Stroke-induced immunodeficiency promotes spontaneous bacterial infections and is mediated by sympathetic activation reversal by poststroke T helper cell type 1-like immunostimulation

Konstantin Prass, Christian Meisel, Conny Höflich, Johann Braun, Elke Halle, Tilo Wolf, Karsten Ruscher, Ilya V Victorov, Josef Priller, Ulrich Dirnagl, Hans-Dieter Volk, Andreas Meisel, Konstantin Prass, Christian Meisel, Conny Höflich, Johann Braun, Elke Halle, Tilo Wolf, Karsten Ruscher, Ilya V Victorov, Josef Priller, Ulrich Dirnagl, Hans-Dieter Volk, Andreas Meisel

Abstract

Infections are a leading cause of death in stroke patients. In a mouse model of focal cerebral ischemia, we tested the hypothesis that a stroke-induced immunodeficiency increases the susceptibility to bacterial infections. 3 d after ischemia, all animals developed spontaneous septicemia and pneumonia. Stroke induced an extensive apoptotic loss of lymphocytes and a shift from T helper cell (Th)1 to Th2 cytokine production. Adoptive transfer of T and natural killer cells from wild-type mice, but not from interferon (IFN)-gamma-deficient mice, or administration of IFN-gamma at day 1 after stroke greatly decreased the bacterial burden. Importantly, the defective IFN-gamma response and the occurrence of bacterial infections were prevented by blocking the sympathetic nervous system but not the hypothalamo-pituitary-adrenal axis. Furthermore, administration of the beta-adrenoreceptor blocker propranolol drastically reduced mortality after stroke. These data suggest that a catecholamine-mediated defect in early lymphocyte activation is the key factor in the impaired antibacterial immune response after stroke.

Figures

Figure 1.
Figure 1.
Stroke induces bacteremia and pneumonia. (a) Lungs and blood samples from sham (n = 10) and MCAO-treated mice (n = 4–14 per group) were collected for bacteriological analysis at the indicated time points after surgery. Data are given in CFU/ml (log 10) blood or lung tissue homogenate. (b) In a different set of experiments, lungs from sham-operated (n = 4) and MCAO mice (n = 4) were collected after 72 h for histological examination. A representative 12-μm section of HE-stained lung from MCAO but not from sham animals revealed signs (thickening of alveolar walls and neutrophilic infiltrates) of E. coli pneumonia. ×160.
Figure 2.
Figure 2.
Stroke induces long-lasting lymphopenia and impaired cytokine expression. Spleen, thymus, and peripheral blood samples of untreated SV129/J mice (control) and sham or MCAO mice were collected at different time points (sham, 12 h; MCAO, as indicated) after surgery. Leukocyte counts in blood (a–c), spleen (d–f), and thymus (g) single cell suspensions were determined as described in Materials and Methods. The lymphocyte subsets were determined by flow cytometry and their absolute numbers were calculated. (h and i) Aliquots of blood samples were stimulated ex vivo with either LPS for analysis of monocytic TNF-α expression or Con A for analysis of IFN-γ and IL-4 synthesis, as described in Materials and Methods. Cytokines were determined in supernatants by ELISA. The Con A–induced lymphokine expression is given as the ratio of IFN-γ and IL-4 production calculated for each individual. Data are shown as mean ± SD. *, results differed from the control group; *, P n = 3–11 per group.
Figure 3.
Figure 3.
Stroke induces increased apoptosis in lymphoid organs. Spleen and thymus from untreated SV129/J mice (control) and sham or MCAO mice were collected 12 h after surgery. Splenocytes (a) and thymocytes (b) were isolated and investigated for apoptosis by annexin V labeling and flow cytometry. Lymphocyte subpopulations were determined by staining with mAbs against cell-type–specific surface markers. Data are shown as mean ± SD. *, results differed from the control group; *, P n = 8 per group. (c) Thymic tissue sections from sham- and MCAO-treated mice were stained with HE (top) or examined by the fluorescent TUNEL method (bottom). Thymi from MCAO mice showed many apoptotic cells with typical features of nuclear condensation and fragmentation (HE; ×400). Note also the high number of TUNEL+ cells (bright red nuclei) in the thymus cortex from MCAO animals as compared with sham mice. ×100.
Figure 4.
Figure 4.
Effects of β-adrenoreceptor and glucocorticoid receptor blockade on stroke-induced immunodepression and bacterial infection. SV129/J mice underwent sham or MCAO surgery (n = 9–12 per group). MCAO mice received the β-adrenoreceptor antagonist propranolol, the glucocorticoid receptor inhibitor RU486, or only diluent before or after surgery, as described in Materials and Methods. Spleens and blood samples were collected 12 h after surgery. (a) Splenocytes were isolated and investigated for apoptosis by annexin V labeling, staining with mAbs against cell-type–specific surface markers, and flow cytometry. (b) Total leukocyte counts in blood samples were determined as described in Materials and Methods. The percentages of different lymphocyte classes were determined by flow cytometry using mAbs against cell-type–specific surface markers and their absolute numbers calculated. (c) For determination of cytokine production, blood samples were processed and cytokine expression was analyzed as described in Fig. 2. Data are shown as mean ± SD. #, results differed from the sham-operated group; #, P < 0.05; ##, P < 0.01; §, results differed from the diluent-treated MCAO group; §, P < 0.05; §§, P < 0.01; §§§, P < 0.001. Mann-Whitney U test, n = 9–12 per group. (d) In a different set of experiments, lungs and blood samples from sham and inhibitor- or diluent-treated MCAO mice were collected for bacteriological analysis 72 h after surgery. Data are shown as box plots in CFU/ml (log 10) blood or lung tissue homogenate.
Figure 5.
Figure 5.
Prevention of bacterial infections and restoration of defective IFN-γ response by SNS inhibitors is dose and time dependent. (a) MCAO mice were treated with diluent or different doses of propranolol (3 × 1–3 × 30 mg/kg BW) commencing immediately after MCAO or 24 h (3 × 30 mg/kg BW) after MCAO, or with 6-OHDA before MCAO as described in Materials and Methods and as indicated (n = 4 in each group). Lungs and blood samples were collected for bacteriological analysis 72 h after MCAO. Data are shown as box plots in CFU/ml (log 10) blood or lung tissue homogenate. (b) In a different experiment, spleens of untreated mice (control), sham-, or MCAO-operated mice, which received diluent, propranolol, or 6-OHDA as described above, were collected 12 or 36 h (delayed propranolol treatment) after surgery. 106/ml spleen cells were stimulated ex vivo with Con A for 24 h and IFN-γ production was analyzed by ELISA, as described in Materials and Methods. Data are shown as mean ± SD. #, results differed from the sham-operated group; #, P < 0.05; §, results differed from the MCAO animals; §, P < 0.05. Mann-Whitney U test, n = 3–6 per group.
Figure 6.
Figure 6.
Blocking the SNS improves survival after experimental stroke. Mice underwent MCAO and received either diluent or 10 mg/kg BW immediately before, 4, and 8 h after MCAO (n = 15 per group). Differences in survival between propranolol-treated versus diluent-treated mice were tested for significance on day 11 after MCAO and are indicated by asterisks. *, P < 0.05. Fisher's exact test.
Figure 7.
Figure 7.
Adoptive lymphocyte transfer after experimental stroke prevents infection. (a) Adoptive lymphocyte transfer was performed 24 h after MCAO using either unseparated (total) or T, B, and NK cell, or T cell–, B cell–, or NK cell–depleted spleen cell suspensions from wild-type SV129/J mice. *, results differed from unseparated splenocyte transfer group. Kruskal-Wallis analysis of variance (ANOVA) after pairwise comparison with Dunn's method test, n = 4 per group. (b) In a different set of experiments, splenocytes from αβ T cell–deficient (αβ-TCR−/−), γδ T cell–deficient (γδ-TCR−/−), or wild-type B6 mice (WT) were transferred into B6 mice 24 h after MCAO. 48 h later, blood and lung samples were analyzed for bacterial counts. Data are shown as box plots in CFU/ml (log 10) blood or lung tissue homogenate. *, results differed from WT splenocyte transfer group. Kruskal-Wallis analysis of variance (ANOVA) after comparison of pairs with Dunn's method test, n = 8 per group.
Figure 8.
Figure 8.
IFN-γ is essential in preventing the bacterial infections. (a) Adoptive lymphocyte transfer experiments 24 h after MCAO revealed an impaired ability of splenocytes from IFN-γ–deficient mice (IFN-γ−/−) to prevent bacterial infection in blood and lung when compared with splenocytes from wild-type B6 mice (n = 8 per group). (b) Administration of 2 μg recombinant IFN-γ 24 h, but not 48 h, after cerebral ischemia reduced the bacterial burden in peripheral blood and lungs as measured 72 h after MCAO (n = 4 per group). For data representation see Fig. 5.

References

    1. Davenport, R.J., M.S. Dennis, I. Wellwood, and C.P. Warlow. 1996. Complications after acute stroke. Stroke. 27:415–420.
    1. Johnston, K.C., J.Y. Li, P.D. Lyden, S.K. Hanson, T.E. Feasby, R.J. Adams, R.E. Faught, Jr., and E.C. Haley, Jr. 1998. Medical and neurological complications of ischemic stroke: experience from the RANTTAS trial. RANTTAS investigators. Stroke. 29:447–453.
    1. Georgilis, K., A. Plomaritoglou, U. Dafni, Y. Bassiakos, and K. Vemmos. 1999. Aetiology of fever in patients with acute stroke. J. Intern. Med. 246:203–209.
    1. Grau, A.J., F. Buggle, P. Schnitzler, M. Spiel, C. Lichy, and W. Hacke. 1999. Fever and infection early after ischemic stroke. J. Neurol. Sci. 171:115–120.
    1. Langhorne, P., D.J. Stott, L. Robertson, J. MacDonald, L. Jones, C. McAlpine, F. Dick, G.S. Taylor, and G. Murray. 2000. Medical complications after stroke: a multicenter study. Stroke. 31:1223–1229.
    1. Kalra, L., A. Evans, I. Perez, M. Knapp, N. Donaldson, and C.G. Swift. 2000. Alternative strategies for stroke care: a prospective randomised controlled study of stroke unit, stroke team, and domiciliary management of stroke. Lancet. 356:894–899.
    1. Henon, H., O. Godefroy, D. Leys, F. Mounier-Vehier, C. Lucas, P. Rondepierre, A. Duhamel, and J.P. Pruvo. 1995. Early predictors of death and disability after acute cerebral ischemic event. Stroke. 26:392–398.
    1. Kalra, L., G. Yu, K. Wilson, and P. Roots. 1995. Medical complications during stroke rehabilitation. Stroke. 26:990–994.
    1. Viitanen, M., B. Winblad, and K. Asplund. 1987. Autopsy-verified causes of death after stroke. Acta Med. Scand. 222:401–408.
    1. Ronning, O.M., and B. Guldvog. 1998. Stroke unit versus general medical wards, II: neurological deficits and activities of daily living: a quasi-randomized controlled trial. Stroke. 29:586–590.
    1. Livingston, D.H., S.H. Appel, S.R. Wellhausen, G. Sonnenfeld, and H.C. Polk, Jr. 1988. Depressed interferon gamma production and monocyte HLA-DR expression after severe injury. Arch. Surg. 123:1309–1312.
    1. Docke, W.D., F. Randow, U. Syrbe, D. Krausch, K. Asadullah, P. Reinke, H.D. Volk, and W. Kox. 1997. Monocyte deactivation in septic patients: restoration by IFN-gamma treatment. Nat. Med. 3:678–681.
    1. Woiciechowsky, C., K. Asadullah, D. Nestler, B. Eberhardt, C. Platzer, B. Schoning, F. Glockner, W.R. Lanksch, H.D. Volk, and W.D. Docke. 1998. Sympathetic activation triggers systemic interleukin-10 release in immunodepression induced by brain injury. Nat. Med. 4: 808–813.
    1. Rodriguez, J.L., K.J. Gibbons, L.G. Bitzer, R.E. Dechert, S.M. Steinberg, and L.M. Flint. 1991. Pneumonia: incidence, risk factors, and outcome in injured patients. J. Trauma. 31:907–912.
    1. Hsieh, A.H., M.J. Bishop, P.S. Kubilis, D.W. Newell, and D.J. Pierson. 1992. Pneumonia following closed head injury. Am. Rev. Respir. Dis. 146:290–294.
    1. Howard, R.J., and R.L. Simmons. 1974. Acquired immunologic deficiencies after trauma and surgical procedures. Surg. Gynecol. Obstet. 139:771–782.
    1. Hara, H., P.L. Huang, N. Panahian, M.C. Fishman, and M.A. Moskowitz. 1996. Reduced brain edema and infarction volume in mice lacking the neuronal isoform of nitric oxide synthase after transient MCA occlusion. J. Cereb. Blood Flow Metab. 16:605–611.
    1. Prass, K., K. Ruscher, M. Karsch, N. Isaev, D. Megow, J. Priller, A. Scharff, U. Dirnagl, and A. Meisel. 2002. Desferrioxamine induces delayed tolerance against cerebral ischemia in vivo and in vitro. J. Cereb. Blood Flow Metab. 22:520–525.
    1. Wang, Q., P. Teder, N.P. Judd, P.W. Noble, and C.M. Doerschuk. 2002. CD44 deficiency leads to enhanced neutrophil migration and lung injury in Escherichia coli pneumonia in mice. Am. J. Pathol. 161:2219–2228.
    1. Matute-Bello, G., C.W. Frevert, W.C. Liles, M. Nakamura, J.T. Ruzinski, K. Ballman, V.A. Wong, C. Vathanaprida, and T.R. Martin. 2001. Fas/Fas ligand system mediates epithelial injury, but not pulmonary host defenses, in response to inhaled bacteria. Infect. Immun. 69:5768–5776.
    1. Asadullah, K., C. Woiciechowsky, W.D. Docke, C. Liebenthal, H. Wauer, W. Kox, H.D. Volk, S. Vogel, and R. von Baehr. 1995. Immunodepression following neurosurgical procedures. Crit. Care Med. 23:1976–1983.
    1. Woiciechowsky, C., B. Schoning, W.R. Lanksch, H.D. Volk, and W.D. Docke. 1999. Mechanisms of brain-mediated systemic anti-inflammatory syndrome causing immunodepression. J. Mol. Med. 77:769–780.
    1. Cao, L., N.M. Filipov, and D.A. Lawrence. 2002. Sympathetic nervous system plays a major role in acute cold/restraint stress inhibition of host resistance to Listeria monocytogenes. J. Neuroimmunol. 125:94–102.
    1. Bernik, T.R., S.G. Friedman, M. Ochani, R. DiRaimo, L. Ulloa, H. Yang, S. Sudan, C.J. Czura, S.M. Ivanova, and K.J. Tracey. 2002. Pharmacological stimulation of the cholinergic antiinflammatory pathway. J. Exp. Med. 195:781–788.
    1. Tracey, K.J. 2002. The inflammatory reflex. Nature. 420:853–859.
    1. Feuerstein, G.Z., T. Liu, and F.C. Barone. 1994. Cytokines, inflammation, and brain injury: role of tumor necrosis factor-alpha. Cerebrovasc. Brain Metab. Rev. 6:341–360.
    1. Tarkowski, E., L. Rosengren, C. Blomstrand, C. Wikkelso, C. Jensen, S. Ekholm, and A. Tarkowski. 1997. Intrathecal release of pro- and anti-inflammatory cytokines during stroke. Clin. Exp. Immunol. 110:492–499.
    1. del Zoppo, G., I. Ginis, J.M. Hallenbeck, C. Iadecola, X. Wang, and G.Z. Feuerstein. 2000. Inflammation and stroke: putative role for cytokines, adhesion molecules and iNOS in brain response to ischemia. Brain Pathol. 10:95–112.
    1. Fassbender, K., R. Schmidt, R. Mossner, M. Daffertshofer, and M. Hennerici. 1994. Pattern of activation of the hypothalamic-pituitary-adrenal axis in acute stroke. Relation to acute confusional state, extent of brain damage, and clinical outcome. Stroke. 25:1105–1108.
    1. Cruse, J.M., R.E. Lewis, Jr., G.R. Bishop, W.F. Kliesch, E. Gaitan, and R. Britt. 1993. Decreased immune reactivity and neuroendocrine alterations related to chronic stress in spinal cord injury and stroke patients. Pathobiology. 61:183–192.
    1. Sander, D., K. Winbeck, J. Klingelhofer, T. Etgen, and B. Conrad. 2001. Prognostic relevance of pathological sympathetic activation after acute thromboembolic stroke. Neurology. 57:833–838.
    1. Myers, M.G., J.W. Norris, V.C. Hachniski, and M.J. Sole. 1981. Plasma norepinephrine in stroke. Stroke. 12:200–204.
    1. Woiciechowsky, C., B. Schoning, N. Daberkow, K. Asche, G. Stoltenburg, W.R. Lanksch, and H.D. Volk. 1999. Brain-IL-1beta induces local inflammation but systemic anti-inflammatory response through stimulation of both hypothalamic-pituitary-adrenal axis and sympathetic nervous system. Brain Res. 816:563–571.
    1. Elenkov, I.J., R.L. Wilder, G.P. Chrousos, and E.S. Vizi. 2000. The sympathetic nerve-an integrative interface between two supersystems: the brain and the immune system. Pharmacol. Rev. 52:595–638.
    1. Abbas, A.K., A.H. Lichtman, and J.S. Pober. 2000. Cellular and Molecular Immunology. Fourth edition. W.B. Saunders, Philadelphia. 235 pp.
    1. O'Sullivan, S.T., J.A. Lederer, A.F. Horgan, D.H. Chin, J.A. Mannick, and M.L. Rodrick. 1995. Major injury leads to predominance of the T helper-2 lymphocyte phenotype and diminished interleukin-12 production associated with decreased resistance to infection. Ann. Surg. 222:482–490.
    1. Decker, D., M. Schondorf, F. Bidlingmaier, A. Hirner, and A.A. von Ruecker. 1996. Surgical stress induces a shift in the type-1/type-2 T-helper cell balance, suggesting down-regulation of cell-mediated and up-regulation of antibody-mediated immunity commensurate to the trauma. Surgery. 119:316–325.
    1. Mack, V.E., M.D. McCarter, H.A. Naama, S.E. Calvano, and J.M. Daly. 1996. Dominance of T-helper 2-type cytokines after severe injury. Arch. Surg. 131:1303–1308.
    1. Zedler, S., R.C. Bone, A.E. Baue, G.H. von Donnersmarck, and E. Faist. 1999. T-cell reactivity and its predictive role in immunosuppression after burns. Crit. Care Med. 27:66–72.
    1. Hotchkiss, R.S., K.W. Tinsley, P.E. Swanson, K.C. Chang, J.P. Cobb, T.G. Buchman, S.J. Korsmeyer, and I.E. Karl. 1999. Prevention of lymphocyte cell death in sepsis improves survival in mice. Proc. Natl. Acad. Sci. USA. 96:14541–14546.
    1. Hotchkiss, R.S., K.C. Chang, P.E. Swanson, K.W. Tinsley, J.J. Hui, P. Klender, S. Xanthoudakis, S. Roy, C. Black, E. Grimm, et al. 2000. Caspase inhibitors improve survival in sepsis: a critical role of the lymphocyte. Nat. Immunol. 1:496–501.
    1. Rubins, J.B., and C. Pomeroy. 1997. Role of gamma interferon in the pathogenesis of bacteremic pneumococcal pneumonia. Infect. Immun. 65:2975–2977.
    1. King, D.P., D.M. Hyde, K.A. Jackson, D.M. Novosad, T.N. Ellis, L. Putney, M.Y. Stovall, L.S. Van Winkle, B.L. Beaman, and D.A. Ferrick. 1999. Cutting edge: protective response to pulmonary injury requires gamma delta T lymphocytes. J. Immunol. 162:5033–5036.
    1. Moore, T.A., B.B. Moore, M.W. Newstead, and T.J. Standiford. 2000. Gamma delta-T cells are critical for survival and early proinflammatory cytokine gene expression during murine Klebsiella pneumonia. J. Immunol. 165:2643–2650.
    1. Gendron, A., J. Teitelbaum, C. Cossette, S. Nuara, M. Dumont, D. Geadah, P. du Souich, and E. Kouassi. 2002. Temporal effects of left versus right middle cerebral artery occlusion on spleen lymphocyte subsets and mitogenic response in Wistar rats. Brain Res. 955:85–97.
    1. Hatcher, J.P., D. Virley, S.J. Hadingham, J. Roberts, A.J. Hunter, and A.A. Parsons. 2002. The behavioural effect of middle cerebral artery occlusion on apolipoprotein-E deficient mice. Behav. Brain Res. 131:139–149.
    1. Wang, R.Y., P.S. Wang, and Y.R. Yang. 2003. Effect of age in rats following middle cerebral artery occlusion. Gerontology. 49:27–32.
    1. Neumann-Haefelin, T., A. Kastrup, A. de Crespigny, M.A. Yenari, T. Ringer, G.H. Sun, and M.E. Moseley. 2000. Serial MRI after transient focal cerebral ischemia in rats: dynamics of tissue injury, blood-brain barrier damage, and edema formation. Stroke. 31:1965–1972.
    1. Aspey, B.S., F.L. Taylor, M. Terruli, and M.J. Harrison. 2000. Temporary middle cerebral artery occlusion in the rat: consistent protocol for a model of stroke and reperfusion. Neuropathol. Appl. Neurobiol. 26:232–242.
    1. Huang, J., D.B. Agus, C.J. Winfree, S. Kiss, W.J. Mack, R.A. McTaggart, T.F. Choudhri, L.J. Kim, J. Mocco, D.J. Pinsky, et al. 2001. Dehydroascorbic acid, a blood-brain barrier transportable form of vitamin C, mediates potent cerebroprotection in experimental stroke. Proc. Natl. Acad. Sci. USA. 98:11720–11724.
    1. Maier, C.M., G.H. Sun, D. Kunis, M.A. Yenari, and G.K. Steinberg. 2001. Delayed induction and long-term effects of mild hypothermia in a focal model of transient cerebral ischemia: neurological outcome and infarct size. J. Neurosurg. 94:90–96.
    1. Kondo, T., A.G. Reaume, T.T. Huang, E. Carlson, K. Murakami, S.F. Chen, E.K. Hoffman, R.W. Scott, C.J. Epstein, and P.H. Chan. 1997. Reduction of CuZn-superoxide dismutase activity exacerbates neuronal cell injury and edema formation after transient focal cerebral ischemia. J. Neurosci. 17:4180–4189.
    1. Schabitz, W.R., F. Li, K. Irie, B.W. Sandage, Jr., K.W. Locke, and M. Fisher. 1999. Synergistic effects of a combination of low-dose basic fibroblast growth factor and citicoline after temporary experimental focal ischemia. Stroke. 30:427–431.
    1. Virley, D., J.S. Beech, S.C. Smart, S.C. Williams, H. Hodges, and A.J. Hunter. 2000. A temporal MRI assessment of neuropathology after transient middle cerebral artery occlusion in the rat: correlations with behavior. J. Cereb. Blood Flow Metab. 20:563–582.
    1. Katzan, I.L., R.D. Cebul, S.H. Husak, N.V. Dawson, and D.W. Baker. 2003. The effect of pneumonia on mortality among patients hospitalized for acute stroke. Neurology. 60:620–625.
    1. Hilker, R., C. Poetter, N. Findeisen, J. Sobesky, A. Jacobs, M. Neveling, and W.D. Heiss. 2003. Nosocomial pneumonia after acute stroke: implications for neurological intensive care medicine. Stroke. 34:975–981.
    1. Perry, L., and C.P. Love. 2001. Screening for dysphagia and aspiration in acute stroke: a systematic review. Dysphagia. 16:7–18.
    1. Finegold, S.M. 1991. Aspiration pneumonia. Rev. Infect. Dis. 13:S737–S742.
    1. Riquelme, R., A. Torres, M. El-Ebiary, J.P. de la Bellacasa, R. Estruch, J. Mensa, J. Fernandez-Sola, C. Hernandez, and R. Rodriguez-Roisin. 1996. Community-acquired pneumonia in the elderly: a multivariate analysis of risk and prognostic factors. Am. J. Respir. Crit. Care Med. 154:1450–1455.
    1. Marik, P.E. 2001. Aspiration pneumonitis and aspiration pneumonia. N. Engl. J. Med. 344:665–671.
    1. Evans, A., I. Perez, F. Harraf, A. Melbourn, J. Steadman, N. Donaldson, and L. Kalra. 2001. Can differences in management processes explain different outcomes between stroke unit and stroke-team care? Lancet. 358:1586–1592.
    1. Hajat, C., S. Hajat, and P. Sharma. 2000. Effects of poststroke pyrexia on stroke outcome: a meta-analysis of studies in patients. Stroke. 31:410–414.

Source: PubMed

3
Předplatit