HELP     Sign In
Search

Relevance to Autism

This gene was identified by TADA (transmission and de novo association) analysis of a combined dataset from the Simons Simplex Collection (SSC) and the Autism Sequencing Consortium (ASC) as a gene strongly enriched for variants likely to affect ASD risk with a false discovery rate (FDR) of < 0.1 (Sanders et al., 2015); among the variants identified in this gene was one de novo loss-of-function (LoF) variant. Additional de novo protein-truncating variants and missense variants in the DNMT3A gene were identified in ASD probands from the SPARK cohort (Feliciano et al., 2019) and the Autism Sequencing Consortium (Satterstrom et al., 2020); TADA analysis in both studies identified DNMT3A as a candidate gene with a false discovery rate (FDR) 0.01. 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 DNMT3A as a gene reaching exome-wide significance (P < 2.5E-06). Heterozygous variants in the DNMT3A gene are responsible for Tatton-Brown-Rahman syndrome (OMIM 615879), an overgrowth intellectual disability syndrome characterized by tall stature, increased head circumference, and distinctive facial appearance (Tatton-Brown et al., 2014). Clinical review of 55 individuals with Tatton-Brown-Rahman syndrome resulting from de novo DNMT3A variants in Tatton-Brown et al., 2018 determined that autism spectrum disorder (ASD) was observed in 20 individuals. De novo gain-of-function missense variants in the DNMT3A gene were observed in three patients presenting with microcephalic dwarfism and developmental delay (Heyn et al., 2018).

Molecular Function

The protein encoded by this gene is required for genome-wide de novo methylation and is essential for the establishment of DNA methylation patterns during development. It plays a role in paternal and maternal imprinting.

External Links

        

References

Type
Title
Type of Disorder
Associated Disorders
Author, Year
Primary
Insights into Autism Spectrum Disorder Genomic Architecture and Biology from 71 Risk Loci.
ASD
Positive Association
Genetic association of DNMT variants can play a critical role in defining the methylation patterns in autism.
ASD
Support
The contribution of de novo coding mutations to autism spectrum disorder
ASD
Support
Integrating de novo and inherited variants in 42
ASD
Support
Exome sequencing of 457 autism families recruited online provides evidence for autism risk genes
ASD
Support
Functional and epigenetic phenotypes of humans and mice with DNMT3A Overgrowth Syndrome
Tatton-Brown-Rahman syndrome
ASD, ADHD, OCD, DD, ID
Support
Synaptic, transcriptional and chromatin genes disrupted in autism.
ASD
Support
Complex Diagnostics of Non-Specific Intellectual Developmental Disorder
DD, ID
Support
Whole genome sequencing and variant discovery in the ASPIRE autism spectrum disorder cohort.
ASD
Support
Tatton-Brown-Rahman syndrome, ADHD, DD
Support
Tatton-Brown-Rahman syndrome with a novel DNMT3A mutation presented severe intellectual disability and autism spectrum disorder
Tatton-Brown-Rahman syndrome
ASD, ID
Support
Mutations in the DNA methyltransferase gene DNMT3A cause an overgrowth syndrome with intellectual disability.
Tatton-Brown-Rahman syndrome
Support
Impaired Neurodevelopmental Genes in Slovenian Autistic Children Elucidate the Comorbidity of Autism With Other Developmental Disorders
ASD
OCD, DD, ID
Support
Gain-of-function DNMT3A mutations cause microcephalic dwarfism and hypermethylation of Polycomb-regulated regions.
DD
Microcephaly, short stature
Support
Tatton-Brown-Rahman syndrome, DD
ID, epilepsy/seizures, autistic features
Support
Rare genetic susceptibility variants assessment in autism spectrum disorder: detection rate and practical use.
ASD
Support
Detection of clinically relevant genetic variants in autism spectrum disorder by whole-genome sequencing.
ASD
Support
The disordered N-terminal domain of DNMT3A recognizes H2AK119ub and is required for postnatal development
Support
Whole genome sequencing resource identifies 18 new candidate genes for autism spectrum disorder
ASD
Support
Heyn-Sproul-Jackson syndrome, DD
Support
Large-Scale Exome Sequencing Study Implicates Both Developmental and Functional Changes in the Neurobiology of Autism
ASD
Support
Diagnostic yield of patients with undiagnosed intellectual disability
DD, ID
Support
Candidate-gene criteria for clinical reporting: diagnostic exome sequencing identifies altered candidate genes among 8% of patients with undiagnose...
ID
Support
Tatton-Brown-Rahman syndrome: cognitive and behavioural phenotypes.
Tatton-Brown-Rahman syndrome
Autistic traits
Support
New Candidates for Autism/Intellectual Disability Identified by Whole-Exome Sequencing
DD, ID
Support
Meta-analysis of 2,104 trios provides support for 10 new genes for intellectual disability
ID
Support
ADHD, DD, ID
Autistic features
Support
Further delineation of neuropsychiatric findings in Tatton-Brown-Rahman syndrome due to disease-causing variants in DNMT3A: seven new patients.
Tatton-Brown-Rahman syndrome
SCZ
Support
Prevalence and phenotypic impact of rare potentially damaging variants in autism spectrum disorder
ASD
Recent Recommendation
The Tatton-Brown-Rahman Syndrome: A clinical study of 55 individuals with de novo constitutive DNMT3A variants.
Tatton-Brown-Rahman syndrome
ASD
Recent Recommendation
ASD, ID

Rare

Variant ID
Variant Type
Allele Change
Residue Change
Inheritance Pattern
Inheritance Association
Family Type
Author, Year
 GEN762R001 
 frameshift_variant 
 c.402dup 
 p.Gly135TrpfsTer17 
 De novo 
  
 Simplex 
 GEN762R002 
 missense_variant 
 c.2204A>G 
 p.Tyr735Cys 
 De novo 
  
  
 GEN762R003 
 missense_variant 
 c.2711C>T 
 p.Pro904Leu 
 De novo 
  
 Simplex 
 GEN762R004 
 missense_variant 
 c.1993G>T 
 p.Val665Leu 
 De novo 
  
 Simplex 
 GEN762R005 
 missense_variant 
 c.2644C>T 
 p.Arg882Cys 
 De novo 
  
  
 GEN762R006 
 missense_variant 
 c.1903C>T 
 p.Arg635Trp 
 De novo 
  
 Simplex 
 GEN762R007 
 missense_variant 
 c.892G>T 
 p.Gly298Trp 
 De novo 
  
  
 GEN762R008 
 frameshift_variant 
  
  
 De novo 
  
  
 GEN762R009 
 missense_variant 
 c.541C>T 
 p.Arg181Cys 
 De novo 
  
  
 GEN762R010 
 frameshift_variant 
 c.421dup 
 p.Glu141GlyfsTer11 
 De novo 
  
  
 GEN762R011 
 inframe_deletion 
 c.551_553del 
 p.Pro184_Met185delinsLeu 
 De novo 
  
  
 GEN762R012 
 missense_variant 
 c.892G>T 
 p.Gly298Trp 
 De novo 
  
  
 GEN762R013 
 missense_variant 
 c.892G>A 
 p.Gly298Arg 
 De novo 
  
  
 GEN762R014 
 missense_variant 
 c.901C>T 
 p.Arg301Trp 
 De novo 
  
  
 GEN762R015 
 stop_gained 
 c.580G>A 
 p.Asp194Asn 
 De novo 
  
  
 GEN762R016 
 missense_variant 
 c.929T>A 
 p.Ile310Asn 
 De novo 
  
  
 GEN762R017 
 frameshift_variant 
 c.596_597insGCAA 
 p.Ser199ArgfsTer18 
 De novo 
  
  
 GEN762R018 
 stop_gained 
 c.941G>A 
 p.Trp314Ter 
 De novo 
  
  
 GEN762R019 
 frameshift_variant 
 c.221del 
 p.Ala74ValfsTer90 
 De novo 
  
  
 GEN762R020 
 missense_variant 
 c.1154C>T 
 p.Pro385Leu 
 De novo 
  
  
 GEN762R021 
 stop_gained 
 c.502C>G 
 p.Arg168Gly 
 De novo 
  
  
 GEN762R022 
 stop_gained 
 c.1320G>A 
 p.Trp440Ter 
 De novo 
  
  
 GEN762R023 
 missense_variant 
 c.729T>C 
 p.Thr243= 
 De novo 
  
  
 GEN762R024 
 missense_variant 
 c.1594G>A 
 p.Gly532Ser 
 De novo 
  
  
 GEN762R025 
 missense_variant 
 c.1594G>A 
 p.Gly532Ser 
 De novo 
  
  
 GEN762R026 
 missense_variant 
 c.1643T>A 
 p.Met548Lys 
 De novo 
  
  
 GEN762R027 
 missense_variant 
 c.1643T>C 
 p.Met548Thr 
 De novo 
  
  
 GEN762R028 
 missense_variant 
 c.1645T>C 
 p.Cys549Arg 
 De novo 
  
  
 GEN762R029 
 missense_variant 
 c.1684T>C 
 p.Cys562Arg 
 De novo 
  
  
 GEN762R030 
 missense_variant 
 c.1743G>C 
 p.Trp581Cys 
 De novo 
  
  
 GEN762R031 
 missense_variant 
 c.1743G>C 
 p.Trp581Cys 
 De novo 
  
  
 GEN762R032 
 missense_variant 
 c.1748G>A 
 p.Cys583Tyr 
 De novo 
  
  
 GEN762R033 
 stop_gained 
 c.1803G>A 
 p.Trp601Ter 
 De novo 
  
  
 GEN762R034 
 splice_site_variant 
 c.1395+3G>C 
  
 De novo 
  
  
 GEN762R035 
 missense_variant 
 c.1943T>C 
 p.Leu648Pro 
 De novo 
  
  
 GEN762R036 
 frameshift_variant 
 c.1262del 
 p.Gln421ArgfsTer78 
 De novo 
  
  
 GEN762R037 
 missense_variant 
 c.2094G>C 
 p.Trp698Cys 
 De novo 
  
  
 GEN762R038 
 missense_variant 
 c.2099C>T 
 p.Pro700Leu 
 De novo 
  
  
 GEN762R039 
 missense_variant 
 c.2141C>G 
 p.Ser714Cys 
 De novo 
  
  
 GEN762R040 
 missense_variant 
 c.2204A>C 
 p.Tyr735Ser 
 De novo 
  
  
 GEN762R041 
 missense_variant 
 c.2207G>A 
 p.Arg736His 
 De novo 
  
  
 GEN762R042 
 missense_variant 
 c.2245C>T 
 p.Arg749Cys 
 De novo 
  
  
 GEN762R043 
 missense_variant 
 c.2245C>T 
 p.Arg749Cys 
 De novo 
  
  
 GEN762R044 
 missense_variant 
 c.1452G>A 
 p.Val484= 
 De novo 
  
  
 GEN762R045 
 inframe_deletion 
 c.1799_1801del 
 p.Phe600del 
 De novo 
  
  
 GEN762R046 
 frameshift_variant 
 c.1505dup 
 p.Ile503HisfsTer13 
 De novo 
  
  
 GEN762R047 
 missense_variant 
 c.2309C>T 
 p.Ser770Leu 
 De novo 
  
  
 GEN762R048 
 missense_variant 
 c.2312G>A 
 p.Arg771Gln 
 De novo 
  
  
 GEN762R049 
 missense_variant 
 c.2401A>G 
 p.Met801Val 
 De novo 
  
  
 GEN762R050 
 missense_variant 
 c.1718A>G 
 p.Glu573Gly 
 De novo 
  
  
 GEN762R051 
 missense_variant 
 c.2188C>T 
 p.Arg730Cys 
 De novo 
  
  
 GEN762R052 
 missense_variant 
 c.2644C>T 
 p.Arg882Cys 
 De novo 
  
  
 GEN762R053 
 missense_variant 
 c.2644C>T 
 p.Arg882Cys 
 De novo 
  
  
 GEN762R054 
 missense_variant 
 c.2645G>A 
 p.Arg882His 
 De novo 
  
  
 GEN762R055 
 stop_gained 
 c.1867-284C>A 
  
 De novo 
  
  
 GEN762R056 
 missense_variant 
 c.1867-254T>C 
  
 De novo 
  
  
 GEN762R057 
 missense_variant 
 c.2711C>T 
 p.Pro904Leu 
 De novo 
  
  
 GEN762R058 
 missense_variant 
 c.2711C>T 
 p.Pro904Leu 
 De novo 
  
  
 GEN762R059 
 copy_number_loss 
  
  
 De novo 
  
  
 GEN762R060 
 copy_number_loss 
  
  
 De novo 
  
  
 GEN762R062 
 copy_number_loss 
  
  
 De novo 
  
  
 GEN762R063 
 missense_variant 
 c.988T>C 
 p.Trp330Arg 
 De novo 
  
  
 GEN762R064 
 missense_variant 
 c.988T>C 
 p.Trp330Arg 
 De novo 
  
  
 GEN762R065 
 missense_variant 
 c.997G>A 
 p.Asp333Asn 
 De novo 
  
  
 GEN762R066 
 missense_variant 
 c.1907C>T 
 p.Ala636Val 
 Unknown 
  
 Simplex 
 GEN762R067 
 missense_variant 
 c.1660G>A 
 p.Gly554Arg 
 De novo 
  
  
 GEN762R068 
 missense_variant 
 c.1570C>T 
 p.Arg524Trp 
 De novo 
  
  
 GEN762R069 
 frameshift_variant 
 c.2246_2247del 
 p.Arg749ProfsTer7 
 De novo 
  
  
 GEN762R070 
 splice_site_variant 
 c.1211+2T>G 
  
 De novo 
  
  
 GEN762R071 
 missense_variant 
 c.2209C>T 
 p.Leu737Phe 
 Unknown 
  
  
 GEN762R072 
 missense_variant 
 c.2207G>A 
 p.Arg736His 
 De novo 
  
  
 GEN762R073 
 missense_variant 
 c.1627G>A 
 p.Gly543Ser 
 De novo 
  
  
 GEN762R074 
 stop_gained 
 c.1681G>T 
 p.Glu561Ter 
 De novo 
  
  
 GEN762R075 
 missense_variant 
 c.919C>T 
 p.Pro307Ser 
 De novo 
  
  
 GEN762R076 
 frameshift_variant 
 c.2296_2297del 
 p.Lys766GlufsTer15 
 De novo 
  
 Simplex 
 GEN762R077 
 missense_variant 
 c.1067T>C 
 p.Leu356Pro 
 De novo 
  
 Simplex 
 GEN762R078 
 missense_variant 
 c.1658T>C 
 p.Ile553Thr 
 De novo 
  
 Simplex 
 GEN762R079 
 missense_variant 
 c.1447C>T 
 p.Arg483Trp 
 De novo 
  
 Simplex 
 GEN762R080 
 stop_gained 
 c.745C>T 
 p.Gln249Ter 
 De novo 
  
 Simplex 
 GEN762R081 
 missense_variant 
 c.1904G>A 
 p.Arg635Gln 
 De novo 
  
 Simplex 
 GEN762R082 
 missense_variant 
  
 p.Arg882His 
 Unknown 
  
  
 GEN762R083 
 missense_variant 
  
 p.Arg882His 
 Unknown 
  
  
 GEN762R084 
 missense_variant 
  
 p.Arg882Cys 
 Unknown 
  
  
 GEN762R085 
 missense_variant 
  
 p.Cys583Tyr 
 Unknown 
  
  
 GEN762R086 
 missense_variant 
  
 p.Arg301Trp 
 Unknown 
  
  
 GEN762R087 
 frameshift_variant 
  
 p.Phe414fsTer7 
 Unknown 
  
  
 GEN762R088 
 missense_variant 
  
 p.Arg736His 
 Unknown 
  
  
 GEN762R089 
 missense_variant 
  
 p.Arg688His 
 Unknown 
  
  
 GEN762R090 
 missense_variant 
  
 p.Tyr660His 
 Unknown 
  
  
 GEN762R091 
 copy_number_loss 
  
  
 Unknown 
  
  
 GEN762R092 
 missense_variant 
  
 p.Ile310Asn 
 Unknown 
  
  
 GEN762R093 
 missense_variant 
 c.1523T>C 
 p.Leu508Pro 
 Unknown 
  
  
 GEN762R094 
 missense_variant 
 c.1597G>A 
 p.Gly533Arg 
 De novo 
  
 Simplex 
 GEN762R095 
 copy_number_loss 
  
  
 De novo 
  
  
 GEN762R096 
 missense_variant 
 c.1969G>A 
 p.Val657Met 
 Unknown 
 Not maternal 
  
 GEN762R097 
 stop_gained 
 c.1443C>A 
 p.Tyr481Ter 
 De novo 
  
 Simplex 
 GEN762R098 
 missense_variant 
 c.2204A>G 
 p.Tyr735Cys 
 De novo 
  
 Simplex 
 GEN762R099 
 missense_variant 
 c.2711C>T 
 p.Pro904Leu 
 De novo 
  
  
 GEN762R100 
 missense_variant 
 c.2012C>T 
 p.Thr671Met 
 De novo 
  
  
 GEN762R101 
 inframe_insertion 
 c.1127_1128insACGACGACGACGGCTACCAGT 
 p.Tyr376delinsTer 
 De novo 
  
  
 GEN762R102 
 splice_region_variant 
 c.1015-3C>G 
  
 De novo 
  
  
 GEN762R103 
 missense_variant 
 c.2711C>T 
 p.Pro904Leu 
 De novo 
  
  
 GEN762R104 
 missense_variant 
 c.2204A>C 
 p.Tyr735Ser 
 De novo 
  
  
 GEN762R105 
 missense_variant 
 c.2050G>A 
 p.Val684Ile 
 De novo 
  
  
 GEN762R106 
 missense_variant 
 c.1919T>C 
 p.Phe640Ser 
 De novo 
  
  
 GEN762R107 
 missense_variant 
 c.1904G>T 
 p.Arg635Leu 
 De novo 
  
  
 GEN762R108 
 splice_site_variant 
 c.1668-1G>A 
  
 De novo 
  
  
 GEN762R109 
 frameshift_variant 
 c.699dup 
 p.Gly234ArgfsTer19 
 De novo 
  
  
 GEN762R110 
 missense_variant 
 c.1034G>T 
 p.Cys345Phe 
 De novo 
  
  
 GEN762R111 
 frameshift_variant 
 c.387del 
 p.Glu129AspfsTer35 
 De novo 
  
  
 GEN762R112 
 splice_site_variant 
 c.556_558+3del 
  
 De novo 
  
 Simplex 
 GEN762R113 
 missense_variant 
 c.2207G>A 
 p.Arg736His 
 De novo 
  
 Simplex 
  et al.  
 GEN762R114 
 splice_site_variant 
 c.2141+1G>A 
  
 De novo 
  
 Simplex 
  et al.  
 GEN762R115 
 missense_variant 
 c.1481G>C 
 p.Cys494Ser 
 De novo 
  
 Multiplex (monozygotic twins) 
  et al.  
 GEN762R116 
 missense_variant 
 c.2036G>A 
 p.Gly679Glu 
 De novo 
  
 Simplex 
  et al.  

Common

Variant ID
Polymorphism
SNP ID
Allele Change
Residue Change
Population Origin
Population Stage
Author, Year
 GEN762C001 
 intron_variant 
 rs2289195 
 c.1717+26C>T;c.2173+26C>T;c.1606+26C>T 
  
 180 autistic patients and 260 healthy controls from a south Indian population 
 Discovery 
Chromosome
CNV Locus
CNV Type
# of studies
Animal Model
2
Deletion
 13
 
2
Duplication
 1
 
2
Duplication
 1
 

Model Summary

Heterozygous DNMT3A knockout mice display growth and behavioral alterations consistent with human phenotypes.

References

Type
Title
Author, Year
Primary
DNA methyltransferases Dnmt3a and Dnmt3b are essential for de novo methylation and mammalian development
Primary
DNMT3A Haploinsufficiency Results in Behavioral Deficits and Global Epigenomic Dysregulation Shared across Neurodevelopmental Disorders
Additional

M_DNMT3A_1_KO_HM

Model Type: Genetic LOF
Model Genotype: Homozygous
Mutation: A 0.3kb region encoding the pc and env motifs of dnmt3a was replaced with a cassette containing an ires-lacz gene and a neo gene. northern blot analysis of total rna showed an absence of transcript in homozygous mutant mice.
Allele Type: Knockout
Strain of Origin: 129S4/SvJae
Genetic Background: 129S4/SvJae
ES Cell Line: NA
Mutant ES Cell Line: J1
Model Source: 33238114

M_DNMT3A_2_KO_HT

Model Type: Genetic LOF
Model Genotype: Heterozygous
Mutation: Mice carrying a constitutive heterozygous deletion of exon 19 of dnmt3a were generated by crossing mice derived from dnmt3a fl/fl strain and cmv-cre (imsr cat# jax:006054, rrid:imsr_jax:006054; balb/cj) mice obtained from the jackson laboratory.
Allele Type: Knockout
Strain of Origin: 129S4/SvJae
Genetic Background: 129S4/SvJae*BALB/cJ
ES Cell Line: NA
Mutant ES Cell Line: J1
Model Source: Kaneda et al., 2004 (Provided by M. Goodell)

M_DNMT3A_3_KI_HT_P904L

Model Type: Genetic
Model Genotype: Heterozygous
Mutation: The DNMT3A P900L mouse model was generated using single guide RNAs (sgRNAs) to create a cytosine to thymine substitution at chr12:3,907,719. This mutation changed the proline CCG codon into a leucine CTG codon, corresponding to the P904L human mutation. Founders were crossed to C57BL6/J females for 5-10 generations before experimental analysis.
Allele Type: ASD mutation
Strain of Origin: C57BL/6J x CBA
Genetic Background: C57BL6/J
ES Cell Line: N2A
Mutant ES Cell Line:
Model Source: Harrison W. Gabel

M_DNMT3A_4_KI_HT_R882H

Model Type: Genetic
Model Genotype: Heterozygous
Mutation: Heterozygous mice carry one allele with a guanine to adenine substitution which results in a arginine to histidine mutation in amino acid 878, which corresponds to the R882H human mutation found in patients with acute myeloid leukemia. Mutant male mice were crossed with C57BL6/J female mice.
Allele Type: Humanized mutation
Strain of Origin: C57BL/6NTac
Genetic Background: C57BL6/J
ES Cell Line: iTL IC1
Mutant ES Cell Line:
Model Source: Timothy J. Ley

M_DNMT3A_1_KO_HM

Category
Entity
Quantity
Experimental Paradigm
Age at Testing
Mortality/lethality: postnatal: incomplete penetrance1
Increased
Description: Most die at about 4 weeks
 General observations
 4 weeks
Size/growth1
Decreased
Description: Decreased size
 General observations
 P18
Targeted expression1
Decreased
Description: Genomic excision of dnmt3a
 Southern blot
 E9.5
DNA methylation1
Decreased
Description: Decreased global dna methylation
 Bisulfite sequencing
 E9.5
 Not Reported:

M_DNMT3A_2_KO_HT

Category
Entity
Quantity
Experimental Paradigm
Age at Testing
General locomotor activity: Ambulatory activity1
Decreased
Description: Decrease in ambulation
 Open field test
 Adult
Climbing1
Decreased
Description: Increase in time taken to climb to the top of a 60 deg and 90 deg inclined screen
 Climbing assay
 Adult
Rearing behavior1
Decreased
Description: Decreased rearings
 Open field test
 Adult
Repetitive digging1
Decreased
Description: Fewer marbles buried
 Morris water maze test
 Adult
Social approach1
Abnormal
Description: Mice spend reduced time investigating mice and objects, with no change in preference index for sociability or novelty under low light
 Three-chamber social approach test
 Adult
Ultrasonic vocalization: Isolation induced1
Decreased
Description: Decreased in calls at p5, p20
 Monitoring ultrasonic vocalizations
 P5, P20
Skeletal development: craniofacial1
Abnormal
Description: Abnormal cranial and mandible morphometric measurements
 Dual-energy X-ray absorptiometry for bone mineral density measurement
 8-25 weeks
Skeletal development: Appendages (limbs, digits, tail)1
Increased
Description: Increased tibia length and a trend toward longer femur length
 Dual-energy X-ray absorptiometry for bone mineral density measurement
 8-25 weeks
Size/growth1
Increased
Description: Increased adult body weight; no change in postnatal body weight
 Body weight measurement
 Adult
Anxiety1
Increased
Description: Decrease in time spent in open arms; no change in number of entries into open arms
 Elevated T-maze test
 Adult
Fear response1
Increased
Description: Increased freezing response during training
 Fear conditioning test
 Adult
Exploratory activity1
Decreased
Description: Decreased exploration
 Open field test
 Adult
Anxiety1
Increased
Description: Decrease in time spent in the center
 Open field test
 Adult
Object recognition memory1
Decreased
Description: Mice spend reduced time investigating objects
 Three-chamber social approach test
 Adult
Extinction of fear memory1
Decreased
Description: Delayed extinction of freezing behavior in response to auditory cue alone
 Fear conditioning test
 Adult
Cued or contextual fear conditioning: Memory of cue1
Increased
Description: Increase in time spent freezing
 Fear conditioning test
 Adult
Spatial working memory1
Decreased
Description: Slower progressive decrease in path distance to escape platform; mice did not learn the position of the hidden platform
 Morris water maze test
 Adult
Cued or contextual fear conditioning: Memory of context1
Increased
Description: Increase in time spent freezing
 Fear conditioning test
 Adult
Differential gene expression1
Abnormal
Description: Genes associated with axon guidance and recognition are differentially expressed; reduced expression of asd genes shank2 and shank3, upregulation of shroom3, latrophilin-2; geo: gse147899
 RNA sequencing
 Adult
Targeted expression1
Decreased
Description: 50% reduction of protein expression
 Western blot
 2 weeks
Chromatin modification: histone acetylation1
Increased
Description: Significant acetylation changes at mecp2-repressed enhancers; enhancers within high mca tads show significantly higher increases in acetylation compared with enhancers in other tads; geo: gse147899
 Chromatin immunoprecipitation sequencing (ChIP-seq)
 Adult
Regulation of gene expression1
Decreased
Description: Differentially methylated cg and ca regions in regulatory elements in the brain; enhancer dysregulation; mecp2-repressed enhancers are particularly susceptible to mca binding site loss; broad mca-associated derepression of enhancers in cx; geo: gse147899
 Bisulfite sequencing
 Adult
Chromatin modification1
Abnormal
Description: Decreased mca methylation of topologically associated domains
 Bisulfite sequencing
 Adult
Transcriptome diversity1
Abnormal
Description: Transcriptomic changes due to differential mcg enhancer methylation
 RNA sequencing
 Adult
DNA methylation1
Decreased
Description: Reduced deposition of neuronal mca throughout the brain; decreased global methylation in mcg in brain but not liver
 Bisulfite sequencing
 Adult
Targeted expression1
Decreased
Description: 50% reduction of rna expression
 Quantitative PCR (qRT-PCR)
 2 weeks
Spatial reference memory1
 No change
 Morris water maze test
 Adult
Grip strength1
 No change
 Inverted grid test
 Adult
Motor coordination and balance1
 No change
 Ledge test
 Adult
Motor coordination and balance1
 No change
 Balance beam test
 Adult
Swimming ability1
 No change
 Morris water maze test
 Adult
Sensorimotor gating1
 No change
 Prepulse inhibition
 Adult
Pain or nociception1
 No change
 Foot shock test
 Adult
 Not Reported:

M_DNMT3A_3_KI_HT_P904L

Category
Entity
Quantity
Experimental Paradigm
Age at Testing
DTI: fractional anisotropy or relative anisotropy in brain regions1
Decreased
Description: P900L mutant mice show a significant decrease in fractional anisotropy.
 Magnetic resonance imaging (MRI)
 8 weeks
Cortical thickness1
Decreased
Description: Heterozygous P900L mice exhibit broad reductions in cortical thickness.
 Magnetic resonance imaging (MRI)
 8 weeks
Brain size1
Decreased
Description: MRI in aged animals revealed significantly reduced brain volume in late adulthood in mutants compared to wildtype mice.
 Magnetic resonance imaging (MRI)
 30-35 weeks
Tactile memory1
Decreased
Description: Tactile discrimination was measured using a textured novel object recognition task. While wildtype mice display preference for novel tactile objects, P900L mutants show no preference.
 Novel texture discrimination
 8 weeks
Homeostasis: adiposity1
Increased
Description: Heterozygous P900L mice show significantly increased fat mass with no change in lean mass, indicating an obesity phenotype.
Exp Paradigm: EchoMRI
 Magnetic resonance imaging (MRI)
 8 weeks
Ultrasonic vocalization: isolation induced1
Decreased
Description: P900L mutant pups make significantly fewer calls when removed from the nest, indicating deficits in early communication behaviors. Mutant pups show decreased volume, and no change in frequency or duration compared to wildtype pups.
 Monitoring ultrasonic vocalizations
 P5-P9
Skeletal development: appendages (limbs, digits, tail)1
Increased
Description: Measurement of femur length using X-ray imaging revealed that heterozygote P900L femurs are significantly longer than those of wildtype littermates.
 Skeletal X-rays
 8 weeks
Procedural learning1
Decreased
Description: P900L mutant mice show a slight increase in path length upon initial task exposure (first cued trial with visible platform) of the Morris water maze, but they show no difference in cued trials 2-4.
Exp Paradigm: visible platform
 Morris water maze test
 8 weeks
Differential gene expression1
Abnormal
Description: RNA sequencing analysis of cerebral cortex from 8-week-old animals was used to define transcriptional alterations in Dnmt3a mutants. P900L mutants displayed significant gene expression changes: 444 upregulated, 182 downregulated.
 RNA sequencing
 8 weeks
Targeted expression1
Decreased
Description: Heterozygous P900L mice show a reduction in DNMT3A protein expression compared to wildtype mice.
 Western blot
 8 weeks
Chromatin modification: histone acetylation1
Increased
Description: Enhancer activity was examined using chromatin immunoprecipitation sequencing (ChIP-seq) analysis of histone H3 lysine 27 acetylation (H3K27ac; a histone modification correlated with active enhancers) in the cortex of 8-week-old P900L mutants Quantification of differential acetylation did not detect significantly altered enhancers in the P900L mutant. Enhancers containing hypo-CG differentially methylated regions showed a trend toward upregulation in the P900L mutant.
 Chromatin immunoprecipitation sequencing (ChIP-seq)
 8 weeks
Protein-DNA complex assembly1
Decreased
Description: ChIP-seq measured changes in MeCP2 binding at enhancers genome-wide. Enhancers with a high density of wildtype methyl-CA sites (mCA/kb) exhibited the largest loss of methyl-CA and MeCP2 binding in P900L mutants.
 Chromatin immunoprecipitation sequencing (ChIP-seq)
 8 weeks
DNA methylation1
Decreased
Description: 
 Bisulfite sequencing
 8 weeks
General characteristics1
 No change
 General observations
 30 weeks
Size/growth1
 No change
 Body weight measurement
 10-30 weeks
Skeletal development: craniofacial1
 No change
 Cranial computerized tomography (CT) scan
 8 weeks
Anxiety1
 No change
 Open field test
 8 weeks
Anxiety1
 No change
 Elevated plus maze test
 8 weeks
Fear response1
 No change
 Fear conditioning test
 8 weeks
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
Object recognition memory1
 No change
 Novel object recognition test
 8 weeks
Spatial learning1
 No change
 Morris water maze test
 8 weeks
Spatial reference memory1
 No change
 Morris water maze test
 8 weeks
Targeted expression1
 No change
 Quantitative PCR (qRT-PCR)
 8 weeks
General locomotor activity: ambulatory activity1
 No change
 Open field test
 8 weeks
General locomotor activity: ambulatory activity1
 No change
 Three-chamber social approach test
 8 weeks
Grip strength1
 No change
 Inverted grid test
 8 weeks
Motor coordination and balance1
 No change
 Ledge test
 8 weeks
Motor coordination and balance1
 No change
 Accelerating rotarod test
 8 weeks
Motor coordination and balance1
 No change
 Vertical pole test
 8 weeks
Brain size1
 No change
 Magnetic resonance imaging (MRI)
 P10, 8 weeks
Brain size1
 No change
 Histology
 8 weeks
Morphology and size of the corpus callosum1
 No change
 Magnetic resonance imaging (MRI)
 8 weeks
Neuronal number1
 No change
 Histology
 8 weeks
Size of cerebral ventricles1
 No change
 Magnetic resonance imaging (MRI)
 8 weeks
Satiety response1
 No change
 Food intake measurements
 30 weeks
Repetitive digging1
 No change
 Marble-burying test
 8 weeks
Social approach1
 No change
 Three-chamber social approach test
 10-12 weeks
Social dominance1
 No change
 Tube test of social dominance
 10-12 weeks
Social memory1
 No change
 Three-chamber social approach test
 10-12 weeks
 Not Reported:

M_DNMT3A_4_KI_HT_R882H

Category
Entity
Quantity
Experimental Paradigm
Age at Testing
Cortical thickness1
Decreased
Description: Heterozygous R787H mice exhibit broad reductions in cortical thickness.
 Magnetic resonance imaging (MRI)
 8 weeks
Brain size1
Decreased
Description: MRI in aged animals revealed significantly reduced brain volume in late adulthood in mutants compared to wildtype mice.
 Magnetic resonance imaging (MRI)
 30-35 weeks
Size of cerebral ventricles1
Decreased
Description: R787H mutant mice had subtle reductions in ventricular volume.
 Magnetic resonance imaging (MRI)
 8 weeks
Tactile memory1
Decreased
Description: Tactile discrimination was measured using a textured novel object recognition task. While wildtype mice display preference for novel tactile objects, R787H mutants show no preference.
 Novel texture discrimination
 8 weeks
Social dominance1
Increased
Description: R787H mutants displayed social dominance, with mutants overwhelmingly winning bouts in the tube test over wildtype mice.
 Tube test of social dominance
 10-12 weeks
Ultrasonic vocalization: isolation induced1
Decreased
Description: Isolation-induced ultrasonic vocalizations are reduced in R878H mutants compared to WT littermates. Mutants show decreased frequency, but no change in volume or duration, compared to wildtype.
 Monitoring ultrasonic vocalizations
 P5-P9
Object recognition memory1
Decreased
Description: Unlike wildtype mice, R787H mutant mice show no preference for visually distinct novel objects.
 Novel object recognition test
 8 weeks
DNA methylation1
Decreased
Description: 
 Bisulfite sequencing
 8 weeks
Differential gene expression1
Abnormal
Description: RNA sequencing analysis of cerebral cortex from 8-week-old animals was used to define transcriptional alterations in Dnmt3a mutants. R787H mutants displayed significant gene expression changes: 797 upregulated, 960 downregulated.
 RNA sequencing
 8 weeks
Chromatin modification: histone acetylation1
Increased
Description: Enhancer activity was examined using chromatin immunoprecipitation sequencing (ChIP-seq) analysis of histone H3 lysine 27 acetylation (H3K27ac; a histone modification correlated with active enhancers40) in the cortex of 8-week-old R878H mutants. Quantification of differential acetylation shows 29 upregulated and 29 downregulated enhancers in the R878H mutant. Enhancers containing hypo-CG differentially methylated regions showed significant increases in H3K27ac in the R878H mutant.
 Chromatin immunoprecipitation sequencing (ChIP-seq)
 8 weeks
Protein-DNA complex assembly1
Decreased
Description: ChIP-seq measured changes in MeCP2 binding at enhancers genome-wide. Enhancers with a high density of wildtype methyl-CA sites (mCA/kb) exhibited the largest loss of methyl-CA and MeCP2 binding in R787H mutants.
 Chromatin immunoprecipitation sequencing (ChIP-seq)
 8 weeks
Size/growth1
 No change
 Body weight measurement
 8 weeks
Brain size1
 No change
 Magnetic resonance imaging (MRI)
 P10, 8 weeks
Brain size1
 No change
 Histology
 8 weeks
DTI: fractional anisotropy or relative anisotropy in brain regions1
 No change
 Magnetic resonance imaging (MRI)
 8 weeks
Morphology and size of the corpus callosum1
 No change
 Magnetic resonance imaging (MRI)
 8 weeks
Neuronal number1
 No change
 Histology
 8 weeks
 Not Reported:

 

No Interactions Available
HELP
Copyright © 2017 MindSpec, Inc.