1
|
Sintsova O, Peigneur S, Kalina R, Otstavnykh N, Garbuz M, Klimovich A, Priymenko N, Shamatova M, Pavlenko A, Kozlov S, Gladkikh I, Isaeva M, Tytgat J, Leychenko E. The major component of Heteractis magnifica sea anemone venom, RpIII, exhibits strong subtype selectivity for insects over mammalian voltage-gated sodium channels. Neuropharmacology 2025; 274:110466. [PMID: 40246274 DOI: 10.1016/j.neuropharm.2025.110466] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2025] [Revised: 03/23/2025] [Accepted: 04/11/2025] [Indexed: 04/19/2025]
Abstract
Voltage-gated sodium channels (NaV) are molecular targets for the development of drugs for the treatment of diseases such as epilepsy, neuropathic pain, long QT syndrome, etc., as well as for insecticides. Therefore, the search for novel selective NaV channel ligands is relevant. Using amplicon deep sequencing of tentacle cDNA libraries from sea anemones Heteractis magnifica, 36 transcripts related to RpIII neurotoxin, a NaV channel modulators, were revealed. The recombinant RpIII was moderately toxic for mice (LD50 0.030 ± 0.004 mg/kg) but did not demonstrate any activity towards NaV in human SH-SY5Y cells. The toxin inhibited inactivation of heterologously expressed mammalian, insect, and arachnid NaV channels with higher specificity to insect channels. Cockroach (Blattella germanica) sodium channel BgNaV1 (EC50 of 2.4 ± 0.2 nM) and yellow fever mosquito (Aedes aegypti) channel AaNaV1 (EC50 of 1.5 ± 0.3 nM) were the most sensitive to RpIII, while mammals NaV had EC50 values above 100 nM except mNaV1.6 (EC50 of 43.8 ± 3.6 nM). The low nanomolar RpIII affinity to insect AaNaV1 may be explained by the extensive intermolecular contacts found by docking study. According to the predicted data, the toxin lands on the ion channel between voltage-sensing domain IV and pore domain I, also known as toxin site 3, followed by stabilizing the channels in the open state what was measured at electrophysiological experiments.
Collapse
Affiliation(s)
- Oksana Sintsova
- G.B. Elyakov Pacific Institute of Bioorganic Chemistry, Far Eastern Branch, Russian Academy of Sciences, 159, Pr. 100 let Vladivostoku, Vladivostok,690022, Russian Federation
| | - Steve Peigneur
- Toxicology and Pharmacology, KU Leuven Campus Gasthuisberg, ON2, Herestraat 49, Box-922, 3000, Leuven, Belgium
| | - Rimma Kalina
- G.B. Elyakov Pacific Institute of Bioorganic Chemistry, Far Eastern Branch, Russian Academy of Sciences, 159, Pr. 100 let Vladivostoku, Vladivostok,690022, Russian Federation
| | - Nadezhda Otstavnykh
- G.B. Elyakov Pacific Institute of Bioorganic Chemistry, Far Eastern Branch, Russian Academy of Sciences, 159, Pr. 100 let Vladivostoku, Vladivostok,690022, Russian Federation
| | - Mikhail Garbuz
- G.B. Elyakov Pacific Institute of Bioorganic Chemistry, Far Eastern Branch, Russian Academy of Sciences, 159, Pr. 100 let Vladivostoku, Vladivostok,690022, Russian Federation
| | - Anna Klimovich
- G.B. Elyakov Pacific Institute of Bioorganic Chemistry, Far Eastern Branch, Russian Academy of Sciences, 159, Pr. 100 let Vladivostoku, Vladivostok,690022, Russian Federation
| | - Nadezhda Priymenko
- G.B. Elyakov Pacific Institute of Bioorganic Chemistry, Far Eastern Branch, Russian Academy of Sciences, 159, Pr. 100 let Vladivostoku, Vladivostok,690022, Russian Federation
| | - Margarita Shamatova
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, ul. Miklukho-Maklaya 16/10, 117997, Moscow, Russian Federation
| | - Aleksandra Pavlenko
- G.B. Elyakov Pacific Institute of Bioorganic Chemistry, Far Eastern Branch, Russian Academy of Sciences, 159, Pr. 100 let Vladivostoku, Vladivostok,690022, Russian Federation
| | - Sergey Kozlov
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, ul. Miklukho-Maklaya 16/10, 117997, Moscow, Russian Federation
| | - Irina Gladkikh
- G.B. Elyakov Pacific Institute of Bioorganic Chemistry, Far Eastern Branch, Russian Academy of Sciences, 159, Pr. 100 let Vladivostoku, Vladivostok,690022, Russian Federation
| | - Marina Isaeva
- G.B. Elyakov Pacific Institute of Bioorganic Chemistry, Far Eastern Branch, Russian Academy of Sciences, 159, Pr. 100 let Vladivostoku, Vladivostok,690022, Russian Federation
| | - Jan Tytgat
- Toxicology and Pharmacology, KU Leuven Campus Gasthuisberg, ON2, Herestraat 49, Box-922, 3000, Leuven, Belgium
| | - Elena Leychenko
- G.B. Elyakov Pacific Institute of Bioorganic Chemistry, Far Eastern Branch, Russian Academy of Sciences, 159, Pr. 100 let Vladivostoku, Vladivostok,690022, Russian Federation.
| |
Collapse
|
2
|
Slowik KM, Edmans JG, Harrison S, Edwards SM, Bolt R, Spain SG, Hatton PV, Murdoch C, Colley HE. Controlled dual drug release from adhesive electrospun patches for prevention and treatment of alveolar osteitis. J Control Release 2024; 376:253-265. [PMID: 39389367 DOI: 10.1016/j.jconrel.2024.09.048] [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: 07/07/2024] [Revised: 09/21/2024] [Accepted: 09/28/2024] [Indexed: 10/12/2024]
Abstract
Approximately one in five individuals experience alveolar osteitis (AO) following wisdom tooth extraction. AO is characterised by loss of the blood clot from the tooth extraction socket leading to infection and pain, resulting in repeated hospital visits that impose a substantial burden on healthcare systems. Current treatments are sub-optimal; to address this we developed a novel drug-loaded mucoadhesive patch composed of dual electrospun polyvinyl pyrrolidone/Eudragit RS100 (PVP/RS100) and poly(N-isopropylacrylamide) (PNIPAM) fibres protected by a poly(ε-caprolactone) (PCL) backing layer. These patches demonstrated controlled release of the long-acting analgesic bupivacaine HCl and the anti-inflammatory drug prednisolone. Topical application of patches to tissue-engineered gingival mucosa showed that patch-released bupivacaine and prednisolone achieved sustained tissue permeation with 54.8 ± 3.3 % bupivacaine HCl and 65.8 ± 5.1 % prednisolone permeating the epithelium after 24 h. The drugs retained their functionality after release; bupivacaine HCl significantly (p < 0.05) inhibited veratridine-induced intracellular calcium flux in SH-SY5Y neuronal cells, while prednisolone significantly reduced gene expression of IL-6 (2-fold; p < 0.001), CXCL8 (5.1-fold; p < 0.01) and TNF-α (1.5-fold; p < 0.001) in stimulated THP-1 monocytes. Taken together, these data show that dual electrospun patches have the potential to provide a mucoadhesive covering to prevent blood clot loss while delivering pain relief and anti-inflammatory therapeutics at tooth extraction sites to prevent and treat AO. This study not only offers a future therapeutic pathway for AO but also contributes valuable insights into future advancements in drug delivery devices for periodontal or oral mucosal tissue.
Collapse
Affiliation(s)
- Klaudia M Slowik
- School of Clinical Dentistry, 19 Claremont Crescent, University of Sheffield, Sheffield S10 2TA, UK
| | - Jake G Edmans
- School of Clinical Dentistry, 19 Claremont Crescent, University of Sheffield, Sheffield S10 2TA, UK; Department of Chemistry, Brook Hill, University of Sheffield, Sheffield S3 7HF, UK
| | - Samuel Harrison
- Department of Chemistry, Brook Hill, University of Sheffield, Sheffield S3 7HF, UK
| | - Sean M Edwards
- Department of Mathematical Sciences, University of Liverpool, Liverpool L69 7ZL, UK
| | - Robert Bolt
- School of Clinical Dentistry, 19 Claremont Crescent, University of Sheffield, Sheffield S10 2TA, UK
| | - Sebastian G Spain
- Department of Chemistry, Brook Hill, University of Sheffield, Sheffield S3 7HF, UK
| | - Paul V Hatton
- School of Clinical Dentistry, 19 Claremont Crescent, University of Sheffield, Sheffield S10 2TA, UK; Insigneo, University of Sheffield, Sheffield, UK
| | - Craig Murdoch
- School of Clinical Dentistry, 19 Claremont Crescent, University of Sheffield, Sheffield S10 2TA, UK; Insigneo, University of Sheffield, Sheffield, UK.
| | - Helen E Colley
- School of Clinical Dentistry, 19 Claremont Crescent, University of Sheffield, Sheffield S10 2TA, UK; Insigneo, University of Sheffield, Sheffield, UK
| |
Collapse
|
3
|
Pukkanasut P, Jaskula-Sztul R, Gomora JC, Velu SE. Therapeutic targeting of voltage-gated sodium channel Na V1.7 for cancer metastasis. Front Pharmacol 2024; 15:1416705. [PMID: 39045054 PMCID: PMC11263763 DOI: 10.3389/fphar.2024.1416705] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2024] [Accepted: 06/12/2024] [Indexed: 07/25/2024] Open
Abstract
This review focuses on the expression and function of voltage-gated sodium channel subtype NaV1.7 in various cancers and explores its impact on the metastasis driving cell functions such as proliferation, migration, and invasiveness. An overview of its structural characteristics, drug binding sites, inhibitors and their likely mechanisms of action are presented. Despite the lack of clarity on the precise mechanism by which NaV1.7 contributes to cancer progression and metastasis; many studies have suggested a connection between NaV1.7 and proteins involved in multiple signaling pathways such as PKA and EGF/EGFR-ERK1/2. Moreover, the functional activity of NaV1.7 appears to elevate the expression levels of MACC1 and NHE-1, which are controlled by p38 MAPK activity, HGF/c-MET signaling and c-Jun activity. This cascade potentially enhances the secretion of extracellular matrix proteases, such as MMPs which play critical roles in cell migration and invasion activities. Furthermore, the NaV1.7 activity may indirectly upregulate Rho GTPases Rac activity, which is critical for cytoskeleton reorganization, cell adhesion, and actin polymerization. The relationship between NaV1.7 and cancer progression has prompted researchers to investigate the therapeutic potential of targeting NaV1.7 using inhibitors. The positive outcome of such studies resulted in the discovery of several inhibitors with the ability to reduce cancer cell migration, invasion, and tumor growth underscoring the significance of NaV1.7 as a promising pharmacological target for attenuating cancer cell proliferation and metastasis. The research findings summarized in this review suggest that the regulation of NaV1.7 expression and function by small molecules and/or by genetic engineering is a viable approach to discover novel therapeutics for the prevention and treatment of metastasis of cancers with elevated NaV1.7 expression.
Collapse
Affiliation(s)
- Piyasuda Pukkanasut
- Department of Chemistry, The University of Alabama at Birmingham, Birmingham, AL, United States
| | - Renata Jaskula-Sztul
- Department of Surgery, The University of Alabama at Birmingham, Birmingham, AL, United States
- O’Neal Comprehensive Cancer Center, The University of Alabama at Birmingham, Birmingham, AL, United States
| | - Juan Carlos Gomora
- Departamento de Neuropatología Molecular, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Sadanandan E. Velu
- Department of Chemistry, The University of Alabama at Birmingham, Birmingham, AL, United States
- O’Neal Comprehensive Cancer Center, The University of Alabama at Birmingham, Birmingham, AL, United States
| |
Collapse
|
4
|
Zosen D, Kondratskaya E, Kaplan-Arabaci O, Haugen F, Paulsen RE. Antidepressants escitalopram and venlafaxine up-regulate BDNF promoter IV but down-regulate neurite outgrowth in differentiating SH-SY5Y neurons. Neurochem Int 2023; 169:105571. [PMID: 37451345 DOI: 10.1016/j.neuint.2023.105571] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2023] [Revised: 06/26/2023] [Accepted: 07/04/2023] [Indexed: 07/18/2023]
Abstract
Antidepressants are used to treat depression and some anxiety disorders, including use in pregnant patients. The pharmacological actions of these drugs generally determine the uptake and metabolism of a series of neurotransmitters, such as serotonin, norepinephrine, or dopamine, along with an increase in BDNF expression. However, many aspects of antidepressant action remain unknown, particularly whether antidepressants interfere with normal neurodevelopment when taken by pregnant women. In order to reveal cellular and molecular implications crucial to the functioning of pathways related to antidepressant effects, we performed an investigation on neuronally differentiating human SH-SY5Y cells. To our knowledge, this is the first time human SH-SY5Y cells in cultures of purely neuronal cells induced by controlled differentiation with retinoic acid are followed by short-term 48-h exposure to 0.1-10 μM escitalopram or venlafaxine. Treatment with antidepressants (1 μM) did not affect the electrophysiological properties of SH-SY5Y cells. However, the percentage of mature neurons exhibiting voltage-gated sodium currents was substantially higher in cultures pre-treated with either antidepressant. After exposure to escitalopram or venlafaxine, we observed a concentration-dependent increase in activity-dependent BDNF promoter IV activation. The assessment of neurite metrics showed significant down-regulation of neurite outgrowth upon exposure to venlafaxine. Identified changes may represent links to molecular processes of importance to depression and be involved in neurodevelopmental alterations observed in postpartum children exposed to antidepressants antenatally.
Collapse
Affiliation(s)
- Denis Zosen
- Section for Pharmacology and Pharmaceutical Biosciences, Department of Pharmacy, Faculty of Mathematics and Natural Sciences, University of Oslo, Oslo, Norway
| | - Elena Kondratskaya
- Department of Medical Genetics, Oslo University Hospital, Oslo, Norway; NORMENT, Institute of Clinical Medicine, University of Oslo, and Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Norway
| | - Oykum Kaplan-Arabaci
- Section for Pharmacology and Pharmaceutical Biosciences, Department of Pharmacy, Faculty of Mathematics and Natural Sciences, University of Oslo, Oslo, Norway
| | - Fred Haugen
- Department of Work Psychology and Physiology, National Institute of Occupational Health (STAMI), Oslo, Norway
| | - Ragnhild Elisabeth Paulsen
- Section for Pharmacology and Pharmaceutical Biosciences, Department of Pharmacy, Faculty of Mathematics and Natural Sciences, University of Oslo, Oslo, Norway.
| |
Collapse
|
5
|
Galante P, Campos GAA, Moser JCG, Martins DB, Dos Santos Cabrera MP, Rangel M, Coelho LC, Simon KS, Amado VM, de A I Muller J, Koehbach J, Lohman RJ, Cabot PJ, Vetter I, Craik DJ, Toffoli-Kadri MC, Monge-Fuentes V, Goulart JT, Schwartz EF, Silva LP, Bocca AL, Mortari MR. Exploring the therapeutic potential of an antinociceptive and anti-inflammatory peptide from wasp venom. Sci Rep 2023; 13:12491. [PMID: 37528129 PMCID: PMC10393941 DOI: 10.1038/s41598-023-38828-w] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2023] [Accepted: 07/16/2023] [Indexed: 08/03/2023] Open
Abstract
Animal venoms are rich sources of neuroactive compounds, including anti-inflammatory, antiepileptic, and antinociceptive molecules. Our study identified a protonectin peptide from the wasp Parachartergus fraternus' venom using mass spectrometry and cDNA library construction. Using this peptide as a template, we designed a new peptide, protonectin-F, which exhibited higher antinociceptive activity and less motor impairment compared to protonectin. In drug interaction experiments with naloxone and AM251, Protonectin-F's activity was decreased by opioid and cannabinoid antagonism, two critical antinociception pathways. Further experiments revealed that this effect is most likely not induced by direct action on receptors but by activation of the descending pain control pathway. We noted that protonectin-F induced less tolerance in mice after repeated administration than morphine. Protonectin-F was also able to decrease TNF-α production in vitro and modulate the inflammatory response, which can further contribute to its antinociceptive activity. These findings suggest that protonectin-F may be a potential molecule for developing drugs to treat pain disorders with fewer adverse effects. Our results reinforce the biotechnological importance of animal venom for developing new molecules of clinical interest.
Collapse
Affiliation(s)
- Priscilla Galante
- Laboratory of Neuropharmacology, Department of Physiological Sciences, University of Brasília, Brasília, DF, 70910-900, Brazil
| | - Gabriel A A Campos
- Laboratory of Neuropharmacology, Department of Physiological Sciences, University of Brasília, Brasília, DF, 70910-900, Brazil
| | - Jacqueline C G Moser
- Laboratory of Neuropharmacology, Department of Physiological Sciences, University of Brasília, Brasília, DF, 70910-900, Brazil
| | - Danubia B Martins
- Department of Physics, IBILCE, São Paulo State University, São José do Rio Preto, SP, 15054-000, Brazil
| | | | - Marisa Rangel
- Immunopathology Laboratory, Butantan Institute, Sao Paulo, SP, 05503-900, Brazil
| | - Luiza C Coelho
- Laboratory of Applied Immunology, Department of Cell Biology, University of Brasilia, Brasilia, DF, 70910-900, Brazil
| | - Karina S Simon
- Laboratory of Applied Immunology, Department of Cell Biology, University of Brasilia, Brasilia, DF, 70910-900, Brazil
| | - Veronica M Amado
- Faculty of Medicine and University Hospital of Brasília, University of Brasilia, Brasilia, DF, 79910-900, Brazil
| | - Jessica de A I Muller
- Laboratory of Pharmacology and Inflammation FACFAN, Federal University of Mato Grosso do Sul, Campo Grande, Mato Grosso do Sul, 79070-900, Brazil
| | - Johannes Koehbach
- Institute for Molecular Bioscience, Australian Research Council Centre of Excellence for Innovations in Peptide and Protein Science, The University of Queensland, Brisbane, QLD, 4072, Australia
| | - Rink-Jan Lohman
- School of Pharmacy, The University of Queensland, Brisbane, QLD, 4072, Australia
| | - Peter J Cabot
- School of Pharmacy, The University of Queensland, Brisbane, QLD, 4072, Australia
| | - Irina Vetter
- Institute for Molecular Bioscience, Australian Research Council Centre of Excellence for Innovations in Peptide and Protein Science, The University of Queensland, Brisbane, QLD, 4072, Australia
| | - 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, QLD, 4072, Australia
| | - Monica C Toffoli-Kadri
- Laboratory of Pharmacology and Inflammation FACFAN, Federal University of Mato Grosso do Sul, Campo Grande, Mato Grosso do Sul, 79070-900, Brazil
| | - Victoria Monge-Fuentes
- Laboratory of Neuropharmacology, Department of Physiological Sciences, University of Brasília, Brasília, DF, 70910-900, Brazil
| | - Jair T Goulart
- Laboratory of Neuropharmacology, Department of Physiological Sciences, University of Brasília, Brasília, DF, 70910-900, Brazil
| | - Elisabeth F Schwartz
- Laboratory of Neuropharmacology, Department of Physiological Sciences, University of Brasília, Brasília, DF, 70910-900, Brazil
| | - Luciano P Silva
- Laboratory of Nanobiotechnology, Embrapa Genetic Resources and Biotechnology, Brasília, DF, 70770917, Brazil
| | - Anamelia L Bocca
- Laboratory of Applied Immunology, Department of Cell Biology, University of Brasilia, Brasilia, DF, 70910-900, Brazil
| | - Márcia R Mortari
- Laboratory of Neuropharmacology, Department of Physiological Sciences, University of Brasília, Brasília, DF, 70910-900, Brazil.
| |
Collapse
|
6
|
Sanchez-Sandoval AL, Hernández-Plata E, Gomora JC. Voltage-gated sodium channels: from roles and mechanisms in the metastatic cell behavior to clinical potential as therapeutic targets. Front Pharmacol 2023; 14:1206136. [PMID: 37456756 PMCID: PMC10348687 DOI: 10.3389/fphar.2023.1206136] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2023] [Accepted: 06/21/2023] [Indexed: 07/18/2023] Open
Abstract
During the second half of the last century, the prevalent knowledge recognized the voltage-gated sodium channels (VGSCs) as the proteins responsible for the generation and propagation of action potentials in excitable cells. However, over the last 25 years, new non-canonical roles of VGSCs in cancer hallmarks have been uncovered. Their dysregulated expression and activity have been associated with aggressive features and cancer progression towards metastatic stages, suggesting the potential use of VGSCs as cancer markers and prognostic factors. Recent work has elicited essential information about the signalling pathways modulated by these channels: coupling membrane activity to transcriptional regulation pathways, intracellular and extracellular pH regulation, invadopodia maturation, and proteolytic activity. In a promising scenario, the inhibition of VGSCs with FDA-approved drugs as well as with new synthetic compounds, reduces cancer cell invasion in vitro and cancer progression in vivo. The purpose of this review is to present an update regarding recent advances and ongoing efforts to have a better understanding of molecular and cellular mechanisms on the involvement of both pore-forming α and auxiliary β subunits of VGSCs in the metastatic processes, with the aim at proposing VGSCs as new oncological markers and targets for anticancer treatments.
Collapse
Affiliation(s)
- Ana Laura Sanchez-Sandoval
- Departamento de Neuropatología Molecular, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Mexico City, Mexico
- Medicina Genómica, Hospital General de México “Dr Eduardo Liceaga”, Mexico City, Mexico
| | - Everardo Hernández-Plata
- Consejo Nacional de Humanidades, Ciencias y Tecnologías and Instituto Nacional de Medicina Genómica, Mexico City, Mexico
| | - Juan Carlos Gomora
- Departamento de Neuropatología Molecular, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Mexico City, Mexico
| |
Collapse
|
7
|
Groome JR. Historical Perspective of the Characterization of Conotoxins Targeting Voltage-Gated Sodium Channels. Mar Drugs 2023; 21:209. [PMID: 37103349 PMCID: PMC10142487 DOI: 10.3390/md21040209] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2023] [Revised: 03/21/2023] [Accepted: 03/24/2023] [Indexed: 03/30/2023] Open
Abstract
Marine toxins have potent actions on diverse sodium ion channels regulated by transmembrane voltage (voltage-gated ion channels) or by neurotransmitters (nicotinic acetylcholine receptor channels). Studies of these toxins have focused on varied aspects of venom peptides ranging from evolutionary relationships of predator and prey, biological actions on excitable tissues, potential application as pharmacological intervention in disease therapy, and as part of multiple experimental approaches towards an understanding of the atomistic characterization of ion channel structure. This review examines the historical perspective of the study of conotoxin peptides active on sodium channels gated by transmembrane voltage, which has led to recent advances in ion channel research made possible with the exploitation of the diversity of these marine toxins.
Collapse
Affiliation(s)
- James R Groome
- Department of Biological Sciences, Idaho State University, Pocatello, ID 83209, USA
| |
Collapse
|
8
|
Chen ZH, Guo YW, Li XW. Recent advances on marine mollusk-derived natural products: chemistry, chemical ecology and therapeutical potential. Nat Prod Rep 2023; 40:509-556. [PMID: 35942896 DOI: 10.1039/d2np00021k] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Covering: 2011-2021Marine mollusks, which are well known as rich sources of diverse and biologically active natural products, have attracted significant attention from researchers due to their chemical and pharmacological properties. The occurrence of some of these marine mollusk-derived natural products in their preys, predators, and associated microorganisms has also gained interest in chemical ecology research. Based on previous reviews, herein, we present a comprehensive summary of the recent advances of interesting secondary metabolites from marine mollusks, focusing on their structural features, possible chemo-ecological significance, and promising biological activities, covering the literature from 2011 to 2021.
Collapse
Affiliation(s)
- Zi-Hui Chen
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zu Chong Zhi Road, Zhangjiang Hi-Tech Park, Shanghai 201203, China.
- University of Chinese Academy of Sciences, No. 19A Yuquan Road, Beijing 100049, China
| | - Yue-Wei Guo
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zu Chong Zhi Road, Zhangjiang Hi-Tech Park, Shanghai 201203, China.
- University of Chinese Academy of Sciences, No. 19A Yuquan Road, Beijing 100049, China
- Shandong Laboratory of Yantai Drug Discovery, Bohai Rim Advanced Research Institute for Drug Discovery, Yantai, Shandong 264117, China
| | - Xu-Wen Li
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zu Chong Zhi Road, Zhangjiang Hi-Tech Park, Shanghai 201203, China.
- University of Chinese Academy of Sciences, No. 19A Yuquan Road, Beijing 100049, China
- Shandong Laboratory of Yantai Drug Discovery, Bohai Rim Advanced Research Institute for Drug Discovery, Yantai, Shandong 264117, China
| |
Collapse
|
9
|
A bivalent remipede toxin promotes calcium release via ryanodine receptor activation. Nat Commun 2023; 14:1036. [PMID: 36823422 PMCID: PMC9950431 DOI: 10.1038/s41467-023-36579-w] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2022] [Accepted: 02/07/2023] [Indexed: 02/25/2023] Open
Abstract
Multivalent ligands of ion channels have proven to be both very rare and highly valuable in yielding unique insights into channel structure and pharmacology. Here, we describe a bivalent peptide from the venom of Xibalbanus tulumensis, a troglobitic arthropod from the enigmatic class Remipedia, that causes persistent calcium release by activation of ion channels involved in muscle contraction. The high-resolution solution structure of φ-Xibalbin3-Xt3a reveals a tandem repeat arrangement of inhibitor-cysteine knot (ICK) domains previously only found in spider venoms. The individual repeats of Xt3a share sequence similarity with a family of scorpion toxins that target ryanodine receptors (RyR). Single-channel electrophysiology and quantification of released Ca2+ stores within skinned muscle fibers confirm Xt3a as a bivalent RyR modulator. Our results reveal convergent evolution of RyR targeting toxins in remipede and scorpion venoms, while the tandem-ICK repeat architecture is an evolutionary innovation that is convergent with toxins from spider venoms.
Collapse
|
10
|
Ciocca M, Marcozzi S, Mariani P, Lacconi V, Di Carlo A, Cinà L, Rosato-Siri MD, Zanon A, Cattelan G, Avancini E, Lugli P, Priya S, Camaioni A, Brown TM. A Polymer Bio–Photoelectrolytic Platform for Electrical Signal Measurement and for Light Modulation of Ion Fluxes and Proliferation in a Neuroblastoma Cell Line. ADVANCED NANOBIOMED RESEARCH 2023. [DOI: 10.1002/anbr.202200127] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Affiliation(s)
- Manuela Ciocca
- Department of Electronic Engineering University of Rome Tor Vergata Via del Politecnico 1 00133 Rome Italy
- Faculty of Science and Technology Free University of Bozen-Bolzano Piazza Università 1 39100 Bolzano Italy
| | - Serena Marcozzi
- Department of Biomedicine and Prevention University of Rome Tor Vergata Via Montpellier 1 00133 Rome Italy
| | - Paolo Mariani
- Department of Electronic Engineering University of Rome Tor Vergata Via del Politecnico 1 00133 Rome Italy
| | - Valentina Lacconi
- Department of Biomedicine and Prevention University of Rome Tor Vergata Via Montpellier 1 00133 Rome Italy
| | - Aldo Di Carlo
- Istituto di Struttura della Materia CNR-ISM via Fosso del Cavaliere 100 00133 Rome Italy
| | - Lucio Cinà
- Cicci Research srl., Via Giordania 227 58100 Grosseto Italy
| | - Marcelo D. Rosato-Siri
- Institute for Biomedicine, Eurac Research Affiliated Institute of the University of Lübeck 39100 Bolzano Italy
| | - Alessandra Zanon
- Institute for Biomedicine, Eurac Research Affiliated Institute of the University of Lübeck 39100 Bolzano Italy
| | - Giada Cattelan
- Institute for Biomedicine, Eurac Research Affiliated Institute of the University of Lübeck 39100 Bolzano Italy
| | - Enrico Avancini
- Faculty of Science and Technology Free University of Bozen-Bolzano Piazza Università 1 39100 Bolzano Italy
| | - Paolo Lugli
- Faculty of Science and Technology Free University of Bozen-Bolzano Piazza Università 1 39100 Bolzano Italy
| | - Shashank Priya
- Department of Materials Science and Engineering Pennsylvania State University University Park PA 16802 USA
| | - Antonella Camaioni
- Department of Biomedicine and Prevention University of Rome Tor Vergata Via Montpellier 1 00133 Rome Italy
| | - Thomas M. Brown
- Department of Electronic Engineering University of Rome Tor Vergata Via del Politecnico 1 00133 Rome Italy
| |
Collapse
|
11
|
Wang D, Ai X, Duan Y, Xian N, Fang RH, Gao W, Zhang L. Neuronal Cellular Nanosponges for Effective Detoxification of Neurotoxins. ACS NANO 2022; 16:19145-19154. [PMID: 36354967 DOI: 10.1021/acsnano.2c08319] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/16/2023]
Abstract
Neurotoxins attack and destruct the nervous system, which can cause serious health problems and security threats. Existing detoxification approaches, such as antibodies and small molecule antidotes, rely on neurotoxin's molecular structure as design cues and require toxin-specific development for each type of toxins. However, the enormous diversity of neurotoxins makes such structure-based development of antitoxin particularly challenging and inefficient. Here, we report on the development and use of neuronal membrane-coated nanosponges (denoted "Neuron-NS") as an effective approach to detoxifying neurotoxins. Specifically, Neuron-NS act as neuron decoys to lure neurotoxins, bind with and neutralize the toxins, and thus block them from attacking the host neuron cells. These nanosponges detoxify neurotoxins regardless of their molecular structures and therefore can overcome the challenge posed by toxin structural diversity. In the study, we fabricate Neuron-NS by coating the membrane of Neuro-2a cells onto polymeric cores. Meanwhile, we select tetrodotoxin (TTX) as a model neurotoxin and demonstrate the detoxification efficacy of the Neuron-NS in a cytotoxicity assay, a calcium flux assay, and a cell osmotic swelling assay in vitro. Additionally, in mouse models of TTX intoxication, the Neuron-NS significantly enhance mouse survival in therapeutic and prophylactic regimens without showing acute toxicity. Overall, the Neuron-NS contribute to the current detoxification arsenal with the potential to treat various injuries and diseases caused by neurotoxins.
Collapse
Affiliation(s)
- Dan Wang
- Department of NanoEngineering and Chemical Engineering Program, University of California San Diego, La Jolla, California 92093, United States
| | - Xiangzhao Ai
- Department of NanoEngineering and Chemical Engineering Program, University of California San Diego, La Jolla, California 92093, United States
| | - Yaou Duan
- Department of NanoEngineering and Chemical Engineering Program, University of California San Diego, La Jolla, California 92093, United States
| | - Nianfei Xian
- Department of NanoEngineering and Chemical Engineering Program, University of California San Diego, La Jolla, California 92093, United States
| | - Ronnie H Fang
- Department of NanoEngineering and Chemical Engineering Program, University of California San Diego, La Jolla, California 92093, United States
| | - Weiwei Gao
- Department of NanoEngineering and Chemical Engineering Program, University of California San Diego, La Jolla, California 92093, United States
| | - Liangfang Zhang
- Department of NanoEngineering and Chemical Engineering Program, University of California San Diego, La Jolla, California 92093, United States
| |
Collapse
|
12
|
Barberio C, Saez J, Withers A, Nair M, Tamagnini F, Owens RM. Conducting Polymer-ECM Scaffolds for Human Neuronal Cell Differentiation. Adv Healthc Mater 2022; 11:e2200941. [PMID: 35904257 DOI: 10.1002/adhm.202200941] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2022] [Revised: 07/23/2022] [Indexed: 01/28/2023]
Abstract
3D cell culture formats more closely resemble tissue architecture complexity than 2D systems, which are lacking most of the cell-cell and cell-microenvironment interactions of the in vivo milieu. Scaffold-based systems integrating natural biomaterials are extensively employed in tissue engineering to improve cell survival and outgrowth, by providing the chemical and physical cues of the natural extracellular matrix (ECM). Using the freeze-drying technique, porous 3D composite scaffolds consisting of poly(3,4-ethylene-dioxythiophene) doped with polystyrene sulfonate (PEDOT:PSS), containing ECM components (i.e., collagen, hyaluronic acid, and laminin) are engineered for hosting neuronal cells. The resulting scaffolds exhibit a highly porous microstructure and good conductivity, determined by scanning electron microscopy and electrochemical impedance spectroscopy, respectively. These supports boast excellent mechanical stability and water uptake capacity, making them ideal candidates for cell infiltration. SH-SY5Y human neuroblastoma cells show enhanced cell survival and proliferation in the presence of ECM compared to PEDOT:PSS alone. Whole-cell patch-clamp recordings acquired from differentiated SHSY5Y cells in the scaffolds demonstrate that ECM constituents promote neuronal differentiation in situ. These findings reinforce the usability of 3D conducting supports as engineered highly biomimetic and functional in vitro tissue-like platforms for drug or disease modeling.
Collapse
Affiliation(s)
- Chiara Barberio
- Bioelectronic Systems and Technology group, Department of Chemical Engineering and Biotechnology, Philippa Fawcett Drive, Cambridge, CB3 0AS, UK
| | - Janire Saez
- Microfluidics Cluster UPV/EHU, BIOMICs Microfluidics Group, Lascaray Research Center, University of the Basque Country UPV/EHU, Vitoria-Gasteiz, 01006, Spain.,Ikerbasque, Basque Foundation for Science, Bilbao, E-48011, Spain
| | - Aimee Withers
- Bioelectronic Systems and Technology group, Department of Chemical Engineering and Biotechnology, Philippa Fawcett Drive, Cambridge, CB3 0AS, UK
| | - Malavika Nair
- Cambridge Centre for Medical Materials, Department of Materials Science and Metallurgy, University of Cambridge, 27 Charles Babbage Road, Cambridge, CB3 0FS, UK
| | - Francesco Tamagnini
- University of Reading, School of Pharmacy, Hopkins Building, Reading, RG6 6LA, UK
| | - Roisin M Owens
- Bioelectronic Systems and Technology group, Department of Chemical Engineering and Biotechnology, Philippa Fawcett Drive, Cambridge, CB3 0AS, UK
| |
Collapse
|
13
|
Louzao MC, Vilariño N, Vale C, Costas C, Cao A, Raposo-Garcia S, Vieytes MR, Botana LM. Current Trends and New Challenges in Marine Phycotoxins. Mar Drugs 2022; 20:md20030198. [PMID: 35323497 PMCID: PMC8950113 DOI: 10.3390/md20030198] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Revised: 03/04/2022] [Accepted: 03/05/2022] [Indexed: 02/04/2023] Open
Abstract
Marine phycotoxins are a multiplicity of bioactive compounds which are produced by microalgae and bioaccumulate in the marine food web. Phycotoxins affect the ecosystem, pose a threat to human health, and have important economic effects on aquaculture and tourism worldwide. However, human health and food safety have been the primary concerns when considering the impacts of phycotoxins. Phycotoxins toxicity information, often used to set regulatory limits for these toxins in shellfish, lacks traceability of toxicity values highlighting the need for predefined toxicological criteria. Toxicity data together with adequate detection methods for monitoring procedures are crucial to protect human health. However, despite technological advances, there are still methodological uncertainties and high demand for universal phycotoxin detectors. This review focuses on these topics, including uncertainties of climate change, providing an overview of the current information as well as future perspectives.
Collapse
Affiliation(s)
- Maria Carmen Louzao
- Departamento de Farmacologia, Facultad de Veterinaria, Universidade de Santiago de Compostela, 27002 Lugo, Spain; (N.V.); (C.V.); (C.C.); (A.C.); (S.R.-G.)
- Correspondence: (M.C.L.); (L.M.B.)
| | - Natalia Vilariño
- Departamento de Farmacologia, Facultad de Veterinaria, Universidade de Santiago de Compostela, 27002 Lugo, Spain; (N.V.); (C.V.); (C.C.); (A.C.); (S.R.-G.)
| | - Carmen Vale
- Departamento de Farmacologia, Facultad de Veterinaria, Universidade de Santiago de Compostela, 27002 Lugo, Spain; (N.V.); (C.V.); (C.C.); (A.C.); (S.R.-G.)
| | - Celia Costas
- Departamento de Farmacologia, Facultad de Veterinaria, Universidade de Santiago de Compostela, 27002 Lugo, Spain; (N.V.); (C.V.); (C.C.); (A.C.); (S.R.-G.)
| | - Alejandro Cao
- Departamento de Farmacologia, Facultad de Veterinaria, Universidade de Santiago de Compostela, 27002 Lugo, Spain; (N.V.); (C.V.); (C.C.); (A.C.); (S.R.-G.)
| | - Sandra Raposo-Garcia
- Departamento de Farmacologia, Facultad de Veterinaria, Universidade de Santiago de Compostela, 27002 Lugo, Spain; (N.V.); (C.V.); (C.C.); (A.C.); (S.R.-G.)
| | - Mercedes R. Vieytes
- Departamento de Fisiologia, Facultad de Veterinaria, Universidade de Santiago de Compostela, 27002 Lugo, Spain;
| | - Luis M. Botana
- Departamento de Farmacologia, Facultad de Veterinaria, Universidade de Santiago de Compostela, 27002 Lugo, Spain; (N.V.); (C.V.); (C.C.); (A.C.); (S.R.-G.)
- Correspondence: (M.C.L.); (L.M.B.)
| |
Collapse
|
14
|
Sánchez Triviño CA, Landinez MP, Duran S, Gomez MDP, Nasi E. Modulation of Gq/PLC-Mediated Signaling by Acute Lithium Exposure. Front Cell Neurosci 2022; 16:838939. [PMID: 35242014 PMCID: PMC8885521 DOI: 10.3389/fncel.2022.838939] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2021] [Accepted: 01/27/2022] [Indexed: 11/13/2022] Open
Abstract
Although lithium has long been one of the most widely used pharmacological agents in psychiatry, its mechanisms of action at the cellular and molecular levels remain poorly understood. One of the targets of Li+ is the phosphoinositide pathway, but whereas the impact of Li+ on inositol lipid metabolism is well documented, information on physiological effects at the cellular level is lacking. We examined in two mammalian cell lines the effect of acute Li+ exposure on the mobilization of internal Ca2+ and phospholipase C (PLC)-dependent membrane conductances. We first corroborated by Western blots and immunofluorescence in HEK293 cells the presence of key signaling elements of a muscarinic PLC pathway (M1AchR, Gq, PLC-β1, and IP3Rs). Stimulation with carbachol evoked a dose-dependent mobilization of Ca, as determined with fluorescent indicators. This was due to release from internal stores and proved susceptible to the PLC antagonist U73122. Li+ exposure reproducibly potentiated the Ca response in a concentration-dependent manner extending to the low millimolar range. To broaden those observations to a neuronal context and probe potential Li modulation of electrical signaling, we next examined the cell line SHsy5y. We replicated the potentiating effects of Li on the mobilization of internal Ca, and, after characterizing the basic properties of the electrical response to cholinergic stimulation, we also demonstrated an equally robust upregulation of muscarinic membrane currents. Finally, by directly stimulating the signaling pathway at different links downstream of the receptor, the site of action of the observed Li effects could be narrowed down to the G protein and its interaction with PLC-β. These observations document a modulation of Gq/PLC/IP3-mediated signaling by acute exposure to lithium, reflected in distinct physiological changes in cellular responses.
Collapse
Affiliation(s)
- Cesar Adolfo Sánchez Triviño
- Departamento de Biología, Universidad Nacional de Colombia, Bogotá, Colombia
- Centro Internacional de Física, Universidad Nacional de Colombia, Bogotá, Colombia
| | - Maria Paula Landinez
- Departamento de Biología, Universidad Nacional de Colombia, Bogotá, Colombia
- Centro Internacional de Física, Universidad Nacional de Colombia, Bogotá, Colombia
| | - Sara Duran
- Departamento de Biología, Universidad Nacional de Colombia, Bogotá, Colombia
- Centro Internacional de Física, Universidad Nacional de Colombia, Bogotá, Colombia
| | - María Del Pilar Gomez
- Departamento de Biología, Universidad Nacional de Colombia, Bogotá, Colombia
- Marine Biological Laboratory, Woods Hole, MA, United States
| | - Enrico Nasi
- Marine Biological Laboratory, Woods Hole, MA, United States
- Instituto de Genética, Universidad Nacional de Colombia, Bogotá, Colombia
| |
Collapse
|
15
|
Pharmacological Dissection of the Crosstalk between Na V and Ca V Channels in GH3b6 Cells. Int J Mol Sci 2022; 23:ijms23020827. [PMID: 35055012 PMCID: PMC8775721 DOI: 10.3390/ijms23020827] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Revised: 01/07/2022] [Accepted: 01/09/2022] [Indexed: 02/01/2023] Open
Abstract
Thanks to the crosstalk between Na+ and Ca2+ channels, Na+ and Ca2+ homeostasis interplay in so-called excitable cells enables the generation of action potential in response to electrical stimulation. Here, we investigated the impact of persistent activation of voltage-gated Na+ (NaV) channels by neurotoxins, such as veratridine (VTD), on intracellular Ca2+ concentration ([Ca2+]i) in a model of excitable cells, the rat pituitary GH3b6 cells, in order to identify the molecular actors involved in Na+-Ca2+ homeostasis crosstalk. By combining RT-qPCR, immunoblotting, immunocytochemistry, and patch-clamp techniques, we showed that GH3b6 cells predominantly express the NaV1.3 channel subtype, which likely endorses their voltage-activated Na+ currents. Notably, these Na+ currents were blocked by ICA-121431 and activated by the β-scorpion toxin Tf2, two selective NaV1.3 channel ligands. Using Fura-2, we showed that VTD induced a [Ca2+]i increase. This effect was suppressed by the selective NaV channel blocker tetrodotoxin, as well by the selective L-type CaV channel (LTCC) blocker nifedipine. We also evidenced that crobenetine, a NaV channel blocker, abolished VTD-induced [Ca2+]i elevation, while it had no effects on LTCC. Altogether, our findings highlight a crosstalk between NaV and LTCC in GH3b6 cells, providing a new insight into the mode of action of neurotoxins.
Collapse
|
16
|
Ridley J, Manyweathers S, Tang R, Goetze T, Becker N, Rinke-Weiß I, Kirby R, Obergrussberger A, Rogers M. Development of ASIC1a ligand-gated ion channel drug screening assays across multiple automated patch clamp platforms. Front Mol Neurosci 2022; 15:982689. [PMID: 36340694 PMCID: PMC9629855 DOI: 10.3389/fnmol.2022.982689] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Accepted: 09/12/2022] [Indexed: 02/05/2023] Open
Abstract
Human acid-sensing ion channels (ASIC) are ligand-gated ionotropic receptors expressed widely in peripheral tissues as well as sensory and central neurons and implicated in detection of inflammation, tissue injury, and hypoxia-induced acidosis. This makes ASIC channels promising targets for drug discovery in oncology, pain and ischemia, and several modulators have progressed into clinical trials. We describe the use of hASIC1a as a case study for the development and validation of low, medium and high throughput automated patch clamp (APC) assays suitable for the screening and mechanistic profiling of new ligands for this important class of ligand-gated ion channel. Initial efforts to expand on previous manual patch work describing an endogenous hASIC1a response in HEK cells were thwarted by low current expression and unusual pharmacology, so subsequent work utilized stable hASIC1a CHO cell lines. Ligand-gated application protocols and screening assays on the Patchliner, QPatch 48, and SyncroPatch 384 were optimized and validated based on pH activation and nM-μM potency of reference antagonists (e.g., Amiloride, Benzamil, Memantine, Mambalgin-3, A-317567, PcTx1). By optimizing single and stacked pipette tip applications available on each APC platform, stable pH-evoked currents during multiple ligand applications enabled cumulative EC50 and IC50 determinations with minimized receptor desensitization. Finally, we successfully demonstrated for the first time on an APC platform the ability to use current clamp to implement the historical technique of input resistance tracking to measure ligand-gated changes in membrane conductance on the Patchliner platform.
Collapse
Affiliation(s)
- John Ridley
- Metrion Biosciences Ltd., Cambridge, United Kingdom
| | | | - Raymond Tang
- Metrion Biosciences Ltd., Cambridge, United Kingdom
| | - Tom Goetze
- Nanion Technologies GmbH, Munich, Germany
| | | | | | - Robert Kirby
- Metrion Biosciences Ltd., Cambridge, United Kingdom
| | | | - Marc Rogers
- Metrion Biosciences Ltd., Cambridge, United Kingdom
- *Correspondence: Marc Rogers,
| |
Collapse
|
17
|
Dongol Y, Choi PM, Wilson DT, Daly NL, Cardoso FC, Lewis RJ. Voltage-Gated Sodium Channel Modulation by a New Spider Toxin Ssp1a Isolated From an Australian Theraphosid. Front Pharmacol 2021; 12:795455. [PMID: 35002728 PMCID: PMC8740163 DOI: 10.3389/fphar.2021.795455] [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: 10/15/2021] [Accepted: 11/22/2021] [Indexed: 11/13/2022] Open
Abstract
Given the important role of voltage-gated sodium (NaV) channel-modulating spider toxins in elucidating the function, pharmacology, and mechanism of action of therapeutically relevant NaV channels, we screened the venom from Australian theraphosid species against the human pain target hNaV1.7. Using assay-guided fractionation, we isolated a 33-residue inhibitor cystine knot (ICK) peptide (Ssp1a) belonging to the NaSpTx1 family. Recombinant Ssp1a (rSsp1a) inhibited neuronal hNaV subtypes with a rank order of potency hNaV1.7 > 1.6 > 1.2 > 1.3 > 1.1. rSsp1a inhibited hNaV1.7, hNaV1.2 and hNaV1.3 without significantly altering the voltage-dependence of activation, inactivation, or delay in recovery from inactivation. However, rSsp1a demonstrated voltage-dependent inhibition at hNaV1.7 and rSsp1a-bound hNaV1.7 opened at extreme depolarizations, suggesting rSsp1a likely interacted with voltage-sensing domain II (VSD II) of hNaV1.7 to trap the channel in its resting state. Nuclear magnetic resonance spectroscopy revealed key structural features of Ssp1a, including an amphipathic surface with hydrophobic and charged patches shown by docking studies to comprise the interacting surface. This study provides the basis for future structure-function studies to guide the development of subtype selective inhibitors.
Collapse
Affiliation(s)
- Yashad Dongol
- Centre for Pain Research, Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD, Australia
| | - Phil M. Choi
- Centre for Pain Research, Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD, Australia
| | - David T. Wilson
- Centre for Molecular Therapeutics, Australian Institute of Tropical Health and Medicine, James Cook University, Cairns, QLD, Australia
| | - Norelle L. Daly
- Centre for Molecular Therapeutics, Australian Institute of Tropical Health and Medicine, James Cook University, Cairns, QLD, Australia
| | - Fernanda C. Cardoso
- Centre for Pain Research, Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD, Australia
| | - Richard J. Lewis
- Centre for Pain Research, Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD, Australia
| |
Collapse
|
18
|
A Deep Learning Approach with Data Augmentation to Predict Novel Spider Neurotoxic Peptides. Int J Mol Sci 2021; 22:ijms222212291. [PMID: 34830173 PMCID: PMC8619404 DOI: 10.3390/ijms222212291] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2021] [Revised: 11/09/2021] [Accepted: 11/11/2021] [Indexed: 11/17/2022] Open
Abstract
As major components of spider venoms, neurotoxic peptides exhibit structural diversity, target specificity, and have great pharmaceutical potential. Deep learning may be an alternative to the laborious and time-consuming methods for identifying these peptides. However, the major hurdle in developing a deep learning model is the limited data on neurotoxic peptides. Here, we present a peptide data augmentation method that improves the recognition of neurotoxic peptides via a convolutional neural network model. The neurotoxic peptides were augmented with the known neurotoxic peptides from UniProt database, and the models were trained using a training set with or without the generated sequences to verify the augmented data. The model trained with the augmented dataset outperformed the one with the unaugmented dataset, achieving accuracy of 0.9953, precision of 0.9922, recall of 0.9984, and F1 score of 0.9953 in simulation dataset. From the set of all RNA transcripts of Callobius koreanus spider, we discovered neurotoxic peptides via the model, resulting in 275 putative peptides of which 252 novel sequences and only 23 sequences showing homology with the known peptides by Basic Local Alignment Search Tool. Among these 275 peptides, four were selected and shown to have neuromodulatory effects on the human neuroblastoma cell line SH-SY5Y. The augmentation method presented here may be applied to the identification of other functional peptides from biological resources with insufficient data.
Collapse
|
19
|
Loser D, Schaefer J, Danker T, Möller C, Brüll M, Suciu I, Ückert AK, Klima S, Leist M, Kraushaar U. Human neuronal signaling and communication assays to assess functional neurotoxicity. Arch Toxicol 2021; 95:229-252. [PMID: 33269408 PMCID: PMC7811517 DOI: 10.1007/s00204-020-02956-3] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2020] [Accepted: 11/16/2020] [Indexed: 01/08/2023]
Abstract
Prediction of drug toxicity on the human nervous system still relies mainly on animal experiments. Here, we developed an alternative system allowing assessment of complex signaling in both individual human neurons and on the network level. The LUHMES cultures used for our approach can be cultured in 384-well plates with high reproducibility. We established here high-throughput quantification of free intracellular Ca2+ concentrations [Ca2+]i as broadly applicable surrogate of neuronal activity and verified the main processes by patch clamp recordings. Initially, we characterized the expression pattern of many neuronal signaling components and selected the purinergic receptors to demonstrate the applicability of the [Ca2+]i signals for quantitative characterization of agonist and antagonist responses on classical ionotropic neurotransmitter receptors. This included receptor sub-typing and the characterization of the anti-parasitic drug suramin as modulator of the cellular response to ATP. To exemplify potential studies on ion channels, we characterized voltage-gated sodium channels and their inhibition by tetrodotoxin, saxitoxin and lidocaine, as well as their opening by the plant alkaloid veratridine and the food-relevant marine biotoxin ciguatoxin. Even broader applicability of [Ca2+]i quantification as an end point was demonstrated by measurements of dopamine transporter activity based on the membrane potential-changing activity of this neurotransmitter carrier. The substrates dopamine or amphetamine triggered [Ca2+]i oscillations that were synchronized over the entire culture dish. We identified compounds that modified these oscillations by interfering with various ion channels. Thus, this new test system allows multiple types of neuronal signaling, within and between cells, to be assessed, quantified and characterized for their potential disturbance.
Collapse
Affiliation(s)
- Dominik Loser
- NMI Natural and Medical Sciences Institute at the University of Tuebingen, 72770, Reutlingen, Germany
- NMI TT GmbH, 72770, Reutlingen, Germany
- Life Sciences Faculty, Albstadt-Sigmaringen University, 72488, Sigmaringen, Germany
| | - Jasmin Schaefer
- NMI Natural and Medical Sciences Institute at the University of Tuebingen, 72770, Reutlingen, Germany
- NMI TT GmbH, 72770, Reutlingen, Germany
| | | | - Clemens Möller
- Life Sciences Faculty, Albstadt-Sigmaringen University, 72488, Sigmaringen, Germany
| | - Markus Brüll
- In Vitro Toxicology and Biomedicine, Department Inaugurated by the Doerenkamp-Zbinden Foundation, University of Konstanz, Universitaetsstr. 10, 78457, Constance, Germany
| | - Ilinca Suciu
- In Vitro Toxicology and Biomedicine, Department Inaugurated by the Doerenkamp-Zbinden Foundation, University of Konstanz, Universitaetsstr. 10, 78457, Constance, Germany
| | - Anna-Katharina Ückert
- In Vitro Toxicology and Biomedicine, Department Inaugurated by the Doerenkamp-Zbinden Foundation, University of Konstanz, Universitaetsstr. 10, 78457, Constance, Germany
| | - Stefanie Klima
- In Vitro Toxicology and Biomedicine, Department Inaugurated by the Doerenkamp-Zbinden Foundation, University of Konstanz, Universitaetsstr. 10, 78457, Constance, Germany
| | - Marcel Leist
- In Vitro Toxicology and Biomedicine, Department Inaugurated by the Doerenkamp-Zbinden Foundation, University of Konstanz, Universitaetsstr. 10, 78457, Constance, Germany.
| | - Udo Kraushaar
- NMI Natural and Medical Sciences Institute at the University of Tuebingen, 72770, Reutlingen, Germany
| |
Collapse
|
20
|
Sun Y, He L, Wang T, Hua W, Qin H, Wang J, Wang L, Gu W, Li T, Li N, Liu X, Chen F, Tang L. Activation of p62-Keap1-Nrf2 Pathway Protects 6-Hydroxydopamine-Induced Ferroptosis in Dopaminergic Cells. Mol Neurobiol 2020; 57:4628-4641. [PMID: 32770451 DOI: 10.1007/s12035-020-02049-3] [Citation(s) in RCA: 109] [Impact Index Per Article: 21.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2020] [Accepted: 07/28/2020] [Indexed: 02/07/2023]
Abstract
Parkinson's disease (PD) is a common neurodegenerative disorder primarily caused by the death of dopaminergic neurons in the substantia nigra pars compacta (SNpc). However, the manner of death of dopaminergic neurons remains indistinct. Ferroptosis is a form of cell death involving in the iron-dependent accumulation of glutathione depletion and lipid peroxide. Besides, previous studies indicated that ferroptosis might be involved in the death of dopaminergic neurons. In this study, we aim to explore the protective effect of the p62-Keap1-Nrf2 pathway against 6-hydroxydopamine (6-OHDA)-induced ferroptosis in dopaminergic cells. Firstly, our results demonstrated that 6-OHDA-induced ferroptosis could be observed in vivo zebrafish and in vitro human dopaminergic cell line (SH-SY5Y cells) model. Moreover, ferroptosis induced by 6-OHDA mitigates in SH-SY5Y cells upon ferrostatin-1 (Fer, an inhibitor of ferroptosis) treatment via upregulating the protein expression of glutathione peroxidase 4 (GPX4). Then, we found that high p62/SQSTM1 (p62) expression could protect SH-SY5Y cells against ferroptosis through promoting Nrf2 nuclear transfer and upregulating the expression of the antioxidant protein heme oxygenase-1 (HO-1). Ultimately, high p62 expression activates the Nrf2/HO-1 signaling pathway through binding to Kelch-like ECH-associated protein 1 (Keap1). Collectively, the activation of the p62-Keap1-Nrf2 pathway prevents 6-OHDA-induced ferroptosis in SH-SY5Y cells, targeting this pathway in combination with a pharmacological inhibitor of ferroptosis can be a potential approach for PD therapy.
Collapse
Affiliation(s)
- Yiran Sun
- Key Laboratory of Bio-Resources and Eco-Environment of Ministry of Education, College of Life Sciences, Sichuan University, Chengdu, 610065, Sichuan, China.,National and Local Joint Engineering Laboratory for Energy Plant Bio-Oil Production and Application, Chengdu, 610065, Sichuan, China
| | - Libo He
- Key Laboratory of Bio-Resources and Eco-Environment of Ministry of Education, College of Life Sciences, Sichuan University, Chengdu, 610065, Sichuan, China.,National and Local Joint Engineering Laboratory for Energy Plant Bio-Oil Production and Application, Chengdu, 610065, Sichuan, China
| | - Taoyu Wang
- Key Laboratory of Bio-Resources and Eco-Environment of Ministry of Education, College of Life Sciences, Sichuan University, Chengdu, 610065, Sichuan, China.,National and Local Joint Engineering Laboratory for Energy Plant Bio-Oil Production and Application, Chengdu, 610065, Sichuan, China
| | - Wan Hua
- Key Laboratory of Bio-Resources and Eco-Environment of Ministry of Education, College of Life Sciences, Sichuan University, Chengdu, 610065, Sichuan, China.,National and Local Joint Engineering Laboratory for Energy Plant Bio-Oil Production and Application, Chengdu, 610065, Sichuan, China
| | - Huan Qin
- Key Laboratory of Bio-Resources and Eco-Environment of Ministry of Education, College of Life Sciences, Sichuan University, Chengdu, 610065, Sichuan, China.,National and Local Joint Engineering Laboratory for Energy Plant Bio-Oil Production and Application, Chengdu, 610065, Sichuan, China
| | - Jingjin Wang
- Key Laboratory of Bio-Resources and Eco-Environment of Ministry of Education, College of Life Sciences, Sichuan University, Chengdu, 610065, Sichuan, China.,National and Local Joint Engineering Laboratory for Energy Plant Bio-Oil Production and Application, Chengdu, 610065, Sichuan, China
| | - Li Wang
- Key Laboratory of Bio-Resources and Eco-Environment of Ministry of Education, College of Life Sciences, Sichuan University, Chengdu, 610065, Sichuan, China.,National and Local Joint Engineering Laboratory for Energy Plant Bio-Oil Production and Application, Chengdu, 610065, Sichuan, China
| | - Wanqin Gu
- Key Laboratory of Bio-Resources and Eco-Environment of Ministry of Education, College of Life Sciences, Sichuan University, Chengdu, 610065, Sichuan, China.,National and Local Joint Engineering Laboratory for Energy Plant Bio-Oil Production and Application, Chengdu, 610065, Sichuan, China
| | - Tingting Li
- Key Laboratory of Bio-Resources and Eco-Environment of Ministry of Education, College of Life Sciences, Sichuan University, Chengdu, 610065, Sichuan, China.,National and Local Joint Engineering Laboratory for Energy Plant Bio-Oil Production and Application, Chengdu, 610065, Sichuan, China
| | - Na Li
- Key Laboratory of Bio-Resources and Eco-Environment of Ministry of Education, College of Life Sciences, Sichuan University, Chengdu, 610065, Sichuan, China.,National and Local Joint Engineering Laboratory for Energy Plant Bio-Oil Production and Application, Chengdu, 610065, Sichuan, China
| | - Xinanbei Liu
- Key Laboratory of Bio-Resources and Eco-Environment of Ministry of Education, College of Life Sciences, Sichuan University, Chengdu, 610065, Sichuan, China.,National and Local Joint Engineering Laboratory for Energy Plant Bio-Oil Production and Application, Chengdu, 610065, Sichuan, China
| | - Fang Chen
- Key Laboratory of Bio-Resources and Eco-Environment of Ministry of Education, College of Life Sciences, Sichuan University, Chengdu, 610065, Sichuan, China.,National and Local Joint Engineering Laboratory for Energy Plant Bio-Oil Production and Application, Chengdu, 610065, Sichuan, China
| | - Lin Tang
- Key Laboratory of Bio-Resources and Eco-Environment of Ministry of Education, College of Life Sciences, Sichuan University, Chengdu, 610065, Sichuan, China. .,National and Local Joint Engineering Laboratory for Energy Plant Bio-Oil Production and Application, Chengdu, 610065, Sichuan, China.
| |
Collapse
|
21
|
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
|
22
|
Wilson DT, Bansal PS, Carter DA, Vetter I, Nicke A, Dutertre S, Daly NL. Characterisation of a Novel A-Superfamily Conotoxin. Biomedicines 2020; 8:biomedicines8050128. [PMID: 32443665 PMCID: PMC7277881 DOI: 10.3390/biomedicines8050128] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2020] [Revised: 05/15/2020] [Accepted: 05/18/2020] [Indexed: 12/13/2022] Open
Abstract
Conopeptides belonging to the A-superfamily from the venomous molluscs, Conus, are typically α-conotoxins. The α-conotoxins are of interest as therapeutic leads and pharmacological tools due to their selectivity and potency at nicotinic acetylcholine receptor (nAChR) subtypes. Structurally, the α-conotoxins have a consensus fold containing two conserved disulfide bonds that define the two-loop framework and brace a helical region. Here we report on a novel α-conotoxin Pl168, identified from the transcriptome of Conus planorbis, which has an unusual 4/8 loop framework. Unexpectedly, NMR determination of its three-dimensional structure reveals a new structural type of A-superfamily conotoxins with a different disulfide-stabilized fold, despite containing the conserved cysteine framework and disulfide connectivity of classical α-conotoxins. The peptide did not demonstrate activity on a range of nAChRs, or Ca2+ and Na+ channels suggesting that it might represent a new pharmacological class of conotoxins.
Collapse
Affiliation(s)
- David T. Wilson
- Centre for Molecular Therapeutics, Australian Institute of Tropical Health and Medicine, James Cook University, Smithfield, QLD 4878, Australia; (D.T.W.); (P.S.B.)
| | - Paramjit S. Bansal
- Centre for Molecular Therapeutics, Australian Institute of Tropical Health and Medicine, James Cook University, Smithfield, QLD 4878, Australia; (D.T.W.); (P.S.B.)
| | - David A. Carter
- Centre for Pain Research, Institute for Molecular Bioscience, The University of Queensland, St Lucia, QLD 4072, Australia; (D.A.C.); (I.V.)
| | - Irina Vetter
- Centre for Pain Research, Institute for Molecular Bioscience, The University of Queensland, St Lucia, QLD 4072, Australia; (D.A.C.); (I.V.)
- School of Pharmacy, The University of Queensland, Woolloongabba, QLD 4102, Australia
| | - Annette Nicke
- Walther Straub Institute of Pharmacology and Toxicology, Faculty of Medicine, LMU Munich, Nußbaumstraße 26, 80336 Munich, Germany;
| | - Sébastien Dutertre
- Institut des Biomolécules Max Mousseron, UMR 5247, Université de Montpellier, CNRS, 34095 Montpellier, France;
| | - Norelle L. Daly
- Centre for Molecular Therapeutics, Australian Institute of Tropical Health and Medicine, James Cook University, Smithfield, QLD 4878, Australia; (D.T.W.); (P.S.B.)
- Correspondence: ; Tel.: +61-7-4232-1815
| |
Collapse
|
23
|
Keenan B, Finol-Urdaneta RK, Hope A, Bremner JB, Kavallaris M, Lucena-Agell D, Oliva MÁ, Díaz JF, Vine KL. N-alkylisatin-based microtubule destabilizers bind to the colchicine site on tubulin and retain efficacy in drug resistant acute lymphoblastic leukemia cell lines with less in vitro neurotoxicity. Cancer Cell Int 2020; 20:170. [PMID: 32467666 PMCID: PMC7229617 DOI: 10.1186/s12935-020-01251-6] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2020] [Accepted: 05/07/2020] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND Drug resistance and chemotherapy-induced peripheral neuropathy continue to be significant problems in the successful treatment of acute lymphoblastic leukemia (ALL). 5,7-Dibromo-N-alkylisatins, a class of potent microtubule destabilizers, are a promising alternative to traditionally used antimitotics with previous demonstrated efficacy against solid tumours in vivo and ability to overcome P-glycoprotein (P-gp) mediated drug resistance in lymphoma and sarcoma cell lines in vitro. In this study, three di-brominated N-alkylisatins were assessed for their ability to retain potency in vincristine (VCR) and 2-methoxyestradiol (2ME2) resistant ALL cell lines. For the first time, in vitro neurotoxicity was also investigated in order to establish their suitability as candidate drugs for future use in ALL treatment. METHODS Vincristine resistant (CEM-VCR R) and 2-methoxyestradiol resistant (CEM/2ME2-28.8R) ALL cell lines were used to investigate the ability of N-alkylisatins to overcome chemoresistance. Interaction of N-alkylisatins with tubulin at the the colchicine-binding site was studied by competitive assay using the fluorescent colchicine analogue MTC. Human neuroblastoma SH-SY5Y cells differentiated into a morphological and functional dopaminergic-like neurotransmitter phenotype were used for neurotoxicity and neurofunctional assays. Two-way ANOVA followed by a Tukey's post hoc test or a two-tailed paired t test was used to determine statistical significance. RESULTS CEM-VCR R and CEM/2ME2-28.8R cells displayed resistance indices of > 100 to VCR and 2-ME2, respectively. CEM-VCR R cells additionally displayed a multi-drug resistant phenotype with significant cross resistance to vinblastine, 2ME2, colchicine and paclitaxel consistent with P-gp overexpression. Despite differences in resistance mechanisms observed between the two cell lines, the N-alkylisatins displayed bioequivalent dose-dependent cytotoxicity to that of the parental control cell line. The N-alkylisatins proved to be significantly less neurotoxic towards differentiated SH-SY5Y cells than VCR and vinblastine, evidenced by increased neurite length and number of neurite branch points. Neuronal cells treated with 5,7-dibromo-N-(p-hydroxymethylbenzyl)isatin showed significantly higher voltage-gated sodium channel function than those treated with Vinca alkaloids, strongly supportive of continued action potential firing. CONCLUSIONS The N-alkylisatins are able to retain cytotoxicity towards ALL cell lines with functionally distinct drug resistance mechanisms and show potential for reduced neurotoxicity. As such they pose as promising candidates for future implementation into anticancer regimes for ALL. Further in vivo studies are therefore warranted.
Collapse
Affiliation(s)
- Bryce Keenan
- School of Chemistry and Molecular Bioscience, Molecular Horizons, Faculty of Science, Medicine and Health, University of Wollongong, Northfields Ave, Wollongong, NSW Australia
- Illawarra Health and Medical Research Institute, Northfields Ave, Wollongong, NSW 2522 Australia
| | - Rocio K. Finol-Urdaneta
- Illawarra Health and Medical Research Institute, Northfields Ave, Wollongong, NSW 2522 Australia
- Electrophysiology Facility for Cell Phenotyping and Drug Discovery, Wollongong, NSW Australia
| | - Ashleigh Hope
- School of Chemistry and Molecular Bioscience, Molecular Horizons, Faculty of Science, Medicine and Health, University of Wollongong, Northfields Ave, Wollongong, NSW Australia
| | - John B. Bremner
- School of Chemistry and Molecular Bioscience, Molecular Horizons, Faculty of Science, Medicine and Health, University of Wollongong, Northfields Ave, Wollongong, NSW Australia
- Illawarra Health and Medical Research Institute, Northfields Ave, Wollongong, NSW 2522 Australia
| | - Maria Kavallaris
- Children’s Cancer Institute, Lowy Cancer Research Centre, UNSW, Sydney, NSW Australia
- ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, UNSW, Sydney, NSW Australia
- School of Women’s and Children’s Health, Faculty of Medicine, UNSW Sydney, Sydney, NSW Australia
| | - Daniel Lucena-Agell
- Centro de Investigaciones Biológicas, Consejo Superior de Investigaciones Científicas, Madrid, Spain
| | - María Ángela Oliva
- Centro de Investigaciones Biológicas, Consejo Superior de Investigaciones Científicas, Madrid, Spain
| | - Jose Fernando Díaz
- Centro de Investigaciones Biológicas, Consejo Superior de Investigaciones Científicas, Madrid, Spain
| | - Kara L. Vine
- School of Chemistry and Molecular Bioscience, Molecular Horizons, Faculty of Science, Medicine and Health, University of Wollongong, Northfields Ave, Wollongong, NSW Australia
- Illawarra Health and Medical Research Institute, Northfields Ave, Wollongong, NSW 2522 Australia
- Centre for Oncology Education and Research Translation (CONCERT), Cancer Institute NSW Translational Cancer Research Centre, NSW, Sydney, Australia
| |
Collapse
|
24
|
Malacrida A, Rivara M, Di Domizio A, Cislaghi G, Miloso M, Zuliani V, Nicolini G. 3D proteome-wide scale screening and activity evaluation of a new ALKBH5 inhibitor in U87 glioblastoma cell line. Bioorg Med Chem 2020; 28:115300. [PMID: 31937477 DOI: 10.1016/j.bmc.2019.115300] [Citation(s) in RCA: 108] [Impact Index Per Article: 21.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2019] [Revised: 12/18/2019] [Accepted: 12/26/2019] [Indexed: 12/14/2022]
Abstract
The imidazobenzoxazin-5-thione MV1035, synthesized as a new sodium channel blocker, has been tested on tumoral cells that differ for origin and for expressed NaV pool (U87-MG, H460 and A549). In this paper we focus on the effect of MV1035 in reducing U87 glioblastoma cell line migration and invasiveness. Since the effect of this compound on U87-MG cells seemed not dependent on its sodium channel blocking capability, alternative off-target interaction for MV1035 have been identified using SPILLO-PBSS software. This software performs a structure-based in silico screening on a proteome-wide scale, that allows to identify off-target interactions. Among the top-ranked off-targets of MV1035, we focused on the RNA demethylase ALKBH5 enzyme, known for playing a key role in cancer. In order to prove the effect of MV1035 on ALKBH5 in vitro coincubation of MV1035 and ALKBH5 has been performed demonstrating a consequent increase of N6-methyladenosine (m6A) RNA. To further validate the pathway involving ALKBH5 inhibition by MV1035 in U87-MG reduced migration and invasiveness, we evaluated CD73 as possible downstream protein. CD73 is an extrinsic protein involved in the generation of adenosine and is overexpressed in several tumors including glioblastoma. We have demonstrated that treating U87-MG with MV1035, CD73 protein expression was reduced without altering CD73 transcription. Our results show that MV1035 is able to significantly reduce U87 cell line migration and invasiveness inhibiting ALKBH5, an RNA demethylase that can be considered an interesting target in fighting glioblastoma aggressiveness. Our data encourage to further investigate the MV1035 inhibitory effect on glioblastoma.
Collapse
Affiliation(s)
- Alessio Malacrida
- School of Medicine and Surgery, Experimental Neurology Unit and Milan Center for Neuroscience, University of Milano-Bicocca, via Cadore 48, 20900 Monza, MB, Italy
| | - Mirko Rivara
- Food and Drug Department, University of Parma, Parco Area delle Scienze 27/A, 43124 Parma, PR, Italy.
| | - Alessandro Di Domizio
- Department of Pharmacological and Biomolecular Sciences, University of Milano, via Balzaretti 9, 20133 Milano, Italy; SPILLOproject, via Stradivari 17, 20037 Paderno Dugnano, Milano, Italy(2)
| | - Giacomo Cislaghi
- SPILLOproject, via Stradivari 17, 20037 Paderno Dugnano, Milano, Italy(2)
| | - Mariarosaria Miloso
- School of Medicine and Surgery, Experimental Neurology Unit and Milan Center for Neuroscience, University of Milano-Bicocca, via Cadore 48, 20900 Monza, MB, Italy
| | - Valentina Zuliani
- Food and Drug Department, University of Parma, Parco Area delle Scienze 27/A, 43124 Parma, PR, Italy
| | - Gabriella Nicolini
- School of Medicine and Surgery, Experimental Neurology Unit and Milan Center for Neuroscience, University of Milano-Bicocca, via Cadore 48, 20900 Monza, MB, Italy
| |
Collapse
|
25
|
High-Throughput Fluorescence Assays for Ion Channels and GPCRs. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1131:27-72. [DOI: 10.1007/978-3-030-12457-1_3] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
|
26
|
Albalawi F, Lim JC, DiRenzo KV, Hersh EV, Mitchell CH. Effects of Lidocaine and Articaine on Neuronal Survival and Recovery. Anesth Prog 2019; 65:82-88. [PMID: 29952644 DOI: 10.2344/anpr-65-02-02] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
The local anesthetics lidocaine and articaine are among the most widely used drugs in the dentist's arsenal, relieving pain by blocking voltage-dependent Na+ channels and thus preventing transmission of the pain signal. Given reports of infrequent but prolonged paresthesias with 4% articaine, we compared its neurotoxicity and functional impairment by screening cultured neural SH-SY5Y cells with formulations used in patients (2% lidocaine + 1:100,000 epinephrine or 4% articaine + 1:100,000 epinephrine) and with pure formulations of the drugs. Voltage-dependent sodium channels Na(v)1.2 and Na(v)1.7 were expressed in SH-SY5Y cells. To test the effects on viability, cells were exposed to drugs for 5 minutes, and after washing, cells were treated with the ratiometric Live/Dead assay. Articaine had no effect on the survival of SH-SY5Y cells, while lidocaine produced a significant reduction only when used as pure powder. To determine reversibility of blockage, wells were exposed to drugs for 5 minutes and returned for medium for 30 minutes, and the calcium elevation induced by depolarizing cells with a high-potassium solution was measured using the calcium indicator Fura-2. High potassium raised calcium in control SH-SY5Y cells and those treated with articaine, but lidocaine treatment significantly reduced the response. In conclusion, articaine does not damage neural cells more than lidocaine in this in vitro model. While this does not question the safety of lidocaine used clinically, it does suggest that articaine is no more neurotoxic, at least in the in vitro setting.
Collapse
Affiliation(s)
- Farraj Albalawi
- Department of Anatomy and Cell Biology, University of Pennsylvania School of Dental Medicine, Philadelphia, Pennsylvania.,Department of Orthodontics, University of Pennsylvania School of Dental Medicine, Philadelphia, Pennsylvania
| | - Jason C Lim
- Department of Anatomy and Cell Biology, University of Pennsylvania School of Dental Medicine, Philadelphia, Pennsylvania
| | - Kyle V DiRenzo
- Department of Anatomy and Cell Biology, University of Pennsylvania School of Dental Medicine, Philadelphia, Pennsylvania
| | - Elliot V Hersh
- Department of Oral & Maxillofacial Surgery/Pharmacology, University of Pennsylvania School of Dental Medicine, Philadelphia, Pennsylvania
| | - Claire H Mitchell
- Department of Anatomy and Cell Biology, University of Pennsylvania School of Dental Medicine, Philadelphia, Pennsylvania.,Department of Physiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, and.,Department of Ophthalmology, Perelman School of Medicine, University of Pennsylvania, Philadelphia
| |
Collapse
|
27
|
Liu J, Tan H, Yang W, Yao S, Hong L. The voltage-gated sodium channel Na v1.7 associated with endometrial cancer. J Cancer 2019; 10:4954-4960. [PMID: 31598168 PMCID: PMC6775510 DOI: 10.7150/jca.31544] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2018] [Accepted: 06/25/2019] [Indexed: 12/15/2022] Open
Abstract
Background: Endometrial cancer is the most common gynecologic malignancy in women in the developed countries. Despite recent progress in functional characterization of voltage-gated sodium channel (Nav) in multiple cancers, very little was known about the expression of Nav in human endometrial cancer. The present study sought to determine the role of Nav and molecular nature of this channel in the endometrial cancer. Methods: PCR approach was introduced to determine expression level of Nav subunits in endometrial cancer specimens. Pharmacological agents were used to investigate Nav function in endometrial cancer cells. Flow cytometry were used to test cancer apoptosis, and invasion assays were applied to test tumor metastasis. Results: Transcriptional levels of the all Nav α and β subunits were determined by real time-PCR in endometrial cancer with pair tissues of carcinoma and adjacent nonneoplastic tissue, Nav1.7 was the most highly expressed Nav subtype in endometrial cancer tissues. Nav1.7 level was closely associated with tumor size, local lymph node metastasis, and 5-year and 10-year survival ratio. Inhibition of this channel by Nav1.7 blocker PF-05089771, promoted cancer apoptosis and attenuated cancer cell invasion. Conclusion: These results establish a relationship between voltage-gated sodium channel protein and endometrial cancer, and suggest that Nav1.7 is a potential prognostic biomarker and could serve as a novel therapeutic target for endometrial cancer.
Collapse
Affiliation(s)
- Junxiu Liu
- Department of Obstetrics and Gynecology, the First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Hao Tan
- Department of Obstetrics and Gynecology, the First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Wancai Yang
- Institute of Precision Medicine, Jining Medical University, Jining, China
| | - Shuzhong Yao
- Department of Obstetrics and Gynecology, the First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Liang Hong
- Institute of Precision Medicine, Jining Medical University, Jining, China
| |
Collapse
|
28
|
Clitherow KH, Murdoch C, Spain SG, Handler AM, Colley HE, Stie MB, Mørck Nielsen H, Janfelt C, Hatton PV, Jacobsen J. Mucoadhesive Electrospun Patch Delivery of Lidocaine to the Oral Mucosa and Investigation of Spatial Distribution in a Tissue Using MALDI-Mass Spectrometry Imaging. Mol Pharm 2019; 16:3948-3956. [PMID: 31361498 PMCID: PMC7007276 DOI: 10.1021/acs.molpharmaceut.9b00535] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
![]()
Many
oral mucosal conditions cause considerable and prolonged pain
that to date has been difficult to alleviate via topical delivery,
and the use of injection causes many patients dental anxiety and needle-prick
pain. Therefore, developing a noninjectable drug delivery system as
an alternative administration procedure may vastly improve the health
and wellbeing of these patients. Recent advances in the development
of mucoadhesive electrospun patches for the direct delivery of therapeutics
to the oral mucosa offer a potential solution, but as yet, the release
of local anesthetics from this system and their uptake by oral tissue
have not been demonstrated. Here, we demonstrate the fabrication of
lidocaine-loaded electrospun fiber patches, drug release, and subsequent
uptake and permeation through the porcine buccal mucosa. Lidocaine
HCl and lidocaine base were incorporated into the electrospun patches
to evaluate the difference in drug permeation for the two drug compositions.
Lidocaine released from the lidocaine HCl-containing electrospun patches
was significantly quicker than from the lidocaine base patches, with
double the amount of drug released from the lidocaine HCl patches
in the first 15 min (0.16 ± 0.04 mg) compared to that from the
lidocaine base patches (0.07 ± 0.01 mg). The permeation of lidocaine
from the lidocaine HCl electrospun patches through ex vivo porcine
buccal mucosa was also detected in 15 min, whereas permeation of lidocaine
from the lidocaine base patch was not detected. Matrix-assisted laser
desorption ionization-mass spectrometry imaging was used to investigate
localization of lidocaine within the oral tissue. Lidocaine in the
solution as well as from the mucoadhesive patch penetrated into the
buccal mucosal tissue in a time-dependent manner and was detectable
in the lamina propria after only 15 min. Moreover, the lidocaine released
from lidocaine HCl electrospun patches retained biological activity,
inhibiting veratridine-mediated opening of voltage-gated sodium channels
in SH-SY5Y neuroblastoma cells. These data suggest that a mucoadhesive
electrospun patch may be used as a vehicle for rapid uptake and sustained
anesthetic drug delivery to treat or prevent oral pain.
Collapse
Affiliation(s)
- Katharina H Clitherow
- School of Clinical Dentistry , University of Sheffield , 19 Claremont Crescent , Sheffield S10 2TA , U.K
| | - Craig Murdoch
- School of Clinical Dentistry , University of Sheffield , 19 Claremont Crescent , Sheffield S10 2TA , U.K
| | - Sebastian Guy Spain
- Department of Chemistry , University of Sheffield , Brook Hill , Sheffield S3 7HF , U.K
| | - Anna Mette Handler
- Department of Pharmacy , University of Copenhagen , 2 Universitetsparken , Copenhagen DK-2100 , Denmark
| | - Helen E Colley
- School of Clinical Dentistry , University of Sheffield , 19 Claremont Crescent , Sheffield S10 2TA , U.K
| | - Mai Bay Stie
- Department of Pharmacy , University of Copenhagen , 2 Universitetsparken , Copenhagen DK-2100 , Denmark
| | - Hanne Mørck Nielsen
- Department of Pharmacy , University of Copenhagen , 2 Universitetsparken , Copenhagen DK-2100 , Denmark
| | - Christian Janfelt
- Department of Pharmacy , University of Copenhagen , 2 Universitetsparken , Copenhagen DK-2100 , Denmark
| | - Paul V Hatton
- School of Clinical Dentistry , University of Sheffield , 19 Claremont Crescent , Sheffield S10 2TA , U.K
| | - Jette Jacobsen
- Department of Pharmacy , University of Copenhagen , 2 Universitetsparken , Copenhagen DK-2100 , Denmark
| |
Collapse
|
29
|
Hompoonsup S, Chambers D, Doherty P, Williams G. No transcriptional evidence for active Na v channels in two classes of cancer cell. Channels (Austin) 2019; 13:311-320. [PMID: 31329011 PMCID: PMC6682260 DOI: 10.1080/19336950.2019.1644858] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022] Open
Abstract
Voltage-gated sodium channel (Nav) expression in non-excitable cells has raised questions regarding their non-canonical roles. Interestingly, a growing body of evidence also points towards the prevalence of aberrant Nav expression in malignant tumors, potentially opening a new therapeutic window. In this study, the transcriptional consequences of channel inhibition were investigated in non-small cell lung carcinoma H460 and neuroblastoma SH-SYSY cell lines, that both express Nav1.7. Channel activity was blocked by the application of both selective, ProTx-II, and non-selective, tetrodotoxin, inhibitors. Global gene expression profiling did not point to any statistically significant inhibition-associated perturbation of the transcriptome. A small subset of genes that showed relatively consistent changes across multiple treatments were further assayed in the context of a multiplex bead expression array which failed to recapitulate the changes seen in the global array. We conclude that there is no robust transcriptional signature associated with the inhibition of two sodium channel expressing cancer cell lines and consequently sodium channel inhibition will not lend itself to therapeutic approaches such as transcription-based drug repurposing.
Collapse
Affiliation(s)
- Supanida Hompoonsup
- a Wolfson Centre for Age-Related Diseases, King's College London , London , UK.,b Learning Institute, King Mongkut's University of Technology Thonburi , Bangkok , Thailand
| | - David Chambers
- a Wolfson Centre for Age-Related Diseases, King's College London , London , UK
| | - Patrick Doherty
- a Wolfson Centre for Age-Related Diseases, King's College London , London , UK
| | - Gareth Williams
- a Wolfson Centre for Age-Related Diseases, King's College London , London , UK
| |
Collapse
|
30
|
Novel conorfamides from Conus austini venom modulate both nicotinic acetylcholine receptors and acid-sensing ion channels. Biochem Pharmacol 2019; 164:342-348. [PMID: 31028742 DOI: 10.1016/j.bcp.2019.04.025] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2019] [Accepted: 04/22/2019] [Indexed: 01/06/2023]
Abstract
Conorfamides are a poorly studied family of cone snail venom peptides with broad biological activities, including inhibition of glutamate receptors, acid-sensing ion channels, and voltage-gated potassium channels. The aim of this study was to characterize the pharmacological activity of two novel linear conorfamides (conorfamide_As1a and conorfamide_As2a) and their non-amidated counterparts (conopeptide_As1b and conopeptide_As2b) that were isolated from the venom of the Mexican cone snail Conus austini. Although As1a, As2a, As1b and As2b were identified by activity-guided fractionation using a high-throughput fluorescence imaging plate reader (FLIPR) assay assessing α7 nAChR activity, sequence determination revealed activity associated with four linear peptides of the conorfamide rather than the anticipated α-conotoxin family. Pharmacological testing revealed that the amidated peptide variants altered desensitization of acid-sensing ion channels (ASICs) 1a and 3, and the native lysine to arginine mutation differentiating As1a and As1b from As2a and As2b introduced ASIC1a peak current potentiation. Surprisingly, these conorfamides also inhibited α7 and muscle-type nicotinic acetylcholine receptors (nAChR) at nanomolar concentrations. This is the first report of conorfamides with dual activity, with the nAChR activity being the most potent molecular target of any conorfamide discovered to date.
Collapse
|
31
|
Ziegman R, Brust A, Jha P, Cardoso FC, Lewis RJ, Alewood PF. 'Messy' Processing of χ-conotoxin MrIA Generates Homologues with Reduced hNET Potency. Mar Drugs 2019; 17:md17030165. [PMID: 30875751 PMCID: PMC6470548 DOI: 10.3390/md17030165] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2019] [Revised: 02/26/2019] [Accepted: 03/12/2019] [Indexed: 12/14/2022] Open
Abstract
Integrated venomics techniques have shown that variable processing of conotoxins from Conus marmoreus resulted in a dramatic expansion in the number of expressed conotoxins. One conotoxin from C. marmoreus, the χ-conotoxin MrIA, is a selective inhibitor of human norepinephrine transporters (hNET) and therefore a drug candidate for attenuating chronic neuropathic pain. It has been found that “messy” processing of the MrIA transcripts results in the expression of MrIA analogs with different truncations of the pro-peptide that contains portions of the MrIA molecule. The aim of this study was to investigate if variable processing of the expressed peptides results in modulation of the existing hNET pharmacology or creates new pharmacologies. To this end, a number of MrIA analogs found in C. marmoreus venom were synthesized and evaluated for their activity at hNET receptors. While several of the analogs exhibited norepinephrine transporter inhibitory activity comparable to that of MrIA, none significantly improved on the potency of conotoxin MrIA, and those analogs with disrupted pharmacophores produced greatly reduced NET inhibition, confirming previous structure-activity relationships seen on χ-class conopeptides. Additionally, analogs were screened for new activities on ion channels using calcium influx assays, although no major new pharmacology was revealed.
Collapse
Affiliation(s)
- Rebekah Ziegman
- Institute for Molecular Bioscience, The University of Queensland, St. Lucia, Queensland 4072, Australia.
| | - Andreas Brust
- Institute for Molecular Bioscience, The University of Queensland, St. Lucia, Queensland 4072, Australia.
| | - Prerna Jha
- Institute for Molecular Bioscience, The University of Queensland, St. Lucia, Queensland 4072, Australia.
| | - Fernanda C Cardoso
- Institute for Molecular Bioscience, The University of Queensland, St. Lucia, Queensland 4072, Australia.
| | - Richard J Lewis
- Institute for Molecular Bioscience, The University of Queensland, St. Lucia, Queensland 4072, Australia.
| | - Paul F Alewood
- Institute for Molecular Bioscience, The University of Queensland, St. Lucia, Queensland 4072, Australia.
| |
Collapse
|
32
|
Tay B, Stewart TA, Davis FM, Deuis JR, Vetter I. Development of a high-throughput fluorescent no-wash sodium influx assay. PLoS One 2019; 14:e0213751. [PMID: 30856233 PMCID: PMC6411159 DOI: 10.1371/journal.pone.0213751] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2018] [Accepted: 02/27/2019] [Indexed: 12/15/2022] Open
Abstract
Voltage-gated sodium channels (NaVs) are key therapeutic targets for pain, epilepsy and cardiac arrhythmias. Here we describe the development of a no-wash fluorescent sodium influx assay suitable for high-throughput screening and characterization of novel drug leads. Addition of red-violet food dyes (peak absorbance range 495–575 nm) to assays in HEK293 cells heterologously expressing hNaV1.1–1.8 effectively quenched background fluorescence of the sodium indicator dye Asante NaTRIUM Green-2 (ANG-2; peak emission 540 nm), negating the need for a wash step. Ponceau 4R (1 mM) was identified as a suitable quencher, which had no direct effect on NaV channels as assessed by patch-clamp experiments, and did not alter the pharmacology of the NaV1.1–1.7 activator veratridine (EC50 10–29 μM) or the NaV1.1–1.8 inhibitor tetracaine (IC50’s 6–66 μM). In addition, we also identified that the food dyes Ponceau 4R, Brilliant Black BN, Allura Red and Amaranth are effective at quenching the background fluorescence of the calcium indicator dyes fluo-4, fura-2 and fura-5F, identifying them as potential inexpensive alternatives to no-wash calcium ion indicator kits. In summary, we have developed a no-wash fluorescent sodium influx assay suitable for high-throughput screening based on the sodium indicator dye ANG-2 and the quencher Ponceau 4R.
Collapse
Affiliation(s)
- Bryan Tay
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland, Australia
| | - Teneale A. Stewart
- Mater Research Institute—The University of Queensland, Faculty of medicine, The University of Queensland, Woolloongabba, QLD, Australia
- Translational Research Institute, Woolloongabba, QLD, Australia
| | - Felicity M. Davis
- Mater Research Institute—The University of Queensland, Faculty of medicine, The University of Queensland, Woolloongabba, QLD, Australia
- Translational Research Institute, Woolloongabba, QLD, Australia
| | - Jennifer R. Deuis
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland, Australia
- * E-mail: (IV); (JD)
| | - Irina Vetter
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland, Australia
- School of Pharmacy, The University of Queensland, Brisbane, QLD, Australia
- * E-mail: (IV); (JD)
| |
Collapse
|
33
|
Agwa AJ, Blomster LV, Craik DJ, King GF, Schroeder CI. Efficient Enzymatic Ligation of Inhibitor Cystine Knot Spider Venom Peptides: Using Sortase A To Form Double-Knottins That Probe Voltage-Gated Sodium Channel NaV1.7. Bioconjug Chem 2018; 29:3309-3319. [DOI: 10.1021/acs.bioconjchem.8b00505] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Affiliation(s)
- Akello J. Agwa
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland 4072, Australia
| | - Linda V. Blomster
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland 4072, Australia
| | - David J. Craik
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland 4072, Australia
| | - Glenn F. King
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland 4072, Australia
| | - Christina I. Schroeder
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland 4072, Australia
| |
Collapse
|
34
|
Cardoso FC, Lewis RJ. Sodium channels and pain: from toxins to therapies. Br J Pharmacol 2018; 175:2138-2157. [PMID: 28749537 PMCID: PMC5980290 DOI: 10.1111/bph.13962] [Citation(s) in RCA: 71] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2017] [Revised: 07/11/2017] [Accepted: 07/17/2017] [Indexed: 12/16/2022] Open
Abstract
Voltage-gated sodium channels (NaV channels) are essential for the initiation and propagation of action potentials that critically influence our ability to respond to a diverse range of stimuli. Physiological and pharmacological studies have linked abnormal function of NaV channels to many human disorders, including chronic neuropathic pain. These findings, along with the description of the functional properties and expression pattern of NaV channel subtypes, are helping to uncover subtype specific roles in acute and chronic pain and revealing potential opportunities to target these with selective inhibitors. High-throughput screens and automated electrophysiology platforms have identified natural toxins as a promising group of molecules for the development of target-specific analgesics. In this review, the role of toxins in defining the contribution of NaV channels in acute and chronic pain states and their potential to be used as analgesic therapies are discussed. LINKED ARTICLES This article is part of a themed section on Recent Advances in Targeting Ion Channels to Treat Chronic Pain. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v175.12/issuetoc.
Collapse
Affiliation(s)
- Fernanda C Cardoso
- Department of Chemistry and Structural Biology, Institute for Molecular BioscienceThe University of QueenslandBrisbaneQLDAustralia
| | - Richard J Lewis
- Department of Chemistry and Structural Biology, Institute for Molecular BioscienceThe University of QueenslandBrisbaneQLDAustralia
| |
Collapse
|
35
|
Starobova H, S. W. A. H, Lewis RJ, Vetter I. Transcriptomics in pain research: insights from new and old technologies. Mol Omics 2018; 14:389-404. [DOI: 10.1039/c8mo00181b] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Physiological and pathological pain involves a complex interplay of multiple cell types and signaling pathways.
Collapse
Affiliation(s)
- H. Starobova
- Centre for Pain Research
- Institute for Molecular Bioscience
- University of Queensland
- St Lucia
- Australia
| | - Himaya S. W. A.
- Centre for Pain Research
- Institute for Molecular Bioscience
- University of Queensland
- St Lucia
- Australia
| | - R. J. Lewis
- Centre for Pain Research
- Institute for Molecular Bioscience
- University of Queensland
- St Lucia
- Australia
| | - I. Vetter
- Centre for Pain Research
- Institute for Molecular Bioscience
- University of Queensland
- St Lucia
- Australia
| |
Collapse
|
36
|
Kocmalova M, Joskova M, Franova S, Banovcin P, Sutovska M. Airway Defense Control Mediated via Voltage-Gated Sodium Channels. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017; 921:71-80. [PMID: 27161110 DOI: 10.1007/5584_2016_244] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Expression of voltage-gated sodium channels (Nav) takes place in the airways and the role of Nav1.7 and Nav1.8 in the control of airway's defense reflexes has been confirmed. The activation of Nav channels is crucial for cough initiation and airway smooth muscle reactivity, but it is unknown whether these channels regulate ciliary beating. This study evaluated the involvement of Nav1.7 and Nav1.8 channels in the airway defense mechanisms using their pharmacological blockers in healthy guinea pigs and in the experimental allergic asthma model. Asthma was modeled by ovalbumin sensitization over a period of 21 days. Blockade of Nav1.7 channels significantly decreased airway smooth muscle reactivity in vivo, the number of cough efforts, and the cilia beat frequency in healthy animals. In the allergic asthma model, blockade of Nav1.8 efficiently relieved symptoms of asthma, without adversely affecting cilia beat frequency. The study demonstrates that Nav1.8 channel antagonism has a potential to alleviate cough and bronchial hyperreactivity in asthma.
Collapse
Affiliation(s)
- M Kocmalova
- Department of Pharmacology, Division of Respirology, BioMed Martin, Jessenius Faculty of Medicine, Comenius University in Bratislava, 4C Mala Hora St, 036 01, Martin, Slovakia
| | - M Joskova
- Department of Pharmacology, Division of Respirology, BioMed Martin, Jessenius Faculty of Medicine, Comenius University in Bratislava, 4C Mala Hora St, 036 01, Martin, Slovakia.
| | - S Franova
- Department of Pharmacology, Division of Respirology, BioMed Martin, Jessenius Faculty of Medicine, Comenius University in Bratislava, 4C Mala Hora St, 036 01, Martin, Slovakia
| | - P Banovcin
- Department of Children and Adolescents, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava, and Martin University Hospital, Martin, Slovakia
| | - M Sutovska
- Department of Pharmacology, Division of Respirology, BioMed Martin, Jessenius Faculty of Medicine, Comenius University in Bratislava, 4C Mala Hora St, 036 01, Martin, Slovakia
| |
Collapse
|
37
|
Abstract
Crotalphine is a structural analogue to a novel analgesic peptide that was first identified in the crude venom from the South American rattlesnake Crotalus durissus terrificus. Although crotalphine's analgesic effect is well established, its direct mechanism of action remains unresolved. The aim of the present study was to investigate the effect of crotalphine on ion channels in peripheral pain pathways. We found that picomolar concentrations of crotalphine selectively activate heterologously expressed and native TRPA1 ion channels. TRPA1 activation by crotalphine required intact N-terminal cysteine residues and was followed by strong and long-lasting desensitization of the channel. Homologous desensitization of recombinant TRPA1 and heterologous desensitization in cultured dorsal root ganglia neurons was observed. Likewise, crotalphine acted on peptidergic TRPA1-expressing nerve endings ex vivo as demonstrated by suppression of calcitonin gene-related peptide release from the trachea and in vivo by inhibition of chemically induced and inflammatory hypersensitivity in mice. The crotalphine-mediated desensitizing effect was abolished by the TRPA1 blocker HC030031 and absent in TRPA1-deficient mice. Taken together, these results suggest that crotalphine is the first peptide to mediate antinociception selectively and at subnanomolar concentrations by targeting TRPA1 ion channels.
Collapse
|
38
|
Sousa SR, Wingerd JS, Brust A, Bladen C, Ragnarsson L, Herzig V, Deuis JR, Dutertre S, Vetter I, Zamponi GW, King GF, Alewood PF, Lewis RJ. Discovery and mode of action of a novel analgesic β-toxin from the African spider Ceratogyrus darlingi. PLoS One 2017; 12:e0182848. [PMID: 28880874 PMCID: PMC5589098 DOI: 10.1371/journal.pone.0182848] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2017] [Accepted: 07/25/2017] [Indexed: 01/13/2023] Open
Abstract
Spider venoms are rich sources of peptidic ion channel modulators with important therapeutical potential. We screened a panel of 60 spider venoms to find modulators of ion channels involved in pain transmission. We isolated, synthesized and pharmacologically characterized Cd1a, a novel peptide from the venom of the spider Ceratogyrus darlingi. Cd1a reversibly paralysed sheep blowflies (PD50 of 1318 pmol/g) and inhibited human Cav2.2 (IC50 2.6 μM) but not Cav1.3 or Cav3.1 (IC50 > 30 μM) in fluorimetric assays. In patch-clamp electrophysiological assays Cd1a inhibited rat Cav2.2 with similar potency (IC50 3 μM) without influencing the voltage dependence of Cav2.2 activation gating, suggesting that Cd1a doesn’t act on Cav2.2 as a classical gating modifier toxin. The Cd1a binding site on Cav2.2 did not overlap with that of the pore blocker ω-conotoxin GVIA, but its activity at Cav2.2-mutant indicated that Cd1a shares some molecular determinants with GVIA and MVIIA, localized near the pore region. Cd1a also inhibited human Nav1.1–1.2 and Nav1.7–1.8 (IC50 0.1–6.9 μM) but not Nav1.3–1.6 (IC50 > 30 μM) in fluorimetric assays. In patch-clamp assays, Cd1a strongly inhibited human Nav1.7 (IC50 16 nM) and produced a 29 mV depolarising shift in Nav1.7 voltage dependence of activation. Cd1a (400 pmol) fully reversed Nav1.7-evoked pain behaviours in mice without producing side effects. In conclusion, Cd1a inhibited two anti-nociceptive targets, appearing to interfere with Cav2.2 inactivation gating, associated with the Cav2.2 α-subunit pore, while altering the activation gating of Nav1.7. Cd1a was inactive at some of the Nav and Cav channels expressed in skeletal and cardiac muscles and nodes of Ranvier, apparently contributing to the lack of side effects at efficacious doses, and suggesting potential as a lead for development of peripheral pain treatments.
Collapse
Affiliation(s)
- Silmara R. Sousa
- IMB Centre for Pain Research, Institute for Molecular Bioscience, The University of Queensland, Brisbane, Australia
| | - Joshua S. Wingerd
- IMB Centre for Pain Research, Institute for Molecular Bioscience, The University of Queensland, Brisbane, Australia
| | - Andreas Brust
- IMB Centre for Pain Research, Institute for Molecular Bioscience, The University of Queensland, Brisbane, Australia
| | - Christopher Bladen
- Department of Physiology and Pharmacology, Hotchkiss Brain Institute and Alberta Children’s Hospital Research Institute, The University of Calgary, Calgary, Canada
| | - Lotten Ragnarsson
- IMB Centre for Pain Research, Institute for Molecular Bioscience, The University of Queensland, Brisbane, Australia
| | - Volker Herzig
- IMB Centre for Pain Research, Institute for Molecular Bioscience, The University of Queensland, Brisbane, Australia
| | - Jennifer R. Deuis
- IMB Centre for Pain Research, Institute for Molecular Bioscience, The University of Queensland, Brisbane, Australia
| | - Sebastien Dutertre
- Institut des Biomolécules Max Mousseron, Université Montpellier - CNRS, Montpellier, France
| | - Irina Vetter
- IMB Centre for Pain Research, Institute for Molecular Bioscience, The University of Queensland, Brisbane, Australia
- School of Pharmacy, The University of Queensland, Brisbane, Australia
| | - Gerald W. Zamponi
- Department of Physiology and Pharmacology, Hotchkiss Brain Institute and Alberta Children’s Hospital Research Institute, The University of Calgary, Calgary, Canada
| | - Glenn F. King
- IMB Centre for Pain Research, Institute for Molecular Bioscience, The University of Queensland, Brisbane, Australia
| | - Paul F. Alewood
- IMB Centre for Pain Research, Institute for Molecular Bioscience, The University of Queensland, Brisbane, Australia
| | - Richard J. Lewis
- IMB Centre for Pain Research, Institute for Molecular Bioscience, The University of Queensland, Brisbane, Australia
- * E-mail:
| |
Collapse
|
39
|
Cardoso FC, Dekan Z, Smith JJ, Deuis JR, Vetter I, Herzig V, Alewood PF, King GF, Lewis RJ. Modulatory features of the novel spider toxin μ-TRTX-Df1a isolated from the venom of the spider Davus fasciatus. Br J Pharmacol 2017; 174:2528-2544. [PMID: 28542706 DOI: 10.1111/bph.13865] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2016] [Revised: 05/01/2017] [Accepted: 05/02/2017] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND AND PURPOSE Naturally occurring dysfunction of voltage-gated sodium (NaV ) channels results in complex disorders such as chronic pain, making these channels an attractive target for new therapies. In the pursuit of novel NaV modulators, we investigated spider venoms for new inhibitors of NaV channels. EXPERIMENTAL APPROACH We used high-throughput screens to identify a NaV modulator in venom of the spider Davus fasciatus. Further characterization of this venom peptide was undertaken using fluorescent and electrophysiological assays, molecular modelling and a rodent pain model. KEY RESULTS We identified a potent NaV inhibitor named μ-TRTX-Df1a. This 34-residue peptide fully inhibited responses mediated by NaV 1.7 endogenously expressed in SH-SY5Y cells. Df1a also inhibited voltage-gated calcium (CaV 3) currents but had no activity against the voltage-gated potassium (KV 2) channel. The modelled structure of Df1a, which contains an inhibitor cystine knot motif, is reminiscent of the NaV channel toxin ProTx-I. Electrophysiology revealed that Df1a inhibits all NaV subtypes tested (hNaV 1.1-1.7). Df1a also slowed fast inactivation of NaV 1.1, NaV 1.3 and NaV 1.5 and modified the voltage-dependence of activation and inactivation of most of the NaV subtypes. Df1a preferentially binds to the domain II voltage-sensor and has additional interactions with the voltage sensors domains III and IV, which probably explains its modulatory features. Df1a was analgesic in vivo, reversing the spontaneous pain behaviours induced by the NaV activator OD1. CONCLUSION AND IMPLICATIONS μ-TRTX-Df1a shows potential as a new molecule for the development of drugs to treat pain disorders mediated by voltage-gated ion channels.
Collapse
Affiliation(s)
- Fernanda C Cardoso
- Institute for Molecular Bioscience, The University of Queensland, St Lucia, QLD, Australia
| | - Zoltan Dekan
- Institute for Molecular Bioscience, The University of Queensland, St Lucia, QLD, Australia
| | - Jennifer J Smith
- Institute for Molecular Bioscience, The University of Queensland, St Lucia, QLD, Australia
| | - Jennifer R Deuis
- Institute for Molecular Bioscience, The University of Queensland, St Lucia, QLD, Australia.,School of Pharmacy, The University of Queensland, Woolloongabba, QLD, Australia
| | - Irina Vetter
- Institute for Molecular Bioscience, The University of Queensland, St Lucia, QLD, Australia.,School of Pharmacy, The University of Queensland, Woolloongabba, QLD, Australia
| | - Volker Herzig
- Institute for Molecular Bioscience, The University of Queensland, St Lucia, QLD, Australia
| | - Paul F Alewood
- 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
| | - Richard J Lewis
- Institute for Molecular Bioscience, The University of Queensland, St Lucia, QLD, Australia
| |
Collapse
|
40
|
Coccini T, Caloni F, De Simone U. Human neuronal cell based assay: A new in vitro model for toxicity evaluation of ciguatoxin. ENVIRONMENTAL TOXICOLOGY AND PHARMACOLOGY 2017; 52:200-213. [PMID: 28437641 DOI: 10.1016/j.etap.2017.04.003] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/23/2016] [Revised: 03/31/2017] [Accepted: 04/02/2017] [Indexed: 06/07/2023]
Abstract
Ciguatoxins (CTXs) are emerging marine neurotoxins representing the main cause of ciguatera fish poisoning, an intoxication syndrome which configures a health emergency and constitutes an evolving issue constantly changing due to new vectors and derivatives of CTXs, as well as their presence in new non-endemic areas. The study applied the neuroblastoma cell model of human origin (SH-SY5Y) to evaluate species-specific mechanistic information on CTX toxicity. Metabolic functionality, cell morphology, cytosolic Ca2+i responses, neuronal cell growth and proliferation were assessed after short- (4-24h) and long-term exposure (10days) to P-CTX-3C. In SH-SY5Y, P-CTX-3C displayed a powerful cytotoxicity requiring the presence of both Veratridine and Ouabain. SH-SY5Y were very sensitive to Ouabain: 10 and 0.25nM appeared the optimal concentrations, for short- and long-term toxicity studies, respectively, to be used in co-incubation with Veratridine (25μM), simulating the physiological and pathological endogenous Ouabain levels in humans. P-CTX-3C cytotoxic effect, on human neurons co-incubated with OV (Ouabain+Veratridine) mix, was expressed starting from 100pM after short- and 25pM after long-term exposure. Notably, P-CTX-3C alone at 25nM induced cytotoxicity after 24h and prolonged exposure. This human brain-derived cell line appears a suitable cell-based-model to evaluate cytotoxicity of CTX present in marine food contaminated at low toxic levels and to characterize the toxicological profile of other/new congeners.
Collapse
Affiliation(s)
- Teresa Coccini
- Laboratory of Clinical and Experimental Toxicology, Poison Control Centre, Toxicology Unit, Maugeri Clinical Scientific Institutes S.p.A.-BS, IRCCS Pavia, Pavia Italy.
| | - Francesca Caloni
- Department of Veterinary Medicine (DIMEVET), Università degli Studi di Milano, Milano, Italy
| | - Uliana De Simone
- Laboratory of Clinical and Experimental Toxicology, Poison Control Centre, Toxicology Unit, Maugeri Clinical Scientific Institutes S.p.A.-BS, IRCCS Pavia, Pavia Italy
| |
Collapse
|
41
|
Wingerd JS, Mozar CA, Ussing CA, Murali SS, Chin YKY, Cristofori-Armstrong B, Durek T, Gilchrist J, Vaughan CW, Bosmans F, Adams DJ, Lewis RJ, Alewood PF, Mobli M, Christie MJ, Rash LD. The tarantula toxin β/δ-TRTX-Pre1a highlights the importance of the S1-S2 voltage-sensor region for sodium channel subtype selectivity. Sci Rep 2017; 7:974. [PMID: 28428547 PMCID: PMC5430537 DOI: 10.1038/s41598-017-01129-0] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2016] [Accepted: 03/27/2017] [Indexed: 11/09/2022] Open
Abstract
Voltage-gated sodium (NaV) channels are essential for the transmission of pain signals in humans making them prime targets for the development of new analgesics. Spider venoms are a rich source of peptide modulators useful to study ion channel structure and function. Here we describe β/δ-TRTX-Pre1a, a 35-residue tarantula peptide that selectively interacts with neuronal NaV channels inhibiting peak current of hNaV1.1, rNaV1.2, hNaV1.6, and hNaV1.7 while concurrently inhibiting fast inactivation of hNaV1.1 and rNaV1.3. The DII and DIV S3-S4 loops of NaV channel voltage sensors are important for the interaction of Pre1a with NaV channels but cannot account for its unique subtype selectivity. Through analysis of the binding regions we ascertained that the variability of the S1-S2 loops between NaV channels contributes substantially to the selectivity profile observed for Pre1a, particularly with regards to fast inactivation. A serine residue on the DIV S2 helix was found to be sufficient to explain Pre1a’s potent and selective inhibitory effect on the fast inactivation process of NaV1.1 and 1.3. This work highlights that interactions with both S1-S2 and S3-S4 of NaV channels may be necessary for functional modulation, and that targeting the diverse S1-S2 region within voltage-sensing domains provides an avenue to develop subtype selective tools.
Collapse
Affiliation(s)
- Joshua S Wingerd
- Institute for Molecular Bioscience, The University of Queensland, St Lucia, QLD, 4072, Australia
| | - Christine A Mozar
- Discipline of Pharmacology, University of Sydney, Camperdown, NSW, 2006, Australia
| | - Christine A Ussing
- Institute for Molecular Bioscience, The University of Queensland, St Lucia, QLD, 4072, Australia.,Novo Nordisk A/S, Copenhagen Area, Capital Region, Denmark
| | - Swetha S Murali
- Discipline of Pharmacology, University of Sydney, Camperdown, NSW, 2006, Australia.,Harvard Medical School, Children's Hospital, 300 Longwood Ave, Boston, MA, 02115, United States
| | - Yanni K-Y Chin
- Institute for Molecular Bioscience, The University of Queensland, St Lucia, QLD, 4072, Australia
| | - Ben Cristofori-Armstrong
- Institute for Molecular Bioscience, The University of Queensland, St Lucia, QLD, 4072, Australia
| | - Thomas Durek
- Institute for Molecular Bioscience, The University of Queensland, St Lucia, QLD, 4072, Australia
| | - John Gilchrist
- Department of Physiology and Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, Maryland, 21205, USA
| | - Christopher W Vaughan
- Pain Management Research Institute, University of Sydney, St Leonards, NSW, 2006, Australia
| | - Frank Bosmans
- Department of Physiology and Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, Maryland, 21205, USA
| | - David J Adams
- Illawarra Health and Medical Research Institute, University of Wollongong, Wollongong, NSW, 2522, Australia
| | - Richard J Lewis
- Institute for Molecular Bioscience, The University of Queensland, St Lucia, QLD, 4072, Australia
| | - Paul F Alewood
- Institute for Molecular Bioscience, The University of Queensland, St Lucia, QLD, 4072, Australia
| | - Mehdi Mobli
- Centre for Advanced Imaging & School of Chemistry and Molecular Biosciences, The University of Queensland, St Lucia, QLD, 4072, Australia
| | - Macdonald J Christie
- Discipline of Pharmacology, University of Sydney, Camperdown, NSW, 2006, Australia
| | - Lachlan D Rash
- Institute for Molecular Bioscience, The University of Queensland, St Lucia, QLD, 4072, Australia. .,School of Biomedical Sciences, The University of Queensland, St Lucia, 4072, QLD, Australia.
| |
Collapse
|
42
|
Deuis JR, Mueller A, Israel MR, Vetter I. The pharmacology of voltage-gated sodium channel activators. Neuropharmacology 2017; 127:87-108. [PMID: 28416444 DOI: 10.1016/j.neuropharm.2017.04.014] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2017] [Revised: 03/28/2017] [Accepted: 04/10/2017] [Indexed: 12/19/2022]
Abstract
Toxins and venom components that target voltage-gated sodium (NaV) channels have evolved numerous times due to the importance of this class of ion channels in the normal physiological function of peripheral and central neurons as well as cardiac and skeletal muscle. NaV channel activators in particular have been isolated from the venom of spiders, wasps, snakes, scorpions, cone snails and sea anemone and are also produced by plants, bacteria and algae. These compounds have provided key insight into the molecular structure, function and pathophysiological roles of NaV channels and are important tools due to their at times exquisite subtype-selectivity. We review the pharmacology of NaV channel activators with particular emphasis on mammalian isoforms and discuss putative applications for these compounds. This article is part of the Special Issue entitled 'Venom-derived Peptides as Pharmacological Tools.'
Collapse
Affiliation(s)
- Jennifer R Deuis
- Centre for Pain Research, Institute for Molecular Bioscience, The University of Queensland, St Lucia, Qld 4072, Australia
| | - Alexander Mueller
- Centre for Pain Research, Institute for Molecular Bioscience, The University of Queensland, St Lucia, Qld 4072, Australia
| | - Mathilde R Israel
- Centre for Pain Research, Institute for Molecular Bioscience, The University of Queensland, St Lucia, Qld 4072, Australia
| | - Irina Vetter
- Centre for Pain Research, Institute for Molecular Bioscience, The University of Queensland, St Lucia, Qld 4072, Australia; School of Pharmacy, The University of Queensland, Woolloongabba, Qld 4102, Australia.
| |
Collapse
|
43
|
O'Neill K, Musgrave IF, Humpage A. Extended Low-Dose Exposure to Saxitoxin Inhibits Neurite Outgrowth in Model Neuronal Cells. Basic Clin Pharmacol Toxicol 2017; 120:390-397. [DOI: 10.1111/bcpt.12701] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2016] [Accepted: 11/02/2016] [Indexed: 11/28/2022]
Affiliation(s)
- Katie O'Neill
- Discipline of Pharmacology; School of Medicine; The University of Adelaide; Adelaide SA Australia
| | - Ian F. Musgrave
- Discipline of Pharmacology; School of Medicine; The University of Adelaide; Adelaide SA Australia
| | - Andrew Humpage
- Australian Water Quality Centre; South Australian Water Corporation; Adelaide SA Australia
| |
Collapse
|
44
|
Gervois P, Wolfs E, Dillen Y, Hilkens P, Ratajczak J, Driesen R, Vangansewinkel T, Bronckaers A, Brône B, Struys T, Lambrichts I. Paracrine Maturation and Migration of SH-SY5Y Cells by Dental Pulp Stem Cells. J Dent Res 2017; 96:654-662. [DOI: 10.1177/0022034517690491] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Neurological disorders are characterized by neurodegeneration and/or loss of neuronal function, which cannot be adequately repaired by the host. Therefore, there is need for novel treatment options such as cell-based therapies that aim to salvage or reconstitute the lost tissue or that stimulate host repair. The present study aimed to evaluate the paracrine effects of human dental pulp stem cells (hDPSCs) on the migration and neural maturation of human SH-SY5Y neuroblastoma cells. The hDPSC secretome had a significant chemoattractive effect on SH-SY5Y cells as shown by a transwell assay. To evaluate neural maturation, SH-SY5Y cells were first induced toward neuronal cells, after which they were exposed to the hDPSC secretome. In addition, SH-SY5Y cells subjected to the hDPSC secretome showed increased neuritogenesis compared with nonexposed cells. Maturated cells were shown to increase immune reactivity for neuronal markers compared with controls. Ultrastructurally, retinoic acid (RA) signaling and subsequent exposure to the hDPSC secretome induced a gradual rise in metabolic activity and neuronal features such as multivesicular bodies and cytoskeletal elements associated with cellular communication. In addition, electrophysiological recordings of differentiating cells demonstrated a transition toward a neuronal electrophysiological profile based on the maximum tetrodotoxin (TTX)–sensitive, Na+ current. Moreover, conditioned medium (CM)–hDPSC–maturated SH-SY5Y cells developed distinct features including, Cd2+-sensitive currents, which suggests that CM-hDPSC–maturated SH-SY5Y acquired voltage-gated Ca2+ channels. The results reported in this study demonstrate the potential of hDPSCs to support differentiation and recruitment of cells with neuronal precursor characteristics in a paracrine manner. Moreover, this in vitro experimental design showed that the widely used SH-SY5Y cell line can improve and simplify the preclinical in vitro research on the molecular mechanisms of stem cell–mediated neuronal regeneration.
Collapse
Affiliation(s)
- P. Gervois
- Morphology Research Group, Biomedical Research Institute, Hasselt University, Diepenbeek, Belgium
| | - E. Wolfs
- Morphology Research Group, Biomedical Research Institute, Hasselt University, Diepenbeek, Belgium
| | - Y. Dillen
- Morphology Research Group, Biomedical Research Institute, Hasselt University, Diepenbeek, Belgium
| | - P. Hilkens
- Morphology Research Group, Biomedical Research Institute, Hasselt University, Diepenbeek, Belgium
| | - J. Ratajczak
- Morphology Research Group, Biomedical Research Institute, Hasselt University, Diepenbeek, Belgium
| | - R.B. Driesen
- Morphology Research Group, Biomedical Research Institute, Hasselt University, Diepenbeek, Belgium
| | - T. Vangansewinkel
- Morphology Research Group, Biomedical Research Institute, Hasselt University, Diepenbeek, Belgium
| | - A. Bronckaers
- Morphology Research Group, Biomedical Research Institute, Hasselt University, Diepenbeek, Belgium
| | - B. Brône
- Group of Physiology, Biomedical Research Institute, Hasselt University, Diepenbeek, Belgium
| | - T. Struys
- Morphology Research Group, Biomedical Research Institute, Hasselt University, Diepenbeek, Belgium
| | - I. Lambrichts
- Morphology Research Group, Biomedical Research Institute, Hasselt University, Diepenbeek, Belgium
| |
Collapse
|
45
|
Spider peptide toxin HwTx-IV engineered to bind to lipid membranes has an increased inhibitory potency at human voltage-gated sodium channel hNa V1.7. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2017; 1859:835-844. [PMID: 28115115 DOI: 10.1016/j.bbamem.2017.01.020] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/20/2016] [Revised: 01/03/2017] [Accepted: 01/19/2017] [Indexed: 01/15/2023]
Abstract
The human voltage-gated sodium channel sub-type 1.7 (hNaV1.7) is emerging as an attractive target for the development of potent and sub-type selective novel analgesics with increased potency and fewer side effects than existing therapeutics. HwTx-IV, a spider derived peptide toxin, inhibits hNaV1.7 with high potency and is therefore of great interest as an analgesic lead. In the current study we examined whether engineering a HwTx-IV analogue with increased ability to bind to lipid membranes would improve its inhibitory potency at hNaV1.7. This hypothesis was explored by comparing HwTx-IV and two analogues [E1PyrE]HwTx-IV (mHwTx-IV) and [E1G,E4G,F6W,Y30W]HwTx-IV (gHwTx-IV) on their membrane-binding affinity and hNaV1.7 inhibitory potency using a range of biophysical techniques including computational analysis, NMR spectroscopy, surface plasmon resonance, and fluorescence spectroscopy. HwTx-IV and mHwTx-IV exhibited weak affinity for lipid membranes, whereas gHwTx-IV showed improved affinity for the model membranes studied. In addition, activity assays using SH-SY5Y neuroblastoma cells expressing hNaV1.7 showed that gHwTx-IV has increased activity at hNaV1.7 compared to HwTx-IV. Based on these results we hypothesize that an increase in the affinity of HwTx-IV for lipid membranes is accompanied by improved inhibitory potency at hNaV1.7 and that increasing the affinity of gating modifier toxins to lipid bilayers is a strategy that may be useful for improving their potency at hNaV1.7.
Collapse
|
46
|
Rogers M, Zidar N, Kikelj D, Kirby RW. Characterization of Endogenous Sodium Channels in the ND7-23 Neuroblastoma Cell Line: Implications for Use as a Heterologous Ion Channel Expression System Suitable for Automated Patch Clamp Screening. Assay Drug Dev Technol 2016; 14:109-30. [PMID: 26991361 PMCID: PMC4800267 DOI: 10.1089/adt.2016.704] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
The rodent neuroblastoma cell line, ND7-23, is used to express voltage-dependent sodium (Nav) and other neuronal ion channels resistant to heterologous expression in Chinese hamster ovary (CHO) or human embryonic kidney (HEK) cells. Their advantage is that they provide endogenous factors and signaling pathways to promote ion channel peptide folding, expression, and function at the cell surface and are also amenable to automated patch clamping. However, ND7-23 cells exhibit endogenous tetrodotoxin (TTX)-sensitive Nav currents, and molecular profiling has revealed the presence of Nav1.2, Nav1.3, Nav1.6, and Nav1.7 transcripts, but no study has determined which subtypes contribute to functional channels at the cell surface. We profiled the repertoire of functional Nav channels endogenously expressed in ND7-23 cells using the QPatch automated patch clamp platform and selective toxins and small molecules. The potency and subtype selectivity of the ligands (Icagen compound 68 from patent US-20060025415-A1-20060202, 4,9 anhydro TTX, and Protoxin-II) were established in human Nav1.3, Nav1.6, and Nav1.7 channel cell lines before application of selective concentrations to ND7-23 cells. Our data confirm previous studies that >97% of macroscopic Nav current in ND7-23 cells is carried by TTX-sensitive channels (300 nM TTX) and that Nav1.7 is the predominant channel contributing to this response (65% of peak inward current), followed by Nav1.6 (∼20%) and negligible Nav1.3 currents (∼2%). In addition, our data are the first to assess the Nav1.6 potency (50% inhibitory concentration [IC50] of 33 nM) and selectivity (50-fold over Nav1.7) of 4,9 anhydro TTX in human Nav channels expressed in mammalian cells, confirming previous studies of rodent Nav channels expressed in oocytes and HEK cells.
Collapse
Affiliation(s)
- Marc Rogers
- 1 Xention Limited , Cambridge, United Kingdom
| | - Nace Zidar
- 2 Faculty of Pharmacy, University of Ljubljana , Ljubljana, Slovenia
| | - Danijel Kikelj
- 2 Faculty of Pharmacy, University of Ljubljana , Ljubljana, Slovenia
| | | |
Collapse
|
47
|
Touchard A, Brust A, Cardoso FC, Chin YKY, Herzig V, Jin AH, Dejean A, Alewood PF, King GF, Orivel J, Escoubas P. Isolation and characterization of a structurally unique β-hairpin venom peptide from the predatory ant Anochetus emarginatus. Biochim Biophys Acta Gen Subj 2016; 1860:2553-2562. [PMID: 27474999 DOI: 10.1016/j.bbagen.2016.07.027] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2016] [Revised: 06/24/2016] [Accepted: 07/26/2016] [Indexed: 12/18/2022]
Abstract
BACKGROUND Most ant venoms consist predominantly of small linear peptides, although some contain disulfide-linked peptides as minor components. However, in striking contrast to other ant species, some Anochetus venoms are composed primarily of disulfide-rich peptides. In this study, we investigated the venom of the ant Anochetus emarginatus with the aim of exploring these novel disulfide-rich peptides. METHODS The venom peptidome was initially investigated using a combination of reversed-phase HPLC and mass spectrometry, then the amino acid sequences of the major peptides were determined using a combination of Edman degradation and de novo MS/MS sequencing. We focused on one of these peptides, U1-PONTX-Ae1a (Ae1a), because of its novel sequence, which we predicted would form a novel 3D fold. Ae1a was chemically synthesized using Fmoc chemistry and its 3D structure was elucidated using NMR spectroscopy. The peptide was then tested for insecticidal activity and its effect on a range of human ion channels. RESULTS Seven peptides named poneritoxins (PONTXs) were isolated and sequenced. The three-dimensional structure of synthetic Ae1a revealed a novel, compact scaffold in which a C-terminal β-hairpin is connected to the N-terminal region via two disulfide bonds. Synthetic Ae1a reversibly paralyzed blowflies and inhibited human L-type voltage-gated calcium channels (CaV1). CONCLUSIONS Poneritoxins from Anochetus emarginatus venom are a novel class of toxins that are structurally unique among animal venoms. GENERAL SIGNIFICANCE This study demonstrates that Anochetus ant venoms are a rich source of novel ion channel modulating peptides, some of which might be useful leads for the development of biopesticides.
Collapse
Affiliation(s)
- Axel Touchard
- CNRS, UMR Ecologie des forêts de Guyane (AgroParisTech, CIRAD, CNRS, INRA, Université de Guyane, Université des Antilles), Campus Agronomique, BP 316, 97379 Kourou, France.
| | - Andreas Brust
- Institute for Molecular Bioscience, The University of Queensland, St Lucia, Queensland 4072, Australia
| | - Fernanda Caldas Cardoso
- Institute for Molecular Bioscience, The University of Queensland, St Lucia, Queensland 4072, Australia
| | - Yanni K-Y Chin
- Institute for Molecular Bioscience, The University of Queensland, St Lucia, Queensland 4072, Australia
| | - Volker Herzig
- Institute for Molecular Bioscience, The University of Queensland, St Lucia, Queensland 4072, Australia
| | - Ai-Hua Jin
- Institute for Molecular Bioscience, The University of Queensland, St Lucia, Queensland 4072, Australia
| | - Alain Dejean
- CNRS, UMR Ecologie des forêts de Guyane (AgroParisTech, CIRAD, CNRS, INRA, Université de Guyane, Université des Antilles), Campus Agronomique, BP 316, 97379 Kourou, France; CNRS, UMR 5245, Laboratoire Écologie Fonctionnelle et Environnement, 118 route de Narbonne, 31062 Toulouse, France; Université de Toulouse, UPS, INP, Ecolab, Toulouse, France
| | - Paul F Alewood
- Institute for Molecular Bioscience, The University of Queensland, St Lucia, Queensland 4072, Australia
| | - Glenn F King
- Institute for Molecular Bioscience, The University of Queensland, St Lucia, Queensland 4072, Australia
| | - Jérôme Orivel
- CNRS, UMR Ecologie des forêts de Guyane (AgroParisTech, CIRAD, CNRS, INRA, Université de Guyane, Université des Antilles), Campus Agronomique, BP 316, 97379 Kourou, France
| | | |
Collapse
|
48
|
Lewis RJ, Inserra M, Vetter I, Holland WC, Hardison DR, Tester PA, Litaker RW. Rapid Extraction and Identification of Maitotoxin and Ciguatoxin-Like Toxins from Caribbean and Pacific Gambierdiscus Using a New Functional Bioassay. PLoS One 2016; 11:e0160006. [PMID: 27467390 PMCID: PMC4965106 DOI: 10.1371/journal.pone.0160006] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2016] [Accepted: 07/12/2016] [Indexed: 11/18/2022] Open
Abstract
Background Ciguatera is a circumtropical disease produced by polyether sodium channel toxins (ciguatoxins) that enter the marine food chain and accumulate in otherwise edible fish. Ciguatoxins, as well as potent water-soluble polyethers known as maitotoxins, are produced by certain dinoflagellate species in the genus Gambierdiscus and Fukuyoa spp. in the Pacific but little is known of the potential of related Caribbean species to produce these toxins. Methods We established a simplified procedure for extracting polyether toxins from Gambierdiscus and Fukuyoa spp. based on the ciguatoxin rapid extraction method (CREM). Fractionated extracts from identified Pacific and Caribbean isolates were analysed using a functional bioassay that recorded intracellular calcium changes (Ca2+) in response to sample addition in SH-SY5Y cells. Maitotoxin directly elevated Ca2+i, while low levels of ciguatoxin-like toxins were detected using veratridine to enhance responses. Results We identified significant maitotoxin production in 11 of 12 isolates analysed, with 6 of 12 producing at least two forms of maitotoxin. In contrast, only 2 Caribbean isolates produced detectable levels of ciguatoxin-like activity despite a detection limit of >30 pM. Significant strain-dependent differences in the levels and types of ciguatoxins and maitotoxins produced by the same Gambierdiscus spp. were also identified. Conclusions The ability to rapidly identify polyether toxins produced by Gambierdiscus spp. in culture has the potential to distinguish ciguatoxin-producing species prior to large-scale culture and in naturally occurring blooms of Gambierdiscus and Fukuyoa spp. Our results have implications for the evaluation of ciguatera risk associated with Gambierdiscus and related species.
Collapse
Affiliation(s)
- Richard J. Lewis
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, 4072, Australia
- * E-mail:
| | - Marco Inserra
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, 4072, Australia
| | - Irina Vetter
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, 4072, Australia
| | - William C. Holland
- National Oceanic and Atmospheric Administration, National Ocean Service, Center for Coastal Fisheries & Habitat Research, 101 Pivers Island Road, Beaufort, NC, 28516, United States of America
| | - D. Ransom Hardison
- National Oceanic and Atmospheric Administration, National Ocean Service, Center for Coastal Fisheries & Habitat Research, 101 Pivers Island Road, Beaufort, NC, 28516, United States of America
| | - Patricia A. Tester
- National Oceanic and Atmospheric Administration, National Ocean Service, Center for Coastal Fisheries & Habitat Research, 101 Pivers Island Road, Beaufort, NC, 28516, United States of America
| | - R. Wayne Litaker
- National Oceanic and Atmospheric Administration, National Ocean Service, Center for Coastal Fisheries & Habitat Research, 101 Pivers Island Road, Beaufort, NC, 28516, United States of America
| |
Collapse
|
49
|
Henriques ST, Deplazes E, Lawrence N, Cheneval O, Chaousis S, Inserra M, Thongyoo P, King GF, Mark AE, Vetter I, Craik DJ, Schroeder CI. Interaction of Tarantula Venom Peptide ProTx-II with Lipid Membranes Is a Prerequisite for Its Inhibition of Human Voltage-gated Sodium Channel NaV1.7. J Biol Chem 2016; 291:17049-65. [PMID: 27311819 DOI: 10.1074/jbc.m116.729095] [Citation(s) in RCA: 57] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2016] [Indexed: 12/11/2022] Open
Abstract
ProTx-II is a disulfide-rich peptide toxin from tarantula venom able to inhibit the human voltage-gated sodium channel 1.7 (hNaV1.7), a channel reported to be involved in nociception, and thus it might have potential as a pain therapeutic. ProTx-II acts by binding to the membrane-embedded voltage sensor domain of hNaV1.7, but the precise peptide channel-binding site and the importance of membrane binding on the inhibitory activity of ProTx-II remain unknown. In this study, we examined the structure and membrane-binding properties of ProTx-II and several analogues using NMR spectroscopy, surface plasmon resonance, fluorescence spectroscopy, and molecular dynamics simulations. Our results show a direct correlation between ProTx-II membrane binding affinity and its potency as an hNaV1.7 channel inhibitor. The data support a model whereby a hydrophobic patch on the ProTx-II surface anchors the molecule at the cell surface in a position that optimizes interaction of the peptide with the binding site on the voltage sensor domain. This is the first study to demonstrate that binding of ProTx-II to the lipid membrane is directly linked to its potency as an hNaV1.7 channel inhibitor.
Collapse
Affiliation(s)
| | - Evelyne Deplazes
- From the Institute for Molecular Bioscience and School of Chemistry and Molecular Biosciences, University of Queensland, Queensland 4072 and
| | | | | | | | | | | | | | - Alan E Mark
- From the Institute for Molecular Bioscience and School of Chemistry and Molecular Biosciences, University of Queensland, Queensland 4072 and
| | - Irina Vetter
- From the Institute for Molecular Bioscience and the School of Pharmacy, University of Queensland, Queensland 4102, Australia
| | | | | |
Collapse
|
50
|
Yin K, Baillie GJ, Vetter I. Neuronal cell lines as model dorsal root ganglion neurons: A transcriptomic comparison. Mol Pain 2016; 12:1744806916646111. [PMID: 27130590 PMCID: PMC4956150 DOI: 10.1177/1744806916646111] [Citation(s) in RCA: 67] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2016] [Revised: 02/24/2016] [Accepted: 03/16/2016] [Indexed: 11/17/2022] Open
Abstract
BACKGROUND Dorsal root ganglion neuron-derived immortal cell lines including ND7/23 and F-11 cells have been used extensively as in vitro model systems of native peripheral sensory neurons. However, while it is clear that some sensory neuron-specific receptors and ion channels are present in these cell lines, a systematic comparison of the molecular targets expressed by these cell lines with those expressed in intact peripheral neurons is lacking. RESULTS In this study, we examined the expression of RNA transcripts in the human neuroblastoma-derived cell line, SH-SY5Y, and two dorsal root ganglion hybridoma cell lines, F-11 and ND7/23, using Illumina next-generation sequencing, and compared the results with native whole murine dorsal root ganglions. The gene expression profiles of these three cell lines did not resemble any specific defined dorsal root ganglion subclass. The cell lines lacked many markers for nociceptive sensory neurons, such as the Transient receptor potential V1 gene, but expressed markers for both myelinated and unmyelinated neurons. Global gene ontology analysis on whole dorsal root ganglions and cell lines showed similar enrichment of biological process terms across all samples. CONCLUSIONS This paper provides insights into the receptor repertoire expressed in common dorsal root ganglion neuron-derived cell lines compared with whole murine dorsal root ganglions, and illustrates the limits and potentials of these cell lines as tools for neuropharmacological exploration.
Collapse
Affiliation(s)
- Kathleen Yin
- Centre for Pain Research, Institute for Molecular Bioscience, University of Queensland, Queensland, Australia Pharmacy Australia Centre of Excellence, University of Queensland, Queensland, Australia
| | - Gregory J Baillie
- Centre for Pain Research, Institute for Molecular Bioscience, University of Queensland, Queensland, Australia
| | - Irina Vetter
- Centre for Pain Research, Institute for Molecular Bioscience, University of Queensland, Queensland, Australia Pharmacy Australia Centre of Excellence, University of Queensland, Queensland, Australia
| |
Collapse
|