1
|
Pei S, Wang N, Mei Z, Zhangsun D, Craik DJ, McIntosh JM, Zhu X, Luo S. Conotoxins Targeting Voltage-Gated Sodium Ion Channels. Pharmacol Rev 2024; 76:828-845. [PMID: 38914468 PMCID: PMC11331937 DOI: 10.1124/pharmrev.123.000923] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2023] [Revised: 06/17/2024] [Accepted: 06/18/2024] [Indexed: 06/26/2024] Open
Abstract
Voltage-gated sodium (NaV) channels are intimately involved in the generation and transmission of action potentials, and dysfunction of these channels may contribute to nervous system diseases, such as epilepsy, neuropathic pain, psychosis, autism, and cardiac arrhythmia. Many venom peptides selectively act on NaV channels. These include conotoxins, which are neurotoxins secreted by cone snails for prey capture or self-defense but which are also valuable pharmacological tools for the identification and/or treatment of human diseases. Typically, conotoxins contain two or three disulfide bonds, and these internal crossbraces contribute to conotoxins having compact, well defined structures and high stability. Of the conotoxins containing three disulfide bonds, some selectively target mammalian NaV channels and can block, stimulate, or modulate these channels. Such conotoxins have great potential to serve as pharmacological tools for studying the functions and characteristics of NaV channels or as drug leads for neurologic diseases related to NaV channels. Accordingly, discovering or designing conotoxins targeting NaV channels with high potency and selectivity is important. The amino acid sequences, disulfide bond connectivity, and three-dimensional structures are key factors that affect the biological activity of conotoxins, and targeted synthetic modifications of conotoxins can greatly improve their activity and selectivity. This review examines NaV channel-targeted conotoxins, focusing on their structures, activities, and designed modifications, with a view toward expanding their applications. SIGNIFICANCE STATEMENT: NaV channels are crucial in various neurologic diseases. Some conotoxins selectively target NaV channels, causing either blockade or activation, thus enabling their use as pharmacological tools for studying the channels' characteristics and functions. Conotoxins also have promising potential to be developed as drug leads. The disulfide bonds in these peptides are important for stabilizing their structures, thus leading to enhanced specificity and potency. Together, conotoxins targeting NaV channels have both immediate research value and promising future application prospects.
Collapse
Affiliation(s)
- Shengrong Pei
- Guangxi Key Laboratory of Special Biomedicine, School of Medicine, Guangxi University, Nanning, China (S.P., N.W., Z.M., D.Z., X.Z., S.L.); Key Laboratory of Tropical Biological Resources of Ministry of Education, Hainan University, Haikou, China (D.Z., S.L.); Institute for Molecular Bioscience, Australian Research Council Centre of Excellence for Innovations in Peptide and Protein Science, The University of Queensland, Brisbane, Queensland, Australia (D.J.C.); Departments of Biology and Psychiatry, University of Utah, Salt Lake City, Utah (J.M.M.); and George E. Wahlen Veterans Affairs Medical Center, Salt Lake City, Utah (J.M.M.)
| | - Nan Wang
- Guangxi Key Laboratory of Special Biomedicine, School of Medicine, Guangxi University, Nanning, China (S.P., N.W., Z.M., D.Z., X.Z., S.L.); Key Laboratory of Tropical Biological Resources of Ministry of Education, Hainan University, Haikou, China (D.Z., S.L.); Institute for Molecular Bioscience, Australian Research Council Centre of Excellence for Innovations in Peptide and Protein Science, The University of Queensland, Brisbane, Queensland, Australia (D.J.C.); Departments of Biology and Psychiatry, University of Utah, Salt Lake City, Utah (J.M.M.); and George E. Wahlen Veterans Affairs Medical Center, Salt Lake City, Utah (J.M.M.)
| | - Zaoli Mei
- Guangxi Key Laboratory of Special Biomedicine, School of Medicine, Guangxi University, Nanning, China (S.P., N.W., Z.M., D.Z., X.Z., S.L.); Key Laboratory of Tropical Biological Resources of Ministry of Education, Hainan University, Haikou, China (D.Z., S.L.); Institute for Molecular Bioscience, Australian Research Council Centre of Excellence for Innovations in Peptide and Protein Science, The University of Queensland, Brisbane, Queensland, Australia (D.J.C.); Departments of Biology and Psychiatry, University of Utah, Salt Lake City, Utah (J.M.M.); and George E. Wahlen Veterans Affairs Medical Center, Salt Lake City, Utah (J.M.M.)
| | - Dongting Zhangsun
- Guangxi Key Laboratory of Special Biomedicine, School of Medicine, Guangxi University, Nanning, China (S.P., N.W., Z.M., D.Z., X.Z., S.L.); Key Laboratory of Tropical Biological Resources of Ministry of Education, Hainan University, Haikou, China (D.Z., S.L.); Institute for Molecular Bioscience, Australian Research Council Centre of Excellence for Innovations in Peptide and Protein Science, The University of Queensland, Brisbane, Queensland, Australia (D.J.C.); Departments of Biology and Psychiatry, University of Utah, Salt Lake City, Utah (J.M.M.); and George E. Wahlen Veterans Affairs Medical Center, Salt Lake City, Utah (J.M.M.)
| | - David J Craik
- Guangxi Key Laboratory of Special Biomedicine, School of Medicine, Guangxi University, Nanning, China (S.P., N.W., Z.M., D.Z., X.Z., S.L.); Key Laboratory of Tropical Biological Resources of Ministry of Education, Hainan University, Haikou, China (D.Z., S.L.); Institute for Molecular Bioscience, Australian Research Council Centre of Excellence for Innovations in Peptide and Protein Science, The University of Queensland, Brisbane, Queensland, Australia (D.J.C.); Departments of Biology and Psychiatry, University of Utah, Salt Lake City, Utah (J.M.M.); and George E. Wahlen Veterans Affairs Medical Center, Salt Lake City, Utah (J.M.M.)
| | - J Michael McIntosh
- Guangxi Key Laboratory of Special Biomedicine, School of Medicine, Guangxi University, Nanning, China (S.P., N.W., Z.M., D.Z., X.Z., S.L.); Key Laboratory of Tropical Biological Resources of Ministry of Education, Hainan University, Haikou, China (D.Z., S.L.); Institute for Molecular Bioscience, Australian Research Council Centre of Excellence for Innovations in Peptide and Protein Science, The University of Queensland, Brisbane, Queensland, Australia (D.J.C.); Departments of Biology and Psychiatry, University of Utah, Salt Lake City, Utah (J.M.M.); and George E. Wahlen Veterans Affairs Medical Center, Salt Lake City, Utah (J.M.M.)
| | - Xiaopeng Zhu
- Guangxi Key Laboratory of Special Biomedicine, School of Medicine, Guangxi University, Nanning, China (S.P., N.W., Z.M., D.Z., X.Z., S.L.); Key Laboratory of Tropical Biological Resources of Ministry of Education, Hainan University, Haikou, China (D.Z., S.L.); Institute for Molecular Bioscience, Australian Research Council Centre of Excellence for Innovations in Peptide and Protein Science, The University of Queensland, Brisbane, Queensland, Australia (D.J.C.); Departments of Biology and Psychiatry, University of Utah, Salt Lake City, Utah (J.M.M.); and George E. Wahlen Veterans Affairs Medical Center, Salt Lake City, Utah (J.M.M.)
| | - Sulan Luo
- Guangxi Key Laboratory of Special Biomedicine, School of Medicine, Guangxi University, Nanning, China (S.P., N.W., Z.M., D.Z., X.Z., S.L.); Key Laboratory of Tropical Biological Resources of Ministry of Education, Hainan University, Haikou, China (D.Z., S.L.); Institute for Molecular Bioscience, Australian Research Council Centre of Excellence for Innovations in Peptide and Protein Science, The University of Queensland, Brisbane, Queensland, Australia (D.J.C.); Departments of Biology and Psychiatry, University of Utah, Salt Lake City, Utah (J.M.M.); and George E. Wahlen Veterans Affairs Medical Center, Salt Lake City, Utah (J.M.M.)
| |
Collapse
|
2
|
McMahon KL, Vetter I, Schroeder CI. Voltage-Gated Sodium Channel Inhibition by µ-Conotoxins. Toxins (Basel) 2024; 16:55. [PMID: 38251271 PMCID: PMC10819908 DOI: 10.3390/toxins16010055] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2023] [Revised: 01/10/2024] [Accepted: 01/11/2024] [Indexed: 01/23/2024] Open
Abstract
µ-Conotoxins are small, potent pore-blocker inhibitors of voltage-gated sodium (NaV) channels, which have been identified as pharmacological probes and putative leads for analgesic development. A limiting factor in their therapeutic development has been their promiscuity for different NaV channel subtypes, which can lead to undesirable side-effects. This review will focus on four areas of µ-conotoxin research: (1) mapping the interactions of µ-conotoxins with different NaV channel subtypes, (2) µ-conotoxin structure-activity relationship studies, (3) observed species selectivity of µ-conotoxins and (4) the effects of µ-conotoxin disulfide connectivity on activity. Our aim is to provide a clear overview of the current status of µ-conotoxin research.
Collapse
Affiliation(s)
- Kirsten L. McMahon
- Institute for Molecular Biosciences, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Irina Vetter
- Institute for Molecular Biosciences, The University of Queensland, Brisbane, QLD 4072, Australia
- The School of Pharmacy, The University of Queensland, Woolloongabba, QLD 4102, Australia
| | - Christina I. Schroeder
- Institute for Molecular Biosciences, The University of Queensland, Brisbane, QLD 4072, Australia
- Genentech, 1 DNA Way, South San Francisco, CA 94080, USA
| |
Collapse
|
3
|
Groome JR. Historical Perspective of the Characterization of Conotoxins Targeting Voltage-Gated Sodium Channels. Mar Drugs 2023; 21:209. [PMID: 37103349 PMCID: PMC10142487 DOI: 10.3390/md21040209] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2023] [Revised: 03/21/2023] [Accepted: 03/24/2023] [Indexed: 03/30/2023] Open
Abstract
Marine toxins have potent actions on diverse sodium ion channels regulated by transmembrane voltage (voltage-gated ion channels) or by neurotransmitters (nicotinic acetylcholine receptor channels). Studies of these toxins have focused on varied aspects of venom peptides ranging from evolutionary relationships of predator and prey, biological actions on excitable tissues, potential application as pharmacological intervention in disease therapy, and as part of multiple experimental approaches towards an understanding of the atomistic characterization of ion channel structure. This review examines the historical perspective of the study of conotoxin peptides active on sodium channels gated by transmembrane voltage, which has led to recent advances in ion channel research made possible with the exploitation of the diversity of these marine toxins.
Collapse
Affiliation(s)
- James R Groome
- Department of Biological Sciences, Idaho State University, Pocatello, ID 83209, USA
| |
Collapse
|
4
|
Pathophysiological Responses to Conotoxin Modulation of Voltage-Gated Ion Currents. Mar Drugs 2022; 20:md20050282. [PMID: 35621933 PMCID: PMC9143252 DOI: 10.3390/md20050282] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2022] [Revised: 04/04/2022] [Accepted: 04/21/2022] [Indexed: 02/04/2023] Open
Abstract
Voltage-gated ion channels are plasma membrane proteins that generate electrical signals following a change in the membrane voltage. Since they are involved in several physiological processes, their dysfunction may be responsible for a series of diseases and pain states particularly related to neuronal and muscular systems. It is well established for decades that bioactive peptides isolated from venoms of marine mollusks belonging to the Conus genus, collectively known as conotoxins, can target different types and isoforms of these channels exerting therapeutic effects and pain relief. For this reason, conotoxins are widely used for either therapeutic purposes or studies on ion channel mechanisms of action disclosure. In addition their positive property, however, conotoxins may generate pathological states through similar ion channel modulation. In this narrative review, we provide pieces of evidence on the pathophysiological impacts that different members of conotoxin families exert by targeting the three most important voltage-gated channels, such as sodium, calcium, and potassium, involved in cellular processes.
Collapse
|
5
|
Tran HNT, McMahon KL, Deuis JR, Vetter I, Schroeder CI. Structural and functional insights into the inhibition of human voltage-gated sodium channels by μ-conotoxin KIIIA disulfide isomers. J Biol Chem 2022; 298:101728. [PMID: 35167877 PMCID: PMC8927997 DOI: 10.1016/j.jbc.2022.101728] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Revised: 02/08/2022] [Accepted: 02/11/2022] [Indexed: 12/13/2022] Open
Abstract
μ-Conotoxins are components of cone snail venom, well-known for their analgesic activity through potent inhibition of voltage-gated sodium channel (NaV) subtypes, including NaV1.7. These small, disulfide-rich peptides are typically stabilized by three disulfide bonds arranged in a ‘native’ CysI-CysIV, CysII-CysV, CysIII-CysVI pattern of disulfide connectivity. However, μ-conotoxin KIIIA, the smallest and most studied μ-conotoxin with inhibitory activity at NaV1.7, forms two distinct disulfide bond isomers during thermodynamic oxidative folding, including Isomer 1 (CysI-CysV, CysII-CysIV, CysIII-CysVI) and Isomer 2 (CysI-CysVI, CysII-CysIV, CysIII-CysV), but not the native μ-conotoxin arrangement. To date, there has been no study on the structure and activity of KIIIA comprising the native μ-conotoxin disulfide bond arrangement. Here, we evaluated the synthesis, potency, sodium channel subtype selectivity, and 3D structure of the three isomers of KIIIA. Using a regioselective disulfide bond-forming strategy, we synthetically produced the three μ-conotoxin KIIIA isomers displaying distinct bioactivity and NaV subtype selectivity across human NaV channel subtypes 1.2, 1.4, and 1.7. We show that Isomer 1 inhibits NaV subtypes with a rank order of potency of NaV1.4 > 1.2 > 1.7 and Isomer 2 in the order of NaV1.4≈1.2 > 1.7, while the native isomer inhibited NaV1.4 > 1.7≈1.2. The three KIIIA isomers were further evaluated by NMR solution structure analysis and molecular docking with hNaV1.2. Our study highlights the importance of investigating alternate disulfide isomers, as disulfide connectivity affects not only the overall structure of the peptides but also the potency and subtype selectivity of μ-conotoxins targeting therapeutically relevant NaV subtypes.
Collapse
Affiliation(s)
- Hue N T Tran
- Institute for Molecular Bioscience, The University of Queensland, St Lucia, Queensland, Australia
| | - Kirsten L McMahon
- Institute for Molecular Bioscience, The University of Queensland, St Lucia, Queensland, Australia
| | - Jennifer R Deuis
- Institute for Molecular Bioscience, The University of Queensland, St Lucia, Queensland, Australia
| | - Irina Vetter
- Institute for Molecular Bioscience, The University of Queensland, St Lucia, Queensland, Australia; School of Pharmacy, The University of Queensland, Woolloongabba, Queensland, Australia.
| | - Christina I Schroeder
- Institute for Molecular Bioscience, The University of Queensland, St Lucia, Queensland, Australia; Center for Cancer Research, National Cancer Institute, National Institutes of Health, Frederick, Maryland, USA.
| |
Collapse
|
6
|
Yang M, Li Y, Liu L, Zhou M. A novel proline-rich M-superfamily conotoxin that can simultaneously affect sodium, potassium and calcium currents. J Venom Anim Toxins Incl Trop Dis 2021; 27:e20200164. [PMID: 34234819 PMCID: PMC8230863 DOI: 10.1590/1678-9199-jvatitd-2020-0164] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2020] [Accepted: 03/01/2021] [Indexed: 12/19/2022] Open
Abstract
Background Conotoxins have become a research hotspot in the neuropharmacology field for their high activity and specificity in targeting ion channels and neurotransmitter receptors. There have been reports of a conotoxin acting on two ion channels, but rare reports of a conotoxin acting on three ion channels. Methods Vr3a, a proline-rich M-superfamily conotoxin from a worm-hunting Conus varius, was obtained by solid-phase synthesis and identified by mass spectrometry. The effects of synthesized Vr3a on sodium, potassium and calcium currents were tested on rat DRG cells by patch clamp experiments. The further effects of Vr3a on human Cav1.2 and Cav2.2 currents were tested on HEK293 cells. Results About 10 μM Vr3a has no effects on the peak sodium currents, but can induce a ~10 mV shift in a polarizing direction in the current-voltage relationship. In addition, 10 μM Vr3a can increase 19.61 ± 5.12% of the peak potassium currents and do not induce a shift in the current-voltage relationship. An amount of 10 μM Vr3a can inhibit 31.26% ± 4.53% of the peak calcium currents and do not induce a shift in the current-voltage relationship. The IC50 value of Vr3a on calcium channel currents in rat DRG neurons is 19.28 ± 4.32 μM. Moreover, 10 μM Vr3a can inhibit 15.32% ± 5.41% of the human Cav1.2 currents and 12.86% ± 4.93% of the human Cav2.2 currents. Conclusions Vr3a can simultaneously affect sodium, potassium and calcium currents. This novel triple-target conotoxin Vr3a expands understanding of conotoxin functions.
Collapse
Affiliation(s)
- Manyi Yang
- Department of Hepatobiliary and Pancreatic Surgery, NHC Key Laboratory of Nanobiological Technology, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Yubin Li
- Department of Oncology, State Local Joint Engineering Laboratory for Anticancer Drugs, NHC Key Laboratory of Cancer Proteomics, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Longfei Liu
- Department of Urology, National Clinical Research Center for Geriatric Disorder, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Maojun Zhou
- Department of Oncology, State Local Joint Engineering Laboratory for Anticancer Drugs, NHC Key Laboratory of Cancer Proteomics, Xiangya Hospital, Central South University, Changsha, Hunan, China
| |
Collapse
|
7
|
Abstract
Snails can provide a considerable variety of bioactive compounds for cosmetic and pharmaceutical industries, useful for the development of new formulations with less toxicity and post effects compared to regular compounds used for the purpose. Compounds from crude extract, mucus, slime consist of glycans, polypeptides, proteins, etc., and can be used for curing diseases like viral lesions, warts, and different dermal problems. Some particular uses of snails involve treating post-traumatic stress. Micro RNA of Lymnaea stagnalis, was known to be responsible for the development of long-term memory and treatment of Alzheimer's and Dementia like diseases. This review explores the application of various bioactive compounds from snails with its potential as new translational medicinal and cosmetic applications. Snail bioactive compounds like ω-MVIIA, μ-SIIIA, μO-MrVIB, Xen2174, δ-EVIA, α-Vc1.1, σ-GVIIA, Conantokin-G, and Contulakin-G, conopeptides can be used for the development of anti-cancer drugs. These compounds target the innate immunity and improve the defense system of humans and provide protection against these life-threatening health concerns.AbbreviationsFDA: Food and Drug Administration; UTI: urinal tract infection; nAChRs: nicotinic acetylcholine receptors; NMDA: N-methyl-D-aspartate; CNS: central nervous system; CAR T: chimeric antigen receptors therapy; Micro RNA: micro ribonucleic acid.
Collapse
Affiliation(s)
- Varun Dhiman
- Department of Environmental Sciences, Central University of Himachal Pradesh, DharamshalaDharamshala, India
| | - Deepak Pant
- School of Chemical Sciences, Central University of Haryana, Mahendragarh, Haryana, India
| |
Collapse
|
8
|
Gallo A, Boni R, Tosti E. Neurobiological activity of conotoxins via sodium channel modulation. Toxicon 2020; 187:47-56. [PMID: 32877656 DOI: 10.1016/j.toxicon.2020.08.019] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2020] [Revised: 07/20/2020] [Accepted: 08/22/2020] [Indexed: 01/02/2023]
Abstract
Conotoxins (CnTX) are bioactive peptides produced by marine molluscs belonging to Conus genus. The biochemical structure of these venomous peptides is characterized by a low number of amino acids linked with disulfide bonds formed by a high degree of post-translational modifications and glycosylation steps which increase the diversity and rate of evolution of these molecules. CnTX different isoforms are known to target ion channels and, in particular, voltage-gated sodium (Na+) channels (Nav channels). These are transmembrane proteins fundamental in excitable cells for generating the depolarization of plasma membrane potential known as action potential which propagates electrical signals in muscles and nerves for physiological functions. Disorders in Nav channel activity have been shown to induce neurological pathologies and pain states. Here, we describe the current knowledge of CnTX isoform modulation of the Nav channel activity, the mechanism of action and the potential therapeutic use of these toxins in counteracting neurological dysfunctions.
Collapse
Affiliation(s)
- Alessandra Gallo
- Department of Biology and Evolution of Marine Organisms, Stazione Zoologica Anton Dohrn, Villa Comunale, 80121, Naples, Italy.
| | - Raffele Boni
- Department of Sciences, University of Basilicata, 85100, Potenza, Italy.
| | - Elisabetta Tosti
- Department of Biology and Evolution of Marine Organisms, Stazione Zoologica Anton Dohrn, Villa Comunale, 80121, Naples, Italy.
| |
Collapse
|
9
|
Kondasinghe TD, Saraha HY, Jackowski ST, Stockdill JL. Raising the Bar On-Bead: Efficient On-Resin Synthesis of α-Conotoxin LvIA. Tetrahedron Lett 2019; 60:23-28. [PMID: 31564757 PMCID: PMC6764457 DOI: 10.1016/j.tetlet.2018.11.048] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
α4/7-Conotoxin LvIA is an isoform-selective inhibitor of the α3β2 nicotinic acetylcholine receptor. An efficient strategy for the synthesis of this toxin is critical to advancing its utility as a probe for receptor function and as a potential pharmaceutical lead target. On-resin methods for peptide synthesis offer potential synthetic advantages; however, strategies for on-resin formation of multiple disulfides have historically been low-yielding. Here, we harness the reactivity of the Allocam protecting group and employ 3-amino acid spacer strategy to synthesize α4/7-conotoxin LvIA via three different on-resin strategies, each of which results in an isolated yield higher than prior fully on-resin approaches.
Collapse
Affiliation(s)
| | - Hasina Y. Saraha
- Department of Chemistry, Wayne State University, Detroit, MI 48202
| | | | | |
Collapse
|
10
|
Cardoso FC, Lewis RJ. Sodium channels and pain: from toxins to therapies. Br J Pharmacol 2018; 175:2138-2157. [PMID: 28749537 PMCID: PMC5980290 DOI: 10.1111/bph.13962] [Citation(s) in RCA: 71] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2017] [Revised: 07/11/2017] [Accepted: 07/17/2017] [Indexed: 12/16/2022] Open
Abstract
Voltage-gated sodium channels (NaV channels) are essential for the initiation and propagation of action potentials that critically influence our ability to respond to a diverse range of stimuli. Physiological and pharmacological studies have linked abnormal function of NaV channels to many human disorders, including chronic neuropathic pain. These findings, along with the description of the functional properties and expression pattern of NaV channel subtypes, are helping to uncover subtype specific roles in acute and chronic pain and revealing potential opportunities to target these with selective inhibitors. High-throughput screens and automated electrophysiology platforms have identified natural toxins as a promising group of molecules for the development of target-specific analgesics. In this review, the role of toxins in defining the contribution of NaV channels in acute and chronic pain states and their potential to be used as analgesic therapies are discussed. LINKED ARTICLES This article is part of a themed section on Recent Advances in Targeting Ion Channels to Treat Chronic Pain. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v175.12/issuetoc.
Collapse
Affiliation(s)
- Fernanda C Cardoso
- Department of Chemistry and Structural Biology, Institute for Molecular BioscienceThe University of QueenslandBrisbaneQLDAustralia
| | - Richard J Lewis
- Department of Chemistry and Structural Biology, Institute for Molecular BioscienceThe University of QueenslandBrisbaneQLDAustralia
| |
Collapse
|
11
|
Wu Y, Ma H, Zhang F, Zhang C, Zou X, Cao Z. Selective Voltage-Gated Sodium Channel Peptide Toxins from Animal Venom: Pharmacological Probes and Analgesic Drug Development. ACS Chem Neurosci 2018; 9:187-197. [PMID: 29161016 DOI: 10.1021/acschemneuro.7b00406] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Voltage-gated sodium channels (Navs) play critical roles in action potential generation and propagation. Nav channelopathy as well as pathological sensitization contribute to allodynia and hyperalgesia. Recent evidence has demonstrated the significant roles of Nav subtypes (Nav1.3, 1.7, 1.8, and 1.9) in nociceptive transduction, and therefore these Navs may represent attractive targets for analgesic drug discovery. Animal toxins are structurally diverse peptides that are highly potent yet selective on ion channel subtypes and therefore represent valuable probes to elucidate the structures, gating properties, and cellular functions of ion channels. In this review, we summarize recent advances on peptide toxins from animal venom that selectively target Nav1.3, 1.7, 1.8, and 1.9, along with their potential in analgesic drug discovery.
Collapse
Affiliation(s)
- Ying Wu
- Jiangsu Provincial Key Laboratory for TCM Evaluation
and Translational Development, China Pharmaceutical University, Nanjing 211198, China
| | - Hui Ma
- Jiangsu Provincial Key Laboratory for TCM Evaluation
and Translational Development, China Pharmaceutical University, Nanjing 211198, China
| | - Fan Zhang
- Jiangsu Provincial Key Laboratory for TCM Evaluation
and Translational Development, China Pharmaceutical University, Nanjing 211198, China
| | - Chunlei Zhang
- Jiangsu Provincial Key Laboratory for TCM Evaluation
and Translational Development, China Pharmaceutical University, Nanjing 211198, China
| | - Xiaohan Zou
- Jiangsu Provincial Key Laboratory for TCM Evaluation
and Translational Development, China Pharmaceutical University, Nanjing 211198, China
| | - Zhengyu Cao
- Jiangsu Provincial Key Laboratory for TCM Evaluation
and Translational Development, China Pharmaceutical University, Nanjing 211198, China
| |
Collapse
|
12
|
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
|
13
|
Tosti E, Boni R, Gallo A. µ-Conotoxins Modulating Sodium Currents in Pain Perception and Transmission: A Therapeutic Potential. Mar Drugs 2017; 15:E295. [PMID: 28937587 PMCID: PMC5666403 DOI: 10.3390/md15100295] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2017] [Revised: 09/12/2017] [Accepted: 09/20/2017] [Indexed: 12/27/2022] Open
Abstract
The Conus genus includes around 500 species of marine mollusks with a peculiar production of venomous peptides known as conotoxins (CTX). Each species is able to produce up to 200 different biological active peptides. Common structure of CTX is the low number of amino acids stabilized by disulfide bridges and post-translational modifications that give rise to different isoforms. µ and µO-CTX are two isoforms that specifically target voltage-gated sodium channels. These, by inducing the entrance of sodium ions in the cell, modulate the neuronal excitability by depolarizing plasma membrane and propagating the action potential. Hyperexcitability and mutations of sodium channels are responsible for perception and transmission of inflammatory and neuropathic pain states. In this review, we describe the current knowledge of µ-CTX interacting with the different sodium channels subtypes, the mechanism of action and their potential therapeutic use as analgesic compounds in the clinical management of pain conditions.
Collapse
Affiliation(s)
- Elisabetta Tosti
- Department of Biology and Evolution of Marine Organisms, Stazione Zoologica Anton Dohrn, Villa Comunale, 80121 Naples, Italy.
| | - Raffaele Boni
- Department of Sciences, University of Basilicata, 75100 Potenza, Italy.
| | - Alessandra Gallo
- Department of Biology and Evolution of Marine Organisms, Stazione Zoologica Anton Dohrn, Villa Comunale, 80121 Naples, Italy.
| |
Collapse
|
14
|
A novel μ-conotoxin from worm-hunting Conus tessulatus that selectively inhibit rat TTX-resistant sodium currents. Toxicon 2017; 130:11-18. [DOI: 10.1016/j.toxicon.2017.02.013] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2016] [Revised: 02/01/2017] [Accepted: 02/16/2017] [Indexed: 12/13/2022]
|
15
|
Wu X, Huang Y, Kaas Q, Craik DJ. Cyclisation of Disulfide‐Rich Conotoxins in Drug Design Applications. European J Org Chem 2016. [DOI: 10.1002/ejoc.201600402] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Affiliation(s)
- Xiaosa Wu
- Institute for Molecular BioscienceThe University of Queensland306 Carmody Road (Building 80)4072BrisbaneAustralia
| | - Yen‐Hua Huang
- Institute for Molecular BioscienceThe University of Queensland306 Carmody Road (Building 80)4072BrisbaneAustralia
| | - Quentin Kaas
- Institute for Molecular BioscienceThe University of Queensland306 Carmody Road (Building 80)4072BrisbaneAustralia
| | - David J. Craik
- Institute for Molecular BioscienceThe University of Queensland306 Carmody Road (Building 80)4072BrisbaneAustralia
| |
Collapse
|
16
|
de Lera Ruiz M, Kraus RL. Voltage-Gated Sodium Channels: Structure, Function, Pharmacology, and Clinical Indications. J Med Chem 2015; 58:7093-118. [PMID: 25927480 DOI: 10.1021/jm501981g] [Citation(s) in RCA: 367] [Impact Index Per Article: 36.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The tremendous therapeutic potential of voltage-gated sodium channels (Na(v)s) has been the subject of many studies in the past and is of intense interest today. Na(v)1.7 channels in particular have received much attention recently because of strong genetic validation of their involvement in nociception. Here we summarize the current status of research in the Na(v) field and present the most relevant recent developments with respect to the molecular structure, general physiology, and pharmacology of distinct Na(v) channel subtypes. We discuss Na(v) channel ligands such as small molecules, toxins isolated from animal venoms, and the recently identified Na(v)1.7-selective antibody. Furthermore, we review eight characterized ligand binding sites on the Na(v) channel α subunit. Finally, we examine possible therapeutic applications of Na(v) ligands and provide an update on current clinical studies.
Collapse
Affiliation(s)
- Manuel de Lera Ruiz
- Merck Research Laboratories , 770 Sumneytown Pike, West Point, Pennsylvania 19486, United States
| | - Richard L Kraus
- Merck Research Laboratories , 770 Sumneytown Pike, West Point, Pennsylvania 19486, United States
| |
Collapse
|
17
|
Heimer P, Tietze AA, Böhm M, Giernoth R, Kuchenbuch A, Stark A, Leipold E, Heinemann SH, Kandt C, Imhof D. Application of Room-Temperature Aprotic and Protic Ionic Liquids for Oxidative Folding of Cysteine-Rich Peptides. Chembiochem 2014; 15:2754-65. [DOI: 10.1002/cbic.201402356] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2014] [Indexed: 11/05/2022]
|
18
|
Akondi KB, Muttenthaler M, Dutertre S, Kaas Q, Craik DJ, Lewis RJ, Alewood PF. Discovery, synthesis, and structure-activity relationships of conotoxins. Chem Rev 2014; 114:5815-47. [PMID: 24720541 PMCID: PMC7610532 DOI: 10.1021/cr400401e] [Citation(s) in RCA: 258] [Impact Index Per Article: 23.5] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Affiliation(s)
| | | | - Sébastien Dutertre
- Institute for Molecular Bioscience, The University of Queensland, Brisbane QLD 4072, Australia
| | - Quentin Kaas
- Institute for Molecular Bioscience, The University of Queensland, Brisbane QLD 4072, Australia
| | - David J Craik
- Institute for Molecular Bioscience, The University of Queensland, Brisbane QLD 4072, Australia
| | - Richard J Lewis
- Institute for Molecular Bioscience, The University of Queensland, Brisbane QLD 4072, Australia
| | - Paul F Alewood
- Institute for Molecular Bioscience, The University of Queensland, Brisbane QLD 4072, Australia
| |
Collapse
|
19
|
Li J, Yan Y, Yu H, Peng X, Zhang Y, Hu W, Duan Z, Wang X, Liang S. Isolation and identification of a sodium channel-inhibiting protein from eggs of black widow spiders. Int J Biol Macromol 2014; 65:115-120. [PMID: 24412150 DOI: 10.1016/j.ijbiomac.2014.01.004] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2013] [Revised: 01/01/2014] [Accepted: 01/02/2014] [Indexed: 11/30/2022]
Abstract
The eggs of black widow spider (L. tredecimguttatus) have been demonstrated to be rich in biologically active components that exhibit great research value and application foreground. In the present study, a protein toxin, named Latroeggtoxin-II, was isolated from the eggs using the combination of gel filtration, ion exchange chromatography and reversed-phase high performance liquid chromatography. Electrospray mass spectrometric analysis indicated that the molecular weight of the protein was 28.69 kDa, and Edman degradation revealed that its N-terminal sequence was ESIQT STYVP NTPNQ KFDYE VGKDY-. After being abdominally injected into mice and P. americana, the protein could make the animals especially P. americana display a series of poisoning symptoms. Electrophysiological experiments demonstrated that the protein could selectively inhibit tetrodotoxin-resistant Na(+) channel currents in rat dorsal root ganglion neurons, without significant effect on the tetrodotoxin-sensitive Na(+) channel currents. Using multiple proteomic strategies, the purified protein was shown to have only a few similarities to the existing proteins in the databases, suggesting that it was a novel protein isolated from the eggs of black widow spiders.
Collapse
Affiliation(s)
- Jianjun Li
- Key Laboratory of Protein Chemistry and Developmental Biology of Ministry of Education, College of Life Sciences, Hunan Normal University, Changsha 410081, PR China
| | - Yizhong Yan
- Key Laboratory of Protein Chemistry and Developmental Biology of Ministry of Education, College of Life Sciences, Hunan Normal University, Changsha 410081, PR China
| | - Hai Yu
- Key Laboratory of Protein Chemistry and Developmental Biology of Ministry of Education, College of Life Sciences, Hunan Normal University, Changsha 410081, PR China
| | - Xiaozhen Peng
- Key Laboratory of Protein Chemistry and Developmental Biology of Ministry of Education, College of Life Sciences, Hunan Normal University, Changsha 410081, PR China
| | - Yiya Zhang
- Key Laboratory of Protein Chemistry and Developmental Biology of Ministry of Education, College of Life Sciences, Hunan Normal University, Changsha 410081, PR China
| | - Weijun Hu
- Key Laboratory of Protein Chemistry and Developmental Biology of Ministry of Education, College of Life Sciences, Hunan Normal University, Changsha 410081, PR China
| | - Zhigui Duan
- Key Laboratory of Protein Chemistry and Developmental Biology of Ministry of Education, College of Life Sciences, Hunan Normal University, Changsha 410081, PR China
| | - Xianchun Wang
- Key Laboratory of Protein Chemistry and Developmental Biology of Ministry of Education, College of Life Sciences, Hunan Normal University, Changsha 410081, PR China.
| | - Songping Liang
- Key Laboratory of Protein Chemistry and Developmental Biology of Ministry of Education, College of Life Sciences, Hunan Normal University, Changsha 410081, PR China.
| |
Collapse
|
20
|
Akondi KB, Lewis RJ, Alewood PF. Re-engineering the μ-conotoxin SIIIA scaffold. Biopolymers 2014; 101:347-54. [DOI: 10.1002/bip.22368] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2013] [Revised: 07/21/2013] [Accepted: 07/26/2013] [Indexed: 12/19/2022]
Affiliation(s)
- K. B. Akondi
- Institute for Molecular Bioscience (IMB); The University of Queensland; Brisbane 4072 Queensland Australia
| | - R. J. Lewis
- Institute for Molecular Bioscience (IMB); The University of Queensland; Brisbane 4072 Queensland Australia
| | - P. F. Alewood
- Institute for Molecular Bioscience (IMB); The University of Queensland; Brisbane 4072 Queensland Australia
| |
Collapse
|
21
|
Fedosov AÉ, Moshkovskiĭ SA, Kuznetsova KG, Olivera BM. [Conotoxins: from the biodiversity of gastropods to new drugs]. BIOMEDIT︠S︡INSKAI︠A︡ KHIMII︠A︡ 2013; 59:267-94. [PMID: 23987066 DOI: 10.18097/pbmc20135903267] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
A review describes general trends in research of conotoxins that are peptide toxins isolated from sea gastropods of the Conus genus, since the toxins were discovered in 1970th. There are disclosed a conotoxin classification, their structure diversity and different ways of action to their molecular targets, mainly, ion channels. In the applied aspect of conotoxin research, drug discovery and development is discussed, the drugs being based on conotoxin structure. A first exemplary drug is a ziconotide, which is an analgesic of new generation.
Collapse
|
22
|
Strategies for the development of conotoxins as new therapeutic leads. Mar Drugs 2013; 11:2293-313. [PMID: 23812174 PMCID: PMC3736424 DOI: 10.3390/md11072293] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2013] [Revised: 05/27/2013] [Accepted: 06/06/2013] [Indexed: 11/24/2022] Open
Abstract
Peptide toxins typically bind to their target ion channels or receptors with high potency and selectivity, making them attractive leads for therapeutic development. In some cases the native peptide as it is found in the venom from which it originates can be used directly, but in many instances it is desirable to truncate and/or stabilize the peptide to improve its therapeutic properties. A complementary strategy is to display the key residues that make up the pharmacophore of the peptide toxin on a non-peptidic scaffold, thereby creating a peptidomimetic. This review exemplifies these approaches with peptide toxins from marine organisms, with a particular focus on conotoxins.
Collapse
|
23
|
Gilchrist J, Bosmans F. Animal toxins can alter the function of Nav1.8 and Nav1.9. Toxins (Basel) 2012; 4:620-32. [PMID: 23012651 PMCID: PMC3446747 DOI: 10.3390/toxins4080620] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2012] [Revised: 07/24/2012] [Accepted: 07/27/2012] [Indexed: 12/19/2022] Open
Abstract
Human voltage-activated sodium (Nav) channels are adept at rapidly transmitting electrical signals across long distances in various excitable tissues. As such, they are amongst the most widely targeted ion channels by drugs and animal toxins. Of the nine isoforms, Nav1.8 and Nav1.9 are preferentially expressed in DRG neurons where they are thought to play an important role in pain signaling. Although the functional properties of Nav1.8 have been relatively well characterized, difficulties with expressing Nav1.9 in established heterologous systems limit our understanding of the gating properties and toxin pharmacology of this particular isoform. This review summarizes our current knowledge of the role of Nav1.8 and Nav1.9 in pain perception and elaborates on the approaches used to identify molecules capable of influencing their function.
Collapse
Affiliation(s)
- John Gilchrist
- Department of Physiology, School of Medicine, Johns Hopkins University, Baltimore, MD 21205, USA;
| | - Frank Bosmans
- Department of Physiology, School of Medicine, Johns Hopkins University, Baltimore, MD 21205, USA;
- Solomon H. Snyder Department of Neuroscience, School of Medicine, Johns Hopkins University, Baltimore, MD 21205, USA
- Author to whom correspondence should be addressed; ; Tel.: +1-410-955-4428
| |
Collapse
|
24
|
Li P, Zhu S. Mutational analysis of the analgesic peptide DrTx(1-42) revealing a functional role of the amino-terminal turn. PLoS One 2012; 7:e31830. [PMID: 22355398 PMCID: PMC3280213 DOI: 10.1371/journal.pone.0031830] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2011] [Accepted: 01/13/2012] [Indexed: 12/19/2022] Open
Abstract
Background DrTx(1-42) (a carboxyl-terminally truncated version of drosotoxin) is a potent and selective blocker of tetrodotoxin-resistant (TTX-R) Na+ channels in rat dorsal root ganglion neurons with analgesic activity. This purpose is to identify key amino acids which are responsible for both blocking and analgesic effects of DrTx(1-42). Methods On the basis of previous study, we designed five mutants of DrTx(1-42) (delN, D8A, D8K, G9A, and G9R) in the amino-terminal turn (N-turn) region, a proposed functional region located in the amino-terminus of the molecule. All these mutants were expressed in E.coli and purified by RP-HPLC. Electrophysiological properties of these analogues were examined by whole-cell patch-clamp recordings and their antinociceptive effects were investigated by the formalin test and acetic acid induced writhing test. Results All the mutants except for G9A possess a similar secondary structure to that of DrTx(1-42), as identified by circular dichroism analysis. Three mutants (delN, D8A and G9A) were found almost inactive to TTX-R Na+ channels, whereas D8K retains similar activity and G9R showed decreased potency when compared with the wild-type molecule. Consistent with the electrophysiological observations, D8K and G9R exhibited antinociceptive effects in the second phase (inflammatory pain) of the formalin test and the acetic acid induced writhing test, while delN, D8A and G9A lack such effects. Conclusions Our results show that the N-turn is closely related to function of DrTx(1-42). The mutant (D8A) as a control peptide further reveals that a charged residue at site 8 of the N-terminus is important for channel blockade and analgesic activity. This study indicates that blocking of voltage-gated TTX-R Na+ channel in DRG neurons contributes to analgesic effect in rat inflammatory pain. Structural and functional data described here offers support for the development of novel analgesic drugs through targeting TTX-R Na+ channels.
Collapse
Affiliation(s)
- Ping Li
- State Key Laboratory of Integrated Management of Pest Insects and Rodents, Institute of Zoology, Chinese Academy of Sciences, Beijing, People's Republic of China
| | - Shunyi Zhu
- State Key Laboratory of Integrated Management of Pest Insects and Rodents, Institute of Zoology, Chinese Academy of Sciences, Beijing, People's Republic of China
- * E-mail:
| |
Collapse
|
25
|
Steiner AM, Bulaj G. Optimization of oxidative folding methods for cysteine-rich peptides: a study of conotoxins containing three disulfide bridges. J Pept Sci 2011; 17:1-7. [PMID: 20814907 DOI: 10.1002/psc.1283] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
The oxidative folding of small, cysteine-rich peptides to selectively achieve the native disulfide bond connectivities is critical for discovery and structure-function studies of many bioactive peptides. As the propensity to acquire the native conformation greatly depends on the peptide sequence, numerous empirical oxidation methods are employed. The context-dependent optimization of these methods has thus far precluded a generalized oxidative folding protocol, in particular for peptides containing more than two disulfides. Herein, we compare the efficacy of optimized solution-phase and polymer-supported oxidation methods using three disulfide-bridged conotoxins, namely µ-SIIIA, µ-KIIIA and ω-GVIA. The use of diselenide bridges as proxies for disulfide bridges is also evaluated. We propose the ClearOx-assisted oxidation of selenopeptides as a fairly generalized oxidative folding protocol.
Collapse
Affiliation(s)
- Andrew M Steiner
- Department of Medicinal Chemistry, University of Utah, Salt Lake City, UT 84108, USA
| | | |
Collapse
|
26
|
Leipold E, Markgraf R, Miloslavina A, Kijas M, Schirmeyer J, Imhof D, Heinemann SH. Molecular determinants for the subtype specificity of μ-conotoxin SIIIA targeting neuronal voltage-gated sodium channels. Neuropharmacology 2011; 61:105-11. [PMID: 21419143 DOI: 10.1016/j.neuropharm.2011.03.008] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2011] [Accepted: 03/09/2011] [Indexed: 11/29/2022]
Abstract
Voltage-gated sodium channels (Na(V) channels) play a pivotal role in neuronal excitability; they are specifically targeted by μ-conotoxins from the venom of marine cone snails. These peptide toxins bind to the outer vestibule of the channel pore thereby blocking ion conduction through Na(V) channels. μ-Conotoxin SIIIA from Conus striatus was shown to be a potent inhibitor of neuronal sodium channels and to display analgesic effects in mice, albeit the molecular targets are not unambiguously known. We therefore studied recombinant Na(V) channels expressed in mammalian cells using the whole-cell patch-clamp method. Synthetic μSIIIA slowly and partially blocked rat Na(V)1.4 channels with an apparent IC(50) of 0.56 ± 0.29 μM; the block was not complete, leaving at high concentration a residual current component of about 10% with a correspondingly reduced single-channel conductance. At 10 μM, μSIIIA potently blocked rat Na(V)1.2, rat and human Na(V)1.4, and mouse Na(V)1.6 channels; human Na(V)1.7 channels were only inhibited by 58.1 ± 4.9%, whereas human Na(V)1.5 as well as rat and human Na(V)1.8 were insensitive. Employing domain chimeras between rNa(V)1.4 and hNa(V)1.5, we located the determinants for μSIIIA specificity in the first half of the channel protein with a major contribution of domain-2 and a minor contribution of domain-1. The latter was largely accounted for by the alteration in the TTX-binding site (Tyr401 in rNa(V)1.4, Cys for Na(V)1.5, and Ser for Na(V)1.8). Introduction of domain-2 pore loops of all tested channel isoforms into rNa(V)1.4 conferred the μSIIIA phenotype of the respective donor channels highlighting the importance of the domain-2 pore loop as the major determinant for μSIIIA's subtype specificity. Single-site substitutions identified residue Ala728 in rNa(V)1.4 as crucial for its high sensitivity toward μSIIIA. Likewise, Asn889 at the homologous position in hNa(V)1.7 is responsible for the channel's reduced μSIIIA sensitivity. These results will pave the way for the rational design of selective Na(V)-channel antagonists for research and medical applications.
Collapse
Affiliation(s)
- Enrico Leipold
- Center for Molecular Biomedicine, Department of Biophysics, Friedrich Schiller University of Jena & University Hospital Jena, Hans-Knoell-Str. 2, D-07745 Jena, Germany
| | | | | | | | | | | | | |
Collapse
|
27
|
Zhu S, Luo L, Li P, Gao B, Zhu L, Yuan Y. DrTx(1–42), a C-terminally truncated analogue of drosotoxin, is a candidate of analgesic drugs. Biochem Pharmacol 2011; 81:425-31. [PMID: 21040712 DOI: 10.1016/j.bcp.2010.10.015] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2010] [Revised: 10/24/2010] [Accepted: 10/25/2010] [Indexed: 12/19/2022]
|
28
|
Isoform-selective voltage-gated Na+ channel modulators as next-generation analgesics. Future Med Chem 2010; 2:775-90. [DOI: 10.4155/fmc.10.26] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
For many patients the current therapies for controlling chronic pain are inadequate. This has driven the search for analgesics with improved efficacy and side effect profiles. Some anticonvulsants have voltage-gated Na+ channels (VGSCs) as their molecular targets and are used to treat pain, but the efficacy seen is marginal and the drugs are generally poorly tolerated. The clinically used VGSC-modulating analgesics show no isoform selectivity, which probably limits their use. Thus, focus has fallen on VGSCs expressed selectively by primary afferent neurons and the search for isoform-selective drugs. In this review, we describe developments in our understanding of the biology of VGSCs, screening technologies and the pharmacological properties of VGSC modulators with promise as analgesics. Also highlighted are the challenges associated with targeting isoform-selective VGSCs.
Collapse
|
29
|
Norton RS. Mu-conotoxins as leads in the development of new analgesics. Molecules 2010; 15:2825-44. [PMID: 20428082 PMCID: PMC6257286 DOI: 10.3390/molecules15042825] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2010] [Revised: 04/06/2010] [Accepted: 04/12/2010] [Indexed: 02/02/2023] Open
Abstract
Voltage-gated sodium channels (VGSCs) contain a specific binding site for a family of cone shell toxins known as mu-conotoxins. As some VGSCs are involved in pain perception and mu-conotoxins are able to block these channels, mu-conotoxins show considerable potential as analgesics. Recent studies have advanced our understanding of the three-dimensional structures and structure-function relationships of the mu-conotoxins, including their interaction with VGSCs. Truncated peptide analogues of the native toxins have been created in which secondary structure elements are stabilized by non-native linkers such as lactam bridges. Ultimately, it would be desirable to capture the favourable analgesic properties of the native toxins, in particular their potency and channel sub-type selectivity, in non-peptide mimetics. Such mimetics would constitute lead compounds in the development of new therapeutics for the treatment of pain.
Collapse
Affiliation(s)
- Raymond S Norton
- Walter and Eliza Hall Institute of Medical Research, Victoria, Australia.
| |
Collapse
|
30
|
Abstract
Drugs inhibiting voltage-gated sodium channels have long been used as analgesics, beginning with the use of local anaesthetics for sensory blockade and then with the discovery that Nav-blocking anticonvulsants also have benefit for pain therapy. These drugs were discovered without knowledge of their molecular target, using traditional pharmacological methods, and their clinical utility is limited by relatively narrow therapeutic windows. Until recently, attempts to develop improved inhibitors using modern molecular-targeted screening approaches have met with limited success. However, in the last few years there has been renewed activity following the discovery of human Nav1.7 mutations that cause striking insensitivity to pain. Together with recent advances in the technologies required to prosecute ion channels as drug targets, this has led to significant progress being made. This article reviews these developments and summarises current findings with these emerging new Nav inhibitors, highlighting some of the unanswered questions and the challenges that remain before they can be developed for clinical use.
Collapse
Affiliation(s)
- Jeffrey J Clare
- Cell-Based Assays Group, Millipore Corporation, St Charles, Missouri 63304, USA.
| |
Collapse
|
31
|
Novel strategies for the treatment of inflammatory hyperalgesia. Eur J Clin Pharmacol 2010; 66:429-44. [DOI: 10.1007/s00228-010-0784-7] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2009] [Accepted: 01/11/2010] [Indexed: 12/24/2022]
|
32
|
Jacob RB, McDougal OM. The M-superfamily of conotoxins: a review. Cell Mol Life Sci 2010; 67:17-27. [PMID: 19705062 PMCID: PMC3741454 DOI: 10.1007/s00018-009-0125-0] [Citation(s) in RCA: 60] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2009] [Revised: 07/30/2009] [Accepted: 08/03/2009] [Indexed: 12/19/2022]
Abstract
The focus of this review is the M-superfamily of Conus venom peptides. Disulfide rich peptides belonging to the M-superfamily have three loop regions and the cysteine arrangement: CC-C-C-CC, where the dashes represent loops one, two, and three, respectively. Characterization of M-superfamily peptides has demonstrated that diversity in cystine connectivity occurs between different branches of peptides even though the cysteine pattern remains consistent. This superfamily is subdivided into five branches, M-1 through M-5, based on the number of residues in the third loop region, between the fourth and fifth cysteine residues. M-superfamily peptides appear to be ubiquitous in Conus venom. They are largely unexplained in indigenous biological function, and they represent an active area of research within the scientific community.
Collapse
Affiliation(s)
- Reed B. Jacob
- Department of Chemistry and Biochemistry, Boise State University, 1910 University Drive, Boise, ID 83725-1520 USA
| | - Owen M. McDougal
- Department of Chemistry and Biochemistry, Boise State University, 1910 University Drive, Boise, ID 83725-1520 USA
| |
Collapse
|
33
|
Khoo KK, Feng ZP, Smith BJ, Zhang MM, Yoshikami D, Olivera BM, Bulaj G, Norton RS. Structure of the analgesic mu-conotoxin KIIIA and effects on the structure and function of disulfide deletion. Biochemistry 2009; 48:1210-9. [PMID: 19170536 DOI: 10.1021/bi801998a] [Citation(s) in RCA: 67] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Mu-conotoxin mu-KIIIA, from Conus kinoshitai, blocks mammalian neuronal voltage-gated sodium channels (VGSCs) and is a potent analgesic following systemic administration in mice. We have determined its solution structure using NMR spectroscopy. Key residues identified previously as being important for activity against VGSCs (Lys7, Trp8, Arg10, Asp11, His12, and Arg14) all reside on an alpha-helix with the exception of Arg14. To further probe structure-activity relationships of this toxin against VGSC subtypes, we have characterized the analogue mu-KIIIA[C1A,C9A], in which the Cys residues involved in one of the three disulfides in mu-KIIIA were replaced with Ala. Its structure is quite similar to that of mu-KIIIA, indicating that the Cys1-Cys9 disulfide bond could be removed without any significant distortion of the alpha-helix bearing the key residues. Consistent with this, mu-KIIIA[C1A,C9A] retained activity against VGSCs, with its rank order of potency being essentially the same as that of mu-KIIIA, namely, Na(V)1.2 > Na(V)1.4 > Na(V)1.7 >or= Na(V)1.1 > Na(V)1.3 > Na(V)1.5. Kinetics of block were obtained for Na(V)1.2, Na(V)1.4, and Na(V)1.7, and in each case, both k(on) and k(off) values of mu-KIIIA[C1A,C9A] were larger than those of mu-KIIIA. Our results show that the key residues for VGSC binding lie mostly on an alpha-helix and that the first disulfide bond can be removed without significantly affecting the structure of this helix, although the modification accelerates the on and off rates of the peptide against all tested VGSC subtypes. These findings lay the groundwork for the design of minimized peptides and helical mimetics as novel analgesics.
Collapse
Affiliation(s)
- Keith K Khoo
- The Walter and Eliza Hall Institute of Medical Research, 1G Royal Parade, Parkville, Victoria 3052, Australia
| | | | | | | | | | | | | | | |
Collapse
|
34
|
Docherty RJ, Farmer CE. The pharmacology of voltage-gated sodium channels in sensory neurones. Handb Exp Pharmacol 2009:519-61. [PMID: 19655117 DOI: 10.1007/978-3-540-79090-7_15] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Voltage-gated sodium channels (VGSCs) are vital for the normal functioning of most excitable cells. At least nine distinct functional subtypes of VGSCs are recognized, corresponding to nine genes for their pore-forming alpha-subunits. These have different developmental expression patterns, different tissue distributions in the adult and are differentially regulated at the cellular level by receptor-coupled cell signalling systems. Unsurprisingly, VGSC blockers are found to be useful as drugs in diverse clinical applications where excessive excitability of tissue leads to pathological dysfunction, e.g. epilepsy or cardiac tachyarrhythmias. The effects of most clinically useful VGSC blockers are use-dependent, i.e. their efficacy depends on channel activity. In addition, many natural toxins have been discovered that interact with VGSCs in complex ways and they have been used as experimental probes to study the structure and function of the channels and to better understand how drugs interact with the channels. Here we have attempted to summarize the properties of VGSCs in sensory neurones, discuss how they are regulated by cell signalling systems and we have considered briefly current concepts of their physiological function. We discuss in detail how drugs and toxins interact with archetypal VGSCs and where possible consider how they act on VGSCs in peripheral sensory neurones. Increasingly, drugs that block VGSCs are being used as systemic analgesic agents in chronic pain syndromes, but the full potential for VGSC blockers in this indication is yet to be realized and other applications in sensory dysfunction are also possible. Drugs targeting VGSC subtypes in sensory neurones are likely to provide novel systemic analgesics that are tissue-specific and perhaps even disease-specific, providing much-needed novel therapeutic approaches for the relief of chronic pain.
Collapse
Affiliation(s)
- Reginald J Docherty
- Neurorestoration Group, Wolfson CARD, King's College London, London SE1 9RT, UK.
| | | |
Collapse
|
35
|
Schroeder CI, Ekberg J, Nielsen KJ, Adams D, Loughnan ML, Thomas L, Adams DJ, Alewood PF, Lewis RJ. Neuronally Selective μ-Conotoxins from Conus striatus Utilize an α-Helical Motif to Target Mammalian Sodium Channels. J Biol Chem 2008; 283:21621-8. [DOI: 10.1074/jbc.m802852200] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
|
36
|
Yuan C, Liao Z, Zeng X, Dai L, Kuang F, Liang S. Jingzhaotoxin-XII, a gating modifier specific for Kv4.1 channels. Toxicon 2007; 50:646-52. [PMID: 17631373 DOI: 10.1016/j.toxicon.2007.05.009] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2007] [Revised: 05/15/2007] [Accepted: 05/29/2007] [Indexed: 01/17/2023]
Abstract
Jingzhaotoxin-XII (JZTX-XII), a 29-residue polypeptide, was purified from the venom of the Chinese tarantula Chilobrachys jingzhao. Electrophysiological recordings carried out in Xenopus laevis oocytes showed that JZTX-XII is specific for Kv4.1 channels, with the IC50 value of 0.363 microM. It interacts with the channels by modifying the gating behavior. JZTX-XII shares 80% sequence identity with phrixotoxin1, a potent inhibitor for Kv4.2 and Kv4.3 channels. Structure analysis indicates that the difference of the charge distribution in the interactive surface perhaps influences the specific pharmacology of the toxins. JZTX-XII should be a valuable tool for the investigation of the Kv4.1 channels.
Collapse
Affiliation(s)
- Chunhua Yuan
- Key Laboratory of Protein Chemistry and Developmental Biology of the Ministry of Education, Life Science College, Hunan Normal University, Changsha 410081, PR China
| | | | | | | | | | | |
Collapse
|
37
|
Zhang MM, Green BR, Catlin P, Fiedler B, Azam L, Chadwick A, Terlau H, McArthur JR, French RJ, Gulyas J, Rivier JE, Smith BJ, Norton RS, Olivera BM, Yoshikami D, Bulaj G. Structure/Function Characterization of μ-Conotoxin KIIIA, an Analgesic, Nearly Irreversible Blocker of Mammalian Neuronal Sodium Channels. J Biol Chem 2007; 282:30699-706. [PMID: 17724025 DOI: 10.1074/jbc.m704616200] [Citation(s) in RCA: 126] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Peptide neurotoxins from cone snails continue to supply compounds with therapeutic potential. Although several analgesic conotoxins have already reached human clinical trials, a continuing need exists for the discovery and development of novel non-opioid analgesics, such as subtype-selective sodium channel blockers. Micro-conotoxin KIIIA is representative of micro-conopeptides previously characterized as inhibitors of tetrodotoxin (TTX)-resistant sodium channels in amphibian dorsal root ganglion neurons. Here, we show that KIIIA has potent analgesic activity in the mouse pain model. Surprisingly, KIIIA was found to block most (>80%) of the TTX-sensitive, but only approximately 20% of the TTX-resistant, sodium current in mouse dorsal root ganglion neurons. KIIIA was tested on cloned mammalian channels expressed in Xenopus oocytes. Both Na(V)1.2 and Na(V)1.6 were strongly blocked; within experimental wash times of 40-60 min, block was reversed very little for Na(V)1.2 and only partially for Na(V)1.6. Other isoforms were blocked reversibly: Na(V)1.3 (IC50 8 microM), Na(V)1.5 (IC50 284 microM), and Na(V)1.4 (IC50 80 nM). "Alanine-walk" and related analogs were synthesized and tested against both Na(V)1.2 and Na(V)1.4; replacement of Trp-8 resulted in reversible block of Na(V)1.2, whereas replacement of Lys-7, Trp-8, or Asp-11 yielded a more profound effect on the block of Na(V)1.4 than of Na(V)1.2. Taken together, these data suggest that KIIIA is an effective tool to study structure and function of Na(V)1.2 and that further engineering of micro-conopeptides belonging to the KIIIA group may provide subtype-selective pharmacological compounds for mammalian neuronal sodium channels and potential therapeutics for the treatment of pain.
Collapse
Affiliation(s)
- Min-Min Zhang
- Department of Biology, University of Utah, Salt Lake City, Utah 84112, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Zeilhofer HU, Brune K. Analgesic strategies beyond the inhibition of cyclooxygenases. Trends Pharmacol Sci 2006; 27:467-74. [PMID: 16876882 DOI: 10.1016/j.tips.2006.07.007] [Citation(s) in RCA: 60] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2006] [Revised: 06/19/2006] [Accepted: 07/17/2006] [Indexed: 12/20/2022]
Abstract
Blocking the formation of prostaglandins with cyclooxygenase (COX) inhibitors has been the treatment of choice for inflammatory pain for more than a century. Although these agents provide profound pain relief, their long-term use is hampered by severe side-effects, mainly ulceration of the upper gastrointestinal tract. The development of COX-2-selective inhibitors ("coxibs") has significantly reduced gastrointestinal toxicity, but evidence from controlled clinical trials and experimental studies indicates that the use of coxibs has a significant cardiovascular risk. Recently, signalling elements downstream of COX-2 inhibition have been identified, which offer a great diversity of possible targets. This review focuses on prostaglandin E synthases, prostaglandin receptors and downstream effectors of prostaglandins in the PNS and CNS, including transient receptor potential channels, tetrodotoxin-resistant Na(+) channels and inhibitory glycine receptors. These novel targets should enable inflammatory pain to be treated with improved specificity and, possibly, fewer side-effects.
Collapse
Affiliation(s)
- Hanns Ulrich Zeilhofer
- Institute of Pharmacology and Toxicology, University of Zürich, and Institute of Pharmaceutical Sciences, ETH Zürich, Winterthurerstrasse 190, CH-8057 Zürich, Switzerland.
| | | |
Collapse
|
39
|
Abstract
In the four decades since toxinologists in Australia and elsewhere started to investigate the active constituents of venomous cone snails, a wealth of information has emerged on the various classes of peptides and proteins that make their venoms such potent bioactive cocktails. This article provides an overview of the current state of knowledge of these venom constituents, several of which are of interest as potential human therapeutics as a consequence of their high potency and exquisite target specificity. With the promise of as many as 50,000 venom components across the entire Conus genus, many more interesting peptides can be anticipated.
Collapse
Affiliation(s)
- Raymond S Norton
- Walter and Eliza Hall Institute of Medical Research, 1G Royal Parade, Parkville 3050, Victoria, Australia.
| | | |
Collapse
|