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.

  • Expert Review
  • Published:

Neurobiology of ARID1B haploinsufficiency related to neurodevelopmental and psychiatric disorders

Abstract

ARID1B haploinsufficiency is a frequent cause of intellectual disability (ID) and autism spectrum disorder (ASD), and also leads to emotional disturbances. In this review, we examine past and present clinical and preclinical research into the neurobiological function of ARID1B. The presentation of ARID1B-related disorders (ARID1B-RD) is highly heterogeneous, including varying degrees of ID, ASD, and physical features. Recent research includes the development of suitable clinical readiness assessments for the treatment of ARID1B-RD, as well as similar neurodevelopmental disorders. Recently developed mouse models of Arid1b haploinsufficiency successfully mirror many of the behavioral phenotypes of ASD and ID. These animal models have helped to solidify the molecular mechanisms by which ARID1B regulates brain development and function, including epigenetic regulation of the Pvalb gene and promotion of Wnt/β-catenin signaling in neural progenitors in the ventral telencephalon. Finally, preclinical studies have identified the use of a positive allosteric modulator of the GABAA receptor as an effective treatment for some Arid1b haploinsufficiency-related behavioral phenotypes, and there is potential for the refinement of this therapy in order to translate it into clinical use.

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

Fig. 1: Abnormal epigenetics and gene expression related to inhibitory neurons.
Fig. 2: Abnormal epigenetics and gene expression related to inhibitory neurons.

Similar content being viewed by others

References

  1. Backx L, Seuntjens E, Devriendt K, Vermeesch J, Van, Esch H. A balanced translocation t(6;14)(q25.3;q13.2) leading to reciprocal fusion transcripts in a patient with intellectual disability and agenesis of corpus callosum. Cytogenet Genome Res. 2011;132:135–43.

    Article  CAS  PubMed  Google Scholar 

  2. Halgren C, Kjaergaard S, Bak M, Hansen C, El-Schich Z, Anderson CM, et al. Corpus callosum abnormalities, intellectual disability, speech impairment, and autism in patients with haploinsufficiency of ARID1B. Clin Genet. 2012;82:248–55.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. Hoyer J, Ekici AB, Endele S, Popp B, Zweier C, Wiesener A, et al. Haploinsufficiency of ARID1B, a member of the SWI/SNF-a chromatin-remodeling complex, is a frequent cause of intellectual disability. Am J Hum Genet. 2012;90:565–72.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. Santen GW, Aten E, Sun Y, Almomani R, Gilissen C, Nielsen M, et al. Mutations in SWI/SNF chromatin remodeling complex gene ARID1B cause Coffin-Siris syndrome. Nat Genet. 2012;44:379–80.

    Article  CAS  PubMed  Google Scholar 

  5. Tsurusaki Y, Okamoto N, Ohashi H, Kosho T, Imai Y, Hibi-Ko Y, et al. Mutations affecting components of the SWI/SNF complex cause Coffin-Siris syndrome. Nat Genet. 2012;44:376–8.

    Article  CAS  PubMed  Google Scholar 

  6. Sim JC, White SM, Fitzpatrick E, Wilson GR, Gillies G, Pope K, et al. Expanding the phenotypic spectrum of ARID1B-mediated disorders and identification of altered cell-cycle dynamics due to ARID1B haploinsufficiency. Orphanet J Rare Dis. 2014;9:43.

    Article  PubMed  PubMed Central  Google Scholar 

  7. Vasileiou G, Ekici AB, Uebe S, Zweier C, Hoyer J, Engels H, et al. Chromatin-remodeling-factor ARID1B represses Wnt/beta-catenin signaling. Am J Hum Genet. 2015;97:445–56.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Ka M, Chopra DA, Dravid SM, Kim WY. Essential roles for ARID1B in dendritic arborization and spine morphology of developing pyramidal neurons. J Neurosci. 2016;36:2723–42.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Jung EM, Moffat JJ, Liu J, Dravid SM, Gurumurthy CB, Kim WY. Arid1b haploinsufficiency disrupts cortical interneuron development and mouse behavior. Nat Neurosci. 2017;20:1694–707.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Yu D, Jiao X, Cao T, Huang F. Serum miRNA expression profiling reveals miR-486-3p may play a significant role in the development of autism by targeting ARID1B. Neuroreport. 2018;29:1431–6.

    Article  CAS  PubMed  Google Scholar 

  11. Demily C, Duwime C, Lopez C, Hemimou C, Poisson A, Plasse J, et al. Corpus callosum metrics predict severity of visuospatial and neuromotor dysfunctions in ARID1B mutations with Coffin-Siris syndrome. Psychiatr Genet. 2019;29:237–42.

    Article  CAS  PubMed  Google Scholar 

  12. Gorokhova S, Mortreux J, Afenjar A, Attie-Bitach T, Blanluet M, Cormier-Daire V, et al. Significant contribution of intragenic deletions to ARID1B mutation spectrum. Genet Med. 2019;21:2654–5.

    Article  PubMed  Google Scholar 

  13. van der Sluijs PJ, Jansen S, Vergano SA, Adachi-Fukuda M, Alanay Y, AlKindy A, et al. The ARID1B spectrum in 143 patients: from nonsyndromic intellectual disability to Coffin-Siris syndrome. Genet Med. 2019;21:1295–307.

    Article  PubMed  Google Scholar 

  14. Boerstler T, Wend H, Krumbiegel M, Kavyanifar A, Reis A, Lie DC, et al. CRISPR/Cas9 mediated generation of human ARID1B heterozygous knockout hESC lines to model Coffin-Siris syndrome. Stem Cell Res. 2020;47:101889.

    Article  CAS  PubMed  Google Scholar 

  15. Kruizinga MD, Zuiker R, Sali E, de Kam ML, Doll RJ, Groeneveld GJ et al. Finding suitable clinical endpoints for a potential treatment of a rare genetic disease: the case of ARID1B. Neurotherapeutics. 2020;17:1300–10.

  16. Liu X, Hu G, Ye J, Ye B, Shen N, Tao Y, et al. De Novo ARID1B mutations cause growth delay associated with aberrant Wnt/beta-catenin signaling. Hum Mutat. 2020;41:1012–24.

    Article  CAS  PubMed  Google Scholar 

  17. Nie Z, Yan Z, Chen EH, Sechi S, Ling C, Zhou S, et al. Novel SWI/SNF chromatin-remodeling complexes contain a mixed-lineage leukemia chromosomal translocation partner. Mol Cell Biol. 2003;23:2942–52.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Wang X, Nagl NG, Wilsker D, Van Scoy M, Pacchione S, Yaciuk P, et al. Two related ARID family proteins are alternative subunits of human SWI/SNF complexes. Biochem J. 2004;383:319–25.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Celen C, Chuang JC, Luo X, Nijem N, Walker AK, Chen F, et al. Arid1b haploinsufficient mice reveal neuropsychiatric phenotypes and reversible causes of growth impairment. Elife. 2017;6:e25730.

  20. Lee AW, Ventola P, Budimirovic D, Berry-Kravis E, Visootsak J. Clinical development of targeted fragile X syndrome treatments: an industry perspective. Brain Sci. 2018;8:214.

  21. Berry-Kravis E, Hagerman R, Visootsak J, Budimirovic D, Kaufmann WE, Cherubini M, et al. Arbaclofen in fragile X syndrome: results of phase 3 trials. J Neurodev Disord. 2017;9:3.

    Article  PubMed  PubMed Central  Google Scholar 

  22. Kapp SK, Gillespie-Lynch K, Sherman LE, Hutman T. Deficit, difference, or both? Autism and neurodiversity. Dev Psychol. 2013;49:59–71.

    Article  PubMed  Google Scholar 

  23. Pantazakos T. Treatment for whom? Towards a phenomenological resolution of controversy within autism treatment. Stud Hist Philos Biol Biomed Sci. 2019;77:101176.

    Article  PubMed  Google Scholar 

  24. Eady N, Courtenay K, Strydom A. Pharmacological management of behavioral and psychiatric symptoms in older adults with intellectual disability. Drugs Aging. 2015;32:95–102.

    Article  CAS  PubMed  Google Scholar 

  25. Ellison JW, Rosenfeld JA, Shaffer LG. Genetic basis of intellectual disability. Annu Rev Med. 2013;64:441–50.

    Article  CAS  PubMed  Google Scholar 

  26. Simonoff E, Pickles A, Chadwick O, Gringras P, Wood N, Higgins S, et al. The Croydon Assessment of Learning Study: prevalence and educational identification of mild mental retardation. J Child Psychol Psychiatry. 2006;47:828–39.

    Article  PubMed  Google Scholar 

  27. Westerinen H, Kaski M, Virta L, Almqvist F, Iivanainen M. Prevalence of intellectual disability: a comprehensive study based on national registers. J Intellect Disabil Res. 2007;51:715–25.

    Article  CAS  PubMed  Google Scholar 

  28. Wright CF, Fitzgerald TW, Jones WD, Clayton S, McRae JF, van Kogelenberg M, et al. Genetic diagnosis of developmental disorders in the DDD study: a scalable analysis of genome-wide research data. Lancet. 2015;385:1305–14.

    Article  PubMed  PubMed Central  Google Scholar 

  29. Vergano SA, van der Sluijs PJ, Santen G. ARID1B-related disorder. BTI - GeneReviews(®). Seattle: University of Washington; 1993.

  30. Santen GW, Clayton-Smith J, consortium ABC. The ARID1B phenotype: what we have learned so far. Am J Med Genet C Semin Med Genet. 2014;166C:276–89.

    Article  PubMed  Google Scholar 

  31. Mignot C, Moutard ML, Rastetter A, Boutaud L, Heide S, Billette T, et al. ARID1B mutations are the major genetic cause of corpus callosum anomalies in patients with intellectual disability. Brain. 2016;139:e64.

    Article  PubMed  Google Scholar 

  32. Santen GW, Aten E, Vulto-van Silfhout AT, Pottinger C, van Bon BW, van Minderhout IJ, et al. Coffin-Siris syndrome and the BAF complex: genotype-phenotype study in 63 patients. Hum Mutat. 2013;34:1519–28.

    Article  CAS  PubMed  Google Scholar 

  33. Wieczorek D, Bogershausen N, Beleggia F, Steiner-Haldenstatt S, Pohl E, Li Y, et al. A comprehensive molecular study on Coffin-Siris and Nicolaides-Baraitser syndromes identifies a broad molecular and clinical spectrum converging on altered chromatin remodeling. Hum Mol Genet. 2013;22:5121–35.

    Article  CAS  PubMed  Google Scholar 

  34. Miyake N, Tsurusaki Y, Matsumoto N, Numerous BAF. complex genes are mutated in Coffin-Siris syndrome. Am J Med Genet C Semin Med Genet. 2014;166C:257–61.

    Article  PubMed  Google Scholar 

  35. Bramswig NC, Caluseriu O, Ludecke HJ, Bolduc FV, Noel NC, Wieland T, et al. Heterozygosity for ARID2 loss-of-function mutations in individuals with a Coffin-Siris syndrome-like phenotype. Hum Genet. 2017;136:297–305.

    Article  CAS  PubMed  Google Scholar 

  36. Vasileiou G, Vergarajauregui S, Endele S, Popp B, Buttner C, Ekici AB, et al. Mutations in the BAF-complex subunit DPF2 are associated with Coffin-Siris Syndrome. Am J Hum Genet. 2018;102:468–79.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Kosho T, Okamoto N.Coffin-Siris Syndrome International Collaborators Genotype-phenotype correlation of Coffin-Siris syndrome caused by mutations in SMARCB1, SMARCA4, SMARCE1, and ARID1A. Am J Med Genet C Semin Med Genet. 2014;166C:262–75.

    Article  PubMed  Google Scholar 

  38. Pasca SP, Veenstra-VanderWeele J, McPartland JC. Research and training in autism spectrum disorder to catalyze the next genomic and neuroscience revolutions. Mol Psychiatry. 2020. https://doi.org/10.1038/s41380-020-0830-5.

  39. Knopf A. Autism prevalence increases from 1 in 60 to 1 in 54: CDC. Brown Univ Child Adolesc Behav Lett. 2020;36:4–4.

    Google Scholar 

  40. Fombonne E. The epidemiology of autism: a review. Psychol Med. 1999;29:769–86.

    Article  CAS  PubMed  Google Scholar 

  41. O’Brien G, Pearson J. Autism and learning disability. Autism. 2004;8:125–40.

    Article  PubMed  Google Scholar 

  42. Perou R, Bitsko RH, Blumberg SJ, Pastor P, Ghandour RM, Gfroerer JC, et al. Mental health surveillance among children-United States, 2005-2011. MMWR Suppl. 2013;62:1–35.

    PubMed  Google Scholar 

  43. Baio J, Wiggins L, Christensen DL, Maenner MJ, Daniels J, Warren Z, et al. Prevalence of autism spectrum disorder among children aged 8 years - autism and developmental disabilities monitoring network, 11 sites, United States, 2014. MMWR Surveill Summ. 2018;67:1–23.

    Article  PubMed  PubMed Central  Google Scholar 

  44. Nord AS, Roeb W, Dickel DE, Walsh T, Kusenda M, O’Connor KL, et al. Reduced transcript expression of genes affected by inherited and de novo CNVs in autism. Eur J Hum Genet. 2011;19:727–31.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Wing L, Gould J. Severe impairments of social interaction and associated abnormalities in children: epidemiology and classification. J Autism Dev Disord. 1979;9:11–29.

    Article  CAS  PubMed  Google Scholar 

  46. Purpura G, Fulceri F, Puglisi V, Masoni P, Contaldo A. Motor coordination impairment in children with autism spectrum disorder: a pilot study using Movement Assessment Battery for Children-2 Checklist. Minerva Pediatr. 2020;72:22–9.

  47. Robertson J, Hatton C, Emerson E, Baines S. Prevalence of epilepsy among people with intellectual disabilities: a systematic review. Seizure. 2015;29:46–62.

    Article  PubMed  Google Scholar 

  48. Kessi M, Xiong J, Wu L, Yang L, He F, Chen C, et al. Rare copy number variations and predictors in children with intellectual disability and epilepsy. Front Neurol. 2018;9:947.

    Article  PubMed  PubMed Central  Google Scholar 

  49. Ronan JL, Wu W, Crabtree GR. From neural development to cognition: unexpected roles for chromatin. Nat Rev Genet. 2013;14:347–59.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  50. Lopez AJ, Wood MA. Role of nucleosome remodeling in neurodevelopmental and intellectual disability disorders. Front Behav Neurosci. 2015;9:100.

    Article  PubMed  PubMed Central  Google Scholar 

  51. Gabriele M, Lopez Tobon A, D’Agostino G, Testa G. The chromatin basis of neurodevelopmental disorders: Rethinking dysfunction along the molecular and temporal axes. Prog Neuropsychopharmacol Biol Psychiatry. 2018;84:306–27.

    Article  CAS  PubMed  Google Scholar 

  52. Shibutani M, Horii T, Shoji H, Morita S, Kimura M, Terawaki N et al. Arid1b haploinsufficiency causes abnormal brain gene expression and autism-related behaviors in mice. Int J Mol Sci. 2017;18:1872.

  53. Daily DK, Ardinger HH, Holmes GE. Identification and evaluation of mental retardation. Am Fam Physician. 2000;61:1059–1067, 1070.

    CAS  PubMed  Google Scholar 

  54. Fakhoury M. Autistic spectrum disorders: a review of clinical features, theories and diagnosis. International journal of developmental neuroscience: the official journal of the International Society for. Dev Neurosci. 2015;43:70–77.

    Article  Google Scholar 

  55. Reid KA, Smiley E, Cooper SA. Prevalence and associations of anxiety disorders in adults with intellectual disabilities. J Intellect Disabil Res. 2011;55:172–81.

    Article  CAS  PubMed  Google Scholar 

  56. Nelson SB, Valakh V. Excitatory/Inhibitory balance and circuit homeostasis in autism spectrum disorders. Neuron. 2015;87:684–98.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  57. Ramamoorthi K, Lin Y. The contribution of GABAergic dysfunction to neurodevelopmental disorders. Trends Mol Med. 2011;17:452–62.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  58. Marin O. Interneuron dysfunction in psychiatric disorders. Nat Rev Neurosci. 2012;13:107–20.

    Article  CAS  PubMed  Google Scholar 

  59. Smith AL, Jung EM, Jeon BT, Kim WY. Arid1b haploinsufficiency in parvalbumin- or somatostatin-expressing interneurons leads to distinct ASD-like and ID-like behavior. Sci Rep. 2020;10:7834.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  60. Nagl NG Jr., Wang X, Patsialou A, Van Scoy M, Moran E. Distinct mammalian SWI/SNF chromatin remodeling complexes with opposing roles in cell-cycle control. EMBO J. 2007;26:752–63.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  61. Yan Z, Wang Z, Sharova L, Sharov AA, Ling C, Piao Y, et al. BAF250B-associated SWI/SNF chromatin-remodeling complex is required to maintain undifferentiated mouse embryonic stem cells. Stem Cells. 2008;26:1155–65.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  62. Wu JI, Lessard J, Olave IA, Qiu Z, Ghosh A, Graef IA, et al. Regulation of dendritic development by neuron-specific chromatin remodeling complexes. Neuron. 2007;56:94–108.

    Article  CAS  PubMed  Google Scholar 

  63. Redmond L, Kashani AH, Ghosh A. Calcium regulation of dendritic growth via CaM kinase IV and CREB-mediated transcription. Neuron. 2002;34:999–1010.

    Article  CAS  PubMed  Google Scholar 

  64. Kwon M, Fernandez JR, Zegarek GF, Lo SB, Firestein BL. BDNF-promoted increases in proximal dendrites occur via CREB-dependent transcriptional regulation of cypin. J Neurosci. 2011;31:9735–45.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  65. Rubio-Garrido P, Perez-de-Manzo F, Porrero C, Galazo MJ, Clasca F. Thalamic input to distal apical dendrites in neocortical layer 1 is massive and highly convergent. Cereb Cortex. 2009;19:2380–95.

    Article  PubMed  Google Scholar 

  66. Gilbert CD, Sigman M. Brain states: top-down influences in sensory processing. Neuron. 2007;54:677–96.

    Article  CAS  PubMed  Google Scholar 

  67. Sjostrom PJ, Hausser M. A cooperative switch determines the sign of synaptic plasticity in distal dendrites of neocortical pyramidal neurons. Neuron. 2006;51:227–38.

    Article  CAS  PubMed  Google Scholar 

  68. Aguirre-Chen C, Stec N, Ramos OM, Kim N, Kramer M, McCarthy S, et al. A Caenorhabditis elegans model for integrating the functions of neuropsychiatric risk genes identifies components required for normal dendritic morphology. G3. 2020;10:1617–28.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  69. John A, Brylka H, Wiegreffe C, Simon R, Liu P, Juttner R, et al. Bcl11a is required for neuronal morphogenesis and sensory circuit formation in dorsal spinal cord development. Development. 2012;139:1831–41.

    Article  CAS  PubMed  Google Scholar 

  70. Aizawa H, Hu SC, Bobb K, Balakrishnan K, Ince G, Gurevich I, et al. Dendrite development regulated by CREST, a calcium-regulated transcriptional activator. Science. 2004;303:197–202.

    Article  CAS  PubMed  Google Scholar 

  71. Martinez-Cerdeno V. Dendrite and spine modifications in autism and related neurodevelopmental disorders in patients and animal models. Dev Neurobiol. 2017;77:393–404.

    Article  PubMed  Google Scholar 

  72. Gilbert J, Man HY. Fundamental elements in autism: from neurogenesis and neurite growth to synaptic plasticity. Front Cell Neurosci. 2017;11:359.

    Article  PubMed  PubMed Central  Google Scholar 

  73. Overstreet-Wadiche L, McBain CJ. Neurogliaform cells in cortical circuits. Nat Rev Neurosci. 2015;16:458–68.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  74. Markram H, Toledo-Rodriguez M, Wang Y, Gupta A, Silberberg G, Wu C. Interneurons of the neocortical inhibitory system. Nat Rev Neurosci. 2004;5:793–807.

    Article  CAS  PubMed  Google Scholar 

  75. Tremblay R, Lee S, Rudy B. GABAergic interneurons in the neocortex: from cellular properties to circuits. Neuron. 2016;91:260–92.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  76. Urban-Ciecko J, Barth AL. Somatostatin-expressing neurons in cortical networks. Nat Rev Neurosci. 2016;17:401–9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  77. Benes FM, Berretta S. GABAergic interneurons: implications for understanding schizophrenia and bipolar disorder. Neuropsychopharmacology. 2001;25:1–27.

    Article  CAS  PubMed  Google Scholar 

  78. Lim L, Mi D, Llorca A, Marin O. Development and functional diversification of cortical interneurons. Neuron. 2018;100:294–313.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  79. Chattopadhyaya B, Cristo GD. GABAergic circuit dysfunctions in neurodevelopmental disorders. Front Psychiatry. 2012;3:51.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  80. Lee E, Lee J, Kim E. Excitation/Inhibition imbalance in animal models of autism spectrum disorders. Biol Psychiatry. 2017;81:838–47.

    Article  PubMed  Google Scholar 

  81. Puts NAJ, Wodka EL, Harris AD, Crocetti D, Tommerdahl M, Mostofsky SH, et al. Reduced GABA and altered somatosensory function in children with autism spectrum disorder. Autism research: official journal of the International Society for Autism. Research. 2017;10:608–19.

    Google Scholar 

  82. Robertson CE, Ratai EM, Kanwisher N. Reduced GABAergic action in the autistic brain. Curr Biol. 2016;26:80–85.

    Article  CAS  PubMed  Google Scholar 

  83. Harada M, Taki MM, Nose A, Kubo H, Mori K, Nishitani H, et al. Non-invasive evaluation of the GABAergic/glutamatergic system in autistic patients observed by MEGA-editing proton MR spectroscopy using a clinical 3 tesla instrument. J Autism Dev Disord. 2011;41:447–54.

    Article  PubMed  Google Scholar 

  84. Gaetz W, Bloy L, Wang DJ, Port RG, Blaskey L, Levy SE, et al. GABA estimation in the brains of children on the autism spectrum: measurement precision and regional cortical variation. NeuroImage. 2014;86:1–9.

    Article  CAS  PubMed  Google Scholar 

  85. Han S, Tai C, Westenbroek RE, Yu FH, Cheah CS, Potter GB, et al. Autistic-like behaviour in Scn1a+/- mice and rescue by enhanced GABA-mediated neurotransmission. Nature. 2012;489:385–90.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  86. Mao W, Watanabe T, Cho S, Frost JL, Truong T, Zhao X, et al. Shank1 regulates excitatory synaptic transmission in mouse hippocampal parvalbumin-expressing inhibitory interneurons. Eur J Neurosci. 2015;41:1025–35.

    Article  PubMed  PubMed Central  Google Scholar 

  87. Lauber E, Filice F, Schwaller B. Dysregulation of parvalbumin expression in the Cntnap2-/- mouse model of autism spectrum disorder. Front Mol Neurosci. 2018;11:262.

    Article  PubMed  PubMed Central  Google Scholar 

  88. Wiebe S, Nagpal A, Truong VT, Park J, Skalecka A, He AJ, et al. Inhibitory interneurons mediate autism-associated behaviors via 4E-BP2. Proc Natl Acad Sci USA. 2019;116:18060–7.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  89. Harris KM, Weinberg RJ. Ultrastructure of synapses in the mammalian brain. Cold Spring Harb Perspect Biol. 2012;4:5.

    Article  Google Scholar 

  90. Zoghbi HY, Bear MF. Synaptic dysfunction in neurodevelopmental disorders associated with autism and intellectual disabilities. Cold Spring Harbor Perspect Biol. 2012;4:152ps117.

    Article  Google Scholar 

  91. Kaeser PS, Deng L, Chavez AE, Liu X, Castillo PE, Sudhof TC. ELKS2alpha/CAST deletion selectively increases neurotransmitter release at inhibitory synapses. Neuron. 2009;64:227–39.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  92. Bao J, Li JJ, Perl ER. Differences in Ca2+ channels governing generation of miniature and evoked excitatory synaptic currents in spinal laminae I and II. J Neurosci. 1998;18:8740–50.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  93. Wall MJ, Usowicz MM. Development of the quantal properties of evoked and spontaneous synaptic currents at a brain synapse. Nat Neurosci. 1998;1:675–82.

    Article  CAS  PubMed  Google Scholar 

  94. Rho HS, Ahn SM, Hwang JS. Inhibitory effect of N-adamantyl-3,4-dihydroxybenzamide on melanogenesis in melan-a cells and brown guinea pigs. Arch Dermatol Res. 2011;303:153–9.

    Article  CAS  PubMed  Google Scholar 

  95. Selten M, van Bokhoven H, Nadif, Kasri N. Inhibitory control of the excitatory/inhibitory balance in psychiatric disorders. F1000Res. 2018;7:23.

    Article  PubMed  PubMed Central  Google Scholar 

  96. Sohal VS, Rubenstein JLR. Excitation-inhibition balance as a framework for investigating mechanisms in neuropsychiatric disorders. Mol Psychiatry. 2019;24:1248–57.

    Article  PubMed  PubMed Central  Google Scholar 

  97. Ajram LA, Horder J, Mendez MA, Galanopoulos A, Brennan LP, Wichers RH, et al. Shifting brain inhibitory balance and connectivity of the prefrontal cortex of adults with autism spectrum disorder. Transl psychiatry. 2017;7:e1137.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  98. Lunden JW, Durens M, Phillips AW, Nestor MW. Cortical interneuron function in autism spectrum condition. Pediatr Res. 2019;85:146–54.

    Article  CAS  PubMed  Google Scholar 

  99. Karayannis T, Au E, Patel JC, Kruglikov I, Markx S, Delorme R, et al. Cntnap4 differentially contributes to GABAergic and dopaminergic synaptic transmission. Nature. 2014;511:236–40.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  100. Gogolla N, Leblanc JJ, Quast KB, Sudhof TC, Fagiolini M, Hensch TK. Common circuit defect of excitatory-inhibitory balance in mouse models of autism. J Neurodev Disord. 2009;1:172–81.

    Article  PubMed  PubMed Central  Google Scholar 

  101. Pizzarelli R, Cherubini E. Alterations of GABAergic signaling in autism spectrum disorders. Neural Plast. 2011;2011:297153.

    Article  PubMed  PubMed Central  Google Scholar 

  102. Wohr M, Orduz D, Gregory P, Moreno H, Khan U, Vorckel KJ, et al. Lack of parvalbumin in mice leads to behavioral deficits relevant to all human autism core symptoms and related neural morphofunctional abnormalities. Transl Psychiatry. 2015;5:e525.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  103. Ito-Ishida A, Ure K, Chen H, Swann JW, Zoghbi HY. Loss of MeCP2 in parvalbumin-and somatostatin-expressing neurons in mice leads to distinct Rett syndrome-like phenotypes. Neuron. 2015;88:651–8.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  104. Selby L, Zhang C, Sun QQ. Major defects in neocortical GABAergic inhibitory circuits in mice lacking the fragile X mental retardation protein. Neurosci Lett. 2007;412:227–32.

    Article  CAS  PubMed  Google Scholar 

  105. Lauber E, Filice F, Schwaller B. Prenatal valproate exposure differentially affects parvalbumin-expressing neurons and related circuits in the cortex and striatum of mice. Front Mol Neurosci. 2016;9:150.

    Article  PubMed  PubMed Central  Google Scholar 

  106. Hargreaves DC, Crabtree GR. ATP-dependent chromatin remodeling: genetics, genomics and mechanisms. Cell Res. 2011;21:396–420.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  107. Clapier CR, Iwasa J, Cairns BR, Peterson CL. Mechanisms of action and regulation of ATP-dependent chromatin-remodelling complexes. Nat Rev Mol Cell Biol. 2017;18:407–22.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  108. Zhou CY, Johnson SL, Gamarra NI, Narlikar GJ. Mechanisms of ATP-dependent chromatin remodeling motors. Annu Rev Biophys. 2016;45:153–81.

    Article  CAS  PubMed  Google Scholar 

  109. Kadoch C, Crabtree GR. Mammalian SWI/SNF chromatin remodeling complexes and cancer: Mechanistic insights gained from human genomics. Sci Adv. 2015;1:e1500447.

    Article  PubMed  PubMed Central  Google Scholar 

  110. Kadoch C, Hargreaves DC, Hodges C, Elias L, Ho L, Ranish J, et al. Proteomic and bioinformatic analysis of mammalian SWI/SNF complexes identifies extensive roles in human malignancy. Nat Genet. 2013;45:592–601.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  111. Masliah-Planchon J, Bieche I, Guinebretiere JM, Bourdeaut F, Delattre O. SWI/SNF chromatin remodeling and human malignancies. Annu Rev Pathol. 2015;10:145–71.

    Article  CAS  PubMed  Google Scholar 

  112. Ho L, Crabtree GR. Chromatin remodelling during development. Nature. 2010;463:474–84.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  113. Dallas PB, Pacchione S, Wilsker D, Bowrin V, Kobayashi R, Moran E. The human SWI-SNF complex protein p270 is an ARID family member with non-sequence-specific DNA binding activity. Mol Cell Biol. 2000;20:3137–46.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  114. He S, Wu Z, Tian Y, Yu Z, Yu J, Wang X, et al. Structure of nucleosome-bound human BAF complex. Science. 2020;367:875–81.

    Article  CAS  PubMed  Google Scholar 

  115. Han Y, Reyes AA, Malik S, He Y. Cryo-EM structure of SWI/SNF complex bound to a nucleosome. Nature. 2020;579:452–5.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  116. Gandal MJ, Nesbitt AM, McCurdy RM, Alter MD. Measuring the maturity of the fast-spiking interneuron transcriptional program in autism, schizophrenia, and bipolar disorder. PLoS ONE. 2012;7:e41215.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  117. Li XS, Trojer P, Matsumura T, Treisman JE, Tanese N. Mammalian SWI/SNF-a subunit BAF250/ARID1 is an E3 ubiquitin ligase that targets histone H2B. Mol Cell Biol. 2010;30:1673–88.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  118. Barker N, Hurlstone A, Musisi H, Miles A, Bienz M, Clevers H. The chromatin remodelling factor Brg-1 interacts with beta-catenin to promote target gene activation. EMBO J. 2001;20:4935–43.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  119. Son EY, Crabtree GR. The role of BAF (mSWI/SNF) complexes in mammalian neural development. Am J Med Genet C Semin Med Genet. 2014;166C:333–49.

    Article  PubMed  Google Scholar 

  120. Budimirovic DB, Berry-Kravis E, Erickson CA, Hall SS, Hessl D, Reiss AL, et al. Updated report on tools to measure outcomes of clinical trials in fragile X syndrome. J Neurodev Disord. 2017;9:14.

    Article  PubMed  PubMed Central  Google Scholar 

  121. Tombaugh TN, Kozak J, Rees L. Normative data stratified by age and education for two measures of verbal fluency: FAS and animal naming. Arch Clin Neuropsychol. 1999;14:167–77.

    CAS  PubMed  Google Scholar 

  122. Basilico B, Morandell J, Novarino G. Molecular mechanisms for targeted ASD treatments. Curr Opin Genet Dev. 2020;65:126–37.

    Article  CAS  PubMed  Google Scholar 

  123. Sgritta M, Dooling SW, Buffington SA, Momin EN, Francis MB, Britton RA, et al. Mechanisms underlying microbial-mediated changes in social behavior in mouse models of autism spectrum disorder. Neuron. 2019;101:246–59 e246.

    Article  CAS  PubMed  Google Scholar 

  124. Cryan JF, O’Riordan KJ, Sandhu K, Peterson V, Dinan TG. The gut microbiome in neurological disorders. Lancet Neurol. 2020;19:179–94.

    Article  CAS  PubMed  Google Scholar 

  125. Han S, Tai C, Jones CJ, Scheuer T, Catterall WA. Enhancement of inhibitory neurotransmission by GABAA receptors having alpha2,3-subunits ameliorates behavioral deficits in a mouse model of autism. Neuron. 2014;81:1282–9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  126. Rhine MA, Parrott JM, Schultz MN, Kazdoba TM, Crawley JN. Hypothesis-driven investigations of diverse pharmacological targets in two mouse models of autism. Autism Res. 2019;12:401–21.

    Article  PubMed  PubMed Central  Google Scholar 

  127. Nardi AE, Perna G. Clonazepam in the treatment of psychiatric disorders: an update. Int Clin Psychopharmacol. 2006;21:131–42.

    Article  PubMed  Google Scholar 

  128. Delbeke J, Hoffman L, Mols K, Braeken D, Prodanov D. And then there was light: perspectives of optogenetics for deep brain stimulation and neuromodulation. Front Neurosci. 2017;11:663.

    Article  PubMed  PubMed Central  Google Scholar 

  129. Demars MP, Morishita H. Cortical parvalbumin and somatostatin GABA neurons express distinct endogenous modulators of nicotinic acetylcholine receptors. Mol Brain. 2014;7:75.

    Article  PubMed  PubMed Central  Google Scholar 

  130. Pirola B, Bortotto L, Giglio S, Piovan E, Janes A, Guerrini R et al. Agenesis of the corpus callosum with Probst bundles owing to haploinsufficiency for a gene in an 8 cM region of 6q25. J Med Genet. 1998;35:1031–33.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  131. Nagamani SC, Erez A, Eng C, Ou Z, Chinault C, Workman L et al. Interstitial deletion of 6q25.2-q25.3: a novel microdeletion syndrome associated with microcephaly, developmental delay, dysmorphic features and hearing loss. Eur J Hum Genet 2009;17:573–81.

    Article  CAS  PubMed  Google Scholar 

  132. Tsurusaki Y, Okamoto N, Ohashi H, Kosho T, Imai Y, Hibi-Ko Y et al. Mutations affecting components of the SWI/SNF complex cause Coffin-Siris syndrome. Nat genet 2012;44:376–8.

    Article  CAS  PubMed  Google Scholar 

  133. Vengoechea J, Carpenter L, Zarate YA. Papillary thyroid cancer in a patient with interstitial 6q25 deletion including ARID1B. Am J Med Genet A 2014;164A:1857–9.

Download references

Acknowledgements

This work was supported by the National Institute of Neurological Disorders and Stroke of the National Institutes of Health under award number R01NS091220 to WYK. Figures were created in part using Biorender.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Woo-Yang Kim.

Ethics declarations

Conflict of 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.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Moffat, J.J., Smith, A.L., Jung, EM. et al. Neurobiology of ARID1B haploinsufficiency related to neurodevelopmental and psychiatric disorders. Mol Psychiatry 27, 476–489 (2022). https://doi.org/10.1038/s41380-021-01060-x

Download citation

  • Received:

  • Revised:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41380-021-01060-x

This article is cited by

Search

Quick links