Skip to main content

Advertisement

Log in

Microglial priming of antigen presentation and adaptive stimulation in Alzheimer’s disease

  • Review
  • Published:
Cellular and Molecular Life Sciences Aims and scope Submit manuscript

Abstract

The prominent pathological consequences of Alzheimer’s disease (AD) are the misfolding and mis-sorting of two cellular proteins, amyloid-β and microtubule-associated protein Tau. The accumulation of toxic phosphorylated Tau inside the neurons induces the increased processing of amyloid-β-associated signaling cascade and vice versa. Neuroinflammation-driven synaptic depletion and cognitive decline are substantiated by the cross talk of activated microglia and astroglia, leading to neuron degeneration. Microglia are the brain-resident immune effectors that prove their diverse functions in maintaining CNS homeostasis via collaboration with astrocytes and T lymphocytes. Age-related senescence and chronic inflammation activate microglia with increased pro-inflammatory markers, oxidative damage and phagocytosis. But the improper processing of misfolded protein via lysosomal pathway destines the spreading of ‘seed’ constituents to the nearby healthy neurons. Primed microglia process and present self-antigen such as amyloid-β and modified Tau to the infiltrated T lymphocytes through MHC I/II molecules. After an effective conversation with CD4+ T cells, microglial phenotype can be altered from pro-active M1 to neuro-protective M2 type, which corresponds to the tissue remodeling and homeostasis. In this review, we are focusing on the change in functionality of microglia from innate to adaptive immune response in the context of neuroprotection, which may help in the search of novel immune therapy in AD.

This is a preview of subscription content, log in via an institution to check access.

Access this article

Price excludes VAT (USA)
Tax calculation will be finalised during checkout.

Instant access to the full article PDF.

Fig. 1
Fig. 2
Fig. 3
Fig. 4

Similar content being viewed by others

Abbreviations

BACE1:

Beta secretase 1

PTMs:

Post-translational modifications

BBB:

Blood–brain barrier

CNS:

Central nervous system

ROS:

Reactive oxygen species

NO:

Nitric oxide

iNOS:

Inorganic nitric oxide synthase

CD:

Cluster of differentiation

MHC:

Major histocompatibility complex

TGFβ:

Transforming growth factor β

TNFα:

Tumor necrosis factor α

IL:

Interleukin

IFN:

Interferon

TH :

Helper T cell

Treg :

Regulatory T cell

KO:

Knockout

CR:

Complement receptor

NFκB:

Nuclear factor κ for B cell

mTOR:

Mammalian target for rapamycin

Akt:

Protein kinase B

ATP:

Adenosine tri-phosphate

APC:

Antigen-presenting cell

TREM2:

Triggering receptor expressed on myeloid cells 2

ApoE:

Apolipoprotein E

NFκB:

Nuclear factor kappa-light-chain-enhancer of activated B cells

p38-MAPK:

p38-mitogen-activated protein kinases

CSF1R:

Colony-stimulating factor-1 receptor

References

  1. Barnham KJ, Masters CL, Bush AI (2004) Neurodegenerative diseases and oxidative stress. Nat Rev Drug Discov 3:205

    Article  CAS  PubMed  Google Scholar 

  2. Soto C (2003) Unfolding the role of protein misfolding in neurodegenerative diseases. Nat Rev Neurosci 4:49

    Article  CAS  PubMed  Google Scholar 

  3. Sheikh S, Haque E, Mir SS (2013) Neurodegenerative diseases: multifactorial conformational diseases and their therapeutic interventions. J Neurodegener Dis 2013:8

    Google Scholar 

  4. Prinz M, Priller J (2014) Microglia and brain macrophages in the molecular age: from origin to neuropsychiatric disease. Nat Rev Neurosci 15:300

    Article  CAS  PubMed  Google Scholar 

  5. Minghetti L, Ajmone-Cat MA, De Berardinis MA, De Simone R (2005) Microglial activation in chronic neurodegenerative diseases: roles of apoptotic neurons and chronic stimulation. Brain Res Rev 48:251–256

    Article  CAS  PubMed  Google Scholar 

  6. Kettenmann H, Hanisch U-K, Noda M, Verkhratsky A (2011) Physiology of microglia. Physiol Rev 91:461–553

    Article  CAS  PubMed  Google Scholar 

  7. Graeber MB, Li W, Rodriguez ML (2011) Role of microglia in CNS inflammation. FEBS Lett 585:3798–3805

    Article  CAS  PubMed  Google Scholar 

  8. Aloisi F, Ria F, Adorini L (2000) Regulation of T-cell responses by CNS antigen-presenting cells: different roles for microglia and astrocytes. Immunol Today 21:141–147

    Article  CAS  PubMed  Google Scholar 

  9. Aguzzi A, Barres BA, Bennett ML (2013) Microglia: scapegoat, saboteur, or something else? Science 339:156–161

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Falsig J, Julius C, Margalith I, Schwarz P, Heppner FL, Aguzzi A (2008) A versatile prion replication assay in organotypic brain slices. Nat Neurosci 11:109–117

    Article  CAS  PubMed  Google Scholar 

  11. Block ML, Hong J-S (2005) Microglia and inflammation-mediated neurodegeneration: multiple triggers with a common mechanism. Prog Neurobiol 76:77–98

    Article  CAS  PubMed  Google Scholar 

  12. Aloisi F, Ria F, Columba-Cabezas S, Hess H, Penna G, Adorini L (1999) Relative efficiency of microglia, astrocytes, dendritic cells and B cells in naive CD4+ T cell priming and Th1/Th2 cell restimulation. Eur J Immunol 29:2705–2714

    Article  CAS  PubMed  Google Scholar 

  13. Selkoe DJ (2000) Toward a comprehensive theory for Alzheimer’s disease. Hypothesis: Alzheimer’s disease is caused by the cerebral accumulation and cytotoxicity of amyloidβ’ protein. Ann N Y Acad Sci 924:17–25

    Article  CAS  PubMed  Google Scholar 

  14. Kametani F, Hasegawa M (2018) Reconsideration of amyloid hypothesis and Tau hypothesis in Alzheimer’s disease. Front Neurosci 12:25

    Article  PubMed  PubMed Central  Google Scholar 

  15. Merezhko M, Brunello CA, Yan X, Vihinen H, Jokitalo E, Uronen R-L, Huttunen HJ (2018) Secretion of tau via an unconventional non-vesicular mechanism. Cell Rep 25(2027–2035):e4

    Google Scholar 

  16. Wang Y et al (2017) The release and trans-synaptic transmission of Tau via exosomes. Mol Neurodegener 12:5

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Mesulam M-M (2000) A plasticity-based theory of the pathogenesis of Alzheimer’s disease. Ann N Y Acad Sci 924:42–52

    Article  CAS  PubMed  Google Scholar 

  18. Giovannetti EA, Fuhrmann M (2018) Unsupervised excitation: GABAergic dysfunctions in Alzheimer’s disease. Brain Res 1707:216–226

    Article  CAS  Google Scholar 

  19. Yang T, Li S, Xu H, Walsh DM, Selkoe DJ (2017) Large soluble oligomers of amyloid β-protein from alzheimer brain are far less neuroactive than the smaller oligomers to which they dissociate. J Neurosci 37:152–163

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Solomon B (2004) Alzheimer’s disease and immunotherapy. Curr Alzheimer Res 1:149–163

    Article  CAS  PubMed  Google Scholar 

  21. Wang H et al (2018) Genome-wide RNAseq study of the molecular mechanisms underlying microglia activation in response to pathological tau perturbation in the rTg4510 tau transgenic animal model. Mol Neurodegener 13:65

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Kang SS et al (2018) Microglial translational profiling reveals a convergent APOE pathway from aging, amyloid, and tau. J Exp Med 215:2235–2245

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Morris G, Berk M, Maes M, Puri BK (2018) Could Alzheimer’s disease originate in the periphery and if so how so? Mol Neurobiol 56(1):406–434

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Boivin G, Coulombe Z, Rivest S (2002) Intranasal herpes simplex virus type 2 inoculation causes a profound thymidine kinase dependent cerebral inflammatory response in the mouse hindbrain. Eur J Neurosci 16:29–43

    Article  PubMed  Google Scholar 

  25. Ilievski V et al (2018) Chronic oral application of a periodontal pathogen results in brain inflammation, neurodegeneration and amyloid beta production in wild type mice. PLoS One 13:e0204941

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Huang C, Irwin MG, Wong GTC, Chang RCC (2018) Evidence of the impact of systemic inflammation on neuroinflammation from a non-bacterial endotoxin animal model. J Neuroinflamm 15:147

    Article  CAS  Google Scholar 

  27. Ishida N et al (2017) Periodontitis induced by bacterial infection exacerbates features of Alzheimer’s disease in transgenic mice. Alzheimer’s Dementia 9:P851

    Article  Google Scholar 

  28. Rakic S et al (2018) Systemic infection modifies the neuroinflammatory response in late stage Alzheimer’s disease. Acta Neuropathol Commun 6:88

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Mucke L, Oldstone MBA (1992) The expression of major histocompatibility complex (MHC) class I antigens in the brain differs markedly in acute and persistent infections with lymphocytic choriomeningitis virus (LCMV). J Neuroimmunol 36:193–198

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Erny D et al (2015) Host microbiota constantly control maturation and function of microglia in the CNS. Nat Neurosci 18:965

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Askew K et al (2017) Coupled proliferation and apoptosis maintain the rapid turnover of microglia in the adult brain. Cell Rep 18:391–405

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Paolicelli RC, Gross CT (2011) Microglia in development: linking brain wiring to brain environment. Neuron Glia Biol 7:77–83

    Article  PubMed  Google Scholar 

  33. Ullian EM, Sapperstein SK, Christopherson KS, Barres BA (2001) Control of synapse number by glia. Science 291:657–661

    Article  CAS  PubMed  Google Scholar 

  34. Whitelaw BS (2017) Microglia-mediated synaptic elimination in neuronal development and disease. J Neurophysiol 119:1–4

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Hanisch U-K, Kettenmann H (2007) Microglia: active sensor and versatile effector cells in the normal and pathologic brain. Nat Neurosci 10:1387

    Article  CAS  PubMed  Google Scholar 

  36. Fuhrmann M et al (2010) Microglial Cx3cr1 knockout prevents neuron loss in a mouse model of Alzheimer’s disease. Nat Neurosci 13:411

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Zheng H, Cheng B, Li Y, Li X, Chen X, Zhang Y-W (2018) TREM2 in Alzheimer’s disease: microglial survival and energy metabolism. Front Aging Neurosci 10:395

    Article  PubMed  PubMed Central  Google Scholar 

  38. Yeh FL, Wang Y, Tom I, Gonzalez LC, Sheng M (2016) TREM2 binds to apolipoproteins, including APOE and CLU/APOJ, and thereby facilitates uptake of amyloid-beta by microglia. Neuron 91:328–340

    Article  CAS  PubMed  Google Scholar 

  39. Rashid K, Wolf A, Langmann T (2018) Microglia activation and immunomodulatory therapies for retinal degenerations. Front Cell Neurosci 12:176

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Rezaie P, Dean A, Male D, Ulfig N (2004) Microglia in the cerebral wall of the human telencephalon at second trimester. Cereb Cortex 15:938–949

    Article  PubMed  Google Scholar 

  41. Yates D (2018) Neurodegenerative disease: a proteostatic boost. Nat Rev Neurosci 19:61

    Article  CAS  PubMed  Google Scholar 

  42. Wang Y, Martinez-Vicente M, Krüger U, Kaushik S, Wong E, Mandelkow E-M, Cuervo AM, Mandelkow E (2009) Tau fragmentation, aggregation and clearance: the dual role of lysosomal processing. Hum Mol Genet 18:4153–4170

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Romero Molina C et al (2018) Distinct microglial responses in two transgenic murine models of TAU pathology. Front Cell Neurosci 12:421

    Article  PubMed  PubMed Central  Google Scholar 

  44. Joseph M, Anglada-Huguet M, Paesler K, Mandelkow E, Mandelkow E-M (2017) Anti-aggregant tau mutant promotes neurogenesis. Mol Neurodegener 12:88

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Dlugosz P, Nimpf J (2018) The reelin receptors apolipoprotein E receptor 2 (ApoER2) and VLDL receptor. Int J Mol Sci 19:3090

    Article  CAS  PubMed Central  Google Scholar 

  46. Bourdenx M, Koulakiotis NS, Sanoudou D, Bezard E, Dehay B, Tsarbopoulos A (2017) Protein aggregation and neurodegeneration in prototypical neurodegenerative diseases: examples of amyloidopathies, tauopathies and synucleinopathies. Prog Neurobiol 155:171–193

    Article  CAS  PubMed  Google Scholar 

  47. Reed MJ et al (2018) Increased hyaluronan and TSG-6 in association with neuropathologic changes of Alzheimer’s disease. J Alzheimer’s Dis 67:91–102

    Article  CAS  Google Scholar 

  48. Wang C, Telpoukhovskaia MA, Bahr BA, Chen X, Gan L (2017) Endo-lysosomal dysfunction: a converging mechanism in neurodegenerative diseases. Curr Opin Neurobiol 48:52–58

    Article  CAS  PubMed  Google Scholar 

  49. Hopp SC, Lin Y, Oakley D, Roe AD, DeVos SL, Hanlon D, Hyman BT (2018) The role of microglia in processing and spreading of bioactive tau seeds in Alzheimer’s disease. J Neuroinflamm 15:269

    Article  CAS  Google Scholar 

  50. Opoku-Nsiah KA, Gestwicki JE (2018) Aim for the core: suitability of the ubiquitin-independent 20S proteasome as a drug target in neurodegeneration. Transl Res 198:48–57

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  51. Neefjes JJ, Momburg F, Hammerling GJ (1993) Selective and ATP-dependent translocation of peptides by the MHC-encoded transporter. Science 261:769–771

    Article  CAS  PubMed  Google Scholar 

  52. Ferrer I, Martín B, Castaño JG, Lucas JJ, Moreno D, Olivé M (2004) Proteasomal expression, induction of immunoproteasome subunits, and local MHC class I presentation in myofibrillar myopathy and inclusion body myositis. J Neuropathol Exp Neurol 63:484–498

    Article  CAS  PubMed  Google Scholar 

  53. Reddy VP, Obrenovich ME, Atwood CS, Perry G, Smith MA (2002) Involvement of Maillard reactions in Alzheimer disease. Neurotox Res 4:191–209

    Article  CAS  PubMed  Google Scholar 

  54. Singh R, Barden A, Mori T, Beilin L (2001) Advanced glycation end-products: a review. Diabetologia 44:129–146

    Article  CAS  PubMed  Google Scholar 

  55. Rothman SM et al (2018) Human Alzheimer’s disease gene expression signatures and immune profile in APP mouse models: a discrete transcriptomic view of Aβ plaque pathology. J Neuroinflamm 15:256

    Article  CAS  Google Scholar 

  56. Patterson SL (2015) Immune dysregulation and cognitive vulnerability in the aging brain: interactions of microglia, IL-1β, BDNF and synaptic plasticity. Neuropharmacology 96:11–18

    Article  CAS  PubMed  Google Scholar 

  57. Colton CA, Wilcock DM (2010) Assessing activation states in microglia. CNS Neurol Disord Drug Targets (Formerly Current Drug Targets-CNS & Neurological Disorders) 9:174–191

    CAS  Google Scholar 

  58. Calvo-Rodríguez M, de la Fuente C, García-Durillo M, García-Rodríguez C, Villalobos C, Núñez L (2017) Aging and amyloid β oligomers enhance TLR4 expression, LPS-induced Ca2+ responses, and neuron cell death in cultured rat hippocampal neurons. J Neuroinflamm 14:24

    Article  CAS  Google Scholar 

  59. Sun Y et al (2017) High-glucose induces tau hyperphosphorylation through activation of TLR9-P38MAPK pathway. Exp Cell Res 359:312–318

    Article  CAS  PubMed  Google Scholar 

  60. Gustin A et al (2015) NLRP3 inflammasome is expressed and functional in mouse brain microglia but not in astrocytes. PLoS One 10:e0130624

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  61. Chan EWL, Krishnansamy S, Wong C, Gan SY (2018) The NLRP3 inflammasome is involved in the neuroprotective mechanism of neural stem cells against microglia-mediated toxicity in SH-SY5Y cells via the attenuation of tau hyperphosphorylation and amyloidogenesis. Neurotoxicology 70:91–98

    Article  CAS  PubMed  Google Scholar 

  62. White CS, Lawrence CB, Brough D, Rivers-Auty J (2017) Inflammasomes as therapeutic targets for A lzheimer’s disease. Brain Pathol 27:223–234

    Article  PubMed  PubMed Central  Google Scholar 

  63. Aso E, Ferrer I (2016) CB2 cannabinoid receptor as potential target against Alzheimer’s disease. Front Neurosci 10:243

    Article  PubMed  PubMed Central  Google Scholar 

  64. Mustaly-Kalimi S, Littlefield AM, Stutzmann GE (2018) Calcium signaling deficits in glia and autophagic pathways contributing to neurodegenerative disease. Antioxid Redox Signal 29:1158–1175

    Article  CAS  PubMed  Google Scholar 

  65. Chiarini A, Armato U, Liu D, Dal Prà I (2016) Calcium-sensing receptors of human neural cells play crucial roles in Alzheimer’s disease. Front Physiol 7:134

    Article  PubMed  PubMed Central  Google Scholar 

  66. Lee M, McGeer E, McGeer PL (2015) Activated human microglia stimulate neuroblastoma cells to upregulate production of beta amyloid protein and tau: implications for Alzheimer’s disease pathogenesis. Neurobiol Aging 36:42–52

    Article  CAS  PubMed  Google Scholar 

  67. Perea JR, Ãvila J, Bolós M (2018) Dephosphorylated rather than hyperphosphorylated Tau triggers a pro-inflammatory profile in microglia through the p38 MAPK pathway. Exp Neurol 310:14–21

    Article  CAS  PubMed  Google Scholar 

  68. Franco Fernandez R et al (2018) N-Methyl-d-aspartate receptor link to the MAP kinase pathway in cortical and hippocampal neurons and microglia is dependent on calcium sensors and is blocked by α-synuclein, tau, and phospho-tau in non-transgenic and transgenic APPSw. Ind mice. Front Pharmacol 11:273

    Google Scholar 

  69. Maphis N et al (2016) Selective suppression of the α isoform of p38 MAPK rescues late-stage tau pathology. Alzheimer’s Res Ther 8:54

    Article  CAS  Google Scholar 

  70. Perea JR, Lleó A, Alcolea D, Fortea J, Ãvila J, Bolós M (2018) Decreased CX3CL1 levels in the cerebrospinal fluid of patients with Alzheimer’s disease. Front Neurosci 12:609

    Article  PubMed  PubMed Central  Google Scholar 

  71. Bemiller SM et al (2018) Genetically enhancing the expression of chemokine domain of CX3CL1 fails to prevent tau pathology in mouse models of tauopathy. J Neuroinflamm 15:278

    Article  CAS  Google Scholar 

  72. Finneran DJ, Morgan D, Gordon MN, Nash KR (2018) CNS-wide over expression of fractalkine improves cognitive functioning in a tauopathy model. J Neuroimmune Pharmacol 1–14

  73. Merino JJ, Muñetón-Gómez V, Alvárez M-I, Toledano-Díaz A (2016) Effects of CX3CR73 and fractalkine chemokines in amyloid beta clearance and p-Tau accumulation in Alzheimer’s disease (AD) rodent models: is fractalkine a systemic biomarker for AD? Curr Alzheimer Res 13:403–412

    Article  CAS  PubMed  Google Scholar 

  74. Haure-Mirande J-V et al (2017) Deficiency of TYROBP, an adapter protein for TREM2 and CR74 receptors, is neuroprotective in a mouse model of early Alzheimer’s pathology. Acta Neuropathol 134:769–788

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  75. Wang Y et al (2015) TREM2 lipid sensing sustains the microglial response in an Alzheimer’s disease model. Cell 160:1061–1071

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  76. Leyns CEG et al (2014) TREM2 deficiency attenuates neuroinflammation and protects against neurodegeneration in a mouse model of tauopathy. Proc Natl Acad Sci 114:11524–11529

    Article  CAS  Google Scholar 

  77. Rauchmann B-S, Schneider-Axmann T, Alexopoulos P, Perneczky R, Alzheimer’s Disease Neuroimaging, I (2018) CSF soluble TREM2 as a measure of immune response along the Alzheimer’s disease continuum. Neurobiol Aging 74:182–190

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  78. Henjum K, Quist-Paulsen E, Zetterberg H, Blennow K, Nilsson LNG, Watne LO (2018) CSF sTREM2 in delirium-relation to Alzheimer’s disease CSF biomarkers Aβ42, t-tau and p-tau. J Neuroinflamm 15:304

    Article  CAS  Google Scholar 

  79. Sayed FA et al (2018) Differential effects of partial and complete loss of TREM2 on microglial injury response and tauopathy. Proc Natl Acad Sci 115:10172–10177

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  80. Hebron M, Peyton M, Liu X, Gao X, Wang R, Lonskaya I, Moussa CEH (2017) Discoidin domain receptor inhibition reduces neuropathology and attenuates inflammation in neurodegeneration models. J Neuroimmunol 311:1–9

    Article  CAS  PubMed  Google Scholar 

  81. Jiang T, Zhang Y-D, Gao Q, Ou Z, Gong P-Y, Shi J-Q, Wu L, Zhou J-S (2018) TREM2 ameliorates neuronal Tau pathology through suppression of microglial inflammatory response. Inflammation 41:811–823

    Article  CAS  PubMed  Google Scholar 

  82. Litvinchuk A et al (2018) Complement c3ar inactivation attenuates tau pathology and reverses an immune network deregulated in tauopathy models and alzheimer’s disease. Neuron 100:1337–1353

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  83. Dejanovic B et al (2018) Changes in the synaptic proteome in tauopathy and rescue of tau-induced synapse loss by C1q antibodies. Neuron 100:1322–1336

    Article  CAS  PubMed  Google Scholar 

  84. Streit WJ, Sammons NW, Kuhns AJ, Sparks DL (2004) Dystrophic microglia in the aging human brain. Glia 45:208–212

    Article  PubMed  Google Scholar 

  85. Streit WJ (2006) Microglial senescence: does the brain’s immune system have an expiration date? Trends Neurosci 29:506–510

    Article  CAS  PubMed  Google Scholar 

  86. Mosher KI, Wyss-Coray T (2014) Microglial dysfunction in brain aging and Alzheimer’s disease. Biochem Pharmacol 88:594–604

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  87. Elmore MRP et al (2018) Replacement of microglia in the aged brain reverses cognitive, synaptic, and neuronal deficits in mice. Aging Cell 17:e12832

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  88. Gomez CR, Nomellini V, Faunce DE, Kovacs EJ (2008) Innate immunity and aging. Exp Gerontol 43:718–728

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  89. Perry VH, Matyszak MK, Fearn S (1993) Altered antigen expression of microglia in the aged rodent CNS. Glia 7:60–67

    Article  CAS  PubMed  Google Scholar 

  90. Streit WJ, Braak H, Xue Q-S, Bechmann I (2009) Dystrophic (senescent) rather than activated microglial cells are associated with tau pathology and likely precede neurodegeneration in Alzheimer’s disease. Acta Neuropathol 118:475–485

    Article  PubMed  PubMed Central  Google Scholar 

  91. Boche D, Nicoll JAR (2008) SYMPOSIUM: clearance of Aβ from the brain in Alzheimer’ disease: the role of the immune system in clearance of Aβ from the brain. Brain Pathol 18:267–278

    Article  PubMed  PubMed Central  Google Scholar 

  92. Elmore MRP et al (2014) Colony-stimulating factor 1 receptor signaling is necessary for microglia viability, unmasking a microglia progenitor cell in the adult brain. Neuron 82:380–397

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  93. Cronk JC et al (2018) Peripherally derived macrophages can engraft the brain independent of irradiation and maintain an identity distinct from microglia. J Exp Med 215:1627–1647

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  94. Viola GG et al (2009) Morphological changes in hippocampal astrocytes induced by environmental enrichment in mice. Brain Res 1274:47–54

    Article  CAS  PubMed  Google Scholar 

  95. Velasco-Estevez M et al (2018) Infection augments expression of mechanosensing Piezo1 channels in amyloid plaque-reactive astrocytes. Front Aging Neurosci 10:332

    Article  PubMed  PubMed Central  Google Scholar 

  96. Liddelow SA et al (2017) Neurotoxic reactive astrocytes are induced by activated microglia. Nature 541:481

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  97. Sofroniew MV (2015) Astrocyte barriers to neurotoxic inflammation. Nat Rev Neurosci 16:249

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  98. Kamphuis W, Kooijman L, Orre M, Stassen O, Pekny M, Hol EM (2015) GFAP and vimentin deficiency alters gene expression in astrocytes and microglia in wild-type mice and changes the transcriptional response of reactive glia in mouse model for Alzheimer’s disease. Glia 63:1036–1056

    Article  PubMed  Google Scholar 

  99. Sharma S, Khadimallah I, Corya AW, Ali YO, Rao X, Liu Y, Lu H-C (2018) Presymptomatic change in microRNAs modulates Tau pathology. Sci Rep 8:9251

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  100. Endo F et al (2015) Astrocyte-derived TGF-β accelerates disease progression in ALS mice by interfering with the neuroprotective functions of microglia and T cells. Cell Rep 11:592–604

    Article  CAS  PubMed  Google Scholar 

  101. Schecter RW, Maher EE, Welsh CA, Stevens B, Erisir A, Bear MF (2017) Experience-dependent synaptic plasticity in V1 occurs without microglial CX3CR101. J Neurosci 37:10541–10553

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  102. Jo S et al (2014) GABA from reactive astrocytes impairs memory in mouse models of Alzheimer’s disease. Nat Med 20:886

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  103. Rustenhoven J et al (2016) TGF-beta1 regulates human brain pericyte inflammatory processes involved in neurovasculature function. J Neuroinflamm 13:37

    Article  CAS  Google Scholar 

  104. Zhou J, Du T, Li B, Rong Y, Verkhratsky A, Peng L (2015) Crosstalk between MAPK/ERK and PI3K/AKT signal pathways during brain ischemia/reperfusion. Asn Neuro 7:1759091415602463

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  105. Brelstaff J, Tolkovsky AM, Ghetti B, Goedert M, Spillantini MG (2018) Living neurons with tau filaments aberrantly expose phosphatidylserine and are phagocytosed by microglia. Cell Rep 24(1939–1948):e4

    Google Scholar 

  106. Lian H, Litvinchuk A, Chiang ACA, Aithmitti N, Jankowsky JL, Zheng H (2016) Astrocyte-microglia cross talk through complement activation modulates amyloid pathology in mouse models of Alzheimer’s disease. J Neurosci 36:577–589

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  107. Torika N, Asraf K, Roasso E, Danon A, Fleisher-Berkovich S (2016) Angiotensin converting enzyme inhibitors ameliorate brain inflammation associated with microglial activation: possible implications for Alzheimer’s disease. J Neuroimmune Pharmacol 11:774–785

    Article  PubMed  Google Scholar 

  108. Kobayashi M, Tamari K, Al Salihi MO, Nishida K, Takeuchi K (2018) Anti-high mobility group box 1 antibody suppresses local inflammatory reaction and facilitates olfactory nerve recovery following injury. J Neuroinflamm 15:124

    Article  CAS  Google Scholar 

  109. Park J, Wetzel I, Marriott I, Dréau D, D’Avanzo C, Kim DY, Tanzi RE, Cho H (2018) A 3D human triculture system modeling neurodegeneration and neuroinflammation in Alzheimer’s disease. Nat Neurosci 21:941

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  110. Bonham LW et al (2018) CXCR110 involvement in neurodegenerative diseases. Transl Psychiatry 8:73

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  111. Guedes JR, Lao T, Cardoso AL, El Khoury J (2018) Roles of microglial and monocyte chemokines and their receptors in regulating alzheimer’s Disease-associated amyloid-β and Tau pathologies. Front Neurol 9:549

    Article  PubMed  PubMed Central  Google Scholar 

  112. Zhu C, Xu B, Sun X, Zhu Q, Sui Y (2017) Targeting CCR112 to reduce amyloid-β production, Tau hyperphosphorylation, and synaptic loss in a mouse model of Alzheimer’s disease. Mol Neurobiol 54:7964–7978

    Article  CAS  PubMed  Google Scholar 

  113. Wolf Y et al (2018) Microglial MHC class II is dispensable for experimental autoimmune encephalomyelitis and cuprizone-induced demyelination. Eur J Immunol 48(8):1308–1318

    Article  CAS  PubMed  Google Scholar 

  114. Walker DG et al (2017) Changes in CD200 and intercellular adhesion molecule-1 (ICAM-1) levels in brains of Lewy body disorder cases are associated with amounts of Alzheimer’s pathology not α-synuclein pathology. Neurobiol Aging 54:175–186

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  115. Janelidze S, Mattsson N, Stomrud E, Lindberg O, Palmqvist S, Zetterberg H, Blennow K, Hansson O (2018) CSF biomarkers of neuroinflammation and cerebrovascular dysfunction in early Alzheimer disease. Neurology 91:e867–e877

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  116. Waisman A, Johann L (2018) Antigen-presenting cell diversity for T cell reactivation in central nervous system autoimmunity. J Mol Med 96:1279–1292

    Article  CAS  PubMed  Google Scholar 

  117. Hutter-Schmid B, Humpel C (2018) Primary mouse brain pericytes isolated from transgenic Alzheimer mice spontaneously differentiate into a CD11b+ microglial-like cell type in vitro. Exp Gerontol 112:30–37

    Article  CAS  PubMed  Google Scholar 

  118. Johnson DB et al (2016) Melanoma-specific MHC-II expression represents a tumour-autonomous phenotype and predicts response to anti-PD-1/PD-L1 therapy. Nat Commun 7:10582

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  119. Wlodarczyk A, Løbner M, Cédile O, Owens T (2014) Comparison of microglia and infiltrating CD11c+ cells as antigen presenting cells for T cell proliferation and cytokine response. J Neuroinflamm 11:57

    Article  CAS  Google Scholar 

  120. Rangachari M, Kerfoot SM, Arbour N, Alvarez JI (2017) Lymphocytes in MS and EAE: more than just a CD4+ world. Front Immunol 8:133

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  121. David S, Kroner A, Greenhalgh AD, Zarruk JG, López-Valesd R (2018) Myeloid cell responses after spinal cord injury. J Neuroimmunol 321:97–108

    Article  CAS  PubMed  Google Scholar 

  122. Wilshusen RA, Mosley RL (2016) Exacerbation of MPTP induced neurodegeneration by a CD4+ effector T cell line specific for N-α-synuclein in a mouse model of Parkinson’s disease. Am Assoc Immnol 196:49.7

    Google Scholar 

  123. Beers DR et al (2017) ALS patients’ regulatory T lymphocytes are dysfunctional, and correlate with disease progression rate and severity. JCI Insight 2(5):e89530

    Article  PubMed  PubMed Central  Google Scholar 

  124. Lueg G et al (2015) Clinical relevance of specific T-cell activation in the blood and cerebrospinal fluid of patients with mild Alzheimer’s disease. Neurobiol Aging 36:81–89

    Article  CAS  PubMed  Google Scholar 

  125. Baruch K et al (2015) Breaking immune tolerance by targeting Foxp3+ regulatory T cells mitigates Alzheimer’s disease pathology. Nat Commun 6:7967

    Article  CAS  PubMed  Google Scholar 

  126. Rosset MB, Lui G, Dansokho C, Chaigneau T, Dorothée G (2015) Vaccine-induced Aβ-specific CD8+ T cells do not trigger autoimmune neuroinflammation in a murine model of Alzheimer’s disease. J Neuroinflamm 12:95

    Article  CAS  Google Scholar 

  127. Oberstein TJ, Taha L, Spitzer P, Hellstern J, Herrmann M, Kornhuber J, Maler JM (2018) Imbalance of circulating Th17 and regulatory T cells in Alzheimer’s disease: a case control study. Front Immunol 9:1213

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  128. Gendelman HE, Mosley RL (2015) A perspective on roles played by innate and adaptive immunity in the pathobiology of neurodegenerative disorders. J Neuroimmune Pharmacol 10:645–650

    Article  PubMed  PubMed Central  Google Scholar 

  129. Ellwardt E, Walsh JT, Kipnis J, Zipp F (2016) Understanding the role of T cells in CNS homeostasis. Trends Immunol 37:154–165

    Article  CAS  PubMed  Google Scholar 

  130. Walsh JT, Zheng J, Smirnov I, Lorenz U, Tung K, Kipnis J (2014) Regulatory T cells in central nervous system injury: a double-edged sword. J Immunol 193:5013–5022

    Article  CAS  PubMed  Google Scholar 

  131. Obregon D et al (2008) CD40L disruption enhances Abeta vaccine-mediated reduction of cerebral amyloidosis while minimizing cerebral amyloid angiopathy and inflammation. Neurobiol Dis 29:336–353

    Article  CAS  PubMed  Google Scholar 

  132. Tang Y, Le W (2016) Differential roles of M1 and M2 microglia in neurodegenerative diseases. Mol Neurobiol 53:1181–1194

    Article  CAS  PubMed  Google Scholar 

  133. Rőszer T (2015) Understanding the mysterious M2 macrophage through activation markers and effector mechanisms. Mediat Inflamm 2015:816460

    Article  CAS  Google Scholar 

  134. Celada LJ et al (2018) PD-1 up-regulation on CD4+ T cells promotes pulmonary fibrosis through STAT3-mediated IL-17A and TGF-β1 production. Sci Transl Med 10:8356

    Article  CAS  Google Scholar 

  135. Réus GZ, Fries GR, Stertz L, Badawy M, Passos IC, Barichello T, Kapczinski F, Quevedo J (2015) The role of inflammation and microglial activation in the pathophysiology of psychiatric disorders. Neuroscience 300:141–154

    Article  CAS  PubMed  Google Scholar 

  136. De Marco D, Taggenbrock R, Crespo R, Koudstaal W, Ramsburg E, Apetri A (2018) Cell-based assay to study antibody-mediated tau clearance by microglia. JoVE J Vis Exp (141):e58576

  137. Florez-Pollack S, Tseng L-C, Kobayashi M, Hosler GA, Ariizumi K, Chong BF (2018) Expansion of myeloid-derived suppressor cells in the peripheral blood and lesional skin of cutaneous lupus patients. J Investig Dermatol 139(2):478–481

    Article  CAS  PubMed  Google Scholar 

  138. Hu J et al (2016) Programmed death ligand-1 on microglia regulates Th1 differentiation via nitric oxide in experimental autoimmune encephalomyelitis. Neurosci Bull 32:70–82

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  139. Villa-Álvarez M, Lorenzo-Herrero S, Gonzalez-Rodriguez AP, López-Soto A, Payer AR, Gonzalez-Garcia E, Huergo-Zapico L, Gonzalez S (2017) Ig-like transcript 2 (ILT2) suppresses T cell function in chronic lymphocytic leukemia. OncoImmunology 6:e1353856

    Article  PubMed  PubMed Central  Google Scholar 

  140. Burel JG, Apte SH, Groves PL, McCarthy JS, Doolan DL (2017) Polyfunctional and IFN-γ monofunctional human CD4+ T cell populations are molecularly distinct. JCI Insight 2:e87499

    Article  PubMed  PubMed Central  Google Scholar 

  141. Bennett RE, Bryant A, Hu M, Robbins AB, Hopp SC, Hyman BT (2018) Partial reduction of microglia does not affect tau pathology in aged mice. J Neuroinflamm 15:311

    Article  CAS  Google Scholar 

  142. Njie EG, Boelen E, Stassen FR, Steinbusch HW, Borchelt DR, Streit WJ (2012) Ex vivo cultures of microglia from young and aged rodent brain reveal age-related changes in microglial function. Neurobiol Aging 33:195.e1–195.e12

    Article  CAS  Google Scholar 

  143. Bussian TJ, Aziz A, Meyer CF, Swenson BL, van Deursen JM, Baker DJ (2018) Clearance of senescent glial cells prevents tau-dependent pathology and cognitive decline. Nature 562:578

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  144. Zhang F, Zhong R, Li S, Fu Z, Cheng C, Cai H, Le W (2017) Acute hypoxia induced an imbalanced M1/M2 activation of microglia through NF-κB signaling in Alzheimer’s disease mice and wild-type littermates. Front Aging Neurosci 9:282

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  145. Shapira R, Solomon B, Efrati S, Frenkel D, Ashery U (2017) Hyperbaric oxygen therapy ameliorates pathophysiology of 3xTg-AD mouse model by attenuating neuroinflammation. Neurobiol Aging 62:105–119

    Article  CAS  PubMed  Google Scholar 

  146. He J, Liao T, Zhong G-X, Zhang J-D, Chen Y-P, Wang Q, Zeng Q-P (2017) Alzheimer’s disease-like early-phase brain pathogenesis: self-curing amelioration of neurodegeneration from pro-inflammatory wounding-to anti-inflammatory-healing. Curr Alzheimer Res 14:1123–1135

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

This project is supported in part by grants from the Department of Biotechnology, Neuroscience Task Force (Medical Biotechnology-Human Development & Disease Biology DBT-HDDB)-BT/PR/15780/MED/122/4/2016 and in-house CSIR-National Chemical Laboratory Grant MLP029526. Special thanks to Ms. Nalini Gorantla, Mr. Abhishek Balmik and Ms. Shweta Sonawane for proofreading the manuscript. Rashmi Das acknowledges the fellowship from University Grant Commission (UGC) India.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Subashchandrabose Chinnathambi.

Ethics declarations

Conflict of interest

The authors declare no conflict of interest.

Additional information

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Das, R., Chinnathambi, S. Microglial priming of antigen presentation and adaptive stimulation in Alzheimer’s disease. Cell. Mol. Life Sci. 76, 3681–3694 (2019). https://doi.org/10.1007/s00018-019-03132-2

Download citation

  • Received:

  • Revised:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1007/s00018-019-03132-2

Keywords

Navigation