HELP     Sign In
Search

Relevance to Autism

Recurrent mutations in the PTEN gene have been identified in multiple individuals with ASD as described below. Deleterious variants in PTEN have been identified in individuals presenting with ASD and macrocephaly in multiple studies (PMIDs 15805158, 18759867, 19265751, 20533527). Two de novo deleterious events in the PTEN gene were identified in exome sequencing studies in simplex ASD cases in 2012: a missense variant (p.Thr167Asn) that was classified as "severe" in O'Roak et al. Nature 2012 (PMID 22495309); and a frameshift variant (p.Cys136MetfsX44) in O'Roak et al. Science 2012 (PMID 23160955). A detailed examination of ASD cases with heterozygous PTEN mutations in Frazier et al., 2015 found that these cases had a high proportion of missense variants, showed reduced PTEN protein levels, and exhibited prominent white-matter and cognitive abnormalities compared to other groups (PMID 25288137). An additional de novo loss-of-function variant in the PTEN gene was subsequently identified in an ASD proband from 2,270 trios screened by the Autism Sequencing Consortium in De Rubeis et al., 2014 (PMID 25363760). Analysis of rare coding variation in 3,871 ASD cases and 9,937 ancestry-matched or paternal controls from the Autism Sequencing Consortium (ASC) in this report furthermore identified PTEN as a gene meeting high statistical significance with a 0.01 < FDR 0.05, meaning that this gene had a 95% chance of being a true autism gene. This gene was identified in Iossifov et al. 2015 as a strong candidate to be an ASD risk gene based on a combination of de novo mutational evidence and the absence or very low frequency of mutations in controls (PMID 26401017). A two-stage analysis of rare de novo and inherited coding variants in 42,607 ASD cases, including 35,130 new cases from the SPARK cohort, in Zhou et al., 2022 identified PTEN as a gene reaching exome-wide significance (P < 2.5E-06). PTEN has also been designated as a syndromic ASD gene, as mutations in the PTEN gene are causative for Cowden syndrome, a disorder in which a subpopulation of individuals with the syndrome develop autism (PMID 11496368).

Molecular Function

The protein encoded this gene is a phosphatidylinositol-3,4,5-trisphosphate 3-phosphatase. It contains a tensin like domain as well as a catalytic domain similar to that of the dual specificity protein tyrosine phosphatases.

External Links

        

References

Type
Title
Type of Disorder
Associated Disorders
Author, Year
Primary
PTEN mutation in a family with Cowden syndrome and autism.
Cowden syndrome
ASD
Positive Association
De novo mutations in epileptic encephalopathies.
Epilepsy
IS, LGS, DD, ID, ASD, ADHD
Positive Association
Subset of individuals with autism spectrum disorders and extreme macrocephaly associated with germline PTEN tumour suppressor gene mutations.
Macrocephaly/autism syndrome
ASD, macrocephaly
Support
Contribution of rare inherited and de novo variants in 2,871 congenital heart disease probands.
Congenital heart disease (CHD)
Neurodevelopmental disorders (NDD)
Support
Recurrent de novo mutations implicate novel genes underlying simplex autism risk.
ASD
Support
Modeling PTEN overexpression-induced microcephaly in human brain organoids
Support
Binding of PTEN to specific PDZ domains contributes to PTEN protein stability and phosphorylation by microtubule-associated serine/threonine kinases.
Support
Targeted resequencing of 358 candidate genes for autism spectrum disorder in a Chinese cohort reveals diagnostic potential and genotype-phenotype c...
ASD
Support
ASD
DD, ID
Support
Targeted DNA Sequencing from Autism Spectrum Disorder Brains Implicates Multiple Genetic Mechanisms.
ASD
Support
Genetic care in geographically isolated small island communities: 8 years of experience in the Dutch Caribbean
DD
Support
Autistic spectrum disorder in a 9-year-old girl with macrocephaly.
ASD
Support
Phenotype-to-genotype approach reveals head-circumference-associated genes in an autism spectrum disorder cohort.
ASD
Macrocephaly
Support
Cowden syndrome 1
Support
Improved diagnostic yield compared with targeted gene sequencing panels suggests a role for whole-genome sequencing as a first-tier genetic test.
DD, macrocephaly
Support
Autism-epilepsy phenotype with macrocephaly suggests PTEN, but not GLIALCAM, genetic screening.
ASD
ID, epilepsy
Support
A Temporal Activity of CA1 Neurons Underlying Short-Term Memory for Social Recognition Altered in PTEN Mouse Models of Autism Spectrum Disorder
ASD
Support
Genetic Diagnostic Evaluation of Trio-Based Whole Exome Sequencing Among Children With Diagnosed or Suspected Autism Spectrum Disorder.
DD/ID, macrocephaly
Autistic features
Support
ASD
Support
Cytoplasm-predominant Pten associates with increased region-specific brain tyrosine hydroxylase and dopamine D2 receptors in mouse model with autis...
Support
Distinct metabolic profiles associated with autism spectrum disorder versus cancer in individuals with germline PTEN mutations
Cowden syndrome
ASD, DD
Support
A mutant form of PTEN linked to autism.
Support
Inherited and De Novo Genetic Risk for Autism Impacts Shared Networks.
ASD
Support
Cowden syndrome
ASD
Support
PTEN Loss Increases the Connectivity of Fast Synaptic Motifs and Functional Connectivity in a Developing Hippocampal Network.
Support
Genotype-phenotype associations in a large PTEN Hamartoma Tumor Syndrome (PHTS) patient cohort
PTEN hamartoma tumor syndrome (PHTS)
DD
Support
Biochemical screening and PTEN mutation analysis in individuals with autism spectrum disorders and macrocephaly.
ASD
DD, epilepsy
Support
Prevalence and clinical/molecular characteristics of PTEN mutations in Turkish children with autism spectrum disorders and macrocephaly
ASD, macrocephaly
ADHD, ID
Support
Clinical spectrum of PTEN mutation in pediatric patients. A bicenter experience.
Macrocephaly
DD, ASD
Support
DD, ID
Autistic features
Support
A recessive form of extreme macrocephaly and mild intellectual disability complements the spectrum of PTEN hamartoma tumour syndrome.
ID, macrocephaly
Support
Mutational Landscape of Autism Spectrum Disorder Brain Tissue
ASD
Support
Confirmation study of PTEN mutations among individuals with autism or developmental delays/mental retardation and macrocephaly.
ASD, DD, ID
Support
Autism-associated missense genetic variants impact locomotion and neurodevelopment in Caenorhabditis elegans.
ASD
Support
Whole genome sequencing resource identifies 18 new candidate genes for autism spectrum disorder
ASD
Support
Integrating de novo and inherited variants in 42
ASD
Support
Multiplex targeted sequencing identifies recurrently mutated genes in autism spectrum disorders.
ASD
Support
Whole genome sequencing of 45 Japanese patients with intellectual disability
DD, ID
Support
A family with PTEN mutations with malignancy and an unusually high number of offspring with autism spectrum disorder: a case report.
Cowden syndrome
ASD, macrocephaly
Support
Molecular Diagnostic Yield of Chromosomal Microarray Analysis and Whole-Exome Sequencing in Children With Autism Spectrum Disorder.
Cowden syndrome
Support
Pten haploinsufficiency causes desynchronized growth of brain areas involved in sensory processing
Support
The prevalence of PTEN mutations in a clinical pediatric cohort with autism spectrum disorders, developmental delay, and macrocephaly.
ASD, DD, ID
Support
Characterization of intellectual disability and autism comorbidity through gene panel sequencing.
ID, ASD or autistic traits
Support
Prevalence of four Mendelian disorders associated with autism in 2392 affected families.
ASD
Support
Complex Diagnostics of Non-Specific Intellectual Developmental Disorder
DD, ID
Support
Novel PTEN germline mutation in a family with mild phenotype: difficulties in genetic counseling.
Learning disabilities, macrocephaly
ID
Support
Polymicrogyria is Associated With Pathogenic Variants in PTEN
Support
Identification of a PTEN mutation with reduced protein stability, phosphatase activity, and nuclear localization in Hong Kong patients with autisti...
PTEN hamartoma tumor syndrome
ASD or autistic features, DD, macrocephaly
Support
ASD, DD, epilepsy/seizures
Support
Integrated analysis of whole-exome sequencing and transcriptome profiling in males with autism spectrum disorders.
ASD
Support
PTEN Regulates Dendritic Arborization by Decreasing Microtubule Polymerization Rate
Support
Novel PTEN mutations in neurodevelopmental disorders and macrocephaly.
ASD
DD, ID
Support
A novel missense PTEN mutation identified in a patient with macrocephaly and developmental delay.
DD
Macrocephaly
Support
A combination of genetic and biochemical analyses for the diagnosis of PI3K-AKT-mTOR pathway-associated megalencephaly.
DD, megalencephaly
Support
Clinical Targeted Panel Sequencing Analysis in Clinical Evaluation of Children with Autism Spectrum Disorder in China
ASD
Support
Sporadic autism exomes reveal a highly interconnected protein network of de novo mutations.
ASD
Support
Next-Generation Sequencing in Korean Children With Autism Spectrum Disorder and Comorbid Epilepsy
ASD
Support
DD
Support
Identification of mutations in the PI3K-AKT-mTOR signalling pathway in patients with macrocephaly and developmental delay and/or autism.
DD, macrocephaly
ASD
Support
Cowden syndrome 1
Support
Excess of rare, inherited truncating mutations in autism.
ASD
Support
Behavioural and psychological features of PTEN mutations: a systematic review of the literature and meta-analysis of the prevalence of autism spectrum disorder characteristics
Cowden syndrome 1
ASD
Support
Mutation screening of the PTEN gene in patients with autism spectrum disorders and macrocephaly.
ASD
Support
Lessons Learned from Large-Scale, First-Tier Clinical Exome Sequencing in a Highly Consanguineous Population.
Macrocephaly, DD, ID
ASD
Support
Altered proliferation and networks in neural cells derived from idiopathic autistic individuals.
ASD
Support
The impact of phosphorylated PTEN at threonine 366 on cortical connectivity and behaviour
Support
Oligogenic heterozygosity in individuals with high-functioning autism spectrum disorders.
ASD
Support
Large-Scale Exome Sequencing Study Implicates Both Developmental and Functional Changes in the Neurobiology of Autism
ASD
Support
Highly Cited
Negative regulation of PKB/Akt-dependent cell survival by the tumor suppressor PTEN.
Highly Cited
The tumor suppressor, PTEN/MMAC1, dephosphorylates the lipid second messenger, phosphatidylinositol 3,4,5-trisphosphate.
Highly Cited
PTEN, a putative protein tyrosine phosphatase gene mutated in human brain, breast, and prostate cancer.
Recent Recommendation
Low load for disruptive mutations in autism genes and their biased transmission.
ASD
Recent Recommendation
Free fatty acids inhibit insulin signaling-stimulated endothelial nitric oxide synthase activation through upregulating PTEN or inhibiting Akt kinase.
Recent Recommendation
Cowden syndrome, ASD
Recent Recommendation
PTEN interacts with histone H1 and controls chromatin condensation.
Recent Recommendation
The parvalbumin/somatostatin ratio is increased in Pten mutant mice and by human PTEN ASD alleles.
Recent Recommendation
Electrical signals control wound healing through phosphatidylinositol-3-OH kinase-gamma and PTEN.
Recent Recommendation
ASD
Recent Recommendation
Loss of mTOR repressors Tsc1 or Pten has divergent effects on excitatory and inhibitory synaptic transmission in single hippocampal neuron cultures.
Recent Recommendation
Neural transcriptome of constitutional Pten dysfunction in mice and its relevance to human idiopathic autism spectrum disorder.
Recent Recommendation
ASD
Recent Recommendation
Characteristic brain magnetic resonance imaging pattern in patients with macrocephaly and PTEN mutations.
Recent Recommendation
Conformational stability and catalytic activity of PTEN variants linked to cancers and autism spectrum disorders.
Recent Recommendation
Nuclear Excluded Autism-Associated Phosphatase and Tensin Homolog Mutations Dysregulate Neuronal Growth.
Recent Recommendation
PTEN knockdown alters dendritic spine/protrusion morphology, not density.
Recent Recommendation
Functionally distinct groups of inherited PTEN mutations in autism and tumour syndromes.
Recent Recommendation
A Retroviral CRISPR-Cas9 System for Cellular Autism-Associated Phenotype Discovery in Developing Neurons.
Recent Recommendation
A secreted PTEN phosphatase that enters cells to alter signaling and survival.
Recent Recommendation
Synaptic, transcriptional and chromatin genes disrupted in autism.
ASD
Recent Recommendation
Autistic-Like Traits and Cerebellar Dysfunction in Purkinje Cell PTEN Knock-Out Mice.
Recent Recommendation
PTEN is recruited to the postsynaptic terminal for NMDA receptor-dependent long-term depression.
Recent Recommendation
Molecular and phenotypic abnormalities in individuals with germline heterozygous PTEN mutations and autism.

Rare

Variant ID
Variant Type
Allele Change
Residue Change
Inheritance Pattern
Inheritance Association
Family Type
Author, Year
 GEN204R001 
 stop_gained 
 c.534T>G 
 p.Tyr178Ter 
 Familial 
 Maternal 
  
 GEN204R002 
 missense_variant 
 c.278A>G 
 p.His93Arg 
 De novo 
  
 Simplex 
 GEN204R003 
 missense_variant 
 c.755A>G 
 p.Asp252Gly 
 De novo 
  
 Simplex 
 GEN204R004 
 missense_variant 
 c.722T>C 
 p.Phe241Ser 
 De novo 
  
 Simplex 
 GEN204R005 
 5_prime_UTR_variant 
 c.-1088C>T 
  
  
  
  
 GEN204R006 
 5_prime_UTR_variant 
  
  
 Familial 
 Maternal 
 Multiplex 
 GEN204R007 
 5_prime_UTR_variant 
 c.-1026C>A 
  
 Familial 
 Maternal 
  
 GEN204R008 
 5_prime_UTR_variant 
 c.-903G>A 
  
  
  
  
 GEN204R009 
 5_prime_UTR_variant 
 c.-903G>A 
  
  
  
  
 GEN204R010 
 missense_variant 
 c.66C>G 
 p.Asp22Glu 
 Familial 
 Paternal 
 Multiplex 
 GEN204R011 
 intron_variant 
 T>C 
  
 Familial 
 Maternal 
  
 GEN204R012 
 missense_variant 
 c.976G>A 
 p.Asp326Asn 
 De novo 
  
  
 GEN204R013 
 stop_gained 
 c.1003C>T 
 p.Arg335Ter 
 De novo 
  
  
 GEN204R014 
 missense_variant 
 c.232A>G 
 p.Thr78Ala 
 De novo 
  
 Simplex 
 GEN204R015 
 missense_variant 
 c.618C>G 
 p.Phe206Leu 
 Unknown 
  
 Simplex 
 GEN204R016 
 copy_number_loss 
  
  
 Unknown 
  
 Simplex 
 GEN204R017 
 missense_variant 
 c.500C>A 
 p.Thr167Asn 
 De novo 
  
 Simplex 
 GEN204R018 
 missense_variant 
 c.392C>T 
 p.Ala131Val 
 De novo 
  
 Simplex 
 GEN204R019 
 frameshift_variant 
 c.-115dup 
  
 De novo 
  
 Simplex 
 GEN204R020 
 stop_gained 
 c.640C>T 
 p.Gln214Ter 
 Familial 
 Maternal 
 Simplex 
 GEN204R021 
 missense_variant 
 c.402G>C 
 p.Glu134Asp 
 Familial 
 Maternal 
 Multiplex 
 GEN204R022 
 frameshift_variant 
 c.420_421insA 
 p.Ala141SerfsTer43 
 Unknown 
  
 Unknown 
 GEN204R023 
 missense_variant 
 c.208C>G 
 p.Pro70Ala 
 Unknown 
  
 Unknown 
 GEN204R024 
 initiator_codon_variant 
 c.3G>T 
 p.Leu1? 
 Unknown 
  
 Unknown 
 GEN204R025 
 stop_gained 
 c.1003C>T 
 p.Arg335Ter 
 Unknown 
  
 Unknown 
 GEN204R026 
 splice_site_variant 
 c.209+5G>A 
  
 Unknown 
  
 Unknown 
 GEN204R027 
 missense_variant 
 c.737C>T 
 p.Pro246Leu 
 De novo 
  
  
 GEN204R028 
 frameshift_variant 
 c.-478del 
  
 Unknown 
  
  
 GEN204R029 
 splice_site_variant 
 c.493-1G>A 
  
 De novo 
  
 Simplex 
 GEN204R030 
 frameshift_variant 
 c.100del 
 p.Ala34LeufsTer20 
 Unknown 
  
 Unknown 
 GEN204R031 
 missense_variant 
 c.400A>T 
 p.Met134Leu 
 Unknown 
  
 Unknown 
 GEN204R032 
 missense_variant 
 c.353A>C 
 p.His118Pro 
 De novo 
  
  
 GEN204R033 
 missense_variant 
 c.527A>G 
 p.Tyr176Cys 
 De novo 
  
  
 GEN204R034 
 missense_variant 
 c.827A>G 
 p.Asn276Ser 
 De novo 
  
  
 GEN204R035 
 stop_gained 
 c.388C>T 
 p.Arg130Ter 
 Familial 
 Paternal 
 Simplex 
 GEN204R036 
 missense_variant 
 c.470A>G 
 p.Gln157Arg 
 De novo 
  
 Multiplex (identical twins) 
 GEN204R037 
 stop_gained 
 c.416T>A 
 p.Leu139Ter 
 Familial 
 Paternal 
 Simplex 
 GEN204R038 
 frameshift_variant 
 c.520dup 
 p.Tyr174LeufsTer6 
 De novo 
  
  
 GEN204R039 
 intron_variant 
 C>G 
  
 De novo 
  
  
 GEN204R040 
 missense_variant 
 c.131G>A 
 p.Gly44Asp 
 Unknown 
  
 Unknown 
 GEN204R041 
 missense_variant 
 c.518G>A 
 p.Arg173His 
 Familial 
 Maternal 
 Multi-generational 
 GEN204R042 
 missense_variant 
 c.605C>T 
 p.Thr202Ile 
 De novo 
  
  
 GEN204R043 
 splice_site_variant 
 G>A 
  
 Unknown 
  
  
 GEN204R044 
 stop_gained 
 c.697C>T 
 p.Arg233Ter 
 Unknown 
 Not paternal 
 Multiplex 
 GEN204R045 
 splice_site_variant 
 A>G 
  
 Familial 
 Paternal 
 Simplex 
 GEN204R046 
 missense_variant 
 c.369C>G 
 p.Tyr123Ter 
 Unknown 
  
 Unknown 
 GEN204R047 
 missense_variant 
 c.518G>A 
 p.Arg173His 
 Familial 
 Paternal 
 Simplex 
 GEN204R048 
 missense_variant 
 c.821G>T 
 p.Trp274Leu 
 Familial 
 Maternal 
 Multi-generational 
 GEN204R049 
 missense_variant 
 c.401T>C 
 p.Met134Thr 
 Familial 
 Maternal 
 Multi-generational 
 GEN204R050 
 inframe_insertion 
 c.239-21G>C 
  
 De novo 
  
 Simplex 
 GEN204R051 
 missense_variant 
 c.274G>A 
 p.Asp92Asn 
 De novo 
  
 Simplex 
 GEN204R052 
 missense_variant 
 c.494G>T 
 p.Gly165Val 
 De novo 
  
  
 GEN204R053a 
 missense_variant 
 c.545T>C 
 p.Val182Ala 
 Familial 
 Both parents 
 Multiplex 
 GEN204R054 
 stop_gained 
 c.195C>A 
 p.Cys65Ter 
 Unknown 
  
 Unknown 
 GEN204R055 
 stop_gained 
 c.640C>T 
 p.Gln214Ter 
 Unknown 
 Not maternal 
 Simplex 
 GEN204R056 
 missense_variant 
 c.741T>C 
 p.Leu247Ser 
 De novo 
  
 Simplex 
 GEN204R057 
 stop_gained 
 NM_000314.5:c.1006C>G 
 p.Tyr336Ter 
 De novo 
  
 Simplex 
 GEN204R058 
 stop_gained 
 c.445C>T 
 p.Gln149Ter 
 De novo 
  
 Simplex 
 GEN204R059 
 missense_variant 
 c.723T>G 
 p.Phe241Leu 
 De novo 
  
 Simplex 
 GEN204R060 
 missense_variant 
 c.723T>G 
 p.Phe241Leu 
 De novo 
  
 Simplex 
 GEN204R061 
 missense_variant 
 c.737C>T 
 p.Pro246Leu 
 De novo 
  
 Simplex 
 GEN204R062 
 splice_site_variant 
 c.80-1G>A 
  
 Familial 
 Maternal 
  
 GEN204R063 
 missense_variant 
 c.149T>C 
 p.Ile50Thr 
 Familial 
 Maternal 
  
 GEN204R064 
 missense_variant 
 c.397G>A 
 p.Ala133Thr 
 Familial 
 Maternal 
  
 GEN204R065 
 missense_variant 
 c.44G>T 
 p.Arg15Ile 
 De novo 
  
  
 GEN204R066 
 stop_gained 
 c.87T>G 
 p.Tyr29Ter 
 De novo 
  
  
 GEN204R067a 
 missense_variant 
 c.315G>T 
 p.Cys105Phe 
 De novo 
  
  
 GEN204R067b 
 missense_variant 
 c.493G>T 
 p.Lys164Asn 
 Familial 
 Maternal 
 Multi-generational 
 GEN204R068 
 missense_variant 
 c.510G>C 
 p.Ser170Thr 
 De novo 
  
  
 GEN204R069 
 frameshift_variant 
 c.549del 
 p.Lys183ArgfsTer16 
 De novo 
  
  
 GEN204R070 
 missense_variant 
 c.204A>G 
 p.Tyr68Cys 
 De novo 
  
  
 GEN204R071 
 frameshift_variant 
 c.329dup 
 p.Gln110ProfsTer5 
 Familial 
  
  
 GEN204R072 
 missense_variant 
 c.303T>C 
 p.Ile101Thr 
 De novo 
  
  
 GEN204R073 
 stop_gained 
 c.1003C>T 
 p.Arg335Ter 
 De novo 
  
  
 GEN204R074 
 missense_variant 
 c.320A>T 
 p.Lys107Met 
 De novo 
  
 Simplex 
 GEN204R075 
 stop_gained 
 c.176C>G 
 p.Pro59Arg 
 Familial 
 Paternal 
 Multi-generational 
 GEN204R076 
 missense_variant 
 c.389G>A 
 p.Arg130Gln 
 Unknown 
 Not maternal 
  
 GEN204R077 
 splice_site_variant 
 c.634+2T>G 
  
 De novo 
  
  
 GEN204R078 
 inframe_deletion 
 c.-479_-477del 
  
 Unknown 
  
  
 GEN204R079 
 stop_gained 
 c.697C>T 
 p.Arg233Ter 
 De novo 
  
  
 GEN204R080 
 stop_gained 
 c.697C>T 
 p.Arg233Ter 
 Familial 
 Paternal 
  
 GEN204R081 
 2KB_upstream_variant 
  
  
 Familial 
 Maternal 
  
 GEN204R082 
 missense_variant 
 c.302T>C 
 p.Ile101Thr 
 De novo 
  
  
 GEN204R083 
 missense_variant 
 c.510T>A 
 p.Ser170Arg 
 Familial 
 Maternal 
  
 GEN204R084 
 stop_gained 
 c.1003C>T 
 p.Arg335Ter 
 De novo 
  
  
 GEN204R085 
 stop_gained 
 c.1003C>T 
 p.Arg335Ter 
 De novo 
  
  
 GEN204R086 
 missense_variant 
 c.75G>T 
 p.Leu25Phe 
 Unknown 
  
  
 GEN204R087 
 missense_variant 
 c.801G>T 
 p.Lys267Asn 
 De novo 
  
  
 GEN204R088 
 inframe_deletion 
 c.7_9del 
 p.Ala3del 
 De novo 
  
  
 GEN204R089 
 inframe_insertion 
 c.512_513insACA 
 p.Leu171_Pro172insHis 
 De novo 
  
  
 GEN204R090 
 stop_gained 
 c.697C>T 
 p.Arg233Ter 
 Unknown 
  
  
 GEN204R091 
 missense_variant 
 c.389G>C 
 p.Arg130Pro 
 De novo 
  
  
 GEN204R092 
 missense_variant 
 c.737C>T 
 p.Pro246Leu 
 De novo 
  
 Simplex 
 GEN204R093 
 frameshift_variant 
 c.405dup 
 p.Cys136MetfsTer44 
 Familial 
 Paternal 
  
 GEN204R094 
 frameshift_variant 
 c.462dup 
 p.Tyr155LeufsTer25 
 Familial 
 Maternal 
  
 GEN204R095 
 missense_variant 
 c.960T>C 
 p.Leu320Ser 
 De novo 
  
 Simplex 
 GEN204R096 
 missense_variant 
 c.235G>A 
 p.Ala79Thr 
 Familial 
 Maternal 
  
 GEN204R097 
 missense_variant 
 c.302T>C 
 p.Ile101Thr 
 De novo 
  
 Multiplex 
 GEN204R098 
 frameshift_variant 
 c.225_226del 
 p.His75LeufsTer2 
 De novo 
  
 Simplex 
 GEN204R099 
 missense_variant 
 c.102C>T 
 p.Ala34Val 
 Unknown 
  
  
 GEN204R100 
 missense_variant 
 c.38A>G 
 p.Lys13Glu 
 Unknown 
  
 Unknown 
 GEN204R101 
 splice_site_variant 
 NM_001304717.3:c.1011+2T>G 
  
 Unknown 
  
 Simplex 
 GEN204R102 
 missense_variant 
 c.182C>T 
 p.His61Tyr 
 Unknown 
  
 Unknown 
 GEN204R103 
 frameshift_variant 
 c.1013del 
 p.Ser338LeufsTer6 
 De novo 
  
 Simplex 
 GEN204R104 
 missense_variant 
 c.810T>A 
 p.Met270Lys 
 De novo 
  
 Simplex 
 GEN204R105 
 stop_gained 
 c.19G>T 
 p.Glu7Ter 
 De novo 
  
 Simplex 
 GEN204R106 
 frameshift_variant 
 c.548del 
 p.Lys183ArgfsTer16 
 De novo 
  
 Simplex 
 GEN204R107 
 missense_variant 
 c.302T>C 
 p.Ile101Thr 
 De novo 
  
 Simplex 
 GEN204R108 
 missense_variant 
 c.449A>G 
 p.Glu150Gly 
 De novo 
  
 Simplex 
 GEN204R109 
 stop_gained 
 c.249C>A 
 p.Cys83Ter 
 Unknown 
  
  
 GEN204R110 
 splice_site_variant 
 c.209+5G>A 
  
 Unknown 
  
 Unknown 
 GEN204R111 
 missense_variant 
 c.380G>C 
 p.Gly127Ala 
 Unknown 
  
 Unknown 
 GEN204R112 
 stop_gained 
 c.-132C>T 
  
 Unknown 
  
 Unknown 
 GEN204R113 
 stop_gained 
 c.388C>T 
 p.Gln130Ter 
 Unknown 
  
 Unknown 
 GEN204R114 
 missense_variant 
 c.389G>A 
 p.Arg130Gln 
 Unknown 
  
 Unknown 
 GEN204R115 
 missense_variant 
 c.406T>C 
 p.Cys136Arg 
 Unknown 
  
 Unknown 
 GEN204R116 
 missense_variant 
 c.464A>C 
 p.Tyr155Ser 
 Unknown 
  
 Unknown 
 GEN204R117 
 missense_variant 
 c.521A>G 
 p.Tyr174Cys 
 Unknown 
  
 Unknown 
 GEN204R118 
 frameshift_variant 
 c.611del 
 p.Pro204GlnfsTer17 
 Unknown 
  
 Unknown 
 GEN204R119 
 missense_variant 
 c.737C>T 
 p.Pro246Leu 
 Unknown 
  
 Unknown 
 GEN204R120 
 frameshift_variant 
 c.955insA 
 p.Thr319AsnfsTer6 
 Unknown 
  
 Unknown 
 GEN204R121 
 frameshift_variant 
 c.1027del 
 p.Val343Ter 
 Unknown 
  
 Unknown 
 GEN204R122 
 2KB_upstream_variant 
 c.-1034_-1030dupGCCCT 
  
 Unknown 
  
 Unknown 
 GEN204R123 
 frameshift_variant 
 c.27del 
 p.Ser10AlafsTer14 
 Unknown 
  
 Unknown 
 GEN204R124 
 splice_site_variant 
 c.164+1G>A 
  
 Unknown 
  
 Unknown 
 GEN204R125 
 missense_variant 
 c.323T>C 
 p.Leu108Pro 
 Unknown 
  
 Unknown 
 GEN204R126 
 stop_gained 
 c.686C>A 
 p.Ser229Ter 
 Unknown 
  
 Unknown 
 GEN204R127 
 missense_variant 
 c.737C>T 
 p.Pro246Leu 
 Unknown 
  
 Unknown 
 GEN204R128 
 splice_site_variant 
 c.1027-1G>A 
  
 Unknown 
  
 Unknown 
 GEN204R129 
 frameshift_variant 
 c.1110_1111insATAGT 
 p.Asp371IlefsTer47 
 Unknown 
  
 Unknown 
 GEN204R130 
 frameshift_variant 
 c.1176del 
 p.Phe392LeufsTer24 
 Unknown 
  
 Unknown 
 GEN204R131 
 copy_number_loss 
  
  
 Unknown 
  
 Unknown 
 GEN204R132 
 frameshift_variant 
 c.253+1dup 
  
 De novo 
  
 Simplex 
 GEN204R133 
 missense_variant 
 c.611C>T 
 p.Pro204Leu 
 De novo 
  
 Simplex 
 GEN204R134 
 missense_variant 
 c.235G>A 
 p.Ala79Thr 
 Unknown 
  
  
 GEN204R135 
 3_prime_UTR_variant 
 c.*10del 
  
 Unknown 
  
  
 GEN204R136 
 stop_gained 
 c.697C>T 
 p.Arg233Ter 
 De novo 
  
 Simplex 
 GEN204R137 
 frameshift_variant 
 c.525_526dup 
 p.Tyr176CysfsTer8 
 De novo 
  
 Simplex 
 GEN204R138 
 stop_gained 
 c.714C>A 
 p.Tyr238Ter 
 Unknown 
  
  
 GEN204R139 
 stop_gained 
 c.1003C>T 
 p.Arg335Ter 
 Unknown 
  
  
 GEN204R140 
 stop_gained 
 c.697C>T 
 p.Arg233Ter 
 Unknown 
  
  
 GEN204R141 
 missense_variant 
 c.737C>T 
 p.Pro246Leu 
 Unknown 
  
 Unknown 
 GEN204R142 
 stop_gained 
 c.388C>T 
 p.Gln130Ter 
 Unknown 
  
 Unknown 
 GEN204R143 
 inframe_deletion 
 c.54_56del 
 p.Gly19del 
 De novo 
  
 Multiplex 
 GEN204R144 
 missense_variant 
 c.97A>G 
 p.Asn33Asp 
 De novo 
  
 Simplex 
 GEN204R145 
 missense_variant 
 c.220A>G 
 p.Arg74Gly 
 De novo 
  
 Simplex 
 GEN204R146 
 missense_variant 
 c.302T>C 
 p.Ile101Thr 
 De novo 
  
 Simplex 
 GEN204R147 
 missense_variant 
 c.320A>T 
 p.Lys107Met 
 De novo 
  
 Simplex 
 GEN204R148 
 splice_site_variant 
 c.493-2A>G 
  
 De novo 
  
 Simplex 
 GEN204R149 
 missense_variant 
 c.723T>G 
 p.Phe241Leu 
 De novo 
  
 Simplex 
 GEN204R150 
 missense_variant 
 c.23T>A 
 p.Ile8Asn 
 De novo 
  
  
 GEN204R151 
 splice_site_variant 
 c.165-2A>G 
  
 De novo 
  
  
 GEN204R152 
 stop_gained 
 c.388C>T 
 p.Gln130Ter 
 De novo 
  
  
 GEN204R153 
 missense_variant 
 c.403A>C 
 p.Ile135Leu 
 De novo 
  
  
 GEN204R154 
 missense_variant 
 c.424C>T 
 p.Arg142Trp 
 De novo 
  
  
 GEN204R155 
 missense_variant 
 c.479C>T 
 p.Pro160Leu 
 De novo 
  
  
 GEN204R156 
 missense_variant 
 c.626G>T 
 p.Gly209Val 
 De novo 
  
  
 GEN204R157 
 splice_site_variant 
 c.1027-2A>G 
  
 De novo 
  
 Multiplex 
 GEN204R158 
 missense_variant 
 c.149T>C 
 p.Ile50Thr 
 De novo 
  
  
 GEN204R159 
 splice_site_variant 
 c.253+1G>A 
  
 De novo 
  
  
 GEN204R160 
 stop_gained 
 c.259C>T 
 p.Gln87Ter 
 De novo 
  
  
 GEN204R161 
 frameshift_variant 
 c.502_503del 
 p.Ser168ProfsTer15 
 De novo 
  
  
 GEN204R162 
 missense_variant 
 c.514A>G 
 p.Arg172Gly 
 De novo 
  
  
 GEN204R163 
 missense_variant 
 c.820T>G 
 p.Trp274Gly 
 De novo 
  
  
 GEN204R164 
 missense_variant 
 c.752G>A 
 p.Gly251Asp 
 Familial 
 Maternal 
 Simplex 
 GEN204R165 
 missense_variant 
 c.77C>T 
 p.Thr26Ile 
 De novo 
  
  
 GEN204R166 
 stop_gained 
 c.1008C>G 
 p.Tyr336Ter 
 Familial 
 Maternal 
  
 GEN204R167 
 frameshift_variant 
 c.27dup 
 p.Ser10Ter 
 De novo 
  
  
 GEN204R168 
 missense_variant 
 c.403A>C 
 p.Ile135Leu 
 Unknown 
  
  
 GEN204R169 
 stop_gained 
 c.388C>T 
 p.Arg130Ter 
 De novo 
  
  
 GEN204R170 
 missense_variant 
 c.518G>T 
 p.Arg173Leu 
 De novo 
  
 Simplex 
 GEN204R171 
 missense_variant 
 c.466G>A 
 p.Gly156Arg 
 De novo 
  
 Simplex 
 GEN204R172 
 missense_variant 
 c.389G>A 
 p.Arg130Gln 
 Unknown 
  
 Simplex 
 GEN204R173 
 missense_variant 
 c.821G>T 
 p.Trp274Leu 
 Unknown 
  
 Simplex 
 GEN204R174 
 frameshift_variant 
 c.361_362dup 
 p.Ser121ArgfsTer55 
 De novo 
  
 Simplex 
  et al.  
 GEN204R175 
 5_prime_UTR_variant 
 c.-835C>T 
  
 Unknown 
  
  
  et al.  
 GEN204R176 
 missense_variant 
 c.278A>G 
 p.His93Arg 
 Unknown 
  
  
  et al.  

Common

No Common Variants Available
Chromosome
CNV Locus
CNV Type
# of studies
Animal Model
10
Duplication
 1
 
10
Deletion
 2
 
10
Deletion
 1
 
10
Deletion
 8
 

Model Summary

Mutant mice display abnormal social interaction and exaggerated responses to sensory stimuli.

References

Type
Title
Author, Year
Primary
Pten regulates neuronal arborization and social interaction in mice.
Primary
Pharmacological inhibition of mTORC1 suppresses anatomical, cellular, and behavioral abnormalities in neural-specific Pten knock-out mice.
Additional
Allele-specific tumor spectrum in pten knockin mice.
Additional
Mitochondrial dysfunction in Pten haplo-insufficient mice with social deficits and repetitive behavior: interplay between Pten and p53.
Additional
Dysregulation of synaptic plasticity precedes appearance of morphological defects in a Pten conditional knockout mouse model of autism.
Additional
Pten haploinsufficient mice show broad brain overgrowth but selective impairments in autism-relevant behavioral tests.
Additional
Molecular and phenotypic abnormalities in individuals with germline heterozygous PTEN mutations and autism.
Additional
Decreased aggression and increased repetitive behavior in Pten haploinsufficient mice.
Additional
Hyperactivity of newborn Pten knock-out neurons results from increased excitatory synaptic drive.
Additional
The parvalbumin/somatostatin ratio is increased in Pten mutant mice and by human PTEN ASD alleles.
Additional
Autistic-Like Traits and Cerebellar Dysfunction in Purkinje Cell PTEN Knock-Out Mice.
Additional
Superimposing Status Epilepticus on Neuron Subset-Specific PTEN Haploinsufficient and Wild Type Mice Results in Long-term Changes in Behavior.
Additional
Intermittent fasting uncovers and rescues cognitive phenotypes in PTEN neuronal haploinsufficient mice.
Additional
PTEN Activity Defines an Axis for Plasticity at Cortico-Amygdala Synapses and Influences Social Behavior.
Additional
Therapeutic inhibition of mTORC2 rescues the behavioral and neurophysiological abnormalities associated with Pten-deficiency.
Additional
Interneuron Transplantation Rescues Social Behavior Deficits Without Restoring Wild-Type Physiology in a Mouse Model of Autism With Excessive Synaptic Inhibition
Additional
Elevated protein synthesis in microglia causes autism-like synaptic and behavioral aberrations
Additional
Pten haploinsufficiency causes desynchronized growth of brain areas involved in sensory processing
Additional
Disruption of mTORC1 rescues neuronal overgrowth and synapse function dysregulated by Pten loss

M_PTEN_1_CKO_HM

Model Type: Genetic
Model Genotype: Homozygous
Mutation: Conditional deletion of exons 4-5 of the Pten gene using Nse-cre, authors in PMID 23487788 (Takeuchi K et al 2013) specifically show loss of Pten in granule cells of dentate gyrus and pyramidal neurons of hippocampal CA3 and a subset of postmitotic neruons in the cortex. Same CKOs produced in PMID:16675393
Allele Type: Conditional loss-of-function
Strain of Origin: C57BL/6
Genetic Background: C57BL/6
ES Cell Line: E14K
Mutant ES Cell Line: Not Specified
Model Source: Not Specified

M_PTEN_19_KO_HM

Model Type: Genetic
Model Genotype: Homozygous
Mutation: Exon 4 and 5 are floxed by introducing loxP sites in introns 3 and 5, homozygous mice are obtained by breeding heterozygous knockouts following germline transmission of exon4-5 deletion.
Allele Type:
Strain of Origin:
Genetic Background: FVB/N *129Sv/Ev* Black swiss
ES Cell Line:
Mutant ES Cell Line:
Model Source: PMID: 20194734

M_PTEN_2_CKO_HT

Model Type: Genetic
Model Genotype: Heterozygous
Mutation: Conditional heterozygous deletion of exon 5 of the Pten gene , that encodes the phosphatase domain, using Nse-cre (or Enolas2-cre) in neurons of the cortex, hippocampus and cerebellum
Allele Type: Conditional loss-of-function
Strain of Origin: BALB/c
Genetic Background: C57BL/6
ES Cell Line: 129S4/SvJae
Mutant ES Cell Line: Not Specified
Model Source: Not specified

M_PTEN_3_CKO_HM

Model Type: Genetic
Model Genotype: Homozygous
Mutation: Conditional deletion of exons 4-5 of the Pten gene using Nse-cre, authors in PMID 23487788 (Takeuchi K et al 2013) specifically show loss of Pten in granule cells of dentate gyrus and pyramidal neurons of hippocampal CA3 and a subset of postmitotic neruons in the cortex. Same CKOs produced in PMID:16675393
Allele Type: Conditional (knockout)
Strain of Origin: Not specified
Genetic Background: C57BL/6
ES Cell Line: Not specified
Mutant ES Cell Line: Not Specified
Model Source: Not Specified

M_PTEN_6_KO_HT

Model Type: Genetic
Model Genotype: Heterozygous
Mutation: Deletion of gene sequence from BglII site in exon 5 to the next BglII site 2.0 kb downstream.
Allele Type: Targeted (knockout)
Strain of Origin: 129Sv/J
Genetic Background: C57BL/6
ES Cell Line: Not Specified
Mutant ES Cell Line: Not Specified
Model Source: Not Specified

M_PTEN_7_CKO_HM

Model Type: Genetic
Model Genotype: Homozygous
Mutation: Conditional deletion of exon 5 of the Pten gene using endogenous Slc6a3 (Dopamine transporter, DAT)-cre, in dopaminergic neurons
Allele Type: Conditional loss-of-function
Strain of Origin: C57BL/6
Genetic Background: C57BL/6
ES Cell Line: Not Specified
Mutant ES Cell Line: Not Specified
Model Source: Not Specified

M_PTEN_4_KI_M3M4_HM

Model Type: Genetic
Model Genotype: Homozygous
Mutation: M3M4 missense double mutations were targeted into the Pten exon 7, which contains the NLS-like sequence.
Allele Type: Targeted (knockin)
Strain of Origin: CD1
Genetic Background: CD1
ES Cell Line: Not Specified
Mutant ES Cell Line: Not Specified
Model Source: Not Specified

M_PTEN_5_KI_M3M4_HT

Model Type: Genetic
Model Genotype: Heterozygous
Mutation: M3M4 missense double mutations were targeted into the Pten exon 7, which contains the NLS-like sequence.
Allele Type: Targeted (knockin)
Strain of Origin: CD1
Genetic Background: CD1
ES Cell Line: Not Specified
Mutant ES Cell Line: Not Specified
Model Source: Not Specified

M_PTEN_8_CKO_HM_DG

Model Type: Genetic
Model Genotype: Homozygous
Mutation: Conditional deletion of exon 5 of the Pten gene using a T2A-cre containing retrovirus injected bilaterally into dentate gyrus at postnatal day 7
Allele Type: Conditional loss-of-function
Strain of Origin: C57BL/6
Genetic Background: C57BL/6
ES Cell Line: Not Specified
Mutant ES Cell Line: Not Specified
Model Source: Not Specified

M_PTEN_10_CKO_HM

Model Type: Genetic
Model Genotype: Homozygous
Mutation: Conditional deletion of exons 4-5 of of PTEN using Dlxl12b-cre mice, in gabaergic interneurons of the forebrain, by E15.5. Authors report assessments looking into the secondary progenitor cells of the subventricular zone of the subpallium
Allele Type: Conditional loss-of-function
Strain of Origin: CD1* C56BL/6
Genetic Background: CD1
ES Cell Line:
Mutant ES Cell Line:
Model Source:

M_PTEN_10_CKO_HT

Model Type: Genetic
Model Genotype: Heterozygous
Mutation: Conditional heterozygous deletion of exons 4-5 of of PTEN using Dlxl12b-cre mice, in gabaergic neurons of the forebrain, by E15.5. Authors report assessments looking into the secondary progenitor cells of the subventricular zone of the subpallium
Allele Type: Conditional loss-of-function
Strain of Origin: CD1* C56BL/6
Genetic Background: CD1
ES Cell Line:
Mutant ES Cell Line:
Model Source:

M_PTEN_11_CKO_HM_SSTN

Model Type: Genetic
Model Genotype: Homozygous
Mutation: Conditional deletion of exons 4 - 5 using SST-IRES-Cre, in postmitotic somatostatin expressing cells as they leave the medial ganglionic eminence. The tdtomato reporter under Ai14 Flox/+ line was used to follow expression of the cre recombinase.
Allele Type: Conditional loss-of-function
Strain of Origin: CD1* C56BL/6
Genetic Background: CD1* C56BL/6
ES Cell Line:
Mutant ES Cell Line:
Model Source:

M_PTEN_11_CKO_HT_SSTN

Model Type: Genetic
Model Genotype: Heterozygous
Mutation: Conditional heterozygous deletion of exons 4 - 5 using SST-IRES-Cre, in postmitotic somatostatin expressing cells as they leave the medial ganglionic eminence. The tdtomato reporter under Ai14 Flox/+ line was used to follow expression of the cre recombinase.
Allele Type: Conditional loss-of-function
Strain of Origin: CD1* C56BL/6
Genetic Background: CD1
ES Cell Line:
Mutant ES Cell Line:
Model Source:

M_PTEN_12_CKO_HM

Model Type: Genetic
Model Genotype: Homozygous
Mutation: In this cell transplant based model, cells from with a homozygous loss of Pten using Dlxl12b-Cre+ were extracted from E12.5 embryos, then transplanted to wild type recipient mice at P1 to see how complete loss of Pten from the secondary progenitors in the subventricular zone (SVZ) of the entire subpallium, affects cell differentiation. The recipient mouse neocortices were then examined 35 days following transplantation.
Allele Type: Conditional loss-of-function
Strain of Origin: CD1* C56BL/6
Genetic Background: CD1
ES Cell Line:
Mutant ES Cell Line:
Model Source:

M_PTEN_12_CKO_HT

Model Type: Genetic
Model Genotype: Heterozygous
Mutation: In this cell transplant based model, cells from with a heterozygous loss of Pten using Dlxl12b-Cre+ were extracted from E12.5 embryos, then transplanted to wild type recipient mice at P1 to see howpartial loss of Pten from the secondary progenitors in the subventricular zone (SVZ) of the entire subpallium, affects cell differentiation. he recipient mouse neocortices were then examined 35 days following transplantation.
Allele Type: Conditional loss-of-function
Strain of Origin: CD1* C56BL/6
Genetic Background: CD1
ES Cell Line:
Mutant ES Cell Line:
Model Source:

M_PTEN_9_CKO_HM

Model Type: Genetic
Model Genotype: Homozygous
Mutation: Conditional deletion of exons 4-5 of the PTEN gene using Nkx2.1-Cre, in the ventricular zone of the medical ganglionic eminence (MGE) that gives rise to interneurons starting E9.5. The tdtomato reporter under Ai14 Flox/+ line was used to follow expression of the cre recombinase.
Allele Type: Conditional loss-of-function
Strain of Origin: CD1* C56BL/6
Genetic Background: CD1
ES Cell Line:
Mutant ES Cell Line:
Model Source:

M_PTEN_9_CKO_HT

Model Type: Genetic
Model Genotype: Heterozygous
Mutation: Conditional heterozygous deletion of exons 4-5 of the PTEN gene using Nkx2.1-Cre in the ventricular zone of the medical ganglionic eminence (MGE) that gives rise to interneurons starting E9.5. The tdtomato reporter under Ai14 Flox/+ line was used to follow expression of the cre recombinase.
Allele Type: Conditional loss-of-function
Strain of Origin: CD1* C56BL/6
Genetic Background: CD1
ES Cell Line:
Mutant ES Cell Line:
Model Source:

M_PTEN_17_CKO_HM_PURKINJEN

Model Type: Genetic
Model Genotype: Homozygous
Mutation: Conditional deletion of exon 5 of the Pten gene using L7-Cre in Purkinje cells of the cerebellum
Allele Type: Conditional loss-of-function
Strain of Origin:
Genetic Background: C57BL/6
ES Cell Line:
Mutant ES Cell Line:
Model Source:

M_PTEN_18_CKO_HT_SEIZURE

Model Type: Genetic
Model Genotype: Heterozygous
Mutation: Multifactorial model, Pten CKO mice, with deletion of exons 4 and 5 using GFAP-cre, authors indicate that the is KO localized to the hippocampal granule cells of the dentate gyrus, by P5. These PTEN het CKO mice were treated with 20mg/kg (in 0.9% saline) of kainate (ip) beginning at P60 (2 months age). At approximately 1 hour following saline (control) or kainate injections, all mice received an ip injection of pentobarbital (20mg/kg) to terminate seizure activity. All behavioral testing was carried out 1 week after seizure induction, only mice that had entered the status epilepticus so that they all fell into the category of highest seizure severity were used in the studies.
Allele Type: Multifactorial
Strain of Origin: 129P2/OlaHsd
Genetic Background: FVB*mixed
ES Cell Line: E14K
Mutant ES Cell Line:
Model Source:

M_PTEN_13_KI_HM

Model Type: Genetic
Model Genotype: Heterozygous
Mutation: Partial loss-of-function knockin mice with truncated Pten gene lacking its C-terminal PDZ motif, Pten^tm(Q399stop)amc, were generated by homologous recombination in iTL1 129S6/SvEvBrdTac(129Sv)-derived embryonic stem cells where residues 399 to 403 were truncated. The PDZ-binding domain was deleted by substituting codon 399 (CAA) with a stop codon (TAA), as previously described (Knafo et al. 2016, PMID 26780512). These mice express normal levels of total Pten protein and normal Pten catalytic activity.
Allele Type: LOF Knockin
Strain of Origin: 129S6/SvEvBrdTac(129Sv)
Genetic Background: C57BL/6
ES Cell Line: 129S6/SvEvBrdTac(129Sv)
Mutant ES Cell Line:
Model Source:

M_PTEN_14_TG

Model Type: Genetic
Model Genotype: Heterozygous
Mutation: Gain-of-function mutant mice with increased Pten copy number (Pten^tg) that overexpress Pten protein were generated by bacterial artificial chromosome (BAC) transgenesis on a C57BL6/CBA genetic background as previously described in Ortega-Molina et al. 2012, PMID 22405073 (gift from the lab of Manuel Serrano, IRB, Barcelona, Spain).
Allele Type: Overexpression
Strain of Origin:
Genetic Background: C57BL6*CBA
ES Cell Line:
Mutant ES Cell Line:
Model Source:

M_PTEN_20_CKO_HM

Model Type: Genetic
Model Genotype: Homozygous
Mutation: Mice with loxp sites flanking exons 4 and 5 in both alleles of Pten were crossed with Camk2a-Cre heterozygous mice to generate mice with Pten deleted in selectively in the forebrain; M_PTEN_1_CKO_HM; M_PTEN_10_CKO_HM; M_PTEN_12_CKO_HM; M_PTEN_9_CKO_HM
Allele Type: Conditional knockout
Strain of Origin:
Genetic Background: C57BL/6
ES Cell Line:
Mutant ES Cell Line:
Model Source: Kwon et al, Nat Gen, 2001

M_PTEN_21_CKO_HM

Model Type: Genetic
Model Genotype: Homozygous
Mutation: At P7, Pten^flx/flx animals were co-injected into the dentate gyrus with a retrovirus encoding a fluorophore (mCherry) with a downstream Cre, and a control retrovirus with just a fluorophore (GFP) and no Cre.
Allele Type: Conditional Knockout
Strain of Origin: 129S4/SvJae
Genetic Background: C57BL/6J
ES Cell Line: LW1
Mutant ES Cell Line:
Model Source: Hong Wu Lab

M_PTEN_22_CKO_HM

Model Type: Genetic
Model Genotype: Homozygous
Mutation: At P7, Pten^flx/flx animals were co-injected into the dentate gyrus with a retrovirus encoding a fluorophore (GFP) with a downstream Cre, and a control retrovirus with just a fluorophore (mCherry) and no Cre.
Allele Type: Conditional Knockout
Strain of Origin: 129S4/SvJae
Genetic Background: C57BL/6J
ES Cell Line: LW1
Mutant ES Cell Line:
Model Source: Hong Wu Lab

M_PTEN_23_CKO_HM

Model Type: Genetic
Model Genotype: Homozygous
Mutation: Conditional knockout mice carrying an inducible microglia-specific Pten deletion were generated by crossing Pten^flox/flox mice (MGI:2156086) with Cx3cr1^CreERT2/+ mice (MGI:5450813). The Pten-flox allele contains loxP sites flanking exon 5 of the Pten gene. The knock-in Cx3cr1-CreERT2 allele replaces exon 2 of the microglia-specific gene Cx3cr1 with a tamoxifen-inducible Cre gene (Cre recombinase fused to an estrogen receptor ligand binding domain). The knock-in Cx3cr1-CreERT2 allele also expresses EYFP. The genotype of the experimental mice is Pten^flox/flox;Cx3cr1^CreERT2/+. The genotype of control mice is Cx3cr1^CreERT2/+. Pten deletion in microglia was induced at postnatal days 0-2 (P0, P1, P2) by injection of 50 micrograms of tamoxifen in sunflower oil into the stomach.
Allele Type: Conditional knockout
Strain of Origin: 129S4/SvJae; 129P2/OlaHsd
Genetic Background:
ES Cell Line: LW1; E14.1
Mutant ES Cell Line:
Model Source: Jackson Laboratory

M_PTEN_1_CKO_HM

Category
Entity
Quantity
Experimental Paradigm
Age at Testing
General locomotor activity1
Abnormal
Description: Abnormal locomotor activity; hyperactive in light boxes, normal in dark boxes
Exp Paradigm: Open field test
 Open field test
 6 weeks
Dendritic architecture: spine density1
Abnormal
Description: Dendritic hypertrophy, ectopy, and increased spine density in cerebral cortex and dentate gyri
Exp Paradigm: Golgi staining; immunohistochemistry with antibodies to dendritic markers; electron microscopic analysis of cortex and dentate gyri
 Immunohistochemistry
 3-9 months
Brain morphology1
Abnormal
Description: Progressive macrocephaly in forebrain; foliation of dg and compression of ca1; increased neuronal size with soma hypertrophy
Exp Paradigm: General observation; hematoxylin/eoslin staining on coronal sections
 Histology
 Unreported
Brain morphology2
Abnormal
Description: Abnormal brain morphology indicated by enlarged dgc and hypertrophied mossy fiber projection axons
Exp Paradigm: H&e staining
 Histology
 5 months
Synapse density1
Abnormal
Description: Hypertrophic and ectopic axonal tract with increased synapses in dentate gyrus
Exp Paradigm: Immunohistochemistry with antibodies to synapsin (presynaptic marker) and calbindin (expressed in soma and processes); electron microscopic analysis of the inner molecular layer of dentate gyri
 Immunohistochemistry
 10.7 months
Synaptic transmission2
Increased
Description: Increased basal synaptic transmission as measured with input/output relation at dgc synapse
Exp Paradigm: Whole cell recordings in the medial prefrontal pathway
 Whole-cell patch clamp
 3.5-5 months
Synaptic plasticity2
Decreased
Description: Decreased synaptic plasticity as assessed by impaired tbs-ltp at dgc synapses
Exp Paradigm: Whole cell recordings in the medial prefrontal pathway
 Whole-cell patch clamp
 3.5-7.5 months
Synaptic plasticity2
Increased
Description: Increased synaptic plasticity as assessed by tbs-ltp at dgc synapse
Exp Paradigm: Whole cell recordings in the medial prefrontal pathway
 Whole-cell patch clamp
 8-12 weeks
Presynaptic function: paired-pulse facilitation2
Decreased
Description: Decreased presynaptic function as measured by paired pulse ratio (ppr) at dgc synapse
Exp Paradigm: Whole cell recordings in the medial prefrontal pathway
 Whole-cell patch clamp
 3.5-5 months
Synaptic plasticity2
Decreased
Description: Decreased synaptic plasticity as assessed through tbs-ltp and hfs-ltp at ca3-ca1 synapses
Exp Paradigm: Whole cell recordings at ca3-ca1 synapses
 Whole-cell patch clamp
 4-5months
Synaptic plasticity2
Decreased
Description: Decreased synaptic plasticity as assessed through mglur-ltd at dgc synapses
Exp Paradigm: Whole cell recordings in the medial prefrontal pathway
 Whole-cell patch clamp
 2-7.5 months
Seizures1
Abnormal
Description: Sporadic seizure propensity during light phase; repetitive spike-wave patterns with continous spike-wave bursting
Exp Paradigm: Subjective observation; analysis of eeg/emg recordings of mice-observation of seizures
 Observation of seizures
 3-9 months
Seizures1
Abnormal
Description: Sporadic seizure propensity during light phase; repetitive spike-wave patterns with continous spike-wave bursting
Exp Paradigm: Subjective observation; analysis of eeg/emg recordings of mice- electroencephalogram/electromyogram (eeg/emg)
 Electroencephalogram/electromyogram (eeg/emg)
 3-9 months
Sensorimotor gating1
Abnormal
Description: Significantly impaired startle response
Exp Paradigm: Prepulse inhibition of the acoustic startle response as a measure of sensorimotor gating.
 Prepulse inhibition
 6 weeks
Startle response: acoustic stimulus1
Increased
Description: Increased startle reflex with habituation to repeated stimuli
Exp Paradigm: Response to 120 db white noise stimulus
 Acoustic startle reflex test
 6 weeks
Social interaction1
Decreased
Description: Decreased social interaction
Exp Paradigm: Direct contacts between genotypically identical test
 Reciprocal social interaction test
 Unreported
Social approach1
Decreased
Description: Decreased social approach
Exp Paradigm: Social preference test in tripartite chamber
 Three-chamber social approach test
 Unreported
Social transmission of food preference1
Decreased
Description: Decreased social learning
Exp Paradigm: Direct contacts between genotypically identical test
 Reciprocal social interaction test
 Unreported
Nest building behavior1
Decreased
Description: Decreased nest building activity
Exp Paradigm: Nest forming test for home cage behavior
 Nest building assay
 Unreported
Social interaction1
Decreased
Description: Decreased social interaction
Exp Paradigm: Social interaction between social target and empty cage in open field
 Reciprocal social interaction test
 Unreported
Anxiety1
Increased
Description: Increased anxiety-like behavior
Exp Paradigm: Open field test; dark/light apparatus-open field test
 Open field test
 Unreported
Anxiety1
Increased
Description: Increased anxiety-like behavior
Exp Paradigm: Open field test; dark/light apparatus- light-dark exploration test
 Light-dark exploration test
 Unreported
Spatial reference memory1
Decreased
Description: Decreased rate of acquisition of spatial reference memory
Exp Paradigm: Morris water maze task
 Morris water maze test
 Unreported
Nursing/lactation1
Abnormal
Description: Abnormal maternal nursing
Exp Paradigm: Pup survival rate
 Survival analysis
 Unreported
Protein modification process1
Increased
Description: Increased phosphorylation of downstream targets of pten such as p-akt, p-s6, p-gsk3, tuberin
Exp Paradigm: Downstream targets phosphorylation
 Immunohistochemistry
 2 months
Protein expression level evidence2
Decreased
Description: Decreased expression of pten in dgc and ca3 pyramidal cells
Exp Paradigm: Pten protein expression
 Immunohistochemistry
 8 weeks
General characteristics1
 No change
 General observations
 4-5 weeks
Anxiety1
 No change
 Elevated plus maze test
 Unreported
Cued or contextual fear conditioning: memory of cue1
 No change
 Fear conditioning test
 Unreported
Motor coordination and balance1
 No change
 Accelerating rotarod test
 Unreported
Motor strength and endurance1
 No change
 Dowel walk test
 Unreported
Motor strength and endurance1
 No change
 Vertical pole test
 Unreported
Presynaptic function: paired-pulse facilitation2
 No change
 Whole-cell patch clamp
 8-30 weeks
Synaptic plasticity2
 No change
 Whole-cell patch clamp
 8-20 weeks
Synaptic plasticity2
 No change
 Whole-cell patch clamp
 8-12 weeks
Synaptic transmission2
 No change
 Whole-cell patch clamp
 8-20 weeks
Synaptic transmission2
 No change
 Whole-cell patch clamp
 8-30 weeks
Olfaction1
 No change
 Buried food test
 Unreported
Inanimate object preference1
 No change
 Three-chamber social approach test
 Unreported
 Not Reported: Circadian sleep/wake cycle, Communications, Developmental profile, Emotion, Immune response, Learning & memory, Maternal behavior, Motor phenotype, Neurophysiology, Physiological parameters, Repetitive behavior, Seizure, Sensory, Social behavior

M_PTEN_19_KO_HM

Category
Entity
Quantity
Experimental Paradigm
Age at Testing
Mortality/lethality: embryonic1
Increased
Description: No live pten del 4-5 homozygous mutants are recovered from heterozygous crosses
Exp Paradigm: NA
 Survival analysis
 E9.5
 Not Reported: Circadian sleep/wake cycle, Communications, Emotion, Immune response, Learning & memory, Maternal behavior, Molecular profile, Motor phenotype, Neuroanatomy / ultrastructure / cytoarchitecture, Neurophysiology, Physiological parameters, Repetitive behavior, Seizure, Sensory, Social behavior

M_PTEN_2_CKO_HT

Category
Entity
Quantity
Experimental Paradigm
Age at Testing
Brain size1
Increased
Description: Increased brain weight (normalized to body weight)
Exp Paradigm: NA
 Measurement of tissue weight
 5-7 months
Brain size2
Increased
Description: Mutants show an increase in total brain and cortical mass compared to controls.
Exp Paradigm: NA
 Measurement of tissue weight
 5-6 months
Brain size1
Increased
Description: Increased enlargement of cortex and hippocampus over cerebellum
Exp Paradigm: NA
 Measurement of tissue weight
 5-7 months
Self grooming: perseveration1
Increased
Description: Increased self grooming indicated by twice the frequency of self grooming
Exp Paradigm: Observation of self-grooming activities
 Grooming behavior assessments
 5-7 months
Social interaction1
Decreased
Description: Decreased social novelty as demonstrated by lower novel to total side time ratio and lower time spent sniffing novel mouse normalized by total sniff time
Exp Paradigm: Social novelty preference trial
 Three-chamber social approach test
 5-7 months
Social interaction1
Decreased
Description: Decreased sociability demonstrated by decreased time spent with social target with same number of entries towards social side and decreased time spent sniffing social target with no change in sniff frequency
Exp Paradigm: Socialization trial with measures of number of entires and time spent in each side of box, and time spent sniffing each wire cage
 Three-chamber social approach test
 5-7 months
Oxidative stress: systemic1
Increased
Description: Increased nitrative/oxidative stress indicated by increased nitrated tyr in atpase beta-subunit
Exp Paradigm: Mitochondrial enzymaic and complex activity measurement from isolated mitochondria from hippocampus and cerebellum
 Mitochondrial respiratory chain enzyme analysis
 5-6 months
Cued or contextual fear conditioning: passive avoidance2
Decreased
Description: Mutants show no change in the latency to move to the dark chamber compared to controls that show increase in latency, in the probe stage. mutants show no change in latency to move to the dark chamber in the exposure stage compared to controls.
Exp Paradigm: NA
 Passive avoidance test
 5-6 months
Protein expression level evidence1
Increased
Description: Increased levels of glial fibrillary acidic protein
Exp Paradigm: Gfap protein levels
 Western blot
 5-7 months
Enzyme activity1
Decreased
Description: Decreased cytochrome c oxidase activity in cerebellum and hippocampus
Exp Paradigm: Cytochrome c oxidase activity
 Mitochondrial respiratory chain enzyme analysis
 5-7 months
Targeted expression2
Decreased
Description: Mutants show lower pten protein levels compared to controls in the cerebral cortex.
Exp Paradigm: NA
 Western blot
 5-6 months
Protein expression level evidence1
Decreased
Description: Decreased p53 protein expression in cerebellum
Exp Paradigm: P53 protein expression
 Western blot
 5-7 months
Signaling2
Increased
Description: Mutants show increase in phosphorylated akt (p-akt^t308) compared to controls.
Exp Paradigm: NA
 Western blot
 5-6 months
Protein expression level evidence1
Decreased
Description: Decreased levels of pten protein expression in hippocampus and cerebellum
Exp Paradigm: Pten protein expression
 Western blot
 4-29 weeks
Signaling1
Decreased
Description: Decreased levels of p21, sco2 in cerebellum and hippocampus
Exp Paradigm: P53 downstream effectors levels
 Western blot
 5-7 months
Mitochondrial dna replication1
Increased
Description: Increased mtdna copy number in cerebellum
Exp Paradigm: Mtdna copy number
 Quantitative pcr (qrt-pcr)
 8-13 weeks
Size/growth2
 No change
 Body weight measurement
 5-6 months
Anxiety2
 No change
 Elevated plus maze test
 5-6 months
Anxiety2
 No change
 Open field test
 5-6 months
Cognitive flexibility: associative learning2
 No change
 Morris water maze test
 5-6 months
Object recognition memory2
 No change
 Novel object recognition test
 5-6 months
Spatial reference memory2
 No change
 Morris water maze test
 5-6 months
Spatial working memory2
 No change
 Morris water maze test
 5-6 months
Enzyme activity1
 No change
 Mitochondrial respiratory chain enzyme analysis
 4-29 weeks
Mitochondrial biogenesis1
 No change
 Mitochondrial respiratory chain enzyme analysis
 4-6 weeks
Mitochondrial biogenesis1
 No change
 Mitochondrial respiratory chain enzyme analysis
 8-13 weeks
Protein expression level evidence1
 No change
 Western blot
 8-13 weeks
Protein expression level evidence1
 No change
 Western blot
 4-29 weeks
Protein expression level evidence1
 No change
 Western blot
 4-6 weeks
Protein expression level evidence1
 No change
 Western blot
 4-29 weeks
Protein localization: synapse2
 No change
 Western blot
 5-6 months
Signaling2
 No change
 Western blot
 5-6 months
General locomotor activity2
 No change
 Open field test
 5-6 months
General locomotor activity: ambulatory activity2
 No change
 Elevated plus maze test
 5-6 months
General locomotor activity: ambulatory activity2
 No change
 Open field test
 5-6 months
Swimming ability2
 No change
 Morris water maze test
 5-6 months
Cerebellar morphology2
 No change
 Measurement of tissue weight
 5-6 months
Hippocampal morphology2
 No change
 Measurement of tissue weight
 5-6 months
Neuroreceptor levels: glutamate receptors: ampa receptors2
 No change
 Western blot
 5-6 months
Neuroreceptor levels: glutamate receptors: nmda receptors2
 No change
 Western blot
 5-6 months
Satiety response2
 No change
 Food intake measurements
 5-6 months
Self grooming: perseveration1
 No change
 Grooming behavior assessments
 8-13 weeks
Startle response2
 No change
 Open field test
 5-6 months
Social interaction1
 No change
 Three-chamber social approach test
 8-13 weeks
Social interaction1
 No change
 Three-chamber social approach test
 8-13 weeks
 Not Reported: Circadian sleep/wake cycle, Communications, Developmental profile, Emotion, Immune response, Learning & memory, Maternal behavior, Motor phenotype, Neurophysiology, Physiological parameters, Repetitive behavior, Seizure, Sensory, Social behavior

M_PTEN_3_CKO_HM

Category
Entity
Quantity
Experimental Paradigm
Age at Testing
Brain morphology1
Abnormal
Description: Abnormal brain morphology indicated by enlarged dgc and hypertrophied mossy fiber projection axons
Exp Paradigm: H&e staining
 Histology
 5 months
Synaptic plasticity1
Decreased
Description: Decreased synaptic plasticity as assessed through tbs-ltp and hfs-ltp at ca3-ca1 synapses
Exp Paradigm: Whole cell recordings at ca3-ca1 synapses
 Whole-cell patch clamp
 4-5months
Synaptic plasticity1
Decreased
Description: Decreased synaptic plasticity as assessed through mglur-ltd at dgc synapses
Exp Paradigm: Whole cell recordings in the medial prefrontal pathway
 Whole-cell patch clamp
 2-7.5 months
Synaptic transmission1
Increased
Description: Increased basal synaptic transmission as measured with input/output relation at dgc synapse
Exp Paradigm: Whole cell recordings in the medial prefrontal pathway
 Whole-cell patch clamp
 3.5-5 months
Synaptic plasticity1
Decreased
Description: Decreased synaptic plasticity as assessed by impaired tbs-ltp at dgc synapses
Exp Paradigm: Whole cell recordings in the medial prefrontal pathway
 Whole-cell patch clamp
 3.5-7.5 months
Synaptic plasticity1
Increased
Description: Increased synaptic plasticity as assessed by tbs-ltp at dgc synapse
Exp Paradigm: Whole cell recordings in the medial prefrontal pathway
 Whole-cell patch clamp
 8-12 weeks
Presynaptic function: paired-pulse facilitation1
Decreased
Description: Decreased presynaptic function as measured by paired pulse ratio (ppr) at dgc synapse
Exp Paradigm: Whole cell recordings in the medial prefrontal pathway
 Whole-cell patch clamp
 3.5-5 months
Protein expression level evidence1
Decreased
Description: Decreased expression of pten in dgc and ca3 pyramidal cells
Exp Paradigm: Pten protein expression
 Immunohistochemistry
 8 weeks
Presynaptic function: paired-pulse facilitation1
 No change
 Whole-cell patch clamp
 8-30 weeks
Synaptic plasticity1
 No change
 Whole-cell patch clamp
 8-20 weeks
Synaptic plasticity1
 No change
 Whole-cell patch clamp
 8-12 weeks
Synaptic transmission1
 No change
 Whole-cell patch clamp
 8-20 weeks
Synaptic transmission1
 No change
 Whole-cell patch clamp
 8-30 weeks
 Not Reported:

M_PTEN_6_KO_HT

Category
Entity
Quantity
Experimental Paradigm
Age at Testing
Locomotor activity in diurnal cycle1
Decreased
Description: In females, decreased activity during light hours, with fewer rotations
Exp Paradigm: Wheel running monitoring
 Running wheel test
 7-20 weeks
Adaptation to dark phase1
Decreased
Description: In females, decreased overall activity during dark hours, with fewer rotation but with increased bouts of activity
Exp Paradigm: Wheel running monitoring
 Running wheel test
 7-20 weeks
Locomotor activity in diurnal cycle1
Increased
Description: In males, a slight increase in activity in light hours
Exp Paradigm: Wheel running monitoring
 Running wheel test
 7-20 weeks
Motor coordination and balance3
increased
Description: female Pten HT mice had shorter latency to fall on the first day of rotarod training
 Accelerating rotarod test
 P 78-128
Brain morphology3
abnormal
Description: divergent growth correlations between brain regions, indicating different patterns of growth between Pten WT and HT mice. Most overgrown areas were the pons, inferior colliculus, and medulla.
 Magnetic resonance imaging (MRI)
 P 7,P 60
Microglial number4
Increased
Description: Increased microglial density in mpfc
Exp Paradigm: NA
 Immunohistochemistry
 2 weeks
Brain size3
increased
Description: significantly increased absolute and relative volume in many brain regions, excluding cerebellar cortex and frontal lobe
 Magnetic resonance imaging (MRI)
 P 7,P 60
Microglial morphology4
Increased
Description: Increase in size of microglia in mpfc, hip, str
Exp Paradigm: NA
 Immunohistochemistry
 2 weeks
Brain size1
Increased
Description: Greater absolute and relative brain mass; both sexes reported, analyzed separately
Exp Paradigm: Brain weighed, body weighed
 General observations
 7-20 weeks
Microglial morphology4
Increased
Description: Increase in size of microglia in mpfc and str
Exp Paradigm: NA
 Immunohistochemistry
 2 weeks
Brain morphology1
Abnormal
Description: Overgrowth in anterior and posterior halves of cortex, in the cerebellum and in the remainder of the brain; only males reported
Exp Paradigm: Measurement of tissue weight
 Measurement of tissue weight
 7-20 weeks
Cued or contextual fear conditioning: memory of context: long term recall3
decreased
Description: male Pten HT froze less in the context test showing impaired remote contextual memory compared to WT males
Exp Paradigm: remote memory for trace fear conditioning
 Fear conditioning test
 P 73-123
Self grooming: perseveration2
Increased
Description: In males, increased self-grooming behavior
Exp Paradigm: Resident-intruder test: 15-minute interaction
 Resident-intruder test
 11-16 weeks
Repetitive digging2
Increased
Description: In males, increased digging behavior
Exp Paradigm: Resident-intruder test: 15-minute interaction
 Resident-intruder test
 11-16 weeks
Repetitive digging1
Increased
Description: In males, increased digging behavior
Exp Paradigm: Marble-burying test
 Marble-burying test
 7-20 weeks
Startle response: acoustic stimulus3
increased
Description: sexual dimmorphism; females are hyerpersensitive (lower startle threshold and increased startle amplitude) to acoustic stimuli of low intensity as juveniles and adults but not as adolescents
Exp Paradigm: low-dB
 Acoustic startle reflex test
 juvenile, adult
Startle response: acoustic stimulus3
decreased
Description: Pten HT male mice are hyporeactive (low startle amplitude and lower maximum startle response) to acoustic stimuli of high intensity throughout development while female only as adults, not juveniles or adolescent
Exp Paradigm: high-dB
 Acoustic startle reflex test
 juvenile, adult
Sensorimotor gating3
decreased
Description: impaired pre-pulse inhibition in Pten HT comared to WT, showing decreased inhibition of startle response
 Prepulse inhibition
 P 57-128
Aggression2
Decreased
Description: In males, residents exhibit less dominance scores, have fewer discrete attacks, spend less time fighting, and have a greater latency to attack
Exp Paradigm: Resident-intruder test: 15-minute interaction
 Resident-intruder test
 11-16 weeks
Social approach3
decreased
Description: female Pten HT mice fail to show social preference at P45 and in adulthood
 Three-chamber social approach test
 P 45, adult
Social memory1
Decreased
Description: Decreased preference for novel social stimulus; both sexes reported, analyzed separately
Exp Paradigm: Three-chamber social approach test
 Three-chamber social approach test
 7-20 weeks
Social approach1
Decreased
Description: Decreased preference for social stimulus; both sexes reported, analyzed separately
Exp Paradigm: Three-chamber social approach test
 Three-chamber social approach test
 7-20 weeks
Social interaction2
Decreased
Description: In males, decreased duration and frequence of solcial behavor
Exp Paradigm: Resident-intruder test: 15-minute interaction
 Resident-intruder test
 11-16 weeks
Rearing behavior2
Increased
Description: In males, increased vertical exploratory activity
Exp Paradigm: Resident-intruder test: 15-minute interaction
 Resident-intruder test
 11-16 weeks
Social habituation1
Decreased
Description: In males, decreased social habituation, compared to male controls
Exp Paradigm: Reciprocal social interaction test: confined stimulus mouse: juvenile conspecific p21-p28, four 5-minute stimulus presentations, 10-minute intertrial intervals
 Reciprocal social interaction test
 7-20 weeks
Response to novelty3
decreased
Description: Pten HT females investigate novel object less than WT females
 Novel object recognition test
 P 84-116
Depression1
Increased
Description: In males, increased immobility in the tail suspension test and the forced swim test
Exp Paradigm: Forced swim test
 Forced swim test
 7-20 weeks
Exploratory activity3
decreased
Description: male Pten HT spend less time than WT investigating the objects across all 3 phases, females Pten HT investigated less than WT in the first phase
 Novel object recognition test
 P 52-116
Anxiety1
Decreased
Description: In males, decreased latency to light compartment, increased number of crossings between light and dark, more time spent in light compartment, more time spent in center of open field
Exp Paradigm: Open field test
 Open field test
 7-20 weeks
Exploratory activity2
Increased
Description: In males, increased horizontal exploratory activity
Exp Paradigm: Resident-intruder test: 15-minute interaction
 Resident-intruder test
 11-16 weeks
Anxiety1
Decreased
Description: In males, decreased latency to light compartment, increased number of crossings between light and dark, more time spent in light compartment, more time spent in center of open field
Exp Paradigm: Light-dark exploration test
 Light-dark exploration test
 7-20 weeks
Depression1
Increased
Description: In males, increased immobility in the tail suspension test and the forced swim test
Exp Paradigm: Tail suspension test
 Tail suspension test
 7-20 weeks
Cued or contextual fear conditioning: trace fear conditioning1
Decreased
Description: In females, decreased freezing behavior when testing auditory cue
Exp Paradigm: Fear conditioning test: trace
 Fear conditioning test
 7-20 weeks
Cued or contextual fear conditioning: memory of context1
Decreased
Description: In females, decreased freezing behavior when testing context
Exp Paradigm: Fear conditioning test: trace
 Fear conditioning test
 7-20 weeks
Spatial working memory3
decreased
Description: female Pten HT mice showed increased latency to find the platform in the reversal stage, they also only showed trends of spending more time in the platform quadrant compared to WT females which spent significantly more time in the platform quardrant when compared to chance
 Morris water maze test
 P 60-89
Cognitive flexibility3
increased
Description: female Pten HT mice were significantly faster than WT on the tissue task which was not explained by differences in locomotion or anxiety
Exp Paradigm: tunnel, tunnel+bedding, tissue, and foam tasks
 Puzzle box test
 P 64-162
Procedural learning3
increased
Description: female Pten HT mice were significantly faster than WT on the tissue task which was not explained by differences in locomotion or anxiety
Exp Paradigm: tunnel, tunnel+bedding, tissue, and foam tasks
 Puzzle box test
 P 64-162
Adaptation to dark phase1
 No change
 Running wheel test
 7-20 weeks
Circadian rhythms: timing/phases of locomotor activity1
 No change
 Running wheel test
 7-20 weeks
Size/growth1
 No change
 General observations
 7-20 weeks
Anxiety1
 No change
 Light-dark exploration test
 7-20 weeks
Anxiety1
 No change
 Open field test
 7-20 weeks
Anxiety3
 no change
 Open field test
 P 71-170
Depression1
 No change
 Forced swim test
 7-20 weeks
Depression1
 No change
 Tail suspension test
 7-20 weeks
Habituation to aversive stimuli3
 no change
 Acoustic startle reflex test
 P 57-128
Habituation to aversive stimuli3
 no change
 Acoustic startle reflex test
 P 61-116
Response to novelty3
 no change
 Novel object recognition test
 P 52-99
Cognitive flexibility3
 no change
 Morris water maze test
 P 52-89
Cognitive flexibility3
 no change
 Puzzle box test
 P 57-99
Cued or contextual fear conditioning: memory of context1
 No change
 Fear conditioning test
 7-20 weeks
Cued or contextual fear conditioning: trace fear conditioning1
 No change
 Fear conditioning test
 7-20 weeks
Extinction of fear memory3
 no change
 Fear conditioning test
 P 62-118
Procedural learning3
 no change
 Puzzle box test
 P 57-99
Spatial learning3
 no change
 Morris water maze test
 P 52-89
Spatial working memory3
 no change
 Morris water maze test
 P 52-89
General locomotor activity3
 no change
 Three-chamber social approach test
 juvenile, P 35, P 45, adult
General locomotor activity: ambulatory activity1
 No change
 Open field test
 7-20 weeks
General locomotor activity: ambulatory activity1
 No change
 Three-chamber social approach test
 7-20 weeks
Motor coordination and balance1
 No change
 Accelerating rotarod test
 7-20 weeks
Motor coordination and balance3
 no change
 Accelerating rotarod test
 P 78-128
Motor coordination and balance: fine motor skills3
 no change
 Single-seed reaching test
 P 68-116
Motor learning3
 no change
 Accelerating rotarod test
 P 78-128
Cued or contextual fear conditioning: memory of context: long term recall3
 no change
 Fear conditioning test
 P 91-141
Cued or contextual fear conditioning: memory of cue: long term recall3
 no change
 Fear conditioning test
 P 91-141
Cued or contextual fear conditioning: memory of cue: long term recall3
 no change
 Fear conditioning test
 P 73-123
Cued or contextual fear conditioning: trace fear conditioning3
 no change
 Fear conditioning test
 P 88-132
Microglial number4
 No change
 Immunohistochemistry
 2 weeks
Repetitive digging1
 No change
 Marble-burying test
 7-20 weeks
Pain or nociception1
 No change
 Hot plate test
 7-20 weeks
Startle response: acoustic stimulus3
 no change
 Acoustic startle reflex test
 juvenile, adolescent, adult
Social approach3
 no change
 Three-chamber social approach test
 juvenile, adult
Social dishabituation1
 No change
 Reciprocal social interaction test
 7-20 weeks
Social habituation1
 No change
 Reciprocal social interaction test
 7-20 weeks
Social interaction1
 No change
 Reciprocal social interaction test
 7-20 weeks
 Not Reported: Circadian sleep/wake cycle, Communications, Developmental profile, Immune response, Learning & memory, Maternal behavior, Molecular profile, Motor phenotype, Neuroanatomy / ultrastructure / cytoarchitecture, Neurophysiology, Physiological parameters, Seizure, Sensory

M_PTEN_7_CKO_HM

Category
Entity
Quantity
Experimental Paradigm
Age at Testing
General locomotor activity: ambulatory activity1
Abnormal
Description: In males, decreased distance traveled during sociability test but not during social novelty test
Exp Paradigm: Three-chamber social approach test
 Three-chamber social approach test
 7-15 weeks
Social approach1
Decreased
Description: In females, decreased preference for social stimulus
Exp Paradigm: Three-chamber social approach test: sociability
 Three-chamber social approach test
 7-15 weeks
Social memory1
Decreased
Description: In males, no preference for novel social stimulus, compared to controls
Exp Paradigm: Three-chamber social approach test: social novelty
 Three-chamber social approach test
 7-15 weeks
General locomotor activity: ambulatory activity1
 No change
 Three-chamber social approach test
 7-15 weeks
Social approach1
 No change
 Three-chamber social approach test
 7-15 weeks
Social memory1
 No change
 Three-chamber social approach test
 7-15 weeks
 Not Reported: Circadian sleep/wake cycle, Communications, Developmental profile, Emotion, Immune response, Learning & memory, Maternal behavior, Molecular profile, Neuroanatomy / ultrastructure / cytoarchitecture, Neurophysiology, Physiological parameters, Repetitive behavior, Seizure, Sensory

M_PTEN_4_KI_M3M4_HM

Category
Entity
Quantity
Experimental Paradigm
Age at Testing
Morphology and size of the corpus callosum1
Increased
Description: Increased corpus callosum thickness compared to wild type and heterozygote
Exp Paradigm: Immunohistochemistry: mbp for corpus callosum, neun for cortex
 Immunohistochemistry
 6 weeks
Myelination1
Increased
Description: Increased ratio of corpus callosum thickness to cortical thickness, compared to wild-types and heterozygotes
Exp Paradigm: Immunohistochemistry: mbp for corpus callosum, neun for cortex
 Immunohistochemistry
 6 weeks
Gene expression1
Abnormal
Description: Differential expression of genes related to myelanation, the majority of which were upregulated; out of all differentially expressed genes there is a significant enrichment in myelination-related genes
Exp Paradigm: Rna sequencing of cortical tissue followed by pathway and gene ontology analysis of differentially expressed genes
 Rna sequencing
 6 weeks
Cortical thickness1
 No change
 Immunohistochemistry
 6 weeks
 Not Reported: Circadian sleep/wake cycle, Communications, Developmental profile, Emotion, Immune response, Learning & memory, Maternal behavior, Motor phenotype, Neurophysiology, Physiological parameters, Repetitive behavior, Seizure, Sensory, Social behavior

M_PTEN_5_KI_M3M4_HT

Category
Entity
Quantity
Experimental Paradigm
Age at Testing
Myelination1
Increased
Description: Increased ratio of corpus callosum thickness to cortical thickness, compared to wild-types
Exp Paradigm: Immunohistochemistry: mbp for corpus callosum, neun for cortex
 Immunohistochemistry
 6 weeks
Morphology and size of the corpus callosum1
Increased
Description: Increased corpus callosum thickness compared to wild type
Exp Paradigm: Immunohistochemistry: mbp for corpus callosum, neun for cortex
 Immunohistochemistry
 6 weeks
Cortical thickness1
 No change
 Immunohistochemistry
 6 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, Social behavior

M_PTEN_8_CKO_HM_DG

Category
Entity
Quantity
Experimental Paradigm
Age at Testing
Synapse density: excitatory1
Increased
Description: Increased number of quantal fractions (aepscs) that fit in an evoked epsc
Exp Paradigm: Whole-cell patch clamp
 Whole-cell patch clamp
 3-4.5 weeks
Dendritic architecture: dendritic tree complexity1
Increased
Description: Increased primary dendrites, number of branching points, number of distal branches, total surface area and dendritic volume
Exp Paradigm: Fluorescence microscopy
 Fluorescence microscopy
 3-4.5 weeks
Neuronal size1
Increased
Description: Increased soma size of neurons with pten deletion
Exp Paradigm: Immunohistochemistry: pten; fluorescence microscopy
 Immunohistochemistry
 3-4.5 weeks
Dendritic architecture: spine density1
Increased
Description: Increased protusion density, increased dendritic caliber
Exp Paradigm: Fluorescence microscopy
 Fluorescence microscopy
 3-4.5 weeks
Dendritic architecture: spine density1
Increased
Description: Increased filopodial density, mushroom spine density, and number of spine maturation events
Exp Paradigm: Fluorescence microscopy
 Fluorescence microscopy
 3-4.5 weeks
Miniature post synaptic current amplitude: excitatory1
Increased
Description: Increased amplitude of miniature epscs
Exp Paradigm: Whole-cell patch clamp
 Whole-cell patch clamp
 3-4.5 weeks
Neuronal activation1
Increased
Description: Increased neuronal activation of neurons with pten deletion, measured as an increase of phospho-s6
Exp Paradigm: Immunohistochemistry: phospho-s6; fluorescence microscopy
 Immunohistochemistry
 3-4.5 weeks
Ion influx and permeability: calcium1
Increased
Description: Neurons have greater calcium transients
Exp Paradigm: Fluorescence microscopy
 Fluorescence microscopy
 3-4.5 weeks
Network excitability1
Increased
Description: Developing granule neurons are more sensitive to to afferent stimulation
Exp Paradigm: Whole-cell patch clamp
 Whole-cell patch clamp
 3-4.5 weeks
Intrinsic membrane properties1
Abnormal
Description: Increase capacitance, decreased input resistance, increased threshold to fire action potential
Exp Paradigm: Whole-cell patch clamp
 Whole-cell patch clamp
 3-4.5 weeks
Synaptic transmission: excitatory1
Increased
Description: Increased amplitude of evoked ampa epscs
Exp Paradigm: Whole-cell patch clamp
 Whole-cell patch clamp
 3-4.5 weeks
Miniature post synaptic current frequency: excitatory1
Increased
Description: Increased frequency of miniature epscs
Exp Paradigm: Whole-cell patch clamp
 Whole-cell patch clamp
 3-4.5 weeks
Action potential property: firing rate1
Increased
Description: Increased threshold of current delivered to afferent stimulation to fire action potential, increased spike amplitude, more active spike rate
Exp Paradigm: Whole-cell patch clamp
 Whole-cell patch clamp
 3-4.5 weeks
Neurotransmitter release: quantal parameters1
Increased
Description: Small increase of asynchronous epscs, which represent quantal fractions
Exp Paradigm: Whole-cell patch clamp
 Whole-cell patch clamp
 3-4.5 weeks
Targeted expression1
Decreased
Description: Null expression of pten in neurons infected with retrovirus containing cre, compared to control without cre
Exp Paradigm: Immunohistochemistry: pten; fluorescence microscopy
 Immunohistochemistry
 2 weeks
Presynaptic function: paired-pulse facilitation1
 No change
 Whole-cell patch clamp
 3-4.5 weeks
Synaptic transmission: inhibitory1
 No change
 Whole-cell patch clamp
 3-4.5 weeks
 Not Reported: Circadian sleep/wake cycle, Communications, Developmental profile, Emotion, Immune response, Learning & memory, Maternal behavior, Motor phenotype, Physiological parameters, Repetitive behavior, Seizure, Sensory, Social behavior

M_PTEN_10_CKO_HM

Category
Entity
Quantity
Experimental Paradigm
Age at Testing
Anatomical projections and connectivity1
Increased
Description: The neuropils of the parvalbumin +ve neurons extend upto layer i, unlike in wt controls where these projections remain within layerii to vi in mice with pten cko homozygous for sub ventricular zone (svz) derived interneurons
Exp Paradigm: NA
 Immunohistochemistry
 P18
Neuronal number: interneurons1
Decreased
Description: There is a very significant decrease in the number of interneurons in the pten cko homozygous mice, that are believed to be the lineage of the dlxl12b-cre expressing cells. there is an overall reduction of 53% in total number of interneurons in these mouse brains as well
Exp Paradigm: Sst
 Immunohistochemistry
 P18
Mortality/lethality1
Increased
Description: The loss of pten from the secondary progenitors of the sub ventricular zone in this model leads to increased mortality as the mice die after p18
Exp Paradigm: NA
 Survival analysis
 P18
 Not Reported: Circadian sleep/wake cycle, Communications, Emotion, Immune response, Learning & memory, Maternal behavior, Molecular profile, Motor phenotype, Neurophysiology, Physiological parameters, Repetitive behavior, Seizure, Sensory, Social behavior

M_PTEN_10_CKO_HT

Category
Entity
Quantity
Experimental Paradigm
Age at Testing
Neuronal number: interneurons1
 No change
 Immunohistochemistry
 4 weeks
Neuronal specification1
 No change
 NA
 4 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, Social behavior

M_PTEN_11_CKO_HM_SSTN

Category
Entity
Quantity
Experimental Paradigm
Age at Testing
Anatomical projections and connectivity1
 No change
 Immunohistochemistry
 4 weeks
Neuronal number: interneurons1
 No change
 Immunohistochemistry
 4 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, Social behavior

M_PTEN_11_CKO_HT_SSTN

Category
Entity
Quantity
Experimental Paradigm
Age at Testing
Neuronal number: interneurons1
 No change
 Immunohistochemistry
 P18
Neuronal specification1
 No change
 NA
 P18
 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, Social behavior

M_PTEN_12_CKO_HM

Category
Entity
Quantity
Experimental Paradigm
Age at Testing
Neuronal size1
Increased
Description: There is also an increase in the soma size of the transplanted pten cko cells, indicating it a cell autonomous phenotype
Exp Paradigm: NA
 Immunohistochemistry
 35dpt
Neuronal differentiation1
Decreased
Description: The transplanted cells lacking pten (pten cko mge cells) recapitulate the phenotype of the donor mice, and have significant reduction in somatostatin expressing interneurons that develop from the pten cko progenitor cells and there is a significant reduction in the ratio of sst/ pv expressing interneurons that develop from the transplants
Exp Paradigm: NA
 NA
 35dpt
Intrinsic membrane properties1
Decreased
Description: The resting membrane potential and the action potential threshold is significantly reduced in the transplanted pten cko neurons present in the brain of wt mice
Exp Paradigm: NA
 Whole-cell patch clamp
 45dpt
Action potential property: firing rate1
 No change
 Whole-cell patch clamp
 45dpt
 Not Reported: Circadian sleep/wake cycle, Communications, Developmental profile, Emotion, Immune response, Learning & memory, Maternal behavior, Molecular profile, Motor phenotype, Physiological parameters, Repetitive behavior, Seizure, Sensory, Social behavior

M_PTEN_12_CKO_HT

Category
Entity
Quantity
Experimental Paradigm
Age at Testing
Neuronal differentiation1
Decreased
Description: Heterozygous pten cko in mge derived progenitors show decrease in the number of somatostatin expressing interneurons arising from the transplants
Exp Paradigm: NA
 NA
 4 weeks
Neuronal size1
 No change
 Immunohistochemistry
 4 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, Social behavior

M_PTEN_9_CKO_HM

Category
Entity
Quantity
Experimental Paradigm
Age at Testing
Neuronal differentiation1
Decreased
Description: There is reduction in sst expressing interneurons in the hippocampus as well.
Exp Paradigm: NA
 NA
 4 weeks
Neuronal differentiation1
Decreased
Description: The reduction in interneuronal number is biased towards loss of somatostatin (sst) expressing interneurons in the cortex, it is first detectable staring e15.5 and continues to be significant through p30. the ratio of sst/pv expressing interneurons is signficantly reduced.
Exp Paradigm: NA
 NA
 E15.5, 4 weeks
Neuronal size1
Increased
Description: The cell/soma size of interneurons that have homozygous loss of pten is significantly increased.
Exp Paradigm: NA
 Immunohistochemistry
 NA
Anatomical projections and connectivity1
Increased
Description: The neuropils of the parvalbumin +ve neurons extend upto layer i, unlike in wt controls where these projections remain within layerii to vi in mice with pten cko (homozygous) mge derived interneurons
Exp Paradigm: NA
 Immunohistochemistry
 4 weeks
Neuronal number: interneurons1
Decreased
Description: There is a very significant decrease in the number of interneurons in the pten cko homozygous mice, that are believed to be the lineage of the nkx2.1-cre expressing cells. there is an overall reduction of 53% in total number of interneurons, a majority of which occurs in between p0 and p8.
Exp Paradigm: Nkx2.1
 Immunohistochemistry
 P0, p8, 4 weeks
Apoptosis: brain cells1
Increased
Description: The interneurons that express somatostatin and are pten cko homozygous, have a much higher probability of expressing cleaved caspace3
Exp Paradigm: Expression of cleaved caspase-3 (cc3)
 Expression of cleaved caspase-3 (cc3)
 P0
Spontaneous post synaptic events: inhibitory currents1
Increased
Description: There is a two fold increase in the frequency of spontaneous ipscs onto layer ii/iii excitatory neurons that are connected to the mge derived interneurons (pten cko, homozygous in this model)
Exp Paradigm: NA
 Whole-cell patch clamp
 4 weeks
Apoptosis: brain cells1
Increased
Description: The interneurons that express somatostatin and are pten cko homozygous, have a much higher probability of expressing cleaved caspace3
Exp Paradigm: Cell counting
 Cell counting
 P0
Electroencephalogram (eeg) signature1
Increased
Description: The pten cko homozygous interneurons had an elevated egg power during behavioral testing in the reciproal social interaction in the gamma frequency band
Exp Paradigm: NA
 Electroencephalogram (eeg)
 4 weeks
Apoptosis: brain cells1
Increased
Description: There is a significant increase in the expression of cleaved caspase3(cc3), an apoptopis marker,in the mfe derived interneurons of the neocortex and lateral cortex marginal zone (nkx2.1-cre/ tdtomato marker expressing). by p0 about 36% of the pten cko homozygous cells express cc3.
Exp Paradigm: NA
 Expression of cleaved caspase-3 (cc3)
 E17.5, p0
Social interaction1
Decreased
Description: Mice with pten cko homozygous mge derived interneurons have significantly reduced preference for a novel mouse compared to control mice
Exp Paradigm: NA
 Reciprocal social interaction test
 P30
Size/growth1
Decreased
Description: There is a 25% reduction in body weight in pten cko compared to control littermates
Exp Paradigm: NA
 Body weight measurement
 4 weeks
Object recognition memory1
Decreased
Description: Mice with pten cko homozygous mge derived interneurons ,spend less time exploring the novel object compared to familiar object
Exp Paradigm: NA
 Novel object recognition test
 NA
Protein phosphorylation1
Increased
Description: There is increased phosphorylated forms of akt (pakt) and gsk3 (pfgsk3) in the medial ganglionic eminence (mge) in mice with pten cko mge interneurons
Exp Paradigm: NA
 Western blot
 E13.5
Mortality/lethality1
 No change
 General observations
 NA
Anxiety1
 No change
 Elevated plus maze test
 NA
Anxiety1
 No change
 Open field test
 NA
Cell proliferation1
 No change
 Edu incorporation
 NA
Cell proliferation1
 No change
 Phospho-histone 3 expression
 NA
General locomotor activity1
 No change
 Open field test
 NA
Brain size1
 No change
 Histology
 4 weeks
Spontaneous post synaptic event amplitude: inhibitory currents1
 No change
 Whole-cell patch clamp
 4 weeks
 Not Reported: Circadian sleep/wake cycle, Communications, Immune response, Maternal behavior, Physiological parameters, Repetitive behavior, Seizure, Sensory

M_PTEN_9_CKO_HT

Category
Entity
Quantity
Experimental Paradigm
Age at Testing
Mortality/lethality1
 No change
 General observations
 NA
Size/growth1
 No change
 Body weight measurement
 4 weeks
Anatomical projections and connectivity1
 No change
 Immunohistochemistry
 4 weeks
Brain size1
 No change
 Histology
 4 weeks
Neuronal differentiation1
 No change
 NA
 4 weeks
Spontaneous post synaptic events: inhibitory currents1
 No change
 Whole-cell patch clamp
 4 weeks
 Not Reported: Circadian sleep/wake cycle, Communications, Emotion, Immune response, Learning & memory, Maternal behavior, Molecular profile, Motor phenotype, Physiological parameters, Repetitive behavior, Seizure, Sensory, Social behavior

M_PTEN_17_CKO_HM_PURKINJEN

Category
Entity
Quantity
Experimental Paradigm
Age at Testing
Motor coordination and balance1
Decreased
Description: Older pten cko males show significant reduction in motor coordination, as they have shorter latency to fall from the accelerating rotarod and also do not improve in performance, unlike wild type controls
Exp Paradigm: Males only
 Accelerating rotarod test
 5-6 months
Dendritic architecture: spine density1
Increased
Description: Pten ko purkinje cells in have hypertrophic spines, notably in the dendrites that are thicker compared to pcs in wild type mice
Exp Paradigm: Males only
 Immunohistochemistry
 2 months, 4 months, 6 months
Dendritic architecture: dendritic tree complexity1
Increased
Description: Pten ko purkinje cells have thicker dendrites with more focal dendritic swellings and torpedoes compared to wild type mice, that increase with age in pten cko mice cerebella
Exp Paradigm: Males only
 Immunohistochemistry
 2 months, 4 months, 6 months
Neuronal size1
Increased
Description: Size of pten ko purkinje cells is increased in all the lobules of the cerebellum by 15% ( 2 months), 50% (4 months) and 60% (9 months)
Exp Paradigm: Males only
 Cell size measurement
 2 months, 4 months, 9 months
Cytoskeletal organization: neuronal1
Increased
Description: There is an increase in the neurofilament content in pten ko purkinje cell compartments, along with an increase in map2 and acetylated alpha-tubulin levels
Exp Paradigm: Males only
 Immunohistochemistry
 NA
Neuronal number: purkinje cells1
Decreased
Description: By 6 months of age there is a significant reduction in purkinje cell number in the cerebella of pten cko mice, they have about 50% less pcs than wt
Exp Paradigm: Males only
 Immunohistochemistry
 6 months
Cerebellar morphology: molecular layer thickness1
Increased
Description: Thickness of the molecular layer is increased in the cerebella of the pten cko mice at 2 and 4 months compared to controls
Exp Paradigm: Males only
 Immunostaining
 2months, 4 months
Apoptosis: brain cells1
Increased
Description: By 9 months of age there is a significant increase in the expression of cleaved caspase-3 in the purkinje cells indicative of apoptosis, while wt controls have no expression of cleaved caspase three
Exp Paradigm: Males only
 Expression of cleaved caspase-3 (cc3)
 9 months
Synaptic transmission: excitatory1
Increased
Description: Epscs have increased amplitude in the parallel fiber- purkinje cell synapse (pf-pc) in pten cko mice cerebellar slices
Exp Paradigm: Males only
 Cell-attached patch clamp
 3-4.5 months
Action potential property: firing rate1
Decreased
Description: Spontaneous as well as evoked action potential firing is decreased in single pten ko purkinje cells
Exp Paradigm: Males only
 Cell-attached patch clamp
 3-4.5 months
Synaptic transmission: excitatory1
Increased
Description: Epscs have increased amplitude in the climbing fiber- purkinje cell synapse (cf-pc) in pten cko mice cerebellar slices
Exp Paradigm: Males only
 Cell-attached patch clamp
 3-4.5 months
Presynaptic function: paired-pulse depression (ppd)1
Decreased
Description: Pten ko purkinje cells had reduced depression (ppd) in the cf- pc synapse
Exp Paradigm: Males only
 Whole-cell patch clamp
 3-4.5 months
Vertical jumping or back flipping1
Increased
Description: Pten cko mice had a significantly increase in jumping or scrabbling behavior compard to wild type controls
Exp Paradigm: Males only
 Observation of repetitive behavior
 3-4months
Self grooming: perseveration1
Decreased
Description: Pten cko mice show decreased time spent in self-grooming compared to wild type controls
Exp Paradigm: Males only
 Grooming behavior assessments
 3-4 months
Social approach1
Decreased
Description: Pten cko mice show no preference to stranger mouse over chamber with an object unlike wild type controls
Exp Paradigm: Males only-three-chamber social approach test
 Three-chamber social approach test
 3-4months
Social approach1
Decreased
Description: Pten cko mice show no preference to stranger mouse over chamber with an object unlike wild type controls
Exp Paradigm: Males only- open field test
 Open field test
 3-4months
Social interaction: opposite sex1
Decreased
Description: Pten cko mice spend significantly less time sniffing, allogrooming or mounting a conspecific female compared to wild type control males
Exp Paradigm: Males only
 Reciprocal social interaction test
 3-4months
Signaling1
Increased
Description: Pten cko mice have increased signaling in the mtor pathway determined by increase in phosphorylated ribosomal protein s6
Exp Paradigm: Males only
 Immunohistochemistry
 4 months
Mortality/lethality1
 No change
 General observations
 Adult
Cognitive flexibility1
 No change
 Water t-maze test
 Adult
Cognitive flexibility1
 No change
 Morris water maze test
 Adult
Spatial learning1
 No change
 Morris water maze test
 Adult
Spatial reference memory1
 No change
 Morris water maze test
 Adult
General locomotor activity1
 No change
 Open field test
 4 months
General locomotor activity1
 No change
 Three-chamber social approach test
 4 months
Motor coordination and balance1
 No change
 Accelerating rotarod test
 2-3 months
Cerebellar morphology1
 No change
 Histology
 2-3 months
Presynaptic function: paired-pulse facilitation1
 No change
 Paired-pulse ratio
 3-4.5 months
Reproductive function1
 No change
 General observations
 Adult
 Not Reported: Circadian sleep/wake cycle, Communications, Emotion, Immune response, Maternal behavior, Physiological parameters, Seizure, Sensory

M_PTEN_18_CKO_HT_SEIZURE

Category
Entity
Quantity
Experimental Paradigm
Age at Testing
Hyperactivity1
Increased
Description: Status epilectus in pten het mice increases distance traveled in the open field as well as in the elevated plus maze as wt controls
Exp Paradigm: Males only- elevated plus maze test
 Elevated plus maze test
 9 weeks
Hyperactivity1
Increased
Description: Status epilectus in pten het mice increases distance traveled in the open field as well as in the elevated plus maze as wt controls
Exp Paradigm: Males only-open field test
 Open field test
 9 weeks
Social approach1
Decreased
Description: Status epilectus in pten het mice causes reduced preference for mouse over object in the three chamber test, compared to control pten hets (which have normal preference to mouse )
Exp Paradigm: Males only
 Three-chamber social approach test
 10 weeks
Cued or contextual fear conditioning: trace fear conditioning1
Increased
Description: Status epilectus in pten het mice causes increased freezing and learning in the trace fear conditioning test compared to wt and pten het controls
Exp Paradigm: Males only
 Fear conditioning test
 12 weeks
Anxiety1
 No change
 Open field test
 9 weeks
Exploratory activity1
 No change
 Three-chamber social approach test
 10 weeks
Cued or contextual fear conditioning: memory of context1
 No change
 Fear conditioning test
 12 weeks
Spatial learning1
 No change
 Morris water maze test
 13 weeks
Spatial reference memory1
 No change
 Morris water maze test
 13 weeks
Protein expression level evidence1
 No change
 Western blot
 14 weeks
Protein localization: synapse1
 No change
 Western blot
 14 weeks
Protein phosphorylation1
 No change
 Western blot
 14 weeks
Swimming ability1
 No change
 Morris water maze test
 12-13 weeks
Circling1
 No change
 Open field test
 9 weeks
Repetitive digging1
 No change
 Marble-burying test
 10 weeks
Self grooming: perseveration1
 No change
 Open field test
 9 weeks
 Not Reported: Circadian sleep/wake cycle, Communications, Developmental profile, Immune response, Maternal behavior, Neuroanatomy / ultrastructure / cytoarchitecture, Neurophysiology, Physiological parameters, Seizure, Sensory

M_PTEN_13_KI_HM

Category
Entity
Quantity
Experimental Paradigm
Age at Testing
Brain size1
Increased
Description: Mutants show small but significant macrocephaly.
Exp Paradigm: Nissl stain
 Stereology
 6-12 months
Synaptic plasticity: ltd1
Decreased
Description: Mutants show impaired nmdar-dependent synaptic depression (ltd) at cortico-amygdala synapses in the lateral amygdala compared with controls.
Exp Paradigm: Ltd was induced at lateral amygdala synapses in coronal slices by stimulating afferents from the auditory cortex (15 min at 1 hz) and recording the fepsps in the la.
 Field potential recordings
 6-12 months
Social approach1
Decreased
Description: Mutants show decreased social interactions compared with controls.
Exp Paradigm: NA
 Three-chamber social approach test
 6-12 months
Ultrasonic vocalization: isolation induced1
 No change
 Monitoring ultrasonic vocalizations;
 P3-p12
Size/growth1
 No change
 Measurement of body weight
 6-12 months
Anxiety1
 No change
 Elevated plus maze test
 6-12 months
Cued or contextual fear conditioning: memory of context1
 No change
 Fear conditioning test
 6-12 months
Cued or contextual fear conditioning: memory of cue1
 No change
 Fear conditioning test
 6-12 months
Morphology of the amygdala1
 No change
 Stereology
 6-12 months
Post-synaptic density size1
 No change
 Electron microscopy
 6-12 months
Synapse density1
 No change
 Electron microscopy
 6-12 months
Synaptic plasticity: ltd1
 No change
 Field potential recordings
 6-12 months
 Not Reported: Circadian sleep/wake cycle, Immune response, Maternal behavior, Molecular profile, Motor phenotype, Physiological parameters, Repetitive behavior, Seizure, Sensory

M_PTEN_14_TG

Category
Entity
Quantity
Experimental Paradigm
Age at Testing
Hippocampal morphology1
Decreased
Description: Mutants show decrease in hippocampal volume.
Exp Paradigm: NA
 Cranial computerized tomography (ct) scan
 6-12 months
Dendritic architecture: spine density1
Decreased
Description: Mutants show decrease in spine density in the la pyramidal neurons and hippocampal neurons, particularly in distal dendrites.
Exp Paradigm: NA
 Sholl analysis
 6-12 months
Synapse density: excitatory1
Decreased
Description: Mutants show decreased excitatory synaptic density in the lateral amygdala and the hippocampus, indicating structural hypoconnectivity.
Exp Paradigm: NA
 Electron microscopy
 6-12 months
Dendritic architecture: dendritic tree complexity1
Decreased
Description: Mutants show decrease in dendritic complexity in la pyramidal neurons.
Exp Paradigm: NA
 Sholl analysis
 6-12 months
Post-synaptic density size1
Decreased
Description: Mutants show decrease in the number of synapses with large psds in the lateral amygdala. mutants show no change in the average length of the psds in the lateral amygdala.
Exp Paradigm: Lateral amygdala
 Electron microscopy
 6-12 months
Brain size1
Decreased
Description: Mutants show microcephaly compared with controls.
Exp Paradigm: NA
 Stereology
 6-12 months
Neuronal activation following behavioral stimulation1
Decreased
Description: Mutants displayed overall less activity induced by fear conditioning in the auditory cortex. mutants show decrease in glucose uptake in brain areas involved in auditory fear conditioning such as the hippocampus, amygdala, and thalamus, during retrieval of conditional cued memory.
Exp Paradigm: Micro-ct-pet was performed using a radiolabeled glucose analogue (2-deoxy-2-[18f]fluoro-d-glucose, 18ffdg) as the tracer; hippocampus, amygdala, and thalamus.
 Positron emission tomography (pet) imaging
 6-12 months
Epsp-spike relationship1
Decreased
Description: Mutants show decrease in basal transmission was at synapses receiving thalamic input in the lateral amygdala.
Exp Paradigm: Thalamus; lateral amygdala
 Field potential recordings
 6-12 months
Synaptic plasticity: ltd1
Increased
Description: Mutants show enhanced synaptic depression at cortico-amygdala (ac-la) synapses compared with controls.
Exp Paradigm: Ltd was induced at lateral amygdala synapses in coronal slices by stimulating afferents from the auditory cortex (15 min at 1 hz) and recording the fepsps in the la.
 Whole-cell patch clamp
 6-12 months
Neuronal activation1
Decreased
Description: Mutants show less activity in the frontal and auditory cortices and in the ventral hippocampus.
Exp Paradigm: Micro-ct-pet was performed using a radiolabeled glucose analogue (2-deoxy-2-[18f]fluoro-d-glucose, 18ffdg) as the tracer.
 Positron emission tomography (pet) imaging
 6-12 months
Synaptic plasticity: hippocampal ltp1
Decreased
Description: Mutants show decreased ltp at hippocampal ca3-ca1 synapses.
Exp Paradigm: Hippocampus
 Whole-cell patch clamp
 6-12 months
Neuronal activation1
Increased
Description: Mutants show increased basal activity in the dorsal hippocampus.
Exp Paradigm: Micro-ct-pet was performed using a radiolabeled glucose analogue (2-deoxy-2-[18f]fluoro-d-glucose, 18ffdg) as the tracer.
 Positron emission tomography (pet) imaging
 6-12 months
Synaptic plasticity: amygdalar ltp1
Decreased
Description: Mutants show significantly less ltp at synapses in the auditory cortex - lateral amygdalar pathway.
Exp Paradigm: Auditory cortex; lateral amygdala
 Whole-cell patch clamp
 6-12 months
Intrinsic membrane properties1
Decreased
Description: Mutants show decreased capacitance in la pyramidal neurons, indicating decrease in total surface area of neuronal membrane.
Exp Paradigm: NA
 Whole-cell patch clamp
 6-12 months
Social approach1
Increased
Description: Mutants spent more time in social interactions compared with controls.
Exp Paradigm: NA
 Three-chamber social approach test
 6-12 months
Ultrasonic vocalization: isolation induced1
Decreased
Description: Mutant pups produced fewer and shorter calls of lower intensity.
Exp Paradigm: NA
 Monitoring ultrasonic vocalizations;
 P3-p12
Size/growth1
Decreased
Description: Mutants show decreased body weight compared with controls.
Exp Paradigm: NA
 Body weight measurement
 6-12 months
Anxiety1
Decreased
Description: Mutants spend more time in the open arms compared with controls.
Exp Paradigm: NA
 Elevated plus maze test
 6-12 months
Spatial working memory1
Decreased
Description: Mutants made more errors in finding the escape hole during the learning phase (days 1-3) and a higher latency in reaching the escape hole. on the last training day (day 4), mutants show no change in latency in reaching the escape hole, indicating a delay in hippocampal-dependent learning.
Exp Paradigm: NA
 Barnes maze test
 6-12 months
Cued or contextual fear conditioning: memory of cue1
Decreased
Description: Mutants show decrease in freezing rates on presentation of the tone indicating impaired amygdala-dependent fear recall.
Exp Paradigm: NA
 Fear conditioning test
 6-12 months
Cognitive flexibility1
Decreased
Description: When the escape hole was moved to the opposite side of the barnes maze, mutant mice made more errors on day 1 but showed no change on day 2, indicating a delay in hippocampal-dependent learning.
Exp Paradigm: NA
 Barnes maze test
 6-12 months
Exploratory activity1
 No change
 Open field test
 6-12 months
Habituation to aversive stimuli1
 No change
 Fear conditioning test
 6-12 months
Object recognition memory1
 No change
 Object-place recognition test
 6-12 months
Spatial reference memory1
 No change
 Barnes maze test
 6-12 months
Morphology of the amygdala1
 No change
 Cranial computerized tomography (ct) scan
 6-12 months
Post-synaptic density size1
 No change
 Electron microscopy
 6-12 months
Epsp-spike relationship1
 No change
 Field potential recordings
 6-12 months
Intrinsic membrane properties1
 No change
 Whole-cell patch clamp
 6-12 months
Neuronal activation1
 No change
 Positron emission tomography (pet) imaging
 6-12 months
Synaptic plasticity: amygdalar ltp1
 No change
 Whole-cell patch clamp
 6-12 months
Synaptic plasticity: ltd1
 No change
 Whole-cell patch clamp
 6-12 months
Self grooming: perseveration1
 No change
 Grooming behavior assessments
 6-12 months
Pain or nociception1
 No change
 Foot shock test
 6-12 months
 Not Reported: Circadian sleep/wake cycle, Immune response, Maternal behavior, Molecular profile, Motor phenotype, Physiological parameters, Seizure

M_PTEN_20_CKO_HM

Category
Entity
Quantity
Experimental Paradigm
Age at Testing
General locomotor activity: ambulatory activity2
Increased
Description: Mutants show increase in total distance travelled.
Exp Paradigm: NA
 Open field test
 1.7 months
Brain size1
Increased
Description: Mutants show increase in brain weight.
Exp Paradigm: NA
 Measurement of tissue weight
 5-7 weeks
Neuronal number: interneurons2
Abnormal
Description: Mutants show reduced somatostatin to parvalbumin interneuron ratios.
Exp Paradigm: NA
 Immunohistochemistry
 1.8 months
Epsp-spike relationship1
Increased
Description: Mutants show increased runaway excitability upon equivalent step injection of currents in ca1 neurons.
Exp Paradigm: NA
 Whole-cell patch clamp
 5-7 weeks
Electroencephalogram (eeg) signature2
Decreased
Description: Mutants show decreased baseline eeg gamma power.
Exp Paradigm: NA
 Electroencephalogram (eeg)
 1.7 months
Spontaneous post synaptic event frequency: inhibitory currents2
Increased
Description: Mutants show increased sipsc frequency in pyramidal neurons of the recipient medial pfc.
Exp Paradigm: NA
 Whole-cell voltage clamp
 1.7 months
Spontaneous post synaptic event frequency: excitatory currents1
Increased
Description: Mutants show increase in frequency of sepscs in hippocampal slices in whole-cell recordings from ca1 neuron.
Exp Paradigm: NA
 Whole-cell patch clamp
 5-7 weeks
Seizures1
Increased
Description: Mutants show increase in seizure frequency.
Exp Paradigm: NA
 Observation of seizures
 5-7 weeks
Electroencephalogram (eeg): signature of seizure/epilepsy1
Increased
Description: Mutants show abnormal interictal spikes, eeg waveforms and tonic-clonic seizures.
Exp Paradigm: NA
 Electroencephalogram (eeg)
 5-7 weeks
Social memory1
Decreased
Description: Mutants show decreased preference for a novel over a familiar mouse.
Exp Paradigm: NA
 Three-chamber social approach test
 5-7 weeks
Social interaction: with juveniles2
Decreased
Description: Mutants spent less time interacting with a juvenile mouse of the same sex in the home cage.
Exp Paradigm: NA
 Reciprocal social interaction test
 1.7 months
Social interaction1
Decreased
Description: Mutants show decrease in direct social interaction.
Exp Paradigm: NA
 Reciprocal social interaction test
 5-7 weeks
Mortality/lethality1
Increased
Description: Mutants show decreased survival.
Exp Paradigm: NA
 Kaplan-meier survival curve
 5-7 weeks
Anxiety2
Increased
Description: Mutants avoided the center of the open field.
Exp Paradigm: NA
 Open field test
 1.7 months
Object recognition memory1
Decreased
Description: Mutants show decrease in preference for novel over familiar object.
Exp Paradigm: NA
 Novel object recognition test
 5-7 weeks
Cued or contextual fear conditioning: memory of context1
Decreased
Description: Mutants show decrease in freezing upon being exposed to the context 24 hours after training.
Exp Paradigm: NA
 Fear conditioning test
 5-7 weeks
Cognitive flexibility1
Decreased
Description: Mutants repeatedly explored the same arm of the maze.
Exp Paradigm: NA
 T-maze test
 5-7 weeks
Metabolite levels: neurometabolites1
Increased
Description: Mutants show increase in glycolytic metabolites in the brain, glucose, glucose 6-p and lactate.
Exp Paradigm: NA
 Comprehensive metabolic evaluation
 5-7 weeks
Signaling: mtor pathway1
Increased
Description: Mutant mice show increase in activity of both mtorc1 measured by the phosphorylation of its downstream target protein s6 at ser 240/244 and mtorc2 measured by the phosphorylation of its downstream target akt at ser 473, in the hippocampus and cortex.
Exp Paradigm: NA
 Western blot
 5-7 weeks
Protein phosphorylation1
Increased
Description: Mutants show a selective increase in phosphorylation of akt-ser473 in the hippocampus but no change in phosphorylation of pkc or ndrg1.
Exp Paradigm: NA
 Western blot
 5-7 weeks
Anxiety2
 No change
 Elevated plus maze test
 1.7 months
Object recognition memory2
 No change
 Novel object recognition test
 1.7 months
Mitochondrial biogenesis1
 No change
 Comprehensive metabolic evaluation
 5-7 weeks
Action potential property: half-width2
 No change
 Whole-cell voltage clamp
 1.7 months
Mitochondrial activity1
 No change
 Comprehensive metabolic evaluation
 5-7 weeks
Spontaneous post synaptic event amplitude: excitatory currents1
 No change
 Whole-cell patch clamp
 5-7 weeks
Spontaneous post synaptic event amplitude: inhibitory currents2
 No change
 Whole-cell voltage clamp
 1.7 months
Spontaneous post synaptic event amplitude: inhibitory currents1
 No change
 Whole-cell patch clamp
 5-7 weeks
Spontaneous post synaptic event frequency: inhibitory currents1
 No change
 Whole-cell patch clamp
 5-7 weeks
Electroencephalogram (eeg): signature of seizure/epilepsy2
 No change
 Whole-cell patch clamp
 1.7 months
Pain or nociception1
 No change
 Fear conditioning test
 5-7 weeks
Social approach1
 No change
 Three-chamber social approach test
 5-7 weeks
 Not Reported: Circadian sleep/wake cycle, Communications, Developmental profile, Emotion, Immune response, Maternal behavior, Molecular profile, Motor phenotype, Physiological parameters, Repetitive behavior, Sensory

M_PTEN_21_CKO_HM

Category
Entity
Quantity
Experimental Paradigm
Age at Testing
Dendritic architecture: dendritic tree complexity1
Increased
Description: Vehicle-treated Pten knockout neurons had an increased number of intersections. Vehicle-treated Pten knockout neurons have more primary dendrites protruding directly out of the soma compared with their wildtype control. Vehicle-treated Pten knockout neurons displayed an increase in the number of branch points (nodes) and endpoints.
 Sholl analysis
 P31
Neuronal size1
Increased
Description: Pten knockout granule neurons had a greater soma size.
Exp Paradigm: GFP, mCherry
 Immunofluorescence staining
 P31
Dendritic architecture: dendritic length1
Increased
Description: Vehicle-treated Pten knockout neurons had increased total dendritic length. Vehicle-treated Pten knockout neurons had increased dendritic growth, confirmed by Sholl analysis of dendritic length dendritic lengths.
Exp Paradigm: GFP, mCherry
 Immunofluorescence staining
 P31
Neuronal migration1
Increased
Description: Vehicle-treated Pten knockout neurons displayed significantly farther migration along the granule cell layer.
Exp Paradigm: GFP, mCherry
 Immunofluorescence staining
 P31
Dendritic architecture: spine density1
Increased
Description: Vehicle-treated Pten knockout neurons had increased spine density in the middle molecular layer.
Exp Paradigm: GFP, mCherry
 Immunofluorescence staining
 P31
 Not Reported:

M_PTEN_22_CKO_HM

Category
Entity
Quantity
Experimental Paradigm
Age at Testing
Dendritic architecture: spine morphology1
Abnormal
Description: Pten knockout neurons had decreased spine head diameter. Pten knockout neurons displayed increased spine length.
Exp Paradigm: GFP, mCherry
 Immunofluorescence staining
 
Dendritic architecture: spine density1
Increased
Description: Pten knockout neurons had greater dendritic spine density.
Exp Paradigm: GFP, mCherry
 Immunofluorescence staining
 P28
Neuronal size1
Increased
Description: Pten knockout neurons had significantly greater soma size compared to wildtype control.
Exp Paradigm: GFP, mCherry
 Immunofluorescence staining
 P28
Dendritic architecture: dendritic tree complexity1
Increased
Description: Pten knockout neurons had an increased number of intersections, when compared with wildtype control neurons. Pten knockout neurons have more primary dendrites protruding directly out of the soma, when compared with their wildtype control. Pten knockout neurons had an increased number of branch points (nodes) and endpoints.
 Sholl analysis
 
Neuronal migration1
Increased
Description: The Pten knockout neurons migrate significantly farther from the hilus along the granule cell layer, when compared with their wildtype control.
Exp Paradigm: GFP, mCherry
 Immunofluorescence staining
 P28
Dendritic architecture: dendritic length1
Increased
Description: The total dendritic length was increased in Pten knockout neurons. Pten knockout neurons had increased dendritic growth, confirmed by Sholl analysis of dendritic length.
Exp Paradigm: GFP, mCherry
 Immunofluorescence staining
 
Signaling: PI3K/AKT pathway1
Increased
Description: Knockout of Pten resulted in an increase in the phosphorylation of AKT Thr308.
Exp Paradigm: phospho-AKT
 Immunohistochemistry
 
Signaling: MTOR pathway1
Increased
Description: Pten knockout neurons showed elevated pS6 fluorescence intensity relative to their in-tissue wildtype controls, indicating increased mTORC1 activity.
Exp Paradigm: phospho-S6
 Immunohistochemistry
 P28
 Not Reported:

M_PTEN_23_CKO_HM

Category
Entity
Quantity
Experimental Paradigm
Age at Testing
Microglial morphology1
Abnormal
Description: PTEN mutant mice microglia from both the somatosensory (S1) and auditory (Au1) cortex showed decreased process length, decreased number of branch points, increased volume, and decreased number of terminal points, as compared to controls.
Exp Paradigm: Iba1, software-based morphometric analysis
 Immunohistochemistry
 2-3 months
Neurotransporter expression1
Decreased
Description: PTEN mutant mice exhibited a significant reduction in presynaptic VGLUT1 and synaptophysin compared to control synaptosomes, as demonstrated by Western blot.
Exp Paradigm: VGLUT1
 Western blot
 2-3 months
Neurotransporter expression1
Decreased
Description: PTEN mutant mice exhibited a reduction in VGLUT1 puncta in the deep layers of somatosensory and auditory cortices compared to controls, as demonstrated by immunostaining of tissue sections.
Exp Paradigm: VGLUT1
 Immunohistochemistry
 P14
Synapse density: excitatory1
Decreased
Description: Excitatory synapses, as identified by colocalized VGLUT1 and PSD95 staining, showed a significantly reduced density in PTEN mutant mice.
Exp Paradigm: VGLUT1, PSD95
 Immunohistochemistry
 2-3 months
Microglial number1
Increased
Description: The coronal sections of PTEN mutant mice exhibited increased accumulation of microglia compared to controls. This effect was predominantly seen at P14, though the density of microglia in the cerebral cortex remained significantly increased at P30 and 2-3 months in mutant mice.
Exp Paradigm: Iba1
 Immunohistochemistry
 P7, P14, P30, 2-3 months
Synapse density1
Decreased
Description: PTEN mutant mice exhibited a significant decrease in presynaptic marker levels (VGLUT1 and synaptophysin) as compared to control synaptosomes. Postsynaptic markers (PSD95, GluN2B, and Gephyrin), on the other hand, showed no significant differences between mutant and control synaptosomes.
 Western blot
 2-3 months
Miniature post synaptic current frequency: excitatory1
Decreased
Description: PTEN mutant mice exhibited significantly reduced frequency of miniature excitatory postsynaptic currents compared to controls, as demonstrated by whole-cell patch clamp recordings in L5 pyramidal neurons in the auditory cortex.
 Whole-cell patch clamp
 P14
Miniature post synaptic current amplitude: excitatory1
Decreased
Description: PTEN mutant mice exhibited significantly reduced amplitude of miniature excitatory postsynaptic currents compared to controls, as demonstrated by whole-cell patch clamp recordings in L5 pyramidal neurons in the auditory cortex.
 Whole-cell patch clamp
 P14
Presynaptic function: paired-pulse facilitation1
Increased
Description: PTEN mutant mice exhibited increased paired-pulse ratios in L5 pyramidal cells compared to controls. This suggests a decrease in release probability of glutamate vesicles from presynaptic terminals.
 Whole-cell patch clamp
 P14
Miniature post synaptic currents: excitatory1
Decreased
Description: PTEN mutant mice exhibited significantly decreased area of miniature excitatory postsynaptic currents compared to controls, as demonstrated by whole-cell patch clamp recordings in L5 pyramidal neurons in the auditory cortex.
 Whole-cell patch clamp
 P14
Social approach1
Decreased
Description: PTEN mutant mice spent a decreased amount of time with a companion mouse compared to controls. Controls exhibited a clear preference for interacting with a companion mouse over an inanimate object; in mutant mice, no preference existed.
 Three-chamber social approach test
 2-3 months
Social memory1
Decreased
Description: Mutant mice spent a decreased amount of time exploring a novel mouse compared to controls. Controls exhibited a clear preference for interacting with a novel mouse over a familiar mouse; in mutant mice, no preference existed.
 Three-chamber social approach test
 2-3 months
Object recognition memory1
Decreased
Description: PTEN mutant mice exhibited a decreased amount of time exploring the novel object compared to a familiar object, failing to show a preference for either. Controls, conversely, displayed a significant preference for the novel over familiar object.
 Novel object recognition test
 2-3 months
Protein expression level evidence1
Increased
Description: PTEN mutant mice microglia expressed higher levels of intracellular CD68 protein at P14 compared to controls.
Exp Paradigm: CD68
 Flow cytometric analysis
 P14
Differential gene expression1
Abnormal
Description: Mutant mice microglia exhibited an increase in the number of genes differentially expressed over time. There was an increase in genes encoding C1q components (C1qa, C1qb, and C1qc), and lysosomal KEEG pathway genes (Cd68 and those encoding cathepsins) at P14 and later. DAM signature genes, which reflect an inflammatory microglia state, were also expressed at higher levels in mutant compared to control microglia at P14 and later. Expression of microglia homeostatic genes (P2ry12, Cx3cr1, Cst3, Tmem119, Olfml3, Hexb, Fcrls, Csf1r, Tgfbr1, Mef2a, Mafb) was either unchanged or increased in mutant microglia. Finally, many mitochondria-related genes were expressed at higher levels in mutant microglia from P14 or young adult mice compared to controls.
 RNA sequencing
 P7, P14, 2-3 months
Targeted expression1
Decreased
Description: Western blotting showed that PTEN protein expression in mutant mice microglia was decreased compared to PTEN protein expression in control microglia at both P7 and at 2-3 months.
Exp Paradigm: PTEN
 Western blot
 P7, 2-3 months
Phagocytosis1
Increased
Description: PTEN mutant mice Iba1-positive microglia contained more VGLUT1 puncta within CD68-positive vesicles. This suggests that mutant microglia exhibited increased phagocytosis of VGLUT1-containing pre-synapses in early development.
Exp Paradigm: VGLUT1, CD68
 Immunohistochemistry
 P14
Targeted expression1
Decreased
Description: Intracellular staining showed that PTEN mutant microglia largely lost PTEN protein as soon as one week after the tamoxifen injection; this decrease remained consistent through one month.
Exp Paradigm: PTEN
 Flow cytometric analysis
 P7, 1 month
Phagocytosis1
Increased
Description: PTEN mutant mice exhibited increased phagocytic capability in microglia compared to controls, as evidenced by an increase in the number of zymosan particles phagocytosed in vitro.
Exp Paradigm: zymosan
 Phagocytosis assay
 P7
Protein expression: in situ protein expression1
Increased
Description: Mutant mice exhibited an increase in C1q protein in the somatosensory cortex compared to controls, as shown by immunostaining on tissue sections.
Exp Paradigm: C1q
 Immunohistochemistry
 2-3 months
Gene expression: activated microglia1
Increased
Description: Mutant mice microglia expressed higher levels of F4/80 and CD86 compared to controls.
Exp Paradigm: F4/80, CD86
 Flow cytometric analysis
 2-3 months
Anxiety1
 No change
 Open field test
 2-3 months
Thigmotaxis1
 No change
 Open field test
 2-3 months
Cell differentiation1
 No change
 RNA sequencing
 P7, P14, 2-3 months
Targeted expression1
 No change
 Immunohistochemistry
 2-3 months
General locomotor activity: ambulatory activity1
 No change
 Open field test
 2-3 months
Astrocyte number1
 No change
 Immunohistochemistry
 2-3 months
Neuronal number1
 No change
 Immunohistochemistry
 2-3 months
Number of oligodendrocytes1
 No change
 Immunohistochemistry
 2-3 months
Repetitive digging1
 No change
 Marble-burying test
 2-3 months
 Not Reported:


Interactor Symbol Interactor Name Interactor Organism Entrez ID Uniprot ID Interaction Type Evidence Reference
AKT1 v-akt murine thymoma viral oncogene homolog 1 207 P31749 IP/WB
Mistafa O , et al. 2010
AMHR2 anti-Mullerian hormone receptor, type II 269 Q16671 Affinity chromatography; MS; in silico target prediction
Crockett DK , et al. 2005
ANG angiogenin, ribonuclease, RNase A family, 5 283 P03950 Y2H
Stelzl U , et al. 2005
AR androgen receptor 367 P10275 GST; IP/WB
Lin HK , et al. 2004
ARHGAP26 Rho GTPase activating protein 26 23092 Q9UNA1 Affinity chromatography; MS; in silico target prediction
Crockett DK , et al. 2005
ATF2 activating transcription factor 2 1386 P15336 ChIP
Shen YH , et al. 2006
ATM ataxia telangiectasia mutated 472 Q13315 in vitro kinase assay
Chen JH , et al. 2015
BAP1 BRCA1 associated protein-1 (ubiquitin carboxy-terminal hydrolase) 8314 Q92560 Affinity chromatography; MS; in silico target prediction
Crockett DK , et al. 2005
Beclin-1 Beclin-1 8678 Q14457 Immunohistochemistry
Ying H , et al. 2015
BMI1 BMI1 polycomb ring finger oncogene 648 P35226 IP/WB
Fan C , et al. 2009
BRAF Serine/threonine-protein kinase B-raf 673 P15056 IP/WB
Silva JM , et al. 2014
C19orf29OS chromosome 19 open reading frame 29 opposite strand 404665 Q8N1I8 Y2H
Sakai Y , et al. 2011
CASP3 caspase 3, apoptosis-related cysteine peptidase 836 P42574 Immunodepletion assay; in vitro proteolysis assay
Torres J , et al. 2003
CASP8 caspase 8, apoptosis-related cysteine peptidase 841 Q14790 Affinity chromatography; MS; IP/WB
Crockett DK , et al. 2005
CAV1 caveolin 1, caveolae protein, 22kDa 857 Q03135 IP/WB
Caselli A , et al. 2002
CAV1 caveolin 1, caveolae protein, 22kDa 857 Q03135 IP/WB
Conde-Perez A , et al. 2015
CBP CREB-binding protein 1387 Q92793 IP/WB
Ding L , et al. 2014
CCNE2 cyclin E2 9134 O96020 Affinity chromatography; MS; IP/WB
Crockett DK , et al. 2005
CDX2 caudal type homeobox 2 1045 Q99626 Immunofluorescence; IP/WB
Liu YQ , et al. 2015
CHD8 chromodomain helicase DNA binding protein 8 57680 Q9HCK8 ChIP-chip
Subtil-Rodrguez A , et al. 2013
CHGB chromogranin B (secretogranin 1) 1114 P05060 Y2H
Stelzl U , et al. 2005
COPS6 COP9 constitutive photomorphogenic homolog subunit 6 (Arabidopsis) 10980 Q7L5N1 Y2H
Stelzl U , et al. 2005
CREB1 cAMP responsive element binding protein 1 1385 P16220 IP/WB
Duan S , et al. 2015
CREBBP CREB binding protein 1387 Q92793 IP/WB
Duan S , et al. 2015
CRKL v-crk sarcoma virus CT10 oncogene homolog (avian)-like 1399 P46109 Affinity chromatography; MS; in silico target prediction
Crockett DK , et al. 2005
CSF1 colony stimulating factor 1 (macrophage) 1435 P09603 Luciferase reporter assay
Mandal CC , et al. 2012
CSNK1E casein kinase 1, epsilon 1454 P49674 LUMIER with BACON
Shnitsar I , et al. 2015
CSNK2A1 casein kinase 2, alpha 1 polypeptide 1457 P68400 in vitro kinase assay; GST; IP/WB
Miller SJ , et al. 2002
CSNK2A2 casein kinase 2, alpha prime polypeptide 1459 P19784 in vitro kinase assay; GST; IP/WB
Miller SJ , et al. 2002
CTNNB1 catenin (cadherin-associated protein), beta 1, 88kDa 1499 Q9WU82 IP/WB
Vogelmann R , et al. 2005
CXXC1 CXXC finger protein 1 30827 Q9P0U4 Y2H; GST
Sakai Y , et al. 2011
DBF4B DBF4 homolog B (S. cerevisiae) 80174 Q8NFT6 Affinity chromatography; MS; in silico target prediction
Crockett DK , et al. 2005
DJ1 Protein deglycase DJ-1 11315 Q99497 X-ray crystallography
Choi MS , et al. 2014
DLC1 DLC1 Rho GTPase activating protein 10395 Q96QB1 IP/WB
Cao X , et al. 2015
DLG1 discs, large homolog 1 (Drosophila) 1739 Q12959 Y2H; Affinity chromatography
Adey NB , et al. 2000
DLG5 discs, large homolog 5 (Drosophila) 9231 Q8TDM6 LUMIER with BACON
Shnitsar I , et al. 2015
DVL2 dishevelled, dsh homolog 2 (Drosophila) 1856 O14641 LUMIER with BACON; IP/WB; in vitro phosphatase assay
Shnitsar I , et al. 2015
DVL3 dishevelled, dsh homolog 3 (Drosophila) 1857 Q92997 LUMIER with BACON; IP/WB
Shnitsar I , et al. 2015
EEF1A2 eukaryotic translation elongation factor 1 alpha 2 1917 Q05639 Affinity chromatography; MS; in silico target prediction
Crockett DK , et al. 2005
EGR1 early growth response 1 1958 P18146 Luciferase reporter assay; ChIP; IP/WB; Fluorescence Polarization (FP)
Tell G , et al. 2004
ELAVL1 ELAV (embryonic lethal, abnormal vision, Drosophila)-like 1 (Hu antigen R) 1994 Q15717 RNP IP; Protein microarray
Abdelmohsen K , et al. 2009
EML1 echinoderm microtubule associated protein like 1 2009 O00423 Y2H
Sakai Y , et al. 2011
EP300 E1A binding protein p300 2033 Q9Y6B2 IP/WB
Li AG , et al. 2006
EphA2 EPH receptor A2 1969 P29317 X-ray crystallography; IP/WB; in vitro kinase assay; in vitro phosphatase assay; GST
Wei Q , et al. 2014
ESR1 estrogen receptor 1 2099 P03372 GST
Lin HK , et al. 2004
FBN2 fibrillin 2 2201 P35556 Affinity chromatography; MS; in silico target prediction
Crockett DK , et al. 2005
FLNA filamin A, alpha 2316 P21333 Y2H
Sakai Y , et al. 2011
FRAT1 frequently rearranged in advanced T-cell lymphomas 10023 Q92837 LUMIER with BACON
Shnitsar I , et al. 2015
FRK fyn-related kinase 2444 P42685 IP/WB; in vitro kinase assay
Yim EK , et al. 2009
G3BP2 GTPase activating protein (SH3 domain) binding protein 2 9908 Q9UN86 Affinity chromatography; MS; in silico target prediction
Crockett DK , et al. 2005
GFRA2 GDNF family receptor alpha 2 2675 E9PD47 Affinity chromatography; MS; in silico target prediction
Crockett DK , et al. 2005
GLTSCR2 glioma tumor suppressor candidate region gene 2 29997 Q9NZM5 GST; Y2H; IP/WB
Okahara F , et al. 2004
GOPC golgi-associated PDZ and coiled-coil motif containing 57120 Q9HD26 LUMIER with BACON
Shnitsar I , et al. 2015
GPR113 G protein-coupled receptor 113 165082 Q8IZF5 Affinity chromatography; MS; in silico target prediction
Crockett DK , et al. 2005
GSK3A glycogen synthase kinase 3 alpha 2931 P49840 LUMIER with BACON
Shnitsar I , et al. 2015
GSK3B glycogen synthase kinase 3 beta 2932 P49841 Metabolic labeling with 32P; MS; WB
Al-Khouri AM , et al. 2005
HBA1 hemoglobin, alpha 1 3039 P69905 Y2H
Stelzl U , et al. 2005
HDAC4 histone deacetylase 4 9759 P56524 ChIP; Bimolecular fluorescence complementation assay; IP/WB; Y2H
Reddy SD , et al. 2012
Histone H1.2 Histone H1.2 3006 P16403 MS; FRAP; ChIP; GST; IP/WB; in vitro binding assay ; Immunofluorescence
Chen ZH , et al. 2014
HP1 alpha Chromobox protein homolog 5 23468 P45973 MS; FRAP; ChIP; GST; IP/WB; in vitro binding assay ; Immunofluorescence
Chen ZH , et al. 2014
IKBKB inhibitor of kappa light polypeptide gene enhancer in B-cells, kinase beta 3551 O14920 LUMIER with BACON
Shnitsar I , et al. 2015
INHBE inhibin, beta E 83729 P58166 Affinity chromatography; MS; in silico target prediction
Crockett DK , et al. 2005
IRS4 insulin receptor substrate 4 8471 O14654 Affinity chromatography; MS; IP/WB
Crockett DK , et al. 2005
KAT2B K(lysine) acetyltransferase 2B 8850 Q92831 IP/WB; in vitro acetylation assay
Okumura K , et al. 2006
KIF1B kinesin family member 1B 23095 O60333 LUMIER with BACON
Shnitsar I , et al. 2015
KRT14 keratin 14 3861 P02533 Affinity chromatography; MS; in silico target prediction
Crockett DK , et al. 2005
LEPREL4 leprecan-like 4 10609 Q92791 Affinity chromatography; MS; in silico target prediction
Crockett DK , et al. 2005
lncRNA-BGL3 beta globin locus transcript 3 (non-protein coding) 103344929 N/A Luciferase reporter assay
Guo G , et al. 2014
MAGI2 membrane associated guanylate kinase, WW and PDZ domain containing 2 9863 Q86UL8 Y2H; GST; IP/WB
Wu X , et al. 2000
MAGI3 membrane associated guanylate kinase, WW and PDZ domain containing 3 260425 Q5TCQ9 Y2H; GST; IP/WB
Wu Y , et al. 2000
MAP2K6 mitogen-activated protein kinase kinase 6 5608 A8K3Y2 Affinity chromatography; MS; in silico target prediction
Crockett DK , et al. 2005
MAPK14 mitogen-activated protein kinase 14 1432 Q16539 IP/WB
Ittner A , et al. 2012
Mast1 microtubule associated serine/threonine kinase 1 56527 Q9R1L5 GST; Y2H; in vitro kinase assay
Valiente M , et al. 2005
MAST2 microtubule associated serine/threonine kinase 2 23139 Q6P0Q8 Y2H; Affinity chromatography
Adey NB , et al. 2000
MAST3 microtubule associated serine/threonine kinase 3 23031 O60307 GST; Y2H; in vitro kinase assay
Valiente M , et al. 2005
MCRS1 microspherule protein 1 10445 Q96EZ8 IP/WB; GST
Okumura K , et al. 2005
MED12 mediator complex subunit 12 9968 Q93074 Affinity chromatography; MS; in silico target prediction
Crockett DK , et al. 2005
miR-106b microRNA 106b 406900 N/A Luciferase reporter assay; Immunohistochemistry
Li KK , et al. 2014
miR-130b microRNA 130b 406920 N/A qRT-PCR; IP/WB; Luciferase reporter assay
Yu T , et al. 2015
miR-21 microRNA 21 406991 N/A Luciferase reporter assay; IP/WB
Zhou X , et al. 2014
miR-221 microRNA 221 407006 IP/WB; qRT-PCR
Xie Q , et al. 2014
miR-518c microRNA 518c 574477 N/A Luciferase reporter assay; IP/WB
Tay Y , et al. 2014
miR-638 microRNA 638 693223 N/A Luciferase reporter assay; IP/WB
Tay Y , et al. 2014
miR-7 leukocyte immunoglobulin-like receptor, subfamily B (with TM and ITIM domains), member 1 10859 Q8NHL6 Luciferase reporter assay; IP/WB
Wu XN , et al. 2014
miR-802 microRNA 802 768219 N/A ChIP
Li N and Qin ZB 2014
miR-92a microRNA 29a 407021 N/A qRT-PCR
Ke TW , et al. 2014
miR-BART7 ebv-mir-BART7 (MI0003729) N/A N/A Peptide microarray; IP/WB; Luciferase reporter assay
Cai LM , et al. 2014
miR1297 microRNA 1297 100302187 N/A Luciferase reporter assay; IP/WB; MTT assay
Yang NQ , et al. 2014
miR20b microRNA 20b 574032 N/A Luciferase reporter assay; IP/WB
Zhou W , et al. 2014
MIR21 microRNA 21 406991 N/A Luciferase reporter assay
Dey N , et al. 2012
miR214 microRNA 214 406996 N/A Luciferase reporter assay; IP/WB
Zou ZJ , et al. 2014
MIR221 microRNA 221 407006 N/A Luciferase reporter assay
Zhao G , et al. 2013
miR26a microRNA 26a-1 407015 N/A Luciferase reporter assay; IP/WB
Zou ZJ , et al. 2014
MIR29C microRNA 29c 407026 N/A Luciferase reporter assay
Tumaneng K , et al. 2012
MIR374A microRNA 374a 442919 N/A Luciferase reporter assay
Cai J , et al. 2013
miR708 microRNA 708 100126333 N/A Luciferase reporter assay; IP/WB; qRT-PCR
Dileepan M , et al. 2014
MKRN1 makorin ring finger protein 1 23608 Q9UHC7 in vitro ubiquitination assay; In vivo ubiquitination assay
Lee MS , et al. 2015
MME membrane metallo-endopeptidase 4311 P08473 IP/WB; GST
Sumitomo M , et al. 2004
MPP2 membrane protein, palmitoylated 2 (MAGUK p55 subfamily member 2) 4355 Q14168 LUMIER with BACON
Shnitsar I , et al. 2015
MPRIP myosin phosphatase Rho interacting protein 23164 Q6WCQ1 Y2H
Sakai Y , et al. 2011
MTA1 metastasis associated 1 9112 Q13330 ChIP; Luciferase reporter assay; Bimolecular fluorescence complementation assay; IP/WB; Y2H
IP/WB; Immunofluorescence
Reddy SD , et al. 2012
MVP major vault protein 9961 Q14764 IP/WB; Y2H; GST
Yu Z , et al. 2002
NDFIP1 Nedd4 family interacting protein 1 80762 Q9BT67 IP/WB
Mund T and Pelham HR 2010
NDFIP2 Nedd4 family interacting protein 2 54602 B4DGY6 IP/WB
Mund T and Pelham HR 2010
NEDD4 neural precursor cell expressed, developmentally down-regulated 4 4734 P46934 WB; in vitro ubiquitination assay; GST
Wang X , et al. 2008
NKD1 naked cuticle homolog 1 (Drosophila) 85407 Q969G9 LUMIER with BACON
Shnitsar I , et al. 2015
NKD2 naked cuticle homolog 2 (Drosophila) 85409 Q969F2 LUMIER with BACON
Shnitsar I , et al. 2015
PARK7 Parkinson disease (autosomal recessive, early onset) 7 11315 Q99497 IP/WB; GST
Kim YC , et al. 2009
PAX7 paired box 7 5081 P23759 ChIP; qRT-PCR
Duan S , et al. 2015
PDGFRA platelet-derived growth factor receptor, alpha polypeptide 5156 P16234 Affinity chromatography; MS; in silico target prediction
Crockett DK , et al. 2005
PDGFRB platelet-derived growth factor receptor, beta polypeptide 5159 P09619 WB; Metabolic labeling with 32P; IP/WB; in vitro binding assay; GST
Mahimainathan L and Choudhury GG 2004
PICK1 protein interacting with PRKCA 1 9463 Q9NRD5 Y2H; GST
Sakai Y , et al. 2011
PIK3R1 phosphoinositide-3-kinase, regulatory subunit 1 (alpha) 5295 P27986 GST; IP/WB
Ittner A , et al. 2012
PIK3R1 phosphoinositide-3-kinase, regulatory subunit 1 (alpha) 5295 P27986 IP/WB
Cao X , et al. 2015
PINK1 PTEN induced putative kinase 1 65018 Q9BXM7 Affinity chromatography; MS; in silico target prediction
Crockett DK , et al. 2005
PKN2 protein kinase N2 5586 Q16513 Affinity chromatography; MS; in silico target prediction
Crockett DK , et al. 2005
Plk1 polo-like kinase 1 5347 P53350 IP/WB; GST; In vivo ubiquitination assay; LC/MS
Li Z , et al. 2014
PPM1B protein phosphatase, Mg2+/Mn2+ dependent, 1B 5495 O75688 LUMIER with BACON
Shnitsar I , et al. 2015
PPP1CA protein phosphatase 1, catalytic subunit, alpha isozyme 5499 P62136 Antibody Microarray; IP/WB
Flores-Delgado G , et al. 2007
PPP1R10 protein phosphatase 1, regulatory subunit 10 5514 Q96QC0 IP/WB; GST
Kavela S , et al. 2012
PPP2R4 protein phosphatase 2A activator, regulatory subunit 4 5524 Q15257 Affinity chromatography; MS; IP/WB
Crockett DK , et al. 2005
PPP3CA protein phosphatase 3, catalytic subunit, alpha isozyme 5530 Q08209 IP/WB
Mistafa O , et al. 2010
PTK2 PTK2 protein tyrosine kinase 2 5747 Q05397 WB
Tamura M , et al. 1998
PTK2B PTK2B protein tyrosine kinase 2 beta 2185 Q14289 Affinity chromatography; MS; in silico target prediction
Crockett DK , et al. 2005
PTPN14 protein tyrosine phosphatase, non-receptor type 14 5784 A8K6H6 Affinity chromatography; MS; in silico target prediction
Crockett DK , et al. 2005
PTPN6 protein tyrosine phosphatase, non-receptor type 6 5777 P29350 IP/WB
Lu Y , et al. 2003
PXN paxillin 5829 P49023 IP/WB
Haier J and Nicolson GL 2002
QRFPR pyroglutamylated RFamide peptide receptor 84109 Q96P65 Affinity chromatography; MS; in silico target prediction
Crockett DK , et al. 2005
RNF146 ring finger protein 146 81847 Q9NTX7 IP/WB
Li N , et al. 2014
RPL14 ribosomal protein L14 9045 P50914 Affinity chromatography; MS; in silico target prediction
Crockett DK , et al. 2005
RPS6KB1 ribosomal protein S6 kinase, 70kDa, polypeptide 1 6198 P23443 IP/WB
Liu JL , et al. 2007
RPS6KB2 ribosomal protein S6 kinase, 70kDa, polypeptide 2 6199 Q9UBS0 IP/WB
Liu JL , et al. 2007
RYBP RING1 and YY1 binding protein 23429 Q8N488 Y2H
Sakai Y , et al. 2011
SHARPIN SHANK-associated RH domain interactor 81858 Q9H0F6 IP/WB
He L , et al. 2010
SLC9A3R1 solute carrier family 9 (sodium/hydrogen exchanger), member 3 regulator 1 9368 O14745 Y2H; Blot overlay assay; GST
Takahashi Y , et al. 2006
SLC9A3R2 solute carrier family 9 (sodium/hydrogen exchanger), member 3 regulator 2 9351 Q6NTG0 Blot overlay assay; GST; IP/WB; Immunodepletion assay
Takahashi Y , et al. 2006
SLUG Zinc finger protein SNAI2 6591 O43623 qRT-PCR; Luciferase reporter assay; IP/WB; ChIP
Uygur B , et al. 2015
SMAD2 SMAD family member 2 4087 Q15796 IP/WB
Hjelmeland AB , et al. 2005
SMAD3 SMAD family member 3 4088 P84022 IP/WB
Hjelmeland AB , et al. 2005
SMTN smoothelin 6525 B4E229 Affinity chromatography; MS; in silico target prediction
Crockett DK , et al. 2005
SNAI1 snail homolog 1 (Drosophila) 6615 O95863 Luciferase reporter assay; EMSA; ChIP
Escriv M , et al. 2008
SNRPN small nuclear ribonucleoprotein polypeptide N 6638 P63162 LUMIER with BACON
Shnitsar I , et al. 2015
SP1 Sp1 transcription factor 6667 P08047 Luciferase reporter assay; ChIP
Kou XX , et al. 2012
STK11 serine/threonine kinase 11 6794 Q15831 Y2H; IP/WB; in vitro kinase assay
Mehenni H , et al. 2005
STK11 serine/threonine kinase 11 6794 Q15831 LUMIER with BACON
Shnitsar I , et al. 2015
STUB1 STIP1 homology and U-box containing protein 1, E3 ubiquitin protein ligase 10273 Q9UNE7 IP/WB; GST; in vitro ubiquitination assay
Ahmed SF , et al. 2012
SUMO1 SMT3 suppressor of mif two 3 homolog 1 (S. cerevisiae) 7341 P63165 in vitro SUMOylation assay; IP/WB
Gonzlez-Santamara J , et al. 2012
SUMO2 SMT3 suppressor of mif two 3 homolog 2 (S. cerevisiae) 6613 P61956 in vitro SUMOylation assay; IP/WB
Gonzlez-Santamara J , et al. 2012
TCEB3C transcription elongation factor B polypeptide 3C (elongin A3) 162699 Q8NG57 Affinity chromatography; MS; in silico target prediction
Crockett DK , et al. 2005
TNKS1 Tankyrase-1 8658 O95271 IP/WB; PARP assay
Li N , et al. 2014
TNKS2 tankyrase, TRF1-interacting ankyrin-related ADP-ribose polymerase 2 80351 Q9H2K2 IP/WB; PARP assay
Li N , et al. 2014
TP53 tumor protein p53 7157 P04637 IP/WB; EMSA; ChIP
Freeman DJ , et al. 2003
TTBK2 tau tubulin kinase 2 146057 Q6IQ55 Affinity chromatography; MS; in silico target prediction
Crockett DK , et al. 2005
UBC ubiquitin C 7316 P63279 WB
Wu W , et al. 2003
UBE2I ubiquitin-conjugating enzyme E2I (UBC9 homolog, yeast) 7329 P63279 IP/WB
Waite KA and Eng C 2003
UBE2L3 ubiquitin-conjugating enzyme E2L 3 7332 P68036 IP/WB
Waite KA and Eng C 2003
UTP14A UTP14, U3 small nucleolar ribonucleoprotein, homolog A (yeast) 10813 Q9BVJ6 Y2H
Stelzl U , et al. 2005
WNT4 wingless-type MMTV integration site family, member 4 54361 P56705 Affinity chromatography; MS; in silico target prediction
Crockett DK , et al. 2005
WWP2 WW domain containing E3 ubiquitin protein ligase 2 11060 B4DHF6 Affinity chromatography; MS; in silico target prediction
Crockett DK , et al. 2005
XIAP X-linked inhibitor of apoptosis 331 P98170 WB; IP/WB; in vitro ubiquitination assay
Van Themsche C , et al. 2009
YAF2 YY1 associated factor 2 10138 Q8IY57 Y2H
Sakai Y , et al. 2011
YY1 YY1 transcription factor 7528 P25490 ChIP; EMSA; Bimolecular fluorescence complementation assay; IP/WB; Y2H
Reddy SD , et al. 2012
ZNF787 zinc finger protein 787 126208 Q6DD87 Affinity chromatography; MS; in silico target prediction
Crockett DK , et al. 2005
Aebp1 AE binding protein 1 11568 Q640N1 IP/WB
Ro HS , et al. 2007
Arx aristaless related homeobox 11878 O35085 ChIP-qPCR
Quill ML , et al. 2011
BMP9 Growth/differentiation factor 2 12165 Q9WV56 Luciferase reporter assay; IP/WB; in vitro phosphatase assay; Immunohistochemistry
Huang J , et al. 2014
Cenpc1 centromere protein C1 12617 P49452 IP/WB
Shen WH , et al. 2007
FMR1 fragile X mental retardation 1 14265 P35922 HITS-CLIP
Darnell JC , et al. 2011
FOXP1 forkhead box P1 108655 P58462 ChIP
Tang B , et al. 2012
Htr2c 5-hydroxytryptamine (serotonin) receptor 2C 15560 P34968 Proximity ligation assay; IP/WB
Kleene R , et al. 2015
Itga3 integrin alpha 3 16400 Q62470 Immunohistochemistry; IP/WB
Yazlovitskaya EM , et al. 2015
Itgb1 Integrin beta-1 16412 P09055 Immunohistochemistry; IP/WB
Yazlovitskaya EM , et al. 2015
Mir17hg Mir17 host gene (non-protein coding) 75957 Luciferase reporter assay
Tung YT , et al. 2015
Mir19a microRNA 19a 723891 Luciferase reporter assay
Zhang L , et al. 2015
Ndfip1 NEDD4 family-interacting protein 1 65113 Q8R0W6 Immunohistochemistry; IP/WB
Howitt J , et al. 2015
Stat3 signal transducer and activator of transcription 3 20848 P42227 ChIP
Zha X , et al. 2011
cyclin D1 G1/S-specific cyclin-D1 595 P24385 IP/WB
Gao L , et al. 2014
Grin2b glutamate receptor, ionotropic, N-methyl D-aspartate 2B 24410 Q00960 IP/WB; in vitro binding assay
Ning K , et al. 2004
Htr2c 5-hydroxytryptamine (serotonin) receptor 2C, G protein-coupled 25187 P08909 IP/WB
Ji SP , et al. 2006
miR-29a microRNA mir-29a 100314230 N/A qRT-PCR; IP/WB; Luciferase reporter assay; Immunofluorescence
Zou H , et al. 2015
p21 cyclin-dependent kinase inhibitor 1A (p21, Cip1) 1026 P38936 IP/WB
Gao L , et al. 2014
EHMT1 G9a 30971 Q95RU8 ChIP-Seq
Kramer JM , et al. 2011
Cbl N/A 100754737 N/A IP/WB
Rodriguez S and Huynh-Do U 2012
PTPRZ1 N/A 100511294 N/A IP/WB; in vitro phosphatase assay
Shen X , et al. 2012

HELP
Copyright © 2017 MindSpec, Inc.