1
|
Phulera S, Dickson CJ, Schwalen CJ, Khoshouei M, Cassell SJ, Sun Y, Condos T, Whicher J, Weihofen WA. Scorpion α-toxin LqhαIT specifically interacts with a glycan at the pore domain of voltage-gated sodium channels. Structure 2024; 32:1611-1620.e4. [PMID: 39181123 DOI: 10.1016/j.str.2024.07.021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2024] [Revised: 06/13/2024] [Accepted: 07/30/2024] [Indexed: 08/27/2024]
Abstract
Voltage-gated sodium (Nav) channels sense membrane potential and drive cellular electrical activity. The deathstalker scorpion α-toxin LqhαIT exerts a strong action potential prolonging effect on Nav channels. To elucidate the mechanism of action of LqhαIT, we determined a 3.9 Å cryoelectron microscopy (cryo-EM) structure of LqhαIT in complex with the Nav channel from Periplaneta americana (NavPas). We found that LqhαIT binds to voltage sensor domain 4 and traps it in an "S4 down" conformation. The functionally essential C-terminal epitope of LqhαIT forms an extensive interface with the glycan scaffold linked to Asn330 of NavPas that augments a small protein-protein interface between NavPas and LqhαIT. A combination of molecular dynamics simulations, structural comparisons, and prior mutagenesis experiments demonstrates the functional importance of this toxin-glycan interaction. These findings establish a structural basis for the specificity achieved by scorpion α-toxins and reveal the conserved glycan as an essential component of the toxin-binding epitope.
Collapse
Affiliation(s)
- Swastik Phulera
- Discovery Sciences, Novartis Biomedical Research, 250 Massachusetts Avenue, Cambridge, MA 02139, USA
| | - Callum J Dickson
- Global Discovery Chemistry, Novartis Biomedical Research, 181 Massachusetts Avenue, Cambridge, MA 02139, USA
| | - Christopher J Schwalen
- Global Discovery Chemistry, Novartis Biomedical Research, 181 Massachusetts Avenue, Cambridge, MA 02139, USA
| | - Maryam Khoshouei
- Discovery Sciences, Novartis Biomedical Research, Novartis Pharma AG, Basel, Switzerland
| | - Samantha J Cassell
- Discovery Sciences, Novartis Biomedical Research, 250 Massachusetts Avenue, Cambridge, MA 02139, USA
| | - Yishan Sun
- Neuroscience, Novartis Biomedical Research, 22 Windsor St, Cambridge, MA 02139, USA
| | - Tara Condos
- Discovery Sciences, Novartis Biomedical Research, 250 Massachusetts Avenue, Cambridge, MA 02139, USA
| | - Jonathan Whicher
- Discovery Sciences, Novartis Biomedical Research, 250 Massachusetts Avenue, Cambridge, MA 02139, USA.
| | - Wilhelm A Weihofen
- Discovery Sciences, Novartis Biomedical Research, 250 Massachusetts Avenue, Cambridge, MA 02139, USA.
| |
Collapse
|
2
|
Hmaidi R, Ksouri A, Benabderrazek R, Antonietti V, Sonnet P, Gautier M, Bouhaouala-Zahar B, Ouadid-Ahidouch H. The Pharmacological and Structural Basis of the AahII–NaV1.5 Interaction and Modulation by the Anti-AahII Nb10 Nanobody. Front Pharmacol 2022; 13:821181. [PMID: 35295326 PMCID: PMC8918821 DOI: 10.3389/fphar.2022.821181] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2021] [Accepted: 01/17/2022] [Indexed: 11/13/2022] Open
Abstract
Scorpion α-toxins are neurotoxins that target the fast inactivation mechanism of voltage-gated sodium (NaV) channels leading to several neuro- and cardiotoxic effects in mammals. The toxin AahII is the most active α-toxin from the North African scorpion Androctonus australis Hector that slows the fast inactivation of NaV channels. To fight scorpion envenomation, an anti-AahII nanobody named NbAahII10 (Nb10) was developed. The efficiency of this nanobody has been evaluated in vivo on mice, but its mechanism of action at the cellular level remains unknown. Here we have shown that AahII toxin slows the fast inactivation of the adult cardiac NaV1.5 channels, expressed in HEK293 cells, in a dose-dependent manner, while current amplitude was not affected. The inactivation of NaV1.5 is slower by a factor of 4, 7, and 35 in the presence of [AahII] at 75, 150, and 300 nM, respectively. The washout partially reversed the toxin effect on inactivation from 8.3 ± 0.9 ms to 5.2 ± 1.2 ms at 75 nM. We have also demonstrated that the highly neutralizing Nb10 can fully reverse the effect of AahII toxin on the channel inactivation kinetics even at the 1:1 M ratio. However, the 1:0.5 M ratio is not able to neutralize completely the AahII effect. Therefore, the application of Nb10 promotes a partial abolishment of AahII action. Bioinformatic analysis and prediction of NaV1.5-driven docking with AahII show that Ala39 and Arg62 of AahII play a crucial role to establish a stable interaction through H-bound interactions with Gln1615 and Lys1616 (S3–S4 extracellular loop) and Asp1553 (S1–S2 loop) from the voltage-sensing domain IV (VSD4) of NaV1.5, respectively. From this, we notice that AahII shares the same contact surface with Nb10. This strongly suggests that Nb10 dynamically replaces AahII toxin from its binding site on the NaV1.5 channel. At the physiopathological level, Nb10 completely neutralized the enhancement of breast cancer cell invasion induced by AahII. In summary, for the first time, we made an electrophysiological and structural characterization of the neutralization potent of Nb10 against the α-scorpion toxin AahII in a cellular model overexpressing NaV1.5 channels.
Collapse
Affiliation(s)
- Riadh Hmaidi
- Laboratory of Biomolecules, Venoms, and Theranostic Applications, Institut Pasteur Tunis, University of Tunis El Manar, Tunis, Tunisia
- Laboratory of Cellular and Molecular Physiology UR 4667, UFR of Sciences, University of Picardie Jules Verne, Amiens, France
| | - Ayoub Ksouri
- Laboratory of Biomolecules, Venoms, and Theranostic Applications, Institut Pasteur Tunis, University of Tunis El Manar, Tunis, Tunisia
| | - Rahma Benabderrazek
- Laboratory of Biomolecules, Venoms, and Theranostic Applications, Institut Pasteur Tunis, University of Tunis El Manar, Tunis, Tunisia
| | - Viviane Antonietti
- Infectious Agents, Resistance and Chemotherapy UR 4294, UFR of Pharmacy, University of Picardie Jules Verne, Amiens, France
| | - Pascal Sonnet
- Infectious Agents, Resistance and Chemotherapy UR 4294, UFR of Pharmacy, University of Picardie Jules Verne, Amiens, France
| | - Mathieu Gautier
- Laboratory of Cellular and Molecular Physiology UR 4667, UFR of Sciences, University of Picardie Jules Verne, Amiens, France
- *Correspondence: Mathieu Gautier, ; Balkiss Bouhaouala-Zahar, ; Halima Ouadid-Ahidouch,
| | - Balkiss Bouhaouala-Zahar
- Laboratory of Biomolecules, Venoms, and Theranostic Applications, Institut Pasteur Tunis, University of Tunis El Manar, Tunis, Tunisia
- Medical School of Tunis, University of Tunis El Manar, Tunis, Tunisia
- *Correspondence: Mathieu Gautier, ; Balkiss Bouhaouala-Zahar, ; Halima Ouadid-Ahidouch,
| | - Halima Ouadid-Ahidouch
- Laboratory of Cellular and Molecular Physiology UR 4667, UFR of Sciences, University of Picardie Jules Verne, Amiens, France
- *Correspondence: Mathieu Gautier, ; Balkiss Bouhaouala-Zahar, ; Halima Ouadid-Ahidouch,
| |
Collapse
|
3
|
Zhu Q, Du Y, Nomura Y, Gao R, Cang Z, Wei GW, Gordon D, Gurevitz M, Groome J, Dong K. Charge substitutions at the voltage-sensing module of domain III enhance actions of site-3 and site-4 toxins on an insect sodium channel. INSECT BIOCHEMISTRY AND MOLECULAR BIOLOGY 2021; 137:103625. [PMID: 34358664 PMCID: PMC9376739 DOI: 10.1016/j.ibmb.2021.103625] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/03/2021] [Revised: 07/29/2021] [Accepted: 07/29/2021] [Indexed: 06/13/2023]
Abstract
Scorpion α-toxins bind at the pharmacologically-defined site-3 on the sodium channel and inhibit channel inactivation by preventing the outward movement of the voltage sensor in domain IV (IVS4), whereas scorpion β-toxins bind at site-4 on the sodium channel and enhance channel activation by trapping the voltage sensor of domain II (IIS4) in its outward position. However, limited information is available on the role of the voltage-sensing modules (VSM, comprising S1-S4) of domains I and III in toxin actions. We have previously shown that charge reversing substitutions of the innermost positively-charged residues in IIIS4 (R4E, R5E) increase the activity of an insect-selective site-4 scorpion toxin, Lqh-dprIT3-c, on BgNav1-1a, a cockroach sodium channel. Here we show that substitutions R4E and R5E in IIIS4 also increase the activity of two site-3 toxins, LqhαIT from Leiurusquinquestriatus hebraeus and insect-selective Av3 from Anemonia viridis. Furthermore, charge reversal of either of two conserved negatively-charged residues, D1K and E2K, in IIIS2 also increase the action of the site-3 and site-4 toxins. Homology modeling suggests that S2-D1 and S2-E2 interact with S4-R4 and S4-R5 in the VSM of domain III (III-VSM), respectively, in the activated state of the channel. However, charge swapping between S2-D1 and S4-R4 had no compensatory effects on gating or toxin actions, suggesting that charged residue interactions are complex. Collectively, our results highlight the involvement of III-VSM in the actions of both site 3 and site 4 toxins, suggesting that charge reversing substitutions in III-VSM allosterically facilitate IIS4 or IVS4 voltage sensor trapping by these toxins.
Collapse
Affiliation(s)
- Qing Zhu
- Key Lab of Food Quality and Safety of Jiangsu Province-State Key Laboratory Breeding Base, Key Laboratory of Control Technology and Standard for Agro-product Safety and Quality, Ministry of Agriculture, Institute of Food Safety and Nutrition, Jiangsu Academy of Agricultural Sciences, Nanjing, China; Department of Entomology, Michigan State University, East Lansing, MI, USA
| | - Yuzhe Du
- Department of Entomology, Michigan State University, East Lansing, MI, USA
| | - Yoshiko Nomura
- Department of Entomology, Michigan State University, East Lansing, MI, USA
| | - Rong Gao
- Department of Hygienic Analysis and Detection, School of Public Health, Nanjing Medical University, 818 Tianyuan East Road, Nanjing, Jiangsu, China
| | - Zixuan Cang
- Department of Mathematics, Michigan State University, East Lansing, MI, USA
| | - Guo-Wei Wei
- Department of Mathematics, Michigan State University, East Lansing, MI, USA
| | - Dalia Gordon
- Department of Plant Molecular Biology & Ecology, George S. Wise Faculty of Life Sciences, Tel-Aviv University, Tel Aviv, Israel
| | - Michael Gurevitz
- Department of Plant Molecular Biology & Ecology, George S. Wise Faculty of Life Sciences, Tel-Aviv University, Tel Aviv, Israel.
| | - James Groome
- Department of Biological Sciences, Idaho State University, Pocatello, ID, USA
| | - Ke Dong
- Department of Entomology, Michigan State University, East Lansing, MI, USA; Department of Biology, Duke University, Durham, NC, USA.
| |
Collapse
|
4
|
Niklas B, Jankowska M, Gordon D, Béress L, Stankiewicz M, Nowak W. Interactions of Sea Anemone Toxins with Insect Sodium Channel-Insights from Electrophysiology and Molecular Docking Studies. Molecules 2021; 26:molecules26051302. [PMID: 33670972 PMCID: PMC7957711 DOI: 10.3390/molecules26051302] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2021] [Revised: 02/23/2021] [Accepted: 02/24/2021] [Indexed: 01/22/2023] Open
Abstract
Animal venoms are considered as a promising source of new drugs. Sea anemones release polypeptides that affect electrical activity of neurons of their prey. Voltage dependent sodium (Nav) channels are the common targets of Av1, Av2, and Av3 toxins from Anemonia viridis and CgNa from Condylactis gigantea. The toxins bind to the extracellular side of a channel and slow its fast inactivation, but molecular details of the binding modes are not known. Electrophysiological measurements on Periplaneta americana neuronal preparation revealed differences in potency of these toxins to increase nerve activity. Av1 and CgNa exhibit the strongest effects, while Av2 the weakest effect. Extensive molecular docking using a modern SMINA computer method revealed only partial overlap among the sets of toxins’ and channel’s amino acid residues responsible for the selectivity and binding modes. Docking positions support earlier supposition that the higher neuronal activity observed in electrophysiology should be attributed to hampering the fast inactivation gate by interactions of an anemone toxin with the voltage driven S4 helix from domain IV of cockroach Nav channel (NavPaS). Our modelling provides new data linking activity of toxins with their mode of binding in site 3 of NavPaS channel.
Collapse
Affiliation(s)
- Beata Niklas
- Institute of Physics, Faculty of Physics, Astronomy and Informatics, Nicolaus Copernicus University, Grudziadzka 5, 87-100 Torun, Poland
- Correspondence: (B.N.); (W.N.)
| | - Milena Jankowska
- Faculty of Biological and Veterinary Sciences, Nicolaus Copernicus University, Lwowska 1, 87-100 Torun, Poland; (M.J.); (M.S.)
| | - Dalia Gordon
- Department of Biomolecular Sciences, Weizmann Institute of Science, Rehovot 76100, Israel;
| | - László Béress
- Department of Internal Medicine, Clinic of Immunology, Division of Experimental and Clinical Peptide Research, Hannover Medical School, 30625 Hannover, Germany;
| | - Maria Stankiewicz
- Faculty of Biological and Veterinary Sciences, Nicolaus Copernicus University, Lwowska 1, 87-100 Torun, Poland; (M.J.); (M.S.)
| | - Wieslaw Nowak
- Institute of Physics, Faculty of Physics, Astronomy and Informatics, Nicolaus Copernicus University, Grudziadzka 5, 87-100 Torun, Poland
- Correspondence: (B.N.); (W.N.)
| |
Collapse
|
5
|
Activation of voltage-gated sodium channels by BmK NT1 augments NMDA receptor function through Src family kinase signaling pathway in primary cerebellar granule cell cultures. Neuropharmacology 2020; 180:108291. [PMID: 32931812 DOI: 10.1016/j.neuropharm.2020.108291] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2020] [Revised: 08/23/2020] [Accepted: 08/26/2020] [Indexed: 12/29/2022]
Abstract
Voltage-gated sodium channels (VGSCs) are responsible for the generation and propagation of action potentials in excitable cells and are the molecular targets of an array of neurotoxins. BmK NT1, an α-scorpion toxin obtained from the scorpion Buthus martensii Karsch (BmK), produces neurotoxicity that is associated with extracellular Ca2+ influx through Na+-Ca2+ exchangers, N-methyl-d-aspartic acid (NMDA) receptors, and L-type Ca2+ channels in cultured cerebellar granule cells (CGCs). In the present study, we demonstrated that BmK NT1 triggered concentration-dependent release of excitatory neurotransmitters, glutamate and aspartate; both effects were eliminated by VGSC blocker, tetrodotoxin. More importantly, we demonstrated that a threshold concentration of BmK NT1 that produced marginal Ca2+ influx and neuronal death augmented glutamate-induced Ca2+ elevation and neuronal death in CGCs. BmK NT1-augmented glutamate-induced Ca2+ influx and neuronal death were suppressed by tetrodotoxin and MK-801 suggesting that the augmentation was through activation of VGSCs and NMDA receptors. Consistently, BmK NT1 also enhanced NMDA-induced Ca2+ influx. Further mechanistic investigations demonstrated that BmK NT1 increased the expression level of NMDA receptors on the plasma membrane and increased the phosphorylation level of NR2B at Tyr1472. Src family kinase inhibitor, 1-tert-butyl-3-(4-chlorophenyl)pyrazolo[3,4-d]pyrimidin-4-yl]amine (PP2), but not the inactive analogue, 4-amino-1-phenylpyrazolo[3,4-d]pyrimidine (PP3), eliminated BmK NT1-triggered NR2B phosphorylation, NMDA receptor trafficking, as well as BmK NT1-augmented NMDA Ca2+ response and neuronal death. Considered together, these data demonstrated that both presynaptic (excitatory amino acid release) and postsynaptic mechanisms (augmentation of NMDA receptor function) are critical for VGSC activation-induced neurotoxicity in primary CGC cultures.
Collapse
|
6
|
Garrison CE, Guan W, Kato M, Tamsett T, Patel T, Sun Y, Pathak TP. Structure-Activity Relationship Evaluation of Wasp Toxin β-PMTX Leads to Analogs with Superior Activity for Human Neuronal Sodium Channels. ACS Med Chem Lett 2020; 11:353-357. [PMID: 32184969 DOI: 10.1021/acsmedchemlett.9b00415] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2019] [Accepted: 10/25/2019] [Indexed: 11/29/2022] Open
Abstract
Beta-pompilidotoxin (β-PMTX) is a 13-amino acid wasp venom peptide that activates human neuronal sodium channel NaV1.1 with weak activity (40% activation at 3.3 μM of β-PMTX). Through rational design of β-PMTX analogs, we have identified peptides with significantly improved activity on human NaV1.1 (1170% activation at 3.3 μM of peptide 18). The underlying structure-activity relationship suggests importance of charge interactions (from residue Lys-3) and lipophilic interactions (from residue Phe-7 and Ser-11). Three top-ranked analogs showed parallel activity improvement for other neuronal sodium channels (human NaV1.2/1.3/1.6/1.7) but not muscular subtypes (NaV1.4/1.5). Finally, we found that analog 16 could partially rescue the pharmacological block imposed by NaV1.1/1.3 selective inhibitor ICA-121431 in cultured mouse cortical GABAergic neurons, demonstrating an activating effect of this peptide on native neuronal sodium channels and its potential utility as a neuropharmacological tool.
Collapse
|
7
|
Ksouri A, Ghedira K, Ben Abderrazek R, Shankar BG, Benkahla A, Bishop OT, Bouhaouala-Zahar B. Homology modeling and docking of AahII-Nanobody complexes reveal the epitope binding site on AahII scorpion toxin. Biochem Biophys Res Commun 2018; 496:1025-1032. [DOI: 10.1016/j.bbrc.2018.01.036] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2017] [Accepted: 01/04/2018] [Indexed: 11/25/2022]
|
8
|
Abstract
Voltage-gated sodium channels (VGSCs) are critical in generation and conduction of electrical signals in multiple excitable tissues. Natural toxins, produced by animal, plant, and microorganisms, target VGSCs through diverse strategies developed over millions of years of evolutions. Studying of the diverse interaction between VGSC and VGSC-targeting toxins has been contributing to the increasing understanding of molecular structure and function, pharmacology, and drug development potential of VGSCs. This chapter aims to summarize some of the current views on the VGSC-toxin interaction based on the established receptor sites of VGSC for natural toxins.
Collapse
Affiliation(s)
- Yonghua Ji
- Laboratory of Neuropharmacology and Neurotoxicology, Shanghai University, Shanghai, China.
| |
Collapse
|
9
|
Zou X, Wu Y, Chen J, Zhao F, Zhang F, Yu B, Cao Z. Activation of sodium channel by a novel α-scorpion toxin, BmK NT2, stimulates ERK1/2 and CERB phosphorylation through a Ca2+ dependent pathway in neocortical neurons. Int J Biol Macromol 2017; 104:70-77. [DOI: 10.1016/j.ijbiomac.2017.05.163] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2017] [Revised: 05/26/2017] [Accepted: 05/28/2017] [Indexed: 12/20/2022]
|
10
|
Hopp BH, Arvidson RS, Adams ME, Razak KA. Arizona bark scorpion venom resistance in the pallid bat, Antrozous pallidus. PLoS One 2017; 12:e0183215. [PMID: 28854259 PMCID: PMC5576675 DOI: 10.1371/journal.pone.0183215] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2016] [Accepted: 08/01/2017] [Indexed: 11/19/2022] Open
Abstract
The pallid bat (Antrozous pallidus), a gleaning bat found in the western United States and Mexico, hunts a wide variety of ground-dwelling prey, including scorpions. Anecdotal evidence suggests that the pallid bat is resistant to scorpion venom, but no systematic study has been performed. Here we show with behavioral measures and direct injection of venom that the pallid bat is resistant to venom of the Arizona bark scorpion, Centruroides sculpturatus. Our results show that the pallid bat is stung multiple times during a hunt without any noticeable effect on behavior. In addition, direct injection of venom at mouse LD50 concentrations (1.5 mg/kg) has no effect on bat behavior. At the highest concentration tested (10 mg/kg), three out of four bats showed no effects. One of the four bats showed a transient effect suggesting that additional studies are required to identify potential regional variation in venom tolerance. Scorpion venom is a cocktail of toxins, some of which activate voltage-gated sodium ion channels, causing intense pain. Dorsal root ganglia (DRG) contain nociceptive neurons and are principal targets of scorpion venom toxins. To understand if mutations in specific ion channels contribute to venom resistance, a pallid bat DRG transcriptome was generated. As sodium channels are a major target of scorpion venom, we identified amino acid substitutions present in the pallid bat that may lead to venom resistance. Some of these substitutions are similar to corresponding amino acids in sodium channel isoforms responsible for reduced venom binding activity. The substitution found previously in the grasshopper mouse providing venom resistance to the bark scorpion is not present in the pallid bat, indicating a potentially novel mechanism for venom resistance in the bat that remains to be identified. Taken together, these results indicate that the pallid bat is resistant to venom of the bark scorpion and altered sodium ion channel function may partly underlie such resistance.
Collapse
Affiliation(s)
- Bradley H. Hopp
- Graduate Neuroscience Program, University of California, Riverside, California, United States of America
| | - Ryan S. Arvidson
- Departments of Entomology and Cell Biology & Neuroscience, University of California, Riverside, California, United States of America
| | - Michael E. Adams
- Graduate Neuroscience Program, University of California, Riverside, California, United States of America
- Departments of Entomology and Cell Biology & Neuroscience, University of California, Riverside, California, United States of America
| | - Khaleel A. Razak
- Graduate Neuroscience Program, University of California, Riverside, California, United States of America
- Department of Psychology, University of California, Riverside, California, United States of America
- * E-mail:
| |
Collapse
|
11
|
Housley DM, Housley GD, Liddell MJ, Jennings EA. Scorpion toxin peptide action at the ion channel subunit level. Neuropharmacology 2016; 127:46-78. [PMID: 27729239 DOI: 10.1016/j.neuropharm.2016.10.004] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2016] [Revised: 09/06/2016] [Accepted: 10/06/2016] [Indexed: 12/19/2022]
Abstract
This review categorizes functionally validated actions of defined scorpion toxin (SCTX) neuropeptides across ion channel subclasses, highlighting key trends in this rapidly evolving field. Scorpion envenomation is a common event in many tropical and subtropical countries, with neuropharmacological actions, particularly autonomic nervous system modulation, causing significant mortality. The primary active agents within scorpion venoms are a diverse group of small neuropeptides that elicit specific potent actions across a wide range of ion channel classes. The identification and functional characterisation of these SCTX peptides has tremendous potential for development of novel pharmaceuticals that advance knowledge of ion channels and establish lead compounds for treatment of excitable tissue disorders. This review delineates the unique specificities of 320 individual SCTX peptides that collectively act on 41 ion channel subclasses. Thus the SCTX research field has significant translational implications for pathophysiology spanning neurotransmission, neurohumoral signalling, sensori-motor systems and excitation-contraction coupling. This article is part of the Special Issue entitled 'Venom-derived Peptides as Pharmacological Tools.'
Collapse
Affiliation(s)
- David M Housley
- College of Medicine and Dentistry, Cairns Campus, James Cook University, Cairns, Queensland 4878, Australia; Translational Neuroscience Facility and Department of Physiology, School of Medical Sciences, UNSW Australia, Sydney, NSW 2052, Australia.
| | - Gary D Housley
- Translational Neuroscience Facility and Department of Physiology, School of Medical Sciences, UNSW Australia, Sydney, NSW 2052, Australia
| | - Michael J Liddell
- Centre for Tropical Environmental and Sustainability Science and College of Science & Engineering, Cairns Campus, James Cook University, Cairns, Queensland 4878, Australia
| | - Ernest A Jennings
- College of Medicine and Dentistry, Cairns Campus, James Cook University, Cairns, Queensland 4878, Australia; Centre for Biodiscovery and Molecular Development of Therapeutics, James Cook University, Queensland 4878, Australia; Australian Institute of Tropical Health and Medicine, James Cook University, Cairns Campus, QLD, Australia
| |
Collapse
|
12
|
Zhang S, Zhu L, Yu J, Xu J, Gao B, Zhou C, Zhu S. Evaluating the potential of a loop-extended scorpion toxin-like peptide as a protein scaffold. Protein Eng Des Sel 2016; 29:607-616. [PMID: 27672050 DOI: 10.1093/protein/gzw051] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2016] [Revised: 08/06/2016] [Accepted: 08/26/2016] [Indexed: 11/14/2022] Open
Abstract
Grafting of exogenous bioactive sites or functional motifs onto structurally stable scaffolds to gain new functions represents an important research direction in protein engineering. Some engineered proteins have been developed into therapeutic drugs. MeuNaTxα-3 (abbreviated as MT-3) is a newly characterized scorpion sodium channel toxin-like peptide isolated from the venom of the scorpion Mesobuthus eupeus, which contains a rigid scaffold highly similar to classical scorpion sodium channel toxins and an extension of eight amino acids in its J-loop region. This extended loop constitutes a flexible region extruded from the scaffold and could be substituted by exogenous functional sequences. In this study, we experimentally evaluated the scaffold potential of MT-3 through grafting two small antimicrobial motifs to replace residues within the loop. Functional assays showed that the two engineered molecules exhibited elevated antimicrobial potency, as compared with the unmodified scaffold, without structural disruption, providing experimental evidence in favor of MT-3 as a promising scaffold in protein engineering.
Collapse
Affiliation(s)
- Shangfei Zhang
- Group of Peptide Biology and Evolution, State Key Laboratory of Integrated Management of Pest Insects and Rodents, Institute of Zoology, Chinese Academy of Sciences, 1 Beichen West Road, Chaoyang District, 100101 Beijing, China
| | - Limei Zhu
- Group of Peptide Biology and Evolution, State Key Laboratory of Integrated Management of Pest Insects and Rodents, Institute of Zoology, Chinese Academy of Sciences, 1 Beichen West Road, Chaoyang District, 100101 Beijing, China
| | - Jie Yu
- State Key Laboratory of Natural Medicines, School of Life Science and Technology, China Pharmaceutical University, 24 TongJiaXiang, 210009 Nanjing, Jiangsu, China
| | - Jun Xu
- State Key Laboratory of Natural Medicines, School of Life Science and Technology, China Pharmaceutical University, 24 TongJiaXiang, 210009 Nanjing, Jiangsu, China
| | - Bin Gao
- Group of Peptide Biology and Evolution, State Key Laboratory of Integrated Management of Pest Insects and Rodents, Institute of Zoology, Chinese Academy of Sciences, 1 Beichen West Road, Chaoyang District, 100101 Beijing, China
| | - Changlin Zhou
- State Key Laboratory of Natural Medicines, School of Life Science and Technology, China Pharmaceutical University, 24 TongJiaXiang, 210009 Nanjing, Jiangsu, China
| | - Shunyi Zhu
- Group of Peptide Biology and Evolution, State Key Laboratory of Integrated Management of Pest Insects and Rodents, Institute of Zoology, Chinese Academy of Sciences, 1 Beichen West Road, Chaoyang District, 100101 Beijing, China
| |
Collapse
|
13
|
Ahern CA, Payandeh J, Bosmans F, Chanda B. The hitchhiker's guide to the voltage-gated sodium channel galaxy. ACTA ACUST UNITED AC 2016; 147:1-24. [PMID: 26712848 PMCID: PMC4692491 DOI: 10.1085/jgp.201511492] [Citation(s) in RCA: 242] [Impact Index Per Article: 26.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Eukaryotic voltage-gated sodium (Nav) channels contribute to the rising phase of action potentials and served as an early muse for biophysicists laying the foundation for our current understanding of electrical signaling. Given their central role in electrical excitability, it is not surprising that (a) inherited mutations in genes encoding for Nav channels and their accessory subunits have been linked to excitability disorders in brain, muscle, and heart; and (b) Nav channels are targeted by various drugs and naturally occurring toxins. Although the overall architecture and behavior of these channels are likely to be similar to the more well-studied voltage-gated potassium channels, eukaryotic Nav channels lack structural and functional symmetry, a notable difference that has implications for gating and selectivity. Activation of voltage-sensing modules of the first three domains in Nav channels is sufficient to open the channel pore, whereas movement of the domain IV voltage sensor is correlated with inactivation. Also, structure–function studies of eukaryotic Nav channels show that a set of amino acids in the selectivity filter, referred to as DEKA locus, is essential for Na+ selectivity. Structures of prokaryotic Nav channels have also shed new light on mechanisms of drug block. These structures exhibit lateral fenestrations that are large enough to allow drugs or lipophilic molecules to gain access into the inner vestibule, suggesting that this might be the passage for drug entry into a closed channel. In this Review, we will synthesize our current understanding of Nav channel gating mechanisms, ion selectivity and permeation, and modulation by therapeutics and toxins in light of the new structures of the prokaryotic Nav channels that, for the time being, serve as structural models of their eukaryotic counterparts.
Collapse
Affiliation(s)
- Christopher A Ahern
- Department of Molecular Physiology and Biophysics, University of Iowa, Iowa City, IA 52242
| | - Jian Payandeh
- Department of Structural Biology, Genentech, Inc., South San Francisco, CA 94080
| | - Frank Bosmans
- Department of Physiology and Solomon H. Snyder Department of Neuroscience, Johns Hopkins University, School of Medicine, Baltimore, MD 21205 Department of Physiology and Solomon H. Snyder Department of Neuroscience, Johns Hopkins University, School of Medicine, Baltimore, MD 21205
| | - Baron Chanda
- Department of Neuroscience and Department of Biomolecular Chemistry, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI 53705 Department of Neuroscience and Department of Biomolecular Chemistry, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI 53705
| |
Collapse
|
14
|
Martin-Eauclaire MF, Salvatierra J, Bosmans F, Bougis PE. The scorpion toxin Bot IX is a potent member of the α-like family and has a unique N-terminal sequence extension. FEBS Lett 2016; 590:3221-32. [DOI: 10.1002/1873-3468.12357] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2016] [Revised: 08/05/2016] [Accepted: 08/05/2016] [Indexed: 11/08/2022]
Affiliation(s)
| | - Juan Salvatierra
- Department of Physiology; School of Medicine; Johns Hopkins University; Baltimore MD USA
| | - Frank Bosmans
- Department of Physiology; School of Medicine; Johns Hopkins University; Baltimore MD USA
- Solomon H. Snyder Department of Neuroscience; School of Medicine; Johns Hopkins University; Baltimore MD USA
| | - Pierre E. Bougis
- Aix Marseille Université; CNRS; CRN2M; UMR7286; PFRN-CAPM; Marseille France
| |
Collapse
|
15
|
Tuluc P, Benedetti B, Coste de Bagneaux P, Grabner M, Flucher BE. Two distinct voltage-sensing domains control voltage sensitivity and kinetics of current activation in CaV1.1 calcium channels. J Gen Physiol 2016; 147:437-49. [PMID: 27185857 PMCID: PMC4886277 DOI: 10.1085/jgp.201611568] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2016] [Accepted: 04/18/2016] [Indexed: 01/25/2023] Open
Abstract
Alternative splicing of the skeletal muscle CaV1.1 voltage-gated calcium channel gives rise to two channel variants with very different gating properties. The currents of both channels activate slowly; however, insertion of exon 29 in the adult splice variant CaV1.1a causes an ∼30-mV right shift in the voltage dependence of activation. Existing evidence suggests that the S3-S4 linker in repeat IV (containing exon 29) regulates voltage sensitivity in this voltage-sensing domain (VSD) by modulating interactions between the adjacent transmembrane segments IVS3 and IVS4. However, activation kinetics are thought to be determined by corresponding structures in repeat I. Here, we use patch-clamp analysis of dysgenic (CaV1.1 null) myotubes reconstituted with CaV1.1 mutants and chimeras to identify the specific roles of these regions in regulating channel gating properties. Using site-directed mutagenesis, we demonstrate that the structure and/or hydrophobicity of the IVS3-S4 linker is critical for regulating voltage sensitivity in the IV VSD, but by itself cannot modulate voltage sensitivity in the I VSD. Swapping sequence domains between the I and the IV VSDs reveals that IVS4 plus the IVS3-S4 linker is sufficient to confer CaV1.1a-like voltage dependence to the I VSD and that the IS3-S4 linker plus IS4 is sufficient to transfer CaV1.1e-like voltage dependence to the IV VSD. Any mismatch of transmembrane helices S3 and S4 from the I and IV VSDs causes a right shift of voltage sensitivity, indicating that regulation of voltage sensitivity by the IVS3-S4 linker requires specific interaction of IVS4 with its corresponding IVS3 segment. In contrast, slow current kinetics are perturbed by any heterologous sequences inserted into the I VSD and cannot be transferred by moving VSD I sequences to VSD IV. Thus, CaV1.1 calcium channels are organized in a modular manner, and control of voltage sensitivity and activation kinetics is accomplished by specific molecular mechanisms within the IV and I VSDs, respectively.
Collapse
Affiliation(s)
- Petronel Tuluc
- Department of Pharmacology and Toxicology, Institute of Pharmacy, University of Innsbruck, A-6020 Innsbruck, Austria
| | - Bruno Benedetti
- Department of Physiology and Medical Physics, Medical University Innsbruck, A-6020 Innsbruck, Austria
| | - Pierre Coste de Bagneaux
- Department of Physiology and Medical Physics, Medical University Innsbruck, A-6020 Innsbruck, Austria
| | - Manfred Grabner
- Department of Medical Genetics, Molecular and Clinical Pharmacology, Medical University Innsbruck, A-6020 Innsbruck, Austria
| | - Bernhard E Flucher
- Department of Physiology and Medical Physics, Medical University Innsbruck, A-6020 Innsbruck, Austria
| |
Collapse
|
16
|
Tao H, Chen X, Lu M, Wu Y, Deng M, Zeng X, Liu Z, Liang S. Molecular determinant for the tarantula toxin Jingzhaotoxin-I slowing the fast inactivation of voltage-gated sodium channels. Toxicon 2015; 111:13-21. [PMID: 26721415 DOI: 10.1016/j.toxicon.2015.12.009] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2015] [Revised: 11/23/2015] [Accepted: 12/16/2015] [Indexed: 12/19/2022]
Abstract
Peptide toxins often have divergent pharmacological functions and are powerful tools for a deep review on the current understanding of the structure-function relationships of voltage-gated sodium channels (VGSCs). However, knowing about the interaction of site 3 toxins from tarantula venoms with VGSCs is not sufficient. In the present study, using whole-cell patch clamp technique, we determined the effects of Jingzhaotoxin-I (JZTX-I) on five VGSC subtypes expressed in HEK293 cells. The results showed that JZTX-I could inhibit the inactivation of rNav1.2, rNav1.3, rNav1.4, hNav1.5 and hNav1.7 channels with the IC50 of 870 ± 8 nM, 845 ± 4 nM, 339 ± 5 nM, 335 ± 9 nM, and 348 ± 6 nM, respectively. The affinity of the toxin interaction with subtypes (rNav1.4, hNav1.5, and hNav1.7) was only 2-fold higher than that for subtypes (rNav1.2 and rNav1.3). The toxin delayed the inactivation of VGSCs without affecting the activation and steady-state inactivation kinetics in the physiological range of voltages. Site-directed mutagenesis indicated that the toxin interacted with site 3 located at the extracellular S3-S4 linker of domain IV, and the acidic residue Asp at the position1609 in hNav1.5 was crucial for JZTX-I activity. Our results provide new insights in single key residue that allows toxins to recognize distinct ion channels with similar potency and enhance our understanding of the structure-function relationships of toxin-channel interactions.
Collapse
Affiliation(s)
- Huai Tao
- Department of Biochemistry and Molecular Biology, Hunan University of Chinese Medicine, Changsha 410208, Hunan, China; Division of Stem Cell Regulation and Application, Hunan University of Chinese Medicine, Changsha 410208, Hunan, China.
| | - Xia Chen
- Department of Orthopedics, The Second Xiangya Hospital of Central South University, Changsha 410011, Hunan, China
| | - Min Lu
- Key Laboratory of Protein Chemistry and Developmental Biology of Ministry of Education, College of Life Sciences, Hunan Normal University, Changsha 410081, Hunan, China
| | - Yuanyuan Wu
- Key Laboratory of Protein Chemistry and Developmental Biology of Ministry of Education, College of Life Sciences, Hunan Normal University, Changsha 410081, Hunan, China
| | - Meichun Deng
- State Key Laboratory of Medical Genetics and School of Life Sciences, Central South University, Changsha 410013, Hunan, China
| | - Xiongzhi Zeng
- Key Laboratory of Protein Chemistry and Developmental Biology of Ministry of Education, College of Life Sciences, Hunan Normal University, Changsha 410081, Hunan, China
| | - Zhonghua Liu
- Key Laboratory of Protein Chemistry and Developmental Biology of Ministry of Education, College of Life Sciences, Hunan Normal University, Changsha 410081, Hunan, China
| | - Songping Liang
- Key Laboratory of Protein Chemistry and Developmental Biology of Ministry of Education, College of Life Sciences, Hunan Normal University, Changsha 410081, Hunan, China.
| |
Collapse
|
17
|
Computational Studies of Venom Peptides Targeting Potassium Channels. Toxins (Basel) 2015; 7:5194-211. [PMID: 26633507 PMCID: PMC4690127 DOI: 10.3390/toxins7124877] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2015] [Revised: 11/13/2015] [Accepted: 11/20/2015] [Indexed: 01/18/2023] Open
Abstract
Small peptides isolated from the venom of animals are potential scaffolds for ion channel drug discovery. This review article mainly focuses on the computational studies that have advanced our understanding of how various toxins interfere with the function of K+ channels. We introduce the computational tools available for the study of toxin-channel interactions. We then discuss how these computational tools have been fruitfully applied to elucidate the mechanisms of action of a wide range of venom peptides from scorpions, spiders, and sea anemone.
Collapse
|
18
|
Feng YJ, Feng Q, Tao J, Zhao R, Ji YH. Allosteric interactions between receptor site 3 and 4 of voltage-gated sodium channels: a novel perspective for the underlying mechanism of scorpion sting-induced pain. J Venom Anim Toxins Incl Trop Dis 2015; 21:42. [PMID: 26491429 PMCID: PMC4612427 DOI: 10.1186/s40409-015-0043-6] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2015] [Accepted: 10/13/2015] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND BmK I, a site-3-specific modulator of voltage-gated sodium channels (VGSCs), causes pain and hyperalgesia in rats, while BmK IT2, a site-4-specific modulator of VGSCs, suppresses pain-related responses. A stronger pain-related effect has been previously attributed to Buthus martensi Karsch (BmK) venom, which points out the joint pharmacological effect in the crude venom. METHODS In order to detect the joint effect of BmK I and BmK IT2 on ND7-23 cells, the membrane current was measured by whole cell recording. BmK I and BmK IT2 were applied successively and jointly, and the synergistic modulations of VGSCs on ND7-23 cells were detected. RESULTS Larger peak INa and more negative half-activation voltage were elicited by joint application of BmK I and BmK IT2 than by application of BmK I or BmK IT2 alone. Compared to the control, co-applied BmK I and BmK IT2 also significantly prolonged the time constant of inactivation. CONCLUSIONS Our results indicated that site-4 toxin (BmK IT2) could enhance the pharmacological effect induced by site-3 toxin (BmK I), suggesting a stronger effect elicited by both toxins that alone usually exhibit opposite pharmacological effects, which is related to the allosteric interaction between receptor site 3 and site 4. Meanwhile, these results may bring a novel perspective for exploring the underlying mechanisms of scorpion sting-induced pain.
Collapse
Affiliation(s)
- Yi-Jun Feng
- Laboratory of Neuropharmacology and Neurotoxicology, Shanghai University, Nanchen Road 333, Shanghai, 200444 China
| | - Qi Feng
- Laboratory of Neuropharmacology and Neurotoxicology, Shanghai University, Nanchen Road 333, Shanghai, 200444 China
| | - Jie Tao
- Department of Nephrology, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, 164 Lanxi Road, Shanghai, 200062 China ; Department of Pharmacology, School of Pharmacy, Fudan University, 826 Zhangheng Road, Shanghai, 201203 China
| | - Rong Zhao
- Department of Neuroscience, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030 USA
| | - Yong-Hua Ji
- Laboratory of Neuropharmacology and Neurotoxicology, Shanghai University, Nanchen Road 333, Shanghai, 200444 China
| |
Collapse
|
19
|
Abstract
It is long known that peptide neurotoxins derived from a diversity of venomous animals evolve by positive selection following gene duplication, yet a force that drives their adaptive evolution remains a mystery. By using maximum-likelihood models of codon substitution, we analyzed molecular adaptation in scorpion sodium channel toxins from a specific species and found ten positively selected sites, six of which are located at the core-domain of scorpion α-toxins, a region known to interact with two adjacent loops in the voltage-sensor domain (DIV) of sodium channels, as validated by our newly constructed computational model of toxin-channel complex. Despite the lack of positive selection signals in these two loops, they accumulated extensive sequence variations by relaxed purifying selection in prey and predators of scorpions. The evolutionary variability in the toxin-bound regions of sodium channels indicates that accelerated substitutions in the multigene family of scorpion toxins is a consequence of dealing with the target diversity. This work presents an example of atypical co-evolution between animal toxins and their molecular targets, in which toxins suffered from more prominent selective pressure from the channels of their competitors. Our discovery helps explain the evolutionary rationality of gene duplication of toxins in a specific venomous species.
Collapse
|
20
|
Martin-Eauclaire MF, Ferracci G, Bosmans F, Bougis PE. A surface plasmon resonance approach to monitor toxin interactions with an isolated voltage-gated sodium channel paddle motif. ACTA ACUST UNITED AC 2015; 145:155-62. [PMID: 25624450 PMCID: PMC4306711 DOI: 10.1085/jgp.201411268] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
The isolated Nav channel domain IV paddle motif remains susceptible to toxins that inhibit fast inactivation. Animal toxins that inhibit voltage-gated sodium (Nav) channel fast inactivation can do so through an interaction with the S3b–S4 helix-turn-helix region, or paddle motif, located in the domain IV voltage sensor. Here, we used surface plasmon resonance (SPR), an optical approach that uses polarized light to measure the refractive index near a sensor surface to which a molecule of interest is attached, to analyze interactions between the isolated domain IV paddle and Nav channel–selective α-scorpion toxins. Our SPR analyses showed that the domain IV paddle can be removed from the Nav channel and immobilized on sensor chips, and suggest that the isolated motif remains susceptible to animal toxins that target the domain IV voltage sensor. As such, our results uncover the inherent pharmacological sensitivities of the isolated domain IV paddle motif, which may be exploited to develop a label-free SPR approach for discovering ligands that target this region.
Collapse
Affiliation(s)
- Marie-France Martin-Eauclaire
- Centre National de la Recherche Scientifique, Centre de Recherche en Neurobiologie et Neurophysiologie de Marseillle, Unité Mixte de Recherche 7286, Plates-Formes de Recherche en Neurosciences-Centre d'Analyse Protéomique de Marseille, Aix Marseille Université, 13344 Marseille, France
| | - Géraldine Ferracci
- Centre National de la Recherche Scientifique, Centre de Recherche en Neurobiologie et Neurophysiologie de Marseillle, Unité Mixte de Recherche 7286, Plates-Formes de Recherche en Neurosciences-Centre d'Analyse Protéomique de Marseille, Aix Marseille Université, 13344 Marseille, France
| | - Frank Bosmans
- Department of Physiology and Solomon H. Snyder Department of Neuroscience, Johns Hopkins University, School of Medicine, Baltimore, MD 21205 Department of Physiology and Solomon H. Snyder Department of Neuroscience, Johns Hopkins University, School of Medicine, Baltimore, MD 21205
| | - Pierre E Bougis
- Centre National de la Recherche Scientifique, Centre de Recherche en Neurobiologie et Neurophysiologie de Marseillle, Unité Mixte de Recherche 7286, Plates-Formes de Recherche en Neurosciences-Centre d'Analyse Protéomique de Marseille, Aix Marseille Université, 13344 Marseille, France
| |
Collapse
|
21
|
Jankowska M, Pawlowska-Mainville A, Stankiewicz M, Rogalska J, Wyszkowska J. Exposure to 50 Hz electromagnetic field changes the efficiency of the scorpion alpha toxin. J Venom Anim Toxins Incl Trop Dis 2015; 21:38. [PMID: 26430395 PMCID: PMC4589959 DOI: 10.1186/s40409-015-0040-9] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2015] [Accepted: 09/24/2015] [Indexed: 11/17/2022] Open
Abstract
BACKGROUND Extremely low-frequency (50 Hz) electromagnetic field (ELF-EMF) is produced by electric power transmission lines and electronic devices of everyday use. Some phenomena are proposed as "first effects" of ELF-EMF: the discrete changes in the membrane potential and the increase of the calcium channel activity as well as the intracellular concentration of Ca(2+). Interaction of the scorpion alpha toxin with the sodium channel depends on the orientation of the charges and may be perturbed by changes in the membrane polarization. The toxin induces overexcitability in the nervous system and an increase in the neurotransmitters released with different consequences, mainly the paralysis of muscles. We assumed that the exposure to ELF-EMF 0.7 mT will change the effects of the insect selective scorpion alpha toxin (recombinant LqhαIT from Leiurus quinquestriatus hebraeus) at the level of the cercal nerve function, the synaptic transmission and on the level of entire insect organism. Taking into account the compensatory mechanisms in organisms, we tested in addition ten times higher ELF-EMF on whole insects. METHODS Experiments were performed in vivo on cockroaches (Periplaneta americana) and in vitro - on isolated cockroach abdominal nerve cord with cerci. In biotests, the effects of LqhαIT (10(-8) M) were estimated on the basis of the insect ability to turn back from dorsal to ventral side. Three groups were compared: the control one and the two exposed to ELF-EMF - 0.7 and 7 mT. Bioelectrical activity of the cercal nerve and of the connective nerve that leaves the terminal abdominal ganglion was recorded using extracellular electrodes. LqhαIT (5 × 10(-8) M) induced modifications of neuronal activity that were observed in the control cockroach preparations and in the ones exposed to ELF-EMF (0.7 mT). The exposure to ELF-EMF was carried out using coils with a size appropriate to the examined objects. RESULTS The exposure to ELF-EMF (0.7 mT) modified the effects of LqhαIT (5 × 10(-8) M) on activity of the cercal nerve and of the connective nerve. We observed a decrease of the toxin effect on the cercal nerve activity, but the toxic effect of LqhαIT on the connective nerve was increased. Biotests showed that toxicity of LqhαIT (10(-8) M) on cockroaches was reduced by the exposure to ELF-EMF (0.7 and 7 mT). CONCLUSIONS The exposure to 50 Hz ELF-EMF modified the mode of action of the anti-insect scorpion alpha toxin LqhαIT at cellular level of the cockroach nervous system and in biotests. Toxin appeared as a usefull tool in distinguishing between the primary and the secondary effects of ELF-EMF.
Collapse
Affiliation(s)
- Milena Jankowska
- />Nicolaus Copernicus University, Faculty of Biology and Environmental Protection, Torun, Poland
| | | | - Maria Stankiewicz
- />Nicolaus Copernicus University, Faculty of Biology and Environmental Protection, Torun, Poland
| | - Justyna Rogalska
- />Nicolaus Copernicus University, Faculty of Biology and Environmental Protection, Torun, Poland
| | - Joanna Wyszkowska
- />Nicolaus Copernicus University, Faculty of Biology and Environmental Protection, Torun, Poland
| |
Collapse
|
22
|
Camargos TS, Bosmans F, Rego SC, Mourão CBF, Schwartz EF. The Scorpion Toxin Tf2 from Tityus fasciolatus Promotes Nav1.3 Opening. PLoS One 2015; 10:e0128578. [PMID: 26083731 PMCID: PMC4470819 DOI: 10.1371/journal.pone.0128578] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2014] [Accepted: 04/29/2015] [Indexed: 11/23/2022] Open
Abstract
We identified Tf2, the first β-scorpion toxin from the venom of the Brazilian scorpion Tityus fasciolatus. Tf2 is identical to Tb2-II found in Tityus bahiensis. We found that Tf2 selectively activates human (h)Nav1.3, a neuronal voltage-gated sodium (Nav) subtype implicated in epilepsy and nociception. Tf2 shifts hNav1.3 activation voltage to more negative values, thereby opening the channel at resting membrane potentials. Seven other tested mammalian Nav channels (Nav1.1-1.2; Nav1.4-1.8) expressed in Xenopus oocytes are insensitive upon application of 1 μM Tf2. Therefore, the identification of Tf2 represents a unique addition to the repertoire of animal toxins that can be used to investigate Nav channel function.
Collapse
Affiliation(s)
- Thalita S. Camargos
- Departamento de Ciências Fisiológicas, Laboratório de Toxinologia, Universidade de Brasília, Brasília, DF, Brazil
| | - Frank Bosmans
- Department of Physiology, Johns Hopkins University—School of Medicine, Baltimore, MD, United States of America
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University—School of Medicine, Baltimore, MD, United States of America
| | - Solange C. Rego
- Departamento de Ciências Fisiológicas, Laboratório de Toxinologia, Universidade de Brasília, Brasília, DF, Brazil
| | - Caroline B. F. Mourão
- Departamento de Ciências Fisiológicas, Laboratório de Toxinologia, Universidade de Brasília, Brasília, DF, Brazil
| | - Elisabeth F. Schwartz
- Departamento de Ciências Fisiológicas, Laboratório de Toxinologia, Universidade de Brasília, Brasília, DF, Brazil
- * E-mail:
| |
Collapse
|
23
|
The specificity of Av3 sea anemone toxin for arthropods is determined at linker DI/SS2-S6 in the pore module of target sodium channels. Biochem J 2014; 463:271-7. [PMID: 25055135 DOI: 10.1042/bj20140576] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Av3 is a peptide neurotoxin from the sea anemone Anemonia viridis that shows specificity for arthropod voltage-gated sodium channels (Navs). Interestingly, Av3 competes with a scorpion α-toxin on binding to insect Navs and similarly inhibits the inactivation process, and thus has been classified as 'receptor site-3 toxin', although the two peptides are structurally unrelated. This raises questions as to commonalities and differences in the way both toxins interact with Navs. Recently, site-3 was partly resolved for scorpion α-toxins highlighting S1-S2 and S3-S4 external linkers at the DIV voltage-sensor module and the juxtaposed external linkers at the DI pore module. To uncover channel determinants involved in Av3 specificity for arthropods, the toxin was examined on channel chimaeras constructed with the external linkers of the mammalian brain Nav1.2a, which is insensitive to Av3, in the background of the Drosophila DmNav1. This approach highlighted the role of linker DI/SS2-S6, adjacent to the channel pore, in determining Av3 specificity. Point mutagenesis at DI/SS2-S6 accompanied by functional assays highlighted Trp404 and His405 as a putative point of Av3 interaction with DmNav1. His405 conservation in arthropod Navs compared with tyrosine in vertebrate Navs may represent an ancient substitution that explains the contemporary selectivity of Av3. Trp404 and His405 localization near the membrane surface and the hydrophobic bioactive surface of Av3 suggest that the toxin possibly binds at a cleft by DI/S6. A partial overlap in receptor site-3 of both toxins nearby DI/S6 may explain their binding competition capabilities.
Collapse
|
24
|
Xu L, Li T, Liu H, Yang F, Liang S, Cao Z, Li W, Wu Y. Functional characterization of two novel scorpion sodium channel toxins from Lychas mucronatus. Toxicon 2014; 90:318-25. [DOI: 10.1016/j.toxicon.2014.08.075] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2014] [Revised: 08/11/2014] [Accepted: 08/27/2014] [Indexed: 10/24/2022]
|
25
|
Wheat germ in vitro translation to produce one of the most toxic sodium channel specific toxins. Biosci Rep 2014; 34:BSR20140050. [PMID: 24924257 PMCID: PMC4114062 DOI: 10.1042/bsr20140050] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Envenoming following scorpion sting is a common emergency in many parts of the world. During scorpion envenoming, highly toxic small polypeptides of the venom diffuse rapidly within the victim causing serious medical problems. The exploration of toxin structure-function relationship would benefit from the generation of soluble recombinant scorpion toxins in Escherichia coli. We developed an in vitro wheat germ translation system for the expression of the highly toxic Aah (Androctonus australis hector)II protein that requires the proper formation of four disulphide bonds. Soluble, recombinant N-terminal GST (glutathione S-transferase)-tagged AahII toxin is obtained in this in vitro translation system. After proteolytic removal of the GST-tag, purified rAahII (recombinant AahII) toxin, which contains two extra amino acids at its N terminal relative to the native AahII, is highly toxic after i.c.v. (intracerebroventricular) injection in Swiss mice. An LD50 (median lethal dose)-value of 10 ng (or 1.33 pmol), close to that of the native toxin (LD50 of 3 ng) indicates that the wheat germ in vitro translation system produces properly folded and biological active rAahII. In addition, NbAahII10 (Androctonus australis hector nanobody 10), a camel single domain antibody fragment, raised against the native AahII toxin, recognizes its cognate conformational epitope on the recombinant toxin and neutralizes the toxicity of purified rAahII upon injection in mice. A wheat germ embryo derived cell-free translation system expresses a biologically active, highly toxic scorpion venom protein that is fully neutralized by a camel single domain antibody fragment raised against the native scorpion toxin.
Collapse
|
26
|
Dong K, Du Y, Rinkevich F, Nomura Y, Xu P, Wang L, Silver K, Zhorov BS. Molecular biology of insect sodium channels and pyrethroid resistance. INSECT BIOCHEMISTRY AND MOLECULAR BIOLOGY 2014; 50:1-17. [PMID: 24704279 PMCID: PMC4484874 DOI: 10.1016/j.ibmb.2014.03.012] [Citation(s) in RCA: 312] [Impact Index Per Article: 28.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/20/2014] [Revised: 03/24/2014] [Accepted: 03/24/2014] [Indexed: 05/06/2023]
Abstract
Voltage-gated sodium channels are essential for the initiation and propagation of the action potential in neurons and other excitable cells. Because of their critical roles in electrical signaling, sodium channels are targets of a variety of naturally occurring and synthetic neurotoxins, including several classes of insecticides. This review is intended to provide an update on the molecular biology of insect sodium channels and the molecular mechanism of pyrethroid resistance. Although mammalian and insect sodium channels share fundamental topological and functional properties, most insect species carry only one sodium channel gene, compared to multiple sodium channel genes found in each mammalian species. Recent studies showed that two posttranscriptional mechanisms, alternative splicing and RNA editing, are involved in generating functional diversity of sodium channels in insects. More than 50 sodium channel mutations have been identified to be responsible for or associated with knockdown resistance (kdr) to pyrethroids in various arthropod pests and disease vectors. Elucidation of molecular mechanism of kdr led to the identification of dual receptor sites of pyrethroids on insect sodium channels. Many of the kdr mutations appear to be located within or close to the two receptor sites. The accumulating knowledge of insect sodium channels and their interactions with insecticides provides a foundation for understanding the neurophysiology of sodium channels in vivo and the development of new and safer insecticides for effective control of arthropod pests and human disease vectors.
Collapse
Affiliation(s)
- Ke Dong
- Department of Entomology, Neuroscience and Genetics Programs, Michigan State University, East Lansing, MI, USA.
| | - Yuzhe Du
- Department of Entomology, Neuroscience and Genetics Programs, Michigan State University, East Lansing, MI, USA
| | - Frank Rinkevich
- Department of Entomology, Neuroscience and Genetics Programs, Michigan State University, East Lansing, MI, USA
| | - Yoshiko Nomura
- Department of Entomology, Neuroscience and Genetics Programs, Michigan State University, East Lansing, MI, USA
| | - Peng Xu
- Department of Entomology, Neuroscience and Genetics Programs, Michigan State University, East Lansing, MI, USA
| | - Lingxin Wang
- Department of Entomology, Neuroscience and Genetics Programs, Michigan State University, East Lansing, MI, USA
| | - Kristopher Silver
- Department of Anatomy and Physiology, Kansas State University, Manhattan, KS, USA
| | - Boris S Zhorov
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Ontario, Canada; Sechenov Institute of Evolutionary Physiology & Biochemistry, Russian Academy of Sciences, St. Petersburg, Russia
| |
Collapse
|
27
|
Gao R, Du Y, Wang L, Nomura Y, Satar G, Gordon D, Gurevitz M, Goldin AL, Dong K. Sequence variations at I260 and A1731 contribute to persistent currents in Drosophila sodium channels. Neuroscience 2014; 268:297-308. [PMID: 24662849 DOI: 10.1016/j.neuroscience.2014.03.028] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2013] [Revised: 02/02/2014] [Accepted: 03/09/2014] [Indexed: 12/19/2022]
Abstract
Tetrodotoxin-sensitive persistent sodium currents, INaP, that activate at subthreshold voltages, have been detected in numerous vertebrate and invertebrate neurons. These currents are believed to be critical for regulating neuronal excitability. However, the molecular mechanism underlying INaP is controversial. In this study, we identified an INaP with a broad range of voltage dependence, from -60mV to 20mV, in a Drosophila sodium channel variant expressed in Xenopus oocytes. Mutational analysis revealed that two variant-specific amino acid changes, I260T in the S4-S5 linker of domain I (ILS4-S5) and A1731V in the voltage sensor S4 of domain IV (IVS4), contribute to the INaP. I260T is critical for the portion of INaP at hyperpolarized potentials. The T260-mediated INaP is likely the result of window currents flowing in the voltage range where the activation and inactivation curves overlap. A1731V is responsible for impaired inactivation and contributes to the portion of INaP at depolarized potentials. Furthermore, A1731V causes enhanced activity of two site-3 toxins which induce persistent currents by inhibiting the outward movement of IVS4, suggesting that A1731V inhibits the outward movement of IVS4. These results provided molecular evidence for the involvement of distinct mechanisms in the generation of INaP: T260 contributes to INaP via enhancement of the window current, whereas V1731 impairs fast inactivation probably by inhibiting the outward movement of IVS4.
Collapse
Affiliation(s)
- R Gao
- Department of Entomology and Neuroscience Program, Michigan State University, East Lansing, MI 48824, United States
| | - Y Du
- Department of Entomology and Neuroscience Program, Michigan State University, East Lansing, MI 48824, United States
| | - L Wang
- Department of Entomology and Neuroscience Program, Michigan State University, East Lansing, MI 48824, United States
| | - Y Nomura
- Department of Entomology and Neuroscience Program, Michigan State University, East Lansing, MI 48824, United States
| | - G Satar
- Department of Entomology and Neuroscience Program, Michigan State University, East Lansing, MI 48824, United States
| | - D Gordon
- Department of Plant Molecular Biology & Ecology, George S. Wise Faculty of Life Sciences, Tel-Aviv University, Israel
| | - M Gurevitz
- Department of Plant Molecular Biology & Ecology, George S. Wise Faculty of Life Sciences, Tel-Aviv University, Israel
| | - A L Goldin
- Department of Microbiology and Molecular Genetics, University of California, Irvine, CA 92697, United States
| | - K Dong
- Department of Entomology and Neuroscience Program, Michigan State University, East Lansing, MI 48824, United States.
| |
Collapse
|
28
|
Characterization of a novel BmαTX47 toxin modulating sodium channels: the crucial role of expression vectors in toxin pharmacological activity. Toxins (Basel) 2014; 6:816-29. [PMID: 24577584 PMCID: PMC3968363 DOI: 10.3390/toxins6030816] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2013] [Revised: 12/30/2013] [Accepted: 01/20/2014] [Indexed: 12/19/2022] Open
Abstract
Long-chain scorpion toxins with four disulfide bridges exhibit various pharmacological features towards the different voltage-gated sodium channel subtypes. However, the toxin production still remains a huge challenge. Here, we reported the effects of different expression vectors on the pharmacological properties of a novel toxin BmαTX47 from the scorpion Buthus martensii Karsch. The recombinant BmαTX47 was obtained using the expression vector pET-14b and pET-28a, respectively. Pharmacological experiments showed that the recombinant BmαTX47 was a new α-scorpion toxin which could inhibit the fast inactivation of rNav1.2, mNav1.4 and hNav1.5 channels. Importantly, the different expression vectors were found to strongly affect BmαTX47 pharmacological activities while toxins were obtained by the same expression and purification procedures. When 10 µM recombinant BmαTX47 from the pET-28a vector was applied, the values of I5ms/Ipeak for rNav1.2, mNav1.4 and hNav1.5 channels were 44.12% ± 3.17%, 25.40% ± 4.89% and 65.34% ± 3.86%, respectively, which were better than those values of 11.33% ± 1.46%, 15.96% ± 1.87% and 5.24% ± 2.38% for rNav1.2, mNav1.4 and hNav1.5 channels delayed by 10 µM recombinant BmαTX47 from the pET-14b vector. The dose-response experiments further indicated the EC50 values of recombinant BmαTX47 from the pET-28a vector were 7262.9 ± 755.9 nM for rNav1.2 channel and 1005.8 ± 118.6 nM for hNav1.5 channel, respectively. Together, these findings highlighted the important role of expression vectors in scorpion toxin pharmacological properties, which would accelerate the understanding of the structure-function relationships of scorpion toxins and promote the potential application of toxins in the near future.
Collapse
|
29
|
Abstract
The mechanism by which voltage-gated ion channels respond to changes in membrane polarization during action potential signaling in excitable cells has been the subject of research attention since the original description of voltage-dependent sodium and potassium flux in the squid giant axon. The cloning of ion channel genes and the identification of point mutations associated with channelopathy diseases in muscle and brain has facilitated an electrophysiological approach to the study of ion channels. Experimental approaches to the study of voltage gating have incorporated the use of thiosulfonate reagents to test accessibility, fluorescent probes, and toxins to define domain-specific roles of voltage-sensing S4 segments. Crystallography, structural and homology modeling, and molecular dynamics simulations have added computational approaches to study the relationship of channel structure to function. These approaches have tested models of voltage sensor translocation in response to membrane depolarization and incorporate the role of negative countercharges in the S1 to S3 segments to define our present understanding of the mechanism by which the voltage sensor module dictates gating particle permissiveness in excitable cells.
Collapse
Affiliation(s)
- James R Groome
- Department of Biological Sciences, Idaho State University, Pocatello, ID, 83209, USA,
| |
Collapse
|
30
|
Ma Z, Kong J, Gordon D, Gurevitz M, Kallen RG. Direct evidence that scorpion α-toxins (site-3) modulate sodium channel inactivation by hindrance of voltage-sensor movements. PLoS One 2013; 8:e77758. [PMID: 24302985 PMCID: PMC3841157 DOI: 10.1371/journal.pone.0077758] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2013] [Accepted: 09/07/2013] [Indexed: 01/09/2023] Open
Abstract
The position of the voltage-sensing transmembrane segment, S4, in voltage-gated ion channels as a function of voltage remains incompletely elucidated. Site-3 toxins bind primarily to the extracellular loops connecting transmembrane helical segments S1-S2 and S3-S4 in Domain 4 (D4) and S5-S6 in Domain 1 (D1) and slow fast-inactivation of voltage-gated sodium channels. As S4 of the human skeletal muscle voltage-gated sodium channel, hNav1.4, moves in response to depolarization from the resting to the inactivated state, two D4S4 reporters (R2C and R3C, Arg1451Cys and Arg1454Cys, respectively) move from internal to external positions as deduced by reactivity to internally or externally applied sulfhydryl group reagents, methane thiosulfonates (MTS). The changes in reporter reactivity, when cycling rapidly between hyperpolarized and depolarized voltages, enabled determination of the positions of the D4 voltage-sensor and of its rate of movement. Scorpion α-toxin binding impedes D4S4 segment movement during inactivation since the modification rates of R3C in hNav1.4 with methanethiosulfonate (CH3SO2SCH2CH2R, where R = -N(CH3)3 (+) trimethylammonium, MTSET) and benzophenone-4-carboxamidocysteine methanethiosulfonate (BPMTS) were slowed ~10-fold in toxin-modified channels. Based upon the different size, hydrophobicity and charge of the two reagents it is unlikely that the change in reactivity is due to direct or indirect blockage of access of this site to reagent in the presence of toxin (Tx), but rather is the result of inability of this segment to move outward to the normal extent and at the normal rate in the toxin-modified channel. Measurements of availability of R3C to internally applied reagent show decreased access (slower rates of thiol reaction) providing further evidence for encumbered D4S4 movement in the presence of toxins consistent with the assignment of at least part of the toxin binding site to the region of D4S4 region of the voltage-sensor module.
Collapse
Affiliation(s)
- Zhongming Ma
- Department of Biochemistry and Biophysics, Perelman School of Medicine University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Jun Kong
- Department of Biochemistry and Biophysics, Perelman School of Medicine University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Dalia Gordon
- Department of Plant Molecular Biology and Ecology, George S. Wise Faculty of Life Sciences, Tel Aviv University, Ramat Aviv, Tel Aviv, Israel
| | - Michael Gurevitz
- Department of Plant Molecular Biology and Ecology, George S. Wise Faculty of Life Sciences, Tel Aviv University, Ramat Aviv, Tel Aviv, Israel
| | - Roland G. Kallen
- Department of Biochemistry and Biophysics, Perelman School of Medicine University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
- The Mahoney Institute for Neuroscience, Perelman School of Medicine University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| |
Collapse
|
31
|
Smith JJ, Herzig V, King GF, Alewood PF. The insecticidal potential of venom peptides. Cell Mol Life Sci 2013; 70:3665-93. [PMID: 23525661 PMCID: PMC11114029 DOI: 10.1007/s00018-013-1315-3] [Citation(s) in RCA: 74] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2012] [Revised: 02/27/2013] [Accepted: 02/28/2013] [Indexed: 12/19/2022]
Abstract
Pest insect species are a burden to humans as they destroy crops and serve as vectors for a wide range of diseases including malaria and dengue. Chemical insecticides are currently the dominant approach for combating these pests. However, the de-registration of key classes of chemical insecticides due to their perceived ecological and human health risks in combination with the development of insecticide resistance in many pest insect populations has created an urgent need for improved methods of insect pest control. The venoms of arthropod predators such as spiders and scorpions are a promising source of novel insecticidal peptides that often have different modes of action to extant chemical insecticides. These peptides have been optimized via a prey-predator arms race spanning hundreds of millions of years to target specific types of insect ion channels and receptors. Here we review the current literature on insecticidal venom peptides, with a particular focus on their structural and pharmacological diversity, and discuss their potential for deployment as insecticides.
Collapse
Affiliation(s)
- Jennifer J. Smith
- Institute for Molecular Bioscience, The University of Queensland, St. Lucia, QLD 4072 Australia
| | - Volker Herzig
- Institute for Molecular Bioscience, The University of Queensland, St. Lucia, QLD 4072 Australia
| | - Glenn F. King
- Institute for Molecular Bioscience, The University of Queensland, St. Lucia, QLD 4072 Australia
| | - Paul F. Alewood
- Institute for Molecular Bioscience, The University of Queensland, St. Lucia, QLD 4072 Australia
| |
Collapse
|
32
|
Chugunov AO, Koromyslova AD, Berkut AA, Peigneur S, Tytgat J, Polyansky AA, Pentkovsky VM, Vassilevski AA, Grishin EV, Efremov RG. Modular organization of α-toxins from scorpion venom mirrors domain structure of their targets, sodium channels. J Biol Chem 2013; 288:19014-27. [PMID: 23637230 DOI: 10.1074/jbc.m112.431650] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
To gain success in the evolutionary "arms race," venomous animals such as scorpions produce diverse neurotoxins selected to hit targets in the nervous system of prey. Scorpion α-toxins affect insect and/or mammalian voltage-gated sodium channels (Na(v)s) and thereby modify the excitability of muscle and nerve cells. Although more than 100 α-toxins are known and a number of them have been studied into detail, the molecular mechanism of their interaction with Na(v)s is still poorly understood. Here, we employ extensive molecular dynamics simulations and spatial mapping of hydrophobic/hydrophilic properties distributed over the molecular surface of α-toxins. It is revealed that despite the small size and relatively rigid structure, these toxins possess modular organization from structural, functional, and evolutionary perspectives. The more conserved and rigid "core module" is supplemented with the "specificity module" (SM) that is comparatively flexible and variable and determines the taxon (mammal versus insect) specificity of α-toxin activity. We further show that SMs in mammal toxins are more flexible and hydrophilic than in insect toxins. Concomitant sequence-based analysis of the extracellular loops of Na(v)s suggests that α-toxins recognize the channels using both modules. We propose that the core module binds to the voltage-sensing domain IV, whereas the more versatile SM interacts with the pore domain in repeat I of Na(v)s. These findings corroborate and expand the hypothesis on different functional epitopes of toxins that has been reported previously. In effect, we propose that the modular structure in toxins evolved to match the domain architecture of Na(v)s.
Collapse
Affiliation(s)
- Anton O Chugunov
- M. M. Shemyakin and Yu. A. Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, 117997 Moscow, Russia.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Chen R, Chung SH. Binding Modes and Functional Surface of Anti-mammalian Scorpion α-Toxins to Sodium Channels. Biochemistry 2012; 51:7775-82. [DOI: 10.1021/bi300776g] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Affiliation(s)
- Rong Chen
- Research School of Biology, Australian National University, Canberra, ACT 0200, Australia
| | - Shin-Ho Chung
- Research School of Biology, Australian National University, Canberra, ACT 0200, Australia
| |
Collapse
|
34
|
Nardi A, Damann N, Hertrampf T, Kless A. Advances in targeting voltage-gated sodium channels with small molecules. ChemMedChem 2012; 7:1712-40. [PMID: 22945552 DOI: 10.1002/cmdc.201200298] [Citation(s) in RCA: 59] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2012] [Revised: 07/30/2012] [Indexed: 12/19/2022]
Abstract
Blockade of voltage-gated sodium channels (VGSCs) has been used successfully in the clinic to enable control of pathological firing patterns that occur in conditions as diverse as chronic pain, epilepsy, and arrhythmias. Herein we review the state of the art in marketed sodium channel inhibitors, including a brief compendium of their binding sites and of the cellular and molecular biology of sodium channels. Despite the preferential action of this drug class toward over-excited cells, which significantly limits potential undesired side effects on other cells, the need to develop a second generation of sodium channel inhibitors to overcome their critical clinical shortcomings is apparent. Current approaches in drug discovery to deliver novel and truly innovative sodium channel inhibitors is next presented by surveying the most recent medicinal chemistry breakthroughs in the field of small molecules and developments in automated patch-clamp platforms. Various strategies aimed at identifying small molecules that target either particular isoforms of sodium channels involved in specific diseases or anomalous sodium channel currents, irrespective of the isoform by which they have been generated, are critically discussed and revised.
Collapse
Affiliation(s)
- Antonio Nardi
- Global Drug Discovery, Department of Medicinal Chemistry, Grünenthal, Zieglerstrasse 6, 52078 Aachen, Germany.
| | | | | | | |
Collapse
|
35
|
Gurevitz M. Mapping of scorpion toxin receptor sites at voltage-gated sodium channels. Toxicon 2012; 60:502-11. [DOI: 10.1016/j.toxicon.2012.03.022] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2012] [Revised: 03/22/2012] [Accepted: 03/27/2012] [Indexed: 10/28/2022]
|
36
|
Zhang JZ, Yarov-Yarovoy V, Scheuer T, Karbat I, Cohen L, Gordon D, Gurevitz M, Catterall WA. Mapping the interaction site for a β-scorpion toxin in the pore module of domain III of voltage-gated Na(+) channels. J Biol Chem 2012; 287:30719-28. [PMID: 22761417 DOI: 10.1074/jbc.m112.370742] [Citation(s) in RCA: 56] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Activation of voltage-gated sodium (Na(v)) channels initiates and propagates action potentials in electrically excitable cells. β-Scorpion toxins, including toxin IV from Centruroides suffusus suffusus (CssIV), enhance activation of Na(V) channels. CssIV stabilizes the voltage sensor in domain II in its activated state via a voltage-sensor trapping mechanism. Amino acid residues required for the action of CssIV have been identified in the S1-S2 and S3-S4 extracellular loops of domain II. The extracellular loops of domain III are also involved in toxin action, but individual amino acid residues have not been identified. We used site-directed mutagenesis and voltage clamp recording to investigate amino acid residues of domain III that are involved in CssIV action. In the IIISS2-S6 loop, five substitutions at four positions altered voltage-sensor trapping by CssIV(E15A). Three substitutions (E1438A, D1445A, and D1445Y) markedly decreased voltage-sensor trapping, whereas the other two substitutions (N1436G and L1439A) increased voltage-sensor trapping. These bidirectional effects suggest that residues in IIISS2-S6 make both positive and negative interactions with CssIV. N1436G enhanced voltage-sensor trapping via increased binding affinity to the resting state, whereas L1439A increased voltage-sensor trapping efficacy. Based on these results, a three-dimensional model of the toxin-channel interaction was developed using the Rosetta modeling method. These data provide additional molecular insight into the voltage-sensor trapping mechanism of toxin action and define a three-point interaction site for β-scorpion toxins on Na(V) channels. Binding of α- and β-scorpion toxins to two distinct, pseudo-symmetrically organized receptor sites on Na(V) channels acts synergistically to modify channel gating and paralyze prey.
Collapse
Affiliation(s)
- Joel Z Zhang
- Department of Pharmacology, University of Washington, Seattle, WA 98195-7280, USA
| | | | | | | | | | | | | | | |
Collapse
|
37
|
Fabrichny IP, Mondielli G, Conrod S, Martin-Eauclaire MF, Bourne Y, Marchot P. Structural insights into antibody sequestering and neutralizing of Na+ channel α-type modulator from old world scorpion venom. J Biol Chem 2012; 287:14136-48. [PMID: 22371498 DOI: 10.1074/jbc.m111.315382] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
The Old World scorpion Androctonus australis hector (Aah) produces one of the most lethal venoms for humans. Peptidic α-toxins AahI to AahIV are responsible for its potency, with AahII accounting for half of it. All four toxins are high affinity blockers of the fast inactivation phase of mammalian voltage-activated Na(+) channels. However, the high antigenic polymorphism of α-toxins prevents production of a polyvalent neutralizing antiserum, whereas the determinants dictating their trapping by neutralizing antibodies remain elusive. From an anti-AahII mAb, we generated an antigen binding fragment (Fab) with high affinity and selectivity for AahII and solved a 2.3 Å-resolution crystal structure of the complex. Sequestering of the C-terminal region of the bound toxin within a groove formed by the Fab combining loops is associated with a toxin orientation and main and side chain conformations that dictate the AahII antigenic specificity and efficient neutralization. From an anti-AahI mAb, we also preformed and crystallized a high affinity AahI-Fab complex. The 1.6 Å-resolution structure solved revealed a Fab molecule devoid of a bound AahI and with combining loops involved in packing interactions, denoting expulsion of the bound antigen upon crystal formation. Comparative analysis of the groove-like combining site of the toxin-bound anti-AahII Fab and planar combining surface of the unbound anti-AahI Fab along with complementary data from a flexible docking approach suggests occurrence of distinctive trapping orientations for the two toxins relative to their respective Fab. This study provides complementary templates for designing new molecules aimed at capturing Aah α-toxins and suitable for immunotherapy.
Collapse
Affiliation(s)
- Igor P Fabrichny
- Faculté de Médecine Secteur Nord, Centre de Recherche en Neurobiologie-Neurophysiologie de Marseille, CRN2M, CNRS/Aix-Marseille Université UMR-6231, Institut Fédératif de Recherche Jean Roche, CS80011, F-13344 Marseille cedex 15, France
| | | | | | | | | | | |
Collapse
|
38
|
Dai H, Yin S, Li T, Cao Z, Ji Y, Wu Y, Li W. Recombinant expression, purification, and characterization of scorpion toxin BmαTX14. Protein Expr Purif 2012; 82:325-31. [PMID: 22343065 DOI: 10.1016/j.pep.2012.02.001] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2011] [Revised: 02/01/2012] [Accepted: 02/02/2012] [Indexed: 12/19/2022]
Abstract
Long-chain and cysteine-rich scorpion toxins exhibit various pharmacological profiles for different voltage-gated sodium channel subtypes. However, the exploration of toxin structure-function relationships has progressed slowly due to the difficulty of obtaining synthetic or recombinant peptides. We now report that we have established an effective expression and purification approach for the novel scorpion toxin BmαTX14. BmαTX14 was over-expressed as inclusion bodies in Escherichia coli. The insoluble pellet was successfully transformed into active peptide by using a refolding procedure. One-step purification by reverse-phase HPLC was sufficient to generate chromatographically pure peptide. The yield of recombinant toxin reached 4mg from 1L LB medium. The pharmacological data further showed that BmαTX14 selectively inhibited the fast inactivation of mNa(v)1.4 (EC(50)=82.3±15.7nM) rather than that of rNa(v)1.2 (EC(50)>30μM), which indicates that BmαTX14 is a new α-like toxin. This work enables further structural, functional, and pharmacological studies of BmαTX14 and similar toxins.
Collapse
Affiliation(s)
- Hui Dai
- State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan 430072, PR China
| | | | | | | | | | | | | |
Collapse
|