Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters










Publication year range
1.
Toxins (Basel) ; 4(11): 1082-119, 2012 Nov 01.
Article in English | MEDLINE | ID: mdl-23202307

ABSTRACT

Scorpion toxins have been central to the investigation and understanding of the physiological role of potassium (K⁺) channels and their expansive function in membrane biophysics. As highly specific probes, toxins have revealed a great deal about channel structure and the correlation between mutations, altered regulation and a number of human pathologies. Radio- and fluorescently-labeled toxin isoforms have contributed to localization studies of channel subtypes in expressing cells, and have been further used in competitive displacement assays for the identification of additional novel ligands for use in research and medicine. Chimeric toxins have been designed from multiple peptide scaffolds to probe channel isoform specificity, while advanced epitope chimerization has aided in the development of novel molecular therapeutics. Peptide backbone cyclization has been utilized to enhance therapeutic efficiency by augmenting serum stability and toxin half-life in vivo as a number of K⁺-channel isoforms have been identified with essential roles in disease states ranging from HIV, T-cell mediated autoimmune disease and hypertension to various cardiac arrhythmias and Malaria. Bioengineered scorpion toxins have been monumental to the evolution of channel science, and are now serving as templates for the development of invaluable experimental molecular therapeutics.


Subject(s)
Bioengineering/methods , Charybdotoxin/chemistry , Potassium Channel Blockers/chemistry , Scorpion Venoms/chemistry , Scorpions/physiology , Animals , Charybdotoxin/genetics , Charybdotoxin/pharmacology , Computer Simulation , Escherichia coli/genetics , HEK293 Cells , Humans , Ligands , Models, Molecular , Potassium Channel Blockers/pharmacology , Potassium Channel Blockers/therapeutic use , Potassium Channels/metabolism , Protein Binding , Protein Conformation , Scorpion Venoms/genetics , Scorpion Venoms/pharmacology , Structure-Activity Relationship , Transfection
2.
J Agric Food Chem ; 58(7): 4051-5, 2010 Apr 14.
Article in English | MEDLINE | ID: mdl-20229998

ABSTRACT

The main objective of the present work was to evaluate a real-time polymerase chain reaction (PCR) method to detect toxigenic Vibrio cholerae in Pangasius hypophthalmus, a freshwater fish cultured mainly in South East Asia. A FDA traditional culture method and a real-time PCR method of the ctx gene were used for detection of V. cholerae in spiked samples of pangasius fish. After an overnight enrichment of samples at 37 degrees C in alkaline peptone water, 2 cfu/25 g of fish was detected with both methods. Although both methods were very sensitive, obtaining results with culture methods may take several days, while real-time PCR takes only a few hours. Furthermore, with traditional methods, complementary techniques such as serotyping, although not available for all serogroups, are needed to identify toxigenic V. cholerae. However, with real-time PCR, toxigenic serogroups are detected in only one step after overnight enrichment.


Subject(s)
Charybdotoxin/genetics , Culture Techniques/methods , Fish Diseases/microbiology , Polymerase Chain Reaction/methods , Vibrio cholerae/isolation & purification , Animals , Bacterial Typing Techniques , Catfishes , Charybdotoxin/analysis , Taq Polymerase/analysis , Vibrio cholerae/chemistry , Vibrio cholerae/genetics
3.
J Chem Inf Model ; 49(7): 1831-8, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19499912

ABSTRACT

The scorpion toxin, charybdotoxin (ChTX), is the first identified peptide inhibitor for the large-conductance Ca2+ and voltage-dependent K+ (BK) channel, and the chemical information of the interaction between ChTX and BK channel remains unclear today. Using combined computational methods, we obtained a ChTX-BK complex structure model, which correlated well with the mutagenesis data. In this complex, ChTX mainly used its beta-sheet domains to associate the BK channel with a conserved pore-blocking Lys27. Another crucial Tyr36 residue of ChTX lied over the loop connecting selectivity filter and S6 helix of BK channel, forming a hydrogen bond with Gly291 of BK channel. Besides, the unique turret region of BK channel was found to be far away from bound ChTX, which could explain the fact that many BK channel blockers show less selectivity over Kv channels. Together, all these information is helpful to reveal the diverse interactions between scorpion toxins and potassium channels and can accelerate the molecular engineering of specific inhibitor design.


Subject(s)
Charybdotoxin/chemistry , Charybdotoxin/metabolism , Potassium Channels, Calcium-Activated/chemistry , Potassium Channels, Calcium-Activated/metabolism , Scorpions/metabolism , Amino Acid Sequence , Animals , Charybdotoxin/genetics , Computer Simulation , Models, Molecular , Molecular Sequence Data , Protein Binding , Protein Conformation , Scorpion Venoms/chemistry , Scorpion Venoms/genetics , Scorpion Venoms/metabolism , Sequence Alignment , Thermodynamics
4.
Biochemistry ; 46(18): 5358-64, 2007 May 08.
Article in English | MEDLINE | ID: mdl-17439246

ABSTRACT

Potassium channel dysfunction underlies diseases such as epilepsy, hypertension, cardiac arrhythmias, and multiple sclerosis. Neurotoxins that selectively inhibit potassium channels, alpha-KTx, have provided invaluable information for dissecting the contribution of different potassium channels to neurotransmission, vasoconstriction, and lymphocyte proliferation. Thus, alpha-KTx specificity comprises an important first step in potassium channel-directed drug discovery for these diseases. Despite extensive functional and structural studies of alpha-KTx-potassium channel complexes, none have predicted the molecular basis of alpha-KTx specificity. Here we show that by minimizing the differences in binding free energy between selective and nonselective alpha-KTx we are able to identify all of the determinants of alpha-KTx specificity for calcium-activated versus voltage-dependent potassium channels. Because these determinants correspond to unique features of the two types of channels, they provide a way to develop more accurate models of alpha-KTx-potassium channel complexes that can be used to design novel selective alpha-KTx inhibitors.


Subject(s)
Kv1.3 Potassium Channel/antagonists & inhibitors , Large-Conductance Calcium-Activated Potassium Channels/antagonists & inhibitors , Potassium Channel Blockers/chemistry , Potassium Channel Blockers/toxicity , Scorpion Venoms/chemistry , Scorpion Venoms/toxicity , Amino Acid Sequence , Amino Acid Substitution/genetics , Animals , CHO Cells , Cell Line , Charybdotoxin/chemistry , Charybdotoxin/genetics , Charybdotoxin/toxicity , Cricetinae , Cricetulus , Humans , Kv1.3 Potassium Channel/chemistry , Large-Conductance Calcium-Activated Potassium Channels/chemistry , Molecular Sequence Data , Peptides/chemistry , Peptides/toxicity , Scorpion Venoms/genetics
6.
Toxicon ; 43(8): 951-60, 2004 Jun 15.
Article in English | MEDLINE | ID: mdl-15208028

ABSTRACT

We have studied the interaction between the SK2 channel and different scorpion toxins in order to find similarity and differences to other K+ channels. Beside apamin, ScTX is a high affinity blocker of the SK2 channel, whereas CTX is unable to block current through SK2. In order to prove that the ScTX affinity can be explained by the character of the different residues in the outer pore of the SK channels we introduced point mutations that render SK2 K+ channel SK1 and SK3 like. Directed by the results of the toxin receptor on the ShakerK+ channel, we changed single amino acids of the SK2 K+ channel that should render it sensitive to other peptide toxins like CTX a blocker of the IK channel, or KTX a blocker of the voltage-dependent channel Kv1.1 and Kv1.3. Amino acids V342G, S344E, and G384D of SK2 were changed to amino acids known from ShakerK+ channel to improve Shaker K+ channel CTX sensitivity. Interestingly SK2 V342G became CTX sensitive with a Kd of 19 nM and was also KTX sensitive Kd=97 nM. SK2 S344E (KdCTX = 105 nM,KdKTX = 144 nM) and G348D (KdCTX = 31 nM,Kd KTX = 89 nM) became also CTX and KTX sensitive with a lower affinity. The mutant channels SK V342G, SK2 S344E and SK2 G348D showed reduced ScTX sensitivity (Kd = 6 nM,Kd = 48 nM, and Kd = 12 nM). Because the exchange of a single residue could create a new high affinity binding site for CTX and KTX we concluded that the outer vestibule around position V342, S344, and G348 of the SK2 K+ channel pore is very similar to those of voltage-gated K+ channels such as the Shaker K+ channel, Kv1.1 and Kv1.3 channels and also to the prokaryotic KcsA channel. From mutant cycle analysis of KTX position H34 and SK2 position V342G, S344E, and G348D we could deduce that KTX binds in a similar way to SK2 channel mutant pore than to the Kv1.1 pore. We have studied the interaction between the SK2 channel and different scorpion toxins in order to find similarity and differences to other K+ channels. Beside apamin, ScTX is a high affinity blocker of the SK2 channel, whereas CTX is unable to block current through SK2. In order to prove that the ScTX affinity can be explained by the character of the different residues in the outer pore of the SK channels we introduced point mutations that render SK2 K+ channel SK1 and SK3 like. Directed by the results of the toxin receptor on the ShakerK+ channel, we changed single amino acids of the SK2 K+ channel that should render it sensitive to other peptide toxins like CTX a blocker of the IK channel, or KTX a blocker of the voltage-dependent channel Kv1.1 and Kv1.3. Amino acids V342G, S344E, and G384D of SK2 were changed to amino acids known from ShakerK+ channel to improve Shaker K+ channel CTX sensitivity. Interestingly SK2 V342G became CTX sensitive with a Kd of 19 nM and was also KTX sensitive Kd = 97 nM. SK2 S344E (KdCTX = 105 nM,KdKTX = 144 nM) and G348D (KdCTX = 31 nM,Kd KTX = 89 nM) became also CTX and KTX sensitive with a lower affinity. The mutant channels SK V342G, SK2 S344E and SK2 G348D showed reduced ScTX sensitivity (Kd = 6 nM,Kd = 48 nM, and Kd = 12 nM). Because the exchange of a single residue could create a new high affinity binding site for CTX and KTX we concluded that the outer vestibule around position V342, S344, and G348 of the SK2 K+ channel pore is very similar to those of voltage-gated K+ channels such as the Shaker K+ channel, Kv1.1 and Kv1.3 channels and also to the prokaryotic KcsA channel. From mutant cycle analysis of KTX position H34 and SK2 position V342G, S344E, and G348D we could deduce that KTX binds in a similar way to SK2 channel mutant pore than to the Kv1.1 pore.


Subject(s)
Charybdotoxin/metabolism , Potassium Channels, Calcium-Activated , Potassium Channels/metabolism , Scorpion Venoms/metabolism , Scorpions/chemistry , Amino Acid Sequence , Amino Acid Substitution , Animals , Apamin/genetics , Apamin/metabolism , Binding Sites/physiology , Calcium/metabolism , Cells, Cultured , Charybdotoxin/genetics , DNA, Complementary/genetics , Electrophysiology , Genetic Vectors , Humans , Hydrogen-Ion Concentration , Intermediate-Conductance Calcium-Activated Potassium Channels , Membrane Potentials/physiology , Molecular Sequence Data , Potassium Channels/genetics , Potassium Channels/physiology , Protein Binding/physiology , Scorpion Venoms/genetics , Sequence Alignment , Small-Conductance Calcium-Activated Potassium Channels , Transfection
7.
Toxicon ; 43(4): 447-53, 2004 Mar 15.
Article in English | MEDLINE | ID: mdl-15051409

ABSTRACT

BmTX3 is a toxin recently characterised from the venom of the Chinese scorpion Buthus martensi Karch, which specifically blocks a transient A-type K+ current in striatum neurons in culture and binds to rat brain synaptosomes with high affinity. With Aa1 and AmmTX3, it belongs to the new alpha-KTx15 subfamily from "short-chain" scorpion toxins, which specifically block different types of K+ channels. Here, a highly specific polyclonal antiserum was raised in rabbit against a C-terminal deleted BmTX3 analogue (BmTX-del YP). Using liquid-phase radioimmunoassay, we have studied its selectivity for the toxins from the alpha-KTx15 subfamily. We have also demonstrated that no/or poor cross-reactivity was observed with a panel of "short-chain" scorpion toxins representative of other structurally different subfamilies. These results suggest that a wide antigenic polymorphism, similar to that previously observed for "long-chain" scorpion toxins acting as modulators of voltage-activated Na+ channels, is also the rule for the "short-chain" scorpion toxins able to block K+ channels.


Subject(s)
Genetic Variation , Immune Sera/immunology , Models, Molecular , Scorpion Venoms/chemistry , Scorpion Venoms/immunology , Scorpions , Amino Acid Sequence , Animals , Binding, Competitive , Charybdotoxin/genetics , Cross Reactions , Enzyme-Linked Immunosorbent Assay , Molecular Sequence Data , Protein Conformation , Radioimmunoassay , Scorpion Venoms/genetics , Sequence Alignment , Sequence Homology
8.
Biochemistry ; 40(37): 10987-97, 2001 Sep 18.
Article in English | MEDLINE | ID: mdl-11551194

ABSTRACT

Noxiustoxin (NxTX) displays an extraordinary ability to discriminate between large conductance, calcium-activated potassium (maxi-K) channels and voltage-gated potassium (Kv1.3) channels. To identify features that contribute to this specificity, we constructed several NxTX mutants and examined their effects on whole cell current through Kv1.3 channels and on current through single maxi-K channels. Recombinant NxTX and the site-specific mutants (P10S, S14W, A25R, A25Delta) all inhibited Kv1.3 channels with Kd values of 6, 30, 0.6, 112, and 166 nM, respectively. In contrast, these same NxTX mutants had no effect on maxi-K channel activity with estimated Kd values exceeding 1 mM. To examine the role of the alpha-carbon backbone in binding specificity, we constructed four NxTX chimeras, which altered the backbone length and the alpha/beta turn. For each of these chimeras, six amino acids comprising the alpha/beta turn in iberiotoxin (IbTX) replaced the corresponding seven amino acids in NxTX (NxTX-YGSSAGA21-27-FGVDRG21-26). The chimeras differed in length of N- and C-terminal residues and in critical contact residues. In contrast to NxTX and its site-directed mutants, all of these chimeras inhibited single maxi-K channels. Under low ionic strength conditions, Kd values ranged from 0.4 to 6 microM, association rate constant values from 3 x 10(7) to 3 x 10(8) M(-1) x s(-1), and time constants for block from 5 to 20 ms. The rapid blocked times suggest that key microscopic interactions at the toxin-maxi-K channel interface may be absent. Under physiologic external ionic strength conditions, these chimera inhibited Kv1.3 channels with Kd values from 30 to 10 000 nM. These results suggest that the extraordinary specificity of NxTX for Kv1.3 over maxi-K channels is controlled, in part, by the toxin alpha-carbon backbone. These differences in the alpha-carbon backbone are likely to reflect fundamental structural differences in the external vestibules of these two channels.


Subject(s)
Potassium Channels, Calcium-Activated , Potassium Channels, Voltage-Gated , Potassium Channels , Scorpion Venoms/antagonists & inhibitors , Scorpion Venoms/genetics , Scorpion Venoms/pharmacology , Binding Sites , Charybdotoxin/genetics , Charybdotoxin/pharmacology , Dose-Response Relationship, Drug , Kv1.3 Potassium Channel , Large-Conductance Calcium-Activated Potassium Channels , Models, Molecular , Osmolar Concentration , Peptides/genetics , Peptides/pharmacology , Potassium Channel Blockers , Protein Binding , Protein Structure, Secondary , Recombinant Fusion Proteins , Thermodynamics
9.
J Membr Biol ; 179(1): 13-25, 2001 Jan 01.
Article in English | MEDLINE | ID: mdl-11155206

ABSTRACT

Pandinus imperator scorpion toxins Pi2 and Pi3 differ only by a single amino acid residue (neutral Pro7 in Pi2 vs. acidic Glu7 in Pi3). The binding kinetics of these toxins to human Kv1.3 showed that the decreased ON rate (k(ON) = 2.18 x 10(8) m(-1)sec(-1) for Pi2 and 1.28 x 10(7) m(-1)sec(-1) for Pi3) was almost entirely responsible for the increased dissociation constant (K(d)) of Pi3 (K(d) = 795 pm) as compared to Pi2 (K(d) = 44 pm). The ionic strength dependence of the association rates was exactly the same for the two toxins indicating that through-space electrostatic interactions can not account for the different ON rates. Results were further analyzed on the basis of the three-dimensional structural models of the toxins. A 3D structure of Pi3 was generated from the NMR spectroscopy coordinates of Pi2 by computer modeling. The Pi3 model resulted in a salt bridge between Glu7 and Lys24 in Pi3. Based on this finding our interpretation of the reduced ON rate of Pi3 is that the intramolecular salt bridge reduces the local positive electrostatic potential around Lys24 resulting in decreased short-range electrostatic interactions during the binding step. To support our finding, we constructed a 3D model of the Ser-10-Asp Charybdotoxin mutant displaying distinctly reduced affinity for Shaker channels. The mutant Charybdotoxin structure also displayed a salt bridge between residues Asp10 and Lys27 equivalent to the one between Glu7 and Lys24 in Pi3.


Subject(s)
Potassium Channel Blockers , Potassium Channels, Voltage-Gated , Scorpion Venoms/pharmacology , Amino Acid Sequence , Animals , Charybdotoxin/chemistry , Charybdotoxin/genetics , Charybdotoxin/metabolism , Charybdotoxin/pharmacology , Glutamic Acid/chemistry , Humans , In Vitro Techniques , Kinetics , Kv1.3 Potassium Channel , Lysine/chemistry , Membrane Potentials , Models, Molecular , Molecular Sequence Data , Point Mutation , Potassium Channels/metabolism , Protein Conformation , Scorpion Venoms/chemistry , Scorpion Venoms/genetics , Scorpion Venoms/metabolism , Sequence Homology, Amino Acid , T-Lymphocytes/drug effects , T-Lymphocytes/metabolism
10.
Article in English | MEDLINE | ID: mdl-10048185

ABSTRACT

The primary structures of four low molecular mass peptides (Bs 6, 8, 10 and 14) from scorpion Buthus sindicus were elucidated via combination of Edman degradation and matrix-assisted laser desorption ionization mass spectrometry. Bs 8 and 14 are cysteine-rich, thermostable peptides composed of 35-36 residues with molecular weights of 3.7 and 3.4 kDa, respectively. These peptides show close sequence homologies (55-78%) with other scorpion chlorotoxin-like short-chain neurotoxins (SCNs) containing four intramolecular disulfide bridges. Despite the sequence variation between these two peptides (37% heterogeneity) their general structural organization is very similar as shown by their clearly related circular dichroism spectra. Furthermore, Bs6 is a minor component, composed of 38 residues (4.1 kDa) containing six half-cystine residues and having close sequence identities (40-80%) with charybdotoxin-like SCNs containing three disulfide bridges. The non-cysteinic, bacic and thermolabile Bs10 is composed of 34 amino acid residues (3.7 kDa), and belongs to a new class of peptides, with no sequence resemblance to any other so far reported sequence isolated from scorpions. Surprisingly, Bs10 shows some limited sequence analogy with oocyte zinc finger proteins. Results of these studies are discussed with respect to their structural similarities within the scorpion LCNs, SCNs and other biologically active peptides.


Subject(s)
Peptide Fragments/analysis , Peptide Fragments/isolation & purification , Scorpion Venoms/chemistry , Animals , Charybdotoxin/chemistry , Charybdotoxin/genetics , Chromatography, High Pressure Liquid , Cysteine , Molecular Sequence Data , Molecular Weight , Peptide Fragments/genetics , Scorpion Venoms/genetics , Scorpions , Sequence Homology, Amino Acid
11.
Bioorg Khim ; 23(9): 710-20, 1997 Sep.
Article in Russian | MEDLINE | ID: mdl-9441593

ABSTRACT

A theoretical method for the rational design of a "universal" scorpion toxin with a wider spectrum of specificity for K+ channels and a more stable alpha/beta-folding than in its natural homologues is described. On the basis of the analysis of molecular hydrophobic potentials (MHP) of the protein spatial structures, structural features for a family of five short scorpion toxins were revealed. The analysis of the maps of two-dimensional intramolecular MHP contacts allowed the identification of amino acid residues responsible for the folding of the protein and/or for the manifestation of its specific function. The theoretically predicted structure-function roles of the residues were compared with experimental data on the mutagenesis of charybdotoxin. Based on the results of MHP calculations and with the theory of protein molecular evolution used as an additional criterion for the selection of mutations, the amino acid sequence and the spatial structure of a "universal" scorpion toxin were determined.


Subject(s)
Potassium Channels/drug effects , Scorpion Venoms/chemistry , Scorpion Venoms/pharmacology , Amino Acid Sequence , Amino Acids/analysis , Animals , Charybdotoxin/chemistry , Charybdotoxin/genetics , Evolution, Molecular , Models, Theoretical , Molecular Sequence Data , Mutagenesis/genetics , Scorpion Venoms/genetics , Sequence Alignment , Sequence Homology, Amino Acid , Structure-Activity Relationship
12.
Biochemistry ; 35(20): 6181-7, 1996 May 21.
Article in English | MEDLINE | ID: mdl-8639558

ABSTRACT

A structural model of charybdotoxin bound to a Shaker K+ channel has emerged from mechanistic and mutagenic analysis of toxin-channel interactions. We test this model by predicting through-space electrostatic interactions between specific pairs of channel-toxin residues. Dissociation constants of channel-toxin variants, determined by radiolabeled toxin binding to Shaker-transfected COS membrane fragments, were used to identify pairs of residues located closely enough to interact electrostatically. The results further refine the structural model of the bound complex and produce a more detailed view of the vestibule of the Shaker channel.


Subject(s)
Charybdotoxin/chemistry , Charybdotoxin/metabolism , Potassium Channels/chemistry , Potassium Channels/metabolism , Animals , Binding Sites , Cell Line , Charybdotoxin/genetics , Electrochemistry , Ion Channel Gating , Magnetic Resonance Spectroscopy , Models, Molecular , Molecular Structure , Potassium Channels/genetics , Protein Conformation , Shaker Superfamily of Potassium Channels , Thermodynamics , Transfection
13.
Neuron ; 16(1): 123-30, 1996 Jan.
Article in English | MEDLINE | ID: mdl-8562075

ABSTRACT

Charybdotoxin, a peptide neurotoxin of known molecular structure, blocks Shaker K+ channels by binding to a receptor at the outer opening of the ion conduction pathway. Analysis of variants of CTX at position 29 and of Shaker at position 449 shows that these two residues interact closely in the channel-toxin complex. The CTX mutation M29I leads to a slight strengthening of block when tested on Shaker-449T; the same CTX mutation weakens block 1700-fold when tested on Shaker-449F. The known position of CTX-29 on the toxin's interaction surface thus locates Shaker-449 within 5 A of the pore axis of the closed channel. All four subunits must carry the 449F mutation to produce a highly toxin-insensitive channel.


Subject(s)
Charybdotoxin/chemistry , Potassium Channels/chemistry , Protein Conformation , Amino Acid Sequence , Animals , Binding Sites , Charybdotoxin/genetics , Charybdotoxin/metabolism , Charybdotoxin/pharmacology , Drosophila Proteins , Drosophila melanogaster/genetics , Ion Channel Gating/drug effects , Lysine/chemistry , Molecular Sequence Data , Oocytes , Phenylalanine/chemistry , Potassium/metabolism , Potassium Channels/drug effects , Potassium Channels/genetics , Potassium Channels/metabolism , Protein Binding , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/metabolism , Scorpions/genetics , Shaker Superfamily of Potassium Channels , Threonine/chemistry , Transfection , Xenopus laevis
14.
Biochemistry ; 34(51): 16563-73, 1995 Dec 26.
Article in English | MEDLINE | ID: mdl-8527429

ABSTRACT

The 3D structure of noxiustoxin, the first identified scorpion toxin acting on K+ channels, has been elucidated by NMR and molecular modeling. Thirty-nine solution structures were calculated using 572 distance and 42 dihedral restraints. The average atomic rms deviation between the refined structures and the mean structure is 0.75 A for the backbone atoms. Noxiustoxin adopts a alpha/beta scaffold constituted of a three-stranded beta-sheet (residues 2-3, 25-30, 33-38) linked to a helix (residues 10-20) through two disulfide bridges. A comparison between the 3D structure of noxiustoxin and those of other structurally and functionally related scorpion toxins (charybdotoxin, PO5-NH2, kaliotoxin) revealed a bending capacity of the helix and a variability in the relative orientations between the helix and the beta-sheet. These two features highlight the plasticity of the alpha/beta scaffold and offer a structural explanation for the capacity of the fold to accommodate an additional alanine residue in the Gly-x-Cys pattern of a previously proposed consensus sequence [Bontems et al. (1991) Science 254, 1521-1523]. Our structural data also emphasize the possibility that the beta-sheet of NTX is implicated in the capacity of NTX to recognize voltage-dependent K+ channels.


Subject(s)
Scorpion Venoms/chemistry , Amino Acid Sequence , Animals , Charybdotoxin/chemistry , Charybdotoxin/genetics , Disulfides/chemistry , Electrochemistry , Hydrogen Bonding , Magnetic Resonance Spectroscopy , Models, Molecular , Molecular Sequence Data , Molecular Structure , Potassium Channels/drug effects , Protein Structure, Secondary , Scorpion Venoms/genetics , Scorpion Venoms/toxicity , Scorpions , Sequence Homology, Amino Acid , Thermodynamics
15.
J Membr Biol ; 147(1): 71-82, 1995 Sep.
Article in English | MEDLINE | ID: mdl-8531201

ABSTRACT

Despite recent progress in the molecular characterization of high-conductance Ca(2+)-activated K+ (maxi-K) channels, the molecular identities of intermediate conductance Ca(2+)-activated K+ channels, including that of mature erythrocytes, remains unknown. We have used various peptide toxins to characterize the intermediate conductance Ca(2+)-activated K+ channels (Gardos pathway) of human and rabbit red cells. With studies on K+ transport and on binding of 125I-charybdotoxin (ChTX) and 125I-kaliotoxin (KTX) binding in red cells, we provide evidence for the distinct nature of the red cell Gardos channel among described Ca(2+)-activated K+ channels based on (i) the characteristic inhibition and binding patterns produced by ChTX analogues, iberiotoxin (IbTX) and IbTX-like ChTX mutants, and KTX (1-37 and 1-38 variants); (ii) the presence of some properties heretofore attributed only to voltage-gated channels, including inhibition of K transport by margatoxin (MgTX) and by stichodactyla toxin (StK); (iii) and the ability of scyllatoxin (ScyTX) and apamin to displace bound 125I-charybdotoxin, a novel property for K+ channels. These unusual pharmacological characteristics suggest a unique structure for the red cell Gardos channel.


Subject(s)
Calcium/metabolism , Erythrocytes/metabolism , Peptides/toxicity , Potassium Channel Blockers , Potassium Channels/metabolism , Scorpion Venoms/toxicity , Animals , Charybdotoxin/genetics , Charybdotoxin/toxicity , Erythrocytes/drug effects , Humans , In Vitro Techniques , Ion Transport/drug effects , Kinetics , Neurotoxins/toxicity , Point Mutation , Potassium/metabolism , Rabbits , Rubidium/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...