1
|
Wells TJ, Esposito T, Henderson IR, Labzin LI. Mechanisms of antibody-dependent enhancement of infectious disease. Nat Rev Immunol 2025; 25:6-21. [PMID: 39122820 DOI: 10.1038/s41577-024-01067-9] [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: 07/04/2024] [Indexed: 08/12/2024]
Abstract
Antibody-dependent enhancement (ADE) of infectious disease is a phenomenon whereby host antibodies increase the severity of an infection. It is well established in viral infections but ADE also has an underappreciated role during bacterial, fungal and parasitic infections. ADE can occur during both primary infections and re-infections with the same or a related pathogen; therefore, understanding the underlying mechanisms of ADE is critical for understanding the pathogenesis and progression of many infectious diseases. Here, we review the four distinct mechanisms by which antibodies increase disease severity during an infection. We discuss the most established mechanistic explanation for ADE, where cross-reactive, disease-enhancing antibodies bound to pathogens interact with Fc receptors, thereby enhancing pathogen entry or replication, ultimately increasing the total pathogen load. Additionally, we explore how some pathogenic antibodies can shield bacteria from complement-dependent killing, thereby enhancing bacterial survival. We interrogate the molecular mechanisms by which antibodies can amplify inflammation to drive severe disease, even in the absence of increased pathogen replication. We also examine emerging roles for autoantibodies in enhancing the pathogenesis of infectious diseases. Finally, we discuss how we can leverage these insights to improve vaccine design and future treatments for infectious diseases.
Collapse
Affiliation(s)
- Timothy J Wells
- Frazer Institute, The University of Queensland, Brisbane, Queensland, Australia.
| | - Tyron Esposito
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland, Australia
| | - Ian R Henderson
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland, Australia
| | - Larisa I Labzin
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland, Australia.
| |
Collapse
|
2
|
Samavati L, Talreja J, Peng C. Sarcoidosis Immunodiagnostic Test Detects IgG Antibodies against Unique Antigenic Epitopes. Am J Respir Crit Care Med 2024; 210:1165-1166. [PMID: 39078236 PMCID: PMC11544355 DOI: 10.1164/rccm.202406-1235le] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2024] [Accepted: 07/29/2024] [Indexed: 07/31/2024] Open
Affiliation(s)
- Lobelia Samavati
- Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Medicine, Wayne State University School of Medicine and Detroit Medical Center, Detroit, Michigan; and
- Center for Molecular Medicine and Genetics, Wayne State University School of Medicine, Detroit, Michigan
| | - Jaya Talreja
- Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Medicine, Wayne State University School of Medicine and Detroit Medical Center, Detroit, Michigan; and
| | - Changya Peng
- Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Medicine, Wayne State University School of Medicine and Detroit Medical Center, Detroit, Michigan; and
| |
Collapse
|
3
|
Lettieri S, Bonella F, Marando VA, Franciosi AN, Corsico AG, Campo I. Pathogenesis-driven treatment of primary pulmonary alveolar proteinosis. Eur Respir Rev 2024; 33:240064. [PMID: 39142709 PMCID: PMC11322829 DOI: 10.1183/16000617.0064-2024] [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: 03/25/2024] [Accepted: 05/28/2024] [Indexed: 08/16/2024] Open
Abstract
Pulmonary alveolar proteinosis (PAP) is a syndrome that results from the accumulation of lipoproteinaceous material in the alveolar space. According to the underlying pathogenetic mechanisms, three different forms have been identified, namely primary, secondary and congenital. Primary PAP is caused by disruption of granulocyte-macrophage colony-stimulating factor (GM-CSF) signalling due to the presence of neutralising autoantibodies (autoimmune PAP) or GM-CSF receptor genetic defects (hereditary PAP), which results in dysfunctional alveolar macrophages with reduced phagocytic clearance of particles, cholesterol and surfactant. The serum level of GM-CSF autoantibody is the only disease-specific biomarker of autoimmune PAP, although it does not correlate with disease severity. In PAP patients with normal serum GM-CSF autoantibody levels, elevated serum GM-CSF levels is highly suspicious for hereditary PAP. Several biomarkers have been correlated with disease severity, although they are not specific for PAP. These include lactate dehydrogenase, cytokeratin 19 fragment 21.1, carcinoembryonic antigen, neuron-specific enolase, surfactant proteins, Krebs von Lungen 6, chitinase-3-like protein 1 and monocyte chemotactic proteins. Finally, increased awareness of the disease mechanisms has led to the development of pathogenesis-based treatments, such as GM-CSF augmentation and cholesterol-targeting therapies.
Collapse
Affiliation(s)
- Sara Lettieri
- Pneumology Unit, IRCCS San Matteo Hospital Foundation, Pavia, Italy
| | - Francesco Bonella
- Center for interstitial and rare lung diseases, Ruhrlandklinik, University of Essen, Essen, Germany
| | | | | | - Angelo Guido Corsico
- Pneumology Unit, IRCCS San Matteo Hospital Foundation, Pavia, Italy
- Department of Internal Medicine, University of Pavia, Pavia, Italy
| | - Ilaria Campo
- Pneumology Unit, IRCCS San Matteo Hospital Foundation, Pavia, Italy
| |
Collapse
|
4
|
Paparoditis P, Shulman Z. The tumor-driven antibody-mediated immune response in cancer. Curr Opin Immunol 2024; 88:102431. [PMID: 38866666 DOI: 10.1016/j.coi.2024.102431] [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: 01/15/2024] [Revised: 05/21/2024] [Accepted: 05/27/2024] [Indexed: 06/14/2024]
Abstract
Immune cells in the tumor microenvironment play a crucial role in cancer prognosis and response to immunotherapy. Recent studies highlight the significance of tumor-infiltrating B cells and tertiary lymphoid structures as markers of favorable prognosis and patient-positive response to immune checkpoint blockers in some types of cancer. Although the presence of germinal center B cells and plasma cells in the tumor microenvironment has been established, determining their tumor reactivity remains challenging. The few known tumor targets range from viral proteins to self and altered self-proteins. The emergence of self-reactive antibodies in patients with cancer, involves the opposing forces of antigen-driven affinity increase and peripheral tolerance mechanisms. Here, B cell tumor antigen specificity and affinity maturation in tumor-directed immune responses in cancer are discussed.
Collapse
Affiliation(s)
- Philipp Paparoditis
- Department of Systems Immunology, The Weizmann Institute of Science, Rehovot 7610001, Israel
| | - Ziv Shulman
- Department of Systems Immunology, The Weizmann Institute of Science, Rehovot 7610001, Israel.
| |
Collapse
|
5
|
Das S, Stamnaes J, Høydahl LS, Skagen C, Lundin KEA, Jahnsen J, Sollid LM, Iversen R. Selective activation of naïve B cells with unique epitope specificity shapes autoantibody formation in celiac disease. J Autoimmun 2024; 146:103241. [PMID: 38754235 DOI: 10.1016/j.jaut.2024.103241] [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: 01/16/2024] [Revised: 03/25/2024] [Accepted: 05/03/2024] [Indexed: 05/18/2024]
Abstract
Many antibody responses induced by infection, vaccination or autoimmunity show signs of convergence across individuals with epitope-dependent selection of particular variable region gene segments and complementarity determining region 3 properties. However, not much is known about the relationship between antigen-specific effector cells and antigen-specific precursors present in the naïve B-cell repertoire. Here, we sought to address this relationship in the context of celiac disease, where there is a stereotyped autoantibody response against the enzyme transglutaminase 2 (TG2). By generating TG2-specific monoclonal antibodies from both duodenal plasma cells and circulating naïve B cells, we demonstrate a discord between the naïve TG2-specific repertoire and the cells that are selected for autoantibody production. Hence, the naïve repertoire does not fully reflect the epitope preference and gene usage observed for memory B cells and plasma cells. Instead, distinct naïve B cells that target particular TG2 epitopes appear to be selectively activated at the expense of TG2-binding B cells targeting other epitopes.
Collapse
Affiliation(s)
- Saykat Das
- Norwegian Coeliac Disease Research Centre, Institute of Clinical Medicine, University of Oslo, Oslo, Norway; Department of Immunology, Oslo University Hospital-Rikshospitalet, Oslo, Norway
| | - Jorunn Stamnaes
- Norwegian Coeliac Disease Research Centre, Institute of Clinical Medicine, University of Oslo, Oslo, Norway; Department of Immunology, Oslo University Hospital-Rikshospitalet, Oslo, Norway
| | - Lene S Høydahl
- Norwegian Coeliac Disease Research Centre, Institute of Clinical Medicine, University of Oslo, Oslo, Norway; Department of Immunology, Oslo University Hospital-Rikshospitalet, Oslo, Norway
| | - Christine Skagen
- Department of Immunology, Oslo University Hospital-Rikshospitalet, Oslo, Norway
| | - Knut E A Lundin
- Norwegian Coeliac Disease Research Centre, Institute of Clinical Medicine, University of Oslo, Oslo, Norway; Department of Gastroenterology, Oslo University Hospital-Rikshospitalet, Oslo, Norway
| | - Jørgen Jahnsen
- Department of Gastroenterology, Akershus University Hospital, Lørenskog, Norway
| | - Ludvig M Sollid
- Norwegian Coeliac Disease Research Centre, Institute of Clinical Medicine, University of Oslo, Oslo, Norway; Department of Immunology, Oslo University Hospital-Rikshospitalet, Oslo, Norway
| | - Rasmus Iversen
- Norwegian Coeliac Disease Research Centre, Institute of Clinical Medicine, University of Oslo, Oslo, Norway; Department of Immunology, Oslo University Hospital-Rikshospitalet, Oslo, Norway.
| |
Collapse
|
6
|
Mabo A, Borie R, Wemeau-Stervinou L, Uzunhan Y, Gomez E, Prevot G, Reynaud-Gaubert M, Traclet J, Bergot E, Cadranel J, Marchand-Adam S, Bergeron A, Blanchard E, Bondue B, Bonniaud P, Bourdin A, Burgel PR, Hirschi S, Marquette CH, Quétant S, Nunes H, Chenivesse C, Crestani B, Guirriec Y, Monnier D, Ménard C, Tattevin P, Cottin V, Luque Paz D, Jouneau S. Infections in autoimmune pulmonary alveolar proteinosis: a large retrospective cohort. Thorax 2023; 79:68-74. [PMID: 37758458 DOI: 10.1136/thorax-2023-220040] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2023] [Accepted: 08/21/2023] [Indexed: 10/03/2023]
Abstract
BACKGROUND Autoimmune pulmonary alveolar proteinosis (aPAP) is a rare disease, predisposing to an increased risk of infection. A complete picture of these infections is lacking. RESEARCH QUESTION Describe the characteristics and clinical outcomes of patients diagnosed with aPAP, and to identify risk factors associated with opportunistic infections. METHODS We conducted a retrospective cohort including all patients diagnosed with aPAP between 2008 and 2018 in France and Belgium. Data were collected using a standardised questionnaire including demographics, comorbidities, imaging features, outcomes and microbiological data. RESULTS We included 104 patients, 2/3 were men and median age at diagnosis was 45 years. With a median follow-up of 3.4 years (IQR 1.7-6.6 years), 60 patients (58%), developed at least one infection, including 23 (22%) with opportunistic infections. Nocardia spp was the main pathogen identified (n=10). Thirty-five (34%) patients were hospitalised due to infection. In univariate analysis, male gender was associated with opportunistic infections (p=0.04, OR=3.88; 95% CI (1.02 to 22.06)). Anti-granulocyte macrophage colony-stimulating factor antibody titre at diagnosis was significantly higher among patients who developed nocardiosis (1058 (316-1591) vs 580 (200-1190), p=0.01). Nine patients had died (9%), but only one death was related to infection. INTERPRETATION Patients with aPAP often presented with opportunistic infections, especially nocardiosis, which highlights the importance of systematic search for slow-growing bacteria in bronchoalveolar lavage or whole lung lavage.
Collapse
Affiliation(s)
- Axelle Mabo
- Service de Pneumologie, Centre de Compétence pour les Maladies Pulmonaires Rares, Hôpital Pontchaillou, CHU Rennes, Rennes, France
| | - Raphael Borie
- Centre de Référence Constitutif des Maladies Pulmonaires Rares, Service de Pneumologie A, Hopital Bichat, APHP, Paris, France
| | - Lidwine Wemeau-Stervinou
- Centre de Référence Constitutif des Maladies Pulmonaires Rares, Institut Cœur-Poumon, Service de Pneumologie et Immuno-Allergologie, CHRU Lille, Lille, France
| | - Yurdagül Uzunhan
- Centre de Référence Constitutif des Maladies Pulmonaires Rares, Service de Pneumologie, Hôpital Avicenne, APHP, Bobigny, France
| | - Emmanuel Gomez
- Centre de Compétence pour les Maladies Pulmonaires Rares, Département de Pneumologie, Hôpitaux de Brabois, CHRU de Nancy, Vandoeuvre-les Nancy, France
| | - Gregoire Prevot
- Service de Pneumologie, Centre de Compétence pour les Maladies Pulmonaires Rares, Hôpital Larry, CHU Toulouse, Toulouse, France
| | - Martine Reynaud-Gaubert
- Service de Pneumologie et Transplantation Pulmonaire, Centre de Compétences des Maladies Rares Pulmonaires et de l'Hypertension Pulmonaire, CHU Nord de Marseille, AP-HM, Aix Marseille Université, Marseille, France
| | - Julie Traclet
- Service de Pneumologie, Centre National Coordonnateur de Référence des Maladies Pulmonaires Rares, Hôpital Louis-Pradel, Hospices Civils de Lyon (HCL), UMR754, INRAE, Université Lyon 1, ERN-LUNG, Lyon, France
| | - Emmanuel Bergot
- Centre de Compétence pour les Maladies Pulmonaires Rares de l'Adulte, Service de Pneumologie et Oncologie Thoracique, Hôpital Côte de Nacre, CHU de Caen, Caen, France
| | - Jacques Cadranel
- Service de Pneumologie et Oncologie Thoracique, Centre Constitutif Maladies Pulmonaires Rares, Hôpital Tenon, APHP, Sorbonne Université, Paris, France
| | - Sylvain Marchand-Adam
- Service de Pneumologie, CHRU de Tours, Université François Rabelais de Tours, INSERM U1100, Tours, France
| | - Anne Bergeron
- Service de Pneumologie, Hôpitaux Universitaires de Genève, Genève, Switzerland
| | - Elodie Blanchard
- Service de Pneumologie, centre de compétence pour les maladies pulmonaires rares, CHU de Bordeaux, Pessac, France
| | - Benjamin Bondue
- Service de Pneumologie, CUB Hôpital Erasme, Université Libre de Bruxelles, Bruxelles, Belgium
| | - Philippe Bonniaud
- Service de Pneumologie et Soins Intensifs Respiratoires, Centre de Référence Constitutif des Maladies Pulmonaires Rares de l'Adulte, CHU Dijon-Bourgogne, Inserm U123, Université de Bourgogne, Dijon, France
| | - Arnaud Bourdin
- Service de Pneumologie, CHU Montpellier, Université de Montpellier, Inserm U1046, Montpellier, France
| | - Pierre Regis Burgel
- Service de Pneumologie, Hopital Cochin Pneumologie, AP-HP, Université Paris Cité Paris, Paris, France
| | - Sandrine Hirschi
- Service de Pneumologie et Transplantation Pulmonaire, Hopitaux universitaires de Strasbourg, Strasbourg, France
| | - Charles Hugo Marquette
- Service de Pneumologie, CHU Nice, Fédération Hospitalo-Universitaire OncoAge, Nice, France
| | - Sébastien Quétant
- Service Hospitalo-Universitaire de Pneumologie et Physiologie, Pôle Thorax et Vaisseaux, CHU de Grenoble-Alpes, La Tronche, Grenoble, France
| | - Hilario Nunes
- Centre de Référence Constitutif des Maladies Pulmonaires Rares, Service de Pneumologie, Hôpital Avicenne, APHP, Bobigny, France
| | - Cécile Chenivesse
- Centre de Référence Constitutif des Maladies Pulmonaires Rares, Institut Cœur-Poumon, Service de Pneumologie et Immuno-Allergologie, CHRU Lille, Lille, France
| | - Bruno Crestani
- Centre de Référence Constitutif des Maladies Pulmonaires Rares, Service de Pneumologie A, Hopital Bichat, APHP, Paris, France
| | - Yoann Guirriec
- Service de Pneumologie, Centre de Compétence pour les Maladies Pulmonaires Rares, Hôpital Pontchaillou, CHU Rennes, Rennes, France
| | - Delphine Monnier
- Service d'Immunologie, Laboratoire de Biologie Médicale de Référence Lipoprotéinose Alvéolaire, Hôpital Pontchaillou, CHU Rennes, Rennes, France
| | - Cédric Ménard
- Service d'Immunologie, Laboratoire de Biologie Médicale de Référence Lipoprotéinose Alvéolaire, Hôpital Pontchaillou, CHU Rennes, Rennes, France
| | - Pierre Tattevin
- Service de Maladies Infectieuses et Réanimation Médicale, Hôpital Pontchaillou, Inserm U1230, Université de Rennes, Rennes, France
| | - Vincent Cottin
- Service de Pneumologie, Centre National Coordonnateur de Référence des Maladies Pulmonaires Rares, Hôpital Louis-Pradel, Hospices Civils de Lyon (HCL), UMR754, INRAE, Université Lyon 1, ERN-LUNG, Lyon, France
| | - David Luque Paz
- Service de Pneumologie, Centre de Compétence pour les Maladies Pulmonaires Rares, Hôpital Pontchaillou, CHU Rennes, Rennes, France
- Service de Maladies Infectieuses et Réanimation Médicale, Hôpital Pontchaillou, Inserm U1230, Université de Rennes, Rennes, France
| | - Stéphane Jouneau
- Service de Pneumologie, Centre de Compétence pour les Maladies Pulmonaires Rares, Hôpital Pontchaillou, CHU Rennes, Rennes, France
- Inserm UMR1085 IRSET, Université de Rennes, EHESP, Rennes, France
| |
Collapse
|
7
|
Taylor PC, Weinblatt ME, McInnes IB, Atsumi T, Strand V, Takeuchi T, Bracher M, Brooks D, Davies J, Goode C, Gupta A, Mukherjee S, O'Shea C, Saurigny D, Schifano LA, Shelton C, Smith JE, Wang M, Wang R, Watts S, Fleischmann RM. Anti-GM-CSF otilimab versus sarilumab or placebo in patients with rheumatoid arthritis and inadequate response to targeted therapies: a phase III randomised trial (contRAst 3). Ann Rheum Dis 2023; 82:1527-1537. [PMID: 37696589 PMCID: PMC10646837 DOI: 10.1136/ard-2023-224449] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2023] [Accepted: 08/23/2023] [Indexed: 09/13/2023]
Abstract
OBJECTIVES To investigate the efficacy and safety of otilimab, an anti-granulocyte-macrophage colony-stimulating factor antibody, in patients with active rheumatoid arthritis and an inadequate response to conventional synthetic (cs) and biologic disease-modifying antirheumatic drugs (DMARDs) and/or Janus kinase inhibitors. METHODS ContRAst 3 was a 24-week, phase III, multicentre, randomised controlled trial. Patients received subcutaneous otilimab (90/150 mg once weekly), subcutaneous sarilumab (200 mg every 2 weeks) or placebo for 12 weeks, in addition to csDMARDs. Patients receiving placebo were switched to active interventions at week 12 and treatment continued to week 24. The primary end point was the proportion of patients achieving an American College of Rheumatology ≥20% response (ACR20) at week 12. RESULTS Overall, 549 patients received treatment. At week 12, there was no significant difference in the proportion of ACR20 responders with otilimab 90 mg and 150 mg versus placebo (45% (p=0.2868) and 51% (p=0.0596) vs 38%, respectively). There were no significant differences in Clinical Disease Activity Index, Health Assessment Questionnaire-Disability Index, pain Visual Analogue Scale or Functional Assessment of Chronic Illness Therapy-Fatigue scores with otilimab versus placebo at week 12. Sarilumab demonstrated superiority to otilimab in ACR20 response and secondary end points. The incidence of adverse or serious adverse events was similar across treatment groups. CONCLUSIONS Otilimab demonstrated an acceptable safety profile but failed to achieve the primary end point of ACR20 and improve secondary end points versus placebo or demonstrate non-inferiority to sarilumab in this patient population. TRIAL REGISTRATION NUMBER NCT04134728.
Collapse
Affiliation(s)
- Peter C Taylor
- Botnar Research Centre, Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, University of Oxford, Oxford, UK
| | - Michael E Weinblatt
- Division of Rheumatology, Inflammation and Immunity, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Iain B McInnes
- College of Medical Veterinary and Life Sciences, University of Glasgow, Glasgow, UK
| | - Tatsuya Atsumi
- Department of Rheumatology, Endocrinology and Nephrology, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Vibeke Strand
- Division of Immunology/Rheumatology, Stanford University, Palo Alto, California, USA
| | - Tsutomu Takeuchi
- Department of Internal Medicine, Division of Rheumatology, Keio University School of Medicine, Tokyo, Japan
- Saitama Medical University, Saitama, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | - Roy M Fleischmann
- Department of Medicine, University of Texas Southwestern Medical Center, Dallas, Texas, USA
- Metroplex Clinical Research Center, Dallas, Texas, USA
| |
Collapse
|
8
|
Taghadosi M, Safarzadeh E, Asgarzadeh A, Roghani SA, Shamsi A, Jalili C, Assar S, Soufivand P, Pournazari M, Feizollahi P, Nicknam MH, Asghariazar V, Vaziri S, Shahriari H, Mohammadi A. Partners in crime: Autoantibodies complicit in COVID-19 pathogenesis. Rev Med Virol 2023; 33:e2412. [PMID: 36471421 PMCID: PMC9877745 DOI: 10.1002/rmv.2412] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2022] [Revised: 11/06/2022] [Accepted: 11/18/2022] [Indexed: 12/12/2022]
Abstract
Autoantibodies (AABs) play a critical role in the pathogenesis of autoimmune diseases (AIDs) and serve as a diagnostic and prognostic tool in assessing these complex disorders. Viral infections have long been recognized as a principal environmental factor affecting the production of AABs and the development of autoimmunity. COVID-19 has primarily been considered a hyperinflammatory syndrome triggered by a cytokine storm. In the following, the role of maladaptive B cell response and AABs became more apparent in COVID-19 pathogenesis. The current review will primarily focus on the role of extrafollicular B cell response, Toll-like receptor-7 (TLR-7) activation, and neutrophil extracellular traps (NETs) formation in the development of AABs following SARS-CoV-2 infection. In the following, this review will clarify how these AABs dysregulate immune response to SARS-CoV-2 by disrupting cytokine function and triggering neutrophil hyper-reactivity. Finally, the pathologic effects of these AABs will be further described in COVID-19 associate clinical manifestations, including venous and arterial thrombosis, a multisystem inflammatory syndrome in children (MIS-C), acute respiratory distress syndrome (ARDS), and recently described post-acute sequelae of COVID-19 (PASC) or long-COVID.
Collapse
Affiliation(s)
- Mahdi Taghadosi
- Immunology Department, Faculty of Medicine, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Elham Safarzadeh
- Department of Microbiology, Parasitology, and Immunology, Ardabil University of Medical Sciences, Ardabil, Iran
| | - Ali Asgarzadeh
- Students Research Committee, School of Medicine, Ardabil University of Medical Sciences, Ardabil, Iran
| | - Seyed Askar Roghani
- Medical Biology Research Center, Health Technology Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran.,Clinical Research Development Center, Imam Reza Hospital, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Afsaneh Shamsi
- Immunology Department, Faculty of Medicine, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Cyrus Jalili
- Department of Anatomy, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Shirin Assar
- Clinical Research Development Center, Imam Reza Hospital, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Parviz Soufivand
- Clinical Research Development Center, Imam Reza Hospital, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Mehran Pournazari
- Clinical Research Development Center, Imam Reza Hospital, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Parisa Feizollahi
- Immunology Department, Faculty of Medicine, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Mohammad Hossein Nicknam
- Department of Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran.,Molecular Immunology Research Centre, Tehran University of Medical Sciences, Tehran, Iran
| | - Vahid Asghariazar
- Deputy of Research and Technology, Ardabil University of Medical Sciences, Ardabil, Iran
| | - Siavash Vaziri
- Infectious Disease Research Center, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Hossein Shahriari
- Clinical Immunology Research Center, Zahedan University of Medical Sciences, Zahedan, Iran
| | - Asadollah Mohammadi
- Cellular and Molecular Research Center, Research Institute for Health Development, Kurdistan University of Medical Sciences, Sanandaj, Iran
| |
Collapse
|
9
|
Li SY, Yoshida Y, Kubota M, Zhang BS, Matsutani T, Ito M, Yajima S, Yoshida K, Mine S, Machida T, Hayashi A, Takemoto M, Yokote K, Ohno M, Nishi E, Kitamura K, Kamitsukasa I, Takizawa H, Sata M, Yamagishi K, Iso H, Sawada N, Tsugane S, Iwase K, Shimada H, Iwadate Y, Hiwasa T. Utility of atherosclerosis-associated serum antibodies against colony-stimulating factor 2 in predicting the onset of acute ischemic stroke and prognosis of colorectal cancer. Front Cardiovasc Med 2023; 10:1042272. [PMID: 36844744 PMCID: PMC9954151 DOI: 10.3389/fcvm.2023.1042272] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2022] [Accepted: 01/11/2023] [Indexed: 02/12/2023] Open
Abstract
Introduction Autoantibodies against inflammatory cytokines may be used for the prevention of atherosclerosis. Preclinical studies consider colony-stimulating factor 2 (CSF2) as an essential cytokine with a causal relationship to atherosclerosis and cancer. We examined the serum anti-CSF2 antibody levels in patients with atherosclerosis or solid cancer. Methods We measured the serum anti-CSF2 antibody levels via amplified luminescent proximity homogeneous assay-linked immunosorbent assay based on the recognition of recombinant glutathione S-transferase-fused CSF2 protein or a CSF2-derived peptide as the antigen. Results The serum anti-CSF2 antibody (s-CSF2-Ab) levels were significantly higher in patients with acute ischemic stroke (AIS), acute myocardial infarction (AMI), diabetes mellitus (DM), and chronic kidney disease (CKD) compared with healthy donors (HDs). In addition, the s-CSF2-Ab levels were associated with intima-media thickness and hypertension. The analyzes of samples obtained from a Japan Public Health Center-based prospective study suggested the utility of s-CSF2-Ab as a risk factor for AIS. Furthermore, the s-CSF2-Ab levels were higher in patients with esophageal, colorectal, gastric, and lung cancer than in HDs but not in those with mammary cancer. In addition, the s-CSF2-Ab levels were associated with unfavorable postoperative prognosis in colorectal cancer (CRC). In CRC, the s-CSF2-Ab levels were more closely associated with poor prognosis in patients with p53-Ab-negative CRC despite the lack of significant association of the anti-p53 antibody (p53-Ab) levels with the overall survival. Conclusion S-CSF2-Ab was useful for the diagnosis of atherosclerosis-related AIS, AMI, DM, and CKD and could discriminate poor prognosis, especially in p53-Ab-negative CRC.
Collapse
Affiliation(s)
- Shu-Yang Li
- Department of Neurological Surgery, Graduate School of Medicine, Chiba University, Chiba, Japan
- Department of Biochemistry and Genetics, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Yoichi Yoshida
- Department of Neurological Surgery, Graduate School of Medicine, Chiba University, Chiba, Japan
- Comprehensive Stroke Center, Chiba University Hospital, Chiba, Japan
| | - Masaaki Kubota
- Department of Neurological Surgery, Graduate School of Medicine, Chiba University, Chiba, Japan
- Department of Biochemistry and Genetics, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Bo-Shi Zhang
- Department of Neurological Surgery, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Tomoo Matsutani
- Department of Neurological Surgery, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Masaaki Ito
- Department of Clinical Oncology, Toho University Graduate School of Medicine, Tokyo, Japan
| | - Satoshi Yajima
- Department of Gastroenterological Surgery, Toho University Graduate School of Medicine, Tokyo, Japan
| | - Kimihiko Yoshida
- Department of Gastroenterological Surgery, Toho University Graduate School of Medicine, Tokyo, Japan
| | - Seiichiro Mine
- Department of Neurological Surgery, Graduate School of Medicine, Chiba University, Chiba, Japan
- Department of Neurological Surgery, Chiba Prefectural Sawara Hospital, Chiba, Japan
- Department of Neurological Surgery, Chiba Cerebral and Cardiovascular Center, Chiba, Japan
| | - Toshio Machida
- Department of Neurological Surgery, Graduate School of Medicine, Chiba University, Chiba, Japan
- Department of Neurological Surgery, Chiba Cerebral and Cardiovascular Center, Chiba, Japan
- Department of Neurosurgery, Eastern Chiba Medical Center, Chiba, Japan
| | - Aiko Hayashi
- Department of Endocrinology, Hematology and Gerontology, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Minoru Takemoto
- Department of Endocrinology, Hematology and Gerontology, Graduate School of Medicine, Chiba University, Chiba, Japan
- Department of Diabetes, Metabolism and Endocrinology, School of Medicine, International University of Health and Welfare, Chiba, Japan
| | - Koutaro Yokote
- Department of Endocrinology, Hematology and Gerontology, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Mikiko Ohno
- Department of Cardiovascular Medicine, Graduate School of Medicine, Kyoto University, Kyoto, Japan
- Department of Pharmacology, Shiga University of Medical Science, Otsu, Shiga, Japan
| | - Eiichiro Nishi
- Department of Cardiovascular Medicine, Graduate School of Medicine, Kyoto University, Kyoto, Japan
- Department of Pharmacology, Shiga University of Medical Science, Otsu, Shiga, Japan
| | | | | | - Hirotaka Takizawa
- Port Square Kashiwado Clinic, Kashiwado Memorial Foundation, Chiba, Japan
| | - Mizuki Sata
- Department of Public Health Medicine, Faculty of Medicine, and Health Services Research and Development Center, University of Tsukuba, Tsukuba, Japan
- Department of Preventive Medicine and Public Health, Keio University School of Medicine, Tokyo, Japan
| | - Kazumasa Yamagishi
- Department of Public Health Medicine, Faculty of Medicine, and Health Services Research and Development Center, University of Tsukuba, Tsukuba, Japan
| | - Hiroyasu Iso
- Public Health, Department of Social Medicine, Osaka University Graduate School of Medicine, Suita, Japan
| | - Norie Sawada
- Division of Cohort Research, National Cancer Center Institute for Cancer Control, Tokyo, Japan
| | - Shoichiro Tsugane
- Division of Cohort Research, National Cancer Center Institute for Cancer Control, Tokyo, Japan
| | - Katsuro Iwase
- Department of Biochemistry and Genetics, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Hideaki Shimada
- Department of Clinical Oncology, Toho University Graduate School of Medicine, Tokyo, Japan
- Department of Gastroenterological Surgery, Toho University Graduate School of Medicine, Tokyo, Japan
| | - Yasuo Iwadate
- Department of Neurological Surgery, Graduate School of Medicine, Chiba University, Chiba, Japan
- Comprehensive Stroke Center, Chiba University Hospital, Chiba, Japan
| | - Takaki Hiwasa
- Department of Neurological Surgery, Graduate School of Medicine, Chiba University, Chiba, Japan
- Department of Biochemistry and Genetics, Graduate School of Medicine, Chiba University, Chiba, Japan
- Comprehensive Stroke Center, Chiba University Hospital, Chiba, Japan
- Department of Clinical Oncology, Toho University Graduate School of Medicine, Tokyo, Japan
| |
Collapse
|
10
|
Salvator H, Cheng A, Rosen LB, Williamson PR, Bennett JE, Kashyap A, Ding L, Kwon-Chung KJ, Namkoong H, Zerbe CS, Holland SM. Neutralizing GM-CSF autoantibodies in pulmonary alveolar proteinosis, cryptococcal meningitis and severe nocardiosis. Respir Res 2022; 23:280. [PMID: 36221098 PMCID: PMC9552154 DOI: 10.1186/s12931-022-02103-9] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2022] [Accepted: 06/30/2022] [Indexed: 12/05/2022] Open
Abstract
Background Anti GM-CSF autoantibodies (aAb) have been related to acquired pulmonary alveolar proteinosis (PAP) and described in cases of severe infections such as cryptococcosis and nocardiosis in previously healthy subjects. Whether there are different anti-GM-CSF autoantibodies corresponding to these phenotypes is unclear. Therefore, we examined anti-GM-CSF autoantibodies to determine whether amount or neutralizing activity could distinguish between groups. Methods Plasma samples gathered in the National Institute of Health from patients with anti GM-CSF aAb and either PAP (n = 15), cryptococcal meningitis (n = 15), severe nocardiosis (n = 5) or overlapping phenotypes (n = 6) were compared. The relative amount of aAb was assessed using a particle-based approach, reported as a mouse monoclonal anti-human GM-CSF as standard curve and expressed in an arbitrary Mouse Monoclonal Antibody Unit (MMAU). The neutralizing activity of the plasma was assessed by inhibition of GM-CSF-induced intracellular phospho-STAT5 (pSTAT5) in monocytes. Results Anti-GM-CSF aAb relative amounts were higher in PAP patients compared to those with cryptococcosis (mean 495 ± 464 MMAU vs 197 ± 159 MMAU, p = 0.02); there was no difference with patients with nocardiosis (430 ± 493 MMAU) nor between the two types of infections. The dilution of plasma resulting in 50% inhibition of GM-CSF-induced pSTAT5 (approximate IC50) did not vary appreciably across groups of patients (1.6 ± 3.1%, 3.9 ± 6% and 1.8 ± 2.2% in PAP patients, cryptococcosis and nocardiosis patients, respectively). Nor was the concentration of GM-CSF necessary to induce 50% of maximal GM-CSF-induced pSTAT5 in the presence of 10 MMAU of anti-GM-CSF aAb (EC50). When studying longitudinal samples from patients with PAP or disseminated nocardiosis, the neutralizing effect of anti-GM-CSF aAb was relatively constant over time despite targeted treatments and variations in aAb levels. Conclusions Despite different clinical manifestations, anti-GM-CSF antibodies were similar across PAP, cryptococcosis and nocardiosis. Underlying host genetics and functional analyses may help further differentiate the biology of these conditions.
Collapse
Affiliation(s)
- Hélène Salvator
- Laboratory of Clinical Immunology and Microbiology, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA.,Department of Respiratory Medicine, Hôpital Foch, Suresnes, France-UMR 0892 VIM Suresnes, INRAE Paris Saclay University, Jouy-en-Josas, France
| | - Aristine Cheng
- Laboratory of Clinical Immunology and Microbiology, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA.,Division of Infectious Diseases, Department of Medicine, National Taiwan University Hospital and National Taiwan University College of Medicine, Taipei, Taiwan
| | - Lindsey B Rosen
- Laboratory of Clinical Immunology and Microbiology, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Peter R Williamson
- Laboratory of Clinical Immunology and Microbiology, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - John E Bennett
- Laboratory of Clinical Immunology and Microbiology, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Anuj Kashyap
- Laboratory of Clinical Immunology and Microbiology, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA.,Department of Analytical Sciences, BioPharmaceuticals Development, BioPharmaceuticals R&D, AstraZeneca, Gaithersburg, MD, USA
| | - Li Ding
- Laboratory of Clinical Immunology and Microbiology, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Kyung J Kwon-Chung
- Laboratory of Clinical Immunology and Microbiology, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Ho Namkoong
- Laboratory of Clinical Immunology and Microbiology, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA.,Department of Infectious Diseases, Keio University School of Medicine, Tokyo, Japan
| | - Christa S Zerbe
- Laboratory of Clinical Immunology and Microbiology, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Steven M Holland
- Laboratory of Clinical Immunology and Microbiology, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA.
| |
Collapse
|
11
|
Mortha A, Remark R, Del Valle DM, Chuang LS, Chai Z, Alves I, Azevedo C, Gaifem J, Martin J, Petralia F, Tuballes K, Barcessat V, Tai SL, Huang HH, Laface I, Jerez YA, Boschetti G, Villaverde N, Wang MD, Korie UM, Murray J, Choung RS, Sato T, Laird RM, Plevy S, Rahman A, Torres J, Porter C, Riddle MS, Kenigsberg E, Pinho SS, Cho JH, Merad M, Colombel JF, Gnjatic S. Neutralizing Anti-Granulocyte Macrophage-Colony Stimulating Factor Autoantibodies Recognize Post-Translational Glycosylations on Granulocyte Macrophage-Colony Stimulating Factor Years Before Diagnosis and Predict Complicated Crohn's Disease. Gastroenterology 2022; 163:659-670. [PMID: 35623454 PMCID: PMC10127946 DOI: 10.1053/j.gastro.2022.05.029] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/20/2021] [Revised: 05/09/2022] [Accepted: 05/12/2022] [Indexed: 02/07/2023]
Abstract
BACKGROUND & AIMS Anti-granulocyte macrophage-colony stimulating factor autoantibodies (aGMAbs) are detected in patients with ileal Crohn's disease (CD). Their induction and mode of action during or before disease are not well understood. We aimed to investigate the underlying mechanisms associated with aGMAb induction, from functional orientation to recognized epitopes, for their impact on intestinal immune homeostasis and use as a predictive biomarker for complicated CD. METHODS We characterized using enzyme-linked immunosorbent assay naturally occurring aGMAbs in longitudinal serum samples from patients archived before the diagnosis of CD (n = 220) as well as from 400 healthy individuals (matched controls) as part of the US Defense Medical Surveillance System. We used biochemical, cellular, and transcriptional analysis to uncover a mechanism that governs the impaired immune balance in CD mucosa after diagnosis. RESULTS Neutralizing aGMAbs were found to be specific for post-translational glycosylation on granulocyte macrophage-colony stimulating factor (GM-CSF), detectable years before diagnosis, and associated with complicated CD at presentation. Glycosylation of GM-CSF was altered in patients with CD, and aGMAb affected myeloid homeostasis and promoted group 1 innate lymphoid cells. Perturbations in immune homeostasis preceded the diagnosis in the serum of patients with CD presenting with aGMAb and were detectable in the noninflamed CD mucosa. CONCLUSIONS Anti-GMAbs predict the diagnosis of complicated CD long before the diagnosis of disease, recognize uniquely glycosylated epitopes, and impair myeloid cell and innate lymphoid cell balance associated with altered intestinal immune homeostasis.
Collapse
Affiliation(s)
- Arthur Mortha
- Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, New York; Tisch Cancer Institute, Division of Hematology/Oncology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York; Department of Immunology, University of Toronto, Toronto, Canada.
| | - Romain Remark
- Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, New York; Innate Pharma, Marseille, France
| | - Diane Marie Del Valle
- Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, New York; Tisch Cancer Institute, Division of Hematology/Oncology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Ling-Shiang Chuang
- Charles Bronfman Institute for Personalized Medicine, Department of Genetics, Icahn School of Medicine at Mount Sinai, New York, New York; Department of Genetics and Genomic Sciences, Icahn Institute for Data Science and Genomic Technology, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Zhi Chai
- Charles Bronfman Institute for Personalized Medicine, Department of Genetics, Icahn School of Medicine at Mount Sinai, New York, New York; Department of Genetics and Genomic Sciences, Icahn Institute for Data Science and Genomic Technology, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Inês Alves
- i3S - Institute for Research and Innovation in Health, University of Porto, Porto, Portugal; Faculty of Medicine, University of Porto, Porto, Portugal
| | - Catarina Azevedo
- i3S - Institute for Research and Innovation in Health, University of Porto, Porto, Portugal; Faculty of Medicine, University of Porto, Porto, Portugal
| | - Joana Gaifem
- i3S - Institute for Research and Innovation in Health, University of Porto, Porto, Portugal
| | - Jerome Martin
- Université de Nantes, Inserm, CHU Nantes, Centre de Recherche en Transplantation et Immunologie, Nantes, France; CHU Nantes, Laboratoire d'Immunologie, CIMNA, Nantes, France
| | - Francesca Petralia
- Department of Genetics and Genomic Sciences, Icahn Institute for Data Science and Genomic Technology, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Kevin Tuballes
- Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, New York; Tisch Cancer Institute, Division of Hematology/Oncology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Vanessa Barcessat
- Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, New York; Tisch Cancer Institute, Division of Hematology/Oncology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Siu Ling Tai
- Department of Immunology, University of Toronto, Toronto, Canada
| | - Hsin-Hui Huang
- Tisch Cancer Institute, Division of Hematology/Oncology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Ilaria Laface
- Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, New York; Tisch Cancer Institute, Division of Hematology/Oncology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Yeray Arteaga Jerez
- Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Gilles Boschetti
- Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, New York; Hépato-Gastroentérologue, Hospices Civils de Lyon, Université Claude Bernard, Lyon, France
| | - Nicole Villaverde
- Charles Bronfman Institute for Personalized Medicine, Department of Genetics, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Mona D Wang
- Department of Immunology, University of Toronto, Toronto, Canada
| | - Ujunwa M Korie
- Charles Bronfman Institute for Personalized Medicine, Department of Genetics, Icahn School of Medicine at Mount Sinai, New York, New York; Department of Genetics and Genomic Sciences, Icahn Institute for Data Science and Genomic Technology, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Joseph Murray
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minnesota
| | - Rok-Seon Choung
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minnesota
| | | | - Renee M Laird
- Naval Medical Research Center, Silver Spring, Maryland
| | | | - Adeeb Rahman
- Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, New York; Human Immune Monitoring Center at Mount Sinai, New York, New York
| | - Joana Torres
- Department of Gastroenterology, Icahn School of Medicine at Mount Sinai, New York, New York; Gastroenterology Division, Hospital Beatriz Ângelo, Loures, Portugal
| | - Chad Porter
- Naval Medical Research Center, Silver Spring, Maryland
| | - Mark S Riddle
- Naval Medical Research Center, Silver Spring, Maryland
| | - Ephraim Kenigsberg
- Charles Bronfman Institute for Personalized Medicine, Department of Genetics, Icahn School of Medicine at Mount Sinai, New York, New York; Human Immune Monitoring Center at Mount Sinai, New York, New York
| | - Salomé S Pinho
- i3S - Institute for Research and Innovation in Health, University of Porto, Porto, Portugal; Faculty of Medicine, University of Porto, Porto, Portugal; School of Medicine and Biomedical Sciences Abel Salazar (ICBAS), University of Porto, Porto, Portugal
| | - Judy H Cho
- Charles Bronfman Institute for Personalized Medicine, Department of Genetics, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Miriam Merad
- Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, New York; Tisch Cancer Institute, Division of Hematology/Oncology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York; Human Immune Monitoring Center at Mount Sinai, New York, New York
| | - Jean-Frederic Colombel
- Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, New York; Department of Gastroenterology, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Sacha Gnjatic
- Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, New York; Tisch Cancer Institute, Division of Hematology/Oncology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York; Human Immune Monitoring Center at Mount Sinai, New York, New York
| |
Collapse
|
12
|
McLachlan G, Alton EWFW, Boyd AC, Clarke NK, Davies JC, Gill DR, Griesenbach U, Hickmott JW, Hyde SC, Miah KM, Molina CJ. Progress in Respiratory Gene Therapy. Hum Gene Ther 2022; 33:893-912. [PMID: 36074947 PMCID: PMC7615302 DOI: 10.1089/hum.2022.172] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
The prospect of gene therapy for inherited and acquired respiratory disease has energized the research community since the 1980s, with cystic fibrosis, as a monogenic disorder, driving early efforts to develop effective strategies. The fact that there are still no approved gene therapy products for the lung, despite many early phase clinical trials, illustrates the scale of the challenge: In the 1990s, first-generation non-viral and viral vector systems demonstrated proof-of-concept but low efficacy. Since then, there has been steady progress toward improved vectors with the capacity to overcome at least some of the formidable barriers presented by the lung. In addition, the inclusion of features such as codon optimization and promoters providing long-term expression have improved the expression characteristics of therapeutic transgenes. Early approaches were based on gene addition, where a new DNA copy of a gene is introduced to complement a genetic mutation: however, the advent of RNA-based products that can directly express a therapeutic protein or manipulate gene expression, together with the expanding range of tools for gene editing, has stimulated the development of alternative approaches. This review discusses the range of vector systems being evaluated for lung delivery; the variety of cargoes they deliver, including DNA, antisense oligonucleotides, messenger RNA (mRNA), small interfering RNA (siRNA), and peptide nucleic acids; and exemplifies progress in selected respiratory disease indications.
Collapse
Affiliation(s)
- Gerry McLachlan
- The Roslin Institute & R(D)SVS, University of Edinburgh, Edinburgh, United Kingdom
- UK Respiratory Gene Therapy Consortium, London, United Kingdom
| | - Eric W F W Alton
- UK Respiratory Gene Therapy Consortium, London, United Kingdom
- Gene Therapy Group, National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - A Christopher Boyd
- UK Respiratory Gene Therapy Consortium, London, United Kingdom
- Centre for Genomic and Experimental Medicine, IGMM, University of Edinburgh, Edinburgh, United Kingdom
| | - Nora K Clarke
- UK Respiratory Gene Therapy Consortium, London, United Kingdom
- Gene Therapy Group, National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - Jane C Davies
- UK Respiratory Gene Therapy Consortium, London, United Kingdom
- Gene Therapy Group, National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - Deborah R Gill
- UK Respiratory Gene Therapy Consortium, London, United Kingdom
- Gene Medicine Group, Radcliffe Department of Medicine (NDCLS), University of Oxford, Oxford, United Kingdom
| | - Uta Griesenbach
- UK Respiratory Gene Therapy Consortium, London, United Kingdom
- Gene Therapy Group, National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - Jack W Hickmott
- UK Respiratory Gene Therapy Consortium, London, United Kingdom
- Gene Therapy Group, National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - Stephen C Hyde
- UK Respiratory Gene Therapy Consortium, London, United Kingdom
- Gene Medicine Group, Radcliffe Department of Medicine (NDCLS), University of Oxford, Oxford, United Kingdom
| | - Kamran M Miah
- UK Respiratory Gene Therapy Consortium, London, United Kingdom
- Gene Medicine Group, Radcliffe Department of Medicine (NDCLS), University of Oxford, Oxford, United Kingdom
| | - Claudia Juarez Molina
- UK Respiratory Gene Therapy Consortium, London, United Kingdom
- Gene Therapy Group, National Heart and Lung Institute, Imperial College London, London, United Kingdom
| |
Collapse
|
13
|
Wang SY, Lo YF, Shih HP, Ho MW, Yeh CF, Peng JJ, Ting HT, Lin KH, Huang WC, Chen YC, Chiu YH, Hsu CW, Tseng YT, Wang LS, Lei WY, Lin CY, Aoh Y, Chou CH, Wu TY, Ding JY, Lo CC, Lin YN, Tu KH, Lei WT, Kuo CY, Chi CY, Ku CL. Cryptococcus gattii Infection as the Major Clinical Manifestation in Patients with Autoantibodies Against Granulocyte-Macrophage Colony-Stimulating Factor. J Clin Immunol 2022; 42:1730-1741. [PMID: 35947322 DOI: 10.1007/s10875-022-01341-2] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2022] [Accepted: 07/20/2022] [Indexed: 11/24/2022]
Abstract
PURPOSE Anti-granulocyte-macrophage colony-stimulating factor autoantibodies (anti-GM-CSF Abs) are a predisposing factor for pulmonary alveolar proteinosis (PAP) and Cryptococcus gattii cryptococcosis. This study aimed to investigate clinical manifestations in anti-GM-CSF Ab-positive patients with C. gattii cryptococcosis and analyze the properties of anti-GM-CSF Abs derived from these patients and patients with PAP. METHODS Thirty-nine patients diagnosed with cryptococcosis (caused by C. neoformans or C. gattii) and 6 with PAP were enrolled in the present study. Clinical information was obtained from medical records. Blood samples were collected for analysis of autoantibody properties. We also explored the National Health Insurance Research Database (NHIRD) of Taiwan to investigate the epidemiology of cryptococcosis and PAP. RESULTS High titers of neutralizing anti-GM-CSF Abs were identified in 15 patients with cryptococcosis (15/39, 38.5%). Most anti-GM-CSF Ab-positive cryptococcosis cases had central nervous system (CNS) involvement (14/15, 93.3%). Eleven out of 14 (78.6%) anti-GM-CSF Ab-positive CNS cryptococcosis patients were confirmed to be infected with C. gattii, and PAP did not occur synchronously or metachronously in a single patient from our cohort. Exploration of an association between HLA and anti-GM-CSF Ab positivity or differential properties of autoantibodies from cryptococcosis patients and PAP yielded no significant results. CONCLUSION Anti-GM-CSF Abs can cause two diseases, C. gattii cryptococcosis and PAP, which seldom occur in the same subject. Current biological evidence regarding the properties of anti-GM-CSF Abs cannot provide clues regarding decisive mechanisms. Further analysis, including more extensive cohort studies and investigations into detailed properties, is mandatory to better understand the pathogenesis of anti-GM-CSF Abs.
Collapse
Affiliation(s)
- Shang-Yu Wang
- Laboratory of Human Immunology and Infectious Diseases, Graduate Institute of Clinical Medical Sciences, Chang Gung University, Taoyuan, Taiwan.,Division of General Surgery, Department of Surgery, Chang Gung Memorial Hospital, Taoyuan, Taiwan
| | - Yu-Fang Lo
- Laboratory of Human Immunology and Infectious Diseases, Graduate Institute of Clinical Medical Sciences, Chang Gung University, Taoyuan, Taiwan
| | - Han-Po Shih
- Laboratory of Human Immunology and Infectious Diseases, Graduate Institute of Clinical Medical Sciences, Chang Gung University, Taoyuan, Taiwan
| | - Mao-Wang Ho
- Division of Infectious Diseases, Department of Internal Medicine, China Medical University Hospital, Taichung, Taiwan
| | - Chun-Fu Yeh
- Laboratory of Human Immunology and Infectious Diseases, Graduate Institute of Clinical Medical Sciences, Chang Gung University, Taoyuan, Taiwan.,Division of Infectious Diseases, Department of Internal Medicine, Chang Gung Memorial Hospital, Taoyuan, Taiwan
| | - Jhan-Jie Peng
- Laboratory of Human Immunology and Infectious Diseases, Graduate Institute of Clinical Medical Sciences, Chang Gung University, Taoyuan, Taiwan
| | - He-Ting Ting
- Laboratory of Human Immunology and Infectious Diseases, Graduate Institute of Clinical Medical Sciences, Chang Gung University, Taoyuan, Taiwan
| | - Kuo-Hsi Lin
- Division of Infectious Diseases, Department of Internal Medicine, Tungs' Taichung MetroHarbor Hospital, Taichung, Taiwan
| | - Wen-Chi Huang
- Division of Infectious Diseases, Department of Internal Medicine, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung, Taiwan
| | - Yi-Chun Chen
- Division of Infectious Diseases, Department of Internal Medicine, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung, Taiwan
| | - Yu-Hsin Chiu
- Division of Infectious Diseases, Department of Internal Medicine, Chi Mei Medical Center, Liouying, Tainan, Taiwan
| | - Chien-Wei Hsu
- Department of Chest Medicine, Kaohsiung Veterans General Hospital, Kaohsiung, Taiwan.,School of Medicine, National Yang-Ming University, Taipei, Taiwan
| | - Yu-Ting Tseng
- Section of Infectious Diseases, Department of Medicine, Kaohsiung Veterans General Hospital, Kaohsiung, Taiwan
| | - Lih-Shinn Wang
- Division of Infectious Disease, Department of Internal Medicine, Buddhist Tzu Chi General Hospital and Tzu Chi University, Hualien, Taiwan
| | - Wei-Yi Lei
- Department of Medicine, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation and Tzu Chi University, Hualien, Taiwan
| | - Chen-Yuan Lin
- Department of Hematology and Oncology, China Medical University Hospital, Taichung, Taiwan.,School of Pharmacy, China Medical University, Taichung, Taiwan
| | - Yu Aoh
- Neuroscience Laboratory, Department of Neurology, China Medical University Hospital, Taichung, Taiwan
| | - Chia-Huei Chou
- Division of Infectious Diseases, Department of Internal Medicine, China Medical University Hospital, Taichung, Taiwan
| | - Tsai-Yi Wu
- Laboratory of Human Immunology and Infectious Diseases, Graduate Institute of Clinical Medical Sciences, Chang Gung University, Taoyuan, Taiwan
| | - Jing-Ya Ding
- Laboratory of Human Immunology and Infectious Diseases, Graduate Institute of Clinical Medical Sciences, Chang Gung University, Taoyuan, Taiwan
| | - Chia-Chi Lo
- Laboratory of Human Immunology and Infectious Diseases, Graduate Institute of Clinical Medical Sciences, Chang Gung University, Taoyuan, Taiwan
| | - You-Ning Lin
- Laboratory of Human Immunology and Infectious Diseases, Graduate Institute of Clinical Medical Sciences, Chang Gung University, Taoyuan, Taiwan
| | - Kun-Hua Tu
- Laboratory of Human Immunology and Infectious Diseases, Graduate Institute of Clinical Medical Sciences, Chang Gung University, Taoyuan, Taiwan.,Kidney Research Center, Department of Nephrology, Chang Gung Memorial Hospital, Taoyuan, Taiwan
| | - Wei-Te Lei
- Laboratory of Human Immunology and Infectious Diseases, Graduate Institute of Clinical Medical Sciences, Chang Gung University, Taoyuan, Taiwan.,Section of Immunology, Rheumatology, and Allergy Department of Pediatrics, Hsinchu Mackay Memorial Hospital, Hsinchu City, Taiwan
| | - Chen-Yen Kuo
- Laboratory of Human Immunology and Infectious Diseases, Graduate Institute of Clinical Medical Sciences, Chang Gung University, Taoyuan, Taiwan.,Division of Infectious Diseases, Department of Pediatrics, Chang Gung Memorial Hospital, Taoyuan city, Taiwan
| | - Chih-Yu Chi
- Laboratory of Human Immunology and Infectious Diseases, Graduate Institute of Clinical Medical Sciences, Chang Gung University, Taoyuan, Taiwan.,Division of Infectious Diseases, Department of Internal Medicine, China Medical University Hospital, Taichung, Taiwan.,School of Medicine, College of Medicine, China Medical University, Taichung, Taiwan
| | - Cheng-Lung Ku
- Laboratory of Human Immunology and Infectious Diseases, Graduate Institute of Clinical Medical Sciences, Chang Gung University, Taoyuan, Taiwan. .,Department of Nephrology, Chang Gung Memorial Hospital, Taoyuan, Taiwan. .,Center for Clinical and Medical Immunology, Chang Gung University, Taoyuan, Taiwan.
| |
Collapse
|
14
|
Shih HP, Ding JY, Sotolongo Bellón J, Lo YF, Chung PH, Ting HT, Peng JJ, Wu TY, Lin CH, Lo CC, Lin YN, Yeh CF, Chen JB, Wu TS, Liu YM, Kuo CY, Wang SY, Tu KH, Ng CY, Lei WT, Tsai YH, Chen JH, Chuang YT, Huang JY, Rey FA, Chen HK, Chang TW, Piehler J, Chi CY, Ku CL. Pathogenic autoantibodies to IFN-γ act through the impedance of receptor assembly and Fc-mediated response. J Exp Med 2022; 219:213354. [PMID: 35833912 PMCID: PMC9287643 DOI: 10.1084/jem.20212126] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2021] [Revised: 03/21/2022] [Accepted: 06/23/2022] [Indexed: 01/16/2023] Open
Abstract
Anti-interferon (IFN)-γ autoantibodies (AIGAs) are a pathogenic factor in late-onset immunodeficiency with disseminated mycobacterial and other opportunistic infections. AIGAs block IFN-γ function, but their effects on IFN-γ signaling are unknown. Using a single-cell capture method, we isolated 19 IFN-γ-reactive monoclonal antibodies (mAbs) from patients with AIGAs. All displayed high-affinity (KD < 10-9 M) binding to IFN-γ, but only eight neutralized IFN-γ-STAT1 signaling and HLA-DR expression. Signal blockade and binding affinity were correlated and attributed to somatic hypermutations. Cross-competition assays identified three nonoverlapping binding sites (I-III) for AIGAs on IFN-γ. We found that site I mAb neutralized IFN-γ by blocking its binding to IFN-γR1. Site II and III mAbs bound the receptor-bound IFN-γ on the cell surface, abolishing IFN-γR1-IFN-γR2 heterodimerization and preventing downstream signaling. Site III mAbs mediated antibody-dependent cellular cytotoxicity, probably through antibody-IFN-γ complexes on cells. Pathogenic AIGAs underlie mycobacterial infections by the dual blockade of IFN-γ signaling and by eliminating IFN-γ-responsive cells.
Collapse
Affiliation(s)
- Han-Po Shih
- Laboratory of Human Immunology and Infectious Disease, Graduate Institute of Clinical Medical Sciences, Chang Gung University, Taoyuan, Taiwan
| | - Jing-Ya Ding
- Laboratory of Human Immunology and Infectious Disease, Graduate Institute of Clinical Medical Sciences, Chang Gung University, Taoyuan, Taiwan
| | - Junel Sotolongo Bellón
- Division of Biophysics, Department of Biology, University of Osnabruck, Osnabruck, Germany
| | - Yu-Fang Lo
- Laboratory of Human Immunology and Infectious Disease, Graduate Institute of Clinical Medical Sciences, Chang Gung University, Taoyuan, Taiwan
| | | | - He-Ting Ting
- Laboratory of Human Immunology and Infectious Disease, Graduate Institute of Clinical Medical Sciences, Chang Gung University, Taoyuan, Taiwan
| | - Jhan-Jie Peng
- Laboratory of Human Immunology and Infectious Disease, Graduate Institute of Clinical Medical Sciences, Chang Gung University, Taoyuan, Taiwan
| | - Tsai-Yi Wu
- Laboratory of Human Immunology and Infectious Disease, Graduate Institute of Clinical Medical Sciences, Chang Gung University, Taoyuan, Taiwan
| | - Chia-Hao Lin
- Laboratory of Human Immunology and Infectious Disease, Graduate Institute of Clinical Medical Sciences, Chang Gung University, Taoyuan, Taiwan
| | - Chia-Chi Lo
- Laboratory of Human Immunology and Infectious Disease, Graduate Institute of Clinical Medical Sciences, Chang Gung University, Taoyuan, Taiwan
| | - You-Ning Lin
- Laboratory of Human Immunology and Infectious Disease, Graduate Institute of Clinical Medical Sciences, Chang Gung University, Taoyuan, Taiwan
| | - Chun-Fu Yeh
- Laboratory of Human Immunology and Infectious Disease, Graduate Institute of Clinical Medical Sciences, Chang Gung University, Taoyuan, Taiwan,Division of Infectious Diseases, Department of Internal Medicine, Chang Gung Memorial Hospital, Linkou, Taiwan
| | - Jiun-Bo Chen
- Genomics Research Center, Academia Sinica, Taipei, Taiwan
| | - Ting-Shu Wu
- Division of Infectious Diseases, Department of Internal Medicine, Chang Gung Memorial Hospital, Linkou, Taiwan,Chang Gung University College of Medicine, Taoyuan, Taiwan
| | - Yuag-Meng Liu
- Division of Infectious Diseases, Department of Internal Medicine, Changhua Christian Hospital, Changhua, Taiwan
| | - Chen-Yen Kuo
- Laboratory of Human Immunology and Infectious Disease, Graduate Institute of Clinical Medical Sciences, Chang Gung University, Taoyuan, Taiwan,Division of Infectious Diseases, Department of Pediatrics, Chang Gung Memorial Hospital, Taoyuan, Taiwan
| | - Shang-Yu Wang
- Laboratory of Human Immunology and Infectious Disease, Graduate Institute of Clinical Medical Sciences, Chang Gung University, Taoyuan, Taiwan,Division of General Surgery, Department of Surgery, Chang Gung Memorial Hospital, Taoyuan, Taiwan
| | - Kun-Hua Tu
- Laboratory of Human Immunology and Infectious Disease, Graduate Institute of Clinical Medical Sciences, Chang Gung University, Taoyuan, Taiwan,Chang Gung University College of Medicine, Taoyuan, Taiwan,Kidney Research Center, Department of Nephrology, Chang Gung Memorial Hospital, Taoyuan, Taiwan
| | - Chau Yee Ng
- Laboratory of Human Immunology and Infectious Disease, Graduate Institute of Clinical Medical Sciences, Chang Gung University, Taoyuan, Taiwan,Department of Dermatology, Chang Gung Memorial Hospital, Taipei, Taiwan
| | - Wei-Te Lei
- Laboratory of Human Immunology and Infectious Disease, Graduate Institute of Clinical Medical Sciences, Chang Gung University, Taoyuan, Taiwan,Department of Pediatrics, Hsinchu MacKay Memorial Hospital, Hsinchu, Taiwan
| | - Yu-Huan Tsai
- Laboratory of Host-Microbe Interactions and Cell Dynamics, Institute of Microbiology and Immunology, College of Life Sciences, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Jou-Han Chen
- Genomics Research Center, Academia Sinica, Taipei, Taiwan
| | - Ya-Ting Chuang
- Department of Medical Research, National Taiwan University Hospital, Taipei, Taiwan
| | | | - Félix A. Rey
- Structural Virology Unit, Department of Virology, Institut Pasteur, Paris, France
| | | | - Tse-Wen Chang
- Genomics Research Center, Academia Sinica, Taipei, Taiwan
| | - Jacob Piehler
- Division of Biophysics, Department of Biology, University of Osnabruck, Osnabruck, Germany
| | - Chih-Yu Chi
- Division of Infectious Diseases, Department of Internal Medicine, China Medical University Hospital, Taichung, Taiwan,School of Medicine, College of Medicine, China Medical University, Taichung, Taiwan,Chih-Yu Chi:
| | - Cheng-Lung Ku
- Laboratory of Human Immunology and Infectious Disease, Graduate Institute of Clinical Medical Sciences, Chang Gung University, Taoyuan, Taiwan,Department of Nephrology, Chang Gung Memorial Hospital, Taoyuan, Taiwan,Center for Molecular and Clinical Immunology, Chang Gung University, Taoyuan, Taiwan,Correspondence to Cheng-Lung Ku:
| |
Collapse
|
15
|
Anti-cytokine autoantibodies and inborn errors of immunity. J Immunol Methods 2022; 508:113313. [PMID: 35817172 DOI: 10.1016/j.jim.2022.113313] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2022] [Revised: 06/06/2022] [Accepted: 06/28/2022] [Indexed: 11/20/2022]
Abstract
The past quarter of a century has witnessed an inordinate increase in our understanding of primary immunodeficiencies / inborn errors of immunity. These include a significant increase in the number of identified conditions, broadening the phenotypes of existing entities, delineation of classical inborn errors of immunity from those with a narrow phenotype, and a gradual shift from supportive to definitive care in patients afflicted with these diseases. It has also seen the discovery of conditions broadly defined as phenocopies of primary immunodeficiencies, where somatic mutations or autoantibodies mimic a recognised primary immunodeficiency's presentation in the absence of the underlying genetic basis for that disease. This article will provide a review of the anti-cytokine autoantibody-mediated phenocopies of inborn errors of immunity and discuss the therapeutic and laboratory aspects of this group of diseases.
Collapse
|
16
|
Prevel R, Guillotin V, Imbert S, Blanco P, Delhaes L, Duffau P. Central Nervous System Cryptococcosis in Patients With Sarcoidosis: Comparison With Non-sarcoidosis Patients and Review of Potential Pathophysiological Mechanisms. Front Med (Lausanne) 2022; 9:836886. [PMID: 35425769 PMCID: PMC9002233 DOI: 10.3389/fmed.2022.836886] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2022] [Accepted: 03/02/2022] [Indexed: 11/13/2022] Open
Abstract
Introduction Cryptococcus spp. infection of the central nervous system (CINS) is a devastating opportunistic infection that was historically described in patients with acquired immunodeficiency syndrome (AIDS). Cryptococcus spp. infections are also associated with sarcoidosis; the impairment of cell-mediated immunity and long-term corticosteroid therapy being evoked to explain this association. Nevertheless, this assertion is debated and the underlying pathophysiological mechanisms are still unknown. The aims of this study were (i) to describe the clinical and biological presentation, treatments, and outcomes of CINS patients with and without sarcoidosis and (ii) to review the pathophysiological evidence underlying this clinical association. Patients and Methods Every patient with positive cerebrospinal fluid (CSF) cryptococcal antigen testing, India ink preparation, and/or culture from January 2015 to December 2020 at a tertiary university hospital were included, and patients with sarcoidosis were compared with non-sarcoidosis patients. Quantitative variables are presented as mean ± SD and are compared using the Mann-Whitney Wilcoxon rank-sum test. Categorical variables are expressed as the number of patients (percentage) and compared using the χ2 or Fisher's tests. Results During the study period, 16 patients experienced CINS, of whom 5 (31%) were associated with sarcoidosis. CINS symptoms, biological, and CSF features were similar between CINS patients with and without sarcoidosis except regarding CD4 cells percentages and CD4/CD8 ratio that was higher in those with sarcoidosis (47 ± 12 vs. 22 ± 18, p = 0.02 and 2.24 ± 1.42 vs. 0.83 ± 1.10, p = 0.03, respectively). CINS patients with sarcoidosis had less often positive blood antigen testing than those without sarcoidosis (2/5 vs. 11/11, p = 0.02). CINS patients with and without sarcoidosis were treated with similar drugs, but patients with sarcoidosis had a shorter length of treatment. CD4 cell levels do not seem to explain the association between sarcoidosis and cryptococcosis. Conclusion Sarcoidosis was the most frequently associated condition with CINS in this study. CINS patients associated with sarcoidosis had overall similar clinical and biological presentation than CINS patients associated with other conditions but exhibited a lower rate of positive blood cryptococcal antigen testing and higher CD4/CD8 T cells ratio. Pathophysiological mechanisms underlying this association remain poorly understood but B-1 cell deficiency or lack of IgM could be a part of the explanation. Another plausible mechanism is the presence of anti-granulocyte-macrophage colony-stimulating factor (GM-CSF) antibodies in a subset of patients with sarcoidosis, which could impair macrophage phagocytic function. Further studies are strongly needed to better understand those mechanisms and to identify at-risk patients.
Collapse
Affiliation(s)
- Renaud Prevel
- CHU Bordeaux, Internal Medicine Department, Bordeaux, France.,Univ Bordeaux, Centre de Recherche Cardio-Thoracique de Bordeaux, Inserm UMR 1045, Bordeaux, France
| | | | - Sébastien Imbert
- Univ Bordeaux, Centre de Recherche Cardio-Thoracique de Bordeaux, Inserm UMR 1045, Bordeaux, France.,CHU Bordeaux, Mycology-Parasitology Department, CIC 1401, Bordeaux, France
| | - Patrick Blanco
- CHU Bordeaux, Immunology Department, Bordeaux, France.,Univ Bordeaux, CNRS ImmunoConcEpT UMR 5164, Bordeaux, France
| | - Laurence Delhaes
- Univ Bordeaux, Centre de Recherche Cardio-Thoracique de Bordeaux, Inserm UMR 1045, Bordeaux, France.,CHU Bordeaux, Mycology-Parasitology Department, CIC 1401, Bordeaux, France
| | - Pierre Duffau
- CHU Bordeaux, Internal Medicine Department, Bordeaux, France.,Univ Bordeaux, CNRS ImmunoConcEpT UMR 5164, Bordeaux, France
| |
Collapse
|
17
|
Tumor-reactive antibodies evolve from non-binding and autoreactive precursors. Cell 2022; 185:1208-1222.e21. [PMID: 35305314 DOI: 10.1016/j.cell.2022.02.012] [Citation(s) in RCA: 78] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2021] [Revised: 12/20/2021] [Accepted: 02/09/2022] [Indexed: 12/27/2022]
Abstract
The tumor microenvironment hosts antibody-secreting cells (ASCs) associated with a favorable prognosis in several types of cancer. Patient-derived antibodies have diagnostic and therapeutic potential; yet, it remains unclear how antibodies gain autoreactivity and target tumors. Here, we found that somatic hypermutations (SHMs) promote antibody antitumor reactivity against surface autoantigens in high-grade serous ovarian carcinoma (HGSOC). Patient-derived tumor cells were frequently coated with IgGs. Intratumoral ASCs in HGSOC were both mutated and clonally expanded and produced tumor-reactive antibodies that targeted MMP14, which is abundantly expressed on the tumor cell surface. The reversion of monoclonal antibodies to their germline configuration revealed two types of classes: one dependent on SHMs for tumor binding and a second with germline-encoded autoreactivity. Thus, tumor-reactive autoantibodies are either naturally occurring or evolve through an antigen-driven selection process. These findings highlight the origin and potential applicability of autoantibodies directed at surface antigens for tumor targeting in cancer patients.
Collapse
|
18
|
Descatoire M, Fritzen R, Rotman S, Kuntzelman G, Leber XC, Droz-Georget S, Thrasher AJ, Traggiai E, Candotti F. Critical role of WASp in germinal center tolerance through regulation of B cell apoptosis and diversification. Cell Rep 2022; 38:110474. [PMID: 35263577 DOI: 10.1016/j.celrep.2022.110474] [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: 04/07/2021] [Revised: 08/18/2021] [Accepted: 02/10/2022] [Indexed: 11/30/2022] Open
Abstract
A main feature of Wiskott-Aldrich syndrome (WAS) is increased susceptibility to autoimmunity. A key contribution of B cells to development of these complications has been demonstrated through studies of samples from affected individuals and mouse models of the disease, but the role of the WAS protein (WASp) in controlling peripheral tolerance has not been specifically explored. Here we show that B cell responses remain T cell dependent in constitutive WASp-deficient mice, whereas selective WASp deletion in germinal center B cells (GCBs) is sufficient to induce broad development of self-reactive antibodies and kidney pathology, pointing to loss of germinal center tolerance as a primary cause leading to autoimmunity. Mechanistically, we show that WASp is upregulated in GCBs and regulates apoptosis and plasma cell differentiation in the germinal center and that the somatic hypermutation-derived diversification is the basis of autoantibody development.
Collapse
Affiliation(s)
- Marc Descatoire
- Laboratory of Inherited Immune Disorders, Division of Immunology and Allergy, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland.
| | | | - Samuel Rotman
- Service of Clinical Pathology, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | | | | | - Stephanie Droz-Georget
- Laboratory of Inherited Immune Disorders, Division of Immunology and Allergy, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Adrian J Thrasher
- University College of London, Great Ormond Street Institute of Child Health, London, UK
| | | | - Fabio Candotti
- Laboratory of Inherited Immune Disorders, Division of Immunology and Allergy, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| |
Collapse
|
19
|
Fisher BA, Veenith T, Slade D, Gaskell C, Rowland M, Whitehouse T, Scriven J, Parekh D, Balasubramaniam MS, Cooke G, Morley N, Gabriel Z, Wise MP, Porter J, McShane H, Ho LP, Newsome PN, Rowe A, Sharpe R, Thickett DR, Bion J, Gates S, Richards D, Kearns P, Turner R, Libri V, Mussai F, Middleton G, Bowden S, Bangash M, Gao-Smith F, Patel J, Sapey E, Thomas M, Coles M, Watkinson P, Rahman N, Angus B, Mentzer AJ, Novak A, Feldman M, Richter A, Faustini S, Bathurst C, Van de Wiel J, Mee S, James K, Rahman B, Turner K, Hill A, Gordon A, Yap C, Matthay M, McAuley D, Hall A, Dark P, McMichael A. Namilumab or infliximab compared with standard of care in hospitalised patients with COVID-19 (CATALYST): a randomised, multicentre, multi-arm, multistage, open-label, adaptive, phase 2, proof-of-concept trial. THE LANCET. RESPIRATORY MEDICINE 2022; 10:255-266. [PMID: 34922649 PMCID: PMC8676420 DOI: 10.1016/s2213-2600(21)00460-4] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/11/2021] [Revised: 10/08/2021] [Accepted: 10/12/2021] [Indexed: 01/05/2023]
Abstract
BACKGROUND Dysregulated inflammation is associated with poor outcomes in COVID-19. We aimed to assess the efficacy of namilumab (a granulocyte-macrophage colony stimulating factor inhibitor) and infliximab (a tumour necrosis factor inhibitor) in hospitalised patients with COVID-19, to prioritise agents for phase 3 trials. METHODS In this randomised, multicentre, multi-arm, multistage, parallel-group, open-label, adaptive, phase 2, proof-of-concept trial (CATALYST), we recruited patients (aged ≥16 years) admitted to hospital with COVID-19 pneumonia and C-reactive protein (CRP) concentrations of 40 mg/L or greater, at nine hospitals in the UK. Participants were randomly assigned with equal probability to usual care or usual care plus a single intravenous dose of namilumab (150 mg) or infliximab (5 mg/kg). Randomisation was stratified by care location within the hospital (ward vs intensive care unit [ICU]). Patients and investigators were not masked to treatment allocation. The primary endpoint was improvement in inflammation, measured by CRP concentration over time, analysed using Bayesian multilevel models. This trial is now complete and is registered with ISRCTN, 40580903. FINDINGS Between June 15, 2020, and Feb 18, 2021, we screened 299 patients and 146 were enrolled and randomly assigned to usual care (n=54), namilumab (n=57), or infliximab (n=35). For the primary outcome, 45 patients in the usual care group were compared with 52 in the namilumab group, and 29 in the usual care group were compared with 28 in the infliximab group. The probabilities that the interventions were superior to usual care alone in reducing CRP concentration over time were 97% for namilumab and 15% for infliximab; the point estimates for treatment-time interactions were -0·09 (95% CI -0·19 to 0·00) for namilumab and 0·06 (-0·05 to 0·17) for infliximab. 134 adverse events occurred in 30 (55%) of 55 patients in the namilumab group compared with 145 in 29 (54%) of 54 in the usual care group. 102 adverse events occurred in 20 (69%) of 29 patients in the infliximab group compared with 112 in 17 (50%) of 34 in the usual care group. Death occurred in six (11%) patients in the namilumab group compared with ten (19%) in the usual care group, and in four (14%) in the infliximab group compared with five (15%) in the usual care group. INTERPRETATION Namilumab, but not infliximab, showed proof-of-concept evidence for reduction in inflammation-as measured by CRP concentration-in hospitalised patients with COVID-19 pneumonia. Namilumab should be prioritised for further investigation in COVID-19. FUNDING Medical Research Council.
Collapse
Affiliation(s)
- Benjamin A Fisher
- Rheumatology Research Group, Institute of Inflammation and Ageing, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK,Cancer Research UK Clinical Trials Unit, Institute of Cancer and Genomic Sciences, University of Birmingham, Birmingham, UK,National Institute for Health Research (NIHR) Birmingham Biomedical Research Centre, University Hospitals Birmingham NHS Foundation Trust, Birmingham, UK,Correspondence to: Dr Benjamin A Fisher, Rheumatology Research Group, Institute of Inflammation and Ageing, College of Medical and Dental Sciences, University of Birmingham, Birmingham B15 2TT, UK
| | - Tonny Veenith
- Birmingham Acute Care Research Group, Institute of Inflammation and Ageing, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK,Department of Critical Care Medicine, University Hospitals Birmingham NHS Foundation Trust, Birmingham, UK
| | - Daniel Slade
- Cancer Research UK Clinical Trials Unit, Institute of Cancer and Genomic Sciences, University of Birmingham, Birmingham, UK
| | - Charlotte Gaskell
- Cancer Research UK Clinical Trials Unit, Institute of Cancer and Genomic Sciences, University of Birmingham, Birmingham, UK
| | - Matthew Rowland
- Kadoorie Centre for Critical Care Research, Nuffield Department of Clinical Neurosciences, John Radcliffe Hospital, University of Oxford, Oxford, UK
| | - Tony Whitehouse
- Birmingham Acute Care Research Group, Institute of Inflammation and Ageing, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK,Department of Critical Care Medicine, University Hospitals Birmingham NHS Foundation Trust, Birmingham, UK
| | - James Scriven
- Institute of Microbiology and Infection, University of Birmingham, Birmingham, UK,Department of Infectious Diseases, University Hospitals Birmingham NHS Foundation Trust, Birmingham, UK
| | - Dhruv Parekh
- Birmingham Acute Care Research Group, Institute of Inflammation and Ageing, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK,Department of Critical Care Medicine, University Hospitals Birmingham NHS Foundation Trust, Birmingham, UK,Department of Respiratory Medicine, University Hospitals Birmingham NHS Foundation Trust, Birmingham, UK
| | | | - Graham Cooke
- Department of Infectious Disease, Imperial College London, London, UK
| | - Nick Morley
- Department of Haematology, Royal Hallamshire Hospital, Sheffield, UK
| | - Zoe Gabriel
- Sheffield Teaching Hospitals NHS Foundation Trust, Sheffield, UK
| | - Matthew P Wise
- Department of Critical Care Medicine, University Hospital of Wales, Cardiff, UK
| | - Joanna Porter
- Department of Respiratory Medicine, University College Hospital, London, UK
| | | | - Ling-Pei Ho
- Medical Research Council Human Immunology Unit, Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, UK,Oxford Interstitial Lung Disease Service, Oxford University Hospitals NHS Foundation Trust, Oxford, UK
| | - Philip N Newsome
- Centre for Liver and Gastrointestinal Research, Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, UK,National Institute for Health Research (NIHR) Birmingham Biomedical Research Centre, University Hospitals Birmingham NHS Foundation Trust, Birmingham, UK
| | - Anna Rowe
- Cancer Research UK Clinical Trials Unit, Institute of Cancer and Genomic Sciences, University of Birmingham, Birmingham, UK,National Institute for Health Research (NIHR) Birmingham Biomedical Research Centre, University Hospitals Birmingham NHS Foundation Trust, Birmingham, UK
| | - Rowena Sharpe
- Cancer Research UK Clinical Trials Unit, Institute of Cancer and Genomic Sciences, University of Birmingham, Birmingham, UK
| | - David R Thickett
- Birmingham Acute Care Research Group, Institute of Inflammation and Ageing, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK,Department of Respiratory Medicine, University Hospitals Birmingham NHS Foundation Trust, Birmingham, UK
| | - Julian Bion
- Birmingham Acute Care Research Group, Institute of Inflammation and Ageing, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK,Department of Critical Care Medicine, University Hospitals Birmingham NHS Foundation Trust, Birmingham, UK
| | - Simon Gates
- Cancer Research UK Clinical Trials Unit, Institute of Cancer and Genomic Sciences, University of Birmingham, Birmingham, UK
| | - Duncan Richards
- Oxford Clinical Trials Research Unit, Botnar Research Centre, University of Oxford, Oxford, UK
| | - Pamela Kearns
- Cancer Research UK Clinical Trials Unit, Institute of Cancer and Genomic Sciences, University of Birmingham, Birmingham, UK,National Institute for Health Research (NIHR) Birmingham Biomedical Research Centre, University Hospitals Birmingham NHS Foundation Trust, Birmingham, UK
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
20
|
An L, Michaeli J, Pallavi P, Breedijk A, Xu X, Dietrich N, Sigl M, Keese M, Nitschke K, Jarczyk J, Nuhn P, Krämer BK, Yard BA, Leipe J. Concurrent stimulation of monocytes with CSF1 and polarizing cytokines reveals phenotypic and functional differences with classical polarized macrophages. J Leukoc Biol 2022; 112:437-447. [DOI: 10.1002/jlb.3a0721-383r] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022] Open
Affiliation(s)
- Liying An
- 5th Medical Department, University Hospital Mannheim Heidelberg University Mannheim Germany
| | - Julia Michaeli
- 5th Medical Department, University Hospital Mannheim Heidelberg University Mannheim Germany
| | - Prama Pallavi
- Department of Surgery, University Hospital Mannheim Heidelberg University Mannheim Germany
- European Center for Angioscience University Hospital Mannheim, Heideleberg University Mannheim Germany
| | - Annette Breedijk
- 5th Medical Department, University Hospital Mannheim Heidelberg University Mannheim Germany
| | - Xin Xu
- 5th Medical Department, University Hospital Mannheim Heidelberg University Mannheim Germany
| | - Nadine Dietrich
- 5th Medical Department, University Hospital Mannheim Heidelberg University Mannheim Germany
| | - Martin Sigl
- 1st Medical Department, Angiology, University Hospital Mannheim Heidelberg University Mannheim Germany
| | - Michael Keese
- Department of Surgery, University Hospital Mannheim Heidelberg University Mannheim Germany
- European Center for Angioscience University Hospital Mannheim, Heideleberg University Mannheim Germany
| | - Katja Nitschke
- Department of Urology, University Hospital Mannheim Heidelberg University Mannheim Germany
| | - Jonas Jarczyk
- Department of Urology, University Hospital Mannheim Heidelberg University Mannheim Germany
| | - Philipp Nuhn
- Department of Urology, University Hospital Mannheim Heidelberg University Mannheim Germany
| | - Bernhard K. Krämer
- 5th Medical Department, University Hospital Mannheim Heidelberg University Mannheim Germany
- European Center for Angioscience University Hospital Mannheim, Heideleberg University Mannheim Germany
- Center for Innate Immunoscience Mannheim Heidelberg University Mannheim Germany
| | - Benito A. Yard
- 5th Medical Department, University Hospital Mannheim Heidelberg University Mannheim Germany
- European Center for Angioscience University Hospital Mannheim, Heideleberg University Mannheim Germany
- Center for Innate Immunoscience Mannheim Heidelberg University Mannheim Germany
| | - Jan Leipe
- 5th Medical Department, University Hospital Mannheim Heidelberg University Mannheim Germany
- Center for Innate Immunoscience Mannheim Heidelberg University Mannheim Germany
| |
Collapse
|
21
|
Chen LF, Yang CD, Cheng XB. Anti-Interferon Autoantibodies in Adult-Onset Immunodeficiency Syndrome and Severe COVID-19 Infection. Front Immunol 2022; 12:788368. [PMID: 35003106 PMCID: PMC8727472 DOI: 10.3389/fimmu.2021.788368] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2021] [Accepted: 11/22/2021] [Indexed: 01/08/2023] Open
Abstract
Adult-onset immunodeficiency syndrome due to anti-interferon (IFN)-γ autoantibodies has attracted much attention in recent years. It usually occurs in previously healthy people and usually presents as chronic, recurrent, and hard-to-control infections that can be effectively treated with aggressive antibiotic therapy. Adult-onset immunodeficiency syndrome is also referred to as AIDS-like syndrome. Anti-type I IFN (IFN-I) autoantibodies have been reported to play a significant role in the pathogenesis of coronavirus disease 2019 (COVID-19) and preexisting anti-IFN-I autoantibodies are associated with an increased risk of severe COVID-19. This review summarizes the effects of anti-IFN autoantibodies on the susceptibility and severity of various infectious diseases, including SARS-CoV-2 infection. In addition, we discuss the role of anti-IFN autoantibodies in the pathogenesis of autoimmune diseases that are characterized by recurrent infections.
Collapse
Affiliation(s)
- Long-Fang Chen
- Department of Rheumatology and Immunology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Cheng-De Yang
- Department of Rheumatology and Immunology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiao-Bing Cheng
- Department of Rheumatology and Immunology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
22
|
Knight V. Immunodeficiency and Autoantibodies to Cytokines. J Appl Lab Med 2022; 7:151-164. [PMID: 34996092 DOI: 10.1093/jalm/jfab139] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2021] [Accepted: 10/11/2021] [Indexed: 11/13/2022]
Abstract
BACKGROUND Anti-cytokine autoantibodies (AAbs) associated with an infectious phenotype are now included along with anti-complement AAbs and somatic pathogenic gene variants as a distinct category termed 'phenocopies of primary immunodeficiencies' in the classification of inborn errors of immunity. Anti-cytokine AAbs target specific cytokine pathways, leading to inordinate susceptibility to specific organisms, generally in the setting of immunocompetence. CONTENT Anti-cytokine AAbs are detected in the majority of healthy individuals and may play a regulatory role in limiting exaggerated responses to cytokines. While it is not well understood why some individuals with anti-cytokine AAbs develop increased susceptibility to organisms of low pathogenicity and others do not, it is likely that genetics and environment play a role. To date, AAbs to interferon gamma (IFNγ), interferon alpha (IFNα), interleukins-17 and 22 (IL-17/IL-22), interleukin-6 and granulocyte macrophage colony stimulating factor (GM-CSF) and their association with increased susceptibility to nontuberculous mycobacteria and other intracellular organisms, viral infections, Candida albicans, Staphylococcus aureus and other pyogenic organisms, and fungal infections respectively, have been described. The clinical phenotype of these patients is very similar to that of individuals with pathogenic gene variants in the specific cytokine pathway that the autoantibody targets, hence the term 'phenocopy.' Recognition of anti-cytokine AAbs as a distinct cause of immunodeficiency or immune dysregulation is important for appropriate management of such patients. SUMMARY Understanding the roles that anti-cytokine AAbs play in health and disease continues to be a fascinating area of research. Evaluating generally immunocompetent individuals who present with chronic, treatment refractory, or unusual infections for anti-cytokine AAbs is critical as it may direct therapy and disease management.
Collapse
Affiliation(s)
- Vijaya Knight
- Department of Pediatrics, University of Colorado School of Medicine, Aurora, CO, USA.,Children's Hospital, Colorado, Aurora, CO, USA
| |
Collapse
|
23
|
Bai JW, Gu SY, Sun XL, Lu HW, Liang S, Xu JF. CYFRA21-1 is a more sensitive biomarker to assess the severity of pulmonary alveolar proteinosis. BMC Pulm Med 2022; 22:2. [PMID: 34980056 PMCID: PMC8725332 DOI: 10.1186/s12890-021-01795-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2021] [Accepted: 12/14/2021] [Indexed: 12/01/2022] Open
Abstract
Background Serum lactate dehydrogenase (LDH), carcinoembryonic antigen (CEA) and CYFRA21-1 are the commonly used biomarkers to identify patients with autoimmune pulmonary alveolar proteinosis (APAP). However, it is not clear which of the biomarkers is more sensitive to the severity of the patient’s condition. Methods APAP patients numbering 151 were enrolled in this study. All patients’ severity was assessed through the severity and prognosis score of PAP (SPSP). According to the respective laboratory upper limits of serum levels of LDH, CEA and CYFRA21-1, APAP patients were divided into higher and lower-level groups. Patients were divided into five groups based on SPSP. 88 patients had completed six months of follow-up. We calculated sensitivity, specificity, and critical point of LDH, CEA and CYFRA21-1 between APAP patients and normal control group, and between grade 1–2 and 3–5 through receiving operating characteristics (ROC) curve. Results Serum LDH, CEA and CYFRA21-1 levels of patients with PAP were higher and distinctly related to PaO2, FVC, FEV1, DLCO, HRCT scores and SPSP. The SPSP of patients in higher-level LDH, CEA and CYFRA21-1 groups were higher than those of corresponding lower-level groups. Based on SPSP results, the patients were divided into five groups (grade I, 20; grade II, 37; grade III, 40; grade IV, 38; grade V, 16). The serum level of CYFRA21-1 of patients with APAP in grade II was higher than that of patients in grade I and lower than that of patients in grade III. Serum CYFRA21-1 of patients with APAP after six months were higher than the baseline among the aggravated group. Serum LDH, CEA and CYFRA21-1 levels after six months among patients in the relieved group of patients with APAP were lower than the baseline. ROC correlating LDH, CEA and CYFRA21-1 values with APAP severity (between grade 1–2 and 3–5) showed an optimal cutoff of LDH of over 203 U/L (< 246 U/L), CEA of over 2.56 ug/L (< 10 ug/L), and CYFRA21-1 of over 5.57 ng/ml (> 3.3 ng/ml) (AUC: 0.815, 95% CI [0.748–0.882], sensitivity: 0.606, specificity: 0.877). Conclusion Serum CYFRA21-1 level was more sensitive in revealing the severity of APAP than LDH and CEA levels among mild to moderate forms of disease.
Collapse
Affiliation(s)
- Jiu-Wu Bai
- Department of Respiratory and Critical Care Medicine, Shanghai Pulmonary Hospital, Tongji University School of Medicine, No. 507 Zhengmin Road, Shanghai, 200433, China
| | - Shui-Yi Gu
- Department of Respiratory and Critical Care Medicine, Shanghai Pulmonary Hospital, Tongji University School of Medicine, No. 507 Zhengmin Road, Shanghai, 200433, China
| | - Xiao-Li Sun
- Department of Respiratory and Critical Care Medicine, Shanghai Pulmonary Hospital, Tongji University School of Medicine, No. 507 Zhengmin Road, Shanghai, 200433, China
| | - Hai-Wen Lu
- Department of Respiratory and Critical Care Medicine, Shanghai Pulmonary Hospital, Tongji University School of Medicine, No. 507 Zhengmin Road, Shanghai, 200433, China
| | - Shuo Liang
- Department of Respiratory and Critical Care Medicine, Shanghai Pulmonary Hospital, Tongji University School of Medicine, No. 507 Zhengmin Road, Shanghai, 200433, China
| | - Jin-Fu Xu
- Department of Respiratory and Critical Care Medicine, Shanghai Pulmonary Hospital, Tongji University School of Medicine, No. 507 Zhengmin Road, Shanghai, 200433, China.
| |
Collapse
|
24
|
Tanaka S, Ise W, Baba Y, Kurosaki T. Silencing and activating anergic B cells. Immunol Rev 2021; 307:43-52. [PMID: 34908172 DOI: 10.1111/imr.13053] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2021] [Accepted: 11/30/2021] [Indexed: 02/06/2023]
Abstract
Despite the existence of central tolerance mechanisms, including clonal deletion and receptor editing to eliminate self-reactive B cells, moderately self-reactive cells still survive in the periphery (about 20% of peripheral B cells). These cells normally exist in a functionally silenced state called anergy; thus, anergy has been thought to contribute to tolerance by active-silencing of potentially dangerous B cells. However, a positive rationale for the existence of these anergic B cells has recently been suggested by discoveries that broadly neutralizing antibodies for HIV and influenza virus possess poly- and/or auto-reactivity. Given the conundrum of generating inherent holes in the immune repertoire, retaining weakly self-reactive BCRs on anergic B cells could allow these antibodies to serve as an effective defense against pathogens, particularly in the case of pathogens that mimic forbidden self-epitopes to evade the host immune system. Thus, anergic B cells should be brought into a silenced or activated state, depending on their contexts. Here, we review recent progress in our understanding of how the anergic B cell state is controlled in B cell-intrinsic and B cell-extrinsic ways.
Collapse
Affiliation(s)
- Shinya Tanaka
- Division of Immunology and Genome Biology, Department of Molecular Genetics, Medical Institute of Bioregulation, Kyushu University, Fukuoka, Japan
| | - Wataru Ise
- Team of Host Defense, Center for Infectious Disease Education and Research, Osaka University, Osaka, Japan.,Laboratory of Lymphocyte Differentiation, WPI Immunology Frontier Research Center, Osaka University, Osaka, Japan
| | - Yoshihiro Baba
- Division of Immunology and Genome Biology, Department of Molecular Genetics, Medical Institute of Bioregulation, Kyushu University, Fukuoka, Japan
| | - Tomohiro Kurosaki
- Laboratory of Lymphocyte Differentiation, WPI Immunology Frontier Research Center, Osaka University, Osaka, Japan.,Division of Microbiology and Immunology, Center for Infectious Disease Education and Research, Osaka University, Osaka, Japan.,Laboratory of Lymphocyte Differentiation, RIKEN Center for Integrative Medical Sciences (IMS), Yokohama, Japan
| |
Collapse
|
25
|
Ataya A, Knight V, Carey BC, Lee E, Tarling EJ, Wang T. The Role of GM-CSF Autoantibodies in Infection and Autoimmune Pulmonary Alveolar Proteinosis: A Concise Review. Front Immunol 2021; 12:752856. [PMID: 34880857 PMCID: PMC8647160 DOI: 10.3389/fimmu.2021.752856] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2021] [Accepted: 11/04/2021] [Indexed: 11/13/2022] Open
Abstract
Autoantibodies to multiple cytokines have been identified and some, including antibodies against granulocyte-macrophage colony-stimulating factor (GM-CSF), have been associated with increased susceptibility to infection. High levels of GM-CSF autoantibodies that neutralize signaling cause autoimmune pulmonary alveolar proteinosis (aPAP), an ultrarare autoimmune disease characterized by accumulation of excess surfactant in the alveoli, leading to pulmonary insufficiency. Defective GM-CSF signaling leads to functional deficits in multiple cell types, including macrophages and neutrophils, with impaired phagocytosis and host immune responses against pulmonary and systemic infections. In this article, we review the role of GM-CSF in aPAP pathogenesis and pulmonary homeostasis along with the increased incidence of infections (particularly opportunistic infections). Therefore, recombinant human GM-CSF products may have potential for treatment of aPAP and possibly other infectious and pulmonary diseases due to its pleotropic immunomodulatory actions.
Collapse
Affiliation(s)
- Ali Ataya
- Division of Pulmonary, Critical Care and Sleep Medicine, University of Florida, Gainesville, FL, United States
| | - Vijaya Knight
- Department of Pediatrics, Section of Allergy and Immunology, University of Colorado School of Medicine and Children's Hospital, Aurora, CO, United States
| | - Brenna C Carey
- Translational Pulmonary Science Center, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, United States
| | - Elinor Lee
- Division of Pulmonary, Critical Care, and Sleep Medicine, David Geffen School of Medicine at University of California, Los Angeles (UCLA), Los Angeles, CA, United States
| | - Elizabeth J Tarling
- Department of Medicine, Division of Cardiology, David Geffen School of Medicine at University of California, Los Angeles (UCLA), Los Angeles, CA, United States
| | - Tisha Wang
- Division of Pulmonary, Critical Care, and Sleep Medicine, David Geffen School of Medicine at University of California, Los Angeles (UCLA), Los Angeles, CA, United States
| |
Collapse
|
26
|
Chronic pharmacological antagonism of the GM-CSF receptor in mice does not replicate the pulmonary alveolar proteinosis phenotype but does alter lung surfactant turnover. Clin Sci (Lond) 2021; 135:2559-2573. [PMID: 34778899 DOI: 10.1042/cs20210713] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2021] [Revised: 11/04/2021] [Accepted: 11/12/2021] [Indexed: 12/19/2022]
Abstract
Granulocyte macrophage colony stimulating factor (GM-CSF) is a key participant in, and a clinical target for, the treatment of inflammatory diseases including rheumatoid arthritis (RA). Therapeutic inhibition of GM-CSF signalling using monoclonal antibodies to the α-subunit of the GM-CSF receptor (GMCSFRα) has shown clear benefit in patients with RA, giant cell arteritis (GCAs) and some efficacy in severe SARS-CoV-2 infection. However, GM-CSF autoantibodies are associated with the development of pulmonary alveolar proteinosis (PAP), a rare lung disease characterised by alveolar macrophage (AM) dysfunction and the accumulation of surfactant lipids. We assessed how the anti-GMCSFRα approach might impact surfactant turnover in the airway. Female C57BL/6J mice received a mouse-GMCSFRα blocking antibody (CAM-3003) twice per week for up to 24 weeks. A parallel, comparator cohort of the mouse PAP model, GM-CSF receptor β subunit (GMCSFRβ) knock-out (KO), was maintained up to 16 weeks. We assessed lung tissue histopathology alongside lung phosphatidylcholine (PC) metabolism using stable isotope lipidomics. GMCSFRβ KO mice reproduced the histopathological and biochemical features of PAP, accumulating surfactant PC in both broncho-alveolar lavage fluid (BALF) and lavaged lung tissue. The incorporation pattern of methyl-D9-choline showed impaired catabolism and not enhanced synthesis. In contrast, chronic supra-pharmacological CAM-3003 exposure (100 mg/kg) over 24 weeks did not elicit a histopathological PAP phenotype despite some changes in lung PC catabolism. Lack of significant impairment of AM catabolic function supports clinical observations that therapeutic antibodies to this pathway have not been associated with PAP in clinical trials.
Collapse
|
27
|
Hong C, Lu H, Jin R, Huang X, Chen M, Dai X, Gong F, Dong H, Wang H, Gao XM. Cytokine Cocktail Promotes Alveolar Macrophage Reconstitution and Functional Maturation in a Murine Model of Haploidentical Bone Marrow Transplantation. Front Immunol 2021; 12:719727. [PMID: 34621268 PMCID: PMC8490745 DOI: 10.3389/fimmu.2021.719727] [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: 06/03/2021] [Accepted: 09/02/2021] [Indexed: 11/23/2022] Open
Abstract
Infectious pneumonia is one of the most common complications after bone marrow transplantation (BMT), which is considered to be associated with poor reconstitution and functional maturation of alveolar macrophages (AMs) post-transplantation. Here, we present evidence showing that lack of IL-13-secreting group 2 innate lymphoid cells (ILC2s) in the lungs may underlay poor AM reconstitution in a mouse model of haploidentical BMT (haplo-BMT). Recombinant murine IL-13 was able to potentiate monocyte-derived AM differentiation in vitro. When intranasally administered, a cocktail of granulocyte-macrophage colony-stimulating factor (GM-CSF), IL-13, and CCL2 not only promoted donor monocyte-derived AM reconstitution in haplo-BMT-recipient mice but also enhanced the innate immunity of the recipient animals against pulmonary bacterial infection. These results provide a useful clue for a clinical strategy to prevent pulmonary bacterial infection at the early stage of recipients post-BMT.
Collapse
Affiliation(s)
- Chao Hong
- Institutes of Biology and Medical Sciences, Soochow University, Suzhou, China
| | - Hongyun Lu
- Institutes of Biology and Medical Sciences, Soochow University, Suzhou, China
| | - Rong Jin
- Institutes of Biology and Medical Sciences, Soochow University, Suzhou, China
| | - Xiaohong Huang
- Institutes of Biology and Medical Sciences, Soochow University, Suzhou, China
| | - Ming Chen
- Institutes of Biology and Medical Sciences, Soochow University, Suzhou, China
| | - Xiaoqiu Dai
- Institutes of Biology and Medical Sciences, Soochow University, Suzhou, China
| | - Fangyuan Gong
- Institutes of Biology and Medical Sciences, Soochow University, Suzhou, China
| | - Hongliang Dong
- Institutes of Biology and Medical Sciences, Soochow University, Suzhou, China
| | - Hongmin Wang
- Institutes of Biology and Medical Sciences, Soochow University, Suzhou, China
| | - Xiao-Ming Gao
- Institutes of Biology and Medical Sciences, Soochow University, Suzhou, China
| |
Collapse
|
28
|
Reijm S, Kissel T, Stoeken-Rijsbergen G, Slot LM, Wortel CM, van Dooren HJ, Levarht NEW, Kampstra ASB, Derksen VFAM, Heer POD, Bang H, Drijfhout JW, Trouw LA, Huizinga TWJ, Rispens T, Scherer HU, Toes REM. Cross-reactivity of IgM anti-modified protein antibodies in rheumatoid arthritis despite limited mutational load. Arthritis Res Ther 2021; 23:230. [PMID: 34479638 PMCID: PMC8413699 DOI: 10.1186/s13075-021-02609-5] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2021] [Accepted: 08/16/2021] [Indexed: 12/16/2022] Open
Abstract
Background Anti-modified protein antibodies (AMPA) targeting citrullinated, acetylated and/or carbamylated self-antigens are hallmarks of rheumatoid arthritis (RA). Although AMPA-IgG cross-reactivity to multiple post-translational modifications (PTMs) is evident, it is unknown whether the first responding B cells, expressing IgM, display similar characteristics or if cross-reactivity is crucially dependent on somatic hypermutation (SHM). We now studied the reactivity of (germline) AMPA-IgM to further understand the breach of B cell tolerance and to identify if cross-reactivity depends on extensive SHM. Moreover, we investigated whether AMPA-IgM can efficiently recruit immune effector mechanisms. Methods Polyclonal AMPA-IgM were isolated from RA patients and assessed for cross-reactivity towards PTM antigens. AMPA-IgM B cell receptor sequences were obtained by single cell isolation using antigen-specific tetramers. Subsequently, pentameric monoclonal AMPA-IgM, their germline counterparts and monomeric IgG variants were generated. The antibodies were analysed on a panel of PTM antigens and tested for complement activation. Results Pentameric monoclonal and polyclonal AMPA-IgM displayed cross-reactivity to multiple antigens and different PTMs. PTM antigen recognition was still present, although reduced, after reverting the IgM into germline. Valency of AMPA-IgM was crucial for antigen recognition as PTM-reactivity significantly decreased when AMPA-IgM were expressed as IgG. Furthermore, AMPA-IgM was 15- to 30-fold more potent in complement-activation compared to AMPA-IgG. Conclusions We provide first evidence that AMPA-IgM are cross-reactive towards different PTMs, indicating that PTM (cross-)reactivity is not confined to IgG and does not necessarily depend on extensive somatic hypermutation. Moreover, our data indicate that a diverse set of PTM antigens could be involved in the initial tolerance breach in RA and suggest that AMPA-IgM can induce complement-activation and thereby inflammation. Supplementary Information The online version contains supplementary material available at 10.1186/s13075-021-02609-5.
Collapse
Affiliation(s)
- Sanne Reijm
- Department of Rheumatology, Leiden University Medical Center, Leiden, The Netherlands.
| | - Theresa Kissel
- Department of Rheumatology, Leiden University Medical Center, Leiden, The Netherlands
| | | | - Linda M Slot
- Department of Rheumatology, Leiden University Medical Center, Leiden, The Netherlands
| | - Corrie M Wortel
- Department of Rheumatology, Leiden University Medical Center, Leiden, The Netherlands
| | - Hugo J van Dooren
- Department of Rheumatology, Leiden University Medical Center, Leiden, The Netherlands
| | - Nivine E W Levarht
- Department of Rheumatology, Leiden University Medical Center, Leiden, The Netherlands
| | - Arieke S B Kampstra
- Department of Rheumatology, Leiden University Medical Center, Leiden, The Netherlands
| | - Veerle F A M Derksen
- Department of Rheumatology, Leiden University Medical Center, Leiden, The Netherlands
| | - Pleuni Ooijevaar-de Heer
- Sanquin Research and Landsteiner Laboratory, Academic Medical Center, Amsterdam, The Netherlands
| | | | - Jan W Drijfhout
- Department of Immunology, Leiden University Medical Center, Leiden, The Netherlands
| | - Leendert A Trouw
- Department of Immunology, Leiden University Medical Center, Leiden, The Netherlands
| | - Tom W J Huizinga
- Department of Rheumatology, Leiden University Medical Center, Leiden, The Netherlands
| | - Theo Rispens
- Sanquin Research and Landsteiner Laboratory, Academic Medical Center, Amsterdam, The Netherlands
| | - Hans U Scherer
- Department of Rheumatology, Leiden University Medical Center, Leiden, The Netherlands
| | - René E M Toes
- Department of Rheumatology, Leiden University Medical Center, Leiden, The Netherlands
| |
Collapse
|
29
|
Fichtner ML, Vieni C, Redler RL, Kolich L, Jiang R, Takata K, Stathopoulos P, Suarez PA, Nowak RJ, Burden SJ, Ekiert DC, O'Connor KC. Affinity maturation is required for pathogenic monovalent IgG4 autoantibody development in myasthenia gravis. J Exp Med 2021; 217:152036. [PMID: 32820331 PMCID: PMC7953735 DOI: 10.1084/jem.20200513] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2020] [Revised: 06/04/2020] [Accepted: 07/16/2020] [Indexed: 12/24/2022] Open
Abstract
Pathogenic muscle-specific tyrosine kinase (MuSK)–specific IgG4 autoantibodies in autoimmune myasthenia gravis (MG) are functionally monovalent as a result of Fab-arm exchange. The development of these unique autoantibodies is not well understood. We examined MG patient–derived monoclonal autoantibodies (mAbs), their corresponding germline-encoded unmutated common ancestors (UCAs), and monovalent antigen-binding fragments (Fabs) to investigate how affinity maturation contributes to binding and immunopathology. Mature mAbs, UCA mAbs, and mature monovalent Fabs bound to MuSK and demonstrated pathogenic capacity. However, monovalent UCA Fabs bound to MuSK but did not have measurable pathogenic capacity. Affinity of the UCA Fabs for MuSK was 100-fold lower than the subnanomolar affinity of the mature Fabs. Crystal structures of two Fabs revealed how mutations acquired during affinity maturation may contribute to increased MuSK-binding affinity. These findings indicate that the autoantigen drives autoimmunity in MuSK MG through the accumulation of somatic mutations such that monovalent IgG4 Fab-arm–exchanged autoantibodies reach a high-affinity threshold required for pathogenic capacity.
Collapse
Affiliation(s)
- Miriam L Fichtner
- Department of Neurology, Yale University School of Medicine, New Haven, CT.,Department of Immunobiology, Yale University School of Medicine, New Haven, CT
| | - Casey Vieni
- Departments of Cell Biology and Microbiology, New York University School of Medicine, New York, NY.,Medical Scientist Training Program, New York University School of Medicine, New York, NY
| | - Rachel L Redler
- Departments of Cell Biology and Microbiology, New York University School of Medicine, New York, NY
| | - Ljuvica Kolich
- Departments of Cell Biology and Microbiology, New York University School of Medicine, New York, NY
| | - Ruoyi Jiang
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT
| | - Kazushiro Takata
- Department of Neurology, Yale University School of Medicine, New Haven, CT.,Department of Immunobiology, Yale University School of Medicine, New Haven, CT
| | - Panos Stathopoulos
- Department of Neurology, Yale University School of Medicine, New Haven, CT.,Department of Immunobiology, Yale University School of Medicine, New Haven, CT
| | - Pablo A Suarez
- Department of Neurology, Yale University School of Medicine, New Haven, CT.,Department of Immunobiology, Yale University School of Medicine, New Haven, CT
| | - Richard J Nowak
- Department of Neurology, Yale University School of Medicine, New Haven, CT
| | - Steven J Burden
- Departments of Cell Biology and Microbiology, New York University School of Medicine, New York, NY
| | - Damian C Ekiert
- Departments of Cell Biology and Microbiology, New York University School of Medicine, New York, NY
| | - Kevin C O'Connor
- Department of Neurology, Yale University School of Medicine, New Haven, CT.,Department of Immunobiology, Yale University School of Medicine, New Haven, CT
| |
Collapse
|
30
|
Berthoux C, Mailhe M, Vély F, Gauthier C, Mège JL, Lagier JC, Melenotte C. Granulocyte Macrophage Colony-Stimulating Factor-Specific Autoantibodies and Cerebral Nocardia With Pulmonary Alveolar Proteinosis. Open Forum Infect Dis 2020; 8:ofaa612. [PMID: 33614812 PMCID: PMC7881751 DOI: 10.1093/ofid/ofaa612] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2020] [Accepted: 12/10/2020] [Indexed: 11/13/2022] Open
Abstract
In this study, we report the history of a 40-year-old man with a primary cerebral abscess caused by Nocardia abscessus that led to the discovery of autoimmune pulmonary alveolar lipoproteinosis (anti-granulocyte-macrophage colony-stimulating factor [GM-CSF] autoantibodies). Anti-GM-CSF autoantibodies promote immunodeficiency and should be monitored to prevent opportunistic and disseminated infections and to diagnose asymptomatic pulmonary alveolar lipoproteinosis.
Collapse
Affiliation(s)
- Charlotte Berthoux
- Aix-Marseille Université, IRD Institut de Recherche et de Developpement, APHM Assistance publique des Hôpitaux de Marseille, MEPHI Microbes, Evolution, Phylogénie et Infection, Marseille, France.,IHU-Méditerranée Infection, Marseille, France
| | - Morgane Mailhe
- Aix-Marseille Université, IRD Institut de Recherche et de Developpement, APHM Assistance publique des Hôpitaux de Marseille, MEPHI Microbes, Evolution, Phylogénie et Infection, Marseille, France.,IHU-Méditerranée Infection, Marseille, France
| | - Frédéric Vély
- Assistance Publique des Hôpitaux de Marseille, Hôpitaux Conception et Timone, Service d'Immunologie, Marseille, France.,Aix-Marseille Université, CNRS, INSERM, CIML, Marseille, France
| | - Clarisse Gauthier
- Assistance Publique des Hôpitaux de Marseille, Hôpital Nord, Service de Pneumologie et Maladies Respiratoires Rares, Marseille, France
| | - Jean-Louis Mège
- Assistance Publique des Hôpitaux de Marseille, Hôpitaux Conception et Timone, Service d'Immunologie, Marseille, France
| | - Jean-Christophe Lagier
- Aix-Marseille Université, IRD Institut de Recherche et de Developpement, APHM Assistance publique des Hôpitaux de Marseille, MEPHI Microbes, Evolution, Phylogénie et Infection, Marseille, France.,IHU-Méditerranée Infection, Marseille, France
| | - Cléa Melenotte
- Aix-Marseille Université, IRD Institut de Recherche et de Developpement, APHM Assistance publique des Hôpitaux de Marseille, MEPHI Microbes, Evolution, Phylogénie et Infection, Marseille, France.,IHU-Méditerranée Infection, Marseille, France
| |
Collapse
|
31
|
Reijm S, Kissel T, Toes R. Checkpoints controlling the induction of B cell mediated autoimmunity in human autoimmune diseases. Eur J Immunol 2020; 50:1885-1894. [DOI: 10.1002/eji.202048820] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2020] [Accepted: 11/04/2020] [Indexed: 12/24/2022]
Affiliation(s)
- S. Reijm
- Department of Rheumatology Leiden University Medical Center Leiden The Netherlands
| | - T. Kissel
- Department of Rheumatology Leiden University Medical Center Leiden The Netherlands
| | - R.E.M. Toes
- Department of Rheumatology Leiden University Medical Center Leiden The Netherlands
| |
Collapse
|
32
|
Hamilton JA. GM-CSF in inflammation. J Exp Med 2020; 217:jem.20190945. [PMID: 31611249 PMCID: PMC7037240 DOI: 10.1084/jem.20190945] [Citation(s) in RCA: 163] [Impact Index Per Article: 32.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2019] [Revised: 07/09/2019] [Accepted: 09/11/2019] [Indexed: 02/06/2023] Open
Abstract
GM-CSF is a potential therapeutic target in inflammation and autoimmunity. This study reviews the literature on the biology of GM-CSF, in particular that describing the research leading to clinical trials targeting GM-CSF and its receptor in numerous inflammatory/autoimmune conditions, such as rheumatoid arthritis. Granulocyte–macrophage colony-stimulating factor (GM-CSF) has many more functions than its original in vitro identification as an inducer of granulocyte and macrophage development from progenitor cells. Key features of GM-CSF biology need to be defined better, such as the responding and producing cell types, its links with other mediators, its prosurvival versus activation/differentiation functions, and when it is relevant in pathology. Significant preclinical data have emerged from GM-CSF deletion/depletion approaches indicating that GM-CSF is a potential target in many inflammatory/autoimmune conditions. Clinical trials targeting GM-CSF or its receptor have shown encouraging efficacy and safety profiles, particularly in rheumatoid arthritis. This review provides an update on the above topics and current issues/questions surrounding GM-CSF biology.
Collapse
Affiliation(s)
- John A Hamilton
- The University of Melbourne, Department of Medicine, Royal Melbourne Hospital, Parkville, Victoria, Australia.,Australian Institute for Musculoskeletal Science, The University of Melbourne and Western Health, St Albans, Victoria, Australia
| |
Collapse
|
33
|
Abstract
Pulmonary alveolar proteinosis (PAP) is a rare respiratory syndrome characterised by the accumulation of surfactant lipoproteins within the alveoli. According to various pathogenetic mechanisms and aetiologies, PAP is classified as primary, secondary or congenital. Primary PAP is led by a granulocyte–macrophage colony-stimulating factor (GM-CSF) signalling disruption; the autoimmune form is driven by the presence of anti GM-CSF autoantibodies and represents 90% of all the PAP cases; and the hereditary form is the result of mutations in genes encoding GM-CSF receptor. Secondary PAP is associated with various diseases causing a reduction in function and/or number of alveolar macrophages. Congenital PAP emerges as a consequence of corrupted surfactant production, due to mutations in surfactant proteins or lipid transporter, or mutations affecting lung development. The clinical manifestations are various, ranging from insidious onset to acute or progressive respiratory failure, including premature death within the first days of life in neonates with congenital surfactant production disorders. The diagnostic workup includes clinical and radiological assessment (respiratory function test, high-resolution chest computed tomography), laboratory tests (anti-GM-CSF autoantibodies dosage, GM-CSF serum level and GM-CSF signalling test), and genetic tests. Whole-lung lavage is the current gold standard of care of PAP; however, the therapeutic approach depends on the pathogenic form and disease severity, including GM-CSF augmentation strategies in autoimmune PAP and other promising new treatments. A concise educational review of pulmonary alveolar proteinosis (PAP), a rare respiratory syndrome with various and heterogeneous aetiologies, caused by the impairment of pulmonary surfactant clearance or by abnormal surfactant productionhttps://bit.ly/3aFpQm9
Collapse
Affiliation(s)
| | - Ilaria Campo
- Pneumology Unit, IRCCS Policlinico San Matteo Hospital Foundation, Pavia, Italy
| |
Collapse
|
34
|
Lang FM, Lee KMC, Teijaro JR, Becher B, Hamilton JA. GM-CSF-based treatments in COVID-19: reconciling opposing therapeutic approaches. Nat Rev Immunol 2020; 20:507-514. [PMID: 32576980 PMCID: PMC7309428 DOI: 10.1038/s41577-020-0357-7] [Citation(s) in RCA: 145] [Impact Index Per Article: 29.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/01/2020] [Indexed: 12/17/2022]
Abstract
Therapeutics against coronavirus disease 2019 (COVID-19) are urgently needed. Granulocyte–macrophage colony-stimulating factor (GM-CSF), a myelopoietic growth factor and pro-inflammatory cytokine, plays a critical role in alveolar macrophage homeostasis, lung inflammation and immunological disease. Both administration and inhibition of GM-CSF are currently being therapeutically tested in COVID-19 clinical trials. This Perspective discusses the pleiotropic biology of GM-CSF and the scientific merits behind these contrasting approaches. Recombinant granulocyte–macrophage colony-stimulating factor (GM-CSF) as well as antibodies targeted at GM-CSF or its receptor are being tested in clinical trials for coronavirus disease 2019 (COVID-19). This Perspective introduces the pleiotropic functions of GM-CSF and explores the rationale behind these different approaches.
Collapse
Affiliation(s)
| | - Kevin M-C Lee
- Department of Medicine, Royal Melbourne Hospital, The University of Melbourne, Parkville, Melbourne, Victoria, Australia
| | - John R Teijaro
- Department of Immunology and Microbiology, Scripps Research Institute, La Jolla, CA, USA
| | - Burkhard Becher
- Institute of Experimental Immunology, University of Zurich, Zurich, Switzerland
| | - John A Hamilton
- Department of Medicine, Royal Melbourne Hospital, The University of Melbourne, Parkville, Melbourne, Victoria, Australia. .,Australian Institute for Musculoskeletal Science, The University of Melbourne and Western Health, St Albans, Melbourne, Victoria, Australia.
| |
Collapse
|
35
|
Autoantibodies against cytokines: phenocopies of primary immunodeficiencies? Hum Genet 2020; 139:783-794. [PMID: 32419033 PMCID: PMC7272486 DOI: 10.1007/s00439-020-02180-0] [Citation(s) in RCA: 62] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2020] [Accepted: 05/05/2020] [Indexed: 01/04/2023]
Abstract
Anti-cytokine autoantibodies may cause immunodeficiency and have been recently recognized as ‘autoimmune phenocopies of primary immunodeficiencies’ and are found in particular, but not exclusively in adult patients. By blocking the cytokine’s biological function, patients with anti-cytokine autoantibodies may present with a similar clinical phenotype as the related inborn genetic disorders. So far, autoantibodies to interferon (IFN)-γ, GM-CSF, to a group of TH-17 cytokines and to IL-6 have been found to be causative or closely associated with susceptibility to infection. This review compares infectious diseases associated with anti-cytokine autoantibodies with primary immunodeficiencies affecting similar cytokines or related pathways.
Collapse
|
36
|
Hashimoto A, Takeuchi S, Kajita R, Yamagata A, Kakui R, Tanaka T, Nakata K. Proteogenomic analysis of granulocyte macrophage colony- stimulating factor autoantibodies in the blood of a patient with autoimmune pulmonary alveolar proteinosis. Sci Rep 2020; 10:4923. [PMID: 32188922 PMCID: PMC7080758 DOI: 10.1038/s41598-020-61934-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2019] [Accepted: 03/02/2020] [Indexed: 11/18/2022] Open
Abstract
Recently, attempts to reveal the structures of autoantibodies comprehensively using improved proteogenomics technology, have become popular. This technology identifies peptides in highly purified antibodies by using an Orbitrap device to compare spectra from liquid chromatography-tandem mass spectrometry against a cDNA database obtained through next-generation sequencing. In this study, we first analyzed granulocyte-macrophage colony-stimulating factor (GM-CSF) autoantibodies in a patient with autoimmune pulmonary alveolar proteinosis, using the trapped ion mobility spectrometry coupled with quadrupole time-of-flight (TIMS-TOF) instrument. The TIMS-TOF instrument identified peptides that partially matched sequences in up to 156 out of 162 cDNA clones. Complementarity-determining region 3 (CDR3) was fully and partially detected in nine and 132 clones, respectively. Moreover, we confirmed one unique framework region 4 (FR4) and at least three unique across CDR3 to FR4 peptides via de novo peptide sequencing. This new technology may thus permit the comprehensive identification of autoantibody structure.
Collapse
Affiliation(s)
| | - Shiho Takeuchi
- Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
| | | | | | | | - Takahiro Tanaka
- Niigata University Medical & Dental Hospital, Niigata, Japan
| | - Koh Nakata
- Niigata University Medical & Dental Hospital, Niigata, Japan.
| |
Collapse
|
37
|
Cotzomi E, Stathopoulos P, Lee CS, Ritchie AM, Soltys JN, Delmotte FR, Oe T, Sng J, Jiang R, Ma AK, Vander Heiden JA, Kleinstein SH, Levy M, Bennett JL, Meffre E, O'Connor KC. Early B cell tolerance defects in neuromyelitis optica favour anti-AQP4 autoantibody production. Brain 2020; 142:1598-1615. [PMID: 31056665 DOI: 10.1093/brain/awz106] [Citation(s) in RCA: 60] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2018] [Revised: 02/13/2019] [Accepted: 02/24/2019] [Indexed: 11/12/2022] Open
Abstract
Neuromyelitis optica spectrum disorders (NMOSD) constitute rare autoimmune disorders of the CNS that are primarily characterized by severe inflammation of the spinal cord and optic nerve. Approximately 75% of NMOSD patients harbour circulating pathogenic autoantibodies targeting the aquaporin-4 water channel (AQP4). The source of these autoantibodies remains unclear, but parallels between NMOSD and other autoantibody-mediated diseases posit compromised B cell tolerance checkpoints as common underlying and contributing factors. Using a well established assay, we assessed tolerance fidelity by creating recombinant antibodies from B cell populations directly downstream of each checkpoint and testing them for polyreactivity and autoreactivity. We examined a total of 863 recombinant antibodies. Those derived from three anti-AQP4-IgG seropositive NMOSD patients (n = 130) were compared to 733 antibodies from 15 healthy donors. We found significantly higher frequencies of poly- and autoreactive new emigrant/transitional and mature naïve B cells in NMOSD patients compared to healthy donors (P-values < 0.003), thereby identifying defects in both central and peripheral B cell tolerance checkpoints in these patients. We next explored whether pathogenic NMOSD anti-AQP4 autoantibodies can originate from the pool of poly- and autoreactive clones that populate the naïve B cell compartment of NMOSD patients. Six human anti-AQP4 autoantibodies that acquired somatic mutations were reverted back to their unmutated germline precursors, which were tested for both binding to AQP4 and poly- or autoreactivity. While the affinity of mature autoantibodies against AQP4 ranged from modest to strong (Kd 15.2-559 nM), none of the germline revertants displayed any detectable binding to AQP4, revealing that somatic hypermutation is required for the generation of anti-AQP4 autoantibodies. However, two (33.3%) germline autoantibody revertants were polyreactive and four (66.7%) were autoreactive, suggesting that pathogenic anti-AQP4 autoantibodies can originate from the pool of autoreactive naïve B cells, which develops as a consequence of impaired early B cell tolerance checkpoints in NMOSD patients.
Collapse
Affiliation(s)
- Elizabeth Cotzomi
- Department of Neurology, Yale University School of Medicine, New Haven, CT, USA.,Department of Immunobiology, Yale University School of Medicine, New Haven, CT, USA
| | - Panos Stathopoulos
- Department of Neurology, Yale University School of Medicine, New Haven, CT, USA.,Department of Immunobiology, Yale University School of Medicine, New Haven, CT, USA
| | - Casey S Lee
- Department of Neurology, Yale University School of Medicine, New Haven, CT, USA.,Department of Immunobiology, Yale University School of Medicine, New Haven, CT, USA
| | - Alanna M Ritchie
- Departments of Neurology and Ophthalmology and Neuroscience Program, University of Colorado, Denver, CO, USA
| | - John N Soltys
- Departments of Neurology and Ophthalmology and Neuroscience Program, University of Colorado, Denver, CO, USA
| | - Fabien R Delmotte
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT, USA
| | - Tyler Oe
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT, USA
| | - Joel Sng
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT, USA
| | - Ruoyi Jiang
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT, USA
| | - Anthony K Ma
- Department of Pathology, Yale University School of Medicine, New Haven, CT, USA
| | | | - Steven H Kleinstein
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT, USA.,Department of Pathology, Yale University School of Medicine, New Haven, CT, USA.,Interdepartmental Program in Computational Biology and Bioinformatics, Yale University School of Medicine, New Haven, CT, USA
| | - Michael Levy
- Department of Neurology, Johns Hopkins, School of Medicine, Baltimore, MD, USA
| | - Jeffrey L Bennett
- Departments of Neurology and Ophthalmology and Neuroscience Program, University of Colorado, Denver, CO, USA
| | - Eric Meffre
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT, USA
| | - Kevin C O'Connor
- Department of Neurology, Yale University School of Medicine, New Haven, CT, USA.,Department of Immunobiology, Yale University School of Medicine, New Haven, CT, USA
| |
Collapse
|
38
|
Natural Autoantibodies in Chronic Pulmonary Diseases. Int J Mol Sci 2020; 21:ijms21031138. [PMID: 32046322 PMCID: PMC7037933 DOI: 10.3390/ijms21031138] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2019] [Revised: 02/06/2020] [Accepted: 02/07/2020] [Indexed: 12/14/2022] Open
Abstract
In autoantibody-mediated autoimmune diseases, pathogenic autoantibodies generated by a failure of central or peripheral tolerance, have different effects mediated by a variety of mechanisms. Interestingly, even non-autoimmune chronic diseases have a set of disease-specific natural autoantibodies that are maintained for a long time. Because most of these natural autoantibodies target intracellular proteins or long non-coding RNAs, they are speculated to be non-pathological and have some important as yet unrecognized physiological functions such as debris clearance. Recently, we revealed a set of disease-specific natural autoantibodies of chronic pulmonary diseases with unknown etiology by protein arrays that enable detection of specific autoantibodies against >8000 targets. Surprisingly, some of the targeted antigens of disease-specific autoantibodies were subsequently reported by other laboratories as strongly associated with the disease, suggesting that these antigens reflect the pathology of each disease. Furthermore, some of these autoantibodies that target extracellular antigens might modify the original course of each disease. Here, we review the disease-specific natural autoantibodies of chronic pulmonary diseases, including chronic fibrosing idiopathic interstitial pneumonias, sarcoidosis, and autoimmune pulmonary alveolar proteinosis, and discuss their utility and effects.
Collapse
|
39
|
Damiani G, McCormick TS, Leal LO, Ghannoum MA. Recombinant human granulocyte macrophage-colony stimulating factor expressed in yeast (sargramostim): A potential ally to combat serious infections. Clin Immunol 2020; 210:108292. [DOI: 10.1016/j.clim.2019.108292] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2019] [Revised: 10/09/2019] [Accepted: 10/23/2019] [Indexed: 12/27/2022]
|
40
|
Toullec L, Batteux F, Santulli P, Chouzenoux S, Jeljeli M, Belmondo T, Hue S, Chapron C. High Levels of Anti-GM-CSF Antibodies in Deep Infiltrating Endometriosis. Reprod Sci 2020; 27:211-217. [PMID: 32046390 DOI: 10.1007/s43032-019-00021-8] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2019] [Accepted: 03/25/2019] [Indexed: 12/19/2022]
Abstract
Endometriosis is a chronic hormono-dependent inflammatory gynecological disease. Endometriosis can be subdivided into three forms: superficial peritoneal implants, endometrioma, and deep infiltrating endometriosis (DIE). Inflammation is a typical feature of endometriosis with overproduction of prostaglandins, chemokines, and cytokines, like granulocyte-macrophage colony-stimulating factor (GM-CSF). GM-CSF is a hematopoietic growth factor and immune modulator which belongs to the group of cytokines that actively participate in inflammatory reactions. GM-CSF autoantibodies (Ab) are described in inflammatory diseases such as Crohn disease and ulcerative colitis where high concentrations of anti-GM-CSF Ab are correlated with severity, complications, and relapses. We have evaluated the presence of anti-GM-CSF Ab in the serum of 106 patients with endometriosis and 92 controls using a home-made enzyme-linked immunosorbent assay (ELISA) and correlated the results with the form and severity of the disease. We found that anti-GM-CSF Ab level is significantly increased in the sera of patients with endometriosis compared to controls and is associated with the severity of the disease especially in patients with deep endometriosis (p < 0.0001) with the highest number of lesions (p = 0.0034), including digestive involvement (p = 0.0041). We also found a correlation between these levels of anti-GM-CSF Ab and the number of lesions in DIE patients (r = 0.913). In this way, searching anti-GM-CSF Ab in endometriosis patient sera could be of value for patient follow-up and put further insight into the role of inflammation and of GM-CSF in endometriosis pathogenesis.
Collapse
Affiliation(s)
- Laurie Toullec
- Department of Immunology, Cochin University Hospital, APHP, Paris, France.,Department of Immuno-hematology, Henri Mondor University Hospital, APHP, Créteil, France
| | - Frédéric Batteux
- Department of Immunology, Cochin University Hospital, APHP, Paris, France. .,INSERM U1016, Institut Cochin, Paris, France.
| | - Pietro Santulli
- Department of Gynecology Obstetrics II and Reproductive Medicine, Cochin University Hospital, Paris, France.,INSERM U1016, Institut Cochin, Paris, France
| | | | - Mohamed Jeljeli
- Department of Immunology, Cochin University Hospital, APHP, Paris, France.,INSERM U1016, Institut Cochin, Paris, France
| | - Thibaut Belmondo
- Department of Immuno-hematology, Henri Mondor University Hospital, APHP, Créteil, France
| | - Sophie Hue
- Department of Immuno-hematology, Henri Mondor University Hospital, APHP, Créteil, France.
| | - Charles Chapron
- Department of Gynecology Obstetrics II and Reproductive Medicine, Cochin University Hospital, Paris, France. .,INSERM U1016, Institut Cochin, Paris, France.
| |
Collapse
|
41
|
Ichiwata T, Ishida M, Itoh Y, Kitamura N, Nakata K. Does maternal autoantibody that transfer to newborn cause disease? Respirol Case Rep 2019; 7:e00494. [PMID: 31632678 PMCID: PMC6788979 DOI: 10.1002/rcr2.494] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2019] [Revised: 08/31/2019] [Accepted: 09/03/2019] [Indexed: 11/23/2022] Open
Abstract
Autoimmune pulmonary alveolar proteinosis (aPAP) is associated with excess amount of granulocyte-macrophage colony-stimulating factor (GM-CSF) autoantibody (GMAb) in the lung and blood. We experienced a female case with severe aPAP who could continue her pregnancy under home oxygen therapy and delivered a newborn by caesarean section. Maternal serum GMAb remained high level for up to one year after the delivery, although aPAP entered remission by whole lung lavage. While the newborn oxygen saturation as well as serum Krebs von den Lungen-6 and surfactant protein-D levels had been normal until one year. As GMAb likely transfer to the newborn and might cause the same disease, we carefully monitored both maternal and the newborn serum GMAb levels after the birth for up to one year. We confirmed that GMAb passively transferred to the newborn circulation but rapidly decreased exponentially to the cut-off level. It is suggested that this rapid decrease might prevent the newborn from developing aPAP.
Collapse
Affiliation(s)
| | - Manabu Ishida
- Hachioji Medical CenterTokyo Medical UniversityTokyoJapan
| | - Yuko Itoh
- Bioscience Medical Research CenterNiigata University Medical and Dental HospitalNiigataJapan
| | - Nobutaka Kitamura
- Bioscience Medical Research CenterNiigata University Medical and Dental HospitalNiigataJapan
| | - Koh Nakata
- Bioscience Medical Research CenterNiigata University Medical and Dental HospitalNiigataJapan
| |
Collapse
|
42
|
Burnett DL, Reed JH, Christ D, Goodnow CC. Clonal redemption and clonal anergy as mechanisms to balance B cell tolerance and immunity. Immunol Rev 2019; 292:61-75. [DOI: 10.1111/imr.12808] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2019] [Revised: 09/10/2019] [Accepted: 09/13/2019] [Indexed: 12/19/2022]
Affiliation(s)
- Deborah L. Burnett
- Garvan Institute of Medical Research Darlinghurst NSW Australia
- St Vincent's Clinical School UNSW Sydney Darlinghurst NSW Australia
| | - Joanne H. Reed
- Garvan Institute of Medical Research Darlinghurst NSW Australia
- St Vincent's Clinical School UNSW Sydney Darlinghurst NSW Australia
| | - Daniel Christ
- Garvan Institute of Medical Research Darlinghurst NSW Australia
- St Vincent's Clinical School UNSW Sydney Darlinghurst NSW Australia
| | - Christopher C. Goodnow
- Garvan Institute of Medical Research Darlinghurst NSW Australia
- St Vincent's Clinical School UNSW Sydney Darlinghurst NSW Australia
| |
Collapse
|
43
|
Singh A, Jindal AK, Joshi V, Anjani G, Rawat A. An updated review on phenocopies of primary immunodeficiency diseases. Genes Dis 2019; 7:12-25. [PMID: 32181272 PMCID: PMC7063430 DOI: 10.1016/j.gendis.2019.09.007] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2019] [Revised: 08/28/2019] [Accepted: 09/04/2019] [Indexed: 02/07/2023] Open
Abstract
Primary immunodeficiency diseases (PIDs) refer to a heterogenous group of disorders characterized clinically by increased susceptibility to infections, autoimmunity and increased risk of malignancies. These group of disorders present with clinical manifestations similar to PIDs with known genetic defects but have either no genetic defect or have a somatic mutation and thus have been labelled as “Phenocopies of PIDs”. These diseases have been further subdivided into those associated with somatic mutations and those associated with presence of auto-antibodies against various cytokines. In this review, we provide an update on clinical manifestations, diagnosis and management of these diseases.
Collapse
Affiliation(s)
- Ankita Singh
- Allergy Immunology Unit, Department of Pediatrics, Advanced Pediatrics Centre, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Ankur K Jindal
- Allergy Immunology Unit, Department of Pediatrics, Advanced Pediatrics Centre, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Vibhu Joshi
- Allergy Immunology Unit, Department of Pediatrics, Advanced Pediatrics Centre, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Gummadi Anjani
- Allergy Immunology Unit, Department of Pediatrics, Advanced Pediatrics Centre, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Amit Rawat
- Allergy Immunology Unit, Department of Pediatrics, Advanced Pediatrics Centre, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| |
Collapse
|
44
|
Merkel PA, Lebo T, Knight V. Functional Analysis of Anti-cytokine Autoantibodies Using Flow Cytometry. Front Immunol 2019; 10:1517. [PMID: 31354706 PMCID: PMC6640114 DOI: 10.3389/fimmu.2019.01517] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2019] [Accepted: 06/18/2019] [Indexed: 12/23/2022] Open
Abstract
Autoantibodies to cytokines are increasingly being detected in association with immunodeficient, autoimmune and immune dysregulated states. Presence of these autoantibodies in an otherwise healthy individual may result in a unique phenotype characterized by predisposition to infection with specific organisms. The ability to detect these autoantibodies is of importance as it may direct treatment toward a combination of anti-microbial agents and immunomodulatory therapies that decrease autoantibody levels, thereby releasing the immune system from autoantibody-mediated inhibition. Ligand binding assays such as ELISA or bead multiplex assays have been used to detect these antibodies. However, not all anti-cytokine autoantibodies have demonstrable function in vitro and therefore their clinical significance is unclear. Assays that evaluate the functionality of anti-cytokine autoantibodies can supplement such ligand binding assays and add valuable functional information that, when viewed in the context of the clinical phenotype, may guide the use of adjunctive immunomodulatory therapy. This mini review provides an overview of anti-cytokine autoantibodies identified to date and their clinical associations. It also describes the use of flow cytometry for the functional analysis of anti-IFNγ and anti-GM-CSF autoantibodies.
Collapse
Affiliation(s)
- Patricia A Merkel
- Section of Allergy and Immunology, Department of Pediatrics, University of Colorado School of Medicine, Denver, CO, United States
| | - Terri Lebo
- Advanced Diagnostic Laboratories, National Jewish Health, Denver, CO, United States
| | - Vijaya Knight
- Section of Allergy and Immunology, Department of Pediatrics, University of Colorado School of Medicine, Denver, CO, United States
| |
Collapse
|
45
|
Nei T, Urano S, Motoi N, Hashimoto A, Kitamura N, Tanaka T, Nakagaki K, Takizawa J, Kaneko C, Tazawa R, Nakata K. Memory B cell pool of autoimmune pulmonary alveolar proteinosis patients contains higher frequency of GM-CSF autoreactive B cells than healthy subjects. Immunol Lett 2019; 212:22-29. [PMID: 31195018 DOI: 10.1016/j.imlet.2019.05.013] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2019] [Revised: 05/12/2019] [Accepted: 05/24/2019] [Indexed: 11/25/2022]
Abstract
The IgG-type neutralizing GM-CSF autoantibody (GMAb) is known to be the causative agent for autoimmune pulmonary alveolar proteinosis (APAP). Previous studies report that serum levels of IgG-GMAb are approximately 50-fold higher in APAP patients than in healthy subjects (HS). Serum levels of IgM-GMAb are also higher in APAP patients than in HS, but this has been assumed to be an etiological bystander. However, the mechanism for the excessive production of IgG-GMAb in APAP remains unclear. To investigate this, we detected putative GMAb-producing B cells (PGMPB) by inoculated B cells from the peripheral blood of APAP patients, HS, and umbilical cord blood mononuclear cells (UCBMNs) with Epstein-Barr virus. Both ELISA and ELISPOT assays showed that IgM-type GMAb was consistently and frequently present in all three groups, whereas IgG-type GMAb was high only in APAP patients, in whom it was exclusively produced in memory B cells and not in naive B cells. Since PGMPB in UCBMNs produced IgM-GMAb, but not IgG-GMAb, to the same extent as in HS and APAP patients, most IgM-GMAb reacted with GM-CSF in a non-specific manner. The memory B cell pool of APAP patients contain higher frequency of PGMPB than that of healthy subjects.
Collapse
Affiliation(s)
- Takahito Nei
- Clinical and Translational Research Center, Niigata University Medical and Dental Hospital, Niigata, Japan; Department of Infection Control and Prevention, Nippon Medical School Hospital, Tokyo, Japan
| | - Shinya Urano
- Clinical and Translational Research Center, Niigata University Medical and Dental Hospital, Niigata, Japan
| | - Natsuki Motoi
- Clinical and Translational Research Center, Niigata University Medical and Dental Hospital, Niigata, Japan
| | - Atsushi Hashimoto
- Clinical and Translational Research Center, Niigata University Medical and Dental Hospital, Niigata, Japan
| | - Nobutaka Kitamura
- Clinical and Translational Research Center, Niigata University Medical and Dental Hospital, Niigata, Japan
| | - Takahiro Tanaka
- Clinical and Translational Research Center, Niigata University Medical and Dental Hospital, Niigata, Japan
| | - Kazuhide Nakagaki
- Clinical and Translational Research Center, Niigata University Medical and Dental Hospital, Niigata, Japan
| | - Jun Takizawa
- Clinical and Translational Research Center, Niigata University Medical and Dental Hospital, Niigata, Japan
| | - Chinatsu Kaneko
- Clinical and Translational Research Center, Niigata University Medical and Dental Hospital, Niigata, Japan
| | - Ryushi Tazawa
- Clinical and Translational Research Center, Niigata University Medical and Dental Hospital, Niigata, Japan
| | - Koh Nakata
- Clinical and Translational Research Center, Niigata University Medical and Dental Hospital, Niigata, Japan.
| |
Collapse
|
46
|
Piloni D, Campo I. Current management strategies and the potential of inhaled GM-CSF for the treatment of autoimmune pulmonary alveolar proteinosis. Expert Opin Orphan Drugs 2019. [DOI: 10.1080/21678707.2019.1589450] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Affiliation(s)
- Davide Piloni
- Pneumology Unit, IRCCS Policlinico San Matteo Hospital Foundation, Pavia, Italy
| | - Ilaria Campo
- Pneumology Unit, IRCCS Policlinico San Matteo Hospital Foundation, Pavia, Italy
| |
Collapse
|
47
|
Trapnell BC, Nakata K, Bonella F, Campo I, Griese M, Hamilton J, Wang T, Morgan C, Cottin V, McCarthy C. Pulmonary alveolar proteinosis. Nat Rev Dis Primers 2019; 5:16. [PMID: 30846703 DOI: 10.1038/s41572-019-0066-3] [Citation(s) in RCA: 214] [Impact Index Per Article: 35.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
Pulmonary alveolar proteinosis (PAP) is a syndrome characterized by the accumulation of alveolar surfactant and dysfunction of alveolar macrophages. PAP results in progressive dyspnoea of insidious onset, hypoxaemic respiratory failure, secondary infections and pulmonary fibrosis. PAP can be classified into different types on the basis of the pathogenetic mechanism: primary PAP is characterized by the disruption of granulocyte-macrophage colony-stimulating factor (GM-CSF) signalling and can be autoimmune (caused by elevated levels of GM-CSF autoantibodies) or hereditary (due to mutations in CSF2RA or CSF2RB, encoding GM-CSF receptor subunits); secondary PAP results from various underlying conditions; and congenital PAP is caused by mutations in genes involved in surfactant production. In most patients, pathogenesis is driven by reduced GM-CSF-dependent cholesterol clearance in alveolar macrophages, which impairs alveolar surfactant clearance. PAP has a prevalence of at least 7 cases per million individuals in large population studies and affects men, women and children of all ages, ethnicities and geographical locations irrespective of socioeconomic status, although it is more-prevalent in smokers. Autoimmune PAP accounts for >90% of all cases. Management aims at improving symptoms and quality of life; whole-lung lavage effectively removes excessive surfactant. Novel pathogenesis-based therapies are in development, targeting GM-CSF signalling, immune modulation and cholesterol homeostasis.
Collapse
Affiliation(s)
- Bruce C Trapnell
- Translational Pulmonary Science Center, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA.
| | - Koh Nakata
- Bioscience Medical Research Center, Niigata University, Niigata, Japan
| | - Francesco Bonella
- Interstitial and Rare Lung Disease Unit, Pneumology Department, Ruhrlandklinik University Hospital, University of Essen, Essen, Germany
| | - Ilaria Campo
- Pneumology Unit, IRCCS San Matteo Hospital Foundation, Pavia, Italy
| | - Matthias Griese
- Pediatric Pneumology, University of Munich, German Center for Lung Research (DZL), Munich, Germany
| | - John Hamilton
- University of Melbourne, Parkville, Victoria, Australia
| | - Tisha Wang
- Department of Medicine, University of California, Los Angeles, CA, USA
| | - Cliff Morgan
- Department of Critical Care and Anaesthesia, Royal Brompton Hospital, London, UK
| | - Vincent Cottin
- National Reference Center for Rare Pulmonary Diseases, University of Lyon, Lyon, France
| | - Cormac McCarthy
- Department of Medicine, St. Vincent's University Hospital and University College Dublin, Dublin, Ireland
| |
Collapse
|
48
|
Lanzavecchia A. Dissecting human antibody responses: useful, basic and surprising findings. EMBO Mol Med 2019; 10:emmm.201808879. [PMID: 29363490 PMCID: PMC5840544 DOI: 10.15252/emmm.201808879] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
Human memory B cells and plasma cells represent a rich source of antibodies that have been selected in response to human pathogens. In the last decade, different methods have been developed to interrogate the human memory repertoire and isolate monoclonal antibodies. I will discuss how a target‐agnostic approach based on high‐throughput screening of antibodies produced by cultured B cells and plasma cells has not only provided potent and broadly neutralizing antibodies against a range of pathogens, but has also advanced our understanding of basic aspects of the immune response, from host–pathogen interaction to the role of somatic mutations in affinity maturation and in the diversification of the antibody response. Most surprisingly, this approach has also revealed a new mechanism of diversification based on templated insertion of non‐Ig DNA into antibody genes that we discovered in the context of the immune response to malaria infection.
Collapse
Affiliation(s)
- Antonio Lanzavecchia
- Institute for Research in Biomedicine, Università della Svizzera Italiana, Bellinzona, Switzerland
| |
Collapse
|
49
|
The role of APRIL - A proliferation inducing ligand - In autoimmune diseases and expectations from its targeting. J Autoimmun 2018; 95:179-190. [DOI: 10.1016/j.jaut.2018.10.016] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2018] [Accepted: 10/17/2018] [Indexed: 12/12/2022]
|
50
|
Dhagat U, Hercus TR, Broughton SE, Nero TL, Cheung Tung Shing KS, Barry EF, Thomson CA, Bryson S, Pai EF, McClure BJ, Schrader JW, Lopez AF, Parker MW. The mechanism of GM-CSF inhibition by human GM-CSF auto-antibodies suggests novel therapeutic opportunities. MAbs 2018; 10:1018-1029. [PMID: 29969365 DOI: 10.1080/19420862.2018.1494107] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/28/2022] Open
Abstract
Granulocyte-macrophage colony-stimulating factor (GM-CSF) is a hematopoietic growth factor that can stimulate a variety of cells, but its overexpression leads to excessive production and activation of granulocytes and macrophages with many pathogenic effects. This cytokine is a therapeutic target in inflammatory diseases, and several anti-GM-CSF antibodies have advanced to Phase 2 clinical trials in patients with such diseases, e.g., rheumatoid arthritis. GM-CSF is also an essential factor in preventing pulmonary alveolar proteinosis (PAP), a disease associated with GM-CSF malfunction arising most typically through the presence of GM-CSF neutralizing auto-antibodies. Understanding the mechanism of action for neutralizing antibodies that target GM-CSF is important for improving their specificity and affinity as therapeutics and, conversely, in devising strategies to reduce the effects of GM-CSF auto-antibodies in PAP. We have solved the crystal structures of human GM-CSF bound to antigen-binding fragments of two neutralizing antibodies, the human auto-antibody F1 and the mouse monoclonal antibody 4D4. Coordinates and structure factors of the crystal structures of the GM-CSF:F1 Fab and the GM-CSF:4D4 Fab complexes have been deposited in the RCSB Protein Data Bank under the accession numbers 6BFQ and 6BFS, respectively. The structures show that these antibodies bind to mutually exclusive epitopes on GM-CSF; however, both prevent the cytokine from interacting with its alpha receptor subunit and hence prevent receptor activation. Importantly, identification of the F1 epitope together with functional analyses highlighted modifications to GM-CSF that would abolish auto-antibody recognition whilst retaining GM-CSF function. These results provide a framework for developing novel GM-CSF molecules for PAP treatment and for optimizing current anti-GM-CSF antibodies for use in treating inflammatory disorders.
Collapse
Affiliation(s)
- Urmi Dhagat
- a St. Vincent's Institute of Medical Research , Australian Cancer Research Foundation Rational Drug Discovery Centre , Fitzroy , Victoria , Australia.,c Department of Biochemistry and Molecular Biology, Bio21 Molecular Science and Biotechnology Institute , University of Melbourne , Parkville , Victoria , Australia
| | - Timothy R Hercus
- b The Centre for Cancer Biology , SA Pathology and the University of South Australia , Adelaide , South Australia , Australia
| | - Sophie E Broughton
- a St. Vincent's Institute of Medical Research , Australian Cancer Research Foundation Rational Drug Discovery Centre , Fitzroy , Victoria , Australia.,c Department of Biochemistry and Molecular Biology, Bio21 Molecular Science and Biotechnology Institute , University of Melbourne , Parkville , Victoria , Australia
| | - Tracy L Nero
- a St. Vincent's Institute of Medical Research , Australian Cancer Research Foundation Rational Drug Discovery Centre , Fitzroy , Victoria , Australia.,c Department of Biochemistry and Molecular Biology, Bio21 Molecular Science and Biotechnology Institute , University of Melbourne , Parkville , Victoria , Australia
| | - Karen S Cheung Tung Shing
- a St. Vincent's Institute of Medical Research , Australian Cancer Research Foundation Rational Drug Discovery Centre , Fitzroy , Victoria , Australia.,c Department of Biochemistry and Molecular Biology, Bio21 Molecular Science and Biotechnology Institute , University of Melbourne , Parkville , Victoria , Australia
| | - Emma F Barry
- b The Centre for Cancer Biology , SA Pathology and the University of South Australia , Adelaide , South Australia , Australia
| | - Christy A Thomson
- d The Biomedical Research Centre , University of British Columbia , Vancouver , British Columbia , Canada
| | - Steve Bryson
- e Princess Margaret Cancer Centre, University Health Network, University of Toronto , Toronto , Ontario , Canada.,f Department of Biochemistry , University of Toronto , Toronto , Ontario , Canada
| | - Emil F Pai
- e Princess Margaret Cancer Centre, University Health Network, University of Toronto , Toronto , Ontario , Canada.,f Department of Biochemistry , University of Toronto , Toronto , Ontario , Canada.,g Department of Medical Biophysics , University of Toronto , Toronto , Ontario , Canada.,h Department of Molecular Genetics , University of Toronto , Toronto , Ontario , Canada
| | - Barbara J McClure
- b The Centre for Cancer Biology , SA Pathology and the University of South Australia , Adelaide , South Australia , Australia
| | - John W Schrader
- d The Biomedical Research Centre , University of British Columbia , Vancouver , British Columbia , Canada.,g Department of Medical Biophysics , University of Toronto , Toronto , Ontario , Canada
| | - Angel F Lopez
- b The Centre for Cancer Biology , SA Pathology and the University of South Australia , Adelaide , South Australia , Australia.,i Department of Medicine , University of Adelaide , Adelaide , South Australia , Australia
| | - Michael W Parker
- a St. Vincent's Institute of Medical Research , Australian Cancer Research Foundation Rational Drug Discovery Centre , Fitzroy , Victoria , Australia.,c Department of Biochemistry and Molecular Biology, Bio21 Molecular Science and Biotechnology Institute , University of Melbourne , Parkville , Victoria , Australia
| |
Collapse
|