Skip to main content
Log in

Scaffolding Proteins of the Post-synaptic Density Contribute to Synaptic Plasticity by Regulating Receptor Localization and Distribution: Relevance for Neuropsychiatric Diseases

  • Overview
  • Published:
Neurochemical Research Aims and scope Submit manuscript

Abstract

Synaptic plasticity represents the long lasting activity-related strengthening or weakening of synaptic transmission, whose well-characterized types are the long term potentiation and depression. Despite this classical definition, however, the molecular mechanisms by which synaptic plasticity may occur appear to be extremely complex and various. The post-synaptic density (PSD) of glutamatergic synapses is a major site for synaptic plasticity processes and alterations of PSD members have been recently implicated in neuropsychiatric diseases where an impairment of synaptic plasticity has also been reported. Among PSD members, scaffolding proteins have been demonstrated to bridge surface receptors with their intracellular effectors and to regulate receptors distribution and localization both at surface membranes and within the PSD. This review will focus on the molecular physiology and pathophysiology of synaptic plasticity processes, which are tuned by scaffolding PSD proteins and their close related partners, through the modulation of receptor localization and distribution at post-synaptic sites. We suggest that, by regulating both the compartmentalization of receptors along surface membrane and their degradation as well as by modulating receptor trafficking into the PSD, postsynaptic scaffolding proteins may contribute to form distinct signaling micro-domains, whose efficacy in transmitting synaptic signals depends on the dynamic stability of the scaffold, which in turn is provided by relative amounts and post-translational modifications of scaffolding members. The putative relevance for neuropsychiatric diseases and possible pathophysiological mechanisms are discussed in the last part of this work.

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.

Institutional subscriptions

Fig. 1
Fig. 2
Fig. 3

Similar content being viewed by others

References

  1. Bliss TV, Gardner-Medwin AR (1973) Long-lasting potentiation of synaptic transmission in the dentate area of the unanaestetized rabbit following stimulation of the perforant path. J Physiol 232(2):357–374

    PubMed  CAS  Google Scholar 

  2. Malenka RC, Bear MF (2004) LTP and LTD: an embarrassment of riches. Neuron 44(1):5–21. doi:10.1016/j.neuron.2004.09.012

    Article  PubMed  CAS  Google Scholar 

  3. Boeckers TM (2006) The postsynaptic density. Cell Tissue Res 326(2):409–422. doi:10.1007/s00441-006-0274-5

    Article  PubMed  CAS  Google Scholar 

  4. Holtmaat A, Svoboda K (2009) Experience-dependent structural synaptic plasticity in the mammalian brain. Nat Rev Neurosci 10(9):647–658. doi:10.1038/nrn2699

    Article  PubMed  CAS  Google Scholar 

  5. Meador-Woodruff JH, Clinton SM, Beneyto M, McCullumsmith RE (2003) Molecular abnormalities of the glutamate synapse in the thalamus in schizophrenia. Ann N Y Acad Sci 1003:75–93

    Article  PubMed  CAS  Google Scholar 

  6. Proctor DT, Coulson EJ, Dodd PR (2010) Reduction in post-synaptic scaffolding PSD-95 and SAP-102 protein levels in the Alzheimer inferior temporal cortex is correlated with disease pathology. J Alzheimer’s Dis JAD 21(3):795–811. doi:10.3233/JAD-2010-100090

    CAS  Google Scholar 

  7. Goto Y, Yang CR, Otani S (2010) Functional and dysfunctional synaptic plasticity in prefrontal cortex: roles in psychiatric disorders. Biol Psychiatry 67(3):199–207. doi:10.1016/j.biopsych.2009.08.026

    Article  PubMed  Google Scholar 

  8. Sheng M, Hoogenraad CC (2007) The postsynaptic architecture of excitatory synapses: a more quantitative view. Annu Rev Biochem 76:823–847. doi:10.1146/annurev.biochem.76.060805.160029

    Article  PubMed  CAS  Google Scholar 

  9. Santucci DM, Raghavachari S (2008) The effects of NR2 subunit-dependent NMDA receptor kinetics on synaptic transmission and CaMKII activation. PLoS Comput Biol 4(10):e1000208. doi:10.1371/journal.pcbi.1000208

    Article  PubMed  CAS  Google Scholar 

  10. Raghavachari S, Lisman JE (2004) Properties of quantal transmission at CA1 synapses. J Neurophysiol 92(4):2456–2467. doi:10.1152/jn.00258.2004

    Article  PubMed  CAS  Google Scholar 

  11. Opazo P, Sainlos M, Choquet D (2012) Regulation of AMPA receptor surface diffusion by PSD-95 slots. Curr Opin Neurobiol 22(3):453–460. doi:10.1016/j.conb.2011.10.010

    Google Scholar 

  12. Chen X, Winters C, Azzam R, Li X, Galbraith JA, Leapman RD, Reese TS (2008) Organization of the core structure of the postsynaptic density. Proc Natl Acad Sci USA 105(11):4453–4458. doi:10.1073/pnas.0800897105

    Article  PubMed  CAS  Google Scholar 

  13. Ehlers MD, Heine M, Groc L, Lee MC, Choquet D (2007) Diffusional trapping of GluR1 AMPA receptors by input-specific synaptic activity. Neuron 54(3):447–460. doi:10.1016/j.neuron.2007.04.010

    Article  PubMed  CAS  Google Scholar 

  14. Borgdorff AJ, Choquet D (2002) Regulation of AMPA receptor lateral movements. Nature 417(6889):649–653. doi:10.1038/nature00780

    Article  PubMed  CAS  Google Scholar 

  15. Heine M, Groc L, Frischknecht R, Beique JC, Lounis B, Rumbaugh G, Huganir RL, Cognet L, Choquet D (2008) Surface mobility of postsynaptic AMPARs tunes synaptic transmission. Science 320(5873):201–205. doi:10.1126/science.1152089

    Article  PubMed  CAS  Google Scholar 

  16. Sharma K, Fong DK, Craig AM (2006) Postsynaptic protein mobility in dendritic spines: long-term regulation by synaptic NMDA receptor activation. Mol Cell Neurosci 31(4):702–712. doi:10.1016/j.mcn.2006.01.010

    Article  PubMed  CAS  Google Scholar 

  17. Tardin C, Cognet L, Bats C, Lounis B, Choquet D (2003) Direct imaging of lateral movements of AMPA receptors inside synapses. EMBO J 22(18):4656–4665. doi:10.1093/emboj/cdg463

    Article  PubMed  CAS  Google Scholar 

  18. Groc L, Heine M, Cousins SL, Stephenson FA, Lounis B, Cognet L, Choquet D (2006) NMDA receptor surface mobility depends on NR2A-2B subunits. Proc Natl Acad Sci USA 103(49):18769–18774. doi:10.1073/pnas.0605238103

    Article  PubMed  CAS  Google Scholar 

  19. Tomita S, Adesnik H, Sekiguchi M, Zhang W, Wada K, Howe JR, Nicoll RA, Bredt DS (2005) Stargazin modulates AMPA receptor gating and trafficking by distinct domains. Nature 435(7045):1052–1058. doi:10.1038/nature03624

    Article  PubMed  CAS  Google Scholar 

  20. Bats C, Groc L, Choquet D (2007) The interaction between Stargazin and PSD-95 regulates AMPA receptor surface trafficking. Neuron 53(5):719–734. doi:10.1016/j.neuron.2007.01.030

    Article  PubMed  CAS  Google Scholar 

  21. Rumbaugh G, Sia GM, Garner CC, Huganir RL (2003) Synapse-associated protein-97 isoform-specific regulation of surface AMPA receptors and synaptic function in cultured neurons. J Neurosci Off J Soc Neurosci 23(11):4567–4576

    CAS  Google Scholar 

  22. Nakagawa T, Futai K, Lashuel HA, Lo I, Okamoto K, Walz T, Hayashi Y, Sheng M (2004) Quaternary structure, protein dynamics, and synaptic function of SAP97 controlled by L27 domain interactions. Neuron 44(3):453–467. doi:10.1016/j.neuron.2004.10.012

    Article  PubMed  CAS  Google Scholar 

  23. Waites CL, Specht CG, Hartel K, Leal-Ortiz S, Genoux D, Li D, Drisdel RC, Jeyifous O, Cheyne JE, Green WN, Montgomery JM, Garner CC (2009) Synaptic SAP97 isoforms regulate AMPA receptor dynamics and access to presynaptic glutamate. J Neurosci Off J Soc Neurosci 29(14):4332–4345. doi:10.1523/JNEUROSCI.4431-08.2009

    Article  CAS  Google Scholar 

  24. Malinow R, Malenka RC (2002) AMPA receptor trafficking and synaptic plasticity. Annu Rev Neurosci 25:103–126. doi:10.1146/annurev.neuro.25.112701.142758

    Article  PubMed  CAS  Google Scholar 

  25. Carroll RC, Beattie EC, von Zastrow M, Malenka RC (2001) Role of AMPA receptor endocytosis in synaptic plasticity. Nat Rev Neurosci 2(5):315–324. doi:10.1038/35072500

    Article  PubMed  CAS  Google Scholar 

  26. Lu J, Helton TD, Blanpied TA, Racz B, Newpher TM, Weinberg RJ, Ehlers MD (2007) Postsynaptic positioning of endocytic zones and AMPA receptor cycling by physical coupling of dynamin-3 to Homer. Neuron 55(6):874–889. doi:10.1016/j.neuron.2007.06.041

    Article  PubMed  CAS  Google Scholar 

  27. de Bartolomeis A, Iasevoli F (2003) The Homer family and the signal transduction system at glutamatergic postsynaptic density: potential role in behavior and pharmacotherapy. Psychopharmacol Bull 37(3):51–83

    PubMed  Google Scholar 

  28. Iasevoli F, Ambesi-Impiombato A, Fiore G, Panariello F, Muscettola G, de Bartolomeis A (2011) Pattern of acute induction of Homer1a gene is preserved after chronic treatment with first- and second-generation antipsychotics: effect of short-term drug discontinuation and comparison with Homer1a-interacting genes. J Psychopharmacol 25(7):875–887. doi:10.1177/0269881109358199

    Article  PubMed  CAS  Google Scholar 

  29. Iasevoli F, Tomasetti C, Marmo F, Bravi D, Arnt J, de Bartolomeis A (2010) Divergent acute and chronic modulation of glutamatergic postsynaptic density genes expression by the antipsychotics haloperidol and sertindole. Psychopharmacology 212(3):329–344

    Article  PubMed  CAS  Google Scholar 

  30. Tomasetti C, Dell’Aversano C, Iasevoli F, Marmo F, de Bartolomeis A (2011) The acute and chronic effects of combined antipsychotic-mood stabilizing treatment on the expression of cortical and striatal postsynaptic density genes. Prog Neuropsychopharmacol Biol Psychiatry 35(1):184–197. doi:10.1016/j.pnpbp.2010.10.025

    Article  PubMed  CAS  Google Scholar 

  31. Bhattacharyya S, Biou V, Xu W, Schluter O, Malenka RC (2009) A critical role for PSD-95/AKAP interactions in endocytosis of synaptic AMPA receptors. Nat Neurosci 12(2):172–181. doi:10.1038/nn.2249

    Article  PubMed  CAS  Google Scholar 

  32. Chowdhury S, Shepherd JD, Okuno H, Lyford G, Petralia RS, Plath N, Kuhl D, Huganir RL, Worley PF (2006) Arc/Arg3.1 interacts with the endocytic machinery to regulate AMPA receptor trafficking. Neuron 52(3):445–459. doi:10.1016/j.neuron.2006.08.033

    Google Scholar 

  33. Fumagalli F, Frasca A, Racagni G, Riva MA (2008) Dynamic regulation of glutamatergic postsynaptic activity in rat prefrontal cortex by repeated administration of antipsychotic drugs. Mol Pharmacol 73(5):1484–1490. doi:10.1124/mol.107.043786

    Article  PubMed  CAS  Google Scholar 

  34. Lu W, Ziff EB (2005) PICK1 interacts with ABP/GRIP to regulate AMPA receptor trafficking. Neuron 47(3):407–421. doi:10.1016/j.neuron.2005.07.006

    Article  PubMed  CAS  Google Scholar 

  35. Citri A, Bhattacharyya S, Ma C, Morishita W, Fang S, Rizo J, Malenka RC (2010) Calcium binding to PICK1 is essential for the intracellular retention of AMPA receptors underlying long-term depression. J Neurosci Off J Soc Neurosci 30(49):16437–16452. doi:10.1523/JNEUROSCI.4478-10.2010

    Article  CAS  Google Scholar 

  36. Hanley LJ, Henley JM (2010) Differential roles of GRIP1a and GRIP1b in AMPA receptor trafficking. Neurosci Lett 485(3):167–172. doi:10.1016/j.neulet.2010.09.003

    Article  PubMed  CAS  Google Scholar 

  37. Kulangara K, Kropf M, Glauser L, Magnin S, Alberi S, Yersin A, Hirling H (2007) Phosphorylation of glutamate receptor interacting protein 1 regulates surface expression of glutamate receptors. J Biol Chem 282(4):2395–2404. doi:10.1074/jbc.M606471200

    Article  PubMed  CAS  Google Scholar 

  38. Jo J, Son GH, Winters BL, Kim MJ, Whitcomb DJ, Dickinson BA, Lee YB, Futai K, Amici M, Sheng M, Collingridge GL, Cho K (2010) Muscarinic receptors induce LTD of NMDAR EPSCs via a mechanism involving hippocalcin, AP2 and PSD-95. Nat Neurosci 13(10):1216–1224. doi:10.1038/nn.2636

    Article  PubMed  CAS  Google Scholar 

  39. Steiner P, Higley MJ, Xu W, Czervionke BL, Malenka RC, Sabatini BL (2008) Destabilization of the postsynaptic density by PSD-95 serine 73 phosphorylation inhibits spine growth and synaptic plasticity. Neuron 60(5):788–802. doi:10.1016/j.neuron.2008.10.014

    Article  PubMed  CAS  Google Scholar 

  40. Verpelli C, Dvoretskova E, Vicidomini C, Rossi F, Chiappalone M, Schoen M, Di Stefano B, Mantegazza R, Broccoli V, Bockers TM, Dityatev A, Sala C (2011) Importance of Shank3 protein in regulating metabotropic glutamate receptor 5 (mGluR5) expression and signaling at synapses. J Biol Chem 286(40):34839–34850. doi:10.1074/jbc.M111.258384

    Article  PubMed  CAS  Google Scholar 

  41. Lennertz L, Wagner M, Wolwer W, Schuhmacher A, Frommann I, Berning J, Schulze-Rauschenbach S, Landsberg MW, Steinbrecher A, Alexander M, Franke PE, Pukrop R, Ruhrmann S, Bechdolf A, Gaebel W, Klosterkotter J, Hafner H, Maier W, Mossner R (2012) A promoter variant of SHANK1 affects auditory working memory in schizophrenia patients and in subjects clinically at risk for psychosis. Eur Arch Psychiatry Clin Neurosci 262(2):117–124. doi:10.1007/s00406-011-0233-3

    Article  PubMed  Google Scholar 

  42. Delahaye A, Toutain A, Aboura A, Dupont C, Tabet AC, Benzacken B, Elion J, Verloes A, Pipiras E, Drunat S (2009) Chromosome 22q13.3 deletion syndrome with a de novo interstitial 22q13.3 cryptic deletion disrupting SHANK3. Eur J Med Genet 52(5):328–332. doi:10.1016/j.ejmg.2009.05.004

    Google Scholar 

  43. Durand CM, Betancur C, Boeckers TM, Bockmann J, Chaste P, Fauchereau F, Nygren G, Rastam M, Gillberg IC, Anckarsater H, Sponheim E, Goubran-Botros H, Delorme R, Chabane N, Mouren-Simeoni MC, de Mas P, Bieth E, Roge B, Heron D, Burglen L, Gillberg C, Leboyer M, Bourgeron T (2007) Mutations in the gene encoding the synaptic scaffolding protein SHANK3 are associated with autism spectrum disorders. Nat Genet 39(1):25–27. doi:10.1038/ng1933

    Article  PubMed  CAS  Google Scholar 

  44. Moessner R, Marshall CR, Sutcliffe JS, Skaug J, Pinto D, Vincent J, Zwaigenbaum L, Fernandez B, Roberts W, Szatmari P, Scherer SW (2007) Contribution of SHANK3 mutations to autism spectrum disorder. Am J Hum Genet 81(6):1289–1297. doi:10.1086/522590

    Article  PubMed  CAS  Google Scholar 

  45. Penzes P, Johnson RC, Alam MR, Kambampati V, Mains RE, Eipper BA (2000) An isoform of kalirin, a brain-specific GDP/GTP exchange factor, is enriched in the postsynaptic density fraction. J Biol Chem 275(9):6395–6403

    Article  PubMed  CAS  Google Scholar 

  46. Penzes P, Cahill ME, Jones KA, VanLeeuwen JE, Woolfrey KM (2011) Dendritic spine pathology in neuropsychiatric disorders. Nat Neurosci 14(3):285–293. doi:10.1038/nn.2741

    Article  PubMed  CAS  Google Scholar 

  47. Penzes P, Johnson RC, Sattler R, Zhang X, Huganir RL, Kambampati V, Mains RE, Eipper BA (2001) The neuronal Rho-GEF Kalirin-7 interacts with PDZ domain-containing proteins and regulates dendritic morphogenesis. Neuron 29(1):229–242

    Article  PubMed  CAS  Google Scholar 

  48. Xie Z, Srivastava DP, Photowala H, Kai L, Cahill ME, Woolfrey KM, Shum CY, Surmeier DJ, Penzes P (2007) Kalirin-7 controls activity-dependent structural and functional plasticity of dendritic spines. Neuron 56(4):640–656. doi:10.1016/j.neuron.2007.10.005

    Article  PubMed  CAS  Google Scholar 

  49. Kiraly DD, Lemtiri-Chlieh F, Levine ES, Mains RE, Eipper BA (2011) Kalirin binds the NR2B subunit of the NMDA receptor, altering its synaptic localization and function. J Neurosci Off J Soc Neurosci 31(35):12554–12565. doi:10.1523/JNEUROSCI.3143-11.2011

    Article  CAS  Google Scholar 

  50. Ma XM, Wang Y, Ferraro F, Mains RE, Eipper BA (2008) Kalirin-7 is an essential component of both shaft and spine excitatory synapses in hippocampal interneurons. J Neurosci 28(3):711–724. doi:10.1523/JNEUROSCI.5283-07.2008

    Article  PubMed  CAS  Google Scholar 

  51. Cahill ME, Xie Z, Day M, Photowala H, Barbolina MV, Miller CA, Weiss C, Radulovic J, Sweatt JD, Disterhoft JF, Surmeier DJ, Penzes P (2009) Kalirin regulates cortical spine morphogenesis and disease-related behavioral phenotypes. Proc Natl Acad Sci USA 106(31):13058–13063. doi:10.1073/pnas.0904636106

    Article  PubMed  CAS  Google Scholar 

  52. Ma XM, Kiraly DD, Gaier ED, Wang Y, Kim EJ, Levine ES, Eipper BA, Mains RE (2008) Kalirin-7 is required for synaptic structure and function. J Neurosci 28(47):12368–12382. doi:10.1523/JNEUROSCI.4269-08.2008

    Article  PubMed  CAS  Google Scholar 

  53. Lemtiri-Chlieh F, Zhao L, Kiraly DD, Eipper BA, Mains RE, Levine ES (2011) Kalirin-7 is necessary for normal NMDA receptor-dependent synaptic plasticity. BMC Neurosci 12:126. doi:10.1186/1471-2202-12-126

    Article  PubMed  CAS  Google Scholar 

  54. Kushima I, Nakamura Y, Aleksic B, Ikeda M, Ito Y, Shiino T, Okochi T, Fukuo Y, Ujike H, Suzuki M, Inada T, Hashimoto R, Takeda M, Kaibuchi K, Iwata N, Ozaki N (2012) Resequencing and association analysis of the KALRN and EPHB1 genes and their contribution to schizophrenia susceptibility. Schizophr Bull 38(3):552–560. doi:10.1093/schbul/sbq118

    Google Scholar 

  55. Zhao Y, Hegde AN, Martin KC (2003) The ubiquitin proteasome system functions as an inhibitory constraint on synaptic strengthening. Curr Biol 13(11):887–898

    Article  PubMed  CAS  Google Scholar 

  56. Yashiro K, Riday TT, Condon KH, Roberts AC, Bernardo DR, Prakash R, Weinberg RJ, Ehlers MD, Philpot BD (2009) Ube3a is required for experience-dependent maturation of the neocortex. Nat Neurosci 12(6):777–783. doi:10.1038/nn.2327

    Article  PubMed  CAS  Google Scholar 

  57. Yeh SH, Mao SC, Lin HC, Gean PW (2006) Synaptic expression of glutamate receptor after encoding of fear memory in the rat amygdala. Mol Pharmacol 69(1):299–308. doi:10.1124/mol.105.017194

    PubMed  CAS  Google Scholar 

  58. Mao SC, Lin HC, Gean PW (2008) Augmentation of fear extinction by D-cycloserine is blocked by proteasome inhibitors. Neuropsychopharmacology Off Publ Am Coll Neuropsychopharmacol 33(13):3085–3095. doi:10.1038/npp.2008.30

    Article  CAS  Google Scholar 

  59. Dong C, Upadhya SC, Ding L, Smith TK, Hegde AN (2008) Proteasome inhibition enhances the induction and impairs the maintenance of late-phase long-term potentiation. Learn Mem 15(5):335–347. doi:10.1101/lm.984508

    Article  PubMed  CAS  Google Scholar 

  60. Kato A, Rouach N, Nicoll RA, Bredt DS (2005) Activity-dependent NMDA receptor degradation mediated by retrotranslocation and ubiquitination. Proc Natl Acad Sci USA 102(15):5600–5605. doi:10.1073/pnas.0501769102

    Article  PubMed  CAS  Google Scholar 

  61. Jurd R, Thornton C, Wang J, Luong K, Phamluong K, Kharazia V, Gibb SL, Ron D (2008) Mind bomb-2 is an E3 ligase that ubiquitinates the N-methyl-d-aspartate receptor NR2B subunit in a phosphorylation-dependent manner. J Biol Chem 283(1):301–310. doi:10.1074/jbc.M705580200

    Article  PubMed  CAS  Google Scholar 

  62. Mao LM, Wang W, Chu XP, Zhang GC, Liu XY, Yang YJ, Haines M, Papasian CJ, Fibuch EE, Buch S, Chen JG, Wang JQ (2009) Stability of surface NMDA receptors controls synaptic and behavioral adaptations to amphetamine. Nat Neurosci 12(5):602–610. doi:10.1038/nn.2300

    Article  PubMed  CAS  Google Scholar 

  63. Park EC, Glodowski DR, Rongo C (2009) The ubiquitin ligase RPM-1 and the p38 MAPK PMK-3 regulate AMPA receptor trafficking. PLoS ONE 4(1):e4284. doi:10.1371/journal.pone.0004284

    Article  PubMed  CAS  Google Scholar 

  64. Burbea M, Dreier L, Dittman JS, Grunwald ME, Kaplan JM (2002) Ubiquitin and AP180 regulate the abundance of GLR-1 glutamate receptors at postsynaptic elements in C. elegans. Neuron 35(1):107–120

    Article  PubMed  CAS  Google Scholar 

  65. Schaefer H, Rongo C (2006) KEL-8 is a substrate receptor for CUL3-dependent ubiquitin ligase that regulates synaptic glutamate receptor turnover. Mol Biol Cell 17(3):1250–1260. doi:10.1091/mbc.E05-08-0794

    Article  PubMed  CAS  Google Scholar 

  66. Pavlopoulos E, Trifilieff P, Chevaleyre V, Fioriti L, Zairis S, Pagano A, Malleret G, Kandel ER (2011) Neuralized1 activates CPEB3: a function for nonproteolytic ubiquitin in synaptic plasticity and memory storage. Cell 147(6):1369–1383. doi:10.1016/j.cell.2011.09.056

    Article  PubMed  CAS  Google Scholar 

  67. Lee SH, Choi JH, Lee N, Lee HR, Kim JI, Yu NK, Choi SL, Kim H, Kaang BK (2008) Synaptic protein degradation underlies destabilization of retrieved fear memory. Science 319(5867):1253–1256. doi:10.1126/science.1150541

    Article  PubMed  CAS  Google Scholar 

  68. Spangler SA, Hoogenraad CC (2007) Liprin-alpha proteins: scaffold molecules for synapse maturation. Biochem Soc Trans 35(Pt 5):1278–1282. doi:10.1042/BST0351278

    PubMed  CAS  Google Scholar 

  69. Wyszynski M, Kim E, Dunah AW, Passafaro M, Valtschanoff JG, Serra-Pages C, Streuli M, Weinberg RJ, Sheng M (2002) Interaction between GRIP and liprin-alpha/SYD2 is required for AMPA receptor targeting. Neuron 34(1):39–52

    Article  PubMed  CAS  Google Scholar 

  70. van Roessel P, Elliott DA, Robinson IM, Prokop A, Brand AH (2004) Independent regulation of synaptic size and activity by the anaphase-promoting complex. Cell 119(5):707–718. doi:10.1016/j.cell.2004.11.028

    Article  PubMed  Google Scholar 

  71. Hoogenraad CC, Feliu-Mojer MI, Spangler SA, Milstein AD, Dunah AW, Hung AY, Sheng M (2007) Liprinalpha1 degradation by calcium/calmodulin-dependent protein kinase II regulates LAR receptor tyrosine phosphatase distribution and dendrite development. Dev Cell 12(4):587–602. doi:10.1016/j.devcel.2007.02.006

    Article  PubMed  CAS  Google Scholar 

  72. Djakovic SN, Schwarz LA, Barylko B, DeMartino GN, Patrick GN (2009) Regulation of the proteasome by neuronal activity and calcium/calmodulin-dependent protein kinase II. J Biol Chem 284(39):26655–26665. doi:10.1074/jbc.M109.021956

    Article  PubMed  CAS  Google Scholar 

  73. Bingol B, Wang CF, Arnott D, Cheng D, Peng J, Sheng M (2010) Autophosphorylated CaMKIIalpha acts as a scaffold to recruit proteasomes to dendritic spines. Cell 140(4):567–578. doi:10.1016/j.cell.2010.01.024

    Article  PubMed  CAS  Google Scholar 

  74. Colledge M, Snyder EM, Crozier RA, Soderling JA, Jin Y, Langeberg LK, Lu H, Bear MF, Scott JD (2003) Ubiquitination regulates PSD-95 degradation and AMPA receptor surface expression. Neuron 40(3):595–607

    Article  PubMed  CAS  Google Scholar 

  75. Guo L, Wang Y (2007) Glutamate stimulates glutamate receptor interacting protein 1 degradation by ubiquitin-proteasome system to regulate surface expression of GluR2. Neuroscience 145(1):100–109. doi:10.1016/j.neuroscience.2006.11.042

    Article  PubMed  CAS  Google Scholar 

  76. Yang H, Takagi H, Konishi Y, Ageta H, Ikegami K, Yao I, Sato S, Hatanaka K, Inokuchi K, Seog DH, Setou M (2008) Transmembrane and ubiquitin-like domain-containing protein 1 (Tmub1/HOPS) facilitates surface expression of GluR2-containing AMPA receptors. PLoS ONE 3(7):e2809. doi:10.1371/journal.pone.0002809

    Article  PubMed  CAS  Google Scholar 

  77. Dodd PR, Scott HL, Westphalen RI (1994) Excitotoxic mechanisms in the pathogenesis of dementia. Neurochem Int 25(3):203–219

    Article  PubMed  CAS  Google Scholar 

  78. Gong Y, Lippa CF (2010) Review: disruption of the postsynaptic density in Alzheimer’s disease and other neurodegenerative dementias. Am J Alzheimers Dis Other Demen 25(7):547–555. doi:10.1177/1533317510382893

    Article  PubMed  Google Scholar 

  79. Ehlers MD (2003) Activity level controls postsynaptic composition and signaling via the ubiquitin-proteasome system. Nat Neurosci 6(3):231–242. doi:10.1038/nn1013

    Article  PubMed  CAS  Google Scholar 

  80. Sjostrom PJ, Rancz EA, Roth A, Hausser M (2008) Dendritic excitability and synaptic plasticity. Physiol Rev 88(2):769–840. doi:10.1152/physrev.00016.2007

    Article  PubMed  CAS  Google Scholar 

  81. Almeida CG, Tampellini D, Takahashi RH, Greengard P, Lin MT, Snyder EM, Gouras GK (2005) Beta-amyloid accumulation in APP mutant neurons reduces PSD-95 and GluR1 in synapses. Neurobiol Dis 20(2):187–198. doi:10.1016/j.nbd.2005.02.008

    Article  PubMed  CAS  Google Scholar 

  82. Louneva N, Cohen JW, Han LY, Talbot K, Wilson RS, Bennett DA, Trojanowski JQ, Arnold SE (2008) Caspase-3 is enriched in postsynaptic densities and increased in Alzheimer’s disease. Am J Pathol 173(5):1488–1495. doi:10.2353/ajpath.2008.080434

    Article  PubMed  CAS  Google Scholar 

  83. Lacor PN, Buniel MC, Chang L, Fernandez SJ, Gong Y, Viola KL, Lambert MP, Velasco PT, Bigio EH, Finch CE, Krafft GA, Klein WL (2004) Synaptic targeting by Alzheimer’s-related amyloid beta oligomers. J Neurosci 24(45):10191–10200. doi:10.1523/JNEUROSCI.3432-04.2004

    Article  PubMed  CAS  Google Scholar 

  84. Gong Y, Lippa CF, Zhu J, Lin Q, Rosso AL (2009) Disruption of glutamate receptors at Shank-postsynaptic platform in Alzheimer’s disease. Brain Res 1292:191–198. doi:10.1016/j.brainres.2009.07.056

    Article  PubMed  CAS  Google Scholar 

  85. Koffie RM, Meyer-Luehmann M, Hashimoto T, Adams KW, Mielke ML, Garcia-Alloza M, Micheva KD, Smith SJ, Kim ML, Lee VM, Hyman BT, Spires-Jones TL (2009) Oligomeric amyloid beta associates with postsynaptic densities and correlates with excitatory synapse loss near senile plaques. Proc Natl Acad Sci U S A 106(10):4012–4017. doi:10.1073/pnas.0811698106

    Article  PubMed  CAS  Google Scholar 

  86. Roselli F, Tirard M, Lu J, Hutzler P, Lamberti P, Livrea P, Morabito M, Almeida OF (2005) Soluble beta-amyloid1-40 induces NMDA-dependent degradation of postsynaptic density-95 at glutamatergic synapses. J Neurosci 25(48):11061–11070. doi:10.1523/JNEUROSCI.3034-05.2005

    Article  PubMed  CAS  Google Scholar 

  87. Gylys KH, Fein JA, Yang F, Wiley DJ, Miller CA, Cole GM (2004) Synaptic changes in Alzheimer’s disease: increased amyloid-beta and gliosis in surviving terminals is accompanied by decreased PSD-95 fluorescence. Am J Pathol 165(5):1809–1817

    Article  PubMed  CAS  Google Scholar 

  88. Leuba G, Savioz A, Vernay A, Carnal B, Kraftsik R, Tardif E, Riederer I, Riederer BM (2008) Differential changes in synaptic proteins in the Alzheimer frontal cortex with marked increase in PSD-95 postsynaptic protein. J Alzheimer’s Dis JAD 15(1):139–151

    CAS  Google Scholar 

  89. Nyffeler M, Zhang WN, Feldon J, Knuesel I (2007) Differential expression of PSD proteins in age-related spatial learning impairments. Neurobiol Aging 28(1):143–155. doi:10.1016/j.neurobiolaging.2005.11.003

    Article  PubMed  CAS  Google Scholar 

  90. Shao CY, Mirra SS, Sait HB, Sacktor TC, Sigurdsson EM (2011) Postsynaptic degeneration as revealed by PSD-95 reduction occurs after advanced Abeta and tau pathology in transgenic mouse models of Alzheimer’s disease. Acta Neuropathol 122(3):285–292. doi:10.1007/s00401-011-0843-x

    Article  PubMed  CAS  Google Scholar 

  91. Roselli F, Livrea P, Almeida OF (2011) CDK5 is essential for soluble amyloid beta-induced degradation of GKAP and remodeling of the synaptic actin cytoskeleton. PLoS ONE 6(7):e23097. doi:10.1371/journal.pone.0023097

    Article  PubMed  CAS  Google Scholar 

  92. Roselli F, Hutzler P, Wegerich Y, Livrea P, Almeida OF (2009) Disassembly of shank and homer synaptic clusters is driven by soluble beta-amyloid(1–40) through divergent NMDAR-dependent signalling pathways. PLoS ONE 4(6):e6011. doi:10.1371/journal.pone.0006011

    Article  PubMed  CAS  Google Scholar 

  93. Dickey CA, Loring JF, Montgomery J, Gordon MN, Eastman PS, Morgan D (2003) Selectively reduced expression of synaptic plasticity-related genes in amyloid precursor protein + presenilin-1 transgenic mice. J Neurosci 23(12):5219–5226

    PubMed  CAS  Google Scholar 

  94. Clemmensen C, Aznar S, Knudsen GM, Klein AB (2012) The microtubule-associated protein 1A (MAP1A) is an early molecular target of soluble abeta-peptide. Cell Mol Neurobiol 32(4):561–566. doi:10.1007/s10571-011-9796-9

    Google Scholar 

  95. Youn H, Ji I, Ji HP, Markesbery WR, Ji TH (2007) Under-expression of Kalirin-7 increases iNOS activity in cultured cells and correlates to elevated iNOS activity in Alzheimer’s disease hippocampus. J Alzheimers Dis 12(3):271–281

    PubMed  CAS  Google Scholar 

  96. Blanpied TA, Ehlers MD (2004) Microanatomy of dendritic spines: emerging principles of synaptic pathology in psychiatric and neurological disease. Biol Psychiatry 55(12):1121–1127. doi:10.1016/j.biopsych.2003.10.006

    Article  PubMed  Google Scholar 

  97. Pfeiffer BE, Huber KM (2009) The state of synapses in fragile X syndrome. Neuroscientist 15(5):549–567. doi:10.1177/1073858409333075

    Article  PubMed  CAS  Google Scholar 

  98. Muddashetty RS, Kelic S, Gross C, Xu M, Bassell GJ (2007) Dysregulated metabotropic glutamate receptor-dependent translation of AMPA receptor and postsynaptic density-95 mRNAs at synapses in a mouse model of fragile X syndrome. J Neurosci 27(20):5338–5348. doi:10.1523/JNEUROSCI.0937-07.2007

    Article  PubMed  CAS  Google Scholar 

  99. Bassell GJ, Warren ST (2008) Fragile X syndrome: loss of local mRNA regulation alters synaptic development and function. Neuron 60(2):201–214. doi:10.1016/j.neuron.2008.10.004

    Article  PubMed  CAS  Google Scholar 

  100. Luscher C, Huber KM (2010) Group 1 mGluR-dependent synaptic long-term depression: mechanisms and implications for circuitry and disease. Neuron 65(4):445–459. doi:10.1016/j.neuron.2010.01.016

    Article  PubMed  CAS  Google Scholar 

  101. Dolen G, Bear MF (2008) Role for metabotropic glutamate receptor 5 (mGluR5) in the pathogenesis of fragile X syndrome. J Physiol 586(6):1503–1508. doi:10.1113/jphysiol.2008.150722

    Article  PubMed  CAS  Google Scholar 

  102. Zalfa F, Eleuteri B, Dickson KS, Mercaldo V, De Rubeis S, di Penta A, Tabolacci E, Chiurazzi P, Neri G, Grant SG, Bagni C (2007) A new function for the fragile X mental retardation protein in regulation of PSD-95 mRNA stability. Nat Neurosci 10(5):578–587. doi:10.1038/nn1893

    Article  PubMed  CAS  Google Scholar 

  103. Tarpey P, Parnau J, Blow M, Woffendin H, Bignell G, Cox C, Cox J, Davies H, Edkins S, Holden S, Korny A, Mallya U, Moon J, O’Meara S, Parker A, Stephens P, Stevens C, Teague J, Donnelly A, Mangelsdorf M, Mulley J, Partington M, Turner G, Stevenson R, Schwartz C, Young I, Easton D, Bobrow M, Futreal PA, Stratton MR, Gecz J, Wooster R, Raymond FL (2004) Mutations in the DLG3 gene cause nonsyndromic X-linked mental retardation. Am J Hum Genet 75(2):318–324. doi:10.1086/422703

    Article  PubMed  CAS  Google Scholar 

  104. Zanni G, van Esch H, Bensalem A, Saillour Y, Poirier K, Castelnau L, Ropers HH, de Brouwer AP, Laumonnier F, Fryns JP, Chelly J (2010) A novel mutation in the DLG3 gene encoding the synapse-associated protein 102 (SAP102) causes non-syndromic mental retardation. Neurogenetics 11(2):251–255. doi:10.1007/s10048-009-0224-y

    Article  PubMed  CAS  Google Scholar 

  105. Cuthbert PC, Stanford LE, Coba MP, Ainge JA, Fink AE, Opazo P, Delgado JY, Komiyama NH, O’Dell TJ, Grant SG (2007) Synapse-associated protein 102/dlgh3 couples the NMDA receptor to specific plasticity pathways and learning strategies. J Neurosci 27(10):2673–2682. doi:10.1523/JNEUROSCI.4457-06.2007

    Article  PubMed  CAS  Google Scholar 

  106. Chen BS, Thomas EV, Sanz-Clemente A, Roche KW (2011) NMDA receptor-dependent regulation of dendritic spine morphology by SAP102 splice variants. J Neurosci 31(1):89–96. doi:10.1523/JNEUROSCI.1034-10.2011

    Article  PubMed  CAS  Google Scholar 

  107. Hung AY, Futai K, Sala C, Valtschanoff JG, Ryu J, Woodworth MA, Kidd FL, Sung CC, Miyakawa T, Bear MF, Weinberg RJ, Sheng M (2008) Smaller dendritic spines, weaker synaptic transmission, but enhanced spatial learning in mice lacking Shank1. J Neurosci 28(7):1697–1708. doi:10.1523/JNEUROSCI.3032-07.2008

    Article  PubMed  CAS  Google Scholar 

  108. Berkel S, Tang W, Trevino M, Vogt M, Obenhaus HA, Gass P, Scherer SW, Sprengel R, Schratt G, Rappold GA (2012) Inherited and de novo SHANK2 variants associated with autism spectrum disorder impair neuronal morphogenesis and physiology. Hum Mol Genet 21(2):344–357. doi:10.1093/hmg/ddr470

    Article  PubMed  CAS  Google Scholar 

  109. Durand CM, Perroy J, Loll F, Perrais D, Fagni L, Bourgeron T, Montcouquiol M, Sans N (2012) SHANK3 mutations identified in autism lead to modification of dendritic spine morphology via an actin-dependent mechanism. Mol Psychiatry 17(1):71–84. doi:10.1038/mp.2011.57

    Article  PubMed  CAS  Google Scholar 

  110. Bangash MA, Park JM, Melnikova T, Wang D, Jeon SK, Lee D, Syeda S, Kim J, Kouser M, Schwartz J, Cui Y, Zhao X, Speed HE, Kee SE, Tu JC, Hu JH, Petralia RS, Linden DJ, Powell CM, Savonenko A, Xiao B, Worley PF (2011) Enhanced polyubiquitination of Shank3 and NMDA receptor in a mouse model of autism. Cell 145(5):758–772. doi:10.1016/j.cell.2011.03.052

    Article  PubMed  CAS  Google Scholar 

  111. Ichtchenko K, Nguyen T, Sudhof TC (1996) Structures, alternative splicing, and neurexin binding of multiple neuroligins. J Biol Chem 271(5):2676–2682

    Article  PubMed  CAS  Google Scholar 

  112. Tabuchi K, Blundell J, Etherton MR, Hammer RE, Liu X, Powell CM, Sudhof TC (2007) A neuroligin-3 mutation implicated in autism increases inhibitory synaptic transmission in mice. Science 318(5847):71–76. doi:10.1126/science.1146221

    Article  PubMed  CAS  Google Scholar 

  113. Mondin M, Labrousse V, Hosy E, Heine M, Tessier B, Levet F, Poujol C, Blanchet C, Choquet D, Thoumine O (2011) Neurexin-neuroligin adhesions capture surface-diffusing AMPA receptors through PSD-95 scaffolds. J Neurosci 31(38):13500–13515. doi:10.1523/JNEUROSCI.6439-10.2011

    Article  PubMed  CAS  Google Scholar 

  114. Tabrizi SJ, Cleeter MW, Xuereb J, Taanman JW, Cooper JM, Schapira AH (1999) Biochemical abnormalities and excitotoxicity in Huntington’s disease brain. Ann Neurol 45(1):25–32

    Article  PubMed  CAS  Google Scholar 

  115. Vonsattel JP, DiFiglia M (1998) Huntington disease. J Neuropathol Exp Neurol 57(5):369–384

    Article  PubMed  CAS  Google Scholar 

  116. Trottier Y, Lutz Y, Stevanin G, Imbert G, Devys D, Cancel G, Saudou F, Weber C, David G, Tora L et al (1995) Polyglutamine expansion as a pathological epitope in Huntington’s disease and four dominant cerebellar ataxias. Nature 378(6555):403–406. doi:10.1038/378403a0

    Article  PubMed  CAS  Google Scholar 

  117. Hodgson JG, Agopyan N, Gutekunst CA, Leavitt BR, LePiane F, Singaraja R, Smith DJ, Bissada N, McCutcheon K, Nasir J, Jamot L, Li XJ, Stevens ME, Rosemond E, Roder JC, Phillips AG, Rubin EM, Hersch SM, Hayden MR (1999) A YAC mouse model for Huntington’s disease with full-length mutant huntingtin, cytoplasmic toxicity, and selective striatal neurodegeneration. Neuron 23(1):181–192

    Article  PubMed  CAS  Google Scholar 

  118. Shehadeh J, Fernandes HB, Zeron Mullins MM, Graham RK, Leavitt BR, Hayden MR, Raymond LA (2006) Striatal neuronal apoptosis is preferentially enhanced by NMDA receptor activation in YAC transgenic mouse model of Huntington disease. Neurobiol Dis 21(2):392–403. doi:10.1016/j.nbd.2005.08.001

    Article  PubMed  CAS  Google Scholar 

  119. Fan MM, Fernandes HB, Zhang LY, Hayden MR, Raymond LA (2007) Altered NMDA receptor trafficking in a yeast artificial chromosome transgenic mouse model of Huntington’s disease. J Neurosci 27(14):3768–3779. doi:10.1523/JNEUROSCI.4356-06.2007

    Article  PubMed  CAS  Google Scholar 

  120. Sun Y, Savanenin A, Reddy PH, Liu YF (2001) Polyglutamine-expanded huntingtin promotes sensitization of N-methyl-d-aspartate receptors via post-synaptic density 95. J Biol Chem 276(27):24713–24718. doi:10.1074/jbc.M103501200

    Article  PubMed  CAS  Google Scholar 

  121. Fan J, Cowan CM, Zhang LY, Hayden MR, Raymond LA (2009) Interaction of postsynaptic density protein-95 with NMDA receptors influences excitotoxicity in the yeast artificial chromosome mouse model of Huntington’s disease. J Neurosci 29(35):10928–10938. doi:10.1523/JNEUROSCI.2491-09.2009

    Article  PubMed  CAS  Google Scholar 

  122. Hallett PJ, Dunah AW, Ravenscroft P, Zhou S, Bezard E, Crossman AR, Brotchie JM, Standaert DG (2005) Alterations of striatal NMDA receptor subunits associated with the development of dyskinesia in the MPTP-lesioned primate model of Parkinson’s disease. Neuropharmacology 48(4):503–516. doi:10.1016/j.neuropharm.2004.11.008

    Article  PubMed  CAS  Google Scholar 

  123. Picconi B, Centonze D, Rossi S, Bernardi G, Calabresi P (2004) Therapeutic doses of l-dopa reverse hypersensitivity of corticostriatal D2-dopamine receptors and glutamatergic overactivity in experimental parkinsonism. Brain 127(Pt 7):1661–1669. doi:10.1093/brain/awh190

    Article  PubMed  Google Scholar 

  124. Gardoni F, Picconi B, Ghiglieri V, Polli F, Bagetta V, Bernardi G, Cattabeni F, Di Luca M, Calabresi P (2006) A critical interaction between NR2B and MAGUK in L-DOPA induced dyskinesia. J Neurosci 26(11):2914–2922. doi:10.1523/JNEUROSCI.5326-05.2006

    Article  PubMed  CAS  Google Scholar 

  125. Cartier AE, Djakovic SN, Salehi A, Wilson SM, Masliah E, Patrick GN (2009) Regulation of synaptic structure by ubiquitin C-terminal hydrolase L1. J Neurosci Off J Soc Neurosci 29(24):7857–7868. doi:10.1523/JNEUROSCI.1817-09.2009

    Article  CAS  Google Scholar 

  126. Shimura H, Hattori N, Kubo S, Mizuno Y, Asakawa S, Minoshima S, Shimizu N, Iwai K, Chiba T, Tanaka K, Suzuki T (2000) Familial Parkinson disease gene product, parkin, is a ubiquitin-protein ligase. Nat Genet 25(3):302–305. doi:10.1038/77060

    Article  PubMed  CAS  Google Scholar 

  127. Joch M, Ase AR, Chen CX, MacDonald PA, Kontogiannea M, Corera AT, Brice A, Seguela P, Fon EA (2007) Parkin-mediated monoubiquitination of the PDZ protein PICK1 regulates the activity of acid-sensing ion channels. Mol Biol Cell 18(8):3105–3118. doi:10.1091/mbc.E05-11-1027

    Article  PubMed  CAS  Google Scholar 

  128. Helton TD, Otsuka T, Lee MC, Mu Y, Ehlers MD (2008) Pruning and loss of excitatory synapses by the parkin ubiquitin ligase. Proc Natl Acad Sci USA 105(49):19492–19497. doi:10.1073/pnas.0802280105

    Article  PubMed  CAS  Google Scholar 

  129. Park JS, Bateman MC, Goldberg MP (1996) Rapid alterations in dendrite morphology during sublethal hypoxia or glutamate receptor activation. Neurobiol Dis 3(3):215–227

    Article  PubMed  CAS  Google Scholar 

  130. Aarts MM, Tymianski M (2004) Molecular mechanisms underlying specificity of excitotoxic signaling in neurons. Curr Mol Med 4(2):137–147

    Article  PubMed  CAS  Google Scholar 

  131. Sattler R, Xiong Z, Lu WY, Hafner M, MacDonald JF, Tymianski M (1999) Specific coupling of NMDA receptor activation to nitric oxide neurotoxicity by PSD-95 protein. Science 284(5421):1845–1848

    Article  PubMed  CAS  Google Scholar 

  132. Aarts M, Liu Y, Liu L, Besshoh S, Arundine M, Gurd JW, Wang YT, Salter MW, Tymianski M (2002) Treatment of ischemic brain damage by perturbing NMDA receptor- PSD-95 protein interactions. Science 298(5594):846–850. doi:10.1126/science.1072873

    Article  PubMed  CAS  Google Scholar 

  133. Cui H, Hayashi A, Sun HS, Belmares MP, Cobey C, Phan T, Schweizer J, Salter MW, Wang YT, Tasker RA, Garman D, Rabinowitz J, Lu PS, Tymianski M (2007) PDZ protein interactions underlying NMDA receptor-mediated excitotoxicity and neuroprotection by PSD-95 inhibitors. J Neurosci 27(37):9901–9915. doi:10.1523/JNEUROSCI.1464-07.2007

    Article  PubMed  CAS  Google Scholar 

  134. Bratane BT, Cui H, Cook DJ, Bouley J, Tymianski M, Fisher M (2011) Neuroprotection by freezing ischemic penumbra evolution without cerebral blood flow augmentation with a postsynaptic density-95 protein inhibitor. Stroke 42(11):3265–3270. doi:10.1161/STROKEAHA.111.618801

    Article  PubMed  CAS  Google Scholar 

  135. Sun HS, Doucette TA, Liu Y, Fang Y, Teves L, Aarts M, Ryan CL, Bernard PB, Lau A, Forder JP, Salter MW, Wang YT, Tasker RA, Tymianski M (2008) Effectiveness of PSD95 inhibitors in permanent and transient focal ischemia in the rat. Stroke 39(9):2544–2553. doi:10.1161/STROKEAHA.107.506048

    Article  PubMed  CAS  Google Scholar 

  136. Zhou L, Li F, Xu HB, Luo CX, Wu HY, Zhu MM, Lu W, Ji X, Zhou QG, Zhu DY (2010) Treatment of cerebral ischemia by disrupting ischemia-induced interaction of nNOS with PSD-95. Nat Med 16(12):1439–1443. doi:10.1038/nm.2245

    Article  PubMed  CAS  Google Scholar 

  137. Kirov G, Pocklington AJ, Holmans P, Ivanov D, Ikeda M, Ruderfer D, Moran J, Chambert K, Toncheva D, Georgieva L, Grozeva D, Fjodorova M, Wollerton R, Rees E, Nikolov I, Lagemaat LN, Bayes A, Fernandez E, Olason PI, Bottcher Y, Komiyama NH, Collins MO, Choudhary J, Stefansson K, Stefansson H, Grant SG, Purcell S, Sklar P, O’Donovan MC, Owen MJ (2012) De novo CNV analysis implicates specific abnormalities of postsynaptic signalling complexes in the pathogenesis of schizophrenia. Mol Psychiatry 17(2):142–153. doi:10.1038/mp.2011.154

    Article  PubMed  CAS  Google Scholar 

  138. Clinton SM, Meador-Woodruff JH (2004) Abnormalities of the NMDA receptor and associated intracellular molecules in the thalamus in schizophrenia and bipolar disorder. Neuropsychopharmacology 29(7):1353–1362. doi:10.1038/sj.npp.1300451

    Article  PubMed  CAS  Google Scholar 

  139. Clinton SM, Haroutunian V, Davis KL, Meador-Woodruff JH (2003) Altered transcript expression of NMDA receptor-associated postsynaptic proteins in the thalamus of subjects with schizophrenia. Am J Psychiatry 160(6):1100–1109

    Article  PubMed  Google Scholar 

  140. Ohnuma T, Kato H, Arai H, Faull RL, McKenna PJ, Emson PC (2000) Gene expression of PSD95 in prefrontal cortex and hippocampus in schizophrenia. NeuroReport 11(14):3133–3137

    Article  PubMed  CAS  Google Scholar 

  141. Dracheva S, Marras SA, Elhakem SL, Kramer FR, Davis KL, Haroutunian V (2001) N-methyl-d-aspartic acid receptor expression in the dorsolateral prefrontal cortex of elderly patients with schizophrenia. Am J Psychiatry 158(9):1400–1410

    Article  PubMed  CAS  Google Scholar 

  142. Cheng MC, Lu CL, Luu SU, Tsai HM, Hsu SH, Chen TT, Chen CH (2010) Genetic and functional analysis of the DLG4 gene encoding the post-synaptic density protein 95 in schizophrenia. PLoS ONE 5(12):e15107. doi:10.1371/journal.pone.0015107

    Article  PubMed  CAS  Google Scholar 

  143. Spellmann I, Rujescu D, Musil R, Mayr A, Giegling I, Genius J, Zill P, Dehning S, Opgen-Rhein M, Cerovecki A, Hartmann AM, Schafer M, Bondy B, Muller N, Moller HJ, Riedel M (2011) Homer-1 polymorphisms are associated with psychopathology and response to treatment in schizophrenic patients. J Psychiatr Res 45(2):234–241. doi:10.1016/j.jpsychires.2010.06.004

    Article  PubMed  Google Scholar 

  144. Deo AJ, Cahill ME, Li S, Goldszer I, Henteleff R, Vanleeuwen JE, Rafalovich I, Gao R, Stachowski EK, Sampson AR, Lewis DA, Penzes P, Sweet RA (2012) Increased expression of Kalirin-9 in the auditory cortex of schizophrenia subjects: its role in dendritic pathology. Neurobiol Dis 45(2):796–803. doi:10.1016/j.nbd.2011.11.003

    Article  PubMed  CAS  Google Scholar 

  145. Kammermeier PJ (2008) Endogenous homer proteins regulate metabotropic glutamate receptor signaling in neurons. J Neurosci 28(34):8560–8567. doi:10.1523/JNEUROSCI.1830-08.2008

    Article  PubMed  CAS  Google Scholar 

  146. Sala C, Piech V, Wilson NR, Passafaro M, Liu G, Sheng M (2001) Regulation of dendritic spine morphology and synaptic function by Shank and Homer. Neuron 31(1):115–130

    Article  PubMed  CAS  Google Scholar 

  147. Sala C, Futai K, Yamamoto K, Worley PF, Hayashi Y, Sheng M (2003) Inhibition of dendritic spine morphogenesis and synaptic transmission by activity-inducible protein Homer1a. J Neurosci 23(15):6327–6337

    PubMed  CAS  Google Scholar 

  148. Celikel T, Marx V, Freudenberg F, Zivkovic A, Resnik E, Hasan MT, Licznerski P, Osten P, Rozov A, Seeburg PH, Schwarz MK (2007) Select overexpression of homer1a in dorsal hippocampus impairs spatial working memory. Front Neurosci 1(1):97–110. doi:10.3389/neuro.01.1.1.007.2007

    Article  PubMed  CAS  Google Scholar 

  149. Ueta Y, Yamamoto R, Sugiura S, Inokuchi K, Kato N (2008) Homer 1a suppresses neocortex long-term depression in a cortical layer-specific manner. J Neurophysiol 99(2):950–957. doi:10.1152/jn.01101.2007

    Article  PubMed  Google Scholar 

  150. Bertaso F, Roussignol G, Worley P, Bockaert J, Fagni L, Ango F (2010) Homer1a-dependent crosstalk between NMDA and metabotropic glutamate receptors in mouse neurons. PLoS ONE 5(3):e9755. doi:10.1371/journal.pone.0009755

    Article  PubMed  CAS  Google Scholar 

  151. Gerstein H, O’Riordan K, Osting S, Schwarz M, Burger C (2012) Rescue of synaptic plasticity and spatial learning deficits in the hippocampus of Homer1 knockout mice by recombinant Adeno-associated viral gene delivery of Homer1c. Neurobiol Learn Mem 97(1):17–29. doi:10.1016/j.nlm.2011.08.009

    Article  PubMed  CAS  Google Scholar 

  152. Szumlinski KK, Lominac KD, Kleschen MJ, Oleson EB, Dehoff MH, Schwarz MK, Seeburg PH, Worley PF, Kalivas PW (2005) Behavioral and neurochemical phenotyping of Homer1 mutant mice: possible relevance to schizophrenia. Genes Brain Behav 4(5):273–288. doi:10.1111/j.1601-183X.2005.00120.x

    Article  PubMed  CAS  Google Scholar 

  153. Hennou S, Kato A, Schneider EM, Lundstrom K, Gahwiler BH, Inokuchi K, Gerber U, Ehrengruber MU (2003) Homer-1a/Vesl-1S enhances hippocampal synaptic transmission. Eur J Neurosci 18(4):811–819

    Article  PubMed  Google Scholar 

  154. Inoue N, Nakao H, Migishima R, Hino T, Matsui M, Hayashi F, Nakao K, Manabe T, Aiba A, Inokuchi K (2009) Requirement of the immediate early gene vesl-1S/homer-1a for fear memory formation. Mol Brain 2:7. doi:10.1186/1756-6606-2-7

    Article  PubMed  CAS  Google Scholar 

  155. Tronson NC, Guzman YF, Guedea AL, Huh KH, Gao C, Schwarz MK, Radulovic J (2010) Metabotropic glutamate receptor 5/Homer interactions underlie stress effects on fear. Biol Psychiatry 68(11):1007–1015. doi:10.1016/j.biopsych.2010.09.004

    Article  PubMed  CAS  Google Scholar 

  156. Okvist A, Fagergren P, Whittard J, Garcia-Osta A, Drakenberg K, Horvath MC, Schmidt CJ, Keller E, Bannon MJ, Hurd YL (2011) Dysregulated postsynaptic density and endocytic zone in the amygdala of human heroin and cocaine abusers. Biol Psychiatry 69(3):245–252. doi:10.1016/j.biopsych.2010.09.037

    Article  PubMed  CAS  Google Scholar 

  157. Blanpied TA, Kerr JM, Ehlers MD (2008) Structural plasticity with preserved topology in the postsynaptic protein network. Proc Natl Acad Sci USA 105(34):12587–12592. doi:10.1073/pnas.0711669105

    Article  PubMed  CAS  Google Scholar 

  158. Newpher TM, Ehlers MD (2009) Spine microdomains for postsynaptic signaling and plasticity. Trends Cell Biol 19(5):218–227. doi:10.1016/j.tcb.2009.02.004

    Article  PubMed  Google Scholar 

  159. de Bartolomeis A, Sarappa C, Magara S, Iasevoli F (2012) Targeting glutamate system for novel antipsychotic approaches: relevance for residual psychotic symptoms and treatment resistant schizophrenia. Eur J Pharmacol 682(1–3):1–11. doi:10.1016/j.ejphar.2012.02.033

    Article  PubMed  CAS  Google Scholar 

  160. Zhang J, Vinuela A, Neely MH, Hallett PJ, Grant SG, Miller GM, Isacson O, Caron MG, Yao WD (2007) Inhibition of the dopamine D1 receptor signaling by PSD-95. J Biol Chem 282(21):15778–15789. doi:10.1074/jbc.M611485200

    Article  PubMed  CAS  Google Scholar 

  161. Yano M, Steiner H (2005) Methylphenidate (Ritalin) induces Homer 1a and zif 268 expression in specific corticostriatal circuits. Neuroscience 132(3):855–865. doi:10.1016/j.neuroscience.2004.12.019

    Article  PubMed  CAS  Google Scholar 

  162. Iasevoli F, Fiore G, Cicale M, Muscettola G, de Bartolomeis A (2010) Haloperidol induces higher Homer1a expression than risperidone, olanzapine and sulpiride in striatal sub-regions. Psychiatry Res 177(1–2):255–260. doi:10.1016/j.psychres.2010.02.009

    Article  PubMed  CAS  Google Scholar 

  163. Abbas AI, Yadav PN, Yao WD, Arbuckle MI, Grant SG, Caron MG, Roth BL (2009) PSD-95 is essential for hallucinogen and atypical antipsychotic drug actions at serotonin receptors. J Neurosci 29(22):7124–7136. doi:10.1523/JNEUROSCI.1090-09.2009

    Article  PubMed  CAS  Google Scholar 

  164. Tomasetti C, Dell’Aversano C, Iasevoli F, de Bartolomeis A (2007) Homer splice variants modulation within cortico-subcortical regions by dopamine D2 antagonists, a partial agonist, and an indirect agonist: implication for glutamatergic postsynaptic density in antipsychotics action. Neuroscience 150(1):144–158. doi:10.1016/j.neuroscience.2007.08.022

    Article  PubMed  CAS  Google Scholar 

  165. Ambesi-Impiombato A, Panariello F, Dell’aversano C, Tomasetti C, Muscettola G, de Bartolomeis A (2007) Differential expression of Homer 1 gene by acute and chronic administration of antipsychotics and dopamine transporter inhibitors in the rat forebrain. Synapse 61(6):429–439. doi:10.1002/syn.20385

    Article  PubMed  CAS  Google Scholar 

  166. Lominac KD, Oleson EB, Pava M, Klugmann M, Schwarz MK, Seeburg PH, During MJ, Worley PF, Kalivas PW, Szumlinski KK (2005) Distinct roles for different Homer1 isoforms in behaviors and associated prefrontal cortex function. J Neurosci 25(50):11586–11594. doi:10.1523/JNEUROSCI.3764-05.2005

    Article  PubMed  CAS  Google Scholar 

  167. Cook DJ, Teves L, Tymianski M (2012) Treatment of stroke with a PSD-95 inhibitor in the gyrencephalic primate brain. Nature 483(7388):213–217. doi:10.1038/nature10841

    Article  PubMed  CAS  Google Scholar 

  168. Bach A, Clausen BH, Moller M, Vestergaard B, Chi CN, Round A, Sorensen PL, Nissen KB, Kastrup JS, Gajhede M, Jemth P, Kristensen AS, Lundstrom P, Lambertsen KL, Stromgaard K (2012) A high-affinity, dimeric inhibitor of PSD-95 bivalently interacts with PDZ1-2 and protects against ischemic brain damage. Proc Natl Acad Sci USA 109(9):3317–3322. doi:10.1073/pnas.1113761109

    Article  PubMed  CAS  Google Scholar 

  169. Thorsen TS, Madsen KL, Rebola N, Rathje M, Anggono V, Bach A, Moreira IS, Stuhr-Hansen N, Dyhring T, Peters D, Beuming T, Huganir R, Weinstein H, Mulle C, Stromgaard K, Ronn LC, Gether U (2010) Identification of a small-molecule inhibitor of the PICK1 PDZ domain that inhibits hippocampal LTP and LTD. Proc Natl Acad Sci USA 107(1):413–418. doi:10.1073/pnas.0902225107

    Article  PubMed  CAS  Google Scholar 

Download references

Conflict of interest

Authors declare they have no conflict of interest.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Felice Iasevoli.

Rights and permissions

Reprints and permissions

About this article

Cite this article

Iasevoli, F., Tomasetti, C. & de Bartolomeis, A. Scaffolding Proteins of the Post-synaptic Density Contribute to Synaptic Plasticity by Regulating Receptor Localization and Distribution: Relevance for Neuropsychiatric Diseases. Neurochem Res 38, 1–22 (2013). https://doi.org/10.1007/s11064-012-0886-y

Download citation

  • Received:

  • Revised:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1007/s11064-012-0886-y

Keywords

Navigation