HELP     Sign In
Search

Relevance to Autism

De novo missense variants in the NEUROD2 gene that were experimentally shown to disrupt protein function in Xenopus were identified in three individuals presenting with developmental delay with or without seizures (Sega et al., 2019; Mis et al., 2021); follow-up of these three individuals in Runge et al., 2021 revealed that two of these patients also presented with autism spectrum disorder. In addition to follow-up of these three previously published patients, Runge et al., 2021 characterized three novel patients with experimentally-confirmed loss-of-function missense variants in NEUROD2 and two patients with large de novo deletions encompassing NEUROD2; all five of these novel patients presented with autism spectrum disorder, intellectual disability, and speech disturbance. Runge et al., 2021 also demonstrated that Neurod2 knockout mice displayed altered laminar organization and cortical gene expression, radial over-migration of cortical projection neurons, altered excitatory synapse density and turnover, social interaction deficits, stereotypies, hyperactivity, and occasionally spontaneous seizures; mice that were heterozygous for Neurod2 had similar deficits, and specific deletion of Neurod2 in forebrain excitatory neurons recapitulated cellular and behavioral phenotypies observed in constitutive knockout mice.

Molecular Function

This gene encodes a member of the neuroD family of neurogenic basic helix-loop-helix (bHLH) proteins. Expression of this gene can induce transcription from neuron-specific promoters, such as the GAP-43 promoter, which contain a specific DNA sequence known as an E-box. The product of the human gene can induce neurogenic differentiation in non-neuronal cells in Xenopus embryos, and is thought to play a role in the determination and maintenance of neuronal cell fates.

External Links

        

References

Type
Title
Type of Disorder
Associated Disorders
Author, Year
Primary
De novo pathogenic variants in neuronal differentiation factor 2 (NEUROD2) cause a form of early infantile epileptic encephalopathy
DD, epilepsy/seizures
ASD
Support
Integrating de novo and inherited variants in 42
ASD
Support
Expansion of NEUROD2 phenotypes to include developmental delay without seizures
ASD, DD
ADHD
Recent Recommendation
Disruption of NEUROD2 causes a neurodevelopmental syndrome with autistic features via cell-autonomous defects in forebrain glutamatergic neurons
ASD, DD, ID
Epilepsy/seizures

Rare

Variant ID
Variant Type
Allele Change
Residue Change
Inheritance Pattern
Inheritance Association
Family Type
Author, Year
 GEN1256R001 
 missense_variant 
 c.388G>C 
 p.Glu130Gln 
 De novo 
  
 Simplex 
 GEN1256R002 
 missense_variant 
 c.401T>C 
 p.Met134Thr 
 De novo 
  
 Simplex 
 GEN1256R003 
 missense_variant 
 c.488T>C 
 p.Leu163Pro 
 De novo 
  
 Simplex 
 GEN1256R004 
 missense_variant 
 c.703G>A 
 p.Ala235Thr 
 Unknown 
  
 Unknown 
 GEN1256R005 
 missense_variant 
 c.385C>T 
 p.Arg129Trp 
 De novo 
  
 Simplex 
 GEN1256R006 
 missense_variant 
 c.388G>C 
 p.Glu130Gln 
 De novo 
  
 Simplex 
 GEN1256R007 
 missense_variant 
 c.804C>A 
 p.His268Gln 
 Familial 
 Paternal 
 Multiplex 
 GEN1256R008 
 copy_number_loss 
  
  
 De novo 
  
 Simplex 
 GEN1256R009 
 copy_number_loss 
  
  
 De novo 
  
 Simplex 
 GEN1256R010 
 stop_lost 
 c.1148G>C 
 p.Ter383SerextTer106 
 De novo 
  
  

Common

No Common Variants Available
Chromosome
CNV Locus
CNV Type
# of studies
Animal Model
17
Deletion-Duplication
 74
 
17
Duplication
 1
 
17
Duplication
 1
 

Model Summary

Neurod2 KO embryos show increased migration of cortical projection neurons and abnormal cortical thickness and lamination. Neurod2 KO juvenile and adult mice show misregulated spine density and turnover in apical but not basal

References

Type
Title
Author, Year
Primary
Disruption of NEUROD2 causes a neurodevelopmental syndrome with autistic features via cell-autonomous defects in forebrain glutamatergic neurons

M_NEUROD2_1_KO_HM

Model Type: Genetic LOF
Model Genotype: Homozygous
Mutation: The entire coding region of the gene was replaced with an in-frame lacZ gene followed by a loxP flanked neomycin selection cassette. For layer cortical morpho-functional analyses,Neurod2 KO mice were bred with mice expressing eGFP under the Thy-1 promoter. AAV-TurboRFP was injected to assess neuronal expression in L2/3 of M1 in Neurod2 WT and KO mice, and measure spine density.
Allele Type: knockout
Strain of Origin: C57BL/6J
Genetic Background: C57BL/6N*129Sv
ES Cell Line: Not specified
Mutant ES Cell Line: Not specified
Model Source: Bormuth I, et al, J Neurosci. 2013;33:641â??51 (23303943); Pieper A, Sci Rep 2019 Feb 5;9(1):1448 (30723302)

M_NEUROD2_2_KO_HT

Model Type: Genetic LOF
Model Genotype: Heterozygous
Mutation: The entire coding region of the gene was replaced with an in-frame lacZ gene followed by a loxP flanked neomycin selection cassette. For layer cortical morpho-functional analyses,Neurod2 KO mice were bred with mice expressing eGFP under the Thy-1 promoter.
Allele Type: knockout
Strain of Origin: C57BL/6J
Genetic Background: C57BL/6N*129Sv
ES Cell Line: Not specified
Mutant ES Cell Line: Not specified
Model Source: Bormuth I, et al, J Neurosci. 2013;33:641â??51 (23303943); Pieper A, Sci Rep 2019 Feb 5;9(1):1448 (30723302)

M_NEUROD2_3_CKO_HM

Model Type: Genetic LOF
Model Genotype: Homozygous
Mutation: Neurod2 Tm1a mice, with a conditional allele based on theâ??knockout-firstâ?? design were generated on a C57BL/6N background using cell clone EPD0422_5_B06 from the Knock Out Mouse Project at UC Davis (http://www.komp.org), in which the only coding exon (exon 2) was flanked by loxP sites. Neurod2^flox/flox mice, or Tm1c mice for Neurod2, were produced by crossing Tm1a mice with actin-Flippase mice (Jackson Stock N° 003800). For forebrain-deletion experiments, Neurod2flox/flox mice were crossed with Emx1IRESCre mice.
Allele Type: conditional knockout
Strain of Origin: C57BL/6N
Genetic Background: Not reported
ES Cell Line: Not specified
Mutant ES Cell Line: Not specified
Model Source: Gorski et al, J Neurosci, 2002 (12151506)

M_NEUROD2_1_KO_HM

Category
Entity
Quantity
Experimental Paradigm
Age at Testing
Rearing behavior1
Increased
Description: Increased rearing both at and outside the walls of the cage
 Open field test
 8â??14-weeks
General locomotor activity1
Increased
Description: Increased velocity of locomotion, decreased time spent in rest
 Open field test
 8â??14-weeks
Cortical thickness1
Decreased
Description: Decreased cortical areas m1 and s1, normal aspect and thickness of the cortical wall at sensory-motor levels at 1 month; decreased thickness of the cortical wall at sensory-motor levels at p7; thicknesses of mz and subsequent l1 were unaltered at e18, p7, and p30
 Immunohistochemistry
 1 month, P7
Morphology and size of the corpus callosum1
Decreased
Description: Thickness of the corpus callosum was slightly reduced
 Immunohistochemistry
 1 month
Cortical lamination1
Abnormal
Description: Tbr1 and bcl11b-expressing deep layers extended superficially; enlargement of l5; thinning of rorβ and cux1-expressing layers 2â??4, which were shifted superficially; no change in numbers and laminar distribution of cpn subtypes in s1 at 1 month; superficial shift at p7
 Immunohistochemistry
 1 month, P7
Neuronal morphology1
Abnormal
Description: Somatic volumes of l4 and l5 cpns were not significantly altered; more cells had elongated morphologies and increased overall areas and perimeters, typical of the multipolar-to-bipolar transition
 Immunohistochemistry
 1 month
Dendritic architecture: spine turnover1
Increased
Description: Abnormal spine turnover rates in the apical compartment; formed spines outnumbered eliminated spines, resulting in a net spine gain
 Immunohistochemistry
 1 month
Neuronal migration1
Increased
Description: Excessive radial migration at e13.5-18.5; excess migration maintained at p7; pro-migratory effect of neurod2 deletion is generalized to other cpn subtypes; over-migration already occurs at 2 days post-electroporation when cells had not started terminal translocation yet
 Immunofluorescence staining
 E13.5-E18.5, P7
Dendritic architecture: spine density1
Abnormal
Description: No change in basal spine density at p30 and p120; apical spine density in cpns decreased at p30 but increased at p120; early onset; no change in spine density in other cpns
 Immunohistochemistry
 1, 4 months
Neocortex morphology1
Abnormal
Description: Cortical layers were shifted towards the pial surface;
 Immunohistochemistry
 1 month
EPSP-spike relationship1
Increased
Description: L5 cpns fired significantly more action potentials in response to depolarizing current injections
 Whole-cell patch clamp
 1 month
Neuronal ion channel activity1
Abnormal
Description: Abnormal gene expression of 6 voltage-gated ion channel genes
 Quantitative PCR (qRT-PCR)
 1 month
Miniature post synaptic current amplitude: inhibitory1
Increased
Description: Small increase in mipsc amplitude
 Whole-cell patch clamp
 1 month
Hyperpolarization activated cation currents1
Increased
Description: Increased ih hyperpolarization activated cation (hcn) current density
 Whole-cell patch clamp
 1 month
Seizures1
Increased
Description: Increased incidence of seizures in a third of all mutants
 Observation of seizures
 8â??14-weeks
Social memory1
Decreased
Description: Decreased preference for novel conspecific
 Three-chamber social approach test
 8â??14-weeks
Social approach1
Decreased
Description: Decreased preference for conspecific
 Three-chamber social approach test
 8â??14-weeks
Mortality/lethality1
Increased
Description: Seizures were lethal for half of ko mice for which seizures were observed
 Observation of seizures
 8â??14-weeks
Differential gene expression1
Abnormal
Description: Altered gene expression in the neocortex - motor and somatosensory cortex (geo:gse67539)
 RNA sequencing
 1 month
Differential gene expression1
Abnormal
Description: Abnormal gene expression levels (gse110491); upregulation of voltage-gated ion channel, cell projection morphogenesis and chemical synaptic transmission; among dex asd genes, 8 were sfari score 1 and 25 sfari score 2â??3
 RNA sequencing
 P28
Differential gene expression1
Abnormal
Description: 11 dex genes representative of different expression profiles and disease associations (kcnh1 and cdh8 with expected l2/3 enrichment, kcnq5 and htr2a with l5 enrichment, scn4b, kcnk4, scn8a, scn1a, cacna1c, and grin2b with l2-6 enrichment) showed abnormal expression level i
 Quantitative PCR (qRT-PCR)
 1 month
Object recognition memory1
 No change
 Novel object recognition test
 8â??14-weeks
Anatomical projections and connectivity1
 No change
 Retrograde labeling assay
 1 month
Cell proliferation: neural precursors1
 No change
 Immunohistochemistry
 E18, P7, P30
Dendritic architecture: dendritic tree complexity1
 No change
 Immunohistochemistry
 1 month
Synapse density: Inhibitory1
 No change
 Immunohistochemistry
 1 month
Action potential property: after hyperpolarization1
 No change
 Whole-cell patch clamp
 1 month
Action potential property: amplitude1
 No change
 Whole-cell patch clamp
 1 month
Action potential property: threshold1
 No change
 Whole-cell patch clamp
 1 month
Intrinsic membrane properties1
 No change
 Whole-cell patch clamp
 1 month
Membrane potential1
 No change
 Whole-cell patch clamp
 1 month
Miniature post synaptic current amplitude: excitatory1
 No change
 Whole-cell patch clamp
 1 month
Miniature post synaptic current frequency: excitatory1
 No change
 Whole-cell patch clamp
 1 month
Miniature post synaptic current frequency: inhibitory1
 No change
 Whole-cell patch clamp
 1 month
Neuronal apoptosis1
 No change
 Immunohistochemistry
 E18, P7, P30
Synaptic transmission1
 No change
 Whole-cell patch clamp
 1 month
Circling1
 No change
 Open field test
 8â??14-weeks
 Not Reported:

M_NEUROD2_2_KO_HT

Category
Entity
Quantity
Experimental Paradigm
Age at Testing
General locomotor activity1
Increased
Description: No change in velocity of locomotion, decrease in time spent at rest
 Open field test
 8â??14-weeks
Neocortex morphology1
Abnormal
Description: More cells had elongated morphologies and increased overall areas and perimeters, typical of the multipolar-to-bipolar transition
 Immunohistochemistry
 1 month
Cortical thickness1
Decreased
Description: Thicknesses of mz and subsequent l1 were unaltered; iz and lower cp showed the strongest reductions in electroporated cell proportions in e13-e18 electroporated brains
 Immunohistochemistry
 E18, P7, and P30
Neuronal migration1
Increased
Description: Excessive radial migration; no presence of ectopic neurons in mz; over-migration already occurs at 2 days post-electroporation when cells had not started terminal translocation yet
 Immunofluorescence staining
 E13.5-E18.5
Neuronal ion channel activity1
Abnormal
Description: Abnormal gene expression of 6 voltage-gated ion channel genes
 Quantitative PCR (qRT-PCR)
 1 month
Circling1
Increased
Description: Increased circling
 Observation of repetitive behavior
 8â??14-weeks
Seizures1
Increased
Description: Increased incidence of seizures in one mutant
 Observation of seizures
 8â??14-weeks
Social memory1
Decreased
Description: Decreased preference for novel conspecific
 Three-chamber social approach test
 8â??14-weeks
Differential gene expression1
Abnormal
Description: 11 dex genes representative of different expression profiles and disease associations (kcnh1 and cdh8 with expected l2/3 enrichment, kcnq5 and htr2a with l5 enrichment, scn4b, kcnk4, scn8a, scn1a, cacna1c, and grin2b with l2-6 enrichment) showed an expression level in het mice that was either intermediate between wt and ko or undistinguishable to the ko level
 Quantitative PCR (qRT-PCR)
 1 month
Mortality/lethality1
 No change
 Observation of seizures
 8â??14-weeks
Object recognition memory1
 No change
 Novel object recognition test
 8â??14-weeks
Rearing behavior1
 No change
 Observation of repetitive behavior
 8â??14-weeks
Social approach1
 No change
 Three-chamber social approach test
 8â??14-weeks
 Not Reported:

M_NEUROD2_3_CKO_HM

Category
Entity
Quantity
Experimental Paradigm
Age at Testing
General locomotor activity1
Increased
Description: Increased velocity of locomotion, decreased time spent in rest
 Open field test
 8â??14-weeks
Seizures1
Increased
Description: Occasional spontaneous seizures
 Observation of seizures
 8â??14-weeks
Social memory1
Decreased
Description: Decreased preference for novel conspecific
 Three-chamber social approach test
 8â??14-weeks
Social approach1
Decreased
Description: Decreased preference for conspecific
 Three-chamber social approach test
 8â??14-weeks
Object recognition memory1
 No change
 Novel object recognition test
 8â??14-weeks
 Not Reported:

No PIN Data Available
HELP
Copyright © 2017 MindSpec, Inc.