Skip to main content

Advertisement

Log in

The synapse as a treatment avenue for Alzheimer’s Disease

  • Review Article
  • Published:
Molecular Psychiatry Submit manuscript

Abstract

Alzheimer’s disease (AD) is a neurodegenerative disorder with devastating symptoms, including memory impairments and cognitive deficits. Hallmarks of AD pathology are amyloid-beta (Aβ) deposition forming neuritic plaques and neurofibrillary tangles (NFTs). For many years, AD drug development has mainly focused on directly targeting the Aβ aggregation or the formation of tau tangles, but this disease has no cure so far. Other common characteristics of AD are synaptic abnormalities and dysfunctions such as synaptic damage, synaptic loss, and structural changes in the synapse. Those anomalies happen in the early stages of the disease before behavioural symptoms have occurred. Therefore, better understanding the mechanisms underlying the synaptic dysfunction found in AD and targeting the synapse, especially using early treatment windows, can lead to finding novel and more effective treatments that could improve the lives of AD patients. Researchers have recently started developing different disease-modifying treatments targeting the synapse to rescue and prevent synaptic dysfunction in AD. The main objectives of these new strategies are to halt synaptic loss, strengthen synaptic connections, and improve synaptic density, potentially leading to the rescue or prevention of cognitive impairments. This article aims to address the mechanisms of synaptic degeneration in AD and discuss current strategies that focus on the synapse for AD therapy. Alzheimer’s disease (AD) is a neurodegenerative disorder that significantly impairs memory and causes cognitive and behavioural deficits. Scientists worldwide have tried to find a treatment that can reverse or rescue AD symptoms, but there is no cure so far. One prominent characteristic of AD is the brain atrophy caused by significant synaptic loss and overall neuronal damage, which starts at the early stages of the disease before other AD hallmarks such as neuritic plaques and NFTs. The present review addresses the underlying mechanisms behind synaptic loss and dysfunction in AD and discusses potential strategies that target the synapse.

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: Oligomeric Aβ and phosphorylated tau induce neural toxicity causing synaptic loss and dysfunction.
Fig. 2: General representation of the role of microglia, astrocytes and ApoE isoform on abnormal synaptic pruning and synaptic dysfunction.

Similar content being viewed by others

References

  1. Jan LY, Jan YN. L-glutamate as an excitatory transmitter at the Drosophila larval neuromuscular junction. J Physiol. 1976;262:215–36.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  2. Takumi Y, Ramirez-Leon V, Laake P, Rinvik E, Ottersen OP. Different modes of expression of AMPA and NMDA receptors in hippocampal synapses. Nat Neurosci. 1999;2:618–24.

    Article  CAS  PubMed  Google Scholar 

  3. Migaud M, Charlesworth P, Dempster M, Webster LC, Watabe AM, Makhinson M, et al. Enhanced long-term potentiation and impaired learning in mice with mutant postsynaptic density-95 protein. Nature. 1998;396:433–9.

    Article  CAS  PubMed  Google Scholar 

  4. Davies CA, Mann DM, Sumpter PQ, Yates PO. A quantitative morphometric analysis of the neuronal and synaptic content of the frontal and temporal cortex in patients with Alzheimer’s disease. J Neurological Sci. 1987;78:151–64.

    Article  CAS  Google Scholar 

  5. Scheff SW, Price DA, Schmitt FA, Mufson EJ. Hippocampal synaptic loss in early Alzheimer’s disease and mild cognitive impairment. Neurobiol Aging. 2006;27:1372–84.

    Article  CAS  PubMed  Google Scholar 

  6. Mucke L, Masliah E, Yu GQ, Mallory M, Rockenstein EM, Tatsuno G, et al. High-level neuronal expression of abeta 1-42 in wild-type human amyloid protein precursor transgenic mice: synaptotoxicity without plaque formation. J Neurosci. 2000;20:4050–8.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Harris JA, Devidze N, Verret L, Ho K, Halabisky B, Thwin MT, et al. Transsynaptic progression of amyloid-beta-induced neuronal dysfunction within the entorhinal-hippocampal network. Neuron. 2010;68:428–41.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Terry RD, Masliah E, Salmon DP, Butters N, DeTeresa R, Hill R, et al. Physical basis of cognitive alterations in Alzheimer’s disease: synapse loss is the major correlate of cognitive impairment. Ann Neurol. 1991;30:572–80.

    Article  CAS  PubMed  Google Scholar 

  9. Robakis NK, Ramakrishna N, Wolfe G, Wisniewski HM. Molecular cloning and characterization of a cDNA encoding the cerebrovascular and the neuritic plaque amyloid peptides. Proc Natl Acad Sci USA. 1987;84:4190–4.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Tanzi RE, Gusella JF, Watkins PC, Bruns GA, St George-Hyslop P, Van Keuren ML, et al. Amyloid beta protein gene: cDNA, mRNA distribution, and genetic linkage near the Alzheimer locus. Science. 1987;235:880–4.

    Article  CAS  PubMed  Google Scholar 

  11. Kang J, Lemaire HG, Unterbeck A, Salbaum JM, Masters CL, Grzeschik KH, et al. The precursor of Alzheimer’s disease amyloid A4 protein resembles a cell-surface receptor. Nature. 1987;325:733–6.

    Article  CAS  PubMed  Google Scholar 

  12. Goldgaber D, Lerman MI, McBride OW, Saffiotti U, Gajdusek DC. Characterization and chromosomal localization of a cDNA encoding brain amyloid of Alzheimer’s disease. Science. 1987;235:877–80.

    Article  CAS  PubMed  Google Scholar 

  13. Li Y, Zhou W, Tong Y, He G, Song W. Control of APP processing and Abeta generation level by BACE1 enzymatic activity and transcription. FASEB J. 2006;20:285–92.

    Article  PubMed  CAS  Google Scholar 

  14. Zhang S, Wang Z, Cai F, Zhang M, Wu Y, Zhang J, et al. BACE1 cleavage site selection critical for amyloidogenesis and Alzheimer’s Pathogenesis. J Neurosci. 2017;37:6915–25.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Deng Y, Wang Z, Wang R, Zhang X, Zhang S, Wu Y, et al. Amyloid-beta protein (Abeta) Glu11 is the major beta-secretase site of beta-site amyloid-beta precursor protein-cleaving enzyme 1(BACE1), and shifting the cleavage site to Abeta Asp1 contributes to Alzheimer pathogenesis. Eur J Neurosci. 2013;37:1962–9.

    Article  PubMed  Google Scholar 

  16. Song W, Nadeau P, Yuan M, Yang X, Shen J, Yankner BA. Proteolytic release and nuclear translocation of Notch-1 are induced by presenilin-1 and impaired by pathogenic presenilin-1 mutations. Proc Natl Acad Sci USA. 1999;96:6959–63.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Zhang Z, Nadeau P, Song W, Donoviel D, Yuan M, Bernstein A, et al. Presenilins are required for gamma-secretase cleavage of beta-APP and transmembrane cleavage of Notch-1. Nat Cell Biol. 2000;2:463–5.

    Article  CAS  PubMed  Google Scholar 

  18. Lue LF, Kuo YM, Roher AE, Brachova L, Shen Y, Sue L, et al. Soluble amyloid beta peptide concentration as a predictor of synaptic change in Alzheimer’s disease. Am J Pathol. 1999;155:853–62.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. McLean CA, Cherny RA, Fraser FW, Fuller SJ, Smith MJ, Beyreuther K, et al. Soluble pool of Abeta amyloid as a determinant of severity of neurodegeneration in Alzheimer’s disease. Ann Neurol. 1999;46:860–6.

    Article  CAS  PubMed  Google Scholar 

  20. Lambert MP, Barlow AK, Chromy BA, Edwards C, Freed R, Liosatos M, et al. Diffusible, nonfibrillar ligands derived from Abeta1-42 are potent central nervous system neurotoxins. Proc Natl Acad Sci USA. 1998;95:6448–53.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Lacor PN, Buniel MC, Furlow PW, Clemente AS, Velasco PT, Wood M, et al. Abeta oligomer-induced aberrations in synapse composition, shape, and density provide a molecular basis for loss of connectivity in Alzheimer’s disease. J Neurosci. 2007;27:796–807.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Koffie RM, Meyer-Luehmann M, Hashimoto T, Adams KW, Mielke ML, Garcia-Alloza M, et al. Oligomeric amyloid beta associates with postsynaptic densities and correlates with excitatory synapse loss near senile plaques. Proc Natl Acad Sci USA. 2009;106:4012–7.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Shankar GM, Li S, Mehta TH, Garcia-Munoz A, Shepardson NE, Smith I, et al. Amyloid-beta protein dimers isolated directly from Alzheimer’s brains impair synaptic plasticity and memory. Nat Med. 2008;14:837–42.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Walsh DM, Klyubin I, Fadeeva JV, Cullen WK, Anwyl R, Wolfe MS, et al. Naturally secreted oligomers of amyloid beta protein potently inhibit hippocampal long-term potentiation in vivo. Nature. 2002;416:535–9.

    Article  CAS  PubMed  Google Scholar 

  25. Klyubin I, Walsh DM, Lemere CA, Cullen WK, Shankar GM, Betts V, et al. Amyloid beta protein immunotherapy neutralizes Abeta oligomers that disrupt synaptic plasticity in vivo. Nat Med. 2005;11:556-61.

    Article  CAS  PubMed  Google Scholar 

  26. Lei M, Xu H, Li Z, Wang Z, O’Malley TT, Zhang D, et al. Soluble Abeta oligomers impair hippocampal LTP by disrupting glutamatergic/GABAergic balance. Neurobiol Dis. 2016;85:111–21.

    Article  CAS  PubMed  Google Scholar 

  27. Zhao D, Watson JB, Xie CW. Amyloid beta prevents activation of calcium/calmodulin-dependent protein kinase II and AMPA receptor phosphorylation during hippocampal long-term potentiation. J Neurophysiol. 2004;92:2853–8.

    Article  CAS  PubMed  Google Scholar 

  28. Li S, Jin M, Koeglsperger T, Shepardson NE, Shankar GM, Selkoe DJ. Soluble Abeta oligomers inhibit long-term potentiation through a mechanism involving excessive activation of extrasynaptic NR2B-containing NMDA receptors. J Neurosci. 2011;31:6627–38.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Takahashi RH, Capetillo-Zarate E, Lin MT, Milner TA, Gouras GK. Co-occurrence of Alzheimer’s disease ss-amyloid and tau pathologies at synapses. Neurobiol Aging. 2010;31:1145–52.

    Article  CAS  PubMed  Google Scholar 

  30. Zempel H, Thies E, Mandelkow E, Mandelkow EM. Abeta oligomers cause localized Ca(2+) elevation, missorting of endogenous Tau into dendrites, Tau phosphorylation, and destruction of microtubules and spines. J Neurosci. 2010;30:11938–50.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Roberson ED, Halabisky B, Yoo JW, Yao J, Chin J, Yan F, et al. Amyloid-beta/Fyn-induced synaptic, network, and cognitive impairments depend on tau levels in multiple mouse models of Alzheimer’s disease. J Neurosci. 2011;31:700–11.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Andorfer C, Acker CM, Kress Y, Hof PR, Duff K, Davies P. Cell-cycle reentry and cell death in transgenic mice expressing nonmutant human tau isoforms. J Neurosci. 2005;25:5446–54.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Tai HC, Wang BY, Serrano-Pozo A, Frosch MP, Spires-Jones TL, Hyman BT. Frequent and symmetric deposition of misfolded tau oligomers within presynaptic and postsynaptic terminals in Alzheimer’s disease. Acta Neuropathologica Commun. 2014;2:146.

    Google Scholar 

  34. Hoffmann NA, Dorostkar MM, Blumenstock S, Goedert M, Herms J. Impaired plasticity of cortical dendritic spines in P301S tau transgenic mice. Acta Neuropathologica Commun. 2013;1:82.

    Article  Google Scholar 

  35. Jaworski T, Lechat B, Demedts D, Gielis L, Devijver H, Borghgraef P, et al. Dendritic degeneration, neurovascular defects, and inflammation precede neuronal loss in a mouse model for tau-mediated neurodegeneration. Am J Pathol. 2011;179:2001–15.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Kopeikina KJ, Polydoro M, Tai HC, Yaeger E, Carlson GA, Pitstick R, et al. Synaptic alterations in the rTg4510 mouse model of tauopathy. J Comp Neurol. 2013;521:1334–53.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Rocher AB, Crimins JL, Amatrudo JM, Kinson MS, Todd-Brown MA, Lewis J, et al. Structural and functional changes in tau mutant mice neurons are not linked to the presence of NFTs. Exp Neurol. 2010;223:385–93.

    Article  CAS  PubMed  Google Scholar 

  38. Dejanovic B, Huntley MA, De Maziere A, Meilandt WJ, Wu T, Srinivasan K, et al. Changes in the synaptic proteome in tauopathy and rescue of Tau-Induced synapse loss by C1q antibodies. Neuron. 2018;100:1322–36 e1327.

    Article  CAS  PubMed  Google Scholar 

  39. Dickstein DL, Brautigam H, Stockton SD Jr, Schmeidler J, Hof PR. Changes in dendritic complexity and spine morphology in transgenic mice expressing human wild-type tau. Brain Struct Funct. 2010;214:161–79.

    Article  PubMed  PubMed Central  Google Scholar 

  40. Polydoro M, Acker CM, Duff K, Castillo PE, Davies P. Age-dependent impairment of cognitive and synaptic function in the htau mouse model of tau pathology. J Neurosci. 2009;29:10741–9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Pickett EK, Henstridge CM, Allison E, Pitstick R, Pooler A, Wegmann S, et al. Spread of tau down neural circuits precedes synapse and neuronal loss in the rTgTauEC mouse model of early Alzheimer’s disease. Synapse. 2017;71:e21965.

    Article  PubMed Central  CAS  Google Scholar 

  42. Zheng L, Duan J, Duan X, Zhou W, Chen C, Li Y, et al. Association of apolipoprotein E (ApoE) Polymorphism with Alzheimer’s Disease in Chinese Population. Curr Alzheimer Res. 2016;13:912–7.

    Article  CAS  PubMed  Google Scholar 

  43. Farrer LA, Cupples LA, Haines JL, Hyman B, Kukull WA, Mayeux R, et al. Effects of age, sex, and ethnicity on the association between apolipoprotein E genotype and Alzheimer disease. A meta-analysis. APOE and Alzheimer Disease Meta Analysis Consortium. JAMA. 1997;278:1349–56.

    Article  CAS  PubMed  Google Scholar 

  44. Reiman EM, Arboleda-Velasquez JF, Quiroz YT, Huentelman MJ, Beach TG, Caselli RJ, et al. Exceptionally low likelihood of Alzheimer’s dementia in APOE2 homozygotes from a 5,000-person neuropathological study. Nat Commun. 2020;11:667.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Strittmatter WJ, Saunders AM, Schmechel D, Pericak-Vance M, Enghild J, Salvesen GS, et al. Apolipoprotein E: high-avidity binding to beta-amyloid and increased frequency of type 4 allele in late-onset familial Alzheimer disease. Proc Natl Acad Sci USA. 1993;90:1977–81.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Hesse R, Hurtado ML, Jackson RJ, Eaton SL, Herrmann AG, Colom-Cadena M, et al. Comparative profiling of the synaptic proteome from Alzheimer’s disease patients with focus on the APOE genotype. Acta Neuropathologica Commun. 2019;7:214.

    Article  CAS  Google Scholar 

  47. Zhao J, Fu Y, Yamazaki Y, Ren Y, Davis MD, Liu CC, et al. APOE4 exacerbates synapse loss and neurodegeneration in Alzheimer’s disease patient iPSC-derived cerebral organoids. Nat Commun. 2020;11:5540.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. Chung WS, Verghese PB, Chakraborty C, Joung J, Hyman BT, Ulrich JD, et al. Novel allele-dependent role for APOE in controlling the rate of synapse pruning by astrocytes. Proc Natl Acad Sci USA. 2016;113:10186–91.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. Jain S, Yoon SY, Leung L, Knoferle J, Huang Y. Cellular source-specific effects of apolipoprotein (apo) E4 on dendrite arborization and dendritic spine development. PLoS ONE. 2013;8:e59478.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  50. Hudry E, Dashkoff J, Roe AD, Takeda S, Koffie RM, Hashimoto T, et al. Gene transfer of human Apoe isoforms results in differential modulation of amyloid deposition and neurotoxicity in mouse brain. Sci Transl Med. 2013;5:212ra161.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  51. Kang SS, Ahn EH, Liu X, Bryson M, Miller GW, Weinshenker D, et al. ApoE4 inhibition of VMAT2 in the locus coeruleus exacerbates Tau pathology in Alzheimer’s disease. Acta Neuropathol. 2021;142:139–58.

    Article  CAS  PubMed  Google Scholar 

  52. Wang C, Xiong M, Gratuze M, Bao X, Shi Y, Andhey PS, et al. Selective removal of astrocytic APOE4 strongly protects against tau-mediated neurodegeneration and decreases synaptic phagocytosis by microglia. Neuron. 2021;109:1657–74 e1657.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  53. Nimmerjahn A, Kirchhoff F, Helmchen F. Resting microglial cells are highly dynamic surveillants of brain parenchyma in vivo. Science. 2005;308:1314–8.

    Article  CAS  PubMed  Google Scholar 

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

    Article  PubMed  Google Scholar 

  55. Stevens B, Allen NJ, Vazquez LE, Howell GR, Christopherson KS, Nouri N, et al. The classical complement cascade mediates CNS synapse elimination. Cell. 2007;131:1164–78.

    Article  CAS  PubMed  Google Scholar 

  56. Stephan AH, Barres BA, Stevens B. The complement system: an unexpected role in synaptic pruning during development and disease. Annu Rev Neurosci. 2012;35:369–89.

    Article  CAS  PubMed  Google Scholar 

  57. Fonseca MI, Chu SH, Hernandez MX, Fang MJ, Modarresi L, Selvan P, et al. Cell-specific deletion of C1qa identifies microglia as the dominant source of C1q in mouse brain. J Neuroinflammation. 2017;14:48.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  58. Haga S, Ikeda K, Sato M, Ishii T. Synthetic Alzheimer amyloid beta/A4 peptides enhance production of complement C3 component by cultured microglial cells. Brain Res. 1993;601:88–94.

    Article  CAS  PubMed  Google Scholar 

  59. Schafer DP, Lehrman EK, Kautzman AG, Koyama R, Mardinly AR, Yamasaki R, et al. Microglia sculpt postnatal neural circuits in an activity and complement-dependent manner. Neuron. 2012;74:691–705.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  60. Stephan AH, Madison DV, Mateos JM, Fraser DA, Lovelett EA, Coutellier L, et al. A dramatic increase of C1q protein in the CNS during normal aging. J Neurosci. 2013;33:13460–74.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  61. Meyer-Luehmann M, Spires-Jones TL, Prada C, Garcia-Alloza M, de Calignon A, Rozkalne A, et al. Rapid appearance and local toxicity of amyloid-beta plaques in a mouse model of Alzheimer’s disease. Nature. 2008;451:720–4.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  62. Condello C, Yuan P, Schain A, Grutzendler J. Microglia constitute a barrier that prevents neurotoxic protofibrillar Abeta42 hotspots around plaques. Nat Commun. 2015;6:6176.

    Article  CAS  PubMed  Google Scholar 

  63. Hong S, Beja-Glasser VF, Nfonoyim BM, Frouin A, Li S, Ramakrishnan S, et al. Complement and microglia mediate early synapse loss in Alzheimer mouse models. Science. 2016;352:712–6.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  64. Reichwald J, Danner S, Wiederhold KH, Staufenbiel M. Expression of complement system components during aging and amyloid deposition in APP transgenic mice. J Neuroinflammation. 2009;6:35.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  65. Fonseca MI, Zhou J, Botto M, Tenner AJ. Absence of C1q leads to less neuropathology in transgenic mouse models of Alzheimer’s disease. J Neurosci. 2004;24:6457–65.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  66. Zhang Y, Dong Z, Song W. NLRP3 inflammasome as a novel therapeutic target for Alzheimer’s disease. Signal Transduct Target Ther. 2020;5:37.

    Article  PubMed  PubMed Central  Google Scholar 

  67. Heneka MT, Kummer MP, Stutz A, Delekate A, Schwartz S, Vieira-Saecker A, et al. NLRP3 is activated in Alzheimer’s disease and contributes to pathology in APP/PS1 mice. Nature. 2013;493:674–8.

    Article  CAS  PubMed  Google Scholar 

  68. Ising C, Venegas C, Zhang S, Scheiblich H, Schmidt SV, Vieira-Saecker A, et al. NLRP3 inflammasome activation drives tau pathology. Nature. 2019;575:669–73.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  69. Lonnemann N, Hosseini S, Marchetti C, Skouras DB, Stefanoni D, D’Alessandro A, et al. The NLRP3 inflammasome inhibitor OLT1177 rescues cognitive impairment in a mouse model of Alzheimer’s disease. Proc Natl Acad Sci USA. 2020;117:32145–54.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  70. Hennequin LF, Allen J, Breed J, Curwen J, Fennell M, Green TP, et al. N-(5-chloro-1,3-benzodioxol-4-yl)-7-[2-(4-methylpiperazin-1-yl)ethoxy] -5-(tetrahydro-2H-pyran-4-yloxy)quinazolin-4-amine, a novel, highly selective, orally available, dual-specific c-Src/Abl kinase inhibitor. J Med Chem. 2006;49:6465–88.

  71. Larson M, Sherman MA, Amar F, Nuvolone M, Schneider JA, Bennett DA, et al. The complex PrPc-Fyn couples human oligomeric Aβ with pathological tau changes in Alzheimer’s disease. J Neurosci. 2012;32:16857–71.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  72. Um JW, Nygaard HB, Heiss JK, Kostylev MA, Stagi M, Vortmeyer A, et al. Alzheimer amyloid-Î 2 oligomer bound to postsynaptic prion protein activates Fyn to impair neurons. Nat Neurosci. 2012;15:1227–35.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  73. Kaufman AC, Salazar SV, Haas LT, Yang J, Kostylev MA, Jeng AT, et al. Fyn inhibition rescues established memory and synapse loss in Alzheimer mice. Ann Neurol. 2015;77:953–71.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  74. Smith LM, Zhu R, Strittmatter SM. Disease-modifying benefit of Fyn blockade persists after washout in mouse Alzheimer’s model. Neuropharmacology. 2018;130:54–61.

    Article  CAS  PubMed  Google Scholar 

  75. Toyonaga T, Smith LM, Finnema SJ, Gallezot JD, Naganawa M, Bini J, et al. In vivo synaptic density imaging with 11C-UCB-J detects treatment effects of saracatinib in a mouse model of Alzheimer disease. J Nucl Med. 2019;60:1780–6.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  76. Nygaard HB, Wagner AF, Bowen GS, Good SP, MacAvoy MG, Strittmatter KA, et al. A phase Ib multiple ascending dose study of the safety, tolerability, and central nervous system availability of AZD0530 (saracatinib) in Alzheimer’s disease. Alzheimers Res Ther. 2015;7:35–35.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  77. Van Dyck CH, Nygaard HB, Chen K, Donohue MC, Raman R, Rissman RA, et al. Effect of AZD0530 on cerebral metabolic decline in Alzheimer Disease: a randomized clinical trial. JAMA Neurol. 2019;76:1219–29.

    Article  PubMed  PubMed Central  Google Scholar 

  78. Trias E, Ibarburu S, Barreto-Núñez R, Varela V, Moura IC, Dubreuil P, et al. Evidence for mast cells contributing to neuromuscular pathology in an inherited model of ALS. JCI Insight. 2017;2:e95934.

    Article  PubMed Central  Google Scholar 

  79. Li T, Martin E, Abada Y-S, Boucher C, Cès A, Youssef I, et al. Effects of chronic masitinib treatment in APPPS1dE9 transgenic mice modeling Alzheimer’s Disease. J Alzheimer’s Dis. 2020;76:1339–45.

    Article  CAS  Google Scholar 

  80. Piette F, Belmin J, Vincent H, Schmidt N, Pariel S, Verny M, et al. Masitinib as an adjunct therapy for mild-to-moderate Alzheimer’s disease: A randomised, placebo-controlled phase 2 trail. Alzheimer’s Res Ther. 2011;3:1–11.

    Google Scholar 

  81. Iannuzzi F, Sirabella R, Canu N, Maier TJ, Annunziato L, Matrone C. Fyn tyrosine kinase elicits amyloid precursor protein Tyr682 phosphorylation in neurons from Alzheimer’s Disease Patients. Cells. 2020;9:1807.

    Article  CAS  PubMed Central  Google Scholar 

  82. Sellers KJ, Elliott C, Jackson J, Ghosh A, Ribe E, Rojo AI, et al. Amyloid β synaptotoxicity is Wnt-PCP dependent and blocked by fasudil. Alzheimers Dement. 2018;14:306–17.

    Article  PubMed  PubMed Central  Google Scholar 

  83. Yu J, Yan Y, Gu Q, Kumar G, Yu H, Zhao Y, et al. Fasudil in combination with Bone Marrow Stromal Cells (BMSCs) Attenuates Alzheimer’s Disease-Related changes through the regulation of the peripheral immune system. Front Aging Neurosci. 2018;10:216–16.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  84. Kumar M, Bansal N. Fasudil hydrochloride ameliorates memory deficits in rat model of streptozotocin-induced Alzheimer’s disease: Involvement of PI3-kinase, eNOS and NFκB. Behavioural Brain Res. 2018;351:4–16.

    Article  CAS  Google Scholar 

  85. Xin YL, Yu JZ, Yang XW, Liu CY, Li YH, Feng L, et al. FSD-C10: a more promising novel ROCK inhibitor than Fasudil for treatment of CNS autoimmunity. Biosci Rep. 2015;35:247.

    Article  CAS  Google Scholar 

  86. Gu QF, Yu JZ, Wu H, Li YH, Liu CY, Feng L, et al. Therapeutic effect of Rho kinase inhibitor FSD-C10 in a mouse model of alzheimer’s disease. Exp Therapeutic Med. 2018;16:3929–38.

    Google Scholar 

  87. Bormann J. Memantine is a potent blocker of N-methyl-D-aspartate (NMDA) receptor channels. Eur J Pharm. 1989;166:591–2.

    Article  CAS  Google Scholar 

  88. Fleischhacker WW, Buchgeher A, Schubert H. Memantine in the treatment of senile dementia of the Alzheimer type. Prog Neuropsychopharmacol Biol Psychiatry. 1986;10:87–93.

    Article  CAS  PubMed  Google Scholar 

  89. Winblad B, Poritis N. Memantine in severe dementia: results of the 9M-Best Study (Benefit and efficacy in severely demented patients during treatment with memantine). Int J Geriatr Psychiatry. 1999;14:135–46.

    Article  CAS  PubMed  Google Scholar 

  90. Reisberg B, Doody R, Stöffler A, Schmitt F, Ferris S, Möbius HJ. Memantine in moderate-to-severe Alzheimer’s disease. N. Engl J Med. 2003;348:1333–41.

    Article  CAS  PubMed  Google Scholar 

  91. Devi L, Ohno M. Cognitive benefits of memantine in Alzheimer’s 5XFAD model mice decline during advanced disease stages. Pharmacol Biochem Behav. 2016;144:60–66.

    Article  CAS  PubMed  Google Scholar 

  92. Folch J, Busquets O, Ettcheto M, Sánchez-López E, Castro-Torres RD, Verdaguer E, et al. Memantine for the treatment of Dementia: a review on its current and future applications. J Alzheimer’s Dis. 2018;62:1223–40.

    Article  CAS  Google Scholar 

  93. Knight R, Khondoker M, Magill N, Stewart R, Landau S. A systematic review and meta-analysis of the effectiveness of acetylcholinesterase inhibitors and memantine in treating the cognitive symptoms of dementia. Dement Geriatr Cogn Disord. 2018;45:131–51.

    Article  CAS  PubMed  Google Scholar 

  94. Tampi RR, van Dyck CH. Memantine: Efficacy and safety in mild-to-severe Alzheimer’s disease. Neuropsychiatr Dis Treat. 2007;3:245–58.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  95. Talantova M, Sanz-Blasco S, Zhang X, Xia P, Akhtar MW, Okamoto S, et al. Aβ induces astrocytic glutamate release, extrasynaptic NMDA receptor activation, and synaptic loss. Proc Natl Acad Sci USA. 2013;110:E2518–2527.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  96. Trudler D, Sanz-Blasco S, Eisele YS, Ghatak S, Bodhinathan K, Akhtar MW, et al. alpha-Synuclein oligomers induce glutamate release from astrocytes and excessive extrasynaptic NMDAR activity in neurons, thus contributing to synapse loss. J Neurosci. 2021;41:2264–73.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  97. Ghatak S, Dolatabadi N, Gao R, Wu Y, Scott H, Trudler D, et al. NitroSynapsin ameliorates hypersynchronous neural network activity in Alzheimer hiPSC models. Mol Psychiatry. 2021;26:5751–65.

    Article  CAS  PubMed  Google Scholar 

  98. Pereira AC, Lambert HK, Grossman YS, Dumitriu D, Waldman R, Jannetty SK, et al. Glutamatergic regulation prevents hippocampal-dependent age-related cognitive decline through dendritic spine clustering. Proc Natl Acad Sci USA. 2014;111:18733–8.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  99. Mokhtari Z, Baluchnejadmojarad T, Nikbakht F, Mansouri M, Roghani M. Riluzole ameliorates learning and memory deficits in Abeta25-35-induced rat model of Alzheimer’s disease and is independent of cholinoceptor activation. Biomed Pharmacother. 2017;87:135–44.

    Article  CAS  PubMed  Google Scholar 

  100. Matthews DC, Mao X, Dowd K, Tsakanikas D, Jiang CS, Meuser C, et al. Riluzole, a glutamate modulator, slows cerebral glucose metabolism decline in patients with Alzheimer’s disease. Brain. 2021;144:3742-55.

    Article  PubMed  Google Scholar 

  101. Burns A, Rossor M, Hecker J, Gauthier S, Petit H, Möller HJ, et al. The effects of donepezil in Alzheimer’s disease - Results from a multinational trial. Dement Geriatr Cogn Disord. 1999;10:237–44.

    Article  CAS  PubMed  Google Scholar 

  102. Johannsen P, Salmon E, Hampel H, Xu Y, Richardson S, Qvitzau S, et al. Assessing therapeutic efficacy in a progressive disease: a study of donepezil in Alzheimer’s disease. CNS Drugs. 2006;20:311–25.

    Article  CAS  PubMed  Google Scholar 

  103. Seltzer B, Zolnouni P, Nunez M, Goldman R, Kumar D, Ieni J, et al. Efficacy of donepezil in early-stage Alzheimer disease: a randomized placebo-controlled trial. Arch Neurol. 2004;61:1852–6.

    Article  PubMed  Google Scholar 

  104. Wilcock GK, Lilienfeld S, Gaens E. Efficacy and safety of galantamine in patients with mild to moderate Alzheimer’s disease: multicentre randomised controlled trial. Br Med J. 2000;321:1445–9.

    Article  CAS  Google Scholar 

  105. Tariot PN, Solomon PR, Morris JC, Kershaw P, Lilienfeld S, Ding C. A 5-month, randomized, placebo-controlled trial of galantamine in AD. Neurology. 2000;54:2269–76.

    Article  CAS  PubMed  Google Scholar 

  106. Rösler M, Anand R, Cicin-Sain A, Gauthier S, Agid Y, Dal-Bianco P, et al. Efficacy and safety of rivastigmine in patients with Alzheimer’s disease: International randomised controlled trial. Br Med J. 1999;318:633–40.

    Article  Google Scholar 

  107. Small GW, Kaufer D, Mendiondo MS, Quarg P, Spiegel R. Cognitive performance in Alzheimer’s disease patients receiving rivastigmine for up to 5 years. Int J Clin Pract. 2005;59:473–7.

    Article  CAS  PubMed  Google Scholar 

  108. Lopez OL, Becker JT, Wahed AS, Saxton J, Sweet RA, Wolk DA, et al. Long-term effects of the concomitant use of memantine with cholinesterase inhibition in Alzheimer disease. J Neurol Neurosurg Psychiatry. 2009;80:600–7.

    Article  CAS  PubMed  Google Scholar 

  109. Riepe MW, Adler G, Ibach B, Weinkauf B, Tracik F, Gunay I. Domain-specific improvement of cognition on memantine in patients with Alzheimer’s disease treated with rivastigmine. Dement Geriatr Cogn Disord. 2007;23:301–6.

    Article  CAS  PubMed  Google Scholar 

  110. Rountree SD, Chan W, Pavlik VN, Darby EJ, Siddiqui S, Doody RS. Persistent treatment with cholinesterase inhibitors and/or memantine slows clinical progression of Alzheimer disease. Alzheimer’s Res Ther. 2009;1:7.

    Article  CAS  Google Scholar 

  111. Tricco AC, Soobiah C, Berliner S, Ho JM, Ng CH, Ashoor HM, et al. Efficacy and safety of cognitive enhancers for patients with mild cognitive impairment: a systematic review and meta-analysis. CMAJ. 2013;185:1393–401.

    Article  PubMed  PubMed Central  Google Scholar 

  112. Davis B, Sadik K. Circadian cholinergic rhythms: Implications for cholinesterase inhibitor therapy. Dement Geriatr Cogn Disord. 2006;21:120–9.

    Article  PubMed  Google Scholar 

  113. Finkel SI, Mintzer JE, Dysken M, Krishnan KRR, Burt T, McRae T. A randomized, placebo-controlled study of the efficacy and safety of sertraline in the treatment of the behavioral manifestations of Alzheimer’s disease in outpatients treated with donepezil. Int J Geriat Psychiat. 2004;19:9–18.

    Article  Google Scholar 

  114. Mowla A, Mosavinasab M, Pani A. Does fluoxetine have any effect on the cognition of patients with mild cognitive impairment? J Clin Psychopharm. 2007;27:67–70.

    Article  CAS  Google Scholar 

  115. Bartels C, Wagner M, Wolfsgruber S, Ehrenreich H, Schneider A. Impact of SSRI therapy on risk of conversion from mild cognitive impairment to Alzheimer’s dementia in individuals with previous depression. Am J Psychiatry. 2018;175:232–41.

    Article  PubMed  Google Scholar 

  116. Banerjee S, Hellier J, Dewey M, Romeo R, Ballard C, Baldwin R, et al. Sertraline or mirtazapine for depression in dementia (HTA-SADD): a randomised, multicentre, double-blind, placebo-controlled trial. Lancet. 2011;378:403–11.

    Article  CAS  PubMed  Google Scholar 

  117. Munro CA. Cognitive response to pharmacological treatment for depression in Alzheimer Disease: secondary outcomes from the Depression in Alzheimer’s Disease Study (DIADS). Am J Geriatr Psychiatry. 2004;12:491–8.

    Article  PubMed  Google Scholar 

  118. Olafsson K, Jørgensen S, Jensen HV, Bille A, Arup P, Andersen J. Fluvoxamine in the treatment of demented elderly patients: a double‐blind, placebo‐controlled study. Acta Psychiatr Scandinavica. 1992;85:453–6.

    Article  CAS  Google Scholar 

  119. Porsteinsson AP, Drye LT, Pollock BG, Devanand DP, Frangakis C, Ismail Z, et al. Effect of citalopram on agitation in Alzheimer disease: The CitAD randomized clinical trial. JAMA - J Am Med Assoc. 2014;311:682–91.

    Article  CAS  Google Scholar 

  120. Ivković M, Damjanović A, Jasović-Gasić M, Paunović VR. The effects of fluoxetine on cognitive functions in animal model of Alzheimer’s disease. Psychiatr Danubina. 2004;16:15–20.

    Google Scholar 

  121. Jin L, Gao LF, Sun DS, Wu H, Wang Q, Ke D, et al. Long-term ameliorative effects of the antidepressant fluoxetine exposure on cognitive deficits in 3 × TgAD Mice. Mol Neurobiol. 2017;54:4160–71.

    Article  CAS  PubMed  Google Scholar 

  122. Zhou CN, Chao FL, Zhang Y, Jiang L, Zhang L, Fan JH, et al. Fluoxetine delays the cognitive function decline and synaptic changes in a transgenic mouse model of early Alzheimer’s disease. J Comp Neurol. 2019;527:1378–87.

    Article  CAS  PubMed  Google Scholar 

  123. Hajszan T, MacLusky NJ, Leranth C. Short-term treatment with the antidepressant fluoxetine triggers pyramidal dendritic spine synapse formation in rat hippocampus. Eur J Neurosci. 2005;21:1299–303.

    Article  PubMed  Google Scholar 

  124. Zheng J, Xu DF, Li K, Wang HT, Shen PC, Lin M, et al. Neonatal exposure to fluoxetine and fluvoxamine alteres spine density in mouse hippocampal CA1 pyramidal neurons. Int J Clin Exp Pathol. 2011;4:162–8.

    CAS  PubMed  PubMed Central  Google Scholar 

  125. Rozzini L, Vicini Chilovi B, Conti M, Bertoletti E, Zanetti M, Trabucchi M, et al. Efficacy of SSRIs on cognition of Alzheimer’s disease patients treated with cholinesterase inhibitors. Int Psychogeriatr. 2010;22:114–9.

    Article  PubMed  Google Scholar 

  126. Orgeta V, Tabet N, Nilforooshan R, Howard R. Efficacy of antidepressants for depression in Alzheimer’s Disease: systematic review and meta-analysis. J Alzheimer’s Dis. 2017;58:725–33.

    Article  CAS  Google Scholar 

  127. Petracca GM, Chemerinski E, Starkstein SE. A double-blind, placebo-controlled study of fluoxetine in depressed patients with Alzheimer’s disease. Int Psychogeriatr. 2001;13:233–40.

    Article  CAS  PubMed  Google Scholar 

  128. Jin K, Peel AL, Mao XO, Xie L, Cottrell BA, Henshall DC, et al. Increased hippocampal neurogenesis in Alzheimer’s disease. Proc Natl Acad Sci USA. 2004;101:343–7.

    Article  CAS  PubMed  Google Scholar 

  129. Li B, Yamamori H, Tatebayashi Y, Shafit-Zagardo B, Tanimukai H, Chen S, et al. Failure of neuronal maturation in Alzheimer disease dentate gyrus. J Neuropathol Exp Neurol. 2008;67:78–84.

    Article  CAS  PubMed  Google Scholar 

  130. Mukaetova-Ladinska EB, Garcia-Siera F, Hurt J, Gertz HJ, Xuereb JH, Hills R, et al. Staging of cytoskeletal and β-amyloid changes in human isocortex reveals biphasic synaptic protein response during progression of Alzheimer’s disease. Am J Pathol. 2000;157:623–36.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  131. Ferrer I, Marín C, Rey MJ, Ribalta T, Goutan E, Blanco R, et al. BDNF and full-length and Truncated TrkB expression in Alzheimer disease. Implications in Therapeutic strategies. J Neuropathol Exp Neurol. 1999;58:729–39.

    Article  CAS  PubMed  Google Scholar 

  132. Cocco C, D’Amato F, Noli B, Ledda A, Brancia C, Bongioanni P, et al. Distribution of VGF peptides in the human cortex and their selective changes in Parkinson’s and Alzheimer’s diseases. J Anat. 2010;217:683–93.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  133. Bruno MA, Leon WC, Fragoso G, Mushynski WE, Almazan G, Cuello AC. Amyloid beta-induced nerve growth factor dysmetabolism in Alzheimer disease. J Neuropathol Exp Neurol. 2009;68:857–69.

    Article  CAS  PubMed  Google Scholar 

  134. Fahnestock M, Michalski B, Xu B, Coughlin MD. The precursor pro-nerve growth factor is the predominant form of nerve growth factor in brain and is increased in Alzheimer’s disease. Mol Cell Neurosci. 2001;18:210–20.

    Article  CAS  PubMed  Google Scholar 

  135. Tuszynski MH, Thal L, Pay M, Salmon DP, Sang U, H., Bakay R, et al. A phase 1 clinical trial of nerve growth factor gene therapy for Alzheimer disease. Nat Med. 2005;11:551–5.

    Article  CAS  PubMed  Google Scholar 

  136. Rafii MS, Baumann TL, Bakay RAE, Ostrove JM, Siffert J, Fleisher AS, et al. A phase1 study of stereotactic gene delivery of AAV2-NGF for Alzheimer’s disease. Alzheimer’s Dement: J Alzheimer’s Assoc. 2014;10:571–81.

    Article  Google Scholar 

  137. Rafii MS, Tuszynski MH, Thomas RG, Barba D, Brewer JB, Rissman RA, et al. Adeno-associated viral vector (serotype 2)-nerve growth factor for patients with Alzheimer disease a randomized clinical trial. JAMA Neurol. 2018;75:834–41.

    Article  PubMed  PubMed Central  Google Scholar 

  138. Baazaoui N, Iqbal K. Prevention of Amyloid-β and Tau pathologies, associated neurodegeneration, and cognitive deficit by early treatment with a neurotrophic compound. J Alzheimer’s Dis. 2017;58:215–30.

    Article  CAS  Google Scholar 

  139. Baazaoui N, Iqbal K. Prevention of dendritic and synaptic deficits and cognitive impairment with a neurotrophic compound. Alzheimer’s Res Ther. 2017;9:45–45.

    Article  CAS  Google Scholar 

  140. Kazim SF, Blanchard J, Dai CL, Tung YC, LaFerla FM, Iqbal IG, et al. Disease modifying effect of chronic oral treatment with a neurotrophic peptidergic compound in a triple transgenic mouse model of Alzheimer’s disease. Neurobiol Dis. 2014;71:110–30.

    Article  CAS  PubMed  Google Scholar 

  141. Li B, Wanka L, Blanchard J, Liu F, Chohan MO, Iqbal K, et al. Neurotrophic peptides incorporating adamantane improve learning and memory, promote neurogenesis and synaptic plasticity in mice. FEBS Lett. 2010;584:3359–65.

    Article  CAS  PubMed  Google Scholar 

  142. Eyjolfsdottir H, Eriksdotter M, Linderoth B, Lind G, Juliusson B, Kusk P, et al. Targeted delivery of nerve growth factor to the cholinergic basal forebrain of Alzheimer’s disease patients: Application of a second-generation encapsulated cell biodelivery device. Alzheimers Res Ther. 2016;8:30–30.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  143. Eriksdotter M, Navarro-Oviedo M, Mitra S, Wahlberg L, Linderoth B, Tjernberg LO, et al. Cerebrospinal fluid from Alzheimer patients affects cell-mediated nerve growth factor production and cell survival in vitro. Exp Cell Res. 2018;371:175–84.

    Article  CAS  PubMed  Google Scholar 

  144. James ML, Belichenko NP, Shuhendler AJ, Hoehne A, Andrews LE, Condon C, et al. [(18)F]GE-180 PET detects reduced microglia activation after LM11A-31 Therapy in a Mouse Model of Alzheimer’s Disease. Theranostics. 2017;7:1422–36.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Funding

This work was supported by the Key Laboratory of Alzheimer’s Disease Of Zhejiang Province, the grants from the Canadian Institutes of Health Research (CIHR) Project Grant PJT-166127 to WS, the National Natural Science Foundation of China (No. 81801091) to LP, and the Natural Science Foundation of Shandong Province of China (No. ZR2019BH076) to LP. WS was the holder of the Tier 1 Canada Research Chair in Alzheime’s Disease, and IBL is supported by the UBC Four Year Fellowship and the Jock & Irene Graham Brain Research Endowment.

Author information

Authors and Affiliations

Authors

Contributions

All authors were involved in literature search and writing of the article. WS supervised the project.

Corresponding author

Correspondence to Weihong Song.

Ethics declarations

Competing interests

The authors declare no competing interests.

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

Peng, L., Bestard-Lorigados, I. & Song, W. The synapse as a treatment avenue for Alzheimer’s Disease. Mol Psychiatry 27, 2940–2949 (2022). https://doi.org/10.1038/s41380-022-01565-z

Download citation

  • Received:

  • Revised:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41380-022-01565-z

  • Springer Nature Limited

This article is cited by

Navigation