Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Publication year range
1.
Rinsho Shinkeigaku ; 60(1): 27-31, 2020 Jan 30.
Article in Japanese | MEDLINE | ID: mdl-31852871

ABSTRACT

BACKGROUND: Metabolic insult causing re-expression of old stroke (MICROS), one of the stroke mimics, is characterized by reappearance of impairment of past stroke and can be mistaken for a stroke recurrence. The aim of the present study was to identify the clinical characteristics of MICROS in emergency stroke care, and to investigate predictive factors for distinguishing MICROS from stroke recurrences. METHODS: In our Stroke Center, 519 consecutive patients admitted with suspected stroke in June 2016 to December 2017. MICROS was defined as an acute deterioration of neurological deficits of the previous stroke despite no evidence for stroke recurrences. Among the 70 patients with a past history of stroke, 14 were MICROS, 5 were transient ischemic attack, 15 were other stroke mimics, and 36 were stroke recurrences, respectively. We evaluated the clinical characteristics of MICROS and compared MICROS with stroke recurrences. RESULTS: The causes of MICROS were infectious disease (including influenza and pneumonia) in 4, transient somnolence after syncope in 4, hypo/hyperglycemia in 2, medication overdoses in 1, and anxiety in 3. Eight of the 14 MICROS patients were admitted within 4 hours after the symptom onset. In MICROS patients, fever (>37°C) was observed more frequently than those with stroke recurrences though the difference was not statistically significant. CONCLUSION: MICROS might be associated with fever, syncope, or serum glucose abnormality. MICROS patients sometimes visit the hospital emergency room within 4 hours, thus, distinction between MICROS and true stroke recurrences is important.


Subject(s)
Stroke/diagnosis , Stroke/etiology , Diagnosis, Differential , Drug Overdose/complications , Humans , Hyperglycemia , Influenza, Human/complications , Pneumonia/complications , Recurrence , Syncope/complications
2.
Nat Neurosci ; 15(4): 574-80, 2012 Feb 26.
Article in English | MEDLINE | ID: mdl-22366758

ABSTRACT

Transient global ischemia in rats induces delayed death of hippocampal CA1 neurons. Early events include caspase activation, cleavage of anti-death Bcl-2 family proteins and large mitochondrial channel activity. However, whether these events have a causal role in ischemia-induced neuronal death is unclear. We found that the Bcl-2 and Bcl-x(L) inhibitor ABT-737, which enhances death of tumor cells, protected rats against neuronal death in a clinically relevant model of brain ischemia. Bcl-x(L) is prominently expressed in adult neurons and can be cleaved by caspases to generate a pro-death fragment, ΔN-Bcl-x(L). We found that ABT-737 administered before or after ischemia inhibited ΔN-Bcl-x(L)-induced mitochondrial channel activity and neuronal death. To establish a causal role for ΔN-Bcl-x(L), we generated knock-in mice expressing a caspase-resistant form of Bcl-x(L). The knock-in mice exhibited markedly reduced mitochondrial channel activity and reduced vulnerability to ischemia-induced neuronal death. These findings suggest that truncated Bcl-x(L) could be a potentially important therapeutic target in ischemic brain injury.


Subject(s)
Brain Ischemia/metabolism , Brain Ischemia/pathology , Neurons/metabolism , Neurons/pathology , bcl-X Protein/physiology , Animals , Biphenyl Compounds/pharmacology , Biphenyl Compounds/therapeutic use , Brain Ischemia/prevention & control , Cell Death/drug effects , Cell Death/genetics , Cells, Cultured , Female , Gene Knock-In Techniques , Male , Mice , Mice, Knockout , Neurons/drug effects , Nitrophenols/pharmacology , Nitrophenols/therapeutic use , Organ Culture Techniques , Piperazines/pharmacology , Piperazines/therapeutic use , Rats , Rats, Sprague-Dawley , Sulfonamides/pharmacology , Sulfonamides/therapeutic use , bcl-X Protein/biosynthesis , bcl-X Protein/genetics
3.
Proc Natl Acad Sci U S A ; 109(16): E962-71, 2012 Apr 17.
Article in English | MEDLINE | ID: mdl-22371606

ABSTRACT

Dysregulation of the transcriptional repressor element-1 silencing transcription factor (REST)/neuron-restrictive silencer factor is important in a broad range of diseases, including cancer, diabetes, and heart disease. The role of REST-dependent epigenetic modifications in neurodegeneration is less clear. Here, we show that neuronal insults trigger activation of REST and CoREST in a clinically relevant model of ischemic stroke and that REST binds a subset of "transcriptionally responsive" genes (gria2, grin1, chrnb2, nefh, nfκb2, trpv1, chrm4, and syt6), of which the AMPA receptor subunit GluA2 is a top hit. Genes with enriched REST exhibited decreased mRNA and protein. We further show that REST assembles with CoREST, mSin3A, histone deacetylases 1 and 2, histone methyl-transferase G9a, and methyl CpG binding protein 2 at the promoters of target genes, where it orchestrates epigenetic remodeling and gene silencing. RNAi-mediated depletion of REST or administration of dominant-negative REST delivered directly into the hippocampus in vivo prevents epigenetic modifications, restores gene expression, and rescues hippocampal neurons. These findings document a causal role for REST-dependent epigenetic remodeling in the neurodegeneration associated with ischemic stroke and identify unique therapeutic targets for the amelioration of hippocampal injury and cognitive deficits.


Subject(s)
Epigenesis, Genetic/genetics , Epigenomics , Neurons/metabolism , Repressor Proteins/genetics , Animals , Blotting, Western , CA1 Region, Hippocampal/metabolism , CA1 Region, Hippocampal/pathology , Cell Death , Cells, Cultured , Co-Repressor Proteins/genetics , Co-Repressor Proteins/metabolism , Gene Expression Regulation , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Humans , Ischemia/complications , Male , Microscopy, Fluorescence , Neurons/pathology , Promoter Regions, Genetic/genetics , Protein Binding , RNA Interference , Rats , Rats, Sprague-Dawley , Receptors, AMPA/genetics , Receptors, AMPA/metabolism , Repressor Proteins/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Stroke/etiology , Stroke/genetics , Stroke/metabolism
4.
Brain Res ; 1321: 1-12, 2010 Mar 19.
Article in English | MEDLINE | ID: mdl-20114038

ABSTRACT

Global ischemia arising during cardiac arrest or cardiac surgery causes highly selective, delayed death of hippocampal CA1 neurons. Exogenous estradiol ameliorates global ischemia-induced neuronal death and cognitive impairment in male and female rodents. However, the molecular mechanisms by which a single acute injection of estradiol administered after the ischemic event intervenes in global ischemia-induced apoptotic cell death are unclear. Here we show that acute estradiol acts via the phosphoinositide 3-kinase (PI3K)/protein kinase B (Akt) signaling cascade to protect CA1 neurons in ovariectomized female rats. We demonstrate that global ischemia promotes early activation of glycogen synthase kinase-3beta (GSK3beta) and forkhead transcription factor of the O class (FOXO)3A, known Akt targets that are related to cell survival, and activation of caspase-3. Estradiol prevents ischemia-induced dephosphorylation and activation of GSK3beta and FOXO3A, and the caspase death cascade. These findings support a model whereby estradiol acts by activation of PI3K/Akt signaling to promote neuronal survival in the face of global ischemia.


Subject(s)
Apoptosis/drug effects , Brain Ischemia/drug therapy , Estradiol/pharmacology , Estrogens/pharmacology , Neurons/drug effects , Signal Transduction/drug effects , Animals , Blotting, Western , Brain Ischemia/pathology , Female , Hippocampus/drug effects , Hippocampus/metabolism , Hippocampus/pathology , Nerve Degeneration/prevention & control , Neurons/metabolism , Neurons/pathology , Ovariectomy , Phosphatidylinositol 3-Kinases/drug effects , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/drug effects , Proto-Oncogene Proteins c-akt/metabolism , Rats
5.
J Neurosci ; 30(2): 694-702, 2010 Jan 13.
Article in English | MEDLINE | ID: mdl-20071534

ABSTRACT

Fragile X syndrome, the most common form of inherited mental retardation and leading genetic cause of autism, is caused by transcriptional silencing of the Fmr1 gene. The fragile X mental retardation protein (FMRP), the gene product of Fmr1, is an RNA binding protein that negatively regulates translation in neurons. The Fmr1 knock-out mouse, a model of fragile X syndrome, exhibits cognitive deficits and exaggerated metabotropic glutamate receptor (mGluR)-dependent long-term depression at CA1 synapses. However, the molecular mechanisms that link loss of function of FMRP to aberrant synaptic plasticity remain unclear. The mammalian target of rapamycin (mTOR) signaling cascade controls initiation of cap-dependent translation and is under control of mGluRs. Here we show that mTOR phosphorylation and activity are elevated in hippocampus of juvenile Fmr1 knock-out mice by four functional readouts: (1) association of mTOR with regulatory associated protein of mTOR; (2) mTOR kinase activity; (3) phosphorylation of mTOR downstream targets S6 kinase and 4E-binding protein; and (4) formation of eukaryotic initiation factor complex 4F, a critical first step in cap-dependent translation. Consistent with this, mGluR long-term depression at CA1 synapses of FMRP-deficient mice is exaggerated and rapamycin insensitive. We further show that the p110 subunit of the upstream kinase phosphatidylinositol 3-kinase (PI3K) and its upstream activator PI3K enhancer PIKE, predicted targets of FMRP, are upregulated in knock-out mice. Elevated mTOR signaling may provide a functional link between overactivation of group I mGluRs and aberrant synaptic plasticity in the fragile X mouse, mechanisms relevant to impaired cognition in fragile X syndrome.


Subject(s)
Fragile X Syndrome/complications , Fragile X Syndrome/metabolism , Signal Transduction/physiology , Sirolimus/metabolism , Adaptor Proteins, Signal Transducing , Animals , CA1 Region, Hippocampal/metabolism , Carrier Proteins/genetics , Carrier Proteins/metabolism , Cell Cycle Proteins , Cognition Disorders/etiology , Disease Models, Animal , Eukaryotic Initiation Factor-4A/genetics , Eukaryotic Initiation Factor-4A/metabolism , Eukaryotic Initiation Factors , Excitatory Postsynaptic Potentials/drug effects , Excitatory Postsynaptic Potentials/genetics , Fragile X Mental Retardation Protein/genetics , Fragile X Syndrome/genetics , Fragile X Syndrome/pathology , Gene Expression Regulation/drug effects , Gene Expression Regulation/genetics , Immunoprecipitation/methods , In Vitro Techniques , Long-Term Synaptic Depression/drug effects , Long-Term Synaptic Depression/genetics , Methoxyhydroxyphenylglycol/analogs & derivatives , Methoxyhydroxyphenylglycol/pharmacology , Mice , Mice, Knockout , Oncogene Protein v-akt/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Phosphoproteins/genetics , Phosphoproteins/metabolism , Phosphorylation/genetics , Receptors, Metabotropic Glutamate/metabolism , Serine/metabolism , Signal Transduction/genetics
6.
Nat Neurosci ; 12(5): 618-26, 2009 May.
Article in English | MEDLINE | ID: mdl-19349976

ABSTRACT

Dysregulation of Akt signaling is important in a broad range of diseases that includes cancer, diabetes and heart disease. The role of Akt signaling in brain disorders is less clear. We found that global ischemia in intact rats triggered expression and activation of the Akt inhibitor CTMP (carboxyl-terminal modulator protein) in vulnerable hippocampal neurons and that CTMP bound and extinguished Akt activity and was essential to ischemia-induced neuronal death. Although ischemia induced a marked phosphorylation and nuclear translocation of Akt, phosphorylated Akt was not active in post-ischemic neurons, as assessed by kinase assays and phosphorylation of the downstream targets GSK-3beta and FOXO3A. RNA interference-mediated depletion of CTMP in a clinically relevant model of stroke restored Akt activity and rescued hippocampal neurons. Our results indicate that CTMP is important in the neurodegeneration that is associated with stroke and identify CTMP as a therapeutic target for the amelioration of hippocampal injury and cognitive deficits.


Subject(s)
Brain Infarction/metabolism , Brain Ischemia/metabolism , Carrier Proteins/metabolism , Nerve Degeneration/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Active Transport, Cell Nucleus/physiology , Animals , Brain Infarction/physiopathology , Brain Ischemia/physiopathology , Carrier Proteins/genetics , Cells, Cultured , Disease Models, Animal , Forkhead Box Protein O3 , Forkhead Transcription Factors/metabolism , Glycogen Synthase Kinase 3/metabolism , Glycogen Synthase Kinase 3 beta , Hippocampus/metabolism , Hippocampus/physiopathology , Male , Nerve Degeneration/physiopathology , Neurons/metabolism , Palmitoyl-CoA Hydrolase , Phosphorylation , Rats , Rats, Sprague-Dawley , Signal Transduction/physiology
7.
Proc Natl Acad Sci U S A ; 105(12): 4892-7, 2008 Mar 25.
Article in English | MEDLINE | ID: mdl-18347331

ABSTRACT

Transient forebrain or global ischemia induces delayed neuronal death in vulnerable CA1 pyramidal cells with many features of apoptosis. A brief period of ischemia, i.e., ischemic preconditioning, affords robust protection of CA1 neurons against a subsequent more prolonged ischemic challenge. Here we show that preconditioning acts via PI3K/Akt signaling to block the ischemia-induced cascade involving mitochondrial translocation of Bad, assembly of Bad with Bcl-x(L), cleavage of Bcl-x(L) to form its prodeath fragment, DeltaN-Bcl-x(L), activation of large-conductance channels in the mitochondrial outer membrane, mitochondrial release of cytochrome c and Smac/DIABLO (second mitochondria-derived activator of caspases/direct IAP-binding protein with low pI), caspase activation, and neuronal death. These findings show how preconditioning acts to prevent the release of cytochrome c and Smac/DIABLO from mitochondria and to preserve the integrity of the mitochondrial membrane. The specific PI3K inhibitor LY294002 administered in vivo 1 h before or immediately after ischemia or up to 120 h later significantly reverses preconditioning-induced protection, indicating a requirement for sustained PI3K signaling in ischemic tolerance. These findings implicate PI3K/Akt signaling in maintenance of the integrity of the mitochondrial outer membrane.


Subject(s)
Hippocampus/metabolism , Ischemic Preconditioning , Large-Conductance Calcium-Activated Potassium Channels/metabolism , Mitochondria/metabolism , Neurons/metabolism , bcl-Associated Death Protein/metabolism , bcl-X Protein/metabolism , Animals , Apoptosis/drug effects , Brain Ischemia/enzymology , Caspase Inhibitors , Chromones/pharmacology , Hippocampus/cytology , Hippocampus/drug effects , Hippocampus/enzymology , Ion Channel Gating/drug effects , Male , Mitochondria/drug effects , Mitochondria/enzymology , Morpholines/pharmacology , Neurons/cytology , Neurons/drug effects , Neurons/enzymology , Phosphoinositide-3 Kinase Inhibitors , Phosphorylation/drug effects , Protein Transport/drug effects , Proto-Oncogene Proteins c-akt/metabolism , Rats , Rats, Sprague-Dawley , Signal Transduction/drug effects
8.
Proc Natl Acad Sci U S A ; 104(10): 4170-5, 2007 Mar 06.
Article in English | MEDLINE | ID: mdl-17360495

ABSTRACT

Transient global ischemia is a neuronal insult that induces delayed, selective death of hippocampal CA1 pyramidal neurons. A mechanism underlying ischemia-induced cell death is activation of the gene silencing transcription factor REST (repressor element-1 silencing transcription factor)/NRSF (neuron-restrictive silencing factor) and REST-dependent suppression of the AMPA receptor subunit GluR2 in CA1 neurons destined to die. Here we show that REST regulates an additional gene target, OPRM1 (mu opioid receptor 1 or MOR-1). MORs are abundantly expressed by basket cells and other inhibitory interneurons of CA1. Global ischemia induces a marked decrease in MOR-1 mRNA and protein expression that is specific to the selectively vulnerable area CA1, as assessed by quantitative real-time RT-PCR, Western blotting, and ChIP. We further show that OPRM1 gene silencing is REST-dependent and occurs via epigenetic modifications. Ischemia promotes deacetylation of core histone proteins H3 and H4 and dimethylation of histone H3 at lysine-9 (H3-K9) over the MOR-1 promoter, an signature of epigenetic gene silencing. Acute knockdown of MOR-1 gene expression by administration of antisense oligodeoxynucleotides to hippocampal slices in vitro or injection of the MOR antagonist naloxone to rats in vivo affords protection against ischemia-induced death of CA1 pyramidal neurons. These findings implicate MORs in ischemia-induced death of CA1 pyramidal neurons and document epigenetic remodeling of expression of OPRM1 in CA1 inhibitory interneurons.


Subject(s)
Epigenesis, Genetic , Hippocampus/metabolism , Ischemia , Neurons/metabolism , Receptors, Opioid, mu/genetics , Receptors, Opioid, mu/physiology , Animals , Cell Survival , Histones/metabolism , Ischemia/metabolism , Male , Naloxone/pharmacology , Narcotic Antagonists/pharmacology , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley , Receptors, Opioid, mu/metabolism , Time Factors
9.
J Neurosci ; 26(25): 6851-62, 2006 Jun 21.
Article in English | MEDLINE | ID: mdl-16793892

ABSTRACT

Transient global ischemia is a neuronal insult that induces delayed cell death. A hallmark event in the early post-ischemic period is enhanced permeability of mitochondrial membranes. The precise mechanisms by which mitochondrial function is disrupted are, as yet, unclear. Here we show that global ischemia promotes alterations in mitochondrial membrane contact points, a rise in intramitochondrial Zn2+, and activation of large, multi-conductance channels in mitochondrial outer membranes by 1 h after insult. Mitochondrial channel activity was associated with enhanced protease activity and proteolytic cleavage of BCL-xL to generate its pro-death counterpart, deltaN-BCL-xL. The findings implicate deltaN-BCL-xL in large, multi-conductance channel activity. Consistent with this, large channel activity was mimicked by introduction of recombinant deltaN-BCL-xL to control mitochondria and blocked by introduction of a functional BCL-xL antibody to post-ischemic mitochondria via the patch pipette. Channel activity was also inhibited by nicotinamide adenine dinucleotide, indicative of a role for the voltage-dependent anion channel (VDAC) of the outer mitochondrial membrane. In vivo administration of the membrane-impermeant Zn2+ chelator CaEDTA before ischemia or in vitro application of the membrane-permeant Zn2+ chelator tetrakis-(2-pyridylmethyl) ethylenediamine attenuated channel activity, suggesting a requirement for Zn2+. These findings reveal a novel mechanism by which ischemic insults disrupt the functional integrity of the outer mitochondrial membrane and implicate deltaN-BCL-xL and VDAC in the large, Zn2+-dependent mitochondrial channels observed in post-ischemic hippocampal mitochondria.


Subject(s)
Brain Ischemia/pathology , Ion Channels/physiology , Mitochondria/physiology , Zinc/metabolism , Animals , Blotting, Western/methods , Caspases/metabolism , Chelating Agents/pharmacology , Diagnostic Imaging/methods , Dose-Response Relationship, Drug , Ethylenediamines/pharmacology , Ion Channel Gating/drug effects , Ion Channel Gating/physiology , Ion Channel Gating/radiation effects , Ion Channels/classification , Male , Membrane Potentials/drug effects , Membrane Potentials/physiology , Membrane Potentials/radiation effects , Microscopy, Electron, Transmission/methods , Mitochondria/drug effects , Mitochondria/pathology , Mitochondria/ultrastructure , NAD/pharmacology , Patch-Clamp Techniques/methods , Rats , Rats, Sprague-Dawley , Synaptosomes/drug effects , Synaptosomes/physiology , Synaptosomes/ultrastructure , Xanthenes
10.
Neurosci Lett ; 364(2): 101-5, 2004 Jul 01.
Article in English | MEDLINE | ID: mdl-15196687

ABSTRACT

Developing animals are known to be resistant to cerebral ischemia. To investigate the mechanisms by which developing animals exhibit ischemic resistance, we examined the changes in intracellular calcium ([Ca2+]i) after oxygen-glucose deprivation (OGD) using hippocampal slices from gerbils. We found that increases of [Ca2+]i in hippocampal CA1 neurons is significantly less after OGD in developing gerbils than in adults. Western blot analysis of AMPA (alpha-amino-3-hydroxy-5-methyl-4-isoxazole-propionic acid) receptors (AMPARs) showed that GluR2 expression, but not that of the other AMPARs is significantly higher in developing gerbils than in adults. Expression of the anti-apoptotic proteins such as HSP70, Bcl-XL, and plasma membrane Ca2+-ATPase type1 (PMCA1) are not higher in the developing gerbils than in adults. These results suggest that the higher expression of GluR2 is important for the smaller increases in [Ca2+]i and enhanced resistance to ischemia-induced neuronal damage in developing animals.


Subject(s)
Aging/physiology , Brain Ischemia/metabolism , Calcium/metabolism , Receptors, AMPA/biosynthesis , Receptors, AMPA/genetics , Up-Regulation/physiology , Animals , Blotting, Western , Calcium-Transporting ATPases/biosynthesis , Calcium-Transporting ATPases/genetics , Carotid Artery, Common/physiology , Fluorescent Dyes , Gerbillinae , HSP70 Heat-Shock Proteins/biosynthesis , HSP70 Heat-Shock Proteins/genetics , Hippocampus/metabolism , In Vitro Techniques
11.
Neurosci Lett ; 362(3): 216-9, 2004 May 27.
Article in English | MEDLINE | ID: mdl-15158018

ABSTRACT

In normal gerbils, intracellular zinc ions ([Zn2+]i) and calcium ions ([Ca2+]i) accumulate in hippocampal CA1 neurons after global ischemia. We examined whether ischemic preconditioning modifies these changes in gerbil hippocampal slices. In normal slices, large increases in [Zn2+]i and [Ca2+]i were observed in the stratum radiatum of the CA1 area after oxygen-glucose deprivation. In preconditioned slices, there were significantly decreased peak levels of [Zn2+]i and [Ca2+]i in CA1. However, there were no differences in the peak levels of these ions in CA3 and dentate gyrus. These results suggest that modified [Zn2+]i and [Ca2+]i accumulation after an ischemic insult might be important for the mechanisms of ischemic tolerance induced by preconditioning.


Subject(s)
Glucose/deficiency , Hippocampus/cytology , Hypoxia/metabolism , Ischemia/metabolism , Neurons/metabolism , Zinc/metabolism , Animals , Calcium/metabolism , Diagnostic Imaging/methods , Extracellular Space/metabolism , Gerbillinae , In Vitro Techniques , Ischemic Preconditioning , Male
12.
Neurosci Lett ; 328(1): 25-8, 2002 Aug 02.
Article in English | MEDLINE | ID: mdl-12123851

ABSTRACT

We studied the effects of a wasp toxin beta-pompilidotoxin (beta-PMTX) on rat hippocampal CA1 interneurons by the current-clamp technique. The firing patterns of pyramidal neurons and pyramidale interneurons were not affected by beta-PMTX, but in oriens and radiatum interneurons, beta-PMTX converted the action potentials to prolonged depolarizing potentials by slowing the inactivation of Na(+) channels. In lacunosum moleculare interneurons, beta-PMTX induced initial bursting spikes followed by block of succeeding spikes. Comparison of beta-PMTX with a sea anemone toxin, ATX II, revealed that ATX II altered the firing properties of pyramidal neurons and pyramidale interneurons that were unchanged by beta-PMTX. Our results suggest that beta-PMTX modulates Na(+) currents in CA1 interneurons differently in various CA1 neurons and the toxin is useful to classify Na(+) channel subtypes.


Subject(s)
Action Potentials/physiology , Hippocampus/metabolism , Interneurons/metabolism , Neural Inhibition/physiology , Neurotoxins/pharmacology , Pyramidal Cells/metabolism , Sodium Channels/metabolism , Action Potentials/drug effects , Animals , Cnidarian Venoms/pharmacology , Excitatory Postsynaptic Potentials/drug effects , Excitatory Postsynaptic Potentials/physiology , Hippocampus/cytology , Hippocampus/drug effects , Insect Proteins , Interneurons/cytology , Interneurons/drug effects , Male , Neural Inhibition/drug effects , Neural Pathways/cytology , Neural Pathways/drug effects , Neural Pathways/metabolism , Organ Culture Techniques , Pyramidal Cells/cytology , Pyramidal Cells/drug effects , Rats , Rats, Wistar , Sodium Channels/drug effects , Synaptic Transmission/drug effects , Synaptic Transmission/physiology , Wasp Venoms
SELECTION OF CITATIONS
SEARCH DETAIL
...