Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 34
Filter
Add more filters










Publication year range
1.
Zygote ; 30(2): 213-216, 2022 Apr.
Article in English | MEDLINE | ID: mdl-34315559

ABSTRACT

In this work, the presence of calcium-dependent calcium channels and their receptors (RyR) has been investigated in Paracentrotus lividus eggs and early embryos, from unfertilized egg to four-blastomere stages. Electrophysiological recordings of RyR single-channel current fluctuations showed that RyRs are functional during the first developmental events with a maximum at zygote stage, c. 40 min after fertilization, corresponding to the first cleavage. The nature of vertebrate-like RyRs active at this stage was established by specific activation/blockade experiments.


Subject(s)
Paracentrotus , Zygote , Animals , Blastomeres/metabolism , Calcium/metabolism , Fertilization/physiology , Oocytes/metabolism , Ryanodine Receptor Calcium Release Channel/metabolism , Sea Urchins/metabolism , Zygote/metabolism
2.
Chem Biol Interact ; 157-158: 305-16, 2005 Dec 15.
Article in English | MEDLINE | ID: mdl-16257396

ABSTRACT

Organophosphate (OP) compounds exert inhibition on cholinesterase (ChE) activity by irreversibly binding to the catalytic site of the enzymes. For this reason, they are employed as insecticides for agricultural, gardening and indoor pest control. The biological function of the ChE enzymes is well known and has been studied since the beginning of the XXth century; in particular, acetylcholinesterase (AChE, E.C. 3.1.1.7) is an enzyme playing a key role in the modulation of neuromuscular impulse transmission. However, in the past decades, there has been increasing interest concerning its role in regulating non-neuromuscular cell-to-cell interactions mediated by electrical events, such as intracellular ion concentration changes, as the ones occurring during gamete interaction and embryonic development. An understanding of the mechanisms of the cholinergic regulation of these events can help us foresee the possible impact on environmental and human health, including gamete efficiency and possible teratogenic effects on different models, and help elucidate the extent to which OP exposure may affect human health. The chosen organophosphates were the ones mainly used in Europe: diazinon, chlorpyriphos, malathion, and phentoate, all of them belonging to the thionophosphate chemical class. This research has focused on the comparison between the effects of exposure on the developing embryos at different stages, identifying biomarkers and determining potential risk factors for sensitive subpopulations. The effects of OP oxonisation were not taken into account at this level, because embryonic responses were directly correlated to the changes of AChE activity, as determined by histochemical localisation and biochemical measurements. The identified biomarkers of effect for in vitro experiments were: cell proliferation/apoptosis as well as cell differentiation. For in vivo experiments, the endpoints were: developmental speed, size and shape of pre-gastrula embryos; developmental anomalies on neural tube, head, eye, heart. In all these events, we had evidence that the effects are mediated by ion channel activation, through the activation/inactivation of acetylcholine receptors (AChRs).


Subject(s)
Cholinesterases/metabolism , Embryonic Development/drug effects , Organophosphorus Compounds/pharmacology , Animals , Apoptosis/drug effects , Cell Differentiation/drug effects , Cell Proliferation/drug effects , Chick Embryo , Gene Expression Regulation, Developmental/drug effects , Time Factors
3.
Toxicon ; 40(5): p.557-62, 2002.
Article in English | Sec. Est. Saúde SP, SESSP-IBPROD, Sec. Est. Saúde SP | ID: but-ib8935
4.
Eur Biophys J ; 29(8): 569-73, 2001.
Article in English | MEDLINE | ID: mdl-11288831

ABSTRACT

Native noxiustoxin (NTX) and synthetic peptides corresponding to its primary sequence, from positions 1-9, 1-14, 1-20, 10-20, 21-39 and 30 39, were prepared and assayed on the K+ currents of cerebellum granular cells, using the patch-clamp technique in the whole-cell configuration system. Native toxin has a reversible inhibitory effect (IC50 = 360 nM), whereas synthetic peptides NTXI-20 and NTX1-9 had a half-effective dose IC50 of approximately 2 and 10 microM, respectively, which correlates with their biological effects in vivo. Synthetic peptide NTX10-20 was quite remarkable in having a preference for the IA current, which was completely inhibited at high peptide concentration. The effects of the other peptides (NTXI 14, NTX21-39 and NTX30-39), although positive and reversible, required higher concentrations (50 200 microM) to block both currents, suggesting no affinity or, at least, much lower specificity for the channels responsible for the potassium currents in the granular cells studied.


Subject(s)
Cerebellum/cytology , Cerebellum/drug effects , Peptides/pharmacology , Potassium Channel Blockers , Potassium/metabolism , Scorpion Venoms/chemistry , Scorpion Venoms/pharmacology , Amino Acid Sequence , Animals , Cells, Cultured , Dose-Response Relationship, Drug , Electrophysiology , Inhibitory Concentration 50 , Molecular Sequence Data , Patch-Clamp Techniques , Peptide Biosynthesis , Rats , Rats, Wistar , Sequence Homology, Amino Acid
5.
Biochim Biophys Acta ; 1468(1-2): 203-12, 2000 Sep 29.
Article in English | MEDLINE | ID: mdl-11018665

ABSTRACT

A novel peptide was purified from the venom of the scorpion Androctonus australis Garzoni (abbreviated Aa1, corresponding to the systematic number alpha KTX4.4). It contains 37 amino acid residues, has a molecular mass of 3850 Da, is closely packed by three disulfide bridges and a blocked N-terminal amino acid. This peptide selectively affects the K(+) currents recorded from cerebellum granular cells. Only the fast activating and inactivating current, with a kinetics similar to I(A)-type current, is completely blocked by the addition of low micromolar concentrations (K(i) value of 150 nM) of peptide Aa1 to the external side of the cell preparation. The blockade is partially reversible in our experimental conditions. Aa1 blocks the channels in both the open and the closed states. The blockage is test potential independent and is not affected by changes in the holding potential. The kinetics of the current are not affected by the addition of Aa1 to the preparation; it means that the block is a simple 'plugging mechanism', in which a single toxin molecule finds a specific receptor site in the external vestibule of the K(+) channel and thereby occludes the outer entry to the K(+) conducting pore.


Subject(s)
Cerebellum/metabolism , Neurotoxins/chemistry , Peptides/pharmacology , Potassium Channel Blockers , Scorpion Venoms/chemistry , Amino Acid Sequence , Animals , Cells, Cultured , Kinetics , Molecular Sequence Data , Patch-Clamp Techniques , Peptides/isolation & purification , Potassium Channels/metabolism , Rats , Rats, Wistar , Serine Endopeptidases
6.
Br J Pharmacol ; 131(2): 335-41, 2000 Sep.
Article in English | MEDLINE | ID: mdl-10991928

ABSTRACT

1. Haloperidol is a drug used in the management of several psychotic disorders and its use has been linked to Neuroleptic Malignant Syndrome. In the present study we have investigated the effect of a commercial preparation of haloperidol, Serenase, on skeletal muscle sarcoplasmic reticulum. 2. Addition of Serenase to isolated terminal cisternae caused a rapid release of calcium. We tested whether the active Ca(2+)-releasing substance was haloperidol or another compound present in the preparation. 3. Our results show that methyl p-hydroxybenzoate, one of the preservatives and a commonly used anti-microbial agent (E-218) is an activator of Ca(2+) release (E.C. 50=2.0 mM), mediated by a ruthenium red-sensitive Ca(2+) release channel present in skeletal muscle terminal cisternae.


Subject(s)
Food Preservatives/pharmacology , Parabens/pharmacology , Ryanodine Receptor Calcium Release Channel/drug effects , Sarcoplasmic Reticulum/drug effects , Animals , Antipsychotic Agents/adverse effects , Antipsychotic Agents/pharmacology , Calcium/metabolism , Dose-Response Relationship, Drug , Haloperidol/adverse effects , Haloperidol/pharmacology , In Vitro Techniques , Neuroleptic Malignant Syndrome/etiology , Rabbits , Ryanodine Receptor Calcium Release Channel/metabolism , Sarcoplasmic Reticulum/metabolism
7.
J Biol Chem ; 274(18): 12278-83, 1999 Apr 30.
Article in English | MEDLINE | ID: mdl-10212196

ABSTRACT

Triadin has been shown to co-localize with the ryanodine receptor in the sarcoplasmic reticulum membrane. We show that immunoprecipitation of solubilized sarcoplasmic reticulum membrane with antibodies directed against triadin or ryanodine receptor, leads to the co-immunoprecipitation of ryanodine receptor and triadin. We then investigated the functional importance of the cytoplasmic domain of triadin (residues 1-47) in the control of Ca2+ release from sarcoplasmic reticulum. We show that antibodies directed against a synthetic peptide encompassing residues 2-17, induce a decrease in the rate of Ca2+ release from sarcoplasmic reticulum vesicles as well as a decrease in the open probability of the ryanodine receptor Ca2+ channel incorporated in lipid bilayers. Using surface plasmon resonance spectroscopy, we defined a discrete domain (residues 18-46) of the cytoplasmic part of triadin interacting with the purified ryanodine receptor. This interaction is optimal at low Ca2+ concentration (up to pCa 5) and inhibited by increasing calcium concentration (IC50 of 300 microM). The direct molecular interaction of this triadin domain with the ryanodine receptor was confirmed by overlay assay and shown to induce the inhibition of the Ca2+ channel activity of purified RyR in bilayer. We propose that this interaction plays a critical role in the control, by triadin, of the Ca2+ channel behavior of the ryanodine receptor and therefore may represent an important step in the regulation process of excitation-contraction coupling in skeletal muscle.


Subject(s)
Carrier Proteins , Cytoplasm/metabolism , Muscle Proteins/metabolism , Muscle, Skeletal/metabolism , Ryanodine Receptor Calcium Release Channel/metabolism , Animals , Antibodies/immunology , Muscle Proteins/chemistry , Muscle Proteins/immunology , Precipitin Tests , Protein Binding , Rabbits , Ryanodine Receptor Calcium Release Channel/immunology
8.
Brain Res ; 815(2): 410-3, 1999 Jan 09.
Article in English | MEDLINE | ID: mdl-9878856

ABSTRACT

Membrane vesicles from rat cerebellum were reconstituted into lipid bilayers. The activity of two different potassium channels was recorded: (1) a small conducting voltage dependent potassium channel insensitive to [Ca2+]i, (2) a calcium and voltage dependent potassium channel (KCa). KCa channels had a conductance of (302+/-15) pS (n=5) and were activated by [Ca2+]i and membrane depolarizations. They were blocked by tetraethylamonium (TEA) and charybdotoxin (CTX) but insensitive to noxiustoxin (NTX). Finally, we showed the blocking effect of Androctonus australis Hector (AaH) scorpion venom on KCa channels from rat cerebellum.


Subject(s)
Calcium/physiology , Cerebellum/metabolism , Ion Channel Gating , Potassium Channels/metabolism , Animals , Cerebellum/drug effects , Charybdotoxin/pharmacology , Ion Channel Gating/drug effects , Lipid Bilayers/metabolism , Membrane Potentials/drug effects , Potassium Channels/isolation & purification , Rats , Scorpion Venoms/pharmacology , Tetraethylammonium/pharmacology
9.
Biochem Biophys Res Commun ; 242(2): 287-91, 1998 Jan 14.
Article in English | MEDLINE | ID: mdl-9446786

ABSTRACT

The Shaker B potassium channel expressed in Xenopus laevis oocytes is blocked, in a total reversible manner from the outside part, by a new toxin (Aa1) composed of 40 amino acid residues, purified from the venom of the North African scorpion Androctonus australis Garzoni. The experiments were performed with patch-clamp technique in the outside-out configuration. The half blocking concentration is approximately 4.5 microM with a 1:1 stoichiometry. The activation and inactivation kinetics of the current are not modified by the blocking mechanism. The binding affinity is not voltage dependent. These results suggest a simple bimolecular mechanism of blockade by which the toxin occludes the external vestibule of the channel and thereby inhibits the K+ ions conduction.


Subject(s)
Potassium Channels/drug effects , Scorpion Venoms/isolation & purification , Toxins, Biological/isolation & purification , Animals , Electrophysiology , Gene Expression/genetics , Kinetics , Membrane Potentials/drug effects , Oocytes/metabolism , Patch-Clamp Techniques , Scorpion Venoms/pharmacology , Shaker Superfamily of Potassium Channels , Toxins, Biological/pharmacology , Xenopus laevis
10.
Eur Biophys J ; 27(1): 69-73, 1998.
Article in English | MEDLINE | ID: mdl-9463891

ABSTRACT

A purified peptide from Androctonus australis Garzoni venom (AaG) affects selectively a K(+)-current recorded from cerebellum granular cells. This current is characterized by fast activating and inactivating kinetics similar to an IA-type current. Addition of 2 microM peptide Aa1 (from Androctonus australis, toxin 1) to the external side of the channel suppressed completely and in a selective manner the IA-type current, with an IC50 value of 130 nM, whereas in the same conditions, the other potassium current, identified as delayed rectifier (Id), was not affected. Additionally, we show that another partially purified peptide (III-12) from the same venom was able to block reversibly both K(+)-currents.


Subject(s)
Cerebellum/drug effects , Potassium Channels/drug effects , Scorpion Venoms/chemistry , Scorpion Venoms/toxicity , Toxins, Biological/isolation & purification , Toxins, Biological/toxicity , Animals , Cells, Cultured , Cerebellum/physiology , Cytoplasmic Granules/drug effects , Cytoplasmic Granules/physiology , Electrophysiology , Potassium/physiology , Potassium Channels/physiology , Rats , Rats, Wistar , Scorpions
11.
Biochemistry ; 36(38): 11496-503, 1997 Sep 23.
Article in English | MEDLINE | ID: mdl-9298970

ABSTRACT

In the present report we studied the interaction between the skeletal muscle ryanodine receptor and the ubiquitous S100A1 Ca2+ binding protein. S100A1 did not affect equilibrium [3H]ryanodine binding to purified rabbit skeletal muscle terminal cisternae at 100 microM free [Ca2+]. At nanomolar free [Ca2+], however, S100A1 activated by 40 +/- 6.7% (mean +/- SE, n = 5) the [3H]ryanodine binding activity; the half-maximal concentration for stimulation of [3H]ryanodine binding was approximately 70 nM, a value well below the estimated S100A1 concentration in skeletal muscle fibers. Scatchard analysis of [3H]ryanodine binding performed in the presence of 100 microM EGTA indicates that S100A1 increases the apparent affinity of the receptor for ryanodine (Kd = 191 vs 383 nM in the presence and in the absence of 100 nM S100A1, respectively). The effect of S100A1 was also tested on the single-channel gating properties of the purified ryanodine receptor after reconstitution into a lipid planar bilayer. Currents carried by purified ryanodine receptor channels were modulated by both cis Ca2+ and ruthenium red. In the presence of nanomolar [Ca2+], S100A1 activated the channel by increasing (6.0 +/- 2.8)-fold (mean +/- SE, n = 3) the normalized open probability. The interaction between S100A1 and the purified RYR was verified using the optical biosensor BIAcore: we show that the two proteins interact directly both at millimolar and at nanomolar calcium concentrations. We next mapped the regions of the skeletal muscle RYR involved in the interaction with S100A1 by performing ligand overlays on a panel RYR of fusion proteins in the presence of 100 nM S100A1. Our results indicate that the skeletal muscle RYR contains three potential S100A1 binding domains. Binding of S100A1 to the RYR fusion proteins occurred at both nanomolar and millimolar free [Ca2+]. S100A1 binding domain 1 binds the ligand in the presence of 1 mM free [Ca2+] or 1 mM EGTA. Maximal binding to S100A1#2 was achieved in the presence of 1 mM free [Ca2+]. The S100A1#3 domain, which overlaps with calcium-dependent calmodulin binding domain 3 (CaM 3), exhibits weak and strong S100A1 binding activity in the presence of either millimolar or nanomolar Ca2+, respectively. The interaction between S100A1 and the purified RYR complex was also investigated by affinity chromatography: in the presence of nanomolar Ca2+, we observed binding of native RYR complex to S100A1-conjugated Sepharose. This interaction could be inhibited by the presence of RYR polypeptides encompassing S100A1 binding sites S100A1#1, S100A1#2, and S100A1#3.


Subject(s)
Biomarkers , Calcium Channels/metabolism , Calcium/metabolism , Muscle Proteins/metabolism , Ryanodine/metabolism , S100 Proteins/metabolism , Sarcoplasmic Reticulum/metabolism , Animals , Binding Sites , Biosensing Techniques , Calcium Channels/genetics , Cattle , Electric Conductivity , Ion Channel Gating , Lipid Bilayers , Male , Muscle Proteins/genetics , Peptide Fragments/genetics , Peptide Fragments/metabolism , Protein Binding , Rabbits , Recombinant Fusion Proteins/metabolism , Ryanodine Receptor Calcium Release Channel
12.
Exp Brain Res ; 110(1): 15-20, 1996 Jun.
Article in English | MEDLINE | ID: mdl-8817251

ABSTRACT

Helothermine (HLTx), a 25.5-kDa peptide toxin isolated from the venom of the Mexican beaded lizard (Heloderma horridum horridum), was found to be an inhibitor of Ca2+ channels in cerebellar granule cells of newborn rats. Macroscopic currents, carried by 10 mM Ba2+, were measured in whole-cell configuration. The toxin at the saturating dose of 2.5 microM reversibly produced an approximately 67% block of the voltage-dependent Ca2+ current by a fast mechanism of action. The current inhibition and recovery were reached in less than 1 min. Inhibition was concentration-dependent, with a half-effective dose of 0.25 microM. The current block was practically voltage-independent, whereas the steady-state inactivation h infinity was significantly affected by HLTx (approximately 10 mV). The toxin did not affect the activation and inactivation kinetics of the Ca2+ current. Experiments with other Ca2+ channel blockers showed that HLTx abolished omega-cono-toxin GVIA-sensitive Ca2+ currents, as well as omega-Aga-IVA- and dihydropyridine-sensitive Ca2+ currents. These drugs had virtually no effect when HLTx was applied first. The present results indicate that HLTx produce a high-potency blockage of the three pharmacologically distinct Ca2+ current components.


Subject(s)
Calcium Channel Blockers/toxicity , Cerebellum/drug effects , Lizards , Neurotoxins/toxicity , Peptides/toxicity , Venoms/chemistry , Animals , Cerebellum/cytology , Dihydropyridines/pharmacology , Mollusk Venoms/toxicity , Neurons/drug effects , Spider Venoms/toxicity , omega-Agatoxin IVA , omega-Conotoxin GVIA
13.
Comp Biochem Physiol A Physiol ; 112(1): 21-8, 1995 Sep.
Article in English | MEDLINE | ID: mdl-7553331

ABSTRACT

A novel toxin was isolated from the venom of the Brazilian scorpion Tityus (T.) bahiensis. The N-terminal amino acid sequence of this toxin was shown to be 80% identical to the corresponding segment of T. serrulatus toxin IV-5. The new toxin was thus named toxin IV-5b. Toxin IV-5b was found to markedly slow inactivation of Na channel in dorsal root ganglion neurons from chick embryo. By contrast, Na channel activation was only negligibly delayed, and deactivation completely unaffected. Similarly unaffected by the toxin were K and Ca currents. The slowing effect of the toxin starts to appear at concentrations of c. 80 nM, and shows a KD of 143 nM. With a toxin concentration of 2.4 microM, the Na channel inactivation time constant was increased c. 3-fold with respect to the control. The slowing of inactivation was voltage dependent, and increased with depolarization.


Subject(s)
Ganglia, Spinal/metabolism , Neurons/metabolism , Scorpion Venoms/pharmacology , Sodium Channels/drug effects , Animals , Chick Embryo , Dose-Response Relationship, Drug , Electrophysiology , Ganglia, Spinal/cytology , Sodium Channels/physiology
14.
Glia ; 15(1): 33-42, 1995 Sep.
Article in English | MEDLINE | ID: mdl-8847099

ABSTRACT

An affinity column prepared with noxiustoxin (NTx), a K+ channel blocker from the venom of the Mexican scorpion Centruroides noxius, was used to purify a functional channel from a detergent extract of Schwann cell membrane of the giant axon of the squid Loligo vulgaris. The purified protein was reconstituted as a functional unit in a planar lipid bilayer and tested with a sequence of potentials to obtain information about single-channel amplitude and kinetics. The reconstituted channel showed delayed rectifier behavior with a slope conductance of 10 pS under 5:1 asymmetric KCl concentrations and a clear tendency to open under negative potentials. The zero-current potential was +36mV, which fitted well with the Nernst equation for the CIS/TRANS K(+)-concentration ratio of 5:1. The channel also showed a strong sensitivity to tetraethylammonium and its activity was inhibited by NTx, as expected from the purification procedure. The behavior of this protein in the presence of 0.5 mM ATP (cis side) was also tested, significantly increasing current fluctuations across the membrane. In order to compare the modulation of the Schwann cell K+ channel with that of the axonal K+ channel, a purified protein from the squid axon membrane was also tested in the presence of ATP. This 10-11 pS, delayed rectifier channel from the squid giant axon (Prestipino et al., FEBS Lett. 250:570-574, 1989) was also tested in the presence of ATP and showed a similar rise in activity.


Subject(s)
Axons/metabolism , Lipid Bilayers/metabolism , Potassium Channels/metabolism , Schwann Cells/metabolism , Animals , Axons/chemistry , Decapodiformes , Electrophysiology , Ion Channel Gating/physiology , Kinetics , Lipid Bilayers/chemistry , Membrane Potentials/physiology , Phosphorylation , Potassium Channels/chemistry , Protein Binding/physiology , Scorpion Venoms/pharmacology
15.
Biochem Biophys Res Commun ; 213(3): 1082-90, 1995 Aug 24.
Article in English | MEDLINE | ID: mdl-7544580

ABSTRACT

In this study terminal cisternae vesicles from rabbit skeletal muscle were fused into planar bilayers and the effect of calmodulin on single Ca2+ release channel currents was investigated. In the presence of 10(-7) and 10(-9) M free [Ca2+], nanomolar concentrations of calmodulin activated the channel by increasing the open probability of single-channel events in a dose dependent manner. The activatory effect of calmodulin was reversed by 10 microM ruthenium red. At 10(-5) M free [Ca2+], calmodulin (0.1-1 microM) inhibited channel activity. Calmodulin overlays were carried out using concentrations of Ca2+ similar to those used for the planar lipid bilayer assays. In the presence of 10(-7) M [Ca2+], calmodulin bound to the ryanodine receptor, to a region defined by residues 2937-3225 and 3546-3655. These results suggest that calmodulin may activate the Ca(2+)-release channel (ryanodine-receptor) by interacting with binding sites localized in the central portion of the RYR protomer.


Subject(s)
Calcium Channels/metabolism , Calcium/metabolism , Calmodulin/pharmacology , Muscle Proteins/metabolism , Muscle, Skeletal/drug effects , Animals , Calmodulin-Binding Proteins/metabolism , Lipid Bilayers , Muscle, Skeletal/metabolism , Rabbits , Ruthenium Red/pharmacology , Ryanodine Receptor Calcium Release Channel
16.
Toxicon ; 32(12): 1487-93, 1994 Dec.
Article in English | MEDLINE | ID: mdl-7725317

ABSTRACT

A novel toxin was isolated and characterized from the venom of the Mexican scorpion Centruroides infamatus infamatus. It has an apparent mol. wt of 7600, compatible with the presence of 66 amino acid residues per molecule. The N-terminal amino acid sequence was determined (up to residue 48) and showed approximately 95% similarity with toxins from other Mexican scorpions of the gnus Centruroides. Experiments conducted with chick dorsal root ganglion cells showed that toxin 1 is a Na+ channel effector, causing a decrease in the peak Na+ permeability, similar to decreases observed for typical beta-scorpion toxins.


Subject(s)
Ganglia, Spinal/drug effects , Scorpion Venoms/metabolism , Sodium/metabolism , Toxins, Biological/isolation & purification , Amino Acid Sequence , Amino Acids/metabolism , Animals , Cell Membrane Permeability/drug effects , Chemical Fractionation , Chick Embryo , Chromatography, Gel , Electrophysiology , Freeze Drying , Ganglia, Spinal/cytology , Ganglia, Spinal/metabolism , Injections, Intraperitoneal , Mice , Molecular Sequence Data , Molecular Weight , Neurons/drug effects , Scorpion Venoms/chemistry , Scorpions , Sequence Homology, Amino Acid , Toxins, Biological/administration & dosage , Toxins, Biological/chemistry , Toxins, Biological/toxicity
17.
Biochem J ; 304 ( Pt 1): 51-6, 1994 Nov 15.
Article in English | MEDLINE | ID: mdl-7998956

ABSTRACT

Two novel toxins were purified from the venom of the Mexican scorpion Centruroides limpidus limpidus, using an immunoassay based on antibodies raised against noxiustoxin (NTX), a known K(+)-channel-blocker-peptide. The primary structure of C. l. limpidus toxin 1 was obtained by Edman degradation and was shown to be composed of 38 amino acid residues, containing six half-cystines. The first 36 residues of C. l. limpidus toxin 2 were also determined. Both toxins are capable of displacing the binding of radio-labelled NTX to rat brain synaptosomes with high affinity (about 100 pM). These toxins are capable of inhibiting transient K(+)-currents (resembling IA-type currents), in cultured rat cerebellar granule cells. About 50% of the peak currents are reduced by application of a 1.5 microM solution of toxins 1 and 2 The K+ current reduction is partially reversible, under washing but not voltage-dependent. Comparison of the primary structure of C. l. limpidus toxin 1 with other known toxins shows 74% identity with margatoxin, 64% with NTX, 51% with kaliotoxin, 39% with iberiotoxin, 37% with charybdotoxin and Lq2, and 29% with leirutoxin 1. The only invariant amino acids in all these toxins are the six cysteines, a glycine in position 26 and two lysines at positions 28 and 33, respectively. The relevance of these differences in terms of possible structure-function relationships is discussed.


Subject(s)
Potassium Channel Blockers , Scorpion Venoms/chemistry , Toxins, Biological/isolation & purification , Amino Acid Sequence , Animals , Antibodies, Monoclonal , Cross Reactions , Molecular Sequence Data , Scorpion Venoms/immunology , Scorpion Venoms/pharmacology , Sequence Alignment , Structure-Activity Relationship , Toxins, Biological/immunology
18.
Biochem Biophys Res Commun ; 201(1): 186-93, 1994 May 30.
Article in English | MEDLINE | ID: mdl-7515231

ABSTRACT

The analysis of the hydropathy profile power spectra provides a basis for studies of pattern matching between the primary and secondary structure of peptides. The structural motif obtained with Noxiustoxin (NTX), the first K+ channel blocking peptide described, is composed of a N-terminal beta-strand, a central alpha-helix and a final beta-strand zone, probably forming a beta-sheet. These results were compared with those of Charybdotoxin (ChTX), a potent inhibitor of the high conductance Ca(2+)-activated K+ channel, which presents about 48% similarity with NTX in the amino acid sequence. Our prediction for ChTX secondary structure, which is known by 2D-NMR spectroscopy, yielded a Chou-Fasman quality index Q = 90%. The comparison between the two toxins has guided the interpretation of the data obtained.


Subject(s)
Scorpion Venoms/chemistry , Amino Acid Sequence , Charybdotoxin , Molecular Sequence Data , Potassium Channel Blockers , Protein Structure, Secondary , Sequence Alignment , Sequence Homology, Amino Acid , Solubility
19.
J Membr Biol ; 139(1): 49-55, 1994 Apr.
Article in English | MEDLINE | ID: mdl-8071987

ABSTRACT

Helothermine, a recently isolated toxin from the venom of the Mexican beaded lizard Heloderma horridum horridum was tested on K+ currents of newborn rat cerebellar granule cells. In whole-cell voltage-clamp experiments, cerebellar granule neurons exhibited at least two different K+ current components: a first transient component which is similar to an IA-type current, is characterized by fast activating and inactivating kinetics and blocked by 4-aminopyridine; a second component which is characterized by noninactivating kinetics, is blocked by tetraetylammonium ions and resembles the classical delayed-rectifier current. When added to the standard external solution at concentrations ranging between 0.1 and 2 microM, helothermine reduced the pharmacologically isolated IA-type current component in a voltage- and dose-dependent way, with a half-maximal inhibitory concentration (IC50) of 0.52 microM. A comparison between control and helothermine-modified peak transient currents shows a slowdown of activation and inactivation kinetics. The delayed-rectifier component inhibition was concentration dependent (IC50 = 0.86 microM) but not voltage dependent. No frequency- or use-dependent block was observed on both K+ current types. Perfusing the cells with control solution resulted in quite a complete current recovery. We conclude that helothermine acts with different affinities on two types of K+ current present in central nervous system neurons.


Subject(s)
Cerebellum/cytology , Peptides/pharmacology , Potassium Channels/physiology , Venoms/pharmacology , 4-Aminopyridine/pharmacology , Animals , Cell Membrane/physiology , Cell Membrane/ultrastructure , Cerebellum/physiology , Cerebellum/ultrastructure , Dose-Response Relationship, Drug , Lizards , Neurons/physiology , Neurons/ultrastructure , Potassium Channels/drug effects , Rats , Rats, Wistar , Tetraethylammonium Compounds/pharmacology
20.
Anal Biochem ; 210(1): 119-22, 1993 Apr.
Article in English | MEDLINE | ID: mdl-8489006

ABSTRACT

We have investigated the ability of the neutral ionophore ETH 129 to translocate Ca2+ across artificial and biological membranes. ETH 129 induces Ca2+ transport across planar lipid bilayer. The zero-current membrane potential in a gradient of Ca2+ concentration exhibits Nernst behavior. The dependence of the membrane conductance on ionophore and Ca2+ concentration indicates that three ionophore molecules are needed to transfer one Ca2+ across the hydrophobic region of the membrane. In mitochondria the neutral Ca2+ ionophore can move Ca2+ inside in response to a negative membrane potential under conditions in which the endogenous uniporter is blocked by ruthenium red. This electrophoretic transport of Ca2+ by ETH 129 occurs at a concentration much lower than the one previously reported with the neutral Ca2+ ionophore ETH 1001. Using sea urchin eggs, we have also shown that the efficiency of ETH 129 in inducing egg activation, as revealed by cortical granules exocytosis, is four orders of magnitude higher than that of the commonly used Ca2+ ionophore A21387. ETH 129 is a very efficient and useful tool for use in the investigation of Ca(2+)-dependent biological processes.


Subject(s)
Calcium/metabolism , Cyclohexanes/pharmacology , Ionophores/pharmacology , Animals , Cell Membrane/drug effects , Cell Membrane/metabolism , Exocytosis/drug effects , Female , In Vitro Techniques , Intracellular Membranes/drug effects , Intracellular Membranes/metabolism , Ion Transport/drug effects , Lipid Bilayers , Membranes, Artificial , Mitochondria, Liver/drug effects , Mitochondria, Liver/metabolism , Ovum/drug effects , Ovum/metabolism , Rats , Sea Urchins
SELECTION OF CITATIONS
SEARCH DETAIL
...