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:

TSHZ3 deletion causes an autism syndrome and defects in cortical projection neurons

Abstract

TSHZ3, which encodes a zinc-finger transcription factor, was recently positioned as a hub gene in a module of the genes with the highest expression in the developing human neocortex, but its functions remained unknown. Here we identify TSHZ3 as the critical region for a syndrome associated with heterozygous deletions at 19q12-q13.11, which includes autism spectrum disorder (ASD). In Tshz3-null mice, differentially expressed genes include layer-specific markers of cerebral cortical projection neurons (CPNs), and the human orthologs of these genes are strongly associated with ASD. Furthermore, mice heterozygous for Tshz3 show functional changes at synapses established by CPNs and exhibit core ASD-like behavioral abnormalities. These findings highlight essential roles for Tshz3 in CPN development and function, whose alterations can account for ASD in the newly defined TSHZ3 deletion syndrome.

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

Access options

Rent or buy this article

Prices vary by article type

from$1.95

to$39.95

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

Figure 1: Schematic of deletions of the TSHZ3 locus and TSHZ3 expression in the human and mouse fetal neocortex.
Figure 2: Tshz3lacZ/lacZ mice show altered gene expression of cortical layer markers at E18.5.
Figure 3: Cortical layering and major axonal tracts are preserved in Tshz3lacZ/lacZ brains at E18.5.
Figure 4: Tshz3lacZ/+ mice exhibit altered expression of genes in the neocortex from embryonic to postnatal stages, but the main projection systems from deep CPNs are preserved.
Figure 5: Altered corticostriatal synaptic transmission and plasticity in Tshz3lacZ/+ mice.
Figure 6: Tshz3lacZ/+ mice display autism-like behavioral deficits.

Similar content being viewed by others

Accession codes

Primary accessions

Gene Expression Omnibus

References

  1. American Psychiatric Association. Diagnostic and Statistical Manual of Mental Disorders, DSM-5 5th edn. (American Psychiatric Association, 2013).

  2. Buxbaum, J.D. et al. The autism sequencing consortium: large-scale, high-throughput sequencing in autism spectrum disorders. Neuron 76, 1052–1056 (2012).

    CAS  PubMed  Google Scholar 

  3. Parikshak, N.N. et al. Integrative functional genomic analyses implicate specific molecular pathways and circuits in autism. Cell 155, 1008–1021 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  4. State, M.W. & Šestan, N. The emerging biology of autism spectrum disorders. Science 337, 1301–1303 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  5. Willsey, A.J. et al. Coexpression networks implicate human midfetal deep cortical projection neurons in the pathogenesis of autism. Cell 155, 997–1007 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  6. Belmonte, M.K. et al. Autism and abnormal development of brain connectivity. J. Neurosci. 24, 9228–9231 (2004).

    CAS  PubMed  PubMed Central  Google Scholar 

  7. Kwan, K.Y., Sestan, N. & Anton, E.S. Transcriptional co-regulation of neuronal migration and laminar identity in the neocortex. Development 139, 1535–1546 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  8. Rosenfeld, J.A. et al. Small deletions of SATB2 cause some of the clinical features of the 2q33.1 microdeletion syndrome. PLoS One 4, e6568 (2009).

    PubMed  PubMed Central  Google Scholar 

  9. Rosenfeld, J.A. et al. Copy number variations associated with autism spectrum disorders contribute to a spectrum of neurodevelopmental disorders. Genet. Med. 12, 694–702 (2010).

    PubMed  Google Scholar 

  10. Wang, K. et al. Common genetic variants on 5p14.1 associate with autism spectrum disorders. Nature 459, 528–533 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  11. O'Roak, B.J. et al. Multiplex targeted sequencing identifies recurrently mutated genes in autism spectrum disorders. Science 338, 1619–1622 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  12. Kang, H.J. et al. Spatio-temporal transcriptome of the human brain. Nature 478, 483–489 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  13. Ariani, F. et al. FOXG1 is responsible for the congenital variant of Rett syndrome. Am. J. Hum. Genet. 83, 89–93 (2008).

    CAS  PubMed  PubMed Central  Google Scholar 

  14. Chen, B., Schaevitz, L.R. & McConnell, S.K. Fezl regulates the differentiation and axon targeting of layer 5 subcortical projection neurons in cerebral cortex. Proc. Natl. Acad. Sci. USA 102, 17184–17189 (2005).

    CAS  PubMed  PubMed Central  Google Scholar 

  15. Chen, J.G., Rasin, M.R., Kwan, K.Y. & Sestan, N. Zfp312 is required for subcortical axonal projections and dendritic morphology of deep-layer pyramidal neurons of the cerebral cortex. Proc. Natl. Acad. Sci. USA 102, 17792–17797 (2005).

    CAS  PubMed  PubMed Central  Google Scholar 

  16. Han, W. et al. TBR1 directly represses Fezf2 to control the laminar origin and development of the corticospinal tract. Proc. Natl. Acad. Sci. USA 108, 3041–3046 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  17. Bedogni, F. et al. Tbr1 regulates regional and laminar identity of postmitotic neurons in developing neocortex. Proc. Natl. Acad. Sci. USA 107, 13129–13134 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  18. Molyneaux, B.J., Arlotta, P., Hirata, T., Hibi, M. & Macklis, J.D. Fezl is required for the birth and specification of corticospinal motor neurons. Neuron 47, 817–831 (2005).

    CAS  PubMed  Google Scholar 

  19. Leone, D.P. et al. Satb2 regulates the differentiation of both callosal and subcerebral projection neurons in the developing cerebral cortex. Cereb. Cortex 25, 3406–3419 (2015).

    PubMed  Google Scholar 

  20. Bishop, K.M., Garel, S., Nakagawa, Y., Rubenstein, J.L. & O'Leary, D.D. Emx1 and Emx2 cooperate to regulate cortical size, lamination, neuronal differentiation, development of cortical efferents, and thalamocortical pathfinding. J. Comp. Neurol. 457, 345–360 (2003).

    CAS  PubMed  Google Scholar 

  21. Britanova, O. et al. Satb2 is a postmitotic determinant for upper-layer neuron specification in the neocortex. Neuron 57, 378–392 (2008).

    CAS  PubMed  Google Scholar 

  22. International Molecular Genetic Study of Autism Consortium. A full genome screen for autism with evidence for linkage to a region on chromosome 7q. Hum. Mol. Genet. 7, 571–578 (1998).

  23. Liu, J. et al. A genomewide screen for autism susceptibility loci. Am. J. Hum. Genet. 69, 327–340 (2001).

    CAS  PubMed  PubMed Central  Google Scholar 

  24. Hussman, J.P. et al. A noise-reduction GWAS analysis implicates altered regulation of neurite outgrowth and guidance in autism. Mol. Autism 2, 1 (2011).

    PubMed  PubMed Central  Google Scholar 

  25. Caubit, X. et al. Teashirt 3 regulates development of neurons involved in both respiratory rhythm and airflow control. J. Neurosci. 30, 9465–9476 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  26. Caubit, X., Tiveron, M.C., Cremer, H. & Fasano, L. Expression patterns of the three Teashirt-related genes define specific boundaries in the developing and postnatal mouse forebrain. J. Comp. Neurol. 486, 76–88 (2005).

    CAS  PubMed  Google Scholar 

  27. Adalat, S., Bockenhauer, D., Ledermann, S.E., Hennekam, R.C. & Woolf, A.S. Renal malformations associated with mutations of developmental genes: messages from the clinic. Pediatr. Nephrol. 25, 2247–2255 (2010).

    PubMed  PubMed Central  Google Scholar 

  28. Chowdhury, S. et al. Phenotypic and molecular characterization of 19q12-q13.1 deletions: a report of five patients. Am. J. Med. Genet. A 164A, 62–69 (2014).

    PubMed  Google Scholar 

  29. Kulharya, A.S., Michaelis, R.C., Norris, K.S., Taylor, H.A. & Garcia-Heras, J. Constitutional del(19)(q12q13.1) in a three-year-old girl with severe phenotypic abnormalities affecting multiple organ systems. Am. J. Med. Genet. 77, 391–394 (1998).

    CAS  PubMed  Google Scholar 

  30. Malan, V. et al. 19q13.11 deletion syndrome: a novel clinically recognisable genetic condition identified by array comparative genomic hybridisation. J. Med. Genet. 46, 635–640 (2009).

    CAS  PubMed  Google Scholar 

  31. Kwan, K.Y. et al. SOX5 postmitotically regulates migration, postmigratory differentiation, and projections of subplate and deep-layer neocortical neurons. Proc. Natl. Acad. Sci. USA 105, 16021–16026 (2008).

    CAS  PubMed  PubMed Central  Google Scholar 

  32. Lai, T. et al. SOX5 controls the sequential generation of distinct corticofugal neuron subtypes. Neuron 57, 232–247 (2008).

    CAS  PubMed  Google Scholar 

  33. Li, H. et al. Transcription factor MEF2C influences neural stem/progenitor cell differentiation and maturation in vivo. Proc. Natl. Acad. Sci. USA 105, 9397–9402 (2008).

    CAS  PubMed  PubMed Central  Google Scholar 

  34. Novara, F. et al. Refining the phenotype associated with MEF2C haploinsufficiency. Clin. Genet. 78, 471–477 (2010).

    CAS  PubMed  Google Scholar 

  35. Arlotta, P. et al. Neuronal subtype-specific genes that control corticospinal motor neuron development in vivo. Neuron 45, 207–221 (2005).

    CAS  PubMed  Google Scholar 

  36. Workman, A.D., Charvet, C.J., Clancy, B., Darlington, R.B. & Finlay, B.L. Modeling transformations of neurodevelopmental sequences across mammalian species. J. Neurosci. 33, 7368–7383 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  37. Hoerder-Suabedissen, A. et al. Expression profiling of mouse subplate reveals a dynamic gene network and disease association with autism and schizophrenia. Proc. Natl. Acad. Sci. USA 110, 3555–3560 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  38. Mi, H., Poudel, S., Muruganujan, A., Casagrande, J.T. & Thomas, P.D. PANTHER version 10: expanded protein families and functions, and analysis tools. Nucleic Acids Res. 44 D1, D336–D342 (2016).

    CAS  PubMed  Google Scholar 

  39. Caubit, X. et al. Teashirt 3 is necessary for ureteral smooth muscle differentiation downstream of SHH and BMP4. Development 135, 3301–3310 (2008).

    CAS  PubMed  Google Scholar 

  40. Shepherd, G.M. Corticostriatal connectivity and its role in disease. Nat. Rev. Neurosci. 14, 278–291 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  41. Sohur, U.S., Padmanabhan, H.K., Kotchetkov, I.S., Menezes, J.R. & Macklis, J.D. Anatomic and molecular development of corticostriatal projection neurons in mice. Cereb. Cortex 24, 293–303 (2014).

    PubMed  Google Scholar 

  42. Arlotta, P., Molyneaux, B.J., Jabaudon, D., Yoshida, Y. & Macklis, J.D. Ctip2 controls the differentiation of medium spiny neurons and the establishment of the cellular architecture of the striatum. J. Neurosci. 28, 622–632 (2008).

    CAS  PubMed  PubMed Central  Google Scholar 

  43. Varlinskaya, E.I. & Spear, L.P. Acute effects of ethanol on social behavior of adolescent and adult rats: role of familiarity of the test situation. Alcohol. Clin. Exp. Res. 26, 1502–1511 (2002).

    PubMed  Google Scholar 

  44. Roubertoux, P.L., Carlier, M. & Tordjman, S. in Organism Models of Autism Spectrum Disorders (ed. Roubertoux, P.L.) 335–370 (Springer, 2015).

  45. Nadler, J.J. et al. Automated apparatus for quantitation of social approach behaviors in mice. Genes Brain Behav. 3, 303–314 (2004).

    CAS  PubMed  Google Scholar 

  46. Moy, S.S. et al. Sociability and preference for social novelty in five inbred strains: an approach to assess autistic-like behavior in mice. Genes Brain Behav. 3, 287–302 (2004).

    CAS  PubMed  Google Scholar 

  47. Irie, F., Badie-Mahdavi, H. & Yamaguchi, Y. Autism-like socio-communicative deficits and stereotypies in mice lacking heparan sulfate. Proc. Natl. Acad. Sci. USA 109, 5052–5056 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  48. Makanjuola, R.O., Hill, G., Maben, I., Dow, R.C. & Ashcroft, G.W. An automated method for studying exploratory and stereotyped behaviour in rats. Psychopharmacology (Berl.) 52, 271–277 (1977).

    CAS  Google Scholar 

  49. Cohen, J. Statistical Power Analysis for the Behavioral Sciences (Routledge, 1988).

  50. Lai, M.C., Lombardo, M.V. & Baron-Cohen, S. Autism. Lancet 383, 896–910 (2014).

    PubMed  Google Scholar 

  51. Jenkins, D. et al. Analysis of TSHZ2 and TSHZ3 genes in congenital pelvi-ureteric junction obstruction. Nephrol. Dial. Transplant. 25, 54–60 (2010).

    CAS  PubMed  Google Scholar 

  52. Brandt, T. et al. Complex autism spectrum disorder in a patient with a 17q12 microduplication. Am. J. Med. Genet. A 158A, 1170–1177 (2012).

    PubMed  Google Scholar 

  53. Handrigan, G.R. et al. Deletions in 16q24.2 are associated with autism spectrum disorder, intellectual disability and congenital renal malformation. J. Med. Genet. 50, 163–173 (2013).

    CAS  PubMed  Google Scholar 

  54. Bodria, M. & Sanna-Cherchi, S. Genetic basis of congenital anomalies of the kidney and urinary tract. G. Ital. Nefrol. 32 (Suppl. 64) (2015).

  55. Mefford, H., Mitchell, E. & Hodge, J. 17q12 recurrent duplication. GeneReviews http://www.ncbi.nlm.nih.gov/books/NBK344340/ (2016).

  56. O'Leary, D.D. & Koester, S.E. Development of projection neuron types, axon pathways, and patterned connections of the mammalian cortex. Neuron 10, 991–1006 (1993).

    CAS  PubMed  Google Scholar 

  57. Delmonte, S., Gallagher, L., O'Hanlon, E., McGrath, J. & Balsters, J.H. Functional and structural connectivity of frontostriatal circuitry in autism spectrum disorder. Front. Hum. Neurosci. 7, 430 (2013).

    PubMed  PubMed Central  Google Scholar 

  58. Peça, J. et al. Shank3 mutant mice display autistic-like behaviours and striatal dysfunction. Nature 472, 437–442 (2011).

    PubMed  PubMed Central  Google Scholar 

  59. Blundell, J. et al. Neuroligin-1 deletion results in impaired spatial memory and increased repetitive behavior. J. Neurosci. 30, 2115–2129 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  60. Portmann, T. et al. Behavioral abnormalities and circuit defects in the basal ganglia of a mouse model of 16p11.2 deletion syndrome. Cell Rep. 7, 1077–1092 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  61. Schuurs-Hoeijmakers, J.H. et al. Refining the critical region of the novel 19q13.11 microdeletion syndrome to 750 Kb. J. Med. Genet. 46, 421–423 (2009).

    CAS  PubMed  Google Scholar 

  62. Gana, S. et al. 19q13.11 cryptic deletion: description of two new cases and indication for a role of WTIP haploinsufficiency in hypospadias. Eur. J. Hum. Genet. 20, 852–856 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  63. Forzano, F. et al. 19q13 microdeletion syndrome: further refining the critical region. Eur. J. Med. Genet. 55, 429–432 (2012).

    PubMed  Google Scholar 

  64. Venegas-Vega, C. et al. 19q13.11 microdeletion concomitant with ins(2;19)(p25.3;q13.1q13.4)dn in a boy: potential role of UBA2 in the associated phenotype. Mol. Cytogenet. 7, 61 (2014).

    PubMed  PubMed Central  Google Scholar 

  65. Paxinos, G. & Franklin, K.B.J. The Mouse Brain in Stereotaxic Coordinates 2nd edn. (Academic Press, 2001).

  66. Kim, D. et al. TopHat2: accurate alignment of transcriptomes in the presence of insertions, deletions and gene fusions. Genome Biol. 14, R36 (2013).

    PubMed  PubMed Central  Google Scholar 

  67. Langmead, B. & Salzberg, S.L. Fast gapped-read alignment with Bowtie 2. Nat. Methods 9, 357–359 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  68. Banerjee-Basu, S. & Packer, A. SFARI gene: an evolving database for the autism research community. Dis. Model. Mech. 3, 133–135 (2010).

    PubMed  Google Scholar 

  69. Chassain, C. et al. Metabolic, synaptic and behavioral impact of 5-week chronic deep brain stimulation in hemiparkinsonian rats. J. Neurochem. 136, 1004–1016 (2016).

    CAS  PubMed  Google Scholar 

  70. Jiang, Z.G. & North, R.A. Membrane properties and synaptic responses of rat striatal neurones in vitro. J. Physiol. (Lond.) 443, 533–553 (1991).

    CAS  Google Scholar 

  71. Beurrier, C. et al. Ciliary neurotrophic factor protects striatal neurons against excitotoxicity by enhancing glial glutamate uptake. PLoS One 5, e8550 (2010).

    PubMed  PubMed Central  Google Scholar 

  72. Thomas, A. et al. Marble burying reflects a repetitive and perseverative behavior more than novelty-induced anxiety. Psychopharmacology (Berl.) 204, 361–373 (2009).

    CAS  Google Scholar 

  73. Ehret, G. & Romand, R. Development of tone response thresholds, latencies and tuning in the mouse inferior colliculus. Brain Res. Dev. Brain Res. 67, 317–326 (1992).

    CAS  PubMed  Google Scholar 

  74. Willott, J.F. The Auditory Psychobiology of the Mouse in The Auditory Psychobiology of the Mouse 305–340 (Charles C Thomas Publishers, 1983).

  75. Yang, M. & Crawley, J.N. Simple behavioral assessment of mouse olfaction. Curr. Protoc. Neurosci. Ch. 8, Unit 8.24 (2009).

Download references

Acknowledgements

We thank M. Galdi, V. Vanoosten and C. Scajola who assisted with behavioral testing. This work was supported by funding from CNRS and Aix-Marseille University to L.K.-L.G., L.F. and M.C.; INSERM and Aix-Marseille University to P.L.R.; Fédération pour la Recherche sur le Cerveau (FRC) to L.F.; National Institutes of Health grants MH103339, MH106934 and MH106874, the Simons Foundation and the Kavli Foundation to N.S.; and the Medical Research Council (MR/L002744/1), the Manchester Biomedical Research Centre and the NIHR Greater Manchester Clinical Research Network to A.S.W. Funding for A.N.G. was provided by a grant from the German Federal Ministry for Education and Research (BMBF, NGFN-plus, 'Alzheimer Disease Integrative Genomics', PNA-01GS08127-3a). Microscopy was performed at the imaging platform of IBDM, supported by the French National Research Agency through the 'Investments for the Future' program (France-BioImaging, ANR-10-INSB-04-01).

Author information

Authors and Affiliations

Authors

Contributions

X.C., P.G., P.L.R., L.H.-A., N.S., A.N.G., L.K.-L.G. and L.F. designed the study. X.C., P.G., P.L.R., J.A., A.N.G., B.J., M.M., L.H.-A., K.Y.K., P.S. and Y.Z. performed experiments. J.A., A.L., E.R., M.S., C.V.-D., J.-M.C., M.-P.L., F.A., B.D., J.-F.L., A.S.W. and D.B. contributed clinical samples and clinical data. A.F., X.C. and L.F. prepared RNA samples, A.F. performed qRT-PCR, and D.S. and E.D. produced RNA–seq data and performed bioinformatics analysis of them (MGX-Montpellier GenomiX). X.C., P.G., P.L.R., L.H.-A., P.L.R., M.C., A.N.G., B.J., K.Y.K., N.S., L.K.-L.G. and L.F. analyzed data. X.C., P.G., P.L.R., L.H.-A., N.S., A.N.G., A.S.W., L.K.-L.G. and L.F. wrote the manuscript.

Corresponding author

Correspondence to Laurent Fasano.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Integrated supplementary information

Supplementary Figure 1 Genes coexpressed with TSHZ3 in the human neocortex and associated human pathologies.

(a) Module of genes coexpressed with TSHZ3 in the human neocortex at mid-gestation. The TSHZ3 module contains 50 genes including TSHZ3. Blue ‘balls’ correspond to genes that have been associated with ASD. Larger font size is used for genes encoding transcription factors known to influence cortical projection neuron differentiation, which all have been associated with ASD. (b) Histogram showing the most represented human pathologies associated with genes present in the TSHZ3 coexpression module (sorted in order of decreasing representation). Each gene is scored on the basis of the number of relevant publications that associate it with a pathology. Scores were given as follows: 1, 1 publication; 2, 2 publications; 3, >2 publications. ASD, autism spectrum disorder; ALS, amyotrophic lateral sclerosis; ADHD, attention deficit hyperactivity disorder.

Supplementary Figure 2 Tshz3lacZ/lacZ mice show altered gene expression at E18.5.

(a) Variation in mRNA levels analyzed by qRT–PCR in the cortex of Tshz3lacZ/lacZ versus wild-type mice. (b) Detection by in situ hybridization of differentially expressed genes and qRT–PCR analyses in the cortex of Tshz3lacZ/lacZ versus wild-type mice. Sagittal brain sections at E18.5 are shown (rostral, left; dorsal, top). Fgf10 expression in L5, which is caudally restricted in wild-type mice, is increased in the rostro-caudal extent of the cortex in Tshz3lacZ/lacZ mutant. The caudal expression of Ngfr in the subplate and L6 is strongly decreased in the mutant. The caudal expression of Col5a1 in the suplate is strongly decreased. Igfbp3 expression is strongly decreased caudally in the subplate and L6. (c) Tshz3 expression in a wild-type sagittal section. Data in a and b are shown as means ± s.e.m. (n = 3). *P < 0.05, **P < 0.02, unpaired two-tailed t test). For the large views in b and c, scale bar = 1 mm; for close-up views, scale bar = 100 μm.

Supplementary Figure 3 Membrane properties of striatal MSNs are similar in wild-type and Tshz3lacZ/+ mice.

(ae) Resting membrane potential (RMP; P = 0.069, Student’s t test) (a), action potential (AP) threshold (P = 0.746, Mann–Whitney test) (b), pattern of AP discharge (traces depict the responses of two MSNs to current steps of –100, +100 and +200 pA) (c), current–voltage relationship (d) and resulting input resistance (Ri) (e) (linear regression and slope comparison between wild-type and Tshz3lacZ/+ mice, F(1,127) = 1.486, P = 0.225). All data are expressed as means ± s.e.m.

Supplementary Figure 4 Visual, auditory and olfactory screening in Tshz3lacZ/+ and wild-type mice.

(ac) No significant differences were found between the two genotypes (n = 11 male mice per group) for visual performance (t < 1; d.f. = 20; P = 0.55) (a), auditory performance (t = 0.46; d.f. = 20; P = 0.65) (b) and olfactory exploration in the habituation/dishabituation test (c). Data are shown as means ± s.e.m. In c, time sniffing non-social (water, violet, vanilla) and social (B6, SWR) odors was analyzed with mixed ANOVA (genotype factor with two levels, Tshz3lacZ/+ and wild type, and 15 odors as repeated measures). The genotype factor was not significant (F < 1; d.f. = 1, 20).

Supplementary Figure 5 Sociability and preference in social novelty of Tshz3lacZ/+ male mice in the three-chamber apparatus.

(a) Time spent in the empty lateral compartments did not differ between the genotypes (F(1, 19) = 2.17; P = 0.16), but, with the effect size η2 being 0.10, we included the total activity in an ANCOVA. (b,c) Mean exploration times (±s.e.m.) that measured sociability (b) and preference for social novelty (c), after controlling for the effect of total activity. A double interaction between the genotype factor and the two condition factors (sociability and preference for social novelty) reached P = 0.005 (F(1, 19) = 10.08), with a large effect size (partial η2 = 0.35). We performed two partial ANOVAs to break out the double interaction. The first partial ANOVA showed an interaction with a large effect size (partial η2 = 0.31) between the genotype factor and the sociability factor (F(1, 19) = 8.51; P = 0.009). Tshz3lacZ/+ males did not explore the CD1 partner and empty box differently. Wild-type males explored the CD1 partner more than the empty box (dependent t(10) = 4.72; P < 0.001) with a large effect size of 0.76. The second partial ANOVA showed that the interaction was not significant between the genotype factor and the social novelty factor (F(1, 19) = 2.08; P = 0.16; partial η2 = 0.10). The two genotypes explored the new CD1 partner more than the already known CD1 partner. However, the difference was significant only for the wild-type male group (dependent t(10) = 3.08; P = 0.01) with an effect size of 0.61. n = 11 male mice per group. *P < 0.01, **P < 0.001. All data are expressed as means ± s.e.m.

Supplementary information

Supplementary Text and Figures

Supplementary Figures 1–5 and Supplementary Tables 2, 6, 7 and 8. (PDF 1301 kb)

Supplementary Table 1

Human brain and nervous system pathologies associated with genes present in the TSHZ3 module. (XLSX 43 kb)

Supplementary Table 3

DEX gene markers in E18.5 Tshz3lacZ/lacZ cortex. (XLSX 21 kb)

Supplementary Table 4

Analysis of pathways represented in and GO terms associated with DEX genes. (XLSX 84 kb)

Supplementary Table 5

Human brain and nervous system pathologies associated with orthologs of mouse Tshz3-regulated genes. (XLSX 57 kb)

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Caubit, X., Gubellini, P., Andrieux, J. et al. TSHZ3 deletion causes an autism syndrome and defects in cortical projection neurons. Nat Genet 48, 1359–1369 (2016). https://doi.org/10.1038/ng.3681

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/ng.3681

This article is cited by

Search

Quick links

Nature Briefing

Sign up for the Nature Briefing newsletter — what matters in science, free to your inbox daily.

Get the most important science stories of the day, free in your inbox. Sign up for Nature Briefing