1
|
Luo S, Zhou X, Wu M, Wang G, Wang L, Feng X, Wu H, Luo R, Lu M, Ju J, Wang W, Yuan L, Luo X, Peng D, Yang L, Zhang Q, Chen M, Liang S, Dong X, Hao G, Zhang Y, Liu Z. Optimizing Nav1.7-Targeted Analgesics: Revealing Off-Target Effects of Spider Venom-Derived Peptide Toxins and Engineering Strategies for Improvement. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2406656. [PMID: 39248322 PMCID: PMC11558128 DOI: 10.1002/advs.202406656] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/16/2024] [Revised: 08/22/2024] [Indexed: 09/10/2024]
Abstract
The inhibition of Nav1.7 is a promising strategy for the development of analgesic treatments. Spider venom-derived peptide toxins are recognized as significant sources of Nav1.7 inhibitors. However, their development has been impeded by limited selectivity. In this study, eight peptide toxins from three distinct spider venom Nav channel families demonstrated robust inhibition of hNav1.7, rKv4.2, and rKv4.3 (rKv4.2/4.3) currents, exhibiting a similar mode of action. The analysis of structure and function relationship revealed a significant overlap in the pharmacophore responsible for inhibiting hNav1.7 and rKv4.2 by HNTX-III, although Lys25 seems to play a more pivotal role in the inhibition of rKv4.2/4.3. Pharmacophore-guided rational design is employed for the development of an mGpTx1 analogue, mGpTx1-SA, which retains its inhibition of hNav1.7 while significantly reducing its inhibition of rKv4.2/4.3 and eliminating cardiotoxicity. Moreover, mGpTx1-SA demonstrates potent analgesic effects in both inflammatory and neuropathic pain models, accompanied by an improved in vivo safety profile. The results suggest that off-target inhibition of rKv4.2/4.3 by specific spider peptide toxins targeting hNav1.7 may arise from a conserved binding motif. This insight promises to facilitate the design of hNav1.7-specific analgesics, aimed at minimizing rKv4.2/4.3 inhibition and associated toxicity, thereby enhancing their suitability for therapeutic applications.
Collapse
|
2
|
Collaço RDC, Van Petegem F, Bosmans F. ω-Grammotoxin-SIA inhibits voltage-gated Na+ channel currents. J Gen Physiol 2024; 156:e202413563. [PMID: 39042091 PMCID: PMC11270453 DOI: 10.1085/jgp.202413563] [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: 02/09/2024] [Revised: 06/14/2024] [Accepted: 07/10/2024] [Indexed: 07/24/2024] Open
Abstract
ω-Grammotoxin-SIA (GrTX-SIA) was originally isolated from the venom of the Chilean rose tarantula and demonstrated to function as a gating modifier of voltage-gated Ca2+ (CaV) channels. Later experiments revealed that GrTX-SIA could also inhibit voltage-gated K+ (KV) channel currents via a similar mechanism of action that involved binding to a conserved S3-S4 region in the voltage-sensing domains (VSDs). Since voltage-gated Na+ (NaV) channels contain homologous structural motifs, we hypothesized that GrTX-SIA could inhibit members of this ion channel family as well. Here, we show that GrTX-SIA can indeed impede the gating process of multiple NaV channel subtypes with NaV1.6 being the most susceptible target. Moreover, molecular docking of GrTX-SIA onto NaV1.6, supported by a p.E1607K mutation, revealed the voltage sensor in domain IV (VSDIV) as being a primary site of action. The biphasic manner in which current inhibition appeared to occur suggested a second, possibly lower-sensitivity binding locus, which was identified as VSDII by using KV2.1/NaV1.6 chimeric voltage-sensor constructs. Subsequently, the NaV1.6p.E782K/p.E838K (VSDII), NaV1.6p.E1607K (VSDIV), and particularly the combined VSDII/VSDIV mutant lost virtually all susceptibility to GrTX-SIA. Together with existing literature, our data suggest that GrTX-SIA recognizes modules in NaV channel VSDs that are conserved among ion channel families, thereby allowing it to act as a comprehensive ion channel gating modifier peptide.
Collapse
Affiliation(s)
- Rita de Cássia Collaço
- Department of Basic and Applied Medical Sciences, Molecular Physiology and Neurophysics Group, Ghent University, Ghent, Belgium
| | - Filip Van Petegem
- Department of Biochemistry and Molecular Biology, University of British Columbia, Vancouver, Canada
- Life Sciences Institute, University of British Columbia, Vancouver, Canada
| | - Frank Bosmans
- Department of Basic and Applied Medical Sciences, Molecular Physiology and Neurophysics Group, Ghent University, Ghent, Belgium
- Faculty of Medicine and Pharmacy, Center for Neurosciences (C4N), Vrije Universiteit Brussel, Brussels, Belgium
| |
Collapse
|
3
|
Hiranuma M, Okuda Y, Fujii Y, Richard JP, Watanabe T. Characterization of human iPSC-derived sensory neurons and their functional assessment using multi electrode array. Sci Rep 2024; 14:6011. [PMID: 38472288 DOI: 10.1038/s41598-024-55602-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2023] [Accepted: 02/26/2024] [Indexed: 03/14/2024] Open
Abstract
Sensory neurons are afferent neurons in sensory systems that convert stimuli and transmit information to the central nervous system as electrical signals. Primary afferent neurons that are affected by non-noxious and noxious stimuli are present in the dorsal root ganglia (DRG), and the DRG sensory neurons are used as an in vitro model of the nociceptive response. However, DRG derived from mouse or rat give a low yield of neurons, and they are difficult to culture. To help alleviate this problem, we characterized human induced pluripotent stem cell (hiPSC) derived sensory neurons. They can solve the problems of interspecies differences and supply stability. We investigated expressions of sensory neuron related proteins and genes, and drug responses by Multi-Electrode Array (MEA) to analyze the properties and functions of sensory neurons. They expressed nociceptor, mechanoreceptor and proprioceptor related genes and proteins. They constitute a heterogeneous population of their subclasses. We confirmed that they could respond to both noxious and non-noxious stimuli. We showed that histamine inhibitors reduced histamine-induced neuronal excitability. Furthermore, incubation with a ProTx-II and Nav1.7 inhibitor reduced the spontaneous neural activity in hiPSC-derived sensory neurons. Their responsiveness was different from each drug. We have demonstrated that hiPSC-derived sensory neurons combined with MEA are good candidates for drug discovery studies where DRG in vitro modeling is necessary.
Collapse
|
4
|
Pan X, Li Y, Huang P, Staecker H, He M. Extracellular vesicles for developing targeted hearing loss therapy. J Control Release 2024; 366:460-478. [PMID: 38182057 DOI: 10.1016/j.jconrel.2023.12.050] [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: 10/12/2023] [Revised: 12/19/2023] [Accepted: 12/28/2023] [Indexed: 01/07/2024]
Abstract
Substantial efforts have been made for local administration of small molecules or biologics in treating hearing loss diseases caused by either trauma, genetic mutations, or drug ototoxicity. Recently, extracellular vesicles (EVs) naturally secreted from cells have drawn increasing attention on attenuating hearing impairment from both preclinical studies and clinical studies. Highly emerging field utilizing diverse bioengineering technologies for developing EVs as the bioderived therapeutic materials, along with artificial intelligence (AI)-based targeting toolkits, shed the light on the unique properties of EVs specific to inner ear delivery. This review will illuminate such exciting research field from fundamentals of hearing protective functions of EVs to biotechnology advancement and potential clinical translation of functionalized EVs. Specifically, the advancements in assessing targeting ligands using AI algorithms are systematically discussed. The overall translational potential of EVs is reviewed in the context of auditory sensing system for developing next generation gene therapy.
Collapse
Affiliation(s)
- Xiaoshu Pan
- Department of Pharmaceutics, College of Pharmacy, University of Florida, Gainesville, Florida 32610, United States
| | - Yanjun Li
- Department of Medicinal Chemistry, Center for Natural Products, Drug Discovery and Development, University of Florida, Gainesville, Florida 32610, United States
| | - Peixin Huang
- Department of Otolaryngology, Head and Neck Surgery, University of Kansas School of Medicine, Kansas City, Kansas 66160, United States
| | - Hinrich Staecker
- Department of Otolaryngology, Head and Neck Surgery, University of Kansas School of Medicine, Kansas City, Kansas 66160, United States.
| | - Mei He
- Department of Pharmaceutics, College of Pharmacy, University of Florida, Gainesville, Florida 32610, United States.
| |
Collapse
|
5
|
Antunes FTT, Campos MM, Carvalho VDPR, da Silva Junior CA, Magno LAV, de Souza AH, Gomez MV. Current Drug Development Overview: Targeting Voltage-Gated Calcium Channels for the Treatment of Pain. Int J Mol Sci 2023; 24:ijms24119223. [PMID: 37298174 DOI: 10.3390/ijms24119223] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2023] [Revised: 04/29/2023] [Accepted: 05/02/2023] [Indexed: 06/12/2023] Open
Abstract
Voltage-gated calcium channels (VGCCs) are targeted to treat pain conditions. Since the discovery of their relation to pain processing control, they are investigated to find new strategies for better pain control. This review provides an overview of naturally based and synthetic VGCC blockers, highlighting new evidence on the development of drugs focusing on the VGCC subtypes as well as mixed targets with pre-clinical and clinical analgesic effects.
Collapse
Affiliation(s)
- Flavia Tasmin Techera Antunes
- Department of Physiology and Pharmacology, University of Calgary, Calgary, AB T2N 1N4, Canada
- Hotchkiss Brain Institute, University of Calgary, Calgary, AB T2N 1N4, Canada
| | - Maria Martha Campos
- Programa de Pós-Graduação em Odontologia, Escola de Ciências da Saúde e da Vida, Pontifícia Universidade Católica do Rio Grande do Sul, Porto Alegre 90619-900, RS, Brazil
| | | | | | - Luiz Alexandre Viana Magno
- Programa de Pós-Graduação em Ciências da Saúde, Faculdade Ciências Médicas de Minas Gerais (FCMMG), Belo Horizonte 30110-005, MG, Brazil
| | - Alessandra Hubner de Souza
- Programa de Pós-Graduação em Ciências da Saúde, Faculdade Ciências Médicas de Minas Gerais (FCMMG), Belo Horizonte 30110-005, MG, Brazil
| | | |
Collapse
|
6
|
T-Type Calcium Channels: A Mixed Blessing. Int J Mol Sci 2022; 23:ijms23179894. [PMID: 36077291 PMCID: PMC9456242 DOI: 10.3390/ijms23179894] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2022] [Revised: 08/26/2022] [Accepted: 08/29/2022] [Indexed: 11/17/2022] Open
Abstract
The role of T-type calcium channels is well established in excitable cells, where they preside over action potential generation, automaticity, and firing. They also contribute to intracellular calcium signaling, cell cycle progression, and cell fate; and, in this sense, they emerge as key regulators also in non-excitable cells. In particular, their expression may be considered a prognostic factor in cancer. Almost all cancer cells express T-type calcium channels to the point that it has been considered a pharmacological target; but, as the drugs used to reduce their expression are not completely selective, several complications develop, especially within the heart. T-type calcium channels are also involved in a specific side effect of several anticancer agents, that act on microtubule transport, increase the expression of the channel, and, thus, the excitability of sensory neurons, and make the patient more sensitive to pain. This review puts into context the relevance of T-type calcium channels in cancer and in chemotherapy side effects, considering also the cardiotoxicity induced by new classes of antineoplastic molecules.
Collapse
|
7
|
McArthur JR, Wen J, Hung A, Finol-Urdaneta RK, Adams DJ. µ-Theraphotoxin Pn3a inhibition of Ca V3.3 channels reveals a novel isoform-selective drug binding site. eLife 2022; 11:e74040. [PMID: 35858123 PMCID: PMC9342953 DOI: 10.7554/elife.74040] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2021] [Accepted: 07/19/2022] [Indexed: 11/13/2022] Open
Abstract
Low voltage-activated calcium currents are mediated by T-type calcium channels CaV3.1, CaV3.2, and CaV3.3, which modulate a variety of physiological processes including sleep, cardiac pace-making, pain, and epilepsy. CaV3 isoforms' biophysical properties, overlapping expression, and lack of subtype-selective pharmacology hinder the determination of their specific physiological roles in health and disease. We have identified μ-theraphotoxin Pn3a as the first subtype-selective spider venom peptide inhibitor of CaV3.3, with >100-fold lower potency against the other T-type isoforms. Pn3a modifies CaV3.3 gating through a depolarizing shift in the voltage dependence of activation thus decreasing CaV3.3-mediated currents in the normal range of activation potentials. Paddle chimeras of KV1.7 channels bearing voltage sensor sequences from all four CaV3.3 domains revealed preferential binding of Pn3a to the S3-S4 region of domain II (CaV3.3DII). This novel T-type channel pharmacological site was explored through computational docking simulations of Pn3a, site-directed mutagenesis, and full domain II swaps between CaV3 channels highlighting it as a subtype-specific pharmacophore. This research expands our understanding of T-type calcium channel pharmacology and supports the suitability of Pn3a as a molecular tool in the study of the physiological roles of CaV3.3 channels.
Collapse
Affiliation(s)
- Jeffrey R McArthur
- Illawarra Health and Medical Research Institute, University of WollongongWollongongAustralia
| | - Jierong Wen
- School of Science, RMIT UniversityMelbourneAustralia
| | - Andrew Hung
- School of Science, RMIT UniversityMelbourneAustralia
| | - Rocio K Finol-Urdaneta
- Illawarra Health and Medical Research Institute, University of WollongongWollongongAustralia
| | - David J Adams
- Illawarra Health and Medical Research Institute, University of WollongongWollongongAustralia
| |
Collapse
|
8
|
Zhang YL, Moran SP, Allen A, Baez-Nieto D, Xu Q, Wang LA, Martenis WE, Sacher JR, Gale JP, Weïwer M, Wagner FF, Pan JQ. Novel Fluorescence-Based High-Throughput FLIPR Assay Utilizing Membrane-Tethered Genetic Calcium Sensors to Identify T-Type Calcium Channel Modulators. ACS Pharmacol Transl Sci 2022; 5:156-168. [PMID: 35311021 PMCID: PMC8923061 DOI: 10.1021/acsptsci.1c00233] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2021] [Indexed: 11/28/2022]
Abstract
T-type voltage-gated Ca2+ channels have been implicated in many human disorders, and there has been increasing interest in developing highly selective and potent T-type Ca2+ channel modulators for potential clinical use. However, the unique biophysical properties of T-type Ca2+ channels are not conducive for developing high-throughput screening (HTS) assays to identify modulators, particularly potentiators. To illustrate, T-type Ca2+ channels are largely inactivated and unable to open to allow Ca2+ influx at -25 mV, the typical resting membrane potential of the cell lines commonly used in cellular screening assays. To address this issue, we developed cell lines that express Kir2.3 channels to hyperpolarize the membrane potential to -70 mV, thus allowing T-type channels to return to their resting state where they can be subsequently activated by membrane depolarization in the presence of extracellular KCl. Furthermore, to simplify the HTS assay and to reduce reagent cost, we stably expressed a membrane-tethered genetic calcium sensor, GCaMP6s-CAAX, that displays superior signal to the background compared to the untethered GCaMP6s or the synthetic Ca2+ sensor Fluo-4AM. Here, we describe a novel GCaMP6s-CAAX-based calcium assay utilizing a high-throughput fluorometric imaging plate reader (Molecular Devices, Sunnyvale, CA) format that can identify both activators and inhibitors of T-type Ca2+ channels. Lastly, we demonstrate the utility of this novel fluorescence-based assay to evaluate the activities of two distinct G-protein-coupled receptors, thus expanding the use of GCaMP6s-CAAX to a wide range of applications relevant for developing cellular assays in drug discovery.
Collapse
Affiliation(s)
- Yan-Ling Zhang
- Stanley Center for Psychiatric
Research, Broad Institute of MIT and Harvard, Cambridge, Massachusetts 02142, United States
| | - Sean P. Moran
- Stanley Center for Psychiatric
Research, Broad Institute of MIT and Harvard, Cambridge, Massachusetts 02142, United States
| | - Andrew Allen
- Stanley Center for Psychiatric
Research, Broad Institute of MIT and Harvard, Cambridge, Massachusetts 02142, United States
| | - David Baez-Nieto
- Stanley Center for Psychiatric
Research, Broad Institute of MIT and Harvard, Cambridge, Massachusetts 02142, United States
| | - Qihong Xu
- Stanley Center for Psychiatric
Research, Broad Institute of MIT and Harvard, Cambridge, Massachusetts 02142, United States
| | - Lei A. Wang
- Stanley Center for Psychiatric
Research, Broad Institute of MIT and Harvard, Cambridge, Massachusetts 02142, United States
| | - William E. Martenis
- Stanley Center for Psychiatric
Research, Broad Institute of MIT and Harvard, Cambridge, Massachusetts 02142, United States
| | - Joshua R. Sacher
- Stanley Center for Psychiatric
Research, Broad Institute of MIT and Harvard, Cambridge, Massachusetts 02142, United States
| | - Jennifer P. Gale
- Stanley Center for Psychiatric
Research, Broad Institute of MIT and Harvard, Cambridge, Massachusetts 02142, United States
| | - Michel Weïwer
- Stanley Center for Psychiatric
Research, Broad Institute of MIT and Harvard, Cambridge, Massachusetts 02142, United States
| | - Florence F. Wagner
- Stanley Center for Psychiatric
Research, Broad Institute of MIT and Harvard, Cambridge, Massachusetts 02142, United States
| | - Jen Q. Pan
- Stanley Center for Psychiatric
Research, Broad Institute of MIT and Harvard, Cambridge, Massachusetts 02142, United States
| |
Collapse
|
9
|
Nguyen PT, Nguyen HM, Wagner KM, Stewart RG, Singh V, Thapa P, Chen YJ, Lillya MW, Ton AT, Kondo R, Ghetti A, Pennington MW, Hammock B, Griffith TN, Sack JT, Wulff H, Yarov-Yarovoy V. Computational design of peptides to target Na V1.7 channel with high potency and selectivity for the treatment of pain. eLife 2022; 11:81727. [PMID: 36576241 PMCID: PMC9831606 DOI: 10.7554/elife.81727] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2022] [Accepted: 12/23/2022] [Indexed: 12/29/2022] Open
Abstract
The voltage-gated sodium NaV1.7 channel plays a key role as a mediator of action potential propagation in C-fiber nociceptors and is an established molecular target for pain therapy. ProTx-II is a potent and moderately selective peptide toxin from tarantula venom that inhibits human NaV1.7 activation. Here we used available structural and experimental data to guide Rosetta design of potent and selective ProTx-II-based peptide inhibitors of human NaV1.7 channels. Functional testing of designed peptides using electrophysiology identified the PTx2-3127 and PTx2-3258 peptides with IC50s of 7 nM and 4 nM for hNaV1.7 and more than 1000-fold selectivity over human NaV1.1, NaV1.3, NaV1.4, NaV1.5, NaV1.8, and NaV1.9 channels. PTx2-3127 inhibits NaV1.7 currents in mouse and human sensory neurons and shows efficacy in rat models of chronic and thermal pain when administered intrathecally. Rationally designed peptide inhibitors of human NaV1.7 channels have transformative potential to define a new class of biologics to treat pain.
Collapse
Affiliation(s)
- Phuong T Nguyen
- Department of Physiology and Membrane Biology, University of California DavisDavisUnited States
| | - Hai M Nguyen
- Department of Pharmacology, University of California DavisDavisUnited States
| | - Karen M Wagner
- Department of Entomology and Nematology & Comprehensive Cancer Center, University of California DavisDavisUnited States
| | - Robert G Stewart
- Department of Physiology and Membrane Biology, University of California DavisDavisUnited States
| | - Vikrant Singh
- Department of Pharmacology, University of California DavisDavisUnited States
| | - Parashar Thapa
- Department of Physiology and Membrane Biology, University of California DavisDavisUnited States
| | - Yi-Je Chen
- Department of Pharmacology, University of California DavisDavisUnited States
| | - Mark W Lillya
- Department of Physiology and Membrane Biology, University of California DavisDavisUnited States
| | | | | | | | | | - Bruce Hammock
- Department of Entomology and Nematology & Comprehensive Cancer Center, University of California DavisDavisUnited States
| | - Theanne N Griffith
- Department of Physiology and Membrane Biology, University of California DavisDavisUnited States
| | - Jon T Sack
- Department of Physiology and Membrane Biology, University of California DavisDavisUnited States,Department of Anesthesiology and Pain Medicine, University of California DavisDavisUnited States
| | - Heike Wulff
- Department of Pharmacology, University of California DavisDavisUnited States
| | - Vladimir Yarov-Yarovoy
- Department of Physiology and Membrane Biology, University of California DavisDavisUnited States,Department of Anesthesiology and Pain Medicine, University of California DavisDavisUnited States,Biophysics Graduate Group, University of California DavisDavisUnited States
| |
Collapse
|
10
|
Diochot S. Pain-related toxins in scorpion and spider venoms: a face to face with ion channels. J Venom Anim Toxins Incl Trop Dis 2021; 27:e20210026. [PMID: 34925480 PMCID: PMC8667759 DOI: 10.1590/1678-9199-jvatitd-2021-0026] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2021] [Revised: 05/10/2021] [Indexed: 12/12/2022] Open
Abstract
Pain is a common symptom induced during envenomation by spiders and scorpions.
Toxins isolated from their venom have become essential tools for studying the
functioning and physiopathological role of ion channels, as they modulate their
activity. In particular, toxins that induce pain relief effects can serve as a
molecular basis for the development of future analgesics in humans. This review
provides a summary of the different scorpion and spider toxins that directly
interact with pain-related ion channels, with inhibitory or stimulatory effects.
Some of these toxins were shown to affect pain modalities in different animal
models providing information on the role played by these channels in the pain
process. The close interaction of certain gating-modifier toxins with membrane
phospholipids close to ion channels is examined along with molecular approaches
to improve selectivity, affinity or bioavailability in vivo for
therapeutic purposes.
Collapse
Affiliation(s)
- Sylvie Diochot
- Institut de Pharmacologie Moléculaire et Cellulaire (IPMC), Centre National de la Recherche Scientifique (CNRS) UMR 7275 et Université Côte d'Azur (UCA), 06560 Valbonne, France. Institut de Pharmacologie Moléculaire et Cellulaire Centre National de la Recherche Scientifique Université Côte d'Azur Valbonne France
| |
Collapse
|
11
|
Hu H, Mawlawi SE, Zhao T, Deuis JR, Jami S, Vetter I, Lewis RJ, Cardoso FC. Engineering of a Spider Peptide via Conserved Structure-Function Traits Optimizes Sodium Channel Inhibition In Vitro and Anti-Nociception In Vivo. Front Mol Biosci 2021; 8:742457. [PMID: 34621788 PMCID: PMC8490825 DOI: 10.3389/fmolb.2021.742457] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2021] [Accepted: 09/06/2021] [Indexed: 12/12/2022] Open
Abstract
Venom peptides are potent and selective modulators of voltage-gated ion channels that regulate neuronal function both in health and in disease. We previously identified the spider venom peptide Tap1a from the Venezuelan tarantula Theraphosa apophysis that targeted multiple voltage-gated sodium and calcium channels in visceral pain pathways and inhibited visceral mechano-sensing neurons contributing to irritable bowel syndrome. In this work, alanine scanning and domain activity analysis revealed Tap1a inhibited sodium channels by binding with nanomolar affinity to the voltage-sensor domain II utilising conserved structure-function features characteristic of spider peptides belonging to family NaSpTx1. In order to speed up the development of optimized NaV-targeting peptides with greater inhibitory potency and enhanced in vivo activity, we tested the hypothesis that incorporating residues identified from other optimized NaSpTx1 peptides into Tap1a could also optimize its potency for NaVs. Applying this approach, we designed the peptides Tap1a-OPT1 and Tap1a-OPT2 exhibiting significant increased potency for NaV1.1, NaV1.2, NaV1.3, NaV1.6 and NaV1.7 involved in several neurological disorders including acute and chronic pain, motor neuron disease and epilepsy. Tap1a-OPT1 showed increased potency for the off-target NaV1.4, while this off-target activity was absent in Tap1a-OPT2. This enhanced potency arose through a slowed off-rate mechanism. Optimized inhibition of NaV channels observed in vitro translated in vivo, with reversal of nocifensive behaviours in a murine model of NaV-mediated pain also enhanced by Tap1a-OPT. Molecular docking studies suggested that improved interactions within loops 3 and 4, and C-terminal of Tap1a-OPT and the NaV channel voltage-sensor domain II were the main drivers of potency optimization. Overall, the rationally designed peptide Tap1a-OPT displayed new and refined structure-function features which are likely the major contributors to its enhanced bioactive properties observed in vivo. This work contributes to the rapid engineering and optimization of potent spider peptides multi-targeting NaV channels, and the research into novel drugs to treat neurological diseases.
Collapse
Affiliation(s)
- H Hu
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD, Australia
| | - S E Mawlawi
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD, Australia
| | - T Zhao
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD, Australia
| | - J R Deuis
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD, Australia
| | - S Jami
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD, Australia
| | - I Vetter
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD, Australia.,School of Pharmacy, The University of Queensland, Brisbane, QLD, Australia
| | - R J Lewis
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD, Australia
| | - F C Cardoso
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD, Australia.,Centre for Innovations in Peptide and Protein Science, The University of Queensland, Brisbane, QLD, Australia
| |
Collapse
|
12
|
Depuydt AS, Rihon J, Cheneval O, Vanmeert M, Schroeder CI, Craik DJ, Lescrinier E, Peigneur S, Tytgat J. Cyclic Peptides as T-Type Calcium Channel Blockers: Characterization and Molecular Mapping of the Binding Site. ACS Pharmacol Transl Sci 2021; 4:1379-1389. [PMID: 34423272 DOI: 10.1021/acsptsci.1c00079] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2021] [Indexed: 01/18/2023]
Abstract
T-type calcium (CaV3) channels play a crucial role in the generation and propagation of action potentials in excitable cells and are considered potential drug targets for the treatment of neurological and cardiovascular diseases. Given the limited pharmacological repertoire for these channels, there is a great need for novel potent and selective CaV3 channel inhibitors. In this study, we used Xenopus oocytes to heterologously express CaV3.1 channels and characterized the interaction with a small cyclic peptide, PnCS1. Using molecular modeling, PnCS1 was docked into the cryo-electron microscopy structure of the human CaV3.1 channel and molecular dynamics were performed on the resultant complex. The binding site of the peptide was mapped with the involvement of critical amino acids located in the pore region and fenestrations of the channel. More specifically, we found that PnCS1 reclines in the central cavity of the pore domain of the CaV3.1 channel and resides stably between the selectivity filter and the intracellular gate, blocking the conduction pathway of the channel. Using Multiple Attribute Positional Scanning approaches, we developed a series of PnCS1 analogues. These analogues had a reduced level of inhibition, confirming the importance of specific residues and corroborating our modeling. In summary, functional studies of PnCS1 on the CaV3.1 channel combined with molecular dynamics results provide the basis for understanding the molecular interactions of PnCS1 with CaV3.1 and are fundamental to structure-based drug discovery for treating CaV3 channelopathies.
Collapse
Affiliation(s)
- Anne-Sophie Depuydt
- Toxicology and Pharmacology, University of Leuven (KU Leuven), Campus Gasthuisberg, Leuven 3000, Belgium
| | - Jérôme Rihon
- Laboratory of Medicinal Chemistry, Rega Institute for Medical Research, KU Leuven, Leuven 3000, Belgium
| | - Olivier Cheneval
- Institute for Molecular Bioscience, Australian Research Council Centre of Excellence for Innovations in Peptide and Protein Science, The University of Queensland, Brisbane, 4072 Queensland Australia
| | - Michiel Vanmeert
- Laboratory of Medicinal Chemistry, Rega Institute for Medical Research, KU Leuven, Leuven 3000, Belgium
| | - Christina I Schroeder
- Institute for Molecular Bioscience, Australian Research Council Centre of Excellence for Innovations in Peptide and Protein Science, The University of Queensland, Brisbane, 4072 Queensland Australia.,National Cancer Institute, National Institutes of Health, Frederick, Maryland 21702, United States
| | - 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, 4072 Queensland Australia
| | - Eveline Lescrinier
- Laboratory of Medicinal Chemistry, Rega Institute for Medical Research, KU Leuven, Leuven 3000, Belgium
| | - Steve Peigneur
- Toxicology and Pharmacology, University of Leuven (KU Leuven), Campus Gasthuisberg, Leuven 3000, Belgium
| | - Jan Tytgat
- Toxicology and Pharmacology, University of Leuven (KU Leuven), Campus Gasthuisberg, Leuven 3000, Belgium
| |
Collapse
|
13
|
Cardoso FC, Castro J, Grundy L, Schober G, Garcia-Caraballo S, Zhao T, Herzig V, King GF, Brierley SM, Lewis RJ. A spider-venom peptide with multitarget activity on sodium and calcium channels alleviates chronic visceral pain in a model of irritable bowel syndrome. Pain 2021; 162:569-581. [PMID: 32826759 DOI: 10.1097/j.pain.0000000000002041] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2020] [Accepted: 08/04/2020] [Indexed: 12/19/2022]
Abstract
ABSTRACT Chronic pain is a serious debilitating condition that affects ∼20% of the world's population. Currently available drugs fail to produce effective pain relief in many patients and have dose-limiting side effects. Several voltage-gated sodium (NaV) and calcium (CaV) channels are implicated in the etiology of chronic pain, particularly NaV1.1, NaV1.3, NaV1.7-NaV1.9, CaV2.2, and CaV3.2. Numerous NaV and CaV modulators have been described, but with few exceptions, they display poor potency and/or selectivity for pain-related channel subtypes. Here, we report the discovery and characterization of 2 novel tarantula-venom peptides (Tap1a and Tap2a) isolated from Theraphosa apophysis venom that modulate the activity of both NaV and CaV3 channels. Tap1a and Tap2a inhibited on-target NaV and CaV3 channels at nanomolar to micromolar concentrations and displayed moderate off-target selectivity for NaV1.6 and weak affinity for NaV1.4 and NaV1.5. The most potent inhibitor, Tap1a, nearly ablated neuronal mechanosensitivity in afferent fibers innervating the colon and the bladder, with in vivo intracolonic administration reversing colonic mechanical hypersensitivity in a mouse model of irritable bowel syndrome. These findings suggest that targeting a specific combination of NaV and CaV3 subtypes provides a novel route for treatment of chronic visceral pain.
Collapse
Affiliation(s)
- Fernanda C Cardoso
- Centre for Pain Research, Institute for Molecular Bioscience, The University of Queensland, St Lucia, Brisbane, Queensland, Australia
| | - Joel Castro
- Visceral Pain Research Group, College of Medicine and Public Health, Flinders Health and Medical Research Institute, Flinders University, South Australia, Australia
- Hopwood Centre for Neurobiology, Lifelong Health Theme, South Australian Health and Medical Research Institute (SAHMRI), North Terrace, Adelaide, South Australia, Australia
| | - Luke Grundy
- Visceral Pain Research Group, College of Medicine and Public Health, Flinders Health and Medical Research Institute, Flinders University, South Australia, Australia
- Hopwood Centre for Neurobiology, Lifelong Health Theme, South Australian Health and Medical Research Institute (SAHMRI), North Terrace, Adelaide, South Australia, Australia
| | - Gudrun Schober
- Visceral Pain Research Group, College of Medicine and Public Health, Flinders Health and Medical Research Institute, Flinders University, South Australia, Australia
- Hopwood Centre for Neurobiology, Lifelong Health Theme, South Australian Health and Medical Research Institute (SAHMRI), North Terrace, Adelaide, South Australia, Australia
| | - Sonia Garcia-Caraballo
- Visceral Pain Research Group, College of Medicine and Public Health, Flinders Health and Medical Research Institute, Flinders University, South Australia, Australia
- Hopwood Centre for Neurobiology, Lifelong Health Theme, South Australian Health and Medical Research Institute (SAHMRI), North Terrace, Adelaide, South Australia, Australia
| | - Tianjiao Zhao
- Centre for Pain Research, Institute for Molecular Bioscience, The University of Queensland, St Lucia, Brisbane, Queensland, Australia
| | - Volker Herzig
- Centre for Pain Research, Institute for Molecular Bioscience, The University of Queensland, St Lucia, Brisbane, Queensland, Australia
- School of Science and Engineering, University of the Sunshine Coast, Sippy Downs, Queensland, Australia
| | - Glenn F King
- Centre for Pain Research, Institute for Molecular Bioscience, The University of Queensland, St Lucia, Brisbane, Queensland, Australia
| | - Stuart M Brierley
- Visceral Pain Research Group, College of Medicine and Public Health, Flinders Health and Medical Research Institute, Flinders University, South Australia, Australia
- Hopwood Centre for Neurobiology, Lifelong Health Theme, South Australian Health and Medical Research Institute (SAHMRI), North Terrace, Adelaide, South Australia, Australia
| | - Richard J Lewis
- Centre for Pain Research, Institute for Molecular Bioscience, The University of Queensland, St Lucia, Brisbane, Queensland, Australia
| |
Collapse
|
14
|
McArthur JR, Munasinghe NR, Finol-Urdaneta RK, Adams DJ, Christie MJ. Spider Venom Peptide Pn3a Inhibition of Primary Afferent High Voltage-Activated Calcium Channels. Front Pharmacol 2021; 11:633679. [PMID: 33584315 PMCID: PMC7875911 DOI: 10.3389/fphar.2020.633679] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2020] [Accepted: 12/30/2020] [Indexed: 11/19/2022] Open
Abstract
Despite potently inhibiting the nociceptive voltage-gated sodium (Nav) channel, Nav1.7, µ-theraphotoxin Pn3a is antinociceptive only upon co-administration with sub-therapeutic opioid agonists, or by itself at doses >3,000-fold greater than its Nav1.7 IC50 by a yet undefined mechanism. Nav channels are structurally related to voltage-gated calcium (Cav) channels, Cav1 and Cav2. These channels mediate the high voltage-activated (HVA) calcium currents (ICa) that orchestrate synaptic transmission in nociceptive dorsal root ganglion (DRG) neurons and are fine-tuned by opioid receptor (OR) activity. Using whole-cell patch clamp recording, we found that Pn3a (10 µM) inhibits ∼55% of rat DRG neuron HVA-ICa and 60–80% of Cav1.2, Cav1.3, Cav2.1, and Cav2.2 mediated currents in HEK293 cells, with no inhibition of Cav2.3. As a major DRG ICa component, Cav2.2 inhibition by Pn3a (IC50 = 3.71 ± 0.21 µM) arises from an 18 mV hyperpolarizing shift in the voltage dependence of inactivation. We observed that co-application of Pn3a and µ-OR agonist DAMGO results in enhanced HVA-ICa inhibition in DRG neurons whereas co-application of Pn3a with the OR antagonist naloxone does not, underscoring HVA channels as shared targets of Pn3a and opioids. We provide evidence that Pn3a inhibits native and recombinant HVA Cavs at previously reportedly antinociceptive concentrations in animal pain models. We show additive modulation of DRG HVA-ICa by sequential application of low Pn3a doses and sub-therapeutic opioids ligands. We propose Pn3a's antinociceptive effects result, at least in part, from direct inhibition of HVA-ICa at high Pn3a doses, or through additive inhibition by low Pn3a and mild OR activation.
Collapse
Affiliation(s)
- Jeffrey R McArthur
- Illawarra Health and Medical Research Institute (IHMRI), University of Wollongong, Wollongong, NSW, Australia
| | - Nehan R Munasinghe
- Discipline of Pharmacology, University of Sydney, Sydney, NSW, Australia
| | - Rocio K Finol-Urdaneta
- Illawarra Health and Medical Research Institute (IHMRI), University of Wollongong, Wollongong, NSW, Australia.,Electrophysiology Facility for Cell Phenotyping and Drug Discovery, IHMRI, Wollongong, NSW, Australia
| | - David J Adams
- Illawarra Health and Medical Research Institute (IHMRI), University of Wollongong, Wollongong, NSW, Australia
| | | |
Collapse
|
15
|
Yousuf A, Sadeghi M, Adams DJ. Venom-Derived Peptides Inhibiting Voltage-Gated Sodium and Calcium Channels in Mammalian Sensory Neurons. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1349:3-19. [DOI: 10.1007/978-981-16-4254-8_1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
|
16
|
Abstract
The delivery of therapies to the cochlea is notoriously challenging. It is an organ protected by a number of barriers that need to be overcome in the drug delivery process. Additionally, there are multiple sites of possible damage within the cochlea. Despite the many potential sites of damage, acquired otologic insults preferentially damage a single location. While progress has been made in techniques for inner ear drug delivery, the current techniques remain non-specific and our ability to deliver therapies in a cell-specific manner are limited. Fortunately, there are proteins specific to various cell-types within the cochlea (e.g., hair cells, spiral ganglion cells, stria vascularis) that function as biomarkers of site-specific damage. These protein biomarkers have potential to serve as targets for cell-specific inner ear drug delivery. In this manuscript, we review the concept of biomarkers and targeted- inner ear drug delivery and the well-characterized protein biomarkers within each of the locations of interest within the cochlea. Our review will focus on targeted drug delivery in the setting of acquired otologic insults (e.g., ototoxicity, noise-induce hearing loss). The goal is not to discuss therapies to treat acquired otologic insults, rather, to establish potential concepts of how to deliver therapies in a targeted, cell-specific manner. Based on our review, it is clear that future of inner ear drug delivery is a discipline filled with potential that will require collaborative efforts among clinicians and scientists to optimize treatment of otologic insults. Graphical Abstract ![]()
Collapse
|
17
|
Characterisation of d-Conotoxin TxVIA as a Mammalian T-Type Calcium Channel Modulator. Mar Drugs 2020; 18:md18070343. [PMID: 32629781 PMCID: PMC7401249 DOI: 10.3390/md18070343] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2020] [Revised: 06/25/2020] [Accepted: 06/25/2020] [Indexed: 12/19/2022] Open
Abstract
The 27-amino acid (aa)-long d-conotoxin TxVIA, originally isolated from the mollusc-hunting cone snail Conus textile, slows voltage-gated sodium (NaV) channel inactivation in molluscan neurons, but its mammalian ion channel targets remain undetermined. In this study, we confirmed that TxVIA was inactive on mammalian NaV1.2 and NaV1.7 even at high concentrations (10 µM). Given the fact that invertebrate NaV channel and T-type calcium channels (CaV3.x) are evolutionarily related, we examined the possibility that TxVIA may act on CaV3.x. Electrophysiological characterisation of the native TxVIA on CaV3.1, 3.2 and 3.3 revealed that TxVIA preferentially inhibits CaV3.2 current (IC50 = 0.24 mM) and enhances CaV3.1 current at higher concentrations. In fish bioassays TxVIA showed little effect on zebrafish behaviours when injected intramuscular at 250 ng/100 mg fish. The binding sites for TxVIA at NaV1.7 and CaV3.1 revealed that their channel binding sites contained a common epitope.
Collapse
|
18
|
Cardoso FC. Multi-targeting sodium and calcium channels using venom peptides for the treatment of complex ion channels-related diseases. Biochem Pharmacol 2020; 181:114107. [PMID: 32579958 DOI: 10.1016/j.bcp.2020.114107] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2020] [Revised: 06/13/2020] [Accepted: 06/17/2020] [Indexed: 02/08/2023]
Abstract
Venom peptides are amongst the most exquisite group of bioactive molecules able to alter the normal physiology of organisms. These bioactive peptides penetrate tissues and blood vessels to encounter a number of receptors and ion channels to which they bind with high affinity and execute modulatory activities. Arachnid is the most diverse class of venomous animals often rich in peptides modulating voltage-gated sodium (NaV), calcium (CaV), and potassium (KV) channels. Spider venoms, in particular, contain potent and selective peptides targeting these channels, with a few displaying interesting multi-target properties for NaV and CaV channels underlying disease mechanisms such as in neuropathic pain, motor neuron disease and cancer. The elucidation of the pharmacology and structure-function properties of these venom peptides are invaluable for the development of effective drugs targeting NaV and CaV channels. This perspective discusses spider venom peptides displaying multi-target properties to modulate NaV and CaV channels in regard to their pharmacological features, structure-function relationships and potential to become the next generation of effective drugs to treat neurological disorders and other multi-ion channels related diseases.
Collapse
Affiliation(s)
- Fernanda C Cardoso
- Institute for Molecular Bioscience, The University of Queensland, 306 Carmody Rd., St Lucia, QLD AU 4072, Australia
| |
Collapse
|
19
|
Mulcahy JV, Pajouhesh H, Beckley JT, Delwig A, Bois JD, Hunter JC. Challenges and Opportunities for Therapeutics Targeting the Voltage-Gated Sodium Channel Isoform Na V1.7. J Med Chem 2019; 62:8695-8710. [PMID: 31012583 PMCID: PMC6786914 DOI: 10.1021/acs.jmedchem.8b01906] [Citation(s) in RCA: 58] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Voltage-gated sodium ion channel subtype 1.7 (NaV1.7) is a high interest target for the discovery of non-opioid analgesics. Compelling evidence from human genetic data, particularly the finding that persons lacking functional NaV1.7 are insensitive to pain, has spurred considerable effort to develop selective inhibitors of this Na+ ion channel target as analgesic medicines. Recent clinical setbacks and disappointing performance of preclinical compounds in animal pain models, however, have led to skepticism around the potential of selective NaV1.7 inhibitors as human therapeutics. In this Perspective, we discuss the attributes and limitations of recently disclosed investigational drugs targeting NaV1.7 and review evidence that, by better understanding the requirements for selectivity and target engagement, the opportunity to deliver effective analgesic medicines targeting NaV1.7 endures.
Collapse
Affiliation(s)
- John V. Mulcahy
- SiteOne Therapeutics, 280 Utah Ave, Suite 250, South San Francisco, CA 94080
| | - Hassan Pajouhesh
- SiteOne Therapeutics, 280 Utah Ave, Suite 250, South San Francisco, CA 94080
| | - Jacob T. Beckley
- SiteOne Therapeutics, 351 Evergreen Drive, Suite B1, Bozeman, MT 59715
| | - Anton Delwig
- SiteOne Therapeutics, 280 Utah Ave, Suite 250, South San Francisco, CA 94080
| | - J. Du Bois
- Stanford University, Lokey Chemistry and Biology, 337 Campus Drive, Stanford, CA 94305
| | - John C. Hunter
- SiteOne Therapeutics, 280 Utah Ave, Suite 250, South San Francisco, CA 94080
| |
Collapse
|
20
|
Saez NJ, Herzig V. Versatile spider venom peptides and their medical and agricultural applications. Toxicon 2018; 158:109-126. [PMID: 30543821 DOI: 10.1016/j.toxicon.2018.11.298] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2018] [Revised: 11/12/2018] [Accepted: 11/14/2018] [Indexed: 02/07/2023]
Abstract
Spiders have been evolving complex and diverse repertoires of peptides in their venoms with vast pharmacological activities for more than 300 million years. Spiders use their venoms for prey capture and defense, hence they contain peptides that target both prey (mainly arthropods) and predators (other arthropods or vertebrates). This includes peptides that potently and selectively modulate a range of targets such as ion channels, receptors and signaling pathways involved in physiological processes. The contribution of these targets in particular disease pathophysiologies makes spider venoms a valuable source of peptides with potential therapeutic use. In addition, peptides with insecticidal activities, used for prey capture, can be exploited for the development of novel bioinsecticides for agricultural use. Although we have already reviewed potential applications of spider venom peptides as therapeutics (in 2010) and as bioinsecticides (in 2012), a considerable number of research articles on both topics have been published since, warranting an updated review. Here we explore the most recent research on the use of spider venom peptides for both medical and agricultural applications.
Collapse
Affiliation(s)
- Natalie J Saez
- Institute for Molecular Bioscience, The University of Queensland, St. Lucia, QLD 4072, Australia.
| | - Volker Herzig
- Institute for Molecular Bioscience, The University of Queensland, St. Lucia, QLD 4072, Australia.
| |
Collapse
|
21
|
Wang D, Ragnarsson L, Lewis RJ. T-type Calcium Channels in Health and Disease. Curr Med Chem 2018; 27:3098-3122. [PMID: 30277145 DOI: 10.2174/0929867325666181001112821] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2018] [Revised: 08/28/2018] [Accepted: 08/30/2018] [Indexed: 12/12/2022]
Abstract
Low Voltage-Activated (LVA) T-type calcium channels are characterized by transient current and Low Threshold Spikes (LTS) that trigger neuronal firing and oscillatory behavior. Combined with their preferential localization in dendrites and their specific "window current", T-type calcium channels are considered to be key players in signal amplification and synaptic integration. Assisted by the emerging pharmacological tools, the structural determinants of channel gating and kinetics, as well as novel physiological and pathological functions of T-type calcium channels, are being uncovered. In this review, we provide an overview of structural determinants in T-type calcium channels, their involvement in disorders and diseases, the development of novel channel modulators, as well as Structure-Activity Relationship (SAR) studies that lead to rational drug design.
Collapse
Affiliation(s)
- Dan Wang
- Division of Chemistry and Structural Biology, Institute for Molecular Bioscience, the University of Queensland, Brisbane Qld 4072, Australia
| | - Lotten Ragnarsson
- Division of Chemistry and Structural Biology, Institute for Molecular Bioscience, the University of Queensland, Brisbane Qld 4072, Australia
| | - Richard J Lewis
- Division of Chemistry and Structural Biology, Institute for Molecular Bioscience, the University of Queensland, Brisbane Qld 4072, Australia
| |
Collapse
|
22
|
Toxins as tools: Fingerprinting neuronal pharmacology. Neurosci Lett 2018; 679:4-14. [DOI: 10.1016/j.neulet.2018.02.001] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2017] [Revised: 01/09/2018] [Accepted: 02/02/2018] [Indexed: 12/30/2022]
|
23
|
|
24
|
Cav3.1 overexpression is associated with negative characteristics and prognosis in non-small cell lung cancer. Oncotarget 2018; 9:8573-8583. [PMID: 29492218 PMCID: PMC5823575 DOI: 10.18632/oncotarget.24194] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2017] [Accepted: 12/05/2017] [Indexed: 11/25/2022] Open
Abstract
Introduction Voltage-gated calcium channels (VGCC) have been found to be differentially expressed in several different tumor types, but their role in tumor growth, malignant invasion, metastases and impact on clinical outcomes has not been clarified. Materials and Methods From a cohort database of 193 patients with early-stage NSCLC, 163 formalin-fixed paraffin-embedded specimens were available for analysis to construct tissue microarrays. Cav3.1 protein expression was detected using fluorescence immunohistochemistry, and quantified using automated image acquisition and analysis. Results Among the cohort of 193 NSCLC patients, adenocarcinoma (53.9%) and squamous cell carcinoma (SCC) (30.1%) were the most common histologies. There was no difference between SCC and non-SCC subtypes in overall survival (OS) or relapse-free survival (RFS); 74.2 vs 90.1 months (p = 0.543) and 48.8 vs 52.6 months (p = 0.766), respectively. T-type VGCC 3.1 (Cav3.1) overexpression was assessed by tissue microarray immunohistochemistry analysis from 163 available patient samples. Eighteen (11.0%) NSCLC primaries were found to have Cav3.1 overexpression levels, and were significantly associated with SCC histology (p < 0.001), larger tumor size (p < 0.001) and later stage disease at diagnosis (p = 0.019). Median OS was 48.6 vs 106.7 months for Cav3.1 overexpressing and non-overexpressing patients, respectively (p = 0.032). Regression analysis revealed a significantly negative effect for Cav3.1 overexpression on RFS (Hazard ratio [HR] = 2.02, p = 0.048). Conclusions Cav3.1 overexpression is a potential biomarker for poorer patient outcomes. These results bring supportive evidence for calcium channels inducing an aggressive phenotype in NSCLC and potentially may serve as a therapeutic target in overexpressing tumors.
Collapse
|
25
|
Israel MR, Tay B, Deuis JR, Vetter I. Sodium Channels and Venom Peptide Pharmacology. ADVANCES IN PHARMACOLOGY (SAN DIEGO, CALIF.) 2017; 79:67-116. [PMID: 28528674 DOI: 10.1016/bs.apha.2017.01.004] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/15/2023]
Abstract
Venomous animals including cone snails, spiders, scorpions, anemones, and snakes have evolved a myriad of components in their venoms that target the opening and/or closing of voltage-gated sodium channels to cause devastating effects on the neuromuscular systems of predators and prey. These venom peptides, through design and serendipity, have not only contributed significantly to our understanding of sodium channel pharmacology and structure, but they also represent some of the most phyla- and isoform-selective molecules that are useful as valuable tool compounds and drug leads. Here, we review our understanding of the basic function of mammalian voltage-gated sodium channel isoforms as well as the pharmacology of venom peptides that act at these key transmembrane proteins.
Collapse
Affiliation(s)
- Mathilde R Israel
- Centre for Pain Research, Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD, Australia
| | - Bryan Tay
- Centre for Pain Research, Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD, Australia
| | - Jennifer R Deuis
- Centre for Pain Research, Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD, Australia.
| | - Irina Vetter
- Centre for Pain Research, Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD, Australia; School of Pharmacy, The University of Queensland, Brisbane, QLD, Australia.
| |
Collapse
|
26
|
Bourinet E, Zamponi GW. Block of voltage-gated calcium channels by peptide toxins. Neuropharmacology 2016; 127:109-115. [PMID: 27756538 DOI: 10.1016/j.neuropharm.2016.10.016] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2016] [Revised: 10/14/2016] [Accepted: 10/15/2016] [Indexed: 12/26/2022]
Abstract
Venoms from various predatory species, such as fish hunting molluscs scorpions, snakes and arachnids contain a large spectrum of toxins that include blockers of voltage-gated calcium channels. These peptide blockers act by two principal manners - physical occlusion of the pore and prevention of activation gating. Many of the calcium channel-blocking peptides have evolved to tightly occupy their binding pocket on the principal pore forming subunit of the channel, often rendering block poorly reversible. Moreover, several of the best characterized blocking peptides have developed a high degree of channel subtype selectivity. Here we give an overview of different types of calcium channel-blocking toxins, their mechanism of action, channel subtype specificity, and potential use as therapeutic agents. This article is part of the Special Issue entitled 'Venom-derived Peptides as Pharmacological Tools.'
Collapse
Affiliation(s)
- Emmanuel Bourinet
- Institute for Functional Genomics, CNRS UMR5203, INSERM U1191, University of Montpellier, LABEX ICST, Montpellier, France
| | - Gerald W Zamponi
- Department of Physiology and Pharmacology, Hotchkiss Brain Institute and Alberta Children's Hospital Research Institute, Cumming School of Medicine, University of Calgary, Calgary, Canada.
| |
Collapse
|
27
|
Salari A, Vega BS, Milescu LS, Milescu M. Molecular Interactions between Tarantula Toxins and Low-Voltage-Activated Calcium Channels. Sci Rep 2016; 6:23894. [PMID: 27045173 PMCID: PMC4820701 DOI: 10.1038/srep23894] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2015] [Accepted: 03/16/2016] [Indexed: 01/26/2023] Open
Abstract
Few gating-modifier toxins have been reported to target low-voltage-activated (LVA) calcium channels, and the structural basis of toxin sensitivity remains incompletely understood. Studies of voltage-gated potassium (Kv) channels have identified the S3b–S4 “paddle motif,” which moves at the protein-lipid interface to drive channel opening, as the target for these amphipathic neurotoxins. Voltage-gated calcium (Cav) channels contain four homologous voltage sensor domains, suggesting multiple toxin binding sites. We show here that the S3–S4 segments within Cav3.1 can be transplanted into Kv2.1 to examine their individual contributions to voltage sensing and pharmacology. With these results, we now have a more complete picture of the conserved nature of the paddle motif in all three major voltage-gated ion channel types (Kv, Nav, and Cav). When screened with tarantula toxins, the four paddle sequences display distinct toxin binding properties, demonstrating that gating-modifier toxins can bind to Cav channels in a domain specific fashion. Domain III was the most commonly and strongly targeted, and mutagenesis revealed an acidic residue that is important for toxin binding. We also measured the lipid partitioning strength of all toxins tested and observed a positive correlation with their inhibition of Cav3.1, suggesting a key role for membrane partitioning.
Collapse
Affiliation(s)
- Autoosa Salari
- University of Missouri, Division of Biological Sciences, Columbia, 65211, USA
| | - Benjamin S Vega
- University of Missouri, Division of Biological Sciences, Columbia, 65211, USA
| | - Lorin S Milescu
- University of Missouri, Division of Biological Sciences, Columbia, 65211, USA
| | - Mirela Milescu
- University of Missouri, Division of Biological Sciences, Columbia, 65211, USA
| |
Collapse
|
28
|
Deplazes E, Henriques ST, Smith JJ, King GF, Craik DJ, Mark AE, Schroeder CI. Membrane-binding properties of gating modifier and pore-blocking toxins: Membrane interaction is not a prerequisite for modification of channel gating. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2016; 1858:872-82. [DOI: 10.1016/j.bbamem.2016.02.002] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/04/2015] [Revised: 01/21/2016] [Accepted: 02/01/2016] [Indexed: 12/30/2022]
|
29
|
Endostatin is protective against monocrotaline-induced right heart disease through the inhibition of T-type Ca(2+) channel. Pflugers Arch 2016; 468:1259-1270. [PMID: 27023352 DOI: 10.1007/s00424-016-1810-0] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2015] [Revised: 03/03/2016] [Accepted: 03/15/2016] [Indexed: 12/27/2022]
Abstract
Endostatin (ES), a C-terminal fragment of collagen XVIIIα1, has a potent anti-angiogenic effect. ES prevents tumor proliferation through inhibiting T-type Ca(2+) channel. T-type Ca(2+) channel is re-expressed during heart diseases including monocrotaline (MCT)-induced right heart failure. The present study aimed to clarify the effects of ES on T-type Ca(2+) channel and pathogenesis of MCT-induced right ventricular disease. MCT or saline was injected intraperitoneally to rats. After cardiomyocytes were isolated from right ventricles (RVs), T-type Ca(2+) channel current (I CaT) was measured by a patch-clamp method. After ES small interfering RNA (siRNA) or control siRNA (20 μg) was administrated for 1 week via the right jugular vein 1 week after MCT injection, echocardiography and histological analysis were done. I CaT was significantly increased in RV from MCT-injected rats, and ES significantly inhibited it. The survival rate of ES siRNA-administrated MCT rats (MCT ES si group) was decreased. In echocardiography, although ES siRNA did not affect pulmonary arterial pressure, RV systolic function was impaired in MCT ES si group compared with control siRNA-administrated MCT rats (MCT cont si group). In the histological analysis of RV, ES expression was increased in MCT cont si group, and ES siRNA inhibited it. Furthermore, although MCT cont si group showed only cardiomyocyte hypertrophy, MCT ES si group showed notable enlargement of intercellular spaces. The present study for the first time revealed that ES inhibits T-type Ca(2+) channel activity in RV from MCT-injected rats. ES gene knockdown deteriorates MCT-induced right heart disease. ES is thus cardioprotective possibly through inhibiting T-type Ca(2+) channel activity.
Collapse
|
30
|
Barbosa C, Cummins TR. Unusual Voltage-Gated Sodium Currents as Targets for Pain. CURRENT TOPICS IN MEMBRANES 2016; 78:599-638. [PMID: 27586296 DOI: 10.1016/bs.ctm.2015.12.005] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Pain is a serious health problem that impacts the lives of many individuals. Hyperexcitability of peripheral sensory neurons contributes to both acute and chronic pain syndromes. Because voltage-gated sodium currents are crucial to the transmission of electrical signals in peripheral sensory neurons, the channels that underlie these currents are attractive targets for pain therapeutics. Sodium currents and channels in peripheral sensory neurons are complex. Multiple-channel isoforms contribute to the macroscopic currents in nociceptive sensory neurons. These different isoforms exhibit substantial variations in their kinetics and pharmacology. Furthermore, sodium current complexity is enhanced by an array of interacting proteins that can substantially modify the properties of voltage-gated sodium channels. Resurgent sodium currents, atypical currents that can enhance recovery from inactivation and neuronal firing, are increasingly being recognized as playing potentially important roles in sensory neuron hyperexcitability and pain sensations. Here we discuss unusual sodium channels and currents that have been identified in nociceptive sensory neurons, describe what is known about the molecular determinants of the complex sodium currents in these neurons. Finally, we provide an overview of therapeutic strategies to target voltage-gated sodium currents in nociceptive neurons.
Collapse
Affiliation(s)
- C Barbosa
- Indiana University School of Medicine, Indianapolis, IN, United States
| | - T R Cummins
- Indiana University School of Medicine, Indianapolis, IN, United States
| |
Collapse
|
31
|
Zamponi GW, Striessnig J, Koschak A, Dolphin AC. The Physiology, Pathology, and Pharmacology of Voltage-Gated Calcium Channels and Their Future Therapeutic Potential. Pharmacol Rev 2015; 67:821-70. [PMID: 26362469 PMCID: PMC4630564 DOI: 10.1124/pr.114.009654] [Citation(s) in RCA: 764] [Impact Index Per Article: 76.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Voltage-gated calcium channels are required for many key functions in the body. In this review, the different subtypes of voltage-gated calcium channels are described and their physiologic roles and pharmacology are outlined. We describe the current uses of drugs interacting with the different calcium channel subtypes and subunits, as well as specific areas in which there is strong potential for future drug development. Current therapeutic agents include drugs targeting L-type Ca(V)1.2 calcium channels, particularly 1,4-dihydropyridines, which are widely used in the treatment of hypertension. T-type (Ca(V)3) channels are a target of ethosuximide, widely used in absence epilepsy. The auxiliary subunit α2δ-1 is the therapeutic target of the gabapentinoid drugs, which are of value in certain epilepsies and chronic neuropathic pain. The limited use of intrathecal ziconotide, a peptide blocker of N-type (Ca(V)2.2) calcium channels, as a treatment of intractable pain, gives an indication that these channels represent excellent drug targets for various pain conditions. We describe how selectivity for different subtypes of calcium channels (e.g., Ca(V)1.2 and Ca(V)1.3 L-type channels) may be achieved in the future by exploiting differences between channel isoforms in terms of sequence and biophysical properties, variation in splicing in different target tissues, and differences in the properties of the target tissues themselves in terms of membrane potential or firing frequency. Thus, use-dependent blockers of the different isoforms could selectively block calcium channels in particular pathologies, such as nociceptive neurons in pain states or in epileptic brain circuits. Of important future potential are selective Ca(V)1.3 blockers for neuropsychiatric diseases, neuroprotection in Parkinson's disease, and resistant hypertension. In addition, selective or nonselective T-type channel blockers are considered potential therapeutic targets in epilepsy, pain, obesity, sleep, and anxiety. Use-dependent N-type calcium channel blockers are likely to be of therapeutic use in chronic pain conditions. Thus, more selective calcium channel blockers hold promise for therapeutic intervention.
Collapse
Affiliation(s)
- Gerald W Zamponi
- Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada (G.W.Z.); Department of Pharmacology and Toxicology, Institute of Pharmacy, Center for Molecular Biosciences, University of Innsbruck, Innsbruck, Austria (J.S., A.K.); and Department of Neuroscience, Physiology, and Pharmacology, Division of Biosciences, University College London, London, United Kingdom (A.C.D.)
| | - Joerg Striessnig
- Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada (G.W.Z.); Department of Pharmacology and Toxicology, Institute of Pharmacy, Center for Molecular Biosciences, University of Innsbruck, Innsbruck, Austria (J.S., A.K.); and Department of Neuroscience, Physiology, and Pharmacology, Division of Biosciences, University College London, London, United Kingdom (A.C.D.)
| | - Alexandra Koschak
- Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada (G.W.Z.); Department of Pharmacology and Toxicology, Institute of Pharmacy, Center for Molecular Biosciences, University of Innsbruck, Innsbruck, Austria (J.S., A.K.); and Department of Neuroscience, Physiology, and Pharmacology, Division of Biosciences, University College London, London, United Kingdom (A.C.D.)
| | - Annette C Dolphin
- Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada (G.W.Z.); Department of Pharmacology and Toxicology, Institute of Pharmacy, Center for Molecular Biosciences, University of Innsbruck, Innsbruck, Austria (J.S., A.K.); and Department of Neuroscience, Physiology, and Pharmacology, Division of Biosciences, University College London, London, United Kingdom (A.C.D.)
| |
Collapse
|
32
|
Stephens RF, Guan W, Zhorov BS, Spafford JD. Selectivity filters and cysteine-rich extracellular loops in voltage-gated sodium, calcium, and NALCN channels. Front Physiol 2015; 6:153. [PMID: 26042044 PMCID: PMC4436565 DOI: 10.3389/fphys.2015.00153] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2015] [Accepted: 04/28/2015] [Indexed: 12/19/2022] Open
Abstract
How nature discriminates sodium from calcium ions in eukaryotic channels has been difficult to resolve because they contain four homologous, but markedly different repeat domains. We glean clues from analyzing the changing pore region in sodium, calcium and NALCN channels, from single-cell eukaryotes to mammals. Alternative splicing in invertebrate homologs provides insights into different structural features underlying calcium and sodium selectivity. NALCN generates alternative ion selectivity with splicing that changes the high field strength (HFS) site at the narrowest level of the hourglass shaped pore where the selectivity filter is located. Alternative splicing creates NALCN isoforms, in which the HFS site has a ring of glutamates contributed by all four repeat domains (EEEE), or three glutamates and a lysine residue in the third (EEKE) or second (EKEE) position. Alternative splicing provides sodium and/or calcium selectivity in T-type channels with extracellular loops between S5 and P-helices (S5P) of different lengths that contain three or five cysteines. All eukaryotic channels have a set of eight core cysteines in extracellular regions, but the T-type channels have an infusion of 4–12 extra cysteines in extracellular regions. The pattern of conservation suggests a possible pairing of long loops in Domains I and III, which are bridged with core cysteines in NALCN, Cav, and Nav channels, and pairing of shorter loops in Domains II and IV in T-type channel through disulfide bonds involving T-type specific cysteines. Extracellular turrets of increasing lengths in potassium channels (Kir2.2, hERG, and K2P1) contribute to a changing landscape above the pore selectivity filter that can limit drug access and serve as an ion pre-filter before ions reach the pore selectivity filter below. Pairing of extended loops likely contributes to the large extracellular appendage as seen in single particle electron cryo-microscopy images of the eel Nav1 channel.
Collapse
Affiliation(s)
| | - W Guan
- Department of Biology, University of Waterloo Waterloo, ON, Canada
| | - Boris S Zhorov
- Department of Biochemistry and Biomedical Sciences, McMaster University Hamilton, ON, Canada ; Sechenov Institute of Evolutionary Physiology and Biochemistry, Russian Academy of Sciences St. Petersburg, Russia
| | - J David Spafford
- Department of Biology, University of Waterloo Waterloo, ON, Canada
| |
Collapse
|
33
|
Yasuda J, Okada M, Yamawaki H. Endostatin inhibits T-type Ca2+ channel current in guinea pig ventricular myocyte. J Vet Med Sci 2015; 77:1289-91. [PMID: 25947888 PMCID: PMC4638298 DOI: 10.1292/jvms.14-0551] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Endostatin, a fragment of collagen XVIII, is known as an endogenous angiogenesis
inhibitor, and its serum concentration increases in various cardiovascular diseases.
T-type Ca2+ channel, low voltage-activated Ca2+ channel, is not
expressed in adult ventricular myocytes. Re-expression of T-type Ca2+ channels
in cardiac myocytes is thought to be involved in the development of cardiac hypertrophy.
We examined the effects of endostatin on T-type Ca2+ channel current by
whole-cell patch clamp technique in freshly isolated adult guinea pig ventricular
myocytes, which exceptionally express T-type Ca2+ channels. Although endostatin
300 ng/ml had no effect on L-type Ca2+
current, it significantly inhibited T-type Ca2+ current. These data indicate
that endostatin can be an endogenous inhibitor of T-type Ca2+ channels in the
cardiac myocytes.
Collapse
Affiliation(s)
- Jumpei Yasuda
- Laboratory of Veterinary Pharmacology, School of Veterinary Medicine, Kitasato University, Higashi 23 bancho 35-1, Towada-shi, Aomori 034-8628, Japan
| | | | | |
Collapse
|
34
|
Berkut AA, Peigneur S, Myshkin MY, Paramonov AS, Lyukmanova EN, Arseniev AS, Grishin EV, Tytgat J, Shenkarev ZO, Vassilevski AA. Structure of membrane-active toxin from crab spider Heriaeus melloteei suggests parallel evolution of sodium channel gating modifiers in Araneomorphae and Mygalomorphae. J Biol Chem 2014; 290:492-504. [PMID: 25352595 DOI: 10.1074/jbc.m114.595678] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
We present a structural and functional study of a sodium channel activation inhibitor from crab spider venom. Hm-3 is an insecticidal peptide toxin consisting of 35 amino acid residues from the spider Heriaeus melloteei (Thomisidae). We produced Hm-3 recombinantly in Escherichia coli and determined its structure by NMR spectroscopy. Typical for spider toxins, Hm-3 was found to adopt the so-called "inhibitor cystine knot" or "knottin" fold stabilized by three disulfide bonds. Its molecule is amphiphilic with a hydrophobic ridge on the surface enriched in aromatic residues and surrounded by positive charges. Correspondingly, Hm-3 binds to both neutral and negatively charged lipid vesicles. Electrophysiological studies showed that at a concentration of 1 μm Hm-3 effectively inhibited a number of mammalian and insect sodium channels. Importantly, Hm-3 shifted the dependence of channel activation to more positive voltages. Moreover, the inhibition was voltage-dependent, and strong depolarizing prepulses attenuated Hm-3 activity. The toxin is therefore concluded to represent the first sodium channel gating modifier from an araneomorph spider and features a "membrane access" mechanism of action. Its amino acid sequence and position of the hydrophobic cluster are notably different from other known gating modifiers from spider venom, all of which are described from mygalomorph species. We hypothesize parallel evolution of inhibitor cystine knot toxins from Araneomorphae and Mygalomorphae suborders.
Collapse
Affiliation(s)
- Antonina A Berkut
- From the M. M. Shemyakin and Yu. A. Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, 117997 Moscow, Russia, Moscow Institute of Physics and Technology (State University), 117303 Moscow, Russia, and
| | - Steve Peigneur
- Toxicology and Pharmacology, University of Leuven, 3000 Leuven, Belgium
| | - Mikhail Yu Myshkin
- From the M. M. Shemyakin and Yu. A. Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, 117997 Moscow, Russia, Moscow Institute of Physics and Technology (State University), 117303 Moscow, Russia, and
| | - Alexander S Paramonov
- From the M. M. Shemyakin and Yu. A. Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, 117997 Moscow, Russia
| | - Ekaterina N Lyukmanova
- From the M. M. Shemyakin and Yu. A. Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, 117997 Moscow, Russia
| | - Alexander S Arseniev
- From the M. M. Shemyakin and Yu. A. Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, 117997 Moscow, Russia, Moscow Institute of Physics and Technology (State University), 117303 Moscow, Russia, and
| | - Eugene V Grishin
- From the M. M. Shemyakin and Yu. A. Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, 117997 Moscow, Russia
| | - Jan Tytgat
- Toxicology and Pharmacology, University of Leuven, 3000 Leuven, Belgium
| | - Zakhar O Shenkarev
- From the M. M. Shemyakin and Yu. A. Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, 117997 Moscow, Russia
| | - Alexander A Vassilevski
- From the M. M. Shemyakin and Yu. A. Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, 117997 Moscow, Russia,
| |
Collapse
|
35
|
Cardiac functions of voltage-gated Ca(2+) channels: role of the pharmacoresistant type (E-/R-Type) in cardiac modulation and putative implication in sudden unexpected death in epilepsy (SUDEP). Rev Physiol Biochem Pharmacol 2014; 167:115-39. [PMID: 25280639 DOI: 10.1007/112_2014_21] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Voltage-gated Ca(2+) channels (VGCCs) are ubiquitous in excitable cells. These channels play key roles in many physiological events like cardiac regulation/pacemaker activity due to intracellular Ca(2+) transients. In the myocardium, the Cav1 subfamily (L-type: Cav1.2 and Cav1.3) is the main contributor to excitation-contraction coupling and/or pacemaking, whereas the Cav3 subfamily (T-type: Cav3.1 and Cav3.2) is important in rhythmically firing of the cardiac nodal cells. No established cardiac function has been attributed to the Cav2 family (E-/R-type: Cav2.3) despite accumulating evidence of cardiac dysregulation observed upon deletion of the Cav2.3 gene, the only member of this family so far detected in cardiomyocytes. In this review, we summarize the pathophysiological changes observed after ablation of the E-/R-type VGCC and propose a cardiac mechanism of action for this channel. Also, considering the role played by this channel in epilepsy and its reported sensitivity to antiepileptic drugs, a putative involvement of this channel in the cardiac mechanism of sudden unexpected death in epilepsy is also discussed.
Collapse
|