1
|
Masi F, Al Qudsi S, Visigalli D, Zardini E, Capello E, Dicembre LP, Colombo E, Uccelli A, Gastaldi M, Inglese M, Franciotta D. Oligoclonal IgM band patterns in multiple sclerosis: A two-center study. J Neuroimmunol 2025; 404:578622. [PMID: 40288070 DOI: 10.1016/j.jneuroim.2025.578622] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2025] [Revised: 04/10/2025] [Accepted: 04/20/2025] [Indexed: 04/29/2025]
Abstract
BACKGROUND Cerebrospinal fluid (CSF) oligoclonal IgM bands (OCMBs) have been suggested as prognostic biomarkers in MS, but serum OCMBs meaning is still uncertain. OBJECTIVES We aimed to assess frequency and clinical relevance of all OCMB patterns. METHODS In this retrospective cohort study, 136 paired sera-CSF from consecutive persons with MS (pwMS) were tested in 2 centers for OCMBs using isoelectric focusing-immunoblotting. Active disease was defined as clinical or radiological relapse occurring during two-year follow-up. Predictors of active disease were analyzed with logistic regressions and Kaplan-Meier survival curves. RESULTS OCMBs were found in 6.6 % of pwMS as unique-to-CSF (pattern #2), and in 20.6 % as identical in serum-CSF (pattern #4), without between-cohort difference. Active disease was more frequent in those with pattern #2 (88.9 %) and #4 (64.3 %) than in those OCMB-negative (33.3 %, p < 0.001). In multivariate analysis, pattern #2 (OR: 15.9; 95 % CI [1.8-136]), and pattern #4 (OR: 3.3 95 % CI [1.3-8.3]) were independent predictors of active disease. In survival analysis, pattern #2 (p < 0.001) and #4 (p = 0.017) predicted radiological relapses. CONCLUSIONS Our data confirm that CSF OCMB marks poor prognosis in MS. However, both OCMB pattern #4 and pattern #2, with different strength prediction, might be useful to stratify pwMS deserving more aggressive treatments, although the stratification could be achieved in the near future with more standardized and easily measurable biomarkers (e.g., serum neurofilaments).
Collapse
Affiliation(s)
- Francesco Masi
- Department of Brain and Behavioral Sciences, University of Pavia, Pavia, Italy.
| | - Sabrina Al Qudsi
- Department of Neuroscience, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health (DINOGMI), Italy
| | - Davide Visigalli
- Department of Neuroscience, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health (DINOGMI), Italy; IRCCS Ospedale Policlinico San Martino, Genova, Italy
| | - Elisabetta Zardini
- Department of Brain and Behavioral Sciences, University of Pavia, Pavia, Italy; Neuroimmunology Research Unit and Multiple Sclerosis Center, IRCCS "C. Mondino" National Neurological Institute, Pavia, Italy
| | - Elisabetta Capello
- Department of Neuroscience, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health (DINOGMI), Italy; IRCCS Ospedale Policlinico San Martino, Genova, Italy
| | - Luca Pio Dicembre
- Department of Clinical Pathology, Santa Chiara Hospital-APSS, Trento, Italy
| | - Elena Colombo
- Multiple Sclerosis Center, IRCCS "C. Mondino" National Neurological Institute, Pavia, Italy
| | - Antonio Uccelli
- Department of Neuroscience, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health (DINOGMI), Italy; IRCCS Ospedale Policlinico San Martino, Genova, Italy
| | - Matteo Gastaldi
- Department of Brain and Behavioral Sciences, University of Pavia, Pavia, Italy; Neuroimmunology Research Unit and Multiple Sclerosis Center, IRCCS "C. Mondino" National Neurological Institute, Pavia, Italy
| | - Matilde Inglese
- Department of Neuroscience, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health (DINOGMI), Italy; IRCCS Ospedale Policlinico San Martino, Genova, Italy
| | - Diego Franciotta
- Department of Neuroscience, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health (DINOGMI), Italy; Department of Clinical Pathology, Santa Chiara Hospital-APSS, Trento, Italy
| |
Collapse
|
2
|
Hu Y, Huang J, Wang S, Sun X, Wang X, Yu H. Deciphering Autoimmune Diseases: Unveiling the Diagnostic, Therapeutic, and Prognostic Potential of Immune Repertoire Sequencing. Inflammation 2025; 48:676-695. [PMID: 38914737 DOI: 10.1007/s10753-024-02079-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Revised: 05/31/2024] [Accepted: 06/08/2024] [Indexed: 06/26/2024]
Abstract
Autoimmune diseases (AIDs) are immune system disorders where the body exhibits an immune response to its own antigens, causing damage to its own tissues and organs. The pathogenesis of AIDs is incompletely understood. However, recent advances in immune repertoire sequencing (IR-seq) technology have opened-up a new avenue to study the IR. These studies have revealed the prevalence in IR alterations, potentially inducing AIDs by disrupting immune tolerance and thereby contributing to our comprehension of AIDs. IR-seq harbors significant potential for the clinical diagnosis, personalized treatment, and prognosis of AIDs. This article reviews the application and progress of IR-seq in diseases, such as multiple sclerosis, systemic lupus erythematosus, rheumatoid arthritis, and type 1 diabetes, to enhance our understanding of the pathogenesis of AIDs and offer valuable references for the diagnosis and treatment of AIDs.
Collapse
Affiliation(s)
- Yuelin Hu
- Department of Immunology, Special Key Laboratory of Ocular Diseases of Guizhou Province, Zunyi Medical University, Zunyi, Guizhou, P.R. China
| | - Jialing Huang
- Department of Immunology, Special Key Laboratory of Ocular Diseases of Guizhou Province, Zunyi Medical University, Zunyi, Guizhou, P.R. China
| | - Shuqing Wang
- Department of Immunology, Special Key Laboratory of Ocular Diseases of Guizhou Province, Zunyi Medical University, Zunyi, Guizhou, P.R. China
| | - Xin Sun
- School of Basic Medical Sciences, Special Key Laboratory of Gene Detection and Therapy of Guizhou Province, Zunyi Medical University, Zunyi, Guizhou, P.R. China
| | - Xin Wang
- School of Basic Medical Sciences, Special Key Laboratory of Gene Detection and Therapy of Guizhou Province, Zunyi Medical University, Zunyi, Guizhou, P.R. China
| | - Hongsong Yu
- Department of Immunology, Special Key Laboratory of Ocular Diseases of Guizhou Province, Zunyi Medical University, Zunyi, Guizhou, P.R. China.
| |
Collapse
|
3
|
Jelcic I, Naghavian R, Fanaswala I, Macnair W, Esposito C, Calini D, Han Y, Marti Z, Raposo C, Sarabia Del Castillo J, Oldrati P, Erny D, Kana V, Zheleznyakova G, Al Nimer F, Tackenberg B, Reichen I, Khademi M, Piehl F, Robinson MD, Jelcic I, Sospedra M, Pelkmans L, Malhotra D, Reynolds R, Jagodic M, Martin R. T-bet+ CXCR3+ B cells drive hyperreactive B-T cell interactions in multiple sclerosis. Cell Rep Med 2025; 6:102027. [PMID: 40107244 PMCID: PMC11970401 DOI: 10.1016/j.xcrm.2025.102027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2023] [Revised: 05/16/2024] [Accepted: 02/20/2025] [Indexed: 03/22/2025]
Abstract
Multiple sclerosis (MS) is an autoimmune disease of the central nervous system (CNS). Self-peptide-dependent autoproliferation (AP) of B and T cells is a key mechanism in MS. Here, we show that pro-inflammatory B-T cell-enriched cell clusters (BTECs) form during AP and mirror features of a germinal center reaction. T-bet+CXCR3+ B cells are the main cell subset amplifying and sustaining their counterpart Th1 cells via interferon (IFN)-γ and are present in highly inflamed meningeal tissue. The underlying B cell activation signature is reflected by epigenetic modifications and receptor-ligand interactions with self-reactive T cells. AP+ CXCR3+ B cells show marked clonal evolution from memory to somatically hypermutated plasmablasts and upregulation of IFN-γ-related genes. Our data underscore a key role of T-bet+CXCR3+ B cells in the pathogenesis of MS in both the peripheral immune system and the CNS compartment, and thus they appear to be involved in both early relapsing-remitting disease and the chronic stage.
Collapse
Affiliation(s)
- Ivan Jelcic
- Neuroimmunology and MS Research Section (NIMS), Neurology Clinic, University of Zurich, University Hospital Zurich, 8091 Zurich, Switzerland; Roche Pharma Research and Early Development (pRED), Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd, Basel, Switzerland.
| | - Reza Naghavian
- Neuroimmunology and MS Research Section (NIMS), Neurology Clinic, University of Zurich, University Hospital Zurich, 8091 Zurich, Switzerland
| | - Imran Fanaswala
- SIB Swiss Institute of Bioinformatics, University of Zurich, Zurich, Switzerland; Department of Molecular Life Sciences, University of Zurich, Zurich, Switzerland
| | - Will Macnair
- Roche Pharma Research and Early Development (pRED), Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd, Basel, Switzerland
| | - Cinzia Esposito
- Department of Molecular Life Sciences, University of Zurich, Zurich, Switzerland
| | - Daniela Calini
- Roche Pharma Research and Early Development (pRED), Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd, Basel, Switzerland
| | - Yanan Han
- Department of Clinical Neuroscience, Karolinska Institutet, Center for Molecular Medicine, Karolinska University Hospital, Stockholm, Sweden
| | - Zoe Marti
- Neuroimmunology and MS Research Section (NIMS), Neurology Clinic, University of Zurich, University Hospital Zurich, 8091 Zurich, Switzerland; Cellerys AG, Schlieren, Switzerland
| | - Catarina Raposo
- Roche Pharma Research and Early Development (pRED), Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd, Basel, Switzerland
| | | | - Pietro Oldrati
- Neuroimmunology and MS Research Section (NIMS), Neurology Clinic, University of Zurich, University Hospital Zurich, 8091 Zurich, Switzerland; Cellerys AG, Schlieren, Switzerland
| | - Daniel Erny
- Neuroimmunology and MS Research Section (NIMS), Neurology Clinic, University of Zurich, University Hospital Zurich, 8091 Zurich, Switzerland; Institute of Neuropathology, University of Freiburg, Freiburg, Germany
| | - Veronika Kana
- Neuroimmunology and MS Research Section (NIMS), Neurology Clinic, University of Zurich, University Hospital Zurich, 8091 Zurich, Switzerland
| | - Galina Zheleznyakova
- Department of Clinical Neuroscience, Karolinska Institutet, Center for Molecular Medicine, Karolinska University Hospital, Stockholm, Sweden
| | - Faiez Al Nimer
- Neuroimmunology and MS Research Section (NIMS), Neurology Clinic, University of Zurich, University Hospital Zurich, 8091 Zurich, Switzerland; Department of Clinical Neuroscience, Karolinska Institutet, Center for Molecular Medicine, Karolinska University Hospital, Stockholm, Sweden
| | - Björn Tackenberg
- Product Development Medical Affairs, Neuroscience and Rare Disease, F. Hoffmann-La Roche Ltd, Basel, Switzerland
| | - Ina Reichen
- Neuroimmunology and MS Research Section (NIMS), Neurology Clinic, University of Zurich, University Hospital Zurich, 8091 Zurich, Switzerland
| | - Mohsen Khademi
- Department of Clinical Neuroscience, Karolinska Institutet, Center for Molecular Medicine, Karolinska University Hospital, Stockholm, Sweden
| | - Fredrik Piehl
- Department of Clinical Neuroscience, Karolinska Institutet, Center for Molecular Medicine, Karolinska University Hospital, Stockholm, Sweden
| | - Mark D Robinson
- SIB Swiss Institute of Bioinformatics, University of Zurich, Zurich, Switzerland; Department of Molecular Life Sciences, University of Zurich, Zurich, Switzerland
| | - Ilijas Jelcic
- Neuroimmunology and MS Research Section (NIMS), Neurology Clinic, University of Zurich, University Hospital Zurich, 8091 Zurich, Switzerland
| | - Mireia Sospedra
- Neuroimmunology and MS Research Section (NIMS), Neurology Clinic, University of Zurich, University Hospital Zurich, 8091 Zurich, Switzerland; Cellerys AG, Schlieren, Switzerland
| | - Lucas Pelkmans
- Department of Molecular Life Sciences, University of Zurich, Zurich, Switzerland
| | - Dheeraj Malhotra
- Roche Pharma Research and Early Development (pRED), Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd, Basel, Switzerland
| | | | - Maja Jagodic
- Department of Clinical Neuroscience, Karolinska Institutet, Center for Molecular Medicine, Karolinska University Hospital, Stockholm, Sweden
| | - Roland Martin
- Neuroimmunology and MS Research Section (NIMS), Neurology Clinic, University of Zurich, University Hospital Zurich, 8091 Zurich, Switzerland; Institute of Experimental Immunology, University of Zurich, Zurich, Switzerland; Therapeutic Design Unit, Center for Molecular Medicine, Department of Clinical Neurosciences, Karolinska Institutet, Stockholm, Sweden; Cellerys AG, Schlieren, Switzerland.
| |
Collapse
|
4
|
Di Sabatino E, Ferraro D, Gaetani L, Emiliano E, Parnetti L, Di Filippo M. CSF biomarkers of B-cell activation in multiple sclerosis: a clinical perspective. J Neurol 2025; 272:211. [PMID: 39960641 PMCID: PMC11832686 DOI: 10.1007/s00415-025-12907-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2024] [Revised: 01/04/2025] [Accepted: 01/07/2025] [Indexed: 02/20/2025]
Abstract
The role of B cells in the pathophysiology of multiple sclerosis (MS) extends beyond antibody synthesis, also involving the modulation of T lymphocytes and myeloid cells. B-cell activation within the Central Nervous System is associated with the release of various antibodies, cytokines, and chemokines, measurable in biofluids, thereby serving as biomarkers of the immune processes responsible for MS. To this purpose, a biomarker-based characterization of the disease through the combination of well-established markers, e.g., immunoglobulin (Ig) G index, IgG oligoclonal bands, Ig free light chains, with new promising markers, namely chemokine (C-X-C motif) ligand 13, and B-cell activating factor/A proliferation-inducing ligand, might represent a significant improvement in the management of people with MS.
Collapse
Affiliation(s)
- Elena Di Sabatino
- Clinica Neurologica, Dipartimento di Medicina e Chirurgia, Università di Perugia, Perugia, Umbria, Italy
| | - Diana Ferraro
- Dipartimento di Neuroscienze, Ospedale Civile di Baggiovara, Azienda Ospedaliera-Università di Modena, Modena, Italy
| | - Lorenzo Gaetani
- Clinica Neurologica, Dipartimento di Medicina e Chirurgia, Università di Perugia, Perugia, Umbria, Italy
| | - Edoardo Emiliano
- Clinica Neurologica, Dipartimento di Medicina e Chirurgia, Università di Perugia, Perugia, Umbria, Italy
| | - Lucilla Parnetti
- Clinica Neurologica, Dipartimento di Medicina e Chirurgia, Università di Perugia, Perugia, Umbria, Italy
| | - Massimiliano Di Filippo
- Clinica Neurologica, Dipartimento di Medicina e Chirurgia, Università di Perugia, Perugia, Umbria, Italy.
| |
Collapse
|
5
|
Pérez-Saldívar M, Nakamura Y, Kiyotani K, Imoto S, Katayama K, Yamaguchi R, Miyano S, Martínez-Barnetche J, Godoy-Lozano EE, Ordoñez G, Sotelo J, González-Conchillos H, Martínez-Palomo A, Flores-Rivera J, Santos-Argumedo L, Sánchez-Salguero ES, Espinosa-Cantellano M. Comparative analysis of the B cell receptor repertoire during relapse and remission in patients with multiple sclerosis. Clin Immunol 2024; 269:110398. [PMID: 39551364 DOI: 10.1016/j.clim.2024.110398] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2024] [Revised: 11/11/2024] [Accepted: 11/12/2024] [Indexed: 11/19/2024]
Abstract
Multiple sclerosis (MS) is a chronic, multifactorial, inflammatory and demyelinating disease of the central nervous system (CNS), which involves an autoimmune response against components of the myelin sheaths. Anti-B cell therapies have been proven to be successful in reducing relapses. Therefore, the study of B cells in both phases of the disease (relapse and remission) is of great importance. Here, we analyzed peripheral blood-cell BCR repertoire from 11 MS patients during a relapse phase and during remission, 6 patients with other inflammatory neurological diseases (OIND) and 10 healthy subjects (HCs), using next generation sequencing. In addition, immunoglobulins G, M, A and D were quantified in the serum of patients and controls, using ELISA. BCR repertoire of relapsing MS patients showed lower diversity, as well as a higher rate of somatic hypermutation compared to the other study groups. Within this group, the highest percentage of shared clonotypes was observed. IGHV4-32 gene was identified as a potential differential biomarker between MS and OIND, as well as IGL3-21 gene as a potential MS biomarker. On the other hand, an elevation of IgG and IgD was found in the serum of MS patients during remission, and the serum IgG was also elevated in MS patients during relapse. In conclusion, these results show the important role of B cells in the pathogenesis of the MS relapses and a new panorama on the analysis of the peripheral blood BCR repertoire to obtain diagnostic tools for MS. Furthermore, this work highlights the need of studies in diverse populations, since results reported in Caucasian populations may not coincide with the immunological course of MS patients in other latitudes, due to differences in genetic background and environmental exposures.
Collapse
Affiliation(s)
- Miriam Pérez-Saldívar
- Department of Infectomics and Molecular Pathogenesis, Center for Research and Advanced Studies (Cinvestav), Mexico City 07360, Mexico
| | - Yusuke Nakamura
- Project for Immunogenomics, Cancer Precision Medicine Center, Japanese Foundation for Cancer Research, Tokyo, Japan.
| | - Kazuma Kiyotani
- Project for Immunogenomics, Cancer Precision Medicine Center, Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Seiya Imoto
- Human Genome Center, Institute of Medical Science, The University of Tokyo, Tokyo 108-8639, Japan
| | - Kotoe Katayama
- Human Genome Center, Institute of Medical Science, The University of Tokyo, Tokyo 108-8639, Japan
| | - Rui Yamaguchi
- Division of Cancer Systems Biology, Aichi Cancer Center Research Institute, Nagoya, Aichi 464-8681, Japan
| | - Satoru Miyano
- Human Genome Center, Institute of Medical Science, The University of Tokyo, Tokyo 108-8639, Japan
| | - Jesús Martínez-Barnetche
- Centro de Investigación Sobre Enfermedades Infecciosas-Instituto Nacional de Salud Pública, Cuernavaca, Morelos 62100, Mexico
| | | | - Graciela Ordoñez
- Department of Neuroimmunology, National Institute of Neurology and Neurosurgery "Manuel Velasco Suarez" (INNN), Mexico City 14269, Mexico
| | - Julio Sotelo
- Department of Neuroimmunology, National Institute of Neurology and Neurosurgery "Manuel Velasco Suarez" (INNN), Mexico City 14269, Mexico
| | - Hugo González-Conchillos
- Department of Infectomics and Molecular Pathogenesis, Center for Research and Advanced Studies (Cinvestav), Mexico City 07360, Mexico
| | - Adolfo Martínez-Palomo
- Department of Infectomics and Molecular Pathogenesis, Center for Research and Advanced Studies (Cinvestav), Mexico City 07360, Mexico
| | - José Flores-Rivera
- Clinical Laboratory of Neurodegenerative Diseases, National Institute of Neurology and Neurosurgery "Manuel Velasco Suarez" (INNN), Mexico City 14269, Mexico
| | - Leopoldo Santos-Argumedo
- Department of Molecular Biomedicine, Center for Research and Advanced Studies (Cinvestav), Mexico City 07360, Mexico
| | - Erick Saúl Sánchez-Salguero
- Department of Molecular Biomedicine, Center for Research and Advanced Studies (Cinvestav), Mexico City 07360, Mexico
| | - Martha Espinosa-Cantellano
- Department of Infectomics and Molecular Pathogenesis, Center for Research and Advanced Studies (Cinvestav), Mexico City 07360, Mexico.
| |
Collapse
|
6
|
Di Filippo M, Gaetani L, Centonze D, Hegen H, Kuhle J, Teunissen CE, Tintoré M, Villar LM, Willemse EA, Zetterberg H, Parnetti L. Fluid biomarkers in multiple sclerosis: from current to future applications. THE LANCET REGIONAL HEALTH. EUROPE 2024; 44:101009. [PMID: 39444698 PMCID: PMC11496979 DOI: 10.1016/j.lanepe.2024.101009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/15/2024] [Revised: 05/21/2024] [Accepted: 07/09/2024] [Indexed: 10/25/2024]
Abstract
Multiple sclerosis (MS) is an immune-mediated inflammatory and degenerative disorder of the central nervous system (CNS) with heterogeneous clinical manifestations. In the last decade, the landscape of cerebrospinal fluid (CSF) and blood biomarkers as potential key tools for MS diagnosis, prognosis and treatment monitoring has evolved considerably, alongside magnetic resonance imaging (MRI). CSF analysis has the potential not only to provide information on the underlying immunopathology of the disease and exclude differential diagnoses, but also to predict the risk of future relapses and disability accrual, guide therapeutic decisions and thus improve patient outcomes. This Series article overviews the biological framework and current applicability of fluid biomarkers for MS, exploring their potential role in the molecular characterisation of the disease. We discuss recent advances in the field of neurochemistry that enabled the detection of brain-derived proteins in blood, opening the door to much more efficient longitudinal disease monitoring. Furthermore, we identify the current challenges in the application of fluid biomarkers for MS in a real-world setting, while offering recommendations for harnessing their full potential as key paraclinical tools to improve patient management and personalise treatment.
Collapse
Affiliation(s)
- Massimiliano Di Filippo
- Section of Neurology, Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Lorenzo Gaetani
- Section of Neurology, Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Diego Centonze
- IRCCS Neuromed, Pozzilli, IS, Italy
- Department of Systems Medicine, Tor Vergata University, Rome, Italy
| | - Harald Hegen
- Department of Neurology, Medical University of Innsbruck, Innsbruck, Austria
| | - Jens Kuhle
- Department of Neurology, University Hospital and University of Basel, Basel, Switzerland
- Multiple Sclerosis Centre and Research Center for Clinical Neuroimmunology and Neuroscience (RC2NB), Departments of Biomedicine and Clinical Research, University Hospital and University of Basel, Basel, Switzerland
| | - Charlotte E. Teunissen
- Neurochemistry Laboratory, Department of Laboratory Medicine, Amsterdam Neuroscience, Amsterdam University Medical Center, Amsterdam, the Netherlands
| | - Mar Tintoré
- Multiple Sclerosis Centre of Catalonia (Cemcat), Department of Neurology, Hospital Universitari Vall D'Hebron, Universitat Autònoma de Barcelona, Universitat de Vic-Universitat Central de Catalunya (UVic-UCC), Barcelona, Spain
| | - Luisa M. Villar
- Departments of Immunology and Neurology, Multiple Sclerosis Unit, Hospital Ramon y Cajal, (IRYCIS), Madrid, Spain
| | - Eline A.J. Willemse
- Department of Neurology, University Hospital and University of Basel, Basel, Switzerland
- Multiple Sclerosis Centre and Research Center for Clinical Neuroimmunology and Neuroscience (RC2NB), Departments of Biomedicine and Clinical Research, University Hospital and University of Basel, Basel, Switzerland
- Neurochemistry Laboratory, Department of Laboratory Medicine, Amsterdam Neuroscience, Amsterdam University Medical Center, Amsterdam, the Netherlands
| | - Henrik Zetterberg
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience & Physiology, Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
- Department of Neurodegenerative Disease, Queen Square Institute of Neurology, University College London, London, UK
- UK Dementia Research Institute, University College London, London, UK
- Hong Kong Center for Neurodegenerative Diseases, Hong Kong, China
- UW Department of Medicine, School of Medicine and Public Health, Madison, WI, USA
| | - Lucilla Parnetti
- Section of Neurology, Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| |
Collapse
|
7
|
Callegari I, Oechtering J, Schneider M, Perriot S, Mathias A, Voortman MM, Cagol A, Lanner U, Diebold M, Holdermann S, Kreiner V, Becher B, Granziera C, Junker A, Du Pasquier R, Khalil M, Kuhle J, Kappos L, Sanderson NSR, Derfuss T. Cell-binding IgM in CSF is distinctive of multiple sclerosis and targets the iron transporter SCARA5. Brain 2024; 147:839-848. [PMID: 38123517 PMCID: PMC10907079 DOI: 10.1093/brain/awad424] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2023] [Revised: 10/27/2023] [Accepted: 11/05/2023] [Indexed: 12/23/2023] Open
Abstract
Intrathecal IgM production in multiple sclerosis is associated with a worse disease course. To investigate pathogenic relevance of autoreactive IgM in multiple sclerosis, CSF from two independent cohorts, including multiple sclerosis patients and controls, were screened for antibody binding to induced pluripotent stem cell-derived neurons and astrocytes, and a panel of CNS-related cell lines. IgM binding to a primitive neuro-ectodermal tumour cell line discriminated 10% of multiple sclerosis donors from controls. Transcriptomes of single IgM producing CSF B cells from patients with cell-binding IgM were sequenced and used to produce recombinant monoclonal antibodies for characterization and antigen identification. We produced five cell-binding recombinant IgM antibodies, of which one, cloned from an HLA-DR + plasma-like B cell, mediated antigen-dependent complement activation. Immunoprecipitation and mass spectrometry, and biochemical and transcriptome analysis of the target cells identified the iron transport scavenger protein SCARA5 as the antigen target of this antibody. Intrathecal injection of a SCARA5 antibody led to an increased T cell infiltration in an experimental autoimmune encephalomyelitis (EAE) model. CSF IgM might contribute to CNS inflammation in multiple sclerosis by binding to cell surface antigens like SCARA5 and activating complement, or by facilitating immune cell migration into the brain.
Collapse
Affiliation(s)
- Ilaria Callegari
- Department of Biomedicine, University of Basel and University Hospital Basel, Basel 4031, Switzerland
- Neurologic Clinic and Policlinic, MS Center and Research Center for Clinical Neuroimmunology and Neuroscience Basel (RC2NB), University Hospital Basel and University of Basel, Basel 4056, Switzerland
| | - Johanna Oechtering
- Neurologic Clinic and Policlinic, MS Center and Research Center for Clinical Neuroimmunology and Neuroscience Basel (RC2NB), University Hospital Basel and University of Basel, Basel 4056, Switzerland
| | - Mika Schneider
- Department of Biomedicine, University of Basel and University Hospital Basel, Basel 4031, Switzerland
| | - Sylvain Perriot
- Department of Clinical Neurosciences, Laboratory of Neuroimmunology, Center of Research in Neurosciences, Lausanne 1011, Switzerland
| | - Amandine Mathias
- Department of Clinical Neurosciences, Laboratory of Neuroimmunology, Center of Research in Neurosciences, Lausanne 1011, Switzerland
| | | | - Alessandro Cagol
- Neurologic Clinic and Policlinic, MS Center and Research Center for Clinical Neuroimmunology and Neuroscience Basel (RC2NB), University Hospital Basel and University of Basel, Basel 4056, Switzerland
- Translational Imaging in Neurology (ThINK) Basel, Department of Biomedical Engineering, Faculty of Medicine, University Hospital Basel, University of Basel, Basel 4123, Switzerland
| | - Ulrike Lanner
- Proteomics Core Facility, Biozentrum, University of Basel, Basel 4056, Switzerland
| | - Martin Diebold
- Neurologic Clinic and Policlinic, MS Center and Research Center for Clinical Neuroimmunology and Neuroscience Basel (RC2NB), University Hospital Basel and University of Basel, Basel 4056, Switzerland
- Institute of Neuropathology, Faculty of Medicine, University of Freiburg, Freiburg 79085, Germany
| | - Sebastian Holdermann
- Department of Biomedicine, University of Basel and University Hospital Basel, Basel 4031, Switzerland
| | - Victor Kreiner
- Institute of Experimental Immunology, University of Zurich, Zurich 8057, Switzerland
| | - Burkhard Becher
- Institute of Experimental Immunology, University of Zurich, Zurich 8057, Switzerland
| | - Cristina Granziera
- Neurologic Clinic and Policlinic, MS Center and Research Center for Clinical Neuroimmunology and Neuroscience Basel (RC2NB), University Hospital Basel and University of Basel, Basel 4056, Switzerland
- Translational Imaging in Neurology (ThINK) Basel, Department of Biomedical Engineering, Faculty of Medicine, University Hospital Basel, University of Basel, Basel 4123, Switzerland
| | - Andreas Junker
- Department of Neuropathology, University Hospital Essen, Essen 45147, Germany
| | - Renaud Du Pasquier
- Department of Clinical Neurosciences, Laboratory of Neuroimmunology, Center of Research in Neurosciences, Lausanne 1011, Switzerland
- Department of Clinical Neurosciences, Service of Neurology, Lausanne 1011, Switzerland
| | - Michael Khalil
- Department of Neurology, Medical University of Graz, Graz 8010, Austria
| | - Jens Kuhle
- Neurologic Clinic and Policlinic, MS Center and Research Center for Clinical Neuroimmunology and Neuroscience Basel (RC2NB), University Hospital Basel and University of Basel, Basel 4056, Switzerland
| | - Ludwig Kappos
- Neurologic Clinic and Policlinic, MS Center and Research Center for Clinical Neuroimmunology and Neuroscience Basel (RC2NB), University Hospital Basel and University of Basel, Basel 4056, Switzerland
| | - Nicholas S R Sanderson
- Department of Biomedicine, University of Basel and University Hospital Basel, Basel 4031, Switzerland
- Neurologic Clinic and Policlinic, MS Center and Research Center for Clinical Neuroimmunology and Neuroscience Basel (RC2NB), University Hospital Basel and University of Basel, Basel 4056, Switzerland
| | - Tobias Derfuss
- Department of Biomedicine, University of Basel and University Hospital Basel, Basel 4031, Switzerland
- Neurologic Clinic and Policlinic, MS Center and Research Center for Clinical Neuroimmunology and Neuroscience Basel (RC2NB), University Hospital Basel and University of Basel, Basel 4056, Switzerland
| |
Collapse
|
8
|
Wang Q, Feng D, Jia S, Lu Q, Zhao M. B-Cell Receptor Repertoire: Recent Advances in Autoimmune Diseases. Clin Rev Allergy Immunol 2024; 66:76-98. [PMID: 38459209 DOI: 10.1007/s12016-024-08984-6] [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] [Accepted: 02/27/2024] [Indexed: 03/10/2024]
Abstract
In the field of contemporary medicine, autoimmune diseases (AIDs) are a prevalent and debilitating group of illnesses. However, they present extensive and profound challenges in terms of etiology, pathogenesis, and treatment. A major reason for this is the elusive pathophysiological mechanisms driving disease onset. Increasing evidence suggests the indispensable role of B cells in the pathogenesis of autoimmune diseases. Interestingly, B-cell receptor (BCR) repertoires in autoimmune diseases display a distinct skewing that can provide insights into disease pathogenesis. Over the past few years, advances in high-throughput sequencing have provided powerful tools for analyzing B-cell repertoire to understand the mechanisms during the period of B-cell immune response. In this paper, we have provided an overview of the mechanisms and analytical methods for generating BCR repertoire diversity and summarize the latest research progress on BCR repertoire in autoimmune diseases, including systemic lupus erythematosus (SLE), rheumatoid arthritis (RA), primary Sjögren's syndrome (pSS), multiple sclerosis (MS), and type 1 diabetes (T1D). Overall, B-cell repertoire analysis is a potent tool to understand the involvement of B cells in autoimmune diseases, facilitating the creation of innovative therapeutic strategies targeting specific B-cell clones or subsets.
Collapse
Affiliation(s)
- Qian Wang
- Department of Dermatology, Hunan Key Laboratory of Medical Epigenomics, the Second Xiangya Hospital of Central South University, Changsha, Hunan, China
- Clinical Medical Research Center of Major Skin Diseases and Skin Health of Hunan Province, Changsha, China
| | - Delong Feng
- Department of Dermatology, Hunan Key Laboratory of Medical Epigenomics, the Second Xiangya Hospital of Central South University, Changsha, Hunan, China
- Clinical Medical Research Center of Major Skin Diseases and Skin Health of Hunan Province, Changsha, China
| | - Sujie Jia
- Department of Pharmacy, Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing, 210042, China
| | - Qianjin Lu
- Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing, 210042, China.
- Key Laboratory of Basic and Translational Research on Immune-Mediated Skin Diseases, Chinese Academy of Medical Sciences, Nanjing, China.
| | - Ming Zhao
- Department of Dermatology, Hunan Key Laboratory of Medical Epigenomics, the Second Xiangya Hospital of Central South University, Changsha, Hunan, China.
- Clinical Medical Research Center of Major Skin Diseases and Skin Health of Hunan Province, Changsha, China.
- Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing, 210042, China.
- Key Laboratory of Basic and Translational Research on Immune-Mediated Skin Diseases, Chinese Academy of Medical Sciences, Nanjing, China.
| |
Collapse
|
9
|
Satitsuksanoa P, Iwasaki S, Boersma J, Bel Imam M, Schneider SR, Chang I, van de Veen W, Akdis M. B cells: The many facets of B cells in allergic diseases. J Allergy Clin Immunol 2023; 152:567-581. [PMID: 37247640 DOI: 10.1016/j.jaci.2023.05.011] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Revised: 03/30/2023] [Accepted: 05/16/2023] [Indexed: 05/31/2023]
Abstract
B cells play a key role in our immune system through their ability to produce antibodies, suppress a proinflammatory state, and contribute to central immune tolerance. We aim to provide an in-depth knowledge of the molecular biology of B cells, including their origin, developmental process, types and subsets, and functions. In allergic diseases, B cells are well known to induce and maintain immune tolerance through the production of suppressor cytokines such as IL-10. Similarly, B cells protect against viral infections such as severe acute respiratory syndrome coronavirus 2 that caused the recent coronavirus disease 2019 pandemic. Considering the unique and multifaceted functions of B cells, we hereby provide a comprehensive overview of the current knowledge of B-cell biology and its clinical applications in allergic diseases, organ transplantation, and cancer.
Collapse
Affiliation(s)
- Pattraporn Satitsuksanoa
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zürich, Davos, Switzerland.
| | - Sayuri Iwasaki
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zürich, Davos, Switzerland; Wageningen University & Research, Wageningen, The Netherlands
| | - Jolien Boersma
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zürich, Davos, Switzerland; Wageningen University & Research, Wageningen, The Netherlands
| | - Manal Bel Imam
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zürich, Davos, Switzerland
| | - Stephan R Schneider
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zürich, Davos, Switzerland
| | - Iris Chang
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zürich, Davos, Switzerland; Sean N. Parker Centre for Allergy and Asthma Research, Department of Medicine, Stanford University, Palo Alto, Calif
| | - Willem van de Veen
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zürich, Davos, Switzerland
| | - Mübeccel Akdis
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zürich, Davos, Switzerland.
| |
Collapse
|
10
|
Lim YW, Ramirez NJ, Asensio MA, Chiang Y, Müller G, Mrovecova P, Mitsuiki N, Krausz M, Camacho-Ordonez N, Warnatz K, Adler AS, Grimbacher B. Sequencing the B Cell Receptor Repertoires of Antibody-Deficient Individuals With and Without Infection Susceptibility. J Clin Immunol 2023; 43:940-950. [PMID: 36826743 PMCID: PMC10276080 DOI: 10.1007/s10875-023-01448-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2022] [Accepted: 02/07/2023] [Indexed: 02/25/2023]
Abstract
PURPOSE Most individuals with antibody deficiency (hypogammaglobulinemia) need immunoglobulin replacement therapy (IgG-RT) from healthy plasma donors to stay clear of infections. However, a small subset of hypogammaglobulinemic patients do not require this substitution therapy. We set out to investigate this clinical conundrum by asking whether the peripheral B cell receptor repertoires differ between antibody-deficient patients who do and do not need IgG-RT. METHODS We sequenced and analyzed IgG and IgM heavy chain B cell receptor repertoires from peripheral blood mononuclear cells (PBMCs) isolated from patients with low serum IgG concentrations who did or did not require IgG-RT. RESULTS Compared to the patients who did not need IgG-RT, those who needed IgG-RT had higher numbers of IgG antibody clones, higher IgM diversity, and less oligoclonal IgG and IgM repertoires. The patient cohorts had different heavy chain variable gene usage, and the patients who needed IgG-RT had elevated frequencies of IgG clones with higher germline identity (i.e., fewer somatic hypermutations). CONCLUSION Antibody-deficient patients with infection susceptibility who needed IgG-RT had more diverse peripheral antibody repertoires that were less diverged from germline and thus may not be as optimal for targeting pathogens, possibly contributing to infection susceptibility.
Collapse
Affiliation(s)
| | - Neftali Jose Ramirez
- Institute for Immunodeficiency, Medical Center, Faculty of Medicine, Albert-Ludwigs University, Freiburg, Germany
- Center for Chronic Immunodeficiency (CCI), Medical Center, Faculty of Medicine, Albert-Ludwigs University, Freiburg, Germany
| | | | - Yao Chiang
- GigaGen, Inc. (A Grifols Company), San Carlos, CA, USA
| | - Gabriele Müller
- Institute for Immunodeficiency, Medical Center, Faculty of Medicine, Albert-Ludwigs University, Freiburg, Germany
- Center for Chronic Immunodeficiency (CCI), Medical Center, Faculty of Medicine, Albert-Ludwigs University, Freiburg, Germany
| | - Pavla Mrovecova
- Institute for Immunodeficiency, Medical Center, Faculty of Medicine, Albert-Ludwigs University, Freiburg, Germany
- Center for Chronic Immunodeficiency (CCI), Medical Center, Faculty of Medicine, Albert-Ludwigs University, Freiburg, Germany
| | - Noriko Mitsuiki
- Institute for Immunodeficiency, Medical Center, Faculty of Medicine, Albert-Ludwigs University, Freiburg, Germany
- Center for Chronic Immunodeficiency (CCI), Medical Center, Faculty of Medicine, Albert-Ludwigs University, Freiburg, Germany
- Department of Pediatrics and Developmental Biology, Graduate School of Medical Sciences, Tokyo Medical and Dental University, Tokyo, Japan
| | - Máté Krausz
- Institute for Immunodeficiency, Medical Center, Faculty of Medicine, Albert-Ludwigs University, Freiburg, Germany
- Center for Chronic Immunodeficiency (CCI), Medical Center, Faculty of Medicine, Albert-Ludwigs University, Freiburg, Germany
- Department of Rheumatology and Clinical Immunology, Medical Center, Faculty of Medicine, Albert-Ludwigs University, Freiburg, Germany
| | - Nadezhda Camacho-Ordonez
- Institute for Immunodeficiency, Medical Center, Faculty of Medicine, Albert-Ludwigs University, Freiburg, Germany
- Center for Chronic Immunodeficiency (CCI), Medical Center, Faculty of Medicine, Albert-Ludwigs University, Freiburg, Germany
- Department of Rheumatology and Clinical Immunology, Medical Center, Faculty of Medicine, Albert-Ludwigs University, Freiburg, Germany
- Faculty of Biology, Albert-Ludwigs University, Freiburg, Germany
| | - Klaus Warnatz
- Center for Chronic Immunodeficiency (CCI), Medical Center, Faculty of Medicine, Albert-Ludwigs University, Freiburg, Germany
- Department of Rheumatology and Clinical Immunology, Medical Center, Faculty of Medicine, Albert-Ludwigs University, Freiburg, Germany
| | - Adam S Adler
- GigaGen, Inc. (A Grifols Company), San Carlos, CA, USA.
| | - Bodo Grimbacher
- Institute for Immunodeficiency, Medical Center, Faculty of Medicine, Albert-Ludwigs University, Freiburg, Germany.
- Center for Chronic Immunodeficiency (CCI), Medical Center, Faculty of Medicine, Albert-Ludwigs University, Freiburg, Germany.
- Department of Rheumatology and Clinical Immunology, Medical Center, Faculty of Medicine, Albert-Ludwigs University, Freiburg, Germany.
- DZIF - German Center for Infection Research, Satellite Center Freiburg, Freiburg im Breisgau, Germany.
- CIBSS - Centre for Integrative Biological Signalling Studies, Albert-Ludwigs University, Freiburg, Germany.
- RESIST - Cluster of Excellence 2155 to Hanover Medical School, Satellite Center, Freiburg, Germany.
| |
Collapse
|
11
|
Cossu D, Tomizawa Y, Yokoyama K, Sakanishi T, Momotani E, Sechi LA, Hattori N. Mycobacterium avium subsp. paratuberculosis Antigens Elicit a Strong IgG4 Response in Patients with Multiple Sclerosis and Exacerbate Experimental Autoimmune Encephalomyelitis. Life (Basel) 2023; 13:1437. [PMID: 37511812 PMCID: PMC10381415 DOI: 10.3390/life13071437] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2023] [Revised: 06/20/2023] [Accepted: 06/23/2023] [Indexed: 07/30/2023] Open
Abstract
Neuroinflammation can be triggered by microbial products disrupting immune regulation. In this study, we investigated the levels of IgG1, IgG2, IgG3, and IgG4 subclasses against the heat shock protein (HSP)70533-545 peptide and lipopentapeptide (MAP_Lp5) derived from Mycobacterium avium subsp. paratuberculosis (MAP) in the blood samples of Japanese and Italian individuals with relapsing remitting multiple sclerosis (MS). Additionally, we examined the impact of this peptide on MOG-induced experimental autoimmune encephalomyelitis (EAE). A total of 130 Japanese and 130 Italian subjects were retrospectively analyzed using the indirect ELISA method. Furthermore, a group of C57BL/6J mice received immunization with the MAP_HSP70533-545 peptide two weeks prior to the active induction of MOG35-55 EAE. The results revealed a significantly robust antibody response against MAP_HSP70533-545 in serum of both Japanese and Italian MS patients compared to their respective control groups. Moreover, heightened levels of serum IgG4 antibodies specific to MAP antigens were correlated with the severity of the disease. Additionally, EAE mice that were immunized with MAP_HSP70533-545 peptide exhibited more severe disease symptoms and increased reactivity of MOG35-55-specific T-cell compared to untreated mice. These findings provide evidence suggesting a potential link between MAP and the development or exacerbation of MS, particularly in a subgroup of MS patients with elevated serum IgG4 levels.
Collapse
Affiliation(s)
- Davide Cossu
- Department of Neurology, Juntendo University, Tokyo 1138431, Japan
- Biomedical Research Core Facilities, Juntendo University, Tokyo 1138431, Japan
- Department of Biomedical Sciences, Sassari University, 07100 Sassari, Italy
| | - Yuji Tomizawa
- Department of Neurology, Juntendo University, Tokyo 1138431, Japan
| | - Kazumasa Yokoyama
- Department of Neurology, Juntendo University, Tokyo 1138431, Japan
- Tosei Center for Neurological Diseases, Shizuoka 4180026, Japan
| | - Tamami Sakanishi
- Division of Cell Biology, Juntendo University, Tokyo 1138431, Japan
| | - Eiichi Momotani
- Department of Neurology, Juntendo University, Tokyo 1138431, Japan
- Comparative Medical Research Institute, Tsukuba 3050856, Japan
| | - Leonardo A Sechi
- Department of Biomedical Sciences, Sassari University, 07100 Sassari, Italy
- SC Microbiology, AOU Sassari, 07100 Sassari, Italy
| | - Nobutaka Hattori
- Department of Neurology, Juntendo University, Tokyo 1138431, Japan
- Neurodegenerative Disorders Collaborative Laboratory, RIKEN Center for Brain Science, Saitama 3510918, Japan
| |
Collapse
|
12
|
Li Y, Noto D, Hoshino Y, Mizuno M, Yoshikawa S, Miyake S. Immunoglobulin directly enhances differentiation of oligodendrocyte-precursor cells and remyelination. Sci Rep 2023; 13:9394. [PMID: 37296298 PMCID: PMC10256778 DOI: 10.1038/s41598-023-36532-3] [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: 02/10/2023] [Accepted: 06/05/2023] [Indexed: 06/12/2023] Open
Abstract
Multiple sclerosis (MS) is an inflammatory demyelinating disease characterized by multiple lesions in the central nervous system. Although the role of B cells in MS pathogenesis has attracted much attention, but the detailed mechanisms remain unclear. To investigate the effects of B cells on demyelination, we analyzed a cuprizone-induced demyelination model, and found that demyelination was significantly exacerbated in B cell-deficient mice. We next investigated whether immunoglobulin affected the myelin formation process using organotypic brain slice cultures and revealed that remyelination was improved in immunoglobulin-treated groups compared with the control group. Analysis of oligodendrocyte-precursor cell (OPC) monocultures showed that immunoglobulins directly affected on OPCs and promoted their differentiation and myelination. Furthermore, OPCs expressed FcγRI and FcγRIII, two receptors that were revealed to mediate the effects of IgG. To the best of our knowledge, this is the first study to demonstrate that B cells act in an inhibitory manner against cuprizone-induced demyelination, while immunoglobulins enhance remyelination following demyelination. Analysis of the culture system revealed that immunoglobulins directly act on OPCs to promote their differentiation and myelination. Future studies to elucidate the effects of immunoglobulins on OPCs in vivo and the detailed mechanisms of these effects may lead to new treatments for demyelinating diseases.
Collapse
Affiliation(s)
- Yaguang Li
- Department of Immunology, Juntendo University School of Medicine, 2-1-1 Hongo, Bunkyo-ku, Tokyo, 113-8421, Japan
| | - Daisuke Noto
- Department of Immunology, Juntendo University School of Medicine, 2-1-1 Hongo, Bunkyo-ku, Tokyo, 113-8421, Japan
| | - Yasunobu Hoshino
- Department of Neurology, Juntendo University School of Medicine, Tokyo, Japan
| | - Miho Mizuno
- Department of Immunology, Juntendo University School of Medicine, 2-1-1 Hongo, Bunkyo-ku, Tokyo, 113-8421, Japan
| | - Soichiro Yoshikawa
- Department of Immunology, Juntendo University School of Medicine, 2-1-1 Hongo, Bunkyo-ku, Tokyo, 113-8421, Japan
| | - Sachiko Miyake
- Department of Immunology, Juntendo University School of Medicine, 2-1-1 Hongo, Bunkyo-ku, Tokyo, 113-8421, Japan.
| |
Collapse
|
13
|
Biljecki M, Eisenhut K, Beltrán E, Winklmeier S, Mader S, Thaller A, Eichhorn P, Steininger P, Flierl-Hecht A, Lewerenz J, Kümpfel T, Kerschensteiner M, Meinl E, Thaler FS. Antibodies Against Glutamic Acid Decarboxylase 65 Are Locally Produced in the CSF and Arise During Affinity Maturation. NEUROLOGY(R) NEUROIMMUNOLOGY & NEUROINFLAMMATION 2023; 10:10/3/e200090. [PMID: 36823135 PMCID: PMC9969496 DOI: 10.1212/nxi.0000000000200090] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Accepted: 12/12/2022] [Indexed: 02/25/2023]
Abstract
BACKGROUND AND OBJECTIVES Antibodies (Abs) against the cytoplasmic protein glutamic acid decarboxylase 65 (GAD65) are detected in patients with neurologic syndromes together referred to as GAD65-Ab spectrum disorders. The response of some of these patients to plasma exchange or immunoglobulins indicates that GAD65-Abs could contribute to disease pathogenesis at least at some stages of disease. However, the involvement of GAD65-reactive B cells in the CNS is incompletely understood. METHODS We studied 7 patients with high levels of GAD65-Abs and generated monoclonal Abs (mAbs) derived from single cells in the CSF. Sequence characteristics, reactivity to GAD65, and the role of somatic hypermutations of the mAbs were analyzed. RESULTS Twelve CSF-derived mAbs were generated originating from 3 patients with short disease duration, and 7/12 of these mAbs (58%) were GAD65 reactive in at least 1 detection assay. Four of 12 (33%) were definitely positive in all 3 detection assays. The intrathecal anti-GAD65 response was polyclonal. GAD65-Abs were mostly of the IgG1 subtype and had undergone affinity maturation. Reversion of 2 GAD65-reactive mAbs to their corresponding germline-encoded unmutated common ancestors abolished GAD65 reactivity. DISCUSSION GAD65-specific B cells are present in the CNS and represent a sizable fraction of CSF B cells early in the disease course. The anti-GAD65 response in the CSF is polyclonal and shows evidence of antigen-driven affinity maturation required for GAD65 recognition. Our data support the hypothesis that the accumulation of GAD65-specific B cells and plasma cells in the CSF is an important feature of early disease stages.
Collapse
Affiliation(s)
- Michelle Biljecki
- From the Institute of Clinical Neuroimmunology (M.B., K.E., E.B., S.W., S.M., A.T., A.F.-H., T.K., M.K., E.M., F.S.T.), University Hospital, Ludwig-Maximilians-Universität Munich; Biomedical Center (BMC) (M.B., K.E., E.B., S.W., S.M., A.T., A.F.-H., T.K., M.K., E.M., F.S.T.), Medical Faculty, Ludwig-Maximilians-Universität Munich, Martinsried; Graduate School of Systemic Neurosciences Ludwig-Maximilians-Universität Munich (M.B., K.E.); Munich Cluster for Systems Neurology (SyNergy) (E.B., M.K., F.S.T.); Innate Immunity Unit (A.T.), Institut Pasteur, Inserm U1223, Paris, France; Université de Paris (A.T.), Sorbonne Paris Cité, France; Institute of Laboratory Medicine (P.E.), University Hospital, LMU Munich; Institute of Clinical and Molecular Virology (P.S.), University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg; and Department of Neurology (J.L.), University Hospital Ulm, Germany
| | - Katharina Eisenhut
- From the Institute of Clinical Neuroimmunology (M.B., K.E., E.B., S.W., S.M., A.T., A.F.-H., T.K., M.K., E.M., F.S.T.), University Hospital, Ludwig-Maximilians-Universität Munich; Biomedical Center (BMC) (M.B., K.E., E.B., S.W., S.M., A.T., A.F.-H., T.K., M.K., E.M., F.S.T.), Medical Faculty, Ludwig-Maximilians-Universität Munich, Martinsried; Graduate School of Systemic Neurosciences Ludwig-Maximilians-Universität Munich (M.B., K.E.); Munich Cluster for Systems Neurology (SyNergy) (E.B., M.K., F.S.T.); Innate Immunity Unit (A.T.), Institut Pasteur, Inserm U1223, Paris, France; Université de Paris (A.T.), Sorbonne Paris Cité, France; Institute of Laboratory Medicine (P.E.), University Hospital, LMU Munich; Institute of Clinical and Molecular Virology (P.S.), University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg; and Department of Neurology (J.L.), University Hospital Ulm, Germany
| | - Eduardo Beltrán
- From the Institute of Clinical Neuroimmunology (M.B., K.E., E.B., S.W., S.M., A.T., A.F.-H., T.K., M.K., E.M., F.S.T.), University Hospital, Ludwig-Maximilians-Universität Munich; Biomedical Center (BMC) (M.B., K.E., E.B., S.W., S.M., A.T., A.F.-H., T.K., M.K., E.M., F.S.T.), Medical Faculty, Ludwig-Maximilians-Universität Munich, Martinsried; Graduate School of Systemic Neurosciences Ludwig-Maximilians-Universität Munich (M.B., K.E.); Munich Cluster for Systems Neurology (SyNergy) (E.B., M.K., F.S.T.); Innate Immunity Unit (A.T.), Institut Pasteur, Inserm U1223, Paris, France; Université de Paris (A.T.), Sorbonne Paris Cité, France; Institute of Laboratory Medicine (P.E.), University Hospital, LMU Munich; Institute of Clinical and Molecular Virology (P.S.), University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg; and Department of Neurology (J.L.), University Hospital Ulm, Germany
| | - Stephan Winklmeier
- From the Institute of Clinical Neuroimmunology (M.B., K.E., E.B., S.W., S.M., A.T., A.F.-H., T.K., M.K., E.M., F.S.T.), University Hospital, Ludwig-Maximilians-Universität Munich; Biomedical Center (BMC) (M.B., K.E., E.B., S.W., S.M., A.T., A.F.-H., T.K., M.K., E.M., F.S.T.), Medical Faculty, Ludwig-Maximilians-Universität Munich, Martinsried; Graduate School of Systemic Neurosciences Ludwig-Maximilians-Universität Munich (M.B., K.E.); Munich Cluster for Systems Neurology (SyNergy) (E.B., M.K., F.S.T.); Innate Immunity Unit (A.T.), Institut Pasteur, Inserm U1223, Paris, France; Université de Paris (A.T.), Sorbonne Paris Cité, France; Institute of Laboratory Medicine (P.E.), University Hospital, LMU Munich; Institute of Clinical and Molecular Virology (P.S.), University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg; and Department of Neurology (J.L.), University Hospital Ulm, Germany
| | - Simone Mader
- From the Institute of Clinical Neuroimmunology (M.B., K.E., E.B., S.W., S.M., A.T., A.F.-H., T.K., M.K., E.M., F.S.T.), University Hospital, Ludwig-Maximilians-Universität Munich; Biomedical Center (BMC) (M.B., K.E., E.B., S.W., S.M., A.T., A.F.-H., T.K., M.K., E.M., F.S.T.), Medical Faculty, Ludwig-Maximilians-Universität Munich, Martinsried; Graduate School of Systemic Neurosciences Ludwig-Maximilians-Universität Munich (M.B., K.E.); Munich Cluster for Systems Neurology (SyNergy) (E.B., M.K., F.S.T.); Innate Immunity Unit (A.T.), Institut Pasteur, Inserm U1223, Paris, France; Université de Paris (A.T.), Sorbonne Paris Cité, France; Institute of Laboratory Medicine (P.E.), University Hospital, LMU Munich; Institute of Clinical and Molecular Virology (P.S.), University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg; and Department of Neurology (J.L.), University Hospital Ulm, Germany
| | - Anna Thaller
- From the Institute of Clinical Neuroimmunology (M.B., K.E., E.B., S.W., S.M., A.T., A.F.-H., T.K., M.K., E.M., F.S.T.), University Hospital, Ludwig-Maximilians-Universität Munich; Biomedical Center (BMC) (M.B., K.E., E.B., S.W., S.M., A.T., A.F.-H., T.K., M.K., E.M., F.S.T.), Medical Faculty, Ludwig-Maximilians-Universität Munich, Martinsried; Graduate School of Systemic Neurosciences Ludwig-Maximilians-Universität Munich (M.B., K.E.); Munich Cluster for Systems Neurology (SyNergy) (E.B., M.K., F.S.T.); Innate Immunity Unit (A.T.), Institut Pasteur, Inserm U1223, Paris, France; Université de Paris (A.T.), Sorbonne Paris Cité, France; Institute of Laboratory Medicine (P.E.), University Hospital, LMU Munich; Institute of Clinical and Molecular Virology (P.S.), University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg; and Department of Neurology (J.L.), University Hospital Ulm, Germany
| | - Peter Eichhorn
- From the Institute of Clinical Neuroimmunology (M.B., K.E., E.B., S.W., S.M., A.T., A.F.-H., T.K., M.K., E.M., F.S.T.), University Hospital, Ludwig-Maximilians-Universität Munich; Biomedical Center (BMC) (M.B., K.E., E.B., S.W., S.M., A.T., A.F.-H., T.K., M.K., E.M., F.S.T.), Medical Faculty, Ludwig-Maximilians-Universität Munich, Martinsried; Graduate School of Systemic Neurosciences Ludwig-Maximilians-Universität Munich (M.B., K.E.); Munich Cluster for Systems Neurology (SyNergy) (E.B., M.K., F.S.T.); Innate Immunity Unit (A.T.), Institut Pasteur, Inserm U1223, Paris, France; Université de Paris (A.T.), Sorbonne Paris Cité, France; Institute of Laboratory Medicine (P.E.), University Hospital, LMU Munich; Institute of Clinical and Molecular Virology (P.S.), University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg; and Department of Neurology (J.L.), University Hospital Ulm, Germany
| | - Philipp Steininger
- From the Institute of Clinical Neuroimmunology (M.B., K.E., E.B., S.W., S.M., A.T., A.F.-H., T.K., M.K., E.M., F.S.T.), University Hospital, Ludwig-Maximilians-Universität Munich; Biomedical Center (BMC) (M.B., K.E., E.B., S.W., S.M., A.T., A.F.-H., T.K., M.K., E.M., F.S.T.), Medical Faculty, Ludwig-Maximilians-Universität Munich, Martinsried; Graduate School of Systemic Neurosciences Ludwig-Maximilians-Universität Munich (M.B., K.E.); Munich Cluster for Systems Neurology (SyNergy) (E.B., M.K., F.S.T.); Innate Immunity Unit (A.T.), Institut Pasteur, Inserm U1223, Paris, France; Université de Paris (A.T.), Sorbonne Paris Cité, France; Institute of Laboratory Medicine (P.E.), University Hospital, LMU Munich; Institute of Clinical and Molecular Virology (P.S.), University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg; and Department of Neurology (J.L.), University Hospital Ulm, Germany
| | - Andrea Flierl-Hecht
- From the Institute of Clinical Neuroimmunology (M.B., K.E., E.B., S.W., S.M., A.T., A.F.-H., T.K., M.K., E.M., F.S.T.), University Hospital, Ludwig-Maximilians-Universität Munich; Biomedical Center (BMC) (M.B., K.E., E.B., S.W., S.M., A.T., A.F.-H., T.K., M.K., E.M., F.S.T.), Medical Faculty, Ludwig-Maximilians-Universität Munich, Martinsried; Graduate School of Systemic Neurosciences Ludwig-Maximilians-Universität Munich (M.B., K.E.); Munich Cluster for Systems Neurology (SyNergy) (E.B., M.K., F.S.T.); Innate Immunity Unit (A.T.), Institut Pasteur, Inserm U1223, Paris, France; Université de Paris (A.T.), Sorbonne Paris Cité, France; Institute of Laboratory Medicine (P.E.), University Hospital, LMU Munich; Institute of Clinical and Molecular Virology (P.S.), University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg; and Department of Neurology (J.L.), University Hospital Ulm, Germany
| | - Jan Lewerenz
- From the Institute of Clinical Neuroimmunology (M.B., K.E., E.B., S.W., S.M., A.T., A.F.-H., T.K., M.K., E.M., F.S.T.), University Hospital, Ludwig-Maximilians-Universität Munich; Biomedical Center (BMC) (M.B., K.E., E.B., S.W., S.M., A.T., A.F.-H., T.K., M.K., E.M., F.S.T.), Medical Faculty, Ludwig-Maximilians-Universität Munich, Martinsried; Graduate School of Systemic Neurosciences Ludwig-Maximilians-Universität Munich (M.B., K.E.); Munich Cluster for Systems Neurology (SyNergy) (E.B., M.K., F.S.T.); Innate Immunity Unit (A.T.), Institut Pasteur, Inserm U1223, Paris, France; Université de Paris (A.T.), Sorbonne Paris Cité, France; Institute of Laboratory Medicine (P.E.), University Hospital, LMU Munich; Institute of Clinical and Molecular Virology (P.S.), University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg; and Department of Neurology (J.L.), University Hospital Ulm, Germany
| | - Tania Kümpfel
- From the Institute of Clinical Neuroimmunology (M.B., K.E., E.B., S.W., S.M., A.T., A.F.-H., T.K., M.K., E.M., F.S.T.), University Hospital, Ludwig-Maximilians-Universität Munich; Biomedical Center (BMC) (M.B., K.E., E.B., S.W., S.M., A.T., A.F.-H., T.K., M.K., E.M., F.S.T.), Medical Faculty, Ludwig-Maximilians-Universität Munich, Martinsried; Graduate School of Systemic Neurosciences Ludwig-Maximilians-Universität Munich (M.B., K.E.); Munich Cluster for Systems Neurology (SyNergy) (E.B., M.K., F.S.T.); Innate Immunity Unit (A.T.), Institut Pasteur, Inserm U1223, Paris, France; Université de Paris (A.T.), Sorbonne Paris Cité, France; Institute of Laboratory Medicine (P.E.), University Hospital, LMU Munich; Institute of Clinical and Molecular Virology (P.S.), University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg; and Department of Neurology (J.L.), University Hospital Ulm, Germany
| | - Martin Kerschensteiner
- From the Institute of Clinical Neuroimmunology (M.B., K.E., E.B., S.W., S.M., A.T., A.F.-H., T.K., M.K., E.M., F.S.T.), University Hospital, Ludwig-Maximilians-Universität Munich; Biomedical Center (BMC) (M.B., K.E., E.B., S.W., S.M., A.T., A.F.-H., T.K., M.K., E.M., F.S.T.), Medical Faculty, Ludwig-Maximilians-Universität Munich, Martinsried; Graduate School of Systemic Neurosciences Ludwig-Maximilians-Universität Munich (M.B., K.E.); Munich Cluster for Systems Neurology (SyNergy) (E.B., M.K., F.S.T.); Innate Immunity Unit (A.T.), Institut Pasteur, Inserm U1223, Paris, France; Université de Paris (A.T.), Sorbonne Paris Cité, France; Institute of Laboratory Medicine (P.E.), University Hospital, LMU Munich; Institute of Clinical and Molecular Virology (P.S.), University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg; and Department of Neurology (J.L.), University Hospital Ulm, Germany
| | - Edgar Meinl
- From the Institute of Clinical Neuroimmunology (M.B., K.E., E.B., S.W., S.M., A.T., A.F.-H., T.K., M.K., E.M., F.S.T.), University Hospital, Ludwig-Maximilians-Universität Munich; Biomedical Center (BMC) (M.B., K.E., E.B., S.W., S.M., A.T., A.F.-H., T.K., M.K., E.M., F.S.T.), Medical Faculty, Ludwig-Maximilians-Universität Munich, Martinsried; Graduate School of Systemic Neurosciences Ludwig-Maximilians-Universität Munich (M.B., K.E.); Munich Cluster for Systems Neurology (SyNergy) (E.B., M.K., F.S.T.); Innate Immunity Unit (A.T.), Institut Pasteur, Inserm U1223, Paris, France; Université de Paris (A.T.), Sorbonne Paris Cité, France; Institute of Laboratory Medicine (P.E.), University Hospital, LMU Munich; Institute of Clinical and Molecular Virology (P.S.), University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg; and Department of Neurology (J.L.), University Hospital Ulm, Germany
| | - Franziska S Thaler
- From the Institute of Clinical Neuroimmunology (M.B., K.E., E.B., S.W., S.M., A.T., A.F.-H., T.K., M.K., E.M., F.S.T.), University Hospital, Ludwig-Maximilians-Universität Munich; Biomedical Center (BMC) (M.B., K.E., E.B., S.W., S.M., A.T., A.F.-H., T.K., M.K., E.M., F.S.T.), Medical Faculty, Ludwig-Maximilians-Universität Munich, Martinsried; Graduate School of Systemic Neurosciences Ludwig-Maximilians-Universität Munich (M.B., K.E.); Munich Cluster for Systems Neurology (SyNergy) (E.B., M.K., F.S.T.); Innate Immunity Unit (A.T.), Institut Pasteur, Inserm U1223, Paris, France; Université de Paris (A.T.), Sorbonne Paris Cité, France; Institute of Laboratory Medicine (P.E.), University Hospital, LMU Munich; Institute of Clinical and Molecular Virology (P.S.), University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg; and Department of Neurology (J.L.), University Hospital Ulm, Germany.
| |
Collapse
|
14
|
The complex role of inflammation and gliotransmitters in Parkinson's disease. Neurobiol Dis 2023; 176:105940. [PMID: 36470499 PMCID: PMC10372760 DOI: 10.1016/j.nbd.2022.105940] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2022] [Revised: 11/28/2022] [Accepted: 12/01/2022] [Indexed: 12/09/2022] Open
Abstract
Our understanding of the role of innate and adaptive immune cell function in brain health and how it goes awry during aging and neurodegenerative diseases is still in its infancy. Inflammation and immunological dysfunction are common components of Parkinson's disease (PD), both in terms of motor and non-motor components of PD. In recent decades, the antiquated notion that the central nervous system (CNS) in disease states is an immune-privileged organ, has been debunked. The immune landscape in the CNS influences peripheral systems, and peripheral immunological changes can alter the CNS in health and disease. Identifying immune and inflammatory pathways that compromise neuronal health and survival is critical in designing innovative and effective strategies to limit their untoward effects on neuronal health.
Collapse
|
15
|
Hvaring C, Alawad N, Salvesen Ø, Hovdal H, White LR, Boullerne AI. Cut-off evaluation of intrathecal oligoclonal bands of IgM in relapsing-remitting multiple sclerosis; a retrospective study. Mult Scler Relat Disord 2022; 68:104188. [PMID: 36179461 DOI: 10.1016/j.msard.2022.104188] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2022] [Accepted: 09/18/2022] [Indexed: 12/15/2022]
Abstract
BACKGROUND Multiple sclerosis (MS) is the most common demyelinating disease and characterized by immunological changes. Oligoclonal bands of IgG in CSF not seen in corresponding serum have been used for many years as part of the diagnostic criteria. However, considerably less is known about the role of IgM, despite several studies showing marked changes to IgM metabolism in MS. Bands of oligoclonal IgM (o-IgM) are more difficult to determine than oligoclonal IgG, thus limiting their study, and there is no agreement as to whether o-IgM in CSF should be part of the clinical work-up of MS. Nevertheless, there is a possibility that such bands might provide a prognostic marker if a cut-off could be established. MATERIALS AND METHODS In this pilot study, paired samples of CSF and serum from 37 patients with relapsing-remitting MS (RRMS) and 57 controls with no subsequent signs of neurological disease were analysed for total IgM, and bands of o-IgM were visualised by isoelectric focusing and western blot. Patient records were used to compare mean changes in Expanded Disability Status Scale (EDSS) over a maximum of 17 years. RESULTS None of the controls displayed extra o-IgM in CSF compared to corresponding serum, whereas additional o-IgM band(s) were seen in CSF in most patient samples (70%). After five years of disease, there was a significant difference in the EDSS between patients with no extra o-IgM compared to patients with at least one extra o-IgM band. This difference increased over time. If a cut-off of two or more extra bands of o-IgM in CSF was applied, this difference was not found. CONCLUSION These exploratory data suggest that o-IgM support the prognostic potential for RRMS, and though tentative, the occurrence of any bands of o-IgM restricted to CSF seems to result in poorer prognosis. Despite the small size of the groups, the data infer that the absence of CSF-restricted o-IgM is good news for the patient. The results need to be reproduced in a more comprehensive study.
Collapse
Affiliation(s)
- Charlotte Hvaring
- Department of Neuromedicine and Movement Science, Norwegian University of Science and Technology, Trondheim N-7491, Norway.
| | - Noor Alawad
- Department of Anesthesiology, University of Illinois at Chicago, Chicago, IL, USA
| | - Øyvind Salvesen
- Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology, Trondheim N-7491, Norway
| | - Harald Hovdal
- Department of Neurology and Clinical Neurophysiology, University Hospital of Trondheim, Trondheim N-7006, Norway
| | - Linda R White
- Department of Neuromedicine and Movement Science, Norwegian University of Science and Technology, Trondheim N-7491, Norway
| | - Anne I Boullerne
- Department of Anesthesiology, University of Illinois at Chicago, Chicago, IL, USA
| |
Collapse
|
16
|
Alcalá Vicente C, Lacruz L, Gascón F, Carratalà S, Quintanilla-Bordás C, Sanz MT, Carcelén-Gadea M, Mallada J, Carreres J, Gabaldón Torres L, Dominguez JA, Cañizares E, Gil-Perotin S, Cubas L, Gasqué Rubio R, Castillo-Villalba J, Pérez-Miralles FC, Casanova B. Oligoclonal M bands and cervical spinal cord lesions predict early secondary progressive multiple sclerosis. Front Neurol 2022; 13:991596. [DOI: 10.3389/fneur.2022.991596] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2022] [Accepted: 09/26/2022] [Indexed: 11/13/2022] Open
Abstract
ObjectiveTo determine baseline cerebrospinal fluid and magnetic resonance imaging (MRI) variables at the onset of a clinically isolated syndrome (CIS) suggestive of multiple sclerosis (MS) that predict evolution to secondary progressive MS (SPMS).Methods276 CIS patients with a minimum follow-up of 10 years were studied. Baseline presence of oligoclonal IgG and IgM bands (OCGB and OCMB respectively); number of brain T2 lesions (B-T2L), brain gadolinium enhancement lesions (brain-GEL), cervical spinal cord T2 lesions (cSC-T2L); and fulfillment of 2017 McDonald criteria among other variables were collected.Results14 patients ended up with a non-MS condition. 138/276 CIS patients fulfilled 2017 McDonald criteria. Mean age was 32.4 years, 185 female. 227 received treatment, 95 as CIS. After a mean follow-up of 12 years, 36 patients developed SPMS. Conversion to SPMS was associated with OCGB (p = 0.02), OCMB (p = 0.0001); ≥ 9 B-T2L (p = 0.03), brain-GEL (p = 0.03), and cSC-T2L (p = 0.03). However, after adjusting for sex, age, BT2L, brain-GEL, SC-T2, and OCMB status, only OCMB (HR 4.4, 1.9–10.6) and cSC-T2L (HR 2.2, 1.0–6.2) suggested an independent association with risk of conversion to SPMS. Patients with both risk factors had a HR of 6.12 (2.8–12.9).DiscussionOCMB and SC-T2 lesions are potential independent predictors of conversion to SPMS.
Collapse
|
17
|
Rosenstein I, Rasch S, Axelsson M, Novakova L, Blennow K, Zetterberg H, Lycke J. Increased intrathecal neurofilament light and immunoglobulin M predict severe disability in relapsing-remitting multiple sclerosis. Front Immunol 2022; 13:967953. [PMID: 36032114 PMCID: PMC9399944 DOI: 10.3389/fimmu.2022.967953] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2022] [Accepted: 07/25/2022] [Indexed: 11/15/2022] Open
Abstract
Background Emerging evidence supports that determination of intrathecal immunoglobulin M (IgM) synthesis (ITMS) and neurofilament light (NfL) concentration in cerebrospinal fluid (CSF) may be clinically useful as disease severity biomarkers in relapsing-remitting multiple sclerosis (RRMS). Methods Monocentric observational longitudinal cohort study in which prospectively collected data were retrospectively retrieved. Included were patients with RRMS (n=457) who had a diagnostic investigation including analysis of ITMS and CSF neurofilament light (cNfL). ITMS was calculated with the linear index formula, the intrathecal fraction of IgM according to Reiber (IgMIF), and by qualitative determination of oligoclonal IgM bands (OCMB). Univariable and multivariable models were performed to predict Evidence of Disease Activity-3 (EDA-3) status within 24 months from onset, and the risk of Expanded Disability Status Score (EDSS) ≥3 and ≥6. Results All investigated methods to calculate ITMS significantly predicted evidence of disease activity (EDA-3) within 24 months. IgMIF>0% showed the strongest association with EDA-3 status (adjusted hazard ratio [aHR] 3.7, 95%CI 2.7-5, p<0.001). Combining IgM-index>0.1 or OCMB with increased cNfL were strong predictors of EDSS≥3 (for cNfL+/IgM-index+: aHR 4.6, 95%CI 2.6-8.2, p<0.001) and EDSS≥6 (aHR 8.2, 95%CI 2.3-30, p<0.001). Conclusions In a real-world setting, ITMS was a useful biomarker in early RRMS to predict disabling MS and its prognostic value was even stronger in combination with cNfL. Our data suggest that determination of ITMS and cNfL should be included in the diagnostic work-up of RRMS for prognostic purposes and in decisions of disease-modifying therapy.
Collapse
Affiliation(s)
- Igal Rosenstein
- Department of Clinical Neuroscience, Institute of Neuroscience and Physiology at Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- *Correspondence: Igal Rosenstein,
| | - Sofia Rasch
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
| | - Markus Axelsson
- Department of Clinical Neuroscience, Institute of Neuroscience and Physiology at Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Lenka Novakova
- Department of Clinical Neuroscience, Institute of Neuroscience and Physiology at Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Kaj Blennow
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, University of Gothenburg, Mölndal, Sweden
| | - Henrik Zetterberg
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, University of Gothenburg, Mölndal, Sweden
- UK Dementia Research Institute at University College London (UCL), London, United Kingdom
- Department of Neurodegenerative Disease, University College London (UCL) Queen Square Institute of Neurology, London, United Kingdom
- Hong Kong Centre for Neurodegenerative Diseases, Hong Kong, Hong Kong SAR, China
| | - Jan Lycke
- Department of Clinical Neuroscience, Institute of Neuroscience and Physiology at Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| |
Collapse
|
18
|
Salvador F, Deramoudt L, Leprêtre F, Figeac M, Guerrier T, Boucher J, Bas M, Journiac N, Peters A, Mars LT, Zéphir H. A Spontaneous Model of Experimental Autoimmune Encephalomyelitis Provides Evidence of MOG-Specific B Cell Recruitment and Clonal Expansion. Front Immunol 2022; 13:755900. [PMID: 35185870 PMCID: PMC8850296 DOI: 10.3389/fimmu.2022.755900] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Accepted: 01/11/2022] [Indexed: 01/02/2023] Open
Abstract
The key role of B cells in the pathophysiology of multiple sclerosis (MS) is supported by the presence of oligoclonal bands in the cerebrospinal fluid, by the association of meningeal ectopic B cell follicles with demyelination, axonal loss and reduction of astrocytes, as well as by the high efficacy of B lymphocyte depletion in controlling inflammatory parameters of MS. Here, we use a spontaneous model of experimental autoimmune encephalomyelitis (EAE) to study the clonality of the B cell response targeting myelin oligodendrocyte glycoprotein (MOG). In particular, 94% of SJL/j mice expressing an I-As: MOG92-106 specific transgenic T cell receptor (TCR1640) spontaneously develop a chronic paralytic EAE between the age of 60-500 days. The immune response is triggered by the microbiota in the gut-associated lymphoid tissue, while there is evidence that the maturation of the autoimmune demyelinating response might occur in the cervical lymph nodes owing to local brain drainage. Using MOG-protein-tetramers we tracked the autoantigen-specific B cells and localized their enrichment to the cervical lymph nodes and among the brain immune infiltrate. MOG-specific IgG1 antibodies were detected in the serum of diseased TCR1640 mice and proved pathogenic upon adoptive transfer into disease-prone recipients. The ontogeny of the MOG-specific humoral response preceded disease onset coherent with their contribution to EAE initiation. This humoral response was, however, not sufficient for disease induction as MOG-antibodies could be detected at the age of 69 days in a model with an average age of onset of 197 days. To assess the MOG-specific B cell repertoire we FACS-sorted MOG-tetramer binding cells and clonally expand them in vitro to sequence the paratopes of the IgG heavy chain and kappa light chains. Despite the fragility of clonally expanding MOG-tetramer binding effector B cells, our results indicate the selection of a common CDR-3 clonotype among the Igk light chains derived from both disease-free and diseased TCR1640 mice. Our study demonstrates the pre-clinical mobilization of the MOG-specific B cell response within the brain-draining cervical lymph nodes, and reiterates that MOG antibodies are a poor biomarker of disease onset and progression.
Collapse
Affiliation(s)
- Florent Salvador
- Univ. Lille, Inserm, CHU Lille, Laboratory of Neuroinflammation and Multiple Sclerosis (NEMESIS), UMR-S1172, Lille Neuroscience & Cognition, LICEND, FHU Imminent, Lille, France
| | - Laure Deramoudt
- Univ. Lille, Inserm, CHU Lille, Laboratory of Neuroinflammation and Multiple Sclerosis (NEMESIS), UMR-S1172, Lille Neuroscience & Cognition, LICEND, FHU Imminent, Lille, France
| | - Frédéric Leprêtre
- UMS2014-US51, Genomics and Structural Platform, Lille University, Lille, France
| | - Martin Figeac
- UMS2014-US51, Genomics and Structural Platform, Lille University, Lille, France
| | - Thomas Guerrier
- Univ. Lille, Inserm, CHU Lille, U1286, INFINITE-Institute for Translational Research in Inflammation, Lille, France
| | - Julie Boucher
- Univ. Lille, Inserm, CHU Lille, Laboratory of Neuroinflammation and Multiple Sclerosis (NEMESIS), UMR-S1172, Lille Neuroscience & Cognition, LICEND, FHU Imminent, Lille, France
| | - Mathilde Bas
- Univ. Lille, Inserm, CHU Lille, Laboratory of Neuroinflammation and Multiple Sclerosis (NEMESIS), UMR-S1172, Lille Neuroscience & Cognition, LICEND, FHU Imminent, Lille, France
| | - Nathalie Journiac
- Univ. Lille, Inserm, CHU Lille, Laboratory of Neuroinflammation and Multiple Sclerosis (NEMESIS), UMR-S1172, Lille Neuroscience & Cognition, LICEND, FHU Imminent, Lille, France
| | - Anneli Peters
- Institute of Clinical Neuroimmunology, Hospital and Biomedical Center of the Ludwig-Maximilian University (LMU), Martinsried, Germany
| | - Lennart T Mars
- Univ. Lille, Inserm, CHU Lille, Laboratory of Neuroinflammation and Multiple Sclerosis (NEMESIS), UMR-S1172, Lille Neuroscience & Cognition, LICEND, FHU Imminent, Lille, France
| | - Hélène Zéphir
- Univ. Lille, Inserm, CHU Lille, Laboratory of Neuroinflammation and Multiple Sclerosis (NEMESIS), UMR-S1172, Lille Neuroscience & Cognition, LICEND, FHU Imminent, Lille, France.,Institute of Clinical Neuroimmunology, Hospital and Biomedical Center of the Ludwig-Maximilian University (LMU), Martinsried, Germany.,CRC-SEP of Lille, CHU of Lille, Lille, France
| |
Collapse
|
19
|
Arneth B, Kraus J. Experimental laboratory biomarkers in multiple sclerosis. Wien Med Wochenschr 2022; 172:346-358. [PMID: 35254566 DOI: 10.1007/s10354-022-00920-7] [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: 10/13/2021] [Accepted: 01/27/2022] [Indexed: 10/18/2022]
Abstract
BACKGROUND Multiple sclerosis (MS) is a chronic autoimmune disorder of the central nervous system; the cause of this condition remains unknown. Researchers have analyzed different biomarkers related to MS. Here, experimental laboratory biomarkers for MS are identified and analyzed. METHODS The current study examined articles investigating biomarkers for MS. Records were obtained from the PubMed, LILACS, and EBSCO databases using an identical search strategy and terms that included "multiple sclerosis," "MS," and "biomarkers." In the current review, we also focus on lesser known biomarkers that have not yet been established for use in clinical practice. RESULTS Previous studies have explored molecular substances that may help diagnose MS and manage its adverse effects. Commonly studied factors include neurofilaments, sCD163, CXCL13, NEO, NF‑L, OPN, B cells, T cells, and integrin-binding proteins. CONCLUSIONS Interactions between environmental and genetic factors have been implicated in the development of MS. Previous investigations have identified a wide range of biomarkers that can be used for diagnosis and disease management. These molecules and their associated studies provide vital insight and data to help primary physicians improve clinical and health outcomes for MS patients.
Collapse
Affiliation(s)
- Borros Arneth
- Institute of Laboratory Medicine and Pathobiochemistry, Justus Liebig University Giessen, Giessen, Germany.
| | - Jörg Kraus
- Department of Laboratory Medicine, Paracelsus Medical University and Salzburger Landeskliniken, Salzburg, Austria.,Department of Neurology, Medical Faculty, Heinrich Heine University, Düsseldorf, Germany
| |
Collapse
|
20
|
Halperin ST, ’t Hart BA, Luchicchi A, Schenk GJ. The Forgotten Brother: The Innate-like B1 Cell in Multiple Sclerosis. Biomedicines 2022; 10:606. [PMID: 35327408 PMCID: PMC8945227 DOI: 10.3390/biomedicines10030606] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2022] [Revised: 02/21/2022] [Accepted: 03/01/2022] [Indexed: 02/04/2023] Open
Abstract
Multiple sclerosis (MS) is a neurodegenerative disease of the central nervous system (CNS), traditionally considered a chronic autoimmune attack against the insulating myelin sheaths around axons. However, the exact etiology has not been identified and is likely multi-factorial. Recently, evidence has been accumulating that implies that autoimmune processes underlying MS may, in fact, be triggered by pathological processes initiated within the CNS. This review focuses on a relatively unexplored immune cell-the "innate-like" B1 lymphocyte. The B1 cell is a primary-natural-antibody- and anti-inflammatory-cytokine-producing cell present in the healthy brain. It has been recently shown that its frequency and function may differ between MS patients and healthy controls, but its exact involvement in the MS pathogenic process remains obscure. In this review, we propose that this enigmatic cell may play a more prominent role in MS pathology than ever imagined. We aim to shed light on the human B1 cell in health and disease, and how dysregulation in its delicate homeostatic role could impact MS. Furthermore, novel therapeutic avenues to restore B1 cells' beneficial functions will be proposed.
Collapse
Affiliation(s)
| | | | - Antonio Luchicchi
- Department of Anatomy and Neurosciences, Amsterdam Neuroscience, MS Center Amsterdam, Amsterdam UMC, Vrije Universiteit, 1081 HZ Amsterdam, The Netherlands; (S.T.H.); (B.A.’t.H.)
| | - Geert J. Schenk
- Department of Anatomy and Neurosciences, Amsterdam Neuroscience, MS Center Amsterdam, Amsterdam UMC, Vrije Universiteit, 1081 HZ Amsterdam, The Netherlands; (S.T.H.); (B.A.’t.H.)
| |
Collapse
|
21
|
Lindeman I, Polak J, Qiao S, Holmøy T, Høglund RA, Vartdal F, Berg‐Hansen P, Sollid LM, Lossius A. Stereotyped B‐cell responses are linked to IgG constant region polymorphisms in multiple sclerosis. Eur J Immunol 2022; 52:550-565. [DOI: 10.1002/eji.202149576] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2021] [Revised: 11/19/2021] [Accepted: 01/10/2022] [Indexed: 11/07/2022]
Affiliation(s)
- Ida Lindeman
- Department of Immunology Oslo University Hospital Oslo Norway
- Department of Immunology Institute of Clinical Medicine University of Oslo Norway
- K.G. Jebsen Coeliac Disease Research Centre University of Oslo Norway
| | - Justyna Polak
- Department of Immunology Institute of Clinical Medicine University of Oslo Norway
- K.G. Jebsen Coeliac Disease Research Centre University of Oslo Norway
| | - Shuo‐Wang Qiao
- Department of Immunology Oslo University Hospital Oslo Norway
- Department of Immunology Institute of Clinical Medicine University of Oslo Norway
- K.G. Jebsen Coeliac Disease Research Centre University of Oslo Norway
| | - Trygve Holmøy
- Department of Neurology Akershus University Hospital Lørenskog Norway
- Department of Neurology Institute of Clinical Medicine University of Oslo Norway
| | - Rune A. Høglund
- Department of Neurology Akershus University Hospital Lørenskog Norway
- Department of Neurology Institute of Clinical Medicine University of Oslo Norway
| | - Frode Vartdal
- Department of Immunology Institute of Clinical Medicine University of Oslo Norway
- K.G. Jebsen Coeliac Disease Research Centre University of Oslo Norway
| | - Pål Berg‐Hansen
- Department of Neurology Oslo University Hospital Oslo Norway
| | - Ludvig M. Sollid
- Department of Immunology Oslo University Hospital Oslo Norway
- Department of Immunology Institute of Clinical Medicine University of Oslo Norway
- K.G. Jebsen Coeliac Disease Research Centre University of Oslo Norway
| | - Andreas Lossius
- K.G. Jebsen Coeliac Disease Research Centre University of Oslo Norway
- Department of Neurology Akershus University Hospital Lørenskog Norway
- Department of Molecular Medicine Institute of Basic Medical Sciences University of Oslo Norway
| |
Collapse
|
22
|
Cerebrospinal Fluid IgM and Oligoclonal IgG Bands in Multiple Sclerosis: A Meta-Analysis of Prevalence and Prognosis. Brain Sci 2021; 11:brainsci11111444. [PMID: 34827444 PMCID: PMC8615995 DOI: 10.3390/brainsci11111444] [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: 10/04/2021] [Revised: 10/25/2021] [Accepted: 10/28/2021] [Indexed: 11/17/2022] Open
Abstract
The presence of intrathecal IgM synthesis (ITMS) has been associated with an aggressive multiple sclerosis (MS) clinical course. In the present systematic review, we aimed at assessing the prevalence of ITMS among different MS phenotypes. Moreover, we aimed at quantifying the risk of a second relapse in ITMS positive and oligoclonal IgG bands (OCGBs)-positive patients. We selected clinical studies reporting the ITMS prevalence assessed as oligoclonal IgM Bands (OCMBs), lipid-specific OCMBs (LS-OCMBs), and/or as an intrathecal IgM production > 0% (IgMLoc, Reiber formula). The overall prevalence of ITMS was higher in relapsing-remitting (RR) than clinically isolated syndrome (CIS) patients (40.1% versus 23.8%, p < 0.00001), while was in line with that detected in primary progressive MS (PPMS, 26.7%). Almost all patients (98%) with ITMS had also OCGBs. The risk of having a second relapse was higher in OCGBs positive patients (HR = 2.18, p = 0.007) but much higher in ITMS positive patients (HR = 3.62, p = 0.0005). This study revealed that the prevalence of ITMS is higher in RRMS patients. It suggests that the risk of having a second relapse, previously ascribed to OCGBs, may, to a certain extent, be related to the presence of intrathecal IgM.
Collapse
|
23
|
Next Generation Sequencing of Cerebrospinal Fluid B Cell Repertoires in Multiple Sclerosis and Other Neuro-Inflammatory Diseases-A Comprehensive Review. Diagnostics (Basel) 2021; 11:diagnostics11101871. [PMID: 34679570 PMCID: PMC8534365 DOI: 10.3390/diagnostics11101871] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2021] [Revised: 09/29/2021] [Accepted: 10/05/2021] [Indexed: 11/17/2022] Open
Abstract
During the last few decades, the role of B cells has been well established and redefined in neuro-inflammatory diseases, including multiple sclerosis and autoantibody-associated diseases. In particular, B cell maturation and trafficking across the blood–brain barrier (BBB) has recently been deciphered with the development of next-generation sequencing (NGS) approaches, which allow the assessment of representative cerebrospinal fluid (CSF) and peripheral blood B cell repertoires. In this review, we perform literature research focusing on NGS studies that allow further insights into B cell pathophysiology during neuro-inflammation. Besides the analysis of CSF B cells, the paralleled assessment of peripheral blood B cell repertoire provides deep insights into not only the CSF compartment, but also in B cell trafficking patterns across the BBB. In multiple sclerosis, CSF-specific B cell maturation, in combination with a bidirectional exchange of B cells across the BBB, is consistently detectable. These data suggest that B cells most likely encounter antigen(s) within the CSF and migrate across the BBB, with further maturation also taking place in the periphery. Autoantibody-mediated diseases, such as neuromyelitis optica spectrum disorder and LGI1 / NMDAR encephalitis, also show features of a CSF-specific B cell maturation and clonal connectivity with peripheral blood. In conclusion, these data suggest an intense exchange of B cells across the BBB, possibly feeding autoimmune circuits. Further developments in sequencing technologies will help to dissect the exact pathophysiologic mechanisms of B cells during neuro-inflammation.
Collapse
|
24
|
Oechtering J, Schaedelin S, Benkert P, Müller S, Achtnichts L, Vehoff J, Disanto G, Findling O, Fischer-Barnicol B, Orleth A, Chan A, Pot C, Barakovic M, Rahmanzadeh R, Galbusera R, Heijnen I, Lalive PH, Wuerfel J, Subramaniam S, Aeschbacher S, Conen D, Naegelin Y, Maceski A, Meier S, Berger K, Wiendl H, Lincke T, Lieb J, Yaldizli Ö, Sinnecker T, Derfuss T, Regeniter A, Zecca C, Gobbi C, Kappos L, Granziera C, Leppert D, Kuhle J. Intrathecal Immunoglobulin M Synthesis is an Independent Biomarker for Higher Disease Activity and Severity in Multiple Sclerosis. Ann Neurol 2021; 90:477-489. [PMID: 34057235 PMCID: PMC8518907 DOI: 10.1002/ana.26137] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2021] [Revised: 05/26/2021] [Accepted: 05/27/2021] [Indexed: 11/12/2022]
Abstract
OBJECTIVE We aimed to determine in relapsing multiple sclerosis (MS) whether intrathecal synthesis of immunoglobulin (Ig) M and IgG is associated with outcomes reflecting inflammatory activity and chronic worsening. METHODS We compared cerebrospinal fluid analysis, clinical and magnetic resonance imaging data, and serum neurofilament light chain (sNfL) levels at baseline and follow-up in 530 patients with relapsing MS. Patients were categorized by the presence of oligoclonal IgG bands (OCGB) and intrathecal synthesis of IgG and IgM (intrathecal fraction [IF]: IgGIF and IgMIF ). Relationships with the time to first relapse, sNfL concentrations, T2-weighted (T2w) lesions, MS Severity Score (MSSS), and time to initiation of high-efficacy therapy were analyzed in covariate-adjusted statistical models. RESULTS By categorical analysis, in patients with IgMIF the median time to first relapse was 28 months shorter and MSSS on average higher by 1.11 steps compared with patients without intrathecal immunoglobulin synthesis. Moreover, patients with IgMIF had higher sNfL concentrations, more new/enlarging T2w lesions, and higher total T2w lesion counts (all p ≤ 0.01). These associations were absent or equally smaller in patients who were positive for only OCGB or OCGB/IgGIF . Furthermore, quantitative analyses revealed that in patients with IgMIF ≥ median, the time to first relapse and to initiation of high-efficacy therapy was shorter by 32 and by 203 months, respectively (both p < 0.01), in comparison to patients with IgMIF < median. Dose-dependent associations were also found for IgMIF but not for IgGIF with magnetic resonance imaging-defined disease activity and sNfL. INTERPRETATION This large study supports the value of intrathecal IgM synthesis as an independent biomarker of disease activity and severity in relapsing MS. ANN NEUROL 2021.
Collapse
Affiliation(s)
- Johanna Oechtering
- Neurology Clinic and Policlinic, MS Center and Research Center for Clinical Neuroimmunology and Neuroscience Basel (RC2NB), University Hospital Basel, University of Basel, Basel, Switzerland
| | - Sabine Schaedelin
- Clinical Trial Unit, Department of Clinical Research, University Hospital Basel, University of Basel, Basel, Switzerland
| | - Pascal Benkert
- Clinical Trial Unit, Department of Clinical Research, University Hospital Basel, University of Basel, Basel, Switzerland
| | - Stefanie Müller
- Department of Neurology, Cantonal Hospital St. Gallen, St. Gallen, Switzerland
| | - Lutz Achtnichts
- Department of Neurology, Cantonal Hospital Aarau, Aarau, Switzerland
| | - Jochen Vehoff
- Department of Neurology, Cantonal Hospital St. Gallen, St. Gallen, Switzerland
| | - Giulio Disanto
- Neurocentre of Southern Switzerland, Multiple Sclerosis Centre, Ospedale Civico, Lugano, Switzerland
| | - Oliver Findling
- Department of Neurology, Cantonal Hospital Aarau, Aarau, Switzerland
| | - Bettina Fischer-Barnicol
- Neurology Clinic and Policlinic, MS Center and Research Center for Clinical Neuroimmunology and Neuroscience Basel (RC2NB), University Hospital Basel, University of Basel, Basel, Switzerland
| | - Annette Orleth
- Neurology Clinic and Policlinic, MS Center and Research Center for Clinical Neuroimmunology and Neuroscience Basel (RC2NB), University Hospital Basel, University of Basel, Basel, Switzerland
| | - Andrew Chan
- Department of Neurology, Inselspital, Bern University Hospital and University of Bern, Bern, Switzerland
| | - Caroline Pot
- Division of Neurology, Department of Clinical Neurosciences, Lausanne University Hospital (CHUV) and University of Lausanne, Lausanne, Switzerland
| | - Muhamed Barakovic
- Neurology Clinic and Policlinic, MS Center and Research Center for Clinical Neuroimmunology and Neuroscience Basel (RC2NB), University Hospital Basel, University of Basel, Basel, Switzerland.,Translational Imaging in Neurology (ThINk) Basel, Department of Biomedical Engineering, Faculty of Medicine, University Hospital Basel and University of Basel, Basel, Switzerland
| | - Reza Rahmanzadeh
- Neurology Clinic and Policlinic, MS Center and Research Center for Clinical Neuroimmunology and Neuroscience Basel (RC2NB), University Hospital Basel, University of Basel, Basel, Switzerland.,Translational Imaging in Neurology (ThINk) Basel, Department of Biomedical Engineering, Faculty of Medicine, University Hospital Basel and University of Basel, Basel, Switzerland
| | - Riccardo Galbusera
- Neurology Clinic and Policlinic, MS Center and Research Center for Clinical Neuroimmunology and Neuroscience Basel (RC2NB), University Hospital Basel, University of Basel, Basel, Switzerland.,Translational Imaging in Neurology (ThINk) Basel, Department of Biomedical Engineering, Faculty of Medicine, University Hospital Basel and University of Basel, Basel, Switzerland
| | - Ingmar Heijnen
- Division of Medical Immunology, Laboratory Medicine, University Hospital Basel, Basel, Switzerland
| | - Patrice H Lalive
- Department of Clinical Neurosciences, Division of Neurology, Geneva University Hospital, Geneva, Switzerland.,Diagnostic Department, Division of Laboratory Medicine, Geneva University Hospital, Geneva, Switzerland.,Department of Pathology and Immunology, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Jens Wuerfel
- Medical Image Analysis Center (MIAC) and Department of Biomedical Engineering, University Basel, Basel, Switzerland
| | - Suvitha Subramaniam
- Clinical Trial Unit, Department of Clinical Research, University Hospital Basel, University of Basel, Basel, Switzerland
| | - Stefanie Aeschbacher
- Cardiovascular Research Institute Basel, University Hospital Basel, Basel, Switzerland
| | - David Conen
- Population Health Research Institute, McMaster University, Hamilton, Ontario, Canada
| | - Yvonne Naegelin
- Neurology Clinic and Policlinic, MS Center and Research Center for Clinical Neuroimmunology and Neuroscience Basel (RC2NB), University Hospital Basel, University of Basel, Basel, Switzerland
| | - Aleksandra Maceski
- Neurology Clinic and Policlinic, MS Center and Research Center for Clinical Neuroimmunology and Neuroscience Basel (RC2NB), University Hospital Basel, University of Basel, Basel, Switzerland
| | - Stephanie Meier
- Neurology Clinic and Policlinic, MS Center and Research Center for Clinical Neuroimmunology and Neuroscience Basel (RC2NB), University Hospital Basel, University of Basel, Basel, Switzerland
| | - Klaus Berger
- Institute of Epidemiology and Social Medicine, University of Münster, Münster, Germany
| | - Heinz Wiendl
- Department of Neurology with Institute of Translational Neurology, University Hospital Münster, Münster, Germany
| | - Therese Lincke
- Division of Neuroradiology, Department of Radiology and Nuclear Medicine, University Hospital Basel, University of Basel, Basel, Switzerland
| | - Johanna Lieb
- Division of Neuroradiology, Department of Radiology and Nuclear Medicine, University Hospital Basel, University of Basel, Basel, Switzerland
| | - Özgür Yaldizli
- Neurology Clinic and Policlinic, MS Center and Research Center for Clinical Neuroimmunology and Neuroscience Basel (RC2NB), University Hospital Basel, University of Basel, Basel, Switzerland.,Translational Imaging in Neurology (ThINk) Basel, Department of Biomedical Engineering, Faculty of Medicine, University Hospital Basel and University of Basel, Basel, Switzerland
| | - Tim Sinnecker
- Neurology Clinic and Policlinic, MS Center and Research Center for Clinical Neuroimmunology and Neuroscience Basel (RC2NB), University Hospital Basel, University of Basel, Basel, Switzerland.,Medical Image Analysis Center (MIAC) and Department of Biomedical Engineering, University Basel, Basel, Switzerland
| | - Tobias Derfuss
- Neurology Clinic and Policlinic, MS Center and Research Center for Clinical Neuroimmunology and Neuroscience Basel (RC2NB), University Hospital Basel, University of Basel, Basel, Switzerland
| | | | - Chiara Zecca
- Neurocentre of Southern Switzerland, Multiple Sclerosis Centre, Ospedale Civico, Lugano, Switzerland.,Faculty of Biomedical Sciences, Università della Svizzera italiana, Lugano, Switzerland
| | - Claudio Gobbi
- Neurology Clinic and Policlinic, MS Center and Research Center for Clinical Neuroimmunology and Neuroscience Basel (RC2NB), University Hospital Basel, University of Basel, Basel, Switzerland.,Neurocentre of Southern Switzerland, Multiple Sclerosis Centre, Ospedale Civico, Lugano, Switzerland.,Faculty of Biomedical Sciences, Università della Svizzera italiana, Lugano, Switzerland
| | - Ludwig Kappos
- Neurology Clinic and Policlinic, MS Center and Research Center for Clinical Neuroimmunology and Neuroscience Basel (RC2NB), University Hospital Basel, University of Basel, Basel, Switzerland
| | - Cristina Granziera
- Neurology Clinic and Policlinic, MS Center and Research Center for Clinical Neuroimmunology and Neuroscience Basel (RC2NB), University Hospital Basel, University of Basel, Basel, Switzerland.,Translational Imaging in Neurology (ThINk) Basel, Department of Biomedical Engineering, Faculty of Medicine, University Hospital Basel and University of Basel, Basel, Switzerland
| | - David Leppert
- Neurology Clinic and Policlinic, MS Center and Research Center for Clinical Neuroimmunology and Neuroscience Basel (RC2NB), University Hospital Basel, University of Basel, Basel, Switzerland
| | - Jens Kuhle
- Neurology Clinic and Policlinic, MS Center and Research Center for Clinical Neuroimmunology and Neuroscience Basel (RC2NB), University Hospital Basel, University of Basel, Basel, Switzerland
| | | |
Collapse
|
25
|
Weng R, Liu S, Gu X, Zhong Z. Characterization of the B cell receptor repertoire of patients with acute coronary syndrome. Genes Genomics 2021; 44:19-28. [PMID: 33974240 DOI: 10.1007/s13258-021-01110-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2020] [Accepted: 04/29/2021] [Indexed: 01/01/2023]
Abstract
BACKGROUND Acute coronary syndrome (ACS) is a complex cardiovascular disease whose development involves the dysregulation of adaptive immune responses. Though it has been proven that T cells associate with inflammation in the development of ACS, the function of B cells in disease remains unclear. OBJECTIVE The aim of this study was to reveal the diversity of the B cell receptor (BCR) repertoire of patients with ACS. METHODS We conducted a pilot study to sequence the immune repertoire of peripheral blood mononuclear cells (PBMCs) from patients with ACS, including acute myocardial infarction (AMI) and unstable angina (UA), and quantitatively characterized BCR repertoires by bioinformatics analysis. RESULTS We found that patients with AMI and UA had lower BCR repertoire diversity compared with controls with normal coronary arteries (NCA). Lower percentages of productive unique BCR nt sequences and higher percentages of top 200 unique BCR sequences were identified in AMI and UA patients than NCA controls. Patients had various preferential usage of V and J genes from B cell clones in accordance with the disease severity of coronary arteries. AMI patients had distinct CDR3 amino acids, and their frequency differed among patients with ACS. CONCLUSIONS Our results indicate that differential BCR signatures represent an imprint of distinct repertoires among ACS patients. This study thereby opens up the prospect of studying disease-relevant B cells to better understand and treat ACS.
Collapse
Affiliation(s)
- Ruiqiang Weng
- Center for Precision Medicine, Meizhou People's Hospital (Huangtang Hospital), Meizhou Hospital Affiliated to Sun Yat-sen University, No. 63 Huangtang Road, Meijiang District, Meizhou, 514031, People's Republic of China
- Guangdong Provincial Engineering and Technological Research Center for Molecular Diagnostics of Cardiovascular Diseases, Meizhou, 514031, People's Republic of China
- Guangdong Provincial Key Laboratory of Precision Medicine and Clinical Translational Research of Hakka Population, Meizhou, 514031, People's Republic of China
| | - Sudong Liu
- Center for Precision Medicine, Meizhou People's Hospital (Huangtang Hospital), Meizhou Hospital Affiliated to Sun Yat-sen University, No. 63 Huangtang Road, Meijiang District, Meizhou, 514031, People's Republic of China
- Guangdong Provincial Engineering and Technological Research Center for Molecular Diagnostics of Cardiovascular Diseases, Meizhou, 514031, People's Republic of China
- Guangdong Provincial Key Laboratory of Precision Medicine and Clinical Translational Research of Hakka Population, Meizhou, 514031, People's Republic of China
| | - Xiaodong Gu
- Center for Precision Medicine, Meizhou People's Hospital (Huangtang Hospital), Meizhou Hospital Affiliated to Sun Yat-sen University, No. 63 Huangtang Road, Meijiang District, Meizhou, 514031, People's Republic of China
- Guangdong Provincial Engineering and Technological Research Center for Molecular Diagnostics of Cardiovascular Diseases, Meizhou, 514031, People's Republic of China
- Guangdong Provincial Key Laboratory of Precision Medicine and Clinical Translational Research of Hakka Population, Meizhou, 514031, People's Republic of China
| | - Zhixiong Zhong
- Center for Precision Medicine, Meizhou People's Hospital (Huangtang Hospital), Meizhou Hospital Affiliated to Sun Yat-sen University, No. 63 Huangtang Road, Meijiang District, Meizhou, 514031, People's Republic of China.
- Guangdong Provincial Engineering and Technological Research Center for Molecular Diagnostics of Cardiovascular Diseases, Meizhou, 514031, People's Republic of China.
- Center for Cardiovascular Diseases, Meizhou People's Hospital (Huangtang Hospital), Meizhou Hospital Affiliated to Sun Yat-sen University, Meizhou, 514031, People's Republic of China.
| |
Collapse
|
26
|
Wu M, Zhao M, Wu H, Lu Q. Immune repertoire: Revealing the "real-time" adaptive immune response in autoimmune diseases. Autoimmunity 2021; 54:61-75. [PMID: 33650440 DOI: 10.1080/08916934.2021.1887149] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
Abstract
The diversity of the immune repertoire (IR) enables the human immune system to distinguish multifarious antigens (Ags) that humans may encounter throughout life. At the same time, bias or abnormalities in the IR also pay a contribution to the pathogenesis of autoimmune diseases. Rapid advancements in high-throughput sequencing (HTS) technology have ushered in a new era of immune studies, revealing novel molecules and pathways that might result in autoimmunity. In the field of IR, HTS can monitor the immune response status and identify disease-specific immune repertoires. In this review, we summarize updated progress on the mechanisms of the IR and current related studies on four autoimmune diseases, particularly focusing on systemic lupus erythematosus (SLE). These autoimmune diseases can exhibit slightly or significantly skewed IRs and provide novel insights that inform our comprehending of disease pathogenesis and provide potential targets for diagnosis and treatment.
Collapse
Affiliation(s)
- Meiyu Wu
- Department of Dermatology, Second Xiangya Hospital, Central South University, Hunan Key Laboratory of Medical Epigenomics, Changsha, Hunan, China
| | - Ming Zhao
- Department of Dermatology, Second Xiangya Hospital, Central South University, Hunan Key Laboratory of Medical Epigenomics, Changsha, Hunan, China
| | - Haijing Wu
- Department of Dermatology, Second Xiangya Hospital, Central South University, Hunan Key Laboratory of Medical Epigenomics, Changsha, Hunan, China
| | - Qianjin Lu
- Department of Dermatology, Second Xiangya Hospital, Central South University, Hunan Key Laboratory of Medical Epigenomics, Changsha, Hunan, China.,Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing, Jiangsu, China
| |
Collapse
|
27
|
Tsamis KI, Sakkas H, Giannakis A, Ryu HS, Gartzonika C, Nikas IP. Evaluating Infectious, Neoplastic, Immunological, and Degenerative Diseases of the Central Nervous System with Cerebrospinal Fluid-Based Next-Generation Sequencing. Mol Diagn Ther 2021; 25:207-229. [PMID: 33646562 PMCID: PMC7917176 DOI: 10.1007/s40291-021-00513-x] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/07/2021] [Indexed: 12/24/2022]
Abstract
Cerebrospinal fluid (CSF) is a clear and paucicellular fluid that circulates within the ventricular system and the subarachnoid space of the central nervous system (CNS), and diverse CNS disorders can impact its composition, volume, and flow. As conventional CSF testing suffers from suboptimal sensitivity, this review aimed to evaluate the role of next-generation sequencing (NGS) in the work-up of infectious, neoplastic, neuroimmunological, and neurodegenerative CNS diseases. Metagenomic NGS showed improved sensitivity—compared to traditional methods—to detect bacterial, viral, parasitic, and fungal infections, while the overall performance was maximized in some studies when all diagnostic modalities were used. In patients with primary CNS cancer, NGS findings in the CSF were largely concordant with the molecular signatures derived from tissue-based molecular analysis; of interest, additional mutations were identified in the CSF in some glioma studies, reflecting intratumoral heterogeneity. In patients with metastasis to the CNS, NGS facilitated diagnosis, prognosis, therapeutic management, and monitoring, exhibiting higher sensitivity than neuroimaging, cytology, and plasma-based molecular analysis. Although evidence is still rudimentary, NGS could enhance the diagnosis and pathogenetic understanding of multiple sclerosis in addition to Alzheimer and Parkinson disease. To conclude, NGS has shown potential to aid the research, facilitate the diagnostic approach, and improve the management outcomes of all the aforementioned CNS diseases. However, to establish its role in clinical practice, the clinical validity and utility of each NGS protocol should be determined. Lastly, as most evidence has been derived from small and retrospective studies, results from randomized control trials could be of significant value.
Collapse
Affiliation(s)
- Konstantinos I Tsamis
- Department of Neurology, University Hospital of Ioannina, 45500, Ioannina, Greece. .,School of Medicine, European University Cyprus, 2404, Nicosia, Cyprus.
| | - Hercules Sakkas
- Microbiology Department, Faculty of Medicine, School of Health Sciences, University of Ioannina, 45110, Ioannina, Greece
| | - Alexandros Giannakis
- Department of Neurology, University Hospital of Ioannina, 45500, Ioannina, Greece
| | - Han Suk Ryu
- Department of Pathology, Seoul National University Hospital, Seoul, 03080, Korea
| | - Constantina Gartzonika
- Microbiology Department, Faculty of Medicine, School of Health Sciences, University of Ioannina, 45110, Ioannina, Greece
| | - Ilias P Nikas
- School of Medicine, European University Cyprus, 2404, Nicosia, Cyprus
| |
Collapse
|
28
|
Simeonova D, Stoyanov D, Leunis JC, Murdjeva M, Maes M. Construction of a nitro-oxidative stress-driven, mechanistic model of mood disorders: A nomothetic network approach. Nitric Oxide 2021; 106:45-54. [PMID: 33186727 DOI: 10.1016/j.niox.2020.11.001] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Accepted: 11/05/2020] [Indexed: 12/17/2022]
Abstract
Major depression is accompanied by increased IgM-mediated autoimmune responses to oxidative specific epitopes (OSEs) and nitric oxide (NO)-adducts. These responses were not examined in bipolar disorder type 1 (BP1) and BP2. IgM responses to malondialdehyde (MDA), phosphatidinylinositol, oleic acid, azelaic acid, and NO-adducts were determined in 35 healthy controls, and 47 major depressed (MDD), 29 BP1, and 25 BP2 patients. We also measured serum peroxides, IgG to oxidized LDL (oxLDL), and IgM/IgA directed to lipopolysaccharides (LPS). IgM responses to OSEs and NO-adducts (OSENO) were significantly higher in MDD and BP1 as compared with controls, and IgM to OSEs higher in MDD than in BP2. Partial Least Squares (PLS) analysis showed that 57.7% of the variance in the clinical phenome of mood disorders was explained by number of episodes, a latent vector extracted from IgM to OSENO, IgG to oxLDL, and peroxides. There were significant specific indirect effects of IgA/IgM to LPS on the clinical phenome, which were mediated by peroxides, IgM OSENO, and IgG oxLDL. Using PLS we have constructed a data-driven nomothetic network which ensembled causome (increased plasma LPS load), adverse outcome pathways (namely neuro-affective toxicity), and clinical phenome features of mood disorders in a data-driven model. Based on those feature sets, cluster analysis discovered a new diagnostic class characterized by increased plasma LPS load, peroxides, autoimmune responses to OSENO, and increased phenome scores. Using the new nomothetic network approach, we constructed a mechanistically transdiagnostic diagnostic class indicating neuro-affective toxicity in 74.3% of the mood disorder patients.
Collapse
Affiliation(s)
- Denitsa Simeonova
- Department of Psychiatry and Medical Psychology, Medical Faculty, Medical University of Plovdiv, Plovdiv, Bulgaria; Research Institute, Medical University, Plovdiv, Bulgaria
| | - Drozdstoy Stoyanov
- Department of Psychiatry and Medical Psychology, Medical Faculty, Medical University of Plovdiv, Plovdiv, Bulgaria; Research Institute, Medical University, Plovdiv, Bulgaria
| | | | - Marianna Murdjeva
- Research Institute, Medical University, Plovdiv, Bulgaria; Department of Microbiology and Immunology, Faculty of Pharmacy, Medical University of Plovdiv, Plovdiv, Bulgaria; Section of Immunological Assessment of Chronic Stress, Technological Center of Emergency Medicine, Plovdiv, Bulgaria
| | - Michael Maes
- Department of Psychiatry and Medical Psychology, Medical Faculty, Medical University of Plovdiv, Plovdiv, Bulgaria; Department of Psychiatry, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand; Impact Research Center, Deakin University, Geelong, Australia.
| |
Collapse
|
29
|
Houen G, Trier NH, Frederiksen JL. Epstein-Barr Virus and Multiple Sclerosis. Front Immunol 2020; 11:587078. [PMID: 33391262 PMCID: PMC7773893 DOI: 10.3389/fimmu.2020.587078] [Citation(s) in RCA: 49] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2020] [Accepted: 11/18/2020] [Indexed: 12/11/2022] Open
Abstract
Multiple sclerosis (MS) is a neurologic disease affecting myelinated nerves in the central nervous system (CNS). The disease often debuts as a clinically isolated syndrome, e.g., optic neuritis (ON), which later develops into relapsing-remitting (RR) MS, with temporal attacks or primary progressive (PP) MS. Characteristic features of MS are inflammatory foci in the CNS and intrathecal synthesis of immunoglobulins (Igs), measured as an IgG index, oligoclonal bands (OCBs), or specific antibody indexes. Major predisposing factors for MS are certain tissue types (e.g., HLA DRB1*15:01), vitamin D deficiency, smoking, obesity, and infection with Epstein-Barr virus (EBV). Many of the clinical signs of MS described above can be explained by chronic/recurrent EBV infection and current models of EBV involvement suggest that RRMS may be caused by repeated entry of EBV-transformed B cells to the CNS in connection with attacks, while PPMS may be caused by more chronic activity of EBV-transformed B cells in the CNS. In line with the model of EBV's role in MS, new treatments based on monoclonal antibodies (MAbs) targeting B cells have shown good efficacy in clinical trials both for RRMS and PPMS, while MAbs inhibiting B cell mobilization and entry to the CNS have shown efficacy in RRMS. Thus, these agents, which are now first line therapy in many patients, may be hypothesized to function by counteracting a chronic EBV infection.
Collapse
Affiliation(s)
- Gunnar Houen
- Institute of Biochemistry and Molecular Biology, University of Southern Denmark, Odense, Denmark
- Department of Neurology, Rigshospitalet, Glostrup, Denmark
| | | | - Jette Lautrup Frederiksen
- Department of Neurology, Rigshospitalet, Glostrup, Denmark
- Institute of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
30
|
Negron A, Stüve O, Forsthuber TG. Ectopic Lymphoid Follicles in Multiple Sclerosis: Centers for Disease Control? Front Neurol 2020; 11:607766. [PMID: 33363512 PMCID: PMC7753025 DOI: 10.3389/fneur.2020.607766] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2020] [Accepted: 11/03/2020] [Indexed: 12/11/2022] Open
Abstract
While the contribution of autoreactive CD4+ T cells to the pathogenesis of Multiple Sclerosis (MS) is widely accepted, the advent of B cell-depleting monoclonal antibody (mAb) therapies has shed new light on the complex cellular mechanisms underlying MS pathogenesis. Evidence supports the involvement of B cells in both antibody-dependent and -independent capacities. T cell-dependent B cell responses originate and take shape in germinal centers (GCs), specialized microenvironments that regulate B cell activation and subsequent differentiation into antibody-secreting cells (ASCs) or memory B cells, a process for which CD4+ T cells, namely follicular T helper (TFH) cells, are indispensable. ASCs carry out their effector function primarily via secreted Ig but also through the secretion of both pro- and anti-inflammatory cytokines. Memory B cells, in addition to being capable of rapidly differentiating into ASCs, can function as potent antigen-presenting cells (APCs) to cognate memory CD4+ T cells. Aberrant B cell responses are prevented, at least in part, by follicular regulatory T (TFR) cells, which are key suppressors of GC-derived autoreactive B cell responses through the expression of inhibitory receptors and cytokines, such as CTLA4 and IL-10, respectively. Therefore, GCs represent a critical site of peripheral B cell tolerance, and their dysregulation has been implicated in the pathogenesis of several autoimmune diseases. In MS patients, the presence of GC-like leptomeningeal ectopic lymphoid follicles (eLFs) has prompted their investigation as potential sources of pathogenic B and T cell responses. This hypothesis is supported by elevated levels of CXCL13 and circulating TFH cells in the cerebrospinal fluid (CSF) of MS patients, both of which are required to initiate and maintain GC reactions. Additionally, eLFs in post-mortem MS patient samples are notably devoid of TFR cells. The ability of GCs to generate and perpetuate, but also regulate autoreactive B and T cell responses driving MS pathology makes them an attractive target for therapeutic intervention. In this review, we will summarize the evidence from both humans and animal models supporting B cells as drivers of MS, the role of GC-like eLFs in the pathogenesis of MS, and mechanisms controlling GC-derived autoreactive B cell responses in MS.
Collapse
Affiliation(s)
- Austin Negron
- Department of Biology, University of Texas at San Antonio, San Antonio, TX, United States
| | - Olaf Stüve
- Department of Neurology, University of Texas Southwestern Medical Center, Dallas, TX, United States.,Neurology Section, Veterans Affairs North Texas Health Care System, Medical Service, Dallas, TX, United States
| | - Thomas G Forsthuber
- Department of Biology, University of Texas at San Antonio, San Antonio, TX, United States
| |
Collapse
|
31
|
Magliozzi R, Mazziotti V, Montibeller L, Pisani AI, Marastoni D, Tamanti A, Rossi S, Crescenzo F, Calabrese M. Cerebrospinal Fluid IgM Levels in Association With Inflammatory Pathways in Multiple Sclerosis Patients. Front Cell Neurosci 2020; 14:569827. [PMID: 33192314 PMCID: PMC7596330 DOI: 10.3389/fncel.2020.569827] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2020] [Accepted: 09/22/2020] [Indexed: 11/23/2022] Open
Abstract
Background Intrathecal immunoglobulin M (IgM) synthesis has been demonstrated in the early disease stages of multiple sclerosis (MS) as a predictor factor of a worsening disease course. Similarly, increased cerebrospinal fluid (CSF) molecules related to B-cell intrathecal activity have been associated with a more severe MS progression. However, whether CSF levels of IgM are linked to specific inflammatory and clinical profile in MS patients at the time of diagnosis remains to be elucidated. Methods Using customized Bio-Plex assay, the protein levels of IgG, IgA, IgM, and of 34 other inflammatory molecules, related to B-cell, T-cell, and monocyte/macrophage activity, were analyzed in the CSF of 103 newly diagnosed relapsing–remitting MS patients and 36 patients with other neurological disorders. CSF IgM levels were also correlated with clinical and neuroradiological measures [advanced 3-T magnetic resonance imaging (MRI) parameters], at diagnosis and after 2 years of follow-up. Results A 45.6% increase in CSF IgM levels was found in MS patients compared to controls (p = 0.013). CSF IgM levels correlated with higher CSF levels of CXCL13 (p = 0.039), CCL21 (p = 0.023), interleukin 10 (IL-10) (p = 0.025), IL-12p70 (p = 0.020), CX3CL1 (p = 0.036), and CHI3L1 (p = 0.048) and were associated with earlier age of patients at diagnosis (p = 0.008), white matter lesion (WML) number (p = 0.039) and disease activity (p = 0.033) after 2 years of follow-up. Conclusion IgMs are the immunoglobulins mostly expressed in the CSF of naive MS patients compared to other neurological conditions at the time of diagnosis. The association between increased CSF IgM levels and molecules related to both B-cell immunity (IL-10) and recruitment (CXCL13 and CCL21) and to macrophage/microglia activity (IL-12p70, CX3CL1, and CHI3L1) suggests possible correlation between humoral and innate intrathecal immunity in early disease stage. Furthermore, the association of IgM levels with WMLs and MS clinical and MRI activity after 2 years supports the idea of key role of IgM in the disease course.
Collapse
Affiliation(s)
- Roberta Magliozzi
- Neurology Section of Department of Neurological and Movement Sciences, University of Verona, Verona, Italy.,Division of Brain Sciences, Department of Medicine, Imperial College London, London, United Kingdom
| | - Valentina Mazziotti
- Neurology Section of Department of Neurological and Movement Sciences, University of Verona, Verona, Italy
| | - Luigi Montibeller
- Neurology Section of Department of Neurological and Movement Sciences, University of Verona, Verona, Italy
| | - Anna I Pisani
- Neurology Section of Department of Neurological and Movement Sciences, University of Verona, Verona, Italy
| | - Damiano Marastoni
- Neurology Section of Department of Neurological and Movement Sciences, University of Verona, Verona, Italy
| | - Agnese Tamanti
- Neurology Section of Department of Neurological and Movement Sciences, University of Verona, Verona, Italy
| | - Stefania Rossi
- Department of Oncology and Molecular Medicine, National Institute of Health, Rome, Italy
| | - Francesco Crescenzo
- Neurology Section of Department of Neurological and Movement Sciences, University of Verona, Verona, Italy
| | - Massimiliano Calabrese
- Neurology Section of Department of Neurological and Movement Sciences, University of Verona, Verona, Italy
| |
Collapse
|
32
|
Abstract
B cells serve as a key weapon against infectious diseases. They also contribute to multiple autoimmune diseases, including multiple sclerosis (MS) where depletion of B cells is a highly effective therapy. We describe a comprehensive profile of central nervous system (CNS)-specific transcriptional B cell phenotypes in MS at single-cell resolution with paired immune repertoires. We reveal a polyclonal immunoglobulin M (IgM) and IgG1 cerebrospinal fluid B cell expansion polarized toward an inflammatory, memory and plasmablast/plasma cell phenotype, with differential up-regulation of specific proinflammatory pathways. We did not find evidence that CNS B cells harbor a neurotropic virus. These data support the targeting of activated resident B cells in the CNS as a potentially effective strategy for control of treatment-resistant chronic disease. Central nervous system B cells have several potential roles in multiple sclerosis (MS): secretors of proinflammatory cytokines and chemokines, presenters of autoantigens to T cells, producers of pathogenic antibodies, and reservoirs for viruses that trigger demyelination. To interrogate these roles, single-cell RNA sequencing (scRNA-Seq) was performed on paired cerebrospinal fluid (CSF) and blood from subjects with relapsing-remitting MS (RRMS; n = 12), other neurologic diseases (ONDs; n = 1), and healthy controls (HCs; n = 3). Single-cell immunoglobulin sequencing (scIg-Seq) was performed on a subset of these subjects and additional RRMS (n = 4), clinically isolated syndrome (n = 2), and OND (n = 2) subjects. Further, paired CSF and blood B cell subsets (RRMS; n = 7) were isolated using fluorescence activated cell sorting for bulk RNA sequencing (RNA-Seq). Independent analyses across technologies demonstrated that nuclear factor kappa B (NF-κB) and cholesterol biosynthesis pathways were activated, and specific cytokine and chemokine receptors were up-regulated in CSF memory B cells. Further, SMAD/TGF-β1 signaling was down-regulated in CSF plasmablasts/plasma cells. Clonally expanded, somatically hypermutated IgM+ and IgG1+ CSF B cells were associated with inflammation, blood–brain barrier breakdown, and intrathecal Ig synthesis. While we identified memory B cells and plasmablast/plasma cells with highly similar Ig heavy-chain sequences across MS subjects, similarities were also identified with ONDs and HCs. No viral transcripts, including from Epstein–Barr virus, were detected. Our findings support the hypothesis that in MS, CSF B cells are driven to an inflammatory and clonally expanded memory and plasmablast/plasma cell phenotype.
Collapse
|
33
|
Alcalá C, Pérez-Miralles FC, Gil-Perotín S, Casanova B. Reader response: Intrathecal IgM production is a strong risk factor for early conversion to multiple sclerosis. Neurology 2020; 95:277. [DOI: 10.1212/wnl.0000000000010137] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
|
34
|
Beltrán E, Gerdes LA, Hansen J, Flierl-Hecht A, Krebs S, Blum H, Ertl-Wagner B, Barkhof F, Kümpfel T, Hohlfeld R, Dornmair K. Early adaptive immune activation detected in monozygotic twins with prodromal multiple sclerosis. J Clin Invest 2020; 129:4758-4768. [PMID: 31566584 DOI: 10.1172/jci128475] [Citation(s) in RCA: 85] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2019] [Accepted: 08/01/2019] [Indexed: 12/31/2022] Open
Abstract
Multiple sclerosis (MS) is a disabling disease of the CNS. Inflammatory features of MS include lymphocyte accumulations in the CNS and cerebrospinal fluid (CSF). The preclinical events leading to established MS are still enigmatic. Here we compared gene expression patterns of CSF cells from MS-discordant monozygotic twin pairs. Six "healthy" co-twins, who carry a maximal familial risk for developing MS, showed subclinical neuroinflammation (SCNI) with small MRI lesions. Four of these subjects had oligoclonal bands (OCBs). By single-cell RNA sequencing of 2752 CSF cells, we identified clonally expanded CD8+ T cells, plasmablasts, and, to a lesser extent, CD4+ T cells not only from MS patients but also from subjects with SCNI. In contrast to nonexpanded T cells, clonally expanded T cells showed characteristics of activated tissue-resident memory T (TRM) cells. The TRM-like phenotype was detectable already in cells from SCNI subjects but more pronounced in cells from patients with definite MS. Expanded plasmablast clones were detected only in MS and SCNI subjects with OCBs. Our data provide evidence for very early concomitant activation of 3 components of the adaptive immune system in MS, with a notable contribution of clonally expanded TRM-like CD8+ cells.
Collapse
Affiliation(s)
- Eduardo Beltrán
- Institute of Clinical Neuroimmunology, Biomedical Center and Hospital
| | - Lisa Ann Gerdes
- Institute of Clinical Neuroimmunology, Biomedical Center and Hospital
| | - Julia Hansen
- Institute of Clinical Neuroimmunology, Biomedical Center and Hospital
| | | | - Stefan Krebs
- Laboratory for Functional Genome Analysis (LAFUGA), Gene Center; and
| | - Helmut Blum
- Laboratory for Functional Genome Analysis (LAFUGA), Gene Center; and
| | - Birgit Ertl-Wagner
- Department of Radiology, Grosshadern Medical Campus; Ludwig Maximilian University of Munich, Munich, Germany.,Department of Medical Imaging, The Hospital for Sick Children, University of Toronto, Toronto, Ontario, Canada
| | - Frederik Barkhof
- Department of Radiology and Nuclear Medicine, VU University Medical Center, Amsterdam, Netherlands.,UCL Institutes of Neurology and Healthcare Engineering, University College London, London, United Kingdom
| | - Tania Kümpfel
- Institute of Clinical Neuroimmunology, Biomedical Center and Hospital
| | - Reinhard Hohlfeld
- Institute of Clinical Neuroimmunology, Biomedical Center and Hospital.,Munich Cluster of Systems Neurology (SyNergy), Munich, Germany
| | - Klaus Dornmair
- Institute of Clinical Neuroimmunology, Biomedical Center and Hospital.,Munich Cluster of Systems Neurology (SyNergy), Munich, Germany
| |
Collapse
|
35
|
Gharibi T, Babaloo Z, Hosseini A, Marofi F, Ebrahimi-Kalan A, Jahandideh S, Baradaran B. The role of B cells in the immunopathogenesis of multiple sclerosis. Immunology 2020; 160:325-335. [PMID: 32249925 DOI: 10.1111/imm.13198] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2019] [Revised: 03/01/2020] [Accepted: 03/25/2020] [Indexed: 02/06/2023] Open
Abstract
There is ongoing debate on how B cells contribute to the pathogenesis of multiple sclerosis (MS). The success of B-cell targeting therapies in MS highlighted the role of B cells, particularly the antibody-independent functions of these cells such as antigen presentation to T cells and modulation of the function of T cells and myeloid cells by secreting pathogenic and/or protective cytokines in the central nervous system. Here, we discuss the role of different antibody-dependent and antibody-independent functions of B cells in MS disease activity and progression proposing new therapeutic strategies for the optimization of B-cell targeting treatments.
Collapse
Affiliation(s)
- Tohid Gharibi
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.,Department of Immunology, School of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran.,Neuroscience Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.,Department of Neurosciences and Cognition, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran.,Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Zohreh Babaloo
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.,Department of Immunology, School of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Arezoo Hosseini
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.,Department of Immunology, School of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran.,Neuroscience Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.,Department of Neurosciences and Cognition, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Faroogh Marofi
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.,Department of Immunology, School of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Abbas Ebrahimi-Kalan
- Neuroscience Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.,Department of Neurosciences and Cognition, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Saeed Jahandideh
- Department of Biochemistry, Pasteur Institute of Iran, Tehran, Iran
| | - Behzad Baradaran
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.,Department of Immunology, School of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
36
|
Meyer Zu Hörste G, Gross CC, Klotz L, Schwab N, Wiendl H. Next-Generation Neuroimmunology: New Technologies to Understand Central Nervous System Autoimmunity. Trends Immunol 2020; 41:341-354. [PMID: 32147112 DOI: 10.1016/j.it.2020.02.005] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2019] [Revised: 02/10/2020] [Accepted: 02/10/2020] [Indexed: 12/11/2022]
Abstract
Understanding neuroimmunological disorders is essential for developing new diagnostic and therapeutic strategies. Rodent models have provided valuable insights, but are sometimes equated with their human counterparts. Here, we summarize how novel technologies may enable an improved human-focused view of immune mechanisms. Recent studies have applied these new technologies to the brain parenchyma, its surrounding cerebrospinal fluid, and peripheral immune compartments. Therapeutic interventions have also facilitated translational understanding in a reverse way. However, with improved technology, access to patient samples remains a rate-limiting step in translational research. We anticipate that next-generation neuroimmunology is likely to integrate, in the immediate future, diverse technical tools for optimal diagnosis, prognosis, and treatment of neuroimmunological disorders.
Collapse
Affiliation(s)
- Gerd Meyer Zu Hörste
- Department of Neurology with Institute of Translational Neurology, University Hospital Münster, University of Münster, Münster, Germany
| | - Catharina C Gross
- Department of Neurology with Institute of Translational Neurology, University Hospital Münster, University of Münster, Münster, Germany
| | - Luisa Klotz
- Department of Neurology with Institute of Translational Neurology, University Hospital Münster, University of Münster, Münster, Germany
| | - Nicholas Schwab
- Department of Neurology with Institute of Translational Neurology, University Hospital Münster, University of Münster, Münster, Germany
| | - Heinz Wiendl
- Department of Neurology with Institute of Translational Neurology, University Hospital Münster, University of Münster, Münster, Germany.
| |
Collapse
|
37
|
B cells in autoimmune and neurodegenerative central nervous system diseases. Nat Rev Neurosci 2019; 20:728-745. [PMID: 31712781 DOI: 10.1038/s41583-019-0233-2] [Citation(s) in RCA: 223] [Impact Index Per Article: 37.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/30/2019] [Indexed: 12/16/2022]
|
38
|
Emerging role of innate B1 cells in the pathophysiology of autoimmune and neuroimmune diseases: Association with inflammation, oxidative and nitrosative stress and autoimmune responses. Pharmacol Res 2019; 148:104408. [PMID: 31454534 DOI: 10.1016/j.phrs.2019.104408] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/16/2019] [Revised: 08/20/2019] [Accepted: 08/21/2019] [Indexed: 12/16/2022]
|
39
|
New Ways of "Seeing" the Mechanistic Heterogeneity of Multiple Sclerosis Plaque Pathogenesis. J Neuroophthalmol 2019; 38:91-100. [PMID: 29438266 DOI: 10.1097/wno.0000000000000633] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
BACKGROUND Over the past few decades, we have witnessed a transformation with respect to the principles and pathobiological underpinnings of multiple sclerosis (MS). From the traditional rubric of MS as an inflammatory and demyelinating disorder restricted to central nervous system (CNS) white matter, our contemporary view has evolved to encompass a broader understanding of the variable mechanisms that contribute to tissue injury, in a disorder now recognized to affect white and grey matter compartments. EVIDENCE ACQUISITION A constellation of inflammation, ion channel derangements, bioenergetic supply: demand mismatches within the intra-axonal compartment, and alterations in the dynamics and oximetry of blood flow in CNS tissue compartments are observed in MS. These findings have raised questions regarding how histopathologic heterogeneity may influence the diverse clinical spectrum of MS; and, accordingly, how individual treatment needs vary from 1 patient to the next. RESULTS We are now on new scaffolding in MS; one that promises to translate key clinical and laboratory observations to the application of emerging patient-centered therapies. CONCLUSIONS This review highlights our current knowledge of the underlying disease mechanisms in MS, explores the inflammatory and neurodegenerative consequences of tissue damage, and examines physiologic factors that contribute to bioenergetic homeostasis within the CNS of affected patients.
Collapse
|
40
|
Baker D, Pryce G, Amor S, Giovannoni G, Schmierer K. Learning from other autoimmunities to understand targeting of B cells to control multiple sclerosis. Brain 2019; 141:2834-2847. [PMID: 30212896 DOI: 10.1093/brain/awy239] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2018] [Accepted: 08/01/2018] [Indexed: 12/15/2022] Open
Abstract
Although many suspected autoimmune diseases are thought to be T cell-mediated, the response to therapy indicates that depletion of B cells consistently inhibits disease activity. In multiple sclerosis, it appears that disease suppression is associated with the long-term reduction of memory B cells, which serves as a biomarker for disease activity in many other CD20+ B cell depletion-sensitive, autoimmune diseases. Following B cell depletion, the rapid repopulation by transitional (immature) and naïve (mature) B cells from the bone marrow masks the marked depletion and slow repopulation of lymphoid tissue-derived, memory B cells. This can provide long-term protection from a short treatment cycle. It seems that memory B cells, possibly via T cell stimulation, drive relapsing disease. However, their sequestration in ectopic follicles and the chronic activity of B cells and plasma cells in the central nervous system may drive progressive neurodegeneration directly via antigen-specific mechanisms or indirectly via glial-dependent mechanisms. While unproven, Epstein-Barr virus may be an aetiological trigger of multiple sclerosis. This infects mature B cells, drives the production of memory B cells and possibly provides co-stimulatory signals promoting T cell-independent activation that breaks immune tolerance to generate autoreactivity. Thus, a memory B cell centric mechanism can integrate: potential aetiology, genetics, pathology and response to therapy in multiple sclerosis and other autoimmune conditions with ectopic B cell activation that are responsive to memory B cell-depleting strategies.
Collapse
Affiliation(s)
- David Baker
- BartsMS, Blizard Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Gareth Pryce
- BartsMS, Blizard Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Sandra Amor
- BartsMS, Blizard Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, UK.,Pathology Department, Free University Amsterdam, 1081 HV Amsterdam, The Netherlands
| | - Gavin Giovannoni
- BartsMS, Blizard Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, UK.,Clinical Board Medicine (Neuroscience), The Royal London Hospital, Barts Health NHS Trust, London, UK
| | - Klaus Schmierer
- BartsMS, Blizard Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, UK.,Clinical Board Medicine (Neuroscience), The Royal London Hospital, Barts Health NHS Trust, London, UK
| |
Collapse
|
41
|
Arneth BM. Impact of B cells to the pathophysiology of multiple sclerosis. J Neuroinflammation 2019; 16:128. [PMID: 31238945 PMCID: PMC6593488 DOI: 10.1186/s12974-019-1517-1] [Citation(s) in RCA: 62] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2019] [Accepted: 06/11/2019] [Indexed: 02/07/2023] Open
Abstract
Introduction Multiple sclerosis (MS) is a chronic autoimmune disorder that affects the central nervous system and compromises the health and well-being of millions of people worldwide. B cells have been linked to MS and its progression. This review aimed to determine the role of B cells in MS development. Methods Articles used in this review were obtained from PubMed, LILACS, and EBSCO. The search terms and phrases included “multiple sclerosis,” “MS,” “B-Cells,” “pathogenesis,” and “development.” Original research studies and articles on MS and B cells published between 2007 and 2018 were included. Results Results from the selected articles showed a significant connection between B cell groups and MS. B cells act as a significant source of plasma cells, which generate antibodies while also regulating autoimmune processes and T cell production. In addition, B cells regulate the release of molecules that affect the proinflammatory actions of other immune cells. Discussion B cells play key roles in immune system functioning and MS. The findings of this review illustrate the complex nature of B cell actions, their effects on the autoimmune system, and the method by which they contribute to MS pathogenesis. Conclusion Previous research implicates biological, genetic, and environmental factors in MS pathogenesis. This review suggests that B cells contribute to MS development and advancement by influencing and regulating autoimmune processes such as T cell production and APC activity.
Collapse
Affiliation(s)
- Borros M Arneth
- Institute of Laboratory Medicine and Pathobiochemistry, Molecular Diagnostics, University Hospital of the Universities of Giessen and Marburg UKGM, Justus Liebig University Giessen, Feulgenstr. 12, 35392, Giessen, Germany.
| |
Collapse
|
42
|
Tomescu-Baciu A, Johansen JN, Holmøy T, Greiff V, Stensland M, de Souza GA, Vartdal F, Lossius A. Persistence of intrathecal oligoclonal B cells and IgG in multiple sclerosis. J Neuroimmunol 2019; 333:576966. [PMID: 31153015 DOI: 10.1016/j.jneuroim.2019.576966] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2019] [Revised: 05/14/2019] [Accepted: 05/14/2019] [Indexed: 11/18/2022]
Abstract
In multiple sclerosis (MS), B cells are trafficking across the blood-brain barrier, but it is not known how this relates to the synthesis of oligoclonal IgG. We used quantitative mass spectrometry of oligoclonal bands and high-throughput sequencing of immunoglobulin heavy-chain variable transcripts to study the longitudinal B cell response in the cerebrospinal fluid (CSF) and blood of two MS patients. Twenty of 22 (91%) and 25 of 28 (89%) of oligoclonal band peptides persisted in samples collected 18 months apart, in spite of a dynamic exchange across the blood-CSF barrier of B lineage cells connecting to oligoclonal IgG.
Collapse
Affiliation(s)
- Alina Tomescu-Baciu
- Department of Immunology and Transfusion Medicine, Faculty of Medicine, University of Oslo and Oslo University Hospital Rikshospitalet, Oslo, Norway
| | - Jorunn N Johansen
- Department of Immunology and Transfusion Medicine, Faculty of Medicine, University of Oslo and Oslo University Hospital Rikshospitalet, Oslo, Norway
| | - Trygve Holmøy
- Department of Neurology, Akershus University Hospital, Lørenskog, Norway; Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Victor Greiff
- Department of Immunology and Transfusion Medicine, Faculty of Medicine, University of Oslo and Oslo University Hospital Rikshospitalet, Oslo, Norway; Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Maria Stensland
- Department of Immunology and Transfusion Medicine, Faculty of Medicine, University of Oslo and Oslo University Hospital Rikshospitalet, Oslo, Norway; Proteomics Core Facility, Oslo University Hospital Rikshospitalet, NO-0372 Oslo, Norway
| | - Gustavo Antonio de Souza
- Department of Immunology and Transfusion Medicine, Faculty of Medicine, University of Oslo and Oslo University Hospital Rikshospitalet, Oslo, Norway; Proteomics Core Facility, Oslo University Hospital Rikshospitalet, NO-0372 Oslo, Norway
| | - Frode Vartdal
- Department of Immunology and Transfusion Medicine, Faculty of Medicine, University of Oslo and Oslo University Hospital Rikshospitalet, Oslo, Norway
| | - Andreas Lossius
- Department of Immunology and Transfusion Medicine, Faculty of Medicine, University of Oslo and Oslo University Hospital Rikshospitalet, Oslo, Norway; Department of Neurology, Akershus University Hospital, Lørenskog, Norway.
| |
Collapse
|
43
|
Senel M, Mojib-Yezdani F, Braisch U, Bachhuber F, Lewerenz J, Ludolph AC, Otto M, Tumani H. CSF Free Light Chains as a Marker of Intrathecal Immunoglobulin Synthesis in Multiple Sclerosis: A Blood-CSF Barrier Related Evaluation in a Large Cohort. Front Immunol 2019; 10:641. [PMID: 30984199 PMCID: PMC6449445 DOI: 10.3389/fimmu.2019.00641] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2018] [Accepted: 03/08/2019] [Indexed: 01/22/2023] Open
Abstract
Objectives: The importance of immunoglobulin G (IgG) oligoclonal bands (OCB) in the diagnosis of multiple sclerosis (MS) was reaffirmed again in the recently revised MS diagnostic criteria. Since OCB testing is based on non-quantitative techniques and demands considerable methodological experience, measurement of CSF immunoglobulin free light chains (FLC) has been suggested as quantitative alternative to OCB. We aimed to establish reference values for FLC measures and evaluate their diagnostic accuracy with regard to the diagnosis of MS. Methods: Immunoglobulin kappa (KFLC) and lambda (LFLC) free light chains were prospectively measured by nephelometry in CSF and serum sample pairs in 1,224 patients. The analyzed cohort included patients with MS, other autoimmune or infectious inflammatory diseases of the nervous system as well as 989 patients without signs for nervous system inflammation. Results: Regarding diagnosis of MS, the diagnostic sensitivity and specificity of intrathecal KFLC ratio were 93.3 and 93.7% using the CSF-serum albumin ratio-dependent reference values, 92.0 and 95.9% for intrathecal KFLC ratio applying the ROC-curve determined cut-off levels, 62.7 and 98.3% for IgG index, 64.0 and 98.8% for intrathecal IgG synthesis according to Reiber diagrams, and 94.7 and 93.3% for OCB. Diagnostic sensitivity and specificity of intrathecal LFLC were clearly lower than KFLC. Conclusions: Intrathecal KFLC and OCB showed the highest diagnostic sensitivities for MS. However, specificity was slightly lower compared to other quantitative IgG parameters. Consequently, CSF FLC may not replace OCB, but it may support diagnosis in MS as a quantitative parameter.
Collapse
Affiliation(s)
- Makbule Senel
- Department of Neurology, University of Ulm, Ulm, Germany
| | | | - Ulrike Braisch
- Institute of Epidemiology and Medical Biometry, Ulm University, Ulm, Germany
| | | | - Jan Lewerenz
- Department of Neurology, University of Ulm, Ulm, Germany
| | | | - Markus Otto
- Department of Neurology, University of Ulm, Ulm, Germany
| | - Hayrettin Tumani
- Department of Neurology, University of Ulm, Ulm, Germany.,Specialty Hospital of Neurology Dietenbronn, Schwendi, Germany
| |
Collapse
|
44
|
Häusser-Kinzel S, Weber MS. The Role of B Cells and Antibodies in Multiple Sclerosis, Neuromyelitis Optica, and Related Disorders. Front Immunol 2019; 10:201. [PMID: 30800132 PMCID: PMC6375838 DOI: 10.3389/fimmu.2019.00201] [Citation(s) in RCA: 96] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2018] [Accepted: 01/23/2019] [Indexed: 12/31/2022] Open
Abstract
Our pathophysiological concept of the most common central nervous system demyelinating disease, multiple sclerosis, strikingly evolved by recent discoveries suggesting that B lymphocytes substantially contribute in its initiation and chronic propagation. In this regard, activated B cells are nowadays considered to act as important antigen-presenting cells for the activation of T cells and as essential source of pro-inflammatory cytokines. Hereby, they create a milieu in which other immune cells differentiate and join an orchestrated inflammatory infiltration of the CNS. Without a doubt, this scientific leap was critically pioneered by the empirical use of anti-CD20 antibodies in recent clinical MS trials, which revealed that the therapeutic removal of immature and mature B cells basically halted development of new inflammatory flares in otherwise relapsing MS patients. This stabilization occurred largely independent of any indirect effect on plasma cell-produced antibody levels. On the contrary, peripherally produced autoantibodies are probably the most important B cell component in two other CNS demyelinating diseases which are currently in the process of being delineated as separate disease entities. The first one is neuromyelitis optica in which an antibody response against aquaporin-4 targets and destroys astrocytes, the second, likely distinct entity embraces a group of patients containing antibodies against myelin oligodendrocyte glycoprotein. In this review, we will describe and summarize pro-inflammatory B cell properties in these three CNS demyelinating disorders; we will however also provide an overview on the emerging concept that B cells or B cell subsets may exert immunologically counterbalancing properties, which may be therapeutically desirable to maintain and foster in inflammatory CNS demyelination. In an outlook, we will discuss accordingly, how this potentially important aspect can be harnessed to advance future B cell-directed therapeutic approaches in multiple sclerosis and related diseases.
Collapse
Affiliation(s)
| | - Martin S Weber
- Institute of Neuropathology, University Medical Center, Göttingen, Germany.,Department of Neurology, University Medical Center, Göttingen, Germany
| |
Collapse
|
45
|
Negron A, Robinson RR, Stüve O, Forsthuber TG. The role of B cells in multiple sclerosis: Current and future therapies. Cell Immunol 2018; 339:10-23. [PMID: 31130183 DOI: 10.1016/j.cellimm.2018.10.006] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2018] [Revised: 10/19/2018] [Accepted: 10/20/2018] [Indexed: 02/07/2023]
Abstract
While it was long held that T cells were the primary mediators of multiple sclerosis (MS) pathogenesis, the beneficial effects observed in response to treatment with Rituximab (RTX), a monoclonal antibody (mAb) targeting CD20, shed light on a key contributor to MS that had been previously underappreciated: B cells. This has been reaffirmed by results from clinical trials testing the efficacy of subsequently developed B cell-depleting mAbs targeting CD20 as well as studies revisiting the effects of previous disease-modifying therapies (DMTs) on B cell subsets thought to modulate disease severity. In this review, we summarize current knowledge regarding the complex roles of B cells in MS pathogenesis and current and potential future B cell-directed therapies.
Collapse
Affiliation(s)
- Austin Negron
- Department of Biology, University of Texas at San Antonio, TX 78249, USA
| | - Rachel R Robinson
- Department of Biology, University of Texas at San Antonio, TX 78249, USA
| | - Olaf Stüve
- Department of Neurology and Neurotherapeutics, University of Texas Southwestern Medical Center, Dallas, TX, USA; Neurology Section, VA North Texas Health Care System, Medical Service, Dallas, TX, USA
| | | |
Collapse
|
46
|
Huss A, Abdelhak A, Halbgebauer S, Mayer B, Senel M, Otto M, Tumani H. Intrathecal immunoglobulin M production: A promising high-risk marker in clinically isolated syndrome patients. Ann Neurol 2018; 83:1032-1036. [DOI: 10.1002/ana.25237] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2017] [Revised: 03/06/2018] [Accepted: 04/07/2018] [Indexed: 01/31/2023]
Affiliation(s)
- André Huss
- Experimental Neurology; University Hospital Ulm; Ulm
| | | | | | - Benjamin Mayer
- Institute of Epidemiology and Medical Biometry; Ulm University; Ulm
| | - Makbule Senel
- Department of Neurology; University Hospital Ulm; Ulm
| | - Markus Otto
- Department of Neurology; University Hospital Ulm; Ulm
| | - Hayrettin Tumani
- Department of Neurology; University Hospital Ulm; Ulm
- Specialty Hospital of Neurology Dietenbronn; Schwendi Germany
| |
Collapse
|
47
|
Bashford-Rogers RJM, Smith KGC, Thomas DC. Antibody repertoire analysis in polygenic autoimmune diseases. Immunology 2018; 155:3-17. [PMID: 29574826 PMCID: PMC6099162 DOI: 10.1111/imm.12927] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2018] [Revised: 03/01/2018] [Accepted: 03/06/2018] [Indexed: 12/18/2022] Open
Abstract
High-throughput sequencing of the DNA/RNA encoding antibody heavy- and light-chains is rapidly transforming the field of adaptive immunity. It can address key questions, including: (i) how the B-cell repertoire differs in health and disease; and (ii) if it does differ, the point(s) in B-cell development at which this occurs. The advent of technologies, such as whole-genome sequencing, offers the chance to link abnormalities in the B-cell antibody repertoire to specific genomic variants and polymorphisms. Here, we discuss the current research using B-cell antibody repertoire sequencing in three polygenic autoimmune diseases where there is good evidence for a pathological role for B-cells, namely systemic lupus erythematosus, multiple sclerosis and rheumatoid arthritis. These autoimmune diseases exhibit significantly skewed B-cell receptor repertoires compared with healthy controls. Interestingly, some common repertoire defects are shared between diseases, such as elevated IGHV4-34 gene usage. B-cell clones have effectively been characterized and tracked between different tissues and blood in autoimmune disease. It has been hypothesized that these differences may signify differences in B-cell tolerance; however, the mechanisms and implications of these defects are not clear.
Collapse
Affiliation(s)
| | | | - David C Thomas
- Department of Medicine, University of Cambridge, Cambridge, UK
| |
Collapse
|
48
|
Hohlfeld R. B-cells as therapeutic targets in neuro-inflammatory diseases. Clin Immunol 2018; 186:51-53. [DOI: 10.1016/j.clim.2017.07.013] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2017] [Accepted: 07/19/2017] [Indexed: 10/19/2022]
|
49
|
Höftberger R, Lassmann H. Inflammatory demyelinating diseases of the central nervous system. HANDBOOK OF CLINICAL NEUROLOGY 2018; 145:263-283. [PMID: 28987175 PMCID: PMC7149979 DOI: 10.1016/b978-0-12-802395-2.00019-5] [Citation(s) in RCA: 112] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Inflammatory demyelinating diseases are a heterogeneous group of disorders, which occur against the background of an acute or chronic inflammatory process. The pathologic hallmark of multiple sclerosis (MS) is the presence of focal demyelinated lesions with partial axonal preservation and reactive astrogliosis. Demyelinated plaques are present in the white as well as gray matter, such as the cerebral or cerebellar cortex and brainstem nuclei. Activity of the disease process is reflected by the presence of lesions with ongoing myelin destruction. Axonal and neuronal destruction in the lesions is a major substrate for permanent neurologic deficit in MS patients. The MS pathology is qualitatively similar in different disease stages, such as relapsing remitting MS or secondary or primary progressive MS, but the prevalence of different lesion types differs quantitatively. Acute MS and Balo's type of concentric sclerosis appear to be variants of classic MS. In contrast, neuromyelitis optica (NMO) and spectrum disorders (NMOSD) are inflammatory diseases with primary injury of astrocytes, mediated by aquaporin-4 antibodies. Finally, we discuss the histopathology of other inflammatory demyelinating diseases such as acute disseminated encephalomyelitis and myelin oligodendrocyte glycoprotein antibody-associated demyelination. Knowledge of the heterogenous immunopathology in demyelinating diseases is important, to understand the clinical presentation and disease course and to find the optimal treatment for an individual patient.
Collapse
Affiliation(s)
- Romana Höftberger
- Institute of Neurology, Medical University of Vienna, Vienna, Austria
| | - Hans Lassmann
- Center for Brain Research, Medical University of Vienna, Vienna, Austria,Correspondence to: Hans Lassmann, MD, Center for Brain Research, Medical University of Vienna, Spitalgasse, 1090 Vienna, Austria
| |
Collapse
|
50
|
Eggers EL, Michel BA, Wu H, Wang SZ, Bevan CJ, Abounasr A, Pierson NS, Bischof A, Kazer M, Leitner E, Greenfield AL, Demuth S, Wilson MR, Henry RG, Cree BA, Hauser SL, von Büdingen HC. Clonal relationships of CSF B cells in treatment-naive multiple sclerosis patients. JCI Insight 2017; 2:92724. [PMID: 29202449 DOI: 10.1172/jci.insight.92724] [Citation(s) in RCA: 70] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2017] [Accepted: 10/17/2017] [Indexed: 12/29/2022] Open
Abstract
A role of B cells in multiple sclerosis (MS) is well established, but there is limited understanding of their involvement during active disease. Here, we examined cerebrospinal fluid (CSF) and peripheral blood (PB) B cells in treatment-naive patients with MS or high-risk clinically isolated syndrome. Using flow cytometry, we found increased CSF lymphocytes with a disproportionate increase of B cells compared with T cells in patients with gadolinium-enhancing (Gd+) lesions on brain MRI. Ig gene heavy chain variable region (Ig-VH) repertoire sequencing of CSF and PB B cells revealed clonal relationships between intrathecal and peripheral B cell populations, which could be consistent with migration of B cells to and activation in the CNS in active MS. In addition, we found evidence for bystander immigration of B cells from the periphery, which could be supported by a CXCL13 gradient between CSF and blood. Understanding what triggers B cells to migrate and home to the CNS may ultimately aid in the rational selection of therapeutic strategies to limit progression in MS.
Collapse
|