1
|
Tang H, Wang Q, Yang M, Jia R, Yuan J, Wang R. Development of sensitive immunoassay for identification and detection of μ-KIIIA-CTX: Insights into antibody discovery, molecular recognition, and immunoassay. Int J Biol Macromol 2025; 310:143346. [PMID: 40254203 DOI: 10.1016/j.ijbiomac.2025.143346] [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: 02/27/2025] [Revised: 03/30/2025] [Accepted: 04/17/2025] [Indexed: 04/22/2025]
Abstract
μ-KIIIA-Conotoxin (KIIIA) is a short, toxic peptide that selectively targets voltage-gated sodium channels, and has enormous potential in analgesic-drugs development and neuroscience research. However, no correlated immunoassays have been reported for identification and detection of KIIIA. Herein, a hybridoma 3E11 that specifically targets KIIIA was screened using hybridoma technology after animal immunization. The subtype of monoclonal antibody(mAb) 3E11 was IgG1, and it exhibited a high affinity constant (Kaff) of 5.838 × 108 L/mol. Meanwhile, the 3D structure of variable regions of mAb 3E11 was modeled, and the detailed molecular recognition mechanisms of mAb 3E11 to KIIIA were further investigated by molecular docking, alanine scanning and disulfide bond quenching. The sequence "KWCRDH" of KIIIA has been identified as the crucial and structural dependent epitope region recognized by mAb 3E11. The principal forces maintaining the interaction are hydrogen bonding, π-π stacking, nonpolar interactions and salt bridges. Consequently, mAb 3E11 exhibited different binding affinities towards epitope-similar antigens from μ-conotoxin family, including SIIIA, CIIIA, CnIIIA, MIIIA and SmIIIA. Among these μ-conotoxins, the binding affinity of mAb 3E11 to SIIIA is nearly equivalent to that observed with KIIIA. Ultimately, an indirect competitive ELISA(ic-ELISA) was developed based on mAb 3E11, and the linear range of ic-ELISA was 0.72 to 33.02 ng/mL with a lower detection limit (LOD) of 0.28 ng/mL. The recovery rates of intra-assays and inter-assays in spiked samples were 101.32 % and 102.47 %, respectively. The developed ic-ELISA demonstrated high accuracy and repeatability, indicating its potential for detecting the content of KIIIA in real samples.
Collapse
Affiliation(s)
- Hengkun Tang
- Guangxi Key Laboratory of Translational Medicine for Treating High-Incidence Infectious Diseases with Integrative Medicine, Guangxi University of Chinese Medicine, Nanning, China; Key Laboratory of Pathogenic Fungi and Mycotoxins of Fujian Province, School of Life Sciences, Fujian Agriculture and Forestry University, Fuzhou 350002, China
| | - Qing Wang
- Key Laboratory of Pathogenic Fungi and Mycotoxins of Fujian Province, School of Life Sciences, Fujian Agriculture and Forestry University, Fuzhou 350002, China
| | - Minyi Yang
- Guangxi Key Laboratory of Translational Medicine for Treating High-Incidence Infectious Diseases with Integrative Medicine, Guangxi University of Chinese Medicine, Nanning, China
| | - Rongye Jia
- Guangxi Key Laboratory of Translational Medicine for Treating High-Incidence Infectious Diseases with Integrative Medicine, Guangxi University of Chinese Medicine, Nanning, China
| | - Jun Yuan
- Guangxi Key Laboratory of Translational Medicine for Treating High-Incidence Infectious Diseases with Integrative Medicine, Guangxi University of Chinese Medicine, Nanning, China
| | - Rongzhi Wang
- Guangxi Key Laboratory of Translational Medicine for Treating High-Incidence Infectious Diseases with Integrative Medicine, Guangxi University of Chinese Medicine, Nanning, China; Key Laboratory of Pathogenic Fungi and Mycotoxins of Fujian Province, School of Life Sciences, Fujian Agriculture and Forestry University, Fuzhou 350002, China.
| |
Collapse
|
2
|
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
|
3
|
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
|
4
|
McMahon KL, O'Brien H, Schroeder CI, Deuis JR, Venkatachalam D, Huang D, Green BR, Bandyopadhyay PK, Li Q, Yandell M, Safavi-Hemami H, Olivera BM, Vetter I, Robinson SD. Identification of sodium channel toxins from marine cone snails of the subgenera Textilia and Afonsoconus. Cell Mol Life Sci 2023; 80:287. [PMID: 37689602 PMCID: PMC10492761 DOI: 10.1007/s00018-023-04935-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2023] [Revised: 08/20/2023] [Accepted: 08/22/2023] [Indexed: 09/11/2023]
Abstract
Voltage-gated sodium (NaV) channels are transmembrane proteins that play a critical role in electrical signaling in the nervous system and other excitable tissues. µ-Conotoxins are peptide toxins from the venoms of marine cone snails (genus Conus) that block NaV channels with nanomolar potency. Most species of the subgenera Textilia and Afonsoconus are difficult to acquire; therefore, their venoms have yet to be comprehensively interrogated for µ-conotoxins. The goal of this study was to find new µ-conotoxins from species of the subgenera Textilia and Afonsoconus and investigate their selectivity at human NaV channels. Using RNA-seq of the venom gland of Conus (Textilia) bullatus, we identified 12 µ-conotoxin (or µ-conotoxin-like) sequences. Based on these sequences we designed primers which we used to identify additional µ-conotoxin sequences from DNA extracted from historical specimens of species from Textilia and Afonsoconus. We synthesized six of these µ-conotoxins and tested their activity on human NaV1.1-NaV1.8. Five of the six synthetic peptides were potent blockers of human NaV channels. Of these, two peptides (BuIIIB and BuIIIE) were potent blockers of hNaV1.3. Three of the peptides (BuIIIB, BuIIIE and AdIIIA) had submicromolar activity at hNaV1.7. This study serves as an example of the identification of new peptide toxins from historical DNA and provides new insights into structure-activity relationships of µ-conotoxins with activity at hNaV1.3 and hNaV1.7.
Collapse
Affiliation(s)
- Kirsten L McMahon
- Institute for Molecular Bioscience, The University of Queensland, St Lucia, QLD, 4072, Australia
| | - Henrik O'Brien
- Biology Department, University of Utah, Salt Lake City, UT, 84112, USA
| | - Christina I Schroeder
- Institute for Molecular Bioscience, The University of Queensland, St Lucia, QLD, 4072, Australia
- Peptide Therapeutics, Genentech, 1 DNA Way, South San Francisco, CA, 94080, USA
| | - Jennifer R Deuis
- Institute for Molecular Bioscience, The University of Queensland, St Lucia, QLD, 4072, Australia
| | | | - Di Huang
- Institute for Molecular Bioscience, The University of Queensland, St Lucia, QLD, 4072, Australia
| | - Brad R Green
- Biology Department, University of Utah, Salt Lake City, UT, 84112, USA
| | | | - Qing Li
- Department of Human Genetics, Utah Center for Genetic Discovery, University of Utah, Salt Lake City, UT, 84112, USA
- Cancer Bioinformatics, Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, 84112, USA
| | - Mark Yandell
- Department of Human Genetics, Utah Center for Genetic Discovery, University of Utah, Salt Lake City, UT, 84112, USA
| | | | | | - Irina Vetter
- Institute for Molecular Bioscience, The University of Queensland, St Lucia, QLD, 4072, Australia
| | - Samuel D Robinson
- Institute for Molecular Bioscience, The University of Queensland, St Lucia, QLD, 4072, Australia.
- Biology Department, University of Utah, Salt Lake City, UT, 84112, USA.
| |
Collapse
|
5
|
Zhao Z, Pan T, Chen S, Harvey PJ, Zhang J, Li X, Yang M, Huang L, Wang S, Craik DJ, Jiang T, Yu R. Design, synthesis, and mechanism of action of novel μ-conotoxin KIIIA analogues for inhibition of the voltage-gated sodium channel Na v1.7. J Biol Chem 2023; 299:103068. [PMID: 36842500 PMCID: PMC10074208 DOI: 10.1016/j.jbc.2023.103068] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2022] [Revised: 02/16/2023] [Accepted: 02/22/2023] [Indexed: 02/28/2023] Open
Abstract
μ-Conotoxin KIIIA, a selective blocker of sodium channels, has strong inhibitory activity against several Nav isoforms, including Nav1.7, and has potent analgesic effects, but it contains three pairs of disulfide bonds, making structural modification difficult and synthesis complex. To circumvent these difficulties, we designed and synthesized three KIIIA analogues with one disulfide bond deleted. The most active analogue, KIIIA-1, was further analyzed, and its binding pattern to hNav1.7 was determined by molecular dynamics simulations. Guided by the molecular dynamics computational model, we designed and tested 32 second-generation and 6 third-generation analogues of KIIIA-1 on hNav1.7 expressed in HEK293 cells. Several analogues showed significantly improved inhibitory activity on hNav1.7, and the most potent peptide, 37, was approximately 4-fold more potent than the KIIIA Isomer I and 8-fold more potent than the wildtype (WT) KIIIA in inhibiting hNav1.7 current. Intraperitoneally injected 37 exhibited potent in vivo analgesic activity in a formalin-induced inflammatory pain model, with activity reaching ∼350-fold of the positive control drug morphine. Overall, peptide 37 has a simplified disulfide-bond framework and exhibits potent in vivo analgesic effects and has promising potential for development as a pain therapy in the future.
Collapse
Affiliation(s)
- Zitong Zhao
- Key Laboratory of Marine Drugs, Chinese Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao, China; Laboratory for Marine Drugs and Bioproducts, Qingdao National Laboratory for Marine Science and Technology, Qingdao, China
| | - Teng Pan
- Key Laboratory of Marine Drugs, Chinese Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao, China; Laboratory for Marine Drugs and Bioproducts, Qingdao National Laboratory for Marine Science and Technology, Qingdao, China
| | - Shen Chen
- Key Laboratory of Marine Drugs, Chinese Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao, China; Laboratory for Marine Drugs and Bioproducts, Qingdao National Laboratory for Marine Science and Technology, Qingdao, China
| | - Peta J Harvey
- Institute for Molecular Bioscience, Australian Research Council Centre of Excellence for Innovations in Peptide and Protein Science, The University of Queensland, Brisbane, Queensland, Australia
| | - Jinghui Zhang
- Key Laboratory of Marine Drugs, Chinese Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao, China; Laboratory for Marine Drugs and Bioproducts, Qingdao National Laboratory for Marine Science and Technology, Qingdao, China
| | - Xiao Li
- Key Laboratory of Marine Drugs, Chinese Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao, China; Laboratory for Marine Drugs and Bioproducts, Qingdao National Laboratory for Marine Science and Technology, Qingdao, China
| | - Mengke Yang
- Key Laboratory of Marine Drugs, Chinese Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao, China; Laboratory for Marine Drugs and Bioproducts, Qingdao National Laboratory for Marine Science and Technology, Qingdao, China
| | - Linhong Huang
- Key Laboratory of Marine Drugs, Chinese Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao, China; Laboratory for Marine Drugs and Bioproducts, Qingdao National Laboratory for Marine Science and Technology, Qingdao, China
| | - Shoushi Wang
- Qingdao Central Hospital, Central Hospital Affiliated to Qingdao University, Qingdao, China
| | - David J Craik
- Institute for Molecular Bioscience, Australian Research Council Centre of Excellence for Innovations in Peptide and Protein Science, The University of Queensland, Brisbane, Queensland, Australia
| | - Tao Jiang
- Key Laboratory of Marine Drugs, Chinese Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao, China; Laboratory for Marine Drugs and Bioproducts, Qingdao National Laboratory for Marine Science and Technology, Qingdao, China
| | - Rilei Yu
- Key Laboratory of Marine Drugs, Chinese Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao, China; Laboratory for Marine Drugs and Bioproducts, Qingdao National Laboratory for Marine Science and Technology, Qingdao, China.
| |
Collapse
|
6
|
Eagles DA, Chow CY, King GF. Fifteen years of Na
V
1.7 channels as an analgesic target: Why has excellent in vitro pharmacology not translated into in vivo analgesic efficacy? Br J Pharmacol 2020; 179:3592-3611. [DOI: 10.1111/bph.15327] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Revised: 09/14/2020] [Accepted: 10/23/2020] [Indexed: 12/16/2022] Open
Affiliation(s)
- David A. Eagles
- Institute for Molecular Bioscience The University of Queensland St Lucia QLD Australia
| | - Chun Yuen Chow
- Institute for Molecular Bioscience The University of Queensland St Lucia QLD Australia
| | - Glenn F. King
- Institute for Molecular Bioscience The University of Queensland St Lucia QLD Australia
| |
Collapse
|
7
|
Yang M, Zhou M. μ-conotoxin TsIIIA, a peptide inhibitor of human voltage-gated sodium channel hNa v1.8. Toxicon 2020; 186:29-34. [PMID: 32758497 DOI: 10.1016/j.toxicon.2020.07.024] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2020] [Revised: 07/19/2020] [Accepted: 07/22/2020] [Indexed: 10/23/2022]
Abstract
TsIIIA, the first μ-conotoxin from Conus tessulatus, can selectively inhibit rat tetrodotoxin-resistant sodium channels. TsIIIA also shows potent analgesic activity in a mice hotplate analgesic assay, but its effect on human sodium channels remains unknown. In this study, eight human sodium channel subtypes, hNav1.1- hNav1.8, were expressed in HEK293 or ND7/23 cells and tested on the chemically synthesized TsIIIA. Patch clamp experiments showed that 10 μM TsIIIA had no effects on the tetrodotoxin-sensitive hNav1.1, hNav1.2, hNav1.3, hNav1.4, hNav1.6 and hNav1.7, as well as tetrodotoxin-resistant hNav1.5. For tetrodotoxin-resistant hNav1.8, concentrations of 1, 5 and 10 μM TsIIIA reduced the hNav1.8 currents to 59.26%, 36.21% and 24.93% respectively. Further detailed dose-effect experiments showed that TsIIIA inhibited hNav1.8 currents with an IC50 value of 2.11 μM. In addition, 2 μM TsIIIA did not induce a shift in the current-voltage relationship of hNav1.8. Taken together, the hNav1.8 peptide inhibitor TsIIIA provides a pharmacological probe for sodium channels and a potential therapeutic agent for pain.
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
| | - 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, 410008, China.
| |
Collapse
|
8
|
Walker AA, Robinson SD, Hamilton BF, Undheim EAB, King GF. Deadly Proteomes: A Practical Guide to Proteotranscriptomics of Animal Venoms. Proteomics 2020; 20:e1900324. [PMID: 32820606 DOI: 10.1002/pmic.201900324] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2020] [Revised: 08/07/2020] [Indexed: 11/11/2022]
Abstract
Animal venoms are renowned for their toxicity, biochemical complexity, and as a source of compounds with potential applications in medicine, agriculture, and industry. Polypeptides underlie much of the pharmacology of animal venoms, and elucidating these arsenals of polypeptide toxins-known as the venom proteome or venome-is an important step in venom research. Proteomics is used for the identification of venom toxins, determination of their primary structure including post-translational modifications, as well as investigations into the physiology underlying their production and delivery. Advances in proteomics and adjacent technologies has led to a recent upsurge in publications reporting venom proteomes. Improved mass spectrometers, better proteomic workflows, and the integration of next-generation sequencing of venom-gland transcriptomes and venomous animal genomes allow quicker and more accurate profiling of venom proteomes with greatly reduced starting material. Technologies such as imaging mass spectrometry are revealing additional insights into the mechanism, location, and kinetics of venom toxin production. However, these numerous new developments may be overwhelming for researchers designing venom proteome studies. Here, the field of venom proteomics is reviewed and some practical solutions for simplifying mass spectrometry workflows to study animal venoms are offered.
Collapse
Affiliation(s)
- Andrew A Walker
- Institute for Molecular Bioscience, The University of Queensland, St. Lucia, Queensland, 4072, Australia
| | - Samuel D Robinson
- Institute for Molecular Bioscience, The University of Queensland, St. Lucia, Queensland, 4072, Australia
| | - Brett F Hamilton
- Centre for Microscopy and Microanalysis, The University of Queensland, St. Lucia, Queensland, 4072, Australia.,Centre for Advanced Imaging, The University of Queensland, St. Lucia, Queensland, 4072, Australia
| | - Eivind A B Undheim
- Centre for Advanced Imaging, The University of Queensland, St. Lucia, Queensland, 4072, Australia.,Department of Biology, Centre for Biodiversity Dynamics, NTNU, Trondheim, 7491, Norway.,Department of Bioscience, Centre for Ecological and Evolutionary Synthesis, University of Oslo, Blindern, Oslo, 0316, Norway
| | - Glenn F King
- Institute for Molecular Bioscience, The University of Queensland, St. Lucia, Queensland, 4072, Australia
| |
Collapse
|
9
|
Inhibitors of blood coagulation factor XIII. Anal Biochem 2020; 605:113708. [PMID: 32335064 DOI: 10.1016/j.ab.2020.113708] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2020] [Revised: 03/11/2020] [Accepted: 03/30/2020] [Indexed: 02/07/2023]
Abstract
The blood coagulation factor XIII (FXIII) plays an essential role in the stabilization of fibrin clots. This factor, belonging to the class of transglutaminases, catalyzes the final step of secondary hemostasis, i.e. the crosslinking of fibrin polymers. These crosslinks protect the clots against premature fibrinolysis. Consequently, FXIII is an interesting target for the therapeutic treatment of cardiovascular diseases. In this context, inhibitors can influence FXIII in the activation process of the enzyme itself or in its catalytic activity. To date, there is no FXIII inhibitor in medical application, but several studies have been conducted in the past. These studies provided a better understanding of FXIII and identified new lead structures for FXIII inhibitors. Next to small molecule inhibitors, the most promising candidates for the development of clinically applicable FXIII inhibitors are the peptide inhibitors tridegin and transglutaminase-inhibiting Michael acceptors (TIMAs) due to their selectivity towards activated FXIII (FXIIIa). In this review, select FXIII inhibitors and their pharmacological potential are discussed.
Collapse
|
10
|
Structure and Activity Studies of Disulfide-Deficient Analogues of αO-Conotoxin GeXIVA. J Med Chem 2020; 63:1564-1575. [DOI: 10.1021/acs.jmedchem.9b01409] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|
11
|
Bäuml CA, Schmitz T, Paul George AA, Sudarsanam M, Hardes K, Steinmetzer T, Holle LA, Wolberg AS, Pötzsch B, Oldenburg J, Biswas A, Imhof D. Coagulation Factor XIIIa Inhibitor Tridegin: On the Role of Disulfide Bonds for Folding, Stability, and Function. J Med Chem 2019; 62:3513-3523. [PMID: 30852892 PMCID: PMC6650289 DOI: 10.1021/acs.jmedchem.8b01982] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Tridegin is a potent and specific 66mer peptide inhibitor of coagulation factor XIIIa with six cysteines involved in three disulfide bonds. Three of the 15 possible 3-disulfide-bonded isomers have been identified, which share a bridge between cysteines 19 and 25. We synthesized the three possible 2-disulfide-bonded analogues using a targeted protecting group strategy to investigate the impact of the C19-C25 bond on tridegin's folding, stability, and function. The FXIIIa inhibitory activity of the analogues was retained, which was shown by in vitro fluorogenic activity and whole blood clotting assays. Molecular dynamics simulations of wild-type tridegin and the analogues as well as molecular docking studies with FXIIIa were performed to elucidate the impact of the C19-C25 bond on conformational stability and binding mode. The strategy of selectively reducing disulfide bonds to facilitate large-scale synthesis, while retaining the functionality of disulfide-bonded peptides, has been demonstrated with our present study.
Collapse
Affiliation(s)
- Charlotte A. Bäuml
- Pharmaceutical Biochemistry and Bioanalytics, Pharmaceutical Institute, University of Bonn, An der Immenburg 4, 53121 Bonn, Germany
| | - Thomas Schmitz
- Pharmaceutical Biochemistry and Bioanalytics, Pharmaceutical Institute, University of Bonn, An der Immenburg 4, 53121 Bonn, Germany
| | - Ajay A. Paul George
- Pharmaceutical Biochemistry and Bioanalytics, Pharmaceutical Institute, University of Bonn, An der Immenburg 4, 53121 Bonn, Germany
| | - Monica Sudarsanam
- Pharmaceutical Biochemistry and Bioanalytics, Pharmaceutical Institute, University of Bonn, An der Immenburg 4, 53121 Bonn, Germany
| | - Kornelia Hardes
- Department of Pharmacy, Institute of Pharmaceutical Chemistry, Philipps University of Marburg, Marbacher Weg 6, 35032 Marburg, Germany
| | - Torsten Steinmetzer
- Department of Pharmacy, Institute of Pharmaceutical Chemistry, Philipps University of Marburg, Marbacher Weg 6, 35032 Marburg, Germany
| | - Lori A. Holle
- Department of Pathology and Laboratory Medicine, University of North Carolina at Chapel Hill, 819 Brinkhous-Bullitt Building, Chapel Hill, NC 27599, USA
| | - Alisa S. Wolberg
- Department of Pathology and Laboratory Medicine, University of North Carolina at Chapel Hill, 819 Brinkhous-Bullitt Building, Chapel Hill, NC 27599, USA
| | - Bernd Pötzsch
- Institute of Experimental Hematology and Transfusion Medicine, University Hospital Bonn, Sigmund-Freud-Str. 25, 53127 Bonn, Germany
| | - Johannes Oldenburg
- Institute of Experimental Hematology and Transfusion Medicine, University Hospital Bonn, Sigmund-Freud-Str. 25, 53127 Bonn, Germany
| | - Arijit Biswas
- Institute of Experimental Hematology and Transfusion Medicine, University Hospital Bonn, Sigmund-Freud-Str. 25, 53127 Bonn, Germany
| | - Diana Imhof
- Pharmaceutical Biochemistry and Bioanalytics, Pharmaceutical Institute, University of Bonn, An der Immenburg 4, 53121 Bonn, Germany
| |
Collapse
|
12
|
Mansbach RA, Travers T, McMahon BH, Fair JM, Gnanakaran S. Snails In Silico: A Review of Computational Studies on the Conopeptides. Mar Drugs 2019; 17:E145. [PMID: 30832207 PMCID: PMC6471681 DOI: 10.3390/md17030145] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2019] [Revised: 02/21/2019] [Accepted: 02/22/2019] [Indexed: 12/26/2022] Open
Abstract
Marine cone snails are carnivorous gastropods that use peptide toxins called conopeptides both as a defense mechanism and as a means to immobilize and kill their prey. These peptide toxins exhibit a large chemical diversity that enables exquisite specificity and potency for target receptor proteins. This diversity arises in terms of variations both in amino acid sequence and length, and in posttranslational modifications, particularly the formation of multiple disulfide linkages. Most of the functionally characterized conopeptides target ion channels of animal nervous systems, which has led to research on their therapeutic applications. Many facets of the underlying molecular mechanisms responsible for the specificity and virulence of conopeptides, however, remain poorly understood. In this review, we will explore the chemical diversity of conopeptides from a computational perspective. First, we discuss current approaches used for classifying conopeptides. Next, we review different computational strategies that have been applied to understanding and predicting their structure and function, from machine learning techniques for predictive classification to docking studies and molecular dynamics simulations for molecular-level understanding. We then review recent novel computational approaches for rapid high-throughput screening and chemical design of conopeptides for particular applications. We close with an assessment of the state of the field, emphasizing important questions for future lines of inquiry.
Collapse
Affiliation(s)
- Rachael A Mansbach
- Theoretical Biology and Biophysics Group, Los Alamos National Laboratory, Los Alamos, NM 87545, USA.
| | - Timothy Travers
- Theoretical Biology and Biophysics Group, Los Alamos National Laboratory, Los Alamos, NM 87545, USA.
- Center for Nonlinear Studies, Los Alamos National Laboratory, Los Alamos, NM 87545, USA.
| | - Benjamin H McMahon
- Theoretical Biology and Biophysics Group, Los Alamos National Laboratory, Los Alamos, NM 87545, USA.
| | - Jeanne M Fair
- Biosecurity and Public Health Group, Los Alamos National Laboratory, Los Alamos, NM 87545, USA.
| | - S Gnanakaran
- Theoretical Biology and Biophysics Group, Los Alamos National Laboratory, Los Alamos, NM 87545, USA.
| |
Collapse
|
13
|
Peigneur S, Cheneval O, Maiti M, Leipold E, Heinemann SH, Lescrinier E, Herdewijn P, De Lima ME, Craik DJ, Schroeder CI, Tytgat J. Where cone snails and spiders meet: design of small cyclic sodium-channel inhibitors. FASEB J 2018; 33:3693-3703. [PMID: 30509130 DOI: 10.1096/fj.201801909r] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
A 13 aa residue voltage-gated sodium (NaV) channel inhibitor peptide, Pn, containing 2 disulfide bridges was designed by using a chimeric approach. This approach was based on a common pharmacophore deduced from sequence and secondary structural homology of 2 NaV inhibitors: Conus kinoshitai toxin IIIA, a 14 residue cone snail peptide with 3 disulfide bonds, and Phoneutria nigriventer toxin 1, a 78 residue spider toxin with 7 disulfide bonds. As with the parent peptides, this novel NaV channel inhibitor was active on NaV1.2. Through the generation of 3 series of peptide mutants, we investigated the role of key residues and cyclization and their influence on NaV inhibition and subtype selectivity. Cyclic PnCS1, a 10 residue peptide cyclized via a disulfide bond, exhibited increased inhibitory activity toward therapeutically relevant NaV channel subtypes, including NaV1.7 and NaV1.9, while displaying remarkable serum stability. These peptides represent the first and the smallest cyclic peptide NaV modulators to date and are promising templates for the development of toxin-based therapeutic agents.-Peigneur, S., Cheneval, O., Maiti, M., Leipold, E., Heinemann, S. H., Lescrinier, E., Herdewijn, P., De Lima, M. E., Craik, D. J., Schroeder, C. I., Tytgat, J. Where cone snails and spiders meet: design of small cyclic sodium-channel inhibitors.
Collapse
Affiliation(s)
- Steve Peigneur
- Toxicology and Pharmacology, Katholieke Universiteit (KU) Leuven, Campus Gasthuisberg, Leuven, Belgium.,Department de Bioquímica e Imunologia, Laboratório de Venenos e Toxinas Animais, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo-Horizonte, Brazil
| | - Olivier Cheneval
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland, Australia
| | - Mohitosh Maiti
- Laboratory of Medicinal Chemistry, Rega Institute for Medical Research, Katholieke Universiteit (KU) Leuven, Leuven, Belgium
| | - Enrico Leipold
- Department of Biophysics, Center for Molecular Biomedicine, Jena University Hospital, Friedrich Schiller University Jena, Germany
| | - Stefan H Heinemann
- Department of Biophysics, Center for Molecular Biomedicine, Jena University Hospital, Friedrich Schiller University Jena, Germany
| | - Eveline Lescrinier
- Laboratory of Medicinal Chemistry, Rega Institute for Medical Research, Katholieke Universiteit (KU) Leuven, Leuven, Belgium
| | - Piet Herdewijn
- Laboratory of Medicinal Chemistry, Rega Institute for Medical Research, Katholieke Universiteit (KU) Leuven, Leuven, Belgium
| | - Maria Elena De Lima
- Department de Bioquímica e Imunologia, Laboratório de Venenos e Toxinas Animais, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo-Horizonte, Brazil.,Programa de Pós-Graduação em Ciências da Saúde, Biomedicina e Medicina, Instituto de Ensino e Pesquisa da Santa Casa de Belo Horizonte, Grupo Santa Casa de Belo Horizonte, Belo Horizonte, Brazil
| | - David J Craik
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland, Australia
| | - Christina I Schroeder
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland, Australia
| | - Jan Tytgat
- Toxicology and Pharmacology, Katholieke Universiteit (KU) Leuven, Campus Gasthuisberg, Leuven, Belgium
| |
Collapse
|
14
|
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: 68] [Impact Index Per Article: 9.7] [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
|
15
|
Han P, Wang K, Dai X, Cao Y, Liu S, Jiang H, Fan C, Wu W, Chen J. The Role of Individual Disulfide Bonds of μ-Conotoxin GIIIA in the Inhibition of Na V1.4. Mar Drugs 2016; 14:md14110213. [PMID: 27869701 PMCID: PMC5128756 DOI: 10.3390/md14110213] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2016] [Revised: 10/31/2016] [Accepted: 11/11/2016] [Indexed: 12/20/2022] Open
Abstract
μ-Conotoxin GIIIA, a peptide toxin isolated from Conus geographus, preferentially blocks the skeletal muscle sodium channel NaV1.4. GIIIA folds compactly to a pyramidal structure stabilized by three disulfide bonds. To assess the contributions of individual disulfide bonds of GIIIA to the blockade of NaV1.4, seven disulfide-deficient analogues were prepared and characterized, each with one, two, or three pairs of disulfide-bonded Cys residues replaced with Ala. The inhibitory potency of the analogues against NaV1.4 was assayed by whole cell patch-clamp on rNaV1.4, heterologously expressed in HEK293 cells. The corresponding IC50 values were 0.069 ± 0.005 μM for GIIIA, 2.1 ± 0.3 μM for GIIIA-1, 3.3 ± 0.2 μM for GIIIA-2, and 15.8 ± 0.8 μM for GIIIA-3 (-1, -2 and -3 represent the removal of disulfide bridges Cys3–Cys15, Cys4–Cys20 and Cys10–Cys21, respectively). Other analogues were not active enough for IC50 measurement. Our results indicate that all three disulfide bonds of GIIIA are required to produce effective inhibition of NaV1.4, and the removal of any one significantly lowers its sodium channel binding affinity. Cys10–Cys21 is the most important for the NaV1.4 potency.
Collapse
Affiliation(s)
- Penggang Han
- College of Science, National University of Defense Technology, Changsha 410073, Hunan, China.
- Beijing Institute of Pharmaceutical Chemistry, Beijing 102205, China.
| | - Kang Wang
- Beijing Institute of Pharmaceutical Chemistry, Beijing 102205, China.
| | - Xiandong Dai
- Beijing Institute of Pharmaceutical Chemistry, Beijing 102205, China.
| | - Ying Cao
- Beijing Institute of Pharmaceutical Chemistry, Beijing 102205, China.
| | - Shangyi Liu
- Beijing Institute of Pharmaceutical Chemistry, Beijing 102205, China.
| | - Hui Jiang
- Beijing Institute of Pharmaceutical Chemistry, Beijing 102205, China.
| | - Chongxu Fan
- Beijing Institute of Pharmaceutical Chemistry, Beijing 102205, China.
| | - Wenjian Wu
- College of Science, National University of Defense Technology, Changsha 410073, Hunan, China.
| | - Jisheng Chen
- College of Science, National University of Defense Technology, Changsha 410073, Hunan, China.
- Beijing Institute of Pharmaceutical Chemistry, Beijing 102205, China.
| |
Collapse
|
16
|
Structure and function of μ-conotoxins, peptide-based sodium channel blockers with analgesic activity. Future Med Chem 2015; 6:1677-98. [PMID: 25406007 DOI: 10.4155/fmc.14.107] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
μ-Conotoxins block voltage-gated sodium channels (VGSCs) and compete with tetrodotoxin for binding to the sodium conductance pore. Early efforts identified µ-conotoxins that preferentially blocked the skeletal muscle subtype (NaV1.4). However, the last decade witnessed a significant increase in the number of µ-conotoxins and the range of VGSC subtypes inhibited (NaV1.2, NaV1.3 or NaV1.7). Twenty µ-conotoxin sequences have been identified to date and structure-activity relationship studies of several of these identified key residues responsible for interactions with VGSC subtypes. Efforts to engineer-in subtype specificity are driven by in vivo analgesic and neuromuscular blocking activities. This review summarizes structural and pharmacological studies of µ-conotoxins, which show promise for development of selective blockers of NaV1.2, and perhaps also NaV1.1,1.3 or 1.7.
Collapse
|
17
|
Kwong K, Carr MJ. Voltage-gated sodium channels. Curr Opin Pharmacol 2015; 22:131-9. [PMID: 26043074 DOI: 10.1016/j.coph.2015.04.007] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2015] [Revised: 04/21/2015] [Accepted: 04/29/2015] [Indexed: 12/11/2022]
Abstract
Voltage-gated sodium channels play a key role in the transmission of sensory information about the status of organs in the periphery. Sensory fibers contain a heterogeneous yet specific distribution of voltage-gated sodium channel isoforms. Major efforts by industry and academic groups are underway to develop medicines that interrupt inappropriate signaling for a number of clinical indications by taking advantage of this specific distribution of channel isoforms. This review highlights recent advances in the study of human channelopathies, animal toxins and channel structure that may facilitate the development of selective voltage-gated sodium channel blockers.
Collapse
|
18
|
Sharma H, Nagaraj R. Human β-defensin 4 with non-native disulfide bridges exhibit antimicrobial activity. PLoS One 2015; 10:e0119525. [PMID: 25785690 PMCID: PMC4364940 DOI: 10.1371/journal.pone.0119525] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2014] [Accepted: 01/14/2015] [Indexed: 01/05/2023] Open
Abstract
Human defensins play multiple roles in innate immunity including direct antimicrobial killing and immunomodulatory activity. They have three disulfide bridges which contribute to the stability of three anti-parallel β-strands. The exact role of disulfide bridges and canonical β-structure in the antimicrobial action is not yet fully understood. In this study, we have explored the antimicrobial activity of human β-defensin 4 (HBD4) analogs that differ in the number and connectivity of disulfide bridges. The cysteine framework was similar to the disulfide bridges present in μ-conotoxins, an unrelated class of peptide toxins. All the analogs possessed enhanced antimicrobial potency as compared to native HBD4. Among the analogs, the single disulfide bridged peptide showed maximum potency. However, there were no marked differences in the secondary structure of the analogs. Subtle variations were observed in the localization and membrane interaction of the analogs with bacteria and Candida albicans, suggesting a role for disulfide bridges in modulating their antimicrobial action. All analogs accumulated in the cytosol where they can bind to anionic molecules such as nucleic acids which would affect several cellular processes leading to cell death. Our study strongly suggests that native disulfide bridges or the canonical β-strands in defensins have not evolved for maximal activity but they play important roles in determining their antimicrobial potency.
Collapse
Affiliation(s)
- Himanshu Sharma
- CSIR-Centre for Cellular and Molecular Biology, Hyderabad, Telangana, India
| | - Ramakrishnan Nagaraj
- CSIR-Centre for Cellular and Molecular Biology, Hyderabad, Telangana, India
- * E-mail:
| |
Collapse
|
19
|
Systematic study of binding of μ-conotoxins to the sodium channel NaV1.4. Toxins (Basel) 2014; 6:3454-70. [PMID: 25529306 PMCID: PMC4280544 DOI: 10.3390/toxins6123454] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2014] [Revised: 12/01/2014] [Accepted: 12/10/2014] [Indexed: 01/25/2023] Open
Abstract
Voltage-gated sodium channels (NaV) are fundamental components of the nervous system. Their dysfunction is implicated in a number of neurological disorders, such as chronic pain, making them potential targets for the treatment of such disorders. The prominence of the NaV channels in the nervous system has been exploited by venomous animals for preying purposes, which have developed toxins that can block the NaV channels, thereby disabling their function. Because of their potency, such toxins could provide drug leads for the treatment of neurological disorders associated with NaV channels. However, most toxins lack selectivity for a given target NaV channel, and improving their selectivity profile among the NaV1 isoforms is essential for their development as drug leads. Computational methods will be very useful in the solution of such design problems, provided accurate models of the protein-ligand complex can be constructed. Using docking and molecular dynamics simulations, we have recently constructed a model for the NaV1.4-μ-conotoxin-GIIIA complex and validated it with the ample mutational data available for this complex. Here, we use the validated NaV1.4 model in a systematic study of binding other μ-conotoxins (PIIIA, KIIIA and BuIIIB) to NaV1.4. The binding mode obtained for each complex is shown to be consistent with the available mutation data and binding constants. We compare the binding modes of PIIIA, KIIIA and BuIIIB to that of GIIIA and point out the similarities and differences among them. The detailed information about NaV1.4-μ-conotoxin interactions provided here will be useful in the design of new NaV channel blocking peptides.
Collapse
|
20
|
Furman JL, Chiu M, Hunter MJ. Early engineering approaches to improve peptide developability and manufacturability. AAPS JOURNAL 2014; 17:111-20. [PMID: 25338742 DOI: 10.1208/s12248-014-9681-9] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/16/2014] [Accepted: 10/02/2014] [Indexed: 02/08/2023]
Abstract
Downstream success in Pharmaceutical Development requires thoughtful molecule design early in the lifetime of any potential therapeutic. Most therapeutic monoclonal antibodies are quite similar with respect to their developability properties. However, the properties of therapeutic peptides tend to be as diverse as the molecules themselves. Analysis of the primary sequence reveals sites of potential adverse posttranslational modifications including asparagine deamidation, aspartic acid isomerization, methionine, tryptophan, and cysteine oxidation and, potentially, chemical and proteolytic degradation liabilities that can impact the developability and manufacturability of a potential therapeutic peptide. Assessing these liabilities, both biophysically and functionally, early in a molecule's lifetime can drive a more effective path forward in the drug discovery process. In addition to these potential liabilities, more complex peptides that contain multiple disulfide bonds can pose particular challenges with respect to production and manufacturability. Approaches to reducing the disulfide bond complexity of these peptides are often explored with mixed success. Proteolytic degradation is a major contributor to decreased half-life and efficacy. Addressing this aspect of peptide stability early in the discovery process increases downstream success. We will address aspects of peptide sequence analysis, molecule complexity, developability analysis, and manufacturing routes that drive the decision making processes during peptide therapeutic development.
Collapse
Affiliation(s)
- Jennifer L Furman
- Janssen Research & Development, LLC, 3210 Merryfield Row, San Diego, California, 92121, USA
| | | | | |
Collapse
|
21
|
Sato K, Yamaguchi Y, Ishida Y, Ohizumi Y. Roles of basic amino acid residues in the activity of μ-conotoxin GIIIA and GIIIB, peptide blockers of muscle sodium channels. Chem Biol Drug Des 2014; 85:488-93. [PMID: 25228447 DOI: 10.1111/cbdd.12433] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2014] [Revised: 06/29/2014] [Accepted: 08/29/2014] [Indexed: 11/29/2022]
Abstract
To study in detail the roles of basic amino acid residues in the activity of μ-conotoxin GIIIA (μ-GIIIA) and GIIIB (μ-GIIIB), specific blockers of muscle sodium channels, seven analogs of μ-GIIIA, and two analogs of μ-GIIIB were synthesized. μ-GIIIA analogs were synthesized by replacing systematically the three Arg residues (Arg1, Arg13, and Arg19) with one, two, and three Lys residues. μ-GIIIB analogs were synthesized by replacing simultaneously all four Lys residues (Lys9, Lys11, Lys16, and Lys19) with Arg residues and further replacement of acidic Asp residues with neutral Ala residues. Circular dichroism spectra of the synthesized analogs suggested that the replacement did not affect the three dimensional structure. The inhibitory effects on the twitch contractions of the rat diaphragm showed that the side chain guanidino group of Arg13 of μ-GIIIA was important for the activity, whereas that of Arg19 had little role for biological activity. Although [Arg9,11,16,19]μ-GIIIB showed higher activity than native μ-GIIIB, highly basic [Ala2,12, Arg9,11,16,19]μ-GIIIB showed lower activity, suggesting that there was an appropriate molecular basicity for the maximum activity.
Collapse
Affiliation(s)
- Kazuki Sato
- Department of Environmental Science, Fukuoka Women's University, 1-1-1 Kasumigaoka, Higashi-ku, Fukuoka, 813-8529, Japan
| | | | | | | |
Collapse
|
22
|
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: 237] [Impact Index Per Article: 21.5] [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
|
23
|
Green BR, Zhang MM, Chhabra S, Robinson SD, Wilson MJ, Redding A, Olivera BM, Yoshikami D, Bulaj G, Norton RS. Interactions of disulfide-deficient selenocysteine analogs of μ-conotoxin BuIIIB with the α-subunit of the voltage-gated sodium channel subtype 1.3. FEBS J 2014; 281:2885-98. [PMID: 24814369 DOI: 10.1111/febs.12835] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2014] [Revised: 04/18/2014] [Accepted: 05/05/2014] [Indexed: 12/17/2022]
Abstract
Inhibitors of the α-subunit of the voltage-gated sodium channel subtype 1.3 (NaV 1.3) are of interest as pharmacological tools for the study of neuropathic pain associated with spinal cord injury and have potential therapeutic applications. The recently described μ-conotoxin BuIIIB (μ-BuIIIB) from Conus bullatus was shown to block NaV 1.3 with submicromolar potency (Kd = 0.2 μm), making it one of the most potent peptidic inhibitors of this subtype described to date. However, oxidative folding of μ-BuIIIB results in numerous folding isoforms, making it difficult to obtain sufficient quantities of the active form of the peptide for detailed structure-activity studies. In the present study, we report the synthesis and characterization of μ-BuIIIB analogs incorporating a disulfide-deficient, diselenide-containing scaffold designed to simplify synthesis and facilitate structure-activity studies directed at identifying amino acid residues involved in NaV 1.3 blockade. Our results indicate that, similar to other μ-conotoxins, the C-terminal residues (Trp16, Arg18 and His20) are most crucial for NaV 1 blockade. At the N-terminus, replacement of Glu3 by Ala resulted in an analog with an increased potency for NaV 1.3 (Kd = 0.07 μm), implicating this position as a potential site for modification for increased potency and/or selectivity. Further examination of this position showed that increased negative charge, through γ-carboxyglutamate replacement, decreased potency (Kd = 0.33 μm), whereas replacement with positively-charged 2,4-diamonobutyric acid increased potency (Kd = 0.036 μm). These results provide a foundation for the design and synthesis of μ-BuIIIB-based analogs with increased potency against NaV 1.3.
Collapse
Affiliation(s)
- Brad R Green
- Medicinal Chemistry, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Vic., Australia; Department of Medicinal Chemistry, College of Pharmacy, University of Utah, Salt Lake City, UT, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
24
|
Lebbe EKM, Peigneur S, Brullot W, Verbiest T, Tytgat J. Ala-7, His-10 and Arg-12 are crucial amino acids for activity of a synthetically engineered μ-conotoxin. Peptides 2014; 53:300-6. [PMID: 23871692 DOI: 10.1016/j.peptides.2013.07.005] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/13/2013] [Revised: 07/01/2013] [Accepted: 07/01/2013] [Indexed: 12/19/2022]
Abstract
Cone snail toxins or conotoxins are often small cysteine-rich peptides which have shown to be highly selective ligands for a wide range of ion channels such as voltage-gated sodium channels (Na(V)s). Na(V)s participate in a wide range of electrophysiological processes. Consequently, their malfunction has been associated with numerous diseases. The development of subtype-selective modulators of Na(V)s remains highly important in the treatment of such disorders. In order to expand our knowledge in the search for novel therapeutics to treat Na(V)-related diseases, we explored the field of peptide engineering. In the current study, the impact of well considered point mutations into a bioactive peptide that was found to be a very potent and selective inhibitor of Na(V)s (i.e. Midi R2) was examined. We designed two peptides, named Midi R2[A7G] and Midi R2[H10A, R12A] which have mutations at position 7, and both 10 and 12, respectively. Electrophysiological recordings indicated that an Ala to Gly mutation at position 7 increased IC50-values from the nanomolar range to the micromolar range. For Midi R2[H10A, R12A] at a concentration of 10 μM, activity is even reduced to 0-10% for all of the tested Na(V)-channels. Circular dichroism measurements proved that overall structural conformations did not change. These findings suggest that the minimal space between the second and the third intercysteine loop of Midi R2 is the sequence RRWARDHSR and that His at position 10 and Arg at position 12 are crucial amino acids for the potency and specificity of Midi R2. In this way, new insights into the structure-activity relationships of μ-conotoxins were found.
Collapse
Affiliation(s)
- Eline K M Lebbe
- Toxicology and Pharmacology, University of Leuven (KU Leuven), Campus Gasthuisberg O&N2, Herestraat 49, P.O. Box 922, 3000 Leuven, Belgium.
| | - Steve Peigneur
- Toxicology and Pharmacology, University of Leuven (KU Leuven), Campus Gasthuisberg O&N2, Herestraat 49, P.O. Box 922, 3000 Leuven, Belgium.
| | - Ward Brullot
- Laboratory for Molecular Electronics and Photonics, Division Molecular Imaging and Photonics, Department of Chemistry, University of Leuven, Celestijnenlaan 200D, P.O. Box 2425, 3001 Heverlee, Belgium.
| | - Thierry Verbiest
- Laboratory for Molecular Electronics and Photonics, Division Molecular Imaging and Photonics, Department of Chemistry, University of Leuven, Celestijnenlaan 200D, P.O. Box 2425, 3001 Heverlee, Belgium.
| | - Jan Tytgat
- Toxicology and Pharmacology, University of Leuven (KU Leuven), Campus Gasthuisberg O&N2, Herestraat 49, P.O. Box 922, 3000 Leuven, Belgium.
| |
Collapse
|
25
|
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
|
26
|
Sato K, Yamaguchi Y, Ishida Y. Roles of Individual Disulfide Bridges in the Conformation and Activity of μ-Conotoxin GIIIA, a Peptide Blocker of Muscle Sodium Channels. Int J Pept Res Ther 2013. [DOI: 10.1007/s10989-013-9389-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
|
27
|
Incorporation of post-translational modified amino acids as an approach to increase both chemical and biological diversity of conotoxins and conopeptides. Amino Acids 2013; 46:125-51. [DOI: 10.1007/s00726-013-1606-x] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2013] [Accepted: 10/17/2013] [Indexed: 02/06/2023]
|
28
|
Zhou M, Wang L, Wu Y, Zhu X, Feng Y, Chen Z, Li Y, Sun D, Ren Z, Xu A. Characterizing the evolution and functions of the M-superfamily conotoxins. Toxicon 2013; 76:150-9. [PMID: 24080356 DOI: 10.1016/j.toxicon.2013.09.020] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2013] [Revised: 08/28/2013] [Accepted: 09/19/2013] [Indexed: 10/26/2022]
Abstract
Conotoxins from cone snails are valuable in physiology research and therapeutic applications. Evolutionary mechanisms of conotoxins have been investigated in several superfamilies, but there is no phylogenetic analysis on M-superfamily conotoxins. In this study, we characterized identical sequences, gene structure, novel cysteine frameworks, functions and evolutionary mechanisms of M-superfamily conotoxins. Identical M-superfamily conotoxins can be found in different Conus species from the analysis of novel 467 M-superfamily conotoxin sequences and other published M-superfamily conotoxins sequences. M-superfamily conotoxin genes consist of two introns and three exons from the results of genome walking. Eighteen cysteine frameworks were identified from the M-superfamily conotoxins, and 10 of the 18 may be generated from framework III. An analysis between diet types and phylogeny of the M-superfamily conotoxins indicate that M-superfamily conotoxins might not evolve in a concerted manner but were subject to birth-and-death evolution. Codon usage analysis shows that position-specific codon conservation is not restricted to cysteines, but also to other conserved residues. By analysing primary structures and physiological functions of M-superfamily conotoxins, we proposed a hypothesis that insertions and deletions, especially insertions in the third cysteine loop, are involved in the creation of new functions and structures of the M-superfamily conotoxins.
Collapse
Affiliation(s)
- Maojun Zhou
- State Key Laboratory of Biocontrol, Guangdong Province Key Laboratory of Pharmaceutical Functional Genes, National Engineering Research Center of South China Sea Marine Biotechnology, Department of Biochemistry, College of Life Sciences, Sun Yat-sen University, Guangzhou 510275, People's Republic of China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
29
|
Stevens M, Timmermans S, Bottelbergs A, Hendriks JJ, Brône B, Baes M, Tytgat J. Block of a subset of sodium channels exacerbates experimental autoimmune encephalomyelitis. J Neuroimmunol 2013; 261:21-8. [DOI: 10.1016/j.jneuroim.2013.04.012] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2012] [Revised: 03/19/2013] [Accepted: 04/11/2013] [Indexed: 10/26/2022]
|
30
|
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
|
31
|
Walewska A, Han TS, Zhang MM, Yoshikami D, Bulaj G, Rolka K. Expanding chemical diversity of conotoxins: peptoid-peptide chimeras of the sodium channel blocker μ-KIIIA and its selenopeptide analogues. Eur J Med Chem 2013; 65:144-50. [PMID: 23707919 DOI: 10.1016/j.ejmech.2013.04.041] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2012] [Revised: 04/19/2013] [Accepted: 04/20/2013] [Indexed: 12/19/2022]
Abstract
The μ-conotoxin KIIIA is a three disulfide-bridged blocker of voltage-gated sodium channels (VGSCs). The Lys(7) residue in KIIIA is an attractive target for manipulating the selectivity and efficacy of this peptide. Here, we report the design and chemical synthesis of μ-conopeptoid analogues (peptomers) in which we replaced Lys(7) with peptoid monomers of increasing side-chain size: N-methylglycine, N-butylglycine and N-octylglycine. In the first series of analogues, the peptide core contained all three disulfide bridges; whereas in the second series, a disulfide-depleted selenoconopeptide core was used to simplify oxidative folding. The analogues were tested for functional activity in blocking the Nav1.2 subtype of mammalian VGSCs exogenously expressed in Xenopus oocytes. All six analogues were active, with the N-methylglycine analogue, [Sar(7)]KIIIA, the most potent in blocking the channels while favouring lower efficacy. Our findings demonstrate that the use of N-substituted Gly residues in conotoxins show promise as a tool to optimize their pharmacological properties as potential analgesic drug leads.
Collapse
Affiliation(s)
- Aleksandra Walewska
- Faculty of Chemistry, University of Gdansk, Sobieskiego 18, 80-952 Gdansk, Poland.
| | | | | | | | | | | |
Collapse
|
32
|
Khoo KK, Gupta K, Green BR, Zhang MM, Watkins M, Olivera BM, Balaram P, Yoshikami D, Bulaj G, Norton RS. Distinct disulfide isomers of μ-conotoxins KIIIA and KIIIB block voltage-gated sodium channels. Biochemistry 2012; 51:9826-35. [PMID: 23167564 DOI: 10.1021/bi301256s] [Citation(s) in RCA: 63] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
In the preparation of synthetic conotoxins containing multiple disulfide bonds, oxidative folding can produce numerous permutations of disulfide bond connectivities. Establishing the native disulfide connectivities thus presents a significant challenge when the venom-derived peptide is not available, as is increasingly the case when conotoxins are identified from cDNA sequences. Here, we investigate the disulfide connectivity of μ-conotoxin KIIIA, which was predicted originally to have a [C1-C9,C2-C15,C4-C16] disulfide pattern based on homology with closely related μ-conotoxins. The two major isomers of synthetic μ-KIIIA formed during oxidative folding were purified and their disulfide connectivities mapped by direct mass spectrometric collision-induced dissociation fragmentation of the disulfide-bonded polypeptides. Our results show that the major oxidative folding product adopts a [C1-C15,C2-C9,C4-C16] disulfide connectivity, while the minor product adopts a [C1-C16,C2-C9,C4-C15] connectivity. Both of these peptides were potent blockers of Na(V)1.2 (K(d) values of 5 and 230 nM, respectively). The solution structure for μ-KIIIA based on nuclear magnetic resonance data was recalculated with the [C1-C15,C2-C9,C4-C16] disulfide pattern; its structure was very similar to the μ-KIIIA structure calculated with the incorrect [C1-C9,C2-C15,C4-C16] disulfide pattern, with an α-helix spanning residues 7-12. In addition, the major folding isomers of μ-KIIIB, an N-terminally extended isoform of μ-KIIIA identified from its cDNA sequence, were isolated. These folding products had the same disulfide connectivities as μ-KIIIA, and both blocked Na(V)1.2 (K(d) values of 470 and 26 nM, respectively). Our results establish that the preferred disulfide pattern of synthetic μ-KIIIA and μ-KIIIB folded in vitro is 1-5/2-4/3-6 but that other disulfide isomers are also potent sodium channel blockers. These findings raise questions about the disulfide pattern(s) of μ-KIIIA in the venom of Conus kinoshitai; indeed, the presence of multiple disulfide isomers in the venom could provide a means of further expanding the snail's repertoire of active peptides.
Collapse
Affiliation(s)
- Keith K Khoo
- Medicinal Chemistry, Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Parade, Parkville, VIC 3052, Australia
| | | | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Knapp O, McArthur JR, Adams DJ. Conotoxins targeting neuronal voltage-gated sodium channel subtypes: potential analgesics? Toxins (Basel) 2012. [PMID: 23202314 PMCID: PMC3509706 DOI: 10.3390/toxins4111236] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Voltage-gated sodium channels (VGSC) are the primary mediators of electrical signal amplification and propagation in excitable cells. VGSC subtypes are diverse, with different biophysical and pharmacological properties, and varied tissue distribution. Altered VGSC expression and/or increased VGSC activity in sensory neurons is characteristic of inflammatory and neuropathic pain states. Therefore, VGSC modulators could be used in prospective analgesic compounds. VGSCs have specific binding sites for four conotoxin families: μ-, μO-, δ- and ί-conotoxins. Various studies have identified that the binding site of these peptide toxins is restricted to well-defined areas or domains. To date, only the μ- and μO-family exhibit analgesic properties in animal pain models. This review will focus on conotoxins from the μ- and μO-families that act on neuronal VGSCs. Examples of how these conotoxins target various pharmacologically important neuronal ion channels, as well as potential problems with the development of drugs from conotoxins, will be discussed.
Collapse
Affiliation(s)
- Oliver Knapp
- Health Innovations Research Institute, RMIT University, Melbourne, Victoria 3083, Australia.
| | | | | |
Collapse
|
34
|
Favreau P, Benoit E, Hocking HG, Carlier L, D' hoedt D, Leipold E, Markgraf R, Schlumberger S, Córdova MA, Gaertner H, Paolini-Bertrand M, Hartley O, Tytgat J, Heinemann SH, Bertrand D, Boelens R, Stöcklin R, Molgó J. A novel µ-conopeptide, CnIIIC, exerts potent and preferential inhibition of NaV1.2/1.4 channels and blocks neuronal nicotinic acetylcholine receptors. Br J Pharmacol 2012; 166:1654-68. [PMID: 22229737 DOI: 10.1111/j.1476-5381.2012.01837.x] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Abstract
BACKGROUND AND PURPOSE The µ-conopeptide family is defined by its ability to block voltage-gated sodium channels (VGSCs), a property that can be used for the development of myorelaxants and analgesics. We characterized the pharmacology of a new µ-conopeptide (µ-CnIIIC) on a range of preparations and molecular targets to assess its potential as a myorelaxant. EXPERIMENTAL APPROACH µ-CnIIIC was sequenced, synthesized and characterized by its direct block of elicited twitch tension in mouse skeletal muscle and action potentials in mouse sciatic and pike olfactory nerves. µ-CnIIIC was also studied on HEK-293 cells expressing various rodent VGSCs and also on voltage-gated potassium channels and nicotinic acetylcholine receptors (nAChRs) to assess cross-interactions. Nuclear magnetic resonance (NMR) experiments were carried out for structural data. KEY RESULTS Synthetic µ-CnIIIC decreased twitch tension in mouse hemidiaphragms (IC(50) = 150 nM), and displayed a higher blocking effect in mouse extensor digitorum longus muscles (IC = 46 nM), compared with µ-SIIIA, µ-SmIIIA and µ-PIIIA. µ-CnIIIC blocked Na(V)1.4 (IC(50) = 1.3 nM) and Na(V)1.2 channels in a long-lasting manner. Cardiac Na(V)1.5 and DRG-specific Na(V)1.8 channels were not blocked at 1 µM. µ-CnIIIC also blocked the α3β2 nAChR subtype (IC(50) = 450 nM) and, to a lesser extent, on the α7 and α4β2 subtypes. Structure determination of µ-CnIIIC revealed some similarities to α-conotoxins acting on nAChRs. CONCLUSION AND IMPLICATIONS µ-CnIIIC potently blocked VGSCs in skeletal muscle and nerve, and hence is applicable to myorelaxation. Its atypical pharmacological profile suggests some common structural features between VGSCs and nAChR channels.
Collapse
|
35
|
Bingham JP, Andrews EA, Kiyabu SM, Cabalteja CC. Drugs from slugs. Part II--conopeptide bioengineering. Chem Biol Interact 2012; 200:92-113. [PMID: 23063744 DOI: 10.1016/j.cbi.2012.09.021] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2012] [Revised: 08/27/2012] [Accepted: 09/17/2012] [Indexed: 11/27/2022]
Abstract
The biological transformation of toxins as research probes, or as pharmaceutical drug leads, is an onerous and drawn out process. Issues regarding changes to pharmacological specificity, desired potency, and bioavailability are compounded naturally by their inherent toxicity. These often scuttle their progress as they move up the narrowing drug development pipeline. Yet one class of peptide toxins, from the genus Conus, has in many ways spearheaded the expansion of new peptide bioengineering techniques to aid peptide toxin pharmaceutical development. What has now emerged is the sequential bioengineering of new research probes and drug leads that owe their lineage to these highly potent and isoform specific peptides. Here we discuss the progressive bioengineering steps that many conopeptides have transitioned through, and specifically illustrate some of the biochemical approaches that have been established to maximize their biological research potential and pharmaceutical worth.
Collapse
Affiliation(s)
- Jon-Paul Bingham
- Department of Molecular Biosciences and Bioengineering, University of Hawaii, Honolulu, HI 96822, USA.
| | | | | | | |
Collapse
|
36
|
Gowd KH, Blais KD, Elmslie KS, Steiner AM, Olivera BM, Bulaj G. Dissecting a role of evolutionary-conserved but noncritical disulfide bridges in cysteine-rich peptides using ω-conotoxin GVIA and its selenocysteine analogs. Biopolymers 2012; 98:212-23. [PMID: 22782563 DOI: 10.1002/bip.22047] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Conotoxins comprise a large group of peptidic neurotoxins that use diverse disulfide-rich scaffolds. Each scaffold is determined by an evolutionarily conserved pattern of cysteine residues. Although many structure-activity relationship studies confirm the functional and structural importance of disulfide crosslinks, there is growing evidence that not all disulfide bridges are critical in maintaining activities of conotoxins. To answer the fundamental biological question of what the role of noncritical disulfide bridges is, we investigated function and folding of disulfide-depleted analogs of ω-conotoxin GVIA (GVIA) that belongs to an inhibitory cystine knot motif family and blocks N-type calcium channels. Removal of a noncritical Cys1-Cys16 disulfide bridge in GVIA or its selenopeptide analog had, as predicted, rather minimal effects on the inhibitory activity on calcium channels, as well as on in vivo activity following intracranial administration. However, the disulfide-depleted GVIA exhibited significantly lower folding yields for forming the remaining two native disulfide bridges. The disulfide-depleted selenoconotoxin GVIA analog also folded with significantly lower yields, suggesting that the functionally noncritical disulfide pair plays an important cooperative role in forming the native disulfide scaffold. Taken together, our results suggest that distinct disulfide bridges may be evolutionarily preserved by the oxidative folding or/and stabilization of the bioactive conformation of a disulfide-rich scaffold.
Collapse
|
37
|
Stevens M, Peigneur S, Dyubankova N, Lescrinier E, Herdewijn P, Tytgat J. Design of bioactive peptides from naturally occurring μ-conotoxin structures. J Biol Chem 2012; 287:31382-92. [PMID: 22773842 DOI: 10.1074/jbc.m112.375733] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
To date, cone snail toxins ("conotoxins") are of great interest in the pursuit of novel subtype-selective modulators of voltage-gated sodium channels (Na(v)s). Na(v)s participate in a wide range of electrophysiological processes. Consequently, their malfunctioning has been associated with numerous diseases. The development of subtype-selective modulators of Na(v)s remains highly important in the treatment of such disorders. In current research, a series of novel, synthetic, and bioactive compounds were designed based on two naturally occurring μ-conotoxins that target Na(v)s. The initial designed peptide contains solely 13 amino acids and was therefore named "Mini peptide." It was derived from the μ-conotoxins KIIIA and BuIIIC. Based on this Mini peptide, 10 analogues were subsequently developed, comprising 12-16 amino acids with two disulfide bridges. Following appropriate folding and mass verification, blocking effects on Na(v)s were investigated. The most promising compound established an IC(50) of 34.1 ± 0.01 nM (R2-Midi on Na(v)1.2). An NMR structure of one of our most promising compounds was determined. Surprisingly, this structure does not reveal an α-helix. We prove that it is possible to design small peptides based on known pharmacophores of μ-conotoxins without losing their potency and selectivity. These data can provide crucial material for further development of conotoxin-based therapeutics.
Collapse
Affiliation(s)
- Marijke Stevens
- Laboratory of Toxicology, Katholieke Universiteit (KU) Leuven, Campus Gasthuisberg O and N2, Herestraat 49 Box 922, 3000 Leuven, Belgium
| | | | | | | | | | | |
Collapse
|
38
|
Steiner AM, Woycechowsky KJ, Olivera BM, Bulaj G. Reagentless Oxidative Folding of Disulfide-Rich Peptides Catalyzed by an Intramolecular Diselenide. Angew Chem Int Ed Engl 2012. [DOI: 10.1002/ange.201200062] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
|
39
|
Steiner AM, Woycechowsky KJ, Olivera BM, Bulaj G. Reagentless oxidative folding of disulfide-rich peptides catalyzed by an intramolecular diselenide. Angew Chem Int Ed Engl 2012; 51:5580-4. [PMID: 22454362 DOI: 10.1002/anie.201200062] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2012] [Revised: 03/03/2012] [Indexed: 11/08/2022]
Affiliation(s)
- Andrew M Steiner
- Department of Medicinal Chemistry, University of Utah, 421 Wakara Way, Suite 360, Salt Lake City, UT 84108, USA
| | | | | | | |
Collapse
|
40
|
Khoo KK, Wilson MJ, Smith BJ, Zhang MM, Gulyas J, Yoshikami D, Rivier JE, Bulaj G, Norton RS. Lactam-stabilized helical analogues of the analgesic μ-conotoxin KIIIA. J Med Chem 2011; 54:7558-66. [PMID: 21962108 DOI: 10.1021/jm200839a] [Citation(s) in RCA: 52] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
μ-Conotoxin KIIIA (μ-KIIIA) blocks mammalian voltage-gated sodium channels (VGSCs) and is a potent analgesic following systemic administration in mice. Previous structure-activity studies of μ-KIIIA identified a helical pharmacophore for VGSC blockade. This suggested a route for designing truncated analogues of μ-KIIIA by incorporating the key residues into an α-helical scaffold. As (i, i+4) lactam bridges constitute a proven approach for stabilizing α-helices, we designed and synthesized six truncated analogues of μ-KIIIA containing single lactam bridges at various locations. The helicity of these lactam analogues was analyzed by NMR spectroscopy, and their activities were tested against mammalian VGSC subtypes Na(V)1.1 through 1.7. Two of the analogues, Ac-cyclo9/13[Asp9,Lys13]KIIIA7-14 and Ac-cyclo9/13[Lys9,Asp13]KIIIA7-14, displayed μM activity against VGSC subtypes Na(V)1.2 and Na(V)1.6; importantly, the subtype selectivity profile for these peptides matched that of μ-KIIIA. Our study highlights structure-activity relationships within these helical mimetics and provides a basis for the design of additional truncated peptides as potential analgesics.
Collapse
Affiliation(s)
- Keith K Khoo
- Monash Institute of Pharmaceutical Sciences, Monash University, Parkville Australia
| | | | | | | | | | | | | | | | | |
Collapse
|
41
|
McArthur JR, Singh G, McMaster D, Winkfein R, Tieleman DP, French RJ. Interactions of key charged residues contributing to selective block of neuronal sodium channels by μ-conotoxin KIIIA. Mol Pharmacol 2011; 80:573-84. [PMID: 21709136 DOI: 10.1124/mol.111.073460] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/14/2025] Open
Abstract
Voltage-gated sodium channels are important in initiating and propagating nerve impulses in various tissues, including cardiac muscle, skeletal muscle, the brain, and the peripheral nerves. Hyperexcitability of these channels leads to such disorders as cardiac arrhythmias (Na(v)1.5), myotonias (Na(v)1.4), epilepsies (Na(v)1.2), and pain (Na(v)1.7). Thus, there is strong motivation to identify isoform-specific blockers and the molecular determinants underlying their selectivity among these channels. μ-Conotoxin KIIIA blocks rNa(v)1.2 (IC(50), 5 nM), rNa(v)1.4 (37 nM), and hNa(v)1.7 (97 nM), expressed in mammalian cells, with high affinity and a maximal block at saturating concentrations of 90 to 95%. Mutations of charged residues on both the toxin and channel modulate the maximal block and/or affinity of KIIIA. Two toxin substitutions, K7A and R10A, modulate the maximal block (52-70%). KIIIA-H12A and R14A were the only derivatives tested that altered Na(v) isoform specificity. KIIIA-R14A showed the highest affinity for Na(v)1.7, a channel involved in pain signaling. Wild-type KIIIA has a 2-fold higher affinity for Na(v)1.4 than for Na(v)1.7, which can be attributed to a missing outer vestibule charge in domain III of Na(v)1.7. Reciprocal mutations Na(v)1.4 D1241I and Na(v)1.7 I1410D remove the affinity differences between these two channels for wild-type KIIIA without affecting their affinities for KIIIA-R14A. KIIIA is the first μ-conotoxin to show enhanced activity as pH is lowered, apparently resulting from titration of the free N terminus. Removal of this free amino group reduced the pH sensitivity by 10-fold. Recognition of these molecular determinants of KIIIA block may facilitate further development of subtype-specific, sodium channel blockers to treat hyperexcitability disorders.
Collapse
Affiliation(s)
- J R McArthur
- Department of Physiology and Pharmacology, and the Hotchkiss Brain Institute, Canada
| | | | | | | | | | | |
Collapse
|
42
|
Van Der Haegen A, Peigneur S, Tytgat J. Importance of position 8 in μ-conotoxin KIIIA for voltage-gated sodium channel selectivity. FEBS J 2011; 278:3408-18. [PMID: 21781281 DOI: 10.1111/j.1742-4658.2011.08264.x] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
μ-Conotoxin KIIIA from Conus kinoshitai is a 16-residue peptide that acts as a potent pore blocker of several voltage-gated sodium channels (Na(v)). In order to obtain more selective blockers and to investigate the role of Trp at position 8, we substituted this residue with Arg, Gln and Glu. KIIIA and analogues were tested on a range of Na(v) expressed in Xenopus laevis oocytes. The rank order of potency for KIIIA was: rNa(v)1.4 ≥ rNa(v)1.2 > mNa(v)1.6 > rNa(v)1.3, with IC(50) values of 48 ± 6 nm, 61 ± 5 nm, 183 ± 31 nm and 3.6 ± 0.3 μm, respectively, whereas no effect was seen on hNa(v)1.5 and hNa(v)1.8 at a concentration of 10 μm. Replacement of Trp8 resulted in more selective blockers with a preference for neuronal sodium channels over the skeletal sodium channel. The activity on rNa(v)1.4 was reduced about 40-, 70- and 200-fold for [W8R]KIIIA, [W8Q]KIIIA and [W8E]KIIIA, respectively. All analogues showed a completely reversible block of rNa(v)1.2, as opposed to the partial reversibility of KIIIA. At saturating concentrations, complete block of rNa(v)1.2 was never achieved. The residual current was lower than 10%, except for [W8E]KIIIA. KIIIA had no effect on the voltage dependence of activation of rNa(v)1.2, whereas all analogues caused a depolarizing shift. Overall, this study shows that Trp8 is a key residue in the pharmacophore. Replacement of Trp8 enables more selective blockers to be obtained for neuronal sodium channels. Trp is a key determinant for the reversibility of block of rNa(v)1.2.
Collapse
|
43
|
Empting M, Avrutina O, Meusinger R, Fabritz S, Reinwarth M, Biesalski M, Voigt S, Buntkowsky G, Kolmar H. "Triazole bridge": disulfide-bond replacement by ruthenium-catalyzed formation of 1,5-disubstituted 1,2,3-triazoles. Angew Chem Int Ed Engl 2011; 50:5207-11. [PMID: 21544910 DOI: 10.1002/anie.201008142] [Citation(s) in RCA: 107] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2010] [Revised: 02/21/2011] [Indexed: 12/20/2022]
Affiliation(s)
- Martin Empting
- Clemens-Schöpf-Institut für Organische Chemie und Biochemie, Technische Universität Darmstadt, Petersenstrasse 22, 64287 Darmstadt, Germany
| | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Empting M, Avrutina O, Meusinger R, Fabritz S, Reinwarth M, Biesalski M, Voigt S, Buntkowsky G, Kolmar H. “Triazolbrücke”: ein Disulfidbrückenersatz durch Ruthenium- katalysierte Bildung von 1,5-disubstituierten 1,2,3-Triazolen. Angew Chem Int Ed Engl 2011. [DOI: 10.1002/ange.201008142] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
|
45
|
Blunt JW, Copp BR, Munro MHG, Northcote PT, Prinsep MR. Marine natural products. Nat Prod Rep 2010; 28:196-268. [PMID: 21152619 DOI: 10.1039/c005001f] [Citation(s) in RCA: 343] [Impact Index Per Article: 22.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Affiliation(s)
- John W Blunt
- Department of Chemistry, University of Canterbury, Christchurch, New Zealand.
| | | | | | | | | |
Collapse
|
46
|
Zhang MM, Han TS, Olivera BM, Bulaj G, Yoshikami D. μ-conotoxin KIIIA derivatives with divergent affinities versus efficacies in blocking voltage-gated sodium channels. Biochemistry 2010; 49:4804-12. [PMID: 20459109 DOI: 10.1021/bi100207k] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
The possibility of independently manipulating the affinity and efficacy of pore-blocking ligands of sodium channels is of interest for the development of new drugs for the treatment of pain. The analgesic mu-conotoxin KIIIA (KIIIA), a 16-residue peptide with three disulfide bridges, is a pore blocker of voltage-gated sodium channels, including neuronal subtype Na(V)1.2 (K(d) = 5 nM). At saturating concentrations, KIIIA incompletely blocks the sodium current of Na(V)1.2, leaving a 5% residual current (rI(Na)). Lys7 is an important residue: the K7A mutation decreases both the efficacy (i.e., increases rI(Na) to 23%) and the affinity of the peptide (K(d) = 115 nM). In this report, various replacements of residue 7 were examined to determine whether affinity and efficacy were inexorably linked. Because of their facile chemical synthesis, KIIIA analogues that had as a core structure the disulfide-depleted KIIIA[C1A,C2U,C9A,C15U] (where U is selenocysteine) or ddKIIIA were used. Analogues ddKIIIA and ddKIIIA[K7X], where X represents one of nine different amino acids, were tested on voltage-clamped Xenopus oocytes expressing rat Na(V)1.2 or Na(V)1.4. Their affinities ranged from 0.01 to 36 muM and rI(Na) values from 2 to 42%, and these two variables appeared to be uncorrelated. Instead, rI(Na) varied inversely with side chain size, and remarkably charge and hydrophobicity appeared to be inconsequential. The ability to manipulate a mu-conopeptide's affinity and efficacy, as well as its capacity to interfere with subsequent tetrodotoxin binding, greatly expands its scope as a reagent for probing sodium channel structure and function and may also lead to the development of mu-conotoxins as safe analgesics.
Collapse
Affiliation(s)
- Min-Min Zhang
- Department of Biology, University of Utah, Salt Lake City, Utah 84112, USA
| | | | | | | | | |
Collapse
|
47
|
Han TS, Zhang MM, Gowd KH, Walewska A, Yoshikami D, Olivera BM, Bulaj G. Disulfide-Depleted Selenoconopeptides: a Minimalist Strategy to Oxidative Folding of Cysteine-Rich Peptides. ACS Med Chem Lett 2010; 1:140-144. [PMID: 20676359 DOI: 10.1021/ml900017q] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Despite the therapeutic promise of disulfide-rich, peptidic natural products, their discovery and structure/function studies have been hampered by inefficient oxidative folding methods for their synthesis. Here we report that converting the three disulfide-bridged mu-conopeptide KIIIA into a disulfide-depleted selenoconopeptide (by removal of a noncritical disulfide bridge and substitution of a disulfide- with a diselenide-bridge) dramatically simplified its oxidative folding while preserving the peptide's ability to block voltage-gated sodium channels. The simplicity of synthesizing disulfide-depleted selenopeptide analogs containing a single disulfide bridge allowed rapid positional scanning at Lys7 of mu-KIIIA, resulting in the identification of K7L as a mutation that improved the peptide's selectivity in blocking a neuronal (Na(v)1.2) over a muscle (Na(v)1.4) subtype of sodium channel. The disulfide-depleted selenopeptide strategy offers regioselective folding compatible with high throughput chemical synthesis and on-resin oxidation methods, and thus shows great promise to accelerate the use of disulfide-rich peptides as research tools and drugs.
Collapse
Affiliation(s)
- Tiffany S. Han
- Department of Biology, University of Utah, Salt Lake City, Utah, 84112
| | - Min-Min Zhang
- Department of Biology, University of Utah, Salt Lake City, Utah, 84112
| | | | - Aleksandra Walewska
- Department of Biology, University of Utah, Salt Lake City, Utah, 84112
- Faculty of Chemistry, University of Gdansk, 80-952 Gdansk, Poland
| | - Doju Yoshikami
- Department of Biology, University of Utah, Salt Lake City, Utah, 84112
| | | | - Grzegorz Bulaj
- Department of Medicinal Chemistry, University of Utah, Salt Lake City, Utah, 84108
| |
Collapse
|
48
|
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: 47] [Impact Index Per Article: 3.1] [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
|
49
|
Holford M, Auer S, Laqua M, Ibañez-Tallon I. Manipulating neuronal circuits with endogenous and recombinant cell-surface tethered modulators. Front Mol Neurosci 2009; 2:21. [PMID: 19915728 PMCID: PMC2776481 DOI: 10.3389/neuro.02.021.2009] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2009] [Accepted: 10/10/2009] [Indexed: 11/26/2022] Open
Abstract
Neuronal circuits depend on the precise regulation of cell-surface receptors and ion channels. An ongoing challenge in neuroscience research is deciphering the functional contribution of specific receptors and ion channels using engineered modulators. A novel strategy, termed “tethered toxins”, was recently developed to characterize neuronal circuits using the evolutionary derived selectivity of venom peptide toxins and endogenous peptide ligands, such as lynx1 prototoxins. Herein, the discovery and engineering of cell-surface tethered peptides is reviewed, with particular attention given to their cell-autonomy, modular composition, and genetic targeting in different model organisms. The relative ease with which tethered peptides can be engineered, coupled with the increasing number of neuroactive venom toxins and ligand peptides being discovered, imply a multitude of potentially innovative applications for manipulating neuronal circuits and tissue-specific cell networks, including treatment of disorders caused by malfunction of receptors and ion channels.
Collapse
Affiliation(s)
- Mandë Holford
- York College and The Graduate Center, The American Museum of Natural History, The City University of New York New York, NY, USA
| | | | | | | |
Collapse
|
50
|
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
|