Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 95
Filter
Add more filters










Publication year range
1.
Blood Adv ; 5(17): 3303-3308, 2021 09 14.
Article in English | MEDLINE | ID: mdl-34468723

ABSTRACT

In patients with Gárdos channelopathy (p.R352H), an increased concentration of intracellular Ca2+ was previously reported. This is a surprising finding because the Gárdos channel (KCa3.1) is a K+ channel. Here, we confirm the increased intracellular Ca2+ for patients with the KCa3.1 mutation p.S314P. Furthermore, we provide the concept of KCa3.1 activity resulting in a flickering of red blood cell (RBC) membranepotential, which activates the CaV2.1 channel allowing Ca2+ to enter the RBC. Activity of the nonselective cation channel Piezo1 modulates the aforementioned interplay in away that a closed Piezo1 is in favor of the KCa3.1-CaV2.1 interaction. In contrast, Piezo1 openings compromise the membrane potential flickering, thus limiting the activity of CaV2.1. With the compound NS309, we mimic a gain-of-function mutation of KCa3.1. Assessing the RBC Ca2+ response by Fluo-4-based flow cytometry and by measuring the membrane potential using the Macey-Bennekou-Egée method, we provide data that support the concept of the KCa3.1/CaV2.1/Piezo1 interplay as a partial explanation for an increased number of high Ca2+ RBCs. With the pharmacological inhibition of KCa3.1 (TRAM34 and Senicapoc), CaV2.1 (ω-agatoxin TK), and Piezo1 (GsMTx-4), we could project the NS309 behavior of healthy RBCs to the RBCs of Gárdos channelopathy patients.


Subject(s)
Channelopathies , Agatoxins , Calcium/metabolism , Erythrocytes/metabolism , Humans , Ion Channels/genetics
2.
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
3.
Gen Comp Endocrinol ; 299: 113609, 2020 12 01.
Article in English | MEDLINE | ID: mdl-32916171

ABSTRACT

Over the past decade, in silico genome and transcriptome mining has led to the identification of many new crustacean peptide families, including the agatoxin-like peptides (ALPs), a group named for their structural similarity to agatoxin, a spider venom component. Here, analysis of publicly accessible transcriptomes was used to expand our understanding of crustacean ALPs. Specifically, transcriptome mining was used to investigate the phylogenetic/structural conservation, tissue localization, and putative functions of ALPs in decapod species. Transcripts encoding putative ALP precursors were identified from one or more members of the Penaeoidea (penaeid shrimp), Sergestoidea (sergestid shrimps), Caridea (caridean shrimp), Astacidea (clawed lobsters and freshwater crayfish), Achelata (spiny/slipper lobsters), and Brachyura (true crabs), suggesting a broad, and perhaps ubiquitous, conservation of ALPs in decapods. Comparison of the predicted mature structures of decapod ALPs revealed high levels of amino acid conservation, including eight identically conserved cysteine residues that presumably allow for the formation of four identically positioned disulfide bridges. All decapod ALPs are predicted to have amidated carboxyl-terminals. Two isoforms of ALP appear to be present in most decapod species, one 44 amino acids long and the other 42 amino acids in length, both likely generated by alternative splicing of a single gene. In carideans, a gene or terminal exon duplication appears to have occurred, with alternative splicing producing four ALPs, two 44 and two 42 amino acid isoforms. The identification of ALP precursor-encoding transcripts in nervous system-specific transcriptomes (e.g., Homarus americanus brain, eyestalk ganglia, and cardiac ganglion assemblies, finding confirmed using RT-PCR) suggests that members of this peptide family may serve as locally-released and/or hormonally-delivered neuromodulators in decapods. Their detection in testis- and hepatopancreas-specific transcriptomes suggests that members of the ALP family may also play roles in male reproduction and innate immunity/detoxification.


Subject(s)
Agatoxins/chemistry , Decapoda/genetics , Decapoda/metabolism , Mass Spectrometry/methods , Peptide Fragments/genetics , Peptide Fragments/metabolism , Transcriptome , Amino Acid Sequence , Animals , Cloning, Molecular , Decapoda/classification , Organ Specificity , Phylogeny
4.
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
5.
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
6.
Sci Rep ; 6: 34706, 2016 10 06.
Article in English | MEDLINE | ID: mdl-27708374

ABSTRACT

The first generation transgenic crops used strong constitutive promoters for transgene expression. However, tissue-specific expression is desirable for more precise targeting of transgenes. Moreover, piercing/sucking insects, which are generally resistant to insecticidal Bacillus thuringiensis (Bt) proteins, have emerged as a major pests since the introduction of transgenic crops expressing these toxins. Phloem-specific promoters isolated from Banana bunchy top virus (BBTV) were used for the expression of two insecticidal proteins, Hadronyche versuta (Blue Mountains funnel-web spider) neurotoxin (Hvt) and onion leaf lectin, in tobacco (Nicotiana tabacum). Here we demonstrate that transgenic plants expressing Hvt alone or in combination with onion leaf lectin are resistant to Phenacoccus solenopsis (cotton mealybug), Myzus persicae (green peach aphids) and Bemisia tabaci (silver leaf whitefly). The expression of both proteins under different phloem-specific promoters resulted in close to 100% mortality and provided more rapid protection than Hvt alone. Our results suggest the employment of the Hvt and onion leaf lectin transgenic constructs at the commercial level will reduce the use of chemical pesticides for control of hemipteran insect pests.


Subject(s)
Agatoxins/metabolism , Aphids/physiology , Insecticides/metabolism , Phloem/metabolism , Plant Lectins/metabolism , Plants, Genetically Modified/growth & development , Promoter Regions, Genetic , Agatoxins/genetics , Animals , Aphids/genetics , Babuvirus/genetics , Crops, Agricultural/genetics , Crops, Agricultural/growth & development , Crops, Agricultural/metabolism , Crops, Agricultural/parasitology , Genes, Viral , Organ Specificity , Pest Control, Biological , Plant Diseases/prevention & control , Plant Lectins/genetics , Plants, Genetically Modified/metabolism , Plants, Genetically Modified/parasitology
7.
Microscopy (Oxf) ; 65(4): 337-40, 2016 08.
Article in English | MEDLINE | ID: mdl-27095701

ABSTRACT

ω-Agatoxin IV A, a peptidyl toxin from Agelenopsis aperta venom, selectively binds to voltage-gated P/Q-type calcium channels. ω-Agatoxin IV A has been used as a selective tool in pharmacological and electrophysiological studies. Visualization of P/Q-type calcium channels has previously been accomplished using biotin-conjugated ω-Agatoxin IV A in freshly prepared mouse cerebellar and hippocampal slices (Nakanishi et al, J. Neurosci. Res., 41: , 532, 1995). Here biotinylated ω-agatoxin IV A was applied to transcardially fixed brain slices prepared with various fixatives. ω-Agatoxin IV A did not bind to fixed tissues from P/Q-type calcium channel knockout mice, confirming that binding to normal, fixed tissues was not an artifact. Using transmission electron microscopy, locations of biotinylated ω-agatoxin IV A binding sites visualized with gold-conjugated streptavidin showed a similar pattern to those visualized with antibody. The ability of biotinylated ω-agatoxin IV A to bind to fixed tissue provides a new cytochemical technique to study molecular architecture of synapses.


Subject(s)
Agatoxins/metabolism , Calcium Channels, P-Type/metabolism , Calcium Channels, Q-Type/metabolism , Synapses/metabolism , Tissue Fixation , Animals , Binding Sites , Biotinylation , Brain Stem/physiology , Mice , Microscopy, Electron, Transmission/methods , Microscopy, Immunoelectron/methods , Neurons/physiology , Protein Binding , Rats , Streptavidin/chemistry
8.
J Proteomics ; 132: 77-84, 2016 Jan 30.
Article in English | MEDLINE | ID: mdl-26626629

ABSTRACT

We investigated the peptide inventory of the corpora cardiaca (CC) of the honey bee, Apis mellifera, by direct tissue profiling using MALDI-TOF MS combined with proteomic approaches focusing on cysteine-containing peptides. An agatoxin-like peptide (ALP) was identified as a component of the glandular part of the CC and was associated with the presence of the adipokinetic hormone in mass spectra. Although abundant in the CC, ALP does not belong to the toxins observed in the venom gland of A. mellifera. Homologs of ALP are highly conserved in major groups of arthropods and in line with this we detected ALP in the CC of non-venomous insects such as cockroaches and silverfish. In the American cockroach, Periplaneta americana, ALP was also identified in the CNS and stomatogastric nervous system. This is the first report that establishes the presence of ALPs in the neuroendocrine tissues of insects and further studies are necessary to reveal common functions of these peptides, e.g. as antimicrobial agents, ion channel modulators or classical neuropeptides. BIOLOGICAL SIGNIFICANCE: Among the messenger molecules of the nervous system, neuropeptides represent the structurally most diverse class and basically participate in the regulation of all physiological processes. The set of neuropeptides, their functions and spatial distribution are particularly well-studied in insects. Until now, however, several potential neuropeptide receptors remained orphan, which indicates the existence of so far unknown ligands. In our study, we used proteomic methods such as cysteine modification, enzymatic digestion and peptide derivatization, combined with direct tissue profiling by MALDI-TOF mass spectrometry, for the discovery of novel putative messenger molecules in the neuroendocrine system. The described presence of agatoxin-like peptides in the nervous system of the honey bee and other insects was overseen so far and is thus a remarkable addition to the very well studied neuropeptidome of insects. It is not yet clear, if these toxin-like peptides act as antimicrobial agents, ion channel modulators or classical neuropeptides.


Subject(s)
Agatoxins/chemistry , Agatoxins/metabolism , Bees/metabolism , Insecta/metabolism , Neurosecretory Systems/metabolism , Peptides/chemistry , Agatoxins/analysis , Amino Acid Sequence , Animals , Conserved Sequence , Molecular Sequence Data , Peptides/analysis , Peptides/metabolism , Species Specificity
9.
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
10.
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
11.
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
12.
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
13.
PLoS One ; 8(4): e60498, 2013.
Article in English | MEDLINE | ID: mdl-23573262

ABSTRACT

BACKGROUND: Cystine-knot miniproteins, also known as knottins, have shown great potential as molecular scaffolds for the development of targeted therapeutics and diagnostic agents. For this purpose, previous protein engineering efforts have focused on knottins based on the Ecballium elaterium trypsin inhibitor (EETI) from squash seeds, the Agouti-related protein (AgRP) neuropeptide from mammals, or the Kalata B1 uterotonic peptide from plants. Here, we demonstrate that Agatoxin (AgTx), an ion channel inhibitor found in spider venom, can be used as a molecular scaffold to engineer knottins that bind with high-affinity to a tumor-associated integrin receptor. METHODOLOGY/PRINCIPAL FINDINGS: We used a rational loop-grafting approach to engineer AgTx variants that bound to αvß3 integrin with affinities in the low nM range. We showed that a disulfide-constrained loop from AgRP, a structurally-related knottin, can be substituted into AgTx to confer its high affinity binding properties. In parallel, we identified amino acid mutations required for efficient in vitro folding of engineered integrin-binding AgTx variants. Molecular imaging was used to evaluate in vivo tumor targeting and biodistribution of an engineered AgTx knottin compared to integrin-binding knottins based on AgRP and EETI. Knottin peptides were chemically synthesized and conjugated to a near-infrared fluorescent dye. Integrin-binding AgTx, AgRP, and EETI knottins all generated high tumor imaging contrast in U87MG glioblastoma xenograft models. Interestingly, EETI-based knottins generated significantly lower non-specific kidney imaging signals compared to AgTx and AgRP-based knottins. CONCLUSIONS/SIGNIFICANCE: In this study, we demonstrate that AgTx, a knottin from spider venom, can be engineered to bind with high affinity to a tumor-associated receptor target. This work validates AgTx as a viable molecular scaffold for protein engineering, and further demonstrates the promise of using tumor-targeting knottins as probes for in vivo molecular imaging.


Subject(s)
Agatoxins , Neoplasms/diagnosis , Agatoxins/chemistry , Agatoxins/genetics , Amino Acid Substitution , Animals , Carbocyanines/chemistry , Cysteine/genetics , Cystine Knot Motifs , Female , Fluorescent Dyes/chemistry , Humans , Integrin alphaVbeta3/metabolism , K562 Cells , Mice , Mice, Nude , Mutagenesis, Site-Directed , Neoplasm Transplantation , Protein Binding , Protein Engineering , Protein Folding
14.
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
15.
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
16.
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
17.
J Neurophysiol ; 105(6): 2897-906, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21471392

ABSTRACT

N-methyl-D-aspartate (NMDA) receptors (NMDARs) are implicated in synaptic plasticity and modulation of glutamatergic excitatory transmission. Effect of NMDAR activation on inhibitory GABAergic transmission remains largely unknown. Here, we report that a brief application of NMDA could induce two distinct actions in CA1 pyramidal neurons in mouse hippocampal slices: 1) an inward current attributed to activation of postsynaptic NMDARs; and 2) fast phasic synaptic currents, namely spontaneous inhibitory postsynaptic currents (sIPSCs), mediated by GABA(A) receptors in pyramidal neurons. The mean amplitude of sIPSCs was also increased by NMDA. This profound increase in the sIPSC frequency and amplitude was markedly suppressed by the sodium channel blocker TTX, whereas the frequency and mean amplitude of miniature IPSCs were not significantly affected by NMDA, suggesting that NMDA elicits repetitive firing in GABAergic interneurons, thereby leading to GABA release from multiple synaptic sites of single GABAergic axons. We found that the NMDAR open-channel blocker MK-801 injected into recorded pyramidal neurons suppressed the NMDA-induced increase of sIPSCs, which raises the possibility that the firing of interneurons may not be the sole factor and certain retrograde messengers may also be involved in the NMDA-mediated enhancement of GABAergic transmission. Our results from pharmacological tests suggest that the nitric oxide signaling pathway is mobilized by NMDAR activation in CA1 pyramidal neurons, which in turn retrogradely facilitates GABA release from the presynaptic terminals. Thus NMDARs at glutamatergic synapses on both CA1 pyramidal neurons and interneurons appear to exert feedback and feedforward inhibition for determining the spike timing of the hippocampal microcircuit.


Subject(s)
Hippocampus/cytology , Presynaptic Terminals/physiology , Pyramidal Cells/cytology , Receptors, N-Methyl-D-Aspartate/metabolism , Synaptic Transmission/physiology , gamma-Aminobutyric Acid/metabolism , Agatoxins , Animals , Animals, Newborn , Calcium Channel Blockers/pharmacology , Cyclic N-Oxides/pharmacology , Drug Interactions , Enzyme Inhibitors/pharmacology , Excitatory Amino Acid Antagonists/pharmacology , Free Radical Scavengers/pharmacology , GABA Agents/pharmacology , Imidazoles/pharmacology , In Vitro Techniques , Inhibitory Postsynaptic Potentials/drug effects , Mice , Mice, Inbred C57BL , NG-Nitroarginine Methyl Ester/pharmacology , Neural Inhibition/drug effects , Neural Inhibition/physiology , Patch-Clamp Techniques , Presynaptic Terminals/drug effects , Spider Venoms/pharmacology , Synaptic Transmission/drug effects , Time Factors , omega-Conotoxin GVIA/pharmacology
18.
Mol Pain ; 6: 48, 2010 Aug 24.
Article in English | MEDLINE | ID: mdl-20735819

ABSTRACT

BACKGROUND: The R192Q mutation of the CACNA1A gene, encoding for the α1 subunit of voltage-gated P/Q Ca2+ channels (Ca(v)2.1), is associated with familial hemiplegic migraine-1. We investigated whether this gain-of-function mutation changed the structure and function of trigeminal neuron P2X3 receptors that are thought to be important contributors to migraine pain. RESULTS: Using in vitro trigeminal sensory neurons of a mouse genetic model knockin for the CACNA1A R192Q mutation, we performed patch clamp recording and intracellular Ca2+ imaging that showed how these knockin ganglion neurons generated P2X3 receptor-mediated responses significantly larger than wt neurons. These enhanced effects were reversed by the Ca(v)2.1 blocker ω-agatoxin. We, thus, explored intracellular signalling dependent on kinases and phosphatases to understand the molecular regulation of P2X3 receptors of knockin neurons. In such cells we observed strong activation of CaMKII reversed by ω-agatoxin treatment. The CaMKII inhibitor KN-93 blocked CaMKII phosphorylation and the hyperesponsive P2X3 phenotype. Although no significant difference in membrane expression of knockin receptors was found, serine phosphorylation of knockin P2X3 receptors was constitutively decreased and restored by KN-93. No change in threonine or tyrosine phosphorylation was detected. Finally, pharmacological inhibitors of the phosphatase calcineurin normalized the enhanced P2X3 receptor responses of knockin neurons and increased their serine phosphorylation. CONCLUSIONS: The present results suggest that the CACNA1A mutation conferred a novel molecular phenotype to P2X3 receptors of trigeminal ganglion neurons via CaMKII-dependent activation of calcineurin that selectively impaired the serine phosphorylation state of such receptors, thus potentiating their effects in transducing trigeminal nociception.


Subject(s)
Calcium Channels, P-Type/genetics , Calcium Channels, Q-Type/genetics , Ganglia, Sensory/metabolism , Ion Channel Gating/physiology , Migraine with Aura/genetics , Mutation/genetics , Pain/physiopathology , Receptors, Purinergic P2X3/metabolism , Adenosine Triphosphate/metabolism , Agatoxins , Amino Acid Substitution/genetics , Animals , Calcineurin/metabolism , Calcium Channels, N-Type , Calcium Channels, P-Type/metabolism , Calcium Channels, Q-Type/metabolism , Calcium Signaling/drug effects , Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Enzyme Activation/drug effects , Ganglia, Sensory/drug effects , Gene Knock-In Techniques , Intracellular Space/drug effects , Intracellular Space/metabolism , Ion Channel Gating/drug effects , Membrane Potentials/drug effects , Mice , Migraine with Aura/complications , Migraine with Aura/physiopathology , Mutant Proteins/metabolism , Pain/complications , Phosphorylation/drug effects , Phosphoserine/metabolism , Potassium/metabolism , Sensory Receptor Cells/drug effects , Sensory Receptor Cells/metabolism , Spider Venoms/pharmacology , Trigeminal Nerve/drug effects , Trigeminal Nerve/enzymology , Trigeminal Nerve/physiopathology
19.
Eur J Neurosci ; 32(4): 579-90, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20718855

ABSTRACT

Whole-cell patch-clamp recordings of non-N-methyl-d-aspartate glutamatergic excitatory postsynaptic currents (EPSCs) were carried out from cholinergic neurons in slices of basal forebrain (BF) of developing rats aged 21-42 postnatal days to elucidate postnatal developmental change in Ca(2+) channel subtypes involved in the transmission as well as that in dopamine D(1)-like receptor-mediated presynaptic inhibition. The amplitude of EPSCs was inhibited by bath application of omega-conotoxin GVIA (omega-CgTX; 3 microM) or omega-agatoxin-TK (omega-Aga-TK; 200 nM) throughout the age range examined, suggesting that multiple types of Ca(2+) channel are involved in the transmission. The EPSC fraction reduced by omega-CgTX decreased with age, whereas that reduced by omega-Aga-TK increased. Inhibition of the EPSCs by a D(1)-like receptor agonist, SKF 81297 (SKF; 30 microM) increased with age in parallel with the increase in omega-Aga-TK-induced inhibition. An activator of the adenylyl cyclase (AC) pathway, forskolin (FK; 10 microM) inhibited the EPSCs, and FK-induced inhibition also increased with age in parallel with the increase in SKF-induced inhibition. Throughout the age range examined, SKF showed no further inhibitory effect on the EPSCs after omega-Aga-TK- or FK-induced effect had reached steady-state. These findings suggest that D(1)-like receptor-mediated presynaptic inhibition of glutamate release onto cholinergic BF neurons increases with age, and that the change is coupled with a developmental increase in the contribution of P/Q-type Ca(2+) channels as well as a developmental increase in AC pathway contribution.


Subject(s)
Calcium Channels, P-Type/metabolism , Calcium Channels, Q-Type/metabolism , Glutamic Acid/metabolism , Prosencephalon/growth & development , Prosencephalon/metabolism , Receptors, Dopamine D1/metabolism , Synaptic Transmission/physiology , Agatoxins , Animals , Benzazepines/pharmacology , Dopamine/metabolism , Dopamine Agonists/pharmacology , Excitatory Postsynaptic Potentials/drug effects , Excitatory Postsynaptic Potentials/physiology , Imines/pharmacology , Patch-Clamp Techniques , Prosencephalon/drug effects , Rats , Spider Venoms/pharmacology , Synaptic Transmission/drug effects , omega-Conotoxin GVIA/pharmacology
20.
Nat Neurosci ; 13(5): 592-600, 2010 May.
Article in English | MEDLINE | ID: mdl-20348918

ABSTRACT

When chronic alterations in neuronal activity occur, network gain is maintained by global homeostatic scaling of synaptic strength, but the stability of microcircuits can be controlled by unique adaptations that differ from the global changes. It is not understood how specificity of synaptic tuning is achieved. We found that, although a large population of inhibitory synapses was homeostatically scaled down after chronic inactivity, decreased endocannabinoid tone specifically strengthened a subset of GABAergic synapses that express cannabinoid receptors. In rat hippocampal slice cultures, a 3-5-d blockade of neuronal firing facilitated uptake and degradation of anandamide. The consequent reduction in basal stimulation of cannabinoid receptors augmented GABA release probability, fostering rapid depression of synaptic inhibition and on-demand disinhibition. This regulatory mechanism, mediated by activity-dependent changes in tonic endocannabinoid level, permits selective local tuning of inhibitory synapses in hippocampal networks.


Subject(s)
Cannabinoid Receptor Modulators/metabolism , Endocannabinoids , Homeostasis/physiology , Neural Inhibition/physiology , Neurons/physiology , Synapses/physiology , Agatoxins , Animals , Arachidonic Acids/pharmacology , Benzamides/pharmacology , Benzoxazines/pharmacology , Biophysics , Calcium/metabolism , Calcium Channel Blockers/pharmacology , Cannabinoid Receptor Modulators/pharmacology , Carbamates/pharmacology , Conotoxins/pharmacology , Dose-Response Relationship, Drug , Down-Regulation/drug effects , Drug Interactions , Electric Stimulation/methods , Enzyme Inhibitors/pharmacology , Excitatory Amino Acid Antagonists/pharmacology , Glycerides/pharmacology , Hippocampus/cytology , Homeostasis/drug effects , In Vitro Techniques , Inhibitory Postsynaptic Potentials/drug effects , Inhibitory Postsynaptic Potentials/physiology , Morpholines/pharmacology , Naphthalenes/pharmacology , Nerve Net/physiology , Neural Inhibition/drug effects , Neurons/drug effects , Patch-Clamp Techniques/methods , Piperidines/pharmacology , Polyamines/pharmacology , Polyunsaturated Alkamides/pharmacology , Pyrazoles/pharmacology , Rats , Receptor, Cannabinoid, CB1/agonists , Receptor, Cannabinoid, CB1/antagonists & inhibitors , Receptor, Cannabinoid, CB1/metabolism , Rimonabant , Sodium Channel Blockers/pharmacology , Synapses/drug effects , Tetrodotoxin/pharmacology , gamma-Aminobutyric Acid/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...