HELP     Sign In
Search

Relevance to Autism

Two de novo frameshift variants in the ARID1B gene were identified by exome sequencing in unrelated simplex ASD cases (PMIDs 22495309 and 23160955). Nord et al., 2011 (PMID 21448237) had previously identified a de novo deletion within the ARID1B gene resulting in reduced transcript expression in a patient with autism, and a de novo translocation and deletions disrupting ARID1B had previously been identified in ASD patients in Halgren et al., 2011 (PMID 21801163). Three additional de novo loss-of-function variants in ARID1B were identified in ASD probands from the Autism Sequencing Consortium (ASC) in De Rubeis et al., 2014. Furthermore, analysis of rare coding variation in 3,871 ASD cases and 9,937 ancestry-matched or paternal controls from ASC in this report identified ARID1B as a gene meeting high statistical significance with a FDR 0.01, meaning that this gene had a 99% chance of being a true autism gene (PMID 25363760). This gene was identified in Iossifov et al. 2015 as a strong candidate to be an ASD risk gene based on a combination of de novo mutational evidence and the absence or very low frequency of mutations in controls (PMID 26401017). A two-stage analysis of rare de novo and inherited coding variants in 42,607 ASD cases, including 35,130 new cases from the SPARK cohort, in Zhou et al., 2022 identified ARID1B as a gene reaching exome-wide significance (P < 2.5E-06). Variants in ARID1B are also responsible for Coffin-Siris syndrome (CSS); a subset of CSS patients have been reported to show ASD or autistic features (PMIDs 22426309, 24569609).

Molecular Function

This locus encodes an AT-rich DNA interacting domain-containing protein. The encoded protein is a component of the SWI/SNF chromatin remodeling complex and may play a role in cell-cycle activation. The protein encoded by this locus is similar to AT-rich interactive domain-containing protein 1A. These two proteins function as alternative, mutually exclusive ARID-subunits of the SWI/SNF complex. The associated complexes play opposing roles.

External Links

        

References

Type
Title
Type of Disorder
Associated Disorders
Author, Year
Primary
Reduced transcript expression of genes affected by inherited and de novo CNVs in autism.
ASD
Support
Comprehensive whole genome sequence analyses yields novel genetic and structural insights for Intellectual Disability.
ASD, DD
Support
Identification of de novo mutations for ARID1B haploinsufficiency associated with Coffin-Siris syndrome 1 in three Chinese families via array-CGH and whole exome sequencing
DD
Epilepsy/seizures
Support
Large-scale discovery of novel genetic causes of developmental disorders.
ASD, DD, ID
Support
Large-Scale Exome Sequencing Study Implicates Both Developmental and Functional Changes in the Neurobiology of Autism
ASD
Support
DD, epilepsy/seizures
Support
High Rate of Recurrent De Novo Mutations in Developmental and Epileptic Encephalopathies.
Epilepsy/seizures
DD/ID
Support
Clinical Targeted Panel Sequencing Analysis in Clinical Evaluation of Children with Autism Spectrum Disorder in China
ASD
Support
High diagnostic yield of syndromic intellectual disability by targeted next-generation sequencing.
Coffin-Siris syndrome
ID
Support
De novo variants in neurodevelopmental disorders-experiences from a tertiary care center
DD
Support
Mutations in SWI/SNF chromatin remodeling complex gene ARID1B cause Coffin-Siris syndrome.
Coffin-Siris syndrome
ID, ASD
Support
Lessons Learned from Large-Scale, First-Tier Clinical Exome Sequencing in a Highly Consanguineous Population.
DD, ID
ADHD, OCD, epilepsy/seizures
Support
The HHID syndrome of hypertrichosis, hyperkeratosis, abnormal corpus callosum, intellectual disability, and minor anomalies is caused by mutations ...
Hypertrichosis, hyperkeratosis, mental retardation
Support
Phenotypic and molecular spectra of patients with switch/sucrose nonfermenting complex-related intellectual disability disorders in Korea
Coffin-Siris syndrome 1, DD
ID, epilepsy/seizures, Afs
Support
De novo mutations in moderate or severe intellectual disability.
ID, epilepsy/seizures
Speech delay, hypotonia
Support
De Novo ARID1B mutations cause growth delay associated with aberrant Wnt/-catenin signaling.
Coffin-Siris syndrome
Support
DD
Support
Genomic Patterns of De Novo Mutation in Simplex Autism
ASD
Support
2022
ASD, SCZ
Support
Meta-analysis of 2,104 trios provides support for 10 new genes for intellectual disability
ID
Support
Differential roles of ARID1B in excitatory and inhibitory neural progenitors in the developing cortex
Support
Mutations affecting components of the SWI/SNF complex cause Coffin-Siris syndrome.
Support
Inherited and multiple de novo mutations in autism/developmental delay risk genes suggest a multifactorial model.
ASD
Support
Whole genome sequencing resource identifies 18 new candidate genes for autism spectrum disorder
ASD
Support
Prevalence and phenotypic impact of rare potentially damaging variants in autism spectrum disorder
ASD
Support
Expanding the phenotypic spectrum of ARID1B-mediated disorders and identification of altered cell-cycle dynamics due to ARID1B haploinsufficiency.
ID
Support
Phenotype-to-genotype approach reveals head-circumference-associated genes in an autism spectrum disorder cohort.
ASD
Macrocephaly
Support
Coffin-Siris syndrome 1, ASD, DD, epilepsy/seizure
Support
Expanding the genetic heterogeneity of intellectual disability.
ID, epilepsy/seizures
Support
Deep phenotyping and whole-exome sequencing improved the diagnostic yield for nuclear pedigrees with neurodevelopmental disorders
ID
Support
Comprehensive molecular testing in patients with high functioning autism spectrum disorder.
ASD
Support
The landscape of somatic mutation in cerebral cortex of autistic and neurotypical individuals revealed by ultra-deep whole-genome sequencing
ASD
Support
Haploinsufficiency of ARID1B, a member of the SWI/SNF-a chromatin-remodeling complex, is a frequent cause of intellectual disability.
ID
Autistic features
Support
Genome sequencing identifies multiple deleterious variants in autism patients with more severe phenotypes.
ASD
Support
Targeted sequencing identifies 91 neurodevelopmental-disorder risk genes with autism and developmental-disability biases.
ASD, DD
Support
A single center experience with publicly funded clinical exome sequencing for neurodevelopmental disorders or multiple congenital anomalies
DD, ID
ASD, epilepsy/seizures
Support
Coffin-Siris Syndrome with obesity, macrocephaly, hepatomegaly and hyperinsulinism caused by a mutation in the ARID1B gene.
Coffin-Siris syndrome
ID, autistic features
Support
Exome sequencing of 457 autism families recruited online provides evidence for autism risk genes
ASD
Support
ID, epilepsy/seizures
Support
Rates, distribution and implications of postzygotic mosaic mutations in autism spectrum disorder.
ASD
Support
Genetic care in geographically isolated small island communities: 8 years of experience in the Dutch Caribbean
DD, ID
Support
Targeted DNA Sequencing from Autism Spectrum Disorder Brains Implicates Multiple Genetic Mechanisms.
ASD
Support
A recurrent PJA1 variant in trigonocephaly and neurodevelopmental disorders
DD
Support
A 69-year-old woman with Coffin-Siris syndrome.
Coffin-Siris syndrome
DD, ID, autistic behavior
Support
Clinical exome sequencing: results from 2819 samples reflecting 1000 families.
DD, ID, epilepsy/seizures
ADHD
Support
Comprehensive genetic analysis confers high diagnostic yield in 16 Japanese patients with corpus callosum anomalies
Corpus callosum anomalies
ASD, DD, ID, epilepsy/seizures
Support
A discovery resource of rare copy number variations in individuals with autism spectrum disorder.
ASD
Support
Impact of on-site clinical genetics consultations on diagnostic rate in children and young adults with autism spectrum disorder.
ASD
Support
ASD
DD, ID
Support
Using medical exome sequencing to identify the causes of neurodevelopmental disorders: experience of two clinical units and 216 patients.
ID
Coffin-Siris syndrome
Support
Mutational Landscape of Autism Spectrum Disorder Brain Tissue
ASD
Support
Insights into Autism Spectrum Disorder Genomic Architecture and Biology from 71 Risk Loci.
ASD
Support
Coffin-Siris Syndrome-1: Report of five cases from Asian populations with truncating mutations in the ARID1B gene
Coffin-Siris syndrome
Support
Coffin-Siris syndrome 1
Support
Risks and Recommendations in Prenatally Detected De Novo Balanced Chromosomal Rearrangements from Assessment of Long-Term Outcomes.
ASD, ID
Support
Integrating de novo and inherited variants in 42
ASD
Support
De novo genic mutations among a Chinese autism spectrum disorder cohort.
ASD
Support
Genotype and phenotype in 18 Chinese patients with Coffin-Siris syndrome
Coffin-Siris syndrome, DD/ID
ASD, epilepsy/seizures
Support
Multiplex targeted sequencing identifies recurrently mutated genes in autism spectrum disorders.
ASD
Support
Inherited and De Novo Genetic Risk for Autism Impacts Shared Networks.
ASD
Support
ID, epilepsy/seizures
Support
Genomic diagnosis for children with intellectual disability and/or developmental delay.
ID
Epilepsy/seizures
Support
Genetic Analysis of Children With Unexplained Developmental Delay and/or Intellectual Disability by Whole-Exome Sequencing
DD
Support
Excess of rare, inherited truncating mutations in autism.
ASD
Support
Excess of de novo variants in genes involved in chromatin remodelling in patients with marfanoid habitus and intellectual disability
ID
Marfanoid habitus
Support
Coffin-Siris syndrome 1, ASD, DD
Support
Diagnostic exome sequencing of syndromic epilepsy patients in clinical practice.
DD, epilepsy/seizures, microcephaly
Support
A novel nonsense variant in ARID1B causing simultaneous RNA decay and exon skipping is associated with Coffin-Siris syndrome
Coffin-Siris syndrome, DD
Support
Mutations in Human Accelerated Regions Disrupt Cognition and Social Behavior.
ASD
Support
Genetic and phenotypic analysis of 101 patients with developmental delay or intellectual disability using whole-exome sequencing
DD, epilepsy/seizures
ASD
Support
Sporadic autism exomes reveal a highly interconnected protein network of de novo mutations.
ASD
Support
Characterization of intellectual disability and autism comorbidity through gene panel sequencing.
ID, ASD or autistic traits
Recent Recommendation
Genome Sequencing of Autism-Affected Families Reveals Disruption of Putative Noncoding Regulatory DNA
ASD
Recent Recommendation
Low load for disruptive mutations in autism genes and their biased transmission.
ASD
Recent Recommendation
ASD
Recent Recommendation
Synaptic, transcriptional and chromatin genes disrupted in autism.
ASD
Recent Recommendation
Recent Recommendation
Corpus callosum abnormalities, intellectual disability, speech impairment, and autism in patients with haploinsufficiency of ARID1B.
ASD or autistic traits
ID
Recent Recommendation
Craniosynostosis
ASD
Recent Recommendation
Arachnoid cysts
ASD, DD, epilepsy/seizures
Recent Recommendation
Autism genes converge on asynchronous development of shared neuron classes
ASD

Rare

Variant ID
Variant Type
Allele Change
Residue Change
Inheritance Pattern
Inheritance Association
Family Type
Author, Year
 GEN357R001 
 copy_number_loss 
  
  
 De novo 
  
  
 GEN357R002 
 translocation 
  
  
 De novo 
  
  
 GEN357R003 
 copy_number_loss 
  
  
 De novo 
  
  
 GEN357R004 
 copy_number_loss 
  
  
 De novo 
  
  
 GEN357R005 
 copy_number_loss 
  
  
 De novo 
  
  
 GEN357R006 
 frameshift_variant 
 c.5509del 
 p.Leu1837CysfsTer54 
 De novo 
  
 Simplex 
 GEN357R007 
 frameshift_variant 
 c.3706dup 
 p.Gln1236ProfsTer14 
 De novo 
  
 Simplex 
 GEN357R008 
 copy_number_loss 
  
  
 Unknown 
  
 Unknown 
 GEN357R009 
 stop_gained 
 c.5329A>T 
 p.Lys1777Ter 
 De novo 
  
 Simplex 
 GEN357R010 
 stop_gained 
 c.3223C>T 
 p.Arg1075Ter 
 De novo 
  
 Simplex 
 GEN357R011 
 frameshift_variant 
 c.4620_4629del 
 p.Met1540IlefsTer4 
 De novo 
  
 Simplex 
 GEN357R012 
 copy_number_gain 
  
  
 De novo 
  
  
 GEN357R013 
 stop_gained 
 c.3919C>T 
 p.Pro1307Ser 
 De novo 
  
  
 GEN357R014 
 frameshift_variant 
 c.6463_6473del 
 p.Leu2155ArgfsTer73 
 De novo 
  
  
 GEN357R015 
 stop_gained 
 c.3304C>T 
 p.Arg1102Ter 
 De novo 
  
  
 GEN357R016 
 frameshift_variant 
 c.3326_3327del 
 p.Arg1109LysfsTer48 
 De novo 
  
  
 GEN357R017 
 splice_site_variant 
 c.4230G>A 
 p.Pro1410= 
 De novo 
  
  
 GEN357R018 
 stop_gained 
 c.4038T>A 
 p.Tyr1346Ter 
 De novo 
  
  
 GEN357R019 
 frameshift_variant 
 c.1114dup 
 p.Arg372ProfsTer163 
 De novo 
  
  
 GEN357R020 
 frameshift_variant 
 c.1584del 
 p.Leu528PhefsTer62 
 De novo 
  
 Simplex 
 GEN357R021 
 copy_number_loss 
  
  
 De novo 
  
  
 GEN357R022 
 frameshift_variant 
 c.3208_3209del 
 p.Ser1070Ter 
 De novo 
  
  
 GEN357R023 
 frameshift_variant 
 c.2306delinsTCCGCAGCCACT 
 p.Ser769IlefsTer67 
 De novo 
  
  
 GEN357R024 
 frameshift_variant 
 c.4274dup 
 p.Arg1426AlafsTer73 
 De novo 
  
  
 GEN357R025 
 stop_gained 
 c.2941C>T 
 p.Gln981Ter 
 De novo 
  
  
 GEN357R026 
 stop_gained 
 c.2977C>T 
 p.Gln993Ter 
 De novo 
  
 Simplex 
 GEN357R027 
 stop_gained 
 c.3020C>A 
 p.Ser1007Ter 
 De novo 
  
 Simplex 
 GEN357R028 
 frameshift_variant 
 c.5235_5236insG 
 p.Pro1746AlafsTer7 
 De novo 
  
 Simplex 
 GEN357R029 
 missense_variant 
 c.5903A>G 
 p.Glu1968Gly 
 De novo 
  
 Simplex 
 GEN357R030 
 missense_variant 
 c.4097G>A 
 p.Arg1366His 
 Familial 
 Maternal 
 Simplex 
 GEN357R031 
 missense_variant 
 c.3791T>G 
 p.Met1264Arg 
 Familial 
 Paternal 
 Simplex 
 GEN357R032 
 missense_variant 
 c.4211A>G 
 p.Tyr1404Cys 
 Familial 
 Maternal 
 Simplex 
 GEN357R033 
 missense_variant 
 c.1880C>T 
 p.Pro627Leu 
 Familial 
 Paternal 
 Simplex 
 GEN357R034 
 missense_variant 
 c.2258C>T 
 p.Pro753Leu 
 Familial 
 Paternal 
 Simplex 
 GEN357R035 
 missense_variant 
 c.1702G>A 
 p.Gly568Arg 
 Unknown 
  
 Unknown 
 GEN357R036 
 missense_variant 
 c.2939A>T 
 p.Gln980Leu 
 Unknown 
  
 Unknown 
 GEN357R037 
 missense_variant 
 c.2404T>C 
 p.Ser802Pro 
 Unknown 
  
 Unknown 
 GEN357R038 
 missense_variant 
 c.3670+36G>A 
  
 Unknown 
  
 Unknown 
 GEN357R039 
 missense_variant 
 c.3254C>T 
 p.Pro1085Leu 
 Unknown 
  
 Unknown 
 GEN357R040 
 stop_gained 
 c.5404C>T 
 p.Arg1802Ter 
 De novo 
  
 Simplex 
 GEN357R041 
 stop_gained 
 c.1914C>A 
 p.Tyr638Ter 
 De novo 
  
 Simplex 
 GEN357R042 
 frameshift_variant 
 c.6562del 
 p.Ala2188ProfsTer2 
 De novo 
  
 Simplex 
 GEN357R043 
 stop_gained 
 c.5776C>T 
 p.Arg1926Ter 
 De novo 
  
 Simplex 
 GEN357R044 
 stop_gained 
 c.4045C>T 
 p.Gln1349Ter 
 De novo 
  
 Simplex 
 GEN357R045 
 splice_site_variant 
 c.4894G>A 
 p.Val1632Ile 
 De novo 
  
 Simplex 
 GEN357R046 
 stop_gained 
 c.1621C>T 
 p.Gln541Ter 
 De novo 
  
 Simplex 
 GEN357R047 
 missense_variant 
 c.2496G>T 
 p.Gln832His 
 De novo 
  
 Multi-generational 
 GEN357R048 
 stop_gained 
 c.2507C>G 
 p.Pro836Arg 
 De novo 
  
 Multi-generational 
 GEN357R049 
 frameshift_variant 
 c.5267_5270del 
 p.Ala1756GlyfsTer49 
 De novo 
  
 Simplex 
 GEN357R050 
 frameshift_variant 
 c.4144del 
 p.Tyr1382MetfsTer106 
 De novo 
  
 Multiplex 
 GEN357R051 
 stop_gained 
 c.1729C>T 
 p.Gln577Ter 
 De novo 
  
 Simplex 
 GEN357R052 
 missense_variant 
 c.5015A>G 
 p.Asn1672Ser 
 De novo 
  
 Simplex 
 GEN357R053 
 missense_variant 
 c.4237C>T 
 p.Pro1413Ser 
 Unknown 
  
 Multiplex or multi-generational 
 GEN357R054 
 missense_variant 
 c.1463C>G 
 p.Pro488Arg 
 Unknown 
  
 Unknown 
 GEN357R055 
 copy_number_loss 
  
  
 Familial 
 Maternal 
 Simplex 
 GEN357R056 
 intergenic_variant 
 c.1999-4542C>A 
  
 De novo 
  
 Simplex 
 GEN357R057 
 missense_variant 
 c.736G>A 
 p.Gly246Ser 
 Familial 
 Paternal 
 Multi-generational 
 GEN357R058 
 missense_variant 
 c.736G>A 
 p.Gly246Ser 
 Unknown 
 Not paternal 
 Simplex 
 GEN357R059 
 missense_variant 
 c.736G>A 
 p.Gly246Ser 
 Familial 
 Paternal 
 Multi-generational 
 GEN357R060 
 missense_variant 
 c.122C>T 
 p.Ser41Phe 
 Familial 
 Paternal 
 Multi-generational 
 GEN357R061 
 frameshift_variant 
 c.5072del 
 p.Ser1691IlefsTer33 
 De novo 
  
  
 GEN357R062 
 stop_gained 
 c.6511C>T 
 p.Gln2171Ter 
 De novo 
  
  
 GEN357R063 
 stop_gained 
 c.4566T>A 
 p.Tyr1522Ter 
 De novo 
  
  
 GEN357R064 
 stop_gained 
 c.2318C>G 
 p.Ser773Ter 
 De novo 
  
  
 GEN357R065 
 stop_gained 
 c.4741C>T 
 p.Gln1581Ter 
 De novo 
  
  
 GEN357R066 
 intergenic_variant 
 delG 
  
  
  
 Unknown 
 GEN357R067 
 frameshift_variant 
 c.4395del 
 p.Tyr1467ThrfsTer21 
 Unknown 
 Not paternal 
  
 GEN357R068 
 frameshift_variant 
 c.5547dup 
 p.Gln1850AlafsTer5 
 De novo 
  
 Simplex 
 GEN357R069 
 frameshift_variant 
 c.5376_5379del 
 p.Ser1792ArgfsTer13 
 De novo 
  
 Simplex 
 GEN357R070 
 splice_site_variant 
 c.1888-2A>G 
  
 De novo 
  
  
 GEN357R071 
 stop_gained 
 c.2938C>T 
 p.Gln980Ter 
 De novo 
  
 Simplex 
 GEN357R072 
 frameshift_variant 
 c.5228_5231del 
 p.Val1743GlufsTer62 
 De novo 
  
  
 GEN357R073 
 stop_gained 
 c.3424C>T 
 p.Arg1142Ter 
 De novo 
  
  
 GEN357R074 
 stop_gained 
 c.2653C>T 
 p.Arg885Ter 
 De novo 
  
  
 GEN357R075 
 stop_gained 
 c.1729C>T 
 p.Gln577Ter 
 De novo 
  
 Simplex 
 GEN357R076 
 stop_gained 
 c.1762G>T 
 p.Gly588Cys 
 Familial 
  
 Simplex 
 GEN357R077 
 stop_gained 
 c.1762G>T 
 p.Gly588Cys 
 Familial 
  
 Simplex 
 GEN357R078 
 splice_site_variant 
 A>G 
 p.? 
 Familial 
  
 Multiplex 
 GEN357R079 
 splice_site_variant 
 c.4133G>A 
 p.Arg1378Lys 
 Familial 
  
 Multiplex 
 GEN357R080 
 stop_gained 
 c.3678G>T 
 p.Ser1226= 
 Unknown 
  
 Simplex 
 GEN357R081 
 frameshift_variant 
 c.5376_5379del 
 p.Ser1792ArgfsTer13 
 De novo 
  
  
 GEN357R082 
 splice_site_variant 
 c.4110G>A 
 p.Pro1370= 
 De novo 
  
  
 GEN357R083 
 frameshift_variant 
 c.3716del 
 p.Gly1239AlafsTer12 
 De novo 
  
 Simplex 
 GEN357R084 
 frameshift_variant 
 c.1595del 
 p.Gly532AlafsTer58 
 De novo 
  
 Simplex 
 GEN357R085 
 stop_gained 
 c.6100C>T 
 p.Gln2034Ter 
 De novo 
  
  
 GEN357R086 
 stop_gained 
 c.2242C>T 
 p.Gln748Ter 
 De novo 
  
  
 GEN357R087 
 stop_gained 
 c.5830C>T 
 p.Pro1944Ser 
 Familial 
 Paternal 
  
 GEN357R088 
 splice_site_variant 
 c.5026G>A 
 p.Ala1676Thr 
 De novo 
  
  
 GEN357R089 
 missense_variant 
 c.6475G>A 
 p.Val2159Ile 
 De novo 
  
 Simplex 
 GEN357R090 
 stop_gained 
 c.5911G>T 
 p.Glu1971Ter 
 De novo 
  
 Simplex 
 GEN357R091 
 frameshift_variant 
 c.1504del 
 p.Ser502AlafsTer21 
 De novo 
  
 Simplex 
 GEN357R092 
 frameshift_variant 
 c.6764del 
 p.Phe2255SerfsTer17 
 De novo 
  
 Simplex 
 GEN357R093 
 splice_site_variant 
 c.4230G>A 
 p.Pro1410= 
 Unknown 
  
  
 GEN357R094 
 translocation 
  
  
 De novo 
  
  
 GEN357R095 
 frameshift_variant 
 c.5063_5064dup 
 p.Ala1689TrpfsTer36 
 Unknown 
  
 Simplex 
 GEN357R096 
 stop_gained 
 c.2528C>T 
 p.Pro843Leu 
 De novo 
  
 Simplex 
 GEN357R097 
 stop_gained 
 c.2536C>T 
 p.Gln846Ter 
 De novo 
  
 Simplex 
 GEN357R098 
 missense_variant 
 c.2056G>A 
 p.Ala686Thr 
 Familial 
 Maternal 
 Simplex 
 GEN357R099 
 missense_variant 
 c.3220G>A 
 p.Asp1074Asn 
 Familial 
 Maternal 
 Simplex 
 GEN357R100 
 stop_gained 
 c.6010G>T 
 p.Glu2004Ter 
 De novo 
  
  
 GEN357R101 
 missense_variant 
 c.4286G>A 
 p.Gly1429Glu 
 Unknown 
  
 Simplex 
 GEN357R102 
 missense_variant 
 c.6707T>C 
 p.Leu2236Ser 
 Unknown 
  
 Unknown 
 GEN357R103 
 frameshift_variant 
 c.5032_5035del 
 p.Arg1678Ter 
 Unknown 
  
 Unknown 
 GEN357R104 
 splice_site_variant 
 c.4110G>A 
 p.Pro1370= 
 De novo 
  
 Simplex 
 GEN357R105 
 splice_site_variant 
 c.2333-2A>C 
  
 Familial 
 Paternal 
 Multiplex 
 GEN357R106 
 missense_variant 
 c.6410A>C 
 p.Asp2137Ala 
 De novo 
  
  
 GEN357R107 
 missense_variant 
 c.3214T>C 
 p.Ser1072Pro 
 De novo 
  
 Simplex 
 GEN357R108 
 stop_gained 
 c.1831C>T 
 p.Pro611Ser 
 De novo 
  
 Simplex 
 GEN357R109 
 stop_gained 
 c.6683C>A 
 p.Ser2228Ter 
 De novo 
  
 Simplex 
 GEN357R110 
 stop_gained 
 c.4845G>T 
 p.Leu1615= 
 De novo 
  
 Unknown 
 GEN357R111 
 stop_gained 
 c.5551G>T 
 p.Glu1851Ter 
 De novo 
  
 Simplex 
 GEN357R112 
 stop_gained 
 c.4129C>T 
 p.Arg1377Ter 
 De novo 
  
 Simplex 
 GEN357R113 
 missense_variant 
 c.2231C>T 
 p.Ala744Val 
 De novo 
  
 Simplex 
 GEN357R114 
 frameshift_variant 
 c.2782_2785del 
 p.Arg928Ter 
 De novo 
  
 Simplex 
 GEN357R115 
 missense_variant 
 c.5855T>C 
 p.Val1952Ala 
 De novo 
  
 Simplex 
 GEN357R116 
 frameshift_variant 
 c.4144del 
 p.Tyr1382MetfsTer106 
 De novo 
  
  
 GEN357R117 
 stop_gained 
 c.1174C>T 
 p.Arg392Ter 
 De novo 
  
  
 GEN357R118 
 stop_gained 
 c.2086C>T 
 p.Gln696Ter 
 De novo 
  
  
 GEN357R119 
 stop_gained 
 c.592C>T 
 p.Gln198Ter 
 Unknown 
 Not maternal 
  
 GEN357R120 
 frameshift_variant 
 c.1402_1409del 
 p.Gly468ArgfsTer147 
 De novo 
  
  
 GEN357R121 
 frameshift_variant 
 c.1990del 
 p.Ser664ValfsTer20 
 De novo 
  
 Simplex 
 GEN357R122 
 stop_gained 
 c.1762G>T 
 p.Glu588Ter 
 De novo 
  
  
 GEN357R123 
 frameshift_variant 
 c.2231_2232dup 
 p.Pro745AsnfsTer7 
 De novo 
  
 Unknown 
 GEN357R124 
 stop_gained 
 c.4009C>T 
 p.Arg1337Ter 
 De novo 
  
 Unknown 
 GEN357R125 
 stop_gained 
 c.6382C>T 
 p.Arg2128Ter 
 De novo 
  
 Simplex 
 GEN357R126 
 frameshift_variant 
 c.3118del 
 p.Glu1040LysfsTer16 
 De novo 
  
 Simplex 
 GEN357R127 
 splice_site_variant 
 c.2986+1G>C 
 p.? 
 De novo 
  
 Simplex 
 GEN357R128 
 stop_gained 
 c.2528C>A 
 p.Ser843Ter 
 De novo 
  
 Simplex 
 GEN357R129 
 stop_gained 
 c.6274C>T 
 p.Pro2092Ser 
 De novo 
  
 Simplex 
 GEN357R130 
 stop_gained 
 c.-42C>T 
  
 De novo 
  
 Simplex 
 GEN357R131 
 stop_gained 
 c.3646C>T 
 p.Arg1216Ter 
 De novo 
  
 Simplex 
 GEN357R132 
 stop_gained 
 c.4252C>T 
 p.Arg1418Ter 
 De novo 
  
 Simplex 
 GEN357R133 
 frameshift_variant 
 c.6726_6730del 
 p.Gly2243SerfsTer70 
 Familial 
 Maternal 
 Multiplex 
 GEN357R134 
 frameshift_variant 
 c.6091dup 
 p.Gln2031ProfsTer36 
 De novo 
  
 Simplex 
 GEN357R135 
 stop_gained 
 c.403C>T 
 p.Arg135Ter 
 De novo 
  
 Simplex 
 GEN357R136 
 frameshift_variant 
 c.2020_2023del 
 p.Gly674ThrfsTer63 
 De novo 
  
 Simplex 
 GEN357R137 
 copy_number_loss 
  
  
 De novo 
  
 Simplex 
 GEN357R138 
 frameshift_variant 
  
 p.Leu2049TrpfsTer50 
 De novo 
  
 Simplex 
 GEN357R139 
 splice_site_variant 
 c.-252-1G>A 
  
 De novo 
  
 Simplex 
 GEN357R140 
 stop_gained 
 c.2406G>A 
 p.Trp802Ter 
 De novo 
  
 Simplex 
 GEN357R141 
 frameshift_variant 
 c.2131del 
 p.Gln711SerfsTer27 
 De novo 
  
 Simplex 
 GEN357R142 
 frameshift_variant 
 c.3809_3810insATGCTCAT 
 p.Tyr1271CysfsTer66 
 De novo 
  
 Simplex 
 GEN357R143 
 frameshift_variant 
 c.4466del 
 p.Tyr1489SerfsTer82 
 De novo 
  
 Simplex 
 GEN357R144 
 frameshift_variant 
 c.5734del 
 p.Ile1912Ter 
 De novo 
  
 Simplex 
 GEN357R145 
 frameshift_variant 
 c.1832del 
 p.Leu611CysfsTer65 
 De novo 
  
 Simplex 
 GEN357R146 
 missense_variant 
 c.6122A>C 
 p.His2041Pro 
 De novo 
  
 Simplex 
 GEN357R147 
 frameshift_variant 
 c.5148_5434del 
 p.Gly1717MetfsTer4 
 De novo 
  
 Simplex 
 GEN357R148 
 frameshift_variant 
 c.6377dup 
 p.Val2128GlyfsTer7 
 De novo 
  
 Simplex 
 GEN357R149 
 frameshift_variant 
 c.2642_2645de 
 p.Arg881ThrfsTer46 
 De novo 
  
 Simplex 
 GEN357R150 
 frameshift_variant 
 c.3286_3287del 
 p.Leu1096AlafsTer21 
 De novo 
  
 Simplex 
 GEN357R151 
 stop_gained 
 c.4378C>T 
 p.Arg1460Ter 
 Unknown 
  
  
 GEN357R152 
 stop_gained 
 c.1311C>G 
 p.Gly437%3D 
 De novo 
  
  
 GEN357R153 
 stop_gained 
 c.1861C>T 
 p.Gln621Ter 
 De novo 
  
  
 GEN357R154 
 stop_gained 
 c.73C>T 
 p.Gln25Ter 
 De novo 
  
  
 GEN357R155 
 stop_gained 
 c.403C>T 
 p.Arg135Ter 
 De novo 
  
  
 GEN357R156 
 copy_number_loss 
  
  
 Unknown 
  
  
 GEN357R157 
 splice_site_variant 
 c.846+1G>A 
  
 De novo 
  
  
 GEN357R158 
 stop_gained 
 c.2719C>T 
 p.Gln907Ter 
 De novo 
  
  
 GEN357R159 
 frameshift_variant 
 c.3227del 
 p.Lys1076SerfsTer4 
 De novo 
  
  
 GEN357R160 
 copy_number_loss 
  
  
 De novo 
  
 Simplex 
 GEN357R161 
 splice_site_variant 
 c.-167G>A 
  
 De novo 
  
  
 GEN357R162 
 stop_gained 
 c.2611C>T 
 p.Gln871Ter 
 De novo 
  
 Simplex 
 GEN357R163 
 stop_gained 
 c.4194T>G 
 p.Tyr1398Ter 
 De novo 
  
  
 GEN357R164 
 missense_variant 
 c.4645C>T 
 p.Pro1549Ser 
 Unknown 
  
  
 GEN357R165 
 frameshift_variant 
 c.6700_6701del 
 p.Phe2234LeufsTer6 
 De novo 
  
  
 GEN357R166 
 frameshift_variant 
 c.1810del 
 p.Met604TrpfsTer2 
 De novo 
  
 Simplex 
 GEN357R167 
 missense_variant 
 c.2170C>T 
 p.Gln724Ter 
 Unknown 
  
  
 GEN357R168 
 missense_variant 
 c.2170C>T 
 p.Gln724Ter 
 Unknown 
  
  
 GEN357R169 
 missense_variant 
 c.1063A>T 
 p.Asn355Tyr 
 Familial 
 Paternal 
  
 GEN357R170 
 missense_variant 
 c.1063A>T 
 p.Asn355Tyr 
 Familial 
 Paternal 
  
 GEN357R171 
 missense_variant 
 c.1562G>A 
 p.Ser521Asn 
 Unknown 
  
  
 GEN357R172 
 missense_variant 
 c.3838G>A 
 p.Glu1280Lys 
 Unknown 
  
  
 GEN357R173 
 missense_variant 
 c.2176G>A 
 p.Gly726Arg 
 Unknown 
  
  
 GEN357R174 
 missense_variant 
 c.2377G>A 
 p.Asp793Asn 
 Unknown 
  
  
 GEN357R175 
 missense_variant 
 c.2377G>A 
 p.Asp793Asn 
 Unknown 
  
  
 GEN357R176 
 missense_variant 
 c.2846A>G 
 p.Asn949Ser 
 Unknown 
  
  
 GEN357R177 
 missense_variant 
 c.3209A>G 
 p.Gln1070Arg 
 Unknown 
  
  
 GEN357R178 
 missense_variant 
 c.5366G>A 
 p.Ser1789Asn 
 Familial 
 Paternal 
  
 GEN357R179 
 missense_variant 
 c.5366G>A 
 p.Ser1789Asn 
 Familial 
 Paternal 
  
 GEN357R180 
 missense_variant 
 c.3644G>A 
 p.Ser1215Asn 
 Unknown 
  
  
 GEN357R181 
 missense_variant 
 c.3887C>G 
 p.Ala1296Gly 
 Unknown 
  
  
 GEN357R182 
 stop_gained 
 c.1986C>A 
 p.Tyr662Ter 
 De novo 
  
  
 GEN357R183 
 stop_gained 
 c.2206C>T 
 p.Gln736Ter 
 De novo 
  
 Simplex 
 GEN357R184 
 missense_variant 
 c.2132A>G 
 p.Gln711Arg 
 De novo 
  
 Simplex 
 GEN357R185 
 synonymous_variant 
 c.2181G>A 
 p.Met727Ile 
 De novo 
  
 Multiplex 
 GEN357R186 
 inframe_deletion 
 c.188_193del 
 p.Asp63_Gly64del 
 De novo 
  
  
 GEN357R187 
 frameshift_variant 
 c.872_890del 
 p.Leu291ArgfsTer52 
 De novo 
  
  
 GEN357R188 
 missense_variant 
 c.1109T>C 
 p.Met370Thr 
 De novo 
  
  
 GEN357R189 
 missense_variant 
 c.1147G>A 
 p.Val383Met 
 De novo 
  
  
 GEN357R190 
 missense_variant 
 c.1339G>A 
 p.Glu447Lys 
 De novo 
  
  
 GEN357R191 
 splice_site_variant 
 c.1137del 
 p.Leu380CysfsTer40 
 De novo 
  
  
 GEN357R192 
 frameshift_variant 
 c.4496del 
 p.Met1499ArgfsTer72 
 De novo 
  
  
 GEN357R193 
 missense_variant 
 c.3012C>A 
 p.Asn1004Lys 
 De novo 
  
  
 GEN357R194 
 inframe_insertion 
 c.6023_6031dup 
 p.Ala2010_Leu2011insSerGlyAla 
 De novo 
  
  
 GEN357R195 
 missense_variant 
 c.6533G>A 
 p.Trp2178Ter 
 De novo 
  
  
 GEN357R196a 
 synonymous_variant 
 c.1704T>G 
 p.Ala568%3D 
 De novo 
  
 Simplex 
 GEN357R196b 
 frameshift_variant 
 c.1331del 
 p.Ser444CysfsTer8 
 De novo 
  
 Simplex 
 GEN357R197 
 frameshift_variant 
 c.1206_1224del 
 p.Ser403GlufsTer43 
 De novo 
  
  
 GEN357R198 
 missense_variant 
 c.1256T>C 
 p.Leu419Pro 
 De novo 
  
  
 GEN357R199 
 splice_site_variant 
 c.1980G>A 
 p.Pro660%3D 
 De novo 
  
  
 GEN357R200 
 stop_gained 
 c.3274C>T 
 p.Arg1092Ter 
 De novo 
  
 Simplex 
 GEN357R201 
 splice_site_variant 
 c.1883+1G>A 
  
 Familial 
 Maternal 
 Multiplex 
 GEN357R202 
 missense_variant 
 c.2253C>A 
 p.Asn751Lys 
 Familial 
 Paternal 
 Simplex 
 GEN357R203 
 frameshift_variant 
 c.2189dup 
 p.His731ThrfsTer18 
 Unknown 
  
 Simplex 
 GEN357R204 
 splice_site_variant 
 c.2287-2A>G 
 p.? 
 Unknown 
  
 Simplex 
 GEN357R205 
 stop_gained 
 c.583C>T 
 p.Gln195Ter 
 Unknown 
  
  
 GEN357R206 
 stop_gained 
 c.2956G>T 
 p.Gly986Ter 
 Unknown 
  
  
 GEN357R207 
 missense_variant 
 c.727G>C 
 p.Gly243Arg 
 Unknown 
  
 Simplex 
 GEN357R208 
 frameshift_variant 
 c.1638_1647del 
 p.Ala547SerfsTer35 
 De novo 
  
 Simplex 
  et al.  
 GEN357R209 
 frameshift_variant 
 c.5804_5805del 
 p.Phe1935Ter 
 Unknown 
  
 Unknown 
  et al.  
 GEN357R210 
 missense_variant 
 c.4173G>A 
 p.Met1391Ile 
 Unknown 
  
  
  et al.  
 GEN357R211 
 frameshift_variant 
 c.1150_1151del 
 p.Tyr384GlnfsTer233 
 De novo 
  
  
  et al.  

Common

No Common Variants Available
Chromosome
CNV Locus
CNV Type
# of studies
Animal Model
6
Duplication
 1
 
6
Duplication
 1
 
6
Deletion
 1
 
6
Deletion
 4
 
6
Deletion
 3
 
6
Deletion-Duplication
 21
 
6
Deletion
 2
 

Model Summary

Arid1b heterozygous mice showed social behavior impairment, altered vocalization, anxiety-like behavior, neuroanatomical abnormalities, and growth impairment. In the brain, Arid1b haploinsufficiency resulted in changes in the expression of SWI/SNF-regulated genes implicated in neuropsychiatric disorders, including deficiency in insulin-like growth factor 1. Supplementation with growth hormone restored muscle weakness and growth retardation in Arid1b heterozygous mice.

References

Type
Title
Author, Year
Primary
Arid1b haploinsufficient mice reveal neuropsychiatric phenotypes and reversible causes of growth impairment.
Additional
Arid1b Haploinsufficiency Causes Abnormal Brain Gene Expression and Autism-Related Behaviors in Mice.
Additional
Arid1b haploinsufficiency disrupts cortical interneuron development and mouse behavior
Additional
Arid1b haploinsufficiency in parvalbumin- or somatostatin-expressing interneurons leads to distinct ASD-like and ID-like behavior
Additional
Neuroanatomy and behavior in mice with a haploinsufficiency of AT-rich interactive domain 1B (ARID1B) throughout development
Additional
Early postnatal serotonin modulation prevents adult-stage deficits in Arid1b-deficient mice through synaptic transcriptional reprogramming

M_ARID1B_1_KO_HT

Model Type: Genetic
Model Genotype: Heterozygous
Mutation: Cas9 germline gene-editing was used to flox exon5 of Arid1b and generate whole-body Arid1b knockout mice with a deleted exon 5.
Allele Type: Knockout
Strain of Origin: C57BL/6J
Genetic Background:
ES Cell Line:
Mutant ES Cell Line:
Model Source:

M_ARID1B_2_KO_HM

Model Type: Genetic
Model Genotype: Homozygous
Mutation: Cas9 germline gene-editing was used to flox exon5 of Arid1b and generate whole-body Arid1b knockout mice with a deleted exon 5.
Allele Type: Knockout
Strain of Origin: C57BL/6J
Genetic Background:
ES Cell Line:
Mutant ES Cell Line:
Model Source:

M_ARID1B_3_CKO_HT

Model Type: Genetic
Model Genotype: Heterozygous
Mutation: Conditional heterozygous deletion of exon 5 (floxed allele generated by Cas9 mediated editing) of the Arid1b gene using Albumin-cre, in the liver
Allele Type: Conditional loss-of-function
Strain of Origin: C57BL/6J
Genetic Background:
ES Cell Line:
Mutant ES Cell Line:
Model Source:

M_ARID1B_4_CKO_HT

Model Type: Genetic
Model Genotype: Heterozygous
Mutation: Conditional heterozygous deletion of exon 5 (floxed allele generated by Cas9 mediated editing) of the Arid1b gene using Nestin-cre, in neuronal, glial and other cell types in the central and peripheral nervous system
Allele Type: Conditional loss-of-function
Strain of Origin: C57BL/6J
Genetic Background:
ES Cell Line:
Mutant ES Cell Line:
Model Source:

M_ARID1B_5_CKO_HT

Model Type: Genetic
Model Genotype: Heterozygous
Mutation: Conditional heterozygous deletion of exon 5 (floxed allele generated by Cas9 mediated editing) of the Arid1b gene using Ckm-Cre, in skeletal muscle
Allele Type: Conditional loss-of-function
Strain of Origin: C57BL/6J
Genetic Background:
ES Cell Line:
Mutant ES Cell Line:
Model Source:

M_ARID1B_6_KO_HT

Model Type: Genetic
Model Genotype: Heterozygous
Mutation: CRISPR/Cas9 double-nicking method of gene editing was used to generate a deletion in exon 3.
Allele Type: Knockout
Strain of Origin: C57BL/6J
Genetic Background:
ES Cell Line:
Mutant ES Cell Line:
Model Source:

M_ARID1B_7_KO_HT

Model Type: Genetic
Model Genotype: Heterozygous
Mutation: Exon 5 of the Arid1b gene was flanked by loxp sites to create a deletion allele. To generate the global Arid1b KO allele, the knockout first allele was crossed with a CMV-Cre mouse strain (Jackson Llaboratory, #006054), and the resulting Arid1b KO allele in which the neo cassette, one FRT, one loxP, and exon 5 were removed was selected by genomic PCR and sequencing. Lack of Arid1b protein in the null mice was confirmed.
Allele Type: Knockout
Strain of Origin: C57BL6
Genetic Background: NA
ES Cell Line: NA
Mutant ES Cell Line: JM8.N4 strain ((EUCOMM)
Model Source: NA

M_ARID1B_8_CKO_HT

Model Type: Genetic
Model Genotype: Heterozygous
Mutation: Mice with exon 5 of Arid1b floxed were crossed with Dlx5/6-Cre-IRES-EGFP
Allele Type: Conditional loss-of-function
Strain of Origin: C57BL6
Genetic Background: NA
ES Cell Line: NA
Mutant ES Cell Line: JM8.N4 strain ((EUCOMM)
Model Source: 29184203 (Jung et al, Nat Neurosci, 2017)

M_ARID1B_9_CKO_HT

Model Type: Genetic
Model Genotype: Heterozygous
Mutation: Mice with exon 5 of Arid1b floxed were crossed with Nkx2.1-Cre mouse line
Allele Type: Conditional loss-of-function
Strain of Origin: C57BL6
Genetic Background: NA
ES Cell Line: NA
Mutant ES Cell Line: JM8.N4 strain ((EUCOMM)
Model Source: 29184203 (Jung et al, Nat Neurosci, 2017)

M_ARID1B_10_CKO_HT

Model Type: Genetic
Model Genotype: Heterozygous
Mutation: The Arid1b-floxed allele was crossed with the PV-Cre (B6;129P2-Pvalbtm1(cre)Arbr/J; JAX 008069) allele. Exon 5 of Arid1b was floxed as in model M_ARID1B_7_KO_HT.
Allele Type: Conditional loss-of-function
Strain of Origin: C57BL6
Genetic Background: NA
ES Cell Line: NA
Mutant ES Cell Line: JM8.N4 strain ((EUCOMM)
Model Source: 29184203 (Jung et al, Nat Neurosci, 2017)

M_ARID1B_11_CKO_HT

Model Type: Genetic
Model Genotype: Heterozygous
Mutation: The Arid1b-floxed allele was crossed with the SST-Cre (B6N.Cg-Ssttm2.1(cre)Zjh/J; JAX 018973) allele. Exon 5 of Arid1b was floxed as in model M_ARID1B_7_KO_HT.
Allele Type: Conditional loss-of-function
Strain of Origin: C57BL6
Genetic Background: NA
ES Cell Line: NA
Mutant ES Cell Line: JM8.N4 strain ((EUCOMM)
Model Source: 29184203 (Jung et al, Nat Neurosci, 2017)

M_ARID1B_12_KO_HM

Model Type: Genetic
Model Genotype: Homozygous
Mutation: Targeting of Arid1b exon 5 employed Cas9 nuclease (D10A) and single-guide RNAs with spacer sequences for targeting exon 5, resulting in deletion of Chr17.
Allele Type: Knockout
Strain of Origin: C57BL/6NCrl
Genetic Background: C57BL/6N
ES Cell Line: NA
Mutant ES Cell Line: NA
Model Source: Toronto Centre for Phenogenomics

M_ARID1B_13_KO_HT

Model Type: Genetic
Model Genotype: Heterozygous
Mutation: Targeting of Arid1b exon 5 employed Cas9 nuclease (D10A) and single-guide RNAs with spacer sequences for targeting exon 5, resulting in deletion of Chr17.
Allele Type: Knockout
Strain of Origin: C57BL/6NCrl
Genetic Background: C57BL/6N
ES Cell Line: NA
Mutant ES Cell Line: NA
Model Source: Toronto Centre for Phenogenomics

M_ARID1B_14_CKO_HT

Model Type: Genetic
Model Genotype: Heterozygous
Mutation: Mice harboring a cassette containing the Ardi1b gene flanked by loxP and lacZ sites flanked by FRT sites were obtained from EUCOMM. This Arid1b^tm1a(EUCOMM)Hmgu mouse line was crossed with Protamine-Flp mice to remove the cassette and produce Arid1bfl/+ mice. Sert-Cre; Arid1b^fl/+ mice were generated by crossing Sert-Cre female mice from Jackson Laboratory (B6.129(Cg)-Slc6a4^tm1(cre)Xz/J) with Arid1b^fl/fl male mice.
Allele Type: Conditional knockout
Strain of Origin: C57BL/6N; 129S1/Sv-Oca2+ Tyr+ Kitl+
Genetic Background: C57BL/6J
ES Cell Line: JM8.N4; W9.5/W95
Mutant ES Cell Line:
Model Source: Helmholtz Zentrum Muenchen GmbH; Xiaoxi Zhuang

M_ARID1B_1_KO_HT

Category
Entity
Quantity
Experimental Paradigm
Age at Testing
Grip strength1
Decreased
Description: Mutants show hypotonia compared to controls.
Exp Paradigm: NA
 Grip strength test
 Not reported
Brain development1
Decreased
Description: Mutants have hydrocephalus.
Exp Paradigm: NA
 Gross necroscopy
 Not reported
Hippocampal morphology1
Decreased
Description: Mutants show reduction in the size of the dentate gyrus compared to controls.
Exp Paradigm: NA
 Immunohistochemistry
 1.7 months
Neuronal number1
Decreased
Description: Mutants show reduced neuronal number in the cerebral cortex compared to controls.
Exp Paradigm: Tbr1 immunostaining
 Immunohistochemistry
 P0-10
Cortical thickness1
Decreased
Description: Mutants show reduced cortical thickness compared to controls.
Exp Paradigm: Hoechst staining
 Immunohistochemistry
 P0-10
Morphology and size of the corpus callosum1
Decreased
Description: Mutants show reduction in the size of the corpus callosum compared to controls.
Exp Paradigm: NA
 Immunohistochemistry
 1.7 months
Cell proliferation: neural precursors1
Decreased
Description: Mutants show fewer ki67 and brdu positive cells compared to controls.
Exp Paradigm: Adult mice received one brdu injection for five consecutive days and three days following the last injection, brains were fixed and harvested. the subgranular zone of the granular layer of the dentate gyrus was analyzed.
 Immunohistochemistry
 1.7 months
Morphology and size of the corpus callosum1
Decreased
Description: Mutants have agenesis or hypoplasia of corpus callosum compared to controls.
Exp Paradigm: NA
 Histology
 Not reported
Self grooming: perseveration1
Increased
Description: Mutants show increased self grooming compared to controls.
Exp Paradigm: NA
 Grooming behavior assessments
 Adult
Repetitive digging1
Decreased
Description: Mutants buried fewer marbles than controls.
Exp Paradigm: NA
 Marble-burying test
 Not reported
Social interaction: with juveniles1
Decreased
Description: Mutants show decreased social interaction with a juvenile target mouse compared to controls.
Exp Paradigm: NA
 Reciprocal social interaction test
 Adult
Hormone levels1
Decreased
Description: Mutants show decreased levels of igf-1 in the plasma compared to controls.
Exp Paradigm: Mice were not fasted.
 Elisa
 0.9-1.4 months
Ultrasonic vocalization: isolation induced1
Abnormal
Description: Mutants emit usvs of longer duration and abnormal pitch, compared to controls. mutants show no change in the total number of usvs emitted compared to controls.
Exp Paradigm: Number, duration, frequency and amplitude of usvs were recorded.
 Monitoring ultrasonic vocalizations
 P4
Audible vocalization1
Decreased
Description: Mutants show abnormal vocalization compared to controls.
Exp Paradigm: NA
 General observations
 Not reported
Cardiovascular development and function1
Decreased
Description: Mutants had disproportionately small hearts compared to controls.
Exp Paradigm: NA
 Gross necroscopy
 P0-10
Size/growth1
Decreased
Description: Mutants show reduced growth compared to controls.
Exp Paradigm: Body length measurement
 Body length measurement
 0.9-1.2 months
Renal morphology1
Decreased
Description: Mutants had disproportionately small kidneys compared to controls.
Exp Paradigm: NA
 Gross necroscopy
 P0-10
Mortality/lethality: neonatal1
Increased
Description: Mutants showed increased mortality rates compared to controls.
Exp Paradigm: NA
 General observations
 Adult
Size/growth1
Decreased
Description: Mutants show reduced growth compared to controls.
Exp Paradigm: Body weight measurement
 Body weight measurement
 0.9-1.2 months
Anxiety1
Increased
Description: Mutants spend less time in the center of the open field, less time in the open arms on the elevated plus maze, and less time in the brightly lit chamber of the light-dark exploration test, compared to controls.
Exp Paradigm: Open field test
 Open field test
 8 weeks
Anxiety1
Increased
Description: Mutants spend less time in the center of the open field, less time in the open arms on the elevated plus maze, and less time in the brightly lit chamber of the light-dark exploration test, compared to controls.
Exp Paradigm: Light-dark exploration test
 Light-dark exploration test
 8 weeks
Anxiety1
Increased
Description: Mutants spend less time in the center of the open field, less time in the open arms on the elevated plus maze, and less time in the brightly lit chamber of the light-dark exploration test, compared to controls.
Exp Paradigm: Elevated plus maze test
 Elevated plus maze test
 8 weeks
Gene expression1
Abnormal
Description: Mutants show over 150 misregulated genes, including 14 asd genes, in the hippocampus compared to controls. mutants show increased gene expression in components of axon guidance signaling, glutamate receptor signaling, ephrina signaling, nnos signaling, and ephrin receptor signaling, compared to controls.
Exp Paradigm: NA
 Rna sequencing
 2.6-2.7 months
Gene expression1
Increased
Description: Mutants show increased levels of igfbp3 transcript in the liver compared to controls.
Exp Paradigm: NA
 Quantitative pcr (qrt-pcr)
 1.4 months
Targeted expression1
Decreased
Description: Mutants show reduced arid1b mrna transcripts in the liver, whole brain, pituitary gland, dentate gyrus, and hypothalamus, compared to controls.
Exp Paradigm: NA
 Western blot
 Not reported
Gene expression1
Decreased
Description: Mutants show decreased levels of igf-1 and growth hormone receptor (ghr) transcript in the liver compared to controls.
Exp Paradigm: NA
 Quantitative pcr (qrt-pcr)
 1.4 months
Protein-dna complex assembly1
Increased
Description: Misregulated genes in mutants show increased binding with smarca4 (brg1) compared to controls, with positional enrichment at transcription start sites.
Exp Paradigm: NA
 Chromatin immunoprecipitation (chip)
 2.6-2.7 months
Digestive system development1
 No change
 Gross necroscopy
 P0-10
General characteristics1
 No change
 General observations
 P0-10
Cued or contextual fear conditioning: memory of context1
 No change
 Fear conditioning test
 8 weeks
Cued or contextual fear conditioning: memory of cue1
 No change
 Fear conditioning test
 8 weeks
Spatial reference memory1
 No change
 Morris water maze test
 2 weeks
Spatial working memory1
 No change
 Morris water maze test
 2 weeks
Enzyme activity1
 No change
 Enzyme assay
 3.4 months
Gene expression1
 No change
 Quantitative pcr (qrt-pcr)
 1.1-1.5 months
Gene expression1
 No change
 Quantitative pcr (qrt-pcr)
 1.1-1.5 months
General locomotor activity: ambulatory activity1
 No change
 Elevated plus maze test
 8 weeks
General locomotor activity: ambulatory activity1
 No change
 Light-dark exploration test
 8 weeks
General locomotor activity: ambulatory activity1
 No change
 Open field test
 8 weeks
Brain size1
 No change
 Gross necroscopy
 P0-10
Homeostasis1
 No change
 Cell counting
 3.4 months
Hormone levels1
 No change
 Elisa
 1.1-1.5 months
Hormone levels1
 No change
 Elisa
 2 weeks, 0.9-1.4 months
Renal function1
 No change
 Urine/blood electrolyte analysis
 3.4 months
Satiety response1
 No change
 Food intake measurements
 1.9 weeks
Pain or nociception1
 No change
 Foot shock test
 8 weeks
 Not Reported: Circadian sleep/wake cycle, Immune response, Maternal behavior, Neurophysiology, Physiological parameters, Seizure

M_ARID1B_2_KO_HM

Category
Entity
Quantity
Experimental Paradigm
Age at Testing
Cortical thickness1
Decreased
Description: Mutants show reduced cortical thickness compared to controls.
Exp Paradigm: Hoechst staining
 Immunohistochemistry
 P0-10
Neuronal number1
Decreased
Description: Mutants show reduced neuronal number in the cerebral cortex compared to controls.
Exp Paradigm: Tbr1 immunostaining
 Immunohistochemistry
 P0-10
Mortality/lethality: neonatal1
Increased
Description: Mutants were born but died perinataly compared to controls.
Exp Paradigm: NA
 General observations
 P0-10
Targeted expression1
Decreased
Description: Mutants show a lack of arid1b protein in whole brain extracts compared to controls.
Exp Paradigm: NA
 Western blot
 P0
 Not Reported: Circadian sleep/wake cycle, Communications, Emotion, Immune response, Learning & memory, Maternal behavior, Motor phenotype, Neurophysiology, Physiological parameters, Repetitive behavior, Seizure, Sensory, Social behavior

M_ARID1B_3_CKO_HT

Category
Entity
Quantity
Experimental Paradigm
Age at Testing
Size/growth1
 No change
 Body weight measurement
 1.1-3.1 weeks
Hormone levels1
 No change
 Elisa
 1.4-1.5 months
 Not Reported: Circadian sleep/wake cycle, Communications, Emotion, Immune response, Learning & memory, Maternal behavior, Molecular profile, Motor phenotype, Neuroanatomy / ultrastructure / cytoarchitecture, Neurophysiology, Physiological parameters, Repetitive behavior, Seizure, Sensory, Social behavior

M_ARID1B_4_CKO_HT

Category
Entity
Quantity
Experimental Paradigm
Age at Testing
Hormone levels1
Decreased
Description: Mutants show reduced plasma levels of igf-1 compared to controls.
Exp Paradigm: NA
 Elisa
 1.4-1.5 months
Hormone levels1
Decreased
Description: Mutants show reduced plasma levels of gh in spite of the reduced igf1 deficiency, compared to controls.
Exp Paradigm: NA
 Elisa
 Not reported
Size/growth1
Decreased
Description: Mutants show reduced body weight compared to controls.
Exp Paradigm: NA
 Body weight measurement
 1.1-5 weeks
 Not Reported: Circadian sleep/wake cycle, Communications, Emotion, Immune response, Learning & memory, Maternal behavior, Molecular profile, Motor phenotype, Neuroanatomy / ultrastructure / cytoarchitecture, Neurophysiology, Physiological parameters, Repetitive behavior, Seizure, Sensory, Social behavior

M_ARID1B_5_CKO_HT

Category
Entity
Quantity
Experimental Paradigm
Age at Testing
General characteristics1
 No change
 General observations
 Not reported
Size/growth1
 No change
 Body weight measurement
 1.1-3.1 weeks
 Not Reported: Circadian sleep/wake cycle, Communications, Emotion, Immune response, Learning & memory, Maternal behavior, Molecular profile, Motor phenotype, Neuroanatomy / ultrastructure / cytoarchitecture, Neurophysiology, Physiological parameters, Repetitive behavior, Seizure, Sensory, Social behavior

M_ARID1B_6_KO_HT

Category
Entity
Quantity
Experimental Paradigm
Age at Testing
General locomotor activity: ambulatory activity1
Decreased
Description: Mutants travel less distance in elevated plus maze compared to controls.
Exp Paradigm: NA
 Elevated plus maze test
 11-14 weeks
Motor coordination and balance1
Decreased
Description: Mutants show slower moving speed, increased number of movements, increased total duration of movements, increased latency of crossing the beam, and increased number of slips in balance beam test compared to controls.
Exp Paradigm: NA
 Balance beam test
 16-19 weeks
General locomotor activity: ambulatory activity1
Decreased
Description: Mutants show decrease in total distance traveled in open field test compared to controls. mutants showed decrease in vertical activity in open field test compared to controls.
Exp Paradigm: NA
 Open field test
 11-13 weeks
Motor coordination and balance1
Decreased
Description: Mutants show lower latency of falling off in rotarod test than controls.
Exp Paradigm: NA
 Accelerating rotarod test
 12-15 weeks
General locomotor activity1
Decreased
Description: Mutants show decrease in overall activity levels when measured over 1 week compared to controls. when averaged over the last three days, mutants show no change in overall activity levels compared to controls.
Exp Paradigm: Home-cage social interaction test
 Reciprocal social interaction test
 39-42 weeks
Grip strength1
Decreased
Description: Mutants show reduced grip strength compared to controls. mutants also show lower latency to fall compared to controls.
Exp Paradigm: NA
 Wire hang test
 1013 weeks
Brain development1
Abnormal
Description: Mutants have hydrocephalus. mutants with hydrocephalus show a cavity with new vascularization. mutants with hydrocephalus show enlarged lateral ventricles. mutants with hydrocephalus show corruption of the pyramidal cell layer of hippocampus.
Exp Paradigm: NA
 General observations
 8-12 weeks
Circling1
Decreased
Description: Mutants show decrease in stereotypic counts (measured by the number of breaks of the same infrared beam) in open field test compared to controls.
Exp Paradigm: NA
 Open field test
 11-13 weeks
Sensorimotor gating1
Decreased
Description: Mutants show decreased 74 db and 78db prepulse inhibition of startle reflex in response to 120 db acoustic stimulus compared to controls.
Exp Paradigm: NA
 Prepulse inhibition
 13-16 weeks
Foot shock sensitivity1
Decreased
Description: Mutants show decreased freezing behavior during fear conditioning session involving foot shock and travel longer distances both during and immediately after foot shock compared to controls.
Exp Paradigm: NA
 Fear conditioning test
 33-36 weeks
Startle response: acoustic stimulus1
Decreased
Description: Mutants show reduced startle reflex response to 110 db acoustic stimulus compared to controls.
Exp Paradigm: NA
 Startle response test
 13-16 weeks
Social interaction1
Increased
Description: Mutants show increased interaction with unfamiliar mouse during the light, but not the dark, phase of the last 3 days compared to controls.
Exp Paradigm: Home-cage social interaction test
 Reciprocal social interaction test
 39-42 weeks
Size/growth1
Decreased
Description: Mutants show reduced body weight compared to controls. mutants with hydrocephalus show reduced body weight compared to controls. mutants with hydrocephalus show reduced body weight compared to mutants without hydrocephalus.
Exp Paradigm: NA
 Body weight measurement
 10-13 weeks
Depression1
Decreased
Description: Mutants show a decrease in immobility and an increase in distance traveled in porsolt forced swim test compared to controls.
Exp Paradigm: NA
 Forced swim test
 13-16 weeks
Anxiety1
Increased
Description: Mutants show a decrease in number of entries into open or closed arms of elevated plus maze compared to controls. mutants show lower percentage of entries into open arms of elevated plus maze compared to controls. mutants spent less time in open arms of elevated plus maze compared to controls.
Exp Paradigm: NA
 Elevated plus maze test
 11-14 weeks
Spatial reference memory1
Increased
Description: Mutants spend more time around the target in probe trial of barnes maze test than controls.
Exp Paradigm: NA
 Barnes maze test
 16-23 weeks
Cued or contextual fear conditioning: memory of cue: long term recall1
Increased
Description: Mutants show no change in freezing behavior in cued test 1 day after conditioning but do show increase in freezing behavior in cued test 30 days after conditioning compared to controls.
Exp Paradigm: NA
 Fear conditioning test
 33-40 weeks
Cognitive flexibility1
Decreased
Description: Mutants show increased latency in reaching target during reversal task of barnes maze test than controls.
Exp Paradigm: NA
 Barnes maze test
 16-23 weeks
Targeted expression1
Decreased
Description: Mutants show reduction in arid1b protein in the whole brain to about 50% compared to controls.
Exp Paradigm: NA
 Western blot
 Not reported
Gene expression1
Decreased
Description: Mutants show reduction in gene expression compared to controls.
Exp Paradigm: NA
 Rna sequencing
 Not reported
Gene expression1
Decreased
Description: Mutants show reduction in arid1b mrna expression in the whole brain to about 70% compared to controls.
Exp Paradigm: Rna sequencing
 Rna sequencing
 Not reported
Gene expression1
Decreased
Description: Mutants show reduction in arid1b mrna expression in the whole brain to about 70% compared to controls.
Exp Paradigm: Quantitative pcr (qrt-pcr)
 Quantitative pcr (qrt-pcr)
 Not reported
Anxiety1
 No change
 Open field test
 11-13 weeks
Anxiety1
 No change
 Light-dark exploration test
 10-13 weeks
Cued or contextual fear conditioning: memory of context1
 No change
 Fear conditioning test
 33-40 weeks
Cued or contextual fear conditioning: memory of context: long term recall1
 No change
 Fear conditioning test
 33-40 weeks
Cued or contextual fear conditioning: memory of cue1
 No change
 Fear conditioning test
 33-40 weeks
Spatial learning1
 No change
 Barnes maze test
 16-23 weeks
General locomotor activity1
 No change
 Reciprocal social interaction test
 12-14 weeks
General locomotor activity: ambulatory activity1
 No change
 Light-dark exploration test
 10-13 weeks
Brain cytoarchitecture1
 No change
 Histology
 8-9 weeks
Core body temperature1
 No change
 Body temperature measurement
 10-13 weeks
Pain or nociception1
 No change
 Hot plate test
 12-14 weeks
Sensorimotor gating1
 No change
 Prepulse inhibition
 13-16 weeks
Startle response: acoustic stimulus1
 No change
 Startle response test
 13-16 weeks
Social approach1
 No change
 Three-chamber social approach test
 13-15 weeks
Social interaction1
 No change
 Reciprocal social interaction test
 12-14 weeks
Social memory1
 No change
 Three-chamber social approach test
 13-15 weeks
 Not Reported: Circadian sleep/wake cycle, Communications, Immune response, Maternal behavior, Neurophysiology, Physiological parameters, Seizure

M_ARID1B_7_KO_HT

Category
Entity
Quantity
Experimental Paradigm
Age at Testing
General locomotor activity: ambulatory activity1
Decreased
Description: Decrease in total distance traveled
Exp Paradigm: NA
 Open field test
 3-4 months
Motor learning1
Decreased
Description: Decrease in latency to fall during the test phase over 13 trial days
Exp Paradigm: NA
 Accelerating rotarod test
 3-4 months
Synapse density: inhibitory1
Decreased
Description: Decreased levels of slc32a1, gad1, gad2, gphn and arhgef9 mrnas
Exp Paradigm: Gad1; gad2
 Quantitative pcr (qrt-pcr)
 3 months
Neuronal number: inhibitory neurons1
Decreased
Description: Decreased gaba- and parvalbumin-positive neuron densities in the cortex at p91; decreased gaba- and parvalbumin-positive neuron densities in the amy, hip and thm at p28 and p91
Exp Paradigm: Gaba; pv
 Immunohistochemistry
 1month; 3 months
Synaptic neuroreceptors1
Decreased
Description: Decreased ntrk2 transcript levels
Exp Paradigm: Ntrk2
 Quantitative pcr (qrt-pcr)
 3 months
Synapse density: inhibitory1
Decreased
Description: Decreased gad1 protein levels
Exp Paradigm: NA
 Western blot
 3 months
Cortical lamination1
Abnormal
Description: Inhibitory interneurons were abnormally positioned in the cortical layers at p91, no change in normal cux1 and tbr1 layering patterns of excitatory pyramidal neurons at p28 and p91
Exp Paradigm: NA
 Immunohistochemistry
 1 month, 3 months
Synaptic morphology: synaptic cleft length1
Increased
Description: Increase in the cleft width of symmetric synapses
Exp Paradigm: NA
 Electron microscopy
 3 months
Post-synaptic density size: inhibitory synapses1
Decreased
Description: Decreased length of postsynaptic density
Exp Paradigm: NA
 Electron microscopy
 3 months
Cell proliferation: neural precursors1
Decreased
Description: Decreased phosphorylated histone-h3 and brdu positive ventral progenitors in the medial ganglionic eminence
Exp Paradigm: Ph-h3; brdu
 Immunohistochemistry
 E12.5
Synapse density: inhibitory1
Decreased
Description: Decreased numbers of inhibitory synaptic puncta positive to viaat and glutamic acid decarboxylase 2
Exp Paradigm: Viaat; gad2
 Immunohistochemistry
 3 months
Neuronal number: interneurons1
Decreased
Description: Decreased interneuron marker transcript levels, pv
Exp Paradigm: Pv
 Quantitative pcr (qrt-pcr)
 3 months
Miniature post synaptic current frequency: inhibitory1
Decreased
Description: Decreased mipsc frequency
Exp Paradigm: NA
 Whole-cell patch clamp
 3-4 months
Apoptosis: brain cells1
Increased
Description: Increased caspase 3 and tunel positive cells in the mge and lge
Exp Paradigm: Cc3; tunel
 Immunohistochemistry
 E12.5
Self grooming: perseveration1
Increased
Description: Increase in time spent grooming
Exp Paradigm: NA
 Grooming behavior assessments
 3-4 months
Social approach1
Decreased
Description: Decreased preference for unfamiliar mouse over empty cage
Exp Paradigm: NA
 Three-chamber social approach test
 3-4 months
Social memory1
Decreased
Description: Decreased preference for unfamiliar mouse over familiar mouse
Exp Paradigm: NA
 Three-chamber social approach test
 3-4 months
Social interaction1
Decreased
Description: Decrease in time spent in general sniffing, anogenital sniffing, and following behavior
Exp Paradigm: NA
 Reciprocal social interaction test
 3-4 months
Size/growth1
Decreased
Description: Decreased male and female body weight at p14, 21, 28, 56 and 63
Exp Paradigm: NA
 Body weight measurement
 P14, 21, 28, 56 and 63
Anxiety1
Increased
Description: Decrease in time spent in the open arms
Exp Paradigm: NA
 Elevated plus maze test
 3-4 months
Anxiety1
Increased
Description: Decrease in time spent in the center
Exp Paradigm: NA
 Open field test
 3-4 months
Depression1
Increased
Description: Increase in time spent immobile
Exp Paradigm: NA
 Forced swim test
 3-4 months
Reward reinforced choice behavior1
Decreased
Description: Decreased success rate of food retrieval
Exp Paradigm: NA
 T-maze test
 3-4 months
Object recognition memory1
Decreased
Description: Decreased preference for exploring familiar and novel objects
Exp Paradigm: NA
 Novel object recognition test
 3-4 months
Spatial working memory1
Decreased
Description: Increase in escape latency from day 4 of the trials onward
Exp Paradigm: NA
 Morris water maze test
 3-4 months
Spatial reference memory1
Decreased
Description: Decrease in crossings of the platform location
Exp Paradigm: NA
 Morris water maze test
 3-4 months
Chromatin modification: histone modification: open chromatin1
Decreased
Description: Decreased levels of transcriptional activator histone h3 trimethylation at lysine 4 (h3k4me3); increased level of transcriptional repressor, histone h3 trimethylation at lysine 27 (h3k27me3)
Exp Paradigm: NA
 Western blot
 3 months
Signaling: wnt pathway1
Decreased
Description: Decreased protein levels of beta-catenin and its target cyclin d1 in ventral telencephalons in a gene dose-dependent manner; increased level of phospho-beta-catenin
Exp Paradigm: NA
 Western blot
 E15.5
Protein binding1
Decreased
Description: Decreased levels of cbp and pcaf bound to h3k9ac
Exp Paradigm: Cbp; pcaf; h3k9ac
 Co-immunoprecipitation
 3 months
Chromatin modification: histone acetylation1
Decreased
Description: Decreased h3k9ac histone h3 acetylation in the pvalb promoter
Exp Paradigm: NA
 Chromatin immunoprecipitation sequencing (chip-seq)
 3 months
Regulation of gene expression1
Decreased
Description: Decreased initiation of pvalb and ntrk2 gene transcriptions measured by decreased levels of phospho-ctd (ser5) rnapol ii associated with the pvalb and ntrk2 promoters; no change in levels of phospho-ctd (ser5) rnapol ii associated with synt2 promoter
Exp Paradigm: NA
 Chromatin immunoprecipitation sequencing (chip-seq)
 3 months
Protein binding1
Decreased
Description: Decreased binding of the hats and histone h3
Exp Paradigm: Hat; h3
 Proximity ligation assay
 3 months
Targeted expression1
Decreased
Description: Decreased arid1b transcript levels
Exp Paradigm: NA
 Quantitative pcr (qrt-pcr)
 E14.5
Protein-dna complex assembly1
Decreased
Description: Decreased arid1b binding to regions 1 and 2 of the pvalb promoter but no change in arid1b binding to region 3 of the pvalb promoter; no change in arid1b binding in the vicinity of transcription start sites of slc17a7 and dlg4; decreased levels of arid1b binding to gad1 or slc32a1 promoters; decreased association of h3k9ac with the pvalb promoter in regions 1 and 2; no change in the level of h3k9ac binding to pv promoters syt2 and ppargc1a;
Exp Paradigm: NA
 Chromatin immunoprecipitation sequencing (chip-seq)
 3 months
Gene expression: wnt pathway1
Decreased
Description: Decreased transcript levels of ctnnb1 and its target genes and wnt family genes
Exp Paradigm: NA
 Quantitative pcr (qrt-pcr)
 E15.5
Targeted expression1
Decreased
Description: Decreased arid1b protein levels
Exp Paradigm: NA
 Western blot
 E14.5
Protein expression level evidence1
Decreased
Description: Decreased protein level of acetyl-creb-binding protein (cbp), which enhances hat function
Exp Paradigm: Cbp
 Western blot
 3 months
Swim distance1
 No change
 Morris water maze test
 3-4 months
Chromatin modification1
 No change
 Quantitative pcr (qrt-pcr)
 3 months
Enzyme activity1
 No change
 Western blot
 3 months
Protein expression level evidence1
 No change
 Western blot
 3 months
Protein localization: nucleus1
 No change
 Western blot
 3 months
Swimming ability1
 No change
 Morris water maze test
 3-4 months
Glial number1
 No change
 Immunohistochemistry
 1month; 3 months
Neuronal number1
 No change
 Immunohistochemistry
 1 month, 3 months
Neuronal number: excitatory neurons1
 No change
 Immunohistochemistry
 1 month, 3 months
Neuronal number: interneurons1
 No change
 Immunohistochemistry
 1month; 3 months
Neuroreceptor levels: gaba-r: gabaa1
 No change
 Quantitative pcr (qrt-pcr)
 3 months
Synapse density: excitatory1
 No change
 Quantitative pcr (qrt-pcr)
 3 months
Synapse density: excitatory1
 No change
 Immunohistochemistry
 3 months
Synapse density: excitatory1
 No change
 Quantitative pcr (qrt-pcr)
 3 months
Miniature post synaptic current amplitude: excitatory1
 No change
 Whole-cell patch clamp
 3-4 months
Miniature post synaptic current amplitude: inhibitory1
 No change
 Whole-cell patch clamp
 3-4 months
Miniature post synaptic current frequency: excitatory1
 No change
 Whole-cell patch clamp
 3-4 months
 Not Reported:

M_ARID1B_8_CKO_HT

Category
Entity
Quantity
Experimental Paradigm
Age at Testing
General locomotor activity: ambulatory activity1
Decreased
Description: Decrease in total distance traveled
Exp Paradigm: NA
 Open field test
 3-4 months
Neuronal number: interneurons1
Decreased
Description: Decreased number of egfp-positive interneurons; no change in syt2 and ppargc1a mrna levels expressed in interneurons
Exp Paradigm: NA
 Immunohistochemistry
 E12.5, e14.5, e16.5 and e18.5
Neuronal number: inhibitory neurons1
Decreased
Description: Decreased gaba- and parvalbumin-positive neuron densities in the cortex at p91; decreased gaba- and parvalbumin-positive neuron densities in the amy, hip and thm at p28 and p91
Exp Paradigm: NA
 Immunohistochemistry
 1month; 3 months
Neuronal morphology1
Decreased
Description: Decreased number and length of egfp-positive interneuron neurites
Exp Paradigm: NA
 Immunohistochemistry
 3 months
Self grooming: perseveration1
Increased
Description: Increase in time spent grooming
Exp Paradigm: NA
 Grooming behavior assessments
 3-4 months
Social memory1
Decreased
Description: Decreased preference for unfamiliar mouse over familiar mouse
Exp Paradigm: NA
 Three-chamber social approach test
 3-4 months
Social approach1
Decreased
Description: Decreased preference for unfamiliar mouse over empty cage
Exp Paradigm: NA
 Three-chamber social approach test
 3-4 months
Anxiety1
Increased
Description: Increase in time spent in the center and the number of entries into the center
Exp Paradigm: NA
 Open field test
 3-4 months
Object recognition memory1
Decreased
Description: Decreased preference for exploring familiar and novel objects
Exp Paradigm: NA
 Novel object recognition test
 3-4 months
Neuronal migration1
 No change
 Immunohistochemistry
 E14.5, e18.5
 Not Reported:

M_ARID1B_9_CKO_HT

Category
Entity
Quantity
Experimental Paradigm
Age at Testing
Neuronal number: inhibitory neurons1
Decreased
Description: Decreased gaba- and parvalbumin-positive neuron densities in the cortex at p91; decreased gaba- and parvalbumin-positive neuron densities in the amy, hip and thm at p28 and p91
Exp Paradigm: NA
 Immunohistochemistry
 1month; 3 months
Self grooming: perseveration1
Increased
Description: Increase in time spent grooming
Exp Paradigm: NA
 Grooming behavior assessments
 3-4 months
 Not Reported:

M_ARID1B_10_CKO_HT

Category
Entity
Quantity
Experimental Paradigm
Age at Testing
Synapse density: inhibitory1
Decreased
Description: Decreased viaat protein levels
Exp Paradigm: Viaat
 Western blot
 2-4 months
Synapse density: inhibitory1
Decreased
Description: Decreased inhibitory synaptic puncta
Exp Paradigm: Viaat; gad2
 Immunohistochemistry
 2-4 months
Social memory1
Decreased
Description: No preference for unfamiliar mouse over familiar mouse
Exp Paradigm: NA
 Three-chamber social approach test
 2-4 months
Social approach1
Decreased
Description: No preference for unfamiliar mouse over empty cage
Exp Paradigm: NA
 Three-chamber social approach test
 2-4 months
Depression1
Increased
Description: Increase in time spent immobile
Exp Paradigm: NA
 Tail suspension test
 2-4 months
Anxiety1
Increased
Description: Decrease in time spent in the open arms
Exp Paradigm: NA
 Elevated plus maze test
 2-4 months
Anxiety1
Increased
Description: Decrease in time spent in the center
Exp Paradigm: NA
 Open field test
 2-4 months
Targeted expression1
Decreased
Description: Decreased cortical expression of arid1b in pv interneurons
Exp Paradigm: Pv
 Immunohistochemistry
 2-4 months
Size/growth1
 No change
 Body weight measurement
 2-4 months
Object recognition memory1
 No change
 Novel object recognition test
 2-4 months
Spatial reference memory1
 No change
 Morris water maze test
 2-4 months
Spatial working memory1
 No change
 Morris water maze test
 2-4 months
Swim distance1
 No change
 Morris water maze test
 2-4 months
General locomotor activity1
 No change
 Open field test
 2-4 months
General locomotor activity: ambulatory activity1
 No change
 Open field test
 2-4 months
Motor learning1
 No change
 Accelerating rotarod test
 2-4 months
Swimming ability1
 No change
 Morris water maze test
 2-4 months
Brain size1
 No change
 Measurement of tissue weight
 2-4 months
Synapse density: excitatory1
 No change
 Immunohistochemistry
 2-4 months
Self grooming: perseveration1
 No change
 Grooming behavior assessments
 2-4 months
 Not Reported:

M_ARID1B_11_CKO_HT

Category
Entity
Quantity
Experimental Paradigm
Age at Testing
Motor learning1
Decreased
Description: No increase in the latency to fall during the testing phase and decrease latency to fall during the testing phase
Exp Paradigm: NA
 Accelerating rotarod test
 2-4 months
Synapse density: inhibitory1
Decreased
Description: Decreased inhibitory synaptic puncta
Exp Paradigm: Viaat; gad2
 Immunohistochemistry
 2-4 months
Synapse density: inhibitory1
Decreased
Description: Decreased viaat protein levels
Exp Paradigm: Viaat
 Western blot
 2-4 months
Self grooming: perseveration1
Increased
Description: Increased time spent grooming
Exp Paradigm: NA
 Grooming behavior assessments
 2-4 months
Spatial working memory1
Decreased
Description: Increase in the latency to reach the platform during the 10 day trial phase; no change in the latency to reach the platform during the first three days
Exp Paradigm: NA
 Morris water maze test
 2-4 months
Spatial reference memory1
Decreased
Description: Decrease in the number of platform crossings
Exp Paradigm: NA
 Morris water maze test
 2-4 months
Object recognition memory1
Decreased
Description: No preference for novel object
Exp Paradigm: NA
 Novel object recognition test
 2-4 months
Targeted expression1
Decreased
Description: Decreased cortical expression of arid1b in sst interneurons
Exp Paradigm: Sst
 Immunohistochemistry
 2-4 months
Size/growth1
 No change
 Body weight measurement
 2-4 months
Anxiety1
 No change
 Open field test
 2-4 months
Anxiety1
 No change
 Elevated plus maze test
 2-4 months
Depression1
 No change
 Tail suspension test
 2-4 months
Swim distance1
 No change
 Morris water maze test
 2-4 months
General locomotor activity1
 No change
 Open field test
 2-4 months
General locomotor activity: ambulatory activity1
 No change
 Open field test
 2-4 months
Swimming ability1
 No change
 Morris water maze test
 2-4 months
Brain size1
 No change
 Measurement of tissue weight
 2-4 months
Synapse density: excitatory1
 No change
 Immunohistochemistry
 2-4 months
Social approach1
 No change
 Three-chamber social approach test
 2-4 months
Social memory1
 No change
 Three-chamber social approach test
 2-4 months
 Not Reported:

M_ARID1B_12_KO_HM

Category
Entity
Quantity
Experimental Paradigm
Age at Testing
Mortality/lethality: embryonic1
Increased
Description: Homozygous mutants died during early embryonic development
 General observations
 E1-E10
Targeted expression2
Decreased
Description: There was undetectable levels of ARID1B protein in Arid1bâ??/â?? (KO) mice at P0.
 Western blot
 P0
 Not Reported:

M_ARID1B_13_KO_HT

Category
Entity
Quantity
Experimental Paradigm
Age at Testing
Hypoactivity1
Increased
Description: Hypoactivity in horizontal and vertical activity
 Open field test
 Adult
Spinal reflex1
Decreased
Description: Delayed motor reflex
 General observations
 P2-12
Grip strength1
Decreased
Description: Decreased maximum forelimb force exerted, indicating reduced muscle strength
 Grip strength test
 17 weeks
Negative Geotaxis1
Decreased
Description: Decreased negative geotaxis at p12
 Negative geotaxis test
 P2-12
General locomotor activity: Ambulatory activity1
Decreased
Description: Decreased total distance traversed in the novel open field
 Open field test
 Adult, 11 weeks
Motor coordination and balance1
Decreased
Description: Decreased limb coordination
 General observations
 P2-12
General locomotor activity1
Decreased
Description: Increased time to traverse out of circle were detected in
 Distance traveled in a grid
 P2-12
Brain anatomy1
Decreased
Description: Decreased volume of brain stem, more widespread effect on female than males; sexual dimorphism
 Magnetic resonance imaging (MRI)
 Adult
Morphology and size of the corpus callosum1
Decreased
Description: Decrease in relative volume
 Magnetic resonance imaging (MRI)
 Adult
Size of cerebral ventricles: lateral ventricle2
Increased
Description: There were moderate increases in the areas of lateral ventricles in Arid1b heterozygous mice compared to wildtype mice.
 Histology
 6 weeks
Cerebellar morphology1
Decreased
Description: Decreased cerebellar volume; affected structures diverged earlier in the males, with differences seen as early as pnd7, while in females, the differences did not emerge until after pnd40
 Magnetic resonance imaging (MRI)
 P2-2 months
Morphology of the basal ganglia: Striatum: Caudoputamen1
Decreased
Description: Decrease in relative volume
 Magnetic resonance imaging (MRI)
 Adult
Hydrocephaly1
Increased
Description: Increased incidence of hydrocephaly; equal rates between sexes
 Histology
 P0-P21
Size of cerebral ventricles: Lateral ventricle1
Increased
Description: Entire brain ventricular system was enlarged throughout development, particularly lateral ventricles
 Histology
 P0-P21
Brain size1
Decreased
Description: Decreased head width at p12 but not in adult
 Macroscopic analysis
 P2-12, adult
Morphology of telencephalon1
Decreased
Description: Smaller hindbrain
 Magnetic resonance imaging (MRI)
 Adult
Hippocampal morphology1
Increased
Description: Increased hippocampal volume; affected structures diverged earlier in the males, with differences seen as early as pnd7, while in females, the differences did not emerge until after pnd40
 Magnetic resonance imaging (MRI)
 P2-2 months
Olfactory bulb morphology1
Decreased
Description: Olfactory bulb was smaller
 Magnetic resonance imaging (MRI)
 Adult
Brain morphology1
Abnormal
Description: White matter fiber tracts showed differences; affected structures diverged earlier in the males, with differences seen as early as pnd7, while in females, the differences did not emerge until after pnd40
 Magnetic resonance imaging (MRI)
 P2-2 months, Adult
Morphology and size of the corpus callosum2
Decreased
Description: There were moderate decreases in corpus callosal areas in Arid1b heterozygous mice compared to wildtype mice.
 Histology
 6 weeks
Synapse density2
Decreased
Description: There was a decrease (30%) in the density of the postsynaptic density (PSD) in the prelimbic layer 2/3 region of the mPFC of juvenile Arid1b heterozygous mice.
 Electron microscopy
 3 weeks
Dendritic architecture: dendritic tree complexity2
Increased
Description: There were moderate increases in apical (not basal) dendritic complexity in hippocampal neurons.
 Sholl analysis
 6 weeks
Neocortex morphology1
Decreased
Description: Decrease in relative volume
 Magnetic resonance imaging (MRI)
 Adult
Miniature post synaptic current frequency: excitatory2
Decreased
Description: Layer 2/3 pyramidal neurons in the prelimbic region of the mPFC in juvenile Arid1b heterozygous mice showed markedly decreased frequency (50%) of miniature excitatory post-synaptic currents (mEPSCs). Layer 5 Arid1b heterozygous pyramidal neurons showed no alterations in mEPSCs. Layer 6 Arid1b heterozygous pyramidal neurons also showed unaltered mEPSCs. In the mutant anterior cingulate cortex (ACC), layer 2/3 or layer 5 pyramidal neurons did not show alterations in mEPSCs. The frequency of mEPSCs was decreased in layer 2/3 pyramidal neurons of adult Arid1b heterozygous mice.
 Whole-cell patch clamp
 3 weeks
Self grooming2
Increased
Description: Arid1b heterozygous mice displayed enhanced repetitive self-grooming.
 Home cage behavior
 2-3 months
Vertical jumping or back flipping1
Decreased
Description: Decreased vertical activity
 Open field test
 Adult
Seizures1
Increased
Description: Faster loss of righting reflexes, tonic extension and death upon ptz injection
 Observation of chemically induced seizures
 18-19 weeks
Social interaction2
Decreased
Description: Arid1b heterozygous mice showed decreased direct social interaction in the dyadic social interaction test, measured by decreased total sniffing, nose-to-nose interaction, nose-to-body interaction and following time.
 Reciprocal social interaction test
 2-3 months
Social interaction: opposite sex1
Decreased
Description: Decreased preference for opposite sex
 Reciprocal social interaction test
 16 weeks
Ultrasonic vocalization: Isolation induced1
Decreased
Description: Fewer calls each day; delayed shift in day of peak calls, no sex differences
 Monitoring ultrasonic vocalizations
 PND2-14
Ultrasonic vocalization: Isolation induced2
Decreased
Description: Arid1b heterozygous mice showed decreased number of ultrasonic vocalization calls when separated from their mothers.
 Monitoring ultrasonic vocalizations
 P3-11
Ultrasonic vocalization: Interaction induced: opposite sex stimulus1
Decreased
Description: Decreased usv in the presence of estrus female
 Monitoring ultrasonic vocalizations
 Adult
Size/growth1
Decreased
Description: Decreased body length at p12
 Body length measurement
 P2-12
Size/growth2
Decreased
Description: Arid1b heterozygous mice showed moderately decreased body weights from P9 compared to wildtype mice.
 Body weight measurement
 P3-P49
Developmental trajectory1
Decreased
Description: Delayed developmental milestones, no sex differences; divergence in physical metrics were detected in mid and late development
 General observations
 P2-P14
Size/growth1
Decreased
Description: Decreased body weight at p12 through adult
 Body weight measurement
 P2-12, Adult
Exploratory activity2
Decreased
Description: Arid1b heterozygous mice exhibited lower levels of exploratory activity.
 Novel object recognition test
 2-3 months
Anxiety1
Increased
Description: Spent less time on the open arm and had fewer total entries calculated by the open and closed arm entries summed
 Elevated plus maze test
 10 weeks
Object recognition memory1
Decreased
Description: No increase in time spent exploring novel object over familiar object
 Novel object recognition test
 13 weeks
Visual discrimination learning1
Increased
Description: Mutant mice required significantly fewer sessions to a stringent criterion of completing at least 30 trials, at an accuracy of 80% or higher, on two consecutive days
 Operant self-learning paradigm
 9-17 weeks
Protein phosphorylation2
Decreased
Description: p-MEF2A levels were decreased in the whole brain of Arid1b heterozygous mice relative to wildtype mice. p-beta-catenin was unchanged.
Exp Paradigm: p-MEF2A
 Western blot
 3-6 months
Differential gene expression2
Abnormal
Description: The numbers of differentially expressed genes (DEGs) with significant FDR values (<0.05) in the P3 whole-brain and P10 whole-brain/mPFC transcripts were small making it difficult to identify significant biological functions.
Exp Paradigm: gene set enrichment analysis (GSEA)
 RNA sequencing
 P3-P10
Targeted expression2
Decreased
Description: There was reduced levels of ARID1B protein in Arid1b heterozygous (HT) mice at P0.
 Western blot
 P0
Protein expression level evidence2
Decreased
Description: HDAC4 levels were decreased in the whole brain and P1 fraction of Arid1b heterozygous mice relative to wildtype mice.
Exp Paradigm: HDAC4
 Western blot
 3-6 months
Differential gene expression1
Abnormal
Description: 373 genes were differentially expressed, gene ontology (go) terms identified axon and synaptic terms among upregulated genes and transcriptional regulation and rna processing enriched among downregulated genes, gse169491
 RNA sequencing
 5 months
Targeted expression1
Decreased
Description: Reduced arid1b transcript
 Quantitative PCR (qRT-PCR)
 Adult
Protein expression level evidence2
Increased
Description: Arc levels were increased in whole brain of Arid1b heterozygous mice relative to wildtype mice. SynGAP levels exhibited a non-significant trend of increase (p=0.06) in Arid1b heterozygous mice relative to wildtype mice.
Exp Paradigm: SynGAP, Arc
 Western blot
 3-6 months
Targeted expression1
Decreased
Description: Reduced arid1b protein
 Western blot
 Adult
Differential gene expression: ASD risk genes2
Abnormal
Description: A ratio comparison of Arid1b heterozygotes to wildtype (Arid1b/WT) whole-brain transcripts at P10 showed negative enrichment for ASD-related gene sets that are upregulated in ASD, positive enrichment for gene sets that are downregulated in ASD, and positive enrichment for ASD-risk gene sets that are thought to be downregulated in ASD through loss-of-function mutations. At P10, Arid1b/WT whole-brain transcripts were positively enriched for neuron (both glutamatergic and GABAergic)-related gene sets and negatively enriched for microglia-related gene sets, a strongly reverse-ASD pattern. Whole brain transcripts at P3 indicated a reverse-ASD pattern that are largely similar to those observed in P10 Arid1b/ WT whole-brain transcript. In the mPFC at P10, there wa
Exp Paradigm: gene set enrichment analysis (GSEA)
 RNA sequencing
 P3-P10
Mortality/lethality2
 No change
 General observations
 P0
Anxiety2
 No change
 Light-dark exploration test
 2-3 months
Anxiety2
 No change
 Elevated plus maze test
 2-3 months
Anxiety2
 No change
 Open field test
 2-3 months
Anxiety1
 No change
 Light-dark exploration test
 10 weeks
Exploratory activity: Habituation1
 No change
 Open field test
 Adult
Cognitive flexibility1
 No change
 Operant self-learning paradigm
 12 weeks
Cued or contextual fear conditioning: Memory of context1
 No change
 Fear conditioning test
 18 weeks
Cued or contextual fear conditioning: Memory of cue1
 No change
 Fear conditioning test
 18 weeks
Object recognition memory2
 No change
 Novel object recognition test
 2-3 months
Gait1
 No change
 Footprint analysis
 Adult
General locomotor activity: ambulatory activity2
 No change
 Open field test
 2-3 months
Limb Posture1
 No change
 General observations
 Adult
Righting response1
 No change
 Righting reflex test
 P2-12
Brain morphology2
 No change
 Histology
 6 weeks
Cortical lamination2
 No change
 Immunohistochemistry
 6 weeks
Cortical thickness2
 No change
 Histology
 6 weeks
Neuronal number2
 No change
 Immunohistochemistry
 6 weeks
Neuronal number: Inhibitory neurons2
 No change
 In situ hybridization (ISH)
 P7, P14, P56
Post-synaptic density size2
 No change
 Electron microscopy
 3 weeks
Size of cerebral ventricles: third ventricle2
 No change
 Histology
 6 weeks
Synapse density: Inhibitory2
 No change
 Electron microscopy
 2 months
Synaptic morphology: synaptic cleft length2
 No change
 Electron microscopy
 3 weeks
Action potential property: firing rate2
 No change
 Whole-cell patch clamp
 2 months
Action potential property: threshold2
 No change
 Whole-cell patch clamp
 2 months
Intrinsic membrane properties2
 No change
 Whole-cell patch clamp
 2 months
Membrane potential2
 No change
 Whole-cell patch clamp
 2 months
Miniature post synaptic current amplitude: excitatory2
 No change
 Whole-cell patch clamp
 3 weeks
Miniature post synaptic current amplitude: inhibitory2
 No change
 Whole-cell patch clamp
 3 weeks
Miniature post synaptic current frequency: inhibitory2
 No change
 Whole-cell patch clamp
 3 weeks
Presynaptic function: paired-pulse facilitation2
 No change
 Whole-cell patch clamp
 2 months
Synaptic transmission: inhibitory2
 No change
 Whole-cell patch clamp
 2 months
Circling1
 No change
 Novel cage test
 Adult
Self grooming1
 No change
 Grooming behavior assessments
 15 weeks
Core body temperature1
 No change
 Body temperature measurement
 PND2-14
Allogrooming1
 No change
 Grooming behavior assessments
 15 weeks
Social approach2
 No change
 Three-chamber social approach test
 2-3 months
Social approach1
 No change
 Three-chamber social approach test
 15 weeks
Social memory2
 No change
 Three-chamber social approach test
 2-3 months
 Not Reported:

M_ARID1B_14_CKO_HT

Category
Entity
Quantity
Experimental Paradigm
Age at Testing
Self grooming1
 No change
 Home cage behavior
 6 weeks
Social interaction1
 No change
 Reciprocal social interaction test
 6 weeks
 Not Reported:


Interactor Symbol Interactor Name Interactor Organism Entrez ID Uniprot ID Interaction Type Evidence Reference
BCL7A B-cell CLL/lymphoma 7A 605 Q4VC05 IP; LC-MS/MS
Huttlin EL , et al. 2015
BCL7C B-cell CLL/lymphoma 7 protein family member C 9274 Q8WUZ0-2 IP; LC-MS/MS
Huttlin EL , et al. 2015
CHD8 chromodomain helicase DNA binding protein 8 57680 Q9HCK8 CHIP-seq
Cotney J , et al. 2015
CUL2 cullin 2 8453 Q13617 IP; MS; in vitro kinase assay
Li XS , et al. 2010
DPF2 D4, zinc and double PHD fingers family 2 5977 Q92785 IP; LC-MS/MS
Huttlin EL , et al. 2015
DPF3 D4, zinc and double PHD fingers, family 3 8110 Q92784 IP; LC-MS/MS
Huttlin EL , et al. 2015
HIST2H2BE histone cluster 2, H2be 8349 Q16778 Ubiquitination assay
Li XS , et al. 2010
KDM5B lysine (K)-specific demethylase 5B 10765 Q9UGL1 Phage display
Zhou W , et al. 2009
NAGK N-acetylglucosamine kinase 55577 Q9UJ70 IP; LC-MS/MS
Huttlin EL , et al. 2015
PRMT5 protein arginine methyltransferase 5 10419 O14744 IP; LC-MS/MS
Inoue H , et al. 2010
QPRT Nicotinate-nucleotide pyrophosphorylase [carboxylating] 23475 Q15274 IP; LC-MS/MS
Huttlin EL , et al. 2015
RBFOX1 RNA binding protein, fox-1 homolog (C. elegans) 1 54715 Q9NWB1 qRT-PCR; IP/WB; in situ hybridization; in silico target prediction
Fogel BL , et al. 2012
SMARCA2 SWI/SNF related, matrix associated, actin dependent regulator of chromatin, subfamily a, member 2 6595 P51531 IP; in vitro kinase assay
Wang X , et al. 2004
SMARCA4 SWI/SNF related, matrix associated, actin dependent regulator of chromatin, subfamily a, member 4 6597 A7E2E1 IP; LC-MS/MS; IP/WB
Y2H; IP/WB
Hurlstone AF , et al. 2002
SMARCB1 SWI/SNF related, matrix associated, actin dependent regulator of chromatin, subfamily b, member 1 6598 Q12824 IP; MS; in vitro kinase assay
et al.
SMARCC1 SWI/SNF related, matrix associated, actin dependent regulator of chromatin, subfamily c, member 1 6599 Q58EY4 IP/WB; IEX-HPLC/sucrose gradient/isoelectric focus/MS
IP; in vitro kinase assay
IP; LC-MS/MS; IP/WB
Wang X , et al. 2004
SMARCC2 SWI/SNF related, matrix associated, actin dependent regulator of chromatin, subfamily c, member 2 6601 Q8TAQ2 IP; LC-MS/MS; IP/WB
Inoue H , et al. 2010
SMARCC2 SWI/SNF related, matrix associated, actin dependent regulator of chromatin, subfamily c, member 2 6601 Q8TAQ2 IP; LC-MS/MS
Huttlin EL , et al. 2015
SMARCD1 SWI/SNF related, matrix associated, actin dependent regulator of chromatin, subfamily d, member 1 6602 Q96GM5 IP; LC-MS/MS
Inoue H , et al. 2010
SMARCD1 SWI/SNF related, matrix associated, actin dependent regulator of chromatin, subfamily d, member 1 6602 Q96GM5 IP; LC-MS/MS
Huttlin EL , et al. 2015
SMARCE1 SWI/SNF related, matrix associated, actin dependent regulator of chromatin, subfamily e, member 1 6605 Q969G3 IP; MS; in vitro kinase assay
Mak AB , et al. 2010
SMARCE1 SWI/SNF related, matrix associated, actin dependent regulator of chromatin, subfamily e, member 1 6605 Q969G3 IP; LC-MS/MS
Huttlin EL , et al. 2015
SS18 synovial sarcoma translocation, chromosome 18 6760 Q15532 TAP; MS
Middeljans E , et al. 2012
SS18 synovial sarcoma translocation, chromosome 18 6760 Q15532 IP; LC-MS/MS
Huttlin EL , et al. 2015
TCEB1 transcription elongation factor B (SIII), polypeptide 1 (15kDa, elongin C) 6921 Q15369 IP; MS; in vitro kinase assay
Li XS , et al. 2010
TOP3B topoisomerase (DNA) III beta 8940 O95985 HITS-CLIP
Xu D , et al. 2013
FMR1 fragile X mental retardation 1 14265 P35922 HITS-CLIP
Darnell JC , et al. 2011

HELP
Copyright © 2017 MindSpec, Inc.