Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
Add more filters










Publication year range
1.
Epilepsia ; 2024 May 18.
Article in English | MEDLINE | ID: mdl-38761065

ABSTRACT

OBJECTIVE: The mechanistic target of rapamycin (mTOR) pathway has been implicated in promoting epileptogenesis in animal models of acquired epilepsy, such as posttraumatic epilepsy (PTE) following traumatic brain injury (TBI). However, the specific anatomical regions and neuronal populations mediating mTOR's role in epileptogenesis are not well defined. In this study, we tested the hypothesis that mTOR activation in dentate gyrus granule cells promotes neuronal death, mossy fiber sprouting, and PTE in the controlled cortical impact (CCI) model of TBI. METHODS: An adeno-associated virus (AAV)-Cre viral vector was injected into the hippocampus of Rptorflox/flox (regulatory-associated protein of mTOR) mutant mice to inhibit mTOR activation in dentate gyrus granule cells. Four weeks after AAV-Cre or AAV-vehicle injection, mice underwent CCI injury and were subsequently assessed for mTOR pathway activation by Western blotting, neuronal death, and mossy fiber sprouting by immunopathological analysis, and posttraumatic seizures by video-electroencephalographic monitoring. RESULTS: AAV-Cre injection primarily affected the dentate gyrus and inhibited hippocampal mTOR activation following CCI injury. AAV-Cre-injected mice had reduced neuronal death in dentate gyrus detected by Fluoro-Jade B staining and decreased mossy fiber sprouting by ZnT3 immunostaining. Finally, AAV-Cre-injected mice exhibited a decrease in incidence of PTE. SIGNIFICANCE: mTOR pathway activation in dentate gyrus granule cells may at least partly mediate pathological abnormalities and epileptogenesis in models of TBI and PTE. Targeted modulation of mTOR activity in this hippocampal network may represent a focused therapeutic approach for antiepileptogenesis and prevention of PTE.

2.
bioRxiv ; 2024 Mar 30.
Article in English | MEDLINE | ID: mdl-38585903

ABSTRACT

GABAergic interneuron deficits have been implicated in the epileptogenesis of multiple neurological diseases. While epileptic seizures are a key clinical hallmark of CLN2 disease, a childhood-onset neurodegenerative lysosomal storage disorder caused by a deficiency of tripeptidyl peptidase 1 (TPP1), the etiology of these seizures remains elusive. Given that Cln2 R207X/R207X mice display fatal spontaneous seizures and an early loss of several cortical interneuron populations, we hypothesized that those two events might be causally related. To address this hypothesis, we first generated an inducible transgenic mouse expressing lysosomal membrane-tethered TPP1 (TPP1LAMP1) on the Cln2 R207X/R207X genetic background to study the cell-autonomous effects of cell-type-specific TPP1 deficiency. We crossed the TPP1LAMP1 mice with Vgat-Cre mice to introduce interneuron-specific TPP1 deficiency. Vgat-Cre ; TPP1LAMP1 mice displayed storage material accumulation in several interneuron populations both in cortex and striatum, and increased susceptibility to die after PTZ-induced seizures. Secondly, to test the role of GABAergic interneuron activity in seizure progression, we selectively activated these cells in Cln2 R207X/R207X mice using Designer Receptor Exclusively Activated by Designer Drugs (DREADDs) in in Vgat-Cre : Cln2 R207X/R207X mice. EEG monitoring revealed that DREADD-mediated activation of interneurons via chronic deschloroclozapine administration accelerated the onset of spontaneous seizures and seizure-associated death in Vgat-Cre : Cln2 R207X/R207X mice, suggesting that modulating interneuron activity can exert influence over epileptiform abnormalities in CLN2 disease. Taken together, these results provide new mechanistic insights into the underlying etiology of seizures and premature death that characterize CLN2 disease.

3.
J Neurophysiol ; 131(3): 529-540, 2024 Mar 01.
Article in English | MEDLINE | ID: mdl-38323322

ABSTRACT

Cortical electroencephalograms (EEGs) may help understanding of neuropsychiatric illness and new treatment mechanisms. The aperiodic component (1/f) of EEG power spectra is often treated as noise, but recent studies suggest that changes to the aperiodic exponent of power spectra may reflect changes in excitation/inhibition balance, a concept linked to antidepressant effects, epilepsy, autism, and other clinical conditions. One confound of previous studies is behavioral state, because factors associated with behavioral state other than excitation/inhibition ratio may alter EEG parameters. Thus, to test the robustness of the aperiodic exponent as a predictor of excitation/inhibition ratio, we analyzed video-EEG during active exploration in mice of both sexes during various pharmacological manipulations with the fitting oscillations and one over f (FOOOF) algorithm. We found that GABAA receptor (GABAAR)-positive allosteric modulators increased the aperiodic exponent, consistent with the hypothesis that an increased exponent signals enhanced cortical inhibition, but other drugs (ketamine and GABAAR antagonists at subconvulsive doses) did not follow the prediction. To tilt excitation/inhibition ratio more selectively toward excitation, we suppressed the activity of parvalbumin-positive interneurons with Designer Receptors Exclusively Activated by Designer Drugs (DREADDs). Contrary to our expectations, circuit disinhibition with the DREADD increased the aperiodic exponent. We conclude that the aperiodic exponent of EEG power spectra does not yield a universally reliable marker of cortical excitation/inhibition ratio.NEW & NOTEWORTHY Neuropsychiatric illness may be associated with altered excitation/inhibition balance. A single electroencephalogram (EEG) parameter, the aperiodic exponent of power spectra, may predict the ratio between excitation and inhibition. Here, we use cortical EEGs in mice to evaluate this hypothesis, using pharmacological manipulations of known mechanism. We show that the aperiodic exponent of EEG power spectra is not a reliable marker of excitation/inhibition ratio. Thus, alternative markers of this ratio must be sought.


Subject(s)
Electroencephalography , Ketamine , Male , Female , Mice , Animals , Receptors, GABA-A , Ketamine/pharmacology , gamma-Aminobutyric Acid
4.
Epilepsia ; 65(2): 483-496, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38049961

ABSTRACT

OBJECTIVE: Tuberous sclerosis complex (TSC) is a genetic disorder, characterized by tumor formation in the brain and other organs, and severe neurological symptoms, such as epilepsy. Abnormal vascular endothelial growth factor (VEGF) expression may promote angiogenesis in kidney and lung tumors in TSC and has been identified in brain specimens from TSC patients, but the role of VEGF and vascular abnormalities in neurological manifestations of TSC is poorly defined. In this study, we investigated abnormalities in brain VEGF expression, cerebral blood vessel anatomy, and blood-brain barrier (BBB) structure and function in a mouse model of TSC. METHODS: Tsc1GFAP CKO mice were used to investigate VEGF expression and vascular abnormalities in the brain by Western blotting and immunohistochemical analysis of vascular and BBB markers. In vivo two-photon imaging was used to assess BBB permeability to normally impenetrable fluorescently labeled compounds. The effect of mechanistic target of rapamycin (mTOR) pathway inhibitors, VEGF receptor antagonists (apatinib), or BBB stabilizers (RepSox) was assessed in some of these assays, as well as on seizures by video-electroencephalography. RESULTS: VEGF expression was elevated in cortex of Tsc1GFAP CKO mice, which was reversed by the mTOR inhibitor rapamycin. Tsc1GFAP CKO mice exhibited increased cerebral angiogenesis and vascular complexity in cortex and hippocampus, which were reversed by the VEGF receptor antagonist apatinib. BBB permeability was abnormally increased and BBB-related tight junction proteins occludin and claudin-5 were decreased in Tsc1GFAP CKO mice, also in an apatinib- and RepSox-dependent manner. The BBB stabilizer (RepSox), but not the VEGF receptor antagonist (apatinib), decreased seizures and improved survival in Tsc1GFAP CKO mice. SIGNIFICANCE: Increased brain VEGF expression is dependent on mTOR pathway activation and promotes cerebral vascular abnormalities and increased BBB permeability in a mouse model of TSC. BBB modulation may affect epileptogenesis and represent a rational treatment for epilepsy in TSC.


Subject(s)
Epilepsy , Tuberous Sclerosis , Humans , Mice , Animals , Blood-Brain Barrier , Vascular Endothelial Growth Factor A/metabolism , Tuberous Sclerosis/complications , Tuberous Sclerosis/genetics , Tumor Suppressor Proteins/genetics , Tuberous Sclerosis Complex 1 Protein/genetics , Tuberous Sclerosis Complex 1 Protein/metabolism , Epilepsy/genetics , Epilepsy/metabolism , Seizures , TOR Serine-Threonine Kinases/genetics , Sirolimus , Receptors, Vascular Endothelial Growth Factor/metabolism
5.
bioRxiv ; 2023 Sep 22.
Article in English | MEDLINE | ID: mdl-37790570

ABSTRACT

Cortical electroencephalograms (EEG) may help understanding of neuropsychiatric illness and new treatment mechanisms. The aperiodic component (1/ f ) of EEG power spectra is often treated as noise, but recent studies suggest that changes to the aperiodic exponent of power spectra may reflect changes in excitation/inhibition (E/I) balance, a concept linked to antidepressant effects, epilepsy, autism, and other clinical conditions. One confound of previous studies is behavioral state, because factors associated with behavioral state other than E/I ratio may alter EEG parameters. Thus, to test the robustness of the aperiodic exponent as a predictor of E/I ratio, we analyzed active exploration in mice using video EEG following various pharmacological manipulations with the Fitting Oscillations & One Over F (FOOOF) algorithm. We found that GABA A receptor (GABA A R) positive allosteric modulators increased the aperiodic exponent, consistent with the hypothesis that an increased exponent signals enhanced cortical inhibition, but other drugs (ketamine and GABA A R antagonists at sub-convulsive doses) did not follow the prediction. To tilt E/I ratio more selectively toward excitation, we suppressed the activity of parvalbumin (PV) interneurons with Designer Receptors Exclusively Activated by Designer Drugs (DREADDs). Contrary to our expectations and studies demonstrating increased cortical activity following PV suppression, circuit disinhibition with the DREADD increased the aperiodic exponent. We conclude that the aperiodic exponent of EEG power spectra does not yield a universally reliable marker of E/I ratio. Alternatively, the concept of E/I state may be sufficiently oversimplified that it cannot be mapped readily onto an EEG parameter. Significance StateBment: Neuropsychiatric illness is widely prevalent and debilitating. Causes are not well understood, but some hypotheses point toward altered excitation/inhibition (E/I) balance. Here, we use cortical electroencephalograms (EEG) in mice, given applicability of cortical EEG across species, and evaluate the impact of validated drugs, including anxiolytics (pentobarbital and diazepam), along with novel rapid-acting antidepressants (ketamine and allopregnanolone). We focus on analyzing the aperiodic component of EEG power spectra, which may be associated with changes in E/I ratio. We show that aperiodic exponent of EEG power spectra is not a reliable marker of E/I ratio. Moreover, the concept of E/I ratio may be too broad and complex to be defined by an EEG parameter.

6.
J Clin Invest ; 133(12)2023 06 15.
Article in English | MEDLINE | ID: mdl-37104037

ABSTRACT

Although a disease-modifying therapy for classic late infantile neuronal ceroid lipofuscinosis (CLN2 disease) exists, poor understanding of cellular pathophysiology has hampered the development of more effective and persistent therapies. Here, we investigated the nature and progression of neurological and underlying neuropathological changes in Cln2R207X mice, which carry one of the most common pathogenic mutations in human patients but are yet to be fully characterized. Long-term electroencephalography recordings revealed progressive epileptiform abnormalities, including spontaneous seizures, providing a robust, quantifiable, and clinically relevant phenotype. These seizures were accompanied by the loss of multiple cortical neuron populations, including those stained for interneuron markers. Further histological analysis revealed early localized microglial activation months before neuron loss started in the thalamocortical system and spinal cord, which was accompanied by astrogliosis. This pathology was more pronounced and occurred in the cortex before the thalamus or spinal cord and differed markedly from the staging seen in mouse models of other forms of neuronal ceroid lipofuscinosis. Neonatal administration of adeno-associated virus serotype 9-mediated gene therapy ameliorated the seizure and gait phenotypes and prolonged the life span of Cln2R207X mice, attenuating most pathological changes. Our findings highlight the importance of clinically relevant outcome measures for judging preclinical efficacy of therapeutic interventions for CLN2 disease.


Subject(s)
Neurons , Seizures , Animals , Humans , Mice , Neurons/pathology , Seizures/genetics , Seizures/therapy , Seizures/pathology , Gliosis/pathology , Interneurons/pathology , Thalamus/pathology , Disease Models, Animal
7.
Neuropsychopharmacology ; 48(2): 371-379, 2023 01.
Article in English | MEDLINE | ID: mdl-36168047

ABSTRACT

Neurosteroids that positively modulate GABAA receptors are among a growing list of rapidly acting antidepressants, including ketamine and psychedelics. To develop increasingly specific treatments with fewer side effects, we explored the possibility of EEG signatures in mice, which could serve as a cross-species screening tool. There are few studies of the impact of non-sedative doses of rapid antidepressants on EEG in either rodents or humans. Here we hypothesize that EEG features may separate a rapid antidepressant neurosteroid, allopregnanolone, from other GABAA positive modulators, pentobarbital and diazepam. Further, we compared the actions GABA modulators with those of ketamine, an NMDA antagonist and prototype rapid antidepressant. We examined EEG spectra during active exploration at two cortical locations and examined cross-regional and cross-frequency interactions. We found that at comparable doses, the effects of allopregnanolone, despite purported selectivity for certain GABAAR subtypes, was indistinguishable from pentobarbital during active waking exploration. The actions of diazepam had recognizable common features with allopregnanolone and pentobarbital but was also distinct, consistent with subunit selectivity of benzodiazepines. Finally, ketamine exhibited no distinguishing overlap with allopregnanolone in the parameters examined. Our results suggest that rapid antidepressants with different molecular substrates may remain separated at the level of large-scale ensemble activity, but the studies leave open the possibility of commonalities in more discrete circuits and/or in the context of a dysfunctional brain.


Subject(s)
Ketamine , Neurosteroids , Humans , Mice , Animals , Pregnanolone/pharmacology , Ketamine/pharmacology , Pentobarbital/pharmacology , Receptors, GABA-A/physiology , Diazepam/pharmacology , Antidepressive Agents/pharmacology , gamma-Aminobutyric Acid , Electroencephalography
8.
Sci Rep ; 12(1): 11286, 2022 07 04.
Article in English | MEDLINE | ID: mdl-35789177

ABSTRACT

Cannabidiol (CBD) has gained attention as a therapeutic agent and is purported to have immunomodulatory, neuroprotective, and anti-seizure effects. Here, we determined the effects of chronic CBD administration in a mouse model of CLN1 disease (Cln1-/-) that simultaneously exhibits neuroinflammation, neurodegeneration, and spontaneous seizures. Proteomic analysis showed that putative CBD receptors are expressed at similar levels in the brains of Cln1-/- mice compared to normal animals. Cln1-/- mice received an oral dose (100 mg/kg/day) of CBD for six months and were evaluated for changes in pathological markers of disease and seizures. Chronic cannabidiol administration was well-tolerated, high levels of CBD were detected in the brain, and markers of astrocytosis and microgliosis were reduced. However, CBD had no apparent effect on seizure frequency or neuron survival. These data are consistent with CBD having immunomodulatory effects. It is possible that a higher dose of CBD could also reduce neurodegeneration and seizure frequency.


Subject(s)
Cannabidiol , Graft vs Host Disease , Animals , Cannabidiol/pharmacology , Cannabidiol/therapeutic use , Disease Models, Animal , Graft vs Host Disease/drug therapy , Mice , Neuroinflammatory Diseases , Neuronal Ceroid-Lipofuscinoses , Proteomics
9.
Brain Pathol ; 31(5): e12949, 2021 09.
Article in English | MEDLINE | ID: mdl-33786950

ABSTRACT

Tuberous sclerosis complex (TSC) is a congenital disorder characterized by cortical malformations and concomitant epilepsy caused by loss-of-function mutations in the mTOR suppressors TSC1 or TSC2. While the underlying molecular changes caused by mTOR activation in TSC have previously been investigated, the drivers of these transcriptional change have not been fully elucidated. A better understanding of the perturbed transcriptional regulation could lead to the identification of novel pathways for therapeutic intervention not only in TSC, but other genetic epilepsies in which mTOR activation plays a key role, such as focal cortical dysplasia 2b (FCD). Here, we analyzed RNA sequencing data from cortical tubers and a tsc2-/- zebrafish. We identified differential expression of the transcription factors (TFs) SPI1/PU.1, IRF8, GBX2, and IKZF1 of which SPI1/PU.1 and IRF8 targets were enriched among the differentially expressed genes. Furthermore, for SPI1/PU.1 these findings were conserved in TSC zebrafish model. Next, we confirmed overexpression of SPI1/PU.1 on the RNA and protein level in a separate cohort of surgically resected TSC tubers and FCD tissue, in fetal TSC tissue, and a Tsc1GFAP-/- mouse model of TSC. Subsequently, we validated the expression of SPI1/PU.1 in dysmorphic cells with mTOR activation in TSC tubers. In fetal TSC, we detected SPI1/PU.1 expression prenatally and elevated RNA Spi1 expression in Tsc1GFAP-/- mice before the development of seizures. Finally, in vitro, we identified that in astrocytes and neurons SPI1 transcription was driven by H2 O2 -induced oxidative stress, independent of mTOR. We identified SPI1/PU.1 as a novel TF involved in the pro-inflammatory gene expression of malformed cells in TSC and FCD 2b. This transcriptional program is activated in response to oxidative stress and already present prenatally. Importantly, SPI1/PU.1 protein appears to be strictly limited to malformed cells, as we did not find SPI1/PU.1 protein expression in mice nor in our in vitro models.


Subject(s)
Oxidative Stress/physiology , Proto-Oncogene Proteins/metabolism , Trans-Activators/metabolism , Tuberous Sclerosis Complex 2 Protein/metabolism , Tuberous Sclerosis/pathology , Animals , Astrocytes/metabolism , Astrocytes/pathology , Brain/metabolism , Brain/pathology , Humans , Malformations of Cortical Development/metabolism , Malformations of Cortical Development/pathology , Mice, Transgenic , Neurons/pathology , Tuberous Sclerosis/genetics , Tuberous Sclerosis/metabolism , Tuberous Sclerosis Complex 2 Protein/genetics , Up-Regulation
10.
Nat Commun ; 11(1): 6164, 2020 12 02.
Article in English | MEDLINE | ID: mdl-33268780

ABSTRACT

Familial hemiplegic migraine is an episodic neurological disorder characterized by transient sensory and motor symptoms and signs. Mutations of the ion pump α2-Na/K ATPase cause familial hemiplegic migraine, but the mechanisms by which α2-Na/K ATPase mutations lead to the migraine phenotype remain incompletely understood. Here, we show that mice in which α2-Na/K ATPase is conditionally deleted in astrocytes display episodic paralysis. Functional neuroimaging reveals that conditional α2-Na/K ATPase knockout triggers spontaneous cortical spreading depression events that are associated with EEG low voltage activity events, which correlate with transient motor impairment in these mice. Transcriptomic and metabolomic analyses show that α2-Na/K ATPase loss alters metabolic gene expression with consequent serine and glycine elevation in the brain. A serine- and glycine-free diet rescues the transient motor impairment in conditional α2-Na/K ATPase knockout mice. Together, our findings define a metabolic mechanism regulated by astrocytic α2-Na/K ATPase that triggers episodic motor paralysis in mice.


Subject(s)
Astrocytes/metabolism , Ataxia/genetics , Metabolome/genetics , Migraine with Aura/genetics , Sodium-Potassium-Exchanging ATPase/genetics , Transcriptome , Animals , Astrocytes/pathology , Ataxia/metabolism , Ataxia/pathology , Brain/metabolism , Brain/pathology , Disease Models, Animal , Electroencephalography , Female , Functional Neuroimaging , Glycine/metabolism , Male , Mice , Mice, Knockout , Migraine with Aura/metabolism , Migraine with Aura/pathology , Rotarod Performance Test , Serine/metabolism , Sodium-Potassium-Exchanging ATPase/deficiency
11.
Epilepsia ; 59(9): 1796-1806, 2018 09.
Article in English | MEDLINE | ID: mdl-30079598

ABSTRACT

OBJECTIVE: Microglial abnormalities have been reported in pathologic specimens from patients with tuberous sclerosis complex (TSC), a genetic disorder characterized by epilepsy, intellectual disability, and autism. However, the pathogenic role of microglia in epilepsy in TSC is poorly understood, particularly whether microglia defects may be a primary contributor to epileptogenesis or are secondary to seizures or simply epiphenomena. In this study, we tested the hypothesis that Tsc1 gene inactivation in microglia is sufficient to cause epilepsy in mouse models of TSC. METHODS: Using a chemokine receptor, Cx3cr1, to target microglia, conventional Tsc1Cx3cr1-Cre CKO (conditional knockout) mice and postnatal-inducible Tsc1Cx3cr1-CreER CKO mice were generated and assessed for molecular and histopathologic evidence of microglial abnormalities, mechanistic target of rapamycin 1 (mTORC1) pathway activation, and epilepsy. RESULTS: Tsc1Cx3cr1-Cre CKO mice exhibited a high efficiency of microglia Tsc1 inactivation, mTORC1 activation, increased microglial size and number, and robust epilepsy, which were rapamycin-dependent. However, Cre reporter studies demonstrated that constitutive Cx3cr1 expression affected not only microglia, but also a large percentage of cortical neurons, confounding the role of microglia in epileptogenesis in Tsc1 Cx3cr1-Cre CKO mice. In contrast, postnatal inactivation of Tsc1 utilizing a tamoxifen-inducible Cx3cr1-CreER resulted in a more-selective microglia Tsc1 inactivation with high efficiency, mTORC1 activation, and increased microglial size and number, but no documented epilepsy. SIGNIFICANCE: Microglia abnormalities may contribute to epileptogenesis in the context of neuronal involvement in TSC mouse models, but selective Tsc1 gene inactivation in microglia alone may not be sufficient to cause epilepsy, suggesting that microglia have more supportive roles in the pathogenesis of seizures in TSC.


Subject(s)
Mechanistic Target of Rapamycin Complex 1/metabolism , Microglia/pathology , Tuberous Sclerosis/pathology , Tuberous Sclerosis/physiopathology , Animals , Animals, Newborn , Brain/pathology , CX3C Chemokine Receptor 1/genetics , CX3C Chemokine Receptor 1/metabolism , Calcium-Binding Proteins/metabolism , Disease Models, Animal , Electroencephalography , Estrogen Antagonists/pharmacology , Gene Expression Regulation/genetics , Glial Fibrillary Acidic Protein/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , Microfilament Proteins/metabolism , Microglia/drug effects , Microglia/metabolism , Phosphopyruvate Hydratase/metabolism , Sirolimus/pharmacology , Statistics, Nonparametric , Tamoxifen/pharmacology , Tuberous Sclerosis/drug therapy , Tuberous Sclerosis/genetics , Tuberous Sclerosis Complex 1 Protein/genetics , Tuberous Sclerosis Complex 1 Protein/metabolism , Video Recording
12.
Neurobiol Dis ; 80: 70-9, 2015 Aug.
Article in English | MEDLINE | ID: mdl-26003087

ABSTRACT

Epilepsy and other neurological deficits are common, disabling manifestations of the genetic disorder, tuberous sclerosis complex (TSC). Brain inflammation has been implicated in contributing to epileptogenesis in acquired epilepsy due to brain injury, but the potential role of inflammatory mechanisms in genetic epilepsies is relatively unexplored. In this study, we investigated activation of inflammatory mediators and tested the effects of anti-inflammatory treatment on epilepsy in the Tsc1-GFAP conditional knock-out mouse model of TSC (Tsc1(GFAP)CKO mice). Real-time quantitative RT-PCR, immunohistochemistry, and Western blotting demonstrated increased expression of specific cytokines and chemokines, particularly IL-1ß and CXCL10, in the neocortex and hippocampus of Tsc1(GFAP)CKO mice, which was reversed by treatment with a mammalian target of rapamycin complex 1 (mTORC1) inhibitor. Double-labeling immunohistochemical studies indicated that the increased IL-1ß was localized primarily to astrocytes. Importantly, the increase in inflammatory markers was also observed in astrocyte culture in vitro and at 2 weeks of age in Tsc1(GFAP)CKO mice before the onset of epilepsy in vivo, indicating that the inflammatory changes were not secondary to seizures. Epicatechin-3-gallate, an inhibitor of IL-1ß and CXCL10, at least partially reversed the elevated cytokine and chemokine levels, reduced seizure frequency, and prolonged survival of Tsc1(GFAP)CKO mice. These findings suggest that mTOR-mediated inflammatory mechanisms may be involved in epileptogenesis in the genetic epilepsy, TSC.


Subject(s)
Encephalitis/metabolism , Seizures/etiology , Tuberous Sclerosis/complications , Tuberous Sclerosis/metabolism , Animals , Anti-Inflammatory Agents/administration & dosage , Catechin/administration & dosage , Catechin/analogs & derivatives , Encephalitis/complications , Hippocampus/drug effects , Hippocampus/metabolism , Inflammation Mediators/metabolism , Mechanistic Target of Rapamycin Complex 1 , Mice , Mice, Knockout , Multiprotein Complexes/antagonists & inhibitors , Neocortex/drug effects , Neocortex/metabolism , Neuroglia/drug effects , Neuroglia/metabolism , Sirolimus/administration & dosage , Survival Analysis , TOR Serine-Threonine Kinases/antagonists & inhibitors , Tuberous Sclerosis/pathology , Tuberous Sclerosis Complex 1 Protein , Tumor Suppressor Proteins/genetics
13.
PLoS One ; 8(2): e57445, 2013.
Article in English | MEDLINE | ID: mdl-23437388

ABSTRACT

Epilepsy is a common neurological disorder and cause of significant morbidity and mortality. Although antiseizure medication is the first-line treatment for epilepsy, currently available medications are ineffective in a significant percentage of patients and have not clearly been demonstrated to have disease-specific effects for epilepsy. While seizures are usually intractable to medication in tuberous sclerosis complex (TSC), a common genetic cause of epilepsy, vigabatrin appears to have unique efficacy for epilepsy in TSC. While vigabatrin increases gamma-aminobutyric acid (GABA) levels, the precise mechanism of action of vigabatrin in TSC is not known. In this study, we investigated the effects of vigabatrin on epilepsy in a knock-out mouse model of TSC and tested the novel hypothesis that vigabatrin inhibits the mammalian target of rapamycin (mTOR) pathway, a key signaling pathway that is dysregulated in TSC. We found that vigabatrin caused a modest increase in brain GABA levels and inhibited seizures in the mouse model of TSC. Furthermore, vigabatrin partially inhibited mTOR pathway activity and glial proliferation in the knock-out mice in vivo, as well as reduced mTOR pathway activation in cultured astrocytes from both knock-out and control mice. This study identifies a potential novel mechanism of action of an antiseizure medication involving the mTOR pathway, which may account for the unique efficacy of this drug for a genetic epilepsy.


Subject(s)
Anticonvulsants/pharmacology , Astrocytes/drug effects , Brain/drug effects , Seizures/drug therapy , TOR Serine-Threonine Kinases/genetics , Tuberous Sclerosis/drug therapy , Vigabatrin/pharmacology , Animals , Astrocytes/metabolism , Astrocytes/pathology , Brain/metabolism , Cell Proliferation/drug effects , Cells, Cultured , Disease Models, Animal , Gene Expression Regulation/drug effects , Humans , Mice , Mice, Knockout , Seizures/genetics , Seizures/metabolism , Seizures/pathology , Signal Transduction/drug effects , TOR Serine-Threonine Kinases/antagonists & inhibitors , TOR Serine-Threonine Kinases/metabolism , Tuberous Sclerosis/genetics , Tuberous Sclerosis/metabolism , Tuberous Sclerosis/pathology , Tuberous Sclerosis Complex 1 Protein , Tumor Suppressor Proteins/deficiency , Tumor Suppressor Proteins/genetics , gamma-Aminobutyric Acid/biosynthesis
14.
Methods Mol Biol ; 821: 373-91, 2012.
Article in English | MEDLINE | ID: mdl-22125079

ABSTRACT

Tuberous Sclerosis Complex (TSC) is a genetic disease involving dysregulation of the mTOR pathway and resulting in disabling neurological manifestations, such as epilepsy. Animal models may recapitulate epilepsy and other behavioral features of TSC and are useful tools for investigating mechanisms of epileptogenesis and other neurological deficits in TSC. In this chapter, methods for performing video-electroencephalography (video-EEG) to characterize epilepsy and neurological dysfunction in rodent models are reviewed. In particular, technical aspects of surgical implantation of EEG electrodes, video-EEG recording, and analysis and interpretation of EEG data are detailed. These methodological approaches should be helpful in characterizing seizures and background EEG abnormalities not only in animal models of TSC but also in many rodent epilepsy models in general.


Subject(s)
Electroencephalography/methods , Epilepsy/physiopathology , Monitoring, Physiologic/methods , TOR Serine-Threonine Kinases/metabolism , Tuberous Sclerosis/physiopathology , Video Recording/methods , Animals , Disease Models, Animal , Electrodes, Implanted , Humans , Magnetic Resonance Imaging , Mice , Rats , TOR Serine-Threonine Kinases/genetics
15.
Neurobiol Dis ; 45(1): 348-55, 2012 Jan.
Article in English | MEDLINE | ID: mdl-21889979

ABSTRACT

Seizures may directly cause brain injury by disrupting the structure and function of synapses. Previous studies using in vivo time-lapse imaging have demonstrated an acute beading of dendrites and loss of dendritic spines immediately following status epilepticus, but the effects of brief seizures and the long-term evolution of this dendritic injury are unknown. Here, we examined the effects of seizures of varying durations on dendritic structure over several weeks using in vivo multiphoton imaging with kainate-induced seizures in mice. The degree of dendritic injury was directly dependent on the duration of the seizures, with seizures lasting more than 30 min (status epilepticus) resulting in a greater than 75% spine loss. However, even brief seizures (<5 min) induced moderate dendritic beading and spine loss. The dendritic injury from brief seizures usually recovered within 2 weeks, whereas status epilepticus-induced injury only partially reversed. These studies demonstrate that seizures of all durations may trigger at least transient neuronal injury.


Subject(s)
Cerebral Cortex/pathology , Dendrites/pathology , Neurons/pathology , Seizures/pathology , Status Epilepticus/pathology , Animals , Dendritic Spines/pathology , Kainic Acid , Mice , Seizures/chemically induced , Status Epilepticus/chemically induced , Time Factors
16.
Epilepsia ; 52(3): e7-11, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21371020

ABSTRACT

The ketogenic diet (KD) is an effective treatment for epilepsy, but its mechanisms of action are poorly understood. We investigated the hypothesis that the KD inhibits mammalian target of rapamycin (mTOR) pathway signaling. The expression of pS6 and pAkt, markers of mTOR pathway activation, was reduced in hippocampus and liver of rats fed KD. In the kainate model of epilepsy, KD blocked the hippocampal pS6 elevation that occurs after status epilepticus. Because mTOR signaling has been implicated in epileptogenesis, these results suggest that the KD may have anticonvulsant or antiepileptogenic actions via mTOR pathway inhibition.


Subject(s)
Epilepsy/diet therapy , Epilepsy/genetics , TOR Serine-Threonine Kinases/genetics , AMP-Activated Protein Kinase Kinases , Animals , Diet, Ketogenic , Disease Models, Animal , Epilepsy/pathology , Gene Expression/genetics , Hippocampus/pathology , Liver/pathology , Protein Kinases/genetics , Rats , Rats, Sprague-Dawley , Signal Transduction/genetics , Status Epilepticus/genetics
17.
Hum Mol Genet ; 20(3): 445-54, 2011 Feb 01.
Article in English | MEDLINE | ID: mdl-21062901

ABSTRACT

Tuberous Sclerosis Complex (TSC) is an autosomal dominant, multi-system disorder, typically involving severe neurological symptoms, such as epilepsy, cognitive deficits and autism. Two genes, TSC1 and TSC2, encoding the proteins hamartin and tuberin, respectively, have been identified as causing TSC. Although there is a substantial overlap in the clinical phenotype produced by TSC1 and TSC2 mutations, accumulating evidence indicates that TSC2 mutations cause more severe neurological manifestations than TSC1 mutations. In this study, the neurological phenotype of a novel mouse model involving conditional inactivation of the Tsc2 gene in glial-fibrillary acidic protein (GFAP)-positive cells (Tsc2(GFAP1)CKO mice) was characterized and compared with previously generated Tsc1(GFAP1)CKO mice. Similar to Tsc1(GFAP1)CKO mice, Tsc2(GFAP1)CKO mice exhibited epilepsy, premature death, progressive megencephaly, diffuse glial proliferation, dispersion of hippocampal pyramidal cells and decreased astrocyte glutamate transporter expression. However, Tsc2(GFAP1)CKO mice had an earlier onset and higher frequency of seizures, as well as significantly more severe histological abnormalities, compared with Tsc1(GFAP1)CKO mice. The differences between Tsc1(GFAP1)CKO and Tsc2(GFAP1)CKO mice were correlated with higher levels of mammalian target of rapamycin (mTOR) activation in Tsc2(GFAP1)CKO mice and were reversed by the mTOR inhibitor, rapamycin. These findings provide novel evidence in mouse models that Tsc2 mutations intrinsically cause a more severe neurological phenotype than Tsc1 mutations and suggest that the difference in phenotype may be related to the degree to which Tsc1 and Tsc2 inactivation causes abnormal mTOR activation.


Subject(s)
Epilepsy/genetics , Gene Silencing , Tuberous Sclerosis/genetics , Tuberous Sclerosis/metabolism , Tumor Suppressor Proteins/genetics , Animals , Blotting, Western , Disease Models, Animal , Epilepsy/metabolism , Fluorescent Antibody Technique , Mice , Mice, Knockout , Mice, Transgenic , Mutation , Phenotype , TOR Serine-Threonine Kinases/genetics , TOR Serine-Threonine Kinases/metabolism , Tuberous Sclerosis Complex 1 Protein , Tuberous Sclerosis Complex 2 Protein , Tumor Suppressor Proteins/metabolism
18.
Cell Cycle ; 9(12): 2281-5, 2010 Jun 15.
Article in English | MEDLINE | ID: mdl-20603607

ABSTRACT

Identification of cell signaling mechanisms mediating seizure-related neuronal death and epileptogenesis is important for developing more effective therapies for epilepsy. The mammalian target of rapamycin (mTOR) pathway has recently been implicated in regulating neuronal death and epileptogenesis in rodent models of epilepsy. In particular, kainate-induced status epilepticus causes abnormal activation of the mTOR pathway, and the mTOR inhibitor, rapamycin, can decrease the development of neuronal death and chronic seizures in the kainate model. Here, we discuss the significance of these findings and extend them further by identifying upstream signaling pathways through which kainate status epilepticus activates the mTOR pathway and by demonstrating limited situations where rapamycin may paradoxically increase mTOR activation and worsen neuronal death in the kainate model. Thus, the regulation of seizure-induced neuronal death and epileptogenesis by mTOR is complex and may have dual, opposing effects depending on the physiological and pathological context. Overall, these findings have important implications for designing potential neuroprotective and antiepileptogenic therapies that modulate the mTOR pathway.


Subject(s)
Epilepsy/metabolism , TOR Serine-Threonine Kinases/metabolism , Animals , Cell Death/drug effects , Epilepsy/pathology , Mice , Neurons/metabolism , Neurons/pathology , Neuroprotective Agents/pharmacology , Phosphatidylinositol 3-Kinase/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Rats , Receptors, Kainic Acid/metabolism , Signal Transduction/drug effects , Status Epilepticus/metabolism
19.
J Neurosci ; 29(21): 6964-72, 2009 May 27.
Article in English | MEDLINE | ID: mdl-19474323

ABSTRACT

Understanding molecular mechanisms mediating epileptogenesis is critical for developing more effective therapies for epilepsy. We recently found that the mammalian target of rapamycin (mTOR) signaling pathway is involved in epileptogenesis, and mTOR inhibitors prevent epilepsy in a mouse model of tuberous sclerosis complex. Here, we investigated the potential role of mTOR in a rat model of temporal lobe epilepsy initiated by status epilepticus. Acute kainate-induced seizures resulted in biphasic activation of the mTOR pathway, as evident by an increase in phospho-S6 (P-S6) expression. An initial rise in P-S6 expression started approximately 1 h after seizure onset, peaked at 3-6 h, and returned to baseline by 24 h in both hippocampus and neocortex, reflecting widespread stimulation of mTOR signaling by acute seizure activity. After resolution of status epilepticus, a second increase in P-S6 was observed in hippocampus only, which started at 3 d, peaked 5-10 d, and persisted for several weeks after kainate injection, correlating with the development of chronic epileptogenesis within hippocampus. The mTOR inhibitor rapamycin, administered before kainate, blocked both the acute and chronic phases of seizure-induced mTOR activation and decreased kainate-induced neuronal cell death, neurogenesis, mossy fiber sprouting, and the development of spontaneous epilepsy. Late rapamycin treatment, after termination of status epilepticus, blocked the chronic phase of mTOR activation and reduced mossy fiber sprouting and epilepsy but not neurogenesis or neuronal death. These findings indicate that mTOR signaling mediates mechanisms of epileptogenesis in the kainate rat model and that mTOR inhibitors have potential antiepileptogenic effects in this model.


Subject(s)
Epilepsy, Temporal Lobe/metabolism , Protein Kinases/metabolism , Signal Transduction/physiology , Analysis of Variance , Animals , Bromodeoxyuridine/metabolism , Cell Death/physiology , Disease Models, Animal , Electroencephalography/methods , Epilepsy, Temporal Lobe/chemically induced , Epilepsy, Temporal Lobe/pathology , Epilepsy, Temporal Lobe/prevention & control , Fluoresceins , Gene Expression Regulation/drug effects , Immunosuppressive Agents/pharmacology , In Situ Nick-End Labeling/methods , Kainic Acid , Male , Mossy Fibers, Hippocampal/drug effects , Mossy Fibers, Hippocampal/physiology , Organic Chemicals/metabolism , Rats , Rats, Sprague-Dawley , Seizures/drug therapy , Seizures/physiopathology , Signal Transduction/drug effects , Sirolimus/administration & dosage , TOR Serine-Threonine Kinases , Time Factors , Video Recording/methods
20.
Mol Cell Pharmacol ; 1(3): 124-129, 2009 Jan 01.
Article in English | MEDLINE | ID: mdl-20419051

ABSTRACT

While current medications for epilepsy are primarily symptomatic treatments that suppress seizures, one of the main goals of future drug development in epilepsy is the identification of antiepileptogenic or disease-modifying therapies that can completely prevent epilepsy or slow its progression. A rational antiepileptogenic strategy is to target primary cell signaling pathways that initially trigger the downstream mechanisms causing epileptogenesis. Recent work implicates the mammalian target of rapamycin (mTOR) pathway as mediating epileptogenesis in a genetic epilepsy, Tuberous Sclerosis Complex (TSC), and suggests that mTOR inhibitors, such as rapamycin, may have antiepileptogenic properties for epilepsy in TSC. As mTOR regulates multiple cellular functions that may contribute to epileptogenesis in general, including ion channel expression, synaptic plasticity, and programmed cell death, mTOR inhibitors might also represent an effective antiepileptogenic therapy for other, more common types of epilepsy, such as acquired epilepsies due to brain injuries. Here, we describe evidence from a recently-published study that mTOR mediates epileptogenesis in a popular animal model of acquired limbic epilepsy due to brain injury following kainate-induced status epilepticus, and that rapamycin has antiepileptogenic effects in this model. Furthermore, putative pathways and mechanisms upstream and downstream from mTOR involved in epileptogenesis in the kainite model are considered, identifying possible additional therapeutic targets. Finally, the potential translational applications of this and other animal model data for developing antiepileptogenic therapies for people with acquired epilepsy due to brain injury are discussed.

SELECTION OF CITATIONS
SEARCH DETAIL
...