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:

Dissecting 16p11.2 hemi-deletion to study sex-specific striatal phenotypes of neurodevelopmental disorders

Abstract

Neurodevelopmental disorders (NDDs) are polygenic in nature and copy number variants (CNVs) are ideal candidates to study the nature of this polygenic risk. The disruption of striatal circuits is considered a central mechanism in NDDs. The 16p11.2 hemi-deletion (16p11.2 del/+) is one of the most common CNVs associated with NDD, and 16p11.2 del/+ mice show sex-specific striatum-related behavioral phenotypes. However, the critical genes among the 27 genes in the 16p11.2 region that underlie these phenotypes remain unknown. Previously, we applied a novel strategy to identify candidate genes associated with the sex-specific phenotypes of 16p11.2 del/+ mice and highlighted three genes within the deleted region: thousand and one amino acid protein kinase 2 (Taok2), seizure-related 6 homolog-like 2 (Sez6l2), and major vault protein (Mvp). Using CRISPR/Cas9, we generated mice carrying null mutations in Taok2, Sez6l2, and Mvp (3 gene hemi-deletion (3g del/+)). Hemi-deletion of these 3 genes recapitulates sex-specific behavioral alterations in striatum-dependent behavioral tasks observed in 16p11.2 del/+ mice, specifically male-specific hyperactivity and impaired motivation for reward seeking. Moreover, RNAseq analysis revealed that 3g del/+ mice exhibit gene expression changes in the striatum similar to 16p11.2 del/+ mice exclusively in males. Subsequent analysis identified translation dysregulation and/or extracellular signal-regulated kinase signaling as plausible molecular mechanisms underlying male-specific, striatum-dependent behavioral alterations. Interestingly, ribosomal profiling supported the notion of translation dysregulation in both 3g del/+ and 16p11.2 del/+ male mice. However, mice carrying a 4-gene deletion (with an additional deletion of Mapk3) exhibited fewer phenotypic similarities with 16p11.2 del/+ mice. Together, the mutation of 3 genes within the 16p11.2 region phenocopies striatal sex-specific phenotypes of 16p11.2 del/+ mice. These results support the importance of a polygenic approach to study NDDs and underscore that the effects of the large genetic deletions result from complex interactions between multiple candidate genes.

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: Generation of 3g del/+ mice.
Fig. 2: 3g del/+ mice reproduced sex-specific changes in striatal behavior.
Fig. 3: Gene expression changes in the striatum of male 3g del/+ mice are consistent with 16p11.2 del/+ males.
Fig. 4: Sex-specific changes in white matter during development in 3g del/+ mice.
Fig. 5: Additional hemi-deletion of Mapk3 does not increase molecular and behavioral alterations observed in 3g del/+ mice.

Similar content being viewed by others

Data availability

The dataset supporting the conclusions of this article is available in the NCBI’s Gene Expression Omnibus repository, GEO Series accession GSE224750.

References

  1. Baxter AJ, Brugha TS, Erskine HE, Scheurer RW, Vos T, Scott JG. The epidemiology and global burden of autism spectrum disorders. Psychol Med. 2015;45:601–613.

    Article  CAS  PubMed  Google Scholar 

  2. Grove J, Ripke S, Als TD, Mattheisen M, Walters RK, Won H, et al. Identification of common genetic risk variants for autism spectrum disorder. Nat Genet. 2019;51:431–444.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. Faraone SV, Larsson H. Genetics of attention deficit hyperactivity disorder. Mol Psychiatry. 2019;24:562–575.

    Article  CAS  PubMed  Google Scholar 

  4. 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;87:1215–1233.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Sebat J, Lakshmi B, Malhotra D, Troge J, Lese-Martin C, Walsh T, et al. Strong association of de novo copy number mutations with autism. Science. 2007;316:445–449.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Fu JM, Satterstrom FK, Peng M, Brand H, Collins RL, Dong S, et al. Rare coding variation provides insight into the genetic architecture and phenotypic context of autism. Nat Genet. 2022;54:1320–1331.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Hanson E, Bernier R, Porche K, Jackson FI, Goin-Kochel RP, Snyder LG, et al. The cognitive and behavioral phenotype of the 16p11.2 deletion in a clinically ascertained population. Biol Psychiatry. 2015;77:785–793.

    Article  CAS  PubMed  Google Scholar 

  8. Rein B, Yan Z. 16p11.2 copy number variations and neurodevelopmental disorders. Trends Neurosci. 2020;43:886–901.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Angelakos CC, Watson AJ, O’Brien WT, Krainock KS, Nickl-Jockschat T, Abel T. Hyperactivity and male-specific sleep deficits in the 16p11.2 deletion mouse model of autism. Autism Res. 2017;10:572–584.

    Article  PubMed  Google Scholar 

  10. Grissom NM, McKee SE, Schoch H, Bowman N, Havekes R, O’Brien WT, et al. Male-specific deficits in natural reward learning in a mouse model of neurodevelopmental disorders. Mol Psychiatry. 2018;23:544–555.

    Article  CAS  PubMed  Google Scholar 

  11. Lynch JF 3rd, Ferri SL, Angelakos C, Schoch H, Nickl-Jockschat T, Gonzalez A, et al. Comprehensive behavioral phenotyping of a 16p11.2 del mouse model for neurodevelopmental disorders. Autism Res. 2020;13:1670–1684.

    Article  PubMed  PubMed Central  Google Scholar 

  12. Sonderby IE, Gustafsson O, Doan NT, Hibar DP, Martin-Brevet S, Abdellaoui A, et al. Dose response of the 16p11.2 distal copy number variant on intracranial volume and basal ganglia. Mol Psychiatry. 2020;25:584–602.

    Article  PubMed  Google Scholar 

  13. Hudac CM, Bove J, Barber S, Duyzend M, Wallace A, Martin CL, et al. Evaluating heterogeneity in ASD symptomatology, cognitive ability, and adaptive functioning among 16p11.2 CNV carriers. Autism Res. 2020;13:1300–1310.

    Article  PubMed  Google Scholar 

  14. Nickl-Jockschat T, Habel U, Michel TM, Manning J, Laird AR, Fox PT, et al. Brain structure anomalies in autism spectrum disorder–a meta-analysis of VBM studies using anatomic likelihood estimation. Hum Brain Mapp. 2012;33:1470–1489.

    Article  PubMed  Google Scholar 

  15. Langen M, Bos D, Noordermeer SD, Nederveen H, van Engeland H, Durston S. Changes in the development of striatum are involved in repetitive behavior in autism. Biol Psychiatry. 2014;76:405–411.

    Article  PubMed  Google Scholar 

  16. Langen M, Schnack HG, Nederveen H, Bos D, Lahuis BE, de Jonge MV, et al. Changes in the developmental trajectories of striatum in autism. Biol Psychiatry. 2009;66:327–333.

    Article  PubMed  Google Scholar 

  17. Haznedar MM, Buchsbaum MS, Hazlett EA, LiCalzi EM, Cartwright C, Hollander E. Volumetric analysis and three-dimensional glucose metabolic mapping of the striatum and thalamus in patients with autism spectrum disorders. Am J Psychiatry. 2006;163:1252–1263.

    Article  PubMed  Google Scholar 

  18. Di Martino A, Zuo XN, Kelly C, Grzadzinski R, Mennes M, Schvarcz A, et al. Shared and distinct intrinsic functional network centrality in autism and attention-deficit/hyperactivity disorder. Biol Psychiatry. 2013;74:623–632.

    Article  PubMed  PubMed Central  Google Scholar 

  19. Langen M, Leemans A, Johnston P, Ecker C, Daly E, Murphy CM, et al. Fronto-striatal circuitry and inhibitory control in autism: findings from diffusion tensor imaging tractography. Cortex. 2012;48:183–193.

    Article  PubMed  Google Scholar 

  20. Castellanos FX, Giedd JN, Eckburg P, Marsh WL, Vaituzis AC, Kaysen D, et al. Quantitative morphology of the caudate nucleus in attention deficit hyperactivity disorder. Am J Psychiatry. 1994;151:1791–1796.

    Article  CAS  PubMed  Google Scholar 

  21. Semrud-Clikeman M, Pliszka SR, Bledsoe J, Lancaster J. Volumetric MRI differences in treatment naive and chronically treated adolescents with ADHD-combined type. J Atten Disord. 2014;18:511–520.

    Article  PubMed  Google Scholar 

  22. Badgaiyan RD, Sinha S, Sajjad M, Wack DS. Attenuated tonic and enhanced phasic release of dopamine in attention deficit hyperactivity disorder. PLoS One. 2015;10:e0137326.

    Article  PubMed  PubMed Central  Google Scholar 

  23. Fuccillo MV. Striatal circuits as a common node for autism pathophysiology. Front Neurosci. 2016;10:27.

    Article  PubMed  PubMed Central  Google Scholar 

  24. Janouschek H, Chase HW, Sharkey RJ, Peterson ZJ, Camilleri JA, Abel T, et al. The functional neural architecture of dysfunctional reward processing in autism. Neuroimage Clin. 2021;31:102700.

    Article  PubMed  PubMed Central  Google Scholar 

  25. Chase HW, Loriemi P, Wensing T, Eickhoff SB, Nickl-Jockschat T. Meta-analytic evidence for altered mesolimbic responses to reward in schizophrenia. Hum Brain Mapp. 2018;39:2917–2928.

    Article  PubMed  PubMed Central  Google Scholar 

  26. Faheem M, Akram W, Akram H, Khan MA, Siddiqui FA, Majeed I. Gender-based differences in prevalence and effects of ADHD in adults: a systematic review. Asian J Psychiatr. 2022;75:103205.

    Article  PubMed  Google Scholar 

  27. Santos S, Ferreira H, Martins J, Goncalves J, Castelo-Branco M. Male sex bias in early and late onset neurodevelopmental disorders: shared aspects and differences in autism spectrum disorder, attention deficit/hyperactivity disorder, and schizophrenia. Neurosci Biobehav Rev. 2022;135:104577.

    Article  CAS  PubMed  Google Scholar 

  28. Werling DM, Geschwind DH. Understanding sex bias in autism spectrum disorder. Proc Natl Acad Sci USA. 2013;110:4868–4869.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Ferri SL, Abel T, Brodkin ES. Sex differences in autism spectrum disorder: a review. Curr Psychiatry Rep. 2018;20:9.

    Article  PubMed  PubMed Central  Google Scholar 

  30. Bolte S, Neufeld J, Marschik PB, Williams ZJ, Gallagher L, Lai MC. Sex and gender in neurodevelopmental conditions. Nat Rev Neurol. 2023;19:136–159.

    Article  PubMed  PubMed Central  Google Scholar 

  31. Agarwalla S, Arroyo NS, Long NE, O’Brien WT, Abel T, Bandyopadhyay S. Male-specific alterations in structure of isolation call sequences of mouse pups with 16p11.2 deletion. Genes Brain Behav. 2020;19:e12681.

    Article  CAS  PubMed  Google Scholar 

  32. Kumar VJ, Grissom NM, McKee SE, Schoch H, Bowman N, Havekes R, et al. Linking spatial gene expression patterns to sex-specific brain structural changes on a mouse model of 16p11.2 hemideletion. Transl Psychiatry. 2018;8:109.

    Article  PubMed  PubMed Central  Google Scholar 

  33. Martin Lorenzo S, Nalesso V, Chevalier C, Birling MC, Herault Y. Targeting the RHOA pathway improves learning and memory in adult Kctd13 and 16p11.2 deletion mouse models. Mol Autism. 2021;12:1.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Richter M, Murtaza N, Scharrenberg R, White SH, Johanns O, Walker S, et al. Altered TAOK2 activity causes autism-related neurodevelopmental and cognitive abnormalities through RhoA signaling. Mol Psychiatry. 2019;24:1329–1350.

    Article  CAS  PubMed  Google Scholar 

  35. de Anda FC, Rosario AL, Durak O, Tran T, Graff J, Meletis K, et al. Autism spectrum disorder susceptibility gene TAOK2 affects basal dendrite formation in the neocortex. Nat Neurosci. 2012;15:1022–1031.

    Article  PubMed  PubMed Central  Google Scholar 

  36. Ip JPK, Nagakura I, Petravicz J, Li K, Wiemer EAC, Sur M. Major vault protein, a candidate gene in 16p11.2 microdeletion syndrome, is required for the homeostatic regulation of visual cortical plasticity. J Neurosci. 2018;38:3890–3900.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Ferguson SM, Fasano S, Yang P, Brambilla R, Robinson TE. Knockout of ERK1 enhances cocaine-evoked immediate early gene expression and behavioral plasticity. Neuropsychopharmacology. 2006;31:2660–2668.

    Article  CAS  PubMed  Google Scholar 

  38. Mazzucchelli C, Vantaggiato C, Ciamei A, Fasano S, Pakhotin P, Krezel W, et al. Knockout of ERK1 MAP kinase enhances synaptic plasticity in the striatum and facilitates striatal-mediated learning and memory. Neuron. 2002;34:807–820.

    Article  CAS  PubMed  Google Scholar 

  39. Kretz PF, Wagner C, Mikhaleva A, Montillot C, Hugel S, Morella I, et al. Dissecting the autism-associated 16p11.2 locus identifies multiple drivers in neuroanatomical phenotypes and unveils a male-specific role for the major vault protein. Genome Biol. 2023;24:261.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Weiner DJ, Ling E, Erdin S, Tai DJC, Yadav R, Grove J, et al. Statistical and functional convergence of common and rare genetic influences on autism at chromosome 16p. Nat Genet. 2022;54:1630–1639.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Yadav S, Oses-Prieto JA, Peters CJ, Zhou J, Pleasure SJ, Burlingame AL, et al. TAOK2 kinase mediates PSD95 stability and dendritic spine maturation through Septin7 phosphorylation. Neuron. 2017;93:379–393.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Miyazaki T, Hashimoto K, Uda A, Sakagami H, Nakamura Y, Saito SY, et al. Disturbance of cerebellar synaptic maturation in mutant mice lacking BSRPs, a novel brain-specific receptor-like protein family. FEBS Lett. 2006;580:4057–4064.

    Article  CAS  PubMed  Google Scholar 

  43. Boonen M, Staudt C, Gilis F, Oorschot V, Klumperman J, Jadot M, et al. and its newly identified transport receptor SEZ6L2 can modulate neurite outgrowth. J Cell Sci. 2016;129:557–568.

    CAS  PubMed  Google Scholar 

  44. Lotsch D, Steiner E, Holzmann K, Spiegl-Kreinecker S, Pirker C, Hlavaty J, et al. Major vault protein supports glioblastoma survival and migration by upregulating the EGFR/PI3K signalling axis. Oncotarget. 2013;4:1904–1918.

    Article  PubMed  PubMed Central  Google Scholar 

  45. Steiner E, Holzmann K, Pirker C, Elbling L, Micksche M, Sutterluty H, et al. The major vault protein is responsive to and interferes with interferon-gamma-mediated STAT1 signals. J Cell Sci. 2006;119:459–469.

    Article  CAS  PubMed  Google Scholar 

  46. Nash A, Aumann TD, Pigoni M, Lichtenthaler SF, Takeshima H, Munro KM, et al. Lack of Sez6 family proteins impairs motor functions, short-term memory, and cognitive flexibility and alters dendritic spine properties. Cereb Cortex. 2020;30:2167–2184.

    Article  PubMed  Google Scholar 

  47. Pinkert CA, Pinkert CA. Transgenic animal technology: a laboratory handbook. 2nd edn. Amsterdam; Boston: Academic Press; 2002.

  48. Angelakos CC, Tudor JC, Ferri SL, Jongens TA, Abel T. Home-cage hypoactivity in mouse genetic models of autism spectrum disorder. Neurobiol Learn Mem. 2019;165:107000.

    Article  PubMed  PubMed Central  Google Scholar 

  49. Brunner D, Kabitzke P, He D, Cox K, Thiede L, Hanania T, et al. Comprehensive analysis of the 16p11.2 deletion and null Cntnap2 mouse models of autism spectrum disorder. PLoS One. 2015;10:e0134572.

    Article  PubMed  PubMed Central  Google Scholar 

  50. Portmann T, Yang M, Mao R, Panagiotakos G, Ellegood J, Dolen G, et al. Behavioral abnormalities and circuit defects in the basal ganglia of a mouse model of 16p11.2 deletion syndrome. Cell Rep. 2014;7:1077–1092.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  51. Bali P, Kenny PJ. Transcriptional mechanisms of drug addiction. Dialogues Clin Neurosci. 2019;21:379–387.

    Article  PubMed  PubMed Central  Google Scholar 

  52. Miyashita Y. Cognitive memory: cellular and network machineries and their top-down control. Science. 2004;306:435–440.

    Article  CAS  PubMed  Google Scholar 

  53. Engeln M, Fox ME, Chandra R, Choi EY, Nam H, Qadir H, et al. Transcriptome profiling of the ventral pallidum reveals a role for pallido-thalamic neurons in cocaine reward. Mol Psychiatry. 2022;27:3980–3991.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. Brar GA, Weissman JS. Ribosome profiling reveals the what, when, where and how of protein synthesis. Nat Rev Mol Cell Biol. 2015;16:651–664.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  55. Meoded A, Huisman T. Diffusion tensor imaging of brain malformations: exploring the internal architecture. Neuroimaging Clin N Am. 2019;29:423–434.

    Article  PubMed  Google Scholar 

  56. Maillard AM, Ruef A, Pizzagalli F, Migliavacca E, Hippolyte L, Adaszewski S, et al. The 16p11.2 locus modulates brain structures common to autism, schizophrenia and obesity. Mol Psychiatry. 2015;20:140–147.

    Article  CAS  PubMed  Google Scholar 

  57. Kolli S, Zito CI, Mossink MH, Wiemer EA, Bennett AM. The major vault protein is a novel substrate for the tyrosine phosphatase SHP-2 and scaffold protein in epidermal growth factor signaling. J Biol Chem. 2004;279:29374–29385.

    Article  CAS  PubMed  Google Scholar 

  58. Kim E, Lee S, Mian MF, Yun SU, Song M, Yi KS, et al. Crosstalk between Src and major vault protein in epidermal growth factor-dependent cell signalling. FEBS J. 2006;273:793–804.

    Article  CAS  PubMed  Google Scholar 

  59. Zhang N, Liu L, Fan N, Zhang Q, Wang W, Zheng M, et al. The requirement of SEPT2 and SEPT7 for migration and invasion in human breast cancer via MEK/ERK activation. Oncotarget. 2016;7:61587–61600.

    Article  PubMed  PubMed Central  Google Scholar 

  60. Tai DJC, Razaz P, Erdin S, Gao D, Wang J, Nuttle X, et al. Tissue- and cell-type-specific molecular and functional signatures of 16p11.2 reciprocal genomic disorder across mouse brain and human neuronal models. Am J Hum Genet. 2022;109:1789–1813.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  61. Hetman M, Slomnicki LP. Ribosomal biogenesis as an emerging target of neurodevelopmental pathologies. J Neurochem. 2019;148:325–347.

    Article  CAS  PubMed  Google Scholar 

  62. Santini E, Klann E. Reciprocal signaling between translational control pathways and synaptic proteins in autism spectrum disorders. Sci Signal. 2014;7:re10.

    Article  PubMed  PubMed Central  Google Scholar 

  63. Tee AR, Blenis J. mTOR, translational control and human disease. Semin Cell Dev Biol. 2005;16:29–37.

    Article  CAS  PubMed  Google Scholar 

  64. Ifrim MF, Williams KR, Bassell GJ. Single-molecule imaging of PSD-95 mRNA translation in dendrites and its dysregulation in a mouse model of fragile X syndrome. J Neurosci. 2015;35:7116–7130.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  65. Jishi A, Qi X, Miranda HC. Implications of mRNA translation dysregulation for neurological disorders. Semin Cell Dev Biol. 2021;114:11–19.

    Article  CAS  PubMed  Google Scholar 

  66. Ceman S, O’Donnell WT, Reed M, Patton S, Pohl J, Warren ST. Phosphorylation influences the translation state of FMRP-associated polyribosomes. Hum Mol Genet. 2003;12:3295–3305.

    Article  CAS  PubMed  Google Scholar 

  67. Inoki K, Li Y, Xu T, Guan KL. Rheb GTPase is a direct target of TSC2 GAP activity and regulates mTOR signaling. Genes Dev. 2003;17:1829–1834.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  68. Xue S, Barna M. Specialized ribosomes: a new frontier in gene regulation and organismal biology. Nat Rev Mol Cell Biol. 2012;13:355–369.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  69. Qureshi AY, Mueller S, Snyder AZ, Mukherjee P, Berman JI, Roberts TP, et al. Opposing brain differences in 16p11.2 deletion and duplication carriers. J Neurosci. 2014;34:11199–11211.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  70. Martin-Brevet S, Rodriguez-Herreros B, Nielsen JA, Moreau C, Modenato C, Maillard AM, et al. Quantifying the effects of 16p11.2 copy number variants on brain structure: a multisite genetic-first study. Biol Psychiatry. 2018;84:253–264.

    Article  CAS  PubMed  Google Scholar 

  71. Horev G, Ellegood J, Lerch JP, Son YE, Muthuswamy L, Vogel H, et al. Dosage-dependent phenotypes in models of 16p11.2 lesions found in autism. Proc Natl Acad Sci USA. 2011;108:17076–17081.

    Article  PubMed  PubMed Central  Google Scholar 

  72. Ellegood J, Anagnostou E, Babineau BA, Crawley JN, Lin L, Genestine M, et al. Clustering autism: using neuroanatomical differences in 26 mouse models to gain insight into the heterogeneity. Mol Psychiatry. 2015;20:118–125.

    Article  CAS  PubMed  Google Scholar 

  73. Pucilowska J, Vithayathil J, Tavares EJ, Kelly C, Karlo JC, Landreth GE. The 16p11.2 deletion mouse model of autism exhibits altered cortical progenitor proliferation and brain cytoarchitecture linked to the ERK MAPK pathway. J Neurosci. 2015;35:3190–3200.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  74. Pucilowska J, Vithayathil J, Pagani M, Kelly C, Karlo JC, Robol C, et al. Pharmacological inhibition of ERK signaling rescues pathophysiology and behavioral phenotype associated with 16p11.2 chromosomal deletion in mice. J Neurosci. 2018;38:6640–6652.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  75. Sweatt JD. The neuronal MAP kinase cascade: a biochemical signal integration system subserving synaptic plasticity and memory. J Neurochem. 2001;76:1–10.

    Article  CAS  PubMed  Google Scholar 

  76. Kelleher RJ 3rd, Govindarajan A, Tonegawa S. Translational regulatory mechanisms in persistent forms of synaptic plasticity. Neuron. 2004;44:59–73.

    Article  CAS  PubMed  Google Scholar 

  77. Shiflett MW, Balleine BW. Contributions of ERK signaling in the striatum to instrumental learning and performance. Behav Brain Res. 2011;218:240–247.

    Article  CAS  PubMed  Google Scholar 

  78. Selcher JC, Nekrasova T, Paylor R, Landreth GE, Sweatt JD. Mice lacking the ERK1 isoform of MAP kinase are unimpaired in emotional learning. Learn Mem. 2001;8:11–19.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  79. Engel SR, Creson TK, Hao Y, Shen Y, Maeng S, Nekrasova T, et al. The extracellular signal-regulated kinase pathway contributes to the control of behavioral excitement. Mol Psychiatry. 2009;14:448–461.

    Article  CAS  PubMed  Google Scholar 

  80. Talley MJ, Nardini D, Qin S, Prada CE, Ehrman LA, Waclaw RR. A role for sustained MAPK activity in the mouse ventral telencephalon. Dev Biol. 2021;476:137–147.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  81. Scharrenberg R, Richter M, Johanns O, Meka DP, Rucker T, Murtaza N, et al. TAOK2 rescues autism-linked developmental deficits in a 16p11.2 microdeletion mouse model. Mol Psychiatry. 2022;27:4707–4721.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

This work was supported by The University of Iowa Hawkeye Intellectual and Developmental Disabilities Research Center (HAWK-IDDRC) P50 HD103556 (TA and Lane Strathearn, PI), the Roy J. Carver Chair in Neuroscience (TA), Interdisciplinary Graduate Program in Genetics at University of Iowa (YV), NIH grant R01 MH 087463 (TA), Simons Foundation Autism Research Initiative (SFARI) grant 345034 (TA), NIH grants T32 GM067795 and F31 MH134542 (BK), Eagles Autism Challenge (TN-J) and the Andrew H. Woods Professorship (TN-J). Transgenic mice were generated at the University of Iowa Genome Editing Core Facility directed by William Paradee, PhD and supported in part by grants from the NIH and from the Roy J. and Lucille A. Carver College of Medicine. We wish to thank Norma Sinclair, Patricia Yarolem, Joanne Schwarting and Rongbin Guan for their technical expertise in generating transgenic mice. The Neural Circuits and Behavior Core in the Iowa Neuroscience Institute provided equipment, facilities, and consultations services to support investigators in performing behavioral tasks. The Iowa Institute of Human Genetics provided Sanger DNA sequencing and RNA sequencing services. The Iowa Magnetic Resonance Research Facility provides access to a small animal MRI scanner as well as the necessary data processing equipment for animal MR imaging. We thank TB-SEQ, Inc. (Palo Alto, CA) for Ribosome profiling and helpful discussion.

Author information

Authors and Affiliations

Authors

Contributions

TA and TN-J conceptualized the project, supervised data collection and analysis. JK, TN-J, and TA wrote the manuscript with inputs from all the authors. JK, BK, and EB performed behavior, biochemical and molecular biology experiments. YV performed the bioinformatic analysis. ZP performed MRI imaging experiments and analysis. MEG conceptualized and designed the novel genetic mouse model.

Corresponding authors

Correspondence to Thomas Nickl-Jockschat or Ted Abel.

Ethics declarations

Competing interests

TA serves on the Scientific Advisory Board of EmbarkNeuro and is a scientific advisor to Aditum Bio and Radius Health.

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

Kim, J., Vanrobaeys, Y., Kelvington, B. et al. Dissecting 16p11.2 hemi-deletion to study sex-specific striatal phenotypes of neurodevelopmental disorders. Mol Psychiatry (2024). https://doi.org/10.1038/s41380-024-02411-0

Download citation

  • Received:

  • Revised:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1038/s41380-024-02411-0

Search

Quick links