Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
1.
PLoS Genet ; 10(7): e1004454, 2014 Jul.
Article in English | MEDLINE | ID: mdl-25010494

ABSTRACT

Absence epilepsy (AE) is a common type of genetic generalized epilepsy (GGE), particularly in children. AE and GGE are complex genetic diseases with few causal variants identified to date. Gria4 deficient mice provide a model of AE, one for which the common laboratory inbred strain C3H/HeJ (HeJ) harbors a natural IAP retrotransposon insertion in Gria4 that reduces its expression 8-fold. Between C3H and non-seizing strains such as C57BL/6, genetic modifiers alter disease severity. Even C3H substrains have surprising variation in the duration and incidence of spike-wave discharges (SWD), the characteristic electroencephalographic feature of absence seizures. Here we discovered extensive IAP retrotransposition in the C3H substrain, and identified a HeJ-private IAP in the Pcnxl2 gene, which encodes a putative multi-transmembrane protein of unknown function, resulting in decreased expression. By creating new Pcnxl2 frameshift alleles using TALEN mutagenesis, we show that Pcnxl2 deficiency is responsible for mitigating the seizure phenotype - making Pcnxl2 the first known modifier gene for absence seizures in any species. This finding gave us a handle on genetic complexity between strains, directing us to use another C3H substrain to map additional modifiers including validation of a Chr 15 locus that profoundly affects the severity of SWD episodes. Together these new findings expand our knowledge of how natural variation modulates seizures, and highlights the feasibility of characterizing and validating modifiers in mouse strains and substrains in the post-genome sequence era.


Subject(s)
Epilepsy, Absence/genetics , Neoplasm Proteins/genetics , Receptors, AMPA/genetics , Seizures/genetics , Alleles , Animals , Chromosome Mapping , Disease Models, Animal , Epilepsy, Absence/pathology , Humans , Mice , Phenotype , Retroelements/genetics , Seizures/pathology
2.
J Physiol ; 591(1): 241-55, 2013 Jan 01.
Article in English | MEDLINE | ID: mdl-23090952

ABSTRACT

Mice deficient for CELF4, a neuronal RNA-binding protein, have a complex seizure disorder that includes both convulsive and non-convulsive seizures, and is dependent upon Celf4 gene dosage and mouse strain background. It was previously shown that Celf4 is expressed predominantly in excitatory neurons, and that deficiency results in abnormal excitatory synaptic neurotransmission. To examine the physiological and molecular basis of this, we studied Celf4-deficient neurons in brain slices. Assessment of intrinsic properties of layer V cortical pyramidal neurons showed that neurons from mutant heterozygotes and homozygotes have a lower action potential (AP) initiation threshold and a larger AP gain when compared with wild-type neurons. Celf4 mutant neurons also demonstrate an increase in persistent sodium current (I(NaP)) and a hyperpolarizing shift in the voltage dependence of activation. As part of a related study, we find that CELF4 directly binds Scn8a mRNA, encoding sodium channel Na(v)1.6, the primary instigator of AP at the axon initial segment (AIS) and the main carrier of I(NaP). In the present study we find that CELF4 deficiency results in a dramatic elevation in the expression of Na(v)1.6 protein at the AIS in both null and heterozygous neurons. Together these results suggest that activation of Na(v)1.6 plays a crucial role in seizure generation in this complex model of neurological disease.


Subject(s)
NAV1.6 Voltage-Gated Sodium Channel/physiology , RNA-Binding Proteins/physiology , Seizures/physiopathology , Animals , Brain/physiology , CELF Proteins , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Neurons/physiology , Tamoxifen/pharmacology
3.
PLoS Genet ; 8(11): e1003067, 2012.
Article in English | MEDLINE | ID: mdl-23209433

ABSTRACT

RNA-binding proteins have emerged as causal agents of complex neurological diseases. Mice deficient for neuronal RNA-binding protein CELF4 have a complex neurological disorder with epilepsy as a prominent feature. Human CELF4 has recently been associated with clinical features similar to those seen in mutant mice. CELF4 is expressed primarily in excitatory neurons, including large pyramidal cells of the cerebral cortex and hippocampus, and it regulates excitatory but not inhibitory neurotransmission. We examined mechanisms underlying neuronal hyperexcitability in Celf4 mutants by identifying CELF4 target mRNAs and assessing their fate in the absence of CELF4 in view of their known functions. CELF4 binds to at least 15%-20% of the transcriptome, with striking specificity for the mRNA 3' untranslated region. CELF4 mRNA targets encode a variety of proteins, many of which are well established in neuron development and function. While the overall abundance of these mRNA targets is often dysregulated in Celf4 deficient mice, the actual expression changes are modest at the steady-state level. In contrast, by examining the transcriptome of polysome fractions and the mRNA distribution along the neuronal cell body-neuropil axis, we found that CELF4 is critical for maintaining mRNA stability and availability for translation. Among biological processes associated with CELF4 targets that accumulate in neuropil of mutants, regulation of synaptic plasticity and transmission are the most prominent. Together with a related study of the impact of CELF4 loss on sodium channel Na(v)1.6 function, we suggest that CELF4 deficiency leads to abnormal neuronal function by combining a specific effect on neuronal excitation with a general impairment of synaptic transmission. These results also expand our understanding of the vital roles RNA-binding proteins play in regulating and shaping the activity of neural circuits.


Subject(s)
Epilepsy , Neurons , Protein Biosynthesis , RNA, Messenger , RNA-Binding Proteins , Animals , CELF Proteins , Cerebral Cortex/metabolism , Epilepsy/genetics , Epilepsy/metabolism , Hippocampus/metabolism , Humans , Mice , NAV1.6 Voltage-Gated Sodium Channel/metabolism , Neurons/cytology , Neurons/metabolism , Pyramidal Cells/metabolism , RNA Stability , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism , Synapses/genetics , Synapses/metabolism , Synaptic Transmission/genetics , Transcriptome
4.
Hum Mol Genet ; 20(5): 988-99, 2011 Mar 01.
Article in English | MEDLINE | ID: mdl-21159799

ABSTRACT

In a phenotype-driven mutagenesis screen, a novel, dominant mouse mutation, Nmf350, caused low seizure threshold, sporadic tonic-clonic seizures, brain enlargement and ectopic neurons in the dentate hilus and molecular layer of the hippocampus. Genetic mapping implicated Akt3, one of four candidates within the critical interval. Sequencing analysis revealed that mutants have a missense mutation in Akt3 (encoding one of three AKT/protein kinase B molecules), leading to a non-synonymous amino acid substitution in the highly conserved protein kinase domain. Previous knockout studies showed that Akt3 is pivotal in postnatal brain development, including a smaller brain, although seizures were not observed. In contrast to Akt3(Nmf350), we find that Akt3 null mice exhibit an elevated seizure threshold. An in vitro kinase assay revealed that Akt3(Nmf350) confers higher enzymatic activity, suggesting that Akt3(Nmf350) might enhance AKT signaling in the brain. In the dentate gyrus of Akt3(Nmf350) homozygotes, we also observed a modest increase in immunoreactivity of phosphorylated ribosomal protein S6, an AKT pathway downstream target. Together these findings suggest that Akt3(Nmf350) confers an increase of AKT3 activity in specific neuronal populations in the brain, and a unique dominant phenotype. Akt3(Nmf350) mice provide a new tool for studying physiological roles of AKT signaling in the brain, and potentially novel mechanisms for epilepsy.


Subject(s)
Disease Susceptibility , Mutation, Missense , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , Seizures/enzymology , Amino Acid Sequence , Animals , Cell Line , Female , Humans , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Molecular Sequence Data , Phosphorylation , Proto-Oncogene Proteins c-akt/chemistry , Ribosomal Protein S6 Kinases/genetics , Ribosomal Protein S6 Kinases/metabolism , Seizures/genetics , Sequence Alignment , Signal Transduction
5.
PLoS Genet ; 6(8)2010 Aug 05.
Article in English | MEDLINE | ID: mdl-20700442

ABSTRACT

Dynamin-1 (Dnm1) encodes a large multimeric GTPase necessary for activity-dependent membrane recycling in neurons, including synaptic vesicle endocytosis. Mice heterozygous for a novel spontaneous Dnm1 mutation--fitful--experience recurrent seizures, and homozygotes have more debilitating, often lethal seizures in addition to severe ataxia and neurosensory deficits. Fitful is a missense mutation in an exon that defines the DNM1a isoform, leaving intact the alternatively spliced exon that encodes DNM1b. The expression of the corresponding alternate transcripts is developmentally regulated, with DNM1b expression highest during early neuronal development and DNM1a expression increasing postnatally with synaptic maturation. Mutant DNM1a does not efficiently self-assemble into higher order complexes known to be necessary for proper dynamin function, and it also interferes with endocytic recycling in cell culture. In mice, the mutation results in defective synaptic transmission characterized by a slower recovery from depression after trains of stimulation. The DNM1a and DNM1b isoform pair is highly conserved in vertebrate evolution, whereas invertebrates have only one isoform. We speculate that the emergence of more specialized forms of DNM1 may be important in organisms with complex neuronal function.


Subject(s)
Dynamin I/genetics , Epilepsy/genetics , Exons , Alternative Splicing , Amino Acid Sequence , Animals , Cell Line , Conserved Sequence , Disease Models, Animal , Dynamin I/metabolism , Epilepsy/metabolism , Epilepsy/physiopathology , Female , Humans , Male , Mice , Mice, Inbred C57BL , Molecular Sequence Data , Mutation, Missense , Protein Isoforms/genetics , Protein Isoforms/metabolism , Sequence Alignment , Synaptic Transmission
6.
Hum Mol Genet ; 17(12): 1738-49, 2008 Jun 15.
Article in English | MEDLINE | ID: mdl-18316356

ABSTRACT

Absence epilepsy, characterized by spike-wave discharges (SWD) in the electroencephalogram, arises from aberrations within the circuitry of the cerebral cortex and thalamus that regulates awareness. The inbred mouse strain C3H/HeJ is prone to absence seizures, with a major susceptibility locus, spkw1, accounting for most of the phenotype. Here we find that spkw1 is associated with a hypomorphic retroviral-like insertion mutation in the Gria4 gene, encoding one of the four amino-3-hydroxy-5-methyl-4isoxazolepropionic acid (AMPA) receptor subunits in the brain. Consistent with this, Gria4 knockout mice also have frequent SWD and do not complement spkw1. In contrast, null mutants for the related gene Gria3 do not have SWD, and Gria3 loss actually lowers SWD of spkw1 homozygotes. Gria3 and Gria4 encode the predominant AMPA receptor subunits in the reticular thalamus, which is thought to play a central role in seizure genesis by inhibiting thalamic relay cells and promoting rebound burst firing responses. In Gria4 mutants, synaptic excitation of inhibitory reticular thalamic neurons is enhanced, with increased duration of synaptic responses-consistent with what might be expected from reduction of the kinetically faster subunit of AMPA receptors encoded by Gria4. These results demonstrate for the first time an essential role for Gria4 in the brain, and suggest that abnormal AMPA receptor-dependent synaptic activity can be involved in the network hypersynchrony that underlies absence seizures.


Subject(s)
Epilepsy, Absence/genetics , Receptors, AMPA/genetics , Receptors, AMPA/metabolism , Animals , Electroencephalography , Epilepsy, Absence/physiopathology , Mice , Mice, Inbred C3H , Mice, Knockout , Molecular Sequence Data , Synapses/physiology , Thalamus/physiology
7.
PLoS Genet ; 3(7): e124, 2007 Jul.
Article in English | MEDLINE | ID: mdl-17677002

ABSTRACT

Idiopathic epilepsy is a common human disorder with a strong genetic component, usually exhibiting complex inheritance. We describe a new mouse mutation in C57BL/6J mice, called frequent-flyer (Ff), in which disruption of the gene encoding RNA-binding protein Bruno-like 4 (Brunol4) leads to limbic and severe tonic-clonic seizures in heterozygous mutants beginning in their third month. Younger heterozygous adults have a reduced seizure threshold. Although homozygotes do not survive well on the C57BL/6J background, on mixed backgrounds homozygotes and some heterozygotes also display spike-wave discharges, the electroencephalographic manifestation of absence epilepsy. Brunol4 is widely expressed in the brain with enrichment in the hippocampus. Gene expression profiling and subsequent analysis revealed the down-regulation of at least four RNA molecules encoding proteins known to be involved in neuroexcitability, particularly in mutant hippocampus. Genetic and phenotypic assessment suggests that Brunol4 deficiency in mice results in a complex seizure phenotype, likely due to the coordinate dysregulation of several molecules, providing a unique new animal model of epilepsy that mimics the complex genetic architecture of common disease.


Subject(s)
Epilepsy/diagnosis , Epilepsy/genetics , Mutation , RNA-Binding Proteins/genetics , Animals , CELF Proteins , Disease Models, Animal , Electroencephalography , Gene Expression Profiling , Homozygote , Mice , Mice, Inbred C57BL , Mice, Transgenic , Molecular Sequence Data , Phenotype , Transgenes
8.
J Biol Chem ; 281(31): 22352-22359, 2006 Aug 04.
Article in English | MEDLINE | ID: mdl-16751186

ABSTRACT

The gene defective in fidget mice encodes fidgetin, a member of the AAA (ATPases associated with diverse cellular activities) family of ATPases. Using a yeast two-hybrid screen, we identified cAMP-dependent protein kinase A anchoring protein 95 kDa (AKAP95) as a potential fidgetin-binding protein. Epitope-tagged fidgetin co-localized with endogenous AKAP95 in the nuclear matrix, and the physical interaction between fidgetin and AKAP95 was further confirmed by reciprocal immunoprecipitation. To evaluate the biological significance of the fidgetin-AKAP95 binding, we created AKAP95 mutant mice through a gene trap strategy. Akap95 mutant mice are surprisingly viable with no overt phenotype. However, a significant number of mice carrying both Akap95 and fidget mutations die soon after birth due to cleft palate, consistent with the overlapping expression of AKAP95 and fidgetin in the branchial arches during mouse embryogenesis. These results expand the spectrum of the pleiotropic phenotypes of fidget mice and provide new leads on the in vivo function of AKAP95.


Subject(s)
Adenosine Triphosphatases/metabolism , DNA-Binding Proteins/metabolism , Morphogenesis , Nuclear Proteins/metabolism , Palate/growth & development , A Kinase Anchor Proteins , ATPases Associated with Diverse Cellular Activities , Adenosine Triphosphatases/genetics , Animals , Cleft Palate/genetics , DNA-Binding Proteins/genetics , Embryonic Development/genetics , Mice , Mice, Mutant Strains , Microtubule-Associated Proteins , Nuclear Matrix/metabolism , Nuclear Proteins/genetics , Phenotype , Protein Binding , Two-Hybrid System Techniques
9.
Exp Cell Res ; 304(1): 50-8, 2005 Mar 10.
Article in English | MEDLINE | ID: mdl-15707573

ABSTRACT

The mouse fidget mutation is an autosomal recessive mutation that renders reduced or absent semicircular canals, microphthalmia, and various skeletal abnormalities to affected mice. We previously identified the defective gene which encodes fidgetin, a new member of the ATPases associated with diverse cellular activities (AAA proteins). Here, we report on the subcellular localization of fidgetin as well as that of two closely related proteins, fidgetin-like 1 and fidgetin-like 2. Epitope-tagging and immunostaining revealed that both fidgetin and fidgetin-like 2 were predominantly localized to the nucleus, whereas fidgetin-like 1 was both nuclear and cytoplasmic. Furthermore, deletion studies identified a putative bipartite nuclear localization signal in the middle portion of the fidgetin protein. Since AAA proteins are known to form functional hetero- or homo-hexamers, we used reciprocal immunoprecipitation to examine the potential interaction among these proteins. We found that fidgetin interacted with itself and this specific interaction was abolished when either the N- or C-terminus of the protein was truncated. Taken together, our results suggest that fidgetin is a nuclear AAA-family protein with the potential to form homo-oligomers, thus representing the first step towards the elucidation of fidgetin's cellular function and the disease mechanism in fidget mutant mice.


Subject(s)
Adenosine Triphosphatases/genetics , Adenosine Triphosphatases/metabolism , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , ATPases Associated with Diverse Cellular Activities , Adenosine Triphosphatases/chemistry , Animals , Mice , Mice, Mutant Strains , Microtubule-Associated Proteins , Nuclear Proteins/chemistry
10.
Proc Natl Acad Sci U S A ; 102(6): 2123-8, 2005 Feb 08.
Article in English | MEDLINE | ID: mdl-15677329

ABSTRACT

The voltage-dependent calcium channel gamma4 subunit protein, CACNG4, is closely related to the gamma2 subunit, CACNG2. Both are expressed primarily in the brain and share 53% amino acid identity. The Cacng2 gene is disrupted in the stargazer mouse, with its distinctive phenotype including ataxia, frequent absence seizure episodes, and head elevation. A disruption within Cacng4 was engineered to assess its particular function. The homozygous Cacng4-targeted mutant mouse appeared normal with no ataxic gait or absence seizures, suggesting that other members of the gamma subunit family might functionally compensate for the absence of CACNG4. To test this hypothesis, the targeted Cacng4 mutation was combined with alleles of Cacng2. Absence seizures were observed in combination with the stargazer 3J mutation, which itself does not have seizures, and increased seizure activity was observed in combination with the waggler allele. Furthermore, within the corticothalamic loop, where absence seizures arise, CACNG4 expression is restricted to the thalamus. Our studies show that the CACNG4 protein has seizure suppressing activity, but this effect is revealed only when CACNG2 expression is also compromised, suggesting that CACNG subunits have in vivo overlapping functions.


Subject(s)
Calcium Channels/metabolism , Mice, Mutant Strains , Mutation , Protein Subunits/metabolism , Seizures , Animals , Brain/cytology , Brain/metabolism , Calcium Channels/genetics , Electrodes, Implanted , Electroencephalography , Embryo, Mammalian/anatomy & histology , Embryo, Mammalian/physiology , Female , Gene Expression Regulation, Developmental , Gene Targeting , Genotype , Male , Maze Learning , Mice , Phenotype , Protein Subunits/genetics , Rotarod Performance Test , Seizures/genetics
11.
Mamm Genome ; 14(8): 506-13, 2003 Aug.
Article in English | MEDLINE | ID: mdl-12925883

ABSTRACT

The stargazer mutant mouse is characterized by its ataxic gait, head tossing, and absence seizures. The mutation was identified in the gamma 2 subunit gene of the high voltage-dependent calcium channel, Cacng2. Subsequently, two allelic variants of stargazer have arisen, waggler and stargazer 3J. In this study, we have compared these new alleles to the original stargazer allele. All three mutations affect the Cacng2 mRNA levels as they all arise from disruptions within the introns of this gene. Our results show that the mutations cause reduced Cacng2 mRNA and protein levels. Stargazer and waggler mice have the least amount of mRNA and undetectable protein, whereas stargazer 3J appears to be the mildest allele, both in terms of the phenotype and protein expression. Electroencephalographic (EEG) analysis confirmed that stargazer has frequent spike-wave discharges (SWDs); the average duration of each discharge burst is 5 seconds and recurs every minute. The waggler allele causes a greater variation in SWD activity depending on the individual mouse, and the stargazer 3J mouse has no SWDs. The preliminary characterization of this heterogeneous allelic series provides a basis to explore more biochemical and physiological parameters relating to the role of the Cacng2 product in calcium channel activity, AMPA receptor localization, and cerebellar disturbances.


Subject(s)
Alleles , Calcium Channels/genetics , Calcium Channels/physiology , Phenotype , Action Potentials/genetics , Action Potentials/physiology , Animals , Blotting, Western , DNA Primers , Electroencephalography , Mice , Mice, Mutant Strains , RNA, Messenger/genetics , RNA, Messenger/physiology , Reverse Transcriptase Polymerase Chain Reaction
12.
Genomics ; 82(3): 254-60, 2003 Sep.
Article in English | MEDLINE | ID: mdl-12906850

ABSTRACT

It is hypothesized that autosomal retroposons compensate for the loss of their inactivated essential X-chromosome progenitors during spermatogenesis. Here we test this Retroposon Compensatory Mechanism (RCM) hypothesis using the Zfy gene family. The mouse autosomal retroposon Zfa is expressed in testes at the same developmental time points at which Zfx levels decline, which correspond to the time of male sex chromosome inactivation, suggesting that Zfa may compensate for the loss of Zfx during spermatogenesis. We examined the effect of Zfa-targeted mutagenesis on spermatogenesis in three genetically distinct mouse strains. Surprisingly, Zfa knockout mice showed no detectable fertility, sperm count, or testes morphology defects. We therefore conclude that Zfa is not an essential gene for spermatogenesis and fertility. This surprising finding now challenges the RCM hypothesis at least for the Zfy gene family. It also forces us to reevaluate the original data underpinning the RCM hypothesis for this family and to propose alternative hypotheses.


Subject(s)
DNA-Binding Proteins/genetics , Fertility/genetics , Retroelements , Spermatogenesis/genetics , Aging/genetics , Aging/physiology , Animals , DNA-Binding Proteins/metabolism , Epididymis/metabolism , Fertility/physiology , Gene Targeting , Male , Mice , Mice, Knockout , Spermatogenesis/physiology , Testis/metabolism
13.
Behav Brain Res ; 132(2): 145-58, 2002 May 14.
Article in English | MEDLINE | ID: mdl-11997145

ABSTRACT

A new spontaneous mouse mutation named fierce (frc) is deleted for the nuclear receptor Nr2e1 gene (also known as Tlx, mouse homolog of Drosophila tailless). The fierce mutation is genetically and phenotypically similar to Nr2e1 targeted mutations previously studied on segregating genetic backgrounds. However, we have characterized the fierce brain, eye, and behavioural phenotypes on three defined genetic backgrounds (C57BL/6J, 129P3/JEms, and B6129F1). The data revealed many novel and background-dependent phenotypic characteristics. Whereas abnormalities in brain development, hypoplasia of cerebrum and olfactory lobes, were consistent on all three backgrounds, our novel finding of enlarged ventricles in 100% and overt hydrocephalus in up to 30% of fierce mice were unique to the C57BL/6J background. Developmental eye abnormalities were also background-dependent with B6129F1-frc mice having less severe thinning of optic layers and less affected electroretinogram responses. Impaired regression of hyaloid vessels was observed in all backgrounds. Furthermore, retinal vessels were deficient in size and number in 129P3/JEms-frc and B6129F1-frc mice but almost entirely absent in C57BL/6J-frc mice. We present the first standardized behavioural tests conducted on Nr2e1 mutant mice and show that C57BL/6J-frc and B6129F1-frc mice have deficits in sensorimotor assays and are hyperaggressive in both sexes and backgrounds. However, C57BL/6J-frc mice were significantly more aggressive than B6129F1-frc mice. Overall, this extensive characterization of the fierce mutation is essential to its application for the study of behavioural, and brain and eye developmental disorders. In addition, the background-dependent differences revealed will enable the identification of important genetic modifiers.


Subject(s)
Aggression , Eye Abnormalities/genetics , Receptors, Cytoplasmic and Nuclear/genetics , Animals , Behavior, Animal/physiology , Blotting, Northern , Blotting, Southern , Brain/growth & development , Brain/pathology , Corticosterone/blood , Electroretinography , Eye Abnormalities/pathology , Female , Gene Deletion , Hearing/physiology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Motor Activity/physiology , Phenotype , Retina/abnormalities , Retina/pathology , Sexual Behavior, Animal/physiology , Smell/genetics , Testosterone/blood
SELECTION OF CITATIONS
SEARCH DETAIL
...