HELP     Sign In
Search

Relevance to Autism

Mutations in TSC2 cause the autosomal dominant disorder tuberous sclerosis (TSC). About 25% of individuals with TSC have autism and 40%-50% meet diagnostic criteria within the autistic spectrum disorders (Smalley et al., 1998). Schaaf et al., 2011 and Kelleher et al., 2012 identified a number of TSC2 missense variants in ASD probands that were not observed in control populations. Rare de novo and inherited missense variants in TSC2 have been identified in ASD probands from the Simons Simplex Collection (ORoak et al., 2012; Bahl et al., 2013; Iossifov et al., 2014), as well as in ASD probands from the Autism Clinical and Genetic Resources in China (ACGC) cohort (Wang et al., 2016). Targeted gene panel screening of a clinical population of 100 children with ASD from the Connecticut Childrens Medical Center Autism Neurogenetics Program in Kalsner et al., 2016 identified 18 rare TSC2 variants in ASD cases (18.0%) compared to 9.8% in the ExAC database (P = 0.0062); the statistical enrichment of rare TSC2 variants in ASD cases remained significant after multiple comparisons correction. Addtional de novo loss-of-function variants and potentially damaging missense variants in the TSC2 gene were reported in ASD probands from the Autism Sequencing Consortium, the MSSNG cohort, and the SPARK cohort in Zhou et al., 2022; a two-stage analysis of rare de novo and inherited coding variants in 42,607 ASD cases, including 35,130 new cases from the SPARK cohort, in this report identified TSC2 as a gene reaching exome-wide significance (P < 2.5E-06).

Molecular Function

The product of this gene is believed to be a tumor suppressor and is able to stimulate specific GTPases.

External Links

        

References

Type
Title
Type of Disorder
Associated Disorders
Author, Year
Primary
Autism and tuberous sclerosis.
Tuberous sclerosis-2
ASD
Positive Association
Association of INPP1, PIK3CG, and TSC2 gene variants with autistic disorder: implications for phosphatidylinositol signalling in autism.
ASD
Negative Association
Lack of association of rare functional variants in TSC1/TSC2 genes with autism spectrum disorder.
ASD
Support
Oligogenic heterozygosity in individuals with high-functioning autism spectrum disorders.
ASD
Support
Tuberous sclerosis
Support
Tuberous sclerosis complex (TSC) inactivation increases neuronal network activity by enhancing Ca 2+ influx via L-type Ca 2+ channels
Tuberous sclerosis complex
Support
Everolimus improves neuropsychiatric symptoms in a patient with tuberous sclerosis carrying a novel TSC2 mutation.
Tuberous sclerosis-2
ASD, DD, epilepsy
Support
Tuberous sclerosis 2
Support
Cerebellar demyelination and neurodegeneration associated with mTORC1 hyperactivity may contribute to the developmental onset of autism-like neurobehavioral phenotype in a rat model
ASD
Support
Neurological Diseases With Autism Spectrum Disorder: Role of ASD Risk Genes.
ASD
Tuberous sclerosis complex, epilepsy/seizures
Support
Tuberous sclerosis complex, ASD
Support
Genome sequencing of 320 Chinese children with epilepsy: a clinical and molecular study
DD, epilepsy/seizures
Support
Next-generation sequencing using a pre-designed gene panel for the molecular diagnosis of congenital disorders in pediatric patients.
Epilepsy/seizures
MCAs
Support
Epilepsy/seizures
Support
Disruption of Amygdala Tsc2 in Adolescence Leads to Changed Prelimbic Cellular Activity and Generalized Fear Responses at Adulthood in Rats
Support
Targeted resequencing of 358 candidate genes for autism spectrum disorder in a Chinese cohort reveals diagnostic potential and genotype-phenotype c...
ASD
Support
Anxiety disorder
BPD, depressive disorder, epilepsy/seizures
Support
Next-Generation Sequencing in Korean Children With Autism Spectrum Disorder and Comorbid Epilepsy
ASD
Epilepsy/seizures, tuberous sclerosis
Support
Excess of rare, inherited truncating mutations in autism.
ASD
Support
Tuberous sclerosis complex
ASD
Support
Sex-Dependent Social and Repetitive Behavior and Neurochemical Profile in Mouse Model of Autism Spectrum Disorder
ASD
Support
Diagnostic exome sequencing of syndromic epilepsy patients in clinical practice.
Epilepsy/seizures
DD, specific learning disability
Support
Developmental effects of constitutive mTORC1 hyperactivity and environmental enrichment on structural synaptic plasticity and behaviour in a rat model of autism spectrum disorder
ASD
Support
Utility of clinical exome sequencing in a complex Emirati pediatric cohort
ASD, DD
Support
Performance comparison of bench-top next generation sequencers using microdroplet PCR-based enrichment for targeted sequencing in patients with aut...
ASD
ID, epilepsy
Support
ASD
ID
Support
Autism spectrum disorder and comorbid neurodevelopmental disorders (ASD-NDDs): Clinical and genetic profile of a pediatric cohort
ASD
Epilepsy/seizures
Support
Somatic Mutations in TSC1 and TSC2 Cause Focal Cortical Dysplasia.
Focal cortical dysplasia
Support
Genetic and Phenotype Analysis of a Chinese Cohort of Infants and Children With Epilepsy
Epilepsy/seizures
ID
Support
Rare genetic susceptibility variants assessment in autism spectrum disorder: detection rate and practical use.
ASD
Support
High-throughput sequencing of mGluR signaling pathway genes reveals enrichment of rare variants in autism.
Non-syndromic ASD
Support
ID, epilepsy/seizures
Support
mTOR-related synaptic pathology causes autism spectrum disorder-associated functional hyperconnectivity
ASD
Support
Targeted sequencing identifies 91 neurodevelopmental-disorder risk genes with autism and developmental-disability biases.
ASD
Support
Drosophila functional screening of de novo variants in autism uncovers damaging variants and facilitates discovery of rare neurodevelopmental diseases
ASD
Support
Autism risk in offspring can be assessed through quantification of male sperm mosaicism.
ASD
Support
Sporadic autism exomes reveal a highly interconnected protein network of de novo mutations.
ASD
Support
DD, epilepsy/seizures
Support
Microglial ASD-related genes are involved in oligodendrocyte differentiation
Support
De novo genic mutations among a Chinese autism spectrum disorder cohort.
ASD
Support
Genetic care in geographically isolated small island communities: 8 years of experience in the Dutch Caribbean
Epilepsy/seizures
Support
Inherited and De Novo Genetic Risk for Autism Impacts Shared Networks.
ASD
Highly Cited
Interaction between hamartin and tuberin, the TSC1 and TSC2 gene products.
Highly Cited
Tsc tumour suppressor proteins antagonize amino-acid-TOR signalling.
Recent Recommendation
Regionally specific TSC1 and TSC2 gene expression in tuberous sclerosis complex.
Recent Recommendation
Tuberous sclerosis complex proteins control axon formation.
Recent Recommendation
Purkinje cells derived from TSC patients display hypoexcitability and synaptic deficits associated with reduced FMRP levels and reversed by rapamycin.
Recent Recommendation
Reversal of learning deficits in a Tsc2 mouse model of tuberous sclerosis.
Recent Recommendation
Genetic testing including targeted gene panel in a diverse clinical population of children with autism spectrum disorder: Findings and implications.
ASD
Recent Recommendation
Regulation of neuronal morphology and function by the tumor suppressors Tsc1 and Tsc2.
Recent Recommendation
Low load for disruptive mutations in autism genes and their biased transmission.
ASD
Recent Recommendation
Neuroepileptic correlates of autistic symptomatology in tuberous sclerosis.
Recent Recommendation
Loss of mTOR-dependent macroautophagy causes autistic-like synaptic pruning deficits.
Recent Recommendation
Identification of regions critical for the integrity of the TSC1-TSC2-TBC1D7 complex.
Recent Recommendation
Integrating de novo and inherited variants in 42
ASD
Recent Recommendation
ARD1 stabilization of TSC2 suppresses tumorigenesis through the mTOR signaling pathway.

Rare

Variant ID
Variant Type
Allele Change
Residue Change
Inheritance Pattern
Inheritance Association
Family Type
Author, Year
 GEN256R001 
  
  
  
  
  
  
 GEN256R002 
 missense_variant 
 c.148A>G 
 p.Met50Val 
 Unknown 
  
 Simplex 
 GEN256R003 
 missense_variant 
 c.1378G>A 
 p.Ala460Thr 
 Unknown 
  
 Simplex 
 GEN256R004 
 missense_variant 
 c.1597A>C 
 p.Lys533Gln 
 Familial 
 Maternal 
 Simplex 
 GEN256R005 
 missense_variant 
 c.1609C>T 
 p.Arg537Cys 
 Unknown 
  
 Simplex 
 GEN256R006 
 missense_variant 
 c.1816A>G 
 p.Ile606Val 
 Unknown 
  
 Simplex 
 GEN256R007 
 missense_variant 
 c.2032G>A 
 p.Ala678Thr 
 Familial (1 case); unknown (1 case) 
 Maternal (1 case) 
 Simplex 
 GEN256R008 
 missense_variant 
 c.2712C>G 
 p.Phe904Leu 
 Unknown 
  
 Simplex 
 GEN256R009 
 missense_variant 
 c.2950G>C 
 p.Glu984Gln 
 Familial 
 Paternal 
 Simplex 
 GEN256R010 
 missense_variant 
 c.3293C>T 
 p.Pro1098Leu 
 Unknown 
  
 Simplex 
 GEN256R011 
 missense_variant 
 c.3422C>T 
 p.Ala1141Val 
 Familial 
 Paternal 
 Simplex 
 GEN256R012 
 missense_variant 
 c.4106G>A 
 p.Arg1369Gln 
 Unknown 
  
 Simplex 
 GEN256R013 
 missense_variant 
 c.4273G>A 
 p.Gly1425Arg 
 Unknown 
  
 Simplex 
 GEN256R014 
 missense_variant 
 c.5094C>A 
 p.Ser1698Arg 
 Familial 
 Maternal 
 Simplex 
 GEN256R015 
 inframe_deletion 
 c.3846_3854del 
 p.Ser1282_Gly1285delinsArg 
 De novo 
  
 Simplex 
 GEN256R016 
 missense_variant 
 c.4738C>T 
 p.Arg1580Trp 
 De novo 
  
 Simplex 
 GEN256R017 
 missense_variant 
 c.433G>A 
 p.Ala145Thr 
  
  
 Multiplex 
 GEN256R018 
 synonymous_variant 
 c.618C>T 
 p.Cys206= 
  
  
 Multiplex 
 GEN256R019 
 synonymous_variant 
 c.1143G>A 
 p.Arg381= 
  
  
 Multiplex 
 GEN256R020 
 missense_variant 
 c.1292C>T 
 p.Ala431Val 
  
  
 Multiplex 
 GEN256R021 
 intron_variant 
 c.1839+6G>A 
  
  
  
 Multiplex 
 GEN256R022 
 missense_variant 
 c.1912G>A 
 p.Val638Met 
  
  
 Multiplex 
 GEN256R023 
 missense_variant 
 c.2155T>C 
 p.Tyr719His 
  
  
 Multiplex 
 GEN256R024 
 missense_variant 
 c.2621C>T 
 p.Pro874Leu 
  
  
 Multiplex 
 GEN256R025 
 synonymous_variant 
 c.3126G>T 
 p.Pro1042= 
  
  
 Multiplex 
 GEN256R026 
 missense_variant 
 c.3252C>G 
 p.Asp1084Glu 
  
  
 Multiplex 
 GEN256R027 
 missense_variant 
 c.3827C>T 
 p.Ser1276Phe 
  
  
 Multiplex 
 GEN256R028 
 missense_variant 
 c.3914C>T 
 p.Pro1305Leu 
  
  
 Multiplex 
 GEN256R029 
 missense_variant 
 c.3974G>A 
 p.Gly1325Asp 
  
  
 Multiplex 
 GEN256R030 
 missense_variant 
 c.4051G>A 
 p.Glu1351Lys 
  
  
 Multiplex 
 GEN256R031 
 missense_variant 
 c.4316G>A 
 p.Gly1439Asp 
  
  
 Multiplex 
 GEN256R032 
 synonymous_variant 
 c.4341C>T 
 p.Ser1447= 
  
  
 Multiplex 
 GEN256R033 
 missense_variant 
 c.4460C>G 
 p.Ser1487Cys 
  
  
 Multiplex 
 GEN256R034 
 synonymous_variant 
 c.5028G>A 
 p.Leu1676= 
  
  
 Multiplex 
 GEN256R035 
 intron_variant 
 c.5069-8C>T 
  
  
  
 Multiplex 
 GEN256R036 
 synonymous_variant 
 c.5175G>A 
 p.Val1725= 
  
  
 Multiplex 
 GEN256R037 
 3_prime_UTR_variant 
 c.*5G>A 
  
  
  
 Multiplex 
 GEN256R038 
 3_prime_UTR_variant 
 c.*26G>A 
  
  
  
 Multiplex 
 GEN256R039 
 missense_variant 
 c.190A>G 
 p.Ile64Val 
 Familial 
 Paternal 
 Simplex 
 GEN256R040 
 missense_variant 
 c.454C>G 
 p.His152Asp 
 Familial 
 Maternal 
 Simplex 
 GEN256R041 
 missense_variant 
 c.1597A>C 
 p.Lys533Gln 
 Familial 
 Maternal 
 Simplex 
 GEN256R042 
 missense_variant 
 c.2861A>G 
 p.Lys954Arg 
 Familial 
 Paternal 
 Simplex 
 GEN256R043 
 missense_variant 
 c.2950G>C 
 p.Glu984Gln 
 Familial 
 Paternal 
 Simplex 
 GEN256R044 
 missense_variant 
 c.4285G>T 
 p.Ala1429Ser 
 Familial 
  
 Simplex 
 GEN256R045 
 missense_variant 
 c.2032G>A 
 p.Ala678Thr 
 Unknown 
  
 Unknown 
 GEN256R046 
 missense_variant 
 c.1643G>T 
 p.Arg548Met 
 De novo 
  
 Simplex 
 GEN256R047 
 frameshift_variant 
 c.3202del 
 p.Thr1068LeufsTer2 
 Unknown 
  
 Unknown 
 GEN256R048 
 frameshift_variant 
 c.538_539del 
 p.Leu180GlyfsTer8 
 Familial 
  
 Multi-generational 
 GEN256R049 
 missense_variant 
 c.2377G>A 
 p.Glu793Lys 
 De novo 
  
  
 GEN256R050 
 missense_variant 
 c.3100G>A 
 p.Val1034Ile 
 Familial 
 Maternal 
  
 GEN256R051 
 missense_variant 
 c.1939G>A 
 p.Asp647Asn 
 Familial 
 Paternal 
  
 GEN256R052 
 stop_gained 
 c.5323A>T 
 p.Lys1775Ter 
 De novo 
  
  
 GEN256R053 
 missense_variant 
 c.4639G>A 
 p.Val1547Ile 
  
  
  
 GEN256R054 
 missense_variant 
 c.1946T>C 
 p.Met649Thr 
 Familial 
 Maternal 
  
 GEN256R055 
 missense_variant 
 c.583A>G 
 p.Ile195Val 
 De novo 
  
  
 GEN256R056 
 missense_variant 
 c.4285G>T 
 p.Ala1429Ser 
 Familial 
 Maternal 
  
 GEN256R057 
 missense_variant 
 c.2153G>C 
 p.Arg718Pro 
 Familial 
 Paternal 
  
 GEN256R058 
 missense_variant 
 c.5359G>A 
 p.Gly1787Ser 
 Unknown 
  
  
 GEN256R059 
 missense_variant 
 c.5359G>A 
 p.Gly1787Ser 
 Familial 
 Maternal 
  
 GEN256R060 
 missense_variant 
 c.5065A>G 
 p.Lys1689Glu 
 Unknown 
  
  
 GEN256R061 
 missense_variant 
 c.1747G>A 
 p.Ala583Thr 
 Familial 
 Maternal 
  
 GEN256R062 
 missense_variant 
 c.2035G>A 
 p.Val679Met 
 Unknown 
  
  
 GEN256R063 
 missense_variant 
 c.5413G>A 
 p.Glu1805Lys 
 Familial 
 Paternal 
  
 GEN256R064 
 missense_variant 
 c.1070C>T 
 p.Ala357Val 
 Familial 
 Paternal 
  
 GEN256R065 
 missense_variant 
 c.1318G>A 
 p.Gly440Ser 
 Unknown 
  
  
 GEN256R066 
 inframe_deletion 
 CCTT>C 
  
 Familial 
 Maternal 
  
 GEN256R067 
 inframe_insertion 
 C>CCAGCGGGTAGGGAATATGGGGCTCCCT 
  
 Familial 
 Paternal 
  
 GEN256R068 
 missense_variant 
 c.1747G>A 
 p.Ala583Thr 
 Unknown 
  
  
 GEN256R069 
 inframe_deletion 
 CCTT>C 
  
 Unknown 
  
  
 GEN256R070 
 missense_variant 
 c.1865G>A 
 p.Arg622Gln 
 Familial 
 Paternal 
  
 GEN256R071 
 missense_variant 
 c.1747G>A 
 p.Ala583Thr 
 Unknown 
  
  
 GEN256R072 
 missense_variant 
 c.886G>A 
 p.Val296Met 
 Familial 
 Paternal 
  
 GEN256R073 
 splice_site_variant 
 c.4662+1G>A 
  
 De novo 
  
  
 GEN256R074 
 frameshift_variant 
 c.4538_4548del 
 p.Glu1513AlafsTer7 
 De novo 
  
  
 GEN256R075 
 missense_variant 
 c.919C>G 
 p.His307Asp 
 Familial 
 Maternal 
  
 GEN256R076 
 missense_variant 
 c.1864C>T 
 p.Arg622Trp 
 Familial 
 Paternal 
  
 GEN256R077 
 missense_variant 
 c.2636C>T 
 p.Ser879Phe 
 De novo 
  
 Multiplex (monozygotic twins) 
 GEN256R078 
 splice_site_variant 
 c.600-1G>A 
  
 De novo 
  
 Simplex 
 GEN256R079 
 frameshift_variant 
 c.3023_3026del 
 p.Val1008AlafsTer7 
 De novo 
  
 Simplex 
 GEN256R080 
 missense_variant 
 c.1754G>A 
 p.Arg585His 
 Unknown 
  
 Simplex 
 GEN256R081 
 inframe_deletion 
 c.4744_4746del 
 p.Ile1582del 
 Unknown 
  
  
 GEN256R082 
 intron_variant 
 c.2838-122G>A 
  
 Unknown 
  
  
 GEN256R083 
 splice_region_variant 
 c.849-3T>G 
  
 Unknown 
  
  
 GEN256R084 
 splice_site_variant 
 c.1839+1G>T 
  
 Unknown 
  
  
 GEN256R085 
 stop_gained 
 c.2353C>T 
 p.Gln785Ter 
 Unknown 
  
  
 GEN256R086 
 splice_site_variant 
 c.2639+2T>C 
  
 Unknown 
  
  
 GEN256R087 
 splice_site_variant 
 c.3284+1G>A 
  
 Familial 
 Paternal 
  
 GEN256R088 
 frameshift_variant 
 c.4589dup 
 p.Val1531GlyfsTer35 
 Unknown 
  
  
 GEN256R089 
 splice_site_variant 
 c.599+1G>A 
  
 Unknown 
  
  
 GEN256R090 
 missense_variant 
 c.1864C>T 
 p.Arg622Trp 
 Familial 
 Paternal 
  
 GEN256R091 
 splice_site_variant 
 c.1443+1G>A 
  
 Unknown 
  
  
 GEN256R092 
 stop_gained 
 c.4183C>T 
 p.Gln1395Ter 
 Unknown 
  
  
 GEN256R093 
 frameshift_variant 
 c.5025_5032dup 
 p.Tyr1678CysfsTer151 
 De novo 
  
  
 GEN256R094 
 frameshift_variant 
 c.2182del 
 p.Cys728AlafsTer43 
 Unknown 
  
  
 GEN256R095 
 missense_variant 
 c.5126C>T 
 p.Pro1709Leu 
 Unknown 
  
  
 GEN256R096 
 missense_variant 
 c.632C>T 
 p.Ser211Phe 
 Unknown 
  
  
 GEN256R097 
 missense_variant 
 c.1168A>T 
 p.Thr390Ser 
 Unknown 
  
  
 GEN256R098 
 missense_variant 
 c.2636C>T 
 p.Ser879Phe 
 De novo 
  
  
 GEN256R099 
 inframe_deletion 
 c.5238_5255del 
 p.His1746_Arg1751del 
 De novo 
  
  
 GEN256R100 
 synonymous_variant 
 c.2586G>A 
 p.Ala862%3D 
 De novo 
  
 Simplex 
 GEN256R101 
 missense_variant 
 c.2044G>C 
 p.Gly682Arg 
 De novo 
  
 Simplex 
 GEN256R102 
 missense_variant 
 c.4470G>C 
 p.Glu1490Asp 
 De novo 
  
 Simplex 
 GEN256R103 
 splice_site_variant 
 c.1444-1G>A 
  
 De novo 
  
  
 GEN256R104 
 missense_variant 
 c.4537G>A 
 p.Glu1513Lys 
 De novo 
  
  
 GEN256R105 
 splice_site_variant 
 c.775-1G>C 
  
 De novo 
  
  
 GEN256R106 
 stop_gained 
 c.1957A>T 
 p.Arg653Ter 
 De novo 
  
  
 GEN256R107 
 frameshift_variant 
 c.2086_2087del 
 p.Cys696LeufsTer6 
 De novo 
  
  
 GEN256R108 
 missense_variant 
 c.2089T>G 
 p.Leu697Val 
 De novo 
  
  
 GEN256R109 
 frameshift_variant 
 c.2475del 
 p.Leu826TrpfsTer3 
 De novo 
  
  
 GEN256R110 
 missense_variant 
 c.2742G>C 
 p.Lys914Asn 
 De novo 
  
  
 GEN256R111 
 missense_variant 
 c.4856T>C 
 p.Phe1619Ser 
 De novo 
  
  
 GEN256R112 
 stop_gained 
 c.3412C>T 
 p.Arg1138Ter 
 Unknown 
  
  
 GEN256R113 
 frameshift_variant 
 c.2859dup 
 p.Lys954GlnfsTer6 
 Unknown 
  
  
 GEN256R114 
 intron_variant 
 c.2838-122G>A 
  
 Unknown 
  
 Simplex 
 GEN256R115 
 missense_variant 
 c.4672G>A 
 p.Glu1558Lys 
 Unknown 
  
  
  et al.  
 GEN256R116 
 missense_variant 
 c.4678G>A 
 p.Ala1560Thr 
 De novo 
  
  
  et al.  
 GEN256R117 
 missense_variant 
 c.2070C>G 
 p.Phe690Leu 
 Familial 
 Maternal 
 Simplex 
  et al.  
 GEN256R118 
 missense_variant 
 c.880G>A 
 p.Gly294Arg 
 De novo 
  
 Simplex 
  et al.  
 GEN256R119 
 splice_site_variant 
 c.5068+2T>C 
  
 Familial 
 Paternal 
 Multiplex 
  et al.  

Common

Variant ID
Polymorphism
SNP ID
Allele Change
Residue Change
Population Origin
Population Stage
Author, Year
 GEN256C001 
 intron_variant 
 rs2073636 
 c.482-348A>G;c.755-348A>G;c.644-348A>G;c.551-348A>G 
 C/T 
 AGRE 
 Discovery 
Chromosome
CNV Locus
CNV Type
# of studies
Animal Model
16
Deletion-Duplication
 68
 
16
Duplication
 3
 
16
Deletion-Duplication
 2
 
16
Deletion
 5
 

Model Summary

To provide an alternative model for Tuberous Sclerosis (TSC).

References

Type
Title
Author, Year
Primary
Tsc2() mice develop tumors in multiple sites that express gelsolin and are influenced by genetic background.
Additional
Increased levels of anxiety-related behaviors in a Tsc2 dominant negative transgenic mouse model of tuberous sclerosis.
Additional
Tsc2 gene inactivation causes a more severe epilepsy phenotype than Tsc1 inactivation in a mouse model of tuberous sclerosis complex.
Additional
Loss of Tsc2 in Purkinje cells is associated with autistic-like behavior in a mouse model of tuberous sclerosis complex.
Additional
Rapamycin reverses impaired social interaction in mouse models of tuberous sclerosis complex.
Loss of mTOR-dependent macroautophagy causes autistic-like synaptic pruning deficits.
Additional
Loss of mTOR-dependent macroautophagy causes autistic-like synaptic pruning deficits.
Additional
Increased Excitation-Inhibition Ratio Stabilizes Synapse and Circuit Excitability in Four Autism Mouse Models.
Additional
Postnatal immune activation causes social deficits in a mouse model of tuberous sclerosis: Role of microglia and clinical implications
Additional
mTOR-related synaptic pathology causes autism spectrum disorder-associated functional hyperconnectivity

M_TSC2_1_KO_HM

Model Type: Genetic
Model Genotype: Homozygous
Mutation: Gene targeted replacement of exon 2 of Tsc2 gene with a neomycin resistance cassette.
Allele Type: Targeted (Knock Out)
Strain of Origin: Not Specified
Genetic Background: C57BL/6J or BALB/cJ
ES Cell Line: J1
Mutant ES Cell Line: Not Specified
Model Source: Not Specified

M_TSC2_2_KO_HT

Model Type: Genetic
Model Genotype: Heterozygous
Mutation: Gene targeted replacement of exon 2 of Tsc2 gene with a neomycin resistance cassette.
Allele Type: Targeted (Knock Out)
Strain of Origin: Not Specified
Genetic Background: 129/SvJae-BALB/cJ or 129/SvJae-C57BL/6J
ES Cell Line: J1
Mutant ES Cell Line: Not Specified
Model Source: Not Specified

M_TSC2_3_KO_HT

Model Type: Genetic
Model Genotype: Heterozygous
Mutation: Gene targeted replacement of exon 2 of Tsc2 gene with a neomycin resistance cassette.
Allele Type: Knockout
Strain of Origin: 129S4/SvJae
Genetic Background: Black Swiss Mice
ES Cell Line: J1
Mutant ES Cell Line: Not Specified
Model Source: David J. Kwiatkowski Lab (PMID 10491404), JAX# 004686

M_TSC2_4_DN_HM

Model Type: Genetic
Model Genotype: Homozygous
Mutation: .
Allele Type: Not Specified
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_TSC2_5_CKO_HM

Model Type: Genetic
Model Genotype: Homozygous
Mutation: Conditional deletion of exons 2-4 of the Tsc2 gene using Gfap-cre, in neurons and glia
Allele Type: Conditional loss-of-function
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_TSC2_6_CKO_HM_PURKINJEN

Model Type: Genetic
Model Genotype: Homozygous
Mutation: Conditional deletion of exons 2-4 of the Tsc2 gene using L7-cre, in Purkinje cells of the cerebellum
Allele Type: Conditional loss-of-function
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_TSC2_6_CKO_HT_PURKINJEN

Model Type: Genetic
Model Genotype: Heterozygous
Mutation: Conditional hetereozygous deletion of exons 2-4 of the Tsc2 gene using L7-cre, in Purkinje cells of the cerebellum
Allele Type: Conditional loss-of-function
Strain of Origin: Not specified
Genetic Background: 129X1/SvJ and C57BL/6J
ES Cell Line: Not specified
Mutant ES Cell Line: Not Specified
Model Source: Not Specified

M_TSC2_8_KO_HT

Model Type: Genetic
Model Genotype: Heterozygous
Mutation: Targeted deletion of a fragment from codon 74 in exon 2 to codon 164 in exon 5 of Tsc2 gene.
Allele Type: Targeted (Knock Out)
Strain of Origin: Not specified
Genetic Background: C57BL/6J
ES Cell Line: J1
Mutant ES Cell Line: Not Specified
Model Source: Not Specified

M_TSC2_10_KO_ATG7_DM

Model Type: Genetic
Model Genotype: Double KO (Heterozygous/ Homozygous)
Mutation: Tsc2 heterozygous mice were bred to Atg7 KO mice lacking Atg7 (macroautophagy deficient mice) specifically in the forebrain excitatory neurons to give rise to Tsc2 het an Atg7 neuronal -null double mutants.
Allele Type: Targeted (Knock out)
Strain of Origin:
Genetic Background: C57BL/6J
ES Cell Line:
Mutant ES Cell Line:
Model Source:

M_TSC2_9_KO_HT_GFP-LC3

Model Type: Genetic
Model Genotype: Heterozygous
Mutation: Tsc2 heterozygous mice crossed with GFP-LC3 mice to visualize the autophagosome marker protein LC3.
Allele Type: Targeted (Knock out)
Strain of Origin:
Genetic Background: C57BL/6J
ES Cell Line:
Mutant ES Cell Line:
Model Source:

M_TSC2_11_KO_HT

Model Type: Genetic
Model Genotype: Heterozygous
Mutation: Mice harborning a neo cassette in the second exon of the Tsc2 gene on chromosome 17.
Allele Type: Knockout
Strain of Origin: 129S4/SvJae
Genetic Background: 129S*C57BL/6J
ES Cell Line: J1
Mutant ES Cell Line:
Model Source: David J. Kwiatkowski Lab (PMID 10491404), JAX# 004686

M_TSC2_12_KO_HT

Model Type: Genetic
Model Genotype: Heterozygous
Mutation: Mice harborning a neo cassette in the second exon of the tsc2 gene for rescue experiments vehicle -treated with vehicle (2% dmso, 30% peg400, 5% tween80) from p21 to p28
Allele Type: Knockout
Strain of Origin: 129S4/SvJae
Genetic Background: C57BL/6J
ES Cell Line: J1
Mutant ES Cell Line:
Model Source: David J. Kwiatkowski Lab (PMID 10491404), JAX# 004686

M_TSC2_13_KO_HT_POLYIC

Model Type: Multifactorial
Model Genotype: Heterozygous
Mutation: Gene targeted replacement of exon 2 of Tsc2 gene with a neomycin resistance cassette, polyinosinic:polycytidylic acid (poly I:C) injection at P3, P7, and P14.
Allele Type: knockout
Strain of Origin: 129S4/SvJae
Genetic Background: C57Bl/6J
ES Cell Line: J1
Mutant ES Cell Line:
Model Source: David J. Kwiatkowski Lab (PMID 10491404), JAX# 004686

M_TSC2_TSC1_DM_HT

Model Type: Genetic
Model Genotype: Heterozygous
Mutation: Heterozygous Tsc1 mice (M_TSC1_7_KO_HT) and heterozygous Tsc2 mice (M_TSC2_8_KO_HT) were crossed to produce double heterozygous mutant mice (Tsc1^+/-;Tsc2^+/-).
Allele Type: Knockout
Strain of Origin:
Genetic Background: C57BL/6J
ES Cell Line: Not Specified
Mutant ES Cell Line:
Model Source: Not Specified

M_TSC2_1_KO_HM

Category
Entity
Quantity
Experimental Paradigm
Age at Testing
General characteristics1
Abnormal
Description: Abnormal general characteristics - hypoplastic liver, poor development of othre abdominal organs, absence of diaphragm, dilated v ascular channels of liver and other abdominal organs
Exp Paradigm: Histological analysis of embryos
 Histology
 E9-e11.5
Cardiovascular development and function1
Abnormal
Description: Abnormally inferiorly shifted heart
Exp Paradigm: Histological analysis of embryos
 Histology
 E9-e11.5
Mortality/lethality1
Increased
Description: Increased embryonic lethality
Exp Paradigm: General observations
 General observations
 E12.5
General characteristics1
Abnormal
Description: Delayed growth by 1-2 embryonic days; pale skin and edematous gross appearance; pericardial effusions; exencephaly
Exp Paradigm: General observations
 General observations
 E8-e12.5
 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_TSC2_2_KO_HT

Category
Entity
Quantity
Experimental Paradigm
Age at Testing
Developmental trajectory1
Abnormal
Description: Abnormal developmental trajectory - renal cysts and adenomas with dense cellular architecture
Exp Paradigm: Histological analysis of kidney
 Histology
 Unreported
Developmental trajectory1
Abnormal
Description: Abnormal developmental trajectory - development of several tumor types: renal cysts, liver hemangiomas, lung tumors, and angiosarcomas
Exp Paradigm: Histological analysis of embryos
 Histology
 15 months
Protein expression level evidence1
Increased
Description: Increased gelsolin expression in liver hemangiomas, lung adenomas, and angiosarcomas
Exp Paradigm: Gelsolin protein expression
 Immunohistochemistry
 Unreported
Protein expression level evidence1
Increased
Description: Increased gelsolin expression in kidneys
Exp Paradigm: Gelsolin protein expression
 Immunohistochemistry
 3, 6, 12 months
Protein expression level evidence1
Decreased
Description: Decreased tuberin expression in cystadenomas
Exp Paradigm: Tuberin expression
 Immunohistochemistry
 Unreported
Mortality/lethality1
 No change
 General observations
 Unreported
 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_TSC2_3_KO_HT

Category
Entity
Quantity
Experimental Paradigm
Age at Testing
Developmental trajectory1
Abnormal
Description: Abnormal developmental trajectory - renal cysts and adenomas greater than 1-10 mm2 size with dense cellular architecture and large solid adenomatous lesions in kidney
Exp Paradigm: Histological analysis of kidney
 Histology
 Unreported
Developmental trajectory1
Abnormal
Description: Abnormal developmental trajectory - increased frequency of angiosarcomas
Exp Paradigm: Histological analyses
 Histology
 Unreported
 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_TSC2_4_DN_HM

Category
Entity
Quantity
Experimental Paradigm
Age at Testing
Social interaction1
Decreased
Description: Decreased exploratory activity
Exp Paradigm: Open field test
 Open field test
 3-6 months
Anxiety1
Increased
Description: Increased levels of anxiety as demonstrated by significantly less time spent in open arms of elevated plus maze
Exp Paradigm: Elevated plus maze test
 Elevated plus maze test
 3-6 months
Cued or contextual fear conditioning1
Abnormal
Description: Abnormal contextual discrimination
Exp Paradigm: Cued and contextual stimulus (ccs) assay
 Fear conditioning test
 3-6 months
Ultrasonic vocalization1
 No change
 Monitoring ultrasonic vocalizations
 P7
Spatial reference memory1
 No change
 Morris water maze test
 3-6 months
Swimming ability1
 No change
 Morris water maze test
 3-6 months
 Not Reported: Circadian sleep/wake cycle, Developmental profile, Immune response, Maternal behavior, Molecular profile, Neuroanatomy / ultrastructure / cytoarchitecture, Neurophysiology, Physiological parameters, Repetitive behavior, Seizure, Sensory

M_TSC2_5_CKO_HM

Category
Entity
Quantity
Experimental Paradigm
Age at Testing
Brain morphology1
Increased
Description: Increased ca1 pyramidal cell layer width
Exp Paradigm: Histological analysis of hippocampus
 Histology
 3 weeks
Brain morphology1
Abnormal
Description: Abnormal morphology demonstrated by progressive neuronal disorganization in hippocampus with a dispersion of the pyramidal cell layer
Exp Paradigm: Histological analysis of hippocampus
 Histology
 Unreported
Brain morphology1
Abnormal
Description: Abnormal morphology demonstrated by increased astrocyte number in neocortex and hippocampus
Exp Paradigm: Histological analysis of neocortex and hippocampus
 Histology
 P7
Brain size1
Increased
Description: Increased brain weight with generalized megencephaly
Exp Paradigm: NA
 NA
 3 weeks
Seizures1
Increased
Description: Increased incidence of observable seizures characterized by head nodding, rearing up on hindlimbs, repetitive forelimb clonus, loss of upright posture
Exp Paradigm: Video-eeg recordings
 Electroencephalogram (eeg)
 P14-p21
Size/growth1
Decreased
Description: Decreased weight gain
Exp Paradigm: General observations
 General observations
 Unreported
Mortality/lethality1
Increased
Description: Increased lethality related to acute seizures/status epilepticus or malnutrition/dehydration
Exp Paradigm: General observations
 General observations
 7-10 weeks
Enzyme activity1
Increased
Description: Increased mtor activation demonstrated by increased phospho-s6 (p-s6) expression
Exp Paradigm: Mtor activation
 Western blot
 Unreported
General characteristics1
 No change
 General observations
 2 weeks
Mortality/lethality1
 No change
 General observations
 8-10 weeks
Brain morphology1
 No change
 Histology
 Unreported
Brain size1
 No change
 NA
 4 weeks
Seizures1
 No change
 Electroencephalogram (eeg)
 3-8 weeks
 Not Reported: Circadian sleep/wake cycle, Communications, Emotion, Immune response, Learning & memory, Maternal behavior, Motor phenotype, Neurophysiology, Physiological parameters, Repetitive behavior, Sensory, Social behavior

M_TSC2_6_CKO_HM_PURKINJEN

Category
Entity
Quantity
Experimental Paradigm
Age at Testing
Social interaction1
Decreased
Description: Decreased sociaiblity indicated by no preference shown for chamber with stranger mouse or inanimate object
Exp Paradigm: Female mice: three chambered apparatus for sociability
 Three-chamber social approach test
 2 months
Social interaction1
Decreased
Description: Decreased social novelty indicated by non-significant preference for novel mouse
Exp Paradigm: Male mice: three chambered apparatus for social novelty
 Three-chamber social approach test
 2 months
Gene expression1
Decreased
Description: Decreased expression of astrocyte glutamate transporter glt-1
Exp Paradigm: Glt-1 expression
 Western blot
 Unreported
General characteristics1
 No change
 General observations
 4 weeks
Anxiety1
 No change
 Open field test
 6 weeks
General locomotor activity1
 No change
 Open field test
 6 weeks
Motor coordination and balance1
 No change
 Accelerating rotarod test
 2 months
Neuronal number: interneurons1
 No change
 NA
 3 months
Stereotypy1
 No change
 Marble-burying test
 6 weeks
Olfaction1
 No change
 Buried food test
 7 weeks
Vision1
 No change
 Morris water maze test
 2 months
Social interaction1
 No change
 Three-chamber social approach test
 2 months
Social interaction1
 No change
 Three-chamber social approach test
 2 months
 Not Reported: Circadian sleep/wake cycle, Communications, Immune response, Learning & memory, Maternal behavior, Neurophysiology, Physiological parameters, Seizure

M_TSC2_6_CKO_HT_PURKINJEN

Category
Entity
Quantity
Experimental Paradigm
Age at Testing
General characteristics1
 No change
 General observations
 4 weeks
Anxiety1
 No change
 Open field test
 6 weeks
General locomotor activity1
 No change
 Open field test
 6 weeks
Motor coordination and balance1
 No change
 Accelerating rotarod test
 2 months
Stereotypy1
 No change
 Marble-burying test
 6 weeks
Olfaction1
 No change
 Buried food test
 7 weeks
Vision1
 No change
 Morris water maze test
 2 months
Social interaction1
 No change
 Three-chamber social approach test
 2 months
Social interaction1
 No change
 Three-chamber social approach test
 2 months
 Not Reported: Circadian sleep/wake cycle, Communications, Immune response, Learning & memory, Maternal behavior, Molecular profile, Neuroanatomy / ultrastructure / cytoarchitecture, Neurophysiology, Physiological parameters, Seizure

M_TSC2_8_KO_HT

Category
Entity
Quantity
Experimental Paradigm
Age at Testing
Dendritic architecture: spine density2
Increased
Description: Tsc2 het mice, after 3 weeks of age -the time period of spinal pruning, show increased number of spines in pyramidal neurons of layer v of the auditory cortex and the secondary somatosensory cortex. compared to wild type (26% reduction in spines) the tsc2 hets only show a reduction of spines or pruning by 3% ( these numbers are from control mice injected with dmso)
Exp Paradigm: NA
 Histology
 4 weeks
Macroautophagy: neuronal2
Decreased
Description: Tsc2 het mice have reduced macroautophagy in the cortex, due to increase in mtor activation. macroautophagy was monitored using the level of protein lc3 known to be associated with autophagosomes.
Exp Paradigm: NA
 Western blot
 4 weeks
Self grooming3
Increased
Description: Tsc2 mutant mice exhibited more frequent self-grooming behavior compared to wildtype controls.
 Grooming behavior assessments
 10 months
Social memory3
Decreased
Description: Tsc2 mutant mice exhibited impaired social memory compared to control mice, spending comparable amounts of time exploring the novel versus familiar cagemate mouse, whereas wildtype mice spent significantly more time exploring the novel mouse. This was further validated by a significant reduction in the approach-avoidance score ((time exploring S3) â?? (time exploring C)) compared to both Tsc1 and wildtype mice.
Exp Paradigm: Novel mouse: stranger (S3); Familiar mouse: cagemate
 Three-chamber social approach test
 3-10 months
Social interaction2
Decreased
Description: Tsc2 het mice spend less time sniffing the new stimulus mouse compared to wild type controls
Exp Paradigm: NA
 Reciprocal social interaction test
 4-5 weeks
Rearing behavior1
Increased
Description: Increased rearing behavior
Exp Paradigm: Social interaction test with novel mouse scored for rearing
 Reciprocal social interaction test
 3-7 months
Social memory3
Decreased
Description: Tsc2 mutant mice exhibited impaired social memory compared to control mice, spending comparable amounts of time exploring the novel versus familiar (non-cagemate) mouse, whereas wildtype mice spent significantly more time exploring the novel mouse.
Exp Paradigm: Novel mouse: stranger (S2); Familiar mouse: non-cagemate previously introduced for 10 mins (S1).
 Three-chamber social approach test
 3-10 months
Social interaction1
Decreased
Description: Decreased social interaction demonstrated by shorter time engaged in active exploration i.e. exploring a novel mouse
Exp Paradigm: Social interaction test with novel mouse scored for sniffing, allo-grooming, mounting, and following
 Reciprocal social interaction test
 3-7 months
Rearing behavior1
Increased
Description: Increased rearing behavior
Exp Paradigm: Social interaction test
 Reciprocal social interaction test
 3-7 months
Social memory2
Decreased
Description: Tsc2 het mice had a lower preference index, spend similar amount of time sniffing, the new social target compared to a familiar social target compared to controls.
Exp Paradigm: NA
 Three-chamber social approach test
 4-5 weeks
Social interaction1
Decreased
Description: Decreased active interaction
Exp Paradigm: Social interaction test
 Reciprocal social interaction test
 3-7 months
Social interaction3
Decreased
Description: Tsc2 mutant mice exhibited a decrease in the amount of time spent interacting with a wildtype mate of the same sex compared to wildtype controls.
 Reciprocal social interaction test
 4-10 months
Social approach2
Decreased
Description: Tsc2 het mice had a lower preference index towards the social target compared to a non social target compared to controls. tsc2 do spend more type with the social target compared to non social target, but the preference index is smaller than controls
Exp Paradigm: NA
 Three-chamber social approach test
 4-5 weeks
Depression1
Increased
Description: Increased depression indicated by longer immobility time
Exp Paradigm: Tail suspension test
 Tail suspension test
 3-7 months
Object recognition memory2
Decreased
Description: Tsc2 het mice spend less time with new object compared to wild type littermate controls
Exp Paradigm: NA
 Novel object recognition test
 4-5 weeks
Gene expression1
Increased
Description: Increased expression of tsc1, gsk3b, mapk1, deptor, and ulk1 mrna
Exp Paradigm: Gene expression
 Quantitative pcr (qrt-pcr)
 Unreported
Protein expression level evidence2
Increased
Description: Tsc2 het mice show increased expression of post synaptic marker psd95
Exp Paradigm: NA
 Immunostaining
 4-5 weeks
Gene expression1
Decreased
Description: Decreased mrna expression of tsc2
Exp Paradigm: Mrna expression intensity
 Gene expression microarray
 Unreported
Protein expression level evidence1
Decreased
Description: Decreased tuberin expression
Exp Paradigm: Tuberin expression
 Western blot
 Unreported
Enzyme activity1
Increased
Description: Increased mtor activation demonstrated by increased phospho-s6k levels
Exp Paradigm: Mtor activation
 Western blot
 Unreported
Protein phosphorylation1
Decreased
Description: Decreased akt phosphorylation with no change in total akt levels
Exp Paradigm: Akt phosphorylation
 Western blot
 Unreported
Gene expression1
Increased
Description: Increased mrna expression of tsc1, gsk3b, mapk1, ulk1, igbp1, rps6, deptor, and eef2k
Exp Paradigm: Mrna expression intensity
 Gene expression microarray
 Unreported
Differential gene expression3
Abnormal
Description: Tsc2 mutant mice exhibited 1225 differentially expressed transcripts compared to wildtype controls.
 RNA sequencing
 3 months
Protein expression level evidence2
Increased
Description: Tsc2 het mice show increased expression of presynaptic marker synaptophysin
Exp Paradigm: NA
 Immunostaining
 4-5 weeks
General characteristics1
 No change
 General observations
 3-7 months
Size/growth1
 No change
 General observations
 3-7 months
Anxiety1
 No change
 Light-dark exploration test
 3-7 months
Anxiety2
 No change
 Open field test
 4-5 weeks
Anxiety3
 No change
 Elevated plus maze test
 3-6 months
Exploratory activity1
 No change
 Home cage behavior
 3-7 months
Protein expression level evidence1
 No change
 Western blot
 Unreported
General locomotor activity1
 No change
 Open field test
 8-10 weeks
General locomotor activity: ambulatory activity2
 No change
 Open field test
 4-5 weeks
General locomotor activity: ambulatory activity3
 No change
 Elevated plus maze test
 3-6 months
Grip strength1
 No change
 Vertical pole test
 8-10 weeks
Motor coordination and balance1
 No change
 Accelerating rotarod test
 8-10 weeks
Rearing behavior3
 No change
 Grooming behavior assessments
 10 months
Dendritic architecture: dendritic tree complexity2
 No change
 Histology
 P19-p20, p30
Dendritic architecture: spine density2
 No change
 Histology
 P19-p30
Repetitive nose pokes3
 No change
 Hole-board test
 3-5 months
Self grooming: perseveration2
 No change
 Grooming behavior assessments
 4-5 weeks
Olfaction1
 No change
 Buried food test
 3-7 months
Pain or nociception1
 No change
 Tail flick test
 3-7 months
Social approach3
 No change
 Three-chamber social approach test
 3-10 months
Social dominance1
 No change
 Tube test of social dominance
 3-7 months
Social transmission of food preference3
 No change
 Social transmission of food preference
 4-5 months
 Not Reported: Circadian sleep/wake cycle, Communications, Developmental profile, Immune response, Learning & memory, Maternal behavior, Neuroanatomy / ultrastructure / cytoarchitecture, Neurophysiology, Physiological parameters, Repetitive behavior, Seizure, Sensory

M_TSC2_10_KO_ATG7_DM

Category
Entity
Quantity
Experimental Paradigm
Age at Testing
Dendritic architecture: spine density1
Increased
Description: By p29 the double knock out mice for tsc2 (het) atg (null) show increased number of spines as a result of lack of spinal pruning that reduces the number of spines by 26% in wild type mice between p20 and p28. in the double knock out there is only a reduction by 2%
Exp Paradigm: NA
 Histology
 4 weeks
Social approach1
Decreased
Description: The double knock out tsc2 het/ atg null mice show impaired social preference or sociability in the three chamber test, towards the social target compared to the non social target
Exp Paradigm: NA
 Three-chamber social approach test
 Unreported
Social memory1
Decreased
Description: The double knock out tsc2 het/ atg null mice show impaired social preference in the three chamber test, towards the new social target compared to the familiar social target
Exp Paradigm: NA
 Three-chamber social approach test
 Unreported
Dendritic architecture: spine density1
 No change
 Histology
 3 weeks
 Not Reported: Circadian sleep/wake cycle, Communications, Developmental profile, Emotion, Immune response, Learning & memory, Maternal behavior, Molecular profile, Motor phenotype, Neurophysiology, Physiological parameters, Repetitive behavior, Seizure, Sensory

M_TSC2_9_KO_HT_GFP-LC3

Category
Entity
Quantity
Experimental Paradigm
Age at Testing
Macroautophagy: neuronal1
Decreased
Description: Tsc2 het mice with gfp tagged lc3 protein show reduced levels of the labeled lc3 puncta confirming a reductionin autophagosome formation and macroautophagy in the cortical pyramidal neurons
Exp Paradigm: NA
 Immunostaining
 Unreported
Protein expression level evidence1
Increased
Description: Tsc2 het mice with gfp tagged lc3 protein have increased levels of phospho s6, indicating increased mtor activation, in cortical pyramidal neurons
Exp Paradigm: NA
 Western blot
 Unreported
 Not Reported: Circadian sleep/wake cycle, Communications, Developmental profile, Emotion, Immune response, Learning & memory, Maternal behavior, Motor phenotype, Neuroanatomy / ultrastructure / cytoarchitecture, Physiological parameters, Repetitive behavior, Seizure, Sensory, Social behavior

M_TSC2_11_KO_HT

Category
Entity
Quantity
Experimental Paradigm
Age at Testing
Stimulus-spike relationship: inhibitory1
Decreased
Description: Mutants show decrease in ipscs compared with controls.
Exp Paradigm: L4-evoked feedforward excitatory and inhibitory currents converging onto single l2/3 pyramidal cells were measured in the s1 somatosensory cortex in acute slices. epsc amplitudes were compared at the minimum l4 stimulation intensity required to evoke a detectable epsc and at peak current amplitude.
 Whole-cell voltage clamp
 P17-p23
Intrinsic membrane properties1
Abnormal
Description: Mutants show no change in specific membrane capacitance (cm), membrane time constant (tao-mem),spike threshold and rheobase but a decrease in input resistance (ri) compared with controls.
Exp Paradigm: S1 somatosensory cortex; acute slices
 Whole-cell voltage clamp
 P17-p23
Spontaneous post synaptic event frequency: inhibitory currents1
Decreased
Description: Mutants show decrease in spontaneous mipsc frequency in l2/l3 pyramidal neurons of the s1 somatosensory cortex compared with controls.
Exp Paradigm: S1 somatosensory cortex; acute slices; l2/3 pyramidal neurons
 Whole-cell voltage clamp
 P17-p23
Decay kinetics of miniature post synaptic currents1
Increased
Description: Mutants show increase in duration of l4 evoked post synaptic potential compared with controls.
Exp Paradigm: Single stimulus l4-evoked post-synaptic potentials recorded in l2/3 pyramidal neurons at 1.4 times the minimum l4 stimulation intensity required to evoke a detectable epsc (e-theta), with nmda currents intact.
 Whole-cell current clamp
 P17-p23
Synaptic transmission1
Increased
Description: Mutants show increase in e-i conductance ratio compared with controls.
Exp Paradigm: L4-evoked feedforward excitatory and inhibitory currents converging onto single l2/3 pyramidal cells were measured in the s1 somatosensory cortex in acute slices. epsc amplitudes were compared at the minimum l4 stimulation intensity required to evoke a detectable epsc and at peak current amplitude.
 Whole-cell voltage clamp
 P17-p23
Spontaneous post synaptic event frequency: excitatory currents1
Decreased
Description: Mutants show reduced spontaneous mepsc frequency in l2/l3 pyramidal neurons of the s1 somatosensory cortex compared with controls.
Exp Paradigm: S1 somatosensory cortex; acute slices; l2/3 pyramidal neurons
 Whole-cell voltage clamp
 P17-p23
Synaptic plasticity1
Increased
Description: Mutants show no change in peak amplitude of postsynaptic potential but show an increase in the number of spikes following a train of l4 stimulation compared with controls indicating no change in temporal summation.
Exp Paradigm: L4 train-evoked postsynaptic potentials and spikes in l2/3 pyramidal cells (5 pulses at 20 hz).
 Whole-cell current clamp
 P17-p23
Spontaneous post synaptic event amplitude: excitatory currents1
Decreased
Description: Mutants show reduction in spontaneous mepsc amplitude in l2/l3 pyramidal neurons of the s1 somatosensory cortex compared with controls.
Exp Paradigm: S1 somatosensory cortex; acute slices; l2/3 pyramidal neurons
 Whole-cell voltage clamp
 P17-p23
Action potential property: amplitude1
 No change
 Whole-cell current clamp
 P17-p23
Action potential property: firing rate1
 No change
 Whole-cell voltage clamp
 P17-p23
Action potential property: firing rate1
 No change
 Cell-attached patch clamp
 P17-p23
Action potential property: firing rate1
 No change
 Whole-cell current clamp
 P17-p23
Action potential property: rate of depolarization1
 No change
 Whole-cell current clamp
 P17-p23
Decay kinetics of miniature post synaptic currents1
 No change
 Whole-cell voltage clamp
 P17-p23
Epsp-spike relationship1
 No change
 Whole-cell voltage clamp
 P17-p23
Membrane potential1
 No change
 Whole-cell voltage clamp
 P17-p23
Network excitability1
 No change
 Whole-cell current clamp
 P17-p23
Neuronal activation1
 No change
 Whole-cell voltage clamp
 P17-p23
Spontaneous post synaptic event amplitude: inhibitory currents1
 No change
 Whole-cell voltage clamp
 P17-p23
 Not Reported: Circadian sleep/wake cycle, Communications, Developmental profile, Emotion, Immune response, Learning & memory, Maternal behavior, Molecular profile, Motor phenotype, Neuroanatomy / ultrastructure / cytoarchitecture, Physiological parameters, Repetitive behavior, Seizure, Sensory, Social behavior

M_TSC2_12_KO_HT

Category
Entity
Quantity
Experimental Paradigm
Age at Testing
Dendritic architecture: spine density2
increased
Description: Tsc2+/- mice exhibit higher spine density of the insular cortex
 Functional magnetic resonance imaging (FMRI)-resting state
 P29
Functional magnetic resonance imaging: connectivity2
increased
Description: Global connectivity analysis revealed prominent foci of increased connectivity in the pfc and insular cortex of tsc2+/- mice; seed based mapping revealed that tsc2+/- mice exhibit functional over-synchronization between the prefrontal cortex and the posterior cingulate, anterior insula, and corticalâ??striatal components of the dmn; similarly, the anterior insula was oversynchronized with prefrontal regions and the retrosplenial cortex
 Functional magnetic resonance imaging (FMRI)-resting state
 P28
Self grooming2
increased
Description: Tsc2+/â?? mutants exhibited increased stereotypical grooming behavior; fronto striatalâ??cortical hyperconnectivity was prominently associated with repetitive motor behavior
 Reciprocal social interaction test
 P29
Social interaction2
decreased
Description: Tsc2+/â?? mutants exhibited significantly decreased social investigation; analysis did not reveal any significant correlation between social scores and rsfmri hyperconnectivity
 Reciprocal social interaction test
 P29
Ultrasonic vocalization1
Abnormal
Description: Tsc2 heterozygous pups exhibit an increase in the amount of short calls, chevron, and complex call types. A decrease in two-syllable calling is also observed.
 Monitoring ultrasonic vocalizations
 P6
Object recognition memory1
 No change
 Novel object recognition test
 month 4-6
Dti: fractional anisotropy or relative anisotropy in brain regions2
 no change
 Diffusion tensor imaging
 P29-34
Myelination2
 no change
 Immunohistochemistry
 P29-34
Neuronal morphology: axonal structure2
 no change
 Retrograde labeling assay
 P29-34
Synaptic neuroreceptor ratio (nmdar/ampar) dependent transmission2
 no change
 Whole-cell patch clamp
 P29-34
Social approach1
 No change
 Three-chamber social approach test
 month 4-6
Social memory1
 No change
 Three-chamber social approach test
 month 4-6
 Not Reported:

M_TSC2_13_KO_HT_POLYIC

Category
Entity
Quantity
Experimental Paradigm
Age at Testing
Social memory1
Decreased
Description: Male Tsc2 heterozygous mice injected with poly I:C early postnatally show social memory deficits; sexual dimorphism. Unlike early postnatal injections, adult poly I:C injections do not trigger chronic deficits in social memory in Tsc2 heterozygous male mice.
 Three-chamber social approach test
 month 4-6
Ultrasonic vocalization1
Abnormal
Description: Poly I:C-treated (at P3) Tsc2 heterozygous pups, but not their WT littermates, show a call repertoire dominated by the short call type and exhibit a significant increase in calls with an abnormal harmonic structure that does not fit into any predefined call type.
 Monitoring ultrasonic vocalizations
 P6
Gene expression: activated microglia1
Increased
Description: 17 genes exhibit differential expression across prefrontal cortex, hippocampus, and cerebellum regions. Known markers of microglia were significantly enriched among up-regulated genes.
 RNA sequencing
 month 4
Object recognition memory1
 No change
 Novel object recognition test
 month 4-6
Social approach1
 No change
 Three-chamber social approach test
 month 4-6
Social memory1
 No change
 Three-chamber social approach test
 month 4-6
 Not Reported:

M_TSC2_TSC1_DM_HT

Category
Entity
Quantity
Experimental Paradigm
Age at Testing
Self grooming1
Increased
Description: TscD mutant mice exhibited more frequent self-grooming behavior compared to wildtype controls.
 Grooming behavior assessments
 10 months
Social memory1
Decreased
Description: TscD mutant mice exhibited impaired social memory compared to control mice, spending comparable amounts of time exploring the novel versus familiar cagemate mouse, whereas wildtype mice spent significantly more time exploring the novel mouse. This was further validated by a significant reduction in the approach-avoidance score ((time exploring S3) â?? (time exploring C)) compared to both Tsc1 and wildtype mice.
Exp Paradigm: Novel mouse: stranger (S3); Familiar mouse: cagemate
 Three-chamber social approach test
 3-10 months
Social memory1
Decreased
Description: TscD mutant mice exhibited impaired social memory compared to control mice, spending comparable amounts of time exploring the novel versus familiar (non-cagemate) mouse, whereas wildtype mice spent significantly more time exploring the novel mouse.
Exp Paradigm: Novel mouse: stranger (S2); Familiar mouse: non-cagemate previously introduced for 10 mins (S1).
 Three-chamber social approach test
 3-10 months
Social interaction1
Decreased
Description: TscD mutant mice exhibited a decrease in the amount of time spent interacting with a wildtype mate of the same sex compared to wildtype controls.
 Reciprocal social interaction test
 4-10 months
Differential gene expression1
Abnormal
Description: TscD mutant mice exhibited 4954 differentially expressed transcripts compared to wildtype controls.
 RNA sequencing
 3 months
Mortality/lethality: embryonic1
 No change
 Genotypic ratio of progeny from heterozygous parents
 not specified
Anxiety1
 No change
 Elevated plus maze test
 3-6 months
General locomotor activity: ambulatory activity1
 No change
 Elevated plus maze test
 3-6 months
Rearing behavior1
 No change
 Grooming behavior assessments
 10 months
Repetitive nose pokes1
 No change
 Hole-board test
 3-5 months
Social approach1
 No change
 Three-chamber social approach test
 3-10 months
Social transmission of food preference1
 No change
 Social transmission of food preference
 4-5 months
 Not Reported:


Interactor Symbol Interactor Name Interactor Organism Entrez ID Uniprot ID Interaction Type Evidence Reference
4-Sep septin 4 5414 O43236 Y2H
Sakai Y , et al. 2011
ACTN2 actinin, alpha 2 88 P35609 Y2H
Sakai Y , et al. 2011
AKT1 v-akt murine thymoma viral oncogene homolog 1 207 P31749 Metabolic labeling with 32P; IP/WB
Dan HC , et al. 2002
ANKRD35 ankyrin repeat domain 35 148741 Q8N283 Y2H
Sakai Y , et al. 2011
AR androgen receptor 367 P10275 ChIP-Seq; Capillary gel electrophoresis (CGE)
Rajan P , et al. 2011
axin1 axin 1 734298 Q9YGY0 IP/WB
Mak BC , et al. 2003
C19ORF75 SIGLEC family-like protein 1 284369 Q8N7X8 IP; LC-MS/MS
Huttlin EL , et al. 2015
C9ORF128 Protein FAM221B 392307 A6H8Z2 IP; LC-MS/MS
Huttlin EL , et al. 2015
CALM1 calmodulin 1 (phosphorylase kinase, delta) 801 P62158 Phage display; GST; Bimolecular fluorescence complementation assay
Noonan DJ , et al. 2002
CAV1 caveolin 1, caveolae protein, 22kDa 857 Q03135 IP/WB
Yamamoto Y , et al. 2002
CCNA2 cyclin A2 890 P20248 IP/WB
Catania MG , et al. 2001
CCNB1 cyclin B1 891 P14635 IP/WB
Catania MG , et al. 2001
CCND1 cyclin D1 595 P24385 IP/WB
Zacharek SJ , et al. 2005
CCND2 cyclin D2 894 P30279 IP/WB
Zacharek SJ , et al. 2005
CCND3 cyclin D3 896 P30281 IP/WB
Zacharek SJ , et al. 2005
CD79B B-cell antigen receptor complex-associated protein beta chain 974 P40259-2 IP; LC-MS/MS
Huttlin EL , et al. 2015
CD83 ITGB7 9308 Q01151 IP; LC-MS/MS
Huttlin EL , et al. 2015
CDK1 cyclin-dependent kinase 1 983 P06493 IP/WB
Catania MG , et al. 2001
CDKN1B cyclin-dependent kinase inhibitor 1B (p27, Kip1) 1027 P46527 IP/WB
Rosner M and Hengstschlger M 2004
COL20A1 collagen, type XX, alpha 1 57642 Q9P218 IP; LC-MS/MS
Huttlin EL , et al. 2015
CRB3 crumbs homolog 3 (Drosophila) 92359 Q9BUF7 IP/WB; GST
Massey-Harroche D , et al. 2007
CUL4A cullin 4A 8451 Q13619 IP/WB
Hu J , et al. 2008
DAPK1 death-associated protein kinase 1 1612 P53355 Solid phase binding assay; IP/WB; Metabolic labeling with 32P; WB
Stevens C , et al. 2008
DDB1 damage-specific DNA binding protein 1, 127kDa 1642 Q16531 IP/WB
Hu J , et al. 2008
DDIT4 DNA-damage-inducible transcript 4 54541 Q9NX09 IP/WB
Vega-Rubin-de-Celis S , et al. 2010
EEF1A1 eukaryotic translation elongation factor 1 alpha 1 1915 P68104 Y2H
Sakai Y , et al. 2011
ESR1 estrogen receptor 1 2099 P03372 IP/WB; GST
York B , et al. 2005
FBXW5 F-box and WD repeat domain containing 5 54661 Q969U6 Y2H; WB; in vitro ubiquitination assay; IP/WB
Hu J , et al. 2008
GAPDH glyceraldehyde-3-phosphate dehydrogenase 2597 P04406 Y2H
Sakai Y , et al. 2011
GRB2 growth factor receptor-bound protein 2 2885 P62993 Y2H
Wang J , et al. 2008
GSK3B glycogen synthase kinase 3 beta 2932 P49841 IP/WB
Mak BC , et al. 2003
HERC1 hect (homologous to the E6-AP (UBE3A) carboxyl terminus) domain and RCC1 (CHC1)-like domain (RLD) 1 8925 Q15751 IP; MS; IP/WB
Chong-Kopera H , et al. 2006
HSPA1A heat shock 70kDa protein 1A 3303 P08107 IP; MS; IP/WB
Nellist M , et al. 2005
HTRA1 HtrA serine peptidase 1 5654 Q92743 Y2H; GST; IP/WB; in vitro proteolysis assay
Campioni M , et al. 2010
INADL InaD-like (Drosophila) 10207 Q8NI35 Y2H; GST; IP/WB
Massey-Harroche D , et al. 2007
LNX1 ligand of numb-protein X 1, E3 ubiquitin protein ligase 84708 Q8TBB1 Y2H; in vitro ubiquitination assay; IP/WB
Guo Z , et al. 2012
MAPK1 mitogen-activated protein kinase 1 5594 P28482 IP/WB; Metabolic labeling with 32P; IP; MS
Ma L , et al. 2005
MAPKAP1 mitogen-activated protein kinase associated protein 1 79109 Q9BPZ7 IP/WB
Huang J , et al. 2009
MDFI MyoD family inhibitor 4188 Q99750 Y2H
Corominas R , et al. 2014
MKRN1 makorin ring finger protein 1 23608 Q9UHC7 Y2H
Sakai Y , et al. 2011
MLST8 MTOR associated protein, LST8 homolog (S. cerevisiae) 64223 Q9BVC4 IP/WB
Huang J , et al. 2009
MRPL21 mitochondrial ribosomal protein L21 219927 Q7Z2W9 Y2H
Sakai Y , et al. 2011
MTOR mechanistic target of rapamycin (serine/threonine kinase) 2475 P42345 IP/WB
Huang J , et al. 2009
MYCBP2 MYC binding protein 2 23077 O75592 Y2H; GST; IP/WB
Murthy V , et al. 2003
NAA10 N(alpha)-acetyltransferase 10, NatA catalytic subunit 8260 A6NM98 GST; IP/WB; in vitro acetylation assay
Kuo HP , et al. 2010
NEK1 NIMA (never in mitosis gene a)-related kinase 1 4750 Q96PY6 Y2H
Surpili MJ , et al. 2003
P4HA2 Prolyl 4-hydroxylase subunit alpha-2 8974 O15460-2 IP; LC-MS/MS
Huttlin EL , et al. 2015
P4HA3 prolyl 4-hydroxylase, alpha polypeptide III 283208 Q7Z4N8 IP; LC-MS/MS
Huttlin EL , et al. 2015
PHLDB1 pleckstrin homology-like domain, family B, member 1 23187 Q86UU1 Y2H
Sakai Y , et al. 2011
PICK1 protein interacting with PRKCA 1 9463 Q9NRD5 Y2H; GST
Sakai Y , et al. 2011
PIN1 peptidylprolyl cis/trans isomerase, NIMA-interacting 1 5300 Q13526 Y2H
Vinayagam A , et al. 2011
PKD1 polycystic kidney disease 1 (autosomal dominant) 5310 P98161 IP/WB
Dere R , et al. 2010
PLK1 polo-like kinase 1 5347 P53350 IP/WB
Astrinidis A , et al. 2005
PNKD Probable hydrolase PNKD 25953 Q8N490-2 IP; LC-MS/MS
Huttlin EL , et al. 2015
PRKCSH protein kinase C substrate 80K-H 5589 P14314 IP; LC-MS/MS
Huttlin EL , et al. 2015
RAB5A RAB5A, member RAS oncogene family 5868 P20339 IP/WB
Yamamoto Y , et al. 2002
RALA v-ral simian leukemia viral oncogene homolog A (ras related) 5898 P11233 IP/WB
Castro AF , et al. 2003
RAP1A RAP1A, member of RAS oncogene family 5906 P62834 IP/WB
Yamamoto Y , et al. 2002
RB1CC1 RB1-inducible coiled-coil 1 9821 Q8TDY2 IP/WB
Gan B , et al. 2005
RHEB Ras homolog enriched in brain 6009 Q15382 GTP hydrolysis assay
Garami A , et al. 2003
RICTOR RPTOR independent companion of MTOR, complex 2 253260 Q6R327 IP/WB
Huang J , et al. 2009
RP2 retinitis pigmentosa 2 (X-linked recessive) 6102 O75695 IP; LC-MS/MS
Huttlin EL , et al. 2015
RPL4 ribosomal protein L4 6124 P36578 Y2H
Sakai Y , et al. 2011
RPS6KA1 ribosomal protein S6 kinase, 90kDa, polypeptide 1 6195 Q15418 Metabolic labeling with 32P; in vitro kinase assay; IP/WB
Roux PP , et al. 2004
RPSA ribosomal protein SA 3921 P08865 Y2H
Sakai Y , et al. 2011
SERPINI1 serpin peptidase inhibitor, clade I (neuroserpin), member 1 5274 Q99574 Y2H; IP/WB
Sakai Y , et al. 2011
SIRT1 sirtuin 1 23411 A8K128 IP/WB
Ghosh HS , et al. 2010
SLC13A3 solute carrier family 13 (sodium-dependent dicarboxylate transporter), member 3 64849 Q8WWT9 Y2H
Sakai Y , et al. 2011
SPERT spermatid associated 220082 Q8NA61 Y2H
Corominas R , et al. 2014
SRCRB4D scavenger receptor cysteine rich domain containing, group B (4 domains) 136853 Q8WTU2 Y2H
Sakai Y , et al. 2011
TACC3 transforming, acidic coiled-coil containing protein 3 10460 Q9Y6A5 Y2H; GST; IP/WB
Gmez-Bald L , et al. 2010
TBC1D7 TBC1 domain family, member 7 51256 Q9P0N9 IP/WB; GST
Nakashima A , et al. 2007
TBC1D7 TBC1 domain family, member 7 51256 Q9P0N9 IP; LC-MS/MS
Huttlin EL , et al. 2015
TFAP4 transcription factor AP-4 (activating enhancer binding protein 4) 7023 Q01664 IP/WB; EMSA
Habib SL , et al. 2010
TK1 thymidine kinase 1, soluble 7083 P04183 Y2H
Vinayagam A , et al. 2011
TSC1 tuberous sclerosis 1 7248 Q92574 Y2H; IP/WB
van Slegtenhorst M , et al. 1998
TSC2 tuberous sclerosis 2 7249 P49815 IP/WB
Hoogeveen-Westerveld M , et al. 2012
UBC ubiquitin C 7316 P63279 IP/WB
Zheng L , et al. 2008
UBE3A ubiquitin protein ligase E3A 7337 Q05086 IP/WB; WB; GST
Zheng L , et al. 2008
US3 N/A 2703401 B9VQJ7 IP/WB; in vitro kinase assay
Chuluunbaatar U , et al. 2010
USPL1 ubiquitin specific peptidase like 1 10208 Q5W0Q7 Y2H
Sakai Y , et al. 2011
YWHAB tyrosine 3-monooxygenase/tryptophan 5-monooxygenase activation protein, beta polypeptide 7529 P31946 GST
Nellist M , et al. 2002
YWHAE tyrosine 3-monooxygenase/tryptophan 5-monooxygenase activation protein, epsilon polypeptide 7531 P62258 GST
Nellist M , et al. 2002
YWHAG tyrosine 3-monooxygenase/tryptophan 5-monooxygenase activation protein, gamma polypeptide 7532 P61981 GST
Nellist M , et al. 2002
YWHAH tyrosine 3-monooxygenase/tryptophan 5-monooxygenase activation protein, eta polypeptide 7533 Q04917 GST
Nellist M , et al. 2002
YWHAQ tyrosine 3-monooxygenase/tryptophan 5-monooxygenase activation protein, theta polypeptide 10971 P27348 GST
Nellist M , et al. 2002
YWHAZ tyrosine 3-monooxygenase/tryptophan 5-monooxygenase activation protein, zeta polypeptide 7534 P63104 Y2H; GST; IP/WB
Nellist M , et al. 2002
FMR1 fragile X mental retardation 1 14265 P35922 HITS-CLIP
Darnell JC , et al. 2011
NGFRAP1 nerve growth factor receptor (TNFRSF16) associated protein 1 27018 Q00994 IP/WB
Yasui S , et al. 2007
PIP4ks Phosphatidylinositol 5-phosphate 4-kinase type-2 gamma 117150 Q91XU3 IP/WB; in vitro kinase assay; LC-MS/MS
Mackey AM , et al. 2014
Ppp2ca protein phosphatase 2 (formerly 2A), catalytic subunit, alpha isoform 19052 P63330 IP; MS; IP/WB
Lee WJ , et al. 2006
Rbfox1 RNA binding protein, fox-1 homolog (C. elegans) 1 268859 Q9JJ43 HITS-CLIP
Weyn-Vanhentenryck SM , et al. 2014
Foxo1 forkhead box O1 56458 Q9R1E0 Y2H; IP/WB; GST
Cao Y , et al. 2006
Mapkapk2 MAP kinase-activated protein kinase 2 17164 P49138 in vitro kinase assay
Li Y , et al. 2003
PRKAA1 protein kinase, AMP-activated, alpha 1 catalytic subunit 5562 Q13131 EMSA; Metabolic labeling with 32P; in vitro kinase assay; 2-D phosphopeptide mapping
Inoki K , et al. 2003
PTK2 PTK2 protein tyrosine kinase 2 5747 Q05397 IP/WB; GST; WB
Gan B , et al. 2006
RABEP1 rabaptin, RAB GTPase binding effector protein 1 9135 Q15276 Y2H
Xiao GH , et al. 1997
ROCK1 Rho-associated, coiled-coil containing protein kinase 1 6093 Q13464 IP/WB; in vitro kinase assay
Park JH , et al. 2011
Sfn stratifin 55948 O70456 GST; IP/WB
Liu MY , et al. 2002
SMAD2 SMAD family member 2 4087 Q15796 IP/WB; GST
Birchenall-Roberts MC , et al. 2004
SMAD3 SMAD family member 3 4088 P84022 IP/WB; GST
Birchenall-Roberts MC , et al. 2004
Spry2 sprouty homolog 2 (Drosophila) 306141 Q5HZA2 IP/WB
Scott CL , et al. 2010
AMPK AMP-activated protein kinase alpha subunit 43904 O18645 IP/WB; in vitro kinase assay
Kim M and Lee JH 2015
ATPsynd ATP synthase subunit d, mitochondrial 42291 Q24251 Peptide microarray; IP/WB
Sun X , et al. 2014

HELP
Copyright © 2017 MindSpec, Inc.