Skip to main content

Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.

  • Article
  • Published:

Brain-specific deletion of GIT1 impairs cognition and alters phosphorylation of synaptic protein networks implicated in schizophrenia susceptibility

Abstract

Despite tremendous effort, the molecular and cellular basis of cognitive deficits in schizophrenia remain poorly understood. Recent progress in elucidating the genetic architecture of schizophrenia has highlighted the association of multiple loci and rare variants that may impact susceptibility. One key example, given their potential etiopathogenic and therapeutic relevance, is a set of genes that encode proteins that regulate excitatory glutamatergic synapses in brain. A critical next step is to delineate specifically how such genetic variation impacts synaptic plasticity and to determine if and how the encoded proteins interact biochemically with one another to control cognitive function in a convergent manner. Towards this goal, here we study the roles of GPCR-kinase interacting protein 1 (GIT1), a synaptic scaffolding and signaling protein with damaging coding variants found in schizophrenia patients, as well as copy number variants found in patients with neurodevelopmental disorders. We generated conditional neural-selective GIT1 knockout mice and found that these mice have deficits in fear conditioning memory recall and spatial memory, as well as reduced cortical neuron dendritic spine density. Using global quantitative phospho-proteomics, we revealed that GIT1 deletion in brain perturbs specific networks of GIT1-interacting synaptic proteins. Importantly, several schizophrenia and neurodevelopmental disorder risk genes are present within these networks. We propose that GIT1 regulates the phosphorylation of a network of synaptic proteins and other critical regulators of neuroplasticity, and that perturbation of these networks may contribute specifically to cognitive deficits observed in schizophrenia and neurodevelopmental disorders.

This is a preview of subscription content, access via your institution

Access options

Buy this article

Prices may be subject to local taxes which are calculated during checkout

Fig. 1: Cognitive deficits observed in GIT1-NKO mice tested in a battery of behavioral paradigms.
Fig. 2: Global functional and synaptic analyses of mouse hippocampal proteome and phospho-proteome.
Fig. 3: Enrichment of synaptic proteins in the set of hippocampal phospho-proteins regulated by GIT1.
Fig. 4: GIT1-regulated synaptic phosphorylation signaling events within a network of neuropsychiatric risk genes.

Similar content being viewed by others

References

  1. Vissers LE, Gilissen C, Veltman JA. Genetic studies in intellectual disability and related disorders. Nat Rev Genet. 2016;17:9–18.

    Article  CAS  PubMed  Google Scholar 

  2. McGrath J, Saha S, Chant D, Welham J. Schizophrenia: a concise overview of incidence, prevalence, and mortality. Epidemiol Rev. 2008;30:67–76.

    Article  PubMed  Google Scholar 

  3. Kahn RS, Keefe RS. Schizophrenia is a cognitive illness: time for a change in focus. JAMA Psychiatry. 2013;70:1107–12.

    Article  PubMed  Google Scholar 

  4. Tandon R, Gaebel W, Barch DM, Bustillo J, Gur RE, Heckers S, et al. Definition and description of schizophrenia in the DSM-5. Schizophr Res. 2013;150:3–10.

    Article  PubMed  Google Scholar 

  5. Schizophrenia Working Group of the Psychiatric Genomics Consortium. Biological insights from 108 schizophrenia-associated genetic loci. Nature. 2014;511:421-7.

  6. Fromer M, Pocklington AJ, Kavanagh DH, Williams HJ, Dwyer S, Gormley P, et al. De novo mutations in schizophrenia implicate synaptic networks. Nature. 2014;506:179–84.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Purcell SM, Moran JL, Fromer M, Ruderfer D, Solovieff N, Roussos P, et al. A polygenic burden of rare disruptive mutations in schizophrenia. Nature. 2014;506:185–90.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Genovese G, Fromer M, Stahl EA, Ruderfer DM, Chambert K, Landen M, et al. Increased burden of ultra-rare protein-altering variants among 4,877 individuals with schizophrenia. Nat Neurosci. 2016;19:1433–41.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Pardinas AF, Holmans P, Pocklington AJ, Escott-Price V, Ripke S, Carrera N, et al. Common schizophrenia alleles are enriched in mutation-intolerant genes and in regions under strong background selection. Nat Genet. 2018;50:381–9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Cristino AS, Williams SM, Hawi Z, An JY, Bellgrove MA, Schwartz CE, et al. Neurodevelopmental and neuropsychiatric disorders represent an interconnected molecular system. Mol Psychiatry. 2014;19:294–301.

    Article  CAS  PubMed  Google Scholar 

  11. Hormozdiari F, Penn O, Borenstein E, Eichler EE. The discovery of integrated gene networks for autism and related disorders. Genome Res. 2015;25:142–54.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Broek JAC, Lin Z, de Gruiter HM, van ‘t Spijker H, Haasdijk ED, Cox D, et al. Synaptic vesicle dynamic changes in a model of fragile X. Mol Autism. 2016;7:17.

    Article  PubMed  PubMed Central  Google Scholar 

  13. Pechstein A, Shupliakov O, Haucke V. Intersectin 1: a versatile actor in the synaptic vesicle cycle. Biochem Soc Trans. 2010;38:181–6.

    Article  CAS  PubMed  Google Scholar 

  14. Egbujo CN, Sinclair D, Hahn CG. Dysregulations of Synaptic Vesicle Trafficking in Schizophrenia. Curr Psychiatry Rep. 2016;18:77.

    Article  PubMed  PubMed Central  Google Scholar 

  15. Volk L, Chiu SL, Sharma K, Huganir RL. Glutamate synapses in human cognitive disorders. Annu Rev Neurosci. 2015;38:127–49.

    Article  CAS  PubMed  Google Scholar 

  16. Hall J, Trent S, Thomas KL, O’Donovan MC, Owen MJ. Genetic risk for schizophrenia: convergence on synaptic pathways involved in plasticity. Biol psychiatry. 2015;77:52–8.

    Article  CAS  PubMed  Google Scholar 

  17. Zhou W, Li X, Premont RT. Expanding functions of GIT Arf GTPase-activating proteins, PIX Rho guanine nucleotide exchange factors and GIT-PIX complexes. J Cell Sci. 2016;129:1963–74.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Kim MJ, Biag J, Fass DM, Lewis MC, Zhang Q, Fleishman M, et al. Functional analysis of rare variants found in schizophrenia implicates a critical role for GIT1-PAK3 signaling in neuroplasticity. Mol Psychiatry. 2017;22:417–29.

    Article  CAS  PubMed  Google Scholar 

  19. Datta D, Arion D, Corradi JP, Lewis DA. Altered expression of CDC42 signaling pathway components in cortical layer 3 pyramidal cells in schizophrenia. Biol psychiatry. 2015;78:775–85.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Uddin M, Pellecchia G, Thiruvahindrapuram B, D’Abate L, Merico D, Chan A, et al. Indexing effects of copy number variation on genes involved in developmental delay. Sci Rep. 2016;6:28663.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Lek M, Karczewski KJ, Minikel EV, Samocha KE, Banks E, Fennell T, et al. Analysis of protein-coding genetic variation in 60,706 humans. Nature. 2016;536:285–91.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Lim J, Ritt DA, Zhou M, Morrison DK. The CNK2 scaffold interacts with vilse and modulates Rac cycling during spine morphogenesis in hippocampal neurons. Curr Biol. 2014;24:786–92.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Houge G, Rasmussen IH, Hovland R. Loss-of-function CNKSR2 mutation is a likely cause of non-syndromic X-linked intellectual disability. Mol Syndromol. 2012;2:60–3.

    Article  CAS  PubMed  Google Scholar 

  24. Ko J, Kim S, Valtschanoff JG, Shin H, Lee JR, Sheng M, et al. Interaction between liprin-alpha and GIT1 is required for AMPA receptor targeting. J Neurosci: Off J Soc Neurosci. 2003;23:1667–77.

    Article  CAS  Google Scholar 

  25. Kutsche K, Yntema H, Brandt A, Jantke I, Nothwang HG, Orth U, et al. Mutations in ARHGEF6, encoding a guanine nucleotide exchange factor for Rho GTPases, in patients with X-linked mental retardation. Nat Genet. 2000;26:247–50.

    Article  CAS  PubMed  Google Scholar 

  26. Bagrodia S, Bailey D, Lenard Z, Hart M, Guan JL, Premont RT, et al. A tyrosine-phosphorylated protein that binds to an important regulatory region on the cool family of p21-activated kinase-binding proteins. J Biol Chem. 1999;274:22393–400.

    Article  CAS  PubMed  Google Scholar 

  27. Allen KM, Gleeson JG, Bagrodia S, Partington MW, MacMillan JC, Cerione RA, et al. PAK3 mutation in nonsyndromic X-linked mental retardation. Nat Genet. 1998;20:25–30.

    Article  CAS  PubMed  Google Scholar 

  28. Zhang H, Webb DJ, Asmussen H, Horwitz AF. Synapse formation is regulated by the signaling adaptor GIT1. J Cell Biol. 2003;161:131–42.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Zhang H, Webb DJ, Asmussen H, Niu S, Horwitz AF. A GIT1/PIX/Rac/PAK signaling module regulates spine morphogenesis and synapse formation through MLC. J Neurosci: Off J Soc Neurosci. 2005;25:3379–88.

    Article  CAS  Google Scholar 

  30. Collins MO, Husi H, Yu L, Brandon JM, Anderson CN, Blackstock WP, et al. Molecular characterization and comparison of the components and multiprotein complexes in the postsynaptic proteome. J Neurochem. 2006;97:16–23.

    Article  CAS  PubMed  Google Scholar 

  31. Kim S, Ko J, Shin H, Lee JR, Lim C, Han JH, et al. The GIT family of proteins forms multimers and associates with the presynaptic cytomatrix protein Piccolo. J Biol Chem. 2003;278:6291–300.

    Article  CAS  PubMed  Google Scholar 

  32. Montesinos MS, Dong W, Goff K, Das B, Guerrero-Given D, Schmalzigaug R, et al. Presynaptic deletion of GIT proteins results in increased synaptic strength at a mammalian central synapse. Neuron. 2015;88:918–25.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Schmalzigaug R, Rodriguiz RM, Bonner PE, Davidson CE, Wetsel WC, Premont RT. Impaired fear response in mice lacking GIT1. Neurosci Lett. 2009;458:79–83.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Won H, Mah W, Kim E, Kim JW, Hahm EK, Kim MH, et al. GIT1 is associated with ADHD in humans and ADHD-like behaviors in mice. Nat Med. 2011;17:566–72.

    Article  CAS  PubMed  Google Scholar 

  35. Martyn AC, Toth K, Schmalzigaug R, Hedrick NG, Rodriguiz RM, Yasuda R, et al. GIT1 regulates synaptic structural plasticity underlying learning. PloS One. 2018;13:e0194350.

    Article  PubMed  PubMed Central  Google Scholar 

  36. Menon P, Deane R, Sagare A, Lane SM, Zarcone TJ, O’Dell MR, et al. Impaired spine formation and learning in GPCR kinase 2 interacting protein-1 (GIT1) knockout mice. Brain Res. 2010;1317:218–26.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Pang J, Hoefen R, Pryhuber GS, Wang J, Yin G, White RJ, et al. G-protein-coupled receptor kinase interacting protein-1 is required for pulmonary vascular development. Circulation. 2009;119:1524–32.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Bates B, Rios M, Trumpp A, Chen C, Fan G, Bishop JM, et al. Neurotrophin-3 is required for proper cerebellar development. Nat Neurosci. 1999;2:115–7.

    Article  CAS  PubMed  Google Scholar 

  39. Dell RB, Holleran S, Ramakrishnan R. Sample size determination. ILAR J. 2002:43:207–13. https://doi.org/10.1093/ilar.43.4.207. Erratum in: ILAR J. 2003;44:239.

  40. Wolf A, Bauer B, Abner EL, Ashkenazy-Frolinger T, Hartz AM. A comprehensive behavioral test battery to assess learning and memory in 129S6/Tg2576 Mice. PloS One. 2016;11:e0147733.

    Article  PubMed  PubMed Central  Google Scholar 

  41. Clark RE, Zola SM, Squire LR. Impaired recognition memory in rats after damage to the hippocampus. J Neurosci: Off J Soc Neurosci. 2000;20:8853–60.

    Article  CAS  Google Scholar 

  42. Lotfipour S, Byun JS, Leach P, Fowler CD, Murphy NP, Kenny PJ. et al. Targeted deletion of the mouse alpha2 nicotinic acetylcholine receptor subunit gene (Chrna2) potentiates nicotine-modulated behaviors. J Neurosci: Off J Soc Neurosci. 2013;33:7728–41.

    Article  CAS  Google Scholar 

  43. Hong ST, Mah W. A critical role of GIT1 in vertebrate and invertebrate brain development. Exp Neurobiol. 2015;24:8–16.

    Article  PubMed  PubMed Central  Google Scholar 

  44. Mertins P, Mani DR, Ruggles KV, Gillette MA, Clauser KR, Wang P, et al. Proteogenomics connects somatic mutations to signalling in breast cancer. Nature. 2016;534:55–62.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Mundt F, Rajput S, Li S, Ruggles KV, Mooradian AD, Mertins P, et al. Mass spectrometry-based proteomics reveals potential roles of NEK9 and MAP2K4 in resistance to PI3K inhibitors in triple negative breast cancers. Cancer Res. 2018:78:2732–46.

  46. Elias JE, Gygi SP. Target-decoy search strategy for mass spectrometry-based proteomics. Methods Mol Biol. 2010;604:55–71.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. Nesvizhskii AI, Aebersold R. Interpretation of shotgun proteomic data: the protein inference problem. Mol Cell Proteom. 2005;4:1419–40.

    Article  CAS  Google Scholar 

  48. Maere S, Heymans K, Kuiper M. BiNGO: a Cytoscape plugin to assess overrepresentation of gene ontology categories in biological networks. Bioinformatics. 2005;21:3448–9.

    Article  CAS  PubMed  Google Scholar 

  49. Cheng C, Fass DM, Folz-Donahue K, MacDonald ME, Haggarty SJ. Highly expandable human iPS cell-derived neural progenitor cells (npc) and neurons for central nervous system disease modeling and high-throughput screening. Curr Protoc Hum Genet. 2017;92:21.8.1–8.

    CAS  PubMed  Google Scholar 

  50. Roux KJ, Kim DI, Burke B, May DG. BioID: a screen for protein-protein interactions. Curr Protoc protein Sci. 2018;91:19.23.1–15.

    CAS  PubMed  Google Scholar 

  51. Pang J, Yan C, Natarajan K, Cavet ME, Massett MP, Yin G, et al. GIT1 mediates HDAC5 activation by angiotensin II in vascular smooth muscle cells. Arterioscler Thromb Vasc Biol. 2008;28:892–8.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  52. Kim JJ, Fanselow MS. Modality-specific retrograde amnesia of fear. Science. 1992;256:675–7.

    Article  CAS  PubMed  Google Scholar 

  53. Muller J, Corodimas KP, Fridel Z, LeDoux JE. Functional inactivation of the lateral and basal nuclei of the amygdala by muscimol infusion prevents fear conditioning to an explicit conditioned stimulus and to contextual stimuli. Behav Neurosci. 1997;111:683–91.

    Article  CAS  PubMed  Google Scholar 

  54. Lalonde R. The neurobiological basis of spontaneous alternation. Neurosci Biobehav Rev. 2002;26:91–104.

    Article  CAS  PubMed  Google Scholar 

  55. Dudchenko PA. An overview of the tasks used to test working memory in rodents. Neurosci Biobehav Rev. 2004;28:699–709.

    Article  PubMed  Google Scholar 

  56. Silverman JL, Yang M, Lord C, Crawley JN. Behavioural phenotyping assays for mouse models of autism. Nat Rev Neurosci. 2010;11:490–502.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  57. Braff D, Stone C, Callaway E, Geyer M, Glick I, Bali L. Prestimulus effects on human startle reflex in normals and schizophrenics. Psychophysiol. 1978;15:339–43.

    Article  CAS  Google Scholar 

  58. Jordi E, Heiman M, Marion-Poll L, Guermonprez P, Cheng SK, Nairn AC, et al. Differential effects of cocaine on histone posttranslational modifications in identified populations of striatal neurons. Proc Natl Acad Sci USA. 2013;110:9511–6.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  59. Vizi ES, Kiss JP. Neurochemistry and pharmacology of the major hippocampal transmitter systems: synaptic and nonsynaptic interactions. Hippocampus. 1998;8:566–607.

    Article  CAS  PubMed  Google Scholar 

  60. Gonzalez-Burgos I, Feria-Velasco A. Serotonin/dopamine interaction in memory formation. Prog Brain Res. 2008;172:603–23.

    Article  CAS  PubMed  Google Scholar 

  61. Harrison PJ. The hippocampus in schizophrenia: a review of the neuropathological evidence and its pathophysiological implications. Psychopharmacol (Berl). 2004;174:151–62.

    Article  CAS  Google Scholar 

  62. Woolfrey KM, Dell’Acqua ML. Coordination of protein phosphorylation and dephosphorylation in synaptic plasticity. J Biol Chem. 2015;290:28604–12.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  63. Boyken J, Gronborg M, Riedel D, Urlaub H, Jahn R, Chua JJ. Molecular profiling of synaptic vesicle docking sites reveals novel proteins but few differences between glutamatergic and GABAergic synapses. Neuron. 2013;78:285–97.

    Article  CAS  PubMed  Google Scholar 

  64. Zhao ZS, Manser E, Loo TH, Lim L. Coupling of PAK-interacting exchange factor PIX to GIT1 promotes focal complex disassembly. Mol Cell Biol. 2000;20:6354–63.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  65. Hsu RM, Tsai MH, Hsieh YJ, Lyu PC, Yu JS. Identification of MYO18A as a novel interacting partner of the PAK2/betaPIX/GIT1 complex and its potential function in modulating epithelial cell migration. Mol Biol Cell. 2010;21:287–301.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  66. Shin H, Wyszynski M, Huh KH, Valtschanoff JG, Lee JR, Ko J, et al. Association of the kinesin motor KIF1A with the multimodular protein liprin-alpha. J Biol Chem. 2003;278:11393–401.

    Article  CAS  PubMed  Google Scholar 

  67. Frese S, Schubert WD, Findeis AC, Marquardt T, Roske YS, Stradal TE, et al. The phosphotyrosine peptide binding specificity of Nck1 and Nck2 Src homology 2 domains. J Biol Chem. 2006;281:18236–45.

    Article  CAS  PubMed  Google Scholar 

  68. Ko J, Na M, Kim S, Lee JR, Kim E. Interaction of the ERC family of RIM-binding proteins with the liprin-alpha family of multidomain proteins. J Biol Chem. 2003;278:42377–85.

    Article  CAS  PubMed  Google Scholar 

  69. Park E, Na M, Choi J, Kim S, Lee JR, Yoon J, et al. The Shank family of postsynaptic density proteins interacts with and promotes synaptic accumulation of the beta PIX guanine nucleotide exchange factor for Rac1 and Cdc42. J Biol Chem. 2003;278:19220–9.

    Article  CAS  PubMed  Google Scholar 

  70. Martin HG, Henley JM, Meyer G. Novel putative targets of N-ethylmaleimide sensitive fusion protein (NSF) and alpha/beta soluble NSF attachment proteins (SNAPs) include the Pak-binding nucleotide exchange factor betaPIX. J Cell Biochem. 2006;99:1203–15.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  71. Fu CA, Shen M, Huang BC, Lasaga J, Payan DG, Luo Y. TNIK, a novel member of the germinal center kinase family that activates the c-Jun N-terminal kinase pathway and regulates the cytoskeleton. J Biol Chem. 1999;274:30729–37.

    Article  CAS  PubMed  Google Scholar 

  72. Wang Q, Amato SP, Rubitski DM, Hayward MM, Kormos BL, Verhoest PR, et al. Identification of phosphorylation consensus sequences and endogenous neuronal substrates of the psychiatric risk kinase TNIK. J Pharm Exp Ther. 2016;356:410–23.

    Article  CAS  Google Scholar 

  73. Yin G, Haendeler J, Yan C, Berk BC. GIT1 functions as a scaffold for MEK1-extracellular signal-regulated kinase 1 and 2 activation by angiotensin II and epidermal growth factor. Mol Cell Biol. 2004;24:875–85.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  74. Yin G, Zheng Q, Yan C, Berk BC. GIT1 is a scaffold for ERK1/2 activation in focal adhesions. J Biol Chem. 2005;280:27705–12.

    Article  CAS  PubMed  Google Scholar 

  75. Zhang N, Cai W, Yin G, Nagel DJ, Berk BC. GIT1 is a novel MEK1-ERK1/2 scaffold that localizes to focal adhesions. Cell Biol Int. 2009;34:41–7.

    PubMed  PubMed Central  Google Scholar 

  76. Versele M, Thorner J. Septin collar formation in budding yeast requires GTP binding and direct phosphorylation by the PAK, Cla4. J Cell Biol. 2004;164:701–15.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  77. Wittmann T, Bokoch GM, Waterman-Storer CM. Regulation of microtubule destabilizing activity of Op18/stathmin downstream of Rac1. J Biol Chem. 2004;279:6196–203.

    Article  CAS  PubMed  Google Scholar 

  78. Aslan JE, Baker SM, Loren CP, Haley KM, Itakura A, Pang J, et al. The PAK system links Rho GTPase signaling to thrombin-mediated platelet activation. Am J Physiol Cell Physiol. 2013;305:C519–28.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  79. Shin EY, Shim ES, Lee CS, Kim HK, Kim EG. Phosphorylation of RhoGDI1 by p21-activated kinase 2 mediates basic fibroblast growth factor-stimulated neurite outgrowth in PC12 cells. Biochem Biophys Res Commun. 2009;379:384–9.

    Article  CAS  PubMed  Google Scholar 

  80. Vadlamudi RK, Manavathi B, Singh RR, Nguyen D, Li F, Kumar R. An essential role of Pak1 phosphorylation of SHARP in Notch signaling. Oncogene. 2005;24:4591–6.

    Article  CAS  PubMed  Google Scholar 

  81. Rennefahrt UE, Deacon SW, Parker SA, Devarajan K, Beeser A, Chernoff J, et al. Specificity profiling of Pak kinases allows identification of novel phosphorylation sites. J Biol Chem. 2007;282:15667–78.

    Article  CAS  PubMed  Google Scholar 

  82. Veeranna, Amin ND, Ahn NG, Jaffe H, Winters CA, Grant P, et al. Mitogen-activated protein kinases (Erk1,2) phosphorylate Lys-Ser-Pro (KSP) repeats in neurofilament proteins NF-H and NF-M. J Neurosci: Off J Soc Neurosci. 1998;18:4008–21.

    Article  CAS  Google Scholar 

  83. Kubiniok P, Finicle BT, Piffaretti F, McCracken AN, Perryman M, Hanessian S, et al. Dynamic phosphoproteomics uncovers signaling pathways modulated by anti-oncogenic sphingolipid analogs. Mol Cell Proteomics. 2019;18:408–22. https://doi.org/10.1074/mcp.RA118.001053.

    Article  CAS  PubMed  Google Scholar 

  84. Westphal RS, Coffee RL Jr, Marotta A, Pelech SL, Wadzinski BE. Identification of kinase-phosphatase signaling modules composed of p70 S6 kinase-protein phosphatase 2A (PP2A) and p21-activated kinase-PP2A. J Biol Chem. 1999;274:687–92. https://doi.org/10.1074/jbc.274.2.687.

  85. Reynhout S, Jansen S, Haesen D, van Belle S, de Munnik SA, Bongers EMHF, et al. De novo mutations affecting the catalytic Cα subunit of PP2A, PPP2CA, cause syndromic intellectual disability resembling other PP2A-related neurodevelopmental disorders. Am J Hum Genet. 2019;104:139–56. https://doi.org/10.1016/j.ajhg.2018.12.002. Erratum in: Am J Hum Genet. 2019;104:357.

    Article  CAS  PubMed  Google Scholar 

  86. Edwards RH. The neurotransmitter cycle and quantal size. Neuron. 2007;55:835–58.

    Article  CAS  PubMed  Google Scholar 

  87. Xue J, Tsang CW, Gai WP, Malladi CS, Trimble WS, Rostas JA, et al. Septin 3 (G-septin) is a developmentally regulated phosphoprotein enriched in presynaptic nerve terminals. J Neurochem. 2004;91:579–90.

    Article  CAS  PubMed  Google Scholar 

  88. Jaudon F, Raynaud F, Wehrle R, Bellanger JM, Doulazmi M, Vodjdani G, et al. The RhoGEF DOCK10 is essential for dendritic spine morphogenesis. Mol Biol Cell. 2015;26:2112–27.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  89. Ka M, Kim WY. Microtubule-actin crosslinking factor 1 is required for dendritic arborization and axon outgrowth in the developing brain. Mol Neurobiol. 2016;53:6018–32.

    Article  CAS  PubMed  Google Scholar 

  90. Doherty JL, O’Donovan MC, Owen MJ. Recent genomic advances in schizophrenia. Clin Genet. 2012;81:103–9.

    Article  CAS  PubMed  Google Scholar 

  91. Yuan H, Low CM, Moody OA, Jenkins A, Traynelis SF. Ionotropic GABA and glutamate receptor mutations and human neurologic diseases. Mol Pharmacol. 2015;88:203–17. https://doi.org/10.1124/mol.115.097998.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  92. Kim MJ, Biag J, Fass DM, Lewis MC, Zhang Q, Fleishman M, et al. Functional analysis of rare variants found in schizophrenia implicates a critical role for GIT1-PAK3 signaling in neuroplasticity. Mol Psychiatry. 2017;22:417–29. https://doi.org/10.1038/mp.2016.98.

    Article  CAS  PubMed  Google Scholar 

  93. Ko J, Kim S, Valtschanoff JG, Shin H, Lee JR, Sheng M, et al. Interaction between liprin-alpha and GIT1 is required for AMPA receptor targeting. J Neurosci. 2003;23:1667–77. https://doi.org/10.1523/JNEUROSCI.23-05-01667.2003.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  94. Nabavi S, Fox R, Proulx CD, Lin JY, Tsien RY, Malinow R. Engineering a memory with LTD and LTP. Nature 2014;511:348–52.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  95. Ramakers GJ, Wolfer D, Rosenberger G, Kuchenbecker K, Kreienkamp HJ, Prange-Kiel J, et al. Dysregulation of Rho GTPases in the alphaPix/Arhgef6 mouse model of X-linked intellectual disability is paralleled by impaired structural and synaptic plasticity and cognitive deficits. Hum Mol Genet. 2012;21:268–86.

    Article  CAS  PubMed  Google Scholar 

  96. Asrar S, Meng Y, Zhou Z, Todorovski Z, Huang WW, Jia Z. Regulation of hippocampal long-term potentiation by p21-activated protein kinase 1 (PAK1). Neuropharmacology. 2009;56:73–80.

    Article  CAS  PubMed  Google Scholar 

  97. Castillo PE, Schoch S, Schmitz F, Sudhof TC, Malenka RC. RIM1alpha is required for presynaptic long-term potentiation. Nature. 2002;415:327–30.

    Article  CAS  PubMed  Google Scholar 

  98. Diering GH, Gustina AS, Huganir RL. PKA-GluA1 coupling via AKAP5 controls AMPA receptor phosphorylation and cell-surface targeting during bidirectional homeostatic plasticity. Neuron. 2014;84:790–805.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  99. Sanderson JL, Gorski JA, Dell’Acqua ML. NMDA receptor-dependent LTD requires transient synaptic incorporation of Ca(2)(+)-permeable AMPARs mediated by AKAP150-anchored PKA and calcineurin. Neuron. 2016;89:1000–15.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  100. Orock A, Logan S, Deak F. Munc18-1 haploinsufficiency impairs learning and memory by reduced synaptic vesicular release in a model of Ohtahara syndrome. Mol Cell Neurosci. 2018;88:33–42.

    Article  CAS  PubMed  Google Scholar 

  101. Takei Y, Kikkawa YS, Atapour N, Hensch TK, Hirokawa N. Defects in Synaptic plasticity, reduced nmda-receptor transport, and instability of postsynaptic density proteins in micE lacking microtubule-associated protein 1A. J Neurosci: Off J Soc Neurosci. 2015;35:15539–54.

    Article  CAS  Google Scholar 

  102. Wu D, Bacaj T, Morishita W, Goswami D, Arendt KL, Xu W, et al. Postsynaptic synaptotagmins mediate AMPA receptor exocytosis during LTP. Nature. 2017;544:316–21.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  103. Shumyatsky GP, Malleret G, Shin RM, Takizawa S, Tully K, Tsvetkov E, et al. stathmin, a gene enriched in the amygdala, controls both learned and innate fear. Cell. 2005;123:697–709.

    Article  CAS  PubMed  Google Scholar 

  104. Spilker C, Nullmeier S, Grochowska KM, Schumacher A, Butnaru I, Macharadze T, et al. A Jacob/Nsmf Gene Knockout Results in Hippocampal Dysplasia and Impaired BDNF Signaling in Dendritogenesis. PLoS Genet. 2016;12:e1005907.

    Article  PubMed  PubMed Central  Google Scholar 

  105. Lisman J, Yasuda R, Raghavachari S. Mechanisms of CaMKII action in long-term potentiation. Nat Rev Neurosci. 2012;13:169–82.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  106. Hilal ML, Moreau MM, Racca C, Pinheiro VL, Piguel NH, Santoni MJ, et al. Activity-Dependent Neuroplasticity Induced by an Enriched Environment Reverses Cognitive Deficits in Scribble Deficient Mouse. Cereb Cortex (N. Y, NY: 1991). 2017;27:5635–51.

    Google Scholar 

  107. Kim IH, Racz B, Wang H, Burianek L, Weinberg R, Yasuda R, et al. Disruption of Arp2/3 results in asymmetric structural plasticity of dendritic spines and progressive synaptic and behavioral abnormalities. The. J Neurosci: Off J Soc Neurosci. 2013;33:6081–92.

    Article  CAS  Google Scholar 

  108. Beaudoin GM 3rd, Schofield CM, Nuwal T, Zang K, Ullian EM, Huang B, et al. Afadin, a Ras/Rap effector that controls cadherin function, promotes spine and excitatory synapse density in the hippocampus. J Neurosci: Off J Soc Neurosci. 2012;32:99–110.

    Article  CAS  Google Scholar 

  109. Endele S, Rosenberger G, Geider K, Popp B, Tamer C, Stefanova I, et al. Mutations in GRIN2A and GRIN2B encoding regulatory subunits of NMDA receptors cause variable neurodevelopmental phenotypes. Nat Genet. 2010;42:1021–6.

    Article  CAS  PubMed  Google Scholar 

  110. Sanders SJ, He X, Willsey AJ, Ercan-Sencicek AG, Samocha KE, Cicek AE, et al. Insights into autism spectrum disorder genomic architecture and biology from 71 risk Loci. Neuron. 2015;87:1215–33.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  111. Sanders SJ, He X, Willsey AJ, Ercan-Sencicek AG, Samocha KE, Cicek AE, et al. Insights into autism spectrum disorder genomic architecture and biology from 71 risk loci. Neuron. 2015;87:1215–33. https://doi.org/10.1016/j.neuron.2015.09.016.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  112. Prevalence and architecture of de novo mutations in developmental disorders. Nature. 2017;542:433–8.

  113. Kim K, Lakhanpal G, Lu HE, Khan M, Suzuki A, Hayashi MK, et al. A temporary gating of actin remodeling during synaptic plasticity consists of the interplay between the kinase and structural functions of CaMKII. Neuron. 2015;87:813–26.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  114. Balu D, Larson JR, Schmidt JV, Wirtshafter D, Yevtodiyenko A, Leonard JP. Behavioral and physiological characterization of PKC-dependent phosphorylation in the Grin2aPKC mouse. Brain Res. 2016;1646:315–26.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  115. Aleman A, Kahn RS. Strange feelings: do amygdala abnormalities dysregulate the emotional brain in schizophrenia? Prog Neurobiol. 2005;77:283–98.

    PubMed  Google Scholar 

  116. Holt DJ, Lebron-Milad K, Milad MR, Rauch SL, Pitman RK, Orr SP, et al. Extinction memory is impaired in schizophrenia. Biol Psychiatry. 2009;65:455–63.

    Article  PubMed  Google Scholar 

  117. Pinkham AE, Liu P, Lu H, Kriegsman M, Simpson C, Tamminga C. Amygdala hyperactivity at rest in paranoid individuals with schizophrenia. Am J Psychiatry. 2015;172:784–92.

    Article  PubMed  Google Scholar 

  118. Potvin S, Tikasz A, Mendrek A. Emotionally neutral stimuli are not neutral in schizophrenia: a mini review of functional neuroimaging studies. Front Psychiatry. 2016;7:115.

    Article  PubMed  PubMed Central  Google Scholar 

  119. Hamdan FF, Srour M, Capo-Chichi JM, Daoud H, Nassif C, Patry L, et al. De novo mutations in moderate or severe intellectual disability. PLoS Genet. 2014;10:e1004772. https://doi.org/10.1371/journal.pgen.1004772.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  120. Meng J, Meng Y, Hanna A, Janus C, Jia Z. Abnormal long-lasting synaptic plasticity and cognition in mice lacking the mental retardation gene Pak3. J Neurosci: Off J Soc Neurosci. 2005;25:6641–50.

    Article  CAS  Google Scholar 

  121. Waites CL, Leal-Ortiz SA, Andlauer TF, Sigrist SJ, Garner CC. Piccolo regulates the dynamic assembly of presynaptic F-actin. J Neurosci: Off J Soc Neurosci. 2011;31:14250–63.

    Article  CAS  Google Scholar 

  122. Kohansal-Nodehi M, Chua JJ, Urlaub H, Jahn R, Czernik D Analysis of protein phosphorylation in nerve terminal reveals extensive changes in active zone proteins upon exocytosis. Elife. 2016;5:e14530.

  123. Xie Z, Photowala H, Cahill ME, Srivastava DP, Woolfrey KM, Shum CY, et al. Coordination of synaptic adhesion with dendritic spine remodeling by AF-6 and kalirin-7. J Neurosci. 2008;28:6079–91. https://doi.org/10.1523/JNEUROSCI.1170-08.2008.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

We thank members of the Stanley Center for Psychiatric Research and the Chemical Neurobiology Laboratory for helpful discussions and critical feedback. Dr. Guoping Feng is thanked for generously sharing GIT1 mice and for his critical feedback. This work was supported by funding from the Stanley Medical Research Institute, the National Institute of Mental Health (R01MH095088), and the Stuart & Suzanne Steele MGH Research Scholar Award (SJH).

Author information

Authors and Affiliations

Authors

Contributions

SJH, EMS, DMF, MCL, RA, MJK, RTP, and SAC designed the study. DMF, MCL, MJS, QZ, MF, DW, JB, RTP, and SJH conducted the experiments and analyzed data. DMF and SJH wrote the manuscript, and all authors edited the manuscript.

Corresponding author

Correspondence to Stephen J. Haggarty.

Ethics declarations

Competing interests

SJH is a member of the SAB and equity holder of Psy Therapeutics, Frequency Therapeutics, Sensorium Therapeutics, 4 M Therapeutics, and Proximity Therapeutics; and has received speaking/consulting fees from AstraZeneca, Amgen, Merck, and Syros, and RBNC. DMF has received consulting fees from Psy Therapeutics. None of these entities were involved in the current study.

Additional information

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

Supplementary information

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Fass, D.M., Lewis, M.C., Ahmad, R. et al. Brain-specific deletion of GIT1 impairs cognition and alters phosphorylation of synaptic protein networks implicated in schizophrenia susceptibility. Mol Psychiatry 27, 3272–3285 (2022). https://doi.org/10.1038/s41380-022-01557-z

Download citation

  • Received:

  • Revised:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41380-022-01557-z

Search

Quick links