1
|
Li L, Zhang M, Gu M, Li J, Li Z, Zhang R, Du C, Lv Y. The causal relationship between autoimmune diseases and age-related macular degeneration: A two-sample mendelian randomization study. PLoS One 2024; 19:e0303170. [PMID: 38857222 PMCID: PMC11164335 DOI: 10.1371/journal.pone.0303170] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2023] [Accepted: 04/18/2024] [Indexed: 06/12/2024] Open
Abstract
OBJECTIVE The aim of this study is to investigate the potential causal relationship between autoimmune diseases, including systemic lupus erythematosus, rheumatoid arthritis, inflammatory bowel disease, multiple sclerosis, and Type 1 diabetes, and age-related macular degeneration (AMD). By utilizing the two-sample Mendelian Randomization (MR) approach, we endeavor to address this complex medical issue. METHODS Genome-wide association study (GWAS) data for autoimmune diseases and AMD were obtained from the IEU Open GWAS database and the FinnGen consortium. A series of stringent SNP filtering steps was applied to ensure the reliability of the genetic instruments. MR analyses were conducted using the TwoSampleMR and MR-PRESSO packages in R. The inverse-variance weighted (IVW) method served as the primary analysis, complemented by multiple supplementary analyses and sensitivity tests. RESULTS Within the discovery sample, only a statistically significant inverse causal relationship between multiple sclerosis (MS) and AMD was observed (OR = 0.92, 95% CI: 0.88-0.97, P = 0.003). This finding was confirmed in the replication sample (OR = 0.85, 95% CI: 0.80-0.89, P = 3.32×10-12). No statistically significant associations were detected between systemic lupus erythematosus, rheumatoid arthritis, inflammatory bowel disease, and Type 1 diabetes and AMD. CONCLUSION Strong evidence is provided by this study to support the existence of an inverse causal relationship between multiple sclerosis and age-related macular degeneration. However, no causal evidence was found linking other autoimmune diseases with AMD. These findings not only offer novel insights into the potential etiological mechanisms underlying AMD but also suggest possible directions for future clinical interventions.
Collapse
Affiliation(s)
- Linrui Li
- Department of Ophthalmology, Fushun People’s Hospital, Fushun, China. Sichuan Province, P.R. China
| | - Mingyue Zhang
- Department of Ophthalmology, Fushun People’s Hospital, Fushun, China. Sichuan Province, P.R. China
| | - Moxiu Gu
- Department of Ophthalmology, Fushun People’s Hospital, Fushun, China. Sichuan Province, P.R. China
| | - Jun Li
- Department of Ophthalmology, Fushun People’s Hospital, Fushun, China. Sichuan Province, P.R. China
| | - Zhiyuan Li
- Department of Ophthalmology, Fushun People’s Hospital, Fushun, China. Sichuan Province, P.R. China
| | - Rong Zhang
- Department of Ophthalmology, Fushun People’s Hospital, Fushun, China. Sichuan Province, P.R. China
| | - Chuanwang Du
- Department of Ophthalmology, Fushun People’s Hospital, Fushun, China. Sichuan Province, P.R. China
| | - Yun Lv
- Department of Ophthalmology, Fushun People’s Hospital, Fushun, China. Sichuan Province, P.R. China
| |
Collapse
|
2
|
Mohammadzamani M, Kazemzadeh K, Chand S, Thapa S, Ebrahimi N, Yazdan Panah M, Shaygannejad V, Mirmosayyeb O. Insights into the interplay between Epstein-Barr virus (EBV) and multiple sclerosis (MS): A state-of-the-art review and implications for vaccine development. Health Sci Rep 2024; 7:e1898. [PMID: 38361801 PMCID: PMC10867693 DOI: 10.1002/hsr2.1898] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2023] [Revised: 11/12/2023] [Accepted: 01/15/2024] [Indexed: 02/17/2024] Open
Abstract
Background and Aims Multiple sclerosis (MS) is a chronic autoimmune disease of the central nervous system (CNS). MS results from an inflammatory process leading to the loss of neural tissue and increased disability over time. The role of Epstein Barr Virus (EBV), as one of the most common global viruses, in MS development has been the subject of several studies. However, many related questions are still unanswered. This study aimed to review the connection between MS and EBV and provide a quick outline of MS prevention using EBV vaccination. Methods For this narrative review, an extensive literature search using specific terms was conducted across online databases, including PubMed/Medline, Scopus, Web of Science, and Google Scholar, to identify pertinent studies. Results Several studies proved that almost 100% of people with MS showed a history of EBV infection, and there was an association between high titers of EBV antibodies and an increased risk of MS development. Various hypotheses are proposed for how EBV may contribute to MS directly and indirectly: (1) Molecular Mimicry, (2) Mistaken Self, (3) Bystander Damage, and (4) Autoreactive B cells infected with EBV. Conclusion Given the infectious nature of EBV and its ability to elude the immune system, EBV emerges as a strong candidate for being the underlying cause of MS. The development of an EBV vaccine holds promise for preventing MS; however, overcoming the challenge of creating a safe and efficacious vaccine presents a significant obstacle.
Collapse
Affiliation(s)
- Mahtab Mohammadzamani
- Isfahan Neurosciences Research CenterIsfahan University of Medical SciencesIsfahanIran
| | - Kimia Kazemzadeh
- Students' Scientific Research CenterTehran University of Medical SciencesTehranIran
| | - Swati Chand
- Westchester Medical CenterNew York Medical CollegeValhallaNew YorkUSA
| | - Sangharsha Thapa
- Department of Neurology, Westchester Medical CenterNew York Medical CollegeValhallaUSA
| | - Narges Ebrahimi
- Isfahan Neurosciences Research CenterIsfahan University of Medical SciencesIsfahanIran
| | | | - Vahid Shaygannejad
- Isfahan Neurosciences Research CenterIsfahan University of Medical SciencesIsfahanIran
- Department of NeurologyIsfahan University of Medical SciencesIsfahanIran
| | - Omid Mirmosayyeb
- Isfahan Neurosciences Research CenterIsfahan University of Medical SciencesIsfahanIran
- Department of NeurologyIsfahan University of Medical SciencesIsfahanIran
| |
Collapse
|
3
|
Kalkowski L, Walczak P, Mycko MP, Malysz-Cymborska I. Reconsidering the route of drug delivery in refractory multiple sclerosis: Toward a more effective drug accumulation in the central nervous system. Med Res Rev 2023; 43:2237-2259. [PMID: 37203228 DOI: 10.1002/med.21973] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2022] [Revised: 03/08/2023] [Accepted: 04/30/2023] [Indexed: 05/20/2023]
Abstract
Multiple sclerosis is a chronic demyelinating disease with different disease phenotypes. The current FDA-approved disease-modifying therapeutics (DMTs) cannot cure the disease, but only alleviate the disease progression. While the majority of patients respond well to treatment, some of them are suffering from rapid progression. Current drug delivery strategies include the oral, intravenous, subdermal, and intramuscular routes, so these drugs are delivered systemically, which is appropriate when the therapeutic targets are peripheral. However, the potential benefits may be diminished when these targets sequester behind the barriers of the central nervous system. Moreover, systemic drug administration is plagued with adverse effects, sometimes severe. In this context, it is prudent to consider other drug delivery strategies improving their accumulation in the brain, thus providing better prospects for patients with rapidly progressing disease course. These targeted drug delivery strategies may also reduce the severity of systemic adverse effects. Here, we discuss the possibilities and indications for reconsideration of drug delivery routes (especially for those "non-responding" patients) and the search for alternative drug delivery strategies. More targeted drug delivery strategies sometimes require quite invasive procedures, but the potential therapeutic benefits and reduction of adverse effects could outweigh the risks. We characterized the major FDA-approved DMTs focusing on their therapeutic mechanism and the potential benefits of improving the accumulation of these drugs in the brain.
Collapse
Affiliation(s)
- Lukasz Kalkowski
- Department of Diagnostic Radiology and Nuclear Medicine, Center for Advanced Imaging Research, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Piotr Walczak
- Department of Diagnostic Radiology and Nuclear Medicine, Center for Advanced Imaging Research, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Marcin P Mycko
- Medical Division, Department of Neurology, Laboratory of Neuroimmunology, Collegium Medicum, University of Warmia and Mazury, Olsztyn, Poland
| | - Izabela Malysz-Cymborska
- Department of Neurosurgery, School of Medicine, Collegium Medicum, University of Warmia and Mazury, Olsztyn, Poland
| |
Collapse
|
4
|
Khakdan F, Javanmard AS, Shahmoradipour P, Jahromi MJ. The fluctuations of expression profiles of critical genes in the miRNA maturation process and pro-and anti-inflammatory cytokines in the pathogenesis and progression of multiple sclerosis. Mol Biol Rep 2023; 50:9405-9416. [PMID: 37823932 DOI: 10.1007/s11033-023-08812-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Accepted: 09/11/2023] [Indexed: 10/13/2023]
Abstract
BACKGROUND Multiple sclerosis (MS) is a central nervous system disease known for immune-mediated demyelination, inflammatory, and neurodegeneration symptoms. Discovering molecular biomarkers to classify RRMS and SPMS patients, monitor the disease activity, and response to particular treatments is one area that has received notable attraction. MicroRNA (miRNA), a single-stranded non-coding RNA molecule, is a significant regulator of gene expression recruited in pathogenic mechanisms in diverse diseases, especially cancer and MS. Also, the relapsing-remitting features of MS exhibit that both inflammatory and anti-inflammatory cytokines are effective in the progression of the disease over time. METHODS AND RESULTS It was assessed the expression patterns of the genes (Drosha, Pasha (DGCR8), and Dicer ) encoding the critical enzymes in the processing steps of miRNA maturation and major pro-inflammatory and anti-inflammatory cytokines (IFN-α, IFN-β, and IL-6) in blood cells of 40 MS patients (two groups of 10 men and women in both clinical courses of RR and SPMS patients) in comparison with 20 healthy control group (10 males and 10 females). The highest transcription activity of Drosha was observed for RRMS patients (4.2 and 3.6-fold, respectively), and the expression ratio was down regulated in male and female patients with SPMS (3.9- and 3.1-fold, respectively). Considering the studied cytokines, the increase in expression ratio of IL-6 in SPMS patients and the decrease in transcript abundance of INF-α, and INF-β cytokines are consistent with the progression of the disease. CONCLUSIONS Our findings showed that the high and low transcriptional levels of the considered genes seem to be effective in the pathogenesis and progression of MS.
Collapse
Affiliation(s)
- Fatemeh Khakdan
- Department of Biology, Farzanegan Campus, Semnan University, Semnan, Iran
| | | | - Parisa Shahmoradipour
- Department of Biotechnology, Institute of Science, High Technology and Environmental Sciences, Graduate University of Advanced Technology, Kerman, Iran
| | | |
Collapse
|
5
|
Londoño AC, Mora CA. Continued dysregulation of the B cell lineage promotes multiple sclerosis activity despite disease modifying therapies. F1000Res 2023; 10:1305. [PMID: 37655229 PMCID: PMC10467621 DOI: 10.12688/f1000research.74506.2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 07/27/2023] [Indexed: 09/02/2023] Open
Abstract
A clear understanding of the origin and role of the different subtypes of the B cell lineage involved in the activity or remission of multiple sclerosis (MS) is important for the treatment and follow-up of patients living with this disease. B cells, however, are dynamic and can play an anti-inflammatory or pro-inflammatory role, depending on their milieu. Depletion of B cells has been effective in controlling the progression of MS, but it can have adverse side effects. A better understanding of the role of the B cell subtypes, through the use of surface biomarkers of cellular activity with special attention to the function of memory and other regulatory B cells (Bregs), will be necessary in order to offer specific treatments without inducing undesirable effects.
Collapse
Affiliation(s)
- Ana C. Londoño
- Neurologia y Neuroimagen, Instituto Neurologico de Colombia (INDEC), Medellin, Antioquia, Colombia
| | - Carlos A. Mora
- Spine & Brain Institute, Ascension St. Vincent's Riverside Hospital, Jacksonville, FL, 32204, USA
| |
Collapse
|
6
|
Zettl UK, Rommer PS, Aktas O, Wagner T, Richter J, Oschmann P, Cepek L, Elias-Hamp B, Gehring K, Chan A, Hecker M. Interferon beta-1a sc at 25 years: a mainstay in the treatment of multiple sclerosis over the period of one generation. Expert Rev Clin Immunol 2023; 19:1343-1359. [PMID: 37694381 DOI: 10.1080/1744666x.2023.2248391] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2023] [Accepted: 08/10/2023] [Indexed: 09/12/2023]
Abstract
INTRODUCTION Interferon beta (IFN beta) preparations are an established group of drugs used for immunomodulation in patients with multiple sclerosis (MS). Subcutaneously (sc) applied interferon beta-1a (IFN beta-1a sc) has been in continuous clinical use for 25 years as a disease-modifying treatment. AREAS COVERED Based on data published since 2018, we discuss recent insights from analyses of the pivotal trial PRISMS and its long-term extension as well as from newer randomized studies with IFN beta-1a sc as the reference treatment, the use of IFN beta-1a sc across the patient life span and as a bridging therapy, recent data regarding the mechanisms of action, and potential benefits of IFN beta-1a sc regarding vaccine responses. EXPERT OPINION IFN beta-1a sc paved the way to effective immunomodulatory treatment of MS, enabled meaningful insights into the disease process, and remains a valid therapeutic option in selected vulnerable MS patient groups.
Collapse
Affiliation(s)
- Uwe Klaus Zettl
- Department of Neurology, Rostock University Medical Center, Rostock, Germany
| | - Paulus Stefan Rommer
- Department of Neurology, Rostock University Medical Center, Rostock, Germany
- Department of Neurology, Medical University of Vienna, Vienna, Austria
| | - Orhan Aktas
- Department of Neurology, Medical Faculty, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | | | | | | | | | | | | | - Andrew Chan
- Department of Neurology, Inselspital Bern, University Hospital Bern, Bern, Switzerland
| | - Michael Hecker
- Department of Neurology, Rostock University Medical Center, Rostock, Germany
| |
Collapse
|
7
|
Bellucci G, Albanese A, Rizzi C, Rinaldi V, Salvetti M, Ristori G. The value of Interferon β in multiple sclerosis and novel opportunities for its anti-viral activity: a narrative literature review. Front Immunol 2023; 14:1161849. [PMID: 37334371 PMCID: PMC10275407 DOI: 10.3389/fimmu.2023.1161849] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2023] [Accepted: 05/24/2023] [Indexed: 06/20/2023] Open
Abstract
Interferon-beta (IFN-β) for Multiple Sclerosis (MS) is turning 30. The COVID-19 pandemic rejuvenated the interest in interferon biology in health and disease, opening translational opportunities beyond neuroinflammation. The antiviral properties of this molecule are in accord with the hypothesis of a viral etiology of MS, for which a credible culprit has been identified in the Epstein-Barr Virus. Likely, IFNs are crucial in the acute phase of SARS-CoV-2 infection, as demonstrated by inherited and acquired impairments of the interferon response that predispose to a severe COVID-19 course. Accordingly, IFN-β exerted protection against SARS-CoV-2 in people with MS (pwMS). In this viewpoint, we summarize the evidence on IFN-β mechanisms of action in MS with a focus on its antiviral properties, especially against EBV. We synopsize the role of IFNs in COVID-19 and the opportunities and challenges of IFN-β usage for this condition. Finally, we leverage the lessons learned in the pandemic to suggest a role of IFN-β in long-COVID-19 and in special MS subpopulations.
Collapse
Affiliation(s)
- Gianmarco Bellucci
- Department of Neurosciences, Mental Health and Sensory Organs, Centre for Experimental Neurological Therapies (CENTERS), Sapienza University of Rome, Rome, Italy
| | - Angela Albanese
- Merck Serono S.p.A., An Affiliate of Merck KGaA, Rome, Italy
- Department of Health Sciences, University of Genoa, Genoa, Italy
| | - Caterina Rizzi
- Merck Serono S.p.A., An Affiliate of Merck KGaA, Rome, Italy
| | - Virginia Rinaldi
- Department of Neurosciences, Mental Health and Sensory Organs, Centre for Experimental Neurological Therapies (CENTERS), Sapienza University of Rome, Rome, Italy
| | - Marco Salvetti
- Department of Neurosciences, Mental Health and Sensory Organs, Centre for Experimental Neurological Therapies (CENTERS), Sapienza University of Rome, Rome, Italy
- Istituti di Ricovero e Cura a Carattere Scientifico (IRCCS) Istituto Neurologico Mediterraneo Neuromed, Pozzilli, Italy
| | - Giovanni Ristori
- Department of Neurosciences, Mental Health and Sensory Organs, Centre for Experimental Neurological Therapies (CENTERS), Sapienza University of Rome, Rome, Italy
- Neuroimmunology Unit, Istituti di Ricovero e Cura a Carattere Scientifico (IRCCS) Fondazione Santa Lucia, Rome, Italy
| |
Collapse
|
8
|
Xavier A, Campagna MP, Maltby VE, Kilpatrick T, Taylor BV, Butzkueven H, Ponsonby AL, Scott RJ, Jokubaitis VG, Lea RA, Lechner-Scott J. Interferon beta treatment is a potent and targeted epigenetic modifier in multiple sclerosis. Front Immunol 2023; 14:1162796. [PMID: 37325639 PMCID: PMC10266220 DOI: 10.3389/fimmu.2023.1162796] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2023] [Accepted: 05/12/2023] [Indexed: 06/17/2023] Open
Abstract
Introduction Multiple Sclerosis (MS) has a complex pathophysiology that involves genetic and environmental factors. DNA methylation (DNAm) is one epigenetic mechanism that can reversibly modulate gene expression. Cell specific DNAm changes have been associated with MS, and some MS therapies such as dimethyl fumarate can influence DNAm. Interferon Beta (IFNβ), was one of the first disease modifying therapies in multiple sclerosis (MS). However, how IFNβ reduces disease burden in MS is not fully understood and little is known about the precise effect of IFNβ treatment on methylation. Methods The objective of this study was to determine the changes in DNAm associated with INFβ use, using methylation arrays and statistical deconvolutions on two separate datasets (total ntreated = 64, nuntreated = 285). Results We show that IFNβ treatment in people with MS modifies the methylation profile of interferon response genes in a strong, targeted, and reproducible manner. Using these identified methylation differences, we constructed a methylation treatment score (MTS) that is an accurate discriminator between untreated and treated patients (Area under the curve = 0.83). This MTS is time-sensitive and in consistent with previously identified IFNβ treatment therapeutic lag. This suggests that methylation changes are required for treatment efficacy. Overrepresentation analysis found that IFNβ treatment recruits the endogenous anti-viral molecular machinery. Finally, statistical deconvolution revealed that dendritic cells and regulatory CD4+ T cells were most affected by IFNβ induced methylation changes. Discussion In conclusion, our study shows that IFNβ treatment is a potent and targeted epigenetic modifier in multiple sclerosis.
Collapse
Affiliation(s)
- Alexandre Xavier
- School of Biomedical Science and Pharmacy, University of Newcastle, Newcastle, NSW, Australia
| | - Maria Pia Campagna
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, VIC, Australia
| | - Vicki E. Maltby
- Hunter Medical Research Institute, Immune Health research program, Newcastle, NSW, Australia
- Department of Neurology, John Hunter Hospital, Newcastle, NSW, Australia
- School of Medicine and Public Health, University of Newcastle, Newcastle, NSW, Australia
| | - Trevor Kilpatrick
- Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Parkville, VIC, Australia
| | - Bruce V. Taylor
- Menzies Institute for Medical Research, University of Tasmania, Hobart, TAS, Australia
| | - Helmut Butzkueven
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, VIC, Australia
- Neuro-Immunology Registry, MSBase Foundation, Melbourne, VIC, Australia
| | - Anne-Louise Ponsonby
- Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Parkville, VIC, Australia
| | - Rodney J. Scott
- School of Biomedical Science and Pharmacy, University of Newcastle, Newcastle, NSW, Australia
- New South Wales (NSW) Health Pathology, John Hunter Hospital, Newcastle, NSW, Australia
| | - Vilija G. Jokubaitis
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, VIC, Australia
| | - Rodney A. Lea
- School of Biomedical Science and Pharmacy, University of Newcastle, Newcastle, NSW, Australia
- Centre of Genomics and Personalised Health, School of Biomedical Sciences, Queensland University of Technology, Kelvin Grove, QLD, Australia
| | - Jeannette Lechner-Scott
- Hunter Medical Research Institute, Immune Health research program, Newcastle, NSW, Australia
- Department of Neurology, John Hunter Hospital, Newcastle, NSW, Australia
- School of Medicine and Public Health, University of Newcastle, Newcastle, NSW, Australia
| |
Collapse
|
9
|
Aloisi F, Giovannoni G, Salvetti M. Epstein-Barr virus as a cause of multiple sclerosis: opportunities for prevention and therapy. Lancet Neurol 2023; 22:338-349. [PMID: 36764322 DOI: 10.1016/s1474-4422(22)00471-9] [Citation(s) in RCA: 46] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Revised: 10/27/2022] [Accepted: 11/04/2022] [Indexed: 02/10/2023]
Abstract
Multiple sclerosis is a chronic inflammatory disease of the CNS that results from the interplay between heritable and environmental factors. Mounting evidence from different fields of research supports the pivotal role of the Epstein-Barr virus (EBV) in the development of multiple sclerosis. However, translating this knowledge into clinically actionable information requires a better understanding of the mechanisms linking EBV to pathophysiology. Ongoing research is trying to clarify whether EBV causes neuroinflammation via autoimmunity or antiviral immunity, and if the interaction of EBV with genetic susceptibility to multiple sclerosis can explain why a ubiquitous virus promotes immune dysfunction in susceptible individuals. If EBV also has a role in driving disease activity, the characterisation of this role will help diagnosis, prognosis, and treatment in people with multiple sclerosis. Ongoing clinical trials targeting EBV and new anti-EBV vaccines provide hope for future treatments and preventive interventions.
Collapse
Affiliation(s)
- Francesca Aloisi
- Department of Neuroscience, Istituto Superiore di Sanità, Rome, Italy.
| | - Gavin Giovannoni
- Preventive Neurology Unit, Wolfson Institute of Preventive Medicine and Blizard Institute, Queen Mary University, London, UK
| | - Marco Salvetti
- Department of Neurosciences, Mental Health and Sensory Organs, Sapienza University of Rome, Rome, Italy; IRCCS Neuromed, Pozzilli, Italy
| |
Collapse
|
10
|
Smith C, Khanna R. Adoptive T-cell therapy targeting Epstein-Barr virus as a treatment for multiple sclerosis. Clin Transl Immunology 2023; 12:e1444. [PMID: 36960148 PMCID: PMC10028422 DOI: 10.1002/cti2.1444] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Revised: 03/06/2023] [Accepted: 03/07/2023] [Indexed: 03/23/2023] Open
Abstract
Emergence of a definitive link between Epstein-Barr virus (EBV) and multiple sclerosis has provided an impetus to develop immune-based therapies to target EBV-infected B cells. Initial studies with autologous EBV-specific T-cell therapy demonstrated that this therapy is safe with minimal side effects and more importantly multiple patients showed both symptomatic and objective neurological improvements including improved quality of life, reduction of fatigue and reduced intrathecal IgG production. These observations have been successfully extended to an 'off-the-shelf' allogeneic EBV-specific T-cell therapy manufactured using peripheral blood lymphocytes of healthy seropositive individuals. This adoptive immunotherapy has also been shown to be safe with encouraging clinical responses. Allogeneic EBV T-cell therapy overcomes some of the limitations of autologous therapy and can be rapidly delivered to patients with improved therapeutic potential.
Collapse
Affiliation(s)
- Corey Smith
- QIMR Berghofer Centre for Immunotherapy and Vaccine Development, Infection and Inflammation ProgramQIMR Berghofer Medical Research InstituteHerstonQLDAustralia
| | - Rajiv Khanna
- QIMR Berghofer Centre for Immunotherapy and Vaccine Development, Infection and Inflammation ProgramQIMR Berghofer Medical Research InstituteHerstonQLDAustralia
| |
Collapse
|
11
|
The BAFF-APRIL System in Cancer. Cancers (Basel) 2023; 15:cancers15061791. [PMID: 36980677 PMCID: PMC10046288 DOI: 10.3390/cancers15061791] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2023] [Revised: 03/13/2023] [Accepted: 03/13/2023] [Indexed: 03/17/2023] Open
Abstract
B cell-activating factor (BAFF; also known as CD257, TNFSF13B, BLyS) and a proliferation-inducing ligand (APRIL; also known as CD256, TNFSF13) belong to the tumor necrosis factor (TNF) family. BAFF was initially discovered as a B-cell survival factor, whereas APRIL was first identified as a protein highly expressed in various cancers. These discoveries were followed by over two decades of extensive research effort, which identified overlapping signaling cascades between BAFF and APRIL, controlling immune homeostasis in health and driving pathogenesis in autoimmunity and cancer, the latter being the focus of this review. High levels of BAFF, APRIL, and their receptors have been detected in different cancers and found to be associated with disease severity and treatment response. Here, we have summarized the role of the BAFF-APRIL system in immune cell differentiation and immune tolerance and detailed its pathogenic functions in hematological and solid cancers. We also highlight the emerging therapeutics targeting the BAFF-APRIL system in different cancer types.
Collapse
|
12
|
Dyer Z, Tscharke D, Sutton I, Massey J. From bedside to bench: how existing therapies inform the relationship between Epstein-Barr virus and multiple sclerosis. Clin Transl Immunology 2023; 12:e1437. [PMID: 36844913 PMCID: PMC9947628 DOI: 10.1002/cti2.1437] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Revised: 01/19/2023] [Accepted: 01/20/2023] [Indexed: 02/25/2023] Open
Abstract
Therapy for relapsing-remitting multiple sclerosis (MS) has advanced dramatically despite incomplete understanding of the cause of the condition. Current treatment involves inducing broad effects on immune cell populations with consequent off-target side effects, and no treatment can completely prevent disability progression. Further therapeutic advancement will require a better understanding of the pathobiology of MS. Interest in the role of Epstein-Barr virus (EBV) in multiple sclerosis has intensified based on strong epidemiological evidence of an association between EBV seroprevalence and MS. Hypotheses proposed to explain the biological relationship between EBV and MS include molecular mimicry, EBV immortalised autoreactive B cells and infection of glial cells by EBV. Examining the interaction between EBV and immunotherapies that have demonstrated efficacy in MS offers clues to the validity of these hypotheses. The efficacy of B cell depleting therapies could be consistent with a hypothesis that EBV-infected B cells drive MS; however, loss of T cell control of B cells does not exacerbate MS. A number of MS therapies invoke change in EBV-specific T cell populations, but pathogenic EBV-specific T cells with cross-reactivity to CNS antigen have not been identified. Immune reconstitution therapies induce EBV viraemia and expansion of EBV-specific T cell clones, but this does not correlate with relapse. Much remains unknown regarding the role of EBV in MS pathogenesis. We discuss future translational research that could fill important knowledge gaps.
Collapse
Affiliation(s)
- Zoe Dyer
- Blood Stem Cell and Cancer Research Group, St Vincent's Centre for Applied Medical ResearchDarlinghurstNSWAustralia,St. Vincent's Clinical School, Faculty of MedicineUniversity of New South Wales (UNSW)DarlinghurstNSWAustralia
| | - David Tscharke
- John Curtin School of Medical ResearchAustralian National UniversityCanberraACTAustralia
| | - Ian Sutton
- St. Vincent's Clinical School, Faculty of MedicineUniversity of New South Wales (UNSW)DarlinghurstNSWAustralia,Department of NeurologySt Vincent's ClinicDarlinghurstNSWAustralia
| | - Jennifer Massey
- Blood Stem Cell and Cancer Research Group, St Vincent's Centre for Applied Medical ResearchDarlinghurstNSWAustralia,St. Vincent's Clinical School, Faculty of MedicineUniversity of New South Wales (UNSW)DarlinghurstNSWAustralia,Department of NeurologySt Vincent's ClinicDarlinghurstNSWAustralia,Department of NeurologySt Vincent's HospitalDarlinghurstNSWAustralia
| |
Collapse
|
13
|
Interferon β1a treatment does not influence serum Epstein-Barr virus antibodies in patients with multiple sclerosis. Mult Scler Relat Disord 2023; 70:104530. [PMID: 36701908 DOI: 10.1016/j.msard.2023.104530] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Accepted: 01/18/2023] [Indexed: 01/22/2023]
Abstract
There is increasing evidence of Epstein-Barr virus (EBV) being conditional in multiple sclerosis (MS) pathogenesis and influential for disease activity. Interferon-beta (IFNβ) is a cytokine with antiviral effects used to treat MS, in which a possible antiviral effect against EBV has been questioned. In this study, we investigated the effect of IFNβ-1a treatment on serum EBV antibody levels in 84 patients with relapsing-remitting MS. In the 18 months following IFNβ-1a treatment initiation, there were no significant associations between treatment and serum levels of Epstein-Barr nuclear antigen 1 (EBNA-1) immunoglobulin (Ig) G, early antigen (EA) IgG, viral capsid antigen (VCA) IgG or VCA IgM. The findings suggest that IFNβ-1a treatment does not influence the humoral response to EBV in patients with MS.
Collapse
|
14
|
Kumar G, Axtell RC. Dual Role of B Cells in Multiple Sclerosis. Int J Mol Sci 2023; 24:2336. [PMID: 36768658 PMCID: PMC9916779 DOI: 10.3390/ijms24032336] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Revised: 01/12/2023] [Accepted: 01/21/2023] [Indexed: 01/26/2023] Open
Abstract
B cells have emerged as an important immune cell type that can be targeted for therapy in multiple sclerosis (MS). Depleting B cells with anti-CD20 antibodies is effective in treating MS. Yet, atacicept treatment, which blocks B-cell Activating Factor (BAFF) and A Proliferation-Inducing Ligand (APRIL), two cytokines important for B cell development and function, paradoxically increases disease activity in MS patients. The reason behind the failure of atacicept is not well understood. The stark differences in clinical outcomes with these therapies demonstrate that B cells have both inflammatory and anti-inflammatory functions in MS. In this review, we summarize the importance of B cells in MS and discuss the different B cell subsets that perform inflammatory and anti-inflammatory functions and how therapies modulate B cell functions in MS patients. Additionally, we discuss the potential anti-inflammatory functions of BAFF and APRIL on MS disease.
Collapse
Affiliation(s)
| | - Robert C. Axtell
- Arthritis and Clinical Immunology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK 73104, USA
| |
Collapse
|
15
|
Attfield KE, Jensen LT, Kaufmann M, Friese MA, Fugger L. The immunology of multiple sclerosis. Nat Rev Immunol 2022; 22:734-750. [PMID: 35508809 DOI: 10.1038/s41577-022-00718-z] [Citation(s) in RCA: 128] [Impact Index Per Article: 42.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/30/2022] [Indexed: 12/11/2022]
Abstract
Our incomplete understanding of the causes and pathways involved in the onset and progression of multiple sclerosis (MS) limits our ability to effectively treat this complex neurological disease. Recent studies explore the role of immune cells at different stages of MS and how they interact with cells of the central nervous system (CNS). The findings presented here begin to question the exclusivity of an antigen-specific cause and highlight how seemingly distinct immune cell types can share common functions that drive disease. Innovative techniques further expose new disease-associated immune cell populations and reinforce how environmental context is critical to their phenotype and subsequent role in disease. Importantly, the differentiation of immune cells into a pathogenic state is potentially reversible through therapeutic manipulation. As such, understanding the mechanisms that provide plasticity to causal cell types is likely key to uncoupling these disease processes and may identify novel therapeutic targets that replace the need for cell ablation.
Collapse
Affiliation(s)
- Kathrine E Attfield
- Oxford Centre for Neuroinflammation, Nuffield Department of Clinical Neurosciences, Oxford University Hospitals, University of Oxford, Oxford, UK
| | - Lise Torp Jensen
- Department of Clinical Medicine, Aarhus University Hospital, Aarhus, Denmark
| | - Max Kaufmann
- Institut für Neuroimmunologie und Multiple Sklerose, Zentrum für Molekulare Neurobiologie Hamburg, Universitätsklinikum Hamburg-Eppendorf, Hamburg, Germany
| | - Manuel A Friese
- Institut für Neuroimmunologie und Multiple Sklerose, Zentrum für Molekulare Neurobiologie Hamburg, Universitätsklinikum Hamburg-Eppendorf, Hamburg, Germany
| | - Lars Fugger
- Oxford Centre for Neuroinflammation, Nuffield Department of Clinical Neurosciences, Oxford University Hospitals, University of Oxford, Oxford, UK.
- MRC Human Immunology Unit, Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, UK.
| |
Collapse
|
16
|
Leffler J, Trend S, Hart PH, French MA. Epstein-Barr virus infection, B-cell dysfunction and other risk factors converge in gut-associated lymphoid tissue to drive the immunopathogenesis of multiple sclerosis: a hypothesis. Clin Transl Immunology 2022; 11:e1418. [PMID: 36325491 PMCID: PMC9621333 DOI: 10.1002/cti2.1418] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2022] [Revised: 09/12/2022] [Accepted: 09/13/2022] [Indexed: 11/10/2022] Open
Abstract
Multiple sclerosis is associated with Epstein-Barr virus (EBV) infection, B-cell dysfunction, gut dysbiosis, and environmental and genetic risk factors, including female sex. A disease model incorporating all these factors remains elusive. Here, we hypothesise that EBV-infected memory B cells (MBCs) migrate to gut-associated lymphoid tissue (GALT) through EBV-induced expression of LPAM-1, where they are subsequently activated by gut microbes and/or their products resulting in EBV reactivation and compartmentalised anti-EBV immune responses. These responses involve marginal zone (MZ) B cells that activate CD4+ T-cell responses, via HLA-DRB1, which promote downstream B-cell differentiation towards CD11c+/T-bet+ MBCs, as well as conventional MBCs. Intrinsic expression of low-affinity B-cell receptors (BCRs) by MZ B cells and CD11c+/T-bet+ MBCs promotes polyreactive BCR/antibody responses against EBV proteins (e.g. EBNA-1) that cross-react with central nervous system (CNS) autoantigens (e.g. GlialCAM). EBV protein/autoantigen-specific CD11c+/T-bet+ MBCs migrate to the meningeal immune system and CNS, facilitated by their expression of CXCR3, and induce cytotoxic CD8+ T-cell responses against CNS autoantigens amplified by BAFF, released from EBV-infected MBCs. An increased abundance of circulating IgA+ MBCs, observed in MS patients, might also reflect GALT-derived immune responses, including disease-enhancing IgA antibody responses against EBV and gut microbiota-specific regulatory IgA+ plasma cells. Female sex increases MZ B-cell and CD11c+/T-bet+ MBC activity while environmental risk factors affect gut dysbiosis. Thus, EBV infection, B-cell dysfunction and other risk factors converge in GALT to generate aberrant B-cell responses that drive pathogenic T-cell responses in the CNS.
Collapse
Affiliation(s)
- Jonatan Leffler
- Telethon Kids InstituteUniversity of Western AustraliaPerthWAAustralia
| | - Stephanie Trend
- Telethon Kids InstituteUniversity of Western AustraliaPerthWAAustralia,Perron Institute for Neurological and Translational ScienceUniversity of Western AustraliaPerthWAAustralia
| | - Prue H Hart
- Telethon Kids InstituteUniversity of Western AustraliaPerthWAAustralia
| | - Martyn A French
- School of Biomedical SciencesUniversity of Western AustraliaPerthWAAustralia,Immunology DivisionPathWest Laboratory MedicinePerthWAAustralia
| |
Collapse
|
17
|
Saitakis G, Chwalisz BK. Treatment and Relapse Prevention of Typical and Atypical Optic Neuritis. Int J Mol Sci 2022; 23:9769. [PMID: 36077167 PMCID: PMC9456305 DOI: 10.3390/ijms23179769] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2022] [Revised: 08/17/2022] [Accepted: 08/18/2022] [Indexed: 12/02/2022] Open
Abstract
Optic neuritis (ON) is an inflammatory condition involving the optic nerve. Several important typical and atypical ON variants are now recognized. Typical ON has a more favorable prognosis; it can be idiopathic or represent an early manifestation of demyelinating diseases, mostly multiple sclerosis (MS). The atypical spectrum includes entities such as antibody-driven ON associated with neuromyelitis optica spectrum disorder (NMOSD) and myelin oligodendrocyte glycoprotein antibody disease (MOGAD), chronic/relapsing inflammatory optic neuropathy (CRION), and sarcoidosis-associated ON. Appropriate and timely diagnosis is essential to rapidly decide on the appropriate treatment, maximize visual recovery, and minimize recurrences. This review paper aims at presenting the currently available state-of-the-art treatment strategies for typical and atypical ON, both in the acute phase and in the long-term. Moreover, emerging therapeutic approaches and novel steps in the direction of achieving remyelination are discussed.
Collapse
Affiliation(s)
- George Saitakis
- Division of Neuro-Ophthalmology, Department of Ophthalmology, Massachusetts Eye & Ear Infirmary, Harvard Medical School, Boston, MA 02115, USA
- Athens Eye Hospital, 166 75 Athens, Greece
| | - Bart K. Chwalisz
- Division of Neuro-Ophthalmology, Department of Ophthalmology, Massachusetts Eye & Ear Infirmary, Harvard Medical School, Boston, MA 02115, USA
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, 15 Parkman Street, Suite 835, Boston, MA 02114, USA
| |
Collapse
|
18
|
Serum levels of IgM to phosphatidylcholine predict the response of multiple sclerosis patients to natalizumab or IFN-β. Sci Rep 2022; 12:13357. [PMID: 35922641 PMCID: PMC9349316 DOI: 10.1038/s41598-022-16218-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2022] [Accepted: 07/06/2022] [Indexed: 11/09/2022] Open
Abstract
We developed an ELISA assay demonstrating the high prevalence of serum IgM to phosphatidylcholine (IgM-PC) in the first stages of multiple sclerosis (MS). We aimed to analyze the role of serum IgM-PC as a biomarker of response to treatment. Paired serum samples from 95 MS patients were obtained before (b.t) and after (a.t) treatment with disease modifying therapies. Patients were classified as non-responders or responders to treatment, according to classical criteria. Serum IgM-PC concentration was analyzed using our house ELISA assay. The level of serum IgM-PC b.t was higher in patients treated later with natalizumab than in those treated with Copaxone (p = 0.011) or interferon-β (p = 0.009). Responders to natalizumab showed higher concentration of serum IgM-PC b.t than those who did not respond to it (p = 0.019). The 73.3% of patients with the highest level of serum IgM-PC b.t responded to natalizumab. IgM-PC level decreased a.t in both cases, non-responders and responders to natalizumab. IgM-PC levels a.t did not decrease in non-responders to interferon-β, but in responders to it the IgM-PC level decreased (p = 0.007). Serum IgM-PC could be a biomarker of response to natalizumab or interferon-β treatment. Further studies would be necessary to validate these results.
Collapse
|
19
|
Márquez AC, Croft C, Shanina I, Horwitz MS. Influence of Type I Interferons in Gammaherpesvirus-68 and Its Influence on EAE Enhancement. Front Immunol 2022; 13:858583. [PMID: 35874728 PMCID: PMC9301468 DOI: 10.3389/fimmu.2022.858583] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Accepted: 06/14/2022] [Indexed: 11/18/2022] Open
Abstract
Epstein-Barr virus (EBV) has been identified as a putative trigger of multiple sclerosis (MS). Previously, we reported that mice latently infected with murine gammaherpesvirus 68 (γHV-68), the murine homolog to EBV, and induced for experimental autoimmune encephalomyelitis (EAE), developed an enhanced disease more reminiscent of MS. These prior results showed that expression of CD40 on CD11b+CD11c+ cells in latently infected mice was required to prime the strong Th1 response driving disease as well as decreasing Treg frequencies in the periphery and CNS. Subsequent work demonstrated that transfer of B cells from latently infected mice was sufficient to enhance disease. Herein, we show that B cells from infected mice do not need type I IFN signaling to drive a strong Th1 response, yet are important in driving infiltration of the CNS by CD8+ T cells. Given the importance of type I IFNs in MS, we used IFNARko mice in order to determine if type I IFN signaling was important in the enhancement of EAE in latently infected mice. We found that while type I IFNs are important for the control of γHV-68 infection and maintenance of latency, they do not have a direct effect in the development of enhanced EAE.
Collapse
Affiliation(s)
- Ana Citlali Márquez
- Department of Microbiology and Immunology, University of British Columbia, Vancouver, BC, Canada
- BC Centre for Disease Control, University of British Columbia, Vancouver, BC, Canada
| | - Carys Croft
- Innate Immunity Unit, Institut Pasteur, Inserm U1223, Paris, France
| | - Iryna Shanina
- Department of Microbiology and Immunology, University of British Columbia, Vancouver, BC, Canada
| | - Marc Steven Horwitz
- Department of Microbiology and Immunology, University of British Columbia, Vancouver, BC, Canada
- *Correspondence: Marc Steven Horwitz,
| |
Collapse
|
20
|
Dumitrescu L, Papathanasiou A, Coclitu C, Constantinescu CS, Popescu BO, Tanasescu R. Beta interferons as immunotherapy in multiple sclerosis: a new outlook on a classic drug during the COVID-19 pandemic. QJM 2021; 114:691-697. [PMID: 33486513 PMCID: PMC7928608 DOI: 10.1093/qjmed/hcaa348] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/12/2020] [Accepted: 12/22/2020] [Indexed: 12/16/2022] Open
Abstract
Beta interferons (IFN-β) are pleiotropic cytokines with antiviral properties. They play important roles in the pathogenesis of multiple sclerosis (MS), an incurable immune-mediated disorder of the central nervous system. The clinical expression of MS is heterogeneous, with relapses of neuroinflammation and with disability accrual in considerable part unrelated to the attacks. The injectable recombinant IFN-β preparations are the first approved disease-modifying treatments for MS. They have moderate efficacy in reducing the frequency of relapses, but good long-term cost-efficacy and safety profiles, so are still widely used. They have some tolerability and adherence issues, partly mitigated in recent years by the introduction of a PEGylated formulation and use of 'smart' autoinjector devices. Their general impact on long-term disability is modest but could be further improved by developing accurate tools for identifying the patient profile of best responders to IFN-β. Here, we present the IFN-β-based immunomodulatory therapeutic approaches in MS, highlighting their place in the current coronavirus disease (COVID-19) pandemic. The potential role of IFN-β in the treatment of COVID-19 is also briefly discussed.
Collapse
Affiliation(s)
- L Dumitrescu
- From the Department of Clinical Neurosciences, University of Medicine and Pharmacy Carol Davila, Bucharest, Romania
- Department of Neurology, Colentina Hospital, Bucharest, Romania
| | - A Papathanasiou
- Department of Neurology, Queen’s Medical Centre, Nottingham University Hospitals, Nottingham, UK
| | - C Coclitu
- Department of Multiple Sclerosis and Neuroimmunology, CHU Grenoble, Grenoble, France
| | - C S Constantinescu
- Department of Neurology, Queen’s Medical Centre, Nottingham University Hospitals, Nottingham, UK
- Academic Clinical Neurology, Division of Clinical Neuroscience, C Floor, South Block, Queen's Medical Centre, Derby Road, NG7 2UH, Nottingham, UK
| | - B O Popescu
- From the Department of Clinical Neurosciences, University of Medicine and Pharmacy Carol Davila, Bucharest, Romania
- Department of Neurology, Colentina Hospital, Bucharest, Romania
| | - R Tanasescu
- Department of Neurology, Queen’s Medical Centre, Nottingham University Hospitals, Nottingham, UK
- Academic Clinical Neurology, Division of Clinical Neuroscience, C Floor, South Block, Queen's Medical Centre, Derby Road, NG7 2UH, Nottingham, UK
| |
Collapse
|
21
|
Cauwels A, Van Lint S, Rogge E, Verhee A, Van Den Eeckhout B, Pang S, Prinz M, Kley N, Uzé G, Tavernier J. Targeting IFN activity to both B cells and plasmacytoid dendritic cells induces a robust tolerogenic response and protection against EAE. Sci Rep 2021; 11:21575. [PMID: 34732771 PMCID: PMC8566508 DOI: 10.1038/s41598-021-00891-6] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2021] [Accepted: 10/06/2021] [Indexed: 02/07/2023] Open
Abstract
Type I Interferon (IFN) was the very first drug approved for the treatment of Multiple Sclerosis (MS), and is still frequently used as a first line therapy. However, systemic IFN also causes considerable side effects, affecting therapy adherence and dose escalation. In addition, the mechanism of action of IFN in MS is multifactorial and still not completely understood. Using AcTaferons (Activity-on-Target IFNs, AFNs), optimized IFN-based immunocytokines that allow cell-specific targeting, we have previously demonstrated that specific targeting of IFN activity to dendritic cells (DCs) can protect against experimental autoimmune encephalitis (EAE), inducing in vivo tolerogenic protective effects, evidenced by increased indoleamine-2,3-dioxygenase (IDO) and transforming growth factor β (TGFβ) release by plasmacytoid (p) DCs and improved immunosuppressive capacity of regulatory T and B cells. We here report that targeting type I IFN activity specifically towards B cells also provides strong protection against EAE, and that targeting pDCs using SiglecH-AFN can significantly add to this protective effect. The superior protection achieved by simultaneous targeting of both B lymphocytes and pDCs correlated with improved IL-10 responses in B cells and conventional cDCs, and with a previously unseen very robust IDO response in several cells, including all B and T lymphocytes, cDC1 and cDC2.
Collapse
Affiliation(s)
- Anje Cauwels
- Cytokine Receptor Laboratory, VIB Medical Biotechnology Center, Ghent University, A. Baertsoenkaai 3, 9000, Ghent, Belgium. .,Orionis Biosciences, 9052, Ghent, Belgium.
| | - Sandra Van Lint
- Cytokine Receptor Laboratory, VIB Medical Biotechnology Center, Ghent University, A. Baertsoenkaai 3, 9000, Ghent, Belgium
| | - Elke Rogge
- Cytokine Receptor Laboratory, VIB Medical Biotechnology Center, Ghent University, A. Baertsoenkaai 3, 9000, Ghent, Belgium.,Orionis Biosciences, 9052, Ghent, Belgium
| | - Annick Verhee
- Cytokine Receptor Laboratory, VIB Medical Biotechnology Center, Ghent University, A. Baertsoenkaai 3, 9000, Ghent, Belgium
| | - Bram Van Den Eeckhout
- Cytokine Receptor Laboratory, VIB Medical Biotechnology Center, Ghent University, A. Baertsoenkaai 3, 9000, Ghent, Belgium
| | - Shengru Pang
- Institute of Neuropathology, Faculty of Medicine, University of Freiburg, 79106, Freiburg, Germany
| | - Marco Prinz
- Institute of Neuropathology, Faculty of Medicine, University of Freiburg, 79106, Freiburg, Germany.,BIOSS Centre for Biological Signalling Studies, University of Freiburg, 79106, Freiburg, Germany
| | - Niko Kley
- Orionis Biosciences, 9052, Ghent, Belgium
| | - Gilles Uzé
- CNRS UMR 5235, University Montpellier, 34095, Montpellier, France
| | - Jan Tavernier
- Cytokine Receptor Laboratory, VIB Medical Biotechnology Center, Ghent University, A. Baertsoenkaai 3, 9000, Ghent, Belgium. .,Orionis Biosciences, 9052, Ghent, Belgium.
| |
Collapse
|
22
|
Yang M, Yi P, Jiang J, Zhao M, Wu H, Lu Q. Dysregulated translational factors and epigenetic regulations orchestrate in B cells contributing to autoimmune diseases. Int Rev Immunol 2021; 42:1-25. [PMID: 34445929 DOI: 10.1080/08830185.2021.1964498] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
B cells play a crucial role in antigen presentation, antibody production and pro-/anti-inflammatory cytokine secretion in adaptive immunity. Several translational factors including transcription factors and cytokines participate in the regulation of B cell development, with the cooperation of epigenetic regulations. Autoimmune diseases are generally characterized with autoreactive B cells and high-level pathogenic autoantibodies. The success of B cell depletion therapy in mouse model and clinical trials has proven the role of B cells in pathogenesis of autoimmune diseases. The failure of B cell tolerance in immune checkpoints results in accumulated autoreactive naïve B (BN) cells with aberrant B cell receptor signaling and dysregulated B cell response, contributing to self-antibody-mediated autoimmune reaction. Dysregulation of translational factors and epigenetic alterations in B cells has been demonstrated to correlate with aberrant B cell compartment in autoimmune diseases, such as systemic lupus erythematosus, rheumatoid arthritis, primary Sjögren's syndrome, multiple sclerosis, diabetes mellitus and pemphigus. This review is intended to summarize the interaction of translational factors and epigenetic regulations that are involved with development and differentiation of B cells, and the mechanism of dysregulation in the pathogenesis of autoimmune diseases.
Collapse
Affiliation(s)
- Ming Yang
- Department of Dermatology, Second Xiangya Hospital, Central South University, Hunan Key Laboratory of Medical Epigenomics, Changsha, Hunan, China
| | - Ping Yi
- Department of Dermatology, Second Xiangya Hospital, Central South University, Hunan Key Laboratory of Medical Epigenomics, Changsha, Hunan, China
| | - Jiao Jiang
- Department of Dermatology, Second Xiangya Hospital, Central South University, Hunan Key Laboratory of Medical Epigenomics, Changsha, Hunan, China
| | - Ming Zhao
- Department of Dermatology, Second Xiangya Hospital, Central South University, Hunan Key Laboratory of Medical Epigenomics, Changsha, Hunan, China
| | - Haijing Wu
- Department of Dermatology, Second Xiangya Hospital, Central South University, Hunan Key Laboratory of Medical Epigenomics, Changsha, Hunan, China
| | - Qianjin Lu
- Department of Dermatology, Second Xiangya Hospital, Central South University, Hunan Key Laboratory of Medical Epigenomics, Changsha, Hunan, China.,Department of Dermatology, Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing, Jiangsu, China
| |
Collapse
|
23
|
Veroni C, Aloisi F. The CD8 T Cell-Epstein-Barr Virus-B Cell Trialogue: A Central Issue in Multiple Sclerosis Pathogenesis. Front Immunol 2021; 12:665718. [PMID: 34305896 PMCID: PMC8292956 DOI: 10.3389/fimmu.2021.665718] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Accepted: 06/07/2021] [Indexed: 12/11/2022] Open
Abstract
The cause and the pathogenic mechanisms leading to multiple sclerosis (MS), a chronic inflammatory disease of the central nervous system (CNS), are still under scrutiny. During the last decade, awareness has increased that multiple genetic and environmental factors act in concert to modulate MS risk. Likewise, the landscape of cells of the adaptive immune system that are believed to play a role in MS immunopathogenesis has expanded by including not only CD4 T helper cells but also cytotoxic CD8 T cells and B cells. Once the key cellular players are identified, the main challenge is to define precisely how they act and interact to induce neuroinflammation and the neurodegenerative cascade in MS. CD8 T cells have been implicated in MS pathogenesis since the 80's when it was shown that CD8 T cells predominate in MS brain lesions. Interest in the role of CD8 T cells in MS was revived in 2000 and the years thereafter by studies showing that CNS-recruited CD8 T cells are clonally expanded and have a memory effector phenotype indicating in situ antigen-driven reactivation. The association of certain MHC class I alleles with MS genetic risk implicates CD8 T cells in disease pathogenesis. Moreover, experimental studies have highlighted the detrimental effects of CD8 T cell activation on neural cells. While the antigens responsible for T cell recruitment and activation in the CNS remain elusive, the high efficacy of B-cell depleting drugs in MS and a growing number of studies implicate B cells and Epstein-Barr virus (EBV), a B-lymphotropic herpesvirus that is strongly associated with MS, in the activation of pathogenic T cells. This article reviews the results of human studies that have contributed to elucidate the role of CD8 T cells in MS immunopathogenesis, and discusses them in light of current understanding of autoreactivity, B-cell and EBV involvement in MS, and mechanism of action of different MS treatments. Based on the available evidences, an immunopathological model of MS is proposed that entails a persistent EBV infection of CNS-infiltrating B cells as the target of a dysregulated cytotoxic CD8 T cell response causing CNS tissue damage.
Collapse
Affiliation(s)
| | - Francesca Aloisi
- Department of Neuroscience, Istituto Superiore di Sanità, Rome, Italy
| |
Collapse
|
24
|
DiSano KD, Gilli F, Pachner AR. Memory B Cells in Multiple Sclerosis: Emerging Players in Disease Pathogenesis. Front Immunol 2021; 12:676686. [PMID: 34168647 PMCID: PMC8217754 DOI: 10.3389/fimmu.2021.676686] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2021] [Accepted: 05/11/2021] [Indexed: 11/25/2022] Open
Abstract
Multiple Sclerosis (MS) is an inflammatory demyelinating disease of the central nervous system. Once thought to be primarily driven by T cells, B cells are emerging as central players in MS immunopathogenesis. Interest in multiple B cell phenotypes in MS expanded following the efficacy of B cell-depleting agents targeting CD20 in relapsing-remitting MS and inflammatory primary progressive MS patients. Interestingly, these therapies primarily target non-antibody secreting cells. Emerging studies seek to explore B cell functions beyond antibody-mediated roles, including cytokine production, antigen presentation, and ectopic follicle-like aggregate formation. Importantly, memory B cells (Bmem) are rising as a key B cell phenotype to investigate in MS due to their antigen-experience, increased lifespan, and rapid response to stimulation. Bmem display diverse effector functions including cytokine production, antigen presentation, and serving as antigen-experienced precursors to antibody-secreting cells. In this review, we explore the cellular and molecular processes involved in Bmem development, Bmem phenotypes, and effector functions. We then examine how these concepts may be applied to the potential role(s) of Bmem in MS pathogenesis. We investigate Bmem both within the periphery and inside the CNS compartment, focusing on Bmem phenotypes and proposed functions in MS and its animal models. Finally, we review how current immunomodulatory therapies, including B cell-directed therapies and other immunomodulatory therapies, modify Bmem and how this knowledge may be harnessed to direct therapeutic strategies in MS.
Collapse
Affiliation(s)
- Krista D. DiSano
- Department of Neurology, Geisel School of Medicine & Dartmouth-Hitchcock Medical Center, Lebanon, NH, United States
| | | | | |
Collapse
|
25
|
Toll-Like Receptor Homolog CD180 Expression Is Diminished on Natural Autoantibody-Producing B Cells of Patients with Autoimmune CNS Disorders. J Immunol Res 2021; 2021:9953317. [PMID: 34124274 PMCID: PMC8169253 DOI: 10.1155/2021/9953317] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2021] [Revised: 04/26/2021] [Accepted: 05/04/2021] [Indexed: 11/17/2022] Open
Abstract
Purpose Decreased expression of TLR homolog CD180 in peripheral blood B cells and its potential role in antibody production have been described in autoimmune diseases. Effectiveness of anti-CD20 therapy in neuromyelitis optica spectrum disorder (NMOSD) and multiple sclerosis (MS) strengthens the role of B cells in the pathogenesis. Therefore, we aimed to investigate the CD180 expression of peripheral blood B cell subsets in NMOSD and MS patients and analyze the levels of natural anti-citrate synthase (CS) IgG autoantibodies and IgG antibodies induced by bacterial infections reported to play a role in the pathogenesis of NMOSD or MS. Methods We analyzed the distribution and CD180 expression of peripheral blood B cell subsets, defined by CD19/CD27/IgD staining, and measured anti-CS IgM/G natural autoantibody and antibacterial IgG serum levels in NMOSD, RRMS, and healthy controls (HC). Results We found decreased naïve and increased memory B cells in NMOSD compared to MS. Among the investigated four B cell subsets, CD180 expression was exclusively decreased in CD19+CD27+IgD+ nonswitched (NS) memory B cells in both NMOSD and MS compared to HC. Furthermore, the anti-CS IgM natural autoantibody serum level was lower in both NMOSD and MS. In addition, we found a tendency of higher anti-CS IgG natural autoantibody levels only in anti-Chlamydia IgG antibody-positive NMOSD and MS patients. Conclusions Our results suggest that reduced CD180 expression of NS B cells could contribute to the deficient natural IgM autoantibody production in NMOSD and MS, whereas natural IgG autoantibody levels show an association with antibacterial antibodies.
Collapse
|
26
|
Lundtoft C, Pucholt P, Imgenberg-Kreuz J, Carlsson-Almlöf J, Eloranta ML, Syvänen AC, Nordmark G, Sandling JK, Kockum I, Olsson T, Rönnblom L, Hagberg N. Function of multiple sclerosis-protective HLA class I alleles revealed by genome-wide protein-quantitative trait loci mapping of interferon signalling. PLoS Genet 2020; 16:e1009199. [PMID: 33104735 PMCID: PMC7644105 DOI: 10.1371/journal.pgen.1009199] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2020] [Revised: 11/05/2020] [Accepted: 10/15/2020] [Indexed: 12/20/2022] Open
Abstract
Interferons (IFNs) are cytokines that are central to the host defence against viruses and other microorganisms. If not properly regulated, IFNs may contribute to the pathogenesis of inflammatory autoimmune, or infectious diseases. To identify genetic polymorphisms regulating the IFN system we performed an unbiased genome-wide protein-quantitative trait loci (pQTL) mapping of cell-type specific type I and type II IFN receptor levels and their responses in immune cells from 303 healthy individuals. Seven genome-wide significant (p < 5.0E-8) pQTLs were identified. Two independent SNPs that tagged the multiple sclerosis (MS)-protective HLA class I alleles A*02/A*68 and B*44, respectively, were associated with increased levels of IFNAR2 in B and T cells, with the most prominent effect in IgD–CD27+ memory B cells. The increased IFNAR2 levels in B cells were replicated in cells from an independent set of healthy individuals and in MS patients. Despite increased IFNAR2 levels, B and T cells carrying the MS-protective alleles displayed a reduced response to type I IFN stimulation. Expression and methylation-QTL analysis demonstrated increased mRNA expression of the pseudogene HLA-J in B cells carrying the MS-protective class I alleles, possibly driven via methylation-dependent transcriptional regulation. Together these data suggest that the MS-protective effects of HLA class I alleles are unrelated to their antigen-presenting function, and propose a previously unappreciated function of type I IFN signalling in B and T cells in MS immune-pathogenesis. Genetic association studies have been very successful in identifying disease-associated single nucleotide polymorphisms (SNPs), but it has been challenging to define the molecular mechanisms underlying these associations. As interferons (IFNs) have a central role in the immune system, we hypothesized that some of the SNPs associated to immune-mediated diseases would affect the IFN system. By combining genetic data with characterization of interferon receptor levels and their responses on the protein level in immune cells from 303 genotyped healthy individuals, we show that two SNPs tagging the HLA class I alleles A*02/A*68 and B*44 are associated with a decreased response to type I IFN stimulation in B cells and T cells. Notably, both HLA-A*02 and HLA-B*44 confer protection from developing multiple sclerosis (MS), which is a chronic inflammatory neurologic disease. In addition to suggesting a pathogenic role of enhanced type I interferon signalling in B cells and T cells in MS, our data emphasize the fact that genetic associations in the HLA locus can affect functions not directly associated to antigen presentation, which conceptually may be important for other diseases genetically associated to the HLA locus.
Collapse
Affiliation(s)
- Christian Lundtoft
- Rheumatology and Science for Life Laboratories, Department of Medical Sciences, Uppsala University, Uppsala, Sweden
| | - Pascal Pucholt
- Rheumatology and Science for Life Laboratories, Department of Medical Sciences, Uppsala University, Uppsala, Sweden
| | - Juliana Imgenberg-Kreuz
- Rheumatology and Science for Life Laboratories, Department of Medical Sciences, Uppsala University, Uppsala, Sweden
| | - Jonas Carlsson-Almlöf
- Molecular Medicine and Science for Life Laboratory, Department of Medical Sciences, Uppsala University, Uppsala, Sweden
| | - Maija-Leena Eloranta
- Rheumatology and Science for Life Laboratories, Department of Medical Sciences, Uppsala University, Uppsala, Sweden
| | - Ann-Christine Syvänen
- Molecular Medicine and Science for Life Laboratory, Department of Medical Sciences, Uppsala University, Uppsala, Sweden
| | - Gunnel Nordmark
- Rheumatology and Science for Life Laboratories, Department of Medical Sciences, Uppsala University, Uppsala, Sweden
| | - Johanna K. Sandling
- Rheumatology and Science for Life Laboratories, Department of Medical Sciences, Uppsala University, Uppsala, Sweden
| | - Ingrid Kockum
- Centre for Molecular Medicine, Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Tomas Olsson
- Centre for Molecular Medicine, Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Lars Rönnblom
- Rheumatology and Science for Life Laboratories, Department of Medical Sciences, Uppsala University, Uppsala, Sweden
| | - Niklas Hagberg
- Rheumatology and Science for Life Laboratories, Department of Medical Sciences, Uppsala University, Uppsala, Sweden
- * E-mail:
| |
Collapse
|
27
|
Kemmerer CL, Pernpeintner V, Ruschil C, Abdelhak A, Scholl M, Ziemann U, Krumbholz M, Hemmer B, Kowarik MC. Differential effects of disease modifying drugs on peripheral blood B cell subsets: A cross sectional study in multiple sclerosis patients treated with interferon-β, glatiramer acetate, dimethyl fumarate, fingolimod or natalizumab. PLoS One 2020; 15:e0235449. [PMID: 32716916 PMCID: PMC7384624 DOI: 10.1371/journal.pone.0235449] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2020] [Accepted: 06/15/2020] [Indexed: 12/14/2022] Open
Abstract
Background Several disease modifying drugs (DMDs) have been approved for the treatment of multiple sclerosis (MS), however, little is known about their differential impact on peripheral blood (PB) B cell subsets. Methods We performed a cross sectional study on PB B cells in MS patients treated with interferon-β (n = 25), glatiramer acetate (n = 19), dimethyl fumarate (n = 15), fingolimod (n = 16) or natalizumab (n = 22), untreated MS patients (n = 20), and in patients with non-inflammatory neurological diseases (n = 12). Besides analyzing routine laboratory data, flow cytometry was performed to analyze naïve B cells (CD19+CD20+CD27-IgD+), non-class switched (CD19+CD20+CD27+IgD+) and class-switched memory B cells (CD19+CD20+CD27+IgD-), double negative B cells (CD19+CD20lowCD27-IgD-) and plasmablasts (CD19+CD20lowCD27+CD38++). Results Treatment associated changes were found for the overall B cell pool as well as for all B cell subsets. Natalizumab increased absolute numbers and percentage of all B cells mainly by expanding the memory B cell pool. Fingolimod decreased absolute numbers of all B cell subsets and the percentage of total B cells. Fingolimod, dimethyl fumarate and interferon-β treatments were associated with an increase in the fraction of naïve B cells while class switched and non-class switched memory B cells showed decreased percentages. Conclusion Our results highlight differential effects of DMDs on the PB B cell compartment. Across the examined treatments, a decreased percentage of memory B cells was found in dimethyl fumarate, interferon-β and fingolimod treated patients which might contribute to the drugs’ mode of action in MS. Further studies are necessary to decipher the exact role of B cell subsets during MS pathogenesis.
Collapse
Affiliation(s)
- C. L. Kemmerer
- Department of Neurology & Stroke, and Hertie-Institute for Clinical Brain Research, Eberhard-Karls University of Tübingen, Tübingen, Germany
| | - V. Pernpeintner
- Department of Neurology, Klinikum rechts der Isar, Technische Universität München, Munich, Germany
| | - C. Ruschil
- Department of Neurology & Stroke, and Hertie-Institute for Clinical Brain Research, Eberhard-Karls University of Tübingen, Tübingen, Germany
| | - A. Abdelhak
- Department of Neurology & Stroke, and Hertie-Institute for Clinical Brain Research, Eberhard-Karls University of Tübingen, Tübingen, Germany
| | - M. Scholl
- Department of Neurology & Stroke, and Hertie-Institute for Clinical Brain Research, Eberhard-Karls University of Tübingen, Tübingen, Germany
| | - U. Ziemann
- Department of Neurology & Stroke, and Hertie-Institute for Clinical Brain Research, Eberhard-Karls University of Tübingen, Tübingen, Germany
| | - M. Krumbholz
- Department of Neurology & Stroke, and Hertie-Institute for Clinical Brain Research, Eberhard-Karls University of Tübingen, Tübingen, Germany
| | - B. Hemmer
- Department of Neurology, Klinikum rechts der Isar, Technische Universität München, Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | - M. C. Kowarik
- Department of Neurology & Stroke, and Hertie-Institute for Clinical Brain Research, Eberhard-Karls University of Tübingen, Tübingen, Germany
- Department of Neurology, Klinikum rechts der Isar, Technische Universität München, Munich, Germany
- * E-mail:
| |
Collapse
|
28
|
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: 4.2] [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
|
29
|
Severa M, Farina C, Salvetti M, Coccia EM. Three Decades of Interferon-β in Multiple Sclerosis: Can We Repurpose This Information for the Management of SARS-CoV2 Infection? Front Immunol 2020; 11:1459. [PMID: 32655578 PMCID: PMC7326001 DOI: 10.3389/fimmu.2020.01459] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2020] [Accepted: 06/04/2020] [Indexed: 12/16/2022] Open
Affiliation(s)
- Martina Severa
- Department of Infectious Diseases, Istituto Superiore di Sanità, Rome, Italy
| | - Cinthia Farina
- Institute of Experimental Neurology (INSpe), Division of Neuroscience, IRCSS San Raffaele Scientific Institute, Milan, Italy
| | - Marco Salvetti
- Center for Experimental Neurological Therapies, Sant'Andrea Hospital, Department of Neurosciences, Mental Health and Sensory Organs, Sapienza University, Rome, Italy.,Istituto Neurologico Mediterraneo (INM) Neuromed, Pozzilli, Italy
| | | |
Collapse
|
30
|
Gharibi T, Babaloo Z, Hosseini A, Marofi F, Ebrahimi-Kalan A, Jahandideh S, Baradaran B. The role of B cells in the immunopathogenesis of multiple sclerosis. Immunology 2020; 160:325-335. [PMID: 32249925 DOI: 10.1111/imm.13198] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2019] [Revised: 03/01/2020] [Accepted: 03/25/2020] [Indexed: 02/06/2023] Open
Abstract
There is ongoing debate on how B cells contribute to the pathogenesis of multiple sclerosis (MS). The success of B-cell targeting therapies in MS highlighted the role of B cells, particularly the antibody-independent functions of these cells such as antigen presentation to T cells and modulation of the function of T cells and myeloid cells by secreting pathogenic and/or protective cytokines in the central nervous system. Here, we discuss the role of different antibody-dependent and antibody-independent functions of B cells in MS disease activity and progression proposing new therapeutic strategies for the optimization of B-cell targeting treatments.
Collapse
Affiliation(s)
- Tohid Gharibi
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.,Department of Immunology, School of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran.,Neuroscience Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.,Department of Neurosciences and Cognition, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran.,Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Zohreh Babaloo
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.,Department of Immunology, School of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Arezoo Hosseini
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.,Department of Immunology, School of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran.,Neuroscience Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.,Department of Neurosciences and Cognition, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Faroogh Marofi
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.,Department of Immunology, School of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Abbas Ebrahimi-Kalan
- Neuroscience Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.,Department of Neurosciences and Cognition, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Saeed Jahandideh
- Department of Biochemistry, Pasteur Institute of Iran, Tehran, Iran
| | - Behzad Baradaran
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.,Department of Immunology, School of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
31
|
Akgün K, Blankenburg J, Marggraf M, Haase R, Ziemssen T. Event-Driven Immunoprofiling Predicts Return of Disease Activity in Alemtuzumab-Treated Multiple Sclerosis. Front Immunol 2020; 11:56. [PMID: 32082320 PMCID: PMC7005935 DOI: 10.3389/fimmu.2020.00056] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2019] [Accepted: 01/09/2020] [Indexed: 12/19/2022] Open
Abstract
Background: Alemtuzumab is a highly effective drug for the treatment of multiple sclerosis (MS), characterized by specific patterns of depletion and repopulation. As an induction-like treatment concept, two mandatory infusion courses can inhibit long-term disease activity in the majority of patients, and additional courses can successfully manage subsequent re-emergence of disease activity. Currently, there are no biomarkers to identify patients with re-emergent disease activity requiring retreatment. Methods: In this study, we systematically characterized 16 MS patients commencing alemtuzumab. Clinical parameters, MRI and detailed immunoprofiling were conducted every 3 months for up to 84 months. Results: Alemtuzumab led to significant decrease in clinical disease activity in all evaluated patients. Nine out of 16 patients presented with no evidence of disease activity (NEDA)-3 up to 84 months (“complete-responder”), while 7 patients demonstrated clinical or/and subclinical MRI disease activity and received alemutzumab retreatment (“partial-responder”). In both response categories, all T- and B-cell subsets were markedly depleted after alemtuzumab therapy. In particular, absolute numbers of Th1 and Th17 cells were markedly decreased and remained stable below baseline levels—this effect was particularly pronounced in complete-responders. While mean cell numbers did not differ significantly between groups, analysis of event-driven immunoprofiling demonstrated that absolute numbers of Th1 and Th17 cells showed a reproducible increase starting 6 months before relapse activity. This change appears to predict emergent disease activity when compared with stable disease. Conclusion: Studies with larger patient populations are needed to confirm that frequent immunoprofiling may assist in evaluating clinical decision-making of alemtuzumab retreatment.
Collapse
Affiliation(s)
- Katja Akgün
- Center of Clinical Neuroscience, University Hospital, Technical University Dresden, Dresden, Germany
| | - Judith Blankenburg
- Center of Clinical Neuroscience, University Hospital, Technical University Dresden, Dresden, Germany
| | - Michaela Marggraf
- Center of Clinical Neuroscience, University Hospital, Technical University Dresden, Dresden, Germany
| | - Rocco Haase
- Center of Clinical Neuroscience, University Hospital, Technical University Dresden, Dresden, Germany
| | - Tjalf Ziemssen
- Center of Clinical Neuroscience, University Hospital, Technical University Dresden, Dresden, Germany
| |
Collapse
|
32
|
Bar-Or A, Pender MP, Khanna R, Steinman L, Hartung HP, Maniar T, Croze E, Aftab BT, Giovannoni G, Joshi MA. Epstein-Barr Virus in Multiple Sclerosis: Theory and Emerging Immunotherapies. Trends Mol Med 2019; 26:296-310. [PMID: 31862243 PMCID: PMC7106557 DOI: 10.1016/j.molmed.2019.11.003] [Citation(s) in RCA: 164] [Impact Index Per Article: 27.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2019] [Revised: 10/31/2019] [Accepted: 11/06/2019] [Indexed: 12/12/2022]
Abstract
New treatments for multiple sclerosis (MS) focused on B cells have created an atmosphere of excitement in the MS community. B cells are now known to play a major role in disease, demonstrated by the highly impactful effect of a B cell-depleting antibody on controlling MS. The idea that a virus may play a role in the development of MS has a long history and is supported mostly by studies demonstrating a link between B cell-tropic Epstein–Barr virus (EBV) and disease onset. Efforts to develop antiviral strategies for treating MS are underway. Although gaps remain in our understanding of the etiology of MS, the role, if any, of viruses in propagating pathogenic immune responses deserves attention.
Collapse
Affiliation(s)
- Amit Bar-Or
- Center for Neuroinflammation and Experimental Therapeutics, Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Michael P Pender
- Faculty of Medicine, University of Queensland, Brisbane, QLD, Australia
| | - Rajiv Khanna
- Centre for Immunotherapy and Vaccine Development, Queensland Institute of Medical Research (QIMR) Berghofer Medical Research Institute, Brisbane, QLD, Australia
| | - Lawrence Steinman
- Department of Neurology and Neurological Sciences, Beckman Center for Molecular Medicine, Stanford University Medical Center, Stanford, CA, USA
| | - Hans-Peter Hartung
- Department of Neurology, Medical Faculty, Heinrich Heine University, Düsseldorf, Germany
| | - Tap Maniar
- Clinical Development, Torque Therapeutics, Boston, MA, USA
| | - Ed Croze
- IRIS-Bay, San Francisco, CA, USA.
| | - Blake T Aftab
- Preclinical Science and Translational Medicine, Atara Biotherapeutics, South San Francisco, CA, USA
| | - Gavin Giovannoni
- Blizard Institute, Queen Mary University London, Barts and the London School of Medicine, London, UK
| | - Manher A Joshi
- Medical Affairs, Atara Biotherapeutics, South San Francisco, CA, USA
| |
Collapse
|
33
|
Maimaitijiang G, Watanabe M, Shinoda K, Isobe N, Nakamura Y, Masaki K, Matsushita T, Yoshikai Y, Kira JI. Long-term use of interferon-β in multiple sclerosis increases Vδ1 -Vδ2 -Vγ9 - γδ T cells that are associated with a better outcome. J Neuroinflammation 2019; 16:179. [PMID: 31519178 PMCID: PMC6743159 DOI: 10.1186/s12974-019-1574-5] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2019] [Accepted: 08/29/2019] [Indexed: 12/27/2022] Open
Abstract
Background We previously reported that Vδ2+Vγ9+ γδ T cells were significantly decreased in multiple sclerosis (MS) patients without disease-modifying therapies (untreated MS) and were negatively correlated with Expanded Disability Status Scale (EDSS) scores, suggesting protective roles of Vδ2+Vγ9+ γδ T cells. Interferon-β (IFN-β) is one of the first-line disease-modifying drugs for MS. However, no previous studies have reported changes in γδ T cell subsets under IFN-β treatment. Therefore, we aimed to clarify the effects of the long-term usage of IFN-β on γδ T cell subsets in MS patients. Methods Comprehensive flow cytometric immunophenotyping was performed in 35 untreated MS and 21 MS patients on IFN-β for more than 2 years (IFN-β-treated MS) including eight super-responders fulfilling no evidence of disease activity criteria, and 44 healthy controls (HCs). Results The percentages of Vδ2+Vγ9+ cells in γδ T cells were significantly lower in untreated and IFN-β-treated MS patients than in HCs. By contrast, the percentages of Vδ1−Vδ2−Vγ9− cells in γδ T cells were markedly higher in IFN-β-treated MS patients than in HCs and untreated MS patients (both p < 0.001). A significant negative correlation between the percentages of Vδ2+Vγ9+ cells in γδ T cells and EDSS scores was confirmed in untreated MS but not evident in IFN-β-treated MS. Moreover, class-switched memory B cells were decreased in IFN-β-treated MS compared with HCs (p < 0.001) and untreated MS patients (p = 0.006). Interestingly, the percentages of Vδ1−Vδ2−Vγ9− cells in γδ T cells were negatively correlated with class-switched memory B cell percentages in all MS patients (r = − 0.369, p = 0.005), and the percentages of Vδ1−Vδ2−Vγ9− cells in Vδ1−Vδ2− γδ T cells were negatively correlated with EDSS scores only in IFN-β super-responders (r = − 0.976, p < 0.001). Conclusions The present study suggests that long-term usage of IFN-β increases Vδ1−Vδ2−Vγ9− γδ T cells, which are associated with a better outcome, especially in IFN-β super-responders. Thus, increased Vδ1−Vδ2−Vγ9− cells together with decreased class-switched memory B cells may contribute to the suppression of disease activity in MS patients under IFN-β treatment. Electronic supplementary material The online version of this article (10.1186/s12974-019-1574-5) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Guzailiayi Maimaitijiang
- Department of Neurology, Neurological Institute, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Mitsuru Watanabe
- Department of Neurology, Neurological Institute, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Koji Shinoda
- Department of Neurology, Neurological Institute, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Noriko Isobe
- Department of Neurological Therapeutics, Neurological Institute, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Yuri Nakamura
- Department of Neurology, Neurological Institute, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Katsuhisa Masaki
- Department of Neurology, Neurological Institute, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Takuya Matsushita
- Department of Neurology, Neurological Institute, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Yasunobu Yoshikai
- Division of Host Defense, Medical Institute of Bioregulation, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Jun-Ichi Kira
- Department of Neurology, Neurological Institute, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan.
| |
Collapse
|
34
|
Malekzadeh A, Leurs C, van Wieringen W, Steenwijk MD, Schoonheim MM, Amann M, Naegelin Y, Kuhle J, Killestein J, Teunissen CE. Plasma proteome in multiple sclerosis disease progression. Ann Clin Transl Neurol 2019; 6:1582-1594. [PMID: 31364818 PMCID: PMC7651845 DOI: 10.1002/acn3.771] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2018] [Revised: 02/28/2019] [Accepted: 03/07/2019] [Indexed: 01/01/2023] Open
Abstract
Background The pathophysiology of multiple sclerosis disease progression remains undetermined. The aim of this study was to identify differences in plasma proteome during different stages of MS disease progression. Methods We used a multiplex aptamer proteomics platform (Somalogic) for sensitive detection of 1129 proteins in plasma. MS patients were selected and categorized based on baseline and a 4‐year follow‐up EDSS (delta EDSS) scores; relapse‐onset (RO) slow progression (n = 31), RO with rapid progression (n = 29), primary progressive (n = 30), and healthy controls (n = 20). The relation of baseline plasma protein levels with delta EDSS and different MRI progression parameters were assessed using linear regression models. Results Regression analyses of plasma proteins with delta EDSS showed six significant associations. Strong associations were found for the proteins LGLAS8 (P = 7.64 × 10−5, q = 0.06), CCL3 (P = 0.0001, q = 0.06), and RGMA (P = 0.0005, q = 0.09). In addition, associations of plasma proteins were found with percentage brain volume for C3 (P = 2,08 × 10−9, q = 1,70 × 10−6), FGF9 (P = 3,42 × 10−9, q = 1,70 × 10−6), and EHMT2 (P = 0.0007, q = 0.01). Most of the significant markers were associated with cell‐cell and cell‐extracellular matrix adhesion, immune system communication, immune system activation, and complement pathways. Conclusions Our results revealed eight novel biomarkers related to clinical and radiological progression in MS. These results indicate that changes in immune system, complement pathway and ECM remodeling proteins contribute to MS progression and may therefore be further explored for use in prognosis of MS.
Collapse
Affiliation(s)
- Arjan Malekzadeh
- Department of Clinical Chemistry, Amsterdam University Medical Centre, Amsterdam, The Netherlands
| | - Cyra Leurs
- Department of Neurology, Amsterdam University Medical Centre, Amsterdam, The Netherlands
| | - Wessel van Wieringen
- Department of Mathematics, Amsterdam University Medical Centre, Amsterdam, The Netherlands
| | - Martijn D Steenwijk
- Department of Anatomy and Neurosciences, Amsterdam Neuroscience, MS Center Amsterdam, Amsterdam University Medical Centre, Amsterdam, The Netherlands
| | - Menno M Schoonheim
- Department of Anatomy and Neurosciences, Amsterdam Neuroscience, MS Center Amsterdam, Amsterdam University Medical Centre, Amsterdam, The Netherlands
| | - Michael Amann
- Division of Diagnostic and Interventional Neuroradiology, Department of Radiology and Nuclear Medicine, University Hospital Basel, Basel, Switzerland.,Medical Image Analysis Center (MIAC AG), Basel, Switzerland
| | - Yvonne Naegelin
- Department of Biomedicine and Clinical Research, University Hospital Basel, Basel, Switzerland
| | - Jens Kuhle
- Department of Biomedicine and Clinical Research, University Hospital Basel, Basel, Switzerland
| | - Joep Killestein
- Department of Neurology, Amsterdam University Medical Centre, Amsterdam, The Netherlands
| | - Charlotte E Teunissen
- Department of Clinical Chemistry, Amsterdam University Medical Centre, Amsterdam, The Netherlands
| |
Collapse
|
35
|
Osherov M, Milo R. B Cell-based Therapies for Multiple Sclerosis. EMERGING DRUGS AND TARGETS FOR MULTIPLE SCLEROSIS 2019. [DOI: 10.1039/9781788016070-00134] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
The traditional view of multiple sclerosis (MS) as a T cell mediated autoimmune disease of the central nervous system (CNS) has evolved into a concept of an immune-mediated disease where complex bi-directional interactions between T cells, B cells and myeloid cells underlie and shape CNS-directed autoimmunity. B cells are now recognized as major contributors to the pathogenesis of MS, largely due to increased understanding of their biology and the profound anti-inflammatory effects demonstrated by B cell depletion in MS. In this chapter we discuss the fundamental roles B cells play in the pathogenesis of MS and review current and future therapeutic strategies targeting B cells in MS, including B cell depletion with various monoclonal antibodies (mAbs) against the B cell surface markers CD20 and CD19, anti-B cell cytokine therapies, blocking Bruton's tyrosine kinase (BTK) in B cells, and various immunomodulatory and immunosuppressive effects exerted on B cells by virtually all other approved therapies for MS.
Collapse
Affiliation(s)
- Michael Osherov
- Department of Neurology, Barzilai University Medical Center 2 Hahistadrut St. Ashkelon 7830604 Israel
| | - Ron Milo
- Department of Neurology, Barzilai University Medical Center 2 Hahistadrut St. Ashkelon 7830604 Israel
- Faculty of Health Sciences, Ben-Gurion University of the Negev Beer-Sheva Israel
| |
Collapse
|
36
|
Severa M, Rizzo F, Srinivasan S, Di Dario M, Giacomini E, Buscarinu MC, Cruciani M, Etna MP, Sandini S, Mechelli R, Farina A, Trivedi P, Hertzog PJ, Salvetti M, Farina C, Coccia EM. A cell type-specific transcriptomic approach to map B cell and monocyte type I interferon-linked pathogenic signatures in Multiple Sclerosis. J Autoimmun 2019; 101:1-16. [PMID: 31047767 DOI: 10.1016/j.jaut.2019.04.006] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2019] [Revised: 04/08/2019] [Accepted: 04/08/2019] [Indexed: 12/15/2022]
Abstract
Alteration in endogenous Interferon (IFN) system may profoundly impact immune cell function in autoimmune diseases. Here, we provide evidence that dysregulation in IFN-regulated genes and pathways are involved in B cell- and monocyte-driven pathogenic contribution to Multiple Sclerosis (MS) development and maintenance. In particular, by using an Interferome-based cell type-specific approach, we characterized an increased susceptibility to an IFN-linked caspase-3 dependent apoptotic cell death in both B cells and monocytes of MS patients that may arise from their chronic activation and persistent stimulation by activated T cells. Ongoing caspase-3 activation functionally impacts on MS monocyte properties influencing the STAT-3/IL-16 axis, thus, driving increased expression and massive release of the bio-active IL-16 triggering and perpetuating CD4+ T cell migration. Importantly, our analysis also identified a previously unknown multi-component defect in type I IFN-mediated signaling and response to virus pathways specific of MS B cells, impacting on induction of anti-viral responses and Epstein-barr virus infection control in patients. Taking advantage of cell type-specific transcriptomics and in-depth functional validation, this study revealed pathogenic contribution of endogenous IFN signaling and IFN-regulated cell processes to MS pathogenesis with implications on fate and functions of B cells and monocytes that may hold therapeutic potential.
Collapse
Affiliation(s)
- Martina Severa
- Department of Infectious Diseases, Istituto Superiore di Sanità, Rome, Italy.
| | - Fabiana Rizzo
- Department of Infectious Diseases, Istituto Superiore di Sanità, Rome, Italy
| | - Sundararajan Srinivasan
- Institute of Experimental Neurology (INSpe), Division of Neuroscience, San Raffaele Scientific Institute, Milan, Italy
| | - Marco Di Dario
- Institute of Experimental Neurology (INSpe), Division of Neuroscience, San Raffaele Scientific Institute, Milan, Italy
| | - Elena Giacomini
- Department of Infectious Diseases, Istituto Superiore di Sanità, Rome, Italy
| | - Maria Chiara Buscarinu
- Center for Experimental Neurological Therapies, Sant'Andrea Hospital, Department of Neurosciences, Mental Health and Sensory Organs, Sapienza University, Rome, Italy
| | - Melania Cruciani
- Department of Infectious Diseases, Istituto Superiore di Sanità, Rome, Italy
| | - Marilena P Etna
- Department of Infectious Diseases, Istituto Superiore di Sanità, Rome, Italy
| | - Silvia Sandini
- Department of Infectious Diseases, Istituto Superiore di Sanità, Rome, Italy
| | - Rosella Mechelli
- Center for Experimental Neurological Therapies, Sant'Andrea Hospital, Department of Neurosciences, Mental Health and Sensory Organs, Sapienza University, Rome, Italy; Department of Human Science and Promotion of Quality of Life, San Raffaele Roma Open University and IRCCS San Raffaele-Pisana, Rome, Italy
| | - Antonella Farina
- Department of Experimental Medicine, Sapienza University, Rome, Italy
| | - Pankaj Trivedi
- Department of Experimental Medicine, Sapienza University, Rome, Italy
| | - Paul J Hertzog
- Department of Molecular and Translational Sciences, Monash University, Clayton, Australia
| | - Marco Salvetti
- Center for Experimental Neurological Therapies, Sant'Andrea Hospital, Department of Neurosciences, Mental Health and Sensory Organs, Sapienza University, Rome, Italy; Istituto Neurologico Mediterraneo (INM) Neuromed, Pozzilli, Isernia, Italy
| | - Cinthia Farina
- Institute of Experimental Neurology (INSpe), Division of Neuroscience, San Raffaele Scientific Institute, Milan, Italy
| | - Eliana M Coccia
- Department of Infectious Diseases, Istituto Superiore di Sanità, Rome, Italy.
| |
Collapse
|
37
|
Guan Y, Jakimovski D, Ramanathan M, Weinstock-Guttman B, Zivadinov R. The role of Epstein-Barr virus in multiple sclerosis: from molecular pathophysiology to in vivo imaging. Neural Regen Res 2019; 14:373-386. [PMID: 30539801 PMCID: PMC6334604 DOI: 10.4103/1673-5374.245462] [Citation(s) in RCA: 104] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2018] [Accepted: 08/31/2018] [Indexed: 12/29/2022] Open
Abstract
Multiple sclerosis (MS) is a disease of the central nervous system characterized by inflammation, demyelination, and neuronal damage. Environmental and genetic factors are associated with the risk of developing MS, but the exact cause still remains unidentified. Epstein-Barr virus (EBV), vitamin D, and smoking are among the most well-established environmental risk factors in MS. Infectious mononucleosis, which is caused by delayed primary EBV infection, increases the risk of developing MS. EBV may also contribute to MS pathogenesis indirectly by activating silent human endogenous retrovirus-W. The emerging B-cell depleting therapies, particularly anti-CD20 agents such as rituximab, ocrelizumab, as well as the fully human ofatumumab, have shown promising clinical and magnetic resonance imaging benefit. One potential effect of these therapies is the depletion of memory B-cells, the primary reservoir site where EBV latency occurs. In addition, EBV potentially interacts with both genetic and other environmental factors to increase susceptibility and disease severity of MS. This review examines the role of EBV in MS pathophysiology and summarizes the recent clinical and radiological findings, with a focus on B-cells and in vivo imaging. Addressing the potential link between EBV and MS allows the better understanding of MS pathogenesis and helps to identify additional disease biomarkers that may be responsive to B-cell depleting intervention.
Collapse
Affiliation(s)
- Yi Guan
- Buffalo Neuroimaging Analysis Center, Department of Neurology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, State University of New York, Buffalo, NY, USA
| | - Dejan Jakimovski
- Buffalo Neuroimaging Analysis Center, Department of Neurology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, State University of New York, Buffalo, NY, USA
| | - Murali Ramanathan
- Jacobs Comprehensive MS Treatment and Research Center, Department of Neurology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, State University of New York, Buffalo, NY, USA
- Department of Pharmaceutical Sciences, State University of New York, Buffalo, NY, USA
| | - Bianca Weinstock-Guttman
- Jacobs Comprehensive MS Treatment and Research Center, Department of Neurology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, State University of New York, Buffalo, NY, USA
| | - Robert Zivadinov
- Buffalo Neuroimaging Analysis Center, Department of Neurology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, State University of New York, Buffalo, NY, USA
- Center for Biomedical Imaging at Clinical Translational Science Institute, University at Buffalo, State University of New York, Buffalo, NY, USA
| |
Collapse
|
38
|
Monteiro A, Cruto C, Rosado P, Rosado L, Fonseca AM, Paiva A. Interferon-beta treated-multiple sclerosis patients exhibit a decreased ratio between immature/transitional B cell subset and plasmablasts. J Neuroimmunol 2019; 326:49-54. [DOI: 10.1016/j.jneuroim.2018.11.001] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2018] [Revised: 10/20/2018] [Accepted: 11/05/2018] [Indexed: 12/20/2022]
|
39
|
Severa M, Zhang J, Giacomini E, Rizzo F, Etna MP, Cruciani M, Garaci E, Chopp M, Coccia EM. Thymosins in multiple sclerosis and its experimental models: moving from basic to clinical application. Mult Scler Relat Disord 2019; 27:52-60. [PMID: 30317071 PMCID: PMC7104151 DOI: 10.1016/j.msard.2018.09.035] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2018] [Revised: 09/20/2018] [Accepted: 09/30/2018] [Indexed: 02/08/2023]
Abstract
BACKGROUND Multiple sclerosis (MS) afflicts more than 2.5 million individuals worldwide and this number is increasing over time. Within the past years, a great number of disease-modifying treatments have emerged; however, efficacious treatments and a cure for MS await discovery. Thymosins, soluble hormone-like peptides produced by the thymus gland, can mediate immune and non-immune physiological processes and have gained interest in recent years as therapeutics in inflammatory and autoimmune diseases. METHODS Pubmed was searched with no time constraints for articles using a combination of the keywords "thymosin/s" or "thymus factor/s" AND "multiple sclerosis", mesh terms with no language restriction. RESULTS Here, we review the state-of-the-art on the effects of thymosins on MS and its experimental models. In particular, we describe what is known in this field on the roles of thymosin-α1 (Tα1) and -β4 (Tβ4) as potential anti-inflammatory as well as neuroprotective and remyelinating molecules and their mechanisms of action. CONCLUSION Based on the data that Tα1 and Tβ4 act as anti-inflammatory molecules and as inducers of myelin repair and neuronal protection, respectively, a possible therapeutic application in MS for Tα1 and Tβ4 alone or combined with other approved drugs may be envisaged. This approach is reasonable in light of the current clinical usage of Tα1 and data demonstrating the safety, tolerability and efficacy of Tβ4 in clinical practice.
Collapse
Affiliation(s)
- Martina Severa
- Department of Infectious Diseases, Istituto Superiore di Sanità, Rome, Italy
| | - Jing Zhang
- Department of Neurology, Henry Ford Hospital, Detroit, MI, USA
| | - Elena Giacomini
- Department of Infectious Diseases, Istituto Superiore di Sanità, Rome, Italy
| | - Fabiana Rizzo
- Department of Infectious Diseases, Istituto Superiore di Sanità, Rome, Italy
| | - Marilena Paola Etna
- Department of Infectious Diseases, Istituto Superiore di Sanità, Rome, Italy
| | - Melania Cruciani
- Department of Infectious Diseases, Istituto Superiore di Sanità, Rome, Italy
| | - Enrico Garaci
- University San Raffaele and IRCCS San Raffaele, Rome, Italy
| | - Michael Chopp
- Department of Neurology, Henry Ford Hospital, Detroit, MI, USA,Department of Physics, Oakland University, Rochester, MI, USA
| | | |
Collapse
|
40
|
Jakimovski D, Kolb C, Ramanathan M, Zivadinov R, Weinstock-Guttman B. Interferon β for Multiple Sclerosis. Cold Spring Harb Perspect Med 2018; 8:cshperspect.a032003. [PMID: 29311124 DOI: 10.1101/cshperspect.a032003] [Citation(s) in RCA: 105] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Despite that the availability of new therapeutic options has expanded the multiple sclerosis (MS) disease-modifying therapy arsenal, interferon β (IFN-β) remains an important therapy option in the current decision-making process. This review will summarize the present knowledge of IFN-β mechanism of action, the overall safety, and the short- and long-term efficacy of its use in relapsing remitting MS and clinically isolated syndromes. Data on secondary progressive MS is also provided, although no clear benefit was identified.
Collapse
Affiliation(s)
- Dejan Jakimovski
- Buffalo Neuroimaging Analysis Center, Department of Neurology, University at Buffalo, State University of New York, Buffalo, New York 14203
| | - Channa Kolb
- Jacobs MS Center, Department of Neurology, University at Buffalo, State University of New York, Buffalo, New York 14202
| | - Murali Ramanathan
- Jacobs MS Center, Department of Neurology, University at Buffalo, State University of New York, Buffalo, New York 14202.,Department of Pharmaceutical Sciences, School of Medicine and Biomedical Sciences, State University of New York, Buffalo, New York 14214
| | - Robert Zivadinov
- Buffalo Neuroimaging Analysis Center, Department of Neurology, University at Buffalo, State University of New York, Buffalo, New York 14203.,MR Imaging Clinical Translational Research Center, School of Medicine and Biomedical Sciences, University at Buffalo, State University of New York, Buffalo, New York 14203
| | - Bianca Weinstock-Guttman
- Jacobs MS Center, Department of Neurology, University at Buffalo, State University of New York, Buffalo, New York 14202
| |
Collapse
|
41
|
Negron A, Robinson RR, Stüve O, Forsthuber TG. The role of B cells in multiple sclerosis: Current and future therapies. Cell Immunol 2018; 339:10-23. [PMID: 31130183 DOI: 10.1016/j.cellimm.2018.10.006] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2018] [Revised: 10/19/2018] [Accepted: 10/20/2018] [Indexed: 02/07/2023]
Abstract
While it was long held that T cells were the primary mediators of multiple sclerosis (MS) pathogenesis, the beneficial effects observed in response to treatment with Rituximab (RTX), a monoclonal antibody (mAb) targeting CD20, shed light on a key contributor to MS that had been previously underappreciated: B cells. This has been reaffirmed by results from clinical trials testing the efficacy of subsequently developed B cell-depleting mAbs targeting CD20 as well as studies revisiting the effects of previous disease-modifying therapies (DMTs) on B cell subsets thought to modulate disease severity. In this review, we summarize current knowledge regarding the complex roles of B cells in MS pathogenesis and current and potential future B cell-directed therapies.
Collapse
Affiliation(s)
- Austin Negron
- Department of Biology, University of Texas at San Antonio, TX 78249, USA
| | - Rachel R Robinson
- Department of Biology, University of Texas at San Antonio, TX 78249, USA
| | - Olaf Stüve
- Department of Neurology and Neurotherapeutics, University of Texas Southwestern Medical Center, Dallas, TX, USA; Neurology Section, VA North Texas Health Care System, Medical Service, Dallas, TX, USA
| | | |
Collapse
|
42
|
Annibali V, Umeton R, Palermo A, Severa M, Etna MP, Giglio S, Romano S, Ferraldeschi M, Buscarinu MC, Vecchione A, Annese A, Policano C, Mechelli R, Pizzolato Umeton R, Fornasiero A, Angelini DF, Guerrera G, Battistini L, Coccia EM, Salvetti M, Ristori G. Analysis of coding and non-coding transcriptome of peripheral B cells reveals an altered interferon response factor (IRF)-1 pathway in multiple sclerosis patients. J Neuroimmunol 2018; 324:165-171. [PMID: 30270021 DOI: 10.1016/j.jneuroim.2018.09.005] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2018] [Revised: 08/13/2018] [Accepted: 09/10/2018] [Indexed: 01/15/2023]
Abstract
Several evidences emphasize B-cell pathogenic roles in multiple sclerosis (MS). We performed transcriptome analyses on peripheral B cells from therapy-free patients and age/sex-matched controls. Down-regulation of two transcripts (interferon response factor 1-IRF1, and C-X-C motif chemokine 10-CXCL10), belonging to the same pathway, was validated by RT-PCR in 26 patients and 21 controls. IRF1 and CXCL10 transcripts share potential seeding sequences for hsa-miR-424, that resulted up-regulated in MS patients. We confirmed this interaction and its functional effect by transfection experiments. Consistent findings indicate down-regulation of IRF1/CXCL10 axis, that may plausibly contribute to a pro-survival status of B cells in MS.
Collapse
Affiliation(s)
- Viviana Annibali
- Center for Experimental Neurological Therapies, Sant'Andrea Hospital, Department of Neurosciences, Mental Health and Sensory Organs (NESMOS), Faculty of Medicine and Psychology, Sapienza University of Rome, Rome, Italy
| | - Renato Umeton
- Department of Informatics, Dana-Farber Cancer Institute, Boston, MA, United States; Massachusetts Institute of Technology, Cambridge, MA, United States
| | - Antonia Palermo
- Department of Mathematics and Computer Science, University of Calabria
| | - Martina Severa
- Department of Infectious, Parasitic and Immune-mediated Disease, Istituto Superiore di Sanità, Rome, Italy
| | - Marilena Paola Etna
- Department of Infectious, Parasitic and Immune-mediated Disease, Istituto Superiore di Sanità, Rome, Italy
| | - Simona Giglio
- Division of Pathology, Department of Clinical and Molecular Medicine, Sant'Andrea Hospital, Faculty of Medicine and Psychology, Sapienza University of Rome, Rome, Italy
| | - Silvia Romano
- Center for Experimental Neurological Therapies, Sant'Andrea Hospital, Department of Neurosciences, Mental Health and Sensory Organs (NESMOS), Faculty of Medicine and Psychology, Sapienza University of Rome, Rome, Italy
| | - Michela Ferraldeschi
- Center for Experimental Neurological Therapies, Sant'Andrea Hospital, Department of Neurosciences, Mental Health and Sensory Organs (NESMOS), Faculty of Medicine and Psychology, Sapienza University of Rome, Rome, Italy
| | - Maria Chiara Buscarinu
- Center for Experimental Neurological Therapies, Sant'Andrea Hospital, Department of Neurosciences, Mental Health and Sensory Organs (NESMOS), Faculty of Medicine and Psychology, Sapienza University of Rome, Rome, Italy
| | - Andrea Vecchione
- Division of Pathology, Department of Clinical and Molecular Medicine, Sant'Andrea Hospital, Faculty of Medicine and Psychology, Sapienza University of Rome, Rome, Italy
| | - Anita Annese
- Center for Experimental Neurological Therapies, Sant'Andrea Hospital, Department of Neurosciences, Mental Health and Sensory Organs (NESMOS), Faculty of Medicine and Psychology, Sapienza University of Rome, Rome, Italy
| | - Claudia Policano
- Center for Experimental Neurological Therapies, Sant'Andrea Hospital, Department of Neurosciences, Mental Health and Sensory Organs (NESMOS), Faculty of Medicine and Psychology, Sapienza University of Rome, Rome, Italy
| | - Rosella Mechelli
- Center for Experimental Neurological Therapies, Sant'Andrea Hospital, Department of Neurosciences, Mental Health and Sensory Organs (NESMOS), Faculty of Medicine and Psychology, Sapienza University of Rome, Rome, Italy
| | | | - Arianna Fornasiero
- Center for Experimental Neurological Therapies, Sant'Andrea Hospital, Department of Neurosciences, Mental Health and Sensory Organs (NESMOS), Faculty of Medicine and Psychology, Sapienza University of Rome, Rome, Italy
| | | | | | | | - Eliana Marina Coccia
- Department of Infectious, Parasitic and Immune-mediated Disease, Istituto Superiore di Sanità, Rome, Italy
| | - Marco Salvetti
- Center for Experimental Neurological Therapies, Sant'Andrea Hospital, Department of Neurosciences, Mental Health and Sensory Organs (NESMOS), Faculty of Medicine and Psychology, Sapienza University of Rome, Rome, Italy; IRCCS Istituto Neurologico Mediterraneo (INM) Neuromed (M.S.), Pozzilli, IS, Italy.
| | - Giovanni Ristori
- Center for Experimental Neurological Therapies, Sant'Andrea Hospital, Department of Neurosciences, Mental Health and Sensory Organs (NESMOS), Faculty of Medicine and Psychology, Sapienza University of Rome, Rome, Italy.
| |
Collapse
|
43
|
Li R, Patterson KR, Bar-Or A. Reassessing B cell contributions in multiple sclerosis. Nat Immunol 2018; 19:696-707. [PMID: 29925992 DOI: 10.1038/s41590-018-0135-x] [Citation(s) in RCA: 271] [Impact Index Per Article: 38.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2018] [Accepted: 05/09/2018] [Indexed: 02/06/2023]
Abstract
There is growing recognition that B cell contributions to normal immune responses extend well beyond their potential to become antibody-producing cells, including roles at the innate-adaptive interface and their potential to modulate the responses of other immune cells such as T cells and myeloid cells. These B cell functions can have both pathogenic and protective effects in the context of central nervous system (CNS) inflammation. Here, we review recent advances in the field of multiple sclerosis (MS), which has traditionally been viewed as primarily a T cell-mediated disease, and we consider antibody-dependent and, particularly, emerging antibody-independent functions of B cells that may be relevant in both the peripheral and CNS disease compartments.
Collapse
Affiliation(s)
- Rui Li
- Center for Neuroinflammation and Experimental Therapeutics (CNET) and Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Kristina R Patterson
- Center for Neuroinflammation and Experimental Therapeutics (CNET) and Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Amit Bar-Or
- Center for Neuroinflammation and Experimental Therapeutics (CNET) and Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
44
|
Janus revisited: The intricate role of the immune system in neovascularization. Int J Cardiol 2018; 260:193-194. [DOI: 10.1016/j.ijcard.2018.03.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/01/2018] [Accepted: 03/05/2018] [Indexed: 11/23/2022]
|
45
|
Dumitrescu L, Constantinescu CS, Tanasescu R. Recent developments in interferon-based therapies for multiple sclerosis. Expert Opin Biol Ther 2018; 18:665-680. [PMID: 29624084 DOI: 10.1080/14712598.2018.1462793] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
INTRODUCTION Multiple sclerosis (MS) is a chronic and disabling immune-mediated disease of the central nervous system. Beta-interferons are the first approved and still the most widely used first-line disease-modifying treatment in MS. AREAS COVERED Here we focus on recent developments in pharmacology and delivery systems of beta-interferons, and discuss their place within current state of the art therapeutic approaches. We briefly review the clinical trials for classical and PEGylated formulations, emphasizing effectiveness, safety concerns, and tolerability. The mechanisms of action of IFN-β in view of MS pathogenesis are also debated EXPERT OPINION Though only modestly efficient in reducing the annualized relapse rate, beta-interferons remain a valid first-line option due to their good long-term safety profile and cost-efficacy. Moreover, they are endogenous class II cytokines essential for mounting an effective antiviral response, and they may interact with putative MS triggering factors such as Epstein-Barr virus infection and human endogenous retroviruses. Recent improvements in formulations, delivery devices and drug regimens tackle the tolerability and adherence issues frequently seen with these drugs, and scientific advances may offer means for a better selection of patients. Although a well-established immunomodulatory treatment, beta-interferons have not said their last word in the management of MS.
Collapse
Affiliation(s)
- Laura Dumitrescu
- a Department of Clinical Neurosciences , University of Medicine and Pharmacy Carol Davila , Bucharest , Romania.,b Department of Neurology , Colentina Hospital , Bucharest , Romania
| | - Cris S Constantinescu
- c Academic Clinical Neurology, Division of Clinical Neuroscience , University of Nottingham , UK
| | - Radu Tanasescu
- a Department of Clinical Neurosciences , University of Medicine and Pharmacy Carol Davila , Bucharest , Romania.,b Department of Neurology , Colentina Hospital , Bucharest , Romania.,c Academic Clinical Neurology, Division of Clinical Neuroscience , University of Nottingham , UK
| |
Collapse
|
46
|
Hammer A, Waschbisch A, Kuhbandner K, Bayas A, Lee DH, Duscha A, Haghikia A, Gold R, Linker RA. The NRF2 pathway as potential biomarker for dimethyl fumarate treatment in multiple sclerosis. Ann Clin Transl Neurol 2018; 5:668-676. [PMID: 29928650 PMCID: PMC5989754 DOI: 10.1002/acn3.553] [Citation(s) in RCA: 63] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2017] [Revised: 01/29/2018] [Accepted: 02/17/2018] [Indexed: 12/11/2022] Open
Abstract
Objective Immunological studies have demonstrated a plethora of beneficial effects of dimethyl fumarate (DMF) on various cell types. However, the cellular and molecular targets are incompletely understood and response markers are scarce. Here, we focus on the relation between nuclear factor (erythroid‐derived 2)‐like 2 (NRF2) pathway induction under DMF therapy and the composition of the blood immune cell compartment and clinical efficacy in relapsing‐remitting multiple sclerosis (MS) patients. Methods We explored effects of DMF on peripheral immune cell subsets by flow cytometric and transcriptional analysis of serial blood samples obtained from 43 MS patients during the first year of therapy. Results Gene expression analysis proved activation of NRF2 signaling under DMF therapy that was paralleled by a temporal expansion of FoxP3+ regulatory T cells, CD56bright natural killer cells, plasmacytoid dendritic cells, and a decrease in CD8+ T cells, B cells, and type 1 myeloid dendritic cells. In a subgroup of 28 patients with completely available clinical data, individuals with higher levels of the NRF2 target gene NAD(P)H quinone dehydrogenase 1 (NQO1) 4–6 weeks after DMF therapy initiation were more likely to achieve no evidence of disease activity status 1 year later. The degree of NQO1 induction further correlated with patient age. Interpretation We demonstrate that positive effects of DMF on the clinical outcome are paralleled by induction of the antioxidant NRF2 transcriptional pathway and a shift toward regulatory immune cell subsets in the periphery. Our data identify a role of the NRF2 pathway as potential biomarker for DMF treatment in MS.
Collapse
Affiliation(s)
- Anna Hammer
- Department of Neurology University Hospital Erlangen Friedrich-Alexander-University Erlangen-Nürnberg Erlangen 91054 Germany
| | - Anne Waschbisch
- Department of Neurology University Hospital Erlangen Friedrich-Alexander-University Erlangen-Nürnberg Erlangen 91054 Germany.,Present address: Department of Neurology University Hospital Rheinisch-Westfälische Technische Hochschule (RWTH) Aachen Aachen 52074 Germany
| | - Kristina Kuhbandner
- Department of Neurology University Hospital Erlangen Friedrich-Alexander-University Erlangen-Nürnberg Erlangen 91054 Germany
| | - Antonios Bayas
- Department of Neurology Hospital Augsburg Augsburg 86156 Germany
| | - De-Hyung Lee
- Department of Neurology University Hospital Erlangen Friedrich-Alexander-University Erlangen-Nürnberg Erlangen 91054 Germany
| | - Alexander Duscha
- Department of Neurology Ruhr-University Bochum Bochum 44791 Germany
| | - Aiden Haghikia
- Department of Neurology Ruhr-University Bochum Bochum 44791 Germany
| | - Ralf Gold
- Department of Neurology Ruhr-University Bochum Bochum 44791 Germany
| | - Ralf A Linker
- Department of Neurology University Hospital Erlangen Friedrich-Alexander-University Erlangen-Nürnberg Erlangen 91054 Germany
| |
Collapse
|
47
|
Cavalcante P, Barzago C, Baggi F, Antozzi C, Maggi L, Mantegazza R, Bernasconi P. Toll-like receptors 7 and 9 in myasthenia gravis thymus: amplifiers of autoimmunity? Ann N Y Acad Sci 2018; 1413:11-24. [PMID: 29363775 DOI: 10.1111/nyas.13534] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2017] [Revised: 09/25/2017] [Accepted: 09/26/2017] [Indexed: 12/12/2022]
Abstract
Pathogen infections and dysregulated Toll-like receptor (TLR)-mediated innate immune responses are suspected to play key roles in autoimmunity. Among TLRs, TLR7 and TLR9 have been implicated in several autoimmune conditions, mainly because of their ability to promote abnormal B cell activation and survival. Recently, we provided evidence of Epstein-Barr virus (EBV) persistence and reactivation in the thymus of myasthenia gravis (MG) patients, suggesting an involvement of EBV in the intrathymic pathogenesis of the disease. Considerable data highlight the existence of pathogenic crosstalk among EBV, TLR7, and TLR9: EBV elicits TLR7/9 signaling, which in turn can enhance B cell dysfunction and autoimmunity. In this article, after a brief summary of data demonstrating TLR activation in MG thymus, we provide an overview on the contribution of TLR7 and TLR9 to autoimmune diseases and discuss our recent findings indicating a pivotal role for these two receptors, along with EBV, in driving, perpetuating, and/or amplifying intrathymic B cell dysregulation and autoimmune responses in MG. Development of therapeutic approaches targeting TLR7 and TLR9 signaling could be a novel strategy for treating the chronic inflammatory autoimmune process in myasthenia gravis.
Collapse
Affiliation(s)
- Paola Cavalcante
- Neurology IV, Neuroimmunology and Neuromuscular Diseases Unit, Fondazione Istituto Neurologico "Carlo Besta", Milan, Italy
| | - Claudia Barzago
- Neurology IV, Neuroimmunology and Neuromuscular Diseases Unit, Fondazione Istituto Neurologico "Carlo Besta", Milan, Italy
| | - Fulvio Baggi
- Neurology IV, Neuroimmunology and Neuromuscular Diseases Unit, Fondazione Istituto Neurologico "Carlo Besta", Milan, Italy
| | - Carlo Antozzi
- Neurology IV, Neuroimmunology and Neuromuscular Diseases Unit, Fondazione Istituto Neurologico "Carlo Besta", Milan, Italy
| | - Lorenzo Maggi
- Neurology IV, Neuroimmunology and Neuromuscular Diseases Unit, Fondazione Istituto Neurologico "Carlo Besta", Milan, Italy
| | - Renato Mantegazza
- Neurology IV, Neuroimmunology and Neuromuscular Diseases Unit, Fondazione Istituto Neurologico "Carlo Besta", Milan, Italy
| | - Pia Bernasconi
- Neurology IV, Neuroimmunology and Neuromuscular Diseases Unit, Fondazione Istituto Neurologico "Carlo Besta", Milan, Italy
| |
Collapse
|
48
|
Pavelek Z, Vyšata O, Klímová B, Andrýs C, Vokurková D, Vališ M. Lymphocytes in the treatment with interferon beta-1 b. Mult Scler Relat Disord 2017; 18:29-32. [PMID: 29141817 DOI: 10.1016/j.msard.2017.08.020] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2017] [Revised: 08/18/2017] [Accepted: 08/31/2017] [Indexed: 11/28/2022]
Abstract
BACKGROUND Multiple sclerosis (MS) is a chronic inflammatory demyelinating and neurodegenerative disease affecting the central nervous system. One of the basic medications for the treatment of a clinically isolated syndrome (CIS) or relapsing-remitting MS is interferon beta (INFβ). Although the exact mechanism of its effects is unknown, the medication has an anti-inflammatory and immunomodulatory effect. The goal of this study was to determine the characters which are affected in patients treated with INFβ. METHODS A total of 97 patients (25 males and 72 females) were included into the study. Patients were treated by INFβ 1-b (subcutaneous injection, 250µg, each other day). Clinical evaluations were performed by an attending neurologist. Peripheral blood samples were obtained just prior to treatment and 5 years after INFβ 1-b. Statistical analysis and processing of the obtained data were performed by using a comprehensive statistical software package MATLAB®. RESULTS A significant decrease of the observed parameters after 5 years' of treatment (significant at the 1% significance level) was found in the absolute and relative CD69 count, absolute cytotoxic/suppressor T lymphocyte count, absolute total leukocyte count, absolute natural killer cells count. A significant decrease of the observed parameters after 5 years' of treatment (significant at the 5% significance level) was found in the absolute lymphocyte count, relative cytotoxic/suppressor T lymphocyte count, relative CD3+CD69+ count and absolute CD8+CD38+ count. CONCLUSION The treatment with interferon beta reduces clinical exacerbations in multiple sclerosis (MS) through several known immunomodulatory mechanisms. However, the exact mechanism of effect of this medication is not known. This study presents some parameters that were affected by the long-term INFβ treatment.
Collapse
Affiliation(s)
- Zbyšek Pavelek
- Department of Neurology, Faculty of Medicine and University Hospital Hradec Králové, Charles University in Prague, Hradec Králové, Sokolská 581, 500 05, Czech Republic.
| | - Oldřich Vyšata
- Department of Neurology, Faculty of Medicine and University Hospital Hradec Králové, Charles University in Prague, Hradec Králové, Sokolská 581, 500 05, Czech Republic
| | - Blanka Klímová
- Department of Neurology, Faculty of Medicine and University Hospital Hradec Králové, Charles University in Prague, Hradec Králové, Sokolská 581, 500 05, Czech Republic
| | - Ctirad Andrýs
- Department of Clinical Immunology and Allergology, University Hospital Hradec Králové, Hradec Králové, Sokolská 581, 500 05, Czech Republic
| | - Doris Vokurková
- Department of Clinical Immunology and Allergology, University Hospital Hradec Králové, Hradec Králové, Sokolská 581, 500 05, Czech Republic
| | - Martin Vališ
- Department of Neurology, Faculty of Medicine and University Hospital Hradec Králové, Charles University in Prague, Hradec Králové, Sokolská 581, 500 05, Czech Republic
| |
Collapse
|
49
|
Role of Immunological Memory Cells as a Therapeutic Target in Multiple Sclerosis. Brain Sci 2017; 7:brainsci7110148. [PMID: 29112130 PMCID: PMC5704155 DOI: 10.3390/brainsci7110148] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2017] [Revised: 10/31/2017] [Accepted: 11/02/2017] [Indexed: 12/14/2022] Open
Abstract
Pharmacological targeting of memory cells is an attractive treatment strategy in various autoimmune diseases, such as psoriasis and rheumatoid arthritis. Multiple sclerosis is the most common inflammatory disorder of the central nervous system, characterized by focal immune cell infiltration, activation of microglia and astrocytes, along with progressive damage to myelin sheaths, axons, and neurons. The current review begins with the identification of memory cell types in the previous literature and a recent description of the modulation of these cell types in T, B, and resident memory cells in the presence of different clinically approved multiple sclerosis drugs. Overall, this review paper tries to determine the potential of memory cells to act as a target for the current or newly-developed drugs.
Collapse
|
50
|
Deciphering the Role of B Cells in Multiple Sclerosis-Towards Specific Targeting of Pathogenic Function. Int J Mol Sci 2017; 18:ijms18102048. [PMID: 28946620 PMCID: PMC5666730 DOI: 10.3390/ijms18102048] [Citation(s) in RCA: 59] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2017] [Revised: 09/19/2017] [Accepted: 09/20/2017] [Indexed: 12/25/2022] Open
Abstract
B cells, plasma cells and antibodies may play a key role in the pathogenesis of multiple sclerosis (MS). This notion is supported by various immunological changes observed in MS patients, such as activation and pro-inflammatory differentiation of peripheral blood B cells, the persistence of clonally expanded plasma cells producing immunoglobulins in the cerebrospinal fluid, as well as the composition of inflammatory central nervous system lesions frequently containing co-localizing antibody depositions and activated complement. In recent years, the perception of a respective pathophysiological B cell involvement was vividly promoted by the empirical success of anti-CD20-mediated B cell depletion in clinical trials; based on these findings, the first monoclonal anti-CD20 antibody—ocrelizumab—is currently in the process of being approved for treatment of MS. In this review, we summarize the current knowledge on the role of B cells, plasma cells and antibodies in MS and elucidate how approved and future treatments, first and foremost anti-CD20 antibodies, therapeutically modify these B cell components. We will furthermore describe regulatory functions of B cells in MS and discuss how the evolving knowledge of these therapeutically desirable B cell properties can be harnessed to improve future safety and efficacy of B cell-directed therapy in MS.
Collapse
|