HELP     Sign In
Search

Relevance to Autism

Two de novo LoF variants in the TCF7L2 gene (both splice-site) were identified in ASD probands from the Simons Simplex Collection (PMID 25363768). TADA analysis of de novo variants from the Simons Simplex Collection and the Autism Sequencing Consortium and protein-truncating variants from iPSYCH in Satterstrom et al., 2020 identified TCF7L2 as a candidate gene with a false discovery rate (FDR) between 0.01 and 0.05 (0.01 < FDR 0.05). Dias et al., 2021 reported 11 individuals with de novo TCF7L2 variants presenting with a syndromic neurodevelopmental disorder; autism spectrum disorder was reported in four of these individuals.

Molecular Function

This gene encodes a high mobility group (HMG) box-containing transcription factor that plays a key role in the Wnt signaling pathway. The protein has been implicated in blood glucose homeostasis. Genetic variants of this gene are associated with increased risk of type 2 diabetes (Diabetes mellitus, non-insulin-dependent (NIDDM) [MIM:125853]).

External Links

        

References

Type
Title
Type of Disorder
Associated Disorders
Author, Year
Primary
The contribution of de novo coding mutations to autism spectrum disorder
ASD
Support
Meta-analysis of 2,104 trios provides support for 10 new genes for intellectual disability
ID
Support
Large-scale discovery of novel genetic causes of developmental disorders.
Unknown diagnosis
Support
Mutational Landscape of Autism Spectrum Disorder Brain Tissue
ASD
Support
Large-scale targeted sequencing identifies risk genes for neurodevelopmental disorders
ASD
DD, ID
Support
Large-Scale Exome Sequencing Study Implicates Both Developmental and Functional Changes in the Neurobiology of Autism
ASD
Support
Inherited and multiple de novo mutations in autism/developmental delay risk genes suggest a multifactorial model.
ASD
Support
NA
ASD
Support
Prevalence and architecture of de novo mutations in developmental disorders
Developmental disorders
Recent Recommendation
ASD, DD, ID
Recent Recommendation
De novo variants in TCF7L2 are associated with a syndromic neurodevelopmental disorder
DD
ASD, ADHD, ID

Rare

Variant ID
Variant Type
Allele Change
Residue Change
Inheritance Pattern
Inheritance Association
Family Type
Author, Year
 GEN652R001 
 splice_site_variant 
 c.1001+1G>A 
  
 De novo 
  
 Simplex 
 GEN652R002 
 splice_site_variant 
 c.685+1G>A 
  
 De novo 
  
 Simplex 
 GEN652R003 
 stop_gained 
 c.1219C>T 
 p.Arg407Ter 
 De novo 
  
 Unknown 
 GEN652R004 
 stop_gained 
 c.1804G>T 
 p.Glu602Ter 
 Familial 
 Paternal 
 Simplex 
 GEN652R005 
 intron_variant 
 c.552+49423C>T 
  
 De novo 
  
 Simplex 
 GEN652R006 
 splice_site_variant 
 c.451-1G>C 
  
 De novo 
  
  
 GEN652R007 
 missense_variant 
 c.1243C>G 
 p.Pro415Ala 
 De novo 
  
  
 GEN652R008 
 missense_variant 
 c.1319-636G>A 
  
 De novo 
  
  
 GEN652R009 
 missense_variant 
 c.1412G>T 
 p.Arg471Leu 
 Unknown 
  
  
 GEN652R010 
 missense_variant 
 c.836C>T 
 p.Pro279Leu 
 Unknown 
  
  
 GEN652R011 
 missense_variant 
 c.866C>T 
 p.Ser289Phe 
 Unknown 
  
  
 GEN652R012 
 missense_variant 
 c.971C>T 
 p.Ser324Phe 
 Unknown 
  
 Simplex 
 GEN652R013 
 missense_variant 
 c.1643C>A 
 p.Ser548Tyr 
 Unknown 
  
  
 GEN652R014 
 missense_variant 
 c.1694C>A 
 p.Ser565Tyr 
 Unknown 
  
  
 GEN652R015 
 missense_variant 
 c.1763C>T 
 p.Ser588Leu 
 Unknown 
  
  
 GEN652R016 
 missense_variant 
 c.526C>T 
 p.Arg176Trp 
 Unknown 
  
  
 GEN652R017 
 missense_variant 
 c.1211G>A 
 p.Arg404Gln 
 Unknown 
  
  
 GEN652R018 
 missense_variant 
 c.490G>A 
 p.Gly164Arg 
 Unknown 
  
  
 GEN652R019 
 missense_variant 
 c.1097C>A 
 p.Ala366Glu 
 Unknown 
  
  
 GEN652R020 
 missense_variant 
 c.1427A>G 
 p.Glu476Gly 
 De novo 
  
  
 GEN652R021 
 missense_variant 
 c.1143C>G 
 p.Asn381Lys 
 De novo 
  
  
 GEN652R022 
 missense_variant 
 c.881C>T 
 p.Pro294Leu 
 De novo 
  
  
 GEN652R023 
 stop_gained 
 c.1219C>T 
 p.Arg407Ter 
 De novo 
  
  
 GEN652R024 
 splice_site_variant 
 c.553-1G>A 
  
 De novo 
  
 Simplex 
 GEN652R025 
 stop_gained 
 c.1269T>G 
 p.Tyr423Ter 
 De novo 
  
 Simplex 
 GEN652R026 
 frameshift_variant 
 c.787del 
 p.Gln263SerfsTer22 
 De novo 
  
  
 GEN652R027 
 stop_gained 
 c.1144C>T 
 p.Gln382Ter 
 De novo 
  
  
 GEN652R028 
 frameshift_variant 
 c.660dup 
 p.Pro221ThrfsTer107 
 De novo 
  
 Simplex 
 GEN652R029 
 splice_site_variant 
 c.875+1G>C 
  
 De novo 
  
 Simplex 
 GEN652R030 
 missense_variant 
 c.1143C>G 
 p.Asn381Lys 
 De novo 
  
  
 GEN652R031 
 missense_variant 
 c.1142A>C 
 p.Asn381Thr 
 De novo 
  
 Simplex 
 GEN652R032 
 missense_variant 
 c.1250G>T 
 p.Trp417Leu 
 De novo 
  
 Simplex 
 GEN652R033 
 missense_variant 
 c.1267T>C 
 p.Tyr423His 
 De novo 
  
 Simplex 
 GEN652R034 
 missense_variant 
 c.1268A>G 
 p.Tyr423Cys 
 De novo 
  
  
 GEN652R035 
 synonymous_variant 
 c.666C>T 
 p.Ala222%3D 
 Unknown 
  
  
 GEN652R036 
 missense_variant 
 c.595C>G 
 p.Pro199Ala 
 De novo 
  
 Simplex 
 GEN652R037a 
 missense_variant 
 c.536C>A 
 p.Ser179Ter 
 Familial 
 Both parents 
  

Common

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

Model Summary

Tcf7l2 is a transcription factor that was identified in a forward genetic screen for ultrasonic vocalization deficits. The mutant identified in the screen exhibits vocalization deficits in pups (isolation-induced) and adults (opposite sex interaction-induced). The mutation affects the number and complexity of ultrasonic vocalizations. Passive social behavior is decreased due to lack of vocalizations. The heterozygous null mutant shows deficits in miniature post synaptic currents in the periaqueductal grey neurons. Humanized mutations found in ASD/NDD probands recapitulate vocalization deficits. There are no social or repetitive behavior deficits in the mutants studied.

References

Type
Title
Author, Year
Additional
NA

M_TCF7L2_10_KI_HT

Model Type: Genetic
Model Genotype: Heterozygous
Mutation: The knockin deletion of exon 5' was generated using CRISPR/Cas9 technology to remove alternative exons 5'-2, 5'-3, 5'-4, 5'-5 through 5'-9.
Allele Type: Knockin
Strain of Origin:
Genetic Background: C57BL/6J
ES Cell Line:
Mutant ES Cell Line:
Model Source: Yichang Jia lab (PMID 36782064)

M_TCF7L2_11_KI_HM

Model Type: Genetic
Model Genotype: Homozygous
Mutation: The knockin deletion of exon 5' was generated using CRISPR/Cas9 technology to remove alternative exons 5'-2, 5'-3, 5'-4, 5'-5 through 5'-9.
Allele Type: Knockin
Strain of Origin:
Genetic Background: C57BL/6J
ES Cell Line:
Mutant ES Cell Line:
Model Source: Yichang Jia lab (PMID 36782064)

M_TCF7L2_12_KI_HT_EXON9-5SSV

Model Type: Genetic
Model Genotype: Heterozygous
Mutation: The humanized knockin mutation c.932 + 1 G > A mutation alters the 5' splicing site (GT) of Tcf7l2 exon 9 to AT, potentially preventing correct splicing from exon 9 to exon 10. This mutation is identified in an ASD proband in Iossifov et al. 2014 (PMID 25363768).
Allele Type: ASD mutation
Strain of Origin:
Genetic Background: C57BL/6J
ES Cell Line:
Mutant ES Cell Line:
Model Source: Yichang Jia lab (PMID 36782064)

M_TCF7L2_13_KI_HT_R384X

Model Type: Genetic
Model Genotype: Heterozygous
Mutation: The humanized knockin mutation c.1150 C > T (R384X) mutation introduces a premature termination codon in Tcf7l2 exon 11. This mutation is identified as a developmental delay mutation in a large-scale discovery study in Deciphering Developmental Disorders, 2015 (PMID 25533962).
Allele Type: NDD mutation
Strain of Origin:
Genetic Background: C57BL/6J
ES Cell Line:
Mutant ES Cell Line:
Model Source: Yichang Jia lab (PMID 36782064)

M_TCF7L2_1_KI_HT

Model Type: Genetic
Model Genotype: Heterozygous
Mutation: The Y337H knockin mutation was identified in an ENU-mutagenesis screen for causing impaired ultrasonic vocalizations. This non-synonymous mutation in the Tcf7l2 gene (c.T1019C, p.Y337H) is always co-segregated with USV impairment.
Allele Type: Knockin
Strain of Origin:
Genetic Background: C57BL/6J
ES Cell Line:
Mutant ES Cell Line:
Model Source: Yichang Jia lab (PMID 36782064)

M_TCF7L2_2_KO_HT

Model Type: Genetic
Model Genotype: Heterozygous
Mutation: The knockout mutation was generated with CRISPR/Cas9 technology by introducing a two-nucleotide deletion in exon 10.
Allele Type: Knockout
Strain of Origin:
Genetic Background: C57BL/6J
ES Cell Line:
Mutant ES Cell Line:
Model Source: Yichang Jia lab (PMID 36782064)

M_TCF7L2_3_CKO_HT

Model Type: Genetic
Model Genotype: Heterozygous
Mutation: The neuronal-specific conditional knockout allele was generated by crossing a conditional ready mouse line, where exon 11 of the Tcf7l2 gene is flanked by loxP sites (MGI:5430317), with a mouse line expressing the Nestin-Cre transgenic construct (MGI:2176173).
Allele Type: Conditional knockout
Strain of Origin:
Genetic Background: C57BL/6J
ES Cell Line:
Mutant ES Cell Line:
Model Source: Richard Liu lab; Jackson Laboratories

M_TCF7L2_4_CKO_HT

Model Type: Genetic
Model Genotype: Heterozygous
Mutation: The Vglut2-specific conditional knockout allele was generated by crossing a conditional ready mouse line, where exon 11 of the Tcf7l2 gene is flanked by loxP sites (MGI:5430317), with a mouse line expressing the Vglut2-Cre transgenic construct (MGI:5141269).
Allele Type: Conditional knockout
Strain of Origin:
Genetic Background: C57BL/6J
ES Cell Line:
Mutant ES Cell Line:
Model Source: Richard Liu lab; Jackson Laboratories

M_TCF7L2_5_CKO_HM

Model Type: Genetic
Model Genotype: Homozygous
Mutation: The Vglut2-specific conditional knockout allele was generated by crossing a conditional ready mouse line, where exon 11 of the Tcf7l2 gene is flanked by loxP sites (MGI:5430317), with a mouse line expressing the Vglut2-Cre transgenic construct (MGI:5141269).
Allele Type: Conditional knockout
Strain of Origin:
Genetic Background: C57BL/6J
ES Cell Line:
Mutant ES Cell Line:
Model Source: Richard Liu lab; Jackson Laboratories

M_TCF7L2_6_KO_HT

Model Type: Genetic
Model Genotype: Heterozygous
Mutation: The fx knockout allele for Tcf7l2 was generated with CRISPR/Cas9 technology, by inverting exons 9 and 10, and placing the inward-facing Lox2272:LoxP sites flanking the two inverted exons. Tcf7l2 expression can be switched on in a Cre-dependent manner. In the absence of Cre, Tcf7l2 is not expressed.
Allele Type: Knockout
Strain of Origin:
Genetic Background: C57BL/6J
ES Cell Line:
Mutant ES Cell Line:
Model Source: Yichang Jia lab (PMID 36782064)

M_TCF7L2_7_CKO_HM

Model Type: Genetic
Model Genotype: Homozygous
Mutation: The midbrain-specific conditional knockout allele was generated by injecting a conditional ready mouse line, where exon 11 of the Tcf7l2 gene is flanked by loxP sites (MGI:5430317) with an AAV-mCherry-Cre construct at age P0. Controls were injected with an AAV-mCherry construct.
Allele Type: Conditional knockout
Strain of Origin:
Genetic Background: C57BL/6J
ES Cell Line:
Mutant ES Cell Line:
Model Source: Richard Liu lab

M_TCF7L2_8_CKO_HM

Model Type: Genetic
Model Genotype: Homozygous
Mutation: The midbrain-specific conditional knockout allele was generated by injecting a conditional ready mouse line, where exon 11 of the Tcf7l2 gene is flanked by loxP sites (MGI:5430317) with an AAV-mCherry-Cre construct at age P17. Controls were injected with an AAV-mCherry construct.
Allele Type: Conditional knockout
Strain of Origin:
Genetic Background: C57BL/6J
ES Cell Line:
Mutant ES Cell Line:
Model Source: Richard Liu lab

M_TCF7L2_9_CKI_HM

Model Type: Genetic
Model Genotype: Homozygous
Mutation: The Vglut2-specific conditional knockin allele was generated by crossing a conditional ready mouse line, where exon 1 of the Tcf7l2 gene is flanked by loxP sites (MGI:4946910), with a mouse line expressing the Vglut2-Cre transgenic construct (MGI:5141269).
Allele Type: Conditional knockin
Strain of Origin: (129S6/SvEvTac x C57BL/6NCrl)F1
Genetic Background: C57BL/6J
ES Cell Line:
Mutant ES Cell Line:
Model Source: Jackson Laboratories

M_TCF7L2_14_CKO_HM

Model Type: Genetic
Model Genotype: Homozygous
Mutation: To generate the Aldh1l1Cre-ERT2:Tcf7l2^fl/fl strain, in which Tcf7l2 knockout is induced in astrocytes after the administration of tamoxifen (75 mg/kg body weight), Tcf7l2^tm1a/+ (MGI:4431951) animals were first crossed with flippase-expressing allele (MGI:2429412) and then with the inducible Cre recombinase-expressing allele (MGI:5806568) under the astrocyte-specific Aldh1/1 promoter.
Allele Type: Conditional knockout
Strain of Origin: C57BL/6N; 129S4/SvJaeSor; FVB/N
Genetic Background: C57BL/6NTac
ES Cell Line: JM8.N4; AK7
Mutant ES Cell Line:
Model Source: Jackson laboratories

M_TCF7L2_15_CKO_HM

Model Type: Genetic
Model Genotype: Homozygous
Mutation: To determine the cell-autonomous impact of TCF7L2 on astrocyte morphology, the adeno-associated virus (AAV)-mediated delivery of CRISPR/Cas9 was used to delete Tcf7l2. On P2, AAVs were intraventricularly injected to express Cre under the astrocyte-specific gfaABC1D promoter and two guide RNAs under the U6 promoter.
Allele Type: Conditional knockout
Strain of Origin: Not applicable
Genetic Background: Not specified
ES Cell Line: Not applicable
Mutant ES Cell Line:
Model Source:

M_TCF7L2_10_KI_HT

Category
Entity
Quantity
Experimental Paradigm
Age at Testing
Targeted expression1
Abnormal
Description: Mutant mice exhibited reduced expression of the short TCF7L2 form and no change in the long TCF7L2 form in the midbrain as compared to wildtype control mice.
 Western blot
 P7
Ultrasonic vocalization: isolation induced1
 No change
 Monitoring ultrasonic vocalizations
 P7
 Not Reported:

M_TCF7L2_11_KI_HM

Category
Entity
Quantity
Experimental Paradigm
Age at Testing
Ultrasonic vocalization: isolation induced1
Decreased
Description: Mutant mice exhibited significantly decreased syllable number, mean frequency (kHz), peak syllable amplitude (dB), and syllable duration (ms) compared to wildtype control mice, indicating that brain-specific dnTCF7L2 is required for mouse ultrasonic vocalization production.
 Monitoring ultrasonic vocalizations
 P7
Targeted expression1
Abnormal
Description: Mutant mice exhibited absent expression of the short TCF7L2 form and no change in the long TCF7L2 form in the midbrain as compared to wildtype control mice.
 Western blot
 P7
 Not Reported:

M_TCF7L2_12_KI_HT_EXON9-5SSV

Category
Entity
Quantity
Experimental Paradigm
Age at Testing
Ultrasonic vocalization: isolation induced1
Decreased
Description: Mutant mice exhibited significantly decreased syllable number, mean frequency (kHz), peak syllable amplitude (dB), and syllable duration (ms) compared to wildtype control mice.
 Monitoring ultrasonic vocalizations
 P7
Targeted expression1
Decreased
Description: Mutant mice exhibited decreased midbrain expression of TCF7L2 compared to wildtype control levels.
 Western blot
 P7
 Not Reported:

M_TCF7L2_13_KI_HT_R384X

Category
Entity
Quantity
Experimental Paradigm
Age at Testing
Ultrasonic vocalization: isolation induced1
Decreased
Description: Mutant mice exhibited significantly decreased syllable number, mean frequency (kHz), peak syllable amplitude (dB), and syllable duration (ms) compared to wildtype control mice.
 Monitoring ultrasonic vocalizations
 P7
Targeted expression1
Decreased
Description: Mutant mice exhibited decreased midbrain expression of TCF7L2 compared to wildtype control levels.
 Western blot
 P7
 Not Reported:

M_TCF7L2_1_KI_HT

Category
Entity
Quantity
Experimental Paradigm
Age at Testing
Passive social behavior1
Decreased
Description: ENU-induced mutant male mice reduced both the sniffing time (s) and preference index (%) of wildtype female mice, compared to the female sniffing time and preference index in response to wildtype males.
 Three-chamber social approach test
 3 months
Ultrasonic vocalization: interaction induced: opposite sex stimulus: complex syllables1
Decreased
Description: ENU-induced mutant male mice produced significantly fewer 'multiple' syllables in the presence of a virgin female (anesthetized and awake), as well as significantly fewer 'upward' syllables (awake only), compared to wildtype control virgin males.
 Monitoring ultrasonic vocalizations
 3 months
Ultrasonic vocalization: isolation induced1
Decreased
Description: ENU-induced mutant mice exhibited multiple ultrasonic vocalization abnormalities compared to wildtype controls, including significantly fewer syllable numbers, lower peak amplitude (dB), and reduced mean frequency (kHz). Mutant mice displayed no change in syllable duration (ms) compared to control mice.
Exp Paradigm: MUPET (Mouse Ultrasonic Profile ExTraction)
 Monitoring ultrasonic vocalizations
 P7
Ultrasonic vocalization: interaction induced: opposite sex stimulus1
Decreased
Description: ENU-induced mutant male mice exhibited multiple ultrasonic vocalization abnormalities when in the presence of wildtype virgin females compared to wildtype controls. These include significantly fewer syllable numbers (anesthetized and awake female), lower peak amplitude (dB) (awake female only), and reduced syllable duration (ms) (anesthetized and awake female). Mutant male mice displayed no change in mean frequency (kHz) compared to wildtype control mice.
 Monitoring ultrasonic vocalizations
 3 months
Ultrasonic vocalization: interaction induced: opposite sex stimulus: syllable transition1
Decreased
Description: ENU-induced mutant male mice exhibited impaired syllable transitions in the presence of both a anesthetized and awake female mice, compared to wildtype control male mice.
 Monitoring ultrasonic vocalizations
 3 months
Ultrasonic vocalization: interaction induced: opposite sex stimulus: simple syllables1
Increased
Description: ENU-induced mutant male mice produced significantly more 'simple' syllables in the presence of a virgin female (anesthetized and awake) compared to wildtype control males.
 Monitoring ultrasonic vocalizations
 3 months
Anxiety1
 No change
 Open field test
 3 months
General locomotor activity: ambulatory activity1
 No change
 Open field test
 3 months
Motor coordination and balance1
 No change
 Accelerating rotarod test
 3 months
Brain morphology1
 No change
 Histology
 P7, 4 months
Repetitive digging1
 No change
 Marble-burying test
 2 months
Self grooming1
 No change
 Grooming behavior assessments
 2-3 months
Social approach1
 No change
 Three-chamber social approach test
 3 months
 Not Reported:

M_TCF7L2_2_KO_HT

Category
Entity
Quantity
Experimental Paradigm
Age at Testing
Miniature post synaptic current frequency: excitatory1
Decreased
Description: Tcf7l2 heterozygous mutant mice exhibited significantly decreased miniature excitatory postsynaptic current (mEPSC) frequency (Hz) in lateral periaqueductal gray (LPAG) neurons compared to wildtype control mice.
 Whole-cell current clamp
 P25-P30
Miniature post synaptic current amplitude: inhibitory1
Decreased
Description: Tcf7l2 heterozygous mutant mice exhibited significantly decreased miniature inhibitory postsynaptic current (mIPSC) amplitude (pA) in lateral periaqueductal gray (LPAG) neurons compared to wildtype control mice.
 Whole-cell current clamp
 P25-P30
Passive social behavior1
Decreased
Description: Tcf7l2 heterozygous mutant male mice reduced both the sniffing time (s) and preference index (%) of wildtype female mice, compared to the female sniffing time and preference index in response to wildtype males.
 Three-chamber social approach test
 3 months
Ultrasonic vocalization: isolation induced1
Decreased
Description: Tcf7l2 heterozygous mutant male mice exhibited impaired ultrasonic vocalization generation, including a significantly decreased number of syllables, reduced peak amplitude (dB), and decreased mean frequency (kHz) compared to wildtype controls.
 Monitoring ultrasonic vocalizations
 P7
Ultrasonic vocalization: interaction induced: opposite sex stimulus1
Decreased
Description: Tcf7l2 heterozygous mutant virgin male mice displayed a significantly decreased number of syllables, decreased peak syllable amplitude (dB), and lower mean frequency (kHz) with awake wildtype females compared to wildtype virgin males.
 Monitoring ultrasonic vocalizations
 3 months
Targeted expression1
Decreased
Description: Tcf7l2 heterozygous mutant mice exhibited reduced expression of high and low molecular weight TCF7L2 in the midbrain compared to wildtype controls.
 Western blot
 P7
Action potential property: after hyperpolarization1
 No change
 Whole-cell current clamp
 P25-P30
Action potential property: amplitude1
 No change
 Whole-cell current clamp
 P25-P30
Action potential property: firing pattern1
 No change
 Whole-cell current clamp
 P25-P30
Action potential property: firing rate1
 No change
 Whole-cell current clamp
 P25-P30
Action potential property: half-width1
 No change
 Whole-cell current clamp
 P25-P30
Miniature post synaptic current amplitude: excitatory1
 No change
 Whole-cell current clamp
 P25-P30
Miniature post synaptic current frequency: inhibitory1
 No change
 Whole-cell current clamp
 P25-P30
Repetitive digging1
 No change
 Marble-burying test
 2 months
Self grooming1
 No change
 Grooming behavior assessments
 2-3 months
Social approach1
 No change
 Three-chamber social approach test
 3 months
 Not Reported:

M_TCF7L2_3_CKO_HT

Category
Entity
Quantity
Experimental Paradigm
Age at Testing
Ultrasonic vocalization: isolation induced1
Decreased
Description: Tcf7l2 conditional heterozygous mutant mice displayed significantly fewer syllables, reduced peak amplitude (dB), lower mean frequency (kHz), and shorter syllable duration (ms) compared to control mice, indicating that Tcf7l2 expression in neuronal progenitors is required for ultrasonic vocalizations.
 Monitoring ultrasonic vocalizations
 P7
Targeted expression1
Decreased
Description: Tcf7l2 conditional heterozygous mutant mice exhibited reduced expression of high and low molecular weight TCF7L2 in the midbrain compared to control mice.
 Western blot
 P7
 Not Reported:

M_TCF7L2_4_CKO_HT

Category
Entity
Quantity
Experimental Paradigm
Age at Testing
Ultrasonic vocalization: isolation induced1
Decreased
Description: Mutant mice displayed significantly fewer syllables, reduced peak amplitude (dB), and shorter syllable duration (ms) compared to control mice, indicating that expression of Tcf7l2 in Vglut2-positive neurons is necessary for ultrasonic vocalizations.
 Monitoring ultrasonic vocalizations
 P7
Targeted expression1
Decreased
Description: Mutant mice exhibited reduced expression of high and low molecular weight TCF7L2 in the midbrain compared to control mice.
 Western blot
 P7
 Not Reported:

M_TCF7L2_5_CKO_HM

Category
Entity
Quantity
Experimental Paradigm
Age at Testing
Differential gene expression1
Abnormal
Description: Mutant mice exhibited several altered biological pathways in the periaqueductal gray (PAG) region relative to control mice.
 RNA sequencing
 P7
 Not Reported:

M_TCF7L2_6_KO_HT

Category
Entity
Quantity
Experimental Paradigm
Age at Testing
Ultrasonic vocalization: isolation induced1
Decreased
Description: In the absence of Cre, mutant mice displayed significantly fewer syllables, reduced peak amplitude (dB), lower mean frequency (kHz), and shorter syllable duration (ms) compared to wildtype controls.
 Monitoring ultrasonic vocalizations
 P7
Targeted expression1
Decreased
Description: In the absence of Cre, mutant mice exhibited reduced expression of both short and long TCF7L2 in the midbrain compared to wildtype controls.
 Western blot
 P7
 Not Reported:

M_TCF7L2_7_CKO_HM

Category
Entity
Quantity
Experimental Paradigm
Age at Testing
Ultrasonic vocalization: isolation induced1
Decreased
Description: Mutant mice displayed significantly fewer syllables compared to control mice, and no change in peak syllable amplitude (dB), syllable duration (ms), or mean frequency (kHz).
 Monitoring ultrasonic vocalizations
 P7
Targeted expression1
Abnormal
Description: Mutant mice exhibited abnormal TCF7L2 expression at P3 and P5 in the midbrain compared to control mice.
 Quantitative PCR (qRT-PCR)
 P3, P5
Targeted expression1
Decreased
Description: Mutant mice exhibited reduced expression of both long and short TCF7L2 in the midbrain compared to control mice.
 Western blot
 P7
 Not Reported:

M_TCF7L2_8_CKO_HM

Category
Entity
Quantity
Experimental Paradigm
Age at Testing
Ultrasonic vocalization: isolation induced1
Decreased
Description: Mutant mice displayed significantly fewer syllables and decreased peak syllable amplitude (dB) compared to control mice. No change was present in syllable duration (ms) or mean frequency (kHz).
 Monitoring ultrasonic vocalizations
 P17
 Not Reported:

M_TCF7L2_9_CKI_HM

Category
Entity
Quantity
Experimental Paradigm
Age at Testing
Ultrasonic vocalization: isolation induced1
Decreased
Description: Mutant mice displayed significantly fewer syllables, reduced peak amplitude (dB), and shorter syllable duration (ms) compared to control mice, indicating that flTCF7L2 is required for mouse ultrasonic vocalization production
 Monitoring ultrasonic vocalizations
 P7
Targeted expression1
Abnormal
Description: Mutant mice exhibited reduced expression of the long TCF7L2 form and no change in the short TCF7L2 form in the midbrain as compared to control mice.
 Western blot
 P7
 Not Reported:

M_TCF7L2_14_CKO_HM

Category
Entity
Quantity
Experimental Paradigm
Age at Testing
Astrocyte morphology1
Abnormal
Description: Tcf7l2 mutant mice exhibited impairments in astrocytic gap junction function or assembly compared to controls, as measured by a reduction in the spreading of dye via gap junctions of coupled astrocytes.
Exp Paradigm: Gap junction assay
 Immunohistochemistry
 P30
Synapse density: inhibitory1
Increased
Description: Tcf7l2 mutant mice exhibited a modest increase in inhibitory synapses compared to controls.
 Immunohistochemistry
 P30
Synapse density: excitatory1
Increased
Description: Tcf7l2 mutant mice exhibited a significant increase in excitatory synaptic puncta compared to controls.
 Immunohistochemistry
 P30
Astrocyte morphology1
Abnormal
Description: Tcf7l2 mutant mice exhibited significantly larger neighboring astrocyte overlap compared with control cells, consistent with a defect in tiling.
 Immunohistochemistry
 P30
Miniature post synaptic current amplitude: excitatory1
Increased
Description: Tcf7l2 mutant mice exhibited an increase in mEPSC amplitude compared to controls.
 Whole-cell patch clamp
 P30
Miniature post synaptic current frequency: excitatory1
Increased
Description: Tcf7l2 mutant mice exhibited an increase in the frequency of mEPSCs in neurons compared with controls.
 Whole-cell patch clamp
 P30
Sensory-evoked response: excitation: olfactory stimulus1
Increased
Description: Tcf7l2 exhibited a significant increase in the density of c-FOS positive neurons in response to a social scent compared to a neutral scent. While control animals exhibited a slight increase in response to a social scent compared to a neutral scent, this trend did not reach statistical significance.
Exp Paradigm: neutral scent (fresh bedding) vs. social scent (used bedding); c-FOS
 Immunohistochemistry
 not specified
Social interaction1
Decreased
Description: Tcf7l2 mutant female mice exhibited a two-fold increase in sociability compared with control mice, as measured by the amount of time each pair of mice spent together throughout day 3 of the testing period.
Exp Paradigm: 4-box naturalistic setting; housing compartments with access to food and water in box 2 and 4, empty box 1 and 3; same strain, sex, and age mouse pairs
 Reciprocal social interaction test
 not specified
Social scent marking or recognition1
Increased
Description: Tcf7l2 mutant male and female mice spent more time around the social scent and visited the social scent area significantly more times compared to controls. No differences between mutant males and females in approach to the social scent were observed.
Exp Paradigm: social scent in chamber 1, neutral scent in chamber 2
 Three-chamber social approach test
 not specified
Social scent marking or recognition1
Increased
Description: Tcf7l2 mutant female mice exhibited a significantly stronger preference for the social scent, spending more time in the box with the social scent compared with control mice.
Exp Paradigm: 4-box naturalistic setting; housing compartments with access to food and water in box 2 and 4, social scent in box 3 and a neutral scent in box 1
 Olfactory discrimination test
 not specified
Social memory1
Increased
Description: Tcf7l2 mutant mice exhibited a significant increase in the ratio of the amount of time spent with the novel mouse compared to the familiar mouse in comparison to controls, suggesting an increased preference for the socially novel mouse.
Exp Paradigm: familiar mouse in chamber 1 (s1), stranger mouse (s2) in chamber 2
 Three-chamber social approach test
 not specified
Exploratory activity: habituation1
Decreased
Description: Tcf7l2 mutant mice exhibited a steady preference for the social stimulus during the entire testing period, while the preference of the control mice for the social scent decreased over the 10-min testing window.
Exp Paradigm: social scent in chamber 1, neutral scent in chamber 2
 Three-chamber social approach test
 not specified
Differential gene expression1
Abnormal
Description: Tcf7l2 mutant mice exhibited changes in the expression of 275 genes, among which 157 were downregulated and 118 were upregulated. Gene ontology analysis showed the significant enrichment of networks that are involved in glial cell differentiation, the development of gap junction, assembly, and the regulation of neuronal function.
 RNA sequencing
 P35
Targeted expression1
Decreased
Description: Tcf7l2 mutant mice exhibited an 85% reduction of cortical astrocytes that expressed TCF7L2 isoforms compared to controls. Tcf7l2 mutant mice exhibited similar levels to controls of TCF7L2 in other cell types in the brain, such as thalamic neurons and oligodendrocyte lineage cells.
 Fluorescence microscopy
 P15
Protein expression: in situ protein expression1
Abnormal
Description: RNA-seq data was validated at the protein level by quantifying the expression of astrocytic Connexins (Connexin-30 and Connexin-43), and it was observed that mutant mice exhibited a significant decrease in the number of Connexin-30 and Connexin-43 puncta compared to controls.
Exp Paradigm: Cx30
 Immunohistochemistry
 P35
Reward reinforced choice behavior1
 No change
 Sucrose preference test
 not specified
General locomotor activity1
 No change
 Reciprocal social interaction test
 not specified
Astrocyte number1
 No change
 Fluorescence microscopy
 P15
Astrocyte number1
 No change
 Immunohistochemistry
 P15
Miniature post synaptic current amplitude: inhibitory1
 No change
 Whole-cell patch clamp
 P30
Miniature post synaptic current frequency: inhibitory1
 No change
 Whole-cell patch clamp
 P30
Social approach1
 No change
 Three-chamber social approach test
 not specified
 Not Reported:

M_TCF7L2_15_CKO_HM

Category
Entity
Quantity
Experimental Paradigm
Age at Testing
Astrocyte morphology1
Abnormal
Description: Tcf7l2 mutant mice exhibited a significantly larger astrocyte volume relative to controls.
 Immunohistochemistry
 P15
 Not Reported:


Interactor Symbol Interactor Name Interactor Organism Entrez ID Uniprot ID Interaction Type Evidence Reference
CHD8 chromodomain helicase DNA binding protein 8 57680 Q9HCK8 ChIP-chip
Subtil-Rodrguez A , et al. 2013
CTBP1 C-terminal binding protein 1 13016 O88712 GST; Y2H; Luciferase reporter assay
Valenta T , et al. 2003
CTNNB1 catenin (cadherin-associated protein), beta 1, 88kDa 1499 Q9WU82 IP/WB; in vitro binding assay
Tzeng SL , et al. 2006
CTNNB1 catenin (cadherin-associated protein), beta 1, 88kDa 1499 Q9WU82 GST; IP/WB; ChIP; High-throughput fluorescence polarization interaction analysis assay
Del Valle-Prez B , et al. 2011
DAXX death-domain associated protein 1616 Q53F85 Y2H; IP/WB; Co-localization; Luciferase reporter assay; EMSA
Tzeng SL , et al. 2006
DAZAP2 DAZ-associated protein 2 9802 Q15038 Y2H; in vitro binding assay; IP/WB; Luciferase reporter assay; ChIP
Lukas J , et al. 2009
ELAVL1 ELAV (embryonic lethal, abnormal vision, Drosophila)-like 1 (Hu antigen R) 1994 Q15717 IP; RNP IP; IP/WB
Abdelmohsen K , et al. 2009
FBLN1 fibulin 1 2192 P23142 Y2H
Wang J , et al. 2011
FMR1 fragile X mental retardation 1 2332 G8JLE9 PAR-CLIP
Ascano M Jr , et al. 2012
HDAC4 histone deacetylase 4 9759 P56524 GST; In vivo acetylation assay; in vitro kinase assay; AMPK kinase assay; IP/WB
Lin YY , et al. 2012
IKZF5 IKAROS family zinc finger 5 (Pegasus) 64376 Q9H5V7 IP; LC-MS/MS
Huttlin EL , et al. 2015
JUP junction plakoglobin 3728 P14923 in vitro binding assay
Miravet S , et al. 2001
MAD2L2 MAD2 mitotic arrest deficient-like 2 (yeast) 10459 Q9UI95 Y2H; IP/WB; Co-localization; Luciferase reporter assay
Hong CF , et al. 2009
NLK nemo-like kinase 51701 Q9UBE8 IP/WB; EMSA
Ishitani T , et al. 2003
PARP1 poly (ADP-ribose) polymerase 1 142 P09874 IP; LC-MS/MS; IP/WB
Idogawa M , et al. 2007
PRMT6 protein arginine methyltransferase 6 55170 Q96LA8 IP; LC-MS/MS
Huttlin EL , et al. 2015
PSEN1 presenilin 1 5663 P49768 IP/WB
Palacino JJ , et al. 2001
PSMA3 proteasome (prosome, macropain) subunit, alpha type, 3 5684 P25788 Y2H
Wang J , et al. 2011
SUMO2 SMT3 suppressor of mif two 3 homolog 2 (S. cerevisiae) 6613 P61956 SILAC; TAP; LC-MS/MS
Tatham MH , et al. 2011
UBC ubiquitin C 7316 P63279 diGly proteomics
Kim W , et al. 2011
UBE2I ubiquitin-conjugating enzyme E2I (UBC9 homolog, yeast) 7329 P63279 Y2H
Wang J , et al. 2011
XRCC5 X-ray repair complementing defective repair in Chinese hamster cells 5 (double-strand-break rejoining) 7520 P13010 IP; LC-MS/MS; IP/WB
Idogawa M , et al. 2007
XRCC6 X-ray repair complementing defective repair in Chinese hamster cells 6 2547 P12956 IP; LC-MS/MS; IP/WB; Luciferase reporter assay
Idogawa M , et al. 2007
ZBTB33 Transcriptional regulator Kaiso 56805 Q8BN78 IP/WB
Del Valle-Prez B , et al. 2011
CEBPA CCAAT/enhancer-binding protein zeta 12606 P53569 Luciferase reporter assay; IP
Kennell JA , et al. 2003
Ctnnb1 catenin (cadherin associated protein), beta 1 12387 Q02248 IP/WB
ChIP-Seq; ChIP-qPCR
Zhao C , et al. 2016
EP300 E1A binding protein p300 2033 Q9Y6B2 Luciferase reporter assay
Kennell JA , et al. 2003
NR5A1 nuclear receptor subfamily 5, group A, member 1 26423 P33242 Luciferase reporter assay
Kennell JA , et al. 2003
Sox10 SRY-box containing gene 10 20665 Q04888 IP/WB; Co-localization
Zhao C , et al. 2016
Zbtb33 Transcriptional regulator Kaiso 56805 Q8BN78 IP/WB; Co-localization
Zhao C , et al. 2016
Axin2 axin 2 29134 O70240 ChIP-Seq; ChIP-qPCR
Zhao C , et al. 2016
Ccnd1 cyclin D1 58919 P39948 ChIP-Seq; ChIP-qPCR
Zhao C , et al. 2016
Ctnnb1 catenin (cadherin-associated protein), beta 1, 88kDa 1499 Q9WU82 IP/WB
ChIP-Seq; ChIP-qPCR
Zhao C , et al. 2016
Lef1 lymphoid enhancer binding factor 1 161452 Q9QXN1 ChIP-Seq; ChIP-qPCR
Zhao C , et al. 2016
Mbp myelin basic protein 24547 P02688 ChIP-Seq
Zhao C , et al. 2016
Myrf myelin regulatory factor 293736 D4A352 ChIP-Seq; ChIP-qPCR
Zhao C , et al. 2016
Olig1 oligodendrocyte transcription factor 1 60394 Q9WUQ3 ChIP-Seq
Zhao C , et al. 2016
Sp5 Sp5 transcription factor 296510 A0A0G2JUC1 ChIP-Seq; ChIP-qPCR
Zhao C , et al. 2016
Ugt8 UDP glycosyltransferase 8 50555 Q09426 ChIP-Seq
Zhao C , et al. 2016
Wnt10a wingless-type MMTV integration site family, member 10A 316527 D3ZRW5 ChIP-Seq; ChIP-qPCR
Zhao C , et al. 2016
Wnt11 wingless-type MMTV integration site family, member 11 140584 G3V819 ChIP-Seq; ChIP-qPCR
Zhao C , et al. 2016
Zfp24 zinc finger protein 24 360204 Q7TNK3 ChIP-Seq
Zhao C , et al. 2016

HELP
Copyright © 2017 MindSpec, Inc.