Functional status of peripheral blood T-cells in ischemic stroke patients

Antje Vogelgesang, Verena E L May, Uwe Grunwald, Maren Bakkeboe, Soenke Langner, Henry Wallaschofski, Christof Kessler, Barbara M Bröker, Alexander Dressel, Antje Vogelgesang, Verena E L May, Uwe Grunwald, Maren Bakkeboe, Soenke Langner, Henry Wallaschofski, Christof Kessler, Barbara M Bröker, Alexander Dressel

Abstract

Stroke is a major cause of disability and leading cause of death in the northern hemisphere. Only recently it became evident that cerebral ischemia not only leads to brain tissue damage and subsequent local inflammation but also to a dramatic loss of peripheral blood T-cells with subsequent infections. However, only scarce information is available on the activation status of surviving T cells. This study therefore addressed the functional consequences of immunological changes induced by stroke in humans. For this purpose peripheral blood T-cells were isolated from 93 stroke patients and the expression of activation makers was determined. In addition ex vivo stimulation assays were applied to asses the functionality of T cells derived from blood of stroke patients. Compared to healthy controls, stroke patients demonstrated an enhanced surface expression of HLA-DR (p<0.0001) and CD25 (p = 0.02) on T cells, revealing that stroke leads to T cell activation, while CTLA-4 remained undetectable. In vitro studies revealed that catecholamines inhibit CTLA-4 upregulation in activated T cells. Ex vivo, T cells of stroke patients proliferated unimpaired and released increased amounts of the proinflammatory cytokine TNF-alpha (p<0.01) and IL-6 (p<0.05). Also, in sera of stroke patients HMGB1 concentrations were increased (p = 0.0002). The data demonstrate that surviving T cells in stroke patients remain fully functional and are primed towards a TH1 response, in addition we provide evidence that catecholamine mediated inhibition of CTLA-4 expression and serum HMGB1 release are possible mediators in stroke induced activation of T cells.

Conflict of interest statement

Competing Interests: The authors have declared that no competing interests exist.

Figures

Figure 1. Th1-type cytokines are downregulated in…
Figure 1. Th1-type cytokines are downregulated in stroke patients.
IL-2, IL-12p70, IFN-γ and TNF-α serum cytokine levels are significantly reduced in sera of stroke patients (grey bars) compared to healthy controls (white bars) in the first two weeks following stroke. * p(IL-2, IL-12p70, TNF-α) = 16; 20; 20; 21; 13; n(IFN-γ) = 16; 20; 20; 19; 12 (control; day 0; day 1; day 7; day14).
Figure 2. HMGB1 is upregulated in serum…
Figure 2. HMGB1 is upregulated in serum of stroke patients.
HMGB1 is upregulated in serum of stroke patients (grey bars) compared to healthy controls (white bars) in the first two weeks following stroke while its decoy receptors sRAGE and esRAGE remain indistinguishable within 48h following stroke. ** p(HMGB1) = 16; 87; 84; 46; 24; n(sRAGE,) 12; 32; 29, esRAGE) = 10; 37; 34, (control; day 0; day 1; day 7; day14).
Figure 3. CD4 + T-cells in the…
Figure 3. CD4+ T-cells in the peripheral blood of stroke patients are activated.
Comparison of expression of activation markers on lymphocytes between healthy controls (white bars) and stroke patients (grey bars) in the first two weeks following stroke reveal that CD4+ T-cells in the peripheral blood are activated. * p<0.05; ** p<0.01; *** p<0.001. Medians and interquartile ranges. n(CD4 + CD25 + , CD25 + %CD4 + ) = 14; 33; 32; 24; 14; n(CD4 + HLA-DR + ) = 12; 64; 66; 42; 22; n(HLA-DR + %CD4 + ) = 12; 63; 66; 43; 22; (control; day 0; day 1; day 7; day14).
Figure 4. Lack of CTLA-4 expression and…
Figure 4. Lack of CTLA-4 expression and inhibition by catecholamines.
A) The percentage of PBMC expressing CTLA-4 is undistinguishable between healthy controls (white bars) and stroke patients (grey bars) in the first two weeks following stroke. Medians and interquartile ranges. n = 9; 31; 33; 27; 20; (control; day 0; day 1; day 7; day14). B) Percentage of CTLA-4 expression on PBMC decreases after in vitro treatment with PHA+epinephrine or PHA+norepinephrine compared to treatment with PHA alone. *** p<0.001. Medians and interquartile ranges. .n = 13; 6; 9; (PHA; PHA+Epi; PHA+Norepi).
Figure 5. Surviving T lymphocytes are functional.
Figure 5. Surviving T lymphocytes are functional.
A) The proliferation expressed as stimulation index of T cells after ex vivo stimulation with PHA reveals normal proliferative responses of stroke patients (grey bars) lymphocytes compared to healthy controls (white bars) in the first week following stroke. Medians and interquartile ranges. n = 7; 36; 35; 32; (control; day 0; day 1; day 7). B–E) Exvivo stimulation of T cells with PHA leads to increased proinflammatory cytokine release in supernatants of stroke patients cells (grey bars) compared to healthy controls (white bars) within the first week following stroke. * p<0.05; ** p<0.01. Medians and interquartile ranges. n(IL-1β, IL-6, TNFα, TNF-β) = 10; 37; 36; 28; (control; day 0; day 1; day 7).
Figure 6. Relative proportion of activated T…
Figure 6. Relative proportion of activated T cells were undistinguishable between patient subgroups.
Activation markers on lymphocytes from stroke patients without subsequent infection (white bars) remained undistinguishable from those of stroke patients with subsequent infection (grey bars) within the relative proportion of lymphocytes in the first two weeks following stroke. * p(CD4 + CD25 + , CD25 + %CD4 + ) = 5; 7; 7; 7; 7; 6; 6; 3; n(CD4 + HLA-DR + , HLA-DR + %CD4 + ) = 14; 12; 16; 11; 12; 9; 8; 5 (no infection day 0; subsequent infection day 0; no infection day 1; subsequent infection day 1; no infection day 7; subsequent infection day 7; no infection day 14; subsequent infection day 14).
Figure 7. Increased metanephrine and cortisol serum…
Figure 7. Increased metanephrine and cortisol serum levels in patients with subsequent infection.
Metanephrine and cortisol serum levels are increased in serum of stroke patients with subsequent infection (grey bars) compared to stroke patients without subsequent infection (white bars) within 24h following stroke. * p(Metanephrine) = 8; 8; 8; 7; n(Cortisol) = 10; 8; 9; 9; (no infection day 0; subsequent infection day 0; no infection day 1; subsequent infection day 1).

References

    1. Donnan GA, Fisher M, Macleod M, Davis SM. Stroke. The Lancet. 2008;371:1612–1623.
    1. Emsley HC, Hopkins SJ. Acute ischaemic stroke and infection: recent and emerging concepts. Lancet Neurol. 2008;7:341–353.
    1. Chamorro A, Urra X, Planas AM. Infection after acute ischemic stroke: a manifestation of brain-induced immunodepression. Stroke. 2007;38:1097–1103.
    1. Prass K, Meisel C, Hoflich C, Braun J, Halle E, et al. Stroke-induced immunodeficiency promotes spontaneous bacterial infections and is mediated by sympathetic activation reversal by poststroke T helper cell type 1-like immunostimulation. J Exp Med. 2003;198:725–736.
    1. Klehmet J, Harms H, Richter M, Prass K, Volk HD, et al. Stroke-induced immunodepression and post-stroke infections: lessons from the preventive antibacterial therapy in stroke trial. Neuroscience. 2009;158:1184–1193.
    1. Urra X, Cervera A, Obach V, Climent N, Planas AM, et al. Monocytes are major players in the prognosis and risk of infection after acute stroke. Stroke. 2009;40:1262–1268.
    1. Vogelgesang A, Grunwald U, Langner S, Jack R, Broker BM, et al. Analysis of lymphocyte subsets in patients with stroke and their influence on infection after stroke. Stroke. 2008;39:237–241.
    1. Qiu J, Nishimura M, Wang Y, Sims JR, Qiu S, et al. Early release of HMGB-1 from neurons after the onset of brain ischemia. J Cereb Blood Flow Metab. 2008;28:927–938.
    1. Erlandsson Harris H, Andersson U. Mini-review: The nuclear protein HMGB1 as a proinflammatory mediator. Eur J Immunol. 2004;34:1503–1512.
    1. Scaffidi P, Misteli T, Bianchi ME. Release of chromatin protein HMGB1 by necrotic cells triggers inflammation. Nature. 2002;418:191–195.
    1. Sundberg E, Fasth AE, Palmblad K, Harris HE, Andersson U. High mobility group box chromosomal protein 1 acts as a proliferation signal for activated T lymphocytes. Immunobiology. 2009;214:303–309.
    1. Dumitriu IE, Baruah P, Valentinis B, Voll RE, Herrmann M, et al. Release of high mobility group box 1 by dendritic cells controls T cell activation via the receptor for advanced glycation end products. J Immunol. 2005;174:7506–7515.
    1. Goldstein RS, Gallowitsch-Puerta M, Yang L, Rosas-Ballina M, Huston JM, et al. Elevated high-mobility group box 1 levels in patients with cerebral and myocardial ischemia. Shock. 2006;25:571–574.
    1. Faraco G, Fossati S, Bianchi ME, Patrone M, Pedrazzi M, et al. High mobility group box 1 protein is released by neural cells upon different stresses and worsens ischemic neurodegeneration in vitro and in vivo. J Neurochem. 2007;103:590–603.
    1. Kim JB, Sig Choi J, Yu YM, Nam K, Piao CS, et al. HMGB1, a novel cytokine-like mediator linking acute neuronal death and delayed neuroinflammation in the postischemic brain. J Neurosci. 2006;26:6413–6421.
    1. Bierhaus A, Humpert PM, Morcos M, Wendt T, Chavakis T, et al. Understanding RAGE, the receptor for advanced glycation end products. J Mol Med. 2005;83:876–886.
    1. Hug A, Dalpke A, Wieczorek N, Giese T, Lorenz A, et al. Infarct Volume is a Major Determiner of Post-Stroke Immune Cell Function and Susceptibility to Infection. Stroke. 2009;40:3226–3232.
    1. Zhang DP, Yan FL, Xu HQ, Zhu YX, Yin Y, et al. A decrease of human leucocyte antigen-DR expression on monocytes in peripheral blood predicts stroke-associated infection in critically-ill patients with acute stroke. Eur J Neurol. 2009;16:498–505.
    1. Urra X, Cervera A, Villamor N, Planas AM, Chamorro A. Harms and benefits of lymphocyte subpopulations in patients with acute stroke. Neuroscience. 2009;158:1174–1183.
    1. Haeusler KG, Schmidt WU, Fohring F, Meisel C, Helms T, et al. Cellular immunodepression preceding infectious complications after acute ischemic stroke in humans. Cerebrovasc Dis. 2008;25:50–58.
    1. Petersen CM, Christensen EI, Andresen BS, Møller BK. Internalization, lysosomal degradation and new synthesis of surface membrane CD4 in phorbol ester-activated T-lymphocytes and U-937 cells. Experimental Cell Research. 1992;201:160–173.
    1. Freeman GJ, Lombard DB, Gimmi CD, Brod SA, Lee K, et al. CTLA-4 and CD28 mRNAs are coexpressed in most activated T cells after activation: Expression of CTLA-4 and CD28 messenger RNA does not correlate with the pattern of lymphokine production. J Immunol. 1992;149:3795–3801.
    1. Zheng Y, Manzotti CN, Burke F, Dussably L, Qureshi O, et al. Acquisition of Suppressive Function by Activated Human CD4+CD25− T Cells Is Associated with the Expression of CTLA-4 Not FoxP3. 2008:1683–1691.
    1. Chamorro Á, Amaro S, Vargas M, Obach V, Cervera Á, et al. Catecholamines, infection, and death in acute ischemic stroke. Journal of the Neurological Sciences. 2007;252:29–35.
    1. Katzan IL, Cebul RD, Husak SH, Dawson NV, Baker DW. The effect of pneumonia on mortality among patients hospitalized for acute stroke. Neurology. 2003;60:620–625.
    1. Vermeij FH, Scholte op Reimer WJ, de Man P, van Oostenbrugge RJ, Franke CL, et al. Stroke-associated infection is an independent risk factor for poor outcome after acute ischemic stroke: data from the Netherlands Stroke Survey. Cerebrovasc Dis. 2009;27:465–471.

Source: PubMed

3
Sottoscrivi