Transplanted neural stem cells modulate regulatory T, γδ T cells and corresponding cytokines after intracerebral hemorrhage in rats

Lu Gao, Qin Lu, Li-Jie Huang, Lin-Hui Ruan, Jian-Jing Yang, Wei-Long Huang, Wei-Shan ZhuGe, Yong-Liang Zhang, Biao Fu, Kun-Lin Jin, Qi-Chuan ZhuGe, Lu Gao, Qin Lu, Li-Jie Huang, Lin-Hui Ruan, Jian-Jing Yang, Wei-Long Huang, Wei-Shan ZhuGe, Yong-Liang Zhang, Biao Fu, Kun-Lin Jin, Qi-Chuan ZhuGe

Abstract

The immune system, particularly T lymphocytes and cytokines, has been implicated in the progression of brain injury after intracerebral hemorrhage (ICH). Although studies have shown that transplanted neural stem cells (NSCs) protect the central nervous system (CNS) from inflammatory damage, their effects on subpopulations of T lymphocytes and their corresponding cytokines are largely unexplored. Here, rats were subjected to ICH and NSCs were intracerebrally injected at 3 h after ICH. The profiles of subpopulations of T cells in the brain and peripheral blood were analyzed by flow cytometry. We found that regulatory T (Treg) cells in the brain and peripheral blood were increased, but γδT cells (gamma delta T cells) were decreased, along with increased anti-inflammatory cytokines (IL-4, IL-10 and TGF-β) and decreased pro-inflammatory cytokines (IL-6, and IFN-γ), compared to the vehicle-treated control. Our data suggest that transplanted NSCs protect brain injury after ICH via modulation of Treg and γδT cell infiltration and anti- and pro-inflammatory cytokine release.

Figures

Figure 1.
Figure 1.
Total numbers of T lymphocyte cells in the brain and peripheral blood. Total numbers of T lymphocytes in the brain were significantly increased in intracerebral hemorrhage (ICH), vehicle-treated (ICH + V) and neural stem cells (NSCs)-treated groups (ICH + NSCs), compared to sham-operated group (Sham) (A); there was no difference of total numbers of T lymphocytes of each group in peripheral blood in ICH, vehicle-treated (ICH + V) and NSCs-treated groups (ICH + NSCs), compared to sham-operated group (Sham) (B), (n = 8 in each group). *p < 0.05 versus sham ICH.
Figure 2.
Figure 2.
Increased absolute numbers of γδT and Treg cells in the brain and peripheral blood after ICH. The absolute numbers of γδT and Treg cells were increased significantly in the brain (A) and peripheral blood; (B) after ICH. (n = 8 in each group) *p < 0.05 versus sham ICH.
Figure 3.
Figure 3.
Behavioral functional test before and after ICH. Transplanted NSCs improve functional recovery after ICH at day 3, 7 and 14. (n = 6 in each group) *p < 0.05 versus ICH and ICH + V.
Figure 4.
Figure 4.
Transplanted NSCs altered γδT and Treg cells in the brain and peripheral blood. Transplanted NSCs reduced the absolute numbers of γδT cells in the brain and peripheral blood (AC); The percentage and the absolute numbers of Treg cells were significantly increased in the brain by transplantation of NSCs (D,E); An increased absolute Treg cell numbers in the peripheral blood after transplanted NSCs (F). (n = 8 in each group) *p < 0.05 versus ICH-vehicle.
Figure 5.
Figure 5.
Expressions of inflammatory cytokine in the cerebral and peripheral blood after transplantation of NSCs. ELASA (A,B) revealed the upregulations of IL-6 and IFN-γ in the haemorrhagic brains and peripheral blood in the groups transplanted witn NSCs, compared to the normal samples. In the peripheral blood, TGF-β and IL-10 were upregulated after ICH followed NSC transplantation. However, IL-4 was unchanged both in the brain and peripheral blood after ICH. In brain and peripheral blood samples, the groups treated with NSCs showed decreases of IL-6 and IFN-γ and increases of IL-4, IL-10 and TGF-β levels, compared with the ICH group. (n = 4 in each group) *p < 0.05 versus normal; **p < 0.05 versus ICH.

References

    1. Broderick J.P., Adams H.P., Barsan W., Feinberg W., Feldmann E., Grotta J., Kase C., Krieger D., Mayberg M., Tilley B., et al. Guidelines for the management of spontaneous intracerebral hemorrhage: A statement for Healthcare Professionals From a Special Writing Group of the Stroke Council American Heart Association. Stroke. 1999;30:905–915.
    1. Qureshi A.I., Mendelow A.D., Hanley D.F. Intracerebral haemorrhage. Lancet. 2009;373:1632–1644.
    1. MacLellan C.L., Silasi G., Poon C.C., Edmundson C.L., Buist R., Peeling J., Colbourne F. Intracerebral hemorrhage models in rat: Comparing collagenase to blood infusion. J. Cereb. Blood Flow Metab. 2007;28:516–525.
    1. Wang J., Doré S. Inflammation after intracerebral hemorrhage. J. Cereb. Blood Flow Metab. 2007;27:894–908.
    1. Aronowski J., Zhao X. Molecular pathophysiology of cerebral hemorrhage: Secondary brain injury. Stroke. 2011;42:1781–1786.
    1. Hallenbeck J., Hansson G., Becker K. Immunology of ischemic vascular disease: Plaque to attack. Trends Immunol. 2005;26:550–556.
    1. Loftspring M.C., McDole J., Lu A., Clark J.F., Johnson A.J. Intracerebral hemorrhage leads to infiltration of several leukocyte populations with concomitant pathophysiological changes. J. Cereb. Blood Flow Metab. 2008;29:137–143.
    1. Yilmaz G. Role of T lymphocytes and interferon-in ischemic stroke. Circulation. 2006;113:2105–2112.
    1. Shichita T., Sugiyama Y., Ooboshi H., Sugimori H., Nakagawa R., Takada I., Iwaki T., Okada Y., Iida M., Cua D.J., et al. Pivotal role of cerebral interleukin-17—Producing γδT cells in the delayed phase of ischemic brain injury. Nat. Med. 2009;15:946–950.
    1. Baird A.E. The forgotten lymphocyte: Immunity and stroke. Circulation. 2006;113:2035–2036.
    1. Yan J., Read S.J., Henderson R.D., Hull R., O’Sullivan J.D., McCombe P.A., Greer J.M. Frequency and function of regulatory T cells after ischaemic stroke in humans. J. Neuroimmunol. 2012;243:89–94.
    1. Vignali D.A.A., Collison L.W., Workman C.J. How regulatory T cells work. Nat. Rev. Immunol. 2008;8:523–532.
    1. Jeong S.W., Chu K., Jung K.H., Kim S.U., Kim M., Roh J.K. Human neural stem cell transplantation promotes functional recovery in rats with experimental intracerebral hemorrhage. Stroke. 2003;34:2258–2263.
    1. Kelly S. Transplanted human fetal neural stem cells survive migrate and differentiate in ischemic rat cerebral cortex. Proc. Natl. Acad. Sci. USA. 2004;101:11839–11844.
    1. Martino G., Pluchino S. The therapeutic potential of neural stem cells. Nat. Rev. Neurosci. 2006;7:395–406.
    1. Lee H.J., Kim K.S., Kim E.J., Choi H.B., Lee K.H., Park I.H., Ko Y., Jeong S.W., Kim S.U. Brain transplantation of immortalized human neural stem cells promotes functional recovery in mouse intracerebral hemorrhage stroke model. Stem Cells. 2007;25:1204–1212.
    1. Pluchino S., Zanotti L., Rossi B., Brambilla E., Ottoboni L., Salani G., Martinello M., Cattalini A., Bergami A., Furlan R., et al. Neurosphere-derived multipotent precursors promote neuroprotection by an immunomodulatory mechanism. Nature. 2005;436:266–271.
    1. Pluchino S., Quattrini A., Brambilla E., Gritti A., Salani G., Dina G., Galli R., del Carro U., Amadio S., Bergami A., et al. Injection of adult neurospheres induces recovery in a chronic model of multiple sclerosis. Nature. 2003;422:688–694.
    1. Lee J.-P., Jeyakumar M., Gonzalez R., Takahashi H., Lee P.-J., Baek R.C., Clark D., Rose H., Fu G., Clarke J., et al. Stem cells act through multiple mechanisms to benefit mice with neurodegenerative metabolic disease. Nat. Med. 2007;13:439–447.
    1. Wang L., Shi J., van Ginkel F.W., Lan L., Niemeyer G., Martin D.R., Snyder E.Y., Cox N.R. Neural stem/progenitor cells modulate immune responses by suppressing T lymphocytes with nitric oxide and prostaglandin E2. Exp. Neurol. 2009;216:177–183.
    1. Kim S.-Y., Cho H.-S., Yang S.-H., Shin J.-Y., Kim J.-S., Lee S.-T., Chu K., Roh J.-K., Kim S.U., Park C.-G. Soluble mediators from human neural stem cells play a critical role in suppression of T-cell activation and proliferation. J. Neurosci. Res. 2009;87:2264–2272.
    1. Gelderblom M., Leypoldt F., Steinbach K., Behrens D., Choe C.U., Siler D.A., Arumugam T.V., Orthey E., Gerloff C., Tolosa E., et al. Temporal and spatial dynamics of cerebral immune cell accumulation in stroke. Stroke. 2009;40:1849–1857.
    1. Dirnagl U., Iadecola C., Moskowitz M.A. Pathobiology of ischaemic stroke: An integrated view. Trends Neurosci. 1999;22:391–397.
    1. Wennersten A., Meier X., Holmin S., Wahlberg L., Mathiesen T. Proliferation migration and differentiation of human neural stem progenitor cells after transplantation into a rat model of traumatic brain injury. J. Neurosurg. 2004;100:88–96.
    1. Knight J., Hackett C., Breton J., Mao-Draayer Y. Cross-talk between CD4+ T-cells and neural stem/progenitor cells. J. Neurol. Sci. 2011;306:121–128.
    1. Xi G., Keep R.F., Hoff J.T. Mechanisms of brain injury after intracerebral haemorrhage. Lancet Neurol. 2006;5:53–63.
    1. Erickson M.A., Dohi K., Banks W.A. Neuroinflammation: A common pathway in CNS diseases as mediated at the blood-brain barrier. Neuroimmunomodulation. 2012;19:121–130.
    1. Elenkov I.J., Wilder R.L., Chrousos G.P., Vizi E.S. The sympathetic nerve—An integrative interface between two supersystems: The brain and the immune syste. Pharmacol. Rev. 2000;52:595–638.
    1. Woiciechowsky C., Asadullah K., Nestler D., Eberhardt B., Platzer C., Schöning B., Glöckner F., Lanksch W.R., Volk H.D., Döcke W.D. Sympathetic activation triggers systemic interleukin-10 release in immunodepression induced by brain injury. Nat. Med. 1998;4:808–813.
    1. Liesz A., Suri-Payer E., Veltkamp C., Doerr H., Sommer C., Rivest S., Giese T., Veltkamp R. Regulatory T cells are key cerebroprotective immunomodulators in acute experimental stroke. Nat. Med. 2009;15:192–199.
    1. Liesz A., Zhou W., Mracsko E., Karcher S., Bauer H., Schwarting S., Sun L., Bruder D., Stegemann S., Cerwenka A., et al. Inhibition of lymphocyte trafficking shields the brain against deleterious neuroinflammation after stroke. Brain. 2011;134:704–720.
    1. Taylor A., Verhagen J., Blaser K., Akdis M., Akdis C.A. Mechanisms of immune suppression by interleukin-10 and transforming growth factor-beta: The role of T regulatory cells. Immunology. 2006;117:433–442.
    1. O’Garra A., Vieira P.L., Vieira P., Goldfeld A.E. IL-10-producing and naturally occurring CD4+ Tregs: Limiting collateral damage. J. Clin. Investig. 2004;114:1372–1378.
    1. Prass K., Meisel C., Hoflich C., Braun J., Halle E., Wolf T., Ruscher K., Victorov I.V., Priller J., Dirnagl U., 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. Chesney J.A., Kondoh T., Conrad J.A., Low W.C. Collagenase-induced intrastriatal hemorrhage in rats results in long-term locomotor deficits. Stroke. 1995;26:312–316.
    1. Del Bigio M.R., Yan H.J., Buist R., Peeling J. Experimental intracerebral hemorrhage in rats: Magnetic resonance imaging and histopathological correlates. Stroke. 1996;27:2312–2319.
    1. Chen J., Li Y., Wang L., Zhang Z., Lu D., Lu M., Chopp M. Therapeutic benefit of intravenous administration of bone marrow stromal cells after cerebral ischemia in rats. Stroke. 2001;32:1005–1011.

Source: PubMed

3
購読する