HELP     Sign In
Search

Relevance to Autism

Rare de novo missnese variants in the CACNA1D gene have been identified in ASD probands from the Simons Simplex Collection (ORoak et al., 2012; Iossifov et al., 2012).

Molecular Function

The encoded protein has low voltage-gated calcium channel activity. Mutations in this gene are responsible for primary aldosteronism with seizures and neurologic abnormalities (PASNA; OMIM 615474).

External Links

        

References

Type
Title
Type of Disorder
Associated Disorders
Author, Year
Primary
Calcium channel activation and self-biting in mice.
Self-biting
Positive Association
Common schizophrenia alleles are enriched in mutation-intolerant genes and in regions under strong background selection.
SCZ
Support
Exome sequencing of ion channel genes reveals complex profiles confounding personal risk assessment in epilepsy.
Epilepsy
Support
New gain-of-function mutation shows CACNA1D as recurrently mutated gene in autism spectrum disorders and epilepsy.
ASD, ID, epilepsy/seizures
Support
Autism-associated missense genetic variants impact locomotion and neurodevelopment in Caenorhabditis elegans.
ASD
Support
Association of CaV1.3 L-type calcium channels with Shank.
Support
Integrating de novo and inherited variants in 42
ASD
Support
An autism-associated mutation in CaV1.3 channels has opposing effects on voltage- and Ca(2)-dependent regulation.
Support
The Clinical and Genetic Features of Co-occurring Epilepsy and Autism Spectrum Disorder in Chinese Children.
ASD, epilepsy/seizures
Support
Contribution of CACNA1H Variants in Autism Spectrum Disorder Susceptibility
ASD
Support
Large-scale discovery of novel genetic causes of developmental disorders.
Unknown diagnosis
Support
Gating defects of disease-causing de novo mutations in Cav1.3 Ca2+ channels.
Support
Identification of ultra-rare disruptive variants in voltage-gated calcium channel-encoding genes in Japanese samples of schizophrenia and autism spectrum disorder
ASD, SCZ
Support
Somatic and germline CACNA1D calcium channel mutations in aldosterone-producing adenomas and primary aldosteronism.
Support
ASD
Support
Diagnostic exome sequencing of syndromic epilepsy patients in clinical practice.
Epilepsy/seizures
Support
Deletion of the voltage-gated calcium channel, Ca V 1.3, causes deficits in motor performance and associative learning
Support
De novo gene disruptions in children on the autistic spectrum.
ASD
Support
Primary aldosteronism with seizures and neurologic
Support
Targeted sequencing and functional analysis reveal brain-size-related genes and their networks in autism spectrum disorders.
ASD
Support
The landscape of somatic mutation in cerebral cortex of autistic and neurotypical individuals revealed by ultra-deep whole-genome sequencing
ASD
Support
Sporadic autism exomes reveal a highly interconnected protein network of de novo mutations.
ASD
Support
ASD, ID
BPD, MDD
Support
Rates, distribution and implications of postzygotic mosaic mutations in autism spectrum disorder.
ASD
Support
Identification of De Novo JAK2 and MAPK7 Mutations Related to Autism Spectrum Disorder Using Whole-Exome Sequencing in a Chinese Child and Adolesce...
ASD
Highly Cited
alpha 1D (Cav1.3) subunits can form l-type Ca2 channels activating at negative voltages.
Recent Recommendation
Synaptic, transcriptional and chromatin genes disrupted in autism.
ASD
Recent Recommendation
Functional roles of Cav1.3(alpha1D) calcium channels in atria: insights gained from gene-targeted null mutant mice.
Recent Recommendation
NA
ASD
Recent Recommendation
CACNA1D de novo mutations in autism spectrum disorders activate Cav1.3 L-type calcium channels.

Rare

Variant ID
Variant Type
Allele Change
Residue Change
Inheritance Pattern
Inheritance Association
Family Type
Author, Year
 GEN032R001 
 missense_variant 
 c.2306C>G 
 p.Ala769Gly 
 De novo 
  
 Simplex 
 GEN032R002 
 missense_variant 
 c.1219G>A 
 p.Gly407Arg 
 De novo 
  
 Simplex 
 GEN032R003 
 synonymous_variant 
 c.1023C>T 
 p.Asn341= 
 Unknown 
  
 Unknown 
 GEN032R004 
 synonymous_variant 
 c.2802C>T 
 p.Phe934= 
 Unknown 
  
 Unknown 
 GEN032R005 
 missense_variant 
 c.1615G>C 
 p.Ala539Pro 
 Unknown 
  
 Unknown 
 GEN032R006 
 missense_variant 
 c.3952C>T 
 p.Pro1318Ser 
 Unknown 
  
 Unknown 
 GEN032R007 
 missense_variant 
 c.5381T>A 
 p.Ile1794Asn 
 Unknown 
  
 Unknown 
 GEN032R008 
 intron_variant 
 c.4262+560C>T 
  
 Unknown 
  
 Unknown 
 GEN032R009 
 stop_gained 
 c.4897C>T 
 p.Gln1633Ter 
 Unknown 
  
 Unknown 
 GEN032R010 
 splice_site_variant 
 G>T 
  
 Unknown 
  
 Unknown 
 GEN032R011 
 missense_variant 
 c.1810G>A 
 p.Val604Ile 
 De novo 
  
 Simplex 
 GEN032R012 
 missense_variant 
 c.1261G>A 
 p.Asp421Asn 
 Familial 
 Paternal 
 Simplex 
 GEN032R013 
 missense_variant 
 c.1792G>A 
 p.Gly598Ser 
 Familial 
 Paternal 
 Simplex 
 GEN032R014 
 missense_variant 
 c.2789G>A 
 p.Arg930His 
 Familial 
 Paternal 
 Simplex 
 GEN032R015 
 missense_variant 
 c.5816G>A 
 p.Arg1939Gln 
 Familial 
 Paternal 
 Simplex 
 GEN032R016 
 missense_variant 
 c.6053C>A 
 p.Thr2018Asn 
 Familial 
 Maternal 
 Simplex 
 GEN032R017 
 missense_variant 
 c.2612T>G 
 p.Leu871Trp 
 Familial 
 Paternal 
 Simplex 
 GEN032R018 
 missense_variant 
 c.2612T>G 
 p.Leu871Trp 
 Familial 
 Maternal 
 Simplex 
 GEN032R019 
 missense_variant 
 c.3433C>G 
 p.Arg1145Gly 
 Familial 
 Paternal 
 Simplex 
 GEN032R020 
 missense_variant 
 c.3607C>T 
 p.Arg1203Cys 
 Familial 
 Paternal 
 Simplex 
 GEN032R021 
 missense_variant 
 c.176C>T 
 p.Ala59Val 
 Unknown 
  
 Unknown 
 GEN032R022 
 missense_variant 
 c.1033A>C 
 p.Thr345Pro 
 Unknown 
  
 Unknown 
 GEN032R023 
 missense_variant 
 c.2612T>G 
 p.Leu871Trp 
 Unknown 
  
 Unknown 
 GEN032R024 
 missense_variant 
 c.2789G>A 
 p.Arg930His 
 Unknown 
  
 Unknown 
 GEN032R025 
 missense_variant 
 c.3578G>A 
 p.Arg1193His 
 Unknown 
  
 Unknown 
 GEN032R026 
 missense_variant 
 c.5990C>G 
 p.Ser1997Trp 
 Unknown 
  
 Unknown 
 GEN032R027 
 missense_variant 
 c.6053C>A 
 p.Thr2018Asn 
 Unknown 
  
 Unknown 
 GEN032R028 
 missense_variant 
 c.6122G>A 
 p.Arg2041His 
 Unknown 
  
 Unknown 
 GEN032R029 
 missense_variant 
 c.826C>T 
 p.Leu276Phe 
 Unknown 
  
 Unknown 
 GEN032R030 
 missense_variant 
 c.1112A>C 
 p.Tyr371Ser 
 Unknown 
  
 Unknown 
 GEN032R031 
 missense_variant 
 c.1810G>A 
 p.Val604Ile 
 Unknown 
  
 Unknown 
 GEN032R032 
 missense_variant 
 c.3788T>C 
 p.Val1263Ala 
 Unknown 
  
 Unknown 
 GEN032R033 
 missense_variant 
 c.5023C>T 
 p.Arg1675Trp 
 Unknown 
  
 Unknown 
 GEN032R034 
 missense_variant 
 c.1105G>A 
 p.Val369Met 
 Unknown 
  
 Unknown 
 GEN032R035 
 missense_variant 
 c.6275G>A 
 p.Gly2092Glu 
 Unknown 
  
 Unknown 
 GEN032R036 
 missense_variant 
 c.2015C>T 
 p.Ser672Leu 
 De novo 
  
 Unknown 
 GEN032R037 
 missense_variant 
 c.1201G>C 
 p.Val401Leu 
 De novo 
  
  
 GEN032R038 
 missense_variant 
 c.1534T>G 
 p.Trp512Gly 
 De novo 
  
  
 GEN032R039 
 missense_variant 
 c.2206A>G 
 p.Met736Val 
 Familial 
  
 Simplex 
 GEN032R040 
 missense_variant 
 c.3187C>T 
 p.Arg1063Cys 
 Familial 
  
 Simplex 
 GEN032R041 
 missense_variant 
 c.3862G>A 
 p.Ala1288Thr 
 Familial 
  
 Simplex 
 GEN032R042 
 missense_variant 
 c.5846G>A 
 p.Arg1949His 
 Familial 
  
 Simplex 
 GEN032R043 
 missense_variant 
 c.790A>G 
 p.Ile264Val 
 Familial 
  
 Simplex 
 GEN032R044 
 missense_variant 
 c.920A>C 
 p.Asp307Ala 
 Familial 
  
 Simplex 
 GEN032R045 
 missense_variant 
 c.1493G>T 
 p.Arg498Leu 
 Unknown 
  
  
 GEN032R046 
 missense_variant 
 c.4444G>C 
 p.Val1482Leu 
 De novo 
  
  
 GEN032R047 
 missense_variant 
 c.4232C>T 
 p.Thr1411Met 
 De novo 
  
 Simplex 
 GEN032R048 
 missense_variant 
 ENSG00000157388:ENST00000422281:exon21:c.A2771G:p.Y924C,ENSG00000157388:ENST00000288139:exon22:c.A28 
  
 De novo 
  
  
 GEN032R049 
 stop_gained 
 c.4413T>A 
 p.Cys1471Ter 
 Unknown 
  
  
 GEN032R050 
 inframe_deletion 
 c.5017_5019del 
 p.Glu1673del 
 Unknown 
  
  
 GEN032R051 
 inframe_deletion 
 c.5017_5019del 
 p.Glu1673del 
 Unknown 
  
  
 GEN032R052 
 missense_variant 
 c.988G>C 
 p.Gly330Arg 
 Unknown 
  
  
 GEN032R053 
 missense_variant 
 c.2206A>G 
 p.Met736Val 
 Unknown 
  
  
 GEN032R054 
 missense_variant 
 c.2242G>A 
 p.Val748Ile 
 Unknown 
  
  
 GEN032R055 
 missense_variant 
 c.2242G>A 
 p.Val748Ile 
 Unknown 
  
  
 GEN032R056 
 missense_variant 
 G>A 
 p.Gly1438Glu 
 Familial 
 Paternal 
  
 GEN032R057 
 missense_variant 
 c.3039G>T 
 p.Arg1013Ser 
 De novo 
  
 Multiplex 
 GEN032R058 
 missense_variant 
 c.2015C>T 
 p.Ser672Leu 
 De novo 
  
  
 GEN032R059 
 missense_variant 
 c.2513A>G 
 p.Tyr838Cys 
 De novo 
  
  
 GEN032R060 
 missense_variant 
 c.3245A>G 
 p.Tyr1082Cys 
 De novo 
  
  
 GEN032R061 
 missense_variant 
 c.3506G>T 
 p.Gly1169Val 
 De novo 
  
  
 GEN032R062 
 missense_variant 
 c.4724G>C 
 p.Arg1575Pro 
 De novo 
  
  
 GEN032R063 
 synonymous_variant 
 c.6033G>A 
 p.Arg2011%3D 
 De novo 
  
  
 GEN032R064 
 missense_variant 
 c.2140G>T 
 p.Ala714Ser 
 De novo 
  
  
 GEN032R065 
 missense_variant 
 c.2015C>T 
 p.Ser672Leu 
 De novo 
  
  
 GEN032R066 
 missense_variant 
 c.2015C>T 
 p.Ser672Leu 
 Familial 
 Paternal 
 Multiplex 
 GEN032R067 
 missense_variant 
 c.4967G>A 
 p.Arg1656His 
 Unknown 
  
  
  et al.  

Common

Variant ID
Polymorphism
SNP ID
Allele Change
Residue Change
Population Origin
Population Stage
Author, Year
 GEN032C001 
 intergenic_variant 
 rs312477 
 G>A 
  
 40,675 SCZ cases and 64,643 controls (CLOZUK and independent PGC datasets) 
 Discovery 
Chromosome
CNV Locus
CNV Type
# of studies
Animal Model
3
Deletion
 7
 
3
Deletion
 2
 
3
Deletion
 2
 
3
Deletion
 1
 

Model Summary

Cacna1d encodes for Cav1.3, an L-type calcium channel expressed primarily in the adrenal glands. Human mutations that affect the channel properties are modeled in mouse. One mutation I770M is associated with primary aldosteronism, seizures, and neurologic abnormalities (PASNA), the other mutation A769G is a de novo mutation found in an individual with ASD. These mutations are considered gain-of-function mutations and result an increase in aldosterone, a decrease in body weight, social behavior and motor phenotypes. Isradipine is a calcium channel blocker that has a partially ameliorates some motor phenotypes, and aldosterone levels.

References

Type
Title
Author, Year
Primary
Additional
NA

M_CACNA1D_1_KI_HT_I770M

Model Type: Genetic
Model Genotype: Heterozygous
Mutation: CRISPR/Cas9-mediated mutagenesis in the Cacna1d gene was performed in mice (I772M) to model human mutation I770M, a gain-of-function CACNA1D mutation that causes a rare syndrome of primary aldosteronism, seizures, and neurologic abnormalities (PASNA). Mice were bred using in vitro fertilization, using C57BL/6J females as egg donors and sperm from heterozygous male Cacna1d^I772M/+ mice.
Allele Type: ASD mutation
Strain of Origin: C57BL/6J
Genetic Background: C57BL/6J
ES Cell Line: Not applicable
Mutant ES Cell Line:
Model Source: Ute I. Scholl

M_CACNA1D_2_KO_HM

Model Type: Genetic
Model Genotype: Homozygous
Mutation: Mice carrying knockout allele with 1 bp deletion, leading to the generation of a stop codon (p.Leu783*).
Allele Type: Knockout
Strain of Origin: Not specified
Genetic Background: Not specified
ES Cell Line: Not specified
Mutant ES Cell Line:
Model Source: Not specified

M_CACNA1D_3_KI_HM_A769G

Model Type: Genetic
Model Genotype: Homozygous
Mutation: A771G mutant mice were generated using CRISPR/Cas9 (MGI:7549317), to model human mutation, A769G, found in an ASD proband.
Allele Type: ASD mutation
Strain of Origin: C57BL/6N
Genetic Background: C57BL/6N
ES Cell Line: Not applicable
Mutant ES Cell Line:
Model Source: Joerg Striessnig

M_CACNA1D_4_KI_HT_A769G

Model Type: Genetic
Model Genotype: Heterozygous
Mutation: A771G mutant mice were generated using CRISPR/Cas9 (MGI:7549317), to model human mutation, A769G, found in an ASD proband.
Allele Type: ASD mutation
Strain of Origin: C57BL/6N
Genetic Background: C57BL/6N
ES Cell Line: Not applicable
Mutant ES Cell Line:
Model Source: Joerg Striessnig

M_CACNA1D_1_KI_HT_I770M

Category
Entity
Quantity
Experimental Paradigm
Age at Testing
Locomotor activity in diurnal cycle1
Increased
Description: In a home cage screening, behavior is automatically categorized based on video recordings of singly housed mice in cages for a period of 24 hours. I772M heterozygous mice show increased locomotion compared with wildtype mice particularly at the start of the dark period (active period of mice), with decreasing locomotion in the later phase of the dark period. A similar peak in activity was observed after the change to the light phase.
 Home cage behavior
 13-18 weeks
Tremor1
Increased
Description: Tremor was almost exclusively observed in I772M heterozygous mice: 36 of 37 in mutants versus 2 of 42 in wildtype.
Exp Paradigm: SHIRPA
 Neurological exam battery
 13-18 weeks
Grip strength1
Decreased
Description: I772M heterozygous mice spent less time hanging at the top of the cage lid in the home cage behavior screening.
 Home cage behavior
 13-18 weeks
Motor strength and endurance1
Decreased
Description: Male heterozygous knockin mice lack the strength and motor coordination for successful mating.
 General observations
 unreported
General locomotor activity: ambulatory activity1
Increased
Description: I772M heterozygous mice exhibited hyper-locomotion.
 Open field test
 13-18 weeks
Motor coordination and balance1
Decreased
Description: I772M heterozygous mice showed impaired coordination in the rotarod test, with reduced times spent on the rod.
 Accelerating rotarod test
 13-18 weeks
Grip strength1
Decreased
Description: I772M mice show reduced grip strength and grasping compared to wildtype.
Exp Paradigm: SHIRPA
 Neurological exam battery
 13-18 weeks
Seizures1
Increased
Description: Upon injection of ketamine and xylazine for anesthesia, 10 of 13 heterozygous knockin mice showed abnormal jerking movements and tonic-clonic seizures, while no seizures were observed in 19 wildtype mice with the same anesthesia treatment.
Exp Paradigm: anesthesia with ketamine and xylazine
 Observation of chemically induced seizures
 13-18 weeks
Social memory1
Increased
Description: I772M heterozygous mice again preferred a novel social stimulus (new unknown mouse) over a known social stimulus, whereas no significant preference was seen in wildtype littermates.
 Three-chamber social approach test
 13-18 weeks
Nest building behavior1
Decreased
Description: I772M heterozygous mice showed severely impaired nest building scores.
 Nest building assay
 13-18 weeks
Social interaction1
Increased
Description: In a social proximity test (forced social interaction in a constrained space), I772M heterozygous mice show significantly more nose-to-anogenital contacts, jump escape events, uprighting events, crawl-over events, and crawl under-events when in contact with an unknown mouse, but there was no significant increase in nose tip-to-nose tip contacts or nose-to-head contacts.
 Reciprocal social interaction test
 13-18 weeks
Social interaction1
Increased
Description: Exploration of the novel social stimulus is mostly limited to olfaction, manifesting as sniffing. Counting the number of sniffing events at the new unknown mouse relative to the known mouse revealed a similar pattern with no difference for wildtype but a preference for the novel stimulus in I772M heterozygous mice.
 Three-chamber social approach test
 13-18 weeks
Social approach1
Increased
Description: I772M heterozygous mice showed a marked preference for a novel social stimulus over a novel empty object, whereas no preference was seen for wildtype mice.
 Three-chamber social approach test
 13-18 weeks
Hormone levels1
Increased
Description: Serum aldosterone levels were significantly elevated in heterozygous knockin mice compared with wildtype.
Exp Paradigm: aldosterone
 ELISA
 13-18 weeks
Mortality/lethality: life span: incomplete penetrance1
Increased
Description: Heterozygous knockin mice show increased mortality between weaning and 100 days of age, after which no further lethality was observed.
 Survival analysis
 P21-P100
Tissue weight1
Decreased
Description: Heart, kidney and pancreas weight was decreased in heterozygous knockin male mice compared to wildtype.
 Measurement of tissue weight
 13-18 weeks
Size/growth1
Decreased
Description: Heterozygous knockin mice show decreased body weight.
 Body weight measurement
 13-18 weeks
Size/growth1
Decreased
Description: Heterozygous knockin mice show decreased body length.
 Body length measurement
 13-18 weeks
Maternal nurturing1
Decreased
Description: Offspring from female heterozygous knockin mice were found dead.
 General observations
 unreported
Calcium ion uptake1
Increased
Description: The pattern of calcium oscillations in the adrenal cortex was largely unaffected, which was measured using variable extracellular concentrations of potassium ion and angiotensin II (Ang II) to model effects of the two main stimuli of aldosterone production. Heterozygous knockin mice showed significantly elevated intracellular calcium levels compared with wildtype across almost all concentrations of Ang II and extracellular potassium.
Exp Paradigm: Fura-2
 Calcium imaging
 13-18 weeks
Adrenal cortex morphology1
 No change
 Histology
 13-18 weeks
Adrenal cortex morphology1
 No change
 In situ hybridization (ISH)
 13-18 weeks
Mortality/lethality: perinatal1
 No change
 General observations
 unreported
Tissue weight1
 No change
 Measurement of tissue weight
 13-18 weeks
Anxiety1
 No change
 Open field test
 13-18 weeks
Object recognition memory1
 No change
 Novel object recognition test
 13-18 weeks
Gene expression1
 No change
 Quantitative PCR (qRT-PCR)
 13-18 weeks
Brain morphology1
 No change
 Histology
 13-18 weeks
Brain size1
 No change
 Measurement of tissue weight
 13-18 weeks
Neuronal number1
 No change
 Histology
 13-18 weeks
Neuronal number: dopaminergic1
 No change
 In situ hybridization (ISH)
 13-18 weeks
Neuronal size1
 No change
 Histology
 13-18 weeks
Bioactive compound levels1
 No change
 ELISA
 13-18 weeks
Glucose levels1
 No change
 Measurement of blood glucose
 13-18 weeks
Hormone levels1
 No change
 ELISA
 13-18 weeks
Renal function1
 No change
 Urine/blood electrolyte analysis
 13-18 weeks
Seizures1
 No change
 Observation of seizures
 13-18 weeks
Olfaction1
 No change
 Buried food test
 13-18 weeks
Startle response: acoustic stimulus1
 No change
 Startle response test
 13-18 weeks
 Not Reported:

M_CACNA1D_2_KO_HM

Category
Entity
Quantity
Experimental Paradigm
Age at Testing
Mortality/lethality1
Increased
Description: No knockout mice were found from 147 mice at weaning from heterozygous backcrossing (25% homozygous offspring expected). At E8.5, one embryo out of 9 genotyped embryos was a homozygous knockout mouse.
 Genotypic ratio of progeny from heterozygous parents
 E8.5, P21
 Not Reported:

M_CACNA1D_3_KI_HM_A769G

Category
Entity
Quantity
Experimental Paradigm
Age at Testing
Locomotor activity in diurnal cycle1
Decreased
Description: Baseline activity was reduced during the first half of the dark phase in homozygous mutants, while it did not differ among genotypes during the light phase.
 Home cage behavior
 12-14 weeks
Hyperactivity1
Increased
Description: Homozygous mutant mice show an increased locomotion in response to handling.
 General observations
 12-14 weeks
General locomotor activity: ambulatory activity1
Increased
Description: Both the total distance traveled (2.3-fold) as well as the average velocity (1.5-fold) were significantly higher in homozygous mutant mice, which also spent significantly less time immobile (decreased by 70%).
 Open field test
 12-14 weeks
Rearing behavior1
Decreased
Description: Quantification of rearing behavior in a novel environment revealed similar time spent in rearing, whereas the ratio of time over frequency was significantly reduced in homozygous mutant mice.
 Novel cage test
 12-14 weeks
Repetitive digging1
Decreased
Description: Homozygous mice did not bury any marbles during the duration of the session, while wildtype mice buried ~12 out of 20 marbles.
 Marble-burying test
 12-14 weeks
Self grooming1
Decreased
Description: Quantification of grooming behavior in a novel environment revealed similar time spent in grooming, whereas the ratio of time over frequency was significantly reduced in homozygous mutant mice.
 Novel cage test
 12-14 weeks
Social interaction1
Decreased
Description: Quantification of direct nose-to-grid interaction time in each chamber revealed homozygous animals did not discriminate between the two grids.
 Three-chamber social approach test
 12-14 weeks
Social approach1
Decreased
Description: Wildtype mice displayed the expected preference to spend more time in the social chamber compared with the nonsocial one. Conversely, homozygous mice spent more time in the nonsocial chamber.
 Three-chamber social approach test
 12-14 weeks
Glucose levels1
Decreased
Description: Homozygous male mutants show significantly lower blood glucose levels at baseline.
 Glucose tolerance test
 26 weeks
Mortality/lethality: embryonic: incomplete penetrance1
Increased
Description: Mutant mice show reduced body weight that was already evident at birth. Weight gain in male and female mice was monitored weekly starting after weaning (4 weeks). At early developmental stages (4 weeks), heterozygous mutants weighed significantly less. At adulthood (11 weeks), the body weight of heterozygous mice had caught up. The reduced body weight was not associated with changes of other morphometric parameters such as body length.
 Genotypic ratio of progeny from heterozygous parents
 P0
Size/growth1
Decreased
Description: Mutant mice show reduced body weight that was already evident at birth. Weight gain in male and female mice was monitored weekly starting after weaning (4 weeks). At early developmental stages (4 weeks), Homozygous mutants weighed significantly less (a more pronounced difference than heterozygous mutants). Homozygous mutants consistently weighed less, even at 1 year of age. The reduced body weight was not associated with changes of other morphometric parameters such as body length.
 Body weight measurement
 4-52 weeks
Anxiety1
Increased
Description: In the light-dark box, where a higher light intensity was used (400 lux compared with 100â??150 lux in the other tests), homozygous mutant mice spent significantly less time in the lit compartment (decreased by 70%).
 Light-dark exploration test
 12-14 weeks
Anxiety1
 No change
 Open field test
 12-14 weeks
Anxiety1
 No change
 Elevated plus maze test
 12-14 weeks
Targeted expression1
 No change
 Western blot
 5-13 weeks
Brain morphology1
 No change
 Histology
 13-15 weeks
Brain size1
 No change
 Measurement of tissue weight
 5-26 weeks
Cortical lamination1
 No change
 Immunohistochemistry
 unreported
Morphology of the basal ganglia: striatum: caudoputamen1
 No change
 Histology
 unreported
Morphology of the basal ganglia: ventral striatum or nucleus accumbens1
 No change
 Histology
 unreported
Neuronal number: dopaminergic1
 No change
 Immunohistochemistry
 unreported
Neuronal number: Purkinje cells1
 No change
 Immunohistochemistry
 unreported
Glucose levels1
 No change
 Glucose tolerance test
 13 weeks
Hormone levels1
 No change
 ELISA
 13-15 weeks
 Not Reported:

M_CACNA1D_4_KI_HT_A769G

Category
Entity
Quantity
Experimental Paradigm
Age at Testing
Rearing behavior1
Decreased
Description: Quantification of rearing behavior in a novel environment revealed similar time spent in rearing, whereas the ratio of time over frequency was significantly reduced in heterozygous mutant mice.
 Novel cage test
 12-14 weeks
Hyperactivity1
Increased
Description: Heterozygous mutant mice show an increased locomotion in response to handling.
 General observations
 12-14 weeks
General locomotor activity: ambulatory activity1
Increased
Description: Both the total distance traveled (1.7-fold) as well as the average velocity (1.2-fold) were significantly higher in heterozygous mutant mice, which also spent significantly less time immobile (decreased by 50%).
 Open field test
 12-14 weeks
Action potential property: firing pattern1
Abnormal
Description: DMS-projecting mSN DA neurons displayed a significantly larger sag component (WT 13.3 mV versus HET 16.0 mV, P = 0.0395, unpaired Studentâ??s t test) associated with shorter rebound delay
 Whole-cell patch clamp
 12-17 weeks
Intrinsic membrane properties1
Abnormal
Description: Neither the time to action potential peak at the rheobase, cell capacitance, nor the action potential shape were affected in mutant slices.
 Whole-cell current clamp
 12-17 weeks
EPSP-spike relationship1
Increased
Description: Plotting the injected current versus the elicited number of action potential spikes (input-output curve) revealed a pronounced hyperexcitability of heterozygous medial spiny neurons.
 Whole-cell current clamp
 12-17 weeks
Action potential property: firing rate1
Increased
Description: The nigrostriatal pathway consist of dopamine neurons in the substantia nigra project either to the dorsal striatum. In ex vivo brain slices and in the absence of synaptic inputs, dopamine neurons showed spontaneous pacemaker firing. Brain slices from heterozygote animals, dopamine neuron populations (medial and lateral) fired with similar mean frequencies. Medial-projecting dopamine neurons expressing mutant Cav1.3 channels displayed a projection-selective gain-of-function phenotype with elevated baseline firing, while the pacemaker rate of lateral-projecting dopamine neurons was not altered by the presence of mutant channels.
 Whole-cell patch clamp
 12-17 weeks
Membrane potential1
Decreased
Description: Lower somatic current injections were required to depolarize the membrane and to elicit action potential firing.
 Whole-cell current clamp
 12-17 weeks
Social approach1
Decreased
Description: Wildtype mice displayed the expected preference to spend more time in the social chamber compared with the nonsocial one. Conversely, no significant preference was found for heterozygous mutants.
 Three-chamber social approach test
 12-14 weeks
Hormone levels1
Increased
Description: Plasma aldosterone levels of adult female heterozygous mutant mice increased by approximately 80%.
 ELISA
 13-15 weeks
Size/growth1
Decreased
Description: Mutant mice show reduced body weight that was already evident at birth. Weight gain in male and female mice was monitored weekly starting after weaning (4 weeks). At early developmental stages (4 weeks), heterozygous mutants weighed significantly less. At adulthood (11 weeks), the body weight of heterozygous mice had caught up. The reduced body weight was not associated with changes of other morphometric parameters such as body length.
 Body weight measurement
 4-52 weeks
Anxiety1
Increased
Description: In the light-dark box, where a higher light intensity was used (400 lux compared with 100â??150 lux in the other tests), heterozygous mutant mice spent significantly less time in the lit compartment (decreased by 50%).
 Light-dark exploration test
 12-14 weeks
Locomotor activity in diurnal cycle1
 No change
 Home cage behavior
 12-14 weeks
Mortality/lethality1
 No change
 Genotypic ratio of progeny from heterozygous parents
 P0
Anxiety1
 No change
 Open field test
 12-14 weeks
Anxiety1
 No change
 Elevated plus maze test
 12-14 weeks
Targeted expression1
 No change
 Western blot
 5-13 weeks
Brain morphology1
 No change
 Histology
 13-15 weeks
Brain morphology1
 No change
 Histology
 unreported
Brain size1
 No change
 Measurement of tissue weight
 5-26 weeks
Cortical lamination1
 No change
 Immunohistochemistry
 unreported
Morphology of the basal ganglia: striatum: caudoputamen1
 No change
 Histology
 unreported
Morphology of the basal ganglia: ventral striatum or nucleus accumbens1
 No change
 Histology
 unreported
Neuronal number: dopaminergic1
 No change
 Immunohistochemistry
 unreported
Neuronal number: Purkinje cells1
 No change
 Immunohistochemistry
 unreported
Action potential property: after hyperpolarization1
 No change
 Whole-cell current clamp
 12-17 weeks
Action potential property: amplitude1
 No change
 Whole-cell current clamp
 12-17 weeks
Action potential property: threshold1
 No change
 Whole-cell current clamp
 12-17 weeks
Glucose levels1
 No change
 Glucose tolerance test
 13-26 weeks
Hormone levels1
 No change
 ELISA
 13-15 weeks
Repetitive digging1
 No change
 Marble-burying test
 12-14 weeks
Self grooming1
 No change
 Novel cage test
 12-14 weeks
Social interaction1
 No change
 Three-chamber social approach test
 12-14 weeks
 Not Reported:

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