1
|
Alvitigala BY, Dissanayake HA, Weeratunga PN, Padmaperuma PACD, Gooneratne LV, Gnanathasan CA. Haemotoxicity of snakes: a review of pathogenesis, clinical manifestations, novel diagnostics and challenges in management. Trans R Soc Trop Med Hyg 2025:trae058. [PMID: 39749491 DOI: 10.1093/trstmh/trae058] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2024] [Revised: 06/29/2024] [Accepted: 08/29/2024] [Indexed: 01/04/2025] Open
Abstract
Haemotoxicity is the most common complication of systemic envenoming following snakebite, leading to diverse clinical syndromes ranging from haemorrhagic to prothrombotic manifestations. Key haematological abnormalities include platelet dysfunction, venom-induced consumption coagulopathy, anticoagulant coagulopathy and organ-threatening thrombotic microangiopathy. Diagnostic methods include the bedside whole blood clotting test, laboratory coagulation screening and other advanced methods such as thromboelastogram and clot strength analysis. The primary management strategies are venom neutralisation with antivenom and correction of coagulopathy with blood component transfusions, while options such as plasma exchange are utilised in certain cases. Recent advancements in understanding the pathogenesis of haemotoxicity have facilitated the development of new diagnostic and treatment modalities. This review summarises current knowledge on the pathogenesis, diagnosis, clinical and laboratory manifestations and treatment of the haematological effects of snake envenoming. Furthermore, it highlights important challenges concerning diagnosis and management. Addressing these challenges is crucial for achieving the WHO's goal of reducing deaths and disabilities caused by snakebites by 2030.
Collapse
Affiliation(s)
| | - Harsha A Dissanayake
- Department of Clinical Medicine, Faculty of Medicine, University of Colombo, P.O. 00800, Sri Lanka
| | - Praveen N Weeratunga
- Department of Clinical Medicine, Faculty of Medicine, University of Colombo, P.O. 00800, Sri Lanka
| | | | | | | |
Collapse
|
2
|
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:d4sc06511e. [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
|
3
|
Wiezel GA, Oliveira IS, Ferreira IG, Bordon KCF, Arantes EC. Hyperglycosylation impairs the inhibitory activity of rCdtPLI2, the first recombinant beta-phospholipase A 2 inhibitor. Int J Biol Macromol 2024; 280:135581. [PMID: 39270892 DOI: 10.1016/j.ijbiomac.2024.135581] [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: 05/01/2024] [Revised: 08/24/2024] [Accepted: 09/10/2024] [Indexed: 09/15/2024]
Abstract
Crotoxin, a phospholipase A2 (PLA2) complex and the major Crotalus venom component, is responsible for the main symptoms described in crotalic snakebite envenomings and a key target for PLA2 inhibitors (PLIs). PLIs comprise the alpha, beta and gamma families, and, due to a lack of reports on beta-PLIs, this study aimed to heterologously express CdtPLI2 from Crotalus durissus terrificus venom gland to improve the knowledge of the neglected beta-PLI family. Thereby, recombinant CdtPLI2 (rCdtPLI2) was produced in the eukaryotic Pichia pastoris system to keep some native post-translational modifications. rCdtPLI2 (~41 kDa) presents both N- and O-linked glycans. Alpha-mannosidase digested-rCdtPLI2 (1 mol) strongly inhibited (73%) CB-Cdc catalytic activity (5 moles), demonstrating that glycosylations performed by P. pastoris affect rCdtPLI2 action. Digested-rCdtPLI2 also inhibited PLA2s from diverse Brazilian snake venoms. Furthermore, rCdtPLI2 (1 mol) abolished the catalytic activity of Lmr-PLA2 (5 moles) and reduced the CTx-Cdc (5 moles) enzyme activity by 65%, suppressing basic and acidic snake venom PLA2s. Additionally, crotalic antivenom did not recognize rCdtPLI2, suggesting a lack of neutralization by antivenom antibodies. These findings demonstrate that studying snake venom components may reveal interesting novel molecules to be studied in the snakebite treatment and help to understand these underexplored inhibitors.
Collapse
Affiliation(s)
- Gisele A Wiezel
- Department of BioMolecular Sciences, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Av. do Café, s/n, 14040-903 Ribeirão Preto, SP, Brazil
| | - Isadora S Oliveira
- Department of BioMolecular Sciences, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Av. do Café, s/n, 14040-903 Ribeirão Preto, SP, Brazil
| | - Isabela G Ferreira
- Department of BioMolecular Sciences, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Av. do Café, s/n, 14040-903 Ribeirão Preto, SP, Brazil
| | - Karla C F Bordon
- Department of BioMolecular Sciences, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Av. do Café, s/n, 14040-903 Ribeirão Preto, SP, Brazil
| | - Eliane C Arantes
- Department of BioMolecular Sciences, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Av. do Café, s/n, 14040-903 Ribeirão Preto, SP, Brazil.
| |
Collapse
|
4
|
Jones L, Lay M, Neri-Castro E, Zarzosa V, Hodgson WC, Lewin M, Fry BG. Breaking muscle: neurotoxic and myotoxic effects of Central American snake venoms and the relative efficacies of antivenom and varespladib. BMC Biol 2024; 22:243. [PMID: 39443999 PMCID: PMC11515554 DOI: 10.1186/s12915-024-02044-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2024] [Accepted: 10/14/2024] [Indexed: 10/25/2024] Open
Abstract
BACKGROUND The snake genera Atropoides, Cerrophidion, and Metlapilcoatlus form a clade of neotropical pit vipers distributed across Mexico and Central America. This study evaluated the myotoxic and neurotoxic effects of nine species of Atropoides, Cerrophidion, and Metlapilcoatlus, and the neutralising efficacy of the ICP antivenom from Costa Rica against these effects, in the chick biventer cervicis nerve-muscle preparation. Given the prominence of PLA2s within the venom proteomes of these species, we also aimed to determine the neutralising potency of the PLA2 inhibitor, varespladib. RESULTS All venoms showed myotoxic and potential neurotoxic effects, with differential intra-genera and inter-genera potency. This variation was also seen in the antivenom ability to neutralise the muscle damaging pathophysiological effects observed. Variation was also seen in the relative response to the PLA2 inhibitor varespladib. While the myotoxic effects of M. mexicanus and M. nummifer venoms were effectively neutralised by varespladib, indicating myotoxicity is PLA2 mediated, those of C. godmani and M. olmec venoms were not, revealing that the myotoxicity is driven by non-PLA2 toxin types. CONCLUSIONS This study characterises the myotoxic and neurotoxic venom activity, as well as neutralisation of venom effects from the Atropoides, Cerrophidion, and Metlapilcoatlus clade of American crotalids. Our findings contribute significant clinical and evolutionary knowledge to a clade of poorly researched snakes. In addition, these results provide a platform for future research into the reciprocal interaction between ecological niche specialisation and venom evolution, as well as highlighting the need to test purified toxins to accurately evaluate the potential effects observed in these venoms.
Collapse
Affiliation(s)
- Lee Jones
- Adaptive Biotoxicology Lab, School of the Environment, University of Queensland, St Lucia, QLD, 4072, Australia.
| | - Mimi Lay
- Monash Venom Group, Department of Pharmacology, Biomedical Discovery Institute, Monash University, Clayton, VIC, 3800, Australia
| | - Edgar Neri-Castro
- Facultad de Ciencias Biológicas, Investigador Por México, CONAHCYT, Universidad Juárez del Estado de Durango, Avenida Universidad S/N. Fracc. Filadelfia, Gómez Palacio, Dgo.,, C.P. 35010, México
| | - Vanessa Zarzosa
- Departamento de Medicina Molecular y Bioprocesos, Instituto de Biotecnología, Universidad Nacional Autónoma de México, Av. Universidad 2001, 62210, Cuernavaca, Mexico
| | - Wayne C Hodgson
- Monash Venom Group, Department of Pharmacology, Biomedical Discovery Institute, Monash University, Clayton, VIC, 3800, Australia
| | | | - Bryan G Fry
- Adaptive Biotoxicology Lab, School of the Environment, University of Queensland, St Lucia, QLD, 4072, Australia.
| |
Collapse
|
5
|
Fu K, Zhao J, Zhong L, Xu H, Yu X, Bi X, Huang C. Dual therapy with phospholipase and metalloproteinase inhibitors from Sinonatrix annularis alleviated acute kidney and liver injury caused by multiple snake venoms. Biomed Pharmacother 2024; 177:116967. [PMID: 38908206 DOI: 10.1016/j.biopha.2024.116967] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Revised: 06/10/2024] [Accepted: 06/15/2024] [Indexed: 06/24/2024] Open
Abstract
Snakebite envenomation often induces acute kidney injury (AKI) and acute liver injury (ALI), leading to augmented injuries and poor rehabilitation. Phospholipase A2 (PLA2) and metalloproteinase (SVMP) present in venom are responsible for the envenomation-associated events. In this study, mice envenomed with Deinagkistrodon acutus, Naja atra, or Agkistrodon halys pallas venom exhibited typical AKI and ALI symptoms, including significantly increased plasma levels of myoglobin, free hemoglobin, uric acid, aspartate aminotransferase, and alanine aminotransferase and upregulated expression of kidney NGAL and KIM-1. These effects were significantly inhibited when the mice were pretreated with natural inhibitors of PLA2 and SVMP isolated from Sinonatrix annularis (SaPLIγ and SaMPI). The inhibitors protected the physiological structural integrity of the renal tubules and glomeruli, alleviating inflammatory infiltration and diffuse hemorrhage in the liver. Furthermore, the dual therapy alleviated oxidative stress and apoptosis in the kidneys and liver by mitigating mitochondrial damage, thereby effectively reducing the lethal effect of snake venom in the inhibitor-treated mouse model. This study showed that dual therapy with inhibitors of metalloproteinase and phospholipase can effectively prevent ALI and AKI caused by snake bites. Our findings suggest that intrinsic inhibitors present in snakes are prospective therapeutic agents for multi-organ injuries caused by snake envenoming.
Collapse
Affiliation(s)
- Kepu Fu
- School of Basic Medical Sciences, Jiangxi Medical college, Nanchang University, Nanchang, Jiangxi 330006, China
| | - Jianqi Zhao
- School of Basic Medical Sciences, Jiangxi Medical college, Nanchang University, Nanchang, Jiangxi 330006, China
| | - Lipeng Zhong
- Clinical Laboratory Center, The First Affiliated Hospital, Jiangxi Medical college, Nanchang University, Nanchang, Jiangxi 330209, China
| | - Haiyan Xu
- Blood Transfusion Department, The First Affiliated Hospital of Xi'an Jiao Tong University, Xi'an, Shaanxi, China
| | - Xinhui Yu
- School of Basic Medical Sciences, Jiangxi Medical college, Nanchang University, Nanchang, Jiangxi 330006, China
| | - Xiaowen Bi
- School of Basic Medical Sciences, Jiangxi Medical college, Nanchang University, Nanchang, Jiangxi 330006, China
| | - Chunhong Huang
- School of Basic Medical Sciences, Jiangxi Medical college, Nanchang University, Nanchang, Jiangxi 330006, China.
| |
Collapse
|
6
|
Bartlett KE, Hall SR, Rasmussen SA, Crittenden E, Dawson CA, Albulescu LO, Laprade W, Harrison RA, Saviola AJ, Modahl CM, Jenkins TP, Wilkinson MC, Gutiérrez JM, Casewell NR. Dermonecrosis caused by a spitting cobra snakebite results from toxin potentiation and is prevented by the repurposed drug varespladib. Proc Natl Acad Sci U S A 2024; 121:e2315597121. [PMID: 38687786 PMCID: PMC11087757 DOI: 10.1073/pnas.2315597121] [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: 09/30/2023] [Accepted: 02/26/2024] [Indexed: 05/02/2024] Open
Abstract
Snakebite envenoming is a neglected tropical disease that causes substantial mortality and morbidity globally. The venom of African spitting cobras often causes permanent injury via tissue-destructive dermonecrosis at the bite site, which is ineffectively treated by current antivenoms. To address this therapeutic gap, we identified the etiological venom toxins in Naja nigricollis venom responsible for causing local dermonecrosis. While cytotoxic three-finger toxins were primarily responsible for causing spitting cobra cytotoxicity in cultured keratinocytes, their potentiation by phospholipases A2 toxins was essential to cause dermonecrosis in vivo. This evidence of probable toxin synergism suggests that a single toxin-family inhibiting drug could prevent local envenoming. We show that local injection with the repurposed phospholipase A2-inhibiting drug varespladib significantly prevents local tissue damage caused by several spitting cobra venoms in murine models of envenoming. Our findings therefore provide a therapeutic strategy that may effectively prevent life-changing morbidity caused by snakebite in rural Africa.
Collapse
Affiliation(s)
- Keirah E. Bartlett
- Centre for Snakebite Research & Interventions, Department of Tropical Disease Biology, Liverpool School of Tropical Medicine, LiverpoolL3 5QA, United Kingdom
| | - Steven R. Hall
- Centre for Snakebite Research & Interventions, Department of Tropical Disease Biology, Liverpool School of Tropical Medicine, LiverpoolL3 5QA, United Kingdom
- Centre for Drugs & Diagnostics, Department of Tropical Disease Biology, Liverpool School of Tropical Medicine, LiverpoolL3 5QA, United Kingdom
| | - Sean A. Rasmussen
- Department of Pathology and Laboratory Medicine, Queen Elizabeth II Health Sciences Centre and Dalhousie University, Halifax, NSB3H 1V8, Canada
| | - Edouard Crittenden
- Centre for Snakebite Research & Interventions, Department of Tropical Disease Biology, Liverpool School of Tropical Medicine, LiverpoolL3 5QA, United Kingdom
| | - Charlotte A. Dawson
- Centre for Snakebite Research & Interventions, Department of Tropical Disease Biology, Liverpool School of Tropical Medicine, LiverpoolL3 5QA, United Kingdom
| | - Laura-Oana Albulescu
- Centre for Snakebite Research & Interventions, Department of Tropical Disease Biology, Liverpool School of Tropical Medicine, LiverpoolL3 5QA, United Kingdom
- Centre for Drugs & Diagnostics, Department of Tropical Disease Biology, Liverpool School of Tropical Medicine, LiverpoolL3 5QA, United Kingdom
| | - William Laprade
- Department of Applied Mathematics and Computer Science, Technical University of Denmark, Kongens LyngbyDK-2800, Denmark
| | - Robert A. Harrison
- Centre for Snakebite Research & Interventions, Department of Tropical Disease Biology, Liverpool School of Tropical Medicine, LiverpoolL3 5QA, United Kingdom
- Centre for Drugs & Diagnostics, Department of Tropical Disease Biology, Liverpool School of Tropical Medicine, LiverpoolL3 5QA, United Kingdom
| | - Anthony J. Saviola
- Department of Biochemistry and Molecular Genetics, University of Colorado Denver, Aurora, CO80045
| | - Cassandra M. Modahl
- Centre for Snakebite Research & Interventions, Department of Tropical Disease Biology, Liverpool School of Tropical Medicine, LiverpoolL3 5QA, United Kingdom
| | - Timothy P. Jenkins
- Department of Biotechnology and Biomedicine, Technical University of Denmark, Kongens LyngbyDK-2800, Denmark
| | - Mark C. Wilkinson
- Centre for Snakebite Research & Interventions, Department of Tropical Disease Biology, Liverpool School of Tropical Medicine, LiverpoolL3 5QA, United Kingdom
| | - José María Gutiérrez
- Instituto Clodomiro Picado, Facultad de Microbiología, Universidad de Costa Rica, San José11501–2060, Costa Rica
| | - Nicholas R. Casewell
- Centre for Snakebite Research & Interventions, Department of Tropical Disease Biology, Liverpool School of Tropical Medicine, LiverpoolL3 5QA, United Kingdom
- Centre for Drugs & Diagnostics, Department of Tropical Disease Biology, Liverpool School of Tropical Medicine, LiverpoolL3 5QA, United Kingdom
| |
Collapse
|
7
|
Zhao W, Liu J, Wang S, Tao Q, Lei Q, Huang C. Varespladib mitigates acute liver injury via suppression of excessive mitophagy on Naja atra envenomed mice by inhibiting PLA 2. Toxicon 2024; 242:107694. [PMID: 38556061 DOI: 10.1016/j.toxicon.2024.107694] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2024] [Revised: 03/05/2024] [Accepted: 03/19/2024] [Indexed: 04/02/2024]
Abstract
Snakebite envenomation often leads to severe visceral injuries, including acute liver injury (ALI). However, the toxicity mechanism remains unclear. Moreover, varespladib can directly inhibit phospholipase A2 (PLA2) in snake venom, but its protective effect on snakebite-induced ALI and the mechanism have not been clarified. Previous studies have shown that snake venom PLA2 leads to neuron cell death via reactive oxygen species (ROS), one of the initial factors related to the mitophagy pathway. The present study group also found that ROS accumulation occurred after Naja atra envenoming. Hematoxylin and eosin (H/E) staining and immunohistochemistry (IHC) were performed to identify the expression of inflammatory factors in the liver tissue, and flow cytometry and immunofluorescence were used to detect ROS levels and mitochondrial function. Immunofluorescence and western blotting were also used for detecting mitophagy pathway-related proteins. The results showed that N. atra bite induced ALI by activating mitophagy and inducing inflammation and that varespladib had a protective effect. Collectively, these results showed the pathological mechanism of ALI caused by N. atra bite and revealed the protective effect of varespladib.
Collapse
Affiliation(s)
- Wenjie Zhao
- School of Basic Medicine Sciences, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, 330006, China
| | - Jiahao Liu
- School of Basic Medicine Sciences, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, 330006, China
| | - Sidan Wang
- Queen Mary School, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, 330006, China
| | - Qinqin Tao
- School of Basic Medicine Sciences, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, 330006, China
| | - Qiongqiong Lei
- School of Nursing, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, 330006, China.
| | - Chunhong Huang
- School of Basic Medicine Sciences, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, 330006, China.
| |
Collapse
|
8
|
Geng H, Li R, Teng L, Yu C, Wang W, Gao K, Li A, Liu S, Xing R, Yu H, Li P. Exploring the Efficacy of Hydroxybenzoic Acid Derivatives in Mitigating Jellyfish Toxin-Induced Skin Damage: Insights into Protective and Reparative Mechanisms. Mar Drugs 2024; 22:205. [PMID: 38786596 PMCID: PMC11122885 DOI: 10.3390/md22050205] [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: 04/07/2024] [Revised: 04/23/2024] [Accepted: 04/25/2024] [Indexed: 05/25/2024] Open
Abstract
The escalation of jellyfish stings has drawn attention to severe skin reactions, underscoring the necessity for novel treatments. This investigation assesses the potential of hydroxybenzoic acid derivatives, specifically protocatechuic acid (PCA) and gentisic acid (DHB), for alleviating Nemopilema nomurai Nematocyst Venom (NnNV)-induced injuries. By employing an in vivo mouse model, the study delves into the therapeutic efficacy of these compounds. Through a combination of ELISA and Western blot analyses, histological examinations, and molecular assays, the study scrutinizes the inflammatory response, assesses skin damage and repair mechanisms, and investigates the compounds' ability to counteract venom effects. Our findings indicate that PCA and DHB significantly mitigate inflammation by modulating critical cytokines and pathways, altering collagen ratios through topical application, and enhancing VEGF and bFGF levels. Furthermore, both compounds demonstrate potential in neutralizing NnNV toxicity by inhibiting metalloproteinases and phospholipase-A2, showcasing the viability of small-molecule compounds in managing toxin-induced injuries.
Collapse
Affiliation(s)
- Hao Geng
- CAS and Shandong Province Key Laboratory of Experimental Marine Biology, Center for Ocean Mega-Science, Institute of Oceanology, Chinese Academy of Sciences, Qingdao 266071, China; (H.G.)
- Laboratory for Marine Drugs and Bioproducts, Pilot National Laboratory for Marine Science and Technology (Qingdao), No. 1 Wenhai Road, Qingdao 266237, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Rongfeng Li
- CAS and Shandong Province Key Laboratory of Experimental Marine Biology, Center for Ocean Mega-Science, Institute of Oceanology, Chinese Academy of Sciences, Qingdao 266071, China; (H.G.)
- Laboratory for Marine Drugs and Bioproducts, Pilot National Laboratory for Marine Science and Technology (Qingdao), No. 1 Wenhai Road, Qingdao 266237, China
| | - Lichao Teng
- CAS and Shandong Province Key Laboratory of Experimental Marine Biology, Center for Ocean Mega-Science, Institute of Oceanology, Chinese Academy of Sciences, Qingdao 266071, China; (H.G.)
- Laboratory for Marine Drugs and Bioproducts, Pilot National Laboratory for Marine Science and Technology (Qingdao), No. 1 Wenhai Road, Qingdao 266237, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Chunlin Yu
- CAS and Shandong Province Key Laboratory of Experimental Marine Biology, Center for Ocean Mega-Science, Institute of Oceanology, Chinese Academy of Sciences, Qingdao 266071, China; (H.G.)
- Laboratory for Marine Drugs and Bioproducts, Pilot National Laboratory for Marine Science and Technology (Qingdao), No. 1 Wenhai Road, Qingdao 266237, China
| | - Wenjie Wang
- CAS and Shandong Province Key Laboratory of Experimental Marine Biology, Center for Ocean Mega-Science, Institute of Oceanology, Chinese Academy of Sciences, Qingdao 266071, China; (H.G.)
- Laboratory for Marine Drugs and Bioproducts, Pilot National Laboratory for Marine Science and Technology (Qingdao), No. 1 Wenhai Road, Qingdao 266237, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Kun Gao
- CAS and Shandong Province Key Laboratory of Experimental Marine Biology, Center for Ocean Mega-Science, Institute of Oceanology, Chinese Academy of Sciences, Qingdao 266071, China; (H.G.)
- Laboratory for Marine Drugs and Bioproducts, Pilot National Laboratory for Marine Science and Technology (Qingdao), No. 1 Wenhai Road, Qingdao 266237, China
| | - Aoyu Li
- CAS and Shandong Province Key Laboratory of Experimental Marine Biology, Center for Ocean Mega-Science, Institute of Oceanology, Chinese Academy of Sciences, Qingdao 266071, China; (H.G.)
- Laboratory for Marine Drugs and Bioproducts, Pilot National Laboratory for Marine Science and Technology (Qingdao), No. 1 Wenhai Road, Qingdao 266237, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Song Liu
- CAS and Shandong Province Key Laboratory of Experimental Marine Biology, Center for Ocean Mega-Science, Institute of Oceanology, Chinese Academy of Sciences, Qingdao 266071, China; (H.G.)
- Laboratory for Marine Drugs and Bioproducts, Pilot National Laboratory for Marine Science and Technology (Qingdao), No. 1 Wenhai Road, Qingdao 266237, China
| | - Ronge Xing
- CAS and Shandong Province Key Laboratory of Experimental Marine Biology, Center for Ocean Mega-Science, Institute of Oceanology, Chinese Academy of Sciences, Qingdao 266071, China; (H.G.)
- Laboratory for Marine Drugs and Bioproducts, Pilot National Laboratory for Marine Science and Technology (Qingdao), No. 1 Wenhai Road, Qingdao 266237, China
| | - Huahua Yu
- CAS and Shandong Province Key Laboratory of Experimental Marine Biology, Center for Ocean Mega-Science, Institute of Oceanology, Chinese Academy of Sciences, Qingdao 266071, China; (H.G.)
- Laboratory for Marine Drugs and Bioproducts, Pilot National Laboratory for Marine Science and Technology (Qingdao), No. 1 Wenhai Road, Qingdao 266237, China
| | - Pengcheng Li
- CAS and Shandong Province Key Laboratory of Experimental Marine Biology, Center for Ocean Mega-Science, Institute of Oceanology, Chinese Academy of Sciences, Qingdao 266071, China; (H.G.)
- Laboratory for Marine Drugs and Bioproducts, Pilot National Laboratory for Marine Science and Technology (Qingdao), No. 1 Wenhai Road, Qingdao 266237, China
| |
Collapse
|
9
|
Kumar A, Madni ZK, Chaturvedi S, Salunke DM. Recombinant human scFv antibody fragments against phospholipase A2 from Naja naja and Echis carinatus snake venoms: In vivo neutralization and mechanistic insights. Mol Immunol 2024; 165:55-67. [PMID: 38154407 DOI: 10.1016/j.molimm.2023.12.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2023] [Revised: 11/07/2023] [Accepted: 12/17/2023] [Indexed: 12/30/2023]
Abstract
Snake envenomation results in a range of clinical sequelae, and widely used animal-based conventional antivenoms exhibit several limitations including the adverse immunological effects in human snake bite victims. Therefore, human monoclonal anti-snake venom antibodies or fragments can be an alternate therapy for overcoming the existing limitations. We developed venom-neutralizing humanized scFv antibodies and analyzed biochemical mechanisms associated with the inhibition of toxicity. Tomlinson I and J human scFv antibody libraries were screened against Naja naja and Echis carinatus venoms, and seven unique scFv antibodies were obtained. Further, specific toxins of snake venom interacting with each of these scFvs were identified, and phospholipase A2 (PLA2) was found to be prominently captured by the phage-anchored scFv antibodies. Our study indicated PLA2 to be one of the abundant toxins in Naja naja and Echis carinatus venom samples. The scFvs binding to PLA2 were used to perform in vivo survival assay using the mouse model and in vitro toxin inhibition assays. scFv N194, which binds to acidic PLA2, protected 50% of mice treated with Naja naja venom. Significant prolongation of survival time and 16% survival were observed in Echis carinatus venom-challenged mice treated with scFv E113 and scFv E10, respectively. However, a combination comprised of an equal amount of two scFvs, E113 and E10, both interacting with basic PLA2, exhibited synergistically enhanced survival of 33% in Echis carinatus venom-challenged mice. No such synergistically enhanced survival was observed in the case of combinatorial treatment with anti-Naja naja scFvs, N194, and N248. These scFvs demonstrated partial inhibition of venom-induced myotoxicity, and E113 also inhibited hemolysis by 50%, which corroborates the enhanced survival during combinatorial treatment in Echis carinatus venom-challenged mice.
Collapse
Affiliation(s)
- Amit Kumar
- International Centre for Genetic Engineering and Biotechnology, Aruna Asaf Ali Marg, New Delhi 110067, India
| | - Zaid Kamal Madni
- International Centre for Genetic Engineering and Biotechnology, Aruna Asaf Ali Marg, New Delhi 110067, India
| | - Shivam Chaturvedi
- International Centre for Genetic Engineering and Biotechnology, Aruna Asaf Ali Marg, New Delhi 110067, India
| | - Dinakar M Salunke
- International Centre for Genetic Engineering and Biotechnology, Aruna Asaf Ali Marg, New Delhi 110067, India.
| |
Collapse
|
10
|
Hall SR, Rasmussen SA, Crittenden E, Dawson CA, Bartlett KE, Westhorpe AP, Albulescu LO, Kool J, Gutiérrez JM, Casewell NR. Repurposed drugs and their combinations prevent morbidity-inducing dermonecrosis caused by diverse cytotoxic snake venoms. Nat Commun 2023; 14:7812. [PMID: 38097534 PMCID: PMC10721902 DOI: 10.1038/s41467-023-43510-w] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Accepted: 11/11/2023] [Indexed: 12/17/2023] Open
Abstract
Morbidity from snakebite envenoming affects approximately 400,000 people annually. Tissue damage at the bite-site often leaves victims with catastrophic life-long injuries and is largely untreatable by current antivenoms. Repurposed small molecule drugs that inhibit specific snake venom toxins show considerable promise for tackling this neglected tropical disease. Using human skin cell assays as an initial model for snakebite-induced dermonecrosis, we show that the drugs 2,3-dimercapto-1-propanesulfonic acid (DMPS), marimastat, and varespladib, alone or in combination, inhibit the cytotoxicity of a broad range of medically important snake venoms. Thereafter, using preclinical mouse models of dermonecrosis, we demonstrate that the dual therapeutic combinations of DMPS or marimastat with varespladib significantly inhibit the dermonecrotic activity of geographically distinct and medically important snake venoms, even when the drug combinations are delivered one hour after envenoming. These findings strongly support the future translation of repurposed drug combinations as broad-spectrum therapeutics for preventing morbidity caused by snakebite.
Collapse
Affiliation(s)
- Steven R Hall
- Centre for Snakebite Research & Interventions, Liverpool School of Tropical Medicine, Pembroke Place, Liverpool, L3 5QA, UK
- Centre for Drugs & Diagnostics, Liverpool School of Tropical Medicine, Pembroke Place, Liverpool, L3 5QA, UK
| | - Sean A Rasmussen
- Department of Pathology and Laboratory Medicine, Queen Elizabeth II Health Sciences Centre and Dalhousie University, 7th Floor of MacKenzie Building, 5788 University Avenue, Halifax, NS, B3H 1V8, Canada
| | - Edouard Crittenden
- Centre for Snakebite Research & Interventions, Liverpool School of Tropical Medicine, Pembroke Place, Liverpool, L3 5QA, UK
| | - Charlotte A Dawson
- Centre for Snakebite Research & Interventions, Liverpool School of Tropical Medicine, Pembroke Place, Liverpool, L3 5QA, UK
| | - Keirah E Bartlett
- Centre for Snakebite Research & Interventions, Liverpool School of Tropical Medicine, Pembroke Place, Liverpool, L3 5QA, UK
| | - Adam P Westhorpe
- Centre for Snakebite Research & Interventions, Liverpool School of Tropical Medicine, Pembroke Place, Liverpool, L3 5QA, UK
| | - Laura-Oana Albulescu
- Centre for Snakebite Research & Interventions, Liverpool School of Tropical Medicine, Pembroke Place, Liverpool, L3 5QA, UK
- Centre for Drugs & Diagnostics, Liverpool School of Tropical Medicine, Pembroke Place, Liverpool, L3 5QA, UK
| | - Jeroen Kool
- Division of BioAnalytical Chemistry, Amsterdam Institute of Molecular and Life Sciences (AIMMS), Vrije Universiteit Amsterdam, De Boelelaan 1085, 1081 HV, Amsterdam, The Netherlands
- Centre for Analytical Sciences Amsterdam (CASA), 1098 XH, Amsterdam, The Netherlands
| | - José María Gutiérrez
- Instituto Clodomiro Picado, Facultad de Microbiología, Universidad de Costa Rica, PO Box 11501-2060, San José, Costa Rica
| | - Nicholas R Casewell
- Centre for Snakebite Research & Interventions, Liverpool School of Tropical Medicine, Pembroke Place, Liverpool, L3 5QA, UK.
- Centre for Drugs & Diagnostics, Liverpool School of Tropical Medicine, Pembroke Place, Liverpool, L3 5QA, UK.
| |
Collapse
|
11
|
Sampat GH, Hiremath K, Dodakallanavar J, Patil VS, Harish DR, Biradar P, Mahadevamurthy RK, Barvaliya M, Roy S. Unraveling snake venom phospholipase A 2: an overview of its structure, pharmacology, and inhibitors. Pharmacol Rep 2023; 75:1454-1473. [PMID: 37926795 DOI: 10.1007/s43440-023-00543-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2023] [Revised: 10/05/2023] [Accepted: 10/06/2023] [Indexed: 11/07/2023]
Abstract
Snake bite is a neglected disease that affects millions of people worldwide. WHO reported approximately 5 million people are bitten by various species of snakes each year, resulting in nearly 1 million deaths and an additional three times cases of permanent disability. Snakes utilize the venom mainly for immobilization and digestion of their prey. Snake venom is a composition of proteins and enzymes which is responsible for its diverse pharmacological action. Snake venom phospholipase A2 (SvPLA2) is an enzyme that is present in every snake species in different quantities and is known to produce remarkable functional diversity and pharmacological action like inflammation, necrosis, myonecrosis, hemorrhage, etc. Arachidonic acid, a precursor to eicosanoids, such as prostaglandins and leukotrienes, is released when SvPLA2 catalyzes the hydrolysis of the sn-2 positions of membrane glycerophospholipids, which is responsible for its actions. Polyvalent antivenom produced from horses or lambs is the standard treatment for snake envenomation, although it has many drawbacks. Traditional medical practitioners treat snake bites using plants and other remedies as a sustainable alternative. More than 500 plant species from more than 100 families reported having venom-neutralizing abilities. Plant-derived secondary metabolites have the ability to reduce the venom's adverse consequences. Numerous studies have documented the ability of plant chemicals to inhibit the enzymes found in snake venom. Research in recent years has shown that various small molecules, such as varespladib and methyl varespladib, effectively inhibit the PLA2 toxin. In the present article, we have overviewed the knowledge of snake venom phospholipase A2, its classification, and the mechanism involved in the pathophysiology of cytotoxicity, myonecrosis, anticoagulation, and inflammation clinical application and inhibitors of SvPLA2, along with the list of studies carried out to evaluate the potency of small molecules like varespladib and secondary metabolites from the traditional medicine for their anti-PLA2 effect.
Collapse
Affiliation(s)
- Ganesh H Sampat
- ICMR-National Institute of Traditional Medicine, Belagavi, Karnataka, 590010, India
- KLE College of Pharmacy, Belagavi, KLE Academy of Higher Education and Research, Belagavi, Karnataka, 590010, India
| | - Kashinath Hiremath
- ICMR-National Institute of Traditional Medicine, Belagavi, Karnataka, 590010, India
- KLE College of Pharmacy, Belagavi, KLE Academy of Higher Education and Research, Belagavi, Karnataka, 590010, India
| | - Jagadeesh Dodakallanavar
- ICMR-National Institute of Traditional Medicine, Belagavi, Karnataka, 590010, India
- KLE College of Pharmacy, Belagavi, KLE Academy of Higher Education and Research, Belagavi, Karnataka, 590010, India
| | - Vishal S Patil
- ICMR-National Institute of Traditional Medicine, Belagavi, Karnataka, 590010, India
- KLE College of Pharmacy, Belagavi, KLE Academy of Higher Education and Research, Belagavi, Karnataka, 590010, India
| | - Darasaguppe R Harish
- ICMR-National Institute of Traditional Medicine, Belagavi, Karnataka, 590010, India.
| | - Prakash Biradar
- KLE College of Pharmacy, Belagavi, KLE Academy of Higher Education and Research, Belagavi, Karnataka, 590010, India.
| | | | - Manish Barvaliya
- ICMR-National Institute of Traditional Medicine, Belagavi, Karnataka, 590010, India
| | - Subarna Roy
- ICMR-National Institute of Traditional Medicine, Belagavi, Karnataka, 590010, India
| |
Collapse
|
12
|
Werner RM, Soffa AN. Considerations for the development of a field-based medical device for the administration of adjunctive therapies for snakebite envenoming. Toxicon X 2023; 20:100169. [PMID: 37661997 PMCID: PMC10474190 DOI: 10.1016/j.toxcx.2023.100169] [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: 05/31/2023] [Revised: 07/27/2023] [Accepted: 08/12/2023] [Indexed: 09/05/2023] Open
Abstract
The timely administration of antivenom is the most effective method currently available to reduce the burden of snakebite envenoming (SBE), a neglected tropical disease that most often affects rural agricultural global populations. There is increasing interest in the development of adjunctive small molecule and biologic therapeutics that target the most problematic venom components to bridge the time-gap between initial SBE and the administration antivenom. Unique combinations of these therapeutics could provide relief from the toxic effects of regional groupings of medically relevant snake species. The application a PRISMA/PICO literature search methodology demonstrated an increasing interest in the rapid administration of therapies to improve patient symptoms and outcomes after SBE. Advice from expert interviews and considerations regarding the potential routes of therapy administration, anatomical bite location, and species-specific venom delivery have provided a framework to identify ideal metrics and potential hurdles for the development of a field-based medical device that could be used immediately after SBE to deliver adjunctive therapies. The use of subcutaneous (SC) or intramuscular (IM) injection were identified as potential routes of administration of both small molecule and biologic therapies. The development of a field-based medical device for the delivery of adjunctive SBE therapies presents unique challenges that will require a collaborative and transdisciplinary approach to be successful.
Collapse
|
13
|
Fu K, Cao L, Tang Y, Zhao J, Xiong K, Hong C, Huang C. The anti-myotoxic effects and mechanisms of Sinonatrix annularis serum and a novel plasma metalloproteinase inhibitor towards Deinagkistrodon acutus envenomation. Toxicol Lett 2023; 388:13-23. [PMID: 37805084 DOI: 10.1016/j.toxlet.2023.10.004] [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: 05/16/2023] [Revised: 09/30/2023] [Accepted: 10/04/2023] [Indexed: 10/09/2023]
Abstract
Non-venomous snakes commonly evolve natural resistance to venom to escape predators. Sinonatrix annularis serum has been shown to inhibit Deinagkistrodon acutus venom-induced hemorrhage and upregulation of serum CK, CK-MB, LDH, AST and ALT levels. Using TMT-labeled proteomics analysis, 168 proteins were found to be altered significantly in the envenomed gastrocnemius muscle and categorized into pathways such as complement and coagulation cascades, leukocyte transendothelial migration, and JAK/STAT signaling. These alterations were mitigated by S. annularis serum. Subsequently, a novel metalloproteinase inhibitor, SaMPI, was isolated from S. annularis serum by two-step chromatography. It showed strong antidotal effects against D. acutus envenomation, including inhibition of subcutaneous bleeding caused by crude venom and DaMP (a metalloproteinase derived from D. acutus) activity in a 1:1 ratio. Histology and immunoblotting analyses demonstrated that SaMPI mitigated myonecrosis, reduced neutrophil infiltration and local inflammatory factor release, and retarded JAK/STAT and MAPK signaling activation. Analysis of the SaMPI gene cloned by 5'-RACE revealed a shared sequence identity of 58-79% with other SVMP inhibitors. These findings demonstrate the protective effects of SaMPI and indicate its potential value as a candidate for viper bite adjuvant therapy.
Collapse
Affiliation(s)
- Kepu Fu
- College of Basic Medical Sciences, Nanchang University, Nanchang 330006, Jiangxi, China
| | - Liyun Cao
- Reproductive Medicine Center, Jiangxi Maternal and Child Health Hospital, Nanchang 330038, Jiangxi, China
| | - Yitao Tang
- College of Basic Medical Sciences, Nanchang University, Nanchang 330006, Jiangxi, China
| | - Jianqi Zhao
- College of Basic Medical Sciences, Nanchang University, Nanchang 330006, Jiangxi, China
| | - Kejia Xiong
- College of Basic Medical Sciences, Nanchang University, Nanchang 330006, Jiangxi, China
| | - Congjiang Hong
- Department of Breast Surgery, Ganxi Cancer Hospital, Pingxiang 337099, Jiangxi, China
| | - Chunhong Huang
- College of Basic Medical Sciences, Nanchang University, Nanchang 330006, Jiangxi, China.
| |
Collapse
|
14
|
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
|
15
|
Murakami M, Sato H, Taketomi Y. Modulation of immunity by the secreted phospholipase A 2 family. Immunol Rev 2023; 317:42-70. [PMID: 37035998 DOI: 10.1111/imr.13205] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2023] [Revised: 03/23/2023] [Accepted: 03/28/2023] [Indexed: 04/11/2023]
Abstract
Among the phospholipase A2 (PLA2 ) superfamily, which typically catalyzes the sn-2 hydrolysis of phospholipids to yield fatty acids and lysophospholipids, the secreted PLA2 (sPLA2 ) family contains 11 isoforms in mammals. Individual sPLA2 s have unique enzymatic specificity toward fatty acids and polar heads of phospholipid substrates and display distinct tissue/cellular distributions, suggesting their distinct physiological functions. Recent studies using knockout and/or transgenic mice for a full set of sPLA2 s have revealed their roles in modulation of immunity and related disorders. Application of mass spectrometric lipidomics to these mice has enabled to identify target substrates and products of individual sPLA2 s in given tissue microenvironments. sPLA2 s hydrolyze not only phospholipids in the plasma membrane of activated, damaged or dying mammalian cells, but also extracellular phospholipids such as those in extracellular vesicles, microbe membranes, lipoproteins, surfactants, and dietary phospholipids, thereby exacerbating or ameliorating various diseases. The actions of sPLA2 s are dependent on, or independent of, the generation of fatty acid- or lysophospholipid-derived lipid mediators according to the pathophysiological contexts. In this review, we make an overview of our current understanding of the roles of individual sPLA2 s in various immune responses and associated diseases.
Collapse
Affiliation(s)
- Makoto Murakami
- Laboratory of Microenvironmental and Metabolic Health Science, Center for Disease Biology and Integrative Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
- AMED-CREST, Japan Agency for Medical Research and Development, Tokyo, Japan
| | - Hiroyasu Sato
- Laboratory of Microenvironmental and Metabolic Health Science, Center for Disease Biology and Integrative Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Yoshitaka Taketomi
- Laboratory of Microenvironmental and Metabolic Health Science, Center for Disease Biology and Integrative Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| |
Collapse
|
16
|
Cardiac Effects of Micrurus corallinus and Micrurus dumerilii carinicauda (Elapidae) Venoms and Neutralization by Brazilian Coralsnake Antivenom and Varespladib. Cardiovasc Toxicol 2023; 23:132-146. [PMID: 36813862 DOI: 10.1007/s12012-023-09786-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/17/2022] [Accepted: 02/09/2023] [Indexed: 02/24/2023]
Abstract
In this work, we examined the action of two South American coralsnake (Micrurus corallinus and Micrurus dumerilii carinicauda) venoms on rat heart function in the absence and presence of treatment with Brazilian coralsnake antivenom (CAV) and varespladib (VPL), a potent phospholipase A2 inhibitor. Anesthetized male Wistar rats were injected with saline (control) or a single dose of venom (1.5 mg/kg, i.m.) and monitored for alterations in echocardiographic parameters, serum CK-MB levels and cardiac histomorphology, the latter using a combination of fractal dimension and histopathological methods. Neither of the venoms caused cardiac functional alterations 2 h after venom injection; however, M. corallinus venom caused tachycardia 2 h after venom injection, with CAV (given i.p. at an antivenom:venom ratio of 1:1.5, v/w), VPL (0.5 mg/kg, i.p.) and CAV + VPL preventing this increase. Both venoms increased the cardiac lesional score and serum CK-MB levels compared to saline-treated rats, but only the combination of CAV + VPL prevented these alterations, although VPL alone was able to attenuate the increase in CK-MB caused by M. corallinus venom. Micrurus corallinus venom increased the heart fractal dimension measurement, but none of the treatments prevented this alteration. In conclusion, M. corallinus and M. d. carinicauda venoms caused no major cardiac functional alterations at the dose tested, although M. corallinus venom caused transient tachycardia. Both venoms caused some cardiac morphological damage, as indicated by histomorphological analyses and the increase in circulating CK-MB levels. These alterations were consistently attenuated by a combination of CAV and VPL.
Collapse
|
17
|
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
|
18
|
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: 21] [Impact Index Per Article: 7.0] [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
|
19
|
Quiroz S, Henao Castañeda IC, Granados J, Patiño AC, Preciado LM, Pereañez JA. Inhibitory Effects of Varespladib, CP471474, and Their Potential Synergistic Activity on Bothrops asper and Crotalus durissus cumanensis Venoms. Molecules 2022; 27:8588. [PMID: 36500682 PMCID: PMC9737558 DOI: 10.3390/molecules27238588] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Revised: 12/02/2022] [Accepted: 12/03/2022] [Indexed: 12/12/2022] Open
Abstract
Snakebite is a neglected tropical disease that causes extensive mortality and morbidity in rural communities. Antivenim sera are the currently approved therapy for snake bites; however, they have some therapeutic limitations that have been extensively documented. Recently, small molecule toxin inhibitors have received significant attention as potential alternatives or co-adjuvant to immunoglobulin-based snakebite therapies. Thus, in this study, we evaluated the inhibitory effects of the phospholipase A2 inhibitor varespladib and the metalloproteinase inhibitor CP471474 and their synergistic effects on the lethal, edema-forming, hemorrhagic, and myotoxic activities of Bothrops asper and Crotalus durissus cumanensis venoms from Colombia. Except for the preincubation assay of the lethal activity with B. asper venom, the mixture showed the best inhibitory activity. Nevertheless, the mix did not display statistically significant differences to varespladib and CP471474 used separately in all assays. In preincubation assays, varespladib showed the best inhibitory activity against the lethal effect induced by B. asper venom. However, in independent injection assays, the mix of the compounds partially inhibited the lethal activity of both venoms (50%). In addition, in the assays to test the inhibition of edema-forming activity, the mixture exhibited the best inhibitory activity, followed by Varespladib, but without statistically significant differences (p > 0.05). The combination also decreased the myotoxic activity of evaluated venoms. In these assays, the mix showed statistical differences regarding CP471474 (p < 0.05). The mixture also abolished the hemorrhagic activity of B. asper venom in preincubation assays, with no statistical differences to CP471474. Finally, the mixture showed inhibition in studies with independent administration in a time-dependent manner. To propose a mode of action of varespladib and CP471474, molecular docking was performed. PLA2s and SVMPs from tested venoms were used as targets. In all cases, our molecular modeling results suggested that inhibitors may occupy the substrate-binding cleft of the enzymes, which was supported by specific interaction with amino acids from the active site, such as His48 for PLA2s and Glu143 for the metalloproteinase. In addition, varespladib and CP471474 also showed interaction with residues from the hydrophobic channel in PLA2s and substrate binding subsites in the SVMP. Our results suggest a synergistic action of the mixed inhibitors and show the potential of varespladib, CP471474, and their mixture to generate new treatments for snakebite envenoming with application in the field or as antivenom co-adjuvants.
Collapse
Affiliation(s)
- Sara Quiroz
- Research Group in Toxinology, Pharmaceutical, and Food Alternatives, Pharmaceutical and Food Sciences Faculty, University of Antioquia, Medellín 50010, Colombia
| | - Isabel C. Henao Castañeda
- Research Group in Marine Natural Products, Pharmaceutical and Food Sciences Faculty, University of Antioquia, Medellín 050010, Colombia
| | - Johan Granados
- Research Group in Pharmaceutical Promotion and Prevention, Universidad de Antioquia, Medellín 050010, Colombia
| | - Arley Camilo Patiño
- Research Group in Toxinology, Pharmaceutical, and Food Alternatives, Pharmaceutical and Food Sciences Faculty, University of Antioquia, Medellín 50010, Colombia
| | - Lina María Preciado
- Research Group in Toxinology, Pharmaceutical, and Food Alternatives, Pharmaceutical and Food Sciences Faculty, University of Antioquia, Medellín 50010, Colombia
| | - Jaime Andrés Pereañez
- Research Group in Toxinology, Pharmaceutical, and Food Alternatives, Pharmaceutical and Food Sciences Faculty, University of Antioquia, Medellín 50010, Colombia
| |
Collapse
|
20
|
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: 28] [Impact Index Per Article: 9.3] [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
|
21
|
Arrahman A, Kazandjian TD, Still KBM, Slagboom J, Somsen GW, Vonk FJ, Casewell NR, Kool J. A Combined Bioassay and Nanofractionation Approach to Investigate the Anticoagulant Toxins of Mamba and Cobra Venoms and Their Inhibition by Varespladib. Toxins (Basel) 2022; 14:736. [PMID: 36355986 PMCID: PMC9695013 DOI: 10.3390/toxins14110736] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2022] [Revised: 10/17/2022] [Accepted: 10/21/2022] [Indexed: 01/26/2023] Open
Abstract
Envenomation by elapid snakes primarily results in neurotoxic symptoms and, consequently, are the primary focus of therapeutic research concerning such venoms. However, mounting evidence suggests these venoms can additionally cause coagulopathic symptoms, as demonstrated by some Asian elapids and African spitting cobras. This study sought to investigate the coagulopathic potential of venoms from medically important elapids of the genera Naja (true cobras), Hemachatus (rinkhals), and Dendroaspis (mambas). Crude venoms were bioassayed for coagulant effects using a plasma coagulation assay before RPLC/MS was used to separate and identify venom toxins in parallel with a nanofractionation module. Subsequently, coagulation bioassays were performed on the nanofractionated toxins, along with in-solution tryptic digestion and proteomics analysis. These experiments were then repeated on both crude venoms and on the nanofractionated venom toxins with the addition of either the phospholipase A2 (PLA2) inhibitor varespladib or the snake venom metalloproteinase (SVMP) inhibitor marimastat. Our results demonstrate that various African elapid venoms have an anticoagulant effect, and that this activity is significantly reduced for cobra venoms by the addition of varespladib, though this inhibitor had no effect against anticoagulation caused by mamba venoms. Marimastat showed limited capacity to reduce anticoagulation in elapids, affecting only N. haje and H. haemachatus venom at higher doses. Proteomic analysis of nanofractionated toxins revealed that the anticoagulant toxins in cobra venoms were both acidic and basic PLA2s, while the causative toxins in mamba venoms remain uncertain. This implies that while PLA2 inhibitors such as varespladib and metalloproteinase inhibitors such as marimastat are viable candidates for novel snakebite treatments, they are not likely to be effective against mamba envenomings.
Collapse
Affiliation(s)
- Arif Arrahman
- Division of Bioanalytical Chemistry, Department of Chemistry and Pharmaceutical Sciences, Faculty of Sciences, Amsterdam Institute of Molecular and Life Sciences (AIMMS), Vrije Universiteit Amsterdam, De Boelelaan 1083, 1081 HV Amsterdam, The Netherlands
- Centre for Analytical Sciences Amsterdam (CASA), 1012 WX Amsterdam, The Netherlands
- Faculty of Pharmacy, Universitas Indonesia, Kampus Baru UI, Depok 16424, Indonesia
| | - Taline D. Kazandjian
- Centre for Snakebite Research and Interventions. Liverpool School of Tropical Medicine, Pembroke Place, Liverpool L3 5QA, UK
| | - Kristina B. M. Still
- Division of Bioanalytical Chemistry, Department of Chemistry and Pharmaceutical Sciences, Faculty of Sciences, Amsterdam Institute of Molecular and Life Sciences (AIMMS), Vrije Universiteit Amsterdam, De Boelelaan 1083, 1081 HV Amsterdam, The Netherlands
- Centre for Analytical Sciences Amsterdam (CASA), 1012 WX Amsterdam, The Netherlands
| | - Julien Slagboom
- Division of Bioanalytical Chemistry, Department of Chemistry and Pharmaceutical Sciences, Faculty of Sciences, Amsterdam Institute of Molecular and Life Sciences (AIMMS), Vrije Universiteit Amsterdam, De Boelelaan 1083, 1081 HV Amsterdam, The Netherlands
- Centre for Analytical Sciences Amsterdam (CASA), 1012 WX Amsterdam, The Netherlands
| | - Govert W. Somsen
- Division of Bioanalytical Chemistry, Department of Chemistry and Pharmaceutical Sciences, Faculty of Sciences, Amsterdam Institute of Molecular and Life Sciences (AIMMS), Vrije Universiteit Amsterdam, De Boelelaan 1083, 1081 HV Amsterdam, The Netherlands
- Centre for Analytical Sciences Amsterdam (CASA), 1012 WX Amsterdam, The Netherlands
| | - Freek J. Vonk
- Division of Bioanalytical Chemistry, Department of Chemistry and Pharmaceutical Sciences, Faculty of Sciences, Amsterdam Institute of Molecular and Life Sciences (AIMMS), Vrije Universiteit Amsterdam, De Boelelaan 1083, 1081 HV Amsterdam, The Netherlands
- Naturalis Biodiversity Centre, Darwinweg 2, 2333 CR Leiden, The Netherlands
| | - Nicholas R. Casewell
- Centre for Snakebite Research and Interventions. Liverpool School of Tropical Medicine, Pembroke Place, Liverpool L3 5QA, UK
| | - Jeroen Kool
- Division of Bioanalytical Chemistry, Department of Chemistry and Pharmaceutical Sciences, Faculty of Sciences, Amsterdam Institute of Molecular and Life Sciences (AIMMS), Vrije Universiteit Amsterdam, De Boelelaan 1083, 1081 HV Amsterdam, The Netherlands
- Centre for Analytical Sciences Amsterdam (CASA), 1012 WX Amsterdam, The Netherlands
| |
Collapse
|
22
|
Wang Y, Zhang J, Zhang D, Xiao H, Xiong S, Huang C. Correction: Wang et al. Exploration of the Inhibitory Potential of Varespladib for Snakebite Envenomation. Molecules 2018, 23, 391. Molecules 2022; 27:molecules27165269. [PMID: 36014594 PMCID: PMC9412874 DOI: 10.3390/molecules27165269] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2021] [Accepted: 08/11/2022] [Indexed: 11/16/2022] Open
Abstract
The authors would like to correct an error in the original publication [...]
Collapse
|
23
|
Sofyantoro F, Yudha DS, Lischer K, Nuringtyas TR, Putri WA, Kusuma WA, Purwestri YA, Swasono RT. Bibliometric Analysis of Literature in Snake Venom-Related Research Worldwide (1933-2022). Animals (Basel) 2022; 12:2058. [PMID: 36009648 PMCID: PMC9405337 DOI: 10.3390/ani12162058] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2022] [Revised: 08/03/2022] [Accepted: 08/09/2022] [Indexed: 11/29/2022] Open
Abstract
Snake envenomation is a severe economic and health concern affecting countries worldwide. Snake venom carries a wide variety of small peptides and proteins with various immunological and pharmacological properties. A few key research areas related to snake venom, including its applications in treating cancer and eradicating antibiotic-resistant bacteria, have been gaining significant attention in recent years. The goal of the current study was to analyze the global profile of literature in snake venom research. This study presents a bibliometric review of snake venom-related research documents indexed in the Scopus database between 1933 and 2022. The overall number of documents published on a global scale was 2999, with an average annual production of 34 documents. Brazil produced the highest number of documents (n = 729), followed by the United States (n = 548), Australia (n = 240), and Costa Rica (n = 235). Since 1963, the number of publications has been steadily increasing globally. At a worldwide level, antivenom, proteomics, and transcriptomics are growing hot issues for research in this field. The current research provides a unique overview of snake venom research at global level from 1933 through 2022, and it may be beneficial in guiding future research.
Collapse
Affiliation(s)
- Fajar Sofyantoro
- Faculty of Biology, Universitas Gadjah Mada, Yogyakarta 55281, Indonesia
| | - Donan Satria Yudha
- Faculty of Biology, Universitas Gadjah Mada, Yogyakarta 55281, Indonesia
| | - Kenny Lischer
- Faculty of Engineering, University of Indonesia, Jakarta 16424, Indonesia
| | - Tri Rini Nuringtyas
- Faculty of Biology, Universitas Gadjah Mada, Yogyakarta 55281, Indonesia
- Research Center for Biotechnology, Universitas Gadjah Mada, Yogyakarta 55281, Indonesia
| | | | - Wisnu Ananta Kusuma
- Department of Computer Science, Faculty of Mathematics and Natural Sciences, IPB University, Bogor 16680, Indonesia
| | - Yekti Asih Purwestri
- Faculty of Biology, Universitas Gadjah Mada, Yogyakarta 55281, Indonesia
- Research Center for Biotechnology, Universitas Gadjah Mada, Yogyakarta 55281, Indonesia
| | - Respati Tri Swasono
- Department of Chemistry, Faculty of Mathematics and Natural Sciences, Universitas Gadjah Mada, Yogyakarta 55281, Indonesia
| |
Collapse
|
24
|
Tiwari N, Aggarwal G, Jain GK, Mittal G. Multi-drug loaded microneedles for emergency treatment of snakebite envenomation. Med Hypotheses 2022. [DOI: 10.1016/j.mehy.2022.110908] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/17/2022]
|
25
|
Menzies SK, Clare RH, Xie C, Westhorpe A, Hall SR, Edge RJ, Alsolaiss J, Crittenden E, Marriott AE, Harrison RA, Kool J, Casewell NR. In vitro and in vivo preclinical venom inhibition assays identify metalloproteinase inhibiting drugs as potential future treatments for snakebite envenoming by Dispholidus typus. Toxicon X 2022; 14:100118. [PMID: 35321116 PMCID: PMC8935517 DOI: 10.1016/j.toxcx.2022.100118] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2021] [Revised: 02/18/2022] [Accepted: 03/07/2022] [Indexed: 11/23/2022] Open
Abstract
Snakebite envenoming affects more than 250,000 people annually in sub-Saharan Africa. Envenoming by Dispholidus typus (boomslang) results in venom-induced consumption coagulopathy (VICC), whereby highly abundant prothrombin-activating snake venom metalloproteinases (SVMPs) consume clotting factors and deplete fibrinogen. The only available treatment for D. typus envenoming is the monovalent SAIMR Boomslang antivenom. Treatment options are urgently required because this antivenom is often difficult to source and, at US$6000/vial, typically unaffordable for most snakebite patients. We therefore investigated the in vitro and in vivo preclinical efficacy of four SVMP inhibitors to neutralise the effects of D. typus venom; the matrix metalloproteinase inhibitors marimastat and prinomastat, and the metal chelators dimercaprol and DMPS. The venom of D. typus exhibited an SVMP-driven procoagulant phenotype in vitro. Marimastat and prinomastat demonstrated equipotent inhibition of the SVMP-mediated procoagulant activity of the venom in vitro, whereas dimercaprol and DMPS showed considerably lower potency. However, when tested in preclinical murine models of envenoming using mixed sex CD1 mice, DMPS and marimastat demonstrated partial protection against venom lethality, demonstrated by prolonged survival times of experimental animals, whereas dimercaprol and prinomastat failed to confer any protection at the doses tested. The preclinical results presented here demonstrate that DMPS and marimastat show potential as novel small molecule-based therapeutics for D. typus snakebite envenoming. These two drugs have been previously shown to be effective against Echis ocellatus VICC in preclinical models, and thus we conclude that marimastat and DMPS should be further explored as potentially valuable early intervention therapeutics to broadly treat VICC following snakebite envenoming in sub-Saharan Africa.
Collapse
Affiliation(s)
- Stefanie K. Menzies
- Centre for Snakebite Research and Interventions, Department of Tropical Disease Biology, Liverpool School of Tropical Medicine, Pembroke Place, Liverpool, L3 5QA, England, UK
- Centre for Drugs and Diagnostics, Department of Tropical Disease Biology, Liverpool School of Tropical Medicine, Pembroke Place, Liverpool, L3 5QA, England, UK
| | - Rachel H. Clare
- Centre for Snakebite Research and Interventions, Department of Tropical Disease Biology, Liverpool School of Tropical Medicine, Pembroke Place, Liverpool, L3 5QA, England, UK
- Centre for Drugs and Diagnostics, Department of Tropical Disease Biology, Liverpool School of Tropical Medicine, Pembroke Place, Liverpool, L3 5QA, England, UK
| | - Chunfang Xie
- Amsterdam Institute of Molecular and Life Sciences, Division of BioAnalytical Chemistry, Department of Chemistry and Pharmaceutical Sciences, Faculty of Science, Vrije Universiteit Amsterdam, De Boelelaan 1085, 1081HV, Amsterdam, the Netherlands
| | - Adam Westhorpe
- Centre for Snakebite Research and Interventions, Department of Tropical Disease Biology, Liverpool School of Tropical Medicine, Pembroke Place, Liverpool, L3 5QA, England, UK
- Centre for Drugs and Diagnostics, Department of Tropical Disease Biology, Liverpool School of Tropical Medicine, Pembroke Place, Liverpool, L3 5QA, England, UK
| | - Steven R. Hall
- Centre for Snakebite Research and Interventions, Department of Tropical Disease Biology, Liverpool School of Tropical Medicine, Pembroke Place, Liverpool, L3 5QA, England, UK
- Centre for Drugs and Diagnostics, Department of Tropical Disease Biology, Liverpool School of Tropical Medicine, Pembroke Place, Liverpool, L3 5QA, England, UK
| | - Rebecca J. Edge
- Centre for Snakebite Research and Interventions, Department of Tropical Disease Biology, Liverpool School of Tropical Medicine, Pembroke Place, Liverpool, L3 5QA, England, UK
- Centre for Drugs and Diagnostics, Department of Tropical Disease Biology, Liverpool School of Tropical Medicine, Pembroke Place, Liverpool, L3 5QA, England, UK
| | - Jaffer Alsolaiss
- Centre for Snakebite Research and Interventions, Department of Tropical Disease Biology, Liverpool School of Tropical Medicine, Pembroke Place, Liverpool, L3 5QA, England, UK
- Centre for Drugs and Diagnostics, Department of Tropical Disease Biology, Liverpool School of Tropical Medicine, Pembroke Place, Liverpool, L3 5QA, England, UK
| | - Edouard Crittenden
- Centre for Snakebite Research and Interventions, Department of Tropical Disease Biology, Liverpool School of Tropical Medicine, Pembroke Place, Liverpool, L3 5QA, England, UK
- Centre for Drugs and Diagnostics, Department of Tropical Disease Biology, Liverpool School of Tropical Medicine, Pembroke Place, Liverpool, L3 5QA, England, UK
| | - Amy E. Marriott
- Centre for Snakebite Research and Interventions, Department of Tropical Disease Biology, Liverpool School of Tropical Medicine, Pembroke Place, Liverpool, L3 5QA, England, UK
- Centre for Drugs and Diagnostics, Department of Tropical Disease Biology, Liverpool School of Tropical Medicine, Pembroke Place, Liverpool, L3 5QA, England, UK
| | - Robert A. Harrison
- Centre for Snakebite Research and Interventions, Department of Tropical Disease Biology, Liverpool School of Tropical Medicine, Pembroke Place, Liverpool, L3 5QA, England, UK
- Centre for Drugs and Diagnostics, Department of Tropical Disease Biology, Liverpool School of Tropical Medicine, Pembroke Place, Liverpool, L3 5QA, England, UK
| | - Jeroen Kool
- Amsterdam Institute of Molecular and Life Sciences, Division of BioAnalytical Chemistry, Department of Chemistry and Pharmaceutical Sciences, Faculty of Science, Vrije Universiteit Amsterdam, De Boelelaan 1085, 1081HV, Amsterdam, the Netherlands
| | - Nicholas R. Casewell
- Centre for Snakebite Research and Interventions, Department of Tropical Disease Biology, Liverpool School of Tropical Medicine, Pembroke Place, Liverpool, L3 5QA, England, UK
- Centre for Drugs and Diagnostics, Department of Tropical Disease Biology, Liverpool School of Tropical Medicine, Pembroke Place, Liverpool, L3 5QA, England, UK
| |
Collapse
|
26
|
de Souza J, Oliveira IC, Yoshida EH, Cantuaria NM, Cogo JC, Torres-Bonilla KA, Hyslop S, Silva Junior NJ, Floriano RS, Gutiérrez JM, Oshima-Franco Y. Effect of the phospholipase A2 inhibitor Varespladib, and its synergism with crotalic antivenom, on the neuromuscular blockade induced by Crotalus durissus terrificus venom (with and without crotamine) in mouse neuromuscular preparations. Toxicon 2022; 214:54-61. [DOI: 10.1016/j.toxicon.2022.05.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2022] [Revised: 05/06/2022] [Accepted: 05/10/2022] [Indexed: 10/18/2022]
|
27
|
Silva-Carvalho R, Gaspar MZ, Quadros LHB, Lobo LGG, Giuffrida R, Santarém CL, Silva EO, Gerez JR, Silva NJ, Hyslop S, Lomonte B, Floriano RS. Partial efficacy of a Brazilian coralsnake antivenom and varespladib in neutralizing distinct toxic effects induced by sublethal Micrurus dumerilii carinicauda envenoming in rats. Toxicon 2022; 213:99-104. [PMID: 35489427 DOI: 10.1016/j.toxicon.2022.04.014] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2022] [Revised: 03/31/2022] [Accepted: 04/20/2022] [Indexed: 11/26/2022]
Abstract
In this work, we report the efficacy of a combination of Brazilian therapeutic coralsnake antivenom (CAV) and varespladib (phospholipase A2 inhibitor - VPL) in partially neutralizing selected toxic effects of Micrurus dumerilii carinicauda coralsnake venom in rats. Venom caused local myonecrosis and systemic neurotoxicity, nephrotoxicity, and hepatotoxicity within 2 h of injection. CAV and VPL administered separately failed to prevent most of these alterations. However, a combination of CAV plus VPL offered variable protection against venom-induced coagulation disturbances, leukocytosis, and renal and hepatic morphological alterations.
Collapse
Affiliation(s)
- Rosimeire Silva-Carvalho
- Laboratory of Toxinology and Cardiovascular Research, Graduate Program in Health Sciences, University of Western São Paulo, Rodovia Raposo Tavares Km 572, B2-205, 19067-175, Presidente Prudente, SP, Brazil
| | - Matheus Z Gaspar
- Laboratory of Toxinology and Cardiovascular Research, Graduate Program in Health Sciences, University of Western São Paulo, Rodovia Raposo Tavares Km 572, B2-205, 19067-175, Presidente Prudente, SP, Brazil
| | - Luiz H B Quadros
- Laboratory of Toxinology and Cardiovascular Research, Graduate Program in Health Sciences, University of Western São Paulo, Rodovia Raposo Tavares Km 572, B2-205, 19067-175, Presidente Prudente, SP, Brazil
| | - Luís G G Lobo
- Laboratory of Toxinology and Cardiovascular Research, Graduate Program in Health Sciences, University of Western São Paulo, Rodovia Raposo Tavares Km 572, B2-205, 19067-175, Presidente Prudente, SP, Brazil
| | - Rogério Giuffrida
- Graduate Program in Animal Sciences, University of Western São Paulo, Rodovia Raposo Tavares Km 572, 19067-175, Presidente Prudente, SP, Brazil
| | - Cecília L Santarém
- Graduate Program in Animal Sciences, University of Western São Paulo, Rodovia Raposo Tavares Km 572, 19067-175, Presidente Prudente, SP, Brazil
| | - Elisangela O Silva
- Graduate Program in Animal Sciences, University of Western São Paulo, Rodovia Raposo Tavares Km 572, 19067-175, Presidente Prudente, SP, Brazil
| | - Juliana R Gerez
- Department of Histology, State University of Londrina (UEL), Rodovia Celso Garcia Cid Km 380, 86057-970, Londrina, PR, Brazil
| | - Nelson J Silva
- Graduate Program in Environmental Sciences and Health, School of Medical, Pharmaceutical and Biomedical Sciences, Pontifical Catholic University of Goiás (PUC-Goiás), 74605-140, Goiânia, GO, Brazil
| | - Stephen Hyslop
- Section of Pharmacology, Department of Translational Medicine, Faculty of Medical Sciences, State University of Campinas (UNICAMP), Rua Tessália Vieira de Camargo, 126, 13083-887, Campinas, SP, Brazil
| | - Bruno Lomonte
- Instituto Clodomiro Picado, Facultad de Microbiología, Universidad de Costa Rica, 11501, San José, Costa Rica.
| | - Rafael S Floriano
- Laboratory of Toxinology and Cardiovascular Research, Graduate Program in Health Sciences, University of Western São Paulo, Rodovia Raposo Tavares Km 572, B2-205, 19067-175, Presidente Prudente, SP, Brazil.
| |
Collapse
|
28
|
A comparative study of endogenous phospholipase A 2 inhibitors in the serum of Brazilian pit vipers. Toxicon 2022; 213:87-91. [PMID: 35487313 DOI: 10.1016/j.toxicon.2022.04.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2021] [Revised: 04/13/2022] [Accepted: 04/14/2022] [Indexed: 11/20/2022]
Abstract
This work compared the presence of phospholipase A2 inhibitors (PLIs) in the serum of 19 snake species maintained at Instituto Butantan to better understand the mechanisms of venom resistance in snakes and improve the treatment of snakebite. PLI was isolated from blood of 19 snake species by one-step chromatography and identified in all samples, besides its identity was confirmed through the interaction with both phospholipase A2 and anti-γPLI. These findings highlight the diversity of snake serum PLIs and emphasize the importance of structure-function studies.
Collapse
|
29
|
Youngman NJ, Lewin MR, Carter R, Naude A, Fry BG. Efficacy and Limitations of Chemically Diverse Small-Molecule Enzyme-Inhibitors against the Synergistic Coagulotoxic Activities of Bitis Viper Venoms. Molecules 2022; 27:1733. [PMID: 35268832 PMCID: PMC8911647 DOI: 10.3390/molecules27051733] [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: 01/10/2022] [Revised: 02/22/2022] [Accepted: 03/02/2022] [Indexed: 11/24/2022] Open
Abstract
Snakebite remains a significant public health burden globally, disproportionately affecting low-income and impoverished regions of the world. Recently, researchers have begun to focus on the use of small-molecule inhibitors as potential candidates for the neutralisation of key snake venom toxins and as potential field therapies. Bitis vipers represent some of the most medically important as well as frequently encountered snake species in Africa, with a number of species possessing anticoagulant phospholipase A2 (PLA2) toxins that prevent the prothrombinase complex from inducing clot formation. Additionally, species within the genus are known to exert pseudo-procoagulant activity, whereby kallikrein enzymatic toxins cleave fibrinogen to form a weak fibrin clot that rapidly degrades, thereby depleting fibrinogen levels and contributing to the net anticoagulant state. Utilising well-validated coagulation assays measuring time until clot formation, this study addresses the in vitro efficacy of three small molecule enzyme inhibitors (marimastat, prinomastat and varespladib) in neutralising these aforementioned activities. The PLA2 inhibitor varespladib showed the greatest efficacy for the neutralisation of PLA2-driven anticoagulant venom activity, with the metalloproteinase inhibitors prinomastat and marimastat both showing low and highly variable degrees of cross-neutralisation with PLA2 anticoagulant toxicity. However, none of the inhibitors showed efficacy in neutralising the pseudo-procoagulant venom activity exerted by the venom of B. caudalis. Our results highlight the complex nature of snake venoms, for which single-compound treatments will not be universally effective, but combinations might prove highly effective. Despite the limitations of these inhibitors with regards to in vitro kallikrein enzyme pseudo-procoagulant venom activity, our results further support the growing body of literature indicating the potential use of small molecule inhibitors to enhance first-aid treatment of snakebite envenoming, particularly in cases where hospital and thus antivenom treatment is either unavailable or far away.
Collapse
Affiliation(s)
- Nicholas J. Youngman
- Venom Evolution Lab, School of Biological Science, 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;
| | | | - Arno Naude
- Snakebite Assist, Pretoria ZA-0001, South Africa;
| | - Bryan G. Fry
- Venom Evolution Lab, School of Biological Science, University of Queensland, St. Lucia, QLD 4072, Australia
| |
Collapse
|
30
|
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
|
31
|
Moreira V, Leiguez E, Janovits PM, Maia-Marques R, Fernandes CM, Teixeira C. Inflammatory Effects of Bothrops Phospholipases A 2: Mechanisms Involved in Biosynthesis of Lipid Mediators and Lipid Accumulation. Toxins (Basel) 2021; 13:toxins13120868. [PMID: 34941706 PMCID: PMC8709003 DOI: 10.3390/toxins13120868] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2021] [Revised: 11/19/2021] [Accepted: 11/30/2021] [Indexed: 02/07/2023] Open
Abstract
Phospholipases A2s (PLA2s) constitute one of the major protein groups present in the venoms of viperid and crotalid snakes. Snake venom PLA2s (svPLA2s) exhibit a remarkable functional diversity, as they have been described to induce a myriad of toxic effects. Local inflammation is an important characteristic of snakebite envenomation inflicted by viperid and crotalid species and diverse svPLA2s have been studied for their proinflammatory properties. Moreover, based on their molecular, structural, and functional properties, the viperid svPLA2s are classified into the group IIA secreted PLA2s, which encompasses mammalian inflammatory sPLA2s. Thus, research on svPLA2s has attained paramount importance for better understanding the role of this class of enzymes in snake envenomation and the participation of GIIA sPLA2s in pathophysiological conditions and for the development of new therapeutic agents. In this review, we highlight studies that have identified the inflammatory activities of svPLA2s, in particular, those from Bothrops genus snakes, which are major medically important snakes in Latin America, and we describe recent advances in our collective understanding of the mechanisms underlying their inflammatory effects. We also discuss studies that dissect the action of these venom enzymes in inflammatory cells focusing on molecular mechanisms and signaling pathways involved in the biosynthesis of lipid mediators and lipid accumulation in immunocompetent cells.
Collapse
Affiliation(s)
- Vanessa Moreira
- Departamento de Farmacologia, Escola Paulista de Medicina, Universidade Federal de Sao Paulo, Sao Paulo 04044-020, Brazil;
| | - Elbio Leiguez
- Laboratório de Farmacologia, Instituto Butantan, Sao Paulo 05503-900, Brazil; (E.L.); (P.M.J.); (R.M.-M.); (C.M.F.)
| | - Priscila Motta Janovits
- Laboratório de Farmacologia, Instituto Butantan, Sao Paulo 05503-900, Brazil; (E.L.); (P.M.J.); (R.M.-M.); (C.M.F.)
| | - Rodrigo Maia-Marques
- Laboratório de Farmacologia, Instituto Butantan, Sao Paulo 05503-900, Brazil; (E.L.); (P.M.J.); (R.M.-M.); (C.M.F.)
| | - Cristina Maria Fernandes
- Laboratório de Farmacologia, Instituto Butantan, Sao Paulo 05503-900, Brazil; (E.L.); (P.M.J.); (R.M.-M.); (C.M.F.)
| | - Catarina Teixeira
- Laboratório de Farmacologia, Instituto Butantan, Sao Paulo 05503-900, Brazil; (E.L.); (P.M.J.); (R.M.-M.); (C.M.F.)
- Correspondence:
| |
Collapse
|
32
|
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
|
33
|
Maciel FV, Ramos Pinto ÊK, Valério Souza NM, Gonçalves de Abreu TA, Ortolani PL, Fortes-Dias CL, Garrido Cavalcante WL. Varespladib (LY315920) prevents neuromuscular blockage and myotoxicity induced by crotoxin on mouse neuromuscular preparations. Toxicon 2021; 202:40-45. [PMID: 34562493 DOI: 10.1016/j.toxicon.2021.09.009] [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: 07/27/2021] [Revised: 09/15/2021] [Accepted: 09/20/2021] [Indexed: 12/26/2022]
Abstract
Varespladib (LY315920) is a synthetic phospholipase A2 (PLA2) inhibitor that has been demonstrating antiophidic potential against snake venoms that present PLA2 neurotoxins. In this study, we evaluate the capacity of Varespladib to inhibit the neuromuscular effects of crotoxin (CTX), the main toxic component of Crotalus durissus terrificus snake venom, and its PLA2 subunit (CB). We performed a myographic study to compare the neuromuscular effects of CTX or CB and the mixture of these substances plus Varespladib in mice phrenic nerve-diaphragm muscle preparations. CTX (5 μg/mL), CB (20 μg/mL), or toxin-inhibitor mixtures pre-incubated with different concentration ratios of Varespladib (1:0.25; 1:0.5; 1:1; w/w) were added to the preparations and maintained throughout the experimentation period. Myotoxicity was assessed by light microscopic analysis of diaphragm muscle after myographic study. CTX and CB blocked the nerve-evoked twitches, and only CTX induced histological alterations in diaphragm muscle. Pre-incubation with Varespladib abolished the muscle-paralyzing activity of CTX and CB, and also the muscle-damaging activity of CTX. These findings emphasize the clinical potential of Varespladib in mitigating the toxic effects of C. d. terrificus snakebites and as a research tool to advance the knowledge of the mechanism of action of snake toxins.
Collapse
Affiliation(s)
- Fernanda Valadares Maciel
- Department of Pharmacology, Institute of Biological Science, Federal University of Minas Gerais (UFMG), Brazil
| | - Êmylle Karoline Ramos Pinto
- Department of Pharmacology, Institute of Biological Science, Federal University of Minas Gerais (UFMG), Brazil
| | | | | | | | | | | |
Collapse
|
34
|
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
|
35
|
Xie C, Bittenbinder MA, Slagboom J, Arrahman A, Bruijns S, Somsen GW, Vonk FJ, Casewell NR, García-Vallejo JJ, Kool J. Erythrocyte haemotoxicity profiling of snake venom toxins after nanofractionation. J Chromatogr B Analyt Technol Biomed Life Sci 2021; 1176:122586. [PMID: 33839052 PMCID: PMC7613003 DOI: 10.1016/j.jchromb.2021.122586] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Revised: 02/01/2021] [Accepted: 02/03/2021] [Indexed: 10/22/2022]
Abstract
Snakebite is classified as a priority Neglected Tropical Disease by the World Health Organization. Understanding the pathology of individual snake venom toxins is of great importance when developing more effective snakebite therapies. Snake venoms may induce a range of pathologies, including haemolytic activity. Although snake venom-induced erythrocyte lysis is not the primary cause of mortality, haemolytic activity can greatly debilitate victims and contributes to systemic haemotoxicity. Current assays designed for studying haemolytic activity are not suitable for rapid screening of large numbers of toxic compounds. Consequently, in this study, a high-throughput haemolytic assay was developed that allows profiling of erythrocyte lysis, and was validated using venom from a number of medically important snake species (Calloselasma rhodostoma, Daboia russelii, Naja mossambica, Naja nigricollis and Naja pallida). The assay was developed in a format enabling direct integration into nanofractionation analytics, which involves liquid chromatographic separation of venom followed by high-resolution fractionation and subsequent bioassaying (and optional proteomics analysis), and parallel mass spectrometric detection. Analysis of the five snake venoms via this nanofractionation approach involving haemolytic assaying provided venom-cytotoxicity profiles and enabled identification of the toxins responsible for haemolytic activity. Our results show that the elapid snake venoms (Naja spp.) contained both direct and indirect lytic toxins, while the viperid venoms (C. rhodostoma and D. russelii) only showed indirect lytic activities, which required the addition of phospholipids to exert cytotoxicity on erythrocytes. The haemolytic venom toxins identified were mainly phospholipase A2s and cytotoxic three finger toxins. Finally, the applicability of this new analytical method was demonstrated using a conventional snakebite antivenom treatment and a small-molecule drug candidate to assess neutralisation of venom cytotoxins.
Collapse
Affiliation(s)
- Chunfang Xie
- Amsterdam Institute of Molecular and Life Sciences, Division of BioAnalytical Chemistry, Department of Chemistry and Pharmaceutical Sciences, Faculty of Science, Vrije Universiteit Amsterdam, De Boelelaan 1085, 1081HV Amsterdam, the Netherlands; Centre for Analytical Sciences Amsterdam (CASA), 1098 XH Amsterdam, the Netherlands
| | - Matyas A Bittenbinder
- Amsterdam Institute of Molecular and Life Sciences, Division of BioAnalytical Chemistry, Department of Chemistry and Pharmaceutical Sciences, Faculty of Science, Vrije Universiteit Amsterdam, De Boelelaan 1085, 1081HV Amsterdam, the Netherlands; Centre for Analytical Sciences Amsterdam (CASA), 1098 XH Amsterdam, the Netherlands; Naturalis Biodiversity Center, Darwinweg 2, 2333 CR Leiden, the Netherlands
| | - Julien Slagboom
- Amsterdam Institute of Molecular and Life Sciences, Division of BioAnalytical Chemistry, Department of Chemistry and Pharmaceutical Sciences, Faculty of Science, Vrije Universiteit Amsterdam, De Boelelaan 1085, 1081HV Amsterdam, the Netherlands; Centre for Analytical Sciences Amsterdam (CASA), 1098 XH Amsterdam, the Netherlands
| | - Arif Arrahman
- Amsterdam Institute of Molecular and Life Sciences, Division of BioAnalytical Chemistry, Department of Chemistry and Pharmaceutical Sciences, Faculty of Science, Vrije Universiteit Amsterdam, De Boelelaan 1085, 1081HV Amsterdam, the Netherlands; Centre for Analytical Sciences Amsterdam (CASA), 1098 XH Amsterdam, the Netherlands
| | - Sven Bruijns
- Department of Molecular Cell Biology and Immunology, Amsterdam Infection and Immunity Institute, Cancer Center Amsterdam, Amsterdam UMC, Vrije Universiteit Amsterdam, 1081 HZ Amsterdam, the Netherlands
| | - Govert W Somsen
- Amsterdam Institute of Molecular and Life Sciences, Division of BioAnalytical Chemistry, Department of Chemistry and Pharmaceutical Sciences, Faculty of Science, Vrije Universiteit Amsterdam, De Boelelaan 1085, 1081HV Amsterdam, the Netherlands; Centre for Analytical Sciences Amsterdam (CASA), 1098 XH Amsterdam, the Netherlands
| | - Freek J Vonk
- Amsterdam Institute of Molecular and Life Sciences, Division of BioAnalytical Chemistry, Department of Chemistry and Pharmaceutical Sciences, Faculty of Science, Vrije Universiteit Amsterdam, De Boelelaan 1085, 1081HV Amsterdam, the Netherlands; Centre for Analytical Sciences Amsterdam (CASA), 1098 XH Amsterdam, the Netherlands; Naturalis Biodiversity Center, Darwinweg 2, 2333 CR Leiden, the Netherlands
| | - Nicholas R Casewell
- Centre for Snakebite Research and Interventions, Liverpool School of Tropical Medicine, Pembroke Place, Liverpool L3 5QA, UK; Centre for Drugs and Diagnostics, Liverpool School of Tropical Medicine, Pembroke Place, Liverpool L3 5QA, UK
| | - Juan J García-Vallejo
- Department of Molecular Cell Biology and Immunology, Amsterdam Infection and Immunity Institute, Cancer Center Amsterdam, Amsterdam UMC, Vrije Universiteit Amsterdam, 1081 HZ Amsterdam, the Netherlands
| | - Jeroen Kool
- Amsterdam Institute of Molecular and Life Sciences, Division of BioAnalytical Chemistry, Department of Chemistry and Pharmaceutical Sciences, Faculty of Science, Vrije Universiteit Amsterdam, De Boelelaan 1085, 1081HV Amsterdam, the Netherlands; Centre for Analytical Sciences Amsterdam (CASA), 1098 XH Amsterdam, the Netherlands.
| |
Collapse
|
36
|
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]
|
37
|
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: 8] [Impact Index Per Article: 2.0] [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
|
38
|
Albulescu LO, Xie C, Ainsworth S, Alsolaiss J, Crittenden E, Dawson CA, Softley R, Bartlett KE, Harrison RA, Kool J, Casewell NR. A therapeutic combination of two small molecule toxin inhibitors provides broad preclinical efficacy against viper snakebite. Nat Commun 2020; 11:6094. [PMID: 33323937 PMCID: PMC7738508 DOI: 10.1038/s41467-020-19981-6] [Citation(s) in RCA: 82] [Impact Index Per Article: 16.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2020] [Accepted: 11/11/2020] [Indexed: 01/21/2023] Open
Abstract
Snakebite is a medical emergency causing high mortality and morbidity in rural tropical communities that typically experience delayed access to unaffordable therapeutics. Viperid snakes are responsible for the majority of envenomings, but extensive interspecific variation in venom composition dictates that different antivenom treatments are used in different parts of the world, resulting in clinical and financial snakebite management challenges. Here, we show that a number of repurposed Phase 2-approved small molecules are capable of broadly neutralizing distinct viper venom bioactivities in vitro by inhibiting different enzymatic toxin families. Furthermore, using murine in vivo models of envenoming, we demonstrate that a single dose of a rationally-selected dual inhibitor combination consisting of marimastat and varespladib prevents murine lethality caused by venom from the most medically-important vipers of Africa, South Asia and Central America. Our findings support the translation of combinations of repurposed small molecule-based toxin inhibitors as broad-spectrum therapeutics for snakebite.
Collapse
Affiliation(s)
- Laura-Oana Albulescu
- Centre for Snakebite Research and Interventions, Liverpool School of Tropical Medicine, Pembroke Place, L3 5QA, Liverpool, UK
| | - Chunfang Xie
- Division of BioAnalytical Chemistry, Amsterdam Institute of Molecular and Life Sciences, Vrije Universiteit Amsterdam, De Boelelaan 1085, 1081 HV, Amsterdam, The Netherlands
- Centre for Analytical Sciences Amsterdam (CASA), 1098 XH, Amsterdam, The Netherlands
| | - Stuart Ainsworth
- Centre for Snakebite Research and Interventions, Liverpool School of Tropical Medicine, Pembroke Place, L3 5QA, Liverpool, UK
| | - Jaffer Alsolaiss
- Centre for Snakebite Research and Interventions, Liverpool School of Tropical Medicine, Pembroke Place, L3 5QA, Liverpool, UK
| | - Edouard Crittenden
- Centre for Snakebite Research and Interventions, Liverpool School of Tropical Medicine, Pembroke Place, L3 5QA, Liverpool, UK
| | - Charlotte A Dawson
- Centre for Snakebite Research and Interventions, Liverpool School of Tropical Medicine, Pembroke Place, L3 5QA, Liverpool, UK
| | - Rowan Softley
- Centre for Snakebite Research and Interventions, Liverpool School of Tropical Medicine, Pembroke Place, L3 5QA, Liverpool, UK
| | - Keirah E Bartlett
- Centre for Snakebite Research and Interventions, Liverpool School of Tropical Medicine, Pembroke Place, L3 5QA, Liverpool, UK
| | - Robert A Harrison
- Centre for Snakebite Research and Interventions, Liverpool School of Tropical Medicine, Pembroke Place, L3 5QA, Liverpool, UK
- Centre for Drugs and Diagnostics, Liverpool School of Tropical Medicine, Pembroke Place, L3 5QA, Liverpool, UK
| | - Jeroen Kool
- Division of BioAnalytical Chemistry, Amsterdam Institute of Molecular and Life Sciences, Vrije Universiteit Amsterdam, De Boelelaan 1085, 1081 HV, Amsterdam, The Netherlands
- Centre for Analytical Sciences Amsterdam (CASA), 1098 XH, Amsterdam, The Netherlands
| | - Nicholas R Casewell
- Centre for Snakebite Research and Interventions, Liverpool School of Tropical Medicine, Pembroke Place, L3 5QA, Liverpool, UK.
- Centre for Drugs and Diagnostics, Liverpool School of Tropical Medicine, Pembroke Place, L3 5QA, Liverpool, UK.
| |
Collapse
|
39
|
Lin B, Zhang JR, Lu HJ, Zhao L, Chen J, Zhang HF, Wei XS, Zhang LY, Wu XB, Lee WH. Immunoreactivity and neutralization study of Chinese Bungarus multicinctus antivenin and lab-prepared anti-bungarotoxin antisera towards purified bungarotoxins and snake venoms. PLoS Negl Trop Dis 2020; 14:e0008873. [PMID: 33253321 PMCID: PMC7728252 DOI: 10.1371/journal.pntd.0008873] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2020] [Revised: 12/10/2020] [Accepted: 10/12/2020] [Indexed: 12/25/2022] Open
Abstract
Bungarus multicinctus is the most venomous snake distributed in China and neighboring countries of Myanmar, Laos, north Vietnam and Thailand. The high mortality rate of B. multicinctus envenomation is attributed to the lethal components of α-, β-, γ- and κ- bungarotoxins contained in the venom. Although anti-B. multicinctus sera were produced in Shanghai, Taiwan and Vietnam, the most widely clinic used product was term as B. multicinctus antivenin and manufactured by Shanghai Serum Bio-technology Co. Ltd. In the present investigation, high purity α-, β- and γ-bungarotoxins were separately isolated from B. multicinctus crude venom. Rabbit anti- α-, β- and γ-bungarotoxin antisera were prepared by common methods, respectively. LD50 values of α-, β- and γ-bungarotoxins were systematically determined via three administration pathways (intraperitoneal, intramuscular and intravenous injections) in Kunming mice. LD50 values of β-bungarotoxin were closely related with injection routines but those of both α- and γ-bungarotoxins were not dependent on the injection routines. Commercial B. multicinctus antivenin showed strong immunoreaction with high molecular weight fractions of the B. multicinctus but weakly recognized low molecular weight fractions like α- and γ-bungarotoxins. Although B. multicinctus antivenin showed immunoreaction with high molecular weight fractions of Bungarus fasciatus, Naja atra, Ophiophagus hannah venoms but the antivenin only demonstrated animal protection efficacy against O. hannah venom. These results indicated that the high molecular weight fractions of the O. hannah played an important role in venom lethality but those of B. fasciatus and N. atra did not have such a role.
Collapse
Affiliation(s)
- Bo Lin
- College of Life Sciences, Anhui Normal University, Wuhu, Anhui, China
- Key Laboratory of Animal Models and Human Disease Mechanisms of The Chinese Academy of Sciences/Key Laboratory of Bioactive Peptides of Yunnan Province, Kunming Institute of Zoology, the Chinese Academy of Sciences, Kunming, Yunnan, China
| | - Jia-Rui Zhang
- Nanshan School, Guangzhou Medical University, Guangzhou, Guandong, China
| | - Hui-Juan Lu
- Key Laboratory of Animal Models and Human Disease Mechanisms of The Chinese Academy of Sciences/Key Laboratory of Bioactive Peptides of Yunnan Province, Kunming Institute of Zoology, the Chinese Academy of Sciences, Kunming, Yunnan, China
| | - Lin Zhao
- College of Life Sciences, Anhui Normal University, Wuhu, Anhui, China
- Key Laboratory of Animal Models and Human Disease Mechanisms of The Chinese Academy of Sciences/Key Laboratory of Bioactive Peptides of Yunnan Province, Kunming Institute of Zoology, the Chinese Academy of Sciences, Kunming, Yunnan, China
| | - Jing Chen
- Key Laboratory of Animal Models and Human Disease Mechanisms of The Chinese Academy of Sciences/Key Laboratory of Bioactive Peptides of Yunnan Province, Kunming Institute of Zoology, the Chinese Academy of Sciences, Kunming, Yunnan, China
- School of Life and Pharmaceutical Sciences, Hainan University, Haikou, Hainan, China
| | - Hong-Fei Zhang
- Key Laboratory of Animal Models and Human Disease Mechanisms of The Chinese Academy of Sciences/Key Laboratory of Bioactive Peptides of Yunnan Province, Kunming Institute of Zoology, the Chinese Academy of Sciences, Kunming, Yunnan, China
| | - Xue-Song Wei
- College of Life Sciences, Anhui Normal University, Wuhu, Anhui, China
- Key Laboratory of Animal Models and Human Disease Mechanisms of The Chinese Academy of Sciences/Key Laboratory of Bioactive Peptides of Yunnan Province, Kunming Institute of Zoology, the Chinese Academy of Sciences, Kunming, Yunnan, China
| | - Liang-Yu Zhang
- Key Laboratory of Animal Models and Human Disease Mechanisms of The Chinese Academy of Sciences/Key Laboratory of Bioactive Peptides of Yunnan Province, Kunming Institute of Zoology, the Chinese Academy of Sciences, Kunming, Yunnan, China
| | - Xiao-Bing Wu
- College of Life Sciences, Anhui Normal University, Wuhu, Anhui, China
| | - Wen-Hui Lee
- Key Laboratory of Animal Models and Human Disease Mechanisms of The Chinese Academy of Sciences/Key Laboratory of Bioactive Peptides of Yunnan Province, Kunming Institute of Zoology, the Chinese Academy of Sciences, Kunming, Yunnan, China
| |
Collapse
|
40
|
Xie C, Slagboom J, Albulescu LO, Somsen GW, Vonk FJ, Casewell NR, Kool J. Neutralising effects of small molecule toxin inhibitors on nanofractionated coagulopathic Crotalinae snake venoms. Acta Pharm Sin B 2020; 10:1835-1845. [PMID: 33163338 PMCID: PMC7606088 DOI: 10.1016/j.apsb.2020.09.005] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2020] [Revised: 09/03/2020] [Accepted: 09/04/2020] [Indexed: 11/29/2022] Open
Abstract
Repurposing small molecule drugs and drug candidates is considered as a promising approach to revolutionise the treatment of snakebite envenoming. In this study, we investigated the inhibiting effects of the small molecules varespladib (nonspecific phospholipase A2 inhibitor), marimastat (broad spectrum matrix metalloprotease inhibitor) and dimercaprol (metal ion chelator) against coagulopathic toxins found in Crotalinae (pit vipers) snake venoms. Venoms from Bothrops asper, Bothrops jararaca, Calloselasma rhodostoma and Deinagkistrodon acutus were separated by liquid chromatography, followed by nanofractionation and mass spectrometry identification undertaken in parallel. Nanofractions of the venom toxins were then subjected to a high-throughput coagulation assay in the presence of different concentrations of the small molecules under study. Anticoagulant venom toxins were mostly identified as phospholipases A2, while procoagulant venom activities were mainly associated with snake venom metalloproteinases and snake venom serine proteases. Varespladib was found to effectively inhibit most anticoagulant venom effects, and also showed some inhibition against procoagulant toxins. Contrastingly, marimastat and dimercaprol were both effective inhibitors of procoagulant venom activities but showed little inhibitory capability against anticoagulant toxins. The information obtained from this study aids our understanding of the mechanisms of action of toxin inhibitor drug candidates, and highlights their potential as future snakebite treatments.
Collapse
Key Words
- ACN, acetonitrile
- Antivenom
- CTL, C-type lectins
- Chelators
- DMSO, dimethyl sulfoxide
- Dimercaprol
- FA, formic acid
- HTS, high-throughput screening
- LC, liquid chromatography
- MS, mass spectrometry
- Marimastat
- NOI, no observed inhibition
- Nanofractionation
- PBS, phosphate buffered saline
- PLA2, phospholipase A2
- PN, partly neutralised at 20 μmol/L inhibitor concentrations
- SVMP, snake venom metalloproteinase
- SVSP, snake venom serine protease
- Snakebite
- TIC, total ion current
- Varespladib
- WHO, World Health Organization
- XIC, extracted ion current
Collapse
Affiliation(s)
- Chunfang Xie
- Amsterdam Institute of Molecular and Life Sciences, Division of BioAnalytical Chemistry, Department of Chemistry and Pharmaceutical Sciences, Faculty of Science, Vrije Universiteit Amsterdam, Amsterdam 1081HV, The Netherlands
- Centre for Analytical Sciences Amsterdam (CASA), Amsterdam 1098 XH, The Netherlands
| | - Julien Slagboom
- Amsterdam Institute of Molecular and Life Sciences, Division of BioAnalytical Chemistry, Department of Chemistry and Pharmaceutical Sciences, Faculty of Science, Vrije Universiteit Amsterdam, Amsterdam 1081HV, The Netherlands
- Centre for Analytical Sciences Amsterdam (CASA), Amsterdam 1098 XH, The Netherlands
| | - Laura-Oana Albulescu
- Centre for Snakebite Research and Interventions, Liverpool School of Tropical Medicine, Liverpool L3 5QA, UK
- Centre for Drugs and Diagnostics, Liverpool School of Tropical Medicine, Liverpool L3 5QA, UK
| | - Govert W. Somsen
- Amsterdam Institute of Molecular and Life Sciences, Division of BioAnalytical Chemistry, Department of Chemistry and Pharmaceutical Sciences, Faculty of Science, Vrije Universiteit Amsterdam, Amsterdam 1081HV, The Netherlands
- Centre for Analytical Sciences Amsterdam (CASA), Amsterdam 1098 XH, The Netherlands
| | - Freek J. Vonk
- Amsterdam Institute of Molecular and Life Sciences, Division of BioAnalytical Chemistry, Department of Chemistry and Pharmaceutical Sciences, Faculty of Science, Vrije Universiteit Amsterdam, Amsterdam 1081HV, The Netherlands
- Centre for Analytical Sciences Amsterdam (CASA), Amsterdam 1098 XH, The Netherlands
- Naturalis Biodiversity Center, Leiden 2333 CR, The Netherlands
| | - Nicholas R. Casewell
- Centre for Snakebite Research and Interventions, Liverpool School of Tropical Medicine, Liverpool L3 5QA, UK
- Centre for Drugs and Diagnostics, Liverpool School of Tropical Medicine, Liverpool L3 5QA, UK
| | - Jeroen Kool
- Amsterdam Institute of Molecular and Life Sciences, Division of BioAnalytical Chemistry, Department of Chemistry and Pharmaceutical Sciences, Faculty of Science, Vrije Universiteit Amsterdam, Amsterdam 1081HV, The Netherlands
- Centre for Analytical Sciences Amsterdam (CASA), Amsterdam 1098 XH, The Netherlands
| |
Collapse
|
41
|
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: 24] [Impact Index Per Article: 4.8] [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
|
42
|
Xie C, Albulescu LO, Bittenbinder MA, Somsen GW, Vonk FJ, Casewell NR, Kool J. Neutralizing Effects of Small Molecule Inhibitors and Metal Chelators on Coagulopathic Viperinae Snake Venom Toxins. Biomedicines 2020; 8:E297. [PMID: 32825484 PMCID: PMC7555180 DOI: 10.3390/biomedicines8090297] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2020] [Revised: 08/13/2020] [Accepted: 08/18/2020] [Indexed: 12/27/2022] Open
Abstract
Animal-derived antivenoms are the only specific therapies currently available for the treatment of snake envenoming, but these products have a number of limitations associated with their efficacy, safety and affordability for use in tropical snakebite victims. Small molecule drugs and drug candidates are regarded as promising alternatives for filling the critical therapeutic gap between snake envenoming and effective treatment. In this study, by using an advanced analytical technique that combines chromatography, mass spectrometry and bioassaying, we investigated the effect of several small molecule inhibitors that target phospholipase A2 (varespladib) and snake venom metalloproteinase (marimastat, dimercaprol and DMPS) toxin families on inhibiting the activities of coagulopathic toxins found in Viperinae snake venoms. The venoms of Echis carinatus, Echis ocellatus, Daboia russelii and Bitis arietans, which are known for their potent haemotoxicities, were fractionated in high resolution onto 384-well plates using liquid chromatography followed by coagulopathic bioassaying of the obtained fractions. Bioassay activities were correlated to parallel recorded mass spectrometric and proteomics data to assign the venom toxins responsible for coagulopathic activity and assess which of these toxins could be neutralized by the inhibitors under investigation. Our results showed that the phospholipase A2-inhibitor varespladib neutralized the vast majority of anticoagulation activities found across all of the tested snake venoms. Of the snake venom metalloproteinase inhibitors, marimastat demonstrated impressive neutralization of the procoagulation activities detected in all of the tested venoms, whereas dimercaprol and DMPS could only partially neutralize these activities at the doses tested. Our results provide additional support for the concept that combinations of small molecules, particularly the combination of varespladib with marimastat, serve as a drug-repurposing opportunity to develop new broad-spectrum inhibitor-based therapies for snakebite envenoming.
Collapse
Affiliation(s)
- Chunfang Xie
- Amsterdam Institute of Molecular and Life Sciences, Division of BioAnalytical Chemistry, Department of Chemistry and Pharmaceutical Sciences, Faculty of Science, Vrije Universiteit Amsterdam, De Boelelaan 1085, 1081HV Amsterdam, The Netherlands; (C.X.); (M.A.B.); (G.W.S.); (F.J.V.)
- Centre for Analytical Sciences Amsterdam (CASA), 1098 XH Amsterdam, The Netherlands
| | - Laura-Oana Albulescu
- Centre for Snakebite Research and Interventions, Liverpool School of Tropical Medicine, Pembroke Place, Liverpool L3 5QA, UK; (L.-O.A.); (N.R.C.)
- Centre for Drugs and Diagnostics, Liverpool School of Tropical Medicine, Pembroke Place, Liverpool L3 5QA, UK
| | - Mátyás A. Bittenbinder
- Amsterdam Institute of Molecular and Life Sciences, Division of BioAnalytical Chemistry, Department of Chemistry and Pharmaceutical Sciences, Faculty of Science, Vrije Universiteit Amsterdam, De Boelelaan 1085, 1081HV Amsterdam, The Netherlands; (C.X.); (M.A.B.); (G.W.S.); (F.J.V.)
- Centre for Analytical Sciences Amsterdam (CASA), 1098 XH Amsterdam, The Netherlands
- Naturalis Biodiversity Center, 2333 CR Leiden, The Netherlands
| | - Govert W. Somsen
- Amsterdam Institute of Molecular and Life Sciences, Division of BioAnalytical Chemistry, Department of Chemistry and Pharmaceutical Sciences, Faculty of Science, Vrije Universiteit Amsterdam, De Boelelaan 1085, 1081HV Amsterdam, The Netherlands; (C.X.); (M.A.B.); (G.W.S.); (F.J.V.)
- Centre for Analytical Sciences Amsterdam (CASA), 1098 XH Amsterdam, The Netherlands
| | - Freek J. Vonk
- Amsterdam Institute of Molecular and Life Sciences, Division of BioAnalytical Chemistry, Department of Chemistry and Pharmaceutical Sciences, Faculty of Science, Vrije Universiteit Amsterdam, De Boelelaan 1085, 1081HV Amsterdam, The Netherlands; (C.X.); (M.A.B.); (G.W.S.); (F.J.V.)
- Centre for Analytical Sciences Amsterdam (CASA), 1098 XH Amsterdam, The Netherlands
| | - Nicholas R. Casewell
- Centre for Snakebite Research and Interventions, Liverpool School of Tropical Medicine, Pembroke Place, Liverpool L3 5QA, UK; (L.-O.A.); (N.R.C.)
- Centre for Drugs and Diagnostics, Liverpool School of Tropical Medicine, Pembroke Place, Liverpool L3 5QA, UK
| | - Jeroen Kool
- Amsterdam Institute of Molecular and Life Sciences, Division of BioAnalytical Chemistry, Department of Chemistry and Pharmaceutical Sciences, Faculty of Science, Vrije Universiteit Amsterdam, De Boelelaan 1085, 1081HV Amsterdam, The Netherlands; (C.X.); (M.A.B.); (G.W.S.); (F.J.V.)
- Centre for Analytical Sciences Amsterdam (CASA), 1098 XH Amsterdam, The Netherlands
| |
Collapse
|
43
|
Xie C, Albulescu LO, Still KBM, Slagboom J, Zhao Y, Jiang Z, Somsen GW, Vonk FJ, Casewell NR, Kool J. Varespladib Inhibits the Phospholipase A 2 and Coagulopathic Activities of Venom Components from Hemotoxic Snakes. Biomedicines 2020; 8:E165. [PMID: 32560391 PMCID: PMC7345350 DOI: 10.3390/biomedicines8060165] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2020] [Revised: 06/04/2020] [Accepted: 06/11/2020] [Indexed: 11/17/2022] Open
Abstract
Phospholipase A2 (PLA2) enzymes are important toxins found in many snake venoms, and they can exhibit a variety of toxic activities including causing hemolysis and/or anticoagulation. In this study, the inhibiting effects of the small molecule PLA2 inhibitor varespladib on snake venom PLA2s was investigated by nanofractionation analytics, which combined chromatography, mass spectrometry (MS), and bioassays. The venoms of the medically important snake species Bothrops asper, Calloselasma rhodostoma, Deinagkistrodon acutus, Daboia russelii, Echis carinatus, Echis ocellatus, and Oxyuranus scutellatus were separated by liquid chromatography (LC) followed by nanofractionation and interrogation of the fractions by a coagulation assay and a PLA2 assay. Next, we assessed the ability of varespladib to inhibit the activity of enzymatic PLA2s and the coagulopathic toxicities induced by fractionated snake venom toxins, and identified these bioactive venom toxins and those inhibited by varespladib by using parallel recorded LC-MS data and proteomics analysis. We demonstrated here that varespladib was not only capable of inhibiting the PLA2 activities of hemotoxic snake venoms, but can also effectively neutralize the coagulopathic toxicities (most profoundly anticoagulation) induced by venom toxins. While varespladib effectively inhibited PLA2 toxins responsible for anticoagulant effects, we also found some evidence that this inhibitory molecule can partially abrogate procoagulant venom effects caused by different toxin families. These findings further emphasize the potential clinical utility of varespladib in mitigating the toxic effects of certain snakebites.
Collapse
Affiliation(s)
- Chunfang Xie
- Amsterdam Institute of Molecular and Life Sciences, Division of BioAnalytical Chemistry, Department of Chemistry and Pharmaceutical Sciences, Faculty of Sciences, Vrije Universiteit Amsterdam, De Boelelaan 1085, 1081 HV Amsterdam, The Netherlands; (C.X.); (K.B.M.S.); (J.S.); (G.W.S.); (F.J.V.)
- Centre for Analytical Sciences Amsterdam (CASA), 1098 XH Amsterdam, The Netherlands
| | - Laura-Oana Albulescu
- Centre for Snakebite Research and Interventions, Liverpool School of Tropical Medicine, Pembroke Place, Liverpool L3 5QA, UK; (L.-O.A.); (N.R.C.)
- Centre for Drugs and Diagnostics, Liverpool School of Tropical Medicine, Pembroke Place, Liverpool L3 5QA, UK
| | - Kristina B. M. Still
- Amsterdam Institute of Molecular and Life Sciences, Division of BioAnalytical Chemistry, Department of Chemistry and Pharmaceutical Sciences, Faculty of Sciences, Vrije Universiteit Amsterdam, De Boelelaan 1085, 1081 HV Amsterdam, The Netherlands; (C.X.); (K.B.M.S.); (J.S.); (G.W.S.); (F.J.V.)
- Centre for Analytical Sciences Amsterdam (CASA), 1098 XH Amsterdam, The Netherlands
| | - Julien Slagboom
- Amsterdam Institute of Molecular and Life Sciences, Division of BioAnalytical Chemistry, Department of Chemistry and Pharmaceutical Sciences, Faculty of Sciences, Vrije Universiteit Amsterdam, De Boelelaan 1085, 1081 HV Amsterdam, The Netherlands; (C.X.); (K.B.M.S.); (J.S.); (G.W.S.); (F.J.V.)
- Centre for Analytical Sciences Amsterdam (CASA), 1098 XH Amsterdam, The Netherlands
| | - Yumei Zhao
- Institute of Pharmaceutical Analysis, College of Pharmacy, Jinan University, Huangpu Avenue West 601, Guangzhou 510632, China; (Y.Z.); (Z.J.)
| | - Zhengjin Jiang
- Institute of Pharmaceutical Analysis, College of Pharmacy, Jinan University, Huangpu Avenue West 601, Guangzhou 510632, China; (Y.Z.); (Z.J.)
| | - Govert W. Somsen
- Amsterdam Institute of Molecular and Life Sciences, Division of BioAnalytical Chemistry, Department of Chemistry and Pharmaceutical Sciences, Faculty of Sciences, Vrije Universiteit Amsterdam, De Boelelaan 1085, 1081 HV Amsterdam, The Netherlands; (C.X.); (K.B.M.S.); (J.S.); (G.W.S.); (F.J.V.)
- Centre for Analytical Sciences Amsterdam (CASA), 1098 XH Amsterdam, The Netherlands
| | - Freek J. Vonk
- Amsterdam Institute of Molecular and Life Sciences, Division of BioAnalytical Chemistry, Department of Chemistry and Pharmaceutical Sciences, Faculty of Sciences, Vrije Universiteit Amsterdam, De Boelelaan 1085, 1081 HV Amsterdam, The Netherlands; (C.X.); (K.B.M.S.); (J.S.); (G.W.S.); (F.J.V.)
- Centre for Analytical Sciences Amsterdam (CASA), 1098 XH Amsterdam, The Netherlands
- Naturalis Biodiversity Center, Darwinweg 2, 2333 CR Leiden, The Netherlands
| | - Nicholas R. Casewell
- Centre for Snakebite Research and Interventions, Liverpool School of Tropical Medicine, Pembroke Place, Liverpool L3 5QA, UK; (L.-O.A.); (N.R.C.)
- Centre for Drugs and Diagnostics, Liverpool School of Tropical Medicine, Pembroke Place, Liverpool L3 5QA, UK
| | - Jeroen Kool
- Amsterdam Institute of Molecular and Life Sciences, Division of BioAnalytical Chemistry, Department of Chemistry and Pharmaceutical Sciences, Faculty of Sciences, Vrije Universiteit Amsterdam, De Boelelaan 1085, 1081 HV Amsterdam, The Netherlands; (C.X.); (K.B.M.S.); (J.S.); (G.W.S.); (F.J.V.)
- Centre for Analytical Sciences Amsterdam (CASA), 1098 XH Amsterdam, The Netherlands
| |
Collapse
|
44
|
Zinenko O, Tovstukha I, Korniyenko Y. PLA 2 Inhibitor Varespladib as an Alternative to the Antivenom Treatment for Bites from Nikolsky's Viper Vipera berus nikolskii. Toxins (Basel) 2020; 12:toxins12060356. [PMID: 32485836 PMCID: PMC7354479 DOI: 10.3390/toxins12060356] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2019] [Revised: 08/27/2019] [Accepted: 08/27/2019] [Indexed: 12/05/2022] Open
Abstract
Although envenoming by a small East European species of viper is rarely severe, and only exceptionally fatal, lack of specific antivenom stocks in a few areas within this region and possible severe side effects of antivenom application leave most bites to be treated only with antihistamines and supportive therapy. Varespladib is an effective inhibitor of snake phospholipase, and, as such, it could be considered as first-line therapy. The Nikolsky’s viper venom contains an extremely high concentration of phospholipase A2 (PLA2), responsible for the toxic effects of the venom, as well as minor amounts of other toxins. If Varespladib can successfully inhibit PLA2 activity, the Nikolsky’s viper could be one of the first venomous snakes having an antitoxin-specific treatment regimen. To assess that, Varespladib was administered alone subcutaneously to adult male CD-1 mice (8 mg/kg) and compared to mice exposed to Vipera berus nikolskii crude venom (8 mg/kg = 10 LD50) or a combination of Varespladib and the same amount of the venom. Experimental animals were monitored for the presence of envenoming symptoms and mortality for 48 h after injection. Eighty percent of mice receiving both Varespladib and venom survived, while 100% of the control group receiving venom alone died within 4 h. Experimental results are consistent with Varespladib acting as an effective antitoxin in the mouse model against Nikolsky’s viper venom. Further studies are needed under experimental conditions that more closely resemble natural envenoming (i.e., delayed administration).
Collapse
Affiliation(s)
- Oleksandr Zinenko
- V. N. Karazin Kharkiv University, 61058 Kharkiv, Ukraine;
- Correspondence:
| | - Igor Tovstukha
- Ukrainian Independent Ecology Institute, 61001 Kharkiv, Ukraine;
| | | |
Collapse
|
45
|
Diagnostic and Therapeutic Value of Aptamers in Envenomation Cases. Int J Mol Sci 2020; 21:ijms21103565. [PMID: 32443562 PMCID: PMC7278915 DOI: 10.3390/ijms21103565] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2020] [Revised: 05/11/2020] [Accepted: 05/13/2020] [Indexed: 02/07/2023] Open
Abstract
It is now more than a century since Albert Calmette from the Institut Pasteur changed the world of envenomation by demonstrating that antibodies raised against animal venoms have the ability to treat human victims of previously fatal bites or stings. Moreover, the research initiated at that time effectively launched the discipline of toxicology, first leading to the search for toxic venom components, followed by the demonstration of venoms that also contained compounds of therapeutic value. Interest from pharmaceutical companies to treat envenomation is, however, declining, mainly for economic reasons, and hence, the World Health Organization has reclassified this public health issue to be a highest priority concern. While the production, storage, and safety of antivenom sera suffer from major inconveniences, alternative chemical and technological approaches to the problem of envenomation need to be considered that bypass the use of antibodies for toxin neutralization. Herein, we review an emerging strategy that relies on the use of aptamers and discuss how close—or otherwise—we are to finding a viable alternative to the use of antibodies for the therapy of human envenomation.
Collapse
|
46
|
Albulescu LO, Hale MS, Ainsworth S, Alsolaiss J, Crittenden E, Calvete JJ, Evans C, Wilkinson MC, Harrison RA, Kool J, Casewell NR. Preclinical validation of a repurposed metal chelator as an early-intervention therapeutic for hemotoxic snakebite. Sci Transl Med 2020; 12:eaay8314. [PMID: 32376771 PMCID: PMC7116364 DOI: 10.1126/scitranslmed.aay8314] [Citation(s) in RCA: 63] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2019] [Revised: 01/22/2020] [Accepted: 03/19/2020] [Indexed: 12/18/2022]
Abstract
Snakebite envenoming causes 138,000 deaths annually, and ~400,000 victims are left with permanent disabilities. Envenoming by saw-scaled vipers (Viperidae: Echis) leads to systemic hemorrhage and coagulopathy and represents a major cause of snakebite mortality and morbidity in Africa and Asia. The only specific treatment for snakebite, antivenom, has poor specificity and low affordability and must be administered in clinical settings because of its intravenous delivery and high rates of adverse reactions. This requirement results in major treatment delays in resource-poor regions and substantially affects patient outcomes after envenoming. Here, we investigated the value of metal ion chelators as prehospital therapeutics for snakebite. Among the tested chelators, dimercaprol (British anti-Lewisite) and its derivative 2,3-dimercapto-1-propanesulfonic acid (DMPS) were found to potently antagonize the activity of Zn2+-dependent snake venom metalloproteinases in vitro. Moreover, DMPS prolonged or conferred complete survival in murine preclinical models of envenoming against a variety of saw-scaled viper venoms. DMPS also considerably extended survival in a "challenge and treat" model, where drug administration was delayed after venom injection and the oral administration of this chelator provided partial protection against envenoming. Last, the potential clinical scenario of early oral DMPS therapy combined with a delayed, intravenous dose of conventional antivenom provided prolonged protection against the lethal effects of envenoming in vivo. Our findings demonstrate that the safe and affordable repurposed metal chelator DMPS can effectively neutralize saw-scaled viper venoms in vitro and in vivo and highlight the promise of this drug as an early, prehospital, therapeutic intervention for hemotoxic snakebite envenoming.
Collapse
Affiliation(s)
- Laura-Oana Albulescu
- Centre for Snakebite Research and Interventions, Liverpool School of Tropical Medicine, Pembroke Place, L3 5QA Liverpool, UK
| | - Melissa S Hale
- Centre for Snakebite Research and Interventions, Liverpool School of Tropical Medicine, Pembroke Place, L3 5QA Liverpool, UK
| | - Stuart Ainsworth
- Centre for Snakebite Research and Interventions, Liverpool School of Tropical Medicine, Pembroke Place, L3 5QA Liverpool, UK
| | - Jaffer Alsolaiss
- Centre for Snakebite Research and Interventions, Liverpool School of Tropical Medicine, Pembroke Place, L3 5QA Liverpool, UK
| | - Edouard Crittenden
- Centre for Snakebite Research and Interventions, Liverpool School of Tropical Medicine, Pembroke Place, L3 5QA Liverpool, UK
| | - Juan J Calvete
- Laboratorio de Venómica Estructural y Funcional, Instituto de Biomedicina de Valencia, CSIC, Valencia 46010, Spain
| | - Chloe Evans
- Centre for Snakebite Research and Interventions, Liverpool School of Tropical Medicine, Pembroke Place, L3 5QA Liverpool, UK
| | - Mark C Wilkinson
- Centre for Snakebite Research and Interventions, Liverpool School of Tropical Medicine, Pembroke Place, L3 5QA Liverpool, UK
| | - Robert A Harrison
- Centre for Snakebite Research and Interventions, Liverpool School of Tropical Medicine, Pembroke Place, L3 5QA Liverpool, UK
- Centre for Drugs and Diagnostics, Liverpool School of Tropical Medicine, Pembroke Place, L3 5QA Liverpool, UK
| | - Jeroen Kool
- Amsterdam Institute for Molecules Medicines and Systems, Division of BioAnalytical Chemistry, Department of Chemistry and Pharmaceutical Sciences, Faculty of Sciences, Vrije Universiteit Amsterdam, De Boelelaan 1083, 1081HV Amsterdam, Netherlands
| | - Nicholas R Casewell
- Centre for Snakebite Research and Interventions, Liverpool School of Tropical Medicine, Pembroke Place, L3 5QA Liverpool, UK.
- Centre for Drugs and Diagnostics, Liverpool School of Tropical Medicine, Pembroke Place, L3 5QA Liverpool, UK
| |
Collapse
|
47
|
Knudsen C, Ledsgaard L, Dehli RI, Ahmadi S, Sørensen CV, Laustsen AH. Engineering and design considerations for next-generation snakebite antivenoms. Toxicon 2019; 167:67-75. [DOI: 10.1016/j.toxicon.2019.06.005] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2019] [Revised: 05/22/2019] [Accepted: 06/03/2019] [Indexed: 11/27/2022]
|
48
|
Williams HF, Layfield HJ, Vallance T, Patel K, Bicknell AB, Trim SA, Vaiyapuri S. The Urgent Need to Develop Novel Strategies for the Diagnosis and Treatment of Snakebites. Toxins (Basel) 2019; 11:E363. [PMID: 31226842 PMCID: PMC6628419 DOI: 10.3390/toxins11060363] [Citation(s) in RCA: 73] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2019] [Revised: 06/18/2019] [Accepted: 06/18/2019] [Indexed: 01/09/2023] Open
Abstract
Snakebite envenoming (SBE) is a priority neglected tropical disease, which kills in excess of 100,000 people per year. Additionally, many millions of survivors also suffer through disabilities and long-term health consequences. The only treatment for SBE, antivenom, has a number of major associated problems, not least, adverse reactions and limited availability. This emphasises the necessity for urgent improvements to the management of this disease. Administration of antivenom is too frequently based on symptomatology, which results in wasting crucial time. The majority of SBE-affected regions rely on broad-spectrum polyvalent antivenoms that have a low content of case-specific efficacious immunoglobulins. Research into small molecular therapeutics such as varespladib/methyl-varespladib (PLA2 inhibitors) and batimastat/marimastat (metalloprotease inhibitors) suggest that such adjunctive treatments could be hugely beneficial to victims. Progress into toxin-specific monoclonal antibodies as well as alternative binding scaffolds such as aptamers hold much promise for future treatment strategies. SBE is not implicit during snakebite, due to venom metering. Thus, the delay between bite and symptom presentation is critical and when symptoms appear it may often already be too late to effectively treat SBE. The development of reliable diagnostical tools could therefore initiate a paradigm shift in the treatment of SBE. While the complete eradication of SBE is an impossibility, mitigation is in the pipeline, with new treatments and diagnostics rapidly emerging. Here we critically review the urgent necessity for the development of diagnostic tools and improved therapeutics to mitigate the deaths and disabilities caused by SBE.
Collapse
Affiliation(s)
| | | | - Thomas Vallance
- School of Pharmacy, University of Reading, Reading RG6 6AH, UK.
| | - Ketan Patel
- School of Biological Sciences, University of Reading, Reading RG6 6AH, UK.
| | - Andrew B Bicknell
- School of Biological Sciences, University of Reading, Reading RG6 6AH, UK.
| | | | | |
Collapse
|
49
|
Nikolaou A, Kokotou MG, Vasilakaki S, Kokotos G. Small-molecule inhibitors as potential therapeutics and as tools to understand the role of phospholipases A 2. Biochim Biophys Acta Mol Cell Biol Lipids 2019; 1864:941-956. [PMID: 30905350 PMCID: PMC7106526 DOI: 10.1016/j.bbalip.2018.08.009] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2018] [Revised: 08/10/2018] [Accepted: 08/16/2018] [Indexed: 11/20/2022]
Abstract
Phospholipase A2 (PLA2) enzymes are involved in various inflammatory pathological conditions including arthritis, cardiovascular and autoimmune diseases. The regulation of their catalytic activity is of high importance and a great effort has been devoted in developing synthetic inhibitors. We summarize the most important small-molecule 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 LpPLA2). We discuss recent applications of inhibitors to understand the role of each PLA2 type and their therapeutic potential. Potent and selective PLA2 inhibitors have been developed. Although some of them have been evaluated in clinical trials, none reached the market yet. Apart from their importance as potential medicinal agents, PLA2 inhibitors are excellent tools to unveil the role that each PLA2 type plays in cells and in vivo. Modern medicinal chemistry approaches are expected to generate improved PLA2 inhibitors as new agents to treat inflammatory diseases.
Collapse
Affiliation(s)
- Aikaterini Nikolaou
- Department of Chemistry, National and Kapodistrian University of Athens, Panepistimiopolis, Athens 15771, Greece
| | - Maroula G Kokotou
- Department of Chemistry, National and Kapodistrian University of Athens, Panepistimiopolis, Athens 15771, Greece
| | - Sofia Vasilakaki
- Department of Chemistry, National and Kapodistrian University of Athens, Panepistimiopolis, Athens 15771, Greece
| | - George Kokotos
- Department of Chemistry, National and Kapodistrian University of Athens, Panepistimiopolis, Athens 15771, Greece.
| |
Collapse
|
50
|
Neutralizing properties of LY315920 toward snake venom group I and II myotoxic phospholipases A2. Toxicon 2019; 157:1-7. [DOI: 10.1016/j.toxicon.2018.11.292] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2018] [Revised: 10/25/2018] [Accepted: 11/09/2018] [Indexed: 12/22/2022]
|