1
|
Choudhury A, Linne K, Bulfone TC, Hossain T, Sina AAI, Bickler PL, Fry BG, Lewin MR. Electrical Cell Impedance Sensing (ECIS): Feasibility of a Novel In Vitro Approach to Studying Venom Toxicity and Potential Therapeutics. Toxins (Basel) 2025; 17:193. [PMID: 40278691 PMCID: PMC12031041 DOI: 10.3390/toxins17040193] [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: 02/18/2025] [Revised: 03/18/2025] [Accepted: 04/09/2025] [Indexed: 04/26/2025] Open
Abstract
Snakebite envenoming is often discussed in terms of lethality and limb loss, but local tissue injury and coagulotoxic effects of venom are significantly more common acute manifestations of snakebite envenoming (SBE). Local tissue injury and the hemorrhagic and coagulotoxic effects of venom are challenging to study in live animals and can be ethically fraught due to animal welfare concerns such that attention to the 3Rs of animal welfare motivates the development of in vitro techniques in this arena. Herein, we tested the use of a wound-healing study technique known as Electric Cell-Substrate Impedance Sensing (ECIS) to assess populations of cultured cells exposed to venom with or without sPLA2 and/or metalloprotease inhibitors (varespladib and marimastat, respectively). For comparison, the StarMax coagulation analyzer for coagulotoxicity was further used to evaluate the venoms and the neutralizing capabilities of the abovementioned direct toxin inhibitors (DTIs) against the same venoms examined using ECIS. Three viper and three elapid venoms that were examined for their effects on H1975 cells were Agkistrodon contortrix (Eastern Copperhead), Crotalus helleri (Southern Pacific Rattlesnake), and Vipera ammodytes (Horned Viper) and Naja atra (Chinese Cobra), Naja mossambica (Mozambique Spitting Cobra), and Naja nigricollis (Black-necked Spitting Cobra), respectively. The combination of cellular and coagulation techniques appears to usefully discriminate the in vitro capabilities and limitations of specific inhibitors to inhibit specific venom effects. This study suggests that ECIS with or without concomitant coagulation testing is a feasible method to generate reproducible, meaningful preclinical data and could be used with any type of cell line. Importantly, this approach is both quantitative and has the potential of reducing animal use and suffering during the evaluation of potential therapeutics. To further evaluate the potential of this method, rescue studies should be performed.
Collapse
Affiliation(s)
- Abhinandan Choudhury
- Adaptive Biotoxicology Lab, University of Queensland, St. Lucia, QLD 4072, Australia;
| | - Kaitlin Linne
- Department of Emergency Medicine, University of California San Francisco Medical Center, San Francisco, CA 94143, USA (T.C.B.); (P.L.B.)
| | - Tommaso C. Bulfone
- Department of Emergency Medicine, University of California San Francisco Medical Center, San Francisco, CA 94143, USA (T.C.B.); (P.L.B.)
| | - Tanvir Hossain
- Australian Institute for Bioengineering and Nanotechnology (AIBN), University of Queensland, St. Lucia, QLD 4072, Australia (A.A.I.S.)
| | - Abu Ali Ibn Sina
- Australian Institute for Bioengineering and Nanotechnology (AIBN), University of Queensland, St. Lucia, QLD 4072, Australia (A.A.I.S.)
| | - Philip L. Bickler
- Department of Emergency Medicine, University of California San Francisco Medical Center, San Francisco, CA 94143, USA (T.C.B.); (P.L.B.)
- California Academy of Sciences, San Francisco, CA 94118, USA
| | - Bryan G. Fry
- Adaptive Biotoxicology Lab, University of Queensland, St. Lucia, QLD 4072, Australia;
| | - Matthew R. Lewin
- California Academy of Sciences, San Francisco, CA 94118, USA
- Ophirex, Inc., Corte Madera, CA 94925, USA
| |
Collapse
|
2
|
Kadler R, Morrison B, Yanagihara AA. Assessing the Utility of Broad-Acting Inhibitors as Therapeutics in Diverse Venoms. Toxins (Basel) 2025; 17:188. [PMID: 40278686 PMCID: PMC12031005 DOI: 10.3390/toxins17040188] [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: 02/28/2025] [Revised: 03/29/2025] [Accepted: 04/07/2025] [Indexed: 04/26/2025] Open
Abstract
Examination of venom constituent bioactivities from diverse venomous animals shows certain highly conserved classes, including enzymes (e.g., phospholipases and metalloproteinases) and pore-forming proteins. While antivenoms targeting other unique and lethal venom components have proven to be life-saving, venom-enzyme-driven tissue damage and morbidity persists. Broad-acting enzyme inhibitors demonstrate the potential to augment antivenom approaches. In this study, we investigate the potential utility of certain broad-acting inhibitors in cubozoa for the first time. Fluorogenic assays were used to determine the phospholipase A2 (PLA2) and matrix metalloproteinase (MMP) activity of the Hawaiian box jellyfish, Alatina alata, and this was compared to representative elapid, viper, and bee venoms. In vitro, evaluation of selected small-molecule inhibitors demonstrated the ability and feasibility of the broad-acting therapeutic doxycycline, which inhibited the PLA2 and MMP activity of A. alata (approximately 50% reduction at 0.1 mM (95% CI 0.06-0.15) and 2.1 mM (95% CI 1.4-3.0), respectively), in addition to both snake venoms. Additionally, copper gluconate broadly inhibited the PLA2 activity of bee, snake, and jellyfish venoms. While all venoms are complex mixtures of bioactive molecules, these studies demonstrate that targeting common class components with broad-acting inhibitors shows promise in clinical and preclinical management.
Collapse
Affiliation(s)
- Raechel Kadler
- Department of Tropical Medicine, Medical Microbiology, and Pharmacology, John A. Burns School of Medicine, University of Hawai’i at Mānoa, Honolulu, HI 96822, USA;
| | - Breanna Morrison
- Department of Public Health, University of Birmingham, Birmingham B15 2TT, UK;
| | - Angel Anne Yanagihara
- Department of Tropical Medicine, Medical Microbiology, and Pharmacology, John A. Burns School of Medicine, University of Hawai’i at Mānoa, Honolulu, HI 96822, USA;
- Pacific Biosciences Research Center (PBRC), School of Ocean and Earth Science and Technology, University of Hawai’i at Mānoa, Honolulu, HI 96822, USA
| |
Collapse
|
3
|
Hearth J, Linne K, Harrison J, Zolfaghari H, Lewin MR. Feasibility study: Varespladib protects CD-1 mice from lethal doses of whole bee ( Apis mellifera) venom. Toxicon X 2025; 25:100214. [PMID: 39936081 PMCID: PMC11808668 DOI: 10.1016/j.toxcx.2025.100214] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2024] [Revised: 12/03/2024] [Accepted: 01/07/2025] [Indexed: 02/13/2025] Open
Abstract
Swarming Hymenoptera attacks can deliver high cumulative doses of venom resulting in death and life-threatening or chronically disabling injuries. Varespladib, a potent inhibitor of snake venom secretory PLA2 (sPLA2), is a relatively weak inhibitor of whole bee venom sPLA2 in vitro (pico-to low nanomolar for snake venom compared to μ M for Apis millera). Animal studies of varespladib against wasp (Vespa mandarinia) venom have shown promise against both nephropathy and coagulopathy, major markers of severe systemic toxicity distinct from hypersensitivity such as anaphylactoid and anaphylaxis reactions. We conducted a simple pilot study to evaluate if varespladib could feasibly decrease mortality against lethal doses of honeybee (Apis mellifera) venom in a murine model. When pre-mixed with a single dose of 10 mg/kg varespladib and administered intravenously (IV), varespladib prevented all mortality (0 of 10) in comparison to a cohort of mice administered lethal doses of whole bee venom alone (6 of 10) during a 24-h study period (N = 10 each group; log rank χ2 = 8.29; p < 0.005), and it eliminated signs of toxicity within 2 h while control animals either died or continued to show signs of toxicity. Survival in these animals despite poor in vitro sPLA2 inhibition suggests that suppression of the host sPLA2 response itself might play a role in the treatment of venom toxicity using an enzyme inhibitor rather than antivenom antibodies. Varespladib could be a useful tool for dissecting fundamental interactions between exogenous toxins and their corresponding endogenous counterparts.
Collapse
Affiliation(s)
| | - Kaitlin Linne
- University of California, San Francisco, United States
| | | | | | | |
Collapse
|
4
|
Yong MY, Tan KY, Tan CH. A genus-wide study on venom proteome variation and phospholipase A 2 inhibition in Asian lance-headed pit vipers (genus: Trimeresurus). Comp Biochem Physiol C Toxicol Pharmacol 2025; 288:110077. [PMID: 39579840 DOI: 10.1016/j.cbpc.2024.110077] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/08/2024] [Revised: 11/01/2024] [Accepted: 11/14/2024] [Indexed: 11/25/2024]
Abstract
High molecular weight proteins are present abundantly in viperid venoms. The amino acid sequence can be highly variable, contributing to the structure and function diversity of snake venom protein. However, this variability remains poorly understood in many species. The study investigated the venom protein variability in a distinct clade of Asian pit vipers (Trimeresurus species complex) through comparative proteomics, applying gel electrophoresis (SDS-PAGE), liquid chromatography-tandem mass spectrometry (LCMS/MS), and bioinformatic approaches. The proteomes revealed a number of conserved protein families, within each are variably expressed protein paralogs that are unrelated to the snake phylogeny and geographic origin. The expression levels of two major enzymes, i.e., snake venom serine proteinase and metalloproteinase, correlate weakly with procoagulant and hemorrhagic activities, implying co-expression of other functionally versatile toxins in the venom. The phospholipase A2 (PLA2) abundance correlates strongly with its enzymatic activity, and a unique phenotype was discovered in two species expressing extremely little PLA2. The commercial mono-specific antivenom effectively neutralized the venoms' procoagulant and hemorrhagic effects but failed to inhibit the PLA2 activities. Instead, the PLA2 activities of all venoms were effectively inhibited by the small molecule inhibitor varespladib, suggesting its potential to be repurposed as a highly potent adjuvant therapeutic in snakebite envenoming.
Collapse
Affiliation(s)
- Mun Yee Yong
- Department of Pharmacology, Faculty of Medicine, Universiti Malaya, Kuala Lumpur, Malaysia
| | - Kae Yi Tan
- Department of Molecular Medicine, Faculty of Medicine, Universiti Malaya, Kuala Lumpur, Malaysia
| | - Choo Hock Tan
- Department of Pharmacology, Faculty of Medicine, Universiti Malaya, Kuala Lumpur, Malaysia; School of Medicine, College of Life Sciences and Medicine, National Tsing Hua University, Hsinchu, Taiwan; Institute of Bioinformatics and Structural Biology, College of Life Sciences and Medicine, National Tsing Hua University, Hsinchu, Taiwan.
| |
Collapse
|
5
|
Castro-Amorim J, Pinto AV, Mukherjee AK, Ramos MJ, Fernandes PA. Beyond Fang's fury: a computational study of the enzyme-membrane interaction and catalytic pathway of the snake venom phospholipase A 2 toxin. Chem Sci 2025; 16:1974-1985. [PMID: 39759936 PMCID: PMC11694569 DOI: 10.1039/d4sc06511e] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2024] [Accepted: 12/19/2024] [Indexed: 01/07/2025] Open
Abstract
Snake venom-secreted phospholipases A2 (svPLA2s) are critical, highly toxic enzymes present in almost all snake venoms. Upon snakebite envenomation, svPLA2s hydrolyze cell membrane phospholipids and induce pathological effects such as paralysis, myonecrosis, inflammation, or pain. Despite its central importance in envenomation, the chemical mechanism of svPLA2s is poorly understood, with detrimental consequences for the design of small-molecule snakebite antidotes, which is highly undesirable given the gravity of the epidemiological data that ranks snakebite as the deadliest neglected tropical disease. We study a member of the svPLA2 family, the Myotoxin-I, which is part of the venom of the Central American pit viper terciopelo (Bothrops asper), a ubiquitous but highly aggressive and dangerous species responsible for the most problematic snakebites in its habitat. Furthermore, PLA2 enzymes are a paradigm of interfacial enzymology, as the complex membrane-enzyme interaction is as important as is crucial for its catalytic process. Here, we explore the detailed interaction between svPLA2 and a 1 : 1 POPC/POPS membrane, and how enzyme binding affects membrane structure and dynamics. We further investigated the two most widely accepted reaction mechanisms for svPLA2s: the 'single-water mechanism' and the 'assisted-water mechanism', using umbrella sampling simulations at the PBE/MM level of theory. We demonstrate that both pathways are catalytically viable. While both pathways occur in two steps, the single-water mechanism yielded a lower activation free energy barrier (20.14 kcal mol-1) for POPC hydrolysis, consistent with experimental and computational values obtained for human PLA2. The reaction mechanisms are similar, albeit not identical, and can be generalized to svPLA2 from most viper species. Furthermore, our findings demonstrate that the sole small molecule inhibitor currently undergoing clinical trials for snakebite is a perfect transition state analog. Thus, understanding snake venom sPLA2 chemistry will help find new, effective small molecule inhibitors with anti-snake venom efficacy.
Collapse
Affiliation(s)
- Juliana Castro-Amorim
- LAQV/Requimte, Departamento de Química e Bioquímica, Faculdade de Ciências da Universidade do Porto Rua do Campo Alegre, s/n 4169-007 Porto Portugal
| | - Alexandre V Pinto
- LAQV/Requimte, Departamento de Química e Bioquímica, Faculdade de Ciências da Universidade do Porto Rua do Campo Alegre, s/n 4169-007 Porto Portugal
| | - Ashis K Mukherjee
- Institute of Advanced Study in Science and Technology Vigyan Path Garchuk, Paschim Boragaon Guwahati-781035 Assam India
| | - Maria J Ramos
- LAQV/Requimte, Departamento de Química e Bioquímica, Faculdade de Ciências da Universidade do Porto Rua do Campo Alegre, s/n 4169-007 Porto Portugal
| | - Pedro A Fernandes
- LAQV/Requimte, Departamento de Química e Bioquímica, Faculdade de Ciências da Universidade do Porto Rua do Campo Alegre, s/n 4169-007 Porto Portugal
| |
Collapse
|
6
|
Salvador GHM, Cardoso FF, Lomonte B, Fontes MRM. Inhibitors and activators for myotoxic phospholipase A 2-like toxins from snake venoms - A structural overview. Biochimie 2024; 227:231-247. [PMID: 39089640 DOI: 10.1016/j.biochi.2024.07.016] [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: 04/10/2024] [Revised: 06/27/2024] [Accepted: 07/29/2024] [Indexed: 08/04/2024]
Abstract
Snakebite envenomations result in acute and chronic physical and psychological health effects on their victims, leading to a substantial socio-economic burden in tropical and subtropical countries. Local necrosis is one of the serious effects caused by envenomation, primarily induced by snake venoms from the Viperidae family through the direct action of components collectively denominated as myotoxins, including the phopholipase A2-like (PLA2-like) toxins. Considering the limitations of antivenoms in preventing the rapid development of local tissue damage caused by envenomation, the use of small molecule therapeutics has been suggested as potential first-aid treatments or as adjuvants to antivenom therapy. In this review, we provide an overview of the structural interactions of molecules exhibiting inhibitory activity toward PLA2-like toxins. Additionally, we discuss the implications for the myotoxic mechanism of PLA2-like toxins and the molecules involved in their activation, highlighting key differences between activators and inhibitors. Finally, we integrate all these results to propose a classification of inhibitors into three different classes and five sub-classes. Taking into account the structural and affinity information, we compare the different inhibitors/ligands to gain a deeper understanding of the structural basis for the effective inhibition of PLA2-like toxins. By offering these insights, we aim to contribute to the search for new and efficient inhibitor molecules to complement and improve current therapy by conventional antivenoms.
Collapse
Affiliation(s)
- Guilherme H M Salvador
- Departamento de Biofísica e Farmacologia, Instituto de Biociências, Universidade Estadual Paulista (UNESP), Botucatu-SP, Brazil
| | - Fábio F Cardoso
- Departamento de Biofísica e Farmacologia, Instituto de Biociências, Universidade Estadual Paulista (UNESP), Botucatu-SP, Brazil
| | - Bruno Lomonte
- Instituto Clodomiro Picado, Facultad de Microbiología, Universidad de Costa Rica, San José, Costa Rica
| | - Marcos R M 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
|
7
|
Chowdhury A, Fry BG, Samuel SP, Bhalla A, Vaiyapuri S, Bhargava P, Carter RW, Lewin MR. In vitro anticoagulant effects of Bungarus venoms on human plasma which are effectively neutralized by the PLA 2-inhibitor varespladib. Toxicon 2024; 252:108178. [PMID: 39547452 DOI: 10.1016/j.toxicon.2024.108178] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2024] [Revised: 10/31/2024] [Accepted: 11/12/2024] [Indexed: 11/17/2024]
Abstract
Bungarus (krait) envenomings are well-known for their life-threatening neurotoxic effects. However, their impact on coagulation remains largely unexplored experimentally or clinically. This study, examined the effect of begins to examine venoms from four Bungarus species-B. caeruleus, B. candidus, B. fasciatus, and B. flaviceps on human platelet poor plasma coagulation parameters using thromboelastography and coagulation inhibition assays. B. flaviceps completely inhibited clotting, while B. caeruleus only delayed clot formation. In contrast, B. candidus and B. fasciatus did not affect clotting. Subsequent examinations into the anticoagulant biochemical mechanisms demonstrated divergent pathophysiological pathways. B. caeruleus venom anticoagulant effects were prevented by the addition of an excess of phospholipids, with anticoagulation thereby the result of phospholipid depletion. In contrast B. flaviceps anticoagulation was not affected by the addition of an excess of phospholipids. Further investigations demonstrated that B. flaviceps mediates its anticoagulant toxicity through the inactivation of coagulation enzymes. The anticoagulant effects of both B. flaviceps and B. caeruleus were nullified by varespladib, a phospholipase A2 (PLA2) inhibitor, revealing the toxin class involved. These results uncover previously unrecognized and unexplored anticoagulant effects of Bungarus venoms.
Collapse
Affiliation(s)
- Abhinandan Chowdhury
- Adaptive Biotoxicology Lab, School of the Environment, University of Queensland, St Lucia, QLD, 4072, Australia; Department of Biochemistry & Microbiology, North South University, Dhaka, Bangladesh
| | - Bryan G Fry
- Adaptive Biotoxicology Lab, School of the Environment, University of Queensland, St Lucia, QLD, 4072, Australia.
| | - Stephen P Samuel
- Ophirex, Inc., Corte Madera, CA, 94925, USA; California Academy of Sciences, San Francisco, CA, 94118, USA
| | - Ashish Bhalla
- Post Graduate Institute of Medical Education and Research, Chandigarh, India
| | | | - Parul Bhargava
- Department of Pathology and Laboratory Medicine, University of San Francisco, California, USA
| | | | - Matthew R Lewin
- Ophirex, Inc., Corte Madera, CA, 94925, USA; California Academy of Sciences, San Francisco, CA, 94118, USA.
| |
Collapse
|
8
|
Lay M, Hodgson WC. Isolation and Pharmacological Characterisation of Pre-Synaptic Neurotoxins from Thai and Javanese Russell's Viper ( Daboia siamensis) Venoms. Toxins (Basel) 2024; 16:405. [PMID: 39330863 PMCID: PMC11436103 DOI: 10.3390/toxins16090405] [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: 07/18/2024] [Revised: 08/28/2024] [Accepted: 09/10/2024] [Indexed: 09/28/2024] Open
Abstract
The widespread geographical distribution of Russell's vipers (Daboia spp.) is associated with marked variations in the clinical outcomes of envenoming by species from different countries. This is likely to be due to differences in the quantity and potency of key toxins and, potentially, the presence or absence of some toxins in venoms across the geographical spectrum. In this study, we aimed to isolate and pharmacologically characterise the major neurotoxic components of D. siamensis venoms from Thailand and Java (Indonesia) and explore the efficacy of antivenom and a PLA2 inhibitor, Varespladib, against the neuromuscular activity. These data will provide insights into the link between venom components and likely clinical outcomes, as well as potential treatment strategies. Venoms were fractionated using RP-HPLC and the in vitro activity of isolated toxins assessed using the chick biventer cervicis nerve-muscle preparation. Two major PLA2 fractions (i.e., fractions 8 and 10) were isolated from each venom. Fraction 8 from both venoms produced pre-synaptic neurotoxicity and myotoxicity, whereas fraction 10 from both venoms was weakly neurotoxic. The removal of the two fractions from each venom abolished the in vitro neurotoxicity, and partially abolished myotoxicity, of the whole venom. A combination of the two fractions from each venom produced neurotoxic activity that was equivalent to the respective whole venom (10 µg/mL), but the myotoxic effects were not additive. The in vitro neurotoxicity of fraction 8 (100 nM) from each venom was prevented by the pre-administration of Thai Russell's viper monovalent antivenom (2× recommended concentration) or preincubation with Varespladib (100 nM). Additionally, the neurotoxicity produced by a combination of the two fractions was partially reversed by the addition of Varespladib (100-300 nM) 60 min after the fractions. The present study demonstrates that the in vitro skeletal muscle effects of Thai and Javanese D. siamensis venoms are primarily due to key PLA2 toxins in each venom.
Collapse
Affiliation(s)
| | - Wayne C. Hodgson
- Monash Venom Group, Department of Pharmacology, Biomedical Discovery Institute, Monash University, Clayton, VIC 3800, Australia;
| |
Collapse
|
9
|
Murphy K, Tasoulis T, Dunstan N, Isbister GK. Anticoagulant activity in Australasian elapid snake venoms and neutralisation with antivenom and varespladib. Toxicon 2024; 247:107836. [PMID: 38945217 DOI: 10.1016/j.toxicon.2024.107836] [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: 04/07/2024] [Revised: 06/04/2024] [Accepted: 06/25/2024] [Indexed: 07/02/2024]
Abstract
The venoms of Australasian elapid snakes are known to possess coagulant activity, including some with strong procoagulant activity and others with anticoagulant activity, although the latter are less well known. This study investigates the anticoagulant activity of Australasian elapid snake venoms, and whether this activity is neutralised by commercial snake antivenom and varespladib (PLA2 inhibiting agent). Clotting assays were completed for 34 species of Australasian elapids. Antivenom neutralisation assays with tiger snake antivenom (TSAV) were performed on five species to determine if there was cross-neutralisation. Varespladib neutralisation assays were also completed for the same five species. All Pseudechis species venoms had anticoagulant activity, except P. porphyriacus, which was procoagulant. Pseudechis species venoms had similar anticoagulant potency ranging from the most potent P. colletti venom to the least potent P. butleri venom. The three Austrelaps (copperhead) species venoms were the next most potent anticoagulants. Six further snakes, Elapognathus coronatus, Acanthophis pyrrhus, A. antarcticus, Suta suta, Denisonia devisi and D. maculata, had weaker anticoagulant activity, except for D. maculata which had similar anticoagulant activity to Pseudechis species. Tiger Snake Antivenom (1200mU/mL) neutralised the anticoagulant effect of P. australis for concentrations up to 1 mg/mL. TSAV (1200mU/mL) also neutralised P. colletti, D. maculata, A. superbus and A. pyrrhus venoms at their EC50, demonstrating cross neutralisation. Varespladib neutralised the anticoagulant effect of P. australis venom at 5 μM and for venoms of P. colletti, D. maculata, A. superbus and A. pyrrhus. We found anticoagulant activity to be present in six genera of Australasian snakes at low concentrations, which can be completely neutralised by both antivenom and varespladib. Anticoagulant activity in Australian elapid venoms was associated with species possessing high PLA2 activity without procoagulant snake venom serine proteases.
Collapse
Affiliation(s)
- Kate Murphy
- Clinical Toxicology Research Group, University of Newcastle, New South Wales, Australia
| | - Theo Tasoulis
- Clinical Toxicology Research Group, University of Newcastle, New South Wales, Australia
| | | | - Geoffrey K Isbister
- Clinical Toxicology Research Group, University of Newcastle, New South Wales, Australia; Department of Clinical Toxicology, Calvary Mater Newcastle, New South Wales, Australia.
| |
Collapse
|
10
|
Diniz EADS, da Silva DP, Ferreira SDS, Fernandes-Pedrosa MDF, Vieira DS. Temperature effect in the inhibition of PLA 2 activity of Bothrops brazili venom by Rosmarinic and Chlorogenic acids, experimental and computational approaches. J Biomol Struct Dyn 2024; 42:5238-5252. [PMID: 37378497 DOI: 10.1080/07391102.2023.2226912] [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: 03/28/2023] [Accepted: 06/09/2023] [Indexed: 06/29/2023]
Abstract
Myotoxicity caused by snakebite envenoming emerges as one of the main problems of ophidic accidents as it is not well neutralized by the current serum therapy. A promising alternative is to search for efficient small molecule inhibitors that can act against multiple venom components. Phospholipase A2 (PLA2) is frequently found in snake venom and is usually associated with myotoxicity. Thus it represents an excellent target for the search of new treatments. This work reports the effect of temperature in the inhibition of catalytic properties of PLA2 from Bothrops brazili venom by Rosmarinic (RSM) and Chlorogenic (CHL) acids through experimental and computational approaches. Three temperatures were evaluated (25, 37 and 50 °C). In the experimental section, enzymatic assays showed that RSM is a better inhibitor in all three temperatures. At 50 °C, the inhibition efficiency decayed significantly for both acids. Docking studies revealed that both ligands bind to the hydrophobic channel of the protein dimer where the phospholipid binds in the catalytic process, interacting with several functional residues. In this context, RSM presents better interaction energies due to stronger interactions with chain B of the dimer. Molecular dynamics simulations showed that RSM can establish selective interactions with ARG112B of PLA2, which is located next to residues of the putative Membrane Disruption Site in PLA2-like structures. The affinity of RSM and CHL acids towards PLA2 is mainly driven by electrostatic interactions, especially salt bridge interactions established with residues ARG33B (for CHL) and ARG112B (RSM) and hydrogen bonds with residue ASP89A. The inability of CHL to establish a stable interaction with ARG112B was identified as the reason for its lower inhibition efficiency compared to RSM at the three temperatures. Furthermore, extensive structural analysis was performed to explain the lower inhibition efficiency at 50 °C for both ligands. The analysis performed in this work provides important information for the future design of new inhibitors.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
| | - 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, Petrópolis, Natal, Brazil
| | - 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, Petrópolis, Natal, 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, Petrópolis, Natal, Brazil
| | - Davi Serradella Vieira
- Institute of Chemistry, Federal University of Rio Grande do Norte, Av Senador Salgado Filho, Natal-RN, Brazil
| |
Collapse
|
11
|
Bittenbinder MA, Wachtel E, Pereira DDC, Slagboom J, Casewell NR, Jennings P, Kool J, Vonk FJ. Development of a membrane-disruption assay using phospholipid vesicles as a proxy for the detection of cellular membrane degradation. Toxicon X 2024; 22:100197. [PMID: 38633504 PMCID: PMC11021370 DOI: 10.1016/j.toxcx.2024.100197] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2024] [Revised: 03/20/2024] [Accepted: 03/22/2024] [Indexed: 04/19/2024] Open
Abstract
Snakebite envenoming is a global health issue that affects millions of people worldwide, and that causes morbidity rates surpassing 450,000 individuals annually. Patients suffering from snakebite morbidities may experience permanent disabilities such as pain, blindness and amputations. The (local) tissue damage that causes these life-long morbidities is the result of cell- and tissue-damaging toxins present in the venoms. These compounds belong to a variety of toxin classes and may affect cells in various ways, for example, by affecting the cell membrane. In this study, we have developed a high-throughput in vitro assay that can be used to study membrane disruption caused by snake venoms using phospholipid vesicles from egg yolk as a substrate. Resuspended chicken egg yolk was used to form these vesicles, which were fluorescently stained to allow monitoring of the degradation of egg yolk vesicles on a plate reader. The assay proved to be suitable for studying phospholipid vesicle degradation of crude venoms and was also tested for its applicability for neutralisation studies of varespladib, which is a PLA2 inhibitor. We additionally made an effort to identify the responsible toxins using liquid chromatography, followed by post-column bioassaying and protein identification using high-throughput venomics. We successfully identified various toxins in the venoms of C. rhodostoma and N. mossambica, which are likely to be involved in the observed vesicle-degrading effect. This indicates that the assay can be used for screening the membrane degrading activity of both crude and fractionated venoms as well as for neutralisation studies.
Collapse
Affiliation(s)
- Mátyás A. Bittenbinder
- Naturalis Biodiversity Center, Leiden, the Netherlands
- AIMMS Division of BioAnalytical Chemistry, Department of Chemistry and Pharmaceutical Sciences, Faculty of Sciences, Vrije Universiteit Amsterdam, Amsterdam, the Netherlands
- Centre for Analytical Sciences Amsterdam (CASA), Amsterdam, the Netherlands
| | - Eric Wachtel
- AIMMS Division of BioAnalytical Chemistry, Department of Chemistry and Pharmaceutical Sciences, Faculty of Sciences, Vrije Universiteit Amsterdam, Amsterdam, the Netherlands
| | - Daniel Da Costa Pereira
- AIMMS Division of Molecular and Computational Toxicology, Department of Chemistry and Pharmaceutical Sciences, Vrije Universiteit Amsterdam, Amsterdam, the Netherlands
| | - Julien Slagboom
- AIMMS Division of BioAnalytical Chemistry, Department of Chemistry and Pharmaceutical Sciences, Faculty of Sciences, Vrije Universiteit Amsterdam, Amsterdam, the Netherlands
- Centre for Analytical Sciences Amsterdam (CASA), Amsterdam, the Netherlands
| | - Nicholas R. Casewell
- Centre for Snakebite Research & Interventions, Liverpool School of Tropical Medicine, Liverpool, United Kingdom
| | - Paul Jennings
- AIMMS Division of Molecular and Computational Toxicology, Department of Chemistry and Pharmaceutical Sciences, Vrije Universiteit Amsterdam, Amsterdam, the Netherlands
| | - Jeroen Kool
- AIMMS Division of BioAnalytical Chemistry, Department of Chemistry and Pharmaceutical Sciences, Faculty of Sciences, Vrije Universiteit Amsterdam, Amsterdam, the Netherlands
- Centre for Analytical Sciences Amsterdam (CASA), Amsterdam, the Netherlands
| | - Freek J. Vonk
- Naturalis Biodiversity Center, Leiden, the Netherlands
- AIMMS Division of BioAnalytical Chemistry, Department of Chemistry and Pharmaceutical Sciences, Faculty of Sciences, Vrije Universiteit Amsterdam, Amsterdam, the Netherlands
- Centre for Analytical Sciences Amsterdam (CASA), Amsterdam, the Netherlands
| |
Collapse
|
12
|
Stazi M, Megighian A, D'Este G, Negro S, Ivanušec A, Lonati D, Pirazzini M, Križaj I, Montecucco C. An agonist of CXCR4 induces a rapid recovery from the neurotoxic effects of Vipera ammodytes and Vipera aspis venoms. J Neurochem 2024; 168:428-440. [PMID: 36912731 DOI: 10.1111/jnc.15803] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2022] [Revised: 01/30/2023] [Accepted: 02/06/2023] [Indexed: 03/14/2023]
Abstract
People bitten by Alpine vipers are usually treated with antivenom antisera to prevent the noxious consequences caused by the injected venom. However, this treatment suffers from a number of drawbacks and additional therapies are necessary. The venoms of Vipera ammodytes and of Vipera aspis are neurotoxic and cause muscle paralysis by inducing neurodegeneration of motor axon terminals because they contain a presynaptic acting sPLA2 neurotoxin. We have recently found that any type of damage to motor axons is followed by the expression and activation of the intercellular signaling axis consisting of the CXCR4 receptor present on the membrane of the axon stump and of its ligand, the chemokine CXCL12 released by activated terminal Schwann cells. We show here that also V. ammodytes and V. aspis venoms cause the expression of the CXCL12-CXCR4 axis. We also show that a small molecule agonist of CXCR4, dubbed NUCC-390, induces a rapid regeneration of the motor axon terminal with functional recovery of the neuromuscular junction. These findings qualify NUCC-390 as a promising novel therapeutics capable of improving the recovery from the paralysis caused by the snakebite of the two neurotoxic Alpine vipers.
Collapse
Affiliation(s)
- M Stazi
- Department of Biomedical Sciences, University of Padova, Padova, Italy
| | - A Megighian
- Department of Biomedical Sciences, University of Padova, Padova, Italy
| | - G D'Este
- Department of Biomedical Sciences, University of Padova, Padova, Italy
| | - S Negro
- Department of Biomedical Sciences, University of Padova, Padova, Italy
| | - A Ivanušec
- Department of Molecular and Biomedical Sciences, Jožef Stefan Institute, Ljubljana, Slovenia
- Doctoral School, Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| | - D Lonati
- Pavia Poison Control Centre - National Toxicology Information Centre - Clinical and Experimental Lab, Toxicology Unit, Istituti Clinici Scientifici Maugeri SpA SB IRCCS, Pavia, Italy
| | - M Pirazzini
- Department of Biomedical Sciences, University of Padova, Padova, Italy
| | - I Križaj
- Department of Molecular and Biomedical Sciences, Jožef Stefan Institute, Ljubljana, Slovenia
| | - C Montecucco
- Department of Biomedical Sciences, University of Padova, Padova, Italy
- National Research Council, Institute of Neuroscience, Padova, Italy
| |
Collapse
|
13
|
Jeong B, Kim JS, Kwon AR, Lee J, Park S, Koo J, Lee WS, Baek JY, Shin WH, Lee JS, Jeong J, Kim WK, Jung CR, Kim NS, Cho SH, Lee DY. Maternal nanoplastic ingestion induces an increase in offspring body weight through altered lipid species and microbiota. ENVIRONMENT INTERNATIONAL 2024; 185:108522. [PMID: 38401434 DOI: 10.1016/j.envint.2024.108522] [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: 11/23/2023] [Revised: 01/13/2024] [Accepted: 02/19/2024] [Indexed: 02/26/2024]
Abstract
The rapidly increasing prevalence of obesity and overweight, especially in children and adolescents, has become a serious societal issue. Although various genetic and environmental risk factors for pediatric obesity and overweight have been identified, the problem has not been solved. In this study, we examined whether environmental nanoplastic (NP) pollutants can act as environmental obesogens using mouse models exposed to NPs derived from polystyrene and polypropylene, which are abundant in the environment. We found abnormal weight gain in the progeny until 6 weeks of age following the oral administration of NPs to the mother during gestation and lactation. Through a series of experiments involving multi-omic analyses, we have demonstrated that NP-induced weight gain is caused by alterations in the lipid composition (lysophosphatidylcholine/phosphatidylcholine ratio) of maternal breast milk and he gut microbiota distribution of the progeny. These data indicate that environmental NPs can act as obesogens in childhood.
Collapse
Affiliation(s)
- Bohyeon Jeong
- Rare Disease Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), 125 Gwahak-ro, Yuseong-gu, Daejeon 34141, South Korea
| | - Ji-Sun Kim
- Korean Collection for Type Cultures, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Jeongeup, 56212, South Korea
| | - A Ra Kwon
- Chemical Analysis Center, Korea Research Institute of Chemical Technology (KRICT), Daejeon 34114, South Korea
| | - Jangjae Lee
- Chemical Analysis Center, Korea Research Institute of Chemical Technology (KRICT), Daejeon 34114, South Korea; Department of Chemistry, Korea University, Seoul 02841, South Korea
| | - Subin Park
- Rare Disease Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), 125 Gwahak-ro, Yuseong-gu, Daejeon 34141, South Korea
| | - Jahong Koo
- Rare Disease Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), 125 Gwahak-ro, Yuseong-gu, Daejeon 34141, South Korea; KRIBB School, Korea University of Science and Technology (UST), Daejeon, South Korea
| | - Wang Sik Lee
- Environmental Disease Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), 125 Gwahak-ro, Yuseong-gu, Daejeon 34141, South Korea
| | - Jeong Yeob Baek
- Rare Disease Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), 125 Gwahak-ro, Yuseong-gu, Daejeon 34141, South Korea
| | - Won-Ho Shin
- Department of Predictive Toxicology, Korea Institute of Toxicology, Daejeon 34114, South Korea
| | - Jung-Sook Lee
- Korean Collection for Type Cultures, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Jeongeup, 56212, South Korea; KRIBB School, Korea University of Science and Technology (UST), Daejeon, South Korea
| | - Jinyoung Jeong
- KRIBB School, Korea University of Science and Technology (UST), Daejeon, South Korea; Environmental Disease Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), 125 Gwahak-ro, Yuseong-gu, Daejeon 34141, South Korea
| | - Won Kon Kim
- KRIBB School, Korea University of Science and Technology (UST), Daejeon, South Korea; Metabolic Regulation Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), 125 Gwahak-ro, Yuseong-gu, Daejeon 34141, South Korea
| | - Cho-Rok Jung
- KRIBB School, Korea University of Science and Technology (UST), Daejeon, South Korea; Stem Cell Convergence Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), 125 Gwahak-ro, Yuseong-gu, Daejeon 34141, South Korea
| | - Nam-Soon Kim
- Rare Disease Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), 125 Gwahak-ro, Yuseong-gu, Daejeon 34141, South Korea; KRIBB School, Korea University of Science and Technology (UST), Daejeon, South Korea
| | - Sung-Hee Cho
- Chemical Analysis Center, Korea Research Institute of Chemical Technology (KRICT), Daejeon 34114, South Korea.
| | - Da Yong Lee
- Rare Disease Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), 125 Gwahak-ro, Yuseong-gu, Daejeon 34141, South Korea; KRIBB School, Korea University of Science and Technology (UST), Daejeon, South Korea.
| |
Collapse
|
14
|
Lay M, Hodgson WC. A Comparison of the Efficacy of Antivenoms and Varespladib against the In Vitro Pre-Synaptic Neurotoxicity of Thai and Javanese Russell's Viper ( Daboia spp.) Venoms. Toxins (Basel) 2024; 16:124. [PMID: 38535790 PMCID: PMC10974476 DOI: 10.3390/toxins16030124] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2024] [Revised: 02/20/2024] [Accepted: 02/27/2024] [Indexed: 04/25/2025] Open
Abstract
The heterogeneity in venom composition and potency in disparate Eastern Russell's viper (Daboia siamensis) populations has repercussions for the efficacy of antivenoms. This is particularly pronounced in geographical areas in which the venom of the local species has not been well studied and locally produced antivenoms are unavailable. In such cases, alternative therapies following envenoming, which are not limited by species specificity, may be employed to complement antivenoms. We studied the neuromuscular activity of D. siamensis venom from Thailand and Java (Indonesia) and the ability of Thai antivenoms and/or Varespladib to prevent or reverse these effects. Both Thai and Javanese D. siamensis venoms displayed potent pre-synaptic neurotoxicity but weak myotoxicity in the chick biventer cervicis nerve-muscle preparation. Whilst the neurotoxicity induced by both venoms was abolished by the prior administration of Thai D. siamensis monovalent antivenom or pre-incubation with Varespladib, Thai neuro-polyvalent antivenom only produced partial protection when added prior to venom. Pre-synaptic neurotoxicity was not reversed by the post-venom addition of either antivenom 30 or 60 min after either venom. Varespladib, when added 60 min after venom, prevented further inhibition of indirect twitches. However, the subsequent addition of additional concentrations of Varespladib did not result in further recovery from neurotoxicity. The combination of Thai monovalent antivenom and Varespladib, added 60 min after venom, resulted in additional recovery of twitches caused by either Thai or Javanese venoms compared with antivenom alone. In conclusion, we have shown that Varespladib can prevent and partially reverse the pre-synaptic neurotoxicity induced by either Thai or Javanese D. siamensis venoms. The efficacy of Thai D. siamensis monovalent antivenom in reversing pre-synaptic neurotoxicity was significantly enhanced by its co-administration with Varespladib. Further work is required to establish the efficacy of Varespladib as a primary or adjunct therapy in human envenoming.
Collapse
Affiliation(s)
| | - Wayne C. Hodgson
- Monash Venom Group, Department of Pharmacology, Biomedical Discovery Institute, Monash University, Clayton, VIC 3800, Australia;
| |
Collapse
|
15
|
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
|
16
|
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
|
17
|
Albulescu LO, Westhorpe A, Clare RH, Woodley CM, James N, Kool J, Berry NG, O’Neill PM, Casewell NR. Optimizing drug discovery for snakebite envenoming via a high-throughput phospholipase A2 screening platform. Front Pharmacol 2024; 14:1331224. [PMID: 38273832 PMCID: PMC10808766 DOI: 10.3389/fphar.2023.1331224] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Accepted: 12/19/2023] [Indexed: 01/27/2024] Open
Abstract
Snakebite envenoming is a neglected tropical disease that causes as many as 1.8 million envenomings and 140,000 deaths annually. To address treatment limitations that exist with current antivenoms, the search for small molecule drug-based inhibitors that can be administered as early interventions has recently gained traction. Snake venoms are complex mixtures of proteins, peptides and small molecules and their composition varies substantially between and within snake species. The phospholipases A2 (PLA2) are one of the main pathogenic toxin classes found in medically important viper and elapid snake venoms, yet varespladib, a drug originally developed for the treatment of acute coronary syndrome, remains the only PLA2 inhibitor shown to effectively neutralise venom toxicity in vitro and in vivo, resulting in an extremely limited drug portfolio. Here, we describe a high-throughput drug screen to identify novel PLA2 inhibitors for repurposing as snakebite treatments. We present method optimisation of a 384-well plate, colorimetric, high-throughput screening assay that allowed for a throughput of ∼2,800 drugs per day, and report on the screening of a ∼3,500 post-phase I repurposed drug library against the venom of the Russell's viper, Daboia russelii. We further explore the broad-spectrum inhibitory potential and efficacy of the resulting top hits against a range of medically important snake venoms and demonstrate the utility of our method in determining drug EC50s. Collectively, our findings support the future application of this method to fully explore the chemical space to discover novel PLA2-inhibiting drugs of value for preventing severe pathology caused by snakebite envenoming.
Collapse
Affiliation(s)
- Laura-Oana Albulescu
- Centre for Snakebite Research and Interventions, Department of Tropical Disease Biology, Liverpool School of Tropical Medicine, Liverpool, United Kingdom
- Centre for Drugs and Diagnostics, Department of Tropical Disease Biology, Liverpool School of Tropical Medicine, Liverpool, United Kingdom
| | - Adam Westhorpe
- Centre for Snakebite Research and Interventions, Department of Tropical Disease Biology, Liverpool School of Tropical Medicine, Liverpool, United Kingdom
- Centre for Drugs and Diagnostics, Department of Tropical Disease Biology, Liverpool School of Tropical Medicine, Liverpool, United Kingdom
| | - Rachel H. Clare
- Centre for Snakebite Research and Interventions, Department of Tropical Disease Biology, Liverpool School of Tropical Medicine, Liverpool, United Kingdom
- Centre for Drugs and Diagnostics, Department of Tropical Disease Biology, Liverpool School of Tropical Medicine, Liverpool, United Kingdom
| | | | - Nivya James
- Department of Chemistry, University of Liverpool, Liverpool, United Kingdom
| | - Jeroen Kool
- Division of BioAnalytical Chemistry, Department of Chemistry and Pharmaceutical Sciences, Faculty of Science, Amsterdam Institute of Molecular and Life Sciences, Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | - Neil G. Berry
- Department of Chemistry, University of Liverpool, Liverpool, United Kingdom
| | - Paul M. O’Neill
- Department of Chemistry, University of Liverpool, Liverpool, United Kingdom
| | - Nicholas R. Casewell
- Centre for Snakebite Research and Interventions, Department of Tropical Disease Biology, Liverpool School of Tropical Medicine, Liverpool, United Kingdom
- Centre for Drugs and Diagnostics, Department of Tropical Disease Biology, Liverpool School of Tropical Medicine, Liverpool, United Kingdom
| |
Collapse
|
18
|
Gilliam LL, Gilliam J, Samuel SP, Carter RW, Ritchey J, Bulfone T, Gutiérrez JM, Williams DJ, Durkin DM, Stephens SI, Lewin MR. Oral and IV Varespladib Rescue Experiments in Juvenile Pigs with Weakness Induced by Australian and Papuan Oxyuranus scutellatus Venoms. Toxins (Basel) 2023; 15:557. [PMID: 37755983 PMCID: PMC10537020 DOI: 10.3390/toxins15090557] [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: 06/08/2023] [Revised: 08/02/2023] [Accepted: 08/17/2023] [Indexed: 09/28/2023] Open
Abstract
Antivenom is currently the standard-of-care treatment for snakebite envenoming, but its efficacy is limited by treatment delays, availability, and in many cases, species specificity. Many of the rapidly lethal effects of envenoming are caused by venom-derived toxins, such as phospholipase A2 (sPLA2); therefore, small molecule direct toxin inhibitors targeting these toxins may have utility as initial and adjunct therapies after envenoming. Varespladib (intravenous, IV) and varespladib-methyl (oral) have been shown to potently inhibit sPLA2s from snake venoms in murine and porcine models, thus supporting their further study as potential treatments for snakebite envenoming. In this pilot study, we tested the ability of these compounds to reverse neurotoxic effects of venom from the Australian and Papuan taipan (Oxyuranus scutellatus) subspecies in juvenile pigs (Sus domesticus). The mean survival time for control animals receiving Australian taipan venom (0.03 mg/kg, n = 3) was 331 min ± 15 min; for those receiving Papuan taipan venom (0.15 mg/kg, n = 3) it was 178 ± 31 min. Thirteen pigs received Australian taipan venom and treatment with either IV or oral varespladib (or with IV to oral transition) and all 13 survived the duration of the study (≥96 h). Eight pigs received Papuan taipan venom followed by treatment: Briefly: Two animals received antivenom immediately and survived to the end of the study. Two animals received antivenom treatment delayed 45 min from envenoming and died within 4 h. Two animals received similarly delayed antivenom treatment and were rescued by varespladib. Two animals were treated with varespladib alone after a 45-min delay. Treatment with varespladib only was effective but required repeat dosing over the course of the study. Findings highlight both the importance of early treatment and, as well, a half-life for the investigational inhibitors now in Phase II clinical trials for snakebite. Varespladib rapidly reversed weakness even when administered many hours post-envenoming and, overall, our results suggest that varespladib and varespladib-methyl could be efficacious tools in the treatment of sPLA2-induced weakness from Oxyuranus envenoming. Further clinical study as initial therapy and as potential method of rescue from some types of antivenom-resistant envenomings are supported by these data.
Collapse
Affiliation(s)
- Lyndi L. Gilliam
- Department of Veterinary Clinical Sciences, Center for Veterinary Health Sciences, Oklahoma State University, Stillwater, OK 74078, USA; (L.L.G.); (J.G.); (J.R.)
| | - John Gilliam
- Department of Veterinary Clinical Sciences, Center for Veterinary Health Sciences, Oklahoma State University, Stillwater, OK 74078, USA; (L.L.G.); (J.G.); (J.R.)
| | - Stephen P. Samuel
- Division of Research Ophirex, Inc., Corte Madera, CA 94925, USA; (S.P.S.); (R.W.C.); (S.I.S.)
| | - Rebecca W. Carter
- Division of Research Ophirex, Inc., Corte Madera, CA 94925, USA; (S.P.S.); (R.W.C.); (S.I.S.)
| | - Jerry Ritchey
- Department of Veterinary Clinical Sciences, Center for Veterinary Health Sciences, Oklahoma State University, Stillwater, OK 74078, USA; (L.L.G.); (J.G.); (J.R.)
| | - Tommaso Bulfone
- Center for Exploration and Travel Health, California Academy of Sciences, San Francisco, CA 94118, USA; (T.B.)
- School of Medicine, University of California, San Francisco, CA 94143, USA
| | - José María Gutiérrez
- Instituto Clodomiro Picado, Facultad de Microbiología, Universidad de Costa Rica, San José 11501-2060, Costa Rica;
| | - David J. Williams
- Regulation and Prequalification Department (RPQ) at the World Health Organization (WHO), 1211 Geneva, Switzerland;
| | - Daniela M. Durkin
- Center for Exploration and Travel Health, California Academy of Sciences, San Francisco, CA 94118, USA; (T.B.)
| | - Sally I. Stephens
- Division of Research Ophirex, Inc., Corte Madera, CA 94925, USA; (S.P.S.); (R.W.C.); (S.I.S.)
| | - Matthew R. Lewin
- Division of Research Ophirex, Inc., Corte Madera, CA 94925, USA; (S.P.S.); (R.W.C.); (S.I.S.)
- Center for Exploration and Travel Health, California Academy of Sciences, San Francisco, CA 94118, USA; (T.B.)
| |
Collapse
|
19
|
Salvador GHM, Pinto ÊKR, Ortolani PL, Fortes-Dias CL, Cavalcante WLG, Soares AM, Lomonte B, Lewin MR, Fontes MRM. Structural basis of the myotoxic inhibition of the Bothrops pirajai PrTX-I by the synthetic varespladib. Biochimie 2023; 207:1-10. [PMID: 36403756 DOI: 10.1016/j.biochi.2022.11.006] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2022] [Revised: 11/08/2022] [Accepted: 11/14/2022] [Indexed: 11/18/2022]
Abstract
Varespladib (LY315920) is a potent inhibitor of human group IIA phospholipase A2 (PLA2) originally developed to control inflammatory cascades of diseases associated with high or dysregulated levels of endogenous PLA2. Recently, varespladib was also found to inhibit snake venom PLA2 and PLA2-like toxins. Herein, ex vivo neuromuscular blocking activity assays were used to test the inhibitory activity of varespladib. The binding affinity between varespladib and a PLA2-like toxin was quantified and compared with other potential inhibitors for this class of proteins. Crystallographic and bioinformatic studies showed that varespladib binds to PrTX-I and BthTX-I into their hydrophobic channels, similarly to other previously characterized PLA2-like myotoxins. However, a new finding is that an additional varespladib binds to the MDiS region, a particular site that is related to muscle cell disruption by these toxins. The present results further advance the characterization of the molecular interactions of varespladib with PLA2-like myotoxins and provide additional evidence for this compound as a promising inhibitor candidate for different PLA2 and PLA2-like toxins.
Collapse
Affiliation(s)
- Guilherme H M Salvador
- Departamento de Biofísica e Farmacologia, Instituto de Biociências, Universidade Estadual Paulista, Botucatu, SP, Brazil
| | - Êmylle K R Pinto
- Departmento de Farmacologia, Instituto de Ciências Biologicas, Universidade Federal de Minas Gerais (UFMG), Brazil
| | - Paula L Ortolani
- Centro de Pesquisa e Desenvolvimento, Fundação Ezequiel Dias (FUNED), Brazil
| | | | - Walter L G Cavalcante
- Departmento de Farmacologia, Instituto de Ciências Biologicas, Universidade Federal de Minas Gerais (UFMG), Brazil
| | - Andreimar M Soares
- Laboratório de Biotecnologia de Proteínas e Compostos Bioativos Aplicados à Saúde, LABIOPROT, Fundação Oswaldo Cruz, FIOCRUZ, unidade Rondônia e Instituto Nacional de Ciência e Tecnologia de Epidemiologia da Amazônia Ocidental, INCT EPIAMO, Porto Velho, RO, Brazil
| | - Bruno Lomonte
- Instituto Clodomiro Picado, Facultad de Microbiología, Universidad de Costa Rica, San José, Costa Rica
| | - Matthew R Lewin
- Ophirex, Inc. Corte Madera, CA, 94925, USA; Center for Exploration and Travel Health, California Academy of Sciences, San Francisco, CA, 94118, USA
| | - Marcos R M Fontes
- Departamento de Biofísica e Farmacologia, Instituto de Biociências, Universidade Estadual Paulista, Botucatu, SP, Brazil.
| |
Collapse
|
20
|
Structural and functional studies of a snake venom phospholipase A 2-like protein complexed to an inhibitor from Tabernaemontana catharinensis. Biochimie 2023; 206:105-115. [PMID: 36273763 DOI: 10.1016/j.biochi.2022.10.011] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2022] [Revised: 09/19/2022] [Accepted: 10/17/2022] [Indexed: 11/22/2022]
Abstract
Snake envenomation is an ongoing global health problem and tropical neglected disease that afflicts millions of people each year. The only specific treatment, antivenom, has several limitations that affects its proper distribution to the victims and its efficacy against local effects, such as myonecrosis. The main responsible for this consequence are the phospholipases A2 (PLA2) and PLA2-like proteins, such as BthTX-I from Bothrops jararacussu. Folk medicine resorts to plants such as Tabernaemontana catharinensis to palliate these and other snakebite effects. Here, we evaluated the effect of its root bark extract and one of its isolated compounds, 12-methoxy-4-methyl-voachalotine (MMV), against the in vitro paralysis and muscle damage induced by BthTX-I. Secondary and quaternary structures of BthTX-I were not modified by the interaction with MMV. Instead, this compound interacted in an unprecedented way with the region inside the toxin hydrophobic channel and promoted a structural change in Val31, loop 58-71 and Membrane Disruption Site. Thus, we hypothesize that MMV inhibits PLA2-like proteins by preventing entrance of fatty acid into the hydrophobic channel. These data may explain the traditional use of T. catharinensis extract and confirm MMV as a promising candidate to complement antivenom or a structural guide to develop more effective inhibitors.
Collapse
|
21
|
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: 24] [Impact Index Per Article: 12.0] [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
|
22
|
In Vitro Efficacy of Antivenom and Varespladib in Neutralising Chinese Russell's Viper ( Daboia siamensis) Venom Toxicity. Toxins (Basel) 2023; 15:toxins15010062. [PMID: 36668882 PMCID: PMC9864994 DOI: 10.3390/toxins15010062] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Revised: 01/08/2023] [Accepted: 01/09/2023] [Indexed: 01/13/2023] Open
Abstract
The venom of the Russell's viper (Daboia siamensis) contains neurotoxic and myotoxic phospholipase A2 toxins which can cause irreversible damage to motor nerve terminals. Due to the time delay between envenoming and antivenom administration, antivenoms may have limited efficacy against some of these venom components. Hence, there is a need for adjunct treatments to circumvent these limitations. In this study, we examined the efficacy of Chinese D. siamensis antivenom alone, and in combination with a PLA2 inhibitor, Varespladib, in reversing the in vitro neuromuscular blockade in the chick biventer cervicis nerve-muscle preparation. Pre-synaptic neurotoxicity and myotoxicity were not reversed by the addition of Chinese D. siamensis antivenom 30 or 60 min after venom (10 µg/mL). The prior addition of Varespladib prevented the neurotoxic and myotoxic activity of venom (10 µg/mL) and was also able to prevent further reductions in neuromuscular block and muscle twitches when added 60 min after venom. The addition of the combination of Varespladib and antivenom 60 min after venom failed to produce further improvements than Varespladib alone. This demonstrates that the window of time in which antivenom remains effective is relatively short compared to Varespladib and small-molecule inhibitors may be effective in abrogating some activities of Chinese D. siamensis venom.
Collapse
|
23
|
Carter RW, Gerardo CJ, Samuel SP, Kumar S, Kotehal SD, Mukherjee PP, Shirazi FM, Akpunonu PD, Bammigatti C, Bhalla A, Manikath N, Platts-Mills TF, Lewin MR. The BRAVO Clinical Study Protocol: Oral Varespladib for Inhibition of Secretory Phospholipase A2 in the Treatment of Snakebite Envenoming. Toxins (Basel) 2022; 15:22. [PMID: 36668842 PMCID: PMC9862656 DOI: 10.3390/toxins15010022] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2022] [Revised: 12/19/2022] [Accepted: 12/24/2022] [Indexed: 12/30/2022] Open
Abstract
INTRODUCTION Snakebite is an urgent, unmet global medical need causing significant morbidity and mortality worldwide. Varespladib is a potent inhibitor of venom secretory phospholipase A2 (sPLA2) that can be administered orally via its prodrug, varespladib-methyl. Extensive preclinical data support clinical evaluation of varespladib as a treatment for snakebite envenoming (SBE). The protocol reported here was designed to evaluate varespladib-methyl for SBE from any snake species in multiple geographies. METHODS AND ANALYSIS BRAVO (Broad-spectrum Rapid Antidote: Varespladib Oral for snakebite) is a multicenter, randomized, double-blind, placebo-controlled, phase 2 study to evaluate the safety, tolerability, and efficacy of oral varespladib-methyl plus standard of care (SoC) vs. SoC plus placebo in patients presenting with acute SBE by any venomous snake species. Male and female patients 5 years of age and older who meet eligibility criteria will be randomly assigned 1:1 to varespladib-methyl or placebo. The primary outcome is the Snakebite Severity Score (SSS) that has been modified for international use. This composite outcome is based on the sum of the pulmonary, cardiovascular, nervous, hematologic, and renal systems components of the updated SSS. ETHICS AND DISSEMINATION This protocol was submitted to regulatory authorities in India and the US. A Clinical Trial No Objection Certificate from the India Central Drugs Standard Control Organisation, Drug Controller General-India, and a Notice to Proceed from the US Food and Drug Administration have been obtained. The study protocol was approved by properly constituted, valid institutional review boards or ethics committees at each study site. This study is being conducted in compliance with the April 1996 ICH Guidance for Industry GCP E6, the Integrated Addendum to ICH E6 (R2) of November 2016, and the applicable regulations of the country in which the study is conducted. The trial is registered on Clinical trials.gov, NCT#04996264 and Clinical Trials Registry-India, 2021/07/045079 000062.
Collapse
Affiliation(s)
| | - Charles J. Gerardo
- Department of Emergency Medicine, Duke University, Durham, NC 27708, USA
| | | | - Surendra Kumar
- Department of Medicine, Sardar Patel Medical College, PBM Hospital, Bikaner 334001, India
| | - Suneetha D. Kotehal
- Department of Medicine, Mysore Medical College and Research Institute, Mysore 570001, India
| | - Partha P. Mukherjee
- Department of General Medicine, Calcutta National Medical College, Kolkata 700014, India
| | - Farshad M. Shirazi
- Arizona Poison & Drug Information Center, College of Pharmacy and University of Arizona College of Medicine, University of Arizona, Tucson, AZ 85721, USA
| | - Peter D. Akpunonu
- Department of Emergency Medicine and Medical Toxicology, University of Kentucky College of Medicine, Lexington, KY 40506, USA
| | - Chanaveerappa Bammigatti
- Department of Medicine, Jawaharlal Institute of Postgraduate Medical Education and Research, Puducherry 605006, India
| | - Ashish Bhalla
- Department of Internal Medicine, Postgraduate Institute of Medical Education and Research, Chandigarh 160012, India
| | - Neeraj Manikath
- Department of Emergency Medicine, Government Medical College, Kozhikode 673008, India
| | | | | |
Collapse
|
24
|
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: 30] [Impact Index Per Article: 10.0] [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
|
25
|
Cerebral Complications of Snakebite Envenoming: Case Studies. Toxins (Basel) 2022; 14:toxins14070436. [PMID: 35878174 PMCID: PMC9320586 DOI: 10.3390/toxins14070436] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2022] [Revised: 06/21/2022] [Accepted: 06/24/2022] [Indexed: 02/01/2023] Open
Abstract
There are an estimated 5.4 million snakebite cases every year. People with snakebite envenoming suffer from severe complications, or even death. Although some review articles cover several topics of snakebite envenoming, a review of the cases regarding cerebral complications, especially rare syndromes, is lacking. Here, we overview 35 cases of snakebite by front-fanged snakes, including Bothrops, Daboia, Cerastes, Deinagkistrodon, Trimeresurus, and Crotalus in the Viperidae family; Bungarus and Naja in the Elapidae family, and Homoroselaps (rare cases) in the Lamprophiidae family. We also review three rare cases of snakebite by rear-fanged snakes, including Oxybelis and Leptodeira in the Colubridae family. In the cases of viper bites, most patients (17/24) were diagnosed with ischemic stroke and intracranial hemorrhage, leading to six deaths. We then discuss the potential underlying molecular mechanisms that cause these complications. In cases of elapid bites, neural, cardiac, and ophthalmic disorders are the main complications. Due to the small amount of venom injection and the inability to deep bite, all the rear-fanged snakebites did not develop any severe complications. To date, antivenom (AV) is the most effective therapy for snakebite envenoming. In the six cases of viper and elapid bites that did not receive AV, three cases (two by viper and one by elapid) resulted in death. This indicates that AV treatment is the key to survival after a venomous snakebite. Lastly, we also discuss several studies of therapeutic agents against snakebite-envenoming-induced complications, which could be potential adjuvants along with AV treatment. This article organizes the diagnosis of hemotoxic and neurotoxic envenoming, which may help ER doctors determine the treatment for unidentified snakebite.
Collapse
|
26
|
Wang R, Gao D, Yu F, Han J, Yuan H, Hu F. Phospholipase A 2 inhibitor varespladib prevents wasp sting-induced nephrotoxicity in rats. Toxicon 2022; 215:69-76. [PMID: 35724947 DOI: 10.1016/j.toxicon.2022.06.007] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2022] [Revised: 06/09/2022] [Accepted: 06/11/2022] [Indexed: 02/07/2023]
Abstract
This study aimed to clarify whether varespladib, a phospholipase A2 (PLA2) inhibitor, can be used as a therapeutic agent for wasp sting-induced acute kidney injury (AKI). Rats were divided into control, AKI, and AKI + varespladib groups. The AKI model was established by subcutaneously injecting wasp venom at five different sites in rats. Varespladib treatment showed a significant inhibitory effect on wasp venom PLA2in vitro and in vivo. Moreover, we observed that varespladib decreased the levels of rhabdomyolysis and hemolysis markers compared with that in the AKI group. Histopathological changes in the kidney decreased significantly, and rat serum creatinine levels were reduced after varespladib administration. The significantly regulated genes in the kidney of the AKI group were mostly involved in inflammatory response pathway, and the administration of varespladib remarkably attenuated the expression of these genes. Therefore, varespladib inhibited wasp sting-induced functional and pathological damage to the kidneys. We propose that the PLA2 inhibitor varespladib protects the kidney tissue in a wasp sting-induced AKI model by inhibiting PLA2 activity.
Collapse
Affiliation(s)
- Rui Wang
- School of Medicine, Wuhan University of Science and Technology, Wuhan, 430065, China; Department of Nephrology, Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Science, Xiangyang, 441000, China
| | - Dan Gao
- Department of Nephrology, Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Science, Xiangyang, 441000, China
| | - Fanglin Yu
- Department of Nephrology, Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Science, Xiangyang, 441000, China
| | - Jiamin Han
- Department of Nephrology, Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Science, Xiangyang, 441000, China
| | - Hai Yuan
- Department of Nephrology, Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Science, Xiangyang, 441000, China.
| | - Fengqi Hu
- Department of Nephrology, Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Science, Xiangyang, 441000, China.
| |
Collapse
|
27
|
Laustsen A, Gless BH, Jenkins TP, Meyhoff-Madsen M, Bjärtun J, Munk AS, Oscoz S, Fernández J, Gutiérrez JM, Lomonte B, Lohse B. In Vivo Neutralization of Myotoxin II, a Phospholipase A 2 Homologue from Bothrops asper Venom, Using Peptides Discovered via Phage Display Technology. ACS OMEGA 2022; 7:15561-15569. [PMID: 35571794 PMCID: PMC9096979 DOI: 10.1021/acsomega.2c00280] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/17/2022] [Accepted: 03/15/2022] [Indexed: 06/15/2023]
Abstract
Many snake venom toxins cause local tissue damage in prey and victims, which constitutes an important pathology that is challenging to treat with existing antivenoms. One of the notorious toxins that causes such effects is myotoxin II present in the venom of the Central and Northern South American viper, Bothrops asper. This Lys49 PLA2 homologue is devoid of enzymatic activity and causes myotoxicity by disrupting the cell membranes of muscle tissue. To improve envenoming therapy, novel approaches are needed, warranting the discovery and development of inhibitors that target key toxins that are currently difficult to neutralize. Here, we report the identification of a new peptide (JB006), discovered using phage display technology, that is capable of binding to and neutralizing the toxic effects of myotoxin II in vitro and in vivo. Through computational modeling, we further identify hypothetical binding interactions between the toxin and the peptide to enable further development of inhibitors that can neutralize myotoxin II.
Collapse
Affiliation(s)
- Andreas
H. Laustsen
- Department
of Biotechnology and Biomedicine, Technical
University of Denmark, Lyngby DK-2800, Denmark
| | - Bengt H. Gless
- Department
of Drug Design and Pharmacology, University
of Copenhagen, Copenhagen DK-2100, Denmark
| | - Timothy P. Jenkins
- Department
of Biotechnology and Biomedicine, Technical
University of Denmark, Lyngby DK-2800, Denmark
| | - Maria Meyhoff-Madsen
- Department
of Drug Design and Pharmacology, University
of Copenhagen, Copenhagen DK-2100, Denmark
| | - Johanna Bjärtun
- Department
of Drug Design and Pharmacology, University
of Copenhagen, Copenhagen DK-2100, Denmark
| | - Andreas S. Munk
- Department
of Biotechnology and Biomedicine, Technical
University of Denmark, Lyngby DK-2800, Denmark
| | - Saioa Oscoz
- Department
of Biotechnology and Biomedicine, Technical
University of Denmark, Lyngby DK-2800, Denmark
| | - Julián Fernández
- Instituto
Clodomiro Picado, Faculty of Microbiology, University of Costa Rica, San
José 11501-2060, Costa Rica
| | - José María Gutiérrez
- Instituto
Clodomiro Picado, Faculty of Microbiology, University of Costa Rica, San
José 11501-2060, Costa Rica
| | - Bruno Lomonte
- Instituto
Clodomiro Picado, Faculty of Microbiology, University of Costa Rica, San
José 11501-2060, Costa Rica
| | - Brian Lohse
- Department
of Drug Design and Pharmacology, University
of Copenhagen, Copenhagen DK-2100, Denmark
| |
Collapse
|
28
|
Thakshila P, Hodgson WC, Isbister GK, Silva A. In Vitro Neutralization of the Myotoxicity of Australian Mulga Snake ( Pseudechis australis) and Sri Lankan Russell's Viper ( Daboia russelii) Venoms by Australian and Indian Polyvalent Antivenoms. Toxins (Basel) 2022; 14:302. [PMID: 35622549 PMCID: PMC9144940 DOI: 10.3390/toxins14050302] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Revised: 04/20/2022] [Accepted: 04/23/2022] [Indexed: 11/16/2022] Open
Abstract
We studied the neutralisation of Sri Lankan Russell's viper (Daboia russelii) and Australian mulga snake (Pseudechis australis) venom-induced myotoxicity by Indian (Vins and Bharat) and Australian (Seqirus) polyvalent antivenoms, using the in vitro chick biventer skeletal muscle preparation. Prior addition of Bharat or Vins antivenoms abolished D. russelii venom (30 µg/mL)-mediated inhibition of direct twitches, while Australian polyvalent antivenom was not protective. Bharat antivenom prevented, while Vins and Australian polyvalent antivenoms partially prevented, the inhibition of responses to exogenous KCl. Myotoxicity of Mulga venom (10 µg/mL) was fully neutralised by the prior addition of Australian polyvalent antivenom, partially neutralised by Vins antivenom but not by Bharat antivenom. Although the myotoxicity of both venoms was partially prevented by homologous antivenoms when added 5 min after the venom, with an increasing time delay between venom and antivenom, the reversal of myotoxicity gradually decreased. However, antivenoms partially prevented myotoxicity even 60 min after venom. The effect of antivenoms on already initiated myotoxicity was comparable to physical removal of the toxins by washing the bath at similar time points, indicating that the action of the antivenoms on myotoxicity is likely to be due to trapping the toxins or steric hindrance within the circulation, not allowing the toxins to reach target sites in muscles.
Collapse
Affiliation(s)
- Prabhani Thakshila
- Department of Parasitology, Faculty of Medicine and Allied Sciences, Rajarata University of Sri Lanka, Anuradhapura 50008, Sri Lanka;
- South Asian Clinical Toxicology Research Collaboration (SACTRC), Faculty of Medicine, University of Peradeniya, Peradeniya 20400, Sri Lanka;
| | - Wayne C. Hodgson
- Monash Venom Group, Department of Pharmacology, Biomedical Discovery Institute, Monash University, Clayton, VIC 3800, Australia;
| | - Geoffrey K. Isbister
- South Asian Clinical Toxicology Research Collaboration (SACTRC), Faculty of Medicine, University of Peradeniya, Peradeniya 20400, Sri Lanka;
- Clinical Toxicology Research Group, University of Newcastle, Newcastle, NSW 2298, Australia
| | - Anjana Silva
- Department of Parasitology, Faculty of Medicine and Allied Sciences, Rajarata University of Sri Lanka, Anuradhapura 50008, Sri Lanka;
- South Asian Clinical Toxicology Research Collaboration (SACTRC), Faculty of Medicine, University of Peradeniya, Peradeniya 20400, Sri Lanka;
- Monash Venom Group, Department of Pharmacology, Biomedical Discovery Institute, Monash University, Clayton, VIC 3800, Australia;
| |
Collapse
|
29
|
Snake Venomics: Fundamentals, Recent Updates, and a Look to the Next Decade. Toxins (Basel) 2022; 14:toxins14040247. [PMID: 35448856 PMCID: PMC9028316 DOI: 10.3390/toxins14040247] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2022] [Revised: 03/18/2022] [Accepted: 03/21/2022] [Indexed: 01/11/2023] Open
Abstract
Venomic research, powered by techniques adapted from proteomics, transcriptomics, and genomics, seeks to unravel the diversity and complexity of venom through which knowledge can be applied in the treatment of envenoming, biodiscovery, and conservation. Snake venom proteomics is most extensively studied, but the methods varied widely, creating a massive amount of information which complicates data comparison and interpretation. Advancement in mass spectrometry technology, accompanied by growing databases and sophisticated bioinformatic tools, has overcome earlier limitations of protein identification. The progress, however, remains challenged by limited accessibility to samples, non-standardized quantitative methods, and biased interpretation of -omic data. Next-generation sequencing (NGS) technologies enable high-throughput venom-gland transcriptomics and genomics, complementing venom proteomics by providing deeper insights into the structural diversity, differential expression, regulation and functional interaction of the toxin genes. Venomic tissue sampling is, however, difficult due to strict regulations on wildlife use and transfer of biological materials in some countries. Limited resources for techniques and funding are among other pertinent issues that impede the progress of venomics, particularly in less developed regions and for neglected species. Genuine collaboration between international researchers, due recognition of regional experts by global organizations (e.g., WHO), and improved distribution of research support, should be embraced.
Collapse
|
30
|
Varespladib (LY315920) rescued mice from fatal neurotoxicity caused by venoms of five major Asiatic kraits (Bungarus spp.) in an experimental envenoming and rescue model. Acta Trop 2022; 227:106289. [PMID: 34929179 DOI: 10.1016/j.actatropica.2021.106289] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2021] [Revised: 12/16/2021] [Accepted: 12/16/2021] [Indexed: 11/23/2022]
Abstract
The venoms of Asiatic kraits (Bungarus spp.) contain various neurotoxic phospholipases A2 (beta-bungarotoxins) which can irreversibly damage motor nerve terminals, resulting in rapidly fatal suffocation by respiratory muscle paralysis or oral airway obstruction. Hence, there is a need of adjunct therapy at the pre-hospital stage to prevent or delay the onset of neurotoxicity, so that antivenom can be given within golden hour before the envenoming becomes antivenom-resistant. This study investigated the efficacy of varespladib, a small molecule PLA2 (phospholipase A2) inhibitor, given as a bolus subcutaneously upon the onset of krait venom-induced paralysis in a mouse experimental envenoming and rescue model, where the severity of neurotoxicity was scored and the survival rate was monitored over 24 h. Varespladib at 10 mg/kg effectively alleviated the neurotoxicity of Bungarus sindanus, Bungarus multicinctus and Bungarus fasciatus venoms, and rescued all mice from venom-induced lethality (100% survival). Varespladib at this dose, however, only partially reduced the neurotoxicity of Bungarus caeruleus and Bungarus candidus venoms, while all challenged mice were dead by 23 h (B. caeruleus) and 12 h (B. candidus). An increased dose of varespladib at 20 mg/kg markedly abated the venom neurotoxicity past 8 h of envenoming, and protected the mice from venom lethality (B. caeruleus: 75% survival; B. candidus: 100% survival). The finding is consistent with previous studies which demonstrated varespladib's inhibitory effect against some snake venoms. The findings suggest varespladib could be repurposed as an emergency drug for prevention or rescue (if given early enough) from the acute, neurotoxic envenoming syndromes caused by various major krait species in Asia.
Collapse
|
31
|
Gutierres PG, Pereira DR, Vieira NL, Arantes LF, Silva NJ, Torres-Bonilla KA, Hyslop S, Morais-Zani K, Nogueira RMB, Rowan EG, Floriano RS. Action of Varespladib (LY-315920), a Phospholipase A 2 Inhibitor, on the Enzymatic, Coagulant and Haemorrhagic Activities of Lachesis muta rhombeata (South-American Bushmaster) Venom. Front Pharmacol 2022; 12:812295. [PMID: 35095526 PMCID: PMC8790531 DOI: 10.3389/fphar.2021.812295] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2021] [Accepted: 12/09/2021] [Indexed: 01/08/2023] Open
Abstract
Varespladib (VPL) was primarily developed to treat inflammatory disturbances associated with high levels of serum phospholipase A2 (PLA2). VPL has also demonstrated to be a potential antivenom support agent to prevent PLA2-dependent effects produced by snake venoms. In this study, we examined the action of VPL on the coagulant, haemorrhagic and enzymatic activities of Lachesis muta rhombeata (South-American bushmaster) venom. Conventional colorimetric enzymatic assays were performed for PLA2, caseinolytic and esterasic activities; in vitro coagulant activities for prothrombin time (PT) and activated partial thromboplastin time (aPTT) were performed in rat citrated plasma through a quick timer coagulometer, whereas the dimensions of haemorrhagic haloes obtained after i.d. injections of venom in Wistar rats were determined using ImageJ software. Venom (1 mg/ml) exhibited accentuated enzymatic activities for proteases and PLA2in vitro, with VPL abolishing the PLA2 activity from 0.01 mM; VPL did not affect caseinolytic and esterasic activities at any tested concentrations (0.001–1 mM). In rat citrated plasma in vitro, VPL (1 mM) alone efficiently prevented the venom (1 mg/ml)-induced procoagulant disorder associated to extrinsic (PT) pathway, whereas its association with a commercial antivenom successfully prevented changes in both intrinsic (aPTT) and extrinsic (PT) pathways; commercial antivenom by itself failed to avoid the procoagulant disorders by this venom. Venom (0.5 mg/kg)-induced hemorrhagic activity was slightly reduced by VPL (1 mM) alone or combined with antivenom (antivenom:venom ratio 1:3 ‘v/w’) in rats, with antivenom alone producing no protective action on this parameter. In conclusion, VPL does not inhibit other major enzymatic groups of L. m. rhombeata venom, with its high PLA2 antagonize activity efficaciously preventing the venom-induced coagulation disturbances.
Collapse
Affiliation(s)
- Pamella G Gutierres
- Laboratory of Toxinology and Cardiovascular Research, University of Western São Paulo, Presidente Prudente, Brazil
| | - Diego R Pereira
- Laboratory of Toxinology and Cardiovascular Research, University of Western São Paulo, Presidente Prudente, Brazil
| | - Nataly L Vieira
- Laboratory of Toxinology and Cardiovascular Research, University of Western São Paulo, Presidente Prudente, Brazil
| | - Lilian F Arantes
- Graduate Program in Zootechnics, Rural Federal University of Pernambuco, Recife, Brazil
| | - Nelson J Silva
- Graduate Program in Environmental Sciences and Health, School of Medical, Pharmaceutical and Biomedical Sciences, Pontifical Catholic University of Goiás, Goiânia, Brazil
| | - Kristian A Torres-Bonilla
- Department of Pharmacology, Faculty of Medical Sciences, State University of Campinas, Campinas, Brazil
| | - Stephen Hyslop
- Department of Pharmacology, Faculty of Medical Sciences, State University of Campinas, Campinas, Brazil
| | | | - Rosa M B Nogueira
- Laboratory of Toxinology and Cardiovascular Research, University of Western São Paulo, Presidente Prudente, Brazil
| | - Edward G Rowan
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow, United Kingdom
| | - Rafael S Floriano
- Laboratory of Toxinology and Cardiovascular Research, University of Western São Paulo, Presidente Prudente, Brazil
| |
Collapse
|
32
|
Saravia-Otten P, Hernández R, Marroquín N, Pereañez JA, Preciado LM, Vásquez A, García G, Nave F, Rochac L, Genovez V, Mérida M, Cruz SM, Orozco N, Cáceres A, Gutiérrez JM. Inhibition of enzymatic activities of Bothrops asper snake venom and docking analysis of compounds from plants used in Central America to treat snakebite envenoming. JOURNAL OF ETHNOPHARMACOLOGY 2022; 283:114710. [PMID: 34626780 DOI: 10.1016/j.jep.2021.114710] [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: 08/30/2021] [Revised: 09/28/2021] [Accepted: 10/04/2021] [Indexed: 06/13/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Snakebite envenoming is a public health problem of high impact in Central America. Bothrops asper, known as barba amarilla, terciopelo, and equis, is the snake species responsible for most snakebites in Central America. In this region, there is a long-standing tradition on the use of plants in the management of snakebites, especially in indigenous communities. Ethnomedical use of Eryngium foetidum L., Neurolaena lobata (L.) Cass. and Pimenta dioica (L.) Merr. to treat snakebite envenoming has been reported in Belice, Guatemala, Nicaragua, and Costa Rica. Extracts of the leaves of these plants have shown anti-venom activities in in vitro assays in previous studies. AIM OF THE STUDY To assess the ability of organic fractions from these three plants to inhibit enzymatic activities associated with toxicity of the venom of B. asper, and to study, by docking analysis, the interaction of metalloproteinase and phospholipases A2 (PLA2) from B. asper venom with secondary metabolites previously described in these plants. MATERIALS AND METHODS Organic fractions were obtained from these three plant species and their ability to neutralize proteolytic, PLA2 and in vitro coagulant activities of B. asper venom was assessed. A phytochemical analysis was carried out in these fractions. The interaction of secondary metabolites previously described in these plants with three toxins from B. asper venom (a metalloproteinase, a PLA2 and a PLA2 homologue) was investigated by docking analysis. RESULTS The inhibitory activity of plants was mainly concentrated in their polar fractions. Acetonic fraction from P. dioica was the most active against PLA2 activity, while the acetonic fraction of E. foetidum completely inhibited the proteolytic activity of the venom. Coagulant activity was partially inhibited only by the acetone and ethyl acetate fractions of P. dioica. Phytochemical analysis of the most bioactive fractions identified flavonoids, saponins, essential oils, coumarins, alkaloids, tannins and sesquiterpene lactones. Docking analysis revealed high affinity interactions of several secondary metabolites of these plants with residues in the vicinity of the catalytic site of these enzymes and, in the case of PLA2 homologue myotoxin II, in the hydrophobic channel. CONCLUSIONS Various fractions from these plants have inhibitory activity against enzymatic actions of B. asper venom which are directly associated with toxicological effects. Docking analysis showed structural evidence of the interaction of secondary metabolites with three toxins. These observations provide support to the potential of these plants to inhibit relevant toxic components of this snake venom.
Collapse
Affiliation(s)
- Patricia Saravia-Otten
- Departamento de Bioquímica, Escuela de Química Biológica, Facultad de Ciencias Químicas y Farmacia, Universidad de San Carlos de Guatemala, Guatemala.
| | - Rosario Hernández
- Departamento de Bioquímica, Escuela de Química Biológica, Facultad de Ciencias Químicas y Farmacia, Universidad de San Carlos de Guatemala, Guatemala
| | - Nereida Marroquín
- Laboratorio de Investigación de Productos Naturales (Lipronat), Escuela de Química Farmacéutica, Facultad de Ciencias Químicas y Farmacia, Universidad de San Carlos de Guatemala, Guatemala
| | - Jaime A Pereañez
- Toxinología, Alternativas Terapeúticas y Alimentarias, Facultad de Ciencias Farmacéuticas y Alimentarias, Universidad de Antioquia, Medellín, Colombia
| | - Lina M Preciado
- Toxinología, Alternativas Terapeúticas y Alimentarias, Facultad de Ciencias Farmacéuticas y Alimentarias, Universidad de Antioquia, Medellín, Colombia
| | - Allan Vásquez
- Facultad de Medicina, Universidad Francisco Marroquín, Guatemala
| | - Gabriela García
- Departamento de Bioquímica, Escuela de Química Biológica, Facultad de Ciencias Químicas y Farmacia, Universidad de San Carlos de Guatemala, Guatemala
| | - Federico Nave
- Dirección General de Investigaciones (DIGI), Universidad de San Carlos de Guatemala, Guatemala
| | - Lorena Rochac
- Laboratorio de Investigación de Productos Naturales (Lipronat), Escuela de Química Farmacéutica, Facultad de Ciencias Químicas y Farmacia, Universidad de San Carlos de Guatemala, Guatemala
| | - Vicente Genovez
- Departamento de Bioquímica, Escuela de Química Biológica, Facultad de Ciencias Químicas y Farmacia, Universidad de San Carlos de Guatemala, Guatemala
| | - Max Mérida
- Laboratorio de Investigación de Productos Naturales (Lipronat), Escuela de Química Farmacéutica, Facultad de Ciencias Químicas y Farmacia, Universidad de San Carlos de Guatemala, Guatemala
| | - Sully M Cruz
- Laboratorio de Investigación de Productos Naturales (Lipronat), Escuela de Química Farmacéutica, Facultad de Ciencias Químicas y Farmacia, Universidad de San Carlos de Guatemala, Guatemala
| | - Nohemí Orozco
- Departamento de Química Orgánica, Escuela de Química, Facultad de Ciencias Químicas y Farmacia, Universidad de San Carlos de Guatemala, Guatemala
| | - Armando Cáceres
- Laboratorio de Investigación de Productos Naturales (Lipronat), Escuela de Química Farmacéutica, Facultad de Ciencias Químicas y Farmacia, Universidad de San Carlos de Guatemala, Guatemala; Laboratorios de Productos Naturales Farmaya, Guatemala
| | - José M Gutiérrez
- Instituto Clodomiro Picado, Facultad de Microbiología, Universidad de Costa Rica, San José, Costa Rica
| |
Collapse
|
33
|
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: 0.3] [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
|
34
|
Barbosa ED, Lima Neto JX, Bezerra KS, Oliveira JIN, Machado LD, Fulco UL. Quantum Biochemical Investigation of Lys49-PLA 2 from Bothrops moojeni. J Phys Chem B 2021; 125:12972-12980. [PMID: 34793159 DOI: 10.1021/acs.jpcb.1c07298] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Envenomation via snakebites occurs largely in areas where it is harder to access the hospital. Its mortality rate and sequelae acquired by the survivors symbolize a big challenge for antivenom therapy. In particular, the homologous phospholipase A2 (Lys49-PLA2) proteins can induce myonecrosis and are not effectively neutralized by current treatments. Thus, by taking advantage of crystallographic structures of Bothrops moojeni Lys49-PLA2 complexed with VRD (varespladib) and AIN (aspirin), a quantum biochemistry study based on the molecular fractionation with conjugate cap scheme within the density functional theory formalism is performed to unveil these complexes' detailed interaction energies. The calculations revealed that important interactions between ligands and the Lys49-PLA2 pocket could occur up to a pocket radius of r = 6.5 (5.0 Å) for VRD (AIN), with the total interaction energy of the VRD ligand being higher than that of the AIN ligand, which is well-correlated with the experimental binding affinity. Furthermore, we have identified the role played by the amino acids LYS0069, LYS0049, LEU0005, ILE0009, CYS0029, GLY0030, HIS0048, PRO0018, ALA0019, CYS0045, TYR0052, TYR0022, PRO0125*, and PHE0126* (LYS0069, LYS0049, GLY0032, LEU0002, and LEU0005) in the VRD↔Lys49-PLA2 (AIN↔Lys49-PLA2) complex. Our simulations are a valuable tool to support the big challenge for neutralizing the damages in victims of snakebites.
Collapse
Affiliation(s)
- E D Barbosa
- Departamento de Biofísica e Farmacologia, Universidade Federal do Rio Grande do Norte, Natal 59072-970, Rio Grande do Norte, Brazil
| | - J X Lima Neto
- Departamento de Biofísica e Farmacologia, Universidade Federal do Rio Grande do Norte, Natal 59072-970, Rio Grande do Norte, Brazil
| | - K S Bezerra
- Departamento de Biofísica e Farmacologia, Universidade Federal do Rio Grande do Norte, Natal 59072-970, Rio Grande do Norte, Brazil
| | - J I N Oliveira
- Departamento de Biofísica e Farmacologia, Universidade Federal do Rio Grande do Norte, Natal 59072-970, Rio Grande do Norte, Brazil
| | - L D Machado
- Departamento de Física Teórica e Experimental, Universidade Federal do Rio Grande do Norte, Natal 59072-970, Rio Grande do Norte, Brazil
| | - U L Fulco
- Departamento de Biofísica e Farmacologia, Universidade Federal do Rio Grande do Norte, Natal 59072-970, Rio Grande do Norte, Brazil
| |
Collapse
|
35
|
BthTX-II from Bothrops jararacussu venom has variants with different oligomeric assemblies: An example of snake venom phospholipases A 2 versatility. Int J Biol Macromol 2021; 191:255-266. [PMID: 34547312 DOI: 10.1016/j.ijbiomac.2021.09.083] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Revised: 09/10/2021] [Accepted: 09/13/2021] [Indexed: 10/20/2022]
Abstract
Phospholipases A2 (PLA2s) are found in almost every venomous snake family. In snakebites, some PLA2s can quickly cause local myonecrosis, which may lead to permanent sequelae if antivenom is administered belatedly. They hydrolyse phospholipids in membranes through a catalytic calcium ions-dependent mechanism. BthTX-II is a basic PLA2 and the second major component in the venom of Bothrops jararacussu. Herein, using the software SEQUENCE SLIDER, which integrates crystallographic, mass spectrometry and genetic data, we characterized the primary, tertiary and quaternary structure of two BthTX-II variants (called a and b), which diverge in 7 residues. Crystallographic structure BthTX-IIa is in a Tense-state with its distorted calcium binding loop buried in the dimer interface, contrarily, the novel BthTX-IIb structure is a monomer in a Relax-state with a fatty acid in the hydrophobic channel. Structural data in solution reveals that both variants are monomeric in neutral physiological conditions and mostly dimeric in an acidic environment, being catalytic active in both situations. Therefore, we propose two myotoxic mechanisms for BthTX-II, a catalytic one associated with the monomeric assembly, whereas the other has a calcium independent activity related to its C-terminal region, adopting a dimeric conformation similar to PLA2-like proteins.
Collapse
|
36
|
In vivo treatment with varespladib, a phospholipase A 2 inhibitor, prevents the peripheral neurotoxicity and systemic disorders induced by Micrurus corallinus (coral snake) venom in rats. Toxicol Lett 2021; 356:54-63. [PMID: 34774704 DOI: 10.1016/j.toxlet.2021.11.003] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2021] [Revised: 10/14/2021] [Accepted: 11/09/2021] [Indexed: 11/23/2022]
Abstract
In this study, we investigated the action of varespladib (VPL) alone or in combination with a coral snake antivenom (CAV) on the local and systemic effects induced by Micrurus corallinus venom in rats. Adult male Wistar rats were exposed to venom (1.5 mg/kg - i.m.) and immediately treated with CAV (antivenom:venom ratio 1:1.5 'v/w' - i.p.), VPL (0.5 mg/kg - i.p.), or both of these treatments. The animals were monitored for 120 min and then anesthetized to collect blood samples used for haematological and serum biochemical analysis; after euthanasia, skeletal muscle, renal and hepatic tissue samples were collected for histopathological analysis. M. corallinus venom caused local oedema without subcutaneous haemorrhage or apparent necrosis formation, although there was accentuated muscle morphological damage; none of the treatments prevented oedema formation but the combination of CAV and VPL reduced venom-induced myonecrosis. Venom caused neuromuscular paralysis and respiratory impairment in approximately 60 min following envenomation; CAV alone did not prevent the neurotoxic action, whereas VPL alone prevented neurotoxic symptoms developing as did the combination of CAV and VPL. Venom induced significant increase of serum CK and AST release, mostly due to local and systemic myotoxicity, which was partially prevented by the combination of CAV and VPL. The release of hepatotoxic serum biomarkers (LDH and ALP) induced by M. corallinus venom was not prevented by CAV and VPL when individually administered; their combination effectively prevented ALP release. The venom-induced nephrotoxicity (increase in serum creatinine concentration) was prevented by all the treatments. VPL alone or in combination with CAV significantly prevented the venom-induced lymphocytosis. In conclusion, VPL shows to be effective at preventing the neurotoxic, nephrotoxic, and inflammatory activities of M. corallinus venom. In addition, VPL acts synergistically with antivenom to prevent a number of systemic effects caused by M. corallinus venom.
Collapse
|
37
|
Chowdhury A, Lewin MR, Zdenek CN, Carter R, Fry BG. The Relative Efficacy of Chemically Diverse Small-Molecule Enzyme-Inhibitors Against Anticoagulant Activities of African Spitting Cobra ( Naja Species) Venoms. Front Immunol 2021; 12:752442. [PMID: 34691069 PMCID: PMC8529177 DOI: 10.3389/fimmu.2021.752442] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2021] [Accepted: 09/22/2021] [Indexed: 11/23/2022] Open
Abstract
African spitting cobras are unique among cobras for their potent anticoagulant venom activity arising from strong inhibition of Factor Xa. This anticoagulant effect is exerted by venom phospholipase A2 (Group I PLA2) toxins whose activity contributes to the lethality of these species. This anticoagulant toxicity is particularly problematic as it is not neutralized by current antivenoms. Previous work demonstrated this trait for Naja mossambica, N. nigricincta, N. nigricollis, and N. pallida. The present work builds upon previous research by testing across the full taxonomical range of African spitting cobras, demonstrating that N. ashei, N. katiensis, and N. nubiae are also potently anticoagulant through the inhibition of Factor Xa, and therefore the amplification of potent anticoagulant activity occurred at the base of the African spitting cobra radiation. Previous work demonstrated that the enzyme-inhibitor varespladib was able to neutralize this toxic action for N. mossambica, N. nigricincta, N. nigricollis, and N. pallida venoms. The current work demonstrates that varespladib was also able to neutralize N. ashei, N. katiensis, and N. nubiae. Thus varespladib is shown to have broad utility across the full range of African spitting cobras. In addition, we examined the cross-reactivity of the metalloprotease inhibitor prinomastat, which had been previously intriguingly indicated as being capable of neutralizing viperid venom PLA2 (Group II PLA2). In this study prinomastat inhibited the FXa-inhibiting PLA2 toxins of all the African spitting cobras at the same concentration at which it has been shown to inhibit metalloproteases, and thus was comparably effective in its cross-reactivity. In addition we showed that the metalloprotease-inhibitor marimastat was also able to cross-neutralize PLA2 but less effectively than prinomastat. Due to logistical (cold-chain requirement) and efficacy (cross-reactivity across snake species) limitations of traditional antivenoms, particularly in developing countries where snakebite is most common, these small molecule inhibitors (SMIs) might hold great promise as initial, field-based, treatments for snakebite envenoming as well as addressing fundamental limitations of antivenom in the clinical setting where certain toxin effects are unneutralized.
Collapse
Affiliation(s)
- Abhinandan Chowdhury
- Venom Evolution Lab, School of Biological Science, University of Queensland, St. Lucia, QLD, Australia.,Department of Biochemistry & Microbiology, North South University, Dhaka, Bangladesh
| | - Matthew R Lewin
- California Academy of Sciences, San Francisco, CA, United States.,Ophirex, Inc., Corte Madera, CA, United States
| | - Christina N Zdenek
- Venom Evolution Lab, School of Biological Science, University of Queensland, St. Lucia, QLD, Australia
| | | | - Bryan G Fry
- Venom Evolution Lab, School of Biological Science, University of Queensland, St. Lucia, QLD, Australia
| |
Collapse
|
38
|
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: 16] [Impact Index Per Article: 4.0] [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
|
39
|
Yusuf AJ, Aleku GA, Bello UR, Liman DU. Prospects and Challenges of Developing Plant-Derived Snake Antivenin Natural Products: A Focus on West Africa. ChemMedChem 2021; 16:3635-3648. [PMID: 34585514 DOI: 10.1002/cmdc.202100478] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2021] [Revised: 09/23/2021] [Indexed: 11/06/2022]
Abstract
Snakebite envenomation (SBE) is an important public health issue that is now receiving renewed attention following its reclassification as a Neglected Tropical Disease (NTD). Most incidences occur in rural areas of resource-limited countries, as such, timely and appropriate medical care for SBE is often inaccessible. The administration of anti-snake venom serum (ASV) is the only effective definitive treatment of SBE, but treatment failure to available ASVs is not uncommon. Emerging evidence highlights the potential of small-molecule compounds as inhibitors against toxins of snake venom. This presents an encouraging prospect to develop an alternative therapeutic option for the treatment SBE, that may be amenable for use at the point of care in resource-constraint settings. In view of the pivotal role of natural products in modern drug discovery programmes, there is considerable interest in ethno-pharmacological mining of medicinal plants and plant-derived medicinal compounds toward developing novel snake venom-neutralising therapeutics. In this review, we compile a collection of medicinal plants used in the treatment of SBE in West Africa and highlight their promise as potential botanical drugs or as sources of novel small-molecule compounds for the treatment of SBE. The challenges that must be surmounted to bring this to fruition including the need for (sub) regional collaboration have been discussed.
Collapse
Affiliation(s)
- Amina J Yusuf
- Department of Pharmaceutical & Medicinal Chemistry, Usmanu Danfodiyo University, Sokoto, Nigeria
| | - Godwin A Aleku
- Department of Biochemistry, University of Cambridge, Cambridge, CB2 1GA, UK
| | - Usman Rabiu Bello
- Biotechnology unit, Department of Life Sciences, Mewar University, Gangrar, Chittorgarh, Rajasthan, India
| | - Dahiru Umar Liman
- Department of Pharmaceutical & Medicinal Chemistry, Usmanu Danfodiyo University, Sokoto, Nigeria
| |
Collapse
|
40
|
Batsika CS, Gerogiannopoulou ADD, Mantzourani C, Vasilakaki S, Kokotos G. The design and discovery of phospholipase A 2 inhibitors for the treatment of inflammatory diseases. Expert Opin Drug Discov 2021; 16:1287-1305. [PMID: 34143707 DOI: 10.1080/17460441.2021.1942835] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
AREAS COVERED This review article summarizes the most important synthetic PLA2 inhibitors developed to target each one of the four major types of human PLA2 (cytosolic cPLA2, calcium-independent iPLA2, secreted sPLA2, and lipoprotein-associated Lp-PLA2), discussing their in vitro and in vivo activities as well as their recent applications and therapeutic properties. Recent findings on the role of PLA2 in the pathobiology of COVID-19 are also discussed. EXPERT OPINION Although a number of PLA2 inhibitors have entered clinical trials, none has reached the market yet. Lipoprotein-associated PLA2 is now considered a biomarker of vascular inflammation rather than a therapeutic target for inhibitors like darapladib. Inhibitors of cytosolic PLA2 may find topical applications for diseases like atopic dermatitis and psoriasis. Inhibitors of secreted PLA2, varespladib and varespladib methyl, are under investigation for repositioning in snakebite envenoming. A deeper understanding of PLA2 enzymes is needed for the development of novel selective inhibitors. Lipidomic technologies combined with medicinal chemistry approaches may be useful tools toward this goal.
Collapse
Affiliation(s)
| | | | - Christiana Mantzourani
- Department of Chemistry, National and Kapodistrian University of Athens, Panepistimiopolis Greece
| | - Sofia Vasilakaki
- Department of Chemistry, National and Kapodistrian University of Athens, Panepistimiopolis Greece
| | - George Kokotos
- Department of Chemistry, National and Kapodistrian University of Athens, Panepistimiopolis Greece
| |
Collapse
|
41
|
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: 48] [Impact Index Per Article: 12.0] [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
|
42
|
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: 2.3] [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
|
43
|
Clare RH, Hall SR, Patel RN, Casewell NR. Small Molecule Drug Discovery for Neglected Tropical Snakebite. Trends Pharmacol Sci 2021; 42:340-353. [DOI: 10.1016/j.tips.2021.02.005] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2020] [Revised: 02/24/2021] [Accepted: 02/26/2021] [Indexed: 12/31/2022]
|
44
|
Kazandjian TD, Arrahman A, Still KBM, Somsen GW, Vonk FJ, Casewell NR, Wilkinson MC, Kool J. Anticoagulant Activity of Naja nigricollis Venom Is Mediated by Phospholipase A2 Toxins and Inhibited by Varespladib. Toxins (Basel) 2021; 13:toxins13050302. [PMID: 33922825 PMCID: PMC8145175 DOI: 10.3390/toxins13050302] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2021] [Revised: 04/07/2021] [Accepted: 04/18/2021] [Indexed: 12/03/2022] Open
Abstract
Bites from elapid snakes typically result in neurotoxic symptoms in snakebite victims. Neurotoxins are, therefore, often the focus of research relating to understanding the pathogenesis of elapid bites. However, recent evidence suggests that some elapid snake venoms contain anticoagulant toxins which may help neurotoxic components spread more rapidly. This study examines the effects of venom from the West African black-necked spitting cobra (Naja nigricollis) on blood coagulation and identifies potential coagulopathic toxins. An integrated RPLC-MS methodology, coupled with nanofractionation, was first used to separate venom components, followed by MS, proteomics and coagulopathic bioassays. Coagulation assays were performed on both crude and nanofractionated N. nigricollis venom toxins as well as PLA2s and 3FTx purified from the venom. Assays were then repeated with the addition of either the phospholipase A2 inhibitor varespladib or the snake venom metalloproteinase inhibitor marimastat to assess whether either toxin inhibitor is capable of neutralizing coagulopathic venom activity. Subsequent proteomic analysis was performed on nanofractionated bioactive venom toxins using tryptic digestion followed by nanoLC-MS/MS measurements, which were then identified using Swiss-Prot and species-specific database searches. Varespladib, but not marimastat, was found to significantly reduce the anticoagulant activity of N. nigricollis venom and MS and proteomics analyses confirmed that the anticoagulant venom components mostly consisted of PLA2 proteins. We, therefore, conclude that PLA2s are the most likely candidates responsible for anticoagulant effects stimulated by N. nigricollis venom.
Collapse
Affiliation(s)
- Taline D. Kazandjian
- Centre for Snakebite Research and Interventions, Liverpool School of Tropical Medicine, Pembroke Place, Liverpool L3 5QA, UK; (T.D.K.); (N.R.C.)
| | - Arif Arrahman
- Department of Chemistry and Pharmaceutical Sciences, Division of Bioanalytical Chemistry, Faculty of Sciences, Amsterdam Institute of Molecular and Life Sciences (AIMMS), Vrije Universiteit Amsterdam, De Boelelaan 1083, 1081HV Amsterdam, The Netherlands; (A.A.); (K.B.M.S.); (G.W.S.)
- Centre for Analytical Sciences Amsterdam (CASA), 1012WX Amsterdam, The Netherlands
- Faculty of Pharmacy, Kampus Baru UI, Universitas Indonesia, Depok 16424, Indonesia
| | - Kristina B. M. Still
- Department of Chemistry and Pharmaceutical Sciences, Division of Bioanalytical Chemistry, Faculty of Sciences, Amsterdam Institute of Molecular and Life Sciences (AIMMS), Vrije Universiteit Amsterdam, De Boelelaan 1083, 1081HV Amsterdam, The Netherlands; (A.A.); (K.B.M.S.); (G.W.S.)
- Centre for Analytical Sciences Amsterdam (CASA), 1012WX Amsterdam, The Netherlands
| | - Govert W. Somsen
- Department of Chemistry and Pharmaceutical Sciences, Division of Bioanalytical Chemistry, Faculty of Sciences, Amsterdam Institute of Molecular and Life Sciences (AIMMS), Vrije Universiteit Amsterdam, De Boelelaan 1083, 1081HV Amsterdam, The Netherlands; (A.A.); (K.B.M.S.); (G.W.S.)
- Centre for Analytical Sciences Amsterdam (CASA), 1012WX Amsterdam, The Netherlands
| | - Freek J. Vonk
- Naturalis Biodiversity Center, Darwinweg 2, 2333CR Leiden, The Netherlands;
| | - Nicholas R. Casewell
- Centre for Snakebite Research and Interventions, Liverpool School of Tropical Medicine, Pembroke Place, Liverpool L3 5QA, UK; (T.D.K.); (N.R.C.)
| | - Mark C. Wilkinson
- Centre for Snakebite Research and Interventions, Liverpool School of Tropical Medicine, Pembroke Place, Liverpool L3 5QA, UK; (T.D.K.); (N.R.C.)
- Correspondence: (M.C.W.); (J.K.)
| | - Jeroen Kool
- Department of Chemistry and Pharmaceutical Sciences, Division of Bioanalytical Chemistry, Faculty of Sciences, Amsterdam Institute of Molecular and Life Sciences (AIMMS), Vrije Universiteit Amsterdam, De Boelelaan 1083, 1081HV Amsterdam, The Netherlands; (A.A.); (K.B.M.S.); (G.W.S.)
- Centre for Analytical Sciences Amsterdam (CASA), 1012WX Amsterdam, The Netherlands
- Correspondence: (M.C.W.); (J.K.)
| |
Collapse
|
45
|
Salvador GHM, Borges RJ, Lomonte B, Lewin MR, Fontes MRM. The synthetic varespladib molecule is a multi-functional inhibitor for PLA 2 and PLA 2-like ophidic toxins. Biochim Biophys Acta Gen Subj 2021; 1865:129913. [PMID: 33865953 DOI: 10.1016/j.bbagen.2021.129913] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2021] [Revised: 04/11/2021] [Accepted: 04/12/2021] [Indexed: 11/28/2022]
Abstract
BACKGROUND The treatment for snakebites is early administration of antivenom, which can be highly effective in inhibiting the systemic effects of snake venoms, but is less effective in the treatment of extra-circulatory and local effects. To complement standard-of-care treatments such as antibody-based antivenoms, natural and synthetic small molecules have been proposed for the inhibition of key venom components such as phospholipase A2 (PLA2) and PLA2-like toxins. Varespladib (compound LY315920) is a synthetic molecule developed and clinically tested aiming to block inflammatory cascades of several diseases associated with high PLA2s. Recent studies have demonstrated this molecule is able to potently inhibit snake venom catalytic PLA2 and PLA2-like toxins. METHODS In vivo and in vitro techniques were used to evaluate the inhibitory effect of varespladib against MjTX-I. X-ray crystallography was used to reveal details of the interaction between these molecules. A new methodology that combines crystallography, mass spectroscopy and phylogenetic data was used to review its primary sequence. RESULTS Varespladib was able to inhibit the myotoxic and cytotoxic effects of MjTX-I. Structural analysis revealed a particular inhibitory mechanism of MjTX-I when compared to other PLA2-like myotoxin, presenting an oligomeric-independent function. CONCLUSION Results suggest the effectiveness of varespladib for the inhibition of MjTX-I, in similarity with other PLA2 and PLA2-like toxins. GENERAL SIGNIFICANCE Varespladib appears to be a promissory molecule in the treatment of local effects led by PLA2 and PLA2-like toxins (oligomeric dependent and independent), indicating that this is a multifunctional or broadly specific inhibitor for different toxins within this superfamily.
Collapse
Affiliation(s)
- Guilherme H M Salvador
- Departamento de Biofísica e Farmacologia, Instituto de Biociências, Universidade Estadual Paulista (UNESP), Botucatu, SP, Brazil
| | - Rafael J Borges
- Departamento de Biofísica e Farmacologia, Instituto de Biociências, Universidade Estadual Paulista (UNESP), Botucatu, SP, Brazil
| | - Bruno Lomonte
- Instituto Clodomiro Picado, Facultad de Microbiología, Universidad de Costa Rica, San José, Costa Rica
| | - Matthew R Lewin
- Center for Exploration and Travel Health, California Academy of Sciences, San Francisco, CA 94118, USA
| | - Marcos R M Fontes
- Departamento de Biofísica e Farmacologia, Instituto de Biociências, Universidade Estadual Paulista (UNESP), Botucatu, SP, Brazil.
| |
Collapse
|
46
|
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: 1.4] [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
|
47
|
Alangode A, Rajan K, Nair BG. Snake antivenom: Challenges and alternate approaches. Biochem Pharmacol 2020; 181:114135. [DOI: 10.1016/j.bcp.2020.114135] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Revised: 06/25/2020] [Accepted: 07/01/2020] [Indexed: 02/06/2023]
|
48
|
Alangode A, Reick M, Reick M. Sodium oleate, arachidonate, and linoleate enhance fibrinogenolysis by Russell's viper venom proteinases and inhibit FXIIIa; a role for phospholipase A 2 in venom induced consumption coagulopathy. Toxicon 2020; 186:83-93. [PMID: 32755649 DOI: 10.1016/j.toxicon.2020.07.011] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2019] [Revised: 06/02/2020] [Accepted: 07/12/2020] [Indexed: 12/22/2022]
Abstract
Life-threatening symptoms produced by Russell's viper (RV, Daboia russelii) envenomation result largely from venom induced consumption coagulopathy (VICC). VICC is thought to be mediated to a large degree by venom serine and metalloproteinases, as well as by snake venom phospholipase A2 (svPLA2), the most abundant constituent of RV venom (RVV). The observation that the phenolic lipid anacardic acid markedly enhances proteolytic degradation of fibrinogen by RVV proteinases led us to characterize the chemical basis of this phenomenon with results indicating that svPLA2 products may be major contributors to VICC. RESULTS: Of the chemical analogs tested, the anionic detergents sodium dodecyl sulfate, sodium deoxycholate, N-lauryl sodium sarcosine, and the sodium salts of the fatty acids arachidonic, oleic and to a lesser extend linoleic acid were able to enhance fibrinogenolysis by RVV proteinases. Enhanced Fibrinogenolysis (EF) was observed with various venom size exclusion fractions containing different proteinases, and also with trypsin, indicating that conformational changes of the substrate and increased accessibility of otherwise cryptic cleavage sites are likely to be responsible for EF. In addition to enhancing fibrinogenolysis, sodium arachidonate and oleate were found to partially inhibit thrombin induced, factor XIIIa (FXIIIa) mediated ligation of fibrin chains. In clotting experiments with fresh blood RVV was found to disrupt normal coagulation, leading to small, partial clot formation, whereas RVV pretreated with the PLA2 inhibitor Varespladib induced rapid and complete clot formation (after 5 min) compared to blood alone. CONCLUSION: The observations that fatty acid anions and anionic detergents induce conformational changes that render fibrin(ogen) more susceptible to proteolysis by RVV proteinases and that RVV-PLA2 activity (which produces FFA) is required to render blood incoagulable in clotting experiments with RVV indicate a mechanism by which the activity of highly abundant RVV-PLA2 promotes degradation and depletion of fibrin(ogen) resulting in incoagulable blood seen following RVV envenomation (VICC).
Collapse
Affiliation(s)
- Aswathy Alangode
- School of Biotechnology, Amrita Vishwa Vidyapeetham, Clappana P.O., Kollam, 690 525, Kerala, India
| | - Margaret Reick
- School of Biotechnology, Amrita Vishwa Vidyapeetham, Clappana P.O., Kollam, 690 525, Kerala, India
| | - Martin Reick
- School of Biotechnology, Amrita Vishwa Vidyapeetham, Clappana P.O., Kollam, 690 525, Kerala, India.
| |
Collapse
|
49
|
Youngman NJ, Walker A, Naude A, Coster K, Sundman E, Fry BG. Varespladib (LY315920) neutralises phospholipase A 2 mediated prothrombinase-inhibition induced by Bitis snake venoms. Comp Biochem Physiol C Toxicol Pharmacol 2020; 236:108818. [PMID: 32512199 DOI: 10.1016/j.cbpc.2020.108818] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/02/2020] [Revised: 05/26/2020] [Accepted: 06/02/2020] [Indexed: 01/28/2023]
Abstract
Anticoagulant toxicity is a common function of venoms produced by species within the Bitis genus. Potent inhibition of the prothrombinase complex is an identified mechanism of action for the dwarf species B. cornuta and B. xeropaga, along with some localities of B. atropos and B. caudalis. Snake venom phospholipase A2 toxins that inhibit the prothrombinase complex have been identified in snake venom, including an isolated phospholipase A2 toxin from B. caudalis. Current research is investigating the ability of the drug varespladib to inhibit snake venom phospholipase A2 toxins and reduce their toxicity. In particular, varespladib is being investigated as a treatment that could be administered prior to hospital referral which is a major necessity for species such as those from the genus Bitis, due to envenomations often occurring in remote regions of Africa where antivenom is unavailable. Using previously validated coagulation assays, this study aimed to determine if the toxins responsible for inhibition of the prothrombinase complex in the venom of four Bitis species are phospholipase A2 toxins, and if varespladib is able to neutralise this anticoagulant activity. Our results demonstrate that varespladib strongly neutralises the prothrombinase-inhibiting effects of all venoms tested in this study, and that this prothrombinase-inhibiting mechanism of anticoagulant activity is driven by phospholipase A2 class toxins in these four species. This study extends previous reports demonstrating varespladib has broad efficacy for treatment of phospholipase A2 rich snake venoms, indicating it also inhibits their anticoagulant effects mediated by prothrombinase-inhibition.
Collapse
Affiliation(s)
- Nicholas J Youngman
- Venom Evolution Lab, School of Biological Sciences, University of Queensland, St Lucia, QLD 4072, Australia
| | - Andrew Walker
- Institute for Molecular Bioscience, University of Queensland, St Lucia, QLD 4072, Australia
| | - Arno Naude
- Snakebite Assist, Pretoria ZA-0001, South Africa
| | | | - Eric Sundman
- Universeum, Södra Vägen 50, 412 54 Gothenburg, Sweden
| | - Bryan G Fry
- Venom Evolution Lab, School of Biological Sciences, University of Queensland, St Lucia, QLD 4072, Australia.
| |
Collapse
|
50
|
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: 2.8] [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
|