340
Views
7
CrossRef citations to date
0
Altmetric
Research Article

Snake Venom Neurotoxins: Pharmacological Classification

Pages 37-96 | Published online: 10 Oct 2008

References

  • Abe T., Limbrick A. R., Miledi R. Acute muscle denervation by β‐bungarotoxin. Proc.Roy.Soc. Lond. (Biol.) 1976; 194: 545–553
  • Aird S. D., Womble G. C., Yates J. R., Yates J. R., III, Griffin P. R. Primary structure of gamma‐bungarotoxin, a new postsynaptic neurotoxin from venom of Bungarus multicinctus. Toxicon 1999; 37(4)609–625, [PUBMED], [INFOTRIEVE], [CSA], [CROSSREF]
  • Anadon A., Martinez‐Larranaga M. R. Effects of crotoxin on autonomic neuro‐ transmission in the guinea pig myenteric plexus and vas deference. Toxicon 1985; 23(6)963–972, [PUBMED], [INFOTRIEVE], [CROSSREF]
  • Anderson A. J., Harvey A. L., Mbugua P. M. Effects of fasciculin‐2 an anticholinesterase polypeptide from green mumba venom, a neuromuscular transmission in mouse diaphragm preparation. Neurosci. Lett. 1985; 54: 123–128, [PUBMED], [INFOTRIEVE]
  • Antil S., Servent D., Menez A. Variability among the sites by which curaremimetic toxins bind to torpedo acetylcholine receptor, as revealed by identification of the functional residues of alpha‐cobrotoxin. J. Biol. Chem. 1999; 274: 34851–34858, [PUBMED], [INFOTRIEVE], [CROSSREF]
  • Aubrey N., Deraux C., Sizaret P. Y., Rochat H., Goyffon M., Billiald P. Design and evaluation of a diabody to improve protection against a potent scorpion neurotoxin. Cell Mol. Life Sci. 2003; 60(3)617–628, [PUBMED], [INFOTRIEVE], [CSA], [CROSSREF]
  • Badin C. A., Vassallo J., Hyslop S., Prodo‐Frnceshi J. Convulxin induced pulmonary thrombosis in mice. 13th World Congress of IST, Paris France, Sept., 18–222000, 181
  • Bengis R. G., Noble D. F. Postsynaptic blockade of neuromuscular transmission by Toxin II from the venom of the South ringhals cobra Hemachatus hemachatus venom. Toxicon 1976; 14: 167–173, [PUBMED], [INFOTRIEVE], [CROSSREF]
  • Benshin C. G. Potassium channel blockade by β‐subunit of β bungarotoxin. Mol. Pharmacol. 1996; 38: 164–169, [CSA]
  • Beven P., Hiestand P., Beta R. T. X. A receptor active protein from Russell's viper (Vipera russelli russelli) venom. J. Biol. Chem. 1983; 258(8)5319–5326
  • Bhattacharya S. Pharmacological studies on the venom of Echis carinatus. Ph.D. pharmacology thesis. University of Bombay, Mumbai 1981
  • Bieber A. L., Tu T., Tu A. T. Studies of an acidic cardiotoxin isolated from the venom of Mojave rattle snake Crotalus scutulatus. Biochem. Biophys. Acta. (Amst.) 1975; 400: 178–188
  • Black A. R., Dolly J. O. Two acceptor sub‐types for dendrotoxin in chick synaptic membranes distinguishable by‐bungarotoxin. Eur. J. Biochem. 1966; 156: 609–617
  • Bloom F. E. Drugs acting on the central nervous system. “Neurotransmission and the central nervous system.”. Goodman and Gilman's the Pharmacological Basis of Therapeutics10th Ed., Hardman, Limbird, Gilman. McGraw Hill. 2001; 298, Ch. 12
  • Bon C., Changeux J. P. An acidic neurotoxin from the venom of Bungarus ceruleus which blocks the response to an agonist without binding to the cholinergic receptor site. FEBS Lett. 1975; 52: 212–216, [CROSSREF]
  • Bon C. Ceruleotoxin, an acidic neurotoxin from Bungarus ceruleus venom which blocks postsynaptically neuromuscular transmission without binding to the cholinergic receptor site. Bull. Inst. Pasteur Lille 1976; 74: 41–45
  • Bon C., Changeux J. P., Jeng T. W., Frankel‐Conrat H. Postsynaptic effects of crotoxin and its isolated subunits. Eur. J. Biochem. 1979; 99: 471, [PUBMED], [INFOTRIEVE]
  • Bourque C. W. Transient calcium dependent potassium current in magnocellular neurosecretory cells of the rat supra optic nucleus. J. Physiol. 1988; 397: 331–347, [PUBMED], [INFOTRIEVE]
  • Bowman W. C., Gibb A. L., Harvey A. L., Marshall I. G. Prejunctional actions of cholinoceptor agonists and antagonists and of anticholinesterase drugs. New Neuromuscular Blocking Agents, Basic and Applied Aspects, A. Kharkevich. Springer‐Verlag, Berlin 1986; 141‐A
  • Bradly K. N., Rowan E. G., Jerusalnsky D., Harvey A. L. Characterisation of muscarinic toxins MT1, MT2, MT3 and MT7. Toxicon 1998; 36(9)1273
  • Brazil O. V. Neurotoxins from the South American rattle snake venoms. Formos. Med. Ass. 1972; 71: 394–400
  • Brazil O. V., Excell B. J. Action of crotoxin and crotactin from the venom of Crotalus durissus terrificus (south rattle snake) on the frog neuromuscular junction. J. Physiol. (Lond.) 1971; 212: 34–35
  • Brazil O. V., Prado‐Franceschi J., Laure C. J. Repetitive muscle responses induced by Crotamine. Toxicon 1979; 17: 61, [PUBMED], [INFOTRIEVE], [CROSSREF]
  • Breithaupt H. Neurotoxic and myotoxic effects of Crotalus phospholipase A and its complex with crotapotin. Naunyn‐Schmiedeberg's Arch. Pharmacol. 1976; 292: 271–276, [CROSSREF]
  • Breithaupt H., Rubesaman K., Habermann E. Biochemistry and pharmacology of the crotoxin complex, biochemical analysis of crotapotin and the basic phospholipase A. Eur. J. Biochem. 1974; 49: 333–345, [PUBMED], [INFOTRIEVE]
  • Breithaupt H., Ouori‐Satosh T., Lang J. Isolation and characterisation of three phospholipase A from the crotoxin complex. Biochem. Biophys. Acta (Amst.). 1975; 403: 355–361
  • Brejc K., Celie P., Dijk P. V., Smit A. B., Sixma T. K. 14th World Congress on Animal, Plant and Microbial Toxins. September, 2003; 78, 05101, 3AH1, Adelaide
  • Brook G. A., Torres L. F., Gopalkrishnakone P., Duchan L. W. Effects of phosopholipase of Enhydrina schistosa venom on nerve, motor end plate and muscle of the mouse. J. Exp. Physiol. 1987; 72: 571–591
  • Brose N., Petrenko A. G., Sudhof T. C., Jahn R. Synaptotagmin, A calcium sensor on the synaptic vesicle surface. Science 1992; 256: 1021–1025, [PUBMED], [INFOTRIEVE]
  • Camillo M. A., Arruda Paes P. C., Troncone L. R., Rogero J. R. Gyroxin fails to modify in vitro release of labelled dopamine and acetylcholine from rat and mouse striatal tissue. Toxicon 2001; 39(6)843–853, [PUBMED], [INFOTRIEVE], [CSA], [CROSSREF]
  • Caputi A., Yamu Y., Otterson O. P. Epileptica limbica en la rata provocada p,or Dendrotoxin un polipeptido ais lado del venenodela Mamba verde (Dendrospis angusticeps). Arch. Biol. Med. Exp. 1980; 12: 125
  • Carsi J. M., Valentine H. H., Valentine H. H., Potter L. T. M2‐toxin: a selective ligand for M2 muscarinic receptors. Mol. Pharmacol. 1999; 56(5)933–937, [PUBMED], [INFOTRIEVE], [CSA]
  • Chaeumet V., Saliou B., Fideler L., Chen Y. C., Gubensek F., Bon C., Delot E. Snake venom PLA2 NTX potentiation of a single chain NTX by the chaperon subunit of two component NTX. J. Biol.Chem. 2001; 276(16)12493–12496, [CSA], [CROSSREF]
  • Chang C. C., Hong S. J. Dissociation of the end‐plate potential run‐down and tetanic fade from the postsynaptic inhibition of acetylcholine receptor by α‐neurotoxins. J. Exp. Neurol. 1987; 98: 509–517, [CROSSREF]
  • Chang C. C., Lee C. Y. Isolation of neurotoxin from the venom of Bungarus ulticinctus and their modes of neuromuscular blocking action. Arch. Int. Pharmacodyn. 1963; 144: 316–342
  • Chang C. C., Lee C. Y. Electrophysiological study of neuromuscular blocking action of cobra neurotoxin. Brit. J. Pharmacol. 1966; 28: 172–181
  • Chang C. C., Chen T. F., Lee C. Y. Studies of the presynaptic effects of β‐bungarotoxin on neuromuscular transmission. J. Pharmacol. Exp.Ther. 1973; 184: 339–345, [PUBMED], [INFOTRIEVE]
  • Chang C. C., Lee J. D., Eaker D., Fohlman J. The presynaptic neuromuscular blocking action of taipoxin. A comparison with β‐bungarotoxin and crotoxin. Toxicon 1977; 15: 571–576, [PUBMED], [INFOTRIEVE], [CROSSREF]
  • Chang L. S., Chou Y. C., Lin S. R., Wu B. N., Lin J., Hong E., Hsiao Y. J. A novel neurotoxin, cobrotoxin b, from Naja naja atra (Taiwan cobra) venom. Purification, characterisation and gene characterisation. J. Biochem. 1997; 122(6)1252–1259, [PUBMED], [INFOTRIEVE]
  • Chang L., Lin S., Wang J., Hu W. P., Wu B., Huang H. Structure function studies on Taiwan cobra long neurotoxin homolog. Biochim. Biophys. Acta. 2000; 1480(1–2)293–301, [PUBMED], [INFOTRIEVE]
  • Changeux J. P. Chemical signalling in the brain. Sci. Am. 1993; 269: 58–62, [PUBMED], [INFOTRIEVE]
  • Changeux J. P., Kasai M., Lee C. Y. Use of a snake venom toxins to characterise the cholinergic receptor protein. Proc. Natl. Acad. Sci. 1970; 67: 1241–1247, [PUBMED], [INFOTRIEVE]
  • Chapell R., Rosenberg P. Specificity of action of β‐bungarotoxin on ACh release from synaptosomes. Toxicon 1992; 30: 621–633, [PUBMED], [INFOTRIEVE], [CROSSREF]
  • Chen J. L., Lee C. Y. Ultrastructural changes in the motor nerve terminals caused by β‐bungarotoxin. Virchows Arch. Abt. Zell Path. 1970; 6: 318–325
  • Cheymol J., Gonsalves J. M., Bourillet F., Roch‐Arveiller M. Action neuromuscularie Comparee de la crotamine et du venin de Crotalus durissus terrificus var. Crotaminicus.I. Sur preparations neuromusculare in situ. Toxicon 1971a; 9: 279–286, [PUBMED], [INFOTRIEVE], [CROSSREF]
  • Cheymol J., et al. Sur Preparations isolees. Toxicon 1971b; 9: 287–289, [PUBMED], [INFOTRIEVE], [CROSSREF]
  • Cheymol J., Tamiya N., Douvillet F., Roch‐Arreiller M. Neuromuscular action of venom of the sea snake erabu Laticauda semifasciata and erabutoxin a and b. Toxicon 1972; 10: 125–131, [PUBMED], [INFOTRIEVE], [CROSSREF]
  • Chiappinelli V. A. Kappa bungarotoxin a probe for the neuronal nicotinic receptor in the avian ciliary ganglion. Brain Res. 1983; 277: 9–22, [PUBMED], [INFOTRIEVE], [CROSSREF]
  • Chiappinelli V. A., Dryer S. E. Nicotinic transmission in sympathetic ganglia blockade by snake venom neurotoxins Kappa neurotoxin. Neurosci. Lett. 1984; 50: 239–244, [PUBMED], [INFOTRIEVE], [CROSSREF]
  • Chiappinelli V. A., Wolf K. M., DeBin J. A., Holt I. L. Kappa‐flavotoxin, isolation of a new neuronal nicotinic receptor antagonist that is structurally related to kappa bungarotoxin. Brain Res. 1987; 402: 21–29, [PUBMED], [INFOTRIEVE], [CROSSREF]
  • Chiappinelli V. A., Weaver W. R., Mclane K. E., Conti Fine B. M., Fiordalisi J. J., Grant J. J. Binding of native neurotoxins and site directed mutants to nicotinic acetylcholine receptors. Toxicon 1996; 34(11–12)1243–1256, [PUBMED], [INFOTRIEVE], [CSA], [CROSSREF]
  • Chung M. C., Tan N. H., Armugarn A. The amino acid sequence of two postsynaptic neurotoxins isolated from Malayan cobra Naja naja sputatrix venom. Toxicon 1994; 32(11)1471–1474, [PUBMED], [INFOTRIEVE], [CSA], [CROSSREF]
  • Cull‐Candy S. G., Fohlman J., Gustavsson D., Rouch R., Thesleff S. The effects of taipoxin and notexin on the function and fine structure of the murine neuromuscular junction. Neuroscience 1976; 1: 175–180, [CROSSREF]
  • Daguillanes V., Eon‐Gerhardt R. Determination of the LD50 of Ls III toxin) from Laticauda semifasciata) and of Cbtx toxin (from Naja kaothia) on sea fish Serranus cabrilla (O Perciforms). Toxicon 1998; 36(12)1734–1737
  • Dajas F., Bolioli B., Castello M., Silveria R. Rat striatal acetylcholinesterase inhibition by fasciculin 2 a polypeptide from green mamba snake venom. Neurosci. Lett. 1987; 77: 87–91, [PUBMED], [INFOTRIEVE], [CROSSREF]
  • Demangel C., Maroun R. C., Rouyne S., Bon C., Mazie J. C., Choumet V. Combining phage display and molecular modeling to map the epitope of neutralising antitoxin antibody. Eur. J. Biochem. 2000; 267: 2365
  • De Weille J. R., Schweitz H., Maes P., Tartar A., Lazdunski M. Calciseptine, a peptide isolated from black mamba venom, is a specific blocker of the L‐type calcium channel. Proc. Natl. Acad. Sci. U. S. A. 1991; 88: 2437–2440, [PUBMED], [INFOTRIEVE], [CSA]
  • Docherty R. J., Dolly J. O., Halliwell J. V., Othman I. Excitatory effects of dendrotoxin on the hippocampus in vitro. J. Physiol. 1983; 336: 58–59
  • Dolly J. O., Halliwell J. V., Black J. D., William R. S., Pelche N., Matthews A., Breeze A. L., Mehraban F., Othman J. B., Black A. R. Botulinum toxin and dendrotoxin as probes for studies on transmitter release. J. Physiol., (Paris) 1984; 79: 280–303
  • Doorty K. B., Beran S., Wadsworth D. D. F., Srong P. N. A new SkCa channel toxin from Oxyuranus scutellatus taipan venom. Toxicon 1998; 36(9)1250
  • Dorandeu F., Pernot‐Marino I., Laaernent I. Biochemical and neurotoxicological characterisation of paradoxin, a neurotoxin from australian inland taipan snake Oxyurunus microlepidotus venom. Toxicon 1996a; 34(3)311, [CROSSREF]
  • Dorandeu F., Pernot‐Marino I., Brochier G., Delorme E., Lallement G. Effects of paradoxin, aneurotoxin from australian inland taipan snake Oxyranus microlepidotus venom: studies in rodent central nervous system. Toxicon 1996b; 34(10)1087–1088, [CROSSREF]
  • Dowdall M. J., Fohlman J. P., Watts A. Presynaptic action of snake venom neurotoxin on cholinergic system. Adv. Cytopharmacol. 1979; 3: 63–76, [PUBMED], [INFOTRIEVE]
  • Dryden W. F., Harvey A. L. Effect of receptor desensitisation on the action of α bungarotoxin on cultured skeletal muscle. Br. J. Pharmacol. 1974; 51: 456–458, [PUBMED], [INFOTRIEVE]
  • Excell B. J., Patel R. Characterisation of toxic fractions of Dendrospis jamensoni venom. J. Physiol. (Lond.) 1972; 229: 29–30
  • Fathi B., Rowan E. G., Harvey A. L. The facilitatory actions of snake venom phospholipase A2 neurotoxins at the neuromuscular junction are not mediated through voltage gated K+ channel. 13th World Congress of International Society of Toxinology, Paris, Sept., 18–222000, 18–22, L‐91
  • Faure G., Copic A., Leporrier S., Gubensek F., Krizaj J., Bon C. Solublisation and purification of the crotoxin acceptor from Torpedo marmorata electric organ. XIII World Congress of IST, Toxins From Animal, Plant and Microbes, Paris, Sept, 18–222000, Abstract: P. 130
  • Fohlman J., Eaker D. Isolation and characterisation of a lethal myotoxic PLA2 from the venom of sea snake Enhydrina schistosa causing myoglobinurea in mice. Toxicon 1977; 13: 345–354
  • Fohlman J., Eaker D., Karlsson E., Thesleff S. Taipoxin an extremely potent presynaptic neurotoxin from the venom of the Australian snake taipen Oxyranus scutellates. Eur. J. Biochem. 1976; 68: 457–469, [PUBMED], [INFOTRIEVE]
  • Francischetti I. M. B., Saliou B., Leduc M. M., Carilini C. R., Hatmi M., Randon J., Faili Bon C. Convulxin, a potent platelet aggregating protein from Crotalus durissus terrificus venom, specifically binds to platelets. Toxicon 1997; 35(8)1217–1228, [PUBMED], [INFOTRIEVE], [CSA], [CROSSREF]
  • Gawade S. P. Pharmacological and Biochemical Studies on the Venom of Enhydrina Schistosa (Common Sea Snake). Ph.D. (Tech.) pharmacology thesis. Bombay Univ, Mumbai 1981
  • Gawade S. P. Comparative evaluation of mechanism of neuromuscular (N–M) blockade between Enhydrotoxin‐a (EsNTx‐a) and D‐tubocurarine (d‐tc). J. Natl. Toxins 1998; 7: 95–96, [CSA]
  • Gawade S. P., Bhide M. B. Chromatographic separation of the venom of Lapemis curtus (short sea snake) and pharmacological characterisation of its toxic components. Preliminary studies. Abstract Toxicon 1978; 16: 134, [CSA]
  • Gawade S. P., Gaitonde B. B. Presynaptic and postsynaptic sites of action of enhydrotoxin‐a (EsNTx‐a) from the venom of Enhydrina schistosa (common sea snake). J. Pharm. Pharmacol. 1982a; 34: 782–787
  • Gawade S. P., Gaitonde B. B. Isolation and characterisation of toxic components from the venom of common Indian sea snake (Enhydrina schistosa). Toxicon 1982b; 20: 797–801, [PUBMED], [INFOTRIEVE], [CSA], [CROSSREF]
  • Gawade S. P., Buduk D. P., Gaitonde B. B. Preparation of monovalent Enhydrina schistosa (common sea snake) antivenin. Ind. J. Med. Res. 1980; 72: 742–752
  • Gawade S. P., Tare T. G., Gaitonde B. B. Simple method of milking of sea snakes. Snake 1981; 13: 154–157
  • Geh S. L., Rampal R. M. Neuromuscular and cardiovascular effects of mulgotoxin isolated from the venom of the Australian king brown snake Pseudechis australis. Toxicon 1997; 35(4)479, [CROSSREF]
  • Geh S. L., Toh H. T. The effects of sea snake neurotoxins and a phospholipase fraction on the ultrastructure of mammalian skeletal muscle. Abstract: South East Asian Western Pacific Regional meeting of Pharmacologists, Singapore, May, 1976, No. 33
  • Geh S. L., Rowan E. G., Harvey A. L. Neuromuscular effects of four phospholipase A2 from the venom of Pseudechis australis, the australian king brown snake. Toxicon 1992; 30: 1051–1057, [PUBMED], [INFOTRIEVE], [CSA], [CROSSREF]
  • Georgieva D. N., Betzel C., Aleksiev B., Genov N. Spectroscopic investigation of calcium binding sites in the neurotoxin vipoxin and its componments relation with the X‐ray structure. Spectrochim. Acta, Part A: Mol. Biomol. Spectrosc. 2000; 56(14)2811–2816, [CSA], [CROSSREF]
  • Germain N., Merienne K., Zinn‐Justin S., Boulain J‐C., Menez A., Ducancel F., Menez A. Molecular and structural basis of the specificity of neutralising acetylcholine receptor mimicking antibody, using combined mutational and molecular modeling analyses. J. Biol.Chem. 2000; 275(28)21578–21586, [PUBMED], [INFOTRIEVE], [CSA], [CROSSREF]
  • Gomes A., Pallabi D. E. Hanahpep: a novel fibrinolytic peptide from the Indian king cobra (Ophiophagus hannah) venom. Biochem. Biophys. Res. Commun. 1999; 266(2)488–491, [PUBMED], [INFOTRIEVE], [CSA], [CROSSREF]
  • Gong N., Armugam A., Jeyaseelan K. Postsynaptic short chain neurotoxin from Pseudonaja textilis, cDNA cloning, expression and protein characterisation. Eur. J. Biochem. 1999; 265(3)982–989, [PUBMED], [INFOTRIEVE], [CROSSREF]
  • Gopalakrinshnan M. The voltage sensitive calcium channels, pharmacology of receptors and ion transporters. Pharma.Soc., G. Jagdeesh. Tamilnadu, India 1995; 189–219
  • Gopalkrishnakone P., Hawgood B. J. Morphological changes induced by crotoxin in murine nerve and neuromuscular junction. Toxicon 1984; 22: 791–804, [CROSSREF]
  • Gopalkrishnakone P., Hawgood B. J., Holbrooke S. E., Marsh N. A., Sautana de Sa S., Tu A. T. Site of action of Mojave toxin isolated from the venom of Crotalus scutalatus scutalatus in comparison with the action of crotoxin from the venom of Crotalus durrisus terrificus. Toxicon 1979; 17(suppl.)
  • Gopalkrishnakone P., Dampstra D. W., Hawgood B. J., Elder H. Y. Cellular and mitochondrial changes induced in the structure of murine skeletal muscle by crotoxin, a neurotoxic phospholipase A2. Toxicon, 22: 85–98, [CSA], [CROSSREF]
  • Grant G. A., Frazier M. W., Chiappinelli V. A. Amino acid sequence of kappa flavotoxin establishment of a new family of snake venom neurotoxins. Biochemistry 1988; 27: 3798–3799
  • Gregoire J., Rochat H. Amino acid sequences of neurotoxins I and III of the elapidae snake Naja mossambica mossambica. Eur. J. Biochem. 1977; 80: 283–293, [PUBMED], [INFOTRIEVE]
  • Halliwell J. V., Othman I. B., Pelche N., Matthew A., Dolly J. O. Central action of dendrotoxin selective reduction of a transient K+ conductance in hippocampus and binding to localised acceptor. Proc. Natl. Acad. Sci. U. S. A. 1986; 83: 493–497, [PUBMED], [INFOTRIEVE]
  • Halpert J., Fohlmen J., Eaker D. Amino acid sequences of a postsynaptic neurotoxin from the venom of the Australian tiger snake Notechis scutatus scutatus
  • Harris J. B. Presynaptically active neurotoxins and the degeneration and regeneration of peripheral neuromuscular systems. 13th World Congress of IST, Paris, Sept., 18–222000, Abstract: L. 84
  • Harris J. B., Zar M. A. The effects of a toxin isolated from Australian tiger snake Notechis scutatus scutatus venom on autonomic neuromuscular transmission. Bri. J. Pharmacol. 1978; 62(3)749–758
  • Harris J. B., Karlsson E., Thesleff S. Effects of isolated toxin from Australian tiger snake (Notechis scutatus scutatus) venom at the mammalian neuromuscular junction. Brit. J. Pharmacol. 1973; 47: 141–146
  • Harris J. B., Prasarnpun S., Dixon R. β‐Bungarotoxin and depletion of synaptic vesicles. 14th World Congress on Animal. Plant and Microbial Toxins, Adelaide, September, 2003, Abstract: 02001., 1CH1 p. 19
  • Harvey A. L., Anderson A. J. Pharmacological actions of facilitatory neurotoxins on peripheral synapses. Neurotoxins and Their Pharmacological Implications, P. G. Jenner. Rowan, New York 1987; 97–114
  • Harvey A. L., Anderson A. J. Dendrotoxins: snake toxins that block potassium channels and facilitate neurotransmitter release. Snake Toxins, A. L. Harvey. Pergamon Press Inc. 1991; 131–164
  • Harvey A. L., Karlsson E. Dendrotoxin from the venom of the green mamba Dendrospis anguisteps. A neurotoxin that enhances acetylcholine release at neuromuscular junctions. Naunyn‐Schmiedeberg's Arch. Pharmacol. 1980; 312: 1–6, [CROSSREF]
  • Harvey A. L., Karlsson E. Protease inhibitor homologous from mamba venoms. Facilitation of acetylcholine release and interactions with prejunctional blocking toxins. Br. J. Pharmacol. 1982; 77: 153–161, [PUBMED], [INFOTRIEVE], [CSA]
  • Ho C. L., Lee C. Y. Mode of neuromuscular blocking action of ceruleotoxin. Toxicon 1983; 21: 527–534, [CSA], [CROSSREF]
  • Ho C. L., Teng C. M., Lee C. Y. Presynaptic and musculotropic effects of a basic phospholipase A2 from the formosan habu Trimeresurus macrosquamatus venom. Toxicon 1984; 22(5)813–816, [PUBMED], [INFOTRIEVE], [CROSSREF]
  • Homsi‐Brandiburgo M.‐I., Queiruz L.‐S., Santo‐neto H., Rodrigues Simioni L., Giglio J. R. Fractionation of Bothrops jarareussu snake venom partial chemical characterisation and biological activity of bothropstoxin. Toxicon 1988; 26: 615–627, [CSA], [CROSSREF]
  • Israel M., Morel N. Mediatophore and a nerve terminal membrane protein supporting the final step of acetylcholine release process. Prog. Brain Res. 1990; 84: 101–110, [PUBMED], [INFOTRIEVE]
  • Jerusalinsky D., Harvey A. Toxins from mamba venoms: small proteins with selectivities for different subtypes of muscarinic acetylcholine receptors. Trends Phar. Sci. 1994; 15: 424–430, [CROSSREF]
  • Jerusalinsky D., Cersenonsky C., Pena C., Raskovsky S., Dajas F. Two polypeptides from Dendrospis angusticeps venom inhibit selectively the binding of central muscarinic cholinergic receptor ligands. Neurochem. Int. 1992; 20: 237–246, [PUBMED], [INFOTRIEVE], [CSA], [CROSSREF]
  • Jerusalinsky D., Harvey A. L., Karlsson E., Potter L. The use of muscarinic toxins in the study of muscarinic receptors. Life Sci. 1997; 60: 1160–1169
  • Jerusalnsky D., Kornisiuk E., Alfaro P., Quillfeldt J., Alonso M., Verde E. R., Cervenansky C., Harvey A. Muscarinic toxin selective for m 4 receptors impairs memory in the rat. NeuroReport 1998; 9(7)1407–1411, [CSA]
  • Jerusalnsky D., Kornisiuk E., Alfaro P., Quillfeldt J., Ferreira A., Verde E. R., Cervenansky C. Muscarinic toxins: novel pharmacological tools for the muscarinic cholinergic system. Toxicon 2000; 38: 747–761, [CSA], [CROSSREF]
  • Jolkkonen M., Toomela T., Rinken A., Satyapan N., Jarr J., Aden A., Karlsson E. Muscarinic toxins from mumba venoms. Toxicon 1993; 31: 497–540, [CROSSREF]
  • Jolkkonen M., Adem A., Hellman U., Wernstedt C., Karlsson E. A snake toxin against muscarinic acetylcholine receptors, amino acid sequence, subtype specificity and effect on guinea pig ileum. Toxicon 1995; 33(4)399–410, [PUBMED], [INFOTRIEVE], [CSA], [CROSSREF]
  • Jolkkonen M., Van Giersbergen P. L., Hellman U., Wernstedt C., Oras A., Satyapanem A., Karlsson E. Muscarinic toxins from the black mamba Dendroaspis polylepis. Eur. J. Biochem. Dec. 1, 1995; 234(2)579–585, [CROSSREF]
  • Joubert E. J., Vijjard N. Snake venoms. The amino acid sequence of two proteinase inhibitor homologous from Dendrospis angusticeps. Hoppe‐Seyler Z. Physiol. Chem. 1980; 361: 661–674
  • Kadlec O., Seferna I., Sevcik I., Somogyi G. T., Vizi E. S. The topographical basis of cholinergic transmission in guniea pig ileum myenteric plexus. Neuroscience 1990; 36: 793–802, [PUBMED], [INFOTRIEVE], [CROSSREF]
  • Kannan H., Tanaka H., Ueta Y., Hayashida Y., Kunitake T., Yamashita H. Effects of centrally administered endothelin‐3 on renal sympathetic nerve activity and renal blood flow in conscious rats. J. Auton. Nerv. Syst. 1994; 49: 105–113, [PUBMED], [INFOTRIEVE], [CSA], [CROSSREF]
  • Karlsson E., Eaker D., Ryden L. Purification of a presynaptic neurotoxin from the venom of the australian tiger snake Notechis scutatus scutatus. Toxicon 1972; 10: 405–413, [PUBMED], [INFOTRIEVE], [CROSSREF]
  • Karlsson E., Eaker D., Fryklund L., Kadin S. Chromatographic separation of Enhydrina schistosa (common sea snake) venom and characterisation of two principal neurotoxins. Biochemistry 1972; 11: 4628, [PUBMED], [INFOTRIEVE]
  • Karlsson E., Mbugua P. M., Rodriguez‐Iithurrralde D. Fasciculins anticholine‐sterase toxins from the venom of the green mamba Dendrospis aunguisticeps. J. Physiol. (Paris) 1984; 79: 232–240
  • Kim H. S., Tamiya N. Isolation, properties and amino acid sequences of a long chain neurotoxin Acanthophis antarcticus b, from the venom of Australian snake the common death adder Acanthophis antarctucus. Biochem. J. 1981a; 193(3)899–906, [PUBMED], [INFOTRIEVE]
  • Kim H. S., Tamiya N. Amino acid sequences and position of the free thiol group of a short chain neurotoxin from common death adder (Acanthphis antarcticus). Biochem. J. 1981b; 199(1)211–218, [PUBMED], [INFOTRIEVE], [CSA]
  • Kornisiuk E., Jerusulinsky D., Cerrenansky C., Harvey A. L. Binding of muscarinic toxins MTx1 and MTx2 from the venom of green mamba Dendrospis angusticeps to cloned human muscarinic cholinoceptors. Toxicon 1995a; 33: 11–18, [PUBMED], [INFOTRIEVE], [CSA], [CROSSREF]
  • Kornisiuk E., Cervenansky C., Alfaro P., Bassarsky M., Harvey A., Jerusalinsky D. Peptide muscarinic toxins with agonist activity that preferentially bind to the m1 receptor subtypes. Commun. Biol. 1995b; 13: 123–133
  • Kriaj I., Ritonja A., Turk D., Gubenek F. Primary structure of ammodytoxin C further reveals the toxin site of ammodytoxin. Biochem. Biophys. Acta. 1989; 999: 198–202
  • Krizaj I., Cople A., Vardjan N., Sirbaj J., Paris A., Sherman N. E., Fox J. W., Gubensek F. Neuronal receptors for ammodytoxin‐C, a presynaptically acting phospholipase A2. 13th World Congress of IST, Paris, Sept., 18–222000, Abstract: L‐86
  • Kuhn P., Deacon A. M., Comoso S., Rajaseger G., Kini R. M., Uson I., Kolatkar P. R. The atomic resolution structure of bucandin, a novel toxin isolated from the Malayan krait, determined by direct methods. Eur. J. Biochem. 1993; 211: 57–62
  • Kukhtina V. V., Weise C., Muranova T. A., Starkov V. G., Franke P., Hucho F., Wnendt S., Gillen C., Tsetilin V. I., Utkin Y. N. Muscarinic toxin‐like proteins from cobra venoms. Eur. J. Biochem. 2000; 267(23)6783–6789
  • Lee C. Y. Chemistry and pharmacology of polypeptide from snake venoms. Ann. Rev. Pharmacol. 1972; 12: 265–286, [PUBMED], [INFOTRIEVE], [CROSSREF]
  • Lee C. Y., Chang C. C. Modes of actions of purified toxins from elapid venoms on neuromuscular transmission. Mem. Inst. Butantan. 1966; 33: 555–572, [PUBMED], [INFOTRIEVE]
  • Lee C. Y., Chen Y. M. Species differences on reversibility of neuromuscular blockade by elapid and sea snake neurotoxins. Animal, Plant and Microbial Toxins, A. Ohsaka, K. Hayashi, Y. Sawai. Plennum Press, New York 1976; Vol. II.: 193–203
  • Lee C. Y., Ho C. L. Pre and postsynaptic action of crotoxin on chick neuromuscular preparation. Natural Toxins, D. Eaker, T. Vadstrom. Pergamon Press, Oxford 1980
  • Lee C. Y., Tseng L. F. Species differences in susceptibility in elapid venoms BuTx like taipoxin and crotoxin, inhibit neuromucular transmission. Toxicon 1969; 7: 89–93, [PUBMED], [INFOTRIEVE], [CROSSREF]
  • Lee C. Y., Chang C. C., Chan Y. M. Reversibility of neuromuscular blockade by neurotoxin from elapid and sea snake venoms. J. Formosan Med. Assoc. 1972; 71: 344–349
  • Lee C. Y., Chen Y. M., Karlsson E. Postsynaptic and musculotropic effects of notexin a presynaptic neuorotoxin from the venom of Notechis scutatus scutatus (Australian tiger snake). Toxicon 1976; 14: 493–494, [PUBMED], [INFOTRIEVE], [CROSSREF]
  • Lee C. Y., Ho C. L., Botes D. P. Sites of action of caudoxin a neurotoxic phospholipase A2 from the from the horned puff adder Bitis caudalis venom. Toxicon 1982; 20: 637–647, [PUBMED], [INFOTRIEVE], [CSA], [CROSSREF]
  • Lee C. Y., Tsai M. C., Tsaur M. L., Lin W. W., Karlsson F. H., Joubert E. J. Pharmacological study on angusticeps type toxins from mamba snake venom. J. Pharmacol. Exp.Ther. 1985; 233: 491–498, [PUBMED], [INFOTRIEVE], [CSA]
  • Lefkowitz R. J., Hoffman B. B., Taylor P. Neurotransmission, the Autonomic and Somatic Motor Nervous System9th Ed., Hardman, Limbird, Milinoff, Raddon, Gilman. McGraw Hill. 1996; 118–119, Ch. 6
  • Lentz T. L., Rosenthal J., Mazarkiewiez J. E. Cytochemical localisation of acetylcholine receptors by means of peroxidase labeled α bungarotoxin. Neurosci. Abst., Soc. Neurosci. 1975; 976, [CSA]
  • Lester H. A. Postsynaptic action of cobra toxin at the myoneural junction from venom of Naja naja atra. Nature (Lond.) 1970; 227: 227–228, [CSA]
  • Lester H. A. Vulnerability of desensitized or curare treated acetylcholine receptors to irreversible blockade by cobra toxin. Molecu. Pharmacol. 1972; 8(6)632–649
  • Lin W. W., Smith L. A., Lee C. Y. A study on the cause of death due to Waglerin I, a toxin from Trimerersurus wagleri. Toxicon 1995; 33(1)111–114, [PUBMED], [INFOTRIEVE], [CSA], [CROSSREF]
  • Loring R. H., Chiappinelli V. A., Zigmond R. E., Cohen J. B. Characterisation of sanke venom neurotoxin which blocks nicotinic transmission in the avian ciliary ganglion. Neuroscience 1984; 11: 989–999, [PUBMED], [INFOTRIEVE], [CROSSREF]
  • Loring R. H., Andrews D., Lane W., Zingmond R. E. Amino acid sequence of Toxin F, a snake venom toxin that blocks neuronal nicotinic receptors. Brain Res. 1986; 385: 30–37, [PUBMED], [INFOTRIEVE], [CROSSREF]
  • Maeda N., Tamiya N. The primary structure of the toxin Laticauda semifasciata III, a weak and reversibly acting neurotoxin from the venom of a sea snake Laticauda semifasciata. Biochem. J. 1974; 141: 389–400, [PUBMED], [INFOTRIEVE], [CSA]
  • Maeda N., Tamiya N. Isolation properties and amino acid sequences of three neurotoxins from the venom of a sea snake Aipysurus laevis. Biochem. J. 1976; 153: 79–87, [PUBMED], [INFOTRIEVE]
  • Maeda N., Tamiya N. Three neurotoxins from the venom of sea snake Astrotia stokesii, including long chain neurotoxic proteins with amidated C terminal. Biochem. J. 1978; 175: 507–517, [PUBMED], [INFOTRIEVE]
  • Maeda N., Takagi K., Tamiya N., Chen Y. M., Lee C. Y. The isolation of easily reversible postsynaptic toxin from the venom of a sea snake Laticauda semifasciata. Biochem. J. 1974; 141: 383–387, [PUBMED], [INFOTRIEVE], [CSA]
  • Mancheva I., Kleinschmidt T., Aleksier B., Braznitzer G. The primary structure of phospholipase A2 of vipoxin from the venom of the Bulgarian viper, Vipera ammodytes. Bio. Chem., Hoppe Seyler 1987; 368: 343–352, [CSA]
  • Marchot P., Bourne Y., Prowse C. N., Bougis P. E., Taylor P. Inhibition of mouse acetylcholinesterase by fasciculin: crystal structure of the complex and mutagenesis of fasciculin. Toxicon 1998; 36(11)1613–1622, [PUBMED], [INFOTRIEVE], [CSA], [CROSSREF]
  • Massoulie J., Pessementi, Bon S., Krejci E., Vallette F. M. Molecular and cellular biology of cholinesterases. Prog. Neurobiol. 1993; 41: 31–91, [PUBMED], [INFOTRIEVE], [CSA], [CROSSREF]
  • Max S. I., Liang J. S., Potter L. T. Purification and properties of m1‐toxin, a specific antagonist of m1 muscarinic receptors. Neuroscience 1993; 13(10)4293–4300, [PUBMED], [INFOTRIEVE]
  • McArdle J. J., Lentz T. L., Witzemann V., Schwarz H., Weinstein S. A., Schmidt J. J., Waglerin I. Selectively blocks the epsilon form of the muscle nicotinic acetylcholine receptor. 1999; 289(1)543–550
  • Mebs D., Narita K., Iwanga S., Samjima Y., Lee C. Y. Purification, properties and amino acid sequences of α‐bungarotoxin from the venom of Bungarus multicinctus. Hoppe‐Seyler Z. Physiol. Chem. 1971; 353: 243–262
  • Mehraban F., Haines A., Dolly J. O. A functional membranous acceptor for dendrotoxin in rat brain: solublisation of the binding component. Biochem. Soc. Trans. 1985; 13: 507–508
  • Melodrama B. S. Action of whole and fractionated Indian cobra Naja naja venom on skeletal muscle. Br. J. Pharmacol. 1967; 25: 197–205
  • Miledi R., Molenuar P. C., Polok R. L. Electrophysiological and chemical determination of acetylcholine release at the frog neuromuscular junction. J. Physiol. 1983; 334: 245–254, [PUBMED], [INFOTRIEVE]
  • Minic J., Krejci E., Faille L., Karlsson E., Molgo J. The modulation of transmitter release from motor nerve terminals by MT3 toxin and drugs that interact with presynaptic muscarinic receptors dependent on functional state of cholinesterase. 13th World Congress of IST, Paris, Sept., 18–222000, Abstract: P. 163
  • Miyoshi S., Tu A. T. Phosphoilipase A2 from Naja naja sputatrix venom is a muscarnic acetylcholine receptor inhibitor. Arch Biochem. Biophys. 1996; 328(1)17–25, [PUBMED], [INFOTRIEVE], [CSA], [CROSSREF]
  • Miyoshi S., Tu A. T. Muscarinic acetylcholine receptor (nAChR) inhibitor from snake venom: interaction with subtypes of human mAChR. Arch. Biochem Biophys. 1999; 369(1)114–118, [PUBMED], [INFOTRIEVE], [CSA], [CROSSREF]
  • Molgo J., Magazanik L. G., Hermel J. M., Juzans P., Stinnake J., Karlsson E. Actions of Fasciculin‐1 and 2 anticholinesterase peptide isolated from green mamba venom at end plate current at the neuromuscular junction. Toxicon, Abstract: 10th Eur Symp. Animal, Plant Microbial Toxins. 1993; Vol. 31 (5): 536
  • Molles B. E., Klines E. F., Rezai P., Nguyen P., Gao S. X., Sine S. M., McArdle J. J., Taylor P. Peptides from the venom of Wagler's pit viper as tools for studying the muscle nicotinic acetylcholine receptor. 13th World Congress of IST, Paris, Sept., 18–222000, Abstract: L110
  • Montecucco C., Rossetto O. How do presynaptic PLA2 neurtotoxin block nerve terminals?. Trends Biochem. Sci. 2000; 25(6)266–270, [PUBMED], [INFOTRIEVE], [CSA], [CROSSREF]
  • Mori N. Isolation and characterisation of Pelamis platurus, yellow bellied sea snake postsynaptic isoneurotoxin. J. Pharm. Pharmacol. 1989; 41: 331–334, [PUBMED], [INFOTRIEVE]
  • Nicholson G. M., Spence I., Lloyd D. R., Wilson H., Tyler M., Howden M. E.H. Presynaptic actions of textilotoxin, a neurotoxin from the Australian common brown snake Pseudonaja textilis. Abstract: Toxicon 10th World Congress of Plant, Animal and Microbial Toxins, Singapore, Nov., 3–81991, 540
  • Nirthanan S., Gopalkrishnakone P., Gwee M. C.E., Knew H. E., Cheah L. S., Kini R. M. Candotoxin a three ‐finger toxin isolated from bungarus candidus snake venom, is a reversible postsynaptic neuromuscular blocker of nicotinic acetylcholine receptor. 13th World Congress of IST, Paris, Sept., 18–222000, Abstract: L 108
  • Nirthanan S., Gao R., Gopalkrishnakone P., Gwee M. C., Khoo H. E., Cheah L. S., Manjunatha, Kini R. Pharmacological characterisation of mikatoxin, an alpha neurotoxin isolated from the venom of new guinean small‐eyed snake Micropechis ikaheka. Toxicon 2002a; 40(7)863–871, [PUBMED], [INFOTRIEVE], [CSA], [CROSSREF]
  • Nirthanan S., Charpantier E., Gopalkrishnakone P., Gwee M. C., Khoo H. E., Cheah L. S., Bertrand D., Kini R. M. Candoxin, a novel toxin from Bungarus candidus, is a reversible antagonist of muscle (αβνδ) but a poorly reversible antagonist of neuronal α‐7 acetylcholine receptors. J. Biol.Chem. 2002b; 277: 17811–17820, [PUBMED], [INFOTRIEVE], [CSA], [CROSSREF]
  • Noetzel C., Chandra V., Perbundt M., Rajashanker K., Singh T., Aleksier B., Kulkura N., Genov N., Betzel C. Enzymatic activity and inhibition of the neurotoxic complex vipoxin from the venom Vipera ammodytes meridionalis. Z. Naturforsch. 2002; 57C: 1078–1083
  • Opi A., Vuernilo N., Gubenek F., Kriaj I. Purification of a novel receptor for secretary PLA2 in porcine cerebral cortex. J. Biol. Chem. 1999; 274: 26315–26320, [CROSSREF]
  • Osaka H., Malany S., Molles B. E., Sine S. M., Taylor O. Pairwise electrostatic interactions between α neurotoxins γ and ε and subunits of the nicotinic acetylcholine receptor. J. Biol. Chem. 2000; 275(8)5478–5484, [PUBMED], [INFOTRIEVE], [CROSSREF]
  • Oshima‐Franco Y., Leite G. B., Silva G. H., Car doso D. F., Hyslop S., Giglio J. R., Cruz Hofling M. A., Rodrigues Simioni L. (2000) Neutralisation of the pharmacological effects of bothropstoxin‐1 from Bothrops jararacussu (jararacucu) venom by crotoxin antiserum and heparin. 13th World Congress of IST, Toxins from Animal, Plant and Microbes. Sept., 18–222000, Abstract: P.240
  • Oshima‐Franco Y., Leite G. B., Silva G. H., Car doso D. F., Hyslop S., Giglio J. R., Cruz Hofling M. A., Rodrigues Simioni L. Neutralisation of the pharmacological effects of bothropstoxin‐1 from Bothrops Jararacussu venom by crotoxin antiserum and heparin. Toxicon 2001; 39(10)1477–1485, [CSA], [CROSSREF]
  • Pallabi D. E., Dasgupta S. C., Gowes A. A novel peptide toxin (Hannah peptide) from the venom of the Indian king Cobra Ophiophagus hannah. Nat. Congre. of Indian Pharmac. Society, Jammu, Nov., 14–161998, 199, Abstract: XXX
  • Panchiappan A., Nirthanan S., Srinivasan K. N., Raghavaendra, Prasad H. S., Gopalkrinshnakone P. Toxicofunctional genomics of human brain cells exposed to candoxin, a novel neurotoxin of bungarus candidus venom and its implication in nAChR mediated neurotransmission. 14th World Congress of Animal, Plant, Microbial Toxins, AdelaideAustralia, Sept., 2003, Abstract: ICH 3 P.22
  • Patel R., Excell B. The effects of lethal component of Dendrospis jamensoni snakevenom on neuromuscular junction and muscle membrane permeability. Toxicon 1975; 13: 245–304, [CROSSREF]
  • Penner R., Peterson M., Pieran F. K., Dreyer F., Dendrotoxin A. Selective blocker of a noninactivating potassium current in guinea pig dorsal root ganglion neurons. Pflugers Arch. 1986; 407: 365–369, [PUBMED], [INFOTRIEVE], [CROSSREF]
  • Possani L. D., Braun M., Atriko M., Mochca‐Movales J., Zamndio F. Z., Gurrola G. B., Brown A. M. Isolation and physiological characterisation of taicatoxin, a complex toxin with specific effects on calcium channels. Toxicon 1992; 30(11)1343–1364, [PUBMED], [INFOTRIEVE], [CROSSREF]
  • Prado‐Franceochi J., Vital Brazil O. Convulxin a new toxin from the venom of the South American rattle snake Crotalus durissus terrifus. Toxicon 1981; 19: 875–887, [CROSSREF]
  • Prado‐Franceochi J., Hyslop S., Cogo J. C., Andrade A. L., Assakura M. T., Reichl A. P., Cruz‐Hofling M. A., Rodrigues Simioni L. Characterisation of a myotoxin from the duvernoy's gland secretion of the xenodontine colubrid Philodryas olefersii (green snake): effects on striated muscle and the neuromuscular junction. Toxicon 1998; 36(10)1407–1421, [CSA], [CROSSREF]
  • Pu X. C., Wong P. T., Gopalkrinshnakone P. A novel analgesic toxin from the venom of king cobra Ophiophagus hannah. Toxicon 1995; 33(11)1425–1431, [PUBMED], [INFOTRIEVE], [CSA], [CROSSREF]
  • Pu X. C., Gopalkrshnakone P., Wong P. T. H. Effects of hannahalgesin from king cobra (Ophiophagus hannah) venom on structure and functin of rat annacoccygeus muscle. Toxicon 1997; 35(4)501, [CROSSREF]
  • Purkerson S. L., Potter L. T. Use of antimuscarinic toxins to facilitate studies of striatal m 4 muscarinic receptors. J. Pharmacol. Exp. Ther. 1998; 284(2)707–713, [PUBMED], [INFOTRIEVE], [CSA]
  • Radic Z., Taylor P. Anticholinesterase inhibition and restricted substrate entry by the snake toxin, fasciculin. 13th World Congress of IST, Toxins from Animal, Plant and Microbes, Paris, Sept., 18–222000, Abstract: L 112
  • Rajashankar K. R., Tsai I. H., Betzel C. Crystallisation and preliminary X‐ray diffraction studies of the myonecrotic, heterodimeric phospholipase A2 from the Taiwan viper (Vipera russelli formosensis). Acta Crystallogr. D. Biol. Crystallogr. 1999; 55(5)1064–1065, [PUBMED], [INFOTRIEVE], [CSA]
  • Ravdin P. M., Nitkin R. M., Berg D. K. Internalisation of α‐bungarotoxin on neurons induced by a neurotoxin that blocks neuronal acetylcholine sensitivity. J. Neurosci., 1: 849–861
  • Ritonja A., Machleidt W., Turk V., Gubensek F. Amino acid sequence of ammodytoxin B partially reveals the location of the site of toxicity of ammodytoxins. Biol. Chem. Hoppe. Seyler 1986; 367(9)919–923, [PUBMED], [INFOTRIEVE]
  • Rodrigues Simioni L., Hawgood B. J., Smith Cristophor H. Properties of the early phases of crotoxin poisoning at frog neuromuscular junction. Toxicon 1990; 28(12)1479–1489, [PUBMED], [INFOTRIEVE], [CSA], [CROSSREF]
  • Rodrigues Simiioni L., Prado‐Franceschi J., Cintra A. C. O., Giglio J. R., Jiang M. S., Fletcher J. E. Two role for enzymatic activity or dandrolene sensitive Ca2 + stores in the muscular effects of bothropstoxin, a Lys‐49 phospholipase A2, myotoxin. Toxicon 1995; 33(11)1479–1489, [CSA], [CROSSREF]
  • Rodriguez‐Ithurralde D., Silveria R., Barbeito L., Dajas F. A powerful anticholinesterase polypeptide from Dendrospis angusticeps venom. Neurochem Int. 1983; 5: 267–274, [CSA], [CROSSREF]
  • Rowan E. G., Harvey A. L. Potassium channel blocking actions of β‐bungarotoxin and related toxins on mouse and frog motor nerve terminals. Br. J. Pharmacol. 1988; 94: 839–847, [PUBMED], [INFOTRIEVE], [CSA]
  • Rowan E. G., Harvey A. L., Takasaki C., Tamiya N. Neuromuscular effects of a toxic phospholipase A2 and its nontoxic homologue from the venom of the sea snake, Laticauda colubrina. Toxicon 1989; 27(5)587–591, [PUBMED], [INFOTRIEVE], [CSA], [CROSSREF]
  • Rowan E. G., Harvey A. L., Menez A. Neuromuscular effects of nigexine, a basic phosphoilipase A2 from Naja nigricollis venom. Toxicon 1991; 29(3)371–374, [PUBMED], [INFOTRIEVE], [CSA], [CROSSREF]
  • Rzhevskii D. I., Murashev A. N., Kukhtina W., Tsetlin V. I., Utkin I. u.N. The weak neurotoxin from Naja kaothia cobra venom decreases arterial blood pressure in rats. Bioorg. Khim 2001; 27(3)221–223, [PUBMED], [INFOTRIEVE], [CSA]
  • Sah D. W. Y., Loring R. H., Zigmond R. E. Long term blockade by Toxin F of nicotinic synaptic potentials in cultured sympathetic neurons. Neuroscience 1987; 20: 867–874, [PUBMED], [INFOTRIEVE], [CROSSREF]
  • Schiavo G., Benfenti F., Poulam B., Rosette O., Polverino, de Lauveto P., Dasgupta B. R., Montecucco C. Tetanus and botulinum neurotoxin block neurotransmitter release by proteolytic cleavage of synaptobrevin. Nature 1992; 339: 832–834, [CROSSREF]
  • Schweitz H., Bidard J. N., Lazdunski M. Purification and pharmacological characterisation of peptide toxins from the black mamba Dendrospis polylepis venom. Toxicon 1990; 28: 847–856, [PUBMED], [INFOTRIEVE], [CSA]
  • Schweitz H., Heurteaux C., Bois P., Moinier D., Romey G., Lazdunski M. Calcicludine, a venom peptide of the Kunitz type protease inhibitor family is a potent blocker of high threshold Ca2 + channel with a high affnity for L‐type channels in cerebellar granule neurons. Proc. Nat. Acad. Sci., U. S. A. 1993; 91: 878–882, [CSA]
  • Servent D., Antil‐Delbeke S., Gailard C., Tamiya T., Corringer P. J., Changeux J. P., Menez A. Molecular diterminants involved in the specificity of interactions of various neurotoxins on different nicotinic receptor subtypes. 13th World Congress of IST, Toxins From Animal, Plant and Microbes, Paris, Sept., 18–222000, Abstract L 109
  • Sheumack D. D., Spence I., Tyler M. I., Howden M. E. The complete amino acid sequence of a post synaptic neurotoxin isolated from the venom of Australian death adder snake Acanthophis antarcticus. Comp. Biochem. Physiol., B 1990; 95(1)45–50, [PUBMED], [INFOTRIEVE], [CSA], [CROSSREF]
  • Siew J. P. Y., Tan P. T. J., Koh J. L. Y., Jeyaseelan K., Brusic V. Systematization of snake venom neurotoxins based on structure function. 14th World Congress on Animal, Plant and Microbial Toxins, AdelaideAustralia, Sept., 14–192003, Abstract: 5PF No. 16401
  • Silveria R., Siciliano J., Abo V., Vieva L., Daja F. Intrastriatal dendrotoxin injection behavioural and neurochemical effects. Toxicon 1988; 26: 1009–1015, [CSA], [CROSSREF]
  • Snyder G. K., Ramsey H. W., Taylor W. J., Chiau C. Y. Neuromuscular blockade of chick biventer cervices muscle preparations by a fraction from coral snake venom. Toxicon 1973; 11: 505–508, [PUBMED], [INFOTRIEVE], [CROSSREF]
  • Soares A. M., Guerra‐sa R., Borja‐Oliveira C. R., Rodrigues V. M., Rodrigues Simioni L., Rodrigues V., Fontes M. R. M., Rodrigues B., Gutierreze J. M., Giglio J. R. Structural and function characterisation of Bn‐SP‐7 and, a Lys 49, myotoxic phospholipase A2 Homologus from Bothorops neuwiedi venom. Arch. Biochem. Biophysics. 2000; 378(2)210–219, [CSA], [CROSSREF]
  • Stansfeld C. E., Marsh S. J., Halliwell J. V., Brown D. A. 4 aminopyridine and dendrotoxin induce repetitive firing in rat visceral sensory neurons by blocking a slowly inactivating outward current. Neurosci. Lett. 1986; 64: 299–304, [PUBMED], [INFOTRIEVE], [CROSSREF]
  • Strydom A. J.C., Botes D. P. Purification, properties and complete amino acid sequences of two toxins from ringhals Hemachatus hemachatus venom. J. Biol. Chem. 1971; 246: 1341–1349, [PUBMED], [INFOTRIEVE]
  • Tamiya N., Arai H. Studies on sea snake venoms. Crystallisation of erabutoxins a and b from Laticauda semifasciata venom. Biochem. J. 1966; 99: 624–630, [PUBMED], [INFOTRIEVE]
  • Tan N. H. Isolation and characterisation of two toxins from the venom of Malyan cobra Naja naja sputatrix. Toxicon 1983; 21(2)201–207, [PUBMED], [INFOTRIEVE], [CSA], [CROSSREF]
  • Taylor P. Agents Acting at the Neuromuscular Junction and Autonomic Ganglia10th Ed., Hardman, Limbird, Gilman. McGraw Hill. 2001a; 194, Ch. 9
  • Taylor P. ‘Anticholinesterase Agents’, Goodman and Gilman's the Pharmacological Basis of Therapeutics10th, Hardman, Limbird, Gilman. McGraw Hill. 2001b; 176–177, Ch. 8
  • Taylor P., Radic Z. The cholinesterase from genes to proteins. Annu. Rev. Pharmacol. Toxicol. 1994; 34: 281–320, [PUBMED], [INFOTRIEVE], [CSA], [CROSSREF]
  • Tchorbavov B., Aleksiev B. A simple procedure from the isolation of vipoxin a neurotoxin with phospholipase A2 activity from the venom of the Bulgarian viper (Vipera ammodytes). J. Appl. BioChem. 1978; 3: 558–561
  • Teerapong P., Harvey A. L., Barkus T. C. Neuromuscular effects of fractions isolated from the venom of Thialand cobra Naja naja siamensis. Toxicon 1978; 16: 551–560, [PUBMED], [INFOTRIEVE], [CROSSREF]
  • Thouin J., Ritonja A., Gubenek F., Russell F. E. Neuromuscular and lethal effects of phospholipase A from Vipera ammodytes venom. Toxicon 1982; 20: 1051–1058, [PUBMED], [INFOTRIEVE], [CSA], [CROSSREF]
  • Thwin M. M, Gopalkrinshnakone P. Inhibition of synaptosomal binding of 125I daboiatoxin by python serum. Toxicon 1997; 34(3)314–334, Y. R.H. T C. [CROSSREF]
  • Thwin M.‐M., Gopalakrishnakone P., Yuen R., Tan C. H. A major lethal factor of the venom of Burmese Russell's viper (Daboia russelli siamensis): isolation, N‐terminal sequencing and biological activities of daboiatoxin. Toxicon 1995a; 33(1)63–76, [CSA], [CROSSREF]
  • Thwin M.‐M., Gopalkrishnakone P., Yuen R., Tan C. H. A major lethal factor of the venom of Burmese Russell's viper (Daboia russelli siamensis): its specific binding to rat brain synaptosomes. Toxicon 1995b; 33(6)716–717, [CROSSREF]
  • Thwin M. M., Gopalkrishnakone P., Yuen R., Tan C. H. Synaptosomal binding of 125I‐labelled daboiatoxin, a new PLA2 neurotoxin from the venom of Daboia russelli siamensis. February, 1996; 34(2)183–199, [CSA]
  • Thwin M.‐M., Gopalkrishnakone P., Yuen R., Tan C. H. Daboiatoxin acceptor in monkey (Maccacus fasiculata) and peafowl (Pavo cristatus) brain synaptosomal membranes. Toxicon 1997; 35(4)492, [CROSSREF]
  • Tsai M. C., Lee C. Y., Bdolah A. Mode of neuromuscular blocking action of a toxic phospholipase A2 from Pseudocerastes fieldi (Field's horned viper) snake venom. Toxicon 1983; 21(A)527–534, [PUBMED], [INFOTRIEVE], [CSA], [CROSSREF]
  • Tsai M. C., Tseil W. H., Smith L. A., Lee C. Y. Effects of Waglerin I on neuromuscular transmission on mouse N–M preparation. Toxicon 1995; 33(2)363–371, [PUBMED], [INFOTRIEVE], [CSA], [CROSSREF]
  • Tseng W. P., Lin‐Shiau S. Y. Activation of NMDA receptor partly involved in Beta Bungarotoxin induced neurotoxicity in cultured primary neurons. Neurochem. Int. 2003; 42(4)333–344, [PUBMED], [INFOTRIEVE], [CSA], [CROSSREF]
  • Tseng L. F., Chiu T. H., Lee C. Y. Absorption and distribution of 131 I‐labeled Cobra venom and its purified Toxins. Toxicol. Appl. Pharmacol. 1968; 12: 526–535, [CROSSREF]
  • Tu T., Tu A. T., Lin T. S. Some pharmacological properties of the venom, venom fraction and pure toxin of the yellow‐bellied sea snake Pelamis platurus. J. Pharm. Pharmacol. 1976; 28: 139–145, [PUBMED], [INFOTRIEVE]
  • Tyler M. I., Spence I., Barnett D., Howden M. E. Pseudonajatoxin b unusual amino acid sequence of a lethal neurotoxin from the venom of the Australian common brown snake Pseudonaja textilis. Eur. J. Biochem. 1987; 166(1)139–143, [PUBMED], [INFOTRIEVE]
  • Tyler M. I., Retsen Y. W., Gipson M. K., Barnett D., Howe E., Stocklin, Tornbull R. K., Kuchel J., Mirtschan P. Isolation and amino acid sequences of a new long chain NTXs with two chromatographic isoforms (Aae1 and Aae2) from the Australian death adder (Acanthphis antarcticus). Toxicon 1997; 35(4)555–562, [PUBMED], [INFOTRIEVE], [CSA], [CROSSREF]
  • Utkin Y. N., Kukhtina V. V., Maslennikov I. V., Elotsky A. V., Starkov V. G., Weise C., Franke J. P., Hucho F., Tsetlin V. I. First tryptophan containing weak neurotoxin from cobra venom. Toxicon 2001a; 39(7)921–927, [PUBMED], [INFOTRIEVE], [CSA], [CROSSREF]
  • Utkin Y. N., Kukhtina V., Kryukova E. V., Chiodini F., Bertrand D., Meth fessel C., Tsetlin V. I. Weak toxin from Naja kaothia is a nontoxic antagonist of α7 and muscle type nicotinic acetylcholine receptors. J. Biol.Chem. 2001b; 276(19)15810–15815, [PUBMED], [INFOTRIEVE], [CSA], [CROSSREF]
  • Vandermeers A., Vandermeers M., Rathe J., Waelbroeck M., Jolkkonen M., Oras A., Karlsson E. Purification and sequence determination of a new muscarinic toxin, (MT4) from the venom of the green mamba (Dendroaspis angusticeps). Toxicon 1995; 33(9)1171–1179, [PUBMED], [INFOTRIEVE], [CSA], [CROSSREF]
  • Van der Wyden L., Hains P., Morris M., Brondy K. Acanthotoxin, a toxic phospholipase A2 from the venom of the common death adder Acanthopis antarctis. Toxicon 1997; 35: 1315–1325, [CSA], [CROSSREF]
  • Vital Brazil O., Excell B. J., Sanatana de SaS. The importance of phospholipase A2 in the action of crotoxin complex at the frog neuromuscular junction. J. Physiol. (Lond.) 1973; 234: 63–64, [CSA]
  • Vucemilo N., Gubensek F., Krizaj J. Neuronal acceptor for ammodytoxin in pig brain cortex. Toxicon 1998; 36(9)1272
  • Vuemilo N., Gubenek F., Kriaj I. Identification of a new high affinity binding protein for nontoxic PLA. Biochem. Biophys. Res. Commun. 1998; 251: 209–212, [CSA], [CROSSREF]
  • Wang Y. N., Lu P. J., Ho C. L., Tsai I. H. Characterisation and molecular cloning of nontoxic phospholipase A2 from Taiwanese Viper (Vipera russelli formosensis). Eur. J. Biochem., 209: 635–641
  • Wilson H. I., Nicolson G. M. Presynaptic snake beta‐neurotoxins produce tetanic fade and end plate potential run down during neuromuscular blockade in mouse diaphragm. Naunyn‐Schmiedeberg's Arch. Pharmacol. 1997; 356(5)626–634
  • Wolf K. M., Clarleglio A., Chiappinelli V. A. Kappa bungarotoxin binding of a neuronal nicotinic receptor antagonist to chick optic lobe and skeletal muscle. Brain Res. 1988; 439: 249–258, [PUBMED], [INFOTRIEVE], [CROSSREF]
  • Wu Y., Li Yu Liang. β‐agkistrodotoxin inhibits fast and Ca2 + activated K+ currents recorded from mouse motor nerve terminals. Toxicon 2000; 38(2)177–185, [PUBMED], [INFOTRIEVE], [CSA], [CROSSREF]
  • Wu Y., Wang Z. F., Shi Y. L. Beta agkistrodotoxin inhibits large conductance calcium activated potassium channels in rat hippocampal CA1 pyramidal neurons. Brain Res. 2002; 14:940(1–2)21–26, [CROSSREF]
  • Yang C. C. Crystallisation and properties of cobrotoxin from Formosan cobra venom. J. Biol. Chem. 1965; 240: 1616–1618, [PUBMED], [INFOTRIEVE]
  • Yang C. C., Chan H. L. Immunological studies on β1‐bungarotoxin using polyclonal and monoclonal antibodies. Toxicon 1999; 37: 729–742, [PUBMED], [INFOTRIEVE], [CSA]
  • Yang C. C., Chang C. C., Hayashi K., Suzuki T. Amino acid composition and end group analysis of cobrotoxin. Toxicon 1969; 7: 43, [PUBMED], [INFOTRIEVE]
  • Yoshizawa T., Shinmi O., Giaid A., Yanagisawa M. M., Gibson S. J., Kimura S., Uchiyama Y., Polak J. M., Masaki T., Kanazawa I. Endothelin: a novel peptide, in the posterior pituitary system. Science 1990; 247: 462–464, [PUBMED], [INFOTRIEVE]
  • Yu N. T., Lin T. S., Tu A. T. Laser Raman Scattering of neurotoxins isolated from the venom of sea snakes Lapemis hardwickii and Enhydrina schistosa. J. Biol. Chem. 1975; 250: 1782, [PUBMED], [INFOTRIEVE]
  • Zaimis E. The neuromuscular junction: area of uncertainty. Neuromuscular Junction, E. Zaimis. Spranger‐Verlag, Berlin 1976; 1–18

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.