1
|
Paparoditis PCG, Fruehwirth A, Bevc K, Low JS, Jerak J, Terzaghi L, Foglierini M, Fernandez B, Jarrossay D, Corti D, Sallusto F, Lanzavecchia A, Cassotta A. Site-specific serology unveils cross-reactive monoclonal antibodies targeting influenza A hemagglutinin epitopes. Eur J Immunol 2024; 54:e2451045. [PMID: 39031535 DOI: 10.1002/eji.202451045] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2024] [Revised: 07/03/2024] [Accepted: 07/05/2024] [Indexed: 07/22/2024]
Abstract
Efficient identification of human monoclonal antibodies targeting specific antigenic sites is pivotal for advancing vaccines and immunotherapies against infectious diseases and cancer. Existing screening techniques, however, limit our ability to discover monoclonal antibodies with desired specificity. In this study, we introduce a novel method, blocking of binding (BoB) enzyme-linked immunoassay (ELISA), enabling the detection of high-avidity human antibodies directed to defined epitopes. Leveraging BoB-ELISA, we analyzed the antibody response to known epitopes of influenza A hemagglutinin (HA) in the serum of vaccinated donors. Our findings revealed that serum antibodies targeting head epitopes were immunodominant, whereas antibodies against the stem epitope, although subdominant, were highly prevalent. Extending our analysis across multiple HA strains, we examined the cross-reactive antibody response targeting the stem epitope. Importantly, employing BoB-ELISA we identified donors harboring potent heterosubtypic antibodies targeting the HA stem. B-cell clonal analysis of these donors revealed three novel, genealogically independent monoclonal antibodies with broad cross-reactivity to multiple HAs. In summary, we demonstrated that BoB-ELISA is a sensitive technique for measuring B-cell epitope immunogenicity, enabling the identification of novel monoclonal antibodies with implications for enhanced vaccine development and immunotherapies.
Collapse
Affiliation(s)
- Philipp C G Paparoditis
- Institute for Research in Biomedicine, Università della Svizzera italiana, Bellinzona, Switzerland
| | - Alexander Fruehwirth
- Institute for Research in Biomedicine, Università della Svizzera italiana, Bellinzona, Switzerland
| | - Kajetana Bevc
- Institute for Research in Biomedicine, Università della Svizzera italiana, Bellinzona, Switzerland
| | - Jun Siong Low
- Institute for Research in Biomedicine, Università della Svizzera italiana, Bellinzona, Switzerland
| | - Josipa Jerak
- Institute for Research in Biomedicine, Università della Svizzera italiana, Bellinzona, Switzerland
| | - Laura Terzaghi
- Institute for Research in Biomedicine, Università della Svizzera italiana, Bellinzona, Switzerland
| | - Mathilde Foglierini
- Institute for Research in Biomedicine, Università della Svizzera italiana, Bellinzona, Switzerland
| | - Blanca Fernandez
- Institute for Research in Biomedicine, Università della Svizzera italiana, Bellinzona, Switzerland
| | - David Jarrossay
- Institute for Research in Biomedicine, Università della Svizzera italiana, Bellinzona, Switzerland
| | - Davide Corti
- Humabs Biomed SA, a subsidiary of Vir Biotechnology, Bellinzona, Switzerland
| | - Federica Sallusto
- Institute for Research in Biomedicine, Università della Svizzera italiana, Bellinzona, Switzerland
- Institute for Microbiology, ETH Zurich, Zurich, Switzerland
| | - Antonio Lanzavecchia
- Humabs Biomed SA, a subsidiary of Vir Biotechnology, Bellinzona, Switzerland
- National Institute of Molecular Genetics, Milano, Italy
| | - Antonino Cassotta
- Institute for Research in Biomedicine, Università della Svizzera italiana, Bellinzona, Switzerland
| |
Collapse
|
2
|
Rohrbacher S, Seefried S, Hartmannsberger B, Annabelle R, Appeltshauser L, Arlt FA, Brämer D, Dresel C, Dorst J, Elmas Z, Franke C, Geis C, Högen T, Krause S, Marziniak M, Mäurer M, Prüss H, Schoeberl F, Schrank B, Steen C, Teichtinger H, Thieme A, Wessely L, Zernecke A, Sommer C, Doppler K. Different Patterns of Autoantibody Secretion by Peripheral Blood Mononuclear Cells in Autoimmune Nodopathies. NEUROLOGY(R) NEUROIMMUNOLOGY & NEUROINFLAMMATION 2024; 11:e200295. [PMID: 39173087 PMCID: PMC11379437 DOI: 10.1212/nxi.0000000000200295] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/24/2024]
Abstract
BACKGROUND AND OBJECTIVES Autoimmune nodopathies with antibodies against the paranodal proteins show a distinct phenotype of a severe sensorimotor neuropathy. In some patients, complete remission can be achieved after treatment with rituximab whereas others show a chronic course. For optimal planning of treatment, predicting the course of disease and therapeutic response is crucial. METHODS We stimulated peripheral blood mononuclear cells in vitro to find out whether secretion of specific autoantibodies may be a predictor of the course of disease and response to rituximab. RESULTS Three patterns could be identified: In most patients with anti-Neurofascin-155-, anti-Contactin-1-, and anti-Caspr1-IgG4 autoantibodies, in vitro production of autoantibodies was detected, indicating autoantigen-specific memory B cells and short-lived plasma cells/plasmablasts as the major source of autoantibodies. These patients generally showed a good response to rituximab. In a subgroup of patients with anti-Neurofascin-155-IgG4 autoantibodies and insufficient response to rituximab, no in vitro autoantibody production was found despite high serum titers, indicating autoantibody secretion by long-lived plasma cells outside the peripheral blood. In the patients with anti-pan-Neurofascin autoantibodies-all with a monophasic course of disease-no in vitro autoantibody production could be measured, suggesting a lack of autoantigen-specific memory B cells. In some of them, autoantibody production by unstimulated cells was detectable, presumably corresponding to high amounts of autoantigen-specific plasmablasts-well in line with a severe but monophasic course of disease. DISCUSSION Our data suggest that different B-cell responses may occur in autoimmune nodopathies and may serve as markers of courses of disease and response to rituximab.
Collapse
Affiliation(s)
- Sophia Rohrbacher
- From the Department of Neurology (S.R., S.S., B.H., L.A., C. Sommer, K.D.); Department of Anaesthesiology, Intensive Care, Emergency and Pain Medicine Centre for Interdisciplinary Pain Medicine (B.H.); Institute of Experimental Biomedicine (R.A., A.Z.), University Hospital Würzburg; German Center for Neurodegenerative Diseases (DZNE) Berlin (F.A.A., H.P.); Department of Neurology and Experimental Neurology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität Berlin; Section Translational Neuroimmunology (D.B., C.G.), Department of Neurology, Jena University Hospital; Department of Neurology (C.D.), University Medical Center of the Johannes Gutenberg University, Mainz; Department of Neurology (J.D., Z.E.), University Hospital Ulm; Department of Neurology (T.H., H.T.), Therapiezentrum Burgau; Department of Neurology (S.K.), Friedrich Baur Institute, LMU University Hospital, LMU Munich; Department of Neurology (M. Marziniak), Kbo-Isar-Amper-Hospital Munich East; Department of Neurology (M. Mäurer), Klinikum Würzburg Mitte gGmbH, Standort Juliusspital; Department of Neurology (F.S.), LMU University Hospital, LMU, Munich; Department of Neurology (B.S.), DKD HELIOS Klinik Wiesbaden; Department of Paediatric and Adolescent Medicine (C. Steen), St Joseph Hospital, Berlin; Department of Neurology (A.T.), HELIOS Klinikum Erfurt; and Neurologische Praxis Dres. Wessely (L.W.), Menden, Germany
| | - Sabine Seefried
- From the Department of Neurology (S.R., S.S., B.H., L.A., C. Sommer, K.D.); Department of Anaesthesiology, Intensive Care, Emergency and Pain Medicine Centre for Interdisciplinary Pain Medicine (B.H.); Institute of Experimental Biomedicine (R.A., A.Z.), University Hospital Würzburg; German Center for Neurodegenerative Diseases (DZNE) Berlin (F.A.A., H.P.); Department of Neurology and Experimental Neurology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität Berlin; Section Translational Neuroimmunology (D.B., C.G.), Department of Neurology, Jena University Hospital; Department of Neurology (C.D.), University Medical Center of the Johannes Gutenberg University, Mainz; Department of Neurology (J.D., Z.E.), University Hospital Ulm; Department of Neurology (T.H., H.T.), Therapiezentrum Burgau; Department of Neurology (S.K.), Friedrich Baur Institute, LMU University Hospital, LMU Munich; Department of Neurology (M. Marziniak), Kbo-Isar-Amper-Hospital Munich East; Department of Neurology (M. Mäurer), Klinikum Würzburg Mitte gGmbH, Standort Juliusspital; Department of Neurology (F.S.), LMU University Hospital, LMU, Munich; Department of Neurology (B.S.), DKD HELIOS Klinik Wiesbaden; Department of Paediatric and Adolescent Medicine (C. Steen), St Joseph Hospital, Berlin; Department of Neurology (A.T.), HELIOS Klinikum Erfurt; and Neurologische Praxis Dres. Wessely (L.W.), Menden, Germany
| | - Beate Hartmannsberger
- From the Department of Neurology (S.R., S.S., B.H., L.A., C. Sommer, K.D.); Department of Anaesthesiology, Intensive Care, Emergency and Pain Medicine Centre for Interdisciplinary Pain Medicine (B.H.); Institute of Experimental Biomedicine (R.A., A.Z.), University Hospital Würzburg; German Center for Neurodegenerative Diseases (DZNE) Berlin (F.A.A., H.P.); Department of Neurology and Experimental Neurology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität Berlin; Section Translational Neuroimmunology (D.B., C.G.), Department of Neurology, Jena University Hospital; Department of Neurology (C.D.), University Medical Center of the Johannes Gutenberg University, Mainz; Department of Neurology (J.D., Z.E.), University Hospital Ulm; Department of Neurology (T.H., H.T.), Therapiezentrum Burgau; Department of Neurology (S.K.), Friedrich Baur Institute, LMU University Hospital, LMU Munich; Department of Neurology (M. Marziniak), Kbo-Isar-Amper-Hospital Munich East; Department of Neurology (M. Mäurer), Klinikum Würzburg Mitte gGmbH, Standort Juliusspital; Department of Neurology (F.S.), LMU University Hospital, LMU, Munich; Department of Neurology (B.S.), DKD HELIOS Klinik Wiesbaden; Department of Paediatric and Adolescent Medicine (C. Steen), St Joseph Hospital, Berlin; Department of Neurology (A.T.), HELIOS Klinikum Erfurt; and Neurologische Praxis Dres. Wessely (L.W.), Menden, Germany
| | - Rosa Annabelle
- From the Department of Neurology (S.R., S.S., B.H., L.A., C. Sommer, K.D.); Department of Anaesthesiology, Intensive Care, Emergency and Pain Medicine Centre for Interdisciplinary Pain Medicine (B.H.); Institute of Experimental Biomedicine (R.A., A.Z.), University Hospital Würzburg; German Center for Neurodegenerative Diseases (DZNE) Berlin (F.A.A., H.P.); Department of Neurology and Experimental Neurology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität Berlin; Section Translational Neuroimmunology (D.B., C.G.), Department of Neurology, Jena University Hospital; Department of Neurology (C.D.), University Medical Center of the Johannes Gutenberg University, Mainz; Department of Neurology (J.D., Z.E.), University Hospital Ulm; Department of Neurology (T.H., H.T.), Therapiezentrum Burgau; Department of Neurology (S.K.), Friedrich Baur Institute, LMU University Hospital, LMU Munich; Department of Neurology (M. Marziniak), Kbo-Isar-Amper-Hospital Munich East; Department of Neurology (M. Mäurer), Klinikum Würzburg Mitte gGmbH, Standort Juliusspital; Department of Neurology (F.S.), LMU University Hospital, LMU, Munich; Department of Neurology (B.S.), DKD HELIOS Klinik Wiesbaden; Department of Paediatric and Adolescent Medicine (C. Steen), St Joseph Hospital, Berlin; Department of Neurology (A.T.), HELIOS Klinikum Erfurt; and Neurologische Praxis Dres. Wessely (L.W.), Menden, Germany
| | - Luise Appeltshauser
- From the Department of Neurology (S.R., S.S., B.H., L.A., C. Sommer, K.D.); Department of Anaesthesiology, Intensive Care, Emergency and Pain Medicine Centre for Interdisciplinary Pain Medicine (B.H.); Institute of Experimental Biomedicine (R.A., A.Z.), University Hospital Würzburg; German Center for Neurodegenerative Diseases (DZNE) Berlin (F.A.A., H.P.); Department of Neurology and Experimental Neurology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität Berlin; Section Translational Neuroimmunology (D.B., C.G.), Department of Neurology, Jena University Hospital; Department of Neurology (C.D.), University Medical Center of the Johannes Gutenberg University, Mainz; Department of Neurology (J.D., Z.E.), University Hospital Ulm; Department of Neurology (T.H., H.T.), Therapiezentrum Burgau; Department of Neurology (S.K.), Friedrich Baur Institute, LMU University Hospital, LMU Munich; Department of Neurology (M. Marziniak), Kbo-Isar-Amper-Hospital Munich East; Department of Neurology (M. Mäurer), Klinikum Würzburg Mitte gGmbH, Standort Juliusspital; Department of Neurology (F.S.), LMU University Hospital, LMU, Munich; Department of Neurology (B.S.), DKD HELIOS Klinik Wiesbaden; Department of Paediatric and Adolescent Medicine (C. Steen), St Joseph Hospital, Berlin; Department of Neurology (A.T.), HELIOS Klinikum Erfurt; and Neurologische Praxis Dres. Wessely (L.W.), Menden, Germany
| | - Friederike A Arlt
- From the Department of Neurology (S.R., S.S., B.H., L.A., C. Sommer, K.D.); Department of Anaesthesiology, Intensive Care, Emergency and Pain Medicine Centre for Interdisciplinary Pain Medicine (B.H.); Institute of Experimental Biomedicine (R.A., A.Z.), University Hospital Würzburg; German Center for Neurodegenerative Diseases (DZNE) Berlin (F.A.A., H.P.); Department of Neurology and Experimental Neurology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität Berlin; Section Translational Neuroimmunology (D.B., C.G.), Department of Neurology, Jena University Hospital; Department of Neurology (C.D.), University Medical Center of the Johannes Gutenberg University, Mainz; Department of Neurology (J.D., Z.E.), University Hospital Ulm; Department of Neurology (T.H., H.T.), Therapiezentrum Burgau; Department of Neurology (S.K.), Friedrich Baur Institute, LMU University Hospital, LMU Munich; Department of Neurology (M. Marziniak), Kbo-Isar-Amper-Hospital Munich East; Department of Neurology (M. Mäurer), Klinikum Würzburg Mitte gGmbH, Standort Juliusspital; Department of Neurology (F.S.), LMU University Hospital, LMU, Munich; Department of Neurology (B.S.), DKD HELIOS Klinik Wiesbaden; Department of Paediatric and Adolescent Medicine (C. Steen), St Joseph Hospital, Berlin; Department of Neurology (A.T.), HELIOS Klinikum Erfurt; and Neurologische Praxis Dres. Wessely (L.W.), Menden, Germany
| | - Dirk Brämer
- From the Department of Neurology (S.R., S.S., B.H., L.A., C. Sommer, K.D.); Department of Anaesthesiology, Intensive Care, Emergency and Pain Medicine Centre for Interdisciplinary Pain Medicine (B.H.); Institute of Experimental Biomedicine (R.A., A.Z.), University Hospital Würzburg; German Center for Neurodegenerative Diseases (DZNE) Berlin (F.A.A., H.P.); Department of Neurology and Experimental Neurology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität Berlin; Section Translational Neuroimmunology (D.B., C.G.), Department of Neurology, Jena University Hospital; Department of Neurology (C.D.), University Medical Center of the Johannes Gutenberg University, Mainz; Department of Neurology (J.D., Z.E.), University Hospital Ulm; Department of Neurology (T.H., H.T.), Therapiezentrum Burgau; Department of Neurology (S.K.), Friedrich Baur Institute, LMU University Hospital, LMU Munich; Department of Neurology (M. Marziniak), Kbo-Isar-Amper-Hospital Munich East; Department of Neurology (M. Mäurer), Klinikum Würzburg Mitte gGmbH, Standort Juliusspital; Department of Neurology (F.S.), LMU University Hospital, LMU, Munich; Department of Neurology (B.S.), DKD HELIOS Klinik Wiesbaden; Department of Paediatric and Adolescent Medicine (C. Steen), St Joseph Hospital, Berlin; Department of Neurology (A.T.), HELIOS Klinikum Erfurt; and Neurologische Praxis Dres. Wessely (L.W.), Menden, Germany
| | - Christian Dresel
- From the Department of Neurology (S.R., S.S., B.H., L.A., C. Sommer, K.D.); Department of Anaesthesiology, Intensive Care, Emergency and Pain Medicine Centre for Interdisciplinary Pain Medicine (B.H.); Institute of Experimental Biomedicine (R.A., A.Z.), University Hospital Würzburg; German Center for Neurodegenerative Diseases (DZNE) Berlin (F.A.A., H.P.); Department of Neurology and Experimental Neurology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität Berlin; Section Translational Neuroimmunology (D.B., C.G.), Department of Neurology, Jena University Hospital; Department of Neurology (C.D.), University Medical Center of the Johannes Gutenberg University, Mainz; Department of Neurology (J.D., Z.E.), University Hospital Ulm; Department of Neurology (T.H., H.T.), Therapiezentrum Burgau; Department of Neurology (S.K.), Friedrich Baur Institute, LMU University Hospital, LMU Munich; Department of Neurology (M. Marziniak), Kbo-Isar-Amper-Hospital Munich East; Department of Neurology (M. Mäurer), Klinikum Würzburg Mitte gGmbH, Standort Juliusspital; Department of Neurology (F.S.), LMU University Hospital, LMU, Munich; Department of Neurology (B.S.), DKD HELIOS Klinik Wiesbaden; Department of Paediatric and Adolescent Medicine (C. Steen), St Joseph Hospital, Berlin; Department of Neurology (A.T.), HELIOS Klinikum Erfurt; and Neurologische Praxis Dres. Wessely (L.W.), Menden, Germany
| | - Johannes Dorst
- From the Department of Neurology (S.R., S.S., B.H., L.A., C. Sommer, K.D.); Department of Anaesthesiology, Intensive Care, Emergency and Pain Medicine Centre for Interdisciplinary Pain Medicine (B.H.); Institute of Experimental Biomedicine (R.A., A.Z.), University Hospital Würzburg; German Center for Neurodegenerative Diseases (DZNE) Berlin (F.A.A., H.P.); Department of Neurology and Experimental Neurology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität Berlin; Section Translational Neuroimmunology (D.B., C.G.), Department of Neurology, Jena University Hospital; Department of Neurology (C.D.), University Medical Center of the Johannes Gutenberg University, Mainz; Department of Neurology (J.D., Z.E.), University Hospital Ulm; Department of Neurology (T.H., H.T.), Therapiezentrum Burgau; Department of Neurology (S.K.), Friedrich Baur Institute, LMU University Hospital, LMU Munich; Department of Neurology (M. Marziniak), Kbo-Isar-Amper-Hospital Munich East; Department of Neurology (M. Mäurer), Klinikum Würzburg Mitte gGmbH, Standort Juliusspital; Department of Neurology (F.S.), LMU University Hospital, LMU, Munich; Department of Neurology (B.S.), DKD HELIOS Klinik Wiesbaden; Department of Paediatric and Adolescent Medicine (C. Steen), St Joseph Hospital, Berlin; Department of Neurology (A.T.), HELIOS Klinikum Erfurt; and Neurologische Praxis Dres. Wessely (L.W.), Menden, Germany
| | - Zeynep Elmas
- From the Department of Neurology (S.R., S.S., B.H., L.A., C. Sommer, K.D.); Department of Anaesthesiology, Intensive Care, Emergency and Pain Medicine Centre for Interdisciplinary Pain Medicine (B.H.); Institute of Experimental Biomedicine (R.A., A.Z.), University Hospital Würzburg; German Center for Neurodegenerative Diseases (DZNE) Berlin (F.A.A., H.P.); Department of Neurology and Experimental Neurology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität Berlin; Section Translational Neuroimmunology (D.B., C.G.), Department of Neurology, Jena University Hospital; Department of Neurology (C.D.), University Medical Center of the Johannes Gutenberg University, Mainz; Department of Neurology (J.D., Z.E.), University Hospital Ulm; Department of Neurology (T.H., H.T.), Therapiezentrum Burgau; Department of Neurology (S.K.), Friedrich Baur Institute, LMU University Hospital, LMU Munich; Department of Neurology (M. Marziniak), Kbo-Isar-Amper-Hospital Munich East; Department of Neurology (M. Mäurer), Klinikum Würzburg Mitte gGmbH, Standort Juliusspital; Department of Neurology (F.S.), LMU University Hospital, LMU, Munich; Department of Neurology (B.S.), DKD HELIOS Klinik Wiesbaden; Department of Paediatric and Adolescent Medicine (C. Steen), St Joseph Hospital, Berlin; Department of Neurology (A.T.), HELIOS Klinikum Erfurt; and Neurologische Praxis Dres. Wessely (L.W.), Menden, Germany
| | - Christiana Franke
- From the Department of Neurology (S.R., S.S., B.H., L.A., C. Sommer, K.D.); Department of Anaesthesiology, Intensive Care, Emergency and Pain Medicine Centre for Interdisciplinary Pain Medicine (B.H.); Institute of Experimental Biomedicine (R.A., A.Z.), University Hospital Würzburg; German Center for Neurodegenerative Diseases (DZNE) Berlin (F.A.A., H.P.); Department of Neurology and Experimental Neurology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität Berlin; Section Translational Neuroimmunology (D.B., C.G.), Department of Neurology, Jena University Hospital; Department of Neurology (C.D.), University Medical Center of the Johannes Gutenberg University, Mainz; Department of Neurology (J.D., Z.E.), University Hospital Ulm; Department of Neurology (T.H., H.T.), Therapiezentrum Burgau; Department of Neurology (S.K.), Friedrich Baur Institute, LMU University Hospital, LMU Munich; Department of Neurology (M. Marziniak), Kbo-Isar-Amper-Hospital Munich East; Department of Neurology (M. Mäurer), Klinikum Würzburg Mitte gGmbH, Standort Juliusspital; Department of Neurology (F.S.), LMU University Hospital, LMU, Munich; Department of Neurology (B.S.), DKD HELIOS Klinik Wiesbaden; Department of Paediatric and Adolescent Medicine (C. Steen), St Joseph Hospital, Berlin; Department of Neurology (A.T.), HELIOS Klinikum Erfurt; and Neurologische Praxis Dres. Wessely (L.W.), Menden, Germany
| | - Christian Geis
- From the Department of Neurology (S.R., S.S., B.H., L.A., C. Sommer, K.D.); Department of Anaesthesiology, Intensive Care, Emergency and Pain Medicine Centre for Interdisciplinary Pain Medicine (B.H.); Institute of Experimental Biomedicine (R.A., A.Z.), University Hospital Würzburg; German Center for Neurodegenerative Diseases (DZNE) Berlin (F.A.A., H.P.); Department of Neurology and Experimental Neurology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität Berlin; Section Translational Neuroimmunology (D.B., C.G.), Department of Neurology, Jena University Hospital; Department of Neurology (C.D.), University Medical Center of the Johannes Gutenberg University, Mainz; Department of Neurology (J.D., Z.E.), University Hospital Ulm; Department of Neurology (T.H., H.T.), Therapiezentrum Burgau; Department of Neurology (S.K.), Friedrich Baur Institute, LMU University Hospital, LMU Munich; Department of Neurology (M. Marziniak), Kbo-Isar-Amper-Hospital Munich East; Department of Neurology (M. Mäurer), Klinikum Würzburg Mitte gGmbH, Standort Juliusspital; Department of Neurology (F.S.), LMU University Hospital, LMU, Munich; Department of Neurology (B.S.), DKD HELIOS Klinik Wiesbaden; Department of Paediatric and Adolescent Medicine (C. Steen), St Joseph Hospital, Berlin; Department of Neurology (A.T.), HELIOS Klinikum Erfurt; and Neurologische Praxis Dres. Wessely (L.W.), Menden, Germany
| | - Tobias Högen
- From the Department of Neurology (S.R., S.S., B.H., L.A., C. Sommer, K.D.); Department of Anaesthesiology, Intensive Care, Emergency and Pain Medicine Centre for Interdisciplinary Pain Medicine (B.H.); Institute of Experimental Biomedicine (R.A., A.Z.), University Hospital Würzburg; German Center for Neurodegenerative Diseases (DZNE) Berlin (F.A.A., H.P.); Department of Neurology and Experimental Neurology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität Berlin; Section Translational Neuroimmunology (D.B., C.G.), Department of Neurology, Jena University Hospital; Department of Neurology (C.D.), University Medical Center of the Johannes Gutenberg University, Mainz; Department of Neurology (J.D., Z.E.), University Hospital Ulm; Department of Neurology (T.H., H.T.), Therapiezentrum Burgau; Department of Neurology (S.K.), Friedrich Baur Institute, LMU University Hospital, LMU Munich; Department of Neurology (M. Marziniak), Kbo-Isar-Amper-Hospital Munich East; Department of Neurology (M. Mäurer), Klinikum Würzburg Mitte gGmbH, Standort Juliusspital; Department of Neurology (F.S.), LMU University Hospital, LMU, Munich; Department of Neurology (B.S.), DKD HELIOS Klinik Wiesbaden; Department of Paediatric and Adolescent Medicine (C. Steen), St Joseph Hospital, Berlin; Department of Neurology (A.T.), HELIOS Klinikum Erfurt; and Neurologische Praxis Dres. Wessely (L.W.), Menden, Germany
| | - Sabine Krause
- From the Department of Neurology (S.R., S.S., B.H., L.A., C. Sommer, K.D.); Department of Anaesthesiology, Intensive Care, Emergency and Pain Medicine Centre for Interdisciplinary Pain Medicine (B.H.); Institute of Experimental Biomedicine (R.A., A.Z.), University Hospital Würzburg; German Center for Neurodegenerative Diseases (DZNE) Berlin (F.A.A., H.P.); Department of Neurology and Experimental Neurology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität Berlin; Section Translational Neuroimmunology (D.B., C.G.), Department of Neurology, Jena University Hospital; Department of Neurology (C.D.), University Medical Center of the Johannes Gutenberg University, Mainz; Department of Neurology (J.D., Z.E.), University Hospital Ulm; Department of Neurology (T.H., H.T.), Therapiezentrum Burgau; Department of Neurology (S.K.), Friedrich Baur Institute, LMU University Hospital, LMU Munich; Department of Neurology (M. Marziniak), Kbo-Isar-Amper-Hospital Munich East; Department of Neurology (M. Mäurer), Klinikum Würzburg Mitte gGmbH, Standort Juliusspital; Department of Neurology (F.S.), LMU University Hospital, LMU, Munich; Department of Neurology (B.S.), DKD HELIOS Klinik Wiesbaden; Department of Paediatric and Adolescent Medicine (C. Steen), St Joseph Hospital, Berlin; Department of Neurology (A.T.), HELIOS Klinikum Erfurt; and Neurologische Praxis Dres. Wessely (L.W.), Menden, Germany
| | - Martin Marziniak
- From the Department of Neurology (S.R., S.S., B.H., L.A., C. Sommer, K.D.); Department of Anaesthesiology, Intensive Care, Emergency and Pain Medicine Centre for Interdisciplinary Pain Medicine (B.H.); Institute of Experimental Biomedicine (R.A., A.Z.), University Hospital Würzburg; German Center for Neurodegenerative Diseases (DZNE) Berlin (F.A.A., H.P.); Department of Neurology and Experimental Neurology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität Berlin; Section Translational Neuroimmunology (D.B., C.G.), Department of Neurology, Jena University Hospital; Department of Neurology (C.D.), University Medical Center of the Johannes Gutenberg University, Mainz; Department of Neurology (J.D., Z.E.), University Hospital Ulm; Department of Neurology (T.H., H.T.), Therapiezentrum Burgau; Department of Neurology (S.K.), Friedrich Baur Institute, LMU University Hospital, LMU Munich; Department of Neurology (M. Marziniak), Kbo-Isar-Amper-Hospital Munich East; Department of Neurology (M. Mäurer), Klinikum Würzburg Mitte gGmbH, Standort Juliusspital; Department of Neurology (F.S.), LMU University Hospital, LMU, Munich; Department of Neurology (B.S.), DKD HELIOS Klinik Wiesbaden; Department of Paediatric and Adolescent Medicine (C. Steen), St Joseph Hospital, Berlin; Department of Neurology (A.T.), HELIOS Klinikum Erfurt; and Neurologische Praxis Dres. Wessely (L.W.), Menden, Germany
| | - Mathias Mäurer
- From the Department of Neurology (S.R., S.S., B.H., L.A., C. Sommer, K.D.); Department of Anaesthesiology, Intensive Care, Emergency and Pain Medicine Centre for Interdisciplinary Pain Medicine (B.H.); Institute of Experimental Biomedicine (R.A., A.Z.), University Hospital Würzburg; German Center for Neurodegenerative Diseases (DZNE) Berlin (F.A.A., H.P.); Department of Neurology and Experimental Neurology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität Berlin; Section Translational Neuroimmunology (D.B., C.G.), Department of Neurology, Jena University Hospital; Department of Neurology (C.D.), University Medical Center of the Johannes Gutenberg University, Mainz; Department of Neurology (J.D., Z.E.), University Hospital Ulm; Department of Neurology (T.H., H.T.), Therapiezentrum Burgau; Department of Neurology (S.K.), Friedrich Baur Institute, LMU University Hospital, LMU Munich; Department of Neurology (M. Marziniak), Kbo-Isar-Amper-Hospital Munich East; Department of Neurology (M. Mäurer), Klinikum Würzburg Mitte gGmbH, Standort Juliusspital; Department of Neurology (F.S.), LMU University Hospital, LMU, Munich; Department of Neurology (B.S.), DKD HELIOS Klinik Wiesbaden; Department of Paediatric and Adolescent Medicine (C. Steen), St Joseph Hospital, Berlin; Department of Neurology (A.T.), HELIOS Klinikum Erfurt; and Neurologische Praxis Dres. Wessely (L.W.), Menden, Germany
| | - Harald Prüss
- From the Department of Neurology (S.R., S.S., B.H., L.A., C. Sommer, K.D.); Department of Anaesthesiology, Intensive Care, Emergency and Pain Medicine Centre for Interdisciplinary Pain Medicine (B.H.); Institute of Experimental Biomedicine (R.A., A.Z.), University Hospital Würzburg; German Center for Neurodegenerative Diseases (DZNE) Berlin (F.A.A., H.P.); Department of Neurology and Experimental Neurology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität Berlin; Section Translational Neuroimmunology (D.B., C.G.), Department of Neurology, Jena University Hospital; Department of Neurology (C.D.), University Medical Center of the Johannes Gutenberg University, Mainz; Department of Neurology (J.D., Z.E.), University Hospital Ulm; Department of Neurology (T.H., H.T.), Therapiezentrum Burgau; Department of Neurology (S.K.), Friedrich Baur Institute, LMU University Hospital, LMU Munich; Department of Neurology (M. Marziniak), Kbo-Isar-Amper-Hospital Munich East; Department of Neurology (M. Mäurer), Klinikum Würzburg Mitte gGmbH, Standort Juliusspital; Department of Neurology (F.S.), LMU University Hospital, LMU, Munich; Department of Neurology (B.S.), DKD HELIOS Klinik Wiesbaden; Department of Paediatric and Adolescent Medicine (C. Steen), St Joseph Hospital, Berlin; Department of Neurology (A.T.), HELIOS Klinikum Erfurt; and Neurologische Praxis Dres. Wessely (L.W.), Menden, Germany
| | - Florian Schoeberl
- From the Department of Neurology (S.R., S.S., B.H., L.A., C. Sommer, K.D.); Department of Anaesthesiology, Intensive Care, Emergency and Pain Medicine Centre for Interdisciplinary Pain Medicine (B.H.); Institute of Experimental Biomedicine (R.A., A.Z.), University Hospital Würzburg; German Center for Neurodegenerative Diseases (DZNE) Berlin (F.A.A., H.P.); Department of Neurology and Experimental Neurology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität Berlin; Section Translational Neuroimmunology (D.B., C.G.), Department of Neurology, Jena University Hospital; Department of Neurology (C.D.), University Medical Center of the Johannes Gutenberg University, Mainz; Department of Neurology (J.D., Z.E.), University Hospital Ulm; Department of Neurology (T.H., H.T.), Therapiezentrum Burgau; Department of Neurology (S.K.), Friedrich Baur Institute, LMU University Hospital, LMU Munich; Department of Neurology (M. Marziniak), Kbo-Isar-Amper-Hospital Munich East; Department of Neurology (M. Mäurer), Klinikum Würzburg Mitte gGmbH, Standort Juliusspital; Department of Neurology (F.S.), LMU University Hospital, LMU, Munich; Department of Neurology (B.S.), DKD HELIOS Klinik Wiesbaden; Department of Paediatric and Adolescent Medicine (C. Steen), St Joseph Hospital, Berlin; Department of Neurology (A.T.), HELIOS Klinikum Erfurt; and Neurologische Praxis Dres. Wessely (L.W.), Menden, Germany
| | - Bertold Schrank
- From the Department of Neurology (S.R., S.S., B.H., L.A., C. Sommer, K.D.); Department of Anaesthesiology, Intensive Care, Emergency and Pain Medicine Centre for Interdisciplinary Pain Medicine (B.H.); Institute of Experimental Biomedicine (R.A., A.Z.), University Hospital Würzburg; German Center for Neurodegenerative Diseases (DZNE) Berlin (F.A.A., H.P.); Department of Neurology and Experimental Neurology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität Berlin; Section Translational Neuroimmunology (D.B., C.G.), Department of Neurology, Jena University Hospital; Department of Neurology (C.D.), University Medical Center of the Johannes Gutenberg University, Mainz; Department of Neurology (J.D., Z.E.), University Hospital Ulm; Department of Neurology (T.H., H.T.), Therapiezentrum Burgau; Department of Neurology (S.K.), Friedrich Baur Institute, LMU University Hospital, LMU Munich; Department of Neurology (M. Marziniak), Kbo-Isar-Amper-Hospital Munich East; Department of Neurology (M. Mäurer), Klinikum Würzburg Mitte gGmbH, Standort Juliusspital; Department of Neurology (F.S.), LMU University Hospital, LMU, Munich; Department of Neurology (B.S.), DKD HELIOS Klinik Wiesbaden; Department of Paediatric and Adolescent Medicine (C. Steen), St Joseph Hospital, Berlin; Department of Neurology (A.T.), HELIOS Klinikum Erfurt; and Neurologische Praxis Dres. Wessely (L.W.), Menden, Germany
| | - Claudia Steen
- From the Department of Neurology (S.R., S.S., B.H., L.A., C. Sommer, K.D.); Department of Anaesthesiology, Intensive Care, Emergency and Pain Medicine Centre for Interdisciplinary Pain Medicine (B.H.); Institute of Experimental Biomedicine (R.A., A.Z.), University Hospital Würzburg; German Center for Neurodegenerative Diseases (DZNE) Berlin (F.A.A., H.P.); Department of Neurology and Experimental Neurology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität Berlin; Section Translational Neuroimmunology (D.B., C.G.), Department of Neurology, Jena University Hospital; Department of Neurology (C.D.), University Medical Center of the Johannes Gutenberg University, Mainz; Department of Neurology (J.D., Z.E.), University Hospital Ulm; Department of Neurology (T.H., H.T.), Therapiezentrum Burgau; Department of Neurology (S.K.), Friedrich Baur Institute, LMU University Hospital, LMU Munich; Department of Neurology (M. Marziniak), Kbo-Isar-Amper-Hospital Munich East; Department of Neurology (M. Mäurer), Klinikum Würzburg Mitte gGmbH, Standort Juliusspital; Department of Neurology (F.S.), LMU University Hospital, LMU, Munich; Department of Neurology (B.S.), DKD HELIOS Klinik Wiesbaden; Department of Paediatric and Adolescent Medicine (C. Steen), St Joseph Hospital, Berlin; Department of Neurology (A.T.), HELIOS Klinikum Erfurt; and Neurologische Praxis Dres. Wessely (L.W.), Menden, Germany
| | - Helena Teichtinger
- From the Department of Neurology (S.R., S.S., B.H., L.A., C. Sommer, K.D.); Department of Anaesthesiology, Intensive Care, Emergency and Pain Medicine Centre for Interdisciplinary Pain Medicine (B.H.); Institute of Experimental Biomedicine (R.A., A.Z.), University Hospital Würzburg; German Center for Neurodegenerative Diseases (DZNE) Berlin (F.A.A., H.P.); Department of Neurology and Experimental Neurology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität Berlin; Section Translational Neuroimmunology (D.B., C.G.), Department of Neurology, Jena University Hospital; Department of Neurology (C.D.), University Medical Center of the Johannes Gutenberg University, Mainz; Department of Neurology (J.D., Z.E.), University Hospital Ulm; Department of Neurology (T.H., H.T.), Therapiezentrum Burgau; Department of Neurology (S.K.), Friedrich Baur Institute, LMU University Hospital, LMU Munich; Department of Neurology (M. Marziniak), Kbo-Isar-Amper-Hospital Munich East; Department of Neurology (M. Mäurer), Klinikum Würzburg Mitte gGmbH, Standort Juliusspital; Department of Neurology (F.S.), LMU University Hospital, LMU, Munich; Department of Neurology (B.S.), DKD HELIOS Klinik Wiesbaden; Department of Paediatric and Adolescent Medicine (C. Steen), St Joseph Hospital, Berlin; Department of Neurology (A.T.), HELIOS Klinikum Erfurt; and Neurologische Praxis Dres. Wessely (L.W.), Menden, Germany
| | - Andrea Thieme
- From the Department of Neurology (S.R., S.S., B.H., L.A., C. Sommer, K.D.); Department of Anaesthesiology, Intensive Care, Emergency and Pain Medicine Centre for Interdisciplinary Pain Medicine (B.H.); Institute of Experimental Biomedicine (R.A., A.Z.), University Hospital Würzburg; German Center for Neurodegenerative Diseases (DZNE) Berlin (F.A.A., H.P.); Department of Neurology and Experimental Neurology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität Berlin; Section Translational Neuroimmunology (D.B., C.G.), Department of Neurology, Jena University Hospital; Department of Neurology (C.D.), University Medical Center of the Johannes Gutenberg University, Mainz; Department of Neurology (J.D., Z.E.), University Hospital Ulm; Department of Neurology (T.H., H.T.), Therapiezentrum Burgau; Department of Neurology (S.K.), Friedrich Baur Institute, LMU University Hospital, LMU Munich; Department of Neurology (M. Marziniak), Kbo-Isar-Amper-Hospital Munich East; Department of Neurology (M. Mäurer), Klinikum Würzburg Mitte gGmbH, Standort Juliusspital; Department of Neurology (F.S.), LMU University Hospital, LMU, Munich; Department of Neurology (B.S.), DKD HELIOS Klinik Wiesbaden; Department of Paediatric and Adolescent Medicine (C. Steen), St Joseph Hospital, Berlin; Department of Neurology (A.T.), HELIOS Klinikum Erfurt; and Neurologische Praxis Dres. Wessely (L.W.), Menden, Germany
| | - Lena Wessely
- From the Department of Neurology (S.R., S.S., B.H., L.A., C. Sommer, K.D.); Department of Anaesthesiology, Intensive Care, Emergency and Pain Medicine Centre for Interdisciplinary Pain Medicine (B.H.); Institute of Experimental Biomedicine (R.A., A.Z.), University Hospital Würzburg; German Center for Neurodegenerative Diseases (DZNE) Berlin (F.A.A., H.P.); Department of Neurology and Experimental Neurology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität Berlin; Section Translational Neuroimmunology (D.B., C.G.), Department of Neurology, Jena University Hospital; Department of Neurology (C.D.), University Medical Center of the Johannes Gutenberg University, Mainz; Department of Neurology (J.D., Z.E.), University Hospital Ulm; Department of Neurology (T.H., H.T.), Therapiezentrum Burgau; Department of Neurology (S.K.), Friedrich Baur Institute, LMU University Hospital, LMU Munich; Department of Neurology (M. Marziniak), Kbo-Isar-Amper-Hospital Munich East; Department of Neurology (M. Mäurer), Klinikum Würzburg Mitte gGmbH, Standort Juliusspital; Department of Neurology (F.S.), LMU University Hospital, LMU, Munich; Department of Neurology (B.S.), DKD HELIOS Klinik Wiesbaden; Department of Paediatric and Adolescent Medicine (C. Steen), St Joseph Hospital, Berlin; Department of Neurology (A.T.), HELIOS Klinikum Erfurt; and Neurologische Praxis Dres. Wessely (L.W.), Menden, Germany
| | - Alma Zernecke
- From the Department of Neurology (S.R., S.S., B.H., L.A., C. Sommer, K.D.); Department of Anaesthesiology, Intensive Care, Emergency and Pain Medicine Centre for Interdisciplinary Pain Medicine (B.H.); Institute of Experimental Biomedicine (R.A., A.Z.), University Hospital Würzburg; German Center for Neurodegenerative Diseases (DZNE) Berlin (F.A.A., H.P.); Department of Neurology and Experimental Neurology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität Berlin; Section Translational Neuroimmunology (D.B., C.G.), Department of Neurology, Jena University Hospital; Department of Neurology (C.D.), University Medical Center of the Johannes Gutenberg University, Mainz; Department of Neurology (J.D., Z.E.), University Hospital Ulm; Department of Neurology (T.H., H.T.), Therapiezentrum Burgau; Department of Neurology (S.K.), Friedrich Baur Institute, LMU University Hospital, LMU Munich; Department of Neurology (M. Marziniak), Kbo-Isar-Amper-Hospital Munich East; Department of Neurology (M. Mäurer), Klinikum Würzburg Mitte gGmbH, Standort Juliusspital; Department of Neurology (F.S.), LMU University Hospital, LMU, Munich; Department of Neurology (B.S.), DKD HELIOS Klinik Wiesbaden; Department of Paediatric and Adolescent Medicine (C. Steen), St Joseph Hospital, Berlin; Department of Neurology (A.T.), HELIOS Klinikum Erfurt; and Neurologische Praxis Dres. Wessely (L.W.), Menden, Germany
| | - Claudia Sommer
- From the Department of Neurology (S.R., S.S., B.H., L.A., C. Sommer, K.D.); Department of Anaesthesiology, Intensive Care, Emergency and Pain Medicine Centre for Interdisciplinary Pain Medicine (B.H.); Institute of Experimental Biomedicine (R.A., A.Z.), University Hospital Würzburg; German Center for Neurodegenerative Diseases (DZNE) Berlin (F.A.A., H.P.); Department of Neurology and Experimental Neurology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität Berlin; Section Translational Neuroimmunology (D.B., C.G.), Department of Neurology, Jena University Hospital; Department of Neurology (C.D.), University Medical Center of the Johannes Gutenberg University, Mainz; Department of Neurology (J.D., Z.E.), University Hospital Ulm; Department of Neurology (T.H., H.T.), Therapiezentrum Burgau; Department of Neurology (S.K.), Friedrich Baur Institute, LMU University Hospital, LMU Munich; Department of Neurology (M. Marziniak), Kbo-Isar-Amper-Hospital Munich East; Department of Neurology (M. Mäurer), Klinikum Würzburg Mitte gGmbH, Standort Juliusspital; Department of Neurology (F.S.), LMU University Hospital, LMU, Munich; Department of Neurology (B.S.), DKD HELIOS Klinik Wiesbaden; Department of Paediatric and Adolescent Medicine (C. Steen), St Joseph Hospital, Berlin; Department of Neurology (A.T.), HELIOS Klinikum Erfurt; and Neurologische Praxis Dres. Wessely (L.W.), Menden, Germany
| | - Kathrin Doppler
- From the Department of Neurology (S.R., S.S., B.H., L.A., C. Sommer, K.D.); Department of Anaesthesiology, Intensive Care, Emergency and Pain Medicine Centre for Interdisciplinary Pain Medicine (B.H.); Institute of Experimental Biomedicine (R.A., A.Z.), University Hospital Würzburg; German Center for Neurodegenerative Diseases (DZNE) Berlin (F.A.A., H.P.); Department of Neurology and Experimental Neurology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität Berlin; Section Translational Neuroimmunology (D.B., C.G.), Department of Neurology, Jena University Hospital; Department of Neurology (C.D.), University Medical Center of the Johannes Gutenberg University, Mainz; Department of Neurology (J.D., Z.E.), University Hospital Ulm; Department of Neurology (T.H., H.T.), Therapiezentrum Burgau; Department of Neurology (S.K.), Friedrich Baur Institute, LMU University Hospital, LMU Munich; Department of Neurology (M. Marziniak), Kbo-Isar-Amper-Hospital Munich East; Department of Neurology (M. Mäurer), Klinikum Würzburg Mitte gGmbH, Standort Juliusspital; Department of Neurology (F.S.), LMU University Hospital, LMU, Munich; Department of Neurology (B.S.), DKD HELIOS Klinik Wiesbaden; Department of Paediatric and Adolescent Medicine (C. Steen), St Joseph Hospital, Berlin; Department of Neurology (A.T.), HELIOS Klinikum Erfurt; and Neurologische Praxis Dres. Wessely (L.W.), Menden, Germany
| |
Collapse
|
3
|
Marti Z, Ruder J, Thomas OG, Bronge M, De La Parra Soto L, Grönlund H, Olsson T, Martin R. Enhanced and cross-reactive in vitro memory B cell response against Epstein-Barr virus nuclear antigen 1 in multiple sclerosis. Front Immunol 2024; 15:1334720. [PMID: 39257578 PMCID: PMC11385009 DOI: 10.3389/fimmu.2024.1334720] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2023] [Accepted: 04/04/2024] [Indexed: 09/12/2024] Open
Abstract
Multiple sclerosis (MS) is a prototypical autoimmune disease of the central nervous system (CNS). In addition to CD4+ T cells, memory B cells are now recognized as a critical cell type in the disease. This is underlined by the fact that the best-characterized environmental risk factor for MS is the Epstein-Barr virus (EBV), which can infect and persist in memory B cells throughout life. Several studies have identified changes in anti-EBV immunity in patients with MS. Examples include elevated titers of anti-EBV nuclear antigen 1 (EBNA1) antibodies, interactions of these with the MS-associated HLA-DR15 haplotype, and molecular mimicry with MS autoantigens like myelin basic protein (MBP), anoctamin-2 (ANO2), glial cell adhesion molecule (GlialCAM), and alpha-crystallin B (CRYAB). In this study, we employ a simple in vitro assay to examine the memory B cell antibody repertoire in MS patients and healthy controls. We replicate previous serological data from MS patients demonstrating an increased secretion of anti-EBNA1380-641 IgG in cell culture supernatants, as well as a positive correlation of these levels with autoantibodies against GlialCAM262-416 and ANO21-275. For EBNA1380-641 and ANO21-275, we provide additional evidence suggesting antibody cross-reactivity between the two targets. Further, we show that two efficacious MS treatments - natalizumab (NAT) and autologous hematopoietic stem cell transplantation (aHSCT) - are associated with distinct changes in the EBNA1-directed B cell response and that these alterations can be attributed to the unique mechanisms of action of these therapies. Using an in vitro system, our study confirms MS-associated changes in the anti-EBNA1 memory B cell response, EBNA1380-641 antibody cross-reactivity with ANO21-275, and reveals treatment-associated changes in the immunoglobulin repertoire in MS.
Collapse
Affiliation(s)
- Zoe Marti
- Institute of Experimental Immunology, University of Zurich, Zurich, Switzerland
- Research and Development, Cellerys, Schlieren, Switzerland
- Department of Neuroimmunology and Multiple Sclerosis Research, University Hospital Zurich, Zurich, Switzerland
| | - Josefine Ruder
- Department of Neuroimmunology and Multiple Sclerosis Research, University Hospital Zurich, Zurich, Switzerland
| | - Olivia G Thomas
- Therapeutic Immune Design Unit, Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Mattias Bronge
- Therapeutic Immune Design Unit, Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
- Department of Neurology, Karolinska University Hospital, Stockholm, Sweden
| | - Lorenzo De La Parra Soto
- Therapeutic Immune Design Unit, Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
- Department of Medical Biochemistry and Microbiology, Uppsala University, Uppsala, Sweden
| | - Hans Grönlund
- Therapeutic Immune Design Unit, Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Tomas Olsson
- Neuroimmunology Unit, Department of Clinical Neurocience, Karolinska Institutet, Stockholm, Sweden
| | - Roland Martin
- Institute of Experimental Immunology, University of Zurich, Zurich, Switzerland
- Research and Development, Cellerys, Schlieren, Switzerland
- Department of Neuroimmunology and Multiple Sclerosis Research, University Hospital Zurich, Zurich, Switzerland
- Therapeutic Immune Design Unit, Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
4
|
Nadat F, Clark B. Forming a new perspective: Post-structural approaches to determination of donor compatibility and post-transplant assessment of allograft health. Int J Immunogenet 2024; 51:195-205. [PMID: 38711186 DOI: 10.1111/iji.12675] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2024] [Accepted: 03/25/2024] [Indexed: 05/08/2024]
Abstract
The purpose of this review is to encourage a new perspective on the question of donor-recipient compatibility and post-transplant assessment of graft health based on functional measures. The premise is that we should be better sighted on what (and how) the immune system responds toward rather than what is merely there. Continuance of the pursuit of further and better definition of antigens and antibodies is not however discouraged but seen as necessary to improved understanding of the structural correlates of functional immunity. There currently exists, in the opinion of the authors, an opportunity for histocompatibility and immunogenetics laboratories to develop and widen their scope of involvement into these new areas of laboratory activity in support and to the benefit of the transplant programmes they serve.
Collapse
Affiliation(s)
- Fatima Nadat
- Functional Assessment of Transplant Immunology Group, St James's University Hospital, Leeds, UK
| | - Brendan Clark
- Functional Assessment of Transplant Immunology Group, St James's University Hospital, Leeds, UK
| |
Collapse
|
5
|
Lee C, Imran I, Thomas S, Nouri-Shirazi M. A comprehensive method for the phenotypical and functional characterization of recalled human memory B and T cells specific to vaccine antigens. J Immunol Methods 2024; 527:113650. [PMID: 38428517 DOI: 10.1016/j.jim.2024.113650] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2023] [Revised: 02/10/2024] [Accepted: 02/25/2024] [Indexed: 03/03/2024]
Abstract
Current methodologies for assessing vaccine effectiveness and longevity primarily center on measuring vaccine-induced neutralizing antibodies in serum or plasma. However, these methods overlook additional parameters such as the presence of memory B cells, even as antibody levels wane, and the pivotal role played by memory T cells in shaping antigen-specific memory B cell responses. Several studies have employed a combination of polyclonal activators, such as CpG and R848, along with various cytokines to provoke the recall of memory B cells from peripheral blood mononuclear cells (PBMCs) into antibody-secreting cells (ASCs). Other studies have examined the use of live attenuated viruses to stimulate antigen-specific memory T cells within PBMCs into effector T cells that produce Th1/Th2 cytokines. However, these studies have not fully elucidated the distinct effects of these polyclonal activators on individual subsets, nor have they evaluated whether the vaccine antigen alone is sufficient to trigger the recall of memory T cells. Thus, in this study, we directly compared the capacity of two B cell polyclonal activators to induce the transition of existing vaccine-specific memory cells present in peripheral blood samples into ASCs. Simultaneously, we also assessed the transition of existing memory T cells into effector subsets in response to vaccine antigens. Our findings demonstrate that both polyclonal activator combinations, CpG with IL-6 and IL-15, as well as R848 with IL-2, effectively induce the terminal differentiation of memory B cells into ASCs. Notably, CpG treatment preferentially expanded naïve and non-class-switched B cells, while R848 expanded class-switched memory cells, plasmablasts, and plasma cells. Consequently, R848 treatment led to a greater overall production of total and antigen-specific IgG immunoglobulins. Additionally, the exposure of isolated PBMCs to vaccine antigens alone proved sufficient for recalling the rare antigen-specific memory T cells into effector subsets, predominantly consisting of IFN-γ-producing CD4 T cells and TNF-β-producing CD8 T cells. This study not only establishes a rationale for the selection of methods to expand and detect antigen-specific lymphocyte subsets but also presents a means to quantify vaccine effectiveness by correlating serum antibody levels with preexisting memory cells within peripheral blood samples.
Collapse
Affiliation(s)
- Czdari Lee
- Florida Atlantic University, Charles E. Schmidt College of Medicine, Department of Medicine, 777 Glades Road, PO Box 3091, Boca Raton, FL 33431, USA
| | - Imtisal Imran
- Florida Atlantic University, Charles E. Schmidt College of Medicine, Department of Medicine, 777 Glades Road, PO Box 3091, Boca Raton, FL 33431, USA
| | - Sara Thomas
- Florida Atlantic University, Charles E. Schmidt College of Medicine, Department of Medicine, 777 Glades Road, PO Box 3091, Boca Raton, FL 33431, USA
| | - Mahyar Nouri-Shirazi
- Florida Atlantic University, Charles E. Schmidt College of Medicine, Department of Medicine, 777 Glades Road, PO Box 3091, Boca Raton, FL 33431, USA.
| |
Collapse
|
6
|
Addetia A, Piccoli L, Case JB, Park YJ, Beltramello M, Guarino B, Dang H, de Melo GD, Pinto D, Sprouse K, Scheaffer SM, Bassi J, Silacci-Fregni C, Muoio F, Dini M, Vincenzetti L, Acosta R, Johnson D, Subramanian S, Saliba C, Giurdanella M, Lombardo G, Leoni G, Culap K, McAlister C, Rajesh A, Dellota E, Zhou J, Farhat N, Bohan D, Noack J, Chen A, Lempp FA, Quispe J, Kergoat L, Larrous F, Cameroni E, Whitener B, Giannini O, Cippà P, Ceschi A, Ferrari P, Franzetti-Pellanda A, Biggiogero M, Garzoni C, Zappi S, Bernasconi L, Kim MJ, Rosen LE, Schnell G, Czudnochowski N, Benigni F, Franko N, Logue JK, Yoshiyama C, Stewart C, Chu H, Bourhy H, Schmid MA, Purcell LA, Snell G, Lanzavecchia A, Diamond MS, Corti D, Veesler D. Neutralization, effector function and immune imprinting of Omicron variants. Nature 2023; 621:592-601. [PMID: 37648855 PMCID: PMC10511321 DOI: 10.1038/s41586-023-06487-6] [Citation(s) in RCA: 74] [Impact Index Per Article: 74.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2023] [Accepted: 07/27/2023] [Indexed: 09/01/2023]
Abstract
Currently circulating SARS-CoV-2 variants have acquired convergent mutations at hot spots in the receptor-binding domain1 (RBD) of the spike protein. The effects of these mutations on viral infection and transmission and the efficacy of vaccines and therapies remains poorly understood. Here we demonstrate that recently emerged BQ.1.1 and XBB.1.5 variants bind host ACE2 with high affinity and promote membrane fusion more efficiently than earlier Omicron variants. Structures of the BQ.1.1, XBB.1 and BN.1 RBDs bound to the fragment antigen-binding region of the S309 antibody (the parent antibody for sotrovimab) and human ACE2 explain the preservation of antibody binding through conformational selection, altered ACE2 recognition and immune evasion. We show that sotrovimab binds avidly to all Omicron variants, promotes Fc-dependent effector functions and protects mice challenged with BQ.1.1 and hamsters challenged with XBB.1.5. Vaccine-elicited human plasma antibodies cross-react with and trigger effector functions against current Omicron variants, despite a reduced neutralizing activity, suggesting a mechanism of protection against disease, exemplified by S309. Cross-reactive RBD-directed human memory B cells remained dominant even after two exposures to Omicron spikes, underscoring the role of persistent immune imprinting.
Collapse
Affiliation(s)
- Amin Addetia
- Department of Biochemistry, University of Washington, Seattle, WA, USA
| | | | - James Brett Case
- Department of Medicine, Washington University School of Medicine, St Louis, MO, USA
| | - Young-Jun Park
- Department of Biochemistry, University of Washington, Seattle, WA, USA
| | | | | | - Ha Dang
- Vir Biotechnology, San Francisco, CA, USA
| | - Guilherme Dias de Melo
- Institut Pasteur, Université Paris Cité, Lyssavirus Epidemiology and Neuropathology Unit, Paris, France
| | | | - Kaitlin Sprouse
- Department of Biochemistry, University of Washington, Seattle, WA, USA
| | - Suzanne M Scheaffer
- Department of Medicine, Washington University School of Medicine, St Louis, MO, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | - Jiayi Zhou
- Vir Biotechnology, San Francisco, CA, USA
| | | | - Dana Bohan
- Vir Biotechnology, San Francisco, CA, USA
| | | | - Alex Chen
- Vir Biotechnology, San Francisco, CA, USA
| | | | - Joel Quispe
- Department of Biochemistry, University of Washington, Seattle, WA, USA
| | - Lauriane Kergoat
- Institut Pasteur, Université Paris Cité, Lyssavirus Epidemiology and Neuropathology Unit, Paris, France
| | - Florence Larrous
- Institut Pasteur, Université Paris Cité, Lyssavirus Epidemiology and Neuropathology Unit, Paris, France
| | | | - Bradley Whitener
- Department of Medicine, Washington University School of Medicine, St Louis, MO, USA
| | - Olivier Giannini
- Faculty of Biomedical Sciences, Università della Svizzera italiana, Lugano, Switzerland
- Department of Medicine, Ente Ospedaliero Cantonale, Bellinzona, Switzerland
| | - Pietro Cippà
- Faculty of Biomedical Sciences, Università della Svizzera italiana, Lugano, Switzerland
- Department of Medicine, Ente Ospedaliero Cantonale, Bellinzona, Switzerland
- Division of Nephrology, Ente Ospedaliero Cantonale, Lugano, Switzerland
| | - Alessandro Ceschi
- Faculty of Biomedical Sciences, Università della Svizzera italiana, Lugano, Switzerland
- Clinical Trial Unit, Ente Ospedaliero Cantonale, Lugano, Switzerland
- Division of Clinical Pharmacology and Toxicology, Institute of Pharmacological Sciences of Southern Switzerland, Ente Ospedaliero Cantonale, Lugano, Switzerland
- Department of Clinical Pharmacology and Toxicology, University Hospital Zurich, Zurich, Switzerland
| | - Paolo Ferrari
- Faculty of Biomedical Sciences, Università della Svizzera italiana, Lugano, Switzerland
- Division of Nephrology, Ente Ospedaliero Cantonale, Lugano, Switzerland
- Clinical School, University of New South Wales, Sydney, New South Wales, Australia
| | | | - Maira Biggiogero
- Clinical Research Unit, Clinica Luganese Moncucco, Lugano, Switzerland
| | - Christian Garzoni
- Clinic of Internal Medicine and Infectious Diseases, Clinica Luganese Moncucco, Lugano, Switzerland
| | - Stephanie Zappi
- Division of Nephrology, Cantonal Hospital Aarau, Aarau, Switzerland
| | - Luca Bernasconi
- Institute of Laboratory Medicine, Cantonal Hospital Aarau, Aarau, Switzerland
| | - Min Jeong Kim
- Division of Nephrology, Cantonal Hospital Aarau, Aarau, Switzerland
| | | | | | | | | | - Nicholas Franko
- Division of Allergy and Infectious Diseases, University of Washington, Seattle, WA, USA
| | - Jennifer K Logue
- Division of Allergy and Infectious Diseases, University of Washington, Seattle, WA, USA
| | | | - Cameron Stewart
- Department of Biochemistry, University of Washington, Seattle, WA, USA
| | - Helen Chu
- Division of Allergy and Infectious Diseases, University of Washington, Seattle, WA, USA
| | - Hervé Bourhy
- Institut Pasteur, Université Paris Cité, Lyssavirus Epidemiology and Neuropathology Unit, Paris, France
| | | | | | | | | | - Michael S Diamond
- Department of Medicine, Washington University School of Medicine, St Louis, MO, USA.
- Department of Pathology and Immunology, Washington University School of Medicine, St Louis, MO, USA.
- Department of Molecular Microbiology, Washington University School of Medicine, St Louis, MO, USA.
- Andrew M. and Jane M. Bursky Center for Human Immunology and Immunotherapy Programs, Washington University School of Medicine, St Louis, MO, USA.
- Center for Vaccines and Immunity to Microbial Pathogens, Washington University School of Medicine, St Louis, MO, USA.
| | | | - David Veesler
- Department of Biochemistry, University of Washington, Seattle, WA, USA.
- Howard Hughes Medical Institute, University of Washington, Seattle, WA, USA.
| |
Collapse
|
7
|
Stensland ZC, Magera CA, Broncucia H, Gomez BD, Rios-Guzman NM, Wells KL, Nicholas CA, Rihanek M, Hunter MJ, Toole KP, Gottlieb PA, Smith MJ. Identification of an anergic BND cell-derived activated B cell population (BND2) in young-onset type 1 diabetes patients. J Exp Med 2023; 220:e20221604. [PMID: 37184563 PMCID: PMC10192302 DOI: 10.1084/jem.20221604] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2022] [Revised: 03/15/2023] [Accepted: 04/21/2023] [Indexed: 05/16/2023] Open
Abstract
Recent evidence suggests a role for B cells in the pathogenesis of young-onset type 1 diabetes (T1D), wherein rapid progression occurs. However, little is known regarding the specificity, phenotype, and function of B cells in young-onset T1D. We performed a cross-sectional analysis comparing insulin-reactive to tetanus-reactive B cells in the blood of T1D and controls using mass cytometry. Unsupervised clustering revealed the existence of a highly activated B cell subset we term BND2 that falls within the previously defined anergic BND subset. We found a specific increase in the frequency of insulin-reactive BND2 cells in the blood of young-onset T1D donors, which was further enriched in the pancreatic lymph nodes of T1D donors. The frequency of insulin-binding BND2 cells correlated with anti-insulin autoantibody levels. We demonstrate BND2 cells are pre-plasma cells and can likely act as APCs to T cells. These findings identify an antigen-specific B cell subset that may play a role in the rapid progression of young-onset T1D.
Collapse
Affiliation(s)
- Zachary C. Stensland
- Department of Pediatrics, University of Colorado School of Medicine, Aurora, CO, USA
- Barbara Davis Center for Diabetes, University of Colorado School of Medicine, Aurora, CO, USA
| | - Christopher A. Magera
- Barbara Davis Center for Diabetes, University of Colorado School of Medicine, Aurora, CO, USA
| | - Hali Broncucia
- Barbara Davis Center for Diabetes, University of Colorado School of Medicine, Aurora, CO, USA
| | - Brittany D. Gomez
- Department of Immunology and Microbiology, University of Colorado School of Medicine, Aurora, CO, USA
| | - Nasha M. Rios-Guzman
- Barbara Davis Center for Diabetes, University of Colorado School of Medicine, Aurora, CO, USA
- Department of Immunology and Microbiology, University of Colorado School of Medicine, Aurora, CO, USA
| | - Kristen L. Wells
- Barbara Davis Center for Diabetes, University of Colorado School of Medicine, Aurora, CO, USA
| | - Catherine A. Nicholas
- Barbara Davis Center for Diabetes, University of Colorado School of Medicine, Aurora, CO, USA
- Department of Biochemistry and Molecular Genetics, University of Colorado School of Medicine, Aurora, CO, USA
| | - Marynette Rihanek
- Barbara Davis Center for Diabetes, University of Colorado School of Medicine, Aurora, CO, USA
| | - Maya J. Hunter
- Barbara Davis Center for Diabetes, University of Colorado School of Medicine, Aurora, CO, USA
| | - Kevin P. Toole
- Barbara Davis Center for Diabetes, University of Colorado School of Medicine, Aurora, CO, USA
| | - Peter A. Gottlieb
- Department of Pediatrics, University of Colorado School of Medicine, Aurora, CO, USA
- Barbara Davis Center for Diabetes, University of Colorado School of Medicine, Aurora, CO, USA
| | - Mia J. Smith
- Department of Pediatrics, University of Colorado School of Medicine, Aurora, CO, USA
- Barbara Davis Center for Diabetes, University of Colorado School of Medicine, Aurora, CO, USA
- Department of Immunology and Microbiology, University of Colorado School of Medicine, Aurora, CO, USA
| |
Collapse
|
8
|
Chunder R, Schropp V, Marzin M, Amor S, Kuerten S. A Dual Role of Osteopontin in Modifying B Cell Responses. Biomedicines 2023; 11:1969. [PMID: 37509608 PMCID: PMC10377065 DOI: 10.3390/biomedicines11071969] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2023] [Revised: 07/06/2023] [Accepted: 07/07/2023] [Indexed: 07/30/2023] Open
Abstract
The occurrence of B cell aggregates within the central nervous system (CNS) has prompted the investigation of the potential sources of pathogenic B cell and T cell responses in a subgroup of secondary progressive multiple sclerosis (MS) patients. Nevertheless, the expression profile of molecules associated with these aggregates and their role in aggregate development and persistence is poorly described. Here, we focused on the expression pattern of osteopontin (OPN), which is a well-described cytokine, in MS brain tissue. Autopsied brain sections from MS cases with and without B cell pathology were screened for the presence of CD20+ B cell aggregates and co-expression of OPN. To demonstrate the effect of OPN on B cells, flow cytometry, ELISA and in vitro aggregation assays were conducted using the peripheral blood of healthy volunteers. Although OPN was expressed in MS brain tissue independent of B cell pathology, it was also highly expressed within B cell aggregates. In vitro studies demonstrated that OPN downregulated the co-stimulatory molecules CD80 and CD86 on B cells. OPN-treated B cells produced significantly lower amounts of IL-6. However, OPN-treated B cells also exhibited a higher tendency to form homotypic cell aggregates in vitro. Taken together, our data indicate a conflicting role of OPN in modulating B cell responses.
Collapse
Affiliation(s)
- Rittika Chunder
- Faculty of Medicine, Institute of Neuroanatomy, University of Bonn, 53115 Bonn, Germany
- University Hospital Bonn, 53127 Bonn, Germany
- Institute of Anatomy and Cell Biology, Friedrich-Alexander-Universität Erlangen-Nürnberg, 91054 Erlangen, Germany
| | - Verena Schropp
- Faculty of Medicine, Institute of Neuroanatomy, University of Bonn, 53115 Bonn, Germany
- University Hospital Bonn, 53127 Bonn, Germany
- Institute of Anatomy and Cell Biology, Friedrich-Alexander-Universität Erlangen-Nürnberg, 91054 Erlangen, Germany
| | - Manuel Marzin
- Department of Pathology, Amsterdam University Medical Center, 1007 MB Amsterdam, The Netherlands
| | - Sandra Amor
- Department of Pathology, Amsterdam University Medical Center, 1007 MB Amsterdam, The Netherlands
- Institute of Anatomy, Rostock University Medical Center, 18057 Rostock, Germany
| | - Stefanie Kuerten
- Faculty of Medicine, Institute of Neuroanatomy, University of Bonn, 53115 Bonn, Germany
- University Hospital Bonn, 53127 Bonn, Germany
- Institute of Anatomy and Cell Biology, Friedrich-Alexander-Universität Erlangen-Nürnberg, 91054 Erlangen, Germany
| |
Collapse
|
9
|
Quan C, Liu Q, Yu L, Li C, Nie K, Ding G, Zhou H, Wang X, Sun W, Wang H, Yue M, Wei L, Zheng W, Lyu Q, Xing W, Zhang Z, Carr MJ, Zhang H, Shi W. SFTSV infection is associated with transient overproliferation of monoclonal lambda-type plasma cells. iScience 2023; 26:106799. [PMID: 37250798 PMCID: PMC10212991 DOI: 10.1016/j.isci.2023.106799] [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: 10/08/2022] [Revised: 04/07/2023] [Accepted: 04/28/2023] [Indexed: 05/31/2023] Open
Abstract
The impairment of antibody-mediated immunity is a major factor associated with fatal cases of severe fever with thrombocytopenia syndrome (SFTS). By collating the clinical diagnosis reports of 30 SFTS cases, we discovered the overproliferation of monoclonal plasma cells (MCP cells, CD38+cLambda+cKappa-) in bone marrow, which has only been reported previously in multiple myeloma. The ratio of CD38+cLambda+ versus CD38+cKappa+ in SFTS cases with MCP cells was significantly higher than that in normal cases. MCP cells presented transient expression in the bone marrow, which was distinctly different from multiple myeloma. Moreover, the SFTS patients with MCP cells had higher clinical severity. Further, the overproliferation of MCP cells was also observed in SFTS virus (SFTSV)-infected mice with lethal infectious doses. Together, SFTSV infection induces transient overproliferation of monoclonal lambda-type plasma cells, which have important implications for the study of SFTSV pathogenesis, prognosis, and the rational development of therapeutics.
Collapse
Affiliation(s)
- Chuansong Quan
- Department of Infectious Disease, The Second Affiliated Hospital of Shandong First Medical University, Taian 271000, China
- Key Laboratory of Emerging Infectious Diseases in Universities of Shandong, School of Clinical and Basic Medical Sciences, Shandong First Medical University & Shandong Academy of Medical Sciences, Taian, 271000, China
| | - Qinghua Liu
- Department of Clinical Laboratory, The Second Affiliated Hospital of Shandong First Medical University, Taian 271000, China
| | - Lijuan Yu
- Key Laboratory of Emerging Infectious Diseases in Universities of Shandong, School of Clinical and Basic Medical Sciences, Shandong First Medical University & Shandong Academy of Medical Sciences, Taian, 271000, China
| | - Chunjing Li
- Department of Hematology, The Second Affiliated Hospital of Shandong First Medical University, Taian 271000, China
| | - Kaixiao Nie
- Key Laboratory of Emerging Infectious Diseases in Universities of Shandong, School of Clinical and Basic Medical Sciences, Shandong First Medical University & Shandong Academy of Medical Sciences, Taian, 271000, China
| | - Guoyong Ding
- School of Public Health, Shandong First Medical University & Shandong Academy of Medical Sciences, Ji’nan 250117, China
| | - Hong Zhou
- Key Laboratory of Emerging Infectious Diseases in Universities of Shandong, School of Clinical and Basic Medical Sciences, Shandong First Medical University & Shandong Academy of Medical Sciences, Taian, 271000, China
| | - Xinli Wang
- Department of Pathology, The Second Affiliated Hospital of Shandong First Medical University, Taian 271000, China
| | - Wenwen Sun
- Department of Pathology, The Second Affiliated Hospital of Shandong First Medical University, Taian 271000, China
| | - Huiliang Wang
- Department of Infectious Disease, The Second Affiliated Hospital of Shandong First Medical University, Taian 271000, China
| | - Maokui Yue
- Department of Critical Care Medicine, The Second Affiliated Hospital of Shandong First Medical University, Taian 271000, China
| | - Li Wei
- Department of Respiratory Medicine, The Second Affiliated Hospital of Shandong First Medical University, Taian 271000, China
| | - Wenjun Zheng
- Key Laboratory of Emerging Infectious Diseases in Universities of Shandong, School of Clinical and Basic Medical Sciences, Shandong First Medical University & Shandong Academy of Medical Sciences, Taian, 271000, China
| | - Qiang Lyu
- School of Public Health, Shandong First Medical University & Shandong Academy of Medical Sciences, Ji’nan 250117, China
| | - Weijia Xing
- School of Public Health, Shandong First Medical University & Shandong Academy of Medical Sciences, Ji’nan 250117, China
| | - Zhenjie Zhang
- Key Laboratory of Emerging Infectious Diseases in Universities of Shandong, School of Clinical and Basic Medical Sciences, Shandong First Medical University & Shandong Academy of Medical Sciences, Taian, 271000, China
| | - Michael J. Carr
- National Virus Reference Laboratory, School of Medicine, University College Dublin, Dublin 4, Ireland
- International Collaboration Unit, International Institute for Zoonosis Control, Hokkaido University, N20 W10 Kita-ku, Sapporo 001-0020, Japan
| | - Hong Zhang
- Department of Infectious Disease, The Second Affiliated Hospital of Shandong First Medical University, Taian 271000, China
- Department of Hematology, The Second Affiliated Hospital of Shandong First Medical University, Taian 271000, China
| | - Weifeng Shi
- Key Laboratory of Emerging Infectious Diseases in Universities of Shandong, School of Clinical and Basic Medical Sciences, Shandong First Medical University & Shandong Academy of Medical Sciences, Taian, 271000, China
- School of Public Health, Shandong First Medical University & Shandong Academy of Medical Sciences, Ji’nan 250117, China
| |
Collapse
|
10
|
Tang Z, Yu P, Guo Q, Chen M, Lei Y, Zhou L, Mai W, Chen L, Deng M, Kong W, Niu C, Xiong X, Li W, Chen C, Lai C, Wang Q, Li B, Ji T. Clinical characteristics and host immunity responses of SARS-CoV-2 Omicron variant BA.2 with deletion of ORF7a, ORF7b and ORF8. Virol J 2023; 20:106. [PMID: 37248496 DOI: 10.1186/s12985-023-02066-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2023] [Accepted: 05/08/2023] [Indexed: 05/31/2023] Open
Abstract
BACKGROUND The pathogenicity and virulence of the Omicron strain have weakened significantly pathogenesis of Omicron variants. Accumulating data indicated accessory proteins play crucial roles in host immune evasion and virus pathogenesis of SARS-CoV-2. Therefore, the impact of simultaneous deletion of accessory protein ORF7a, ORF7b and ORF8 on the clinical characteristics and specific immunity in Omicron breakthrough infected patients (BIPs) need to be verified. METHODS Herein, plasma cytokines were identified using a commercial Multi-cytokine detection kit. Enzyme-linked immunosorbent assay and pseudovirus neutralization assays were utilized to determine the titers of SARS-CoV-2 specific binding antibodies and neutralizing antibodies, respectively. In addition, an enzyme-linked immunospot assay was used to quantify SARS-CoV-2 specific T cells and memory B cells. RESULTS A local COVID-19 outbreak was caused by the Omicron BA.2 variant, which featured a deletion of 871 base pairs (∆871 BA.2), resulting in the removal of ORF7a, ORF7b, and ORF8. We found that hospitalized patients with ∆871 BA.2 had significantly shorter hospital stays than those with wild-type (WT) BA.2. Plasma cytokine levels in both ∆871 BA.2 and WT BA.2 patients were within the normal range of reference, and there was no notable difference in the titers of SARS-CoV-2 ancestor or Omicron-specific binding IgG antibodies, neutralizing antibody titers, effector T cells, and memory B cells frequencies between ∆871 BA.2 and WT BA.2 infected adult patients. However, antibody titers in ∆871 BA.2 infected adolescents were higher than in adults. CONCLUSIONS The simultaneous deletion of ORF7a, ORF7b, and ORF8 facilitates the rapid clearance of the BA.2 variant, without impacting cytokine levels or affecting SARS-CoV-2 specific humoral and cellular immunity in Omicron-infected individuals.
Collapse
Affiliation(s)
- Zhizhong Tang
- Urology Surgery Department, Maoming People's Hospital, Maoming, 525000, People's Republic of China
| | - Pei Yu
- Clinical Laboratory Medicine Department, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510260, People's Republic of China
| | - Qianfang Guo
- Guangdong Provincial Key Laboratory of Pathogen Detection for Emerging Infectious Disease Response, Institute of Microbiology, Guangdong Provincial Center for Disease Control and Prevention, Guangdong, 511430, People's Republic of China
| | - Mingxiao Chen
- Clinical Laboratory Medicine Department, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510260, People's Republic of China
| | - Yu Lei
- Clinical Laboratory Medicine Department, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510260, People's Republic of China
| | - Lei Zhou
- Department Of Pathology Laboratory, Maoming People's Hospital, Maoming, 525000, People's Republic of China
| | - Weikang Mai
- Clinical Laboratory Medicine Department, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510260, People's Republic of China
| | - Lu Chen
- Clinical Laboratory Medicine Department, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510260, People's Republic of China
| | - Min Deng
- Clinical Laboratory Medicine Department, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510260, People's Republic of China
| | - Weiya Kong
- Clinical Laboratory Medicine Department, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510260, People's Republic of China
| | - Chuanying Niu
- State Key Laboratory of Respiratory Disease, CAS Key Laboratory of Regenerative Biology, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Guangdong Provincial Key Laboratory of Biocomputing, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510535, People's Republic of China
| | - Xiaoli Xiong
- State Key Laboratory of Respiratory Disease, CAS Key Laboratory of Regenerative Biology, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Guangdong Provincial Key Laboratory of Biocomputing, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510535, People's Republic of China
- Bioland Laboratory (Guangzhou Regenerative Medicine and Health-Guangdong Laboratory), Guangzhou, 510005, People's Republic of China
| | - Wenrui Li
- Clinical Laboratory Medicine Department, Dongguan Ninth People's Hospital, Dongguan, 523016, People's Republic of China
| | - Chunbo Chen
- Intensive Care Unit Department, Maoming People's Hospital, Maoming, 525000, People's Republic of China
| | - Changchun Lai
- Clinical Laboratory Medicine Department, Maoming People's Hospital, Maoming, 525000, People's Republic of China.
| | - Qian Wang
- State Key Laboratory of Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510120, People's Republic of China.
| | - Baisheng Li
- Guangdong Provincial Key Laboratory of Pathogen Detection for Emerging Infectious Disease Response, Institute of Microbiology, Guangdong Provincial Center for Disease Control and Prevention, Guangdong, 511430, People's Republic of China.
| | - Tianxing Ji
- Clinical Laboratory Medicine Department, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510260, People's Republic of China.
- Guangzhou Key Laboratory for Clinical Rapid Diagnosis and Early Warning of Infectious Diseases, KingMed School of Laboratory Medicine, Guangzhou Medical University, Guangzhou, 511495, People's Republic of China.
| |
Collapse
|
11
|
Raju S, Adams LJ, Earnest JT, Warfield K, Vang L, Crowe JE, Fremont DH, Diamond MS. A chikungunya virus-like particle vaccine induces broadly neutralizing and protective antibodies against alphaviruses in humans. Sci Transl Med 2023; 15:eade8273. [PMID: 37196061 PMCID: PMC10562830 DOI: 10.1126/scitranslmed.ade8273] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2022] [Accepted: 04/27/2023] [Indexed: 05/19/2023]
Abstract
Chikungunya virus (CHIKV) is a mosquito-transmitted alphavirus that causes epidemics of acute and chronic musculoskeletal disease. Here, we analyzed the human B cell response to a CHIKV-like particle-adjuvanted vaccine (PXVX0317) from samples obtained from a phase 2 clinical trial in humans (NCT03483961). Immunization with PXVX0317 induced high levels of neutralizing antibody in serum against CHIKV and circulating antigen-specific B cells up to 6 months after immunization. Monoclonal antibodies (mAbs) generated from peripheral blood B cells of three PXVX0317-vaccinated individuals on day 57 after immunization potently neutralized CHIKV infection, and a subset of these inhibited multiple related arthritogenic alphaviruses. Epitope mapping and cryo-electron microscopy defined two broadly neutralizing mAbs that uniquely bind to the apex of the B domain of the E2 glycoprotein. These results demonstrate the inhibitory breadth and activity of the human B cell response induced by the PXVX0317 vaccine against CHIKV and potentially other related alphaviruses.
Collapse
Affiliation(s)
- Saravanan Raju
- Department of Pathology & Immunology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Lucas J. Adams
- Department of Pathology & Immunology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - James T. Earnest
- Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
| | | | - Lo Vang
- Emergent BioSolutions, Gaithersburg, MD 20879, USA
| | - James E. Crowe
- Vanderbilt Vaccine Center, Vanderbilt University Medical Center, Nashville, TN 37232, USA
- Department of Pediatrics, Vanderbilt University Medical Center, Nashville, TN 37232, USA
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Daved H. Fremont
- Department of Pathology & Immunology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Michael S. Diamond
- Department of Pathology & Immunology, Washington University School of Medicine, St. Louis, MO 63110, USA
- Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
- Andrew M. and Jane M. Bursky Center for Human Immunology and Immunotherapy Programs, Washington University School of Medicine, St. Louis, MO 63110, USA
- Center for Vaccines and Immunity to Microbial Pathogens, Washington University School of Medicine, Saint Louis, MO 63110, USA
- Department of Molecular Microbiology, Washington University School of Medicine, St. Louis, MO 63110, USA
| |
Collapse
|
12
|
Choi HL, Yang HR, Shin HG, Hwang K, Kim JW, Lee JH, Ryu T, Jung Y, Lee S. Generation and Next-Generation Sequencing-Based Characterization of a Large Human Combinatorial Antibody Library. Int J Mol Sci 2023; 24:ijms24066011. [PMID: 36983085 PMCID: PMC10057307 DOI: 10.3390/ijms24066011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Revised: 03/04/2023] [Accepted: 03/21/2023] [Indexed: 03/30/2023] Open
Abstract
Antibody phage display is a key technology for the discovery and development of target-specific monoclonal antibodies (mAbs) for use in research, diagnostics, and therapy. The construction of a high-quality antibody library, with larger and more diverse antibody repertoires, is essential for the successful development of phage display-derived mAbs. In this study, a large human combinatorial single-chain variable fragment library (1.5 × 1011 colonies) was constructed from Epstein-Barr virus-infected human peripheral blood mononuclear cells stimulated with a combination of two of the activators of human B cells, the Toll-like receptor 7/8 agonist R848 and interleukin-2. Next-generation sequencing analysis with approximately 1.9 × 106 and 2.7 × 106 full-length sequences of heavy chain variable (VH) and κ light chain variable (Vκ) domains, respectively, revealed that the library consists of unique VH (approximately 94%) and Vκ (approximately 91%) sequences with greater diversity than germline sequences. Lastly, multiple unique mAbs with high affinity and broad cross-species reactivity could be isolated from the library against two therapeutically relevant target antigens, validating the library quality. These findings suggest that the novel antibody library we have developed may be useful for the rapid development of target-specific phage display-derived recombinant human mAbs for use in therapeutic and diagnostic applications.
Collapse
Affiliation(s)
- Hye Lim Choi
- Department of Biopharmaceutical Chemistry, Kookmin University, Seoul 02707, Republic of Korea
| | - Ha Rim Yang
- Department of Biopharmaceutical Chemistry, Kookmin University, Seoul 02707, Republic of Korea
| | - Ha Gyeong Shin
- Department of Biopharmaceutical Chemistry, Kookmin University, Seoul 02707, Republic of Korea
| | - Kyusang Hwang
- Department of Biopharmaceutical Chemistry, Kookmin University, Seoul 02707, Republic of Korea
| | - Ji Woong Kim
- Department of Chemistry, Kookmin University, Seoul 02707, Republic of Korea
| | - Ji Hyun Lee
- Department of Biopharmaceutical Chemistry, Kookmin University, Seoul 02707, Republic of Korea
| | - Taehoon Ryu
- ATG Lifetech Inc., Seoul 08507, Republic of Korea
| | - Yushin Jung
- ATG Lifetech Inc., Seoul 08507, Republic of Korea
| | - Sukmook Lee
- Department of Biopharmaceutical Chemistry, Kookmin University, Seoul 02707, Republic of Korea
- Department of Applied Chemistry, Kookmin University, Seoul 02707, Republic of Korea
- Antibody Research Institute, Kookmin University, Seoul 02707, Republic of Korea
| |
Collapse
|
13
|
Powers JM, Lyski ZL, Weber WC, Denton M, Streblow MM, Mayo AT, Haese NN, Nix CD, Rodríguez-Santiago R, Alvarado LI, Rivera-Amill V, Messer WB, Streblow DN. Infection with chikungunya virus confers heterotypic cross-neutralizing antibodies and memory B-cells against other arthritogenic alphaviruses predominantly through the B domain of the E2 glycoprotein. PLoS Negl Trop Dis 2023; 17:e0011154. [PMID: 36913428 PMCID: PMC10036167 DOI: 10.1371/journal.pntd.0011154] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2022] [Revised: 03/23/2023] [Accepted: 02/09/2023] [Indexed: 03/14/2023] Open
Abstract
Infections with Chikungunya virus, a mosquito-borne alphavirus, cause an acute febrile syndrome often followed by chronic arthritis that persists for months to years post-infection. Neutralizing antibodies are the primary immune correlate of protection elicited by infection, and the major goal of vaccinations in development. Using convalescent blood samples collected from both endemic and non-endemic human subjects at multiple timepoints following suspected or confirmed chikungunya infection, we identified antibodies with broad neutralizing properties against other alphaviruses within the Semliki Forest complex. Cross-neutralization generally did not extend to the Venezuelan Equine Encephalitis virus (VEEV) complex, although some subjects had low levels of VEEV-neutralizing antibodies. This suggests that broadly neutralizing antibodies elicited following natural infection are largely complex restricted. In addition to serology, we also performed memory B-cell analysis, finding chikungunya-specific memory B-cells in all subjects in this study as remotely as 24 years post-infection. We functionally assessed the ability of memory B-cell derived antibodies to bind to chikungunya virus, and related Mayaro virus, as well as the highly conserved B domain of the E2 glycoprotein thought to contribute to cross-reactivity between related Old-World alphaviruses. To specifically assess the role of the E2 B domain in cross-neutralization, we depleted Mayaro and Chikungunya virus E2 B domain specific antibodies from convalescent sera, finding E2B depletion significantly decreases Mayaro virus specific cross-neutralizing antibody titers with no significant effect on chikungunya virus neutralization, indicating that the E2 B domain is a key target of cross-neutralizing and potentially cross-protective neutralizing antibodies.
Collapse
Affiliation(s)
- John M. Powers
- Vaccine and Gene Therapy Institute, Oregon Health and Science University, Beaverton, Oregon, United States of America
| | - Zoe L. Lyski
- Department of Molecular Microbiology and Immunology, Oregon Health and Science University, Portland, Oregon, United States of America
| | - Whitney C. Weber
- Vaccine and Gene Therapy Institute, Oregon Health and Science University, Beaverton, Oregon, United States of America
- Department of Molecular Microbiology and Immunology, Oregon Health and Science University, Portland, Oregon, United States of America
| | - Michael Denton
- Vaccine and Gene Therapy Institute, Oregon Health and Science University, Beaverton, Oregon, United States of America
| | - Magdalene M. Streblow
- Vaccine and Gene Therapy Institute, Oregon Health and Science University, Beaverton, Oregon, United States of America
| | - Adam T. Mayo
- Vaccine and Gene Therapy Institute, Oregon Health and Science University, Beaverton, Oregon, United States of America
| | - Nicole N. Haese
- Vaccine and Gene Therapy Institute, Oregon Health and Science University, Beaverton, Oregon, United States of America
| | - Chad D. Nix
- Department of Molecular Microbiology and Immunology, Oregon Health and Science University, Portland, Oregon, United States of America
| | | | - Luisa I. Alvarado
- Ponce Health Sciences University/ Ponce Research Institute, Ponce, Puerto Rico
| | | | - William B. Messer
- Department of Molecular Microbiology and Immunology, Oregon Health and Science University, Portland, Oregon, United States of America
- Department of Medicine, Division of Infectious Disease Oregon Health and Science University, Portland, Oregon, United States of America
- OHSU-PSU School of Public Health, Program in Epidemiology, Oregon Health and Science University, Portland, Oregon, United States of America
| | - Daniel N. Streblow
- Vaccine and Gene Therapy Institute, Oregon Health and Science University, Beaverton, Oregon, United States of America
- Division of Pathobiology and Immunology, Oregon National Primate Research Center, Beaverton, Oregon, United States of America
| |
Collapse
|
14
|
Marzi R, Bassi J, Silacci-Fregni C, Bartha I, Muoio F, Culap K, Sprugasci N, Lombardo G, Saliba C, Cameroni E, Cassotta A, Low JS, Walls AC, McCallum M, Tortorici MA, Bowen JE, Dellota EA, Dillen JR, Czudnochowski N, Pertusini L, Terrot T, Lepori V, Tarkowski M, Riva A, Biggiogero M, Franzetti-Pellanda A, Garzoni C, Ferrari P, Ceschi A, Giannini O, Havenar-Daughton C, Telenti A, Arvin A, Virgin HW, Sallusto F, Veesler D, Lanzavecchia A, Corti D, Piccoli L. Maturation of SARS-CoV-2 Spike-specific memory B cells drives resilience to viral escape. iScience 2023; 26:105726. [PMID: 36507220 PMCID: PMC9721160 DOI: 10.1016/j.isci.2022.105726] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Revised: 10/21/2022] [Accepted: 12/01/2022] [Indexed: 12/12/2022] Open
Abstract
Memory B cells (MBCs) generate rapid antibody responses upon secondary encounter with a pathogen. Here, we investigated the kinetics, avidity, and cross-reactivity of serum antibodies and MBCs in 155 SARS-CoV-2 infected and vaccinated individuals over a 16-month time frame. SARS-CoV-2-specific MBCs and serum antibodies reached steady-state titers with comparable kinetics in infected and vaccinated individuals. Whereas MBCs of infected individuals targeted both prefusion and postfusion Spike (S), most vaccine-elicited MBCs were specific for prefusion S, consistent with the use of prefusion-stabilized S in mRNA vaccines. Furthermore, a large fraction of MBCs recognizing postfusion S cross-reacted with human betacoronaviruses. The avidity of MBC-derived and serum antibodies increased over time resulting in enhanced resilience to viral escape by SARS-CoV-2 variants, including Omicron BA.1 and BA.2 sublineages, albeit only partially for BA.4 and BA.5 sublineages. Overall, the maturation of high-affinity and broadly reactive MBCs provides the basis for effective recall responses to future SARS-CoV-2 variants.
Collapse
Affiliation(s)
- Roberta Marzi
- Humabs BioMed SA, a subsidiary of Vir Biotechnology, Bellinzona, Switzerland
| | - Jessica Bassi
- Humabs BioMed SA, a subsidiary of Vir Biotechnology, Bellinzona, Switzerland
| | | | - Istvan Bartha
- Humabs BioMed SA, a subsidiary of Vir Biotechnology, Bellinzona, Switzerland
| | - Francesco Muoio
- Humabs BioMed SA, a subsidiary of Vir Biotechnology, Bellinzona, Switzerland
| | - Katja Culap
- Humabs BioMed SA, a subsidiary of Vir Biotechnology, Bellinzona, Switzerland
| | - Nicole Sprugasci
- Humabs BioMed SA, a subsidiary of Vir Biotechnology, Bellinzona, Switzerland
| | - Gloria Lombardo
- Humabs BioMed SA, a subsidiary of Vir Biotechnology, Bellinzona, Switzerland
| | - Christian Saliba
- Humabs BioMed SA, a subsidiary of Vir Biotechnology, Bellinzona, Switzerland
| | - Elisabetta Cameroni
- Humabs BioMed SA, a subsidiary of Vir Biotechnology, Bellinzona, Switzerland
| | - Antonino Cassotta
- Institute for Research in Biomedicine, Università della Svizzera italiana, Bellinzona, Switzerland
| | - Jun Siong Low
- Institute for Research in Biomedicine, Università della Svizzera italiana, Bellinzona, Switzerland
- Institute of Microbiology, ETH Zurich, Zurich, Switzerland
| | | | - Matthew McCallum
- Department of Biochemistry, University of Washington, Seattle, WA, USA
| | | | - John E. Bowen
- Department of Biochemistry, University of Washington, Seattle, WA, USA
| | | | | | | | - Laura Pertusini
- Division of Nephrology, Ente Ospedaliero Cantonale, Lugano, Switzerland
| | - Tatiana Terrot
- Clinical Trial Unit, Ente Ospedaliero Cantonale, Lugano, Switzerland
| | | | - Maciej Tarkowski
- Department of Biomedical and Clinical Sciences “L. Sacco”, University of Milan, Milan, Italy
| | - Agostino Riva
- Department of Biomedical and Clinical Sciences “L. Sacco”, University of Milan, Milan, Italy
- III Division of Infectious Diseases, ASST Fatebenefratelli Sacco, Luigi Sacco Hospital, Milan, Italy
| | - Maira Biggiogero
- Clinical Research Unit, Clinica Luganese Moncucco, Lugano, Switzerland
| | | | - Christian Garzoni
- Clinic of Internal Medicine and Infectious Diseases, Clinica Luganese Moncucco, Lugano, Switzerland
| | - Paolo Ferrari
- Division of Nephrology, Ente Ospedaliero Cantonale, Lugano, Switzerland
- Faculty of Biomedical Sciences, Università della Svizzera italiana, Lugano, Switzerland
- Clinical School, University of New South Wales, Sydney, NSW, Australia
| | - Alessandro Ceschi
- Clinical Trial Unit, Ente Ospedaliero Cantonale, Lugano, Switzerland
- Faculty of Biomedical Sciences, Università della Svizzera italiana, Lugano, Switzerland
- Division of Clinical Pharmacology and Toxicology, Institute of Pharmacological Sciences of Southern Switzerland, Ente Ospedaliero Cantonale, Lugano, Switzerland
- Department of Clinical Pharmacology and Toxicology, University Hospital Zurich, Zurich, Switzerland
| | - Olivier Giannini
- Faculty of Biomedical Sciences, Università della Svizzera italiana, Lugano, Switzerland
- Department of Medicine, Ente Ospedaliero Cantonale, Bellinzona, Switzerland
| | | | | | - Ann Arvin
- Vir Biotechnology, San Francisco, CA, USA
| | - Herbert W. Virgin
- Vir Biotechnology, San Francisco, CA, USA
- Department of Pathology and Immunology, Washington University School of Medicine, St Louis, MO, USA
- Department of Internal Medicine, UT Southwestern Medical Center, Dallas, TX, USA
| | - Federica Sallusto
- Institute for Research in Biomedicine, Università della Svizzera italiana, Bellinzona, Switzerland
- Institute of Microbiology, ETH Zurich, Zurich, Switzerland
| | - David Veesler
- Department of Biochemistry, University of Washington, Seattle, WA, USA
| | | | - Davide Corti
- Humabs BioMed SA, a subsidiary of Vir Biotechnology, Bellinzona, Switzerland
| | - Luca Piccoli
- Humabs BioMed SA, a subsidiary of Vir Biotechnology, Bellinzona, Switzerland
| |
Collapse
|
15
|
Yu P, Liu Z, Zhu Z, Yang J, Deng M, Chen M, Lai C, Kong W, Xiong S, Wan L, Mai W, Chen L, Lei Y, Khan SA, Ruan J, Kang A, Guo X, Zhou Q, Li W, Chen Z, Liang Y, Li P, Zhang L, Ji T. Omicron variants breakthrough infection elicited higher specific memory immunity than third dose booster in healthy vaccinees. Virol Sin 2023; 38:233-243. [PMID: 36603767 PMCID: PMC10176432 DOI: 10.1016/j.virs.2022.12.008] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2022] [Accepted: 12/30/2022] [Indexed: 01/03/2023] Open
Abstract
Homologous booster, heterologous booster, and Omicron variants breakthrough infection (OBI) could improve the humoral immunity against Omicron variants. Questions concerning about memory B cells (MBCs) and T cells immunity against Omicron variants, features of long-term immunity, after booster and OBI, needs to be explored. Here, comparative analysis demonstrate antibody and T cell immunity against ancestral strain, Delta and Omicron variants in Omicron breakthrough infected patients (OBIPs) are comparable to that in Ad5-nCoV boosted healthy volunteers (HVs), higher than that in inactivated vaccine (InV) boosted HVs. However, memory B cells (MBCs) immunity against Omicron variants was highest in OBIPs, followed by Ad5-nCoV boosted and InV boosted HVs. OBIPs and Ad5-nCoV boosted HVs have higher classical MBCs and activated MBCs, and lower naïve MBCs and atypical MBCs relative to both vaccine boosted HVs. Collectively, these data indicate Omicron breakthrough infection elicit higher MBCs and T cells against SARS-CoV-2 especially Omicron variants relative to homologous InV booster and heterologous Ad5-nCoV booster.
Collapse
Affiliation(s)
- Pei Yu
- Clinical Laboratory Medicine Department, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510260, China
| | - Zijian Liu
- State Key Laboratories of Respiratory Diseases, Guangdong-Hong Kong-Macao Joint Laboratory of Infectious Respiratory Disease, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510535, China
| | - Zhuoqi Zhu
- Clinical Laboratory Medicine Department, Dongguan Ninth People's Hospital, Dongguan, 523016, China
| | - Jiaqing Yang
- State Key Laboratory of Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510120, China
| | - Min Deng
- Clinical Laboratory Medicine Department, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510260, China
| | - Mingxiao Chen
- Clinical Laboratory Medicine Department, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510260, China
| | - Changchun Lai
- Clinical Laboratory Medicine Department, Maoming People's Hospital, Maoming, 525000, China
| | - Weiya Kong
- Clinical Laboratory Medicine Department, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510260, China
| | - Shilong Xiong
- Clinical Laboratory Medicine Department, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510260, China
| | - Li Wan
- Clinical Laboratory Medicine Department, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510260, China
| | - Weikang Mai
- Clinical Laboratory Medicine Department, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510260, China
| | - Lu Chen
- Clinical Laboratory Medicine Department, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510260, China
| | - Yu Lei
- Clinical Laboratory Medicine Department, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510260, China
| | - Shahzad Akbar Khan
- Laboratory of Pathology, Department of Pathobiology, University of Poonch Rawalakot Azad Kashmir Pakistan 12350, Pakistan
| | - Jianfeng Ruan
- Hospital Infection-Control Department, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510260, China
| | - An Kang
- Medical Examination Department, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510260, China
| | - Xuguang Guo
- Department of Clinical Laboratory Medicine, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510140, China
| | - Qiang Zhou
- Clinical Laboratory Medicine Department, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510260, China
| | - Wenrui Li
- Clinical Laboratory Medicine Department, Dongguan Ninth People's Hospital, Dongguan, 523016, China
| | - Zheng Chen
- Kidney Transplant Department, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510260, China.
| | - Yuemei Liang
- Clinical Laboratory Medicine Department, Dongguan Ninth People's Hospital, Dongguan, 523016, China.
| | - Pingchao Li
- State Key Laboratories of Respiratory Diseases, Guangdong-Hong Kong-Macao Joint Laboratory of Infectious Respiratory Disease, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510535, China.
| | - Lei Zhang
- Kidney Transplant Department, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510260, China; Department of Organ Transplantation, The Third Affiliated Hospital of Sun Yat-sen University, 510630, China.
| | - Tianxing Ji
- Clinical Laboratory Medicine Department, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510260, China; Guangzhou Key Laboratory for Clinical Rapid Diagnosis and Early Warning of Infectious Diseases, KingMed School of Laboratory Medicine, Guangzhou Medical University, Guangzhou, 511436, China.
| |
Collapse
|
16
|
Disanto G, Galante A, Cantu' M, Sacco R, Mele F, Eisler JJ, Keller F, Bernasconi E, Sallusto F, Zecca C, Gobbi C. Longitudinal Postvaccine SARS-CoV-2 Immunoglobulin G Titers, Memory B-Cell Responses, and Risk of COVID-19 in Multiple Sclerosis Over 1 Year. NEUROLOGY(R) NEUROIMMUNOLOGY & NEUROINFLAMMATION 2023; 10:e200043. [PMID: 36396447 PMCID: PMC9747147 DOI: 10.1212/nxi.0000000000200043] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/08/2022] [Accepted: 08/29/2022] [Indexed: 11/19/2022]
Abstract
BACKGROUND AND OBJECTIVES Some disease-modifying treatments impair response to severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) vaccines in multiple sclerosis (MS), potentially increasing the risk of breakthrough infections. We aimed to investigate longitudinal SARS-CoV-2 antibody dynamics and memory B cells after 2 and 3 messenger RNA (mRNA) vaccine doses and their association with the risk of COVID-19 in patients with MS on different treatments over 1 year. METHODS Prospective observational cohort study in patients with MS undergoing SARS-CoV-2 mRNA vaccinations. Antispike (anti-S) immunoglobulin G (IgG) titers were measured by chemiluminescence microparticle immunoassay. Frequencies of spike-specific memory B cells were measured on polyclonal stimulation of peripheral blood mononuclear cells and screening of secreted antibodies by ELISA. RESULTS We recruited 120 patients with MS (58 on anti-CD20 antibodies, 9 on sphingosine 1-phosphate (S1P) receptor modulators, 15 on cladribine, 24 on teriflunomide (TFL), and 14 untreated) and collected 392 samples up to 10.8 months after 2 vaccine doses. When compared with untreated patients, anti-CD20 antibodies (β = -2.07, p < 0.001) and S1P modulators (β = -2.02, p < 0.001) were associated with lower anti-S IgG, while TFL and cladribine were not. Anti-S IgG decreased with months since vaccine (β = -0.14, p < 0.001), independently of treatments. Within anti-CD20 patients, anti-S IgG remained higher in those with greater baseline B-cell counts and were not influenced by postvaccine anti-CD20 infusions. Anti-S IgG increase after a 3rd vaccine was mild on anti-CD20 and S1P modulators. Spike-specific memory B-cell responses were weaker on S1P modulators and anti-CD20 than on TFL and influenced by postvaccine anti-CD20 infusions. The frequency of breakthrough infections was comparable between DMTs, but the risk of COVID-19 was predicted by the last measured anti-S IgG titer before infection (OR = 0.56, 95% CI = 0.37-0.86, p = 0.008). DISCUSSION Postvaccine anti-S IgG titers decrease over time regardless of MS treatment and are associated with breakthrough COVID-19. Both humoral and specific memory B-cell responses are diminished on S1P modulators. Within anti-CD20-treated patients, B-cell count at first vaccine determines anti-S IgG production, whereas postvaccine anti-CD20 infusions negatively affect spike-specific memory B cells.
Collapse
Affiliation(s)
- Giulio Disanto
- From the Multiple Sclerosis Center (G.D., R.S., J.J.E., C.Z., C.G.), Neurocenter of Southern Switzerland, Ente Ospedaliero Cantonale, Lugano; Institute for Research in Biomedicine (A.G., F.M., F.S.), Università Della Svizzera Italiana, Bellinzona; Institute of Laboratory Medicine (M.C., F.K.), Ente Ospedaliero Cantonale, Bellinzona; Department of Medicine (E.B.), Ente Ospedaliero Cantonale, Lugano; Faculty of Biomedical Sciences (E.B., F.S., C.Z., C.G.), Università Della Svizzera Italiana, Lugano; and Institute of Microbiology (F.S.), ETH Zurich, Switzerland
| | - Alice Galante
- From the Multiple Sclerosis Center (G.D., R.S., J.J.E., C.Z., C.G.), Neurocenter of Southern Switzerland, Ente Ospedaliero Cantonale, Lugano; Institute for Research in Biomedicine (A.G., F.M., F.S.), Università Della Svizzera Italiana, Bellinzona; Institute of Laboratory Medicine (M.C., F.K.), Ente Ospedaliero Cantonale, Bellinzona; Department of Medicine (E.B.), Ente Ospedaliero Cantonale, Lugano; Faculty of Biomedical Sciences (E.B., F.S., C.Z., C.G.), Università Della Svizzera Italiana, Lugano; and Institute of Microbiology (F.S.), ETH Zurich, Switzerland
| | - Marco Cantu'
- From the Multiple Sclerosis Center (G.D., R.S., J.J.E., C.Z., C.G.), Neurocenter of Southern Switzerland, Ente Ospedaliero Cantonale, Lugano; Institute for Research in Biomedicine (A.G., F.M., F.S.), Università Della Svizzera Italiana, Bellinzona; Institute of Laboratory Medicine (M.C., F.K.), Ente Ospedaliero Cantonale, Bellinzona; Department of Medicine (E.B.), Ente Ospedaliero Cantonale, Lugano; Faculty of Biomedical Sciences (E.B., F.S., C.Z., C.G.), Università Della Svizzera Italiana, Lugano; and Institute of Microbiology (F.S.), ETH Zurich, Switzerland
| | - Rosaria Sacco
- From the Multiple Sclerosis Center (G.D., R.S., J.J.E., C.Z., C.G.), Neurocenter of Southern Switzerland, Ente Ospedaliero Cantonale, Lugano; Institute for Research in Biomedicine (A.G., F.M., F.S.), Università Della Svizzera Italiana, Bellinzona; Institute of Laboratory Medicine (M.C., F.K.), Ente Ospedaliero Cantonale, Bellinzona; Department of Medicine (E.B.), Ente Ospedaliero Cantonale, Lugano; Faculty of Biomedical Sciences (E.B., F.S., C.Z., C.G.), Università Della Svizzera Italiana, Lugano; and Institute of Microbiology (F.S.), ETH Zurich, Switzerland
| | - Federico Mele
- From the Multiple Sclerosis Center (G.D., R.S., J.J.E., C.Z., C.G.), Neurocenter of Southern Switzerland, Ente Ospedaliero Cantonale, Lugano; Institute for Research in Biomedicine (A.G., F.M., F.S.), Università Della Svizzera Italiana, Bellinzona; Institute of Laboratory Medicine (M.C., F.K.), Ente Ospedaliero Cantonale, Bellinzona; Department of Medicine (E.B.), Ente Ospedaliero Cantonale, Lugano; Faculty of Biomedical Sciences (E.B., F.S., C.Z., C.G.), Università Della Svizzera Italiana, Lugano; and Institute of Microbiology (F.S.), ETH Zurich, Switzerland
| | - Jennifer Jessica Eisler
- From the Multiple Sclerosis Center (G.D., R.S., J.J.E., C.Z., C.G.), Neurocenter of Southern Switzerland, Ente Ospedaliero Cantonale, Lugano; Institute for Research in Biomedicine (A.G., F.M., F.S.), Università Della Svizzera Italiana, Bellinzona; Institute of Laboratory Medicine (M.C., F.K.), Ente Ospedaliero Cantonale, Bellinzona; Department of Medicine (E.B.), Ente Ospedaliero Cantonale, Lugano; Faculty of Biomedical Sciences (E.B., F.S., C.Z., C.G.), Università Della Svizzera Italiana, Lugano; and Institute of Microbiology (F.S.), ETH Zurich, Switzerland
| | - Franco Keller
- From the Multiple Sclerosis Center (G.D., R.S., J.J.E., C.Z., C.G.), Neurocenter of Southern Switzerland, Ente Ospedaliero Cantonale, Lugano; Institute for Research in Biomedicine (A.G., F.M., F.S.), Università Della Svizzera Italiana, Bellinzona; Institute of Laboratory Medicine (M.C., F.K.), Ente Ospedaliero Cantonale, Bellinzona; Department of Medicine (E.B.), Ente Ospedaliero Cantonale, Lugano; Faculty of Biomedical Sciences (E.B., F.S., C.Z., C.G.), Università Della Svizzera Italiana, Lugano; and Institute of Microbiology (F.S.), ETH Zurich, Switzerland
| | - Enos Bernasconi
- From the Multiple Sclerosis Center (G.D., R.S., J.J.E., C.Z., C.G.), Neurocenter of Southern Switzerland, Ente Ospedaliero Cantonale, Lugano; Institute for Research in Biomedicine (A.G., F.M., F.S.), Università Della Svizzera Italiana, Bellinzona; Institute of Laboratory Medicine (M.C., F.K.), Ente Ospedaliero Cantonale, Bellinzona; Department of Medicine (E.B.), Ente Ospedaliero Cantonale, Lugano; Faculty of Biomedical Sciences (E.B., F.S., C.Z., C.G.), Università Della Svizzera Italiana, Lugano; and Institute of Microbiology (F.S.), ETH Zurich, Switzerland
| | - Federica Sallusto
- From the Multiple Sclerosis Center (G.D., R.S., J.J.E., C.Z., C.G.), Neurocenter of Southern Switzerland, Ente Ospedaliero Cantonale, Lugano; Institute for Research in Biomedicine (A.G., F.M., F.S.), Università Della Svizzera Italiana, Bellinzona; Institute of Laboratory Medicine (M.C., F.K.), Ente Ospedaliero Cantonale, Bellinzona; Department of Medicine (E.B.), Ente Ospedaliero Cantonale, Lugano; Faculty of Biomedical Sciences (E.B., F.S., C.Z., C.G.), Università Della Svizzera Italiana, Lugano; and Institute of Microbiology (F.S.), ETH Zurich, Switzerland
| | - Chiara Zecca
- From the Multiple Sclerosis Center (G.D., R.S., J.J.E., C.Z., C.G.), Neurocenter of Southern Switzerland, Ente Ospedaliero Cantonale, Lugano; Institute for Research in Biomedicine (A.G., F.M., F.S.), Università Della Svizzera Italiana, Bellinzona; Institute of Laboratory Medicine (M.C., F.K.), Ente Ospedaliero Cantonale, Bellinzona; Department of Medicine (E.B.), Ente Ospedaliero Cantonale, Lugano; Faculty of Biomedical Sciences (E.B., F.S., C.Z., C.G.), Università Della Svizzera Italiana, Lugano; and Institute of Microbiology (F.S.), ETH Zurich, Switzerland
| | - Claudio Gobbi
- From the Multiple Sclerosis Center (G.D., R.S., J.J.E., C.Z., C.G.), Neurocenter of Southern Switzerland, Ente Ospedaliero Cantonale, Lugano; Institute for Research in Biomedicine (A.G., F.M., F.S.), Università Della Svizzera Italiana, Bellinzona; Institute of Laboratory Medicine (M.C., F.K.), Ente Ospedaliero Cantonale, Bellinzona; Department of Medicine (E.B.), Ente Ospedaliero Cantonale, Lugano; Faculty of Biomedical Sciences (E.B., F.S., C.Z., C.G.), Università Della Svizzera Italiana, Lugano; and Institute of Microbiology (F.S.), ETH Zurich, Switzerland.
| |
Collapse
|
17
|
Wolf C, Köppert S, Becza N, Kuerten S, Kirchenbaum GA, Lehmann PV. Antibody Levels Poorly Reflect on the Frequency of Memory B Cells Generated following SARS-CoV-2, Seasonal Influenza, or EBV Infection. Cells 2022; 11:cells11223662. [PMID: 36429090 PMCID: PMC9688940 DOI: 10.3390/cells11223662] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2022] [Revised: 11/14/2022] [Accepted: 11/15/2022] [Indexed: 11/19/2022] Open
Abstract
The scope of immune monitoring is to define the existence, magnitude, and quality of immune mechanisms operational in a host. In clinical trials and praxis, the assessment of humoral immunity is commonly confined to measurements of serum antibody reactivity without accounting for the memory B cell potential. Relying on fundamentally different mechanisms, however, passive immunity conveyed by pre-existing antibodies needs to be distinguished from active B cell memory. Here, we tested whether, in healthy human individuals, the antibody titers to SARS-CoV-2, seasonal influenza, or Epstein-Barr virus antigens correlated with the frequency of recirculating memory B cells reactive with the respective antigens. Weak correlations were found. The data suggest that the assessment of humoral immunity by measurement of antibody levels does not reflect on memory B cell frequencies and thus an individual's potential to engage in an anamnestic antibody response against the same or an antigenically related virus. Direct monitoring of the antigen-reactive memory B cell compartment is both required and feasible towards that goal.
Collapse
Affiliation(s)
- Carla Wolf
- Research and Development, Cellular Technology Ltd. (CTL), Shaker Heights, OH 44122, USA
- Institute of Anatomy and Cell Biology, Friedrich-Alexander University Erlangen-Nürnberg, 91054 Erlangen, Germany
| | - Sebastian Köppert
- Research and Development, Cellular Technology Ltd. (CTL), Shaker Heights, OH 44122, USA
- Institute of Anatomy and Cell Biology, Friedrich-Alexander University Erlangen-Nürnberg, 91054 Erlangen, Germany
| | - Noémi Becza
- Research and Development, Cellular Technology Ltd. (CTL), Shaker Heights, OH 44122, USA
| | - Stefanie Kuerten
- Institute of Anatomy and Cell Biology, Friedrich-Alexander University Erlangen-Nürnberg, 91054 Erlangen, Germany
- Institute of Neuroanatomy, Medical Faculty, University of Bonn, 53115 Bonn, Germany
| | - Greg A. Kirchenbaum
- Research and Development, Cellular Technology Ltd. (CTL), Shaker Heights, OH 44122, USA
| | - Paul V. Lehmann
- Research and Development, Cellular Technology Ltd. (CTL), Shaker Heights, OH 44122, USA
- Correspondence: ; Tel.: +1-(216)-791-5084
| |
Collapse
|
18
|
Park YJ, Pinto D, Walls AC, Liu Z, De Marco A, Benigni F, Zatta F, Silacci-Fregni C, Bassi J, Sprouse KR, Addetia A, Bowen JE, Stewart C, Giurdanella M, Saliba C, Guarino B, Schmid MA, Franko NM, Logue JK, Dang HV, Hauser K, di Iulio J, Rivera W, Schnell G, Rajesh A, Zhou J, Farhat N, Kaiser H, Montiel-Ruiz M, Noack J, Lempp FA, Janer J, Abdelnabi R, Maes P, Ferrari P, Ceschi A, Giannini O, de Melo GD, Kergoat L, Bourhy H, Neyts J, Soriaga L, Purcell LA, Snell G, Whelan SPJ, Lanzavecchia A, Virgin HW, Piccoli L, Chu HY, Pizzuto MS, Corti D, Veesler D. Imprinted antibody responses against SARS-CoV-2 Omicron sublineages. Science 2022; 378:619-627. [PMID: 36264829 DOI: 10.1126/science.adc9127] [Citation(s) in RCA: 106] [Impact Index Per Article: 53.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) Omicron sublineages carry distinct spike mutations resulting in escape from antibodies induced by previous infection or vaccination. We show that hybrid immunity or vaccine boosters elicit plasma-neutralizing antibodies against Omicron BA.1, BA.2, BA.2.12.1, and BA.4/5, and that breakthrough infections, but not vaccination alone, induce neutralizing antibodies in the nasal mucosa. Consistent with immunological imprinting, most antibodies derived from memory B cells or plasma cells of Omicron breakthrough cases cross-react with the Wuhan-Hu-1, BA.1, BA.2, and BA.4/5 receptor-binding domains, whereas Omicron primary infections elicit B cells of narrow specificity up to 6 months after infection. Although most clinical antibodies have reduced neutralization of Omicron, we identified an ultrapotent pan-variant-neutralizing antibody that is a strong candidate for clinical development.
Collapse
Affiliation(s)
- Young-Jun Park
- Department of Biochemistry, University of Washington, Seattle, WA, USA
- Howard Hughes Medical Institute, University of Washington, Seattle, WA, USA
| | - Dora Pinto
- Humabs Biomed SA, Subsidiary of Vir Biotechnology, Bellinzona, Switzerland
| | - Alexandra C Walls
- Department of Biochemistry, University of Washington, Seattle, WA, USA
- Howard Hughes Medical Institute, University of Washington, Seattle, WA, USA
| | - Zhuoming Liu
- Department of Molecular Microbiology, Washington University School of Medicine, St. Louis, MO, USA
| | - Anna De Marco
- Humabs Biomed SA, Subsidiary of Vir Biotechnology, Bellinzona, Switzerland
| | - Fabio Benigni
- Humabs Biomed SA, Subsidiary of Vir Biotechnology, Bellinzona, Switzerland
| | - Fabrizia Zatta
- Humabs Biomed SA, Subsidiary of Vir Biotechnology, Bellinzona, Switzerland
| | | | - Jessica Bassi
- Humabs Biomed SA, Subsidiary of Vir Biotechnology, Bellinzona, Switzerland
| | - Kaitlin R Sprouse
- Department of Biochemistry, University of Washington, Seattle, WA, USA
| | - Amin Addetia
- Department of Biochemistry, University of Washington, Seattle, WA, USA
| | - John E Bowen
- Department of Biochemistry, University of Washington, Seattle, WA, USA
| | - Cameron Stewart
- Department of Biochemistry, University of Washington, Seattle, WA, USA
| | | | - Christian Saliba
- Humabs Biomed SA, Subsidiary of Vir Biotechnology, Bellinzona, Switzerland
| | - Barbara Guarino
- Humabs Biomed SA, Subsidiary of Vir Biotechnology, Bellinzona, Switzerland
| | - Michael A Schmid
- Humabs Biomed SA, Subsidiary of Vir Biotechnology, Bellinzona, Switzerland
| | - Nicholas M Franko
- Division of Allergy and Infectious Diseases, University of Washington, Seattle, WA, USA
| | - Jennifer K Logue
- Division of Allergy and Infectious Diseases, University of Washington, Seattle, WA, USA
| | - Ha V Dang
- Vir Biotechnology, San Francisco, CA, USA
| | | | | | | | | | | | - Jiayi Zhou
- Vir Biotechnology, San Francisco, CA, USA
| | | | | | | | | | | | - Javier Janer
- Department of Molecular Microbiology, Washington University School of Medicine, St. Louis, MO, USA
| | - Rana Abdelnabi
- KU Leuven Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, Laboratory of Virology and Chemotherapy, B-3000 Leuven, Belgium
| | - Piet Maes
- KU Leuven Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, Laboratory of Virology and Chemotherapy, B-3000 Leuven, Belgium
| | - Paolo Ferrari
- Faculty of Biomedical Sciences, Università della Svizzera italiana, Lugano, Switzerland
- Division of Nephrology, Ente Ospedaliero Cantonale, Lugano, Switzerland
- Clinical School, University of New South Wales, Sydney, New South Wales, Australia
| | - Alessandro Ceschi
- Faculty of Biomedical Sciences, Università della Svizzera italiana, Lugano, Switzerland
- Clinical Trial Unit, Ente Ospedaliero Cantonale, Lugano, Switzerland
- Division of Clinical Pharmacology and Toxicology, Institute of Pharmacological Sciences of Southern Switzerland, Ente Ospedaliero Cantonale, Lugano, Switzerland
- Department of Clinical Pharmacology and Toxicology, University Hospital Zurich, Zurich, Switzerland
| | - Olivier Giannini
- Faculty of Biomedical Sciences, Università della Svizzera italiana, Lugano, Switzerland
- Department of Medicine, Ente Ospedaliero Cantonale, Bellinzona, Switzerland
| | - Guilherme Dias de Melo
- Institut Pasteur, Université Paris Cité, Lyssavirus Epidemiology and Neuropathology Unit, F-75015 Paris, France
| | - Lauriane Kergoat
- Institut Pasteur, Université Paris Cité, Lyssavirus Epidemiology and Neuropathology Unit, F-75015 Paris, France
| | - Hervé Bourhy
- Institut Pasteur, Université Paris Cité, Lyssavirus Epidemiology and Neuropathology Unit, F-75015 Paris, France
| | - Johan Neyts
- KU Leuven Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, Laboratory of Virology and Chemotherapy, B-3000 Leuven, Belgium
| | | | | | | | - Sean P J Whelan
- Department of Molecular Microbiology, Washington University School of Medicine, St. Louis, MO, USA
| | | | - Herbert W Virgin
- Vir Biotechnology, San Francisco, CA, USA
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, USA
- Department of Internal Medicine, UT Southwestern Medical Center, Dallas, TX, USA
| | - Luca Piccoli
- Humabs Biomed SA, Subsidiary of Vir Biotechnology, Bellinzona, Switzerland
| | - Helen Y Chu
- Division of Allergy and Infectious Diseases, University of Washington, Seattle, WA, USA
| | | | - Davide Corti
- Humabs Biomed SA, Subsidiary of Vir Biotechnology, Bellinzona, Switzerland
| | - David Veesler
- Department of Biochemistry, University of Washington, Seattle, WA, USA
- Howard Hughes Medical Institute, University of Washington, Seattle, WA, USA
| |
Collapse
|
19
|
Chunder R, Schropp V, Jabari S, Marzin M, Amor S, Kuerten S. Identification of a novel role for matrix metalloproteinase-3 in the modulation of B cell responses in multiple sclerosis. Front Immunol 2022; 13:1025377. [PMID: 36389698 PMCID: PMC9644161 DOI: 10.3389/fimmu.2022.1025377] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2022] [Accepted: 09/26/2022] [Indexed: 07/30/2023] Open
Abstract
There has been a growing interest in the presence and role of B cell aggregates within the central nervous system of multiple sclerosis patients. However, very little is known about the expression profile of molecules associated with these aggregates and how they might be influencing aggregate development or persistence in the brain. The current study focuses on the effect of matrix metalloproteinase-3, which is associated with B cell aggregates in autopsied multiple sclerosis brain tissue, on B cells. Autopsied brain sections from multiple sclerosis cases and controls were screened for the presence of CD20+ B cell aggregates and expression of matrix metalloproteinase-3. Using flow cytometry, enzyme-linked immunosorbent assay and gene array as methods, in vitro studies were conducted using peripheral blood of healthy volunteers to demonstrate the effect of matrix metalloproteinase-3 on B cells. Autopsied brain sections from multiple sclerosis patients containing aggregates of B cells expressed a significantly higher amount of matrix metalloproteinase-3 compared to controls. In vitro experiments demonstrated that matrix metalloproteinase-3 dampened the overall activation status of B cells by downregulating CD69, CD80 and CD86. Furthermore, matrix metalloproteinase-3-treated B cells produced significantly lower amounts of interleukin-6. Gene array data confirmed that matrix metalloproteinase-3 altered the proliferation and survival profiles of B cells. Taken together, out data indicate a role for B cell modulatory properties of matrix metalloproteinase-3.
Collapse
Affiliation(s)
- Rittika Chunder
- Institute of Neuroanatomy, Medical Faculty, University of Bonn, Bonn, Germany
- Institute of Anatomy and Cell Biology, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Verena Schropp
- Institute of Neuroanatomy, Medical Faculty, University of Bonn, Bonn, Germany
- Institute of Anatomy and Cell Biology, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Samir Jabari
- Institute of Neuropathology, University Hospitals Erlangen, Erlangen, Germany
| | - Manuel Marzin
- Department of Pathology, Amsterdam University Medical Center, Amsterdam, Netherlands
| | - Sandra Amor
- Department of Pathology, Amsterdam University Medical Center, Amsterdam, Netherlands
| | - Stefanie Kuerten
- Institute of Neuroanatomy, Medical Faculty, University of Bonn, Bonn, Germany
- Institute of Anatomy and Cell Biology, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
| |
Collapse
|
20
|
Wang H, Liu H, Zhou L, Wang D, Wang S, Liu Q, Wu Y, Tu M, Sun Z, Zheng X, Fu B, Wang B, Wei H. Cytomegalovirus-specific neutralizing antibodies effectively prevent uncontrolled infection after allogeneic hematopoietic stem cell transplantation. iScience 2022; 25:105065. [PMID: 36147955 PMCID: PMC9485910 DOI: 10.1016/j.isci.2022.105065] [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: 03/28/2022] [Revised: 07/06/2022] [Accepted: 08/30/2022] [Indexed: 11/24/2022] Open
Abstract
Cytomegalovirus (CMV) infection remains one of the most frequent and life-threatening infectious complications after allogeneic hematopoietic stem cell transplantation (allo-HSCT). Herein, we comprehensively compared the immune cells of patients with uncontrolled and controlled CMV infection post-allo-HSCT and found that B-cells were extraordinarily insufficient because of impaired B-cells reconstitution in the uncontrolled infection group. Furthermore, in the controlled infection group, reconstructed B-cells showed signatures of mature B-cells, high expression of CXCR4 and IFITM1, and enrichment of CMV-associated B-cell receptors, which were lacking in the uncontrolled infection group. Consistently, sera from the uncontrolled infection group failed to inhibit CMV infection via neutralizing virus in vitro because of its lower content of anti-CMV-specific immunoglobulin G (IgG) than the controlled infection group. Overall, these results highlighted the contribution of B cells and anti-CMV-specific neutralizing IgGs to the restraint of CMV infection post-allo-HSCT, suggesting their potential as a supplementary treatment to improve outcomes.
Collapse
Affiliation(s)
- Huiru Wang
- Department of Transfusion, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230001, China.,Institute of Immunology, University of Science and Technology of China, Hefei 230027, China
| | - Huilan Liu
- Department of Transfusion, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230001, China.,Department of Hematology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230001, China
| | - Li Zhou
- Institute of Immunology, University of Science and Technology of China, Hefei 230027, China.,Department of Hematology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230001, China
| | - Dongyao Wang
- Institute of Immunology, University of Science and Technology of China, Hefei 230027, China.,Department of Hematology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230001, China
| | - Shushu Wang
- Institute of Immunology, University of Science and Technology of China, Hefei 230027, China.,Department of Pediatrics, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230001, China
| | - Qian Liu
- Organ Transplant Center, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230036, China
| | - Yun Wu
- Department of Hematology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230001, China
| | - Meijuan Tu
- Department of Hematology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230001, China
| | - Zimin Sun
- Department of Hematology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230001, China
| | - Xiaohu Zheng
- Institute of Immunology, University of Science and Technology of China, Hefei 230027, China
| | - Binqing Fu
- Institute of Immunology, University of Science and Technology of China, Hefei 230027, China
| | - Baolong Wang
- Department of Laboratory Medicine, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230001, China
| | - Haiming Wei
- Institute of Immunology, University of Science and Technology of China, Hefei 230027, China.,Department of Geriatrics, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230001, China
| |
Collapse
|
21
|
Thawornpan P, Changrob S, Kochayoo P, Wangriatisak K, Ntumngia FB, De SL, Han ET, Adams JH, Chootong P. Cross-reactive inhibitory antibody and memory B cell responses to variant strains of Duffy binding protein II at post-Plasmodium vivax infection. PLoS One 2022; 17:e0276335. [PMID: 36256619 PMCID: PMC9578595 DOI: 10.1371/journal.pone.0276335] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Accepted: 10/04/2022] [Indexed: 11/05/2022] Open
Abstract
Duffy binding protein region II (DBPII) is considered a strong potential vaccine candidate of blood-stage P. vivax. However, the highly polymorphic nature of this protein often misdirects immune responses, leading them to be strain-specific. Details of cross-reactive humoral immunity to DBPII variants have therefore become an important focus for the development of broadly protective vaccines. Here, cross-reactive humoral immunity against a panel of Thai DBPII variants (DBL-THs) was demonstrated in immunized BALB/c mice and P. vivax patients, by in vitro erythrocyte-binding inhibition assay. Sera from immunized animals showed both strain-transcending (anti-DBL-TH2 and -TH4) and strain-specific (anti-DBL-TH5, -TH6 and -TH9) binding to DBL-TH variants. Using anti-DBL-TH sera at 50% inhibitory concentration (IC50) of the homologous strain, anti-DBL-TH2 sera showed cross inhibition to heterologous DBL-TH strains, whereas anti-DBL-TH5 sera exhibited only strain-specific inhibition. In P. vivax patients, 6 of 15 subjects produced and maintained cross-reactive anti-DBL-TH inhibitory antibodies through the 1-year post-infection timepoint. Cross-reactive memory B cell (MBC) responses to DBL-TH variants were analyzed in subjects recovered from P. vivax infection (RC). The plasma samples from 5 RC subjects showed broad inhibition. However, MBC-derived antibodies of these patients did not reveal cross-inhibition. Altogether, broadly anti-DBP variant inhibitory antibodies developed and persisted in P. vivax infections. However, the presence of cross-reactive anti-DBL-TH inhibitory function post-infection was not related with MBC responses to these variants. More detailed investigation of long-lasting, broadly protective antibodies to DBPII will guide the design of vivax malaria vaccines.
Collapse
Affiliation(s)
- Pongsakorn Thawornpan
- Department of Clinical Microbiology and Applied Technology, Faculty of Medical Technology, Mahidol University, Bangkok, Thailand
| | - Siriruk Changrob
- Department of Clinical Microbiology and Applied Technology, Faculty of Medical Technology, Mahidol University, Bangkok, Thailand
| | - Piyawan Kochayoo
- Department of Clinical Microbiology and Applied Technology, Faculty of Medical Technology, Mahidol University, Bangkok, Thailand
| | - Kittikorn Wangriatisak
- Department of Clinical Microbiology and Applied Technology, Faculty of Medical Technology, Mahidol University, Bangkok, Thailand
| | - Francis B. Ntumngia
- Center for Global Health and Infectious Diseases Research and USF Genomics Program, College of Public Health, University of South Florida, Tampa, Florida, United States of America
| | - Sai Lata De
- Center for Global Health and Infectious Diseases Research and USF Genomics Program, College of Public Health, University of South Florida, Tampa, Florida, United States of America
| | - Eun-Taek Han
- Department of Medical Environmental Biology and Tropical Medicine, School of Medicine, Kangwon National University, Chuncheon, Gangwon-do, Republic of Korea
| | - John H. Adams
- Center for Global Health and Infectious Diseases Research and USF Genomics Program, College of Public Health, University of South Florida, Tampa, Florida, United States of America
| | - Patchanee Chootong
- Department of Clinical Microbiology and Applied Technology, Faculty of Medical Technology, Mahidol University, Bangkok, Thailand
- * E-mail:
| |
Collapse
|
22
|
Marzi R, Bassi J, Silacci-Fregni C, Bartha I, Muoio F, Culap K, Sprugasci N, Lombardo G, Saliba C, Cameroni E, Cassotta A, Low JS, Walls AC, McCallum M, Tortorici MA, Bowen JE, Dellota EA, Dillen JR, Czudnochowski N, Pertusini L, Terrot T, Lepori V, Tarkowski M, Riva A, Biggiogero M, Pellanda AF, Garzoni C, Ferrari P, Ceschi A, Giannini O, Havenar-Daughton C, Telenti A, Arvin A, Virgin HW, Sallusto F, Veesler D, Lanzavecchia A, Corti D, Piccoli L. Maturation of SARS-CoV-2 Spike-specific memory B cells drives resilience to viral escape. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2022:2022.09.30.509852. [PMID: 36203553 PMCID: PMC9536037 DOI: 10.1101/2022.09.30.509852] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
Memory B cells (MBCs) generate rapid antibody responses upon secondary encounter with a pathogen. Here, we investigated the kinetics, avidity and cross-reactivity of serum antibodies and MBCs in 155 SARS-CoV-2 infected and vaccinated individuals over a 16-month timeframe. SARS-CoV-2-specific MBCs and serum antibodies reached steady-state titers with comparable kinetics in infected and vaccinated individuals. Whereas MBCs of infected individuals targeted both pre- and postfusion Spike (S), most vaccine-elicited MBCs were specific for prefusion S, consistent with the use of prefusion-stabilized S in mRNA vaccines. Furthermore, a large fraction of MBCs recognizing postfusion S cross-reacted with human betacoronaviruses. The avidity of MBC-derived and serum antibodies increased over time resulting in enhanced resilience to viral escape by SARS-CoV-2 variants, including Omicron BA.1 and BA.2 sub-lineages, albeit only partially for BA.4 and BA.5 sublineages. Overall, the maturation of high-affinity and broadly-reactive MBCs provides the basis for effective recall responses to future SARS-CoV-2 variants.
Collapse
Affiliation(s)
- Roberta Marzi
- Humabs BioMed SA, a subsidiary of Vir Biotechnology, Bellinzona, Switzerland
| | - Jessica Bassi
- Humabs BioMed SA, a subsidiary of Vir Biotechnology, Bellinzona, Switzerland
| | | | - Istvan Bartha
- Humabs BioMed SA, a subsidiary of Vir Biotechnology, Bellinzona, Switzerland
| | - Francesco Muoio
- Humabs BioMed SA, a subsidiary of Vir Biotechnology, Bellinzona, Switzerland
| | - Katja Culap
- Humabs BioMed SA, a subsidiary of Vir Biotechnology, Bellinzona, Switzerland
| | - Nicole Sprugasci
- Humabs BioMed SA, a subsidiary of Vir Biotechnology, Bellinzona, Switzerland
| | - Gloria Lombardo
- Humabs BioMed SA, a subsidiary of Vir Biotechnology, Bellinzona, Switzerland
| | - Christian Saliba
- Humabs BioMed SA, a subsidiary of Vir Biotechnology, Bellinzona, Switzerland
| | - Elisabetta Cameroni
- Humabs BioMed SA, a subsidiary of Vir Biotechnology, Bellinzona, Switzerland
| | - Antonino Cassotta
- Institute for Research in Biomedicine, Università della Svizzera italiana, Bellinzona, Switzerland
| | - Jun Siong Low
- Institute for Research in Biomedicine, Università della Svizzera italiana, Bellinzona, Switzerland
| | - Alexandra C Walls
- Department of Biochemistry, University of Washington, Seattle, WA, United States of America
| | - Matthew McCallum
- Department of Biochemistry, University of Washington, Seattle, WA, United States of America
| | - M Alejandra Tortorici
- Department of Biochemistry, University of Washington, Seattle, WA, United States of America
| | - John E Bowen
- Department of Biochemistry, University of Washington, Seattle, WA, United States of America
| | | | - Josh R Dillen
- Vir Biotechnology, San Francisco, CA, United States of America
| | | | - Laura Pertusini
- Division of Nephrology, Ente Ospedaliero Cantonale, Lugano, Switzerland
| | - Tatiana Terrot
- Clinical Trial Unit, Ente Ospedaliero Cantonale, Lugano, Switzerland
| | | | - Maciej Tarkowski
- III Division of Infectious Diseases, ASST Fatebenefratelli Sacco, Luigi Sacco Hospital, Milan, Italy
| | - Agostino Riva
- III Division of Infectious Diseases, ASST Fatebenefratelli Sacco, Luigi Sacco Hospital, Milan, Italy
| | - Maira Biggiogero
- Clinic of Internal Medicine and Infectious Diseases, Clinica Luganese Moncucco, Lugano, Switzerland
| | | | - Christian Garzoni
- Clinic of Internal Medicine and Infectious Diseases, Clinica Luganese Moncucco, Lugano, Switzerland
| | - Paolo Ferrari
- Division of Nephrology, Ente Ospedaliero Cantonale, Lugano, Switzerland
- Faculty of Biomedical Sciences, Università della Svizzera italiana, Lugano, Switzerland
- Clinical School, University of New South Wales, Sydney, Australia
| | - Alessandro Ceschi
- Clinical Trial Unit, Ente Ospedaliero Cantonale, Lugano, Switzerland
- Faculty of Biomedical Sciences, Università della Svizzera italiana, Lugano, Switzerland
- Division of Clinical Pharmacology and Toxicology, Institute of Pharmacological Science of Southern Switzerland, Ente Ospedaliero Cantonale, Lugano, Switzerland
- Department of Clinical Pharmacology and Toxicology, University Hospital Zurich, Zurich, Switzerland
| | - Olivier Giannini
- Faculty of Biomedical Sciences, Università della Svizzera italiana, Lugano, Switzerland
- Department of Medicine, Ente Ospedaliero Cantonale, Bellinzona, Switzerland
| | | | - Amalio Telenti
- Vir Biotechnology, San Francisco, CA, United States of America
| | - Ann Arvin
- Vir Biotechnology, San Francisco, CA, United States of America
| | - Herbert W Virgin
- Vir Biotechnology, San Francisco, CA, United States of America
- Department of Pathology and Immunology, Washington University School of Medicine, St Louis, MO, United States of America
- Department of Internal Medicine, UT Southwestern Medical Center, Dallas, TX, United States of America
| | - Federica Sallusto
- Institute for Research in Biomedicine, Università della Svizzera italiana, Bellinzona, Switzerland
- Institute of Microbiology, ETH Zurich, Zurich, Switzerland
| | - David Veesler
- Department of Biochemistry, University of Washington, Seattle, WA, United States of America
| | | | - Davide Corti
- Humabs BioMed SA, a subsidiary of Vir Biotechnology, Bellinzona, Switzerland
| | - Luca Piccoli
- Humabs BioMed SA, a subsidiary of Vir Biotechnology, Bellinzona, Switzerland
| |
Collapse
|
23
|
Park YJ, Pinto D, Walls AC, Liu Z, Marco AD, Benigni F, Zatta F, Silacci-Fregni C, Bassi J, Sprouse KR, Addetia A, Bowen JE, Stewart C, Giurdanella M, Saliba C, Guarino B, Schmid MA, Franko N, Logue J, Dang HV, Hauser K, Iulio JD, Rivera W, Schnell G, Rajesh A, Zhou J, Farhat N, Kaiser H, Montiel-Ruiz M, Noack J, Lempp FA, Janer J, Abdelnabi R, Maes P, Ferrari P, Ceschi A, Giannini O, de Melo GD, Kergoat L, Bourhy H, Neyts J, Soriaga L, Purcell LA, Snell G, Whelan SPJ, Lanzavecchia A, Virgin HW, Piccoli L, Chu H, Pizzuto MS, Corti D, Veesler D. Imprinted antibody responses against SARS-CoV-2 Omicron sublineages. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2022:2022.05.08.491108. [PMID: 35677069 PMCID: PMC9176643 DOI: 10.1101/2022.05.08.491108] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
SARS-CoV-2 Omicron sublineages carry distinct spike mutations and represent an antigenic shift resulting in escape from antibodies induced by previous infection or vaccination. We show that hybrid immunity or vaccine boosters result in potent plasma neutralizing activity against Omicron BA.1 and BA.2 and that breakthrough infections, but not vaccination-only, induce neutralizing activity in the nasal mucosa. Consistent with immunological imprinting, most antibodies derived from memory B cells or plasma cells of Omicron breakthrough cases cross-react with the Wuhan-Hu-1, BA.1 and BA.2 receptor-binding domains whereas Omicron primary infections elicit B cells of narrow specificity. While most clinical antibodies have reduced neutralization of Omicron, we identified an ultrapotent pan-variant antibody, that is unaffected by any Omicron lineage spike mutations and is a strong candidate for clinical development.
Collapse
Affiliation(s)
- Young-Jun Park
- Department of Biochemistry, University of Washington, Seattle, WA 98195, USA
- Howard Hughes Medical Institute, University of Washington, Seattle, WA 98195, USA
| | - Dora Pinto
- Humabs Biomed SA, a subsidiary of Vir Biotechnology, 6500 Bellinzona, Switzerland
| | - Alexandra C Walls
- Department of Biochemistry, University of Washington, Seattle, WA 98195, USA
- Howard Hughes Medical Institute, University of Washington, Seattle, WA 98195, USA
| | - Zhuoming Liu
- Department of Molecular Microbiology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Anna De Marco
- Humabs Biomed SA, a subsidiary of Vir Biotechnology, 6500 Bellinzona, Switzerland
| | - Fabio Benigni
- Humabs Biomed SA, a subsidiary of Vir Biotechnology, 6500 Bellinzona, Switzerland
| | - Fabrizia Zatta
- Humabs Biomed SA, a subsidiary of Vir Biotechnology, 6500 Bellinzona, Switzerland
| | | | - Jessica Bassi
- Humabs Biomed SA, a subsidiary of Vir Biotechnology, 6500 Bellinzona, Switzerland
| | - Kaitlin R Sprouse
- Department of Biochemistry, University of Washington, Seattle, WA 98195, USA
| | - Amin Addetia
- Department of Biochemistry, University of Washington, Seattle, WA 98195, USA
| | - John E Bowen
- Department of Biochemistry, University of Washington, Seattle, WA 98195, USA
| | - Cameron Stewart
- Department of Biochemistry, University of Washington, Seattle, WA 98195, USA
| | - Martina Giurdanella
- Humabs Biomed SA, a subsidiary of Vir Biotechnology, 6500 Bellinzona, Switzerland
| | - Christian Saliba
- Humabs Biomed SA, a subsidiary of Vir Biotechnology, 6500 Bellinzona, Switzerland
| | - Barbara Guarino
- Humabs Biomed SA, a subsidiary of Vir Biotechnology, 6500 Bellinzona, Switzerland
| | - Michael A Schmid
- Humabs Biomed SA, a subsidiary of Vir Biotechnology, 6500 Bellinzona, Switzerland
| | - Nicholas Franko
- Division of Allergy and Infectious Diseases, University of Washington, Seattle, WA 98195, USA
| | - Jennifer Logue
- Division of Allergy and Infectious Diseases, University of Washington, Seattle, WA 98195, USA
| | - Ha V Dang
- Vir Biotechnology, San Francisco, CA 94158, USA
| | | | | | | | | | | | - Jiayi Zhou
- Vir Biotechnology, San Francisco, CA 94158, USA
| | | | | | | | - Julia Noack
- Vir Biotechnology, San Francisco, CA 94158, USA
| | | | - Javier Janer
- Department of Molecular Microbiology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Rana Abdelnabi
- Laboratory of Virology and Chemotherapy, Rega Institute for Medical Research, KU Leuven, 3000 Leuven, Belgium
| | - Piet Maes
- Laboratory of Virology and Chemotherapy, Rega Institute for Medical Research, KU Leuven, 3000 Leuven, Belgium
| | - Paolo Ferrari
- Faculty of Biomedical Sciences, Università della Svizzera italiana, Lugano, Switzerland
- Division of Nephrology, Ente Ospedaliero Cantonale, Lugano, Switzerland
- Clinical School, University of New South Wales, Sydney, New South Wales, Australia
| | - Alessandro Ceschi
- Faculty of Biomedical Sciences, Università della Svizzera italiana, Lugano, Switzerland
- Clinical Trial Unit, Ente Ospedaliero Cantonale, Lugano, Switzerland
- Division of Clinical Pharmacology and Toxicology, Institute of Pharmacological Sciences of Southern Switzerland, Ente Ospedaliero Cantonale, Lugano, Switzerland
- Department of Clinical Pharmacology and Toxicology, University Hospital Zurich, Zurich, Switzerland
| | - Olivier Giannini
- Faculty of Biomedical Sciences, Università della Svizzera italiana, Lugano, Switzerland
- Department of Medicine, Ente Ospedaliero Cantonale, Bellinzona, Switzerland
| | - Guilherme Dias de Melo
- Institut Pasteur, Université de Paris Cité, Lyssavirus Epidemiology and Neuropathology Unit, Paris, F-75015, France
| | - Lauriane Kergoat
- Institut Pasteur, Université de Paris Cité, Lyssavirus Epidemiology and Neuropathology Unit, Paris, F-75015, France
| | - Hervé Bourhy
- Institut Pasteur, Université de Paris Cité, Lyssavirus Epidemiology and Neuropathology Unit, Paris, F-75015, France
| | - Johan Neyts
- Laboratory of Virology and Chemotherapy, Rega Institute for Medical Research, KU Leuven, 3000 Leuven, Belgium
| | | | | | | | - Sean P J Whelan
- Department of Molecular Microbiology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Antonio Lanzavecchia
- Humabs Biomed SA, a subsidiary of Vir Biotechnology, 6500 Bellinzona, Switzerland
| | - Herbert W Virgin
- Vir Biotechnology, San Francisco, CA 94158, USA
- Department of Pathology and Immunology, Washington University School of Medicine, St Louis, MO, USA
- Department of Internal Medicine, UT Southwestern Medical Center, Dallas, TX, USA
| | - Luca Piccoli
- Humabs Biomed SA, a subsidiary of Vir Biotechnology, 6500 Bellinzona, Switzerland
| | - Helen Chu
- Division of Allergy and Infectious Diseases, University of Washington, Seattle, WA 98195, USA
| | | | - Davide Corti
- Humabs Biomed SA, a subsidiary of Vir Biotechnology, 6500 Bellinzona, Switzerland
| | - David Veesler
- Department of Biochemistry, University of Washington, Seattle, WA 98195, USA
- Howard Hughes Medical Institute, University of Washington, Seattle, WA 98195, USA
| |
Collapse
|
24
|
Low JS, Jerak J, Tortorici MA, McCallum M, Pinto D, Cassotta A, Foglierini M, Mele F, Abdelnabi R, Weynand B, Noack J, Montiel-Ruiz M, Bianchi S, Benigni F, Sprugasci N, Joshi A, Bowen JE, Stewart C, Rexhepaj M, Walls AC, Jarrossay D, Morone D, Paparoditis P, Garzoni C, Ferrari P, Ceschi A, Neyts J, Purcell LA, Snell G, Corti D, Lanzavecchia A, Veesler D, Sallusto F. ACE2-binding exposes the SARS-CoV-2 fusion peptide to broadly neutralizing coronavirus antibodies. Science 2022; 377:735-742. [PMID: 35857703 PMCID: PMC9348755 DOI: 10.1126/science.abq2679] [Citation(s) in RCA: 91] [Impact Index Per Article: 45.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Accepted: 07/03/2022] [Indexed: 12/14/2022]
Abstract
The coronavirus spike glycoprotein attaches to host receptors and mediates viral fusion. Using a broad screening approach, we isolated seven monoclonal antibodies (mAbs) that bind to all human-infecting coronavirus spike proteins from severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) immune donors. These mAbs recognize the fusion peptide and acquire affinity and breadth through somatic mutations. Despite targeting a conserved motif, only some mAbs show broad neutralizing activity in vitro against alpha- and betacoronaviruses, including animal coronaviruses WIV-1 and PDF-2180. Two selected mAbs also neutralize Omicron BA.1 and BA.2 authentic viruses and reduce viral burden and pathology in vivo. Structural and functional analyses showed that the fusion peptide-specific mAbs bound with different modalities to a cryptic epitope hidden in prefusion stabilized spike, which became exposed upon binding of angiotensin-converting enzyme 2 (ACE2) or ACE2-mimicking mAbs.
Collapse
Affiliation(s)
- Jun Siong Low
- Institute for Research in Biomedicine, Università della Svizzera Italiana, 6500 Bellinzona, Switzerland
- Institute of Microbiology, ETH Zürich, 8093 Zurich, Switzerland
| | - Josipa Jerak
- Institute for Research in Biomedicine, Università della Svizzera Italiana, 6500 Bellinzona, Switzerland
- Institute of Microbiology, ETH Zürich, 8093 Zurich, Switzerland
| | | | - Matthew McCallum
- Department of Biochemistry, University of Washington, Seattle, WA 98195, USA
| | - Dora Pinto
- Humabs BioMed SA (subsidiary of Vir Biotechnology), 6500 Bellinzona, Switzerland
| | - Antonino Cassotta
- Institute for Research in Biomedicine, Università della Svizzera Italiana, 6500 Bellinzona, Switzerland
| | - Mathilde Foglierini
- Institute for Research in Biomedicine, Università della Svizzera Italiana, 6500 Bellinzona, Switzerland
| | - Federico Mele
- Institute for Research in Biomedicine, Università della Svizzera Italiana, 6500 Bellinzona, Switzerland
| | - Rana Abdelnabi
- KU Leuven Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, Laboratory of Virology and Chemotherapy, B-3000 Leuven, Belgium
| | - Birgit Weynand
- KU Leuven Department of Imaging and Pathology, Translational Cell and Tissue Research, B-3000 Leuven, Belgium
| | - Julia Noack
- Vir Biotechnology, San Francisco, CA 94158, USA
| | | | - Siro Bianchi
- Humabs BioMed SA (subsidiary of Vir Biotechnology), 6500 Bellinzona, Switzerland
| | - Fabio Benigni
- Humabs BioMed SA (subsidiary of Vir Biotechnology), 6500 Bellinzona, Switzerland
| | - Nicole Sprugasci
- Humabs BioMed SA (subsidiary of Vir Biotechnology), 6500 Bellinzona, Switzerland
| | - Anshu Joshi
- Department of Biochemistry, University of Washington, Seattle, WA 98195, USA
| | - John E. Bowen
- Department of Biochemistry, University of Washington, Seattle, WA 98195, USA
| | - Cameron Stewart
- Department of Biochemistry, University of Washington, Seattle, WA 98195, USA
| | - Megi Rexhepaj
- Department of Biochemistry, University of Washington, Seattle, WA 98195, USA
| | - Alexandra C. Walls
- Department of Biochemistry, University of Washington, Seattle, WA 98195, USA
- Howard Hughes Medical Institute, Seattle, WA 98195, USA
| | - David Jarrossay
- Institute for Research in Biomedicine, Università della Svizzera Italiana, 6500 Bellinzona, Switzerland
| | - Diego Morone
- Institute for Research in Biomedicine, Università della Svizzera Italiana, 6500 Bellinzona, Switzerland
| | - Philipp Paparoditis
- Institute for Research in Biomedicine, Università della Svizzera Italiana, 6500 Bellinzona, Switzerland
| | - Christian Garzoni
- Clinic of Internal Medicine and Infectious Diseases, Clinica Luganese Moncucco; 6900 Lugano, Switzerland
| | - Paolo Ferrari
- Faculty of Biomedical Sciences, Università della Svizzera Italiana, 6900 Lugano, Switzerland
- Department of Internal Medicine, Ente Ospedaliero Cantonale, 6500 Bellinzona, Switzerland
- Prince of Wales Hospital Clinical School, University of New South Wales, Sydney, New South Wales 2052, Australia
| | - Alessandro Ceschi
- Faculty of Biomedical Sciences, Università della Svizzera Italiana, 6900 Lugano, Switzerland
- Division of Clinical Pharmacology and Toxicology, Institute of Pharmacological Sciences of Southern Switzerland, Ente Ospedaliero Cantonale, 6900 Lugano, Switzerland
- Clinical Trial Unit, Ente Ospedaliero Cantonale, 6500 Bellinzona, Switzerland
- Department of Clinical Pharmacology and Toxicology, University Hospital Zurich, 8091 Zurich, Switzerland
| | - Johan Neyts
- KU Leuven Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, Laboratory of Virology and Chemotherapy, B-3000 Leuven, Belgium
- Global Virus Network, Baltimore, MD 21201, USA
| | | | | | - Davide Corti
- Humabs BioMed SA (subsidiary of Vir Biotechnology), 6500 Bellinzona, Switzerland
| | - Antonio Lanzavecchia
- Humabs BioMed SA (subsidiary of Vir Biotechnology), 6500 Bellinzona, Switzerland
- National Institute of Molecular Genetics, 20122 Milano, Italy
| | - David Veesler
- Department of Biochemistry, University of Washington, Seattle, WA 98195, USA
- Howard Hughes Medical Institute, Seattle, WA 98195, USA
| | - Federica Sallusto
- Institute for Research in Biomedicine, Università della Svizzera Italiana, 6500 Bellinzona, Switzerland
- Institute of Microbiology, ETH Zürich, 8093 Zurich, Switzerland
| |
Collapse
|
25
|
Novel CD81 Mutations in a Chinese Patient Led to IgA Nephropathy and Impaired BCR Signaling. J Clin Immunol 2022; 42:1672-1684. [PMID: 35849269 DOI: 10.1007/s10875-022-01333-2] [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: 04/28/2022] [Accepted: 07/06/2022] [Indexed: 10/17/2022]
Abstract
PURPOSE CD81 deficiency is an extremely rare primary immunodeficiency disease characterized by severe and recurrent infections, IgA-related nephropathy, and profound hypogammaglobulinemia. Only one patient has been reported so far, and the pathogenesis remains unclear. Here, we identified a new case of CD81 deficiency and described its pathogenesis. METHODS We analyzed the clinical, genetic, and immunological features of the patient with CD81 deficiency, and explored the pathogenesis of her antibody deficiencies. RESULTS The major manifestation of this patient was unexpectedly not recurrent infections but IgA nephropathy with aberrant serum galactose-deficient IgA1. Whole-exome sequencing revealed novel biallelic mutations in CD81 gene that abolished the surface expression of CD81. B cells from the patient lack membrane CD19 and showed reduced switched memory B cells and transitional B cells. Decreased expression of key molecules pY and pBTK in BCR signaling were demonstrated by confocal microscopy. RNA sequencing revealed that genes associated with BCR signaling and immunoglobulins were downregulated in CD81-deficient B cells. In addition, the patient showed increased frequency of T follicular helper cells that biased to Th1-like subsets. CONCLUSION We reported the second patient with CD81 deficiency in the world and illustrated aberrant BCR signaling in the patient, therefore helping to unravel the mechanism of antibody deficiency in CD81-deficient patients.
Collapse
|
26
|
Preisendörfer S, Ishikawa Y, Hennen E, Winklmeier S, Schupp JC, Knüppel L, Fernandez IE, Binzenhöfer L, Flatley A, Juan-Guardela BM, Ruppert C, Guenther A, Frankenberger M, Hatz RA, Kneidinger N, Behr J, Feederle R, Schepers A, Hilgendorff A, Kaminski N, Meinl E, Bächinger HP, Eickelberg O, Staab-Weijnitz CA. FK506-Binding Protein 11 Is a Novel Plasma Cell-Specific Antibody Folding Catalyst with Increased Expression in Idiopathic Pulmonary Fibrosis. Cells 2022; 11:1341. [PMID: 35456020 PMCID: PMC9027113 DOI: 10.3390/cells11081341] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2022] [Revised: 04/08/2022] [Accepted: 04/12/2022] [Indexed: 02/01/2023] Open
Abstract
Antibodies are central effectors of the adaptive immune response, widespread used therapeutics, but also potentially disease-causing biomolecules. Antibody folding catalysts in the plasma cell are incompletely defined. Idiopathic pulmonary fibrosis (IPF) is a fatal chronic lung disease with increasingly recognized autoimmune features. We found elevated expression of FK506-binding protein 11 (FKBP11) in IPF lungs where FKBP11 specifically localized to antibody-producing plasma cells. Suggesting a general role in plasma cells, plasma cell-specific FKBP11 expression was equally observed in lymphatic tissues, and in vitro B cell to plasma cell differentiation was accompanied by induction of FKBP11 expression. Recombinant human FKBP11 was able to refold IgG antibody in vitro and inhibited by FK506, strongly supporting a function as antibody peptidyl-prolyl cis-trans isomerase. Induction of ER stress in cell lines demonstrated induction of FKBP11 in the context of the unfolded protein response in an X-box-binding protein 1 (XBP1)-dependent manner. While deficiency of FKBP11 increased susceptibility to ER stress-mediated cell death in an alveolar epithelial cell line, FKBP11 knockdown in an antibody-producing hybridoma cell line neither induced cell death nor decreased expression or secretion of IgG antibody. Similarly, antibody secretion by the same hybridoma cell line was not affected by knockdown of the established antibody peptidyl-prolyl isomerase cyclophilin B. The results are consistent with FKBP11 as a novel XBP1-regulated antibody peptidyl-prolyl cis-trans isomerase and indicate significant redundancy in the ER-resident folding machinery of antibody-producing hybridoma cells.
Collapse
Affiliation(s)
- Stefan Preisendörfer
- Institute of Lung Health and Immunity and Comprehensive Pneumology Center with the CPC-M bioArchive, Member of the German Center of Lung Research (DZL), Helmholtz-Zentrum München, 81377 Munich, Germany; (S.P.); (E.H.); (L.K.); (I.E.F.); (L.B.); (M.F.); (A.H.); (O.E.)
| | - Yoshihiro Ishikawa
- Department of Biochemistry and Molecular Biology, Oregon Health & Science University, Portland, OR 97239, USA; (Y.I.); (H.P.B.)
| | - Elisabeth Hennen
- Institute of Lung Health and Immunity and Comprehensive Pneumology Center with the CPC-M bioArchive, Member of the German Center of Lung Research (DZL), Helmholtz-Zentrum München, 81377 Munich, Germany; (S.P.); (E.H.); (L.K.); (I.E.F.); (L.B.); (M.F.); (A.H.); (O.E.)
| | - Stephan Winklmeier
- Institute of Clinical Neuroimmunology, Biomedical Center and LMU Klinikum, Ludwig-Maximilians-Universität München, 81377 Munich, Germany; (S.W.); (E.M.)
| | - Jonas C. Schupp
- Pulmonary, Critical Care and Sleep Medicine, Yale School of Medicine, New Haven, CT 06520, USA; (J.C.S.); (B.M.J.-G.); (N.K.)
- Department of Respiratory Medicine, Hannover Medical School, Biomedical Research in End-Stage and Obstructive Lung Disease Hannover, Member of the German Center for Lung Research (DZL), 30625 Hannover, Germany
| | - Larissa Knüppel
- Institute of Lung Health and Immunity and Comprehensive Pneumology Center with the CPC-M bioArchive, Member of the German Center of Lung Research (DZL), Helmholtz-Zentrum München, 81377 Munich, Germany; (S.P.); (E.H.); (L.K.); (I.E.F.); (L.B.); (M.F.); (A.H.); (O.E.)
| | - Isis E. Fernandez
- Institute of Lung Health and Immunity and Comprehensive Pneumology Center with the CPC-M bioArchive, Member of the German Center of Lung Research (DZL), Helmholtz-Zentrum München, 81377 Munich, Germany; (S.P.); (E.H.); (L.K.); (I.E.F.); (L.B.); (M.F.); (A.H.); (O.E.)
- Department of Medicine V, LMU Klinikum, Ludwig-Maximilians-Universität München, Member of the German Center of Lung Research (DZL), 81377 Munich, Germany; (N.K.); (J.B.)
| | - Leonhard Binzenhöfer
- Institute of Lung Health and Immunity and Comprehensive Pneumology Center with the CPC-M bioArchive, Member of the German Center of Lung Research (DZL), Helmholtz-Zentrum München, 81377 Munich, Germany; (S.P.); (E.H.); (L.K.); (I.E.F.); (L.B.); (M.F.); (A.H.); (O.E.)
| | - Andrew Flatley
- Monoclonal Antibody Core Facility, Institute for Diabetes and Obesity, Helmholtz-Zentrum München, 85764 Neuherberg, Germany; (A.F.); (R.F.); (A.S.)
| | - Brenda M. Juan-Guardela
- Pulmonary, Critical Care and Sleep Medicine, Yale School of Medicine, New Haven, CT 06520, USA; (J.C.S.); (B.M.J.-G.); (N.K.)
| | - Clemens Ruppert
- Department of Internal Medicine, Medizinische Klinik II, Member of the German Center of Lung Research (DZL), 35392 Giessen, Germany; (C.R.); (A.G.)
| | - Andreas Guenther
- Department of Internal Medicine, Medizinische Klinik II, Member of the German Center of Lung Research (DZL), 35392 Giessen, Germany; (C.R.); (A.G.)
| | - Marion Frankenberger
- Institute of Lung Health and Immunity and Comprehensive Pneumology Center with the CPC-M bioArchive, Member of the German Center of Lung Research (DZL), Helmholtz-Zentrum München, 81377 Munich, Germany; (S.P.); (E.H.); (L.K.); (I.E.F.); (L.B.); (M.F.); (A.H.); (O.E.)
| | - Rudolf A. Hatz
- Thoraxchirurgisches Zentrum, Klinik für Allgemeine-, Viszeral-, Transplantations-, Gefäß- und Thoraxchirurgie, LMU Klinikum, Ludwig-Maximilians-Universität München, 81377 Munich, Germany;
- Asklepios Fachkliniken München-Gauting, 82131 Gauting, Germany
| | - Nikolaus Kneidinger
- Department of Medicine V, LMU Klinikum, Ludwig-Maximilians-Universität München, Member of the German Center of Lung Research (DZL), 81377 Munich, Germany; (N.K.); (J.B.)
| | - Jürgen Behr
- Department of Medicine V, LMU Klinikum, Ludwig-Maximilians-Universität München, Member of the German Center of Lung Research (DZL), 81377 Munich, Germany; (N.K.); (J.B.)
| | - Regina Feederle
- Monoclonal Antibody Core Facility, Institute for Diabetes and Obesity, Helmholtz-Zentrum München, 85764 Neuherberg, Germany; (A.F.); (R.F.); (A.S.)
| | - Aloys Schepers
- Monoclonal Antibody Core Facility, Institute for Diabetes and Obesity, Helmholtz-Zentrum München, 85764 Neuherberg, Germany; (A.F.); (R.F.); (A.S.)
| | - Anne Hilgendorff
- Institute of Lung Health and Immunity and Comprehensive Pneumology Center with the CPC-M bioArchive, Member of the German Center of Lung Research (DZL), Helmholtz-Zentrum München, 81377 Munich, Germany; (S.P.); (E.H.); (L.K.); (I.E.F.); (L.B.); (M.F.); (A.H.); (O.E.)
| | - Naftali Kaminski
- Pulmonary, Critical Care and Sleep Medicine, Yale School of Medicine, New Haven, CT 06520, USA; (J.C.S.); (B.M.J.-G.); (N.K.)
| | - Edgar Meinl
- Institute of Clinical Neuroimmunology, Biomedical Center and LMU Klinikum, Ludwig-Maximilians-Universität München, 81377 Munich, Germany; (S.W.); (E.M.)
| | - Hans Peter Bächinger
- Department of Biochemistry and Molecular Biology, Oregon Health & Science University, Portland, OR 97239, USA; (Y.I.); (H.P.B.)
| | - Oliver Eickelberg
- Institute of Lung Health and Immunity and Comprehensive Pneumology Center with the CPC-M bioArchive, Member of the German Center of Lung Research (DZL), Helmholtz-Zentrum München, 81377 Munich, Germany; (S.P.); (E.H.); (L.K.); (I.E.F.); (L.B.); (M.F.); (A.H.); (O.E.)
| | - Claudia A. Staab-Weijnitz
- Institute of Lung Health and Immunity and Comprehensive Pneumology Center with the CPC-M bioArchive, Member of the German Center of Lung Research (DZL), Helmholtz-Zentrum München, 81377 Munich, Germany; (S.P.); (E.H.); (L.K.); (I.E.F.); (L.B.); (M.F.); (A.H.); (O.E.)
| |
Collapse
|
27
|
Lyski ZL, Brunton AE, Strnad MI, Sullivan PE, Siegel SAR, Tafesse FG, Slifka MK, Messer WB. Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2)-Specific Memory B Cells From Individuals With Diverse Disease Severities Recognize SARS-CoV-2 Variants of Concern. J Infect Dis 2022; 225:947-956. [PMID: 34865053 PMCID: PMC8922005 DOI: 10.1093/infdis/jiab585] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2021] [Accepted: 11/29/2021] [Indexed: 11/12/2022] Open
Abstract
The unprecedented severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) pandemic has called for substantial investigations into the capacity of the human immune system to protect against reinfection and keep pace with the evolution of SARS-CoV-2. We evaluated the magnitude and durability of the SARS-CoV-2-specific antibody responses against parental WA-1 SARS-CoV-2 receptor-binding domain (RBD) and a representative variant of concern (VoC) RBD using antibodies from 2 antibody compartments: long-lived plasma cell-derived plasma antibodies and antibodies encoded by SARS-CoV-2-specific memory B cells (MBCs). Thirty-five participants naturally infected with SARS-CoV-2 were evaluated; although only 25 of 35 participants had VoC RBD-reactive plasma antibodies, 34 of 35 (97%) participants had VoC RBD-reactive MBC-derived antibodies. Our finding that 97% of previously infected individuals have MBCs specific for variant RBDs provides reason for optimism regarding the capacity of vaccination, prior infection, and/or both, to elicit immunity with the capacity to limit disease severity and transmission of VoCs as they arise and circulate.
Collapse
Affiliation(s)
- Zoe L Lyski
- Department of Molecular Microbiology and Immunology, Oregon Health and Science University, Portland, Oregon, USA
| | - Amanda E Brunton
- Oregon Health and Science University–Portland State University School of Public Health, Portland, Oregon, USA
| | - Matt I Strnad
- Oregon Health and Science University–Portland State University School of Public Health, Portland, Oregon, USA
| | - Peter E Sullivan
- Oregon Health and Science University–Portland State University School of Public Health, Portland, Oregon, USA
| | - Sarah A R Siegel
- Oregon Health and Science University–Portland State University School of Public Health, Portland, Oregon, USA
| | - Fikadu G Tafesse
- Department of Molecular Microbiology and Immunology, Oregon Health and Science University, Portland, Oregon, USA
| | - Mark K Slifka
- Division of Neuroscience, Oregon National Primate Research Center, Oregon Health and Science University, Beaverton, Oregon, USA
| | - William B Messer
- Department of Molecular Microbiology and Immunology, Oregon Health and Science University, Portland, Oregon, USA
- Oregon Health and Science University–Portland State University School of Public Health, Portland, Oregon, USA
- Department of Medicine, Division of Infectious Diseases, Oregon Health and Science University, Portland, Oregon, USA
| |
Collapse
|
28
|
Karagiannis P, Correa I, Chauhan J, Cheung A, Dominguez-Rodriguez D, Terranova-Barberio M, Harris RJ, Crescioli S, Spicer J, Bokemeyer C, Lacy KE, Karagiannis SN. Innate stimulation of B cells ex vivo enhances antibody secretion and identifies tumour-reactive antibodies from cancer patients. Clin Exp Immunol 2022; 207:84-94. [PMID: 35020866 PMCID: PMC8802180 DOI: 10.1093/cei/uxab005] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2021] [Revised: 09/26/2021] [Accepted: 10/14/2021] [Indexed: 11/13/2022] Open
Abstract
Human B cells and their expressed antibodies are crucial in conferring immune protection. Identifying pathogen-specific antibodies following infection is possible due to enhanced humoral immunity against well-described molecules on the pathogen surface. However, screening for cancer-reactive antibodies remains challenging since target antigens are often not identified a priori and the frequency of circulating B cells recognizing cancer cells is likely very low. We investigated whether combined ex vivo culture of human B cells with three innate stimuli, interleukin-17 (IL-17), B-cell activation factor (BAFF), and the toll-like receptor 9 (TLR-9) agonist DNA motif CpG ODN 2006 (CpG), each known to activate B cells through different signalling pathways, promote cell activation, proliferation, and antibody production. Combined IL-17+BAFF+CpG prolonged B-cell survival and increased proliferation compared with single stimuli. IL-17+BAFF+CpG triggered higher IgG secretion, likely by activating differentiated, memory and class-switched CD19+CD20+CD27+IgD- B cells. Regardless of anti-FOLR antibody seropositive status, IL-17+BAFF+CpG combined with a monovalent tumour-associated antigen (folate receptor alpha [FOLR]) led to secreted antibodies recognizing the antigen and the antigen-expressing IGROV1 cancer cells. In a seropositive individual, FOLR stimulation favoured class-switched memory B-cell precursors (CD27-CD38-IgD-), class-switched memory B cells and anti-FOLR antibody production, while IL-17+BAFF+CpG combined with FOLR, promoted class-switched memory B-cell precursors and antibody-secreting (CD138+IgD-) plasma cells. Furthermore, IL-17+BAFF+CpG stimulation of peripheral blood B cells from patients with melanoma revealed tumour cell-reactive antibodies in culture supernatants. These findings suggest that innate signals stimulate B-cell survival and antibody production and may help identify low-frequency antigen-reactive humoral responses.
Collapse
Affiliation(s)
- Panagiotis Karagiannis
- Department of Oncology, Haematology and Bone Marrow Transplantation with Section of Pneumology, University Medical Centre Hamburg-Eppendorf, Hamburg, Germany.,St. John's Institute of Dermatology, School of Basic and Medical Biosciences, King's College London, Guy's Hospital, London, UK
| | - Isabel Correa
- St. John's Institute of Dermatology, School of Basic and Medical Biosciences, King's College London, Guy's Hospital, London, UK
| | - Jitesh Chauhan
- St. John's Institute of Dermatology, School of Basic and Medical Biosciences, King's College London, Guy's Hospital, London, UK
| | - Anthony Cheung
- St. John's Institute of Dermatology, School of Basic and Medical Biosciences, King's College London, Guy's Hospital, London, UK.,Breast Cancer Now Research Unit, School of Cancer and Pharmaceutical Sciences, King's College London, Guy's Cancer Centre, London, UK
| | - Diana Dominguez-Rodriguez
- St. John's Institute of Dermatology, School of Basic and Medical Biosciences, King's College London, Guy's Hospital, London, UK
| | - Manuela Terranova-Barberio
- St. John's Institute of Dermatology, School of Basic and Medical Biosciences, King's College London, Guy's Hospital, London, UK
| | - Robert J Harris
- St. John's Institute of Dermatology, School of Basic and Medical Biosciences, King's College London, Guy's Hospital, London, UK
| | - Silvia Crescioli
- St. John's Institute of Dermatology, School of Basic and Medical Biosciences, King's College London, Guy's Hospital, London, UK
| | - James Spicer
- School of Cancer and Pharmaceutical Sciences, King's College London, Guy's Hospital, London, UK
| | - Carsten Bokemeyer
- Department of Oncology, Haematology and Bone Marrow Transplantation with Section of Pneumology, University Medical Centre Hamburg-Eppendorf, Hamburg, Germany
| | - Katie E Lacy
- St. John's Institute of Dermatology, School of Basic and Medical Biosciences, King's College London, Guy's Hospital, London, UK
| | - Sophia N Karagiannis
- St. John's Institute of Dermatology, School of Basic and Medical Biosciences, King's College London, Guy's Hospital, London, UK.,Breast Cancer Now Research Unit, School of Cancer and Pharmaceutical Sciences, King's College London, Guy's Cancer Centre, London, UK
| |
Collapse
|
29
|
Winklmeier S, Eisenhut K, Taskin D, Rübsamen H, Gerhards R, Schneider C, Wratil PR, Stern M, Eichhorn P, Keppler OT, Klein M, Mader S, Kümpfel T, Meinl E. Persistence of functional memory B cells recognizing SARS-CoV-2 variants despite loss of specific IgG. iScience 2022; 25:103659. [PMID: 34957380 PMCID: PMC8686444 DOI: 10.1016/j.isci.2021.103659] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2021] [Revised: 11/17/2021] [Accepted: 12/15/2021] [Indexed: 01/22/2023] Open
Abstract
Although some COVID-19 patients maintain SARS-CoV-2-specific serum immunoglobulin G (IgG) for more than 6 months postinfection, others eventually lose IgG levels. We assessed the persistence of SARS-CoV-2-specific B cells in 17 patients, 5 of whom had lost specific IgGs after 5-8 months. Differentiation of blood-derived B cells in vitro revealed persistent SARS-CoV-2-specific IgG B cells in all patients, whereas IgA B cells were maintained in 11. Antibodies derived from cultured B cells blocked binding of viral receptor-binding domain (RBD) to the cellular receptor ACE-2, had neutralizing activity to authentic virus, and recognized the RBD of the variant of concern Alpha similarly to the wild type, whereas reactivity to Beta and Gamma were decreased. Thus, differentiation of memory B cells could be more sensitive for detecting previous infection than measuring serum antibodies. Understanding the persistence of SARS-CoV-2-specific B cells even in the absence of specific serum IgG will help to promote long-term immunity.
Collapse
Affiliation(s)
- Stephan Winklmeier
- Institute of Clinical Neuroimmunology, University Hospital, LMU Munich, 81377 Munich, Germany
- Biomedical Center (BMC), Faculty of Medicine, LMU Munich, 82152 Martinsried, Germany
| | - Katharina Eisenhut
- Institute of Clinical Neuroimmunology, University Hospital, LMU Munich, 81377 Munich, Germany
- Biomedical Center (BMC), Faculty of Medicine, LMU Munich, 82152 Martinsried, Germany
| | - Damla Taskin
- Institute of Clinical Neuroimmunology, University Hospital, LMU Munich, 81377 Munich, Germany
- Biomedical Center (BMC), Faculty of Medicine, LMU Munich, 82152 Martinsried, Germany
| | - Heike Rübsamen
- Institute of Clinical Neuroimmunology, University Hospital, LMU Munich, 81377 Munich, Germany
- Biomedical Center (BMC), Faculty of Medicine, LMU Munich, 82152 Martinsried, Germany
| | - Ramona Gerhards
- Institute of Clinical Neuroimmunology, University Hospital, LMU Munich, 81377 Munich, Germany
- Biomedical Center (BMC), Faculty of Medicine, LMU Munich, 82152 Martinsried, Germany
| | - Celine Schneider
- Institute of Clinical Neuroimmunology, University Hospital, LMU Munich, 81377 Munich, Germany
- Biomedical Center (BMC), Faculty of Medicine, LMU Munich, 82152 Martinsried, Germany
| | - Paul R. Wratil
- Max von Pettenkofer Institute & GeneCenter, Virology, LMU Munich, 80336 Munich, Germany
| | - Marcel Stern
- Max von Pettenkofer Institute & GeneCenter, Virology, LMU Munich, 80336 Munich, Germany
| | - Peter Eichhorn
- Institute of Laboratory Medicine, University Hospital, LMU Munich, 81377 Munich, Germany
| | - Oliver T. Keppler
- Max von Pettenkofer Institute & GeneCenter, Virology, LMU Munich, 80336 Munich, Germany
| | - Matthias Klein
- Department of Neurology, University Hospital, LMU Munich, 81377 Munich, Germany
| | - Simone Mader
- Institute of Clinical Neuroimmunology, University Hospital, LMU Munich, 81377 Munich, Germany
- Biomedical Center (BMC), Faculty of Medicine, LMU Munich, 82152 Martinsried, Germany
| | - Tania Kümpfel
- Institute of Clinical Neuroimmunology, University Hospital, LMU Munich, 81377 Munich, Germany
- Biomedical Center (BMC), Faculty of Medicine, LMU Munich, 82152 Martinsried, Germany
| | - Edgar Meinl
- Institute of Clinical Neuroimmunology, University Hospital, LMU Munich, 81377 Munich, Germany
- Biomedical Center (BMC), Faculty of Medicine, LMU Munich, 82152 Martinsried, Germany
- Corresponding author
| |
Collapse
|
30
|
Matochko WL, Nelep C, Chen WC, Grauer S, McFadden K, Wilson V, Oxenoid K. CellCelector™ as a platform in isolating primary B cells for antibody discovery. Antib Ther 2022; 5:11-17. [PMID: 35059561 PMCID: PMC8764991 DOI: 10.1093/abt/tbab030] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2021] [Revised: 12/10/2021] [Accepted: 12/17/2021] [Indexed: 01/02/2023] Open
Abstract
The most robust strategy in antibody discovery is the use of immunized animals and the ability to isolate and immortalize immune B-cells to hybridoma for further interrogation. However, capturing the full repertoire of an immunized animal is labor intensive, time consuming and limited in throughput. Therefore, techniques to directly mine the antibody repertoire of primary B-cells are of great importance in antibody discovery. In the current study, we present a method to isolate individual antigen-specific primary B-cells using the CellCellector™ single-cell isolation platform from XenoMouse® (XM) immunized with a recombinant therapeutic protein, EGFR. We screened a subset of CD138+ B-cells and identified 238 potential EGFR-specific B-cells from 1189 antibody-secreting cells (ASCs) and isolated 94 by CellCellector. We identified a diverse set of heavy chain complementarity-determining region sequences and cloned and expressed 20 into a standard human immunoglobulin G1 antibody format. We further characterized and identified 13 recombinant antibodies that engage soluble and native forms of EGFR. By extrapolating the method to all 400 000 CD138+ B-cells extracted from one EGFR immunized XM, a potential 1196 unique EGFR-specific antibodies could be discovered. CellCelector allows for interrogating the B-cell pool directly and isolating B-cells specific to the therapeutic target of interest. Furthermore, antibody sequences recovered from isolated B-cells engage the native and recombinant target, demonstrating the CellCellector can serve as a platform in antibody discovery.
Collapse
Affiliation(s)
- Wadim L Matochko
- Biologics Discovery, Department of Therapeutic Discovery, Amgen British Columbia Inc., Burnaby, Canada
| | - Constantin Nelep
- Marketing and Application Development, ALS Automated Lab Solutions GmbH, Jena, Germany
| | - Weihsu C Chen
- Biologics Discovery, Department of Therapeutic Discovery, Amgen British Columbia Inc., Burnaby, Canada
| | - Stephanie Grauer
- Biologics Discovery, Department of Therapeutic Discovery, Amgen British Columbia Inc., Burnaby, Canada
| | - Karyn McFadden
- Biologics Discovery, Department of Therapeutic Discovery, Amgen British Columbia Inc., Burnaby, Canada
| | - Vicki Wilson
- Biologics Discovery, Department of Therapeutic Discovery, Amgen British Columbia Inc., Burnaby, Canada
| | - Kirill Oxenoid
- Biologics Discovery, Department of Therapeutic Discovery, Amgen British Columbia Inc., Burnaby, Canada
| |
Collapse
|
31
|
Safety and Immunogenicity of M2-Deficient, Single Replication, Live Influenza Vaccine (M2SR) in Adults. Vaccines (Basel) 2021; 9:vaccines9121388. [PMID: 34960134 PMCID: PMC8707871 DOI: 10.3390/vaccines9121388] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2021] [Revised: 11/09/2021] [Accepted: 11/18/2021] [Indexed: 11/17/2022] Open
Abstract
M2SR (M2-deficient single replication) is an investigational live intranasal vaccine that protects against multiple influenza A subtypes in influenza-naïve and previously infected ferrets. We conducted a phase 1, first-in-human, randomized, dose-escalation, placebo-controlled study of M2SR safety and immunogenicity. Adult subjects received a single intranasal administration with either placebo or one of three M2SR dose levels (106, 107 or 108 tissue culture infectious dose (TCID50)) expressing hemagglutinin and neuraminidase from A/Brisbane/10/2007 (H3N2) (24 subjects per group). Subjects were evaluated for virus replication, local and systemic reactions, adverse events (AE), and immune responses post-vaccination. Infectious virus was not detected in nasal swabs from vaccinated subjects. At least one AE (most commonly mild nasal rhinorrhea/congestion) was reported among 29%, 58%, and 83% of M2SR subjects administered a low, medium or high dose, respectively, and among 46% of placebo subjects. No subject had fever or a severe reaction to the vaccine. Influenza-specific serum and mucosal antibody responses and B- and T-cell responses were significantly more frequent among vaccinated subjects vs. placebo recipients. The M2SR vaccine was safe and well tolerated and generated dose-dependent durable serum antibody responses against diverse H3N2 influenza strains. M2SR demonstrated a multi-faceted immune response in seronegative and seropositive subjects.
Collapse
|
32
|
Pinto D, Sauer MM, Czudnochowski N, Low JS, Tortorici MA, Housley MP, Noack J, Walls AC, Bowen JE, Guarino B, Rosen LE, di Iulio J, Jerak J, Kaiser H, Islam S, Jaconi S, Sprugasci N, Culap K, Abdelnabi R, Foo C, Coelmont L, Bartha I, Bianchi S, Silacci-Fregni C, Bassi J, Marzi R, Vetti E, Cassotta A, Ceschi A, Ferrari P, Cippà PE, Giannini O, Ceruti S, Garzoni C, Riva A, Benigni F, Cameroni E, Piccoli L, Pizzuto MS, Smithey M, Hong D, Telenti A, Lempp FA, Neyts J, Havenar-Daughton C, Lanzavecchia A, Sallusto F, Snell G, Virgin HW, Beltramello M, Corti D, Veesler D. Broad betacoronavirus neutralization by a stem helix-specific human antibody. Science 2021; 373:1109-1116. [PMID: 34344823 PMCID: PMC9268357 DOI: 10.1126/science.abj3321] [Citation(s) in RCA: 270] [Impact Index Per Article: 90.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2021] [Accepted: 07/29/2021] [Indexed: 12/11/2022]
Abstract
The spillovers of betacoronaviruses in humans and the emergence of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) variants highlight the need for broad coronavirus countermeasures. We describe five monoclonal antibodies (mAbs) cross-reacting with the stem helix of multiple betacoronavirus spike glycoproteins isolated from COVID-19 convalescent individuals. Using structural and functional studies, we show that the mAb with the greatest breadth (S2P6) neutralizes pseudotyped viruses from three different subgenera through the inhibition of membrane fusion, and we delineate the molecular basis for its cross-reactivity. S2P6 reduces viral burden in hamsters challenged with SARS-CoV-2 through viral neutralization and Fc-mediated effector functions. Stem helix antibodies are rare, oftentimes of narrow specificity, and can acquire neutralization breadth through somatic mutations. These data provide a framework for structure-guided design of pan-betacoronavirus vaccines eliciting broad protection.
Collapse
Affiliation(s)
- Dora Pinto
- Humabs Biomed SA, a subsidiary of Vir Biotechnology, 6500 Bellinzona, Switzerland
| | - Maximilian M. Sauer
- Department of Biochemistry, University of Washington, Seattle, WA 98195, USA
| | | | - Jun Siong Low
- Institute for Research in Biomedicine, Università della Svizzera italiana, 6500 Bellinzona, Switzerland
| | | | | | - Julia Noack
- Vir Biotechnology, San Francisco, CA 94158, USA
| | - Alexandra C. Walls
- Department of Biochemistry, University of Washington, Seattle, WA 98195, USA
| | - John E. Bowen
- Department of Biochemistry, University of Washington, Seattle, WA 98195, USA
| | - Barbara Guarino
- Humabs Biomed SA, a subsidiary of Vir Biotechnology, 6500 Bellinzona, Switzerland
| | | | | | - Josipa Jerak
- Institute for Research in Biomedicine, Università della Svizzera italiana, 6500 Bellinzona, Switzerland
| | | | | | - Stefano Jaconi
- Humabs Biomed SA, a subsidiary of Vir Biotechnology, 6500 Bellinzona, Switzerland
| | - Nicole Sprugasci
- Humabs Biomed SA, a subsidiary of Vir Biotechnology, 6500 Bellinzona, Switzerland
| | - Katja Culap
- Humabs Biomed SA, a subsidiary of Vir Biotechnology, 6500 Bellinzona, Switzerland
| | - Rana Abdelnabi
- Rega Institute for Medical Research, Laboratory of Virology and Chemotherapy, KU Leuven, 3000 Leuven, Belgium
| | - Caroline Foo
- Rega Institute for Medical Research, Laboratory of Virology and Chemotherapy, KU Leuven, 3000 Leuven, Belgium
| | - Lotte Coelmont
- Rega Institute for Medical Research, Laboratory of Virology and Chemotherapy, KU Leuven, 3000 Leuven, Belgium
| | - Istvan Bartha
- Humabs Biomed SA, a subsidiary of Vir Biotechnology, 6500 Bellinzona, Switzerland
| | - Siro Bianchi
- Humabs Biomed SA, a subsidiary of Vir Biotechnology, 6500 Bellinzona, Switzerland
| | | | - Jessica Bassi
- Humabs Biomed SA, a subsidiary of Vir Biotechnology, 6500 Bellinzona, Switzerland
| | - Roberta Marzi
- Humabs Biomed SA, a subsidiary of Vir Biotechnology, 6500 Bellinzona, Switzerland
| | - Eneida Vetti
- Humabs Biomed SA, a subsidiary of Vir Biotechnology, 6500 Bellinzona, Switzerland
| | - Antonino Cassotta
- Institute for Research in Biomedicine, Università della Svizzera italiana, 6500 Bellinzona, Switzerland
| | - Alessandro Ceschi
- Clinical Trial Unit, Ente Ospedaliero Cantonale, 6900 Lugano, Switzerland
- Division of Clinical Pharmacology and Toxicology, Institute of Pharmacological Sciences of Southern Switzerland, Ente Ospedaliero Cantonale, 6900 Lugano, Switzerland
- Department of Clinical Pharmacology and Toxicology, University Hospital Zurich, 8091 Zurich, Switzerland
- Faculty of Biomedical Sciences, Università della Svizzera italiana, 6900 Lugano, Switzerland
| | - Paolo Ferrari
- Faculty of Biomedical Sciences, Università della Svizzera italiana, 6900 Lugano, Switzerland
- Department of Medicine, Ente Ospedaliero Cantonale, 6500 Bellinzona, Switzerland
- Clinical School, University of New South Wales, Sydney, NSW, 2052, Australia
| | - Pietro E. Cippà
- Department of Medicine, Ente Ospedaliero Cantonale, 6500 Bellinzona, Switzerland
- Faculty of Medicine, University of Zurich, 8057 Zurich, Switzerland
| | - Olivier Giannini
- Faculty of Biomedical Sciences, Università della Svizzera italiana, 6900 Lugano, Switzerland
- Department of Medicine, Ente Ospedaliero Cantonale, 6500 Bellinzona, Switzerland
| | - Samuele Ceruti
- Intensive Care Unit, Clinica Luganese Moncucco, 6900 Lugano, Switzerland
| | - Christian Garzoni
- Clinic of Internal Medicine and Infectious Diseases, Clinica Luganese Moncucco, 6900 Lugano, Switzerland
| | - Agostino Riva
- III Division of Infectious Diseases, ASST Fatebenefratelli Sacco, Luigi Sacco Hospital, 20157 Milan, Italy
| | - Fabio Benigni
- Humabs Biomed SA, a subsidiary of Vir Biotechnology, 6500 Bellinzona, Switzerland
| | - Elisabetta Cameroni
- Humabs Biomed SA, a subsidiary of Vir Biotechnology, 6500 Bellinzona, Switzerland
| | - Luca Piccoli
- Humabs Biomed SA, a subsidiary of Vir Biotechnology, 6500 Bellinzona, Switzerland
| | - Matteo S. Pizzuto
- Humabs Biomed SA, a subsidiary of Vir Biotechnology, 6500 Bellinzona, Switzerland
| | | | - David Hong
- Vir Biotechnology, San Francisco, CA 94158, USA
| | | | | | - Johan Neyts
- Rega Institute for Medical Research, Laboratory of Virology and Chemotherapy, KU Leuven, 3000 Leuven, Belgium
| | | | - Antonio Lanzavecchia
- Humabs Biomed SA, a subsidiary of Vir Biotechnology, 6500 Bellinzona, Switzerland
| | - Federica Sallusto
- Institute for Research in Biomedicine, Università della Svizzera italiana, 6500 Bellinzona, Switzerland
- Institute of Microbiology, ETH Zurich, 8093 Zurich, Switzerland
| | | | - Herbert W. Virgin
- Vir Biotechnology, San Francisco, CA 94158, USA
- UT Southwestern Medical Center, Dallas, TX 75390, USA
- Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Martina Beltramello
- Humabs Biomed SA, a subsidiary of Vir Biotechnology, 6500 Bellinzona, Switzerland
| | - Davide Corti
- Humabs Biomed SA, a subsidiary of Vir Biotechnology, 6500 Bellinzona, Switzerland
| | - David Veesler
- Department of Biochemistry, University of Washington, Seattle, WA 98195, USA
| |
Collapse
|
33
|
Bohannon CD, Ende Z, Cao W, Mboko WP, Ranjan P, Kumar A, Mishina M, Amoah S, Gangappa S, Mittal SK, Lovell JF, García‐Sastre A, Pfeifer BA, Davidson BA, Knight P, Sambhara S. Influenza Virus Infects and Depletes Activated Adaptive Immune Responders. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2021; 8:e2100693. [PMID: 34189857 PMCID: PMC8373117 DOI: 10.1002/advs.202100693] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/19/2021] [Revised: 05/18/2021] [Indexed: 05/14/2023]
Abstract
Influenza infections cause several million cases of severe respiratory illness, hospitalizations, and hundreds of thousands of deaths globally. Secondary infections are a leading cause of influenza's high morbidity and mortality, and significantly factored into the severity of the 1918, 1968, and 2009 pandemics. Furthermore, there is an increased incidence of other respiratory infections even in vaccinated individuals during influenza season. Putative mechanisms responsible for vaccine failures against influenza as well as other respiratory infections during influenza season are investigated. Peripheral blood mononuclear cells (PBMCs) are used from influenza vaccinated individuals to assess antigen-specific responses to influenza, measles, and varicella. The observations made in humans to a mouse model to unravel the mechanism is confirmed and extended. Infection with influenza virus suppresses an ongoing adaptive response to vaccination against influenza as well as other respiratory pathogens, i.e., Adenovirus and Streptococcus pneumoniae by preferentially infecting and killing activated lymphocytes which express elevated levels of sialic acid receptors. These findings propose a new mechanism for the high incidence of secondary respiratory infections due to bacteria and other viruses as well as vaccine failures to influenza and other respiratory pathogens even in immune individuals due to influenza viral infections.
Collapse
Affiliation(s)
- Caitlin D. Bohannon
- Influenza DivisionCenters for Disease Control and PreventionAtlantaGA30329USA
- Oak Ridge Institute for Science and Education (ORISE)CDC Fellowship ProgramOak RidgeTN37831USA
| | - Zachary Ende
- Influenza DivisionCenters for Disease Control and PreventionAtlantaGA30329USA
- Oak Ridge Institute for Science and Education (ORISE)CDC Fellowship ProgramOak RidgeTN37831USA
| | - Weiping Cao
- Influenza DivisionCenters for Disease Control and PreventionAtlantaGA30329USA
| | - Wadzanai P. Mboko
- Influenza DivisionCenters for Disease Control and PreventionAtlantaGA30329USA
- Department of Comparative Pathobiology and Purdue Institute for InflammationImmunologyand Infectious DiseasePurdue UniversityWest LafayetteIN47907USA
| | - Priya Ranjan
- Influenza DivisionCenters for Disease Control and PreventionAtlantaGA30329USA
| | - Amrita Kumar
- Influenza DivisionCenters for Disease Control and PreventionAtlantaGA30329USA
| | - Margarita Mishina
- Influenza DivisionCenters for Disease Control and PreventionAtlantaGA30329USA
| | - Samuel Amoah
- Influenza DivisionCenters for Disease Control and PreventionAtlantaGA30329USA
| | | | - Suresh K. Mittal
- Department of Comparative Pathobiology and Purdue Institute for InflammationImmunologyand Infectious DiseasePurdue UniversityWest LafayetteIN47907USA
| | - Jonathan F. Lovell
- Department of Biomedical EngineeringState University of New York at BuffaloBuffaloNY14260USA
| | - Adolfo García‐Sastre
- Global Health and Emerging Pathogens InstituteIcahn School of Medicine at Mount SinaiNew YorkNY10029USA
- Department of MicrobiologyIcahn School of Medicine at Mount SinaiNew YorkNY10029USA
- Department of MedicineDivision of Infectious DiseasesIcahn School of Medicine at Mount SinaiNew YorkNY10029USA
- The Tisch Cancer InstituteIcahn School of Medicine at Mount SinaiNew YorkNY10029USA
| | - Blaine A. Pfeifer
- Department of Chemical and Biological EngineeringSchool of Engineering and Applied SciencesState University of New York at BuffaloBuffaloNY14260USA
| | - Bruce A. Davidson
- Department of AnesthesiologyJacobs School of Medicine and Biomedical SciencesState University of New York at BuffaloBuffaloNY14260USA
- Department of Pathology and Anatomical SciencesSchool of Medicine and Biomedical SciencesState University of New York at BuffaloBuffaloNY14260USA
- Research ServiceVeterans AdministrationWestern New York Healthcare SystemBuffaloNY14215USA
| | - Paul Knight
- Department of AnesthesiologyJacobs School of Medicine and Biomedical SciencesState University of New York at BuffaloBuffaloNY14260USA
| | | |
Collapse
|
34
|
Köppert S, Wolf C, Becza N, Sautto GA, Franke F, Kuerten S, Ross TM, Lehmann PV, Kirchenbaum GA. Affinity Tag Coating Enables Reliable Detection of Antigen-Specific B Cells in Immunospot Assays. Cells 2021; 10:cells10081843. [PMID: 34440612 PMCID: PMC8394687 DOI: 10.3390/cells10081843] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Revised: 07/09/2021] [Accepted: 07/16/2021] [Indexed: 11/26/2022] Open
Abstract
Assessment of humoral immunity to SARS-CoV-2 and other infectious agents is typically restricted to detecting antigen-specific antibodies in the serum. Rarely does immune monitoring entail assessment of the memory B-cell compartment itself, although it is these cells that engage in secondary antibody responses capable of mediating immune protection when pre-existing antibodies fail to prevent re-infection. There are few techniques that are capable of detecting rare antigen-specific B cells while also providing information regarding their relative abundance, class/subclass usage and functional affinity. In theory, the ELISPOT/FluoroSpot (collectively ImmunoSpot) assay platform is ideally suited for antigen-specific B-cell assessments since it provides this information at single-cell resolution for individual antibody-secreting cells (ASC). Here, we tested the hypothesis that antigen-coating efficiency could be universally improved across a diverse set of viral antigens if the standard direct (non-specific, low affinity) antigen absorption to the membrane was substituted by high-affinity capture. Specifically, we report an enhancement in assay sensitivity and a reduction in required protein concentrations through the capture of recombinant proteins via their encoded hexahistidine (6XHis) affinity tag. Affinity tag antigen coating enabled detection of SARS-CoV-2 Spike receptor binding domain (RBD)-reactive ASC, and also significantly improved assay performance using additional control antigens. Collectively, establishment of a universal antigen-coating approach streamlines characterization of the memory B-cell compartment after SARS-CoV-2 infection or COVID-19 vaccinations, and facilitates high-throughput immune-monitoring efforts of large donor cohorts in general.
Collapse
Affiliation(s)
- Sebastian Köppert
- Research & Development Department, Cellular Technology Limited, Shaker Heights, OH 44122, USA; (S.K.); (C.W.); (N.B.); (F.F.); (P.V.L.)
- Institute of Anatomy and Cell Biology, Friedrich-Alexander University Erlangen-Nürnberg, 91054 Erlangen, Germany;
| | - Carla Wolf
- Research & Development Department, Cellular Technology Limited, Shaker Heights, OH 44122, USA; (S.K.); (C.W.); (N.B.); (F.F.); (P.V.L.)
- Institute of Anatomy and Cell Biology, Friedrich-Alexander University Erlangen-Nürnberg, 91054 Erlangen, Germany;
| | - Noémi Becza
- Research & Development Department, Cellular Technology Limited, Shaker Heights, OH 44122, USA; (S.K.); (C.W.); (N.B.); (F.F.); (P.V.L.)
| | - Giuseppe A. Sautto
- Center for Vaccines and Immunology, University of Georgia, Athens, GA 30602, USA; (G.A.S.); (T.M.R.)
| | - Fridolin Franke
- Research & Development Department, Cellular Technology Limited, Shaker Heights, OH 44122, USA; (S.K.); (C.W.); (N.B.); (F.F.); (P.V.L.)
| | - Stefanie Kuerten
- Institute of Anatomy and Cell Biology, Friedrich-Alexander University Erlangen-Nürnberg, 91054 Erlangen, Germany;
- Institute of Neuroanatomy, Medical Faculty, University of Bonn, 53115 Bonn, Germany
| | - Ted M. Ross
- Center for Vaccines and Immunology, University of Georgia, Athens, GA 30602, USA; (G.A.S.); (T.M.R.)
- Department of Infectious Diseases, University of Georgia, Athens, GA 30602, USA
| | - Paul V. Lehmann
- Research & Development Department, Cellular Technology Limited, Shaker Heights, OH 44122, USA; (S.K.); (C.W.); (N.B.); (F.F.); (P.V.L.)
| | - Greg A. Kirchenbaum
- Research & Development Department, Cellular Technology Limited, Shaker Heights, OH 44122, USA; (S.K.); (C.W.); (N.B.); (F.F.); (P.V.L.)
- Correspondence: ; Tel.: +1-(216)-791-5084
| |
Collapse
|
35
|
Soe PT, Hanthamrongwit J, Saelee C, Kyaw SP, Khaenam P, Warit S, Satproedprai N, Mahasirimongkol S, Yanai H, Chootong P, Leepiyasakulchai C. Circulating IgA/IgG memory B cells against Mycobacterium tuberculosis dormancy-associated antigens Rv2659c and Rv3128c in active and latent tuberculosis. Int J Infect Dis 2021; 110:75-82. [PMID: 34284090 DOI: 10.1016/j.ijid.2021.07.033] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2021] [Revised: 07/12/2021] [Accepted: 07/13/2021] [Indexed: 12/13/2022] Open
Abstract
OBJECTIVE To elucidate the antigenic potential of dormancy-associated antigens Rv2659c and Rv3128c of Mycobacterium tuberculosis by examining the persistence of specific IgG and IgA memory B cells (MBCs) among patients with active tuberculosis (TB), household contacts with latent tuberculosis (LTBI), and an endemic healthy control group. METHODS Fresh peripheral blood mononuclear cells from the three study groups were used to enumerate the numbers of IgG and IgA MBCs specific to recombinant protein Rv2659c and Rv3128c by ELISpot assay. The composition of MBC subsets IgA+ and IgG + was analyzed by flow cytometry. RESULTS The number of IgA MBCs specific to antigen Rv2659c was significantly higher in the LTBI group than the TB group. In contrast, no significant difference was found in IgA or IgG MBCs against antigen Rv3128c. The number of IgA+ MBCs was significantly higher than that of IgG+ MBCs in the classical MBC subset of the LTBI group. CONCLUSION The results indicated that the dormancy-associated antigen Rv2659c induced an IgA MBCs response in individuals with latent TB, and IgA+ classical MBCs formed a major portion of the MBCs subset. This new knowledge will be beneficial for the development of novel TB vaccines and their control of latent TB.
Collapse
Affiliation(s)
- Phyu Thwe Soe
- Department of Clinical Microbiology and Applied Technology, Faculty of Medical Technology, Mahidol University, Bangkok, Thailand; Department of Medical Laboratory Technology, University of Medical Technology, Mandalay, Myanmar
| | - Jariya Hanthamrongwit
- Department of Clinical Microbiology and Applied Technology, Faculty of Medical Technology, Mahidol University, Bangkok, Thailand
| | - Chutiphon Saelee
- Department of Clinical Microbiology and Applied Technology, Faculty of Medical Technology, Mahidol University, Bangkok, Thailand
| | - Soe Paing Kyaw
- Clinical Pathology Laboratory, (1000) Bedded General Hospital, Nay Pyi Taw, Myanmar
| | - Prasong Khaenam
- Center of Standardization and Product Validation, Faculty of Medical Technology, Mahidol University, Bangkok, Thailand
| | - Saradee Warit
- Industrial Tuberculosis Team, IMMBRG, National Center for Genetic Engineering and Biotechnology (BIOTEC), NSTDA, Pathum Thani, Thailand
| | - Nusara Satproedprai
- Genomic Medicine and Innovation Support Division, Department of Medical Sciences, Ministry of Public Health, Thailand
| | - Surakameth Mahasirimongkol
- Genomic Medicine and Innovation Support Division, Department of Medical Sciences, Ministry of Public Health, Thailand
| | - Hideki Yanai
- Department of Clinical Laboratory, Fukujuji Hospital, Japan Anti-Tuberculosis Association (JATA), Tokyo, Japan
| | - Patchanee Chootong
- Department of Clinical Microbiology and Applied Technology, Faculty of Medical Technology, Mahidol University, Bangkok, Thailand
| | - Chaniya Leepiyasakulchai
- Department of Clinical Microbiology and Applied Technology, Faculty of Medical Technology, Mahidol University, Bangkok, Thailand.
| |
Collapse
|
36
|
Cherukuri A, Salama AD, Mehta R, Mohib K, Zheng L, Magee C, Harber M, Stauss H, Baker RJ, Tevar A, Landsittel D, Lakkis FG, Hariharan S, Rothstein DM. Transitional B cell cytokines predict renal allograft outcomes. Sci Transl Med 2021; 13:13/582/eabe4929. [PMID: 33627487 DOI: 10.1126/scitranslmed.abe4929] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2020] [Accepted: 01/25/2021] [Indexed: 12/12/2022]
Abstract
Early immunological biomarkers that predict rejection and chronic allograft loss are needed to inform preemptive therapy and improve long-term outcomes. Here, we prospectively examined the ratio of interleukin-10 (IL-10) to tumor necrosis factor-α (TNFα) produced by transitional-1 B cells (T1B) 3 months after transplantation as a predictive biomarker for clinical and subclinical renal allograft rejection and subsequent clinical course. In both Training (n = 162) and Internal Validation (n = 82) Sets, the T1B IL-10/TNFα ratio 3 months after transplantation predicted both clinical and subclinical rejection anytime in the first year. The biomarker also predicted subsequent late rejection with a lead time averaging 8 months. Among biomarker high-risk patients, 60% had early rejection, of which 48% recurred later in the first posttransplant year. Among high-risk patients without early rejection, 74% developed rejection later in the first year. In contrast, only 5% of low-risk patients had early and 5% late rejection. The biomarker also predicted rejection in an External Validation Set (n = 95) and in key patient subgroups, confirming generalizability. Biomarker high-risk patients exhibited progressively worse renal function and decreased 5-year graft survival compared to low-risk patients. Treatment of B cells with anti-TNFα in vitro augmented the IL-10/TNFα ratio, restored regulatory activity, and inhibited plasmablast differentiation. To conclude, the T1B IL-10/TNFα ratio was validated as a strong predictive biomarker of renal allograft outcomes and provides a rationale for preemptive therapeutic intervention with TNF blockade.
Collapse
Affiliation(s)
- Aravind Cherukuri
- Thomas E. Starzl Transplantation Institute, Department of Surgery, University of Pittsburgh, Pittsburgh, PA 15261, USA.,Renal and Electrolyte Division, Department of Medicine, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Alan D Salama
- University College of London Department of Renal Medicine, Royal Free Hospital, London NW3 2QG, UK
| | - Rajil Mehta
- Thomas E. Starzl Transplantation Institute, Department of Surgery, University of Pittsburgh, Pittsburgh, PA 15261, USA.,Renal and Electrolyte Division, Department of Medicine, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Kanishka Mohib
- Thomas E. Starzl Transplantation Institute, Department of Surgery, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Leting Zheng
- Thomas E. Starzl Transplantation Institute, Department of Surgery, University of Pittsburgh, Pittsburgh, PA 15261, USA.,Department of Rheumatology and Immunology, First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi 530021, China
| | - Ciara Magee
- University College of London Department of Renal Medicine, Royal Free Hospital, London NW3 2QG, UK
| | - Mark Harber
- University College of London Department of Renal Medicine, Royal Free Hospital, London NW3 2QG, UK
| | - Hans Stauss
- UCL Institute of Immunity and Transplantation, Royal Free Hospital, London NW3 2QG, UK
| | - Richard J Baker
- Renal Unit, St. James's University Hospital, Leeds LS9 7TF, UK
| | - Amit Tevar
- Thomas E. Starzl Transplantation Institute, Department of Surgery, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Douglas Landsittel
- Thomas E. Starzl Transplantation Institute, Department of Surgery, University of Pittsburgh, Pittsburgh, PA 15261, USA.,Department of Biomedical Informatics, University of Pittsburgh, Pittsburgh, PA 15206, USA
| | - Fadi G Lakkis
- Thomas E. Starzl Transplantation Institute, Department of Surgery, University of Pittsburgh, Pittsburgh, PA 15261, USA.,Renal and Electrolyte Division, Department of Medicine, University of Pittsburgh, Pittsburgh, PA 15261, USA.,Department of Immunology, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Sundaram Hariharan
- Thomas E. Starzl Transplantation Institute, Department of Surgery, University of Pittsburgh, Pittsburgh, PA 15261, USA.,Renal and Electrolyte Division, Department of Medicine, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - David M Rothstein
- Thomas E. Starzl Transplantation Institute, Department of Surgery, University of Pittsburgh, Pittsburgh, PA 15261, USA. .,Renal and Electrolyte Division, Department of Medicine, University of Pittsburgh, Pittsburgh, PA 15261, USA.,Department of Immunology, University of Pittsburgh, Pittsburgh, PA 15213, USA
| |
Collapse
|
37
|
Valgardsdottir R, Cattaneo I, Napolitano G, Raglio A, Spinelli O, Salmoiraghi S, Castilletti C, Lapa D, Capobianchi MR, Farina C, Golay J. Identification of Human SARS-CoV-2 Monoclonal Antibodies from Convalescent Patients Using EBV Immortalization. Antibodies (Basel) 2021; 10:26. [PMID: 34287229 PMCID: PMC8293222 DOI: 10.3390/antib10030026] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2021] [Revised: 06/02/2021] [Accepted: 06/30/2021] [Indexed: 12/27/2022] Open
Abstract
We report the isolation of two human IgG1k monoclonal antibodies (mAbs) directed against the SARS-CoV-2 spike protein. These mAbs were isolated from two donors who had recovered from COVID-19 infection during the first pandemic peak in the Lombardy region of Italy, the first European and initially most affected region in March 2020. We used the method of EBV immortalization of purified memory B cells and supernatant screening with a spike S1/2 assay for mAb isolation. This method allowed rapid isolation of clones, with one donor showing about 7% of clones positive against spike protein, whereas the other donor did not produce positive clones out of 91 tested. RNA was extracted from positive clones 39-47 days post-EBV infection, allowing VH and VL sequencing. The same clones were sequenced again after a further 100 days in culture, showing that no mutation had taken place during in vitro expansion. The B cell clones could be expanded in culture for more than 4 months after EBV immortalization and secreted the antibodies stably during that time, allowing to purify mg quantities of each mAb for functional assays without generating recombinant proteins. Unfortunately, neither mAb had significant neutralizing activity in a virus infection assay with several different SARS-CoV-2 isolates. The antibody sequences are made freely available.
Collapse
Affiliation(s)
- Rut Valgardsdottir
- Center of Cellular Therapy “G. Lanzani”, Division of Hematology, ASST Papa Giovanni XXIII, 24127 Bergamo, Italy; (R.V.); (I.C.); (O.S.); (S.S.)
| | - Irene Cattaneo
- Center of Cellular Therapy “G. Lanzani”, Division of Hematology, ASST Papa Giovanni XXIII, 24127 Bergamo, Italy; (R.V.); (I.C.); (O.S.); (S.S.)
| | - Gavino Napolitano
- Division of Microbiology and Virology, ASST Papa Giovanni XXIII, 24127 Bergamo, Italy; (G.N.); (A.R.); (C.F.)
| | - Annibale Raglio
- Division of Microbiology and Virology, ASST Papa Giovanni XXIII, 24127 Bergamo, Italy; (G.N.); (A.R.); (C.F.)
| | - Orietta Spinelli
- Center of Cellular Therapy “G. Lanzani”, Division of Hematology, ASST Papa Giovanni XXIII, 24127 Bergamo, Italy; (R.V.); (I.C.); (O.S.); (S.S.)
| | - Silvia Salmoiraghi
- Center of Cellular Therapy “G. Lanzani”, Division of Hematology, ASST Papa Giovanni XXIII, 24127 Bergamo, Italy; (R.V.); (I.C.); (O.S.); (S.S.)
- Fondazione per la Ricerca Ospedale Maggiore, 24127 Bergamo, Italy
| | - Concetta Castilletti
- Virology Laboratory, INMI-IRCCS “L. Spallanzani”, 00149 Roma, Italy; (C.C.); (D.L.); (M.R.C.)
| | - Daniele Lapa
- Virology Laboratory, INMI-IRCCS “L. Spallanzani”, 00149 Roma, Italy; (C.C.); (D.L.); (M.R.C.)
| | | | - Claudio Farina
- Division of Microbiology and Virology, ASST Papa Giovanni XXIII, 24127 Bergamo, Italy; (G.N.); (A.R.); (C.F.)
| | - Josee Golay
- Center of Cellular Therapy “G. Lanzani”, Division of Hematology, ASST Papa Giovanni XXIII, 24127 Bergamo, Italy; (R.V.); (I.C.); (O.S.); (S.S.)
- Fondazione per la Ricerca Ospedale Maggiore, 24127 Bergamo, Italy
| |
Collapse
|
38
|
Matsuda Y, Watanabe T, Li XK. Approaches for Controlling Antibody-Mediated Allograft Rejection Through Targeting B Cells. Front Immunol 2021; 12:682334. [PMID: 34276669 PMCID: PMC8282180 DOI: 10.3389/fimmu.2021.682334] [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: 03/18/2021] [Accepted: 06/17/2021] [Indexed: 01/14/2023] Open
Abstract
Both acute and chronic antibody-mediated allograft rejection (AMR), which are directly mediated by B cells, remain difficult to treat. Long-lived plasma cells (LLPCs) in bone marrow (BM) play a crucial role in the production of the antibodies that induce AMR. However, LLPCs survive through a T cell-independent mechanism and resist conventional immunosuppressive therapy. Desensitization therapy is therefore performed, although it is accompanied by severe side effects and the pathological condition may be at an irreversible stage when these antibodies, which induce AMR development, are detected in the serum. In other words, AMR control requires the development of a diagnostic method that predicts its onset before LLPC differentiation and enables therapeutic intervention and the establishment of humoral immune monitoring methods providing more detailed information, including individual differences in the susceptibility to immunosuppressive agents and the pathological conditions. In this study, we reviewed recent studies related to the direct or indirect involvement of immunocompetent cells in the differentiation of naïve-B cells into LLPCs, the limitations of conventional methods, and the possible development of novel control methods in the context of AMR. This information will significantly contribute to the development of clinical applications for AMR and improve the prognosis of patients who undergo organ transplantation.
Collapse
Affiliation(s)
- Yoshiko Matsuda
- Division of Transplantation Immunology, National Research Institute for Child Health and Development, Tokyo, Japan
| | - Takeshi Watanabe
- Laboratory of Immunology, Institute for Frontier Life and Medical Sciences, Kyoto University, Kyoto, Japan
| | - Xiao-Kang Li
- Division of Transplantation Immunology, National Research Institute for Child Health and Development, Tokyo, Japan
| |
Collapse
|
39
|
Woopen C, Schleußner K, Akgün K, Ziemssen T. Approach to SARS-CoV-2 Vaccination in Patients With Multiple Sclerosis. Front Immunol 2021; 12:701752. [PMID: 34234787 PMCID: PMC8256163 DOI: 10.3389/fimmu.2021.701752] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Accepted: 06/07/2021] [Indexed: 12/18/2022] Open
Abstract
For more than a year now, severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has been causing the coronavirus disease (COVID-19) pandemic with high mortality and detrimental effects on society, economy, and individual lives. Great hopes are being placed on vaccination as one of the most potent escape strategies from the pandemic and multiple vaccines are already in clinical use. However, there is still a lot of insecurity about the safety and efficacy of vaccines in patients with autoimmune diseases like multiple sclerosis (MS), especially under treatment with immunomodulatory or immunosuppressive drugs. We propose strategic approaches to SARS-CoV-2 vaccination management in MS patients and encourage fellow physicians to measure the immune response in their patients. Notably, both humoral and cellular responses should be considered since the immunological equivalent for protection from SARS-CoV-2 after infection or vaccination still remains undefined and will most likely involve antiviral cellular immunity. It is important to gain insights into the vaccine response of immunocompromised patients in order to be able to deduce sensible strategies for vaccination in the future.
Collapse
Affiliation(s)
| | | | | | - Tjalf Ziemssen
- Center of Clinical Neuroscience, Department of Neurology, University Hospital Carl Gustav Carus Dresden, Technical University of Dresden, Dresden, Germany
| |
Collapse
|
40
|
Lyski ZL, Brunton AE, Strnad MI, Sullivan PE, Siegel SA, Tafesse FG, Slifka MK, Messer WB. SARS-CoV-2 specific memory B-cells from individuals with diverse disease severities recognize SARS-CoV-2 variants of concern. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2021:2021.05.28.21258025. [PMID: 34100028 PMCID: PMC8183027 DOI: 10.1101/2021.05.28.21258025] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
In this investigation we examined the magnitude, breadth, and durability of SARS-CoV-2 specific antibodies in two distinct B-cell compartments: long-lived plasma cell-derived antibodies in the plasma, and peripheral memory B-cells along with their associated antibody profiles elicited after in vitro stimulation. We found that magnitude varied amongst individuals, but was the highest in hospitalized subjects. Variants of concern (VoC) -RBD-reactive antibodies were found in the plasma of 72% of samples in this investigation, and VoC-RBD-reactive memory B-cells were found in all but 1 subject at a single time-point. This finding, that VoC-RBD-reactive MBCs are present in the peripheral blood of all subjects including those that experienced asymptomatic or mild disease, provides a reason for optimism regarding the capacity of vaccination, prior infection, and/or both, to limit disease severity and transmission of variants of concern as they continue to arise and circulate.
Collapse
Affiliation(s)
- Zoe L. Lyski
- Department of Molecular Microbiology & Immunology, Oregon Health & Science University (OHSU), Portland, OR 97239, USA
| | | | - Matt I. Strnad
- OHSU-PSU School of Public Health, Portland, OR 97239, USA
| | | | | | - Fikadu G. Tafesse
- Department of Molecular Microbiology & Immunology, Oregon Health & Science University (OHSU), Portland, OR 97239, USA
| | - Mark K. Slifka
- Division of Neuroscience, Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, Oregon, USA
| | - William B. Messer
- Department of Molecular Microbiology & Immunology, Oregon Health & Science University (OHSU), Portland, OR 97239, USA
- OHSU-PSU School of Public Health, Portland, OR 97239, USA
- Department of Medicine, Division of Infectious Diseases, Oregon Health & Science University (OHSU), Portland, OR 97239, USA
| |
Collapse
|
41
|
Abstract
As one of the most important weapons against infectious diseases, vaccines have saved countless lives since their first use in the late eighteenth century. Antibodies produced by effector B cells upon vaccination play a critical role in mediating protection. The past several decades of research have led to a revolution in our understanding of B cell response to vaccination. Vaccines against SARS-CoV-2 coronavirus were developed at an unprecedented speed to power our global fight against COVID-19 pandemic. Nevertheless, we still face many challenges in the development of vaccines against many other deadly viruses, such as human immunodeficiency virus (HIV) and influenza virus. In this review, we summarize the latest findings on B cell response to vaccination and pathogen infection. We also discuss the current challenges in the field and the potential strategies targeting B cell response to improve vaccine efficacy.Key abbreviations box: BCR: B cell receptor; bNAb: broadly neutralizing antibody; DC: dendritic cells; DZ: dark zone; EF response: extrafollicular response; FDC: follicular dendritic cell; GC: germinal center; HIV: human immunodeficiency virus; IC: immune complex; LLPC: long-lived plasma cell; LZ: light zone; MBC: memory B cell; SLPB: short-lived plasmablast; TFH: T follicular helper cells; TLR: Toll-like receptor.
Collapse
Affiliation(s)
- Wei Luo
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Qian Yin
- Institute for Immunity, Transplantation & Infection, Stanford University School of Medicine, Stanford, California, USA
| |
Collapse
|
42
|
Immune memory in convalescent patients with asymptomatic or mild COVID-19. Cell Discov 2021; 7:18. [PMID: 33767156 PMCID: PMC7993859 DOI: 10.1038/s41421-021-00250-9] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2020] [Accepted: 02/09/2021] [Indexed: 12/29/2022] Open
Abstract
It is important to evaluate the durability of the protective immune response elicited by primary infection with severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). Here, we systematically evaluated the SARS-CoV-2-specific memory B cell and T cell responses in healthy controls and individuals recovered from asymptomatic or symptomatic infection approximately 6 months prior. Comparatively low frequencies of memory B cells specific for the receptor-binding domain (RBD) of spike glycoprotein (S) persisted in the peripheral blood of individuals who recovered from infection (median 0.62%, interquartile range 0.48-0.69). The SARS-CoV-2 RBD-specific memory B cell response was detected in 2 of 13 individuals who recovered from asymptomatic infection and 10 of 20 individuals who recovered from symptomatic infection. T cell responses induced by S, membrane (M), and nucleocapsid (N) peptide libraries from SARS-CoV-2 were observed in individuals recovered from coronavirus disease 2019 (COVID-19), and cross-reactive T cell responses to SARS-CoV-2 were also detected in healthy controls.
Collapse
|
43
|
Suda Y, Miyazaki A, Miyazawa K, Shibahara T, Ohashi S. Systemic and intestinal porcine epidemic diarrhea virus-specific antibody response and distribution of antibody-secreting cells in experimentally infected conventional pigs. Vet Res 2021; 52:2. [PMID: 33397461 PMCID: PMC7780908 DOI: 10.1186/s13567-020-00880-z] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2020] [Accepted: 12/15/2020] [Indexed: 01/03/2023] Open
Abstract
Porcine epidemic diarrhea (PED) is a coronavirus disease characterized by the rapid spread of severe diarrhea among pigs. PED virus (PEDV) infects and replicates mainly in the epithelial cells of the duodenum, jejunum, ileum and colon. Serum or mucosal IgA antibody levels have been used to predict both vaccine efficacy and the level of protective immunity to enteric infectious diseases in individuals or herds. Details of the B-cell immune response upon PEDV infection, such as the systemic and mucosal PEDV IgA antibody response, the distribution of IgA antibody-secreting cells (ASCs), and their role in virus clearance are not yet clear. In this experimental infection study, we observed similar fluctuations in PEDV IgA antibody levels in serum and intestinal contents of the upper and lower jejunum and ileum, but not fecal samples, over the 4-week experimental course. ASCs that actively secrete PEDV IgA antibody without in vitro stimulation were distributed mainly in the upper jejunum, whereas memory B cells that showed enhanced PEDV IgA antibody production upon in vitro stimulation were observed in mesenteric lymph nodes and the ileum. Our findings will contribute to the development of effective vaccines and diagnostic methods for PEDV.
Collapse
Affiliation(s)
- Yuto Suda
- Kyushu Research Station, National Institute of Animal Health (NIAH), National Agriculture and Food Research Organization (NARO), 2702 Chuzan, Kagoshima, Kagoshima, 891-0105, Japan. .,Division of Viral Disease and Epidemiology, NIAH, NARO, 3-1-5 Kannondai, Tsukuba, Ibaraki, 305-0856, Japan.
| | - Ayako Miyazaki
- Division of Viral Disease and Epidemiology, NIAH, NARO, 3-1-5 Kannondai, Tsukuba, Ibaraki, 305-0856, Japan
| | - Kohtaro Miyazawa
- Division of Viral Disease and Epidemiology, NIAH, NARO, 3-1-5 Kannondai, Tsukuba, Ibaraki, 305-0856, Japan
| | - Tomoyuki Shibahara
- Division of Pathology and Pathophysiology, NIAH, NARO, 3-1-5 Kannondai, Tsukuba, Ibaraki, 305-0856, Japan.,Department of Veterinary Science, Graduate School of Life and Environmental Sciences, Osaka Prefecture University, 1-58 Rinku-oraikita, Izumisano, Osaka, 598-8531, Japan
| | - Seiichi Ohashi
- Division of Viral Disease and Epidemiology, NIAH, NARO, 3-1-5 Kannondai, Tsukuba, Ibaraki, 305-0856, Japan
| |
Collapse
|
44
|
van Dam LS, Oskam JM, Kamerling SWA, Arends EJ, Bredewold OW, Berkowska MA, van Dongen JJM, Rabelink TJ, van Kooten C, Teng YKO. Highly Sensitive Flow Cytometric Detection of Residual B-Cells After Rituximab in Anti-Neutrophil Cytoplasmic Antibodies-Associated Vasculitis Patients. Front Immunol 2020; 11:566732. [PMID: 33384685 PMCID: PMC7770159 DOI: 10.3389/fimmu.2020.566732] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2020] [Accepted: 11/03/2020] [Indexed: 12/20/2022] Open
Abstract
Background B-cell depletion with rituximab (RTX) is an effective treatment for anti-neutrophil cytoplasmic antibodies (ANCA)-associated vasculitis (AAV) patients. Nevertheless, relapses are frequent after RTX, often preceded by B-cell repopulation suggesting that residual autoreactive B-cells persist despite therapy. Therefore, this study aimed to identify minimal residual autoimmunity (MRA) in the B-cell compartment of AAV patients treated with RTX. Methods EuroFlow-based highly-sensitive flow cytometry (HSFC) was employed to study B-cell and plasma cell (PC) subsets in-depth in AAV patients before and after RTX treatment. Additionally, peripheral blood mononuclear cells (PBMCs) of these RTX-treated AAV patients were cultured and in vitro stimulated with CpG, IL-2, and IL-21 to induce antibody-secreting cells (ASC). (ANCA)-IgG was measured in these supernatants by ELISA. Results By employing EuroFlow-based HSFC, we detected circulating CD19+ B-cells at all timepoints after RTX treatment, in contrast to conventional low-sensitive flow cytometry. Pre-germinal center (Pre-GC) B-cells, memory B-cells and CD20+CD138− plasmablasts (PBs) were rapidly and strongly reduced, while CD20−CD138− PrePC and CD20-CD138+ mature (m)PCs were reduced slower and remained detectable. Both memory B-cells and CD20− PCs remained detectable after RTX. Serum ANCA-IgG decreased significantly upon RTX. Changes in ANCA levels strongly correlated with changes in naive, switched CD27+ and CD27− (double-negative) memory B-cells, but not with plasma cells. Lastly, we demonstrated in vitro ANCA production by AAV PBMCs, 24 and 48 weeks after RTX treatment reflecting MRA in the memory compartment of AAV patients. Conclusion We demonstrated that RTX induced strong reductions in circulating B-cells, but never resulted in complete B-cell depletion. Despite strongly reduced B-cell numbers after RTX, ANCA-specific memory B-cells were still detectable in AAV patients. Thus, MRA is identifiable in AAV and can provide a potential novel approach in personalizing RTX treatment in AAV patients.
Collapse
Affiliation(s)
- Laura S van Dam
- Centre of Expertise for Lupus-, Vasculitis-, and Complement-Mediated Systemic Autoimmune Diseases (LuVaCs), Department of Internal Medicine, section Nephrology, Leiden University Medical Center, Leiden, Netherlands
| | - Jelle M Oskam
- Centre of Expertise for Lupus-, Vasculitis-, and Complement-Mediated Systemic Autoimmune Diseases (LuVaCs), Department of Internal Medicine, section Nephrology, Leiden University Medical Center, Leiden, Netherlands
| | - Sylvia W A Kamerling
- Centre of Expertise for Lupus-, Vasculitis-, and Complement-Mediated Systemic Autoimmune Diseases (LuVaCs), Department of Internal Medicine, section Nephrology, Leiden University Medical Center, Leiden, Netherlands
| | - Eline J Arends
- Centre of Expertise for Lupus-, Vasculitis-, and Complement-Mediated Systemic Autoimmune Diseases (LuVaCs), Department of Internal Medicine, section Nephrology, Leiden University Medical Center, Leiden, Netherlands
| | - O W Bredewold
- Centre of Expertise for Lupus-, Vasculitis-, and Complement-Mediated Systemic Autoimmune Diseases (LuVaCs), Department of Internal Medicine, section Nephrology, Leiden University Medical Center, Leiden, Netherlands
| | - Magdalena A Berkowska
- Immunomonitoring group, Department of Immunohematology and Bloodtransfusion, Leiden University Medical Center, Leiden, Netherlands
| | - Jacques J M van Dongen
- Immunomonitoring group, Department of Immunohematology and Bloodtransfusion, Leiden University Medical Center, Leiden, Netherlands
| | - Ton J Rabelink
- Centre of Expertise for Lupus-, Vasculitis-, and Complement-Mediated Systemic Autoimmune Diseases (LuVaCs), Department of Internal Medicine, section Nephrology, Leiden University Medical Center, Leiden, Netherlands
| | - Cees van Kooten
- Centre of Expertise for Lupus-, Vasculitis-, and Complement-Mediated Systemic Autoimmune Diseases (LuVaCs), Department of Internal Medicine, section Nephrology, Leiden University Medical Center, Leiden, Netherlands
| | - Y K Onno Teng
- Centre of Expertise for Lupus-, Vasculitis-, and Complement-Mediated Systemic Autoimmune Diseases (LuVaCs), Department of Internal Medicine, section Nephrology, Leiden University Medical Center, Leiden, Netherlands
| |
Collapse
|
45
|
Nguyen-Contant P, Embong AK, Topham DJ, Sangster MY. Analysis of Antigen-Specific Human Memory B Cell Populations Based on In Vitro Polyclonal Stimulation. CURRENT PROTOCOLS IN IMMUNOLOGY 2020; 131:e109. [PMID: 33017091 PMCID: PMC7647051 DOI: 10.1002/cpim.109] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Antigen-specific memory B cell (MBC) populations mediate the rapid, strong, and high-affinity secondary antibody responses that play a key role in combating infection and generating protective responses to vaccination. Recently, cell staining with fluorochrome-labeled antigens together with sequencing methods such as Drop-seq and CITE-seq have provided information on the specificity, phenotype, and transcriptome of single MBCs. However, characterization of MBCs at the level of antigen-reactive populations remains an important tool for assessing an individual's B cell immunity and responses to antigen exposure. This is readily performed using a long-established method based on in vitro polyclonal stimulation of MBCs to induce division and differentiation into antibody-secreting cells (ASCs). Post-stimulation antigen-specific measurement of the MBC-derived ASCs (or the secreted antibodies) indicates the size of precursor MBC populations. Additional information about the character of antigen-reactive MBC populations is provided by analysis of MBC-derived antibodies of particular specificities for binding avidity and functionality. This article outlines a simple and reliable strategy for efficient in vitro MBC stimulation and use of the ELISpot assay as a post-stimulation readout to determine the size of antigen-specific MBC populations. Other applications of the in vitro stimulation technique for MBC analysis are discussed. The following protocols are included. © 2020 Wiley Periodicals LLC Basic Protocol 1: Polyclonal stimulation of memory B cells using unfractionated PBMCs Alternate Protocol: Stimulation of small PBMC numbers using 96-well plates with U-bottom wells Basic Protocol 2: ELISpot assay for enumeration of memory B cell-derived antibody-secreting cells.
Collapse
Affiliation(s)
- Phuong Nguyen-Contant
- David H. Smith Center for Vaccine Biology and Immunology, Department of Microbiology and Immunology, University of Rochester Medical Center, Rochester, NY 14642, USA
| | - A. Karim Embong
- David H. Smith Center for Vaccine Biology and Immunology, Department of Microbiology and Immunology, University of Rochester Medical Center, Rochester, NY 14642, USA
| | - David J. Topham
- David H. Smith Center for Vaccine Biology and Immunology, Department of Microbiology and Immunology, University of Rochester Medical Center, Rochester, NY 14642, USA
| | - Mark Y. Sangster
- David H. Smith Center for Vaccine Biology and Immunology, Department of Microbiology and Immunology, University of Rochester Medical Center, Rochester, NY 14642, USA
| |
Collapse
|
46
|
Gerhards R, Pfeffer LK, Lorenz J, Starost L, Nowack L, Thaler FS, Schlüter M, Rübsamen H, Macrini C, Winklmeier S, Mader S, Bronge M, Grönlund H, Feederle R, Hsia HE, Lichtenthaler SF, Merl-Pham J, Hauck SM, Kuhlmann T, Bauer IJ, Beltran E, Gerdes LA, Mezydlo A, Bar-Or A, Banwell B, Khademi M, Olsson T, Hohlfeld R, Lassmann H, Kümpfel T, Kawakami N, Meinl E. Oligodendrocyte myelin glycoprotein as a novel target for pathogenic autoimmunity in the CNS. Acta Neuropathol Commun 2020; 8:207. [PMID: 33256847 PMCID: PMC7706210 DOI: 10.1186/s40478-020-01086-2] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2020] [Accepted: 11/18/2020] [Indexed: 12/19/2022] Open
Abstract
Autoimmune disorders of the central nervous system (CNS) comprise a broad spectrum of clinical entities. The stratification of patients based on the recognized autoantigen is of great importance for therapy optimization and for concepts of pathogenicity, but for most of these patients, the actual target of their autoimmune response is unknown. Here we investigated oligodendrocyte myelin glycoprotein (OMGP) as autoimmune target, because OMGP is expressed specifically in the CNS and there on oligodendrocytes and neurons. Using a stringent cell-based assay, we detected autoantibodies to OMGP in serum of 8/352 patients with multiple sclerosis, 1/28 children with acute disseminated encephalomyelitis and unexpectedly, also in one patient with psychosis, but in none of 114 healthy controls. Since OMGP is GPI-anchored, we validated its recognition also in GPI-anchored form. The autoantibodies to OMGP were largely IgG1 with a contribution of IgG4, indicating cognate T cell help. We found high levels of soluble OMGP in human spinal fluid, presumably due to shedding of the GPI-linked OMGP. Analyzing the pathogenic relevance of autoimmunity to OMGP in an animal model, we found that OMGP-specific T cells induce a novel type of experimental autoimmune encephalomyelitis dominated by meningitis above the cortical convexities. This unusual localization may be directed by intrathecal uptake and presentation of OMGP by meningeal phagocytes. Together, OMGP-directed autoimmunity provides a new element of heterogeneity, helping to improve the stratification of patients for diagnostic and therapeutic purposes.
Collapse
|
47
|
Wong HY, Prasad A, Gan SU, Chua JJE, Schwarz H. Identification of CD137-Expressing B Cells in Multiple Sclerosis Which Secrete IL-6 Upon Engagement by CD137 Ligand. Front Immunol 2020; 11:571964. [PMID: 33240262 PMCID: PMC7677239 DOI: 10.3389/fimmu.2020.571964] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2020] [Accepted: 10/12/2020] [Indexed: 12/21/2022] Open
Abstract
The potent costimulatory effect of CD137 has been implicated in several murine autoimmune disease models. CD137 costimulates and polarizes antigen-specific T cells toward a potent Th1/Tc1 response, and is essential for the development of experimental autoimmune encephalomyelitis (EAE), a murine model of Multiple Sclerosis (MS). This study aimed to investigate a role of CD137 in MS. Immunohistochemical and immunofluorescence staining of MS brain tissues was used to identify expression of CD137. CD137+ cells were identified in MS brain samples, with active lesions having the highest frequency of CD137+ cells. CD137 expression was found on several leukocyte subsets, including T cells, B cells and endothelial cells. In particular, CD137+ B cells were found in meningeal infiltrates. In vitro experiments showed that CD137 engagement on activated B cells increased early TNF and persistent IL-6 secretion with increased cell proliferation. These CD137+ B cells could interact with CD137L-expressing cells, secrete pro-inflammatory cytokines and accumulate in the meningeal infiltrate. This study demonstrates CD137 expression by activated B cells, enhancement of the inflammatory activity of B cells upon CD137 engagement, and provides evidence for a pathogenic role of CD137+ B cells in MS.
Collapse
Affiliation(s)
- Hiu Yi Wong
- Department of Physiology, National University of Singapore, Singapore, Singapore.,Immunology Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Ankshita Prasad
- Institute for Health Innovation and Technology, National University of Singapore, Singapore, Singapore
| | - Shu Uin Gan
- Department of Surgery, National University of Singapore, Singapore, Singapore
| | - John Jia En Chua
- Department of Physiology, National University of Singapore, Singapore, Singapore.,Institute for Health Innovation and Technology, National University of Singapore, Singapore, Singapore.,LSI Neurobiology Programme, National University of Singapore, Singapore, Singapore
| | - Herbert Schwarz
- Department of Physiology, National University of Singapore, Singapore, Singapore.,Immunology Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| |
Collapse
|
48
|
Autoantibodies against central nervous system antigens in a subset of B cell-dominant multiple sclerosis patients. Proc Natl Acad Sci U S A 2020; 117:21512-21518. [PMID: 32817492 DOI: 10.1073/pnas.2011249117] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Multiple sclerosis (MS) is a chronic autoimmune disease of the central nervous system (CNS), with characteristic inflammatory lesions and demyelination. The clinical benefit of cell-depleting therapies targeting CD20 has emphasized the role of B cells and autoantibodies in MS pathogenesis. We previously introduced an enzyme-linked immunospot spot (ELISpot)-based assay to measure CNS antigen-specific B cells in the blood of MS patients and demonstrated its usefulness as a predictive biomarker for disease activity in measuring the successful outcome of disease-modifying therapies (DMTs). Here we used a planar protein array to investigate CNS-reactive antibodies in the serum of MS patients as well as in B cell culture supernatants after polyclonal stimulation. Anti-CNS antibody reactivity was evident in the sera of the MS cohort, and the antibodies bound a heterogeneous set of molecules, including myelin, axonal cytoskeleton, and ion channel antigens, in individual patients. Immunoglobulin reactivity in supernatants of stimulated B cells was directed against a broad range of CNS antigens. A group of MS patients with a highly active B cell component was identified by the ELISpot assay. Those antibody reactivities remained stable over time. These assays with protein arrays identify MS patients with a highly active B cell population with antibodies directed against a swathe of CNS proteins.
Collapse
|
49
|
Davis CW, Jackson KJL, McCausland MM, Darce J, Chang C, Linderman SL, Chennareddy C, Gerkin R, Brown SJ, Wrammert J, Mehta AK, Cheung WC, Boyd SD, Waller EK, Ahmed R. Influenza vaccine-induced human bone marrow plasma cells decline within a year after vaccination. Science 2020; 370:237-241. [PMID: 32792465 PMCID: PMC10155619 DOI: 10.1126/science.aaz8432] [Citation(s) in RCA: 73] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2019] [Accepted: 08/02/2020] [Indexed: 01/09/2023]
Abstract
A universal vaccine against influenza would ideally generate protective immune responses that are not only broadly reactive against multiple influenza strains but also long-lasting. Because long-term serum antibody levels are maintained by bone marrow plasma cells (BMPCs), we investigated the production and maintenance of these cells after influenza vaccination. We found increased numbers of influenza-specific BMPCs 4 weeks after immunization with the seasonal inactivated influenza vaccine, but numbers returned to near their prevaccination levels after 1 year. This decline was driven by the loss of BMPCs induced by the vaccine, whereas preexisting BMPCs were maintained. Our results suggest that most BMPCs generated by influenza vaccination in adults are short-lived. Designing strategies to enhance their persistence will be a key challenge for the next generation of influenza vaccines.
Collapse
Affiliation(s)
- Carl W Davis
- Emory Vaccine Center and Department of Microbiology and Immunology, Emory University, Atlanta, GA, USA.,Emory-UGA Center of Excellence of Influenza Research and Surveillance (CEIRS), Atlanta GA, USA
| | | | - Megan M McCausland
- Emory Vaccine Center and Department of Microbiology and Immunology, Emory University, Atlanta, GA, USA.,Emory-UGA Center of Excellence of Influenza Research and Surveillance (CEIRS), Atlanta GA, USA
| | - Jaime Darce
- Cell Signaling Technology, Inc., Danvers, MA, USA
| | - Cathy Chang
- Emory Vaccine Center and Department of Microbiology and Immunology, Emory University, Atlanta, GA, USA.,Emory-UGA Center of Excellence of Influenza Research and Surveillance (CEIRS), Atlanta GA, USA
| | - Susanne L Linderman
- Emory Vaccine Center and Department of Microbiology and Immunology, Emory University, Atlanta, GA, USA.,Emory-UGA Center of Excellence of Influenza Research and Surveillance (CEIRS), Atlanta GA, USA
| | - Chakravarthy Chennareddy
- Emory Vaccine Center and Department of Microbiology and Immunology, Emory University, Atlanta, GA, USA.,Emory-UGA Center of Excellence of Influenza Research and Surveillance (CEIRS), Atlanta GA, USA
| | - Rebecca Gerkin
- Emory-UGA Center of Excellence of Influenza Research and Surveillance (CEIRS), Atlanta GA, USA.,Department of Hematology and Oncology, Winship Cancer Institute, Emory University School of Medicine, Atlanta, GA, USA
| | - Shantoria J Brown
- Emory-UGA Center of Excellence of Influenza Research and Surveillance (CEIRS), Atlanta GA, USA.,Department of Hematology and Oncology, Winship Cancer Institute, Emory University School of Medicine, Atlanta, GA, USA
| | - Jens Wrammert
- Emory Vaccine Center and Department of Microbiology and Immunology, Emory University, Atlanta, GA, USA.,Department of Pediatrics, Division of Infectious Disease, Emory University School of Medicine, Atlanta, GA, USA
| | - Aneesh K Mehta
- Emory-UGA Center of Excellence of Influenza Research and Surveillance (CEIRS), Atlanta GA, USA.,Division of Infectious Diseases, School of Medicine, Emory University, Atlanta, GA, USA
| | | | - Scott D Boyd
- Department of Pathology, Stanford University, Stanford, CA, USA
| | - Edmund K Waller
- Emory-UGA Center of Excellence of Influenza Research and Surveillance (CEIRS), Atlanta GA, USA.,Department of Hematology and Oncology, Winship Cancer Institute, Emory University School of Medicine, Atlanta, GA, USA
| | - Rafi Ahmed
- Emory Vaccine Center and Department of Microbiology and Immunology, Emory University, Atlanta, GA, USA. .,Emory-UGA Center of Excellence of Influenza Research and Surveillance (CEIRS), Atlanta GA, USA
| |
Collapse
|
50
|
Association of Antibody-Secreting Cells With Allograft Rejection After Renal Transplantation. Transplant Proc 2020; 52:1729-1733. [DOI: 10.1016/j.transproceed.2019.12.059] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2019] [Accepted: 12/06/2019] [Indexed: 12/20/2022]
|