HELP     Sign In
Search

Relevance to Autism

A de novo loss-of-function (LoF) variant in the KMT5B gene (previously known as SUV420H1) was identified in an autistic proband from a simplex family in Iossifov et al., 2012 (PMID 22542183). In addition, two missense variants in the KMT5B gene that were predicted to be deleterious was also identified in ASD probands from the Simons Simplex Collection in Sanders et al., 2012 (PMID 22495306). Although population rate is not provided, many missense variants are listed in the NHLBI Exome Sequencing Project's Exome Variant Viewer for this gene. Two additional de novo LoF variants in this gene were identified in ASD probands from 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 identified KMT5B as a gene meeting high statistical significance with a FDR 0.01, meaning that this gene had a 99% chance of being a true autism gene (PMID 25363760). 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 KMT5B as a gene reaching exome-wide significance (P < 2.5E-06). Additional de novo variants in KMT5B have been identified in individuals with ASD and/or developmental delay/intellectual disability (Stessman et al., 2017; Yuen et al., 2017).

Molecular Function

Histone methyltransferase that specifically trimethylates 'Lys-20' of histone H4. H4 'Lys-20' trimethylation represents a specific tag for epigenetic transcriptional repression. Mainly functions in pericentric heterochromatin regions, thereby playing a central role in the establishment of constitutive heterochromatin in these regions. KMT5B is targeted to histone H3 via its interaction with RB1 family proteins (RB1, RBL1 and RBL2). Plays a role in myogenesis by regulating the expression of target genes, such as EID3.

External Links

        

References

Type
Title
Type of Disorder
Associated Disorders
Author, Year
Primary
De novo mutations revealed by whole-exome sequencing are strongly associated with autism.
ASD
Support
Prevalence and phenotypic impact of rare potentially damaging variants in autism spectrum disorder
ASD
Support
DD, ID
Support
Histone Lysine Methylases and Demethylases in the Landscape of Human Developmental Disorders.
DD, ID
ASD, epilepsy/seizures
Support
A single center experience with publicly funded clinical exome sequencing for neurodevelopmental disorders or multiple congenital anomalies
DD, ID
Support
Genomic diagnosis for children with intellectual disability and/or developmental delay.
ID
Support
Autism risk gene KMT5B deficiency in prefrontal cortex induces synaptic dysfunction and social deficits via alterations of DNA repair and gene transcription
ASD
Support
KMT5B is required for early motor development
Support
Whole genome sequencing resource identifies 18 new candidate genes for autism spectrum disorder
ASD
Support
Large-scale targeted sequencing identifies risk genes for neurodevelopmental disorders
ASD, DD
Support
Integrating de novo and inherited variants in 42
ASD
Support
De novo gene disruptions in children on the autistic spectrum.
ASD
Support
Large-Scale Exome Sequencing Study Implicates Both Developmental and Functional Changes in the Neurobiology of Autism
ASD
Support
Refining the Phenotypic Spectrum of KMT5B-Associated Developmental Delay
DD, ID
Epilepsy/seizures
Support
ADHD, DD
Support
Inherited and De Novo Genetic Risk for Autism Impacts Shared Networks.
ASD
Support
Loss-of-function of KMT5B leads to neurodevelopmental disorder and impairs neuronal development and neurogenesis
ASD
Support
DD, ID, epilepsy/seizures
Autistic features
Support
Inherited and multiple de novo mutations in autism/developmental delay risk genes suggest a multifactorial model.
ASD
Recent Recommendation
Targeted sequencing identifies 91 neurodevelopmental-disorder risk genes with autism and developmental-disability biases.
ASD, DD, ID
Recent Recommendation
Low load for disruptive mutations in autism genes and their biased transmission.
ASD
Recent Recommendation
Incorporating Functional Information in Tests of Excess De Novo Mutational Load.
Recent Recommendation
Synaptic, transcriptional and chromatin genes disrupted in autism.
ASD
Recent Recommendation
Craniosynostosis
ASD
Recent Recommendation
DD
ASD, ID, epilepsy/seizures
Recent Recommendation
Autism genes converge on asynchronous development of shared neuron classes
ASD

Rare

Variant ID
Variant Type
Allele Change
Residue Change
Inheritance Pattern
Inheritance Association
Family Type
Author, Year
 GEN336R001 
 missense_variant 
 c.791G>C 
 p.Trp264Ser 
 De novo 
  
 Simplex 
 GEN336R002 
 missense_variant 
 c.1538C>T 
 p.Ala513Val 
 De novo 
  
 Simplex 
 GEN336R003 
 splice_site_variant 
 c.461+1G>A 
  
 De novo 
  
 Simplex 
 GEN336R004 
 stop_gained 
 c.2347C>T 
 p.Arg783Ter 
 De novo 
  
 Simplex 
 GEN336R005 
 frameshift_variant 
 c.555_558del 
 p.Leu186GlufsTer26 
 De novo 
  
 Simplex 
 GEN336R006 
 missense_variant 
 c.2548A>G 
 p.Ile850Val 
 Unknown 
  
 Unknown 
 GEN336R007 
 missense_variant 
 c.1619G>A 
 p.Arg540Gln 
 Familial 
 Maternal 
  
 GEN336R008 
 frameshift_variant 
 c.725del 
 p.Leu242HisfsTer30 
 De novo 
  
  
 GEN336R009 
 frameshift_variant 
 c.1557_1558del 
 p.Asn520SerfsTer33 
 De novo 
  
  
 GEN336R010 
 frameshift_variant 
 c.572dup 
 p.Leu192ValfsTer3 
 Unknown 
  
  
 GEN336R011 
 missense_variant 
 c.676G>A 
 p.Asp226Asn 
 De novo 
  
  
 GEN336R012 
 frameshift_variant 
 NM_001300908.2:c.953_1028del;c.1157_1232del;c.1673_1748del 
 p.Asn387CysfsTer37 
 De novo 
  
 Multiplex 
 GEN336R013 
 missense_variant 
 c.2596T>C 
 p.Ser866Pro 
 De novo 
  
 Simplex 
 GEN336R014 
 stop_gained 
 c.1981G>T 
 p.Val661Leu 
 De novo 
  
  
 GEN336R015 
 frameshift_variant 
 c.-366del 
  
 De novo 
  
  
 GEN336R016 
 stop_gained 
 c.559C>T 
 p.Arg187Ter 
 De novo 
  
  
 GEN336R017 
 copy_number_loss 
  
  
 De novo 
  
  
 GEN336R018 
 copy_number_loss 
  
  
 De novo 
  
  
 GEN336R019 
 splice_site_variant 
 c.978-1G>A 
  
 Familial 
 Maternal 
 Simplex 
 GEN336R020 
 frameshift_variant 
 c.1205_1206insGCGTAAAA 
 p.Lys403ArgfsTer11 
 Familial 
 Paternal 
 Simplex 
 GEN336R021 
 frameshift_variant 
 c.-276-4_-276-1del 
  
 Familial 
 Maternal 
 Multi-generational 
 GEN336R022 
 frameshift_variant 
 c.459del 
 p.Phe154SerfsTer4 
 De novo 
  
 Simplex 
 GEN336R023 
 frameshift_variant 
 c.1150dup 
 p.Thr384AsnfsTer10 
 De novo 
  
 Simplex 
 GEN336R024 
 frameshift_variant 
 c.340del 
 p.Ser114GlnfsTer12 
 De novo 
  
 Simplex 
 GEN336R025 
 frameshift_variant 
 c.1279del 
 p.Glu427ArgfsTer22 
 De novo 
  
  
 GEN336R026 
 frameshift_variant 
 c.264_265del 
 p.Ala89SerfsTer13 
 De novo 
  
  
 GEN336R027 
 splice_site_variant 
 c.27+1G>A 
  
 De novo 
  
  
 GEN336R028 
 frameshift_variant 
 c.234_235del 
 p.Cys78Ter 
 De novo 
  
  
 GEN336R029 
 frameshift_variant 
 c.234_235del 
 p.Cys78Ter 
 De novo 
  
  
 GEN336R030 
 frameshift_variant 
 c.87del 
 p.Gln29HisfsTer12 
 Unknown 
  
  
 GEN336R031 
 initiator_codon_variant 
 c.2T>C 
 p.Met1? 
 Unknown 
  
  
 GEN336R032 
 missense_variant 
 c.2570G>A 
 p.Arg857His 
 Unknown 
  
  
 GEN336R033 
 missense_variant 
 c.973G>A 
 p.Glu325Lys 
 Unknown 
  
  
 GEN336R034 
 missense_variant 
 c.938G>A 
 p.Gly313Glu 
 Unknown 
  
 Simplex 
 GEN336R035 
 frameshift_variant 
 c.1572_1573del 
 p.Ala525SerfsTer28 
 Unknown 
  
  
 GEN336R036 
 frameshift_variant 
 c.983del 
 p.Gly328AlafsTer31 
 Unknown 
  
  
 GEN336R037 
 stop_lost 
 c.2656T>C 
 p.Ter886GlnextTer25 
 Unknown 
  
  
 GEN336R038 
 missense_variant 
 c.1618C>T 
 p.Arg540Trp 
 Unknown 
  
  
 GEN336R039 
 missense_variant 
 c.560G>A 
 p.Arg187Gln 
 Unknown 
  
  
 GEN336R040 
 missense_variant 
 c.1616G>A 
 p.Arg539Gln 
 Unknown 
  
  
 GEN336R041 
 missense_variant 
 c.2162G>A 
 p.Arg721His 
 Unknown 
  
  
 GEN336R042 
 missense_variant 
 c.833A>T 
 p.Asn278Ile 
 De novo 
  
 Simplex 
 GEN336R043 
 missense_variant 
 c.541C>G 
 p.His181Asp 
 De novo 
  
 Simplex 
 GEN336R044 
 frameshift_variant 
 c.1150dup 
 p.Thr384AsnfsTer11 
 Unknown 
  
  
 GEN336R045 
 frameshift_variant 
 c.340del 
 p.Ser114GlnfsTer12 
 Unknown 
  
  
 GEN336R046 
 missense_variant 
 c.788T>C 
 p.Leu263Pro 
 De novo 
  
 Simplex 
 GEN336R047 
 frameshift_variant 
 c.-126_-123del 
  
 De novo 
  
 Simplex 
 GEN336R048 
 synonymous_variant 
 c.1539G>A 
 p.Ala513%3D 
 De novo 
  
 Simplex 
 GEN336R049 
 inframe_deletion 
 c.715_717del 
 p.Glu239del 
 De novo 
  
  
 GEN336R050 
 splice_site_variant 
 c.-425+5G>A 
  
 De novo 
  
  
 GEN336R051 
 stop_gained 
 c.1843C>T 
 p.Arg615Ter 
 De novo 
  
  
 GEN336R052 
 frameshift_variant 
 c.541del 
 p.His181MetfsTer32 
 De novo 
  
  
 GEN336R053 
 missense_variant 
 c.220G>A 
 p.Ala74Thr 
 De novo 
  
  
 GEN336R054 
 frameshift_variant 
 c.391_394del 
 p.Lys131GlufsTer6 
 Unknown 
  
  
 GEN336R055 
 stop_gained 
 c.1282A>T 
 p.Lys428Ter 
 De novo 
  
 Simplex 
 GEN336R056 
 frameshift_variant 
 c.930del 
 p.Phe311SerfsTer48 
 Unknown 
  
  
 GEN336R057 
 missense_variant 
 c.818A>G 
 p.His273Arg 
 De novo 
  
  
 GEN336R058 
 missense_variant 
 c.598T>C 
 p.Cys200Arg 
 De novo 
  
  
 GEN336R059 
 missense_variant 
 c.254C>T 
 p.Thr85Ile 
 De novo 
  
  
 GEN336R060 
 missense_variant 
 c.1073T>C 
 p.Leu358Ser 
 Unknown 
  
  
 GEN336R061 
 frameshift_variant 
 c.397del 
 p.Cys133ValfsTer54 
 Unknown 
  
  
 GEN336R062 
 stop_gained 
 c.1057G>T 
 p.Glu353Ter 
 De novo 
  
  
 GEN336R063 
 splice_site_variant 
 c.304+1del 
  
 De novo 
  
  
 GEN336R064 
 missense_variant 
 c.742A>G 
 p.Asn248Asp 
 Unknown 
  
  
 GEN336R065 
 frameshift_variant 
 c.-58del 
  
 De novo 
  
  
 GEN336R066 
 copy_number_loss 
  
  
 De novo 
  
  
 GEN336R067 
 frameshift_variant 
 c.687_694del 
 p.Cys230ArgfsTer6 
 De novo 
  
  
 GEN336R068 
 frameshift_variant 
 c.1660_1661delinsC 
 p.Ser554ProfsTer13 
 De novo 
  
  
 GEN336R069 
 stop_gained 
 c.856C>T 
 p.Arg286Ter 
 De novo 
  
  
 GEN336R070 
 frameshift_variant 
 c.-94_-91del 
  
 De novo 
  
  
 GEN336R071 
 missense_variant 
 c.581G>A 
 p.Gly194Glu 
 De novo 
  
  
 GEN336R072 
 stop_gained 
 c.658C>T 
 p.Arg220Ter 
 De novo 
  
  
 GEN336R073 
 missense_variant 
 c.664G>T 
 p.Asp222Tyr 
 Familial 
 Maternal 
  
 GEN336R074 
 stop_gained 
 c.1183C>T 
 p.Arg395Ter 
 Unknown 
  
  
 GEN336R075 
 stop_gained 
 c.1183C>T 
 p.Arg395Ter 
 De novo 
  
  
 GEN336R076 
 stop_gained 
 c.2347C>T 
 p.Arg783Ter 
 Unknown 
  
  
 GEN336R077 
 missense_variant 
 c.1331C>G 
 p.Pro444Arg 
 Familial 
 Paternal 
  
 GEN336R078 
 missense_variant 
 c.813A>G 
 p.Ile271Met 
 Unknown 
  
  
 GEN336R079 
 missense_variant 
 c.220G>A 
 p.Ala74Thr 
 De novo 
  
  
 GEN336R080 
 inframe_deletion 
 c.199_201del 
 p.Glu67del 
 De novo 
  
  
 GEN336R081 
 missense_variant 
 c.697G>A 
 p.Glu233Lys 
 De novo 
  
  
 GEN336R082 
 frameshift_variant 
 c.650dup 
 p.Asn217LysfsTer6 
 De novo 
  
  
 GEN336R083 
 splice_site_variant 
 c.324+1_324+5del 
  
 De novo 
  
  
 GEN336R084 
 stop_gained 
 c.559C>T 
 p.Arg187Ter 
 De novo 
  
  
 GEN336R085 
 copy_number_loss 
 c.-76-3276_141del 
  
 De novo 
  
  
 GEN336R086 
 stop_gained 
 c.1183C>T 
 p.Arg395Ter 
 Familial 
 Maternal 
  
 GEN336R087 
 stop_gained 
 c.433A>T 
 p.Lys145Ter 
 De novo 
  
  
 GEN336R088 
 frameshift_variant 
 c.602del 
 p.Ser201IlefsTer29 
 De novo 
  
  
 GEN336R089 
 splice_site_variant 
 c.544-1G>A 
  
 De novo 
  
  
 GEN336R090 
 stop_gained 
 c.658C>T 
 p.Arg220Ter 
 De novo (germline mosaicism) 
  
 Multiplex 
 GEN336R091 
 missense_variant 
 c.904G>A 
 p.Glu302Lys 
 De novo 
  
  
 GEN336R092 
 frameshift_variant 
 c.2273_2274del 
 p.Tyr758CysfsTer5 
 De novo 
  
  
 GEN336R093 
 stop_gained 
 c.856C>T 
 p.Arg286Ter 
 De novo 
  
  
 GEN336R094 
 missense_variant 
 c.218C>T 
 p.Ser73Phe 
 Unknown 
 Not maternal 
  
 GEN336R095 
 missense_variant 
 c.1069C>G 
 p.Arg357Gly 
 De novo 
  
  
 GEN336R096 
 frameshift_variant 
 c.537_538del 
 p.Glu181AsnfsTer3 
 Unknown 
 Not maternal 
  
 GEN336R097 
 inframe_deletion 
 c.1175-6_1183del 
  
 De novo 
  
  
 GEN336R098 
 missense_variant 
 c.1285C>G 
 p.Leu429Val 
 Unknown 
  
  
  et al.  

Common

No Common Variants Available
Chromosome
CNV Locus
CNV Type
# of studies
Animal Model
11
Deletion-Duplication
 1
 
11
Deletion-Duplication
 18
 
11
Deletion
 1
 
11
Deletion
 3
 

Model Summary

KMT5B het KO mice show decrease in size starting at postnatal day 10 through young adulthood, decrease in brain size (i.e., microcephaly) which was more severe in males than females. delayed eye opening and significantly weaker reflexes in males, abnormal anxiety, depression, fear, extinction learning, increased grooming. Several sexually dimorphic differences were noted including increased repetitive grooming behavior in het females and an increased latency to hot plate response in HET females versus a decreased latency in HET males (Wickramasekara RN, Aut Res, 2021).

References

Type
Title
Author, Year
Primary
Differential effects by sex with Kmt5b loss
Additional
KMT5B is required for early motor development

M_KMT5B_1_KO_HT

Model Type: Genetic
Model Genotype: Heterozygous
Mutation: The gene trapping strategy makes Kmt5b^tm1a(KOMP)Wtsi mice effectively heterozygous for Kmt5b. Insertion of the gene trap cassette between Kmt5b exons 4 and 5 results in splicing from exon 4 to the en2 sa (splice acceptor) site in the cassette which drives the expression of the LacZ/Neo selectable marker immediately terminated by a polyadenylation signal. The construct creates an early truncation upstream of the Kmt5b functional set domain.
Allele Type: Knockout
Strain of Origin: C57BL/6N
Genetic Background: C57BL/6N
ES Cell Line: JM8.N4
Mutant ES Cell Line: NA
Model Source: Wellcome Trust Sanger Institute

M_KMT5B_2_KO_HM

Model Type: Genetic
Model Genotype: Homozygous
Mutation: The gene trapping strategy makes Kmt5b^tm1a(KOMP)Wtsi mice effectively heterozygous for Kmt5b. Insertion of the gene trap cassette between Kmt5b exons 4 and 5 results in splicing from exon 4 to the en2 sa (splice acceptor) site in the cassette which drives the expression of the LacZ/Neo selectable marker immediately terminated by a polyadenylation signal. The construct creates an early truncation upstream of the Kmt5b functional set domain.
Allele Type: Knockout
Strain of Origin: C57BL/6N
Genetic Background: C57BL/6N
ES Cell Line: JM8.N4
Mutant ES Cell Line: NA
Model Source: Wellcome Trust Sanger Institute

M_KMT5B_1_KO_HT

Category
Entity
Quantity
Experimental Paradigm
Age at Testing
Grip strength2
Decreased
Description: Male het but not female het mice show decreased grip strength; sexual dimorphism
 Grip strength test
 P10
Grip strength2
Decreased
Description: Male het animals were less capable of resisting the force of gravity with their fore and hindlimbs; sexual dimorphism
 Wire hang test
 P10
Righting response2
Decreased
Description: Linear regression analysis between body weight and time to right showed a significant correlation among het females; male het but not female het mice are significantly slower to right from a supine position; sexual dimorphism
 Righting reflex test
 P10
Brain size2
Decreased
Description: Microcephaly more severe in males than females; sexual dimorphism; more severe in males
 Gross necroscopy
 Adult
Brain size1
Increased
Description: Kmt5b male heterozygous mouse brains were significantly heavier compared to wildtype mice at P17 after normalization to body weight.
 Measurement of tissue weight
 P17
Morphology of neuromuscular junction1
Abnormal
Description: Neuromuscular junction (NMJ) occupancy (defined as the ratio of nerve/vesicular area vs. muscular NMJ area) did not significantly differ in the soleus (SOL) or the extensor digitorum longus (EDL) of Kmt5b heterozygotes compared to wildtype mice. No significant differences were identified in nerve/vesicular NMJ area or continuity in either muscle. In the muscle portion of the NMJ, SOL NMJ areas did not differ, but heterozygote NMJs had increased discontinuity compared to wildtype NMJs. A trend toward increased discontinuity among heterozygotes was noted in the EDL but was not statistically significant.
 Immunohistochemistry
 P44
Self grooming: artificial stress evoked2
Increased
Description: Increased number head grooming initiations in het females compared to wt females after stress stimulus ; no change in novelty-induced or stress-induced grooming; repeated head-body grooming pattern increased in het compared to wt females; sexual dimorphism
 Grooming behavior assessments
 Adult
Pain or nociception2
Increased
Description: Hets had increased freezing time following foot shock in both sexes
 Foot shock test
 Adult
Pain or nociception: thermal2
Increased
Description: Male hets had a significant decrease in latency
 Hot plate test
 Adult
Pain or nociception: thermal2
Decreased
Description: Female hets showed an increase in latency
 Hot plate test
 Adult
Social memory2
Decreased
Description: No social preference for novel compared to familiar mouse in het females; social dimorphism
 Three-chamber social approach test
 Adult
Reproductive function2
Decreased
Description: Intercrosses of het mice were often non-productive and produced fewer viable offspring than het x wt mating
 Genotypic ratio of progeny from heterozygous parents
 Adult
Skeletal muscle morphology1
Decreased
Description: There was a significant decrease in soleus muscle (SOL) weight in Kmt5b heterozygous male mice but no differences in extensor digitorum longus muscle (EDL) weight compared to wildtype mice at P17. In addition, both SOL and EDL muscles in Kmt5b heterozygous mice weighed less than in wildtype mice at P44. After normalization to body weight, SOL muscle weight of Kmt5b heterozygous male mice at P17 were much lower compared to wildtype male mice, but no differences were seen in normalized EDL weights of mice. There were no significant differences in normalized P44 SOL weights of Kmt5b heterozygous mice. However, female heterozygous normalized P44 EDL weights remained modestly lower after post hoc testing.
 Measurement of tissue weight
 P17, P44
Progressive muscle weakness1
Increased
Description: There were no significant differences in neuromuscular strength in Kmt5b heterozygous mice at P17. However, Kmt5b heterozygous mice displayed less neuromuscular strength compared to wildtype mice at P44.
 Grip strength test
 P17, P44
Size/growth1
Decreased
Description: Kmt5b heterozygous mice displayed significantly decreased body weights compared to wildtype mice.
 Body weight measurement
 unreported
Muscular fiber morphology1
Abnormal
Description: In the slow-twitch soleus (SOL) muscle, there were no differences in myofiber number at P17. There were no significant differences among relative proportions either by count or area of myofiber compositions of either male or female SOL or extensor digitorum longus (EDL) muscles of Kmt5b heterozygous and wildtype mice at P17. However, P44 Kmt5b heterozygous females had increased SOL type IIa myofiber number compared to wildtype females. In the fast-twitch EDL muscle, Kmt5b heterozygous mice had significantly greater type IIb myofiber number at P44 compared to wildtype mice. Kmt5b heterozygous females had increased EDL type IIb myofibers at P44 compared to wildtype females. There were no significant differences among relative proportions either by count or ar
 Immunohistochemistry
 P17, P44
Size/growth2
Decreased
Description: Decreased body weight in both male and female hets; more significant in male
 Body weight measurement
 P10-P44
Developmental trajectory2
Decreased
Description: No differences in incisor eruption, pinna folding, or vaginal opening in females, delayed eye opening primarily in males, sexual dimorphism
 Developmental milestone measurements
 P0-P21
Size/growth2
Decreased
Description: Decreased body length in both male and female hets at p44; driven primarily by the male hets
 Body length measurement
 P10-P44
Anxiety2
Decreased
Description: Reduced preference for the dark box
 Light-dark exploration test
 Adult
Extinction learning2
Decreased
Description: Extinction learning may be affected in het males but not females; sexual dimorphism
 Fear conditioning test
 Adult
Chromatin modification: Histone modification: Open chromatin2
Increased
Description: H4k20me2 and h4k20me3 protein levels were not significantly different; h4k20me1 levels were increased
 Western blot
 E14.5
Targeted expression1
Decreased
Description: Kmt5b heterozygous mice displayed decreased gene expression of Kmt5b on mRNA extracted from soleus (SOL) and extensor digitorum longus (EDL) hindlimb muscles.
 Quantitative PCR (qRT-PCR)
 P17
Targeted expression2
Decreased
Description: Decreased kmt5b transcripts
 Quantitative PCR (qRT-PCR)
 E14.5
Cardiovascular development and function1
 No change
 Measurement of tissue weight
 P17
Digestive system morphology: liver1
 No change
 Measurement of tissue weight
 P17
Renal morphology1
 No change
 Measurement of tissue weight
 P17
Anxiety2
 No change
 Elevated plus maze test
 Adult
Anxiety2
 No change
 Open field test
 Adult
Depression2
 No change
 Forced swim test
 Adult
Cued or contextual fear conditioning: Memory of context2
 No change
 Fear conditioning test
 Adult
Cued or contextual fear conditioning: Memory of cue2
 No change
 Fear conditioning test
 Adult
Spatial learning2
 No change
 Barnes maze test
 Adult
Gene expression1
 No change
 Quantitative PCR (qRT-PCR)
 P17
Clasping reflex2
 No change
 Grasping reflex test
 P10
General locomotor activity2
 No change
 Open field test
 Adult
General locomotor activity2
 No change
 Elevated plus maze test
 Adult
General locomotor activity: Ambulatory activity2
 No change
 Open field test
 Adult
Grip strength2
 No change
 Grip strength test
 P10
Grip strength2
 No change
 Wire hang test
 P10
Motor coordination and balance2
 No change
 Accelerating rotarod test
 Adult
Motor coordination and balance2
 No change
 Balance beam test
 Adult
Motor learning2
 No change
 Accelerating rotarod test
 Adult
Repetitive digging2
 No change
 Marble-burying test
 Adult
Olfaction2
 No change
 Olfactory habituation-dishabituation test
 Adult
Olfaction2
 No change
 Buried food test
 Adult
Social approach2
 No change
 Three-chamber social approach test
 Adult
 Not Reported:

M_KMT5B_2_KO_HM

Category
Entity
Quantity
Experimental Paradigm
Age at Testing
Mortality/lethality: neonatal1
Increased
Description: Reduced mendelian ratios for the ko genotype suggesting late-term or perinatal lethality;
 Genotypic ratio of progeny from heterozygous parents
 P0-P21
Targeted expression1
Decreased
Description: Decreased kmt5b transcripts
 Quantitative PCR (qRT-PCR)
 E14.5
Chromatin modification: Histone modification: Open chromatin1
Increased
Description: H4k20me2 and h4k20me3 protein levels were not significantly different; h4k20me1 levels were increased
 Western blot
 E14.5
General characteristics1
 No change
 General observations
 E18.5
 Not Reported:

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