201
|
Saheb Sharif-Askari N, Saheb Sharif-Askari F, Bayram OS, Hafezi S, Alsayed HAH, Kasim F, Mdkhana B, Selvakumar B, Alsafar HS, Halwani R. Salivary autoantibodies to type I IFNs: Mirror plasma levels, predispose to severe COVID-19, and enhance feasibility for clinical screening. Heart Lung 2024; 66:31-36. [PMID: 38547583 DOI: 10.1016/j.hrtlng.2024.03.005] [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: 01/05/2024] [Revised: 03/06/2024] [Accepted: 03/18/2024] [Indexed: 05/14/2024]
Abstract
BACKGROUND Autoantibodies have been demonstrated to dampen the interferon (IFN) response in viral infections. Elevated levels of these preexisting autoantibodies (aAbs) decrease basal interferon levels, increasing susceptibility to severe infections. OBJECTIVES This study aimed to evaluate the prevalence of type I IFN aAbs in both plasma and saliva from COVID-19 patients, analyze their neutralizing activity, and examine their associations with clinical outcomes, including the need for mechanical ventilation and in-hospital mortality. METHODS Prospective analyses of patients admitted to intensive care units in three UAE hospitals from June 2020 to March 2021 were performed to measure aAbs using enzyme-linked immunosorbent assay (ELISA), assess aAbs activity via neutralization assays, and correlate aAbs with clinical outcomes. RESULTS Type I IFN aAbs (α2 and/or ω) were measured in plasma samples from 213 ICU patients, and positive results were obtained for 20 % (n = 42) of the patients, with half exhibiting neutralizing activity. Saliva samples from a subgroup of 24 patients reflected plasma levels. In multivariate regression analyses, presence of type I IFN aAbs was associated with a higher need for mechanical ventilation (OR 2.58; 95 % CI 1.07-6.22) and greater in-hospital mortality (OR 2.40; 95 % CI 1.13 - 5.07; P = 0.022). Similarly, positive neutralizing aAbs (naAbs) were associated with a greater need for mechanical ventilation (OR 4.96; 95 % CI 1.12-22.07; P = 0.035) and greater odds of in-hospital mortality (OR 2.87; 95 % CI 1.05-7.89; P = 0.041). CONCLUSIONS Type I IFN autoantibodies can be detected in noninvasive saliva samples, alongside conventional plasma samples, from COVID-19 patients and are associated with worse outcomes, such as greater mechanical ventilation needs and in-hospital mortality.
Collapse
Affiliation(s)
- Narjes Saheb Sharif-Askari
- Research Institute for Medical and Health Sciences, University of Sharjah, Sharjah, United Arab Emirates; Department of Clinical Sciences, College of Medicine, University of Sharjah, Sharjah, United Arab Emirates
| | - Fatemeh Saheb Sharif-Askari
- Research Institute for Medical and Health Sciences, University of Sharjah, Sharjah, United Arab Emirates; Department of pharmacy practice and pharmacotherapeutics, College of pharmacy, University of Sharjah, Sharjah, United Arab Emirates
| | - Ola Salam Bayram
- Department of Clinical Sciences, College of Medicine, University of Sharjah, Sharjah, United Arab Emirates
| | - Shirin Hafezi
- Research Institute for Medical and Health Sciences, University of Sharjah, Sharjah, United Arab Emirates
| | - Hawra Ali Hussain Alsayed
- Department of Pharmacy, Rashid Hospital, Dubai Academic Health Corporation, Dubai, United Arab Emirates
| | - Fathima Kasim
- Department of internal medicine, Zulekha Hospitals, Dubai, United Arab Emirates
| | - Bushra Mdkhana
- Research Institute for Medical and Health Sciences, University of Sharjah, Sharjah, United Arab Emirates
| | - Balachandar Selvakumar
- Research Institute for Medical and Health Sciences, University of Sharjah, Sharjah, United Arab Emirates
| | - Habiba S Alsafar
- Center for Biotechnology, Khalifa University of Science and Technology, Abu Dhabi, United Arab Emirates; Department of Biomedical Engineering, College of Engineering, Khalifa University of Science and Technology, Abu Dhabi, United Arab Emirates; Department of Genetics and Molecular Biology, College of Medicine and Health Sciences, Khalifa University of Science and Technology, Abu Dhabi, United Arab Emirates
| | - Rabih Halwani
- Research Institute for Medical and Health Sciences, University of Sharjah, Sharjah, United Arab Emirates; Department of Clinical Sciences, College of Medicine, University of Sharjah, Sharjah, United Arab Emirates; Prince Abdullah Ben Khaled Celiac Disease Research Chair, department of pediatrics, Faculty of Medicine, King Saud University, Saudi Arabia.
| |
Collapse
|
202
|
Malireddi RKS, Sharma BR, Kanneganti TD. Innate Immunity in Protection and Pathogenesis During Coronavirus Infections and COVID-19. Annu Rev Immunol 2024; 42:615-645. [PMID: 38941608 PMCID: PMC11373870 DOI: 10.1146/annurev-immunol-083122-043545] [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] [Indexed: 06/30/2024]
Abstract
The COVID-19 pandemic was caused by the recently emerged β-coronavirus SARS-CoV-2. SARS-CoV-2 has had a catastrophic impact, resulting in nearly 7 million fatalities worldwide to date. The innate immune system is the first line of defense against infections, including the detection and response to SARS-CoV-2. Here, we discuss the innate immune mechanisms that sense coronaviruses, with a focus on SARS-CoV-2 infection and how these protective responses can become detrimental in severe cases of COVID-19, contributing to cytokine storm, inflammation, long-COVID, and other complications. We also highlight the complex cross talk among cytokines and the cellular components of the innate immune system, which can aid in viral clearance but also contribute to inflammatory cell death, cytokine storm, and organ damage in severe COVID-19 pathogenesis. Furthermore, we discuss how SARS-CoV-2 evades key protective innate immune mechanisms to enhance its virulence and pathogenicity, as well as how innate immunity can be therapeutically targeted as part of the vaccination and treatment strategy. Overall, we highlight how a comprehensive understanding of innate immune mechanisms has been crucial in the fight against SARS-CoV-2 infections and the development of novel host-directed immunotherapeutic strategies for various diseases.
Collapse
Affiliation(s)
- R K Subbarao Malireddi
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, Tennessee, USA;
| | - Bhesh Raj Sharma
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, Tennessee, USA;
| | | |
Collapse
|
203
|
Khawaja T, Kajova M, Levonen I, Pietilä JP, Välimaa H, Paajanen J, Pakkanen SH, Patjas A, Montonen R, Miettinen S, Virtanen J, Smura T, Sironen T, Fagerlund R, Ugurlu H, Iheozor-Ejiofor R, Saksela K, Vahlberg T, Ranki A, Vierikko A, Ihalainen J, Vapalahti O, Kantele A. Double-blinded, randomised, placebo-controlled trial of convalescent plasma for COVID-19: analyses by neutralising antibodies homologous to recipients' variants. Infect Dis (Lond) 2024; 56:423-433. [PMID: 38513074 DOI: 10.1080/23744235.2024.2329957] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/18/2023] [Revised: 02/04/2024] [Accepted: 03/08/2024] [Indexed: 03/23/2024] Open
Abstract
INTRODUCTION Convalescent plasma (CP) emerged as potential treatment for COVID-19 early in the pandemic. While efficacy in hospitalised patients has been lacklustre, CP may be beneficial at the first stages of disease. Despite multiple new variants emerging, no trials have involved analyses on variant-specific antibody titres of CP. METHODS We recruited hospitalised COVID-19 patients within 10 days of symptom onset and, employing a double-blinded approach, randomised them to receive 200 ml convalescent plasma with high (HCP) or low (LCP) neutralising antibody (NAb) titre against the ancestral strain (Wuhan-like variant) or placebo in 1:1:1 ratio. Primary endpoints comprised intubation, corticosteroids for symptom aggravation, and safety assessed as serious adverse events. For a preplanned ad hoc analysis, the patients were regrouped by infused CP's NAb titers to variants infecting the recipients i.e. by titres of homologous HCP (hHCP) or LCP (hLCP). RESULTS Of the 57 patients, 18 received HCP, 19 LCP and 20 placebo, all groups smaller than planned. No significant differences were found for primary endpoints. In ad hoc analysis, hHCPrecipients needed significantly less respiratory support, and appeared to be given corticosteroids less frequently (1/14; 7.1%) than those receiving hLCP (9/23; 39.1%) or placebo (8/20; 40%), (p = 0.077). DISCUSSION Our double-blinded, placebo-controlled CP therapy trial remained underpowered and does not allow any firm conclusions for early-stage hospitalised COVID-19 patients. Interestingly, however, regrouping by homologous - recipients' variant-specific - CP titres suggested benefits for hHCP. We encourage similar re-analysis of ongoing/previous larger CP studies. TRIAL REGISTRATION ClinTrials.gov identifier: NCT0473040.
Collapse
Affiliation(s)
- T Khawaja
- Meilahti Vaccine Research Center, MeVac, Department of Infectious Diseases, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
- Department of Infectious Diseases, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
- Human Microbiome Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
- FIMAR, Multidisciplinary Center of Excellence in Antimicrobial Resistance Research, University of Helsinki, Helsinki, Finland
| | - M Kajova
- Meilahti Vaccine Research Center, MeVac, Department of Infectious Diseases, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
- Department of Infectious Diseases, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
- Human Microbiome Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
- FIMAR, Multidisciplinary Center of Excellence in Antimicrobial Resistance Research, University of Helsinki, Helsinki, Finland
| | - I Levonen
- Meilahti Vaccine Research Center, MeVac, Department of Infectious Diseases, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
- Human Microbiome Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - J P Pietilä
- Meilahti Vaccine Research Center, MeVac, Department of Infectious Diseases, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
- Human Microbiome Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
- FIMAR, Multidisciplinary Center of Excellence in Antimicrobial Resistance Research, University of Helsinki, Helsinki, Finland
| | - H Välimaa
- Meilahti Vaccine Research Center, MeVac, Department of Infectious Diseases, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
- Department of Virology, University of Helsinki, Helsinki, Finland
| | - J Paajanen
- Meilahti Vaccine Research Center, MeVac, Department of Infectious Diseases, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
- Department of Pulmonary Medicine, Heart and Lung Center, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - S H Pakkanen
- Meilahti Vaccine Research Center, MeVac, Department of Infectious Diseases, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
- Human Microbiome Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
- FIMAR, Multidisciplinary Center of Excellence in Antimicrobial Resistance Research, University of Helsinki, Helsinki, Finland
| | - A Patjas
- Meilahti Vaccine Research Center, MeVac, Department of Infectious Diseases, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
- Human Microbiome Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
- FIMAR, Multidisciplinary Center of Excellence in Antimicrobial Resistance Research, University of Helsinki, Helsinki, Finland
| | - R Montonen
- Department of Virology, University of Helsinki, Helsinki, Finland
| | - S Miettinen
- Department of Virology, University of Helsinki, Helsinki, Finland
- Department of Veterinary Biosciences, Faculty of Veterinary Medicine, University of Helsinki, Helsinki, Finland
| | - J Virtanen
- Department of Virology, University of Helsinki, Helsinki, Finland
- Department of Veterinary Biosciences, Faculty of Veterinary Medicine, University of Helsinki, Helsinki, Finland
| | - T Smura
- Department of Virology, University of Helsinki, Helsinki, Finland
| | - T Sironen
- Department of Virology, University of Helsinki, Helsinki, Finland
- Department of Veterinary Biosciences, Faculty of Veterinary Medicine, University of Helsinki, Helsinki, Finland
| | - R Fagerlund
- Department of Virology, University of Helsinki, Helsinki, Finland
| | - H Ugurlu
- Department of Virology, University of Helsinki, Helsinki, Finland
| | - R Iheozor-Ejiofor
- Department of Virology, University of Helsinki, Helsinki, Finland
- Department of Veterinary Biosciences, Faculty of Veterinary Medicine, University of Helsinki, Helsinki, Finland
| | - K Saksela
- Department of Virology, University of Helsinki, Helsinki, Finland
- HUS Diagnostic Centre, HUSLAB, Clinical Microbiology, Helsinki University Hospital, Helsinki, Finland
| | - T Vahlberg
- Department of Biostatistics, University of Turku and Turku University Hospital, Turku, Finland
| | - A Ranki
- Department of Dermatology, Allergology and Venereology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - A Vierikko
- Finnish Red Cross Blood Service, Helsinki, Finland
| | - J Ihalainen
- Finnish Red Cross Blood Service, Helsinki, Finland
| | - O Vapalahti
- Department of Virology, University of Helsinki, Helsinki, Finland
- Department of Veterinary Biosciences, Faculty of Veterinary Medicine, University of Helsinki, Helsinki, Finland
- HUS Diagnostic Centre, HUSLAB, Clinical Microbiology, Helsinki University Hospital, Helsinki, Finland
| | - A Kantele
- Meilahti Vaccine Research Center, MeVac, Department of Infectious Diseases, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
- Department of Infectious Diseases, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
- Human Microbiome Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
- FIMAR, Multidisciplinary Center of Excellence in Antimicrobial Resistance Research, University of Helsinki, Helsinki, Finland
| |
Collapse
|
204
|
Hoenigsperger H, Sivarajan R, Sparrer KM. Differences and similarities between innate immune evasion strategies of human coronaviruses. Curr Opin Microbiol 2024; 79:102466. [PMID: 38555743 DOI: 10.1016/j.mib.2024.102466] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2024] [Revised: 02/20/2024] [Accepted: 03/12/2024] [Indexed: 04/02/2024]
Abstract
So far, seven coronaviruses have emerged in humans. Four recurring endemic coronaviruses cause mild respiratory symptoms. Infections with epidemic Middle East respiratory syndrome-related coronavirus or severe acute respiratory syndrome coronavirus (SARS-CoV)-1 are associated with high mortality rates. SARS-CoV-2 is the causative agent of the coronavirus disease 2019 pandemic. To establish an infection, coronaviruses evade restriction by human innate immune defenses, such as the interferon system, autophagy and the inflammasome. Here, we review similar and distinct innate immune manipulation strategies employed by the seven human coronaviruses. We further discuss the impact on pathogenesis, zoonotic emergence and adaptation. Understanding the nature of the interplay between endemic/epidemic/pandemic coronaviruses and host defenses may help to better assess the pandemic potential of emerging coronaviruses.
Collapse
Affiliation(s)
- Helene Hoenigsperger
- Institute of Molecular Virology, Ulm University Medical Center, 89081 Ulm, Germany
| | - Rinu Sivarajan
- Institute of Molecular Virology, Ulm University Medical Center, 89081 Ulm, Germany
| | | |
Collapse
|
205
|
Turvey SE, Biggs CM, James EL, Hildebrand KJ. Should "primary immune disorder" replace "inborn error of immunity"? Names matter, but there is room for both. J Allergy Clin Immunol 2024; 153:1546-1547. [PMID: 38649118 DOI: 10.1016/j.jaci.2024.04.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2024] [Revised: 04/15/2024] [Accepted: 04/17/2024] [Indexed: 04/25/2024]
Affiliation(s)
- Stuart E Turvey
- Division of Immunology, Department of Pediatrics, BC Children's Hospital, The University of British Columbia, Vancouver, British Columbia, Canada.
| | - Catherine M Biggs
- Division of Immunology, Department of Pediatrics, BC Children's Hospital, The University of British Columbia, Vancouver, British Columbia, Canada
| | - Elliot L James
- Division of Immunology, Department of Pediatrics, BC Children's Hospital, The University of British Columbia, Vancouver, British Columbia, Canada
| | - Kyla J Hildebrand
- Division of Immunology, Department of Pediatrics, BC Children's Hospital, The University of British Columbia, Vancouver, British Columbia, Canada
| |
Collapse
|
206
|
Kerdiles T, Lejeune S, Portais A, Bourgeois G, Lefevre B, Charmillon A, Sixt T, Moretto F, Cornille C, Vidal M, Coustillères F, Martellosio JP, Quenet M, Belan M, Andry F, Jaffal K, Pinazo-Melia A, Rondeau P, Luque Paz D, Jouneau S, Borie R, Monnier D, Lebeaux D. Nocardia Infection in Patients With Anti-Granulocyte-Macrophage Colony-Stimulating Factor Autoantibodies: A Prospective Multicenter French Study. Open Forum Infect Dis 2024; 11:ofae269. [PMID: 38915339 PMCID: PMC11194753 DOI: 10.1093/ofid/ofae269] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Accepted: 05/05/2024] [Indexed: 06/26/2024] Open
Abstract
Background Nocardiosis, a bacterial opportunistic infection caused by Nocardia spp, has recently been reported in patients with anti-granulocyte-macrophage colony-stimulating factor (GM-CSF) autoantibodies, but insufficient data are available about disease presentation, outcomes, and occurrence of autoimmune pulmonary alveolar proteinosis (aPAP) in this population. Methods We performed a prospective, multicenter, nationwide study in France and included patients with a Nocardia infection who had anti-GM-CSF autoantibodies. We describe their clinical, microbiological, and radiological characteristics, and their outcome at 1 year of follow-up. Results Twenty patients (18 [90%] male) were included, with a median age of 69 (interquartile range, 44-75) years. The organs most frequently involved were the brain (14/20 [70%]) and the lung (12/20 [60%]). Half of the infections were disseminated (10/20 [50%]). Nocardia identification was predominantly made in abscess fluid (17/20 [85%]), among which 10 (59%) were brain abscesses. The 1-year all-cause mortality was 5% (1/20), and only 1 case of aPAP (1/20 [5%]) occurred during the follow-up period. Conclusions Nocardiosis with anti-GM-CSF autoantibodies is associated with a low mortality rate despite a high incidence of brain involvement. Although the occurrence of aPAP was infrequent during the 1-year follow-up period, long-term clinical data are needed to fully understand the potential relationship between nocardiosis, anti-GM-CSF autoantibodies, and aPAP.
Collapse
Affiliation(s)
- Thibault Kerdiles
- AP-HP, Département des Maladies Infectieuses et Tropicales, Hôpital Saint-Louis, Lariboisière, Paris, France
- Faculté de Médecine, Sorbonne Université, Paris, France
| | - Sophie Lejeune
- Service de maladies infectieuses et tropicales, CHU Grenoble Alpes, Grenoble, France
| | - Antoine Portais
- Service de maladies infectieuses et tropicales, CHU Grenoble Alpes, Grenoble, France
| | - Gaelle Bourgeois
- Service de Maladies Infectieuses, Centre Hospitalier Metropole Savoie, Chambéry, France
| | - Benjamin Lefevre
- Service des Maladies Infectieuses et Tropicales, CHRU-Nancy, Université de Lorraine, Nancy, France
- Université de Lorraine, Inserm, INSPIIRE, Nancy, France
| | - Alexandre Charmillon
- Service des Maladies Infectieuses et Tropicales, CHRU-Nancy, Université de Lorraine, Nancy, France
| | - Thibault Sixt
- Infectious Diseases Department, Dijon University Hospital, Dijon, France
| | - Florian Moretto
- Infectious Diseases Department, Dijon University Hospital, Dijon, France
| | - Cyril Cornille
- Service des maladies infectieuses et tropicales, Hôpital universitaire de Clermont-Ferrand, Clermont-Ferrand, France
| | - Magali Vidal
- Service des maladies infectieuses et tropicales, Hôpital universitaire de Clermont-Ferrand, Clermont-Ferrand, France
| | | | - Jean-Philippe Martellosio
- Service de médecine interne, maladies infectieuses et tropicales, CHU de Poitiers, Université de Poitiers, Poitiers, France
| | - Marion Quenet
- Service de Médecine Polyvalente, Centre Hospitalier Yves Le Foll, Saint-Brieuc, France
| | - Martin Belan
- Equipe Mobile d’Infectiologie, Hôpitaux Universitaires Paris Centre-Cochin Port Royal, Assistance publique Hôpitaux de Paris (AP-HP), Paris, France
| | - Fanny Andry
- Service de Maladies Infectieuses et Dermatologie, CHU de la Réunion, Saint Pierre, France
| | - Karim Jaffal
- Service des Maladies Infectieuses et Tropicales, Centre Hospitalier Universitaire Raymond Poincaré, Assistance publique Hôpitaux de Paris (AP-HP), Garches, France
| | | | - Paul Rondeau
- Service de Médecine interne, Hôpital Saint-Camille, Bry-sur-Marne, France
| | - David Luque Paz
- Infectious Diseases and Intensive Care Unit, Pontchaillou University Hospital, Rennes, France
- Bacterial Regulatory RNAs and Medicine, University of Rennes, UMR 1230, Inserm, Rennes, France
| | - Stephane Jouneau
- Department of Respiratory Medicine, CHU Rennes, Rennes, France
- Institut de recherche en santé, environnement et travail, Université Rennes, CHU Rennes, Inserm, EHESP, UMR_S 1085, Rennes, France
| | - Raphael Borie
- Service de Pneumologie A Hôpital Bichat, Assistance publique Hôpitaux de Paris (AP-HP), Université Paris Cité, Inserm, PHERE, Université Paris Cité, Paris, France
| | | | - David Lebeaux
- AP-HP, Département des Maladies Infectieuses et Tropicales, Hôpital Saint-Louis, Lariboisière, Paris, France
- Genetics of Biofilms Laboratory, Institut Pasteur, Université Paris Cité, CNRS UMR 6047, Paris, France
| |
Collapse
|
207
|
Trivedi PJ, Hirschfield GM, Adams DH, Vierling JM. Immunopathogenesis of Primary Biliary Cholangitis, Primary Sclerosing Cholangitis and Autoimmune Hepatitis: Themes and Concepts. Gastroenterology 2024; 166:995-1019. [PMID: 38342195 DOI: 10.1053/j.gastro.2024.01.049] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/25/2023] [Revised: 01/21/2024] [Accepted: 01/28/2024] [Indexed: 02/13/2024]
Abstract
Autoimmune liver diseases include primary biliary cholangitis, primary sclerosing cholangitis, and autoimmune hepatitis, a family of chronic immune-mediated disorders that target hepatocytes and cholangiocytes. Treatments remain nonspecific, variably effective, and noncurative, and the need for liver transplantation is disproportionate to their rarity. Development of effective therapies requires better knowledge of pathogenic mechanisms, including the roles of genetic risk, and how the environment and gut dysbiosis cause immune cell dysfunction and aberrant bile acid signaling. This review summarizes key etiologic and pathogenic concepts and themes relevant for clinical practice and how such learning can guide the development of new therapies for people living with autoimmune liver diseases.
Collapse
Affiliation(s)
- Palak J Trivedi
- National Institute for Health Research Birmingham Biomedical Research Centre, Centre for Liver and Gastrointestinal Research, Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, United Kingdom; Liver Unit, University Hospitals Birmingham, Birmingham, United Kingdom; Institute of Translational Medicine, University of Birmingham, Birmingham, United Kingdom.
| | - Gideon M Hirschfield
- Division of Gastroenterology and Hepatology, Toronto Centre for Liver Disease, University of Toronto, Toronto, Ontario, Canada
| | - David H Adams
- National Institute for Health Research Birmingham Biomedical Research Centre, Centre for Liver and Gastrointestinal Research, Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, United Kingdom; Liver Unit, University Hospitals Birmingham, Birmingham, United Kingdom
| | - John M Vierling
- Section of Gastroenterology and Hepatology, Department of Medicine, Baylor College of Medicine, Houston, Texas; Division of Abdominal Transplantation, Department of Surgery, Baylor College of Medicine, Houston, Texas.
| |
Collapse
|
208
|
Delmonte OM, Oguz C, Dobbs K, Myint-Hpu K, Palterer B, Abers MS, Draper D, Truong M, Kaplan IM, Gittelman RM, Zhang Y, Rosen LB, Snow AL, Dalgard CL, Burbelo PD, Imberti L, Sottini A, Quiros-Roldan E, Castelli F, Rossi C, Brugnoni D, Biondi A, Bettini LR, D'Angio M, Bonfanti P, Anderson MV, Saracino A, Chironna M, Di Stefano M, Fiore JR, Santantonio T, Castagnoli R, Marseglia GL, Magliocco M, Bosticardo M, Pala F, Shaw E, Matthews H, Weber SE, Xirasagar S, Barnett J, Oler AJ, Dimitrova D, Bergerson JRE, McDermott DH, Rao VK, Murphy PM, Holland SM, Lisco A, Su HC, Lionakis MS, Cohen JI, Freeman AF, Snyder TM, Lack J, Notarangelo LD. Perturbations of the T-cell receptor repertoire in response to SARS-CoV-2 in immunocompetent and immunocompromised individuals. J Allergy Clin Immunol 2024; 153:1655-1667. [PMID: 38154666 PMCID: PMC11162338 DOI: 10.1016/j.jaci.2023.12.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2023] [Revised: 12/13/2023] [Accepted: 12/19/2023] [Indexed: 12/30/2023]
Abstract
BACKGROUND Functional T-cell responses are essential for virus clearance and long-term protection after severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection, whereas certain clinical factors, such as older age and immunocompromise, are associated with worse outcome. OBJECTIVE We sought to study the breadth and magnitude of T-cell responses in patients with coronavirus disease 2019 (COVID-19) and in individuals with inborn errors of immunity (IEIs) who had received COVID-19 mRNA vaccine. METHODS Using high-throughput sequencing and bioinformatics tools to characterize the T-cell receptor β repertoire signatures in 540 individuals after SARS-CoV-2 infection, 31 IEI recipients of COVID-19 mRNA vaccine, and healthy controls, we quantified HLA class I- and class II-restricted SARS-CoV-2-specific responses and also identified several HLA allele-clonotype motif associations in patients with COVID-19, including a subcohort of anti-type 1 interferon (IFN-1)-positive patients. RESULTS Our analysis revealed that elderly patients with COVID-19 with critical disease manifested lower SARS-CoV-2 T-cell clonotype diversity as well as T-cell responses with reduced magnitude, whereas the SARS-CoV-2-specific clonotypes targeted a broad range of HLA class I- and class II-restricted epitopes across the viral proteome. The presence of anti-IFN-I antibodies was associated with certain HLA alleles. Finally, COVID-19 mRNA immunization induced an increase in the breadth of SARS-CoV-2-specific clonotypes in patients with IEIs, including those who had failed to seroconvert. CONCLUSIONS Elderly individuals have impaired capacity to develop broad and sustained T-cell responses after SARS-CoV-2 infection. Genetic factors may play a role in the production of anti-IFN-1 antibodies. COVID-19 mRNA vaccines are effective in inducing T-cell responses in patients with IEIs.
Collapse
Affiliation(s)
- Ottavia M Delmonte
- Laboratory of Clinical Immunology and Microbiology National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Md.
| | - Cihan Oguz
- Integrated Data Sciences Section, Research Technology Branch, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Md
| | - Kerry Dobbs
- Laboratory of Clinical Immunology and Microbiology National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Md
| | - Katherine Myint-Hpu
- Laboratory of Clinical Immunology and Microbiology National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Md
| | - Boaz Palterer
- Laboratory of Clinical Immunology and Microbiology National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Md
| | - Michael S Abers
- Laboratory of Clinical Immunology and Microbiology National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Md
| | - Deborah Draper
- Laboratory of Clinical Immunology and Microbiology National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Md
| | - Meng Truong
- Laboratory of Clinical Immunology and Microbiology National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Md
| | | | | | - Yu Zhang
- Laboratory of Clinical Immunology and Microbiology National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Md
| | - Lindsey B Rosen
- Laboratory of Clinical Immunology and Microbiology National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Md
| | - Andrew L Snow
- Laboratory of Clinical Immunology and Microbiology National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Md; Department of Pharmacology & Molecular Therapeutics, Uniformed Services University of the Health Sciences, Bethesda, Md
| | - Clifton L Dalgard
- Department of Anatomy, Physiology & Genetics, Uniformed Services University of the Health Sciences, Bethesda, Md; The American Genome Center, Uniformed Services University of the Health Sciences, Bethesda, Md
| | - Peter D Burbelo
- Adeno-Associated Virus Biology Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, Md
| | - Luisa Imberti
- Section of Microbiology, University of Brescia, ASST Spedali Civili, Brescia, Italy
| | - Alessandra Sottini
- Section of Microbiology, University of Brescia, ASST Spedali Civili, Brescia, Italy
| | - Eugenia Quiros-Roldan
- Department of Infectious and Tropical Diseases, University of Brescia, ASST Spedali Civili, Brescia, Italy
| | - Francesco Castelli
- Department of Infectious and Tropical Diseases, University of Brescia, ASST Spedali Civili, Brescia, Italy
| | - Camillo Rossi
- Direzione Sanitaria, ASST Spedali Civili, Brescia, Italy
| | - Duilio Brugnoni
- Laboratorio Analisi Chimico-Cliniche, ASST Spedali Civili, Brescia, Italy
| | - Andrea Biondi
- Pediatric Department and Centro Tettamanti-European Reference Network on Paediatric Cancer, European Reference Network on Haematological Diseases, and European Reference Network on Hereditary Metabolic Disorders, University of Milano-Bicocca-Fondazione MBBM, Monza, Italy
| | - Laura Rachele Bettini
- Pediatric Department and Centro Tettamanti-European Reference Network on Paediatric Cancer, European Reference Network on Haematological Diseases, and European Reference Network on Hereditary Metabolic Disorders, University of Milano-Bicocca-Fondazione MBBM, Monza, Italy
| | - Mariella D'Angio
- Pediatric Department and Centro Tettamanti-European Reference Network on Paediatric Cancer, European Reference Network on Haematological Diseases, and European Reference Network on Hereditary Metabolic Disorders, University of Milano-Bicocca-Fondazione MBBM, Monza, Italy
| | - Paolo Bonfanti
- Department of Infectious Diseases, San Gerardo Hospital, University of Milano-Bicocca, Monza, Italy
| | - Megan V Anderson
- Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Md
| | - Annalisa Saracino
- Clinic of Infectious Diseases, Azienda Ospedaliero-Universitaria Consorziale Policlinico di Bari, University of Bari, Bari, Italy
| | - Maria Chironna
- Hygiene Section, Department of Interdisciplinary Medicine, University of Bari Aldo Moro, Bari, Italy
| | - Mariantonietta Di Stefano
- Department of Medical and Surgical Sciences, Section of Infectious Diseases, University of Foggia, Foggia, Italy
| | - Jose Ramon Fiore
- Department of Medical and Surgical Sciences, Section of Infectious Diseases, University of Foggia, Foggia, Italy
| | - Teresa Santantonio
- Department of Medical and Surgical Sciences, Section of Infectious Diseases, University of Foggia, Foggia, Italy
| | - Riccardo Castagnoli
- Department of Clinical, Surgical, Diagnostic and Pediatric Sciences, University of Pavia, Pavia, Italy; Pediatric Clinic, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | - Gian Luigi Marseglia
- Department of Clinical, Surgical, Diagnostic and Pediatric Sciences, University of Pavia, Pavia, Italy; Pediatric Clinic, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | - Mary Magliocco
- Molecular Development of the Immune System Section, Laboratory of Immune System Biology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Md
| | - Marita Bosticardo
- Laboratory of Clinical Immunology and Microbiology National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Md
| | - Francesca Pala
- Laboratory of Clinical Immunology and Microbiology National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Md
| | - Elana Shaw
- Laboratory of Clinical Immunology and Microbiology National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Md
| | - Helen Matthews
- Molecular Development of the Immune System Section, Laboratory of Immune System Biology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Md
| | - Sarah E Weber
- Molecular Development of the Immune System Section, Laboratory of Immune System Biology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Md
| | - Sandhya Xirasagar
- Bioinformatics and Computational Biosciences Branch, Office of Cyber Infrastructure and Computational Biology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Md
| | - Jason Barnett
- Bioinformatics and Computational Biosciences Branch, Office of Cyber Infrastructure and Computational Biology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Md
| | - Andrew J Oler
- Bioinformatics and Computational Biosciences Branch, Office of Cyber Infrastructure and Computational Biology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Md
| | - Dimana Dimitrova
- Center for Immuno-Oncology, National Cancer Institute, National Institutes of Health, Bethesda, Md
| | - Jenna R E Bergerson
- Laboratory of Clinical Immunology and Microbiology National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Md
| | - David H McDermott
- Laboratory of Molecular Immunology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Md
| | - V Koneti Rao
- Laboratory of Clinical Immunology and Microbiology National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Md
| | - Philip M Murphy
- Laboratory of Molecular Immunology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Md
| | - Steven M Holland
- Laboratory of Clinical Immunology and Microbiology National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Md
| | - Andrea Lisco
- Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Md
| | - Helen C Su
- Laboratory of Clinical Immunology and Microbiology National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Md
| | - Michail S Lionakis
- Laboratory of Clinical Immunology and Microbiology National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Md
| | - Jeffrey I Cohen
- Laboratory of Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Md
| | - Alexandra F Freeman
- Laboratory of Clinical Immunology and Microbiology National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Md
| | | | - Justin Lack
- Integrated Data Sciences Section, Research Technology Branch, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Md
| | - Luigi D Notarangelo
- Laboratory of Clinical Immunology and Microbiology National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Md.
| |
Collapse
|
209
|
Miller CN, Waterfield MR, Gardner JM, Anderson MS. Aire in Autoimmunity. Annu Rev Immunol 2024; 42:427-53. [PMID: 38360547 PMCID: PMC11774315 DOI: 10.1146/annurev-immunol-090222-101050] [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] [Indexed: 02/17/2024]
Abstract
The role of the autoimmune regulator (Aire) in central immune tolerance and thymic self-representation was first described more than 20 years ago, but fascinating new insights into its biology continue to emerge, particularly in the era of advanced single-cell genomics. We briefly describe the role of human genetics in the discovery of Aire, as well as insights into its function gained from genotype-phenotype correlations and the spectrum of Aire-associated autoimmunity-including insights from patients with Aire mutations with broad and diverse implications for human health. We then highlight emerging trends in Aire biology, focusing on three topic areas. First, we discuss medullary thymic epithelial diversity and the role of Aire in thymic epithelial development. Second, we highlight recent developments regarding the molecular mechanisms of Aire and its binding partners. Finally, we describe the rapidly evolving biology of the identity and function of extrathymic Aire-expressing cells (eTACs), and a novel eTAC subset called Janus cells, as well as their potential roles in immune homeostasis.
Collapse
Affiliation(s)
- Corey N Miller
- Diabetes Center, University of California, San Francisco, California, USA; ,
- Department of Medicine, University of California, San Francisco, California, USA
| | - Michael R Waterfield
- Department of Pediatrics, University of California, San Francisco, California, USA
| | - James M Gardner
- Diabetes Center, University of California, San Francisco, California, USA; ,
- Department of Surgery, University of California, San Francisco, California, USA
| | - Mark S Anderson
- Diabetes Center, University of California, San Francisco, California, USA; ,
- Department of Medicine, University of California, San Francisco, California, USA
| |
Collapse
|
210
|
Edner NM, Houghton LP, Ntavli E, Rees-Spear C, Petersone L, Wang C, Fabri A, Elfaki Y, Rueda Gonzalez A, Brown R, Kisand K, Peterson P, McCoy LE, Walker LSK. TIGIT +Tfh show poor B-helper function and negatively correlate with SARS-CoV-2 antibody titre. Front Immunol 2024; 15:1395684. [PMID: 38868776 PMCID: PMC11167088 DOI: 10.3389/fimmu.2024.1395684] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2024] [Accepted: 05/15/2024] [Indexed: 06/14/2024] Open
Abstract
Circulating follicular helper T cells (cTfh) can show phenotypic alterations in disease settings, including in the context of tissue-damaging autoimmune or anti-viral responses. Using severe COVID-19 as a paradigm of immune dysregulation, we have explored how cTfh phenotype relates to the titre and quality of antibody responses. Severe disease was associated with higher titres of neutralising S1 IgG and evidence of increased T cell activation. ICOS, CD38 and HLA-DR expressing cTfh correlated with serum S1 IgG titres and neutralising strength, and interestingly expression of TIGIT by cTfh showed a negative correlation. TIGIT+cTfh expressed increased IFNγ and decreased IL-17 compared to their TIGIT-cTfh counterparts, and showed reduced capacity to help B cells in vitro. Additionally, TIGIT+cTfh expressed lower levels of CD40L than TIGIT-cTfh, providing a potential explanation for their poor B-helper function. These data identify phenotypic changes in polyclonal cTfh that correlate with specific antibody responses and reveal TIGIT as a marker of cTfh with altered function.
Collapse
Affiliation(s)
- Natalie M. Edner
- Division of Infection and Immunity, Institute of Immunity and Transplantation, University College London, London, United Kingdom
| | - Luke P. Houghton
- Division of Infection and Immunity, Institute of Immunity and Transplantation, University College London, London, United Kingdom
| | - Elisavet Ntavli
- Division of Infection and Immunity, Institute of Immunity and Transplantation, University College London, London, United Kingdom
| | - Chloe Rees-Spear
- Division of Infection and Immunity, Institute of Immunity and Transplantation, University College London, London, United Kingdom
| | - Lina Petersone
- Division of Infection and Immunity, Institute of Immunity and Transplantation, University College London, London, United Kingdom
| | - Chunjing Wang
- Division of Infection and Immunity, Institute of Immunity and Transplantation, University College London, London, United Kingdom
| | - Astrid Fabri
- Division of Infection and Immunity, Institute of Immunity and Transplantation, University College London, London, United Kingdom
| | - Yassin Elfaki
- Division of Infection and Immunity, Institute of Immunity and Transplantation, University College London, London, United Kingdom
| | - Andrea Rueda Gonzalez
- Division of Infection and Immunity, Institute of Immunity and Transplantation, University College London, London, United Kingdom
| | - Rachel Brown
- Division of Infection and Immunity, Institute of Immunity and Transplantation, University College London, London, United Kingdom
- Queen Square Institute of Neurology, University College London, London, United Kingdom
| | - Kai Kisand
- Institute of Biomedicine and Translational Medicine, University of Tartu, Tartu, Estonia
| | - Pärt Peterson
- Institute of Biomedicine and Translational Medicine, University of Tartu, Tartu, Estonia
| | - Laura E. McCoy
- Division of Infection and Immunity, Institute of Immunity and Transplantation, University College London, London, United Kingdom
| | - Lucy S. K. Walker
- Division of Infection and Immunity, Institute of Immunity and Transplantation, University College London, London, United Kingdom
| |
Collapse
|
211
|
Brunet-Ratnasingham E, Morin S, Randolph HE, Labrecque M, Bélair J, Lima-Barbosa R, Pagliuzza A, Marchitto L, Hultström M, Niessl J, Cloutier R, Sreng Flores AM, Brassard N, Benlarbi M, Prévost J, Ding S, Anand SP, Sannier G, Marks A, Wågsäter D, Bareke E, Zeberg H, Lipcsey M, Frithiof R, Larsson A, Zhou S, Nakanishi T, Morrison D, Vezina D, Bourassa C, Gendron-Lepage G, Medjahed H, Point F, Richard J, Larochelle C, Prat A, Cunningham JL, Arbour N, Durand M, Richards JB, Moon K, Chomont N, Finzi A, Tétreault M, Barreiro L, Wolf G, Kaufmann DE. Sustained IFN signaling is associated with delayed development of SARS-CoV-2-specific immunity. Nat Commun 2024; 15:4177. [PMID: 38755196 PMCID: PMC11522391 DOI: 10.1038/s41467-024-48556-y] [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: 06/01/2023] [Accepted: 05/06/2024] [Indexed: 05/18/2024] Open
Abstract
Plasma RNAemia, delayed antibody responses and inflammation predict COVID-19 outcomes, but the mechanisms underlying these immunovirological patterns are poorly understood. We profile 782 longitudinal plasma samples from 318 hospitalized patients with COVID-19. Integrated analysis using k-means reveals four patient clusters in a discovery cohort: mechanically ventilated critically-ill cases are subdivided into good prognosis and high-fatality clusters (reproduced in a validation cohort), while non-critical survivors segregate into high and low early antibody responders. Only the high-fatality cluster is enriched for transcriptomic signatures associated with COVID-19 severity, and each cluster has distinct RBD-specific antibody elicitation kinetics. Both critical and non-critical clusters with delayed antibody responses exhibit sustained IFN signatures, which negatively correlate with contemporaneous RBD-specific IgG levels and absolute SARS-CoV-2-specific B and CD4+ T cell frequencies. These data suggest that the "Interferon paradox" previously described in murine LCMV models is operative in COVID-19, with excessive IFN signaling delaying development of adaptive virus-specific immunity.
Collapse
Affiliation(s)
- Elsa Brunet-Ratnasingham
- Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Montreal, QC, Canada
- Département de Microbiologie, Infectiologie et Immunologie, Université de Montréal, Montreal, QC, Canada
- Department of Medicine, University of California, San Francisco, CA, USA
| | - Sacha Morin
- Department of Computer Science and Operations Research, Université de Montréal, Montreal, QC, Canada
- Mila-Quebec AI Institute, Montreal, QC, Canada
| | - Haley E Randolph
- Committee on Genetics, Genomics, and Systems Biology, University of Chicago, Chicago, IL, USA
| | - Marjorie Labrecque
- Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Montreal, QC, Canada
- Bioinformatics Program, Department of Biochemistry and Molecular Medicine, Université de Montréal, Montreal, QC, Canada
| | - Justin Bélair
- Department of Mathematics and Statistics, Université de Montréal, Montreal, QC, Canada
- Independent Data Scientist, JB Consulting, Montreal, QC, H3S1K8, Canada
| | - Raphaël Lima-Barbosa
- Department of Mathematics and Statistics, Université de Montréal, Montreal, QC, Canada
- Independent Data Scientist, JB Consulting, Montreal, QC, H3S1K8, Canada
| | - Amélie Pagliuzza
- Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Montreal, QC, Canada
| | - Lorie Marchitto
- Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Montreal, QC, Canada
- Département de Microbiologie, Infectiologie et Immunologie, Université de Montréal, Montreal, QC, Canada
| | - Michael Hultström
- Anaesthesiology and Intensive Care Medicine, Department of Surgical Sciences, Uppsala University, Uppsala, Sweden.
- Integrative Physiology, Department of Medical Cell Biology, Uppsala University, Uppsala, Sweden.
- Department of Epidemiology, Biostatistics and Occupational Health, McGill University, Montreal, QC, Canada.
- Lady Davis Institute for Medical Research, Jewish General Hospital, Montreal, QC, Canada.
| | - Julia Niessl
- Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Montreal, QC, Canada
- Département de Microbiologie, Infectiologie et Immunologie, Université de Montréal, Montreal, QC, Canada
- BioNTech SE, Mainz, Germany
| | - Rose Cloutier
- Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Montreal, QC, Canada
| | - Alina M Sreng Flores
- Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Montreal, QC, Canada
| | - Nathalie Brassard
- Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Montreal, QC, Canada
| | - Mehdi Benlarbi
- Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Montreal, QC, Canada
- Département de Microbiologie, Infectiologie et Immunologie, Université de Montréal, Montreal, QC, Canada
| | - Jérémie Prévost
- Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Montreal, QC, Canada
- Département de Microbiologie, Infectiologie et Immunologie, Université de Montréal, Montreal, QC, Canada
| | - Shilei Ding
- Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Montreal, QC, Canada
| | - Sai Priya Anand
- Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Montreal, QC, Canada
- Département de Microbiologie, Infectiologie et Immunologie, Université de Montréal, Montreal, QC, Canada
| | - Gérémy Sannier
- Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Montreal, QC, Canada
- Département de Microbiologie, Infectiologie et Immunologie, Université de Montréal, Montreal, QC, Canada
| | - Amanda Marks
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - Dick Wågsäter
- Integrative Physiology, Department of Medical Cell Biology, Uppsala University, Uppsala, Sweden
| | - Eric Bareke
- Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Montreal, QC, Canada
| | - Hugo Zeberg
- Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden
- Department of Evolutionary Genetics, Max Planck Institute for Evolutionary Anthropology, Leipzig, Germany
| | - Miklos Lipcsey
- Anaesthesiology and Intensive Care Medicine, Department of Surgical Sciences, Uppsala University, Uppsala, Sweden
- Hedenstierna Laboratory, Department of Surgical Sciences, Uppsala University, Uppsala, Sweden
| | - Robert Frithiof
- Anaesthesiology and Intensive Care Medicine, Department of Surgical Sciences, Uppsala University, Uppsala, Sweden
| | - Anders Larsson
- Clinical Chemistry, Department of Medical Sciences, Uppsala University, Uppsala, Sweden
| | - Sirui Zhou
- Lady Davis Institute for Medical Research, Jewish General Hospital, Montreal, QC, Canada
- Department of Human Genetics, McGill University, Montreal, QC, Canada
| | - Tomoko Nakanishi
- Lady Davis Institute for Medical Research, Jewish General Hospital, Montreal, QC, Canada
- Department of Human Genetics, McGill University, Montreal, QC, Canada
- Kyoto-McGill International Collaborative School in Genomic Medicine, Gaduate School of Medicine, Kyoto University, Kyoto, Japan
- Research Fellow, Japan Society for the Promotion of Science, Tokyo, Japan
| | - David Morrison
- Lady Davis Institute for Medical Research, Jewish General Hospital, Montreal, QC, Canada
| | - Dani Vezina
- Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Montreal, QC, Canada
- Département de Microbiologie, Infectiologie et Immunologie, Université de Montréal, Montreal, QC, Canada
| | - Catherine Bourassa
- Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Montreal, QC, Canada
| | - Gabrielle Gendron-Lepage
- Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Montreal, QC, Canada
| | - Halima Medjahed
- Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Montreal, QC, Canada
| | - Floriane Point
- Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Montreal, QC, Canada
| | - Jonathan Richard
- Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Montreal, QC, Canada
| | - Catherine Larochelle
- Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Montreal, QC, Canada
- Department of Neurosciences, Université de Montréal, Montreal, QC, Canada
| | - Alexandre Prat
- Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Montreal, QC, Canada
- Department of Neurosciences, Université de Montréal, Montreal, QC, Canada
| | - Janet L Cunningham
- Department of Medical Sciences, Psychiatry, Uppsala University, Uppsala, Sweden
| | - Nathalie Arbour
- Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Montreal, QC, Canada
- Department of Neurosciences, Université de Montréal, Montreal, QC, Canada
| | - Madeleine Durand
- Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Montreal, QC, Canada
- Centre Hospitalier de l'Université de Montréal (CHUM), Montreal, QC, Canada
| | - J Brent Richards
- Department of Epidemiology, Biostatistics and Occupational Health, McGill University, Montreal, QC, Canada
- Lady Davis Institute for Medical Research, Jewish General Hospital, Montreal, QC, Canada
- Department of Human Genetics, McGill University, Montreal, QC, Canada
- Department of Twin Research, King's College London, London, UK
| | - Kevin Moon
- Department of Mathematics and Statistics, Utah State University, Logan, UT, USA
| | - Nicolas Chomont
- Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Montreal, QC, Canada
- Département de Microbiologie, Infectiologie et Immunologie, Université de Montréal, Montreal, QC, Canada
| | - Andrés Finzi
- Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Montreal, QC, Canada
- Département de Microbiologie, Infectiologie et Immunologie, Université de Montréal, Montreal, QC, Canada
- Department of Microbiology and Immunology, McGill University, Montreal, QC, H3A 2B4, Canada
| | - Martine Tétreault
- Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Montreal, QC, Canada
- Department of Neurosciences, Université de Montréal, Montreal, QC, Canada
| | - Luis Barreiro
- Committee on Genetics, Genomics, and Systems Biology, University of Chicago, Chicago, IL, USA
- Section of Genetic Medicine, Department of Medicine, University of Chicago, Chicago, IL, USA
- Committee on Immunology, University of Chicago, Chicago, IL, USA
| | - Guy Wolf
- Department of Computer Science and Operations Research, Université de Montréal, Montreal, QC, Canada.
- Mila-Quebec AI Institute, Montreal, QC, Canada.
- Department of Mathematics and Statistics, Université de Montréal, Montreal, QC, Canada.
| | - Daniel E Kaufmann
- Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Montreal, QC, Canada.
- Département de Médecine, Université de Montréal, Montreal, QC, Canada.
- Division of Infectious Diseases, Department of Medicine, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland.
| |
Collapse
|
212
|
Qaqish A, Abbas MM, Alkhateeb M, Al-Tamimi M, Mustafa M, Al-Shudifat AE, Tarawneh S, Dawoud R, Mryyian A, Al-Ajaleen M. Anti_spike and anti_nucleocapsid IgG responses to SARS-CoV-2 in children of Jordan. Heliyon 2024; 10:e30631. [PMID: 38765100 PMCID: PMC11101777 DOI: 10.1016/j.heliyon.2024.e30631] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2023] [Revised: 04/30/2024] [Accepted: 05/01/2024] [Indexed: 05/21/2024] Open
Abstract
Background It is proven that children have significantly milder COVID-19 disease compared to adults. Various immunological characteristics influence this age-related difference in protection against COVID-19. Pediatric COVID-19 in Jordan is extremely under reported. Objectives The primary goal of this work is to identify the anti_S and anti_N antibody responses in a random group of children in Jordan and compare it to that of naturally infected-unvaccinated adults. Methods 151 unvaccinated children, 4 days to 18 years old, were screened for anti_S and anti_N antibodies. History of COVID-19 infection or exposure to infection and symptom severity were reported by parents on a special questionnaire. Results 78.9 % and 65.3 % of participants were seropositive for anti_S IgG and anti_N Abs, respectively. There was a remarkable association between age and anti_S IgG and anti_N IgG antibody titers, as children aged 12 years or older had increased anti_S IgG titers (mean = 19.3 BAU/mL) compared to younger groups (means of 10.15, 9.24, 7.91 BAU/mL for age groups 6-12, 1-6, less than 1 year, respectively). Gender did not show a statistically important role in anti_S and anti_N IgG seropositivity rates or titers. Children displayed significantly elevated anti_S titers (mean = 13.23 BAU/mL) compared to naturally infected adults (mean = 9.72 BAU/mL), in contrast, adults' anti_N titers (mean = 39.64 U/mL) were significantly higher compared to those of children (mean = 10.77 U/mL). Conclusions The current work provides evidence of distinctly robust and persistent humoral immunity displayed by high anti_S and anti_N IgG in children, even >12 months post-infection. Age was the only factor that had a significant statistical impact on anti_S and anti_N Ab levels among the pediatric group in this study. Children exhibited significantly higher anti_S titers than naturally infected adults. In contrast, adults' anti_N titers were significantly higher. Such information can assist direct pediatric SARS-CoV-2 immunization programs, with implications for creating age-targeted strategies for diagnostic and population protection measures.
Collapse
Affiliation(s)
- Arwa Qaqish
- Department of Biology and Biotechnology, Faculty of Science, The Hashemite University, Zarqa, Jordan
- Department of Cellular Therapy and Applied Genomics, King Hussein Cancer Center (KHCC), Amman, Jordan
| | - Manal Mohammad Abbas
- Department of Medical Laboratory Sciences, Faculty of Allied Medical Sciences, Al-Ahliyya Amman University, Amman, Jordan
| | - Mohammad Alkhateeb
- Department of Internal Medicine, King Hussein Cancer Center (KHCC), Amman, Jordan
| | - Mohammad Al-Tamimi
- Department of Microbiology, Pathology and Forensic Medicine, Faculty of Medicine, The Hashemite University, Zarqa, Jordan
| | - Minas Mustafa
- Department of Medical Laboratory Sciences, Faculty of Applied Health Sciences, The Hashemite University, Zarqa, Jordan
| | - Abdel-Ellah Al-Shudifat
- Department of Internal and Family Medicine, Faculty of Medicine, The Hashemite University, Zarqa, Jordan
| | - Shahd Tarawneh
- Department of Microbiology, Pathology and Forensic Medicine, Faculty of Medicine, The Hashemite University, Zarqa, Jordan
| | - Rand Dawoud
- Institute for Family Health, King Hussein Foundation, Amman, Jordan
| | - Amel Mryyian
- Department of Pediatrics, King Hussein Cancer Center (KHCC), Amman, Jordan
| | - Mu'ath Al-Ajaleen
- Department of Biology and Biotechnology, Faculty of Science, The Hashemite University, Zarqa, Jordan
| |
Collapse
|
213
|
Viox EG, Bosinger SE, Douek DC, Schreiber G, Paiardini M. Harnessing the power of IFN for therapeutic approaches to COVID-19. J Virol 2024; 98:e0120423. [PMID: 38651899 PMCID: PMC11092331 DOI: 10.1128/jvi.01204-23] [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] [Indexed: 04/25/2024] Open
Abstract
Interferons (IFNs) are essential for defense against viral infections but also drive recruitment of inflammatory cells to sites of infection, a key feature of severe COVID-19. Here, we explore the complexity of the IFN response in COVID-19, examine the effects of manipulating IFN on SARS-CoV-2 viral replication and pathogenesis, and highlight pre-clinical and clinical studies evaluating the therapeutic efficacy of IFN in limiting COVID-19 severity.
Collapse
Affiliation(s)
- Elise G. Viox
- Division of Microbiology and Immunology, Emory National Primate Research Center, Emory University, Atlanta, Georgia, USA
| | - Steven E. Bosinger
- Division of Microbiology and Immunology, Emory National Primate Research Center, Emory University, Atlanta, Georgia, USA
- Emory NPRC Genomics Core Emory National Primate Research Center, Emory University, Atlanta, Georgia, USA
- Department of Pathology and Laboratory Medicine, School of Medicine, Emory University, Atlanta, Georgia, USA
| | - Daniel C. Douek
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Gideon Schreiber
- Department of Biomolecular Sciences, Weizmann Institute of Science, Rehovot, Israel
| | - Mirko Paiardini
- Division of Microbiology and Immunology, Emory National Primate Research Center, Emory University, Atlanta, Georgia, USA
- Department of Pathology and Laboratory Medicine, School of Medicine, Emory University, Atlanta, Georgia, USA
| |
Collapse
|
214
|
Wang S, Li W, Wang L, Tiwari SK, Bray W, Wu L, Li N, Hui H, Clark AE, Zhang Q, Zhang L, Carlin AF, Rana TM. Interferon-Inducible Guanylate-Binding Protein 5 Inhibits Replication of Multiple Viruses by Binding to the Oligosaccharyltransferase Complex and Inhibiting Glycoprotein Maturation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.01.591800. [PMID: 38746287 PMCID: PMC11092618 DOI: 10.1101/2024.05.01.591800] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/16/2024]
Abstract
Viral infection induces production of type I interferons and expression of interferon-stimulated genes (ISGs) that play key roles in inhibiting viral infection. Here, we show that the ISG guanylate-binding protein 5 (GBP5) inhibits N-linked glycosylation of key proteins in multiple viruses, including SARS-CoV-2 spike protein. GBP5 binds to accessory subunits of the host oligosaccharyltransferase (OST) complex and blocks its interaction with the spike protein, which results in misfolding and retention of spike protein in the endoplasmic reticulum likely due to decreased N-glycan transfer, and reduces the assembly and release of infectious virions. Consistent with these observations, pharmacological inhibition of the OST complex with NGI-1 potently inhibits glycosylation of other viral proteins, including MERS-CoV spike protein, HIV-1 gp160, and IAV hemagglutinin, and prevents the production of infectious virions. Our results identify a novel strategy by which ISGs restrict virus infection and provide a rationale for targeting glycosylation as a broad antiviral therapeutic strategy.
Collapse
Affiliation(s)
- Shaobo Wang
- Division of Genetics, Department of Pediatrics, Program in Immunology, Bioinformatics and Systems Biology Program, Institute for Genomic Medicine, UCSD Center for AIDS Research, University of California San Diego, 9500 Gilman Drive MC 0762, La Jolla, California 92093, USA
- These authors contributed equally to this work
| | - Wanyu Li
- Division of Genetics, Department of Pediatrics, Program in Immunology, Bioinformatics and Systems Biology Program, Institute for Genomic Medicine, UCSD Center for AIDS Research, University of California San Diego, 9500 Gilman Drive MC 0762, La Jolla, California 92093, USA
- These authors contributed equally to this work
| | - Lingling Wang
- Division of Genetics, Department of Pediatrics, Program in Immunology, Bioinformatics and Systems Biology Program, Institute for Genomic Medicine, UCSD Center for AIDS Research, University of California San Diego, 9500 Gilman Drive MC 0762, La Jolla, California 92093, USA
| | - Shashi Kant Tiwari
- Division of Genetics, Department of Pediatrics, Program in Immunology, Bioinformatics and Systems Biology Program, Institute for Genomic Medicine, UCSD Center for AIDS Research, University of California San Diego, 9500 Gilman Drive MC 0762, La Jolla, California 92093, USA
| | - William Bray
- Division of Genetics, Department of Pediatrics, Program in Immunology, Bioinformatics and Systems Biology Program, Institute for Genomic Medicine, UCSD Center for AIDS Research, University of California San Diego, 9500 Gilman Drive MC 0762, La Jolla, California 92093, USA
| | - Lujing Wu
- Division of Genetics, Department of Pediatrics, Program in Immunology, Bioinformatics and Systems Biology Program, Institute for Genomic Medicine, UCSD Center for AIDS Research, University of California San Diego, 9500 Gilman Drive MC 0762, La Jolla, California 92093, USA
| | - Na Li
- Division of Genetics, Department of Pediatrics, Program in Immunology, Bioinformatics and Systems Biology Program, Institute for Genomic Medicine, UCSD Center for AIDS Research, University of California San Diego, 9500 Gilman Drive MC 0762, La Jolla, California 92093, USA
| | - Hui Hui
- Division of Genetics, Department of Pediatrics, Program in Immunology, Bioinformatics and Systems Biology Program, Institute for Genomic Medicine, UCSD Center for AIDS Research, University of California San Diego, 9500 Gilman Drive MC 0762, La Jolla, California 92093, USA
| | - Alex E. Clark
- Division of Infectious Diseases and Global Public Health, Department of Medicine; University of California San Diego, 9500 Gilman Drive MC 0762, La Jolla, California 92093, USA
| | - Qiong Zhang
- Division of Genetics, Department of Pediatrics, Program in Immunology, Bioinformatics and Systems Biology Program, Institute for Genomic Medicine, UCSD Center for AIDS Research, University of California San Diego, 9500 Gilman Drive MC 0762, La Jolla, California 92093, USA
| | - Lingzhi Zhang
- Division of Genetics, Department of Pediatrics, Program in Immunology, Bioinformatics and Systems Biology Program, Institute for Genomic Medicine, UCSD Center for AIDS Research, University of California San Diego, 9500 Gilman Drive MC 0762, La Jolla, California 92093, USA
| | - Aaron F. Carlin
- Division of Infectious Diseases and Global Public Health, Department of Medicine; University of California San Diego, 9500 Gilman Drive MC 0762, La Jolla, California 92093, USA
| | - Tariq M. Rana
- Division of Genetics, Department of Pediatrics, Program in Immunology, Bioinformatics and Systems Biology Program, Institute for Genomic Medicine, UCSD Center for AIDS Research, University of California San Diego, 9500 Gilman Drive MC 0762, La Jolla, California 92093, USA
| |
Collapse
|
215
|
Zanoni I. Aged nasal epithelium is more prone to severe COVID-19. Nat Immunol 2024; 25:722-724. [PMID: 38641720 DOI: 10.1038/s41590-024-01824-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/21/2024]
Affiliation(s)
- Ivan Zanoni
- Harvard Medical School, and Boston Children's Hospital, Division of Immunology, Division of Gastroenterology, Boston, MA, USA.
| |
Collapse
|
216
|
Gygi JP, Maguire C, Patel RK, Shinde P, Konstorum A, Shannon CP, Xu L, Hoch A, Jayavelu ND, Haddad EK, IMPACC Network, Reed EF, Kraft M, McComsey GA, Metcalf JP, Ozonoff A, Esserman D, Cairns CB, Rouphael N, Bosinger SE, Kim-Schulze S, Krammer F, Rosen LB, van Bakel H, Wilson M, Eckalbar WL, Maecker HT, Langelier CR, Steen H, Altman MC, Montgomery RR, Levy O, Melamed E, Pulendran B, Diray-Arce J, Smolen KK, Fragiadakis GK, Becker PM, Sekaly RP, Ehrlich LI, Fourati S, Peters B, Kleinstein SH, Guan L. Integrated longitudinal multiomics study identifies immune programs associated with acute COVID-19 severity and mortality. J Clin Invest 2024; 134:e176640. [PMID: 38690733 PMCID: PMC11060740 DOI: 10.1172/jci176640] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2023] [Accepted: 03/12/2024] [Indexed: 05/03/2024] Open
Abstract
BACKGROUNDPatients hospitalized for COVID-19 exhibit diverse clinical outcomes, with outcomes for some individuals diverging over time even though their initial disease severity appears similar to that of other patients. A systematic evaluation of molecular and cellular profiles over the full disease course can link immune programs and their coordination with progression heterogeneity.METHODSWe performed deep immunophenotyping and conducted longitudinal multiomics modeling, integrating 10 assays for 1,152 Immunophenotyping Assessment in a COVID-19 Cohort (IMPACC) study participants and identifying several immune cascades that were significant drivers of differential clinical outcomes.RESULTSIncreasing disease severity was driven by a temporal pattern that began with the early upregulation of immunosuppressive metabolites and then elevated levels of inflammatory cytokines, signatures of coagulation, formation of neutrophil extracellular traps, and T cell functional dysregulation. A second immune cascade, predictive of 28-day mortality among critically ill patients, was characterized by reduced total plasma Igs and B cells and dysregulated IFN responsiveness. We demonstrated that the balance disruption between IFN-stimulated genes and IFN inhibitors is a crucial biomarker of COVID-19 mortality, potentially contributing to failure of viral clearance in patients with fatal illness.CONCLUSIONOur longitudinal multiomics profiling study revealed temporal coordination across diverse omics that potentially explain the disease progression, providing insights that can inform the targeted development of therapies for patients hospitalized with COVID-19, especially those who are critically ill.TRIAL REGISTRATIONClinicalTrials.gov NCT04378777.FUNDINGNIH (5R01AI135803-03, 5U19AI118608-04, 5U19AI128910-04, 4U19AI090023-11, 4U19AI118610-06, R01AI145835-01A1S1, 5U19AI062629-17, 5U19AI057229-17, 5U19AI125357-05, 5U19AI128913-03, 3U19AI077439-13, 5U54AI142766-03, 5R01AI104870-07, 3U19AI089992-09, 3U19AI128913-03, and 5T32DA018926-18); NIAID, NIH (3U19AI1289130, U19AI128913-04S1, and R01AI122220); and National Science Foundation (DMS2310836).
Collapse
Affiliation(s)
| | - Cole Maguire
- The University of Texas at Austin, Austin, Texas, USA
| | | | - Pramod Shinde
- La Jolla Institute for Immunology, La Jolla, California, USA
| | | | - Casey P. Shannon
- Centre for Heart Lung Innovation, University of British Columbia, Vancouver, Canada
- Prevention of Organ Failure (PROOF) Centre of Excellence, Providence Research, Vancouver, British Columbia, Canada
| | - Leqi Xu
- Yale School of Public Health, New Haven, Connecticut, USA
| | - Annmarie Hoch
- Clinical and Data Coordinating Center (CDCC) and
- Precision Vaccines Program, Boston Children’s Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | | | - Elias K. Haddad
- Drexel University, Tower Health Hospital, Philadelphia, Pennsylvania, USA
| | - IMPACC Network
- The Immunophenotyping Assessment in a COVID-19 Cohort (IMPACC) Network is detailed in Supplemental Acknowledgments
| | - Elaine F. Reed
- David Geffen School of Medicine at the UCLA, Los Angeles, California, USA
| | - Monica Kraft
- Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Grace A. McComsey
- Case Western Reserve University and University Hospitals of Cleveland, Cleveland, Ohio, USA
| | - Jordan P. Metcalf
- Oklahoma University Health Sciences Center, Oklahoma City, Oklahoma, USA
| | - Al Ozonoff
- Clinical and Data Coordinating Center (CDCC) and
- Precision Vaccines Program, Boston Children’s Hospital and Harvard Medical School, Boston, Massachusetts, USA
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
- Department of Pediatrics, Boston Children’s Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | | | - Charles B. Cairns
- Drexel University, Tower Health Hospital, Philadelphia, Pennsylvania, USA
| | | | | | | | - Florian Krammer
- Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Ignaz Semmelweis Institute, Interuniversity Institute for Infection Research, Medical University of Vienna, Vienna, Austria
| | - Lindsey B. Rosen
- National Institute of Allergy and Infectious Diseases (NIAID), NIH, Bethesda, Maryland, USA
| | - Harm van Bakel
- Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | | | | | | | | | - Hanno Steen
- Precision Vaccines Program, Boston Children’s Hospital and Harvard Medical School, Boston, Massachusetts, USA
- Department of Pathology, Boston Children’s Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | | | | | - Ofer Levy
- Precision Vaccines Program, Boston Children’s Hospital and Harvard Medical School, Boston, Massachusetts, USA
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
- Department of Pediatrics, Boston Children’s Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | | | - Bali Pulendran
- Stanford University School of Medicine, Palo Alto, California, USA
| | - Joann Diray-Arce
- Clinical and Data Coordinating Center (CDCC) and
- Precision Vaccines Program, Boston Children’s Hospital and Harvard Medical School, Boston, Massachusetts, USA
- Department of Pediatrics, Boston Children’s Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Kinga K. Smolen
- Precision Vaccines Program, Boston Children’s Hospital and Harvard Medical School, Boston, Massachusetts, USA
- Department of Pediatrics, Boston Children’s Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | | | - Patrice M. Becker
- National Institute of Allergy and Infectious Diseases (NIAID), NIH, Bethesda, Maryland, USA
| | - Rafick P. Sekaly
- Case Western Reserve University and University Hospitals of Cleveland, Cleveland, Ohio, USA
| | | | - Slim Fourati
- Case Western Reserve University and University Hospitals of Cleveland, Cleveland, Ohio, USA
| | - Bjoern Peters
- La Jolla Institute for Immunology, La Jolla, California, USA
- Department of Medicine, UCSD, La Jolla, California, USA
| | | | - Leying Guan
- Yale School of Public Health, New Haven, Connecticut, USA
| |
Collapse
|
217
|
Mohamed ON, Mohamed MRM, Hassan IG, Alakkad AF, Othman A, Setouhi A, Issa AS. The Relationship of Fetuin-A with Coronary Calcification, Carotid Atherosclerosis, and Mortality Risk in Non-Dialysis Chronic Kidney Disease. J Lipid Atheroscler 2024; 13:194-211. [PMID: 38826181 PMCID: PMC11140250 DOI: 10.12997/jla.2024.13.2.194] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2023] [Revised: 12/24/2023] [Accepted: 01/09/2024] [Indexed: 06/04/2024] Open
Abstract
Objective This study investigated the relationship of fetuin-A with coronary calcification, carotid atherosclerosis, and mortality risk in non-dialysis chronic kidney disease (CKD). Methods The study included 135 adult patients with CKD at stages 3-5, who were divided into coronary artery calcification (CAC) and non-CAC groups. We excluded current smokers and individuals with diabetes mellitus, inflammatory conditions, liver diseases, acute kidney failure, chronic hemodialysis, and cancer. We conducted kidney function tests, complete blood counts, and measured serum levels of fetuin-A, tumor necrosis factor-alpha (TNF-α), high-sensitivity C-reactive protein (hs-CRP), interleukin-6 (IL-6), total cholesterol (TC), total triglycerides (TG), high-density lipoprotein cholesterol, and low-density lipoprotein cholesterol. Cardiac spiral computed tomography was used to calculate the CAC score, employing the Agatston method. Carotid ultrasonography was performed to assess carotid intima-media thickness (CIMT) and to detect the presence of plaques. Results CAC patients had considerably higher levels of TNF-α (p<0.001), IL-6 (p<0.001), hs-CRP (p=0.006), TC, TG, parathyroid hormone (PTH) (p<0.001) and phosphorus (p<0.001) than non-CAC patients. They also had significantly lower levels of fetuin-A (p<0.001). Fetuin-A was considerably lower in CKD subgroups as CKD progressed. Fetuin-A (p=0.046), age (p=0.009), TNF-α (p=0.027), IL-6 (p=0.005), TG (p=0.002), PTH (p=0.002), and phosphorus (p=0.004) were significant predictors of CAC. CAC and fetuin-A were strong predictors of all-cause mortality and cardiovascular (CV) mortality. Fetuin-A was a significant predictor of CIMT (p=0.045). Conclusion Fetuin-A reliably predicted CAC and CIMT. Fetuin-A and CAC emerged as significant risk factors for all-cause and CV mortality in non-dialysis CKD.
Collapse
Affiliation(s)
- Osama Nady Mohamed
- Department of Internal Medicine, Faculty of Medicine, Minia University, Minya, Egypt
| | | | - Israa Gamal Hassan
- Department of Internal Medicine, Faculty of Medicine, Minia University, Minya, Egypt
| | - Atef Farouk Alakkad
- Department of Internal Medicine, Faculty of Medicine, Minia University, Minya, Egypt
| | - Ashraf Othman
- Department of Clinical Pathology, Faculty of Medicine, Minia University, Minya, Egypt
| | - Amr Setouhi
- Department of Cardiology, Faculty of Medicine, Minia University, Minya, Egypt
| | - Ahmed S. Issa
- Department of Radiology, Faculty of Medicine, Minia University, Minya, Egypt
| |
Collapse
|
218
|
Xu HC, Pandey P, Ward H, Gorzkiewicz M, Abromavičiūtė D, Tinz C, Müller L, Meyer C, Pandyra AA, Yavas A, Borkhardt A, Esposito I, Lang KS, Lang PA. High-Affinity-Mediated Viral Entry Triggers Innate Affinity Escape Resulting in Type I IFN Resistance and Impaired T Cell Immunity. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2024; 212:1457-1466. [PMID: 38497668 PMCID: PMC11016594 DOI: 10.4049/jimmunol.2300637] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/28/2023] [Accepted: 02/23/2024] [Indexed: 03/19/2024]
Abstract
Increased receptor binding affinity may allow viruses to escape from Ab-mediated inhibition. However, how high-affinity receptor binding affects innate immune escape and T cell function is poorly understood. In this study, we used the lymphocytic choriomeningitis virus (LCMV) murine infection model system to create a mutated LCMV exhibiting higher affinity for the entry receptor α-dystroglycan (LCMV-GPH155Y). We show that high-affinity receptor binding results in increased viral entry, which is associated with type I IFN (IFN-I) resistance, whereas initial innate immune activation was not impaired during high-affinity virus infection in mice. Consequently, IFN-I resistance led to defective antiviral T cell immunity, reduced type II IFN, and prolonged viral replication in this murine model system. Taken together, we show that high-affinity receptor binding of viruses can trigger innate affinity escape including resistance to IFN-I resulting in prolonged viral replication.
Collapse
Affiliation(s)
- Haifeng C. Xu
- Department of Molecular Medicine II, Medical Faculty, Heinrich Heine University, Düsseldorf, Germany
| | - Piyush Pandey
- Department of Molecular Medicine II, Medical Faculty, Heinrich Heine University, Düsseldorf, Germany
| | - Harry Ward
- Department of Molecular Medicine II, Medical Faculty, Heinrich Heine University, Düsseldorf, Germany
| | - Michal Gorzkiewicz
- Department of Molecular Medicine II, Medical Faculty, Heinrich Heine University, Düsseldorf, Germany
- Department of General Biophysics, Faculty of Biology and Environmental Protection, University of Lodz, Lodz, Poland
| | - Džiuljeta Abromavičiūtė
- Department of Molecular Medicine II, Medical Faculty, Heinrich Heine University, Düsseldorf, Germany
| | - Constanze Tinz
- Department of Molecular Medicine II, Medical Faculty, Heinrich Heine University, Düsseldorf, Germany
| | - Lisa Müller
- Institute of Virology, University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Caroline Meyer
- Institute of Virology, University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Aleksandra A. Pandyra
- Department of Pediatric Oncology, Hematology and Clinical Immunology, Medical Faculty, Center of Child and Adolescent Health, Heinrich Heine University, Düsseldorf, Germany
- Institute of Clinical Chemistry and Clinical Pharmacology, University Hospital Bonn, Bonn, Germany
- German Center for Infection Research, Partner Site Bonn-Cologne, Bonn, Germany
| | - Aslihan Yavas
- Institute of Pathology, Medical Faculty, Heinrich Heine University and University Hospital of Düsseldorf, Düsseldorf, Germany
| | - Arndt Borkhardt
- Department of Pediatric Oncology, Hematology and Clinical Immunology, Medical Faculty, Center of Child and Adolescent Health, Heinrich Heine University, Düsseldorf, Germany
| | - Irene Esposito
- Institute of Pathology, Medical Faculty, Heinrich Heine University and University Hospital of Düsseldorf, Düsseldorf, Germany
| | - Karl S. Lang
- Institute of Immunology, Medical Faculty, University of Duisburg-Essen, Essen, Germany
| | - Philipp A. Lang
- Department of Molecular Medicine II, Medical Faculty, Heinrich Heine University, Düsseldorf, Germany
| |
Collapse
|
219
|
Fracella M, Mancino E, Nenna R, Virgillito C, Frasca F, D'Auria A, Sorrentino L, Petrarca L, La Regina D, Matera L, Di Mattia G, Caputo B, Antonelli G, Pierangeli A, Viscidi RP, Midulla F, Scagnolari C. Age-related transcript changes in type I interferon signaling in children and adolescents with long COVID. Eur J Immunol 2024; 54:e2350682. [PMID: 38522030 DOI: 10.1002/eji.202350682] [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: 07/25/2023] [Revised: 02/26/2024] [Accepted: 02/26/2024] [Indexed: 03/25/2024]
Abstract
SARS-CoV-2 typically causes mild symptoms in children, but evidence suggests that persistent immunopathological changes may lead to long COVID (LC). To explore the interplay between LC and innate immunity, we assessed the type I interferon (IFN-I) response in children and adolescents with LC symptoms (LC; n = 28). This was compared with age-matched SARS-CoV-2 recovered participants without LC symptoms (MC; n = 28) and healthy controls (HC; n = 18). We measured the mRNA expression of IFN-I (IFN-α/β/ε/ω), IFN-I receptor (IFNAR1/2), and ISGs (ISG15, ISG56, MxA, IFI27, BST2, LY6E, OAS1, OAS2, OAS3, and MDA5) in PBMCs collected 3-6 months after COVID-19. LC adolescents (12-17 years) had higher transcript levels of IFN-β, IFN-ε, and IFN-ω than HC, whereas LC children (6-11 years) had lower levels than HC. In adolescents, increased levels of IFN-α, IFN-β, and IFN-ω mRNAs were found in the LC group compared with MC, while lower levels were observed in LC children than MC. Adolescents with neurological symptoms had higher IFN-α/β mRNA levels than MC. LC and MC participants showed decreased expression of ISGs and IFNAR1, but increased expression of IFNAR2, than HC. Our results show age-related changes in the expression of transcripts involved in the IFN-I signaling pathway in children and adolescents with LC.
Collapse
Affiliation(s)
- Matteo Fracella
- Department of Molecular Medicine, Laboratory of Virology, Sapienza University of Rome, Rome, Italy
| | - Enrica Mancino
- Department of Pediatrics and Infantile Neuropsychiatry, Sapienza University of Rome, Rome, Italy
| | - Raffaella Nenna
- Department of Pediatrics and Infantile Neuropsychiatry, Sapienza University of Rome, Rome, Italy
| | - Chiara Virgillito
- Department of Public Health and Infectious Diseases, Sapienza University of Rome, Rome, Italy
| | - Federica Frasca
- Department of Molecular Medicine, Laboratory of Virology, Sapienza University of Rome, Rome, Italy
- Department of Public Health and Infectious Diseases, Sapienza University of Rome, Rome, Italy
| | - Alessandra D'Auria
- Department of Molecular Medicine, Laboratory of Virology, Sapienza University of Rome, Rome, Italy
| | - Leonardo Sorrentino
- Department of Molecular Medicine, Laboratory of Virology, Sapienza University of Rome, Rome, Italy
| | - Laura Petrarca
- Department of Pediatrics and Infantile Neuropsychiatry, Sapienza University of Rome, Rome, Italy
| | - Domenico La Regina
- Department of Pediatrics and Infantile Neuropsychiatry, Sapienza University of Rome, Rome, Italy
| | - Luigi Matera
- Department of Pediatrics and Infantile Neuropsychiatry, Sapienza University of Rome, Rome, Italy
| | - Greta Di Mattia
- Department of Pediatrics and Infantile Neuropsychiatry, Sapienza University of Rome, Rome, Italy
| | - Beniamino Caputo
- Department of Public Health and Infectious Diseases, Sapienza University of Rome, Rome, Italy
| | - Guido Antonelli
- Department of Molecular Medicine, Laboratory of Virology, Sapienza University of Rome, Rome, Italy
| | - Alessandra Pierangeli
- Department of Molecular Medicine, Laboratory of Virology, Sapienza University of Rome, Rome, Italy
| | - Raphael P Viscidi
- Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Fabio Midulla
- Department of Pediatrics and Infantile Neuropsychiatry, Sapienza University of Rome, Rome, Italy
| | - Carolina Scagnolari
- Department of Molecular Medicine, Laboratory of Virology, Sapienza University of Rome, Rome, Italy
| |
Collapse
|
220
|
Williams GP, Yu ED, Shapiro K, Wang E, Freuchet A, Frazier A, Lindestam Arlehamn CS, Sette A, da Silva Antunes R. Investigating viral and autoimmune T cell responses associated with post-acute sequelae of COVID-19. Hum Immunol 2024; 85:110770. [PMID: 38433036 PMCID: PMC11144566 DOI: 10.1016/j.humimm.2024.110770] [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: 09/24/2023] [Revised: 02/26/2024] [Accepted: 02/26/2024] [Indexed: 03/05/2024]
Abstract
Post-acute sequelae of COVID-19 (PASC), or Long COVID, is a chronic condition following acute SARS-CoV-2 infection. Symptoms include exertion fatigue, respiratory issues, myalgia, and neurological manifestations such as 'brain fog,' posing concern for their debilitating nature and potential role in other neurological disorders. However, the underlying potential pathogenic mechanisms of the neurological complications of PASC is largely unknown. Herein, we investigated differences in antigen-specific T cell responses from the peripheral blood towards SARS-CoV-2, latent viruses, or neuronal antigens in 14 PASC individuals with neurological manifestations (PASC-N) versus 22 individuals fully recovered from COVID-19. We employed Activation Induced Marker (AIM), ICS and FluoroSpot assays to determine the specificity and magnitude of CD4+ and CD8+ T cell responses towards SARS-CoV-2 (Spike and rest of proteome), latent viruses (CMV, EBV), and several neuronal antigens. Overall, we observed similar antigen-specific T cell frequencies and cytokine effector T cell responses between PASC donors compared to recovered controls for all antigens tested (viral or autoantigen) in both CD4+ and CD8+ T cell compartments. Our findings suggest that PASC-N does not appear to be associated with changes in antigen-specific T cell responses towards a subset of disease-relevant targets, but more studies in a larger cohort are needed to confirm these unaltered responses.
Collapse
Affiliation(s)
- Gregory P Williams
- Division of Vaccine Discovery, La Jolla Institute for Immunology, La Jolla, San Diego, CA, USA
| | - Esther Dawen Yu
- Division of Vaccine Discovery, La Jolla Institute for Immunology, La Jolla, San Diego, CA, USA
| | - Kendra Shapiro
- Division of Vaccine Discovery, La Jolla Institute for Immunology, La Jolla, San Diego, CA, USA
| | - Eric Wang
- Division of Vaccine Discovery, La Jolla Institute for Immunology, La Jolla, San Diego, CA, USA
| | - Antoine Freuchet
- Division of Vaccine Discovery, La Jolla Institute for Immunology, La Jolla, San Diego, CA, USA
| | - April Frazier
- Division of Vaccine Discovery, La Jolla Institute for Immunology, La Jolla, San Diego, CA, USA
| | | | - Alessandro Sette
- Division of Vaccine Discovery, La Jolla Institute for Immunology, La Jolla, San Diego, CA, USA; University of California San Diego School of Medicine, La Jolla, San Diego, CA, USA
| | | |
Collapse
|
221
|
Mao T, Kim J, Peña-Hernández MA, Valle G, Moriyama M, Luyten S, Ott IM, Gomez-Calvo ML, Gehlhausen JR, Baker E, Israelow B, Slade M, Sharma L, Liu W, Ryu C, Korde A, Lee CJ, Silva Monteiro V, Lucas C, Dong H, Yang Y, Yale SARS-CoV-2 Genomic Surveillance Initiative, Gopinath S, Wilen CB, Palm N, Dela Cruz CS, Iwasaki A. Intranasal neomycin evokes broad-spectrum antiviral immunity in the upper respiratory tract. Proc Natl Acad Sci U S A 2024; 121:e2319566121. [PMID: 38648490 PMCID: PMC11067057 DOI: 10.1073/pnas.2319566121] [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: 11/07/2023] [Accepted: 03/15/2024] [Indexed: 04/25/2024] Open
Abstract
Respiratory virus infections in humans cause a broad-spectrum of diseases that result in substantial morbidity and mortality annually worldwide. To reduce the global burden of respiratory viral diseases, preventative and therapeutic interventions that are accessible and effective are urgently needed, especially in countries that are disproportionately affected. Repurposing generic medicine has the potential to bring new treatments for infectious diseases to patients efficiently and equitably. In this study, we found that intranasal delivery of neomycin, a generic aminoglycoside antibiotic, induces the expression of interferon-stimulated genes (ISGs) in the nasal mucosa that is independent of the commensal microbiota. Prophylactic or therapeutic administration of neomycin provided significant protection against upper respiratory infection and lethal disease in a mouse model of COVID-19. Furthermore, neomycin treatment protected Mx1 congenic mice from upper and lower respiratory infections with a highly virulent strain of influenza A virus. In Syrian hamsters, neomycin treatment potently mitigated contact transmission of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). In healthy humans, intranasal application of neomycin-containing Neosporin ointment was well tolerated and effective at inducing ISG expression in the nose in a subset of participants. These findings suggest that neomycin has the potential to be harnessed as a host-directed antiviral strategy for the prevention and treatment of respiratory viral infections.
Collapse
Affiliation(s)
- Tianyang Mao
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT06510
| | - Jooyoung Kim
- Department of Internal Medicine, Section of Pulmonary, Critical Care and Sleep Medicine, Yale University School of Medicine, New Haven, CT06510
- Division of Pulmonary, Allergy, Critical Care, and Sleep Medicine, Department of Medicine, University of Pittsburgh, PittsburghPA15213
| | - Mario A. Peña-Hernández
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT06510
- Department of Microbial Pathogenesis, Yale University School of Medicine, New HavenCT06510
| | - Gabrielee Valle
- Department of Internal Medicine, Section of Pulmonary, Critical Care and Sleep Medicine, Yale University School of Medicine, New Haven, CT06510
| | - Miyu Moriyama
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT06510
| | - Sophia Luyten
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT06510
| | - Isabel M. Ott
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT06510
| | | | - Jeff R Gehlhausen
- Department of Dermatology, Yale University School of Medicine, New Haven, CT06510
| | - Emily Baker
- Department of Dermatology, Yale University School of Medicine, New Haven, CT06510
| | - Benjamin Israelow
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT06510
- Department of Internal Medicine, Section of Infectious Diseases, Yale University School of Medicine, New Haven, CT06510
| | - Martin Slade
- Department of Internal Medicine, Section of Occupational Medicine, Yale University School of Medicine, New Haven, CT06510
| | - Lokesh Sharma
- Department of Internal Medicine, Section of Pulmonary, Critical Care and Sleep Medicine, Yale University School of Medicine, New Haven, CT06510
- Division of Pulmonary, Allergy, Critical Care, and Sleep Medicine, Department of Medicine, University of Pittsburgh, PittsburghPA15213
| | - Wei Liu
- Department of Internal Medicine, Section of Pulmonary, Critical Care and Sleep Medicine, Yale University School of Medicine, New Haven, CT06510
| | - Changwan Ryu
- Department of Internal Medicine, Section of Pulmonary, Critical Care and Sleep Medicine, Yale University School of Medicine, New Haven, CT06510
| | - Asawari Korde
- Department of Internal Medicine, Section of Pulmonary, Critical Care and Sleep Medicine, Yale University School of Medicine, New Haven, CT06510
| | - Chris J. Lee
- Department of Internal Medicine, Section of Pulmonary, Critical Care and Sleep Medicine, Yale University School of Medicine, New Haven, CT06510
| | | | - Carolina Lucas
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT06510
| | - Huiping Dong
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT06510
| | - Yi Yang
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT06510
| | | | - Smita Gopinath
- Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health, Harvard University, Boston, MA02115
| | - Craig B. Wilen
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT06510
- Department of Laboratory Medicine, Yale University School of Medicine, New Haven, CT06510
| | - Noah Palm
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT06510
| | - Charles S. Dela Cruz
- Department of Internal Medicine, Section of Pulmonary, Critical Care and Sleep Medicine, Yale University School of Medicine, New Haven, CT06510
- Division of Pulmonary, Allergy, Critical Care, and Sleep Medicine, Department of Medicine, University of Pittsburgh, PittsburghPA15213
- Veterans Affairs Pittsburgh Healthcare System, Pittsburgh, PA15240
| | - Akiko Iwasaki
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT06510
- Department of Dermatology, Yale University School of Medicine, New Haven, CT06510
- Center for Infection and Immunity, Yale University School of Medicine, New Haven, CT06510
- HHMI, Chevy Chase, MD20815
| |
Collapse
|
222
|
Singh L, Kumar A, Rai M, Basnet B, Rai N, Khanal P, Lai KS, Cheng WH, Asaad AM, Ansari S. Spectrum of COVID-19 induced liver injury: A review report. World J Hepatol 2024; 16:517-536. [PMID: 38689748 PMCID: PMC11056898 DOI: 10.4254/wjh.v16.i4.517] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/25/2023] [Revised: 01/20/2024] [Accepted: 02/28/2024] [Indexed: 04/24/2024] Open
Abstract
The coronavirus disease 2019 (COVID-19) pandemic has caused changes in the global health system, causing significant setbacks in healthcare systems worldwide. This pandemic has also shown resilience, flexibility, and creativity in reacting to the tragedy. The severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection targets most of the respiratory tract, resulting in a severe sickness called acute respiratory distress syndrome that may be fatal in some individuals. Although the lung is the primary organ targeted by COVID-19 viruses, the clinical aspect of the disease is varied and ranges from asymptomatic to respiratory failure. However, due to an unorganized immune response and several affected mechanisms, the liver may also experience liver cell injury, ischemic liver dysfunction, and drug-induced liver injury, which can result in respiratory failure because of the immune system's disordered response and other compromised processes that can end in multisystem organ failure. Patients with liver cirrhosis or those who have impaired immune systems may be more likely than other groups to experience worse results from the SARS-CoV-2 infection. We thus intend to examine the pathogenesis, current therapy, and consequences of liver damage concerning COVID-19.
Collapse
Affiliation(s)
- Lokjan Singh
- Department of Microbiology, Karnali Academy of Health Science, Teaching Hospital, Jumla 21200, Karnali, Nepal
| | - Anil Kumar
- Department of Microbiology, Karnali Academy of Health Science, Teaching Hospital, Jumla 21200, Karnali, Nepal
| | - Maya Rai
- Department of Microbiology, Karnali Academy of Health Science, Teaching Hospital, Jumla 21200, Karnali, Nepal
| | - Bibek Basnet
- Health Sciences, Asian College of Advance Studies, Purbanchal University, Satdobato 24122, Lalitpur, Nepal
| | - Nishant Rai
- Department of Biotechnology, Graphic Era (Deemed to be University), Dehradun 248002, Uttarakhand, India
| | - Pukar Khanal
- Department of Pharmacology & Toxicology, KLE College of Pharmacy, Belagavi, KLE Academy of Higher Education and Research, Belagavi 590010, Karnataka, India
| | - Kok-Song Lai
- Division of Health Sciences, Abu Dhabi Women's College, Higher Colleges of Technology, Abu Dhabi 41012, United Arab Emirates
| | - Wan-Hee Cheng
- Health and Life Sciences, INTI International University, Nilai 71800, Malaysia
| | - Ahmed Morad Asaad
- Department of Microbiology, College of Medicine, Zagazig University, Zagazig 44519, Egypt
| | - Shamshul Ansari
- Division of Health Sciences, Abu Dhabi Women's College, Higher Colleges of Technology, Abu Dhabi 41012, United Arab Emirates.
| |
Collapse
|
223
|
Aoki A, Iwamura C, Kiuchi M, Tsuji K, Sasaki A, Hishiya T, Hirasawa R, Kokubo K, Kuriyama S, Onodera A, Shimada T, Nagaoka T, Ishikawa S, Kojima A, Mito H, Hase R, Kasahara Y, Kuriyama N, Nakamura S, Urushibara T, Kaneda S, Sakao S, Nishida O, Takahashi K, Kimura MY, Motohashi S, Igari H, Ikehara Y, Nakajima H, Suzuki T, Hanaoka H, Nakada TA, Kikuchi T, Nakayama T, Yokote K, Hirahara K. Suppression of Type I Interferon Signaling in Myeloid Cells by Autoantibodies in Severe COVID-19 Patients. J Clin Immunol 2024; 44:104. [PMID: 38647550 PMCID: PMC11035476 DOI: 10.1007/s10875-024-01708-7] [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: 06/07/2023] [Accepted: 04/10/2024] [Indexed: 04/25/2024]
Abstract
PURPOSE Auto-antibodies (auto-abs) to type I interferons (IFNs) have been identified in patients with life-threatening coronavirus disease 2019 (COVID-19), suggesting that the presence of auto-abs may be a risk factor for disease severity. We therefore investigated the mechanism underlying COVID-19 exacerbation induced by auto-abs to type I IFNs. METHODS We evaluated plasma from 123 patients with COVID-19 to measure auto-abs to type I IFNs. We performed single-cell RNA sequencing (scRNA-seq) of peripheral blood mononuclear cells from the patients with auto-abs and conducted epitope mapping of the auto-abs. RESULTS Three of 19 severe and 4 of 42 critical COVID-19 patients had neutralizing auto-abs to type I IFNs. Patients with auto-abs to type I IFNs showed no characteristic clinical features. scRNA-seq from 38 patients with COVID-19 revealed that IFN signaling in conventional dendritic cells and canonical monocytes was attenuated, and SARS-CoV-2-specific BCR repertoires were decreased in patients with auto-abs. Furthermore, auto-abs to IFN-α2 from COVID-19 patients with auto-abs recognized characteristic epitopes of IFN-α2, which binds to the receptor. CONCLUSION Auto-abs to type I IFN found in COVID-19 patients inhibited IFN signaling in dendritic cells and monocytes by blocking the binding of type I IFN to its receptor. The failure to properly induce production of an antibody to SARS-CoV-2 may be a causative factor of COVID-19 severity.
Collapse
Grants
- (S) 26221305 Ministry of Education, Culture, Sports, Science and Technology (MEXT Japan) Grants-in-Aid for Scientific Research
- (B) 20H03685 Ministry of Education, Culture, Sports, Science and Technology (MEXT Japan) Grants-in-Aid for Scientific Research
- (C) 17K08876 Ministry of Education, Culture, Sports, Science and Technology (MEXT Japan) Grants-in-Aid for Scientific Research
- (C) 18K07164 Ministry of Education, Culture, Sports, Science and Technology (MEXT Japan) Grants-in-Aid for Scientific Research
- 19K16683 Ministry of Education, Culture, Sports, Science and Technology (MEXT Japan) Grants-in-Aid for Scientific Research
- (B) JP21H05120 Transformative Research Areas
- (B) JP21H05121 Transformative Research Areas
- JP21ek0410060 Practical Research Project for Allergic Diseases and Immunology (Research on Allergic Diseases and Immunology) from the Japan Agency for Medical Research and Development, AMED
- JP21ek0410082 Practical Research Project for Allergic Diseases and Immunology (Research on Allergic Diseases and Immunology) from the Japan Agency for Medical Research and Development, AMED
- JP19ek0410045 Practical Research Project for Allergic Diseases and Immunology (Research on Allergic Diseases and Immunology) from the Japan Agency for Medical Research and Development, AMED
- JP20gm6110005 AMED-PRIME
- JP21gm1210003 AMED-CREST
- JPMJFR200R JST FOREST Project
- Ministry of Education, Culture, Sports, Science and Technology (MEXT Japan) Grants-in-Aid for Scientific Research
- Transformative Research Areas
- Practical Research Project for Allergic Diseases and Immunology (Research on Allergic Diseases and Immunology) from the Japan Agency for Medical Research and Development, AMED
- JST FOREST Project
- Mochida Memorial Foundation for Medical and Pharmaceutical Research
- MSD Life Science Foundation, Public Interest Incorporated Foundation
- Japanese Respiratory Foundation
- Takeda Science Foundation
- The Japanese Association for Infectious Diseases, Grant for Clinical Research Promotion
Collapse
Affiliation(s)
- Ami Aoki
- Department of Immunology, Graduate School of Medicine, Chiba University, Chiba, 260-8670, Japan
- Department of Respiratory Medicine and Infectious Diseases, Niigata University Graduate School of Medical and Dental Sciences, Niigata, 951-8510, Japan
| | - Chiaki Iwamura
- Department of Immunology, Graduate School of Medicine, Chiba University, Chiba, 260-8670, Japan
- Synergy Institute for Futuristic Mucosal Vaccine Research and Development, Chiba University, Chiba, Japan
| | - Masahiro Kiuchi
- Department of Immunology, Graduate School of Medicine, Chiba University, Chiba, 260-8670, Japan
| | - Kaori Tsuji
- Department of Immunology, Graduate School of Medicine, Chiba University, Chiba, 260-8670, Japan
| | - Atsushi Sasaki
- Department of Immunology, Graduate School of Medicine, Chiba University, Chiba, 260-8670, Japan
| | - Takahisa Hishiya
- Department of Immunology, Graduate School of Medicine, Chiba University, Chiba, 260-8670, Japan
| | - Rui Hirasawa
- Department of Immunology, Graduate School of Medicine, Chiba University, Chiba, 260-8670, Japan
| | - Kota Kokubo
- Department of Immunology, Graduate School of Medicine, Chiba University, Chiba, 260-8670, Japan
| | - Sachiko Kuriyama
- Department of Immunology, Graduate School of Medicine, Chiba University, Chiba, 260-8670, Japan
| | - Atsushi Onodera
- Department of Immunology, Graduate School of Medicine, Chiba University, Chiba, 260-8670, Japan
| | - Tadanaga Shimada
- Department of Emergency and Critical Care Medicine, Graduate School of Medicine, Chiba University, Chiba, 260-8670, Japan
| | - Tetsutaro Nagaoka
- Department of Respiratory Medicine, Juntendo University Faculty of Medicine and Graduate School of Medicine, Tokyo, 113-8431, Japan
| | | | - Akira Kojima
- Funabashi Central Hospital, Chiba, 273-8556, Japan
| | - Haruki Mito
- Department of Infectious Diseases, Japanese Red Cross Narita Hospital, Chiba, 286-0041, Japan
| | - Ryota Hase
- Department of Infectious Diseases, Japanese Red Cross Narita Hospital, Chiba, 286-0041, Japan
| | - Yasunori Kasahara
- Department of Respiratory Medicine, Eastern Chiba Medical Center, Chiba, 283-8686, Japan
| | - Naohide Kuriyama
- Department of Anesthesiology and Critical Care Medicine, School of Medicine, Fujita Health University, Toyoake, Aichi, 470-1192, Japan
| | | | | | - Satoru Kaneda
- Department of Gastroenterology, NHO Chiba Medical Center, Chiba, 260-8606, Japan
| | - Seiichiro Sakao
- Department of Pulmonary Medicine, International University of Health and Welfare Narita Hospital, Chiba, 286-8520, Japan
| | - Osamu Nishida
- Department of Anesthesiology and Critical Care Medicine, School of Medicine, Fujita Health University, Toyoake, Aichi, 470-1192, Japan
| | - Kazuhisa Takahashi
- Department of Respiratory Medicine, Juntendo University Faculty of Medicine and Graduate School of Medicine, Tokyo, 113-8431, Japan
| | - Motoko Y Kimura
- Synergy Institute for Futuristic Mucosal Vaccine Research and Development, Chiba University, Chiba, Japan
- Department of Experimental Immunology, Graduate School of Medicine, Chiba University, Chiba, 260-8670, Japan
| | - Shinichiro Motohashi
- Department of Medical Immunology, Graduate School of Medicine, Chiba University, Chiba, 260-8670, Japan
| | - Hidetoshi Igari
- Department of Infectious Diseases, Chiba University Hospital, Chiba, 260-8677, Japan
- COVID-19 Vaccine Center, Chiba University Hospital, Chiba, 260-8677, Japan
| | - Yuzuru Ikehara
- Department of Pathology, Graduate School of Medicine, Chiba University, Chiba, 260-8670, Japan
| | - Hiroshi Nakajima
- Synergy Institute for Futuristic Mucosal Vaccine Research and Development, Chiba University, Chiba, Japan
- COVID-19 Vaccine Center, Chiba University Hospital, Chiba, 260-8677, Japan
- Department of Allergy and Clinical Immunology, Graduate School of Medicine, Chiba University, Chiba, 260-8670, Japan
| | - Takuji Suzuki
- Synergy Institute for Futuristic Mucosal Vaccine Research and Development, Chiba University, Chiba, Japan
- Department of Respirology, Graduate School of Medicine, Chiba University, Chiba, 260-8670, Japan
| | - Hideki Hanaoka
- Synergy Institute for Futuristic Mucosal Vaccine Research and Development, Chiba University, Chiba, Japan
- Clinical Research Center, Chiba University Hospital, Chiba, 260-8677, Japan
| | - Taka-Aki Nakada
- Department of Emergency and Critical Care Medicine, Graduate School of Medicine, Chiba University, Chiba, 260-8670, Japan
| | - Toshiaki Kikuchi
- Department of Respiratory Medicine and Infectious Diseases, Niigata University Graduate School of Medical and Dental Sciences, Niigata, 951-8510, Japan
| | - Toshinori Nakayama
- Department of Immunology, Graduate School of Medicine, Chiba University, Chiba, 260-8670, Japan.
- AMED-CREST, AMED, 1-8-1 Inohana, Chuo-ku, Chiba, 260-8670, Japan.
| | - Koutaro Yokote
- Department of Endocrinology, Hematology and Gerontology, Graduate School of Medicine, Chiba University, Chiba, 260-8670, Japan
| | - Kiyoshi Hirahara
- Department of Immunology, Graduate School of Medicine, Chiba University, Chiba, 260-8670, Japan.
- Synergy Institute for Futuristic Mucosal Vaccine Research and Development, Chiba University, Chiba, Japan.
- AMED-CREST, AMED, 1-8-1 Inohana, Chuo-ku, Chiba, 260-8670, Japan.
| |
Collapse
|
224
|
Casanova JL, MacMicking JD, Nathan CF. Interferon- γ and infectious diseases: Lessons and prospects. Science 2024; 384:eadl2016. [PMID: 38635718 DOI: 10.1126/science.adl2016] [Citation(s) in RCA: 39] [Impact Index Per Article: 39.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2023] [Accepted: 03/13/2024] [Indexed: 04/20/2024]
Abstract
Infectious diseases continue to claim many lives. Prevention of morbidity and mortality from these diseases would benefit not just from new medicines and vaccines but also from a better understanding of what constitutes protective immunity. Among the major immune signals that mobilize host defense against infection is interferon-γ (IFN-γ), a protein secreted by lymphocytes. Forty years ago, IFN-γ was identified as a macrophage-activating factor, and, in recent years, there has been a resurgent interest in IFN-γ biology and its role in human defense. Here we assess the current understanding of IFN-γ, revisit its designation as an "interferon," and weigh its prospects as a therapeutic against globally pervasive microbial pathogens.
Collapse
Affiliation(s)
- Jean-Laurent Casanova
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, The Rockefeller University, New York, NY 10065, USA
- Howard Hughes Medical Institute, The Rockefeller University, New York, NY 10065, USA
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM, Necker Hospital for Sick Children, 75015 Paris, France
- Imagine Institute, Paris Cité University, 75015 Paris, France
- Department of Pediatrics, Necker Hospital for Sick Children, 75015 Paris, France
| | - John D MacMicking
- Department of Microbial Pathogenesis, Yale University School of Medicine, New Haven, CT 06510, USA
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT 06510, USA
- Yale Systems Biology Institute, Yale University, West Haven, CT 06477, USA
- Howard Hughes Medical Institute, Yale University School of Medicine, New Haven, CT 06510, USA
| | - Carl F Nathan
- Department of Microbiology and Immunology, Weill Cornell Medicine, New York, NY 10065, USA
| |
Collapse
|
225
|
Van Phan H, Tsitsiklis A, Maguire CP, Haddad EK, Becker PM, Kim-Schulze S, Lee B, Chen J, Hoch A, Pickering H, van Zalm P, Altman MC, Augustine AD, Calfee CS, Bosinger S, Cairns CB, Eckalbar W, Guan L, Doni Jayavelu N, Kleinstein SH, Krammer F, Maecker HT, Ozonoff A, Peters B, Rouphael N, IMPACC Network, Montgomery RR, Reed E, Schaenman J, Steen H, Levy O, Diray-Arce J, Langelier CR. Host-microbe multiomic profiling reveals age-dependent immune dysregulation associated with COVID-19 immunopathology. Sci Transl Med 2024; 16:eadj5154. [PMID: 38630846 PMCID: PMC11931290 DOI: 10.1126/scitranslmed.adj5154] [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: 06/30/2023] [Accepted: 03/15/2024] [Indexed: 04/19/2024]
Abstract
Age is a major risk factor for severe coronavirus disease 2019 (COVID-19), yet the mechanisms behind this relationship have remained incompletely understood. To address this, we evaluated the impact of aging on host immune response in the blood and the upper airway, as well as the nasal microbiome in a prospective, multicenter cohort of 1031 vaccine-naïve patients hospitalized for COVID-19 between 18 and 96 years old. We performed mass cytometry, serum protein profiling, anti-severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) antibody assays, and blood and nasal transcriptomics. We found that older age correlated with increased SARS-CoV-2 viral abundance upon hospital admission, delayed viral clearance, and increased type I interferon gene expression in both the blood and upper airway. We also observed age-dependent up-regulation of innate immune signaling pathways and down-regulation of adaptive immune signaling pathways. Older adults had lower naïve T and B cell populations and higher monocyte populations. Over time, older adults demonstrated a sustained induction of pro-inflammatory genes and serum chemokines compared with younger individuals, suggesting an age-dependent impairment in inflammation resolution. Transcriptional and protein biomarkers of disease severity differed with age, with the oldest adults exhibiting greater expression of pro-inflammatory genes and proteins in severe disease. Together, our study finds that aging is associated with impaired viral clearance, dysregulated immune signaling, and persistent and potentially pathologic activation of pro-inflammatory genes and proteins.
Collapse
Affiliation(s)
- Hoang Van Phan
- University of California San Francisco, San Francisco, CA
94115, USA
| | | | | | - Elias K. Haddad
- Drexel University, Tower Health Hospital, Philadelphia, PA
19104, USA
| | - Patrice M. Becker
- National Institute of Allergy and Infectious Diseases,
National Institutes of Health, Bethesda, MD 20814, USA
| | | | - Brian Lee
- Icahn School of Medicine at Mount Sinai, New York, NY
10029, USA
| | - Jing Chen
- Precision Vaccines Program, Boston Children’s
Hospital, Harvard Medical School, Boston, MA 02115, USA
- Research Computing, Department of Information Technology,
Boston Children’s Hospital, Harvard Medical School, Boston, MA 02115,
USA
| | - Annmarie Hoch
- Precision Vaccines Program, Boston Children’s
Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Harry Pickering
- David Geffen School of Medicine, University of California
Los Angeles, Los Angeles, CA 90095, USA
| | - Patrick van Zalm
- Precision Vaccines Program, Boston Children’s
Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Matthew C. Altman
- Benaroya Research Institute, University of Washington,
Seattle, WA 98101, USA
| | - Alison D. Augustine
- National Institute of Allergy and Infectious Diseases,
National Institutes of Health, Bethesda, MD 20814, USA
| | - Carolyn S. Calfee
- University of California San Francisco, San Francisco, CA
94115, USA
| | | | - Charles B. Cairns
- Drexel University, Tower Health Hospital, Philadelphia, PA
19104, USA
| | - Walter Eckalbar
- University of California San Francisco, San Francisco, CA
94115, USA
| | - Leying Guan
- Yale School of Public Health, New Haven, CT 06510,
USA
| | | | | | - Florian Krammer
- Icahn School of Medicine at Mount Sinai, New York, NY
10029, USA
| | | | - Al Ozonoff
- Precision Vaccines Program, Boston Children’s
Hospital, Harvard Medical School, Boston, MA 02115, USA
- Research Computing, Department of Information Technology,
Boston Children’s Hospital, Harvard Medical School, Boston, MA 02115,
USA
| | - Bjoern Peters
- La Jolla Institute for Immunology, La Jolla, CA 92037,
USA
| | | | | | | | - Elaine Reed
- David Geffen School of Medicine, University of California
Los Angeles, Los Angeles, CA 90095, USA
| | - Joanna Schaenman
- David Geffen School of Medicine, University of California
Los Angeles, Los Angeles, CA 90095, USA
| | - Hanno Steen
- Precision Vaccines Program, Boston Children’s
Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Ofer Levy
- Precision Vaccines Program, Boston Children’s
Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Joann Diray-Arce
- Precision Vaccines Program, Boston Children’s
Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Charles R. Langelier
- University of California San Francisco, San Francisco, CA
94115, USA
- Chan Zuckerberg Biohub San Francisco, San Francisco, CA
94158, USA
| |
Collapse
|
226
|
Rossini L, Bonardi CM, Bresolin S, Trevisson E, Marzollo A. Severe Herpes Simplex Encephalitis: an Unusual Presentation of IPEX. J Clin Immunol 2024; 44:100. [PMID: 38625673 DOI: 10.1007/s10875-024-01702-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2023] [Accepted: 04/05/2024] [Indexed: 04/17/2024]
Affiliation(s)
- Linda Rossini
- Pediatric Hematology, Oncology and Stem Cell Transplant Division, University Hospital of Padova, Padova, Italy
- Department of Women's and Children's Health, University Hospital of Padova, Padova, Italy
| | - Claudia Maria Bonardi
- Pediatric Intensive Care Unit, Department of Women's and Children's Health, University Hospital of Padova, Via Giustiniani, 3, 35128, Padova, Italy.
| | - Silvia Bresolin
- Onco-Hematology, Stem Cell Transplant and Gene Therapy, Istituto di Ricerca Pediatrica Foundation - Città della Speranza, Padova, Italy
| | - Eva Trevisson
- Clinical Genetics Unit, Department of Women's and Children's Health, University Hospital of Padova, Padova, Italy
| | - Antonio Marzollo
- Pediatric Hematology, Oncology and Stem Cell Transplant Division, University Hospital of Padova, Padova, Italy
| |
Collapse
|
227
|
Tang S, Man Q, Zhu D, Yu X, Chen R, Wang S, Lu Y, Shi Q, Suo C, Xiong L. Risk factors for progression to severe infection and prolonged viral clearance time in hospitalized elderly patients infected with the Omicron variant of SARS-CoV-2: a retrospective study at Shanghai Fourth People's Hospital, School of Medicine, Tongji University. Front Microbiol 2024; 15:1361197. [PMID: 38686116 PMCID: PMC11056568 DOI: 10.3389/fmicb.2024.1361197] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2023] [Accepted: 03/20/2024] [Indexed: 05/02/2024] Open
Abstract
Introduction In elderly patients infected with the Omicron variant, disease progression to severe infection can result in poor outcomes. This study aimed to identify risk and protective factors associated with disease progression to severe infection and viral clearance time in elderly Omicron-infected patients. Methods Shanghai Fourth People's Hospital, School of Medicine, Tongji University, was officially designated to provide treatment to patients with COVID-19. This study was conducted on confirmed Omicron cases admitted to the hospital between 10 April 2022 and 21 June 2022. In total, 1,568 patients aged 65 years or older were included. We conducted a retrospective, observational study using logistic regression to analyze risk and protective factors for the development of severe disease and Cox proportional hazards regression models to analyze factors influencing viral clearance time. Results Aged over 80 years, having 2 or more comorbidities, combined cerebrovascular disease, chronic neurological disease, and mental disorders were associated with the development of severe disease, and full vaccination was a protective factor. Furthermore, aged over 80 years, combined chronic respiratory disease, chronic renal disease, cerebrovascular disease, mental disorders, and high viral load were associated with prolonged viral clearance time, and full vaccination was a protective factor. Discussion This study analyzed risk factors for progression to severe infection and prolonged viral clearance time in hospitalized elderly Omicron-infected patients. Aged patients with comorbidities had a higher risk of developing severe infection and had longer viral clearance, while vaccination protected them against the Omicron infection.
Collapse
Affiliation(s)
- Siqi Tang
- Shanghai Institute of Infectious Disease and Biosecurity, Fudan University, Shanghai, China
- Department of Epidemiology, Ministry of Education, Key Laboratory of Public Health Safety, School of Public Health, Fudan University, Shanghai, China
| | - Qiuhong Man
- Department of Clinical Laboratory, Shanghai Fourth People's Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Dongliang Zhu
- Department of Epidemiology, Ministry of Education, Key Laboratory of Public Health Safety, School of Public Health, Fudan University, Shanghai, China
| | - Xueying Yu
- Department of Clinical Laboratory, Shanghai Fourth People's Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Ruilin Chen
- Department of Epidemiology, Ministry of Education, Key Laboratory of Public Health Safety, School of Public Health, Fudan University, Shanghai, China
| | - Shuo Wang
- Department of Epidemiology, Ministry of Education, Key Laboratory of Public Health Safety, School of Public Health, Fudan University, Shanghai, China
| | - Yihan Lu
- Shanghai Institute of Infectious Disease and Biosecurity, Fudan University, Shanghai, China
- Department of Epidemiology, Ministry of Education, Key Laboratory of Public Health Safety, School of Public Health, Fudan University, Shanghai, China
| | - Qiqing Shi
- Department of Anesthesiology and Perioperative Medicine, Shanghai Fourth People's Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Chen Suo
- Shanghai Institute of Infectious Disease and Biosecurity, Fudan University, Shanghai, China
- Department of Epidemiology, Ministry of Education, Key Laboratory of Public Health Safety, School of Public Health, Fudan University, Shanghai, China
| | - Lize Xiong
- Department of Anesthesiology and Perioperative Medicine, Shanghai Fourth People's Hospital, School of Medicine, Tongji University, Shanghai, China
- Shanghai Key Laboratory of Anesthesiology and Brain Functional Modulation, Translational Research Institute of Brain and Brain-Like Intelligence, Shanghai Fourth People's Hospital, School of Medicine, Tongji University, Shanghai, China
| |
Collapse
|
228
|
Alba-Cano T, Fernández-Cruz E, Alonso R, Muñoz-Gómez S, Pérez de Diego R, García Martínez E, Sánchez-Mateos P, Navarro Caspistegui J, Martín López M, Gil-Herrera J. Lack of Specific Immune Response after Five Doses of mRNA SARS-CoV-2 Vaccine in a Patient with CD4 + T-Cell Lymphopenia but Preserved Responses to CMV. Vaccines (Basel) 2024; 12:386. [PMID: 38675768 PMCID: PMC11054516 DOI: 10.3390/vaccines12040386] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2024] [Revised: 03/15/2024] [Accepted: 03/21/2024] [Indexed: 04/28/2024] Open
Abstract
Immunogenicity of SARS-CoV-2 mRNA vaccines is highly heterogeneous in patients with inborn errors of immunity (IEIs). This case report analyzes the immune response to mRNA COVID-19 two-dose primary vaccination followed by three boosters in an IEI patient with marked CD4+ T-cell cytopenia and diminished thymic output, in comparison with that raised against latent, chronic cytomegalovirus (CMV) infection. Serum IgG antibodies anti-spike (S) protein of SARS-CoV-2 and anti-CMV were both determined by chemiluminescent microparticle immunoassays (CMIAs). SARS-CoV-2 and CMV memory CD4+ T-cell responses were simultaneously evaluated in vitro using an activation-induced marker (AIM) assay via multicolor flow cytometry. Throughout the 2-year follow-up that included the administration of five doses of SARS-CoV-2 mRNA vaccines, cellular anti-SARS-CoV-2-specific responses remained consistently negative, with extremely weak humoral responses, while the patient showed in vitro persistent CD4+ T-cell reactivity to CMV peptides and high-IgG CMV-specific titers. The assessment of immune responses to vaccines and prevalent viruses is essential in IEI patients in order to take adequate preventive measures.
Collapse
Affiliation(s)
- Trinidad Alba-Cano
- Division of Immunology, Hospital General Universitario “Gregorio Marañón”, 28007 Madrid, Spain; (T.A.-C.); (E.F.-C.); (S.M.-G.); (E.G.M.); (J.N.C.); (M.M.L.)
| | - Eduardo Fernández-Cruz
- Division of Immunology, Hospital General Universitario “Gregorio Marañón”, 28007 Madrid, Spain; (T.A.-C.); (E.F.-C.); (S.M.-G.); (E.G.M.); (J.N.C.); (M.M.L.)
- Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), 28007 Madrid, Spain
| | - Roberto Alonso
- Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), 28007 Madrid, Spain
- Department of Clinical Microbiology and Infectious Diseases, Hospital General Universitario Gregorio Marañón and CIBER (Centro de Investigación Biomédicas en Red) de Enfermedades Respiratorias, CIBERES, 08028 Barcelona, Spain;
| | - Sara Muñoz-Gómez
- Division of Immunology, Hospital General Universitario “Gregorio Marañón”, 28007 Madrid, Spain; (T.A.-C.); (E.F.-C.); (S.M.-G.); (E.G.M.); (J.N.C.); (M.M.L.)
| | - Rebeca Pérez de Diego
- Laboratory of Immunogenetics of Human Diseases, Innate Immunity Group, IdiPAZ Institute for Health Research, La Paz Hospital, 28046 Madrid, Spain;
| | - Elena García Martínez
- Division of Immunology, Hospital General Universitario “Gregorio Marañón”, 28007 Madrid, Spain; (T.A.-C.); (E.F.-C.); (S.M.-G.); (E.G.M.); (J.N.C.); (M.M.L.)
| | - Paloma Sánchez-Mateos
- Department of Immunology, Ophthalmology and ENT, School of Medicine, Universidad Complutense, 28040 Madrid, Spain;
| | - Joaquín Navarro Caspistegui
- Division of Immunology, Hospital General Universitario “Gregorio Marañón”, 28007 Madrid, Spain; (T.A.-C.); (E.F.-C.); (S.M.-G.); (E.G.M.); (J.N.C.); (M.M.L.)
- Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), 28007 Madrid, Spain
| | - Mónica Martín López
- Division of Immunology, Hospital General Universitario “Gregorio Marañón”, 28007 Madrid, Spain; (T.A.-C.); (E.F.-C.); (S.M.-G.); (E.G.M.); (J.N.C.); (M.M.L.)
| | - Juana Gil-Herrera
- Division of Immunology, Hospital General Universitario “Gregorio Marañón”, 28007 Madrid, Spain; (T.A.-C.); (E.F.-C.); (S.M.-G.); (E.G.M.); (J.N.C.); (M.M.L.)
- Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), 28007 Madrid, Spain
- Department of Immunology, Ophthalmology and ENT, School of Medicine, Universidad Complutense, 28040 Madrid, Spain;
| |
Collapse
|
229
|
Mahla RS, Dustin LB. COVID-19 vaccination and autoimmunity: Do we need more evidence? J Med Virol 2024; 96:e29623. [PMID: 38639266 DOI: 10.1002/jmv.29623] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2023] [Revised: 04/09/2024] [Accepted: 04/12/2024] [Indexed: 04/20/2024]
Affiliation(s)
- Ranjeet Singh Mahla
- Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, Medical Science Division, Kennedy Institute of Rheumatology, University of Oxford, Oxford, UK
| | - Lynn B Dustin
- Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, Medical Science Division, Kennedy Institute of Rheumatology, University of Oxford, Oxford, UK
| |
Collapse
|
230
|
Zhang Q, Kisand K, Feng Y, Rinchai D, Jouanguy E, Cobat A, Casanova JL, Zhang SY. In search of a function for human type III interferons: insights from inherited and acquired deficits. Curr Opin Immunol 2024; 87:102427. [PMID: 38781720 PMCID: PMC11209856 DOI: 10.1016/j.coi.2024.102427] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2023] [Revised: 03/19/2024] [Accepted: 05/06/2024] [Indexed: 05/25/2024]
Abstract
The essential and redundant functions of human type I and II interferons (IFNs) have been delineated over the last three decades by studies of patients with inborn errors of immunity or their autoimmune phenocopies, but much less is known about type III IFNs. Patients with cells that do not respond to type III IFNs due to inherited IL10RB deficiency display no overt viral disease, and their inflammatory disease phenotypes can be explained by defective signaling via other interleukine10RB-dependent pathways. Moreover, patients with inherited deficiencies of interferon-stimulated gene factor 3 (ISGF-3) (STAT1, STAT2, IRF9) present viral diseases also seen in patients with inherited deficiencies of the type I IFN receptor (IFNAR1/2). Finally, patients with autoantibodies neutralizing type III IFNs have no obvious predisposition to viral disease. Current findings thus suggest that type III IFNs are largely redundant in humans. The essential functions of human type III IFNs, particularly in antiviral defenses, remain to be discovered.
Collapse
Affiliation(s)
- Qian Zhang
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, The Rockefeller University, New York, USA; Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM U1163, Necker Hospital for Sick Children, Paris, France; Paris Cité University, Imagine Institute, Paris, France.
| | - Kai Kisand
- Institute of Biomedicine and Translational Medicine, University of Tartu, Tartu, Estonia
| | - Yi Feng
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, The Rockefeller University, New York, USA
| | - Darawan Rinchai
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, The Rockefeller University, New York, USA
| | - Emmanuelle Jouanguy
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, The Rockefeller University, New York, USA; Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM U1163, Necker Hospital for Sick Children, Paris, France; Paris Cité University, Imagine Institute, Paris, France
| | - Aurélie Cobat
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, The Rockefeller University, New York, USA; Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM U1163, Necker Hospital for Sick Children, Paris, France; Paris Cité University, Imagine Institute, Paris, France
| | - Jean-Laurent Casanova
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, The Rockefeller University, New York, USA; Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM U1163, Necker Hospital for Sick Children, Paris, France; Paris Cité University, Imagine Institute, Paris, France; Department of Pediatrics, Necker Hospital for Sick Children, AP-HP, Paris, France; Howard Hughes Medical Institute, New York, USA
| | - Shen-Ying Zhang
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, The Rockefeller University, New York, USA; Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM U1163, Necker Hospital for Sick Children, Paris, France; Paris Cité University, Imagine Institute, Paris, France
| |
Collapse
|
231
|
Tóth E, Fagyas M, Nagy B, Siket IM, Szőke B, Mártha L, Mahdi M, Erdősi G, Pólik Z, Kappelmayer J, Papp Z, Borbély A, Szabó T, Balla J, Balla G, Bácsi A, Szekanecz Z, Bai P, Tóth A. Distinct subsets of anti-pulmonary autoantibodies correlate with disease severity and survival in severe COVID-19 patients. GeroScience 2024; 46:1561-1574. [PMID: 37656328 PMCID: PMC10828368 DOI: 10.1007/s11357-023-00887-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2023] [Accepted: 07/24/2023] [Indexed: 09/02/2023] Open
Abstract
Autoantibodies targeting the lung tissue were identified in severe COVID-19 patients in this retrospective study. Fifty-three percent of 104 patients developed anti-pulmonary antibodies, the majority of which were IgM class, suggesting that they developed upon infection with SARS-CoV-2. Anti-pulmonary antibodies correlated with worse pulmonary function and a higher risk of multiorgan failure that was further aggravated if 3 or more autoantibody clones were simultaneously present (multi-producers). Multi-producer patients were older than the patients with less or no autoantibodies. One of the identified autoantibodies (targeting a pulmonary protein of ~ 50 kDa) associated with worse clinical outcomes, including mortality. In summary, severe COVID-19 is associated with the development of lung-specific autoantibodies, which may worsen the clinical outcome. Tissue proteome-wide tests, such as the ones applied here, can be used to detect autoimmunity in the post-COVID state to identify the cause of symptoms and to reveal a new target for treatment.
Collapse
Affiliation(s)
- Emese Tóth
- Department of Medical Chemistry, Faculty of Medicine, University of Debrecen, 4032, Debrecen, Hungary
- Center of Excellence, The Hungarian Academy of Sciences, Budapest, Hungary
| | - Miklós Fagyas
- Division of Cardiology, Department of Cardiology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
- Division of Clinical Physiology, Department of Cardiology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Béla Nagy
- Department of Laboratory Medicine, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Ivetta Mányiné Siket
- Division of Clinical Physiology, Department of Cardiology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Blanka Szőke
- Division of Clinical Physiology, Department of Cardiology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Lilla Mártha
- Division of Clinical Physiology, Department of Cardiology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Mohamed Mahdi
- Department of Biochemistry and Molecular Biology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Gábor Erdősi
- Division of Clinical Physiology, Department of Cardiology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Zsófia Pólik
- Division of Clinical Physiology, Department of Cardiology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - János Kappelmayer
- Department of Laboratory Medicine, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Zoltán Papp
- Division of Clinical Physiology, Department of Cardiology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
- ELKH-UD Vascular Biology and Myocardial Pathophysiology Research Group, Hungarian Academy of Sciences, Budapest, Hungary
- Research Center for Molecular Medicine, Faculty of Medicine, University of Debrecen, 4032, Debrecen, Hungary
| | - Attila Borbély
- Division of Cardiology, Department of Cardiology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Tamás Szabó
- Department of Pediatrics, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - József Balla
- ELKH-UD Vascular Biology and Myocardial Pathophysiology Research Group, Hungarian Academy of Sciences, Budapest, Hungary
- Department of Internal Medicine, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - György Balla
- ELKH-UD Vascular Biology and Myocardial Pathophysiology Research Group, Hungarian Academy of Sciences, Budapest, Hungary
- Department of Pediatrics, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Attila Bácsi
- Department of Immunology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Zoltán Szekanecz
- Department of Rheumatology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Péter Bai
- Department of Medical Chemistry, Faculty of Medicine, University of Debrecen, 4032, Debrecen, Hungary.
- Center of Excellence, The Hungarian Academy of Sciences, Budapest, Hungary.
- Research Center for Molecular Medicine, Faculty of Medicine, University of Debrecen, 4032, Debrecen, Hungary.
- MTA-DE Cell Biology and Signaling Research Group ELKH, Debrecen, 4032, Hungary.
- MTA-DE Lendület Laboratory of Cellular Metabolism, 4032, Debrecen, Hungary.
| | - Attila Tóth
- Division of Clinical Physiology, Department of Cardiology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary.
- ELKH-UD Vascular Biology and Myocardial Pathophysiology Research Group, Hungarian Academy of Sciences, Budapest, Hungary.
- Research Center for Molecular Medicine, Faculty of Medicine, University of Debrecen, 4032, Debrecen, Hungary.
| |
Collapse
|
232
|
Svensson Akusjärvi S, Zanoni I. Yin and yang of interferons: lessons from the coronavirus disease 2019 (COVID-19) pandemic. Curr Opin Immunol 2024; 87:102423. [PMID: 38776716 PMCID: PMC11162909 DOI: 10.1016/j.coi.2024.102423] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2023] [Revised: 03/05/2024] [Accepted: 05/06/2024] [Indexed: 05/25/2024]
Abstract
The host immune response against severe acute respiratory syndrome coronavirus 2 includes the induction of a group of natural antiviral cytokines called interferons (IFNs). Although originally recognized for their ability to potently counteract infections, the mechanistic functions of IFNs in patients with varying severities of coronavirus disease 2019 (COVID-19) have highlighted a more complex scenario. Cellular and molecular analyses have revealed that timing, location, and subtypes of IFNs produced during severe acute respiratory syndrome coronavirus 2 infection play a major role in determining disease progression and severity. In this review, we summarize what the COVID-19 pandemic has taught us about the protective and detrimental roles of IFNs during the inflammatory response elicited against a new respiratory virus across different ages and its longitudinal consequences in driving the development of long COVID-19.
Collapse
Affiliation(s)
- Sara Svensson Akusjärvi
- Harvard Medical School, Division of Immunology, Boston Children's Hospital, Boston, MA, USA; Division of Immunology and Allergy, Department of Medicine Solna, Karolinska Institutet, Stockholm, Sweden
| | - Ivan Zanoni
- Harvard Medical School, Division of Immunology, Boston Children's Hospital, Boston, MA, USA; Division of Gastroenterology, Boston Children's Hospital, Boston, MA, USA.
| |
Collapse
|
233
|
Kundura L, Cezar R, Ballongue E, André S, Michel M, Mettling C, Lozano C, Vincent T, Muller L, Lefrant JY, Roger C, Claret PG, Duvnjak S, Loubet P, Sotto A, Tran TA, Estaquier J, Corbeau P. Low Percentage of Perforin-Expressing NK Cells during Severe SARS-CoV-2 Infection: Consumption Rather than Primary Deficiency. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2024; 212:1105-1112. [PMID: 38345346 DOI: 10.4049/jimmunol.2300359] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/25/2023] [Accepted: 12/05/2023] [Indexed: 03/20/2024]
Abstract
Genetic defects in the ability to deliver effective perforin have been reported in patients with hemophagocytic lymphohistiocytosis. We tested the hypothesis that a primary perforin deficiency might also be causal in severe SARS-CoV-2 infection. We recruited 54 volunteers confirmed as being SARS-CoV-2-infected by RT-PCR and admitted to intensive care units or non-intensive care units and age- and sex-matched healthy controls. Compared with healthy controls, the percentage of perforin-expressing CD3-CD56+ NK cells quantified by flow cytometry was low in COVID-19 patients (69.9 ± 17.7 versus 78.6 ± 14.6%, p = 0.026). There was no correlation between the proportions of perforin-positive NK cells and T8 lymphocytes. Moreover, the frequency of NK cells producing perforin was neither linked to disease severity nor predictive of death. Although IL-6 is known to downregulate perforin production in NK cells, we did not find any link between perforin expression and IL-6 plasma level. However, we unveiled a negative correlation between the degranulation marker CD107a and perforin expression in NK cells (r = -0.488, p = 10-4). PRF1 gene expression and the frequency of NK cells harboring perforin were normal in patients 1 y after acute SARS-CoV-2 infection. A primary perforin defect does not seem to be a driver of COVID-19 because NK perforin expression is 1) linked neither to T8 perforin expression nor to disease severity, 2) inversely correlated with NK degranulation, and 3) normalized at distance from acute infection. Thus, the cause of low frequency of perforin-positive NK cells appears, rather, to be consumption.
Collapse
Affiliation(s)
- Lucy Kundura
- Institute of Human Genetics, UMR9002, CNRS and Montpellier University, Montpellier, France
| | - Renaud Cezar
- Immunology Department, Nîmes University Hospital, Nîmes, France
| | - Emma Ballongue
- Institute of Human Genetics, UMR9002, CNRS and Montpellier University, Montpellier, France
| | - Sonia André
- INSERM U1124, Université de Paris, Paris, France
| | - Moïse Michel
- Immunology Department, Nîmes University Hospital, Nîmes, France
| | - Clément Mettling
- Institute of Human Genetics, UMR9002, CNRS and Montpellier University, Montpellier, France
| | - Claire Lozano
- Immunology Department, Montpellier University Hospital, Montpellier, France
| | - Thierry Vincent
- Immunology Department, Montpellier University Hospital, Montpellier, France
| | - Laurent Muller
- Surgical Intensive Care Department, Nîmes University Hospital, Nîmes, France
| | - Jean-Yves Lefrant
- Surgical Intensive Care Department, Nîmes University Hospital, Nîmes, France
| | - Claire Roger
- Surgical Intensive Care Department, Nîmes University Hospital, Nîmes, France
| | - Pierre-Géraud Claret
- Medical and Surgical Emergency Department, Nîmes University Hospital, Nîmes, France
| | - Sandra Duvnjak
- Gerontology Department, Nîmes University Hospital, Nîmes, France
| | - Paul Loubet
- *Infectious Diseases Department, Nîmes University Hospital, Nîmes, France
| | - Albert Sotto
- *Infectious Diseases Department, Nîmes University Hospital, Nîmes, France
| | - Tu-Anh Tran
- Pediatrics Department, Nîmes University Hospital, Nîmes, France
| | - Jérôme Estaquier
- INSERM U1124, Université de Paris, Paris, France
- Laval University Research Center; Quebec City, Quebec, Canada
| | - Pierre Corbeau
- Institute of Human Genetics, UMR9002, CNRS and Montpellier University, Montpellier, France
- Immunology Department, Nîmes University Hospital, Nîmes, France
| |
Collapse
|
234
|
Berber E, Mulik S, Rouse BT. Meeting the Challenge of Controlling Viral Immunopathology. Int J Mol Sci 2024; 25:3935. [PMID: 38612744 PMCID: PMC11011832 DOI: 10.3390/ijms25073935] [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: 02/14/2024] [Revised: 03/27/2024] [Accepted: 03/29/2024] [Indexed: 04/14/2024] Open
Abstract
The mission of this review is to identify immune-damaging participants involved in antiviral immunoinflammatory lesions. We argue these could be targeted and their activity changed selectively by maneuvers that, at the same time, may not diminish the impact of components that help resolve lesions. Ideally, we need to identify therapeutic approaches that can reverse ongoing lesions that lack unwanted side effects and are affordable to use. By understanding the delicate balance between immune responses that cause tissue damage and those that aid in resolution, novel strategies can be developed to target detrimental immune components while preserving the beneficial ones. Some strategies involve rebalancing the participation of immune components using various approaches, such as removing or blocking proinflammatory T cell products, expanding regulatory cells, restoring lost protective cell function, using monoclonal antibodies (moAb) to counteract inhibitory molecules, and exploiting metabolic differences between inflammatory and immuno-protective responses. These strategies can help reverse ongoing viral infections. We explain various approaches, from model studies and some clinical evidence, that achieve innate and adaptive immune rebalancing, offering insights into potential applications for controlling chronic viral-induced lesions.
Collapse
Affiliation(s)
- Engin Berber
- Infection Biology, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA;
| | - Sachin Mulik
- Center for Biomedical Research, The University of Texas Health Science Center at Tyler, Tyler, TX 75708, USA;
| | - Barry T. Rouse
- College of Veterinary Medicine, University of Tennessee, Knoxville, TN 37996, USA
| |
Collapse
|
235
|
Phan T, Zitzmann C, Chew KW, Smith DM, Daar ES, Wohl DA, Eron JJ, Currier JS, Hughes MD, Choudhary MC, Deo R, Li JZ, Ribeiro RM, Ke R, Perelson AS, for the ACTIV-2/A5401 Study Team. Modeling the emergence of viral resistance for SARS-CoV-2 during treatment with an anti-spike monoclonal antibody. PLoS Pathog 2024; 20:e1011680. [PMID: 38635853 PMCID: PMC11060554 DOI: 10.1371/journal.ppat.1011680] [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: 09/13/2023] [Revised: 04/30/2024] [Accepted: 03/18/2024] [Indexed: 04/20/2024] Open
Abstract
To mitigate the loss of lives during the COVID-19 pandemic, emergency use authorization was given to several anti-SARS-CoV-2 monoclonal antibody (mAb) therapies for the treatment of mild-to-moderate COVID-19 in patients with a high risk of progressing to severe disease. Monoclonal antibodies used to treat SARS-CoV-2 target the spike protein of the virus and block its ability to enter and infect target cells. Monoclonal antibody therapy can thus accelerate the decline in viral load and lower hospitalization rates among high-risk patients with variants susceptible to mAb therapy. However, viral resistance has been observed, in some cases leading to a transient viral rebound that can be as large as 3-4 orders of magnitude. As mAbs represent a proven treatment choice for SARS-CoV-2 and other viral infections, evaluation of treatment-emergent mAb resistance can help uncover underlying pathobiology of SARS-CoV-2 infection and may also help in the development of the next generation of mAb therapies. Although resistance can be expected, the large rebounds observed are much more difficult to explain. We hypothesize replenishment of target cells is necessary to generate the high transient viral rebound. Thus, we formulated two models with different mechanisms for target cell replenishment (homeostatic proliferation and return from an innate immune response antiviral state) and fit them to data from persons with SARS-CoV-2 treated with a mAb. We showed that both models can explain the emergence of resistant virus associated with high transient viral rebounds. We found that variations in the target cell supply rate and adaptive immunity parameters have a strong impact on the magnitude or observability of the viral rebound associated with the emergence of resistant virus. Both variations in target cell supply rate and adaptive immunity parameters may explain why only some individuals develop observable transient resistant viral rebound. Our study highlights the conditions that can lead to resistance and subsequent viral rebound in mAb treatments during acute infection.
Collapse
Affiliation(s)
- Tin Phan
- Theoretical Biology & Biophysics, Los Alamos National Laboratory, Los Alamos, New Mexico, United States of America
| | - Carolin Zitzmann
- Theoretical Biology & Biophysics, Los Alamos National Laboratory, Los Alamos, New Mexico, United States of America
| | - Kara W. Chew
- Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, California, United States of America
| | - Davey M. Smith
- Department of Medicine, University of California, San Diego, California, United States of America
| | - Eric S. Daar
- Lundquist Institute at Harbor-UCLA Medical Center, Torrance, California, United States of America
| | - David A. Wohl
- Department of Medicine, University of North Carolina at Chapel Hill School of Medicine, Chapel Hill, North Carolina, United States of America
| | - Joseph J. Eron
- Department of Medicine, University of North Carolina at Chapel Hill School of Medicine, Chapel Hill, North Carolina, United States of America
| | - Judith S. Currier
- Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, California, United States of America
| | - Michael D. Hughes
- Harvard T.H. Chan School of Public Health, Boston, Massachusetts, United States of America
| | - Manish C. Choudhary
- Department of Medicine, Division of Infectious Diseases, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Rinki Deo
- Department of Medicine, Division of Infectious Diseases, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Jonathan Z. Li
- Department of Medicine, Division of Infectious Diseases, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Ruy M. Ribeiro
- Theoretical Biology & Biophysics, Los Alamos National Laboratory, Los Alamos, New Mexico, United States of America
| | - Ruian Ke
- Theoretical Biology & Biophysics, Los Alamos National Laboratory, Los Alamos, New Mexico, United States of America
| | - Alan S. Perelson
- Theoretical Biology & Biophysics, Los Alamos National Laboratory, Los Alamos, New Mexico, United States of America
- Santa Fe Institute, Santa Fe, New Mexico, United States of America
| | | |
Collapse
|
236
|
Morand E, Merola JF, Tanaka Y, Gladman D, Fleischmann R. TYK2: an emerging therapeutic target in rheumatic disease. Nat Rev Rheumatol 2024; 20:232-240. [PMID: 38467779 DOI: 10.1038/s41584-024-01093-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/09/2024] [Indexed: 03/13/2024]
Abstract
Tyrosine kinase 2 (TYK2) is a member of the JAK kinase family of intracellular signalling molecules. By participating in signalling pathways downstream of type I interferons, IL-12, IL-23 and IL-10, TYK2 elicits a distinct set of immune events to JAK1, JAK2 and JAK3. TYK2 polymorphisms have been associated with susceptibility to various rheumatic diseases including systemic lupus erythematosus and dermatomyositis. In vitro and animal studies substantiate these findings, highlighting a role for TYK2 in diseases currently managed by antagonists of cytokines that signal through TYK2. Various inhibitors of TYK2 have now been studied in human disease, and one of these inhibitors, deucravacitinib, has now been approved for the treatment of psoriasis. Phase II trials of deucravacitinib have also reported positive results in the treatment of psoriatic arthritis and systemic lupus erythematosus, with a preliminary safety profile that seems to differ from that of the JAK1, JAK2 and JAK3 inhibitors. Two other inhibitors of TYK2, brepocitinib and ropsacitinib, are also in earlier stages of clinical trials. Overall, TYK2 inhibitors hold promise for the treatment of a distinct spectrum of autoimmune diseases and could potentially have a safety profile that differs from other JAK inhibitors.
Collapse
Affiliation(s)
- Eric Morand
- Centre for Inflammatory Diseases, Monash University, and Department of Rheumatology, Monash Health, Clayton, Victoria, Australia.
| | - Joseph F Merola
- Department of Medicine, Division of Rheumatology and Department of Dermatology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Yoshiya Tanaka
- The First Department of Internal Medicine, University of Occupational and Environmental Health, Japan, Kitakyushu, Japan
| | - Dafna Gladman
- Schroeder Arthritis Institute, Krembil Research Institute, Toronto Western Hospital, Division of Rheumatology University of Toronto, Toronto, Ontario, Canada
| | - Roy Fleischmann
- Metroplex Clinical Research Center, University of Texas Southwestern Medical Center, Dallas, TX, USA
| |
Collapse
|
237
|
Martin E, Winter S, Garcin C, Tanita K, Hoshino A, Lenoir C, Fournier B, Migaud M, Boutboul D, Simonin M, Fernandes A, Bastard P, Le Voyer T, Roupie AL, Ben Ahmed Y, Leruez-Ville M, Burgard M, Rao G, Ma CS, Masson C, Soudais C, Picard C, Bustamante J, Tangye SG, Cheikh N, Seppänen M, Puel A, Daly M, Casanova JL, Neven B, Fischer A, Latour S. Role of IL-27 in Epstein-Barr virus infection revealed by IL-27RA deficiency. Nature 2024; 628:620-629. [PMID: 38509369 DOI: 10.1038/s41586-024-07213-6] [Citation(s) in RCA: 16] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2022] [Accepted: 02/20/2024] [Indexed: 03/22/2024]
Abstract
Epstein-Barr virus (EBV) infection can engender severe B cell lymphoproliferative diseases1,2. The primary infection is often asymptomatic or causes infectious mononucleosis (IM), a self-limiting lymphoproliferative disorder3. Selective vulnerability to EBV has been reported in association with inherited mutations impairing T cell immunity to EBV4. Here we report biallelic loss-of-function variants in IL27RA that underlie an acute and severe primary EBV infection with a nevertheless favourable outcome requiring a minimal treatment. One mutant allele (rs201107107) was enriched in the Finnish population (minor allele frequency = 0.0068) and carried a high risk of severe infectious mononucleosis when homozygous. IL27RA encodes the IL-27 receptor alpha subunit5,6. In the absence of IL-27RA, phosphorylation of STAT1 and STAT3 by IL-27 is abolished in T cells. In in vitro studies, IL-27 exerts a synergistic effect on T-cell-receptor-dependent T cell proliferation7 that is deficient in cells from the patients, leading to impaired expansion of potent anti-EBV effector cytotoxic CD8+ T cells. IL-27 is produced by EBV-infected B lymphocytes and an IL-27RA-IL-27 autocrine loop is required for the maintenance of EBV-transformed B cells. This potentially explains the eventual favourable outcome of the EBV-induced viral disease in patients with IL-27RA deficiency. Furthermore, we identified neutralizing anti-IL-27 autoantibodies in most individuals who developed sporadic infectious mononucleosis and chronic EBV infection. These results demonstrate the critical role of IL-27RA-IL-27 in immunity to EBV, but also the hijacking of this defence by EBV to promote the expansion of infected transformed B cells.
Collapse
Affiliation(s)
- Emmanuel Martin
- Laboratory of Lymphocyte Activation and Susceptibility to EBV infection, INSERM UMR 1163, Imagine Institute, Paris, France
| | - Sarah Winter
- Laboratory of Lymphocyte Activation and Susceptibility to EBV infection, INSERM UMR 1163, Imagine Institute, Paris, France
- Université Paris Cité, Paris, France
| | - Cécile Garcin
- Laboratory of Lymphocyte Activation and Susceptibility to EBV infection, INSERM UMR 1163, Imagine Institute, Paris, France
- Université Paris Cité, Paris, France
| | - Kay Tanita
- Laboratory of Lymphocyte Activation and Susceptibility to EBV infection, INSERM UMR 1163, Imagine Institute, Paris, France
| | - Akihiro Hoshino
- Laboratory of Lymphocyte Activation and Susceptibility to EBV infection, INSERM UMR 1163, Imagine Institute, Paris, France
| | - Christelle Lenoir
- Laboratory of Lymphocyte Activation and Susceptibility to EBV infection, INSERM UMR 1163, Imagine Institute, Paris, France
| | - Benjamin Fournier
- Laboratory of Lymphocyte Activation and Susceptibility to EBV infection, INSERM UMR 1163, Imagine Institute, Paris, France
- Department of Pediatric Immunology, Hematology and Rheumatology, Necker-Enfants Malades Hospital, Assistance Publique-Hôpitaux de Paris (APHP), Paris, France
| | - Mélanie Migaud
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM UMR 1163, Imagine Institute, Paris, France
| | - David Boutboul
- Université Paris Cité, Paris, France
- Department of Hematology, Cochin Hospital, AP-HP, Paris, France
| | - Mathieu Simonin
- Laboratory of Lymphocyte Activation and Susceptibility to EBV infection, INSERM UMR 1163, Imagine Institute, Paris, France
| | - Alicia Fernandes
- Plateforme Vecteurs Viraux et Transfert de Gènes, Institut Necker Enfants Malades, Necker-Enfants Malades Hospital, APHP, Paris, France
| | - Paul Bastard
- Université Paris Cité, Paris, France
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM UMR 1163, Imagine Institute, Paris, France
| | - Tom Le Voyer
- Université Paris Cité, Paris, France
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM UMR 1163, Imagine Institute, Paris, France
| | - Anne-Laure Roupie
- Laboratory of Lymphocyte Activation and Susceptibility to EBV infection, INSERM UMR 1163, Imagine Institute, Paris, France
- Université Paris Cité, Paris, France
| | - Yassine Ben Ahmed
- Laboratory of Lymphocyte Activation and Susceptibility to EBV infection, INSERM UMR 1163, Imagine Institute, Paris, France
| | - Marianne Leruez-Ville
- Service de Bactériologie, Virologie, Parasitologie et Hygiène, Necker-Enfants Malades Hospital, Paris, France
| | - Marianne Burgard
- Service de Bactériologie, Virologie, Parasitologie et Hygiène, Necker-Enfants Malades Hospital, Paris, France
| | - Geetha Rao
- Garvan Institute of Medical Research, Darlinghurst, Sydney, New South Wales, Australia
| | - Cindy S Ma
- Garvan Institute of Medical Research, Darlinghurst, Sydney, New South Wales, Australia
- St Vincent's Clinical School, Faculty of Medicine and Health, Sydney, New South Wales, Australia
| | - Cécile Masson
- Plateforme de Bioinformatique, INSERM UMR1163, Université de Paris, Imagine Institute, Paris, France
| | - Claire Soudais
- Laboratory of Lymphocyte Activation and Susceptibility to EBV infection, INSERM UMR 1163, Imagine Institute, Paris, France
- Université Paris Cité, Paris, France
| | - Capucine Picard
- Laboratory of Lymphocyte Activation and Susceptibility to EBV infection, INSERM UMR 1163, Imagine Institute, Paris, France
- Université Paris Cité, Paris, France
- Study Center for Primary Immunodeficiencies, Necker-Enfants Malades Hospital, APHP, Paris, France
| | - Jacinta Bustamante
- Université Paris Cité, Paris, France
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM UMR 1163, Imagine Institute, Paris, France
- Study Center for Primary Immunodeficiencies, Necker-Enfants Malades Hospital, APHP, Paris, France
- St Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, The Rockefeller University, New York, NY, USA
| | - Stuart G Tangye
- Garvan Institute of Medical Research, Darlinghurst, Sydney, New South Wales, Australia
- St Vincent's Clinical School, Faculty of Medicine and Health, Sydney, New South Wales, Australia
| | - Nathalie Cheikh
- Hôpital Jean Minjoz, Centre Hospitalo-Universitaire de Besançon, Besançon, France
| | - Mikko Seppänen
- Pediatric Research Center and Rare Disease Center, New Children's Hospital, University of Helsinki and HUS Helsinki University Hospital, Helsinki, Finland
| | - Anne Puel
- Université Paris Cité, Paris, France
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM UMR 1163, Imagine Institute, Paris, France
- St Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, The Rockefeller University, New York, NY, USA
| | - Mark Daly
- Institut for Molecular Medecine Finland, University of Helsinki, Helsinki, Finland
| | - Jean-Laurent Casanova
- Université Paris Cité, Paris, France
- Department of Pediatric Immunology, Hematology and Rheumatology, Necker-Enfants Malades Hospital, Assistance Publique-Hôpitaux de Paris (APHP), Paris, France
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM UMR 1163, Imagine Institute, Paris, France
- St Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, The Rockefeller University, New York, NY, USA
- Howard Hughes Medical Institute, New York, NY, USA
| | - Bénédicte Neven
- Department of Pediatric Immunology, Hematology and Rheumatology, Necker-Enfants Malades Hospital, Assistance Publique-Hôpitaux de Paris (APHP), Paris, France
| | - Alain Fischer
- Department of Pediatric Immunology, Hematology and Rheumatology, Necker-Enfants Malades Hospital, Assistance Publique-Hôpitaux de Paris (APHP), Paris, France
- Collège de France, Paris, France
- Imagine Institute, INSERM UMR 1163, Paris, France
| | - Sylvain Latour
- Laboratory of Lymphocyte Activation and Susceptibility to EBV infection, INSERM UMR 1163, Imagine Institute, Paris, France.
- Université Paris Cité, Paris, France.
| |
Collapse
|
238
|
Jiang Y, Sadun RE. What the SARS-CoV-2 Pandemic Has Taught Us About Immunosuppression, Vaccinations, and Immune Dysregulation: The Rheumatology Experience. Curr Allergy Asthma Rep 2024; 24:221-232. [PMID: 38568321 PMCID: PMC11245734 DOI: 10.1007/s11882-024-01139-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/26/2024] [Indexed: 04/11/2024]
Abstract
PURPOSE OF REVIEW This review reflects on the impact of the COVID-19 pandemic on the field of rheumatology, emphasizing resulting insights related to the risks of viral infections in immunosuppressed patients, vaccine immunogenicity in immunocompromised patients, and immune dysregulation in the setting of viral infection. RECENT FINDINGS During the pandemic, global patient registries provided real-time insights into the risk factors associated with severe COVID-19 outcomes in rheumatology patients. Updated evidence-based recommendations from the American College of Rheumatology (ACR) guided rheumatology practice during a time of considerable uncertainty. Studies on COVID-19 vaccines in immunocompromised populations enhanced our understanding of specific immunosuppressive therapies on vaccine efficacy. The immune dysregulation seen in severe COVID-19 underscored a role for immunomodulation in this and other severe infections. Furthermore, novel post-infectious conditions, namely multisystem inflammatory syndrome in children (MIS-C) and Long COVID, reshaped our understanding of post-viral syndromes and revealed novel pathological mechanisms. Lessons from the COVID-19 pandemic demonstrate the power of collaborative research. The scientific revelations from this dreadful time will, nonetheless, benefit the practice of rheumatology for years to come.
Collapse
Affiliation(s)
- Yike Jiang
- Division of Pediatric Rheumatology, Department of Pediatrics, Duke University School of Medicine, Durham, NC, USA
| | - Rebecca E Sadun
- Division of Pediatric Rheumatology, Department of Pediatrics, Duke University School of Medicine, Durham, NC, USA.
- Division of Rheumatology and Immunology, Department of Medicine, Duke University School of Medicine, Durham, NC, USA.
| |
Collapse
|
239
|
Makhluf H, Madany H, Kim K. Long COVID: Long-Term Impact of SARS-CoV2. Diagnostics (Basel) 2024; 14:711. [PMID: 38611624 PMCID: PMC11011397 DOI: 10.3390/diagnostics14070711] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2024] [Revised: 03/23/2024] [Accepted: 03/25/2024] [Indexed: 04/14/2024] Open
Abstract
Four years post-pandemic, SARS-CoV-2 continues to affect many lives across the globe. An estimated 65 million people suffer from long COVID, a term used to encapsulate the post-acute sequelae of SARS-CoV-2 infections that affect multiple organ systems. Known symptoms include chronic fatigue syndrome, brain fog, cardiovascular issues, autoimmunity, dysautonomia, and clotting due to inflammation. Herein, we review long COVID symptoms, the proposed theories behind the pathology, diagnostics, treatments, and the clinical trials underway to explore treatments for viral persistence, autonomic and cognitive dysfunctions, sleep disturbances, fatigue, and exercise intolerance.
Collapse
Affiliation(s)
- Huda Makhluf
- Department of Mathematics and Natural Sciences, National University, San Diego, CA 92123, USA
- Center for Infectious Disease, La Jolla Institute, La Jolla, CA 92037, USA; (H.M.); (K.K.)
| | - Henry Madany
- Center for Infectious Disease, La Jolla Institute, La Jolla, CA 92037, USA; (H.M.); (K.K.)
- Public Health Sciences, University of California, Irvine, CA 92697, USA
| | - Kenneth Kim
- Center for Infectious Disease, La Jolla Institute, La Jolla, CA 92037, USA; (H.M.); (K.K.)
| |
Collapse
|
240
|
Baker PJ, Bohrer AC, Castro E, Amaral EP, Snow-Smith M, Torres-Juárez F, Gould ST, Queiroz ATL, Fukutani ER, Jordan CM, Khillan JS, Cho K, Barber DL, Andrade BB, Johnson RF, Hilligan KL, Mayer-Barber KD. The inflammatory microenvironment of the lung at the time of infection governs innate control of SARS-CoV-2 replication. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.03.27.586885. [PMID: 38585846 PMCID: PMC10996686 DOI: 10.1101/2024.03.27.586885] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/09/2024]
Abstract
SARS-CoV-2 infection leads to vastly divergent clinical outcomes ranging from asymptomatic infection to fatal disease. Co-morbidities, sex, age, host genetics and vaccine status are known to affect disease severity. Yet, how the inflammatory milieu of the lung at the time of SARS-CoV-2 exposure impacts the control of viral replication remains poorly understood. We demonstrate here that immune events in the mouse lung closely preceding SARS-CoV-2 infection significantly impact viral control and we identify key innate immune pathways required to limit viral replication. A diverse set of pulmonary inflammatory stimuli, including resolved antecedent respiratory infections with S. aureus or influenza, ongoing pulmonary M. tuberculosis infection, ovalbumin/alum-induced asthma or airway administration of defined TLR ligands and recombinant cytokines, all establish an antiviral state in the lung that restricts SARS-CoV-2 replication upon infection. In addition to antiviral type I interferons, the broadly inducible inflammatory cytokines TNFα and IL-1 precondition the lung for enhanced viral control. Collectively, our work shows that SARS-CoV-2 may benefit from an immunologically quiescent lung microenvironment and suggests that heterogeneity in pulmonary inflammation that precedes or accompanies SARS-CoV-2 exposure may be a significant factor contributing to the population-wide variability in COVID-19 disease outcomes.
Collapse
Affiliation(s)
- Paul J. Baker
- Inflammation and Innate Immunity Unit, Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, Maryland 20892, USA
- Current Address: Centre for Innate Immunity and Infectious Diseases, Hudson Institute of Medical Research, Clayton, Victoria 3168, Australia
| | - Andrea C. Bohrer
- Inflammation and Innate Immunity Unit, Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, Maryland 20892, USA
| | - Ehydel Castro
- Inflammation and Innate Immunity Unit, Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, Maryland 20892, USA
| | - Eduardo P. Amaral
- Inflammation and Innate Immunity Unit, Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, Maryland 20892, USA
| | - Maryonne Snow-Smith
- Inflammation and Innate Immunity Unit, Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, Maryland 20892, USA
- Human Eosinophil Section, Laboratory of Parasitic Diseases, NIAID, NIH, Bethesda, Maryland 20892, USA
| | - Flor Torres-Juárez
- Inflammation and Innate Immunity Unit, Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, Maryland 20892, USA
| | - Sydnee T. Gould
- T Lymphocyte Biology Section, Laboratory of Parasitic Diseases, NIAID, NIH, Bethesda, Maryland 20892, USA
- Current Address: Department of Molecular and Cell Biology, University of California, Berkeley, CA 94720, USA
| | - Artur T. L. Queiroz
- Multinational Organization Network Sponsoring Translational and Epidemiological Research Initiative, Salvador, Bahia 41810-710, Brazil
- Laboratory of Clinical and Translational Research, Gonçalo Moniz Institute, Oswaldo Cruz Foundation, Salvador, Bahia 40296-710, Brazil
| | - Eduardo R. Fukutani
- Multinational Organization Network Sponsoring Translational and Epidemiological Research Initiative, Salvador, Bahia 41810-710, Brazil
- Laboratory of Clinical and Translational Research, Gonçalo Moniz Institute, Oswaldo Cruz Foundation, Salvador, Bahia 40296-710, Brazil
| | - Cassandra M. Jordan
- Inflammation and Innate Immunity Unit, Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, Maryland 20892, USA
| | - Jaspal S. Khillan
- Mouse Genetics and Gene Modification Section, Comparative Medicine Branch, NIAID, NIH, Rockville, Maryland 20852, USA
| | - Kyoungin Cho
- Mouse Genetics and Gene Modification Section, Comparative Medicine Branch, NIAID, NIH, Rockville, Maryland 20852, USA
| | - Daniel L. Barber
- T Lymphocyte Biology Section, Laboratory of Parasitic Diseases, NIAID, NIH, Bethesda, Maryland 20892, USA
| | - Bruno B. Andrade
- Multinational Organization Network Sponsoring Translational and Epidemiological Research Initiative, Salvador, Bahia 41810-710, Brazil
- Laboratory of Clinical and Translational Research, Gonçalo Moniz Institute, Oswaldo Cruz Foundation, Salvador, Bahia 40296-710, Brazil
| | - Reed F. Johnson
- SCV2 Virology Core, Laboratory of Viral Diseases, NIAID, NIH, Bethesda, Maryland 20892, USA
| | - Kerry L. Hilligan
- Malaghan Institute of Medical Research, Wellington 6012, New Zealand
| | - Katrin D. Mayer-Barber
- Inflammation and Innate Immunity Unit, Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, Maryland 20892, USA
| |
Collapse
|
241
|
Netea MG, van de Veerdonk FL. Anti-Interleukin-23 Autoantibodies and Severe Infections. N Engl J Med 2024; 390:1143-1146. [PMID: 38507758 DOI: 10.1056/nejme2400475] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 03/22/2024]
Affiliation(s)
- Mihai G Netea
- From the Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, the Netherlands (M.G.N. and F.L.V); and the Department of Immunology and the Metabolism, Life and Medical Sciences Institute, University of Bonn, Bonn, Germany (M.G.N.)
| | - Frank L van de Veerdonk
- From the Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, the Netherlands (M.G.N. and F.L.V); and the Department of Immunology and the Metabolism, Life and Medical Sciences Institute, University of Bonn, Bonn, Germany (M.G.N.)
| |
Collapse
|
242
|
Cheng A, Kashyap A, Salvator H, Rosen LB, Colby D, Ardeshir-Larijani F, Loehrer PJ, Ding L, Lugo Reyes SO, Riminton S, Ballman M, Rocco JM, Marciano BE, Freeman AF, Browne SK, Hsu AP, Zelazny A, Rajan A, Sereti I, Zerbe CS, Lionakis MS, Holland SM. Anti-Interleukin-23 Autoantibodies in Adult-Onset Immunodeficiency. N Engl J Med 2024; 390:1105-1117. [PMID: 38507753 DOI: 10.1056/nejmoa2210665] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 03/22/2024]
Abstract
BACKGROUND Autoantibodies against interleukin-12 (anti-interleukin-12) are often identified in patients with thymoma, but opportunistic infections develop in only some of these patients. Interleukin-12 (with subunits p40 and p35) shares a common subunit with interleukin-23 (subunits p40 and p19). In a patient with disseminated Burkholderia gladioli infection, the identification of both anti-interleukin-23 and anti-interleukin-12 prompted further investigation. METHODS Among the patients (most of whom had thymoma) who were known to have anti-interleukin-12, we screened for autoantibodies against interleukin-23 (anti-interleukin-23). To validate the potential role of anti-interleukin-23 with respect to opportunistic infection, we tested a second cohort of patients with thymoma as well as patients without either thymoma or known anti-interleukin-12 who had unusual infections. RESULTS Among 30 patients with anti-interleukin-12 who had severe mycobacterial, bacterial, or fungal infections, 15 (50%) also had autoantibodies that neutralized interleukin-23. The potency of such neutralization was correlated with the severity of these infections. The neutralizing activity of anti-interleukin-12 alone was not associated with infection. In the validation cohort of 91 patients with thymoma, the presence of anti-interleukin-23 was associated with infection status in 74 patients (81%). Overall, neutralizing anti-interleukin-23 was detected in 30 of 116 patients (26%) with thymoma and in 30 of 36 patients (83%) with disseminated, cerebral, or pulmonary infections. Anti-interleukin-23 was present in 6 of 32 patients (19%) with severe intracellular infections and in 2 of 16 patients (12%) with unusual intracranial infections, including Cladophialophora bantiana and Mycobacterium avium complex. CONCLUSIONS Among patients with a variety of mycobacterial, bacterial, or fungal infections, the presence of neutralizing anti-interleukin-23 was associated with severe, persistent opportunistic infections. (Funded by the National Institute of Allergy and Infectious Diseases and others.).
Collapse
Affiliation(s)
- Aristine Cheng
- From the Division of Intramural Research, National Institute of Allergy and Infectious Diseases (A.C., A.K., H.S., L.B.R., D.C., L.D., J.M.R., B.E.M., A.F.F., S.K.B., A.P.H., A.Z., I.S., C.S.Z., M.S.L., S.M.H.), and the Thoracic and GI Malignancies Branch, Center for Cancer Research, National Cancer Institute (M.B., A.R.), National Institutes of Health, Bethesda, MD; the Division of Infectious Diseases, Department of Medicine, National Taiwan University Hospital and National Taiwan University College of Medicine, Taipei, Taiwan (A.C.); the Department of Respiratory Medicine, Hôpital Foch, Unité Mixte de Recherche 0892, Virology and Molecular Immunology Laboratory, Suresnes Paris-Saclay University, Suresnes, France (H.S.); Indiana University Melvin and Bren Comprehensive Cancer Center, Indiana University School of Medicine, Indianapolis (F.A.-L., P.J.L.); Immune Deficiencies Laboratory, National Institute of Pediatrics, Mexico City (S.O.L.R.); and the Department of Immunology, Repatriation General Hospital Concord, University of Sydney, Concord, NSW, Australia (S.R.)
| | - Anuj Kashyap
- From the Division of Intramural Research, National Institute of Allergy and Infectious Diseases (A.C., A.K., H.S., L.B.R., D.C., L.D., J.M.R., B.E.M., A.F.F., S.K.B., A.P.H., A.Z., I.S., C.S.Z., M.S.L., S.M.H.), and the Thoracic and GI Malignancies Branch, Center for Cancer Research, National Cancer Institute (M.B., A.R.), National Institutes of Health, Bethesda, MD; the Division of Infectious Diseases, Department of Medicine, National Taiwan University Hospital and National Taiwan University College of Medicine, Taipei, Taiwan (A.C.); the Department of Respiratory Medicine, Hôpital Foch, Unité Mixte de Recherche 0892, Virology and Molecular Immunology Laboratory, Suresnes Paris-Saclay University, Suresnes, France (H.S.); Indiana University Melvin and Bren Comprehensive Cancer Center, Indiana University School of Medicine, Indianapolis (F.A.-L., P.J.L.); Immune Deficiencies Laboratory, National Institute of Pediatrics, Mexico City (S.O.L.R.); and the Department of Immunology, Repatriation General Hospital Concord, University of Sydney, Concord, NSW, Australia (S.R.)
| | - Helene Salvator
- From the Division of Intramural Research, National Institute of Allergy and Infectious Diseases (A.C., A.K., H.S., L.B.R., D.C., L.D., J.M.R., B.E.M., A.F.F., S.K.B., A.P.H., A.Z., I.S., C.S.Z., M.S.L., S.M.H.), and the Thoracic and GI Malignancies Branch, Center for Cancer Research, National Cancer Institute (M.B., A.R.), National Institutes of Health, Bethesda, MD; the Division of Infectious Diseases, Department of Medicine, National Taiwan University Hospital and National Taiwan University College of Medicine, Taipei, Taiwan (A.C.); the Department of Respiratory Medicine, Hôpital Foch, Unité Mixte de Recherche 0892, Virology and Molecular Immunology Laboratory, Suresnes Paris-Saclay University, Suresnes, France (H.S.); Indiana University Melvin and Bren Comprehensive Cancer Center, Indiana University School of Medicine, Indianapolis (F.A.-L., P.J.L.); Immune Deficiencies Laboratory, National Institute of Pediatrics, Mexico City (S.O.L.R.); and the Department of Immunology, Repatriation General Hospital Concord, University of Sydney, Concord, NSW, Australia (S.R.)
| | - Lindsey B Rosen
- From the Division of Intramural Research, National Institute of Allergy and Infectious Diseases (A.C., A.K., H.S., L.B.R., D.C., L.D., J.M.R., B.E.M., A.F.F., S.K.B., A.P.H., A.Z., I.S., C.S.Z., M.S.L., S.M.H.), and the Thoracic and GI Malignancies Branch, Center for Cancer Research, National Cancer Institute (M.B., A.R.), National Institutes of Health, Bethesda, MD; the Division of Infectious Diseases, Department of Medicine, National Taiwan University Hospital and National Taiwan University College of Medicine, Taipei, Taiwan (A.C.); the Department of Respiratory Medicine, Hôpital Foch, Unité Mixte de Recherche 0892, Virology and Molecular Immunology Laboratory, Suresnes Paris-Saclay University, Suresnes, France (H.S.); Indiana University Melvin and Bren Comprehensive Cancer Center, Indiana University School of Medicine, Indianapolis (F.A.-L., P.J.L.); Immune Deficiencies Laboratory, National Institute of Pediatrics, Mexico City (S.O.L.R.); and the Department of Immunology, Repatriation General Hospital Concord, University of Sydney, Concord, NSW, Australia (S.R.)
| | - Devon Colby
- From the Division of Intramural Research, National Institute of Allergy and Infectious Diseases (A.C., A.K., H.S., L.B.R., D.C., L.D., J.M.R., B.E.M., A.F.F., S.K.B., A.P.H., A.Z., I.S., C.S.Z., M.S.L., S.M.H.), and the Thoracic and GI Malignancies Branch, Center for Cancer Research, National Cancer Institute (M.B., A.R.), National Institutes of Health, Bethesda, MD; the Division of Infectious Diseases, Department of Medicine, National Taiwan University Hospital and National Taiwan University College of Medicine, Taipei, Taiwan (A.C.); the Department of Respiratory Medicine, Hôpital Foch, Unité Mixte de Recherche 0892, Virology and Molecular Immunology Laboratory, Suresnes Paris-Saclay University, Suresnes, France (H.S.); Indiana University Melvin and Bren Comprehensive Cancer Center, Indiana University School of Medicine, Indianapolis (F.A.-L., P.J.L.); Immune Deficiencies Laboratory, National Institute of Pediatrics, Mexico City (S.O.L.R.); and the Department of Immunology, Repatriation General Hospital Concord, University of Sydney, Concord, NSW, Australia (S.R.)
| | - Fatemeh Ardeshir-Larijani
- From the Division of Intramural Research, National Institute of Allergy and Infectious Diseases (A.C., A.K., H.S., L.B.R., D.C., L.D., J.M.R., B.E.M., A.F.F., S.K.B., A.P.H., A.Z., I.S., C.S.Z., M.S.L., S.M.H.), and the Thoracic and GI Malignancies Branch, Center for Cancer Research, National Cancer Institute (M.B., A.R.), National Institutes of Health, Bethesda, MD; the Division of Infectious Diseases, Department of Medicine, National Taiwan University Hospital and National Taiwan University College of Medicine, Taipei, Taiwan (A.C.); the Department of Respiratory Medicine, Hôpital Foch, Unité Mixte de Recherche 0892, Virology and Molecular Immunology Laboratory, Suresnes Paris-Saclay University, Suresnes, France (H.S.); Indiana University Melvin and Bren Comprehensive Cancer Center, Indiana University School of Medicine, Indianapolis (F.A.-L., P.J.L.); Immune Deficiencies Laboratory, National Institute of Pediatrics, Mexico City (S.O.L.R.); and the Department of Immunology, Repatriation General Hospital Concord, University of Sydney, Concord, NSW, Australia (S.R.)
| | - Patrick J Loehrer
- From the Division of Intramural Research, National Institute of Allergy and Infectious Diseases (A.C., A.K., H.S., L.B.R., D.C., L.D., J.M.R., B.E.M., A.F.F., S.K.B., A.P.H., A.Z., I.S., C.S.Z., M.S.L., S.M.H.), and the Thoracic and GI Malignancies Branch, Center for Cancer Research, National Cancer Institute (M.B., A.R.), National Institutes of Health, Bethesda, MD; the Division of Infectious Diseases, Department of Medicine, National Taiwan University Hospital and National Taiwan University College of Medicine, Taipei, Taiwan (A.C.); the Department of Respiratory Medicine, Hôpital Foch, Unité Mixte de Recherche 0892, Virology and Molecular Immunology Laboratory, Suresnes Paris-Saclay University, Suresnes, France (H.S.); Indiana University Melvin and Bren Comprehensive Cancer Center, Indiana University School of Medicine, Indianapolis (F.A.-L., P.J.L.); Immune Deficiencies Laboratory, National Institute of Pediatrics, Mexico City (S.O.L.R.); and the Department of Immunology, Repatriation General Hospital Concord, University of Sydney, Concord, NSW, Australia (S.R.)
| | - Li Ding
- From the Division of Intramural Research, National Institute of Allergy and Infectious Diseases (A.C., A.K., H.S., L.B.R., D.C., L.D., J.M.R., B.E.M., A.F.F., S.K.B., A.P.H., A.Z., I.S., C.S.Z., M.S.L., S.M.H.), and the Thoracic and GI Malignancies Branch, Center for Cancer Research, National Cancer Institute (M.B., A.R.), National Institutes of Health, Bethesda, MD; the Division of Infectious Diseases, Department of Medicine, National Taiwan University Hospital and National Taiwan University College of Medicine, Taipei, Taiwan (A.C.); the Department of Respiratory Medicine, Hôpital Foch, Unité Mixte de Recherche 0892, Virology and Molecular Immunology Laboratory, Suresnes Paris-Saclay University, Suresnes, France (H.S.); Indiana University Melvin and Bren Comprehensive Cancer Center, Indiana University School of Medicine, Indianapolis (F.A.-L., P.J.L.); Immune Deficiencies Laboratory, National Institute of Pediatrics, Mexico City (S.O.L.R.); and the Department of Immunology, Repatriation General Hospital Concord, University of Sydney, Concord, NSW, Australia (S.R.)
| | - Saul O Lugo Reyes
- From the Division of Intramural Research, National Institute of Allergy and Infectious Diseases (A.C., A.K., H.S., L.B.R., D.C., L.D., J.M.R., B.E.M., A.F.F., S.K.B., A.P.H., A.Z., I.S., C.S.Z., M.S.L., S.M.H.), and the Thoracic and GI Malignancies Branch, Center for Cancer Research, National Cancer Institute (M.B., A.R.), National Institutes of Health, Bethesda, MD; the Division of Infectious Diseases, Department of Medicine, National Taiwan University Hospital and National Taiwan University College of Medicine, Taipei, Taiwan (A.C.); the Department of Respiratory Medicine, Hôpital Foch, Unité Mixte de Recherche 0892, Virology and Molecular Immunology Laboratory, Suresnes Paris-Saclay University, Suresnes, France (H.S.); Indiana University Melvin and Bren Comprehensive Cancer Center, Indiana University School of Medicine, Indianapolis (F.A.-L., P.J.L.); Immune Deficiencies Laboratory, National Institute of Pediatrics, Mexico City (S.O.L.R.); and the Department of Immunology, Repatriation General Hospital Concord, University of Sydney, Concord, NSW, Australia (S.R.)
| | - Sean Riminton
- From the Division of Intramural Research, National Institute of Allergy and Infectious Diseases (A.C., A.K., H.S., L.B.R., D.C., L.D., J.M.R., B.E.M., A.F.F., S.K.B., A.P.H., A.Z., I.S., C.S.Z., M.S.L., S.M.H.), and the Thoracic and GI Malignancies Branch, Center for Cancer Research, National Cancer Institute (M.B., A.R.), National Institutes of Health, Bethesda, MD; the Division of Infectious Diseases, Department of Medicine, National Taiwan University Hospital and National Taiwan University College of Medicine, Taipei, Taiwan (A.C.); the Department of Respiratory Medicine, Hôpital Foch, Unité Mixte de Recherche 0892, Virology and Molecular Immunology Laboratory, Suresnes Paris-Saclay University, Suresnes, France (H.S.); Indiana University Melvin and Bren Comprehensive Cancer Center, Indiana University School of Medicine, Indianapolis (F.A.-L., P.J.L.); Immune Deficiencies Laboratory, National Institute of Pediatrics, Mexico City (S.O.L.R.); and the Department of Immunology, Repatriation General Hospital Concord, University of Sydney, Concord, NSW, Australia (S.R.)
| | - Madison Ballman
- From the Division of Intramural Research, National Institute of Allergy and Infectious Diseases (A.C., A.K., H.S., L.B.R., D.C., L.D., J.M.R., B.E.M., A.F.F., S.K.B., A.P.H., A.Z., I.S., C.S.Z., M.S.L., S.M.H.), and the Thoracic and GI Malignancies Branch, Center for Cancer Research, National Cancer Institute (M.B., A.R.), National Institutes of Health, Bethesda, MD; the Division of Infectious Diseases, Department of Medicine, National Taiwan University Hospital and National Taiwan University College of Medicine, Taipei, Taiwan (A.C.); the Department of Respiratory Medicine, Hôpital Foch, Unité Mixte de Recherche 0892, Virology and Molecular Immunology Laboratory, Suresnes Paris-Saclay University, Suresnes, France (H.S.); Indiana University Melvin and Bren Comprehensive Cancer Center, Indiana University School of Medicine, Indianapolis (F.A.-L., P.J.L.); Immune Deficiencies Laboratory, National Institute of Pediatrics, Mexico City (S.O.L.R.); and the Department of Immunology, Repatriation General Hospital Concord, University of Sydney, Concord, NSW, Australia (S.R.)
| | - Joseph M Rocco
- From the Division of Intramural Research, National Institute of Allergy and Infectious Diseases (A.C., A.K., H.S., L.B.R., D.C., L.D., J.M.R., B.E.M., A.F.F., S.K.B., A.P.H., A.Z., I.S., C.S.Z., M.S.L., S.M.H.), and the Thoracic and GI Malignancies Branch, Center for Cancer Research, National Cancer Institute (M.B., A.R.), National Institutes of Health, Bethesda, MD; the Division of Infectious Diseases, Department of Medicine, National Taiwan University Hospital and National Taiwan University College of Medicine, Taipei, Taiwan (A.C.); the Department of Respiratory Medicine, Hôpital Foch, Unité Mixte de Recherche 0892, Virology and Molecular Immunology Laboratory, Suresnes Paris-Saclay University, Suresnes, France (H.S.); Indiana University Melvin and Bren Comprehensive Cancer Center, Indiana University School of Medicine, Indianapolis (F.A.-L., P.J.L.); Immune Deficiencies Laboratory, National Institute of Pediatrics, Mexico City (S.O.L.R.); and the Department of Immunology, Repatriation General Hospital Concord, University of Sydney, Concord, NSW, Australia (S.R.)
| | - Beatriz E Marciano
- From the Division of Intramural Research, National Institute of Allergy and Infectious Diseases (A.C., A.K., H.S., L.B.R., D.C., L.D., J.M.R., B.E.M., A.F.F., S.K.B., A.P.H., A.Z., I.S., C.S.Z., M.S.L., S.M.H.), and the Thoracic and GI Malignancies Branch, Center for Cancer Research, National Cancer Institute (M.B., A.R.), National Institutes of Health, Bethesda, MD; the Division of Infectious Diseases, Department of Medicine, National Taiwan University Hospital and National Taiwan University College of Medicine, Taipei, Taiwan (A.C.); the Department of Respiratory Medicine, Hôpital Foch, Unité Mixte de Recherche 0892, Virology and Molecular Immunology Laboratory, Suresnes Paris-Saclay University, Suresnes, France (H.S.); Indiana University Melvin and Bren Comprehensive Cancer Center, Indiana University School of Medicine, Indianapolis (F.A.-L., P.J.L.); Immune Deficiencies Laboratory, National Institute of Pediatrics, Mexico City (S.O.L.R.); and the Department of Immunology, Repatriation General Hospital Concord, University of Sydney, Concord, NSW, Australia (S.R.)
| | - Alexandra F Freeman
- From the Division of Intramural Research, National Institute of Allergy and Infectious Diseases (A.C., A.K., H.S., L.B.R., D.C., L.D., J.M.R., B.E.M., A.F.F., S.K.B., A.P.H., A.Z., I.S., C.S.Z., M.S.L., S.M.H.), and the Thoracic and GI Malignancies Branch, Center for Cancer Research, National Cancer Institute (M.B., A.R.), National Institutes of Health, Bethesda, MD; the Division of Infectious Diseases, Department of Medicine, National Taiwan University Hospital and National Taiwan University College of Medicine, Taipei, Taiwan (A.C.); the Department of Respiratory Medicine, Hôpital Foch, Unité Mixte de Recherche 0892, Virology and Molecular Immunology Laboratory, Suresnes Paris-Saclay University, Suresnes, France (H.S.); Indiana University Melvin and Bren Comprehensive Cancer Center, Indiana University School of Medicine, Indianapolis (F.A.-L., P.J.L.); Immune Deficiencies Laboratory, National Institute of Pediatrics, Mexico City (S.O.L.R.); and the Department of Immunology, Repatriation General Hospital Concord, University of Sydney, Concord, NSW, Australia (S.R.)
| | - Sarah K Browne
- From the Division of Intramural Research, National Institute of Allergy and Infectious Diseases (A.C., A.K., H.S., L.B.R., D.C., L.D., J.M.R., B.E.M., A.F.F., S.K.B., A.P.H., A.Z., I.S., C.S.Z., M.S.L., S.M.H.), and the Thoracic and GI Malignancies Branch, Center for Cancer Research, National Cancer Institute (M.B., A.R.), National Institutes of Health, Bethesda, MD; the Division of Infectious Diseases, Department of Medicine, National Taiwan University Hospital and National Taiwan University College of Medicine, Taipei, Taiwan (A.C.); the Department of Respiratory Medicine, Hôpital Foch, Unité Mixte de Recherche 0892, Virology and Molecular Immunology Laboratory, Suresnes Paris-Saclay University, Suresnes, France (H.S.); Indiana University Melvin and Bren Comprehensive Cancer Center, Indiana University School of Medicine, Indianapolis (F.A.-L., P.J.L.); Immune Deficiencies Laboratory, National Institute of Pediatrics, Mexico City (S.O.L.R.); and the Department of Immunology, Repatriation General Hospital Concord, University of Sydney, Concord, NSW, Australia (S.R.)
| | - Amy P Hsu
- From the Division of Intramural Research, National Institute of Allergy and Infectious Diseases (A.C., A.K., H.S., L.B.R., D.C., L.D., J.M.R., B.E.M., A.F.F., S.K.B., A.P.H., A.Z., I.S., C.S.Z., M.S.L., S.M.H.), and the Thoracic and GI Malignancies Branch, Center for Cancer Research, National Cancer Institute (M.B., A.R.), National Institutes of Health, Bethesda, MD; the Division of Infectious Diseases, Department of Medicine, National Taiwan University Hospital and National Taiwan University College of Medicine, Taipei, Taiwan (A.C.); the Department of Respiratory Medicine, Hôpital Foch, Unité Mixte de Recherche 0892, Virology and Molecular Immunology Laboratory, Suresnes Paris-Saclay University, Suresnes, France (H.S.); Indiana University Melvin and Bren Comprehensive Cancer Center, Indiana University School of Medicine, Indianapolis (F.A.-L., P.J.L.); Immune Deficiencies Laboratory, National Institute of Pediatrics, Mexico City (S.O.L.R.); and the Department of Immunology, Repatriation General Hospital Concord, University of Sydney, Concord, NSW, Australia (S.R.)
| | - Adrian Zelazny
- From the Division of Intramural Research, National Institute of Allergy and Infectious Diseases (A.C., A.K., H.S., L.B.R., D.C., L.D., J.M.R., B.E.M., A.F.F., S.K.B., A.P.H., A.Z., I.S., C.S.Z., M.S.L., S.M.H.), and the Thoracic and GI Malignancies Branch, Center for Cancer Research, National Cancer Institute (M.B., A.R.), National Institutes of Health, Bethesda, MD; the Division of Infectious Diseases, Department of Medicine, National Taiwan University Hospital and National Taiwan University College of Medicine, Taipei, Taiwan (A.C.); the Department of Respiratory Medicine, Hôpital Foch, Unité Mixte de Recherche 0892, Virology and Molecular Immunology Laboratory, Suresnes Paris-Saclay University, Suresnes, France (H.S.); Indiana University Melvin and Bren Comprehensive Cancer Center, Indiana University School of Medicine, Indianapolis (F.A.-L., P.J.L.); Immune Deficiencies Laboratory, National Institute of Pediatrics, Mexico City (S.O.L.R.); and the Department of Immunology, Repatriation General Hospital Concord, University of Sydney, Concord, NSW, Australia (S.R.)
| | - Arun Rajan
- From the Division of Intramural Research, National Institute of Allergy and Infectious Diseases (A.C., A.K., H.S., L.B.R., D.C., L.D., J.M.R., B.E.M., A.F.F., S.K.B., A.P.H., A.Z., I.S., C.S.Z., M.S.L., S.M.H.), and the Thoracic and GI Malignancies Branch, Center for Cancer Research, National Cancer Institute (M.B., A.R.), National Institutes of Health, Bethesda, MD; the Division of Infectious Diseases, Department of Medicine, National Taiwan University Hospital and National Taiwan University College of Medicine, Taipei, Taiwan (A.C.); the Department of Respiratory Medicine, Hôpital Foch, Unité Mixte de Recherche 0892, Virology and Molecular Immunology Laboratory, Suresnes Paris-Saclay University, Suresnes, France (H.S.); Indiana University Melvin and Bren Comprehensive Cancer Center, Indiana University School of Medicine, Indianapolis (F.A.-L., P.J.L.); Immune Deficiencies Laboratory, National Institute of Pediatrics, Mexico City (S.O.L.R.); and the Department of Immunology, Repatriation General Hospital Concord, University of Sydney, Concord, NSW, Australia (S.R.)
| | - Irini Sereti
- From the Division of Intramural Research, National Institute of Allergy and Infectious Diseases (A.C., A.K., H.S., L.B.R., D.C., L.D., J.M.R., B.E.M., A.F.F., S.K.B., A.P.H., A.Z., I.S., C.S.Z., M.S.L., S.M.H.), and the Thoracic and GI Malignancies Branch, Center for Cancer Research, National Cancer Institute (M.B., A.R.), National Institutes of Health, Bethesda, MD; the Division of Infectious Diseases, Department of Medicine, National Taiwan University Hospital and National Taiwan University College of Medicine, Taipei, Taiwan (A.C.); the Department of Respiratory Medicine, Hôpital Foch, Unité Mixte de Recherche 0892, Virology and Molecular Immunology Laboratory, Suresnes Paris-Saclay University, Suresnes, France (H.S.); Indiana University Melvin and Bren Comprehensive Cancer Center, Indiana University School of Medicine, Indianapolis (F.A.-L., P.J.L.); Immune Deficiencies Laboratory, National Institute of Pediatrics, Mexico City (S.O.L.R.); and the Department of Immunology, Repatriation General Hospital Concord, University of Sydney, Concord, NSW, Australia (S.R.)
| | - Christa S Zerbe
- From the Division of Intramural Research, National Institute of Allergy and Infectious Diseases (A.C., A.K., H.S., L.B.R., D.C., L.D., J.M.R., B.E.M., A.F.F., S.K.B., A.P.H., A.Z., I.S., C.S.Z., M.S.L., S.M.H.), and the Thoracic and GI Malignancies Branch, Center for Cancer Research, National Cancer Institute (M.B., A.R.), National Institutes of Health, Bethesda, MD; the Division of Infectious Diseases, Department of Medicine, National Taiwan University Hospital and National Taiwan University College of Medicine, Taipei, Taiwan (A.C.); the Department of Respiratory Medicine, Hôpital Foch, Unité Mixte de Recherche 0892, Virology and Molecular Immunology Laboratory, Suresnes Paris-Saclay University, Suresnes, France (H.S.); Indiana University Melvin and Bren Comprehensive Cancer Center, Indiana University School of Medicine, Indianapolis (F.A.-L., P.J.L.); Immune Deficiencies Laboratory, National Institute of Pediatrics, Mexico City (S.O.L.R.); and the Department of Immunology, Repatriation General Hospital Concord, University of Sydney, Concord, NSW, Australia (S.R.)
| | - Michail S Lionakis
- From the Division of Intramural Research, National Institute of Allergy and Infectious Diseases (A.C., A.K., H.S., L.B.R., D.C., L.D., J.M.R., B.E.M., A.F.F., S.K.B., A.P.H., A.Z., I.S., C.S.Z., M.S.L., S.M.H.), and the Thoracic and GI Malignancies Branch, Center for Cancer Research, National Cancer Institute (M.B., A.R.), National Institutes of Health, Bethesda, MD; the Division of Infectious Diseases, Department of Medicine, National Taiwan University Hospital and National Taiwan University College of Medicine, Taipei, Taiwan (A.C.); the Department of Respiratory Medicine, Hôpital Foch, Unité Mixte de Recherche 0892, Virology and Molecular Immunology Laboratory, Suresnes Paris-Saclay University, Suresnes, France (H.S.); Indiana University Melvin and Bren Comprehensive Cancer Center, Indiana University School of Medicine, Indianapolis (F.A.-L., P.J.L.); Immune Deficiencies Laboratory, National Institute of Pediatrics, Mexico City (S.O.L.R.); and the Department of Immunology, Repatriation General Hospital Concord, University of Sydney, Concord, NSW, Australia (S.R.)
| | - Steven M Holland
- From the Division of Intramural Research, National Institute of Allergy and Infectious Diseases (A.C., A.K., H.S., L.B.R., D.C., L.D., J.M.R., B.E.M., A.F.F., S.K.B., A.P.H., A.Z., I.S., C.S.Z., M.S.L., S.M.H.), and the Thoracic and GI Malignancies Branch, Center for Cancer Research, National Cancer Institute (M.B., A.R.), National Institutes of Health, Bethesda, MD; the Division of Infectious Diseases, Department of Medicine, National Taiwan University Hospital and National Taiwan University College of Medicine, Taipei, Taiwan (A.C.); the Department of Respiratory Medicine, Hôpital Foch, Unité Mixte de Recherche 0892, Virology and Molecular Immunology Laboratory, Suresnes Paris-Saclay University, Suresnes, France (H.S.); Indiana University Melvin and Bren Comprehensive Cancer Center, Indiana University School of Medicine, Indianapolis (F.A.-L., P.J.L.); Immune Deficiencies Laboratory, National Institute of Pediatrics, Mexico City (S.O.L.R.); and the Department of Immunology, Repatriation General Hospital Concord, University of Sydney, Concord, NSW, Australia (S.R.)
| |
Collapse
|
243
|
Jonassen TB, Jørgensen SE, Mitchell NH, Mogensen TH, Berg RMG, Ronit A, Plovsing RR. Alveolar cytokines and interferon autoantibodies in COVID-19 ARDS. Front Immunol 2024; 15:1353012. [PMID: 38571960 PMCID: PMC10987806 DOI: 10.3389/fimmu.2024.1353012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2023] [Accepted: 02/26/2024] [Indexed: 04/05/2024] Open
Abstract
Background Type I interferon (IFN-I) and IFN autoantibodies play a crucial role in controlling SARS-CoV-2 infection. The levels of these mediators have only rarely been studied in the alveolar compartment in patients with COVID-19 acute respiratory distress syndrome (CARDS) but have not been compared across different ARDS etiologies, and the potential effect of dexamethasone (DXM) on these mediators is not known. Methods We assessed the integrity of the alveolo-capillary membrane, interleukins, type I, II, and III IFNs, and IFN autoantibodies by studying the epithelial lining fluid (ELF) volumes, alveolar concentration of protein, and ELF-corrected concentrations of cytokines in two patient subgroups and controls. Results A total of 16 patients with CARDS (four without and 12 with DXM treatment), eight with non-CARDS, and 15 healthy controls were included. The highest ELF volumes and protein levels were observed in CARDS. Systemic and ELF-corrected alveolar concentrations of interleukin (IL)-6 appeared to be particularly low in patients with CARDS receiving DXM, whereas alveolar levels of IL-8 were high regardless of DXM treatment. Alveolar levels of IFNs were similar between CARDS and non-CARDS patients, and IFNα and IFNω autoantibody levels were higher in patients with CARDS and non-CARDS than in healthy controls. Conclusions Patients with CARDS exhibited greater alveolo-capillary barrier disruption with compartmentalization of IL-8, regardless of DXM treatment, whereas systemic and alveolar levels of IL-6 were lower in the DXM-treated subgroup. IFN-I autoantibodies were higher in the BALF of CARDS patients, independent of DXM, whereas IFN autoantibodies in plasma were similar to those in controls.
Collapse
Affiliation(s)
- Trine B. Jonassen
- Department of Anesthesiology and Intensive Care, Copenhagen University Hospital-Amager and Hvidovre Hospitals, Hvidovre, Denmark
| | - Sofie E. Jørgensen
- Department of Infectious Diseases, Aarhus University Hospital (AUH), Aarhus, Denmark
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
| | - Nikki H. Mitchell
- Department of Clinical Biochemistry, Copenhagen University Hospital-Amager and Hvidovre, Hvidovre, Denmark
| | - Trine H. Mogensen
- Department of Infectious Diseases, Aarhus University Hospital (AUH), Aarhus, Denmark
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
| | - Ronan M. G. Berg
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Department of Clinical Physiology and Nuclear Medicine, Copenhagen University Hospital-Rigshospitalet, Copenhagen, Denmark
- Centre for Physical Activity Research, Copenhagen University Hospital-Rigshospitalet, Copenhagen, Denmark
- Neurovascular Research Laboratory, Faculty of Life Sciences and Education, University of South Wales, Pontypridd, United Kingdom
| | - Andreas Ronit
- Department of Infectious Diseases, Copenhagen University Hospital-Amager and Hvidovre Hospitals, Hvidovre, Denmark
| | - Ronni R. Plovsing
- Department of Anesthesiology and Intensive Care, Copenhagen University Hospital-Amager and Hvidovre Hospitals, Hvidovre, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
244
|
Kazer SW, Match CM, Langan EM, Messou MA, LaSalle TJ, O’Leary E, Marbourg J, Naughton K, von Andrian UH, Ordovas-Montanes J. Primary nasal viral infection rewires the tissue-scale memory response. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.05.11.539887. [PMID: 38562902 PMCID: PMC10983857 DOI: 10.1101/2023.05.11.539887] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/04/2024]
Abstract
The nasal mucosa is frequently the initial site of respiratory viral infection, replication, and transmission. Recent work has started to clarify the independent responses of epithelial, myeloid, and lymphoid cells to viral infection in the nasal mucosa, but their spatiotemporal coordination and relative contributions remain unclear. Furthermore, understanding whether and how primary infection shapes tissue-scale memory responses to secondary challenge is critical for the rational design of nasal-targeting therapeutics and vaccines. Here, we generated a single-cell RNA-sequencing (scRNA-seq) atlas of the murine nasal mucosa sampling three distinct regions before and during primary and secondary influenza infection. Primary infection was largely restricted to respiratory mucosa and induced stepwise changes in cell type, subset, and state composition over time. Type I Interferon (IFN)-responsive neutrophils appeared 2 days post infection (dpi) and preceded transient IFN-responsive/cycling epithelial cell responses 5 dpi, which coincided with broader antiviral monocyte and NK cell accumulation. By 8 dpi, monocyte-derived macrophages (MDMs) expressing Cxcl9 and Cxcl16 arose alongside effector cytotoxic CD8 and Ifng-expressing CD4 T cells. Following viral clearance (14 dpi), rare, previously undescribed Krt13+ nasal immune-interacting floor epithelial (KNIIFE) cells expressing multiple genes with immune communication potential increased concurrently with tissue-resident memory T (TRM)-like cells and early IgG+/IgA+ plasmablasts. Proportionality analysis coupled with cell-cell communication inference, alongside validation by in situ microscopy, underscored the CXCL16-CXCR6 signaling axis between MDMs and effector CD8 T cells 8dpi and KNIIFE cells and TRM cells 14 dpi. Secondary influenza challenge with a homologous or heterologous strain administered 60 dpi induced an accelerated and coordinated myeloid and lymphoid response without epithelial proliferation, illustrating how tissue-scale memory to natural infection engages both myeloid and lymphoid cells to reduce epithelial regenerative burden. Together, this atlas serves as a reference for viral infection in the upper respiratory tract and highlights the efficacy of local coordinated memory responses upon rechallenge.
Collapse
Affiliation(s)
- Samuel W. Kazer
- Division of Gastroenterology, Hepatology, and Nutrition, Boston Children’s Hospital, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Immunology, Harvard Medical School, Boston, MA, USA
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA, USA
| | - Colette Matysiak Match
- Department of Immunology, Harvard Medical School, Boston, MA, USA
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA, USA
| | - Erica M. Langan
- Division of Gastroenterology, Hepatology, and Nutrition, Boston Children’s Hospital, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Marie-Angèle Messou
- Department of Immunology, Harvard Medical School, Boston, MA, USA
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA, USA
| | - Thomas J. LaSalle
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Program in Health Sciences and Technology, Harvard Medical School & Massachusetts Institute of Technology, Boston, MA, USA
| | - Elise O’Leary
- Department of Immunology, Harvard Medical School, Boston, MA, USA
| | | | | | - Ulrich H. von Andrian
- Department of Immunology, Harvard Medical School, Boston, MA, USA
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA, USA
| | - Jose Ordovas-Montanes
- Division of Gastroenterology, Hepatology, and Nutrition, Boston Children’s Hospital, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA, USA
- Program in Immunology, Harvard Medical School, Boston, MA 02115, USA
- Harvard Stem Cell Institute, Harvard University, Cambridge, MA, USA
| |
Collapse
|
245
|
Wu M, Fan Y, Li L, Yuan J. Bi-directional regulation of type I interferon signaling by heme oxygenase-1. iScience 2024; 27:109185. [PMID: 38420586 PMCID: PMC10901085 DOI: 10.1016/j.isci.2024.109185] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2023] [Revised: 12/23/2023] [Accepted: 02/06/2024] [Indexed: 03/02/2024] Open
Abstract
Moderate activation of IFN-I contributes to the body's immune response, but its abnormal expression, stimulated by oxidative stress or other factors causes pathological damage. Heme oxygenase-1 (HO-1), induced by stress stimuli in the body, exerts a central role in cellular protection. Here we showed that HO-1 could promote IFN-1 under Spring Viremia of Carp virus (SVCV) infection and concomitantly attenuate the replication of SVCV. Further characterization of truncated mutants of HO-1 confirmed that intact HO-1 was essential for its antiviral function via IFN-I. Importantly, HO-1 inhibited the IFN-I signal by degrading the IRF3/7 through the autophagy pathway when it was triggered by H2O2 treatment. The iron ion-binding site (His28) was critical for HO-1 to degrade IRF3/7. HO-1 degradation of IRF3/7 is conserved in fish and mammals. Collectively, HO-1 regulates IFN-I positively under viral infection and negatively under oxidative stress, elucidating a mechanism by which HO-1 regulates IFN-I signaling in bi-directions.
Collapse
Affiliation(s)
- Miaomiao Wu
- Department of Aquatic Animal Medicine, College of Fisheries, Huazhong Agricultural University, Wuhan 430070, People’s Republic of China
- Hubei Engineering Research Center for Aquatic Animal Diseases Control and Prevention, Wuhan 430070, People’s Republic of China
| | - Yihui Fan
- Hubei Engineering Research Center for Aquatic Animal Diseases Control and Prevention, Wuhan 430070, People’s Republic of China
- National Aquatic Animal Diseases Para-reference laboratory (HZAU), Wuhan 430070, People’s Republic of China
| | - Lijuan Li
- Department of Aquatic Animal Medicine, College of Fisheries, Huazhong Agricultural University, Wuhan 430070, People’s Republic of China
- Hubei Engineering Research Center for Aquatic Animal Diseases Control and Prevention, Wuhan 430070, People’s Republic of China
- National Aquatic Animal Diseases Para-reference laboratory (HZAU), Wuhan 430070, People’s Republic of China
| | - Junfa Yuan
- Department of Aquatic Animal Medicine, College of Fisheries, Huazhong Agricultural University, Wuhan 430070, People’s Republic of China
- Hubei Engineering Research Center for Aquatic Animal Diseases Control and Prevention, Wuhan 430070, People’s Republic of China
- National Aquatic Animal Diseases Para-reference laboratory (HZAU), Wuhan 430070, People’s Republic of China
| |
Collapse
|
246
|
Geanes ES, McLennan R, LeMaster C, Bradley T. Autoantibodies to ACE2 and immune molecules are associated with COVID-19 disease severity. COMMUNICATIONS MEDICINE 2024; 4:47. [PMID: 38491326 PMCID: PMC10943194 DOI: 10.1038/s43856-024-00477-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2023] [Accepted: 03/05/2024] [Indexed: 03/18/2024] Open
Abstract
BACKGROUND Increased inflammation caused by SARS-CoV-2 infection can lead to severe coronavirus disease 2019 (COVID-19) and long-term disease manifestations. The mechanisms of this variable long-term immune activation are poorly defined. One feature of this increased inflammation is elevated levels of proinflammatory cytokines and chemokines. Autoantibodies targeting immune factors such as cytokines, as well as the viral host cell receptor, angiotensin-converting enzyme 2 (ACE2), have been observed after SARS-CoV-2 infection. Autoantibodies to immune factors and ACE2 could interfere with normal immune regulation and lead to increased inflammation, severe COVID-19, and long-term complications. METHODS Here, we deeply profiled the features of ACE2, cytokine, and chemokine autoantibodies in samples from patients recovering from severe COVID-19. We measured the levels of immunoglobulin subclasses (IgG, IgA, IgM) in the peripheral blood against ACE2 and 23 cytokines and other immune molecules. We then utilized an ACE2 peptide microarray to map the linear epitopes targeted by ACE2 autoantibodies. RESULTS We demonstrate that ACE2 autoantibody levels are increased in individuals with severe COVID-19 compared with those with mild infection or no prior infection. We identify epitopes near the catalytic domain of ACE2 targeted by these antibodies. Levels of autoantibodies targeting ACE2 and other immune factors could serve as determinants of COVID-19 disease severity, and represent a natural immunoregulatory mechanism in response to viral infection. CONCLUSIONS These results demonstrate that SARS-CoV-2 infection can increase autoantibody levels to ACE2 and other immune factors. The levels of these autoantibodies are associated with COVID-19 disease severity.
Collapse
Affiliation(s)
- Eric S Geanes
- Genomic Medicine Center, Children's Mercy Research Institute, Kansas City, MO, USA
| | - Rebecca McLennan
- Genomic Medicine Center, Children's Mercy Research Institute, Kansas City, MO, USA
| | - Cas LeMaster
- Genomic Medicine Center, Children's Mercy Research Institute, Kansas City, MO, USA
| | - Todd Bradley
- Genomic Medicine Center, Children's Mercy Research Institute, Kansas City, MO, USA.
- Department of Pediatrics, University of Missouri, Kansas City, MO, USA.
- Department of Pediatrics, University of Kansas Medical Center, Kansas City, KS, USA.
- Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, KS, USA.
| |
Collapse
|
247
|
Wang W, Wang YH, Huang CH, Hsieh TH, Ibarburu GH, Wei JCC. Paxlovid use is associated with lower risk of cardiovascular diseases in COVID-19 patients with autoimmune rheumatic diseases: a retrospective cohort study. BMC Med 2024; 22:117. [PMID: 38481216 PMCID: PMC10938827 DOI: 10.1186/s12916-024-03331-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/08/2023] [Accepted: 02/29/2024] [Indexed: 03/17/2024] Open
Abstract
BACKGROUND Paxlovid has been shown to be effective in reducing mortality and hospitalization rates in patients with coronavirus disease 2019 (COVID-19). It is not known whether Paxlovid can reduce the risk of cardiovascular diseases (CVD) in COVID-19-surviving patients with autoimmune rheumatic diseases (AIRDs). METHODS TriNetX data from the US Collaborative Network were used in this study. A total of 5,671,395 patients with AIRDs were enrolled between January 1, 2010, and December 31, 2021. People diagnosed with COVID-19 were included in the cohort (n = 238,142) from January 1, 2022, to December 31, 2022. The Study population was divided into two groups based on Paxlovid use. Propensity score matching was used to generate groups with matched baseline characteristics. The hazard ratios (HRs) and 95% confidence intervals of cardiovascular outcomes, admission rate, mortality rate, and intensive care unit (ICU) admission rate were calculated between Paxlovid and non-Paxlovid groups. Subgroup analyses on sex, age, race, autoimmune diseases group, and sensitivity analyses for Paxlovid use within the first day or within 2-5 days of COVID-19 diagnosis were performed. RESULTS Paxlovid use was associated with lower risks of cerebrovascular complications (HR = 0.65 [0.47-0.88]), arrhythmia outcomes (HR = 0.81 [0.68-0.94]), ischemic heart disease, other cardiac disorders (HR = 0.51 [0.35-0.74]) naming heart failure (HR = 0.41 [0.26-0.63]) and deep vein thrombosis (HR = 0.46 [0.24-0.87]) belonging to thrombotic disorders in AIRD patients with COVID-19. Compared with the Non-Paxlovid group, risks of major adverse cardiac events (HR = 0.56 [0.44-0.70]) and any cardiovascular outcome mentioned above (HR = 0.76 [0.66-0.86]) were lower in the Paxlovid group. Moreover, the mortality (HR = 0.21 [0.11-0.40]), admission (HR = 0.68 [0.60-0.76]), and ICU admission rates (HR = 0.52 [0.33-0.80]) were significantly lower in the Paxlovid group than in the non-Paxlovid group. Paxlovid appears to be more effective in male, older, and Black patients with AIRD. The risks of cardiovascular outcomes and severe conditions were reduced significantly with Paxlovid prescribed within the first day of COVID-19 diagnosis. CONCLUSIONS Paxlovid use is associated with a lower risk of CVDs and severe conditions in COVID-19-surviving patients with AIRD.
Collapse
Affiliation(s)
- Weijie Wang
- Department of Rheumatology, The Second Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, China
- Institute of Basic Theory for Chinese Medicine, China Academy of Chinese Medical Science, Beijing, China
| | - Yu-Hsun Wang
- Department of Medical Research, Chung Shan Medical University Hospital, Taichung, Taiwan
| | - Ching-Hua Huang
- Department of Allergy, Immunology & Rheumatology, Chung Shan Medical University Hospital, Taichung, Taiwan.
- Department of Pharmacy, Chung Shan Medical University Hospital, Taichung, Taiwan.
| | - Tsung-Hsueh Hsieh
- Institute of Medicine, Chung Shan Medical University, Taichung, Taiwan
| | | | - James Cheng-Chung Wei
- Department of Allergy, Immunology & Rheumatology, Chung Shan Medical University Hospital, Taichung, Taiwan.
- Institute of Medicine, Chung Shan Medical University, Taichung, Taiwan.
- Graduate Institute of Integrated Medicine, China Medical University, Taichung, Taiwan.
- Department of Medical Research, Taichung Veterans General Hospital, Taichung, Taiwan.
| |
Collapse
|
248
|
Peel JN, Yang R, Le Voyer T, Gervais A, Rosain J, Bastard P, Behere A, Cederholm A, Bodansky A, Seeleuthner Y, Conil C, Ding JY, Lei WT, Bizien L, Soudee C, Migaud M, Ogishi M, Yatim A, Lee D, Bohlen J, Perpoint T, Perez L, Messina F, Genet R, Karkowski L, Blot M, Lafont E, Toullec L, Goulvestre C, Mehlal-Sedkaoui S, Sallette J, Martin F, Puel A, Jouanguy E, CONSTANCES cohort, 3C-Dijon Study, Etablissement du Sang study group, Anderson MS, Landegren N, Tiberghien P, Abel L, Boisson-Dupuis S, Bustamante J, Ku CL, Casanova JL. Neutralizing IFN-γ autoantibodies are rare and pathogenic in HLA-DRB1*15:02 or 16:02 individuals. J Clin Invest 2024; 134:e178263. [PMID: 38470480 PMCID: PMC11014650 DOI: 10.1172/jci178263] [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: 12/07/2023] [Accepted: 03/01/2024] [Indexed: 03/13/2024] Open
Abstract
BACKGROUNDWeakly virulent environmental mycobacteria (EM) can cause severe disease in HLA-DRB1*15:02 or 16:02 adults harboring neutralizing anti-IFN-γ autoantibodies (nAIGAs). The overall prevalence of nAIGAs in the general population is unknown, as are the penetrance of nAIGAs in HLA-DRB1*15:02 or 16:02 individuals and the proportion of patients with unexplained, adult-onset EM infections carrying nAIGAs.METHODSThis study analyzed the detection and neutralization of anti-IFN-γ autoantibodies (auto-Abs) from 8,430 healthy individuals of the general population, 257 HLA-DRB1*15:02 or 16:02 carriers, 1,063 patients with autoimmune disease, and 497 patients with unexplained severe disease due to EM.RESULTSWe found that anti-IFN-γ auto-Abs detected in 4,148 of 8,430 healthy individuals (49.2%) from the general population of an unknown HLA-DRB1 genotype were not neutralizing. Moreover, we did not find nAIGAs in 257 individuals carrying HLA-DRB1* 15:02 or 16:02. Additionally, nAIGAs were absent in 1,063 patients with an autoimmune disease. Finally, 7 of 497 patients (1.4%) with unexplained severe disease due to EM harbored nAIGAs.CONCLUSIONThese findings suggest that nAIGAs are isolated and that their penetrance in HLA-DRB1*15:02 or 16:02 individuals is low, implying that they may be triggered by rare germline or somatic variants. In contrast, the risk of mycobacterial disease in patients with nAIGAs is high, confirming that these nAIGAs are the cause of EM disease.FUNDINGThe Laboratory of Human Genetics of Infectious Diseases is supported by the Howard Hughes Medical Institute, the Rockefeller University, the St. Giles Foundation, the National Institutes of Health (NIH) (R01AI095983 and U19AIN1625568), the National Center for Advancing Translational Sciences (NCATS), the NIH Clinical and Translational Science Award (CTSA) program (UL1 TR001866), the French National Research Agency (ANR) under the "Investments for the Future" program (ANR-10-IAHU-01), the Integrative Biology of Emerging Infectious Diseases Laboratory of Excellence (ANR-10-LABX-62-IBEID), ANR-GENMSMD (ANR-16-CE17-0005-01), ANR-MAFMACRO (ANR-22-CE92-0008), ANRSECTZ170784, the French Foundation for Medical Research (FRM) (EQU201903007798), the ANRS-COV05, ANR GENVIR (ANR-20-CE93-003), and ANR AI2D (ANR-22-CE15-0046) projects, the ANR-RHU program (ANR-21-RHUS-08-COVIFERON), the European Union's Horizon 2020 research and innovation program under grant agreement no. 824110 (EASI-genomics), the Square Foundation, Grandir - Fonds de solidarité pour l'enfance, the Fondation du Souffle, the SCOR Corporate Foundation for Science, the Battersea & Bowery Advisory Group, William E. Ford, General Atlantic's Chairman and Chief Executive Officer, Gabriel Caillaux, General Atlantic's Co-President, Managing Director, and Head of business in EMEA, and the General Atlantic Foundation, Institut National de la Santé et de la Recherche Médicale (INSERM) and of Paris Cité University. JR was supported by the INSERM PhD program for doctors of pharmacy (poste d'accueil INSERM). JR and TLV were supported by the Bettencourt-Schueller Foundation and the MD-PhD program of the Imagine Institute. MO was supported by the David Rockefeller Graduate Program, the Funai Foundation for Information Technology (FFIT), the Honjo International Scholarship Foundation (HISF), and the New York Hideyo Noguchi Memorial Society (HNMS).
Collapse
Affiliation(s)
- Jessica N. Peel
- St. Giles Laboratory of Human Genetics of Infectious Diseases, The Rockefeller University, New York, New York, USA
| | - Rui Yang
- St. Giles Laboratory of Human Genetics of Infectious Diseases, The Rockefeller University, New York, New York, USA
| | - Tom Le Voyer
- St. Giles Laboratory of Human Genetics of Infectious Diseases, The Rockefeller University, New York, New York, USA
- Laboratory of Human Genetics of Infectious Diseases, Inserm U1163, Paris, France
- Paris Cité University, Imagine Institute, Paris, France
- Clinical Immunology Department, Assistance Publique Hôpitaux de Paris (AP-HP), Saint-Louis Hospital, Paris, France
| | - Adrian Gervais
- Laboratory of Human Genetics of Infectious Diseases, Inserm U1163, Paris, France
- Paris Cité University, Imagine Institute, Paris, France
| | - Jérémie Rosain
- Laboratory of Human Genetics of Infectious Diseases, Inserm U1163, Paris, France
- Paris Cité University, Imagine Institute, Paris, France
- Study Center for Primary Immunodeficiencies and
| | - Paul Bastard
- St. Giles Laboratory of Human Genetics of Infectious Diseases, The Rockefeller University, New York, New York, USA
- Laboratory of Human Genetics of Infectious Diseases, Inserm U1163, Paris, France
- Paris Cité University, Imagine Institute, Paris, France
- Pediatric Hematology-Immunology and Rheumatology Unit, Necker Hospital for Sick Children, Assistante Publique-Hôpitaux de Paris (AP-HP), Paris, France
| | - Anish Behere
- Science for Life Laboratory, Department of Medical Biochemistry and Microbiology, Uppsala University, Uppsala, Sweden
| | - Axel Cederholm
- Science for Life Laboratory, Department of Medical Biochemistry and Microbiology, Uppsala University, Uppsala, Sweden
| | - Aaron Bodansky
- Department of Pediatric Critical Care Medicine and
- Department of Medicine, UCSF, San Francisco, California, USA
| | - Yoann Seeleuthner
- Laboratory of Human Genetics of Infectious Diseases, Inserm U1163, Paris, France
- Paris Cité University, Imagine Institute, Paris, France
| | - Clément Conil
- Laboratory of Human Genetics of Infectious Diseases, Inserm U1163, Paris, France
- Paris Cité University, Imagine Institute, Paris, France
| | - Jing-Ya Ding
- Laboratory of Human Immunology and Infectious Disease, Graduate Institute of Clinical Medical Sciences; Center for Molecular and Clinical Immunology, Chang Gung University, Taoyuan, Taiwan
| | - Wei-Te Lei
- Laboratory of Human Immunology and Infectious Disease, Graduate Institute of Clinical Medical Sciences; Center for Molecular and Clinical Immunology, Chang Gung University, Taoyuan, Taiwan
- Department of Pediatrics, Hsinchu Municipal MacKay Children’s Hospital, Hsinchu, Taiwan
| | - Lucy Bizien
- Laboratory of Human Genetics of Infectious Diseases, Inserm U1163, Paris, France
- Paris Cité University, Imagine Institute, Paris, France
| | - Camille Soudee
- Laboratory of Human Genetics of Infectious Diseases, Inserm U1163, Paris, France
- Paris Cité University, Imagine Institute, Paris, France
| | - Mélanie Migaud
- Laboratory of Human Genetics of Infectious Diseases, Inserm U1163, Paris, France
- Paris Cité University, Imagine Institute, Paris, France
| | - Masato Ogishi
- St. Giles Laboratory of Human Genetics of Infectious Diseases, The Rockefeller University, New York, New York, USA
| | - Ahmad Yatim
- St. Giles Laboratory of Human Genetics of Infectious Diseases, The Rockefeller University, New York, New York, USA
| | - Danyel Lee
- St. Giles Laboratory of Human Genetics of Infectious Diseases, The Rockefeller University, New York, New York, USA
- Laboratory of Human Genetics of Infectious Diseases, Inserm U1163, Paris, France
- Paris Cité University, Imagine Institute, Paris, France
| | - Jonathan Bohlen
- Laboratory of Human Genetics of Infectious Diseases, Inserm U1163, Paris, France
- Paris Cité University, Imagine Institute, Paris, France
| | - Thomas Perpoint
- Infectious and Tropical Diseases Service, Hospices Civils of Lyon, Lyon, France
| | - Laura Perez
- Immunology and Rheumatology Unit, Prof. Dr. Juan P. Garrahan National Hospital of Pediatrics, Buenos Aires, Argentina
| | - Fernando Messina
- Mycology Unit, Dr. Francisco J. Muñiz Hospital, Buenos Aires, Argentina
| | - Roxana Genet
- Infectious Diseases Service, Regional Hospital of Metz-Thionville, France
| | - Ludovic Karkowski
- Deparement of Internal Medicine, Sainte Anne Armed Forces Teaching Hospital, Toulon, France
| | - Mathieu Blot
- Department of Infectious Diseases, Dijon-Bourgogne University Hospital, Dijon, France
| | - Emmanuel Lafont
- Department of Infectious Diseases and Tropical Medicine, Paris Cité University, Necker Hospital for Sick Children and
| | - Laurie Toullec
- Laboratory of Immunology, Cochin hospital, AP-HP, Paris, France
| | | | | | | | | | - Anne Puel
- St. Giles Laboratory of Human Genetics of Infectious Diseases, The Rockefeller University, New York, New York, USA
- Laboratory of Human Genetics of Infectious Diseases, Inserm U1163, Paris, France
- Paris Cité University, Imagine Institute, Paris, France
| | - Emmanuelle Jouanguy
- St. Giles Laboratory of Human Genetics of Infectious Diseases, The Rockefeller University, New York, New York, USA
- Laboratory of Human Genetics of Infectious Diseases, Inserm U1163, Paris, France
- Paris Cité University, Imagine Institute, Paris, France
| | | | - 3C-Dijon Study
- Details are available in the Supplemental Acknowledgments
| | | | - Mark S. Anderson
- Department of Pediatric Critical Care Medicine and
- Department of Medicine, UCSF, San Francisco, California, USA
| | - Nils Landegren
- Science for Life Laboratory, Department of Medical Biochemistry and Microbiology, Uppsala University, Uppsala, Sweden
- Centre for Molecular Medicine, Department of Medicine (Solna), Karolinska Institute, Stockholm, Sweden
| | - Pierre Tiberghien
- Etablissement Français Du Sang, La Plaine Saint-Denis, France
- 20UMR1098 RIGHT, INSERM, EFS, Université de Franche-Comté, Besançon, France
| | - Laurent Abel
- St. Giles Laboratory of Human Genetics of Infectious Diseases, The Rockefeller University, New York, New York, USA
- Laboratory of Human Genetics of Infectious Diseases, Inserm U1163, Paris, France
- Paris Cité University, Imagine Institute, Paris, France
| | - Stéphanie Boisson-Dupuis
- St. Giles Laboratory of Human Genetics of Infectious Diseases, The Rockefeller University, New York, New York, USA
- Laboratory of Human Genetics of Infectious Diseases, Inserm U1163, Paris, France
- Paris Cité University, Imagine Institute, Paris, France
| | - Jacinta Bustamante
- St. Giles Laboratory of Human Genetics of Infectious Diseases, The Rockefeller University, New York, New York, USA
- Laboratory of Human Genetics of Infectious Diseases, Inserm U1163, Paris, France
- Paris Cité University, Imagine Institute, Paris, France
- Study Center for Primary Immunodeficiencies and
| | - Cheng-Lung Ku
- Laboratory of Human Immunology and Infectious Disease, Graduate Institute of Clinical Medical Sciences; Center for Molecular and Clinical Immunology, Chang Gung University, Taoyuan, Taiwan
- Department of Nephrology, Chang Gung Memorial Hospital, Taoyuan, Taiwan
| | - Jean-Laurent Casanova
- St. Giles Laboratory of Human Genetics of Infectious Diseases, The Rockefeller University, New York, New York, USA
- Laboratory of Human Genetics of Infectious Diseases, Inserm U1163, Paris, France
- Paris Cité University, Imagine Institute, Paris, France
- Pediatric Hematology-Immunology and Rheumatology Unit, Necker Hospital for Sick Children, Assistante Publique-Hôpitaux de Paris (AP-HP), Paris, France
- Howard Hughes Medical Institute, New York, New York, USA
| |
Collapse
|
249
|
Shi D, Chen J, Zhao M, Tang Y, Zhao C, Jin Y, Tian D, Liao Y, Wang X, Wang W, Fan X, Yi Z, Chen X, Ling Y. Prevalence of Neutralizing Autoantibodies Against Type I Interferon in a Multicenter Cohort of Severe or Critical COVID-19 Cases in Shanghai. J Clin Immunol 2024; 44:80. [PMID: 38462559 PMCID: PMC10925575 DOI: 10.1007/s10875-024-01683-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2023] [Accepted: 03/04/2024] [Indexed: 03/12/2024]
Abstract
OBJECTIVE We sought to explore the prevalence of type I interferon-neutralizing antibodies in a Chinese cohort and its clinical implications during the Omicron variant wave of SARS-CoV-2. METHODS Type I interferon (IFN) autoantibodies possessing neutralizing capabilities were identified using luciferase assays. The capacity of the autoantibodies for in vitro interference with antiviral activity of IFN was assessed by using a SARS-CoV-2 replicon system. An analysis of the demographic and clinical profiles of patients exhibiting neutralizing antibodies was also conducted. RESULTS In this cohort, 11.8% of severe/critical cases exhibited the existence of type I IFN-neutralizing antibodies, specifically targeting IFN-α2, IFN-ω, or both, with an elderly male patient tendency. Notably, these antibodies exerted a pronounced inhibitory effect on the antiviral activity of IFN against SARS-CoV-2 under controlled in vitro conditions. Furthermore, a noteworthy correlation was discerned between the presence of these neutralizing antibodies and critical clinical parameters, including C-reactive protein (CRP) levels, D-dimer levels, and lymphocyte counts. CONCLUSION The presence of type I IFN-neutralizing antibodies is a pervasive risk factor for severe/critical COVID-19 in the Chinese population.
Collapse
Affiliation(s)
- Dongling Shi
- Department of Infectious Diseases, Shanghai Public Health Clinical Center, Fudan University, Shanghai, China
- Scientific Research Center, Shanghai Public Health Clinical Center, Fudan University, Shanghai, China
| | - Jie Chen
- Department of Infectious Diseases, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Meng Zhao
- Key Laboratory of Medical Molecular Virology (MOE/NHC/CAMS), School of Basic Medical Sciences, and Shanghai Institute of Infectious Disease and Biosecurity, Fudan University, Shanghai, China
| | - Yuanjia Tang
- Shanghai Institute of Rheumatology, Renji Hospital, Shanghai Jiao Tong University School of Medicine (SJTUSM), Shanghai, China
| | - Chen Zhao
- Scientific Research Center, Shanghai Public Health Clinical Center, Fudan University, Shanghai, China
| | - Yinpeng Jin
- Liver Disease Center, Shanghai Public Health Clinical Center, Fudan University, Shanghai, China
| | - Di Tian
- Scientific Research Center, Shanghai Public Health Clinical Center, Fudan University, Shanghai, China
| | - Yixin Liao
- Scientific Research Center, Shanghai Public Health Clinical Center, Fudan University, Shanghai, China
| | - Xuebi Wang
- Department of Infectious Diseases, Shanghai Public Health Clinical Center, Fudan University, Shanghai, China
- Scientific Research Center, Shanghai Public Health Clinical Center, Fudan University, Shanghai, China
| | - Wei Wang
- Scientific Research Center, Shanghai Public Health Clinical Center, Fudan University, Shanghai, China
| | - Xiaohong Fan
- Department of Respiratory Medicine, Shanghai Public Health Clinical Center, Fudan University, Shanghai, China
| | - Zhigang Yi
- Scientific Research Center, Shanghai Public Health Clinical Center, Fudan University, Shanghai, China.
- Key Laboratory of Medical Molecular Virology (MOE/NHC/CAMS), School of Basic Medical Sciences, and Shanghai Institute of Infectious Disease and Biosecurity, Fudan University, Shanghai, China.
| | - Xiaohua Chen
- Department of Infectious Diseases, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Yun Ling
- Department of Infectious Diseases, Shanghai Public Health Clinical Center, Fudan University, Shanghai, China.
- Scientific Research Center, Shanghai Public Health Clinical Center, Fudan University, Shanghai, China.
| |
Collapse
|
250
|
Tsukalov I, Sánchez-Cerrillo I, Rajas O, Avalos E, Iturricastillo G, Esparcia L, Buzón MJ, Genescà M, Scagnetti C, Popova O, Martin-Cófreces N, Calvet-Mirabent M, Marcos-Jimenez A, Martínez-Fleta P, Delgado-Arévalo C, de Los Santos I, Muñoz-Calleja C, Calzada MJ, González Álvaro I, Palacios-Calvo J, Alfranca A, Ancochea J, Sánchez-Madrid F, Martin-Gayo E. NFκB and NLRP3/NLRC4 inflammasomes regulate differentiation, activation and functional properties of monocytes in response to distinct SARS-CoV-2 proteins. Nat Commun 2024; 15:2100. [PMID: 38453949 PMCID: PMC10920883 DOI: 10.1038/s41467-024-46322-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2023] [Accepted: 02/22/2024] [Indexed: 03/09/2024] Open
Abstract
Increased recruitment of transitional and non-classical monocytes in the lung during SARS-CoV-2 infection is associated with COVID-19 severity. However, whether specific innate sensors mediate the activation or differentiation of monocytes in response to different SARS-CoV-2 proteins remain poorly characterized. Here, we show that SARS-CoV-2 Spike 1 but not nucleoprotein induce differentiation of monocytes into transitional or non-classical subsets from both peripheral blood and COVID-19 bronchoalveolar lavage samples in a NFκB-dependent manner, but this process does not require inflammasome activation. However, NLRP3 and NLRC4 differentially regulated CD86 expression in monocytes in response to Spike 1 and Nucleoprotein, respectively. Moreover, monocytes exposed to Spike 1 induce significantly higher proportions of Th1 and Th17 CD4 + T cells. In contrast, monocytes exposed to Nucleoprotein reduce the degranulation of CD8 + T cells from severe COVID-19 patients. Our study provides insights in the differential impact of innate sensors in regulating monocytes in response to different SARS-CoV-2 proteins, which might be useful to better understand COVID-19 immunopathology and identify therapeutic targets.
Collapse
Affiliation(s)
- Ilya Tsukalov
- Medicine Faculty, Universidad Autónoma de Madrid, Madrid, Spain
| | - Ildefonso Sánchez-Cerrillo
- Immunology Unit from Hospital Universitario La Princesa, Instituto Investigación Sanitaria-Princesa IIS-IP, Madrid, Spain
- CIBER Infectious Diseases (CIBERINFECC), Instituto de Salud Carlos III, Madrid, Spain
| | - Olga Rajas
- Pneumology Unit from Hospital Universitario La Princesa, Madrid, Spain
| | - Elena Avalos
- Pneumology Unit from Hospital Universitario La Princesa, Madrid, Spain
| | | | - Laura Esparcia
- Medicine Faculty, Universidad Autónoma de Madrid, Madrid, Spain
- Immunology Unit from Hospital Universitario La Princesa, Instituto Investigación Sanitaria-Princesa IIS-IP, Madrid, Spain
| | - María José Buzón
- Infectious Diseases Department, Institut de Recerca Hospital Univesritari Vall d'Hebrón (VHIR), Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Meritxell Genescà
- Infectious Diseases Department, Institut de Recerca Hospital Univesritari Vall d'Hebrón (VHIR), Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Camila Scagnetti
- Immunology Unit from Hospital Universitario La Princesa, Instituto Investigación Sanitaria-Princesa IIS-IP, Madrid, Spain
| | - Olga Popova
- Medicine Faculty, Universidad Autónoma de Madrid, Madrid, Spain
| | - Noa Martin-Cófreces
- Medicine Faculty, Universidad Autónoma de Madrid, Madrid, Spain
- Immunology Unit from Hospital Universitario La Princesa, Instituto Investigación Sanitaria-Princesa IIS-IP, Madrid, Spain
| | - Marta Calvet-Mirabent
- Medicine Faculty, Universidad Autónoma de Madrid, Madrid, Spain
- Immunology Unit from Hospital Universitario La Princesa, Instituto Investigación Sanitaria-Princesa IIS-IP, Madrid, Spain
| | - Ana Marcos-Jimenez
- Medicine Faculty, Universidad Autónoma de Madrid, Madrid, Spain
- Immunology Unit from Hospital Universitario La Princesa, Instituto Investigación Sanitaria-Princesa IIS-IP, Madrid, Spain
| | - Pedro Martínez-Fleta
- Medicine Faculty, Universidad Autónoma de Madrid, Madrid, Spain
- Immunology Unit from Hospital Universitario La Princesa, Instituto Investigación Sanitaria-Princesa IIS-IP, Madrid, Spain
| | - Cristina Delgado-Arévalo
- Immunology Unit from Hospital Universitario La Princesa, Instituto Investigación Sanitaria-Princesa IIS-IP, Madrid, Spain
| | - Ignacio de Los Santos
- CIBER Infectious Diseases (CIBERINFECC), Instituto de Salud Carlos III, Madrid, Spain
- Infectious Diseases Unit from Hospital Universitario La Princesa, Madrid, Spain
| | - Cecilia Muñoz-Calleja
- Immunology Unit from Hospital Universitario La Princesa, Instituto Investigación Sanitaria-Princesa IIS-IP, Madrid, Spain
- CIBER Infectious Diseases (CIBERINFECC), Instituto de Salud Carlos III, Madrid, Spain
| | - María José Calzada
- Medicine Faculty, Universidad Autónoma de Madrid, Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Respiratorias (CIBERES), Instituto de Salud Carlos III, Madrid, Spain
| | - Isidoro González Álvaro
- Rheumatology Department from Hospital Universitario La Princesa. Instituto de Investigación Sanitaria-Princesa IIS-IP, Madrid, Spain
| | - José Palacios-Calvo
- Department of Pathology, Hospital Universitario Ramón y Cajal. Instituto Ramón y Cajal de Investigaciones Sanitarias (IRYCIS), Universidad de Alcalá. Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Madrid, Spain
| | - Arantzazu Alfranca
- Immunology Unit from Hospital Universitario La Princesa, Instituto Investigación Sanitaria-Princesa IIS-IP, Madrid, Spain
- CIBER Cardiovascular, Instituto de Salud Carlos III, Madrid, Spain
| | - Julio Ancochea
- Pneumology Unit from Hospital Universitario La Princesa, Madrid, Spain
| | - Francisco Sánchez-Madrid
- Medicine Faculty, Universidad Autónoma de Madrid, Madrid, Spain
- Immunology Unit from Hospital Universitario La Princesa, Instituto Investigación Sanitaria-Princesa IIS-IP, Madrid, Spain
- CIBER Cardiovascular, Instituto de Salud Carlos III, Madrid, Spain
| | - Enrique Martin-Gayo
- Medicine Faculty, Universidad Autónoma de Madrid, Madrid, Spain.
- Immunology Unit from Hospital Universitario La Princesa, Instituto Investigación Sanitaria-Princesa IIS-IP, Madrid, Spain.
- CIBER Infectious Diseases (CIBERINFECC), Instituto de Salud Carlos III, Madrid, Spain.
| |
Collapse
|