1932

Abstract

Genetic studies of autosomal dominant Alzheimer's disease (AD) revealed that β-amyloid is central to disease pathogenesis. However, amyloid-targeted therapies have generally failed to slow progression in patients with symptomatic disease. This result suggests a transition from an early amyloid-dependent phase to a later amyloid-independent one, during which neurodegeneration occurs and symptoms arise. Microglia, the brain's resident myeloid cells, envelop amyloid and express the majority of genes linked to risk for sporadic late-onset AD. Their activation is associated spatially and temporally with the accumulation of pathological tau. Microglial facilitation of tau pathology may involve apolipoprotein E, the most important genetic risk factor for AD. Once formed, pathological tau spreads between connected neurons, eventually accumulating in the somatic compartment where catastrophic nuclear damage ensues. This emerging understanding of the postamyloid processes leading to neurodegeneration affords the opportunity to develop therapeutics that interrupt this pathological cascade and prevent or delay dementia, even after amyloid deposition.

Loading

Article metrics loading...

/content/journals/10.1146/annurev-med-052919-120219
2021-01-27
2024-04-24
Loading full text...

Full text loading...

/deliver/fulltext/med/72/1/annurev-med-052919-120219.html?itemId=/content/journals/10.1146/annurev-med-052919-120219&mimeType=html&fmt=ahah

Literature Cited

  1. 1. 
    Alzheimer's Association 2020. 2020 Alzheimer's disease facts and figures. Alzheimer's Dement 16:391–460
    [Google Scholar]
  2. 2. 
    Cummings J, Lee G, Ritter A et al. 2019. Alzheimer's disease drug development pipeline: 2019. Alzheimer's Dement 5:272–93
    [Google Scholar]
  3. 3. 
    Selkoe DJ, Hardy J. 2016. The amyloid hypothesis of Alzheimer's disease at 25 years. EMBO Mol. Med. 8:595–608
    [Google Scholar]
  4. 4. 
    Hardy J, De Strooper B 2017. Alzheimer's disease: where next for anti-amyloid therapies. Brain 140:853–55
    [Google Scholar]
  5. 5. 
    Egan MF, Kost J, Tariot PN et al. 2018. Randomized trial of verubecestat for mild-to-moderate Alzheimer's disease. N. Engl. J. Med. 378:1691–703
    [Google Scholar]
  6. 6. 
    Sevigny J, Chiao P, Bussière T et al. 2016. The antibody aducanumab reduces Aβ plaques in Alzheimer's disease. Nature 537:50–56
    [Google Scholar]
  7. 7. 
    Rogers MB. 2019. ‘Reports of my death are greatly exaggerated.’ Signed, aducanumab. Alzforum https://www.alzforum.org/news/research-news/reports-my-death-are-greatly-exaggerated-signed-aducanumab
    [Google Scholar]
  8. 8. 
    Arboleda-Velasquez JF, Lopera F, O'Hare M et al. 2019. Resistance to autosomal dominant Alzheimer's disease in an APOE3 Christchurch homozygote: a case report. Nat. Med. 25:1680–83
    [Google Scholar]
  9. 9. 
    Yamazaki Y, Painter MM, Bu G, Kanekiyo T 2016. Apolipoprotein E as a therapeutic target in Alzheimer's disease: a review of basic research and clinical evidence. CNS Drugs 30:773–89
    [Google Scholar]
  10. 10. 
    Lee SH, Harold D, Nyholt DR et al. 2013. Estimation and partitioning of polygenic variation captured by common SNPs for Alzheimer's disease, multiple sclerosis and endometriosis. Hum. Mol. Genet. 22:832–41
    [Google Scholar]
  11. 11. 
    Efthymiou AG, Goate AM. 2017. Late onset Alzheimer's disease genetics implicates microglial pathways in disease risk. Mol. Neurodegener. 12:43
    [Google Scholar]
  12. 12. 
    Li Q, Barres BA. 2018. Microglia and macrophages in brain homeostasis and disease. Nat. Rev. Immunol. 18:225–42
    [Google Scholar]
  13. 13. 
    Krasemann S, Madore C, Cialic R et al. 2017. The TREM2-APOE pathway drives the transcriptional phenotype of dysfunctional microglia in neurodegenerative diseases. Immunity 47:566–81.e9
    [Google Scholar]
  14. 14. 
    Keren-Shaul H, Spinrad A, Weiner A et al. 2017. A unique microglia type associated with restricting development of Alzheimer's disease. Cell 169:1276–90.e17
    [Google Scholar]
  15. 15. 
    Rangaraju S, Dammer EB, Raza SA et al. 2018. Quantitative proteomics of acutely-isolated mouse microglia identifies novel immune Alzheimer's disease-related proteins. Mol. Neurodegener. 13:34
    [Google Scholar]
  16. 16. 
    Perry VH, Nicoll JAR, Holmes C 2010. Microglia in neurodegenerative disease. Nat. Rev. Neurol. 6:193–201
    [Google Scholar]
  17. 17. 
    Holtman IR, Raj DD, Miller JA et al. 2015. Induction of a common microglia gene expression signature by aging and neurodegenerative conditions: a co-expression meta-analysis. Acta Neuropathol. Commun. 3:31
    [Google Scholar]
  18. 18. 
    Shirotani K, Hori Y, Yoshizaki R et al. 2019. Aminophospholipids are signal-transducing TREM2 ligands on apoptotic cells. Sci. Rep. 9:7508
    [Google Scholar]
  19. 19. 
    Pottier C, Ravenscroft TA, Brown PH et al. 2016. TYROBP genetic variants in early-onset Alzheimer's disease. Neurobiol. Aging 48:222.e9–e15
    [Google Scholar]
  20. 20. 
    Lambert JC, Ibrahim-Verbaas CA, Harold D et al. 2013. Meta-analysis of 74,046 individuals identifies 11 new susceptibility loci for Alzheimer's disease. Nat. Genet. 45:1452–58
    [Google Scholar]
  21. 21. 
    Thornton P, Sevalle J, Deery MJ et al. 2017. TREM2 shedding by cleavage at the H157-S158 bond is accelerated for the Alzheimer's disease-associated H157Y variant. EMBO Mol. Med. 9:1366–78
    [Google Scholar]
  22. 22. 
    Paloneva J, Manninen T, Christman G et al. 2002. Mutations in two genes encoding different subunits of a receptor signaling complex result in an identical disease phenotype. Am. J. Hum. Genet. 71:656–62
    [Google Scholar]
  23. 23. 
    Ewers M, Franzmeier N, Suárez-Calvet M et al. 2019. Increased soluble TREM2 in cerebrospinal fluid is associated with reduced cognitive and clinical decline in Alzheimer's disease. Sci. Transl. Med. 11:eaav6221
    [Google Scholar]
  24. 24. 
    Ulland TK, Song WM, Huang SC et al. 2017. TREM2 maintains microglial metabolic fitness in Alzheimer's disease. Cell 170:649–63.e13
    [Google Scholar]
  25. 25. 
    Yuan P, Condello C, Keene CD et al. 2016. TREM2 haplodeficiency in mice and humans impairs the microglia barrier function leading to decreased amyloid compaction and severe axonal dystrophy. Neuron 90:724–39
    [Google Scholar]
  26. 26. 
    Leyns CEG, Gratuze M, Narasimhan S et al. 2019. TREM2 function impedes tau seeding in neuritic plaques. Nat. Neurosci. 22:1217–22
    [Google Scholar]
  27. 27. 
    Alector 2019. Alector R&D Day, Dec. 13, New York, NY. https://investors.alector.com/static-files/7418b689-c5b7-43ac-a16a-3c64e1a14e80
  28. 28. 
    Kariolis MS, Wells RC, Getz JA et al. 2020. Brain delivery of therapeutic proteins using an Fc fragment blood-brain barrier transport vehicle in mice and monkeys. Sci. Transl. Med. 12:eaay1359
    [Google Scholar]
  29. 29. 
    Schlepckow K, Monroe KM, Kleinberger G et al. 2020. Enhancing protective microglial activities with a dual function TREM2 antibody to the stalk region. EMBO Mol. Med. 12:e11227
    [Google Scholar]
  30. 30. 
    Götzl JK, Brendel M, Werner G et al. 2019. Opposite microglial activation stages upon loss of PGRN or TREM2 result in reduced cerebral glucose metabolism. EMBO Mol. Med. 11:e9711
    [Google Scholar]
  31. 31. 
    Hollingworth P, Harold D, Sims R et al. 2011. Common variants at ABCA7, MS4A6A/MS4A4E, EPHA1, CD33 and CD2AP are associated with Alzheimer's disease. Nat. Genet. 43:429–35
    [Google Scholar]
  32. 32. 
    Griciuc A, Patel S, Federico AN et al. 2019. TREM2 acts downstream of CD33 in modulating microglial pathology in Alzheimer's disease. Neuron 103:820–35.e7
    [Google Scholar]
  33. 33. 
    Miles LA, Hermans SJ, Crespi GAN et al. 2019. Small molecule binding to Alzheimer risk factor CD33 promotes Aβ phagocytosis. iScience 19:110–18
    [Google Scholar]
  34. 34. 
    Novikova G, Kapoor M, TCW J et al. 2019. Integration of Alzheimer's disease genetics and myeloid cell genomics identifies novel causal variants, regulatory elements, genes and pathways. bioRxiv 694281. https://doi.org/10.1101/694281
    [Crossref]
  35. 35. 
    Deming Y, Filipello F, Cignarella F et al. 2019. The MS4A gene cluster is a key modulator of soluble TREM2 and Alzheimer's disease risk. Sci. Transl. Med. 11:eaau2291
    [Google Scholar]
  36. 36. 
    Greer PL, Bear DM, Lassance JM et al. 2016. A family of non-GPCR chemosensors defines an alternative logic for mammalian olfaction. Cell 165:1734–48
    [Google Scholar]
  37. 37. 
    Elmore MR, Najafi AR, Koike MA et al. 2014. Colony-stimulating factor 1 receptor signaling is necessary for microglia viability, unmasking a microglia progenitor cell in the adult brain. Neuron 82:380–97
    [Google Scholar]
  38. 38. 
    Spangenberg EE, Green KN. 2017. Inflammation in Alzheimer's disease: lessons learned from microglia-depletion models. Brain Behav. Immun. 61:1–11
    [Google Scholar]
  39. 39. 
    Mancuso R, Fryatt G, Cleal M et al. 2019. CSF1R inhibitor JNJ-40346527 attenuates microglial proliferation and neurodegeneration in P301S mice. Brain 142:3243–64
    [Google Scholar]
  40. 40. 
    Halle A, Hornung V, Petzold GC et al. 2008. The NALP3 inflammasome is involved in the innate immune response to amyloid-beta. Nat. Immunol. 9:857–65
    [Google Scholar]
  41. 41. 
    Stancu IC, Cremers N, Vanrusselt H et al. 2019. Aggregated tau activates NLRP3-ASC inflammasome exacerbating exogenously seeded and non-exogenously seeded Tau pathology in vivo. Acta Neuropathol 137:599–617
    [Google Scholar]
  42. 42. 
    Heneka MT, Kummer MP, Stutz A et al. 2013. NLRP3 is activated in Alzheimer's disease and contributes to pathology in APP/PS1 mice. Nature 493:674–78
    [Google Scholar]
  43. 43. 
    Ising C, Venegas C, Zhang S et al. 2019. NLRP3 inflammasome activation drives tau pathology. Nature 575:669–73
    [Google Scholar]
  44. 44. 
    Coll RC, Robertson AA, Chae JJ et al. 2015. A small-molecule inhibitor of the NLRP3 inflammasome for the treatment of inflammatory diseases. Nat. Med. 21:248–55
    [Google Scholar]
  45. 45. 
    Jin H, Han J, Resing D et al. 2018. Synthesis and in vitro characterization of a P2X7 radioligand [123I]TZ6019 and its response to neuroinflammation in a mouse model of Alzheimer disease. Eur. J. Pharmacol. 820:8–17
    [Google Scholar]
  46. 46. 
    Degterev A, Ofengeim D, Yuan J 2019. Targeting RIPK1 for the treatment of human diseases. PNAS 116:9714–22
    [Google Scholar]
  47. 47. 
    Weisel K, Berger S, Papp K et al. 2020. Response to inhibition of receptor-interacting protein kinase 1 (RIPK1) in active plaque psoriasis: a randomized placebo-controlled study. Clin. Pharmacol. Ther 108:808–16
    [Google Scholar]
  48. 48. 
    Ofengeim D, Mazzitelli S, Ito Y et al. 2017. RIPK1 mediates a disease-associated microglial response in Alzheimer's disease. PNAS 114:E8788–97
    [Google Scholar]
  49. 49. 
  50. 50. 
    Itzhaki RF. 2017. Herpes simplex virus type 1 and Alzheimer's disease: possible mechanisms and signposts. FASEB J 31:3216–26
    [Google Scholar]
  51. 51. 
    Eimer WA, Vijaya Kumar DK, Navalpur Shanmugam NK et al. 2018. Alzheimer's disease-associated β-amyloid is rapidly seeded by Herpesviridae to protect against brain infection. Neuron 99:56–63.e3
    [Google Scholar]
  52. 52. 
    Readhead B, Haure-Mirande JV, Funk CC et al. 2018. Multiscale analysis of independent Alzheimer's cohorts finds disruption of molecular, genetic, and clinical networks by human herpesvirus. Neuron 99:64–82.e7
    [Google Scholar]
  53. 53. 
    Ashraf GM, Tarasov VV, Makhmutovа A et al. 2019. The possibility of an infectious etiology of Alzheimer disease. Mol. Neurobiol. 56:4479–91
    [Google Scholar]
  54. 54. 
    Dominy SS, Lynch C, Ermini F et al. 2019. Porphyromonas gingivalis in Alzheimer's disease brains: evidence for disease causation and treatment with small-molecule inhibitors. Sci. Adv. 5:eaau3333
    [Google Scholar]
  55. 55. 
    Fries P, Nikolić D, Singer W 2007. The gamma cycle. Trends Neurosci 30:309–16
    [Google Scholar]
  56. 56. 
    Verret L, Mann EO, Hang GB et al. 2012. Inhibitory interneuron deficit links altered network activity and cognitive dysfunction in Alzheimer model. Cell 149:708–21
    [Google Scholar]
  57. 57. 
    Martorell AJ, Paulson AL, Suk HJ et al. 2019. Multi-sensory gamma stimulation ameliorates Alzheimer's-associated pathology and improves cognition. Cell 177:256–71.e22
    [Google Scholar]
  58. 58. 
    Liu L, Drouet V, Wu JW et al. 2012. Trans-synaptic spread of tau pathology in vivo. PLOS ONE 7:e31302
    [Google Scholar]
  59. 59. 
    Vogel JW, Iturria-Medina Y, Strandberg OT et al. 2020. Spread of pathological tau proteins through communicating neurons in human Alzheimer's disease. Nat. Commun. 11:2612
    [Google Scholar]
  60. 60. 
    Duka V, Lee JH, Credle J et al. 2013. Identification of the sites of tau hyperphosphorylation and activation of tau kinases in synucleinopathies and Alzheimer's diseases. PLOS ONE 8:e75025
    [Google Scholar]
  61. 61. 
    Liu F, Shi J, Tanimukai H et al. 2009. Reduced O-GlcNAcylation links lower brain glucose metabolism and tau pathology in Alzheimer's disease. Brain 132:1820–32
    [Google Scholar]
  62. 62. 
    Selnick HG, Hess JF, Tang C et al. 2019. Discovery of MK-8719, a potent O-GlcNAcase inhibitor as a potential treatment for tauopathies. J. Med. Chem. 62:10062–97
    [Google Scholar]
  63. 63. 
    Chen JL, Zhang P, Abe M et al. 2020. Design, optimization, and study of small molecules that target tau pre-mRNA and affect splicing. J. Am. Chem. Soc. 142:8706–27
    [Google Scholar]
  64. 64. 
    Kaufman SK, Sanders DW, Thomas TL et al. 2016. Tau prion strains dictate patterns of cell pathology, progression rate, and regional vulnerability in vivo. Neuron 92:796–812
    [Google Scholar]
  65. 65. 
    Schroeder SK, Joly-Amado A, Gordon MN, Morgan D 2016. Tau-directed immunotherapy: a promising strategy for treating Alzheimer's disease and other tauopathies. J. Neuroimmune Pharmacol. 11:9–25
    [Google Scholar]
  66. 66. 
    Kontsekova E, Zilka N, Kovacech B et al. 2014. First-in-man tau vaccine targeting structural determinants essential for pathological tau-tau interaction reduces tau oligomerisation and neurofibrillary degeneration in an Alzheimer's disease model. Alzheimer's Res. Ther. 6:44
    [Google Scholar]
  67. 67. 
    Lee SH, Le Pichon CE, Adolfsson O et al. 2016. Antibody-mediated targeting of tau in vivo does not require effector function and microglial engagement. Cell Rep 16:1690–700
    [Google Scholar]
  68. 68. 
    Alam R, Driver D, Wu S et al. 2017. Preclinical characterization of an antibody (LY3303560) targeting aggregated tau. Poster O2–14–05. Alzheimer's Dement 13:P592–P593
    [Google Scholar]
  69. 69. 
    Alzforum 2018. To block tau's proteopathic spread, antibody must attack its mid-region. https://www.alzforum.org/news/conference-coverage/block-taus-proteopathic-spread-antibody-must-attack-its-mid-region
  70. 70. 
    Holmes BB, DeVos SL, Kfoury N et al. 2013. Heparan sulfate proteoglycans mediate internalization and propagation of specific proteopathic seeds. PNAS 110:E3138–47
    [Google Scholar]
  71. 71. 
    Rauch JN, Luna G, Guzman E et al. 2020. LRP1 is a master regulator of tau uptake and spread. Nature 580:381–85
    [Google Scholar]
  72. 72. 
    Petros AM, Korepanova A, Jakob CG et al. 2019. Fragment-based discovery of an apolipoprotein E4 (apoE4) stabilizer. J. Med. Chem. 62:4120–30
    [Google Scholar]
  73. 73. 
    Harada A, Oguchi K, Okabe S et al. 1994. Altered microtubule organization in small-calibre axons of mice lacking tau protein. Nature 369:488–91
    [Google Scholar]
  74. 74. 
    Roberson ED, Scearce-Levie K, Palop JJ et al. 2007. Reducing endogenous tau ameliorates amyloid beta-induced deficits in an Alzheimer's disease mouse model. Science 316:750–54
    [Google Scholar]
  75. 75. 
    Ittner LM, Ke YD, Delerue F et al. 2010. Dendritic function of tau mediates amyloid-beta toxicity in Alzheimer's disease mouse models. Cell 142:387–97
    [Google Scholar]
  76. 76. 
    DeVos SL, Miller RL, Schoch KM et al. 2017. Tau reduction prevents neuronal loss and reverses pathological tau deposition and seeding in mice with tauopathy. Sci. Transl. Med. 9:eaag0481
    [Google Scholar]
  77. 77. 
    Cornelison GL, Levy SA, Jenson T, Frost B 2019. Tau-induced nuclear envelope invagination causes a toxic accumulation of mRNA in Drosophila. . Aging Cell 18:e12847
    [Google Scholar]
  78. 78. 
    Paonessa F, Evans LD, Solanki R et al. 2019. Microtubules deform the nuclear membrane and disrupt nucleocytoplasmic transport in tau-mediated frontotemporal dementia. Cell Rep 26:582–93.e5
    [Google Scholar]
  79. 79. 
    Frost B, Bardai FH, Feany MB 2016. Lamin dysfunction mediates neurodegeneration in tauopathies. Curr. Biol. 26:129–36
    [Google Scholar]
  80. 80. 
    Gonzalo S, Kreienkamp R, Askjaer P 2017. Hutchinson-Gilford progeria syndrome: a premature aging disease caused by LMNA gene mutations. Ageing Res. Rev. 33:18–29
    [Google Scholar]
  81. 81. 
    Frost B. 2016. Alzheimer's disease: an acquired neurodegenerative laminopathy. Nucleus 7:275–83
    [Google Scholar]
  82. 82. 
    Frost B, Hemberg M, Lewis J, Feany MB 2014. Tau promotes neurodegeneration through global chromatin relaxation. Nat. Neurosci. 17:357–66
    [Google Scholar]
  83. 83. 
    Sun W, Samimi H, Gamez M et al. 2018. Pathogenic tau-induced piRNA depletion promotes neuronal death through transposable element dysregulation in neurodegenerative tauopathies. Nat. Neurosci. 21:1038–48
    [Google Scholar]
  84. 84. 
    Guo C, Jeong HH, Hsieh YC et al. 2018. Tau activates transposable elements in Alzheimer's disease. Cell Rep 23:2874–80
    [Google Scholar]
  85. 85. 
    Montagne A, Nation DA, Sagare AP et al. 2020. APOE4 leads to blood-brain barrier dysfunction predicting cognitive decline. Nature 581:71–76
    [Google Scholar]
  86. 86. 
    Halliday MR, Rege SV, Ma Q et al. 2016. Accelerated pericyte degeneration and blood-brain barrier breakdown in apolipoprotein E4 carriers with Alzheimer's disease. J. Cereb. Blood Flow Metab. 36:216–27
    [Google Scholar]
/content/journals/10.1146/annurev-med-052919-120219
Loading
/content/journals/10.1146/annurev-med-052919-120219
Loading

Data & Media loading...

  • Article Type: Review Article
This is a required field
Please enter a valid email address
Approval was a Success
Invalid data
An Error Occurred
Approval was partially successful, following selected items could not be processed due to error