1
|
Dalmau J, Dalakas MC, Kolson DL, Pröbstel AK, Paul F, Zamvil SS. Ten Years of Neurology® Neuroimmunology & Neuroinflammation: Decade in Review. NEUROLOGY(R) NEUROIMMUNOLOGY & NEUROINFLAMMATION 2025; 12:e200363. [PMID: 39724529 DOI: 10.1212/nxi.0000000000200363] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/28/2024]
Affiliation(s)
- Josep Dalmau
- IDIBAPS-CaixaResearch Institute, University Hospital Clínic of Barcelona, Barcelona, Spain
- University of Pennsylvania, Philadelphia
| | - Marinos C Dalakas
- University of Athens Medical School, Greece
- Jefferson University, Philadelphia, PA
| | | | - Anne-Katrin Pröbstel
- Departments of Neurology, University Hospital of Basel, Switzerland
- Departments of Biomedicine and Clinical Research, University Hospital Basel and University of Basel, Switzerland
- Research Center for Clinical Neuroimmunology and Neuroscience Basel (RC2NB), University Hospital Basel and University of Basel, Switzerland
| | | | - Scott S Zamvil
- Department of Neurology, University of California, San Francisco
| |
Collapse
|
2
|
Evans EF, Saraph A, Tokuyama M. Transactivation of Human Endogenous Retroviruses by Viruses. Viruses 2024; 16:1649. [PMID: 39599764 PMCID: PMC11599155 DOI: 10.3390/v16111649] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2024] [Revised: 10/12/2024] [Accepted: 10/16/2024] [Indexed: 11/29/2024] Open
Abstract
Human endogenous retroviruses (HERVs) are remnants of ancient retroviral infections that are part the human genome and are normally silenced through epigenetic mechanisms. However, HERVs can be induced by various host and environmental factors, including viral infection, and transcriptionally active HERVs have been implicated in various physiological processes. In this review, we summarize mounting evidence of transactivation of HERVs by a wide range of DNA and RNA viruses. Though a mechanistic understanding of this phenomenon and the biological implications are still largely missing, the link between exogenous and endogenous viruses is intriguing. Considering the increasing recognition of the role of viral infections in disease, understanding these interactions provides novel insights into human health.
Collapse
Affiliation(s)
| | | | - Maria Tokuyama
- Department of Microbiology and Immunology, Life Sciences Institute, The University of British Columbia, Vancouver, BC V6T 1Z4, Canada
| |
Collapse
|
3
|
Stys PK, Tsutsui S, Gafson AR, ‘t Hart BA, Belachew S, Geurts JJG. New views on the complex interplay between degeneration and autoimmunity in multiple sclerosis. Front Cell Neurosci 2024; 18:1426231. [PMID: 39161786 PMCID: PMC11330826 DOI: 10.3389/fncel.2024.1426231] [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: 05/01/2024] [Accepted: 06/14/2024] [Indexed: 08/21/2024] Open
Abstract
Multiple sclerosis (MS) is a frequently disabling neurological disorder characterized by symptoms, clinical signs and imaging abnormalities that typically fluctuate over time, affecting any level of the CNS. Prominent lymphocytic inflammation, many genetic susceptibility variants involving immune pathways, as well as potent responses of the neuroinflammatory component to immunomodulating drugs, have led to the natural conclusion that this disease is driven by a primary autoimmune process. In this Hypothesis and Theory article, we discuss emerging data that cast doubt on this assumption. After three decades of therapeutic experience, what has become clear is that potent immune modulators are highly effective at suppressing inflammatory relapses, yet exhibit very limited effects on the later progressive phase of MS. Moreover, neuropathological examination of MS tissue indicates that degeneration, CNS atrophy, and myelin loss are most prominent in the progressive stage, when lymphocytic inflammation paradoxically wanes. Finally, emerging clinical observations such as "progression independent of relapse activity" and "silent progression," now thought to take hold very early in the course, together argue that an underlying "cytodegenerative" process, likely targeting the myelinating unit, may in fact represent the most proximal step in a complex pathophysiological cascade exacerbated by an autoimmune inflammatory overlay. Parallels are drawn with more traditional neurodegenerative disorders, where a progressive proteopathy with prion-like propagation of toxic misfolded species is now known to play a key role. A potentially pivotal contribution of the Epstein-Barr virus and B cells in this process is also discussed.
Collapse
Affiliation(s)
- Peter K. Stys
- Department of Clinical Neurosciences, Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Shigeki Tsutsui
- Department of Clinical Neurosciences, Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Arie R. Gafson
- Biogen Digital Health, Biogen, Cambridge, MA, United States
| | - Bert A. ‘t Hart
- Department of Anatomy and Neurosciences, Amsterdam University Medical Centers (location VUmc), Amsterdam, Netherlands
| | - Shibeshih Belachew
- TheraPanacea, Paris, France
- Indivi (DBA of Healios AG), Basel, Switzerland
| | - Jeroen J. G. Geurts
- Department of Anatomy and Neurosciences, Amsterdam University Medical Centers (location VUmc), Amsterdam, Netherlands
| |
Collapse
|
4
|
Soldan SS, Su C, Monaco MC, Yoon L, Kannan T, Zankharia U, Patel RJ, Dheekollu J, Vladimirova O, Dowling JW, Thebault S, Brown N, Clauze A, Andrada F, Feder A, Planet PJ, Kossenkov A, Schäffer DE, Ohayon J, Auslander N, Jacobson S, Lieberman PM. Multiple sclerosis patient-derived spontaneous B cells have distinct EBV and host gene expression profiles in active disease. Nat Microbiol 2024; 9:1540-1554. [PMID: 38806670 DOI: 10.1038/s41564-024-01699-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Accepted: 04/11/2024] [Indexed: 05/30/2024]
Abstract
Epstein-Barr virus (EBV) is an aetiologic risk factor for the development of multiple sclerosis (MS). However, the role of EBV-infected B cells in the immunopathology of MS is not well understood. Here we characterized spontaneous lymphoblastoid cell lines (SLCLs) isolated from MS patients and healthy controls (HC) ex vivo to study EBV and host gene expression in the context of an individual's endogenous EBV. SLCLs derived from MS patient B cells during active disease had higher EBV lytic gene expression than SLCLs from MS patients with stable disease or HCs. Host gene expression analysis revealed activation of pathways associated with hypercytokinemia and interferon signalling in MS SLCLs and upregulation of forkhead box protein 1 (FOXP1), which contributes to EBV lytic gene expression. We demonstrate that antiviral approaches targeting EBV replication decreased cytokine production and autologous CD4+ T cell responses in this ex vivo model. These data suggest that dysregulation of intrinsic B cell control of EBV gene expression drives a pro-inflammatory, pathogenic B cell phenotype that can be attenuated by suppressing EBV lytic gene expression.
Collapse
Affiliation(s)
| | - Chenhe Su
- The Wistar Institute, Philadelphia, PA, USA
| | - Maria Chiara Monaco
- Neuroimmunology Branch, National Institute of Neurological Disorders and Stroke, NIH, Bethesda, MD, USA
| | - Leena Yoon
- The Wistar Institute, Philadelphia, PA, USA
| | | | | | | | | | | | - Jack W Dowling
- The Wistar Institute, Philadelphia, PA, USA
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Simon Thebault
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | | | - Annaliese Clauze
- Neuroimmunology Clinic, National Institute of Neurological Disorders and Stroke, NIH, Bethesda, MD, USA
| | - Frances Andrada
- Neuroimmunology Clinic, National Institute of Neurological Disorders and Stroke, NIH, Bethesda, MD, USA
| | - Andries Feder
- Division of Infectious Diseases, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Paul J Planet
- Division of Infectious Diseases, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | | | | | - Joan Ohayon
- Neuroimmunology Clinic, National Institute of Neurological Disorders and Stroke, NIH, Bethesda, MD, USA
| | | | - Steven Jacobson
- Neuroimmunology Branch, National Institute of Neurological Disorders and Stroke, NIH, Bethesda, MD, USA
| | | |
Collapse
|
5
|
James LM, Tsilibary EP, Wanberg EJ, Georgopoulos AP. Negative Association of Cognitive Performance With Blood Serum Neurotoxicity and Its Modulation by Human Herpes Virus 5 (HHV5) Seropositivity in Healthy Women. Neurosci Insights 2024; 19:26331055241258436. [PMID: 38827247 PMCID: PMC11143810 DOI: 10.1177/26331055241258436] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Accepted: 05/15/2024] [Indexed: 06/04/2024] Open
Abstract
Identification of early influences on cognitive decline is of paramount importance in order to stem the impacts of decrements in cognitive functioning and to potentially intervene. Thus, here we focused on 132 healthy adult women (age range 26-98 years) to (a) determine whether factors circulating in serum may exert neurotoxic effects in vitro, (b) evaluate associations between serum neurotoxicity and cognitive performance, and (c) assess the influence of human herpes virus (HHV) seroprevalence and other factors on apoptosis and cognitive performance. The results documented that the addition of serum from healthy adult women to neural cell cultures resulted in apoptosis, indicating the presence of circulating neurotoxic factors in the serum. Furthermore, apoptosis increased with age, and was associated with decreased cognitive performance. Stepwise regression evaluating the influence of 6 HHVs on apoptosis and cognitive function revealed that only HHV5 (cytomegalovirus; CMV) seropositivity was significantly associated with apoptosis and cognitive decline, controlling for age. These findings document neurotoxic effects of serum from healthy women across the adult lifespan and suggest a unique detrimental influence associated with CMV seropositivity.
Collapse
Affiliation(s)
- Lisa M James
- The Healthy Brain Aging Group, Brain Sciences Center, Department of Veterans Affairs Health Care System, Minneapolis, MN, USA
- Department of Neuroscience, University of Minnesota Medical School, Minneapolis, MN, USA
- Department of Psychiatry, University of Minnesota Medical School, Minneapolis, MN, USA
| | - Effie-Photini Tsilibary
- The Healthy Brain Aging Group, Brain Sciences Center, Department of Veterans Affairs Health Care System, Minneapolis, MN, USA
- Department of Neuroscience, University of Minnesota Medical School, Minneapolis, MN, USA
| | - Erik J Wanberg
- The Healthy Brain Aging Group, Brain Sciences Center, Department of Veterans Affairs Health Care System, Minneapolis, MN, USA
| | - Apostolos P Georgopoulos
- The Healthy Brain Aging Group, Brain Sciences Center, Department of Veterans Affairs Health Care System, Minneapolis, MN, USA
- Department of Neuroscience, University of Minnesota Medical School, Minneapolis, MN, USA
- Department of Psychiatry, University of Minnesota Medical School, Minneapolis, MN, USA
- Department of Neurology, University of Minnesota Medical School, Minneapolis, MN, USA
| |
Collapse
|
6
|
Münz C. Altered EBV specific immune control in multiple sclerosis. J Neuroimmunol 2024; 390:578343. [PMID: 38615370 DOI: 10.1016/j.jneuroim.2024.578343] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2024] [Revised: 03/23/2024] [Accepted: 04/04/2024] [Indexed: 04/16/2024]
Abstract
Since the 1980s it is known that immune responses to the Epstein-Barr virus (EBV) are elevated in multiple sclerosis (MS) patients. Recent seroepidemiologial data have shown that this alteration after primary EBV infection identifies individuals with a more than 30-fold increased risk to develop MS. The mechanisms by which EBV infection might erode tolerance for the central nervous system (CNS) in these individuals, years prior to clinical MS onset, remain unclear. In this review I will discuss altered frequencies of EBV life cycle stages and their tissue distribution, EBV with CNS autoantigen cross-reactive immune responses and loss of immune control for autoreactive B and T cells as possible mechanisms. This discussion is intended to stimulate future studies into these mechanisms with the aim to identify candidates for interventions that might correct EBV specific immune control and/or resulting cross-reactivities with CNS autoantigens in MS patients and thereby ameliorate disease activity.
Collapse
Affiliation(s)
- Christian Münz
- Viral Immunobiology, Institute of Experimental Immunology, University of Zürich, Switzerland.
| |
Collapse
|
7
|
Serafini B, Benincasa L, Rosicarelli B, Aloisi F. EBV infected cells in the multiple sclerosis brain express PD-L1: How the virus and its niche may escape immune surveillance. J Neuroimmunol 2024; 389:578314. [PMID: 38422689 DOI: 10.1016/j.jneuroim.2024.578314] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Revised: 01/23/2024] [Accepted: 02/13/2024] [Indexed: 03/02/2024]
Abstract
The presence of EBV infected B cells in postmortem multiple sclerosis (MS) brain tissue suggests immune evasion strategies. Using immunohistochemical techniques we analysed the expression of the immune checkpoint molecule PD-L1 and its receptor PD-1 in MS brains containing B cell-enriched perivascular infiltrates and meningeal follicles, a major EBV reservoir. PD-1 and PD-L1 immunoreactivities were restricted to CNS-infiltrating immune cells. PD-L1 was expressed on B cells, including EBV infected B cells, while PD-1 was expressed on many CD8+ T cells, including EBV-specific CD8+ T-cells, and fewer CD4+ T cells. PD-L1+ cells and EBV infected cells were in close contact with PD-1+ T cells. PD-L1 expressed by EBV infected B cells could favour local immune evasion leading to EBV persistence and immunopathology in the MS brain.
Collapse
Affiliation(s)
- Barbara Serafini
- Department of Neuroscience, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161 Rome, Italy.
| | - Lucia Benincasa
- Department of Neuroscience, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161 Rome, Italy.
| | - Barbara Rosicarelli
- Department of Neuroscience, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161 Rome, Italy.
| | - Francesca Aloisi
- Department of Neuroscience, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161 Rome, Italy.
| |
Collapse
|
8
|
Alghanimy A, Work LM, Holmes WM. The glymphatic system and multiple sclerosis: An evolving connection. Mult Scler Relat Disord 2024; 83:105456. [PMID: 38266608 DOI: 10.1016/j.msard.2024.105456] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2024] [Accepted: 01/18/2024] [Indexed: 01/26/2024]
Abstract
Multiple sclerosis (MS) is a complex autoimmune disorder that affects the central nervous system, resulting in demyelination and an array of neurological manifestations. Recently, there has been significant scientific interest in the glymphatic system, which operates as a waste-clearance system for the brain. This article reviews the existing literature, and explores potential links between the glymphatic system and MS, shedding light on its evolving significance in the context of MS pathogenesis. The authors consider the pathophysiological implications of glymphatic dysfunction in MS, the impact of disrupted sleep on glymphatic function, and the bidirectional relationship between MS and sleep disturbances. By offering an understanding of the intricate interplay between the glymphatic system and MS, this review provides valuable insights which may lead to improved diagnostic techniques and more effective therapeutic interventions.
Collapse
Affiliation(s)
- Alaa Alghanimy
- School of Psychology and Neuroscience, College of Medicine, Veterinary and Life Science, University of Glasgow, Glasgow G61 1QH, United Kingdom; Radiological Sciences Department, College of Applied Medical Sciences, King Saud University, Riyadh, Saudi Arabia.
| | - Lorraine M Work
- School of Cardiovascular and Metabolic Health, College of Medicine, Veterinary and Life Science, University of Glasgow, Glasgow G12 8TA, United Kingdom
| | - William M Holmes
- School of Psychology and Neuroscience, College of Medicine, Veterinary and Life Science, University of Glasgow, Glasgow G61 1QH, United Kingdom
| |
Collapse
|
9
|
Gottlieb A, Pham HPT, Saltarrelli JG, Lindsey JW. Expanded T lymphocytes in the cerebrospinal fluid of multiple sclerosis patients are specific for Epstein-Barr-virus-infected B cells. Proc Natl Acad Sci U S A 2024; 121:e2315857121. [PMID: 38190525 PMCID: PMC10801919 DOI: 10.1073/pnas.2315857121] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2023] [Accepted: 11/27/2023] [Indexed: 01/10/2024] Open
Abstract
Epstein-Barr virus (EBV) infection has long been associated with multiple sclerosis (MS), but the role of EBV in the pathogenesis of MS is not clear. Our hypothesis is that a major fraction of the expanded clones of T lymphocytes in the cerebrospinal fluid (CSF) are specific for autologous EBV-infected B cells. We obtained blood and CSF samples from eight relapsing-remitting patients in the process of diagnosis. We stimulated cells from the blood with autologous EBV-infected lymphoblastoid cell lines (LCL), EBV, varicella zoster virus, influenza, and candida and sorted the responding cells with flow cytometry after 6 d. We sequenced the RNA for T cell receptors (TCR) from CSF, unselected blood cells, and the antigen-specific cells. We used the TCR Vβ CDR3 sequences from the antigen-specific cells to assign antigen specificity to the sequences from the CSF and blood. LCL-specific cells comprised 13.0 ± 4.3% (mean ± SD) of the total reads present in CSF and 13.3 ± 7.5% of the reads present in blood. The next most abundant antigen specificity was flu, which was 4.7 ± 1.7% of the reads in the CSF and 9.3 ± 6.6% in the blood. The prominence of LCL-specific reads was even more marked in the top 1% most abundant CSF clones with statistically significant 47% mean overlap with LCL. We conclude that LCL-specific sequences form a major portion of the TCR repertoire in both CSF and blood and that expanded clones specific for LCL are present in MS CSF. This has important implications for the pathogenesis of MS.
Collapse
Affiliation(s)
- Assaf Gottlieb
- Center for Precision Health, School of Biomedical Informatics, University of Texas Health Science Center at Houston, Houston, TX77030
| | - H. Phuong T. Pham
- Division of Multiple Sclerosis and Neuroimmunology, Department of Neurology, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX77030
| | - Jerome G. Saltarrelli
- Department of Surgery, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX77030
| | - J. William Lindsey
- Division of Multiple Sclerosis and Neuroimmunology, Department of Neurology, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX77030
| |
Collapse
|
10
|
Papetti L, Panella E, Monte G, Ferilli MAN, Tarantino S, Checchi MP, Valeriani M. Pediatric Onset Multiple Sclerosis and Obesity: Defining the Silhouette of Disease Features in Overweight Patients. Nutrients 2023; 15:4880. [PMID: 38068737 PMCID: PMC10707944 DOI: 10.3390/nu15234880] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2023] [Revised: 11/13/2023] [Accepted: 11/20/2023] [Indexed: 12/18/2023] Open
Abstract
Obesity has been suggested as an environmental risk factor for multiple sclerosis (MS) and may negatively effect the progression of the disease. The aim of this study is to determine any correlation between overweight/obesity and the clinical and neuroradiological features at the onset of pediatric onset multiple sclerosis (POMS). Were included patients referred to the POMS Unit of the Bambino Gesù Children's Hospital between June 2012 and June 2021. The diagnosis of MS with an onset of less than 18 years was required. For all included subjects, we considered for the analysis the following data at the onset of symptoms: general data (age, sex, functional system compromised by neurological signs, weight and height), brain and spinal magnetic resonance imaging (MRI), cerebrospinal fluid exams. We identified 55 pediatric cases of POMS and divided them into two groups according to the body mass index (BMI): 60% were healthy weight (HW) and 40% were overweight/obese (OW/O). OW/O patients experienced a two-year age difference in disease onset compared to the HW patients (12.7 ± 3.8 years vs. 14.6 ± 4.1 years; p < 0.05). Onset of polyfocal symptoms was seen more frequently in OW/O patients than in HW (72.7% vs. 21.2%; p < 0.05). The pyramidal functions were involved more frequently in the OW/O group than in the HW group (50% vs. 25%; p < 0.005). Black holes were detected more frequently in OW/O patients in onset MRI scans compared to the HW group (50% vs. 15.5%; p < 0.05). Our findings suggest that being overweight/obese affects the risk of developing MS at an earlier age and is associated with an unfavorable clinical-radiological features at onset. Weight control can be considered as a preventive/therapeutic treatment.
Collapse
Affiliation(s)
- Laura Papetti
- Developmental Neurology Unit, Bambino Gesù Children’s Hospital, IRCCS, 00165 Rome, Italy; (G.M.); (M.A.N.F.); (S.T.); (M.P.C.); (M.V.)
| | - Elena Panella
- Child Neurology and Psychiatry Unit, Systems Medicine Department, Hospital of Rome, Tor Vergata University, 00133 Rome, Italy;
| | - Gabriele Monte
- Developmental Neurology Unit, Bambino Gesù Children’s Hospital, IRCCS, 00165 Rome, Italy; (G.M.); (M.A.N.F.); (S.T.); (M.P.C.); (M.V.)
| | - Michela Ada Noris Ferilli
- Developmental Neurology Unit, Bambino Gesù Children’s Hospital, IRCCS, 00165 Rome, Italy; (G.M.); (M.A.N.F.); (S.T.); (M.P.C.); (M.V.)
| | - Samuela Tarantino
- Developmental Neurology Unit, Bambino Gesù Children’s Hospital, IRCCS, 00165 Rome, Italy; (G.M.); (M.A.N.F.); (S.T.); (M.P.C.); (M.V.)
| | - Martina Proietti Checchi
- Developmental Neurology Unit, Bambino Gesù Children’s Hospital, IRCCS, 00165 Rome, Italy; (G.M.); (M.A.N.F.); (S.T.); (M.P.C.); (M.V.)
| | - Massimiliano Valeriani
- Developmental Neurology Unit, Bambino Gesù Children’s Hospital, IRCCS, 00165 Rome, Italy; (G.M.); (M.A.N.F.); (S.T.); (M.P.C.); (M.V.)
- Center for Sensory Motor Interaction, Aalborg University, DK-9220 Aalborg, Denmark
| |
Collapse
|
11
|
Hedström AK. Risk factors for multiple sclerosis in the context of Epstein-Barr virus infection. Front Immunol 2023; 14:1212676. [PMID: 37554326 PMCID: PMC10406387 DOI: 10.3389/fimmu.2023.1212676] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2023] [Accepted: 06/26/2023] [Indexed: 08/10/2023] Open
Abstract
Compelling evidence indicates that Epstein Barr virus (EBV) infection is a prerequisite for multiple sclerosis (MS). The disease may arise from a complex interplay between latent EBV infection, genetic predisposition, and various environmental and lifestyle factors that negatively affect immune control of the infection. Evidence of gene-environment interactions and epigenetic modifications triggered by environmental factors in genetically susceptible individuals supports this view. This review gives a short introduction to EBV and host immunity and discusses evidence indicating EBV as a prerequisite for MS. The role of genetic and environmental risk factors, and their interactions, in MS pathogenesis is reviewed and put in the context of EBV infection. Finally, possible preventive measures are discussed based on the findings presented.
Collapse
Affiliation(s)
- Anna Karin Hedström
- Department of Clinical Neuroscience, Karolinska Institute, Stockholm, Sweden
| |
Collapse
|
12
|
Andersen O, Ernberg I, Hedström AK. Treatment Options for Epstein-Barr Virus-Related Disorders of the Central Nervous System. Infect Drug Resist 2023; 16:4599-4620. [PMID: 37465179 PMCID: PMC10351589 DOI: 10.2147/idr.s375624] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2023] [Accepted: 06/28/2023] [Indexed: 07/20/2023] Open
Abstract
Epstein-Barr virus (EBV), a causative agent for several types of lymphomas and mucosal cancers, is a human lymphotropic herpesvirus with the capacity to establish lifelong latent infection. More than 90% of the human population worldwide is infected. The primary infection is usually asymptomatic in childhood, whereas infectious mononucleosis (IM) is common when the infection occurs in adolescence. Primary EBV infection, with or without IM, or reactivation of latent infection in immunocompromised individuals have been associated with a wide range of neurologic conditions, such as encephalitis, meningitis, acute disseminated encephalomyelitis, and cerebellitis. EBV is also involved in malignant lymphomas in the brain. An increasing number of reports on EBV-related disorders of the central nervous system (CNS) including the convincing association with multiple sclerosis (MS) have put in focus EBV-related conditions beyond its established link to malignancies. In this review, we present the clinical manifestations of EBV-related CNS-disorders, put them in the context of known EBV biology and focus on available treatment options and future therapeutic approaches.
Collapse
Affiliation(s)
- Oluf Andersen
- Department of Clinical Neuroscience, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Ingemar Ernberg
- Department of Microbiology, Tumor and Cell Biology, Biomedicum Q8C, Karolinska Institutet, Stockholm, 171 77, Sweden
| | - Anna Karin Hedström
- Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
13
|
Lambert N, El Moussaoui M, Baron F, Maquet P, Darcis G. Virus-Specific T-Cell Therapy for Viral Infections of the Central Nervous System: A Review. Viruses 2023; 15:1510. [PMID: 37515196 PMCID: PMC10383098 DOI: 10.3390/v15071510] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2023] [Revised: 06/29/2023] [Accepted: 07/03/2023] [Indexed: 07/30/2023] Open
Abstract
Opportunistic viral infections of the central nervous system represent a significant cause of morbidity and mortality among an increasing number of immunocompromised patients. Since antiviral treatments are usually poorly effective, the prognosis generally relies on the ability to achieve timely immune reconstitution. Hence, strategies aimed at reinvigorating antiviral immune activity have recently emerged. Among these, virus-specific T-cells are increasingly perceived as a principled and valuable tool to treat opportunistic viral infections. Here we briefly discuss how to develop and select virus-specific T-cells, then review their main indications in central nervous system infections, including progressive multifocal leukoencephalopathy, CMV infection, and adenovirus infection. We also discuss their potential interest in the treatment of progressive multiple sclerosis, or EBV-associated central nervous system inflammatory disease. We finish with the key future milestones of this promising treatment strategy.
Collapse
Affiliation(s)
- Nicolas Lambert
- Department of Neurology, University Hospital of Liège, 4000 Liège, Belgium
| | - Majdouline El Moussaoui
- Department of General Internal Medicine and Infectious Diseases, University Hospital of Liège, 4000 Liège, Belgium
| | - Frédéric Baron
- Department of Hematology, University Hospital of Liège, 4000 Liège, Belgium
| | - Pierre Maquet
- Department of Neurology, University Hospital of Liège, 4000 Liège, Belgium
| | - Gilles Darcis
- Department of General Internal Medicine and Infectious Diseases, University Hospital of Liège, 4000 Liège, Belgium
| |
Collapse
|
14
|
Debuysschere C, Nekoua MP, Hober D. Markers of Epstein-Barr Virus Infection in Patients with Multiple Sclerosis. Microorganisms 2023; 11:1262. [PMID: 37317236 DOI: 10.3390/microorganisms11051262] [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: 03/31/2023] [Revised: 05/09/2023] [Accepted: 05/09/2023] [Indexed: 06/16/2023] Open
Abstract
Viral infections have been suspected of being involved in the pathogenesis of certain autoimmune diseases for many years. Epstein-Barr virus (EBV), a DNA virus belonging to the Herpesviridae family, is thought to be associated with the onset and/or the progression of multiple sclerosis (MS), systemic lupus erythematosus, rheumatoid arthritis, Sjögren's syndrome and type 1 diabetes. The lifecycle of EBV consists of lytic cycles and latency programmes (0, I, II and III) occurring in infected B-cells. During this lifecycle, viral proteins and miRNAs are produced. This review provides an overview of the detection of EBV infection, focusing on markers of latency and lytic phases in MS. In MS patients, the presence of latency proteins and antibodies has been associated with lesions and dysfunctions of the central nervous system (CNS). In addition, miRNAs, expressed during lytic and latency phases, may be detected in the CNS of MS patients. Lytic reactivations of EBV can occur in the CNS of patients as well, with the presence of lytic proteins and T-cells reacting to this protein in the CNS of MS patients. In conclusion, markers of EBV infection can be found in MS patients, which argues in favour of a relationship between EBV and MS.
Collapse
Affiliation(s)
- Cyril Debuysschere
- Laboratoire de Virologie ULR3610, Université de Lille, CHU Lille, 59000 Lille, France
| | | | - Didier Hober
- Laboratoire de Virologie ULR3610, Université de Lille, CHU Lille, 59000 Lille, France
| |
Collapse
|
15
|
Teleanu RI, Niculescu AG, Vladacenco OA, Roza E, Perjoc RS, Teleanu DM. The State of the Art of Pediatric Multiple Sclerosis. Int J Mol Sci 2023; 24:ijms24098251. [PMID: 37175954 PMCID: PMC10179691 DOI: 10.3390/ijms24098251] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Revised: 04/28/2023] [Accepted: 05/01/2023] [Indexed: 05/15/2023] Open
Abstract
Multiple sclerosis (MS) represents a chronic immune-mediated neurodegenerative disease of the central nervous system that generally debuts around the age of 20-30 years. Still, in recent years, MS has been increasingly recognized among the pediatric population, being characterized by several peculiar features compared to adult-onset disease. Unfortunately, the etiology and disease mechanisms are poorly understood, rendering the already limited MS treatment options with uncertain efficacy and safety in pediatric patients. Thus, this review aims to shed some light on the progress in MS therapeutic strategies specifically addressed to children and adolescents. In this regard, the present paper briefly discusses the etiology, risk factors, comorbidities, and diagnosis possibilities for pediatric-onset MS (POMS), further moving to a detailed presentation of current treatment strategies, recent clinical trials, and emerging alternatives. Particularly, promising care solutions are indicated, including new treatment formulations, stem cell therapies, and cognitive training methods.
Collapse
Affiliation(s)
- Raluca Ioana Teleanu
- "Carol Davila" University of Medicine and Pharmacy, 020021 Bucharest, Romania
- Department of Pediatric Neurology, "Dr. Victor Gomoiu" Children's Hospital, 022102 Bucharest, Romania
| | - Adelina-Gabriela Niculescu
- Research Institute of the University of Bucharest-ICUB, University of Bucharest, 050657 Bucharest, Romania
- Department of Science and Engineering of Oxide Materials and Nanomaterials, Politehnica University of Bucharest, 011061 Bucharest, Romania
| | - Oana Aurelia Vladacenco
- "Carol Davila" University of Medicine and Pharmacy, 020021 Bucharest, Romania
- Department of Pediatric Neurology, "Dr. Victor Gomoiu" Children's Hospital, 022102 Bucharest, Romania
| | - Eugenia Roza
- "Carol Davila" University of Medicine and Pharmacy, 020021 Bucharest, Romania
- Department of Pediatric Neurology, "Dr. Victor Gomoiu" Children's Hospital, 022102 Bucharest, Romania
| | - Radu-Stefan Perjoc
- "Carol Davila" University of Medicine and Pharmacy, 020021 Bucharest, Romania
- Department of Pediatric Neurology, "Dr. Victor Gomoiu" Children's Hospital, 022102 Bucharest, Romania
| | - Daniel Mihai Teleanu
- "Carol Davila" University of Medicine and Pharmacy, 020021 Bucharest, Romania
- Department of Neurosurgery, Emergency University Hospital, 050098 Bucharest, Romania
| |
Collapse
|
16
|
Ortega-Hernandez OD, Martínez-Cáceres EM, Presas-Rodríguez S, Ramo-Tello C. Epstein-Barr Virus and Multiple Sclerosis: A Convoluted Interaction and the Opportunity to Unravel Predictive Biomarkers. Int J Mol Sci 2023; 24:ijms24087407. [PMID: 37108566 PMCID: PMC10138841 DOI: 10.3390/ijms24087407] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2023] [Revised: 04/06/2023] [Accepted: 04/13/2023] [Indexed: 04/29/2023] Open
Abstract
Since the early 1980s, Epstein-Barr virus (EBV) infection has been described as one of the main risk factors for developing multiple sclerosis (MS), and recently, new epidemiological evidence has reinforced this premise. EBV seroconversion precedes almost 99% of the new cases of MS and likely predates the first clinical symptoms. The molecular mechanisms of this association are complex and may involve different immunological routes, perhaps all running in parallel (i.e., molecular mimicry, the bystander damage theory, abnormal cytokine networks, and coinfection of EBV with retroviruses, among others). However, despite the large amount of evidence available on these topics, the ultimate role of EBV in the pathogenesis of MS is not fully understood. For instance, it is unclear why after EBV infection some individuals develop MS while others evolve to lymphoproliferative disorders or systemic autoimmune diseases. In this regard, recent studies suggest that the virus may exert epigenetic control over MS susceptibility genes by means of specific virulence factors. Such genetic manipulation has been described in virally-infected memory B cells from patients with MS and are thought to be the main source of autoreactive immune responses. Yet, the role of EBV infection in the natural history of MS and in the initiation of neurodegeneration is even less clear. In this narrative review, we will discuss the available evidence on these topics and the possibility of harnessing such immunological alterations to uncover predictive biomarkers for the onset of MS and perhaps facilitate prognostication of the clinical course.
Collapse
Affiliation(s)
- Oscar-Danilo Ortega-Hernandez
- Multiple Sclerosis Unit, Department of Neurosciences, Hospital Universitari Germans Trias i Pujol-IGTP, 08916 Badalona, Spain
| | - Eva M Martínez-Cáceres
- Department of Immunology, Hospital Universitari Germans Trias i Pujol-IGTP, Universitat Autònoma de Barcelona, 08916 Badalona, Spain
| | - Silvia Presas-Rodríguez
- Multiple Sclerosis Unit, Department of Neurosciences, Hospital Universitari Germans Trias i Pujol-IGTP, 08916 Badalona, Spain
| | - Cristina Ramo-Tello
- Multiple Sclerosis Unit, Department of Neurosciences, Hospital Universitari Germans Trias i Pujol-IGTP, 08916 Badalona, Spain
| |
Collapse
|
17
|
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: 42] [Impact Index Per Article: 21.0] [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
|
18
|
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
|
19
|
Serafini B, Rosicarelli B, Veroni C, Aloisi F. Tissue-resident memory T cells in the multiple sclerosis brain and their relationship to Epstein-Barr virus infected B cells. J Neuroimmunol 2023; 376:578036. [PMID: 36753806 DOI: 10.1016/j.jneuroim.2023.578036] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2022] [Revised: 01/23/2023] [Accepted: 01/23/2023] [Indexed: 01/27/2023]
Abstract
Presence of EBV infected B cells and EBV-specific CD8 T cells in the multiple sclerosis (MS) brain suggests a role for virus-driven immunopathology in brain inflammation. Tissue-resident memory (Trm) T cells differentiating in MS lesions could provide local protection against EBV reactivation. Using immunohistochemical techniques to analyse canonical tissue residency markers in postmortem brains from control and MS cases, we report that CD103 and/or CD69 are mainly expressed in a subset of CD8+ T cells that intermingle with and contact EBV infected B cells in the infiltrated MS white matter and meninges, including B-cell follicles. Some Trm-like cells were found to express granzyme B and PD-1, mainly in white matter lesions. In the MS brain, Trm cells could fail to constrain EBV infection while contributing to sustain inflammation.
Collapse
Affiliation(s)
- Barbara Serafini
- Department of Neuroscience, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161 Rome, Italy.
| | - Barbara Rosicarelli
- Department of Neuroscience, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161 Rome, Italy.
| | - Caterina Veroni
- Department of Neuroscience, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161 Rome, Italy.
| | - Francesca Aloisi
- Department of Neuroscience, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161 Rome, Italy.
| |
Collapse
|
20
|
Bjornevik K, Münz C, Cohen JI, Ascherio A. Epstein-Barr virus as a leading cause of multiple sclerosis: mechanisms and implications. Nat Rev Neurol 2023; 19:160-171. [PMID: 36759741 DOI: 10.1038/s41582-023-00775-5] [Citation(s) in RCA: 66] [Impact Index Per Article: 33.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/11/2023] [Indexed: 02/11/2023]
Abstract
Epidemiological studies have provided compelling evidence that multiple sclerosis (MS) is a rare complication of infection with the Epstein-Barr virus (EBV), a herpesvirus that infects more than 90% of the global population. This link was long suspected because the risk of MS increases markedly after infectious mononucleosis (symptomatic primary EBV infection) and with high titres of antibodies to specific EBV antigens. However, it was not until 2022 that a longitudinal study demonstrated that MS risk is minimal in individuals who are not infected with EBV and that it increases over 30-fold following EBV infection. Over the past few years, a number of studies have provided clues on the underlying mechanisms, which might help us to develop more targeted treatments for MS. In this Review, we discuss the evidence linking EBV to the development of MS and the mechanisms by which the virus is thought to cause the disease. Furthermore, we discuss implications for the treatment and prevention of MS, including the use of antivirals and vaccines.
Collapse
Affiliation(s)
- Kjetil Bjornevik
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA, USA
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Christian Münz
- Viral Immunobiology, Institute of Experimental Immunology, University of Zürich, Zürich, Switzerland
| | - Jeffrey I Cohen
- Laboratory of Infectious Diseases, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, MD, USA
| | - Alberto Ascherio
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA, USA.
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA.
- Channing Laboratory, Department of Medicine, Brigham and Women's Hospital, and Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
21
|
Microglia drive transient insult-induced brain injury by chemotactic recruitment of CD8 + T lymphocytes. Neuron 2023; 111:696-710.e9. [PMID: 36603584 DOI: 10.1016/j.neuron.2022.12.009] [Citation(s) in RCA: 53] [Impact Index Per Article: 26.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2022] [Revised: 09/03/2022] [Accepted: 12/05/2022] [Indexed: 01/06/2023]
Abstract
The crosstalk between the nervous and immune systems has gained increasing attention for its emerging role in neurological diseases. Radiation-induced brain injury (RIBI) remains the most common medical complication of cranial radiotherapy, and its pathological mechanisms have yet to be elucidated. Here, using single-cell RNA and T cell receptor sequencing, we found infiltration and clonal expansion of CD8+ T lymphocytes in the lesioned brain tissues of RIBI patients. Furthermore, by strategies of genetic or pharmacologic interruption, we identified a chemotactic action of microglia-derived CCL2/CCL8 chemokines in mediating the infiltration of CCR2+/CCR5+ CD8+ T cells and tissue damage in RIBI mice. Such a chemotactic axis also participated in the progression of cerebral infarction in the mouse model of ischemic injury. Our findings therefore highlight the critical role of microglia in mediating the dysregulation of adaptive immune responses and reveal a potential therapeutic strategy for non-infectious brain diseases.
Collapse
|
22
|
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
|
23
|
Lanz TV, Robinson WH, Ho PP, Steinman L. Roadmap for understanding mechanisms on how Epstein-Barr virus triggers multiple sclerosis and for translating these discoveries in clinical trials. Clin Transl Immunology 2023; 12:e1438. [PMID: 36815946 PMCID: PMC9933111 DOI: 10.1002/cti2.1438] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2022] [Revised: 02/03/2023] [Accepted: 02/03/2023] [Indexed: 02/18/2023] Open
Abstract
Here, we offer a roadmap for what might be studied next in understanding how EBV triggers MS. We focus on two areas: The first area concerns the molecular mechanisms underlying how clonal antibody in the CSF emanates in widespread molecular mimicry to key antigens in the nervous system including GlialCAM, a protein associated with chloride channels. A second and equally high priority in the roadmap concerns various therapeutic approaches that are related to blocking the mechanisms whereby EBV triggers MS. Therapies deserving of attention include clinical trials with antivirals and the development of 'inverse' vaccines based on nucleic acid technologies to control or to eradicate the consequences of EBV infection. High enthusiasm is given to continuation of ongoing clinical trials of cellular adoptive therapy to attack EBV-infected cells. Clinical trials of vaccines to EBV are another area deserving attention. These suggested topics involving research on mechanism, and the design, implementation and performance of well-designed trials are not intended to be an exhaustive list. We have splendid tools available to our community of medical scientists to tackle how EBV triggers MS and then to perhaps change the world with new therapies to potentially eradicate MS, as we have done with nearly complete success for poliomyelitis.
Collapse
|
24
|
Sato Y. [Epidemiological and mechanistic links between Epstein-Barr virus and multiple sclerosis]. Uirusu 2023; 73:147-152. [PMID: 39343549 DOI: 10.2222/jsv.73.147] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/01/2024]
Abstract
Epstein-Barr virus (EBV) is a ubiquitous human lymphotropic herpesvirus that causes several malignancies. EBV infects approximately 90% of individuals worldwide. Recent studies have provided robust evidence for a causal role of EBV in multiple sclerosis. Multiple sclerosis is the most prevalent chronic inflammatory and degenerative disease of the central nerve system (CNS) that progresses over time to progressive neurodegeneration and disability. Here, I review how a ubiquitous virus can elicit autoreactive antibodies through molecular mimicry between viral and host CNS antigens, triggering multiple sclerosis.
Collapse
Affiliation(s)
- Yoshitaka Sato
- Department of Virology, Nagoya University Graduate School of Medicine
| |
Collapse
|
25
|
Sedighi S, Gholizadeh O, Yasamineh S, Akbarzadeh S, Amini P, Favakehi P, Afkhami H, Firouzi-Amandi A, Pahlevan D, Eslami M, Yousefi B, Poortahmasebi V, Dadashpour M. Comprehensive Investigations Relationship Between Viral Infections and Multiple Sclerosis Pathogenesis. Curr Microbiol 2023; 80:15. [PMID: 36459252 PMCID: PMC9716500 DOI: 10.1007/s00284-022-03112-z] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2022] [Accepted: 11/05/2022] [Indexed: 12/04/2022]
Abstract
Multiple sclerosis (MS) is a chronic autoimmune disease that affects the central nervous system (CNS). Compared to other types of self-limiting myelin disorders, MS compartmentalizes and maintains chronic inflammation in the CNS. Even though the exact cause of MS is unclear, it is assumed that genetic and environmental factors play an important role in susceptibility to this disease. The progression of MS is triggered by certain environmental factors, such as viral infections. The most important viruses that affect MS are Epstein-Barr virus (EBV), human herpes virus 6 (HHV-6), human endogenous retrovirus (HERV), cytomegalovirus (CMV), and varicella zoster virus (VZV). These viruses all have latent stages that allow them to escape immune detection and reactivate after exposure to various stimuli. Furthermore, their tropism for CNS and immune system cells explains their possible deleterious function in neuroinflammation. In this study, the effect of viral infections on MS disease focuses on the details of viruses that can change the risk of the disease. Paying attention to the most recent articles on the role of SARS-CoV-2 in MS disease, laboratory indicators show the interaction of the immune system with the virus. Also, strategies to prevent viruses that play a role in triggering MS are discussed, such as EBV, which is one of the most important.
Collapse
Affiliation(s)
- Somayeh Sedighi
- Department of Immunology, Faculty of Medicine, Medical Science of Mashhad, Mashhad, Iran
| | - Omid Gholizadeh
- Department of Bacteriology and Virology, Faculty of Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
- Research Center for Clinical Virology, Tehran University of Medical Sciences, Tehran, Iran
| | - Saman Yasamineh
- Research Center for Clinical Virology, Tehran University of Medical Sciences, Tehran, Iran
| | - Sama Akbarzadeh
- Department of Animal Biology, Faculty of Natural Science, University of Tabriz, Tabriz, Iran
| | - Parya Amini
- Research Center for Clinical Virology, Tehran University of Medical Sciences, Tehran, Iran
| | - Parnia Favakehi
- Department of Microbiology, Falavargan Branch, Islamic Azad University, Isfahan, Iran
| | - Hamed Afkhami
- Department of Bacteriology, Faculty of Medicine, Medical Science of Shahed, Tehran, Iran
| | - Akram Firouzi-Amandi
- Department of Immunology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Daryoush Pahlevan
- Determinants of Health Research Center, Semnan University of Medical Sciences, Semnan, Iran
| | - Majid Eslami
- Department of Bacteriology and Virology, Semnan University of Medical Sciences, Semnan, Iran
| | - Bahman Yousefi
- Cancer Research Center, Semnan University of Medical Sciences, Semnan, Iran
| | - Vahdat Poortahmasebi
- Department of Bacteriology and Virology, Faculty of Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran.
| | - Mehdi Dadashpour
- Cancer Research Center, Semnan University of Medical Sciences, Semnan, Iran.
- Department of Medical Biotechnology, Faculty of Medicine, Semnan University of Medical Sciences, Semnan, Iran.
| |
Collapse
|
26
|
Alvarez-Sanchez N, Dunn SE. Potential biological contributers to the sex difference in multiple sclerosis progression. Front Immunol 2023; 14:1175874. [PMID: 37122747 PMCID: PMC10140530 DOI: 10.3389/fimmu.2023.1175874] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Accepted: 04/03/2023] [Indexed: 05/02/2023] Open
Abstract
Multiple sclerosis (MS) is an immune-mediated disease that targets the myelin sheath of central nervous system (CNS) neurons leading to axon injury, neuronal death, and neurological progression. Though women are more highly susceptible to developing MS, men that develop this disease exhibit greater cognitive impairment and accumulate disability more rapidly than women. Magnetic resonance imaging and pathology studies have revealed that the greater neurological progression seen in males correlates with chronic immune activation and increased iron accumulation at the rims of chronic white matter lesions as well as more intensive whole brain and grey matter atrophy and axon loss. Studies in humans and in animal models of MS suggest that male aged microglia do not have a higher propensity for inflammation, but may become more re-active at the rim of white matter lesions as a result of the presence of pro-inflammatory T cells, greater astrocyte activation or iron release from oligodendrocytes in the males. There is also evidence that remyelination is more efficient in aged female than aged male rodents and that male neurons are more susceptible to oxidative and nitrosative stress. Both sex chromosome complement and sex hormones contribute to these sex differences in biology.
Collapse
Affiliation(s)
- Nuria Alvarez-Sanchez
- Keenan Research Centre for Biomedical Science, St. Michael’s Hospital, Toronto, ON, Canada
- Department of Immunology, 1 King’s College Circle, Toronto, ON, Canada
| | - Shannon E. Dunn
- Keenan Research Centre for Biomedical Science, St. Michael’s Hospital, Toronto, ON, Canada
- Department of Immunology, 1 King’s College Circle, Toronto, ON, Canada
- Women's College Research Institute, Women's College Hospital, Toronto, ON, Canada
- *Correspondence: Shannon E. Dunn,
| |
Collapse
|
27
|
Abstract
Epstein-Barr virus (EBV) is a ubiquitous human lymphotropic herpesvirus with a well-established causal role in several cancers. Recent studies have provided compelling epidemiological and mechanistic evidence for a causal role of EBV in multiple sclerosis (MS). MS is the most prevalent chronic inflammatory and neurodegenerative disease of the central nervous system and is thought to be triggered in genetically predisposed individuals by an infectious agent, with EBV as the lead candidate. How a ubiquitous virus that typically leads to benign latent infections can promote cancer and autoimmune disease in at-risk populations is not fully understood. Here we review the evidence that EBV is a causal agent for MS and how various risk factors may affect EBV infection and immune control. We focus on EBV contributing to MS through reprogramming of latently infected B lymphocytes and the chronic presentation of viral antigens as a potential source of autoreactivity through molecular mimicry. We consider how knowledge of EBV-associated cancers may be instructive for understanding the role of EBV in MS and discuss the potential for therapies that target EBV to treat MS.
Collapse
Affiliation(s)
- Samantha S. Soldan
- grid.251075.40000 0001 1956 6678The Wistar Institute, Philadelphia, PA USA
| | - Paul M. Lieberman
- grid.251075.40000 0001 1956 6678The Wistar Institute, Philadelphia, PA USA
| |
Collapse
|
28
|
Schneider-Hohendorf T, Gerdes LA, Pignolet B, Gittelman R, Ostkamp P, Rubelt F, Raposo C, Tackenberg B, Riepenhausen M, Janoschka C, Wünsch C, Bucciarelli F, Flierl-Hecht A, Beltrán E, Kümpfel T, Anslinger K, Gross CC, Chapman H, Kaplan I, Brassat D, Wekerle H, Kerschensteiner M, Klotz L, Lünemann JD, Hohlfeld R, Liblau R, Wiendl H, Schwab N. Broader Epstein-Barr virus-specific T cell receptor repertoire in patients with multiple sclerosis. J Exp Med 2022; 219:e20220650. [PMID: 36048016 PMCID: PMC9437111 DOI: 10.1084/jem.20220650] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2022] [Revised: 06/30/2022] [Accepted: 08/08/2022] [Indexed: 12/14/2022] Open
Abstract
Epstein-Barr virus (EBV) infection precedes multiple sclerosis (MS) pathology and cross-reactive antibodies might link EBV infection to CNS autoimmunity. As an altered anti-EBV T cell reaction was suggested in MS, we queried peripheral blood T cell receptor β chain (TCRβ) repertoires of 1,395 MS patients, 887 controls, and 35 monozygotic, MS-discordant twin pairs for multimer-confirmed, viral antigen-specific TCRβ sequences. We detected more MHC-I-restricted EBV-specific TCRβ sequences in MS patients. Differences in genetics or upbringing could be excluded by validation in monozygotic twin pairs discordant for MS. Anti-VLA-4 treatment amplified this observation, while interferon β- or anti-CD20 treatment did not modulate EBV-specific T cell occurrence. In healthy individuals, EBV-specific CD8+ T cells were of an effector-memory phenotype in peripheral blood and cerebrospinal fluid. In MS patients, cerebrospinal fluid also contained EBV-specific central-memory CD8+ T cells, suggesting recent priming. Therefore, MS is not only preceded by EBV infection, but also associated with broader EBV-specific TCR repertoires, consistent with an ongoing anti-EBV immune reaction in MS.
Collapse
Affiliation(s)
- Tilman Schneider-Hohendorf
- Department of Neurology with Institute of Translational Neurology, University of Münster, Münster, Germany
| | - Lisa Ann Gerdes
- Institute of Clinical Neuroimmunology, University Hospital and Biomedical Center, Ludwig-Maximilians Universität München, Munich, Germany
- Biomedical Center, Faculty of Medicine, Ludwig-Maximilians Universität München, Martinsried, Germany
- Munich Cluster of Systems Neurology (SyNergy), Munich, Germany
| | - Béatrice Pignolet
- Toulouse Institute for infectious and inflammatory diseases (Infinity), University of Toulouse, Centre National de la Recherche Scientifique, Institut National de la Santé et de la Recherche Médicale, Université Paul Sabatier, Toulouse, France
| | | | - Patrick Ostkamp
- Department of Neurology with Institute of Translational Neurology, University of Münster, Münster, Germany
| | | | | | - Björn Tackenberg
- F. Hoffmann-La Roche Ltd, Basel, Switzerland
- Philipps-University, Department of Neurology, Marburg, Germany
| | - Marianne Riepenhausen
- Department of Neurology with Institute of Translational Neurology, University of Münster, Münster, Germany
| | - Claudia Janoschka
- Department of Neurology with Institute of Translational Neurology, University of Münster, Münster, Germany
| | - Christian Wünsch
- Department of Neurology with Institute of Translational Neurology, University of Münster, Münster, Germany
| | - Florence Bucciarelli
- Toulouse Institute for infectious and inflammatory diseases (Infinity), University of Toulouse, Centre National de la Recherche Scientifique, Institut National de la Santé et de la Recherche Médicale, Université Paul Sabatier, Toulouse, France
| | - Andrea Flierl-Hecht
- Institute of Clinical Neuroimmunology, University Hospital and Biomedical Center, Ludwig-Maximilians Universität München, Munich, Germany
- Biomedical Center, Faculty of Medicine, Ludwig-Maximilians Universität München, Martinsried, Germany
- Munich Cluster of Systems Neurology (SyNergy), Munich, Germany
| | - Eduardo Beltrán
- Institute of Clinical Neuroimmunology, University Hospital and Biomedical Center, Ludwig-Maximilians Universität München, Munich, Germany
- Biomedical Center, Faculty of Medicine, Ludwig-Maximilians Universität München, Martinsried, Germany
- Munich Cluster of Systems Neurology (SyNergy), Munich, Germany
| | - Tania Kümpfel
- Institute of Clinical Neuroimmunology, University Hospital and Biomedical Center, Ludwig-Maximilians Universität München, Munich, Germany
- Biomedical Center, Faculty of Medicine, Ludwig-Maximilians Universität München, Martinsried, Germany
- Munich Cluster of Systems Neurology (SyNergy), Munich, Germany
| | - Katja Anslinger
- Institute of Legal Medicine, Ludwig-Maximilians Universität München, Munich, Germany
| | - Catharina C. Gross
- Department of Neurology with Institute of Translational Neurology, University of Münster, Münster, Germany
| | | | | | | | - Hartmut Wekerle
- Institute of Clinical Neuroimmunology, University Hospital and Biomedical Center, Ludwig-Maximilians Universität München, Munich, Germany
- Institute for Biological Intelligence, Martinsried, Germany
| | - Martin Kerschensteiner
- Institute of Clinical Neuroimmunology, University Hospital and Biomedical Center, Ludwig-Maximilians Universität München, Munich, Germany
- Biomedical Center, Faculty of Medicine, Ludwig-Maximilians Universität München, Martinsried, Germany
- Munich Cluster of Systems Neurology (SyNergy), Munich, Germany
| | - Luisa Klotz
- Department of Neurology with Institute of Translational Neurology, University of Münster, Münster, Germany
| | - Jan D. Lünemann
- Department of Neurology with Institute of Translational Neurology, University of Münster, Münster, Germany
| | - Reinhard Hohlfeld
- Institute of Clinical Neuroimmunology, University Hospital and Biomedical Center, Ludwig-Maximilians Universität München, Munich, Germany
- Biomedical Center, Faculty of Medicine, Ludwig-Maximilians Universität München, Martinsried, Germany
| | - Roland Liblau
- Toulouse Institute for infectious and inflammatory diseases (Infinity), University of Toulouse, Centre National de la Recherche Scientifique, Institut National de la Santé et de la Recherche Médicale, Université Paul Sabatier, Toulouse, France
| | - Heinz Wiendl
- Department of Neurology with Institute of Translational Neurology, University of Münster, Münster, Germany
| | - Nicholas Schwab
- Department of Neurology with Institute of Translational Neurology, University of Münster, Münster, Germany
| |
Collapse
|
29
|
Giovannoni G, Vanderdonckt P, Hartung HP, Lassmann H, Comi G. EBV and multiple sclerosis: Setting the research agenda. Mult Scler Relat Disord 2022; 67:104158. [PMID: 36116382 DOI: 10.1016/j.msard.2022.104158] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Accepted: 09/04/2022] [Indexed: 01/21/2023]
Affiliation(s)
- Gavin Giovannoni
- Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK.
| | | | - Hans-Peter Hartung
- Hans-Peter Hartung, Klinik für Neurologie, Heinrich-Heine Universität Düsseldorf, Germany
| | - Hans Lassmann
- Hans Lassmann, Center for Brain Research, Medical University of Vienna, A-1090 Wien, Austria
| | - Giancarlo Comi
- Giancarlo Comi, Multiple Sclerosis Centres of Gallarate, and Casa di Cura del Policlinico, Milan, Italy
| |
Collapse
|
30
|
Schönrich G, Abdelaziz MO, Raftery MJ. Epstein-Barr virus, interleukin-10 and multiple sclerosis: A ménage à trois. Front Immunol 2022; 13:1028972. [PMID: 36275700 PMCID: PMC9585213 DOI: 10.3389/fimmu.2022.1028972] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2022] [Accepted: 09/23/2022] [Indexed: 12/30/2022] Open
Abstract
Multiple Sclerosis (MS) is an autoimmune disease that is characterized by inflammation and demyelination of nerve cells. There is strong evidence that Epstein-Barr virus (EBV), a human herpesvirus infecting B cells, greatly increases the risk of subsequent MS. Intriguingly, EBV not only induces human interleukin-10 but also encodes a homologue of this molecule, which is a key anti-inflammatory cytokine of the immune system. Although EBV-encoded IL-10 (ebvIL-10) has a high amino acid identity with its cellular counterpart (cIL-10), it shows more restricted and partially weaker functionality. We propose that both EBV-induced cIL-10 and ebvIL-10 act in a temporally and functionally coordinated manner helping the pathogen to establish latency in B cells and, at the same time, to balance the function of antiviral T cells. As a result, the EBV load persisting in the immune system is kept at a constant but individually different level (set point). During this immunological tug of war between virus and host, however, MS can be induced as collateral damage if the set point is too high. Here, we discuss a possible role of ebvIL-10 and EBV-induced cIL-10 in EBV-driven pathogenesis of MS.
Collapse
Affiliation(s)
- Günther Schönrich
- Institute of Virology, Charité– Universitätsmedizin Berlin, Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin Institute of Health, Berlin, Germany,*Correspondence: Günther Schönrich,
| | - Mohammed O. Abdelaziz
- Institute of Virology, Charité– Universitätsmedizin Berlin, Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin Institute of Health, Berlin, Germany
| | - Martin J. Raftery
- Institute of Virology, Charité– Universitätsmedizin Berlin, Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin Institute of Health, Berlin, Germany,Department of Hematology, Oncology and Tumor Immunology (CCM), Charité– Universitätsmedizin Berlin, Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin Institute of Health, Berlin, Germany
| |
Collapse
|
31
|
Liu R, Du S, Zhao L, Jain S, Sahay K, Rizvanov A, Lezhnyova V, Khaibullin T, Martynova E, Khaiboullina S, Baranwal M. Autoreactive lymphocytes in multiple sclerosis: Pathogenesis and treatment target. Front Immunol 2022; 13:996469. [PMID: 36211343 PMCID: PMC9539795 DOI: 10.3389/fimmu.2022.996469] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2022] [Accepted: 09/08/2022] [Indexed: 11/13/2022] Open
Abstract
Multiple sclerosis (MS) is a chronic inflammatory disease of the central nervous system (CNS) characterized by destruction of the myelin sheath structure. The loss of myelin leads to damage of a neuron’s axon and cell body, which is identified as brain lesions on magnetic resonance image (MRI). The pathogenesis of MS remains largely unknown. However, immune mechanisms, especially those linked to the aberrant lymphocyte activity, are mainly responsible for neuronal damage. Th1 and Th17 populations of lymphocytes were primarily associated with MS pathogenesis. These lymphocytes are essential for differentiation of encephalitogenic CD8+ T cell and Th17 lymphocyte crossing the blood brain barrier and targeting myelin sheath in the CNS. B-lymphocytes could also contribute to MS pathogenesis by producing anti-myelin basic protein antibodies. In later studies, aberrant function of Treg and Th9 cells was identified as contributing to MS. This review summarizes the aberrant function and count of lymphocyte, and the contributions of these cell to the mechanisms of MS. Additionally, we have outlined the novel MS therapeutics aimed to amend the aberrant function or counts of these lymphocytes.
Collapse
Affiliation(s)
- Rongzeng Liu
- Department of Immunology, School of Basic Medical Sciences, Henan University of Science and Technology, Luoyang, China
| | - Shushu Du
- Department of Immunology, School of Basic Medical Sciences, Henan University of Science and Technology, Luoyang, China
| | - Lili Zhao
- Department of Immunology, School of Basic Medical Sciences, Henan University of Science and Technology, Luoyang, China
| | - Sahil Jain
- Department of Biochemistry and Molecular Biology, Faculty of Life Sciences, Tel-Aviv University, Tel-Aviv, Israel
| | - Kritika Sahay
- Department of Biotechnology, Thapar Institute of Engineering and Technology, Patiala, India
| | - Albert Rizvanov
- Gene and cell Department, Kazan Federal University, Kazan, Russia
| | - Vera Lezhnyova
- Gene and cell Department, Kazan Federal University, Kazan, Russia
| | - Timur Khaibullin
- Neurological Department, Republican Clinical Neurological Center, Kazan, Russia
| | | | - Svetlana Khaiboullina
- Gene and cell Department, Kazan Federal University, Kazan, Russia
- *Correspondence: Svetlana Khaiboullina, ; Manoj Baranwal, ;
| | - Manoj Baranwal
- Department of Biotechnology, Thapar Institute of Engineering and Technology, Patiala, India
- *Correspondence: Svetlana Khaiboullina, ; Manoj Baranwal, ;
| |
Collapse
|
32
|
Altered Immune Response to the Epstein-Barr Virus as a Prerequisite for Multiple Sclerosis. Cells 2022; 11:cells11172757. [PMID: 36078165 PMCID: PMC9454695 DOI: 10.3390/cells11172757] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2022] [Revised: 08/30/2022] [Accepted: 08/31/2022] [Indexed: 11/17/2022] Open
Abstract
Strong epidemiologic evidence links Epstein–Barr virus (EBV) infection and its altered immune control to multiple sclerosis (MS) development. Clinical MS onset occurs years after primary EBV infection and the mechanisms linking them remain largely unclear. This review summarizes the epidemiological evidence for this association and how the EBV specific immune control is altered in MS patients. The two main possibilities of mechanisms for this association are further discussed. Firstly, immune responses that are induced during a symptomatic primary EBV infection, namely infectious mononucleosis, might be amplified during the following years to finally cause central nervous system (CNS) inflammation and demyelination. Secondly, genetic predisposition and environmental factors might not allow for an efficient immune control of the EBV-infected B cells that might drive autoimmune T cell stimulation or CNS inflammation. These two main hypotheses for explaining the association of the EBV with MS would implicate opposite therapeutic interventions, namely either dampening CNS inflammatory EBV-reactive immune responses or strengthening them to eliminate the autoimmunity stimulating EBV-infected B cell compartment. Nevertheless, recent findings suggest that EBV is an important puzzle piece in the pathogenesis of MS, and understanding its contribution could open new treatment possibilities for this autoimmune disease.
Collapse
|
33
|
Loosen SH, Doege C, Meuth SG, Luedde T, Kostev K, Roderburg C. Infectious mononucleosis is associated with an increased incidence of multiple sclerosis: Results from a cohort study of 32,116 outpatients in Germany. Front Immunol 2022; 13:937583. [PMID: 35983044 PMCID: PMC9379368 DOI: 10.3389/fimmu.2022.937583] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2022] [Accepted: 07/07/2022] [Indexed: 11/29/2022] Open
Abstract
Background The pathogenesis of multiple sclerosis (MS) has not yet been fully uncovered. There is increasing evidence that Epstein-Barr-Virus (EBV) infection, which affects over 90% of people during life and causes infectious mononucleosis, leads to an increased incidence of MS, and thus may play a crucial role in the pathophysiology of the disease. Methods Using the Disease Analyzer database (IQVIA) featuring diagnoses as well as basic medical and demographic data of outpatients from general practices in Germany, we identified a total of 16,058 patients with infectious mononucleosis that were matched to a cohort of equal size without infectious mononucleosis based on patients’ age, sex, index year and yearly consultation frequency. Incidence of MS was compared within a 10-year follow-up period. Results Within 10 years from the index date, the incidence of MS was 22.6 cases per 100,000 person-years among patient with infectious mononucleosis but only 11.9 cases per 100,000 person-years among individuals without infectious mononucleosis. In regression analysis, infectious mononucleosis was significantly associated with the incidence of MS (HR: 1.86, 95% CI: 1.09-3.16). Subgroup analysis revealed the strongest association between infectious mononucleosis and MS in the age group between 14 and 20 years (HR: 3.52, 95% CI: 1.00-12.37) as well as a stronger association in men compared to women. Conclusion Infectious mononucleosis is associated with an increased incidence of MS especially in younger individuals. Our data support the growing evidence of a decisive involvement of EBV in the currently unknown pathophysiology of MS and should trigger further research efforts to better understand and potentially prevent cases of this disabling disease in future.
Collapse
Affiliation(s)
- Sven H. Loosen
- Clinic for Gastroenterology, Hepatology and Infectious Diseases, University Hospital Düsseldorf, Medical Faculty of Heinrich Heine University Düsseldorf, Düsseldorf, Germany
- *Correspondence: Tom Luedde, ; Sven H. Loosen,
| | - Corinna Doege
- Department of Pediatric Neurology, Center of Pediatrics and Adolescent Medicine, Central Hospital Bremen, Bremen, Germany
| | - Sven G. Meuth
- Department of Neurology, Medical Faculty, University Hospital, Heinrich-Heine-University, Düsseldorf, Germany
| | - Tom Luedde
- Clinic for Gastroenterology, Hepatology and Infectious Diseases, University Hospital Düsseldorf, Medical Faculty of Heinrich Heine University Düsseldorf, Düsseldorf, Germany
- *Correspondence: Tom Luedde, ; Sven H. Loosen,
| | | | - Christoph Roderburg
- Clinic for Gastroenterology, Hepatology and Infectious Diseases, University Hospital Düsseldorf, Medical Faculty of Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| |
Collapse
|
34
|
Abstract
PURPOSE OF REVIEW This article provides an overview of genetic, environmental, and lifestyle risk factors affecting the disease course of multiple sclerosis (MS) and reviews the pathophysiologic characteristics of both relapsing and progressive MS. RECENT FINDINGS The prevalence of MS has increased in recent decades, and costs of care for patients with MS have risen dramatically. Black, Asian, and Hispanic individuals may be at risk for more severe MS-related disability. Multiple genetic MS risk factors have been identified. Factors such as low vitamin D levels and a history of Epstein-Barr virus, smoking, and obesity, especially during childhood, also influence MS risk. Traditionally thought to be a T-cell-mediated disease, recent research has highlighted the additional roles of B cells and microglia in both relapsing and progressive MS. SUMMARY Complex interactions between genetic, environmental, and lifestyle factors affect the risk for MS as well as the disease course. People of color have historically been underrepresented in both MS clinical trials and literature, but current research is attempting to better clarify unique considerations in these groups. MS pathology consists of the focal inflammatory lesions that have been well characterized in relapsing MS, as well as a more widespread neurodegenerative component that is posited to drive progressive disease. Recent advances in characterization of both the inflammatory and neurodegenerative aspects of MS pathophysiology have yielded potential targets for future therapeutic options.
Collapse
|
35
|
Aloisi F, Veroni C, Serafini B. EBV as the 'gluten of MS' hypothesis: Bypassing autoimmunity. Mult Scler Relat Disord 2022; 66:104069. [PMID: 35908445 DOI: 10.1016/j.msard.2022.104069] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2022] [Accepted: 07/21/2022] [Indexed: 11/30/2022]
Abstract
The EBV as the 'gluten of MS' hypothesis discussed by Drosu et al. in a recent Editorial envisages the existence of similar mechanisms leading to celiac disease and multiple sclerosis, such as induction of immunity against an ubiquitous exogenous antigen - gluten and EBV, respectively - and subsequent development of autoimmunity that is maintained by persistence of the initial trigger. While this hypothesis provides the rationale for treating MS with antivirals to lower EBV load, it can be misleading when trying to translate concepts of T cell-B cell interaction and autoimmunity development in celiac disease to multiple sclerosis. Here, we propose that EBV might act as the driver of multiple sclerosis without involving autoimmunity.
Collapse
Affiliation(s)
- Francesca Aloisi
- Department of Neuroscience, Istituto Superiore di Sanità, Viale Regina Elena 299, Rome 00161, Italy.
| | - Caterina Veroni
- Department of Neuroscience, Istituto Superiore di Sanità, Viale Regina Elena 299, Rome 00161, Italy
| | - Barbara Serafini
- Department of Neuroscience, Istituto Superiore di Sanità, Viale Regina Elena 299, Rome 00161, Italy
| |
Collapse
|
36
|
Bhargava P, Hartung HP, Calabresi PA. Contribution of B cells to cortical damage in multiple sclerosis. Brain 2022; 145:3363-3373. [PMID: 35775595 DOI: 10.1093/brain/awac233] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2022] [Revised: 05/06/2022] [Accepted: 06/17/2022] [Indexed: 11/14/2022] Open
Abstract
Multiple sclerosis is associated with lesions not just in the white matter, but also involving the cortex. Cortical involvement has been linked to greater disease severity and hence understanding the factor underlying cortical pathology could help identify new therapeutic strategies for multiple sclerosis. The critical role of B cells in multiple sclerosis has been clarified by multiple pivotal trials of B cell depletion in people with multiple sclerosis. The presence of B cell rich areas of meningeal inflammation in multiple sclerosis has been identified at all stages of multiple sclerosis. Leptomeningeal inflammation is associated with greater extent of cortical demyelination and neuronal loss and with greater disease severity. Recent studies have identified several potential mechanisms by which B cells may mediate cortical injury including antibody production, extracellular vesicles containing neurotoxic substances and production of pro-inflammatory cytokines. Additionally, B cells may indirectly mediate cortical damage through effects on T cells, macrophages or microglia. Several animal models replicate the meningeal inflammation and cortical injury noted in people with multiple sclerosis. Studies in these models have identified BTK inhibition and type II anti-CD20 antibodies as potential agents that can impact meningeal inflammation. Trials of anti-CD20 monoclonal antibodies in people with multiple sclerosis have unsuccessfully attempted to eliminate B cells in the leptomeninges. New strategies to target B cells in multiple sclerosis include BTK inhibition and cell-based therapies aimed at B cells infected with Epstein Barr virus. Future studies will clarify the mechanisms by which B cells mediate cortical injury and treatment strategies that can target B cells in the leptomeninges and CNS parenchyma.
Collapse
Affiliation(s)
- Pavan Bhargava
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Hans Peter Hartung
- Department of Neurology, Heinrich-Heine University, Dusseldorf, Germany.,Brain and Mind Center, University of Sydney, Sydney, Australia.,Department of Neurology, Medical University of Vienna, Vienna, Austria.,Department of Neurology, Palacky University Olomouc, Olomouc, Czech Republic
| | - Peter A Calabresi
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| |
Collapse
|
37
|
Yu H, Bai S, Hao Y, Guan Y. Fatty acids role in multiple sclerosis as "metabokines". J Neuroinflammation 2022; 19:157. [PMID: 35715809 PMCID: PMC9205055 DOI: 10.1186/s12974-022-02502-1] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2021] [Accepted: 06/01/2022] [Indexed: 12/21/2022] Open
Abstract
Multiple sclerosis (MS), as an autoimmune neurological disease with both genetic and environmental contribution, still lacks effective treatment options among progressive patients, highlighting the need to re-evaluate disease innate properties in search for novel therapeutic targets. Fatty acids (FA) and MS bear an interesting intimate connection. FA and FA metabolism are highly associated with autoimmunity, as the diet-derived circulatory and tissue-resident FAs level and composition can modulate immune cells polarization, differentiation and function, suggesting their broad regulatory role as “metabokines”. In addition, FAs are indeed protective factors for blood–brain barrier integrity, crucial contributors of central nervous system (CNS) chronic inflammation and progressive degeneration, as well as important materials for remyelination. The remaining area of ambiguity requires further exploration into this arena to validate the existed phenomenon, develop novel therapies, and confirm the safety and efficacy of therapeutic intervention targeting FA metabolism.
Collapse
Affiliation(s)
- Haojun Yu
- Department of Neurology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, 160 Pujian Road, Pudong, Shanghai, 200127, China
| | - Shuwei Bai
- Department of Neurology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, 160 Pujian Road, Pudong, Shanghai, 200127, China
| | - Yong Hao
- Department of Neurology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, 160 Pujian Road, Pudong, Shanghai, 200127, China.
| | - Yangtai Guan
- Department of Neurology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, 160 Pujian Road, Pudong, Shanghai, 200127, China.
| |
Collapse
|
38
|
Bjornevik K, Cortese M, Healy BC, Kuhle J, Mina MJ, Leng Y, Elledge SJ, Niebuhr DW, Scher AI, Munger KL, Ascherio A. Longitudinal analysis reveals high prevalence of Epstein-Barr virus associated with multiple sclerosis. Science 2022; 375:296-301. [PMID: 35025605 DOI: 10.1126/science.abj8222] [Citation(s) in RCA: 964] [Impact Index Per Article: 321.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
Multiple sclerosis (MS) is a chronic inflammatory demyelinating disease of the central nervous system of unknown etiology. We tested the hypothesis that MS is caused by Epstein-Barr virus (EBV) in a cohort comprising more than 10 million young adults on active duty in the US military, 955 of whom were diagnosed with MS during their period of service. Risk of MS increased 32-fold after infection with EBV but was not increased after infection with other viruses, including the similarly transmitted cytomegalovirus. Serum levels of neurofilament light chain, a biomarker of neuroaxonal degeneration, increased only after EBV seroconversion. These findings cannot be explained by any known risk factor for MS and suggest EBV as the leading cause of MS.
Collapse
Affiliation(s)
- Kjetil Bjornevik
- Department of Nutrition, Harvard T. H. Chan School of Public Health, Boston, MA, USA
| | - Marianna Cortese
- Department of Nutrition, Harvard T. H. Chan School of Public Health, Boston, MA, USA
| | - Brian C Healy
- Partners Multiple Sclerosis Center, Brigham and Women's Hospital, Boston, MA, USA.,Department of Neurology, Harvard Medical School, Boston, MA, USA.,Biostatistics Center, Massachusetts General Hospital, Boston, MA, USA
| | - Jens Kuhle
- Neurologic Clinic and Policlinic, MS Center and Research Center for Clinical Neuroimmunology and Neuroscience Basel (RC2NB), University Hospital Basel, University of Basel, Basel, Switzerland
| | - Michael J Mina
- Division of Genetics, Brigham and Women's Hospital, Howard Hughes Medical Institute, Department of Genetics, and Program in Virology, Harvard Medical School, Boston, MA, USA.,Center for Communicable Disease Dynamics, Department of Epidemiology, and Department of Immunology and Infectious Diseases, Harvard T. H. Chan School of Public Health, Boston, MA, USA.,Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Yumei Leng
- Division of Genetics, Brigham and Women's Hospital, Howard Hughes Medical Institute, Department of Genetics, and Program in Virology, Harvard Medical School, Boston, MA, USA
| | - Stephen J Elledge
- Division of Genetics, Brigham and Women's Hospital, Howard Hughes Medical Institute, Department of Genetics, and Program in Virology, Harvard Medical School, Boston, MA, USA
| | - David W Niebuhr
- Department of Preventive Medicine and Biostatistics, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
| | - Ann I Scher
- Department of Preventive Medicine and Biostatistics, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
| | - Kassandra L Munger
- Department of Nutrition, Harvard T. H. Chan School of Public Health, Boston, MA, USA
| | - Alberto Ascherio
- Department of Nutrition, Harvard T. H. Chan School of Public Health, Boston, MA, USA.,Department of Epidemiology, Harvard T. H. Chan School of Public Health, Boston, MA, USA.,Channing Laboratory, Department of Medicine, Brigham and Women's Hospital, and Harvard Medical School, Boston, MA, USA
| |
Collapse
|
39
|
Abstract
[Figure: see text].
Collapse
Affiliation(s)
- William H Robinson
- Division of Immunology and Rheumatology, Department of Medicine, Stanford University, Stanford, CA, USA.,VA Palo Alto Health Care System, Palo Alto, CA, USA
| | - Lawrence Steinman
- Department of Neurology and Neurological Sciences, Stanford University, Stanford, CA, USA
| |
Collapse
|
40
|
Spirin N, Kiselev D, Baranova N, Karpova M. Nociceptive and mixed pains in patients with multiple sclerosis. Zh Nevrol Psikhiatr Im S S Korsakova 2022; 122:44-51. [DOI: 10.17116/jnevro202212207244] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
|
41
|
Do Epstein–Barr Virus Mutations and Natural Genome Sequence Variations Contribute to Disease? Biomolecules 2021; 12:biom12010017. [PMID: 35053165 PMCID: PMC8774192 DOI: 10.3390/biom12010017] [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: 11/29/2021] [Revised: 12/21/2021] [Accepted: 12/22/2021] [Indexed: 12/15/2022] Open
Abstract
Most of the world’s population is infected by the Epstein–Barr virus (EBV), but the incidence of the diseases associated with EBV infection differs greatly in different parts of the world. Many factors may determine those differences, but variation in the virus genome is likely to be a contributing factor for some of the diseases. Here, we describe the main forms of EBV genome sequence variation, and the mechanisms by which variations in the virus genome are likely to contribute to disease. EBV genome deletions or polymorphisms can also provide useful markers for monitoring disease. If some EBV strains prove to be more pathogenic than others, this suggests the possible value of immunising people against infection by those pathogenic strains.
Collapse
|
42
|
Hassani A, Reguraman N, Shehab S, Khan G. Primary Peripheral Epstein-Barr Virus Infection Can Lead to CNS Infection and Neuroinflammation in a Rabbit Model: Implications for Multiple Sclerosis Pathogenesis. Front Immunol 2021; 12:764937. [PMID: 34899715 PMCID: PMC8656284 DOI: 10.3389/fimmu.2021.764937] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2021] [Accepted: 11/08/2021] [Indexed: 01/04/2023] Open
Abstract
Epstein-Barr virus (EBV) is a common herpesvirus associated with malignant and non-malignant conditions. An accumulating body of evidence supports a role for EBV in the pathogenesis of multiple sclerosis (MS), a demyelinating disease of the CNS. However, little is known about the details of the link between EBV and MS. One obstacle which has hindered research in this area has been the lack of a suitable animal model recapitulating natural infection in humans. We have recently shown that healthy rabbits are susceptible to EBV infection, and viral persistence in these animals mimics latent infection in humans. We used the rabbit model to investigate if peripheral EBV infection can lead to infection of the CNS and its potential consequences. We injected EBV intravenously in one group of animals, and phosphate-buffered saline (PBS) in another, with and without immunosuppression. Histopathological changes and viral dynamics were examined in peripheral blood, spleen, brain, and spinal cord, using a range of molecular and histopathology techniques. Our investigations uncovered important findings that could not be previously addressed. We showed that primary peripheral EBV infection can lead to the virus traversing the CNS. Cell associated, but not free virus in the plasma, correlated with CNS infection. The infected cells within the brain were found to be B-lymphocytes. Most notably, animals injected with EBV, but not PBS, developed inflammatory cellular aggregates in the CNS. The incidence of these aggregates increased in the immunosuppressed animals. The cellular aggregates contained compact clusters of macrophages surrounded by reactive astrocytes and dispersed B and T lymphocytes, but not myelinated nerve fibers. Moreover, studying EBV infection over a span of 28 days, revealed that the peak point for viral load in the periphery and CNS coincides with increased occurrence of cellular aggregates in the brain. Finally, peripheral EBV infection triggered temporal changes in the expression of latent viral transcripts and cytokines in the brain. The present study provides the first direct in vivo evidence for the role of peripheral EBV infection in CNS pathology, and highlights a unique model to dissect viral mechanisms contributing to the development of MS.
Collapse
Affiliation(s)
- Asma Hassani
- Department of Medical Microbiology & Immunology, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain, United Arab Emirates
| | - Narendran Reguraman
- Department of Medical Microbiology & Immunology, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain, United Arab Emirates
| | - Safa Shehab
- Department of Anatomy, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain, United Arab Emirates
| | - Gulfaraz Khan
- Department of Medical Microbiology & Immunology, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain, United Arab Emirates.,Zayed Center for Health Sciences, United Arab Emirates University, Al Ain, United Arab Emirates
| |
Collapse
|
43
|
Meier UC, Cipian RC, Karimi A, Ramasamy R, Middeldorp JM. Cumulative Roles for Epstein-Barr Virus, Human Endogenous Retroviruses, and Human Herpes Virus-6 in Driving an Inflammatory Cascade Underlying MS Pathogenesis. Front Immunol 2021; 12:757302. [PMID: 34790199 PMCID: PMC8592026 DOI: 10.3389/fimmu.2021.757302] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2021] [Accepted: 10/11/2021] [Indexed: 12/12/2022] Open
Abstract
Roles for viral infections and aberrant immune responses in driving localized neuroinflammation and neurodegeneration in multiple sclerosis (MS) are the focus of intense research. Epstein-Barr virus (EBV), as a persistent and frequently reactivating virus with major immunogenic influences and a near 100% epidemiological association with MS, is considered to play a leading role in MS pathogenesis, triggering localized inflammation near or within the central nervous system (CNS). This triggering may occur directly via viral products (RNA and protein) and/or indirectly via antigenic mimicry involving B-cells, T-cells and cytokine-activated astrocytes and microglia cells damaging the myelin sheath of neurons. The genetic MS-risk factor HLA-DR2b (DRB1*1501β, DRA1*0101α) may contribute to aberrant EBV antigen-presentation and anti-EBV reactivity but also to mimicry-induced autoimmune responses characteristic of MS. A central role is proposed for inflammatory EBER1, EBV-miRNA and LMP1 containing exosomes secreted by viable reactivating EBV+ B-cells and repetitive release of EBNA1-DNA complexes from apoptotic EBV+ B-cells, forming reactive immune complexes with EBNA1-IgG and complement. This may be accompanied by cytokine- or EBV-induced expression of human endogenous retrovirus-W/-K (HERV-W/-K) elements and possibly by activation of human herpesvirus-6A (HHV-6A) in early-stage CNS lesions, each contributing to an inflammatory cascade causing the relapsing-remitting neuro-inflammatory and/or progressive features characteristic of MS. Elimination of EBV-carrying B-cells by antibody- and EBV-specific T-cell therapy may hold the promise of reducing EBV activity in the CNS, thereby limiting CNS inflammation, MS symptoms and possibly reversing disease. Other approaches targeting HHV-6 and HERV-W and limiting inflammatory kinase-signaling to treat MS are also being tested with promising results. This article presents an overview of the evidence that EBV, HHV-6, and HERV-W may have a pathogenic role in initiating and promoting MS and possible approaches to mitigate development of the disease.
Collapse
Affiliation(s)
- Ute-Christiane Meier
- Institut für Laboratoriumsmedizin, Klinikum der Universität München, München, Germany.,Department of Psychological Medicine, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, United Kingdom
| | | | - Abbas Karimi
- Department of Molecular Medicine, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | | |
Collapse
|
44
|
Läderach F, Münz C. Epstein Barr Virus Exploits Genetic Susceptibility to Increase Multiple Sclerosis Risk. Microorganisms 2021; 9:2191. [PMID: 34835317 PMCID: PMC8625064 DOI: 10.3390/microorganisms9112191] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Revised: 10/12/2021] [Accepted: 10/13/2021] [Indexed: 02/06/2023] Open
Abstract
Multiple sclerosis (MS) is an autoimmune disease of the central nervous system (CNS) for which both genetic and environmental risk factors have been identified. The strongest synergy among them exists between the MHC class II haplotype and infection with the Epstein Barr virus (EBV), especially symptomatic primary EBV infection (infectious mononucleosis) and elevated EBV-specific antibodies. In this review, we will summarize the epidemiological evidence that EBV infection is a prerequisite for MS development, describe altered EBV specific immune responses in MS patients, and speculate about possible pathogenic mechanisms for the synergy between EBV infection and the MS-associated MHC class II haplotype. We will also discuss how at least one of these mechanisms might explain the recent success of B cell-depleting therapies for MS. While a better mechanistic understanding of the role of EBV infection and its immune control during MS pathogenesis is required and calls for the development of innovative experimental systems to test the proposed mechanisms, therapies targeting EBV-infected B cells are already starting to be explored in MS patients.
Collapse
Affiliation(s)
| | - Christian Münz
- Viral Immunobiology, Institute of Experimental Immunology, University of Zurich, 8057 Zurich, Switzerland; or
| |
Collapse
|
45
|
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
|
46
|
Jakhmola S, Upadhyay A, Jain K, Mishra A, Jha HC. Herpesviruses and the hidden links to Multiple Sclerosis neuropathology. J Neuroimmunol 2021; 358:577636. [PMID: 34174587 DOI: 10.1016/j.jneuroim.2021.577636] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2021] [Revised: 06/01/2021] [Accepted: 06/17/2021] [Indexed: 01/08/2023]
Abstract
Herpesviruses like Epstein-Barr virus, human herpesvirus (HHV)-6, HHV-1, VZV, and human endogenous retroviruses, have an age-old clinical association with multiple sclerosis (MS). MS is an autoimmune disease of the nervous system wherein the myelin sheath deteriorates. The most popular mode of virus mediated immune system manipulation is molecular mimicry. Numerous herpesvirus antigens are similar to myelin proteins. Other mechanisms described here include the activity of cytokines and autoantibodies produced by the autoreactive T and B cells, respectively, viral déjà vu, epitope spreading, CD46 receptor engagement, impaired remyelination etc. Overall, this review addresses the host-parasite association of viruses with MS.
Collapse
Affiliation(s)
- Shweta Jakhmola
- Infection Bioengineering Group, Department of Biosciences and Biomedical Engineering, Indian Institute of Technology Indore, India
| | - Arun Upadhyay
- Cellular and Molecular Neurobiology Unit, Indian Institute of Technology, Jodhpur, India
| | - Khushboo Jain
- Infection Bioengineering Group, Department of Biosciences and Biomedical Engineering, Indian Institute of Technology Indore, India
| | - Amit Mishra
- Cellular and Molecular Neurobiology Unit, Indian Institute of Technology, Jodhpur, India
| | - Hem Chandra Jha
- Infection Bioengineering Group, Department of Biosciences and Biomedical Engineering, Indian Institute of Technology Indore, India.
| |
Collapse
|
47
|
Mrad MF, Saba ES, Nakib L, Khoury SJ. Exosomes From Subjects With Multiple Sclerosis Express EBV-Derived Proteins and Activate Monocyte-Derived Macrophages. NEUROLOGY-NEUROIMMUNOLOGY & NEUROINFLAMMATION 2021; 8:8/4/e1004. [PMID: 34006621 PMCID: PMC8130999 DOI: 10.1212/nxi.0000000000001004] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Accepted: 02/26/2021] [Indexed: 12/19/2022]
Abstract
Objective To investigate in a cross-sectional study the effect of serum-derived exosomes on primary human blood monocyte-derived macrophages (MDMs) comparing exosomes from healthy donors vs patients with relapsing-remitting multiple sclerosis in remission and in relapse and to assess whether the response correlates with exosomal Epstein-Barr virus (EBV) protein expression. Methods A total of 45 serum-derived exosome preparations were isolated from patients and healthy controls and verified for the expression of exosomal and EBV markers. MDMs were differentiated from monocytes for 7 days and incubated for 24 hours with exosomes, and then, cell supernatants were collected for cytokine measurement by cytometric bead array. Cells were immunophenotyped before and after differentiation. Results Serum-derived exosomes of patients with multiple sclerosis (MS) expressed higher levels of EBV proteins than healthy controls. Of interest, expression of EBV nuclear antigen EBNA1 and latent membrane proteins LMP1 and 2A was higher on exosomes derived from patients with active RRMS compared with healthy controls and stable patients. After data normalization, we observed that incubation with EBV(+) exosomes induced CXCL10 and CCL2 secretion by MDMs. MDMs differentiated from patients with active disease were better secretors of CXCL10 and other interferon-γ–inducible chemokines, including CCL2 and CXCL9, than MDMs from healthy and stable MS groups. MDMs from active patients had a higher frequency of a CD14(++) subset that correlated with the secreted CXCL10. Conclusion Exosomes expressing EBV proteins correlate with disease activity and induce an inflammatory response in MDMs that is compounded by the origin of the responder cells.
Collapse
Affiliation(s)
- May F Mrad
- From the Nehme and Therese Tohme Multiple Sclerosis Center (M.F.M.), Faculty of Medicine, American University of Beirut Medical Center; Department of Experimental Pathology (E.S.S., L.N.), Immunology and Microbiology, Faculty of Medicine, American University of Beirut; and Nehme and Therese Tohme Multiple Sclerosis Center (S.J.K.), and Abu Haidar Neuroscience Institute, Faculty of Medicine, American University of Beirut Medical Center, Lebanon
| | - Esber S Saba
- From the Nehme and Therese Tohme Multiple Sclerosis Center (M.F.M.), Faculty of Medicine, American University of Beirut Medical Center; Department of Experimental Pathology (E.S.S., L.N.), Immunology and Microbiology, Faculty of Medicine, American University of Beirut; and Nehme and Therese Tohme Multiple Sclerosis Center (S.J.K.), and Abu Haidar Neuroscience Institute, Faculty of Medicine, American University of Beirut Medical Center, Lebanon
| | - Layane Nakib
- From the Nehme and Therese Tohme Multiple Sclerosis Center (M.F.M.), Faculty of Medicine, American University of Beirut Medical Center; Department of Experimental Pathology (E.S.S., L.N.), Immunology and Microbiology, Faculty of Medicine, American University of Beirut; and Nehme and Therese Tohme Multiple Sclerosis Center (S.J.K.), and Abu Haidar Neuroscience Institute, Faculty of Medicine, American University of Beirut Medical Center, Lebanon
| | - Samia J Khoury
- From the Nehme and Therese Tohme Multiple Sclerosis Center (M.F.M.), Faculty of Medicine, American University of Beirut Medical Center; Department of Experimental Pathology (E.S.S., L.N.), Immunology and Microbiology, Faculty of Medicine, American University of Beirut; and Nehme and Therese Tohme Multiple Sclerosis Center (S.J.K.), and Abu Haidar Neuroscience Institute, Faculty of Medicine, American University of Beirut Medical Center, Lebanon.
| |
Collapse
|
48
|
Ioannides ZA, Csurhes PA, Douglas NL, Mackenroth G, Swayne A, Thompson KM, Hopkins TJ, Green KA, Blum S, Hooper KD, Wyssusek KH, Coulthard A, Pender MP. Sustained Clinical Improvement in a Subset of Patients With Progressive Multiple Sclerosis Treated With Epstein-Barr Virus-Specific T Cell Therapy. Front Neurol 2021; 12:652811. [PMID: 33790852 PMCID: PMC8005645 DOI: 10.3389/fneur.2021.652811] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2021] [Accepted: 02/22/2021] [Indexed: 12/02/2022] Open
Abstract
Background: Increasing evidence indicates a role for Epstein–Barr virus (EBV) in the pathogenesis of multiple sclerosis (MS). EBV-infected autoreactive B cells might accumulate in the central nervous system because of defective cytotoxic CD8+ T cell immunity. We have previously reported results of a phase I clinical trial of autologous EBV-specific T cell therapy in MS 6 months after treatment. Objective: To investigate longer-term outcomes in MS patients who received autologous EBV-specific T cell therapy. Methods: We assessed participants 2 and 3 years after completion of T cell therapy. Results: We collected data from all 10 treated participants at year 2 and from 9 participants at year 3. No serious treatment-related adverse events were observed. Four participants had at least some sustained clinical improvement at year 2, including reduced fatigue in three participants, and reduced Expanded Disability Status Scale score in two participants. Three participants experienced a sustained improvement in at least some symptoms at year 3. More sustained improvement was associated with higher EBV-specific CD8+ T cell reactivity in the administered T cell product. Conclusion: Autologous EBV-specific T cell therapy is well-tolerated, and some degree of clinical improvement can be sustained for up to 3 years after treatment.
Collapse
Affiliation(s)
- Zara A Ioannides
- Faculty of Medicine, The University of Queensland, Brisbane, QLD, Australia.,Department of Neurology, Royal Brisbane and Women's Hospital, Herston, QLD, Australia.,The University of Queensland Centre for Clinical Research, Herston, QLD, Australia
| | - Peter A Csurhes
- Faculty of Medicine, The University of Queensland, Brisbane, QLD, Australia.,The University of Queensland Centre for Clinical Research, Herston, QLD, Australia
| | - Nanette L Douglas
- Faculty of Medicine, The University of Queensland, Brisbane, QLD, Australia.,Department of Neurology, Royal Brisbane and Women's Hospital, Herston, QLD, Australia.,The University of Queensland Centre for Clinical Research, Herston, QLD, Australia
| | - Gem Mackenroth
- Faculty of Medicine, The University of Queensland, Brisbane, QLD, Australia.,Department of Neurology, Royal Brisbane and Women's Hospital, Herston, QLD, Australia.,The University of Queensland Centre for Clinical Research, Herston, QLD, Australia
| | - Andrew Swayne
- Faculty of Medicine, The University of Queensland, Brisbane, QLD, Australia.,Mater Centre for Neurosciences, Mater Hospital, Brisbane, QLD, Australia.,Neurology Department, Princess Alexandra Hospital, Woolloongabba, QLD, Australia
| | - Kate M Thompson
- Department of Psychology, Royal Brisbane and Women's Hospital, Herston, QLD, Australia.,School of Psychology, The University of Queensland, Brisbane, QLD, Australia
| | - Tracey J Hopkins
- Internal Medicine Day Treatment Unit, Royal Brisbane and Women's Hospital, Herston, QLD, Australia
| | - Kerryn A Green
- Faculty of Medicine, The University of Queensland, Brisbane, QLD, Australia.,Department of Neurology, Royal Brisbane and Women's Hospital, Herston, QLD, Australia
| | - Stefan Blum
- Faculty of Medicine, The University of Queensland, Brisbane, QLD, Australia.,Mater Centre for Neurosciences, Mater Hospital, Brisbane, QLD, Australia.,Neurology Department, Princess Alexandra Hospital, Woolloongabba, QLD, Australia
| | - Kaye D Hooper
- Faculty of Medicine, The University of Queensland, Brisbane, QLD, Australia.,Department of Neurology, Royal Brisbane and Women's Hospital, Herston, QLD, Australia.,The University of Queensland Centre for Clinical Research, Herston, QLD, Australia
| | - Kerstin H Wyssusek
- Faculty of Medicine, The University of Queensland, Brisbane, QLD, Australia.,Department of Anesthesia and Perioperative Medicine, Royal Brisbane and Women's Hospital, Herston, QLD, Australia
| | - Alan Coulthard
- Faculty of Medicine, The University of Queensland, Brisbane, QLD, Australia.,Department of Medical Imaging, Royal Brisbane and Women's Hospital, Herston, QLD, Australia
| | - Michael P Pender
- Faculty of Medicine, The University of Queensland, Brisbane, QLD, Australia.,Department of Neurology, Royal Brisbane and Women's Hospital, Herston, QLD, Australia
| |
Collapse
|
49
|
Wang L, Zhang J, Deng ZR, Zu MD, Wang Y. The epidemiology of primary headaches in patients with multiple sclerosis. Brain Behav 2021; 11:e01830. [PMID: 33295123 PMCID: PMC7821604 DOI: 10.1002/brb3.1830] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/10/2020] [Revised: 07/28/2020] [Accepted: 08/17/2020] [Indexed: 12/26/2022] Open
Abstract
OBJECTIVE Recent studies have shown a pathophysiologic link between headache and multiple sclerosis (MS), but the prevalence of primary headaches among patients with MS differs substantially across studies. This meta-analysis aimed to comprehensively gather available evidence to estimate the prevalence of primary headaches among patients with MS. METHOD We systematically searched the electronic databases including PubMed, Embase, and Scopus for cohort, case-control, cross-sectional studies that measured the prevalence of headache among patients with MS. Two reviewers independently screened titles and abstracts to identify the eligible studies and the full texts of the included studies were reviewed. Newcastle-Ottawa Scale (NOS) was used to assess the risk of bias of the included literatures. We then conducted a meta-analysis using Stata Software 15.0 to calculate the pooled prevalence of headaches among patients with MS and assess the source of heterogeneity. RESULTS We identified 16 eligible studies covering a total of 3,560 patients with MS. The pooled estimated prevalence of primary headaches among patients with MS was 56%. The statistical heterogeneity was moderate with I2 of 82.1% (p < .001). Both a visual inspection of the funnel plot and Egger' regression tests revealed no significant publication bias (p = .44). The pooled estimated prevalence of migraine (55%) was higher in comparison with that of tension-type headache (20%). The prevalence of migraine subtype was 16% and 10% for migraine without aura and migraine with aura, respectively. The pooled prevalence of primary headache in case-control group (57%) was approximately in line with the cross-sectional group (56%). CONCLUSION The overall prevalence of primary headaches among patients with MS was considerably high. Clinical screening of headache among patients with MS will be helpful to formulate an individualized treatment plans and alleviate the physical and mental impact of the disease.
Collapse
Affiliation(s)
- Long Wang
- Department of Neurology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Juan Zhang
- Department of Neurology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Zi-Ru Deng
- Department of Neurology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Mei-Dan Zu
- Department of Neurology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Yu Wang
- Department of Neurology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| |
Collapse
|
50
|
Ruprecht K. The role of Epstein-Barr virus in the etiology of multiple sclerosis: a current review. Expert Rev Clin Immunol 2020; 16:1143-1157. [PMID: 33152255 DOI: 10.1080/1744666x.2021.1847642] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Introduction: Multiple sclerosis (MS) is a chronic inflammatory demyelinating disease of the central nervous system. While its exact etiology is unknown, it is generally believed that MS is caused by environmental triggers in genetically predisposed individuals. Strong and consistent evidence suggests a key role of Epstein-Barr virus (EBV), a B lymphotropic human gammaherpesvirus, in the etiology of MS. Areas covered: This review summarizes recent developments in the field of EBV and MS with a focus on potential mechanisms underlying the role of EBV in MS. PubMed was searched for the terms 'Epstein-Barr virus' and 'multiple sclerosis'. Expert opinion: The current evidence is compatible with the working hypothesis that MS is a rare complication of EBV infection. Under the premise of a causative role of EBV in MS, it needs to be postulated that EBV causes a specific, and likely persistent, change(s) that is necessarily required for the development of MS. However, although progress has been made, the nature of that change and thus the precise mechanism explaining the role of EBV in MS remain elusive. The mechanism of EBV in MS therefore is a pressing question, whose clarification may substantially advance the pathophysiological understanding, rational therapies, and prevention of MS.
Collapse
Affiliation(s)
- Klemens Ruprecht
- Department of Neurology, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health , Berlin, Germany
| |
Collapse
|