HELP     Sign In
Search

Relevance to Autism

Two de novo loss-of-function (LoF) variants in the ASH1L gene were identified in ASD probands from the Simons Simplex Collection (PMIDs 24267886, 25363768), while a third de novo LoF variant in this gene was identified in one ASD proband from 2,270 trios screened by the Autism Sequencing Consortium in De Rubeis et al., 2014 (PMID 25363760). Analysis of rare coding variation in 3,871 ASD cases and 9,937 ancestry-matched or paternal controls from the Autism Sequencing Consortium (ASC) in this report identified ASH1L as a gene meeting high statistical significance with a 0.05 < FDR 0.1, meaning that this gene had a 90% chance of being a true autism gene. A fourth de novo LoF variant in the ASH1L gene was identified in an ASD proband in Tammimies et al., 2015 (PMID 26325558). 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). An additional de novo LoF variant in ASH1L was identifed in a proband from the Pediatric Cardiac Genetics Consortium who presented with ASD, developmental delay, and intellectual disability in addition to congenital heart disease (Homsy et al., 2015). De novo and inherited missense variants that were predicted to be deleterious were identified in ASD probands from the Autism Clinical and Genetic Resources in China (ACGC) cohort in (PMID 27824329). De novo LoF variants in ASH1L have also been identified in individuals with intellectual disability in Stessman et al., 2017 (PMID 28191889) and Okamoto et al., 2017 (PMID 28394464). 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 ASH1L as a gene reaching exome-wide significance (P < 2.5E-06).

Molecular Function

This gene encodes a member of the trithorax group of transcriptional activators. The encoded product functions as a histone methyltransferase specifically methylating 'Lys-36' of histone H3 (H3K36me).

External Links

        

References

Type
Title
Type of Disorder
Associated Disorders
Author, Year
Primary
Coexpression networks implicate human midfetal deep cortical projection neurons in the pathogenesis of autism.
ASD
Positive Association
ASH1L may contribute to the risk of Tourette syndrome: Combination of family-based analysis and case-control study
Tourette syndrome
Support
De novo genic mutations among a Chinese autism spectrum disorder cohort.
ASD
Support
ASD
Support
Characterization of intellectual disability and autism comorbidity through gene panel sequencing.
ID, ASD or autistic traits
Support
High prevalence of multilocus pathogenic variation in neurodevelopmental disorders in the Turkish population
DD, epilepsy/seizures
Support
Molecular Diagnostic Yield of Chromosomal Microarray Analysis and Whole-Exome Sequencing in Children With Autism Spectrum Disorder.
ASD
Support
Integrating de novo and inherited variants in 42
ASD
Support
Inherited and multiple de novo mutations in autism/developmental delay risk genes suggest a multifactorial model.
ASD
Support
Contribution of Multiple Inherited Variants to Autism Spectrum Disorder (ASD) in a Family with 3 Affected Siblings
ASD
Support
Neural Hyperactivity Is a Core Pathophysiological Change Induced by Deletion of a High Autism Risk Gene Ash1L in the Mouse Brain
ASD
Support
De novo loss-of-function variants of ASH1L are associated with an emergent neurodevelopmental disorder.
DD, ID, ADHD
Support
ASD
OCD, ID, epilepsy/seizures
Support
Loss of histone methyltransferase ASH1L in the developing mouse brain causes autistic-like behaviors
ASD
Support
Rare coding variants in ten genes confer substantial risk for schizophrenia
Schizophrenia
Support
Histone Lysine Methylases and Demethylases in the Landscape of Human Developmental Disorders.
DD, ID
ASD, epilepsy/seizures
Support
ASD
DD, ID
Support
Large-scale targeted sequencing identifies risk genes for neurodevelopmental disorders
ASD, DD
Support
Counteracting epigenetic mechanisms regulate the structural development of neuronal circuitry in human neurons
Support
Novel MCA/ID syndrome with ASH1L mutation.
ID
Microcephaly, dysmorphic features, MCA
Support
Phenotypic and genetic spectrum of epilepsy with myoclonic atonic seizures
ASD, ADHD, ID, epilepsy/seizures
Support
Rare variants in the outcome of social skills group training for autism
ASD
Support
Whole genome sequencing resource identifies 18 new candidate genes for autism spectrum disorder
ASD
Support
ASD
ID
Support
Large-Scale Exome Sequencing Study Implicates Both Developmental and Functional Changes in the Neurobiology of Autism
ASD
Support
Deficiency of autism risk factor ASH1L in prefrontal cortex induces epigenetic aberrations and seizures
ASD
Epilepsy/seizures
Recent Recommendation
The contribution of de novo coding mutations to autism spectrum disorder
ASD
Recent Recommendation
Synaptic, transcriptional and chromatin genes disrupted in autism.
ASD
Recent Recommendation
ASH1L haploinsufficiency results in autistic-like phenotypes in mice and links Eph receptor gene to autism spectrum disorder
ASD
Recent Recommendation
Mutations in ASH1L confer susceptibility to Tourette syndrome.
TS
Recent Recommendation
Targeted sequencing identifies 91 neurodevelopmental-disorder risk genes with autism and developmental-disability biases.
ASD, DD, ID
Recent Recommendation
De novo mutations in congenital heart disease with neurodevelopmental and other congenital anomalies.
Congenital heart disease (CHD)
ASD, DD, ID
Recent Recommendation
Low load for disruptive mutations in autism genes and their biased transmission.
ASD

Rare

Variant ID
Variant Type
Allele Change
Residue Change
Inheritance Pattern
Inheritance Association
Family Type
Author, Year
 GEN642R001 
 frameshift_variant 
 c.7764_7765insCAAGG 
 p.Lys2589GlnfsTer8 
 De novo 
  
 Simplex 
 GEN642R002 
 stop_gained 
 c.6427G>T 
 p.Glu2143Ter 
 De novo 
  
 Simplex 
 GEN642R003 
 frameshift_variant 
 c.3744_3745del 
 p.His1248GlnfsTer5 
 De novo 
  
 Simplex 
 GEN642R004 
 missense_variant 
 c.5260A>T 
 p.Ser1754Cys 
 Familial 
 Paternal 
 Simplex 
 GEN642R005 
 missense_variant 
 c.5260A>T 
 p.Ser1754Cys 
 Familial 
 Maternal 
 Simplex 
 GEN642R006 
 missense_variant 
 c.3857A>T 
 p.Asp1286Val 
 Familial 
 Maternal 
 Simplex 
 GEN642R007 
 missense_variant 
 c.3854C>T 
 p.Pro1285Leu 
 Familial 
 Maternal 
 Simplex 
 GEN642R008 
 missense_variant 
 c.205G>T 
 p.Ala69Ser 
 Familial 
 Maternal 
 Simplex 
 GEN642R009 
 missense_variant 
 c.8375T>G 
 p.Ile2792Ser 
 Familial 
 Maternal 
 Simplex 
 GEN642R010 
 missense_variant 
 c.4361C>G 
 p.Thr1454Arg 
 Familial 
 Paternal 
 Simplex 
 GEN642R011 
 missense_variant 
 c.7598G>A 
 p.Arg2533His 
 Familial 
 Maternal 
 Simplex 
 GEN642R012 
 stop_gained 
 c.817A>T 
 p.Lys273Ter 
 De novo 
  
 Unknown 
 GEN642R013 
 missense_variant 
 c.7094A>G 
 p.Asn2365Ser 
 Unknown 
  
 Unknown 
 GEN642R014 
 missense_variant 
 c.5260A>T 
 p.Ser1754Cys 
 Unknown 
  
 Unknown 
 GEN642R015 
 missense_variant 
 c.4927C>T 
 p.Arg1643Trp 
 Unknown 
  
 Unknown 
 GEN642R016 
 missense_variant 
 c.4478G>A 
 p.Arg1493His 
 Unknown 
  
 Unknown 
 GEN642R017 
 missense_variant 
 c.4008C>A 
 p.Asp1336Glu 
 Unknown 
  
 Unknown 
 GEN642R018 
 missense_variant 
 c.3449C>G 
 p.Ala1150Gly 
 Unknown 
  
 Unknown 
 GEN642R019 
 missense_variant 
 c.431A>G 
 p.Lys144Arg 
 Unknown 
  
 Unknown 
 GEN642R020 
 missense_variant 
 c.8747G>A 
 p.Arg2916Gln 
 Unknown 
  
 Unknown 
 GEN642R021 
 missense_variant 
 c.4477C>T 
 p.Arg1493Cys 
 Unknown 
  
 Unknown 
 GEN642R022 
 missense_variant 
 c.4456C>T 
 p.Arg1486Cys 
 Unknown 
  
 Unknown 
 GEN642R023 
 stop_gained 
 c.7189C>T 
 p.Arg2397Ter 
 De novo 
  
  
 GEN642R024 
 frameshift_variant 
 c.4046_4050del 
 p.Lys1349ArgfsTer5 
 De novo 
  
  
 GEN642R025 
 missense_variant 
 c.6238G>A 
 p.Val2080Ile 
 De novo 
  
  
 GEN642R026 
 missense_variant 
 c.221C>T 
 p.Ser74Leu 
 De novo 
  
  
 GEN642R027 
 missense_variant 
 c.221C>T 
 p.Ser74Leu 
 Familial 
 Maternal 
  
 GEN642R028 
 missense_variant 
 c.6232G>T 
 p.Val2078Phe 
 Familial 
 Maternal 
  
 GEN642R029 
 missense_variant 
 c.6238G>A 
 p.Val2080Ile 
 Unknown 
 Not maternal 
  
 GEN642R030 
 missense_variant 
 c.7889G>C 
 p.Arg2630Thr 
 Familial 
 Paternal 
  
 GEN642R031 
 frameshift_variant 
 c.8869dup 
 p.Ile2957AsnfsTer14 
 De novo 
  
  
 GEN642R032 
 missense_variant 
 c.7172G>A 
 p.Arg2391His 
 Unknown 
  
  
 GEN642R033 
 frameshift_variant 
 c.3704del 
 p.Pro1235LeufsTer7 
 De novo 
  
  
 GEN642R034 
 frameshift_variant 
 c.1516_1517del 
 p.Phe506LeufsTer4 
 Unknown 
  
 Simplex 
 GEN642R035 
 stop_gained 
 c.8887C>T 
 p.Arg2963Ter 
 Unknown 
  
 Simplex 
 GEN642R036 
 stop_gained 
 c.8887C>T 
 p.Arg2963Ter 
 Unknown 
  
 Simplex 
 GEN642R037 
 missense_variant 
 c.8356G>A 
 p.Ala2786Thr 
 De novo 
  
  
 GEN642R038 
 frameshift_variant 
 c.3033del 
 p.Val1014CysfsTer24 
 De novo 
  
  
 GEN642R039 
 stop_gained 
 c.7276C>T 
 p.Arg2426Ter 
 De novo 
  
  
 GEN642R040 
 copy_number_loss 
  
  
 De novo 
  
  
 GEN642R041 
 frameshift_variant 
 c.2422_2423delinsT 
 p.Lys808TyrfsTer40 
 De novo 
  
  
 GEN642R042 
 missense_variant 
 c.8342G>A 
 p.Arg2781Gln 
 Familial 
 Maternal 
 Simplex 
 GEN642R043 
 missense_variant 
 c.5744A>G 
 p.Lys1915Arg 
 De novo 
  
 Simplex 
 GEN642R044 
 missense_variant 
 c.3319A>G 
 p.Ile1107Val 
 De novo 
  
  
 GEN642R045 
 missense_variant 
 c.221C>T 
 p.Ser74Leu 
 Familial 
 Maternal 
 Simplex 
 GEN642R046 
 missense_variant 
 c.4546C>T 
 p.Arg1516Cys 
 Familial 
 Paternal 
 Simplex 
 GEN642R047 
 missense_variant 
 c.4639A>G 
 p.Lys1547Glu 
 De novo 
  
 Simplex 
 GEN642R048 
 missense_variant 
 c.6230A>T 
 p.Tyr2077Phe 
 Familial 
 Maternal 
 Simplex 
 GEN642R049 
 missense_variant 
 c.7889G>C 
 p.Arg2630Thr 
 Familial 
 Paternal 
 Simplex 
 GEN642R050 
 missense_variant 
 c.7975C>T 
 p.Arg2659Cys 
 Unknown 
  
  
 GEN642R051 
 missense_variant 
 c.7916A>C 
 p.His2639Pro 
 Unknown 
  
  
 GEN642R052 
 missense_variant 
 c.7502A>G 
 p.Lys2501Arg 
 Unknown 
  
  
 GEN642R053 
 missense_variant 
 c.6598A>G 
 p.Ser2200Gly 
 Unknown 
  
  
 GEN642R054 
 missense_variant 
 c.5744A>G 
 p.Lys1915Arg 
 Unknown 
  
  
 GEN642R055 
 missense_variant 
 c.5384T>C 
 p.Ile1795Thr 
 Unknown 
  
  
 GEN642R056 
 missense_variant 
 c.5158C>T 
 p.Arg1720Trp 
 Unknown 
  
  
 GEN642R057 
 missense_variant 
 c.4583A>G 
 p.Tyr1528Cys 
 Unknown 
  
  
 GEN642R058 
 missense_variant 
 c.1673C>T 
 p.Pro558Leu 
 Unknown 
  
  
 GEN642R059 
 missense_variant 
 c.1518C>G 
 p.Phe506Leu 
 Unknown 
  
  
 GEN642R060 
 missense_variant 
 c.1318T>G 
 p.Cys440Gly 
 Unknown 
  
  
 GEN642R061 
 splice_site_variant 
 c.6009-2A>G 
  
 Unknown 
  
  
 GEN642R062 
 stop_gained 
 c.2309_2310insCATGATCTACAAATGA 
 p.Asp771MetfsTer5 
 Unknown 
  
  
 GEN642R063 
 frameshift_variant 
 c.2134dup 
 p.Arg712LysfsTer10 
 Unknown 
  
  
 GEN642R064 
 missense_variant 
 c.4546C>T 
 p.Arg1516Cys 
 Unknown 
  
  
 GEN642R065 
 missense_variant 
 c.221C>T 
 p.Ser74Leu 
 Unknown 
  
  
 GEN642R066 
 frameshift_variant 
 c.5081dup 
 p.Thr1695AsnfsTer18 
 De novo 
  
 Simplex 
 GEN642R067 
 missense_variant 
 c.5831T>G 
 p.Phe1944Cys 
 De novo 
  
 Simplex 
 GEN642R068 
 stop_gained 
 c.4024C>T 
 p.Arg1342Ter 
 De novo 
  
  
 GEN642R069 
 stop_gained 
 c.7618C>T 
 p.Arg2540Ter 
 De novo 
  
  
 GEN642R070 
 stop_gained 
 c.7204C>T 
 p.Arg2402Ter 
 De novo 
  
  
 GEN642R071 
 frameshift_variant 
 c.4039_4043del 
 p.Lys1347GlufsTer7 
 De novo 
  
  
 GEN642R072 
 frameshift_variant 
 c.3567dup 
 p.Glu1190Ter 
 De novo 
  
  
 GEN642R073 
 stop_gained 
 c.1348C>T 
 p.Gln450Ter 
 De novo 
  
  
 GEN642R074 
 frameshift_variant 
 c.921dup 
 p.Leu308ThrfsTer11 
 De novo 
  
  
 GEN642R075 
 frameshift_variant 
 c.8698dup 
 p.Thr2900AsnfsTer5 
 Unknown 
  
  
 GEN642R076 
 stop_gained 
 c.8902C>T 
 p.Arg2968Ter 
 Unknown 
  
  
 GEN642R077 
 stop_gained 
 c.8902C>T 
 p.Arg2968Ter 
 Unknown 
  
  
 GEN642R078 
 frameshift_variant 
 c.6247del 
 p.Leu2083PhefsTer30 
 Unknown 
  
  
 GEN642R079 
 splice_site_variant 
 c.420+2T>C 
  
 Unknown 
  
  
 GEN642R080 
 frameshift_variant 
 c.5446dup 
 p.Ile1816AsnfsTer13 
 Unknown 
  
  
 GEN642R081 
 missense_variant 
 c.4025G>A 
 p.Arg1342Gln 
 Familial 
 Paternal 
  
 GEN642R082 
 missense_variant 
 c.6169C>G 
 p.His2057Asp 
 Familial 
 Paternal 
  
 GEN642R083 
 missense_variant 
 c.4546C>T 
 p.Arg1516Cys 
 Familial 
 Paternal 
  
 GEN642R084 
 missense_variant 
 c.6931C>T 
 p.Arg2311Trp 
 Familial 
 Paternal 
  
 GEN642R085 
 missense_variant 
 c.4604G>T 
 p.Arg1535Leu 
 Unknown 
  
  
 GEN642R086 
 missense_variant 
 c.4870C>T 
 p.Arg1624Trp 
 Unknown 
  
  
 GEN642R087 
 missense_variant 
 c.4870C>T 
 p.Arg1624Trp 
 Unknown 
  
  
 GEN642R088 
 missense_variant 
 c.7598G>A 
 p.Arg2533His 
 Unknown 
  
  
 GEN642R089 
 missense_variant 
 c.7619G>A 
 p.Arg2540Gln 
 Unknown 
  
  
 GEN642R090 
 missense_variant 
 c.8429C>T 
 p.Ala2810Val 
 Unknown 
  
  
 GEN642R091 
 missense_variant 
 c.3905G>A 
 p.Arg1302Gln 
 Unknown 
  
  
 GEN642R092 
 missense_variant 
 c.7759C>T 
 p.Arg2587Cys 
 Unknown 
  
  
 GEN642R093 
 missense_variant 
 c.7313A>G 
 p.Glu2438Gly 
 Unknown 
  
  
 GEN642R094 
 missense_variant 
 c.8459C>T 
 p.Ser2820Leu 
 Unknown 
  
  
 GEN642R095 
 missense_variant 
 c.8536C>T 
 p.Arg2846Cys 
 Unknown 
  
  
 GEN642R096 
 missense_variant 
 c.8536C>T 
 p.Arg2846Cys 
 Unknown 
  
  
 GEN642R097 
 missense_variant 
 c.8536C>T 
 p.Arg2846Cys 
 Unknown 
  
  
 GEN642R098 
 missense_variant 
 c.7598G>A 
 p.Gly2533Glu 
 Unknown 
  
  
 GEN642R099 
 missense_variant 
 c.8201G>A 
 p.Arg2734His 
 Unknown 
  
  
 GEN642R100 
 missense_variant 
 c.8201G>A 
 p.Arg2734His 
 Unknown 
  
  
 GEN642R101 
 missense_variant 
 c.3905G>C 
 p.Arg1302Pro 
 Unknown 
  
  
 GEN642R102 
 missense_variant 
 c.7760G>A 
 p.Arg2587His 
 Unknown 
 Not maternal 
  
 GEN642R103 
 missense_variant 
 c.6931C>T 
 p.Arg2311Trp 
 Unknown 
  
  
 GEN642R104 
 missense_variant 
 c.3760C>T 
 p.Arg1254Cys 
 Unknown 
  
  
 GEN642R105 
 missense_variant 
 c.3760C>T 
 p.Arg1254Cys 
 Unknown 
  
 Simplex 
 GEN642R106 
 missense_variant 
 c.3760C>T 
 p.Arg1254Cys 
 Unknown 
  
  
 GEN642R107 
 frameshift_variant 
 c.8698dup 
 p.Thr2900AsnfsTer5 
 Unknown 
  
  
 GEN642R108 
 frameshift_variant 
 c.8698del 
 p.Thr2900GlnfsTer44 
 Unknown 
  
  
 GEN642R109 
 frameshift_variant 
 c.3681del 
 p.Phe1227LeufsTer6 
 Unknown 
  
  
 GEN642R110 
 frameshift_variant 
 c.4651_4654del 
 p.Asn1551GlufsTer60 
 Unknown 
  
  
 GEN642R111 
 missense_variant 
 c.8482T>C 
 p.Tyr2828His 
 Unknown 
  
  
 GEN642R112 
 missense_variant 
 c.8764G>A 
 p.Glu2922Lys 
 Unknown 
  
  
 GEN642R113 
 missense_variant 
 c.7640G>A 
 p.Arg2547Gln 
 Unknown 
  
  
 GEN642R114 
 missense_variant 
 c.6007G>A 
 p.Asp2003Asn 
 Unknown 
  
  
 GEN642R115 
 missense_variant 
 c.6007G>A 
 p.Asp2003Asn 
 Unknown 
  
  
 GEN642R116 
 missense_variant 
 c.46G>A 
 p.Glu16Lys 
 Unknown 
  
  
 GEN642R117 
 missense_variant 
 c.8030G>A 
 p.Arg2677His 
 Unknown 
  
  
 GEN642R118 
 missense_variant 
 c.7558C>T 
 p.Arg2520Trp 
 Unknown 
  
  
 GEN642R119 
 missense_variant 
 c.6869G>A 
 p.Arg2290His 
 Unknown 
  
  
 GEN642R120 
 missense_variant 
 c.5050C>T 
 p.Arg1684Trp 
 Unknown 
  
  
 GEN642R121 
 missense_variant 
 c.3904C>T 
 p.Arg1302Trp 
 Unknown 
  
  
 GEN642R122 
 missense_variant 
 c.7187G>A 
 p.Arg2396His 
 Unknown 
  
  
 GEN642R123 
 missense_variant 
 c.7187G>A 
 p.Arg2396His 
 Unknown 
  
  
 GEN642R124 
 missense_variant 
 c.3905G>A 
 p.Arg1302Gln 
 Unknown 
  
  
 GEN642R125 
 missense_variant 
 c.7759C>T 
 p.Arg2587Cys 
 Unknown 
  
  
 GEN642R126 
 missense_variant 
 c.7637C>T 
 p.Ala2546Val 
 Unknown 
  
  
 GEN642R127 
 missense_variant 
 c.6868C>T 
 p.Arg2290Cys 
 Unknown 
  
  
 GEN642R128 
 missense_variant 
 c.8459C>T 
 p.Ser2820Leu 
 Unknown 
  
  
 GEN642R129 
 missense_variant 
 c.7286G>A 
 p.Arg2429Gln 
 Unknown 
  
  
 GEN642R130 
 missense_variant 
 c.7286G>A 
 p.Arg2429Gln 
 Unknown 
  
  
 GEN642R131 
 missense_variant 
 c.5051G>A 
 p.Arg1684Gln 
 Unknown 
  
  
 GEN642R132 
 missense_variant 
 c.5422C>T 
 p.Arg1808Trp 
 Unknown 
  
  
 GEN642R133 
 missense_variant 
 c.1298A>C 
 p.Gln433Pro 
 Familial 
 Maternal 
 Multiplex 
 GEN642R134a 
 missense_variant 
 c.4025G>A 
 p.Arg1342Gln 
 Familial 
 Both parents 
 Simplex 
 GEN642R135 
 frameshift_variant 
 c.8854_8855insT 
 p.Gly2952ValfsTer24 
 Unknown 
  
  
 GEN642R136 
 frameshift_variant 
 c.4902_4903del 
 p.Ser1635CysfsTer18 
 De novo 
  
 Simplex 
 GEN642R137 
 frameshift_variant 
 c.1958dup 
 p.Pro654AlafsTer6 
 De novo 
  
 Simplex 
 GEN642R138 
 frameshift_variant 
 c.773del 
 p.Gly258ValfsTer5 
 De novo 
  
 Multiplex 
 GEN642R139 
 splice_region_variant 
 c.6238+3G>C 
  
 De novo 
  
  
 GEN642R140 
 missense_variant 
 c.3515C>T 
 p.Ser1172Leu 
 De novo 
  
  
 GEN642R141 
 missense_variant 
 c.102G>C 
 p.Lys34Asn 
 Familial 
 Paternal 
  
 GEN642R142 
 frameshift_variant 
 c.8595del 
 p.Glu2866ArgfsTer3 
 De novo 
  
  
 GEN642R143 
 missense_variant 
 c.229G>A 
 p.Glu77Lys 
 Familial 
 Paternal 
 Simplex 
 GEN642R144 
 stop_gained 
 c.1577dup 
 p.Tyr526Ter 
 Unknown 
  
 Simplex 
 GEN642R145 
 missense_variant 
 c.3771T>G 
 p.His1257Gln 
 Familial 
 Paternal 
 Simplex 
  et al.  

Common

Variant ID
Polymorphism
SNP ID
Allele Change
Residue Change
Population Origin
Population Stage
Author, Year
 GEN642C001 
 intron_variant 
 rs35615695 
 c.-99-9605A>G 
  
 271 Tourette syndrome nuclear family trios and 337 control subjects of Han Chinese descent from the Affiliated Hospital of Qingdao University; all TS patients diagnosed according to DSM-V criteria. 
 Discovery 
 GEN642C002 
 intron_variant 
 rs5005770 
 c.6332+3029T>C 
  
 272 Tourette syndrome nuclear family trios and 337 control subjects of Han Chinese descent from the Affiliated Hospital of Qingdao University; all TS patients diagnosed according to DSM-V criteria. 
 Discovery 
Chromosome
CNV Locus
CNV Type
# of studies
Animal Model
1
Duplication
 44
 
1
Duplication
 1
 
1
Deletion
 3
 
1
Duplication
 1
 
1
Deletion-Duplication
 11
 

Model Summary

Ash1l null mice exhibit Increased premature lethality starts after P7 and growth retardation. Surviving mutants have skeletal abnormalities, blepharitis, and infertility caused by developmental defects in both male and female reproductive organs. The malformed reproductive organs observed in Ash1l null mice might result from the decreased expression of Hoxa10 in uterin or Hoxa11 and Hoxd10 in epididymis.

References

Type
Title
Author, Year
Additional
Histone methyltransferase Ash1l suppresses interleukin-6 production and inflammatory autoimmune diseases by inducing the ubiquitin-editing enzyme A20.
Primary
The Histone Methyltransferase Gene Absent, Small, or Homeotic Discs-1 Like Is Required for Normal Hox Gene Expression and Fertility in Mice.
Additional
Histone methyltransferase Ash1L mediates activity-dependent repression of neurexin-1.
additional
Loss of histone methyltransferase ASH1L in the developing mouse brain causes autistic-like behaviors
Additional
ASH1L haploinsufficiency results in autistic-like phenotypes in mice and links Eph receptor gene to autism spectrum disorder
Neural Hyperactivity Is a Core Pathophysiological Change Induced by Deletion of a High Autism Risk Gene Ash1L in the Mouse Brain

M_ASH1L_2_KO_HM

Model Type: Genetic
Model Genotype: Homozygous
Mutation: A transposon- mediated (piggyBac) DNA insertion of actin-promoter-driven RFP gene between exon 15 and 16 of Ash1l allele, which results in reduced expression of Ash1l mRNA.
Allele Type: Transgenic
Strain of Origin: FVB
Genetic Background: C57BL/6J
ES Cell Line:
Mutant ES Cell Line:
Model Source:

M_ASH1L_1_KO_HM

Model Type: Genetic
Model Genotype: Homozygous
Mutation: A gene trap vector, pGT01xf, containing the beta-geo reporter gene (beta-galactosidase and neomycin resistance fusion gene) was inserted into intron 1 of the Ash1l gene, which results in mis-splicing leading to a truncated product missing AT Hook domains, the SET associated domains, the bromodomain, and the PHD domain.
Allele Type: Gene trapped
Strain of Origin: Not specified
Genetic Background: C57BL/6J and DBA/2J
ES Cell Line: 129P2/OlaHsd
Mutant ES Cell Line: Not specified
Model Source: Not specified

M_ASH1L_1_KO_HT

Model Type: Genetic
Model Genotype: Heterozygous
Mutation: A gene trap vector, pGT01xf, containing the beta-geo reporter gene (beta-galactosidase and neomycin resistance fusion gene) was inserted into intron 1 of the Ash1l gene, which results in mis-splicing leading to a truncated product missing AT Hook domains, the SET associated domains, the bromodomain, and the PHD domain.
Allele Type: Gene trapped
Strain of Origin: Not specified
Genetic Background: C57BL/6J and DBA/2J
ES Cell Line: 129P2/OlaHsd
Mutant ES Cell Line: Not specified
Model Source: Not specified

M_ASH1L_3_KO_HM

Model Type: Genetic
Model Genotype: Homozygous
Mutation: Deletion of 11 bp in the exon 2 of Ash1l gene was generated by pronuclei injection of a targeting sgRNA and Cas9-coding mRNA (CRISPR/Cas9 system), which resulted in a null allelle of Ash1L.
Allele Type: Targeted (knockout)
Strain of Origin:
Genetic Background: C57BL/6
ES Cell Line:
Mutant ES Cell Line:
Model Source:

M_ASH1L_3_KO_HT

Model Type: Genetic
Model Genotype: Heterozygous
Mutation: Deletion of 11 bp in the exon 2 of Ash1l gene was generated by pronuclei injection of a targeting sgRNA and Cas9-coding mRNA (CRISPR/Cas9 system), which resulted in a null allelle of Ash1L.
Allele Type: Targeted (knockout)
Strain of Origin:
Genetic Background: C57BL/6
ES Cell Line:
Mutant ES Cell Line:
Model Source:

M_ASH1L_4_KO_HM

Model Type: Genetic LOF
Model Genotype: Homozygous
Mutation: Ash1l conditional knockout (cko) mouse line with a cre recombinase-mediated deletion of exon 4 that results in altered splicing of mrna that creates a premature stop codon before the sequences encoding the first functional aws (associated with set), set, bromo, bah and phd domains. the truncated ash1l protein contains the n-terminal 1,694 amino acids but no functional domains, thus mimicking the disruptive mutations found in patients. (ash1l+/2f)
Allele Type: Knockout
Strain of Origin: Not reported
Genetic Background: C57BL/6
ES Cell Line: NA
Mutant ES Cell Line: NA
Model Source: 34145365

M_ASH1L_5_KO_HT

Model Type: Genetic LOF
Model Genotype: Heterozygous
Mutation: Ash1l conditional knockout (cko) mouse line with a cre recombinase-mediated deletion of exon 4 that results in altered splicing of mrna that creates a premature stop codon before the sequences encoding the first functional aws (associated with set), set, bromo, bah and phd domains. the truncated ash1l protein contains the n-terminal 1,694 amino acids but no functional domains, thus mimicking the disruptive mutations found in patients. heterozygous ash1l-ko mice (ash1l+/1f) were obtained by crossing the wildtype ash1l+/2f mice with cmv-cre mice, through which one allele of ash1l gene was deleted in both germlines and somatic cells in progenies.
Allele Type: Knockout
Strain of Origin: Not reported
Genetic Background: C57BL/6
ES Cell Line: NA
Mutant ES Cell Line: NA
Model Source: 34145365

M_ASH1L_6_CKO_HM

Model Type: Genetic LOF
Model Genotype: Homozygous
Mutation: Mice with ash1l deleted in the npcs and npc-derived neuronal and glial lineages in the developing mouse brain by crossing the ash1l-conditional ready mice bearing floxed exon 4 with a neural progenitor cell (npc)-specific cre (nestin-cre) mouse line. homozygous ash1l-cko (ash1l-nes-cko, ash1l2f/2f;nestin-cre+/â??)
Allele Type: Conditional knockout
Strain of Origin: Not reported
Genetic Background: C57BL/6
ES Cell Line: NA
Mutant ES Cell Line: NA
Model Source: 34145365

M_ASH1L_7_CKO_HT

Model Type: Genetic LOF
Model Genotype: Heterozygous
Mutation: Mice with ash1l deleted in the npcs and npc-derived neuronal and glial lineages in the developing mouse brain by crossing the ash1l-conditional ready mice bearing floxed exon 4 with a neural progenitor cell (npc)-specific cre (nestin-cre) mouse line. heterozygous (ash1l2f/+;nestin-cre+/â??)
Allele Type: Conditional knockout
Strain of Origin: Not reported
Genetic Background: C57BL/6
ES Cell Line: NA
Mutant ES Cell Line: NA
Model Source: 34145365

M_ASH1L_8_CKO_HM

Model Type: Genetic LOF
Model Genotype: Homozygous
Mutation: Tamoxifen-inducible ash1l-cko mouse line (ash1l2f/2f;rosa26-creert2+/+) generated by crossing the ash1l-cko mice bearing floxed exon 4 with a rosa26-creert2 line.
Allele Type: Conditional knockout
Strain of Origin: Not reported
Genetic Background: C57BL/6
ES Cell Line: NA
Mutant ES Cell Line: NA
Model Source: 34145365

M_ASH1L_9_CKO_HM

Model Type: Genetic
Model Genotype: Homozygous
Mutation: Exon 2 of Ash1l was flanked with one pair of loxP sites. Ash1l fl/fl females were crossed with Emx1-Cre then Ash1lfl/+ (Ash1lEmx1-het) males were crossed to obtain homozygous mice with conditional deletion of Ash1l in neurons of forebrain, especially neocortex and hippocampus.
Allele Type: conditional knockout
Strain of Origin:
Genetic Background: C57BL/6
ES Cell Line:
Mutant ES Cell Line:
Model Source:

M_ASH1L_2_KO_HM

Category
Entity
Quantity
Experimental Paradigm
Age at Testing
Cytokine levels: pro-inflammatory1
Increased
Description: Peritoneal macrophages from ash1l-silenced mice produce more il-6 and tnf in response to lps or poly(i:c) than those from wildtype controls; the serum of ash1l-silenced mice contains increased levels of il-6 and tnf in response to injected e. coli (10e8) , lps, or poly(i:c) relative to wildtype controls
Exp Paradigm: Immunoassay: cytometric bead array (cba): il-6, tnf; thioglycolate-elicited mouse peritoneal macrophages are treated with lps (100 ng/ml) or poly(i:c) (10 microgram/ml); mice are injected intraperitoneally with e. coli (10e8) , lps (15 mg/kg), or poly(i:c) (20 mg/kg)
 Immunoassay
 6-8 weeks
Autoimmune reaction1
Increased
Description: Ash1l-silenced aged mice have higher immunoglobulins (igm, igg1, igg2a, igg2b, and iga) and il-6 in serum, have more severe infiltration of mononuclear cells in various organs, exhibit increased susceptibility to membranous glomerulonephritis, and have more deposition of immune complexes of autoantibodies in the glomerulis; higher incidence of ash1l-silenced mice are subject to collagen ii-induced arthritis (i.e. increased thickness in hind ankle joints and more severe swelling in the paws, increased concentration of il-6 in serum, and more severe cartilage and bone destruction and mononuclear cells infiltration in joint capsule) relative to wildtype controls
Exp Paradigm: Histology; elisa; collagen ii-induced arthritis (cia)
 Collagen ii-induced arthritis (cia)
 8 months
Innate immune response1
Increased
Description: Ash1l-silenced mice have severe inflammatory hyperemia in the lungs with increased infiltration of mononuclear cells and red blood cells 6 hr after e. coli or lps injection; ash1l-silenced mice have higher bacterial load in the blood and are more susceptible to e. coli-induced sepsis
Exp Paradigm: Histology
 Histology
 6-8 weeks
Protein expression level evidence1
Decreased
Description: Ash1l-silenced macrophages exhibit reduced a20 expression upon lps and poly(i:c) stimulation as compared to wildtype controls
Exp Paradigm: Lps or poly(i:c) induced peritoneal macrophages; nuclear run-on assay: tnfaip3; western blot: a20 (tnfaip3 protein)-nuclear run-on assay
 Nuclear run-on assay
 6-8 weeks
Protein expression level evidence1
Decreased
Description: Ash1l-silenced macrophages exhibit reduced a20 expression upon lps and poly(i:c) stimulation as compared to wildtype controls
Exp Paradigm: Lps or poly(i:c) induced peritoneal macrophages; nuclear run-on assay: tnfaip3; western blot: a20 (tnfaip3 protein)- western blot
 Western blot
 6-8 weeks
Chromatin modification1
Decreased
Description: The chromatin accessibility and the concentration of h3k4me3 modification of the tnfaip3 promoter is significantly decreased in ash1l-silenced macrophages upon tlr stimulation as compared to wildtype controls
Exp Paradigm: Dnase i hypersensitive sites sequencing (dnase-seq); chromatin immunoprecipitation (chip)
 Dnase i hypersensitive sites sequencing (dnase-seq)
 6-8 weeks
Signaling1
Increased
Description: Enhanced nf-kappab and mapk signalings in ash1l-silenced macrophages in response to lps or tnf compared to wildtype controls
Exp Paradigm: Western blot: phosphorylation of ikkalpha, ikkbeta, ikappabalpha, and p65 (the nuclear translocation of p65 is also assayed) as well as phosphorlation of erk, jnk, and p38; k63-ubiquitination on nemo and traf6
 Western blot
 6-8 weeks
Development of immune cells and primary lymphoid organs1
 No change
 Flow cytometric analysis
 6-8 weeks
Gene expression1
 No change
 Quantitative pcr (qrt-pcr)
 6-8 weeks
 Not Reported: Circadian sleep/wake cycle, Communications, Developmental profile, Emotion, Learning & memory, Maternal behavior, Motor phenotype, Neuroanatomy / ultrastructure / cytoarchitecture, Neurophysiology, Physiological parameters, Repetitive behavior, Seizure, Sensory, Social behavior

M_ASH1L_1_KO_HM

Category
Entity
Quantity
Experimental Paradigm
Age at Testing
Homeostasis: adiposity1
Decreased
Description: Decreased abdominal fat in the knockout mice compared to littermate controls
Exp Paradigm: Males
 Measurement of tissue weight
 2-3 months
Reproductive function: uterus1
Decreased
Description: The uterine function in supporting implantation and decidualization were impaired in the mutant mice.
Exp Paradigm: Histology: chicago sky blue 6b staining
 Histology
 6-8 weeks
Apoptosis1
Increased
Description: Increased apoptotic cells in the caudal epididymis of the mutant mice compared to heterozygous controls
Exp Paradigm: Activated caspase 3 immunostaining
 Expression of cleaved caspase-3 (cc3)
 Adult
Reproductive function1
Decreased
Description: Female knockout mice were infertile with no change in the birth rate of the pups that developed from the mutant blastocysts in foster mothers; there is no change in the in vitro fertilization rate by using ash1l null eggs; no change in the estrous cycle in ash1l nulls
Exp Paradigm: Female only; embryo transfer
 Vaginal smear
 Unreported
Reproductive function1
Decreased
Description: Decreased litters produced per male mutant mouse; decreased count of motile sperm collected from the mutant vas deferens; no significant change in the sperm count at epididymis; no change in the in vitro fertilization rate
Exp Paradigm: Male only; motile sperm count; h&e histochemistry
 Motile sperm count
 Unreported
Size/growth1
Decreased
Description: Decreased body weight was observed at 2 weeks or 3 weeks old knockout mice (compared to littermate controls)
Exp Paradigm: Body weight measurement
 Body weight measurement
 P14-p21
Eye development: eyelid1
Decreased
Description: 4 out of 7 knockout mice have an abnormal expansion of the corneal epithelium, disorganization of the basal lamina, and an apparent reduction in sebaceous glands
Exp Paradigm: General observations-general observations
 General observations
 2 months
Reproductive system development: uterus morphology1
Decreased
Description: Uteri are smaller and twisted in the mutants. the stromal layer of the uterus was thinner in mutants and the uterine glands were 4-fold less than in the littermate controls
Exp Paradigm: General observations
 Histology
 3-4 months
Eye development: eyelid1
Decreased
Description: 4 out of 7 knockout mice have an abnormal expansion of the corneal epithelium, disorganization of the basal lamina, and an apparent reduction in sebaceous glands
Exp Paradigm: General observations- histology
 Histology
 2 months
Reproductive system development: epididymis morphology1
Decreased
Description: 17 out of 20 epididymides from mutants had a abnormal morphology from corpus to caput, a widening of the cauda, and a twisting of the ductus deferens
Exp Paradigm: The gross structure of the isolated tissue was visualized by x-gal staining
 General observations
 Adult
Skeletal development1
Decreased
Description: Knockout embryos (60%) had an eighth rib attached to the sternum on either the right side (25%) left side (25%), or both sides (10%) as well as the ribs were attached in a staggered manner to the sternum
Exp Paradigm: Alcian blue/alizarin red staining
 Alcian blue/alizarin red staining
 E18.5
Mortality/lethality1
Increased
Description: Increased premature lethality starts after p7
Exp Paradigm: Genotypic ratio of progeny from heterozygous parents
 Genotypic ratio of progeny from heterozygous parents
 E11.5-e18.5; p7-p21
Protein expression: in situ protein expression1
Decreased
Description: Decreased protein levels of hoxd10 in initial segment, caput, and ductus deferens; decreased protein levels of hoxa11 in the initial segment of the mutant epididymis
Exp Paradigm: Quantitative pcr (qrt-pcr)
 Quantitative pcr (qrt-pcr)
 P21
Protein expression: in situ protein expression1
Decreased
Description: Decreased protein levels of either foxa2 or hoxa10 at the mutant uterine glands or the stroma, respectively
Exp Paradigm: Immunostaining: foxa2, hoxa10
 Immunostaining
 P21
Targeted expression1
Decreased
Description: Decreased ash1l protein expression at the cells lining epididymal tubules in mutant mice
Exp Paradigm: Immunostaining; rt-qpcr-immunostaining
 Immunostaining
 Adult
Targeted expression1
Decreased
Description: Decreased ash1l protein expression at the cells lining epididymal tubules in mutant mice
Exp Paradigm: Immunostaining; rt-qpcr- quantitative pcr (qrt-pcr)
 Quantitative pcr (qrt-pcr)
 Adult
Size/growth1
 No change
 Body weight measurement
 P1-p7
Protein expression: in situ protein expression1
 No change
 Immunostaining
 P21
 Not Reported: Circadian sleep/wake cycle, Communications, Emotion, Immune response, Learning & memory, Maternal behavior, Motor phenotype, Neuroanatomy / ultrastructure / cytoarchitecture, Neurophysiology, Physiological parameters, Repetitive behavior, Seizure, Sensory, Social behavior

M_ASH1L_1_KO_HT

Category
Entity
Quantity
Experimental Paradigm
Age at Testing
Mortality/lethality1
 No change
 Genotypic ratio of progeny from heterozygous parents
 E11.5-e18.5; 1 week; 1-3 weeks
Reproductive system development: epididymis morphology1
 No change
 Histology
 Adult
Reproductive system development: epididymis morphology1
 No change
 General observations
 Adult
Size/growth1
 No change
 Body weight measurement
 P1-p21
Reproductive function1
 No change
 General observations
 Adult
 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_ASH1L_3_KO_HM

Category
Entity
Quantity
Experimental Paradigm
Age at Testing
Neuronal activity dependent gene regulation1
Decreased
Description: Ash1l null hippocampal neurons exhibit no activity-dependent repression of nrxn1alpha relative to wildtype controls
Exp Paradigm: Quantitative pcr (qrt-pcr): nrxn1alpha; qrt-pcrs are conducted 24 hours after high potassium stimulation or by optical stimulation (primary cultured neurons tansfected by lentiviral vector, lv: camkii-chr2)
 Quantitative pcr (qrt-pcr)
 E14
Mortality/lethality: embryonic2
increased
Description: Ash1l KO mice dies in utero
 General observations
 Unreported
Mortality/lethality1
Increased
Description: No homozygous ash1l nulls are observed at 1 week of age from intercrosses of ash1l hets
Exp Paradigm: Genotypic ratio of progeny from heterozygous parents
 Genotypic ratio of progeny from heterozygous parents
 P7
 Not Reported: Circadian sleep/wake cycle, Communications, Emotion, Immune response, Learning & memory, Maternal behavior, Molecular profile, Motor phenotype, Neuroanatomy / ultrastructure / cytoarchitecture, Physiological parameters, Repetitive behavior, Seizure, Sensory, Social behavior

M_ASH1L_3_KO_HT

Category
Entity
Quantity
Experimental Paradigm
Age at Testing
Cued or contextual fear conditioning: memory of context2
decreased
Description: Ash1l het mice showed reduced freezing on day 3 during learning; difference in freezing was not observed between distinct contexts indicating impairment of discrimination ability
 Fear conditioning test
 3-6-month
Synapse density2
abnormal
Description: elimination rates of boutons at 24 h after conditioning were significantly lower in Ash1l het mice than that in WT mice, while the bouton formation rates remained unchanged; tone-dependent fear conditioning learning induced an increase in total bouton number in Ash1l het mice than in WT mice at 24 h after conditioning; in WT but not in Ash1l het mice, density of axonal boutons in activated AUD-LA projections (mCherry+) was significantly lower than that in non-activated AUD-LA projections after tone-dependent fear conditioning
Exp Paradigm: axons labeled by AAV-EF1a-DIO-EYFP in Rbp4-Cre mice; Ash1l crossed to FosTRAP mice
 Two-photon microscopy
 3-6-month
Chromatin modification: histone modification: open chromatin2
decreased
Description: Learning-induced significant accumulation of the repressive marker H3K27me3 on EphA7 gene; learning also induced a higher level of accumulation of EZH2 on EphA7 promoter region in Ash1l het mice compared with the WT mice; H3K27me3 levels in the EphA7 promoter region and adjacent gene body regions were increased in the Ash1l mutant mice
Exp Paradigm: tone dependent Go-No-go task; ephrin-A5 Fc was bilaterally infused in the auditory cortex immediately after the training phase
 Chromatin immunoprecipitation sequencing (ChIP-seq)
 3-6-month
Dendritic architecture: spine density2
increased
Description: Ash1l het mice had cortical neurons and dorsal striatal medium spiny neurons with significantly increased spine density
 Golgi-Cox staining
 1 month
Chromatin modification: histone modification: open chromatin2
decreased
Description: H3K36me2, the direct target of Ash1l, showed no difference around EphA7 gene loci between WT and Ash1l het mice in auditory cortex; H3K27me3 signal showed significant increase in EphA7 gene loci of Ash1l het mice compared with WT mice; H3K27 acetylation was decreased near the EphA7 promoter region in Ash1l het mice compared with the WT mice
 Chromatin immunoprecipitation sequencing (ChIP-seq)
 3-6-month
Cued or contextual fear conditioning: memory of cue2
abnormal
Description: Ash1l het mice showed comparable freezing with WT for the conditioned CS+ stimulus but showed significantly increased freezing with the CS- stimulus; Ash1l het mice showed impaired discrimination ability
 Fear conditioning test
 3-6-month
Ultrasonic vocalization: isolation induced2
decreased
Description: Ash1l het mice had fewer neonatal ultrasonic vocalizations
 Monitoring ultrasonic vocalizations
 P7-9
Repetitive digging2
increased
Description: Ash1l het mice showed a high degree of digging in novel contexts
 Marble burying test
 3-6-month
Social memory2
decreased
Description: Ash1l het mice showed deficits in social novelty discrimination
 Three-chamber social approach test
 3-6-month
Anxiety2
increased
Description: Ash1l het mice spent less time at the center of the open field
 Open field test
 3-6-month
Reward reinforced choice behavior2
abnormal
Description: Ash1l het mice had performance similar to WT mice; Ash1l het mice showed higher level of licking probability in many untrained auditory signals compared to WT mice
Exp Paradigm: tone dependent Go-No-go task
 Go-no-go test
 3-6-month
Gene expression2
decreased
Description: EphA7 was downregulated in both auditory cortex and dorsal straitum at both ages tested in Ash1l het mice
 RNA sequencing
 1 month, 1 year
Targeted expression1
Decreased
Description: Decreased ash1l expression at the hippocampus of ash1l hets relative to wildtype controls
Exp Paradigm: Western blot: ash1l; quantitative pcr (qrt-pcr): ash1l- quantitative pcr (qrt-pcr)
 Quantitative pcr (qrt-pcr)
 3 months; e14
Differential gene expression2
abnormal
Description: transcriptome analysis showed 563 differentially expressed genes in the auditory cortex and 155 in the dorsal striatum; Ash1l downregulated genes involved in regulation of synaptic transmission, synapse structure, axon part, and synaptic membrane in the auditory cortex and dorsal striatum
 RNA sequencing
 1 month
Neuronal activity dependent gene regulation2
decreased
Description: activated neurons in Ash1l het mice showed a much lower expression level of EphA7 than that in WT mice and did not show elevated expression of EphA7 compared with silent neurons; After tone dependent fear conditioning training for 12h, the expression of EphA7 was significantly reduced in Ash1l het mice compared to WT mice in the task-activated neurons
Exp Paradigm: mice crossed with EGR1-EGFP transgenic line; fluorescence-activated cell sorting (FACS)
 Western blot
 3-6-month
Differential gene expression2
abnormal
Description: transcriptome analysis showed 1691 differentially expressed genes in the auditory cortex and 861 in the dorsal striatum; Ash1l downregulated genes involved in regulation of synaptic transmission, synapse structure, axon part, and synaptic membrane in the auditory cortex and dorsal striatum
 RNA sequencing
 1 year
Gene expression1
Increased
Description: Increased neurexin 1alpha mrna in the hippocampus of ash1l hets relative to wildtype controls
Exp Paradigm: Quantitative pcr (qrt-pcr): nrxn1alpha
 Quantitative pcr (qrt-pcr)
 3 months
Chromatin modification1
Decreased
Description: Ash1l hets exhibit decreased h3k36me2 at nrxn1alpha promoter region relative to wildtype controls in hippocampus; overall epigenetic histone modification in the hippocampus of ash1l hets is not changed
Exp Paradigm: Chromatin immunoprecipitation (chip): h3k36me2, h3k4me3
 Chromatin immunoprecipitation (chip)
 3 months
Targeted expression1
Decreased
Description: Decreased ash1l expression at the hippocampus of ash1l hets relative to wildtype controls
Exp Paradigm: Western blot: ash1l; quantitative pcr (qrt-pcr): ash1l-western blot
 Western blot
 3 months; e14
Chromatin modification1
 No change
 Chromatin immunoprecipitation (chip)
 3 months
Targeted expression1
 No change
 Western blot
 3 months
General locomotor activity2
 no change
 Open field test
 3-6-month
Brain size1
 No change
 Measurement of tissue weight
 3 months
Exploratory activity: habituation2
 no change
 Three-chamber social approach test
 3-6-month
Hippocampal morphology1
 No change
 Immunostaining
 3 months
Social approach2
 no change
 Three-chamber social approach test
 3-6-month
 Not Reported: Circadian sleep/wake cycle, Communications, Developmental profile, Emotion, Immune response, Learning & memory, Maternal behavior, Motor phenotype, Neurophysiology, Physiological parameters, Repetitive behavior, Seizure, Sensory, Social behavior

M_ASH1L_4_KO_HM

Category
Entity
Quantity
Experimental Paradigm
Age at Testing
Cortical lamination1
Decreased
Description: Some satb2+ neurons were not properly located on the upper layers and scattered in the bottom layers suggesting delayed lamination of neuronal cells during embryonic cortical layer formation
 Immunohistochemistry
 Adult
Neocortex morphology1
Abnormal
Description: Cortices were disorganized and lost its minicolumnar arrangement
 Histology
 Adult
Mortality/lethality: neonatal1
Abnormal
Description: Without maternal uterine support, all ash1l-ko newborns died within 24 h after birth suggesting ash1l might be critical for establishing and maintaining a stable physiological condition for neonatal survival
 General observations
 P0
Developmental trajectory1
Decreased
Description: Delayed embryonic and postnatal brain development
 Gross necroscopy
 P0
Skeletal development1
Abnormal
Description: Majority of ash1l-ko newborns displayed aberrant rib numbers
 Bone histomorphometry
 Not reported
Targeted expression1
Decreased
Description: Truncated ash1l protein
 Western blot
 Not reported
Targeted expression1
Decreased
Description: Truncated ash1l contains the n-terminal 1,694 amino acids no functional domains, mimicking the disruptive mutations found in patients
 Polymerase chain reaction (PCR)
 Not reported
General characteristics1
 No change
 General observations
 E13.5
Mortality/lethality1
 No change
 Genotypic ratio of progeny from heterozygous parents
 Adult
Size/growth1
 No change
 Body weight measurement
 P0
Brain size1
 No change
 Measurement of tissue weight
 P0
 Not Reported:

M_ASH1L_5_KO_HT

Category
Entity
Quantity
Experimental Paradigm
Age at Testing
Targeted expression1
Decreased
Description: Truncated ash1l contains the n-terminal 1,694 amino acids no functional domains, mimicking the disruptive mutations found in patients
 Polymerase chain reaction (PCR)
 Not reported
General characteristics1
 No change
 General observations
 E13.5
Mortality/lethality1
 No change
 Genotypic ratio of progeny from heterozygous parents
 Adult
Size/growth1
 No change
 Body weight measurement
 P0
 Not Reported:

M_ASH1L_6_CKO_HM

Category
Entity
Quantity
Experimental Paradigm
Age at Testing
Locomotor activity in diurnal cycle: light phase1
Increased
Description: Mutant mice maintained higher locomotor activities during the light-on cycle.
Exp Paradigm: TSE PhenoMaster/LabMaster System
 Home cage behavior
 month 4
General locomotor activity: Ambulatory activity3
Increased
Description: No change in distance travelled in 5 mins, increased distance travelled in 10 mins
 Open field test
 Adult
General locomotor activity1
Increased
Description: Both male and female mutant mice had significantly more locomotor activity and longer running distances.
Exp Paradigm: TSE PhenoMaster/LabMaster System
 Home cage behavior
 month 4
Myelination3
Decreased
Description: Levels of myelination in the cortices were significantly lower at p21 but reached to a comparable level around postnatal two months, indicating delayed myelination
 Immunohistochemistry
 P0-2month
Neuronal activation1
Increased
Description: Mutant mice had significant increased c-Fos positive cells in motor cortices, amygdalae, and hypothalami.
 Immunofluorescence staining
 P30, P60
Self grooming3
Increased
Description: Increased grooming episodes resulted in skin lesions
 Grooming behavior assessments
 Adult
Stereotypy3
Increased
Description: Mild or severe hind paw clasping when suspended by tails
 Observation of repetitive behavior
 Adult
Seizure threshold1
Decreased
Description: Male and female mutant mice displayed much more severe epileptic behaviors including heavy myoclonic jerks, lying on belly with rapid body twitches, and clonic-tonic spasm. Mutant mice had spike-wave electrical discharges with increased amplitude, which was consistent with the severe epileptic behaviors observed in these mice.
Exp Paradigm: intraperitoneal injection of pentylenetetrazole
 Electroencephalogram (EEG)
 month 2
Social memory3
Decreased
Description: No preference for the novel social stimulus
 Three-chamber social approach test
 Adult
Social approach3
Decreased
Description: No preference for the social stimulus
 Three-chamber social approach test
 Adult
Metabolic function1
Increased
Description: Mutant mice had higher heat generation.
Exp Paradigm: TSE PhenoMaster/LabMaster System
 Energy homeostasis analysis
 month 4
Homeostasis: adiposity1
Decreased
Description: Mutant mice show markedly reduced subcutaneous and visceral adipose tissue depots. Change appeared to mainly affect white adipose tissues but not brown adipose tissues.
 Measurement of tissue weight
 month 8
Satiety response1
Decreased
Description: Mutant mice had higher food intake compared to their wildtype littermates.
Exp Paradigm: TSE PhenoMaster/LabMaster System
 Food intake measurements
 month 4
Respiratory function1
Increased
Description: Mutant mice had higher oxygen consumption, carbon dioxide production, respiration exchange ratios.
Exp Paradigm: TSE PhenoMaster/LabMaster System
 Respiration rate and pattern analysis
 month 4
Mortality/lethality: postnatal3
Increased
Description: Survived through early postnatal days; 10% died before weaning; the majority of surviving pups were able to grow to adulthood
 General observations
 P0-P21
Skeletal development: craniofacial3
Abnormal
Description: Abnormal craniofacial appearance; reduced eye-to-mouth distance caused by shortened nose bones
 Micro-CT scanning
 Adult
Size/growth3
Decreased
Description: No change in body weight at birth; growth retardation by 2-3 weeks; final body weight 5â??10% lower than that of the wild-type adult
 Body weight measurement
 P0-P21, Adult
Size/growth1
Decreased
Description: Adult mutant mice had significantly lower body weight.
 Body weight measurement
 month 8
Anxiety3
Increased
Description: Spent less time exploring the arena center
 Open field test
 Adult
Object recognition memory3
Decreased
Description: Reduced discrimination index
 Novel object recognition test
 Adult
Mortality/lethality2
 No change
 General observations
 P0-P2
 Not Reported:

M_ASH1L_7_CKO_HT

Category
Entity
Quantity
Experimental Paradigm
Age at Testing
Mortality/lethality
 No change
 
 
 Not Reported:

M_ASH1L_8_CKO_HM

Category
Entity
Quantity
Experimental Paradigm
Age at Testing
Gene expression1
Decreased
Description: Emx2, pcdh10, and foxg1 had similar reduced expression in both ash1l-ko cortices and differentiating npcs
 Quantitative PCR (qRT-PCR)
 E16.5
Differential gene expression1
Abnormal
Description: Abnormal gene expression levels (geo accession: gse173262)
 RNA sequencing
 E16.5
Targeted expression1
Decreased
Description: Ash1l protein expressed decreased
 Western blot
 Adult
Targeted expression1
Decreased
Description: Ash1l mrna expressed decreased
 Quantitative PCR (qRT-PCR)
 Adult
 Not Reported:

M_ASH1L_9_CKO_HM

Category
Entity
Quantity
Experimental Paradigm
Age at Testing
General locomotor activity1
decreased
Description: Ash1l cko mice showed deficits in exploratory behavior
 Open field test
 3-6-month
Synapse density1
abnormal
Description: compared with normal neurons, the elimination rate of axonal boutons in Ash1l-deficient neurons was significantly lower at 24 h after conditioning; no difference was detected for newly emerged boutons between Ash1l-deficient and Ash1l-normal neurons
Exp Paradigm: injected AAV-hSyn-Cre and AAVEF1a-DIO-EYFP into the right side of AUD
 Two-photon microscopy
 3-6-month
Cued or contextual fear conditioning: memory of cue1
abnormal
Description: Ash1l cko mice showed comparable freezing to WT in response to the CS+ stimulus but CS- provoked higher level of freezing in the Ash1l cko mice; Ash1l cko showed significantly impaired discrimination ability
 Fear conditioning test
 3-6-month
Social memory1
decreased
Description: Ash1l cko mice showed deficits in social novelty discrimination
 Three-chamber social approach test
 3-6-month
Anxiety1
increased
Description: Ash1l cko mice spent less time at the center of the open field
 Open field test
 3-6-month
Anxiety1
increased
Description: Ash1l cko mice explored the open arms of the maze significanly less than the closed arms
 Elevated plus maze
 3-6-month
Targeted expression1
decreased
Description: Protein levels of Ash1l were consistently reduced in cortex and hippocampus but not in striatum
 Western blot
 3-6-month
Social approach1
 no change
 Three-chamber social approach test
 3-6-month
 Not Reported:


Interactor Symbol Interactor Name Interactor Organism Entrez ID Uniprot ID Interaction Type Evidence Reference
CHD8 chromodomain helicase DNA binding protein 8 57680 Q9HCK8 CHIP-seq
Cotney J , et al. 2015
H3F3A H3 histone, family 3A 3020 P84243 in vitro methylation assay; M2 affinity gel assay; MS
Heo K , et al. 2007
h3f3a H3 histone, family 3A 399418 Q6PI79 in vitro methylation assay
Yuan G , et al. 2013
HIST1H3A histone cluster 1, H3a 8350 P68431 ChIP; in vitro methylation assay; GST; IP/WB
Gregory GD , et al. 2007
HOXB6 homeobox B6 3216 P17509 ChIP-qPCR; Genome-wide RNAi screen; qRT-PCR
Tanaka Y , et al. 2011
HOXC8 homeobox C8 NM_022658 P31273 ChIP-qPCR; Luciferase reporter assay; Genome-wide RNAi screen; qRT-PCR
Tanaka Y , et al. 2011
MIR142 microRNA 142 406934 Luciferase reporter assay; Ectopic expression/WB
Colamaio M , et al. 2014
MORF4L1 mortality factor 4 like 1 10933 B7Z6R1 IP; LC-MS/MS
Huttlin EL , et al. 2015
MORF4L2 mortality factor 4 like 2 9643 Q15014 IP; LC-MS/MS
Huttlin EL , et al. 2015
NXF2 Nuclear RNA export factor 2 56001 Q9GZY0 IP; LC-MS/MS
Huttlin EL , et al. 2015
SMAD7 SMAD family member 7 4092 K7EQ10 Y2H
Colland F , et al. 2004
SUMO2 SMT3 suppressor of mif two 3 homolog 2 (S. cerevisiae) 6613 P61956 SILAC; TAP; LC-MS/MS
Tatham MH , et al. 2011
THAP7 THAP domain containing 7 80764 Q9BT49 IP; LC-MS/MS
Huttlin EL , et al. 2015
TOP3B topoisomerase (DNA) III beta 8940 O95985 HITS-CLIP
Xu D , et al. 2013
UBC ubiquitin C 7316 P63279 LC-MS/MS
Danielsen JM , et al. 2010
FMR1 fragile X mental retardation 1 14265 P35922 HITS-CLIP
Darnell JC , et al. 2011
H3F3A H3 histone, family 3A 3020 P84243 in vitro methylation assay
Miyazaki H , et al. 2013
Hoxd4 homeobox D4 15436 P10628 ChIP-qPCR; qRT-PCR
Miyazaki H , et al. 2013
Tnfaip3 tumor necrosis factor, alpha-induced protein 3 21929 Q60769 ChIP-qPCR; Genome-wide RNAi screen
Xia M , et al. 2013
DBET D4Z4 binding element transcript (non-protein coding) 100419743 RNA immunoprecipitation (RIP)/qRT-PCR; RNA pull down/qRT-PCR
Cabianca DS , et al. 2012

HELP
Copyright © 2017 MindSpec, Inc.