Skip to main content
Immunology logoLink to Immunology
. 2014 Feb 10;141(3):340–344. doi: 10.1111/imm.12187

T cells in the central nervous system: messengers of destruction or purveyors of protection?

James T Walsh 1,2,3, Nikki Watson 4, Jonathan Kipnis 1,2,3,
PMCID: PMC3930372  PMID: 24708415

Abstract

Although the destructive effects of an overactive adaptive immune system have been well established, especially in the context of autoimmune diseases, recently an understanding of the beneficial effects of the adaptive immunity in central nervous system (CNS) injuries has emerged. CD4+ T cells have been shown to benefit injured CNS tissue through various mechanisms; both traditional cytokine signalling and by modulating the phenotype of neural cells in the injury site. One of the major targets of the cytokine signalling in the CNS are myeloid cells, both resident microglia and monocytes, that infiltrate the tissue after injury and whose phenotype; protective or destructive, appears to be directly influenced by T cells. This cross-talk between the adaptive and innate immune systems presents potential new targets that could provide tangible benefits in pathologies that currently have few treatment options.

Keywords: CNS injury, neuroimmunology, neuroprotection, regulatory T cells, T cells

Introduction

Our understanding of the immune system, and the many components that make up the innate and adaptive arms of this complex system, is constantly evolving. For years, the assumption was that the mere presence of activated immune cells in the central nervous system (CNS) was a hallmark of ongoing pathology.1 This view was particularly applicable in the injured CNS, where the presence of activated T lymphocytes was consistently associated with a poor prognosis and exacerbated neuronal loss.2 However, recently this thinking has been challenged, and it is becoming evident that T cells play a critical role in resolving damage after CNS injury. Early works hinting at this demonstrated that the presence of activated immune cells improved recovery after nerve injury.37 Since these early works, further evidence has begun to accumulate in support of a beneficial role for T cells in a wide range of normal brain functions, and that malfunctions in these homeostatic roles may contribute to neurodegenerative disorders,810 and a range of developmental disorders formerly believed to be purely neurological in nature.11,12 This review will highlight emerging data in support of a beneficial role for different T-cell subtypes in CNS acute and chronic neurodegeneration.

Neurodegeneration in the CNS

Injury in the CNS, as in the periphery, results in a cascade of cellular and molecular responses that amplify tissue damage beyond that expected from the severity of the initial injury itself.13,14 This process, called secondary degeneration, can lead to severe neurodegeneration even when the initial insult may have only involved partial injury to the nerve or spinal cord.9,15 Strikingly, however, the secondary degeneration is more extensive in animals lacking an adaptive immune system than in their wild-type counterparts, suggesting a previously unknown neuroprotective role for immune cells.9,15 Restoration of the immune system, and particularly of the T-cell compartment, in immune-deficient mice restores their normal response to CNS injury,16,17 further suggesting that an endogenous immune response to CNS injury is neuroprotective. Importantly, it was discovered that not all T cells can mediate this neuroprotective effect, but that the T cells need to be specific to brain-restricted antigens,4 which probably governs their migration to, and accumulation in, the injured CNS.18,19 Hence, transfer of autoreactive T cells directed against the CNS antigen, myelin basic protein, reduced the secondary degeneration after nerve injury in rats, and this neuroprotection could be provided through both active immunization (via immunization with the myelin basic protein and adjuvant), or passive immunizations (through the transfer of pre-activated myelin basic protein-specific T cells) 4,5,20 (Fig.1).

Figure 1.

Figure 1

Neuroprotective mode(s) of T-cell action in the injured central nervous system (CNS). T cells in the injured CNS are able to act both directly and indirectly to promote neuroprotection. They produce neuroprotective molecules such as brain-derived neurotrophic factor, which can directly promote neuronal survival. In addition, they are able to potentiate neuronal survival after CNS injury by influencing myeloid cell phenotype at the site of injury by promoting a protective M2 skew though production of interleukin-4 (IL-4) and IL-13. These M2 myeloid cells are then able to promote neuroprotection through production of neuroprotective molecules such as transforming growth factor β (TGF-β), insulin-like growth factor 1 (IGF1) and IL-10.

Regulatory T cells – regulators of the immune response to injury

These findings present something of a quandary because there is significant pathology associated with autoreactive T cells in the brain, in such conditions as multiple sclerosis and neuromyelitis optica.21,22 These conditions, unlike the CNS injuries described above, are improved or eliminated by removal of autoreactive T cells. Furthermore, many of the treatments that are approved for use in autoimmune diseases have proven to be detrimental in CNS injury.23 What then causes autoreactive T cells to be beneficial in injury conditions, yet be detrimental in these autoimmune diseases? A key player in controlling autoimmune responses that might hold the answer to this quandary was uncovered some 40 years ago with the discovery that a population of lymphocytes could control adaptive immune responses24 and, some 20 years later, with our understanding of the molecular identity of these CD25+ regulatory T (Treg) cells.25

These cells are marked by expression of the transcription factor FoxP32628 and have been proposed as the key player in controlling autoimmune responses by the adaptive immune system. This subset of T cells acts not to increase the activity of the immune system to salient stimuli, but instead acts as an endogenous brake to ensure that adaptive immune responses are correctly measured in response to their stimulus. Therefore, because it is their role to suppress autoimmune responses, it stands to reason that Treg cells which are necessary for optimal neuro- protection in CNS injury would be detrimental to the autoreactive T-cell response. However, much controversy still exists about the exact role of Treg cells in CNS injury. For example, early work in models of stroke and Parkinson's disease, showed that depletion of Treg cells led to increased neurodegeneration, but increases in Treg cell numbers and function, improved disease outcome, in contrast to what might have been anticipated.29,30 More recent work, using the same models, has shown that Treg cells play a detrimental role after CNS injuries,31 perhaps more in keeping with the idea that they are suppressing a beneficial autoimmune response. While researchers attempt to resolve these discrepancies, there are several factors that have contributed to the disparate findings. Treg cells from naive mice can exhibit plasticity, especially when placed in lymphopenic or inflammatory conditions,3234 probably because of heterogeneity in the fate commitment of the Treg cell population.35 Furthermore, there are two methods commonly used to deplete Treg cells, and both have significant experimental limitations: anti-CD25 antibodies will deplete activated effector cells as well as regulatory T cells,36 while DEREG mice (which express a diphtheria toxin receptor–green fluorescent protein fusion protein under the control of the FoxP3 promoter37) are susceptible to non-specific effects of diphtheria toxin.38,39 In addition to the experimental challenges that have arisen from working with Treg cells, it has become clear that there exists a balance between Treg cell and effector T-cell CD4+ CD25+ Foxp3 activity, which makes it an over-simplification to say that Treg cells are only neuroprotective or only neurodestructive – too little Treg cell activity and the immune response cannot resolve properly, leading to autoimmune disease,25 whereas too much Treg cell activity and the immune response is not sufficient to promote optimal survival.17 It seems that the immune system has evolved not to exert optimal recovery from CNS injuries, but rather to minimize the risk of autoimmune diseases. Hence, the regulation of the immune response to injury appears to be sub-optimal in physiological processes and is a viable therapeutic target given that the risk for autoimmunity can be mitigated.

Mechanistic insight into T-cell-mediated neuroprotection

This still leaves open the question of what functions of T cells mediate this neuroprotection in situations of CNS injury. There are several functions of T cells, both canonical (cytokine-producing) and non-canonical, that can contribute to their neuroprotective phenotype after CNS injury. The non-canonical effects of T cells that have been shown to contribute to the recovery from injury often rely on interactions with neural cells or the injured neurons themselves. For instance, T cells can protect neurons directly through production of neurotrophic factors, such as brain-derived neurotrophic factor (BDNF), when they become activated.4,40 This immune-derived BDNF plays a functional role in the injured CNS, and mice that lack BDNF in their myeloid and T-cell populations exhibit deficits in functional outcomes after inflammatory injury.41 Additionally, glial cells are involved in the T-cell protective response, as they can signal to astrocytes to up-regulate the production of protective thiol compounds and increase their buffering of glutamate.42,43 In addition to these cytokine-independent mechanisms for T-cell-mediated neuroprotection, several recent works have begun to unravel importance cytokine-dependent interactions between T cells and myeloid cells in the outcome of CNS injury. Monocytes that are recruited to the site of injury in a CCR2-dependent manner take on an alternatively activated phenotype as shown by expression of interleukin-10, whereas the resident microglia do not display this alternatively activated phenotype.44 Additionally, the peak of alternative activation of myeloid cells after injury coincides with the peak of T-cell infiltration into the CNS, suggesting a beneficial T-cell effect on myeloid phenotype.45,46 These alternatively activated macrophages are considered tissue building, and can produce trophic molecules, such as insulin-like growth factor 1 and transforming growth factor β, which are important in growth and development and promote recovery in injured CNS tissue.47 Importantly, one of the best-studied routes to produce alternatively activated macrophages is through signalling by interleukin-4, a prototypical T helper type 2 (Th2) cytokine.48 T cells, then, can infiltrate into the tissue and produce Th2 cytokines, contributing to the protective alternatively activated macrophage skew. This response is controlled by Treg cell suppression of the immune response; slight decreases in Treg cells allows for a more efficient Th2 response, which promotes increased alternative activation of macrophages, while complete depletion of Treg cells permits an uncontrolled immune response that decreases the alternative activation of macrophages at the site of injury (unpublished personal communication). Furthermore, mice deficient in T cells show both fewer infiltrating macrophages to the site of injury and greater pro-inflammatory skew of the cells that do enter. T cells are clearly complex cells that play a multi-faceted role in CNS injury that continues to be an active area of research.

Conclusions

Injury to the CNS spontaneously activates a neuroprotective T-cell response, marked by activation and infiltration of T cells into the injured tissue. The exact identity of the T cells that are mediating this response endogenously remains unknown, but auto-antigen-specific T cells have proven to be particularly potent at promoting neuroprotection. This neuroprotective role of effector T cells is regulated by Treg cells, though the varied physiological roles of Treg cells make studying their involvement in injury difficult. In fact, Treg cells could have different roles at the beginning compared with at the end of the immune response to injury. In the initial stages, Treg cell deactivation would allow autoimmune T cells to mediate their beneficial effects, but later the restoration of their suppressive activity would enable the autoimmune injury response to be controlled or terminated. The mechanisms underlying T-cell-mediated neuroprotection remain unclear, and are likely to be multifaceted, however, their interaction with myeloid cells has recently provided great insights. Clearly further studies are needed, but it is becoming evident that appropriate manipulations of this balance between effector and regulatory T cells may provide powerful new treatment options for an array of neurodegenerative and autoimmune pathologies.

Acknowledgments

This work was primarily supported by a grant from the National Institute of Neurological Disorders and Stroke, NIH (NS061973 award to JK).

Disclosures

The authors declare no conflict of interests.

References

  • 1.D'Souza S, Alinauskas K, McCrea E, Goodyer C, Antel JP. Differential susceptibility of human CNS-derived cell populations to TNF-dependent and independent immune-mediated injury. J Neurosci. 1995;15:7293–300. doi: 10.1523/JNEUROSCI.15-11-07293.1995. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.Popovich PG, Stokes BT, Whitacre CC. Concept of autoimmunity following spinal cord injury: possible roles for T lymphocytes in the traumatized central nervous system. J Neurosci Res. 1996;45:349–63. doi: 10.1002/(SICI)1097-4547(19960815)45:4<349::AID-JNR4>3.0.CO;2-9. [DOI] [PubMed] [Google Scholar]
  • 3.Rapalino O, Lazarov-Spiegler O, Agranov E, et al. Implantation of stimulated homologous macrophages results in partial recovery of paraplegic rats. Nat Med. 1998;4:814–21. doi: 10.1038/nm0798-814. [DOI] [PubMed] [Google Scholar]
  • 4.Moalem G, Leibowitz-Amit R, Yoles E, Mor F, Cohen IR, Schwartz M. Autoimmune T cells protect neurons from secondary degeneration after central nervous system axotomy. Nat Med. 1999;5:49–55. doi: 10.1038/4734. [DOI] [PubMed] [Google Scholar]
  • 5.Hauben E, Butovsky O, Nevo U, et al. Passive or active immunization with myelin basic protein promotes recovery from spinal cord contusion. J Neurosci. 2000;20:6421–30. doi: 10.1523/JNEUROSCI.20-17-06421.2000. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Schori H, Kipnis J, Yoles E, WoldeMussie E, Ruiz G, Wheeler LA, Schwartz M. Vaccination for protection of retinal ganglion cells against death from glutamate cytotoxicity and ocular hypertension: implications for glaucoma. Proc Natl Acad Sci USA. 2001;98:3398–403. doi: 10.1073/pnas.041609498. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Avidan H, Kipnis J, Butovsky O, Caspi RR, Schwartz M. Vaccination with autoantigen protects against aggregated β-amyloid and glutamate toxicity by controlling microglia: effect of CD4+ CD25+ T cells. Eur J Immunol. 2004;34:3434–45. doi: 10.1002/eji.200424883. [DOI] [PubMed] [Google Scholar]
  • 8.Frenkel D, Maron R, Burt DS, Weiner HL. Nasal vaccination with a proteosome-based adjuvant and glatiramer acetate clears β-amyloid in a mouse model of Alzheimer disease. J Clin Invest. 2005;115:2423–33. doi: 10.1172/JCI23241. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Serpe CJ, Kohm AP, Huppenbauer CB, Sanders VM, Jones KJ. Exacerbation of facial motoneuron loss after facial nerve transection in severe combined immunodeficient (scid) mice. J Neurosci. 1999;19:Rc7. doi: 10.1523/JNEUROSCI.19-11-j0004.1999. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Zhao W, Beers DR, Liao B, Henkel JS, Appel SH. Regulatory T lymphocytes from ALS mice suppress microglia and effector T lymphocytes through different cytokine-mediated mechanisms. Neurobiol Dis. 2012;48:418–28. doi: 10.1016/j.nbd.2012.07.008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Derecki NC, Cronk JC, Lu Z, Xu E, Abbott SB, Guyenet PG, Kipnis J. Wild-type microglia arrest pathology in a mouse model of Rett syndrome. Nature. 2012;484:105–9. doi: 10.1038/nature10907. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Simard AR, Soulet D, Gowing G, Julien JP, Rivest S. Bone marrow-derived microglia play a critical role in restricting senile plaque formation in Alzheimer's disease. Neuron. 2006;49:489–502. doi: 10.1016/j.neuron.2006.01.022. [DOI] [PubMed] [Google Scholar]
  • 13.Yoles E, Schwartz M. Degeneration of spared axons following partial white matter lesion: implications for optic nerve neuropathies. Exp Neurol. 1998;153:1–7. doi: 10.1006/exnr.1998.6811. [DOI] [PubMed] [Google Scholar]
  • 14.Crowe MJ, Bresnahan JC, Shuman SL, Masters JN, Beattie MS. Apoptosis and delayed degeneration after spinal cord injury in rats and monkeys. Nat Med. 1997;3:73–6. doi: 10.1038/nm0197-73. [DOI] [PubMed] [Google Scholar]
  • 15.Yoles E, Hauben E, Palgi O, et al. Protective autoimmunity is a physiological response to CNS trauma. J Neurosci. 2001;21:3740–8. doi: 10.1523/JNEUROSCI.21-11-03740.2001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Serpe CJ, Coers S, Sanders VM, Jones KJ. CD4+ T, but not CD8+ or B, lymphocytes mediate facial motoneuron survival after facial nerve transection. Brain Behav Immun. 2003;17:393–402. doi: 10.1016/s0889-1591(03)00028-x. [DOI] [PubMed] [Google Scholar]
  • 17.Kipnis J, Mizrahi T, Hauben E, Shaked I, Shevach E, Schwartz M. Neuroprotective autoimmunity: naturally occurring CD4+ CD25+ regulatory T cells suppress the ability to withstand injury to the central nervous system. Proc Natl Acad Sci USA. 2002;99:15620–5. doi: 10.1073/pnas.232565399. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Ling C, Sandor M, Suresh M, Fabry Z. Traumatic injury and the presence of antigen differentially contribute to T-cell recruitment in the CNS. J Neurosci. 2006;26:731–41. doi: 10.1523/JNEUROSCI.3502-05.2006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Archambault AS, Sim J, Gimenez MA, Russell JH. Defining antigen-dependent stages of T cell migration from the blood to the central nervous system parenchyma. Eur J Immunol. 2005;35:1076–85. doi: 10.1002/eji.200425864. [DOI] [PubMed] [Google Scholar]
  • 20.Byram SC, Carson MJ, DeBoy CA, Serpe CJ, Sanders VM, Jones KJ. CD4-positive T cell-mediated neuroprotection requires dual compartment antigen presentation. J Neurosci. 2004;24:4333–9. doi: 10.1523/JNEUROSCI.5276-03.2004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Bettelli E, Baeten D, Jäger A, Sobel RA, Kuchroo VK. Myelin oligodendrocyte glycoprotein-specific T and B cells cooperate to induce a Devic-like disease in mice. J Clin Invest. 2006;116:2393–402. doi: 10.1172/JCI28334. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Paterson PY. Transfer of allergic encephalomyelitis in rats by means of lymph node cells. J Exp Med. 1960;111:119–36. doi: 10.1084/jem.111.1.119. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Yu-Wai-Man P, Griffiths PG. Steroids for traumatic optic neuropathy. Cochrane Database Syst Rev. 2013;6:Cd006032. doi: 10.1002/14651858.CD006032.pub4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Gershon RK, Cohen P, Hencin R, Liebhaber SA. Suppressor T cells. J Immunol. 1972;108:586–90. [PubMed] [Google Scholar]
  • 25.Sakaguchi S, Sakaguchi N, Asano M, Itoh M, Toda M. Immunologic self-tolerance maintained by activated T cells expressing IL-2 receptor α-chains (CD25). Breakdown of a single mechanism of self-tolerance causes various autoimmune diseases. J Immunol. 1995;155:1151–64. [PubMed] [Google Scholar]
  • 26.Fontenot JD, Gavin MA, Rudensky AY. Foxp3 programs the development and function of CD4+ CD25+ regulatory T cells. Nat Immunol. 2003;4:330–6. doi: 10.1038/ni904. [DOI] [PubMed] [Google Scholar]
  • 27.Hori S, Nomura T, Sakaguchi S. Control of regulatory T cell development by the transcription factor Foxp3. Science. 2003;299:1057–61. doi: 10.1126/science.1079490. [DOI] [PubMed] [Google Scholar]
  • 28.Khattri R, Cox T, Yasayko SA, Ramsdell F. An essential role for Scurfin in CD4+ CD25+ T regulatory cells. Nat Immunol. 2003;4:337–42. doi: 10.1038/ni909. [DOI] [PubMed] [Google Scholar]
  • 29.Reynolds AD, Banerjee R, Liu JN, Gendelman HE, Mosley RL. Neuroprotective activities of CD4+ CD25+ regulatory T cells in an animal model of Parkinson's disease. J Leukoc Biol. 2007;82:1083–94. doi: 10.1189/jlb.0507296. [DOI] [PubMed] [Google Scholar]
  • 30.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;15:192–9. doi: 10.1038/nm.1927. [DOI] [PubMed] [Google Scholar]
  • 31.Kleinschnitz C, Kraft P, Dreykluft A, et al. Regulatory T cells are strong promoters of acute ischemic stroke in mice by inducing dysfunction of the cerebral microvasculature. Blood. 2013;121:679–91. doi: 10.1182/blood-2012-04-426734. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Zhou X, Bailey-Bucktrout SL, Jeker LT, et al. Instability of the transcription factor Foxp3 leads to the generation of pathogenic memory T cells in vivo. Nat Immunol. 2009;10:1000–7. doi: 10.1038/ni.1774. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Zhou X, Kong N, Wang J, et al. Cutting edge: all-trans retinoic acid sustains the stability and function of natural regulatory T cells in an inflammatory milieu. J Immunol. 2010;185:2675–9. doi: 10.4049/jimmunol.1000598. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Komatsu N, Mariotti-Ferrandiz ME, Wang Y, Malissen B, Waldmann H, Hori S. Heterogeneity of natural Foxp3+ T cells: a committed regulatory T-cell lineage and an uncommitted minor population retaining plasticity. Proc Natl Acad Sci USA. 2009;106:1903–8. doi: 10.1073/pnas.0811556106. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.d'Hennezel E, Yurchenko E, Sgouroudis E, Hay V, Piccirillo CA. Single-cell analysis of the human T regulatory population uncovers functional heterogeneity and instability within FOXP3+ cells. J Immunol. 2011;186:6788–97. doi: 10.4049/jimmunol.1100269. [DOI] [PubMed] [Google Scholar]
  • 36.Couper KN, Lanthier PA, Perona-Wright G, Kummer LW, Chen W, Smiley ST, Mohrs M, Johnson LL. Anti-CD25 antibody-mediated depletion of effector T cell populations enhances susceptibility of mice to acute but not chronic Toxoplasma gondii infection. J Immunol. 2009;182:3985–94. doi: 10.4049/jimmunol.0803053. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Lahl K, Loddenkemper C, Drouin C, et al. Selective depletion of Foxp3+ regulatory T cells induces a scurfy-like disease. J Exp Med. 2007;204:57–63. doi: 10.1084/jem.20061852. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Meyer Zu, Horste G, Zozulya AL, El-Haddad H, Lehmann HC, Hartung HP, Wiendl H, Kieseier BC. Active immunization induces toxicity of diphtheria toxin in diphtheria resistant mice – implications for neuroinflammatory models. J Immunol Methods. 2010;354:80–4. doi: 10.1016/j.jim.2010.01.012. [DOI] [PubMed] [Google Scholar]
  • 39.Lahl K, Sparwasser T. In vivo depletion of FoxP3+ Tregs using the DEREG mouse model. Methods Mol Biol. 2011;707:157–72. doi: 10.1007/978-1-61737-979-6_10. [DOI] [PubMed] [Google Scholar]
  • 40.Kerschensteiner M, Gallmeier E, Behrens L, et al. Activated human T cells, B cells, and monocytes produce brain-derived neurotrophic factor in vitro and in inflammatory brain lesions: a neuroprotective role of inflammation? J Exp Med. 1999;189:865–70. doi: 10.1084/jem.189.5.865. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Linker RA, Lee DH, Demir S, et al. Functional role of brain-derived neurotrophic factor in neuroprotective autoimmunity: therapeutic implications in a model of multiple sclerosis. Brain. 2010;133(Pt 8):2248–63. doi: 10.1093/brain/awq179. [DOI] [PubMed] [Google Scholar]
  • 42.Garg SK, Banerjee R, Kipnis J. Neuroprotective immunity: T cell-derived glutamate endows astrocytes with a neuroprotective phenotype. J Immunol. 2008;180:3866–73. doi: 10.4049/jimmunol.180.6.3866. [DOI] [PubMed] [Google Scholar]
  • 43.Garg SK, Kipnis J, Banerjee R. IFN-γ and IL-4 differentially shape metabolic responses and neuroprotective phenotype of astrocytes. J Neurochem. 2009;108:1155–66. doi: 10.1111/j.1471-4159.2009.05872.x. [DOI] [PubMed] [Google Scholar]
  • 44.Shechter R, London A, Varol C, et al. Infiltrating blood-derived macrophages are vital cells playing an anti-inflammatory role in recovery from spinal cord injury in mice. PLoS Med. 2009;6:e1000113. doi: 10.1371/journal.pmed.1000113. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Miron VE, Boyd A, Zhao JW, et al. M2 microglia and macrophages drive oligodendrocyte differentiation during CNS remyelination. Nat Neurosci. 2013;16:1211–8. doi: 10.1038/nn.3469. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Popovich PG, Wei P, Stokes BT. Cellular inflammatory response after spinal cord injury in Sprague-Dawley and Lewis rats. J Comp Neurol. 1997;377:443–64. doi: 10.1002/(sici)1096-9861(19970120)377:3<443::aid-cne10>3.0.co;2-s. [DOI] [PubMed] [Google Scholar]
  • 47.Kigerl KA, Gensel JC, Ankeny DP, Alexander JK, Donnelly DJ, Popovich PG. Identification of two distinct macrophage subsets with divergent effects causing either neurotoxicity or regeneration in the injured mouse spinal cord. J Neurosci. 2009;29:13435–44. doi: 10.1523/JNEUROSCI.3257-09.2009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Anthony RM, Urban JF, Jr, Alem F, Hamed HA, Rozo CT, Boucher JL, Van Rooijen N, Gause WC. Memory TH2 cells induce alternatively activated macrophages to mediate protection against nematode parasites. Nat Med. 2006;12:955–60. doi: 10.1038/nm1451. [DOI] [PMC free article] [PubMed] [Google Scholar]

Articles from Immunology are provided here courtesy of British Society for Immunology

RESOURCES