HELP     Sign In
Search

Relevance to Autism

Rare variants in the MEF2C gene have been identified with autism (Novara et al., 2010; Neale et al., 2012) as well as with mental retardation and developmental delay.

Molecular Function

The encoded protein is a transcription factor involved in diverse developmental processes including hematopoiesis, cardiogenesis and neurogenesis.

External Links

        

References

Type
Title
Type of Disorder
Associated Disorders
Author, Year
Primary
Refining the phenotype associated with MEF2C haploinsufficiency.
ASD
MR, epilepsy
Positive Association
Common schizophrenia alleles are enriched in mutation-intolerant genes and in regions under strong background selection.
SCZ
Support
Rates, distribution and implications of postzygotic mosaic mutations in autism spectrum disorder.
ASD
Support
Patterns and rates of exonic de novo mutations in autism spectrum disorders.
ASD
Support
Epilepsy/seizures
Support
Neurological Diseases With Autism Spectrum Disorder: Role of ASD Risk Genes.
ASD
ID
Support
Large-scale discovery of novel genetic causes of developmental disorders.
Epilepsy/seizures
Support
Genotypes and Phenotypes of MEF2C Haploinsufficiency Syndrome: New Cases and Novel Point Mutations
DD, epilepsy/seizures
Autistic features, stereotypy
Support
Genomic diagnosis for children with intellectual disability and/or developmental delay.
ID, epilepsy/seizures
Macrocephaly, hypotonia
Support
Whole genome paired-end sequencing elucidates functional and phenotypic consequences of balanced chromosomal rearrangement in patients with develop...
ID, epilepsy/seizures
ASD
Support
Refining analyses of copy number variation identifies specific genes associated with developmental delay.
ASD, DD, ID
Support
Confirming the contribution and genetic spectrum of de novo mutation in infantile spasms: Evidence from a Chinese cohort
Epilepsy/seizures
ASD, DD
Support
Diagnostic Targeted Resequencing in 349 Patients with Drug-Resistant Pediatric Epilepsies Identifies Causative Mutations in 30 Different Genes.
Epilepsy/seizures
ASD
Support
Integrating de novo and inherited variants in 42
ASD
Support
Targeted resequencing of 358 candidate genes for autism spectrum disorder in a Chinese cohort reveals diagnostic potential and genotype-phenotype c...
ASD
Support
Targeted resequencing in epileptic encephalopathies identifies de novo mutations in CHD2 and SYNGAP1.
Epilepsy
ID, ASD, DD
Support
Large-scale targeted sequencing identifies risk genes for neurodevelopmental disorders
ASD, DD
Support
MEF2C regulates cortical inhibitory and excitatory synapses and behaviors relevant to neurodevelopmental disorders.
Support
Clinical findings from the landmark MEF2C-related disorders natural history study
Neurodevelopmental disorder with hypotonia, stereo
ASD, epilepsy/seizures, stereotypy
Support
Genome sequencing identifies multiple deleterious variants in autism patients with more severe phenotypes.
ASD
Support
MEF2C Haploinsufficiency features consistent hyperkinesis, variable epilepsy, and has a role in dorsal and ventral neuronal developmental pathways.
Epilepsy/seizures
Stereotypies, absent speech
Support
DD
ADHD
Support
Genes influenced by MEF2C contribute to neurodevelopmental disease via gene expression changes that affect multiple types of cortical excitatory neurons
Support
Mutations in Human Accelerated Regions Disrupt Cognition and Social Behavior.
ASD
Support
Autism spectrum disorder and comorbid neurodevelopmental disorders (ASD-NDDs): Clinical and genetic profile of a pediatric cohort
ASD
Epilepsy/seizures
Support
Novel MEF2C point mutations in Chinese patients with Rett (-like) syndrome or non-syndromic intellectual disability: insights into genotype-phenoty...
ID, epilepsy/seizures
Autistic features
Support
Multiple autism-linked genes mediate synapse elimination via proteasomal degradation of a synaptic scaffold PSD-95.
Support
Neurodevelopmental disorder with hypotonia, stereo
Support
MEF2C Hypofunction in Neuronal and Neuroimmune Populations Produces MEF2C Haploinsufficiency Syndrome-like Behaviors in Mice
Autosomal dominant mental retardation syndrome-20
ASD, stereotypy
Support
Genome-wide characteristics of de novo mutations in autism
ASD
Support
Prevalence and phenotypic impact of rare potentially damaging variants in autism spectrum disorder
ASD
Support
High Rate of Recurrent De Novo Mutations in Developmental and Epileptic Encephalopathies.
Epilepsy/seizures
DD/ID
Support
Refining the phenotype associated with MEF2C point mutations.
ID
Epilepsy, autistic behaviors
Support
ASD
Support
Disruption of chromatin organisation causes MEF2C gene overexpression in intellectual disability: a case report.
ID
DD, ADHD
Support
MEF2C haploinsufficiency syndrome: Report of a new MEF2C mutation and review.
ID, epilepsy/seizures
Autistic features
Support
A single center experience with publicly funded clinical exome sequencing for neurodevelopmental disorders or multiple congenital anomalies
ASD, DD, ID, epilepsy/seizures
Highly Cited
Control of mouse cardiac morphogenesis and myogenesis by transcription factor MEF2C.
Highly Cited
Activation of the transcription factor MEF2C by the MAP kinase p38 in inflammation.
Recent Recommendation
The genomic landscape of balanced cytogenetic abnormalities associated with human congenital anomalies.
DD, ID, epilepsy/seizures
ASD
Recent Recommendation
Regulation of progenitor cell proliferation and granulocyte function by microRNA-223.
Recent Recommendation
Foxp2 controls synaptic wiring of corticostriatal circuits and vocal communication by opposing Mef2c.
Recent Recommendation
ASD
Recent Recommendation
Clinically relevant single gene or intragenic deletions encompassing critical neurodevelopmental genes in patients with developmental delay, mental...
DD
Recent Recommendation
ASD
Recent Recommendation
Mutations in MEF2C from the 5q14.3q15 microdeletion syndrome region are a frequent cause of severe mental retardation and diminish MECP2 and CDKL5 ...
ID
Recent Recommendation
Transcriptional and functional consequences of alterations to MEF2C and its topological organization in neuronal models
Recent Recommendation
Fragile X mental retardation protein is required for synapse elimination by the activity-dependent transcription factor MEF2.
Recent Recommendation
Non-coding region variants upstream of MEF2C cause severe developmental disorder through three distinct loss-of-function mechanisms
DD, epilepsy/seizures
ASD or autistic features, stereotypy
Recent Recommendation
Genome-wide analysis of MEF2 transcriptional program reveals synaptic target genes and neuronal activity-dependent polyadenylation site selection.
Recent Recommendation
Experience-Dependent and Differential Regulation of Local and Long-Range Excitatory Neocortical Circuits by Postsynaptic Mef2c.
Recent Recommendation
Transcription factor MEF2C influences neural stem/progenitor cell differentiation and maturation in vivo.

Rare

Variant ID
Variant Type
Allele Change
Residue Change
Inheritance Pattern
Inheritance Association
Family Type
Author, Year
 GEN159R001 
 copy_number_loss 
  
  
 De novo 
  
  
 GEN159R002 
 copy_number_loss 
  
  
 Unknown 
 Not maternal 
  
 GEN159R003 
 missense_variant 
 c.113T>A 
 p.Leu38Gln 
 De novo 
  
  
 GEN159R004 
 frameshift_variant 
 c.70dup 
 p.Arg24LysfsTer11 
 De novo 
  
  
 GEN159R005 
 frameshift_variant 
 c.226_236del 
 p.His76AspfsTer18 
 De novo 
  
  
 GEN159R006 
 missense_variant 
 c.80G>C 
 p.Gly27Ala 
 De novo 
  
  
 GEN159R007 
 copy_number_loss 
  
  
 Unknown 
 Not maternal 
  
 GEN159R008 
 copy_number_loss 
  
  
 De novo 
  
  
 GEN159R009 
 copy_number_loss 
  
  
 De novo 
  
  
 GEN159R010 
 missense_variant 
 c.241A>G 
 p.Asn81Asp 
 De novo 
  
 Simplex 
 GEN159R011 
 frameshift_variant 
 c.397del 
 p.Ala133LeufsTer52 
 De novo 
  
 Simplex 
 GEN159R012 
 missense_variant 
 c.115T>C 
 p.Cys39Arg 
 De novo 
  
  
 GEN159R013 
 stop_lost 
  
 p.Ter464SerextTer? 
 De novo 
  
  
 GEN159R014 
 copy_number_loss 
  
  
 2 de novo, 8 unknown 
  
 Unknown 
 GEN159R015 
 frameshift_variant 
 c.297del 
 p.Asp100IlefsTer26 
 De novo 
  
 Simplex 
 GEN159R016 
 missense_variant 
 c.9A>T 
 p.Arg3Ser 
 De novo 
  
 Simplex 
 GEN159R017 
 intron_variant 
 c.259-213T>G 
  
 De novo 
  
 Simplex 
 GEN159R018a 
 intergenic_variant 
 T>G 
  
 Familial 
 Both parents 
 Multiplex 
 GEN159R019 
 copy_number_loss 
  
  
 Unknown 
  
 Unknown 
 GEN159R020 
 copy_number_loss 
  
  
 Unknown 
  
 Unknown 
 GEN159R021 
 copy_number_loss 
  
  
 Unknown 
  
 Unknown 
 GEN159R022 
 copy_number_loss 
  
  
 Unknown 
  
 Unknown 
 GEN159R023 
 copy_number_loss 
  
  
 Unknown 
  
 Unknown 
 GEN159R024 
 copy_number_loss 
  
  
 Unknown 
  
 Unknown 
 GEN159R025 
 frameshift_variant 
 c.773del 
 p.Pro258HisfsTer11 
 Unknown 
  
 Unknown 
 GEN159R026 
 copy_number_loss 
  
  
 Unknown 
  
 Unknown 
 GEN159R027 
 copy_number_loss 
  
  
 Unknown 
  
 Unknown 
 GEN159R028 
 copy_number_loss 
  
  
 Unknown 
  
 Unknown 
 GEN159R029 
 copy_number_loss 
  
  
 Unknown 
  
 Unknown 
 GEN159R030 
 copy_number_loss 
  
  
 Unknown 
  
 Unknown 
 GEN159R031 
 copy_number_loss 
  
  
 Unknown 
  
 Unknown 
 GEN159R032 
 copy_number_loss 
  
  
 Unknown 
  
 Unknown 
 GEN159R033 
 copy_number_loss 
  
  
 Unknown 
  
 Unknown 
 GEN159R034 
 copy_number_loss 
  
  
 Unknown 
  
 Unknown 
 GEN159R035 
 complex_structural_alteration 
  
  
 De novo 
  
  
 GEN159R036 
 translocation 
  
  
 De novo 
  
  
 GEN159R037 
 translocation 
  
  
 De novo 
  
  
 GEN159R038 
 inversion 
  
  
 De novo 
  
  
 GEN159R039 
 translocation 
  
  
 De novo 
  
  
 GEN159R040 
 complex_structural_alteration 
  
  
 De novo 
  
  
 GEN159R041 
 translocation 
  
  
 Unknown 
  
  
 GEN159R042 
 translocation 
  
  
 De novo 
  
  
 GEN159R043 
 missense_variant 
 c.108C>A 
 p.Ser36Arg 
 De novo 
  
  
 GEN159R044 
 missense_variant 
 c.71G>A 
 p.Arg24Lys 
 De novo 
  
  
 GEN159R045 
 missense_variant 
 c.1276C>G 
 p.Arg426Gly 
 De novo 
  
 Simplex 
 GEN159R046 
 missense_variant 
 c.169T>G 
 p.Tyr57Asp 
 De novo 
  
 Simplex 
 GEN159R047 
 missense_variant 
 c.48C>G 
 p.Asn16Lys 
 De novo 
  
 Simplex 
 GEN159R048 
 stop_gained 
 c.565C>T 
 p.Arg189Ter 
 Unknown 
 Not maternal 
 Simplex 
 GEN159R049 
 stop_gained 
 c.334G>T 
 p.Glu112Ter 
 Unknown 
 Not paternal 
 Simplex 
 GEN159R050 
 splice_site_variant 
 c.403-1G>T 
 p.? 
 De novo 
  
 Simplex 
 GEN159R051 
 stop_gained 
 c.766C>T 
 p.Arg256Ter 
 De novo 
  
 Simplex 
 GEN159R052 
 missense_variant 
 c.43C>T 
 p.Arg15Cys 
 De novo 
  
 Simplex 
 GEN159R053 
 stop_gained 
 c.766C>T 
 p.Arg256Ter 
 De novo 
  
  
 GEN159R054 
 splice_site_variant 
 c.403-1G>T 
 p.? 
 De novo 
  
 Multi-generational 
 GEN159R055 
 insertion 
  
  
 De novo 
  
  
 GEN159R056 
 translocation 
  
  
 De novo 
  
  
 GEN159R057 
 complex_structural_alteration 
  
  
 De novo 
  
  
 GEN159R058 
 missense_variant 
 c.122G>A 
 p.Cys41Tyr 
 De novo 
  
  
 GEN159R059 
 translocation 
  
  
 De novo 
  
  
 GEN159R060 
 inframe_insertion 
 c.120_121insCTGTGA 
 p.Asp40_Cys41insLeuTer 
 Familial 
 Paternal 
  
 GEN159R061 
 missense_variant 
 c.90G>T 
 p.Lys30Asn 
 De novo 
  
  
 GEN159R062 
 missense_variant 
 c.137T>C 
 p.Ile46Thr 
 Unknown 
 Not maternal 
  
 GEN159R063 
 missense_variant 
 c.43C>T 
 p.Arg15Cys 
 De novo 
  
  
 GEN159R064 
 missense_variant 
 c.58A>G 
 p.Thr20Ala 
 De novo 
  
  
 GEN159R065 
 frameshift_variant 
 c.851del 
 p.Thr284ArgfsTer5 
 De novo 
  
  
 GEN159R066 
 missense_variant 
 c.109G>A 
 p.Val37Met 
 Familial 
 Maternal 
  
 GEN159R067 
 stop_gained 
 c.1323C>G 
 p.Tyr441Ter 
 Unknown 
  
  
 GEN159R068 
 stop_gained 
 c.977C>A 
 p.Ser326Ter 
 Unknown 
  
  
 GEN159R069 
 missense_variant 
 c.116G>T 
 p.Cys39Phe 
 Unknown 
  
  
 GEN159R070 
 missense_variant 
 c.44G>C 
 p.Arg15Pro 
 De novo 
  
 Simplex 
 GEN159R071 
 5_prime_UTR_variant 
 c.-66A>T 
  
 De novo 
  
  
 GEN159R072 
 5_prime_UTR_variant 
 c.-103G>A 
  
 De novo 
  
  
 GEN159R073 
 5_prime_UTR_variant 
 c.-8C>T 
  
 De novo 
  
  
 GEN159R074 
 5_prime_UTR_variant 
 c.-8C>T 
  
 De novo 
  
  
 GEN159R075 
 5_prime_UTR_variant 
 c.-8C>T 
  
 De novo 
  
  
 GEN159R076 
 5_prime_UTR_variant 
 c.-26C>T 
  
 De novo 
  
  
 GEN159R077 
 5_prime_UTR_variant 
 c.-26C>T 
  
 De novo 
  
  
 GEN159R078 
 5_prime_UTR_variant 
 c.-26C>T 
  
 De novo 
  
  
 GEN159R079 
 copy_number_loss 
  
  
 De novo 
  
  
 GEN159R080 
 copy_number_loss 
  
  
 De novo 
  
  
 GEN159R081 
 missense_variant 
 c.9A>T 
 p.Arg3Ser 
 Familial 
 Maternal 
  
 GEN159R082 
 frameshift_variant 
 c.78del 
 p.Phe26LeufsTer3 
 De novo 
  
  
 GEN159R083 
 frameshift_variant 
 c.805-866_805-865dup 
  
 De novo 
  
  
 GEN159R084 
 splice_site_variant 
 c.55-2A>G 
  
 De novo 
  
  
 GEN159R085 
 missense_variant 
 c.44G>C 
 p.Arg15Pro 
 De novo 
  
  
 GEN159R086 
 initiator_codon_variant 
 c.1A>G 
 p.Met1? 
 De novo 
  
  
 GEN159R087 
 initiator_codon_variant 
 c.3G>A 
 p.Met1? 
 Familial 
 Maternal 
  
 GEN159R088 
 copy_number_loss 
  
  
 De novo 
  
 Simplex 
 GEN159R089 
 missense_variant 
 c.26C>T 
 p.Thr9Met 
 Unknown 
  
  
 GEN159R090 
 missense_variant 
 c.122G>A 
 p.Cys41Tyr 
 De novo 
  
  
 GEN159R091 
 frameshift_variant 
 c.51_54del 
 p.Gln18Ter 
 De novo 
  
  
 GEN159R092 
 missense_variant 
 c.1162T>A 
 p.Ser388Thr 
 De novo 
  
  
 GEN159R093 
 splice_site_variant 
 c.865-1G>T 
  
 De novo 
  
  
 GEN159R094 
 missense_variant 
 c.44G>A 
 p.Arg15His 
 De novo 
  
  
 GEN159R095 
 missense_variant 
 c.65C>G 
 p.Thr22Arg 
 Unknown 
  
 Simplex 
 GEN159R096 
 missense_variant 
 c.104T>C 
 p.Leu35Pro 
 De novo 
  
 Simplex 
  et al.  
 GEN159R097 
 splice_site_variant 
 c.258+5G>C 
  
 Unknown 
  
  
  et al.  
 GEN159R098 
 missense_variant 
 c.311A>C 
 p.Asp104Ala 
 Unknown 
  
  
  et al.  

Common

Variant ID
Polymorphism
SNP ID
Allele Change
Residue Change
Population Origin
Population Stage
Author, Year
 GEN159C001 
 intergenic_variant 
 rs254782 
 A>G 
  
 40,675 SCZ cases and 64,643 controls (CLOZUK and independent PGC datasets) 
 Discovery 
Chromosome
CNV Locus
CNV Type
# of studies
Animal Model
5
Duplication
 1
 
5
Duplication
 1
 
5
Deletion
 1
 
5
Deletion-Duplication
 21
 
5
Deletion-Duplication
 2
 
5
Deletion
 1
 

Model Summary

Mef2c is required for the early development of the heart, it is not necessary for the formation of the heart after looping morphogenesis. Conditional embryonic deletion of Mef2c in cortical and hippocampal excitatory neurons decreases cortical network activity by increasing inhibitory and decreasing excitatory synaptic transmission. Mef2c mutant mice exhibit deficits in social approach, learnig and memory, and ultrasonic vocalization. Mef2c mutant mice also display stereotypy and hyperactivity.

References

Type
Title
Author, Year
Primary
Generation of conditional Mef2cloxP/loxP mice for temporal- and tissue-specific analyses.
Additional
MEF2C, a transcription factor that facilitates learning and memory by negative regulation of synapse numbers and function.
Additional
MEF2C regulates cortical inhibitory and excitatory synapses and behaviors relevant to neurodevelopmental disorders.
Additional
NitroSynapsin therapy for a mouse MEF2C haploinsufficiency model of human autism.
Additional
MEF2C Hypofunction in Neuronal and Neuroimmune Populations Produces MEF2C Haploinsufficiency Syndrome-like Behaviors in Mice
Additional

M_MEF2C_1_KO_HM

Model Type: Genetic
Model Genotype: Homozygous
Mutation: Cre-loxP mediated deletion of exon 2 of Mef2c gene with Cre recombinase under control of EIIA- promoter which is expressed in all tissues, including germ cells.
Allele Type: Targeted (Conditional)
Strain of Origin: 129S6
Genetic Background: Not Specified
ES Cell Line: W4
Mutant ES Cell Line: Not Specified
Model Source: Not Specified

M_MEF2C_2_CKO_HM

Model Type: Genetic
Model Genotype: Homozygous
Mutation: Conditional deletion of exon 2 of the Mef2c gene using aMyHC-cre (MCH-cre), in cardiac muscle cells
Allele Type: Conditional loss-of-function
Strain of Origin: 129S6
Genetic Background: Not Specified
ES Cell Line: W4
Mutant ES Cell Line: Not Specified
Model Source: Not Specified

M_MEF2C_3_CKO_HM

Model Type: Genetic
Model Genotype: Homozygous
Mutation: Conditional deletion of exon 2 of the Mef2c gene using MLC2V-cre(Myl2-cre), in cardiac ventricular muscle starting E8
Allele Type: Conditional loss-of-function
Strain of Origin: 129S6
Genetic Background: Not Specified
ES Cell Line: W4
Mutant ES Cell Line: Not Specified
Model Source: Not Specified

M_MEF2C_4_KO_HM

Model Type: Genetic
Model Genotype: Homozygous
Mutation: Cre-loxP mediated deletion of exon 2 of Mef2c gene specific to radial glial cells during late embryogenesis with cre under the control of a human GFAP promoter.
Allele Type: Targeted (Conditional)
Strain of Origin: Not Specified
Genetic Background: Not Specified
ES Cell Line: 129SvEv
Mutant ES Cell Line: Not Specified
Model Source: Not Specified

M_MEF2C_5_VP16_TG

Model Type: Genetic
Model Genotype: Homozygous
Mutation: Overexpression of superactive Mef2c-VP16 protein under the control of the rat neuron-specific enolase (NSE) promoter/enhancer.
Allele Type: Targeted (Overexpression)
Strain of Origin: Not Specified
Genetic Background: Not Specified
ES Cell Line: Not Specified
Mutant ES Cell Line: Not Specified
Model Source: Not Specified

M_MEF2C_6_CKO_HM

Model Type: Genetic
Model Genotype: Homozygous
Mutation: Conditional deletion of exon 2 of the Mef2c gene using Emx1-cre, in the neurons and glia of the neocortex, hippocampus and pallium starting E10.5
Allele Type: Conditional loss-of-function
Strain of Origin: SVeV-129/C57BL/6J
Genetic Background: SVeV-129/C57BL/6J
ES Cell Line: 129SvEv
Mutant ES Cell Line: Not Specified
Model Source: Not Specified

M_MEF2C_7_KO_HT

Model Type: Genetic
Model Genotype: Heterozygous
Mutation: Mef2c heterozygous knockout mice were generated by crossing mice with the second Mef2c exon deleted (PMID 9162005) with their WT littermates.
Allele Type: Knockout
Strain of Origin: C57BL/6J
Genetic Background: C57BL/6J
ES Cell Line: Not Specified
Mutant ES Cell Line: Not Specified
Model Source: PMID 9162005

M_MEF2C_10_CKO_HT

Model Type: Genetic LOF
Model Genotype: Heterozygous
Mutation: Mef2c conditional heterozygous (mef2c-chet) mice were generated by crossing mef2c-flox mice with cell typeâ??selective cre-expressing transgenic mice [pcp2-cre]
Allele Type: Conditional knockout
Strain of Origin: 129S7/SvEvBrd-Hprt+
Genetic Background: C57BL/6J
ES Cell Line: Not Specified
Mutant ES Cell Line: Not Specified
Model Source: The Jackson Laboratory

M_MEF2C_11_CKO_HT

Model Type: Genetic LOF
Model Genotype: Heterozygous
Mutation: Mef2c conditional heterozygous (mef2c-chet) mice were generated by crossing mef2c-flox mice with cell typeâ??selective cre-expressing transgenic mice [cx3cr1creer/creer]. recombination was induced at p1-p3 by tamoxifen treatment in cx3cr1creer;eyfp; ai14 mice and tdtomato expression was used as a repotter.
Allele Type: Conditional knockout
Strain of Origin: 129S7/SvEvBrd-Hprt+
Genetic Background: C57BL/6J
ES Cell Line: Not Specified
Mutant ES Cell Line: Not Specified
Model Source: The Jackson Laboratory

M_MEF2C_8_CKO_HT

Model Type: Genetic LOF
Model Genotype: Heterozygous
Mutation: Mef2c conditional heterozygous (mef2c-chet) mice were generated by crossing mef2c-flox mice with cell typeâ??selective cre-expressing transgenic mice [emx1-cre]. construct is identical to model m_mef2c_6_cko_hm but on a different background.
Allele Type: Conditional knockout
Strain of Origin: 129S7/SvEvBrd-Hprt+
Genetic Background: C57BL/6J
ES Cell Line: Not Specified
Mutant ES Cell Line: Not Specified
Model Source: The Jackson Laboratory

M_MEF2C_9_CKO_HT

Model Type: Genetic LOF
Model Genotype: Heterozygous
Mutation: Mef2c conditional heterozygous (mef2c-chet) mice were generated by crossing mef2c-flox mice with cell typeâ??selective cre-expressing transgenic mice [pv-cre]
Allele Type: Conditional knockout
Strain of Origin: 129S7/SvEvBrd-Hprt+
Genetic Background: C57BL/6J
ES Cell Line: Not Specified
Mutant ES Cell Line: Not Specified
Model Source: The Jackson Laboratory

M_MEF2C_12_KI_HT_L35P

Model Type: Genetic
Model Genotype: Heterozygous
Mutation: Mef2c^L35P knock-in mice were generated using CRISPR-Cas9-mediated gene targeting. The targeted allele has a missense mutation c.104T>C in exon 4, that results in amino acid change in the MEF2C product p.Leu35Pro. The heterozygous model has one wildtype allele and one knock-in allele.
Allele Type: ASD mutation
Strain of Origin: C57BL/6
Genetic Background: C57BL/6
ES Cell Line: Not specified
Mutant ES Cell Line:
Model Source: Zi-Long Qiu

M_MEF2C_1_KO_HM

Category
Entity
Quantity
Experimental Paradigm
Age at Testing
Cardiovascular development and function1
Abnormal
Description: Abnormal cardiovascular development - thin myocardium, lack of trabeculation in the ventricle
Exp Paradigm: Histological analysis of sagittal section of embryo
 Histology
 E9.5
Cardiovascular development and function1
Abnormal
Description: Abnormal cardiovascular development - hypoplastic ventricular region and enlarged sinoatrial region
Exp Paradigm: General observations
 General observations
 E9.5
Size/growth1
Decreased
Description: Decreased size - growth retarded
Exp Paradigm: General observations
 General observations
 E9.5
 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_MEF2C_2_CKO_HM

Category
Entity
Quantity
Experimental Paradigm
Age at Testing
Cardiovascular development and function1
 No change
 Electrocardiogram (ecg)
 14-40 weeks
Cardiovascular development and function1
 No change
 Histology
 14-40 weeks
General characteristics1
 No change
 General observations
 Unreported
Size/growth1
 No change
 General observations
 14-40 weeks
Protein expression level evidence1
 No change
 Quantitative pcr (qrt-pcr)
 15-40 weeks
 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_MEF2C_3_CKO_HM

Category
Entity
Quantity
Experimental Paradigm
Age at Testing
Cardiovascular development and function1
 No change
 Electrocardiogram (ecg)
 14-40 weeks
Cardiovascular development and function1
 No change
 Histology
 14-40 weeks
General characteristics1
 No change
 General observations
 Unreported
Size/growth1
 No change
 General observations
 14-40 weeks
Protein expression level evidence1
 No change
 Quantitative pcr (qrt-pcr)
 15-40 weeks
 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_MEF2C_4_KO_HM

Category
Entity
Quantity
Experimental Paradigm
Age at Testing
Motor coordination and balance1
Abnormal
Description: Abnormal muscle coordination demonstrated by impaired balance beam walking
Exp Paradigm: Balance beam walking
 Balance beam test
 4 weeks
Dendritic architecture: spine density1
Increased
Description: Increased density of dendritic spines in dentate gyrus neurons
Exp Paradigm: Analysis of image stacks from z-series of golgi-stained dentate gyrus
 Golgi-cox staining
 3 weeks
Synaptic plasticity1
Decreased
Description: Decreased paired pulse facilitation (ppf)
Exp Paradigm: Whole-cell voltage clamp recordings of dentate gyrus granule neurons in response to second of two paired stimuli
 Whole-cell patch clamp
 P12-p21
Miniature post synaptic current frequency: excitatory1
Abnormal
Description: Increased frequency of mepsc
Exp Paradigm: Whole-cell voltage clamp recordings of dentate gyrus granule neurons
 Whole-cell patch clamp
 P12-p21
Size/growth1
Decreased
Description: Decreased size
Exp Paradigm: General observations
 General observations
 Unreported
Developmental trajectory1
Abnormal
Description: Abnormal hind/forelimb clasping reflex when suspended by tail
Exp Paradigm: General observations
 General observations
 2-3 weeks
Cued or contextual fear conditioning1
Abnormal
Description: Decreased context-dependent fear conditioning, no change in cue-dependent fear conditioning
Exp Paradigm: Contextual- and cue- dependent learning
 Fear conditioning test
 8 weeks
Protein expression level evidence1
Decreased
Description: Decreased mef2c expression in dentate gyrus, amygdala, dorsolateral amygdaloid nuclei
Exp Paradigm: Mef2c protein expression
 In situ hybridization (ish)
 2 months
General characteristics1
 No change
 General observations
 Unreported
Anxiety1
 No change
 Light-dark exploration test
 2 months
Anxiety1
 No change
 Elevated plus maze test
 2 months
Anxiety1
 No change
 Open field test
 2 months
General locomotor activity1
 No change
 Open field test
 Unreported
Brain anatomy1
 No change
 Histology
 2 months
Synaptic morphology1
 No change
 Electron microscopy
 3 weeks
Apoptosis1
 No change
 Tunel assay
 P14-p42
Pain or nociception1
 No change
 Foot shock test
 2 months
 Not Reported: Circadian sleep/wake cycle, Communications, Immune response, Maternal behavior, Physiological parameters, Repetitive behavior, Seizure, Social behavior

M_MEF2C_5_VP16_TG

Category
Entity
Quantity
Experimental Paradigm
Age at Testing
Dendritic architecture: spine density1
Decreased
Description: Decreased density of dendritic spines in dentate gyrus granule neurons
Exp Paradigm: Analysis of image stacks from z-series of golgi-stained dentate gyrus
 Golgi-cox staining
 P21
Miniature post synaptic current frequency: excitatory1
Decreased
Description: Decreased frequency of mepsc in dentate granule neurons
Exp Paradigm: Whole-cell voltage clamp recordings of dentate gyrus granule neurons
 Whole-cell patch clamp
 P12-p21
Synaptic transmission1
Decreased
Description: Decrease in amplitude of both ampa and nmda mediated epsc in dentate granule neurons
Exp Paradigm: Whole-cell voltage clamp recordings of dentate gyrus granule neurons
 Whole-cell patch clamp
 P12-p21
General characteristics1
 No change
 General observations
 Unreported
Size/growth1
 No change
 General observations
 Unreported
Cued or contextual fear conditioning1
 No change
 Fear conditioning test
 2 months
General locomotor activity1
 No change
 Open field test
 Unreported
Motor coordination and balance1
 No change
 Accelerating rotarod test
 4 weeks
Synaptic plasticity1
 No change
 Whole-cell patch clamp
 P12-p21
 Not Reported: Circadian sleep/wake cycle, Communications, Emotion, Immune response, Maternal behavior, Molecular profile, Physiological parameters, Repetitive behavior, Seizure, Sensory, Social behavior

M_MEF2C_6_CKO_HM

Category
Entity
Quantity
Experimental Paradigm
Age at Testing
Hyperactivity1
Increased
Description: Mef2c cko mice exhibit hyperactivity in a novel environment compared to wildtype controls
Exp Paradigm: Open field test
 Open field test
 812 week
Cortical thickness1
Decreased
Description: Mef2c cko mice exhibit a decrease (~10%) in neocortical thickness compared to controls
Exp Paradigm: Histology
 Histology
 NA
Synapse density: inhibitory1
Increased
Description: Increased dendritic gabaergic synapse density (2 folds) on mef2c cko cortical pyramidal neurons relative to wildtype controls
Exp Paradigm: Immunostaining: gad65 and gabrg2
 Immunostaining
 NA
Dendritic architecture: spine density1
Decreased
Description: Decreased dendritic spin density on mef2c cko cortical pyramidal neurons relative to wildtype controls
Exp Paradigm: Histology
 Histology
 NA
Miniature post synaptic current amplitude: inhibitory1
Increased
Description: Increased mipsc amplitude measured in mef2c cko mice relative to wild-type controls
Exp Paradigm: Whole-cell patch clamp: somatosensory cortical layer 2/3 pyramidal neurons
 Whole-cell patch clamp
 3 weeks
Miniature post synaptic current amplitude: excitatory1
Decreased
Description: Decreased mepsc amplitude in mef2c cko mice relative to wild-type controls
Exp Paradigm: Whole-cell patch clamp: somatosensory cortical layer 2/3 pyramidal neurons
 Whole-cell patch clamp
 3 weeks
Network excitability1
Decreased
Description: Decreased frequency (90%), decreased duration (50%), and decreased amplitude (50%) of spontaneous up states in acute slices from the somatosensory cortex of mef2c cko mice
Exp Paradigm: Field potential recordings
 Field potential recordings
 3 weeks
Miniature post synaptic current frequency: inhibitory1
Increased
Description: Increased mipsc frequency measured in mef2c cko mice relative to wild-type controls
Exp Paradigm: Whole-cell patch clamp: somatosensory cortical layer 2/3 pyramidal neurons
 Whole-cell patch clamp
 3 weeks
Vertical jumping or back flipping1
Increased
Description: Increased repetitive jumping behavior (~3-fold) in mef2c cko mice
Exp Paradigm: Observation of repetitive behavior
 Observation of repetitive behavior
 12 week
Stereotypy1
Increased
Description: Increased repetitive fine motor movements in mef2c cko mice
Exp Paradigm: Observation of repetitive behavior
 Observation of repetitive behavior
 12 week
Social approach1
Decreased
Description: Mef2c cko mice spent less time interacting with the social animal than the control mice
Exp Paradigm: Three-chamber social approach test
 Three-chamber social approach test
 12 week
Ultrasonic vocalization: isolation induced1
Decreased
Description: Decreased number of isolation-induced ultrasonic vocalizations (usvs) in mef2c cko pups relative to wildtype controls
Exp Paradigm: Monitoring ultrasonic vocalizations
 Monitoring ultrasonic vocalizations
 P4-p10
Ultrasonic vocalization: interaction induced: opposite sex stimulus1
Decreased
Description: Decreased number of opposite-sex-induced ultrasonic vocalizations (usvs) in mef2c cko male mice, featured as a 3-fold increase in the latency to the first call, 5-fold increase in unstructured usvs, a decrease in complex, but no change in simple, usvs, and a decrease in the maximum usv frequency and the mean frequency at the end of calls
Exp Paradigm: Monitoring ultrasonic vocalizations
 Monitoring ultrasonic vocalizations
 Adult
Reward reinforced choice behavior1
Decreased
Description: Mef2c cko mice exhibit reduced appetitive reward-related behavior
Exp Paradigm: Sucrose preference test
 Sucrose preference test
 12 week
Cued or contextual fear conditioning: memory of cue1
Decreased
Description: Mef2c cko mice have decreased learning and memory in cued fear conditioning tests relative to wildtype controls
Exp Paradigm: Fear conditioning test: a training protocol is used to pair three white noise tones (cs) with a foot shock (us) presented 1 min after the end of the cs; the memory is tested 24 hr after training sessions
 Fear conditioning test
 12 week
Cued or contextual fear conditioning: memory of context1
Decreased
Description: Mef2c cko mice have decreased learning and memory in contextual fear conditioning tests relative to wildtype controls
Exp Paradigm: Fear conditioning test: a training protocol is used to pair three white noise tones (cs) with a foot shock (us) presented 1 min after the end of the cs; the memory is tested 24 hr after training sessions
 Fear conditioning test
 12 week
Gene expression1
Increased
Description: Increased expression of gabra5 and nos1 (gabaergic transmission regulatory genes) and pcdh10 (mef2/fmrp-dependent glutamatergic synapse elimination) in mef2c cko somatosensory cortex
Exp Paradigm: Rna sequencing; quantitative pcr (qrt-pcr): somatosensory cortex-rna sequencing
 Rna sequencing
 P21; adult
Gene expression1
Increased
Description: Increased expression of gabra5 and nos1 (gabaergic transmission regulatory genes) and pcdh10 (mef2/fmrp-dependent glutamatergic synapse elimination) in mef2c cko somatosensory cortex
Exp Paradigm: Rna sequencing; quantitative pcr (qrt-pcr): somatosensory cortex- quantitative pcr (qrt-pcr)
 Quantitative pcr (qrt-pcr)
 P21; adult
Gene expression1
Decreased
Description: Decreased expression of some autism-linked genes (ntng1, nlgn1, nrxn1, nrxn3, pcdh19, shank2, shank3, pten and htr1b) in mef2c cko somatosensory cortex
Exp Paradigm: Rna sequencing; quantitative pcr (qrt-pcr): somatosensory cortex-rna sequencing
 Rna sequencing
 P21; adult
Targeted expression1
Decreased
Description: Decreased expression level of mef2c protein throughout the cortex and hippocampus of mef2c cko mice
Exp Paradigm: Western blot: mef2c
 Western blot
 812 week
Gene expression1
Decreased
Description: Decreased expression of some autism-linked genes (ntng1, nlgn1, nrxn1, nrxn3, pcdh19, shank2, shank3, pten and htr1b) in mef2c cko somatosensory cortex
Exp Paradigm: Rna sequencing; quantitative pcr (qrt-pcr): somatosensory cortex- quantitative pcr (qrt-pcr)
 Quantitative pcr (qrt-pcr)
 P21; adult
Developmental trajectory1
 No change
 General observations
 NA
Mortality/lethality1
 No change
 General observations
 NA
Size/growth1
 No change
 Body weight measurement
 3 months
Motor coordination and balance1
 No change
 Accelerating rotarod test
 812 week
Brain morphology1
 No change
 Histology
 NA
Cortical lamination1
 No change
 Histology
 NA
Dendritic architecture: dendritic tree complexity1
 No change
 Histology
 NA
Miniature post synaptic current frequency: excitatory1
 No change
 Whole-cell patch clamp
 3 weeks
Neuronal activity dependent gene regulation1
 No change
 Luciferase reporter assay
 NA
Repetitive digging1
 No change
 Observation of repetitive behavior
 12 week
Self grooming: perseveration1
 No change
 Observation of repetitive behavior
 12 week
Foot shock sensitivity1
 No change
 Foot shock test
 12 week
Olfaction1
 No change
 Olfactory discrimination test
 12 week
Startle response: acoustic stimulus1
 No change
 Startle response test
 12 week
Taste1
 No change
 Sucrose preference test
 12 week
Nest building behavior1
 No change
 Nest building assay
 12 week
Social dominance1
 No change
 Tube test of social dominance
 12 week
Social scent marking or recognition1
 No change
 Olfactory discrimination test
 12 week
 Not Reported: Circadian sleep/wake cycle, Emotion, Immune response, Maternal behavior, Physiological parameters, Seizure

M_MEF2C_7_KO_HT

Category
Entity
Quantity
Experimental Paradigm
Age at Testing
General locomotor activity: Ambulatory activity2
Increased
Description: Increase in total distance travelled in a novel environment
 Novel cage test
 Adult
Clasping reflex1
Increased
Description: Mutants show increased paw clasping compared to controls.
Exp Paradigm: NA
 Tail suspension test
 3 months
Clasping reflex1
Abnormal
Description: Mutant mice show abnormal paw clasping reflex compared to controls.
Exp Paradigm: NA
 General observations
 >3 months
Neuronal number1
Decreased
Description: Mutants show decreased neuronal numbers in the hippocampus and frontal cortex compared to controls.
Exp Paradigm: NA
 Immunohistochemistry
 3 months
Synapse density: excitatory1
Increased
Description: Mutants show increased vglut2 immunoreactivity in the ca1 region of the hippocampus compared to controls.
Exp Paradigm: NA
 Immunohistochemistry
 3 months
Dendritic architecture: dendritic tree complexity1
Decreased
Description: Mutants show decreased complexity of the dendritic tree compared to controls. mutants show decreased dendritic interactions and decreased numbers of secondary, tertiary, quaternary and quinary dendritic branches compared to controls.
Exp Paradigm: Sholl analysis was performed.
 Golgi-cox staining
 3 months
Neuronal size1
Decreased
Description: Mutants show decreased neuronal cell size compared to controls.
Exp Paradigm: Visualized via retroviral-mediated gene transduction of mcherry.
 Immunofluorescence staining
 3 months
Dendritic architecture: dendritic length1
Decreased
Description: Mutants show decreased dendritic length compared to controls.
Exp Paradigm: Sholl analysis was performed.
 Golgi-cox staining
 3 months
Synaptic morphology1
Decreased
Description: Mutants show decreased synaptophysin at synapses in the hippocampus compared to controls.
Exp Paradigm: NA
 Immunohistochemistry
 3 months
Neuronal number: interneurons1
Decreased
Description: Mutants show decrease in parvalbumin positive basket interneurons and synapses compared to controls.
Exp Paradigm: Pv
 Immunohistochemistry
 3 months
Cell proliferation: neural precursors1
Decreased
Description: Mutants show decrease in the number of proliferating cells (pcna), developing neurons (dcx) and brdu labelled neun cells compared to controls.
Exp Paradigm: 8-week-old mice were injected intraperitoneally twice daily for 5 consecutive days with brdu.
 Golgi-cox staining
 Adult
Synapse density: inhibitory1
Decreased
Description: Mutants show decrease in the expression of vgat in the hippocampus, a marker of inhibitory synapses, compared to controls.
Exp Paradigm: NA
 Immunohistochemistry
 3 months
Miniature post synaptic current frequency: excitatory1
Increased
Description: Mutants show an increase in mepsc frequency and decreased inter-event interval compared to controls.
Exp Paradigm: Acsf was used as the external bath solution, with 50 m picrotoxin and 1 m tetrodotoxin (ttx) to isolate spontaneous mepscs.
 Whole-cell voltage clamp
 1-6 months
Apoptosis: brain cells1
Increased
Description: Mutants show increase in the number of neurons staining for active caspase-3 and for terminal deoxynucleotidyl transferase dutp nick end labeling (tunel) in the ca3 region of the hippocampus, compared to controls.
Exp Paradigm: Immunohistochemistry: caspase-3
 Immunohistochemistry
 3 months
Presynaptic function: paired-pulse facilitation1
Decreased
Description: Mutants show decreased paired-pulse facilitation compared to controls.
Exp Paradigm: Ppf was tested by applying two pulses with interstimulus intervals ranging from 20 to 200 ms.
 Whole-cell voltage clamp
 1-6 months
Miniature post synaptic current frequency: excitatory2
Increased
Description: Increase in layers 2/3 mepsc frequency; no change in layer5 mepsc frequency
 Whole-cell voltage clamp
 P35-40
Synaptic transmission: excitatory1
Increased
Description: Mutants show increased e/i frequency and e/i amplitude ratios compared to controls, indicating a functional e/i imbalance.
Exp Paradigm: Acsf was used as the external bath solution, with 50 m picrotoxin and 1 m tetrodotoxin (ttx) to isolate spontaneous mepscs.
 Whole-cell voltage clamp
 1-6 months
Apoptosis: brain cells1
Increased
Description: Mutants show increase in the number of neurons staining for active caspase-3 and for terminal deoxynucleotidyl transferase dutp nick end labeling (tunel) in the ca3 region of the hippocampus, compared to controls.
Exp Paradigm: Detection of apoptosis using the tunel assay
 Tunel assay
 3 months
Presynaptic function: paired-pulse facilitation2
Increased
Description: Increase in paired pulse facilitation ratio of local horizontal inputs indicating a decrease in presynaptic release probability when stimulated in layer 2/3 but not layer 4
 Whole-cell voltage clamp
 P35-40
Miniature post synaptic current amplitude: excitatory1
Decreased
Description: Mutants show a slight reduction in mepsc amplitudes compared to controls.
Exp Paradigm: Acsf was used as the external bath solution, with 50 m picrotoxin and 1 m tetrodotoxin (ttx) to isolate spontaneous mepscs.
 Whole-cell voltage clamp
 1-6 months
Miniature post synaptic current amplitude: inhibitory1
Decreased
Description: Mutants show decreased mipsc amplitude compared to controls.
Exp Paradigm: Acsf was used as the external bath solution, with 10 m 6-cyano-7-nitroquinoxaline-2,3-dione (cnqx), 50 m (2r)-amino-5- phosphonopentanoate(ap5), and 1 m ttx to isolate spontaneous mipscs.
 Whole-cell voltage clamp
 1-6 months
Synaptic transmission: excitatory2
Abnormal
Description: Deficits in cortical excitatory synaptic transmission as shown by an increase in mepsc frequency and a decrease in mepsc amplitude
 
 Adult
Miniature post synaptic current frequency: inhibitory1
Decreased
Description: Mutants show decreased mipsc frequency and increased inter-event interval compared to controls.
Exp Paradigm: Acsf was used as the external bath solution, with 10 m 6-cyano-7-nitroquinoxaline-2,3-dione (cnqx), 50 m (2r)-amino-5- phosphonopentanoate(ap5), and 1 m ttx to isolate spontaneous mipscs.
 Whole-cell voltage clamp
 1-6 months
Miniature post synaptic current amplitude: excitatory2
Decreased
Description: Decreased input-selective glutamatergic synaptic transmission indicated by reduction in the amplitude of evoked excitatory postsynaptic currents, suggesting a reduction in presynaptic and/or postsynaptic transmission when stimulated in layer 2/3 and layer 5 but not layer 4; decreased ampa mediated mepsc amplitude indicating decreased ampa-mediated postsynaptic strength
 Whole-cell voltage clamp
 P35-40
Synaptic plasticity: hippocampal ltp1
Decreased
Description: Mutants show decreased ltp in the hippocampal ca1 region compared to controls.
Exp Paradigm: Ltp was induced by applying high-frequency stimulation consisting of three 100 hz pulses (duration: 1 s, interval: 20 s).
 Whole-cell voltage clamp
 1-6 months
Head bobbing1
Increased
Description: Mutant mice show increased head dipping compared to controls.
Exp Paradigm: NA
 Hole-board test
 >3 months
Vertical jumping or back flipping2
Increased
Description: Increased number of rearings
 Novel cage test
 Adult
Head bobbing1
Increased
Description: Mutants show increased head dipping compared to controls.
Exp Paradigm: NA
 Hole-board test
 3 months
Pain or nociception2
Decreased
Description: Decrease in response to foot shock
 Foot shock test
 Adult
Social approach1
Decreased
Description: Mutants spend less time interacting with an unfamiliar mouse compared to controls.
Exp Paradigm: Durations and number of visits to the chamber with the unfamiliar mouse were recorded.
 Three-chamber social approach test
 3 months
Social approach2
Decreased
Description: Decreased preference for a novel same-sex mouse over a novel object
 Three-chamber social approach test
 Adult
Ultrasonic vocalization: Isolation induced2
Decreased
Description: Fewer usv calls when separated
 Monitoring ultrasonic vocalizations
 P7-10
Ultrasonic vocalization: Interaction induced: opposite sex stimulus2
Decreased
Description: Fewer usv calls in the presence of estrus female
 Monitoring ultrasonic vocalizations
 Adult
Size/growth1
Decreased
Description: Mutants show decreased body weight compared to controls.
Exp Paradigm: NA
 Body weight measurement
 3 months
Mortality/lethality: embryonic: incomplete penetrance2
Increased
Description: Non-mendelian frequency of het mice suggesting partial embryonic lethality
 Genotypic ratio of progeny from heterozygous parents
 Adult
Mortality/lethality1
Increased
Description: Mutants show decreased lifespan compared to controls.
Exp Paradigm: NA
 Survival analysis
 3 weeks, 3 months
Anxiety1
Decreased
Description: Mutants spend more time in the center of the open field compared to controls.
Exp Paradigm: NA
 Open field test
 3 months
Anxiety2
Decreased
Description: Increase in time spent in the open arms
 Elevated plus maze test
 Adult
Innate immune response2
Increased
Description: Increased expression of complement-related genes linked previously to synaptic pruning and/or asd risk, including c1qb, c1qc, and c4b
 Quantitative PCR (qRT-PCR)
 P35-40
Astrogliosis1
Increased
Description: Mutants show increased gfap positive cells compared to controls, indicating astrogliosis.
Exp Paradigm: NA
 Immunohistochemistry
 3 months
Spatial reference memory1
Decreased
Description: Mutants showed no preference for the target quadrant over the opposite quadrant compared to controls, during the probe test trials.
Exp Paradigm: NA
 Morris water maze test
 3 months
Spatial reference memory1
Decreased
Description: Mutant mice showed no preference for the target over the opposite quadrant compared to controls, in probe tests.
Exp Paradigm: NA
 Barnes maze test
 >3 months
Spatial working memory1
Decreased
Description: Mutants take longer time to find the hidden platform than controls.
Exp Paradigm: NA
 Morris water maze test
 3 months
Object recognition memory2
Increased
Description: Increased preference for the novel object
 Novel object recognition test
 Adult
Spatial working memory1
Decreased
Description: Mutant mice took longer to find the escape tunnel during training sessions compared to controls.
Exp Paradigm: NA
 Barnes maze test
 >3 months
Gene expression: activated microglia2
Abnormal
Description: Dysregulation of microglial genes
 RNA sequencing
 P35-40
Targeted expression1
Decreased
Description: Mutants show decreased expression of mef2c protein compared to controls. mutants show decreased expression of mef2c transcript compared to controls.
Exp Paradigm: Western blot
 Western blot
 3 months
Differential gene expression2
Abnormal
Description: Dysregulation of 490 cortical genes; gse139419; single cell rna seq shows enrichment for cortical excitatory neuron genes and microglial genes
 RNA sequencing
 P35-40
Protein expression level evidence1
Increased
Description: Mutants show increase in protein levels of vglut2 compared to controls.
Exp Paradigm: Immunoblots were performed on synaptosome-enriched hippocampal lysates.
 Western blot
 3 months
Gene expression1
Decreased
Description: Mutants show decreased transcript levels for vesicular -aminobutyric acid (gaba) transporter vgat (encoded by slc32a1) and ccl9 compared to controls.
Exp Paradigm: NA
 Quantitative pcr (qrt-pcr)
 3 months
Targeted expression1
Decreased
Description: Mutants show decreased expression of mef2c protein compared to controls. mutants show decreased expression of mef2c transcript compared to controls.
Exp Paradigm: Quantitative pcr (qrt-pcr)
 Quantitative pcr (qrt-pcr)
 3 months
Differential gene expression2
Abnormal
Description: Degs were associated with asd risk, fmrp binding, microglia and cellular function
 Quantitative PCR (qRT-PCR)
 P35-40
Protein expression level evidence1
Decreased
Description: Mutants show decrease in the levels of the synaptosomal proteins synaptophysin and gad65 compared to controls.
Exp Paradigm: Immunoblots were performed on synaptosome-enriched hippocampal lysates. gad65 is a marker of inhibitory synapses.
 Western blot
 3 months
Gene expression1
Abnormal
Description: Mutants mice show abnormal gene expression in biogroups of neurogenesis, neuronal differentiation, synaptic function (downregulated), and neuronal cell death (upregulated), compared to controls.
Exp Paradigm: NA
 Gene expression microarray
 >3 months
Targeted expression2
Decreased
Description: Decreased wildtype mef2c protein, increased mutant mef2c protein with ex2 deleted
 Western blot
 Adult
Gene expression: activated microglia2
Increased
Description: Increase in iba1 expression in cx and hip, suggesting microglial activation
 Immunohistochemistry
 P35-40
Gene expression1
Increased
Description: Mutant mice show increased expression of vglut2 compared to controls.
Exp Paradigm: NA
 Quantitative pcr (qrt-pcr)
 3 months
Protein-DNA complex assembly2
Decreased
Description: Mutant mef2c lacks dna binding ability and does not interfere with dna binding ability of wt mef2c
 Chromatin immunoprecipitation (ChIP)
 Adult
Mortality/lethality1
 No change
 Survival analysis
 E18
Depression2
 No change
 Sucrose preference test
 Adult
CNS inflammation2
 No change
 Quantitative PCR (qRT-PCR)
 P35-40
Cytokine levels2
 No change
 Gene expression microarray
 P35-40
Cognitive flexibility2
 No change
 Operant self-learning paradigm
 Adult
Cognitive flexibility2
 No change
 Y-maze test
 Adult
Cued or contextual fear conditioning: Memory of context2
 No change
 Fear conditioning test
 Adult
Cued or contextual fear conditioning: Memory of cue2
 No change
 Fear conditioning test
 Adult
Discrimination learning2
 No change
 Operant self-learning paradigm
 Adult
Extinction learning2
 No change
 Operant self-learning paradigm
 Adult
Reward reinforced choice behavior: long-term memory2
 No change
 Operant self-learning paradigm
 Adult
Reward reinforced choice behavior: short-term memory2
 No change
 Operant self-learning paradigm
 Adult
Spatial working memory2
 No change
 Barnes maze test
 Adult
Gene expression1
 No change
 Quantitative pcr (qrt-pcr)
 3 months
Protein expression level evidence1
 No change
 Western blot
 3 months
General locomotor activity: ambulatory activity1
 No change
 Open field test
 3 months
Grip strength1
 No change
 Grip strength test
 3 months
Motor coordination and balance1
 No change
 Balance beam test
 3 months
Motor coordination and balance1
 No change
 Traction test
 3 months
Motor coordination and balance1
 No change
 Vertical pole test
 3 months
Motor coordination and balance2
 No change
 Accelerating rotarod test
 Adult
Swimming ability1
 No change
 Morris water maze test
 3 months
Cortical thickness2
 No change
 Histology
 Adult
Dendritic architecture: spine density2
 No change
 Whole-cell voltage clamp
 P35-40
Dendritic architecture: spine morphology2
 No change
 Whole-cell voltage clamp
 P35-40
Microglial morphology2
 No change
 Immunohistochemistry
 P35-40
Microglial number2
 No change
 Immunohistochemistry
 P35-40
Somatosensory cortical map architecture2
 No change
 Immunohistochemistry
 P35-40
Synapse density: excitatory1
 No change
 Immunohistochemistry
 3 months
Synaptic morphology1
 No change
 Immunohistochemistry
 3 months
Action potential property: firing rate2
 No change
 Whole-cell voltage clamp
 P35-40
Miniature post synaptic current amplitude: inhibitory2
 No change
 Whole-cell voltage clamp
 P35-40
Miniature post synaptic current frequency: inhibitory2
 No change
 Whole-cell voltage clamp
 P35-40
Startle response: acoustic stimulus2
 No change
 Acoustic startle reflex test
 Adult
 Not Reported: Circadian sleep/wake cycle, Communications, Maternal behavior, Physiological parameters, Seizure, Sensory

M_MEF2C_10_CKO_HT

Category
Entity
Quantity
Experimental Paradigm
Age at Testing
Anxiety1
 No change
 Elevated plus maze test
 Adult
General locomotor activity: Ambulatory activity1
 No change
 Novel cage test
 Adult
Vertical jumping or back flipping1
 No change
 Novel cage test
 Adult
Pain or nociception1
 No change
 Foot shock test
 Adult
Social approach1
 No change
 Three-chamber social approach test
 Adult
 Not Reported:

M_MEF2C_11_CKO_HT

Category
Entity
Quantity
Experimental Paradigm
Age at Testing
Miniature post synaptic current amplitude: excitatory1
Decreased
Description: Decreased evoked mepsc amplitude in layer 2/3 pyramidal neurons
 Whole-cell voltage clamp
 Adult
Vertical jumping or back flipping1
Increased
Description: Increased jump counts
 Novel cage test
 Adult
Social approach1
Decreased
Description: Decreased preference for a novel same-sex mouse over a novel object
 Three-chamber social approach test
 Adult
Protein expression: In situ protein expression1
Increased
Description: Reporter tdtomato is expressed in iba1 positive cells in hip and sc
 Immunohistochemistry
 Adult
Anxiety1
 No change
 Elevated plus maze test
 Adult
General locomotor activity: Ambulatory activity1
 No change
 Novel cage test
 Adult
Miniature post synaptic current amplitude: excitatory1
 No change
 Whole-cell voltage clamp
 Adult
Presynaptic function: paired-pulse facilitation1
 No change
 Whole-cell voltage clamp
 Adult
Vertical jumping or back flipping1
 No change
 Novel cage test
 Adult
Pain or nociception1
 No change
 Foot shock test
 Adult
 Not Reported:

M_MEF2C_8_CKO_HT

Category
Entity
Quantity
Experimental Paradigm
Age at Testing
General locomotor activity: Ambulatory activity1
Increased
Description: Increase in distance travelled
 Novel cage test
 Adult
Miniature post synaptic current amplitude: excitatory1
Decreased
Description: Decreased mepsc amplitude in layer 2/3 pyramidal neurons
 Whole-cell voltage clamp
 Adult
Synaptic transmission: excitatory1
Decreased
Description: Reduced mepsc frequency and amplitude and decreased evoked mepsc amplitude in layer 2/3 pyramidal neurons indicate a decrease in excitatory synaptic transmission
 
 Adult
Miniature post synaptic current amplitude: excitatory1
Decreased
Description: Decreased evoked mepsc amplitude in layer 2/3 pyramidal neurons
 Whole-cell voltage clamp
 Adult
Miniature post synaptic current frequency: excitatory1
Decreased
Description: Reduced mepsc frequency
 Whole-cell voltage clamp
 Adult
Miniature post synaptic current frequency: excitatory1
Decreased
Description: Decreased mepsc frequency in layer 2/3 pyramidal neurons
 Whole-cell voltage clamp
 Adult
Vertical jumping or back flipping1
Increased
Description: Increased jump counts
 Novel cage test
 Adult
Anxiety1
Decreased
Description: Increase in time spent in the open arms
 Elevated plus maze test
 Adult
Differential gene expression1
Abnormal
Description: Dysregulation of cortical genes; enrichment for microglia genes was not detected on degs, gse139419
 RNA sequencing
 P35-40
General locomotor activity: Ambulatory activity1
 No change
 Novel cage test
 Adult
Vertical jumping or back flipping1
 No change
 Novel cage test
 Adult
Pain or nociception1
 No change
 Foot shock test
 Adult
Social approach1
 No change
 Three-chamber social approach test
 Adult
 Not Reported:

M_MEF2C_9_CKO_HT

Category
Entity
Quantity
Experimental Paradigm
Age at Testing
Anxiety1
 No change
 Elevated plus maze test
 Adult
General locomotor activity: Ambulatory activity1
 No change
 Novel cage test
 Adult
Vertical jumping or back flipping1
 No change
 Novel cage test
 Adult
Pain or nociception1
 No change
 Foot shock test
 Adult
Social approach1
 No change
 Three-chamber social approach test
 Adult
 Not Reported:

M_MEF2C_12_KI_HT_L35P

Category
Entity
Quantity
Experimental Paradigm
Age at Testing
General locomotor activity: ambulatory activity1
Increased
Description: Mef2c heterozygous knockin mice show an increase total distance traveled and mean velocity in the open field test.
 Open field test
 8 weeks
Neuronal number: interneurons1
Decreased
Description: Mef2c heterozygous knockin mice show a decrease in parvalbumin-positive neuronal density in retrosplenial cortex, visual cortex, somatosensory cortex and dentate gyrus, but no change in somatostatin-positive interneuron density in retrosplenial cortex, hippocampus, somatosensory cortex.
Exp Paradigm: parvalbumin, somatostatin
 Immunohistochemistry
 10-12 weeks
Dendritic architecture: spine density1
Decreased
Description: Mef2c heterozygous knockin mice show a decrease in spine density and mushroom-type spine density.
 Fluorescence microscopy
 10-12 weeks
Miniature post synaptic current frequency: excitatory1
Decreased
Description: Mef2c heterozygous knockin mice show a decrease in mEPSC frequency in the medial prefrontal cortex.
Exp Paradigm: mPFC
 Whole-cell patch clamp
 10-12 weeks
Self grooming1
Increased
Description: Mef2c heterozygous knockin male mice show increased time grooming and scratching.
 Grooming behavior assessments
 8 weeks
Social memory1
Decreased
Description: Mef2c heterozygous knockin mice show no preference for novel conspecific over a familiar one, indicating a decrease in social memory.
 Three-chamber social approach test
 8 weeks
Social memory1
Decreased
Description: Mef2c heterozygous knockin mice show decreased social recognition in the resident intruder test. Test consisted in presenting an intruder mice in 4 consecutive trials, and a new intruder in a fifth trial. Mef2c heterozygous mice did not show an increase in sniffing behavior in the fifth trial with the novel intruder.
 Resident-intruder test
 8 weeks
Social interaction1
Decreased
Description: Mef2c heterozygous knockin mice show decreased social interaction in the resident intruder test, measured by cumulative sniffing time. Test consisted in presenting an intruder mice in 4 consecutive trials, and a new intruder in a fifth trial.
 Resident-intruder test
 8 weeks
Anxiety1
Decreased
Description: Mef2c heterozygous knockin mice show increased time in open arms and decreased time in closed arms, indicating a decrease in anxiety-like behavior.
 Elevated plus maze test
 8 weeks
Object recognition memory1
Decreased
Description: Mef2c heterozygous knockin female mice do not show a preference for a new object over a familiar one.
 Novel object recognition test
 8 weeks
Targeted expression1
Decreased
Description: Mef2c heterozygous knockin mice show markedly decreased expression of MEF2C protein in the prefrontal cortex and hippocampus.
 Western blot
 10-12 weeks
Targeted expression1
Decreased
Description: Mef2c heterozygous knockin mice show a decrease in MEF2C expression in somatosensory cortex, dentate gyrus and lateral amygdala.
Exp Paradigm: Mef2c
 Immunohistochemistry
 10-12 weeks
Size/growth1
 No change
 Body weight measurement
 P0-9 weeks
Anxiety1
 No change
 Open field test
 8 weeks
Anxiety1
 No change
 Light-dark exploration test
 8 weeks
Object recognition memory1
 No change
 Novel object recognition test
 8 weeks
Spatial learning1
 No change
 Barnes maze test
 8 weeks
Spatial reference memory1
 No change
 Barnes maze test
 8 weeks
Miniature post synaptic current amplitude: excitatory1
 No change
 Whole-cell patch clamp
 10-12 weeks
Miniature post synaptic current amplitude: inhibitory1
 No change
 Whole-cell patch clamp
 10-12 weeks
Miniature post synaptic current frequency: inhibitory1
 No change
 Whole-cell patch clamp
 10-12 weeks
Social approach1
 No change
 Three-chamber social approach test
 8 weeks
 Not Reported:


Interactor Symbol Interactor Name Interactor Organism Entrez ID Uniprot ID Interaction Type Evidence Reference
CDKL5 cyclin-dependent kinase-like 5 6792 O76039 Luciferase reporter assay
Zweier M , et al. 2010
HDAC4 histone deacetylase 4 9759 P56524 IP; Luciferase reporter assay
Wang AH , et al. 1999
HDAC9 histone deacetylase 9 9734 Q9UKV0 M2H
Ravasi T , et al. 2010
MBD5 methyl-CpG binding domain protein 5 55777 Q9P267 Y2H
Bandyopadhyay S , et al. 2010
MECP2 methyl CpG binding protein 2 (Rett syndrome) 4204 P51608 Reporter assay
Zweier M , et al. 2010
MEF2B Myocyte-specific enhancer factor 2B 100271849 Q02080 IP; LC-MS/MS
Huttlin EL , et al. 2015
PLA2G12A phospholipase A2, group XIIA 81579 Q542Y6 Y2H
Wang J , et al. 2011
SIRT5 NAD-dependent protein deacylase sirtuin-5, mitochondrial 23408 Q9NXA8 GST; In vivo acetylation assay; in vitro kinase assay; AMPK kinase assay; IP/WB
Lin YY , et al. 2012
SKP2 S-phase kinase-associated protein 2, E3 ubiquitin protein ligase 6502 B4DJT4 IP/WB
Di Giorgio E , et al. 2015
UBC ubiquitin C 7316 P63279 MS
Wagner SA , et al. 2011
Ash2l ash2 (absent, small, or homeotic)-like (Drosophila) 23808 Q91X20 IP/WB
Jung Y , et al. 2016
RBFOX1 RNA binding protein, fox-1 homolog (C. elegans) 1 268859 Q9JJ43 Transfection analyses
Hakim NH , et al. 2010
Smarca4 SWI/SNF related, matrix associated, actin dependent regulator of chromatin, subfamily a, member 4 20586 Q3TKT4 IP/WB
Zhang Z , et al. 2015
MIB1 mindbomb E3 ubiquitin protein ligase 1 307594 D3ZUV2 Affinity chromatography; LC-MS/MS
Mertz J , et al. 2015

HELP
Copyright © 2017 MindSpec, Inc.