HELP     Sign In
Search

Relevance to Autism

Analysis of 135 ASD probands from consanguineous marriages recruited for the Simons Recessive Autism Cohort (SRAC) in Wenderski et al., 2000 identified six families with segregating loss-of-function variants in ACTL6B; when considering all coding genes, ACTL6B showed genome-wide significance for variants in the SRAC. In the same report, Actl6b knockout mice on two genetic backgrounds displayed ASD-associated behaviors, including social and memory impairments, repetitive behaviors, hyperactivity, and hypoplasia of the corpus callosum. Bell et al., 2019 had previously demonstrated that individuals harboring biallelic mutations in the ACTL6B gene presented with a neurodevelopmental disorder characterized by global developmental delay, epileptic encephalopathy, axial hypotonia, and spasticity, whereas individuals with de novo heterozygous missense variants in the same gene presented with intellectual disability, developmental delay, delayed or absent speech, ambulation deficits, hypotonia, autism or autistic features, Rett-like stereotypies such as handwringing, and minor facial dysmorphisms (wide mouth, diastema, bulbous nose). Homozygous variants in the ACTL6B gene had previously been identified in three individuals from two families presenting with severe developmental and epileptic encephalopathy in Fichera et al., 2019, as well as in two siblings presenting with intellectual disability, seizures, and autistic behaviors (Karaca et al., 2015) and a female patient diagnosed with atypical Rett syndrome (Sajan et al., 2017). A postzygotic mosaic coding-synonymous variant that was predicted to create a new exonic splicing site in the ACTL6B gene was observed in an ASD proband from the Simons Simplex Collection in Krupp et al., 2017.

Molecular Function

The protein encoded by this gene is a member of a family of actin-related proteins (ARPs) which share significant amino acid sequence identity to conventional actins. Both actins and ARPs have an actin fold, which is an ATP-binding cleft, as a common feature. The ARPs are involved in diverse cellular processes, including vesicular transport, spindle orientation, nuclear migration and chromatin remodeling. This gene encodes a subunit of the BAF (BRG1/brm-associated factor) complex in mammals, which is functionally related to SWI/SNF complex in S. cerevisiae and Drosophila; the latter is thought to facilitate transcriptional activation of specific genes by antagonizing chromatin-mediated transcriptional repression. This subunit may be involved in the regulation of genes by structural modulation of their chromatin, specifically in the brain.

External Links

        

References

Type
Title
Type of Disorder
Associated Disorders
Author, Year
Primary
Mutations in ACTL6B Cause Neurodevelopmental Deficits and Epilepsy and Lead to Loss of Dendrites in Human Neurons.
DD, ID
Epilepsy/seizures, ASD or autistic features
Support
Lessons Learned from Large-Scale, First-Tier Clinical Exome Sequencing in a Highly Consanguineous Population.
Motor delay
Hypertonia, GERD
Support
Variantrecurrence in neurodevelopmental disorders: the use of publicly available genomic data identifies clinically relevant pathogenic missense v...
DD, hypotonia
Support
Integrating de novo and inherited variants in 42
ASD
Support
Mutations in ACTL6B, coding for a subunit of the neuron-specific chromatin remodeling complex nBAF, cause early onset severe developmental and epil...
DD, epilepsy/seizures
Support
Complex Diagnostics of Non-Specific Intellectual Developmental Disorder
DD, ID
Epilepsy/seizures
Support
Exonic Mosaic Mutations Contribute Risk for Autism Spectrum Disorder.
ASD
Support
Impaired Neurodevelopmental Genes in Slovenian Autistic Children Elucidate the Comorbidity of Autism With Other Developmental Disorders
ASD
ID
Support
Enrichment of mutations in chromatin regulators in people with Rett syndrome lacking mutations in MECP2.
Rett syndrome
Support
Rare variants in the outcome of social skills group training for autism
ASD
Support
Genes that Affect Brain Structure and Function Identified by Rare Variant Analyses of Mendelian Neurologic Disease.
ID, epilepsy/seizures
Autistic behavior, microcephaly
Support
Pathogenic homozygous variations in ACTL6B cause DECAM syndrome: Developmental delay, Epileptic encephalopathy, Cerebral Atrophy, and abnormal Myel...
DD, epilepsy/seizures
Recent Recommendation
Recent Recommendation
Loss of the neural-specific BAF subunit ACTL6B relieves repression of early response genes and causes recessive autism
ASD
DD, ID, epilepsy/seizures

Rare

Variant ID
Variant Type
Allele Change
Residue Change
Inheritance Pattern
Inheritance Association
Family Type
Author, Year
 GEN1087R001a 
 inframe_deletion 
 c.441_443del 
 p.Phe147del 
 Familial 
 Both parents 
 Unknown 
 GEN1087R002a 
 frameshift_variant 
 c.695del 
 p.Pro232GlnfsTer24 
 Familial 
  
 Multiplex 
 GEN1087R002b 
 stop_gained 
 c.1275C>A 
 p.Cys425Ter 
 Familial 
  
 Multiplex 
 GEN1087R003a 
 frameshift_variant 
 c.1279del 
 p.Ter427AspfsTer34 
 Familial 
 Both parents 
 Multiplex 
 GEN1087R004a 
 missense_variant 
 c.389G>A 
 p.Arg130Gln 
 Familial 
  
 Simplex 
 GEN1087R004b 
 stop_gained 
 c.556C>T 
 p.Gln186Ter 
 Familial 
  
 Simplex 
 GEN1087R005a 
 stop_gained 
 c.852C>G 
 p.Tyr284Ter 
 Familial 
  
 Simplex 
 GEN1087R005b 
 stop_gained 
 c.740G>A 
 p.Trp247Ter 
 Familial 
  
 Simplex 
 GEN1087R006a 
 stop_gained 
 c.1231C>T 
 p.Gln411Ter 
 Familial 
  
 Simplex 
 GEN1087R006b 
 splice_site_variant 
 c.669+1G>A 
  
 Familial 
  
 Simplex 
 GEN1087R007a 
 stop_gained 
 c.289C>T 
 p.Arg97Ter 
 Familial 
 Both parents 
 Simplex 
 GEN1087R008a 
 missense_variant 
 c.1045G>A 
 p.Gly349Ser 
 Familial 
 Both parents 
 Multiplex 
 GEN1087R009a 
 stop_gained 
 c.724C>T 
 p.Gln242Ter 
 Familial 
  
 Simplex 
 GEN1087R009b 
 missense_variant 
 c.617T>C 
 p.Leu206Pro 
 Familial 
  
 Simplex 
 GEN1087R010a 
 frameshift_variant 
 c.1279del 
 p.Ter427AspfsTer34 
 Familial 
 Both parents 
 Simplex 
 GEN1087R011 
 missense_variant 
 c.1027G>A 
 p.Gly343Arg 
 De novo 
  
  
 GEN1087R012 
 missense_variant 
 c.1027G>A 
 p.Gly343Arg 
 De novo 
  
  
 GEN1087R013 
 missense_variant 
 c.1027G>A 
 p.Gly343Arg 
 De novo 
  
  
 GEN1087R014 
 missense_variant 
 c.1027G>A 
 p.Gly343Arg 
 De novo 
  
  
 GEN1087R015 
 missense_variant 
 c.1027G>A 
 p.Gly343Arg 
 De novo 
  
  
 GEN1087R016 
 missense_variant 
 c.1027G>A 
 p.Gly343Arg 
 De novo 
  
  
 GEN1087R017 
 missense_variant 
 c.1027G>A 
 p.Gly343Arg 
 De novo 
  
  
 GEN1087R018 
 missense_variant 
 c.1027G>A 
 p.Gly343Arg 
 De novo 
  
  
 GEN1087R019 
 missense_variant 
 c.230A>G 
 p.Asp77Gly 
 De novo 
  
  
 GEN1087R020 
 missense_variant 
 c.1027G>A 
 p.Gly343Arg 
 De novo 
  
  
 GEN1087R021a 
 stop_gained 
 c.820C>T 
 p.Gln274Ter 
 Familial 
 Both parents 
 Multiplex 
 GEN1087R022a 
 missense_variant 
 c.1045G>A 
 p.Gly349Ser 
 Familial 
 Both parents 
 Multiplex 
 GEN1087R023 
 synonymous_variant 
 c.360C>T 
 p.Ser120= 
 De novo 
  
 Simplex 
 GEN1087R024a 
 missense_variant 
 c.893G>A 
 p.Arg298Gln 
 Familial 
 Both parents 
 Multiplex 
 GEN1087R025a 
 frameshift_variant 
 c.1279del 
 p.Ter427AspfsTer34 
 Familial 
 Both parents 
 Simplex 
 GEN1087R026 
 missense_variant 
 c.1027G>A 
 p.Gly343Arg 
 Unknown 
 Not maternal 
 Multi-generational 
 GEN1087R027a 
 stop_gained 
 c.999T>A 
 p.Cys333Ter 
 Familial 
 Both parents 
 Not simplex (positive family history) 
 GEN1087R028a 
 inframe_deletion 
 c.1261_1275del 
 p.Val421_Cys425del 
 Familial 
 Both parents 
 Extended multiplex 
 GEN1087R029a 
 missense_variant 
 c.1177G>A 
 p.Gly393Arg 
 Familial 
 Both parents 
 Multiplex 
 GEN1087R030a 
 missense_variant 
 c.460C>T 
 p.Leu154Phe 
 Familial 
 Both parents 
 Multiplex 
 GEN1087R031a 
 stop_gained 
 c.892C>T 
 p.Arg298Ter 
 Familial 
 Both parents 
 Multiplex 
 GEN1087R032a 
 missense_variant 
 c.523A>C 
 p.Thr175Pro 
 Familial 
 Both parents 
 Multiplex 
 GEN1087R033a 
 frameshift_variant 
 c.465del 
 p.Ala156ProfsTer64 
 Familial 
 Both parents 
 Simplex 
 GEN1087R034a 
 missense_variant 
 c.1249G>T 
 p.Gly417Trp 
 Familial 
 Both parents 
 Simplex 
 GEN1087R035 
 missense_variant 
 c.694C>A 
 p.Pro232Thr 
 Unknown 
  
  
 GEN1087R036 
 missense_variant 
 c.694C>A 
 p.Pro232Thr 
 Unknown 
  
  
 GEN1087R037a 
 missense_variant 
 c.554T>C 
 p.Leu185Pro 
 Familial 
 Both parents 
 Multiplex 
 GEN1087R038 
 synonymous_variant 
 c.930C>T 
 p.Asn310%3D 
 De novo 
  
 Simplex 

Common

No Common Variants Available
Chromosome
CNV Locus
CNV Type
# of studies
Animal Model
7
Deletion
 2
 
7
Duplication
 2
 
7
Deletion-Duplication
 28
 

Model Summary

Actl6b KO mice show decreased survival, corpus callosum hypoplasia, thinner myelin sheaths, decreased sociability, decreaed social interaction with a juvenile, decreased reciprocal social interaction, impaired spatial memory, increase in distance travelled, repetitive behaviors, hyperactivity, reduced affinity for neuronal chromatin.

References

Type
Title
Author, Year
Primary
Loss of the neural-specific BAF subunit ACTL6B relieves repression of early response genes and causes recessive autism

M_ACTL6B_1_KO_HM

Model Type: Genetic
Model Genotype: Homozygous
Mutation: Actl6b KO mice where exons 2 and 3 are replaced with a neo cassette. Mice were provided recovery gel after weaning to increase survival.
Allele Type: Knockout
Strain of Origin: 129S6/SvEV*C57BL/6
Genetic Background: C57BL/6
ES Cell Line: NA
Mutant ES Cell Line: NA
Model Source: Stanford University

M_ACTL6B_2_KO_HT

Model Type: Genetic
Model Genotype: Heterozygous
Mutation: Actl6b KO mice where exons 2 and 3 are replaced with a neo cassette
Allele Type: Knockout
Strain of Origin: 129S6/SvEV*C57BL/6
Genetic Background: C57BL/6
ES Cell Line: NA
Mutant ES Cell Line: NA
Model Source: Stanford University

M_ACTL6B_3_KO_HM

Model Type: Genetic
Model Genotype: Homozygous
Mutation: Actl6b KO mice where exons 2 and 3 are replaced with a neo cassette. Male F1 hybrids were used in the experiment.
Allele Type: Knockout
Strain of Origin: 129S6/SvEV*C57BL/6
Genetic Background: 129S6/SvEV*C57BL/6
ES Cell Line: NA
Mutant ES Cell Line: NA
Model Source: Fujita Health University

M_ACTL6B_1_KO_HM

Category
Entity
Quantity
Experimental Paradigm
Age at Testing
General locomotor activity: ambulatory activity1
Increased
Description: Increase in distance travelled
Exp Paradigm: NA
 Open field test
 7-11 weeks
Morphology and size of the corpus callosum1
Decreased
Description: Hypoplasia of the corpus callosum
Exp Paradigm: NA
 Immunohistochemistry
 7-11 weeks
Rearing behavior1
Increased
Description: Increased vertical activity
Exp Paradigm: NA
 Open field test
 7-11 weeks
Social interaction: with juveniles1
Decreased
Description: Decrease in interaction with juvenile
Exp Paradigm: NA
 Reciprocal social interaction test
 7 weeks
Social approach1
Decreased
Description: Decrease in social preference
Exp Paradigm: NA
 Three-chamber social approach test
 7-11 weeks
Anxiety1
Increased
Description: Decrease in time spent in the center of the field
Exp Paradigm: NA
 Open field test
 7-11 weeks
Protein expression level evidence1
Increased
Description: Increased actl6a expression in cerebellar and hippocampal extracts
Exp Paradigm: NA
 Western blot
 P0
Protein binding1
Increased
Description: Increased actl6a incorporation in baf complexes immunoprecipitated with nbaf-specific ss18l1 antibody from p0 cerebellar, cortical, or hippocampal nuclear extracts
Exp Paradigm: NA
 Western blot
 P0
Differential gene expression1
Abnormal
Description: Abnormally regulated genes geo accession no gse147056
Exp Paradigm: NA
 Rna sequencing
 E16.5
Protein-dna complex assembly1
Decreased
Description: Decreased nbaf complex stability with increased salt concentration
Exp Paradigm: NA
 Western blot
 E18.5
Mortality/lethality1
 No change
 Survival analysis
 7-11 weeks
Anxiety1
 No change
 Open field test
 7-11 weeks
Cortical thickness1
 No change
 Immunohistochemistry
 7-11 weeks
Inanimate object preference1
 No change
 Novel object interaction test
 7 weeks
 Not Reported:

M_ACTL6B_2_KO_HT

Category
Entity
Quantity
Experimental Paradigm
Age at Testing
Social interaction: with juveniles1
Decreased
Description: Decrease in interaction with juvenile
Exp Paradigm: NA
 Reciprocal social interaction test
 7 weeks
Social approach1
Decreased
Description: Decrease in social preference
Exp Paradigm: NA
 Three-chamber social approach test
 7-11 weeks
Protein-dna complex assembly1
Decreased
Description: Decreased nbaf complex stability with increased salt concentration
Exp Paradigm: NA
 Western blot
 E18.5
Protein binding1
Increased
Description: Increased actl6a incorporation in baf complexes immunoprecipitated with nbaf-specific ss18l1 antibody from p0 cerebellar, cortical, or hippocampal nuclear extracts
Exp Paradigm: NA
 Western blot
 P0
Mortality/lethality1
 No change
 Survival analysis
 7-11 weeks
Anxiety1
 No change
 Open field test
 7-11 weeks
Protein expression level evidence1
 No change
 Western blot
 P0
Cortical thickness1
 No change
 Immunohistochemistry
 7-11 weeks
Morphology and size of the corpus callosum1
 No change
 Immunohistochemistry
 7-11 weeks
Inanimate object preference1
 No change
 Novel object interaction test
 7 weeks
 Not Reported:

M_ACTL6B_3_KO_HM

Category
Entity
Quantity
Experimental Paradigm
Age at Testing
General locomotor activity: ambulatory activity1
Increased
Description: Increase in distance travelled
Exp Paradigm: NA
 Open field test
 12-16 weeks
Stereotypy1
Increased
Description: Increase in infrared beam breaks
Exp Paradigm: NA
 Open field test
 12-16 weeks
Social interaction1
Decreased
Description: Decreased adult social interaction
Exp Paradigm: NA
 Reciprocal social interaction test
 12-16 weeks
Anxiety1
Decreased
Description: Increase in time spent in the center of the field
Exp Paradigm: NA
 Open field test
 12-16 weeks
Spatial working memory1
Decreased
Description: Decreased time spent around the target hole
Exp Paradigm: NA
 Barnes maze test
 12-16 weeks
Reward reinforced choice behavior: short-term memory1
Decreased
Description: Decrease in the number of correct responses to reach food pellet dispenser
Exp Paradigm: NA
 T-maze test
 12-16 weeks
 Not Reported:

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