1
|
Muller JAI, Bourke LA, Campbell SID, Cardoso FC. Venom peptides regulating Ca 2+ homeostasis: neuroprotective potential. Trends Pharmacol Sci 2025:S0165-6147(25)00047-1. [PMID: 40240234 DOI: 10.1016/j.tips.2025.03.007] [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: 12/18/2024] [Revised: 03/14/2025] [Accepted: 03/17/2025] [Indexed: 04/18/2025]
Abstract
Venom peptides specialized in modulating intracellular calcium ([Ca2+]i) offer a treasure trove of pharmacological properties to regulate aberrant Ca2+ homeostasis in disease. Combined with emerging advances across peptide optimization, disease models, and functional bioassays, these venom peptides could unlock new therapies restoring Ca2+ homeostasis. In this opinion, we explore the pharmacology of venom peptides modulating [Ca2+]i signaling along with recent breakthroughs propelling venom peptide-based drug discovery. We predict a transformative era in therapeutic development harnessing venom peptides targeting dysfunctional Ca2+ signaling in intractable conditions such as neurodegenerative diseases.
Collapse
Affiliation(s)
- Jessica A I Muller
- Institute for Molecular Bioscience, The University of Queensland, St. Lucia, Queensland, 4072, Australia
| | - Lachlan A Bourke
- School of the Environment, The University of Queensland, St. Lucia, Queensland, 4072, Australia
| | - Sam I D Campbell
- School of the Environment, 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.
| |
Collapse
|
2
|
Luo S, Zhou X, Wu M, Wang G, Wang L, Feng X, Wu H, Luo R, Lu M, Ju J, Wang W, Yuan L, Luo X, Peng D, Yang L, Zhang Q, Chen M, Liang S, Dong X, Hao G, Zhang Y, Liu Z. Optimizing Nav1.7-Targeted Analgesics: Revealing Off-Target Effects of Spider Venom-Derived Peptide Toxins and Engineering Strategies for Improvement. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2406656. [PMID: 39248322 PMCID: PMC11558128 DOI: 10.1002/advs.202406656] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/16/2024] [Revised: 08/22/2024] [Indexed: 09/10/2024]
Abstract
The inhibition of Nav1.7 is a promising strategy for the development of analgesic treatments. Spider venom-derived peptide toxins are recognized as significant sources of Nav1.7 inhibitors. However, their development has been impeded by limited selectivity. In this study, eight peptide toxins from three distinct spider venom Nav channel families demonstrated robust inhibition of hNav1.7, rKv4.2, and rKv4.3 (rKv4.2/4.3) currents, exhibiting a similar mode of action. The analysis of structure and function relationship revealed a significant overlap in the pharmacophore responsible for inhibiting hNav1.7 and rKv4.2 by HNTX-III, although Lys25 seems to play a more pivotal role in the inhibition of rKv4.2/4.3. Pharmacophore-guided rational design is employed for the development of an mGpTx1 analogue, mGpTx1-SA, which retains its inhibition of hNav1.7 while significantly reducing its inhibition of rKv4.2/4.3 and eliminating cardiotoxicity. Moreover, mGpTx1-SA demonstrates potent analgesic effects in both inflammatory and neuropathic pain models, accompanied by an improved in vivo safety profile. The results suggest that off-target inhibition of rKv4.2/4.3 by specific spider peptide toxins targeting hNav1.7 may arise from a conserved binding motif. This insight promises to facilitate the design of hNav1.7-specific analgesics, aimed at minimizing rKv4.2/4.3 inhibition and associated toxicity, thereby enhancing their suitability for therapeutic applications.
Collapse
|
3
|
Wang SY, Zhang YZ, Liu XH, Guo XC, Wang XF, Wang JR, Liu BJ, Han FT, Zhang Y, Wang CL. BNT12, a novel hybrid peptide of opioid and neurotensin pharmacophores, produces potent central antinociception with limited side effects. Eur J Pharmacol 2024; 978:176775. [PMID: 38925288 DOI: 10.1016/j.ejphar.2024.176775] [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: 01/22/2024] [Revised: 06/17/2024] [Accepted: 06/23/2024] [Indexed: 06/28/2024]
Abstract
The development of multitarget opioid drugs has emerged as an attractive approach for innovative pain management with reduced side effects. In the present study, a novel hybrid peptide BNT12 containing the opioid and neurotensin (NT)-like fragments was synthesized and pharmacologically characterized. In acute radiant heat paw withdrawal test, intracerebroventricular (i.c.v.) administration of BNT12 produced potent antinociception in mice. The central antinociceptive activity of BNT12 was mainly mediated by μ-, δ-opioid receptor, neurotensin receptor type 1 (NTSR1) and 2 (NTSR2), supporting a multifunctional agonism of BNT12 in the functional assays. BNT12 also exhibited significant antinociceptive effects in spared nerve injury (SNI)-neuropathic pain, complete Freund's adjuvant (CFA)-induced inflammatory pain, acetic acid-induced visceral and formalin-induced pain after i.c.v. administration. Furthermore, BNT12 exhibited substantial reduction of acute antinociceptive tolerance, shifted the dose-response curve to the right by only 1.3-fold. It is noteworthy that BNT12 showed insignificant chronic antinociceptive tolerance at the supraspinal level. In addition, BNT12 exhibited reduced or no opioid-like side effects on conditioned place preference (CPP) response, naloxone-precipitated withdrawal response, acute hyperlocomotion, motor coordination, gastrointestinal transit, and cardiovascular responses. The present investigation demonstrated that the novel hybrid peptide BNT12 might serve as a promising analgesic candidate with limited opioid-like side effects.
Collapse
Affiliation(s)
- Si-Yu Wang
- School of Life Science and Technology, Harbin Institute of Technology, 92 West Dazhi Street, Harbin, 150001, China
| | - Yu-Zhe Zhang
- School of Life Science and Technology, Harbin Institute of Technology, 92 West Dazhi Street, Harbin, 150001, China
| | - Xiao-Han Liu
- School of Life Science and Technology, Harbin Institute of Technology, 92 West Dazhi Street, Harbin, 150001, China
| | - Xue-Ci Guo
- School of Life Science and Technology, Harbin Institute of Technology, 92 West Dazhi Street, Harbin, 150001, China
| | | | - Jia-Ran Wang
- School of Life Science and Technology, Harbin Institute of Technology, 92 West Dazhi Street, Harbin, 150001, China
| | - Bing-Jie Liu
- School of Life Science and Technology, Harbin Institute of Technology, 92 West Dazhi Street, Harbin, 150001, China
| | - Feng-Tong Han
- School of Life Science and Technology, Harbin Institute of Technology, 92 West Dazhi Street, Harbin, 150001, China
| | - Yao Zhang
- School of Life Science and Technology, Harbin Institute of Technology, 92 West Dazhi Street, Harbin, 150001, China
| | - Chang-Lin Wang
- School of Life Science and Technology, Harbin Institute of Technology, 92 West Dazhi Street, Harbin, 150001, China; State Key Laboratory of Urban Water Resource and Environment, Harbin Institute of Technology, Harbin, 150090, China; State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources, Guangxi Normal University, Guilin, China.
| |
Collapse
|
4
|
Le Franc A, Da Silva A, Lepetre-Mouelhi S. Nanomedicine and voltage-gated sodium channel blockers in pain management: a game changer or a lost cause? Drug Deliv Transl Res 2024; 14:2112-2145. [PMID: 38861139 DOI: 10.1007/s13346-024-01615-9] [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] [Accepted: 04/25/2024] [Indexed: 06/12/2024]
Abstract
Pain, a complex and debilitating condition affecting millions globally, is a significant concern, especially in the context of post-operative recovery. This comprehensive review explores the complexity of pain and its global impact, emphasizing the modulation of voltage-gated sodium channels (VGSC or NaV channels) as a promising avenue for pain management with the aim of reducing reliance on opioids. The article delves into the role of specific NaV isoforms, particularly NaV 1.7, NaV 1.8, and NaV 1.9, in pain process and discusses the development of sodium channel blockers to target these isoforms precisely. Traditional local anesthetics and selective NaV isoform inhibitors, despite showing varying efficacy in pain management, face challenges in systemic distribution and potential side effects. The review highlights the potential of nanomedicine in improving the delivery of local anesthetics, toxins and selective NaV isoform inhibitors for a targeted and sustained release at the site of pain. This innovative strategy seeks to improve drug bioavailability, minimize systemic exposure, and optimize therapeutic outcomes, holding significant promise for secure pain management and enhancing the quality of life for individuals recovering from surgical procedures or suffering from chronic pain.
Collapse
Affiliation(s)
- Adélaïde Le Franc
- Université Paris-Saclay, CNRS, Institut Galien Paris-Saclay, 91400, Orsay, France
| | - Alexandre Da Silva
- Université Paris-Saclay, CNRS, Institut Galien Paris-Saclay, 91400, Orsay, France
| | | |
Collapse
|
5
|
Kramer S, Kotapati C, Cao Y, Fry BG, Palpant NJ, King GF, Cardoso FC. High-content fluorescence bioassay investigates pore formation, ion channel modulation and cell membrane lysis induced by venoms. Toxicon X 2024; 21:100184. [PMID: 38389571 PMCID: PMC10882159 DOI: 10.1016/j.toxcx.2024.100184] [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: 08/15/2023] [Revised: 01/07/2024] [Accepted: 01/30/2024] [Indexed: 02/24/2024] Open
Abstract
Venoms comprise highly sophisticated bioactive molecules modulating ion channels, receptors, coagulation factors, and the cellular membranes. This array of targets and bioactivities requires advanced high-content bioassays to facilitate the development of novel envenomation treatments and biotechnological and pharmacological agents. In response to the existing gap in venom research, we developed a cutting-edge fluorescence-based high-throughput and high-content cellular assay. This assay enables the simultaneous identification of prevalent cellular activities induced by venoms such as membrane lysis, pore formation, and ion channel modulation. By integrating intracellular calcium with extracellular nucleic acid measurements, we have successfully distinguished these venom mechanisms within a single cellular assay. Our high-content bioassay was applied across three cell types exposed to venom components representing lytic, ion pore-forming or ion channel modulator toxins. Beyond unveiling distinct profiles for these action mechanisms, we found that the pore-forming latrotoxin α-Lt1a prefers human neuroblastoma to kidney cells and cardiomyocytes, while the lytic bee peptide melittin is not selective. Furthermore, evaluation of snake venoms showed that Elapid species induced rapid membrane lysis, while Viper species showed variable to no activity on neuroblastoma cells. These findings underscore the ability of our high-content bioassay to discriminate between clades and interspecific traits, aligning with clinical observations at venom level, beyond discriminating among ion pore-forming, membrane lysis and ion channel modulation. We hope our research will expedite the comprehension of venom biology and the diversity of toxins that elicit cytotoxic, cardiotoxic and neurotoxic effects, and assist in identifying venom components that hold the potential to benefit humankind.
Collapse
Affiliation(s)
- Simon Kramer
- Institute for Molecular Bioscience, The University of Queensland, St Lucia, Brisbane, Queensland, Australia, 4072
| | - Charan Kotapati
- Institute for Molecular Bioscience, The University of Queensland, St Lucia, Brisbane, Queensland, Australia, 4072
| | - Yuanzhao Cao
- Institute for Molecular Bioscience, The University of Queensland, St Lucia, Brisbane, Queensland, Australia, 4072
| | - Bryan G Fry
- School of Biological Sciences, The University of Queensland, St Lucia, Brisbane, Queensland, Australia, 4072
| | - Nathan J Palpant
- Institute for Molecular Bioscience, The University of Queensland, St Lucia, Brisbane, Queensland, Australia, 4072
| | - Glenn F King
- Institute for Molecular Bioscience, The University of Queensland, St Lucia, Brisbane, Queensland, Australia, 4072
| | - Fernanda C Cardoso
- Institute for Molecular Bioscience, The University of Queensland, St Lucia, Brisbane, Queensland, Australia, 4072
| |
Collapse
|
6
|
Yu N, Yan Y, Han Q, Zhang L, Liu Z. Insecticidal toxicity of ω-Atypitoxin-Cs1a and its inhibitory effects on insect voltage-gated calcium channels. PEST MANAGEMENT SCIENCE 2023; 79:4879-4885. [PMID: 37506304 DOI: 10.1002/ps.7689] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Revised: 07/15/2023] [Accepted: 07/29/2023] [Indexed: 07/30/2023]
Abstract
BACKGROUND Excessive use of chemical insecticides raises concerns about insecticide resistance, urging the development of novel insecticides. Peptide neurotoxins from spider venom are an incredibly rich source of ion channel modulators with potent insecticidal activity. A neurotoxin U1-Atypitoxin-Cs1a from the spider Calommata signata was annotated previously. It was of interest to investigate its insecticidal activity and potential molecular targets. RESULTS Cs1a was heterologously expressed, purified and pharmacologically characterized here. The recombinant neurotoxin inhibited high-voltage-activated calcium channel currents with an median inhibitory concentration (IC50 ) value of 0.182 ± 0.026 μm on cockroach DUM neurons and thus was designated as ω-Atypitoxin-Cs1a. The recombinant Cs1a was toxic to three insect pests of agricultural importance, Nilaparvata lugens, Spodoptera frugiperda and Plutella xylostella with median lethal concentration (LD50 ) values of 0.121, 0.172 and 0.356 nmol g-1 , respectively, at 24 h postinjection. Cs1a was equivalently toxic to both insecticide-susceptible and -resistant insects. Cs1a exhibited low toxicity to Danio rerio with an LD50 of 2.316 nmol g-1 . CONCLUSION Our results suggest that ω-Atypitoxin-Cs1a is a potent CaV channel inhibitor and an attractive candidate reagent for pest control and resistance management. © 2023 Society of Chemical Industry.
Collapse
Affiliation(s)
- Na Yu
- Key Laboratory of Integrated Management of Crop Diseases and Pests (Ministry of Education), College of Plant Protection, Nanjing Agricultural University, Nanjing, China
| | - Yangyang Yan
- Key Laboratory of Integrated Management of Crop Diseases and Pests (Ministry of Education), College of Plant Protection, Nanjing Agricultural University, Nanjing, China
| | - Qianqian Han
- Key Laboratory of Integrated Management of Crop Diseases and Pests (Ministry of Education), College of Plant Protection, Nanjing Agricultural University, Nanjing, China
| | - Lingchun Zhang
- Key Laboratory of Integrated Management of Crop Diseases and Pests (Ministry of Education), College of Plant Protection, Nanjing Agricultural University, Nanjing, China
| | - Zewen Liu
- Key Laboratory of Integrated Management of Crop Diseases and Pests (Ministry of Education), College of Plant Protection, Nanjing Agricultural University, Nanjing, China
| |
Collapse
|
7
|
Ahmadi S, Benard-Valle M, Boddum K, Cardoso FC, King GF, Laustsen AH, Ljungars A. From squid giant axon to automated patch-clamp: electrophysiology in venom and antivenom research. Front Pharmacol 2023; 14:1249336. [PMID: 37693897 PMCID: PMC10484000 DOI: 10.3389/fphar.2023.1249336] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2023] [Accepted: 08/11/2023] [Indexed: 09/12/2023] Open
Abstract
Ion channels play a crucial role in diverse physiological processes, including neurotransmission and muscle contraction. Venomous creatures exploit the vital function of ion channels by producing toxins in their venoms that specifically target these ion channels to facilitate prey capture upon a bite or a sting. Envenoming can therefore lead to ion channel dysregulation, which for humans can result in severe medical complications that often necessitate interventions such as antivenom administration. Conversely, the discovery of highly potent and selective venom toxins with the capability of distinguishing between different isoforms and subtypes of ion channels has led to the development of beneficial therapeutics that are now in the clinic. This review encompasses the historical evolution of electrophysiology methodologies, highlighting their contributions to venom and antivenom research, including venom-based drug discovery and evaluation of antivenom efficacy. By discussing the applications and advancements in patch-clamp techniques, this review underscores the profound impact of electrophysiology in unravelling the intricate interplay between ion channels and venom toxins, ultimately leading to the development of drugs for envenoming and ion channel-related pathologies.
Collapse
Affiliation(s)
- Shirin Ahmadi
- Department of Biotechnology and Biomedicine, Technical University of Denmark, Kongens Lyngby, Denmark
| | - Melisa Benard-Valle
- Department of Biotechnology and Biomedicine, Technical University of Denmark, Kongens Lyngby, Denmark
| | | | - Fernanda C. Cardoso
- Institute for Molecular Bioscience, University of Queensland, St Lucia, QLD, Australia
- Australian Research Council Centre of Excellence for Innovations in Protein and Peptide Science, University of Queensland, St Lucia, QLD, Australia
| | - Glenn F. King
- Institute for Molecular Bioscience, University of Queensland, St Lucia, QLD, Australia
- Australian Research Council Centre of Excellence for Innovations in Protein and Peptide Science, University of Queensland, St Lucia, QLD, Australia
| | - Andreas Hougaard Laustsen
- Department of Biotechnology and Biomedicine, Technical University of Denmark, Kongens Lyngby, Denmark
| | - Anne Ljungars
- Department of Biotechnology and Biomedicine, Technical University of Denmark, Kongens Lyngby, Denmark
| |
Collapse
|
8
|
Tran P, Crawford T, Ragnarsson L, Deuis JR, Mobli M, Sharpe SJ, Schroeder CI, Vetter I. Structural Conformation and Activity of Spider-Derived Inhibitory Cystine Knot Peptide Pn3a Are Modulated by pH. ACS OMEGA 2023; 8:26276-26286. [PMID: 37521635 PMCID: PMC10373202 DOI: 10.1021/acsomega.3c02664] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/18/2023] [Accepted: 06/22/2023] [Indexed: 08/01/2023]
Abstract
Numerous spider venom-derived gating modifier toxins exhibit conformational heterogeneity during purification by reversed-phase high-performance liquid chromatography (RP-HPLC). This conformational exchange is especially peculiar for peptides containing an inhibitor cystine knot motif, which confers excellent structural stability under conditions that are not conducive to disulfide shuffling. This phenomenon is often attributed to proline cis/trans isomerization but has also been observed in peptides that do not contain a proline residue. Pn3a is one such peptide forming two chromatographically distinguishable peaks that readily interconvert following the purification of either conformer. The nature of this exchange was previously uncharacterized due to the fast rate of conversion in solution, making isolation of the conformers impossible. In the present study, an N-terminal modification of Pn3a enabled the isolation of the individual conformers, allowing activity assays to be conducted on the individual conformers using electrophysiology. The conformers were analyzed separately by nuclear magnetic resonance spectroscopy (NMR) to study their structural differences. RP-HPLC and NMR were used to study the mechanism of exchange. The later-eluting conformer was the active conformer with a rigid structure that corresponds to the published structure of Pn3a, while NMR analysis revealed the earlier-eluting conformer to be inactive and disordered. The exchange was found to be pH-dependent, arising in acidic solutions, possibly due to reversible disruption and formation of intramolecular salt bridges. This study reveals the nature of non-proline conformational exchange observed in Pn3a and possibly other disulfide-rich peptides, highlighting that the structure and activity of some disulfide-stabilized peptides can be dramatically susceptible to disruption.
Collapse
Affiliation(s)
- Poanna Tran
- Institute
for Molecular Bioscience, The University
of Queensland, Brisbane, Queensland 4072, Australia
| | - Theo Crawford
- Centre
for Advanced Imaging, The University of
Queensland, Brisbane, Queensland 4072, Australia
| | - Lotten Ragnarsson
- Institute
for Molecular Bioscience, The University
of Queensland, Brisbane, Queensland 4072, Australia
| | - Jennifer R. Deuis
- Institute
for Molecular Bioscience, The University
of Queensland, Brisbane, Queensland 4072, Australia
| | - Mehdi Mobli
- Centre
for Advanced Imaging, The University of
Queensland, Brisbane, Queensland 4072, Australia
| | - Simon J. Sharpe
- Molecular
Medicine Program, Research Institute, The
Hospital for Sick Children, Toronto, Ontario M5G 0A4, Canada
- Department
of Biochemistry, University of Toronto, Toronto, Ontario M5S 1A8, Canada
| | - Christina I. Schroeder
- Institute
for Molecular Bioscience, The University
of Queensland, Brisbane, Queensland 4072, Australia
- Center
for Cancer Research, National Cancer Institute, National Institutes of Health, Frederick, Maryland 21702-1201, United States
- Genentech, 1 DNA Way, South San Francisco, California 94080, United States
| | - Irina Vetter
- Institute
for Molecular Bioscience, The University
of Queensland, Brisbane, Queensland 4072, Australia
- School
of Pharmacy, The University of Queensland, Brisbane, Queensland 4102, Australia
| |
Collapse
|
9
|
Antunes FTT, Campos MM, Carvalho VDPR, da Silva Junior CA, Magno LAV, de Souza AH, Gomez MV. Current Drug Development Overview: Targeting Voltage-Gated Calcium Channels for the Treatment of Pain. Int J Mol Sci 2023; 24:ijms24119223. [PMID: 37298174 DOI: 10.3390/ijms24119223] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2023] [Revised: 04/29/2023] [Accepted: 05/02/2023] [Indexed: 06/12/2023] Open
Abstract
Voltage-gated calcium channels (VGCCs) are targeted to treat pain conditions. Since the discovery of their relation to pain processing control, they are investigated to find new strategies for better pain control. This review provides an overview of naturally based and synthetic VGCC blockers, highlighting new evidence on the development of drugs focusing on the VGCC subtypes as well as mixed targets with pre-clinical and clinical analgesic effects.
Collapse
Affiliation(s)
- Flavia Tasmin Techera Antunes
- Department of Physiology and Pharmacology, University of Calgary, Calgary, AB T2N 1N4, Canada
- Hotchkiss Brain Institute, University of Calgary, Calgary, AB T2N 1N4, Canada
| | - Maria Martha Campos
- Programa de Pós-Graduação em Odontologia, Escola de Ciências da Saúde e da Vida, Pontifícia Universidade Católica do Rio Grande do Sul, Porto Alegre 90619-900, RS, Brazil
| | | | | | - Luiz Alexandre Viana Magno
- Programa de Pós-Graduação em Ciências da Saúde, Faculdade Ciências Médicas de Minas Gerais (FCMMG), Belo Horizonte 30110-005, MG, Brazil
| | - Alessandra Hubner de Souza
- Programa de Pós-Graduação em Ciências da Saúde, Faculdade Ciências Médicas de Minas Gerais (FCMMG), Belo Horizonte 30110-005, MG, Brazil
| | | |
Collapse
|
10
|
Cardoso FC, Walker AA, King GF, Gomez MV. Holistic profiling of the venom from the Brazilian wandering spider Phoneutria nigriventer by combining high-throughput ion channel screens with venomics. Front Mol Biosci 2023; 10:1069764. [PMID: 36865382 PMCID: PMC9972223 DOI: 10.3389/fmolb.2023.1069764] [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: 10/14/2022] [Accepted: 01/30/2023] [Indexed: 02/16/2023] Open
Abstract
Introduction: Spider venoms are a unique source of bioactive peptides, many of which display remarkable biological stability and neuroactivity. Phoneutria nigriventer, often referred to as the Brazilian wandering spider, banana spider or "armed" spider, is endemic to South America and amongst the most dangerous venomous spiders in the world. There are 4,000 envenomation accidents with P. nigriventer each year in Brazil, which can lead to symptoms including priapism, hypertension, blurred vision, sweating, and vomiting. In addition to its clinical relevance, P. nigriventer venom contains peptides that provide therapeutic effects in a range of disease models. Methods: In this study, we explored the neuroactivity and molecular diversity of P. nigriventer venom using fractionation-guided high-throughput cellular assays coupled to proteomics and multi-pharmacology activity to broaden the knowledge about this venom and its therapeutic potential and provide a proof-of-concept for an investigative pipeline to study spider-venom derived neuroactive peptides. We coupled proteomics with ion channel assays using a neuroblastoma cell line to identify venom compounds that modulate the activity of voltage-gated sodium and calcium channels, as well as the nicotinic acetylcholine receptor. Results: Our data revealed that P. nigriventer venom is highly complex compared to other neurotoxin-rich venoms and contains potent modulators of voltage-gated ion channels which were classified into four families of neuroactive peptides based on their activity and structures. In addition to the reported P. nigriventer neuroactive peptides, we identified at least 27 novel cysteine-rich venom peptides for which their activity and molecular target remains to be determined. Discussion: Our findings provide a platform for studying the bioactivity of known and novel neuroactive components in the venom of P. nigriventer and other spiders and suggest that our discovery pipeline can be used to identify ion channel-targeting venom peptides with potential as pharmacological tools and to drug leads.
Collapse
Affiliation(s)
- F. C. Cardoso
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Australia,Centre of Excellence for Innovations in Peptide and Protein Science, The University of Queensland, Brisbane, Australia,*Correspondence: F. C. Cardoso,
| | - A. A. Walker
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Australia,Centre of Excellence for Innovations in Peptide and Protein Science, The University of Queensland, Brisbane, Australia
| | - G. F. King
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Australia,Centre of Excellence for Innovations in Peptide and Protein Science, The University of Queensland, Brisbane, Australia
| | - M. V. Gomez
- Department of Neurotransmitters, Institute of Education and Research, Santa Casa, Belo Horizonte, Brazil
| |
Collapse
|
11
|
Shin A, Kashyap PC. Multi-omics for biomarker approaches in the diagnostic evaluation and management of abdominal pain and irritable bowel syndrome: what lies ahead. Gut Microbes 2023; 15:2195792. [PMID: 37009874 PMCID: PMC10072066 DOI: 10.1080/19490976.2023.2195792] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/20/2022] [Accepted: 03/23/2023] [Indexed: 04/04/2023] Open
Abstract
Reliable biomarkers for common disorders of gut-brain interaction characterized by abdominal pain, including irritable bowel syndrome (IBS), are critically needed to enhance care and develop individualized therapies. The dynamic and heterogeneous nature of the pathophysiological mechanisms that underlie visceral hypersensitivity have challenged successful biomarker development. Consequently, effective therapies for pain in IBS are lacking. However, recent advances in modern omics technologies offer new opportunities to acquire deep biological insights into mechanisms of pain and nociception. Newer methods for large-scale data integration of complementary omics approaches have further expanded our ability to build a holistic understanding of complex biological networks and their co-contributions to abdominal pain. Here, we review the mechanisms of visceral hypersensitivity, focusing on IBS. We discuss candidate biomarkers for pain in IBS identified through single omics studies and summarize emerging multi-omics approaches for developing novel biomarkers that may transform clinical care for patients with IBS and abdominal pain.
Collapse
Affiliation(s)
- Andrea Shin
- Division of Gastroenterology and Hepatology, Department of Medicine, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Purna C. Kashyap
- Clinical Enteric Neuroscience Translational and Epidemiological Research Program, Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN, USA
| |
Collapse
|
12
|
The Deadly Toxin Arsenal of the Tree-Dwelling Australian Funnel-Web Spiders. Int J Mol Sci 2022; 23:ijms232113077. [PMID: 36361863 PMCID: PMC9658043 DOI: 10.3390/ijms232113077] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2022] [Revised: 10/18/2022] [Accepted: 10/24/2022] [Indexed: 11/23/2022] Open
Abstract
Australian funnel-web spiders are amongst the most dangerous venomous animals. Their venoms induce potentially deadly symptoms, including hyper- and hypotension, tachycardia, bradycardia and pulmonary oedema. Human envenomation is more frequent with the ground-dwelling species, including the infamous Sydney funnel-web spider (Atrax robustus); although, only two tree-dwelling species induce more severe envenomation. To unravel the mechanisms that lead to this stark difference in clinical outcomes, we investigated the venom transcriptome and proteome of arboreal Hadronyche cerberea and H. formidabilis. Overall, Hadronyche venoms comprised 44 toxin superfamilies, with 12 being exclusive to tree-dwellers. Surprisingly, the major venom components were neprilysins and uncharacterized peptides, in addition to the well-known ω- and δ-hexatoxins and double-knot peptides. The insecticidal effects of Hadronyche venom on sheep blowflies were more potent than Atrax venom, and the venom of both tree- and ground-dwelling species potently modulated human voltage-gated sodium channels, particularly NaV1.2. Only the venom of tree-dwellers exhibited potent modulation of voltage-gated calcium channels. H. formidabilis appeared to be under less diversifying selection pressure compared to the newly adapted tree-dweller, H. cerberea. Thus, this study contributes to unravelling the fascinating molecular and pharmacological basis for the severe envenomation caused by the Australian tree-dwelling funnel-web spiders.
Collapse
|
13
|
The T-type calcium channel Ca V 3.2 regulates bladder afferent responses to mechanical stimuli. Pain 2022; 164:1012-1026. [PMID: 36279179 PMCID: PMC10108591 DOI: 10.1097/j.pain.0000000000002795] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2021] [Accepted: 09/09/2022] [Indexed: 11/06/2022]
Abstract
ABSTRACT The bladder wall is innervated by a complex network of afferent nerves that detect bladder stretch during filling. Sensory signals, generated in response to distension, are relayed to the spinal cord and brain to evoke physiological and painful sensations and regulate urine storage and voiding. Hyperexcitability of these sensory pathways is a key component in the development of chronic bladder hypersensitivity disorders including interstitial cystitis/bladder pain syndrome and overactive bladder syndrome. Despite this, the full array of ion channels that regulate bladder afferent responses to mechanical stimuli have yet to be determined. Here, we investigated the role of low-voltage-activated T-type calcium (Ca V 3) channels in regulating bladder afferent responses to distension. Using single-cell reverse-transcription polymerase chain reaction and immunofluorescence, we revealed ubiquitous expression of Ca V 3.2, but not Ca V 3.1 or Ca V 3.3, in individual bladder-innervating dorsal root ganglia neurons. Pharmacological inhibition of Ca V 3.2 with TTA-A2 and ABT-639, selective blockers of T-type calcium channels, dose-dependently attenuated ex-vivo bladder afferent responses to distension in the absence of changes to muscle compliance. Further evaluation revealed that Ca V 3.2 blockers significantly inhibited both low- and high-threshold afferents, decreasing peak responses to distension, and delayed activation thresholds, thereby attenuating bladder afferent responses to both physiological and noxious distension. Nocifensive visceromotor responses to noxious bladder distension in vivo were also significantly reduced by inhibition of Ca V 3 with TTA-A2. Together, these data provide evidence of a major role for Ca V 3.2 in regulating bladder afferent responses to bladder distension and nociceptive signalling to the spinal cord.
Collapse
|
14
|
LMWP (S3-3) from the Larvae of Musca domestica Alleviate D-IBS by Adjusting the Gut Microbiota. Molecules 2022; 27:molecules27144517. [PMID: 35889391 PMCID: PMC9324334 DOI: 10.3390/molecules27144517] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2022] [Revised: 07/03/2022] [Accepted: 07/11/2022] [Indexed: 11/17/2022] Open
Abstract
Diarrhea-based Irritable Bowel Syndrome (D-IBS) and diarrhea are both associated with ecological imbalance of the gut microbiota. Low Molecular Weight Peptides (LMWP) from the larvae of Musca domestica have been shown to be effective in the treatment of diarrhea and regulation of gut microbiota. Meanwhile, the single polypeptide S3-3 was successfully isolated and identified from LMWP in our previous studies. It remains unclear exactly whether and how LMWP (S3-3) alleviate D-IBS through regulating gut microbiota. We evaluated the gut microbiota and pharmacology to determine the regulation of gut microbiota structure and the alleviating effect on D-IBS through LMWP (S3-3). The rates of loose stools, abdominal withdrawal reflex (AWR) and intestinal tract motility results revealed that LMWP (S3-3) from the larvae of Musca domestica had a regulating effect against diarrhea, visceral hypersensitivity and gastrointestinal (GI) dysfunction in D-IBS model mice. Additionally, 16S rRNA gene sequencing was utilized to examine the gut microbiota, which suggests that LMWP induce structural changes in the gut microbiota and alter the levels of the following gut microbiota: Bacteroidetes, Proteobacteria and Verrucomicrobia. LMWP putatively functioned through regulating 5-HT, SERT, 5-HT2AR, 5-HT3AR and 5-HT4R according to the results of ELISA, qRT-PCR and IHC. The findings of this study will contribute to further understanding how LMWP (S3-3) attenuate the effects of D-IBS on diarrhea, visceral hypersensitivity and GI dysfunction.
Collapse
|
15
|
Cardoso FC, Servent D, de Lima ME. Editorial: Venom Peptides: A Rich Combinatorial Library for Drug Development. Front Mol Biosci 2022; 9:924023. [PMID: 35647027 PMCID: PMC9136682 DOI: 10.3389/fmolb.2022.924023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2022] [Accepted: 04/27/2022] [Indexed: 12/02/2022] Open
Affiliation(s)
- Fernanda C. Cardoso
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD, Australia
- *Correspondence: Fernanda C. Cardoso,
| | - Denis Servent
- CEA, Département Médicaments et Technologies pour La Santé (DMTS), SIMoS, Université Paris-Saclay, Gif-sur-Yvette, France
| | - Maria Elena de Lima
- Programa de Pós Graduação em Medicina e Biomedicina, Santa Casa de Belo Horizonte, Belo Horizonte, Brazil
- Departamento de Bioquímica e Imunologia, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| |
Collapse
|
16
|
da Silva CN, Nunes KP, Dourado LFN, Vieira TO, Mariano XM, Cunha Junior ADS, de Lima ME. From the PnTx2-6 Toxin to the PnPP-19 Engineered Peptide: Therapeutic Potential in Erectile Dysfunction, Nociception, and Glaucoma. Front Mol Biosci 2022; 9:831823. [PMID: 35480885 PMCID: PMC9035689 DOI: 10.3389/fmolb.2022.831823] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2021] [Accepted: 03/21/2022] [Indexed: 11/13/2022] Open
Abstract
The venom of the “armed” spider Phoneutria nigriventer comprises several potent toxins. One of the most toxic components from this venom is the neurotoxin PnTx2-6 (LD50 = ∼ 0.7 μg/mouse, 48 residues, five disulfide bridges, MW = 5,289.31 Da), which slows down the inactivation of various Na+ channels. In mice and rats, this toxin causes priapism, an involuntary and painful erection, similar to what is observed in humans bitten by P. nigriventer. While not completely elucidated, it is clear that PnTx2-6 potentiates erectile function via NO/cGMP signaling, but it has many off-target effects. Seeking to obtain a simpler and less toxic molecule able to retain the pharmacological properties of this toxin, we designed and synthesized the peptide PnPP-19 (19 residues, MW = 2,485.6 Da), representing a discontinuous epitope of PnTx2-6. This synthetic peptide also potentiates erectile function via NO/cGMP, but it does not target Na+ channels, and therefore, it displays nontoxic properties in animals even at high doses. PnPP-19 effectively potentiates erectile function not only after subcutaneous or intravenous administration but also following topical application. Surprisingly, PnPP-19 showed central and peripheral antinociceptive activity involving the opioid and cannabinoid systems, suggesting applicability in nociception. Furthermore, considering that PnPP-19 increases NO availability in the corpus cavernosum, this peptide was also tested in a model of induced intraocular hypertension, characterized by low NO levels, and it showed promising results by decreasing the intraocular pressure which prevents retinal damage. Herein, we discuss how was engineered this smaller active non-toxic peptide with promising results in the treatment of erectile dysfunction, nociception, and glaucoma from the noxious PnTx2-6, as well as the pitfalls of this ongoing journey.
Collapse
Affiliation(s)
- Carolina Nunes da Silva
- Departmentamento de Bioquímica e Imunologia, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
- Faculdade de Farmácia, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
- *Correspondence: Maria Elena de Lima, ; Carolina Nunes da Silva, ; Kenia Pedrosa Nunes,
| | - Kenia Pedrosa Nunes
- Department of Biomedical and Chemical Engineering and Sciences, Florida Institute of Technology, Melbourne, FL, United States
- *Correspondence: Maria Elena de Lima, ; Carolina Nunes da Silva, ; Kenia Pedrosa Nunes,
| | | | - Thayllon Oliveira Vieira
- Programa de Pós-Graduação em Medicina e Biomedicina Faculdade Santa Casa de Belo Horizonte, Belo Horizonte, Brazil
| | - Xavier Maia Mariano
- Programa de Pós-Graduação em Medicina e Biomedicina Faculdade Santa Casa de Belo Horizonte, Belo Horizonte, Brazil
| | | | - Maria Elena de Lima
- Departmentamento de Bioquímica e Imunologia, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
- Programa de Pós-Graduação em Medicina e Biomedicina Faculdade Santa Casa de Belo Horizonte, Belo Horizonte, Brazil
- *Correspondence: Maria Elena de Lima, ; Carolina Nunes da Silva, ; Kenia Pedrosa Nunes,
| |
Collapse
|
17
|
Contrasting patterns of venom regeneration in a centipede (Scolopendra viridis) and a scorpion (Centruroides hentzi). Toxicon 2022; 210:132-140. [PMID: 35245607 DOI: 10.1016/j.toxicon.2022.02.022] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2021] [Revised: 02/22/2022] [Accepted: 02/28/2022] [Indexed: 11/23/2022]
Abstract
As biochemical traits with clear fitness consequences, venoms serve a critical ecological role for the animals that produce them. Understanding how venoms are maintained and regenerated after use will, therefore, provide valuable insight into the ecology of venomous animals. Furthermore, most studies on venomous organisms often require removing animals from the wild and waiting extended periods of time between venom extractions. Uncovering the patterns of venom regeneration across different species will likely lead to the development of more efficient venom extraction protocols, reducing both experimental time and the number of animals required. Using reversed-phase high-performance liquid chromatography, we identified asynchronous regeneration of venom protein component abundances in the centipede Scolopendra viridis but found no evidence for asynchronous venom regeneration in the scorpion Centruroides hentzi. We also observed high levels of intraspecific venom variation in C. hentzi, emphasizing the importance of testing for intraspecific venom variation in studies evaluating the synchronicity of venom regeneration. Although the regeneration of relative venom protein component abundances is an asynchronous process in S. viridis, we provide evidence that the presence-absence of major venom components is not an asynchronous process and suggest that studies relying on just the presence/absence of individual proteins (e.g. bioprospecting, drug discovery) could use catch-and-release methods of venom extraction to reduce the number of animals removed from the wild.
Collapse
|
18
|
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
|
19
|
Diochot S. Pain-related toxins in scorpion and spider venoms: a face to face with ion channels. J Venom Anim Toxins Incl Trop Dis 2021; 27:e20210026. [PMID: 34925480 PMCID: PMC8667759 DOI: 10.1590/1678-9199-jvatitd-2021-0026] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2021] [Revised: 05/10/2021] [Indexed: 12/12/2022] Open
Abstract
Pain is a common symptom induced during envenomation by spiders and scorpions.
Toxins isolated from their venom have become essential tools for studying the
functioning and physiopathological role of ion channels, as they modulate their
activity. In particular, toxins that induce pain relief effects can serve as a
molecular basis for the development of future analgesics in humans. This review
provides a summary of the different scorpion and spider toxins that directly
interact with pain-related ion channels, with inhibitory or stimulatory effects.
Some of these toxins were shown to affect pain modalities in different animal
models providing information on the role played by these channels in the pain
process. The close interaction of certain gating-modifier toxins with membrane
phospholipids close to ion channels is examined along with molecular approaches
to improve selectivity, affinity or bioavailability in vivo for
therapeutic purposes.
Collapse
Affiliation(s)
- Sylvie Diochot
- Institut de Pharmacologie Moléculaire et Cellulaire (IPMC), Centre National de la Recherche Scientifique (CNRS) UMR 7275 et Université Côte d'Azur (UCA), 06560 Valbonne, France. Institut de Pharmacologie Moléculaire et Cellulaire Centre National de la Recherche Scientifique Université Côte d'Azur Valbonne France
| |
Collapse
|
20
|
Alles SRA, Smith PA. Peripheral Voltage-Gated Cation Channels in Neuropathic Pain and Their Potential as Therapeutic Targets. FRONTIERS IN PAIN RESEARCH 2021; 2:750583. [PMID: 35295464 PMCID: PMC8915663 DOI: 10.3389/fpain.2021.750583] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Accepted: 11/10/2021] [Indexed: 11/25/2022] Open
Abstract
The persistence of increased excitability and spontaneous activity in injured peripheral neurons is imperative for the development and persistence of many forms of neuropathic pain. This aberrant activity involves increased activity and/or expression of voltage-gated Na+ and Ca2+ channels and hyperpolarization activated cyclic nucleotide gated (HCN) channels as well as decreased function of K+ channels. Because they display limited central side effects, peripherally restricted Na+ and Ca2+ channel blockers and K+ channel activators offer potential therapeutic approaches to pain management. This review outlines the current status and future therapeutic promise of peripherally acting channel modulators. Selective blockers of Nav1.3, Nav1.7, Nav1.8, Cav3.2, and HCN2 and activators of Kv7.2 abrogate signs of neuropathic pain in animal models. Unfortunately, their performance in the clinic has been disappointing; some substances fail to meet therapeutic end points whereas others produce dose-limiting side effects. Despite this, peripheral voltage-gated cation channels retain their promise as therapeutic targets. The way forward may include (i) further structural refinement of K+ channel activators such as retigabine and ASP0819 to improve selectivity and limit toxicity; use or modification of Na+ channel blockers such as vixotrigine, PF-05089771, A803467, PF-01247324, VX-150 or arachnid toxins such as Tap1a; the use of Ca2+ channel blockers such as TTA-P2, TTA-A2, Z 944, ACT709478, and CNCB-2; (ii) improving methods for assessing "pain" as opposed to nociception in rodent models; (iii) recognizing sex differences in pain etiology; (iv) tailoring of therapeutic approaches to meet the symptoms and etiology of pain in individual patients via quantitative sensory testing and other personalized medicine approaches; (v) targeting genetic and biochemical mechanisms controlling channel expression using anti-NGF antibodies such as tanezumab or re-purposed drugs such as vorinostat, a histone methyltransferase inhibitor used in the management of T-cell lymphoma, or cercosporamide a MNK 1/2 inhibitor used in treatment of rheumatoid arthritis; (vi) combination therapy using drugs that are selective for different channel types or regulatory processes; (vii) directing preclinical validation work toward the use of human or human-derived tissue samples; and (viii) application of molecular biological approaches such as clustered regularly interspaced short palindromic repeats (CRISPR) technology.
Collapse
Affiliation(s)
- Sascha R A Alles
- Department of Anesthesiology and Critical Care Medicine, University of New Mexico School of Medicine, Albuquerque, NM, United States
| | - Peter A Smith
- Department of Pharmacology, Neuroscience and Mental Health Institute, University of Alberta, Edmonton, AB, Canada
| |
Collapse
|
21
|
Macedo KWR, Costa LJDL, de Souza JO, de Vasconcelos IA, de Castro JS, de Santana CJC, Magalhães ACM, Castro MDS, Pires OR. Brazilian Theraphosidae: a toxicological point of view. J Venom Anim Toxins Incl Trop Dis 2021; 27:e20210004. [PMID: 34868282 PMCID: PMC8610171 DOI: 10.1590/1678-9199-jvatitd-2021-0004] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2021] [Accepted: 04/08/2021] [Indexed: 11/30/2022] Open
Abstract
The Theraphosidae family includes the largest number of species of the
Mygalomorphae infraorder, with hundreds of species currently catalogued.
However, there is a huge lack on physiologic and even ecologic information
available, especially in Brazil, which is the most biodiverse country in the
world. Over the years, spiders have been presented as a source of multiple
biologically active compounds with basic roles, such as primary defense against
pathogenic microorganisms or modulation of metabolic pathways and as specialized
hunters. Spider venoms also evolved in order to enable the capture of prey by
interaction with a diversity of molecular targets of interest, raising their
pharmaceutical potential for the development of new drugs. Among the activities
found in compounds isolated from venoms and hemocytes of Brazilian Theraphosidae
there are antimicrobial, antifungal, antiparasitic and antitumoral, as well as
properties related to proteinase action and neuromuscular blockage modulated by
ionic voltage-gated channel interaction. These characteristics are present in
different species from multiple genera, which is strong evidence of the
important role in spider survival. The present review aims to compile the main
results of studies from the last decades on Brazilian Theraphosidae with special
focus on results obtained with the crude venom or compounds isolated from both
venom and hemocytes, and their physiological and chemical characterization.
Collapse
Affiliation(s)
- Keven Wender Rodrigues Macedo
- Laboratory of Toxinology, Department of Physiological Sciences, Institute of Biology, University of Brasília (UnB), Brasília, DF, Brazil
| | - Lucas Jeferson de Lima Costa
- Laboratory of Toxinology, Department of Physiological Sciences, Institute of Biology, University of Brasília (UnB), Brasília, DF, Brazil
| | - Jéssica Oliveira de Souza
- Laboratory of Toxinology, Department of Physiological Sciences, Institute of Biology, University of Brasília (UnB), Brasília, DF, Brazil
| | - Isadora Alves de Vasconcelos
- Laboratory of Toxinology, Department of Physiological Sciences, Institute of Biology, University of Brasília (UnB), Brasília, DF, Brazil
| | - Jessica Schneider de Castro
- Laboratory of Toxinology, Department of Physiological Sciences, Institute of Biology, University of Brasília (UnB), Brasília, DF, Brazil
| | - Carlos José Correia de Santana
- Laboratory of Toxinology, Department of Physiological Sciences, Institute of Biology, University of Brasília (UnB), Brasília, DF, Brazil.,Laboratory of Biochemistry and Protein Chemistry, Department of Cell Biology, Institute of Biology, University of Brasília (UnB), Brasília, DF, Brazil
| | - Ana Carolina Martins Magalhães
- Laboratory of Toxinology, Department of Physiological Sciences, Institute of Biology, University of Brasília (UnB), Brasília, DF, Brazil
| | - Mariana de Souza Castro
- Laboratory of Toxinology, Department of Physiological Sciences, Institute of Biology, University of Brasília (UnB), Brasília, DF, Brazil.,Laboratory of Biochemistry and Protein Chemistry, Department of Cell Biology, Institute of Biology, University of Brasília (UnB), Brasília, DF, Brazil
| | - Osmindo Rodrigues Pires
- Laboratory of Toxinology, Department of Physiological Sciences, Institute of Biology, University of Brasília (UnB), Brasília, DF, Brazil
| |
Collapse
|
22
|
Hu H, Mawlawi SE, Zhao T, Deuis JR, Jami S, Vetter I, Lewis RJ, Cardoso FC. Engineering of a Spider Peptide via Conserved Structure-Function Traits Optimizes Sodium Channel Inhibition In Vitro and Anti-Nociception In Vivo. Front Mol Biosci 2021; 8:742457. [PMID: 34621788 PMCID: PMC8490825 DOI: 10.3389/fmolb.2021.742457] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2021] [Accepted: 09/06/2021] [Indexed: 12/12/2022] Open
Abstract
Venom peptides are potent and selective modulators of voltage-gated ion channels that regulate neuronal function both in health and in disease. We previously identified the spider venom peptide Tap1a from the Venezuelan tarantula Theraphosa apophysis that targeted multiple voltage-gated sodium and calcium channels in visceral pain pathways and inhibited visceral mechano-sensing neurons contributing to irritable bowel syndrome. In this work, alanine scanning and domain activity analysis revealed Tap1a inhibited sodium channels by binding with nanomolar affinity to the voltage-sensor domain II utilising conserved structure-function features characteristic of spider peptides belonging to family NaSpTx1. In order to speed up the development of optimized NaV-targeting peptides with greater inhibitory potency and enhanced in vivo activity, we tested the hypothesis that incorporating residues identified from other optimized NaSpTx1 peptides into Tap1a could also optimize its potency for NaVs. Applying this approach, we designed the peptides Tap1a-OPT1 and Tap1a-OPT2 exhibiting significant increased potency for NaV1.1, NaV1.2, NaV1.3, NaV1.6 and NaV1.7 involved in several neurological disorders including acute and chronic pain, motor neuron disease and epilepsy. Tap1a-OPT1 showed increased potency for the off-target NaV1.4, while this off-target activity was absent in Tap1a-OPT2. This enhanced potency arose through a slowed off-rate mechanism. Optimized inhibition of NaV channels observed in vitro translated in vivo, with reversal of nocifensive behaviours in a murine model of NaV-mediated pain also enhanced by Tap1a-OPT. Molecular docking studies suggested that improved interactions within loops 3 and 4, and C-terminal of Tap1a-OPT and the NaV channel voltage-sensor domain II were the main drivers of potency optimization. Overall, the rationally designed peptide Tap1a-OPT displayed new and refined structure-function features which are likely the major contributors to its enhanced bioactive properties observed in vivo. This work contributes to the rapid engineering and optimization of potent spider peptides multi-targeting NaV channels, and the research into novel drugs to treat neurological diseases.
Collapse
Affiliation(s)
- H Hu
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD, Australia
| | - S E Mawlawi
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD, Australia
| | - T Zhao
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD, Australia
| | - J R Deuis
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD, Australia
| | - S Jami
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD, Australia
| | - I Vetter
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD, Australia.,School of Pharmacy, The University of Queensland, Brisbane, QLD, Australia
| | - R J Lewis
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD, Australia
| | - F C Cardoso
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD, Australia.,Centre for Innovations in Peptide and Protein Science, The University of Queensland, Brisbane, QLD, Australia
| |
Collapse
|
23
|
V V, Achar RR, M.U H, N A, T YS, Kameshwar VH, Byrappa K, Ramadas D. Venom peptides - A comprehensive translational perspective in pain management. Curr Res Toxicol 2021; 2:329-340. [PMID: 34604795 PMCID: PMC8473576 DOI: 10.1016/j.crtox.2021.09.001] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2021] [Revised: 09/02/2021] [Accepted: 09/08/2021] [Indexed: 12/21/2022] Open
Abstract
Venom peptides have been evolving complex therapeutic interventions that potently and selectively modulate a range of targets such as ion channels, receptors, and signaling pathways of physiological processes making it potential therapeutic. Several venom peptides were deduced in vivo for clinical development targeting pain management, diabetes, cardiovascular diseases, antimicrobial activity. Several contributions have been detailed for a clear perspective for a better understanding of venomous animals, their venom, and their pharmacological effects. Here we unravel and summarize the recent advances in wide venom peptides across varieties of species for their therapeutics prospects.
Collapse
Affiliation(s)
- Vidya V
- K. S Hegde Medical Academy, NITTE (Deemed to be) University, Mangalore 575015, Karnataka, India
| | - Raghu Ram Achar
- Division of Biochemistry, School of Life Sciences, JSS Academy of Higher Education & Research, S.S. Nagar, Mysuru 570 015, Karnataka, India
| | - Himathi M.U
- Division of Biochemistry, School of Life Sciences, JSS Academy of Higher Education & Research, S.S. Nagar, Mysuru 570 015, Karnataka, India
| | - Akshita N
- Division of Biochemistry, School of Life Sciences, JSS Academy of Higher Education & Research, S.S. Nagar, Mysuru 570 015, Karnataka, India
| | - Yogish Somayaji T
- Department of Post Graduate Studies and Research in Biochemistry, St. Aloysius College (Autonomous), Mangalore 575003, Karnataka, India
| | - Vivek Hamse Kameshwar
- School of Natural Science, Adichunchanagiri University, B.G. Nagara-571448, Nangamangala, Mandya, India
- School of Natural Sciences, ACU-CRI, Adichunchanagiri University, BGSIT Campus, B.G. Nagara-571448, Nagamangala, Mandya, India
| | - K. Byrappa
- School of Natural Sciences, ACU-CRI, Adichunchanagiri University, BGSIT Campus, B.G. Nagara-571448, Nagamangala, Mandya, India
- Center for Material Science and Technology, Vijnana Bhavan, University of Mysore, Mysuru, Karnataka, India
| | - Dinesha Ramadas
- Adichunchanagiri Institute for Molecular Medicine, AIMS, Adichunchanagiri University, B.G. Nagara-571448, Nagamangala, Mandya, India
| |
Collapse
|
24
|
Dobson JS, Harris RJ, Zdenek CN, Huynh T, Hodgson WC, Bosmans F, Fourmy R, Violette A, Fry BG. The Dragon's Paralysing Spell: Evidence of Sodium and Calcium Ion Channel Binding Neurotoxins in Helodermatid and Varanid Lizard Venoms. Toxins (Basel) 2021; 13:toxins13080549. [PMID: 34437420 PMCID: PMC8402328 DOI: 10.3390/toxins13080549] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2021] [Revised: 07/26/2021] [Accepted: 07/27/2021] [Indexed: 12/19/2022] Open
Abstract
Bites from helodermatid lizards can cause pain, paresthesia, paralysis, and tachycardia, as well as other symptoms consistent with neurotoxicity. Furthermore, in vitro studies have shown that Heloderma horridum venom inhibits ion flux and blocks the electrical stimulation of skeletal muscles. Helodermatids have long been considered the only venomous lizards, but a large body of robust evidence has demonstrated venom to be a basal trait of Anguimorpha. This clade includes varanid lizards, whose bites have been reported to cause anticoagulation, pain, and occasionally paralysis and tachycardia. Despite the evolutionary novelty of these lizard venoms, their neuromuscular targets have yet to be identified, even for the iconic helodermatid lizards. Therefore, to fill this knowledge gap, the venoms of three Heloderma species (H. exasperatum, H. horridum and H. suspectum) and two Varanus species (V. salvadorii and V. varius) were investigated using Gallus gallus chick biventer cervicis nerve–muscle preparations and biolayer interferometry assays for binding to mammalian ion channels. Incubation with Heloderma venoms caused the reduction in nerve-mediated muscle twitches post initial response of avian skeletal muscle tissue preparation assays suggesting voltage-gated sodium (NaV) channel binding. Congruent with the flaccid paralysis inducing blockage of electrical stimulation in the skeletal muscle preparations, the biolayer interferometry tests with Heloderma suspectum venom revealed binding to the S3–S4 loop within voltage-sensing domain IV of the skeletal muscle channel subtype, NaV1.4. Consistent with tachycardia reported in clinical cases, the venom also bound to voltage-sensing domain IV of the cardiac smooth muscle calcium channel, CaV1.2. While Varanus varius venom did not have discernable effects in the avian tissue preparation assay at the concentration tested, in the biointerferometry assay both V. varius and V. salvadorii bound to voltage-sensing domain IV of both NaV1.4 and CaV1.2, similar to H. suspectum venom. The ability of varanid venoms to bind to mammalian ion channels but not to the avian tissue preparation suggests prey-selective actions, as did the differential potency within the Heloderma venoms for avian versus mammalian pathophysiological targets. This study thus presents the detailed characterization of Heloderma venom ion channel neurotoxicity and offers the first evidence of varanid lizard venom neurotoxicity. In addition, the data not only provide information useful to understanding the clinical effects produced by envenomations, but also reveal their utility as physiological probes, and underscore the potential utility of neglected venomous lineages in the drug design and development pipeline.
Collapse
Affiliation(s)
- James S. Dobson
- Venom Evolution Lab, School of Biological Sciences, University of Queensland, St. Lucia, QLD 4072, Australia; (J.S.D.); (R.J.H.); (C.N.Z.)
| | - Richard J. Harris
- Venom Evolution Lab, School of Biological Sciences, University of Queensland, St. Lucia, QLD 4072, Australia; (J.S.D.); (R.J.H.); (C.N.Z.)
| | - Christina N. Zdenek
- Venom Evolution Lab, School of Biological Sciences, University of Queensland, St. Lucia, QLD 4072, Australia; (J.S.D.); (R.J.H.); (C.N.Z.)
| | - Tam Huynh
- Department of Pharmacology, Biomedicine Discovery Institute, Monash University, Clayton, VIC 3800, Australia; (T.H.); (W.C.H.)
| | - Wayne C. Hodgson
- Department of Pharmacology, Biomedicine Discovery Institute, Monash University, Clayton, VIC 3800, Australia; (T.H.); (W.C.H.)
| | - Frank Bosmans
- Department of Basic and Applied Medical Sciences, Ghent University, 9000 Ghent, Belgium;
| | - Rudy Fourmy
- Alphabiotoxine Laboratory sprl, Barberie 15, 7911 Montroeul-au-Bois, Belgium; (R.F.); (A.V.)
| | - Aude Violette
- Alphabiotoxine Laboratory sprl, Barberie 15, 7911 Montroeul-au-Bois, Belgium; (R.F.); (A.V.)
| | - Bryan G. Fry
- Venom Evolution Lab, School of Biological Sciences, University of Queensland, St. Lucia, QLD 4072, Australia; (J.S.D.); (R.J.H.); (C.N.Z.)
- Correspondence: ; Tel.: +61-7-336-58515
| |
Collapse
|
25
|
Muratspahić E, Tomašević N, Koehbach J, Duerrauer L, Hadžić S, Castro J, Schober G, Sideromenos S, Clark RJ, Brierley SM, Craik DJ, Gruber CW. Design of a Stable Cyclic Peptide Analgesic Derived from Sunflower Seeds that Targets the κ-Opioid Receptor for the Treatment of Chronic Abdominal Pain. J Med Chem 2021; 64:9042-9055. [PMID: 34162205 PMCID: PMC8273886 DOI: 10.1021/acs.jmedchem.1c00158] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2021] [Indexed: 02/01/2023]
Abstract
The rising opioid crisis has become a worldwide societal and public health burden, resulting from the abuse of prescription opioids. Targeting the κ-opioid receptor (KOR) in the periphery has emerged as a powerful approach to develop novel pain medications without central side effects. Inspired by the traditional use of sunflower (Helianthus annuus) preparations for analgesic purposes, we developed novel stabilized KOR ligands (termed as helianorphins) by incorporating different dynorphin A sequence fragments into a cyclic sunflower peptide scaffold. As a result, helianorphin-19 selectively bound to and fully activated the KOR with nanomolar potency. Importantly, helianorphin-19 exhibited strong KOR-specific peripheral analgesic activity in a mouse model of chronic visceral pain, without inducing unwanted central effects on motor coordination/sedation. Our study provides a proof of principle that cyclic peptides from plants may be used as templates to develop potent and stable peptide analgesics applicable via enteric administration by targeting the peripheral KOR for the treatment of chronic abdominal pain.
Collapse
MESH Headings
- Abdominal Pain/drug therapy
- Analgesics/chemical synthesis
- Analgesics/chemistry
- Analgesics/pharmacology
- Animals
- Cells, Cultured
- Chronic Disease
- Dose-Response Relationship, Drug
- Drug Design
- HEK293 Cells
- Helianthus/chemistry
- Humans
- Male
- Mice
- Mice, Inbred C57BL
- Molecular Structure
- Peptides, Cyclic/chemical synthesis
- Peptides, Cyclic/chemistry
- Peptides, Cyclic/pharmacology
- Plant Extracts/chemical synthesis
- Plant Extracts/chemistry
- Plant Extracts/pharmacology
- Receptors, Opioid, kappa/antagonists & inhibitors
- Receptors, Opioid, kappa/metabolism
- Seeds/chemistry
- Structure-Activity Relationship
Collapse
Affiliation(s)
- Edin Muratspahić
- Center
for Physiology and Pharmacology, Institute of Pharmacology, Medical University of Vienna, 1090 Vienna, Austria
| | - Nataša Tomašević
- Center
for Physiology and Pharmacology, Institute of Pharmacology, Medical University of Vienna, 1090 Vienna, Austria
| | - Johannes Koehbach
- Institute
for Molecular Bioscience, Australian Research Council Centre of Excellence
for Innovations in Peptide and Protein Science, The University of Queensland, Brisbane, Queensland 4072, Australia
| | - Leopold Duerrauer
- Center
for Physiology and Pharmacology, Institute of Pharmacology, Medical University of Vienna, 1090 Vienna, Austria
- School
of Biomedical Sciences, Faculty of Medicine, The University of Queensland, Brisbane, Queensland 4072, Australia
| | - Seid Hadžić
- Center
for Physiology and Pharmacology, Institute of Pharmacology, Medical University of Vienna, 1090 Vienna, Austria
| | - Joel Castro
- Visceral
Pain Research Group, College of Medicine and Public Health, Flinders
Health and Medical Research Institute (FHMRI), Flinders University, Bedford
Park, South Australia 5042, Australia
- Hopwood
Centre for Neurobiology, Lifelong Health Theme, South Australian Health and Medical Research Institute (SAHMRI), North Terrace, Adelaide, South Australia 5000, Australia
| | - Gudrun Schober
- Visceral
Pain Research Group, College of Medicine and Public Health, Flinders
Health and Medical Research Institute (FHMRI), Flinders University, Bedford
Park, South Australia 5042, Australia
- Hopwood
Centre for Neurobiology, Lifelong Health Theme, South Australian Health and Medical Research Institute (SAHMRI), North Terrace, Adelaide, South Australia 5000, Australia
| | - Spyridon Sideromenos
- Center for
Physiology and Pharmacology, Department of Neurophysiology and Neuropharmacology, Medical University of Vienna, 1090 Vienna, Austria
| | - Richard J. Clark
- School
of Biomedical Sciences, Faculty of Medicine, The University of Queensland, Brisbane, Queensland 4072, Australia
| | - Stuart M. Brierley
- Visceral
Pain Research Group, College of Medicine and Public Health, Flinders
Health and Medical Research Institute (FHMRI), Flinders University, Bedford
Park, South Australia 5042, Australia
- Hopwood
Centre for Neurobiology, Lifelong Health Theme, South Australian Health and Medical Research Institute (SAHMRI), North Terrace, Adelaide, South Australia 5000, Australia
- Discipline
of Medicine, University of Adelaide, North Terrace, Adelaide, South Australia 5000, 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, Queensland 4072, Australia
| | - Christian W. Gruber
- Center
for Physiology and Pharmacology, Institute of Pharmacology, Medical University of Vienna, 1090 Vienna, Austria
| |
Collapse
|
26
|
Yang M, Li Y, Liu L, Zhou M. A novel proline-rich M-superfamily conotoxin that can simultaneously affect sodium, potassium and calcium currents. J Venom Anim Toxins Incl Trop Dis 2021; 27:e20200164. [PMID: 34234819 PMCID: PMC8230863 DOI: 10.1590/1678-9199-jvatitd-2020-0164] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2020] [Accepted: 03/01/2021] [Indexed: 12/19/2022] Open
Abstract
Background Conotoxins have become a research hotspot in the neuropharmacology field for their high activity and specificity in targeting ion channels and neurotransmitter receptors. There have been reports of a conotoxin acting on two ion channels, but rare reports of a conotoxin acting on three ion channels. Methods Vr3a, a proline-rich M-superfamily conotoxin from a worm-hunting Conus varius, was obtained by solid-phase synthesis and identified by mass spectrometry. The effects of synthesized Vr3a on sodium, potassium and calcium currents were tested on rat DRG cells by patch clamp experiments. The further effects of Vr3a on human Cav1.2 and Cav2.2 currents were tested on HEK293 cells. Results About 10 μM Vr3a has no effects on the peak sodium currents, but can induce a ~10 mV shift in a polarizing direction in the current-voltage relationship. In addition, 10 μM Vr3a can increase 19.61 ± 5.12% of the peak potassium currents and do not induce a shift in the current-voltage relationship. An amount of 10 μM Vr3a can inhibit 31.26% ± 4.53% of the peak calcium currents and do not induce a shift in the current-voltage relationship. The IC50 value of Vr3a on calcium channel currents in rat DRG neurons is 19.28 ± 4.32 μM. Moreover, 10 μM Vr3a can inhibit 15.32% ± 5.41% of the human Cav1.2 currents and 12.86% ± 4.93% of the human Cav2.2 currents. Conclusions Vr3a can simultaneously affect sodium, potassium and calcium currents. This novel triple-target conotoxin Vr3a expands understanding of conotoxin functions.
Collapse
Affiliation(s)
- Manyi Yang
- Department of Hepatobiliary and Pancreatic Surgery, NHC Key Laboratory of Nanobiological Technology, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Yubin Li
- Department of Oncology, State Local Joint Engineering Laboratory for Anticancer Drugs, NHC Key Laboratory of Cancer Proteomics, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Longfei Liu
- Department of Urology, National Clinical Research Center for Geriatric Disorder, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Maojun Zhou
- Department of Oncology, State Local Joint Engineering Laboratory for Anticancer Drugs, NHC Key Laboratory of Cancer Proteomics, Xiangya Hospital, Central South University, Changsha, Hunan, China
| |
Collapse
|