HELP     Sign In
Search

Relevance to Autism

A de novo missense variant in the ASPM gene was identified in an ASD proband from the Simons Simplex Collection in Iossifov et al., 2014. Rare inherited loss-of-function and damaging missense variants in the ASPM gene were identified in ASD probands from the Simons Simplex Collection in Krumm et al., 2015 and in a cohort of Chinese ASD probands in Guo et al., 2017. Subsequent Transmission and De Novo Association (TADA) analysis in Guo et al., 2017 identified ASPM as an ASD candidate gene, with a PTADA of 0.001826 in the Chinese ASD case-control cohort and a PTADA of 0.001356 in a combined cohort of Chinese ASD probands and controls, as well as ASD probands and controls from the Simons Simplex Collection and the Autism Sequencing Consortium.

Molecular Function

This gene is the human ortholog of the Drosophila melanogaster 'abnormal spindle' gene (asp), which is essential for normal mitotic spindle function in embryonic neuroblasts. Studies in mouse also suggest a role of this gene in mitotic spindle regulation, with a preferential role in regulating neurogenesis. Biallelic mutations in the ASPM gene are responsible for a form of autosomal recessive primary microcephaly (MCPH5; OMIM 608716), a disorder associated with intellectual disability and speech delay (Bond et al., 2002; Bond et al., 2003).

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
Elucidation of the phenotypic spectrum and genetic landscape in primary and secondary microcephaly.
Microcephaly
DD, epilepsy/seizures
Support
Excess of rare, inherited truncating mutations in autism.
ASD
Support
Protein-truncating mutations in ASPM cause variable reduction in brain size.
Autosomal recessive primary microcephaly-5 (MCPH5)
Support
Integrating de novo and inherited variants in 42
ASD
Support
Mutational Landscape of Autism Spectrum Disorder Brain Tissue
ASD
Support
A truncating Aspm allele leads to a complex cognitive phenotype and region-specific reductions in parvalbuminergic neurons.
Support
Inherited and De Novo Genetic Risk for Autism Impacts Shared Networks.
ASD
Support
Autosomal recessive primary microcephaly 5
Highly Cited
ASPM is a major determinant of cerebral cortical size.
Autosomal recessive primary microcephaly-5 (MCPH5)
Recent Recommendation
Targeted sequencing and functional analysis reveal brain-size-related genes and their networks in autism spectrum disorders.
ASD

Rare

Variant ID
Variant Type
Allele Change
Residue Change
Inheritance Pattern
Inheritance Association
Family Type
Author, Year
 GEN939R001 
 missense_variant 
 c.4669T>C 
 p.Cys1557Arg 
 De novo 
  
 Simplex 
 GEN939R002 
 missense_variant 
 c.4892G>A 
 p.Arg1631Lys 
 Familial 
 Maternal 
 Simplex 
 GEN939R003 
 frameshift_variant 
 c.4066-5164del 
  
 Familial 
 Paternal 
 Simplex 
 GEN939R004 
 frameshift_variant 
 c.4066-5302del 
  
 Familial 
 Maternal 
 Simplex 
 GEN939R005 
 stop_gained 
 c.4066-5522T>G 
  
 Familial 
 Maternal 
 Simplex 
 GEN939R006 
 stop_gained 
 c.4066-5522T>G 
  
 Familial 
 Maternal 
 Simplex 
 GEN939R007 
 missense_variant 
 C>T 
 p.Ala1877Thr 
 Familial 
 Paternal 
 Simplex 
 GEN939R008 
 missense_variant 
 C>T 
 p.Ala1877Thr 
 Familial 
 Maternal 
 Simplex 
 GEN939R009 
 missense_variant 
 c.4066-7458G>A 
  
 Familial 
 Paternal 
 Simplex 
 GEN939R010 
 missense_variant 
 c.4066-8871G>A 
  
 Familial 
 Maternal 
 Simplex 
 GEN939R011 
 missense_variant 
 C>T 
 p.Arg1264His 
 Familial 
 Paternal 
 Simplex 
 GEN939R012 
 missense_variant 
 C>T 
 p.Arg1264His 
 Familial 
 Paternal 
 Simplex 
 GEN939R013 
 missense_variant 
 c.3791G>A 
 p.Arg1264His 
 Familial 
 Paternal 
 Simplex 
 GEN939R014 
 missense_variant 
 c.3624G>T 
 p.Glu1208Asp 
 Familial 
 Maternal 
 Simplex 
 GEN939R015 
 splice_site_variant 
 c.3599-2A>G 
  
 Familial 
 Paternal 
 Simplex 
 GEN939R016 
 missense_variant 
 c.3497C>T 
 p.Thr1166Ile 
 Familial 
 Maternal 
 Simplex 
 GEN939R017 
 frameshift_variant 
 c.3485_3486del 
 p.Cys1162SerfsTer22 
 Familial 
 Maternal 
 Simplex 
 GEN939R018 
 missense_variant 
 c.3395A>T 
 p.Glu1132Val 
 Familial 
 Maternal 
 Simplex 
 GEN939R019 
 missense_variant 
 c.2824C>T 
 p.Arg942Cys 
 Familial 
 Paternal 
 Simplex 
 GEN939R020 
 missense_variant 
 c.2746G>A 
 p.Asp916Asn 
 Familial 
 Maternal 
 Simplex 
 GEN939R021 
 missense_variant 
 c.2621A>G 
 p.Tyr874Cys 
 Familial 
 Maternal 
 Simplex 
 GEN939R022 
 missense_variant 
 c.2612C>T 
 p.Pro871Leu 
 Familial 
 Maternal 
 Simplex 
 GEN939R023 
 missense_variant 
 c.2339A>C 
 p.Lys780Thr 
 Familial 
 Paternal 
 Simplex 
 GEN939R024 
 missense_variant 
 c.2300G>A 
 p.Arg767His 
 Familial 
 Paternal 
 Simplex 
 GEN939R025 
 missense_variant 
 c.2267A>G 
 p.Tyr756Cys 
 Familial 
 Paternal 
 Simplex 
 GEN939R026 
 missense_variant 
 c.2047C>T 
 p.Pro683Ser 
 Familial 
 Paternal 
 Simplex 
 GEN939R027 
 missense_variant 
 C>A 
 p.Ala663Ser 
 Familial 
 Paternal 
 Simplex 
 GEN939R028 
 missense_variant 
 C>A 
 p.Ala663Ser 
 Familial 
 Maternal 
 Simplex 
 GEN939R029 
 missense_variant 
 c.1987G>T 
 p.Ala663Ser 
 Familial 
 Paternal 
 Simplex 
 GEN939R030 
 frameshift_variant 
 c.1632_1636del 
 p.Leu545AsnfsTer8 
 Familial 
 Maternal 
 Simplex 
 GEN939R031 
 stop_gained 
 c.4786C>T 
 p.Arg1596Ter 
 Familial 
 Paternal 
 Simplex 
 GEN939R032 
 frameshift_variant 
 c.4066-5025dup 
  
 Familial 
 Maternal 
 Simplex 
 GEN939R033 
 frameshift_variant 
 c.4066-5164del 
  
 Familial 
 Paternal 
 Simplex 
 GEN939R034 
 frameshift_variant 
 c.4066-6166_4066-6165del 
  
 Familial 
 Maternal 
 Simplex 
 GEN939R035 
 missense_variant 
 c.4066-7430G>T 
  
 Familial 
 Maternal 
 Simplex 
 GEN939R036 
 missense_variant 
 c.4066-7458G>A 
  
 Familial 
 Paternal 
 Simplex 
 GEN939R037 
 missense_variant 
 c.4066-7458G>A 
  
 Familial 
 Paternal 
 Simplex 
 GEN939R038 
 missense_variant 
 c.3829T>C 
 p.Trp1277Arg 
 Familial 
 Maternal 
 Simplex 
 GEN939R039 
 missense_variant 
 c.3791G>A 
 p.Arg1264His 
 Familial 
 Maternal 
 Simplex 
 GEN939R040 
 missense_variant 
 c.3791G>A 
 p.Arg1264His 
 Familial 
 Maternal 
 Simplex 
 GEN939R041 
 missense_variant 
 c.3406G>A 
 p.Val1136Met 
 Familial 
 Maternal 
 Simplex 
 GEN939R042 
 missense_variant 
 c.3091A>G 
 p.Ile1031Val 
 Familial 
 Maternal 
 Simplex 
 GEN939R043 
 missense_variant 
 c.2275C>T 
 p.Arg759Trp 
 Familial 
 Maternal 
 Simplex 
 GEN939R044 
 frameshift_variant 
 c.2094del 
 p.Lys698AsnfsTer10 
 Familial 
 Maternal 
 Simplex 
 GEN939R045 
 missense_variant 
 c.1987G>T 
 p.Ala663Ser 
 Familial 
 Paternal 
 Simplex 
 GEN939R046 
 missense_variant 
 c.1987G>T 
 p.Ala663Ser 
 Familial 
 Paternal 
 Simplex 
 GEN939R047 
 frameshift_variant 
 c.4066-5305_4066-5304del 
  
 Familial 
  
  
 GEN939R048 
 frameshift_variant 
 c.7782_7783del 
 p.Lys2595SerfsTer6 
 Familial 
  
  
 GEN939R049 
 frameshift_variant 
 c.5810_5811insC 
 p.Arg1938LysfsTer9 
 Familial 
  
  
 GEN939R050 
 frameshift_variant 
 c.2672del 
 p.Leu891CysfsTer44 
 Familial 
  
  
 GEN939R051 
 frameshift_variant 
 c.1592_1595del 
 p.Val531GlufsTer17 
 Familial 
  
  
 GEN939R052 
 missense_variant 
 c.5000G>A 
 p.Arg1667His 
 Familial 
  
  
 GEN939R053 
 missense_variant 
 c.5000G>A 
 p.Arg1667His 
 Familial 
  
  
 GEN939R054 
 missense_variant 
 c.3395A>G 
 p.Glu1132Gly 
 Familial 
  
  
 GEN939R055 
 missense_variant 
 c.1468C>T 
 p.Arg490Cys 
 Familial 
  
  
 GEN939R056a 
 stop_gained 
 c.3796G>T 
 p.Glu1266Ter 
 Familial 
 Both parents 
  
 GEN939R057 
 frameshift_variant 
 c.4066-4953_4066-4950del 
  
 Familial 
 Paternal 
 Multiplex (monozygotic twins) 
 GEN939R058 
 frameshift_variant 
 c.4066-5303_4066-5302del 
  
 Familial 
 Paternal 
 Multiplex 
 GEN939R059 
 missense_variant 
 c.4296A>C 
 p.Arg1432Ser 
 Unknown 
  
  
 GEN939R060 
 missense_variant 
 c.5849C>T 
 p.Ala1950Val 
 De novo 
  
  
 GEN939R061 
 missense_variant 
 c.1880G>A 
 p.Arg627His 
 De novo 
  
  
 GEN939R062 
 missense_variant 
 c.1567A>C 
 p.Ser523Arg 
 De novo 
  
 Simplex 
 GEN939R063 
 missense_variant 
 c.2353C>T 
 p.Leu785Phe 
 De novo 
  
 Simplex 
 GEN939R064 
 stop_gained 
 c.4423C>T 
 p.Gln1475Ter 
 Familial 
 Maternal 
 Multiplex 
 GEN939R065 
 stop_gained 
 c.4066-6024C>T 
  
 Familial 
 Paternal 
 Multiplex 
 GEN939R066 
 frameshift_variant 
 c.6686_6689del 
 p.Arg2229ThrfsTer10 
 Familial 
 Maternal 
 Multiplex 
 GEN939R067 
 frameshift_variant 
 c.6591_6594del 
 p.Ser2198ThrfsTer14 
 Familial 
 Paternal 
 Multiplex 
 GEN939R068 
 frameshift_variant 
 c.6562_6565del 
 p.Lys2188CysfsTer24 
 De novo 
  
 Multiplex 
 GEN939R069 
 frameshift_variant 
 c.4528del 
 p.Arg1510GlufsTer6 
 Familial 
 Paternal 
 Multiplex (monozygotic twins) 
 GEN939R070 
 stop_gained 
 c.710T>A 
 p.Leu237Ter 
 Familial 
 Paternal 
 Multiplex 

Common

No Common Variants Available
Chromosome
CNV Locus
CNV Type
# of studies
Animal Model
1
Duplication
 44
 
1
Deletion
 1
 
1
Duplication
 1
 
1
Deletion-Duplication
 17
 

Model Summary

Mice with a truncating Aspm1â??7 truncation mutation show impaired short- and long-term object recognition memory, increased place learning, ventriculomegaly, corpus callosum dysgenesis and decreased parvalbumin (PV)+ interneuron numbers in the hippocampal CA regions and thalamic reticular nucleus.

References

Type
Title
Author, Year
Additional
A truncating Aspm allele leads to a complex cognitive phenotype and region-specific reductions in parvalbuminergic neurons.

M_ASPM_1_KO_HM

Model Type: Genetic LOF
Model Genotype: Homozygous
Mutation: Mice with an aspm truncation mutation where a gene trap vector was integrated between exons 7 and 8 such that the protein product contains only the microtubule binding domain.
Allele Type: Genetic lof
Strain of Origin: 129P2/OlaHsd
Genetic Background: C57BL/6JOlaHsd
ES Cell Line: E14TG2a.4 (ES Cell)
Mutant ES Cell Line:
Model Source: 20823249

M_ASPM_1_KO_HM

Category
Entity
Quantity
Experimental Paradigm
Age at Testing
General locomotor activity1
Decreased
Description: Decreased arm entries
 Y-maze test
 28 weeks
General locomotor activity1
Decreased
Description: Decrease in locomotor activity and speed in the mutant mice
 Open field test
 27 weeks
Hypoactivity1
Increased
Description: Overall sluggish behavior
 Novel cage test
 57 weeks
Morphology and size of the corpus callosum1
Decreased
Description: Corpus callosum dysgenesis, volume of the corpus callosum was clearly smaller
 Histology
 60 weeks
Cell proliferation: neural precursors1
Decreased
Description: Decreased number of dcx+ cells in the dorsal dentate gyrus as a marker of adult neurogenesis
 Immunohistochemistry
 60 weeks
Size of cerebral ventricles: Lateral ventricle1
Increased
Description: Ventriculomegaly, lateral ventricles were significantly larger
 Histology
 60 weeks
Brain size1
Decreased
Description: Mild microcephaly
 Histology
 60 weeks
Neuronal number: Inhibitory neurons1
Decreased
Description: Decreased parvalbumin (pv)+ interneuron numbers in the hippocampal cornu ammonis (ca1,2,3) region and thalamic reticular nucleus (trn) but not in the acc
 Histology
 60 weeks
Exploratory activity1
Decreased
Description: Decreased corner visits, nosepokes and percent corner visits with nosepokes during the first hour on introduction into the intellicage; reduced nosepoke number when lights were switched off at the beginning of the active phase of the cycle
 Novel cage test
 57 weeks
Anxiety1
Increased
Description: Increased corner visits
 Object-place recognition test
 37-39 weeks
Spatial working memory1
Increased
Description: Decreased percent error rate; enhanced place learning in the intellicage
 Object-place recognition test
 57-59 weeks
Object recognition memory: long-term recall1
Decreased
Description: Impaired short- and long-term object recognition memory
 Novel object recognition test
 37-39 weeks
Object recognition memory1
Decreased
Description: Impaired short- and long-term object recognition memory
 Novel object recognition test
 37-39 weeks
Targeted expression1
Decreased
Description: Endogenous aspm mrna was lost in the mutant mice and only genetrapped transcript expressed
 Semi-quantitative PCR (qRT-PCR)
 23 weeks
Targeted expression1
Abnormal
Description: Expression of the truncated aspm protein tagged with lacz
 Immunohistochemistry
 23 weeks
Circadian rhythms: timing/phases of locomotor activity1
 No change
 Running wheel test
 23-27 weeks
Cognitive flexibility1
 No change
 Y-maze test
 28 weeks
Cognitive flexibility1
 No change
 Object-place recognition test
 37-39 weeks
Hippocampal morphology1
 No change
 Immunohistochemistry
 60 weeks
Social memory1
 No change
 Social recognition test
 30-31 weeks
 Not Reported:

 

No Interactions Available
HELP
Copyright © 2017 MindSpec, Inc.