Skip to main content
Top
Published in: Journal of Neuroinflammation 1/2022

Open Access 01-12-2022 | Central Nervous System Trauma | Research

PD-L1 signaling in reactive astrocytes counteracts neuroinflammation and ameliorates neuronal damage after traumatic brain injury

Authors: Xiang Gao, Wei Li, Fahim Syed, Fang Yuan, Ping Li, Qigui Yu

Published in: Journal of Neuroinflammation | Issue 1/2022

Login to get access

Abstract

Background

Tissue damage and cellular destruction are the major events in traumatic brain injury (TBI), which trigger sterile neuroimmune and neuroinflammatory responses in the brain. While appropriate acute and transient neuroimmune and neuroinflammatory responses facilitate the repair and adaptation of injured brain tissues, prolonged and excessive neuroimmune and neuroinflammatory responses exacerbate brain damage. The mechanisms that control the intensity and duration of neuroimmune and neuroinflammatory responses in TBI largely remain elusive.

Methods

We used the controlled cortical impact (CCI) model of TBI to study the role of immune checkpoints (ICPs), key regulators of immune homeostasis, in the regulation of neuroimmune and neuroinflammatory responses in the brain in vivo.

Results

We found that de novo expression of PD-L1, a potent inhibitory ICP, was robustly and transiently induced in reactive astrocytes, but not in microglia, neurons, or oligodendrocyte progenitor cells (OPCs). These PD-L1+ reactive astrocytes were highly enriched to form a dense zone around the TBI lesion. Blockade of PD-L1 signaling enlarged brain tissue cavity size, increased infiltration of inflammatory Ly-6CHigh monocytes/macrophages (M/Mɸ) but not tissue-repairing Ly-6CLowF4/80+ M/Mɸ, and worsened TBI outcomes in mice. PD-L1 gene knockout enhanced production of CCL2 that is best known for its ability to interact with its cognate receptor CCR2 on Ly-6CHigh M/Mϕ to chemotactically recruit these cells into inflammatory sites. Mechanically, PD-L1 signaling in astrocytes likely exhibits dual inhibitory activities for the prevention of excessive neuroimmune and neuroinflammatory responses to TBI through (1) the PD-1/PD-L1 axis to suppress the activity of brain-infiltrating PD-1+ immune cells, such as PD-1+ T cells, and (2) PD-L1 intrinsic signaling to regulate the timing and intensity of astrocyte reactions to TBI.

Conclusions

PD-L1+ astrocytes act as a gatekeeper to the brain to control TBI-related neuroimmune and neuroinflammatory responses, thereby opening a novel avenue to study the role of ICP–neuroimmune axes in the pathophysiology of TBI and other neurological disorders.
Appendix
Available only for authorised users
Literature
1.
go back to reference Kolias AG, Rubiano AM, Figaji A, Servadei F, Hutchinson PJ. Traumatic brain injury: global collaboration for a global challenge. Lancet Neurol. 2019;18:136–7.PubMed Kolias AG, Rubiano AM, Figaji A, Servadei F, Hutchinson PJ. Traumatic brain injury: global collaboration for a global challenge. Lancet Neurol. 2019;18:136–7.PubMed
2.
go back to reference Johnson WD, Griswold DP. Traumatic brain injury: a global challenge. Lancet Neurol. 2017;16:949–50.PubMed Johnson WD, Griswold DP. Traumatic brain injury: a global challenge. Lancet Neurol. 2017;16:949–50.PubMed
3.
go back to reference Hyder AA, Wunderlich CA, Puvanachandra P, Gururaj G, Kobusingye OC. The impact of traumatic brain injuries: a global perspective. NeuroRehabilitation. 2007;22:341–53.PubMed Hyder AA, Wunderlich CA, Puvanachandra P, Gururaj G, Kobusingye OC. The impact of traumatic brain injuries: a global perspective. NeuroRehabilitation. 2007;22:341–53.PubMed
4.
go back to reference Nasr IW, Chun Y, Kannan S. Neuroimmune responses in the developing brain following traumatic brain injury. Exp Neurol. 2019;320:112957.PubMed Nasr IW, Chun Y, Kannan S. Neuroimmune responses in the developing brain following traumatic brain injury. Exp Neurol. 2019;320:112957.PubMed
5.
go back to reference Jassam YN, Izzy S, Whalen M, McGavern DB, El Khoury J. Neuroimmunology of traumatic brain injury: time for a paradigm shift. Neuron. 2017;95:1246–65.PubMedPubMedCentral Jassam YN, Izzy S, Whalen M, McGavern DB, El Khoury J. Neuroimmunology of traumatic brain injury: time for a paradigm shift. Neuron. 2017;95:1246–65.PubMedPubMedCentral
7.
go back to reference Corps KN, Roth TL, McGavern DB. Inflammation and neuroprotection in traumatic brain injury. JAMA Neurol. 2015;72:355–62.PubMedPubMedCentral Corps KN, Roth TL, McGavern DB. Inflammation and neuroprotection in traumatic brain injury. JAMA Neurol. 2015;72:355–62.PubMedPubMedCentral
8.
go back to reference Morganti-Kossmann MC, Satgunaseelan L, Bye N, Kossmann T. Modulation of immune response by head injury. Injury. 2007;38:1392–400.PubMed Morganti-Kossmann MC, Satgunaseelan L, Bye N, Kossmann T. Modulation of immune response by head injury. Injury. 2007;38:1392–400.PubMed
9.
go back to reference Xiong Y, Mahmood A, Chopp M. Current understanding of neuroinflammation after traumatic brain injury and cell-based therapeutic opportunities. Chin J Traumatol. 2018;21:137–51.PubMedPubMedCentral Xiong Y, Mahmood A, Chopp M. Current understanding of neuroinflammation after traumatic brain injury and cell-based therapeutic opportunities. Chin J Traumatol. 2018;21:137–51.PubMedPubMedCentral
10.
go back to reference Kumar A, Loane DJ. Neuroinflammation after traumatic brain injury: opportunities for therapeutic intervention. Brain Behav Immun. 2012;26:1191–201.PubMed Kumar A, Loane DJ. Neuroinflammation after traumatic brain injury: opportunities for therapeutic intervention. Brain Behav Immun. 2012;26:1191–201.PubMed
11.
go back to reference Kumar A, et al. Microglial-derived microparticles mediate neuroinflammation after traumatic brain injury. J Neuroinflamm. 2017;14:47. Kumar A, et al. Microglial-derived microparticles mediate neuroinflammation after traumatic brain injury. J Neuroinflamm. 2017;14:47.
12.
go back to reference Davalos D, et al. ATP mediates rapid microglial response to local brain injury in vivo. Nat Neurosci. 2005;8:752–8.PubMed Davalos D, et al. ATP mediates rapid microglial response to local brain injury in vivo. Nat Neurosci. 2005;8:752–8.PubMed
13.
go back to reference Block ML, Zecca L, Hong JS. Microglia-mediated neurotoxicity: uncovering the molecular mechanisms. Nat Rev Neurosci. 2007;8:57–69.PubMed Block ML, Zecca L, Hong JS. Microglia-mediated neurotoxicity: uncovering the molecular mechanisms. Nat Rev Neurosci. 2007;8:57–69.PubMed
14.
go back to reference Zhang D, Hu X, Qian L, O’Callaghan JP, Hong JS. Astrogliosis in CNS pathologies: is there a role for microglia? Mol Neurobiol. 2010;41:232–41.PubMedPubMedCentral Zhang D, Hu X, Qian L, O’Callaghan JP, Hong JS. Astrogliosis in CNS pathologies: is there a role for microglia? Mol Neurobiol. 2010;41:232–41.PubMedPubMedCentral
15.
go back to reference Berghoff AS, Venur VA, Preusser M, Ahluwalia MS. Immune checkpoint inhibitors in brain metastases: from biology to treatment. Am Soc Clin Oncol Educ Book. 2016;35:e116-122.PubMed Berghoff AS, Venur VA, Preusser M, Ahluwalia MS. Immune checkpoint inhibitors in brain metastases: from biology to treatment. Am Soc Clin Oncol Educ Book. 2016;35:e116-122.PubMed
16.
go back to reference Galea I, Bechmann I, Perry VH. What is immune privilege (not)? Trends Immunol. 2007;28:12–8.PubMed Galea I, Bechmann I, Perry VH. What is immune privilege (not)? Trends Immunol. 2007;28:12–8.PubMed
17.
go back to reference Sanmamed MF, Chen L. A paradigm shift in cancer immunotherapy: from enhancement to normalization. Cell. 2018;175:313–26.PubMedPubMedCentral Sanmamed MF, Chen L. A paradigm shift in cancer immunotherapy: from enhancement to normalization. Cell. 2018;175:313–26.PubMedPubMedCentral
18.
19.
go back to reference Sharma P, Allison JP. The future of immune checkpoint therapy. Science. 2015;348:56–61.PubMed Sharma P, Allison JP. The future of immune checkpoint therapy. Science. 2015;348:56–61.PubMed
20.
go back to reference Wei SC, Duffy CR, Allison JP. Fundamental mechanisms of immune checkpoint blockade therapy. Cancer Discov. 2018;8:1069–86.PubMed Wei SC, Duffy CR, Allison JP. Fundamental mechanisms of immune checkpoint blockade therapy. Cancer Discov. 2018;8:1069–86.PubMed
21.
go back to reference Ishida Y, Agata Y, Shibahara K, Honjo T. Induced expression of PD-1, a novel member of the immunoglobulin gene superfamily, upon programmed cell death. EMBO J. 1992;11:3887–95.PubMedPubMedCentral Ishida Y, Agata Y, Shibahara K, Honjo T. Induced expression of PD-1, a novel member of the immunoglobulin gene superfamily, upon programmed cell death. EMBO J. 1992;11:3887–95.PubMedPubMedCentral
22.
go back to reference Wykes MN, Lewin SR. Immune checkpoint blockade in infectious diseases. Nat Rev Immunol. 2018;18:91–104.PubMed Wykes MN, Lewin SR. Immune checkpoint blockade in infectious diseases. Nat Rev Immunol. 2018;18:91–104.PubMed
23.
go back to reference Dyck L, Mills KHG. Immune checkpoints and their inhibition in cancer and infectious diseases. Eur J Immunol. 2017;47:765–79.PubMed Dyck L, Mills KHG. Immune checkpoints and their inhibition in cancer and infectious diseases. Eur J Immunol. 2017;47:765–79.PubMed
24.
go back to reference Naran K, Nundalall T, Chetty S, Barth S. Principles of immunotherapy: implications for treatment strategies in cancer and infectious diseases. Front Microbiol. 2018;9:3158.PubMedPubMedCentral Naran K, Nundalall T, Chetty S, Barth S. Principles of immunotherapy: implications for treatment strategies in cancer and infectious diseases. Front Microbiol. 2018;9:3158.PubMedPubMedCentral
25.
go back to reference Rao M, Valentini D, Dodoo E, Zumla A, Maeurer M. Anti-PD-1/PD-L1 therapy for infectious diseases: learning from the cancer paradigm. Int J Infect Dis. 2017;56:221–8.PubMed Rao M, Valentini D, Dodoo E, Zumla A, Maeurer M. Anti-PD-1/PD-L1 therapy for infectious diseases: learning from the cancer paradigm. Int J Infect Dis. 2017;56:221–8.PubMed
26.
go back to reference Trautmann L, et al. Upregulation of PD-1 expression on HIV-specific CD8+ T cells leads to reversible immune dysfunction. Nat Med. 2006;12:1198–202.PubMed Trautmann L, et al. Upregulation of PD-1 expression on HIV-specific CD8+ T cells leads to reversible immune dysfunction. Nat Med. 2006;12:1198–202.PubMed
27.
go back to reference Kaufmann DE, et al. Upregulation of CTLA-4 by HIV-specific CD4+ T cells correlates with disease progression and defines a reversible immune dysfunction. Nat Immunol. 2007;8:1246–54.PubMed Kaufmann DE, et al. Upregulation of CTLA-4 by HIV-specific CD4+ T cells correlates with disease progression and defines a reversible immune dysfunction. Nat Immunol. 2007;8:1246–54.PubMed
28.
go back to reference Chew GM, et al. TIGIT marks exhausted t cells, correlates with disease progression, and serves as a target for immune restoration in HIV and SIV infection. PLoS Pathog. 2016;12:e1005349.PubMedPubMedCentral Chew GM, et al. TIGIT marks exhausted t cells, correlates with disease progression, and serves as a target for immune restoration in HIV and SIV infection. PLoS Pathog. 2016;12:e1005349.PubMedPubMedCentral
29.
go back to reference Topalian SL, Drake CG, Pardoll DM. Immune checkpoint blockade: a common denominator approach to cancer therapy. Cancer Cell. 2015;27:450–61.PubMedPubMedCentral Topalian SL, Drake CG, Pardoll DM. Immune checkpoint blockade: a common denominator approach to cancer therapy. Cancer Cell. 2015;27:450–61.PubMedPubMedCentral
31.
go back to reference Hargadon KM, Johnson CE, Williams CJ. Immune checkpoint blockade therapy for cancer: an overview of FDA-approved immune checkpoint inhibitors. Int Immunopharmacol. 2018;62:29–39.PubMed Hargadon KM, Johnson CE, Williams CJ. Immune checkpoint blockade therapy for cancer: an overview of FDA-approved immune checkpoint inhibitors. Int Immunopharmacol. 2018;62:29–39.PubMed
32.
go back to reference Frost RB, Farrer TJ, Primosch M, Hedges DW. Prevalence of traumatic brain injury in the general adult population: a meta-analysis. Neuroepidemiology. 2013;40:154–9.PubMed Frost RB, Farrer TJ, Primosch M, Hedges DW. Prevalence of traumatic brain injury in the general adult population: a meta-analysis. Neuroepidemiology. 2013;40:154–9.PubMed
33.
go back to reference Lagbas C, Bazargan-Hejazi S, Shaheen M, Kermah D, Pan D. Traumatic brain injury related hospitalization and mortality in California. Biomed Res Int. 2013;2013:143092.PubMedPubMedCentral Lagbas C, Bazargan-Hejazi S, Shaheen M, Kermah D, Pan D. Traumatic brain injury related hospitalization and mortality in California. Biomed Res Int. 2013;2013:143092.PubMedPubMedCentral
34.
go back to reference Gao X, Wang X, Xiong W, Chen J. In vivo reprogramming reactive glia into iPSCs to produce new neurons in the cortex following traumatic brain injury. Sci Rep. 2016;6:22490.PubMedPubMedCentral Gao X, Wang X, Xiong W, Chen J. In vivo reprogramming reactive glia into iPSCs to produce new neurons in the cortex following traumatic brain injury. Sci Rep. 2016;6:22490.PubMedPubMedCentral
35.
go back to reference Zhao S, Gao X, Dong W, Chen J. The role of 7,8-dihydroxyflavone in preventing dendrite degeneration in cortex after moderate traumatic brain injury. Mol Neurobiol. 2016;53:1884–95.PubMed Zhao S, Gao X, Dong W, Chen J. The role of 7,8-dihydroxyflavone in preventing dendrite degeneration in cortex after moderate traumatic brain injury. Mol Neurobiol. 2016;53:1884–95.PubMed
36.
go back to reference Gao X, Deng P, Xu ZC, Chen J. Moderate traumatic brain injury causes acute dendritic and synaptic degeneration in the hippocampal dentate gyrus. PLoS One. 2011;6:e24566.PubMedPubMedCentral Gao X, Deng P, Xu ZC, Chen J. Moderate traumatic brain injury causes acute dendritic and synaptic degeneration in the hippocampal dentate gyrus. PLoS One. 2011;6:e24566.PubMedPubMedCentral
37.
go back to reference Han R, Luo J, Shi Y, Yao Y, Hao J. PD-L1 (Programmed Death Ligand 1) protects against experimental intracerebral hemorrhage-induced brain injury. Stroke. 2017;48:2255–62.PubMed Han R, Luo J, Shi Y, Yao Y, Hao J. PD-L1 (Programmed Death Ligand 1) protects against experimental intracerebral hemorrhage-induced brain injury. Stroke. 2017;48:2255–62.PubMed
38.
go back to reference Bodhankar S, et al. PD-L1 monoclonal antibody treats ischemic stroke by controlling central nervous system inflammation. Stroke. 2015;46:2926–34.PubMedPubMedCentral Bodhankar S, et al. PD-L1 monoclonal antibody treats ischemic stroke by controlling central nervous system inflammation. Stroke. 2015;46:2926–34.PubMedPubMedCentral
39.
go back to reference Gao X, Chen J. Moderate traumatic brain injury promotes neural precursor proliferation without increasing neurogenesis in the adult hippocampus. Exp Neurol. 2013;239:38–48.PubMed Gao X, Chen J. Moderate traumatic brain injury promotes neural precursor proliferation without increasing neurogenesis in the adult hippocampus. Exp Neurol. 2013;239:38–48.PubMed
40.
go back to reference Smith DH, et al. A model of parasagittal controlled cortical impact in the mouse: cognitive and histopathologic effects. J Neurotrauma. 1995;12:169–78.PubMed Smith DH, et al. A model of parasagittal controlled cortical impact in the mouse: cognitive and histopathologic effects. J Neurotrauma. 1995;12:169–78.PubMed
41.
go back to reference Hamm RJ, Pike BR, O’Dell DM, Lyeth BG, Jenkins LW. The rotarod test: an evaluation of its effectiveness in assessing motor deficits following traumatic brain injury. J Neurotrauma. 1994;11:187–96.PubMed Hamm RJ, Pike BR, O’Dell DM, Lyeth BG, Jenkins LW. The rotarod test: an evaluation of its effectiveness in assessing motor deficits following traumatic brain injury. J Neurotrauma. 1994;11:187–96.PubMed
42.
go back to reference Rochat L, et al. Assessment of impulsivity after moderate to severe traumatic brain injury. Neuropsychol Rehabil. 2010;20:778–97.PubMed Rochat L, et al. Assessment of impulsivity after moderate to severe traumatic brain injury. Neuropsychol Rehabil. 2010;20:778–97.PubMed
43.
go back to reference Wu D, Qiu Y, Gao X, Yuan XB, Zhai Q. Overexpression of SIRT1 in mouse forebrain impairs lipid/glucose metabolism and motor function. PLoS One. 2011;6:e21759.PubMedPubMedCentral Wu D, Qiu Y, Gao X, Yuan XB, Zhai Q. Overexpression of SIRT1 in mouse forebrain impairs lipid/glucose metabolism and motor function. PLoS One. 2011;6:e21759.PubMedPubMedCentral
44.
go back to reference Popovitz J, Mysore SP, Adwanikar H. Long-term effects of traumatic brain injury on anxiety-like behaviors in mice: behavioral and neural correlates. Front Behav Neurosci. 2019;13:6.PubMedPubMedCentral Popovitz J, Mysore SP, Adwanikar H. Long-term effects of traumatic brain injury on anxiety-like behaviors in mice: behavioral and neural correlates. Front Behav Neurosci. 2019;13:6.PubMedPubMedCentral
46.
go back to reference Glosson-Byers NL, et al. Th17 cells demonstrate stable cytokine production in a proallergic environment. J Immunol. 2014;193:2631–40.PubMed Glosson-Byers NL, et al. Th17 cells demonstrate stable cytokine production in a proallergic environment. J Immunol. 2014;193:2631–40.PubMed
47.
go back to reference Fabrizio FP, et al. Gene code CD274/PD-L1: from molecular basis toward cancer immunotherapy. Ther Adv Med Oncol. 2018;10:1758835918815598.PubMedPubMedCentral Fabrizio FP, et al. Gene code CD274/PD-L1: from molecular basis toward cancer immunotherapy. Ther Adv Med Oncol. 2018;10:1758835918815598.PubMedPubMedCentral
48.
go back to reference Mitchell AL, et al. Programmed death ligand 1 (PD-L1) gene variants contribute to autoimmune Addison’s disease and Graves’ disease susceptibility. J Clin Endocrinol Metab. 2009;94:5139–45.PubMed Mitchell AL, et al. Programmed death ligand 1 (PD-L1) gene variants contribute to autoimmune Addison’s disease and Graves’ disease susceptibility. J Clin Endocrinol Metab. 2009;94:5139–45.PubMed
49.
go back to reference Mahoney KM, et al. A secreted PD-L1 splice variant that covalently dimerizes and mediates immunosuppression. Cancer Immunol Immunother. 2019;68:421–32.PubMed Mahoney KM, et al. A secreted PD-L1 splice variant that covalently dimerizes and mediates immunosuppression. Cancer Immunol Immunother. 2019;68:421–32.PubMed
50.
52.
go back to reference Lagos-Cabre R, Burgos-Bravo F, Avalos AM, Leyton L. Connexins in astrocyte migration. Front Pharmacol. 2019;10:1546.PubMed Lagos-Cabre R, Burgos-Bravo F, Avalos AM, Leyton L. Connexins in astrocyte migration. Front Pharmacol. 2019;10:1546.PubMed
53.
go back to reference Zhao S, Wang X, Gao X, Chen J. Delayed and progressive damages to juvenile mice after moderate traumatic brain injury. Sci Rep. 2018;8:7339.PubMedPubMedCentral Zhao S, Wang X, Gao X, Chen J. Delayed and progressive damages to juvenile mice after moderate traumatic brain injury. Sci Rep. 2018;8:7339.PubMedPubMedCentral
54.
go back to reference Jia T, et al. Additive roles for MCP-1 and MCP-3 in CCR2-mediated recruitment of inflammatory monocytes during Listeria monocytogenes infection. J Immunol. 2008;180:6846–53.PubMed Jia T, et al. Additive roles for MCP-1 and MCP-3 in CCR2-mediated recruitment of inflammatory monocytes during Listeria monocytogenes infection. J Immunol. 2008;180:6846–53.PubMed
55.
go back to reference Hammond MD, et al. CCR2+ Ly6C(hi) inflammatory monocyte recruitment exacerbates acute disability following intracerebral hemorrhage. J Neurosci. 2014;34:3901–9.PubMedPubMedCentral Hammond MD, et al. CCR2+ Ly6C(hi) inflammatory monocyte recruitment exacerbates acute disability following intracerebral hemorrhage. J Neurosci. 2014;34:3901–9.PubMedPubMedCentral
56.
go back to reference Bengsch B, et al. Bioenergetic insufficiencies due to metabolic alterations regulated by the inhibitory receptor PD-1 are an early driver of CD8(+) T cell exhaustion. Immunity. 2016;45:358–73.PubMedPubMedCentral Bengsch B, et al. Bioenergetic insufficiencies due to metabolic alterations regulated by the inhibitory receptor PD-1 are an early driver of CD8(+) T cell exhaustion. Immunity. 2016;45:358–73.PubMedPubMedCentral
57.
go back to reference Patsoukis N, et al. PD-1 alters T-cell metabolic reprogramming by inhibiting glycolysis and promoting lipolysis and fatty acid oxidation. Nat Commun. 2015;6:6692.PubMed Patsoukis N, et al. PD-1 alters T-cell metabolic reprogramming by inhibiting glycolysis and promoting lipolysis and fatty acid oxidation. Nat Commun. 2015;6:6692.PubMed
58.
go back to reference Freeman GJ, et al. Engagement of the PD-1 immunoinhibitory receptor by a novel B7 family member leads to negative regulation of lymphocyte activation. J Exp Med. 2000;192:1027–34.PubMedPubMedCentral Freeman GJ, et al. Engagement of the PD-1 immunoinhibitory receptor by a novel B7 family member leads to negative regulation of lymphocyte activation. J Exp Med. 2000;192:1027–34.PubMedPubMedCentral
59.
60.
go back to reference Dong H, et al. Tumor-associated B7–H1 promotes T-cell apoptosis: a potential mechanism of immune evasion. Nat Med. 2002;8:793–800.PubMed Dong H, et al. Tumor-associated B7–H1 promotes T-cell apoptosis: a potential mechanism of immune evasion. Nat Med. 2002;8:793–800.PubMed
61.
go back to reference Keir ME, Butte MJ, Freeman GJ, Sharpe AH. PD-1 and its ligands in tolerance and immunity. Annu Rev Immunol. 2008;26:677–704.PubMed Keir ME, Butte MJ, Freeman GJ, Sharpe AH. PD-1 and its ligands in tolerance and immunity. Annu Rev Immunol. 2008;26:677–704.PubMed
62.
go back to reference Dong H, Zhu G, Tamada K, Chen L. B7–H1, a third member of the B7 family, co-stimulates T-cell proliferation and interleukin-10 secretion. Nat Med. 1999;5:1365–9.PubMed Dong H, Zhu G, Tamada K, Chen L. B7–H1, a third member of the B7 family, co-stimulates T-cell proliferation and interleukin-10 secretion. Nat Med. 1999;5:1365–9.PubMed
63.
go back to reference Chemnitz JM, Parry RV, Nichols KE, June CH, Riley JL. SHP-1 and SHP-2 associate with immunoreceptor tyrosine-based switch motif of programmed death 1 upon primary human T cell stimulation, but only receptor ligation prevents T cell activation. J Immunol. 2004;173:945–54.PubMed Chemnitz JM, Parry RV, Nichols KE, June CH, Riley JL. SHP-1 and SHP-2 associate with immunoreceptor tyrosine-based switch motif of programmed death 1 upon primary human T cell stimulation, but only receptor ligation prevents T cell activation. J Immunol. 2004;173:945–54.PubMed
64.
go back to reference Yokosuka T, et al. Programmed cell death 1 forms negative costimulatory microclusters that directly inhibit T cell receptor signaling by recruiting phosphatase SHP2. J Exp Med. 2012;209:1201–17.PubMedPubMedCentral Yokosuka T, et al. Programmed cell death 1 forms negative costimulatory microclusters that directly inhibit T cell receptor signaling by recruiting phosphatase SHP2. J Exp Med. 2012;209:1201–17.PubMedPubMedCentral
65.
go back to reference Gato-Canas M, et al. PDL1 signals through conserved sequence motifs to overcome interferon-mediated cytotoxicity. Cell Rep. 2017;20:1818–29.PubMed Gato-Canas M, et al. PDL1 signals through conserved sequence motifs to overcome interferon-mediated cytotoxicity. Cell Rep. 2017;20:1818–29.PubMed
66.
go back to reference Dong P, Xiong Y, Yue J, Hanley SJB, Watari H. Tumor-iIntrinsic PD-L1 signaling in cancer initiation, development and treatment: beyond immune evasion. Front Oncol. 2018;8:386.PubMedPubMedCentral Dong P, Xiong Y, Yue J, Hanley SJB, Watari H. Tumor-iIntrinsic PD-L1 signaling in cancer initiation, development and treatment: beyond immune evasion. Front Oncol. 2018;8:386.PubMedPubMedCentral
67.
go back to reference Poggio M, et al. Suppression of exosomal PD-L1 induces systemic anti-tumor immunity and memory. Cell. 2019;177:414-427.e413.PubMedPubMedCentral Poggio M, et al. Suppression of exosomal PD-L1 induces systemic anti-tumor immunity and memory. Cell. 2019;177:414-427.e413.PubMedPubMedCentral
68.
go back to reference Hartley GP, Chow L, Ammons DT, Wheat WH, Dow SW. Programmed Cell Death Ligand 1 (PD-L1) signaling regulates macrophage proliferation and activation. Cancer Immunol Res. 2018;6:1260–73.PubMed Hartley GP, Chow L, Ammons DT, Wheat WH, Dow SW. Programmed Cell Death Ligand 1 (PD-L1) signaling regulates macrophage proliferation and activation. Cancer Immunol Res. 2018;6:1260–73.PubMed
69.
go back to reference Jalali S, et al. Reverse signaling via PD-L1 supports malignant cell growth and survival in classical Hodgkin lymphoma. Blood Cancer J. 2019;9:22.PubMedPubMedCentral Jalali S, et al. Reverse signaling via PD-L1 supports malignant cell growth and survival in classical Hodgkin lymphoma. Blood Cancer J. 2019;9:22.PubMedPubMedCentral
70.
go back to reference Lecis D, Sangaletti S, Colombo MP, Chiodoni C. Immune checkpoint ligand reverse signaling: looking back to go forward in cancer therapy. Cancers (Basel). 2019;11:624. Lecis D, Sangaletti S, Colombo MP, Chiodoni C. Immune checkpoint ligand reverse signaling: looking back to go forward in cancer therapy. Cancers (Basel). 2019;11:624.
71.
go back to reference Lomas-Neira J, et al. Novel role for PD-1:PD-L1 as mediator of pulmonary vascular endothelial cell functions in pathogenesis of indirect ARDS in mice. Front Immunol. 2018;9:3030.PubMedPubMedCentral Lomas-Neira J, et al. Novel role for PD-1:PD-L1 as mediator of pulmonary vascular endothelial cell functions in pathogenesis of indirect ARDS in mice. Front Immunol. 2018;9:3030.PubMedPubMedCentral
72.
go back to reference Butte MJ, Keir ME, Phamduy TB, Sharpe AH, Freeman GJ. Programmed death-1 ligand 1 interacts specifically with the B7–1 costimulatory molecule to inhibit T cell responses. Immunity. 2007;27:111–22.PubMedPubMedCentral Butte MJ, Keir ME, Phamduy TB, Sharpe AH, Freeman GJ. Programmed death-1 ligand 1 interacts specifically with the B7–1 costimulatory molecule to inhibit T cell responses. Immunity. 2007;27:111–22.PubMedPubMedCentral
73.
go back to reference Burda JE, Sofroniew MV. Reactive gliosis and the multicellular response to CNS damage and disease. Neuron. 2014;81:229–48.PubMedPubMedCentral Burda JE, Sofroniew MV. Reactive gliosis and the multicellular response to CNS damage and disease. Neuron. 2014;81:229–48.PubMedPubMedCentral
74.
go back to reference Sofroniew MV. Molecular dissection of reactive astrogliosis and glial scar formation. Trends Neurosci. 2009;32:638–47.PubMedPubMedCentral Sofroniew MV. Molecular dissection of reactive astrogliosis and glial scar formation. Trends Neurosci. 2009;32:638–47.PubMedPubMedCentral
75.
go back to reference Sofroniew MV, Vinters HV. Astrocytes: biology and pathology. Acta Neuropathol. 2010;119:7–35.PubMed Sofroniew MV, Vinters HV. Astrocytes: biology and pathology. Acta Neuropathol. 2010;119:7–35.PubMed
76.
go back to reference Wanner IB, et al. Glial scar borders are formed by newly proliferated, elongated astrocytes that interact to corral inflammatory and fibrotic cells via STAT3-dependent mechanisms after spinal cord injury. J Neurosci. 2013;33:12870–86.PubMedPubMedCentral Wanner IB, et al. Glial scar borders are formed by newly proliferated, elongated astrocytes that interact to corral inflammatory and fibrotic cells via STAT3-dependent mechanisms after spinal cord injury. J Neurosci. 2013;33:12870–86.PubMedPubMedCentral
77.
go back to reference Burda JE, Bernstein AM, Sofroniew MV. Astrocyte roles in traumatic brain injury. Exp Neurol. 2016;275(Pt 3):305–15.PubMed Burda JE, Bernstein AM, Sofroniew MV. Astrocyte roles in traumatic brain injury. Exp Neurol. 2016;275(Pt 3):305–15.PubMed
78.
go back to reference Piccinini AM, Midwood KS. DAMPening inflammation by modulating TLR signalling. Mediators Inflamm. 2010;2010:1. Piccinini AM, Midwood KS. DAMPening inflammation by modulating TLR signalling. Mediators Inflamm. 2010;2010:1.
79.
go back to reference Thakur KK, et al. Therapeutic implications of toll-like receptors in peripheral neuropathic pain. Pharmacol Res. 2017;115:224–32.PubMed Thakur KK, et al. Therapeutic implications of toll-like receptors in peripheral neuropathic pain. Pharmacol Res. 2017;115:224–32.PubMed
80.
go back to reference Anders HJ, Schaefer L. Beyond tissue injury-damage-associated molecular patterns, toll-like receptors, and inflammasomes also drive regeneration and fibrosis. J Am Soc Nephrol. 2014;25:1387–400.PubMedPubMedCentral Anders HJ, Schaefer L. Beyond tissue injury-damage-associated molecular patterns, toll-like receptors, and inflammasomes also drive regeneration and fibrosis. J Am Soc Nephrol. 2014;25:1387–400.PubMedPubMedCentral
81.
go back to reference Farina C, Aloisi F, Meinl E. Astrocytes are active players in cerebral innate immunity. Trends Immunol. 2007;28:138–45.PubMed Farina C, Aloisi F, Meinl E. Astrocytes are active players in cerebral innate immunity. Trends Immunol. 2007;28:138–45.PubMed
82.
go back to reference Lehnardt S. Innate immunity and neuroinflammation in the CNS: the role of microglia in Toll-like receptor-mediated neuronal injury. Glia. 2010;58:253–63.PubMed Lehnardt S. Innate immunity and neuroinflammation in the CNS: the role of microglia in Toll-like receptor-mediated neuronal injury. Glia. 2010;58:253–63.PubMed
83.
go back to reference Rivest S. Regulation of innate immune responses in the brain. Nat Rev Immunol. 2009;9:429–39.PubMed Rivest S. Regulation of innate immune responses in the brain. Nat Rev Immunol. 2009;9:429–39.PubMed
84.
go back to reference Laird MD, et al. High mobility group box protein-1 promotes cerebral edema after traumatic brain injury via activation of toll-like receptor 4. Glia. 2014;62:26–38.PubMed Laird MD, et al. High mobility group box protein-1 promotes cerebral edema after traumatic brain injury via activation of toll-like receptor 4. Glia. 2014;62:26–38.PubMed
85.
go back to reference Wang KY, Yu GF, Zhang ZY, Huang Q, Dong XQ. Plasma high-mobility group box 1 levels and prediction of outcome in patients with traumatic brain injury. Clin Chim Acta. 2012;413:1737–41.PubMed Wang KY, Yu GF, Zhang ZY, Huang Q, Dong XQ. Plasma high-mobility group box 1 levels and prediction of outcome in patients with traumatic brain injury. Clin Chim Acta. 2012;413:1737–41.PubMed
86.
go back to reference Au AK, et al. Cerebrospinal fluid levels of high-mobility group box 1 and cytochrome C predict outcome after pediatric traumatic brain injury. J Neurotrauma. 2012;29:2013–21.PubMedPubMedCentral Au AK, et al. Cerebrospinal fluid levels of high-mobility group box 1 and cytochrome C predict outcome after pediatric traumatic brain injury. J Neurotrauma. 2012;29:2013–21.PubMedPubMedCentral
87.
go back to reference Hayakawa K, Pham LD, Katusic ZS, Arai K, Lo EH. Astrocytic high-mobility group box 1 promotes endothelial progenitor cell-mediated neurovascular remodeling during stroke recovery. Proc Natl Acad Sci U S A. 2012;109:7505–10.PubMedPubMedCentral Hayakawa K, Pham LD, Katusic ZS, Arai K, Lo EH. Astrocytic high-mobility group box 1 promotes endothelial progenitor cell-mediated neurovascular remodeling during stroke recovery. Proc Natl Acad Sci U S A. 2012;109:7505–10.PubMedPubMedCentral
88.
go back to reference Hayakawa K, et al. High-mobility group box 1 from reactive astrocytes enhances the accumulation of endothelial progenitor cells in damaged white matter. J Neurochem. 2013;125:273–80.PubMed Hayakawa K, et al. High-mobility group box 1 from reactive astrocytes enhances the accumulation of endothelial progenitor cells in damaged white matter. J Neurochem. 2013;125:273–80.PubMed
89.
go back to reference Nutma E, van Gent D, Amor S, Peferoen LAN. Astrocyte and oligodendrocyte cross-talk in the central nervous system. Cells. 2020;9:600.PubMedCentral Nutma E, van Gent D, Amor S, Peferoen LAN. Astrocyte and oligodendrocyte cross-talk in the central nervous system. Cells. 2020;9:600.PubMedCentral
90.
go back to reference Olingy CE, et al. Non-classical monocytes are biased progenitors of wound healing macrophages during soft tissue injury. Sci Rep. 2017;7:447.PubMedPubMedCentral Olingy CE, et al. Non-classical monocytes are biased progenitors of wound healing macrophages during soft tissue injury. Sci Rep. 2017;7:447.PubMedPubMedCentral
91.
go back to reference Hashioka S, et al. Interferon-gamma-induced neurotoxicity of human astrocytes. CNS Neurol Disord Drug Targets. 2015;14:251–6.PubMed Hashioka S, et al. Interferon-gamma-induced neurotoxicity of human astrocytes. CNS Neurol Disord Drug Targets. 2015;14:251–6.PubMed
92.
go back to reference Michalska A, Blaszczyk K, Wesoly J, Bluyssen HAR. A positive feedback amplifier circuit that regulates interferon (IFN)-stimulated gene expression and controls type i and type II IFN responses. Front Immunol. 2018;9:1135.PubMedPubMedCentral Michalska A, Blaszczyk K, Wesoly J, Bluyssen HAR. A positive feedback amplifier circuit that regulates interferon (IFN)-stimulated gene expression and controls type i and type II IFN responses. Front Immunol. 2018;9:1135.PubMedPubMedCentral
93.
go back to reference Hottinger AF. Neurologic complications of immune checkpoint inhibitors. Curr Opin Neurol. 2016;29:806–12.PubMed Hottinger AF. Neurologic complications of immune checkpoint inhibitors. Curr Opin Neurol. 2016;29:806–12.PubMed
94.
go back to reference Fellner A, et al. Neurologic complications of immune checkpoint inhibitors. J Neurooncol. 2018;137:601–9.PubMed Fellner A, et al. Neurologic complications of immune checkpoint inhibitors. J Neurooncol. 2018;137:601–9.PubMed
95.
go back to reference Dalakas MC. Neurological complications of immune checkpoint inhibitors: what happens when you “take the brakes off” the immune system. Ther Adv Neurol Disord. 2018;11:1756286418799864.PubMedPubMedCentral Dalakas MC. Neurological complications of immune checkpoint inhibitors: what happens when you “take the brakes off” the immune system. Ther Adv Neurol Disord. 2018;11:1756286418799864.PubMedPubMedCentral
Metadata
Title
PD-L1 signaling in reactive astrocytes counteracts neuroinflammation and ameliorates neuronal damage after traumatic brain injury
Authors
Xiang Gao
Wei Li
Fahim Syed
Fang Yuan
Ping Li
Qigui Yu
Publication date
01-12-2022
Publisher
BioMed Central
Published in
Journal of Neuroinflammation / Issue 1/2022
Electronic ISSN: 1742-2094
DOI
https://doi.org/10.1186/s12974-022-02398-x

Other articles of this Issue 1/2022

Journal of Neuroinflammation 1/2022 Go to the issue