HELP     Sign In
Search

Relevance to Autism

A de novo LoF variant and two de novo missense variants that were predicted to be damaging were observed in ASD probands (PMID 22495306, 22495309, 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) identified TRIO 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 (PMID 25363760). De novo likely damaging missense variants in this gene have also been identified in patients presenting with intellectual disability (PMID 23033978) and epilepsy (PMID 23934111). An additional de novo likely damaging missense variant in TRIO was identified in an ASD proband from the Autism Genetic Resource Exchange (AGRE) in Stessman et al., 2017. Sadybekov et al., 2017 identified an enrichment of ASD- and NDD-associated missense and loss-of-function (LoF) variants in the GEF1/DH1 subdomain of TRIO; no missense or LoF variants in this domain were observed in 9,937 control exomes. Additional functional analysis of one frameshift variant and three missense variants residing in the Trio GEF1/DH1 subdomain in this report demonstrated effects on Rac1 activation and synaptic function; one of the missense variants that exhibited reduced Rac1 activation and impaired synaptic function, suggestive of a loss-of-function effect, had previously been observed de novo in an ASD proband from the Simons Simplex Collection in Sanders et al., 2012. Barbosa et al., 2020 reported 24 individuals with confirmed pathogenic missense or nonsense variants in the TRIO gene and noted that stereotypy (27%) and autistic behavior (31%) were among the recurrent behavioral phentotypes observed in this cohort; furthermore, the authors found that while all individuals in this cohort presented with developmental delay, individuals with a pathogenic variant in the seventh spectrin repeat (which was shown by functional analysis to cause TRIO-mediated hyper-activation of RAC1) presented with a more severe intellectual disability associated with macrocephaly, whereas individuals with variants in the RAC1-activating GEFD1 domain (which were shown to cause TRIO-mediated hypo-activation of RAC1) presented with milder ID and microcephaly.

Molecular Function

Promotes the exchange of GDP by GTP. Together with leukocyte antigen-related (LAR) protein, it could play a role in coordinating cell-matrix and cytoskeletal rearrangements necessary for cell migration and cell growth.

External Links

        

References

Type
Title
Type of Disorder
Associated Disorders
Author, Year
Primary
Synaptic, transcriptional and chromatin genes disrupted in autism.
ASD
Support
The contribution of de novo coding mutations to autism spectrum disorder
ASD
Support
RhoGEF Trio Regulates Radial Migration of Projection Neurons via Its Distinct Domains
Support
Targeted sequencing identifies 91 neurodevelopmental-disorder risk genes with autism and developmental-disability biases.
ASD
Support
Integrating de novo and inherited variants in 42
ASD
Support
Characterization of intellectual disability and autism comorbidity through gene panel sequencing.
ID, ASD or autistic traits
Support
De novo mutations in epileptic encephalopathies.
Epilepsy
Support
Prevalence and phenotypic impact of rare potentially damaging variants in autism spectrum disorder
ASD
Support
De novo genic mutations among a Chinese autism spectrum disorder cohort.
ASD
Support
Autoinhibition of the GEF activity of cytoskeletal regulatory protein Trio is disrupted in neurodevelopmental disorder-related genetic variants
DD, ID
ASD or autistic features, stereotypy, epilepsy/sei
Support
Elucidation of the phenotypic spectrum and genetic landscape in primary and secondary microcephaly.
Microcephaly
ADHD, learning disabilities
Support
Diagnostic exome sequencing in persons with severe intellectual disability.
ID
Support
More evidence on TRIO missense mutations in the spectrin repeat domain causing severe developmental delay and recognizable facial dysmorphism with macrocephaly
DD
Epilepsy/seizures, stereotypy
Support
Meta-analysis of 2,104 trios provides support for 10 new genes for intellectual disability
ID
Support
Clinical Targeted Panel Sequencing Analysis in Clinical Evaluation of Children with Autism Spectrum Disorder in China
ASD
Support
Inherited and multiple de novo mutations in autism/developmental delay risk genes suggest a multifactorial model.
ASD
Support
Sporadic autism exomes reveal a highly interconnected protein network of de novo mutations.
ASD
Support
ASD, DD, ID
Support
Comorbidities associated with genetic abnormalities in children with intellectual disability
DD/ID
Support
Mutations specific to the Rac-GEF domain of TRIO cause intellectual disability and microcephaly.
DD
Microcephaly
Support
Trio-based exome sequencing reveals a high rate of the de novo variants in intellectual disability
ID
Support
Integrative Analyses of De Novo Mutations Provide Deeper Biological Insights into Autism Spectrum Disorder.
ASD
Support
De novo mutations revealed by whole-exome sequencing are strongly associated with autism.
ASD
Support
ASD
DD, ID
Support
De novo variants in neurodevelopmental disorders-experiences from a tertiary care center
DD
Support
Insights into Autism Spectrum Disorder Genomic Architecture and Biology from 71 Risk Loci.
ASD
Support
Mutational Landscape of Autism Spectrum Disorder Brain Tissue
ASD
Support
Improved diagnostic yield compared with targeted gene sequencing panels suggests a role for whole-genome sequencing as a first-tier genetic test.
DD, microcephaly
Support
Large-scale targeted sequencing identifies risk genes for neurodevelopmental disorders
ASD, DD
Support
Large-scale discovery of novel genetic causes of developmental disorders.
DD, ID
Support
Phenotype and genotype spectrum of variants in guanine nucleotide exchange factor genes in a broad cohort of Iranian patients
ADHD, ID
Support
Hotspots of missense mutation identify neurodevelopmental disorder genes and functional domains.
DD
Support
DD
ASD, ADHD, epilepsy/seizures, stereotypy
Support
Mutations in ASH1L confer susceptibility to Tourette syndrome.
TS
Recent Recommendation
TRIO loss of function is associated with mild intellectual disability and affects dendritic branching and synapse function.
DD, ID
ADHD, autistic behaviors
Recent Recommendation
DD, ID
ASD, ADHD, epilepsy/seizures
Recent Recommendation
Low load for disruptive mutations in autism genes and their biased transmission.
ASD
Recent Recommendation
Rare coding variants in ten genes confer substantial risk for schizophrenia
Schizophrenia
Recent Recommendation
Incorporating Functional Information in Tests of Excess De Novo Mutational Load.
Recent recommendation
Opposite Modulation of RAC1 by Mutations in TRIO Is Associated with Distinct, Domain-Specific Neurodevelopmental Disorders.
DD, ID
Autistic features, stereotypy
Recent Recommendation
Neurodevelopmental disease-associated de novo mutations and rare sequence variants affect TRIO GDP/GTP exchange factor activity.
Recent Recommendation
An autism spectrum disorder-related de novo mutation hotspot discovered in the GEF1 domain of Trio.
Recent Recommendation
Genome Sequencing of Autism-Affected Families Reveals Disruption of Putative Noncoding Regulatory DNA
ASD

Rare

Variant ID
Variant Type
Allele Change
Residue Change
Inheritance Pattern
Inheritance Association
Family Type
Author, Year
 GEN664R001 
 frameshift_variant 
 c.3985del 
 p.Ile1329PhefsTer6 
 De novo 
  
 Simplex 
 GEN664R002 
 missense_variant 
 c.6658G>C 
 p.Val2220Leu 
 De novo 
  
 Simplex 
 GEN664R003 
 missense_variant 
 c.4292A>T 
 p.Lys1431Met 
 De novo 
  
 Simplex 
 GEN664R004 
 missense_variant 
 c.3934C>T 
 p.Arg1312Trp 
 De novo 
  
 Simplex 
 GEN664R005 
 missense_variant 
 c.4103A>T 
 p.Asp1368Val 
 De novo 
  
 Simplex 
 GEN664R006 
 missense_variant 
 c.7688C>T 
 p.Thr2563Met 
 De novo 
  
 Simplex 
 GEN664R007 
 missense_variant 
 c.8834C>T 
 p.Thr2945Met 
 De novo 
  
 Simplex 
 GEN664R008 
 stop_gained 
 c.7632+713G>A 
  
 Familial 
 Maternal 
 Multiplex 
 GEN664R009 
 missense_variant 
 c.2533G>A 
 p.Val845Ile 
 Familial 
 Paternal 
 Simplex 
 GEN664R010 
 missense_variant 
 c.6262G>A 
 p.Gly2088Ser 
 Familial 
 Maternal 
 Simplex 
 GEN664R011 
 missense_variant 
 c.6274C>A 
 p.Gln2092Lys 
 Familial 
 Paternal 
 Simplex 
 GEN664R012 
 missense_variant 
 c.6274C>A 
 p.Gln2092Lys 
 Familial 
 Maternal 
 Simplex 
 GEN664R013 
 missense_variant 
 c.7632+731T>A 
  
 Familial 
 Maternal 
 Simplex 
 GEN664R014 
 missense_variant 
 c.7632+791C>A 
  
 Familial 
 Maternal 
 Simplex 
 GEN664R015 
 missense_variant 
 c.9157G>A 
 p.Gly3053Ser 
 Familial 
 Paternal 
 Simplex 
 GEN664R016 
 missense_variant 
 c.5671T>C 
 p.Ser1891Pro 
 Familial 
 Maternal 
 Simplex 
 GEN664R017 
 missense_variant 
 c.7079A>C 
 p.Gln2360Pro 
 Familial 
 Maternal 
 Simplex 
 GEN664R018 
 missense_variant 
 c.7922A>C 
 p.Lys2641Thr 
 Familial 
 Maternal 
 Simplex 
 GEN664R019 
 missense_variant 
 c.440A>T 
 p.Glu147Val 
 Familial 
 Paternal 
 Simplex 
 GEN664R020 
 missense_variant 
 c.7361C>T 
 p.Ala2454Val 
 Familial 
 Paternal 
 Simplex 
 GEN664R021 
 missense_variant 
 c.7361C>T 
 p.Ala2454Val 
 Familial 
 Paternal 
 Multiplex 
 GEN664R022 
 missense_variant 
 c.925C>T 
 p.Pro309Ser 
 Unknown 
  
 Unknown 
 GEN664R023 
 missense_variant 
 c.2237A>G 
 p.Asn746Ser 
 Unknown 
  
 Unknown 
 GEN664R024 
 missense_variant 
 c.3149A>T 
 p.Asn1050Ile 
 Unknown 
  
 Unknown 
 GEN664R025 
 missense_variant 
 c.6353C>G 
 p.Ser2118Cys 
 Unknown 
  
 Unknown 
 GEN664R026 
 missense_variant 
 c.6611A>G 
 p.Lys2204Arg 
 Unknown 
  
 Unknown 
 GEN664R027 
 missense_variant 
 c.7447G>C 
 p.Gly2483Arg 
 Unknown 
  
 Unknown 
 GEN664R028 
 missense_variant 
 c.7910G>A 
 p.Arg2637His 
 Unknown 
  
 Unknown 
 GEN664R029 
 missense_variant 
 c.463G>A 
 p.Val155Met 
 Unknown 
  
 Unknown 
 GEN664R030 
 missense_variant 
 c.8027A>G 
 p.Asn2676Ser 
 Unknown 
  
 Unknown 
 GEN664R031 
 missense_variant 
 c.2699G>A 
 p.Arg900Gln 
 Unknown 
  
 Unknown 
 GEN664R032 
 missense_variant 
 c.946G>A 
 p.Asp316Asn 
 Unknown 
  
 Unknown 
 GEN664R033 
 missense_variant 
 c.3251T>A 
 p.Phe1084Tyr 
 Unknown 
  
 Unknown 
 GEN664R034 
 missense_variant 
 c.3397C>T 
 p.Arg1133Trp 
 Unknown 
  
 Unknown 
 GEN664R035 
 missense_variant 
 c.3749C>T 
 p.Ser1250Leu 
 Unknown 
  
 Unknown 
 GEN664R036 
 missense_variant 
 c.9157G>A 
 p.Gly3053Ser 
 Unknown 
  
 Unknown 
 GEN664R037 
 missense_variant 
 c.2649G>T 
 p.Glu883Asp 
 De novo 
  
 Simplex 
 GEN664R038 
 missense_variant 
 c.4381C>A 
 p.Pro1461Thr 
 De novo 
  
 Simplex 
 GEN664R039 
 missense_variant 
 c.4382C>T 
 p.Pro1461Leu 
 De novo 
  
 Simplex 
 GEN664R040 
 copy_number_loss 
  
  
 De novo 
  
  
 GEN664R041 
 stop_gained 
 c.649A>T 
 p.Arg217Ter 
 Familial 
 Paternal 
 Multiplex 
 GEN664R042 
 frameshift_variant 
 c.3752del 
 p.Asp1251ValfsTer11 
 De novo 
  
  
 GEN664R043 
 stop_gained 
 c.4128G>A 
 p.Trp1376Ter 
 De novo 
  
  
 GEN664R044 
 missense_variant 
 c.2770C>A 
 p.Arg924Ser 
 Familial 
 Paternal 
  
 GEN664R045 
 missense_variant 
 c.3712T>C 
 p.Tyr1238His 
 Unknown 
  
  
 GEN664R046 
 missense_variant 
 c.5764G>A 
 p.Ala1922Thr 
 Familial 
 Maternal 
  
 GEN664R047 
 missense_variant 
 c.5816G>A 
 p.Ser1939Asn 
 Familial 
 Paternal 
  
 GEN664R048 
 missense_variant 
 c.6601C>G 
 p.Leu2201Val 
 Unknown 
  
  
 GEN664R049 
 missense_variant 
 c.6741G>C 
 p.Glu2247Asp 
 Unknown 
  
  
 GEN664R050 
 missense_variant 
 c.8120G>A 
 p.Arg2707Gln 
 Unknown 
  
  
 GEN664R051 
 copy_number_gain 
  
  
 Familial 
 Maternal 
 Simplex 
 GEN664R052 
 frameshift_variant 
 c.4090del 
 p.Gln1364SerfsTer33 
 Familial 
 Paternal 
 Extended multiplex 
 GEN664R053 
 missense_variant 
 c.4283G>A 
 p.Arg1428Gln 
 De novo 
  
  
 GEN664R054 
 missense_variant 
 c.3239A>T 
 p.Asn1080Ile 
 De novo 
  
  
 GEN664R055 
 missense_variant 
 c.748T>G 
 p.Leu250Val 
 De novo 
  
  
 GEN664R056 
 missense_variant 
 c.6545G>A 
 p.Arg2182His 
 Familial 
 Maternal 
  
 GEN664R057 
 missense_variant 
 c.6422G>T 
 p.Arg2141Leu 
 Familial 
 Paternal 
  
 GEN664R058 
 missense_variant 
 c.8692G>A 
 p.Glu2898Lys 
 Unknown 
 Not maternal 
  
 GEN664R059 
 missense_variant 
 c.5303G>A 
 p.Arg1768Gln 
 De novo 
  
  
 GEN664R060 
 missense_variant 
 c.4375A>G 
 p.Ser1459Gly 
 Unknown 
  
  
 GEN664R061 
 splice_site_variant 
 c.3936G>A 
 p.Arg1312= 
 De novo 
  
  
 GEN664R062 
 missense_variant 
 c.131C>T 
 p.Ala44Val 
 De novo 
  
 Simplex 
 GEN664R063 
 missense_variant 
 c.8729G>A 
 p.Arg2910Lys 
 Unknown 
 Not paternal 
 Simplex 
 GEN664R064 
 splice_site_variant 
 c.4615-2del 
  
 De novo 
  
 Simplex 
 GEN664R065 
 missense_variant 
 c.4111C>T 
 p.His1371Tyr 
 De novo 
  
  
 GEN664R066 
 stop_gained 
 c.4231C>T 
 p.Arg1411Ter 
 Familial 
 Maternal 
 Simplex 
 GEN664R067 
 missense_variant 
 c.8500C>T 
 p.Arg2834Cys 
 De novo 
  
 Simplex 
 GEN664R068 
 missense_variant 
 c.3224C>T 
 p.Thr1075Ile 
 Unknown 
  
  
 GEN664R069 
 missense_variant 
 c.3232C>T 
 p.Arg1078Trp 
 De novo 
  
  
 GEN664R070 
 missense_variant 
 c.3232C>T 
 p.Arg1078Trp 
 De novo 
  
  
 GEN664R071 
 missense_variant 
 c.2848C>T 
 p.His950Tyr 
 De novo 
  
  
 GEN664R072 
 missense_variant 
 c.2848C>T 
 p.His950Tyr 
 De novo 
  
  
 GEN664R073 
 missense_variant 
 c.2848C>G 
 p.His950Asp 
 De novo 
  
  
 GEN664R074 
 missense_variant 
 c.3233G>A 
 p.Arg1078Gln 
 De novo 
  
  
 GEN664R075 
 missense_variant 
 c.3233G>A 
 p.Arg1078Gln 
 De novo 
  
  
 GEN664R076 
 missense_variant 
 c.3895G>A 
 p.Glu1299Lys 
 De novo 
  
  
 GEN664R077 
 missense_variant 
 c.4283G>A 
 p.Arg1428Gln 
 De novo 
  
  
 GEN664R078 
 missense_variant 
 c.4406A>G 
 p.His1469Arg 
 De novo 
  
  
 GEN664R079 
 missense_variant 
 c.4382C>T 
 p.Pro1461Leu 
 De novo 
  
  
 GEN664R080 
 missense_variant 
 c.4382C>T 
 p.Pro1461Leu 
 De novo 
  
  
 GEN664R081 
 stop_gained 
 c.2302C>T 
 p.Gln768Ter 
 Unknown 
  
  
 GEN664R082 
 splice_site_variant 
 c.4860-2A>G 
  
 De novo 
  
  
 GEN664R083 
 frameshift_variant 
 c.5708dup 
 p.Pro1904ThrfsTer11 
 De novo 
  
  
 GEN664R084 
 frameshift_variant 
 c.6667del 
 p.Leu2223PhefsTer16 
 De novo 
  
  
 GEN664R085 
 frameshift_variant 
 c.7078del 
 p.Gln2360ArgfsTer53 
 Unknown 
  
  
 GEN664R086 
 missense_variant 
 c.4283G>A 
 p.Arg1428Gln 
 De novo 
  
  
 GEN664R087 
 missense_variant 
 c.2465C>A 
 p.Thr822Lys 
 De novo 
  
  
 GEN664R088 
 frameshift_variant 
 c.6400del 
 p.Val2134SerfsTer4 
 Familial 
 Maternal 
  
 GEN664R089 
 frameshift_variant 
 c.6400del 
 p.Val2134SerfsTer4 
 Familial 
 Maternal 
  
 GEN664R090 
 frameshift_variant 
 c.3657dup 
 p.Cys1220MetfsTer7 
 Unknown 
  
  
 GEN664R091 
 stop_gained 
 c.5497A>T 
 p.Arg1833Ter 
 Unknown 
  
  
 GEN664R092 
 missense_variant 
 c.3869C>T 
 p.Ala1290Val 
 Unknown 
  
  
 GEN664R093 
 missense_variant 
 c.4073A>G 
 p.Glu1358Gly 
 Unknown 
  
  
 GEN664R094 
 missense_variant 
 c.1072C>T 
 p.His358Tyr 
 Unknown 
  
  
 GEN664R095 
 missense_variant 
 c.3299C>T 
 p.Thr1100Met 
 Unknown 
  
  
 GEN664R096 
 missense_variant 
 c.3695C>T 
 p.Ser1232Phe 
 Unknown 
  
  
 GEN664R097 
 missense_variant 
 c.5855C>T 
 p.Ser1952Phe 
 Unknown 
  
  
 GEN664R098 
 missense_variant 
 c.6559C>T 
 p.Arg2187Cys 
 Unknown 
  
  
 GEN664R099 
 missense_variant 
 c.2479C>T 
 p.Arg827Cys 
 Unknown 
  
  
 GEN664R100 
 missense_variant 
 c.8131C>T 
 p.Arg2711Cys 
 Unknown 
  
  
 GEN664R101 
 missense_variant 
 c.8131C>T 
 p.Arg2711Cys 
 Unknown 
  
  
 GEN664R102 
 missense_variant 
 c.3868G>T 
 p.Ala1290Ser 
 Unknown 
  
  
 GEN664R103 
 missense_variant 
 c.2276C>T 
 p.Thr759Met 
 Unknown 
  
  
 GEN664R104 
 stop_gained 
 c.5032C>T 
 p.Arg1678Ter 
 Unknown 
  
  
 GEN664R105 
 frameshift_variant 
 c.1061_1064del 
 p.Asp354GlyfsTer10 
 Unknown 
  
  
 GEN664R106 
 frameshift_variant 
 c.433del 
 p.Leu145CysfsTer13 
 Unknown 
  
  
 GEN664R107 
 splice_site_variant 
 c.347+1G>C 
  
 Unknown 
  
  
 GEN664R108 
 stop_gained 
 c.1690C>T 
 p.Gln564Ter 
 Unknown 
  
  
 GEN664R109 
 stop_gained 
 c.4673G>A 
 p.Trp1558Ter 
 Unknown 
  
  
 GEN664R110 
 stop_gained 
 c.5581G>T 
 p.Glu1861Ter 
 Unknown 
  
  
 GEN664R111 
 stop_gained 
 c.4219G>T 
 p.Glu1407Ter 
 Unknown 
  
  
 GEN664R112 
 missense_variant 
 c.2365C>T 
 p.Arg789Cys 
 Unknown 
  
  
 GEN664R113 
 missense_variant 
 c.2699G>T 
 p.Arg900Leu 
 Unknown 
  
  
 GEN664R114 
 missense_variant 
 c.4759C>T 
 p.Arg1587Cys 
 Unknown 
  
  
 GEN664R115 
 missense_variant 
 c.316A>G 
 p.Arg106Gly 
 Unknown 
  
  
 GEN664R116 
 missense_variant 
 c.697C>T 
 p.Arg233Trp 
 Unknown 
  
  
 GEN664R117 
 missense_variant 
 c.1307C>A 
 p.Ala436Asp 
 Unknown 
  
  
 GEN664R118 
 missense_variant 
 c.1981C>T 
 p.Arg661Trp 
 Unknown 
  
  
 GEN664R119 
 missense_variant 
 c.2729G>A 
 p.Arg910His 
 Unknown 
  
  
 GEN664R120 
 missense_variant 
 c.2956C>T 
 p.Arg986Trp 
 Unknown 
  
  
 GEN664R121 
 missense_variant 
 c.3256A>G 
 p.Lys1086Glu 
 Unknown 
  
  
 GEN664R122 
 missense_variant 
 c.2479C>T 
 p.Arg827Cys 
 Unknown 
  
  
 GEN664R123 
 missense_variant 
 c.5834C>T 
 p.Ser1945Leu 
 Unknown 
  
  
 GEN664R124 
 missense_variant 
 c.5834C>T 
 p.Ser1945Leu 
 Unknown 
  
  
 GEN664R125 
 missense_variant 
 c.5834C>T 
 p.Ser1945Leu 
 Unknown 
  
  
 GEN664R126 
 missense_variant 
 c.3232C>T 
 p.Arg1078Trp 
 De novo 
  
 Unknown 
 GEN664R127 
 missense_variant 
 c.6727G>A 
 p.Gly2243Ser 
 Unknown 
  
 Unknown 
 GEN664R128 
 missense_variant 
 c.3220T>C 
 p.Cys1074Arg 
 De novo 
  
  
 GEN664R129 
 missense_variant 
 c.3224C>T 
 p.Thr1075Ile 
 De novo 
  
  
 GEN664R130 
 missense_variant 
 c.3251T>A 
 p.Phe1084Tyr 
 Unknown 
  
  
 GEN664R131 
 frameshift_variant 
 c.5091_5101delinsTCTGGTGCGGACC 
 p.Val1698LeufsTer61 
 De novo 
  
 Simplex 
 GEN664R132 
 synonymous_variant 
 c.2547G>A 
 p.Gly849%3D 
 Unknown 
  
  
 GEN664R133 
 missense_variant 
 c.3232C>T 
 p.Arg1078Trp 
 De novo 
  
  
 GEN664R134 
 stop_gained 
 c.9238C>T 
 p.Arg3080Ter 
 Familial 
 Paternal 
  
 GEN664R135 
 missense_variant 
 c.6082G>A 
 p.Asp2028Asn 
 Unknown 
  
  
 GEN664R136 
 missense_variant 
 c.3800G>A 
 p.Ser1267Asn 
 Unknown 
  
  
 GEN664R137 
 synonymous_variant 
 c.3165C>T 
 p.His1055%3D 
 De novo 
  
  
 GEN664R138 
 missense_variant 
 c.7147C>G 
 p.Pro2383Ala 
 De novo 
  
 Simplex 
 GEN664R139 
 missense_variant 
 c.3211C>G 
 p.Leu1071Val 
 De novo 
  
  
 GEN664R140 
 missense_variant 
 c.3229G>C 
 p.Ala1077Pro 
 De novo 
  
  
 GEN664R141 
 missense_variant 
 c.3232C>T 
 p.Arg1078Trp 
 De novo 
  
  
 GEN664R142 
 missense_variant 
 c.3232C>T 
 p.Arg1078Trp 
 De novo 
  
  
 GEN664R143 
 missense_variant 
 c.3232C>T 
 p.Arg1078Trp 
 Unknown 
  
  
 GEN664R144 
 missense_variant 
 c.3232C>T 
 p.Arg1078Trp 
 De novo 
  
  
 GEN664R145 
 missense_variant 
 c.3371T>C 
 p.Leu1124Ser 
 Unknown 
  
  
 GEN664R146 
 missense_variant 
 c.3421G>A 
 p.Val1141Met 
 De novo 
  
  
 GEN664R147 
 missense_variant 
 c.3475G>A 
 p.Glu1159Lys 
 De novo 
  
  
 GEN664R148 
 missense_variant 
 c.4342G>A 
 p.Gly1448Arg 
 Unknown 
  
  
 GEN664R149 
 missense_variant 
 c.3211C>G 
 p.Leu1071Val 
 De novo 
  
  
 GEN664R150 
 missense_variant 
 c.3229G>C 
 p.Ala1077Pro 
 De novo 
  
  
 GEN664R151 
 missense_variant 
 c.3232C>T 
 p.Arg1078Trp 
 De novo 
  
  
 GEN664R152 
 missense_variant 
 c.3232C>T 
 p.Arg1078Trp 
 De novo 
  
  
 GEN664R153 
 missense_variant 
 c.3232C>T 
 p.Arg1078Trp 
 De novo 
  
  
 GEN664R154 
 missense_variant 
 c.3232C>T 
 p.Arg1078Trp 
 De novo 
  
  
 GEN664R155 
 missense_variant 
 c.3371T>C 
 p.Leu1124Ser 
 Unknown 
  
  
 GEN664R156 
 missense_variant 
 c.3421G>A 
 p.Val1141Met 
 De novo 
  
  
 GEN664R157 
 missense_variant 
 c.3475G>A 
 p.Glu1159Lys 
 De novo 
  
  
 GEN664R158 
 missense_variant 
 c.4112A>G 
 p.His1371Arg 
 De novo 
  
  
 GEN664R159 
 missense_variant 
 c.4283G>A 
 p.Arg1428Gln 
 De novo 
  
  
 GEN664R160 
 missense_variant 
 c.4283G>A 
 p.Arg1428Gln 
 De novo 
  
  
 GEN664R161 
 missense_variant 
 c.4342G>A 
 p.Gly1448Arg 
 De novo 
  
  
 GEN664R162 
 missense_variant 
 c.4394A>G 
 p.Asn1465Ser 
 Unknown 
  
  
 GEN664R163 
 missense_variant 
 c.4394A>G 
 p.Asn1465Ser 
 Unknown 
  
  
 GEN664R164 
 missense_variant 
 c.6239T>C 
 p.Phe2080Ser 
 De novo 
  
  
 GEN664R165 
 frameshift_variant 
 c.2926del 
 p.Gln976ArgfsTer9 
 De novo 
  
  
 GEN664R166 
 frameshift_variant 
 c.3727del 
 p.Glu1243LysfsTer19 
 De novo 
  
  
 GEN664R167 
 copy_number_loss 
 c.3949-122_4312-240del 
  
 De novo 
  
  
 GEN664R168 
 stop_gained 
 c.4231C>T 
 p.Arg1411Ter 
 De novo 
  
  
 GEN664R169 
 splice_site_variant 
 c.5203+1dup 
  
 Familial 
 Unknown 
  
 GEN664R170 
 frameshift_variant 
 c.5419del 
 p.Arg1807AlafsTer33 
 Unknown 
  
  
 GEN664R171 
 splice_site_variant 
 c.6244-2A>G 
  
 De novo 
  
  
 GEN664R172 
 frameshift_variant 
 c.6554_6557del 
 p.Glu2185GlyfsTer35 
 Familial 
 Maternal 
  
 GEN664R173 
 frameshift_variant 
 c.6995del 
 p.Ser2332ThrfsTer81 
 De novo 
  
  
 GEN664R174 
 missense_variant 
 c.5303G>A 
 p.Arg1768Gln 
 De novo 
  
 Multiplex 
 GEN664R175 
 frameshift_variant 
 c.6400del 
 p.Val2134SerfsTer4 
 Familial 
 Maternal 
 Multiplex 
 GEN664R176 
 missense_variant 
 c.7193A>C 
 p.Asp2398Ala 
 Unknown 
  
 Simplex 
 GEN664R177 
 frameshift_variant 
 c.2638_2641dup 
 p.Leu881ArgfsTer8 
 Unknown 
  
  
  et al.  

Common

No Common Variants Available
Chromosome
CNV Locus
CNV Type
# of studies
Animal Model
5
Deletion-Duplication
 37
 
5
Deletion
 1
 
5
Duplication
 4
 
5
Duplication
 2
 
5
Duplication
 1
 
5
Duplication
 2
 
5
Deletion
 1
 
5
Deletion
 2
 
5
Deletion
 3
 
5
Deletion
 4
 

Model Summary

Mice with selective Trio heterozygous deletion in the dorsal telencephalon through Nex-Cre recombination show decrease in social approach, motor coordination, brain weight and neuron size, apical and basal dendritic length, dendritic branching, and LTP, abnormal synaptic anatomy, function and plasticity, and increased dendritic spine density and increased anxiety in males. Mice with selective Trio homozygous deletion in the dorsal telencephalon through Nex-Cre recombination show increase in syna

References

Type
Title
Author, Year
Primary
Trio Haploinsufficiency Causes Neurodevelopmental Disease-Associated Deficits.

M_TRIO_1_CKO_HT

Model Type: Genetic
Model Genotype: Heterozygous
Mutation: Neurod6-Trio heterozygous mice with selective deletion of Trio in excitatory neurons were generated by crossing mice bearing a conditional Trio allele in which exons 2225 are deleted using NEX(Neurod6)-Cre mice creating a premature stop codon at the start of the Trio GEF1 domain.Thy1-GFP reporter mice were used to label neurons.
Allele Type: Conditional (knockout)
Strain of Origin:
Genetic Background: C57BL/6*129/SvJ
ES Cell Line:
Mutant ES Cell Line:
Model Source: PMIDs 25727174 (Zong W, et al., Brain Res., 2015), 17146780, (Goebbels S., et al., Genesis, 2006), 20510892 (Porrero C, et al., Brain Res., 2010)

M_TRIO_2_CKO_HM

Model Type: Genetic
Model Genotype: Heterozygous
Mutation: Neurod6-Trio heterozygous mice with selective deletion of Trio in excitatory neurons were generated by crossing mice bearing a conditional Trio allele in which exons 2225 are deleted using NEX(Neurod6)-Cre mice creating a premature stop codon at the start of the Trio GEF1 domain.Thy1-GFP reporter mice were used to label neurons.
Allele Type: Conditional knockout
Strain of Origin:
Genetic Background: C57BL/6*129/SvJ
ES Cell Line:
Mutant ES Cell Line:
Model Source: PMIDs 25727174 (Zong W, et al., Brain Res., 2015), 17146780, (Goebbels S., et al., Genesis, 2006), 20510892 (Porrero C, et al., Brain Res., 2010)

M_TRIO_1_CKO_HT

Category
Entity
Quantity
Experimental Paradigm
Age at Testing
General locomotor activity: ambulatory activity1
Decreased
Description: Mutants show decrease in total distance traveled compared with controls.
Exp Paradigm: NA
 Open field test
 6 -10 weeks
General locomotor activity1
Decreased
Description: Mutants show decrease in time spent in ambulation compared with controls.
Exp Paradigm: NA
 Open field test
 6 -10 weeks
Motor coordination and balance1
Decreased
Description: Mutants show decrease in latency to fall off the rotarod compared with controls.
Exp Paradigm: NA
 Accelerating rotarod test
 6 -10 weeks
Dendritic architecture: dendritic tree complexity1
Decreased
Description: Mutants show decrease in dendritic arborization and branch points compared with controls.
Exp Paradigm: Dendritic arborization was measured in layer 5 pyramidal neurons (l5 pns) in motor cortex (regions m1 and m2).
 Sholl analysis
 6 weeks
Synapse density1
Increased
Description: Mutants show no significant increase in synaptic density (p=0.085) compared with controls, on l5 pyramidal neurons in the motor cortex.
Exp Paradigm: Motor cortex
 Electron microscopy
 6 weeks
Dendritic architecture: dendritic length1
Decreased
Description: Mutants show decrease in apical and basal dendritic lengths compared with controls.
Exp Paradigm: Dendritic length was measured in layer 5 pyramidal neurons (l5 pns) in motor cortex (regions m1 and m2).
 Sholl analysis
 6 weeks
Post-synaptic density size1
Decreased
Description: Mutants show decrease in the length of the post-synaptic density, spine area and bouton area compared with controls.
Exp Paradigm: NA
 Electron microscopy
 6 weeks
Brain size1
Decreased
Description: Mutants show decrease in total brain weight compared with controls. mutants show decrease in the combined weight of the cerebral cortex and hippocampus compared with controls.
Exp Paradigm: Brain; cerebral cortex; hippocampus
 Measurement of tissue weight
 6 weeks, 3 weeks
Dendritic architecture: spine density1
Increased
Description: Mutants show increased dendritic spine density on basal dendrites of l5 pns in motor cortex compared with controls. mutants show no change in spine density in the hippocampus compared with controls.
Exp Paradigm: Motor cortex; hippocampus
 Immunohistochemistry
 6 weeks
Synaptic neuroreceptor ratio (nmdar/ampar) dependent transmission1
Increased
Description: Mutants show increase in nmdar/ampar mediated evoked epsc ratio compared with controls in the motor cortex but not in the hippocampus.
Exp Paradigm: Evoked and miniature excitatory postsynaptic currents (eepscs) in motor cortex l5 pns
 Whole-cell patch clamp
 5.3 weeks - 6.4 weeks
Presynaptic function: paired-pulse facilitation1
Increased
Description: Mutants show an increase in paired-pulse ratio compared with controls, in the motor cortex but not in the hippocampus indicating a deficit in presynaptic release probability in the motor cortex.
Exp Paradigm: L5 pns in motor cortex; hippocampus
 Whole-cell patch clamp
 5.3 weeks - 6.4 weeks
Miniature post synaptic current frequency: excitatory1
Decreased
Description: Mutants show a decrease in ampa and nmda mepsc frequencies compared with controls.
Exp Paradigm: Evoked and miniature excitatory postsynaptic currents (eepscs) in motor cortex l5 pns
 Whole-cell patch clamp
 5.3 weeks - 6.4 weeks
Synaptic plasticity: hippocampal ltp1
Decreased
Description: Mutants show a lack of induction and maintenance of theta burst stimulated-ltp compared with controls.
Exp Paradigm: L5 pns in motor cortex; hippocampus
 Whole-cell patch clamp
 5.3 weeks - 6.4 weeks
Miniature post synaptic current amplitude: excitatory1
Decreased
Description: Mutants show a decrease in ampar mepsc amplitudes but no significant decrease in nmdar mepsc amplitudes compared with controls in the motor cortex and hippocampus.
Exp Paradigm: Evoked and miniature excitatory postsynaptic currents (eepscs) in motor cortex l5 pns
 Whole-cell patch clamp
 5.3 weeks - 6.4 weeksmale
Social approach1
Decreased
Description: Mutants show no preference for an unfamiliar mouse over an object compared with controls.
Exp Paradigm: NA
 Partition test
 6 -10 weeks
Rearing behavior1
Decreased
Description: Mutants show decrease in time spent in vertical activity compared with controls.
Exp Paradigm: NA
 Open field test
 6 -10 weeks
Nest building behavior1
Increased
Description: Male mutants show an increase in the percent nestlet shredded compared with controls.
Exp Paradigm: NA
 Nest building assay
 6 -10 weeks
Anxiety1
Increased
Description: Mutants show decrease in time spent in the open arms compared with controls.
Exp Paradigm: NA
 Elevated plus maze test
 6 -10 weeks
Anxiety1
Increased
Description: Mutants show decrease in time spent in the center and decrease in percent movement in the center compared with controls.
Exp Paradigm: NA
 Open field test
 6 -10 weeks
Targeted expression1
Decreased
Description: Mutants show decrease in trio protein levels compared with controls.
Exp Paradigm: Motor cortex
 Mass spectrometry (ms)
 6 weeks
Signaling: pka pathway1
Increased
Description: Mutants show increased levels of 9 kinase-substrate pairs compared with controls, including pka and its substrates in the motor cortex.
Exp Paradigm: Motor cortex
 Mass spectrometry (ms)
 6 weeks
Protein expression level evidence1
Decreased
Description: Mutants show decrease in protein levels of pde4a5 compared with controls, in a dosage dependent manner.
Exp Paradigm: Motor cortex
 Western blot
 6 weeks
Targeted expression1
Decreased
Description: Mutants show decreased expression of trio protein in the cerebral cortex and the hippocampus compared with controls.
Exp Paradigm: Cerebral cortex and hippocampus
 Western blot
 6 weeks
Protein expression level evidence1
Abnormal
Description: Mutants show 193 upregulated and 101 downregulated proteins in the motor cortex compared with controls, including chc, pde4a, and pak4.
Exp Paradigm: NA
 Mass spectrometry (ms)
 6 weeks
Mortality/lethality1
 No change
 General observations
 Adult
Size/growth1
 No change
 Body weight measurement
 6 weeks, 3 weeks
Depression1
 No change
 Forced swim test
 6 -10 weeks
Object recognition memory1
 No change
 Novel object recognition test
 6 -10 weeks
Targeted expression1
 No change
 Western blot
 6 weeks
Grip strength1
 No change
 Grip strength test
 6 -10 weeks
Motor learning1
 No change
 Accelerating rotarod test
 6 -10 weeks
Decay kinetics of evoked post synaptic currents1
 No change
 Whole-cell patch clamp
 5.3 weeks - 6.4 weeks
Sensorimotor gating1
 No change
 Prepulse inhibition
 6 -10 weeks
Nest building behavior1
 No change
 Nest building assay
 6 -10 weeks
 Not Reported:

M_TRIO_2_CKO_HM

Category
Entity
Quantity
Experimental Paradigm
Age at Testing
Motor learning1
Decreased
Description: Mutants showed show decrease in the rate of motor learning compared with controls.
Exp Paradigm: NA
 Accelerating rotarod test
 6-10 weeks
Motor coordination and balance1
Decreased
Description: Mutants show decrease in latency to fall off the rotarod compared with controls.
Exp Paradigm: NA
 Accelerating rotarod test
 6-10 weeks
General locomotor activity1
Increased
Description: Mutants show increase in time spent in ambulation compared with controls.
Exp Paradigm: NA
 Open field test
 6 -10 weeks
Brain size1
Decreased
Description: Mutants show decrease in total brain weight compared with controls. mutants show decrease in the combined weight of the cerebral cortex and hippocampus compared with controls.
Exp Paradigm: Brain; cerebral cortex; hippocampus
 Measurement of tissue weight
 6 weeks
Dendritic architecture: spine density1
Increased
Description: Mutants show increased dendritic spine density on basal dendrites of l5 pns in motor cortex and apical and basal dendrites of hippocampal ca1 neurons compared with controls.
Exp Paradigm: Motor cortex; hippocampus
 Immunocytochemistry
 6 weeks
Dendritic architecture: dendritic tree complexity1
Decreased
Description: Mutants show decrease in the dendritic arborization compared with controls.
Exp Paradigm: Dendritic arborization was measured in layer 5 pyramidal neurons (l5 pns) in motor cortex (regions m1 and m2).
 Sholl analysis
 6 weeks
Synapse density1
Increased
Description: Mutants show increased synaptic density compared with controls, on l5 pyramidal neurons in the motor cortex.
Exp Paradigm: NA
 Electron microscopy
 6 weeks
Dendritic architecture: dendritic length1
Decreased
Description: Mutants show decrease in apical and basal dendritic lengths compared with controls.
Exp Paradigm: Dendritic length was measured in layer 5 pyramidal neurons (l5 pns) in motor cortex (regions m1 and m2).
 Sholl analysis
 6 weeks
Post-synaptic density size1
Decreased
Description: Mutants show decrease in the length of the post-synaptic density, spine area and bouton area compared with controls.
Exp Paradigm: NA
 Electron microscopy
 6 weeks
Synaptic plasticity: hippocampal ltp1
Decreased
Description: Mutants show a lack of induction and maintenance of tbs-ltp compared with controls.
Exp Paradigm: L5 pns in motor cortex; hippocampus
 Whole-cell patch clamp
 5.3 weeks - 6.4 weeks
Miniature post synaptic current amplitude: excitatory1
Decreased
Description: Mutants show a decrease in ampa and nmda current amplitudes compared with controls.
Exp Paradigm: Evoked excitatory postsynaptic currents (eepscs) in motor cortex l5 pns
 Whole-cell patch clamp
 5.3 weeks - 6.4 weeks
Synaptic neuroreceptor ratio (nmdar/ampar) dependent transmission1
Decreased
Description: Mutants show decrease in nmdar/ampar mediated evoked epsc ratio compared with controls in the motor cortex but not in the hippocampus.
Exp Paradigm: Evoked excitatory postsynaptic currents (eepscs) in motor cortex l5 pns
 Whole-cell patch clamp
 5.3 weeks - 6.4 weeks
Decay kinetics of miniature post synaptic currents1
Increased
Description: Mutants show an increase in nmdar-mepsc decay time but no change in ampar-mepsc decay time compared with controls.
Exp Paradigm: Evoked and miniature excitatory postsynaptic currents (eepscs) in motor cortex l5 pns
 Mass spectrometry (ms)
 5.3 weeks - 6.4 weeks
Presynaptic function: paired-pulse facilitation1
Increased
Description: Mutants show an increase in paired-pulse ratio compared with controls in the motor cortex and hippocampus indicating a deficit in presynaptic release probability.
Exp Paradigm: L5 pyramidal neurons in motor cortex
 Whole-cell patch clamp
 5.3 weeks - 6.4 weeks
Miniature post synaptic current frequency: excitatory1
Decreased
Description: Mutants show a decrease in ampa and nmda mepsc frequencies compared with controls.
Exp Paradigm: Evoked and miniature excitatory postsynaptic currents (eepscs) in motor cortex l5 pns
 Whole-cell patch clamp
 5.3 weeks - 6.4 weeks
Sensorimotor gating1
Decreased
Description: Male mutants show decrease in prepulse inhibition compared with controls, at 6 db prepulse intensity with 30 ms inter-stimulus interval.
Exp Paradigm: 30 ms or 100 ms interstimulus intervals for prepulse intensities (6, 12, and 16 db)
 Prepulse inhibition
 6 -10 weeks
Rearing behavior1
Decreased
Description: Mutants show decrease in time spent in vertical activity compared with controls.
Exp Paradigm: NA
 Open field test
 6 -10 weeks
Nest building behavior1
Increased
Description: Male mutants show an increase in the percent nestlet shredded compared with controls.
Exp Paradigm: NA
 Nest building assay
 6 -10 weeks
Social approach1
Decreased
Description: Mutants show no preference for an unfamiliar mouse over an object compared with controls.
Exp Paradigm: NA
 Partition test
 6-10 weeks
Size/growth1
Decreased
Description: Mutants show decrease in body weight compared with controls.
Exp Paradigm: NA
 Body weight measurement
 3 weeks
Anxiety1
Decreased
Description: Female mutants show increase in time spent in the center and increase in percent movement in the center compared with controls.
Exp Paradigm: NA
 Open field test
 6-10 weeks
Depression1
Decreased
Description: Mutants show decrease in time spent immobile compared with controls.
Exp Paradigm: NA
 Forced swim test
 6 -10 weeks
Anxiety1
Increased
Description: Male mutants show decrease in time spent in the open arms compared with controls.
Exp Paradigm: NA
 Elevated plus maze test
 6-10 weeks
Protein expression level evidence1
Decreased
Description: Mutants show decrease in protein levels including pde4d, chc, pde4a, pak4 and pde4a5. mutants show decrease in ped4a5 in a dosage dependent manner.
Exp Paradigm: Brain
 Western blot
 6 weeks
Targeted expression1
Decreased
Description: Mutants show decreased expression of trio protein in the cerebral cortex and the hippocampus compared with controls.
Exp Paradigm: Hippocampus
 Western blot
 6 weeks
Protein expression level evidence1
Abnormal
Description: Mutants show 193 upregulated and 101 downregulated proteins in the motor cortex compared with controls, including pde4d, chc, pde4a, and pak4.
Exp Paradigm: Motor cortex
 Mass spectrometry (ms)
 6 weeks
Targeted expression1
Decreased
Description: Mutants show decrease in trio protein levels compared with controls.
Exp Paradigm: Motor cortex
 Mass spectrometry (ms)
 6 weeks
Signaling: pka pathway1
Increased
Description: Mutants show increased levels of 9 kinase-substrate pairs compared with controls, including pka and its substrates in the motor cortex.
Exp Paradigm: Motor cortex
 Mass spectrometry (ms)
 6 weeks
Mortality/lethality1
 No change
 General observations
 Adult
Size/growth1
 No change
 Body weight measurement
 6 weeks, 3 weeks
Anxiety1
 No change
 Elevated plus maze test
 6-10 weeks
Anxiety1
 No change
 Open field test
 6-10 weeks
Object recognition memory1
 No change
 Novel object recognition test
 6 -10 weeks
Targeted expression1
 No change
 Western blot
 6 weeks
General locomotor activity: ambulatory activity1
 No change
 Open field test
 6 -10 weeks
Grip strength1
 No change
 Grip strength test
 6-10 weeks
Sensorimotor gating1
 No change
 Prepulse inhibition
 6 -10 weeks
Nest building behavior1
 No change
 Nest building assay
 6 -10 weeks
 Not Reported:


Interactor Symbol Interactor Name Interactor Organism Entrez ID Uniprot ID Interaction Type Evidence Reference
CDH5 Cadherin-5 1003 P33151 IP/WB; in vitro binding assay; Co-localization
Timmerman I , et al. 2015
CHD8 chromodomain helicase DNA binding protein 8 57680 Q9HCK8 CHIP-seq
Cotney J , et al. 2015
DISC1 disrupted in schizophrenia 1 27185 Q9NRI5 Y2H
Camargo LM , et al. 2006
GNAQ guanine nucleotide binding protein (G protein), q polypeptide 2776 P50148 FCPIA; IP/WB
Lutz S , et al. 2007
TOP3B topoisomerase (DNA) III beta 8940 O95985 HITS-CLIP
Xu D , et al. 2013
ZNF232 Zinc finger protein 232 7775 Q9UNY5-2 IP; LC-MS/MS
Huttlin EL , et al. 2015
ZNF785 Zinc finger protein 785 146540 A8K8V0-2 IP; LC-MS/MS
Huttlin EL , et al. 2015

HELP
Copyright © 2017 MindSpec, Inc.