Conus venoms: a rich source of novel ion channel-targeted peptides.

The cone snails (genus Conus) are venomous marine molluscs that use small, structured peptide toxins (conotoxins) for prey capture, defense, and competitor deterrence. Each of the 500 Conus can express approximately 100 different conotoxins, with little overlap between species. An overwhelming majority of these peptides are probably targeted selectively to a specific ion channel. Because conotoxins discriminate between closely related subtypes of ion channels, they are widely used as pharmacological agents in ion channel research, and several have direct diagnostic and therapeutic potential. Large conotoxin families can comprise hundreds or thousands of different peptides; most families have a corresponding ion channel family target (i.e., omega-conotoxins and Ca channels, alpha-conotoxins and nicotinic receptors). Different conotoxin families may have different ligand binding sites on the same ion channel target (i.e., mu-conotoxins and delta-conotoxins to sites 1 and 6 of Na channels, respectively). The individual peptides in a conotoxin family are typically each selectively targeted to a diverse set of different molecular isoforms within the same ion channel family. This review focuses on the targeting specificity of conotoxins and their differential binding to different states of an ion channel.

[1]  H. Nakamura,et al.  The occurrence of arachidonic acid in the venom duct of the marine snailConus textile , 1982, Experientia.

[2]  D. Hornung,et al.  Use dependence of sodium current inhibition by tetrodotoxin in rat cardiac muscle: influence of channel state , 1990, Pflügers Archiv.

[3]  W. Agnew,et al.  The μI skeletal muscle sodium channel: Mutation E403Q eliminates sensitivity to tetrodotoxin but not to μ-conotoxins GIIIA and GIIIB , 2004, The Journal of Membrane Biology.

[4]  I. Huys,et al.  A novel conotoxin inhibiting vertebrate voltage-sensitive potassium channels. , 2003, Toxicon : official journal of the International Society on Toxinology.

[5]  C. Ireland,et al.  Characterization and three-dimensional structure determination of psi-conotoxin Piiif, a novel noncompetitive antagonist of nicotinic acetylcholine receptors. , 2003, Biochemistry.

[6]  C. Ireland,et al.  An Improved Solution Structure for ψ-Conotoxin Piiie†,‡ , 2003 .

[7]  D. Yoshikami,et al.  Novel excitatory Conus peptides define a new conotoxin superfamily , 2003, Journal of neurochemistry.

[8]  Youxing Jiang,et al.  The principle of gating charge movement in a voltage-dependent K+ channel , 2003, Nature.

[9]  M. Cadene,et al.  X-ray structure of a voltage-dependent K+ channel , 2003, Nature.

[10]  J. Tytgat,et al.  A Novel Conotoxin from Conus betulinus, κ-BtX, Unique in Cysteine Pattern and in Function as a Specific BK Channel Modulator* , 2003, The Journal of Biological Chemistry.

[11]  P. Ascenzi,et al.  Contryphan-Vn: a modulator of Ca2+-dependent K+ channels. , 2003, Biochemical and biophysical research communications.

[12]  David John Adams,et al.  ω-Conotoxin CVID Inhibits a Pharmacologically Distinct Voltage-sensitive Calcium Channel Associated with Transmitter Release from Preganglionic Nerve Terminals* , 2003, The Journal of Biological Chemistry.

[13]  Michael P. Ferber,et al.  A Novel Conus Peptide Ligand for K+Channels* , 2003, The Journal of Biological Chemistry.

[14]  J. McIntosh,et al.  α-Conotoxins ImI and ImII , 2003, The Journal of Biological Chemistry.

[15]  G. S. Begley,et al.  Expression and characterization of recombinant vitamin K-dependent gamma-glutamyl carboxylase from an invertebrate, Conus textile. , 2002, European journal of biochemistry.

[16]  D. Yoshikami,et al.  Mu-conotoxin SmIIIA, a potent inhibitor of tetrodotoxin-resistant sodium channels in amphibian sympathetic and sensory neurons. , 2002, Biochemistry.

[17]  B. Olivera Conus Venom Peptides: Reflections from the Biology of Clades and Species , 2002 .

[18]  M. Zoli,et al.  Identification of the Nicotinic Receptor Subtypes Expressed on Dopaminergic Terminals in the Rat Striatum , 2002, The Journal of Neuroscience.

[19]  Li-Ming Zhou,et al.  Conantokin-L, a new NMDA receptor antagonist: determinants for anticonvulsant potency , 2002, Epilepsy Research.

[20]  D. Craik,et al.  Solution Structure of μ-Conotoxin PIIIA, a Preferential Inhibitor of Persistent Tetrodotoxin-sensitive Sodium Channels* , 2002, The Journal of Biological Chemistry.

[21]  B. Olivera,et al.  Conophysin-R, a Conus radiatus venom peptide belonging to the neurophysin family. , 2002, Toxicon : official journal of the International Society on Toxinology.

[22]  F. Tortella,et al.  Intrathecal CGX-1007 is neuroprotective in a rat model of focal cerebral ischemia , 2002, Neuroreport.

[23]  G. Bulaj,et al.  Conorfamide, a Conus venom peptide belonging to the RFamide family of neuropeptides. , 2002, Toxicon : official journal of the International Society on Toxinology.

[24]  Nobukuni Ogata,et al.  Molecular diversity of structure and function of the voltage-gated Na+ channels. , 2002, Japanese journal of pharmacology.

[25]  A. C. Collins,et al.  Involvement of the α3 Subunit in Central Nicotinic Binding Populations , 2002, The Journal of Neuroscience.

[26]  A. Robertson,et al.  The novel N-type calcium channel blocker, AM336, produces potent dose-dependent antinociception after intrathecal dosing in rats and inhibits substance P release in rat spinal cord slices , 2002, Pain.

[27]  J. Changeux,et al.  Distribution and Pharmacology of α6-Containing Nicotinic Acetylcholine Receptors Analyzed with Mutant Mice , 2002, The Journal of Neuroscience.

[28]  Reshma P Shetty,et al.  γ-Glutamyl carboxylation: An extracellular posttranslational modification that antedates the divergence of molluscs, arthropods, and chordates , 2002, Proceedings of the National Academy of Sciences of the United States of America.

[29]  H. Fozzard,et al.  Electrostatic and Steric Contributions to Block of the Skeletal Muscle Sodium Channel by μ-Conotoxin , 2002, The Journal of general physiology.

[30]  R. Schoenfeld,et al.  Post-translational modification: A two-dimensional strategy for molecular diversity of Conus peptides , 2002 .

[31]  A. Ménez Perspectives in molecular toxinology , 2002 .

[32]  B. Olivera,et al.  Venomous cone snails: molecular phylogeny and the generation of toxin diversity. , 2001, Toxicon : official journal of the International Society on Toxinology.

[33]  P. Ascenzi,et al.  Contryphan-Vn: a novel peptide from the venom of the Mediterranean snail Conus ventricosus. , 2001, Biochemical and biophysical research communications.

[34]  D. Yoshikami,et al.  δ-Conotoxin Structure/Function through a Cladistic Analysis† , 2001 .

[35]  L. Otvos,et al.  Point mutations identify the glutamate binding pocket of the N-methyl-d-aspartate receptor as major site of Conantokin-G inhibition , 2001, Neuropharmacology.

[36]  R. MacKinnon,et al.  Chemistry of ion coordination and hydration revealed by a K+ channel–Fab complex at 2.0 Å resolution , 2001, Nature.

[37]  Roderick MacKinnon,et al.  Energetic optimization of ion conduction rate by the K+ selectivity filter , 2001, Nature.

[38]  D. Craik,et al.  Two new classes of conopeptides inhibit the α1-adrenoceptor and noradrenaline transporter , 2001, Nature Neuroscience.

[39]  A. Kohn Maximal species richness in Conus: diversity, diet and habitat on reefs of northeast Papua New Guinea , 2001, Coral Reefs.

[40]  H. Fozzard,et al.  The Selectivity Filter of the Voltage-gated Sodium Channel Is Involved in Channel Activation* , 2001, The Journal of Biological Chemistry.

[41]  F. Castellino,et al.  The Amino Acid Residue at Sequence Position 5 in the Conantokin Peptides Partially Governs Subunit-selective Antagonism of RecombinantN-Methyl-d-aspartate Receptors* , 2001, The Journal of Biological Chemistry.

[42]  S. Schultz Principles of Neural Science, 4th ed. , 2001 .

[43]  Y. Gilad,et al.  Mechanisms for evolving hypervariability: the case of conopeptides. , 2001, Molecular biology and evolution.

[44]  M.,et al.  A Nicotinic Acetylcholine Receptor Ligand of Unique Specificity, a-Conotoxin ImI* , 2001 .

[45]  B. Olivera,et al.  Conotoxins, in retrospect. , 2001, Toxicon : official journal of the International Society on Toxinology.

[46]  A. L. Goldin,et al.  Resurgence of sodium channel research. , 2001, Annual review of physiology.

[47]  P. Andrews,et al.  Novel ω-Conotoxins from Conus catus Discriminate among Neuronal Calcium Channel Subtypes* , 2000, The Journal of Biological Chemistry.

[48]  H. Fozzard,et al.  μ-Conotoxin Giiia Interactions with the Voltage-Gated Na+ Channel Predict a Clockwise Arrangement of the Domains , 2000, The Journal of general physiology.

[49]  D. Yoshikami,et al.  Isolation and Characterization of a Novel ConusPeptide with Apparent Antinociceptive Activity* , 2000, The Journal of Biological Chemistry.

[50]  S. Donevan,et al.  Conantokin G is an NR2B-selective competitive antagonist of N-methyl-D-aspartate receptors. , 2000, Molecular pharmacology.

[51]  Single Amino Acid Substitutions in κ-Conotoxin PVIIA Disrupt Interaction with the Shaker K+ Channel* , 2000, The Journal of Biological Chemistry.

[52]  M. De Waard,et al.  Mechanisms of maurotoxin action on Shaker potassium channels. , 2000, Biophysical journal.

[53]  R M Jones,et al.  Conotoxins - new vistas for peptide therapeutics. , 2000, Current pharmaceutical design.

[54]  F. Tortella,et al.  Neuroprotective efficacy and therapeutic window of the high-affinity N-methyl-D-aspartate antagonist conantokin-G: in vitro (primary cerebellar neurons) and in vivo (rat model of transient focal brain ischemia) studies. , 2000, The Journal of pharmacology and experimental therapeutics.

[55]  Hydrophobic Pairwise Interactions Stabilize α-Conotoxin MI in the Muscle Acetylcholine Receptor Binding Site* , 2000, The Journal of Biological Chemistry.

[56]  W. Catterall,et al.  From Ionic Currents to Molecular Mechanisms The Structure and Function of Voltage-Gated Sodium Channels , 2000, Neuron.

[57]  Gail Mandel,et al.  Nomenclature of Voltage-Gated Sodium Channels , 2000, Neuron.

[58]  S. Sine,et al.  Pairwise Interactions between Neuronal α7Acetylcholine Receptors and α-Conotoxin PnIB* , 2000, The Journal of Biological Chemistry.

[59]  B. Olivera,et al.  The spasmodic peptide defines a new conotoxin superfamily. , 2000, Biochemistry.

[60]  A. Shcherbatko,et al.  Distinction among Neuronal Subtypes of Voltage-Activated Sodium Channels by μ-Conotoxin PIIIA , 2000, The Journal of Neuroscience.

[61]  S. Arneric,et al.  Agonists and Antagonists of Nicotinic Acetylcholine Receptors , 2000 .

[62]  B. Olivera ω-Conotoxin MVIIA: From Marine Snail Venom to Analgesic Drug , 2000 .

[63]  J. McIntosh Toxin antagonists of the neuronal nicotinic acetylcholine receptor , 2000 .

[64]  In vitro and in vivo characterization of conantokin-R, a selective NMDA receptor antagonist isolated from the venom of the fish-hunting snail Conus radiatus. , 2000, The Journal of pharmacology and experimental therapeutics.

[65]  D. Yoshikami,et al.  Effects of Conus peptides on the behavior of mice , 1999, Current Opinion in Neurobiology.

[66]  F. Castellino,et al.  Inhibition of NMDA-induced currents by conantokin-G and conantokin-T in cultured embryonic murine hippocampal neurons , 1999, Neuropharmacology.

[67]  B. Olivera,et al.  The T-superfamily of Conotoxins* , 1999, The Journal of Biological Chemistry.

[68]  D. Yoshikami,et al.  Single-Residue Alteration in α-Conotoxin PnIA Switches Its nAChR Subtype Selectivity† , 1999 .

[69]  D. Yoshikami,et al.  Critical residues influence the affinity and selectivity of alpha-conotoxin MI for nicotinic acetylcholine receptors. , 1999, Biochemistry.

[70]  A. Ménez,et al.  A new conotoxin isolated from Conus consors venom acting selectively on axons and motor nerve terminals through a Na+‐dependent mechanism , 1999, The European journal of neuroscience.

[71]  B. Olivera,et al.  Post-translationally modified neuropeptides from Conus venoms. , 1999, European journal of biochemistry.

[72]  S. Sine,et al.  Pairwise Interactions between Neuronal α7Acetylcholine Receptors and α-Conotoxin ImI* , 1999, The Journal of Biological Chemistry.

[73]  The Block of Shaker K+ Channels by κ-Conotoxin Pviia Is State Dependent , 1999, The Journal of general physiology.

[74]  S. Palumbi,et al.  Molecular genetics of ecological diversification: duplication and rapid evolution of toxin genes of the venomous gastropod Conus. , 1999, Proceedings of the National Academy of Sciences of the United States of America.

[75]  B. Olivera,et al.  Contulakin-G, an O-Glycosylated Invertebrate Neurotensin* , 1999, The Journal of Biological Chemistry.

[76]  P. Fossier,et al.  A conotoxin from Conus textile with unusual posttranslational modifications reduces presynaptic Ca2+ influx. , 1999, Proceedings of the National Academy of Sciences of the United States of America.

[77]  B. Olivera,et al.  Speciation of Cone Snails and Interspecific Hyperdivergence of Their Venom Peptides: Potential Evolutionary Significance of Introns a , 1999, Annals of the New York Academy of Sciences.

[78]  D. Craik,et al.  Effects of chirality at Tyr13 on the structure-activity relationships of omega-conotoxins from Conus magus. , 1999, Biochemistry.

[79]  J. McIntosh,et al.  Conus peptides as probes for ion channels. , 1999, Methods in enzymology.

[80]  J. McIntosh,et al.  Conus peptides targeted to specific nicotinic acetylcholine receptor subtypes. , 1999, Annual review of biochemistry.

[81]  B. Olivera,et al.  The contryphans, a D-tryptophan-containing family of Conus peptides: interconversion between conformers. , 2009, The journal of peptide research : official journal of the American Peptide Society.

[82]  D. Yoshikami,et al.  An O-glycosylated neuroexcitatory conus peptide. , 1998, Biochemistry.

[83]  D. Yoshikami,et al.  α-Conotoxin AuIB Selectively Blocks α3β4 Nicotinic Acetylcholine Receptors and Nicotine-Evoked Norepinephrine Release , 1998, The Journal of Neuroscience.

[84]  J. Kehoe,et al.  Two Distinct Nicotinic Receptors, One Pharmacologically Similar to the Vertebrate α7-Containing Receptor, Mediate Cl Currents inAplysia Neurons , 1998, The Journal of Neuroscience.

[85]  D. Julius,et al.  Inactivation of a serotonin-gated ion channel by a polypeptide toxin from marine snails. , 1998, Science.

[86]  R. Kallen,et al.  Extrapore residues of the S5-S6 loop of domain 2 of the voltage-gated skeletal muscle sodium channel (rSkM1) contribute to the mu-conotoxin GIIIA binding site. , 1998, Biophysical journal.

[87]  L. Pedersen,et al.  Conformational Changes in Conantokin-G Induced upon Binding of Calcium and Magnesium as Revealed by NMR Structural Analysis* , 1998, The Journal of Biological Chemistry.

[88]  David John Adams,et al.  α-Conotoxin EpI, a Novel Sulfated Peptide from Conus episcopatus That Selectively Targets Neuronal Nicotinic Acetylcholine Receptors* , 1998, The Journal of Biological Chemistry.

[89]  D. Yoshikami,et al.  μ-Conotoxin PIIIA, a New Peptide for Discriminating among Tetrodotoxin-Sensitive Na Channel Subtypes , 1998, The Journal of Neuroscience.

[90]  S. Sine,et al.  Structural Elements in α-Conotoxin ImI Essential for Binding to Neuronal α7 Receptors* , 1998, The Journal of Biological Chemistry.

[91]  S. Sine,et al.  Identification of Residues in the Neuronal α7Acetylcholine Receptor That Confer Selectivity for Conotoxin ImI* , 1998, The Journal of Biological Chemistry.

[92]  S. Zinn-Justin,et al.  Three-dimensional structure of kappa-conotoxin PVIIA, a novel potassium channel-blocking toxin from cone snails. , 1998, Biochemistry.

[93]  B. Chait,et al.  The structure of the potassium channel: molecular basis of K+ conduction and selectivity. , 1998, Science.

[94]  P. Taylor,et al.  Residues at the subunit interfaces of the nicotinic acetylcholine receptor that contribute to alpha-conotoxin M1 binding. , 1998, Molecular pharmacology.

[95]  B. Olivera,et al.  Conantokin-G Precursor and Its Role in γ-Carboxylation by a Vitamin K-dependent Carboxylase from a ConusSnail* , 1998, The Journal of Biological Chemistry.

[96]  J. C. Lodder,et al.  gamma-Conotoxin-PnVIIA, a gamma-carboxyglutamate-containing peptide agonist of neuronal pacemaker cation currents. , 1998, Biochemistry.

[97]  M. Foster,et al.  Three-dimensional solution structure of conotoxin psi-PIIIE, an acetylcholine gated ion channel antagonist. , 1998, Biochemistry.

[98]  W. Stühmer,et al.  κ-Conotoxin Pviia Is a Peptide Inhibiting theShaker K+ Channel* , 1998, The Journal of Biological Chemistry.

[99]  B. Olivera,et al.  E.E. Just Lecture, 1996. Conus venom peptides, receptor and ion channel targets, and drug design: 50 million years of neuropharmacology. , 1997, Molecular biology of the cell.

[100]  R. Harris-Warrick,et al.  Alternative Splicing in the Pore-Forming Region of shakerPotassium Channels , 1997, The Journal of Neuroscience.

[101]  Differential Targeting of Nicotinic Acetylcholine Receptors by Novel αA-Conotoxins* , 1997, The Journal of Biological Chemistry.

[102]  D. Yoshikami,et al.  A noncompetitive peptide inhibitor of the nicotinic acetylcholine receptor from Conus purpurascens venom. , 1997, Biochemistry.

[103]  J. McIntosh,et al.  α-Conotoxin MII Blocks Nicotine-Stimulated Dopamine Release in Rat Striatal Synaptosomes , 1997, The Journal of Neuroscience.

[104]  F. Ni,et al.  The NMR solution structure of the NMDA receptor antagonist, conantokin‐T, in the absence of divalent metal ions , 1997, FEBS letters.

[105]  J. Baleja,et al.  Three-dimensional structure of a gamma-carboxyglutamic acid-containing conotoxin, conantokin G, from the marine snail Conus geographus: the metal-free conformer. , 1997, Biochemistry.

[106]  R. Norton,et al.  Structure-function relationships of omega-conotoxin GVIA. Synthesis, structure, calcium channel binding, and functional assay of alanine-substituted analogues. , 1997, The Journal of biological chemistry.

[107]  P. Lyu,et al.  Role of modified glutamic acid in the helical structure of conantokin‐T , 1997, FEBS letters.

[108]  T. Stanley,et al.  Identification of a vitamin K‐dependent carboxylase in the venom duct of a Conus snail , 1997, FEBS letters.

[109]  S. Tavazoie,et al.  Differential block of nicotinic synapses on B versus C neurones in sympathetic ganglia of frog by α‐conotoxins MII and ImI , 1997, British Journal of Pharmacology.

[110]  H. Hirai,et al.  Molecular Determinants of Agonist Discrimination by NMDA Receptor Subunits: Analysis of the Glutamate Binding Site on the NR2B Subunit , 1997, Neuron.

[111]  J. Rivier,et al.  A Novel Post-translational Modification Involving Bromination of Tryptophan , 1997, The Journal of Biological Chemistry.

[112]  K. Han,et al.  NMR structure determination of a novel conotoxin, [Pro 7,13] alpha A-conotoxin PIVA. , 1997, Biochemistry.

[113]  C. Roumestand,et al.  On the Convergent Evolution of Animal Toxins , 1997, The Journal of Biological Chemistry.

[114]  J. Rivier,et al.  Bromocontryphan: post-translational bromination of tryptophan. , 1997, Biochemistry.

[115]  D. Craik,et al.  Determination of the Solution Structures of Conantokin-G and Conantokin-T by CD and NMR Spectroscopy* , 1997, The Journal of Biological Chemistry.

[116]  Y. Jan,et al.  Cloned potassium channels from eukaryotes and prokaryotes. , 1997, Annual review of neuroscience.

[117]  B. Olivera,et al.  Contryphan Is a D-Tryptophan-containing Conus Peptide* , 1996, The Journal of Biological Chemistry.

[118]  F. Conti,et al.  Use dependence of tetrodotoxin block of sodium channels: a revival of the trapped-ion mechanism. , 1996, Biophysical journal.

[119]  M. Stocker,et al.  MicroO-conotoxin MrVIA inhibits mammalian sodium channels, but not through site I. , 1996, Journal of neurophysiology.

[120]  J. C. Lodder,et al.  A novel hydrophobic omega-conotoxin blocks molluscan dihydropyridine-sensitive calcium channels. , 1996, Biochemistry.

[121]  Walter Stühmer,et al.  Strategy for rapid immobilization of prey by a fish-hunting marine snail , 1996, Nature.

[122]  D. Yoshikami,et al.  A New -Conotoxin Which Targets 32 Nicotinic Acetylcholine Receptors (*) , 1996, The Journal of Biological Chemistry.

[123]  R. Horn,et al.  Interactions between a Pore-Blocking Peptide and the Voltage Sensor of the Sodium Channel: An Electrostatic Approach to Channel Geometry , 1996, Neuron.

[124]  D. Groebe,et al.  alpha-Conotoxin EI, a new nicotinic acetylcholine receptor antagonist with novel selectivity. , 1995, Biochemistry.

[125]  H. Fozzard,et al.  A mu-conotoxin-insensitive Na+ channel mutant: possible localization of a binding site at the outer vestibule. , 1995, Biophysical journal.

[126]  D. Yoshikami,et al.  A New Family of Conus Peptides Targeted to the Nicotinic Acetylcholine Receptor (*) , 1995, The Journal of Biological Chemistry.

[127]  M. Spira,et al.  A New Family of Conotoxins That Blocks Voltage-gated Sodium Channels (*) , 1995, The Journal of Biological Chemistry.

[128]  J. C. Lodder,et al.  A new cysteine framework in sodium channel blocking conotoxins. , 1995, Biochemistry.

[129]  R. Maeda,et al.  Molecular dissection of subunit interfaces in the acetylcholine receptor: Identification of determinants of α-Conotoxin M1 selectivity , 1995, Neuron.

[130]  K. Tarczy-Hornoch,et al.  Structure-activity analysis of a Conus peptide blocker of N-type neuronal calcium channels. , 1995, Biochemistry.

[131]  W R Gray,et al.  Purification, characterization, synthesis, and cloning of the lockjaw peptide from Conus purpurascens venom. , 1995, Biochemistry.

[132]  R. MacKinnon,et al.  Revealing the architecture of a K+ channel pore through mutant cycles with a peptide inhibitor. , 1995, Science.

[133]  M. Spira,et al.  Alterations of voltage-activated sodium current by a novel conotoxin from the venom of Conus gloriamaris. , 1995, Journal of neurophysiology.

[134]  M. Gelb,et al.  Conodipine-M, a Novel Phospholipase A2 Isolated from the Venom of the Marine Snail Conus magus(*) , 1995, The Journal of Biological Chemistry.

[135]  J. Luebke,et al.  Exocytotic Ca2+ channels in mammalian central neurons , 1995, Trends in Neurosciences.

[136]  A. Kohn,et al.  Manual of the living conidae , 1995 .

[137]  G Schreiber,et al.  Energetics of protein-protein interactions: analysis of the barnase-barstar interface by single mutations and double mutant cycles. , 1995, Journal of molecular biology.

[138]  M. Adams,et al.  Neurotoxins: overview of an emerging research technology , 1994, Trends in Neurosciences.

[139]  A. Ogura,et al.  Hydroxyl group of Tyr13 is essential for the activity of omega-conotoxin GVIA, a peptide toxin for N-type calcium channel. , 1994, The Journal of biological chemistry.

[140]  W R Gray,et al.  Delta-conotoxin GmVIA, a novel peptide from the venom of Conus gloriamaris. , 1994, Biochemistry.

[141]  A. Burlingame,et al.  New mollusc-specific alpha-conotoxins block Aplysia neuronal acetylcholine receptors. , 1994, Biochemistry.

[142]  D. Yoshikami,et al.  A nicotinic acetylcholine receptor ligand of unique specificity, alpha-conotoxin ImI. , 1994, The Journal of biological chemistry.

[143]  R. Rogart,et al.  Post-repolarization block of cloned sodium channels by saxitoxin: the contribution of pore-region amino acids. , 1994, Biophysical journal.

[144]  M. Adams,et al.  CALCIUM CHANNEL DIVERSITY AND NEUROTRANSMITTER RELEASE : THE OMEGA -CONOTOXINS AND OMEGA -AGATOXINS , 1994 .

[145]  P. Skolnick,et al.  Noncompetitive Inhibition of N‐Methyl‐D‐Aspartate by Conantokin‐G: Evidence for an Allosteric Interaction at Polyamine Sites , 1992, Journal of neurochemistry.

[146]  W. Catterall,et al.  Cellular and molecular biology of voltage-gated sodium channels. , 1992, Physiological reviews.

[147]  R. Horn,et al.  Chimeric study of sodium channels from rat skeletal and cardiac muscle , 1992, FEBS letters.

[148]  R. Gordon,et al.  Action of derivatives of mu-conotoxin GIIIA on sodium channels. Single amino acid substitutions in the toxin separately affect association and dissociation rates. , 1992, Biochemistry.

[149]  D. Yoshikami,et al.  Conantokin-G selectively inhibits N-methyl-D-aspartate-induced currents in Xenopus oocytes injected with mouse brain mRNA. , 1992, European journal of pharmacology.

[150]  B. Bean,et al.  A new conus peptide ligand for mammalian presynaptic Ca2+ channels , 1992, Neuron.

[151]  J. Rivier,et al.  Conantokin-G: A novel peptide antagonist to the N-methyl-d-aspartic acid (NMDA) receptor , 1990, Neuroscience Letters.

[152]  S. Woodward,et al.  Diversity of Conus neuropeptides. , 1990, Science.

[153]  J. Haack,et al.  Conantokin-T. A gamma-carboxyglutamate containing peptide with N-methyl-d-aspartate antagonist activity. , 1990, The Journal of biological chemistry.

[154]  S. Woodward,et al.  Constant and hypervariable regions in conotoxin propeptides. , 1990, The EMBO journal.

[155]  Robert W. Williams,et al.  Peptides from Conus Venoms which Affect Ca++ Entry into Neurons , 1990 .

[156]  U. Lönnendonker Use-dependent block of sodium channels in frog myelinated nerve by tetrodotoxin and saxitoxin at negative holding potentials. , 1989, Biochimica et biophysica acta.

[157]  J. Rivier,et al.  mu-conotoxin GIIIA, a peptide ligand for muscle sodium channels: chemical synthesis, radiolabeling, and receptor characterization. , 1989, Biochemistry.

[158]  S. Woodward,et al.  A molluscivorous Conus toxin: conserved frameworks in conotoxins. , 1989, Biochemistry.

[159]  W R Gray,et al.  Peptide toxins from venomous Conus snails. , 1988, Annual review of biochemistry.

[160]  B. Olivera,et al.  Invertebrate vasopressin/oxytocin homologs. Characterization of peptides from Conus geographus and Conus straitus venoms. , 1987, The Journal of biological chemistry.

[161]  J. McIntosh,et al.  Neuronal calcium channel antagonists. Discrimination between calcium channel subtypes using omega-conotoxin from Conus magus venom. , 1987, Biochemistry.

[162]  E. Carmeliet Voltage-dependent block by tetrodotoxin of the sodium channel in rabbit cardiac Purkinje fibers. , 1987, Biophysical journal.

[163]  T. Narahashi,et al.  Use‐ and Voltage‐Dependent Block of the Sodium Channel by Saxitoxin , 1986, Annals of the New York Academy of Sciences.

[164]  C. Y. Kao,et al.  Tetrodotoxin, saxitoxin, and the molecular biology of the sodium channel. , 1986, Annals of the New York Academy of Sciences.

[165]  G. Strichartz,et al.  Discrimination of muscle and neuronal Na-channel subtypes by binding competition between [3H]saxitoxin and mu-conotoxins. , 1986, Proceedings of the National Academy of Sciences of the United States of America.

[166]  M. Alexander,et al.  Principles of Neural Science , 1981 .

[167]  W R Gray,et al.  Peptide neurotoxins from fish-hunting cone snails. , 1985, Science.

[168]  D. Yoshikami,et al.  Conus geographus toxins that discriminate between neuronal and muscle sodium channels. , 1985, The Journal of biological chemistry.

[169]  D. Middlemas,et al.  A sleep-inducing peptide from Conus geographus venom. , 1985, Toxicon : official journal of the International Society on Toxinology.

[170]  B. Hille,et al.  Ionic channels of excitable membranes , 2001 .

[171]  J. McIntosh,et al.  Gamma-carboxyglutamate in a neuroactive toxin. , 1984, The Journal of biological chemistry.

[172]  J. McIntosh,et al.  Purification and sequence of a presynaptic peptide toxin from Conus geographus venom. , 1984, Biochemistry.

[173]  S. Yoshiba [An estimation of the most dangerous species of cone shell, Conus (Gastridium) geographus Linne, 1758, venom's lethal dose in humans]. , 1984, Nihon eiseigaku zasshi. Japanese journal of hygiene.

[174]  Y. Ohizumi,et al.  The amino acid sequences of homologous hydroxyproline‐containing myotoxins from the marine snal Conus geographus venom , 1983, FEBS letters.

[175]  M. Hunkapiller,et al.  Isolation and structure of a peptide toxin from the marine snail Conus magus. , 1982, Archives of biochemistry and biophysics.

[176]  R. Tsien,et al.  Tetrodotoxin block of sodium channels in rabbit Purkinje fibers. Interactions between toxin binding and channel gating , 1981, The Journal of general physiology.

[177]  B. Olivera,et al.  Peptide toxins from Conus geographus venom. , 1981, The Journal of biological chemistry.

[178]  B. Olivera,et al.  Purification and properties of a myotoxin from Conus geographus venom. , 1978, Archives of biochemistry and biophysics.

[179]  G. Ruggieri Drugs from the sea. , 1976, Science.

[180]  H. Reuter,et al.  Voltage-dependent action of tetrodotoxin in mammalian cardiac muscle , 1976, Nature.

[181]  R. Endean,et al.  Pharmacology of the venom of Conus geographus. , 1974, Toxicon : official journal of the International Society on Toxinology.

[182]  A. Kohn,et al.  PRELIMINARY STUDIES ON THE VENOM OF THE MARINE SNAIL CONUS * , 1960, Annals of the New York Academy of Sciences.

[183]  A. Kohn The Ecology of Conus in Hawaii , 1959 .

[184]  A. Hodgkin,et al.  Measurement of current‐voltage relations in the membrane of the giant axon of Loligo , 1952, The Journal of physiology.

[185]  A. Hodgkin,et al.  Currents carried by sodium and potassium ions through the membrane of the giant axon of Loligo , 1952, The Journal of physiology.

[186]  J. McIntosh,et al.  Carboxyglutamate in a Neuroactive Toxin , 2022 .