Naive CD4 T-cell activation identifies MS patients having rapid transition to progressive MS

Evelyn Zastepa, Leslie Fitz-Gerald, Michael Hallett, Jack Antel, Amit Bar-Or, Sergio Baranzini, Yves Lapierre, David G Haegert, Evelyn Zastepa, Leslie Fitz-Gerald, Michael Hallett, Jack Antel, Amit Bar-Or, Sergio Baranzini, Yves Lapierre, David G Haegert

Abstract

Objective: Our objective was to determine whether altered naive CD4 T-cell biology contributes to development of disease progression in secondary progressive multiple sclerosis (SPMS).

Methods: We compared the naive CD4 T-cell gene expression profiles of 19 patients with SPMS and 14 healthy controls (HCs) using a whole-genome microarray approach. We analyzed surface protein expression of critical genes by flow cytometry after T-cell receptor (TCR) stimulation of naive CD4 T cells isolated from HCs and patients with SPMS.

Results: Hierarchical clustering segregated patients with SPMS into 2 subgroups: SP-1, which had a short duration of relapsing-remitting multiple sclerosis (MS), and SP-2, which had a long duration of relapsing-remitting MS. SP-1 patients upregulated numerous immune genes, including genes within TCR and toll-like receptor (TLR) signaling pathways. SP-2 patients showed immune gene downregulation in comparison with HCs. We identified an SP-1-specific transcriptional signature of 3 genes (TLR4, TLR2, and chemokine receptor 1), and these genes had higher surface protein expression in SP-1 than in SP-2. After TCR stimulation for 48 hours, only SP-1 showed a progressive linear increase in TLR2 and TLR4 protein expression.

Conclusions: Differences in naive CD4 T-cell biology, notably of TCR and TLR signaling pathways, identified patients with MS with more rapid conversion to secondary progression, a critical determinant of long-term disability in MS.

Figures

Figure 1. Hierarchical clustering segregated HCs from…
Figure 1. Hierarchical clustering segregated HCs from patients with SPMS and identified 2 SPMS subgroups
Unsupervised hierarchical clustering (Ward clustering method and Euclidean distance metric) of 1,439 genes having the greatest variance across all samples segregated HCs from patients with SPMS and identified 2 SPMS subgroups (SP-1 and SP-2). Patients with SPMS clustering with HCs are indicated in gray. HC = healthy control; SPMS = secondary progressive multiple sclerosis.
Figure 2. Transcriptional signature of 3 genes…
Figure 2. Transcriptional signature of 3 genes distinguishes SP-1 patients from other patients with MS and HCs
(A) SP-1 patients have higher expression of TLR2, TLR4, and CCR1 genes than either HCs or SP-2 patients. Mean fold change values are shown for SP-1 vs SP-2. (B) Hierarchical clustering based on the 3-gene list distinguishes SP-1 patients. Hierarchical clustering segregated SP-1 from HCs, SP-2, patients with stable RRMS, and patients with PPMS. HC = healthy control; MS = multiple sclerosis; PPMS = primary progressive MS; RRMS = relapsing-remitting MS.
Figure 3. Surface expression of 3-gene signature…
Figure 3. Surface expression of 3-gene signature and CD28
(A) Increased protein expression on freshly isolated naive CD4 T cells from SP-1. SP-1 patients had significantly higher percentage of TLR2, TLR4, and CCR1 than HCs and SP-2 patients (95% confidence interval not shown) and higher TLR4 MFI than HCs and SP-2 patients (p < 0.05, both comparisons). (B) TCR stimulation increased TLR2 surface expression in SP-1. At 48 hours, SP-1 had increased percentage of TLR2 and MFI (p < 0.05, both comparisons) compared with unstimulated cells, with a positive linear increase in percentage of TLR2 from 0 to 48 hours (p = 0.02). At 48 hours, SP-1 had higher percentage of TLR2 expression than HCs and SP-2 (p < 0.001, both comparisons) and higher TLR2 MFI than HCs and SP-2 (p < 0.05, both comparisons). (C) TCR stimulation increased TLR4 surface expression in SP-1 patients. The percentage of TLR4 increased in HCs (p < 0.05) and SP-2 (p < 0.01) from 0 to 24 hours, then decreased from 24 to 48 hours. In SP-1, percentage TLR4 showed a positive linear increase from 0 to 48 hours (p = 0.0025). At 48 hours, SP-1 had higher percentage of TLR4 than HCs or SP-2 (p < 0.01, both comparisons) and higher TLR4 MFI than HCs or SP-2 (p < 0.05, both comparisons). (D) CD28 surface expression. TCR stimulation for 24 hours decreased percentage CD28 (p < 0.001) and CD28 MFI (p < 0.05, not shown) in all groups, but at 24 hours, SP-1 had higher percentage CD28 than HCs (p < 0.05) or SP-2 (p < 0.01) plus higher CD28 MFI than HCs (p < 0.05, not shown). HC = healthy control; MFI = mean or median fluorescence intensity; TCR = T-cell receptor.

References

    1. Muraro PA, Pette M, Bielekova B, McFarland HF, Martin R. Human autoreactive CD4+ T cells from naive CD45RA+ and memory CD45RO+ subsets differ with respect to epitope specificity and functional antigen avidity. J Immunol 2000;164:5474–5481
    1. Haegert DG, Hackenbroch JD, Duszczyszyn D, et al. Reduced thymic output and peripheral naive CD4 T-cell alterations in primary progressive multiple sclerosis (PPMS). J Neuroimmunol 2011;233:233–239
    1. Khoruts A, Fraser JM. A causal link between lymphopenia and autoimmunity. Immunol Lett 2005;98:23–31
    1. King C, Ilic A, Koelsch K, Sarvetnick N. Homeostatic expansion of T cells during immune insufficiency generates autoimmunity. Cell 2004;117:265–277
    1. Corvol JC, Pelletier D, Henry RG, et al. Abrogation of T cell quiescence characterizes patients at high risk for multiple sclerosis after the initial neurological event. Proc Natl Acad Sci USA 2008;105:11839–11844
    1. Confavreux C, Vukusic S, Adeleine P. Early clinical predictors and progression of irreversible disability in multiple sclerosis: an amnesic process. Brain 2003;126:770–782
    1. McDonald WI, Compston A, Edan G, et al. Recommended diagnostic criteria for multiple sclerosis: guidelines from the International Panel on the diagnosis of multiple sclerosis. Ann Neurol 2001;50:121–127
    1. Whitney AR, Diehn M, Popper SJ, et al. Individuality and variation in gene expression patterns in human blood. Proc Natl Acad Sci USA 2003;100:1896–1901
    1. Gautier L, Cope L, Bolstad BM, Irizarry RA. Affy: analysis of Affymetrix GeneChip data at the probe level. Bioinformatics 2004;20:307–315
    1. R Development Core Team R: a language and environment for statistical computing [online]. Available at: . Accessed February 1, 2011
    1. Yang Y, Paquet A, Dudoit S. Marray: exploratory analysis for two-colour spotted microarray data [online]. Available at: . Accessed February 10, 2011
    1. Jain A. Data clustering: 50 years beyond K-means. Pattern Recognit Lett 2010;31:651–666
    1. Mootha VK, Lindgren CM, Eriksson KF, et al. PGC-1alpha-responsive genes involved in oxidative phosphorylation are coordinately downregulated in human diabetes. Nat Genet 2003;34:267–273
    1. Subramanian A, Kuehn H, Gould J, Tamayo P, Mesirov JP. GSEA-P: a desktop application for gene set enrichment analysis. Bioinformatics 2007;23:3251–3253
    1. Schmittgen TD, Livak KJ. Analyzing real-time PCR data by the comparative C(T) method. Nat Protoc 2008;3:1101–1108
    1. Suzuki R, Shimodaira H. Pvclust: an R package for hierarchical clustering with p-values [online]. Available at: . Accessed April 15, 2010
    1. Wang M, Windgassen D, Papoutsakis ET. Comparative analysis of transcriptional profiling of CD3+, CD4+ and CD8+ T cells identifies novel immune response players in T-cell activation. BMC Genomics 2008;9:225.
    1. Chtanova T, Newton R, Liu SM, et al. Identification of T cell-restricted genes, and signatures for different T cell responses, using a comprehensive collection of microarray datasets. J Immunol 2005;175:7837–7847
    1. Sawcer S, Hellenthal G, Pirinen M, et al. Genetic risk and a primary role for cell-mediated immune mechanisms in multiple sclerosis. Nature 2011;476:214–219
    1. von Essen MR, Kongsbak M, Schjerling P, Olgaard K, Odum N, Geisler C. Vitamin D controls T cell antigen receptor signaling and activation of human T cells. Nat Immunol 2010;11:344–349
    1. Gonzalez-Navajas JM, Fine S, Law J, et al. TLR4 signaling in effector CD4+ T cells regulates TCR activation and experimental colitis in mice. J Clin Invest 2010;120:570–581
    1. Komai-Koma M, Jones L, Ogg GS, Xu D, Liew FY. TLR2 is expressed on activated T cells as a costimulatory receptor. Proc Natl Acad Sci USA 2004;101:3029–3034
    1. MacLeod H, Wetzler LM. T cell activation by TLRs: a role for TLRs in the adaptive immune response. Sci STKE 2007;2007:pe48.
    1. Wahlstrom J, Berlin M, Skold CM, Wigzell H, Eklund A, Grunewald J. Phenotypic analysis of lymphocytes and monocytes/macrophages in peripheral blood and bronchoalveolar lavage fluid from patients with pulmonary sarcoidosis. Thorax 1999;54:339–346
    1. Tremlett H, Yinshan Z, Devonshire V. Natural history of secondary-progressive multiple sclerosis. Mult Scler 2008;14:314–324
    1. Gandhi KS, McKay FC, Cox M, et al. The multiple sclerosis whole blood mRNA transcriptome and genetic associations indicate dysregulation of specific T cell pathways in pathogenesis. Hum Mol Genet 2010;19:2134–2143
    1. Duszczyszyn DA, Beck JD, Antel J, et al. Altered naive CD4 and CD8 T cell homeostasis in patients with relapsing-remitting multiple sclerosis: thymic versus peripheral (non-thymic) mechanisms. Clin Exp Immunol 2006;143:305–313
    1. Duszczyszyn DA, Williams JL, Mason H, Lapierre Y, Antel J, Haegert DG. Thymic involution and proliferative T-cell responses in multiple sclerosis. J Neuroimmunol 2010;221:73–80
    1. Babu S, Blauvelt CP, Kumaraswami V, Nutman TB. Cutting edge: diminished T cell TLR expression and function modulates the immune response in human filarial infection. J Immunol 2006;176:3885–3889
    1. Linsley PS, Bradshaw J, Urnes M, Grosmaire L, Ledbetter JA. CD28 engagement by B7/BB-1 induces transient down-regulation of CD28 synthesis and prolonged unresponsiveness to CD28 signaling. J Immunol 1993;150:3161–3169
    1. Achiron A, Grotto I, Balicer R, Magalashvili D, Feldman A, Gurevich M. Microarray analysis identifies altered regulation of nuclear receptor family members in the pre-disease state of multiple sclerosis. Neurobiol Dis 2010;38:201–209
    1. Satoh J, Nakanishi M, Koike F, et al. T cell gene expression profiling identifies distinct subgroups of Japanese multiple sclerosis patients. J Neuroimmunol 2006;174:108–118
    1. Panitch H, Miller A, Paty D, Weinshenker B. Interferon beta-1b in secondary progressive MS: results from a 3-year controlled study. Neurology 2004;63:1788–1795
    1. European Study Group on Interferon Beta-1b in Secondary Progressive MS Placebo-controlled multicentre randomised trial of interferon beta-1b in treatment of secondary progressive multiple sclerosis. Lancet 1998;352:1491–1497
    1. Kappos L, Weinshenker B, Pozzilli C, et al. Interferon beta-1b in secondary progressive MS: a combined analysis of the two trials. Neurology 2004;63:1779–1787

Source: PubMed

3
Předplatit