1
|
Fischbach F, Richter J, Pfeffer LK, Fehse B, Berger SC, Reinhardt S, Kuhle J, Badbaran A, Rathje K, Gagelmann N, Borie D, Seibel J, Ayuk F, Friese MA, Heesen C, Kröger N. CD19-targeted chimeric antigen receptor T cell therapy in two patients with multiple sclerosis. MED 2024; 5:550-558.e2. [PMID: 38554710 DOI: 10.1016/j.medj.2024.03.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2024] [Revised: 02/12/2024] [Accepted: 03/06/2024] [Indexed: 04/02/2024]
Abstract
BACKGROUND Progressive multiple sclerosis (MS) is characterized by compartmentalized smoldering neuroinflammation caused by the proliferation of immune cells residing in the central nervous system (CNS), including B cells. Although inflammatory activity can be prevented by immunomodulatory therapies during early disease, such therapies typically fail to halt disease progression. CD19 chimeric antigen receptor (CAR)-T cell therapies have revolutionized the field of hematologic malignancies. Although generally considered efficacious, serious adverse events associated with CAR-T cell therapies such as immune effector cell-associated neurotoxicity syndrome (ICANS) have been observed. Successful use of CD19 CAR-T cells in rheumatic diseases like systemic lupus erythematosus and neuroimmunological diseases like myasthenia gravis have recently been observed, suggesting possible application in other autoimmune diseases. METHODS Here, we report the first individual treatment with a fully human CD19 CAR-T cell therapy (KYV-101) in two patients with progressive MS. FINDINGS CD19 CAR-T cell administration resulted in acceptable safety profiles for both patients. No ICANS was observed despite detection of CD19 CAR-T cells in the cerebrospinal fluid. In case 1, intrathecal antibody production in the cerebrospinal fluid decreased notably after CAR-T cell infusion and was sustained through day 64. CONCLUSIONS CD19 CAR-T cell administration in progressive MS resulted in an acceptable safety profile. CAR-T cell presence and expansion were observed in the cerebrospinal fluid without clinical signs of neurotoxicity, which, along with intrathecal antibody reduction, indicates expansion-dependent effects of CAR-T cells on CD19+ target cells in the CNS. Larger clinical studies assessing CD19 CAR-T cells in MS are warranted. FUNDING Both individual treatments as well the generated data were not based on external funding.
Collapse
Affiliation(s)
- Felix Fischbach
- Institute of Neuroimmunology and Multiple Sclerosis and Department of Neurology, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Johanna Richter
- Department for Stem Cell Transplantation, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Lena Kristina Pfeffer
- Institute of Neuroimmunology and Multiple Sclerosis and Department of Neurology, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Boris Fehse
- Department for Stem Cell Transplantation, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Susanna Carolina Berger
- Department for Stem Cell Transplantation, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Stefanie Reinhardt
- Institute of Neuroimmunology and Multiple Sclerosis and Department of Neurology, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Jens Kuhle
- Multiple Sclerosis Center and Research Center for Clinical Neuroimmunology and Neuroscience Basel (RC2NB), Departments of Head, Spine and Neuromedicine, Biomedicine and Clinical Research, University Hospital Basel and University of Basel, 4031 Basel, Switzerland
| | - Anita Badbaran
- Department for Stem Cell Transplantation, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Kristin Rathje
- Department for Stem Cell Transplantation, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Nico Gagelmann
- Department for Stem Cell Transplantation, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | | | - Johan Seibel
- Institute for Transfusion Medicine, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Francis Ayuk
- Department for Stem Cell Transplantation, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Manuel A Friese
- Institute of Neuroimmunology and Multiple Sclerosis and Department of Neurology, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Christoph Heesen
- Institute of Neuroimmunology and Multiple Sclerosis and Department of Neurology, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany.
| | - Nicolaus Kröger
- Department for Stem Cell Transplantation, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany.
| |
Collapse
|
2
|
Tolkovsky A, Pardo K, Hellmann M, Lotan I, Auriel E, Wilf-Yarkoni A. Association between clinical characteristics, acute steroid treatment and oligoclonal bands result in multiple sclerosis: A retrospective study. Mult Scler Relat Disord 2024; 85:105554. [PMID: 38537510 DOI: 10.1016/j.msard.2024.105554] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2023] [Revised: 03/11/2024] [Accepted: 03/15/2024] [Indexed: 05/07/2024]
Abstract
BACKGROUND Detection of oligoclonal bands (OCBs) in the cerebrospinal fluid (CSF) is important for diagnosis of multiple sclerosis (MS). Previous studies reported that treatment with intravenous methylprednisolone (IVMP) before lumber puncture (LP) could suppress OCBs production. The aim of this study was to assess whether IVMP initiation prior to CSF collection affects OCBs results in patients with an acute demyelinating event. Additionally, we examined which clinical characteristics are associated with the presence of OCBs in the CSF. METHODS We retrospectively evaluated patients admitted to the neurology department at rabin medical center (RMC) between 2010 and 2022 who underwent LP with OCBs analysis as part of their demyelinating attack workup. Patients were divided into OCB-positive and OCB-negative groups and demographical and clinical characteristics (including timing and duration of acute steroid treatment and history of prior demyelinating attacks) were analyzed for association with OCBs results. RESULTS A total of 342 patients were included with a median age of 35 years (IQR, 27-46). Two hundred thirty-eight (69.6 %) were OCB-positive. Initiation of IVMP before LP was not associated with negative OCBs (11.8 % Vs. 13.5 %, P = 0.721), nor was it correlated with OCBs positivity (OR=0.86, P = 0.66). CSF cell count was higher in OCB-positive patients (5 Vs. 3, P = 0.001), and a history of prior demyelinating attacks was associated with- (33.6 % Vs. 20.2 %, P = 0.014) and predictive of OCBs positivity (OR=2, P = 0.013). CONCLUSIONS Timing of steroids was not associated with OCB positivity. However, pleocytosis and a prior attack were associated with OCB positivity in this cohort. Our results suggest that steroid treatment is unlikely to affect OCBs results. Ideally, larger prospective studies would be needed to confirm our observations.
Collapse
Affiliation(s)
- Assaf Tolkovsky
- Departmet of Neurology, Rabin Medical Center, Petah Tikva, Israel; Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel.
| | - Keshet Pardo
- Departmet of Neurology, Rabin Medical Center, Petah Tikva, Israel; Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Mark Hellmann
- Departmet of Neurology, Rabin Medical Center, Petah Tikva, Israel; Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Itay Lotan
- Departmet of Neurology, Rabin Medical Center, Petah Tikva, Israel; Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Eitan Auriel
- Departmet of Neurology, Rabin Medical Center, Petah Tikva, Israel; Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Adi Wilf-Yarkoni
- Departmet of Neurology, Rabin Medical Center, Petah Tikva, Israel; Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| |
Collapse
|
3
|
El Mahdaoui S, Hansen MM, von Essen MR, Hvalkof VH, Holm Hansen R, Mahler MR, Jennum P, Sellebjerg F, Romme Christensen J. CD11c + B cells in relapsing-remitting multiple sclerosis and effects of anti-CD20 therapy. Ann Clin Transl Neurol 2024; 11:926-937. [PMID: 38332555 PMCID: PMC11021659 DOI: 10.1002/acn3.52009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2023] [Revised: 01/10/2024] [Accepted: 01/12/2024] [Indexed: 02/10/2024] Open
Abstract
OBJECTIVES B cells are important in the pathogenesis of multiple sclerosis. It is yet unknown which subsets may be involved, but atypical B cells have been proposed as mediators of autoimmunity. In this study, we investigated differences in B-cell subsets between controls and patients with untreated and anti-CD20-treated multiple sclerosis. METHODS We recruited 155 participants for an exploratory cohort comprising peripheral blood and cerebrospinal fluid, and a validation cohort comprising peripheral blood. Flow cytometry was used to characterize B-cell phenotypes and effector functions of CD11c+ atypical B cells. RESULTS There were no differences in circulating B cells between controls and untreated multiple sclerosis. As expected, anti-CD20-treated patients had a markedly lower B-cell count. Of B cells remaining after treatment, we observed higher proportions of CD11c+ B cells and plasmablasts. CD11c+ B cells were expanded in cerebrospinal fluid compared to peripheral blood in controls and untreated multiple sclerosis. Surprisingly, the proportion of CD11c+ cerebrospinal fluid B cells was higher in controls and after anti-CD20 therapy than in untreated multiple sclerosis. Apart from the presence of plasmablasts, the cerebrospinal fluid B-cell composition after anti-CD20 therapy resembled that of controls. CD11c+ B cells demonstrated a high potential for both proinflammatory and regulatory cytokine production. INTERPRETATION The study demonstrates that CD11c+ B cells and plasmablasts are less efficiently depleted by anti-CD20 therapy, and that CD11c+ B cells comprise a phenotypically and functionally distinct, albeit heterogenous, B-cell subset with the capacity of exerting both proinflammatory and regulatory functions.
Collapse
Affiliation(s)
- Sahla El Mahdaoui
- Department of NeurologyDanish Multiple Sclerosis Center, Copenhagen University Hospital – RigshospitaletGlostrup2600Denmark
| | - Marie Mathilde Hansen
- Department of NeurologyDanish Multiple Sclerosis Center, Copenhagen University Hospital – RigshospitaletGlostrup2600Denmark
| | - Marina Rode von Essen
- Department of NeurologyDanish Multiple Sclerosis Center, Copenhagen University Hospital – RigshospitaletGlostrup2600Denmark
| | - Victoria Hyslop Hvalkof
- Department of NeurologyDanish Multiple Sclerosis Center, Copenhagen University Hospital – RigshospitaletGlostrup2600Denmark
| | - Rikke Holm Hansen
- Department of NeurologyDanish Multiple Sclerosis Center, Copenhagen University Hospital – RigshospitaletGlostrup2600Denmark
| | - Mie Reith Mahler
- Department of NeurologyDanish Multiple Sclerosis Center, Copenhagen University Hospital – RigshospitaletGlostrup2600Denmark
| | - Poul Jennum
- Department of NeurologyDanish Center for Sleep Medicine, Copenhagen University Hospital – RigshospitaletGlostrup2600Denmark
- Department of Clinical MedicineUniversity of CopenhagenCopenhagen2200Denmark
| | - Finn Sellebjerg
- Department of NeurologyDanish Multiple Sclerosis Center, Copenhagen University Hospital – RigshospitaletGlostrup2600Denmark
- Department of Clinical MedicineUniversity of CopenhagenCopenhagen2200Denmark
| | - Jeppe Romme Christensen
- Department of NeurologyDanish Multiple Sclerosis Center, Copenhagen University Hospital – RigshospitaletGlostrup2600Denmark
| |
Collapse
|
4
|
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
|
5
|
Gottlieb A, Pham HPT, Saltarrelli JG, Lindsey JW. Expanded T lymphocytes in the cerebrospinal fluid of multiple sclerosis patients are specific for Epstein-Barr-virus-infected B cells. Proc Natl Acad Sci U S A 2024; 121:e2315857121. [PMID: 38190525 PMCID: PMC10801919 DOI: 10.1073/pnas.2315857121] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2023] [Accepted: 11/27/2023] [Indexed: 01/10/2024] Open
Abstract
Epstein-Barr virus (EBV) infection has long been associated with multiple sclerosis (MS), but the role of EBV in the pathogenesis of MS is not clear. Our hypothesis is that a major fraction of the expanded clones of T lymphocytes in the cerebrospinal fluid (CSF) are specific for autologous EBV-infected B cells. We obtained blood and CSF samples from eight relapsing-remitting patients in the process of diagnosis. We stimulated cells from the blood with autologous EBV-infected lymphoblastoid cell lines (LCL), EBV, varicella zoster virus, influenza, and candida and sorted the responding cells with flow cytometry after 6 d. We sequenced the RNA for T cell receptors (TCR) from CSF, unselected blood cells, and the antigen-specific cells. We used the TCR Vβ CDR3 sequences from the antigen-specific cells to assign antigen specificity to the sequences from the CSF and blood. LCL-specific cells comprised 13.0 ± 4.3% (mean ± SD) of the total reads present in CSF and 13.3 ± 7.5% of the reads present in blood. The next most abundant antigen specificity was flu, which was 4.7 ± 1.7% of the reads in the CSF and 9.3 ± 6.6% in the blood. The prominence of LCL-specific reads was even more marked in the top 1% most abundant CSF clones with statistically significant 47% mean overlap with LCL. We conclude that LCL-specific sequences form a major portion of the TCR repertoire in both CSF and blood and that expanded clones specific for LCL are present in MS CSF. This has important implications for the pathogenesis of MS.
Collapse
Affiliation(s)
- Assaf Gottlieb
- Center for Precision Health, School of Biomedical Informatics, University of Texas Health Science Center at Houston, Houston, TX77030
| | - H. Phuong T. Pham
- Division of Multiple Sclerosis and Neuroimmunology, Department of Neurology, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX77030
| | - Jerome G. Saltarrelli
- Department of Surgery, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX77030
| | - J. William Lindsey
- Division of Multiple Sclerosis and Neuroimmunology, Department of Neurology, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX77030
| |
Collapse
|
6
|
Asplund Högelin K, Isac B, Khademi M, Al Nimer F. B cell activating factor levels are linked to distinct B cell markers in multiple sclerosis and following B cell depletion and repopulation. Clin Immunol 2024; 258:109870. [PMID: 38101497 DOI: 10.1016/j.clim.2023.109870] [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: 09/12/2023] [Revised: 11/07/2023] [Accepted: 11/17/2023] [Indexed: 12/17/2023]
Abstract
Recent studies have highlighted the important role of B cells in the pathogenesis of multiple sclerosis (MS). B cell activating factor (BAFF) and A proliferation inducing ligand (APRIL) play a major role in B cell survival and homeostasis. Here, we studied the association of BAFF and APRIL with B cell immune markers in MS and following B cell depletion and repopulation. We found that BAFF but not APRIL was significantly higher in plasma in untreated MS compared to controls. BAFF increased after rituximab treatment and decreased again during repopulation displaying an inverse correlation with B cell numbers, and more specifically switched memory B cell numbers. Cerebrospinal fluid BAFF inversely correlated with IgG index. BAFF displayed an inverse association to anti-EBV-CA antibodies. In summary, our study identified immune cells and factors that might regulate or be regulated by BAFF and APRIL levels in MS, and during B cell depletion and repopulation.
Collapse
Affiliation(s)
- Klara Asplund Högelin
- Neuroimmunology Unit, Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden.
| | - Beshoy Isac
- Biomedical Laboratory Science, Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Mohsen Khademi
- Neuroimmunology Unit, Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Faiez Al Nimer
- Neuroimmunology Unit, Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
7
|
Arroyo-Pereiro P, García-Serrano L, Morandeira F, Urban B, Mas V, Framil M, León I, Muñoz-Vendrell A, Matas E, Romero-Pinel L, Martínez-Yélamos A, Martínez-Yélamos S, Bau L. Kappa free light chains index in multiple sclerosis very long-term prognosis. Front Immunol 2023; 14:1223514. [PMID: 37885887 PMCID: PMC10598843 DOI: 10.3389/fimmu.2023.1223514] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2023] [Accepted: 09/11/2023] [Indexed: 10/28/2023] Open
Abstract
Introduction The role of the kappa-free light chain (kFLC) in the diagnosis of multiple sclerosis (MS) and, to a lesser extent, its role as a medium-term prognostic marker have been extensively studied. This study aimed to explore its potential as a long-term prognostic marker for MS. Methods We performed an exploratory retrospective observational study by selecting patients systemically followed up in our MS unit with available cerebrospinal fluid and serum samples at the time of initial evaluation. Two groups were defined: benign MS (bMS), defined as patients with Expanded Disability Status Scale (EDSS) ≤ 3 at 10 years of follow-up, and aggressive MS (aMS), defined as patients with EDSS ≥ 6 at 15 years of follow-up. Clinical variables were collected, and the immunoglobulin G (IgG) index, kFLC index, and oligoclonal bands (OCB) were determined for all patients and compared between the groups. Results Twenty bMS and 15 aMS patients were included in this study. Sixty percent (21/35) were female, and the mean age at the time of the first symptom was 31.5 ± 9.45 years, with no statistical differences between groups. Median follow-up time was 19.8 years (Interquartile range, IQR 15.9-24.6). The median EDSS scores at the last follow-up were 1.5 and 7.5 in the bMS and the aMS group, respectively. No statistically significant differences were found in the kFLC index between the two groups (136.6 vs. 140.27, p=0.59). The IgG index was positive in 62.9% of patients (55% bMS vs. 73.3% aMS, p>0.05), and OCB was positive in 88.6% (90% bMS vs. 86.7% aMS, p>0.05). A significant positive correlation was found between IgG and kFLC indices (rs = 0.85, p<0.001). Conclusion Given the absence of differences between the two groups with opposite disease courses, it is unlikely that the kFLC index is a reliable and powerful marker of long-term prognosis in MS.
Collapse
Affiliation(s)
- Pablo Arroyo-Pereiro
- Multiple Sclerosis Unit, Department of Neurology, Hospital Universitari de Bellvitge- Institut d’Investigació Biomèdica de Bellvitge (IDIBELL), L’Hospitalet de Llobregat, Spain
| | - Lydia García-Serrano
- Department of Immunology, Hospital Universitari de Bellvitge – Institut d’Investigació Biomédica de Bellvitge (IDIBELL), L’Hospitalet de Llobregat, Spain
| | - Francisco Morandeira
- Department of Immunology, Hospital Universitari de Bellvitge – Institut d’Investigació Biomédica de Bellvitge (IDIBELL), L’Hospitalet de Llobregat, Spain
| | - Blanca Urban
- Department of Immunology, Hospital Universitari de Bellvitge – Institut d’Investigació Biomédica de Bellvitge (IDIBELL), L’Hospitalet de Llobregat, Spain
| | - Virginia Mas
- Department of Immunology, Hospital Universitari de Bellvitge – Institut d’Investigació Biomédica de Bellvitge (IDIBELL), L’Hospitalet de Llobregat, Spain
| | - Mario Framil
- Department of Immunology, Hospital Universitari de Bellvitge – Institut d’Investigació Biomédica de Bellvitge (IDIBELL), L’Hospitalet de Llobregat, Spain
| | - Isabel León
- Multiple Sclerosis Unit, Department of Neurology, Hospital Universitari de Bellvitge- Institut d’Investigació Biomèdica de Bellvitge (IDIBELL), L’Hospitalet de Llobregat, Spain
| | - Albert Muñoz-Vendrell
- Multiple Sclerosis Unit, Department of Neurology, Hospital Universitari de Bellvitge- Institut d’Investigació Biomèdica de Bellvitge (IDIBELL), L’Hospitalet de Llobregat, Spain
| | - Elisabet Matas
- Multiple Sclerosis Unit, Department of Neurology, Hospital Universitari de Bellvitge- Institut d’Investigació Biomèdica de Bellvitge (IDIBELL), L’Hospitalet de Llobregat, Spain
| | - Lucía Romero-Pinel
- Multiple Sclerosis Unit, Department of Neurology, Hospital Universitari de Bellvitge- Institut d’Investigació Biomèdica de Bellvitge (IDIBELL), L’Hospitalet de Llobregat, Spain
| | - Antonio Martínez-Yélamos
- Multiple Sclerosis Unit, Department of Neurology, Hospital Universitari de Bellvitge- Institut d’Investigació Biomèdica de Bellvitge (IDIBELL), L’Hospitalet de Llobregat, Spain
- Departament of Clinical Sciences, Facultat de Medicina i Ciències de la Salut, Universitat de Barcelona (UB), Barcelona, Spain
| | - Sergio Martínez-Yélamos
- Multiple Sclerosis Unit, Department of Neurology, Hospital Universitari de Bellvitge- Institut d’Investigació Biomèdica de Bellvitge (IDIBELL), L’Hospitalet de Llobregat, Spain
- Departament of Clinical Sciences, Facultat de Medicina i Ciències de la Salut, Universitat de Barcelona (UB), Barcelona, Spain
| | - Laura Bau
- Multiple Sclerosis Unit, Department of Neurology, Hospital Universitari de Bellvitge- Institut d’Investigació Biomèdica de Bellvitge (IDIBELL), L’Hospitalet de Llobregat, Spain
| |
Collapse
|
8
|
Touil H, Li R, Zuroff L, Moore CS, Healy L, Cignarella F, Piccio L, Ludwin S, Prat A, Gommerman J, Bennett FC, Jacobs D, Benjamins JA, Lisak RP, Antel JP, Bar-Or A. Cross-talk between B cells, microglia and macrophages, and implications to central nervous system compartmentalized inflammation and progressive multiple sclerosis. EBioMedicine 2023; 96:104789. [PMID: 37703640 PMCID: PMC10505984 DOI: 10.1016/j.ebiom.2023.104789] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Revised: 08/07/2023] [Accepted: 08/22/2023] [Indexed: 09/15/2023] Open
Abstract
BACKGROUND B cells can be enriched within meningeal immune-cell aggregates of multiple sclerosis (MS) patients, adjacent to subpial cortical demyelinating lesions now recognized as important contributors to progressive disease. This subpial demyelination is notable for a 'surface-in' gradient of neuronal loss and microglial activation, potentially reflecting the effects of soluble factors secreted into the CSF. We previously demonstrated that MS B-cell secreted products are toxic to oligodendrocytes and neurons. The potential for B-cell-myeloid cell interactions to propagate progressive MS is of considerable interest. METHODS Secreted products of MS-implicated pro-inflammatory effector B cells or IL-10-expressing B cells with regulatory potential were applied to human brain-derived microglia or monocyte-derived macrophages, with subsequent assessment of myeloid phenotype and function through measurement of their expression of pro-inflammatory, anti-inflammatory and homeostatic/quiescent molecules, and phagocytosis (using flow cytometry, ELISA and fluorescently-labeled myelin). Effects of secreted products of differentially activated microglia on B-cell survival and activation were further studied. FINDINGS Secreted products of MS-implicated pro-inflammatory B cells (but not IL-10 expressing B cells) substantially induce pro-inflammatory cytokine (IL-12, IL-6, TNFα) expression by both human microglia and macrophage (in a GM-CSF dependent manner), while down-regulating their expression of IL-10 and of quiescence-associated molecules, and suppressing their myelin phagocytosis. In contrast, secreted products of IL-10 expressing B cells upregulate both human microglia and macrophage expression of quiescence-associated molecules and enhance their myelin phagocytosis. Secreted factors from pro-inflammatory microglia enhance B-cell activation. INTERPRETATION Potential cross-talk between disease-relevant human B-cell subsets and both resident CNS microglia and infiltrating macrophages may propagate CNS-compartmentalized inflammation and injury associated with MS disease progression. These interaction represents an attractive therapeutic target for agents such as Bruton's tyrosine kinase inhibitors (BTKi) that modulate responses of both B cells and myeloid cells. FUNDING Stated in Acknowledgments section of manuscript.
Collapse
Affiliation(s)
- Hanane Touil
- Department of Neurology and Center for Neuroinflammation and Experimental Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Rui Li
- Department of Neurology and Center for Neuroinflammation and Experimental Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Leah Zuroff
- Department of Neurology and Center for Neuroinflammation and Experimental Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Craig S Moore
- Division of BioMedical Sciences, Faculty of Medicine, Memorial University of Newfoundland, St. John's, NL, Canada
| | - Luke Healy
- Neuroimmunology Unit, Montréal Neurological Institute, McGill University, Canada
| | - Francesca Cignarella
- Department of Neurology, Washington University School of Medicine, 660 South Euclid Avenue, St Louis, MO, USA
| | - Laura Piccio
- Charles Perkins Centre and School of Medical Sciences, The University of Sydney, Camperdown, NSW, Australia
| | - Samuel Ludwin
- Department of Pathology and Molecular Medicine, Queen's University, Kingston, ON, K7L 3N6, Canada
| | - Alexandre Prat
- Université de Montréal Centre de Recherche du CHUM (CRCHUM) and Department of Neuroscience, Université de Montréal, 900 Saint Denis Street, Montréal, QC, H2X 0A9, Canada
| | - Jennifer Gommerman
- Department of Immunology, University of Toronto, Toronto, ON, M5S 1A8, Canada
| | - Frederick C Bennett
- Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Dina Jacobs
- Department of Neurology and Center for Neuroinflammation and Experimental Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Joyce A Benjamins
- Departments of Neurology and Biochemistry, Immunology and Microbiology, Wayne State University School of Medicine, Detroit, MI, USA
| | - Robert P Lisak
- Departments of Neurology and Biochemistry, Immunology and Microbiology, Wayne State University School of Medicine, Detroit, MI, USA
| | - Jack P Antel
- Neuroimmunology Unit, Montréal Neurological Institute, McGill University, Canada
| | - Amit Bar-Or
- Department of Neurology and Center for Neuroinflammation and Experimental Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
9
|
Geladaris A, Häusser-Kinzel S, Pretzsch R, Nissimov N, Lehmann-Horn K, Häusler D, Weber MS. IL-10-providing B cells govern pro-inflammatory activity of macrophages and microglia in CNS autoimmunity. Acta Neuropathol 2023; 145:461-477. [PMID: 36854993 PMCID: PMC10020302 DOI: 10.1007/s00401-023-02552-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2022] [Revised: 02/16/2023] [Accepted: 02/16/2023] [Indexed: 03/02/2023]
Abstract
B cells contribute to chronic inflammatory conditions as source of antibody-secreting plasma cells and as antigen-presenting cells activating T cells, making anti-CD20-mediated B cell depletion a widely used therapeutic option. B cells or B cell subsets may, however, exert regulatory effects, while to date, the immunological and/or clinical impact of these observations remained unclear. We found that in multiple sclerosis (MS) patients, B cells contain regulatory features and that their removal enhanced activity of monocytes. Using a co-culture system, we identified B cell-provided interleukin (IL)-10 as key factor in controlling pro-inflammatory activity of peripheral myeloid cells as well as microglia. Depleting B cells via anti-CD20 in a mouse model of MS unleashed the activity of myeloid cells and microglia and accelerated disease severity; in contrast, adoptive transfer of IL-10-providing B cells restored in vivo control of central nervous system (CNS) macrophages and microglia and reversed clinical exacerbation. These findings suggest that B cells exert meaningful regulatory properties, which should be considered when designing novel B cell-directed agents.
Collapse
Affiliation(s)
- Anastasia Geladaris
- Institute of Neuropathology, University Medical Centre Göttingen, Göttingen, Germany
- Fraunhofer-Institute for Translational Medicine and Pharmacology ITMP, Göttingen, Germany
| | - Silke Häusser-Kinzel
- Institute of Neuropathology, University Medical Centre Göttingen, Göttingen, Germany
| | - Roxanne Pretzsch
- Institute of Neuropathology, University Medical Centre Göttingen, Göttingen, Germany
- Department of Neurology, University Medical Centre Göttingen, Göttingen, Germany
| | - Nitzan Nissimov
- Institute of Neuropathology, University Medical Centre Göttingen, Göttingen, Germany
- Department of Neurosurgery, Charité-Universitätsmedizin Berlin Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Klaus Lehmann-Horn
- Department of Neurology, School of Medicine, Technical University of Munich, Munich, Germany
| | - Darius Häusler
- Institute of Neuropathology, University Medical Centre Göttingen, Göttingen, Germany
- Fraunhofer-Institute for Translational Medicine and Pharmacology ITMP, Göttingen, Germany
| | - Martin S Weber
- Institute of Neuropathology, University Medical Centre Göttingen, Göttingen, Germany.
- Fraunhofer-Institute for Translational Medicine and Pharmacology ITMP, Göttingen, Germany.
- Department of Neurology, University Medical Centre Göttingen, Göttingen, Germany.
| |
Collapse
|
10
|
Bogers L, Engelenburg HJ, Janssen M, Unger PPA, Melief MJ, Wierenga-Wolf AF, Hsiao CC, Mason MRJ, Hamann J, van Langelaar J, Smolders J, van Luijn MM. Selective emergence of antibody-secreting cells in the multiple sclerosis brain. EBioMedicine 2023; 89:104465. [PMID: 36796230 PMCID: PMC9958261 DOI: 10.1016/j.ebiom.2023.104465] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Revised: 01/17/2023] [Accepted: 01/23/2023] [Indexed: 02/16/2023] Open
Abstract
BACKGROUND Although distinct brain-homing B cells have been identified in multiple sclerosis (MS), it is unknown how these further evolve to contribute to local pathology. We explored B-cell maturation in the central nervous system (CNS) of MS patients and determined their association with immunoglobulin (Ig) production, T-cell presence, and lesion formation. METHODS Ex vivo flow cytometry was performed on post-mortem blood, cerebrospinal fluid (CSF), meninges and white matter from 28 MS and 10 control brain donors to characterize B cells and antibody-secreting cells (ASCs). MS brain tissue sections were analysed with immunostainings and microarrays. IgG index and CSF oligoclonal bands were measured with nephelometry, isoelectric focusing, and immunoblotting. Blood-derived B cells were cocultured under T follicular helper-like conditions to evaluate their ASC-differentiating capacity in vitro. FINDINGS ASC versus B-cell ratios were increased in post-mortem CNS compartments of MS but not control donors. Local presence of ASCs associated with a mature CD45low phenotype, focal MS lesional activity, lesional Ig gene expression, and CSF IgG levels as well as clonality. In vitro B-cell maturation into ASCs did not differ between MS and control donors. Notably, lesional CD4+ memory T cells positively correlated with ASC presence, reflected by local interplay with T cells. INTERPRETATION These findings provide evidence that local B cells at least in late-stage MS preferentially mature into ASCs, which are largely responsible for intrathecal and local Ig production. This is especially seen in active MS white matter lesions and likely depends on the interaction with CD4+ memory T cells. FUNDING Stichting MS Research (19-1057 MS; 20-490f MS), National MS Fonds (OZ2018-003).
Collapse
Affiliation(s)
- Laurens Bogers
- Department of Immunology, MS Center ErasMS, Erasmus MC, University Medical Center Rotterdam, 3015 CN, Rotterdam, The Netherlands
| | - Hendrik J Engelenburg
- Neuroimmunology Research Group, Netherlands Institute for Neuroscience, 1105 BA, Amsterdam, The Netherlands
| | - Malou Janssen
- Department of Immunology, MS Center ErasMS, Erasmus MC, University Medical Center Rotterdam, 3015 CN, Rotterdam, The Netherlands; Department of Neurology, MS Center ErasMS, Erasmus MC, University Medical Center Rotterdam, 3015 CN, Rotterdam, The Netherlands
| | - Peter-Paul A Unger
- Department of Viroscience, Erasmus MC, University Medical Center Rotterdam, 3015 CN, Rotterdam, The Netherlands
| | - Marie-José Melief
- Department of Immunology, MS Center ErasMS, Erasmus MC, University Medical Center Rotterdam, 3015 CN, Rotterdam, The Netherlands
| | - Annet F Wierenga-Wolf
- Department of Immunology, MS Center ErasMS, Erasmus MC, University Medical Center Rotterdam, 3015 CN, Rotterdam, The Netherlands
| | - Cheng-Chih Hsiao
- Neuroimmunology Research Group, Netherlands Institute for Neuroscience, 1105 BA, Amsterdam, The Netherlands
| | - Matthew R J Mason
- Neuroimmunology Research Group, Netherlands Institute for Neuroscience, 1105 BA, Amsterdam, The Netherlands
| | - Jörg Hamann
- Neuroimmunology Research Group, Netherlands Institute for Neuroscience, 1105 BA, Amsterdam, The Netherlands; Department of Experimental Immunology, Amsterdam Institute for Infection and Immunity, Amsterdam University Medical Centers, 1007 MB, Amsterdam, The Netherlands
| | - Jamie van Langelaar
- Department of Immunology, MS Center ErasMS, Erasmus MC, University Medical Center Rotterdam, 3015 CN, Rotterdam, The Netherlands
| | - Joost Smolders
- Department of Immunology, MS Center ErasMS, Erasmus MC, University Medical Center Rotterdam, 3015 CN, Rotterdam, The Netherlands; Neuroimmunology Research Group, Netherlands Institute for Neuroscience, 1105 BA, Amsterdam, The Netherlands; Department of Neurology, MS Center ErasMS, Erasmus MC, University Medical Center Rotterdam, 3015 CN, Rotterdam, The Netherlands
| | - Marvin M van Luijn
- Department of Immunology, MS Center ErasMS, Erasmus MC, University Medical Center Rotterdam, 3015 CN, Rotterdam, The Netherlands.
| |
Collapse
|
11
|
Wang C, Zhou Y, Feinstein A. Neuro-immune crosstalk in depressive symptoms of multiple sclerosis. Neurobiol Dis 2023; 177:106005. [PMID: 36680805 DOI: 10.1016/j.nbd.2023.106005] [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: 05/23/2022] [Revised: 01/10/2023] [Accepted: 01/17/2023] [Indexed: 01/20/2023] Open
Abstract
Depressive disorders can occur in up to 50% of people with multiple sclerosis in their lifetime. If left untreated, comorbid major depressive disorders may not spontaneously remit and is associated with an increased morbidity and mortality. Conversely, epidemiological evidence supports increased psychiatric visit as a significant prodromal event prior to diagnosis of MS. Are there common molecular pathways that contribute to the co-development of MS and psychiatric illnesses? We discuss immune cells that are dysregulated in MS and how such dysregulation can induce or protect against depressive symptoms. This is not meant to be a comprehensive review of all molecular pathways but rather a framework to guide future investigations of immune responses in depressed versus euthymic people with MS. Currently, there is weak evidence supporting the use of antidepressant medication in comorbid MS patients. It is our hope that by better understanding the neuroimmune crosstalk in the context of depression in MS, we can enhance the potential for future therapeutic options.
Collapse
Affiliation(s)
- Chao Wang
- Biological Sciences Platform, Sunnybrook Research Institute, Toronto, ON, Canada; Department of Immunology, University of Toronto, Toronto, ON, Canada
| | - Yulin Zhou
- Biological Sciences Platform, Sunnybrook Research Institute, Toronto, ON, Canada; Department of Immunology, University of Toronto, Toronto, ON, Canada
| | - Anthony Feinstein
- Department of Psychiatry, Sunnybrook Health Sciences Centre and University of Toronto, Toronto, ON, Canada.
| |
Collapse
|
12
|
Mey GM, Mahajan KR, DeSilva TM. Neurodegeneration in multiple sclerosis. WIREs Mech Dis 2023; 15:e1583. [PMID: 35948371 PMCID: PMC9839517 DOI: 10.1002/wsbm.1583] [Citation(s) in RCA: 29] [Impact Index Per Article: 29.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2022] [Revised: 06/28/2022] [Accepted: 07/11/2022] [Indexed: 01/31/2023]
Abstract
Axonal loss in multiple sclerosis (MS) is a key component of disease progression and permanent neurologic disability. MS is a heterogeneous demyelinating and neurodegenerative disease of the central nervous system (CNS) with varying presentation, disease courses, and prognosis. Immunomodulatory therapies reduce the frequency and severity of inflammatory demyelinating events that are a hallmark of MS, but there is minimal therapy to treat progressive disease and there is no cure. Data from patients with MS, post-mortem histological analysis, and animal models of demyelinating disease have elucidated patterns of MS pathogenesis and underlying mechanisms of neurodegeneration. MRI and molecular biomarkers have been proposed to identify predictors of neurodegeneration and risk factors for disease progression. Early signs of axonal dysfunction have come to light including impaired mitochondrial trafficking, structural axonal changes, and synaptic alterations. With sustained inflammation as well as impaired remyelination, axons succumb to degeneration contributing to CNS atrophy and worsening of disease. These studies highlight the role of chronic demyelination in the CNS in perpetuating axonal loss, and the difficulty in promoting remyelination and repair amidst persistent inflammatory insult. Regenerative and neuroprotective strategies are essential to overcome this barrier, with early intervention being critical to rescue axonal integrity and function. The clinical and basic research studies discussed in this review have set the stage for identifying key propagators of neurodegeneration in MS, leading the way for neuroprotective therapeutic development. This article is categorized under: Immune System Diseases > Molecular and Cellular Physiology Neurological Diseases > Molecular and Cellular Physiology.
Collapse
Affiliation(s)
- Gabrielle M. Mey
- Department of NeurosciencesLerner Research Institute, Cleveland Clinic Foundation, and Case Western Reserve UniversityClevelandOhioUSA
| | - Kedar R. Mahajan
- Department of NeurosciencesLerner Research Institute, Cleveland Clinic Foundation, and Case Western Reserve UniversityClevelandOhioUSA
- Mellen Center for MS Treatment and ResearchNeurological Institute, Cleveland Clinic FoundationClevelandOhioUSA
| | - Tara M. DeSilva
- Department of NeurosciencesLerner Research Institute, Cleveland Clinic Foundation, and Case Western Reserve UniversityClevelandOhioUSA
| |
Collapse
|
13
|
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: 0.5] [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
|
14
|
Chisari CG, Sgarlata E, Arena S, Toscano S, Luca M, Patti F. Rituximab for the treatment of multiple sclerosis: a review. J Neurol 2022; 269:159-183. [PMID: 33416999 PMCID: PMC7790722 DOI: 10.1007/s00415-020-10362-z] [Citation(s) in RCA: 88] [Impact Index Per Article: 44.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2020] [Revised: 12/03/2020] [Accepted: 12/08/2020] [Indexed: 01/07/2023]
Abstract
In the last decades, evidence suggesting the direct or indirect involvement of B cells on multiple sclerosis (MS) pathogenesis has accumulated. The increased amount of data on the efficacy and safety of B-cell-depleting therapies from several studies has suggested the addition of these drugs as treatment options to the current armamentarium of disease modifying therapies (DMTs) for MS. Particularly, rituximab (RTX), a chimeric monoclonal antibody directed at CD20 positive B lymphocytes resulting in cell-mediated apoptosis, has been demonstrated to reduce inflammatory activity, incidence of relapses and new brain lesions on magnetic resonance imaging (MRI) in patients with relapsing-remitting MS (RRMS). Additional evidence also demonstrated that patients with progressive MS (PMS) may benefit from RTX, which also showed to be well tolerated, with acceptable safety risks and favorable cost-effectiveness profile.Despite these encouraging results, RTX is currently approved for non-Hodgkin's lymphoma, chronic lymphocytic leukemia, several forms of vasculitis and rheumatoid arthritis, while it can only be administered off-label for MS treatment. Between Northern European countries exist different rules for using not licensed drug for treating MS. The Sweden MS register reports a high rate (53.5%) of off-label RTX prescriptions in relation to other annually started DMTs to treat MS patients, while Danish and Norwegian neurologists have to use other anti-CD20 drugs, as ocrelizumab, in most of the cases.In this paper, we review the pharmacokinetics, pharmacodynamics, clinical efficacy, safety profile and cost effectiveness aspects of RTX for the treatment of MS. Particularly, with the approval of new anti-CD20 DMTs, the recent worldwide COVID-19 emergency and the possible increased risk of infection with this class of drugs, this review sheds light on the use of RTX as an alternative treatment option for MS management, while commenting the gaps of knowledge regarding this drug.
Collapse
Affiliation(s)
- Clara Grazia Chisari
- Department "GF Ingrassia", Section of Neurosciences, University of Catania, Catania, Italy
| | - Eleonora Sgarlata
- Department "GF Ingrassia", Section of Neurosciences, University of Catania, Catania, Italy
- Stroke Unit, Department of Medicine, Umberto I Hospital, Siracusa, Italy
| | - Sebastiano Arena
- Department "GF Ingrassia", Section of Neurosciences, University of Catania, Catania, Italy
| | - Simona Toscano
- Department "GF Ingrassia", Section of Neurosciences, University of Catania, Catania, Italy
| | - Maria Luca
- Department "GF Ingrassia", Section of Neurosciences, University of Catania, Catania, Italy
| | - Francesco Patti
- Department "GF Ingrassia", Section of Neurosciences, University of Catania, Catania, Italy.
| |
Collapse
|
15
|
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: 1.0] [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
|
16
|
Muñoz U, Sebal C, Escudero E, Esiri M, Tzartos J, Sloan C, Sadaba MC. Main Role of Antibodies in Demyelination and Axonal Damage in Multiple Sclerosis. Cell Mol Neurobiol 2021; 42:1809-1827. [PMID: 33625628 DOI: 10.1007/s10571-021-01059-6] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2020] [Accepted: 02/11/2021] [Indexed: 11/28/2022]
Abstract
Antibodies and oxidative stress are hallmarks of multiple sclerosis (MS) lesions. We aimed to clarify the relation between them, their role in MS patients and to investigate their specificity, comparing MS with classical neurodegenerative diseases (ND). Brain samples from 14 MS cases, 6 with ND and 9 controls (without neurological diseases). Immunohistochemistry assays were used to detect oxidized lipids (EO6), IgG and IgM, oligodendrocytes (Olig2), axons (NF, neurofilament) and cellular (TUNEL) and axonal damage (APP, amyloid precursor protein). We did not observe EO6 in controls. All samples from MS patients showed EO6 in oligodendrocytes and axons within lesions. We did not detect co-localization between EO6 and antibodies. Neither did we between EO6 and TUNEL or APP. 94.4% of TUNEL-positive cells in normal appearing white matter were also stained for IgG and 75.5% for IgM. IgM, but not IgG, co-localized with APP. EO6 was associated with axonal damage in amyotrophic lateral sclerosis (ALS). We did not observe association between antibodies and cellular or axonal damage in ND patients. MS patients showed a higher number of B cells and plasma cells in the lesions and meninges than controls. The number of B cells and plasma cells was associated with the presence of antibodies and with the activity of the lesions. We observed a main role of B lymphocytes in the development of MS lesions. Antibodies contribute to the oligodendrocyte and axonal damage in MS. Oxidative stress was associated with axonal damage in ALS.
Collapse
Affiliation(s)
- Ursula Muñoz
- Facultad de Medicina Instituto de Medicina Molecular Aplicada (IMMA), Universidad San Pablo-CEU, CEU Universities, Madrid, Spain.
| | - Cristina Sebal
- Facultad de Medicina Instituto de Medicina Molecular Aplicada (IMMA), Universidad San Pablo-CEU, CEU Universities, Madrid, Spain
| | - Esther Escudero
- Facultad de Medicina Instituto de Medicina Molecular Aplicada (IMMA), Universidad San Pablo-CEU, CEU Universities, Madrid, Spain
| | - Margaret Esiri
- Nuffield Department of Clinical Neuroscience, University of Oxford, Oxford, UK
| | | | - Carolyn Sloan
- Nuffield Department of Clinical Neuroscience, University of Oxford, Oxford, UK
| | - Mari Cruz Sadaba
- Facultad de Medicina Instituto de Medicina Molecular Aplicada (IMMA), Universidad San Pablo-CEU, CEU Universities, Madrid, Spain.
| |
Collapse
|
17
|
Bakhuraysah MM, Theotokis P, Lee JY, Alrehaili AA, Aui PM, Figgett WA, Azari MF, Abou-Afech JP, Mackay F, Siatskas C, Alderuccio F, Strittmatter SM, Grigoriadis N, Petratos S. B-cells expressing NgR1 and NgR3 are localized to EAE-induced inflammatory infiltrates and are stimulated by BAFF. Sci Rep 2021; 11:2890. [PMID: 33536561 PMCID: PMC7858582 DOI: 10.1038/s41598-021-82346-6] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2019] [Accepted: 01/14/2021] [Indexed: 02/01/2023] Open
Abstract
We have previously reported evidence that Nogo-A activation of Nogo-receptor 1 (NgR1) can drive axonal dystrophy during the neurological progression of experimental autoimmune encephalomyelitis (EAE). However, the B-cell activating factor (BAFF/BlyS) may also be an important ligand of NgR during neuroinflammation. In the current study we define that NgR1 and its homologs may contribute to immune cell signaling during EAE. Meningeal B-cells expressing NgR1 and NgR3 were identified within the lumbosacral spinal cords of ngr1+/+ EAE-induced mice at clinical score 1. Furthermore, increased secretion of immunoglobulins that bound to central nervous system myelin were shown to be generated from isolated NgR1- and NgR3-expressing B-cells of ngr1+/+ EAE-induced mice. In vitro BAFF stimulation of NgR1- and NgR3-expressing B cells, directed them into the cell cycle DNA synthesis phase. However, when we antagonized BAFF signaling by co-incubation with recombinant BAFF-R, NgR1-Fc, or NgR3 peptides, the B cells remained in the G0/G1 phase. The data suggest that B cells express NgR1 and NgR3 during EAE, being localized to infiltrates of the meninges and that their regulation is governed by BAFF signaling.
Collapse
Affiliation(s)
- Maha M Bakhuraysah
- Department of Neuroscience, Central Clinical School, Monash University, Prahran, VIC, 3004, Australia
- Faculty of Applied Medical Sciences, Taif University, Taif, 26521, Kingdom of Saudi Arabia
| | - Paschalis Theotokis
- Laboratory of Experimental Neurology and Neuroimmunology, Department of Neurology, AHEPA University Hospital, 54636, Thessaloniki, Macedonia, Greece
| | - Jae Young Lee
- Department of Neuroscience, Central Clinical School, Monash University, Prahran, VIC, 3004, Australia
- Toolgen Inc., Gasan Digital-Ro, 08594, Geumcheon, Seoul, Korea
| | - Amani A Alrehaili
- Department of Neuroscience, Central Clinical School, Monash University, Prahran, VIC, 3004, Australia
- Faculty of Applied Medical Sciences, Taif University, Taif, 26521, Kingdom of Saudi Arabia
| | - Pei-Mun Aui
- Department of Neuroscience, Central Clinical School, Monash University, Prahran, VIC, 3004, Australia
| | - William A Figgett
- Department of Microbiology and Immunology, School of Biomedical Science, Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, VIC, 3000, Australia
| | - Michael F Azari
- Department of Neuroscience, Central Clinical School, Monash University, Prahran, VIC, 3004, Australia
| | - John-Paul Abou-Afech
- Department of Neuroscience, Central Clinical School, Monash University, Prahran, VIC, 3004, Australia
| | - Fabienne Mackay
- Department of Microbiology and Immunology, School of Biomedical Science, Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, VIC, 3000, Australia
- QIMR Berghofer Medical Research Institute, Herston, QLD, 4006, Australia
| | | | - Frank Alderuccio
- Department of Immunology and Pathology, Central Clinical School, Monash University, Prahran, VIC, 3004, Australia
| | - Stephen M Strittmatter
- Program in Cellular Neuroscience, Neurodegeneration and Repair, Yale University School of Medicine, New Haven, CT, 06536, USA
| | - Nikolaos Grigoriadis
- Laboratory of Experimental Neurology and Neuroimmunology, Department of Neurology, AHEPA University Hospital, 54636, Thessaloniki, Macedonia, Greece
| | - Steven Petratos
- Department of Neuroscience, Central Clinical School, Monash University, Prahran, VIC, 3004, Australia.
| |
Collapse
|
18
|
Armangue T, Capobianco M, de Chalus A, Laetitia G, Deiva K. E.U. paediatric MOG consortium consensus: Part 3 - Biomarkers of paediatric myelin oligodendrocyte glycoprotein antibody-associated disorders. Eur J Paediatr Neurol 2020; 29:22-31. [PMID: 33191096 DOI: 10.1016/j.ejpn.2020.11.001] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/30/2020] [Revised: 10/31/2020] [Accepted: 11/02/2020] [Indexed: 12/12/2022]
Abstract
A first episode of acquired demyelinating disorder (ADS) in children is a diagnostic challenge as different diseases can express similar clinical features. Recently, antibodies against myelin oligodendrocyte glycoprotein (MOG) have emerged as a new ADS biomarker, which clearly allow the identification of monophasic and relapsing ADS forms different from MS predominantly in children. Due to the novelty of this antibody there are still challenges and controversies about its pathogenicity and best technique to detect it. In this manuscript we will discuss the recommendations and caveats on MOG antibody assays, role in the pathogenesis, and additionally discuss the usefulness of other potential new biomarkers in MOG-antibody associated disorders (MOGAD).
Collapse
Affiliation(s)
- Thaís Armangue
- Neuroimmunology Program, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Hospital Clínic, Universitat de Barcelona, Barcelona, Spain; Pediatric Neuroimmunology Unit, Neurology Department, Sant Joan de Déu (SJD) Children's Hospital, University of Barcelona, Barcelona, Spain.
| | - Marco Capobianco
- Department of Neurology and Regional Multiple Sclerosis Centre, University Hospital San Luigi Gonzaga, Orbassano, Italy
| | - Aliénor de Chalus
- Assistance Publique-Hôpitaux de Paris, Pediatric Neurology Department, University Hospitals Paris Saclay, Bicêtre Hospital, Le Kremlin Bicêtre, France
| | - Giorgi Laetitia
- Assistance Publique-Hôpitaux de Paris, Pediatric Neurology Department, University Hospitals Paris Saclay, Bicêtre Hospital, Le Kremlin Bicêtre, France
| | - Kumaran Deiva
- Assistance Publique-Hôpitaux de Paris, Pediatric Neurology Department, University Hospitals Paris Saclay, Bicêtre Hospital, Le Kremlin Bicêtre, France; French Reference Network of Rare Inflammatory Brain and Spinal Diseases, Le Kremlin Bicêtre, France and European Reference Network-RITA, France
| | | |
Collapse
|
19
|
Wang A, Rojas O, Lee D, Gommerman JL. Regulation of neuroinflammation by B cells and plasma cells. Immunol Rev 2020; 299:45-60. [PMID: 33107072 DOI: 10.1111/imr.12929] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2020] [Accepted: 09/07/2020] [Indexed: 02/06/2023]
Abstract
The remarkable success of anti-CD20 B cell depletion therapies in reducing the burden of multiple sclerosis (MS) disease has prompted significant interest in how B cells contribute to neuroinflammation. Most focus has been on identifying pathogenic CD20+ B cells. However, an increasing number of studies have also identified regulatory functions of B lineage cells, particularly the production of IL-10, as being associated with disease remission in anti-CD20-treated MS patients. Moreover, IL-10-producing B cells have been linked to the attenuation of inflammation in experimental autoimmune encephalomyelitis (EAE), the animal model of MS. In addition to IL-10-producing B cells, antibody-producing plasma cells (PCs) have also been implicated in suppressing neuroinflammation. This review will examine regulatory roles for B cells and PCs in MS and EAE. In addition, we speculate on the involvement of regulatory PCs and the cytokine BAFF in the context of anti-CD20 treatment. Lastly, we explore how the microbiota could influence anti-inflammatory B cell behavior. A better understanding of the contributions of different B cell subsets to the regulation of neuroinflammation, and factors that impact the development, maintenance, and migration of such subsets, will be important for rationalizing next-generation B cell-directed therapies for the treatment of MS.
Collapse
Affiliation(s)
- Angela Wang
- Department of Immunology, University of Toronto, Toronto, ON, Canada
| | - Olga Rojas
- Department of Immunology, University of Toronto, Toronto, ON, Canada
| | - Dennis Lee
- Department of Immunology, University of Toronto, Toronto, ON, Canada
| | | |
Collapse
|
20
|
Yu X, Graner M, Kennedy PGE, Liu Y. The Role of Antibodies in the Pathogenesis of Multiple Sclerosis. Front Neurol 2020; 11:533388. [PMID: 33192968 PMCID: PMC7606501 DOI: 10.3389/fneur.2020.533388] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2020] [Accepted: 09/08/2020] [Indexed: 01/09/2023] Open
Abstract
The presence of persistent intrathecal oligoclonal immunoglobulin G (IgG) bands (OCBs) and lesional IgG deposition are seminal features of multiple sclerosis (MS) disease pathology. Despite extensive investigations, the role of antibodies, the products of mature CD19+ B cells, in disease development is still controversial and under significant debate. Recent success of B cell depletion therapies has revealed that CD20+ B cells contribute to MS pathogenesis via both antigen-presentation and T-cell-regulation. However, the limited efficacy of CD20+ B cell depletion therapies for the treatment of progressive MS indicates that additional mechanisms are involved. In this review, we present findings suggesting a potential pathological role for increased intrathecal IgGs, the relation of circulating antibodies to intrathecal IgGs, and the selective elevation of IgG1 and IgG3 subclasses in MS. We propose a working hypothesis that circulating B cells and antibodies contribute significantly to intrathecal IgGs, thereby exerting primary and pathogenic effects in MS development. Increased levels of IgG1 and IgG3 antibodies induce potent antibody-mediated cytotoxicity to central nervous system (CNS) cells and/or reduce the threshold required for antigen-driven antibody clustering leading to optimal activation of immune responses. Direct proof of the pathogenic roles of antibodies in MS may provide opportunities for novel blood biomarker identification as well as strategies for the development of effective therapeutic interventions.
Collapse
Affiliation(s)
- Xiaoli Yu
- Department of Neurosurgery, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Michael Graner
- Department of Neurosurgery, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Peter G E Kennedy
- Institute of Infection, Immunity and Inflammation, University of Glasgow, Glasgow, United Kingdom
| | - Yiting Liu
- Department of Neurology, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| |
Collapse
|
21
|
Mahler MR, Søndergaard HB, Buhelt S, von Essen MR, Romme Christensen J, Enevold C, Sellebjerg F. Multiplex assessment of cerebrospinal fluid biomarkers in multiple sclerosis. Mult Scler Relat Disord 2020; 45:102391. [DOI: 10.1016/j.msard.2020.102391] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2020] [Revised: 07/09/2020] [Accepted: 07/10/2020] [Indexed: 11/25/2022]
|
22
|
Differential Effects of MS Therapeutics on B Cells-Implications for Their Use and Failure in AQP4-Positive NMOSD Patients. Int J Mol Sci 2020; 21:ijms21145021. [PMID: 32708663 PMCID: PMC7404039 DOI: 10.3390/ijms21145021] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2020] [Revised: 07/11/2020] [Accepted: 07/13/2020] [Indexed: 12/25/2022] Open
Abstract
B cells are considered major contributors to multiple sclerosis (MS) pathophysiology. While lately approved disease-modifying drugs like ocrelizumab deplete B cells directly, most MS medications were not primarily designed to target B cells. Here, we review the current understanding how approved MS medications affect peripheral B lymphocytes in humans. These highly contrasting effects are of substantial importance when considering these drugs as therapy for neuromyelitis optica spectrum disorders (NMOSD), a frequent differential diagnosis to MS, which is considered being a primarily B cell- and antibody-driven diseases. Data indicates that MS medications, which deplete B cells or induce an anti-inflammatory phenotype of the remaining ones, were effective and safe in aquaporin-4 antibody positive NMOSD. In contrast, drugs such as natalizumab and interferon-β, which lead to activation and accumulation of B cells in the peripheral blood, lack efficacy or even induce catastrophic disease activity in NMOSD. Hence, we conclude that the differential effect of MS drugs on B cells is one potential parameter determining the therapeutic efficacy or failure in antibody-dependent diseases like seropositive NMOSD.
Collapse
|
23
|
Gross CC, Pawlitzki M, Schulte-Mecklenbeck A, Rolfes L, Ruck T, Hundehege P, Wiendl H, Herty M, Meuth SG. Generation of a Model to Predict Differentiation and Migration of Lymphocyte Subsets under Homeostatic and CNS Autoinflammatory Conditions. Int J Mol Sci 2020; 21:ijms21062046. [PMID: 32192056 PMCID: PMC7139518 DOI: 10.3390/ijms21062046] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2020] [Revised: 03/12/2020] [Accepted: 03/13/2020] [Indexed: 12/15/2022] Open
Abstract
The central nervous system (CNS) is an immune-privileged compartment that is separated from the circulating blood and the peripheral organs by the blood–brain and the blood–cerebrospinal fluid (CSF) barriers. Transmigration of lymphocyte subsets across these barriers and their activation/differentiation within the periphery and intrathecal compartments in health and autoinflammatory CNS disease are complex. Mathematical models are warranted that qualitatively and quantitatively predict the distribution and differentiation stages of lymphocyte subsets in the blood and CSF. Here, we propose a probabilistic mathematical model that (i) correctly reproduces acquired data on location and differentiation states of distinct lymphocyte subsets under homeostatic and neuroinflammatory conditions, (ii) provides a quantitative assessment of differentiation and transmigration rates under these conditions, (iii) correctly predicts the qualitative behavior of immune-modulating therapies, (iv) and enables simulation-based prediction of distribution and differentiation stages of lymphocyte subsets in the case of limited access to biomaterial. Taken together, this model might reduce future measurements in the CSF compartment and allows for the assessment of the effectiveness of different immune-modulating therapies.
Collapse
Affiliation(s)
- Catharina C. Gross
- Department of Neurology with Institute of Translational Neurology, University Hospital Münster, Albert-Schweitzer-Campus 1, Building A01, D-48149 Münster, Germany; (M.P.); (A.S.-M.); (L.R.); (T.R.); (P.H.); (H.W.); (S.G.M.)
- Correspondence:
| | - Marc Pawlitzki
- Department of Neurology with Institute of Translational Neurology, University Hospital Münster, Albert-Schweitzer-Campus 1, Building A01, D-48149 Münster, Germany; (M.P.); (A.S.-M.); (L.R.); (T.R.); (P.H.); (H.W.); (S.G.M.)
| | - Andreas Schulte-Mecklenbeck
- Department of Neurology with Institute of Translational Neurology, University Hospital Münster, Albert-Schweitzer-Campus 1, Building A01, D-48149 Münster, Germany; (M.P.); (A.S.-M.); (L.R.); (T.R.); (P.H.); (H.W.); (S.G.M.)
| | - Leoni Rolfes
- Department of Neurology with Institute of Translational Neurology, University Hospital Münster, Albert-Schweitzer-Campus 1, Building A01, D-48149 Münster, Germany; (M.P.); (A.S.-M.); (L.R.); (T.R.); (P.H.); (H.W.); (S.G.M.)
| | - Tobias Ruck
- Department of Neurology with Institute of Translational Neurology, University Hospital Münster, Albert-Schweitzer-Campus 1, Building A01, D-48149 Münster, Germany; (M.P.); (A.S.-M.); (L.R.); (T.R.); (P.H.); (H.W.); (S.G.M.)
| | - Petra Hundehege
- Department of Neurology with Institute of Translational Neurology, University Hospital Münster, Albert-Schweitzer-Campus 1, Building A01, D-48149 Münster, Germany; (M.P.); (A.S.-M.); (L.R.); (T.R.); (P.H.); (H.W.); (S.G.M.)
| | - Heinz Wiendl
- Department of Neurology with Institute of Translational Neurology, University Hospital Münster, Albert-Schweitzer-Campus 1, Building A01, D-48149 Münster, Germany; (M.P.); (A.S.-M.); (L.R.); (T.R.); (P.H.); (H.W.); (S.G.M.)
| | - Michael Herty
- Institute of Geometry and Applied Mathematics, RWTH Aachen University, Templergraben 55, D-52056 Aachen, Germany;
| | - Sven G. Meuth
- Department of Neurology with Institute of Translational Neurology, University Hospital Münster, Albert-Schweitzer-Campus 1, Building A01, D-48149 Münster, Germany; (M.P.); (A.S.-M.); (L.R.); (T.R.); (P.H.); (H.W.); (S.G.M.)
| |
Collapse
|
24
|
An Overview of the Intrinsic Role of Citrullination in Autoimmune Disorders. J Immunol Res 2019; 2019:7592851. [PMID: 31886309 PMCID: PMC6899306 DOI: 10.1155/2019/7592851] [Citation(s) in RCA: 58] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2018] [Revised: 04/03/2019] [Accepted: 09/28/2019] [Indexed: 02/07/2023] Open
Abstract
A protein undergoes many types of posttranslation modification. Citrullination is one of these modifications, where an arginine amino acid is converted to a citrulline amino acid. This process depends on catalytic enzymes such as peptidylarginine deiminase enzymes (PADs). This modification leads to a charge shift, which affects the protein structure, protein-protein interactions, and hydrogen bond formation, and it may cause protein denaturation. The irreversible citrullination reaction is not limited to a specific protein, cell, or tissue. It can target a wide range of proteins in the cell membrane, cytoplasm, nucleus, and mitochondria. Citrullination is a normal reaction during cell death. Apoptosis is normally accompanied with a clearance process via scavenger cells. A defect in the clearance system either in terms of efficiency or capacity may occur due to massive cell death, which may result in the accumulation and leakage of PAD enzymes and the citrullinated peptide from the necrotized cell which could be recognized by the immune system, where the immunological tolerance will be avoided and the autoimmune disorders will be subsequently triggered. The induction of autoimmune responses, autoantibody production, and cytokines involved in the major autoimmune diseases will be discussed.
Collapse
|
25
|
B cells in autoimmune and neurodegenerative central nervous system diseases. Nat Rev Neurosci 2019; 20:728-745. [DOI: 10.1038/s41583-019-0233-2] [Citation(s) in RCA: 128] [Impact Index Per Article: 25.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/30/2019] [Indexed: 12/16/2022]
|
26
|
Huss A, Mojib-Yezdani F, Bachhuber F, Fangerau T, Lewerenz J, Otto M, Tumani H, Senel M. Association of cerebrospinal fluid kappa free light chains with the intrathecal polyspecific antiviral immune response in multiple sclerosis. Clin Chim Acta 2019; 498:148-153. [PMID: 31437445 DOI: 10.1016/j.cca.2019.08.016] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2018] [Revised: 08/15/2019] [Accepted: 08/18/2019] [Indexed: 11/20/2022]
Abstract
The polyspecific B-lymphocyte response to neurotropic viruses such as measles (M), rubella (R) and varicella zoster (Z), known as MRZ reaction, is to-date the most specific neurochemical marker for multiple sclerosis (MS). The aim of this study was to investigate a possible association of immunoglobulin (Ig) kappa (κ-) and lambda (λ-) free light chains (FLC) with the presence of the MRZ reaction in multiple sclerosis. Immunoglobulin κ- and λ-FLC, MRZ reaction, oligoclonal IgG bands (OCB), and cerebrospinal fluid (CSF) routine parameters were measured in 65 MS patients. OCB were detected in 97% of MS patients, intrathecal IgG synthesis according to Reiber was detectable in 57%, an elevated IgG index (>0.7) in 66% and the MRZR was positive in 45%. All investigated κ-values (CSF κFLC, CSF-serum ratio of κFLCs (QκFLC), and κFLC index (κFLC/QAlbumin)) were significantly higher in patients with positive MRZ reaction as compared to MRZ negative MS patients. In contrast, λ-values showed no significant differences. Additionally to the putative diagnostic sensitivity and prognostic value of κFLC, the association of κFLC with a highly specific neurochemical marker for MS - the MRZ reaction, especially the determination of κFLCs is an informative tool to assess the B-cell response and determine its extent in MS patients.
Collapse
Affiliation(s)
- André Huss
- Department of Neurology, University of Ulm, Ulm, Germany
| | | | | | - Tanja Fangerau
- Department of Neurology, University of Ulm, 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
| | - Makbule Senel
- Department of Neurology, University of Ulm, Ulm, Germany.
| |
Collapse
|
27
|
|
28
|
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: 37] [Impact Index Per Article: 7.4] [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
|
29
|
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: 49] [Impact Index Per Article: 9.8] [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
|
30
|
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: 13] [Impact Index Per Article: 2.6] [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
|
31
|
D'Amico E, Zanghì A, Gastaldi M, Patti F, Zappia M, Franciotta D. Placing CD20-targeted B cell depletion in multiple sclerosis therapeutic scenario: Present and future perspectives. Autoimmun Rev 2019; 18:665-672. [PMID: 31059839 DOI: 10.1016/j.autrev.2019.05.003] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2019] [Accepted: 01/18/2019] [Indexed: 12/26/2022]
Abstract
Multiple sclerosis (MS) is an acquired demyelinating disease of the central nervous system (CNS) that traditionally has been considered to be mediated primarily by T cells. Increasing evidence, however, suggests the fundamental role of B cells in the pathogenesis and development of the disease. Recently, anti-CD20 B cell-based therapies have demonstrated impressive and somewhat surprising results in MS, showing profound anti-inflammatory effects with a favorable risk-benefit ratio. Moreover, for the first time in the MS therapeutic scenario, the anti-CD20 monoclonal antibody ocrelizumab has been granted for the treatment of the primary progressive form of the disease. In this review, we provide a brief overview about anti-CD20 B cell-based therapies in MS, in the perspective of their influence on the future management of the disease, and of their possible positioning in a new wider therapeutic scenario.
Collapse
Affiliation(s)
| | - Aurora Zanghì
- Department G.F.Ingrassia, University of Catania, Italy
| | - Matteo Gastaldi
- Neuroimmunology Laboratory, IRCCS Mondino Foundation, Pavia, Italy
| | | | - Mario Zappia
- Department G.F.Ingrassia, University of Catania, Italy
| | - Diego Franciotta
- Neuroimmunology Laboratory, IRCCS Mondino Foundation, Pavia, Italy
| |
Collapse
|
32
|
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: 4.8] [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
|
33
|
Greenfield AL, Dandekar R, Ramesh A, Eggers EL, Wu H, Laurent S, Harkin W, Pierson NS, Weber MS, Henry RG, Bischof A, Cree BA, Hauser SL, Wilson MR, von Büdingen HC. Longitudinally persistent cerebrospinal fluid B cells can resist treatment in multiple sclerosis. JCI Insight 2019; 4:126599. [PMID: 30747723 DOI: 10.1172/jci.insight.126599] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2018] [Accepted: 02/05/2019] [Indexed: 12/27/2022] Open
Abstract
B cells are key contributors to chronic autoimmune pathology in multiple sclerosis (MS). Clonally related B cells exist in the cerebrospinal fluid (CSF), meninges, and CNS parenchyma of MS patients. We sought to investigate the presence of clonally related B cells over time by performing Ig heavy chain variable region repertoire sequencing on B cells from longitudinally collected blood and CSF samples of MS patients (n = 10). All patients were untreated at the time of the initial sampling; the majority (n = 7) were treated with immune-modulating therapies 1.2 (±0.3 SD) years later during the second sampling. We found clonal persistence of B cells in the CSF of 5 patients; these B cells were frequently Ig class-switched and CD27+. Specific blood B cell subsets appear to provide input into CNS repertoires over time. We demonstrate complex patterns of clonal B cell persistence in CSF and blood, even in patients on immune-modulating therapy. Our findings support the concept that peripheral B cell activation and CNS-compartmentalized immune mechanisms can in part be therapy resistant.
Collapse
Affiliation(s)
- Ariele L Greenfield
- UCSF Weill Institute for Neurosciences, Department of Neurology, UCSF, San Francisco, California, USA
| | - Ravi Dandekar
- UCSF Weill Institute for Neurosciences, Department of Neurology, UCSF, San Francisco, California, USA
| | - Akshaya Ramesh
- UCSF Weill Institute for Neurosciences, Department of Neurology, UCSF, San Francisco, California, USA
| | - Erica L Eggers
- UCSF Weill Institute for Neurosciences, Department of Neurology, UCSF, San Francisco, California, USA
| | - Hao Wu
- UCSF Weill Institute for Neurosciences, Department of Neurology, UCSF, San Francisco, California, USA
| | - Sarah Laurent
- UCSF Weill Institute for Neurosciences, Department of Neurology, UCSF, San Francisco, California, USA
| | - William Harkin
- UCSF Weill Institute for Neurosciences, Department of Neurology, UCSF, San Francisco, California, USA
| | - Natalie S Pierson
- UCSF Weill Institute for Neurosciences, Department of Neurology, UCSF, San Francisco, California, USA
| | - Martin S Weber
- Institute of Neuropathology, Department of Neurology, University Medical Center Göttingen, Germany
| | - Roland G Henry
- UCSF Weill Institute for Neurosciences, Department of Neurology, UCSF, San Francisco, California, USA
| | - Antje Bischof
- UCSF Weill Institute for Neurosciences, Department of Neurology, UCSF, San Francisco, California, USA
| | - Bruce Ac Cree
- UCSF Weill Institute for Neurosciences, Department of Neurology, UCSF, San Francisco, California, USA
| | - Stephen L Hauser
- UCSF Weill Institute for Neurosciences, Department of Neurology, UCSF, San Francisco, California, USA
| | - Michael R Wilson
- UCSF Weill Institute for Neurosciences, Department of Neurology, UCSF, San Francisco, California, USA
| | - H-Christian von Büdingen
- UCSF Weill Institute for Neurosciences, Department of Neurology, UCSF, San Francisco, California, USA
| |
Collapse
|
34
|
Giotaki I, Lange P, Weber MS. Modification of CSF findings in multiple sclerosis in the era of rapidly expanding treatment options. Expert Rev Neurother 2019; 19:277-283. [PMID: 30786785 DOI: 10.1080/14737175.2019.1583557] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
BACKGROUND The detection of cerebrospinal fluid (CSF)-specific oligoclonal bands (OCB) as well as an elevated antibody production directed against neurotrophic viruses remain hallmark findings in multiple sclerosis (MS). In general, individual patients show a unique pattern of OCB, which is believed to persist over time. Here, the authors revisited this assumption and investigated to what extent common anti-inflammatory MS medications can alter the level of immunoglobulins produced within the CSF. METHODS The authors analyzed three patients and compared CSF findings longitudinally. Two of these patients received corticosteroids, either systemically or intrathecally, the third patient was treated continuously with natalizumab. RESULTS In line with reports from other groups, the authors observed that continuous natalizumab treatment dampened the intrathecal immunoglobulin production in our patient. Exceeding this anticipated scenario, the authors detected that the continued administration of corticosteroids similarly reduced the production of CSF immunoglobulins, down to a level at which these parameters are considered normal. CONCLUSION These observations suggest that inflammatory CSF findings are more accessible to immunomodulatory MS treatment than previously thought, and that accordingly, their significance may transition from a sole diagnostic finding to a valuable therapeutic biomarker which may help to assess effective targeting of CNS-established inflammation in MS.
Collapse
Affiliation(s)
- Ioanna Giotaki
- a Department of Neurology , University Medical Center , Göttingen , Germany
| | - Peter Lange
- a Department of Neurology , University Medical Center , Göttingen , Germany
| | - Martin S Weber
- a Department of Neurology , University Medical Center , Göttingen , Germany.,b Institute of Neuropathology , University Medical Center , Göttingen , Germany
| |
Collapse
|
35
|
Myhr KM, Torkildsen Ø, Lossius A, Bø L, Holmøy T. B cell depletion in the treatment of multiple sclerosis. Expert Opin Biol Ther 2019; 19:261-271. [PMID: 30632834 DOI: 10.1080/14712598.2019.1568407] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/09/2023]
Abstract
INTRODUCTION Multiple sclerosis (MS) is a chronic inflammatory disease of the central nervous system. The latest development of B-cell depletion by anti-CD20 monoclonal antibodies has been a large step forward in the treatment of this devastating disease. AREAS COVERED In this manuscript, we review mechanisms of action, efficacy, safety, and tolerance of anti-CD20 therapies for MS, including rituximab, ocrelizumab, and ofatumumab. EXPERT OPINION B-cell depletion efficiently suppresses acute inflammatory disease activity in relapsing-remitting MS (RRMS), and may slowdown progression in primary progressive MS (PPMS). The treatment is generally well tolerated, with manageable adverse events related to infusion reactions and infections. Ocrelizumab, a humanized anti-CD20 monoclonal antibody, is the first therapy to be approved for the treatment of both RRMS and PPMS.
Collapse
Affiliation(s)
- Kjell-Morten Myhr
- a Department of Clinical Medicine , University of Bergen , Bergen , Norway.,b Department of Neurology , Haukeland University Hospital , Bergen , Norway
| | - Øivind Torkildsen
- a Department of Clinical Medicine , University of Bergen , Bergen , Norway.,b Department of Neurology , Haukeland University Hospital , Bergen , Norway
| | - Andreas Lossius
- c Department of Neurology , Akershus University Hospital , Lørenskog , Norway.,d Department of Immunology and Transfusion Medicine, Faculty of Medicine , University of Oslo and Oslo University Hospital Rikshospitalet , Oslo , Norway
| | - Lars Bø
- a Department of Clinical Medicine , University of Bergen , Bergen , Norway.,b Department of Neurology , Haukeland University Hospital , Bergen , Norway
| | - Trygve Holmøy
- c Department of Neurology , Akershus University Hospital , Lørenskog , Norway.,e Department of Clinical Medicine , University of Oslo , Oslo , Norway
| |
Collapse
|
36
|
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: 4.2] [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
|
37
|
Oligoclonal bands in multiple sclerosis; Functional significance and therapeutic implications. Does the specificity matter? Mult Scler Relat Disord 2018; 25:131-137. [DOI: 10.1016/j.msard.2018.07.030] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2018] [Revised: 07/13/2018] [Accepted: 07/16/2018] [Indexed: 01/24/2023]
|
38
|
Høglund RA, Polak J, Vartdal F, Holmøy T, Lossius A. B-cell composition in the blood and cerebrospinal fluid of multiple sclerosis patients treated with dimethyl fumarate. Mult Scler Relat Disord 2018; 26:90-95. [PMID: 30243235 DOI: 10.1016/j.msard.2018.08.032] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2018] [Revised: 08/22/2018] [Accepted: 08/30/2018] [Indexed: 12/18/2022]
Abstract
BACKGROUND B cells may contribute to the immunopathogenesis of multiple sclerosis (MS). Dimethyl fumarate (DMF) has recently been shown to reduce the frequency of memory B cells in blood, but it is not known whether the drug influences the cellular composition in the cerebrospinal fluid (CSF). METHODS A cross-sectional study examining the cellular composition in blood and cerebrospinal fluid (CSF) from 10 patients treated with DMF and 18 patients receiving other disease modifying drugs or no treatment. RESULTS Patients treated with DMF had reduced proportions of memory B cells in blood compared to other MS patients (p = 0.0007), and the reduction correlated with treatment duration (rs = -0.75, p = 0.021). In the CSF, the absolute number of mononuclear cells were significantly lower in DMF-treated patients compared to the other patients (p = 0.023), and there was a disproportionate decrease of plasmablasts (p = 0.031). CONCLUSION The results of this exploratory study support a B-cell mediated mechanism of action for DMF in both blood and CSF.
Collapse
Affiliation(s)
- Rune A Høglund
- Department of Neurology, Akershus University Hospital, Lørenskog, Norway; Institute of Clinical Medicine, University of Oslo, Oslo, Norway.
| | - Justyna Polak
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway; Department of Immunology and Transfusion Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Frode Vartdal
- Department of Immunology and Transfusion Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Trygve Holmøy
- Department of Neurology, Akershus University Hospital, Lørenskog, Norway; Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Andreas Lossius
- Department of Neurology, Akershus University Hospital, Lørenskog, Norway; Department of Immunology and Transfusion Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway.
| |
Collapse
|
39
|
Machado-Santos J, Saji E, Tröscher AR, Paunovic M, Liblau R, Gabriely G, Bien CG, Bauer J, Lassmann H. The compartmentalized inflammatory response in the multiple sclerosis brain is composed of tissue-resident CD8+ T lymphocytes and B cells. Brain 2018; 141:2066-2082. [PMID: 29873694 PMCID: PMC6022681 DOI: 10.1093/brain/awy151] [Citation(s) in RCA: 346] [Impact Index Per Article: 57.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2018] [Revised: 03/22/2018] [Accepted: 04/15/2018] [Indexed: 12/15/2022] Open
Abstract
Multiple sclerosis is an inflammatory demyelinating disease in which active demyelination and neurodegeneration are associated with lymphocyte infiltrates in the brain. However, so far little is known regarding the phenotype and function of these infiltrating lymphocyte populations. In this study, we performed an in-depth phenotypic characterization of T and B cell infiltrates in a large set of multiple sclerosis cases with different disease and lesion stages and compared the findings with those seen in inflammatory, non-inflammatory and normal human controls. In multiple sclerosis lesions, we found a dominance of CD8+ T cells and a prominent contribution of CD20+ B cells in all disease courses and lesion stages, including acute multiple sclerosis cases with very short disease duration, while CD4+ T cells were sparse. A dominance of CD8+ T cells was also seen in other inflammatory controls, such as Rasmussen's encephalitis and viral encephalitis, but the contribution of B cells in these diseases was modest. Phenotypic analysis of the CD8+ T cells suggested that part of the infiltrating cells in active lesions proliferate, show an activated cytotoxic phenotype and are in part destroyed by apoptosis. Further characterization of the remaining cells suggest that CD8+ T cells acquire features of tissue-resident memory cells, which may be focally reactivated in active lesions of acute, relapsing and progressive multiple sclerosis, while B cells, at least in part, gradually transform into plasma cells. The loss of surface molecules involved in the egress of leucocytes from inflamed tissue, such as S1P1 or CCR7, and the upregulation of CD103 expression may be responsible for the compartmentalization of the inflammatory response in established lesions. Similar phenotypic changes of tissue-infiltrating CD8+ T cells were also seen in Rasmussen's encephalitis. Our data underline the potential importance of CD8+ T lymphocytes and B cells in the inflammatory response in established multiple sclerosis lesions. Tissue-resident T and B cells may represent guardians of previous inflammatory brain disease, which can be reactivated and sustain the inflammatory response, when they are re-exposed to their specific antigen.
Collapse
Affiliation(s)
- Joana Machado-Santos
- Department of Neuroimmunology, Center for Brain Research, Medical University of Vienna, Vienna, Austria
| | - Etsuji Saji
- Department of Neurology, Brain Research Institute, Niigata University, Niigata, Japan
| | - Anna R Tröscher
- Department of Neuroimmunology, Center for Brain Research, Medical University of Vienna, Vienna, Austria
| | - Manuela Paunovic
- Department of Neuroimmunology, Center for Brain Research, Medical University of Vienna, Vienna, Austria
| | - Roland Liblau
- INSERM U1043 - CNRS UMR 5282, Centre de Physiopathologie Toulouse-Purpan, Université Toulouse III, Toulouse, F-31000, France
| | - Galina Gabriely
- Department of Neurology, Anne Romney Center for Neurologic Disease, Harvard Medical School, Boston, USA
| | | | - Jan Bauer
- Department of Neuroimmunology, Center for Brain Research, Medical University of Vienna, Vienna, Austria
| | - Hans Lassmann
- Department of Neuroimmunology, Center for Brain Research, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
40
|
Touil H, Kobert A, Lebeurrier N, Rieger A, Saikali P, Lambert C, Fawaz L, Moore CS, Prat A, Gommerman J, Antel JP, Itoyama Y, Nakashima I, Bar-Or A. Human central nervous system astrocytes support survival and activation of B cells: implications for MS pathogenesis. J Neuroinflammation 2018; 15:114. [PMID: 29673365 PMCID: PMC5907187 DOI: 10.1186/s12974-018-1136-2] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2018] [Accepted: 03/22/2018] [Indexed: 12/22/2022] Open
Abstract
Background The success of clinical trials of selective B cell depletion in patients with relapsing multiple sclerosis (MS) indicates B cells are important contributors to peripheral immune responses involved in the development of new relapses. Such B cell contribution to peripheral inflammation likely involves antibody-independent mechanisms. Of growing interest is the potential that B cells, within the MS central nervous system (CNS), may also contribute to the propagation of CNS-compartmentalized inflammation in progressive (non-relapsing) disease. B cells are known to persist in the inflamed MS CNS and are more recently described as concentrated in meningeal immune-cell aggregates, adjacent to the subpial cortical injury which has been associated with progressive disease. How B cells are fostered within the MS CNS and how they may contribute locally to the propagation of CNS-compartmentalized inflammation remain to be elucidated. Methods We considered whether activated human astrocytes might contribute to B cell survival and function through soluble factors. B cells from healthy controls (HC) and untreated MS patients were exposed to primary human astrocytes that were either maintained under basal culture conditions (non-activated) or pre-activated with standard inflammatory signals. B cell exposure to astrocytes included direct co-culture, co-culture in transwells, or exposure to astrocyte-conditioned medium. Following the different exposures, B cell survival and expression of T cell co-stimulatory molecules were assessed by flow cytometry, as was the ability of differentially exposed B cells to induce activation of allogeneic T cells. Results Secreted factors from both non-activated and activated human astrocytes robustly supported human B cell survival. Soluble products of pre-activated astrocytes also induced B cell upregulation of antigen-presenting cell machinery, and these B cells, in turn, were more efficient activators of T cells. Astrocyte-soluble factors could support survival and activation of B cell subsets implicated in MS, including memory B cells from patients with both relapsing and progressive forms of disease. Conclusions Our findings point to a potential mechanism whereby activated astrocytes in the inflamed MS CNS not only promote a B cell fostering environment, but also actively support the ability of B cells to contribute to the propagation of CNS-compartmentalized inflammation, now thought to play key roles in progressive disease. Electronic supplementary material The online version of this article (10.1186/s12974-018-1136-2) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Hanane Touil
- Neuroimmunology Unit, Montreal Neurological Institute, McGill University, 3801 University Street, Room 111, Montréal , QC, H3A 2B3, Canada.,Department of Neurology and Center for NeuroInflammation and Experimental Therapeutics (CNET), Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Antonia Kobert
- Neuroimmunology Unit, Montreal Neurological Institute, McGill University, 3801 University Street, Room 111, Montréal , QC, H3A 2B3, Canada
| | - Nathalie Lebeurrier
- Neuroimmunology Unit, Montreal Neurological Institute, McGill University, 3801 University Street, Room 111, Montréal , QC, H3A 2B3, Canada
| | - Aja Rieger
- Neuroimmunology Unit, Montreal Neurological Institute, McGill University, 3801 University Street, Room 111, Montréal , QC, H3A 2B3, Canada
| | - Philippe Saikali
- Neuroimmunology Unit, Montreal Neurological Institute, McGill University, 3801 University Street, Room 111, Montréal , QC, H3A 2B3, Canada
| | - Caroline Lambert
- Neuroimmunology Unit, Montreal Neurological Institute, McGill University, 3801 University Street, Room 111, Montréal , QC, H3A 2B3, Canada
| | - Lama Fawaz
- Neuroimmunology Unit, Montreal Neurological Institute, McGill University, 3801 University Street, Room 111, Montréal , QC, H3A 2B3, Canada
| | - Craig S Moore
- Neuroimmunology Unit, Montreal Neurological Institute, McGill University, 3801 University Street, Room 111, Montréal , QC, H3A 2B3, Canada.,Division of BioMedical Sciences, Faculty of Medicine, Memorial University of Newfoundland, St. John's, NF, Canada
| | - Alexandre Prat
- Université de Montréal Centre de Recherche du CHUM (CRCHUM) and Department of Neuroscience, Université de Montréal, 900 Saint Denis Street, Montreal, QC, H2X 0A9, Canada
| | - Jennifer Gommerman
- Department of Immunology, Medical Sciences Building, University of Toronto, Toronto, ON, M5S 1A8, Canada
| | - Jack P Antel
- Neuroimmunology Unit, Montreal Neurological Institute, McGill University, 3801 University Street, Room 111, Montréal , QC, H3A 2B3, Canada
| | - Yasuto Itoyama
- Department of Neurology, School of Medicine, Tohoku University, 1-1 Seiryo-machi, Aoba-ku, Sendai, Japan
| | - Ichiro Nakashima
- Neuroimmunology Unit, Montreal Neurological Institute, McGill University, 3801 University Street, Room 111, Montréal , QC, H3A 2B3, Canada.,Department of Neurology, School of Medicine, Tohoku University, 1-1 Seiryo-machi, Aoba-ku, Sendai, Japan
| | - Amit Bar-Or
- Neuroimmunology Unit, Montreal Neurological Institute, McGill University, 3801 University Street, Room 111, Montréal , QC, H3A 2B3, Canada. .,Department of Neurology and Center for NeuroInflammation and Experimental Therapeutics (CNET), Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| | | |
Collapse
|
41
|
Greenfield AL, Hauser SL. B-cell Therapy for Multiple Sclerosis: Entering an era. Ann Neurol 2018; 83:13-26. [PMID: 29244240 PMCID: PMC5876115 DOI: 10.1002/ana.25119] [Citation(s) in RCA: 159] [Impact Index Per Article: 26.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2017] [Revised: 12/01/2017] [Accepted: 12/09/2017] [Indexed: 12/12/2022]
Abstract
Monoclonal antibodies that target CD20 expressing B cells represent an important new treatment option for patients with multiple sclerosis (MS). B-cell-depleting therapy is highly effective against relapsing forms of the disease and is also the first treatment approach proven to protect against disability worsening in primary progressive MS. Moreover, evolving clinical experience with B-cell therapy, combined with a more sophisticated understanding of humoral immunity in preclinical models and in patients with MS, has led to major progress in deciphering the immune pathogenesis of MS. Here, we review the nuanced roles of B cells in MS autoimmunity, the clinical data supporting use of ocrelizumab and other anti-CD20 therapies in the treatment of MS, as well as safety and practical considerations for prescribing. Last, we summarize remaining unanswered questions regarding the proper role of anti-CD20 therapy in MS, its limitations, and the future landscape of B-cell-based approaches to treatment. Ann Neurol 2018;83:13-26.
Collapse
Affiliation(s)
- Ariele L. Greenfield
- Weill Institute for Neurosciences, Department of Neurology, University of California, San Francisco, San Francisco, California
| | - Stephen L. Hauser
- Weill Institute for Neurosciences, Department of Neurology, University of California, San Francisco, San Francisco, California
| |
Collapse
|
42
|
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: 105] [Impact Index Per Article: 17.5] [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
|
43
|
Pollok K, Mothes R, Ulbricht C, Liebheit A, Gerken JD, Uhlmann S, Paul F, Niesner R, Radbruch H, Hauser AE. The chronically inflamed central nervous system provides niches for long-lived plasma cells. Acta Neuropathol Commun 2017; 5:88. [PMID: 29178933 PMCID: PMC5702095 DOI: 10.1186/s40478-017-0487-8] [Citation(s) in RCA: 50] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2017] [Accepted: 10/30/2017] [Indexed: 11/11/2022] Open
Abstract
Although oligoclonal bands in the cerebrospinal fluid have been a hallmark of multiple sclerosis diagnosis for over three decades, the role of antibody-secreting cells in multiple sclerosis remains unclear. T and B cells are critical for multiple sclerosis pathogenesis, but increasing evidence suggests that plasma cells also contribute, through secretion of autoantibodies. Long-lived plasma cells are known to drive various chronic inflammatory conditions as e.g. systemic lupus erythematosus, however, to what extent they are present in autoimmune central nervous system inflammation has not yet been investigated. In brain biopsies from multiple sclerosis patients and other neurological diseases, we could detect non-proliferating plasma cells (CD138+Ki67−) in the parenchyma. Based on this finding, we hypothesized that long-lived plasma cells can persist in the central nervous system (CNS). In order to test this hypothesis, we adapted the multiple sclerosis mouse model experimental autoimmune encephalomyelitis to generate a B cell memory response. Plasma cells were found in the meninges and the parenchyma of the inflamed spinal cord, surrounded by tissue areas resembling survival niches for these cells, characterized by an up-regulation of chemokines (CXCL12), adhesion molecules (VCAM-1) and survival factors (APRIL and BAFF). In order to determine the lifetime of plasma cells in the chronically inflamed CNS, we labeled the DNA of proliferating cells with 5-ethynyl-2′-deoxyuridine (EdU). Up to five weeks later, we could detect EdU+ long-lived plasma cells in the murine CNS. To our knowledge, this is the first study describing non-proliferating plasma cells directly in the target tissue of a chronic inflammation in humans, as well as the first evidence demonstrating the ability of plasma cells to persist in the CNS, and the ability of the chronically inflamed CNS tissue to promote this persistence. Hence, our results suggest that the CNS provides survival niches for long-lived plasma cells, similar to the niches found in other organs. Targeting these cells in the CNS offers new perspectives for treatment of chronic autoimmune neuroinflammatory diseases, especially in patients who do not respond to conventional therapies.
Collapse
|
44
|
Dubey D, Forsthuber T, Flanagan EP, Pittock SJ, Stüve O. B-cell-targeted therapies in relapsing forms of MS. NEUROLOGY-NEUROIMMUNOLOGY & NEUROINFLAMMATION 2017; 4:e405. [PMID: 29082296 PMCID: PMC5656409 DOI: 10.1212/nxi.0000000000000405] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/11/2017] [Accepted: 07/13/2017] [Indexed: 01/04/2023]
Abstract
In recent years, there has been a significant increase in the therapeutic options available for the management of relapsing forms of MS. Therapies primarily targeting B cells, including therapeutic anti-CD20 monoclonal antibodies, have been evaluated in phase I, phase II, and phase III clinical trials. Results of these trials have shown their efficacy and relatively tolerable adverse effect profiles, suggesting a favorable benefit-to-risk ratio. In this review, we discuss the pathogenic role of B cells in MS and the rationale behind the utilization of B-cell depletion as a therapeutic cellular option. We also discuss the data of clinical trials for anti-CD20 antibodies in relapsing forms of MS and existing evidence for other B-cell–directed therapeutic strategies.
Collapse
Affiliation(s)
- Divyanshu Dubey
- Department of Neurology (D.B., E.P.F., S.J.P.), and Department of Laboratory Medicine and Pathology (S.J.P.), Mayo Clinic, Rochester, MN; Department of Biology (T.F.), University of Texas at San Antonio; Department of Neurology and Neurotherapeutics (O.S.), University of Texas Southwestern Medical Center, Dallas; Neurology Section (O.S.), VA North Texas Health Care System, Dallas VA Medical Center, TX; and Department of Neurology (O.S.), Klinikum rechts der Isar, Technische Universität München, Germany
| | - Thomas Forsthuber
- Department of Neurology (D.B., E.P.F., S.J.P.), and Department of Laboratory Medicine and Pathology (S.J.P.), Mayo Clinic, Rochester, MN; Department of Biology (T.F.), University of Texas at San Antonio; Department of Neurology and Neurotherapeutics (O.S.), University of Texas Southwestern Medical Center, Dallas; Neurology Section (O.S.), VA North Texas Health Care System, Dallas VA Medical Center, TX; and Department of Neurology (O.S.), Klinikum rechts der Isar, Technische Universität München, Germany
| | - Eoin P Flanagan
- Department of Neurology (D.B., E.P.F., S.J.P.), and Department of Laboratory Medicine and Pathology (S.J.P.), Mayo Clinic, Rochester, MN; Department of Biology (T.F.), University of Texas at San Antonio; Department of Neurology and Neurotherapeutics (O.S.), University of Texas Southwestern Medical Center, Dallas; Neurology Section (O.S.), VA North Texas Health Care System, Dallas VA Medical Center, TX; and Department of Neurology (O.S.), Klinikum rechts der Isar, Technische Universität München, Germany
| | - Sean J Pittock
- Department of Neurology (D.B., E.P.F., S.J.P.), and Department of Laboratory Medicine and Pathology (S.J.P.), Mayo Clinic, Rochester, MN; Department of Biology (T.F.), University of Texas at San Antonio; Department of Neurology and Neurotherapeutics (O.S.), University of Texas Southwestern Medical Center, Dallas; Neurology Section (O.S.), VA North Texas Health Care System, Dallas VA Medical Center, TX; and Department of Neurology (O.S.), Klinikum rechts der Isar, Technische Universität München, Germany
| | - Olaf Stüve
- Department of Neurology (D.B., E.P.F., S.J.P.), and Department of Laboratory Medicine and Pathology (S.J.P.), Mayo Clinic, Rochester, MN; Department of Biology (T.F.), University of Texas at San Antonio; Department of Neurology and Neurotherapeutics (O.S.), University of Texas Southwestern Medical Center, Dallas; Neurology Section (O.S.), VA North Texas Health Care System, Dallas VA Medical Center, TX; and Department of Neurology (O.S.), Klinikum rechts der Isar, Technische Universität München, Germany
| |
Collapse
|
45
|
Abstract
Highly effective anti-inflammatory therapies have so far been developed for patients with relapsing/remitting multiple sclerosis, which also show some benefits in the early progressive stage of the disease. However, treatment options for patients, who have entered the progressive phase, are still limited. Disease starts as an inflammatory process, which induces focal demyelinating lesions in the gray and white matter. This stage of the disease dominates in the relapsing phase, extends into the early stages of progressive disease, and can be targeted by current anti-inflammatory treatments. In parallel, inflammation accumulates behind a closed or repaired blood brain barrier, and this process peaks in the late relapsing and early progressive stage and then declines. Some data suggest that this process may be targeted by immune ablation and hematopoietic stem cell transplantation. In the late stage, inflammation may decline to levels seen in age-matched controls, but age and disease burden–related neurodegeneration ensues. Such neurodegeneration affects the damaged brain and spinal cord, in which functional reserve capacity is exhausted, giving rise to further disability progression. Anti-inflammatory treatments are unlikely to be beneficial in this stage of the disease, but neuroprotective and repair-inducing strategies may still be effective.
Collapse
Affiliation(s)
- Hans Lassmann
- Center for Brain Research, Medical University of Vienna, Wien, Austria
| |
Collapse
|
46
|
Høglund RA, Lossius A, Johansen JN, Homan J, Benth JŠ, Robins H, Bogen B, Bremel RD, Holmøy T. In Silico Prediction Analysis of Idiotope-Driven T-B Cell Collaboration in Multiple Sclerosis. Front Immunol 2017; 8:1255. [PMID: 29038659 PMCID: PMC5630699 DOI: 10.3389/fimmu.2017.01255] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2017] [Accepted: 09/20/2017] [Indexed: 12/02/2022] Open
Abstract
Memory B cells acting as antigen-presenting cells are believed to be important in multiple sclerosis (MS), but the antigen they present remains unknown. We hypothesized that B cells may activate CD4+ T cells in the central nervous system of MS patients by presenting idiotopes from their own immunoglobulin variable regions on human leukocyte antigen (HLA) class II molecules. Here, we use bioinformatics prediction analysis of B cell immunoglobulin variable regions from 11 MS patients and 6 controls with other inflammatory neurological disorders (OINDs), to assess whether the prerequisites for such idiotope-driven T–B cell collaboration are present. Our findings indicate that idiotopes from the complementarity determining region (CDR) 3 of MS patients on average have high predicted affinities for disease associated HLA-DRB1*15:01 molecules and are predicted to be endosomally processed by cathepsin S and L in positions that allows such HLA binding to occur. Additionally, complementarity determining region 3 sequences from cerebrospinal fluid (CSF) B cells from MS patients contain on average more rare T cell-exposed motifs that could potentially escape tolerance and stimulate CD4+ T cells than CSF B cells from OIND patients. Many of these features were associated with preferential use of the IGHV4 gene family by CSF B cells from MS patients. This is the first study to combine high-throughput sequencing of patient immune repertoires with large-scale prediction analysis and provides key indicators for future in vitro and in vivo analyses.
Collapse
Affiliation(s)
- Rune A Høglund
- Department of Neurology, Akershus University Hospital, Lørenskog, Norway.,Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Andreas Lossius
- Department of Neurology, Akershus University Hospital, Lørenskog, Norway.,Faculty of Medicine, Department of Immunology and Transfusion Medicine, University of Oslo and Oslo University Hospital Rikshospitalet, Oslo, Norway
| | - Jorunn N Johansen
- Faculty of Medicine, Department of Immunology and Transfusion Medicine, University of Oslo and Oslo University Hospital Rikshospitalet, Oslo, Norway
| | - Jane Homan
- EigenBio LLC, Madison, WI, United States
| | - Jūratė Šaltytė Benth
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway.,Health Services Research Unit, Akershus University Hospital, Lørenskog, Norway
| | - Harlan Robins
- Adaptive Biotechnologies, Seattle, WA, United States
| | - Bjarne Bogen
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway.,Faculty of Medicine, Department of Immunology and Transfusion Medicine, University of Oslo and Oslo University Hospital Rikshospitalet, Oslo, Norway.,Centre for Immune Regulation, University of Oslo, Oslo, Norway
| | | | - Trygve Holmøy
- Department of Neurology, Akershus University Hospital, Lørenskog, Norway.,Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| |
Collapse
|
47
|
Mader S, Brimberg L, Diamond B. The Role of Brain-Reactive Autoantibodies in Brain Pathology and Cognitive Impairment. Front Immunol 2017; 8:1101. [PMID: 28955334 PMCID: PMC5601985 DOI: 10.3389/fimmu.2017.01101] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2017] [Accepted: 08/22/2017] [Indexed: 12/15/2022] Open
Abstract
Antibodies to different brain proteins have been recently found to be associated with an increasing number of different autoimmune diseases. They need to penetrate the blood–brain barrier (BBB) in order to bind antigens within the central nervous system (CNS). They can target either neuronal or non-neuronal antigen and result in damage either by themselves or in synergy with other inflammatory mediators. Antibodies can lead to acute brain pathology, which may be reversible; alternatively, they may trigger irreversible damage that persists even though the antibodies are no longer present. In this review, we will describe two different autoimmune conditions and the role of their antibodies in causing brain pathology. In systemic lupus erythematosus (SLE), patients can have double stranded DNA antibodies that cross react with the neuronal N-methyl-d-aspartate receptor (NMDAR), which have been recently linked to neurocognitive dysfunction. In neuromyelitis optica (NMO), antibodies to astrocytic aquaporin-4 (AQP4) are diagnostic of disease. There is emerging evidence that pathogenic T cells also play an important role for the disease pathogenesis in NMO since they infiltrate in the CNS. In order to enable appropriate and less invasive treatment for antibody-mediated diseases, we need to understand the mechanisms of antibody-mediated pathology, the acute and chronic effects of antibody exposure, if the antibodies are produced intrathecally or systemically, their target antigen, and what triggers their production. Emerging data also show that in utero exposure to some brain-reactive antibodies, such as those found in SLE, can cause neurodevelopmental impairment since they can penetrate the embryonic BBB. If the antibody exposure occurs at a critical time of development, this can result in irreversible damage of the offspring that persists throughout adulthood.
Collapse
Affiliation(s)
- Simone Mader
- The Feinstein Institute for Medical Research, The Center for Autoimmune, Musculoskeletal and Hematopoietic Diseases, Northwell Health System, Manhasset, NY, United States
| | - Lior Brimberg
- The Feinstein Institute for Medical Research, The Center for Autoimmune, Musculoskeletal and Hematopoietic Diseases, Northwell Health System, Manhasset, NY, United States
| | - Betty Diamond
- The Feinstein Institute for Medical Research, The Center for Autoimmune, Musculoskeletal and Hematopoietic Diseases, Northwell Health System, Manhasset, NY, United States
| |
Collapse
|
48
|
Wurth S, Kuenz B, Bsteh G, Ehling R, Di Pauli F, Hegen H, Auer M, Gredler V, Deisenhammer F, Reindl M, Berger T. Cerebrospinal fluid B cells and disease progression in multiple sclerosis - A longitudinal prospective study. PLoS One 2017; 12:e0182462. [PMID: 28777826 PMCID: PMC5544180 DOI: 10.1371/journal.pone.0182462] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2017] [Accepted: 07/18/2017] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND There is evidence that B cells play an important role in disease pathology of multiple sclerosis (MS). The aim of this prospective observational study was to determine the predictive value of cerebrospinal fluid (CSF) B cell subtypes in disease evolution of patients with MS. MATERIALS AND METHODS 128 patients were included between 2004 and 2012. Median follow up time was 7.9 years (range 3.3-10.8 years). 10 patients were lost to follow-up. 32 clinically isolated syndrome- (CIS), 25 relapsing remitting MS- (RRMS), 2 secondary progressive MS- (SPMS) and 9 primary progressive MS- (PPMS) patients were included. The control group consisted of 40 patients with other neurological diseases (OND). CSF samples were analyzed for routine diagnostic parameters. B cell phenotypes were characterized by flow cytometry using CD19 and CD138 specific antibodies. Standardized baseline brain MRI was conducted at the time of diagnostic lumbar puncture. Main outcome variables were likelihood of progressive disease course, EDSS progression, conversion to clinical definite MS (CDMS) and relapse rate. RESULTS CSF mature B cells (CD19+CD138-) were increased in bout-onset MS compared to PPMS (p<0.05) and OND (p<0.001), whereas plasma blasts (CD19+CD138+) were increased in bout-onset MS (p<0.001) and PPMS (p<0.05) compared to OND. CSF B cells did not predict a progressive disease course, EDSS progression, an increased relapse rate or the conversion to CDMS. Likelihood of progressive disease course (p<0.05) and EDSS (p<0.01) was predicted by higher age at baseline, whereas conversion to CDMS was predicted by a lower age at onset (p<0.01) and the presence of ≥9 MRI T2 lesions (p<0.05). CONCLUSION We detected significant differences in the CSF B cell subsets between different clinical MS subtypes and OND patients. CSF B cells were neither predictive for disease and EDSS progression nor conversion to CDMS after a CIS.
Collapse
Affiliation(s)
- Sebastian Wurth
- Clinical Department of Neurology, Medical University of Innsbruck, Innsbruck, Tirol, Austria
- * E-mail:
| | - Bettina Kuenz
- Clinical Department of Neurology, Medical University of Innsbruck, Innsbruck, Tirol, Austria
| | - Gabriel Bsteh
- Clinical Department of Neurology, Medical University of Innsbruck, Innsbruck, Tirol, Austria
| | - Rainer Ehling
- Department of Neurology, Clinic for Rehabilitation Münster, Münster, Tirol, Austria
| | - Franziska Di Pauli
- Clinical Department of Neurology, Medical University of Innsbruck, Innsbruck, Tirol, Austria
| | - Harald Hegen
- Clinical Department of Neurology, Medical University of Innsbruck, Innsbruck, Tirol, Austria
| | - Michael Auer
- Clinical Department of Neurology, Medical University of Innsbruck, Innsbruck, Tirol, Austria
| | - Viktoria Gredler
- Clinical Department of Neurology, Medical University of Innsbruck, Innsbruck, Tirol, Austria
| | - Florian Deisenhammer
- Clinical Department of Neurology, Medical University of Innsbruck, Innsbruck, Tirol, Austria
| | - Markus Reindl
- Clinical Department of Neurology, Medical University of Innsbruck, Innsbruck, Tirol, Austria
| | - Thomas Berger
- Clinical Department of Neurology, Medical University of Innsbruck, Innsbruck, Tirol, Austria
| |
Collapse
|
49
|
Gaudet AD, Fonken LK, Watkins LR, Nelson RJ, Popovich PG. MicroRNAs: Roles in Regulating Neuroinflammation. Neuroscientist 2017; 24:221-245. [PMID: 28737113 DOI: 10.1177/1073858417721150] [Citation(s) in RCA: 167] [Impact Index Per Article: 23.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
MicroRNAs (miRNAs) are small noncoding RNAs that broadly affect cellular and physiological function in all multicellular organisms. Here, the role of miRNAs in neuroinflammation is considered. miRNAs are 21- to 23-oligonucleotide RNAs that regulate translation of specific RNAs by binding to complementary regulatory RNA sequences, thereby causing mRNA degradation or sequestration. More than 5000 miRNAs likely exist in humans, and each miRNA binds an average of 200 RNAs. Specific immunomodulatory miRNAs can regulate a set of RNAs in a coordinated manner, suggesting that effective miRNA-based therapeutic manipulations for neuroinflammatory conditions may be revealed. For instance, miRNAs that preferentially inhibit translation of many cellular anti-inflammatory proteins could drive a pro-inflammatory response. Key pro-inflammatory ( miR-155, miR-27b, miR-326), anti-inflammatory ( miR-124, miR-146a, miR-21, miR-223), and mixed immunomodulatory ( let-7 family) miRNAs regulate neuroinflammation in various pathologies, including spinal cord injury, multiple sclerosis, ischemic stroke, and Alzheimer's disease. miRNAs represent a newly revealed layer of physiological complexity, the therapeutic benefits of which remain to be fully explored and exploited. In this review, we discuss the role of miRNAs in neuroinflammatory regulation and discuss how controlling miRNAs could alter cellular machinery to improve neuroinflammatory dynamics.
Collapse
Affiliation(s)
- Andrew D Gaudet
- 1 Center for Neuroscience, University of Colorado Boulder, CO, USA.,2 Department of Psychology and Neuroscience, University of Colorado Boulder, CO, USA
| | - Laura K Fonken
- 1 Center for Neuroscience, University of Colorado Boulder, CO, USA.,2 Department of Psychology and Neuroscience, University of Colorado Boulder, CO, USA
| | - Linda R Watkins
- 1 Center for Neuroscience, University of Colorado Boulder, CO, USA.,2 Department of Psychology and Neuroscience, University of Colorado Boulder, CO, USA
| | - Randy J Nelson
- 3 Department of Neuroscience, Wexner Medical Center, The Ohio State University, Columbus, OH, USA.,4 Institute for Behavioral Medicine Research, Wexner Medical Center, The Ohio State University, Columbus, OH, USA
| | - Phillip G Popovich
- 3 Department of Neuroscience, Wexner Medical Center, The Ohio State University, Columbus, OH, USA.,4 Institute for Behavioral Medicine Research, Wexner Medical Center, The Ohio State University, Columbus, OH, USA.,5 Center for Brain and Spinal Cord Repair, Wexner Medical Center, The Ohio State University, Columbus, OH, USA
| |
Collapse
|
50
|
A Haystack Heuristic for Autoimmune Disease Biomarker Discovery Using Next-Gen Immune Repertoire Sequencing Data. Sci Rep 2017; 7:5338. [PMID: 28706301 PMCID: PMC5509648 DOI: 10.1038/s41598-017-04439-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2016] [Accepted: 05/16/2017] [Indexed: 01/03/2023] Open
Abstract
Large-scale DNA sequencing of immunological repertoires offers an opportunity for the discovery of novel biomarkers for autoimmune disease. Available bioinformatics techniques however, are not adequately suited for elucidating possible biomarker candidates from within large immunosequencing datasets due to unsatisfactory scalability and sensitivity. Here, we present the Haystack Heuristic, an algorithm customized to computationally extract disease-associated motifs from next-generation-sequenced repertoires by contrasting disease and healthy subjects. This technique employs a local-search graph-theory approach to discover novel motifs in patient data. We apply the Haystack Heuristic to nine million B-cell receptor sequences obtained from nearly 100 individuals in order to elucidate a new motif that is significantly associated with multiple sclerosis. Our results demonstrate the effectiveness of the Haystack Heuristic in computing possible biomarker candidates from high throughput sequencing data and could be generalized to other datasets.
Collapse
|