Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
Nat Neurosci ; 27(6): 1039, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38849586
2.
Nat Neurosci ; 27(5): 811, 2024 May.
Article in English | MEDLINE | ID: mdl-38724600
3.
Nat Neurosci ; 27(4): 606, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38589547
5.
Commun Biol ; 4(1): 591, 2021 05 13.
Article in English | MEDLINE | ID: mdl-33986447

Subject(s)
Deep Learning , Humans
6.
Neuropsychopharmacology ; 46(11): 2011-2020, 2021 10.
Article in English | MEDLINE | ID: mdl-33658654

ABSTRACT

Patients with SCN8A epileptic encephalopathy exhibit a range of clinical features, including multiple seizure types, movement disorders, and behavioral abnormalities, such as developmental delay, mild-to-severe intellectual disability, and autism. Recently, the de novo heterozygous SCN8A R1620L mutation was identified in an individual with autism, intellectual disability, and behavioral seizures without accompanying electrographic seizure activity. To date, the effects of SCN8A mutations that are primarily associated with behavioral abnormalities have not been studied in a mouse model. To better understand the phenotypic and functional consequences of the R1620L mutation, we used CRISPR/Cas9 technology to generate mice expressing the corresponding SCN8A amino acid substitution. Homozygous mutants exhibit tremors and a maximum lifespan of 22 days, while heterozygous mutants (RL/+) exhibit autistic-like behaviors, such as hyperactivity and learning and social deficits, increased seizure susceptibility, and spontaneous seizures. Current clamp analyses revealed a reduced threshold for firing action potentials in heterozygous CA3 pyramidal neurons and reduced firing frequency, suggesting that the R1620L mutation has both gain- and loss-of-function effects. In vivo calcium imaging using miniscopes in freely moving RL/+ mutants showed hyperexcitability of cortical excitatory neurons that is likely to increase seizure susceptibility. Finally, we found that oxcarbazepine and Huperzine A, a sodium channel blocker and reversible acetylcholinesterase inhibitor, respectively, were capable of conferring robust protection against induced seizures in RL/+ mutants. This mouse line will provide the opportunity to better understand the range of clinical phenotypes associated with SCN8A mutations and to develop new therapeutic approaches.


Subject(s)
Autistic Disorder , Epilepsy , Animals , Humans , Mice , Mutation/genetics , NAV1.6 Voltage-Gated Sodium Channel/genetics , Neurons , Seizures/genetics
8.
Hum Mol Genet ; 29(15): 2579-2595, 2020 08 29.
Article in English | MEDLINE | ID: mdl-32794569

ABSTRACT

GABAergic interneurons (GINs) are a heterogeneous population of inhibitory neurons that collectively contribute to the maintenance of normal neuronal excitability and network activity. Identification of the genetic regulatory elements and transcription factors that contribute toward GIN function may provide new insight into the pathways underlying proper GIN activity while also indicating potential therapeutic targets for GIN-associated disorders, such as schizophrenia and epilepsy. In this study, we examined the temporal changes in gene expression and chromatin accessibility during GIN development by performing transcriptomic and epigenomic analyses on human induced pluripotent stem cell-derived neurons at 22, 50 and 78 days (D) post-differentiation. We observed 13 221 differentially accessible regions (DARs) of chromatin that associate with temporal changes in gene expression at D78 and D50, relative to D22. We also classified families of transcription factors that are increasingly enriched at DARs during differentiation, indicating regulatory networks that likely drive GIN development. Collectively, these data provide a resource for examining the molecular networks regulating GIN functionality.


Subject(s)
Epigenome/genetics , GABAergic Neurons/metabolism , Interneurons/metabolism , Transcriptome/genetics , Cell Differentiation/genetics , Chromatin , Computational Biology , GABAergic Neurons/cytology , Humans , Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/metabolism , Interneurons/cytology , Transcription Factors/genetics
9.
Genes Brain Behav ; 19(4): e12612, 2020 04.
Article in English | MEDLINE | ID: mdl-31605437

ABSTRACT

Mutations in the voltage-gated sodium channel gene SCN8A cause a broad range of human diseases, including epilepsy, intellectual disability, and ataxia. Here we describe three mouse lines on the C57BL/6J background with novel, overlapping mutations in the Scn8a DIIS4 voltage sensor: an in-frame 9 bp deletion (Δ9), an in-frame 3 bp insertion (∇3) and a 35 bp deletion that results in a frameshift and the generation of a null allele (Δ35). Scn8a Δ9/+ and Scn8a ∇3/+ heterozygous mutants display subtle motor deficits, reduced acoustic startle response, and are resistant to induced seizures, suggesting that these mutations reduce activity of the Scn8a channel protein, Nav 1.6. Heterozygous Scn8a Δ35/+ mutants show no alterations in motor function or acoustic startle response, but are resistant to induced seizures. Homozygous mutants from each line exhibit premature lethality and severe motor impairments, ranging from uncoordinated gait with tremor (Δ9 and ∇3) to loss of hindlimb control (Δ35). Scn8a Δ9/Δ9 and Scn8a ∇3/∇3 homozygous mutants also exhibit impaired nerve conduction velocity, while normal nerve conduction was observed in Scn8a Δ35/Δ35 homozygous mice. Our results suggest that hypomorphic mutations that reduce Nav 1.6 activity will likely result in different clinical phenotypes compared to null alleles. These three mouse lines represent a valuable opportunity to examine the phenotypic impacts of hypomorphic and null Scn8a mutations without the confound of strain-specific differences.


Subject(s)
Movement , Mutation , NAV1.6 Voltage-Gated Sodium Channel/genetics , Action Potentials , Animals , Homozygote , Male , Mice , Mice, Inbred C57BL , NAV1.6 Voltage-Gated Sodium Channel/chemistry , Phenotype , Protein Domains
10.
Epilepsia ; 59(9): e135-e141, 2018 09.
Article in English | MEDLINE | ID: mdl-30132828

ABSTRACT

Previous reports have identified SLC6A1 variants in patients with generalized epilepsies, such as myoclonic-atonic epilepsy and childhood absence epilepsy. However, to date, none of the identified SLC6A1 variants has been functionally tested for an effect on GAT-1 transporter activity. The purpose of this study was to determine the incidence of SLC6A1 variants in 460 unselected epilepsy patients and to evaluate the impact of the identified variants on γ-aminobutyric acid (GABA)transport. Targeted resequencing was used to screen 460 unselected epilepsy patients for variants in SLC6A1. Five missense variants, one in-frame deletion, one nonsense variant, and one intronic splice-site variant were identified, representing a 1.7% diagnostic yield. Using a [3 H]-GABA transport assay, the seven identified exonic variants were found to reduce GABA transport activity. A minigene splicing assay revealed that the splice-site variant disrupted canonical splicing of exon 9 in the mRNA transcript, leading to premature protein truncation. These findings demonstrate that SLC6A1 is an important contributor to childhood epilepsy and that reduced GAT-1 function is a common consequence of epilepsy-causing SLC6A1 variants.


Subject(s)
Epilepsy/genetics , Epilepsy/metabolism , GABA Plasma Membrane Transport Proteins/metabolism , Gene Expression Regulation/genetics , Mutation/genetics , Cohort Studies , DNA Mutational Analysis , Female , GABA Plasma Membrane Transport Proteins/genetics , Genetic Predisposition to Disease/genetics , HEK293 Cells , HeLa Cells , Humans , Male , RNA, Messenger/metabolism , Transfection , Tritium/pharmacokinetics , gamma-Aminobutyric Acid/metabolism
11.
Hum Mol Genet ; 26(19): 3663-3681, 2017 10 01.
Article in English | MEDLINE | ID: mdl-28666327

ABSTRACT

A number of mutations in genes that encode ubiquitously expressed RNA-binding proteins cause tissue specific disease. Many of these diseases are neurological in nature revealing critical roles for this class of proteins in the brain. We recently identified mutations in a gene that encodes a ubiquitously expressed polyadenosine RNA-binding protein, ZC3H14 (Zinc finger CysCysCysHis domain-containing protein 14), that cause a nonsyndromic, autosomal recessive form of intellectual disability. This finding reveals the molecular basis for disease and provides evidence that ZC3H14 is essential for proper brain function. To investigate the role of ZC3H14 in the mammalian brain, we generated a mouse in which the first common exon of the ZC3H14 gene, exon 13 is removed (Zc3h14Δex13/Δex13) leading to a truncated ZC3H14 protein. We report here that, as in the patients, Zc3h14 is not essential in mice. Utilizing these Zc3h14Δex13/Δex13mice, we provide the first in vivo functional characterization of ZC3H14 as a regulator of RNA poly(A) tail length. The Zc3h14Δex13/Δex13 mice show enlarged lateral ventricles in the brain as well as impaired working memory. Proteomic analysis comparing the hippocampi of Zc3h14+/+ and Zc3h14Δex13/Δex13 mice reveals dysregulation of several pathways that are important for proper brain function and thus sheds light onto which pathways are most affected by the loss of ZC3H14. Among the proteins increased in the hippocampi of Zc3h14Δex13/Δex13 mice compared to control are key synaptic proteins including CaMK2a. This newly generated mouse serves as a tool to study the function of ZC3H14 in vivo.


Subject(s)
Brain/physiology , Nuclear Proteins/metabolism , RNA, Messenger/metabolism , RNA-Binding Proteins/metabolism , Animals , Brain/metabolism , Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Cell Nucleus/metabolism , Conserved Sequence , Exons , Female , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Models, Animal , Nuclear Proteins/genetics , Poly(A)-Binding Proteins , Protein Isoforms , RNA/metabolism , RNA, Messenger/genetics , RNA-Binding Proteins/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...