Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 32
Filter
Add more filters










Publication year range
1.
J Neurosci ; 42(30): 5843-5859, 2022 07 27.
Article in English | MEDLINE | ID: mdl-35732494

ABSTRACT

Temporal lobe epilepsy (TLE), the most common focal seizure disorder in adults, can be instigated in experimental animals by convulsant-induced status epilepticus (SE). Principal hippocampal neurons from SE-experienced epileptic male rats (post-SE neurons) display markedly augmented spike output compared with neurons from nonepileptic animals (non-SE neurons). This enhanced firing results from a cAMP-dependent protein kinase A-mediated inhibition of KCa3.1, a subclass of Ca2+-gated K+ channels generating the slow afterhyperpolarizing Ca2+-gated K+ current (IsAHP). The inhibition of KCa3.1 in post-SE neurons leads to a marked reduction in amplitude of the IsAHP that evolves during repetitive firing, as well as in amplitude of the associated Ca2+-dependent component of the slow afterhyperpolarization potential (KCa-sAHP). Here we show that KCa3.1 inhibition in post-SE neurons is induced by corticotropin releasing factor (CRF) through its Type 1 receptor (CRF1R). Acute application of CRF1R antagonists restores KCa3.1 activity in post-SE neurons, normalizing KCa-sAHP/IsAHP amplitudes and neuronal spike output, without affecting these variables in non-SE neurons. Moreover, pharmacological antagonism of CRF1Rs in vivo reduces the frequency of spontaneous recurrent seizures in post-SE chronically epileptic rats. These findings may provide a new vista for treating TLE.SIGNIFICANCE STATEMENT Epilepsy, a common neurologic disorder, often develops following a brain insult. Identifying key cellular mechanisms underlying acquired epilepsy is critical for developing effective antiepileptic therapies. In an experimental model of acquired epilepsy, principal hippocampal neurons manifest hyperexcitability because of downregulation of KCa3.1, a subtype of Ca2+-gated K+ ion channels. We show that KCa3.1 downregulation is mediated by corticotropin releasing factor (CRF) acting through its Type 1 receptor (CRF1R). Congruently, acute application of selective CRF1R antagonists restores KCa3.1 channel activity, leading to normalization of neuronal excitability. In the same model, injection of a CRF1R antagonist to epileptic animals markedly decreases the frequency of electrographic seizures. Therefore, targeting CRF1Rs may provide a new strategy in the treatment of acquired epilepsy.


Subject(s)
Corticotropin-Releasing Hormone , Epilepsy, Temporal Lobe , Epilepsy , Intermediate-Conductance Calcium-Activated Potassium Channels , Status Epilepticus , Animals , Corticotropin-Releasing Hormone/metabolism , Disease Models, Animal , Epilepsy/drug therapy , Epilepsy/metabolism , Epilepsy, Temporal Lobe/metabolism , Hippocampus/metabolism , Intermediate-Conductance Calcium-Activated Potassium Channels/metabolism , Male , Neurons/physiology , Rats , Status Epilepticus/metabolism
2.
J Physiol ; 599(15): 3735-3754, 2021 08.
Article in English | MEDLINE | ID: mdl-34148230

ABSTRACT

KEY POINTS: Stimulation of postsynaptic muscarinic receptors was shown to excite principal hippocampal neurons by modulating several membrane ion conductances. We show here that activation of postsynaptic muscarinic receptors also causes neuronal excitation by inhibiting Na+ /K+ -ATPase activity. Muscarinic Na+ /K+ -ATPase inhibition is mediated by two separate signalling pathways that lead downstream to enhanced Na+ /K+ -ATPase phosphorylation by activating protein kinase C and protein kinase G. Muscarinic excitation through Na+ /K+ -ATPase inhibition is probably involved in cholinergic modulation of hippocampal activity and may turn out to be a widespread mechanism of neuronal excitation in the brain. ABSTRACT: Stimulation of muscarinic cholinergic receptors on principal hippocampal neurons enhances intrinsic neuronal excitability by modulating several membrane ion conductances. The electrogenic Na+ /K+ -ATPase (NKA; the 'Na+ pump') is a ubiquitous regulator of intrinsic neuronal excitability, generating a hyperpolarizing current to thwart excessive neuronal firing. Using electrophysiological and pharmacological methodologies in rat hippocampal slices, we show that neuronal NKA pumping activity is also subjected to cholinergic regulation. Stimulation of postsynaptic muscarinic, but not nicotinic, cholinergic receptors activates membrane-bound phospholipase C and hydrolysis of membrane-integral phosphatidylinositol 4,5-bisphosphate into diacylglycerol (DAG) and inositol 1,4,5-triphosphate (IP3 ). Along one signalling pathway, DAG activates protein kinase C (PKC). Along a second signalling pathway, IP3 causes Ca2+ release from the endoplasmic reticulum, facilitating nitric oxide (NO) production. The rise in NO levels stimulates cGMP synthesis by guanylate-cyclase, activating protein kinase G (PKG). The two pathways converge to cause partial NKA inhibition through enzyme phosphorylation by PKC and PKG, leading to a marked increase in intrinsic neuronal excitability. This novel mechanism of neuronal NKA regulation probably contributes to the cholinergic modulation of hippocampal activity in spatial navigation, learning and memory.


Subject(s)
Hippocampus , Sodium-Potassium-Exchanging ATPase , Animals , Cholinergic Agents , Cyclic GMP-Dependent Protein Kinases , Hippocampus/metabolism , Neurons/metabolism , Rats , Sodium-Potassium-Exchanging ATPase/metabolism
3.
J Neurosci ; 40(5): 974-995, 2020 01 29.
Article in English | MEDLINE | ID: mdl-31959697

ABSTRACT

Multiple insults to the brain lead to neuronal cell death, thus raising the question to what extent can lost neurons be replenished by adult neurogenesis. Here we focused on the hippocampus and especially the dentate gyrus (DG), a vulnerable brain region and one of the two sites where adult neuronal stem cells (NSCs) reside. While adult hippocampal neurogenesis was extensively studied with regard to its contribution to cognitive enhancement, we focused on their underestimated capability to repair a massively injured, nonfunctional DG. To address this issue, we inflicted substantial DG-specific damage in mice of either sex either by diphtheria toxin-based ablation of >50% of mature DG granule cells (GCs) or by prolonged brain-specific VEGF overexpression culminating in extensive, highly selective loss of DG GCs (thereby also reinforcing the notion of selective DG vulnerability). The neurogenic system promoted effective regeneration by increasing NSCs proliferation/survival rates, restoring a nearly original DG mass, promoting proper rewiring of regenerated neurons to their afferent and efferent partners, and regaining of lost spatial memory. Notably, concomitantly with the natural age-related decline in the levels of neurogenesis, the regenerative capacity of the hippocampus also subsided with age. The study thus revealed an unappreciated regenerative potential of the young DG and suggests hippocampal NSCs as a critical reservoir enabling recovery from catastrophic DG damage.SIGNIFICANCE STATEMENT Adult hippocampal neurogenesis has been extensively studied in the context of its role in cognitive enhancement, but whether, and to what extent can dentate gyrus (DG)-resident neural stem cells drive regeneration of an injured DG has remained unclear. Here we show that DG neurogenesis acts to replace lost neurons and restore lost functions even following massive (>50%) neuronal loss. Age-related decline of neurogenesis is paralleled by a progressive decline of regenerative capacity. Considering also the exceptional vulnerability of the DG to insults, these findings provide a further rationale for maintaining DG neurogenesis in adult life.


Subject(s)
Dentate Gyrus/physiopathology , Neural Stem Cells/physiology , Neurogenesis/physiology , Animals , Cell Proliferation , Cell Survival , Dentate Gyrus/injuries , Dentate Gyrus/pathology , Female , Male , Mice, Transgenic
4.
J Neurosci ; 39(50): 9914-9926, 2019 12 11.
Article in English | MEDLINE | ID: mdl-31672789

ABSTRACT

Brain insults, such as trauma, stroke, anoxia, and status epilepticus (SE), cause multiple changes in synaptic function and intrinsic properties of surviving neurons that may lead to the development of epilepsy. Experimentally, a single SE episode, induced by the convulsant pilocarpine, initiates the development of an epileptic condition resembling human temporal lobe epilepsy (TLE). Principal hippocampal neurons from such epileptic animals display enhanced spike output in response to excitatory stimuli compared with neurons from nonepileptic animals. This enhanced firing is negatively related to the size of the slow afterhyperpolarization (sAHP), which is reduced in the epileptic neurons. The sAHP is an intrinsic neuronal negative feedback mechanism consisting normally of two partially overlapping components produced by disparate mechanisms. One component is generated by activation of Ca2+-gated K+ (KCa) channels, likely KCa3.1, consequent to spike Ca2+ influx (the KCa-sAHP component). The second component is generated by enhancement of the electrogenic Na+/K+ ATPase (NKA) by spike Na+ influx (NKA-sAHP component). Here we show that the KCa-sAHP component is markedly reduced in male rat epileptic neurons, whereas the NKA-sAHP component is not altered. The KCa-sAHP reduction is due to the downregulation of KCa3.1 channels, mediated by cAMP-dependent protein kinase A (PKA). This sustained effect can be acutely reversed by applying PKA inhibitors, leading also to normalization of the spike output of epileptic neurons. We propose that the novel "acquired channelopathy" described here, namely, PKA-mediated downregulation of KCa3.1 activity, provides an innovative target for developing new treatments for TLE, hopefully overcoming the pharmacoresistance to traditional drugs.SIGNIFICANCE STATEMENT Epilepsy, a common neurological disorder, often develops following a brain insult. Identifying key molecular and cellular mechanisms underlying acquired epilepsy is critical for developing effective antiepileptic therapies. In an experimental model of acquired epilepsy, we show that principal hippocampal neurons become intrinsically hyperexcitable. This alteration is due predominantly to the downregulation of a ubiquitous class of potassium ion channels, KCa3.1, whose main function is to dampen neuronal excitability. KCa3.1 downregulation is mediated by the cAMP-dependent protein kinase A (PKA) signaling pathway. Most importantly, it can be acutely reversed by PKA inhibitors, leading to recovery of KCa3.1 function and normalization of neuronal excitability. The discovery of this novel epileptogenic mechanism hopefully will facilitate the development of more efficient pharmacotherapy for acquired epilepsy.


Subject(s)
Action Potentials/physiology , Cyclic AMP-Dependent Protein Kinases/metabolism , Epilepsy, Temporal Lobe/physiopathology , Hippocampus/physiopathology , Intermediate-Conductance Calcium-Activated Potassium Channels/metabolism , Neurons/physiology , Animals , Disease Models, Animal , Epilepsy, Temporal Lobe/metabolism , Hippocampus/metabolism , Male , Rats , Rats, Wistar
5.
J Neurosci ; 39(28): 5440-5451, 2019 07 10.
Article in English | MEDLINE | ID: mdl-31085608

ABSTRACT

The Na+/K+-ATPase (NKA) is a ubiquitous membrane-bound enzyme responsible for generating and maintaining the Na+ and K+ electrochemical gradients across the plasmalemma of living cells. Numerous studies in non-neuronal tissues have shown that this transport mechanism is reversibly regulated by phosphorylation/dephosphorylation of the catalytic α subunit and/or associated proteins. In neurons, Na+/K+ transport by NKA is essential for almost all neuronal operations, consuming up to two-thirds of the neuron's energy expenditure. However, little is known about its cellular regulatory mechanisms. Here we have used an electrophysiological approach to monitor NKA transport activity in male rat hippocampal neurons in situ We report that this activity is regulated by a balance between serine/threonine phosphorylation and dephosphorylation. Phosphorylation by the protein kinases PKG and PKC inhibits NKA activity, whereas dephosphorylation by the protein phosphatases PP-1 and PP-2B (calcineurin) reverses this effect. Given that these kinases and phosphatases serve as downstream effectors in key neuronal signaling pathways, they may mediate the coupling of primary messengers, such as neurotransmitters, hormones, and growth factors, to the NKAs, through which multiple brain functions can be regulated or dysregulated.SIGNIFICANCE STATEMENT The Na+/K+-ATPase (NKA), known as the "Na+ pump," is a ubiquitous membrane-bound enzyme responsible for generating and maintaining the Na+ and K+ electrochemical gradients across the plasma membrane of living cells. In neurons, as in most types of cells, the NKA generates the negative resting membrane potential, which is the basis for almost all aspects of cellular function. Here we used an electrophysiological approach to monitor physiological NKA transport activity in single hippocampal pyramidal cells in situ We have found that neuronal NKA activity is oppositely regulated by phosphorylation and dephosphorylation, and we have identified the main protein kinases and phosphatases mediating this regulation. This fundamental form of NKA regulation likely plays a role in multiple brain functions.


Subject(s)
Calcineurin/metabolism , Cyclic GMP-Dependent Protein Kinases/metabolism , Protein Kinase C/metabolism , Protein Phosphatase 1/metabolism , Sodium-Potassium-Exchanging ATPase/metabolism , Animals , Hippocampus/metabolism , Hippocampus/physiology , Male , Membrane Potentials , Neurons/metabolism , Neurons/physiology , Phosphorylation , Rats , Rats, Wistar
6.
Hippocampus ; 28(5): 338-357, 2018 05.
Article in English | MEDLINE | ID: mdl-29431274

ABSTRACT

In many types of CNS neurons, repetitive spiking produces a slow afterhyperpolarization (sAHP), providing sustained, intrinsically generated negative feedback to neuronal excitation. Changes in the sAHP have been implicated in learning behaviors, in cognitive decline in aging, and in epileptogenesis. Despite its importance in brain function, the mechanisms generating the sAHP are still controversial. Here we have addressed the roles of M-type K+ current (IM ), Ca2+ -gated K+ currents (ICa(K) 's) and Na+ /K+ -ATPases (NKAs) current to sAHP generation in adult rat CA1 pyramidal cells maintained at near-physiological temperature (35 °C). No evidence for IM contribution to the sAHP was found in these neurons. Both ICa(K) 's and NKA current contributed to sAHP generation, the latter being the predominant generator of the sAHP, particularly when evoked with short trains of spikes. Of the different NKA isoenzymes, α1 -NKA played the key role, endowing the sAHP a steep voltage-dependence. Thus normal and pathological changes in α1 -NKA expression or function may affect cognitive processes by modulating the inhibitory efficacy of the sAHP.


Subject(s)
CA1 Region, Hippocampal/metabolism , Membrane Potentials/physiology , Potassium Channels, Calcium-Activated/metabolism , Pyramidal Cells/metabolism , Sodium-Potassium-Exchanging ATPase/metabolism , Animals , CA1 Region, Hippocampal/drug effects , Central Nervous System Agents/pharmacology , Feedback, Physiological/drug effects , Male , Membrane Potentials/drug effects , Patch-Clamp Techniques , Potassium Channels, Calcium-Activated/antagonists & inhibitors , Pyramidal Cells/drug effects , Rats, Wistar , Sodium-Potassium-Exchanging ATPase/antagonists & inhibitors , Tissue Culture Techniques
7.
Neuroscience ; 357: 325-337, 2017 08 15.
Article in English | MEDLINE | ID: mdl-28624573

ABSTRACT

OBJECTIVE: In experimental models of temporal lobe epilepsy (TLE), brain neurons manifest multiple changes in intrinsic excitability that contribute to neuronal network hyperexcitability. We have investigated whether the intrinsic firing response gain, quantified by the slope of the function relating the number of evoked spikes (Ns) to input excitatory current intensity (I), is modified in principal rat hippocampal neurons in the pilocarpine-status epilepticus (SE) model of TLE. METHODS: Intracellular recordings were made in CA3 and CA1 pyramidal cells (PCs) and dentate granule cells (GCs) in acute hippocampal slices obtained 7-36days after pilocarpine-SE. Firing response gains were determined empirically from Ns/I relationships and compared to other measured neuronal properties. RESULTS: The firing response gain in all three types of principal neurons, particularly in CA3 PCs, was markedly multiplied following pilocarpine-SE. Analyses of persistent changes in active and passive properties of CA3 PCs suggested that this increase is multifactorial in origin, the major factors being a reduction in amplitude of the slow afterhyperpolarization and an increase in the fraction of bursting neurons. SIGNIFICANCE: Here we show that pilocarpine-SE causes multiplication of the firing response gain in the three principal neurons in the hippocampal trisynaptic pathway. This alteration undoubtedly would contribute to hippocampal hyperexcitability in SE-induced TLE.


Subject(s)
Action Potentials/physiology , Hippocampus/physiopathology , Neuronal Plasticity/physiology , Neurons/physiology , Status Epilepticus/physiopathology , Animals , Disease Models, Animal , Male , Pilocarpine , Rats, Wistar , Tissue Culture Techniques
8.
J Physiol ; 595(3): 713-738, 2017 02 01.
Article in English | MEDLINE | ID: mdl-27506492

ABSTRACT

KEY POINTS: Neuroinflammation associated with CNS insults leads to neuronal hyperexcitability, which may culminate in epileptiform discharges. Application of the endotoxin lipopolysaccharide (LPS) to brain tissue initiates a neuroinflammatory cascade, providing an experimental model to study the mechanisms of neuroinflammatory neuronal hyperexcitability. Here we show that LPS application to hippocampal slices markedly enhances the excitability of CA1 pyramidal cells by inhibiting a specific potassium current, the M-current, generated by KV 7/M channels, which controls the excitability of almost every neuron in the CNS. The LPS-induced M-current inhibition is triggered by sequential activation of microglia, astrocytes and pyramidal cells, mediated by metabotropic purinergic and glutamatergic transmission, leading to blockade of KV 7/M channels by calcium released from intracellular stores. The identification of the downstream molecular target of neuroinflammation, namely the KV 7/M channel, potentially has far reaching implications for the understanding and treatment of many acute and chronic brain disorders. ABSTRACT: Acute brain insults and many chronic brain diseases manifest an innate inflammatory response. The hallmark of this response is glia activation, which promotes repair of damaged tissue, but also induces structural and functional changes that may lead to an increase in neuronal excitability. We have investigated the mechanisms involved in the modulation of neuronal activity by acute inflammation. Initiating inflammatory responses in hippocampal tissue rapidly led to neuronal depolarization and repetitive firing even in the absence of active synaptic transmission. This action was mediated by a complex metabotropic purinergic and glutamatergic glia-to-neuron signalling cascade, leading to the blockade of neuronal KV 7/M channels by Ca2+ released from internal stores. These channels generate the low voltage-activating, non-inactivating M-type K+ current (M-current) that controls intrinsic neuronal excitability, and its inhibition was the predominant cause of the inflammation-induced hyperexcitability. Our discovery that the ubiquitous KV 7/M channels are the downstream target of the inflammation-induced cascade, has far reaching implications for the understanding and treatment of many acute and chronic brain disorders.


Subject(s)
KCNQ Potassium Channels/physiology , Lipopolysaccharides/pharmacology , Pyramidal Cells/drug effects , Animals , Astrocytes/drug effects , Astrocytes/physiology , CA1 Region, Hippocampal/cytology , Calcium/physiology , Male , Pyramidal Cells/physiology , Rats, Sprague-Dawley , Receptor, Metabotropic Glutamate 5/physiology , Receptors, Metabotropic Glutamate/physiology , Receptors, Purinergic P2Y1/physiology
9.
J Neurosci ; 35(46): 15240-53, 2015 Nov 18.
Article in English | MEDLINE | ID: mdl-26586813

ABSTRACT

Dendritic voltage-gated ion channels profoundly shape the integrative properties of neuronal dendrites. In epilepsy, numerous changes in dendritic ion channels have been described, all of them due to either their altered transcription or phosphorylation. In pilocarpine-treated chronically epileptic rats, we describe a novel mechanism that causes an increased proximal dendritic persistent Na(+) current (INaP). We demonstrate using a combination of electrophysiology and molecular approaches that the upregulation of dendritic INaP is due to a relief from polyamine-dependent inhibition. The polyamine deficit in hippocampal neurons is likely caused by an upregulation of the degrading enzyme spermidine/spermine acetyltransferase. Multiphoton glutamate uncaging experiments revealed that the increase in dendritic INaP causes augmented dendritic summation of excitatory inputs. These results establish a novel post-transcriptional modification of ion channels in chronic epilepsy and may provide a novel avenue for treatment of temporal lobe epilepsy. SIGNIFICANCE STATEMENT: In this paper, we describe a novel mechanism that causes increased dendritic persistent Na(+) current. We demonstrate using a combination of electrophysiology and molecular approaches that the upregulation of persistent Na(+) currents is due to a relief from polyamine-dependent inhibition. The polyamine deficit in hippocampal neurons is likely caused by an upregulation of the degrading enzyme spermidine/spermine acetyltransferase. Multiphoton glutamate uncaging experiments revealed that the increase in dendritic persistent Na current causes augmented dendritic summation of excitatory inputs. We believe that these results establish a novel post-transcriptional modification of ion channels in chronic epilepsy.


Subject(s)
CA1 Region, Hippocampal/pathology , Dendrites/physiology , Down-Regulation/physiology , Sodium Channels/physiology , Spermine/metabolism , Status Epilepticus/pathology , Action Potentials/drug effects , Action Potentials/genetics , Analysis of Variance , Animals , Dendrites/drug effects , Disease Models, Animal , Down-Regulation/drug effects , Humans , In Vitro Techniques , Male , Muscarinic Agonists/toxicity , Pilocarpine/toxicity , RNA, Messenger/metabolism , Rats , Rats, Wistar , Sodium Channel Blockers/pharmacology , Sodium Channels/drug effects , Statistics, Nonparametric , Status Epilepticus/chemically induced , Synaptophysin/metabolism , Tetrodotoxin/pharmacology , Up-Regulation/drug effects , Up-Regulation/physiology
10.
Nat Commun ; 6: 8688, 2015 Oct 26.
Article in English | MEDLINE | ID: mdl-26498180

ABSTRACT

Temporal lobe epilepsy (TLE) is the most common focal seizure disorder in adults. In many patients, transient brain insults, including status epilepticus (SE), are followed by a latent period of epileptogenesis, preceding the emergence of clinical seizures. In experimental animals, transcriptional upregulation of CaV3.2 T-type Ca(2+)-channels, resulting in an increased propensity for burst discharges of hippocampal neurons, is an important trigger for epileptogenesis. Here we provide evidence that the metal-regulatory transcription factor 1 (MTF1) mediates the increase of CaV3.2 mRNA and intrinsic excitability consequent to a rise in intracellular Zn(2+) that is associated with SE. Adeno-associated viral (rAAV) transfer of MTF1 into murine hippocampi leads to increased CaV3.2 mRNA. Conversely, rAAV-mediated expression of a dominant-negative MTF1 abolishes SE-induced CaV3.2 mRNA upregulation and attenuates epileptogenesis. Finally, data from resected human hippocampi surgically treated for pharmacoresistant TLE support the Zn(2+)-MTF1-CaV3.2 cascade, thus providing new vistas for preventing and treating TLE.


Subject(s)
Calcium Channels, T-Type/genetics , DNA-Binding Proteins/metabolism , Epilepsy, Temporal Lobe/genetics , Status Epilepticus/genetics , Transcription Factors/metabolism , Zinc/metabolism , Animals , Calcium Channels, T-Type/metabolism , DNA-Binding Proteins/genetics , Epilepsy, Temporal Lobe/metabolism , Gene Expression Regulation , Humans , Male , Mice , Mice, Inbred C57BL , Rats , Status Epilepticus/metabolism , Transcription Factors/genetics , Transcription Factor MTF-1
11.
J Neurosci ; 34(24): 8219-30, 2014 Jun 11.
Article in English | MEDLINE | ID: mdl-24920626

ABSTRACT

Small-conductance Ca(2+)-activated K(+) (SK or K(Ca)2) channels are widely expressed in the CNS. In several types of neurons, these channels were shown to become activated during repetitive firing, causing early spike frequency adaptation. In CA1 pyramidal cells, SK channels in dendritic spines were shown to regulate synaptic transmission. However, the presence of functional SK channels in the somata and their role in controlling the intrinsic firing of these neurons has been controversial. Using whole-cell voltage-clamp and current-clamp recordings in acute hippocampal slices and focal applications of irreversible and reversible SK channel blockers, we provide evidence that functional SK channels are expressed in the somata and proximal dendrites of adult rat CA1 pyramidal cells. Although these channels can generate a medium duration afterhyperpolarizing current, they play only an auxiliary role in controlling the intrinsic excitability of these neurons, secondary to the low voltage-activating, noninactivating K(V)7/M channels. As long as K(V)7/M channels are operative, activation of SK channels during repetitive firing does not notably affect the spike output of CA1 pyramidal cells. However, when K(V)7/M channel activity is compromised, SK channel activation significantly and uniquely reduces spike output of these neurons. Therefore, proximal SK channels provide a "second line of defense" against intrinsic hyperexcitability, which may play a role in multiple conditions in which K(V)7/M channels activity is compromised, such as hyposmolarity.


Subject(s)
Action Potentials/physiology , CA1 Region, Hippocampal/cytology , Dendrites/metabolism , Pyramidal Cells/physiology , Small-Conductance Calcium-Activated Potassium Channels/metabolism , 4-Aminopyridine/pharmacology , Action Potentials/drug effects , Analysis of Variance , Animals , Anthracenes/pharmacology , Apamin/pharmacology , Biophysics , Dendrites/drug effects , Electric Stimulation , In Vitro Techniques , Male , Patch-Clamp Techniques , Potassium Channel Blockers/pharmacology , Potassium Chloride/pharmacology , Pyramidal Cells/cytology , Pyramidal Cells/drug effects , Rats , Time Factors
12.
Toxicology ; 323: 19-25, 2014 Sep 02.
Article in English | MEDLINE | ID: mdl-24881594

ABSTRACT

Poisoning with organophosphates (OPs) may induce status epilepticus (SE), leading to severe brain damage. Our objectives were to investigate whether OP-induced SE leads to the emergence of spontaneous recurrent seizures (SRSs), the hallmark of chronic epilepsy, and if so, to assess the efficacy of benzodiazepine therapy following SE onset in preventing the epileptogenesis. We also explored early changes in hippocampal pyramidal cells excitability in this model. Adult rats were poisoned with the paraoxon (450µg/kg) and immediately treated with atropine (3mg/kg) and obidoxime (20mg/kg) to reduce acute mortality due to peripheral acetylcholinesterase inhibition. Electrical brain activity was assessed for two weeks during weeks 4-6 after poisoning using telemetric electrocorticographic intracranial recordings. All OP-poisoned animals developed SE, which could be suppressed by midazolam. Most (88%) rats which were not treated with midazolam developed SRSs, indicating that they have become chronically epileptic. Application of midazolam 1min following SE onset had a significant antiepileptogenic effect (only 11% of the rats became epileptic; p=0.001 compared to non-midazolam-treated rats). Applying midazolam 30min after SE onset did not significantly prevent chronic epilepsy. The electrophysiological properties of CA1 pyramidal cells, assessed electrophysiologically in hippocampal slices, were not altered by OP-induced SE. Thus we show for the first time that a single episode of OP-induced SE in rats leads to the acquisition of chronic epilepsy, and that this epileptogenic outcome can be largely prevented by immediate, but not delayed, administration of midazolam. Extrapolating these results to humans would suggest that midazolam should be provided together with atropine and an oxime in the immediate pharmacological treatment of OP poisoning.


Subject(s)
Antidotes/therapeutic use , Cholinesterase Inhibitors/toxicity , Epilepsy/prevention & control , Midazolam/therapeutic use , Paraoxon/toxicity , Status Epilepticus/chemically induced , Animals , Atropine/therapeutic use , Cholinesterase Reactivators/therapeutic use , Chronic Disease , Epilepsy/chemically induced , Muscarinic Agonists , Obidoxime Chloride/therapeutic use , Pesticides/toxicity , Pilocarpine , Rats , Rats, Sprague-Dawley , Status Epilepticus/physiopathology
13.
J Physiol ; 590(22): 5895-905, 2012 Nov 15.
Article in English | MEDLINE | ID: mdl-22930274

ABSTRACT

Extracellular zinc can induce numerous acute and persistent physiological and toxic effects in neurons by acting at their plasma membrane or intracellularly following permeation or uptake into them. Zinc acutely and reversibly blocks T-type voltage-gated calcium current (I(CaT)), but the long-term effect of zinc on this current has not been studied. Because chemically induced status epilepticus (SE) results in the release of zinc into the extracellular space, as well as in a long-lasting increase in I(CaT) in CA1 pyramidal cells, we hypothesized that zinc may play a causative role in I(CaT) upregulation. We tested this hypothesis by monitoring for 18 days the effects of zinc and ibotenic acid (a neurotoxic agent serving as control for zinc), injected into the right lateral ventricle, on I(CaT) in rat CA1 pyramidal cells. Both zinc and ibotenic acid caused marked hippocampal lesions on the side of injection, but only minor damage to contralateral hippocampi. Zinc, but not ibotenic acid, caused upregulation of a nickel-sensitive I(CaT) in a subset of contralateral CA1 pyramidal cells, appearing 2 days after injection and lasting for about 2 weeks thereafter. In contrast, acute application of zinc to CA1 pyramidal cells promptly blocked I(CaT). These data indicate that extracellular zinc has a dual effect on I(CaT), blocking it acutely while causing its long-term upregulation. Through the latter effect, zinc may regulate the intrinsic excitability of principal neurons, particularly in pathological conditions associated with enhanced release of zinc, such as SE.


Subject(s)
Action Potentials/drug effects , Calcium Channels, T-Type/drug effects , Hippocampus/physiology , Pyramidal Cells/physiology , Zinc/pharmacology , Action Potentials/physiology , Animals , Calcium Channels, T-Type/physiology , Cell Death , Ibotenic Acid/pharmacology , Male , Nickel/pharmacology , Pyramidal Cells/drug effects , Rats , Zinc/toxicity
14.
J Biol Chem ; 287(19): 15489-501, 2012 May 04.
Article in English | MEDLINE | ID: mdl-22431737

ABSTRACT

The pore-forming Ca(2+) channel subunit Ca(V)3.2 mediates a low voltage-activated (T-type) Ca(2+) current (I(CaT)) that contributes pivotally to neuronal and cardiac pacemaker activity. Despite the importance of tightly regulated Ca(V)3.2 levels, the mechanisms regulating its transcriptional dynamics are not well understood. Here, we have identified two key factors that up- and down-regulate the expression of the gene encoding Ca(V)3.2 (Cacna1h). First, we determined the promoter region and observed several stimulatory and inhibitory clusters. Furthermore, we found binding sites for the transcription factor early growth response 1 (Egr1/Zif268/Krox-24) to be highly overrepresented within the Ca(V)3.2 promoter region. mRNA expression analyses and dual-luciferase promoter assays revealed that the Ca(V)3.2 promoter was strongly activated by Egr1 overexpression in vitro and in vivo. Subsequent chromatin immunoprecipitation assays in NG108-15 cells and mouse hippocampi confirmed specific Egr1 binding to the Ca(V)3.2 promoter. Congruently, whole-cell I(CaT) values were significantly larger after Egr1 overexpression. Intriguingly, Egr1-induced activation of the Ca(V)3.2 promoter was effectively counteracted by the repressor element 1-silencing transcription factor (REST). Thus, Egr1 and REST can bi-directionally regulate Ca(V)3.2 promoter activity and mRNA expression and, hence, the size of I(CaT). This mechanism has critical implications for the regulation of neuronal and cardiac Ca(2+) homeostasis under physiological conditions and in episodic disorders such as arrhythmias and epilepsy.


Subject(s)
Calcium Channels, T-Type/genetics , Early Growth Response Protein 1/metabolism , Gene Expression Regulation , Promoter Regions, Genetic/genetics , Repressor Proteins/metabolism , Animals , Base Sequence , Binding Sites/genetics , Brain/metabolism , Calcium Channels, T-Type/physiology , Cell Line, Tumor , Chromatin Immunoprecipitation , Early Growth Response Protein 1/genetics , HEK293 Cells , Humans , Membrane Potentials , Mice , Molecular Sequence Data , Patch-Clamp Techniques , Protein Binding , Rats , Regulatory Sequences, Nucleic Acid/genetics , Repressor Proteins/genetics , Reverse Transcriptase Polymerase Chain Reaction , Transcription, Genetic , Transfection
15.
Brain Pathol ; 22(3): 280-94, 2012 May.
Article in English | MEDLINE | ID: mdl-21929559

ABSTRACT

α-Synuclein (α-Syn) is a neuronal protein that accumulates progressively in Parkinson's disease (PD) and related synucleinopathies. Attempting to identify cellular factors that affect α-Syn neuropathology, we previously reported that polyunsaturated fatty acids (PUFAs) promote α-Syn oligomerization and aggregation in cultured cells. We now report that docosahexaenoic acid (DHA), a 22:6 PUFA, affects α-Syn oligomerization by activating retinoic X receptor (RXR) and peroxisome proliferator-activated receptor γ2 (PPARγ2). In addition, we show that dietary changes in brain DHA levels affect α-Syn cytopathology in mice transgenic for the PD-causing A53T mutation in human α-Syn. A diet enriched in DHA, an activating ligand of RXR, increased the accumulation of soluble and insoluble neuronal α-Syn, neuritic injury and astrocytosis. Conversely, abnormal accumulations of α-Syn and its deleterious effects were significantly attenuated by low dietary DHA levels. Our results suggest a role for activated RXR/PPARγ 2, obtained by elevated brain PUFA levels, in α-Syn neuropathology.


Subject(s)
Brain/metabolism , Docosahexaenoic Acids/metabolism , Parkinson Disease/metabolism , Receptors, Cytoplasmic and Nuclear/metabolism , alpha-Synuclein/metabolism , Animals , Brain/pathology , Disease Models, Animal , Mice , Mice, Transgenic , Neurons/metabolism , Neurons/pathology , Parkinson Disease/genetics , Parkinson Disease/pathology , Receptors, Cytoplasmic and Nuclear/genetics , alpha-Synuclein/genetics
16.
J Neurophysiol ; 105(1): 117-29, 2011 Jan.
Article in English | MEDLINE | ID: mdl-20980543

ABSTRACT

Brain damage causes multiple changes in synaptic function and intrinsic properties of surviving neurons, leading to the development of chronic epilepsy. In the widely used pilocarpine-status epilepticus (SE) rat model of temporal lobe epilepsy (TLE), a major alteration is the marked increase in the fraction of intrinsically bursting CA1 pyramidal cells. Here we have differentiated between two types of bursting phenotypes: 1) bursting in response to threshold-straddling excitatory current pulses (low-threshold bursting) and 2) bursting only in response to suprathreshold stimuli (high-threshold bursting). Low-threshold bursting prevailed in 46.5% of SE-experienced neurons sampled 1-4 wk after pilocarpine-SE, but was rarely seen in control neurons (1.9%). As previously shown, it appeared to be driven predominantly by a T-type Ca(2+) current (I(CaT)) in the apical dendrites. After blocking low-threshold bursting with Ni(2+), the same neurons still manifested a high-threshold bursting phenotype. Another 40.1% of SE-experienced neurons displayed only a high-threshold bursting phenotype and the remaining 13.4% of these neurons were nonbursters. Altogether, high-threshold bursting prevailed in 86.6% of SE-experienced neurons, but only in 33.0% of control neurons. Several lines of evidence indicated that high-threshold bursting is driven by persistent Na(+) current (I(NaP)) at or near the soma. Congruently, I(NaP) was 1.5-fold larger in SE-experienced versus control neurons. We conclude that an increase in I(NaP), conjointly with an increase in I(CaT), strongly contributes to the predominance of bursting phenotypes in CA1 pyramidal cells early after pilocarpine-SE and thus likely plays a role in the development of a chronic epileptic condition in this TLE model.


Subject(s)
CA1 Region, Hippocampal/physiopathology , Neurons/physiology , Sodium Channels/physiology , Status Epilepticus/physiopathology , Animals , CA1 Region, Hippocampal/cytology , CA1 Region, Hippocampal/drug effects , Calcium Channel Blockers/pharmacology , Calcium Channels, T-Type/drug effects , Calcium Channels, T-Type/physiology , Male , Models, Animal , Neurons/drug effects , Patch-Clamp Techniques , Pilocarpine/adverse effects , Rats , Rats, Inbred Strains , Rats, Wistar , Sodium Channel Blockers/pharmacology , Sodium Channels/drug effects , Status Epilepticus/chemically induced , Tetrodotoxin/pharmacology , Time Factors
17.
J Neurosci ; 29(36): 11098-111, 2009 Sep 09.
Article in English | MEDLINE | ID: mdl-19741116

ABSTRACT

Modest decreases in extracellular osmolarity induce brain hyperexcitability that may culminate in epileptic seizures. At the cellular level, moderate hyposmolarity markedly potentiates the intrinsic neuronal excitability of principal cortical neurons without significantly affecting their volume. The most conspicuous cellular effect of hyposmolarity is converting regular firing neurons to burst-firing mode. This effect is underlain by hyposmotic facilitation of the spike afterdepolarization (ADP), but its ionic mechanism is unknown. Because blockers of K(V)7 (KCNQ) channels underlying neuronal M-type K(+) currents (K(V)7/M channels) also cause spike ADP facilitation and bursting, we hypothesized that lowering osmolarity inhibits these channels. Using current- and voltage-clamp recordings in CA1 pyramidal cells in situ, we have confirmed this hypothesis. Furthermore, we show that hyposmotic inhibition of K(V)7/M channels is mediated by an increase in intracellular Ca(2+) concentration via release from internal stores but not via influx of extracellular Ca(2+). Finally, we show that interfering with internal Ca(2+)-mediated inhibition of K(V)7/M channels entirely protects against hyposmotic ADP facilitation and bursting, indicating the exclusivity of this novel mechanism in producing intrinsic neuronal hyperexcitability in hyposmotic conditions.


Subject(s)
KCNQ Potassium Channels/physiology , Neurons/metabolism , Osmosis/physiology , Action Potentials/physiology , Animals , Ion Channel Gating/physiology , Male , Neurons/physiology , Patch-Clamp Techniques , Pyramidal Cells/metabolism , Rats
18.
J Neurosci ; 28(49): 13341-53, 2008 Dec 03.
Article in English | MEDLINE | ID: mdl-19052226

ABSTRACT

In both humans and animals, an insult to the brain can lead, after a variable latent period, to the appearance of spontaneous epileptic seizures that persist for life. The underlying processes, collectively referred to as epileptogenesis, include multiple structural and functional neuronal alterations. We have identified the T-type Ca(2+) channel Ca(v)3.2 as a central player in epileptogenesis. We show that a transient and selective upregulation of Ca(v)3.2 subunits on the mRNA and protein levels after status epilepticus causes an increase in cellular T-type Ca(2+) currents and a transitional increase in intrinsic burst firing. These functional changes are absent in mice lacking Ca(v)3.2 subunits. Intriguingly, the development of neuropathological hallmarks of chronic epilepsy, such as subfield-specific neuron loss in the hippocampal formation and mossy fiber sprouting, was virtually completely absent in Ca(v)3.2(-/-) mice. In addition, the appearance of spontaneous seizures was dramatically reduced in these mice. Together, these data establish transcriptional induction of Ca(v)3.2 as a critical step in epileptogenesis and neuronal vulnerability.


Subject(s)
Calcium Channels, T-Type/genetics , Calcium Signaling/genetics , Epilepsy, Temporal Lobe/genetics , Hippocampus/metabolism , Neurons/metabolism , Up-Regulation/genetics , Animals , Calcium Channels, T-Type/metabolism , Channelopathies/genetics , Channelopathies/metabolism , Channelopathies/physiopathology , Disease Models, Animal , Epilepsy, Temporal Lobe/chemically induced , Epilepsy, Temporal Lobe/physiopathology , Gene Expression Regulation/drug effects , Gene Expression Regulation/genetics , Genetic Predisposition to Disease/genetics , Hippocampus/physiopathology , Male , Mice , Mice, Knockout , Mossy Fibers, Hippocampal/metabolism , Mossy Fibers, Hippocampal/physiopathology , Muscarinic Agonists/pharmacology , Nerve Degeneration/genetics , Nerve Degeneration/metabolism , Nerve Degeneration/physiopathology , Neurons/drug effects , Pilocarpine/pharmacology , Protein Subunits/genetics , Protein Subunits/metabolism , Rats , Rats, Wistar , Transcriptional Activation/genetics
19.
J Physiol ; 586(22): 5437-53, 2008 Nov 15.
Article in English | MEDLINE | ID: mdl-18801845

ABSTRACT

Early in development, network activity in the hippocampus is characterized by recurrent synchronous bursts, whose cellular correlates are giant depolarizing potentials (GDPs). The propensity for generating GDPs is attributed to GABAergic synaptic transmission being depolarizing and excitatory in neonatal neurons. However, developmental regulation of intrinsic conductances may also influence GDPs generation. A likely candidate is the non-inactivating, low-threshold, muscarinic-sensitive K(+) current (M current; I(m)), which down-regulates intrinsic bursting activity in adult hippocampal pyramidal neurons. Western blot analysis of homogenates of the CA3 hippocampal region showed that expression of the Kv7.2 subunit, one of the constituents of neuronal M channels, is weak in neonatal neurons, and markedly increases after the first postnatal week. Likewise, the density of I(m) was very low in neonatal CA3 pyramidal cells and increased later on. Spontaneously occurring intrinsic bursts in neonatal neurons were longer and more robust, and recurred more regularly, than in juvenile neurons. The I(m) blocker linopirdine only mildly affected intrinsic bursting in neonatal neurons, but strongly facilitated and regularized it in juvenile neurons. We conclude that the low expression of Kv7/M channels and the depolarizing action of GABA early after birth enhance intrinsic bursting and neuronal synchronization leading to generation of GDPs within the hippocampal network.


Subject(s)
Hippocampus/metabolism , KCNQ2 Potassium Channel/metabolism , Action Potentials/drug effects , Animals , Animals, Newborn , Carbamates/pharmacology , Hippocampus/cytology , Hippocampus/drug effects , Hippocampus/growth & development , In Vitro Techniques , Indoles/pharmacology , KCNQ2 Potassium Channel/agonists , KCNQ2 Potassium Channel/antagonists & inhibitors , Kinetics , Nerve Net/cytology , Nerve Net/growth & development , Nerve Net/metabolism , Phenylenediamines/pharmacology , Potassium Channel Blockers/pharmacology , Pyramidal Cells/drug effects , Pyramidal Cells/metabolism , Pyridines/pharmacology , Rats , Rats, Wistar , Sodium Potassium Chloride Symporter Inhibitors , Sodium-Potassium-Chloride Symporters/metabolism , Solute Carrier Family 12, Member 2 , Synaptic Transmission/drug effects , gamma-Aminobutyric Acid/metabolism , gamma-Aminobutyric Acid/pharmacology
20.
J Neurophysiol ; 100(4): 2361-80, 2008 Oct.
Article in English | MEDLINE | ID: mdl-18650312

ABSTRACT

In many neuron types, the axon initial segment (AIS) has the lowest threshold for action potential generation. Its active properties are determined by the targeted expression of specific voltage-gated channel subunits. We show that the Na+ channel NaV1.6 displays a striking aggregation at the AIS of cortical neurons. To assess the functional role of this subunit, we used Scn8amed mice that are deficient for NaV1.6 subunits but still display prominent Na+ channel aggregation at the AIS. In CA1 pyramidal cells from Scn8amed mice, we found a depolarizing shift in the voltage dependence of activation of the transient Na+ current (INaT), indicating that NaV1.6 subunits activate at more negative voltages than other NaV subunits. Additionally, persistent and resurgent Na+ currents were significantly reduced. Current-clamp recordings revealed a significant elevation of spike threshold in Scn8amed mice as well as a shortening of the estimated delay between spike initiation at the AIS and its arrival at the soma. In combination with simulations using a realistic computer model of a CA1 pyramidal cell, our results imply that a hyperpolarized voltage dependence of activation of AIS NaV1.6 channels is important both in determining spike threshold and localizing spike initiation to the AIS. In addition to altered spike initiation, Scn8amed mice also showed a strongly reduced spike gain as expected with combined changes in persistent and resurgent currents and spike threshold. These results suggest that NaV1.6 subunits at the AIS contribute significantly to its role as spike trigger zone and shape repetitive discharge properties of CA1 neurons.


Subject(s)
Axons/physiology , Cerebral Cortex/physiology , Nerve Tissue Proteins/physiology , Pyramidal Cells/physiology , Sodium Channels/physiology , Action Potentials/physiology , Algorithms , Animals , Calcium Channels/physiology , Calcium Signaling/physiology , Cerebral Cortex/cytology , Computer Simulation , Electrophysiology , Immunohistochemistry , Membrane Potentials/physiology , Mice , Mice, Knockout , Models, Neurological , Models, Statistical , Motor Endplate/genetics , Motor Endplate/physiology , NAV1.6 Voltage-Gated Sodium Channel , Nerve Tissue Proteins/genetics , Patch-Clamp Techniques , Potassium Channels/physiology , Pyramidal Cells/ultrastructure , Sodium Channels/genetics , Temperature
SELECTION OF CITATIONS
SEARCH DETAIL
...