Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 29
Filter
Add more filters










Publication year range
1.
Chemistry ; 7(1): 193-9, 2001 Jan 05.
Article in English | MEDLINE | ID: mdl-11205011

ABSTRACT

Hydrolysis and hydrogenation of [RhCl(tppms)3] (1) and trans-[RhCl(CO)(tppms)2] (2) was studied in aqueous solutions in a wide pH range (2 < pH < 11) in the presence of excess TPPMS (3-diphenylphosphinyl-benzenesulfonic acid sodium salt). In acidic solutions hydrogenation of 1 yields a mixture of cis-mer- and cis-fac-[RhClH2(tppms)3] (3a,b) while in strongly basic solutions [RhH(H2O)(tppms)3] (4) is obtained, the midpoint of the equilibrium between these hydride species being at pH 8.2. The paper gives the first successful 1H and 31P NMR spectroscopic characterization of a water soluble rhodium(I)-monohydride (4) bearing only monodentate phosphine ligands. Hydrolysis of 2 is negligible below pH 9 and its hydrogenation results in formation of [Rh(CO)H(tppms)3] (5), which is an analogue to the well known and industrially used hydroformylation catalyst [Rh(CO)H(tppts)3] (6) (TPPTS = 3,3',3''-phosphinetriyltris(benzenesulfonic acid) trisodium salt). It was shown by pH-potentiometric measurements that formation of 5 is strongly pH dependent in the pH 5-9 range, this gives an explanation for the observed but previously unexplained pH dependence of several hydroformylation reactions. Conversely, the effect of pH on the rate of hydrogenation of maleic and fumaric acid catalyzed by 1 in the 2 < pH < 7 range can be adequately described by considering solely the changes in the ionization state of these substrates. All these results warrant the use of buffered (pH-controlled) solutions for aqueous organometallic catalysis.

2.
Peptides ; 21(4): 491-501, 2000 Apr.
Article in English | MEDLINE | ID: mdl-10822104

ABSTRACT

Ziconotide is a selective peptide antagonist of the N-type calcium channel currently in clinical trials for analgesia. Ziconotide reached a maximal brain concentration of between 0.003 and 0.006% of the injected material per gram of tissue at 3-20 min after i.v. injection, and this decayed to below 0.001%/g after 2 h. The structurally distinct conopeptide SNX-185 (synthetic TVIA) was considerably more persistent in brain after i.v. administration, with 0.0035% of the injected material present at 2-4 h after i.v. injection, and 0.0015% present at 24 h. Similar results (i.e. greater persistence of SNX-185) were obtained when the peptides were perfused through in vivo dialysis probes implanted into the hippocampus. Image analysis and serial sectioning showed that diffusion of Ziconotide in the extracellular fluid around the dialysis probe was minimal, with the peptide located within 1 mm of the probe after 2 h. In vitro diffusion through cultured bovine brain microvessel endothelial cells (BBMEC) verified that a close structural analog of Ziconotide (SNX-194) passed through this blood-brain barrier (BBB) model as expected for peptides of similar physical properties (permeability coefficient of 6.5 x 10(-4) cm/g). Passage from blood to brain was also verified by in situ perfusion through the carotid artery. A statistically greater amount of radioactivity was found to cross the BBB after perfusion of radioiodinated Ziconotide compared to [14C]inulin. Capillary depletion experiments and HPLC analysis defined the brain location and stability.


Subject(s)
Brain/metabolism , Calcium Channel Blockers/pharmacokinetics , Neuroprotective Agents/pharmacokinetics , omega-Conotoxins/pharmacokinetics , Amino Acid Sequence , Animals , Biological Availability , Blood-Brain Barrier , Calcium Channel Blockers/blood , Cattle , Chromatography, High Pressure Liquid , Diffusion , Endothelium, Vascular/metabolism , Extracellular Space/metabolism , In Vitro Techniques , Injections, Intravenous , Microdialysis , Molecular Sequence Data , Neuroprotective Agents/blood , Perfusion , Rats , Rats, Sprague-Dawley , omega-Conotoxins/blood
3.
Inorg Chem ; 39(22): 5083-8, 2000 Oct 30.
Article in English | MEDLINE | ID: mdl-11233205

ABSTRACT

The water-soluble tertiary phosphine complex of ruthenium(II), [RuCl2(PTA)4], (PTA = 1,3,5-triaza-7-phosphaadamantane) was used as catalyst precursor for hydrogenation of CO2 and bicarbonate in aqueous solution, in the absence of amine or other additives, under mild conditions. Reaction of [RuCl2(PTA)4] and H2 (60 bar) gives the hydrides [RuH2(PTA)4] (at pH = 12.0) and [RuH(PTA)4X] (X = Cl- or H2O) (at pH = 2.0). In presence of excess PTA, formation of the unparalleled cationic pentakis-phosphino species, [HRu(PTA)5]+, was unambiguously established by 1H and 31P NMR measurements. The same hydrides were observed when [Ru(H2O)6][tos]2 (tos = toluene-4-sulfonate) reacted with PTA under H2 pressure. The rate of CO2 hydrogenation strongly depends on the pH. The highest initial reaction rate (TOF = 807.3 h(-1)) was determined for a 10% HCO3-/90% CO2 mixture (pH = 5.86), whereas the reduction was very slow both at low and high pH (CO2 and Na2CO3 solutions, respectively). 1H and 31P NMR studies together with the kinetic measurements suggested that HCO3- was the real substrate and [RuH(PTA)4X] the catalytically active hydride species in this reaction. Hydrogenation of HCO3- showed an induction period which could be ascribed to the slow formation of the catalytically active hydride species.

4.
Biochemistry ; 37(44): 15353-62, 1998 Nov 03.
Article in English | MEDLINE | ID: mdl-9799496

ABSTRACT

We describe the first potent and selective blocker of the class E Ca2+channel. SNX-482, a novel 41 amino acid peptide present in the venom of the African tarantula, Hysterocrates gigas, was identified through its ability to inhibit human class E Ca2+ channels stably expressed in a mammalian cell line. An IC50 of 15-30 nM was obtained for block of the class E Ca2+ channel, using either patch clamp electrophysiology or K+-evoked Ca2+ flux. At low nanomolar concentrations, SNX-482 also blocked a native resistant or R-type Ca2+ current in rat neurohypophyseal nerve terminals, but concentrations of 200-500 nM had no effect on R-type Ca2+ currents in several types of rat central neurons. The peptide has the sequence GVDKAGCRYMFGGCSVNDDCCPRLGCHSLFSYCAWDLTFSD-OH and is homologous to the spider peptides grammatoxin S1A and hanatoxin, both peptides with very different ion channel blocking selectivities. No effect of SNX-482 was observed on the following ion channel activities: Na+ or K+ currents in several cultured cell types (up to 500 nM); K+ current through cloned potassium channels Kv1.1 and Kv1. 4 expressed in Xenopus oocytes (up to 140 nM); Ca2+ flux through L- and T-type Ca2+ channels in an anterior pituitary cell line (GH3, up to 500 nM); and Ba2+ current through class A Ca2+ channels expressed in Xenopus oocytes (up to 280 nM). A weak effect was noted on Ca2+ current through cloned and stably expressed class B Ca2+ channels (IC50 > 500 nM). The unique selectivity of SNX-482 suggests its usefulness in studying the diversity, function, and pharmacology of class E and/or R-type Ca2+ channels.


Subject(s)
Calcium Channel Blockers/chemistry , Peptides/chemistry , Spider Venoms/chemistry , Amino Acid Sequence , Animals , Calcium Channel Blockers/isolation & purification , Calcium Channel Blockers/pharmacology , Calcium Channels/physiology , Cell Line , Humans , Male , Molecular Sequence Data , Oocytes/physiology , Patch-Clamp Techniques , Peptides/isolation & purification , Peptides/physiology , Potassium Channel Blockers , Rats , Rats, Sprague-Dawley , Sodium Channel Blockers , Spider Venoms/isolation & purification , Spider Venoms/pharmacology , Transfection , Tumor Cells, Cultured , Xenopus
5.
Proc Natl Acad Sci U S A ; 95(7): 3513-8, 1998 Mar 31.
Article in English | MEDLINE | ID: mdl-9520397

ABSTRACT

The fluidity of Synechocystis membranes was adjusted in vivo by temperature acclimation, addition of fluidizer agent benzyl alcohol, or catalytic lipid hydrogenation specific to plasma membranes. The reduced membrane physical order in thylakoids obtained by either downshifting growth temperature or administration of benzyl alcohol was paralleled with enhanced thermosensitivity of the photosynthetic membrane. Simultaneously, the stress-sensing system leading to the cellular heat shock (HS) response also has been altered. There was a close correlation between thylakoid fluidity levels, monitored by steady-state 1,6-diphenyl-1,3,5-hexatriene anisotropy, and threshold temperatures required for maximal activation of all of the HS-inducible genes investigated, including dnaK, groESL, cpn60, and hsp17. The causal relationship between the pre-existing thylakoid physical order and temperature set point of both the transcriptional activation and the de novo protein synthesis was the most striking for the 17-kDa HS protein (HSP17) associated mostly with the thylakoid membranes. These findings together with the fact that the in vivo modulation of lipid saturation within cytoplasmic membrane had no effect on HS response suggest that thylakoid acts as a cellular thermometer where thermal stress is sensed and transduced into a cellular signal leading to the activation of HS genes.


Subject(s)
Cell Membrane/chemistry , Cell Membrane/metabolism , Eukaryota/genetics , Eukaryota/metabolism , Heat-Shock Proteins/genetics , Heat-Shock Proteins/metabolism , Membrane Fluidity/genetics , Signal Transduction/genetics , Cell Membrane/genetics , Gene Expression Regulation, Plant
6.
Naunyn Schmiedebergs Arch Pharmacol ; 357(2): 159-68, 1998 Feb.
Article in English | MEDLINE | ID: mdl-9521489

ABSTRACT

The ability of a series of synthetic analogues of omega-conopeptides MVIIA (SNX-111) and TVIA (SNX-185) to prevent electrically-evoked norepinephrine release from rat tail artery and hippocampal slice preparations was determined in an effort to identify voltage-sensitive calcium channel (VSCC) blockers that selectively target N-type VSCCs in central nervous system tissue. Electrical field stimulation (3 Hz, 1 ms in duration. 80 V for 1 min) caused a high and consistent tritium outflow from rat tail artery and hippocampal slice preparations preloaded with [3H]-norepinephrine. All conopeptides, chosen for their selective affinities for high-affinity SNX-111 binding sites (i.e., N-type VSCCs) over high-affinity omega-conopeptides MVIIC (SNX-230) binding sites (i.e., P/Q-type VSCCs), produced a concentration-dependent inhibition of calcium dependent electrically-evoked tritium outflow from both tail arteries and hippocampal slices: IC50s ranged from 1.2 nM to 1.2 microM. Blocking potencies (IC50s) in the tail artery assay were significantly correlated with those measured in the hippocampal slice preparation (r = 0.91, P = 0.00000012). There was a significant correlation between IC50s for blockade of hippocampal norepinephrine release and the inhibition of high-affinity [125I]-SNX-I11 binding in rat brain synaptosomes (r = 0.76, P = 0.00028). Blockade of hippocampal norepinephrine release was not significantly correlated with the inhibition of high-affinity SNX-230 binding (r = 0.46, P = 0.056). Maximum inhibition of tritium outflow in the tail artery assay was 22+/-1.4% of control, approximating the value (20.9+/-16.0% of control) obtained in the absence of extracellular Ca2+. In contrast, the maximum inhibition of tritium release from hippocampal slices was 36.8+/-2.5% of control (P < 0.05, compared to that of the tail artery assay). These results suggest that (1) N-type VSCCs alone mediate low frequency electrical stimulation-evoked neurotransmitter release from peripheral sympathetic efferents (tail artery) while both N-type and non-N type(s) mediate neurotransmitter release from CNS neurons (hippocampus); and (2) analogues of omega-conopeptides MVIIA and TVIA do not differentiate between N-type VSCCs mediating norepinephrine release from central and peripheral neural tissues.


Subject(s)
Calcium Channel Blockers/pharmacology , Central Nervous System/drug effects , Peripheral Nervous System/drug effects , Animals , Arteries/drug effects , Arteries/metabolism , Blood Pressure/drug effects , Decerebrate State/physiopathology , Electric Stimulation , Electrophysiology , Hippocampus/drug effects , Hippocampus/metabolism , In Vitro Techniques , Male , Muscle, Smooth, Vascular/drug effects , Muscle, Smooth, Vascular/metabolism , Norepinephrine/metabolism , Radioligand Assay , Rats , Rats, Sprague-Dawley , Synaptosomes/drug effects , Synaptosomes/metabolism
7.
J Physiol ; 502 ( Pt 2): 351-63, 1997 Jul 15.
Article in English | MEDLINE | ID: mdl-9263915

ABSTRACT

1. The nerve endings of rat neurohypophyses were acutely dissociated and a combination of pharmacological, biophysical and biochemical techniques was used to determine which classes of Ca2+ channels on these central nervous system (CNS) terminals contribute functionally to arginine vasopressin (AVP) and oxytocin (OT) secretion. 2. Purified neurohypophysial plasma membranes not only had a single high-affinity binding site for the N-channel-specific omega-conopeptide MVIIA, but also a distinct high-affinity site for another omega-conopeptide (MVIIC), which affects both N- and P/Q-channels. 3. Neurohypophysial terminals exhibited, besides L- and N-type currents, another component of the Ca2+ current that was only blocked by low concentrations of MVIIC or by high concentrations of omega-AgaIVA, a P/Q-channel-selective spider toxin. 4. This Ca2+ current component had pharmacological and biophysical properties similar to those described for the fast-inactivating form of the P/Q-channel class, suggesting that in the neurohypophysial terminals this current is mediated by a 'Q'-type channel. 5. Pharmacological additivity studies showed that this Q-component contributed to rises in intraterminal Ca2+ concentration ([Ca2+]i) in only half of the terminals tested. 6. Furthermore, the non-L- and non-N-component of Ca(2+)-dependent AVP release, but not OT release, was effectively abolished by the same blockers of Q-type current. 7. Thus Q-channels are present on a subset of the neurohypophysial terminals where, in combination with N- and L-channels, they control AVP but not OT peptide neurosecretion.


Subject(s)
Arginine Vasopressin/metabolism , Calcium Channels/physiology , Pituitary Gland, Posterior/physiology , omega-Conotoxins , Animals , Calcium/metabolism , Calcium Channel Blockers/pharmacology , Calcium Channels/drug effects , Cattle , Cell Membrane/physiology , Membrane Potentials/drug effects , Mice , Nerve Endings/physiology , Oxytocin/metabolism , Peptides/pharmacology , Pituitary Gland, Posterior/drug effects , Pituitary Gland, Posterior/metabolism , Rats , Spider Venoms/pharmacology , omega-Agatoxin IVA
8.
J Neurosci ; 17(9): 3002-13, 1997 May 01.
Article in English | MEDLINE | ID: mdl-9096136

ABSTRACT

The selective block of N-type Ca2+ channels by omega-conotoxins has been a hallmark of these channels, critical in delineating their biological roles and molecular characteristics. Here we report that the omega-conotoxin-channel interaction depends strongly on channel gating. N-type channels (alpha1B, alpha2, and beta1) expressed in Xenopus oocytes were blocked with a variety of omega-conotoxins, including omega-CTx-GVIA, omega-CTx-MVIIA, and SNX-331, a derivative of omega-CTx-MVIIC. Changes in holding potential (HP) markedly altered the severity of toxin block and the kinetics of its onset and removal. Notably, strong hyperpolarization renders omega-conotoxin block completely reversible. These effects could be accounted for by a modulated receptor model, in which toxin dissociation from the inactivated state is approximately 60-fold slower than from the resting state. Because omega-conotoxins act exclusively outside cells, our results suggest that voltage-dependent inactivation of Ca2+ channels must be associated with an externally detectable conformational change.


Subject(s)
Calcium Channel Blockers/pharmacology , Calcium Channels/drug effects , Peptides/pharmacology , omega-Conotoxins , Animals , Dose-Response Relationship, Drug , Female , Xenopus laevis , omega-Conotoxin GVIA
9.
Int J Pept Protein Res ; 46(3-4): 320-5, 1995.
Article in English | MEDLINE | ID: mdl-8537186

ABSTRACT

Synthetic versions of seven naturally occurring omega-conopeptides were subjected to structural analyses in order to determine their disulfide bridge pattern. The method applied in this study uses a combination of amino-acid composition and peptide sequence analysis of various peptide fragments generated by different enzymatic digestions. A temperature modification in the Edman degradation cycles of a protein sequencer allowed the unambiguous detection of the cleavage of cystine residues. The appearance of the cystine residues in particular cycles of the sequence analysis was characteristic of one or several of the theoretically possible 15 isomers. In the case of multiple choices, possible isomers were further eliminated by the amino-acid and sequence analysis of peptide fragments generated by the enzymatic digestion. All synthetic peptides, SNX-111, -157, -159, -183, -185, -230 and -231, were found to have the same disulfide bridge pattern as determined for the naturally occurring omega-conopeptide G-VI-A, i.e. disulfide bridges between the half-cystines 1-16, 8-20 and 15-25 (using the amino-acid numbering of SNX-111).


Subject(s)
Disulfides/chemistry , Mollusk Venoms/chemistry , Peptide Fragments/chemistry , Peptides/chemistry , omega-Conotoxins , Amino Acid Sequence , Calcium Channel Blockers/chemistry , Chromatography, High Pressure Liquid , Cystine/chemistry , Endopeptidases/metabolism , Molecular Sequence Data , Peptide Fragments/metabolism , Peptides/metabolism
10.
FEBS Lett ; 370(3): 163-9, 1995 Aug 21.
Article in English | MEDLINE | ID: mdl-7656969

ABSTRACT

The solution structure of omega-conotoxin MVIIA (SNX-111), a peptide toxin from the fish hunting cone snail Conus magus and a high-affinity blocker of N-type calcium channels, was determined by 2D NMR spectroscopy. The backbones of the best 44 structures match with an average pairwise RMSD of 0.59 angstroms. The structures contain a short segment of triple-stranded beta-sheet involving residues 6-8, 20-21, and 24-25. The structure of this toxin is very similar to that of omega-conotoxin GVIA with which is has only 40% sequence homology, but very similar calcium channel binding affinity and selectivity.


Subject(s)
Magnetic Resonance Spectroscopy/methods , Peptides/chemistry , omega-Conotoxins , Amino Acid Sequence , Animals , Calcium Channel Blockers/metabolism , Chemical Phenomena , Chemistry, Physical , Hydrogen/chemistry , Models, Molecular , Molecular Sequence Data , Peptides/chemical synthesis , Peptides/metabolism , Protein Conformation , Sequence Alignment , Sequence Homology, Amino Acid , Snails/chemistry , Software , omega-Conotoxin GVIA
11.
Biochemistry ; 34(26): 8341-7, 1995 Jul 04.
Article in English | MEDLINE | ID: mdl-7541240

ABSTRACT

A novel selective calcium channel antagonist peptide, SNX-325, has been isolated from the venom of the spider Segestria florentina. The peptide was isolated using as bioassays the displacement of radioiodinated omega-conopeptide SNX-230 (MVIIC) from rat brain synaptosomal membranes, as well as the inhibition of the barium current through cloned expressed calcium channels in oocytes. The primary sequence of SNX-325 is GSCIESGKSCTHSRSMKNGLCCPKSRCNCRQIQHRHDYLGKRKYSCRCS, which is a novel amino acid sequence. Solid-phase synthesis resulted in a peptide that is chromatographically identical with the native peptide and which has the same configuration of cysteine residues as the spider venom peptide omega-Aga-IVa [Mintz, I. M., et al., (1992) Nature 355, 827-829]. At micromolar concentrations, SNX-325 is an inhibitor of most calcium, but not sodium or potassium, currents. At nanomolar concentrations, SNX-325 is a selective blocker of the cloned expressed class B (N-type), but not class C (cardiac L), A, or E, calcium channels. SNX-325 is approximately equipotent with the N-channel selective omega-conopeptides (GVIA and MVIIA as well as closely related synthetic derivatives) in blocking the potassium induced release of tritiated norepinephrine from hippocampal slices (IC50s, 0.1-0.5 nM) and in blocking the barium current through cloned expressed N-channels in oocytes (IC50s 3-30 nM). By contrast, SNX-325 is 4-5 orders of magnitude less potent than is SNX-111 (synthetic MVIIA) at displacing radioiodinated SNX-111 from rat brain synaptosomal membranes. SNX-325 will be a useful comparative tool in further defining the function and pharmacology of the N- and possibly other types of high-voltage activated calcium channels.


Subject(s)
Calcium Channel Blockers/chemistry , Calcium Channels/physiology , Peptides/chemistry , Spider Venoms , omega-Conotoxins , Amino Acid Sequence , Animals , Binding, Competitive , Brain/metabolism , Calcium Channel Blockers/isolation & purification , Calcium Channels/drug effects , Carboxypeptidases , Cathepsin A , Chymotrypsin , Female , Iodine Radioisotopes , Ion Channels/drug effects , Ion Channels/physiology , Molecular Sequence Data , Oocytes/drug effects , Oocytes/physiology , Organ Specificity , Peptide Fragments/chemistry , Peptide Fragments/isolation & purification , Peptides/isolation & purification , Peptides/metabolism , Peptides/pharmacology , Rats , Sequence Homology, Amino Acid , Spiders , Synaptosomes/metabolism , Xenopus
12.
Biochemistry ; 34(25): 8076-81, 1995 Jun 27.
Article in English | MEDLINE | ID: mdl-7794920

ABSTRACT

The synthetic peptide SNX-111 corresponding to the sequence of the omega-conopeptide MVIIA from the venom of the marine snail Conus magus is a highly potent and selective antagonist of N-type calcium channels. We have synthesized and characterized a large number of analogs of SNX-111 in order to elucidate the structural features of the peptide involved in blocking N-type calcium channels. Comparison of the binding of SNX-111 and its analogs to rat brain synaptosomal membranes rich in N-type channels revealed that, among the four lysines and two arginines in the molecule, lysine in position 2 and arginines at position 10 and 21 are important for the interaction of SNX-111 with N-type channels. The importance of the middle segment from residues 9 through 14 for this binding interaction was revealed by substitution of the individual residues as well as by the construction of hybrid peptides in which the residues 9-12 in SNX-111 and another conopeptide, SNX-183, corresponding to a peptide SVIB from Conus striatus, were interchanged. Introduction of the sequence SRLM from SNX-111 in place of RKTS in position 9-12 in SNX-183 resulted in a 38-fold increase in affinity.


Subject(s)
Calcium Channel Blockers/chemistry , Neurons/chemistry , Peptides/chemistry , Peptides/metabolism , omega-Conotoxins , Amino Acid Sequence , Binding Sites , Brain/metabolism , Calcium/metabolism , Calcium Channel Blockers/metabolism , Calcium Channels/metabolism , Molecular Sequence Data , Peptides/chemical synthesis , Protein Conformation , Structure-Activity Relationship , Synaptic Membranes/metabolism
13.
J Mol Biol ; 248(1): 106-24, 1995 Apr 21.
Article in English | MEDLINE | ID: mdl-7731037

ABSTRACT

We have determined the solution structure of the omega-conotoxin MVIIC from Conus magus by 1H NMR. This conopeptide preferentially blocks P and Q type Ca2+ currents by binding with high affinity to voltage-sensitive Ca2+ channels in neurons. This 26 residue peptide with three disulfide bonds was chemically synthesized and refolded for NMR structural studies. The 1H NMR NOESY spectrum of this peptide was completely assigned, with stereospecific assignments made for 12 of the beta prochiral centers. Complete relaxation matrix analysis using MARDIGRAS was used to obtain initial interproton distances from peak intensities. The correlation time necessary for these calculations was determined by measuring 13C relaxation times using inversely detected natural abundance spectra. Distances were input to DG, which provided 15 starting structures which were then subjected to restrained molecular dynamics calculations using SANDER with the AMBER 91 force field in vacuo. 1H-1H vicinal coupling constants were obtained using a combination of line fitting of both E. COSY and NOESY spectra and used to generate angle restraints that were included explicitly in the restrained molecular dynamics calculations. The final set of the 15 best structures had a backbone rmsd of 0.84 A. The ensemble R1/6 factor calculated by CORMA for the final 15 structures was 11%. The final structure consists of an anti-parallel, triple-stranded beta-sheet, with four turns. In spite of significant differences in amino acid sequence and affinities for calcium channel subtypes, the backbone structure of omega-conotoxin MVIIC is very similar to the previously reported structure of omega-conotoxin GVIA.


Subject(s)
Calcium Channel Blockers/chemistry , Calcium Channels/metabolism , Peptides/chemistry , Protein Conformation , omega-Conotoxins , Amino Acid Sequence , Animals , Hydrogen , Hydrogen Bonding , Magnetic Resonance Spectroscopy/methods , Models, Molecular , Molecular Sequence Data , Mollusca , Mollusk Venoms/chemistry , Peptides/isolation & purification , Peptides/metabolism , Sequence Homology, Amino Acid , Solutions , omega-Conotoxin GVIA
14.
J Pharmacol Exp Ther ; 273(1): 248-56, 1995 Apr.
Article in English | MEDLINE | ID: mdl-7714772

ABSTRACT

This investigation assessed the ability of a variety of calcium channel blocking peptides to block synaptic transmission in the isolated mouse phrenic nerve-hemidiaphragm. The synthetic version of the naturally occurring N-type voltage-sensitive calcium channel (VSCC) blocker omega-conopeptide MVIIA (SNX-111) had no effect on nerve-evoked muscle contractions. The non-N-, non-L-type VSCC blocker, omega-conopeptide MVIIC (SNX-230), blocked neuromuscular transmission completely, as did the selective P-type VSCC blocker, omega-Aga-IVA. Subsequent evaluation of other synthetic omega-conopeptides and analogs disclosed a significant positive correlation between the test compounds' affinities for high-affinity SNX-230 brain binding sites and their neuromuscular blocking potencies. Quantal analysis of transmitter release showed that SNX-230 abolished evoked endplate potentials completely, but had little effect on the amplitude and frequency of spontaneous miniature endplate potentials. Perineural focal recordings of presynaptic currents showed that SNX-230 did not block the neuronal action potential. These and other findings indicated that SNX-230 prevents transmitter release at the mouse neuromuscular junction by blocking calcium channels at presynaptic nerve endings. These calcium channels correspond pharmacologically to VSCCs associated with high-affinity binding sites in rat brain and are most probably either of the P- or Q-type.


Subject(s)
Calcium Channel Blockers/pharmacology , Neuromuscular Junction/drug effects , Peptides/pharmacology , Spider Venoms/pharmacology , omega-Conotoxins , Amino Acid Sequence , Animals , Dose-Response Relationship, Drug , In Vitro Techniques , Male , Mice , Molecular Sequence Data , Muscle Contraction/drug effects , Neuromuscular Junction/physiology , omega-Agatoxin IVA
15.
Mol Cell Neurosci ; 5(3): 219-28, 1994 Jun.
Article in English | MEDLINE | ID: mdl-8087420

ABSTRACT

The interaction of two synthetic omega-conopeptides SNX-111 (MVIIA) and SNX-230 (MVIIC) both derived from the marine snail Conus magus, with non-L-type neuronal voltage-sensitive calcium channels (VSCC) in rat brain synaptosomal preparations has been investigated with the aid of well-characterized 125I derivatives of the two peptides. To assess the effects of iodination on the binding characteristics of SNX-111 and SNX-230, the corresponding peptides containing monoiodotyrosine in place of tyrosine, namely, SNX-259 ([127I]SNX-111) and SNX-260 ([127I]SNX-230), respectively, were prepared by solid-phase synthesis. Saturation analysis showed that [125I]SNX-111 and [125I]SNX-230 bound to two distinct classes of high-affinity sites with apparent Kd's of 9 and 11 pM and Bmax's of 0.54 and 2.2 pmol/mg protein, respectively. Kinetic analysis revealed that both peptides exhibited high association rates as well as rapid dissociation rates in contrast to the 125I derivative of the synthetic omega-conopeptide from Conus geographus, GVIA (SNX-124), which binds irreversibly to N-type channels in rat brain synaptosomes. Competition binding experiments with [125I]SNX-111 and [125I]SNX-124 established that both of them bind to the same site, namely, N-type VSCC. The site detected by the binding of [125I]SNX-230 is distinct from N-type VSCC since SNX-111 has very low affinity (K(i) = 135 nM) in competition studies. Recent findings that a novel high-voltage-activated calcium channel in rat cerebellar granule neurons is resistant to blockers of L-, N-, and P-type VSCC but is highly sensitive to SNX-230 suggest that the [125I]SNX-230 binding site may represent this novel type of calcium channel or another, as yet undescribed, VSCC.


Subject(s)
Calcium Channel Blockers/metabolism , Calcium Channels/metabolism , Neurons/metabolism , Peptides/metabolism , omega-Conotoxins , Animals , Binding Sites , Binding, Competitive , Calcium/pharmacology , Calcium Channel Blockers/pharmacology , Cations, Monovalent/pharmacology , Dizocilpine Maleate/pharmacology , Male , Methionine/analogs & derivatives , Mollusk Venoms/metabolism , Monoiodotyrosine/chemistry , Peptides/chemical synthesis , Protein Binding/drug effects , Rats , Rats, Sprague-Dawley , Synaptosomes/metabolism
16.
Biochemistry ; 32(29): 7396-405, 1993 Jul 27.
Article in English | MEDLINE | ID: mdl-8338837

ABSTRACT

We report here the solution structure of omega-conotoxin GVIA, a peptide antagonist of the N-type neuronal voltage-sensitive calcium channel. The structure was determined using two-dimensional NMR in combination with distance geometry and restrained molecular dynamics. The full relaxation matrix analysis program MARDIGRAS was used to generate maximum and minimum distance restraints from the crosspeak intensities in NOESY spectra. The 187 restraints obtained were used in conjunction with 23 angle restraints from vicinal coupling constants as input for the structure calculations. The backbones of the best 21 structures match with an average pairwise RMSD of 0.58 A. The structures contain a short segment of triple-stranded beta-sheet involving residues 6-8, 18-21, and 24-27, making this the smallest published peptide structure to contain a triple-stranded beta-sheet. Conotoxins have been shown to be effective neuroprotective agents in animal models of brain ischemia. Our results should aid in the design of novel nonpeptide compounds with potential therapeutic utility.


Subject(s)
Magnetic Resonance Spectroscopy , Peptides/chemistry , Chemical Phenomena , Chemistry, Physical , Chromatography, High Pressure Liquid , Disulfides/chemistry , Models, Molecular , Molecular Structure , Protein Structure, Secondary , Software , Solutions , omega-Conotoxin GVIA
17.
Biochemistry ; 31(41): 9919-26, 1992 Oct 20.
Article in English | MEDLINE | ID: mdl-1390774

ABSTRACT

Three neurotoxic peptides from the venom of Conus striatus have been purified, biochemically characterized, and chemically synthesized. One of these, an acetylcholine receptor blocker designated alpha-conotoxin SII, has the sequence GCCCNPACGPNYGCGTSCS. In contrast to all other alpha-conotoxins, SII has three disulfide bonds (instead of two), has no net positive charge, and has a free C-terminus. The other two paralytic peptides are Ca channel-targeted omega-conotoxins, SVIA and SVIB. omega-SVIA is the smallest natural omega-conotoxin so far characterized and has the sequence CRSSGSPCGVTSICCGRCYRGKCT-NH2. Although omega-conotoxin SVIA is a potent paralytic toxic in lower vertebrate species, it was much less effective in mammals. The third toxin, omega-conotoxin SVIB, has the sequence CKLKGQSCRKTSYDCCSGSCGRSGKC-NH2. This peptide has a different pharmacological specificity from other omega-conotoxins previously purified from Conus venoms; only omega-conotoxin SVIB has proven to be lethal to mice upon ic injection. Binding competition experiments with rat brain synaptosomal membranes indicate that the high-affinity binding site for omega-conotoxin SVIB is distinct from the high-affinity omega-conotoxin GVIA or MVIIA site.


Subject(s)
Conotoxins , Mollusk Venoms/chemistry , Peptides/chemistry , omega-Conotoxins , Amino Acid Sequence , Animals , Binding Sites , Binding, Competitive , Brain/metabolism , Bungarotoxins/metabolism , Mice , Molecular Sequence Data , Neuromuscular Junction/physiology , Peptides/metabolism , Peptides/pharmacology , Rana pipiens , Rats , Receptors, Cholinergic/physiology , Spectrometry, Mass, Fast Atom Bombardment , Synaptic Membranes/metabolism
18.
Neuron ; 9(1): 69-77, 1992 Jul.
Article in English | MEDLINE | ID: mdl-1352986

ABSTRACT

Voltage-sensitive Ca2+ channels that control neurotransmitter release are blocked by omega-conotoxin (omega-CgTx) GVIA from the marine snail Conus geographus, the most widely used inhibitor of neurotransmitter release. However, many mammalian synapses are omega-CgTx-GVIA insensitive. We describe a new Conus peptide, omega-CgTx-MVIIC, that is an effective inhibitor of omega-CgTx-GVIA-resistant synaptic transmission. Ca2+ channel targets that are inhibited by omega-CgTx-MVIIC but not by omega-CgTx-GVIA include those mediating depolarization-induced 45Ca2+ uptake in rat synaptosome preparations, "P" currents in cerebellar Purkinje cells, and a subset of omega-CgTx-GVIA-resistant currents in CA1 hippocampal pyramidal cells. The characterization of omega-CgTx-MVIIC by a combination of molecular genetics and chemical synthesis defines a general approach for obtaining ligands with novel receptor subtype specificity from Conus.


Subject(s)
Calcium Channels/drug effects , Peptides/pharmacology , Synapses/ultrastructure , omega-Conotoxins , Amino Acid Sequence , Animals , Base Sequence , Calcium Channel Blockers/pharmacology , Calcium Channels/metabolism , Calcium Channels/physiology , Cells, Cultured , DNA/analysis , DNA/genetics , Hippocampus/cytology , Hippocampus/drug effects , Hippocampus/ultrastructure , Ligands , Molecular Sequence Data , Neurons/cytology , Neurons/drug effects , Neurons/ultrastructure , Neurotransmitter Agents/antagonists & inhibitors , Neurotransmitter Agents/metabolism , Peptides/genetics , Peptides/metabolism , Peptides, Cyclic/pharmacology , Purkinje Cells/cytology , Purkinje Cells/drug effects , Purkinje Cells/ultrastructure , Rats , Snails , Synapses/drug effects , Synapses/metabolism , omega-Conotoxin GVIA
19.
J Cardiovasc Pharmacol ; 20(5): 756-64, 1992.
Article in English | MEDLINE | ID: mdl-1280738

ABSTRACT

We examined the effects of omega-conopeptides, a novel class of neuronal voltage-gated calcium channel antagonists, on hemodynamic responses in rats. Intravenous (i.v.) injections of SNX-111 (omega-conopeptide MVIIA) dose-dependently decreased arterial blood pressure (BP) in conscious rats. Intracerebroventricular (i.c.v.) SNX-111 injections (580 pmol) tended to increase BP and, after an initial decrease, to increase heart rate (HR). The dose-response curve for SNX-111 administered i.v. in conscious rats was biphasic. Beginning at subdepressor doses, SNX-111 caused a long-lasting blockade of pressor responses elicited by sympathetic nerve stimulation in pithed animals but did not prevent increases in BP evoked by exogenously administered norepinephrine (NE). Pretreatment of rats with histamine antagonists partially blocked the hypotensive effects of the higher (870 and 2,900 nmol/kg) doses of SNX-111. Substitution of alanine for arginine at position 10 ([Ala10]-MVIIA) markedly attenuated the histamine-mediated component of the vasodepressor response. Together, these findings indicate that SNX-111 administered i.v. decreases systemic BP by a combination of blockade of sympathetic neurotransmission and mast cell degranulation; the latter function appears to be dependent on the arginine residue in position 10 of the amino acid sequence of SNX-111.


Subject(s)
Blood Pressure/drug effects , Calcium Channel Blockers/pharmacology , Heart Rate/drug effects , Peptides, Cyclic/pharmacology , Peptides/pharmacology , omega-Conotoxins , Amino Acid Sequence , Animals , Chlorpheniramine/pharmacology , Cimetidine/pharmacology , Decerebrate State , Dose-Response Relationship, Drug , Injections, Intraventricular , Male , Molecular Sequence Data , Norepinephrine/pharmacology , Peptides/administration & dosage , Peptides, Cyclic/administration & dosage , Rats , Rats, Sprague-Dawley
20.
Pharm Res ; 7(12): 1213-21, 1990 Dec.
Article in English | MEDLINE | ID: mdl-1965663

ABSTRACT

The G protein coupled receptors (GPC-Rs) comprise a large superfamily of genes encoding numerous receptors which all show common structural features, e.g., seven putative membrane spanning domains. Their biological functions are extremely diverse, ranging from vision and olfaction to neuronal and endocrine signaling. The GPC-Rs couple via multiple G proteins to a growing number of recognized second messenger pathway, e.g., cAMP and phosphatidyl inositol turnover. This review summarizes our current knowledge of the molecular mechanisms by which the GPC-Rs activate second messenger systems, and it addresses their regulation and structure.


Subject(s)
GTP-Binding Proteins/genetics , Receptors, Adrenergic, alpha/genetics , Receptors, Cell Surface/genetics , Receptors, Muscarinic/genetics , Receptors, Neurotransmitter/genetics , Signal Transduction , Amino Acid Sequence , Base Sequence , Cyclic AMP/metabolism , GTP-Binding Proteins/metabolism , GTP-Binding Proteins/physiology , Humans , Molecular Sequence Data , Phosphatidylinositols/metabolism , Protein Binding , Receptors, Adrenergic, alpha/metabolism , Receptors, Adrenergic, alpha/physiology , Receptors, Cell Surface/metabolism , Receptors, Cell Surface/physiology , Receptors, Muscarinic/metabolism , Receptors, Muscarinic/physiology , Receptors, Neurotransmitter/metabolism , Receptors, Neurotransmitter/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...