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:

Decreased CNNM2 expression in prefrontal cortex affects sensorimotor gating function, cognition, dendritic spine morphogenesis and risk of schizophrenia

Abstract

Genome-wide association studies (GWASs) have reported multiple single nucleotide polymorphisms (SNPs) associated with schizophrenia, yet the underlying molecular mechanisms are largely unknown. In this study, we aimed to identify schizophrenia relevant genes showing alterations in mRNA and protein expression associated with risk SNPs at the 10q24.32-33 GWAS locus. We carried out the quantitative trait loci (QTL) and summary data-based Mendelian randomization (SMR) analyses, using the PsychENCODE dorsolateral prefrontal cortex (DLPFC) expression QTL (eQTL) database, as well as the ROSMAP and Banner DLPFC protein QTL (pQTL) datasets. The gene CNNM2 (encoding a magnesium transporter) at 10q24.32-33 was identified to be a robust schizophrenia risk gene, and was highly expressed in human neurons according to single cell RNA-seq (scRNA-seq) data. We further revealed that reduced Cnnm2 in the mPFC of mice led to impaired cognition and compromised sensorimotor gating function, and decreased Cnnm2 in primary cortical neurons altered dendritic spine morphogenesis, confirming the link between CNNM2 and endophenotypes of schizophrenia. Proteomics analyses showed that reduced Cnnm2 level changed expression of proteins associated with neuronal structure and function. Together, these results identify a robust gene in the pathogenesis of schizophrenia.

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: Genetic and molecular characterizations of 10q24.32-33 GWAS locus.
Fig. 2: SMR analyses through integrating schizophrenia PGC3 GWAS with different brain QTL datasets.
Fig. 3: Knockdown of Cnnm2 in the mPFC of mice results in behavioral abnormalities associated with schizophrenia.
Fig. 4: Knockdown of Cnnm2 expression in rat primary cortical neurons and their impacts on dendritic spine morphogenesis (n ≥ 70 neurons in each experimental group).
Fig. 5: overview of the altered proteome of mPFC in Cnnm2 knockdown mice.

Similar content being viewed by others

References

  1. Sullivan PF, Kendler KS, Neale MC. Schizophrenia as a complex trait: evidence from a meta-analysis of twin studies. Arch Gen Psychiatry. 2003;60:1187–92.

    Article  PubMed  Google Scholar 

  2. Trubetskoy V, Pardinas AF, Qi T, Panagiotaropoulou G, Awasthi S, Bigdeli TB, et al. Mapping genomic loci implicates genes and synaptic biology in schizophrenia. Nature. 2022;604:502–8.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. Edwards SL, Beesley J, French JD, Dunning AM. Beyond GWASs: illuminating the dark road from association to function. Am J Hum Genet. 2013;93:779–97.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. Gusev A, Mancuso N, Won H, Kousi M, Finucane HK, Reshef Y, et al. Transcriptome-wide association study of schizophrenia and chromatin activity yields mechanistic disease insights. Nat Genet. 2018;50:538–48.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Huckins LM, Dobbyn A, Ruderfer DM, Hoffman G, Wang W, Pardinas AF, et al. Gene expression imputation across multiple brain regions provides insights into schizophrenia risk. Nat Genet. 2019;51:659–74.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Fromer M, Roussos P, Sieberts SK, Johnson JS, Kavanagh DH, Perumal TM, et al. Gene expression elucidates functional impact of polygenic risk for schizophrenia. Nat Neurosci. 2016;19:1442–53.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Jaffe AE, Straub RE, Shin JH, Tao R, Gao Y, Collado-Torres L, et al. Developmental and genetic regulation of the human cortex transcriptome illuminate schizophrenia pathogenesis. Nat Neurosci. 2018;21:1117–25.

    Article  CAS  PubMed Central  Google Scholar 

  8. Schizophrenia Psychiatric Genome-Wide Association Study Consortium. Genome-wide association study identifies five new schizophrenia loci. Nat Genet. 2011;43:969–76.

    Article  Google Scholar 

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

    Article  PubMed Central  Google Scholar 

  10. Wynn JK, Dawson ME, Schell AM, McGee M, Salveson D, Green MF. Prepulse facilitation and prepulse inhibition in schizophrenia patients and their unaffected siblings. Biol Psychiatry. 2004;55:518–23.

    Article  PubMed  Google Scholar 

  11. San-Martin R, Castro LA, Menezes PR, Fraga FJ, Simoes PW, Salum C. Meta-analysis of sensorimotor gating deficits in patients with schizophrenia evaluated by prepulse inhibition test. Schizophr Bull. 2020;46:1482–97.

    Article  PubMed  PubMed Central  Google Scholar 

  12. Swerdlow NR, Weber M, Qu Y, Light GA, Braff DL. Realistic expectations of prepulse inhibition in translational models for schizophrenia research. Psychopharmacology (Berl). 2008;199:331–88.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Huang Y, Huang J, Zhou QX, Yang CX, Yang CP, Mei WY, et al. ZFP804A mutant mice display sex-dependent schizophrenia-like behaviors. Mol Psychiatry. 2021;26:2514–32.

    Article  CAS  PubMed  Google Scholar 

  14. Miro X, Meier S, Dreisow ML, Frank J, Strohmaier J, Breuer R, et al. Studies in humans and mice implicate neurocan in the etiology of mania. Am J Psychiatry. 2012;169:982–90.

    Article  PubMed  Google Scholar 

  15. Carr GV, Chen J, Yang F, Ren M, Yuan P, Tian Q, et al. KCNH2-3.1 expression impairs cognition and alters neuronal function in a model of molecular pathology associated with schizophrenia. Mol Psychiatry. 2016;21:1517–26.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Weinberger DR. Implications of normal brain development for the pathogenesis of schizophrenia. Arch Gen Psychiatry. 1987;44:660–9.

    Article  CAS  PubMed  Google Scholar 

  17. Glantz LA, Lewis DA. Decreased dendritic spine density on prefrontal cortical pyramidal neurons in schizophrenia. Arch Gen Psychiatry. 2000;57:65–73.

    Article  CAS  PubMed  Google Scholar 

  18. Osimo EF, Beck K, Reis Marques T, Howes OD. Synaptic loss in schizophrenia: a meta-analysis and systematic review of synaptic protein and mRNA measures. Mol Psychiatry. 2019;24:549–61.

    Article  CAS  PubMed  Google Scholar 

  19. Berdenis van Berlekom A, Muflihah CH, Snijders G, MacGillavry HD, Middeldorp J, Hol EM, et al. Synapse pathology in schizophrenia: a meta-analysis of postsynaptic elements in postmortem brain studies. Schizophr Bull. 2020;46:374–86.

    PubMed  Google Scholar 

  20. MacDonald ML, Alhassan J, Newman JT, Richard M, Gu H, Kelly RM, et al. Selective loss of smaller spines in schizophrenia. Am J Psychiatry. 2017;174:586–94.

    Article  PubMed  PubMed Central  Google Scholar 

  21. Penzes P, Cahill ME, Jones KA, VanLeeuwen JE, Woolfrey KM. Dendritic spine pathology in neuropsychiatric disorders. Nat Neurosci. 2011;14:285–93.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Forrest MP, Parnell E, Penzes P. Dendritic structural plasticity and neuropsychiatric disease. Nat Rev Neurosci. 2018;19:215–34.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Glausier JR, Lewis DA. Dendritic spine pathology in schizophrenia. Neuroscience. 2013;251:90–107.

    Article  CAS  PubMed  Google Scholar 

  24. Smith KR, Kopeikina KJ, Fawcett-Patel JM, Leaderbrand K, Gao R, Schurmann B, et al. Psychiatric risk factor ANK3/ankyrin-G nanodomains regulate the structure and function of glutamatergic synapses. Neuron. 2014;84:399–415.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Hayashi-Takagi A, Takaki M, Graziane N, Seshadri S, Murdoch H, Dunlop AJ, et al. Disrupted-in-Schizophrenia 1 (DISC1) regulates spines of the glutamate synapse via Rac1. Nat Neurosci. 2010;13:327–32.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Deans PJM, Raval P, Sellers KJ, Gatford NJF, Halai S, Duarte RRR, et al. Psychosis risk candidate ZNF804A localizes to synapses and regulates neurite formation and dendritic spine structure. Biol Psychiatry. 2017;82:49–61.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Zhou D, Xiao X, Li M. The schizophrenia risk isoform ZNF804AE3E4 affects dendritic spine. Schizophr Res. 2020;218:324–5.

    Article  PubMed  Google Scholar 

  28. Wang D, Liu S, Warrell J, Won H, Shi X, Navarro FCP, et al. Comprehensive functional genomic resource and integrative model for the human brain. Science. 2018;362:eaat8464.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Ongen H, Buil A, Brown AA, Dermitzakis ET, Delaneau O. Fast and efficient QTL mapper for thousands of molecular phenotypes. Bioinformatics. 2016;32:1479–85.

    Article  CAS  PubMed  Google Scholar 

  30. Stegle O, Parts L, Durbin R, Winn J. A Bayesian framework to account for complex non-genetic factors in gene expression levels greatly increases power in eQTL studies. PLoS Comput Biol. 2010;6:e1000770.

    Article  PubMed  PubMed Central  Google Scholar 

  31. Purcell S, Neale B, Todd-Brown K, Thomas L, Ferreira MA, Bender D, et al. PLINK: a tool set for whole-genome association and population-based linkage analyses. Am J Hum Genet. 2007;81:559–75.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Wingo AP, Liu Y, Gerasimov ES, Gockley J, Logsdon BA, Duong DM, et al. Integrating human brain proteomes with genome-wide association data implicates new proteins in Alzheimer’s disease pathogenesis. Nat Genet. 2021;53:143–6.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Robins C, Liu Y, Fan W, Duong DM, Meigs J, Harerimana NV, et al. Genetic control of the human brain proteome. Am J Hum Genet. 2021;108:400–10.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Zhu Z, Zhang F, Hu H, Bakshi A, Robinson MR, Powell JE, et al. Integration of summary data from GWAS and eQTL studies predicts complex trait gene targets. Nat Genet. 2016;48:481–7.

    Article  CAS  PubMed  Google Scholar 

  35. GTEx Consortium. The Genotype-Tissue Expression (GTEx) project. Nat Genet. 2013;45:580–5.

    Article  Google Scholar 

  36. Lake BB, Ai R, Kaeser GE, Salathia NS, Yung YC, Liu R, et al. Neuronal subtypes and diversity revealed by single-nucleus RNA sequencing of the human brain. Science. 2016;352:1586–90.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Darmanis S, Sloan SA, Zhang Y, Enge M, Caneda C, Shuer LM, et al. A survey of human brain transcriptome diversity at the single cell level. Proc Natl Acad Sci USA. 2015;112:7285–90.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Lake BB, Chen S, Sos BC, Fan J, Kaeser GE, Yung YC, et al. Integrative single-cell analysis of transcriptional and epigenetic states in the human adult brain. Nature Biotechnol. 2018;36:70–80.

    Article  CAS  Google Scholar 

  39. Lei Y, Wang J, Wang D, Li C, Liu B, Fang X, et al. SIRT1 in forebrain excitatory neurons produces sexually dimorphic effects on depression-related behaviors and modulates neuronal excitability and synaptic transmission in the medial prefrontal cortex. Mol Psychiatry. 2020;25:1094–111.

    Article  CAS  PubMed  Google Scholar 

  40. Zhang Z, Ye M, Li Q, You Y, Yu H, Ma Y, et al. The schizophrenia susceptibility gene OPCML regulates spine maturation and cognitive behaviors through Eph-Cofilin signaling. Cell Rep. 2019;29:49–61.e47.

    Article  CAS  PubMed  Google Scholar 

  41. Cai X, Yang ZH, Li HJ, Xiao X, Li M, Chang H. A human-specific schizophrenia risk tandem repeat affects alternative splicing of a human-unique isoform AS3MTd2d3 and mushroom dendritic spine density. Schizophr Bull. 2021;41:219–27.

    Article  Google Scholar 

  42. Yang Z, Zhou D, Li H, Cai X, Liu W, Wang L, et al. The genome-wide risk alleles for psychiatric disorders at 3p21.1 show convergent effects on mRNA expression, cognitive function and mushroom dendritic spine. Mol Psychiatry. 2020;25:48–66.

    Article  CAS  PubMed  Google Scholar 

  43. Srivastava DP, Woolfrey KM, Penzes P. Analysis of dendritic spine morphology in cultured CNS neurons. J Vis Exp. 2011;53:e2794.

    Google Scholar 

  44. Rodriguez A, Ehlenberger DB, Dickstein DL, Hof PR, Wearne SL. Automated three-dimensional detection and shape classification of dendritic spines from fluorescence microscopy images. PLoS One. 2008;3:e1997.

    Article  PubMed  PubMed Central  Google Scholar 

  45. Conesa A, Gotz S, Garcia-Gomez JM, Terol J, Talon M, Robles M. Blast2GO: a universal tool for annotation, visualization and analysis in functional genomics research. Bioinformatics. 2005;21:3674–6.

    Article  CAS  PubMed  Google Scholar 

  46. O’Brien HE, Hannon E, Hill MJ, Toste CC, Robertson MJ, Morgan JE, et al. Expression quantitative trait loci in the developing human brain and their enrichment in neuropsychiatric disorders. Genome Biol. 2018;19:194.

    Article  PubMed  PubMed Central  Google Scholar 

  47. Jerber J, Seaton DD, Cuomo ASE, Kumasaka N, Haldane J, Steer J, et al. Population-scale single-cell RNA-seq profiling across dopaminergic neuron differentiation. Nat Genet. 2021;53:304–12.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. Bryois J, Calini D, Macnair W, Foo L, Urich E, Ortmann W, et al. Cell-type-specific cis-eQTLs in eight human brain cell types identify novel risk genes for psychiatric and neurological disorders. Nat Neurosci. 2022;25:1104–12.

    Article  CAS  PubMed  Google Scholar 

  49. Aygun N, Elwell AL, Liang D, Lafferty MJ, Cheek KE, Courtney KP, et al. Brain-trait-associated variants impact cell-type-specific gene regulation during neurogenesis. Am J Hum Genet. 2021;108:1647–68.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  50. Walker RL, Ramaswami G, Hartl C, Mancuso N, Gandal MJ, de la Torre-Ubieta L, et al. Genetic control of expression and splicing in developing human brain informs disease mechanisms. Cell. 2019;179:750–771.e722.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  51. Vogel C, Marcotte EM. Insights into the regulation of protein abundance from proteomic and transcriptomic analyses. Nat Rev Genet. 2012;13:227–32.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  52. Wingo TS, Liu Y, Gerasimov ES, Gockley J, Logsdon BA, Duong DM, et al. Brain proteome-wide association study implicates novel proteins in depression pathogenesis. Nat Neurosci. 2021;24:810–7.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  53. Liu J, Li X, Luo XJ. Proteome-wide association study provides insights into the genetic component of protein abundance in psychiatric disorders. Biol Psychiatry. 2021;90:781–9.

    Article  CAS  PubMed  Google Scholar 

  54. Padmanabhan S, Dominiczak AF. Genomics of hypertension: the road to precision medicine. Nat Rev Cardiol. 2021;18:235–50.

    Article  CAS  PubMed  Google Scholar 

  55. Arjona FJ, de Baaij JH, Schlingmann KP, Lameris AL, van Wijk E, Flik G, et al. CNNM2 mutations cause impaired brain development and seizures in patients with hypomagnesemia. PLoS Genet. 2014;10:e1004267.

    Article  PubMed  PubMed Central  Google Scholar 

  56. Yamanaka R, Shindo Y, Oka K. Magnesium is a key player in neuronal maturation and neuropathology. Int J Mol Sci. 2019;20:3439.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  57. Moghaddam B, Homayoun H. Divergent plasticity of prefrontal cortex networks. Neuropsychopharmacology. 2008;33:42–55.

    Article  PubMed  Google Scholar 

  58. Goverti D, Buyukluoglu N, Kaya H, Yuksel RN, Yucel C, Goka E. Neuronal pentraxin-2 (NPTX2) serum levels during an acute psychotic episode in patients with schizophrenia. Psychopharmacology (Berl). 2022;239:2585–91.

    Article  CAS  PubMed  Google Scholar 

  59. Nakai T, Nagai T, Tanaka M, Itoh N, Asai N, Enomoto A, et al. Girdin phosphorylation is crucial for synaptic plasticity and memory: a potential role in the interaction of BDNF/TrkB/Akt signaling with NMDA receptor. J Neurosci. 2014;34:14995–5008.

    Article  PubMed  PubMed Central  Google Scholar 

  60. Liu S, Chen Y, Wang F, Jiang Y, Duan F, Xia Y, et al. Brain transcriptional regulatory architecture and schizophrenia etiology converge between East Asian and European ancestral populations. 2021. bioRxiv: https://doi.org/10.1101/2021.02.04.922880.

  61. Thyme SB, Pieper LM, Li EH, Pandey S, Wang Y, Morris NS, et al. Phenotypic landscape of schizophrenia-associated genes defines candidates and their shared functions. Cell. 2019;177:478–91.e420.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  62. Collingridge GL, Abraham WC. Glutamate receptors and synaptic plasticity: the impact of Evans and Watkins. Neuropharmacology. 2022;206:108922.

    Article  CAS  PubMed  Google Scholar 

  63. Lynch MA. Long-term potentiation and memory. Physiol Rev. 2004;84:87–136.

    Article  CAS  PubMed  Google Scholar 

  64. Harris KM, Jensen FE, Tsao B. Three-dimensional structure of dendritic spines and synapses in rat hippocampus (CA1) at postnatal day 15 and adult ages: implications for the maturation of synaptic physiology and long-term potentiation. J Neurosci. 1992;12:2685–705.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  65. Hering H, Sheng M. Dendritic spines: structure, dynamics and regulation. Nat Rev Neurosci. 2001;2:880–8.

    Article  CAS  PubMed  Google Scholar 

  66. Helm MS, Dankovich TM, Mandad S, Rammner B, Jähne S, Salimi V, et al. A large-scale nanoscopy and biochemistry analysis of postsynaptic dendritic spines. Nat Neurosci. 2021;24:1151–62.

    Article  CAS  PubMed  Google Scholar 

  67. Sanchez-Gonzalez A, Thougaard E, Tapias-Espinosa C, Canete T, Sampedro-Viana D, Saunders JM, et al. Increased thin-spine density in frontal cortex pyramidal neurons in a genetic rat model of schizophrenia-relevant features. Eur Neuropsychopharmacol. 2021;44:79–91.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  68. Glantz LA, Lewis DA. Dendritic spine density in schizophrenia and depression. Arch Gen Psychiatry. 2001;58:203.

    Article  CAS  PubMed  Google Scholar 

  69. Li W, Lv L, Luo XJ. In vivo study sheds new light on the dendritic spine pathology hypothesis of schizophrenia. Mol Psychiatry. 2022;27:1866–8.

    Article  PubMed  Google Scholar 

  70. Wratten NS, Memoli H, Huang Y, Dulencin AM, Matteson PG, Cornacchia MA, et al. Identification of a schizophrenia-associated functional noncoding variant in NOS1AP. Am J Psychiatry. 2009;166:434–41.

    Article  PubMed  PubMed Central  Google Scholar 

  71. Hernandez K, Swiatkowski P, Patel MV, Liang C, Dudzinski NR, Brzustowicz LM, et al. Overexpression of isoforms of nitric oxide synthase 1 adaptor protein, encoded by a risk gene for schizophrenia, alters actin dynamics and synaptic function. Front Cell Neurosci. 2016;10:6.

    Article  PubMed  PubMed Central  Google Scholar 

  72. Hood VL, Paterson C, Law AJ. PI3Kinase-p110delta overexpression impairs dendritic morphogenesis and increases dendritic spine density. Front Mol Neurosci. 2020;13:29.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  73. Badowska DM, Brzozka MM, Kannaiyan N, Thomas C, Dibaj P, Chowdhury A, et al. Modulation of cognition and neuronal plasticity in gain- and loss-of-function mouse models of the schizophrenia risk gene Tcf4. Transl Psychiatry. 2020;10:343.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  74. Dong F, Mao J, Chen M, Yoon J, Mao Y. Schizophrenia risk ZNF804A interacts with its associated proteins to modulate dendritic morphology and synaptic development. Mol Brain. 2021;14:12.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

The authors sincerely acknowledge the important contributions of many publicly available datasets, including the PGC, the ROSMAP and Banner, and the PsychENCODE Consortium. We thank the participants of the ROS, MAP, Mayo, Mount Sinai Brain Bank and Banner Sun Health Research Institute Brain and Body Donation Program for their time and participation. Data were generated as part of the PsychENCODE Consortium, supported by: U01MH103392, U01MH103365, U01MH103346, U01MH103340, U01MH103339, R21MH109956, R21MH105881, R21MH105853, R21MH103877, R21MH102791, R01MH111721, R01MH110928, R01MH110927, R01MH110926, R01MH110921, R01MH110920, R01MH110905, R01MH109715, R01MH109677, R01MH105898, R01MH105898, R01MH094714, P50MH106934, U01MH116488, U01MH116487, U01MH116492, U01MH116489, U01MH116438, U01MH116441, U01MH116442, R01MH114911, R01MH114899, R01MH114901, R01MH117293, R01MH117291, R01MH117292 awarded to: Schahram Akbarian (Icahn School of Medicine at Mount Sinai), Gregory Crawford (Duke University), Stella Dracheva (Icahn School of Medicine at Mount Sinai), Peggy Farnham (University of Southern California), Mark Gerstein (Yale University), Daniel Geschwind (University of California, Los Angeles), Fernando Goes (Johns Hopkins University), Thomas M. Hyde (Lieber Institute for Brain Development), Andrew Jaffe (Lieber Institute for Brain Development), James A. Knowles (University of Southern California), Chunyu Liu (SUNY Upstate Medical University), Dalila Pinto (Icahn School of Medicine at Mount Sinai), Panos Roussos (Icahn School of Medicine at Mount Sinai), Stephan Sanders (University of California, San Francisco), Nenad Sestan (Yale University), Pamela Sklar (Icahn School of Medicine at Mount Sinai), Matthew State (University of California, San Francisco), Patrick Sullivan (University of North Carolina), Flora Vaccarino (Yale University), Daniel Weinberger (Lieber Institute for Brain Development), Sherman Weissman (Yale University), Kevin White (University of Chicago), Jeremy Willsey (University of California, San Francisco), and Peter Zandi (Johns Hopkins University).

Funding

This work was supported by grants from the National Natural Science Foundation of China (U22A20304 and 81971252 to LXL, 82001407 to XS, 32100482 to LW, 81903688 to MLS), Science and Technology Project of Henan Province (212102310587 to MS, 222102310130 to XS), Spring City Plan: the High-level Talent Promotion and Training Project of Kunming (2022SCP001), Yunnan Fundamental Research Projects (202101AT070283 to LW), Youth Project of Medical Science and Technology of Henan Province (SBGJ202103094 to XS), Open Project of Henan Key Laboratory of Biological Psychiatry (ZDSYS2022002 to MS), High Scientific and Technological Research Fund of Xinxiang Medical University (2017ZDCG-04 to LXL), Major Science and Technology Projects of Henan Province (201300310200 to LXL), Project (joint construction) of Medical Science and Technology in Henan Province (LHGJ20190475 to MLS).

Author information

Authors and Affiliations

Authors

Contributions

ML, LXL and MS designed the study and interpreted the results. DYZ, XS and YW performed the primary experiments and analyses. YFY, LWZ, SMC, MLS, WQL, ZHZ and LW contributed to the analyses and results interpretation. DYZ, MS, ML and XS drafted the first manuscript, and all authors contributed to the final version of the paper.

Corresponding authors

Correspondence to Ming Li or Meng Song.

Ethics declarations

Competing interest

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.

Supplementary information

Rights and permissions

Springer Nature or its licensor (e.g. a society or other partner) holds exclusive rights to this article under a publishing agreement with the author(s) or other rightsholder(s); author self-archiving of the accepted manuscript version of this article is solely governed by the terms of such publishing agreement and applicable law.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Zhou, DY., Su, X., Wu, Y. et al. Decreased CNNM2 expression in prefrontal cortex affects sensorimotor gating function, cognition, dendritic spine morphogenesis and risk of schizophrenia. Neuropsychopharmacol. 49, 433–442 (2024). https://doi.org/10.1038/s41386-023-01732-y

Download citation

  • Received:

  • Revised:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41386-023-01732-y

Search

Quick links