Defective T-cell control of Epstein-Barr virus infection in multiple sclerosis

Michael P Pender, Peter A Csurhes, Jacqueline M Burrows, Scott R Burrows, Michael P Pender, Peter A Csurhes, Jacqueline M Burrows, Scott R Burrows

Abstract

Mounting evidence indicates that infection with Epstein-Barr virus (EBV) has a major role in the pathogenesis of multiple sclerosis (MS). Defective elimination of EBV-infected B cells by CD8+ T cells might cause MS by allowing EBV-infected autoreactive B cells to accumulate in the brain. Here we undertake a comprehensive analysis of the T-cell response to EBV in MS, using flow cytometry and intracellular IFN-γ staining to measure T-cell responses to EBV-infected autologous lymphoblastoid cell lines and pools of human leukocyte antigen (HLA)-class-I-restricted peptides from EBV lytic or latent proteins and cytomegalovirus (CMV), in 95 patients and 56 EBV-seropositive healthy subjects. In 20 HLA-A2+ healthy subjects and 20 HLA-A2+ patients we also analysed CD8+ T cells specific for individual peptides, measured by binding to HLA-peptide complexes and production of IFN-γ, TNF-α and IL-2. We found a decreased CD8+ T-cell response to EBV lytic, but not CMV lytic, antigens at the onset of MS and at all subsequent disease stages. CD8+ T cells directed against EBV latent antigens were increased but had reduced cytokine polyfunctionality indicating T-cell exhaustion. During attacks the EBV-specific CD4+ and CD8+ T-cell populations expanded, with increased functionality of latent-specific CD8+ T cells. With increasing disease duration, EBV-specific CD4+ and CD8+ T cells progressively declined, consistent with T-cell exhaustion. The anti-EBNA1 IgG titre correlated inversely with the EBV-specific CD8+ T-cell frequency. We postulate that defective CD8+ T-cell control of EBV reactivation leads to an expanded population of latently infected cells, including autoreactive B cells.

Conflict of interest statement

The authors declare no conflict of interest.

Figures

Figure 1
Figure 1
Frequencies of T cells producing IFN-γ in response to autologous EBV-infected LCL. (a) Median percentages of T cells of different phenotypes producing IFN-γ in response to autologous EBV-infected LCL in the PBMC in EBV-seropositive healthy controls (HC), patients with clinically isolated syndrome (CIS), patients with relapsing–remitting (RR), secondary progressive (SP) and primary progressive (PP) MS, as well as patients during a clinical attack (Attack) and during remission (Remission). (b) Percentages of LCL-specific T cells in the PBMC in patients not having a clinical attack (Remission+SP+PP) compared with HC (medians with interquartile ranges indicated by black horizontal lines), with bracketed P values determined by the Mann–Whitney test. (c) Percentage of LCL-specific CD8+ EM/EMRA T cells in the PBMC plotted against the percentage of total CD8+ EM/EMRA T cells in the PBMC in healthy subjects (HC) and the total group of MS patients (MS). (d) Percentages of LCL-specific cells within the CD3+, CD4+, CD8+, CD4+ EM, CD4+ EMRA+, CD4+ CM, CD4+ naive, CD8+ EM, CD8+ EMRA, CD8+ CM and CD8+ naive T-cell populations in patients during a clinical attack (Attack) compared with patients not having an attack (Remission+SP+PP) (medians with interquartile ranges indicated by black horizontal lines), with bracketed P values determined by the Mann–Whitney test. (e) The frequency of LCL-specific T cells within the CD8+ population strongly correlated with the frequency of LCL-specific T cells within the CD4+ population in MS patients whereas this correlation was weaker in healthy subjects. On multiple linear regression analysis, the slope of the regression line in the MS patients was significantly greater than that in healthy subjects (P=0.006).
Figure 2
Figure 2
The relationship between the frequency of LCL-specific T cells and duration of MS. (ac) In the total group of patients, the percentage of LCL-specific T cells within the CD8+ CM population progressively decreased with increasing duration of MS (a), as did the frequency of LCL-specific T cells within the CD8+ EM population (b), and within the CD4+ EM population (c). (d) In patients having an attack the percentage of LCL-specific T cells within the CD4+ EM population also declined with increasing disease duration.
Figure 3
Figure 3
Decreased CD8+ T-cell response to EBV lytic phase antigens throughout the course of MS. (a) Median percentages of CD8+ T cells of different phenotypes producing IFN-γ in response to a pool of HLA-class-I-restricted EBV lytic peptides in the PBMC in EBV-seropositive healthy controls (HC), the total group of MS patients (All MS), patients with clinically isolated syndrome (CIS), patients with relapsing–remitting (RR), secondary progressive (SP) and primary progressive (PP) MS, as well as patients during a clinical attack (Attack) and during remission (Remission). As expected, there was no CD4+ T-cell response to these HLA-class-I-restricted peptides. (b) Percentages of EBV-lytic-specific CD8+ T cells in the PBMC in the total group of patients (MS) compared with healthy controls (HC) (medians with interquartile ranges indicated by black horizontal lines), with bracketed P values determined by the Mann–Whitney test. (c) Percentages of EBV-lytic-specific CD8+ T cells in the PBMC in HLA-A*02+ patients (A2+ MS) compared with HLA-A*02+ healthy controls (A2+ HC) (medians with interquartile ranges indicated by black horizontal lines), with bracketedP values determined by the Mann–Whitney test. (d) Percentages of CD8+ T cells of different phenotypes producing IFN-γ in response to a pool of HLA-class-I-restricted CMV peptides in the PBMC in CMV-seropositive HC, and CMV-seropositive MS patients (MS) (medians with interquartile ranges indicated by black horizontal lines), with bracketedP-values determined by the Mann–Whitney test. There was no CD4+ T-cell response.
Figure 4
Figure 4
Skewing of the EBV-specific CD8+ T-cell response in MS away from lytic to latent antigens. (a) Median percentages of CD8+ T cells within the different phenotypes producing IFN-γ in response to a pool of HLA-class-I-restricted EBV latent peptides in EBV-seropositive healthy controls (HC), the total group of MS patients (All MS), patients with clinically isolated syndrome (CIS), patients with relapsing–remitting (RR), secondary progressive (SP) and primary progressive (PP) MS, as well as patients during a clinical attack (Attack) and during remission (Remission). As expected, there was no CD4+ T-cell response to these HLA-class-I-restricted peptides. (b) Percentages of latent-specific cells within the CD8+, CD8+ EM, CD8+ EMRA, CD8+ CM and CD8+ EM/EMRA T-cell populations in patients with MS (MS) compared with healthy controls (HC) (medians with interquartile ranges indicated by black horizontal lines), with bracketedP-values determined by the Mann–Whitney test. (c) Median lytic/latent ratio within the different phenotypes in EBV-seropositive HC, the total group of MS patients (All MS), patients with CIS, patients with RR, SP and PP MS, as well as patients during a clinical attack (Attack) and during remission (Remission). For each subject the ratio was calculated by dividing the frequency of CD8+ T cells producing IFN-γ in response to pooled lytic peptides by the frequency of CD8+ T cells producing IFN-γ in response to pooled latent peptides. (d) Lytic/latent ratio within the CD8+, CD8+ EM, CD8+ EMRA, CD8+ CM and CD8+ EM/EMRA T-cell populations in the total group of MS patients (MS) compared with HC (medians with interquartile ranges indicated by black horizontal lines), with bracketedP values determined by the Mann–Whitney test. (e andf) Percentages of T cells specific for individual EBV latent (LLD, CLG and FLY) and lytic (GLC) peptides in the CD8+ population in 20 HLA-A*02+ EBV-seropositive healthy subjects (HC) and 20 HLA-A*02+ MS patients (MS) measured by binding to peptide-HLA-A*02 Dextramers (e) and IFN-γ production (f) (medians with interquartile ranges indicated by black horizontal lines), with bracketed P-values determined by the Mann–Whitney test. (gand h) Percentages of T cells specific for individual EBV latent and lytic peptides in the CD8+ population in HLA-A*02+ patients during clinical attacks (Attack) or during remission (Remission) measured by binding to peptide-HLA-A*02 Dextramers (g) and IFN-γ production (h) (medians with interquartile ranges indicated by black horizontal lines), with bracketed P-values determined by the Mann–Whitney test.
Figure 5
Figure 5
EBV-specific CD8+ T-cell polyfunctionality in MS. (a andb) Polyfunctionality of CD8+ T cells specific for individual EBV latent (LLD, CLG and FLY) and lytic (GLC) peptides in 20 HLA-A*02+ EBV-seropositive healthy subjects (HC) and 20 HLA-A*02+ MS patients (MS) determined by measuring the frequencies of T cells producing IL-2, TNF-α and IFN-γ in response to stimulation with each peptide and, in a different tube of cells, the frequencies of T cells binding to the respective peptide-HLA-A*02 Dextramer. (a) Ratio of the polyfunctionality index to the percentage of T cells binding the respective peptide-HLA-A*02 Dextramer in the CD8+ population (medians with interquartile ranges indicated by black horizontal lines), with bracketed P-values determined by the Mann–Whitney test. (b) Pie charts showing the mean percentages of Dextramer+ CD8+ T cells producing zero, one, two or three cytokines. (c and d) Polyfunctionality of CD8+ T cells specific for individual EBV latent and lytic peptides in HLA-A*02+ MS patients during clinical attacks (Attack) or during remission (Remission). (c) Ratio of the polyfunctionality index to the percentage of T cells binding the respective peptide-HLA-A*02 Dextramer in the CD8+ population (medians with interquartile ranges indicated by black horizontal lines), with bracketed P-values determined by the Mann–Whitney test. (d) Pie charts showing the mean percentages of Dextramer+ CD8+ T cells producing zero, one, two or three cytokines.
Figure 6
Figure 6
The relationships of EBV genome load and anti-EBV antibody titres with the frequency of EBV-specific CD8+ T cells in MS. (a) EBV DNA copy number in the PBMC in healthy EBV-seropositive subjects (healthy controls (HC)) and patients with MS (MS) (medians with interquartile ranges indicated by black horizontal lines), with P-value determined by the Mann–Whitney test. (b) Relationship between the EBV genome load and the LCL-specific CD8+ T-cell frequency in MS patients. (c) Titres of anti-EBNA1 IgG and anti-VCA IgG in the sera of healthy EBV-seropositive subjects (HC) and patients with MS (MS) (medians with interquartile ranges indicated by black horizontal lines), with bracketed P-values determined by the Mann–Whitney test. (d) Relationship between the anti-EBNA1 IgG titre and the LCL-specific CD8+ EMRA T-cell frequency in the PBMC in MS patients. (e) Relationship between the anti-VCA IgG titre and the LCL-specific CD8+ EMRA T-cell frequency in the PBMC in MS patients (f) Relationship between the anti-VCA IgG titre and the EBV genome load in HC and patients with MS (MS). (g) Relationship between the anti-VCA IgG titre and the EBV genome load in the combined groups of HC and patients with MS. (h) Relationship between the anti-EBNA1 IgG titre and the EBV genome load in HC and patients with MS.
Figure 7
Figure 7
Proposed model of defective CD8+ T-cell control of EBV infection in MS. In healthy EBV carriers, (a) there is a dynamic equilibrium between the EBV-infected cell populations and the T-cell response. EBV-specific CD8+ T cells (T cell) exert a key role in controlling EBV infection by killing infected cells in the B blast, germinal centre (GC) B cell, plasma cell and tonsil epithelial cell, but not memory B cell, populations. The large arrows indicate the cycle of EBV infection: virion→B blast→GC B cell→memory B cell→plasma cell→virion→epithelial cell→virion→B blast. Smaller arrows indicate stimulation of T cells by EBV antigens from the infected populations. The relative sizes of the different EBV-infected cell populations are indicated by the circle sizes, based on the study by Hawkins et al. The relative sizes of the EBV-specific CD8+ T-cell populations are also indicated by the circle sizes; however, it is important to note that the EBV-specific CD8+ T-cell population is several orders of magnitude larger than the EBV-infected cell population, a distinction not depicted here. For simplicity, the EBV-specific CD4+ T-cell population and anti-EBV antibody response are not shown. At all stages of MS (bd) the EBV-lytic-specific CD8+ T-cell population is decreased, allowing increased production of virions which infect naive B cells driving them into the blast phase. The resultant expansion of the infected blast population stimulates EBV-latent-specific CD8+ T cells which proliferate and restrict this expansion, but not without increased flow out of infected blast cells into a consequently enlarged EBV-infected GC cell population, which in turn is partially controlled by the augmented EBV-latent-specific CD8+ T-cell population. In the same way the EBV-infected memory B cell pool also grows, as does the population of plasma cells reactivating EBV infection. During clinical attacks of MS (c) there is increased differentiation of EBV-infected memory B cells into lytically infected plasma cells as a result of the various microbial infections that trigger attacks of MS. This EBV reactivation is inadequately regulated by the already deficient EBV-lytic-specific CD8+ T-cell response, resulting in increased virion production and increased infection of the blast pool, this in turn stimulating proliferation of the EBV-latent-specific CD8+ T-cell population which restricts further growth of the infected blast population. In progressive MS (d) the EBV-latent-specific CD8+ T-cell response becomes exhausted (indicated by fading), resulting in unchecked expansion of the infected GC population and the development of EBV-infected lymphoid tissue in the CNS.

References

    1. Ascherio A, Munger KL, Lünemann JD. The initiation and prevention of multiple sclerosis. Nat Rev Neurol 2012; 8: 602–612.
    1. Pender MP, Burrows SR. Epstein–Barr virus and multiple sclerosis: potential opportunities for immunotherapy. Clin Transl Immunol 2014; 3: e27.
    1. Thorley-Lawson DA, Gross A. Persistence of the Epstein-Barr virus and the origins of associated lymphomas. N Engl J Med 2004; 350: 1328–1337.
    1. Laichalk LL, Thorley-Lawson DA. Terminal differentiation into plasma cells initiates the replicative cycle of Epstein-Barr virus in vivo. J Virol 2005; 79: 1296–1307.
    1. Hadinoto V, Shapiro M, Sun CC, Thorley-Lawson DA. The dynamics of EBV shedding implicate a central role for epithelial cells in amplifying viral output. PLoS Pathog 2009; 5: e1000496.
    1. Hawkins JB, Delgado-Eckert E, Thorley-Lawson DA, Shapiro M. The cycle of EBV infection explains persistence, the sizes of the infected cell populations and which come under CTL regulation. PLoS Pathog 2013; 9: e1003685.
    1. Khanna R, Burrows SR. Role of cytotoxic T lymphocytes in Epstein-Barr virus-associated diseases. Annu Rev Microbiol 2000; 54: 19–48.
    1. Hislop AD, Taylor GS, Sauce D, Rickinson AB. Cellular responses to viral infection in humans: lessons from Epstein-Barr virus. Annu Rev Immunol 2007; 25: 587–617.
    1. Pender MP. Infection of autoreactive B lymphocytes with EBV, causing chronic autoimmune diseases. Trends Immunol 2003; 24: 584–588.
    1. Pender MP. The essential role of Epstein–Barr virus in the pathogenesis of multiple sclerosis. Neuroscientist 2011; 17: 351–367.
    1. Sallusto F, Lenig D, Förster R, Lipp M, Lanzavecchia A. Two subsets of memory T lymphocytes with distinct homing potentials and effector functions. Nature 1999; 401: 708–712.
    1. Amyes E, McMichael AJ, Callan MFC. Human CD4+ T cells are predominantly distributed among six phenotypically and functionally distinct subsets. J Immunol 2005; 175: 5765–5773.
    1. Pender MP, Csurhes PA, Pfluger CMM, Burrows SR. Deficiency of CD8+ effector memory T cells is an early and persistent feature of multiple sclerosis. Mult Scler 2014; 20: 1825–1832.
    1. Bergmann CC, Altman JD, Hinton D, Stohlman SA. Inverted immunodominance and impaired cytolytic function of CD8+ T cells during viral persistence in the central nervous system. J Immunol 1999; 163: 3379–3387.
    1. Ifergan I, Kebir H, Alvarez JI, Marceau G, Bernard M, Bourbonnière L et al. Central nervous system recruitment of effector memory CD8+ T lymphocytes during neuroinflammation is dependent on α4 integrin. Brain 2011; 134: 3560–3577.
    1. Craig JC, Haire M, Millar JHD, Fraser KB, Merrett JD. Immunological control of Epstein–Barr virus-transformed lymphocytes in multiple sclerosis. Clin Immunol Immunopathol 1983; 29: 86–93.
    1. Craig JC, Hawkins SA, Swallow MW, Lyttle JA, Patterson VH, Merrett JD et al. Subsets of T lymphocytes in relation to T lymphocyte function in multiple sclerosis. Clin Exp Immunol 1985; 61: 548–555.
    1. Craig JC, Haire M, Merrett JD. T-cell-mediated suppression of Epstein-Barr virus-induced B lymphocyte activation in multiple sclerosis. Clin Immunol Immunopathol 1988; 48: 253–260.
    1. Pender MP, Csurhes PA, Lenarczyk A, Pfluger CMM, Burrows SR. Decreased T cell reactivity to Epstein-Barr virus infected lymphoblastoid cell lines in multiple sclerosis. J Neurol Neurosurg Psychiatry 2009; 80: 498–505.
    1. Lindsey JW, Hatfield LM. Epstein-Barr virus and multiple sclerosis: cellular immune response and cross-reactivity. J Neuroimmunol 2010; 229: 238–242.
    1. Jilek S, Schluep M, Harari A, Canales M, Lysandropoulos A, Zekeridou A et al. HLA-B7-restricted EBV-specific CD8+ T cells are dysregulated in multiple sclerosis. J Immunol 2012; 188: 4671–4680.
    1. Gronen F, Ruprecht K, Weissbrich B, Klinker E, Kroner A, Hofstetter HH et al. Frequency analysis of HLA-B7-restricted Epstein-Barr virus-specific cytotoxic T lymphocytes in patients with multiple sclerosis and healthy controls. J Neuroimmunol 2006; 180: 185–192.
    1. Höllsberg P, Hansen HJ, Haahr S. Altered CD8+ T cell responses to selected Epstein-Barr virus immunodominant epitopes in patients with multiple sclerosis. Clin Exp Immunol 2003; 132: 137–143.
    1. Cepok S, Zhou D, Srivastava R, Nessler S, Stei S, Büssow K et al. Identification of Epstein-Barr virus proteins as putative targets of the immune response in multiple sclerosis. J Clin Invest 2005; 115: 1352–1360.
    1. Lünemann JD, Edwards N, Muraro PA, Hayashi S, Cohen JI, Münz C et al. Increased frequency and broadened specificity of latent EBV nuclear antigen-1-specific T cells in multiple sclerosis. Brain 2006; 129: 1493–1506.
    1. Jilek S, Schluep M, Meylan P, Vingerhoets F, Guignard L, Monney A et al. Strong EBV-specific CD8+ T-cell response in patients with early multiple sclerosis. Brain 2008; 131: 1712–1721.
    1. Angelini DF, Serafini B, Piras E, Severa M, Coccia EM, Rosicarelli B et al. Increased CD8+ T cell response to Epstein-Barr virus lytic antigens in the active phase of multiple sclerosis. PLoS Pathog 2013; 9: e1003220.
    1. Pudney VA, Leese AM, Rickinson AB, Hislop AD. CD8+ immunodominance among Epstein-Barr virus lytic cycle antigens directly reflects the efficiency of antigen presentation in lytically infected cells. J Exp Med 2005; 201: 349–360.
    1. Tynan FE, Elhassen D, Purcell AW, Burrows JM, Borg NA, Miles JJ et al. The immunogenicity of a viral cytotoxic T cell epitope is controlled by its MHC-bound conformation. J Exp Med 2005; 202: 1249–1260.
    1. Pender MP, Csurhes PA, Pfluger CMM, Burrows SR. CD8+ T cells far predominate over CD4+ T cells in healthy immune response to Epstein-Barr virus infected lymphoblastoid cell lines. Blood 2012; 120: 5085–5087.
    1. Brynedal B, Duvefelt K, Jonasdottir G, Roos IM, Åkesson E, Palmgren J et al. HLA-A confers an HLA-DRB1 independent influence on the risk of multiple sclerosis. PLoS ONE 2007; 2: e664.
    1. Scalfari A, Neuhaus A, Degenhardt A, Rice GP, Muraro PA, Daumer M et al. The natural history of multiple sclerosis, a geographically based study 10: relapses and long-term disability. Brain 2010; 133: 1914–1929.
    1. Wherry EJ, Blattman JN, Murali-Krishna K, van der Most R, Ahmed R. Viral persistence alters CD8 T-cell immunodominance and tissue distribution and results in distinct stages of functional impairment. J Virol 2003; 77: 4911–4927.
    1. Kahan SM, Wherry EJ, Zajac AJ. T cell exhaustion during persistent viral infections. Virology 2015; 479–480: 180–193.
    1. Larsen M, Sauce D, Arnaud L, Fastenackels S, Appay V, Gorochov G. Evaluating cellular polyfunctionality with a novel polyfunctionality index. PLoS ONE 2012; 7: e42403.
    1. Ryan JL, Fan H, Glaser SL, Schichman SA, Raab-Traub N, Gulley ML. Epstein-Barr virus quantitation by real-time PCR targeting multiple gene segments: a novel approach to screen for the virus in paraffin-embedded tissue and plasma. J Mol Diagn 2004; 6: 378–385.
    1. Yea C, Tellier R, Chong P, Westmacott G, Marrie RA, Bar-Or A et al. Epstein-Barr virus in oral shedding of children with multiple sclerosis. Neurology 2013; 81: 1392–1399.
    1. Hopwood PA, Brooks L, Parratt R, Hunt BJ, Maria B, Alero TJ et al. Persistent Epstein-Barr virus infection: unrestricted latent and lytic viral gene expression in healthy immunosuppressed transplant recipients. Transplantation 2002; 74: 194–202.
    1. Alfieri C, Birkenbach M, Kieff E. Early events in Epstein–Barr virus infection of human B lymphocytes. Virology 1991; 181: 595–608.
    1. Serafini B, Rosicarelli B, Franciotta D, Magliozzi R, Reynolds R, Cinque P et al. Dysregulated Epstein-Barr virus infection in the multiple sclerosis brain. J Exp Med 2007; 204: 2899–2912.
    1. Fraser KB, Millar JHD, Haire M, McCrea S. Increased tendency to spontaneous in-vitro lymphocyte transformation in clinically active multiple sclerosis. Lancet 1979; 314: 715–717.
    1. Munch M, Møller-Larsen A, Christensen T, Morling N, Hansen HJ, Haahr S. B-lymphoblastoid cell lines from multiple sclerosis patients and a healthy control producing a putative new human retrovirus and Epstein–Barr virus. Mult Scler 1995; 1: 78–81.
    1. Tørring C, Andreasen C, Gehr N, Bjerg L, Petersen T, Höllsberg P. Higher incidence of Epstein–Barr virus-induced lymphocyte transformation in multiple sclerosis. Acta Neurol Scand 2014; 130: 90–96.
    1. Tzartos JS, Khan G, Vossenkamper A, Cruz-Sadaba M, Lonardi S, Sefia E et al. Association of innate immune activation with latent Epstein-Barr virus in active MS lesions. Neurology 2012; 78: 15–23.
    1. Serafini B, Muzio L, Rosicarelli B, Aloisi F. Radioactive in situ hybridization for Epstein-Barr virus-encoded small RNA supports presence of Epstein-Barr virus in the multiple sclerosis brain. Brain 2013; 136: e233.
    1. Latham LB, Lee MJ, Lincoln JA, Ji N, Forsthuber TG, Lindsey JW. Antivirus immune activity in multiple sclerosis correlates with MRI activity. Acta Neurol Scand 2016; 133: 17–24.
    1. Sibley WA, Bamford CR, Clark K. Clinical viral infections and multiple sclerosis. Lancet 1985; 325: 1313–1315.
    1. Panitch HS. Influence of infection on exacerbations of multiple sclerosis. Ann Neurol 1994; 36: S25–S28.
    1. Correale J, Fiol M, Gilmore W. The risk of relapses in multiple sclerosis during systemic infections. Neurology 2006; 67: 652–659.
    1. Aalto SM, Linnavuori K, Peltola H, Vuori E, Weissbrich B, Schubert J et al. Immunoreactivation of Epstein-Barr virus due to cytomegalovirus primary infection. J Med Virol 1998; 56: 186–191.
    1. Donati D, Espmark E, Kironde F, Mbidde EK, Kamya M, Lundkvist Å et al. Clearance of circulating Epstein-Barr virus DNA in children with acute malaria after antimalaria treatment. J Infect Dis 2006; 193: 971–977.
    1. Tuuminen T, Kekäläinen E, Mäkelä S, Ala-Houhala I, Ennis FA, Hedman K et al. Human CD8+ T cell memory generation in Puumala hantavirus infection occurs after the acute phase and is associated with boosting of EBV-specific CD8+ memory T cells. J Immunol 2007; 179: 1988–1995.
    1. Ueda S, Uchiyama S, Azzi T, Gysin C, Berger C, Bernasconi M et al. Oropharyngeal Group A streptococcal colonization disrupts latent Epstein-Barr virus infection. J Infect Dis 2014; 209: 255–264.
    1. Walton AH, Muenzer JT, Rasche D, Boomer JS, Sato B, Brownstein BH et al. Reactivation of multiple viruses in patients with sepsis. PLoS ONE 2014; 9: e98819.
    1. Pender MP, Csurhes PA, Greer JM, Mowat PD, Henderson RD, Cameron KD et al. Surges of increased T cell reactivity to an encephalitogenic region of myelin proteolipid protein occur more often in patients with multiple sclerosis than in healthy subjects. J Immunol 2000; 165: 5322–5331.
    1. Farez MF, Correale J. Yellow fever vaccination and increased relapse rate in travelers with multiple sclerosis. Arch Neurol 2011; 68: 1267–1271.
    1. Gudgeon NH, Taylor GS, Long HM, Haigh TA, Rickinson AB. Regression of Epstein-Barr virus-induced B-cell transformation in vitro involves virus-specific CD8+ T cells as the principal effectors and a novel CD4+ T-cell reactivity. J Virol 2005; 79: 5477–5488.
    1. West EE, Youngblood B, Tan WG, Jin H-T, Araki K, Alexe G et al. Tight regulation of memory CD8+ T cells limits their effectiveness during sustained high viral load. Immunity 2011; 35: 285–298.
    1. Kolumam GA, Thomas S, Thompson LJ, Sprent J, Murali-Krishna K. Type I interferons act directly on CD8 T cells to allow clonal expansion and memory formation in response to viral infection. J Exp Med 2005; 202: 637–650.
    1. Wandinger K-P, Wessel K, Neustock P, Siekhaus A, Kirchner H. Diminished production of type-I interferons and interleukin-2 in patients with multiple sclerosis. J Neurol Sci 1997; 149: 87–93.
    1. Stasiolek M, Bayas A, Kruse N, Wieczarkowiecz A, Toyka KV, Gold R et al. Impaired maturation and altered regulatory function of plasmacytoid dendritic cells in multiple sclerosis. Brain 2006; 129: 1293–1305.
    1. Klenerman P, Oxenius A. T cell responses to cytomegalovirus. Nat Rev Immunol 2016; 16: 367–377.
    1. Lenman JAR, Peters TJ. Herpes zoster and multiple sclerosis. Br Med J 1969; 2: 218–220.
    1. Berr C, Puel J, Clanet M, Ruidavets JB, Mas JL, Alperovitch A. Risk factors in multiple sclerosis: a population-based case-control study in Hautes-Pyrénées, France. Acta Neurol Scand 1989; 80: 46–50.
    1. Ross RT, Cheang M, Landry G, Klassen L, Doerksen K. Herpes zoster and multiple sclerosis. Can J Neurol Sci 1999; 26: 29–32.
    1. Kang J-H, Sheu J-J, Kao S, Lin H-C. Increased risk of multiple sclerosis following herpes zoster: a nationwide, population-based study. J Infect Dis 2011; 204: 188–192.
    1. Mackay LK, Long HM, Brooks JM, Taylor GS, Leung CS, Chen A et al. T cell detection of a B-cell tropic virus infection: newly-synthesised versus mature viral proteins as antigen sources for CD4 and CD8 epitope display. PLoS Pathog 2009; 5: e1000699.
    1. Lünemann JD, Tintoré M, Messmer B, Strowig T, Rovira A, Perkal H et al. Elevated Epstein–Barr virus-encoded nuclear antigen-1 immune responses predict conversion to multiple sclerosis. Ann Neurol 2010; 67: 159–169.
    1. Ascherio A, Munger KL, Lennette ET, Spiegelman D, Hernán MA, Olek MJ et al. Epstein-Barr virus antibodies and risk of multiple sclerosis: a prospective study. JAMA 2001; 286: 3083–3088.
    1. Sundström P, Juto P, Wadell G, Hallmans G, Svenningsson A, Nyström L et al. An altered immune response to Epstein-Barr virus in multiple sclerosis: a prospective study. Neurology 2004; 62: 2277–2282.
    1. Levin LI, Munger KL, Rubertone MV, Peck CA, Lennette ET, Spiegelman D et al. Temporal relationship between elevation of Epstein-Barr virus antibody titers and initial onset of neurological symptoms in multiple sclerosis. JAMA 2005; 293: 2496–2500.
    1. Levin LI, Munger KL, O’Reilly EJ, Falk KI, Ascherio A. Primary infection with the Epstein-Barr virus and risk of multiple sclerosis. Ann Neurol 2010; 67: 824–830.
    1. Besson C, Amiel C, Le-Pendeven C, Brice P, Fermé C, Carde P et al. Positive correlation between Epstein-Barr virus viral load and anti-viral capsid immunoglobulin G titers determined for Hodgkin’s lymphoma patients and their relatives. J Clin Microbiol 2006; 44: 47–50.
    1. Jabs DA, Arnett FC, Bias WB, Beale MG. Familial abnormalities of lymphocyte function in a large Sjögren’s syndrome kindred. J Rheumatol 1986; 13: 320–326.
    1. Tosato G, Steinberg AD, Yarchoan R, Heilman CA, Pike SE, De Seau V et al. Abnormally elevated frequency of Epstein-Barr virus-infected B cells in the blood of patients with rheumatoid arthritis. J Clin Invest 1984; 73: 1789–1795.
    1. Gross AJ, Hochberg D, Rand WM, Thorley-Lawson DA. EBV and systemic lupus erythematosus: a new perspective. J Immunol 2005; 174: 6599–6607.
    1. Croia C, Serafini B, Bombardieri M, Kelly S, Humby F, Severa M et al. Epstein–Barr virus persistence and infection of autoreactive plasma cells in synovial lymphoid structures in rheumatoid arthritis. Ann Rheum Dis 2013; 72: 1559–1568.
    1. Croia C, Astorri E, Murray-Brown W, Willis A, Brokstad KA, Sutcliffe N et al. Implication of Epstein-Barr virus infection in disease-specific autoreactive B cell activation in ectopic lymphoid structures of Sjögren’s syndrome. Arthritis Rheumatol 2014; 66: 2545–2557.
    1. Pender MP, Csurhes PA, Smith C, Beagley L, Hooper KD, Raj M et al. Epstein–Barr virus-specific adoptive immunotherapy for progressive multiple sclerosis. Mult Scler 2014; 20: 1541–1544.
    1. Polman CH, Reingold SC, Edan G, Filippi M, Hartung H-P, Kappos L et al. Diagnostic criteria for multiple sclerosis: 2005 Revisions to the ‘McDonald Criteria’. Ann Neurol 2005; 58: 840–846.
    1. Polman CH, Reingold SC, Banwell B, Clanet M, Cohen JA, Filippi M et al. Diagnostic criteria for multiple sclerosis: 2010 Revisions to the McDonald Criteria. Ann Neurol 2011; 69: 292–302.
    1. Kurtzke JF. Rating neurologic impairment in multiple sclerosis: an expanded disability status scale (EDSS). Neurology 1983; 33: 1444–1452.
    1. Roxburgh RH, Seaman SR, Masterman T, Hensiek AE, Sawcer SJ, Vukusic S et al. Multiple Sclerosis Severity Score: using disability and disease duration to rate disease severity. Neurology 2005; 64: 1144–1151.
    1. Gandhi MK, Khanna R. Human cytomegalovirus: clinical aspects, immune regulation, and emerging treatments. Lancet Infect Dis 2004; 4: 725–738.
    1. Walker S, Fazou C, Crough T, Holdsworth R, Kiely P, Veale M et al. Ex vivo monitoring of human cytomegalovirus-specific CD8+ T-cell responses using QuantiFERON-CMV. Transpl Infect Dis 2007; 9: 165–170.

Source: PubMed

3
Abonnieren