiency (n=3), and STAT1 gain-of-function (n=1). Humoral immunity was assessed via multiplex microarray for Spike, Nucleocapsid, RBD-Wuhan, RBD-Delta, RBD-BA.1, RBD-BA.2 and RBD-BA.5 neutralizing antibodies. T-cell responses to Spike and Nucleocapsid were assessed using ELISpot. Results Patients with IEI exhibited significantly lower levels of Nucleocapsid and RBD-neutralizing antibodies (p < 0.05). Notable differences in RBD-BA.2 (p = 0.008) and IgG-Nucleocapsid (p = 0.010) levels emerged over time. T-cell responses to Spike were stronger in patients with IEI post-booster (405 vs. 149 spot-forming cells/million PBMC; p = 0.002). Both groups showed enhanced Nucleocapsid-specific cellular responses over time (p = 0.017). COVID-19 hospitalization rates among patients with IEI with SARS-CoV-2 diagnosis dropped from 33.3% to zero after the first booster dose. Conclusions While humoral responses to SARS-CoV-2 vaccines were weaker in patients with IEI, their cellular immunity was similar to controls. Boosters enhanced both humoral and cellular responses. After completion of the vaccination protocol, none of the patients with IEI were hospitalized with COVID-19. Robust T-cell responses may play a critical role in protecting patients with IEI from severe COVID-19 and mortality.
Collapse
Affiliation(s)
| | | | - Kathleen E. Sullivan
- The Children’s Hospital of Philadelphia, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Júlia Barbate
- Departamento de Pediatria, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Maria Izabel de Haro Azinar
- Departamento de Pediatria, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Carolina Sanchez Aranda
- Departamento de Pediatria, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, Brazil
| | | |
Collapse
|
43
|
IJspeert H, Edwards ESJ, O'Hehir RE, Dalm VASH, van Zelm MC. Update on inborn errors of immunity. J Allergy Clin Immunol 2025; 155:740-751. [PMID: 39724969 DOI: 10.1016/j.jaci.2024.12.1075] [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: 11/15/2024] [Revised: 12/10/2024] [Accepted: 12/13/2024] [Indexed: 12/28/2024]
Abstract
Ever since the first description of an inherited immunodeficiency in 1952 in a boy with gammaglobulin deficiency, new insights have progressed rapidly in disorders that are now referred to as inborn errors of immunity. In a field where fundamental molecular biology, genetics, immune signaling, and clinical care are tightly intertwined, 2022-24 saw a multitude of advances. Here we report a selection of research updates with a main focus on (1) diagnosis and screening, (2) new genetic defects, (3) susceptibility to severe coronavirus disease 2019 infection and impact of vaccination, and (4) treatment. Importantly, new pathogenic insights more rapidly affect treatment outcomes, either through an earlier and more precise diagnosis or through implementation of novel, personalized treatment. The field is growing rapidly, so awareness, communication, and collaboration are key to improving treatment outcomes.
Collapse
Affiliation(s)
- Hanna IJspeert
- Department of Clinical Genetics, Erasmus MC, University Medical Center, Rotterdam, The Netherlands
| | - Emily S J Edwards
- Department of Immunology, School of Translational Medicine, Monash University, Melbourne, Australia; Allergy, Asthma and Clinical Immunology, Alfred Health, Melbourne, Australia; Jeffrey Modell Center, Melbourne, Australia
| | - Robyn E O'Hehir
- Department of Immunology, School of Translational Medicine, Monash University, Melbourne, Australia; Allergy, Asthma and Clinical Immunology, Alfred Health, Melbourne, Australia; Jeffrey Modell Center, Melbourne, Australia
| | - Virgil A S H Dalm
- Department of Immunology, Erasmus MC, University Medical Center, Rotterdam, The Netherlands; Department of Internal Medicine, Division of Allergy & Clinical Immunology, Erasmus MC, University Medical Center, Rotterdam, The Netherlands
| | - Menno C van Zelm
- Department of Immunology, Erasmus MC, University Medical Center, Rotterdam, The Netherlands; Department of Immunology, School of Translational Medicine, Monash University, Melbourne, Australia; Allergy, Asthma and Clinical Immunology, Alfred Health, Melbourne, Australia; Jeffrey Modell Center, Melbourne, Australia.
| |
Collapse
|
44
|
Boisson-Dupuis S, Bastard P, Béziat V, Bustamante J, Cobat A, Jouanguy E, Puel A, Rosain J, Zhang Q, Zhang SY, Boisson B. The monogenic landscape of human infectious diseases. J Allergy Clin Immunol 2025; 155:768-783. [PMID: 39724971 PMCID: PMC11875930 DOI: 10.1016/j.jaci.2024.12.1078] [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: 06/24/2024] [Revised: 12/18/2024] [Accepted: 12/20/2024] [Indexed: 12/28/2024]
Abstract
The spectrum of known monogenic inborn errors of immunity is growing, with certain disorders underlying a specific and narrow range of infectious diseases. These disorders reveal the core mechanisms by which these infections occur in various settings, including inherited and acquired immunodeficiencies, thereby delineating the essential mechanisms of protective immunity to the corresponding pathogens. These findings also have medical implications, facilitating diagnosis and improving the management of individuals at risk of disease.
Collapse
Affiliation(s)
- Stéphanie Boisson-Dupuis
- St Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, The Rockefeller University, New York, NY; Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM U1163, Paris, France; Paris Cité University, Imagine Institute, Paris, France
| | - Paul Bastard
- St Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, The Rockefeller University, New York, NY; Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM U1163, Paris, France; Paris Cité University, Imagine Institute, Paris, France; Pediatric Hematology-Immunology and Rheumatology Unit, Necker Hospital for Sick Children, Assistance Publique-Hôpitaux de Paris, Paris, France
| | - Vivien Béziat
- St Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, The Rockefeller University, New York, NY; Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM U1163, Paris, France; Paris Cité University, Imagine Institute, Paris, France
| | - Jacinta Bustamante
- St Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, The Rockefeller University, New York, NY; Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM U1163, Paris, France; Paris Cité University, Imagine Institute, Paris, France; Study Center for Primary Immunodeficiencies, Necker Hospital for Sick Children, Assistance Publique-Hôpitaux de Paris, Paris, France
| | - Aurélie Cobat
- St Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, The Rockefeller University, New York, NY; Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM U1163, Paris, France; Paris Cité University, Imagine Institute, Paris, France
| | - Emmanuelle Jouanguy
- St Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, The Rockefeller University, New York, NY; Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM U1163, Paris, France; Paris Cité University, Imagine Institute, Paris, France
| | - Anne Puel
- St Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, The Rockefeller University, New York, NY; Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM U1163, Paris, France; Paris Cité University, Imagine Institute, Paris, France
| | - Jérémie Rosain
- St Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, The Rockefeller University, New York, NY; Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM U1163, Paris, France; Paris Cité University, Imagine Institute, Paris, France; Study Center for Primary Immunodeficiencies, Necker Hospital for Sick Children, Assistance Publique-Hôpitaux de Paris, Paris, France
| | - Qian Zhang
- St Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, The Rockefeller University, New York, NY; Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM U1163, Paris, France; Paris Cité University, Imagine Institute, Paris, France
| | - Shen-Ying Zhang
- St Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, The Rockefeller University, New York, NY; Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM U1163, Paris, France; Paris Cité University, Imagine Institute, Paris, France
| | - Bertrand Boisson
- St Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, The Rockefeller University, New York, NY; Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM U1163, Paris, France; Paris Cité University, Imagine Institute, Paris, France.
| |
Collapse
|
45
|
Alvarez-Sierra D, Martínez-Gallo M, Sánchez-Montalvá A, Fernández-Sanmartín M, Colobran R, Espinosa-Pereiro J, Poyatos-Canton E, Zurera-Egea C, Sánchez-Pla A, Violan C, Parra R, Alzayat H, Vivancos A, Morandeira-Rego F, Urban-Vargas B, Martínez-Cáceres E, Hernández-González M, Bas-Minguet J, Katsikis PD, Teniente-Serra A, Pujol-Borrell R. The immune response to SARS-CoV-2 in COVID-19 as a recall response susceptible to immune imprinting: A prospective cohort study. Clin Immunol 2025; 272:110429. [PMID: 39842683 DOI: 10.1016/j.clim.2025.110429] [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: 11/20/2024] [Revised: 01/12/2025] [Accepted: 01/14/2025] [Indexed: 01/24/2025]
Abstract
The antibody response to SARS-CoV-2 does not follow the immunoglobulin isotype pattern of primary responses, conflicting with the current interpretation of COVID-19. METHODS Prospective cohort study of 191 SARS-CoV-2 infection cases and 44 controls from the second wave of COVID-19. The study stratified patients by severity and analyzed the trajectories of SARS-CoV-2 antibodies and multiple immune variables. RESULTS Isotype-specific antibody time course profiles to SARS-CoV-2 revealed a pattern of recall response in 94.2 % of cases. The time course profiles of plasmablasts, B cells, cTfh high-resolution subsets, and cytokines indicated a secondary response. The transcriptomic data showed that this cohort is strictly comparable to contemporary cohorts. CONCLUSIONS In most cases, the immune response to SARS-CoV-2 is a recall response. This constitutes a favorable scenario for most COVID-19 cases to be subjected to immune imprinting by endemic coronavirus, which, in turn, can influence the immune response to SARS-CoV-2.
Collapse
Affiliation(s)
- Daniel Alvarez-Sierra
- Translational Immunology Research Group, Vall d'Hebron Research Institute (VHIR), Campus Vall d'Hebron, Barcelona, Spain.
| | - Mónica Martínez-Gallo
- Translational Immunology Research Group, Vall d'Hebron Research Institute (VHIR), Campus Vall d'Hebron, Barcelona, Spain; Immunology Department, Hospital Universitari Vall d'Hebron, Campus Vall d'Hebron, Barcelona, Spain; Department of Cell Biology, Physiology, and Immunology, Universitat Autònoma Barcelona, Campus Vall d'Hebron and Campus Bellaterra, Barcelona, Spain
| | - Adrián Sánchez-Montalvá
- Infectious Disease Department, Hospital Universitari Vall d'Hebron, Campus Vall d'Hebró, Barcelona, Spain; International Health Program, Institut Català de la Salut, Vall d'Hebron Research Institute (VHIR), Campus Vall d'Hebron, Barcelona, Spain; Centro de Investigación Biomédica en Red de Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III, Campus Vall d'Hebron, Barcelona, Spain
| | - Marco Fernández-Sanmartín
- Flow Cytometry Laboratory, Germans Trias i Pujol Research Institute (IGTP), Badalona, Barcelona, Spain
| | - Roger Colobran
- Translational Immunology Research Group, Vall d'Hebron Research Institute (VHIR), Campus Vall d'Hebron, Barcelona, Spain; Immunology Department, Hospital Universitari Vall d'Hebron, Campus Vall d'Hebron, Barcelona, Spain; Department of Cell Biology, Physiology, and Immunology, Universitat Autònoma Barcelona, Campus Vall d'Hebron and Campus Bellaterra, Barcelona, Spain
| | - Juan Espinosa-Pereiro
- Infectious Disease Department, Hospital Universitari Vall d'Hebron, Campus Vall d'Hebró, Barcelona, Spain; International Health Program, Institut Català de la Salut, Vall d'Hebron Research Institute (VHIR), Campus Vall d'Hebron, Barcelona, Spain; Centro de Investigación Biomédica en Red de Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III, Campus Vall d'Hebron, Barcelona, Spain
| | - Elísabet Poyatos-Canton
- Immunology Division, Bellvitge University Hospital, Hospitalet de Llobregat, Barcelona, Spain
| | - Coral Zurera-Egea
- Immunology and Inflammation Research Group, Germans Trias i Pujol Research Institute (IGTP), Badalona, Barcelona, Spain
| | - Alex Sánchez-Pla
- Bioinformatics and Statistics Group, University of Barcelona, Barcelona, Spain
| | - Concepción Violan
- Unitat de Suport a la Recerca Metropolitana Nord, Institut Universitari d'Investigació en Atenció Primària Jordi Gol (IDIAP Jordi Gol), Mataró, Barcelona, Spain
| | - Rafael Parra
- Banc de Sang i Teixits, Hospital Universitari Vall Hebron, Barcelona, Spain
| | - Hammad Alzayat
- Flow Cytometry Laboratory, Germans Trias i Pujol Research Institute (IGTP), Badalona, Barcelona, Spain
| | - Ana Vivancos
- Tumor Immunology and Immunotherapy Group, Vall Hebron Institut Oncology (VHIO), Campus Vall d'Hebron, Barcelona, Spain
| | | | - Blanca Urban-Vargas
- Immunology Division, Bellvitge University Hospital, Hospitalet de Llobregat, Barcelona, Spain
| | - Eva Martínez-Cáceres
- Department of Cell Biology, Physiology, and Immunology, Universitat Autònoma Barcelona, Campus Vall d'Hebron and Campus Bellaterra, Barcelona, Spain; Immunology Department, Hospital Universitari Germans Trias I Pujol, Barcelona, Spain; Immunology and Inflammation Research Group, Germans Trias i Pujol Research Institute (IGTP), Badalona, Barcelona, Spain
| | - Manuel Hernández-González
- Translational Immunology Research Group, Vall d'Hebron Research Institute (VHIR), Campus Vall d'Hebron, Barcelona, Spain; Immunology Department, Hospital Universitari Vall d'Hebron, Campus Vall d'Hebron, Barcelona, Spain; Department of Cell Biology, Physiology, and Immunology, Universitat Autònoma Barcelona, Campus Vall d'Hebron and Campus Bellaterra, Barcelona, Spain
| | - Jordi Bas-Minguet
- Immunology Division, Bellvitge University Hospital, Hospitalet de Llobregat, Barcelona, Spain
| | - Peter D Katsikis
- Dept. of Immunology, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Aina Teniente-Serra
- Department of Cell Biology, Physiology, and Immunology, Universitat Autònoma Barcelona, Campus Vall d'Hebron and Campus Bellaterra, Barcelona, Spain; Immunology Department, Hospital Universitari Germans Trias I Pujol, Barcelona, Spain; Immunology and Inflammation Research Group, Germans Trias i Pujol Research Institute (IGTP), Badalona, Barcelona, Spain
| | - Ricardo Pujol-Borrell
- Immunology Department, Hospital Universitari Vall d'Hebron, Campus Vall d'Hebron, Barcelona, Spain; Department of Cell Biology, Physiology, and Immunology, Universitat Autònoma Barcelona, Campus Vall d'Hebron and Campus Bellaterra, Barcelona, Spain; Tumor Immunology and Immunotherapy Group, Vall Hebron Institut Oncology (VHIO), Campus Vall d'Hebron, Barcelona, Spain.
| |
Collapse
|
46
|
Sahli W, Vitte J, Desnues B. Eosinophils and COVID-19: Insights into immune complexity and vaccine safety. Clin Transl Allergy 2025; 15:e70050. [PMID: 40120088 PMCID: PMC11929522 DOI: 10.1002/clt2.70050] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2024] [Revised: 02/23/2025] [Accepted: 03/10/2025] [Indexed: 03/25/2025] Open
Abstract
BACKGROUND COVID-19 exhibits a variety of symptoms and may lead to multi-organ failure and death. This clinical complexity is exacerbated by significant immune dysregulation affecting nearly all cells of the innate and adaptive immune system. Granulocytes, including eosinophils, are affected by SARS-CoV-2. OBJECTIVES Eosinophil responses remain poorly understood despite early recognition of eosinopenia as a hallmark feature of COVID-19 severity. RESULTS The heterogeneous nature of eosinophil responses categorizes them as dual-function cells with contradictory effects. Eosinophil activation can suppress virus-induced inflammation by releasing type 2 cytokines like IL-13 and granular proteins with antiviral action such as eosinophil-derived neurotoxins and eosinophil cationic protein, and also by acting as antigen-presenting cells. In contrast, eosinophil accumulation in the lungs can induce tissue damage triggered by cytokines or hormones like IFN-γ and leptin. Additionally, they can affect adaptive immune functions by interacting with T cells through direct formation of membrane complexes or soluble mediator action. Individuals with allergic disorders who have elevated levels of eosinophils in tissues and blood, such as asthma, do not appear to be at an increased risk of developing severe COVID-19 following SARS-CoV-2 infection. However, the SARS-CoV-2 vaccine appears to be associated with complications and eosinophilic infiltrate-induced immunopathogenicity, which can be mitigated by corticosteroid, anti-histamines and anti-IL-5 therapy and avoided by modifying adjuvants or excipients. CONCLUSION This review highlights the importance of eosinophils in COVID-19 and contributes to a better understanding of their role during natural infection and vaccination.
Collapse
Affiliation(s)
- Wided Sahli
- Aix Marseille UniversityMEPHIMarseilleFrance
- IHU‐Méditerranée InfectionMarseilleFrance
| | - Joana Vitte
- Laboratory of ImmunologyUniversity Hospital of ReimsReimsFrance
- INSERM UMR‐S 1250 P3CELLUniversity of ReimsReimsFrance
| | - Benoit Desnues
- Aix Marseille UniversityMEPHIMarseilleFrance
- IHU‐Méditerranée InfectionMarseilleFrance
| |
Collapse
|
47
|
Galipeau Y, Castonguay N, McCluskie PS, Sonoda MT, Keeshan A, Collins E, Arnold C, Pelchat M, Burns K, Cooper C, Langlois M. Autoantibodies targeting angiotensin-converting enzyme 2 are prevalent and not induced by SARS-CoV-2 infection. FASEB J 2025; 39:e70390. [PMID: 39950298 PMCID: PMC11826374 DOI: 10.1096/fj.202402694r] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2024] [Revised: 01/07/2025] [Accepted: 02/03/2025] [Indexed: 02/16/2025]
Abstract
Clinical outcomes resulting from SARS-CoV-2 infection vary widely, ranging from asymptomatic cases to the development of mild to severe respiratory illness, and in some instances, chronic lingering disease and mortality. The underlying biological mechanisms driving this wide spectrum of pathogenicity among certain individuals and demographics remain elusive. Autoantibodies have emerged as potential contributors to the severity of COVID-19. Although preliminary reports have suggested the induction of antibodies targeting Angiotensin-Converting Enzyme II (ACE2) post-infection, this assertion lacks confirmation in large-scale studies. In this study, our objective is to comprehensively characterize and quantify the prevalence and expression levels of autoantibodies directed against ACE2 in a sizable cohort (n = 464). Our findings reveal that ACE2-reactive IgM antibodies are the most prevalent, with an overall seroprevalence of 18.8%, followed by IgG at 10.3% and IgA at 6.3%. Longitudinal analysis of individuals with multiple blood draws showed stable ACE2 IgG and IgA levels over time. Upon stratifying individuals based on molecular testing for SARS-CoV-2 or serological evidence of past infection, no significant differences were observed between groups. Functional assessment of ACE2 autoantibodies demonstrated that they are non-neutralizing and failed to inhibit spike-ACE2 interaction or affect the enzymatic activity of ACE2. Our results highlight that ACE2 autoantibodies are prevalent in the general population and were not induced by SARS-CoV-2 infection in our cohort. Notably, we found no substantiated evidence supporting a direct role for ACE2 autoantibodies in SARS-CoV-2 pathogenesis.
Collapse
Affiliation(s)
- Yannick Galipeau
- Department of Biochemistry, Microbiology & Immunology, Faculty of MedicineUniversity of OttawaOttawaOntarioCanada
| | - Nicolas Castonguay
- Department of Biochemistry, Microbiology & Immunology, Faculty of MedicineUniversity of OttawaOttawaOntarioCanada
| | - Pauline S. McCluskie
- Department of Biochemistry, Microbiology & Immunology, Faculty of MedicineUniversity of OttawaOttawaOntarioCanada
| | | | - Alexa Keeshan
- School of Epidemiology and Public Health, Faculty of MedicineUniversity of OttawaOttawaOntarioCanada
- Clinical EpidemiologyOttawa Hospital Research InstituteOttawaOntarioCanada
| | - Erin Collins
- School of Epidemiology and Public Health, Faculty of MedicineUniversity of OttawaOttawaOntarioCanada
- Clinical EpidemiologyOttawa Hospital Research InstituteOttawaOntarioCanada
| | - Corey Arnold
- Department of Biochemistry, Microbiology & Immunology, Faculty of MedicineUniversity of OttawaOttawaOntarioCanada
| | - Martin Pelchat
- Department of Biochemistry, Microbiology & Immunology, Faculty of MedicineUniversity of OttawaOttawaOntarioCanada
- Centre for Infection, Immunity and Inflammation (CI3)University of OttawaOttawaOntarioCanada
| | - Kevin Burns
- Division of Nephrology, Department of MedicineUniversity of OttawaOttawaOntarioCanada
- Ottawa Hospital Research InstituteOttawaOntarioCanada
| | - Curtis Cooper
- School of Epidemiology and Public Health, Faculty of MedicineUniversity of OttawaOttawaOntarioCanada
- Ottawa Hospital Research InstituteOttawaOntarioCanada
- Division of Infectious Diseases, Department of MedicineUniversity of OttawaOttawaOntarioCanada
| | - Marc‐André Langlois
- Department of Biochemistry, Microbiology & Immunology, Faculty of MedicineUniversity of OttawaOttawaOntarioCanada
- Centre for Infection, Immunity and Inflammation (CI3)University of OttawaOttawaOntarioCanada
| |
Collapse
|
48
|
Zhu L, Li L, Wu J. FcRn inhibitors: Transformative advances and significant impacts on IgG-mediated autoimmune diseases. Autoimmun Rev 2025; 24:103719. [PMID: 39672251 DOI: 10.1016/j.autrev.2024.103719] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2024] [Revised: 12/06/2024] [Accepted: 12/06/2024] [Indexed: 12/15/2024]
Abstract
Pathogenic IgG autoantibodies play a crucial role in the pathogenesis of autoimmune diseases, and removal of pathogenic IgG autoantibodies is an important therapeutic approach and tool for such diseases. The neonatal Fc receptor (FcRn) interacts with IgG and protects it from lysosomal degradation. FcRn inhibitors accelerate the clearance of IgG antibodies, including pathogenic IgG autoantibodies, by targeting and blocking the binding of FcRn to IgG. Theoretically, FcRn inhibitors can be applied for the treatment of IgG-mediated autoimmune diseases. With successful completion of multiple relevant clinical trials, key evidence-based data have been provided for FcRn inhibitors in the treatment of IgG-mediated autoimmune diseases, and several FcRn inhibitors have been approved for these indications. Additional trials are being planned or conducted. This review examines all available high-quality clinical trials of FcRn inhibitors assessing IgG-mediated autoimmune diseases.
Collapse
Affiliation(s)
- Lina Zhu
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, 450052 Zhengzhou, China
| | - Lanjun Li
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, 450052 Zhengzhou, China
| | - Jun Wu
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, 450052 Zhengzhou, China..
| |
Collapse
|
49
|
Dai J, He F, Chen Q, Li Q, Zhao L, Du Y. Animal models of post-acute COVID-19 syndrome: a call for longitudinal animal studies. Front Immunol 2025; 16:1521029. [PMID: 40079013 PMCID: PMC11897041 DOI: 10.3389/fimmu.2025.1521029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2024] [Accepted: 01/29/2025] [Indexed: 03/14/2025] Open
Abstract
Animal models are indispensable for unraveling the mechanisms underlying post-acute sequelae of COVID-19 (PASC). This review evaluates recent research on PASC-related perturbations in animal models, drawing comparisons with clinical findings. Despite the limited number of studies on post-COVID conditions, particularly those extending beyond three months, these studies provide valuable insights. Three hallmark features of PASC-lung fibrosis, hyperglycemia, and neurological sequelae-have been successfully replicated in animal models, paving the way for mechanistic discoveries and future medical interventions. Although most studies have reported post-COVID conditions within 14-60 days post-infection, they still offer critical reference for future long-term research. This review also explores potential mechanisms of persisting immune misfiring, a key factor in the chronicity of PASC symptoms. Moreover, challenges in modeling PASC are also discussed, including the limited genetic diversity in inbred animal strains and difficulties in accurately identifying PASC-affected individuals. To address these issues, we propose methodological improvements, such as comparing individual animal parameters with control averages and incorporating genetically diverse populations like collaborative cross models. These strategies will enhance the identification and characterization of PASC endotypes in animal studies. By integrating findings from animal models with clinical manifestations of PASC, future research can provide more valuable insights into its mechanisms and support the development of effective therapeutic strategies. Finally, we emphasize the urgent need for longitudinal studies in animal models to fully uncover the mechanisms driving PASC and guide interventions to mitigate its public health impact.
Collapse
Affiliation(s)
- Jingyi Dai
- Department of Public Laboratory, The Third People’s Hospital of Kunming City/Infectious Disease Clinical Medical Center of Yunnan Province, Kunming, Yunnan, China
- International Research Fellow, Prince of Songkla University, Hat Yai, Songkhla, Thailand
| | - Fanyi He
- Department of Public Laboratory, The Third People’s Hospital of Kunming City/Infectious Disease Clinical Medical Center of Yunnan Province, Kunming, Yunnan, China
| | - Qian Chen
- Department of Public Laboratory, The Third People’s Hospital of Kunming City/Infectious Disease Clinical Medical Center of Yunnan Province, Kunming, Yunnan, China
| | - Qiujing Li
- Department of Public Laboratory, The Third People’s Hospital of Kunming City/Infectious Disease Clinical Medical Center of Yunnan Province, Kunming, Yunnan, China
| | - Liping Zhao
- Department of Public Laboratory, The Third People’s Hospital of Kunming City/Infectious Disease Clinical Medical Center of Yunnan Province, Kunming, Yunnan, China
| | - Yingrong Du
- Department of Public Laboratory, The Third People’s Hospital of Kunming City/Infectious Disease Clinical Medical Center of Yunnan Province, Kunming, Yunnan, China
| |
Collapse
|
50
|
Hromić-Jahjefendić A, Sezer A, Mahmuljin I. The impact of COVID-19 on autoimmune diseases. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2025; 213:315-345. [PMID: 40246348 DOI: 10.1016/bs.pmbts.2025.01.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/19/2025]
Abstract
Various autoantibodies, such as antinuclear antibodies (ANA), anti-Ro/SSA, rheumatoid factor, lupus anticoagulant, and antibodies against interferon type I (IFN-I), have been frequently detected in COVID-19 patients, indicating a significant prevalence of autoimmune reactions following viral exposure. Additionally, the identification of human proteins with structural similarities to SARS-CoV-2 peptides as potential autoantigens underscores the complex interplay between the virus and the immune system in triggering autoimmunity. The chapter discusses probable pathways contributing to COVID-19-related autoimmunity, including bystander activation due to hyperinflammatory states, viral persistence, and the formation of neutrophil extracellular traps. These mechanisms illuminate a spectrum of autoimmune-related symptoms that can manifest, ranging from organ-specific to systemic autoimmune and inflammatory diseases. Importantly, there is emerging evidence of de novo autoimmunity arising after COVID-19 infection or vaccination, where new autoimmune conditions develop in previously healthy individuals. While various COVID-19 vaccines have received emergency use authorization, concerns regarding potential autoimmune side effects persist. Ongoing research is crucial to clarify these relationships and enhance our understanding of the risks associated with COVID-19 infections and vaccinations.
Collapse
Affiliation(s)
- Altijana Hromić-Jahjefendić
- Department of Genetics and Bioengineering, Faculty of Engineering and Natural Sciences, International University of Sarajevo, Sarajevo, Bosnia and Herzegovina.
| | - Abas Sezer
- Department of Genetics and Bioengineering, Faculty of Engineering and Natural Sciences, International University of Sarajevo, Sarajevo, Bosnia and Herzegovina
| | - Irma Mahmuljin
- Association of Biologists in Bosnia and Herzegovina, Sarajevo, Bosnia and Herzegovina
| |
Collapse
|