1
|
Riaño-Umbarila L, Olamendi-Portugal T, Romero-Moreno JA, Delgado-Prudencio G, Zamudio FZ, Becerril B, Possani LD. Toxic Peptides from the Mexican Scorpion Centruroides villegasi: Chemical Structure and Evaluation of Recognition by Human Single-Chain Antibodies. Toxins (Basel) 2024; 16:301. [PMID: 39057941 PMCID: PMC11280942 DOI: 10.3390/toxins16070301] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Revised: 06/26/2024] [Accepted: 06/28/2024] [Indexed: 07/28/2024] Open
Abstract
Alternative recombinant sources of antivenoms have been successfully generated. The application of such strategies requires the characterization of the venoms for the development of specific neutralizing molecules against the toxic components. Five toxic peptides to mammals from the Mexican scorpion Centruroides villegasi were isolated by chromatographic procedures by means of gel filtration on Sephadex G-50, followed by ion-exchange columns on carboxy-methyl-cellulose (CMC) resins and finally purified by high-performance chromatography (HPLC) columns. Their primary structures were determined by Edman degradation. They contain 66 amino acids and are maintained well packed by four disulfide bridges, with molecular mass from 7511.3 to 7750.1 Da. They are all relatively toxic and deadly to mice and show high sequence identity with known peptides that are specific modifiers of the gating mechanisms of Na+ ion channels of type beta-toxin (β-ScTx). They were named Cv1 to Cv5 and used to test their recognition by single-chain variable fragments (scFv) of antibodies, using surface plasmon resonance. Three different scFvs generated in our laboratory (10FG2, HV, LR) were tested for recognizing the various new peptides described here, paving the way for the development of a novel type of scorpion antivenom.
Collapse
Affiliation(s)
- Lidia Riaño-Umbarila
- Investigadora por México, CONAHCYT, Mexico City 03940, Mexico;
- Departamento de Medicina Molecular y Bioprocesos, Instituto de Biotecnología, Universidad Nacional Autónoma de México, Cuernavaca 62210, Mexico; (T.O.-P.); (J.A.R.-M.); (G.D.-P.); (F.Z.Z.)
| | - Timoteo Olamendi-Portugal
- Departamento de Medicina Molecular y Bioprocesos, Instituto de Biotecnología, Universidad Nacional Autónoma de México, Cuernavaca 62210, Mexico; (T.O.-P.); (J.A.R.-M.); (G.D.-P.); (F.Z.Z.)
| | - José Alberto Romero-Moreno
- Departamento de Medicina Molecular y Bioprocesos, Instituto de Biotecnología, Universidad Nacional Autónoma de México, Cuernavaca 62210, Mexico; (T.O.-P.); (J.A.R.-M.); (G.D.-P.); (F.Z.Z.)
| | - Gustavo Delgado-Prudencio
- Departamento de Medicina Molecular y Bioprocesos, Instituto de Biotecnología, Universidad Nacional Autónoma de México, Cuernavaca 62210, Mexico; (T.O.-P.); (J.A.R.-M.); (G.D.-P.); (F.Z.Z.)
| | - Fernando Z. Zamudio
- Departamento de Medicina Molecular y Bioprocesos, Instituto de Biotecnología, Universidad Nacional Autónoma de México, Cuernavaca 62210, Mexico; (T.O.-P.); (J.A.R.-M.); (G.D.-P.); (F.Z.Z.)
| | - Baltazar Becerril
- Departamento de Medicina Molecular y Bioprocesos, Instituto de Biotecnología, Universidad Nacional Autónoma de México, Cuernavaca 62210, Mexico; (T.O.-P.); (J.A.R.-M.); (G.D.-P.); (F.Z.Z.)
| | - Lourival D. Possani
- Departamento de Medicina Molecular y Bioprocesos, Instituto de Biotecnología, Universidad Nacional Autónoma de México, Cuernavaca 62210, Mexico; (T.O.-P.); (J.A.R.-M.); (G.D.-P.); (F.Z.Z.)
| |
Collapse
|
2
|
Chen M, Lu M, Feng X, Wu M, Luo X, Xiang R, Luo R, Wu H, Liu Z, Wang M, Zhou X. LmNaTx15, a novel scorpion toxin, enhances the activity of Nav channels and induces pain in mice. Toxicon 2023; 236:107331. [PMID: 37918718 DOI: 10.1016/j.toxicon.2023.107331] [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: 09/16/2023] [Revised: 10/30/2023] [Accepted: 10/31/2023] [Indexed: 11/04/2023]
Abstract
Polypeptide toxins are major bioactive components found in venomous animals. Many polypeptide toxins can specifically act on targets, such as ion channels and voltage-gated sodium (Nav) channels, in the nervous, muscle, and cardiovascular systems of the recipient to increase defense and predation efficiency. In this study, a novel polypeptide toxin, LmNaTx15, was isolated from the venom of the scorpion Lychas mucronatus, and its activity was analyzed. LmNaTx15 slowed the fast inactivation of Nav1.2, Nav1.3, Nav1.4, Nav1.5, and Nav1.7 and inhibited the peak current of Nav1.5, but it did not affect Nav1.8. In addition, LmNaTx15 altered the voltage-dependent activation and inactivation of these Nav channel subtypes. Furthermore, like site 3 neurotoxins, LmNaTx15 induced pain in mice. These results show a novel scorpion toxin with a modulatory effect on specific Nav channel subtypes and pain induction in mice. Therefore, LmNaTx15 may be a key bioactive component for scorpion defense and predation. Besides, this study provides a basis for analyzing structure-function relationships of the scorpion toxins affecting Nav channel activity.
Collapse
Affiliation(s)
- Minzhi Chen
- The National & Local Joint Engineering Laboratory of Animal Peptide Drug Development, College of Life Sciences, Hunan Normal University, Changsha, Hunan, 410081, China; Institute of Interdisciplinary Studies, Hunan Normal University, Changsha, 410081, China; Peptide and Small Molecule Drug R&D Plateform, Furong Laboratory, Hunan Normal University, Changsha, 410081, Hunan, China
| | - Minjuan Lu
- The National & Local Joint Engineering Laboratory of Animal Peptide Drug Development, College of Life Sciences, Hunan Normal University, Changsha, Hunan, 410081, China; Institute of Interdisciplinary Studies, Hunan Normal University, Changsha, 410081, China; Peptide and Small Molecule Drug R&D Plateform, Furong Laboratory, Hunan Normal University, Changsha, 410081, Hunan, China
| | - Xujun Feng
- The National & Local Joint Engineering Laboratory of Animal Peptide Drug Development, College of Life Sciences, Hunan Normal University, Changsha, Hunan, 410081, China; Institute of Interdisciplinary Studies, Hunan Normal University, Changsha, 410081, China; Peptide and Small Molecule Drug R&D Plateform, Furong Laboratory, Hunan Normal University, Changsha, 410081, Hunan, China
| | - Meijing Wu
- The National & Local Joint Engineering Laboratory of Animal Peptide Drug Development, College of Life Sciences, Hunan Normal University, Changsha, Hunan, 410081, China; Institute of Interdisciplinary Studies, Hunan Normal University, Changsha, 410081, China; Peptide and Small Molecule Drug R&D Plateform, Furong Laboratory, Hunan Normal University, Changsha, 410081, Hunan, China
| | - Xiaoqing Luo
- The National & Local Joint Engineering Laboratory of Animal Peptide Drug Development, College of Life Sciences, Hunan Normal University, Changsha, Hunan, 410081, China; Institute of Interdisciplinary Studies, Hunan Normal University, Changsha, 410081, China; Peptide and Small Molecule Drug R&D Plateform, Furong Laboratory, Hunan Normal University, Changsha, 410081, Hunan, China
| | - Ruiqi Xiang
- The National & Local Joint Engineering Laboratory of Animal Peptide Drug Development, College of Life Sciences, Hunan Normal University, Changsha, Hunan, 410081, China; Institute of Interdisciplinary Studies, Hunan Normal University, Changsha, 410081, China; Peptide and Small Molecule Drug R&D Plateform, Furong Laboratory, Hunan Normal University, Changsha, 410081, Hunan, China
| | - Ren Luo
- The National & Local Joint Engineering Laboratory of Animal Peptide Drug Development, College of Life Sciences, Hunan Normal University, Changsha, Hunan, 410081, China; Institute of Interdisciplinary Studies, Hunan Normal University, Changsha, 410081, China; Peptide and Small Molecule Drug R&D Plateform, Furong Laboratory, Hunan Normal University, Changsha, 410081, Hunan, China
| | - Hang Wu
- The National & Local Joint Engineering Laboratory of Animal Peptide Drug Development, College of Life Sciences, Hunan Normal University, Changsha, Hunan, 410081, China; Institute of Interdisciplinary Studies, Hunan Normal University, Changsha, 410081, China; Peptide and Small Molecule Drug R&D Plateform, Furong Laboratory, Hunan Normal University, Changsha, 410081, Hunan, China
| | - Zhonghua Liu
- The National & Local Joint Engineering Laboratory of Animal Peptide Drug Development, College of Life Sciences, Hunan Normal University, Changsha, Hunan, 410081, China; Institute of Interdisciplinary Studies, Hunan Normal University, Changsha, 410081, China; Peptide and Small Molecule Drug R&D Plateform, Furong Laboratory, Hunan Normal University, Changsha, 410081, Hunan, China
| | - Meichi Wang
- The National & Local Joint Engineering Laboratory of Animal Peptide Drug Development, College of Life Sciences, Hunan Normal University, Changsha, Hunan, 410081, China; Institute of Interdisciplinary Studies, Hunan Normal University, Changsha, 410081, China; Peptide and Small Molecule Drug R&D Plateform, Furong Laboratory, Hunan Normal University, Changsha, 410081, Hunan, China.
| | - Xi Zhou
- The National & Local Joint Engineering Laboratory of Animal Peptide Drug Development, College of Life Sciences, Hunan Normal University, Changsha, Hunan, 410081, China; Institute of Interdisciplinary Studies, Hunan Normal University, Changsha, 410081, China; Peptide and Small Molecule Drug R&D Plateform, Furong Laboratory, Hunan Normal University, Changsha, 410081, Hunan, China.
| |
Collapse
|
3
|
Mendes LC, Viana GMM, Nencioni ALA, Pimenta DC, Beraldo-Neto E. Scorpion Peptides and Ion Channels: An Insightful Review of Mechanisms and Drug Development. Toxins (Basel) 2023; 15:238. [PMID: 37104176 PMCID: PMC10145618 DOI: 10.3390/toxins15040238] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2023] [Revised: 03/21/2023] [Accepted: 03/22/2023] [Indexed: 04/28/2023] Open
Abstract
The Buthidae family of scorpions consists of arthropods with significant medical relevance, as their venom contains a diverse range of biomolecules, including neurotoxins that selectively target ion channels in cell membranes. These ion channels play a crucial role in regulating physiological processes, and any disturbance in their activity can result in channelopathies, which can lead to various diseases such as autoimmune, cardiovascular, immunological, neurological, and neoplastic conditions. Given the importance of ion channels, scorpion peptides represent a valuable resource for developing drugs with targeted specificity for these channels. This review provides a comprehensive overview of the structure and classification of ion channels, the action of scorpion toxins on these channels, and potential avenues for future research. Overall, this review highlights the significance of scorpion venom as a promising source for discovering novel drugs with therapeutic potential for treating channelopathies.
Collapse
Affiliation(s)
- Lais Campelo Mendes
- Programa de Pós-Graduação em Ciências—Toxinologia do Instituto Butantan, São Paulo 05503-900, Brazil
- Laboratório de Bioquímica do Instituto Butantan, São Paulo 05503-900, Brazil
| | | | | | | | - Emidio Beraldo-Neto
- Laboratório de Bioquímica do Instituto Butantan, São Paulo 05503-900, Brazil
| |
Collapse
|
4
|
Kalapothakis Y, Miranda K, Molina DAM, Conceição IMCA, Larangote D, Op den Camp HJM, Kalapothakis E, Chávez-Olórtegui C, Borges A. An overview of Tityus cisandinus scorpion venom: Transcriptome and mass fingerprinting reveal conserved toxin homologs across the Amazon region and novel lipolytic components. Int J Biol Macromol 2023; 225:1246-1266. [PMID: 36427608 DOI: 10.1016/j.ijbiomac.2022.11.185] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2022] [Revised: 11/09/2022] [Accepted: 11/18/2022] [Indexed: 11/23/2022]
Abstract
Tityus cisandinus, a neglected medically important scorpion in Ecuadorian and Peruvian Amazonia, belongs to a complex of species related to the eastern Amazon endemic Tityus obscurus, spanning a distribution of ca. 4000 km. Despite high morbidity and mortality rates, no effective scorpion antivenom is currently available in the Amazon region. Knowledge of the structural/functional relationships between T. cisandinus venom components and those from related Amazonian species is crucial for designing region-specific therapeutic antivenoms. In this work, we carried out the first venom gland transcriptomic study of an Amazonian scorpion outside Brazil, T. cisandinus. We also fingerprinted its total venom through MALDI-TOF MS, which supported our transcriptomic findings. We identified and calculated the expression level of 94 components: 60 toxins, 25 metalloproteases, five disulfide isomerases, three amidating enzymes, one hyaluronidase, and also uncovered transcripts encoding novel lipolytic beta subunits produced by New World buthid scorpions. This study demonstrates the high similarity between T. cisandinus and T. obscurus venoms, reinforcing the existence of a neglected complex of genetically and toxinologically related Amazonian scorpions of medical importance. Finally, we demonstrated the low recognition of currently available therapeutic sera against T. cisandinus and T. obscurus venoms, and concluded that these should be improved to protect against envenomation by Amazonian Tityus spp.
Collapse
Affiliation(s)
- Yan Kalapothakis
- Departamento de Genética, Ecologia e Evolução, ICB, Universidade Federal de Minas Gerais, Av. Antônio Carlos 6627, CEP: 31270-901 Belo Horizonte, Minas Gerais, Brazil
| | - Kelton Miranda
- Departamento de Genética, Ecologia e Evolução, ICB, Universidade Federal de Minas Gerais, Av. Antônio Carlos 6627, CEP: 31270-901 Belo Horizonte, Minas Gerais, Brazil
| | - Denis Alexis Molina Molina
- Departamento de Bioquímica e Imunologia, ICB, Universidade Federal de Minas Gerais, Av. Antônio Carlos 6627, CEP: 31270-901 Belo Horizonte, Minas Gerais, Brazil
| | - Izabela Mamede Costa Andrade Conceição
- Departamento de Bioquímica e Imunologia, ICB, Universidade Federal de Minas Gerais, Av. Antônio Carlos 6627, CEP: 31270-901 Belo Horizonte, Minas Gerais, Brazil
| | - Débora Larangote
- Departamento de Genética, Ecologia e Evolução, ICB, Universidade Federal de Minas Gerais, Av. Antônio Carlos 6627, CEP: 31270-901 Belo Horizonte, Minas Gerais, Brazil
| | - Huub J M Op den Camp
- Department of Microbiology, Faculty of Science, Radboud University Nijmegen, Heyendaalseweg 135, NL-6525 AJ Nijmegen, the Netherlands
| | - Evanguedes Kalapothakis
- Departamento de Genética, Ecologia e Evolução, ICB, Universidade Federal de Minas Gerais, Av. Antônio Carlos 6627, CEP: 31270-901 Belo Horizonte, Minas Gerais, Brazil
| | - Carlos Chávez-Olórtegui
- Departamento de Bioquímica e Imunologia, ICB, Universidade Federal de Minas Gerais, Av. Antônio Carlos 6627, CEP: 31270-901 Belo Horizonte, Minas Gerais, Brazil
| | - Adolfo Borges
- Instituto de Medicina Experimental, Universidad Central de Venezuela, Caracas, Venezuela; Centro para el Desarrollo de la Investigación Científica, CEDIC, Asunción 1255, Paraguay.
| |
Collapse
|
5
|
Zhao F, Fang L, Wang Q, Ye Q, He Y, Xu W, Song Y. Exploring the Pivotal Components Influencing the Side Effects Induced by an Analgesic-Antitumor Peptide from Scorpion Venom on Human Voltage-Gated Sodium Channels 1.4 and 1.5 through Computational Simulation. Toxins (Basel) 2022; 15:33. [PMID: 36668853 PMCID: PMC9864070 DOI: 10.3390/toxins15010033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Revised: 12/28/2022] [Accepted: 12/29/2022] [Indexed: 01/04/2023] Open
Abstract
Voltage-gated sodium channels (VGSCs, or Nav) are important determinants of action potential generation and propagation. Efforts are underway to develop medicines targeting different channel subtypes for the treatment of related channelopathies. However, a high degree of conservation across its nine subtypes could lead to the off-target adverse effects on skeletal and cardiac muscles due to acting on primary skeletal muscle sodium channel Nav1.4 and cardiac muscle sodium channel Nav1.5, respectively. For a long evolutionary process, some peptide toxins from venoms have been found to be highly potent yet selective on ion channel subtypes and, therefore, hold the promising potential to be developed into therapeutic agents. In this research, all-atom molecular dynamic methods were used to elucidate the selective mechanisms of an analgesic-antitumor β-scorpion toxin (AGAP) with human Nav1.4 and Nav1.5 in order to unravel the primary reason for the production of its adverse reactions on the skeletal and cardiac muscles. Our results suggest that the rational distribution of residues with ring structures near position 38 and positive residues in the C-terminal on AGAP are critical factors to ensure its analgesic efficacy. Moreover, the substitution for residues with benzene is beneficial to reduce its side effects.
Collapse
Affiliation(s)
- Fan Zhao
- School of Life Science and Biopharmaceutics, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang 110016, China
| | - Liangyi Fang
- School of Life Science and Biopharmaceutics, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang 110016, China
| | - Qi Wang
- School of Life Science and Biopharmaceutics, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang 110016, China
| | - Qi Ye
- School of Life Science and Biopharmaceutics, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang 110016, China
| | - Yanan He
- School of Life Science and Biopharmaceutics, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang 110016, China
| | - Weizhuo Xu
- Faculty of Functional Food and Wine, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang 110016, China
| | - Yongbo Song
- School of Life Science and Biopharmaceutics, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang 110016, China
| |
Collapse
|
6
|
Ding X, Yu F, He X, Xu S, Yang G, Ren W. Rubbing Salt in the Wound: Molecular Evolutionary Analysis of Pain-Related Genes Reveals the Pain Adaptation of Cetaceans in Seawater. Animals (Basel) 2022; 12:3571. [PMID: 36552490 PMCID: PMC9774174 DOI: 10.3390/ani12243571] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2022] [Revised: 11/26/2022] [Accepted: 12/14/2022] [Indexed: 12/23/2022] Open
Abstract
Pain, usually caused by a strong or disruptive stimulus, is an unpleasant sensation that serves as a warning to organisms. To adapt to extreme environments, some terrestrial animals have evolved to be inherently insensitive to pain. Cetaceans are known as supposedly indifferent to pain from soft tissue injury representatives of marine mammals. However, the molecular mechanisms that explain how cetaceans are adapted to pain in response to seawater environment remain unclear. Here, we performed a molecular evolutionary analysis of pain-related genes in selected representatives of cetaceans. ASIC4 gene was identified to be pseudogenized in all odontocetes (toothed whales) except from Physeter macrocephalus (sperm whales), and relaxed selection of this gene was detected in toothed whales with pseudogenized ASIC4. In addition, positive selection was detected in pain perception (i.e., ASIC3, ANO1, CCK, and SCN9A) and analgesia (i.e., ASIC3, ANO1, CCK, and SCN9A) genes among the examined cetaceans. In this study, potential convergent amino acid substitutions within predicted proteins were found among the examined cetaceans and other terrestrial mammals, inhabiting extreme environments (e.g., V441I of TRPV1 in cetaceans and naked mole rats). Moreover, specific amino acid substitutions within predicted sequences of several proteins were found in the studied representatives of cetaceans (e.g., F56L and D163A of ASIC3, E88G of GRK2, and F159L of OPRD1). Most of the substitutions were located within important functional domains of proteins, affecting their protein functions. The above evidence suggests that cetaceans might have undergone adaptive molecular evolution in pain-related genes through different evolutionary patterns to adapt to pain, resulting in greater sensitivity to pain and more effective analgesia. This study could have implications for diagnosis and treatment of human pain.
Collapse
Affiliation(s)
- Xiaoyue Ding
- Jiangsu Key Laboratory for Biodiversity and Biotechnology, College of Life Sciences, Nanjing Normal University, Nanjing 210000, China
| | - Fangfang Yu
- Jiangsu Key Laboratory for Biodiversity and Biotechnology, College of Life Sciences, Nanjing Normal University, Nanjing 210000, China
| | - Xiaofang He
- Jiangsu Key Laboratory for Biodiversity and Biotechnology, College of Life Sciences, Nanjing Normal University, Nanjing 210000, China
| | - Shixia Xu
- Jiangsu Key Laboratory for Biodiversity and Biotechnology, College of Life Sciences, Nanjing Normal University, Nanjing 210000, China
| | - Guang Yang
- Jiangsu Key Laboratory for Biodiversity and Biotechnology, College of Life Sciences, Nanjing Normal University, Nanjing 210000, China
| | - Wenhua Ren
- Jiangsu Key Laboratory for Biodiversity and Biotechnology, College of Life Sciences, Nanjing Normal University, Nanjing 210000, China
| |
Collapse
|
7
|
Cui Y, Wang T, Hao Z, Zhang J, Zhao Y. Methionine 58 is a key residue in the modulation of BmK scorpion toxin AGP-SYPU2 activity through in silico and in vivo studies. J Biomol Struct Dyn 2022; 40:2955-2962. [PMID: 33228478 DOI: 10.1080/07391102.2020.1848626] [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] [Indexed: 10/22/2022]
Abstract
Protein dynamic networks play an important role in the regulation of many protein systems. Some residues that are far away from the interface between proteins and their targets have a critical role in modulating the activity of some scorpion toxins. Here, conservation analysis combined with an in vivo experiment has reveals that Met58 is a key residue of BmK scorpion toxin AGP-SYPU2 in the modulation of analgesic activity. Molecular dynamics simulations clearly reveal the conformational changes that allow the loop between the β2 and β3 sheets to be exposed on the toxin surface to interact with its targets. Our results emphasize specific roles for the residue Met58 in the NC domain and our work gives valuable information for further modification of scorpion toxins to obtain new analgesic peptides with enhanced activity. Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Yong Cui
- School of Medical Devices, Shenyang Pharmaceutical University, Benxi, Liaoning Province, PR China
| | - Ting Wang
- School of Life Science and Bio-pharmaceutics, Shenyang Pharmaceutical University, Shenyang, Liaoning Province, PR China
| | - Zhihui Hao
- School of Life Science and Bio-pharmaceutics, Shenyang Pharmaceutical University, Shenyang, Liaoning Province, PR China
| | - Jinghai Zhang
- School of Medical Devices, Shenyang Pharmaceutical University, Benxi, Liaoning Province, PR China
| | - Yongshan Zhao
- School of Life Science and Bio-pharmaceutics, Shenyang Pharmaceutical University, Shenyang, Liaoning Province, PR China
| |
Collapse
|
8
|
Reduced Toxicity of Centruroides vittatus (Say, 1821) May Result from Lowered Sodium β Toxin Gene Expression and Toxin Protein Production. Toxins (Basel) 2021; 13:toxins13110828. [PMID: 34822614 PMCID: PMC8619477 DOI: 10.3390/toxins13110828] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2021] [Revised: 10/28/2021] [Accepted: 11/09/2021] [Indexed: 11/17/2022] Open
Abstract
Body tissue and venom glands from an eastern population of the scorpion Centruroides vittatus (Say, 1821) were homogenized and molecular constituents removed to characterize putative sodium β toxin gene diversity, RT-qPCR, transcriptomic, and proteomic variation. We cloned sodium β toxins from genomic DNA, conducted RT-qPCR experiments with seven sodium β toxin variants, performed venom gland tissue RNA-seq, and isolated venom proteins for mass spectrophotometry. We identified >70 putative novel sodium β toxin genes, 111 toxin gene transcripts, 24 different toxin proteins, and quantified sodium β toxin gene expression variation among individuals and between sexes. Our analyses contribute to the growing evidence that venom toxicity among scorpion taxa and their populations may be associated with toxin gene diversity, specific toxin transcripts variation, and subsequent protein production. Here, slight transcript variation among toxin gene variants may contribute to the major toxin protein variation in individual scorpion venom composition.
Collapse
|
9
|
Khoshdel Nezamiha F, Imani S, Arabi Mianroodi R, Tirgari S, Shahbazzadeh D. OdTx12/GNA, a chimeric variant of a β excitatory toxin from Odontobuthus doriae, reveals oral toxicity towards Locusta migratoria and Tenebrio molitor. Toxicon 2021; 202:13-19. [PMID: 34537212 DOI: 10.1016/j.toxicon.2021.09.001] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2021] [Revised: 08/26/2021] [Accepted: 09/08/2021] [Indexed: 11/18/2022]
Abstract
OdTx12, a beta excitatory toxin from Odontobothus doriae had previously been identified and characterized. It had been shown that OdTx12 causes significant lethal effects on insects by injection but does not show any toxicity on mice. Due to the natural ineffectiveness of scorpion toxins to act as oral toxins, OdTx12 was fused to Galanthus nivalis agglutinin (GNA), a protein with the potential to cross the insect gut. The sequence of OdTx12/GNA gene was chemically synthesized, cloned in Escherichia coli, and expressed. The effect of the purified fusion protein (OdTx12/GNA) was assessed on the insect and mammalian cell lines, insect larvae and mice. Toxicity assay on insect cell culture (SF9 cell line) showed comparable toxicity between OdTx12 and OdTx12/GNA (LD50 of 0.0030 and 0.0048 μM, respectively). Also very similar mortality rates were observed by injecting OdTx12 and OdTx12/GNA to Locusta migratoria and Tenebrio molitor. Oral administration of OdTx12/GNA, after five days of feeding, resulted in 96.6% and 98.3% mortality of L. migratoria and T. molitor larvae with an LC50 of 0.69 and 0.43 nmol/g of insect food, respectively, while OdTx12 alone did not cause any toxic effects on the larvae orally, suggesting the role of GNA in delivering the toxin to the insect's haemolymph. No toxicity or mortality was observed after toxicity testing of OdTx12/GNA on a mammalian cell line (HEP-2) or any mortality in vivo, by testing the protein in the laboratory mouse. Herein, we demonstrated that the fusion protein OdTx12/GNA could be considered an effective toxin for the biological control of insects.
Collapse
Affiliation(s)
| | - Sohrab Imani
- Department of Plant Protection, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | - Reza Arabi Mianroodi
- R&D Department, Production and Research Complex, Pasteur Institute of Iran, Tehran, Iran.
| | | | - Delavar Shahbazzadeh
- Department of Medical Biotechnology of Iran, Venom and Therapeutic Biomolecules Lab, Institute Pasteur of Iran, Tehran, Iran
| |
Collapse
|
10
|
Zhu Q, Du Y, Nomura Y, Gao R, Cang Z, Wei GW, Gordon D, Gurevitz M, Groome J, Dong K. Charge substitutions at the voltage-sensing module of domain III enhance actions of site-3 and site-4 toxins on an insect sodium channel. INSECT BIOCHEMISTRY AND MOLECULAR BIOLOGY 2021; 137:103625. [PMID: 34358664 PMCID: PMC9376739 DOI: 10.1016/j.ibmb.2021.103625] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/03/2021] [Revised: 07/29/2021] [Accepted: 07/29/2021] [Indexed: 06/13/2023]
Abstract
Scorpion α-toxins bind at the pharmacologically-defined site-3 on the sodium channel and inhibit channel inactivation by preventing the outward movement of the voltage sensor in domain IV (IVS4), whereas scorpion β-toxins bind at site-4 on the sodium channel and enhance channel activation by trapping the voltage sensor of domain II (IIS4) in its outward position. However, limited information is available on the role of the voltage-sensing modules (VSM, comprising S1-S4) of domains I and III in toxin actions. We have previously shown that charge reversing substitutions of the innermost positively-charged residues in IIIS4 (R4E, R5E) increase the activity of an insect-selective site-4 scorpion toxin, Lqh-dprIT3-c, on BgNav1-1a, a cockroach sodium channel. Here we show that substitutions R4E and R5E in IIIS4 also increase the activity of two site-3 toxins, LqhαIT from Leiurusquinquestriatus hebraeus and insect-selective Av3 from Anemonia viridis. Furthermore, charge reversal of either of two conserved negatively-charged residues, D1K and E2K, in IIIS2 also increase the action of the site-3 and site-4 toxins. Homology modeling suggests that S2-D1 and S2-E2 interact with S4-R4 and S4-R5 in the VSM of domain III (III-VSM), respectively, in the activated state of the channel. However, charge swapping between S2-D1 and S4-R4 had no compensatory effects on gating or toxin actions, suggesting that charged residue interactions are complex. Collectively, our results highlight the involvement of III-VSM in the actions of both site 3 and site 4 toxins, suggesting that charge reversing substitutions in III-VSM allosterically facilitate IIS4 or IVS4 voltage sensor trapping by these toxins.
Collapse
Affiliation(s)
- Qing Zhu
- Key Lab of Food Quality and Safety of Jiangsu Province-State Key Laboratory Breeding Base, Key Laboratory of Control Technology and Standard for Agro-product Safety and Quality, Ministry of Agriculture, Institute of Food Safety and Nutrition, Jiangsu Academy of Agricultural Sciences, Nanjing, China; Department of Entomology, Michigan State University, East Lansing, MI, USA
| | - Yuzhe Du
- Department of Entomology, Michigan State University, East Lansing, MI, USA
| | - Yoshiko Nomura
- Department of Entomology, Michigan State University, East Lansing, MI, USA
| | - Rong Gao
- Department of Hygienic Analysis and Detection, School of Public Health, Nanjing Medical University, 818 Tianyuan East Road, Nanjing, Jiangsu, China
| | - Zixuan Cang
- Department of Mathematics, Michigan State University, East Lansing, MI, USA
| | - Guo-Wei Wei
- Department of Mathematics, Michigan State University, East Lansing, MI, USA
| | - Dalia Gordon
- Department of Plant Molecular Biology & Ecology, George S. Wise Faculty of Life Sciences, Tel-Aviv University, Tel Aviv, Israel
| | - Michael Gurevitz
- Department of Plant Molecular Biology & Ecology, George S. Wise Faculty of Life Sciences, Tel-Aviv University, Tel Aviv, Israel.
| | - James Groome
- Department of Biological Sciences, Idaho State University, Pocatello, ID, USA
| | - Ke Dong
- Department of Entomology, Michigan State University, East Lansing, MI, USA; Department of Biology, Duke University, Durham, NC, USA.
| |
Collapse
|
11
|
Mapping the interaction surface of scorpion β-toxins with an insect sodium channel. Biochem J 2021; 478:2843-2869. [PMID: 34195804 PMCID: PMC10081811 DOI: 10.1042/bcj20210336] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2021] [Revised: 06/21/2021] [Accepted: 06/24/2021] [Indexed: 02/07/2023]
Abstract
The interaction of insect-selective scorpion depressant β-toxins (LqhIT2 and Lqh-dprIT3 from Leiurus quinquestriatus hebraeus) with the Blattella germanica sodium channel, BgNav1-1a, was investigated using site-directed mutagenesis, electrophysiological analyses, and structural modeling. Focusing on the pharmacologically defined binding site-4 of scorpion β-toxins at the voltage-sensing domain II (VSD-II), we found that charge neutralization of D802 in VSD-II greatly enhanced the channel sensitivity to Lqh-dprIT3. This was consistent with the high sensitivity of the splice variant BgNav2-1, bearing G802, to Lqh-dprIT3, and low sensitivity of BgNav2-1 mutant, G802D, to the toxin. Further mutational and electrophysiological analyses revealed that the sensitivity of the WT = D802E < D802G < D802A < D802K channel mutants to Lqh-dprIT3 correlated with the depolarizing shifts of activation in toxin-free channels. However, the sensitivity of single mutants involving IIS4 basic residues (K4E = WT << R1E < R2E < R3E) or double mutants (D802K = K4E/D802K = R3E/D802K > R2E/D802K > R1E/D802K > WT) did not correlate with the activation shifts. Using the cryo-EM structure of the Periplaneta americana channel, NavPaS, as a template and the crystal structure of LqhIT2, we constructed structural models of LqhIT2 and Lqh-dprIT3-c in complex with BgNav1-1a. These models along with the mutational analysis suggest that depressant toxins approach the salt-bridge between R1 and D802 at VSD-II to form contacts with linkers IIS1-S2, IIS3-S4, IIIP5-P1 and IIIP2-S6. Elimination of this salt-bridge enables deeper penetration of the toxin into a VSD-II gorge to form new contacts with the channel, leading to increased channel sensitivity to Lqh-dprIT3.
Collapse
|
12
|
Xu Y, Sun J, Yu Y, Kong X, Meng X, Liu Y, Cui Y, Su Y, Zhao M, Zhang J. Trp: a conserved aromatic residue crucial to the interaction of a scorpion peptide with sodium channels. J Biochem 2020; 168:633-641. [PMID: 32730584 DOI: 10.1093/jb/mvaa088] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2020] [Accepted: 07/03/2020] [Indexed: 11/14/2022] Open
Abstract
Anti-tumour-analgesic peptide (AGAP), one scorpion toxin purified from Buthus martensii Karsch, was known as its analgesic and anti-tumour activities. Trp38, a conserved aromatic residue of AGAP, might play important roles in its interaction with sodium channels. In this study, a mutant W38F was generated and effects of W38F were examined on hNav1.4, hNav1.5 and hNav1.7 by using whole-cell patch-clamp, which were closely associated to the biotoxicity of skeletal and cardiac muscles and pain signalling. The data showed that W38F decreased the inhibition effects of peak currents of hNav1.7, hNav1.4 and hNav1.5 compared with AGAP, notably, W38F reduced the analgesic activity compared with AGAP. The results suggested that Trp38 be a crucial amino acid involved in the interaction with these three sodium channels. The decreased analgesic activity of W38F might result from its much less inhibition of hNav1.7. These findings provided more information about the relationship between structure and function of AGAP and may facilitate the modification of other scorpion toxins with pharmacological effects.
Collapse
Affiliation(s)
- Yijia Xu
- School of Life Sciences and Biopharmaceutical Science, Shenyang Pharmaceutical University, 103 Wenhua road, Shenyang, Liaoning 110016, China
| | - Jianfang Sun
- College of Life and Health Sciences, Northeastern University, 195 Chuangxin road, Shenyang, Liaoning 110004, China
| | - Yue Yu
- College of Traditional Chinese Medicine, Shenyang Pharmaceutical University, 103 Wenhua road, Shenyang, Liaoning 110016, China
| | - Xiaohua Kong
- School of Life Sciences and Biopharmaceutical Science, Shenyang Pharmaceutical University, 103 Wenhua road, Shenyang, Liaoning 110016, China
| | - Xiangxue Meng
- School of Life Sciences and Biopharmaceutical Science, Shenyang Pharmaceutical University, 103 Wenhua road, Shenyang, Liaoning 110016, China
| | - Yanfeng Liu
- School of Life Sciences and Biopharmaceutical Science, Shenyang Pharmaceutical University, 103 Wenhua road, Shenyang, Liaoning 110016, China
| | - Yong Cui
- School of Medical Devices, Shenyang Pharmaceutical University, 103 Wenhua road, Shenyang, Liaoning 110016, China
| | - Yang Su
- Department of General Surgery, Shengjing Hospital of China Medical University, 36 Sanhao street, Shenyang, Liaoning 110004, China
| | - Mingyi Zhao
- School of Life Sciences and Biopharmaceutical Science, Shenyang Pharmaceutical University, 103 Wenhua road, Shenyang, Liaoning 110016, China
| | - Jinghai Zhang
- School of Medical Devices, Shenyang Pharmaceutical University, 103 Wenhua road, Shenyang, Liaoning 110016, China
| |
Collapse
|
13
|
Activation of voltage-gated sodium channels by BmK NT1 augments NMDA receptor function through Src family kinase signaling pathway in primary cerebellar granule cell cultures. Neuropharmacology 2020; 180:108291. [PMID: 32931812 DOI: 10.1016/j.neuropharm.2020.108291] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2020] [Revised: 08/23/2020] [Accepted: 08/26/2020] [Indexed: 12/29/2022]
Abstract
Voltage-gated sodium channels (VGSCs) are responsible for the generation and propagation of action potentials in excitable cells and are the molecular targets of an array of neurotoxins. BmK NT1, an α-scorpion toxin obtained from the scorpion Buthus martensii Karsch (BmK), produces neurotoxicity that is associated with extracellular Ca2+ influx through Na+-Ca2+ exchangers, N-methyl-d-aspartic acid (NMDA) receptors, and L-type Ca2+ channels in cultured cerebellar granule cells (CGCs). In the present study, we demonstrated that BmK NT1 triggered concentration-dependent release of excitatory neurotransmitters, glutamate and aspartate; both effects were eliminated by VGSC blocker, tetrodotoxin. More importantly, we demonstrated that a threshold concentration of BmK NT1 that produced marginal Ca2+ influx and neuronal death augmented glutamate-induced Ca2+ elevation and neuronal death in CGCs. BmK NT1-augmented glutamate-induced Ca2+ influx and neuronal death were suppressed by tetrodotoxin and MK-801 suggesting that the augmentation was through activation of VGSCs and NMDA receptors. Consistently, BmK NT1 also enhanced NMDA-induced Ca2+ influx. Further mechanistic investigations demonstrated that BmK NT1 increased the expression level of NMDA receptors on the plasma membrane and increased the phosphorylation level of NR2B at Tyr1472. Src family kinase inhibitor, 1-tert-butyl-3-(4-chlorophenyl)pyrazolo[3,4-d]pyrimidin-4-yl]amine (PP2), but not the inactive analogue, 4-amino-1-phenylpyrazolo[3,4-d]pyrimidine (PP3), eliminated BmK NT1-triggered NR2B phosphorylation, NMDA receptor trafficking, as well as BmK NT1-augmented NMDA Ca2+ response and neuronal death. Considered together, these data demonstrated that both presynaptic (excitatory amino acid release) and postsynaptic mechanisms (augmentation of NMDA receptor function) are critical for VGSC activation-induced neurotoxicity in primary CGC cultures.
Collapse
|
14
|
Khoshdel Nezamiha F, Imani S, Shahbazzadeh D, Tirgari S, Arabi Mianroodi R. Cloning and expression of OdTx12, a β excitatory toxin from Odontobuthus doriae, in Escherichia coli and evaluation of its bioactivity in Locusta migratoria. Toxicon 2020; 183:20-28. [PMID: 32442468 DOI: 10.1016/j.toxicon.2020.05.003] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2019] [Revised: 04/06/2020] [Accepted: 05/06/2020] [Indexed: 01/11/2023]
Abstract
The venom of Odontobuthus doriae contains several peptide toxins that interfere with the sodium channel function of cell membranes, some of which specifically act on the insect's sodium channel without affecting mammalian cells. In this study sodium channel toxins of Odontobuthus doriae were aligned to other closely related toxins by BLAST and ClustalW servers. Among these toxins, NaTx12 (OdTx12) showed more than 90% similarity to the most known beta excitatory toxin, AaHIT1; furthermore, our modeling studies confirmed high tertiary structure similarity of these proteins. OdTx12 was cloned and expressed in E.coli, using pET26-b and pET28-a expression vectors. Tris-tricine SDS-PAGE and Western blot analysis showed OdTx12 expression by pET28-a, only. After purification, bioactivity of the purified protein was analyzed by injection and oral administration to Locusta migratoria larvae, and toxicity to mammals was tested on mice. Injection of OdTx12 resulted in the killing of larvae with LD50 of 0.4 and 0.2 after 48 and 72 h respectively, but oral administration of OdTx12 had no significant effect on Locusta migratoria, nor did the injection to mice show any signs of toxicity. These results showed that OdTx12, as a novel β excitatory toxin can be considered as a candidate for insect control purposes.
Collapse
Affiliation(s)
| | - Sohrab Imani
- Department of Entomology, Islamic Azad University (Science and Research Branch), Tehran, Iran
| | - Delavar Shahbazzadeh
- Department of Medical Biotechnology of Iran, Venom and Therapeutic Biomolecules Lab, Institute Pasteur of Iran, Tehran, Iran
| | | | - Reza Arabi Mianroodi
- R&D Department, Production and Research Complex, Pasteur Institute of Iran, Tehran, Iran.
| |
Collapse
|
15
|
Biochemical characterization of the venom from the Mexican scorpion Centruroides ornatus, a dangerous species to humans. Toxicon 2020; 173:27-38. [DOI: 10.1016/j.toxicon.2019.11.004] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2019] [Revised: 11/08/2019] [Accepted: 11/11/2019] [Indexed: 11/19/2022]
|
16
|
Characterization, molecular modeling and phylogenetic analysis of a long mammalian neurotoxin from the venom of the Iranian scorpion Androctonus crassicauda. Biologia (Bratisl) 2019. [DOI: 10.2478/s11756-019-00400-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
|
17
|
Delgado-Prudencio G, Possani LD, Becerril B, Ortiz E. The Dual α-Amidation System in Scorpion Venom Glands. Toxins (Basel) 2019; 11:toxins11070425. [PMID: 31330798 PMCID: PMC6669573 DOI: 10.3390/toxins11070425] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2019] [Revised: 07/18/2019] [Accepted: 07/18/2019] [Indexed: 12/14/2022] Open
Abstract
Many peptides in scorpion venoms are amidated at their C-termini. This post-translational modification is paramount for the correct biological function of ion channel toxins and antimicrobial peptides, among others. The discovery of canonical amidation sequences in transcriptome-derived scorpion proproteins suggests that a conserved enzymatic α-amidation system must be responsible for this modification of scorpion peptides. A transcriptomic approach was employed to identify sequences putatively encoding enzymes of the α-amidation pathway. A dual enzymatic α-amidation system was found, consisting of the membrane-anchored, bifunctional, peptidylglycine α-amidating monooxygenase (PAM) and its paralogs, soluble monofunctional peptidylglycine α-hydroxylating monooxygenase (PHMm) and peptidyl-α-hydroxyglycine α-amidating lyase (PALm). Independent genes encode these three enzymes. Amino acid residues responsible for ion coordination and enzymatic activity are conserved in these sequences, suggesting that the enzymes are functional. Potential endoproteolytic recognition sites for proprotein convertases in the PAM sequence indicate that PAM-derived soluble isoforms may also be expressed. Sequences potentially encoding proprotein convertases (PC1 and PC2), carboxypeptidase E (CPE), and other enzymes of the α-amidation pathway, were also found, confirming the presence of this pathway in scorpions.
Collapse
Affiliation(s)
- Gustavo Delgado-Prudencio
- Departamento de Medicina Molecular y Bioprocesos, Instituto de Biotecnología, Universidad Nacional Autónoma de México, Avenida Universidad 2001, Colonia Chamilpa, Cuernavaca, Morelos 62210, Mexico
| | - Lourival D Possani
- Departamento de Medicina Molecular y Bioprocesos, Instituto de Biotecnología, Universidad Nacional Autónoma de México, Avenida Universidad 2001, Colonia Chamilpa, Cuernavaca, Morelos 62210, Mexico
| | - Baltazar Becerril
- Departamento de Medicina Molecular y Bioprocesos, Instituto de Biotecnología, Universidad Nacional Autónoma de México, Avenida Universidad 2001, Colonia Chamilpa, Cuernavaca, Morelos 62210, Mexico
| | - Ernesto Ortiz
- Departamento de Medicina Molecular y Bioprocesos, Instituto de Biotecnología, Universidad Nacional Autónoma de México, Avenida Universidad 2001, Colonia Chamilpa, Cuernavaca, Morelos 62210, Mexico.
| |
Collapse
|
18
|
Anti-tumoral effect of scorpion peptides: Emerging new cellular targets and signaling pathways. Cell Calcium 2019; 80:160-174. [DOI: 10.1016/j.ceca.2019.05.003] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2019] [Revised: 05/10/2019] [Accepted: 05/10/2019] [Indexed: 12/31/2022]
|
19
|
Zhao F, Wang JL, Ming HY, Zhang YN, Dun YQ, Zhang JH, Song YB. Insights into the binding mode and functional components of the analgesic-antitumour peptide from Buthus martensii Karsch to human voltage-gated sodium channel 1.7 based on dynamic simulation analysis. J Biomol Struct Dyn 2019; 38:1868-1879. [PMID: 31099313 DOI: 10.1080/07391102.2019.1620126] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Abstract
Voltage-gated sodium (Nav) channels are transmembrane proteins composed of four homologous domains (DI-DIV) that play important roles in membrane excitability in neurons and muscles. Analgesic-antitumour peptide (AGAP) is a neurotoxin from the scorpion Buthus martensii Karsch, and has been shown to exert analgesic effect by binding on site 4 of human Nav1.7 (hNav1.7). Mechanistic details about this binding, however, remain unclear. To address this issue, we compared the binding modes of AGAP/AGAPW38G/AGAPW38F and the hNav1.7 voltage-sensing domain on DII (VSD2hNav1.7) using homology modeling, molecular docking, molecular dynamics simulation and steered molecular dynamics. Results revealed the key role of tryptophan at position 38 on the binding of AGAP to VSD2hNav1.7. Pivotal roles are played also by residues on the β-turn and negatively charged residues at the C-terminal. We further show that electrostatic interaction is the main contributor to the binding free energy of the complex. Agreement between our computational simulation findings and prior experimental data supports the accuracy of the described mechanism. Accordingly, these results can provide valuable information for designing potent toxin analgesics targeting hNav1.7 with high affinity.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Fan Zhao
- School of Life Science and Bio-pharmaceutics, Shenyang Pharmaceutical University, Shenyang, China
| | - Jin-Long Wang
- School of Life Science and Bio-pharmaceutics, Shenyang Pharmaceutical University, Shenyang, China
| | - Hong-Yan Ming
- The First Affiliated Hospital of Xinxiang Medical University, Weihui, China
| | - Ya-Nan Zhang
- School of Life Science and Bio-pharmaceutics, Shenyang Pharmaceutical University, Shenyang, China
| | - Ying-Qiao Dun
- School of Life Science and Bio-pharmaceutics, Shenyang Pharmaceutical University, Shenyang, China
| | - Jing-Hai Zhang
- School of Life Science and Bio-pharmaceutics, Shenyang Pharmaceutical University, Shenyang, China
| | - Yong-Bo Song
- School of Life Science and Bio-pharmaceutics, Shenyang Pharmaceutical University, Shenyang, China
| |
Collapse
|
20
|
Romero-Gutiérrez MT, Santibáñez-López CE, Jiménez-Vargas JM, Batista CVF, Ortiz E, Possani LD. Transcriptomic and Proteomic Analyses Reveal the Diversity of Venom Components from the Vaejovid Scorpion Serradigitus gertschi. Toxins (Basel) 2018; 10:E359. [PMID: 30189638 PMCID: PMC6162517 DOI: 10.3390/toxins10090359] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2018] [Revised: 08/29/2018] [Accepted: 09/01/2018] [Indexed: 12/22/2022] Open
Abstract
To understand the diversity of scorpion venom, RNA from venomous glands from a sawfinger scorpion, Serradigitus gertschi, of the family Vaejovidae, was extracted and used for transcriptomic analysis. A total of 84,835 transcripts were assembled after Illumina sequencing. From those, 119 transcripts were annotated and found to putatively code for peptides or proteins that share sequence similarities with the previously reported venom components of other species. In accordance with sequence similarity, the transcripts were classified as potentially coding for 37 ion channel toxins; 17 host defense peptides; 28 enzymes, including phospholipases, hyaluronidases, metalloproteases, and serine proteases; nine protease inhibitor-like peptides; 10 peptides of the cysteine-rich secretory proteins, antigen 5, and pathogenesis-related 1 protein superfamily; seven La1-like peptides; and 11 sequences classified as "other venom components". A mass fingerprint performed by mass spectrometry identified 204 components with molecular masses varying from 444.26 Da to 12,432.80 Da, plus several higher molecular weight proteins whose precise masses were not determined. The LC-MS/MS analysis of a tryptic digestion of the soluble venom resulted in the de novo determination of 16,840 peptide sequences, 24 of which matched sequences predicted from the translated transcriptome. The database presented here increases our general knowledge of the biodiversity of venom components from neglected non-buthid scorpions.
Collapse
Affiliation(s)
- Maria Teresa Romero-Gutiérrez
- Departamento de Medicina Molecular y Bioprocesos, Instituto de Biotecnología, Universidad Nacional Autónoma de México, Avenida Universidad 2001, Apartado Postal 510-3, Cuernavaca, Morelos 62210, Mexico.
| | - Carlos Eduardo Santibáñez-López
- Departamento de Medicina Molecular y Bioprocesos, Instituto de Biotecnología, Universidad Nacional Autónoma de México, Avenida Universidad 2001, Apartado Postal 510-3, Cuernavaca, Morelos 62210, Mexico.
- Department of Integrative Biology, University of Wisconsin⁻Madison, Madison, WI 53706, USA.
| | - Juana María Jiménez-Vargas
- Departamento de Medicina Molecular y Bioprocesos, Instituto de Biotecnología, Universidad Nacional Autónoma de México, Avenida Universidad 2001, Apartado Postal 510-3, Cuernavaca, Morelos 62210, Mexico.
| | - Cesar Vicente Ferreira Batista
- Laboratorio Universitario de Proteómica, Instituto de Biotecnología, Universidad Nacional Autónoma de México, Avenida Universidad 2001, Apartado Postal 510-3, Cuernavaca, Morelos 62210, Mexico.
| | - Ernesto Ortiz
- Departamento de Medicina Molecular y Bioprocesos, Instituto de Biotecnología, Universidad Nacional Autónoma de México, Avenida Universidad 2001, Apartado Postal 510-3, Cuernavaca, Morelos 62210, Mexico.
| | - Lourival Domingos Possani
- Departamento de Medicina Molecular y Bioprocesos, Instituto de Biotecnología, Universidad Nacional Autónoma de México, Avenida Universidad 2001, Apartado Postal 510-3, Cuernavaca, Morelos 62210, Mexico.
| |
Collapse
|
21
|
The diversity of venom components of the scorpion species Paravaejovis schwenkmeyeri (Scorpiones: Vaejovidae) revealed by transcriptome and proteome analyses. Toxicon 2018; 151:47-62. [DOI: 10.1016/j.toxicon.2018.06.085] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2018] [Revised: 06/20/2018] [Accepted: 06/25/2018] [Indexed: 12/11/2022]
|
22
|
Wang L, Zellmer SG, Printzenhoff DM, Castle NA. PF-06526290 can both enhance and inhibit conduction through voltage-gated sodium channels. Br J Pharmacol 2018; 175:2926-2939. [PMID: 29791744 DOI: 10.1111/bph.14338] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2017] [Revised: 03/06/2018] [Accepted: 03/17/2018] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND AND PURPOSE Pharmacological agents that either inhibit or enhance flux of ions through voltage-gated sodium (Nav ) channels may provide opportunities for treatment of human health disorders. During studies to characterize agents that modulate Nav 1.3 function, we identified a compound that appears to exhibit both enhancement and inhibition of sodium ion conduction that appeared to be dependent on the gating state that the channel was in. The objective of the current study was to determine if these different modulatory effects are mediated by the same or distinct interactions with the channel. EXPERIMENTAL APPROACH Electrophysiology and site-directed mutation were used to investigate the effects of PF-06526290 on Nav channel function. KEY RESULTS PF-06526290 greatly slows inactivation of Nav channels in a subtype-independent manner. However, upon prolonged depolarization to induce inactivation, PF-06526290 becomes a Nav subtype-selective inhibitor. Mutation of the domain 4 voltage sensor modulates inhibition of Nav 1.3 or Nav 1.7 channels by PF-06526290 but has no effect on PF-06526290 mediated slowing of inactivation. CONCLUSIONS AND IMPLICATIONS These findings suggest that distinct interactions may underlie the two modes of Nav channel modulation by PF-06526290 and that a single compound can affect sodium channel function in several ways.
Collapse
Affiliation(s)
- Lingxin Wang
- Department of Molecular and Cellular Physiology, Stanford University, Stanford, CA, USA
| | | | | | | |
Collapse
|
23
|
Carcamo-Noriega EN, Olamendi-Portugal T, Restano-Cassulini R, Rowe A, Uribe-Romero SJ, Becerril B, Possani LD. Intraspecific variation of Centruroides sculpturatus scorpion venom from two regions of Arizona. Arch Biochem Biophys 2018; 638:52-57. [DOI: 10.1016/j.abb.2017.12.012] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2017] [Revised: 12/12/2017] [Accepted: 12/15/2017] [Indexed: 10/18/2022]
|
24
|
Abstract
Voltage-gated sodium channels (VGSCs) are critical in generation and conduction of electrical signals in multiple excitable tissues. Natural toxins, produced by animal, plant, and microorganisms, target VGSCs through diverse strategies developed over millions of years of evolutions. Studying of the diverse interaction between VGSC and VGSC-targeting toxins has been contributing to the increasing understanding of molecular structure and function, pharmacology, and drug development potential of VGSCs. This chapter aims to summarize some of the current views on the VGSC-toxin interaction based on the established receptor sites of VGSC for natural toxins.
Collapse
Affiliation(s)
- Yonghua Ji
- Laboratory of Neuropharmacology and Neurotoxicology, Shanghai University, Shanghai, China.
| |
Collapse
|
25
|
Yuan S, Gao B, Zhu S. Molecular Dynamics Simulation Reveals Specific Interaction Sites between Scorpion Toxins and K v1.2 Channel: Implications for Design of Highly Selective Drugs. Toxins (Basel) 2017; 9:toxins9110354. [PMID: 29104247 PMCID: PMC5705969 DOI: 10.3390/toxins9110354] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2017] [Revised: 10/15/2017] [Accepted: 10/19/2017] [Indexed: 01/06/2023] Open
Abstract
The Kv1.2 channel plays an important role in the maintenance of resting membrane potential and the regulation of the cellular excitability of neurons, whose silencing or mutations can elicit neuropathic pain or neurological diseases (e.g., epilepsy and ataxia). Scorpion venom contains a variety of peptide toxins targeting the pore region of this channel. Despite a large amount of structural and functional data currently available, their detailed interaction modes are poorly understood. In this work, we choose four Kv1.2-targeted scorpion toxins (Margatoxin, Agitoxin-2, OsK-1, and Mesomartoxin) to construct their complexes with Kv1.2 based on the experimental structure of ChTx-Kv1.2. Molecular dynamics simulation of these complexes lead to the identification of hydrophobic patches, hydrogen-bonds, and salt bridges as three essential forces mediating the interactions between this channel and the toxins, in which four Kv1.2-specific interacting amino acids (D353, Q358, V381, and T383) are identified for the first time. This discovery might help design highly selective Kv1.2-channel inhibitors by altering amino acids of these toxins binding to the four channel residues. Finally, our results provide new evidence in favor of an induced fit model between scorpion toxins and K+ channel interactions.
Collapse
Affiliation(s)
- Shouli Yuan
- Group of Peptide Biology and Evolution, State Key Laboratory of Integrated Management of Pest Insects and Rodents, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China.
- College of Resources and Environment, University of Chinese Academy of Sciences, Beijing 100049, China.
| | - Bin Gao
- Group of Peptide Biology and Evolution, State Key Laboratory of Integrated Management of Pest Insects and Rodents, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China.
| | - Shunyi Zhu
- Group of Peptide Biology and Evolution, State Key Laboratory of Integrated Management of Pest Insects and Rodents, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China.
| |
Collapse
|
26
|
Duque HM, Mourão CBF, Tibery DV, Barbosa EA, Campos LA, Schwartz EF. To4, the first Tityus obscurus β-toxin fully electrophysiologically characterized on human sodium channel isoforms. Peptides 2017; 95:106-115. [PMID: 28735770 DOI: 10.1016/j.peptides.2017.07.010] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/19/2017] [Revised: 07/13/2017] [Accepted: 07/14/2017] [Indexed: 12/19/2022]
Abstract
Many scorpion toxins that act on sodium channels (NaScTxs) have been characterized till date. These toxins may act modulating the inactivation or the activation of sodium channels and are named α- or β-types, respectively. Some venom toxins from Tityus obscurus (Buthidae), a scorpion widely distributed in the Brazilian Amazon, have been partially characterized in previous studies; however, little information about their electrophysiological role on sodium ion channels has been published. In the present study, we describe the purification, identification and electrophysiological characterization of a NaScTx, which was first described as Tc54 and further fully sequenced and renamed To4. This toxin shows a marked β-type effect on different sodium channel subtypes (hNav1.1-hNav1.7) at low concentrations, and has more pronounced activity on hNav1.1, hNav1.2 and hNav1.4. By comparing To4 primary structure with other Tityus β-toxins which have already been electrophysiologically tested, it is possible to establish some key amino acid residues for the sodium channel activity. Thus, To4 is the first toxin from T. obscurus fully electrophysiologically characterized on different human sodium channel isoforms.
Collapse
Affiliation(s)
- Harry Morales Duque
- Laboratório de Neurofarmacologia, Departamento de Ciências Fisiológicas, Universidade de Brasília, Instituto de Ciências Biológicas, Brasília, 70910-900, DF, Brazil
| | - Caroline Barbosa Farias Mourão
- Laboratório de Neurofarmacologia, Departamento de Ciências Fisiológicas, Universidade de Brasília, Instituto de Ciências Biológicas, Brasília, 70910-900, DF, Brazil; Instituto Federal de Educação, Ciência e Tecnologia de Brasília, Campus Ceilândia, Brasília 72220-260, DF, Brazil
| | - Diogo Vieira Tibery
- Laboratório de Neurofarmacologia, Departamento de Ciências Fisiológicas, Universidade de Brasília, Instituto de Ciências Biológicas, Brasília, 70910-900, DF, Brazil
| | - Eder Alves Barbosa
- LSAB - Laboratório de Síntese e Análise de Biomoléculas, Instituto de Química, Universidade de Brasília, Brasília 70910-900, DF, Brazil
| | - Leandro Ambrósio Campos
- Laboratório de Neurofarmacologia, Departamento de Ciências Fisiológicas, Universidade de Brasília, Instituto de Ciências Biológicas, Brasília, 70910-900, DF, Brazil
| | - Elisabeth Ferroni Schwartz
- Laboratório de Neurofarmacologia, Departamento de Ciências Fisiológicas, Universidade de Brasília, Instituto de Ciências Biológicas, Brasília, 70910-900, DF, Brazil.
| |
Collapse
|
27
|
Hopp BH, Arvidson RS, Adams ME, Razak KA. Arizona bark scorpion venom resistance in the pallid bat, Antrozous pallidus. PLoS One 2017; 12:e0183215. [PMID: 28854259 PMCID: PMC5576675 DOI: 10.1371/journal.pone.0183215] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2016] [Accepted: 08/01/2017] [Indexed: 11/19/2022] Open
Abstract
The pallid bat (Antrozous pallidus), a gleaning bat found in the western United States and Mexico, hunts a wide variety of ground-dwelling prey, including scorpions. Anecdotal evidence suggests that the pallid bat is resistant to scorpion venom, but no systematic study has been performed. Here we show with behavioral measures and direct injection of venom that the pallid bat is resistant to venom of the Arizona bark scorpion, Centruroides sculpturatus. Our results show that the pallid bat is stung multiple times during a hunt without any noticeable effect on behavior. In addition, direct injection of venom at mouse LD50 concentrations (1.5 mg/kg) has no effect on bat behavior. At the highest concentration tested (10 mg/kg), three out of four bats showed no effects. One of the four bats showed a transient effect suggesting that additional studies are required to identify potential regional variation in venom tolerance. Scorpion venom is a cocktail of toxins, some of which activate voltage-gated sodium ion channels, causing intense pain. Dorsal root ganglia (DRG) contain nociceptive neurons and are principal targets of scorpion venom toxins. To understand if mutations in specific ion channels contribute to venom resistance, a pallid bat DRG transcriptome was generated. As sodium channels are a major target of scorpion venom, we identified amino acid substitutions present in the pallid bat that may lead to venom resistance. Some of these substitutions are similar to corresponding amino acids in sodium channel isoforms responsible for reduced venom binding activity. The substitution found previously in the grasshopper mouse providing venom resistance to the bark scorpion is not present in the pallid bat, indicating a potentially novel mechanism for venom resistance in the bat that remains to be identified. Taken together, these results indicate that the pallid bat is resistant to venom of the bark scorpion and altered sodium ion channel function may partly underlie such resistance.
Collapse
Affiliation(s)
- Bradley H. Hopp
- Graduate Neuroscience Program, University of California, Riverside, California, United States of America
| | - Ryan S. Arvidson
- Departments of Entomology and Cell Biology & Neuroscience, University of California, Riverside, California, United States of America
| | - Michael E. Adams
- Graduate Neuroscience Program, University of California, Riverside, California, United States of America
- Departments of Entomology and Cell Biology & Neuroscience, University of California, Riverside, California, United States of America
| | - Khaleel A. Razak
- Graduate Neuroscience Program, University of California, Riverside, California, United States of America
- Department of Psychology, University of California, Riverside, California, United States of America
- * E-mail:
| |
Collapse
|
28
|
Meng X, Xu Y, Wang F, Zhao M, Hou X, Ma Y, Jin Y, Liu Y, Song Y, Zhang J. The roles of conserved aromatic residues (Tyr5 and Tyr42) in interaction of scorpion toxin BmK AGP-SYPU1 with human Na v 1.7. Int J Biol Macromol 2017; 99:105-111. [DOI: 10.1016/j.ijbiomac.2017.01.020] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2016] [Revised: 12/25/2016] [Accepted: 01/04/2017] [Indexed: 12/20/2022]
|
29
|
Paramonov AS, Lyukmanova EN, Myshkin MY, Shulepko MA, Kulbatskii DS, Petrosian NS, Chugunov AO, Dolgikh DA, Kirpichnikov MP, Arseniev AS, Shenkarev ZO. NMR investigation of the isolated second voltage-sensing domain of human Nav1.4 channel. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2017; 1859:493-506. [PMID: 28065835 DOI: 10.1016/j.bbamem.2017.01.004] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/30/2016] [Revised: 12/06/2016] [Accepted: 01/04/2017] [Indexed: 10/20/2022]
Abstract
Voltage-gated Na+ channels are essential for the functioning of cardiovascular, muscular, and nervous systems. The α-subunit of eukaryotic Na+ channel consists of ~2000 amino acid residues and encloses 24 transmembrane (TM) helices, which form five membrane domains: four voltage-sensing (VSD) and one pore domain. The structural complexity significantly impedes recombinant production and structural studies of full-sized Na+ channels. Modular organization of voltage-gated channels gives an idea for studying of the isolated second VSD of human skeletal muscle Nav1.4 channel (VSD-II). Several variants of VSD-II (~150a.a., four TM helices) with different N- and C-termini were produced by cell-free expression. Screening of membrane mimetics revealed low stability of VSD-II samples in media containing phospholipids (bicelles, nanodiscs) associated with the aggregation of electrically neutral domain molecules. The almost complete resonance assignment of 13C,15N-labeled VSD-II was obtained in LPPG micelles. The secondary structure of VSD-II showed similarity with the structures of bacterial Na+ channels. The fragment of S4 TM helix between the first and second conserved Arg residues probably adopts 310-helical conformation. Water accessibility of S3 helix, observed by the Mn2+ titration, pointed to the formation of water-filled crevices in the micelle embedded VSD-II. 15N relaxation data revealed characteristic pattern of μs-ms time scale motions in the VSD-II regions sharing expected interhelical contacts. VSD-II demonstrated enhanced mobility at ps-ns time scale as compared to isolated VSDs of K+ channels. These results validate structural studies of isolated VSDs of Na+ channels and show possible pitfalls in application of this 'divide and conquer' approach.
Collapse
Affiliation(s)
- A S Paramonov
- Shemyakin and Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Miklukho-Maklaya str., 16/10, Moscow 117997, Russia; Lomonosov Moscow State University, Moscow 119991, Russia
| | - E N Lyukmanova
- Shemyakin and Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Miklukho-Maklaya str., 16/10, Moscow 117997, Russia; Lomonosov Moscow State University, Moscow 119991, Russia
| | - M Yu Myshkin
- Shemyakin and Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Miklukho-Maklaya str., 16/10, Moscow 117997, Russia; Moscow Institute of Physics and Technology (State University), Institutskiy Pereulok 9, Dolgoprudny, Moscow Region 141700, Russia
| | - M A Shulepko
- Shemyakin and Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Miklukho-Maklaya str., 16/10, Moscow 117997, Russia; Lomonosov Moscow State University, Moscow 119991, Russia
| | - D S Kulbatskii
- Shemyakin and Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Miklukho-Maklaya str., 16/10, Moscow 117997, Russia; Lomonosov Moscow State University, Moscow 119991, Russia
| | - N S Petrosian
- Shemyakin and Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Miklukho-Maklaya str., 16/10, Moscow 117997, Russia; Moscow Institute of Physics and Technology (State University), Institutskiy Pereulok 9, Dolgoprudny, Moscow Region 141700, Russia
| | - A O Chugunov
- Shemyakin and Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Miklukho-Maklaya str., 16/10, Moscow 117997, Russia
| | - D A Dolgikh
- Shemyakin and Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Miklukho-Maklaya str., 16/10, Moscow 117997, Russia; Lomonosov Moscow State University, Moscow 119991, Russia
| | - M P Kirpichnikov
- Shemyakin and Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Miklukho-Maklaya str., 16/10, Moscow 117997, Russia; Lomonosov Moscow State University, Moscow 119991, Russia
| | - A S Arseniev
- Shemyakin and Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Miklukho-Maklaya str., 16/10, Moscow 117997, Russia; Moscow Institute of Physics and Technology (State University), Institutskiy Pereulok 9, Dolgoprudny, Moscow Region 141700, Russia
| | - Z O Shenkarev
- Shemyakin and Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Miklukho-Maklaya str., 16/10, Moscow 117997, Russia; Moscow Institute of Physics and Technology (State University), Institutskiy Pereulok 9, Dolgoprudny, Moscow Region 141700, Russia.
| |
Collapse
|
30
|
Olamendi-Portugal T, Restano-Cassulini R, Riaño-Umbarila L, Becerril B, Possani LD. Functional and immuno-reactive characterization of a previously undescribed peptide from the venom of the scorpion Centruroides limpidus. Peptides 2017; 87:34-40. [PMID: 27871874 DOI: 10.1016/j.peptides.2016.11.008] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/12/2016] [Revised: 11/11/2016] [Accepted: 11/14/2016] [Indexed: 11/23/2022]
Abstract
A previously undescribed toxic peptide named Cl13 was purified from the venom of the Mexican scorpion Centruroides limpidus. It contains 66 amino acid residues, including four disulfide bonds. The physiological effects assayed in 7 different subtypes of voltage gated Na+-channels, showed that it belongs to the β-scorpion toxin type. The most notorious effects were observed in subtypes Nav1.4, Nav1.5 and Nav1.6. Although having important sequence similarities with two other lethal toxins from this scorpion species (Cll1m and Cll2), the recently developed single chain antibody fragments (scFv) of human origin were not capable of protecting against Cl13. At the amino acid sequence level, in 3 stretches of peptide Cl13 (positions 7-9, 30-38 and 62-66) some differences with respect to other similar toxins are observed. Some of these differences coincide with contact points with the human antibody fragments.
Collapse
Affiliation(s)
- Timoteo Olamendi-Portugal
- Departamento de Medicina Molecular, Instituto de Biotecnología, Universidad Nacional Autónoma de México, Avenida Universidad, 2001, Apartado Postal 510.3 Cuernavaca, Morelos, 62210, Mexico
| | - Rita Restano-Cassulini
- Departamento de Medicina Molecular, Instituto de Biotecnología, Universidad Nacional Autónoma de México, Avenida Universidad, 2001, Apartado Postal 510.3 Cuernavaca, Morelos, 62210, Mexico
| | - Lidia Riaño-Umbarila
- CONACYT, Instituto de Biotecnología,Universidad Nacional Autónoma de México, Mexico
| | - Baltazar Becerril
- Departamento de Medicina Molecular, Instituto de Biotecnología, Universidad Nacional Autónoma de México, Avenida Universidad, 2001, Apartado Postal 510.3 Cuernavaca, Morelos, 62210, Mexico
| | - Lourival D Possani
- Departamento de Medicina Molecular, Instituto de Biotecnología, Universidad Nacional Autónoma de México, Avenida Universidad, 2001, Apartado Postal 510.3 Cuernavaca, Morelos, 62210, Mexico.
| |
Collapse
|
31
|
Zhang S, Gao B, Zhu S. Independent Origins of Scorpion Toxins Affecting Potassium and Sodium Channels. EVOLUTION OF VENOMOUS ANIMALS AND THEIR TOXINS 2017. [DOI: 10.1007/978-94-007-6458-3_12] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
|
32
|
Xu X, Zhang B, Yang S, An S, Ribeiro JMC, Andersen JF. Structure and Function of FS50, a salivary protein from the flea Xenopsylla cheopis that blocks the sodium channel Na V1.5. Sci Rep 2016; 6:36574. [PMID: 27819327 PMCID: PMC5098211 DOI: 10.1038/srep36574] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2016] [Accepted: 10/14/2016] [Indexed: 11/10/2022] Open
Abstract
Naturally occurring toxins have been invaluable tools for the study of structural and functional relationships of voltage-gated sodium channels (VGSC). Few studies have been made of potential channel-modulating substances from blood-feeding arthropods. He we describe the characterization FS50, a salivary protein from the flea, Xenopsylla cheopis, that exhibits an inhibitory activity against the NaV1.5 channel with an IC50 of 1.58 μM. The pore-blocking mechanism of this toxin is evident from the kinetics of activation and inactivation suggesting that FS50 does not interfere with the voltage sensor of NaV1.5. FS50 exhibits high specificity for NaV1.5, since 10 μM FS50 had no discernable effect on voltage-gated Na+, K+ and Ca2+ channels in rat dorsal root ganglia or VGSC forms individually expressed in HEK 293T cells. Furthermore, intravenous injection of FS50 into rats and monkeys elicited recovery from arrhythmia induced by BaCl2, as would be expected from a blockade of NaV1.5. The crystal structure of FS50 revealed a βαββ domain similar to that of scorpion β toxin and a small N-terminal βαβ domain. Site-directed mutagenesis experiments have implicated a basic surface including the side chains of Arg 6, His 11 and Lys 32 as potentially important in the FS50 NaV1.5 interaction.
Collapse
Affiliation(s)
- Xueqing Xu
- Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, Guangdong, China.,The Laboratory of Malaria and Vector Research, NIAID, National Institutes of Health, Rockville, Maryland 20852 USA
| | - Bei Zhang
- Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, Guangdong, China
| | - Shilong Yang
- The Laboratory of Animal Models and Human Disease Mechanisms, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming 650223, Yunnan, China
| | - Su An
- The Laboratory of Animal Models and Human Disease Mechanisms, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming 650223, Yunnan, China
| | - José M C Ribeiro
- The Laboratory of Malaria and Vector Research, NIAID, National Institutes of Health, Rockville, Maryland 20852 USA
| | - John F Andersen
- The Laboratory of Malaria and Vector Research, NIAID, National Institutes of Health, Rockville, Maryland 20852 USA
| |
Collapse
|
33
|
Housley DM, Housley GD, Liddell MJ, Jennings EA. Scorpion toxin peptide action at the ion channel subunit level. Neuropharmacology 2016; 127:46-78. [PMID: 27729239 DOI: 10.1016/j.neuropharm.2016.10.004] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2016] [Revised: 09/06/2016] [Accepted: 10/06/2016] [Indexed: 12/19/2022]
Abstract
This review categorizes functionally validated actions of defined scorpion toxin (SCTX) neuropeptides across ion channel subclasses, highlighting key trends in this rapidly evolving field. Scorpion envenomation is a common event in many tropical and subtropical countries, with neuropharmacological actions, particularly autonomic nervous system modulation, causing significant mortality. The primary active agents within scorpion venoms are a diverse group of small neuropeptides that elicit specific potent actions across a wide range of ion channel classes. The identification and functional characterisation of these SCTX peptides has tremendous potential for development of novel pharmaceuticals that advance knowledge of ion channels and establish lead compounds for treatment of excitable tissue disorders. This review delineates the unique specificities of 320 individual SCTX peptides that collectively act on 41 ion channel subclasses. Thus the SCTX research field has significant translational implications for pathophysiology spanning neurotransmission, neurohumoral signalling, sensori-motor systems and excitation-contraction coupling. This article is part of the Special Issue entitled 'Venom-derived Peptides as Pharmacological Tools.'
Collapse
Affiliation(s)
- David M Housley
- College of Medicine and Dentistry, Cairns Campus, James Cook University, Cairns, Queensland 4878, Australia; Translational Neuroscience Facility and Department of Physiology, School of Medical Sciences, UNSW Australia, Sydney, NSW 2052, Australia.
| | - Gary D Housley
- Translational Neuroscience Facility and Department of Physiology, School of Medical Sciences, UNSW Australia, Sydney, NSW 2052, Australia
| | - Michael J Liddell
- Centre for Tropical Environmental and Sustainability Science and College of Science & Engineering, Cairns Campus, James Cook University, Cairns, Queensland 4878, Australia
| | - Ernest A Jennings
- College of Medicine and Dentistry, Cairns Campus, James Cook University, Cairns, Queensland 4878, Australia; Centre for Biodiscovery and Molecular Development of Therapeutics, James Cook University, Queensland 4878, Australia; Australian Institute of Tropical Health and Medicine, James Cook University, Cairns Campus, QLD, Australia
| |
Collapse
|
34
|
Munasinghe NR, Christie MJ. Conotoxins That Could Provide Analgesia through Voltage Gated Sodium Channel Inhibition. Toxins (Basel) 2015; 7:5386-407. [PMID: 26690478 PMCID: PMC4690140 DOI: 10.3390/toxins7124890] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2015] [Revised: 10/23/2015] [Accepted: 11/19/2015] [Indexed: 12/19/2022] Open
Abstract
Chronic pain creates a large socio-economic burden around the world. It is physically and mentally debilitating, and many sufferers are unresponsive to current therapeutics. Many drugs that provide pain relief have adverse side effects and addiction liabilities. Therefore, a great need has risen for alternative treatment strategies. One rich source of potential analgesic compounds that has emerged over the past few decades are conotoxins. These toxins are extremely diverse and display selective activity at ion channels. Voltage gated sodium (NaV) channels are one such group of ion channels that play a significant role in multiple pain pathways. This review will explore the literature around conotoxins that bind NaV channels and determine their analgesic potential.
Collapse
Affiliation(s)
- Nehan R Munasinghe
- Discipline of Pharmacology, The University of Sydney, Sydney, NSW 2006, Australia.
| | - MacDonald J Christie
- Discipline of Pharmacology, The University of Sydney, Sydney, NSW 2006, Australia.
| |
Collapse
|
35
|
Motin L, Durek T, Adams DJ. Modulation of human Nav1.7 channel gating by synthetic α-scorpion toxin OD1 and its analogs. Channels (Austin) 2015; 10:139-47. [PMID: 26646206 DOI: 10.1080/19336950.2015.1120392] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Nine different voltage-gated sodium channel isoforms are responsible for inducing and propagating action potentials in the mammalian nervous system. The Nav1.7 channel isoform plays an important role in conducting nociceptive signals. Specific mutations of this isoform may impair gating behavior of the channel resulting in several pain syndromes. In addition to channel mutations, similar or opposite changes in gating may be produced by spider and scorpion toxins binding to different parts of the voltage-gated sodium channel. In the present study, we analyzed the effects of the α-scorpion toxin OD1 and 2 synthetic toxin analogs on the gating properties of the Nav1.7 sodium channel. All toxins potently inhibited channel inactivation, however, both toxin analogs showed substantially increased potency by more than one order of magnitude when compared with that of wild-type OD1. The decay phase of the whole-cell Na(+) current was substantially slower in the presence of toxins than in their absence. Single-channel recordings in the presence of the toxins revealed that Na(+) current inactivation slowed due to prolonged flickering of the channel between open and closed states. Our findings support the voltage-sensor trapping model of α-scorpion toxin action, in which the toxin prevents a conformational change in the domain IV voltage sensor that normally leads to fast channel inactivation.
Collapse
Affiliation(s)
- Leonid Motin
- a Health Innovations Research Institute, RMIT University , Melbourne , Victoria, Australia
| | - Thomas Durek
- b Division of Chemistry & Structural Biology, Institute for Molecular Bioscience, The University of Queensland, Brisbane , Queensland, Australia
| | - David J Adams
- a Health Innovations Research Institute, RMIT University , Melbourne , Victoria, Australia
| |
Collapse
|
36
|
Computational Studies of Venom Peptides Targeting Potassium Channels. Toxins (Basel) 2015; 7:5194-211. [PMID: 26633507 PMCID: PMC4690127 DOI: 10.3390/toxins7124877] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2015] [Revised: 11/13/2015] [Accepted: 11/20/2015] [Indexed: 01/18/2023] Open
Abstract
Small peptides isolated from the venom of animals are potential scaffolds for ion channel drug discovery. This review article mainly focuses on the computational studies that have advanced our understanding of how various toxins interfere with the function of K+ channels. We introduce the computational tools available for the study of toxin-channel interactions. We then discuss how these computational tools have been fruitfully applied to elucidate the mechanisms of action of a wide range of venom peptides from scorpions, spiders, and sea anemone.
Collapse
|
37
|
Rodríguez-Ravelo R, Batista CV, Coronas FI, Zamudio FZ, Hernández-Orihuela L, Espinosa-López G, Ruiz-Urquiola A, Possani LD. Comparative proteomic analysis of male and female venoms from the Cuban scorpion Rhopalurus junceus. Toxicon 2015; 107:327-34. [DOI: 10.1016/j.toxicon.2015.06.026] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2015] [Revised: 06/24/2015] [Accepted: 06/30/2015] [Indexed: 10/23/2022]
|
38
|
Riaño-Umbarila L, Ledezma-Candanoza LM, Serrano-Posada H, Fernández-Taboada G, Olamendi-Portugal T, Rojas-Trejo S, Gómez-Ramírez IV, Rudiño-Piñera E, Possani LD, Becerril B. Optimal Neutralization of Centruroides noxius Venom Is Understood through a Structural Complex between Two Antibody Fragments and the Cn2 Toxin. J Biol Chem 2015; 291:1619-1630. [PMID: 26589800 DOI: 10.1074/jbc.m115.685297] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2015] [Indexed: 12/19/2022] Open
Abstract
The current trend of using recombinant antibody fragments in research to develop novel antidotes against scorpion stings has achieved excellent results. The polyclonal character of commercial antivenoms, obtained through the immunization of animals and which contain several neutralizing antibodies that recognize different epitopes on the toxins, guarantees the neutralization of the venoms. To avoid the use of animals, we aimed to develop an equivalent recombinant antivenom composed of a few neutralizing single chain antibody fragments (scFvs) that bind to two different epitopes on the scorpion toxins. In this study, we obtained scFv RU1 derived from scFv C1. RU1 showed a good capacity to neutralize the Cn2 toxin and whole venom of the scorpion Centruroides noxius. Previously, we had produced scFv LR, obtained from a different parental fragment (scFv 3F). LR also showed a similar neutralizing capacity. The simultaneous administration of both scFvs resulted in improved protection, which was translated as a rapid recovery of previously poisoned animals. The crystallographic structure of the ternary complex scFv LR-Cn2-scFv RU1 allowed us to identify the areas of interaction of both scFvs with the toxin, which correspond to non-overlapping sites. The epitope recognized by scFv RU1 seems to be related to a greater efficiency in the neutralization of the whole venom. In addition, the structural analysis of the complex helped us to explain the cross-reactivity of these scFvs and how they neutralize the venom.
Collapse
Affiliation(s)
- Lidia Riaño-Umbarila
- From the Departamento de Medicina Molecular y Bioprocesos, Instituto de Biotecnología, Universidad Nacional Autónoma de México, Apartado Postal 510-3, Cuernavaca, Morelos 62250, México and
| | - Luis M Ledezma-Candanoza
- From the Departamento de Medicina Molecular y Bioprocesos, Instituto de Biotecnología, Universidad Nacional Autónoma de México, Apartado Postal 510-3, Cuernavaca, Morelos 62250, México and
| | - Hugo Serrano-Posada
- the Laboratorio de Bioingeniería, Universidad de Colima, Km. 9 carretera Coquimatlán-Colima, C.P. 28400 Coquimatlán, Colima, México
| | - Guillermo Fernández-Taboada
- From the Departamento de Medicina Molecular y Bioprocesos, Instituto de Biotecnología, Universidad Nacional Autónoma de México, Apartado Postal 510-3, Cuernavaca, Morelos 62250, México and
| | - Timoteo Olamendi-Portugal
- From the Departamento de Medicina Molecular y Bioprocesos, Instituto de Biotecnología, Universidad Nacional Autónoma de México, Apartado Postal 510-3, Cuernavaca, Morelos 62250, México and
| | - Sonia Rojas-Trejo
- From the Departamento de Medicina Molecular y Bioprocesos, Instituto de Biotecnología, Universidad Nacional Autónoma de México, Apartado Postal 510-3, Cuernavaca, Morelos 62250, México and
| | - Ilse V Gómez-Ramírez
- From the Departamento de Medicina Molecular y Bioprocesos, Instituto de Biotecnología, Universidad Nacional Autónoma de México, Apartado Postal 510-3, Cuernavaca, Morelos 62250, México and
| | - Enrique Rudiño-Piñera
- From the Departamento de Medicina Molecular y Bioprocesos, Instituto de Biotecnología, Universidad Nacional Autónoma de México, Apartado Postal 510-3, Cuernavaca, Morelos 62250, México and
| | - Lourival D Possani
- From the Departamento de Medicina Molecular y Bioprocesos, Instituto de Biotecnología, Universidad Nacional Autónoma de México, Apartado Postal 510-3, Cuernavaca, Morelos 62250, México and
| | - Baltazar Becerril
- From the Departamento de Medicina Molecular y Bioprocesos, Instituto de Biotecnología, Universidad Nacional Autónoma de México, Apartado Postal 510-3, Cuernavaca, Morelos 62250, México and.
| |
Collapse
|
39
|
Wang L, Zellmer SG, Printzenhoff DM, Castle NA. Addition of a single methyl group to a small molecule sodium channel inhibitor introduces a new mode of gating modulation. Br J Pharmacol 2015. [PMID: 26220736 DOI: 10.1111/bph.13259] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
Abstract
BACKGROUND AND PURPOSE Aryl sulfonamide Nav 1.3 or Nav 1.7 voltage-gated sodium (Nav ) channel inhibitors interact with the Domain 4 voltage sensor domain (D4 VSD). During studies to better understand the structure-activity relationship of this interaction, an additional mode of channel modulation, specifically slowing of inactivation, was revealed by addition of a single methyl moiety. The objective of the current study was to determine if these different modulatory effects are mediated by the same or distinct interactions with the channel. EXPERIMENTAL APPROACH Electrophysiology and site-directed mutation were used to compare the effects of PF-06526290 and its desmethyl analogue PF-05661014 on Nav channel function. KEY RESULTS PF-05661014 selectively inhibits Nav 1.3 versus Nav 1.7 currents by stabilizing inactivated channels via interaction with D4 VSD. In contrast, PF-06526290, which differs from PF-05661014 by a single methyl group, exhibits a dual effect. It greatly slows inactivation of Nav channels in a subtype-independent manner. However, upon prolonged depolarization to induce inactivation, PF-06526290 becomes a Nav subtype selective inhibitor similar to PF-05661014. Mutation of the D4 VSD modulates inhibition of Nav 1.3 or Nav 1.7 by both PF-05661014 and PF-06526290, but has no effect on the inactivation slowing produced by PF-06526290. This finding, along with the absence of functional inhibition of PF-06526290-induced inactivation slowing by PF-05661014, suggests that distinct interactions underlie the two modes of Nav channel modulation. CONCLUSIONS AND IMPLICATIONS Addition of a methyl group to a Nav channel inhibitor introduces an additional mode of gating modulation, implying that a single compound can affect sodium channel function in multiple ways.
Collapse
|
40
|
Abstract
It is long known that peptide neurotoxins derived from a diversity of venomous animals evolve by positive selection following gene duplication, yet a force that drives their adaptive evolution remains a mystery. By using maximum-likelihood models of codon substitution, we analyzed molecular adaptation in scorpion sodium channel toxins from a specific species and found ten positively selected sites, six of which are located at the core-domain of scorpion α-toxins, a region known to interact with two adjacent loops in the voltage-sensor domain (DIV) of sodium channels, as validated by our newly constructed computational model of toxin-channel complex. Despite the lack of positive selection signals in these two loops, they accumulated extensive sequence variations by relaxed purifying selection in prey and predators of scorpions. The evolutionary variability in the toxin-bound regions of sodium channels indicates that accelerated substitutions in the multigene family of scorpion toxins is a consequence of dealing with the target diversity. This work presents an example of atypical co-evolution between animal toxins and their molecular targets, in which toxins suffered from more prominent selective pressure from the channels of their competitors. Our discovery helps explain the evolutionary rationality of gene duplication of toxins in a specific venomous species.
Collapse
|
41
|
Affiliation(s)
- William A Catterall
- Department of Pharmacology, University of Washington, Seattle, Washington 98195-7280.
| |
Collapse
|
42
|
Coronas FIV, Diego-García E, Restano-Cassulini R, de Roodt AR, Possani LD. Biochemical and physiological characterization of a new Na(+)-channel specific peptide from the venom of the Argentinean scorpion Tityus trivittatus. Peptides 2015; 68:11-6. [PMID: 24862827 DOI: 10.1016/j.peptides.2014.05.002] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/10/2014] [Revised: 05/13/2014] [Accepted: 05/14/2014] [Indexed: 01/12/2023]
Abstract
A new peptide with 61 amino acids cross-linked by 4 disulfide bridges, with molecular weight of 6938.12Da, and an amidated C-terminal amino acid residue was purified and characterized. The primary structure was obtained by direct Edman degradation and sequencing its gene. The peptide is lethal to mammals and was shown to be similar (95% identity) to toxin Ts1 (gamma toxin) from the Brazilian scorpion Tityus serrulatus; it was named Tt1g (from T. trivittatus toxin 1 gamma-like). Tt1g was assayed on several sub-types of Na(+)-channels showing displacement of the currents to more negative voltages, being the hNav1.3 the most affected channel. This toxin displays characteristics typical to the β-type sodium scorpion toxins. Lethality tests and physiological assays indicate that this peptide is probably the most important toxic component of this species of scorpion, known for causing human fatalities in the South American continent.
Collapse
Affiliation(s)
- Fredy I V Coronas
- Departamento de Medicina Molecular y Bioprocesos, Instituto de Biotecnología, Universidad Nacional Autónoma de México, Mexico
| | - Elia Diego-García
- Departamento de Medicina Molecular y Bioprocesos, Instituto de Biotecnología, Universidad Nacional Autónoma de México, Mexico
| | - Rita Restano-Cassulini
- Departamento de Medicina Molecular y Bioprocesos, Instituto de Biotecnología, Universidad Nacional Autónoma de México, Mexico
| | - Adolfo R de Roodt
- Laboratorio de Toxinopatología, Centro de Patología Experimental y Aplicada, Facultad de Medicina, Universidad de Buenos Aires and Ministerio de Salud de la Nación, Argentina.
| | - Lourival D Possani
- Departamento de Medicina Molecular y Bioprocesos, Instituto de Biotecnología, Universidad Nacional Autónoma de México, Mexico.
| |
Collapse
|
43
|
Meng X, Xu Y, Zhao M, Wang F, Ma Y, Jin Y, Liu Y, Song Y, Zhang J. The Functional Property Changes of Muscular Nav1.4 and Cardiac Nav1.5 Induced by Scorpion Toxin BmK AGP-SYPU1 Mutants Y42F and Y5F. Biochemistry 2015; 54:2988-96. [DOI: 10.1021/acs.biochem.5b00067] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Affiliation(s)
- Xiangxue Meng
- School of Life Sciences & Biopharmaceutical Science, Shenyang Pharmaceutical University, 103 Wenhua Road, 110016 Shenyang, PR China
- Benxi
Medicine Institute, Shenyang Pharmaceutical University, Shiqiaozi, 177005 Benxi, PR China
| | - Yijia Xu
- School of Life Sciences & Biopharmaceutical Science, Shenyang Pharmaceutical University, 103 Wenhua Road, 110016 Shenyang, PR China
| | - Mingyi Zhao
- School of Life Sciences & Biopharmaceutical Science, Shenyang Pharmaceutical University, 103 Wenhua Road, 110016 Shenyang, PR China
| | - Fangyang Wang
- School of Life Sciences & Biopharmaceutical Science, Shenyang Pharmaceutical University, 103 Wenhua Road, 110016 Shenyang, PR China
| | - Yuanyuan Ma
- School of Life Sciences & Biopharmaceutical Science, Shenyang Pharmaceutical University, 103 Wenhua Road, 110016 Shenyang, PR China
| | - Yao Jin
- School of Life Sciences & Biopharmaceutical Science, Shenyang Pharmaceutical University, 103 Wenhua Road, 110016 Shenyang, PR China
| | - Yanfeng Liu
- School of Life Sciences & Biopharmaceutical Science, Shenyang Pharmaceutical University, 103 Wenhua Road, 110016 Shenyang, PR China
| | - Yongbo Song
- School of Life Sciences & Biopharmaceutical Science, Shenyang Pharmaceutical University, 103 Wenhua Road, 110016 Shenyang, PR China
| | - Jinghai Zhang
- School of Life Sciences & Biopharmaceutical Science, Shenyang Pharmaceutical University, 103 Wenhua Road, 110016 Shenyang, PR China
- School
of Medical Devices, Shenyang Pharmaceutical University, 103 Wenhua
Road, 110016 Shenyang, PR China
- Benxi
Medicine Institute, Shenyang Pharmaceutical University, Shiqiaozi, 177005 Benxi, PR China
| |
Collapse
|
44
|
The transcriptome recipe for the venom cocktail of Tityus bahiensis scorpion. Toxicon 2015; 95:52-61. [DOI: 10.1016/j.toxicon.2014.12.013] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2014] [Revised: 12/13/2014] [Accepted: 12/27/2014] [Indexed: 12/23/2022]
|
45
|
Escalona MP, Batista CV, Cassulini RR, Rios MS, Coronas FI, Possani LD. A proteomic analysis of the early secondary molecular effects caused by Cn2 scorpion toxin on neuroblastoma cells. J Proteomics 2014; 111:212-23. [DOI: 10.1016/j.jprot.2014.04.035] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2014] [Revised: 04/20/2014] [Accepted: 04/22/2014] [Indexed: 01/16/2023]
|
46
|
Tang C, Zhou X, Huang Y, Zhang Y, Hu Z, Wang M, Chen P, Liu Z, Liang S. The tarantula toxin jingzhaotoxin-XI (κ-theraphotoxin-Cj1a) regulates the activation and inactivation of the voltage-gated sodium channel Nav1.5. Toxicon 2014; 92:6-13. [DOI: 10.1016/j.toxicon.2014.09.002] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2014] [Revised: 09/01/2014] [Accepted: 09/09/2014] [Indexed: 01/28/2023]
|
47
|
Xu L, Li T, Liu H, Yang F, Liang S, Cao Z, Li W, Wu Y. Functional characterization of two novel scorpion sodium channel toxins from Lychas mucronatus. Toxicon 2014; 90:318-25. [DOI: 10.1016/j.toxicon.2014.08.075] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2014] [Revised: 08/11/2014] [Accepted: 08/27/2014] [Indexed: 10/24/2022]
|
48
|
Dong K, Du Y, Rinkevich F, Nomura Y, Xu P, Wang L, Silver K, Zhorov BS. Molecular biology of insect sodium channels and pyrethroid resistance. INSECT BIOCHEMISTRY AND MOLECULAR BIOLOGY 2014; 50:1-17. [PMID: 24704279 PMCID: PMC4484874 DOI: 10.1016/j.ibmb.2014.03.012] [Citation(s) in RCA: 312] [Impact Index Per Article: 28.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/20/2014] [Revised: 03/24/2014] [Accepted: 03/24/2014] [Indexed: 05/06/2023]
Abstract
Voltage-gated sodium channels are essential for the initiation and propagation of the action potential in neurons and other excitable cells. Because of their critical roles in electrical signaling, sodium channels are targets of a variety of naturally occurring and synthetic neurotoxins, including several classes of insecticides. This review is intended to provide an update on the molecular biology of insect sodium channels and the molecular mechanism of pyrethroid resistance. Although mammalian and insect sodium channels share fundamental topological and functional properties, most insect species carry only one sodium channel gene, compared to multiple sodium channel genes found in each mammalian species. Recent studies showed that two posttranscriptional mechanisms, alternative splicing and RNA editing, are involved in generating functional diversity of sodium channels in insects. More than 50 sodium channel mutations have been identified to be responsible for or associated with knockdown resistance (kdr) to pyrethroids in various arthropod pests and disease vectors. Elucidation of molecular mechanism of kdr led to the identification of dual receptor sites of pyrethroids on insect sodium channels. Many of the kdr mutations appear to be located within or close to the two receptor sites. The accumulating knowledge of insect sodium channels and their interactions with insecticides provides a foundation for understanding the neurophysiology of sodium channels in vivo and the development of new and safer insecticides for effective control of arthropod pests and human disease vectors.
Collapse
Affiliation(s)
- Ke Dong
- Department of Entomology, Neuroscience and Genetics Programs, Michigan State University, East Lansing, MI, USA.
| | - Yuzhe Du
- Department of Entomology, Neuroscience and Genetics Programs, Michigan State University, East Lansing, MI, USA
| | - Frank Rinkevich
- Department of Entomology, Neuroscience and Genetics Programs, Michigan State University, East Lansing, MI, USA
| | - Yoshiko Nomura
- Department of Entomology, Neuroscience and Genetics Programs, Michigan State University, East Lansing, MI, USA
| | - Peng Xu
- Department of Entomology, Neuroscience and Genetics Programs, Michigan State University, East Lansing, MI, USA
| | - Lingxin Wang
- Department of Entomology, Neuroscience and Genetics Programs, Michigan State University, East Lansing, MI, USA
| | - Kristopher Silver
- Department of Anatomy and Physiology, Kansas State University, Manhattan, KS, USA
| | - Boris S Zhorov
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Ontario, Canada; Sechenov Institute of Evolutionary Physiology & Biochemistry, Russian Academy of Sciences, St. Petersburg, Russia
| |
Collapse
|
49
|
Bladen C, Hamid J, Souza IA, Zamponi GW. Block of T-type calcium channels by protoxins I and II. Mol Brain 2014; 7:36. [PMID: 24886690 PMCID: PMC4030284 DOI: 10.1186/1756-6606-7-36] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2014] [Accepted: 05/05/2014] [Indexed: 01/08/2023] Open
Abstract
Background Low-voltage-activated (T-type) calcium channels play a crucial role in a number of physiological processes, including neuronal and cardiac pacemaker activity and nociception. Therefore, finding specific modulators and/or blockers of T-type channels has become an important field of drug discovery. One characteristic of T-type calcium channels is that they share several structural similarities with voltage-gated sodium channels (VGSCs). We therefore hypothesized that binding sites for certain sodium channel blocking peptide toxins may be present in T-type calcium channels. Findings The sodium channel blocker ProTx I tonically blocked native and transiently expressed T-type channels in the sub- to low micro molar range with at least a ten-fold selectivity for the T-type calcium channel hCav3.1 over hCav3.3, and more than one hundred fold selectivity over hCav3.2. Using chimeras of hCav3.1 and hCav3.3, we determined that the domain IV region of hCav3.1 is a major determinant of toxin affinity, with a minor contribution from domain II. Further analysis revealed several residues in a highly conserved region between T-type and sodium channels that may correspond to toxin binding sites. Mutagenesis of several of these residues on an individual basis, however, did not alter the blocking effects of the toxin. ProTx II on the other hand preferentially blocked hCav3.2 and significantly shifted the steady state inactivation of this channel. Conclusions ProTx I blocks hCav3.1 both selectively and with high affinity. Domain IV appears to play a major role in this selectivity with some contribution from domain II. Given the structural similarities between sodium and T-type calcium channels and the apparent conservation in toxin binding sites, these data could provide insights into the development and synthesis of novel T-type channel antagonists.
Collapse
Affiliation(s)
| | | | | | - Gerald W Zamponi
- Department of Physiology & Pharmacology, Hotchkiss Brain Institute, University of Calgary, 3330 Hospital Drive, Calgary, NW, Canada.
| |
Collapse
|
50
|
Rodríguez-Ravelo R, Restano-Cassulini R, Zamudio FZ, Coronas FIV, Espinosa-López G, Possani LD. A K⁺ channel blocking peptide from the Cuban scorpion Rhopalurus garridoi. Peptides 2014; 53:42-7. [PMID: 24512947 DOI: 10.1016/j.peptides.2013.10.010] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/06/2013] [Revised: 10/15/2013] [Accepted: 10/15/2013] [Indexed: 11/25/2022]
Abstract
A proteomic analysis of the venom obtained from the Cuban scorpion Rhopalurus garridoi was performed. Venom was obtained by electrical stimulation, separated by high performance liquid chromatography, and the molecular masses of their 50 protein components were identified by mass spectrometry. A peptide of 3940 Da molecular mass was obtained in pure form and its primary structure determined. It contains 37 amino acid residues, including three disulfide bridges. Electrophysiological experiments showed that this peptide is capable of blocking reversibly K(+)-channels hKv1.1 with a Kd close to 1 μM, but is not effective against hKv1.4, hERG1 and EAG currents, at the same concentration. This is the first protein component ever isolated from this species of scorpion and was assigned the systematic number α-KTx 2.14.
Collapse
Affiliation(s)
- Rodolfo Rodríguez-Ravelo
- Center for Mountain Development, Ministry of Science, Technology and, Environment, Limonar de Monte Roux, El Salvador Guantánamo, Cuba.
| | - Rita Restano-Cassulini
- Department of Molecular Medicine and Bioprocesses, Biotechnology, Institute, National Autonomous University of Mexico, Avenida Universidad 2001, Colonia Chamilpa, Apartado Postal 510-3, Cuernavaca, Morelos 62210, Mexico.
| | - Fernando Z Zamudio
- Department of Molecular Medicine and Bioprocesses, Biotechnology, Institute, National Autonomous University of Mexico, Avenida Universidad 2001, Colonia Chamilpa, Apartado Postal 510-3, Cuernavaca, Morelos 62210, Mexico.
| | - Fredy I V Coronas
- Department of Molecular Medicine and Bioprocesses, Biotechnology, Institute, National Autonomous University of Mexico, Avenida Universidad 2001, Colonia Chamilpa, Apartado Postal 510-3, Cuernavaca, Morelos 62210, Mexico.
| | | | - Lourival D Possani
- Department of Molecular Medicine and Bioprocesses, Biotechnology, Institute, National Autonomous University of Mexico, Avenida Universidad 2001, Colonia Chamilpa, Apartado Postal 510-3, Cuernavaca, Morelos 62210, Mexico.
| |
Collapse
|