Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 53
Filter
Add more filters










Publication year range
1.
Xenobiotica ; 41(3): 198-211, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21070145

ABSTRACT

1. PAP-1 (5-(4-phenoxybutoxy)psoralen), a potent small-molecule blocker of the voltage-gated potassium Kv1.3 channel, is currently in preclinical development for psoriasis. This study was undertaken to identify the major phase I metabolites of PAP-1 in Sprague-Dawley (SD) rats. 2. Five phase I metabolites, that is 5-(oxybutyric-acid)psoralen (M1), 5-[4-(4-hydroxybutoxy)]psoralen (M2), 5-[4-(4-hydroxyphenoxy)butoxy]psoralen (M3), 5-[4-(3-hydroxyphenoxy)butoxy]psoralen (M4), and 8-hydroxyl-5-(4-phenoxybutoxy)psoralen (M5), were isolated from the bile of rats and identified by mass spectrometry and NMR spectroscopy. The last four metabolites are new compounds. 3. Incubation of PAP-1 with SD rat liver microsomes rendered the same five major metabolites in a nicotinamide adenine dinucleotide phosphate (NADPH)-dependent manner suggesting that cytochrome P450 (CYP) enzymes are involved in PAP-1 metabolism. Inhibitors of rat CYP1A1/2 (alpha-naphthoflavone) and CYP3A (ketoconazole) but not CYP2D6 (quinidine), CYP2E (diethyldithiocarbamate), or CYP2C9 (sulphaphenazole) blocked the metabolism of PAP-1 in rat microsomes. 4. Of the five metabolites M3, M4, and M5 were found to inhibit Kv1.3 currents with nanomolar IC50s, while M1 and M2 were inactive. Our results identified the Kv1.3-inactive M1 as the major phase I metabolite, and suggest that hydroxylation and O-dealkylation are the major pathways of PAP-1 metabolism. 5. We further conducted a 6-month repeat-dose toxicity study with PAP-1 at 50 mg/kg in both male and female Lewis rats and did not observe any toxic effects.


Subject(s)
Ficusin/toxicity , Kv1.3 Potassium Channel/metabolism , Metabolic Detoxication, Phase I , Potassium Channel Blockers/toxicity , Animals , Bile/metabolism , Cytochrome P-450 Enzyme System/metabolism , Dose-Response Relationship, Drug , Feces/chemistry , Ficusin/chemistry , Ficusin/metabolism , Male , Microsomes, Liver/drug effects , Microsomes, Liver/metabolism , Pancreatitis-Associated Proteins , Potassium Channel Blockers/chemistry , Potassium Channel Blockers/metabolism , Rats , Rats, Sprague-Dawley , Toxicity Tests, Chronic
2.
Curr Med Chem ; 11(23): 3029-40, 2004 Dec.
Article in English | MEDLINE | ID: mdl-15578997

ABSTRACT

Ziconotide (PRIALT) is a neuroactive peptide in the final stages of clinical development as a novel non-opioid treatment for severe chronic pain. It is the synthetic equivalent of omega-MVIIA, a component of the venom of the marine snail, Conus magus. The mechanism of action underlying ziconotide's therapeutic profile derives from its potent and selective blockade of neuronal N-type voltage-sensitive calcium channels (N-VSCCs). Direct blockade of N-VSCCs inhibits the activity of a subset of neurons, including pain-sensing primary nociceptors. This mechanism of action distinguishes ziconotide from all other analgesics, including opioid analgesics. In fact, ziconotide is potently anti-nociceptive in animal models of pain in which morphine exhibits poor anti-nociceptive activity. Moreover, in contrast to opiates, tolerance to ziconotide is not observed. Clinical studies of ziconotide in more than 2,000 patients reveal important correlations to ziconotide's non-clinical pharmacology. For example, ziconotide provides significant pain relief to severe chronic pain sufferers who have failed to obtain relief from opiate therapy and no evidence of tolerance to ziconotide is seen in these patients. Contingent on regulatory approval, ziconotide will be the first in a new class of neurological drugs: the N-type calcium channel blockers, or NCCBs. Its novel mechanism of action as a non-opioid analgesic suggests ziconotide has the potential to play a valuable role in treatment regimens for severe chronic pain. If approved for clinical use, ziconotide will further validate the neuroactive venom peptides as a source of new and useful medicines.


Subject(s)
Calcium Channel Blockers/therapeutic use , Pain/drug therapy , omega-Conotoxins/therapeutic use , Amino Acid Sequence/genetics , Animals , Calcium Channel Blockers/chemistry , Chronic Disease , Humans , Molecular Sequence Data , omega-Conotoxins/chemistry , omega-Conotoxins/genetics
3.
Eur J Biochem ; 268(4): 1066-75, 2001 Feb.
Article in English | MEDLINE | ID: mdl-11179973

ABSTRACT

Among voltage-gated Ca2+ channels the non-dihydropyridine-sensitive alpha1E subunit is functionally less well characterized than the structurally related alpha1A (omega-agatoxin-IVA sensitive, P- /Q-type) and alpha1B (omega-conotoxin-GVIA sensitive, N-type) subunits. In the rat insulinoma cell line, INS-1, a tissue-specific splice variant of alpha1E (alpha1Ee) has been characterized at the mRNA and protein levels, suggesting that INS-1 cells are a suitable model for investigating the function of alpha1Ee. In alpha1E-transfected human embryonic kidney (HEK-293) cells the alpha1E-selective peptide antagonist SNX-482 (100 nM) reduces alpha1Ed- and alpha1Ee-induced Ba2+ inward currents in the absence and presence of the auxiliary subunits beta3 and alpha2delta-2 by more than 80%. The inhibition is fast and only partially reversible. No effect of SNX-482 was detected on the recombinant T-type Ca2+ channel subunits alpha1G, alpha1H, and alpha1I showing that the toxin from the venom of Hysterocrates gigas is useful as an alpha1E-selective antagonist. After blocking known components of Ca2+ channel inward current in INS-1 cells by 2 microM (+/-)-isradipine plus 0.5 microM omega-conotoxin-MVIIC, the remaining current is reduced by 100 nM SNX-482 from -12.4 +/- 1.2 pA/pF to -7.6 +/- 0.5 pA/pF (n = 9). Furthermore, in INS-1 cells, glucose- and KCl-induced insulin release are reduced by SNX-482 in a dose-dependent manner leading to the conclusion that alpha1E, in addition to L-type and non-L-type (alpha1A-mediated) Ca2+ currents, is involved in Ca2+ dependent insulin secretion of INS-1 cells.


Subject(s)
Calcium Channels, R-Type/physiology , Calcium Channels/physiology , Cation Transport Proteins , Insulin/metabolism , Insulinoma/metabolism , Islets of Langerhans/metabolism , Alternative Splicing , Animals , Barium/metabolism , Calcium Channel Blockers/pharmacology , Calcium Channels/genetics , Calcium Channels, R-Type/genetics , Cell Culture Techniques/methods , Cells, Cultured , Electric Conductivity , Glucose/pharmacology , Humans , Insulin Secretion , Islets of Langerhans/drug effects , Mice , Patch-Clamp Techniques , Potassium Chloride/pharmacology , Protein Isoforms/genetics , Protein Isoforms/metabolism , Protein Isoforms/physiology , Spider Venoms/pharmacology , Transfection , Tumor Cells, Cultured
5.
Drug Des Discov ; 17(1): 85-93, 2000.
Article in English | MEDLINE | ID: mdl-10928451

ABSTRACT

Several novel N-type voltage sensitive calcium channel blockers showed high affinity in the IMR32 assay and efficacy in the anti-writhing model. Herein, we describe the design, synthesis, SAR studies, biological data, physicochemical properties and pharmacokinetics of this 4-piperidinylaniline series.


Subject(s)
Analgesics, Non-Narcotic/pharmacology , Aniline Compounds/pharmacology , Calcium Channel Blockers/pharmacology , Calcium Channels, N-Type/drug effects , Animals , Calcium Channel Blockers/chemical synthesis , Mice , Structure-Activity Relationship
6.
Bioorg Med Chem ; 8(6): 1203-12, 2000 Jun.
Article in English | MEDLINE | ID: mdl-10896101

ABSTRACT

Our drug discovery efforts for N-type calcium channel blockers in the 4-piperidinylaniline series led to the discovery of an orally active analgesic agent 26.1-[4-Dimethylamino-benzyl)-piperidin-4-yl]-[4-(3,3-dimethyl-but yl)-phenyl]-(3-methyl-but-2-enyl)amine (26) showed high affinity to functionally block N-type calcium channels (IC50=0.7 microM in the IMR32 assay) and exhibited high efficacy in the anti-writhing analgesia test with mice (ED50=12 mg/kg by po and 4 mg/kg by iv). In this report, the rationale for the design, synthesis, biological evaluation, and pharmacokinetics of this series of blockers is described.


Subject(s)
Analgesics/chemistry , Analgesics/pharmacology , Aniline Compounds/chemistry , Aniline Compounds/pharmacology , Calcium Channel Blockers/chemistry , Calcium Channel Blockers/pharmacology , Calcium Channels, N-Type/drug effects , Piperidines/chemistry , Piperidines/pharmacology , Administration, Oral , Analgesics/chemical synthesis , Aniline Compounds/chemical synthesis , Animals , Calcium Channel Blockers/administration & dosage , Calcium Channel Blockers/chemical synthesis , Cell Line , Magnetic Resonance Spectroscopy , Male , Mass Spectrometry , Mice , Molecular Structure , Piperidines/chemical synthesis , Rats , Rats, Wistar
7.
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
9.
J Med Chem ; 42(20): 4239-49, 1999 Oct 07.
Article in English | MEDLINE | ID: mdl-10514294

ABSTRACT

In this article, the rationale for the design, synthesis, and biological evaluation of a series of N-type voltage-sensitive calcium channel (VSCC) blockers is described. N-Type VSCC blockers, such as ziconotide, have shown utility in several models of stroke and pain. Modification of the previously reported lead, 1a, led to several 4-(4-benzyloxylphenyl)piperidine structures with potent in vitro and in vivo activities. In this series, the most interesting compound, (S)-2-amino-1-{4-[(4-benzyloxy-phenyl)-(3-methyl-but-2-enyl)-amino]-p iperidin-1-yl}-4-methyl-pentan-1-one (11), blocked N-type calcium channels (IC(50) = 0.67 microM in the IMR32 assay) and was efficacious in the audiogenic DBA/2 seizure mouse model (ED(50) = 6 mg/kg, iv) as well as the antiwrithing model (ED(50) = 6 mg/kg, iv). Whole-cell voltage-clamp electrophysiology experiments demonstrated that compound 11 blocked N-type Ca(2+) channels and Na(+) channels in superior cervical ganglion neurons at similar concentrations. Compound 11, which showed superior in vivo efficacy, stands out as an interesting lead for further development of neurotherapeutic agents in this series.


Subject(s)
Analgesics, Non-Narcotic/chemical synthesis , Anticonvulsants/chemical synthesis , Calcium Channel Blockers/chemical synthesis , Neurons/metabolism , Piperidines/chemical synthesis , Acoustic Stimulation , Analgesics, Non-Narcotic/chemistry , Analgesics, Non-Narcotic/pharmacokinetics , Analgesics, Non-Narcotic/pharmacology , Animals , Anticonvulsants/chemistry , Anticonvulsants/pharmacokinetics , Anticonvulsants/pharmacology , Blood Pressure/drug effects , Calcium Channel Blockers/chemistry , Calcium Channel Blockers/pharmacokinetics , Calcium Channel Blockers/pharmacology , Cell Line , Heart Rate/drug effects , Humans , In Vitro Techniques , Ion Channel Gating , Male , Mice , Mice, Inbred DBA , Microsomes, Liver/metabolism , Pain Measurement , Patch-Clamp Techniques , Piperidines/chemistry , Piperidines/pharmacokinetics , Piperidines/pharmacology , Rats , Rats, Sprague-Dawley , Rats, Wistar , Seizures/drug therapy
11.
Bioorg Med Chem Lett ; 9(15): 2151-6, 1999 Aug 02.
Article in English | MEDLINE | ID: mdl-10465535

ABSTRACT

Selective N-type voltage sensitive calcium channel (VSCC) blockers have shown efficacy in several animal models of stroke and pain. In the process of searching for small molecule N-type calcium channel blockers, we have identified a series of N-methyl-N-aralkyl-peptidylamines with potent functional activity at N-type VSCCs. The most active compound discovered in this series is PD 173212 (11, IC50 = 36 nM in the IMR-32 assays). SAR and pharmacological evaluation of this series are described.


Subject(s)
Calcium Channel Blockers/pharmacology , Calcium Channels/metabolism , Dipeptides/pharmacology , Animals , Calcium Channel Blockers/therapeutic use , Calcium Channels/drug effects , Dipeptides/chemistry , Disease Models, Animal , Humans , Mice , Seizures/drug therapy , Structure-Activity Relationship , Tumor Cells, Cultured
12.
Bioorg Med Chem Lett ; 9(6): 907-12, 1999 Mar 22.
Article in English | MEDLINE | ID: mdl-10206559

ABSTRACT

Selective N-type voltage sensitive calcium channel (VSCC) blockers have shown utility in several models of stroke and pain. We are especially interested in small molecule N-type calcium channel blockers for therapeutic use. Herein, we report a series of N,N-dialkyl-dipeptidylamines with potent functional activity at N-type VSCCs and in vivo efficacy. The synthesis, SAR, and pharmacological evaluation of this series are discussed.


Subject(s)
Calcium Channel Blockers/chemical synthesis , Diamines/chemical synthesis , Diamines/pharmacology , Dipeptides/chemical synthesis , Dipeptides/pharmacology , Animals , Disease Models, Animal , Mice , Mice, Inbred DBA , Models, Chemical , Seizures/drug therapy
13.
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
14.
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
15.
Neuropharmacology ; 36(6): 865-72, 1997 Jun.
Article in English | MEDLINE | ID: mdl-9225314

ABSTRACT

We have used whole-cell patch clamp recordings and pharmacological blockers of Ca channels to compare the pharmacology of Ca channels that mediate synaptic transmission at the three types of synapses innervating Purkinje cells in rat cerebellar slices. Both parallel fiber and climbing fiber excitatory synapses were sensitive to the P-type Ca channel blocker, omega-AgaIVA and the P/Q/N-type channel blocker, omega-conotoxin MVIIC. Transmission at inhibitory interneuronal synapses was not suppressed by these toxins, or by the N-type (omega-conotoxins GVIA and MVIIA) or L-type (nimodipine) channel blockers. Inhibitory transmission could be inhibited by Ni2+ and amiloride, but only at concentrations (IC50 approximately 300 microM) that affect other types of Ca channels. These results indicate that excitatory and inhibitory presynaptic terminals of the cerebellar cortex possess different types of voltage-gated Ca channels. The excitatory terminals contain P-type, Q-type and N-type Ca channels, with P-type channels playing the most prominent role. The inhibitory terminals possess quite different type(s) of Ca channel. The heterogeneous distribution of Ca channel types should impart unique properties to transmitter release from the excitatory and inhibitory terminals.


Subject(s)
Calcium Channels/drug effects , Cerebellum/physiology , Presynaptic Terminals/physiology , Synapses/drug effects , Synaptic Transmission/drug effects , omega-Conotoxins , Animals , Calcium Channel Blockers/pharmacology , Calcium Channels/physiology , Cerebellum/drug effects , In Vitro Techniques , Patch-Clamp Techniques , Peptides/pharmacology , Presynaptic Terminals/drug effects , Rats , Rats, Sprague-Dawley , Spider Venoms/pharmacology , omega-Agatoxin IVA
16.
Biochemistry ; 36(14): 4317-26, 1997 Apr 08.
Article in English | MEDLINE | ID: mdl-9100028

ABSTRACT

The interaction between the proteins syntaxin 1A and SNAP-25 is a key step in synaptic vesicle docking and fusion. To define the SNAP-25 binding domain on syntaxin, we have prepared peptides that span the syntaxin H3 domain (residues 191-266), the region previously shown to be important for binding to SNAP-25, and then determined the affinities of these peptides for binding to SNAP-25. A minimal binding domain was identified within a region of 32 amino acids (residues 189-220). Its affinity for SNAP-25 is substantially enhanced by C-terminal extension (residues 221-266). Circular dichroism revealed the presence of substantial alpha-helicity in the H3 domain and in the 32-mer minimal binding domain, but not in H3 peptides that do not bind to SNAP-25. At temperatures that denature the alpha-helix of the minimal binding domain peptide, SNAP-25 binding is lost. Selected mutations in evolutionarily conserved residues of the amphiphilic alpha-helix within the minimal binding domain (e.g., residues 205 and 209) greatly reduce the affinity for SNAP-25 but have no major effect on secondary structure, suggesting that these residues may interact directly with SNAP-25. The H3 domain peptide and the minimal binding domain peptide inhibit norepinephrine release from PC12 cells. These results suggest that specific amino acid residues in the H3 domain, positioned by the underlying alpha-helical structure, are important for its binding to SNAP-25 and support the notion that this interaction is important for presynaptic vesicular exocytosis.


Subject(s)
Antigens, Surface/chemistry , Membrane Proteins , Nerve Tissue Proteins/chemistry , Nerve Tissue Proteins/metabolism , Amino Acid Sequence , Animals , Antigens, Surface/metabolism , Binding Sites , Circular Dichroism , Exocytosis/physiology , Molecular Sequence Data , Mutation , Norepinephrine/antagonists & inhibitors , Norepinephrine/metabolism , PC12 Cells/metabolism , Peptide Fragments/chemistry , Peptide Fragments/metabolism , Protein Binding , Protein Denaturation , Protein Structure, Secondary , Rats , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Synaptic Vesicles/metabolism , Synaptosomal-Associated Protein 25 , Syntaxin 1 , Temperature
17.
Drug Metab Dispos ; 25(3): 379-83, 1997 Mar.
Article in English | MEDLINE | ID: mdl-9172958

ABSTRACT

SNX-111, a selective N-type voltage-sensitive calcium channel blocker, is in clinical trials for the treatment of ischemia-induced brain injury and chronic pain. Pharmacokinetic studies were conducted in rats and cynomologus monkeys to determine the disposition of this compound when it is administered for 24 hr by continuous, constant-rate intravenous infusion. Venous blood samples for determination of SNX-111 plasma levels were collected at regular intervals immediately before, during, and after dosing. Plasma concentrations of SNX-111 equivalents were measured by radioimmunoassay. Pharmacokinetic parameters were derived from plasma SNX-111 concentration-time data using a two-compartment pharmacokinetic model. Results showed close correspondences between pharmacokinetic parameters determined for both species. There were no consistent gender- or dose-related differences in calculated kinetic parameters. In all cases, apparent steady-state plasma SNX-111 concentrations were achieved within 2-4 hr of initiating SNX-111 infusion. Steady-state volume of distribution values were approximately 40% of body weight, indicating extravascular dissemination of SNX-111 to both extracellular and intracellular fluids. Elimination curves contained two exponential components. The fast component (rat t1/2, alpha = 0.375 hr; monkey t1/2, alpha = 0.730 hr) accounted for approximately 97% of the unit impulse disposition function. The apparent terminal half-life ranged from 4.61 hr (rat) to 6.48 hr (monkey). Current findings constitute the first description of the pharmacokinetics of a member of the omega-conopeptide family of neuronal calcium channel blockers.


Subject(s)
Calcium Channel Blockers/pharmacokinetics , Peptides/pharmacokinetics , omega-Conotoxins , Animals , Calcium Channel Blockers/blood , Female , Infusions, Intravenous , Macaca fascicularis , Male , Neurons/drug effects , Neurons/metabolism , Peptides/blood , Rats , Rats, Sprague-Dawley
18.
J Biol Chem ; 270(36): 21234-42, 1995 Sep 08.
Article in English | MEDLINE | ID: mdl-7673157

ABSTRACT

Biochemical properties of the alpha 1 subunits of class A brain calcium channels (alpha 1A) were examined in adult rat brain membrane fractions using a site-directed anti-peptide antibody (anti-CNA3) specific for alpha 1A. Anti-CNA3 specifically immunoprecipitated high affinity receptor sites for omega-conotoxin MVIIC (Kd approximately 100 pM), but not receptor sites for the dihydropyridine isradipine or for omega-conotoxin GVIA. In immunoblotting and immunoprecipitation experiments, anti-CNA3 recognized at least two distinct immunoreactive alpha 1A polypeptides, a major form with an apparent molecular mass of 190 kDa and a minor, full-length form with an apparent molecular mass of 220 kDa. The 220- and 190-kDa alpha 1A polypeptides were also specifically recognized by both anti-BI-Nt and anti-BI-1-Ct antibodies, which are directed against the NH2- and COOH-terminal ends of alpha 1A predicted from cDNA sequence, respectively. These data indicate that the predicted NH2 and COOH termini are present in both size forms and therefore that these isoforms of alpha 1A are created by alternative RNA splicing rather than post-translational proteolytic processing of the NH2 or COOH termini. The 220-kDa form was phosphorylated preferentially by cAMP-dependent protein kinase, whereas protein kinase C and cGMP-dependent protein kinase preferentially phosphorylated the 190-kDa form. Our results identify at least two distinct alpha 1A subunits with different molecular mass, demonstrate that they may result from alternative mRNA splicing, and suggest that they may be differentially regulated by protein phosphorylation.


Subject(s)
Alternative Splicing , Brain/metabolism , Calcium Channels/metabolism , omega-Conotoxins , Amino Acid Sequence , Animals , Calcium Channel Blockers/metabolism , Calcium Channels/genetics , Dihydropyridines/metabolism , Enzyme Activation , Immunochemistry , Molecular Sequence Data , Peptides/metabolism , Phosphorylation , Protein Binding , Protein Kinases/metabolism , Rats , Rats, Sprague-Dawley , Second Messenger Systems , omega-Conotoxin GVIA
19.
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
20.
N Engl J Med ; 332(22): 1467-74, 1995 Jun 01.
Article in English | MEDLINE | ID: mdl-7739683

ABSTRACT

BACKGROUND: Voltage-gated calcium channels in small-cell lung carcinomas may initiate autoimmunity in the paraneoplastic neuromuscular disorder Lambert-Eaton syndrome. The calcium-channel subtype that is responsible is not known. METHODS: We compared the effects of antagonists of L-type, N-type, and P/Q-type neuronal calcium channels on the depolarization-dependent influx of calcium-45 in cultured carcinoma cells. Serum samples from patients with various disorders were tested for reactivity with P/Q-type channels solubilized from carcinoma and cerebellar membranes and N-type channels from cerebral cortex. RESULTS: P/Q-type calcium-channel antagonists were the most potent inhibitors of depolarization-induced 45Ca influx in cultured small-cell carcinoma cell lines. Anti-P/Q-type calcium-channel antibodies were found in serum from all 32 patients with Lambert-Eaton syndrome and a diagnosis of cancer and in 91 percent of the 33 patients with Lambert-Eaton syndrome without cancer. Anti-N-type calcium-channel antibodies were found in 49 percent of the 65 patients with the Lambert-Eaton Syndrome. Lower titers of anti-P/Q-type and anti-N-type calcium-channel antibodies were found in 54 percent of 70 patients with a paraneoplastic encephalomyeloneuropathic complication of lung, ovarian, or breast carcinoma, 24 percent of 90 patients with cancer but no evident neurologic complications, 23 percent of 78 patients with sporadic amyotrophic lateral sclerosis, and less than 3 percent of 69 patients with myasthenia gravis, epilepsy, or scleroderma. CONCLUSIONS: The high frequency of P/Q-type calcium-channel antibodies found in patients with Lambert-Eaton syndrome implies that antibodies of this specificity have a role in the presynaptic pathophysiology of this disorder.


Subject(s)
Autoantibodies/analysis , Calcium Channels/immunology , Lambert-Eaton Myasthenic Syndrome/immunology , Neurons/immunology , Paraneoplastic Syndromes/immunology , Calcium Channels/drug effects , Carcinoma, Small Cell/immunology , Carcinoma, Small Cell/pathology , Humans , Lung Neoplasms/immunology , Lung Neoplasms/pathology , Tumor Cells, Cultured
SELECTION OF CITATIONS
SEARCH DETAIL
...