CHD8 and Autism Spectrum Disorder: Comparison
Please note this is a comparison between Version 2 by Conner Chen and Version 1 by Dietmar Spengler.

Autism spectrum disorder (ASD) encompasses a spectrum of early-onset neurodevelopmental disorders with an estimated prevalence of ~1.5% in developed countries. Patients present early deficits in social interaction and communication, repetitive patterns of behavior, and restricted interests and activities.Chromodomain helicase domain 8 (CHD8) is one of the most frequently mutated and most penetrant genes in the autism spectrum disorder (ASD). Individuals with CHD8 mutations show leading symptoms of autism, macrocephaly, and facial dysmorphisms. The molecular and cellular mechanisms underpinning the early onset and development of these symptoms are still poorly understood and prevent timely and more efficient therapies of patients.

  • ASD
  • CHD8
  • single-cell sequencing
  • excitatory/inhibitory imbalance
Please wait, diff process is still running!

References

  1. Hoffmann, A.; Ziller, M.; Spengler, D. Focus on Causality in ESC/iPSC-Based Modeling of Psychiatric Disorders. Cells 2020, 9, 366.
  2. Wade, A.A.; Lim, K.; Catta-Preta, R.; Nord, A.S. Common CHD8 Genomic Targets Contrast With Model-Specific Transcriptional Impacts of CHD8 Haploinsufficiency. Front. Mol. Neurosci. 2018, 11, 481.
  3. Jaenisch, R.; Bird, A. Epigenetic regulation of gene expression: How the genome integrates intrinsic and environmental signals. Nat. Genet. 2003, 33, 245–254.
  4. Tyagi, M.; Imam, N.; Verma, K.; Patel, A.K. Chromatin remodelers: We are the drivers! Nucleus 2016, 7, 388–404.
  5. Murgatroyd, C.; Spengler, D. Genetic variation in the epigenetic machinery and mental health. Curr. Psychiatry Rep. 2012, 14, 138–149.
  6. Krumm, N.; O’Roak, B.J.; Shendure, J.; Eichler, E.E. A de novo convergence of autism genetics and molecular neuroscience. Trends Neurosci. 2014, 37, 95–105.
  7. Clapier, C.R.; Cairns, B.R. The biology of chromatin remodeling complexes. Annu. Rev. Biochem. 2009, 78, 273–304.
  8. Singleton, M.R.; Dillingham, M.S.; Wigley, D.B. Structure and mechanism of helicases and nucleic acid translocases. Annu. Rev. Biochem. 2007, 76, 23–50.
  9. Bernier, R.; Golzio, C.; Xiong, B.; Stessman, H.A.; Coe, B.P.; Penn, O.; Witherspoon, K.; Gerdts, J.; Baker, C.; Vulto-van Silfhout, A.T.; et al. Disruptive CHD8 mutations define a subtype of autism early in development. Cell 2014, 158, 263–276.
  10. Barnard, R.A.; Pomaville, M.B.; O’Roak, B.J. Mutations and Modeling of the Chromatin Remodeler CHD8 Define an Emerging Autism Etiology. Front. Neurosci. 2015, 9, 477.
  11. Hu, Y.; Lai, Y.; Zhu, D. Transcription regulation by CHD proteins to control plant development. Front. Plant Sci. 2014, 5, 223.
  12. Hoffmann, A.; Spengler, D. Chromatin Remodeling Complex NuRD in Neurodevelopment and Neurodevelopmental Disorders. Front. Genet. 2019, 10, 682.
  13. Iossifov, I.; Ronemus, M.; Levy, D.; Wang, Z.; Hakker, I.; Rosenbaum, J.; Yamrom, B.; Lee, Y.-H.; Narzisi, G.; Leotta, A.; et al. De novo gene disruptions in children on the autistic spectrum. Neuron 2012, 74, 285–299.
  14. Neale, B.M.; Kou, Y.; Liu, L.; Ma’ayan, A.; Samocha, K.E.; Sabo, A.; Lin, C.-F.; Stevens, C.; Wang, L.-S.; Makarov, V.; et al. Patterns and rates of exonic de novo mutations in autism spectrum disorders. Nature 2012, 485, 242–245.
  15. Gaugler, T.; Klei, L.; Sanders, S.J.; Bodea, C.A.; Goldberg, A.P.; Lee, A.B.; Mahajan, M.; Manaa, D.; Pawitan, Y.; Reichert, J.; et al. Most genetic risk for autism resides with common variation. Nat. Genet. 2014, 46, 881–885.
  16. Grove, J.; Ripke, S.; Als, T.D.; Mattheisen, M.; Walters, R.K.; Won, H.; Pallesen, J.; Agerbo, E.; Andreassen, O.A.; Anney, R.; et al. Identification of common genetic risk variants for autism spectrum disorder. Nat. Genet. 2019, 51, 431–444.
  17. Satterstrom, F.K.; Kosmicki, J.A.; Wang, J.; Breen, M.S.; De Rubeis, S.; An, J.-Y.; Peng, M.; Collins, R.; Grove, J.; Klei, L.; et al. Large-Scale Exome Sequencing Study Implicates Both Developmental and Functional Changes in the Neurobiology of Autism. Cell 2020, 180, 568–584.e23.
  18. O’Roak, B.J.; Vives, L.; Girirajan, S.; Karakoc, E.; Krumm, N.; Coe, B.P.; Levy, R.; Ko, A.; Lee, C.; Smith, J.D.; et al. Sporadic autism exomes reveal a highly interconnected protein network of de novo mutations. Nature 2012, 485, 246–250.
  19. Sanders, S.J.; Murtha, M.T.; Gupta, A.R.; Murdoch, J.D.; Raubeson, M.J.; Willsey, A.J.; Ercan-Sencicek, A.G.; DiLullo, N.M.; Parikshak, N.N.; Stein, J.L.; et al. De novo mutations revealed by whole-exome sequencing are strongly associated with autism. Nature 2012, 485, 237–241.
  20. O’Roak, B.J.; Stessman, H.A.; Boyle, E.A.; Witherspoon, K.T.; Martin, B.; Lee, C.; Vives, L.; Baker, C.; Hiatt, J.B.; Nickerson, D.A.; et al. Recurrent de novo mutations implicate novel genes underlying simplex autism risk. Nat. Commun. 2014, 5, 5595.
  21. Wang, T.; Guo, H.; Xiong, B.; Stessman, H.A.F.; Wu, H.; Coe, B.P.; Turner, T.N.; Liu, Y.; Zhao, W.; Hoekzema, K.; et al. De novo genic mutations among a Chinese autism spectrum disorder cohort. Nat. Commun. 2016, 7, 13316.
  22. Stessman, H.A.F.; Xiong, B.; Coe, B.P.; Wang, T.; Hoekzema, K.; Fenckova, M.; Kvarnung, M.; Gerdts, J.; Trinh, S.; Cosemans, N.; et al. Targeted sequencing identifies 91 neurodevelopmental-disorder risk genes with autism and developmental-disability biases. Nat. Genet. 2017, 49, 515–526.
  23. Talkowski, M.E.; Rosenfeld, J.A.; Blumenthal, I.; Pillalamarri, V.; Chiang, C.; Heilbut, A.; Ernst, C.; Hanscom, C.; Rossin, E.; Lindgren, A.M.; et al. Sequencing chromosomal abnormalities reveals neurodevelopmental loci that confer risk across diagnostic boundaries. Cell 2012, 149, 525–537.
  24. Yasin, H.; Gibson, W.T.; Langlois, S.; Stowe, R.M.; Tsang, E.S.; Lee, L.; Poon, J.; Tran, G.; Tyson, C.; Wong, C.K.; et al. A distinct neurodevelopmental syndrome with intellectual disability, autism spectrum disorder, characteristic facies, and macrocephaly is caused by defects in CHD8. J. Hum. Genet. 2019, 64, 271–280.
  25. An, Y.; Zhang, L.; Liu, W.; Jiang, Y.; Chen, X.; Lan, X.; Li, G.; Hang, Q.; Wang, J.; Gusella, J.F.; et al. De novo variants in the Helicase-C domain of CHD8 are associated with severe phenotypes including autism, language disability and overgrowth. Hum. Genet. 2020, 139, 499–512.
  26. Beighley, J.S.; Hudac, C.M.; Arnett, A.B.; Peterson, J.L.; Gerdts, J.; Wallace, A.S.; Mefford, H.C.; Hoekzema, K.; Turner, T.N.; O’Roak, B.J.; et al. Clinical Phenotypes of Carriers of Mutations in CHD8 or Its Conserved Target Genes. Biol. Psychiatry 2020, 87, 123–131.
  27. Ostrowski, P.J.; Zachariou, A.; Loveday, C.; Beleza-Meireles, A.; Bertoli, M.; Dean, J.; Douglas, A.G.L.; Ellis, I.; Foster, A.; Graham, J.M.; et al. The CHD8 overgrowth syndrome: A detailed evaluation of an emerging overgrowth phenotype in 27 patients. Am. J. Med. Genet. C Semin. Med. Genet. 2019, 181, 557–564.
  28. Wu, H.; Li, H.; Bai, T.; Han, L.; Ou, J.; Xun, G.; Zhang, Y.; Wang, Y.; Duan, G.; Zhao, N.; et al. Phenotype-to-genotype approach reveals head-circumference-associated genes in an autism spectrum disorder cohort. Clin. Genet. 2020, 97, 338–346.
  29. Sohal, V.S.; Zhang, F.; Yizhar, O.; Deisseroth, K. Parvalbumin neurons and gamma rhythms enhance cortical circuit performance. Nature 2009, 459, 698–702.
  30. Iascone, D.M.; Li, Y.; Sümbül, U.; Doron, M.; Chen, H.; Andreu, V.; Goudy, F.; Blockus, H.; Abbott, L.F.; Segev, I.; et al. Whole-Neuron Synaptic Mapping Reveals Spatially Precise Excitatory/Inhibitory Balance Limiting Dendritic and Somatic Spiking. Neuron 2020, 106, 566–578.e8.
  31. Kavalali, E.T.; Monteggia, L.M. Targeting Homeostatic Synaptic Plasticity for Treatment of Mood Disorders. Neuron 2020, 106, 715–726.
  32. Gao, R.; Penzes, P. Common mechanisms of excitatory and inhibitory imbalance in schizophrenia and autism spectrum disorders. Curr. Mol. Med. 2015, 15, 146–167.
  33. Lee, E.; Lee, J.; Kim, E. Excitation/Inhibition Imbalance in Animal Models of Autism Spectrum Disorders. Biol. Psychiatry 2017, 81, 838–847.
  34. Oliveira, B.; Mitjans, M.; Nitsche, M.A.; Kuo, M.-F.; Ehrenreich, H. Excitation-inhibition dysbalance as predictor of autistic phenotypes. J. Psychiatr. Res. 2018, 104, 96–99.
  35. Port, R.G.; Oberman, L.M.; Roberts, T.P. Revisiting the excitation/inhibition imbalance hypothesis of ASD through a clinical lens. Br. J. Radiol. 2019, 92, 20180944.
  36. Edgar, J.C.; Fisk Iv, C.L.; Berman, J.I.; Chudnovskaya, D.; Liu, S.; Pandey, J.; Herrington, J.D.; Port, R.G.; Schultz, R.T.; Roberts, T.P.L. Auditory encoding abnormalities in children with autism spectrum disorder suggest delayed development of auditory cortex. Mol. Autism 2015, 6, 69.
  37. Port, R.G.; Edgar, J.C.; Ku, M.; Bloy, L.; Murray, R.; Blaskey, L.; Levy, S.E.; Roberts, T.P.L. Maturation of auditory neural processes in autism spectrum disorder—A longitudinal MEG study. Neuroimage Clin. 2016, 11, 566–577.
  38. Brown, M.S.; Singel, D.; Hepburn, S.; Rojas, D.C. Increased glutamate concentration in the auditory cortex of persons with autism and first-degree relatives: A (1)H-MRS study. Autism Res. 2013, 6, 1–10.
  39. Horder, J.; Petrinovic, M.M.; Mendez, M.A.; Bruns, A.; Takumi, T.; Spooren, W.; Barker, G.J.; Künnecke, B.; Murphy, D.G. Glutamate and GABA in autism spectrum disorder-a translational magnetic resonance spectroscopy study in man and rodent models. Transl. Psychiatry 2018, 8, 106.
  40. Kubas, B.; Kułak, W.; Sobaniec, W.; Tarasow, E.; Lebkowska, U.; Walecki, J. Metabolite alterations in autistic children: A 1H MR spectroscopy study. Adv. Med. Sci. 2012, 57, 152–156.
  41. Mescher, M.; Merkle, H.; Kirsch, J.; Garwood, M.; Gruetter, R. Simultaneous in vivo spectral editing and water suppression. NMR Biomed. 1998, 11, 266–272.
  42. Parikshak, N.N.; Luo, R.; Zhang, A.; Won, H.; Lowe, J.K.; Chandran, V.; Horvath, S.; Geschwind, D.H. Integrative functional genomic analyses implicate specific molecular pathways and circuits in autism. Cell 2013, 155, 1008–1021.
  43. Willsey, A.J.; Sanders, S.J.; Li, M.; Dong, S.; Tebbenkamp, A.T.; Muhle, R.A.; Reilly, S.K.; Lin, L.; Fertuzinhos, S.; Miller, J.A.; et al. Coexpression networks implicate human midfetal deep cortical projection neurons in the pathogenesis of autism. Cell 2013, 155, 997–1007.
  44. Bogdan, R.; Salmeron, B.J.; Carey, C.E.; Agrawal, A.; Calhoun, V.D.; Garavan, H.; Hariri, A.R.; Heinz, A.; Hill, M.N.; Holmes, A.; et al. Imaging Genetics and Genomics in Psychiatry: A Critical Review of Progress and Potential. Biol. Psychiatry 2017, 82, 165–175.
  45. Berto, S.; Treacher, A.; Caglayan, E.; Luo, D.; Haney, J.R.; Gandal, M.J.; Geschwind, D.H.; Montillo, A.; Konopka, G. Association between resting-state functional brain connectivity and gene expression is altered in autism spectrum disorder. medRxiv 2021.
  46. Platt, R.J.; Zhou, Y.; Slaymaker, I.M.; Shetty, A.S.; Weisbach, N.R.; Kim, J.-A.; Sharma, J.; Desai, M.; Sood, S.; Kempton, H.R.; et al. Chd8 mutation leads to autistic-like behaviors and impaired striatal circuits. Cell Rep. 2017, 19, 335–350.
  47. Jung, H.; Park, H.; Choi, Y.; Kang, H.; Lee, E.; Kweon, H.; Roh, J.D.; Ellegood, J.; Choi, W.; Kang, J.; et al. Sexually dimorphic behavior, neuronal activity, and gene expression in Chd8-mutant mice. Nat. Neurosci. 2018, 21, 1218–1228.
  48. Ellingford, R.A.; de Meritens, E.R.; Shaunak, R.; Naybour, L.; Basson, M.A.; Andreae, L.C. Cell-type-specific synaptic imbalance and disrupted homeostatic plasticity in cortical circuits of ASD-associated Chd8 haploinsufficient mice. bioRxiv 2020.
  49. Armand, E.J.; Li, J.; Xie, F.; Luo, C.; Mukamel, E.A. Single-Cell Sequencing of Brain Cell Transcriptomes and Epigenomes. Neuron 2021, 109, 11–26.
  50. Jin, X.; Simmons, S.K.; Guo, A.; Shetty, A.S.; Ko, M.; Nguyen, L.; Jokhi, V.; Robinson, E.; Oyler, P.; Curry, N.; et al. In vivo Perturb-Seq reveals neuronal and glial abnormalities associated with autism risk genes. Science 2020, 370, eaaz6063.
  51. Marie, C.; Clavairoly, A.; Frah, M.; Hmidan, H.; Yan, J.; Zhao, C.; Van Steenwinckel, J.; Daveau, R.; Zalc, B.; Hassan, B.; et al. Oligodendrocyte precursor survival and differentiation requires chromatin remodeling by Chd7 and Chd8. Proc. Natl. Acad. Sci. USA 2018, 115, E8246–E8255.
  52. Zhao, C.; Dong, C.; Frah, M.; Deng, Y.; Marie, C.; Zhang, F.; Xu, L.; Ma, Z.; Dong, X.; Lin, Y.; et al. Dual Requirement of CHD8 for Chromatin Landscape Establishment and Histone Methyltransferase Recruitment to Promote CNS Myelination and Repair. Dev. Cell 2018, 45, 753–768.e8.
  53. Hardan, A.Y.; Fung, L.K.; Frazier, T.; Berquist, S.W.; Minshew, N.J.; Keshavan, M.S.; Stanley, J.A. A proton spectroscopy study of white matter in children with autism. Prog. Neuropsychopharmacol. Biol. Psychiatry 2016, 66, 48–53.
  54. Deoni, S.; Dean, D.; Joelson, S.; O’Regan, J.; Schneider, N. Early nutrition influences developmental myelination and cognition in infants and young children. Neuroimage 2018, 178, 649–659.
  55. Douzgou, S.; Liang, H.W.; Metcalfe, K.; Somarathi, S.; Tischkowitz, M.; Mohamed, W.; Kini, U.; McKee, S.; Yates, L.; Bertoli, M.; et al. The clinical presentation caused by truncating CHD8 variants. Clin. Genet. 2019, 96, 72–84.
  56. Ardhanareeswaran, K.; Mariani, J.; Coppola, G.; Abyzov, A.; Vaccarino, F.M. Human induced pluripotent stem cells for modelling neurodevelopmental disorders. Nat. Rev. Neurol. 2017, 13, 265–278.
  57. Gollo, L.L.; Roberts, J.A.; Cropley, V.L.; Di Biase, M.A.; Pantelis, C.; Zalesky, A.; Breakspear, M. Fragility and volatility of structural hubs in the human connectome. Nat. Neurosci. 2018, 21, 1107–1116.
  58. Chiaradia, I.; Lancaster, M.A. Brain organoids for the study of human neurobiology at the interface of in vitro and in vivo. Nat. Neurosci. 2020, 23, 1496–1508.
  59. Tanaka, Y.; Cakir, B.; Xiang, Y.; Sullivan, G.J.; Park, I.-H. Synthetic Analyses of Single-Cell Transcriptomes from Multiple Brain Organoids and Fetal Brain. Cell Rep. 2020, 30, 1682–1689.e3.
  60. Quadrato, G.; Nguyen, T.; Macosko, E.Z.; Sherwood, J.L.; Min Yang, S.; Berger, D.R.; Maria, N.; Scholvin, J.; Goldman, M.; Kinney, J.P.; et al. Cell diversity and network dynamics in photosensitive human brain organoids. Nature 2017, 545, 48–53.
  61. Qian, X.; Su, Y.; Adam, C.D.; Deutschmann, A.U.; Pather, S.R.; Goldberg, E.M.; Su, K.; Li, S.; Lu, L.; Jacob, F.; et al. Sliced Human Cortical Organoids for Modeling Distinct Cortical Layer Formation. Cell Stem Cell 2020, 26, 766–781.e9.
  62. Velasco, S.; Kedaigle, A.J.; Simmons, S.K.; Nash, A.; Rocha, M.; Quadrato, G.; Paulsen, B.; Nguyen, L.; Adiconis, X.; Regev, A.; et al. Individual brain organoids reproducibly form cell diversity of the human cerebral cortex. Nature 2019, 570, 523–527.
  63. Wang, P.; Mokhtari, R.; Pedrosa, E.; Kirschenbaum, M.; Bayrak, C.; Zheng, D.; Lachman, H.M. CRISPR/Cas9-mediated heterozygous knockout of the autism gene CHD8 and characterization of its transcriptional networks in cerebral organoids derived from iPS cells. Mol. Autism 2017, 8, 11.
  64. Wang, P.; Lin, M.; Pedrosa, E.; Hrabovsky, A.; Zhang, Z.; Guo, W.; Lachman, H.M.; Zheng, D. CRISPR/Cas9-mediated heterozygous knockout of the autism gene CHD8 and characterization of its transcriptional networks in neurodevelopment. Mol. Autism 2015, 6, 55.
  65. Hoffmann, A.; Sportelli, V.; Ziller, M.; Spengler, D. Switch-Like Roles for Polycomb Proteins from Neurodevelopment to Neurodegeneration. Epigenomes 2017, 1, 21.
  66. Kraus, P.; Lufkin, T. Dlx homeobox gene control of mammalian limb and craniofacial development. Am. J. Med. Genet. A 2006, 140, 1366–1374.
  67. Feng, J.; Bi, C.; Clark, B.S.; Mady, R.; Shah, P.; Kohtz, J.D. The Evf-2 noncoding RNA is transcribed from the Dlx-5/6 ultraconserved region and functions as a Dlx-2 transcriptional coactivator. Genes Dev. 2006, 20, 1470–1484.
  68. Paina, S.; Garzotto, D.; DeMarchis, S.; Marino, M.; Moiana, A.; Conti, L.; Cattaneo, E.; Perera, M.; Corte, G.; Calautti, E.; et al. Wnt5a is a transcriptional target of Dlx homeogenes and promotes differentiation of interneuron progenitors in vitro and in vivo. J. Neurosci. 2011, 31, 2675–2687.
  69. Poitras, L.; Yu, M.; Lesage-Pelletier, C.; Macdonald, R.B.; Gagné, J.-P.; Hatch, G.; Kelly, I.; Hamilton, S.P.; Rubenstein, J.L.R.; Poirier, G.G.; et al. An SNP in an ultraconserved regulatory element affects Dlx5/Dlx6 regulation in the forebrain. Development 2010, 137, 3089–3097.
  70. Mariani, J.; Coppola, G.; Zhang, P.; Abyzov, A.; Provini, L.; Tomasini, L.; Amenduni, M.; Szekely, A.; Palejev, D.; Wilson, M.; et al. FOXG1-Dependent Dysregulation of GABA/Glutamate Neuron Differentiation in Autism Spectrum Disorders. Cell 2015, 162, 375–390.
  71. Villa, C.E.; Cheroni, C.; López-Tóbon, A.; Dotter, C.P.; Oliveira, B.; Sacco, R.; Yahya, A.C.; Morandell, J.; Gabriele, M.; Sommer, C.; et al. CHD8 haploinsufficiency alters the developmental trajectories of human excitatory and inhibitory neurons linking autism phenotypes with transient cellular defects. bioRxiv 2020.
  72. Hoffmann, A.; Spengler, D. Chromatin Remodeler CHD8 in Autism and Brain Development. J. Clin. Med. 2021, 10, 366.
  73. Benito-Kwiecinski, S.; Giandomenico, S.L.; Sutcliffe, M.; Riis, E.S.; Freire-Pritchett, P.; Kelava, I.; Wunderlich, S.; Martin, U.; Wray, G.; Lancaster, M.A. An early cell shape transition drives evolutionary expansion of the human forebrain. bioRxiv 2020.
More
Video Production Service