1
|
Sarkkinen J, Yohannes DA, Kreivi N, Dürnsteiner P, Elsakova A, Huuhtanen J, Nowlan K, Kurdo G, Linden R, Saarela M, Tienari PJ, Kekäläinen E, Perdomo M, Laakso SM. Altered immune landscape of cervical lymph nodes reveals Epstein-Barr virus signature in multiple sclerosis. Sci Immunol 2025; 10:eadl3604. [PMID: 39982975 DOI: 10.1126/sciimmunol.adl3604] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2023] [Revised: 07/17/2024] [Accepted: 01/29/2025] [Indexed: 02/23/2025]
Abstract
Multiple sclerosis (MS) is an autoimmune disease of the central nervous system, and Epstein-Barr virus (EBV) infection is a prerequisite for developing the disease. However, the pathogenic mechanisms that lead to MS remain to be determined. Here, we characterized the immune landscape of deep cervical lymph nodes (dcLNs) in newly diagnosed untreated patients with MS (pwMS) using fine-needle aspirations. By combining single-cell RNA sequencing and cellular indexing of transcriptomes and epitopes by sequencing, we observed increased memory B cells and reduced germinal center B cells with decreased clonality in pwMS. Double-negative memory B cells were increased in pwMS that transcriptionally resembled B cells with a lytic EBV infection. Moreover, EBV-targeting memory CD8 T cells were detected in a subset of pwMS. We also detected increased EBV DNA in dcLNs and elevated viral loads in patient saliva. These findings suggest that EBV-driven B cell dysregulation is a critical mechanism in MS pathogenesis.
Collapse
Affiliation(s)
- Joona Sarkkinen
- Translational Immunology Research Program, University of Helsinki, Helsinki, Finland
| | - Dawit A Yohannes
- Translational Immunology Research Program, University of Helsinki, Helsinki, Finland
| | - Nea Kreivi
- Translational Immunology Research Program, University of Helsinki, Helsinki, Finland
| | - Pia Dürnsteiner
- Translational Immunology Research Program, University of Helsinki, Helsinki, Finland
| | - Alexandra Elsakova
- Translational Immunology Research Program, University of Helsinki, Helsinki, Finland
| | - Jani Huuhtanen
- Translational Immunology Research Program, University of Helsinki, Helsinki, Finland
- Hematology Research Unit Helsinki, Department of Hematology, University of Helsinki and Helsinki University Hospital Comprehensive Cancer Center, Helsinki, Finland
- ICAN Digital Precision Cancer Medicine Flagship, University of Helsinki and Helsinki University Hospital Comprehensive Cancer Center, Helsinki, Finland
- Department of Computer Science, Aalto University School of Science, Espoo, Finland
| | - Kirsten Nowlan
- Translational Immunology Research Program, University of Helsinki, Helsinki, Finland
| | - Goran Kurdo
- Department of Radiology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Riikka Linden
- Department of Radiology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Mika Saarela
- Department of Neurology, Brain Center, Helsinki University Hospital, Helsinki, Finland
| | - Pentti J Tienari
- Translational Immunology Research Program, University of Helsinki, Helsinki, Finland
- Department of Neurology, Brain Center, Helsinki University Hospital, Helsinki, Finland
| | - Eliisa Kekäläinen
- Translational Immunology Research Program, University of Helsinki, Helsinki, Finland
| | - Maria Perdomo
- Department of Virology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Sini M Laakso
- Translational Immunology Research Program, University of Helsinki, Helsinki, Finland
- Department of Neurology, Brain Center, Helsinki University Hospital, Helsinki, Finland
| |
Collapse
|
2
|
Mittl K, Hayashi F, Dandekar R, Schubert RD, Gerdts J, Oshiro L, Loudermilk R, Greenfield A, Augusto DG, Ramesh A, Tran E, Koshal K, Kizer K, Dreux J, Cagalingan A, Schustek F, Flood L, Moore T, Kirkemo LL, Cooper T, Harms M, Gomez R, Sibener L, Cree BAC, Hauser SL, Hollenbach JA, Gee M, Wilson MR, Zamvil SS, Sabatino JJ. Antigen specificity of clonally-enriched CD8+ T cells in multiple sclerosis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.07.611010. [PMID: 39282370 PMCID: PMC11398516 DOI: 10.1101/2024.09.07.611010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 09/19/2024]
Abstract
CD8+ T cells are the dominant lymphocyte population in multiple sclerosis (MS) lesions where they are highly clonally expanded. The clonal identity, function, and antigen specificity of CD8+ T cells in MS are not well understood. Here we report a comprehensive single-cell RNA-seq and T cell receptor (TCR)-seq analysis of the cerebrospinal fluid (CSF) and blood from a cohort of treatment-naïve MS patients and control participants. A small subset of highly expanded and activated CD8+ T cells were enriched in the CSF in MS that displayed high activation, cytotoxicity and tissue-homing transcriptional profiles. Using a combination of unbiased and targeted antigen discovery approaches, MS-derived CD8+ T cell clonotypes recognizing Epstein-Barr virus (EBV) antigens and multiple novel mimotopes were identified. These findings shed vital insight into the role of CD8+ T cells in MS and pave the way towards disease biomarkers and therapeutic targets.
Collapse
|
3
|
Bogers L, Rip J, Rijvers L, van Langelaar J, Koetzier SC, Kuiper KL, Meerdink V, Wierenga-Wolf AF, Melief MJ, Marques AM, Smolders J, van Luijn MM. Impact of coding risk variant IFNGR2 on the B cell-intrinsic IFN-γ signaling pathway in multiple sclerosis. J Autoimmun 2024; 148:103279. [PMID: 38972102 DOI: 10.1016/j.jaut.2024.103279] [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: 02/13/2024] [Revised: 06/17/2024] [Accepted: 06/27/2024] [Indexed: 07/09/2024]
Abstract
B cells of people with multiple sclerosis (MS) are more responsive to IFN-γ, corresponding to their brain-homing potential. We studied how a coding single nucleotide polymorphism (SNP) in IFNGR2 (rs9808753) co-operates with Epstein-Barr virus (EBV) infection as MS risk factors to affect the IFN-γ signaling pathway in human B cells. In both cell lines and primary cells, EBV infection positively associated with IFN-γ receptor expression and STAT1 phosphorylation. The IFNGR2 risk SNP selectively promoted downstream signaling via STAT1, particularly in transitional B cells. Altogether, EBV and the IFNGR2 risk SNP independently amplify IFN-γ signaling, potentially driving B cells to enter the MS brain.
Collapse
Affiliation(s)
- Laurens Bogers
- Department of Immunology, MS Center ErasMS, Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands
| | - Jasper Rip
- Department of Immunology, MS Center ErasMS, Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands
| | - Liza Rijvers
- Department of Immunology, MS Center ErasMS, Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands
| | - Jamie van Langelaar
- Department of Immunology, MS Center ErasMS, Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands
| | - Steven C Koetzier
- Department of Immunology, MS Center ErasMS, Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands
| | - Kirsten L Kuiper
- Department of Immunology, MS Center ErasMS, Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands
| | - Veronique Meerdink
- Department of Immunology, MS Center ErasMS, Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands
| | - Annet F Wierenga-Wolf
- Department of Immunology, MS Center ErasMS, Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands
| | - Marie-José Melief
- Department of Immunology, MS Center ErasMS, Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands
| | - Ana M Marques
- Department of Immunology, MS Center ErasMS, Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands
| | - Joost Smolders
- Department of Immunology, MS Center ErasMS, Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands; Department of Neurology, MS Center ErasMS, Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands; Neuroimmunology Research Group, Netherlands Institute for Neuroscience, Amsterdam, the Netherlands
| | - Marvin M van Luijn
- Department of Immunology, MS Center ErasMS, Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands.
| |
Collapse
|
4
|
Wang C, Zhang H, Lu W, Zhan Y. The EBV connection: a severe case of GFAP-A with central hypoventilation unresponsive to IVIG and literature review. Eur J Med Res 2024; 29:415. [PMID: 39135139 PMCID: PMC11320868 DOI: 10.1186/s40001-024-01926-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2024] [Accepted: 06/06/2024] [Indexed: 08/15/2024] Open
Abstract
PURPOSE Glial fibrillary acidic protein astrocytopathy (GFAP-A) pathogenesis remains uncertain, with potential viral involvement. More clinical cases are needed to deepen our understanding of this disease, along with the exploration of more effective treatment options to provide clinicians with additional choices. METHODS We report a severe case of GFAP-A secondary to EBV infection, characterized predominantly by central respiratory failure. Additionally, we conducted a literature review summarizing the characteristics of GFAP-IgG-positive patients associated with EBV infection. RESULTS Among the 13 patients identified, fever (92.3%) and headache (84.6%) were the most common initial symptoms, while urinary dysfunction was universally present in all patients. Over half of the patients with altered consciousness required endotracheal intubation (7/11, 63.6%), with only one individual experiencing complete resolution without any residual sequela. Only two patients (16.7%) displayed the classic feature of periventricular enhancement on neuroimaging, whereas T2-FLAIR hyperintensities were more prevalent. All patients tested positive for GFAP-IgG in CSF, and 91.7% (11/12) had detectable serum GFAP-IgG antibodies. Three patients (23.1%) achieved full recovery solely through antiviral therapy. In patients receiving various immunotherapies, 60% (6/10) still had residual sequelae. CONCLUSION EBV infection may contribute to the pathogenesis of GFAP-A. GFAP antibody testing is recommended for diagnostic evaluation in cases of central nervous system viral infections presenting with respiratory insufficiency. For severe GFAP-A patients, Protein A immunoadsorption (Protein A IA).
Collapse
Affiliation(s)
- Changlun Wang
- Department of Neurology, Second Xiangya Hospital, Central South University, No. 139, Renmin Middle Road, Changsha, China
- Clinical Medical Research Center for Stroke Prevention and Treatment of Hunan Province, Department of Neurology, Second Xiangya Hospital, Central South University, Changsha, China
| | - Hainan Zhang
- Department of Neurology, Second Xiangya Hospital, Central South University, No. 139, Renmin Middle Road, Changsha, China
- Clinical Medical Research Center for Stroke Prevention and Treatment of Hunan Province, Department of Neurology, Second Xiangya Hospital, Central South University, Changsha, China
| | - Wei Lu
- Department of Neurology, Second Xiangya Hospital, Central South University, No. 139, Renmin Middle Road, Changsha, China
- Clinical Medical Research Center for Stroke Prevention and Treatment of Hunan Province, Department of Neurology, Second Xiangya Hospital, Central South University, Changsha, China
| | - Yajing Zhan
- Department of Neurology, Second Xiangya Hospital, Central South University, No. 139, Renmin Middle Road, Changsha, China.
- Clinical Medical Research Center for Stroke Prevention and Treatment of Hunan Province, Department of Neurology, Second Xiangya Hospital, Central South University, Changsha, China.
| |
Collapse
|
5
|
Abbadessa G, Lepore MT, Bruzzaniti S, Piemonte E, Miele G, Signoriello E, Perna F, De Falco C, Lus G, Matarese G, Bonavita S, Galgani M. Ocrelizumab Alters Cytotoxic Lymphocyte Function While Reducing EBV-Specific CD8 + T-Cell Proliferation in Patients With Multiple Sclerosis. NEUROLOGY(R) NEUROIMMUNOLOGY & NEUROINFLAMMATION 2024; 11:e200250. [PMID: 38662990 PMCID: PMC11087045 DOI: 10.1212/nxi.0000000000200250] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/22/2023] [Accepted: 03/08/2024] [Indexed: 05/12/2024]
Abstract
BACKGROUND AND OBJECTIVES The role of B cells in the pathogenic events leading to relapsing multiple sclerosis (R-MS) has only been recently elucidated. A pivotal step in defining this role has been provided by therapeutic efficacy of anti-CD20 monoclonal antibodies. Indeed, treatment with anti-CD20 can also alter number and function of other immune cells not directly expressing CD20 on their cell surface, whose activities can contribute to unknown aspects influencing therapeutic efficacy. We examined the phenotype and function of cytotoxic lymphocytes and Epstein-Barr virus (EBV)-specific immune responses in people with R-MS before and after ocrelizumab treatment. METHODS In this prospective study, we collected blood samples from people with R-MS (n = 41) before and 6 and 12 months after initiating ocrelizumab to assess the immune phenotype and the indirect impact on cytotoxic functions of CD8+ T and NK cells. In addition, we evaluated the specific anti-EBV proliferative responses of both CD8+ T and NK lymphocytes as surrogate markers of anti-EBV activity. RESULTS We observed that while ocrelizumab depleted circulating B cells, it also reduced the expression of activation and migratory markers on both CD8+ T and NK cells as well as their in vitro cytotoxic activity. A comparable pattern in the modulation of immune molecules by ocrelizumab was observed in cytotoxic cells even when patients with R-MS were divided into groups based on their prior disease-modifying treatment. These effects were accompanied by a significant and selective reduction of CD8+ T-cell proliferation in response to EBV antigenic peptides. DISCUSSION Taken together, our findings suggest that ocrelizumab-while depleting B cells-affects the cytotoxic function of CD8+ and NK cells, whose reduced cross-activity against myelin antigens might also contribute to its therapeutic efficacy during MS.
Collapse
MESH Headings
- Humans
- Antibodies, Monoclonal, Humanized/pharmacology
- Female
- Adult
- Male
- Herpesvirus 4, Human/immunology
- CD8-Positive T-Lymphocytes/drug effects
- CD8-Positive T-Lymphocytes/immunology
- Middle Aged
- Immunologic Factors/pharmacology
- Multiple Sclerosis, Relapsing-Remitting/drug therapy
- Multiple Sclerosis, Relapsing-Remitting/immunology
- Multiple Sclerosis, Relapsing-Remitting/blood
- T-Lymphocytes, Cytotoxic/drug effects
- T-Lymphocytes, Cytotoxic/immunology
- Prospective Studies
- Cell Proliferation/drug effects
- Killer Cells, Natural/drug effects
- Killer Cells, Natural/immunology
- Lymphocyte Activation/drug effects
Collapse
Affiliation(s)
- Gianmarco Abbadessa
- From the Department of Advanced Medical and Surgical Sciences (G.A., G. Miele, E.S., G.L., S. Bonavita), University of Campania Luigi Vanvitelli; Istituto per l'Endocrinologia e l'Oncologia Sperimentale "G.Salvatore" - Consiglio Nazionale delle Ricerche (M.T.L., S. Bruzzaniti, G. Matarese, M.G.); Department of Molecular Medicine and Medical Biotechnologies (E.P., G. Matarese, M.G.); Department of Clinical Medicine and Surgery (F.P.), University of Naples "Federico II"; and UOC Biochimica Clinica - Ospedali dei Colli (C.D.F.), Naples, Italy
| | - Maria Teresa Lepore
- From the Department of Advanced Medical and Surgical Sciences (G.A., G. Miele, E.S., G.L., S. Bonavita), University of Campania Luigi Vanvitelli; Istituto per l'Endocrinologia e l'Oncologia Sperimentale "G.Salvatore" - Consiglio Nazionale delle Ricerche (M.T.L., S. Bruzzaniti, G. Matarese, M.G.); Department of Molecular Medicine and Medical Biotechnologies (E.P., G. Matarese, M.G.); Department of Clinical Medicine and Surgery (F.P.), University of Naples "Federico II"; and UOC Biochimica Clinica - Ospedali dei Colli (C.D.F.), Naples, Italy
| | - Sara Bruzzaniti
- From the Department of Advanced Medical and Surgical Sciences (G.A., G. Miele, E.S., G.L., S. Bonavita), University of Campania Luigi Vanvitelli; Istituto per l'Endocrinologia e l'Oncologia Sperimentale "G.Salvatore" - Consiglio Nazionale delle Ricerche (M.T.L., S. Bruzzaniti, G. Matarese, M.G.); Department of Molecular Medicine and Medical Biotechnologies (E.P., G. Matarese, M.G.); Department of Clinical Medicine and Surgery (F.P.), University of Naples "Federico II"; and UOC Biochimica Clinica - Ospedali dei Colli (C.D.F.), Naples, Italy
| | - Erica Piemonte
- From the Department of Advanced Medical and Surgical Sciences (G.A., G. Miele, E.S., G.L., S. Bonavita), University of Campania Luigi Vanvitelli; Istituto per l'Endocrinologia e l'Oncologia Sperimentale "G.Salvatore" - Consiglio Nazionale delle Ricerche (M.T.L., S. Bruzzaniti, G. Matarese, M.G.); Department of Molecular Medicine and Medical Biotechnologies (E.P., G. Matarese, M.G.); Department of Clinical Medicine and Surgery (F.P.), University of Naples "Federico II"; and UOC Biochimica Clinica - Ospedali dei Colli (C.D.F.), Naples, Italy
| | - Giuseppina Miele
- From the Department of Advanced Medical and Surgical Sciences (G.A., G. Miele, E.S., G.L., S. Bonavita), University of Campania Luigi Vanvitelli; Istituto per l'Endocrinologia e l'Oncologia Sperimentale "G.Salvatore" - Consiglio Nazionale delle Ricerche (M.T.L., S. Bruzzaniti, G. Matarese, M.G.); Department of Molecular Medicine and Medical Biotechnologies (E.P., G. Matarese, M.G.); Department of Clinical Medicine and Surgery (F.P.), University of Naples "Federico II"; and UOC Biochimica Clinica - Ospedali dei Colli (C.D.F.), Naples, Italy
| | - Elisabetta Signoriello
- From the Department of Advanced Medical and Surgical Sciences (G.A., G. Miele, E.S., G.L., S. Bonavita), University of Campania Luigi Vanvitelli; Istituto per l'Endocrinologia e l'Oncologia Sperimentale "G.Salvatore" - Consiglio Nazionale delle Ricerche (M.T.L., S. Bruzzaniti, G. Matarese, M.G.); Department of Molecular Medicine and Medical Biotechnologies (E.P., G. Matarese, M.G.); Department of Clinical Medicine and Surgery (F.P.), University of Naples "Federico II"; and UOC Biochimica Clinica - Ospedali dei Colli (C.D.F.), Naples, Italy
| | - Francesco Perna
- From the Department of Advanced Medical and Surgical Sciences (G.A., G. Miele, E.S., G.L., S. Bonavita), University of Campania Luigi Vanvitelli; Istituto per l'Endocrinologia e l'Oncologia Sperimentale "G.Salvatore" - Consiglio Nazionale delle Ricerche (M.T.L., S. Bruzzaniti, G. Matarese, M.G.); Department of Molecular Medicine and Medical Biotechnologies (E.P., G. Matarese, M.G.); Department of Clinical Medicine and Surgery (F.P.), University of Naples "Federico II"; and UOC Biochimica Clinica - Ospedali dei Colli (C.D.F.), Naples, Italy
| | - Chiara De Falco
- From the Department of Advanced Medical and Surgical Sciences (G.A., G. Miele, E.S., G.L., S. Bonavita), University of Campania Luigi Vanvitelli; Istituto per l'Endocrinologia e l'Oncologia Sperimentale "G.Salvatore" - Consiglio Nazionale delle Ricerche (M.T.L., S. Bruzzaniti, G. Matarese, M.G.); Department of Molecular Medicine and Medical Biotechnologies (E.P., G. Matarese, M.G.); Department of Clinical Medicine and Surgery (F.P.), University of Naples "Federico II"; and UOC Biochimica Clinica - Ospedali dei Colli (C.D.F.), Naples, Italy
| | - G Lus
- From the Department of Advanced Medical and Surgical Sciences (G.A., G. Miele, E.S., G.L., S. Bonavita), University of Campania Luigi Vanvitelli; Istituto per l'Endocrinologia e l'Oncologia Sperimentale "G.Salvatore" - Consiglio Nazionale delle Ricerche (M.T.L., S. Bruzzaniti, G. Matarese, M.G.); Department of Molecular Medicine and Medical Biotechnologies (E.P., G. Matarese, M.G.); Department of Clinical Medicine and Surgery (F.P.), University of Naples "Federico II"; and UOC Biochimica Clinica - Ospedali dei Colli (C.D.F.), Naples, Italy
| | - Giuseppe Matarese
- From the Department of Advanced Medical and Surgical Sciences (G.A., G. Miele, E.S., G.L., S. Bonavita), University of Campania Luigi Vanvitelli; Istituto per l'Endocrinologia e l'Oncologia Sperimentale "G.Salvatore" - Consiglio Nazionale delle Ricerche (M.T.L., S. Bruzzaniti, G. Matarese, M.G.); Department of Molecular Medicine and Medical Biotechnologies (E.P., G. Matarese, M.G.); Department of Clinical Medicine and Surgery (F.P.), University of Naples "Federico II"; and UOC Biochimica Clinica - Ospedali dei Colli (C.D.F.), Naples, Italy
| | - Simona Bonavita
- From the Department of Advanced Medical and Surgical Sciences (G.A., G. Miele, E.S., G.L., S. Bonavita), University of Campania Luigi Vanvitelli; Istituto per l'Endocrinologia e l'Oncologia Sperimentale "G.Salvatore" - Consiglio Nazionale delle Ricerche (M.T.L., S. Bruzzaniti, G. Matarese, M.G.); Department of Molecular Medicine and Medical Biotechnologies (E.P., G. Matarese, M.G.); Department of Clinical Medicine and Surgery (F.P.), University of Naples "Federico II"; and UOC Biochimica Clinica - Ospedali dei Colli (C.D.F.), Naples, Italy
| | - Mario Galgani
- From the Department of Advanced Medical and Surgical Sciences (G.A., G. Miele, E.S., G.L., S. Bonavita), University of Campania Luigi Vanvitelli; Istituto per l'Endocrinologia e l'Oncologia Sperimentale "G.Salvatore" - Consiglio Nazionale delle Ricerche (M.T.L., S. Bruzzaniti, G. Matarese, M.G.); Department of Molecular Medicine and Medical Biotechnologies (E.P., G. Matarese, M.G.); Department of Clinical Medicine and Surgery (F.P.), University of Naples "Federico II"; and UOC Biochimica Clinica - Ospedali dei Colli (C.D.F.), Naples, Italy
| |
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
|
Dungan L, Dunne J, Savio M, Kalaszi M, McElheron M, Lynagh Y, O'Driscoll K, Roche C, Qureshi A, Crowley B, Conlon N, Kearney H. Disease-Modifying Treatments for Multiple Sclerosis Affect Measures of Cellular Immune Responses to EBNA-1 Peptides. NEUROLOGY(R) NEUROIMMUNOLOGY & NEUROINFLAMMATION 2024; 11:e200217. [PMID: 38547427 DOI: 10.1212/nxi.0000000000200217] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/27/2023] [Accepted: 01/19/2024] [Indexed: 04/02/2024]
Abstract
BACKGROUND AND OBJECTIVES Epstein-Barr virus (EBV) has been strongly implicated in the pathogenesis of multiple sclerosis (MS). Despite this, there are no routinely used tests to measure cellular response to EBV. In this study, we analyzed the cellular response to EBV nuclear antigen-1 (EBNA-1) in people with MS (pwMS) using a whole blood assay. METHODS This cross-sectional study took place in a dedicated MS clinic in a university hospital. We recruited healthy controls, people with epilepsy (PWE), and pwMS taking a range of disease-modifying treatments (DMTs) including natalizumab, anti-CD20 monoclonal antibodies (mAbs), dimethyl fumarate (DMF), and also treatment naïve. Whole blood samples were stimulated with commercially available PepTivator EBNA1 peptides and a control virus-cytomegalovirus (CMV) peptide. We recorded the cellular response to stimulation with both interferon gamma (IFN-γ) and interleukin-2 (IL-2). We also compared the cellular responses to EBNA1 with IgG responses to EBNA1, viral capsid antigen (VCA), and EBV viral load. RESULTS We recruited 86 pwMS, with relapsing remitting MS, in this group, and we observed a higher level of cellular response recorded with IFN-γ (0.79 IU/mL ± 1.36) vs healthy controls (0.29 IU/mL ± 0.90, p = 0.0048) and PWE (0.17 IU/mL ± 0.33, p = 0.0088). Treatment with either anti-CD20 mAbs (0.28 IU/mL ± 0.57) or DMF (0.07 IU/mL ± 0.15) resulted in a cellular response equivalent to control levels or in PWE (p = 0.26). The results of recording IL-2 response were concordant with IFN-γ: with suppression also seen with anti-CD20 mAbs and DMF. By contrast, we did not record any differential effect of DMTs on the levels of IgG to either EBNA-1 or VCA. Nor did we observe differences in cellular response to cytomegalovirus between groups. DISCUSSION This study demonstrates how testing and recording the cellular response to EBNA-1 in pwMS may be beneficial. EBNA-1 stimulation of whole blood samples produced higher levels of IFN-γ and IL-2 in pwMS compared with controls and PWE. In addition, we show a differential effect of currently available DMTs on this response. The functional assay deployed uses whole blood samples with minimal preprocessing suggesting that employment as a treatment response measure in clinical trials targeting EBV may be possible.
Collapse
Affiliation(s)
- Lara Dungan
- From the Department of Immunology (L.D., J.D., N.C.), St James's Hospital; School of Medicine (M.S., N.C.), Trinity College Dublin; MS Unit (M.K., H.K.), Department of Neurology, St James's Hospital; Department of Medical Gerontology (M.M.), School of Medicine, Trinity Translational Medicine Institute, Trinity College Dublin; Virology Laboratory (Y.L., K.O.D., C.R., A.Q., B.C.), St James's Hospital; and FutureNeuro SFI Research Centre (H.K.), Academic Unit of Neurology, School of Medicine, Trinity College Dublin, Ireland
| | - Jean Dunne
- From the Department of Immunology (L.D., J.D., N.C.), St James's Hospital; School of Medicine (M.S., N.C.), Trinity College Dublin; MS Unit (M.K., H.K.), Department of Neurology, St James's Hospital; Department of Medical Gerontology (M.M.), School of Medicine, Trinity Translational Medicine Institute, Trinity College Dublin; Virology Laboratory (Y.L., K.O.D., C.R., A.Q., B.C.), St James's Hospital; and FutureNeuro SFI Research Centre (H.K.), Academic Unit of Neurology, School of Medicine, Trinity College Dublin, Ireland
| | - Michael Savio
- From the Department of Immunology (L.D., J.D., N.C.), St James's Hospital; School of Medicine (M.S., N.C.), Trinity College Dublin; MS Unit (M.K., H.K.), Department of Neurology, St James's Hospital; Department of Medical Gerontology (M.M.), School of Medicine, Trinity Translational Medicine Institute, Trinity College Dublin; Virology Laboratory (Y.L., K.O.D., C.R., A.Q., B.C.), St James's Hospital; and FutureNeuro SFI Research Centre (H.K.), Academic Unit of Neurology, School of Medicine, Trinity College Dublin, Ireland
| | - Marianna Kalaszi
- From the Department of Immunology (L.D., J.D., N.C.), St James's Hospital; School of Medicine (M.S., N.C.), Trinity College Dublin; MS Unit (M.K., H.K.), Department of Neurology, St James's Hospital; Department of Medical Gerontology (M.M.), School of Medicine, Trinity Translational Medicine Institute, Trinity College Dublin; Virology Laboratory (Y.L., K.O.D., C.R., A.Q., B.C.), St James's Hospital; and FutureNeuro SFI Research Centre (H.K.), Academic Unit of Neurology, School of Medicine, Trinity College Dublin, Ireland
| | - Matt McElheron
- From the Department of Immunology (L.D., J.D., N.C.), St James's Hospital; School of Medicine (M.S., N.C.), Trinity College Dublin; MS Unit (M.K., H.K.), Department of Neurology, St James's Hospital; Department of Medical Gerontology (M.M.), School of Medicine, Trinity Translational Medicine Institute, Trinity College Dublin; Virology Laboratory (Y.L., K.O.D., C.R., A.Q., B.C.), St James's Hospital; and FutureNeuro SFI Research Centre (H.K.), Academic Unit of Neurology, School of Medicine, Trinity College Dublin, Ireland
| | - Yvonne Lynagh
- From the Department of Immunology (L.D., J.D., N.C.), St James's Hospital; School of Medicine (M.S., N.C.), Trinity College Dublin; MS Unit (M.K., H.K.), Department of Neurology, St James's Hospital; Department of Medical Gerontology (M.M.), School of Medicine, Trinity Translational Medicine Institute, Trinity College Dublin; Virology Laboratory (Y.L., K.O.D., C.R., A.Q., B.C.), St James's Hospital; and FutureNeuro SFI Research Centre (H.K.), Academic Unit of Neurology, School of Medicine, Trinity College Dublin, Ireland
| | - Kate O'Driscoll
- From the Department of Immunology (L.D., J.D., N.C.), St James's Hospital; School of Medicine (M.S., N.C.), Trinity College Dublin; MS Unit (M.K., H.K.), Department of Neurology, St James's Hospital; Department of Medical Gerontology (M.M.), School of Medicine, Trinity Translational Medicine Institute, Trinity College Dublin; Virology Laboratory (Y.L., K.O.D., C.R., A.Q., B.C.), St James's Hospital; and FutureNeuro SFI Research Centre (H.K.), Academic Unit of Neurology, School of Medicine, Trinity College Dublin, Ireland
| | - Carmel Roche
- From the Department of Immunology (L.D., J.D., N.C.), St James's Hospital; School of Medicine (M.S., N.C.), Trinity College Dublin; MS Unit (M.K., H.K.), Department of Neurology, St James's Hospital; Department of Medical Gerontology (M.M.), School of Medicine, Trinity Translational Medicine Institute, Trinity College Dublin; Virology Laboratory (Y.L., K.O.D., C.R., A.Q., B.C.), St James's Hospital; and FutureNeuro SFI Research Centre (H.K.), Academic Unit of Neurology, School of Medicine, Trinity College Dublin, Ireland
| | - Ammara Qureshi
- From the Department of Immunology (L.D., J.D., N.C.), St James's Hospital; School of Medicine (M.S., N.C.), Trinity College Dublin; MS Unit (M.K., H.K.), Department of Neurology, St James's Hospital; Department of Medical Gerontology (M.M.), School of Medicine, Trinity Translational Medicine Institute, Trinity College Dublin; Virology Laboratory (Y.L., K.O.D., C.R., A.Q., B.C.), St James's Hospital; and FutureNeuro SFI Research Centre (H.K.), Academic Unit of Neurology, School of Medicine, Trinity College Dublin, Ireland
| | - Brendan Crowley
- From the Department of Immunology (L.D., J.D., N.C.), St James's Hospital; School of Medicine (M.S., N.C.), Trinity College Dublin; MS Unit (M.K., H.K.), Department of Neurology, St James's Hospital; Department of Medical Gerontology (M.M.), School of Medicine, Trinity Translational Medicine Institute, Trinity College Dublin; Virology Laboratory (Y.L., K.O.D., C.R., A.Q., B.C.), St James's Hospital; and FutureNeuro SFI Research Centre (H.K.), Academic Unit of Neurology, School of Medicine, Trinity College Dublin, Ireland
| | - Niall Conlon
- From the Department of Immunology (L.D., J.D., N.C.), St James's Hospital; School of Medicine (M.S., N.C.), Trinity College Dublin; MS Unit (M.K., H.K.), Department of Neurology, St James's Hospital; Department of Medical Gerontology (M.M.), School of Medicine, Trinity Translational Medicine Institute, Trinity College Dublin; Virology Laboratory (Y.L., K.O.D., C.R., A.Q., B.C.), St James's Hospital; and FutureNeuro SFI Research Centre (H.K.), Academic Unit of Neurology, School of Medicine, Trinity College Dublin, Ireland
| | - Hugh Kearney
- From the Department of Immunology (L.D., J.D., N.C.), St James's Hospital; School of Medicine (M.S., N.C.), Trinity College Dublin; MS Unit (M.K., H.K.), Department of Neurology, St James's Hospital; Department of Medical Gerontology (M.M.), School of Medicine, Trinity Translational Medicine Institute, Trinity College Dublin; Virology Laboratory (Y.L., K.O.D., C.R., A.Q., B.C.), St James's Hospital; and FutureNeuro SFI Research Centre (H.K.), Academic Unit of Neurology, School of Medicine, Trinity College Dublin, Ireland
| |
Collapse
|
8
|
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
|
9
|
Yin R, Melton S, Huseby ES, Kardar M, Chakraborty AK. How persistent infection overcomes peripheral tolerance mechanisms to cause T cell-mediated autoimmune disease. Proc Natl Acad Sci U S A 2024; 121:e2318599121. [PMID: 38446856 PMCID: PMC10945823 DOI: 10.1073/pnas.2318599121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2023] [Accepted: 02/06/2024] [Indexed: 03/08/2024] Open
Abstract
T cells help orchestrate immune responses to pathogens, and their aberrant regulation can trigger autoimmunity. Recent studies highlight that a threshold number of T cells (a quorum) must be activated in a tissue to mount a functional immune response. These collective effects allow the T cell repertoire to respond to pathogens while suppressing autoimmunity due to circulating autoreactive T cells. Our computational studies show that increasing numbers of pathogenic peptides targeted by T cells during persistent or severe viral infections increase the probability of activating T cells that are weakly reactive to self-antigens (molecular mimicry). These T cells are easily re-activated by the self-antigens and contribute to exceeding the quorum threshold required to mount autoimmune responses. Rare peptides that activate many T cells are sampled more readily during severe/persistent infections than in acute infections, which amplifies these effects. Experiments in mice to test predictions from these mechanistic insights are suggested.
Collapse
Affiliation(s)
- Rose Yin
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA02139
| | - Samuel Melton
- Physics of Living Systems, Department of Physics, Massachusetts Institute of Technology, Cambridge, MA02139
| | - Eric S. Huseby
- Basic Pathology, Department of Pathology, University of Massachusetts Medical School, Worcester, MA01655
| | - Mehran Kardar
- Physics of Living Systems, Department of Physics, Massachusetts Institute of Technology, Cambridge, MA02139
| | - Arup K. Chakraborty
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA02139
- Physics of Living Systems, Department of Physics, Massachusetts Institute of Technology, Cambridge, MA02139
- Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology and Harvard University, Cambridge, MA02139
- Department of Chemistry, Massachusetts Institute of Technology, Cambridge, MA02139
| |
Collapse
|
10
|
Gutiérrez-Guerrero A, Espinosa-Padilla SE, Lugo-Reyes SO. [Anything that can go wrong: cytotoxic cells and their control of Epstein-Barr virus]. REVISTA ALERGIA MÉXICO 2024; 71:29-39. [PMID: 38683066 DOI: 10.29262/ram.v71i1.1276] [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: 06/30/2023] [Accepted: 08/31/2023] [Indexed: 05/01/2024] Open
Abstract
Epstein-Barr virus (EBV) is an gamma of herpes virus affecting exclusively humans, was the first oncogenic virus described and is associated with over seven different cancers. Curiously, the exchange of genes during viral infections has enabled the evolution of other cellular organisms, favoring new functions and the survival of the host. EBV has been co-evolving with mammals for hundreds of millions of years, and more than 95% of adults have been infected in one moment of their life. The infection is acquired primarily during childhood, in most cases as an asymptomatic infection. However, during adolescence or young adulthood, around 10 to 30% develop infectious mononucleosis. The NK and CD8+ T cells are the cytotoxic cells of the immune system that focus on antiviral responses. Importantly, an essential role of NK and CD8+ T cells has been demonstrated during the control and elimination of EBV-infected cells. Nonetheless, when the cytotoxic function of these cells is compromised, the infection increases the risk of developing lymphoproliferative diseases and cancer, often fatal. In this review, we delineate EBV infection and the importance of cytotoxic responses by NK and CD8+ T cells during the control and elimination of EBV-infected cells. Furthermore, we briefly discuss the main inborn errors of immunity that compromise cytotoxic responses by NK and CD8+ T cells, and how this scenario affects the antiviral response during EBV infection. Finally, we conclude the review by underlying the need for an effective EBV vaccine capable of preventing infection and the consequent development of malignancies and autoimmune diseases.
Collapse
Affiliation(s)
- Arturo Gutiérrez-Guerrero
- Laboratorio de Inmunodeficiencias, Instituto Nacional de Pediatría, Secretaría de Salud, Ciudad de México
| | - Sara Elva Espinosa-Padilla
- Laboratorio de Inmunodeficiencias, Instituto Nacional de Pediatría, Secretaría de Salud, Ciudad de México
| | - Saúl Oswaldo Lugo-Reyes
- Laboratorio de Inmunodeficiencias, Instituto Nacional de Pediatría, Secretaría de Salud, Ciudad de México
| |
Collapse
|
11
|
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
|
12
|
Vasileiou ES, Fitzgerald KC. Multiple Sclerosis Pathogenesis and Updates in Targeted Therapeutic Approaches. Curr Allergy Asthma Rep 2023; 23:481-496. [PMID: 37402064 DOI: 10.1007/s11882-023-01102-0] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/06/2023] [Indexed: 07/05/2023]
Abstract
PURPOSE OF REVIEW In this review, we provide a comprehensive update on current scientific advances and emerging therapeutic approaches in the field of multiple sclerosis. RECENT FINDINGS Multiple sclerosis (MS) is a common disorder characterized by inflammation and degeneration within the central nervous system (CNS). MS is the leading cause of non-traumatic disability in the young adult population. Through ongoing research, an improved understanding of the disease underlying mechanisms and contributing factors has been achieved. As a result, therapeutic advancements and interventions have been developed specifically targeting the inflammatory components that influence disease outcome. Recently, a new type of immunomodulatory treatment, known as Bruton tyrosine kinase (BTK) inhibitors, has surfaced as a promising tool to combat disease outcomes. Additionally, there is a renewed interested in Epstein-Barr virus (EBV) as a major potentiator of MS. Current research efforts are focused on addressing the gaps in our understanding of the pathogenesis of MS, particularly with respect to non-inflammatory drivers. Significant and compelling evidence suggests that the pathogenesis of MS is complex and requires a comprehensive, multilevel intervention strategy. This review aims to provide an overview of MS pathophysiology and highlights the most recent advances in disease-modifying therapies and other therapeutic interventions.
Collapse
Affiliation(s)
- Eleni S Vasileiou
- Department of Neurology, Johns Hopkins University, Baltimore, MD, USA
| | - Kathryn C Fitzgerald
- Department of Neurology, Johns Hopkins University, Baltimore, MD, USA.
- Department of Epidemiology, Johns Hopkins University, Baltimore, MD, USA.
| |
Collapse
|
13
|
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
|
14
|
Chen B, Julg B, Mohandas S, Bradfute SB. Viral persistence, reactivation, and mechanisms of long COVID. eLife 2023; 12:e86015. [PMID: 37140960 PMCID: PMC10159620 DOI: 10.7554/elife.86015;] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2023] [Accepted: 04/24/2023] [Indexed: 08/28/2024] Open
Abstract
The COVID-19 global pandemic caused by the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection has infected hundreds of millions of individuals. Following COVID-19 infection, a subset can develop a wide range of chronic symptoms affecting diverse organ systems referred to as post-acute sequelae of SARS-CoV-2 infection (PASC), also known as long COVID. A National Institutes of Health-sponsored initiative, RECOVER: Researching COVID to Enhance Recovery, has sought to understand the basis of long COVID in a large cohort. Given the range of symptoms that occur in long COVID, the mechanisms that may underlie these diverse symptoms may also be diverse. In this review, we focus on the emerging literature supporting the role(s) that viral persistence or reactivation of viruses may play in PASC. Persistence of SARS-CoV-2 RNA or antigens is reported in some organs, yet the mechanism by which they do so and how they may be associated with pathogenic immune responses is unclear. Understanding the mechanisms of persistence of RNA, antigen or other reactivated viruses and how they may relate to specific inflammatory responses that drive symptoms of PASC may provide a rationale for treatment.
Collapse
Affiliation(s)
- Benjamin Chen
- Division of Infectious Diseases, Department of Medicine, Icahn School of Medicine at Mount SinaiNew YorkUnited States
| | - Boris Julg
- Infectious Diseases Division, Massachusetts General Hospital, Ragon Institute of Mass General, MIT and HarvardBostonUnited States
| | - Sindhu Mohandas
- Division of Infectious Diseases, Department of Pediatrics, Children’s Hospital Los Angeles, Keck School of Medicine, University of Southern CaliforniaLos AngelesUnited States
| | - Steven B Bradfute
- Center for Global Health, Department of Internal Medicine, University of New Mexico Health Sciences CenterAlbuquerqueUnited States
| |
Collapse
|
15
|
Chen B, Julg B, Mohandas S, Bradfute SB. Viral persistence, reactivation, and mechanisms of long COVID. eLife 2023; 12:e86015. [PMID: 37140960 PMCID: PMC10159620 DOI: 10.7554/elife.86015] [Citation(s) in RCA: 87] [Impact Index Per Article: 43.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2023] [Accepted: 04/24/2023] [Indexed: 05/05/2023] Open
Abstract
The COVID-19 global pandemic caused by the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection has infected hundreds of millions of individuals. Following COVID-19 infection, a subset can develop a wide range of chronic symptoms affecting diverse organ systems referred to as post-acute sequelae of SARS-CoV-2 infection (PASC), also known as long COVID. A National Institutes of Health-sponsored initiative, RECOVER: Researching COVID to Enhance Recovery, has sought to understand the basis of long COVID in a large cohort. Given the range of symptoms that occur in long COVID, the mechanisms that may underlie these diverse symptoms may also be diverse. In this review, we focus on the emerging literature supporting the role(s) that viral persistence or reactivation of viruses may play in PASC. Persistence of SARS-CoV-2 RNA or antigens is reported in some organs, yet the mechanism by which they do so and how they may be associated with pathogenic immune responses is unclear. Understanding the mechanisms of persistence of RNA, antigen or other reactivated viruses and how they may relate to specific inflammatory responses that drive symptoms of PASC may provide a rationale for treatment.
Collapse
Affiliation(s)
- Benjamin Chen
- Division of Infectious Diseases, Department of Medicine, Icahn School of Medicine at Mount SinaiNew YorkUnited States
| | - Boris Julg
- Infectious Diseases Division, Massachusetts General Hospital, Ragon Institute of Mass General, MIT and HarvardBostonUnited States
| | - Sindhu Mohandas
- Division of Infectious Diseases, Department of Pediatrics, Children’s Hospital Los Angeles, Keck School of Medicine, University of Southern CaliforniaLos AngelesUnited States
| | - Steven B Bradfute
- Center for Global Health, Department of Internal Medicine, University of New Mexico Health Sciences CenterAlbuquerqueUnited States
| |
Collapse
|
16
|
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: 49] [Impact Index Per Article: 24.5] [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
|
17
|
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: 12] [Impact Index Per Article: 6.0] [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
|
18
|
Sundaresan B, Shirafkan F, Ripperger K, Rattay K. The Role of Viral Infections in the Onset of Autoimmune Diseases. Viruses 2023; 15:v15030782. [PMID: 36992490 PMCID: PMC10051805 DOI: 10.3390/v15030782] [Citation(s) in RCA: 27] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2023] [Revised: 03/16/2023] [Accepted: 03/17/2023] [Indexed: 03/31/2023] Open
Abstract
Autoimmune diseases (AIDs) are the consequence of a breach in immune tolerance, leading to the inability to sufficiently differentiate between self and non-self. Immune reactions that are targeted towards self-antigens can ultimately lead to the destruction of the host's cells and the development of autoimmune diseases. Although autoimmune disorders are comparatively rare, the worldwide incidence and prevalence is increasing, and they have major adverse implications for mortality and morbidity. Genetic and environmental factors are thought to be the major factors contributing to the development of autoimmunity. Viral infections are one of the environmental triggers that can lead to autoimmunity. Current research suggests that several mechanisms, such as molecular mimicry, epitope spreading, and bystander activation, can cause viral-induced autoimmunity. Here we describe the latest insights into the pathomechanisms of viral-induced autoimmune diseases and discuss recent findings on COVID-19 infections and the development of AIDs.
Collapse
Affiliation(s)
- Bhargavi Sundaresan
- Institute of Pharmacology, Biochemical Pharmacological Center, University of Marburg, 35043 Marburg, Germany
| | - Fatemeh Shirafkan
- Institute of Pharmacology, Biochemical Pharmacological Center, University of Marburg, 35043 Marburg, Germany
| | - Kevin Ripperger
- Institute of Pharmacology, Biochemical Pharmacological Center, University of Marburg, 35043 Marburg, Germany
| | - Kristin Rattay
- Institute of Pharmacology, Biochemical Pharmacological Center, University of Marburg, 35043 Marburg, Germany
| |
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
|
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
|
21
|
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: 31] [Impact Index Per Article: 10.3] [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
|
22
|
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
|
23
|
MINI-review of Epstein-Barr virus involvement in multiple sclerosis etiology and pathogenesis. J Neuroimmunol 2022; 371:577935. [DOI: 10.1016/j.jneuroim.2022.577935] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2022] [Revised: 07/11/2022] [Accepted: 07/24/2022] [Indexed: 11/18/2022]
|
24
|
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: 35] [Impact Index Per Article: 8.8] [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
|
25
|
Wang Z, Kennedy PG, Dupree C, Wang M, Lee C, Pointon T, Langford TD, Graner MW, Yu X. Antibodies from Multiple Sclerosis Brain Identified Epstein-Barr Virus Nuclear Antigen 1 & 2 Epitopes which Are Recognized by Oligoclonal Bands. J Neuroimmune Pharmacol 2021; 16:567-580. [PMID: 32808238 PMCID: PMC7431217 DOI: 10.1007/s11481-020-09948-1] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2020] [Accepted: 07/27/2020] [Indexed: 12/30/2022]
Abstract
Multiple sclerosis (MS) is a chronic inflammatory demyelinating disease of the central nervous system (CNS), the etiology of which is poorly understood. The most common laboratory abnormality associated with MS is increased intrathecal immunoglobulin G (IgG) synthesis and the presence of oligoclonal bands (OCBs) in the brain and cerebrospinal fluid (CSF). However, the major antigenic targets of these antibody responses are unknown. The risk of MS is increased after infectious mononucleosis (IM) due to EBV infection, and MS patients have higher serum titers of anti-EBV antibodies than control populations. Our goal was to identify disease-relevant epitopes of IgG antibodies in MS; to do so, we screened phage-displayed random peptide libraries (12-mer) with total IgG antibodies purified from the brain of a patient with acute MS. We identified and characterized the phage peptides for binding specificity to intrathecal IgG from patients with MS and from controls by ELISA, phage-mediated Immuno-PCR, and isoelectric focusing. We identified two phage peptides that share sequence homologies with EBV nuclear antigens 1 and 2 (EBNA1 and EBNA2), respectively. The specificity of the EBV epitopes found by panning with MS brain IgG was confirmed by ELISA and competitive inhibition assays. Using a highly sensitive phage-mediated immuno-PCR assay, we determined specific bindings of the two EBV epitopes to IgG from CSF from 46 MS and 5 inflammatory control (IC) patients. MS CSF IgG have significantly higher bindings to EBNA1 epitope than to EBNA2 epitope, whereas EBNA1 and EBNA2 did not significantly differ in binding to IC CSF IgG. Further, the EBNA1 epitope was recognized by OCBs from multiple MS CSF as shown in blotting assays with samples separated by isoelectric focusing. The EBNA1 epitope is reactive to MS intrathecal antibodies corresponding to oligoclonal bands. This reinforces the potential role of EBV in the etiology of MS. Graphical abstract Antibodies purified from an MS brain plaque were panned by phage display peptide libraries to discern potential antigens. Phage displaying peptide sequences resembling Epstein-Barr Virus Nuclear Antigens 1 & 2 (EBNA1 & 2) epitopes were identified. Antibodies from sera and CSF from other MS patients also reacted to those epitopes.
Collapse
Affiliation(s)
- Zhe Wang
- National Engineering Research Center for Protein Drugs, Beijing, 102206, China
| | - Peter Ge Kennedy
- Institute of Infection, Immunity and Inflammation, University of Glasgow, Glasgow, UK
| | - Cecily Dupree
- Department of Neurology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Min Wang
- Immunoah Therapeutics, Inc., 12635 East Montview Boulevard, Aurora, CO, 80045, USA
| | - Catherin Lee
- Department of Neurology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Tiffany Pointon
- Department of Neurology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - T Dianne Langford
- Lewis Katz School of Medicine, Temple University, 3500 N. Broad St, Philadelphia, PA, 19140, USA
| | - Michael W Graner
- Department of Neurosurgery, University of Colorado Anschutz Medical Campus, Aurora, CO, USA.
| | - Xiaoli Yu
- Department of Neurosurgery, University of Colorado Anschutz Medical Campus, Aurora, CO, USA.
| |
Collapse
|
26
|
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
|
27
|
Houen G, Trier NH, Frederiksen JL. Epstein-Barr Virus and Multiple Sclerosis. Front Immunol 2020; 11:587078. [PMID: 33391262 PMCID: PMC7773893 DOI: 10.3389/fimmu.2020.587078] [Citation(s) in RCA: 49] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2020] [Accepted: 11/18/2020] [Indexed: 12/11/2022] Open
Abstract
Multiple sclerosis (MS) is a neurologic disease affecting myelinated nerves in the central nervous system (CNS). The disease often debuts as a clinically isolated syndrome, e.g., optic neuritis (ON), which later develops into relapsing-remitting (RR) MS, with temporal attacks or primary progressive (PP) MS. Characteristic features of MS are inflammatory foci in the CNS and intrathecal synthesis of immunoglobulins (Igs), measured as an IgG index, oligoclonal bands (OCBs), or specific antibody indexes. Major predisposing factors for MS are certain tissue types (e.g., HLA DRB1*15:01), vitamin D deficiency, smoking, obesity, and infection with Epstein-Barr virus (EBV). Many of the clinical signs of MS described above can be explained by chronic/recurrent EBV infection and current models of EBV involvement suggest that RRMS may be caused by repeated entry of EBV-transformed B cells to the CNS in connection with attacks, while PPMS may be caused by more chronic activity of EBV-transformed B cells in the CNS. In line with the model of EBV's role in MS, new treatments based on monoclonal antibodies (MAbs) targeting B cells have shown good efficacy in clinical trials both for RRMS and PPMS, while MAbs inhibiting B cell mobilization and entry to the CNS have shown efficacy in RRMS. Thus, these agents, which are now first line therapy in many patients, may be hypothesized to function by counteracting a chronic EBV infection.
Collapse
Affiliation(s)
- Gunnar Houen
- Institute of Biochemistry and Molecular Biology, University of Southern Denmark, Odense, Denmark
- Department of Neurology, Rigshospitalet, Glostrup, Denmark
| | | | - Jette Lautrup Frederiksen
- Department of Neurology, Rigshospitalet, Glostrup, Denmark
- Institute of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
28
|
Soldan SS, Lieberman PM. Epstein-Barr Virus Infection in the Development of Neurological Disorders. ACTA ACUST UNITED AC 2020; 32:35-52. [PMID: 33897799 DOI: 10.1016/j.ddmod.2020.01.001] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Epstein-Barr Virus (EBV) is a ubiquitous human herpesvirus that contributes to the etiology of diverse human cancers and auto-immune diseases. EBV establishes a relatively benign, long-term latent infection in over 90 percent of the adult population. Yet, it also increases risk for certain cancers and auto-immune disorders depending on complex viral, host, and environmental factors that are only partly understood. EBV latent infection is found predominantly in memory B-cells, but the natural infection cycle and pathological aberrations enable EBV to infect numerous other cell types, including oral, nasopharyngeal, and gastric epithelia, B-, T-, and NK-lymphoid cells, myocytes, adipocytes, astrocytes, and neurons. EBV infected cells, free virus, and gene products can also be found in the CNS. In addition to the direct effects of EBV on infected cells and tissue, the effect of chronic EBV infection on the immune system is also thought to contribute to pathogenesis, especially auto-immune disease. Here, we review properties of EBV infection that may shed light on its potential pathogenic role in neurological disorders.
Collapse
|
29
|
Epstein-Barr Virus-Specific CD8 T Cells Selectively Infiltrate the Brain in Multiple Sclerosis and Interact Locally with Virus-Infected Cells: Clue for a Virus-Driven Immunopathological Mechanism. J Virol 2019; 93:JVI.00980-19. [PMID: 31578295 PMCID: PMC6880158 DOI: 10.1128/jvi.00980-19] [Citation(s) in RCA: 64] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2019] [Accepted: 09/24/2019] [Indexed: 12/14/2022] Open
Abstract
EBV establishes a lifelong and asymptomatic infection in most individuals and more rarely causes infectious mononucleosis and malignancies, like lymphomas. The virus is also strongly associated with MS, a chronic neuroinflammatory disease with unknown etiology. Infectious mononucleosis increases the risk of developing MS, and immune reactivity toward EBV is higher in persons with MS, indicating inadequate control of the virus. Previous studies have suggested that persistent EBV infection in the CNS stimulates an immunopathological response, causing bystander neural cell damage. To verify this, we need to identify the immune culprits responsible for the detrimental antiviral response in the CNS. In this study, we analyzed postmortem brains donated by persons with MS and show that CD8 cytotoxic T cells recognizing EBV enter the brain and interact locally with the virus-infected cells. This antiviral CD8 T cell-mediated immune response likely contributes to MS pathology. Epstein-Barr virus (EBV) is a ubiquitous herpesvirus strongly associated with multiple sclerosis (MS), a chronic inflammatory disease of the central nervous system (CNS). However, the mechanisms linking EBV infection to MS pathology are uncertain. Neuropathological and immunological studies suggest that a persistent EBV infection in the CNS can stimulate a CD8 T-cell response aimed at clearing the virus but inadvertently causing CNS injury. Inasmuch as in situ demonstration of EBV-specific CD8 T cells and their effector function is missing, we searched for EBV-specific CD8 T cells in MS brain tissue using the pentamer technique. Postmortem brain samples from 12 donors with progressive MS and known HLA class I genotype were analyzed. Brain sections were stained with HLA-matched pentamers coupled with immunogenic peptides from EBV-encoded proteins, control virus (cytomegalovirus and influenza A virus) proteins, and myelin basic protein. CD8 T cells recognizing proteins expressed in the latent and lytic phases of the EBV life cycle were visualized in white matter lesions and/or meninges of 11/12 MS donors. The fraction (median value) of CD8 T cells recognizing individual EBV epitopes ranged from 0.5 to 2.5% of CNS-infiltrating CD8 T cells. Cytomegalovirus-specific CD8 T cells were detected at a lower frequency (≤0.3%) in brain sections from 4/12 MS donors. CNS-infiltrating EBV-specific CD8 T cells were CD107a positive, suggesting a cytotoxic phenotype, and stuck to EBV-infected cells. Together with local EBV dysregulation, selective enrichment of EBV-specific CD8 T cells in the MS brain supports the notion that skewed immune responses toward EBV contribute to inflammation causing CNS injury. IMPORTANCE EBV establishes a lifelong and asymptomatic infection in most individuals and more rarely causes infectious mononucleosis and malignancies, like lymphomas. The virus is also strongly associated with MS, a chronic neuroinflammatory disease with unknown etiology. Infectious mononucleosis increases the risk of developing MS, and immune reactivity toward EBV is higher in persons with MS, indicating inadequate control of the virus. Previous studies have suggested that persistent EBV infection in the CNS stimulates an immunopathological response, causing bystander neural cell damage. To verify this, we need to identify the immune culprits responsible for the detrimental antiviral response in the CNS. In this study, we analyzed postmortem brains donated by persons with MS and show that CD8 cytotoxic T cells recognizing EBV enter the brain and interact locally with the virus-infected cells. This antiviral CD8 T cell-mediated immune response likely contributes to MS pathology.
Collapse
|
30
|
Forrest C, Hislop AD, Rickinson AB, Zuo J. Proteome-wide analysis of CD8+ T cell responses to EBV reveals differences between primary and persistent infection. PLoS Pathog 2018; 14:e1007110. [PMID: 30248160 PMCID: PMC6171963 DOI: 10.1371/journal.ppat.1007110] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2018] [Revised: 10/04/2018] [Accepted: 08/27/2018] [Indexed: 02/07/2023] Open
Abstract
Human herpesviruses are antigenically rich agents that induce strong CD8+T cell responses in primary infection yet persist for life, continually challenging T cell memory through recurrent lytic replication and potentially influencing the spectrum of antigen-specific responses. Here we describe the first lytic proteome-wide analysis of CD8+ T cell responses to a gamma1-herpesvirus, Epstein-Barr virus (EBV), and the first such proteome-wide analysis of primary versus memory CD8+ T cell responses to any human herpesvirus. Primary effector preparations were generated directly from activated CD8+ T cells in the blood of infectious mononucleosis (IM) patients by in vitro mitogenic expansion. For memory preparations, EBV-specific cells in the blood of long-term virus carriers were first re-stimulated in vitro by autologous dendritic cells loaded with a lysate of lytically-infected cells, then expanded as for IM cells. Preparations from 7 donors of each type were screened against each of 70 EBV lytic cycle proteins in combination with the donor's individual HLA class I alleles. Multiple reactivities against immediate early (IE), early (E) and late (L) lytic cycle proteins, including many hitherto unrecognised targets, were detected in both contexts. Interestingly however, the two donor cohorts showed a different balance between IE, E and L reactivities. Primary responses targeted IE and a small group of E proteins preferentially, seemingly in line with their better presentation on the infected cell surface before later-expressed viral evasins take full hold. By contrast, target choice equilibrates in virus carriage with responses to key IE and E antigens still present but with responses to a select subset of L proteins now often prominent. We infer that, for EBV at least, long-term virus carriage with its low level virus replication and lytic antigen release is associated with a re-shaping of the virus-specific response.
Collapse
Affiliation(s)
- Calum Forrest
- Institute for Immunology and Immunotherapy, College of Medical and Dental Sciences, University of Birmingham, Birmingham, United Kingdom
| | - Andrew D. Hislop
- Institute for Immunology and Immunotherapy, College of Medical and Dental Sciences, University of Birmingham, Birmingham, United Kingdom
| | - Alan B. Rickinson
- Institute for Immunology and Immunotherapy, College of Medical and Dental Sciences, University of Birmingham, Birmingham, United Kingdom
| | - Jianmin Zuo
- Institute for Immunology and Immunotherapy, College of Medical and Dental Sciences, University of Birmingham, Birmingham, United Kingdom
| |
Collapse
|
31
|
van Langelaar J, van der Vuurst de Vries RM, Janssen M, Wierenga-Wolf AF, Spilt IM, Siepman TA, Dankers W, Verjans GMGM, de Vries HE, Lubberts E, Hintzen RQ, van Luijn MM. T helper 17.1 cells associate with multiple sclerosis disease activity: perspectives for early intervention. Brain 2018; 141:1334-1349. [DOI: 10.1093/brain/awy069] [Citation(s) in RCA: 109] [Impact Index Per Article: 15.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2017] [Accepted: 01/19/2018] [Indexed: 12/13/2022] Open
Affiliation(s)
- Jamie van Langelaar
- Department of Immunology, Erasmus MC, University Medical Center, Rotterdam, The Netherlands
- MS Center ErasMS at Erasmus MC, University Medical Center, Rotterdam, The Netherlands
| | - Roos M van der Vuurst de Vries
- MS Center ErasMS at Erasmus MC, University Medical Center, Rotterdam, The Netherlands
- Department of Neurology, Erasmus MC, University Medical Center, Rotterdam, The Netherlands
| | - Malou Janssen
- Department of Immunology, Erasmus MC, University Medical Center, Rotterdam, The Netherlands
- MS Center ErasMS at Erasmus MC, University Medical Center, Rotterdam, The Netherlands
- Department of Neurology, Erasmus MC, University Medical Center, Rotterdam, The Netherlands
| | - Annet F Wierenga-Wolf
- Department of Immunology, Erasmus MC, University Medical Center, Rotterdam, The Netherlands
- MS Center ErasMS at Erasmus MC, University Medical Center, Rotterdam, The Netherlands
| | - Isis M Spilt
- Department of Immunology, Erasmus MC, University Medical Center, Rotterdam, The Netherlands
- MS Center ErasMS at Erasmus MC, University Medical Center, Rotterdam, The Netherlands
| | - Theodora A Siepman
- MS Center ErasMS at Erasmus MC, University Medical Center, Rotterdam, The Netherlands
- Department of Neurology, Erasmus MC, University Medical Center, Rotterdam, The Netherlands
| | - Wendy Dankers
- Department of Rheumatology, Erasmus MC, University Medical Center, Rotterdam, The Netherlands
| | - Georges M G M Verjans
- Department of Viroscience, Erasmus MC, University Medical Center, Rotterdam, The Netherlands
- Research Center for Emerging Infections and Zoonosis, University of Veterinary Medicine, Hannover, Germany
| | - Helga E de Vries
- Department of Molecular Cell Biology and Immunology; Neuroscience Campus Amsterdam, VU University Medical Center, Amsterdam, The Netherlands
| | - Erik Lubberts
- Department of Rheumatology, Erasmus MC, University Medical Center, Rotterdam, The Netherlands
| | - Rogier Q Hintzen
- Department of Immunology, Erasmus MC, University Medical Center, Rotterdam, The Netherlands
- MS Center ErasMS at Erasmus MC, University Medical Center, Rotterdam, The Netherlands
- Department of Neurology, Erasmus MC, University Medical Center, Rotterdam, The Netherlands
| | - Marvin M van Luijn
- Department of Immunology, Erasmus MC, University Medical Center, Rotterdam, The Netherlands
- MS Center ErasMS at Erasmus MC, University Medical Center, Rotterdam, The Netherlands
| |
Collapse
|
32
|
Serafini B, Zandee S, Rosicarelli B, Scorsi E, Veroni C, Larochelle C, D'Alfonso S, Prat A, Aloisi F. Epstein-Barr virus-associated immune reconstitution inflammatory syndrome as possible cause of fulminant multiple sclerosis relapse after natalizumab interruption. J Neuroimmunol 2018; 319:9-12. [PMID: 29685294 DOI: 10.1016/j.jneuroim.2018.03.011] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2018] [Revised: 03/12/2018] [Accepted: 03/16/2018] [Indexed: 10/17/2022]
Affiliation(s)
- Barbara Serafini
- Department of Neuroscience, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161 Rome, Italy.
| | - Stephanie Zandee
- Neuroimmunology Research Laboratory, Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), 900 Rue Saint-Denis, Tour Viger, Montréal, QC H2X 0A9, Canada
| | - Barbara Rosicarelli
- Department of Neuroscience, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161 Rome, Italy.
| | - Eleonora Scorsi
- 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.
| | - Catherine Larochelle
- Neuroimmunology Research Laboratory, Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), 900 Rue Saint-Denis, Tour Viger, Montréal, QC H2X 0A9, Canada.
| | - Sandra D'Alfonso
- Department of Health Sciences, University of Eastern Piedmont, Novara, Italy.
| | - Alexandre Prat
- Neuroimmunology Research Laboratory, Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), 900 Rue Saint-Denis, Tour Viger, Montréal, QC H2X 0A9, Canada; Department of Neurosciences, Faculty of Medicine, Université de Montréal, Montreal, QC H3T 1J4, Canada.
| | - Francesca Aloisi
- Department of Neuroscience, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161 Rome, Italy.
| |
Collapse
|
33
|
Veroni C, Serafini B, Rosicarelli B, Fagnani C, Aloisi F. Transcriptional profile and Epstein-Barr virus infection status of laser-cut immune infiltrates from the brain of patients with progressive multiple sclerosis. J Neuroinflammation 2018; 15:18. [PMID: 29338732 PMCID: PMC5771146 DOI: 10.1186/s12974-017-1049-5] [Citation(s) in RCA: 53] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2017] [Accepted: 12/21/2017] [Indexed: 01/01/2023] Open
Abstract
Background It is debated whether multiple sclerosis (MS) might result from an immunopathological response toward an active Epstein-Barr virus (EBV) infection brought into the central nervous system (CNS) by immigrating B cells. Based on this model, a relationship should exist between the local immune milieu and EBV infection status in the MS brain. To test this hypothesis, we analyzed expression of viral and cellular genes in brain-infiltrating immune cells. Methods Twenty-three postmortem snap-frozen brain tissue blocks from 11 patients with progressive MS were selected based on good RNA quality and prominent immune cell infiltration. White matter perivascular and intrameningeal immune infiltrates, including B cell follicle-like structures, were isolated from brain sections using laser capture microdissection. Enhanced PCR-based methods were used to investigate expression of 75 immune-related genes and 6 EBV genes associated with latent and lytic infection. Data were analyzed using univariate and multivariate statistical methods. Results Genes related to T cell activation, cytotoxic cell-mediated (or type 1) immunity, B cell growth and differentiation, pathogen recognition, myeloid cell function, type I interferon pathway activation, and leukocyte recruitment were found expressed at different levels in most or all MS brain immune infiltrates. EBV genes were detected in brain samples from 9 of 11 MS patients with expression patterns suggestive of in situ activation of latent infection and, less frequently, entry into the lytic cycle. Comparison of data obtained in meningeal and white matter infiltrates revealed higher expression of genes related to interferonγ production, B cell differentiation, cell proliferation, lipid antigen presentation, and T cell and myeloid cell recruitment, as well as more widespread EBV infection in the meningeal samples. Multivariate analysis grouped genes expressed in meningeal and white matter immune infiltrates into artificial factors that were characterized primarily by genes involved in type 1 immunity effector mechanisms and type I interferon pathway activation. Conclusion These results confirm profound in situ EBV deregulation and suggest orchestration of local antiviral function in the MS brain, lending support to a model of MS pathogenesis that involves EBV as possible antigenic stimulus of the persistent immune response in the central nervous system. Electronic supplementary material The online version of this article (10.1186/s12974-017-1049-5) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Caterina Veroni
- Department of Neuroscience, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161, Rome, Italy
| | - 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
| | - Corrado Fagnani
- Centre for Behavioural Sciences and Mental Health, 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
|
34
|
Cencioni MT, Magliozzi R, Nicholas R, Ali R, Malik O, Reynolds R, Borsellino G, Battistini L, Muraro PA. Programmed death 1 is highly expressed on CD8 + CD57 + T cells in patients with stable multiple sclerosis and inhibits their cytotoxic response to Epstein-Barr virus. Immunology 2017; 152:660-676. [PMID: 28767147 DOI: 10.1111/imm.12808] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2017] [Revised: 06/27/2017] [Accepted: 07/20/2017] [Indexed: 02/06/2023] Open
Abstract
Growing evidence points to a deregulated response to Epstein-Barr virus (EBV) in the central nervous system of patients with multiple sclerosis (MS) as a possible cause of disease. We have investigated the response of a subpopulation of effector CD8+ T cells to EBV in 36 healthy donors and in 35 patients with MS in active and inactive disease. We have measured the expression of markers of degranulation, the release of cytokines, cytotoxicity and the regulation of effector functions by inhibitory receptors, such as programmed death 1 (PD-1) and human inhibitor receptor immunoglobulin-like transcript 2 (ILT2). We demonstrate that polyfunctional cytotoxic CD8+ CD57+ T cells are able to kill EBV-infected cells in healthy donors. In contrast, an anergic exhaustion-like phenotype of CD8+ CD57+ T cells with high expression of PD-1 was observed in inactive patients with MS compared with active patients with MS or healthy donors. Detection of CD8+ CD57+ T cells in meningeal inflammatory infiltrates from post-mortem MS tissue confirmed the association of this cell phenotype with the disease pathological process. The overall results suggest that ineffective immune control of EBV in patietns with MS during remission may be one factor preceding and enabling the reactivation of the virus in the central nervous system and may cause exacerbation of the disease.
Collapse
Affiliation(s)
- Maria T Cencioni
- Department of Medicine, Division of Brain Sciences, Centre for Neuroscience, Wolfson Neuroscience Laboratories, Imperial College London, London, UK
| | - Roberta Magliozzi
- Department of Medicine, Division of Brain Sciences, Centre for Neuroscience, Wolfson Neuroscience Laboratories, Imperial College London, London, UK.,Department of Neurosciences, Biomedicine and Movement, University of Verona, Verona, Italy
| | - Richard Nicholas
- Department of Medicine, Division of Brain Sciences, Centre for Neuroscience, Wolfson Neuroscience Laboratories, Imperial College London, London, UK.,Department of Neurosciences, Imperial College Healthcare NHS Trust, London, UK
| | - Rehiana Ali
- Department of Medicine, Division of Brain Sciences, Centre for Neuroscience, Wolfson Neuroscience Laboratories, Imperial College London, London, UK.,Department of Neurosciences, Imperial College Healthcare NHS Trust, London, UK
| | - Omar Malik
- Department of Medicine, Division of Brain Sciences, Centre for Neuroscience, Wolfson Neuroscience Laboratories, Imperial College London, London, UK.,Department of Neurosciences, Imperial College Healthcare NHS Trust, London, UK
| | - Richard Reynolds
- Department of Medicine, Division of Brain Sciences, Centre for Neuroscience, Wolfson Neuroscience Laboratories, Imperial College London, London, UK
| | | | - Luca Battistini
- Neuroimmunology Unit, IRCCS Santa Lucia Foundation, Rome, Italy
| | - Paolo A Muraro
- Department of Medicine, Division of Brain Sciences, Centre for Neuroscience, Wolfson Neuroscience Laboratories, Imperial College London, London, UK.,Department of Neurosciences, Imperial College Healthcare NHS Trust, London, UK
| |
Collapse
|
35
|
Abstract
T cells are considered pivotal in the pathology of multiple sclerosis (MS), but their function and antigen specificity are unknown. To unravel the role of T cells in MS pathology, we performed a comprehensive analysis on T cells recovered from paired blood, cerebrospinal fluid (CSF), normal-appearing white matter (NAWM) and white matter lesions (WML) from 27 MS patients with advanced disease shortly after death. The differentiation status of T cells in these compartments was determined by ex vivo flow cytometry and immunohistochemistry. T-cell reactivity in short-term T-cell lines (TCL), generated by non-specific stimulation of T cells recovered from the same compartments, was determined by intracellular cytokine flow cytometry. Central memory T cells predominated in CSF and effector memory T cells were enriched in NAWM and WML. WML-derived CD8+ T cells represent chronically activated T cells expressing a cytotoxic effector phenotype (CD95L and granzyme B) indicative for local antigenic stimulation (CD137). The same lesions also contained higher CD8+ T-cell frequencies expressing co-inhibitory (TIM3 and PD1) and co-stimulatory (ICOS) T-cell receptors, yet no evidence for T-cell senescence (CD57) was observed. The oligoclonal T-cell receptor (TCR) repertoire, particularly among CD8+ T cells, correlated between TCL generated from anatomically separated WML of the same MS patient, but not between paired NAWM and WML. Whereas no substantial T-cell reactivity was detected towards seven candidate human MS-associated autoantigens (cMSAg), brisk CD8+ T-cell reactivity was detected in multiple WML-derived TCL towards autologous Epstein–Barr virus (EBV) infected B cells (autoBLCL). In one MS patient, the T-cell response towards autoBLCL in paired intra-lesional TCL was dominated by TCRVβ2+CD8+ T cells, which were localized in the parenchyma of the respective tissues expressing a polarized TCR and CD8 expression suggesting immunological synapse formation in situ. Collectively, the data suggest the involvement of effector memory cytotoxic T cells recognizing antigens expressed by autoBLCL, but not the assayed human cMSAg, in WML of MS patients.
Collapse
|
36
|
Impact of aging immune system on neurodegeneration and potential immunotherapies. Prog Neurobiol 2017; 157:2-28. [PMID: 28782588 DOI: 10.1016/j.pneurobio.2017.07.006] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2015] [Revised: 07/25/2017] [Accepted: 07/28/2017] [Indexed: 12/19/2022]
Abstract
The interaction between the nervous and immune systems during aging is an area of avid interest, but many aspects remain unclear. This is due, not only to the complexity of the aging process, but also to a mutual dependency and reciprocal causation of alterations and diseases between both the nervous and immune systems. Aging of the brain drives whole body systemic aging, including aging-related changes of the immune system. In turn, the immune system aging, particularly immunosenescence and T cell aging initiated by thymic involution that are sources of chronic inflammation in the elderly (termed inflammaging), potentially induces brain aging and memory loss in a reciprocal manner. Therefore, immunotherapeutics including modulation of inflammation, vaccination, cellular immune therapies and "protective autoimmunity" provide promising approaches to rejuvenate neuroinflammatory disorders and repair brain injury. In this review, we summarize recent discoveries linking the aging immune system with the development of neurodegeneration. Additionally, we discuss potential rejuvenation strategies, focusing aimed at targeting the aging immune system in an effort to prevent acute brain injury and chronic neurodegeneration during aging.
Collapse
|
37
|
Burnard S, Lechner-Scott J, Scott RJ. EBV and MS: Major cause, minor contribution or red-herring? Mult Scler Relat Disord 2017; 16:24-30. [DOI: 10.1016/j.msard.2017.06.002] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2017] [Revised: 06/05/2017] [Accepted: 06/09/2017] [Indexed: 10/19/2022]
|
38
|
Massive intracerebral Epstein-Barr virus reactivation in lethal multiple sclerosis relapse after natalizumab withdrawal. J Neuroimmunol 2017; 307:14-17. [PMID: 28495131 DOI: 10.1016/j.jneuroim.2017.03.013] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2017] [Revised: 03/08/2017] [Accepted: 03/15/2017] [Indexed: 01/06/2023]
Abstract
Rebound of disease activity in multiple sclerosis patients after natalizumab withdrawal is a potentially life-threatening event. To verify whether highly destructive inflammation after natalizumab withdrawal is associated with Epstein-Barr virus (EBV) reactivation in central nervous system infiltrating B-lineage cells and cytotoxic immunity, we analyzed post-mortem brain tissue from a patient who died during a fulminating MS relapse following natalizumab withdrawal. Numerous EBV infected B cells/plasma cells and CD8+ T cells infiltrated all white matter lesions; the highest frequency of EBV lytically infected cells and granzyme B+ CD8+ T cells was observed in actively demyelinating lesions. These results may encourage switching to B-cell depleting therapy after natalizumab discontinuation.
Collapse
|
39
|
Fierz W. Multiple sclerosis: an example of pathogenic viral interaction? Virol J 2017; 14:42. [PMID: 28241767 PMCID: PMC5330019 DOI: 10.1186/s12985-017-0719-3] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2016] [Accepted: 02/25/2017] [Indexed: 12/21/2022] Open
Abstract
A hypothesis is formulated on viral interaction between HHV-6A and EBV as a pathogenic mechanism in Multiple Sclerosis (MS). Evidence of molecular and genetic mechanisms suggests a link between HHV-6A infection and EBV activation in the brain of MS patients leading to intrathecal B-cell transformation. Consequent T-cell immune response against the EBV-infected cells is postulated as a pathogenic basis for inflammatory lesion formation in the brain of susceptible individuals. A further link between HHV-6A and EBV involves their induction of expression of the human endogenous retrovirus HERV-K18-encoded superantigen. Such virally induced T-cell responses might secondarily also lead to local autoimmune phenomena. Finally, research recommendations are formulated for substantiating the hypothesis on several levels: epidemiologically, genetically, and viral expression in the brain.
Collapse
Affiliation(s)
- Walter Fierz
- labormedizinisches zentrum Dr Risch, Landstr. 157, 9494, Schaan, Fürstentum, Liechtenstein.
| |
Collapse
|
40
|
Reiss CS. Virus-Induced Demyelination: The Case for Virus(es) in Multiple Sclerosis. NEUROTROPIC VIRAL INFECTIONS 2016. [PMCID: PMC7122906 DOI: 10.1007/978-3-319-33189-8_6] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Multiple Sclerosis (MS) is the most common demyelinating disease of man with over 400,000 cases in the United States and over 2.5 million cases worldwide. There are over 64,000 citations in Pubmed dating back as far as 1887. Much has been learned over the past 129 years with a recent burst in therapeutic options (mostly anti-inflammatory) with newer medications in development that are neuroprotective and/or neuroreparative. However, with all these advancements the cause of MS remains elusive. There is a clear interplay of genetic, immunologic, and environmental factors that influences both the development and progression of this disorder. This chapter will give a brief overview of the history and pathogenesis of MS with attention to how host immune responses in genetically susceptible individuals contribute to the MS disease process. In addition, we will explore the role of infectious agents in MS as potential “triggers” of disease. Models of virus-induced demyelination will be discussed, with an emphasis on the recent interest in human herpesviruses and the role they may play in MS disease pathogenesis. Although we remain circumspect as to the role of any microbial pathogen in MS, we suggest that only through well-controlled serological, cellular immune, molecular, and animal studies we will be able to identify candidate agents. Ultimately, clinical interventional trials that either target a specific pathogen or class of pathogens will be required to make definitive links between the suspected agent and MS.
Collapse
Affiliation(s)
- Carol Shoshkes Reiss
- Departments of Biology and Neural Science, New York University, New York, New York USA
| |
Collapse
|
41
|
Veroni C, Marnetto F, Granieri L, Bertolotto A, Ballerini C, Repice AM, Schirru L, Coghe G, Cocco E, Anastasiadou E, Puopolo M, Aloisi F. Immune and Epstein-Barr virus gene expression in cerebrospinal fluid and peripheral blood mononuclear cells from patients with relapsing-remitting multiple sclerosis. J Neuroinflammation 2015; 12:132. [PMID: 26169064 PMCID: PMC4501166 DOI: 10.1186/s12974-015-0353-1] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2015] [Accepted: 06/24/2015] [Indexed: 12/19/2022] Open
Abstract
Background Gene expression analyses in paired cerebrospinal fluid (CSF) and peripheral blood mononuclear cells (PBMC) from patients with multiple sclerosis (MS) are restrained by the low RNA amounts from CSF cells and low expression levels of certain genes. Here, we applied a Taqman-based pre-amplification real-time reverse-transcription polymerase chain reaction (RT-PCR) (PreAmp RT-PCR) to cDNA from CSF cells and PBMC of MS patients and analyzed multiple genes related to immune system function and genes expressed by Epstein-Barr virus (EBV), a herpesvirus showing strong association with MS. Using this enhanced RT-PCR method, we aimed at the following: (1) identifying gene signatures potentially useful for patient stratification, (2) understanding whether EBV infection is perturbed in CSF and/or blood, and (3) finding a link between immune and EBV infection status. Methods Thirty-one therapy-free patients with relapsing-remitting MS were included in the study. Paired CSF cells and PBMC were collected and expression of 41 immune-related cellular genes and 7 EBV genes associated with latent or lytic viral infection were determined by PreAmp RT-PCR. Clinical, radiological, CSF, and gene expression data were analyzed using univariate and multivariate (cluster analysis, factor analysis) statistical approaches. Results Several immune-related genes were differentially expressed between CSF cells and PBMC from the whole MS cohort. By univariate analysis, no or only minor differences in gene expression were found associated with sex, clinical, or radiological condition. Cluster analysis on CSF gene expression data grouped patients into three clusters; clusters 1 and 2 differed by expression of genes that are related mainly to innate immunity, irrespective of sex and disease characteristics. By factor analysis, two factors grouping genes involved in antiviral immunity and immune regulation, respectively, accurately discriminated cluster 1 and cluster 2 patients. Despite the use of an enhanced RT-PCR method, EBV transcripts were detected in a minority of patients (5 of 31), with evidence of viral latency activation in CSF cells or PBMC and of lytic infection in one patient with active disease only. Conclusions Analysis of multiple cellular and EBV genes in paired CSF cell and PBMC samples using PreAmp RT-PCR may yield new information on the complex interplay between biological processes underlying MS and help in biomarker identification. Electronic supplementary material The online version of this article (doi:10.1186/s12974-015-0353-1) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Caterina Veroni
- Department of Cell Biology and Neuroscience, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161, Rome, Italy.
| | - Fabiana Marnetto
- Neurology 2-CRESM (Multiple Sclerosis Regional Reference Center), AOU San Luigi Gonzaga, Regione Gonzole 10, 10043, Orbassano, Italy.
| | - Letizia Granieri
- Neurology 2-CRESM (Multiple Sclerosis Regional Reference Center), AOU San Luigi Gonzaga, Regione Gonzole 10, 10043, Orbassano, Italy.
| | - Antonio Bertolotto
- Neurology 2-CRESM (Multiple Sclerosis Regional Reference Center), AOU San Luigi Gonzaga, Regione Gonzole 10, 10043, Orbassano, Italy.
| | - Clara Ballerini
- Department of Neuroscience, Drug and Child Health (NEUROFARBA), University of Florence, Viale Pieraccini 6, 50137, Florence, Italy.
| | - Anna Maria Repice
- Multiple Sclerosis Center, Neurology 2 Division, Careggi University Hospital, University of Florence, Viale Morgagni 85, 50134, Florence, Italy.
| | - Lucia Schirru
- Department of Public Health, Clinical and Molecular Medicine, Multiple Sclerosis Center, University of Cagliari, Via Is Guadazzonis, 2, 09126, Cagliari, Italy.
| | - Giancarlo Coghe
- Department of Public Health, Clinical and Molecular Medicine, Multiple Sclerosis Center, University of Cagliari, Via Is Guadazzonis, 2, 09126, Cagliari, Italy.
| | - Eleonora Cocco
- Department of Public Health, Clinical and Molecular Medicine, Multiple Sclerosis Center, University of Cagliari, Via Is Guadazzonis, 2, 09126, Cagliari, Italy.
| | - Eleni Anastasiadou
- Department of Experimental Medicine, La Sapienza University, Viale Regina Elena 324, 00161, Rome, Italy. .,Department of Pathology, Beth Israel Deaconess Medical Center/Harvard Medical School, 330 Brookline Avenue, Boston, MA, 02215, USA.
| | - Maria Puopolo
- Department of Cell Biology and Neuroscience, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161, Rome, Italy.
| | - Francesca Aloisi
- Department of Cell Biology and Neuroscience, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161, Rome, Italy.
| |
Collapse
|