A translational exploration of the effects of WNT2 variants on altered cortical structures in autism spectrum disorder

Yi-Ling Chien, Yu-Chieh Chen, Yen-Nan Chiu, Wen-Che Tsai, Susan Shur-Fen Gau, Yi-Ling Chien, Yu-Chieh Chen, Yen-Nan Chiu, Wen-Che Tsai, Susan Shur-Fen Gau

Abstract

Background: Evidence suggests that cortical anatomy may be aytpical in autism spectrum disorder. The wingless-type MMTV integration site family, member 2 (WNT2), a candidate gene for autism spectrum disorder, may regulate cortical development. However, it is unclear whether WNT2 variants are associated with altered cortical thickness in autism spectrum disorder.

Methods: In a sample of 118 people with autism spectrum disorder and 122 typically developing controls, we investigated cortical thickness using FreeSurfer software. We then examined the main effects of the WNT2 variants and the interactions of group × SNP and age × SNP for each hemisphere and brain region that was altered in people with autism spectrum disorder.

Results: Compared to neurotypical controls, people with autism spectrum disorder showed reduced mean cortical thickness in both hemispheres and 9 cortical regions after false discovery rate correction, including the right cingulate gyrus, the orbital gyrus, the insula, the inferior frontal gyrus (orbital part and triangular part), the lateral occipitotemporal gyrus, the posterior transverse collateral sulcus, the lateral sulcus and the superior temporal sulcus. In the full sample, 2 SNPs of WNT2 (rs6950765 and rs2896218) showed age × SNP interactions for the mean cortical thickness of both hemispheres, the middle-posterior cingulate cortex and the superior temporal cortex.

Limitations: We examined the genetic effect for each hemisphere and the 9 regions that were altered in autism spectrum disorder. The age effect we found in this cross-sectional study needs to be examined in longitudinal studies.

Conclusion: Based on neuroimaging and genetic data, our findings suggest that WNT2 variants might be associated with altered cortical thickness in autism spectrum disorder. Whether and how these WNT2 variants might involve cortical thinning requires further investigation.

Trial registration: ClinicalTrials.gov no. NCT01582256.

Protocol registration: National Institutes of Health no. NCT00494754.

Conflict of interest statement

Competing interests: None declared.

© 2021 CMA Joule Inc. or its licensors.

Figures

Figure 1
Figure 1
The 9 cortical regions identified with reduced cortical thickness, presented on (A) a standard brain cortex and (B) an inflated brain cortex.
Figure 2
Figure 2
The genotypes rs6950765 and rs2896218 were associated with mean cortical thickness in both hemispheres. (A) Genotype rs6950765 and the mean thickness of the left hemisphere. (B) Genotype rs6950765 and the mean thickness of the right hemisphere. (C) Genotype rs2896218 and the mean thickness of the left hemisphere. (D) Genotype rs2896218 and the mean thickness of the right hemisphere. The CG of rs6950765 and the AG of rs2896218 showed greater cortical thinning of both hemispheres with age.

References

    1. Nunes AS, Vakorin VA, Kozhemiako N, et al. . Atypical age-related changes in cortical thickness in autism spectrum disorder. Sci Rep 2020;10:11067.
    1. van Rooij D, Anagnostou E, Arango C, et al. . Cortical and subcortical brain morphometry differences between patients with autism spectrum disorder and healthy individuals across the lifespan: results from the ENIGMA ASD Working Group. Am J Psychiatry 2018;175:359–69.
    1. Baribeau DA, Anagnostou E. A comparison of neuroimaging findings in childhood onset schizophrenia and autism spectrum disorder: a review of the literature. Front Psychiatry 2013;4:175.
    1. Courchesne E, Karns CM, Davis HR, et al. . Unusual brain growth patterns in early life in patients with autistic disorder: an MRI study. Neurology 2001;57:245–54.
    1. Zielinski BA, Prigge MB, Nielsen JA, et al. . Longitudinal changes in cortical thickness in autism and typical development. Brain 2014;137:1799–812.
    1. Wallace GL, Dankner N, Kenworthy L, et al. . Age-related temporal and parietal cortical thinning in autism spectrum disorders. Brain 2010;133:3745–54.
    1. Yang DY, Beam D, Pelphrey KA, et al. . Cortical morphological markers in children with autism: a structural magnetic resonance imaging study of thickness, area, volume, and gyrification. Mol Autism 2016;7:11.
    1. Bedford SA, Park MTM, Devenyi GA, et al. . Large-scale analyses of the relationship between sex, age and intelligence quotient heterogeneity and cortical morphometry in autism spectrum disorder. Mol Psychiatry 2020;25:614–28.
    1. Chen CH, Fiecas M, Gutierrez ED, et al. . Genetic topography of brain morphology. Proc Natl Acad Sci U S A 2013;110:17089–94.
    1. Valk SL, Xu T, Margulies DS, et al. . Shaping brain structure: genetic and phylogenetic axes of macroscale organization of cortical thickness. Sci Adv 2020;6:eabb3417.
    1. Chen CH, Gutierrez ED, Thompson W, et al. . Hierarchical genetic organization of human cortical surface area. Science 2012;335:1634–6.
    1. Chen CH, Panizzon MS, Eyler LT, et al. . Genetic influences on cortical regionalization in the human brain. Neuron 2011;72:537–44.
    1. Fjell AM, Grydeland H, Krogsrud SK, et al. . Development and aging of cortical thickness correspond to genetic organization patterns. Proc Natl Acad Sci U S A 2015;112:15462–7.
    1. Udden J, Snijders TM, Fisher SE, et al. . A common variant of the CNTNAP2 gene is associated with structural variation in the left superior occipital gyrus. Brain Lang 2017;172:16–21.
    1. Zhu B, Chen C, Xue G, et al. . Associations between the CNTNAP2 gene, dorsolateral prefrontal cortex, and cognitive performance on the Stroop task. Neuroscience 2017;343:21–9.
    1. Clemm von Hohenberg C, Wigand MC, Kubicki M, et al. . CNTNAP2 polymorphisms and structural brain connectivity: a diffusion-tensor imaging study. J Psychiatr Res 2013;47:1349–56.
    1. Dennis EL, Jahanshad N, Rudie JD, et al. . Altered structural brain connectivity in healthy carriers of the autism risk gene, CNTNAP2. Brain Connect 2011;1:447–59.
    1. Rakic P. Defects of neuronal migration and the pathogenesis of cortical malformations. Prog Brain Res 1988;73:15–37.
    1. Huttenlocher PR. Morphometric study of human cerebral cortex development. Neuropsychologia 1990;28:517–27.
    1. Okerlund ND, Cheyette BN. Synaptic Wnt signaling—a contributor to major psychiatric disorders? J Neurodev Disord 2011;3:162–74.
    1. Steinecke A, Gampe C, Nitzsche F, et al. . Drosoph Inf ServC1 knockdown impairs the tangential migration of cortical interneurons by affecting the actin cytoskeleton. Front Cell Neurosci 2014;8:190.
    1. Hiester BG, Galati DF, Salinas PC, et al. . Neurotrophin and Wnt signaling cooperatively regulate dendritic spine formation. Mol Cell Neurosci 2013;56:115–27.
    1. Ishizuka K, Kamiya A, Oh EC, et al. . Drosoph Inf ServC1-dependent switch from progenitor proliferation to migration in the developing cortex. Nature 2011;473:92–6.
    1. Schneider T, Przewlocki R. Behavioral alterations in rats prenatally exposed to valproic acid: animal model of autism. Neuropsychopharmacology 2005;30:80–9.
    1. Wang Z, Xu L, Zhu X, et al. . Demethylation of specific Wnt/beta-catenin pathway genes and its upregulation in rat brain induced by prenatal valproate exposure. Anat Rec (Hoboken) 2010;293:1947–53.
    1. Chien YL, Wu YY, Chiu YN, et al. . Association study of the CNS patterning genes and autism in Han Chinese in Taiwan. Prog Neuropsychopharmacol Biol Psychiatry 2011;35:1512–7.
    1. Marui T, Funatogawa I, Koishi S, et al. . Association between autism and variants in the wingless-type MMTV integration site family member 2 (WNT2) gene. Int J Neuropsychopharmacol 2010;13:443–9.
    1. Wassink TH, Piven J, Vieland VJ, et al. . Evidence supporting WNT2 as an autism susceptibility gene. Am J Med Genet 2001;105:406–13.
    1. McCoy PA, Shao Y, Wolpert CM, et al. . No association between the WNT2 gene and autistic disorder. Am J Med Genet 2002;114:106–9.
    1. Chien YL, Wu SW, Chu CP, et al. . Attenuated contact heat-evoked potentials associated with sensory and social-emotional symptoms in individuals with autism spectrum disorder. Sci Rep 2017;7:36887.
    1. American Psychiatric Association. Diagnostic and statistical manual of mental disorders. Fifth edition.Arlington (VA): American Psychiatric Association Publishing; 2013.
    1. Lord C, Rutter M, Le Couteur A. Autism Diagnostic Interview — Revised: a revised version of a diagnostic interview for caregivers of individuals with possible pervasive developmental disorders. J Autism Dev Disord 1994;24:659–85.
    1. Chen YL, Shen LJ, Gau SS. The Mandarin version of the Kiddie-Schedule for Affective Disorders and Schizophrenia–Epidemiological version for DSM-5—a psychometric study. J Formos Med Assoc 2017;116:671–8.
    1. Chien WH, Wu YY, Gau SSF, et al. . Association study of the SLC25A12 gene and autism in Han Chinese in Taiwan. Prog Neuropsychopharmacol Biol Psychiatry 2010;34:189–92.
    1. Gau SS, Liu LT, Wu YY, et al. . Psychometric properties of the Chinese version of the social responsiveness scale. Res Autism Spectr Disord 2013;7:349–60.
    1. Xu H, Gregory SG, Hauser ER, et al. . SNPselector: a web tool for selecting SNPs for genetic association studies. Bioinformatics 2005; 21: 4181–6.
    1. Dale AM, Fischl B, Sereno MI. Cortical surface-based analysis. I. Segmentation and surface reconstruction. Neuroimage 1999;9:179–94.
    1. Fischl B. FreeSurfer. Neuroimage 2012;62:774–81.
    1. Fischl B, Salat DH, Busa E, et al. . Whole brain segmentation: automated labeling of neuroanatomical structures in the human brain. Neuron 2002;33:341–55.
    1. Fischl B, van der Kouwe A, Destrieux C, et al. . Automatically parcellating the human cerebral cortex. Cereb Cortex 2004;14:11–22.
    1. Rosen AFG, Roalf DR, Ruparel K, et al. . Quantitative assessment of structural image quality. Neuroimage 2018;169:407–18.
    1. Desikan RS, Segonne F, Fischl B, et al. . An automated labeling system for subdividing the human cerebral cortex on MRI scans into gyral based regions of interest. Neuroimage 2006;31:968–80.
    1. Fischl B, Dale AM. Measuring the thickness of the human cerebral cortex from magnetic resonance images. Proc Natl Acad Sci U S A 2000;97: 11050–5.
    1. Han X, Jovicich J, Salat D, et al. . Reliability of MRI-derived measurements of human cerebral cortical thickness: the effects of field strength, scanner upgrade and manufacturer. Neuroimage 2006;32: 180–94.
    1. Zhao K, Liu H, Yan R, et al. . Cortical thickness and subcortical structure volume abnormalities in patients with major depression with and without anxious symptoms. Brain Behav 2017;7:e00754.
    1. Stephen R, Liu Y, Ngandu T, et al. . Brain volumes and cortical thickness on MRI in the Finnish Geriatric Intervention Study to Prevent Cognitive Impairment and Disability (FINGER). Alzheimers Res Ther 2019;11:53.
    1. Sanchis-Segura C, Ibanez-Gual MV, Aguirre N, et al. . Author correction. Effects of different intracranial volume correction methods on univariate sex differences in grey matter volume and multivariate sex prediction. Sci Rep 2020;10:18937.
    1. Hardan AY, Muddasani S, Vemulapalli M, et al. . An MRI study of increased cortical thickness in autism. Am J Psychiatry 2006;163:1290–2.
    1. Hadjikhani N, Joseph RM, Snyder J, et al. . Anatomical differences in the mirror neuron system and social cognition network in autism. Cereb Cortex 2006;16:1276–82.
    1. Caria A, de Falco S. Anterior insular cortex regulation in autism spectrum disorders. Front Behav Neurosci 2015;9:38.
    1. Odriozola P, Uddin LQ, Lynch CJ, et al. . Insula response and connectivity during social and non-social attention in children with autism. Soc Cogn Affect Neurosci 2016;11:433–44.
    1. Wang Q, Zhang Z, Dong F, et al. . Anterior insula GABA levels correlate with emotional aspects of empathy: a proton magnetic resonance spectroscopy study. PLoS One 2014;9:e113845.
    1. Muller-Pinzler L, Krach S, Kramer UM, et al. . The social neuroscience of interpersonal emotions. Curr Top Behav Neurosci 2017;30:241–56.
    1. Mak-Fan KM, Taylor MJ, Roberts W, et al. . Measures of cortical grey matter structure and development in children with autism spectrum disorder. J Autism Dev Disord 2012;42:419–27.
    1. Raznahan A, Toro R, Daly E, et al. . Cortical anatomy in autism spectrum disorder: an in vivo MRI study on the effect of age. Cereb Cortex 2010;20:1332–40.
    1. Scheel C, Rotarska-Jagiela A, Schilbach L, et al. . Imaging derived cortical thickness reduction in high-functioning autism: key regions and temporal slope. Neuroimage 2011;58:391–400.
    1. Postema MC, van Rooij D, Anagnostou E, et al. . Altered structural brain asymmetry in autism spectrum disorder in a study of 54 datasets. Nat Commun 2019;10:4958.
    1. Lin PI, Chien YL, Wu YY, et al. . The WNT2 gene polymorphism associated with speech delay inherent to autism. Res Dev Disabil 2012;33:1533–40.
    1. Phillips JE, Corces VG. CTCF: master weaver of the genome. Cell 2009;137:1194–211.
    1. Ohlsson R, Renkawitz R, Lobanenkov V. CTCF is a uniquely versatile transcription regulator linked to epigenetics and disease. Trends Genet 2001;17:520–7.
    1. Bell AC, West AG, Felsenfeld G. Insulators and boundaries: versatile regulatory elements in the eukaryotic genome. Science 2001; 291: 447–50.
    1. West AG, Gaszner M, Felsenfeld G. Insulators: many functions, many mechanisms. Genes Dev 2002;16:271–88.
    1. Botta M, Haider S, Leung IX, et al. . Intra- and inter-chromosomal interactions correlate with CTCF binding genome wide. Mol Syst Biol 2010;6:426.
    1. Dixon JR, Selvaraj S, Yue F, et al. . Topological domains in Mamm genomes identified by analysis of chromatin interactions. Nature 2012;485:376–80.
    1. Lieberman-Aiden E, van Berkum NL, Williams L, et al. . Comprehensive mapping of long-range interactions reveals folding principles of the human genome. Science 2009;326:289–93.
    1. Swafford D, Manicassamy S. Wnt signaling in dendritic cells: its role in regulation of immunity and tolerance. Discov Med 2015;19:303–10.
    1. Gatica-Andrades M, Vagenas D, Kling J, et al. . WNT ligands contribute to the immune response during septic shock and amplify endotoxemia-driven inflammation in mice. Blood Adv 2017;1:1274–86.

Source: PubMed

3
Subscribe