1
|
Ding SM, Yap MKK. Deciphering toxico-proteomics of Asiatic medically significant venomous snake species: A systematic review and interactive data dashboard. Toxicon 2024; 250:108120. [PMID: 39393539 DOI: 10.1016/j.toxicon.2024.108120] [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: 08/07/2024] [Revised: 09/30/2024] [Accepted: 10/06/2024] [Indexed: 10/13/2024]
Abstract
Snakebite envenomation (SBE) is a neglected tropical disease (NTD) with an approximate 1.8 million cases annually. The tremendous figure is concerning, and the currently available treatment for snakebite envenomation is antivenom. However, the current antivenom has limited cross-neutralisation activity due to the variations in snake venom composition across species and geographical locations. The proteomics of medically important venomous species is essential as they study the venom compositions within and among different species. The advancement of sophisticated proteomic approaches allows intensive investigation of snake venoms. Nevertheless, there is a need to consolidate the venom proteomics profiles and distribution analysis to examine their variability patterns. This review systematically analysed the proteomics and toxicity profiles of medically important venomous species from Asia across different geographical locations. An interactive dashboard - Asiatic Proteomics Interactive Datasets was curated to consolidate the distribution patterns of the venom compositions, serve as a comprehensive directory for large-scale comparative meta-analyses. The population proteomics demonstrate higher diversities in the predominant venom toxins. Besides, inter-regional differences were also observed in Bungarus sp., Naja sp., Calliophis sp., and Ophiophagus hannah venoms. The elapid venoms are predominated with three-finger toxins (3FTXs) and phospholipase A2 (PLA2). Intra-regional variation is only significantly observed in Naja naja venoms. Proteomics diversity is more prominent in viper venoms, with widespread dominance observed in snake venom metalloproteinase (SVMP) and snake venom serine protease (SVSP). Correlations exist between the proteomics profiles and the toxicity (LD50) of the medically important venomous species. Additionally, the predominant toxins, alongside their pathophysiological effects, were highlighted and discussed as well. The insights of interactive toxico-proteomics datasets provide comprehensive frameworks of venom dynamics and contribute to developing antivenoms for snakebite envenomation. This could reduce misdiagnosis of SBE and accelerate the researchers' data mining process.
Collapse
Affiliation(s)
- Sher Min Ding
- School of Science, Monash University Malaysia, Bandar Sunway, Malaysia
| | | |
Collapse
|
2
|
Sukumaran S, Prasanna VM, Panicker LK, Nair AS, Oommen OV. Discovery of a new Daboia russelli viper venom PLA 2 inhibitor using virtual screening of pharmacophoric features of co-crystallized compound. J Biomol Struct Dyn 2024; 42:6954-6967. [PMID: 37490072 DOI: 10.1080/07391102.2023.2238072] [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: 12/23/2022] [Accepted: 07/11/2023] [Indexed: 07/26/2023]
Abstract
Snake venom PLA2, a member of the group of hydrolase enzymes, has been recognized as a promising drug target for snake envenomation. In the present study, an attempt was made to identify potential inhibitors of snake venom PLA2 by employing a pharmacophore-based virtual screening, docking, and dynamics approach. A receptor-based pharmacophore model was generated based on the features of the established and bound co-crystal ligand (2-carbamoylmethyl-5-propyl-octahydro-indol-7-yl)-acetic acid in the PLA2 complex. The best pharmacophore model (ADDH) derived, consisted of four features, namely one hydrogen bond acceptor, two hydrogen bond donors, and one hydrophobic region. This common pharmacophore was then used to perform virtual screening against a drug-like diverse database, with due consideration to the Lipinski 'rule of five', so as to obtain a pool of lead molecules. The short-listed lead molecules were then subjected to docking analysis with that of the Daboia russelli viper venom PLA2 followed by a molecular simulation study for a duration of 100 ns. CAP04815700 was chosen as the best compound based on the simulation parameters, which were then taken for MM/PBSA calculation, and it was revealed that it has a similar effective inhibitory potential as that of the crystal ligand. Further, the cluster analysis also revealed the structural significance of the backbone protein after the interaction with CAP04815700. This study will continue to explore its bioactivity in vitro and in vivo.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Suveena Sukumaran
- Centre for venom informatics, Department of Computational Biology and Bioinformatics, University of Kerala, Trivandrum, Kerala, India
- Department of Computational Biology & Bioinformatics, University of Kerala, Trivandrum, Kerala, India
| | - Vinod Manoharan Prasanna
- Centre for venom informatics, Department of Computational Biology and Bioinformatics, University of Kerala, Trivandrum, Kerala, India
| | - Laladhas Krishna Panicker
- Centre for venom informatics, Department of Computational Biology and Bioinformatics, University of Kerala, Trivandrum, Kerala, India
- Department of Computational Biology & Bioinformatics, University of Kerala, Trivandrum, Kerala, India
| | - Achuthsankar S Nair
- Centre for venom informatics, Department of Computational Biology and Bioinformatics, University of Kerala, Trivandrum, Kerala, India
| | - Oommen V Oommen
- Centre for venom informatics, Department of Computational Biology and Bioinformatics, University of Kerala, Trivandrum, Kerala, India
- Department of Computational Biology & Bioinformatics, University of Kerala, Trivandrum, Kerala, India
| |
Collapse
|
3
|
López-Dávila AJ, Lomonte B, Gutiérrez JM. Alterations of the skeletal muscle contractile apparatus in necrosis induced by myotoxic snake venom phospholipases A 2: a mini-review. J Muscle Res Cell Motil 2024; 45:69-77. [PMID: 38063951 PMCID: PMC11096208 DOI: 10.1007/s10974-023-09662-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2023] [Accepted: 11/07/2023] [Indexed: 05/16/2024]
Abstract
Skeletal muscle necrosis is a common clinical manifestation of snakebite envenoming. The predominant myotoxic components in snake venoms are catalytically-active phospholipases A2 (PLA2) and PLA2 homologs devoid of enzymatic activity, which have been used as models to investigate various aspects of muscle degeneration. This review addresses the changes in the contractile apparatus of skeletal muscle induced by these toxins. Myotoxic components initially disrupt the integrity of sarcolemma, generating a calcium influx that causes various degenerative events, including hypercontraction of myofilaments. There is removal of specific sarcomeric proteins, owing to the hydrolytic action of muscle calpains and proteinases from invading inflammatory cells, causing an initial redistribution followed by widespread degradation of myofibrillar material. Experiments using skinned cardiomyocytes and skeletal muscle fibers show that these myotoxins do not directly affect the contractile apparatus, implying that hypercontraction is due to cytosolic calcium increase secondary to sarcolemmal damage. Such drastic hypercontraction may contribute to muscle damage by generating mechanical stress and further sarcolemmal damage.
Collapse
Affiliation(s)
- Alfredo Jesús López-Dávila
- Institute of Molecular and Cell Physiology, Hannover Medical School, Carl-Neuberg-Str. 1, 30625, Hannover, Germany.
| | - Bruno Lomonte
- Instituto Clodomiro Picado, Facultad de Microbiología, Universidad de Costa Rica, San José, 11501, Costa Rica
| | - José María Gutiérrez
- Instituto Clodomiro Picado, Facultad de Microbiología, Universidad de Costa Rica, San José, 11501, Costa Rica
| |
Collapse
|
4
|
Santos LC, Oliveira VQ, Teixeira SC, Correia TML, Andrade LOSB, Polloni L, Marques LM, Clissa PB, Baldo C, Ferro EAV, Gusmão ACMDM, Silva MJB, Sanabani SS, Ávila VDMR, Lopes DS. PLA 2-MjTX-II from Bothrops moojeni snake venom exhibits antimetastatic and antiangiogenic effects on human lung cancer cells. Toxicon 2024; 243:107742. [PMID: 38705486 DOI: 10.1016/j.toxicon.2024.107742] [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: 02/07/2024] [Revised: 04/29/2024] [Accepted: 04/30/2024] [Indexed: 05/07/2024]
Abstract
Phospholipases A2 (PLA2s) from snake venom possess antitumor and antiangiogenic properties. In this study, we evaluated the antimetastatic and antiangiogenic effects of MjTX-II, a Lys49 PLA2 isolated from Bothrops moojeni venom, on lung cancer and endothelial cells. Using in vitro and ex vivo approaches, we demonstrated that MjTX-II reduced cell proliferation and inhibited fundamental processes for lung cancer cells (A549) growth and metastasis, such as adhesion, migration, invasion, and actin cytoskeleton decrease, without significantly interfering with non-tumorigenic lung cells (BEAS-2B). Furthermore, MjTX-II caused cell cycle alterations, increased reactive oxygen species production, modulated the expression of pro- and antiangiogenic genes, and decreased vascular endothelial growth factor (VEGF) expression in HUVECs. Finally, MjTX-II inhibited ex vivo angiogenesis processes in an aortic ring model. Therefore, we conclude that MjTX-II exhibits antimetastatic and antiangiogenic effects in vitro and ex vivo and represents a molecule that hold promise as a pharmacological model for antitumor therapy.
Collapse
Affiliation(s)
- Luísa Carregosa Santos
- Institute Multidisciplinary in Health, Federal University of Bahia (UFBA), Vitória da Conquista, BA, Brazil
| | - Vinícius Queiroz Oliveira
- Institute Multidisciplinary in Health, Federal University of Bahia (UFBA), Vitória da Conquista, BA, Brazil
| | - Samuel Cota Teixeira
- Department of Immunology, Institute of Biomedical Sciences, Federal University of Uberlândia (UFU), Uberlândia, MG, Brazil
| | | | | | - Lorena Polloni
- Laboratory of Biochemistry and Animal Toxins, Institute of Biotechnology, Federal University of Uberlândia, (UFU), Uberlândia, MG, Brazil
| | - Lucas Miranda Marques
- Institute Multidisciplinary in Health, Federal University of Bahia (UFBA), Vitória da Conquista, BA, Brazil
| | | | - Cristiani Baldo
- Department of Biochemistry and Biotechnology, State University of Londrina (UEL), Londrina, PR, Brazil
| | - Eloisa Amália Vieira Ferro
- Department of Immunology, Institute of Biomedical Sciences, Federal University of Uberlândia (UFU), Uberlândia, MG, Brazil
| | | | - Marcelo José Barbosa Silva
- Department of Immunology, Institute of Biomedical Sciences, Federal University of Uberlândia (UFU), Uberlândia, MG, Brazil
| | - Sabri Saeed Sanabani
- Laboratory of Medical Investigation in Dermatology and Immunodeficiency, São Paulo Institute of Tropical Medicine, School of Medicine, University of São Paulo (USP), São Paulo, SP, Brazil
| | - Veridiana de Melo Rodrigues Ávila
- Laboratory of Biochemistry and Animal Toxins, Institute of Biotechnology, Federal University of Uberlândia, (UFU), Uberlândia, MG, Brazil.
| | - Daiana Silva Lopes
- Institute Multidisciplinary in Health, Federal University of Bahia (UFBA), Vitória da Conquista, BA, Brazil.
| |
Collapse
|
5
|
Zhao W, Liu J, Wang S, Tao Q, Lei Q, Huang C. Varespladib mitigates acute liver injury via suppression of excessive mitophagy on Naja atra envenomed mice by inhibiting PLA 2. Toxicon 2024; 242:107694. [PMID: 38556061 DOI: 10.1016/j.toxicon.2024.107694] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2024] [Revised: 03/05/2024] [Accepted: 03/19/2024] [Indexed: 04/02/2024]
Abstract
Snakebite envenomation often leads to severe visceral injuries, including acute liver injury (ALI). However, the toxicity mechanism remains unclear. Moreover, varespladib can directly inhibit phospholipase A2 (PLA2) in snake venom, but its protective effect on snakebite-induced ALI and the mechanism have not been clarified. Previous studies have shown that snake venom PLA2 leads to neuron cell death via reactive oxygen species (ROS), one of the initial factors related to the mitophagy pathway. The present study group also found that ROS accumulation occurred after Naja atra envenoming. Hematoxylin and eosin (H/E) staining and immunohistochemistry (IHC) were performed to identify the expression of inflammatory factors in the liver tissue, and flow cytometry and immunofluorescence were used to detect ROS levels and mitochondrial function. Immunofluorescence and western blotting were also used for detecting mitophagy pathway-related proteins. The results showed that N. atra bite induced ALI by activating mitophagy and inducing inflammation and that varespladib had a protective effect. Collectively, these results showed the pathological mechanism of ALI caused by N. atra bite and revealed the protective effect of varespladib.
Collapse
Affiliation(s)
- Wenjie Zhao
- School of Basic Medicine Sciences, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, 330006, China
| | - Jiahao Liu
- School of Basic Medicine Sciences, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, 330006, China
| | - Sidan Wang
- Queen Mary School, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, 330006, China
| | - Qinqin Tao
- School of Basic Medicine Sciences, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, 330006, China
| | - Qiongqiong Lei
- School of Nursing, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, 330006, China.
| | - Chunhong Huang
- School of Basic Medicine Sciences, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, 330006, China.
| |
Collapse
|
6
|
Bin Haidar H, Almeida JR, Williams J, Guo B, Bigot A, Senthilkumaran S, Vaiyapuri S, Patel K. Differential effects of the venoms of Russell's viper and Indian cobra on human myoblasts. Sci Rep 2024; 14:3184. [PMID: 38326450 PMCID: PMC10850160 DOI: 10.1038/s41598-024-53366-9] [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: 07/27/2023] [Accepted: 01/31/2024] [Indexed: 02/09/2024] Open
Abstract
Local tissue damage following snakebite envenoming remains a poorly researched area. To develop better strategies to treat snakebites, it is critical to understand the mechanisms through which venom toxins induce envenomation effects including local tissue damage. Here, we demonstrate how the venoms of two medically important Indian snakes (Russell's viper and cobra) affect human skeletal muscle using a cultured human myoblast cell line. The data suggest that both venoms affect the viability of myoblasts. Russell's viper venom reduced the total number of cells, their migration, and the area of focal adhesions. It also suppressed myogenic differentiation and induced muscle atrophy. While cobra venom decreased the viability, it did not largely affect cell migration and focal adhesions. Cobra venom affected the formation of myotubes and induced atrophy. Cobra venom-induced atrophy could not be reversed by small molecule inhibitors such as varespladib (a phospholipase A2 inhibitor) and prinomastat (a metalloprotease inhibitor), and soluble activin type IIb receptor (a molecule used to promote regeneration of skeletal muscle), although the antivenom (raised against the Indian 'Big Four' snakes) has attenuated the effects. However, all these molecules rescued the myotubes from Russell's viper venom-induced atrophy. This study demonstrates key steps in the muscle regeneration process that are affected by both Indian Russell's viper and cobra venoms and offers insights into the potential causes of clinical features displayed in envenomed victims. Further research is required to investigate the molecular mechanisms of venom-induced myotoxicity under in vivo settings and develop better therapies for snakebite-induced muscle damage.
Collapse
Affiliation(s)
- Husain Bin Haidar
- School of Biological Sciences, University of Reading, Reading, RG6 6UB, UK
- Kuwait Cancer Control Centre, Ministry of Health, Kuwait City, Kuwait
| | - José R Almeida
- School of Pharmacy, University of Reading, Reading, RG6 6UB, UK
| | - Jarred Williams
- School of Pharmacy, University of Reading, Reading, RG6 6UB, UK
| | - Bokai Guo
- School of Biological Sciences, University of Reading, Reading, RG6 6UB, UK
| | - Anne Bigot
- INSERM, CNRS, Institute of Myology, Centre of Research in Myology, Sorbonne Universities, UPMC University Paris, Paris, France
| | | | | | - Ketan Patel
- School of Biological Sciences, University of Reading, Reading, RG6 6UB, UK.
| |
Collapse
|
7
|
Freitas-de-Sousa LA, Colombini M, Souza VC, Silva JPC, Mota-da-Silva A, Almeida MRN, Machado RA, Fonseca WL, Sartim MA, Sachett J, Serrano SMT, Junqueira-de-Azevedo ILM, Grazziotin FG, Monteiro WM, Bernarde PS, Moura-da-Silva AM. Venom Composition of Neglected Bothropoid Snakes from the Amazon Rainforest: Ecological and Toxinological Implications. Toxins (Basel) 2024; 16:83. [PMID: 38393161 PMCID: PMC10891915 DOI: 10.3390/toxins16020083] [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: 12/11/2023] [Revised: 01/18/2024] [Accepted: 01/30/2024] [Indexed: 02/25/2024] Open
Abstract
Snake venoms have evolved in several families of Caenophidae, and their toxins have been assumed to be biochemical weapons with a role as a trophic adaptation. However, it remains unclear how venom contributes to the success of venomous species for adaptation to different environments. Here we compared the venoms from Bothrocophias hyoprora, Bothrops taeniatus, Bothrops bilineatus smaragdinus, Bothrops brazili, and Bothrops atrox collected in the Amazon Rainforest, aiming to understand the ecological and toxinological consequences of venom composition. Transcriptomic and proteomic analyses indicated that the venoms presented the same toxin groups characteristic from bothropoids, but with distinct isoforms with variable qualitative and quantitative abundances, contributing to distinct enzymatic and toxic effects. Despite the particularities of each venom, commercial Bothrops antivenom recognized the venom components and neutralized the lethality of all species. No clear features could be observed between venoms from arboreal and terrestrial habitats, nor in the dispersion of the species throughout the Amazon habitats, supporting the notion that venom composition may not shape the ecological or toxinological characteristics of these snake species and that other factors influence their foraging or dispersal in different ecological niches.
Collapse
Affiliation(s)
| | - Mônica Colombini
- Laboratório de Imunopatologia, Instituto Butantan, São Paulo 05503-900, SP, Brazil; (L.A.F.-d.-S.); (M.C.)
| | - Vinicius C. Souza
- Laboratório de Toxinologia Aplicada, Instituto Butantan, São Paulo 05503-900, SP, Brazil; (V.C.S.); (J.P.C.S.); (S.M.T.S.); (I.L.M.J.-d.-A.)
| | - Joanderson P. C. Silva
- Laboratório de Toxinologia Aplicada, Instituto Butantan, São Paulo 05503-900, SP, Brazil; (V.C.S.); (J.P.C.S.); (S.M.T.S.); (I.L.M.J.-d.-A.)
| | - Ageane Mota-da-Silva
- Instituto Federal do Acre, Campus de Cruzeiro do Sul, Cruzeiro do Sul 69980-000, AC, Brazil;
| | - Marllus R. N. Almeida
- Laboratório de Herpetologia, Universidade Federal do Acre, Campus Floresta, Cruzeiro do Sul 69895-000, AC, Brazil; (M.R.N.A.); (R.A.M.); (W.L.F.); (P.S.B.)
| | - Reginaldo A. Machado
- Laboratório de Herpetologia, Universidade Federal do Acre, Campus Floresta, Cruzeiro do Sul 69895-000, AC, Brazil; (M.R.N.A.); (R.A.M.); (W.L.F.); (P.S.B.)
| | - Wirven L. Fonseca
- Laboratório de Herpetologia, Universidade Federal do Acre, Campus Floresta, Cruzeiro do Sul 69895-000, AC, Brazil; (M.R.N.A.); (R.A.M.); (W.L.F.); (P.S.B.)
| | - Marco A. Sartim
- Instituto de Pesquisa Clínica Carlos Borborema, Fundação de Medicina Tropical Dr. Heitor Vieira Dourado, Manaus 69040-000, AM, Brazil; (M.A.S.); (J.S.); (W.M.M.)
| | - Jacqueline Sachett
- Instituto de Pesquisa Clínica Carlos Borborema, Fundação de Medicina Tropical Dr. Heitor Vieira Dourado, Manaus 69040-000, AM, Brazil; (M.A.S.); (J.S.); (W.M.M.)
| | - Solange M. T. Serrano
- Laboratório de Toxinologia Aplicada, Instituto Butantan, São Paulo 05503-900, SP, Brazil; (V.C.S.); (J.P.C.S.); (S.M.T.S.); (I.L.M.J.-d.-A.)
| | - Inácio L. M. Junqueira-de-Azevedo
- Laboratório de Toxinologia Aplicada, Instituto Butantan, São Paulo 05503-900, SP, Brazil; (V.C.S.); (J.P.C.S.); (S.M.T.S.); (I.L.M.J.-d.-A.)
| | - Felipe G. Grazziotin
- Laboratório de Coleções Zoológicas, Instituto Butantan, São Paulo 05503-900, SP, Brazil;
| | - Wuelton M. Monteiro
- Instituto de Pesquisa Clínica Carlos Borborema, Fundação de Medicina Tropical Dr. Heitor Vieira Dourado, Manaus 69040-000, AM, Brazil; (M.A.S.); (J.S.); (W.M.M.)
| | - Paulo S. Bernarde
- Laboratório de Herpetologia, Universidade Federal do Acre, Campus Floresta, Cruzeiro do Sul 69895-000, AC, Brazil; (M.R.N.A.); (R.A.M.); (W.L.F.); (P.S.B.)
| | - Ana M. Moura-da-Silva
- Laboratório de Imunopatologia, Instituto Butantan, São Paulo 05503-900, SP, Brazil; (L.A.F.-d.-S.); (M.C.)
| |
Collapse
|
8
|
Cardoso FF, Salvador GHM, Cavalcante WLG, Dal-Pai M, Fontes MRDM. BthTX-I, a phospholipase A 2-like toxin, is inhibited by the plant cinnamic acid derivative: chlorogenic acid. BIOCHIMICA ET BIOPHYSICA ACTA. PROTEINS AND PROTEOMICS 2024; 1872:140988. [PMID: 38142025 DOI: 10.1016/j.bbapap.2023.140988] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/05/2023] [Revised: 12/11/2023] [Accepted: 12/18/2023] [Indexed: 12/25/2023]
Abstract
Snakebite is a significant health concern in tropical and subtropical regions, particularly in Africa, Asia, and Latin America, resulting in more than 2.7 million envenomations and an estimated one hundred thousand fatalities annually. The Bothrops genus is responsible for the majority of snakebite envenomings in Latin America and Caribbean countries. Accidents involving snakes from this genus are characterized by local symptoms that often lead to permanent sequelae and death. However, specific antivenoms exhibit limited effectiveness in inhibiting local tissue damage. Phospholipase A2-like (PLA2-like) toxins emerge as significant contributors to local myotoxicity in accidents involving Bothrops species. As a result, they represent a crucial target for prospective treatments. Some natural and synthetic compounds have shown the ability to reduce or abolish the myotoxic effects of PLA2-like proteins. In this study, we employed a combination approach involving myographic, morphological, biophysical and bioinformatic techniques to investigate the interaction between chlorogenic acid (CGA) and BthTX-I, a PLA2-like toxin. CGA provided a protection of 71.8% on muscle damage in a pre-incubation treatment. Microscale thermophoresis and circular dichroism experiments revealed that CGA interacted with the BthTX-I while preserving its secondary structure. CGA exhibited an affinity to the toxin that ranks among the highest observed for a natural compound. Bioinformatics simulations indicated that CGA inhibitor binds to the toxin's hydrophobic channel in a manner similar to other phenolic compounds previously investigated. These findings suggest that CGA interferes with the allosteric transition of the non-activated toxin, and the stability of the dimeric assembly of its activated state.
Collapse
Affiliation(s)
- Fábio Florença Cardoso
- Departamento de Biofísica e Farmacologia, Instituto de Biociências, Universidade Estadual Paulista (UNESP), Botucatu, SP, Brazil
| | | | - Walter Luís Garrido Cavalcante
- Departamento de Biofísica e Farmacologia, Instituto de Biociências, Universidade Estadual Paulista (UNESP), Botucatu, SP, Brazil; Departamento de Farmacologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, MG, Brazil
| | - Maeli Dal-Pai
- Departamento de Biologia Estrutural e Funcional, Instituto de Biociências, Universidade Estadual Paulista (UNESP), Botucatu, SP, Brazil
| | - Marcos Roberto de Mattos Fontes
- Departamento de Biofísica e Farmacologia, Instituto de Biociências, Universidade Estadual Paulista (UNESP), Botucatu, SP, Brazil; Instituto de Estudos Avançados do Mar (IEAMar), Universidade Estadual Paulista (UNESP), São Vicente, SP, Brazil.
| |
Collapse
|
9
|
de Oliveira ALN, Lacerda MT, Ramos MJ, Fernandes PA. Viper Venom Phospholipase A2 Database: The Structural and Functional Anatomy of a Primary Toxin in Envenomation. Toxins (Basel) 2024; 16:71. [PMID: 38393149 PMCID: PMC10893444 DOI: 10.3390/toxins16020071] [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: 09/25/2023] [Revised: 01/04/2024] [Accepted: 01/13/2024] [Indexed: 02/25/2024] Open
Abstract
Viper venom phospholipase A2 enzymes (vvPLA2s) and phospholipase A2-like (PLA2-like) proteins are two of the principal toxins in viper venom that are responsible for the severe myotoxic and neurotoxic effects caused by snakebite envenoming, among other pathologies. As snakebite envenoming is the deadliest neglected tropical disease, a complete understanding of these proteins' properties and their mechanisms of action is urgently needed. Therefore, we created a database comprising information on the holo-form, cofactor-bound 3D structure of 217 vvPLA2 and PLA2-like proteins in their physiologic environment, as well as 79 membrane-bound viper species from 24 genera, which we have made available to the scientific community to accelerate the development of new anti-snakebite drugs. In addition, the analysis of the sequenced, 3D structure of the database proteins reveals essential aspects of the anatomy of the proteins, their toxicity mechanisms, and the conserved binding site areas that may anchor universal interspecific inhibitors. Moreover, it pinpoints hypotheses for the molecular origin of the myotoxicity of the PLA2-like proteins. Altogether, this study provides an understanding of the diversity of these toxins and how they are conserved, and it indicates how to develop broad, interspecies, efficient small-molecule inhibitors to target the toxin's many mechanisms of action.
Collapse
Affiliation(s)
| | | | | | - Pedro A. Fernandes
- Requimte-Faculty of Sciences, University of Porto, Rua do Campo Alegre s/n, 4169-000 Porto, Portugal; (A.L.N.d.O.); (M.T.L.); (M.J.R.)
| |
Collapse
|
10
|
Dingwoke EJ, Adamude FA, Salihu A, Abubakar MS, Sallau AB. Toxicological analyses of the venoms of Nigerian vipers Echis ocellatus and Bitis arietans. Trop Med Health 2024; 52:15. [PMID: 38282015 PMCID: PMC10823708 DOI: 10.1186/s41182-024-00581-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Accepted: 01/16/2024] [Indexed: 01/30/2024] Open
Abstract
BACKGROUND Among the medically important snakes in Nigeria, Echis ocellatus and Bitis arietans have the most lethal venom. These venoms were classified according to the presence of snake venom metalloproteinases (SVMPs), snake venom phospholipase A2 (PLA2s), and snake venom serine proteases (SVSPs). Toxicological analyzes were performed to understand the significance of different protein families in venoms. METHODS Proteins were separated from venom using column chromatography. The skin and footpad of mice were used to determine hemorrhagic and edematogenic activities. Caprine blood plasma was used to test fibrinolytic activity in vitro. RESULTS The results showed that, compared to the crude venom, the SVMP fraction induced hemorrhagic effects with a diameter of 26.00 ± 1.00 mm in E. ocellatus and 21.33 ± 1.52 mm in B. arietans. Both SVSP and SVMP had anticoagulant effects; however, the SVSP fraction had a stronger effect, with a longer anticoagulation time of 30.00 ± 3.00 min in E. ocellatus and 26.00 ± 2.00 min in B. arietans. These main venom toxins, SVMPs, SVSPs, and PLA2, were found to have edema-forming effects that were optimal at 2 h after envenomation. PLA2s had the highest edema-inducing activity, with onset 30 min after envenomation. CONCLUSIONS Given the importance of SVMPs in altering the integrity of the membrane structure and impairing the blood coagulation system, an antivenom that can specifically neutralize its activity could inhibit the hemorrhage effects of the venoms.
Collapse
Affiliation(s)
- Emeka John Dingwoke
- Department of Tropical Diseases, UNESCO-International Center for Biotechnology, University of Nigeria, Nsukka, Enugu State, Nigeria.
- Department of Biochemistry, Faculty of Life Sciences, Ahmadu Bello University, Zaria, Kaduna State, Nigeria.
- Venom, Antivenom and Natural Toxins Research Centre, Ahmadu Bello University, Zaria, Kaduna State, Nigeria.
| | - Fatima Adis Adamude
- Department of Biochemistry, Faculty of Sciences, Federal University, Lafia, Nasarawa State, Nigeria
- Venom, Antivenom and Natural Toxins Research Centre, Ahmadu Bello University, Zaria, Kaduna State, Nigeria
| | - Aliyu Salihu
- Department of Biochemistry, Faculty of Life Sciences, Ahmadu Bello University, Zaria, Kaduna State, Nigeria
| | - Mujitaba Suleiman Abubakar
- Department of Pharmacognosy and Drug Development, Faculty of Pharmaceutical Sciences, Ahmadu Bello University, Zaria, Kaduna State, Nigeria
- Venom, Antivenom and Natural Toxins Research Centre, Ahmadu Bello University, Zaria, Kaduna State, Nigeria
| | - Abdullahi Balarabe Sallau
- Department of Biochemistry, Faculty of Life Sciences, Ahmadu Bello University, Zaria, Kaduna State, Nigeria.
- Venom, Antivenom and Natural Toxins Research Centre, Ahmadu Bello University, Zaria, Kaduna State, Nigeria.
| |
Collapse
|
11
|
Kumar A, Madni ZK, Chaturvedi S, Salunke DM. Recombinant human scFv antibody fragments against phospholipase A2 from Naja naja and Echis carinatus snake venoms: In vivo neutralization and mechanistic insights. Mol Immunol 2024; 165:55-67. [PMID: 38154407 DOI: 10.1016/j.molimm.2023.12.006] [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: 08/03/2023] [Revised: 11/07/2023] [Accepted: 12/17/2023] [Indexed: 12/30/2023]
Abstract
Snake envenomation results in a range of clinical sequelae, and widely used animal-based conventional antivenoms exhibit several limitations including the adverse immunological effects in human snake bite victims. Therefore, human monoclonal anti-snake venom antibodies or fragments can be an alternate therapy for overcoming the existing limitations. We developed venom-neutralizing humanized scFv antibodies and analyzed biochemical mechanisms associated with the inhibition of toxicity. Tomlinson I and J human scFv antibody libraries were screened against Naja naja and Echis carinatus venoms, and seven unique scFv antibodies were obtained. Further, specific toxins of snake venom interacting with each of these scFvs were identified, and phospholipase A2 (PLA2) was found to be prominently captured by the phage-anchored scFv antibodies. Our study indicated PLA2 to be one of the abundant toxins in Naja naja and Echis carinatus venom samples. The scFvs binding to PLA2 were used to perform in vivo survival assay using the mouse model and in vitro toxin inhibition assays. scFv N194, which binds to acidic PLA2, protected 50% of mice treated with Naja naja venom. Significant prolongation of survival time and 16% survival were observed in Echis carinatus venom-challenged mice treated with scFv E113 and scFv E10, respectively. However, a combination comprised of an equal amount of two scFvs, E113 and E10, both interacting with basic PLA2, exhibited synergistically enhanced survival of 33% in Echis carinatus venom-challenged mice. No such synergistically enhanced survival was observed in the case of combinatorial treatment with anti-Naja naja scFvs, N194, and N248. These scFvs demonstrated partial inhibition of venom-induced myotoxicity, and E113 also inhibited hemolysis by 50%, which corroborates the enhanced survival during combinatorial treatment in Echis carinatus venom-challenged mice.
Collapse
Affiliation(s)
- Amit Kumar
- International Centre for Genetic Engineering and Biotechnology, Aruna Asaf Ali Marg, New Delhi 110067, India
| | - Zaid Kamal Madni
- International Centre for Genetic Engineering and Biotechnology, Aruna Asaf Ali Marg, New Delhi 110067, India
| | - Shivam Chaturvedi
- International Centre for Genetic Engineering and Biotechnology, Aruna Asaf Ali Marg, New Delhi 110067, India
| | - Dinakar M Salunke
- International Centre for Genetic Engineering and Biotechnology, Aruna Asaf Ali Marg, New Delhi 110067, India.
| |
Collapse
|
12
|
Sampat GH, Hiremath K, Dodakallanavar J, Patil VS, Harish DR, Biradar P, Mahadevamurthy RK, Barvaliya M, Roy S. Unraveling snake venom phospholipase A 2: an overview of its structure, pharmacology, and inhibitors. Pharmacol Rep 2023; 75:1454-1473. [PMID: 37926795 DOI: 10.1007/s43440-023-00543-8] [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: 06/12/2023] [Revised: 10/05/2023] [Accepted: 10/06/2023] [Indexed: 11/07/2023]
Abstract
Snake bite is a neglected disease that affects millions of people worldwide. WHO reported approximately 5 million people are bitten by various species of snakes each year, resulting in nearly 1 million deaths and an additional three times cases of permanent disability. Snakes utilize the venom mainly for immobilization and digestion of their prey. Snake venom is a composition of proteins and enzymes which is responsible for its diverse pharmacological action. Snake venom phospholipase A2 (SvPLA2) is an enzyme that is present in every snake species in different quantities and is known to produce remarkable functional diversity and pharmacological action like inflammation, necrosis, myonecrosis, hemorrhage, etc. Arachidonic acid, a precursor to eicosanoids, such as prostaglandins and leukotrienes, is released when SvPLA2 catalyzes the hydrolysis of the sn-2 positions of membrane glycerophospholipids, which is responsible for its actions. Polyvalent antivenom produced from horses or lambs is the standard treatment for snake envenomation, although it has many drawbacks. Traditional medical practitioners treat snake bites using plants and other remedies as a sustainable alternative. More than 500 plant species from more than 100 families reported having venom-neutralizing abilities. Plant-derived secondary metabolites have the ability to reduce the venom's adverse consequences. Numerous studies have documented the ability of plant chemicals to inhibit the enzymes found in snake venom. Research in recent years has shown that various small molecules, such as varespladib and methyl varespladib, effectively inhibit the PLA2 toxin. In the present article, we have overviewed the knowledge of snake venom phospholipase A2, its classification, and the mechanism involved in the pathophysiology of cytotoxicity, myonecrosis, anticoagulation, and inflammation clinical application and inhibitors of SvPLA2, along with the list of studies carried out to evaluate the potency of small molecules like varespladib and secondary metabolites from the traditional medicine for their anti-PLA2 effect.
Collapse
Affiliation(s)
- Ganesh H Sampat
- ICMR-National Institute of Traditional Medicine, Belagavi, Karnataka, 590010, India
- KLE College of Pharmacy, Belagavi, KLE Academy of Higher Education and Research, Belagavi, Karnataka, 590010, India
| | - Kashinath Hiremath
- ICMR-National Institute of Traditional Medicine, Belagavi, Karnataka, 590010, India
- KLE College of Pharmacy, Belagavi, KLE Academy of Higher Education and Research, Belagavi, Karnataka, 590010, India
| | - Jagadeesh Dodakallanavar
- ICMR-National Institute of Traditional Medicine, Belagavi, Karnataka, 590010, India
- KLE College of Pharmacy, Belagavi, KLE Academy of Higher Education and Research, Belagavi, Karnataka, 590010, India
| | - Vishal S Patil
- ICMR-National Institute of Traditional Medicine, Belagavi, Karnataka, 590010, India
- KLE College of Pharmacy, Belagavi, KLE Academy of Higher Education and Research, Belagavi, Karnataka, 590010, India
| | - Darasaguppe R Harish
- ICMR-National Institute of Traditional Medicine, Belagavi, Karnataka, 590010, India.
| | - Prakash Biradar
- KLE College of Pharmacy, Belagavi, KLE Academy of Higher Education and Research, Belagavi, Karnataka, 590010, India.
| | | | - Manish Barvaliya
- ICMR-National Institute of Traditional Medicine, Belagavi, Karnataka, 590010, India
| | - Subarna Roy
- ICMR-National Institute of Traditional Medicine, Belagavi, Karnataka, 590010, India
| |
Collapse
|
13
|
López-Dávila AJ, Weber N, Nayak A, Fritz L, Moustafa KR, Gand LV, Wehry E, Kraft T, Thum T, Fernández J, Gutiérrez JM, Lomonte B. Skeletal muscle fiber hypercontraction induced by Bothrops asper myotoxic phospholipases A 2 ex vivo does not involve a direct action on the contractile apparatus. Pflugers Arch 2023; 475:1193-1202. [PMID: 37474774 PMCID: PMC10499977 DOI: 10.1007/s00424-023-02840-w] [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: 04/29/2023] [Revised: 06/19/2023] [Accepted: 07/10/2023] [Indexed: 07/22/2023]
Abstract
Myonecrosis is a frequent clinical manifestation of envenomings by Viperidae snakes, mainly caused by the toxic actions of secreted phospholipase A2 (sPLA2) enzymes and sPLA2-like homologs on skeletal muscle fibers. A hallmark of the necrotic process induced by these myotoxins is the rapid appearance of hypercontracted muscle fibers, attributed to the massive influx of Ca2+ resulting from cell membrane damage. However, the possibility of myotoxins having, in addition, a direct effect on the contractile machinery of skeletal muscle fibers when internalized has not been investigated. This question is here addressed by using an ex vivo model of single-skinned muscle fibers, which lack membranes but retain an intact contractile apparatus. Rabbit psoas skinned fibers were exposed to two types of myotoxins of Bothrops asper venom: Mt-I, a catalytically active Asp49 sPLA2 enzyme, and Mt-II, a Lys49 sPLA2-like protein devoid of phospholipolytic activity. Neither of these myotoxins affected the main parameters of force development in striated muscle sarcomeres of the skinned fibers. Moreover, no microscopical alterations were evidenced after their exposure to Mt-I or Mt-II. In contrast to the lack of effects on skinned muscle fibers, both myotoxins induced a strong hypercontraction in myotubes differentiated from murine C2C12 myoblasts, with drastic morphological alterations that reproduce those described in myonecrotic tissue in vivo. As neither Mt-I nor Mt-II showed direct effects upon the contractile apparatus of skinned fibers, it is concluded that the mechanism of hypercontraction triggered by both myotoxins in patients involves indirect effects, i.e., the large cytosolic Ca2+ increase after sarcolemma permeabilization.
Collapse
Affiliation(s)
- Alfredo Jesús López-Dávila
- Institute of Molecular and Cell Physiology, Hannover Medical School, Carl-Neuberg-Str. 1, 30625, Hannover, Germany.
| | - Natalie Weber
- Institute of Molecular and Translational Therapeutic Strategies, Hannover Medical School, Carl-Neuberg-Str. 1, 30625, Hannover, Germany
| | - Arnab Nayak
- Institute of Molecular and Cell Physiology, Hannover Medical School, Carl-Neuberg-Str. 1, 30625, Hannover, Germany
| | - Leon Fritz
- Institute of Molecular and Cell Physiology, Hannover Medical School, Carl-Neuberg-Str. 1, 30625, Hannover, Germany
| | - Kian Rami Moustafa
- Institute of Molecular and Translational Therapeutic Strategies, Hannover Medical School, Carl-Neuberg-Str. 1, 30625, Hannover, Germany
| | - Luis Vincens Gand
- Institute of Molecular and Cell Physiology, Hannover Medical School, Carl-Neuberg-Str. 1, 30625, Hannover, Germany
| | - Enke Wehry
- Institute of Molecular and Cell Physiology, Hannover Medical School, Carl-Neuberg-Str. 1, 30625, Hannover, Germany
| | - Theresia Kraft
- Institute of Molecular and Cell Physiology, Hannover Medical School, Carl-Neuberg-Str. 1, 30625, Hannover, Germany
| | - Thomas Thum
- Institute of Molecular and Translational Therapeutic Strategies, Hannover Medical School, Carl-Neuberg-Str. 1, 30625, Hannover, Germany
| | - Julián Fernández
- Instituto Clodomiro Picado, Facultad de Microbiología, Universidad de Costa Rica, San José, 11501, Costa Rica
| | - José María Gutiérrez
- Instituto Clodomiro Picado, Facultad de Microbiología, Universidad de Costa Rica, San José, 11501, Costa Rica
| | - Bruno Lomonte
- Instituto Clodomiro Picado, Facultad de Microbiología, Universidad de Costa Rica, San José, 11501, Costa Rica
| |
Collapse
|
14
|
Henrickson A, Montina T, Hazendonk P, Lomonte B, Neves-Ferreira AGC, Demeler B. SDS-induced hexameric oligomerization of myotoxin-II from Bothrops asper assessed by sedimentation velocity and nuclear magnetic resonance. EUROPEAN BIOPHYSICS JOURNAL : EBJ 2023; 52:445-457. [PMID: 37209172 PMCID: PMC10526984 DOI: 10.1007/s00249-023-01658-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/31/2022] [Revised: 04/21/2023] [Accepted: 04/30/2023] [Indexed: 05/22/2023]
Abstract
We report the solution behavior, oligomerization state, and structural details of myotoxin-II purified from the venom of Bothrops asper in the presence and absence of sodium dodecyl sulfate (SDS) and multiple lipids, as examined by analytical ultracentrifugation and nuclear magnetic resonance. Molecular functional and structural details of the myotoxic mechanism of group II Lys-49 phospholipase A2 homologues have been only partially elucidated so far, and conflicting observations have been reported in the literature regarding the monomeric vs. oligomeric state of these toxins in solution. We observed the formation of a stable and discrete, hexameric form of myotoxin-II, but only in the presence of small amounts of SDS. In SDS-free medium, myotoxin-II was insensitive to mass action and remained monomeric at all concentrations examined (up to 3 mg/ml, 218.2 μM). At SDS concentrations above the critical micelle concentration, only dimers and trimers were observed, and at intermediate SDS concentrations, aggregates larger than hexamers were observed. We found that the amount of SDS required to form a stable hexamer varies with protein concentration, suggesting the need for a precise stoichiometry of free SDS molecules. The discovery of a stable hexameric species in the presence of a phospholipid mimetic suggests a possible physiological role for this oligomeric form, and may shed light on the poorly understood membrane-disrupting mechanism of this myotoxic protein class.
Collapse
Affiliation(s)
- Amy Henrickson
- Department of Chemistry and Biochemistry, University of Lethbridge, Lethbridge, AB, Canada
| | - Tony Montina
- Department of Chemistry and Biochemistry, University of Lethbridge, Lethbridge, AB, Canada
| | - Paul Hazendonk
- Department of Chemistry and Biochemistry, University of Lethbridge, Lethbridge, AB, Canada
| | - Bruno Lomonte
- Instituto Clodomiro Picado, Facultad de Microbiología San José, Universidad de Costa Rica, San José, Costa Rica
| | | | - Borries Demeler
- Department of Chemistry and Biochemistry, University of Lethbridge, Lethbridge, AB, Canada.
- Department of Chemistry and Biochemistry, University of Montana, Missoula, MT, USA.
| |
Collapse
|
15
|
Lomonte B. Lys49 myotoxins, secreted phospholipase A 2-like proteins of viperid venoms: A comprehensive review. Toxicon 2023; 224:107024. [PMID: 36632869 DOI: 10.1016/j.toxicon.2023.107024] [Citation(s) in RCA: 21] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Revised: 01/06/2023] [Accepted: 01/06/2023] [Indexed: 01/11/2023]
Abstract
Muscle necrosis is a potential clinical complication of snakebite envenomings, which in severe cases can lead to functional or physical sequelae such as disability or amputation. Snake venom proteins with the ability to directly damage skeletal muscle fibers are collectively referred to as myotoxins, and include three main types: cytolysins of the "three-finger toxin" protein family expressed in many elapid venoms, the so-called "small" myotoxins found in a number of rattlesnake venoms, and the widespread secreted phospholipase A2 (sPLA2) molecules. Among the latter, protein variants that conserve the sPLA2 structure, but lack such enzymatic activity, have been increasingly found in the venoms of many viperid species. Intriguingly, these sPLA2-like proteins are able to induce muscle necrosis by a mechanism independent of phospholipid hydrolysis. They are commonly referred to as "Lys49 myotoxins" since they most often present, among other substitutions, the replacement of the otherwise invariant residue Asp49 of sPLA2s by Lys. This work comprehensively reviews the historical developments and current knowledge towards deciphering the mechanism of action of Lys49 sPLA2-like myotoxins, and points out main gaps to be filled for a better understanding of these multifaceted snake venom proteins, to hopefully lead to improved treatments for snakebites.
Collapse
Affiliation(s)
- Bruno Lomonte
- Instituto Clodomiro Picado, Facultad de Microbiología, Universidad de Costa Rica, San José, 11501, Costa Rica.
| |
Collapse
|
16
|
Secretory Phospholipases A2, from Snakebite Envenoming to a Myriad of Inflammation Associated Human Diseases-What Is the Secret of Their Activity? Int J Mol Sci 2023; 24:ijms24021579. [PMID: 36675102 PMCID: PMC9863470 DOI: 10.3390/ijms24021579] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Revised: 01/05/2023] [Accepted: 01/11/2023] [Indexed: 01/15/2023] Open
Abstract
Secreted phospholipases of type A2 (sPLA2s) are proteins of 14-16 kDa present in mammals in different forms and at different body sites. They are involved in lipid transformation processes, and consequently in various immune, inflammatory, and metabolic processes. sPLA2s are also major components of snake venoms, endowed with various toxic and pharmacological properties. The activity of sPLA2s is not limited to the enzymatic one but, through interaction with different types of molecules, they exert other activities that are still little known and explored, both outside and inside the cells, as they can be endocytosed. The aim of this review is to analyze three features of sPLA2s, yet under-explored, knowledge of which could be crucial to understanding the activity of these proteins. The first feature is their disulphide bridge pattern, which has always been considered immutable and necessary for their stability, but which might instead be modulable. The second characteristic is their ability to undergo various post-translational modifications that would control their interaction with other molecules. The third feature is their ability to participate in active molecular condensates both on the surface and within the cell. Finally, the implications of these features in the design of anti-inflammatory drugs are discussed.
Collapse
|
17
|
Lewin MR, Carter RW, Matteo IA, Samuel SP, Rao S, Fry BG, Bickler PE. Varespladib in the Treatment of Snakebite Envenoming: Development History and Preclinical Evidence Supporting Advancement to Clinical Trials in Patients Bitten by Venomous Snakes. Toxins (Basel) 2022; 14:783. [PMID: 36422958 PMCID: PMC9695340 DOI: 10.3390/toxins14110783] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2022] [Revised: 10/26/2022] [Accepted: 11/01/2022] [Indexed: 11/16/2022] Open
Abstract
The availability of effective, reliably accessible, and affordable treatments for snakebite envenoming is a critical and long unmet medical need. Recently, small, synthetic toxin-specific inhibitors with oral bioavailability used in conjunction with antivenom have been identified as having the potential to greatly improve outcomes after snakebite. Varespladib, a small, synthetic molecule that broadly and potently inhibits secreted phospholipase A2 (sPLA2s) venom toxins has renewed interest in this class of inhibitors due to its potential utility in the treatment of snakebite envenoming. The development of varespladib and its oral dosage form, varespladib-methyl, has been accelerated by previous clinical development campaigns to treat non-envenoming conditions related to ulcerative colitis, rheumatoid arthritis, asthma, sepsis, and acute coronary syndrome. To date, twenty-nine clinical studies evaluating the safety, pharmacokinetics (PK), and efficacy of varespladib for non-snakebite envenoming conditions have been completed in more than 4600 human subjects, and the drugs were generally well-tolerated and considered safe for use in humans. Since 2016, more than 30 publications describing the structure, function, and efficacy of varespladib have directly addressed its potential for the treatment of snakebite. This review summarizes preclinical findings and outlines the scientific support, the potential limitations, and the next steps in the development of varespladib's use as a snakebite treatment, which is now in Phase 2 human clinical trials in the United States and India.
Collapse
Affiliation(s)
- Matthew R. Lewin
- Division of Research, Ophirex, Inc., Corte Madera, CA 94925, USA
- Center for Exploration and Travel Health, California Academy of Sciences, San Francisco, CA 94118, USA
| | | | - Isabel A. Matteo
- Center for Exploration and Travel Health, California Academy of Sciences, San Francisco, CA 94118, USA
| | | | - Sunita Rao
- Division of Research, Ophirex, Inc., Corte Madera, CA 94925, USA
| | - Bryan G. Fry
- Venom Evolution Lab, School of Biological Science, University of Queensland, St. Lucia, QLD 4072, Australia
| | - Philip E. Bickler
- Center for Exploration and Travel Health, California Academy of Sciences, San Francisco, CA 94118, USA
- Department of Anesthesia and Perioperative Care, University of California San Francisco School of Medicine, San Francisco, CA 94143, USA
| |
Collapse
|
18
|
Mora-Obando D, Lomonte B, Pla D, Guerrero-Vargas JA, Ayerbe-González S, Gutiérrez JM, Sasa M, Calvete JJ. Half a century of research on Bothrops asper venom variation: Biological and biomedical implications. Toxicon 2022; 221:106983. [DOI: 10.1016/j.toxicon.2022.106983] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2022] [Revised: 11/21/2022] [Accepted: 11/22/2022] [Indexed: 11/24/2022]
|
19
|
Luo W, Xu Y, Liu R, Liao Y, Wang S, Zhang H, Li X, Wang H. Retinoic acid and RARγ maintain satellite cell quiescence through regulation of translation initiation. Cell Death Dis 2022; 13:838. [PMID: 36175396 PMCID: PMC9522790 DOI: 10.1038/s41419-022-05284-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2022] [Revised: 09/18/2022] [Accepted: 09/20/2022] [Indexed: 01/23/2023]
Abstract
In adult skeletal muscle, satellite cells are in a quiescent state, which is essential for the future activation of muscle homeostasis and regeneration. Multiple studies have investigated satellite cell proliferation and differentiation, but the molecular mechanisms that safeguard the quiescence of satellite cells remain largely unknown. In this study, we purposely activated dormant satellite cells by using various stimuli and captured the in vivo-preserved features from quiescence to activation transitions. We found that retinoic acid signaling was required for quiescence maintenance. Mechanistically, retinoic acid receptor gamma (RARγ) binds to and stimulates genes responsible for Akt dephosphorylation and subsequently inhibits overall protein translation initiation in satellite cells. Furthermore, the alleviation of retinoic acid signaling released the satellite cells from quiescence, but this restraint was lost in aged cells. Retinoic acid also preserves the quiescent state during satellite cell isolation, overcoming the cellular stress caused by the isolation process. We conclude that active retinoic acid signaling contributes to the maintenance of the quiescent state of satellite cells through regulation of the protein translation initiation process.
Collapse
Affiliation(s)
- Wenzhe Luo
- grid.35155.370000 0004 1790 4137Key Laboratory of Agricultural Animal Genetics, Breeding, and Reproduction of the Ministry of Education, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, China ,grid.440622.60000 0000 9482 4676College of Animal Science and Technology, Shandong Agricultural University, Taian, China
| | - Yueyuan Xu
- grid.35155.370000 0004 1790 4137Key Laboratory of Agricultural Animal Genetics, Breeding, and Reproduction of the Ministry of Education, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, China
| | - Ruige Liu
- grid.35155.370000 0004 1790 4137Key Laboratory of Agricultural Animal Genetics, Breeding, and Reproduction of the Ministry of Education, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, China
| | - Yinlong Liao
- grid.35155.370000 0004 1790 4137Key Laboratory of Agricultural Animal Genetics, Breeding, and Reproduction of the Ministry of Education, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, China
| | - Sheng Wang
- grid.35155.370000 0004 1790 4137Key Laboratory of Agricultural Animal Genetics, Breeding, and Reproduction of the Ministry of Education, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, China
| | - Haoyuan Zhang
- grid.35155.370000 0004 1790 4137Key Laboratory of Agricultural Animal Genetics, Breeding, and Reproduction of the Ministry of Education, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, China
| | - Xinyun Li
- grid.35155.370000 0004 1790 4137Key Laboratory of Agricultural Animal Genetics, Breeding, and Reproduction of the Ministry of Education, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, China
| | - Heng Wang
- grid.35155.370000 0004 1790 4137Key Laboratory of Agricultural Animal Genetics, Breeding, and Reproduction of the Ministry of Education, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, China ,grid.440622.60000 0000 9482 4676College of Animal Science and Technology, Shandong Agricultural University, Taian, China
| |
Collapse
|
20
|
Diversity of Phospholipases A2 from Bothrops atrox Snake Venom: Adaptive Advantages for Snakes Compromising Treatments for Snakebite Patients. Toxins (Basel) 2022; 14:toxins14080543. [PMID: 36006204 PMCID: PMC9414272 DOI: 10.3390/toxins14080543] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2022] [Revised: 08/02/2022] [Accepted: 08/04/2022] [Indexed: 11/25/2022] Open
Abstract
The evolution of snake venoms resulted in multigene toxin families that code for structurally similar isoforms eventually harboring distinct functions. PLA2s are dominant toxins in viper venoms, and little is known about the impact of their diversity on human envenomings and neutralization by antivenoms. Here, we show the isolation of three distinct PLA2s from B. atrox venom. FA1 is a Lys-49 homologue, and FA3 and FA4 are catalytic Asp-49 PLA2s. FA1 and FA3 are basic myotoxic proteins, while FA4 is an acid non-myotoxic PLA2. FA3 was the most potent toxin, inducing higher levels of edema, inflammatory nociception, indirect hemolysis, and anticoagulant activity on human, rat, and chicken plasmas. FA4 presented lower anticoagulant activity, and FA1 had only a slight effect on human and rat plasmas. PLA2s presented differential reactivities with antivenoms, with an emphasis on FA3, which was not recognized or neutralized by the antivenoms used in this study. Our findings reveal the functional and antigenic diversity among PLA2s from B. atrox venom, highlighting the importance of assessing venom variability for understanding human envenomations and treatment with antivenoms, particularly evident here as the antivenom fails to recognize FA3, the most active multifunctional toxin described.
Collapse
|
21
|
Pharmacological Screening of Venoms from Five Brazilian Micrurus Species on Different Ion Channels. Int J Mol Sci 2022; 23:ijms23147714. [PMID: 35887062 PMCID: PMC9318628 DOI: 10.3390/ijms23147714] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Revised: 07/05/2022] [Accepted: 07/07/2022] [Indexed: 12/05/2022] Open
Abstract
Coral snake venoms from the Micrurus genus are a natural library of components with multiple targets, yet are poorly explored. In Brazil, 34 Micrurus species are currently described, and just a few have been investigated for their venom activities. Micrurus venoms are composed mainly of phospholipases A2 and three-finger toxins, which are responsible for neuromuscular blockade—the main envenomation outcome in humans. Beyond these two major toxin families, minor components are also important for the global venom activity, including Kunitz-peptides, serine proteases, 5′ nucleotidases, among others. In the present study, we used the two-microelectrode voltage clamp technique to explore the crude venom activities of five different Micrurus species from the south and southeast of Brazil: M. altirostris, M. corallinus, M. frontalis, M. carvalhoi and M. decoratus. All five venoms induced full inhibition of the muscle-type α1β1δε nAChR with different levels of reversibility. We found M. altirostris and M. frontalis venoms acting as partial inhibitors of the neuronal-type α7 nAChR with an interesting subsequent potentiation after one washout. We discovered that M. altirostris and M. corallinus venoms modulate the α1β2 GABAAR. Interestingly, the screening on KV1.3 showed that all five Micrurus venoms act as inhibitors, being totally reversible after the washout. Since this activity seems to be conserved among different species, we hypothesized that the Micrurus venoms may rely on potassium channel inhibitory activity as an important feature of their envenomation strategy. Finally, tests on NaV1.2 and NaV1.4 showed that these channels do not seem to be targeted by Micrurus venoms. In summary, the venoms tested are multifunctional, each of them acting on at least two different types of targets.
Collapse
|
22
|
Jia Y, Vega C. Biochemical and computational approaches to understand venom toxin-toxin interaction. Toxicon 2022; 216:11-14. [PMID: 35772507 DOI: 10.1016/j.toxicon.2022.06.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2022] [Revised: 06/20/2022] [Accepted: 06/21/2022] [Indexed: 11/30/2022]
Abstract
Snake venoms are mainly composed of proteins and peptides (venom toxins). The venom transcriptomes and proteomes have been extensively investigated; however, venom toxin-toxin interactions remain poorly characterized. We detected the interaction of venom Asp49-PLA2 and 3FTx using biochemical and computational approaches. A stable structure of Asp49-PLA2-3FTx was identified, and the interface of Asp49-PLA2 and 3FTx was analyzed. The approaches will shed light on understanding the venom complexity and deciphering the synergistic effects of venom toxins.
Collapse
Affiliation(s)
- Ying Jia
- Department of Biology, The University of Texas Rio Grande Valley, Brownsville, TX, 78520, USA.
| | - Christine Vega
- Department of Biology, The University of Texas Rio Grande Valley, Brownsville, TX, 78520, USA
| |
Collapse
|
23
|
Oliveira AL, Viegas MF, da Silva SL, Soares AM, Ramos MJ, Fernandes PA. The chemistry of snake venom and its medicinal potential. Nat Rev Chem 2022; 6:451-469. [PMID: 35702592 PMCID: PMC9185726 DOI: 10.1038/s41570-022-00393-7] [Citation(s) in RCA: 81] [Impact Index Per Article: 40.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/26/2022] [Indexed: 12/15/2022]
Abstract
The fascination and fear of snakes dates back to time immemorial, with the first scientific treatise on snakebite envenoming, the Brooklyn Medical Papyrus, dating from ancient Egypt. Owing to their lethality, snakes have often been associated with images of perfidy, treachery and death. However, snakes did not always have such negative connotations. The curative capacity of venom has been known since antiquity, also making the snake a symbol of pharmacy and medicine. Today, there is renewed interest in pursuing snake-venom-based therapies. This Review focuses on the chemistry of snake venom and the potential for venom to be exploited for medicinal purposes in the development of drugs. The mixture of toxins that constitute snake venom is examined, focusing on the molecular structure, chemical reactivity and target recognition of the most bioactive toxins, from which bioactive drugs might be developed. The design and working mechanisms of snake-venom-derived drugs are illustrated, and the strategies by which toxins are transformed into therapeutics are analysed. Finally, the challenges in realizing the immense curative potential of snake venom are discussed, and chemical strategies by which a plethora of new drugs could be derived from snake venom are proposed.
Collapse
Affiliation(s)
- Ana L. Oliveira
- Department of Chemistry and Biochemistry, Faculty of Sciences, University of Porto, Porto, Portugal
- LAQV/Requimte, University of Porto, Porto, Portugal
| | - Matilde F. Viegas
- Department of Chemistry and Biochemistry, Faculty of Sciences, University of Porto, Porto, Portugal
- LAQV/Requimte, University of Porto, Porto, Portugal
| | - Saulo L. da Silva
- Department of Chemistry and Biochemistry, Faculty of Sciences, University of Porto, Porto, Portugal
- LAQV/Requimte, University of Porto, Porto, Portugal
| | - Andreimar M. Soares
- Biotechnology Laboratory for Proteins and Bioactive Compounds from the Western Amazon, Oswaldo Cruz Foundation, National Institute of Epidemiology in the Western Amazon (INCT-EpiAmO), Porto Velho, Brazil
- Sao Lucas Universitary Center (UniSL), Porto Velho, Brazil
| | - Maria J. Ramos
- Department of Chemistry and Biochemistry, Faculty of Sciences, University of Porto, Porto, Portugal
- LAQV/Requimte, University of Porto, Porto, Portugal
| | - Pedro A. Fernandes
- Department of Chemistry and Biochemistry, Faculty of Sciences, University of Porto, Porto, Portugal
- LAQV/Requimte, University of Porto, Porto, Portugal
| |
Collapse
|
24
|
Solving the microheterogeneity of Bothrops asper myotoxin-II by high-resolution mass spectrometry: Insights into C-terminal region variability in Lys49-phospholipase A2 homologs. Toxicon 2022; 210:123-131. [DOI: 10.1016/j.toxicon.2022.02.024] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Revised: 02/27/2022] [Accepted: 02/28/2022] [Indexed: 11/21/2022]
|
25
|
Proleón A, Torrejón D, Urra FA, Lazo F, López-Torres C, Fuentes-Retamal S, Quispe E, Bautista L, Agurto A, Gavilan RG, Sandoval GA, Rodríguez E, Sánchez EF, Yarlequé A, Vivas-Ruiz DE. Functional, immunological characterization, and anticancer activity of BaMtx: A new Lys49- PLA 2 homologue isolated from the venom of Peruvian Bothrops atrox snake (Serpentes: Viperidae). Int J Biol Macromol 2022; 206:990-1002. [PMID: 35321814 DOI: 10.1016/j.ijbiomac.2022.03.111] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2021] [Accepted: 03/17/2022] [Indexed: 12/13/2022]
Abstract
Bothorps atrox is responsible for most of the ophidism cases in Perú. As part of the envenoming, myotoxicity is one of the most recurrent and destructive effects. In this study, a myotoxin, named BaMtx, was purified from B. atrox venom to elucidate its biological, immunological, and molecular characteristics. BaMtx was purified using CM-Sephadex-C-25 ion-exchange resin and SDS-PAGE analysis showed a unique protein band of 13 kDa or 24 kDa under reducing or non-reducing conditions, respectively. cDNA sequence codified a 122-aa mature protein with high homology with other Lys49-PLA2s; modeled structure showed a N-terminal helix, a β-wing region, and a C-terminal random coil. This protein has a poor phospholipase A2 enzymatic activity. BaMtx has myotoxic (DMM = 12.30 ± 0.95 μg) and edema-forming (DEM = 26.00 ± 1.15 μg) activities. Rabbit immunization with purified enzyme produced anti-BaMtx antibodies that reduced 50.28 ± 10.15% of myotoxic activity and showed significant cross-reactivity against B. brazili and B pictus venoms. On the other hand, BaMtx exhibits mild anti-proliferative and anti-migratory effects on breast cancer cells, affecting the ROS and NADH levels, which may reduce mitochondrial respiration. These results contribute to the understanding of B. atrox Lys49-PLA2 effects and establish the anticancer potential de BaMtx.
Collapse
Affiliation(s)
- Alex Proleón
- Laboratorio de Biología Molecular, Facultad de Ciencias Biológicas, Universidad Nacional Mayor de San Marcos, Av. Venezuela Cdra 34 S/N, Ciudad Universitaria, Lima 01, Perú
| | - Daniel Torrejón
- Laboratorio de Biología Molecular, Facultad de Ciencias Biológicas, Universidad Nacional Mayor de San Marcos, Av. Venezuela Cdra 34 S/N, Ciudad Universitaria, Lima 01, Perú
| | - Felix A Urra
- Laboratorio de Plasticidad Metabólica y Bioenergética, Programa de Farmacología Clínica y Molecular, Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Santiago 8380453, Chile
| | - Fanny Lazo
- Laboratorio de Biología Molecular, Facultad de Ciencias Biológicas, Universidad Nacional Mayor de San Marcos, Av. Venezuela Cdra 34 S/N, Ciudad Universitaria, Lima 01, Perú
| | - Camila López-Torres
- Laboratorio de Plasticidad Metabólica y Bioenergética, Programa de Farmacología Clínica y Molecular, Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Santiago 8380453, Chile
| | - Sebastián Fuentes-Retamal
- Laboratorio de Plasticidad Metabólica y Bioenergética, Programa de Farmacología Clínica y Molecular, Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Santiago 8380453, Chile
| | - Edwin Quispe
- Laboratorio de Biología Molecular, Facultad de Ciencias Biológicas, Universidad Nacional Mayor de San Marcos, Av. Venezuela Cdra 34 S/N, Ciudad Universitaria, Lima 01, Perú
| | - Lorgio Bautista
- Laboratorio de Biología Molecular, Facultad de Ciencias Biológicas, Universidad Nacional Mayor de San Marcos, Av. Venezuela Cdra 34 S/N, Ciudad Universitaria, Lima 01, Perú
| | - Andrés Agurto
- Laboratorio de Biología Molecular, Facultad de Ciencias Biológicas, Universidad Nacional Mayor de San Marcos, Av. Venezuela Cdra 34 S/N, Ciudad Universitaria, Lima 01, Perú
| | - Ronnie G Gavilan
- Centro Nacional de Salud Pública, Instituto Nacional de Salud-Perú, Jesús María, Lima, Peru; Escuela Profesional de Medicina Humana, Universidad Privada San Juan Bautista, Lima, Peru
| | - Gustavo A Sandoval
- Laboratorio de Biología Molecular, Facultad de Ciencias Biológicas, Universidad Nacional Mayor de San Marcos, Av. Venezuela Cdra 34 S/N, Ciudad Universitaria, Lima 01, Perú
| | - Edith Rodríguez
- Laboratorio de Biología Molecular, Facultad de Ciencias Biológicas, Universidad Nacional Mayor de San Marcos, Av. Venezuela Cdra 34 S/N, Ciudad Universitaria, Lima 01, Perú
| | - Eladio F Sánchez
- Research and Development Center, Ezequiel Dias Foundation, 30510-010 Belo Horizonte, MG, Brazil
| | - Armando Yarlequé
- Laboratorio de Biología Molecular, Facultad de Ciencias Biológicas, Universidad Nacional Mayor de San Marcos, Av. Venezuela Cdra 34 S/N, Ciudad Universitaria, Lima 01, Perú
| | - Dan E Vivas-Ruiz
- Laboratorio de Biología Molecular, Facultad de Ciencias Biológicas, Universidad Nacional Mayor de San Marcos, Av. Venezuela Cdra 34 S/N, Ciudad Universitaria, Lima 01, Perú.
| |
Collapse
|
26
|
Soares BS, Rocha SLG, Bastos VA, Lima DB, Carvalho PC, Gozzo FC, Demeler B, Williams TL, Arnold J, Henrickson A, Jørgensen TJD, Souza TACB, Perales J, Valente RH, Lomonte B, Gomes-Neto F, Neves-Ferreira AGC. Molecular Architecture of the Antiophidic Protein DM64 and its Binding Specificity to Myotoxin II From Bothrops asper Venom. Front Mol Biosci 2022; 8:787368. [PMID: 35155563 PMCID: PMC8830425 DOI: 10.3389/fmolb.2021.787368] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Accepted: 12/07/2021] [Indexed: 01/11/2023] Open
Abstract
DM64 is a toxin-neutralizing serum glycoprotein isolated from Didelphis aurita, an ophiophagous marsupial naturally resistant to snake envenomation. This 64 kDa antitoxin targets myotoxic phospholipases A2, which account for most local tissue damage of viperid snakebites. We investigated the noncovalent complex formed between native DM64 and myotoxin II, a myotoxic phospholipase-like protein from Bothrops asper venom. Analytical ultracentrifugation (AUC) and size exclusion chromatography indicated that DM64 is monomeric in solution and binds equimolar amounts of the toxin. Attempts to crystallize native DM64 for X-ray diffraction were unsuccessful. Obtaining recombinant protein to pursue structural studies was also challenging. Classical molecular modeling techniques were impaired by the lack of templates with more than 25% sequence identity with DM64. An integrative structural biology approach was then applied to generate a three-dimensional model of the inhibitor bound to myotoxin II. I-TASSER individually modeled the five immunoglobulin-like domains of DM64. Distance constraints generated by cross-linking mass spectrometry of the complex guided the docking of DM64 domains to the crystal structure of myotoxin II, using Rosetta. AUC, small-angle X-ray scattering (SAXS), molecular modeling, and molecular dynamics simulations indicated that the DM64-myotoxin II complex is structured, shows flexibility, and has an anisotropic shape. Inter-protein cross-links and limited hydrolysis analyses shed light on the inhibitor's regions involved with toxin interaction, revealing the critical participation of the first, third, and fifth domains of DM64. Our data showed that the fifth domain of DM64 binds to myotoxin II amino-terminal and beta-wing regions. The third domain of the inhibitor acts in a complementary way to the fifth domain. Their binding to these toxin regions presumably precludes dimerization, thus interfering with toxicity, which is related to the quaternary structure of the toxin. The first domain of DM64 interacts with the functional site of the toxin putatively associated with membrane anchorage. We propose that both mechanisms concur to inhibit myotoxin II toxicity by DM64 binding. The present topological characterization of this toxin-antitoxin complex constitutes an essential step toward the rational design of novel peptide-based antivenom therapies targeting snake venom myotoxins.
Collapse
Affiliation(s)
- Barbara S. Soares
- Laboratory of Toxinology, Oswaldo Cruz Institute, Rio de Janeiro, Brazil
| | | | - Viviane A. Bastos
- Laboratory of Toxinology, Oswaldo Cruz Institute, Rio de Janeiro, Brazil
| | - Diogo B. Lima
- Department of Chemical Biology, Leibniz Forschungsinstitut für Molekulare Pharmakologie (FMP), Berlin, Germany
| | - Paulo C. Carvalho
- Laboratory for Structural and Computational Proteomics, Carlos Chagas Institute, Curitiba, Brazil
| | - Fabio C. Gozzo
- Dalton Mass Spectrometry Laboratory, University of Campinas, Campinas, Brazil
| | - Borries Demeler
- Department of Biochemistry and Structural Biology, University of Texas Health Science Center at San Antonio, San Antonio, TX, United States
- Department of Chemistry and Biochemistry, University of Lethbridge, Lethbridge, AB, Canada
- Department of Chemistry and Biochemistry, University of Montana, Missoula, MT, United States
| | - Tayler L. Williams
- Department of Biochemistry and Structural Biology, University of Texas Health Science Center at San Antonio, San Antonio, TX, United States
| | - Janelle Arnold
- Department of Environmental Science, Princeton University, Princeton, NJ, United States
| | - Amy Henrickson
- Department of Chemistry and Biochemistry, University of Lethbridge, Lethbridge, AB, Canada
| | - Thomas J. D. Jørgensen
- Department of Biochemistry and Molecular Biology, University of Southern Denmark, Odense, Denmark
| | - Tatiana A. C. B. Souza
- Laboratory for Structural and Computational Proteomics, Carlos Chagas Institute, Curitiba, Brazil
| | - Jonas Perales
- Laboratory of Toxinology, Oswaldo Cruz Institute, Rio de Janeiro, Brazil
| | - Richard H. Valente
- Laboratory of Toxinology, Oswaldo Cruz Institute, Rio de Janeiro, Brazil
| | - Bruno Lomonte
- Clodomiro Picado Institute, University of Costa Rica, San José, Costa Rica
| | | | | |
Collapse
|
27
|
Ferreira SDS, Silva DPD, Torres-Rêgo M, Silva-Júnior AAD, Fernandes-Pedrosa MDF. The potential of phenolic acids in therapy against snakebites: A review. Toxicon 2021; 208:1-12. [PMID: 34979199 DOI: 10.1016/j.toxicon.2021.12.019] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2021] [Revised: 12/06/2021] [Accepted: 12/27/2021] [Indexed: 02/07/2023]
Abstract
Ophidism is a serious health problem worldwide and is included in the World Health Organization's (WHO's) list of Neglected Tropical Diseases. Although snakebite envenoming requires emergency treatment, currently the only treatment recommended by WHO is serotherapy, which has some disadvantages such as low access to the rural population, low effectiveness in neutralizing local effects, and high cost. In this context, new alternatives for the treatment of snakebites are required. The use of plant-derived compounds to inhibit the effects caused by snake venoms has been the object of a number of studies in recent years. This review aims to provide an up-to-date overview of the use of phenolic acids with therapeutic application against envenomation by snakes of different species. In this sense, structural analysis in silico and biological activities in vivo and in vitro were reported. The acids were subdivided into derivatives of benzoic and cinnamic acids, with derivatives of cinnamic acids being the most studied. Studies have revealed that these compounds are capable of inhibiting local and systemic effects induced by envenomation, and structural analyses indicate that the acids interact with important sites responsible for the action of toxins. Thus, it was reported that phenolic acids showed antiophidic potential, providing insights for future research to develop complementary drugs for the treatment of snakebites.
Collapse
Affiliation(s)
- Sarah de Sousa Ferreira
- Laboratory of Technology and Pharmaceutical Biotechnology (Tecbiofar), College of Pharmacy, Federal University of Rio Grande do Norte, Avenue General Gustavo Cordeiro de Farias, S/N, Petrópolis, Natal, 59012-570, Brazil.
| | - Diana Pontes da Silva
- Laboratory of Technology and Pharmaceutical Biotechnology (Tecbiofar), College of Pharmacy, Federal University of Rio Grande do Norte, Avenue General Gustavo Cordeiro de Farias, S/N, Petrópolis, Natal, 59012-570, Brazil.
| | - Manoela Torres-Rêgo
- Laboratory of Technology and Pharmaceutical Biotechnology (Tecbiofar), College of Pharmacy, Federal University of Rio Grande do Norte, Avenue General Gustavo Cordeiro de Farias, S/N, Petrópolis, Natal, 59012-570, Brazil; Graduate Program of Chemistry, Chemistry Institute, Federal University of Rio Grande do Norte, Avenue Senador Salgado Filho, 3000, Lagoa Nova, Natal, 59072-970, Brazil.
| | - Arnóbio Antônio da Silva-Júnior
- Laboratory of Technology and Pharmaceutical Biotechnology (Tecbiofar), College of Pharmacy, Federal University of Rio Grande do Norte, Avenue General Gustavo Cordeiro de Farias, S/N, Petrópolis, Natal, 59012-570, Brazil.
| | - Matheus de Freitas Fernandes-Pedrosa
- Laboratory of Technology and Pharmaceutical Biotechnology (Tecbiofar), College of Pharmacy, Federal University of Rio Grande do Norte, Avenue General Gustavo Cordeiro de Farias, S/N, Petrópolis, Natal, 59012-570, Brazil.
| |
Collapse
|
28
|
Lessons from a Single Amino Acid Substitution: Anticancer and Antibacterial Properties of Two Phospholipase A2-Derived Peptides. Curr Issues Mol Biol 2021; 44:46-62. [PMID: 35723383 PMCID: PMC8929095 DOI: 10.3390/cimb44010004] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2021] [Revised: 12/13/2021] [Accepted: 12/15/2021] [Indexed: 12/15/2022] Open
Abstract
The membrane-active nature of phospholipase A2-derived peptides makes them potential candidates for antineoplastic and antibacterial therapies. Two short 13-mer C-terminal fragments taken from snake venom Lys49-PLA2 toxins (p-AppK and p-Acl), differing by a leucine/phenylalanine substitution, were synthesized and their bioactivity was evaluated. Their capacity to interfere with the survival of Gram-positive and Gram-negative bacteria as well as with solid and liquid tumors was assessed in vitro. Toxicity to red blood cells was investigated via in silico and in vitro techniques. The mode of action was mainly studied by molecular dynamics simulations and membrane permeabilization assays. Briefly, both peptides have dual activity, i.e., they act against both bacteria, including multidrug-resistant strains and tumor cells. All tested bacteria were susceptible to both peptides, Pseudomonas aeruginosa being the most affected. RAMOS, K562, NB4, and CEM cells were the main leukemic targets of the peptides. In general, p-Acl showed more significant activity, suggesting that phenylalanine confers advantages to the antibacterial and antitumor mechanism, particularly for osteosarcoma lines (HOS and MG63). Peptide-based treatment increased the uptake of a DNA-intercalating dye by bacteria, suggesting membrane damage. Indeed, p-AppK and p-Acl did not disrupt erythrocyte membranes, in agreement with in silico predictions. The latter revealed that the peptides deform the membrane and increase its permeability by facilitating solvent penetration. This phenomenon is expected to catalyze the permeation of solutes that otherwise could not cross the hydrophobic membrane core. In conclusion, the present study highlights the role of a single amino acid substitution present in natural sequences towards the development of dual-action agents. In other words, dissecting and fine-tuning biomembrane remodeling proteins, such as snake venom phospholipase A2 isoforms, is again demonstrated as a valuable source of therapeutic peptides.
Collapse
|
29
|
Antiprotozoal Effect of Snake Venoms and Their Fractions: A Systematic Review. Pathogens 2021; 10:pathogens10121632. [PMID: 34959587 PMCID: PMC8707848 DOI: 10.3390/pathogens10121632] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2021] [Revised: 12/08/2021] [Accepted: 12/13/2021] [Indexed: 11/17/2022] Open
Abstract
Background: Protozoal infection is a lingering public health issue of great concern, despite efforts to produce drugs and vaccines against it. Recent breakthrough research has discovered alternative antiprotozoal agents encompassing the use of snake venoms and their components to cure these infections. This study collated the existing literature to examine the antiprotozoal effect of snake venoms and their fractions. Methods: We conducted a systematic review following the PRISMA guidelines. The PubMed and Embase databases were searched from their inception until 13 October 2021. Articles were screened at the title, abstract and full-text phases. Some additional studies were obtained through the manual search process. Results: We identified 331 studies via the electronic database and manual searches, of which 55 reporting the antiprotozoal effect of snake venoms and their components were included in the review. Around 38% of studies examined the effect of whole crude venoms, and a similar percentage evaluated the effect of a proportion of enzymatic phospholipase A2 (PLA2). In particular, this review reports around 36 PLA2 activities and 29 snake crude venom activities. We also report the notable phenomenon of synergism with PLA2 isoforms of Bothrops asper. Importantly, limited attention has been given so far to the antiprotozoal efficacies of metalloproteinase, serine protease and three-finger toxins, although these venom components have been identified as significant components of the dominant venom families. Conclusion: This study highlights the impact of snake venoms and their fractions on controlling protozoal infections and suggests the need to examine further the effectiveness of other venom components, such as metalloproteinase, serine protease and three-finger toxins. Future research questions in this field must be redirected toward synergism in snake venom components, based on pharmacological usage and in the context of toxicology. Ascertaining the effects of snake venoms and their components on other protozoal species that have not yet been studied is imperative.
Collapse
|
30
|
Bernarde PS, Pucca MB, Mota-da-Silva A, da Fonseca WL, de Almeida MRN, de Oliveira IS, Cerni FA, Gobbi Grazziotin F, Sartim MA, Sachett J, Wen FH, Moura-da-Silva AM, Monteiro WM. Bothrops bilineatus: An Arboreal Pitviper in the Amazon and Atlantic Forest. Front Immunol 2021; 12:778302. [PMID: 34975866 PMCID: PMC8714932 DOI: 10.3389/fimmu.2021.778302] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2021] [Accepted: 11/25/2021] [Indexed: 11/13/2022] Open
Abstract
The two-striped forest-pitviper (Bothrops bilineatus) is an arboreal snake that is currently represented by two subspecies (B. b. bilineatus and B. b. smaragdinus) that comprise a species complex, and its distribution is in the Amazon and the Atlantic Forest. The rarity of encounters with this snake is reflected in the low occurrence of cases of snakebites throughout its geographic distribution and the resulting low number of published clinical reports. However, in some areas, B. bilineatus proves to be more frequent and causes envenomations in a greater proportion. Herein, we review the main aspects of the species complex B. bilineatus, including its biology, ecology, taxonomy, morphology, genetic and molecular studies, geographic distribution, conservation status, venom, pathophysiology and clinical aspects, and epidemiology. In addition, the different antivenoms available for the treatment of envenomations caused by B. bilineatus are presented along with suggestions for future studies that are needed for a better understanding of the snakebites caused by this snake.
Collapse
Affiliation(s)
- Paulo Sérgio Bernarde
- Laboratório de Herpetologia, Universidade Federal do Acre, Cruzeiro do Sul, Brazil
- *Correspondence: Paulo Sérgio Bernarde, ; Wuelton M. Monteiro,
| | - Manuela Berto Pucca
- Curso de Medicina, Universidade Federal de Roraima, Boa Vista, Brazil
- Programa de Pós-graduação em Ciências da Saúde, Universidade Federal de Roraima, Boa Vista, Brazil
| | | | | | | | - Isadora Sousa de Oliveira
- Departamento de Ciências Biomoleculares, Faculdade de Ciências Farmacêuticas de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, Brazil
| | - Felipe Augusto Cerni
- Programa de Pós-graduação em Ciências da Saúde, Universidade Federal de Roraima, Boa Vista, Brazil
| | | | - Marco A. Sartim
- Departamento de Ensino e Pesquisa, Fundação de Medicina Tropical Dr. Heitor Vieira Dourado, Manaus, Brazil
- Instituto de Ciências Biológicas, Universidade Federal do Amazonas, Manaus, Brazil
- Departamento de Pós-Graduação, Universidade Nilton Lins, Manaus, Brazil
| | - Jacqueline Sachett
- Escola Superior de Ciências da Saúde, Universidade do Estado do Amazonas, Manaus, Brazil
- Departamento de Ensino e Pesquisa, Fundação Alfredo da Matta, Manaus, Brazil
| | - Fan Hui Wen
- Núcleo Estratégico de Venenos e Antivenenos, Instituto Butantan, São Paulo, Brazil
| | - Ana Maria Moura-da-Silva
- Departamento de Ensino e Pesquisa, Fundação de Medicina Tropical Dr. Heitor Vieira Dourado, Manaus, Brazil
- Laboratório de Imunopatologia, Instituto Butantan, São Paulo, Brazil
| | - Wuelton M. Monteiro
- Departamento de Ensino e Pesquisa, Fundação de Medicina Tropical Dr. Heitor Vieira Dourado, Manaus, Brazil
- Escola Superior de Ciências da Saúde, Universidade do Estado do Amazonas, Manaus, Brazil
- *Correspondence: Paulo Sérgio Bernarde, ; Wuelton M. Monteiro,
| |
Collapse
|
31
|
Ogawa T, Tobishima Y, Kamata S, Matsuda Y, Muramoto K, Hidaka M, Futai E, Kuraishi T, Yokota S, Ohno M, Hattori S. Focused Proteomics Analysis of Habu Snake ( Protobothrops flavoviridis) Venom Using Antivenom-Based Affinity Chromatography Reveals Novel Myonecrosis-Enhancing Activity of Thrombin-Like Serine Proteases. Front Pharmacol 2021; 12:766406. [PMID: 34803710 PMCID: PMC8599580 DOI: 10.3389/fphar.2021.766406] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2021] [Accepted: 10/18/2021] [Indexed: 11/13/2022] Open
Abstract
Snakebites are one of the major causes of death and long-term disability in the developing countries due to the presence of various bioactive peptides and proteins in snake venom. In Japan, the venom of the habu snake (Protobothrops flavoviridis) causes severe permanent damage due to its myonecrotic toxins. Antivenom immunoglobulins are an effective therapy for snakebites, and antivenom was recently developed with effective suppressive activity against myonecrosis induced by snake venom. To compare the properties of an antivenom having anti-myonecrotic activity with those of conventional antivenom with no anti-myonecrotic activity, this study applied focused proteomics analysis of habu venom proteins using 2D gel electrophoresis. As a target protein for antivenom immunoglobulins with anti-myonecrotic activity, we identified a thrombin-like serine protease, TLSP2 (TLf2), which was an inactive proteolytic isoform due to the replacement of the active site, His43 with Arg. Additionally, we identified the unique properties and a novel synergistic function of pseudoenzyme TLf2 as a myonecrosis-enhancing factor. To our knowledge, this is the first report of a function of a catalytically inactive snake serine protease.
Collapse
Affiliation(s)
- Tomohisa Ogawa
- Laboratory of Enzymology, Graduate School of Agricultural Science, Tohoku University, Sendai, Japan.,Graduate School of Life Sciences, Tohoku University, Sendai, Japan
| | - Yu Tobishima
- Graduate School of Life Sciences, Tohoku University, Sendai, Japan
| | - Shizuka Kamata
- Graduate School of Life Sciences, Tohoku University, Sendai, Japan
| | - Youhei Matsuda
- Graduate School of Life Sciences, Tohoku University, Sendai, Japan
| | - Koji Muramoto
- Graduate School of Life Sciences, Tohoku University, Sendai, Japan
| | - Masafumi Hidaka
- Laboratory of Enzymology, Graduate School of Agricultural Science, Tohoku University, Sendai, Japan
| | - Eugene Futai
- Laboratory of Enzymology, Graduate School of Agricultural Science, Tohoku University, Sendai, Japan
| | - Takeshi Kuraishi
- Institute of Medical Science, University of Tokyo, Kagoshima, Japan
| | - Shinichi Yokota
- Institute of Medical Science, University of Tokyo, Kagoshima, Japan
| | - Motonori Ohno
- Department of Applied Life Science, Faculty of Bioscience and Biotechnology, Sojo University, Kumamoto, Japan
| | - Shosaku Hattori
- Institute of Medical Science, University of Tokyo, Kagoshima, Japan
| |
Collapse
|
32
|
Cavalcante JDS, Nogueira Júnior FA, Bezerra Jorge RJ, Almeida C. Pain modulated by Bothrops snake venoms: Mechanisms of nociceptive signaling and therapeutic perspectives. Toxicon 2021; 201:105-114. [PMID: 34425141 DOI: 10.1016/j.toxicon.2021.08.016] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2021] [Revised: 08/11/2021] [Accepted: 08/16/2021] [Indexed: 12/17/2022]
Abstract
Snake venoms are substances mostly composed by proteins and peptides with high biological activity. Local and systemic effects culminate in clinical manifestations induced by these substances. Pain is the most uncomfortable condition, but it has not been well investigated. This review discusses Bothrops snakebite-induced nociception, highlighting molecules involved in the mediation of this process and perspectives in treatment of pain induced by Bothrops snake venoms (B. alternatus, B. asper, B. atrox, B. insularis, B. jararaca, B. pirajai, B. jararacussu, B. lanceolatus, B. leucurus, B. mattogrossensis, B. moojeni). We highlight, the understanding of the nociceptive signaling, especially in snakebite, enables more efficient treatment approaches. Finally, future perspectives for pain treatment concerning snakebite patients are discussed.
Collapse
Affiliation(s)
- Joeliton Dos Santos Cavalcante
- Graduate Program in Tropical Diseases, Botucatu Medical School (FMB), São Paulo State University, Botucatu, São Paulo, Brazil.
| | - Francisco Assis Nogueira Júnior
- Department of Physiology and Pharmacology and Drug Research and Development Center Federal University of Ceará, Fortaleza, Ceará, Brazil
| | - Roberta Jeane Bezerra Jorge
- Department of Physiology and Pharmacology and Drug Research and Development Center Federal University of Ceará, Fortaleza, Ceará, Brazil
| | - Cayo Almeida
- Center of Mathematics, Computing Sciences and Cognition, Federal University of ABC, São Paulo, Brazil.
| |
Collapse
|
33
|
Puzari U, Fernandes PA, Mukherjee AK. Advances in the Therapeutic Application of Small-Molecule Inhibitors and Repurposed Drugs against Snakebite. J Med Chem 2021; 64:13938-13979. [PMID: 34565143 DOI: 10.1021/acs.jmedchem.1c00266] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The World Health Organization has declared snakebite as a neglected tropical disease. Antivenom administration is the sole therapy against venomous snakebite; however, several limitations of this therapy reinforce the dire need for an alternative and/or additional treatment against envenomation. Inhibitors against snake venoms have been explored from natural resources and are synthesized in the laboratory; however, repurposing of small-molecule therapeutics (SMTs) against the principal toxins of snake venoms to inhibit their lethality and/or obnoxious effect of envenomation has been garnering greater attention owing to their established pharmacokinetic properties, low-risk attributes, cost-effectiveness, ease of administration, and storage stability. Nevertheless, SMTs are yet to be approved and commercialized for snakebite treatment. Therefore, we have systematically reviewed and critically analyzed the scenario of small synthetic inhibitors and repurposed drugs against snake envenomation from 2005 to date and proposed novel approaches and commercialization strategies for the development of efficacious therapies against snake envenomation.
Collapse
Affiliation(s)
- Upasana Puzari
- Department of Molecular Biology and Biotechnology, School of Sciences, Tezpur University, Tezpur-784028, Assam, India
| | - Pedro Alexandrino Fernandes
- LAQV@REQUIMTE, Departamento de Química e Bioquímica, Faculdade de Ciências, Universidade do Porto, Rua Do Campo Alegre S/N, 4169-007 Porto, Portugal
| | - Ashis K Mukherjee
- Department of Molecular Biology and Biotechnology, School of Sciences, Tezpur University, Tezpur-784028, Assam, India.,Institute of Advanced Study in Science and Technology, Vigyan Path Garchuk, Paschim Boragaon, Guwahati-781035, Assam, India
| |
Collapse
|
34
|
López-Dávila AJ, Weber N, Kraft T, Matinmehr F, Arias-Hidalgo M, Fernández J, Lomonte B, Gutiérrez JM. Cytotoxicity of snake venom Lys49 PLA2-like myotoxin on rat cardiomyocytes ex vivo does not involve a direct action on the contractile apparatus. Sci Rep 2021; 11:19452. [PMID: 34593882 PMCID: PMC8484475 DOI: 10.1038/s41598-021-98594-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2021] [Accepted: 09/07/2021] [Indexed: 11/09/2022] Open
Abstract
Viperid snake venoms contain a unique family of cytotoxic proteins, the Lys49 PLA2 homologs, which are devoid of enzymatic activity but disrupt the integrity of cell membranes. They are known to induce skeletal muscle damage and are therefore named 'myotoxins'. Single intact and skinned (devoid of membranes and cytoplasm but with intact sarcomeric proteins) rat cardiomyocytes were used to analyze the cytotoxic action of a myotoxin, from the venom of Bothrops asper. The toxin induced rapid hypercontraction of intact cardiomyocytes, associated with an increase in the cytosolic concentration of calcium and with cell membrane disruption. Hypercontraction of intact cardiomyocytes was abrogated by the myosin inhibitor para-aminoblebbistatin (AmBleb). No toxin-induced changes of key parameters of force development were observed in skinned cardiomyocytes. Thus, although myosin is a key effector of the observed hypercontraction, a direct effect of the toxin on the sarcomeric proteins -including the actomyosin complex- is not part of the mechanism of cytotoxicity. Owing to the sensitivity of intact cardiomyocytes to the cytotoxic action of myotoxin, this ex vivo model is a valuable tool to explore in further detail the mechanism of action of this group of snake venom toxins.
Collapse
Affiliation(s)
| | - Natalie Weber
- Institute of Molecular and Cell Physiology, Hannover Medical School, 30625, Hannover, Germany
- Institute of Molecular and Translational Therapeutic Strategies, Hannover Medical School, 30625, Hannover, Germany
| | - Theresia Kraft
- Institute of Molecular and Cell Physiology, Hannover Medical School, 30625, Hannover, Germany
| | - Faramarz Matinmehr
- Institute of Molecular and Cell Physiology, Hannover Medical School, 30625, Hannover, Germany
| | - Mariela Arias-Hidalgo
- Departamento de Fisiología, Escuela de Medicina, Universidad de Costa Rica, San José, 11501, Costa Rica
| | - Julián Fernández
- Instituto Clodomiro Picado, Facultad de Microbiología, Universidad de Costa Rica, San José, 11501, Costa Rica
| | - Bruno Lomonte
- Instituto Clodomiro Picado, Facultad de Microbiología, Universidad de Costa Rica, San José, 11501, Costa Rica
| | - José María Gutiérrez
- Instituto Clodomiro Picado, Facultad de Microbiología, Universidad de Costa Rica, San José, 11501, Costa Rica
| |
Collapse
|
35
|
Teixeira SC, da Silva MS, Gomes AAS, Moretti NS, Lopes DS, Ferro EAV, Rodrigues VDM. Panacea within a Pandora's box: the antiparasitic effects of phospholipases A 2 (PLA 2s) from snake venoms. Trends Parasitol 2021; 38:80-94. [PMID: 34364805 DOI: 10.1016/j.pt.2021.07.004] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2021] [Revised: 07/07/2021] [Accepted: 07/08/2021] [Indexed: 10/20/2022]
Abstract
Parasitic diseases affect millions of individuals worldwide, mainly in low-income regions. There is no cure for most of these diseases, and the treatment relies on drugs that have side effects and lead to drug resistance, emphasizing the urgency to find new treatments. Snake venom has been gaining prominence as a rich source of molecules with antiparasitic potentials, such as phospholipases A2 (PLA2s). Here, we compile the findings involving PLA2s with antiparasitic activities against helminths, Plasmodium, Toxoplasma, and trypanosomatids. We indicate their molecular features, highlighting the possible antiparasitic mechanisms of action of these proteins. We also demonstrate interactions between PLA2s and some parasite membrane components, shedding light on potential targets for drug design that may provide better treatment for the illnesses caused by parasites.
Collapse
Affiliation(s)
- Samuel Cota Teixeira
- Department of Immunology, Institute of Biomedical Science, Federal University of Uberlândia, Uberlândia (UFU), MG, Brazil.
| | - Marcelo Santos da Silva
- DNA Replication and Repair Laboratory (DRRL), Department of Chemical and Biological Sciences, Biosciences Institute, São Paulo State University (UNESP), Botucatu, SP, Brazil
| | | | - Nilmar Silvio Moretti
- Laboratório de Biologia Molecular de Patógenos (LBMP), Departamento de Microbiologia, Imunologia e Parasitologia, Escola Paulista de Medicina, Universidade Federal de São Paulo (UNIFESP), São Paulo, SP, Brazil
| | - Daiana Silva Lopes
- Multidisciplinary Institute of Health, Anísio Teixeira Campus, Federal University of Bahia (UFBA), Vitória da Conquista, BA, Brazil
| | - Eloisa Amália Vieira Ferro
- Department of Immunology, Institute of Biomedical Science, Federal University of Uberlândia, Uberlândia (UFU), MG, Brazil
| | - Veridiana de Melo Rodrigues
- Laboratory of Biochemistry and Animal Toxins, Institute of Biotechnology, Federal University of Uberlândia (UFU), Uberlândia, MG, Brazil.
| |
Collapse
|
36
|
Simões-Silva R, Alfonso JJ, Gómez AF, Sobrinho JC, Kayano AM, de Medeiros DSS, Teles CBG, Quintero A, Fuly AL, Gómez CV, Pereira SS, da Silva SL, Stábeli RG, Soares AM. Synergism of in vitro plasmodicidal activity of phospholipase A2 isoforms isolated from panamanian Bothrops asper venom. Chem Biol Interact 2021; 346:109581. [PMID: 34302801 DOI: 10.1016/j.cbi.2021.109581] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2021] [Revised: 06/11/2021] [Accepted: 07/16/2021] [Indexed: 10/20/2022]
Abstract
Bothrops asper is one of the most important snake species in Central America, mainly because of its medical importance in countries like Ecuador, Panama and Costa Rica, where this species causes a high number of snakebite accidents. Several basic phospholipases A2 (PLA2s) have been previously characterized from B. asper venom, but few studies have been carried out with its acidic isoforms. In addition, since snake venom is a rich source of bioactive substances, it is necessary to investigate the biotechnological potential of its components. In this context, this study aimed to carry out the biochemical characterization of PLA2 isoforms isolated from B. asper venom and to evaluate the antiparasitic potential of these toxins. The venom and key fractions were subjected to different chromatographic steps, obtaining nine PLA2s, four acidic ones (BaspAc-I, BaspAc-II, BaspAc-III and BaspAc-IV) and five basic ones (BaspB-I, BaspB-II, BaspB-III, BaspB-IV and BaspB-V). The isoelectric points of the acidic PLA2s were also determined, which presented values ranging between 4.5 and 5. The findings indicated the isolation of five unpublished isoforms, four Asp49-PLA, corresponding to the group of acidic isoforms, and one Lys49-PLA2-like. Acidic PLA2s catalyzed the degradation of all substrates evaluated; however, for the basic PLA2s, there was a preference for phosphatidylglycerol and phosphatidic acid. The antiparasitic potential of the toxins was evaluated, and the acidic PLA2s demonstrated action against the epimastigote forms of T. cruzi and promastigote forms of L. infantum, while the basic PLA2s BaspB-II and BaspB-IV showed activity against P. falciparum. The results indicated an increase of up to 10 times in antiplasmodial activity, when the Asp49-PLA2 and Lys49-PLA2 were associated with one another, denoting synergistic action between these PLA2 isoforms. These findings correspond to the first report of synergistic antiplasmodial action for svPLA2s, demonstrating that these molecules may be important targets in the search for new antiparasitic agents.
Collapse
Affiliation(s)
- Rodrigo Simões-Silva
- Centro de Estudos de Biomoléculas Aplicadas à Saúde, CEBio, Laboratório de Biotecnologia de Proteínas e Compostos Bioativos da Amazônia Ocidental, LaBioProt, Fundação Oswaldo Cruz, FIOCRUZ, Unidade Rondônia, Porto Velho, RO, Brazil; Instituto Federal de Educação, Ciência e Tecnologia de Rondônia, IFRO, Campus Vilhena, Vilhena, RO, Brazil
| | - Jorge Javier Alfonso
- Centro de Estudos de Biomoléculas Aplicadas à Saúde, CEBio, Laboratório de Biotecnologia de Proteínas e Compostos Bioativos da Amazônia Ocidental, LaBioProt, Fundação Oswaldo Cruz, FIOCRUZ, Unidade Rondônia, Porto Velho, RO, Brazil; Centro Para el Desarrollo de la Investigación Científica (CEDIC), Asunción, Paraguay
| | - Ana F Gómez
- Centro de Estudos de Biomoléculas Aplicadas à Saúde, CEBio, Laboratório de Biotecnologia de Proteínas e Compostos Bioativos da Amazônia Ocidental, LaBioProt, Fundação Oswaldo Cruz, FIOCRUZ, Unidade Rondônia, Porto Velho, RO, Brazil; Centro Para el Desarrollo de la Investigación Científica (CEDIC), Asunción, Paraguay
| | - Juliana C Sobrinho
- Centro de Estudos de Biomoléculas Aplicadas à Saúde, CEBio, Laboratório de Biotecnologia de Proteínas e Compostos Bioativos da Amazônia Ocidental, LaBioProt, Fundação Oswaldo Cruz, FIOCRUZ, Unidade Rondônia, Porto Velho, RO, Brazil
| | - Anderson M Kayano
- Centro de Estudos de Biomoléculas Aplicadas à Saúde, CEBio, Laboratório de Biotecnologia de Proteínas e Compostos Bioativos da Amazônia Ocidental, LaBioProt, Fundação Oswaldo Cruz, FIOCRUZ, Unidade Rondônia, Porto Velho, RO, Brazil; Centro de Pesquisa em Medicina Tropical, CEPEM-SESAU/RO, Porto Velho, RO, Brazil
| | - Daniel S S de Medeiros
- Plataforma de Bioensaios em Malária e Leishmanioses, Fundação Oswaldo Cruz, FIOCRUZ, Unidade Rondônia, Porto Velho, RO, Brazil; Programa de Pós-graduação em Biologia Experimental, Universidade Federal de Rondônia, Porto Velho, RO, Brazil
| | - Carolina B G Teles
- Plataforma de Bioensaios em Malária e Leishmanioses, Fundação Oswaldo Cruz, FIOCRUZ, Unidade Rondônia, Porto Velho, RO, Brazil; Programa de Pós-graduação em Biologia Experimental, Universidade Federal de Rondônia, Porto Velho, RO, Brazil; Instituto Nacional de Ciência e Tecnologia em Epidemiologia da Amazônia Ocidental (INCT-EpiAmO), Brazil
| | - Aristides Quintero
- Centro de Informaciones e Investigaciones Toxicológicas y Químicas Aplicadas (CEIITOXQUIA) and Departamento de Química, FCNYE, Universidad Autónoma de Chiriquí, UNACHI, David, Panama
| | - André L Fuly
- Universidade Federal Fluminense, UFF, Niteroi, RJ, Brazil
| | - Celeste Vega Gómez
- Centro Para el Desarrollo de la Investigación Científica (CEDIC), Asunción, Paraguay
| | - Soraya S Pereira
- Laboratório de Engenharia de Anticorpos, Fundação Oswaldo Cruz, FIOCRUZ, Fiocruz Rondônia, Porto Velho, RO, Brazil
| | - Saulo L da Silva
- Faculty of Chemical Sciences, University of Cuenca, Cuenca, Azuay, Ecuador; LAQV-REQUIMTE, Department of Chemistry and Biochemistry, Faculty of Sciences, University of Porto, Rua do Campo Alegre s/n, Porto, Portugal
| | - Rodrigo G Stábeli
- Fundação Oswaldo Cruz, FIOCRUZ, Plataforma Bi-institucional de Medicina Translacional. Ribeirão Preto, SP, Brazil
| | - Andreimar M Soares
- Centro de Estudos de Biomoléculas Aplicadas à Saúde, CEBio, Laboratório de Biotecnologia de Proteínas e Compostos Bioativos da Amazônia Ocidental, LaBioProt, Fundação Oswaldo Cruz, FIOCRUZ, Unidade Rondônia, Porto Velho, RO, Brazil; Programa de Pós-graduação em Biologia Experimental, Universidade Federal de Rondônia, Porto Velho, RO, Brazil; Instituto Nacional de Ciência e Tecnologia em Epidemiologia da Amazônia Ocidental (INCT-EpiAmO), Brazil; Centro Universitário São Lucas, UniSL, Porto Velho, RO, Brazil.
| |
Collapse
|
37
|
Gutiérrez JM, Albulescu LO, Clare RH, Casewell NR, Abd El-Aziz TM, Escalante T, Rucavado A. The Search for Natural and Synthetic Inhibitors That Would Complement Antivenoms as Therapeutics for Snakebite Envenoming. Toxins (Basel) 2021; 13:451. [PMID: 34209691 PMCID: PMC8309910 DOI: 10.3390/toxins13070451] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2021] [Revised: 06/23/2021] [Accepted: 06/27/2021] [Indexed: 12/28/2022] Open
Abstract
A global strategy, under the coordination of the World Health Organization, is being unfolded to reduce the impact of snakebite envenoming. One of the pillars of this strategy is to ensure safe and effective treatments. The mainstay in the therapy of snakebite envenoming is the administration of animal-derived antivenoms. In addition, new therapeutic options are being explored, including recombinant antibodies and natural and synthetic toxin inhibitors. In this review, snake venom toxins are classified in terms of their abundance and toxicity, and priority actions are being proposed in the search for snake venom metalloproteinase (SVMP), phospholipase A2 (PLA2), three-finger toxin (3FTx), and serine proteinase (SVSP) inhibitors. Natural inhibitors include compounds isolated from plants, animal sera, and mast cells, whereas synthetic inhibitors comprise a wide range of molecules of a variable chemical nature. Some of the most promising inhibitors, especially SVMP and PLA2 inhibitors, have been developed for other diseases and are being repurposed for snakebite envenoming. In addition, the search for drugs aimed at controlling endogenous processes generated in the course of envenoming is being pursued. The present review summarizes some of the most promising developments in this field and discusses issues that need to be considered for the effective translation of this knowledge to improve therapies for tackling snakebite envenoming.
Collapse
Affiliation(s)
- José María Gutiérrez
- Facultad de Microbiología, Instituto Clodomiro Picado, Universidad de Costa Rica, San José 11501, Costa Rica; (T.E.); (A.R.)
| | - Laura-Oana Albulescu
- Centre for Snakebite Research & Interventions, Liverpool School of Tropical Medicine, Liverpool L3 5QA, UK; (L.-O.A.); (R.H.C.); (N.R.C.)
| | - Rachel H. Clare
- Centre for Snakebite Research & Interventions, Liverpool School of Tropical Medicine, Liverpool L3 5QA, UK; (L.-O.A.); (R.H.C.); (N.R.C.)
| | - Nicholas R. Casewell
- Centre for Snakebite Research & Interventions, Liverpool School of Tropical Medicine, Liverpool L3 5QA, UK; (L.-O.A.); (R.H.C.); (N.R.C.)
| | - Tarek Mohamed Abd El-Aziz
- Zoology Department, Faculty of Science, Minia University, El-Minia 61519, Egypt;
- Department of Cellular and Integrative Physiology, University of Texas Health Science Center at San Antonio, San Antonio, TX 78229-3900, USA
| | - Teresa Escalante
- Facultad de Microbiología, Instituto Clodomiro Picado, Universidad de Costa Rica, San José 11501, Costa Rica; (T.E.); (A.R.)
| | - Alexandra Rucavado
- Facultad de Microbiología, Instituto Clodomiro Picado, Universidad de Costa Rica, San José 11501, Costa Rica; (T.E.); (A.R.)
| |
Collapse
|
38
|
Costa TR, Francisco AF, Cardoso FF, Moreira-Dill LS, Fernandes CAH, Gomes AAS, Guimarães CLS, Marcussi S, Pereira PS, Oliveira HC, Fontes MRM, Silva SL, Zuliani JP, Soares AM. Gallic acid anti-myotoxic activity and mechanism of action, a snake venom phospholipase A 2 toxin inhibitor, isolated from the medicinal plant Anacardium humile. Int J Biol Macromol 2021; 185:494-512. [PMID: 34197854 DOI: 10.1016/j.ijbiomac.2021.06.163] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Revised: 05/31/2021] [Accepted: 06/24/2021] [Indexed: 11/17/2022]
Abstract
Snakebite envenoming is the cause of an ongoing health crisis in several regions of the world, particularly in tropical and neotropical countries. This scenario creates an urgent necessity for new practical solutions to address the limitations of current therapies. The current study investigated the isolation, phytochemical characterization, and myotoxicity inhibition mechanism of gallic acid (GA), a myotoxin inhibitor obtained from Anacardium humile. The identification and isolation of GA was achieved by employing analytical chromatographic separation, which exhibited a compound with retention time and nuclear magnetic resonance spectra compatible with GA's commercial standard and data from the literature. GA alone was able to inhibit the myotoxic activity induced by the crude venom of Bothrops jararacussu and its two main myotoxins, BthTX-I and BthTX-II. Circular dichroism (CD), fluorescence spectroscopy (FS), dynamic light scattering (DLS), and interaction studies by molecular docking suggested that GA forms a complex with BthTX-I and II. Surface plasmon resonance (SPR) kinetics assays showed that GA has a high affinity for BthTX-I with a KD of 9.146 × 10-7 M. Taken together, the two-state reaction mode of GA binding to BthTX-I, and CD, FS and DLS assays, suggest that GA is able to induce oligomerization and secondary structure changes for BthTX-I and -II. GA and other tannins have been shown to be effective inhibitors of snake venoms' toxic effects, and herein we demonstrated GA's ability to bind to and inhibit a snake venom PLA2, thus proposing a new mechanism of PLA2 inhibition, and presenting more evidence of GA's potential as an antivenom compound.
Collapse
Affiliation(s)
- Tássia R Costa
- Instituto de Genética e Bioquímica, Universidade Federal de Uberlândia, UFU, Uberlândia, MG, Brazil
| | - Aleff F Francisco
- Departamento de Biofísica e Farmacologia, Instituto de Biociências, Universidade Estadual Paulista, UNESP, Botucatu, SP, Brazil; Laboratório de Biotecnologia de Proteínas e Compostos Bioativos, LABIOPROT, Centro de Estudos de Biomoléculas Aplicadas à Saúde, CEBio, Laboratório de Imunologia Celular Aplicada à Saúde, Fundação Oswaldo Cruz, FIOCRUZ, Unidade Rondônia, Porto Velho, RO, Brazil
| | - Fábio F Cardoso
- Departamento de Biofísica e Farmacologia, Instituto de Biociências, Universidade Estadual Paulista, UNESP, Botucatu, SP, Brazil
| | - Leandro S Moreira-Dill
- Laboratório de Biotecnologia de Proteínas e Compostos Bioativos, LABIOPROT, Centro de Estudos de Biomoléculas Aplicadas à Saúde, CEBio, Laboratório de Imunologia Celular Aplicada à Saúde, Fundação Oswaldo Cruz, FIOCRUZ, Unidade Rondônia, Porto Velho, RO, Brazil
| | - Carlos A H Fernandes
- Departamento de Biofísica e Farmacologia, Instituto de Biociências, Universidade Estadual Paulista, UNESP, Botucatu, SP, Brazil
| | - Antoniel A S Gomes
- Departamento de Biofísica e Farmacologia, Instituto de Biociências, Universidade Estadual Paulista, UNESP, Botucatu, SP, Brazil
| | - César L S Guimarães
- Laboratório de Biotecnologia de Proteínas e Compostos Bioativos, LABIOPROT, Centro de Estudos de Biomoléculas Aplicadas à Saúde, CEBio, Laboratório de Imunologia Celular Aplicada à Saúde, Fundação Oswaldo Cruz, FIOCRUZ, Unidade Rondônia, Porto Velho, RO, Brazil; Instituto Brasileiro do Meio Ambiente e dos Recursos Naturais Renováveis, IBAMA, Porto Velho, RO, Brazil
| | - Silvana Marcussi
- Departamento de Química, Universidade Federal de Lavras, UFLA, Lavras, MG, Brazil
| | | | - Hamine C Oliveira
- Departamento de Biofísica e Farmacologia, Instituto de Biociências, Universidade Estadual Paulista, UNESP, Botucatu, SP, Brazil
| | - Marcos R M Fontes
- Departamento de Biofísica e Farmacologia, Instituto de Biociências, Universidade Estadual Paulista, UNESP, Botucatu, SP, Brazil
| | - Saulo L Silva
- Faculty of Chemical Sciences, University of Cuenca, Cuenca, Azuay, Ecuador; LAQV/Requimte, Faculty of Sciences University of Porto, Porto, Portugal
| | - Juliana P Zuliani
- Laboratório de Biotecnologia de Proteínas e Compostos Bioativos, LABIOPROT, Centro de Estudos de Biomoléculas Aplicadas à Saúde, CEBio, Laboratório de Imunologia Celular Aplicada à Saúde, Fundação Oswaldo Cruz, FIOCRUZ, Unidade Rondônia, Porto Velho, RO, Brazil
| | - Andreimar M Soares
- Laboratório de Biotecnologia de Proteínas e Compostos Bioativos, LABIOPROT, Centro de Estudos de Biomoléculas Aplicadas à Saúde, CEBio, Laboratório de Imunologia Celular Aplicada à Saúde, Fundação Oswaldo Cruz, FIOCRUZ, Unidade Rondônia, Porto Velho, RO, Brazil; Centro Universitário São Lucas, UniSL, Porto Velho, RO, Brazil; Instituto Nacional de Ciência e Tecnologia em Epidemiologia da Amazônia Ocidental (INCT-EpiAmO), Brazil.
| |
Collapse
|
39
|
Giribaldi J, Smith JJ, Schroeder CI. Recent developments in animal venom peptide nanotherapeutics with improved selectivity for cancer cells. Biotechnol Adv 2021; 50:107769. [PMID: 33989705 DOI: 10.1016/j.biotechadv.2021.107769] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2021] [Revised: 05/06/2021] [Accepted: 05/08/2021] [Indexed: 02/07/2023]
Abstract
Animal venoms are a rich source of bioactive peptides that efficiently modulate key receptors and ion channels involved in cellular excitability to rapidly neutralize their prey or predators. As such, they have been a wellspring of highly useful pharmacological tools for decades. Besides targeting ion channels, some venom peptides exhibit strong cytotoxic activity and preferentially affect cancer over healthy cells. This is unlikely to be driven by an evolutionary impetus, and differences in tumor cells and the tumor microenvironment are probably behind the serendipitous selectivity shown by some venom peptides. However, strategies such as bioconjugation and nanotechnologies are showing potential to improve their selectivity and potency, thereby paving the way to efficiently harness new anticancer mechanisms offered by venom peptides. This review aims to highlight advances in nano- and chemotherapeutic tools and prospective anti-cancer drug leads derived from animal venom peptides.
Collapse
Affiliation(s)
- Julien Giribaldi
- Center for Cancer Research, National Cancer Institute, National Institutes of Health, Frederick, MD 21702, USA
| | - Jennifer J Smith
- Center for Cancer Research, National Cancer Institute, National Institutes of Health, Frederick, MD 21702, USA
| | - Christina I Schroeder
- Center for Cancer Research, National Cancer Institute, National Institutes of Health, Frederick, MD 21702, USA.
| |
Collapse
|
40
|
Short Linear Motifs Characterizing Snake Venom and Mammalian Phospholipases A2. Toxins (Basel) 2021; 13:toxins13040290. [PMID: 33923919 PMCID: PMC8073766 DOI: 10.3390/toxins13040290] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2021] [Revised: 04/14/2021] [Accepted: 04/16/2021] [Indexed: 12/12/2022] Open
Abstract
Snake venom phospholipases A2 (PLA2s) have sequences and structures very similar to those of mammalian group I and II secretory PLA2s, but they possess many toxic properties, ranging from the inhibition of coagulation to the blockage of nerve transmission, and the induction of muscle necrosis. The biological properties of these proteins are not only due to their enzymatic activity, but also to protein–protein interactions which are still unidentified. Here, we compare sequence alignments of snake venom and mammalian PLA2s, grouped according to their structure and biological activity, looking for differences that can justify their different behavior. This bioinformatics analysis has evidenced three distinct regions, two central and one C-terminal, having amino acid compositions that distinguish the different categories of PLA2s. In these regions, we identified short linear motifs (SLiMs), peptide modules involved in protein–protein interactions, conserved in mammalian and not in snake venom PLA2s, or vice versa. The different content in the SLiMs of snake venom with respect to mammalian PLA2s may result in the formation of protein membrane complexes having a toxic activity, or in the formation of complexes whose activity cannot be blocked due to the lack of switches in the toxic PLA2s, as the motif recognized by the prolyl isomerase Pin1.
Collapse
|
41
|
Sanchez-Castro EE, Pajuelo-Reyes C, Tejedo R, Soria-Juan B, Tapia-Limonchi R, Andreu E, Hitos AB, Martin F, Cahuana GM, Guerra-Duarte C, de Assis TCS, Bedoya FJ, Soria B, Chávez-Olórtegui C, Tejedo JR. Mesenchymal Stromal Cell-Based Therapies as Promising Treatments for Muscle Regeneration After Snakebite Envenoming. Front Immunol 2021; 11:609961. [PMID: 33633730 PMCID: PMC7902043 DOI: 10.3389/fimmu.2020.609961] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2020] [Accepted: 12/17/2020] [Indexed: 12/16/2022] Open
Abstract
Snakebite envenoming is a global neglected disease with an incidence of up to 2.7 million new cases every year. Although antivenoms are so-far the most effective treatment to reverse the acute systemic effects induced by snakebite envenoming, they have a limited therapeutic potential, being unable to completely neutralize the local venom effects. Local damage, such as dermonecrosis and myonecrosis, can lead to permanent sequelae with physical, social, and psychological implications. The strong inflammatory process induced by snake venoms is associated with poor tissue regeneration, in particular the lack of or reduced skeletal muscle regeneration. Mesenchymal stromal cells (MSCs)-based therapies have shown both anti-inflammatory and pro-regenerative properties. We postulate that using allogeneic MSCs or their cell-free products can induce skeletal muscle regeneration in snakebite victims, improving all the three steps of the skeletal muscle regeneration process, mainly by anti-inflammatory activity, paracrine effects, neovascularization induction, and inhibition of tissue damage, instrumental for microenvironment remodeling and regeneration. Since snakebite envenoming occurs mainly in areas with poor healthcare, we enlist the principles and potential of MSCs-based therapies and discuss regulatory issues, good manufacturing practices, transportation, storage, and related-procedures that could allow the administration of these therapies, looking forward to a safe and cost-effective treatment for a so far unsolved and neglected health problem.
Collapse
Affiliation(s)
| | - Cecilia Pajuelo-Reyes
- Institute of Tropical Diseases, Universidad Nacional Toribio Rodríguez de Mendoza de Amazonas, Chachapoyas, Peru
| | - Rebeca Tejedo
- Faculty of Medicine, Universidad Privada San Juan Bautista, Lima, Peru
| | - Bárbara Soria-Juan
- Department of Molecular Biology and Biochemical Engineering, Universidad Pablo de Olavide, Seville, Spain.,Department of Surgery, Fundación Jiménez Díaz, Unidad de Terapias Avanzadas, Universidad Autónoma de Madrid, Madrid, Spain
| | - Rafael Tapia-Limonchi
- Institute of Tropical Diseases, Universidad Nacional Toribio Rodríguez de Mendoza de Amazonas, Chachapoyas, Peru
| | - Etelvina Andreu
- ISABIAL-Hospital General y Universitario de Alicante, Alicante, Spain.,Departmento de Fisica Aplicadas, University Miguel Hernández, Alicante, Spain
| | - Ana B Hitos
- Department of Cell Regeneration and Advanced Therapies, Andalusian Center of Molecular Biology and Regenerative Medicine-CABIMER, University of Pablo de Olavide-University of Sevilla-CSIC, Seville, Spain.,Biomedical Research Network for Diabetes and Related Metabolic Diseases-CIBERDEM, Instituto de Salud Carlos III, Madrid, Spain
| | - Franz Martin
- Department of Molecular Biology and Biochemical Engineering, Universidad Pablo de Olavide, Seville, Spain.,Department of Cell Regeneration and Advanced Therapies, Andalusian Center of Molecular Biology and Regenerative Medicine-CABIMER, University of Pablo de Olavide-University of Sevilla-CSIC, Seville, Spain.,Biomedical Research Network for Diabetes and Related Metabolic Diseases-CIBERDEM, Instituto de Salud Carlos III, Madrid, Spain
| | - Gladys M Cahuana
- Department of Molecular Biology and Biochemical Engineering, Universidad Pablo de Olavide, Seville, Spain.,Department of Cell Regeneration and Advanced Therapies, Andalusian Center of Molecular Biology and Regenerative Medicine-CABIMER, University of Pablo de Olavide-University of Sevilla-CSIC, Seville, Spain
| | - Clara Guerra-Duarte
- Center of Research and Development, Fundação Ezequiel Dias, Belo Horizonte, Brazil
| | - Thamyres C Silva de Assis
- Departament of Biochemistry and Immunology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Francisco J Bedoya
- Department of Molecular Biology and Biochemical Engineering, Universidad Pablo de Olavide, Seville, Spain.,Department of Cell Regeneration and Advanced Therapies, Andalusian Center of Molecular Biology and Regenerative Medicine-CABIMER, University of Pablo de Olavide-University of Sevilla-CSIC, Seville, Spain.,Biomedical Research Network for Diabetes and Related Metabolic Diseases-CIBERDEM, Instituto de Salud Carlos III, Madrid, Spain
| | - Bernat Soria
- Department of Molecular Biology and Biochemical Engineering, Universidad Pablo de Olavide, Seville, Spain.,ISABIAL-Hospital General y Universitario de Alicante, Alicante, Spain.,Biomedical Research Network for Diabetes and Related Metabolic Diseases-CIBERDEM, Instituto de Salud Carlos III, Madrid, Spain.,Institute of Bioengineering, University Miguel Hernandez de Elche, Alicante, Spain
| | - Carlos Chávez-Olórtegui
- Departament of Biochemistry and Immunology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Juan R Tejedo
- Institute of Tropical Diseases, Universidad Nacional Toribio Rodríguez de Mendoza de Amazonas, Chachapoyas, Peru.,Department of Molecular Biology and Biochemical Engineering, Universidad Pablo de Olavide, Seville, Spain.,Department of Cell Regeneration and Advanced Therapies, Andalusian Center of Molecular Biology and Regenerative Medicine-CABIMER, University of Pablo de Olavide-University of Sevilla-CSIC, Seville, Spain.,Biomedical Research Network for Diabetes and Related Metabolic Diseases-CIBERDEM, Instituto de Salud Carlos III, Madrid, Spain
| |
Collapse
|
42
|
Antiangiogenic effects of phospholipase A 2 Lys49 BnSP-7 from Bothrops pauloensis snake venom on endothelial cells: An in vitro and ex vivo approach. Toxicol In Vitro 2021; 72:105099. [PMID: 33486049 DOI: 10.1016/j.tiv.2021.105099] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2020] [Revised: 12/22/2020] [Accepted: 01/15/2021] [Indexed: 11/20/2022]
Abstract
Antiangiogenic strategies are promising tools for cancer treatment and several other disorders. In this sense, phospholipases A2 (PLA2s) from snake venom have been described to possess antiangiogenic properties. In this study, we evaluated both in vitro and ex vivo antiangiogenic effects induced by BnSP-7, a Lys49 PLA2 isolated from Bothrops pauloensis snake venom. BnSP-7 was able to inhibit endothelial cell (HUVEC) proliferation, which was indeed confirmed by a modulation of cell cycle progression. Interestingly, BnSP-7 also inhibited the adhesion and migration of HUVECs and blocked in vitro angiogenesis in a VEGF-dependent manner, an important proangiogenic factor. Finally, BnSP-7 was capable of inhibiting sprouting angiogenic process through an ex vivo aortic ring assay. Taken together, these results indicate that BnSP-7 has potent in vitro and ex vivo antiangiogenic effect.
Collapse
|
43
|
In-solution structural studies involving a phospholipase A 2-like myotoxin and a natural inhibitor: Plasticity of oligomeric assembly affects mechanisms of inhibition. Biochimie 2020; 181:145-153. [PMID: 33333169 DOI: 10.1016/j.biochi.2020.12.008] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2020] [Revised: 12/11/2020] [Accepted: 12/12/2020] [Indexed: 02/02/2023]
Abstract
Snakebite envenomation has been categorized by World Health Organization as a category A neglected tropical disease, since it causes chronic psychological disorders, physical disablement and death. Ophidian accidents may cause local myonecrosis that cause drastic sequelae, which are not efficiently neutralized via serum therapy. Phospholipase A2-like (PLA2-like) myotoxins have a major role in the local effects caused by several snake venoms. We previously demonstrated that chicoric acid (CA) is an efficient inhibitor of the BthTX-I myotoxin and solved the X-ray structure of complex. Herein, we assess the oligomeric behavior of the BthTX-I/CA complex in solution under different physical-chemical conditions and using toxin obtained by two different biochemical methodologies to fully elucidate structural bases of inhibition of myotoxins by CA. We demonstrated the ability of PLA2-like proteins to form different oligomeric assemblies in the presence of certain inhibitors, which can also be modulated by buffer polarity change. In the presence of ethanol, BthTX-I/CA remains predominantly in a monomeric conformation, which prevents it from being in its active form (dimeric conformation). In contrast, in the absence of ethanol, the tetramer assembly was observed, which hid key regions of the protein responsible for docking and disruption of the muscle membrane. Therefore, the "plasticity" of these proteins with regard to their abilities to form oligomeric assemblies is a key issue for the future development of therapeutic agents to complement of serum therapy.
Collapse
|
44
|
Ghezellou P, Albuquerque W, Garikapati V, Casewell NR, Kazemi SM, Ghassempour A, Spengler B. Integrating Top-Down and Bottom-Up Mass Spectrometric Strategies for Proteomic Profiling of Iranian Saw-Scaled Viper, Echis carinatus sochureki, Venom. J Proteome Res 2020; 20:895-908. [PMID: 33225711 DOI: 10.1021/acs.jproteome.0c00687] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Saw-scaled or carpet vipers (genus Echis) are considered to cause a higher global snakebite mortality than any other snake. Echis carinatus sochureki (ECS) is a widely distributed snake species, also found across the thirteen provinces of Iran, where it is assumed to be responsible for the most snakebite envenomings. Here, we collected the Iranian specimens of ECS from three different geographically distinct populations, investigated food habits, and performed toxicity assessment and venom proteome profiling to better understand saw-scaled viper life. Our results show that the prey items most commonly found in all populations were arthropods, with scorpions from the family Buthidae particularly well represented. LD50 (median lethal dose) values of the crude venom demonstrate highly comparable venom toxicities in mammals. Consistent with this finding, venom characterization via top-down and bottom-up proteomics, applied to both crude venoms and size-exclusion chromatographic fractions, revealed highly comparable venom compositions among the different populations. By combining all proteomics data, we identified 22 protein families from 102 liquid chromatography and tandem mass spectrometry (LC-MS/MS) raw files, including the most abundant snake venom metalloproteinases (SVMPs, 29-34%); phospholipase A2 (PLA2s, 26-31%); snake venom serine proteinases (SVSPs, 11-12%); l-amino acid oxidases (LAOs, 8-11%), C-type lectins/lectin-like (CTLs, 7-9%) protein families, and many newly detected ones, e.g., renin-like aspartic proteases (RLAPs), fibroblast growth factors (FGFs), peptidyl-prolyl cis-trans isomerases (PPIs), and venom vasodilator peptides (VVPs). Furthermore, we identified and characterized methylated, acetylated, and oxidized proteoforms relating to the PLA2 and disintegrin toxin families and the site of their modifications. It thus seems that post-translational modifications (PTMs) of toxins, particularly target lysine residues, may play an essential role in the structural and functional properties of venom proteins and might be able to influence the therapeutic response of antivenoms, to be investigated in future studies.
Collapse
Affiliation(s)
- Parviz Ghezellou
- Institute of Inorganic and Analytical Chemistry, Justus Liebig University Giessen, Giessen 35392, Germany
| | - Wendell Albuquerque
- Institute of Food Chemistry and Food Biotechnology, Justus Liebig University Giessen, Giessen 35392, Germany
| | - Vannuruswamy Garikapati
- Institute of Inorganic and Analytical Chemistry, Justus Liebig University Giessen, Giessen 35392, Germany
| | - Nicholas R Casewell
- Centre for Snakebite Research & Interventions, Liverpool School of Tropical Medicine, Pembroke Place, Liverpool, L3 5QA, U.K
| | - Seyed Mahdi Kazemi
- Medicinal Plants and Drugs Research Institute, Shahid Beheshti University, Tehran 1983963113, Iran
| | - Alireza Ghassempour
- Medicinal Plants and Drugs Research Institute, Shahid Beheshti University, Tehran 1983963113, Iran
| | - Bernhard Spengler
- Institute of Inorganic and Analytical Chemistry, Justus Liebig University Giessen, Giessen 35392, Germany
| |
Collapse
|
45
|
Ghazaryan NA, Kishmiryan A, Ayvazyan NM. The Synergy of Membranotropic Effect of the Two Pla 2 Fractions of Macrovipera lebetina obtusa Venom on the Model Membranes. J Membr Biol 2020; 253:609-616. [PMID: 33089393 DOI: 10.1007/s00232-020-00144-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2020] [Accepted: 10/07/2020] [Indexed: 11/29/2022]
Abstract
It is known that snake venoms are a complex of enzymes and proteins and the interaction of different venom components with the membranes could be significantly enhanced in course of their action in an orchestra. The aim of the proposed investigation is to obtain detailed information about the mechanism and topology of two snake venom PLA2 isoforms from the Macrovipera lebetina obtusa venom in the membrane-binding process. We investigated the impact of the interaction on the properties of the model membrane (namely, GUVs and erythrocytes ghost) for each of these isoforms, as well as their synergetic action if they act simultaneously. The 6-lauroyl-2-dimethylaminonaphthalene and 6-propionyl-2-dimethylaminonaphthalene fluorescence probes were used to allow us to determine the membrane polarity more accurately via a generalized polarization function. Our results show that two types of PLA2 bring viscosity reduction in GUVs membrane and the effect became more potent when these PLA2 acts together. Intriguingly, we have not observed any significant difference in the case of the erythrocytes ghost membrane.
Collapse
Affiliation(s)
- N A Ghazaryan
- Laboratory of Toxinology and Molecular Systematics, Institute of Physiology, 0028, Yerevan, Armenia.
| | - A Kishmiryan
- Laboratory of Toxinology and Molecular Systematics, Institute of Physiology, 0028, Yerevan, Armenia
| | - N M Ayvazyan
- Laboratory of Toxinology and Molecular Systematics, Institute of Physiology, 0028, Yerevan, Armenia
| |
Collapse
|
46
|
The allosteric activation mechanism of a phospholipase A 2-like toxin from Bothrops jararacussu venom: a dynamic description. Sci Rep 2020; 10:16252. [PMID: 33004851 PMCID: PMC7529814 DOI: 10.1038/s41598-020-73134-9] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2020] [Accepted: 08/24/2020] [Indexed: 11/08/2022] Open
Abstract
The activation process of phospholipase A2-like (PLA2-like) toxins is a key step in their molecular mechanism, which involves oligomeric changes leading to the exposure of specific sites. Few studies have focused on the characterization of allosteric activators and the features that distinguish them from inhibitors. Herein, a comprehensive study with the BthTX-I toxin from Bothrops jararacussu venom bound or unbound to α-tocopherol (αT) was carried out. The oligomerization state of BthTX-I bound or unbound to αT in solution was studied and indicated that the toxin is predominantly monomeric but tends to oligomerize when complexed with αT. In silico molecular simulations showed the toxin presents higher conformational changes in the absence of αT,
which suggests that it is important to stabilize the structure of the toxin. The transition between the two states (active/inactive) was also studied, showing that only the unbound BthTX-I system could migrate to the inactive state. In contrast, the presence of αT induces the toxin to leave the inactive state, guiding it towards the active state, with more regions exposed to the solvent, particularly its active site. Finally, the structural determinants necessary for a molecule to be an inhibitor or activator were analyzed in light of the obtained results.
Collapse
|
47
|
Mamede CCN, de Sousa Simamoto BB, da Cunha Pereira DF, de Oliveira Costa J, Ribeiro MSM, de Oliveira F. Edema, hyperalgesia and myonecrosis induced by Brazilian bothropic venoms: overview of the last decade. Toxicon 2020; 187:10-18. [PMID: 32846146 DOI: 10.1016/j.toxicon.2020.08.016] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2020] [Revised: 08/15/2020] [Accepted: 08/19/2020] [Indexed: 01/19/2023]
Abstract
Snakebite accidents are considered serious public health problems. They are often neglected, and individuals who have received insufficient treatment are subjected to various disabling alterations. Snake venoms are secretions composed of biologically active molecules capable of triggering local and systemic effects in envenomation victims. Bothropic snakes are responsible for most of the ophidian accidents in Brazil; their venoms are mainly related to local manifestations, due to a composition that is especially rich in proteases and phospholipases A2. The most common local damages are inflammation, with consequent cellular activation and release of inflammatory mediators, hemorrhage, edema, pain and (myo)necrosis, which may lead to amputation of the affected areas. Antivenom therapy is the main treatment for snakebites. However, the efficiency is mainly due to the neutralization of the toxins responsible for the systemic alterations. Thus, the local damages can evolve to markedly compromise the tissue. The complexity of these local effects associated with the toxicity of the snake venom components of the genus Bothrops, arouse interest in the study of the biochemical and pathophysiological mechanisms involved with the actions caused by toxins of the venom. Therefore, this review aims to analyze the edematogenic, hyperalgesic and myotoxic effects caused by Brazilian bothropic venoms in order to contribute to the study and elucidation of the mechanisms of action of its components and, consequently, enable discoveries of more effective combined therapies in the treatment of local damages resulting from envenoming.
Collapse
Affiliation(s)
| | | | | | - Júnia de Oliveira Costa
- Instituto Federal de Educação, Ciência e Tecnologia do Triângulo Mineiro (IFTM), Ituiutaba, MG, Brazil
| | | | - Fabio de Oliveira
- Instituto de Ciências Biomédicas, Universidade Federal de Uberlândia (UFU), Uberlândia, MG, Brazil
| |
Collapse
|
48
|
Acidic Phospholipase A2-Peptide Derivative Modulates Oxidative Status and Microstructural Reorganization of Scar Tissue after Cutaneous Injury. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2020; 2020:8273986. [PMID: 32733589 PMCID: PMC7369679 DOI: 10.1155/2020/8273986] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/29/2020] [Accepted: 06/01/2020] [Indexed: 12/19/2022]
Abstract
From in vitro and in vivo models, the proliferative and healing potential of an acidic phospholipase A2 (LAPLA2) from Lachesis muta venom was investigated. The LAPLA2 proliferative activity was evaluated on fibroblasts and keratinocytes cultured, and the antioxidant and regenerative potential of LAPLA2 was analyzed in a murine model. The animal study consisted of four groups: C (negative control): 0.9% NaCl; SS (positive control): 1% silver sulfadiazine; L1 group: 0.5% LAPLA2; and L2 group: 0.25% LAPLA2. Wounds were topically treated daily for 12 days, and scar tissue samples were collected every 4 days. In vitro, LAPLA2 stimulated marked time-dependent cell proliferation. In vivo, it increased the antioxidant activity of superoxide dismutase (SOD) and catalase (CAT) and decreased malondialdehyde (MDA) and carbonyl protein (CP) levels in scar tissue treated with LAPLA2 at 0.5%. This peptide was effective in stimulating cellular proliferation, neoangiogenesis, type I and III collagen deposition, and maturation in a time-dependent-way, reducing the time required for wound closure. Our results indicated that LAPLA2 presented a remarkable potential in improving the oxidative status and microstructural reorganization of the scar tissue by stimulation of cellularity, angiogenesis, colagenogenesis, and wound contraction, suggesting that the peptide could be a potential candidate for a new healing drug.
Collapse
|
49
|
Sicherer ST, Venkatarama RS, Grasman JM. Recent Trends in Injury Models to Study Skeletal Muscle Regeneration and Repair. Bioengineering (Basel) 2020; 7:bioengineering7030076. [PMID: 32698352 PMCID: PMC7552705 DOI: 10.3390/bioengineering7030076] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Revised: 07/14/2020] [Accepted: 07/18/2020] [Indexed: 12/22/2022] Open
Abstract
Skeletal muscle injuries that occur from traumatic incidents, such as those caused by car accidents or surgical resections, or from injuries sustained on the battlefield, result in the loss of functionality of the injured muscle. To understand skeletal muscle regeneration and to better treat these large scale injuries, termed volumetric muscle loss (VML), in vivo injury models exploring the innate mechanisms of muscle injury and repair are essential for the creation of clinically applicable treatments. While the end result of a muscle injury is often the destruction of muscle tissue, the manner in which these injuries are induced as well as the response from the innate repair mechanisms found in muscle in each animal models can vary. This targeted review describes injury models that assess both skeletal muscle regeneration (i.e., the response of muscle to myotoxin or ischemic injury) and skeletal muscle repair (i.e., VML injury). We aimed to summarize the injury models used in the field of skeletal muscle tissue engineering, paying particular attention to strategies to induce muscle damage and how to standardize injury conditions for future experiments.
Collapse
|
50
|
Teixeira SC, Borges BC, Oliveira VQ, Carregosa LS, Bastos LA, Santos IA, Jardim ACG, Melo FF, Freitas LM, Rodrigues VM, Lopes DS. Insights into the antiviral activity of phospholipases A 2 (PLA 2s) from snake venoms. Int J Biol Macromol 2020; 164:616-625. [PMID: 32698062 PMCID: PMC7368918 DOI: 10.1016/j.ijbiomac.2020.07.178] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2020] [Revised: 07/08/2020] [Accepted: 07/14/2020] [Indexed: 12/18/2022]
Abstract
Viruses are associated with several human diseases that infect a large number of individuals, hence directly affecting global health and economy. Owing to the lack of efficient vaccines, antiviral therapy and emerging resistance strains, many viruses are considered as a potential threat to public health. Therefore, researches have been developed to identify new drug candidates for future treatments. Among them, antiviral research based on natural molecules is a promising approach. Phospholipases A2 (PLA2s) isolated from snake venom have shown significant antiviral activity against some viruses such as Dengue virus, Human Immunodeficiency virus, Hepatitis C virus and Yellow fever virus, and have emerged as an attractive alternative strategy for the development of novel antiviral therapy. Thus, this review provides an overview of remarkable findings involving PLA2s from snake venom that possess antiviral activity, and discusses the mechanisms of action mediated by PLA2s against different stages of virus replication cycle. Additionally, molecular docking simulations were performed by interacting between phospholipids from Dengue virus envelope and PLA2s from Bothrops asper snake venom. Studies on snake venom PLA2s highlight the potential use of these proteins for the development of broad-spectrum antiviral drugs.
Collapse
Affiliation(s)
- S C Teixeira
- Department of Immunology, Institute of Biomedical Science, Federal University of Uberlândia, Uberlândia, MG, Brazil
| | - B C Borges
- Department of Immunology, Institute of Biomedical Science, Federal University of Uberlândia, Uberlândia, MG, Brazil
| | - V Q Oliveira
- Multidisciplinary Institute of Health, Anísio Teixeira Campus, Federal University of Bahia, Vitória da Conquista, BA, Brazil
| | - L S Carregosa
- Multidisciplinary Institute of Health, Anísio Teixeira Campus, Federal University of Bahia, Vitória da Conquista, BA, Brazil
| | - L A Bastos
- Multidisciplinary Institute of Health, Anísio Teixeira Campus, Federal University of Bahia, Vitória da Conquista, BA, Brazil
| | - I A Santos
- Laboratory of Virology, Institute of Biomedical Science, Federal University of Uberlândia, Uberlândia, MG, Brazil
| | - A C G Jardim
- Laboratory of Virology, Institute of Biomedical Science, Federal University of Uberlândia, Uberlândia, MG, Brazil
| | - F F Melo
- Multidisciplinary Institute of Health, Anísio Teixeira Campus, Federal University of Bahia, Vitória da Conquista, BA, Brazil
| | - L M Freitas
- Multidisciplinary Institute of Health, Anísio Teixeira Campus, Federal University of Bahia, Vitória da Conquista, BA, Brazil
| | - V M Rodrigues
- Laboratory of Biochemistry and Animal Toxins, Institute of Biotechnology, Federal University of Uberlândia, Uberlândia, MG, Brazil.
| | - D S Lopes
- Multidisciplinary Institute of Health, Anísio Teixeira Campus, Federal University of Bahia, Vitória da Conquista, BA, Brazil; Institute of Health Sciences, Department of Bio-Function, Federal University of Bahia, Salvador, BA, Brazil.
| |
Collapse
|