Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
Add more filters










Publication year range
1.
J Biol Chem ; 299(11): 105299, 2023 11.
Article in English | MEDLINE | ID: mdl-37777156

ABSTRACT

Microbes living in the intestine can regulate key signaling processes in the central nervous system that directly impact brain health. This gut-brain signaling axis is partially mediated by microbe-host-dependent immune regulation, gut-innervating neuronal communication, and endocrine-like small molecule metabolites that originate from bacteria to ultimately cross the blood-brain barrier. Given the mounting evidence of gut-brain crosstalk, a new therapeutic approach of "psychobiotics" has emerged, whereby strategies designed to primarily modify the gut microbiome have been shown to improve mental health or slow neurodegenerative diseases. Diet is one of the most powerful determinants of gut microbiome community structure, and dietary habits are associated with brain health and disease. Recently, the metaorganismal (i.e., diet-microbe-host) trimethylamine N-oxide (TMAO) pathway has been linked to the development of several brain diseases including Alzheimer's, Parkinson's, and ischemic stroke. However, it is poorly understood how metaorganismal TMAO production influences brain function under normal physiological conditions. To address this, here we have reduced TMAO levels by inhibiting gut microbe-driven choline conversion to trimethylamine (TMA), and then performed comprehensive behavioral phenotyping in mice. Unexpectedly, we find that TMAO is particularly enriched in the murine olfactory bulb, and when TMAO production is blunted at the level of bacterial choline TMA lyase (CutC/D), olfactory perception is altered. Taken together, our studies demonstrate a previously underappreciated role for the TMAO pathway in olfactory-related behaviors.


Subject(s)
Olfactory Perception , Animals , Mice , Bacteria/metabolism , Choline/metabolism , Methylamines/metabolism , Female , Mice, Inbred C57BL
2.
Biomed Opt Express ; 12(8): 4901-4919, 2021 Aug 01.
Article in English | MEDLINE | ID: mdl-34513232

ABSTRACT

Stroke is a leading cause of disability in the Western world. Current post-stroke rehabilitation treatments are only effective in approximately half of the patients. Therefore, there is a pressing clinical need for developing new rehabilitation approaches for enhancing the recovery process, which requires the use of appropriate animal models. Here, we demonstrate the use of nonlinear microscopy of calcium sensors in the rat brain to study the effects of ischemic stroke injury on cortical activity patterns. We longitudinally recorded from thousands of neurons labeled with a genetically-encoded calcium indicator before and after an ischemic stroke injury in the primary motor cortex. We show that this injury has an effect on the activity patterns of neurons not only in the motor and somatosensory cortices, but also in the more distant visual cortex, and that these changes include modified firing rates and kinetics of neuronal activity patterns in response to a sensory stimulus. Changes in neuronal population activity provided animal-specific, circuit-level information on the post-stroke cortical reorganization process, which may be essential for evaluating the efficacy of new approaches for enhancing the recovery process.

3.
eNeuro ; 7(3)2020.
Article in English | MEDLINE | ID: mdl-32327468

ABSTRACT

Several genes are associated with increased risk for autism spectrum disorder (ASD), neurodevelopmental disorders that present with repetitive movements and restricted interests along with deficits in social interaction/communication. While genetic alterations associated with ASD are present early in life, ASD-like behaviors are difficult to detect in early infancy. This raises the issue of whether reversal of an ASD-associated genetic alteration early in life can prevent the onset of ASD-like behaviors. Genetic alterations of SHANK3, a well-characterized gene encoding a postsynaptic scaffolding protein, are estimated to contribute to ∼0.5% of ASD and remain one of the more replicated and well-characterized genetic defects in ASD. Here, we investigate whether early genetic reversal of a Shank3 mutation can prevent the onset of ASD-like behaviors in a mouse model. Previously, we have demonstrated that mice deficient in Shank3 display a wide range of behavioral abnormalities such as repetitive grooming, social deficits, anxiety, and motor abnormalities. In this study, we replicate many of these behaviors in Shank3 mutant mice. With early genetic restoration of wild-type (WT) Shank3, we rescue behaviors including repetitive grooming and social, locomotor, and rearing deficits. Our findings support the idea that the underlying mechanisms involving ASD behaviors in mice deficient in Shank3 are susceptible to early genetic correction of Shank3 mutations.


Subject(s)
Autism Spectrum Disorder , Animals , Autism Spectrum Disorder/genetics , Disease Models, Animal , Mice , Mice, Knockout , Microfilament Proteins , Nerve Tissue Proteins/genetics , Phenotype
4.
Autism Res ; 11(2): 234-244, 2018 02.
Article in English | MEDLINE | ID: mdl-29028156

ABSTRACT

Neuroligin-3 (NLGN3) is a postsynaptic cell adhesion protein that interacts with presynaptic ligands including neurexin-1 (NRXN1) [Ichtchenko et al., Journal of Biological Chemistry, 271, 2676-2682, 1996]. Mice harboring a mutation in the NLGN3 gene (NL3R451C) mimicking a mutation found in two brothers with autism spectrum disorder (ASD) were previously generated and behaviorally phenotyped for autism-related behaviors. In these NL3R451C mice generated and tested on a hybrid C57BL6J/129S2/SvPasCrl background, we observed enhanced spatial memory and reduced social interaction [Tabuchi et al., Science, 318, 71-76, 2007]. Curiously, an independently generated second line of mice harboring the same mutation on a C57BL6J background exhibited minimal aberrant behavior, thereby providing apparently discrepant results. To investigate the origin of the discrepancy, we previously replicated the original findings of Tabuchi et al. by studying the same NL3R451C mutation on a pure 129S2/SvPasCrl genetic background. Here we complete the behavioral characterization of the NL3R451C mutation on a pure C57BL6J genetic background to determine if background genetics play a role in the discrepant behavioral outcomes involving NL3R451C mice. NL3R451C mutant mice on a pure C57BL6J background did not display spatial memory enhancements or social interaction deficits. We only observed a decreased startle response and mildly increased locomotor activity in these mice suggesting that background genetics influences behavioral outcomes involving the NL3R451C mutation. Autism Res 2018, 11: 234-244. © 2017 International Society for Autism Research, Wiley Periodicals, Inc. LAY SUMMARY: Behavioral symptoms of autism can be highly variable, even in cases that involve identical genetic mutations. Previous studies in mice with a mutation of the Neuroligin-3 gene showed enhanced learning and social deficits. We replicated these findings on the same and different genetic backgrounds. In this study, however, the same mutation in mice on a different genetic background did not reproduce our previous findings. Our results suggest that genetic background influences behavioral symptoms of this autism-associated mutation.


Subject(s)
Autism Spectrum Disorder/genetics , Cell Adhesion Molecules, Neuronal/genetics , Membrane Proteins/genetics , Nerve Tissue Proteins/genetics , Problem Behavior/psychology , Animals , Autism Spectrum Disorder/psychology , Disease Models, Animal , Female , Genetic Background , Interpersonal Relations , Learning/physiology , Male , Mental Recall/physiology , Mice , Mice, Inbred C57BL , Mice, Mutant Strains , Mutation , Phenotype , Reflex, Startle/genetics , Spatial Learning/physiology
5.
Autism Res ; 10(1): 42-65, 2017 Jan.
Article in English | MEDLINE | ID: mdl-27492494

ABSTRACT

Mutations/deletions in the SHANK3 gene are associated with autism spectrum disorders and intellectual disability. Here, we present electrophysiological and behavioral consequences in novel heterozygous and homozygous mice with a transcriptional stop cassette inserted upstream of the PDZ domain-coding exons in Shank3 (Shank3E13 ). Insertion of a transcriptional stop cassette prior to exon 13 leads to loss of the two higher molecular weight isoforms of Shank3. Behaviorally, both Shank3E13 heterozygous (HET) and homozygous knockout (KO) mice display increased repetitive grooming, deficits in social interaction tasks, and decreased rearing. Shank3E13 KO mice also display deficits in spatial memory in the Morris water maze task. Baseline hippocampal synaptic transmission and short-term plasticity are preserved in Shank3E13 HET and KO mice, while both HET and KO mice exhibit impaired hippocampal long-term plasticity. Additionally, Shank3E13 HET and KO mice display impaired striatal glutamatergic synaptic transmission. These results demonstrate for the first time in this novel Shank3 mutant that both homozygous and heterozygous mutation of Shank3 lead to behavioral abnormalities with face validity for autism along with widespread synaptic dysfunction. Autism Res 2017, 10: 42-65. © 2016 International Society for Autism Research, Wiley Periodicals, Inc.


Subject(s)
Autistic Disorder/genetics , Autistic Disorder/physiopathology , Corpus Striatum/physiopathology , Hippocampus/physiopathology , Mutation/genetics , Nerve Tissue Proteins/genetics , Animals , Behavior, Animal , Blotting, Western , Disease Models, Animal , Exons , Female , Male , Mice , Mice, Knockout , Microfilament Proteins , Reproducibility of Results , Sequence Deletion , Synaptic Transmission/physiology
6.
Neurobiol Dis ; 85: 81-92, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26459112

ABSTRACT

Absence seizures occur in several types of human epilepsy and result from widespread, synchronous feedback between the cortex and thalamus that produces brief episodes of loss of consciousness. Genetic rodent models have been invaluable for investigating the pathophysiological basis of these seizures. Here, we identify tetratricopeptide-containing Rab8b-interacting protein (TRIP8b) knockout mice as a new model of absence epilepsy, featuring spontaneous spike-wave discharges on electroencephalography (EEG) that are the electrographic hallmark of absence seizures. TRIP8b is an auxiliary subunit of the hyperpolarization-activated cyclic-nucleotide-gated (HCN) channels, which have previously been implicated in the pathogenesis of absence seizures. In contrast to mice lacking the pore-forming HCN channel subunit HCN2, TRIP8b knockout mice exhibited normal cardiac and motor function and a less severe seizure phenotype. Evaluating the circuit that underlies absence seizures, we found that TRIP8b knockout mice had significantly reduced HCN channel expression and function in thalamic-projecting cortical layer 5b neurons and thalamic relay neurons, but preserved function in inhibitory neurons of the reticular thalamic nucleus. Our results expand the known roles of TRIP8b and provide new insight into the region-specific functions of TRIP8b and HCN channels in constraining cortico-thalamo-cortical excitability.


Subject(s)
Cerebral Cortex/physiopathology , Epilepsy, Absence/physiopathology , Membrane Proteins/deficiency , Neurons/physiology , Thalamus/physiopathology , Animals , Blotting, Western , Disease Models, Animal , Electrocardiography , Electrocorticography , Electrodes, Implanted , Epilepsy, Absence/genetics , Immunohistochemistry , Male , Membrane Potentials/physiology , Membrane Proteins/genetics , Mice, Knockout , Motor Activity/physiology , Patch-Clamp Techniques , Peroxins , Rotarod Performance Test , Sequence Deletion , Tissue Culture Techniques
7.
Autism Res ; 9(3): 350-75, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26559786

ABSTRACT

Shank3 is a multi-domain, synaptic scaffolding protein that organizes proteins in the postsynaptic density of excitatory synapses. Clinical studies suggest that ∼ 0.5% of autism spectrum disorder (ASD) cases may involve SHANK3 mutation/deletion. Patients with SHANK3 mutations exhibit deficits in cognition along with delayed/impaired speech/language and repetitive and obsessive/compulsive-like (OCD-like) behaviors. To examine how mutation/deletion of SHANK3 might alter brain function leading to ASD, we have independently created mice with deletion of Shank3 exons 4-9, a region implicated in ASD patients. We find that homozygous deletion of exons 4-9 (Shank3(e4-9) KO) results in loss of the two highest molecular weight isoforms of Shank3 and a significant reduction in other isoforms. Behaviorally, both Shank3(e4-9) heterozygous (HET) and Shank3(e4-9) KO mice display increased repetitive grooming, deficits in novel and spatial object recognition learning and memory, and abnormal ultrasonic vocalizations. Shank3(e4-9) KO mice also display abnormal social interaction when paired with one another. Analysis of synaptosome fractions from striata of Shank3(e4-9) KO mice reveals decreased Homer1b/c, GluA2, and GluA3 expression. Both Shank3(e4-9) HET and KO demonstrated a significant reduction in NMDA/AMPA ratio at excitatory synapses onto striatal medium spiny neurons. Furthermore, Shank3(e4-9) KO mice displayed reduced hippocampal LTP despite normal baseline synaptic transmission. Collectively these behavioral, biochemical and physiological changes suggest Shank3 isoforms have region-specific roles in regulation of AMPAR subunit localization and NMDAR function in the Shank3(e4-9) mutant mouse model of autism.


Subject(s)
Autistic Disorder/genetics , Behavior, Animal/physiology , Corpus Striatum/physiopathology , Exons/genetics , Nerve Tissue Proteins/genetics , Synaptic Transmission/physiology , Animals , Disease Models, Animal , Female , Male , Mice , Microfilament Proteins , Sequence Deletion , Synapses/physiology
8.
Neurosci Lett ; 591: 86-92, 2015 Mar 30.
Article in English | MEDLINE | ID: mdl-25668489

ABSTRACT

Down syndrome (DS) is the most common genetic cause of intellectual disability and developmental delay. In addition to cognitive dysfunction, DS patients are marked by diminished neurogenesis, a neuropathological feature also found in the Ts65Dn mouse model of DS. Interestingly, manipulations that enhance neurogenesis - like environmental enrichment or pharmacological agents - improve cognition in Ts65Dn mice. P7C3 is a proneurogenic compound that enhances hippocampal neurogenesis, cell survival, and promotes cognition in aged animals. However, this compound has not been tested in the Ts65Dn mouse model of DS. We hypothesized that P7C3 treatment would reverse or ameliorate the neurogenic deficits in Ts65Dn mice. To test this, adult Ts65Dn and age-matched wild-type (WT) mice were administered vehicle or P7C3 twice daily for 3 months. After 3 months, brains were examined for indices of neurogenesis, including quantification of Ki67, DCX, activated caspase-3 (AC3), and surviving BrdU-immunoreactive(+) cells in the granule cell layer (GCL) of the hippocampal dentate gyrus. P7C3 had no effect on total Ki67+, DCX+, AC3+, or surviving BrdU+ cells in WT mice relative to vehicle. GCL volume was also not changed. In keeping with our hypothesis, however, P7C3-treated Ts65Dn mice had a significant increase in total Ki67+, DCX+, and surviving BrdU+ cells relative to vehicle. P7C3 treatment also decreased AC3+ cell number but had no effect on total GCL volume in Ts65Dn mice. Our findings show 3 months of P7C3 is sufficient to restore the neurogenic deficits observed in the Ts65Dn mouse model of DS.


Subject(s)
Carbazoles/pharmacology , Hippocampus/drug effects , Neuroprotective Agents/pharmacology , Animals , Carbazoles/therapeutic use , Doublecortin Protein , Down Syndrome/drug therapy , Down Syndrome/pathology , Female , Hippocampus/pathology , Male , Mice , Neurogenesis , Neuroprotective Agents/therapeutic use
9.
Autism Res ; 7(2): 264-72, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24619977

ABSTRACT

Multiple candidate genes have been identified for autism spectrum disorders. While some of these genes reach genome-wide significance, others, such as the R451C point mutation in the synaptic cell adhesion molecule neuroligin-3, appear to be rare. Interestingly, two brothers with the same R451C point mutation in neuroligin-3 present clinically on seemingly disparate sides of the autism spectrum. These clinical findings suggest genetic background may play a role in modifying the penetrance of a particular autism-associated mutation. Animal models may contribute additional support for such mutations as functionally relevant and can provide mechanistic insights. Previously, in collaboration with the Südhof laboratory, we reported that mice with an R451C substitution in neuroligin-3 displayed social deficits and enhanced spatial learning. While some of these behavioral abnormalities have since been replicated independently in the Südhof laboratory, observations from the Crawley laboratory failed to replicate these findings in a similar neuroligin-3 mutant mouse model and suggested that genetic background may contribute to variation in observations across laboratories. Therefore, we sought to replicate our findings in the neuroligin-3 R451C point mutant knock-in mouse model (NL3R451C) in a different genetic background. We backcrossed our NL3R451C mouse line onto a 129S2/SvPasCrl genetic background and repeated a subset of our previous behavioral testing. NL3R451C mice on a 129S2/SvPasCrl displayed social deficits, enhanced spatial learning, and increased locomotor activity. These data extend our previous findings that NL3R451C mice exhibit autism-relevant behavioral abnormalities and further suggest that different genetic backgrounds can modify this behavioral phenotype through epistatic genetic interactions.


Subject(s)
Cell Adhesion Molecules, Neuronal/genetics , Child Development Disorders, Pervasive/genetics , DNA Mutational Analysis , Membrane Proteins/genetics , Nerve Tissue Proteins/genetics , Point Mutation/genetics , Social Behavior , Spatial Learning , Animals , Female , Genetic Association Studies , Male , Mice , Mice, Knockout , Models, Genetic
10.
J Comp Neurol ; 522(5): 1171-90, 2014 Apr 01.
Article in English | MEDLINE | ID: mdl-24264880

ABSTRACT

Mutations in phosphatase and tensin homolog deleted on chromosome 10 (PTEN) are implicated in neuropsychiatric disorders including autism. Previous studies report that PTEN knockdown in neurons in vivo leads to increased spine density and synaptic activity. To better characterize synaptic changes in neurons lacking PTEN, we examined the effects of shRNA knockdown of PTEN in basolateral amygdala neurons on synaptic spine density and morphology by using fluorescent dye confocal imaging. Contrary to previous studies in the dentate gyrus, we find that knockdown of PTEN in basolateral amygdala leads to a significant decrease in total spine density in distal dendrites. Curiously, this decreased spine density is associated with increased miniature excitatory postsynaptic current frequency and amplitude, suggesting an increase in number and function of mature spines. These seemingly contradictory findings were reconciled by spine morphology analysis demonstrating increased mushroom spine density and size with correspondingly decreased thin protrusion density at more distal segments. The same analysis of PTEN conditional deletion in the dentate gyrus demonstrated that loss of PTEN does not significantly alter total density of dendritic protrusions in the dentate gyrus, but does decrease thin protrusion density and increases density of more mature mushroom spines. These findings suggest that, contrary to previous reports, PTEN knockdown may not induce de novo spinogenesis, but instead may increase synaptic activity by inducing morphological and functional maturation of spines. Furthermore, behavioral analysis of basolateral amygdala PTEN knockdown suggests that these changes limited only to the basolateral amygdala complex may not be sufficient to induce increased anxiety-related behaviors.


Subject(s)
Amygdala/cytology , Dendritic Spines/physiology , Hippocampus/cytology , Mutation/genetics , Neurons/ultrastructure , PTEN Phosphohydrolase/genetics , Animals , Anxiety/genetics , Behavior, Animal/physiology , Conditioning, Classical/physiology , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , In Vitro Techniques , Locomotion/genetics , Mice , Mice, Transgenic , Miniature Postsynaptic Potentials/genetics , Oncogene Protein v-akt/metabolism , Phosphopyruvate Hydratase/metabolism , Reflex, Startle/genetics , Transduction, Genetic
11.
J Neurosci ; 33(47): 18448-68, 2013 Nov 20.
Article in English | MEDLINE | ID: mdl-24259569

ABSTRACT

The Shank3 gene encodes a scaffolding protein that anchors multiple elements of the postsynaptic density at the synapse. Previous attempts to delete the Shank3 gene have not resulted in a complete loss of the predominant naturally occurring Shank3 isoforms. We have now characterized a homozygous Shank3 mutation in mice that deletes exon 21, including the Homer binding domain. In the homozygous state, deletion of exon 21 results in loss of the major naturally occurring Shank3 protein bands detected by C-terminal and N-terminal antibodies, allowing us to more definitively examine the role of Shank3 in synaptic function and behavior. This loss of Shank3 leads to an increased localization of mGluR5 to both synaptosome and postsynaptic density-enriched fractions in the hippocampus. These mice exhibit a decrease in NMDA/AMPA excitatory postsynaptic current ratio in area CA1 of the hippocampus, reduced long-term potentiation in area CA1, and deficits in hippocampus-dependent spatial learning and memory. In addition, these mice also exhibit motor-coordination deficits, hypersensitivity to heat, novelty avoidance, altered locomotor response to novelty, and minimal social abnormalities. These data suggest that Shank3 isoforms are required for normal synaptic transmission/plasticity in the hippocampus, as well as hippocampus-dependent spatial learning and memory.


Subject(s)
Behavioral Symptoms/genetics , Behavioral Symptoms/pathology , Hippocampus/metabolism , Nerve Tissue Proteins/metabolism , Synaptic Transmission/physiology , Adaptation, Physiological/genetics , Animals , Behavioral Symptoms/metabolism , Exploratory Behavior/physiology , Hippocampus/pathology , Locomotion/genetics , Male , Maze Learning , Mice , Mice, Inbred C57BL , Mice, Transgenic , Microfilament Proteins , Motor Activity/genetics , Mutation/genetics , Nerve Tissue Proteins/genetics , Post-Synaptic Density/genetics , Post-Synaptic Density/metabolism , Protein Isoforms/genetics , Protein Isoforms/metabolism , Psychomotor Performance/physiology , Receptor, Metabotropic Glutamate 5/metabolism , Reflex, Startle/genetics , Synaptic Transmission/genetics , Synaptosomes/metabolism , Synaptosomes/ultrastructure
12.
Mol Neurodegener ; 5: 31, 2010 Aug 23.
Article in English | MEDLINE | ID: mdl-20731874

ABSTRACT

BACKGROUND: BACE1 is a key enzyme in the generation of the Abeta peptide that plays a central role in the pathogenesis of Alzheimer's disease. While BACE1 is an attractive therapeutic target, its normal physiological function remains largely unknown. Examination of BACE1-/- mice can provide insight into this function and also help anticipate consequences of BACE1 inhibition. Here we report a seizure-susceptibility phenotype that we have identified and characterized in BACE1-/- mice. RESULTS: We find that electroencephalographic recordings reveal epileptiform abnormalities in some BACE1-/- mice, occasionally including generalized tonic-clonic and absence seizures. In addition, we find that kainic acid injection induces seizures of greater severity in BACE1-/- mice relative to BACE1+/+ littermates, and causes excitotoxic cell death in a subset of BACE1-/- mice. This hyperexcitability phenotype is variable and appears to be manifest in approximately 30% of BACE1-/- mice. Finally, examination of the expression and localization of the voltage-gated sodium channel alpha-subunit Nav1.2 reveals no correlation with BACE1 genotype or any measure of seizure susceptibility. CONCLUSIONS: Our data indicate that BACE1 deficiency predisposes mice to spontaneous and pharmacologically-induced seizure activity. This finding has implications for the development of safe therapeutic strategies for reducing Abeta levels in Alzheimer's disease. Further, we demonstrate that altered sodium channel expression and axonal localization are insufficient to account for the observed effect, warranting investigation of alternative mechanisms.

13.
Neurobiol Dis ; 33(3): 499-508, 2009 Mar.
Article in English | MEDLINE | ID: mdl-19150498

ABSTRACT

Analysis of naturally occurring mutations that cause seizures in rodents has advanced understanding of the molecular mechanisms underlying epilepsy. Abnormalities of I(h) and h channel expression have been found in many animal models of absence epilepsy. We characterized a novel spontaneous mutant mouse, apathetic (ap/ap), and identified the ap mutation as a 4 base pair insertion within the coding region of Hcn2, the gene encoding the h channel subunit 2 (HCN2). We demonstrated that Hcn2(ap) mRNA is reduced by 90% compared to wild type, and the predicted truncated HCN2(ap) protein is absent from the brain tissue of mice carrying the ap allele. ap/ap mice exhibited ataxia, generalized spike-wave absence seizures, and rare generalized tonic-clonic seizures. ap/+ mice had a normal gait, occasional absence seizures and an increased severity of chemoconvulsant-induced seizures. These findings help elucidate basic mechanisms of absence epilepsy and suggest HCN2 may be a target for therapeutic intervention.


Subject(s)
Epilepsy, Absence/genetics , Ion Channels/genetics , Amino Acid Sequence , Animals , Ataxia/genetics , Base Sequence , Body Size , Brain/metabolism , Brain/physiopathology , COS Cells , Chlorocebus aethiops , Convulsants/administration & dosage , Cyclic Nucleotide-Gated Cation Channels/genetics , Cyclic Nucleotide-Gated Cation Channels/metabolism , Epilepsy, Absence/physiopathology , Female , Frameshift Mutation , Gait , Hyperpolarization-Activated Cyclic Nucleotide-Gated Channels , Ion Channels/chemistry , Male , Mice , Mice, Mutant Strains , Molecular Sequence Data , Phenotype , Potassium Channels/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , Seizures/chemically induced , Seizures/genetics
14.
Neurobiol Dis ; 32(1): 26-36, 2008 Oct.
Article in English | MEDLINE | ID: mdl-18657617

ABSTRACT

Many animal models of temporal lobe epilepsy (TLE) begin with status epilepticus (SE) followed by a latency period. Increased hippocampal pyramidal neuron excitability may contribute to seizures in TLE. I(h), mediated by h channels, regulates intrinsic membrane excitability by modulating synaptic integration and dampening dendritic calcium signaling. In a rat model of TLE, we found bidirectional changes in h channel function in CA1 pyramidal neurons. 1-2 d after SE, before onset of spontaneous seizures, physiological parameters dependent upon h channels were augmented and h channel subunit surface expression was increased. 28-30 d following SE, after onset of spontaneous seizures, h channel function in dendrites was reduced, coupled with diminished h channel subunit surface expression and relocalization of subunits from distal dendrites to soma. These results implicate h channel localization as a molecular mechanism influencing CA1 excitability in TLE.


Subject(s)
Channelopathies/metabolism , Channelopathies/pathology , Cyclic Nucleotide-Gated Cation Channels/metabolism , Epilepsy, Temporal Lobe/metabolism , Epilepsy, Temporal Lobe/pathology , Potassium Channels/metabolism , Protein Subunits/metabolism , Animals , Channelopathies/physiopathology , Cyclic Nucleotide-Gated Cation Channels/physiology , Epilepsy, Temporal Lobe/physiopathology , Hyperpolarization-Activated Cyclic Nucleotide-Gated Channels , Male , Membrane Proteins/metabolism , Membrane Proteins/physiology , Mice , Nerve Tissue Proteins/metabolism , Nerve Tissue Proteins/physiology , Potassium Channels/physiology , Protein Subunits/physiology , Protein Transport/genetics , Rats , Rats, Sprague-Dawley
SELECTION OF CITATIONS
SEARCH DETAIL