Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 75
Filter
Add more filters










Publication year range
1.
Toxicon ; 60(8): 1415-27, 2012 Dec 15.
Article in English | MEDLINE | ID: mdl-23085191

ABSTRACT

Anti-insect depressant toxins represent a subfamily of scorpion venom-derived ß-toxins that are polypeptides composed of 61-65 amino acid residues stabilized by four disulfide bridges. These toxins affect the activation of voltage-sensitive sodium channels (NaScTx) and exhibit the preferential ability to induce flaccid paralysis in insect larvae. Here we demonstrate the recombinant expression of the novel cardiac inotropic peptide (Bj-IP) that was classified as an anti-insect depressant ßNaScTx isolated from the venom of Hottentotta judaicus. By using "splicing by overlap extension" (SOE)-PCR, allowing for the first time one step de novo synthesis of long-chain scorpion toxin genes, we generated a codon-optimized DNA fragment of Bj-IP for cloning into the Escherichia coli vector pQE30. Moreover, the gene of interest was fused to a 6xHis coding DNA sequence. Subsequent recombinant expression was performed in E. coli KRX. The purification of the polypeptide was achieved by a combination of NiNTA agarose columns and RP (C(18)) high-performance liquid chromatography. The purified fusion protein was digested with factor Xa resulting in the elution of Bj-IP. The yield of recombinant Bj-IP expression was approximately 4.5 mg per liter of culture. Mass spectrometry confirmed the theoretical total mass of Bj-IP (6608 Da). Tag-free Bj-IP was refolded in guanidine chloride buffer with a glutathione redox system which was supplemented with different additives at 16 °C. Supplementation with 10% glycerol produced Bj-IP folding forms that exhibited reproducible biological activity in mouse cardiomyocytes. Cell contractility was increased by almost 3-fold and decay kinetics were hasten by 47% after administration of Bj-IP. Taken together, here we show the recombinant expression of the functionally active cardiac inotropic peptide Bj-IP, a new ßNaScTx from H. judaicus, for promising pharmacological applications. Furthermore, our data suggest that the use of SOE-PCR may help to facilitate in future the high throughput of cloning and/or modification of scorpion toxin genes.


Subject(s)
Peptides/genetics , Peptides/toxicity , Scorpion Venoms/chemistry , Scorpions , Animals , Base Sequence , Blotting, Western , Chromatography, Affinity , Chromatography, High Pressure Liquid , Cloning, Molecular , DNA , DNA Primers , Electrophoresis, Polyacrylamide Gel , Escherichia coli/genetics , Mass Spectrometry , Mutagenesis , Polymerase Chain Reaction , Sequence Homology, Nucleic Acid
2.
Cell Mol Biol Lett ; 6(4): 971-84, 2001.
Article in English | MEDLINE | ID: mdl-11753441

ABSTRACT

Pahutoxin a quaternary ammonium salt surfactant serves as an active ingredient in the defensive skin secretion of various marine trunkfish (Ostracociidae). In the defensive skin secretion of the Red Sea trunkfish, Ostracion cubicus the effect of PHN is amplified due to the existence of non toxic polypeptides which act as (a) PHN - chelators and (b) potentiators. The secretion of the Red Sea trunkfish includes an additional category of pharmacologically active polypeptides represented by boxin [7] which similarly to PHN they independently kill fish exclusively through medium application. By the aid of radiolabeled PHN and a fish gill membrane preparation a series of equilibrium saturation binding assays were carry out which demonstrate that PHN performs its biological defensive function via receptors and not due to its surface activity. The gill membranes of the trunkfish were shown to be devoid of PHNreceptors. The pharmacological, ecological and environmental implications of the above data are discussed.


Subject(s)
Choline/analogs & derivatives , Choline/metabolism , Marine Toxins/metabolism , Tetraodontiformes/metabolism , Animals , Choline/isolation & purification , Choline/toxicity , Gills/metabolism , In Vitro Techniques , Kinetics , Marine Toxins/isolation & purification , Marine Toxins/toxicity , Proteins/isolation & purification , Proteins/metabolism , Sea Bream/metabolism , Skin/metabolism , Surface-Active Agents/isolation & purification , Surface-Active Agents/metabolism , Surface-Active Agents/toxicity
3.
J Exp Biol ; 204(Pt 15): 2637-45, 2001 Aug.
Article in English | MEDLINE | ID: mdl-11533113

ABSTRACT

AaIT, an insect-selective neurotoxic polypeptide derived from scorpion venom, has recently been used to engineer recombinant baculoviruses for insect pest control. Lepidopterous larvae infected with an AaIT-expressing baculovirus reveal symptoms of paralysis identical to those induced by injection of the native toxin. However, the paralyzed larvae treated by the recombinant virus possess an approximately 50-fold lower hemolymph toxin concentration than insects paralyzed by the native toxin. The mechanism of this potentiation effect was studied using immunocytochemistry, electrophysiology and toxicity assays. (i) Light microscopy, using peroxidase-conjugated antibodies, revealed the presence of toxin in virus-susceptible tissues, including tracheal epithelia located close to the central nervous system and beyond its lamellar enveloping sheath. (ii) High-resolution immunogold electron microscopical cytochemistry clearly revealed the presence of recombinant AaIT toxin inside the thoracic and abdominal ganglia on neuronal cell bodies and axonal membranes. (iii) Ventral nerve cords dissected from silkworm larvae infected with the recombinant baculovirus exhibited a high degree of excitability, expressed as enhanced frequency and bursting mode of their spontaneous activity, when compared to nerve cords infected with the wild-type virus. We conclude that the recombinant-virus-infected tracheal epithelia, outbranching in the body of an infected insect, (i) locally supply a continuous, freshly produced toxin to its neuronal receptors and (ii) introduce the expressed toxin to the insect central nervous system, thus providing it with critical target sites that are inaccessible to the native toxin.


Subject(s)
Baculoviridae/genetics , Gene Expression , Gene Targeting , Lepidoptera/genetics , Scorpion Venoms/genetics , Animals , Axons/chemistry , Epithelium/ultrastructure , Ganglia, Invertebrate/chemistry , Genetic Vectors , Insect Control , Larva/genetics , Lepidoptera/growth & development , Microscopy, Immunoelectron , Neurons/chemistry , Recombinant Proteins/analysis , Scorpion Venoms/analysis , Scorpion Venoms/chemistry , Trachea/ultrastructure , Transfection
4.
Biochimie ; 82(9-10): 869-81, 2000.
Article in English | MEDLINE | ID: mdl-11086217

ABSTRACT

AaIT is a single chain neurotoxic polypeptide derived from the venom of the Buthid scorpion Androctonus australis Hector, composed of 70 amino acids cross-linked by four disulfide bridges. Its strict selectivity for insects has been documented by toxicity, electrophysiological and ligand receptor binding assays. These last have shown that various insect neuronal membranes possess a single class of non-interacting AaIT binding sites of high affinity (K(D) = 1-3(n)M) and low capacity (0.5-2.0 pmol/mg prot.). The fast excitatory paralysis induced by AaIT is a result of a presynaptic effect, namely the induction of a repetitive firing in the terminal branches of the insect's motor nerves resulting in a massive and uncoordinated stimulation of the respective skeletal muscles. The neuronal repetitive activity is attributed to an exclusive and specific perturbation of sodium conductance as a consequence of toxin binding to external loops of the insect voltage-dependent sodium channel and modification of its gating mechanism. From a strictly agrotechnical point of view AaIT involvement in plant protection has taken the following two complementary forms: firstly, as a factor for the genetic engineering of insect infective baculoviruses resulting in potent and selective bio-insecticides. The efficacy of the AaIT-expressing, recombinant baculovirus is attributed mainly to its ability to continuously provide and translocate the gene of the expressed toxin to the insect central nervous system; secondly, based on the pharmacological flexibility of the voltage-gated sodium channel, as a device for insecticide resistance management. Channel mutations conferring resistance to a given class of insecticidal agents (such as the KDR phenomenon) may greatly increase susceptibility to the AaIT expressing bioinsecticides. Thus the AaIT is a pharmacological tool for the study of insect neuronal excitability and chemical ecology and the development of new approaches to insect control.


Subject(s)
Insecticides/pharmacology , Neurotoxins/pharmacology , Scorpion Venoms/pharmacology , Amino Acid Sequence , Animals , Binding Sites , Insecticide Resistance , Insecticides/chemistry , Insecticides/metabolism , Ion Channel Gating , Molecular Sequence Data , Neurotoxins/chemistry , Neurotoxins/metabolism , Scorpion Venoms/chemistry , Scorpion Venoms/metabolism , Sodium Channels/drug effects
5.
Insect Biochem Mol Biol ; 29(10): 849-53, 1999 Oct.
Article in English | MEDLINE | ID: mdl-10528405

ABSTRACT

AaIT is an insect selective neurotoxic polypeptide shown to affect insect neuronal sodium conductance by binding to excitable sodium channels. In the present study the paralytic potency of AaIT to wild type and various mutant strains of houseflies (Musca domestica) and fruitflies (Drosophila melanogaster) was examined and it has been shown that: On the basis of body weight when compared to published data on Sarcophaga falculata blowflies, the Musca and Drosophila flies reveal at least two orders of magnitude decreased susceptibility to the AaIT. When compared to wild type flies the toxicity of AaIT is greatly altered in knockdown resistant fly strains which are mutated in their para gene encoding the voltage gated sodium channel. Several strains, with genetically mapped para mutations conferring pyrethroid resistance, exhibited opposing response to AaIT. The para ts2 Drosophila strain, with a point of mutation in domain I of the para gene conferring a 6-fold resistance to deltamethrin also showed about 15-fold tolerance to AaIT. On the other hand the Musca kdr and super-kdr flies, with a single or a double point mutation, respectively in domain II of the para gene, are about 9- and 14-fold more susceptible to AaIT, respectively. The above data are interpreted in terms of the pharmacological diversity and flexibility ("allosteric coupling") of voltage gated sodium channels and their implications for the management of pesticide resistance are discussed.


Subject(s)
Drosophila melanogaster , Houseflies , Insecticides , Ion Channel Gating/physiology , Scorpion Venoms , Animals , Insecticide Resistance , Neurotoxins
6.
Eur J Neurosci ; 11(4): 1449-60, 1999 Apr.
Article in English | MEDLINE | ID: mdl-10103139

ABSTRACT

A scorpion alpha-toxin-sensitive background sodium channel was characterized in short-term cultured adult cockroach dorsal unpaired median (DUM) neurons using the cell-attached patch-clamp configuration. Under control conditions, spontaneous sodium currents were recorded at different steady-state holding potentials, including the range of normal resting membrane potential. At -50 mV, the sodium current was observed as unclustered, single openings. For potentials more negative than -70 mV, investigated patches contained large unitary current steps appearing generally in bursts. These background channels were blocked by tetrodotoxin (TTX, 100 nm), and replacing sodium with TMA-Cl led to a complete loss of channel activity. The current-voltage relationship has a slope conductance of 36 pS. At -50 mV, the mean open time constant was 0.22 +/- 0.05 ms (n = 5). The curve of the open probability versus holding potentials was bell-shaped, with its maximum (0.008 +/- 0.004; n = 5) at -50 mV. LqhalphaIT (10-8 m) altered the background channel activity in a time-dependent manner. At -50 mV, the channel activity appeared in bursts. The linear current-voltage relationship of the LqhalphaIT-modified sodium current determined for the first three well-resolved open states gave three conductance levels: 34, 69 and 104 pS, and reversed at the same extrapolated reversal potential (+52 mV). LqhalphaIT increased the open probability but did not affect either the bell-shaped voltage dependence or the open time constant. Mammal toxin AaHII induced very similar effects on background sodium channels but at a concentration 100 x higher than LqhalphaIT. At 10-7 m, LqhalphaIT produced longer silence periods interrupted by bursts of increased channel activity. Whole-cell experiments suggested that background sodium channels can provide the depolarizing drive for DUM neurons essential to maintain beating pacemaker activity, and revealed that 10-7 m LqhalphaIT transformed a beating pacemaker activity into a rhythmic bursting.


Subject(s)
Biological Clocks , Ganglia, Invertebrate/drug effects , Neurons/drug effects , Neurosecretory Systems/drug effects , Neurotoxins/pharmacology , Scorpion Venoms/pharmacology , Sodium Channels/drug effects , Animals , Biophysical Phenomena , Biophysics , Cockroaches , Ganglia, Invertebrate/cytology , Kinetics , Membrane Potentials/drug effects , Patch-Clamp Techniques , Probability , Reptilian Proteins , Tetrodotoxin/pharmacology
7.
Annu Rev Entomol ; 44: 429-55, 1999.
Article in English | MEDLINE | ID: mdl-9990721

ABSTRACT

Examination of the function, chemistry, and pharmacology of the voltage-gated insect sodium channel (ISC) reveals that the ISC closely resembles its vertebrate counterpart in electrophysiology and ion conductance, primary structure and allocation of all functional domains, and its pharmacological diversity and flexibility exhibited by the occurrence of different allosterically coupled receptor-binding sites for various neurotoxicants. The toxicants include several groups of insecticides, namely DDT and its analogues, pyrethroids, N-alkylamides, and dihydropyrazoles, which affect channel gating and ion permeability. Despite their similarity, the insect and vertebrate channels are pharmacologically distinguishable, as revealed by the responsiveness of the heterologously expressed Drosophila para clone to channel modifiers and blockers and the occurrence of the insect-selective sodium channel neurotoxins derived from arachnid venoms presently used for the design of recombinant baculovirus-mediated selective bioinsecticides. The pharmacological specificity of the ISC may lead to the design of insect-selective toxicants, and its pharmacological flexibility may direct the use of ISC insecticides for resistance management. Insecticide resistance [such as knockdown resistance (KDR)] is acquired by natural selection and operated by increased metabolism, channel mutagenesis, or both. The resistance issue can be dealt with in several ways. One is by simultaneous application of low doses of synergistic, allosterically coupled mixtures (thus delaying or preventing the onset of resistance). An alternative is to replace an insecticide to which resistance was acquired by channel mutation with a different ISC toxicant to which increased susceptibility was conferred by the same mutation. Such a possibility was exemplified by a significant increase in susceptibility to N-alkylamides, as well as an insect-selective neurotoxin revealed by KDR insects. Third, both of these methods can be combined. Thus owing to its pharmacological uniqueness, the ISC may serve as a high-priority target for future selective and resistance-manageable insecticides.


Subject(s)
Insecta/drug effects , Insecta/metabolism , Insecticides/pharmacology , Sodium Channels/drug effects , Animals , Binding Sites , Drosophila/drug effects , Drosophila/genetics , Drosophila/metabolism , Gene Expression , Genes, Insect , Insecta/genetics , Insecticide Resistance , Insecticides/metabolism , Neurotoxins/metabolism , Neurotoxins/pharmacology , Sodium Channels/genetics , Sodium Channels/metabolism , Vertebrates
8.
Biochemistry ; 37(6): 1470-7, 1998 Feb 10.
Article in English | MEDLINE | ID: mdl-9484216

ABSTRACT

A novel gamma-carboxyglutamate-containing peptide, designated gamma-conotoxin-PnVIIA, is described from the venom of the molluscivorous snail Conus pennaceus. gamma PnVIIA, triggers depolarization and firing of action potential bursts in the caudodorsal neurons of Lymnaea. This effect is due to activation or enhancement of a slow inward cation current that may underly endogenous bursting activity of these neurons. The amino acid sequence of gamma PnVIIA was determined as DCTSWFGRCTVNS gamma CCSNSCDQTYC gamma-LYAFOS (where gamma is gamma-carboxyglutamate, O is trans-4-hydroxyproline), thus gamma PnVIIA belongs to the six cysteine four loop structural family of conotoxins, and is most homologous to the previously described excitatory conotoxin-TxVIIA. Interestingly, TxVIIA did not induce action potentials in Lymnaea caudodorsal neurons. gamma PnVIIA is the prototype of a new class of gamma-conotoxins that will provide tools for the study of voltage-gated pacemaker channels, which underly bursting processes in excitable systems.


Subject(s)
1-Carboxyglutamic Acid/agonists , Conotoxins , Ion Channels/drug effects , Mollusk Venoms/agonists , Mollusk Venoms/isolation & purification , Neurons/drug effects , Peptides/agonists , Peptides/isolation & purification , Action Potentials/drug effects , Amino Acid Sequence , Animals , Ion Channels/physiology , Lymnaea , Molecular Sequence Data , Mollusk Venoms/chemistry , Neurons/physiology , Neurosecretory Systems/drug effects , Neurosecretory Systems/physiology , Neurotoxins/agonists , Neurotoxins/chemistry , Neurotoxins/isolation & purification , Paralysis/chemically induced , Peptides/chemistry
9.
J Exp Biol ; 200(Pt 7): 1115-23, 1997.
Article in English | MEDLINE | ID: mdl-9318940

ABSTRACT

Androctonus australis insect toxin (AaIT) is an insect-selective neurotoxic polypeptide from scorpion venom used to probe insect Na+ channels and to design insecticidal recombinant baculoviruses. When injected into susceptible insects (such as flies or cockroaches), nanogram doses of the toxin induce a rapid paralysis within seconds. More tolerant insects respond to microgram doses by developing either a slow progressive paralysis, as in lepidopterous larvae, or a rapid but reversible paralysis, as in Trachyderma philistina, a tenebrionid beetle. Using toxicity and binding assays, microscopy and chromatography, we show that the tolerance of insects to AaIT occurs at both the pharmacokinetic and pharmacodynamic levels. Pharmacokinetic effects occur in Trachyderma philistina in which the toxin undergoes a progressive process of degradation and elimination from the hemolymph, resulting in the loss of 95­97 % of toxin activity 6 h after injection. The pharmacodynamic aspect was demonstrated in studies of the kinetics of binding dissociation of [125I]AaIT from neuronal membranes of susceptible and tolerant insects. Stable binding is shown in susceptible insects such as cockroaches and locusts, which have a dissociation half-time of approximately 9 and 5 min, respectively. This contrasts strongly with the fast half-time of dissociation of 7 s for Spodoptera littoralis larvae and 9 s for Trachyderma philistina, which are both relatively tolerant to AaIT. These differences in binding kinetics may reflect a structural and functional diversity of Na+ channels in different insects that is responsible for their diverse susceptibility to neurotoxic polypeptides.

10.
J Neurochem ; 67(6): 2451-60, 1996 Dec.
Article in English | MEDLINE | ID: mdl-8931478

ABSTRACT

The delta-conotoxin-TxVIA from Conus textile (delta TxVIA) is a mollusk-specific conotoxin that slows sodium channel inactivation exclusively in mollusk neuronal membranes but reveals high-affinity binding to both mollusk (effective binding) and rat brain (silent binding) neuronal membranes, despite not having any toxic effect in vertebrates in vivo and in vitro. Using binding studies with radioactive delta TxVIA we demonstrate that a different mollusk-specific conotoxin, delta-conotoxin-GmVIA from the venom of Conus gloriamaris, possesses "silent" and effective binding properties in rat brain and mollusk sodium channels, respectively. Binding studies and electrophysiological tests with both vertebrate muscle and insect neuronal preparations have indicated that the silent binding sites of delta TxVIA are highly conserved in a wide range of distinct vertebrate and insect sodium channels. Direct probing of receptor site 2 by a tritiated derivative of batrachotoxin ([3H]BTX-B) revealed that [3H]BTX-B binding in mollusk sodium channels is of high affinity with no addition of enhancing ligands, unlike [3H]BTX-B binding in rat brain. In contrast to the negative allosteric modulation of delta TxVIA binding by veratridine, delta TxVIA is not able to affect the binding of [3H]BTX-B in mollusk neuronal membranes but reduces [3H]BTX-B binding in rat brain in the presence of alpha-scorpion toxins. The latter finding indicates the existence of a pharmacological distinction between the silent and effective binding sites of delta TxVIA and points out possible functionally important structural differences between molluscan and rat brain sodium channels.


Subject(s)
Alkaloids/metabolism , Conotoxins , Peptides, Cyclic/chemistry , Peptides, Cyclic/metabolism , Sodium Channels/metabolism , Alkaloids/chemistry , Alkaloids/pharmacology , Animals , Anura , Axons/chemistry , Axons/drug effects , Batrachotoxins/pharmacology , Binding Sites/drug effects , Binding Sites/physiology , Binding, Competitive/physiology , Cell Membrane/chemistry , Cell Membrane/drug effects , Cell Membrane/physiology , Cockroaches , Electrophysiology , Grasshoppers , Helix, Snails , Ion Channel Gating/physiology , Membrane Potentials/drug effects , Mollusk Venoms/chemistry , Mollusk Venoms/metabolism , Mollusk Venoms/pharmacology , Neurotoxins/chemistry , Neurotoxins/metabolism , Neurotoxins/pharmacology , Peptides, Cyclic/pharmacology , Rats , Rats, Inbred Strains , Sodium Channel Agonists , Sodium Channel Blockers , Torpedo , Tritium
11.
J Exp Zool ; 275(6): 444-51, 1996 Aug 15.
Article in English | MEDLINE | ID: mdl-8795288

ABSTRACT

With the aid of dialysis and ion exchange chromatography, a new polypeptide toxin was purified from the tentacles of the Mediterranean jellyfish Rhopilema nomadica. The amino acid sequence of the N-terminal segment of the new toxin revealed that it is a phospholipase A2 (PhA2) toxin closely resembling those previously isolated from reptile and hymenopterous venoms. The occurrence of a PhA2 toxin in the jellyfish tentacles may explain both their local (dermanecrotic) and systemic (cardiac-respiratory) effects upon human envenomation. We used an antibody raised against the above toxin as a probe to explore, for the first time, the site of toxin allocation in cnidarian nematocysts and its morphological route of delivery. Our immunocytochemical approach revealed that the toxin is stored on the outer ("cytoplasmic") surface of the inverted tubule folded in the capsule of the resting nematocyst. During discharge the toxin is translocated to the internal surface surrounding the lumen of the everting tubule, and its delivery via extended spirally arrayed barbs is apparently propelled by the high hydrostatic pressure of the capsule. This is a unique example where subcellular translocation and transfer of a polypetide is driven by mechanical forces.


Subject(s)
Cnidarian Venoms/metabolism , Marine Toxins/metabolism , Scyphozoa/metabolism , Amino Acid Sequence , Animals , Cnidarian Venoms/chemistry , Cnidarian Venoms/isolation & purification , Immunohistochemistry , Marine Toxins/chemistry , Marine Toxins/isolation & purification , Microscopy, Immunoelectron , Molecular Sequence Data , Phospholipases A/chemistry , Phospholipases A/isolation & purification , Phospholipases A/metabolism , Phospholipases A2 , Scyphozoa/anatomy & histology
12.
Biochemistry ; 35(31): 10215-22, 1996 Aug 06.
Article in English | MEDLINE | ID: mdl-8756487

ABSTRACT

The alpha neurotoxin Lqh alpha IT is toxic to both insects and mammals but exhibits a bioactivity ratio favoring insects (insect/mammal approximately 2). With the objective of increasing this ratio by genetic manipulation of the amino acid sequence, a cDNA clone encoding Lqh alpha IT was used to produce recombinant variants of the toxin in a high efficiency bacterial expression system. The unmodified recombinant toxin, isolated from inclusion bodies and renatured in vitro, exhibited chemical and biological properties indistinguishable from those of the authentic native toxin. Alteration of the toxin by site-directed mutagenesis led to a substantial reduction in anti-mammalian toxicity (mouse LD50 reduced 6.4-fold) but only a slight reduction (x 1.5) in the insect ED50 value for paralysis. The reduction in anti-mammalian toxicity was correlated with a approximately 2-fold reduction of its potency for slowing of sodium channel inactivation in mammalian neurons, while no change in mutant toxin binding affinity to insect neuronal receptors was registered. These results demonstrate for the first time expression of a recombinant sodium channel neurotoxin in Escherichia coli and the use of site-directed mutagenesis to improve phylogenetic selectivity. This recombinant approach provides a promising strategy for optimizing the selective toxicity of peptide neurotoxins.


Subject(s)
Neurons/physiology , Neurotoxins/toxicity , Scorpion Venoms/biosynthesis , Amino Acid Sequence , Animals , Base Sequence , Chromatography, High Pressure Liquid , Electrophoresis, Polyacrylamide Gel , Escherichia coli/ultrastructure , Ganglia, Spinal/physiology , Insecta , Isopropyl Thiogalactoside , Mammals , Microscopy, Electron , Molecular Sequence Data , Mutagenesis, Site-Directed , Neurons/drug effects , Neurons/pathology , Neurotoxins/chemistry , Polymerase Chain Reaction , Rats , Recombinant Proteins/biosynthesis , Recombinant Proteins/chemistry , Recombinant Proteins/toxicity , Scorpion Venoms/chemistry , Scorpion Venoms/toxicity , Scorpions , Sequence Homology, Amino Acid , Sequence Homology, Nucleic Acid , Sodium Channels/drug effects , Species Specificity
13.
Toxicon ; 34(4): 435-42, 1996 Apr.
Article in English | MEDLINE | ID: mdl-8735242

ABSTRACT

The effects of scorpion venom Leiurus quinquestriatus hebreus were studied on cardiac cells grown in culture. The venom (30 micrograms/ml) increased significantly (P < 0.05) Ca2+ uptake into intact cardiocytes and to sarcoplasmic reticulum of skinned cells. [3H]Deoxyglucose uptake was also increased significantly (P < 0.05) in venom treated cardiocytes. It was found that fractions I and III of the venom, separated by gel filtration and ion exchange chromatography, are responsible for the increased Ca2+ uptake by the sarcoplasmic reticulum, whereas fraction IIb, III and IV are responsible for the accelerated rate of uptake of 45Ca and [3H]deoxyglucose by intact cells. Ca channel blockers prevented these effects and similar results were obtained by propranolol. Thus, it is concluded that the venom exerts its effect through activation of beta-adrenoceptors which causes the opening of L-type Ca channels.


Subject(s)
Calcium/metabolism , Deoxyglucose/metabolism , Heart/drug effects , Myocardium/metabolism , Scorpion Venoms/toxicity , Adrenergic beta-Antagonists/pharmacology , Animals , Calcium Channel Blockers/pharmacology , Calcium Channels/drug effects , Cells, Cultured , Chemical Fractionation , Chromatography, Gel , Chromatography, Ion Exchange , Dose-Response Relationship, Drug , Myocardial Contraction/drug effects , Myocardium/cytology , Propranolol/pharmacology , Rats , Sarcoplasmic Reticulum/drug effects , Sarcoplasmic Reticulum/metabolism , Scorpion Venoms/chemistry , Scorpion Venoms/metabolism , Sodium-Potassium-Exchanging ATPase/metabolism
15.
FEBS Lett ; 376(3): 181-4, 1995 Dec 04.
Article in English | MEDLINE | ID: mdl-7498537

ABSTRACT

The Leiurus quinquestriatus hebraeus alpha anti-insect toxin (Lqh alpha IT) cDNA was engineered into the Autographa californica Nuclear Polyhedrosis Virus (AcNPV) genome. Insect cells infected with the recombinant virus secreted a functional Lqh alpha IT polypeptide. Spodoptera littoralis and Heliothis armigera larvae injected with recombinant budded virus, showed typical intoxication symptoms. This recombinant virus showed enhanced insecticidal potency against H. armigera larvae compared with wild type AcNPV. The present expression system will facilitate: (1) the future elucidation of structural elements involved in its prominent anti-insect toxicity; and (2) the future design of genetically modified alpha toxins with improved anti-insect selectivity.


Subject(s)
Neurotoxins/genetics , Scorpion Venoms/genetics , Animals , Base Sequence , Cloning, Molecular , DNA Primers/chemistry , Diptera/drug effects , Genetic Vectors , Larva , Molecular Sequence Data , Moths/drug effects , Neurotoxins/toxicity , Nucleopolyhedroviruses/genetics , Recombinant Proteins , Scorpion Venoms/toxicity , Spodoptera/drug effects
16.
Toxicon ; 33(8): 1099-102, 1995 Aug.
Article in English | MEDLINE | ID: mdl-8533143

ABSTRACT

The insecticidal activity of scorpion neurotoxic polypeptides increased 5-10-fold with no apparent increase in mammalian toxicity when a combination of two toxins was injected. Synergistic combinations could be predicted from binding studies and competitive displacement assays. Our results indicate that simultaneous expression in baculovirus or other transgenic organisms of the synergistic combinations of insecticidal toxins may result in more potent insect-selective biopesticides.


Subject(s)
Diptera/drug effects , Moths/drug effects , Neurotoxins/pharmacology , Pest Control, Biological , Pesticide Synergists/pharmacology , Spider Venoms/pharmacology , Animals , Scorpions
17.
Biochemistry ; 34(27): 8649-56, 1995 Jul 11.
Article in English | MEDLINE | ID: mdl-7612605

ABSTRACT

Two novel sodium channel blocking peptides from the venom of the molluscivorous snail Conus pennaceus, muPnIVA and muPnIVB, are described. Elucidation of their amino acid sequences was complicated by a previously undescribed anomalous product of reduction and pyridylethylation, which occurs on N-terminal cysteine residues and gives a PTH derivative eluting at the same position as PTH-Trp in reverse-phase chromatography. The amino acid sequences of the toxins were determined by a combination of Edman degradation and mass-spectrometric techniques as CCKYGWTCLLGCSPCGC (PnIVA) and CCKYGWTCWLGCSPCGC (PnIVB). These toxins block sodium channels in molluscan neurons, but have no effect on sodium currents in bovine chromaffin cells or in rat brain synaptosomes. Although there is only one amino acid difference in the two sequences, PnIVB is approximately 6 times more potent than PnIVA in blockade of the sodium current in Lymnaea neurons. The PnIV sequences reveal a new cysteine residue framework for conotoxins (CC-----C---C--C-C). Strikingly, the only charged residue in PnIVA/B is Lys3. Iodination reaction experiments on the adjacent Tyr4 suggest that this region of the peptide must be solvent exposed and essential for activity. These structurally novel mu-conotoxins target a sodium channel subtype with low affinity for tetrodotoxin and therefore provide new probes for functional studies on sodium channels.


Subject(s)
Conotoxins , Cysteine/chemistry , Mollusk Venoms/chemistry , Oligopeptides/pharmacology , Sodium Channel Blockers , Amino Acid Sequence , Animals , Cattle , Lymnaea , Mass Spectrometry , Molecular Sequence Data , Neurons/drug effects , Neurons/metabolism , Oligopeptides/chemistry , Paralysis/chemically induced , Rats
19.
Eur J Neurosci ; 7(4): 815-8, 1995 Apr 01.
Article in English | MEDLINE | ID: mdl-7620628

ABSTRACT

A novel peptide toxin, PnIVB, isolated from the venom of Conus pennaceus blocks voltage-gated sodium current in Aplysia neurons. Complete blockade is obtained at a PnIVB concentration of 80 +/- 2.2 nM and 50% blockade at 16 +/- 0.86 nM. The potency of PnIVB in blocking Aplysia sodium current is four orders of magnitude larger than that of tetrodotoxin. The toxin has no paralytic activity when injected into fish. The rapid blockade of sodium current by PnIVB is not associated with a change in the activation or inactivation kinetics of the current, or with the reversal potential. Sodium current blockade is reversible after a 30 min wash with 50 times the bath volume. The novel conotoxin PnIVB can be used as a powerful tool for mollusc neurobiological research and as a molecular probe to explore the structure-function relations of voltage-gated sodium channel subtypes.


Subject(s)
Aplysia/physiology , Conotoxins , Mollusk Venoms/pharmacology , Oligopeptides/pharmacology , Snails/physiology , Sodium Channel Blockers , Action Potentials/drug effects , Animals , Cells, Cultured , Electrophysiology , Ion Channel Gating/drug effects , Mollusk Venoms/isolation & purification , Neurons/drug effects , Neurons/metabolism , Oligopeptides/isolation & purification , Patch-Clamp Techniques , Potassium Channels/drug effects , Tetrodotoxin/pharmacology
20.
J Biol Chem ; 270(3): 1123-9, 1995 Jan 20.
Article in English | MEDLINE | ID: mdl-7836370

ABSTRACT

We describe a new peptide conotoxin affecting sodium current inactivation, that competes on binding with delta-conotoxin TxVIA (delta TxVIA). The amino acid sequence of the new toxin, designated conotoxin NgVIA (NgVIA), is SKCFSOGTFCGIKOGLCCSVRCFSLFCISFE (where O is trans-4-hydroxyproline). The primary structure of NgVIA has an identical cysteine framework and similar hydrophobicity as delta TxVIA but differs in its net charge. NgVIA competes with delta TxVIA on binding to rat brain synaptosomes and molluscan central nervous system and strongly inhibits sodium current inactivation in snail neurons, as does delta TxVIA. In contrast to delta TxVIA, NgVIA is a potent paralytic toxin in vertebrate systems, its binding appears to be voltage-dependent, and it synergically increases veratridine-induced sodium influx to rat brain synaptosomes. delta TxVIA acts as a partial antagonist to NgVIA in rat brain in vivo. NgVIA appears to act via a receptor site distinct from that of delta TxVIA but similar to that of Conus striatus toxin. This new toxin provides a lead for structure-function relationship studies in the delta-conotoxins and will enable analysis of the functional significance of this complex of receptor sites in gating mechanisms of sodium channels.


Subject(s)
Conotoxins , Peptides/metabolism , Receptors, Neurotransmitter/metabolism , Sodium Channel Blockers , Sodium/metabolism , Amino Acid Sequence , Animals , Brain/drug effects , Brain/metabolism , Molecular Sequence Data , Peptides/chemistry , Peptides/isolation & purification , Peptides/toxicity , Protein Binding , Rats
SELECTION OF CITATIONS
SEARCH DETAIL
...