Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
1.
Br J Pharmacol ; 166(5): 1654-68, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22229737

ABSTRACT

BACKGROUND AND PURPOSE: The µ-conopeptide family is defined by its ability to block voltage-gated sodium channels (VGSCs), a property that can be used for the development of myorelaxants and analgesics. We characterized the pharmacology of a new µ-conopeptide (µ-CnIIIC) on a range of preparations and molecular targets to assess its potential as a myorelaxant. EXPERIMENTAL APPROACH: µ-CnIIIC was sequenced, synthesized and characterized by its direct block of elicited twitch tension in mouse skeletal muscle and action potentials in mouse sciatic and pike olfactory nerves. µ-CnIIIC was also studied on HEK-293 cells expressing various rodent VGSCs and also on voltage-gated potassium channels and nicotinic acetylcholine receptors (nAChRs) to assess cross-interactions. Nuclear magnetic resonance (NMR) experiments were carried out for structural data. KEY RESULTS: Synthetic µ-CnIIIC decreased twitch tension in mouse hemidiaphragms (IC(50) = 150 nM), and displayed a higher blocking effect in mouse extensor digitorum longus muscles (IC = 46 nM), compared with µ-SIIIA, µ-SmIIIA and µ-PIIIA. µ-CnIIIC blocked Na(V)1.4 (IC(50) = 1.3 nM) and Na(V)1.2 channels in a long-lasting manner. Cardiac Na(V)1.5 and DRG-specific Na(V)1.8 channels were not blocked at 1 µM. µ-CnIIIC also blocked the α3ß2 nAChR subtype (IC(50) = 450 nM) and, to a lesser extent, on the α7 and α4ß2 subtypes. Structure determination of µ-CnIIIC revealed some similarities to α-conotoxins acting on nAChRs. CONCLUSION AND IMPLICATIONS: µ-CnIIIC potently blocked VGSCs in skeletal muscle and nerve, and hence is applicable to myorelaxation. Its atypical pharmacological profile suggests some common structural features between VGSCs and nAChR channels.


Subject(s)
Conotoxins/pharmacology , Conus Snail , Nicotinic Antagonists/pharmacology , Peptides/pharmacology , Sodium Channel Blockers/pharmacology , Amino Acid Sequence , Animals , Conotoxins/chemistry , Esocidae , Female , HEK293 Cells , Humans , In Vitro Techniques , Male , Mice , Molecular Sequence Data , Muscle Contraction/drug effects , Muscle, Skeletal/drug effects , Muscle, Skeletal/physiology , Nicotinic Antagonists/chemistry , Olfactory Nerve/drug effects , Olfactory Nerve/physiology , Oocytes , Peptides/chemistry , Protein Conformation , Receptors, Nicotinic/physiology , Sciatic Nerve/drug effects , Sciatic Nerve/physiology , Sodium Channel Blockers/chemistry , Sodium Channels/physiology , Xenopus laevis
2.
J Biol Chem ; 286(12): 10618-27, 2011 Mar 25.
Article in English | MEDLINE | ID: mdl-21252236

ABSTRACT

Discovery of proteins expressed in the central nervous system sharing the three-finger structure with snake α-neurotoxins provoked much interest to their role in brain functions. Prototoxin LYNX1, having homology both to Ly6 proteins and three-finger neurotoxins, is the first identified member of this family membrane-tethered by a GPI anchor, which considerably complicates in vitro studies. We report for the first time the NMR spatial structure for the water-soluble domain of human LYNX1 lacking a GPI anchor (ws-LYNX1) and its concentration-dependent activity on nicotinic acetylcholine receptors (nAChRs). At 5-30 µM, ws-LYNX1 competed with (125)I-α-bungarotoxin for binding to the acetylcholine-binding proteins (AChBPs) and to Torpedo nAChR. Exposure of Xenopus oocytes expressing α7 nAChRs to 1 µM ws-LYNX1 enhanced the response to acetylcholine, but no effect was detected on α4ß2 and α3ß2 nAChRs. Increasing ws-LYNX1 concentration to 10 µM caused a modest inhibition of these three nAChR subtypes. A common feature for ws-LYNX1 and LYNX1 is a decrease of nAChR sensitivity to high concentrations of acetylcholine. NMR and functional analysis both demonstrate that ws-LYNX1 is an appropriate model to shed light on the mechanism of LYNX1 action. Computer modeling, based on ws-LYNX1 NMR structure and AChBP x-ray structure, revealed a possible mode of ws-LYNX1 binding.


Subject(s)
GPI-Linked Proteins/chemistry , Models, Molecular , Receptors, Nicotinic/chemistry , Adaptor Proteins, Signal Transducing , Animals , Bungarotoxins/chemistry , Bungarotoxins/pharmacology , GPI-Linked Proteins/genetics , GPI-Linked Proteins/metabolism , Humans , Nuclear Magnetic Resonance, Biomolecular , Oocytes , Protein Binding , Protein Structure, Quaternary , Protein Structure, Tertiary , Receptors, Nicotinic/genetics , Receptors, Nicotinic/metabolism , Solubility , Xenopus laevis
3.
J Biol Chem ; 285(11): 8302-15, 2010 Mar 12.
Article in English | MEDLINE | ID: mdl-20071329

ABSTRACT

Snake venoms are a mixture of pharmacologically active proteins and polypeptides that have led to the development of molecular probes and therapeutic agents. Here, we describe the structural and functional characterization of a novel neurotoxin, haditoxin, from the venom of Ophiophagus hannah (King cobra). Haditoxin exhibited novel pharmacology with antagonism toward muscle (alphabetagammadelta) and neuronal (alpha(7), alpha(3)beta(2), and alpha(4)beta(2)) nicotinic acetylcholine receptors (nAChRs) with highest affinity for alpha(7)-nAChRs. The high resolution (1.5 A) crystal structure revealed haditoxin to be a homodimer, like kappa-neurotoxins, which target neuronal alpha(3)beta(2)- and alpha(4)beta(2)-nAChRs. Interestingly however, the monomeric subunits of haditoxin were composed of a three-finger protein fold typical of curaremimetic short-chain alpha-neurotoxins. Biochemical studies confirmed that it existed as a non-covalent dimer species in solution. Its structural similarity to short-chain alpha-neurotoxins and kappa-neurotoxins notwithstanding, haditoxin exhibited unique blockade of alpha(7)-nAChRs (IC(50) 180 nm), which is recognized by neither short-chain alpha-neurotoxins nor kappa-neurotoxins. This is the first report of a dimeric short-chain alpha-neurotoxin interacting with neuronal alpha(7)-nAChRs as well as the first homodimeric three-finger toxin to interact with muscle nAChRs.


Subject(s)
Cobra Neurotoxin Proteins/chemistry , Elapid Venoms/chemistry , Elapidae , Nicotinic Antagonists/chemistry , Receptors, Nicotinic/physiology , Amino Acid Sequence , Animals , Chickens , Cobra Neurotoxin Proteins/genetics , Cobra Neurotoxin Proteins/pharmacology , Crystallography, X-Ray , Diaphragm/drug effects , Diaphragm/physiology , Dimerization , Elapid Venoms/genetics , Elapid Venoms/pharmacology , Gene Library , Humans , Mice , Molecular Sequence Data , Muscle, Smooth/drug effects , Muscle, Smooth/physiology , Nicotinic Antagonists/pharmacology , Oocytes/physiology , Patch-Clamp Techniques , Protein Conformation , Rats , Rats, Sprague-Dawley , Structure-Activity Relationship , Xenopus , alpha7 Nicotinic Acetylcholine Receptor
4.
J Neurochem ; 111(4): 934-44, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19712060

ABSTRACT

alpha-Conotoxins interact with nicotinic acetylcholine receptors (nAChRs) and acetylcholine-binding proteins (AChBPs) at the sites for agonists/competitive antagonists. alpha-Conotoxins blocking muscle-type or alpha7 nAChRs compete with alpha-bungarotoxin. However, alpha-conotoxin ImII, a close homolog of the alpha7 nAChR-targeting alpha-conotoxin ImI, blocked alpha7 and muscle nAChRs without displacing alpha-bungarotoxin (Ellison et al. 2003, 2004), suggesting binding at a different site. We synthesized alpha-conotoxin ImII, its ribbon isomer (ImIIiso), 'mutant' ImII(W10Y) and found similar potencies in blocking human alpha7 and muscle nAChRs in Xenopus oocytes. Both isomers displaced [(125)I]-alpha-bungarotoxin from human alpha7 nAChRs in the cell line GH(4)C(1) (IC(50) 17 and 23 microM, respectively) and from Lymnaea stagnalis and Aplysia californica AChBPs (IC(50) 2.0-9.0 microM). According to SPR measurements, both isomers bound to immobilized AChBPs and competed with AChBP for immobilized alpha-bungarotoxin (K(d) and IC(50) 2.5-8.2 microM). On Torpedo nAChR, alpha-conotoxin [(125)I]-ImII(W10Y) revealed specific binding (K(d) 1.5-6.1 microM) and could be displaced by alpha-conotoxin ImII, ImIIiso and ImII(W10Y) with IC(50) 2.7, 2.2 and 3.1 microM, respectively. As alpha-cobratoxin and alpha-conotoxin ImI displaced [(125)I]-ImII(W10Y) only at higher concentrations (IC(50)> or = 90 microM), our results indicate that alpha-conotoxin ImII and its congeners have an additional binding site on Torpedo nAChR distinct from the site for agonists/competitive antagonists.


Subject(s)
Carrier Proteins/metabolism , Conotoxins/chemistry , Conotoxins/metabolism , Receptors, Nicotinic/metabolism , Torpedo/metabolism , Acetylcholine/pharmacology , Amino Acid Sequence , Animals , Aplysia , Binding Sites/drug effects , Binding Sites/physiology , Binding, Competitive/drug effects , Bungarotoxins/metabolism , Dose-Response Relationship, Drug , Humans , Inhibitory Concentration 50 , Iodine Isotopes/metabolism , Molecular Sequence Data , Oocytes , Radioligand Assay/methods , Receptors, Nicotinic/genetics , Serine Endopeptidases , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization/methods , Surface Plasmon Resonance/methods , Xenopus laevis , alpha7 Nicotinic Acetylcholine Receptor
5.
Expert Opin Ther Targets ; 13(4): 395-411, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19335063

ABSTRACT

BACKGROUND: Nicotinic acetylcholine receptors (nAChRs) are ligand-gated ion channels that participate in many physiological functions. Receptors result from the assembly of five homologous or heterologous subunits that form the ligand-binding site and an ionic pore. In vertebrates, 17 subunits have been identified, alpha (1 - 10), beta (1 - 4), gamma, delta and epsilon. Assembly of different subunit combinations allows a diversity of physiological and pharmacological properties. OBJECTIVE: To review the putative involvement of nAChRs in several diseases. METHODS: We discuss the expression pattern of the subunits, the pharmacological tools for distinguishing them and their role in pathogenesis. RESULTS/CONCLUSION: Long-standing efforts in this field should soon result in the finding of new molecules that might be applicable to situations ranging from neurological diseases to immune treatments.


Subject(s)
Nicotinic Agonists/pharmacology , Nicotinic Antagonists/pharmacology , Receptors, Nicotinic/drug effects , Allosteric Regulation/drug effects , Analgesics/pharmacology , Analgesics/therapeutic use , Bridged Bicyclo Compounds, Heterocyclic/adverse effects , Bridged Bicyclo Compounds, Heterocyclic/pharmacology , Bridged Bicyclo Compounds, Heterocyclic/therapeutic use , Cardiovascular Diseases/drug therapy , Cardiovascular Diseases/etiology , Drug Discovery , Hearing/physiology , Humans , Myasthenia Gravis/etiology , Myasthenia Gravis/immunology , Neurodegenerative Diseases/drug therapy , Neurodegenerative Diseases/physiopathology , Neurodegenerative Diseases/prevention & control , Nicotine/adverse effects , Nicotine/pharmacology , Nicotine/therapeutic use , Nicotinic Agonists/therapeutic use , Nicotinic Antagonists/therapeutic use , Pain/drug therapy , Pain/physiopathology , Protein Subunits/drug effects , Psoriasis/etiology , Pyridines/adverse effects , Pyridines/pharmacology , Pyridines/therapeutic use , Receptors, Nicotinic/classification , Receptors, Nicotinic/immunology , Receptors, Nicotinic/physiology , Schizophrenia/drug therapy , Schizophrenia/etiology , Schizophrenia/physiopathology , Smoking/adverse effects , Smoking Cessation , Tobacco Use Disorder/drug therapy , Tobacco Use Disorder/physiopathology
6.
J Biol Chem ; 283(10): 6272-80, 2008 Mar 07.
Article in English | MEDLINE | ID: mdl-18174177

ABSTRACT

TRPV4, a member of the vanilloid subfamily of the transient receptor potential (TRP) channels, is activated by a variety of stimuli, including cell swelling, moderate heat, and chemical compounds such as synthetic 4alpha-phorbol esters. TRPV4 displays a widespread expression in various cells and tissues and has been implicated in diverse physiological processes, including osmotic homeostasis, thermo- and mechanosensation, vasorelaxation, tuning of neuronal excitability, and bladder voiding. The mechanisms that regulate TRPV4 in these different physiological settings are currently poorly understood. We have recently shown that the relative amount of TRPV4 in the plasma membrane is enhanced by interaction with the SH3 domain of PACSIN 3, a member of the PACSIN family of proteins involved in synaptic vesicular membrane trafficking and endocytosis. Here we demonstrate that PACSIN 3 strongly inhibits the basal activity of TRPV4 and its activation by cell swelling and heat, while leaving channel gating induced by the synthetic ligand 4alpha-phorbol 12,13-didecanoate unaffected. A single proline mutation in the SH3 domain of PACSIN 3 abolishes its inhibitory effect on TRPV4, indicating that PACSIN 3 must bind to the channel to modulate its function. In line herewith, mutations at specific proline residues in the N terminus of TRPV4 abolish binding of PACSIN 3 and render the channel insensitive to PACSIN 3-induced inhibition. Taken together, these data suggest that PACSIN 3 acts as an auxiliary protein of TRPV4 channel that not only affects the channel's subcellular localization but also modulates its function in a stimulus-specific manner.


Subject(s)
Cell Membrane/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Phosphoproteins/metabolism , TRPV Cation Channels/metabolism , Adaptor Proteins, Signal Transducing , Animals , Carcinogens/pharmacology , Cell Line , Cell Membrane/genetics , Cytoskeletal Proteins , Homeostasis/drug effects , Homeostasis/physiology , Hot Temperature , Humans , Intracellular Signaling Peptides and Proteins/genetics , Mechanotransduction, Cellular/drug effects , Mechanotransduction, Cellular/physiology , Mice , Neurons/metabolism , Organ Specificity/physiology , Phorbol Esters/pharmacology , Phosphoproteins/genetics , Protein Binding/physiology , Protein Structure, Tertiary/physiology , TRPV Cation Channels/genetics
7.
J Neurosci ; 28(3): 576-86, 2008 Jan 16.
Article in English | MEDLINE | ID: mdl-18199759

ABSTRACT

Clotrimazole (CLT) is a widely used drug for the topical treatment of yeast infections of skin, vagina, and mouth. Common side effects of topical CLT application include irritation and burning pain of the skin and mucous membranes. Here, we provide evidence that transient receptor potential (TRP) channels in primary sensory neurons underlie these unwanted effects of CLT. We found that clinically relevant CLT concentrations activate heterologously expressed TRPV1 and TRPA1, two TRP channels that act as receptors of irritant chemical and/or thermal stimuli in nociceptive neurons. In line herewith, CLT stimulated a subset of capsaicin-sensitive and mustard oil-sensitive trigeminal neurons, and evoked nocifensive behavior and thermal hypersensitivity with intraplantar injection in mice. Notably, CLT-induced pain behavior was suppressed by the TRPV1-antagonist BCTC [(N-(-4-tertiarybutylphenyl)-4-(3-cholorpyridin-2-yl)tetrahydropyrazine-1(2H)-carboxamide)] and absent in TRPV1-deficient mice. In addition, CLT inhibited the cold and menthol receptor TRPM8, and blocked menthol-induced responses in capsaicin- and mustard oil-insensitive trigeminal neurons. The concentration for 50% inhibition (IC50) of inward TRPM8 current was approximately 200 nM, making CLT the most potent known TRPM8 antagonist and a useful tool to discriminate between TRPM8- and TRPA1-mediated responses. Together, our results identify TRP channels in sensory neurons as molecular targets of CLT, and offer means to develop novel CLT preparations with fewer unwanted sensory side effects.


Subject(s)
Anti-Infective Agents, Local/pharmacology , Clotrimazole/pharmacology , Neurons, Afferent/drug effects , TRPV Cation Channels/physiology , Transient Receptor Potential Channels/physiology , Animals , Avoidance Learning/drug effects , Behavior, Animal/drug effects , Calcium/metabolism , Cells, Cultured , Dose-Response Relationship, Drug , Gene Expression/drug effects , Gene Expression/physiology , Humans , Membrane Potentials/drug effects , Membrane Potentials/physiology , Membrane Potentials/radiation effects , Mice , Mice, Knockout , Neurons, Afferent/physiology , Patch-Clamp Techniques/methods , Reaction Time/drug effects , Reaction Time/physiology , TRPA1 Cation Channel , TRPM Cation Channels/genetics , TRPM Cation Channels/metabolism , TRPV Cation Channels/genetics , Time Factors , Transfection/methods , Transient Receptor Potential Channels/deficiency , Trigeminal Ganglion/cytology
8.
J Biol Chem ; 282(6): 3478-86, 2007 Feb 09.
Article in English | MEDLINE | ID: mdl-17142458

ABSTRACT

Small conductance calcium-activated potassium channels (SK, K(Ca)) are a family of voltage-independent K+ channels with a distinct physiology and pharmacology. The bee venom toxin apamin inhibits exclusively the three cloned SK channel subtypes (SK1, SK2, and SK3) with different affinity, highest for SK2, lowest for SK1, and intermediate for SK3 channels. The high selectivity of apamin made it a valuable tool to study the molecular makeup and function of native SK channels. Three amino acids located in the outer vestibule of the pore are of particular importance for the different apamin sensitivities of SK channels. Chimeric SK1 channels, enabling the homomeric expression of the rat SK1 (rSK1) subunit and containing the core domain (S1-S6) of rSK1, are apamin-insensitive. By contrast, channels formed by the human orthologue human SK1 (hSK1) are sensitive to apamin. This finding hinted at the involvement of regions beyond the pore as determinants of apamin sensitivity, because hSK1 and rSK1 have an identical amino acid sequence in the pore region. Here we investigated which parts of the channels outside the pore region are important for apamin sensitivity by constructing chimeras between apamin-insensitive and -sensitive SK channel subunits and by introducing point mutations. We demonstrate that a single amino acid situated in the extracellular loop between the transmembrane segments S3 and S4 has a major impact on apamin sensitivity. Our findings enabled us to convert the hSK1 channel into a channel that was as sensitive for apamin as SK2, the SK channel with the highest sensitivity.


Subject(s)
Amino Acids/physiology , Apamin/pharmacology , Small-Conductance Calcium-Activated Potassium Channels/chemistry , Small-Conductance Calcium-Activated Potassium Channels/physiology , Amino Acid Sequence , Amino Acids/chemistry , Amino Acids/genetics , Animals , CHO Cells , Cell Line , Cricetinae , Cricetulus , Germinal Center Kinases , Humans , Mice , Molecular Sequence Data , Point Mutation , Protein Serine-Threonine Kinases/antagonists & inhibitors , Protein Serine-Threonine Kinases/biosynthesis , Protein Serine-Threonine Kinases/physiology , Protein Structure, Tertiary/drug effects , Protein Structure, Tertiary/physiology , Rats , Small-Conductance Calcium-Activated Potassium Channels/antagonists & inhibitors , Small-Conductance Calcium-Activated Potassium Channels/biosynthesis
9.
J Biol Chem ; 281(27): 18753-62, 2006 Jul 07.
Article in English | MEDLINE | ID: mdl-16627472

ABSTRACT

TRPV4 is a cation channel that responds to a variety of stimuli including mechanical forces, temperature, and ligand binding. We set out to identify TRPV4-interacting proteins by performing yeast two-hybrid screens, and we isolated with the avian TRPV4 amino terminus the chicken orthologues of mammalian PACSINs 1 and 3. The PACSINs are a protein family consisting of three members that have been implicated in synaptic vesicular membrane trafficking and regulation of dynamin-mediated endocytotic processes. In biochemical interaction assays we found that all three murine PACSIN isoforms can bind to the amino terminus of rodent TRPV4. No member of the PACSIN protein family was able to biochemically interact with TRPV1 and TRPV2. Co-expression of PACSIN 3, but not PACSINs 1 and 2, shifted the ratio of plasma membrane-associated versus cytosolic TRPV4 toward an apparent increase of plasma membrane-associated TRPV4 protein. A similar shift was also observable when we blocked dynamin-mediated endocytotic processes, suggesting that PACSIN 3 specifically affects the endocytosis of TRPV4, thereby modulating the subcellular localization of the ion channel. Mutational analysis shows that the interaction of the two proteins requires both a TRPV4-specific proline-rich domain upstream of the ankyrin repeats of the channel and the carboxyl-terminal Src homology 3 domain of PACSIN 3. Such a functional interaction could be important in cell types that show distribution of both proteins to the same subcellular regions such as renal tubule cells where the proteins are associated with the luminal plasma membrane.


Subject(s)
Phosphoproteins/metabolism , TRPV Cation Channels/metabolism , Adaptor Proteins, Signal Transducing , Amino Acid Sequence , Animals , Binding Sites , Cell Compartmentation , Cell Line , Cell Membrane/chemistry , Cell Membrane/metabolism , Chickens , Cytoskeletal Proteins , Humans , Intracellular Signaling Peptides and Proteins , Mice , Molecular Sequence Data , Mutation , Neuropeptides/chemistry , Neuropeptides/genetics , Neuropeptides/metabolism , Phosphoproteins/chemistry , Phosphoproteins/genetics , Protein Binding , Protein Transport , Proteins/chemistry , Proteins/genetics , Proteins/metabolism , Rats , TRPV Cation Channels/chemistry , TRPV Cation Channels/genetics
10.
Br J Pharmacol ; 143(6): 753-64, 2004 Nov.
Article in English | MEDLINE | ID: mdl-15504758

ABSTRACT

We have recently shown that the alkaloid methyl-laudanosine blocks SK channel-mediated afterhyperpolarizations (AHPs) in midbrain dopaminergic neurones. However, the relative potency of the compound on the SK channel subtypes and its ability to block AHPs of other neurones were unknown. Using whole-cell patch-clamp experiments in transfected cell lines, we found that the compound blocks SK1, SK2 and SK3 currents with equal potency: its mean IC(50)s were 1.2, 0.8 and 1.8 microM, respectively. IK currents were unaffected. In rat brain slices, methyl-laudanosine blocked apamin-sensitive AHPs in serotonergic neurones of the dorsal raphe and noradrenergic neurones of the locus coeruleus with IC(50)s of 21 and 19 microM, as compared to 15 microM in dopaminergic neurones. However, at 100 microM, methyl-laudanosine elicited a constant hyperpolarization of serotonergic neurones of about 9 mV, which was inconsistently (i.e. not in a reproducible manner) antagonized by atropine and hence partly due to the activation of muscarinic receptors. While exploring the pharmacology of related compounds, we found that methyl-noscapine also blocked SK channels. In cell lines, methyl-noscapine blocked SK1, SK2 and SK3 currents with mean IC(50)s of 5.9, 5.6 and 3.9 microM, respectively. It also did not block IK currents. Methyl-noscapine was slightly less potent than methyl-laudanosine in blocking AHPs in brain slices, its IC(50)s being 42, 37 and 29 microM in dopaminergic, serotonergic and noradrenergic neurones, respectively. Interestingly, no significant non-SK effects were observed with methyl-noscapine in slices. At a concentration of 300 microM, methyl-noscapine elicited the same changes in excitability in the three neuronal types than did a supramaximal concentration of apamin (300 nM). Methyl-laudanosine and methyl-noscapine produced a rapidly reversible blockade of SK channels as compared with apamin. The difference between the IC(50)s of apamin (0.45 nM) and methyl-laudanosine (1.8 microM) in SK3 cells was essentially due to a major difference in their k(-1) (0.028 s(-1) for apamin and >or=20 s(-1) for methyl-laudanosine). These experiments demonstrate that both methyl-laudanosine and methyl-noscapine are medium potency, quickly dissociating, SK channel blockers with a similar potency on the three SK subtypes. Methyl-noscapine may be superior in terms of specificity for the SK channels.


Subject(s)
Brain/drug effects , Isoquinolines/pharmacology , Noscapine/pharmacology , Potassium Channels, Calcium-Activated/antagonists & inhibitors , Potassium Channels, Calcium-Activated/physiology , Animals , Brain/physiology , CHO Cells , Cell Line , Cricetinae , Dose-Response Relationship, Drug , Electrophysiology , In Vitro Techniques , Isoquinolines/chemistry , Male , Noscapine/chemistry , Potassium Channel Blockers/chemistry , Potassium Channel Blockers/pharmacology , Rats , Rats, Wistar , Small-Conductance Calcium-Activated Potassium Channels
11.
Toxicon ; 43(8): 933-49, 2004 Jun 15.
Article in English | MEDLINE | ID: mdl-15208027

ABSTRACT

Potassium channels regulate the membrane excitability of neurons, play a major role in shaping action potentials, determining firing patterns and regulating neurotransmitter release, and thus significantly contribute to neuronal signal encoding and integration. This review focuses on the molecular and cellular basis for the specific function of small-conductance calcium-activated potassium channels (SK channels) in the nervous system. SK channels are activated by an intracellular increase of free calcium during action potentials. They mediate currents that modulate the firing frequency of neurons. Three SK channel subunits have been cloned and form channels, which are voltage-insensitive, activated by submicromolar intracellular calcium concentrations, and are blocked, with different affinities, by a number of toxins and organic compounds. Different neurons in the central and peripheral nervous system express distinct subsets of SK channel subunits. Recent progress has been made in relating cloned SK channels to their native counterparts. These findings argue in favour of regulatory mechanisms conferring to native SK channels with specific subunit compositions distinct and specific functional profiles in different neurons.


Subject(s)
Calcium/metabolism , Neurons/physiology , Potassium Channels/metabolism , Toxins, Biological/metabolism , Animals , Large-Conductance Calcium-Activated Potassium Channels , Membrane Potentials/physiology , Neurons/metabolism , Potassium Channels/genetics , Potassium Channels, Calcium-Activated/genetics , Potassium Channels, Calcium-Activated/metabolism , Small-Conductance Calcium-Activated Potassium Channels , Structure-Activity Relationship
12.
J Biol Chem ; 279(13): 12088-92, 2004 Mar 26.
Article in English | MEDLINE | ID: mdl-14761961

ABSTRACT

Two small conductance, calcium-activated potassium channels (SK channels), SK2 and SK3, have been shown to contribute to the afterhyperpolarization (AHP) and to shape the firing behavior in neurons for example in the hippocampal formation, the dorsal vagal nucleus, the subthalamic nucleus, and the cerebellum. In heterologous expression systems, SK2 and SK3 currents are blocked by the bee venom toxin apamin, just as well as the corresponding neuronal AHP currents. However, the functional role and pharmacological profile of SK1 channels from rat brain (rSK1) is still largely unknown, as so far rSK1 homomeric channels could not be functionally expressed. We have performed a domain analysis to elucidate the pharmacological profile and the molecular determinants of rSK1 channel expression by using channel chimeras in combination with immunocytochemistry, immunoblot analysis, and electrophysiology. Our results reveal that the rSK1 subunit is synthesized in cells but does not form functional homomeric channels. Exchanging the carboxyl terminus of rSK1 for that of hSK1 or rSK2 is sufficient to rescue the functional expression of rSK1 channels. Additionally, transplantation of both amino and carboxyl termini of rSK1 onto hSK1 subunits, normally forming functional homomeric channel, hinders their functional expression, while hSK1 channels containing only the rSK1 carboxyl terminus are functional. These results suggest that the lack of functional expression of rSK1 channels is probably due to problems in their assembly and tetramerization but not in their calmodulin-dependent gating. Finally, we show that chimeric channels containing the core domain (S1-S6) of rSK1, unlike hSK1, are apamin-insensitive.


Subject(s)
Brain/metabolism , Potassium Channels, Calcium-Activated , Potassium Channels/chemistry , Amino Acid Sequence , Animals , Apamin/pharmacology , Calmodulin/pharmacology , Cell Line , DNA/chemistry , Electrophysiology , Humans , Immunohistochemistry , Microscopy, Fluorescence , Molecular Sequence Data , Neurons/metabolism , Potassium Channels/biosynthesis , Potassium Channels/genetics , Protein Structure, Tertiary , Rats , Recombinant Fusion Proteins/metabolism , Sequence Homology, Amino Acid , Small-Conductance Calcium-Activated Potassium Channels , Transfection , Tubocurarine/pharmacology
13.
J Biol Chem ; 277(48): 46101-9, 2002 Nov 29.
Article in English | MEDLINE | ID: mdl-12239213

ABSTRACT

The biophysical properties of small conductance Ca(2+)-activated K(+) (SK) channels are well suited to underlie afterhyperpolarizations (AHPs) shaping the firing patterns of a conspicuous number of central and peripheral neurons. We have identified a new scorpion toxin (tamapin) that binds to SK channels with high affinity and inhibits SK channel-mediated currents in pyramidal neurons of the hippocampus as well as in cell lines expressing distinct SK channel subunits. This toxin distinguished between the SK channels underlying the apamin-sensitive I(AHP) and the Ca(2+)-activated K(+) channels mediating the slow I(AHP) (sI(AHP)) in hippocampal neurons. Compared with related scorpion toxins, tamapin displayed a unique, remarkable selectivity for SK2 versus SK1 ( approximately 1750-fold) and SK3 ( approximately 70-fold) channels and is the most potent SK2 channel blocker characterized so far (IC(50) for SK2 channels = 24 pm). Tamapin will facilitate the characterization of the subunit composition of native SK channels and help determine their involvement in electrical and biochemical signaling.


Subject(s)
Calcium/metabolism , Membrane Potentials/drug effects , Neurons/drug effects , Neurotoxins/pharmacology , Potassium Channels/drug effects , Scorpion Venoms/chemistry , Amino Acid Sequence , Animals , Apamin/metabolism , Cell Line , Humans , Molecular Sequence Data , Neurons/physiology , Neurotoxins/chemistry , Neurotoxins/isolation & purification , Rats , Rats, Wistar , Scorpion Venoms/isolation & purification , Scorpion Venoms/pharmacology , Scorpions , Sequence Homology, Amino Acid , Spectrometry, Mass, Electrospray Ionization
SELECTION OF CITATIONS
SEARCH DETAIL
...