Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
1.
J Neurosci ; 34(21): 7047-58, 2014 May 21.
Article in English | MEDLINE | ID: mdl-24849341

ABSTRACT

Specific missense mutations in the CACNA1A gene, which encodes a subunit of voltage-gated CaV2.1 channels, are associated with familial hemiplegic migraine type 1 (FHM1), a rare monogenic subtype of common migraine with aura. We used transgenic knock-in (KI) mice harboring the human pathogenic FHM1 mutation S218L to study presynaptic Ca(2+) currents, EPSCs, and in vivo activity at the calyx of Held synapse. Whole-cell patch-clamp recordings of presynaptic terminals from S218L KI mice showed a strong shift of the calcium current I-V curve to more negative potentials, leading to an increase in basal [Ca(2+)]i, increased levels of spontaneous transmitter release, faster recovery from synaptic depression, and enhanced synaptic strength despite smaller action-potential-elicited Ca(2+) currents. The gain-of-function of transmitter release of the S218L mutant was reproduced in vivo, including evidence for an increased release probability, demonstrating its relevance for glutamatergic transmission. This synaptic phenotype may explain the misbalance between excitation and inhibition in neuronal circuits resulting in a persistent hyperexcitability state and other migraine-relevant mechanisms such as an increased susceptibility to cortical spreading depression.


Subject(s)
Brain Stem/physiology , Calcium Channels, N-Type/genetics , Calcium/metabolism , Migraine with Aura/genetics , Migraine with Aura/metabolism , Mutation/genetics , Synapses/physiology , Agatoxins/pharmacology , Animals , Brain Stem/cytology , Disease Models, Animal , Humans , In Vitro Techniques , Mice , Mice, Inbred C57BL , Mice, Transgenic , Migraine with Aura/pathology , Migraine with Aura/physiopathology , Neurotoxins/pharmacology , Sodium Channel Blockers/pharmacology , Synapses/drug effects , Synapses/genetics , Tetrodotoxin/pharmacology , Time Factors
2.
J Neurosci ; 34(34): 11385-98, 2014 Aug 20.
Article in English | MEDLINE | ID: mdl-25143618

ABSTRACT

Replay of neuronal activity during hippocampal sharp wave-ripples (SWRs) is essential in memory formation. To understand the mechanisms underlying the initiation of irregularly occurring SWRs and the generation of periodic ripples, we selectively manipulated different components of the CA3 network in mouse hippocampal slices. We recorded EPSCs and IPSCs to examine the buildup of neuronal activity preceding SWRs and analyzed the distribution of time intervals between subsequent SWR events. Our results suggest that SWRs are initiated through a combined refractory and stochastic mechanism. SWRs initiate when firing in a set of spontaneously active pyramidal cells triggers a gradual, exponential buildup of activity in the recurrent CA3 network. We showed that this tonic excitatory envelope drives reciprocally connected parvalbumin-positive basket cells, which start ripple-frequency spiking that is phase-locked through reciprocal inhibition. The synchronized GABA(A) receptor-mediated currents give rise to a major component of the ripple-frequency oscillation in the local field potential and organize the phase-locked spiking of pyramidal cells. Optogenetic stimulation of parvalbumin-positive cells evoked full SWRs and EPSC sequences in pyramidal cells. Even with excitation blocked, tonic driving of parvalbumin-positive cells evoked ripple oscillations. Conversely, optogenetic silencing of parvalbumin-positive cells interrupted the SWRs or inhibited their occurrence. Local drug applications and modeling experiments confirmed that the activity of parvalbumin-positive perisomatic inhibitory neurons is both necessary and sufficient for ripple-frequency current and rhythm generation. These interneurons are thus essential in organizing pyramidal cell activity not only during gamma oscillation, but, in a different configuration, during SWRs.


Subject(s)
Action Potentials/physiology , CA3 Region, Hippocampal/cytology , CA3 Region, Hippocampal/physiology , Neurons/physiology , Vestibular Evoked Myogenic Potentials/physiology , Action Potentials/drug effects , Agatoxins/pharmacology , Anesthetics, Local/pharmacology , Animals , Animals, Newborn , Ankyrins/metabolism , CA3 Region, Hippocampal/drug effects , Calcium Channel Blockers/pharmacology , Channelrhodopsins , Excitatory Postsynaptic Potentials/drug effects , Female , Inhibitory Postsynaptic Potentials/drug effects , Male , Mice , Mice, Transgenic , Models, Neurological , Neurons/drug effects , Parvalbumins/genetics , Signal Detection, Psychological , Tetrodotoxin/pharmacology , Vestibular Evoked Myogenic Potentials/drug effects
3.
Cell Physiol Biochem ; 31(6): 883-91, 2013.
Article in English | MEDLINE | ID: mdl-23817128

ABSTRACT

BACKGROUND/AIMS: Protein kinase Cα (PKCα) is activated by an increase in cytosolic Ca(2+) in red blood cells (RBCs). Previous work has suggested that PKCα directly stimulates the CaV2.1 channel, whereas other studies revealed that CaV2.1 is insensitive to activation by PKC. The aim of this study was to resolve this discrepancy. METHODS: We performed experiments based on a single cell read-out of the intracellular Ca(2+) concentration in terms of Fluo-4 fluorescence intensity and phosphatidylserine exposure to the external membrane leaflet. Measurement modalities included flow cytometry and live cell imaging. RESULTS: Treatment of RBCs with phorbol 12-myristate 13-acetate (PMA) led to two distinct populations of cells with an increase in intracellular Ca(2+): a weak-responding and a strong-responding population. The EC50 of PMA for the number of cells with Ca(2+) elevation was 2.7±1.2 µM; for phosphatidylserine exposure to the external membrane surface, it was 2.8±0.5 µM; and for RBC haemolysis, it was 2.9±0.5 µM. Using pharmacological manipulation with the CaV2.1 inhibitor ω-agatoxin TK and the broad protein kinase C inhibitor Gö6983, we are able to show that there are two independent PMA-activated Ca(2+) entry processes: the first is independent of CaV2.1 and directly PKCα-activated, while the second is associated with a likely indirect activation of CaV2.1. Further studies using lysophosphatidic acid (LPA) as a stimulation agent have provided additional evidence that PKCα and CaV2.1 are not directly interconnected in a signalling chain. CONCLUSION: Although we provide evidence for a lack of interaction between PKCα and CaV2.1 in RBCs, further studies are required to decipher the signalling relationship between LPA, PKCα and CaV2.1.


Subject(s)
Calcium Channels, N-Type/metabolism , Erythrocytes/metabolism , Protein Kinase C-alpha/metabolism , Agatoxins/pharmacology , Aniline Compounds/chemistry , Calcium/metabolism , Calcium Channels, N-Type/chemistry , Calcium Signaling/drug effects , Cell Membrane/drug effects , Erythrocytes/cytology , Erythrocytes/drug effects , Flow Cytometry , Hemolysis , Humans , Indoles/pharmacology , Kinetics , Lysophospholipids/pharmacology , Maleimides/pharmacology , Phosphatidylserines/pharmacology , Protein Kinase C-alpha/antagonists & inhibitors , Tetradecanoylphorbol Acetate/analogs & derivatives , Tetradecanoylphorbol Acetate/pharmacology , Xanthenes/chemistry
4.
Peptides ; 145: 170622, 2021 11.
Article in English | MEDLINE | ID: mdl-34363923

ABSTRACT

Peptides isolated from spider venoms are of pharmacological interest due to their neurotoxic activity, acting on voltage-dependent ion channels present in different types of human body tissues. Three peptide toxins titled as Ap2, Ap3 and Ap5 were purified by RP-HPLC from Acanthoscurria paulensis venom. They were partially sequenced by MALDI In-source Decay method and their sequences were completed and confirmed by transcriptome analysis of the venom gland. The Ap2, Ap3 and Ap5 peptides have, respectively, 42, 41 and 46 amino acid residues, and experimental molecular masses of 4886.3, 4883.7 and 5454.7 Da, with the Ap2 peptide presenting an amidated C-terminus. Amongst the assayed channels - NaV1.1, NaV1.5, NaV1.7, CaV1.2, CaV2.1 and CaV2.2 - Ap2, Ap3 and Ap5 inhibited 20-30 % of CaV2.1 current at 1 µM concentration. Ap3 also inhibited sodium current in NaV1.1, Nav1.5 and Nav1.7 channels by 6.6 ± 1.91 % (p = 0.0276), 4.2 ± 1.09 % (p = 0.0185) and 16.05 ± 2.75 % (p = 0.0282), respectively. Considering that Ap2, Ap3 and Ap5 belong to the 'U'-unknown family of spider toxins, which has few descriptions of biological activity, the present work contributes to the knowledge of these peptides and demonstrates this potential as channel modulators.


Subject(s)
Agatoxins/isolation & purification , Agatoxins/pharmacology , Spider Venoms/chemistry , Agatoxins/chemistry , Animals , CHO Cells , Calcium Channels, N-Type/metabolism , Cricetulus , HEK293 Cells , Humans , Peptides/chemistry , Peptides/isolation & purification , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization , Spiders , Voltage-Gated Sodium Channel Blockers/chemistry , Voltage-Gated Sodium Channel Blockers/pharmacology , Voltage-Gated Sodium Channels/genetics , Voltage-Gated Sodium Channels/metabolism
5.
Life Sci ; 188: 110-117, 2017 Nov 01.
Article in English | MEDLINE | ID: mdl-28882644

ABSTRACT

AIMS: Both N-type and P/Q-type voltage-gated Ca2+ channels (VGCCs) are involved in the induction of long-term potentiation (LTP), the long-lasting increase of synaptic strength, in the central nervous system. To provide further information on the roles of N-type and P/Q-type VGCCs in the induction of LTP at excitatory synapses of trigeminal primary afferents in the spinal trigeminal subnucleus oralis (Vo), we investigated whether they contribute to the induction of LTP by activation of group I metabotropic glutamate receptors (mGluRs). MAIN METHODS: (S)-3,5-Dihydroxyphenylglycine (DHPG; 10µM for 5min), the group I mGluR agonist, was used to induce LTP of excitatory postsynaptic currents that were evoked in the Vo neurons by stimulating the trigeminal track. KEY FINDINGS: Weak blockade of the N-type or P/Q-type VGCCs by ω-conotoxin GVIA or ω-agatoxin IVA, respectively, which inhibited only 20-40% of Ca2+ currents recorded in isolated trigeminal ganglion neurons but had no effect on the basal excitatory synaptic transmission, completely blocked the induction of LTP. In contrast, stronger blockade of the channels, which inhibited >50% of Ca2+ currents and about 30% of basal synaptic transmission, resulted in the development of long-term depression (LTD), the long-lasting decrease of synaptic strength. Interestingly, the postsynaptic mechanism of DHPG-induced LTP, which was determined by paired-pulse ratio, disappeared when LTP was blocked, or LTD occurred, while a presynaptic mechanism still remained. SIGNIFICANCE: Our data suggest that postsynaptic N-type and P/Q-type VGCCs mediate the DHPG-induced LTP at the trigeminal afferent synapses in the Vo.


Subject(s)
Calcium Channels, N-Type/physiology , Calcium Channels, P-Type/physiology , Calcium Channels, Q-Type/physiology , Long-Term Potentiation/physiology , Receptors, Metabotropic Glutamate/physiology , Trigeminal Nucleus, Spinal/physiology , Agatoxins/pharmacology , Animals , Calcium Channel Agonists/pharmacology , Calcium Channel Blockers , Chromones/pharmacology , Female , Long-Term Potentiation/drug effects , Long-Term Synaptic Depression/drug effects , Long-Term Synaptic Depression/physiology , Male , Presynaptic Terminals/physiology , Rats , Receptors, Metabotropic Glutamate/agonists , Synaptic Potentials/physiology , Synaptic Transmission/drug effects , Trigeminal Nucleus, Spinal/drug effects , omega-Conotoxins/pharmacology
6.
Neuroscience ; 339: 109-123, 2016 Dec 17.
Article in English | MEDLINE | ID: mdl-27702646

ABSTRACT

The medial prefrontal cortex (mPFC) plays a key role in higher functions such as memory and attention. In order to demonstrate sensory responses in the mPFC, we used electrophysiological recordings of urethane-anesthetized rats to record somatosensory-evoked potentials (SEPs) or auditory-evoked potentials (AEPs) elicited by whisker deflections and click stimulation, respectively. Contralateral whisker stimulation or auditory stimuli were also applied to study sensory interference in the mPFC. Interference with other sensory stimuli or recent stimulation history reduced whisker responses in the infralimbic and prelimbic cortices of the ventral mPFC. This effect could be mediated by activation of parvalbumin (PV) interneurons since the effect was blocked by the P/Q calcium channel antagonist ω-agatoxin. In contrast, sensory interference or the recent stimulation history was not detected by the dorsal mPFC or the primary somatosensory cortex. Results obtained from retrograde tracer injections in the dorsal and ventral regions of the mPFC indicated that somatosensory and auditory sensory inputs may arrive at the dorsal mPFC through secondary sensory cortical areas, and through the insular and temporal cortical areas. The ventral mPFC may receive sensory information through the strong anatomical connections between the dorsal and ventral mPFC areas. In conclusion, results suggest mPFC plays an important role in sensory processing, which may have important implications in attentional and memory processes.


Subject(s)
Auditory Perception/physiology , Prefrontal Cortex/physiology , Touch Perception/physiology , Adaptation, Physiological/drug effects , Adaptation, Physiological/physiology , Agatoxins/pharmacology , Anesthetics, Intravenous/pharmacology , Animals , Auditory Perception/drug effects , Calcium Channel Blockers/pharmacology , Calcium Channels, P-Type/metabolism , Calcium Channels, Q-Type/metabolism , Evoked Potentials, Auditory/drug effects , Evoked Potentials, Auditory/physiology , Evoked Potentials, Somatosensory/drug effects , Evoked Potentials, Somatosensory/physiology , Female , Male , Neurons/cytology , Neurons/drug effects , Neurons/metabolism , Prefrontal Cortex/cytology , Prefrontal Cortex/drug effects , Rats, Sprague-Dawley , Touch Perception/drug effects , Urethane/pharmacology , Vibrissae/physiology
7.
Neuroscience ; 256: 292-301, 2014 Jan 03.
Article in English | MEDLINE | ID: mdl-24505607

ABSTRACT

Modulation of L-type Ca²âº-channel function by dopamine is a major determinant of the rate of action potential firing by striatal medium spiny neurons. However, the role of these channels in modulating GABA release by nerve terminals in the basal ganglia is unknown. We found that depolarization-induced [³H]GABA release in both the substantia nigra reticulata and the external globus pallidus (GPe), was depressed by about 50% by either the selective L-channel dihydropyridine blocker nifedipine or the P/Q channel blocker ω-agatoxin TK. The effects of these blockers were additive and together eliminated about 90% of depolarization-induced [³H]GABA release. In addition, in the substantia nigra reticulata, dihydropyridines prevented both the stimulation of [³H]GABA release produced by dopamine D1 receptor activation and the inhibition caused by D4 receptor activation. In the GP nifedipine blocked the effects of D2 and A2(A) receptor coactivation as well as the effects of activating adenylyl cyclase with forskolin. ω-Agatoxin TK did not interfere with the action of these modulatory agents. The L-type Ca²âº-channel agonist BAYK 8644 stimulated GABA release in both substantia nigra reticulata and GP. Because dihydropyridine sensitivity is a key criterion to identify L-type Ca²âº-channel activity, our results imply that these channels are determinant of GABA release modulation by dopamine in striatonigral, striatopallidal and pallidonigral terminals.


Subject(s)
Calcium Channels, L-Type/metabolism , Dopamine/pharmacology , Globus Pallidus/drug effects , Substantia Nigra/drug effects , gamma-Aminobutyric Acid/metabolism , 3-Pyridinecarboxylic acid, 1,4-dihydro-2,6-dimethyl-5-nitro-4-(2-(trifluoromethyl)phenyl)-, Methyl ester/pharmacology , Agatoxins/pharmacology , Analysis of Variance , Animals , Calcium Channel Agonists/pharmacology , Dopamine Agents/pharmacology , In Vitro Techniques , Male , Nifedipine/pharmacology , Potassium Chloride/pharmacology , Rats , Rats, Wistar , Tritium/metabolism
8.
Neurochem Int ; 61(5): 632-9, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22819793

ABSTRACT

The structure of the toxin ω-agatoxin IVB, extracted from the venom of funnel-web spider Agelenopsis aperta, is an important lead structure when considering the design of modulators of synaptic transmission which largely involves P/Q-type (CaV2.1) voltage gated calcium channels (VGCC) at central synapses. Focusing on the loop 2 of the ω-agatoxin IVB that seems to be the most preeminent interacting domain of the toxin with the CaV2.1 VGCC, cyclooctapeptides mimicking this loop were synthesized. While (14)Trp is essential for the binding of the neurotoxin to the CaV2.1 VGCC, the substitution of the (12)Cys for a glycidyl residue led to a cyclooctapeptide named EP14 able to enhance CaV2.1 VGCC-associated currents measured with patch-clamp recordings and to evoke ω-agatoxin IVA-sensitive intracellular Ca(2+) increase as measured by fura-2 spectrofluoroimaging. Furthermore, this cyclooctapeptide was able to potentiate spontaneous excitatory synaptic transmission in a network of cultured hippocampal neurons, consistent with the activation of presynaptic VGCC by EP14. In addition, this peptide did not affect cell survival measured with the MTT assay. Therefore, such new cyclopeptidic structures are potential good candidates for synthesis of new agents aimed at the restoration deficient excitatory synaptic transmission.


Subject(s)
Agatoxins/chemical synthesis , Calcium Channels, N-Type/metabolism , Hippocampus/drug effects , Hippocampus/metabolism , Neurons/drug effects , Neurons/metabolism , Agatoxins/pharmacology , Animals , Cell Survival/drug effects , Cell Survival/physiology , Cells, Cultured , Excitatory Postsynaptic Potentials/drug effects , Excitatory Postsynaptic Potentials/physiology , Hippocampus/cytology , Rats , Rats, Sprague-Dawley
9.
Neuron ; 74(2): 384-96, 2012 Apr 26.
Article in English | MEDLINE | ID: mdl-22542190

ABSTRACT

Functional neural circuit formation during development involves massive elimination of redundant synapses. In the cerebellum, one-to-one connection from excitatory climbing fiber (CF) to Purkinje cell (PC) is established by elimination of early-formed surplus CFs. This process depends on glutamatergic excitatory inputs, but contribution of GABAergic transmission remains unclear. Here, we demonstrate impaired CF synapse elimination in mouse models with diminished GABAergic transmission by mutation of a single allele for the GABA synthesizing enzyme GAD67, by conditional deletion of GAD67 from PCs and GABAergic interneurons or by pharmacological inhibition of cerebellar GAD activity. The impaired CF synapse elimination was rescued by enhancing GABA(A) receptor sensitivity in the cerebellum by locally applied diazepam. Our electrophysiological and Ca2+ imaging data suggest that GABA(A) receptor-mediated inhibition onto the PC soma from molecular layer interneurons influences CF-induced Ca2+ transients in the soma and regulates CF synapse elimination from postnatal day 10 (P10) to around P16.


Subject(s)
Cerebellum/cytology , Cerebellum/growth & development , Purkinje Cells/cytology , Synapses/physiology , gamma-Aminobutyric Acid/metabolism , Agatoxins/pharmacology , Age Factors , Analysis of Variance , Animals , Animals, Newborn , Bicuculline/pharmacology , Biophysical Phenomena/drug effects , Biophysical Phenomena/genetics , Biophysics , Calbindins , Calcium/metabolism , Calcium Channels/metabolism , Chromones/pharmacology , Cytochromes c/pharmacology , Dendrites/ultrastructure , Diazepam/pharmacology , Electric Stimulation , Excitatory Amino Acid Antagonists/pharmacology , Excitatory Postsynaptic Potentials/drug effects , Excitatory Postsynaptic Potentials/genetics , GABA Modulators/pharmacology , GABA-A Receptor Antagonists/pharmacology , Gene Expression Regulation, Developmental/genetics , Gene Expression Regulation, Developmental/physiology , Glutamate Decarboxylase/genetics , Green Fluorescent Proteins/genetics , In Vitro Techniques , Membrane Potentials/genetics , Membrane Potentials/physiology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Microscopy, Confocal , Microscopy, Electron, Transmission , Nerve Tissue Proteins/metabolism , Neurotoxins/pharmacology , Patch-Clamp Techniques , Phospholipase C beta/metabolism , Protein Kinase C/metabolism , Purkinje Cells/physiology , Quinoxalines/pharmacology , Receptors, Glutamate/metabolism , Receptors, Metabotropic Glutamate/metabolism , S100 Calcium Binding Protein G/metabolism , Vesicular Inhibitory Amino Acid Transport Proteins/metabolism
10.
Toxins (Basel) ; 3(1): 17-42, 2011 01.
Article in English | MEDLINE | ID: mdl-22069688

ABSTRACT

Peptide neurotoxins found in animal venoms have gained great interest in the field of neurotransmission. As they are high affinity ligands for calcium, potassium and sodium channels, they have become useful tools for studying channel structure and activity. Peptide neurotoxins represent the clinical potential of ion-channel modulators across several therapeutic fields, especially in developing new strategies for treatment of ion channel-related diseases. The aim of this review is to overview the latest updates in the domain of peptide neurotoxins that affect voltage-gated calcium channels, with a special focus on ω-agatoxins.


Subject(s)
Agatoxins/pharmacology , Calcium Channel Blockers/pharmacology , Calcium Channels , Neurotoxins/pharmacology , Peptides/pharmacology , Agatoxins/isolation & purification , Amino Acid Sequence , Animals , Binding Sites , Calcium Channel Blockers/isolation & purification , Calcium Channels/chemistry , Calcium Channels/metabolism , Calcium Channels/physiology , Ligands , Models, Molecular , Molecular Sequence Data , Neurotoxins/isolation & purification , Peptides/isolation & purification , Protein Binding
SELECTION OF CITATIONS
SEARCH DETAIL