Elsevier

Journal of Neuroimmunology

Volume 318, 15 May 2018, Pages 36-44
Journal of Neuroimmunology

FasL incapacitation alleviates CD4+ T cells-induced brain injury through remodeling of microglia polarization in mouse ischemic stroke

https://doi.org/10.1016/j.jneuroim.2018.01.017Get rights and content

Highlights

  • FasL deficiency changes the subsets of CD4+ T cells after MCAO.

  • CD4+ T cells facilitated microglial pro-inflammation through NF-κB pathway.

  • FasL of CD4+ T cells played an important role in M1 microglia polarization.

  • FasL-mediated crosstalk between CD4+ T cells and microglia promoted neuronal injury.

Abstract

Inflammation responses involving the crosstalk between infiltrated T cells and microglia play crucial roles in ischemia stroke. Recent studies showed that Fas ligand (FasL) mutation could reduce post-stroke T cell invasion and microglia activation. In this study, we demonstrated that CD4+ T cells could induce M1 microglia polarization through NF-κB signaling pathway, whereas FasL mutant CD4+ T cells significantly reversed this effect. Besides, Th17/Treg cells balance was skewed into Treg cells after FasL mutation. In addition, conditioned medium from co-culture of FasL mutant CD4+ T cells and microglia could alleviate neuronal injury. Collectively, FasL incapacitation could alleviate CD4+ T cells-induced inflammation through remodeling microglia polarization, suggesting a therapeutic potential for control of inflammation responses after ischemic stroke.

Introduction

Ischemic stroke is one of the leading causes of death and mortality worldwide (Tsai et al., 2015). Inflammation, which occurs throughout the whole process of cerebral ischemia reperfusion, is considered to be the most important pathological mechanism of ischemic stroke (Chamorro et al., 2012). After brain ischemia, neuronal death triggers local immune responses, leading to microglia activation and peripheral inflammatory cells migration into the lesion area (Waisman et al., 2015). Activated microglia act synergistically with infiltrating inflammatory cells and release large amounts of pro-inflammatory mediators such as interleukin-1 (IL-1), IL-6, interferon-γ (IFN-γ), tumor necrosis factor-α (TNF-α) and other deleterious substances (Iadecola and Anrather, 2011), resulting in additional neuronal death, disruption of blood brain barrier (BBB), brain edema and aggravation of ischemic brain damage (Brait et al., 2010; Ceulemans et al., 2010).

Microglia are resident innate immune cells in the central nervous system which act as a sensor (Czeh et al., 2011). In recent years, large amounts of studies indicate that microglia are highly plastic cells and can perform different phenotypes and functions in response to different environment stimulus (Perry et al., 2010). Activated microglia can polarize to pro-inflammatory (classically activated, or M1) or anti-inflammatory (alternatively activated, or M2) phenotypes (David and Kroner, 2011; Mosser and Edwards, 2008). M1 microglia produce high levels of pro-inflammatory cytokines which are essential for the clearance of invading pathogens and injured tissue products, but resulting in collateral tissue damage at the same time (Benarroch, 2013). Inversely, M2 microglia secret anti-inflammatory cytokines, such as transforming growth factor-β (TGF-β) and neurotrophins, play crucial roles in promoting wound healing and suppressing destructive immune responses (Martinez et al., 2009). These dual and opposing activation phenotypes of microglia have been found in ischemic stroke and determine the regulation of inflammatory responses after brain infarction (Hu et al., 2012; Yenari et al., 2010). However, the mechanisms of how microglia change phenotypes after ischemic stroke are still unknown.

As the major effector cells of lymphocytes, CD4+ T cells (helper T cells, Th) are involved in the evolution of ischemic stroke and accompanying neurological deficits. CD4+ T cells invade into the ischemic brain within 24 h and peak on day 3 after focal infarction (Gelderblom et al., 2009; Jander et al., 1995). Infiltrated CD4+ T cells regulate brain inflammation though their cytokines production. Typically, CD4+ T cells can be divided into Th1, Th2, Th17, and regulatory T (Treg) cells based on their cytokine secretion profiles (Zhu et al., 2010). Recent studies suggest that different CD4+ T cell subsets may have distinct effects on the injured brain. Th1 and Th17 cells produce specific cytokines such as IL-1, IFN-γ, IL-17, IL-22 and so on, so they are characterized by high pro-inflammatory function and aggravating brain damage (Arumugam et al., 2005; Stockinger et al., 2007). On the contrary, Th2 and Treg cells are characterized by suppressing excessive inflammation, achieving self-tolerance and brain repair via producing anti-inflammatory cytokines such as IL-4, IL-10 and TGF-β (Liesz et al., 2009; Xie et al., 2015; Yilmaz and Granger, 2010). The balance between Th1/Th17 and Th2/Treg cells predominantly affect the function of CD4+ T cells and determine the outcome of brain inflammation (Afzali et al., 2007; Gu et al., 2015). Substantial evidence proves that CD4+ T cells can interact with microglia after brain injury, resulting in injurious or neurotrophic outcomes in vicinity (Strachan-Whaley et al., 2014). However, the molecular mechanisms of their crosstalk are still debated.

Fas ligand (FasL), a member of TNF protein family, has been shown to induce apoptosis in Fas-expressing cells and serves as a key death factor in the immune system (Lettau et al., 2008). Emerging evidence suggests that FasL plays important roles in inflammation since its capacity of inducing production of pro-inflammatory cytokines and leukocyte infiltration (Kunes et al., 2009). In previous studies, we proved that FasL mutation profoundly reduced infarct volume, neutrophil recruitment and microglia activation after ischemic stroke (Meng et al., 2016; Niu et al., 2012). However, little is known about the effect of FasL regulating CD4+ T cell subsets and crosstalk between CD4+ T cells and microglia.

In the present study, we intended to investigate the influence of FasL depletion on brain inflammation by virtue of ischemic stroke using middle cerebral artery occlusion (MCAO) models and explore the underlying mechanism of microglia polarization using co-culturing system involving CD4+ T cells from FasL-mutant (gld) mice and primary microglia. As a result, we found that FasL mutant CD4+ T cells could remodel microglia polarization. More importantly, we showed that proportion of CD4+ T cells were reduced both in the brain and periphery of FasL mutant mice after middle cerebral artery occlusion (MCAO), and that Th17/Treg cells balance was skewed into Treg cells.

Section snippets

Middle cerebral artery occlusion model in mice

Fourteen-week-old male recombination activating gene 1–deficient (Rag1−/−) mice, FasL mutant (gld) mice (22-24 g) and their wild-type control C57BL/6 J (B6) mice were purchased from the Animal Model Research Center of Nanjing University. All animal experiments were approved by the Animal Care Committee in China and performed according to institutional guidelines. Mice were subjected to middle cerebral artery occlusion (MCAO) by intraluminal filament technique for 60 min as previously described (

FasL incapacitation remodels CD4+ T cells reversed M1 microglia polarization

Initially,the crosstalk between CD4+ T cells and microglia were explored using co-culturing models involving CD4+ T cells from mice and primary microglia induced by LPS into an inflammatory state before co-culture. And gene expression of M1 makers (iNOS, TNF-α and CD86) and M2 makers (CD206 and Ym1/2) of microglia were measured by q-PCR. We showed that CD4+ T cells from wild type mice of MCAO models could significantly up-regulate mRNA expression of M1 markers and down-regulate M2 markers of

Discussion

In this study, we reported some novel findings including: (1) FasL deficiency changes the subsets of CD4+ T cells and alleviates inflammatory reaction after MCAO. (2) CD4+ T cells facilitated microglial pro-inflammation through NF-κB signaling pathway and FasL of CD4+ T cells played an important role in M1 microglia polarization. (3) FasL-mediated crosstalk between CD4+ T cells and microglia aggravated neuronal death and apoptosis.

In central nervous system, FasL has been observed to elevate in

Acknowledgments

This research was supported by the National Natural Science Foundation of China (81230026, 81630028, 81402065), the Science and Technology Department of Jiangsu Province (BE2016610) and Jiangsu Province Key Medical Discipline (ZDXKA2016020) and Nanjing Medical Science and technique Development Foundation (QRX17118).

Conflicts of interest

The authors declare no conflicts of interest.

References (61)

  • Z.H. Zhang et al.

    Hydroxy-safflor yellow A attenuates Abeta(1)(−)(4)(2)-induced inflammation by modulating the JAK2/STAT3/NF-kappaB pathway

    Brain Res.

    (2014)
  • B. Afzali et al.

    The role of T helper 17 (Th17) and regulatory T cells (Treg) in human organ transplantation and autoimmune disease

    Clin. Exp. Immunol.

    (2007)
  • T.V. Arumugam et al.

    Stroke and T-cells

    NeuroMolecular Med.

    (2005)
  • E.E. Benarroch

    Microglia: multiple roles in surveillance, circuit shaping, and response to injury

    Neurology

    (2013)
  • V.H. Brait et al.

    Mechanisms contributing to cerebral infarct size after stroke: gender, reperfusion, T lymphocytes, and Nox2-derived superoxide

    J. Cereb. Blood Flow Metab.

    (2010)
  • V.H. Brait et al.

    Importance of T lymphocytes in brain injury, immunodeficiency, and recovery after cerebral ischemia

    J. Cereb. Blood Flow Metab.

    (2012)
  • A.G. Ceulemans et al.

    The dual role of the neuroinflammatory response after ischemic stroke: modulatory effects of hypothermia

    J. Neuroinflammation

    (2010)
  • S. Chabot et al.

    Mechanisms of IL-10 production in human microglia-T cell interaction

    J. Immunol.

    (1999)
  • A. Chamorro et al.

    The immunology of acute stroke

    Nat. Rev. Neurol.

    (2012)
  • J. Chen et al.

    Therapeutic benefit of intravenous administration of bone marrow stromal cells after cerebral ischemia in rats

    Stroke

    (2001)
  • W. Chen et al.

    Role of double-negative regulatory T cells in long-term cardiac xenograft survival

    J. Immunol.

    (2003)
  • W. Chen et al.

    Donor lymphocyte infusion induces long-term donor-specific cardiac xenograft survival through activation of recipient double-negative regulatory T cells

    J. Immunol.

    (2005)
  • Z.B. Chen et al.

    Human urinary kallidinogenase suppresses cerebral inflammation in experimental stroke and downregulates nuclear factor-kappaB

    J. Cereb. Blood Flow Metab.

    (2010)
  • P.L. Cohen et al.

    Lpr and gld: single gene models of systemic autoimmunity and lymphoproliferative disease

    Annu. Rev. Immunol.

    (1991)
  • M. Czeh et al.

    The yin and yang of microglia

    Dev. Neurosci.

    (2011)
  • S. David et al.

    Repertoire of microglial and macrophage responses after spinal cord injury

    Nat. Rev. Neurosci.

    (2011)
  • M. Gelderblom et al.

    Temporal and spatial dynamics of cerebral immune cell accumulation in stroke

    Stroke

    (2009)
  • L. Gu et al.

    T cells and cerebral ischemic stroke

    Neurochem. Res.

    (2015)
  • R.J. Hamm et al.

    The rotarod test: an evaluation of its effectiveness in assessing motor deficits following traumatic brain injury

    J. Neurotrauma

    (1994)
  • O.A. Harari et al.

    NF-kappaB and innate immunity in ischemic stroke

    Ann. N. Y. Acad. Sci.

    (2010)
  • Cited by (20)

    • Resveratrol Alleviates Ischemic Brain Injury by Inhibiting the Activation of Pro-Inflammatory Microglia Via the CD147/MMP-9 Pathway

      2022, Journal of Stroke and Cerebrovascular Diseases
      Citation Excerpt :

      All animals were euthanized by intraperitoneal injection of pentobarbital sodium ≥ 100 mg/kg to collect the tissue. Referring to the previous studies,18,19 unilateral focal cerebral ischemia induced by MCAO was performed in 8-week-old mice using a filament. Briefly, mice were anesthetized with 2% isoflurane at a flow rate of 1.8 L/min in 100% oxygen.

    • Modulation of astrocyte phenotype in response to T-cell interaction

      2021, Journal of Neuroimmunology
      Citation Excerpt :

      There is increasing evidence that the brain is not devoid of peripheral immune cells. Immune cells can enter the brain via the lymphatic vessels, interacting with neurons and glial cells (Lopes Pinheiro et al., 2016; Schachtele et al., 2014), playing vital roles in immune surveillance (Louveau et al., 2018; Louveau et al., 2015) and bi-directional communication between the immune system and central nervous system (CNS) (Engelhardt and Ransohoff, 2012; Zhao et al., 2018; Chabot et al., 1999; Gimsa et al., 2004). T-cells have been identified as resident cells in the brains of humans and rodents (Smolders et al., 2018; Xie et al., 2015).

    • Neuroinflammation and fibrosis in stroke: The good, the bad and the ugly

      2020, Journal of Neuroimmunology
      Citation Excerpt :

      Fas ligand (FasL) belongs to the TNF family of cytokines and plays a critical role in stroke pathology. FasL mutation reduces stroke injury by attenuating CD4+ T cells and nuclear factor-kappa B (NF-κB)-mediated M1 microglia polarization in mice model of MCAO (Zhao et al., 2018). PDPK1 is downstream of FasL signaling and inhibition of PDPK1, by treatment with its inhibitor BX-912, reduces cytotoxicity of CD8+ T cells after stroke in mice (Fan et al., 2020).

    • EZH2 inhibitor DZNep modulates microglial activation and protects against ischaemic brain injury after experimental stroke

      2019, European Journal of Pharmacology
      Citation Excerpt :

      During the reperfusion stage, the plates were then returned to normal medium in normal incubator for 12–24 h. Unilateral focal cerebral ischemia was induced in eight-week-old mice by transient middle cerebral artery occlusion (MCAO) using a filament as described previously (Zhao et al., 2018). Briefly, mice were anesthetized with 2% isoflurane in 100% oxygen at a flow rate of 1.8 L/min.

    • Chronic colitis induces meninges traffic of gut-derived T cells, unbalances M1 and M2 microglia/macrophage and increases ischemic brain injury in mice

      2019, Brain Research
      Citation Excerpt :

      The shifting of microglia/macrophage phenotype is mediated by CD4+ T cells after brain injury, resulting in injurious or neurotrophic outcomes in vicinity (Wang et al., 2016). Emerging evidences suggest that CD4+ T cells could induce pro-inflammatory cytokine production from M1 microglia after brain injury (Dasgupta et al., 2003; Prajeeth et al., 2014; Zhao et al., 2018). Ischemic stroke can cause gut microbiota dysbiosis that influences stroke outcomes by promoting activated CD4+ T cell migration (Benakis et al., 2016; Qin et al., 2007; Singh et al., 2016; Zonis et al., 2015).

    View all citing articles on Scopus
    1

    These authors contributed equally to this study.

    View full text