1
|
Smith CF, Baldé MA, French S, Modahl CM, Bahrabadi L, Amponsah-Asamoah M, Larson KY, Maroney SP, Galindo DC, Millimouno M, Camara N, Benjamin J, Brandehoff NP, McCabe MC, Cohen MJ, Jackson K, Baldé C, Castoe TA, Mackessy SP, Hansen KC, Saviola AJ. Discerning specific thrombolytic activities and blood clot degradomes of diverse snake venoms with untargeted peptidomics. J Thromb Haemost 2025:S1538-7836(25)00187-4. [PMID: 40122467 DOI: 10.1016/j.jtha.2025.03.012] [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: 10/25/2024] [Revised: 02/20/2025] [Accepted: 03/11/2025] [Indexed: 03/25/2025]
Abstract
BACKGROUND Many snake venoms have been shown to possess thrombolytic activity. However, it remains unclear if actions on other clot-stabilizing proteins beyond fibrin chains contribute significantly to venom-induced thrombolysis because the clot-wide targets of venom proteases and the mechanisms responsible for thrombolysis are not well understood. OBJECTIVES Here, we utilized a high-throughput, time-based thrombolysis assay in combination with untargeted peptidomics to provide comprehensive insight into the effects of venom from 5 snake species on blood clot degradation. METHODS We compared thrombolytic profiles across venoms with variable levels of proteases and generated venom-specific fingerprints of cleavage specificity. We also compared the specific effects of venoms that possess a range of thrombolytic activity on fibrin chains and other clot-bound proteins involved in clot structure. RESULTS Protease-rich venom more effectively degraded blood clots. Venoms with higher thrombolytic activity demonstrated an enhanced ability to target multiple sites across fibrin chains critical to clot stability and structure, as well as clot-stabilizing proteins including factor XIII, fibronectin, and vitronectin. CONCLUSION Collectively, this study significantly expands our understanding of the thrombolytic and fibrinolytic effects of snake venom by determining the full suite of clot-specific venom targets that are involved in clot formation and stability. This has important implications for the treatment of snake envenomation, the bioprospecting of therapeutically useful molecules, and the development of research tools for investigating hematologic disorders.
Collapse
Affiliation(s)
- Cara F Smith
- Department of Biochemistry and Molecular Genetics, University of Colorado Denver, Aurora, Colorado, USA; Asclepius Snakebite Foundation, Aurora, Colorado, USA
| | - Mamadou Alpha Baldé
- Asclepius Snakebite Foundation, Aurora, Colorado, USA; Snakebite Center of Excellence, Institut de Recherche en Biologie Appliqueé de Guinée (IRBAG), Kindia, Guinea
| | - Stephanie French
- Department of Tropical Disease Biology, Centre for Snakebite Research and Interventions, Liverpool School of Tropical Medicine, Liverpool, United Kingdom
| | - Cassandra M Modahl
- Department of Tropical Disease Biology, Centre for Snakebite Research and Interventions, Liverpool School of Tropical Medicine, Liverpool, United Kingdom
| | - Lilyrose Bahrabadi
- Department of Biochemistry and Molecular Genetics, University of Colorado Denver, Aurora, Colorado, USA
| | - Merilyn Amponsah-Asamoah
- Department of Biochemistry and Molecular Genetics, University of Colorado Denver, Aurora, Colorado, USA
| | - Keira Y Larson
- Department of Biochemistry and Molecular Genetics, University of Colorado Denver, Aurora, Colorado, USA
| | - Sean P Maroney
- Department of Biochemistry and Molecular Genetics, University of Colorado Denver, Aurora, Colorado, USA
| | - David Ceja Galindo
- Department of Biochemistry and Molecular Genetics, University of Colorado Denver, Aurora, Colorado, USA
| | - Martin Millimouno
- Asclepius Snakebite Foundation, Aurora, Colorado, USA; Snakebite Center of Excellence, Institut de Recherche en Biologie Appliqueé de Guinée (IRBAG), Kindia, Guinea
| | - Naby Camara
- Asclepius Snakebite Foundation, Aurora, Colorado, USA; Snakebite Center of Excellence, Institut de Recherche en Biologie Appliqueé de Guinée (IRBAG), Kindia, Guinea
| | | | - Nicklaus P Brandehoff
- Asclepius Snakebite Foundation, Aurora, Colorado, USA; Rocky Mountain Poison and Drug Center, Denver Health and Hospital Authority, Denver, Colorado, USA
| | - Maxwell C McCabe
- Department of Biochemistry and Molecular Genetics, University of Colorado Denver, Aurora, Colorado, USA
| | - Mitchell J Cohen
- Department of Surgery, University of Colorado Denver, Aurora, Colorado, USA
| | - Kate Jackson
- Asclepius Snakebite Foundation, Aurora, Colorado, USA; Biology Department, Whitman College, Walla Walla, Washington, USA
| | - Cellou Baldé
- Asclepius Snakebite Foundation, Aurora, Colorado, USA; Snakebite Center of Excellence, Institut de Recherche en Biologie Appliqueé de Guinée (IRBAG), Kindia, Guinea
| | - Todd A Castoe
- Department of Biology, University of Texas at Arlington, Arlington, Texas, USA
| | - Stephen P Mackessy
- Department of Biological Sciences, University of Northern Colorado, Greeley, Colorado, USA
| | - Kirk C Hansen
- Department of Biochemistry and Molecular Genetics, University of Colorado Denver, Aurora, Colorado, USA
| | - Anthony J Saviola
- Department of Biochemistry and Molecular Genetics, University of Colorado Denver, Aurora, Colorado, USA.
| |
Collapse
|
2
|
Jia Y, Garcia A, Reyes E. Single-Chain Variable Fragments: Targeting Snake Venom Phospholipase A 2 and Serine Protease. Toxins (Basel) 2025; 17:55. [PMID: 39998072 PMCID: PMC11860530 DOI: 10.3390/toxins17020055] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2024] [Revised: 01/16/2025] [Accepted: 01/22/2025] [Indexed: 02/26/2025] Open
Abstract
Snakebite is a critical global public health issue, causing substantial mortality and morbidity, particularly in tropical and subtropical regions. The development of innovative antivenoms targeting snake venom toxins is therefore of paramount importance. In this study, we adopted an epitope-directed approach to design three degenerate 15-mer peptides based on amino acid sequence alignments of snake venom phospholipase A2s (PLA2s) and snake venom serine proteases (SVSPs) from snake (Crotalus atrox). By leveraging their immunogenic and inhibitory profiles, these peptides were specifically designed to target the Asp49 and Lys49 variants of PLA2 and SVSP toxins. Groups of five mice were immunized with each peptide, and IgG mRNA was subsequently extracted from peripheral blood mononuclear cells (PBMCs) and spleen lymphocytes of the top three responders. The extracted mRNA was reverse-transcribed into complementary DNA (cDNA), and the variable regions of the IgG heavy and kappa chains were amplified using polymerase chain reaction (PCR). These amplified regions were then linked with a 66-nucleotide spacer to construct single-chain variable fragments (scFvs). Sequence analysis of 48 randomly selected plasmids from each PLA2 and SVSP scFv library revealed that over 80% contained scFv sequences with notable diversity observed in the complementarity-determining regions (CDRs), particularly CDR3. Enzyme-linked immunosorbent assay (ELISA) results demonstrated that the SP peptide elicited a broader immune response in mice compared to the Asp49 peptide, implying the strong immunogenicity of the SP peptide. These scFvs represent a promising foundation for the development of recombinant human monoclonal antibodies targeting snake PLA2 and SVSP toxins, providing a potential therapeutic strategy for the treatment of snakebites.
Collapse
Affiliation(s)
- Ying Jia
- School of Integrative Biological and Chemical Sciences, University of Texas Rio Grande Valley, Brownsville, TX 78520, USA; (A.G.); (E.R.)
| | | | | |
Collapse
|
3
|
Vidal JFD, Schwartz MF, Garay AV, Valadares NF, Bueno RV, Monteiro ACL, de Freitas SM, Barbosa JARG. Exploring the Diversity and Function of Serine Proteases in Toxicofera Reptile Venoms: A Comprehensive Overview. Toxins (Basel) 2024; 16:428. [PMID: 39453204 PMCID: PMC11511063 DOI: 10.3390/toxins16100428] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2024] [Revised: 07/05/2024] [Accepted: 07/08/2024] [Indexed: 10/26/2024] Open
Abstract
Toxicofera reptile venoms are composed of several toxins, including serine proteases. These proteases are glycosylated enzymes that affect the prey's hemostatic system. Their actions extend across the coagulation cascade, the kallikrein-kinin system, and platelet activation. Despite their specificity for different substrates, these enzymes are homologous across all toxicoferans and display high sequence similarity. The aim of this review is to compile decades of knowledge about venom serine proteases, showing the diversity of biochemically and biophysically characterized enzymes, their structural characteristics, advances in understanding their origin and evolution, as well as methods of obtaining enzymes and their biotechnological applications.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - João Alexandre R. G. Barbosa
- Laboratory of Molecular Biophysics, Department of Cell Biology, Institute of Biological Sciences, Darcy Ribeiro Campus, University of Brasília, Asa Norte, Brasilia 70910-900, DF, Brazil
| |
Collapse
|
4
|
Dobson J, Chowdhury A, Tai-A-Pin J, van der Ploeg H, Gillett A, Fry BG. The Clot Thickens: Differential Coagulotoxic and Cardiotoxic Activities of Anguimorpha Lizard Venoms. Toxins (Basel) 2024; 16:283. [PMID: 38922177 PMCID: PMC11209219 DOI: 10.3390/toxins16060283] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Revised: 06/11/2024] [Accepted: 06/17/2024] [Indexed: 06/27/2024] Open
Abstract
Despite their evolutionary novelty, lizard venoms are much less studied in comparison to the intense research on snake venoms. While the venoms of helodermatid lizards have long been assumed to be for defensive purposes, there is increasing evidence of toxic activities more useful for predation than defence (such as paralytic neurotoxicity). This study aimed to ascertain the effects of Heloderma, Lanthanotus, and Varanus lizard venoms on the coagulation and cardiovascular systems. Anticoagulant toxicity was demonstrated for the Varanus species studied, with the venoms prolonging clotting times in human and bird plasma due to the destructive cleavage of fibrinogen. In contrast, thromboelastographic analyses on human and bird plasmas in this study demonstrated a procoagulant bioactivity for Heloderma venoms. A previous study on Heloderma venom using factor-depleted plasmas as a proxy model suggested a procoagulant factor was present that activated either Factor XI or Factor XII, but could not ascertain the precise target. Our activation studies using purified zymogens confirmed FXII activation. Comparisons of neonate and adult H. exasperatum, revealed the neonates to be more potent in the ability to activate FXII, being more similar to the venom of the smaller species H. suspectum than the adult H. exasperatum. This suggests potent FXII activation a basal trait in the genus, present in the small bodied last common ancestor. This also indicates an ontogenetic difference in prey preferences in the larger Heloderma species paralleing the change in venom biochemistry. In addition, as birds lack Factor XII, the ability to clot avian plasma suggested an additional procoagulant site of action, which was revealed to be the activation of Factor VII, with H. horridum being the most potent. This study also examined the effects upon the cardiovascular system, including the liberation of kinins from kininogen, which contributes to hypotension induction. This form of toxicity was previously described for Heloderma venoms, and was revealed in this study was to also be a pathophysiological effect of Lanthanotus and Varanus venoms. This suggests that this toxic activity was present in the venom of the last common ancestor of the anguimorph lizards, which is consistent with kallikrein enzymes being a shared toxin trait. This study therefore uncovered novel actions of anguimorph lizard venoms, not only contributing to the evolutionary biology body of knowledge but also revealing novel activities to mine for drug design lead compounds.
Collapse
Affiliation(s)
- James Dobson
- Adaptive Biotoxicology Lab, School of the Environment, University of Queensland, St Lucia, QLD 4072, Australia; (J.D.); (A.C.)
| | - Abhinandan Chowdhury
- Adaptive Biotoxicology Lab, School of the Environment, University of Queensland, St Lucia, QLD 4072, Australia; (J.D.); (A.C.)
| | | | - Harold van der Ploeg
- Working Group Adder Research Netherlands, RAVON, 6525 ED Nijmegen, The Netherlands;
| | - Amber Gillett
- FaunaVet Wildlife Consultancy, Glass House Mountains, QLD 4518, Australia;
| | - Bryan G. Fry
- Adaptive Biotoxicology Lab, School of the Environment, University of Queensland, St Lucia, QLD 4072, Australia; (J.D.); (A.C.)
| |
Collapse
|
5
|
Cañas CA, Castaño-Valencia S, Castro-Herrera F. The Colombian bushmasters Lachesis acrochorda (García, 1896) and Lachesis muta (Linnaeus, 1766): Snake species, venoms, envenomation, and its management. Toxicon 2023; 230:107152. [PMID: 37178796 DOI: 10.1016/j.toxicon.2023.107152] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2023] [Revised: 05/02/2023] [Accepted: 05/04/2023] [Indexed: 05/15/2023]
Abstract
In Colombia, there are two species of bushmaster snakes, Lachesis acrochorda, which is distributed mainly in the west of the country (in the Choco region), and Lachesis muta in the southeast (in the Amazon and Orinoquia region), whose presence has been reduced due to the destruction of their habitats. Captive maintenance is challenging, making it difficult to obtain their venom for study and antivenom manufacturing. They are the largest vipers in the world. The occurrence of human envenomation is quite rare, but when it occurs, it is associated with high mortality. Bushmaster venom is necrotizing, hemorrhagic, myotoxic, hemolytic, and cardiovascular depressant. Due to the presence of bradycardia, hypotension, emesis, and diarrhea in some patients (Lachesis syndrome), the possibility of a vagal or cholinergic effect is raised. The treatment of envenomation is hindered by the scarcity of antivenom and the need to use high doses. A review of the most relevant biological and medical aspects of bushmaster snakes is presented, mainly for those occurring in Colombia, to facilitate their recognition and raise awareness about the need for special attention to improve their conservation and advance scientific knowledge, in particular, about their venom.
Collapse
Affiliation(s)
- Carlos A Cañas
- Universidad Icesi, CIRAT: Centro de Investigación en Reumatología, Autoinmunidad y Medicina Traslacional, Cali, 760031, Colombia; Fundación Valle del Lili, Departamento de Reumatología, Cali, 760026, Colombia.
| | - Santiago Castaño-Valencia
- Department of Physiological Sciences, Department of Health Sciences, Universidad del Valle. Cali 760042, Colombia; Laboratorio de Herpetología y Toxinología, Universidad del Valle. Cali 760042, Colombia
| | | |
Collapse
|
6
|
Diniz-Sousa R, Silva CCA, Pereira SS, da Silva SL, Fernandes PA, Teixeira LMC, Zuliani JP, Soares AM. Therapeutic applications of snake venoms: An invaluable potential of new drug candidates. Int J Biol Macromol 2023; 238:124357. [PMID: 37028634 DOI: 10.1016/j.ijbiomac.2023.124357] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2022] [Revised: 04/01/2023] [Accepted: 04/03/2023] [Indexed: 04/09/2023]
Abstract
Animal venoms and their chemical compounds have aroused both empirical and scientific attention for ages. However, there has been a significant increase in scientific investigations in recent decades, allowing the production of various formulations that are helping in the development of many important tools for biotechnological, diagnostic, or therapeutic use, both in human and animal health, as well as in plants. Venoms are composed of biomolecules and inorganic compounds that may have physiological and pharmacological activities that are not related to their principal actions (prey immobilization, digestion, and defense). Snake venom toxins, mainly enzymatic and non-enzymatic proteins, and peptides have been identified as potential prototypes for new drugs and/or models for the development of pharmacologically active structural domains for the treatment of cancer, cardiovascular diseases, neurodegenerative and autoimmune diseases, pain, and infectious-parasitic diseases. This minireview aims to provide an overview of the biotechnological potential of animal venoms, with a focus on snakes, and to introduce the reader to the fascinating world of Applied Toxinology, where animal biodiversity can be used to develop therapeutic and diagnostic applications for humans.
Collapse
Affiliation(s)
- Rafaela Diniz-Sousa
- Laboratório de Biotecnologia de Proteínas e Compostos Bioativos Aplicados à Saúde (LABIOPROT), Fundação Oswaldo Cruz, FIOCRUZ, Unidade Rondônia, Porto Velho, Rondônia, Brazil; Centro Universitário São Lucas (UniSL), Porto Velho, Rondônia, Brazil
| | - Cleópatra C A Silva
- Laboratório de Biotecnologia de Proteínas e Compostos Bioativos Aplicados à Saúde (LABIOPROT), Fundação Oswaldo Cruz, FIOCRUZ, Unidade Rondônia, Porto Velho, Rondônia, Brazil; Instituto Nacional de Ciência e Tecnologia de Epidemiologia da Amazônia Ocidental (INCT-EpiAmO), Porto Velho, Rondônia, Brazil
| | - Soraya S Pereira
- Laboratório de Engenharia de Anticorpos, Fundação Oswaldo Cruz, FIOCRUZ, Unidade Rondônia, Porto Velho, Rondônia, Brazil
| | - Saulo L da Silva
- LAQV/Requimte, University of Porto, Rua do Campo Alegre s/n, Porto, Portugal; Department of Chemistry and Biochemistry, Faculty of Sciences, University of Porto, Rua do Campo Alegre s/n, Porto, Portugal; Faculty of Chemical Sciences, University of Cuenca, Cuenca, Azuay, Ecuador
| | - Pedro A Fernandes
- LAQV/Requimte, University of Porto, Rua do Campo Alegre s/n, Porto, Portugal; Department of Chemistry and Biochemistry, Faculty of Sciences, University of Porto, Rua do Campo Alegre s/n, Porto, Portugal
| | - Luís M C Teixeira
- LAQV/Requimte, University of Porto, Rua do Campo Alegre s/n, Porto, Portugal; Department of Chemistry and Biochemistry, Faculty of Sciences, University of Porto, Rua do Campo Alegre s/n, Porto, Portugal
| | - Juliana P Zuliani
- Laboratório de Imunologia Celular Aplicada à Saúde, Fundação Oswaldo Cruz, FIOCRUZ, Unidade Rondônia, Porto Velho, Rondônia, Brazil
| | - Andreimar M Soares
- Laboratório de Biotecnologia de Proteínas e Compostos Bioativos Aplicados à Saúde (LABIOPROT), Fundação Oswaldo Cruz, FIOCRUZ, Unidade Rondônia, Porto Velho, Rondônia, Brazil; Centro Universitário São Lucas (UniSL), Porto Velho, Rondônia, Brazil; Instituto Nacional de Ciência e Tecnologia de Epidemiologia da Amazônia Ocidental (INCT-EpiAmO), Porto Velho, Rondônia, Brazil; Faculdade Católica de Rondônia (FCR), Porto Velho, Rondônia, Brazil.
| |
Collapse
|
7
|
Teixeira-Cruz JM, Martins-Ferreira J, Monteiro-Machado M, Strauch MA, de Moraes JA, Amaral LS, Valente RC, Melo PA, Quintas LEM. Heparin prevents the cytotoxic activity of Bothrops jararacussu and Apis mellifera venoms in renal cells. Toxicon 2023; 223:107011. [PMID: 36584790 DOI: 10.1016/j.toxicon.2022.107011] [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: 10/03/2022] [Revised: 12/20/2022] [Accepted: 12/26/2022] [Indexed: 12/29/2022]
Abstract
Envenomation by Bothrops snakes and Apis mellifera bee may imply systemic disorders which affect well-perfused organs such as kidneys, a process that can lead to acute renal failure. Nevertheless, there is scarce information regarding a direct renal cell effect and the putative antagonism by antivenoms. Here the cytotoxic effect of B. jararacussu and A. mellifera venoms was evaluated in the renal proximal tubule cell line LLC-PK1, as well as the antagonism of this effect by heparin. B. jararacussu venom showed significant cytotoxicity as assessed by LDH release and MTT reduction, with a sharp decline of the cell number after 180 min (>90% at 50 μg/mL). A. mellifera venom produced a much faster and potent cytotoxic activity, conferring almost no viable cells after 15 min at 25 μg/mL. Phase contrast microscopy revealed that while B. jararacussu venom induced a progressive loss of cell adhesion and detachment, A. mellifera venom promoted a rapid plasma membrane disruption and nuclear condensation suggestive of necrotic cell death. Pre-incubation of both venoms with heparin for 30 min significantly reduced cytotoxicity. Our results demonstrate direct toxicity of B. jararacussu and A. mellifera venoms toward renal cells but with distinct kinetics and cell pattern, suggesting different mechanisms of action. In addition, the antagonistic, cytoprotective effect of heparin ascribes such compound as a promising drug for preventing renal failure from envenomation.
Collapse
Affiliation(s)
- Jhonatha M Teixeira-Cruz
- Programa de Pós-Graduação em Farmacologia e Química Medicinal, Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Jainne Martins-Ferreira
- Programa de Pós-Graduação em Farmacologia e Química Medicinal, Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Marcos Monteiro-Machado
- Programa de Pós-Graduação em Farmacologia e Química Medicinal, Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Marcelo A Strauch
- Programa de Pós-Graduação em Farmacologia e Química Medicinal, Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, Brazil; Instituto Vital Brazil, Niterói, RJ, Brazil
| | - João Alfredo de Moraes
- Programa de Pós-Graduação em Farmacologia e Química Medicinal, Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Luciana S Amaral
- Programa de Pós-Graduação em Farmacologia e Química Medicinal, Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Raphael C Valente
- Núcleo Multidisciplinar de Pesquisa Em Biologia, Universidade Federal do Rio de Janeiro - Campus Duque de Caxias Professor Geraldo Cidade, Rio de Janeiro, Brazil
| | - Paulo A Melo
- Programa de Pós-Graduação em Farmacologia e Química Medicinal, Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Luis Eduardo M Quintas
- Programa de Pós-Graduação em Farmacologia e Química Medicinal, Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, Brazil.
| |
Collapse
|
8
|
Nguyen GTT, O'Brien C, Wouters Y, Seneci L, Gallissà-Calzado A, Campos-Pinto I, Ahmadi S, Laustsen AH, Ljungars A. High-throughput proteomics and in vitro functional characterization of the 26 medically most important elapids and vipers from sub-Saharan Africa. Gigascience 2022; 11:giac121. [PMID: 36509548 PMCID: PMC9744630 DOI: 10.1093/gigascience/giac121] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2022] [Revised: 10/06/2022] [Accepted: 11/14/2022] [Indexed: 12/15/2022] Open
Abstract
Venomous snakes are important parts of the ecosystem, and their behavior and evolution have been shaped by their surrounding environments over the eons. This is reflected in their venoms, which are typically highly adapted for their biological niche, including their diet and defense mechanisms for deterring predators. Sub-Saharan Africa is rich in venomous snake species, of which many are dangerous to humans due to the high toxicity of their venoms and their ability to effectively deliver large amounts of venom into their victims via their bite. In this study, the venoms of 26 of sub-Saharan Africa's medically most relevant elapid and viper species were subjected to parallelized toxicovenomics analysis. The analysis included venom proteomics and in vitro functional characterization of whole venom toxicities, enabling a robust comparison of venom profiles between species. The data presented here corroborate previous studies and provide biochemical details for the clinical manifestations observed in envenomings by the 26 snake species. Moreover, two new venom proteomes (Naja anchietae and Echis leucogaster) are presented here for the first time. Combined, the presented data can help shine light on snake venom evolutionary trends and possibly be used to further improve or develop novel antivenoms.
Collapse
Affiliation(s)
- Giang Thi Tuyet Nguyen
- Department of Biotechnology and Biomedicine, Technical University of Denmark, DK-2800 Kongens Lyngby, Denmark
| | - Carol O'Brien
- Department of Biotechnology and Biomedicine, Technical University of Denmark, DK-2800 Kongens Lyngby, Denmark
| | - Yessica Wouters
- Department of Biotechnology and Biomedicine, Technical University of Denmark, DK-2800 Kongens Lyngby, Denmark
| | - Lorenzo Seneci
- Department of Biotechnology and Biomedicine, Technical University of Denmark, DK-2800 Kongens Lyngby, Denmark
| | - Alex Gallissà-Calzado
- Department of Biotechnology and Biomedicine, Technical University of Denmark, DK-2800 Kongens Lyngby, Denmark
| | - Isabel Campos-Pinto
- Department of Biotechnology and Biomedicine, Technical University of Denmark, DK-2800 Kongens Lyngby, Denmark
| | - Shirin Ahmadi
- Department of Biotechnology and Biomedicine, Technical University of Denmark, DK-2800 Kongens Lyngby, Denmark
| | - Andreas H Laustsen
- Department of Biotechnology and Biomedicine, Technical University of Denmark, DK-2800 Kongens Lyngby, Denmark
| | - Anne Ljungars
- Department of Biotechnology and Biomedicine, Technical University of Denmark, DK-2800 Kongens Lyngby, Denmark
| |
Collapse
|
9
|
Nasri Nasrabadi N, Mohammadpour Dounighi N, Ahmadinejad M, Rabiei H, Tabarzad M, Najafi M, Vatanpour H. Isolation of the Anticoagulant and Procoagulant Fractions of the Venom of Iranian Endemic Echis carinatus. IRANIAN JOURNAL OF PHARMACEUTICAL RESEARCH : IJPR 2022; 21:e127240. [PMID: 36942067 PMCID: PMC10024320 DOI: 10.5812/ijpr-127240] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Revised: 07/13/2022] [Accepted: 07/17/2022] [Indexed: 11/16/2022]
Abstract
Background The venom of Echis carinatus contains both procoagulant and anticoagulant components that can either promote or block the blood coagulation cascade, and some of these components affect platelet function in different ways. Objectives The present study focuses on setting up a procedure for the purification of crude venom and designing appropriate clotting tests in order to characterize the procoagulant and anticoagulant fractions of E. carinatus venom. Methods Chromatographic methods, including gel filtration, ion-exchange chromatography, and reverse-phase high-performance liquid chromatography (HPLC), were applied for purifying these fractions. Coagulant activity testing, prothrombin time (PT), and activated partial thromboplastin time (APTT) were used to determine procoagulant and anticoagulant properties. For measuring molecular weight, 15% SDS-PAGE electrophoresis with a molecular weight standard ranging from 6.5 to 200 kDa was used. Results We obtained five fractions named F1, F2, F3, F4, and F5. The F1 and F2 fractions showed procoagulant activity, and the F5 fraction had anticoagulant activity. The molecular weight of F2.4.2 from fraction F2 and F5.1 from fraction F5 were analyzed by SDS-PAGE electrophoresis under the reducing condition. These factors were identified as a single protein band at the end of purification. The molecular weights of these purified fractions were estimated to be 7.5 kDa and 38 kDa for F5.1(b) and F2.4.2(b), respectively. Conclusions Our findings suggest an efficient and suitable procedure for the identification and purification of the procoagulant and anticoagulant factors of the venom of Iranian E. carinatus using the PT and APTT assays.
Collapse
Affiliation(s)
- Nafiseh Nasri Nasrabadi
- Student Research Commitee, Pharmaceutical Sciences Research Centre, School of Pharmacy, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Nasser Mohammadpour Dounighi
- Department of Venomous Animals and Anti-venom Production, Razi Vaccine and Serum Research Institute, Karaj, Iran
| | - Minoo Ahmadinejad
- Pathology Blood Transfusion Research Center, High Institute for Research and Education in Transfusion Medicine, Tehran, Iran
| | - Hadi Rabiei
- Department of Venomous Animals and Anti-venom Production, Razi Vaccine and Serum Research Institute, Karaj, Iran
| | - Maryam Tabarzad
- Protein Technology Research Center, Shahid Behashti University of Medical Sciences, Tehran, Iran
| | - Mojtaba Najafi
- Gorgan University of Agricultural Sciences and Natural Resources, Gorgan, Iran
| | - Hossein Vatanpour
- Department of Toxicology, School of Pharmacy, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Corresponding Author: Department of Toxicology, School of Pharmacy, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
10
|
von Reumont BM, Anderluh G, Antunes A, Ayvazyan N, Beis D, Caliskan F, Crnković A, Damm M, Dutertre S, Ellgaard L, Gajski G, German H, Halassy B, Hempel BF, Hucho T, Igci N, Ikonomopoulou MP, Karbat I, Klapa MI, Koludarov I, Kool J, Lüddecke T, Ben Mansour R, Vittoria Modica M, Moran Y, Nalbantsoy A, Ibáñez MEP, Panagiotopoulos A, Reuveny E, Céspedes JS, Sombke A, Surm JM, Undheim EAB, Verdes A, Zancolli G. Modern venomics-Current insights, novel methods, and future perspectives in biological and applied animal venom research. Gigascience 2022; 11:giac048. [PMID: 35640874 PMCID: PMC9155608 DOI: 10.1093/gigascience/giac048] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2022] [Revised: 04/10/2022] [Accepted: 04/12/2022] [Indexed: 12/11/2022] Open
Abstract
Venoms have evolved >100 times in all major animal groups, and their components, known as toxins, have been fine-tuned over millions of years into highly effective biochemical weapons. There are many outstanding questions on the evolution of toxin arsenals, such as how venom genes originate, how venom contributes to the fitness of venomous species, and which modifications at the genomic, transcriptomic, and protein level drive their evolution. These questions have received particularly little attention outside of snakes, cone snails, spiders, and scorpions. Venom compounds have further become a source of inspiration for translational research using their diverse bioactivities for various applications. We highlight here recent advances and new strategies in modern venomics and discuss how recent technological innovations and multi-omic methods dramatically improve research on venomous animals. The study of genomes and their modifications through CRISPR and knockdown technologies will increase our understanding of how toxins evolve and which functions they have in the different ontogenetic stages during the development of venomous animals. Mass spectrometry imaging combined with spatial transcriptomics, in situ hybridization techniques, and modern computer tomography gives us further insights into the spatial distribution of toxins in the venom system and the function of the venom apparatus. All these evolutionary and biological insights contribute to more efficiently identify venom compounds, which can then be synthesized or produced in adapted expression systems to test their bioactivity. Finally, we critically discuss recent agrochemical, pharmaceutical, therapeutic, and diagnostic (so-called translational) aspects of venoms from which humans benefit.
Collapse
Affiliation(s)
- Bjoern M von Reumont
- Goethe University Frankfurt, Institute for Cell Biology and Neuroscience, Department for Applied Bioinformatics, 60438 Frankfurt am Main, Germany
- LOEWE Centre for Translational Biodiversity Genomics, Senckenberg Frankfurt, Senckenberganlage 25, 60235 Frankfurt, Germany
- Justus Liebig University Giessen, Institute for Insectbiotechnology, Heinrich Buff Ring 26-32, 35396 Giessen, Germany
| | - Gregor Anderluh
- Department of Molecular Biology and Nanobiotechnology, National Institute of Chemistry, 1000 Ljubljana, Slovenia
| | - Agostinho Antunes
- CIIMAR/CIMAR, Interdisciplinary Centre of Marine and Environmental Research, University of Porto, Terminal de Cruzeiros do Porto de Leixões, Av. General Norton de Matos, s/n, 4450–208 Porto, Portugal
- Department of Biology, Faculty of Sciences, University of Porto, Rua do Campo Alegre, 4169-007 Porto, Portugal
| | - Naira Ayvazyan
- Orbeli Institute of Physiology of NAS RA, Orbeli ave. 22, 0028 Yerevan, Armenia
| | - Dimitris Beis
- Developmental Biology, Centre for Clinical, Experimental Surgery and Translational Research, Biomedical Research Foundation Academy of Athens, Athens 11527, Greece
| | - Figen Caliskan
- Department of Biology, Faculty of Science and Letters, Eskisehir Osmangazi University, TR-26040 Eskisehir, Turkey
| | - Ana Crnković
- Department of Molecular Biology and Nanobiotechnology, National Institute of Chemistry, 1000 Ljubljana, Slovenia
| | - Maik Damm
- Technische Universität Berlin, Department of Chemistry, Straße des 17. Juni 135, 10623 Berlin, Germany
| | | | - Lars Ellgaard
- Department of Biology, University of Copenhagen, DK-2200 Copenhagen, Denmark
| | - Goran Gajski
- Institute for Medical Research and Occupational Health, Mutagenesis Unit, Ksaverska cesta 2, 10000 Zagreb, Croatia
| | - Hannah German
- Amsterdam Institute of Molecular and Life Sciences, Division of BioAnalytical Chemistry, Faculty of Science, Vrije Universiteit Amsterdam, De Boelelaan 1085, 1081HV Amsterdam, The Netherlands
| | - Beata Halassy
- University of Zagreb, Centre for Research and Knowledge Transfer in Biotechnology, Trg Republike Hrvatske 14, 10000 Zagreb, Croatia
| | - Benjamin-Florian Hempel
- BIH Center for Regenerative Therapies BCRT, Charité - Universitätsmedizin Berlin, Augustenburger Platz 1, 13353 Berlin, Germany
| | - Tim Hucho
- Translational Pain Research, Department of Anesthesiology and Intensive Care Medicine, Faculty of Medicine and University Hospital Cologne, University of Cologne, 50931 Cologne, Germany
| | - Nasit Igci
- Nevsehir Haci Bektas Veli University, Faculty of Arts and Sciences, Department of Molecular Biology and Genetics, 50300 Nevsehir, Turkey
| | - Maria P Ikonomopoulou
- Madrid Institute for Advanced Studies in Food, Madrid,E28049, Spain
- The University of Queensland, St Lucia, QLD 4072, Australia
| | - Izhar Karbat
- Department of Biomolecular Sciences, Weizmann Institute of Science, Rehovot 76100, Israel
| | - Maria I Klapa
- Metabolic Engineering and Systems Biology Laboratory, Institute of Chemical Engineering Sciences, Foundation for Research & Technology Hellas (FORTH/ICE-HT), Patras GR-26504, Greece
| | - Ivan Koludarov
- Justus Liebig University Giessen, Institute for Insectbiotechnology, Heinrich Buff Ring 26-32, 35396 Giessen, Germany
| | - Jeroen Kool
- Amsterdam Institute of Molecular and Life Sciences, Division of BioAnalytical Chemistry, Faculty of Science, Vrije Universiteit Amsterdam, De Boelelaan 1085, 1081HV Amsterdam, The Netherlands
| | - Tim Lüddecke
- LOEWE Centre for Translational Biodiversity Genomics, Senckenberg Frankfurt, Senckenberganlage 25, 60235 Frankfurt, Germany
- Department of Bioresources, Fraunhofer Institute for Molecular Biology and Applied Ecology, 35392 Gießen, Germany
| | - Riadh Ben Mansour
- Department of Life Sciences, Faculty of Sciences, Gafsa University, Campus Universitaire Siidi Ahmed Zarrouk, 2112 Gafsa, Tunisia
| | - Maria Vittoria Modica
- Dept. of Biology and Evolution of Marine Organisms (BEOM), Stazione Zoologica Anton Dohrn, Via Po 25c, I-00198 Roma, Italy
| | - Yehu Moran
- Department of Ecology, Evolution and Behavior, Alexander Silberman Institute of Life Sciences, Faculty of Science, The Hebrew University of Jerusalem, Jerusalem 9190401, Israel
| | - Ayse Nalbantsoy
- Department of Bioengineering, Faculty of Engineering, Ege University, 35100 Bornova, Izmir, Turkey
| | - María Eugenia Pachón Ibáñez
- Unit of Infectious Diseases, Microbiology, and Preventive Medicine, Virgen del Rocío University Hospital, Institute of Biomedicine of Seville, 41013 Sevilla, Spain
- CIBER de Enfermedades Infecciosas, Instituto de Salud Carlos III, Madrid, Spain
| | - Alexios Panagiotopoulos
- Metabolic Engineering and Systems Biology Laboratory, Institute of Chemical Engineering Sciences, Foundation for Research & Technology Hellas (FORTH/ICE-HT), Patras GR-26504, Greece
- Animal Biology Division, Department of Biology, University of Patras, Patras, GR-26500, Greece
| | - Eitan Reuveny
- Department of Biomolecular Sciences, Weizmann Institute of Science, Rehovot 76100, Israel
| | - Javier Sánchez Céspedes
- Unit of Infectious Diseases, Microbiology, and Preventive Medicine, Virgen del Rocío University Hospital, Institute of Biomedicine of Seville, 41013 Sevilla, Spain
- CIBER de Enfermedades Infecciosas, Instituto de Salud Carlos III, Madrid, Spain
| | - Andy Sombke
- Department of Evolutionary Biology, University of Vienna, Djerassiplatz 1, 1030 Vienna, Austria
| | - Joachim M Surm
- Department of Ecology, Evolution and Behavior, Alexander Silberman Institute of Life Sciences, Faculty of Science, The Hebrew University of Jerusalem, Jerusalem 9190401, Israel
| | - Eivind A B Undheim
- University of Oslo, Centre for Ecological and Evolutionary Synthesis, Postboks 1066 Blindern 0316 Oslo, Norway
| | - Aida Verdes
- Department of Biodiversity and Evolutionary Biology, Museo Nacional de Ciencias Naturales, José Gutiérrez Abascal 2, 28006 Madrid, Spain
| | - Giulia Zancolli
- Department of Ecology and Evolution, University of Lausanne, 1015 Lausanne, Switzerland
- Swiss Institute of Bioinformatics, 1015 Lausanne, Switzerland
| |
Collapse
|
11
|
Bitiscetin-3, a Novel C-Type Lectin-like Protein Cloned from the Venom Gland of the Viper Bitis arietans, Induces Platelet Agglutination and Inhibits Binding of Von Willebrand Factor to Collagen. Toxins (Basel) 2022; 14:toxins14040236. [PMID: 35448845 PMCID: PMC9024624 DOI: 10.3390/toxins14040236] [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: 03/03/2022] [Revised: 03/18/2022] [Accepted: 03/18/2022] [Indexed: 02/04/2023] Open
Abstract
Bitiscetin-1 (aka bitiscetin) and bitiscetin-2 are C-type lectin-like proteins purified from the venom of Bitis arietans (puff adder). They bind to von Willebrand factor (VWF) and—at least bitiscetin-1—induce platelet agglutination via enhancement of VWF binding to platelet glycoprotein Ib (GPIb). Bitiscetin-1 and -2 bind the VWF A1 and A3 domains, respectively. The A3 domain includes the major site of VWF for binding collagen, explaining why bitiscetin-2 blocks VWF-to-collagen binding. In the present study, sequences for a novel bitiscetin protein—bitiscetin-3—were identified in cDNA constructed from the B. arietans venom gland. The deduced amino acid sequences of bitiscetin-3 subunits α and β share 79 and 80% identity with those of bitiscetin-1, respectively. Expression vectors for bitiscetin-3α and -3β were co-transfected to 293T cells, producing the heterodimer protein recombinant bitiscetin-3 (rBit-3). Functionally, purified rBit-3 (1) induced platelet agglutination involving VWF and GPIb, (2) did not compete with bitiscetin-1 for binding to VWF, (3) blocked VWF-to-collagen binding, and (4) lost its platelet agglutination inducing ability in the presence of an anti-VWF monoclonal antibody that blocked VWF-to-collagen binding. These combined results suggest that bitiscetin-3 binds to the A3 domain, as does bitiscetin-2. Except for a small N-terminal fragment of a single subunit—which differs from that of both bitiscetin-3 subunits—the sequences of bitiscetin-2 have never been determined. Therefore, by identifying and analyzing bitiscetin-3, the present study is the first to present the full-length α- and β-subunit sequences and recombinant expression of a bitiscetin-family toxin that blocks the binding of VWF to collagen.
Collapse
|
12
|
Chérifi F, Laraba-Djebari F. Bioactive Molecules Derived from Snake Venoms with Therapeutic Potential for the Treatment of Thrombo-Cardiovascular Disorders Associated with COVID-19. Protein J 2021; 40:799-841. [PMID: 34499333 PMCID: PMC8427918 DOI: 10.1007/s10930-021-10019-4] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/24/2021] [Indexed: 01/08/2023]
Abstract
As expected, several new variants of Severe Acute Respiratory Syndrome-CoronaVirus-2 (SARS-CoV-2) emerged and have been detected around the world throughout this Coronavirus Disease of 2019 (COVID-19) pandemic. Currently, there is no specific developed drug against COVID-19 and the challenge of developing effective antiviral strategies based on natural agents with different mechanisms of action becomes an urgent need and requires identification of genetic differences among variants. Such data is used to improve therapeutics to combat SARS-CoV-2 variants. Nature is known to offer many biotherapeutics from animal venoms, algae and plant that have been historically used in traditional medicine. Among these bioresources, snake venom displays many bioactivities of interest such as antiviral, antiplatelet, antithrombotic, anti-inflammatory, antimicrobial and antitumoral. COVID-19 is a viral respiratory sickness due to SARS-CoV-2 which induces thrombotic disorders due to cytokine storm, platelet hyperactivation and endothelial dysfunction. This review aims to: (1) present an overview on the infection, the developed thrombo-inflammatory responses and mechanisms of induced thrombosis of COVID-19 compared to other similar pathogenesis; (2) underline the role of natural compounds such as anticoagulant, antiplatelet and thrombolytic agents; (3) investigate the management of coagulopathy related to COVID-19 and provide insight on therapeutic such as venom compounds. We also summarize the updated advances on antiviral proteins and peptides derived from snake venoms that could weaken coagulopathy characterizing COVID-19.
Collapse
Affiliation(s)
- Fatah Chérifi
- USTHB, Faculty of Biological Sciences, Laboratory of Cellular and Molecular Biology, USTHB, BP 32, El-Alia, Bab Ezzouar, Algiers, Algeria
| | - Fatima Laraba-Djebari
- USTHB, Faculty of Biological Sciences, Laboratory of Cellular and Molecular Biology, USTHB, BP 32, El-Alia, Bab Ezzouar, Algiers, Algeria.
| |
Collapse
|
13
|
Bothrops Jararaca Snake Venom Modulates Key Cancer-Related Proteins in Breast Tumor Cell Lines. Toxins (Basel) 2021; 13:toxins13080519. [PMID: 34437390 PMCID: PMC8402457 DOI: 10.3390/toxins13080519] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2021] [Accepted: 07/19/2021] [Indexed: 12/15/2022] Open
Abstract
Cancer is characterized by the development of abnormal cells that divide in an uncontrolled way and may spread into other tissues where they may infiltrate and destroy normal body tissue. Several previous reports have described biochemical anti-tumorigenic properties of crude snake venom or its components, including their capability of inhibiting cell proliferation and promoting cell death. However, to the best of our knowledge, there is no work describing cancer cell proteomic changes following treatment with snake venoms. In this work we describe the quantitative changes in proteomics of MCF7 and MDA-MB-231 breast tumor cell lines following treatment with Bothrops jararaca snake venom, as well as the functional implications of the proteomic changes. Cell lines were treated with sub-toxic doses at either 0.63 μg/mL (low) or 2.5 μg/mL (high) of B. jararaca venom for 24 h, conditions that cause no cell death per se. Proteomics analysis was conducted on a nano-scale liquid chromatography coupled on-line with mass spectrometry (nLC-MS/MS). More than 1000 proteins were identified and evaluated from each cell line treated with either the low or high dose of the snake venom. Protein profiling upon venom treatment showed differential expression of several proteins related to cancer cell metabolism, immune response, and inflammation. Among the identified proteins we highlight histone H3, SNX3, HEL-S-156an, MTCH2, RPS, MCC2, IGF2BP1, and GSTM3. These data suggest that sub-toxic doses of B. jararaca venom have potential to modulate cancer-development related protein targets in cancer cells. This work illustrates a novel biochemical strategy to identify therapeutic targets against cancer cell growth and survival.
Collapse
|
14
|
Lima-Oliveira G, Brennan-Bourdon LM, Varela B, Arredondo ME, Aranda E, Flores S, Ochoa P. Clot activators and anticoagulant additives for blood collection. A critical review on behalf of COLABIOCLI WG-PRE-LATAM. Crit Rev Clin Lab Sci 2020; 58:207-224. [PMID: 33929278 DOI: 10.1080/10408363.2020.1849008] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
In the clinical laboratory, knowledge of and the correct use of clot activators and anticoagulant additives are critical to preserve and maintain samples in optimal conditions prior to analysis. In 2017, the Latin America Confederation of Clinical Biochemistry (COLABIOCLI) commissioned the Latin American Working Group for Preanalytical Phase (WG-PRE-LATAM) to study preanalytical variability and establish guidelines for preanalytical procedures to be applied by clinical laboratories and health care professionals. The aim of this critical review, on behalf of COLABIOCLI WG-PRE-LATAM, is to provide information to understand the mechanisms of the interactions and reactions that occur between blood and clot activators and anticoagulant additives inside evacuated tubes used for laboratory testing. Clot activators - glass, silica, kaolin, bentonite, and diatomaceous earth - work by surface dependent mechanism whereas extrinsic biomolecules - thrombin, snake venoms, ellagic acid, and thromboplastin - start in vitro coagulation when added to blood. Few manufacturers of evacuated tubes state the type and concentration of clot activators used in their products. With respect to anticoagulant additives, sodium citrate and oxalate complex free calcium and ethylenediaminetetraacetic acid chelates calcium. Heparin potentiates antithrombin and hirudin binds to active thrombin, inactivating the thrombin irreversibly. Blood collection tubes have improved continually over the years, from the glass tubes containing clot activators or anticoagulant additives that were prepared by laboratory personnel to the current standardized evacuated systems that permit more precise blood/additive ratios. Each clot activator and anticoagulant additive demonstrates specific functionality, and both manufacturers of tubes and laboratory professional strive to provide suitable interference-free sample matrices for laboratory testing. Both manufacturers of in vitro diagnostic devices and laboratory professionals need to understand all aspects of venous blood sampling so that they do not underestimate the impact of tube additives on laboratory testing.
Collapse
Affiliation(s)
- G Lima-Oliveira
- Latin American Working Group for Preanalytical Phase (WG-PRE-LATAM), Latin America Confederation of Clinical Biochemistry (COLABIOCLI), Montevideo, Uruguay.,Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona, Verona, Italy
| | - L M Brennan-Bourdon
- Latin American Working Group for Preanalytical Phase (WG-PRE-LATAM), Latin America Confederation of Clinical Biochemistry (COLABIOCLI), Montevideo, Uruguay.,Comisión Para la Protección Contra Riesgos Sanitarios del Estado de Jalisco (COPRISJAL), Secretaria de Salud, Guadalajara, México
| | - B Varela
- Latin American Working Group for Preanalytical Phase (WG-PRE-LATAM), Latin America Confederation of Clinical Biochemistry (COLABIOCLI), Montevideo, Uruguay.,Quality Assurance, LAC, Montevideo, Uruguay
| | - M E Arredondo
- Latin American Working Group for Preanalytical Phase (WG-PRE-LATAM), Latin America Confederation of Clinical Biochemistry (COLABIOCLI), Montevideo, Uruguay.,Management Area, Clinical Laboratory, BIONET S.A, Santiago, Chile
| | - E Aranda
- Latin American Working Group for Preanalytical Phase (WG-PRE-LATAM), Latin America Confederation of Clinical Biochemistry (COLABIOCLI), Montevideo, Uruguay.,Laboratory of Thrombosis and Hemostasis, Department of Hematology-Oncology, School of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - S Flores
- Latin American Working Group for Preanalytical Phase (WG-PRE-LATAM), Latin America Confederation of Clinical Biochemistry (COLABIOCLI), Montevideo, Uruguay.,Clinical Laboratory, Universidad Peruana Cayetano Heredia, Lima, Perú
| | - P Ochoa
- Latin American Working Group for Preanalytical Phase (WG-PRE-LATAM), Latin America Confederation of Clinical Biochemistry (COLABIOCLI), Montevideo, Uruguay.,Facultad de Medicina, Universidad Católica de Cuenca, Cuenca, Ecuador
| |
Collapse
|
15
|
Herzig V, Cristofori-Armstrong B, Israel MR, Nixon SA, Vetter I, King GF. Animal toxins - Nature's evolutionary-refined toolkit for basic research and drug discovery. Biochem Pharmacol 2020; 181:114096. [PMID: 32535105 PMCID: PMC7290223 DOI: 10.1016/j.bcp.2020.114096] [Citation(s) in RCA: 112] [Impact Index Per Article: 22.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2020] [Revised: 06/06/2020] [Accepted: 06/09/2020] [Indexed: 12/27/2022]
Abstract
Venomous animals have evolved toxins that interfere with specific components of their victim's core physiological systems, thereby causing biological dysfunction that aids in prey capture, defense against predators, or other roles such as intraspecific competition. Many animal lineages evolved venom systems independently, highlighting the success of this strategy. Over the course of evolution, toxins with exceptional specificity and high potency for their intended molecular targets have prevailed, making venoms an invaluable and almost inexhaustible source of bioactive molecules, some of which have found use as pharmacological tools, human therapeutics, and bioinsecticides. Current biomedically-focused research on venoms is directed towards their use in delineating the physiological role of toxin molecular targets such as ion channels and receptors, studying or treating human diseases, targeting vectors of human diseases, and treating microbial and parasitic infections. We provide examples of each of these areas of venom research, highlighting the potential that venom molecules hold for basic research and drug development.
Collapse
Affiliation(s)
- Volker Herzig
- School of Science & Engineering, University of the Sunshine Coast, Sippy Downs, QLD, Australia; Institute for Molecular Bioscience, The University of Queensland, St. Lucia, QLD, Australia.
| | | | - Mathilde R Israel
- Institute for Molecular Bioscience, The University of Queensland, St. Lucia, QLD, Australia
| | - Samantha A Nixon
- Institute for Molecular Bioscience, The University of Queensland, St. Lucia, QLD, Australia
| | - Irina Vetter
- Institute for Molecular Bioscience, The University of Queensland, St. Lucia, QLD, Australia
| | - Glenn F King
- Institute for Molecular Bioscience, The University of Queensland, St. Lucia, QLD, Australia.
| |
Collapse
|
16
|
An Y, Bai Z, Xu X, Guo X, Romeiro FG, Philips CA, Li Y, Wu Y, Qi X. No Benefit of Hemostatic Drugs on Acute Upper Gastrointestinal Bleeding in Cirrhosis. BIOMED RESEARCH INTERNATIONAL 2020; 2020:4097170. [PMID: 32685481 PMCID: PMC7336197 DOI: 10.1155/2020/4097170] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/28/2020] [Revised: 05/25/2020] [Accepted: 06/01/2020] [Indexed: 02/07/2023]
Abstract
BACKGROUND AND AIMS Acute upper gastrointestinal bleeding (AUGIB) is one of the most life-threatening emergency conditions. Hemostatic drugs are often prescribed to control AUGIB in clinical practice but have not been recommended by major guidelines and consensus. The aim of this study was to investigate the therapeutic effect of hemostatic drugs on AUGIB in cirrhosis. METHODS All cirrhotic patients with AUGIB who were admitted to our hospital from January 2010 to June 2014 were retrospectively included. Patients were divided into hemostatic drugs and no hemostatic drug groups. A 1 : 1 propensity score matching (PSM) analysis was performed by adjusting age, gender, etiology of liver disease, Child-Pugh score, MELD score, hematemesis, red blood cell transfusion, vasoactive drugs, antibiotics, proton pump inhibitors, and endoscopic variceal therapy. Primary outcomes included 5-day rebleeding and in-hospital mortality. RESULTS Overall, 982 cirrhotic patients with AUGIB were included (870 in hemostatic drugs group and 112 in no hemostatic drug group). In overall analyses, hemostatic drugs group had a significantly higher 5-day rebleeding rate (18.10% versus 5.40%, P = 0.001) than no hemostatic drug group; in-hospital mortality was not significantly different between them (7.10% versus 4.50%, P = 0.293). In PSM analyses, 172 patients were included (86 patients in each group). Hemostatic drugs group still had a significantly higher 5-day rebleeding rate (15.10% versus 5.80%, P = 0.046); in-hospital mortality remained not significantly different (7.00% versus 3.50%, P = 0.304) between them. Statistical results remained in PSM analyses according to the type of hemostatic drugs. CONCLUSIONS The use of hemostatic drugs did not improve the in-hospital outcomes of cirrhotic patients with AUGIB.
Collapse
Affiliation(s)
- Yang An
- Department of Gastroenterology, General Hospital of Northern Theater Command (formerly General Hospital of Shenyang Military Area), Shenyang 110840, China
- Postgraduate College, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Zhaohui Bai
- Department of Gastroenterology, General Hospital of Northern Theater Command (formerly General Hospital of Shenyang Military Area), Shenyang 110840, China
- Postgraduate College, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Xiangbo Xu
- Department of Gastroenterology, General Hospital of Northern Theater Command (formerly General Hospital of Shenyang Military Area), Shenyang 110840, China
- Postgraduate College, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Xiaozhong Guo
- Department of Gastroenterology, General Hospital of Northern Theater Command (formerly General Hospital of Shenyang Military Area), Shenyang 110840, China
| | - Fernando Gomes Romeiro
- Department of Internal Medicine, Botucatu Medical School, UNESP-Univ Estadual Paulista. Av. Prof. Mário Rubens Guimarães Montenegro, s/n Distrito de Rubião Jr, Botucatu, Brazil
| | - Cyriac Abby Philips
- The Liver Unit and Monarch Liver Lab, Cochin Gastroenterology Group, Ernakulam Medical Center, Kochi, 682028 Kerala, India
| | - Yingying Li
- Department of Gastroenterology, The First People's Hospital of Huainan, Huainan 232007, China
| | - Yanyan Wu
- Department of Gastroenterology, General Hospital of Northern Theater Command (formerly General Hospital of Shenyang Military Area), Shenyang 110840, China
- Postgraduate College, Jinzhou Medical University, Jinzhou 121001, China
| | - Xingshun Qi
- Department of Gastroenterology, General Hospital of Northern Theater Command (formerly General Hospital of Shenyang Military Area), Shenyang 110840, China
| |
Collapse
|
17
|
Vivas-Ruiz DE, Sandoval GA, Gonzalez-Kozlova E, Zarria-Romero J, Lazo F, Rodríguez E, Magalhães HPB, Chávez-Olortegui C, Oliveira LS, Alvarenga VG, Urra FA, Toledo J, Yarlequé A, Eble JA, Sanchez EF. Fibrinogen-clotting enzyme, pictobin, from Bothrops pictus snake venom. Structural and functional characterization. Int J Biol Macromol 2020; 153:779-795. [PMID: 32169454 DOI: 10.1016/j.ijbiomac.2020.03.055] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2019] [Revised: 03/09/2020] [Accepted: 03/09/2020] [Indexed: 12/14/2022]
Abstract
A thrombin-like enzyme, pictobin, was purified from Bothrops pictus snake venom. It is a 41-kDa monomeric glycoprotein as showed by mass spectrometry and contains approx. 45% carbohydrate by mass which could be removed with N-glycosidase. Pictobin coagulates plasma and fibrinogen, releasing fibrinopeptide A and induces the formation of a friable/porous fibrin network as visualized by SEM. The enzyme promoted platelet aggregation in human PRP and defibrination in mouse model and showed catalytic activity on chromogenic substrates S-2266, S-2366, S-2160 and S-2238. Pictobin interacts with the plasma inhibitor α2-macroglobulin, which blocks its interaction with fibrinogen but not with the small substrate BApNA. Heparin does not affect its enzymatic activity. Pictobin cross reacted with polyvalent bothropic antivenom, and its deglycosylated form reduced its catalytic action and antivenom reaction. In breast and lung cancer cells, pictobin inhibits the fibronectin-stimulated migration. Moreover, it produces strong NADH oxidation, mitochondrial depolarization, ATP decrease and fragmentation of mitochondrial network. These results suggest by first time that a snake venom serinprotease produces mitochondrial dysfunction by affecting mitochondrial dynamics and bioenergetics. Structural model of pictobin reveals a conserved chymotrypsin fold β/β hydrolase. These data indicate that pictobin has therapeutic potential in the treatment of cardiovascular disorders and metastatic disease.
Collapse
Affiliation(s)
- Dan E Vivas-Ruiz
- Laboratorio de Biología Molecular, Facultad de Ciencias Biológicas, Universidad Nacional Mayor de San Marcos, Lima, Perú, Av. Venezuela Cdra 34 S/N, Ciudad Universitaria, Lima 01, Lima, Peru.
| | - Gustavo A Sandoval
- Laboratorio de Biología Molecular, Facultad de Ciencias Biológicas, Universidad Nacional Mayor de San Marcos, Lima, Perú, Av. Venezuela Cdra 34 S/N, Ciudad Universitaria, Lima 01, Lima, Peru
| | - Edgar Gonzalez-Kozlova
- Department of Genetics and Genomic Sciences, Icahn School for Data Science and Genomic Technology, New York, NYC, USA
| | - Jacquelyne Zarria-Romero
- Laboratorio de Reproducción y Biología del Desarrollo, Facultad de Ciencias Biológicas, Universidad Nacional Mayor de San Marcos, Lima, Perú, Av. Venezuela ra 34 S/N, Ciudad Universitaria, Lima 01, Lima, Peru
| | - Fanny Lazo
- Laboratorio de Biología Molecular, Facultad de Ciencias Biológicas, Universidad Nacional Mayor de San Marcos, Lima, Perú, Av. Venezuela Cdra 34 S/N, Ciudad Universitaria, Lima 01, Lima, Peru
| | - Edith Rodríguez
- Laboratorio de Biología Molecular, Facultad de Ciencias Biológicas, Universidad Nacional Mayor de San Marcos, Lima, Perú, Av. Venezuela Cdra 34 S/N, Ciudad Universitaria, Lima 01, Lima, Peru
| | - Henrique P B Magalhães
- Departamento de Análises Clínicas e Toxicológicas, Faculdade de Farmácia, Universidade Federal de Minas Gerais, Belo Horizonte 31270-901, Minas Gerais, Brazil
| | - Carlos Chávez-Olortegui
- Departamento de Bioquímica-Inmunología, Instituto de Ciências Biologicas, Universidade Federal de Minas Gerais, Belo Horizonte 31270-901, Minas Gerais, Brazil
| | - Luciana S Oliveira
- Research and Development Center, Ezequiel Dias Foundation, 30510-010 Belo Horizonte, MG, Brazil
| | - Valeria G Alvarenga
- Research and Development Center, Ezequiel Dias Foundation, 30510-010 Belo Horizonte, MG, Brazil
| | - Félix A Urra
- Programa de Farmacología Clínica y Molecular, Instituto de Ciencias Biomédicas (ICBM), Facultad de Medicina, Universidad de Chile, Independencia 1027, Casilla 7, Santiago 7800003, Chile
| | - Jorge Toledo
- Instituto de Neurociencia Biomédica, Facultad de Medicina, Universidad de Chile, Independencia 1027, Santiago 8380453, Chile; Facultad de Ciencias de la Salud, Universidad San Sebastián, Lota 2465, Providencia, Santiago 7510157, Chile
| | - Armando Yarlequé
- Laboratorio de Biología Molecular, Facultad de Ciencias Biológicas, Universidad Nacional Mayor de San Marcos, Lima, Perú, Av. Venezuela Cdra 34 S/N, Ciudad Universitaria, Lima 01, Lima, Peru
| | - Johannes A Eble
- Institute for Physiological Chemistry and Pathobiochemistry, University of Münster, 48149 Münster, Germany
| | - Eladio F Sanchez
- Research and Development Center, Ezequiel Dias Foundation, 30510-010 Belo Horizonte, MG, Brazil
| |
Collapse
|
18
|
Liu Y, Zhang XH, Yu Y, Chen HX, Zhou YL, Zhang XX. Snake venom characteristic peptides: novel fingerprints for species identification by sheathless capillary electrophoresis-electrospray ionization-mass spectrometry. Analyst 2020; 145:5027-5031. [PMID: 32525167 DOI: 10.1039/d0an00461h] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Snake venom is a complex mixture mainly consisting of proteins and peptides which varies with different species. These variations lead to different toxic mechanisms and different anti-venom serums for treatment and the determination of their use as drugs. Hence, it is important to develop a sensitive and reliable method to identify the species of snakes from venoms. Herein, we present a novel strategy based on the sheathless capillary electrophoresis-electrospray ionization-mass spectrometry (CESI-MS) system to characterize snake venom proteins. Through the determination of peptides, we found the characteristic peptides of 8 different snakes with high sensitivity (1 μg mL-1) and high selectivity, which provided a reliable method for the species identification and purity detection of snake venom samples.
Collapse
Affiliation(s)
- Ying Liu
- Beijing National Laboratory for Molecular Sciences (BNLMS), Key Laboratory of Bioorganic Chemistry and Molecular Engineering of Ministry of Education, College of Chemistry and Molecular Engineering, Peking University, Beijing 100871, China.
| | | | | | | | | | | |
Collapse
|
19
|
Kini RM. Toxinology provides multidirectional and multidimensional opportunities: A personal perspective. Toxicon X 2020; 6:100039. [PMID: 32550594 PMCID: PMC7285919 DOI: 10.1016/j.toxcx.2020.100039] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2020] [Revised: 04/28/2020] [Accepted: 05/05/2020] [Indexed: 01/16/2023] Open
Abstract
In nature, toxins have evolved as weapons to capture and subdue the prey or to counter predators or competitors. When they are inadvertently injected into humans, they cause symptoms ranging from mild discomfort to debilitation and death. Toxinology is the science of studying venoms and toxins that are produced by a wide variety of organisms. In the past, the structure, function and mechanisms of most abundant and/or most toxic components were characterized to understand and to develop strategies to neutralize their toxicity. With recent technical advances, we are able to evaluate and determine the toxin profiles using transcriptomes of venom glands and proteomes of tiny amounts of venom. Enormous amounts of data from these studies have opened tremendous opportunities in many directions of basic and applied research. The lower costs for profiling venoms will further fuel the expansion of toxin database, which in turn will provide greater exciting and bright opportunities in toxin research.
Collapse
Affiliation(s)
- R. Manjunatha Kini
- Protein Science Laboratory, Department of Biological Sciences, Faculty of Science and Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| |
Collapse
|
20
|
Park EJ, Choi S, Kim HH, Jung YS. Novel Treatment Strategy for Patients with Venom-Induced Consumptive Coagulopathy from a Pit Viper Bite. Toxins (Basel) 2020; 12:toxins12050295. [PMID: 32380672 PMCID: PMC7290867 DOI: 10.3390/toxins12050295] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2020] [Revised: 05/01/2020] [Accepted: 05/02/2020] [Indexed: 12/17/2022] Open
Abstract
Pit viper venom commonly causes venom-induced consumptive coagulopathy (VICC), which can be complicated by life-threatening hemorrhage. VICC has a complex pathophysiology affecting multiple steps of the coagulation pathway. Early detection of VICC is challenging because conventional blood tests such as prothrombin time (PT) and activated partial thromboplastin time (aPTT) are unreliable for early-stage monitoring of VICC progress. As the effects on the coagulation cascade may differ, even in the same species, the traditional coagulation pathways cannot fully explain the mechanisms involved in VICC or may be too slow to have any clinical utility. Antivenom should be promptly administered to neutralize the lethal toxins, although its efficacy remains controversial. Transfusion, including fresh frozen plasma, cryoprecipitate, and specific clotting factors, has also been performed in patients with bleeding. The effectiveness of viscoelastic monitoring in the treatment of VICC remains poorly understood. The development of VICC can be clarified using thromboelastography (TEG), which shows the procoagulant and anticoagulant effects of snake venom. Therefore, we believe that TEG may be able to be used to guide hemostatic resuscitation in victims of VICC. Here, we aim to discuss the advantages of TEG by comparing it with traditional coagulation tests and propose potential treatment options for VICC.
Collapse
|
21
|
An Appraisal of Antidotes' Effectiveness: Evidence of the Use of Phyto-Antidotes and Biotechnological Advancements. Molecules 2020; 25:molecules25071516. [PMID: 32225103 PMCID: PMC7181008 DOI: 10.3390/molecules25071516] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2019] [Revised: 02/10/2020] [Accepted: 02/12/2020] [Indexed: 12/20/2022] Open
Abstract
Poisoning is the greatest source of avoidable death in the world and can result from industrial exhausts, incessant bush burning, drug overdose, accidental toxication or snake envenomation. Since the advent of Albert Calmette’s cobra venom antidote, efforts have been geared towards antidotes development for various poisons to date. While there are resources and facilities to tackle poisoning in urban areas, rural areas and developing countries are challenged with poisoning management due to either the absence of or inadequate facilities and this has paved the way for phyto-antidotes, some of which have been scientifically validated. This review presents the scope of antidotes’ effectiveness in different experimental models and biotechnological advancements in antidote research for future applications. While pockets of evidence of the effectiveness of antidotes exist in vitro and in vivo with ample biotechnological developments, the utilization of analytic assays on existing and newly developed antidotes that have surpassed the proof of concept stage, as well as the inclusion of antidote’s short and long-term risk assessment report, will help in providing the required scientific evidence(s) prior to regulatory authorities’ approval.
Collapse
|
22
|
Gremski LH, da Justa HC, da Silva TP, Polli NLC, Antunes BC, Minozzo JC, Wille ACM, Senff-Ribeiro A, Arni RK, Veiga SS. Forty Years of the Description of Brown Spider Venom Phospholipases-D. Toxins (Basel) 2020; 12:toxins12030164. [PMID: 32155765 PMCID: PMC7150852 DOI: 10.3390/toxins12030164] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2020] [Revised: 02/28/2020] [Accepted: 03/02/2020] [Indexed: 01/24/2023] Open
Abstract
Spiders of the genus Loxosceles, popularly known as Brown spiders, are considered a serious public health issue, especially in regions of hot or temperate climates, such as parts of North and South America. Although the venoms of these arachnids are complex in molecular composition, often containing proteins with distinct biochemical characteristics, the literature has primarily described a family of toxins, the Phospholipases-D (PLDs), which are highly conserved in all Loxosceles species. PLDs trigger most of the major clinical symptoms of loxoscelism i.e., dermonecrosis, thrombocytopenia, hemolysis, and acute renal failure. The key role played by PLDs in the symptomatology of loxoscelism was first described 40 years ago, when researches purified a hemolytic toxin that cleaved sphingomyelin and generated choline, and was referred to as a Sphingomyelinase-D, which was subsequently changed to Phospholipase-D when it was demonstrated that the enzyme also cleaved other cellular phospholipids. In this review, we present the information gleaned over the last 40 years about PLDs from Loxosceles venoms especially with regard to the production and characterization of recombinant isoforms. The history of obtaining these toxins is discussed, as well as their molecular organization and mechanisms of interaction with their substrates. We will address cellular biology aspects of these toxins and how they can be used in the development of drugs to address inflammatory processes and loxoscelism. Present and future aspects of loxoscelism diagnosis will be discussed, as well as their biotechnological applications and actions expected for the future in this field.
Collapse
Affiliation(s)
- Luiza Helena Gremski
- Departamento de Biologia Celular, Universidade Federal do Paraná (UFPR), Curitiba 81531-980, PR, Brazil; (L.H.G.); (H.C.d.J.); (T.P.d.S.); (N.L.C.P.); (B.C.A.); (A.S.-R.)
| | - Hanna Câmara da Justa
- Departamento de Biologia Celular, Universidade Federal do Paraná (UFPR), Curitiba 81531-980, PR, Brazil; (L.H.G.); (H.C.d.J.); (T.P.d.S.); (N.L.C.P.); (B.C.A.); (A.S.-R.)
| | - Thaís Pereira da Silva
- Departamento de Biologia Celular, Universidade Federal do Paraná (UFPR), Curitiba 81531-980, PR, Brazil; (L.H.G.); (H.C.d.J.); (T.P.d.S.); (N.L.C.P.); (B.C.A.); (A.S.-R.)
| | - Nayanne Louise Costacurta Polli
- Departamento de Biologia Celular, Universidade Federal do Paraná (UFPR), Curitiba 81531-980, PR, Brazil; (L.H.G.); (H.C.d.J.); (T.P.d.S.); (N.L.C.P.); (B.C.A.); (A.S.-R.)
| | - Bruno César Antunes
- Departamento de Biologia Celular, Universidade Federal do Paraná (UFPR), Curitiba 81531-980, PR, Brazil; (L.H.G.); (H.C.d.J.); (T.P.d.S.); (N.L.C.P.); (B.C.A.); (A.S.-R.)
- Centro de Produção e Pesquisa de Imunobiológicos (CPPI), Piraquara 83302-200, PR, Brazil;
| | - João Carlos Minozzo
- Centro de Produção e Pesquisa de Imunobiológicos (CPPI), Piraquara 83302-200, PR, Brazil;
| | - Ana Carolina Martins Wille
- Departamento de Biologia Estrutural, Molecular e Genética, Universidade Estadual de Ponta Grossa, Ponta Grossa 84030-900, PR, Brazil;
| | - Andrea Senff-Ribeiro
- Departamento de Biologia Celular, Universidade Federal do Paraná (UFPR), Curitiba 81531-980, PR, Brazil; (L.H.G.); (H.C.d.J.); (T.P.d.S.); (N.L.C.P.); (B.C.A.); (A.S.-R.)
| | - Raghuvir Krishnaswamy Arni
- Centro Multiusuário de Inovação Biomolecular, Departamento de Física, Universidade Estadual Paulista (UNESP), São José do Rio Preto 15054-000, SP, Brazil;
| | - Silvio Sanches Veiga
- Departamento de Biologia Celular, Universidade Federal do Paraná (UFPR), Curitiba 81531-980, PR, Brazil; (L.H.G.); (H.C.d.J.); (T.P.d.S.); (N.L.C.P.); (B.C.A.); (A.S.-R.)
- Correspondence: ; Tel.: +55-(41)-3361-1776
| |
Collapse
|
23
|
Yakubu A, Musa Y, Maiyaki AS, Tambuwal SH. Hemoperitoneum complicating venomous snakebite: A case report. SAHEL MEDICAL JOURNAL 2020. [DOI: 10.4103/smj.smj_62_19] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
|
24
|
Das S, Patel B. Marine resources and animals in modern biotechnology. Anim Biotechnol 2020. [DOI: 10.1016/b978-0-12-811710-1.00027-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
|
25
|
Oliveira LS, Estevão-Costa MI, Alvarenga VG, Vivas-Ruiz DE, Yarleque A, Lima AM, Cavaco A, Eble JA, Sanchez EF. Atroxlysin-III, A Metalloproteinase from the Venom of the Peruvian Pit Viper Snake Bothrops atrox (Jergón) Induces Glycoprotein VI Shedding and Impairs Platelet Function. Molecules 2019; 24:molecules24193489. [PMID: 31561469 PMCID: PMC6803841 DOI: 10.3390/molecules24193489] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2019] [Revised: 09/23/2019] [Accepted: 09/24/2019] [Indexed: 01/28/2023] Open
Abstract
Atroxlysin-III (Atr-III) was purified from the venom of Bothrops atrox. This 56-kDa protein bears N-linked glycoconjugates and is a P-III hemorrhagic metalloproteinase. Its cDNA-deduced amino acid sequence reveals a multidomain structure including a proprotein, a metalloproteinase, a disintegrin-like and a cysteine-rich domain. Its identity with bothropasin and jararhagin from Bothrops jararaca is 97% and 95%, respectively. Its enzymatic activity is metal ion-dependent. The divalent cations, Mg2+ and Ca2+, enhance its activity, whereas excess Zn2+ inhibits it. Chemical modification of the Zn2+-complexing histidine residues within the active site by using diethylpyrocarbonate (DEPC) inactivates it. Atr-III degrades plasma fibronectin, type I-collagen, and mainly the α-chains of fibrinogen and fibrin. The von Willebrand factor (vWF) A1-domain, which harbors the binding site for GPIb, is not hydrolyzed. Platelets interact with collagen via receptors for collagen, glycoprotein VI (GPVI), and α2β1 integrin. Neither the α2β1 integrin nor its collagen-binding A-domain is fragmented by Atr-III. In contrast, Atr-III cleaves glycoprotein VI (GPVI) into a soluble ~55-kDa fragment (sGPVI). Thereby, it inhibits aggregation of platelets which had been stimulated by convulxin, a GPVI agonist. Selectively, Atr-III targets GPVI antagonistically and thus contributes to the antithrombotic effect of envenomation by Bothrops atrox.
Collapse
Affiliation(s)
- Luciana S Oliveira
- Research and Development Center, Ezequiel Dias Foundation, 30510-010 Belo Horizonte, MG, Brazil.
| | - Maria Inácia Estevão-Costa
- Institute of Physiological Chemistry and Pathobiochemistry, University of Münster, 48149 Münster, Germany.
| | - Valéria G Alvarenga
- Research and Development Center, Ezequiel Dias Foundation, 30510-010 Belo Horizonte, MG, Brazil.
| | - Dan E Vivas-Ruiz
- Laboratorio de Biología Molecular-Facultad de Ciencias Biológicas, Universidad Nacional Mayor de San Marcos, Av. Venezuela Cdra 34 S/N, Ciudad Universitaria, Lima 01, Lima 14-0576, Peru.
| | - Armando Yarleque
- Laboratorio de Biología Molecular-Facultad de Ciencias Biológicas, Universidad Nacional Mayor de San Marcos, Av. Venezuela Cdra 34 S/N, Ciudad Universitaria, Lima 01, Lima 14-0576, Peru.
| | - Augusto Martins Lima
- Laboratory of Hemodynamics and Cardiovascular Technology, École Polytechnique Fédérale de Lausanne, 1015 Lausanne, Switzerland.
| | - Ana Cavaco
- Institute of Physiological Chemistry and Pathobiochemistry, University of Münster, 48149 Münster, Germany.
| | - Johannes A Eble
- Institute of Physiological Chemistry and Pathobiochemistry, University of Münster, 48149 Münster, Germany.
| | - Eladio F Sanchez
- Research and Development Center, Ezequiel Dias Foundation, 30510-010 Belo Horizonte, MG, Brazil.
| |
Collapse
|
26
|
Snake Venoms in Drug Discovery: Valuable Therapeutic Tools for Life Saving. Toxins (Basel) 2019; 11:toxins11100564. [PMID: 31557973 PMCID: PMC6832721 DOI: 10.3390/toxins11100564] [Citation(s) in RCA: 101] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2019] [Revised: 09/20/2019] [Accepted: 09/22/2019] [Indexed: 12/16/2022] Open
Abstract
Animal venoms are used as defense mechanisms or to immobilize and digest prey. In fact, venoms are complex mixtures of enzymatic and non-enzymatic components with specific pathophysiological functions. Peptide toxins isolated from animal venoms target mainly ion channels, membrane receptors and components of the hemostatic system with high selectivity and affinity. The present review shows an up-to-date survey on the pharmacology of snake-venom bioactive components and evaluates their therapeutic perspectives against a wide range of pathophysiological conditions. Snake venoms have also been used as medical tools for thousands of years especially in tradition Chinese medicine. Consequently, snake venoms can be considered as mini-drug libraries in which each drug is pharmacologically active. However, less than 0.01% of these toxins have been identified and characterized. For instance, Captopril® (Enalapril), Integrilin® (Eptifibatide) and Aggrastat® (Tirofiban) are drugs based on snake venoms, which have been approved by the FDA. In addition to these approved drugs, many other snake venom components are now involved in preclinical or clinical trials for a variety of therapeutic applications. These examples show that snake venoms can be a valuable source of new principle components in drug discovery.
Collapse
|
27
|
Sachetto A, Mackman N. Modulation of the mammalian coagulation system by venoms and other proteins from snakes, arthropods, nematodes and insects. Thromb Res 2019; 178:145-154. [DOI: 10.1016/j.thromres.2019.04.019] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2019] [Revised: 04/04/2019] [Accepted: 04/15/2019] [Indexed: 12/22/2022]
|
28
|
Ozverel CS, Damm M, Hempel BF, Göçmen B, Sroka R, Süssmuth RD, Nalbantsoy A. Investigating the cytotoxic effects of the venom proteome of two species of the Viperidae family (Cerastes cerastes and Cryptelytrops purpureomaculatus) from various habitats. Comp Biochem Physiol C Toxicol Pharmacol 2019; 220:20-30. [PMID: 30825636 DOI: 10.1016/j.cbpc.2019.02.013] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/23/2018] [Revised: 02/20/2019] [Accepted: 02/25/2019] [Indexed: 12/20/2022]
Abstract
Animal secretions are of great interest in terms of drug development due to their complex protein and peptide composition. Especially, in the field of therapeutic medications such as anti-cancer drugs snake venoms receive attention. In this study, we address two Viperidae species from various habitats with a particular focus on the cytotoxic potential along with the decomplexation of the venom proteome: the horned desert viper (Cerastes cerastes), native to desert regions of North Africa and the mangrove pit viper (Cryptelytrops purpureomaculatus), found in coastal forests of Southeast Asia. Initial cytotoxic screenings of the crude venoms revealed diverse activity, with the highest effect against SHSY5Y human glioblastoma carcinoma cells compared to other cancerous and non-cancerous cell lines. In-depth cytotoxicity studies of SHSY5Y cells with purified venom fractions revealed heterodimeric disintegrins from C. cerastes venom, which exerted a high cytotoxic activity with IC50 values from 0.11 to 0.58 μM and a disintegrin-like effect on SHSY5Y morphology was observed due to cell detachment. Furthermore, two polyproline BPP-related peptides, one PLA2 and a peptide-rich fraction were determined for C. purpureomaculatus with moderate IC50 values between 3 and 51 μM. Additionally, the decryption of the venom proteomes by snake venomic mass spectrometry and comparison of the same species from different habitats revealed slight differences in the composition.
Collapse
Affiliation(s)
- Cenk Serhan Ozverel
- Department of Biology, Faculty of Science, Ege University, Bornova, 35100 Izmir, Turkey
| | - Maik Damm
- Technische Universität Berlin, Institut für Chemie, Strasse des 17. Juni 124, 10623 Berlin, Germany
| | - Benjamin-Florian Hempel
- Technische Universität Berlin, Institut für Chemie, Strasse des 17. Juni 124, 10623 Berlin, Germany.
| | - Bayram Göçmen
- Zoology Section, Department of Biology, Faculty of Science, Ege University, Bornova, 35100 Izmir, Turkey
| | - Robert Sroka
- Technische Universität Berlin, Institut für Chemie, Strasse des 17. Juni 124, 10623 Berlin, Germany
| | - Roderich D Süssmuth
- Technische Universität Berlin, Institut für Chemie, Strasse des 17. Juni 124, 10623 Berlin, Germany
| | - Ayse Nalbantsoy
- Department of Bioengineering, Faculty of Engineering, Ege University, Bornova, 35100 Izmir, Turkey.
| |
Collapse
|
29
|
Dobson JS, Zdenek CN, Hay C, Violette A, Fourmy R, Cochran C, Fry BG. Varanid Lizard Venoms Disrupt the Clotting Ability of Human Fibrinogen through Destructive Cleavage. Toxins (Basel) 2019; 11:E255. [PMID: 31067768 PMCID: PMC6563220 DOI: 10.3390/toxins11050255] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2019] [Revised: 04/30/2019] [Accepted: 05/01/2019] [Indexed: 02/06/2023] Open
Abstract
The functional activities of Anguimorpha lizard venoms have received less attention compared to serpent lineages. Bite victims of varanid lizards often report persistent bleeding exceeding that expected for the mechanical damage of the bite. Research to date has identified the blockage of platelet aggregation as one bleeding-inducing activity, and destructive cleavage of fibrinogen as another. However, the ability of the venoms to prevent clot formation has not been directly investigated. Using a thromboelastograph (TEG5000), clot strength was measured after incubating human fibrinogen with Heloderma and Varanus lizard venoms. Clot strengths were found to be highly variable, with the most potent effects produced by incubation with Varanus venoms from the Odatria and Euprepriosaurus clades. The most fibrinogenolytically active venoms belonged to arboreal species and therefore prey escape potential is likely a strong evolutionary selection pressure. The results are also consistent with reports of profusive bleeding from bites from other notably fibrinogenolytic species, such as V. giganteus. Our results provide evidence in favour of the predatory role of venom in varanid lizards, thus shedding light on the evolution of venom in reptiles and revealing potential new sources of bioactive molecules useful as lead compounds in drug design and development.
Collapse
Affiliation(s)
- James S Dobson
- Venom Evolution Lab, School of Biological Sciences, University of Queensland, St Lucia, QLD 4072, Australia.
| | - Christina N Zdenek
- Venom Evolution Lab, School of Biological Sciences, University of Queensland, St Lucia, QLD 4072, Australia.
| | - Chris Hay
- Venom Evolution Lab, School of Biological Sciences, University of Queensland, St Lucia, QLD 4072, Australia.
| | - Aude Violette
- Alphabiotoxine Laboratory sprl, Barberie 15, 7911 Montroeul-au-bois, Belgium.
| | - Rudy Fourmy
- Alphabiotoxine Laboratory sprl, Barberie 15, 7911 Montroeul-au-bois, Belgium.
| | - Chip Cochran
- Department of Earth and Biological Sciences, Loma Linda University, Loma Linda, CA 92350, USA.
| | - Bryan G Fry
- Venom Evolution Lab, School of Biological Sciences, University of Queensland, St Lucia, QLD 4072, Australia.
| |
Collapse
|
30
|
Kunalan S, Othman I, Syed Hassan S, Hodgson WC. Proteomic Characterization of Two Medically Important Malaysian Snake Venoms, Calloselasma rhodostoma (Malayan Pit Viper) and Ophiophagus hannah (King Cobra). Toxins (Basel) 2018; 10:toxins10110434. [PMID: 30373186 PMCID: PMC6266455 DOI: 10.3390/toxins10110434] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2018] [Revised: 10/23/2018] [Accepted: 10/23/2018] [Indexed: 02/07/2023] Open
Abstract
Calloselasma rhodostoma (CR) and Ophiophagus hannah (OH) are two medically important snakes found in Malaysia. While some studies have described the biological properties of these venoms, feeding and environmental conditions also influence the concentration and distribution of snake venom toxins, resulting in variations in venom composition. Therefore, a combined proteomic approach using shotgun and gel filtration chromatography, analyzed by tandem mass spectrometry, was used to examine the composition of venoms from these Malaysian snakes. The analysis revealed 114 proteins (15 toxin families) and 176 proteins (20 toxin families) in Malaysian Calloselasma rhodostoma and Ophiophagus hannah species, respectively. Flavin monoamine oxidase, phospholipase A2, phosphodiesterase, snake venom metalloproteinase, and serine protease toxin families were identified in both venoms. Aminopeptidase, glutaminyl-peptide cyclotransferase along with ankyrin repeats were identified for the first time in CR venom, and insulin, c-type lectins/snaclecs, hepatocyte growth factor, and macrophage colony-stimulating factor together with tumor necrosis factor were identified in OH venom for the first time. Our combined proteomic approach has identified a comprehensive arsenal of toxins in CR and OH venoms. These data may be utilized for improved antivenom production, understanding pathological effects of envenoming, and the discovery of biologically active peptides with medical and/or biotechnological value.
Collapse
Affiliation(s)
- Sugita Kunalan
- Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Jalan Lagoon Selatan, Bandar Sunway, Selangor Darul Ehsan 47500, Malaysia.
| | - Iekhsan Othman
- Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Jalan Lagoon Selatan, Bandar Sunway, Selangor Darul Ehsan 47500, Malaysia.
| | - Sharifah Syed Hassan
- Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Jalan Lagoon Selatan, Bandar Sunway, Selangor Darul Ehsan 47500, Malaysia.
| | - Wayne C Hodgson
- Monash Venom Group, Department of Pharmacology, Faculty of Medicine, Nursing and Health Sciences, Monash University, Victoria 3800, Australia.
| |
Collapse
|
31
|
Estevão-Costa MI, Sanz-Soler R, Johanningmeier B, Eble JA. Snake venom components in medicine: From the symbolic rod of Asclepius to tangible medical research and application. Int J Biochem Cell Biol 2018; 104:94-113. [PMID: 30261311 DOI: 10.1016/j.biocel.2018.09.011] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2018] [Revised: 09/03/2018] [Accepted: 09/19/2018] [Indexed: 12/21/2022]
Abstract
Both mythologically and logically, snakes have always fascinated man. Snakes have attracted both awe and fear not only because of the elegant movement of their limbless bodies, but also because of the potency of their deadly venoms. Practically, in 2017, the world health organization (WHO) listed snake envenomation as a high priority neglected disease, as snakes inflict up to 2.7 million poisonous bites, around 100.000 casualties, and about three times as many invalidities on man. The venoms of poisonous snakes are a cocktail of potent compounds which specifically and avidly target numerous essential molecules with high efficacy. The individual effects of all venom toxins integrate into lethal dysfunctions of almost any organ system. It is this efficacy and specificity of each venom component, which after analysis of its structure and activity may serve as a potential lead structure for chemical imitation. Such toxin mimetics may help in influencing a specific body function pharmaceutically for the sake of man's health. In this review article, we will give some examples of snake venom components which have spurred the development of novel pharmaceutical compounds. Moreover, we will provide examples where such snake toxin-derived mimetics are in clinical use, trials, or consideration for further pharmaceutical exploitation, especially in the fields of hemostasis, thrombosis, coagulation, and metastasis. Thus, it becomes clear why a snake captured its symbolic place at the Asclepius rod with good reason still nowadays.
Collapse
Affiliation(s)
- Maria-Inacia Estevão-Costa
- Institute of Physiological Chemistry and Pathobiochemistry, University of Münster, Waldeyerstr. 15, 48149, Münster, Germany
| | - Raquel Sanz-Soler
- Institute of Physiological Chemistry and Pathobiochemistry, University of Münster, Waldeyerstr. 15, 48149, Münster, Germany
| | - Benjamin Johanningmeier
- Institute of Physiological Chemistry and Pathobiochemistry, University of Münster, Waldeyerstr. 15, 48149, Münster, Germany
| | - Johannes A Eble
- Institute of Physiological Chemistry and Pathobiochemistry, University of Münster, Waldeyerstr. 15, 48149, Münster, Germany.
| |
Collapse
|
32
|
Song SY, Bae K, Shin KH, Yoo HS. A Case Series of Snake Venom Pharmacopuncture for Chemotherapy-Induced Peripheral Neuropathy: A Retrospective Observational Study. J Pharmacopuncture 2018; 20:280-286. [PMID: 30151298 PMCID: PMC6104720 DOI: 10.3831/kpi.2017.20.034] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2017] [Revised: 11/17/2017] [Accepted: 11/28/2017] [Indexed: 11/16/2022] Open
Abstract
Objective This case series aims to report the efficacy and the safety of using snake venom pharmacopuncture (SVP) for chemotherapy-induced peripheral neuropathy (CIPN). Methods Three heterogeneous cancer (1 endometrium, 1 cervix, and 1 prostate cancer) patients were referred to the East-West Cancer Center (EWCC), Dunsan Korean Medicine Hospital of Daejeon University, from August 02, 2017, to September 15, 2017, for treatment with SVP, and they were treated with SVP 4 times, 6 times, and 8 times, respectively. During the treatment period, the efficacy of SVP therapy was assessed by using the Numerical Rating Scale (NRS) and the Common Terminology Criteria for Adverse Events (CTCAE), and the stability was evaluated by using blood tests. Following each session, all patients were examined closely for any allergenic responses or adverse effects. Results All patients showed noticeable improvements of their NRS and CTCAE scores. Except for bleeding and bruising at the SVP injection site, no major side effects were noted. One of the patients reported mild chilling and a sore throat after receiving the second treatment; those symptoms went away after a few hours. No hematologic toxicity, hepatotoxicity, or nephrotoxicity was found on the blood test. Conclusion The results of this research suggest positive potential benefits of using SVP for treating patients with CIPN. Also, the excellent safety results of SVP seen in this research should lead to larger clinical trials aimed at developing SVP into a potential intervention for managing patients with the symptoms of CIPN.
Collapse
Affiliation(s)
- Si Yeon Song
- East-West Cancer Center, Dunsan Korean Medicine Hospital of Daejeon University, Daejeon, The Republic of Korea
| | - Kyeore Bae
- East-West Cancer Center, Dunsan Korean Medicine Hospital of Daejeon University, Daejeon, The Republic of Korea
| | - Kwhang Ho Shin
- Dalraechon Korean Medicine Clinic, Yangyang-gun, Gangwon-do, The Republic of Korea
| | - Hwa-Seung Yoo
- East-West Cancer Center, Dunsan Korean Medicine Hospital of Daejeon University, Daejeon, The Republic of Korea
| |
Collapse
|
33
|
Chérifi F, Saoud S, Laraba-Djebari F. Molecular modeling, biochemical characterization, and pharmacological properties of Cc 3 -SPase: A platelet-aggregating thrombin-like enzyme purified from Cerastes cerastes venom. J Biochem Mol Toxicol 2018; 32:e22165. [PMID: 29979475 DOI: 10.1002/jbt.22165] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2018] [Revised: 05/23/2018] [Accepted: 06/15/2018] [Indexed: 01/25/2023]
Abstract
Cc3 -SPase (30 kDa-proteinase; pI 5.98) was isolated from Cerastes cerastes venom. Its sequence of 271 residues yielded from LC-MALDI-TOF showed high degrees of homology when aligned with other proteinases. Cc3 -SPase cleaved natural and synthetic proteins such as casein and fibrinogen leaving fibrin clots unaffected. Cc3 -SPase was fully abolished by ion chelators, whereas aprotinin, antithrombin III (Sigma Aldrich, Saint-Louis, Missouri, USA), and heparin were ineffective. Affinity of Cc3 -SPase to benzamidine indicated the presence of an aspartate residue in the catalytic site as confirmed by three-dimensional structure consisting of 14 β-strands and four α-helices. Molecular mechanisms revealed that Cc3 -SPase is capable of promoting dysfunctional platelet aggregation via two signaling pathways mediated by the G-coupled protein receptors and αIIbβ3 integrin. Cc3 -SPase is involved in both extrinsic/intrinsic coagulation pathways in deficient plasmas by replacing defective/lacking factors FII, FVII, and FVIII but not FX. Cc3 -SPase could substitute missing factors in blood diseases related to plasma factor deficiencies.
Collapse
Affiliation(s)
- Fatah Chérifi
- USTHB, Faculty of Biological Sciences, Laboratory of Cellular and Molecular Biology, Bab Ezzouar, Algiers, Algeria
| | - Samah Saoud
- USTHB, Faculty of Biological Sciences, Laboratory of Cellular and Molecular Biology, Bab Ezzouar, Algiers, Algeria
| | - Fatima Laraba-Djebari
- USTHB, Faculty of Biological Sciences, Laboratory of Cellular and Molecular Biology, Bab Ezzouar, Algiers, Algeria
| |
Collapse
|
34
|
Li H, Huang Y, Wu X, Wu T, Cao Y, Wang Q, Qiu Y, Fu W, Zhang Q, Pang J. Effects of hemocoagulase agkistrodon on the coagulation factors and its procoagulant activities. DRUG DESIGN DEVELOPMENT AND THERAPY 2018; 12:1385-1398. [PMID: 29872262 PMCID: PMC5973382 DOI: 10.2147/dddt.s159210] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
Objective Hemocoagulase agkistrodon (HCA), a thrombin-like enzyme (TLE) from the venom of the Chinese moccasin snake (Deinagkistrodon acutus), has been used in clinical practice as a hemostatic compound. The aim of this study was to further investigate the pharmacological properties of HCA. Materials and methods Sodium dodecyl sulfate or native polyacrylamide gel electrophoresis (SDS- or N-PAGE) as well as enzyme linked immunosorbent assays (ELISAs) were conducted to study the effects of HCA on the human plasma fibrinogen and prothrombin levels, as well as its in vitro interactions with some coagulation factors. In addition, the bleeding time effects of HCA in the mouse tail-bleeding model as well as its effects on the fibrinogen levels in rabbits were determined in vivo. Results In vitro results revealed that HCA exerts its procoagulant activities by hydrolyzing fibrinogen into segments that are easier to be absorbed, reducing the risk of thrombus formation. Besides, HCA could significantly inhibit the activation of prothrombin at the concentration of 0.3 μM. Unexpectedly, we also found that HCA was able to strongly bind to factor X/Xa (in a ratio of 1:1) and thus inhibit the acceleration of active factor X to tissue plasminogen activator-catalyzed plasminogen activation, demonstrating that it could be less likely to lead to thrombus formation. Finally, in vivo results indicated that HCA could significantly shorten the bleeding time in the mouse tail-bleeding model and had no effect on the fibrinogen levels in rabbits. Conclusion In summary, HCA, a unique and new family member of TLEs, may become a new clinical drug for the prevention and treatment of hemorrhage due to its unique and complex interactions with the blood system. Clarification of these features will enable us to further understand the mechanism of action of HCA and then promote its further application in clinical practice as a therapeutic drug.
Collapse
Affiliation(s)
- Haixin Li
- Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, People's Republic of China
| | - Ying Huang
- Department of Pharmacy, Shenzhen Hospital, Southern Medical University, Shenzhen, People's Republic of China
| | - Xian Wu
- Department of Pharmacy, The Third Affiliated Hospital, Southern Medical University, Guangzhou, People's Republic of China
| | - Ting Wu
- Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, People's Republic of China
| | - Ying Cao
- Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, People's Republic of China
| | - Qimei Wang
- Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, People's Republic of China
| | - Yuchang Qiu
- Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, People's Republic of China
| | - Weiming Fu
- Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, People's Republic of China
| | - Qun Zhang
- Good Clinical Practice Development, The Third Affiliated Hospital, Southern Medical University, Guangzhou, People's Republic of China
| | - Jianxin Pang
- Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, People's Republic of China
| |
Collapse
|
35
|
Ullah A, Masood R, Ali I, Ullah K, Ali H, Akbar H, Betzel C. Thrombin-like enzymes from snake venom: Structural characterization and mechanism of action. Int J Biol Macromol 2018; 114:788-811. [PMID: 29604354 DOI: 10.1016/j.ijbiomac.2018.03.164] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2018] [Revised: 03/08/2018] [Accepted: 03/23/2018] [Indexed: 01/15/2023]
Abstract
Snake venom thrombin-like enzymes (SVTLEs) constitute the major portion (10-24%) of snake venom and these are the second most abundant enzymes present in the crude venom. During envenomation, these enzymes had shown prominently the various pathological effects, such as disturbance in hemostatic system, fibrinogenolysis, fibrinolysis, platelet aggregation, thrombosis, neurologic disorders, activation of coagulation factors, coagulant, procoagulant etc. These enzymes also been used as a therapeutic agent for the treatment of various diseases such as congestive heart failure, ischemic stroke, thrombotic disorders etc. Although the crystal structures of five SVTLEs are available in the Protein Data Bank (PDB), there is no single article present in the literature that has described all of them. The current work describes the structural aspects, structure-based mechanism of action, processing and inhibition of these enzymes. The sequence analysis indicates that these enzymes show a high sequence identity (57-85%) with each other and low sequence identity with trypsin (36-43%), human alpha-thrombin (29-36%) and other snake venom serine proteinases (57-85%). Three-dimensional structural analysis indicates that the loops surrounding the active site are variable both in amino acids composition and length that may convey variable substrate specificity to these enzymes. The surface charge distributions also vary in these enzymes. Docking analysis with suramin shows that this inhibitor preferably binds to the C-terminal region of these enzymes and causes the destabilization of their three-dimensional structure.
Collapse
Affiliation(s)
- Anwar Ullah
- Department of Biosciences, COMSATS Institute of Information Technology Park Road, Islamabad 45550, Pakistan.
| | - Rehana Masood
- Department of Biochemistry, Shaheed Benazir Bhutto Women University, Peshawar 25000, Pakistan
| | - Ijaz Ali
- Department of Biosciences, COMSATS Institute of Information Technology Park Road, Islamabad 45550, Pakistan
| | - Kifayat Ullah
- Department of Biosciences, COMSATS Institute of Information Technology Park Road, Islamabad 45550, Pakistan
| | - Hamid Ali
- Department of Biosciences, COMSATS Institute of Information Technology Park Road, Islamabad 45550, Pakistan
| | - Haji Akbar
- Department of Biosciences, COMSATS Institute of Information Technology Park Road, Islamabad 45550, Pakistan
| | - Christian Betzel
- Laboratory for Structural Biology of Infection and Inflammation, University of Hamburg, c/o DESY, Notkestrasse 85, 22603 Hamburg, Germany
| |
Collapse
|
36
|
Gowda R, Rajaiah R, Angaswamy N, Krishna S, Bannikuppe Sannanayak V. Biochemical and pharmacological characterization of Trimersurus malabaricus snake venom. J Cell Biochem 2018. [PMID: 29528146 DOI: 10.1002/jcb.26782] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Trimeresurus malabaricus is a venomous pit viper species endemic to southwestern part of India. In earlier reports, we have shown that envenomation by T. malabaricus venom leading to strong local tissue damage but the mechanism of action is not clearly revealed. Local tissue damage affected by T. malabaricus venom is of great importance since the poison has serious systemic effects including death in the case of multiple attacks. The present study details the major manifestations of T. malabaricus venom and the induction of local tissue damage, which suggests that most toxins are present in the form of hydrolytic enzymes. Hydrolytic activity of the enzymes was measured and the data indicated that protease and phospholipase A2 activity was high which is responsible for local tissue damage. Furthermore, the role of hydrolytic enzymes in the induction of pathological events such as hemorrhage, edema, myotoxicity, and blood coagulation examination were assessed through animal models.
Collapse
Affiliation(s)
- Raghavendra Gowda
- Department of Pharmacology, Penn State College of Medicine, Penn State University, Hershey, Pennsylvania
| | - Rajesh Rajaiah
- Department of Studies in Biochemistry, University of Mysore, Manasagangotri, Mysuru, Karnataka, India
| | - Nataraj Angaswamy
- Department of Pharmacology, Penn State College of Medicine, Penn State University, Hershey, Pennsylvania
| | - Sharath Krishna
- Department of Natural Sciences, Central State University, Wilberforce, Ohio
| | | |
Collapse
|
37
|
A New Platelet-Aggregation-Inhibiting Factor Isolated from Bothrops moojeni Snake Venom. BIOMED RESEARCH INTERNATIONAL 2017; 2017:4315832. [PMID: 29226136 PMCID: PMC5687129 DOI: 10.1155/2017/4315832] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/07/2017] [Revised: 09/12/2017] [Accepted: 09/24/2017] [Indexed: 11/29/2022]
Abstract
This work reports the purification and functional characterization of BmooPAi, a platelet-aggregation-inhibiting factor from Bothrops moojeni snake venom. The toxin was purified by a combination of three chromatographic steps (ion-exchange on DEAE-Sephacel, molecular exclusion on Sephadex G-75, and affinity chromatography on HiTrap™ Heparin HP). BmooPAi was found to be a single-chain protein with an apparent molecular mass of 32 kDa on 14% SDS-PAGE, under reducing conditions. Sequencing of BmooPAi by Edman degradation revealed the amino acid sequence LGPDIVPPNELLEVM. The toxin was devoid of proteolytic, haemorrhagic, defibrinating, or coagulant activities and induced no significant oedema or hyperalgesia. BmooPAi showed a rather specific inhibitory effect on ristocetin-induced platelet aggregation in human platelet-rich plasma, whereas it had little or no effect on platelet aggregation induced by collagen and adenosine diphosphate. The results presented in this work suggest that BmooPAi is a toxin comprised of disintegrin-like and cysteine-rich domains, originating from autolysis/proteolysis of PIII SVMPs from B. moojeni snake venom. This toxin may be of medical interest because it is a platelet aggregation inhibitor, which could potentially be developed as a novel therapeutic agent to prevent and/or treat patients with thrombotic disorders.
Collapse
|
38
|
Evaluation of the physiological activity of venom from the Eurasian water shrew Neomys fodiens. Front Zool 2017; 14:46. [PMID: 29026428 PMCID: PMC5622582 DOI: 10.1186/s12983-017-0230-0] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2017] [Accepted: 09/18/2017] [Indexed: 11/10/2022] Open
Abstract
Background Animal toxins can have medical and therapeutic applications. Principally, toxins produced by insects, arachnids, snakes and frogs have been characterized. Venomous mammals are rare, and their venoms have not been comprehensively investigated. Among shrews, only the venom of Blarina brevicauda has been analysed so far, and blarina toxin has been proven to be its main toxic component. It is assumed that Neomys fodiens employs its venom to hunt larger prey. However, the toxic profile, properties and mode of action of its venom are largely unknown. Therefore, we analysed the cardio-, myo- and neurotropic properties of N. fodiens venom and saliva of non-venomous Sorex araneus (control tests) in vitro in physiological bioassays carried out on two model organisms: beetles and frogs. For the first time, we fractionated N. fodiens venom and S. araneus saliva by performing chromatographic separation. Next, the properties of selected compounds were analysed in cardiotropic bioassays in the Tenebrio molitor heart. Results The venom of N. fodiens caused a high decrease in the conduction velocity of the frog sciatic nerve, as well as a significant decrease in the force of frog calf muscle contraction. We also recorded a significant decrease in the frog heart contractile activity. Most of the selected compounds from N. fodiens venom displayed a positive chronotropic effect on the beetle heart. However, one fraction caused a strong decrease in the T. molitor heart contractile activity coupled with a reversible cardiac arrest. We did not observe any responses of the insect heart and frog organs to the saliva of S. araneus. Preliminary mass spectrometry analysis revealed that calmodulin-like protein, thymosin β-10, hyaluronidase, lysozyme C and phospholipase A2 are present in the venom of N. fodiens, whereas thymosin β4, lysozyme C and β-defensin are present in S. araneus saliva. Conclusion Our results showed that N. fodiens venom has stronger paralytic properties and lower cardioinhibitory activity. Therefore, it is highly probable that N. fodiens might use its venom as a prey immobilizing agent. We also confirmed that S. araneus is not a venomous mammal because its saliva did not exhibit any toxic effects. Electronic supplementary material The online version of this article (10.1186/s12983-017-0230-0) contains supplementary material, which is available to authorized users.
Collapse
|
39
|
Koludarov I, Jackson TN, Brouw BOD, Dobson J, Dashevsky D, Arbuckle K, Clemente CJ, Stockdale EJ, Cochran C, Debono J, Stephens C, Panagides N, Li B, Manchadi MLR, Violette A, Fourmy R, Hendrikx I, Nouwens A, Clements J, Martelli P, Kwok HF, Fry BG. Enter the Dragon: The Dynamic and Multifunctional Evolution of Anguimorpha Lizard Venoms. Toxins (Basel) 2017; 9:E242. [PMID: 28783084 PMCID: PMC5577576 DOI: 10.3390/toxins9080242] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2017] [Revised: 08/04/2017] [Accepted: 08/04/2017] [Indexed: 01/08/2023] Open
Abstract
While snake venoms have been the subject of intense study, comparatively little work has been done on lizard venoms. In this study, we have examined the structural and functional diversification of anguimorph lizard venoms and associated toxins, and related these results to dentition and predatory ecology. Venom composition was shown to be highly variable across the 20 species of Heloderma, Lanthanotus, and Varanus included in our study. While kallikrein enzymes were ubiquitous, they were also a particularly multifunctional toxin type, with differential activities on enzyme substrates and also ability to degrade alpha or beta chains of fibrinogen that reflects structural variability. Examination of other toxin types also revealed similar variability in their presence and activity levels. The high level of venom chemistry variation in varanid lizards compared to that of helodermatid lizards suggests that venom may be subject to different selection pressures in these two families. These results not only contribute to our understanding of venom evolution but also reveal anguimorph lizard venoms to be rich sources of novel bioactive molecules with potential as drug design and development lead compounds.
Collapse
Affiliation(s)
- Ivan Koludarov
- Venom Evolution Lab, School of Biological Sciences, University of Queensland, St. Lucia QLD 4072, Australia.
| | - Timothy Nw Jackson
- Venom Evolution Lab, School of Biological Sciences, University of Queensland, St. Lucia QLD 4072, Australia.
- Australian Venom Research Unit, School of Biomedical Sciences, Level 2 Medical Building, University of Melbourne, Victoria 3010, Australia.
| | - Bianca Op den Brouw
- Venom Evolution Lab, School of Biological Sciences, University of Queensland, St. Lucia QLD 4072, Australia.
| | - James Dobson
- Venom Evolution Lab, School of Biological Sciences, University of Queensland, St. Lucia QLD 4072, Australia.
| | - Daniel Dashevsky
- Venom Evolution Lab, School of Biological Sciences, University of Queensland, St. Lucia QLD 4072, Australia.
| | - Kevin Arbuckle
- Department of Biosciences, College of Science, Swansea University, Swansea SA2 8PP, UK.
| | - Christofer J Clemente
- University of the Sunshine Coast, School of Science and Engineering, Sippy Downs, Queensland 4558, Australia.
| | | | - Chip Cochran
- Department of Earth and Biological Sciences, Loma Linda University, Loma Linda, CA 92350, USA.
| | - Jordan Debono
- Venom Evolution Lab, School of Biological Sciences, University of Queensland, St. Lucia QLD 4072, Australia.
| | - Carson Stephens
- School of Biomedical Sciences, Queensland University of Technology, Brisbane QLD 4001, Australia.
| | - Nadya Panagides
- Venom Evolution Lab, School of Biological Sciences, University of Queensland, St. Lucia QLD 4072, Australia.
| | - Bin Li
- Faculty of Health Sciences, University of Macau, Avenida da Universidade, Taipa, Macau.
| | | | - Aude Violette
- Alphabiotoxine Laboratory sprl, Barberie 15, 7911 Montroeul-au-bois, Belgium.
| | - Rudy Fourmy
- Alphabiotoxine Laboratory sprl, Barberie 15, 7911 Montroeul-au-bois, Belgium.
| | - Iwan Hendrikx
- Venom Evolution Lab, School of Biological Sciences, University of Queensland, St. Lucia QLD 4072, Australia.
| | - Amanda Nouwens
- School of Chemistry and Molecular Biology, University of Queenslnd, St. Lucia QLD 4072, Australia.
| | - Judith Clements
- School of Biomedical Sciences, Queensland University of Technology, Brisbane QLD 4001, Australia.
| | | | - Hang Fai Kwok
- Faculty of Health Sciences, University of Macau, Avenida da Universidade, Taipa, Macau.
| | - Bryan G Fry
- Venom Evolution Lab, School of Biological Sciences, University of Queensland, St. Lucia QLD 4072, Australia.
| |
Collapse
|
40
|
Yadav PK, Antonyraj CB, Basheer Ahamed SI, Srinivas S. Understanding Russell's viper venom factor V activator's substrate specificity by surface plasmon resonance and in-silico studies. PLoS One 2017; 12:e0181216. [PMID: 28732041 PMCID: PMC5521794 DOI: 10.1371/journal.pone.0181216] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2017] [Accepted: 06/28/2017] [Indexed: 11/18/2022] Open
Abstract
Blood coagulation factor V (FV) is activated either by Factor X or thrombin, cleaving at three different sites viz., Site I (Arg709-Ser710), site II (Arg1018-Thr1019), and site III (Arg1545-Ser1546). Russell's viper venom factor V activator (RVV-V) is a thrombin-like serine proteinase that activates FV with selective, single cleavage at site III. A long lasting effort is being pending in understanding the 'selective' binding specificity of the RVV-V towards site III. Here, we present the binding kinetic study of RVV-V with two designed peptides corresponding to the regions from site I (Gln699-Asn713) and site II (1008Lys-Pro1022), respectively, that include 15 amino acids. Our investigation for justifying the binding efficacy and kinetics of peptides includes SPR method, protein-peptide docking, molecular dynamics simulation, and principal component analysis (PCA). Surprisingly, the SPR experiment disclosed that the Peptide II showed a lower binding affinity with KD of 2.775 mM while the Peptide I showed none. Docking and simulation of both the peptides with RVV-V engaged either rooted or shallow binding for Peptide II and Peptide I respectively. The peptide binding resulted in global conformational changes in the native fold of RVV-V, whereas the similar studies for thrombin failed to make major changes in the native fold. In support, the PCA analysis for RVV-V showed the dislocation of catalytic triad upon binding both the peptides. Hence, RVV-V, a serine protease, is incompetent in cleaving these two sites. This study suggests a transition in RVV-V from the native rigid to the distorted flexible structure and paves a way to design a new peptide substrate/inhibitor.
Collapse
Affiliation(s)
- Pradeep K. Yadav
- Centre for Bioinformatics, Pondicherry University, Pondicherry, India
| | | | | | - Sistla Srinivas
- GE Healthcare Life Sciences, John F Welch Technology Centre, EPIP, Bengaluru, India
| |
Collapse
|
41
|
Yang ZM, Yu H, Liu ZZ, Pei JZ, Yang YE, Yan SX, Zhang C, Zhao WL, Wang ZZ, Wang YM, Tsai IH. Serine protease isoforms in Gloydius intermedius venom: Full sequences, molecular phylogeny and evolutionary implications. J Proteomics 2017; 164:19-32. [DOI: 10.1016/j.jprot.2017.05.024] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2017] [Revised: 05/18/2017] [Accepted: 05/25/2017] [Indexed: 02/07/2023]
|
42
|
Slagboom J, Kool J, Harrison RA, Casewell NR. Haemotoxic snake venoms: their functional activity, impact on snakebite victims and pharmaceutical promise. Br J Haematol 2017; 177:947-959. [PMID: 28233897 PMCID: PMC5484289 DOI: 10.1111/bjh.14591] [Citation(s) in RCA: 167] [Impact Index Per Article: 20.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2016] [Accepted: 01/09/2017] [Indexed: 12/22/2022]
Abstract
Snake venoms are mixtures of numerous proteinacious components that exert diverse functional activities on a variety of physiological targets. Because the toxic constituents found in venom vary from species to species, snakebite victims can present with a variety of life-threatening pathologies related to the neurotoxic, cytotoxic and haemotoxic effects of venom. Of the 1·8 million people envenomed by snakes every year, up to 125 000 die, while hundreds of thousands survive only to suffer with life-changing long-term morbidity. Consequently, snakebite is one of the world's most severe neglected tropical diseases. Many snake venoms exhibit strong haemotoxic properties by interfering with blood pressure, clotting factors and platelets, and by directly causing haemorrhage. In this review we provide an overview of the functional activities of haemotoxic venom proteins, the pathologies they cause in snakebite victims and how their exquisite selectivity and potency make them amenable for use as therapeutic and diagnostic tools relevant for human medicine.
Collapse
Affiliation(s)
- Julien Slagboom
- Alistair Reid Venom Research Unit, Parasitology Department, Liverpool School of Tropical Medicine, Liverpool, UK
- Division of BioAnalytical Chemistry, Department of Chemistry and Pharmaceutical Sciences, Faculty of Sciences, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Jeroen Kool
- Division of BioAnalytical Chemistry, Department of Chemistry and Pharmaceutical Sciences, Faculty of Sciences, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Robert A Harrison
- Alistair Reid Venom Research Unit, Parasitology Department, Liverpool School of Tropical Medicine, Liverpool, UK
| | - Nicholas R Casewell
- Alistair Reid Venom Research Unit, Parasitology Department, Liverpool School of Tropical Medicine, Liverpool, UK
| |
Collapse
|
43
|
Nguyen TTN, Ha TT, Nguyen TH, Vu TH, Truong NH, Chu HH, Van Quyen D. Peptide Fraction pOh2 Exerts Antiadipogenic Activity through Inhibition of C/EBP- α and PPAR- γ Expression in 3T3-L1 Adipocytes. BIOMED RESEARCH INTERNATIONAL 2017; 2017:4826595. [PMID: 28424783 PMCID: PMC5382294 DOI: 10.1155/2017/4826595] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/22/2016] [Revised: 02/15/2017] [Accepted: 03/15/2017] [Indexed: 01/14/2023]
Abstract
Many studies have comprehensively examined the venom of Ophiophagus hannah snake. Its venom comprises different compounds exhibiting a wide range of pharmacological activities. In this investigation, four peptide fractions (PFs), ranging from 3 kDa to 10 kDa, isolated from the Vietnamese snake venom of O. hannah were separated by HPLC and investigated for their inhibitory activity on adipogenesis in 3T3-L1 adipocytes. The most effective PF was then further purified, generating two peptides, pOh1 and pOh2. Upon investigation of these two peptides on 3T3-L1 adipocytes, it was revealed that, at 10 μg/mL, pOh2 was able to inhibit the lipid accumulation in 3T3-L1 adipocytes by up to 56%, without affecting cell viability. Furthermore, the pOh2 downregulated the gene expression of important transcription factors C/EBP-α and PPAR-γ. In addition, aP2 and GPDH adipocyte-specific markers were also significantly reduced compared to untreated differentiated cells. Taken together, pOh2 inhibited the expression of key transcription factors C/EBP-α and PPAR-γ and their target genes, aP2 and GPDH, thereby blocking the adipocyte differentiation. In conclusion, this novel class of peptide might have potential for in vivo antiobesity effects.
Collapse
Affiliation(s)
- Thi Tuyet Nhung Nguyen
- Institute of Biotechnology (IBT), Vietnam Academy of Science and Technology (VAST), 18 Hoang Quoc Viet Street, Cau Giay District, Hanoi 100000, Vietnam
| | - Thi Thu Ha
- Institute of Biotechnology (IBT), Vietnam Academy of Science and Technology (VAST), 18 Hoang Quoc Viet Street, Cau Giay District, Hanoi 100000, Vietnam
| | - Thi Hoa Nguyen
- Institute of Biotechnology (IBT), Vietnam Academy of Science and Technology (VAST), 18 Hoang Quoc Viet Street, Cau Giay District, Hanoi 100000, Vietnam
| | - Thi Hien Vu
- Institute of Biotechnology (IBT), Vietnam Academy of Science and Technology (VAST), 18 Hoang Quoc Viet Street, Cau Giay District, Hanoi 100000, Vietnam
| | - Nam Hai Truong
- Institute of Biotechnology (IBT), Vietnam Academy of Science and Technology (VAST), 18 Hoang Quoc Viet Street, Cau Giay District, Hanoi 100000, Vietnam
| | - Hoang Ha Chu
- Institute of Biotechnology (IBT), Vietnam Academy of Science and Technology (VAST), 18 Hoang Quoc Viet Street, Cau Giay District, Hanoi 100000, Vietnam
| | - Dong Van Quyen
- Institute of Biotechnology (IBT), Vietnam Academy of Science and Technology (VAST), 18 Hoang Quoc Viet Street, Cau Giay District, Hanoi 100000, Vietnam
| |
Collapse
|
44
|
Shih CH, Chiang TB, Wang WJ. Convulxin, a C-type lectin-like protein, inhibits HCASMCs functions via WAD-motif/integrin-αv interaction and NF-κB-independent gene suppression of GRO and IL-8. Exp Cell Res 2017; 352:234-244. [PMID: 28192121 DOI: 10.1016/j.yexcr.2017.02.008] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2016] [Revised: 01/25/2017] [Accepted: 02/07/2017] [Indexed: 01/13/2023]
Abstract
Convulxin (CVX), a C-type lectin-like protein (CLPs), is a potent platelet aggregation inducer. To evaluate its potential applications in angiogenic diseases, the multimeric CVX were further explored on its mode of actions toward human coronary artery smooth muscle cells (HCASMCs). The N-terminus of β-chain of CVX (CVX-β) contains a putative disintegrin-like domain with a conserved motif upon the sequence comparison with other CLPs. Importantly, native CVX had no cytotoxic activity as examined by electrophoretic pattern. A Trp-Ala-Asp (WAD)-containing octapeptide, MTWADAEK, was thereafter synthesized and analyzed in functional assays. In the case of specific integrin antagonists as positive controls, the anti-angiogenic effects of CVX on HCASMCs were investigated by series of functional analyses. CVX showed to exhibit multiple inhibitory activities toward HCASMCs proliferation, adhesion and invasion with a dose- and integrin αvβ3-dependent fashion. However, the WAD-octapeptide exerting a minor potency could also work as an active peptidomimetic. In addition, flow cytometric analysis demonstrated both the intact CVX and synthetic peptide can specifically interact with integrin-αv on HCASMCs and CVX was shown to have a down-regulatory effect on the gene expression of CXC-chemokines, such as growth-related oncogene and interleukin-8. According to nuclear factor-κB (NF-κB) p65 translocation assay and Western blotting analysis, the NF-κB activation was not involved in the signaling events of CVX-induced gene expression. In conclusion, CVX may act as a disintegrin-like protein via the interactions of WAD-motif in CVX-β with integrin-αv on HCASMCs and it also is a gene suppressor with the ability to diminish the expression of two CXC-chemokines in a NF-κB-independent manner. Indeed, more extensive investigations are needed and might create a new avenue for the development of a novel angiostatic agent.
Collapse
Affiliation(s)
- Chun-Ho Shih
- Chang Gung University of Science and Technology, Guishan Dist., Taoyuan City, Taiwan
| | - Tin-Bin Chiang
- Chang Gung University of Science and Technology, Guishan Dist., Taoyuan City, Taiwan
| | - Wen-Jeng Wang
- Chang Gung University of Science and Technology, Guishan Dist., Taoyuan City, Taiwan; Department of Neurological Surgery, Chang Gung Memorial Hospital, Guishan Dist., Taoyuan City, Taiwan.
| |
Collapse
|
45
|
Al-Asmari AK, Riyasdeen A, Al-Shahrani MH, Islam M. Snake venom causes apoptosis by increasing the reactive oxygen species in colorectal and breast cancer cell lines. Onco Targets Ther 2016; 9:6485-6498. [PMID: 27799796 PMCID: PMC5079696 DOI: 10.2147/ott.s115055] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
Snake venom possesses various kinds of proteins and neurotoxic polypeptides, which can negatively interfere with the neurotransmitter signaling cascade. This phenomenon occurs mainly due to the blocking of ion channels in the body system. Envenomation prevents or severely interrupts nerve impulses from being transmitted, inhibition of adenosine triphosphate synthesis, and proper functioning of the cardiac muscles. However, some beneficial properties of venoms have also been reported. The aim of this study was to examine the snake venom as an anticancer agent due to its inhibitory effects on cancer progression such as cell motility, cell invasion, and colony formation. In this study, the effect of venoms on phenotypic changes and the change on molecular level in colorectal and breast cancer cell lines were examined. A reduction of 60%–90% in cell motility, colony formation, and cell invasion was observed when these cell lines were treated with different concentrations of snake venom. In addition, the increase in oxidative stress that results in an increase in the number of apoptotic cancer cells was significantly higher in the venom-treated cell lines. Further analysis showed that there was a decrease in the expression of pro-inflammatory cytokines and signaling proteins, strongly suggesting a promising role for snake venom against breast and colorectal cancer cell progression. In conclusion, the snake venoms used in this study showed significant anticancer properties against colorectal and breast cancer cell lines.
Collapse
Affiliation(s)
| | | | - Mohammad Hamed Al-Shahrani
- Pediatric Hematology/Oncology and Bone Marrow Transplant Unit, Prince Sultan Military Medical City, Riyadh, Kingdom of Saudi Arabia
| | | |
Collapse
|
46
|
Metalloproteases Affecting Blood Coagulation, Fibrinolysis and Platelet Aggregation from Snake Venoms: Definition and Nomenclature of Interaction Sites. Toxins (Basel) 2016; 8:toxins8100284. [PMID: 27690102 PMCID: PMC5086644 DOI: 10.3390/toxins8100284] [Citation(s) in RCA: 114] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2016] [Revised: 09/21/2016] [Accepted: 09/22/2016] [Indexed: 01/08/2023] Open
Abstract
Snake venom metalloproteases, in addition to their contribution to the digestion of the prey, affect various physiological functions by cleaving specific proteins. They exhibit their activities through activation of zymogens of coagulation factors, and precursors of integrins or receptors. Based on their structure–function relationships and mechanism of action, we have defined classification and nomenclature of functional sites of proteases. These metalloproteases are useful as research tools and in diagnosis and treatment of various thrombotic and hemostatic conditions. They also contribute to our understanding of molecular details in the activation of specific factors involved in coagulation, platelet aggregation and matrix biology. This review provides a ready reference for metalloproteases that interfere in blood coagulation, fibrinolysis and platelet aggregation.
Collapse
|
47
|
de Oliveira F, de Sousa BB, Mamede CCN, de Morais NCG, de Queiroz MR, da Cunha Pereira DF, Matias MS, Homi Brandeburgo MI. Biochemical and functional characterization of BmooSP, a new serine protease from Bothrops moojeni snake venom. Toxicon 2016; 111:130-8. [PMID: 26797102 DOI: 10.1016/j.toxicon.2016.01.055] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2015] [Revised: 12/30/2015] [Accepted: 01/10/2016] [Indexed: 09/30/2022]
Abstract
In this work, we describe the purification and characterization of a new serine protease enzyme from Bothrops moojeni snake venom (BmooSP). On SDS-PAGE, BmooSP was found to be a single-chain protein with an apparent molecular mass of 36,000 and 32,000 under reduced and non-reduced conditions, respectively. Mass spectrometry analysis showed that the BmooSP is composed by two isoforms with molecular mass of 30,363 and 30,070, respectively. The purified enzyme consists of 277 amino acid residues, disregarding the cysteine and tryptophan residues that have been degraded by acid hydrolysis, and its N-terminal sequence showed similarity with other serine protease enzymes. BmooSP induced blood-clotting in vitro, defibrination in vivo, caseinolytic and fibrin(ogen)olytic activities. The enzyme is stable at high temperatures (up to 100 °C) and shows maximum activity at pH around 7.0. Preliminary results show that BmooSP can induce the formation of a stable fibrin clot for more than 10 days. BmooSP presents medical interest because it can be used as biodegradable fibrin glue and for the treatment and prevention of cardiovascular disorders because of its ability to promote the defibrination in vivo, decreasing blood viscosity and improving blood circulation.
Collapse
Affiliation(s)
- Fábio de Oliveira
- Instituto de Ciências Biomédicas, Universidade Federal de Uberlândia, Uberlândia MG, Brazil; Instituto Nacional de Ciência e Tecnologia em Nano-Biofarmacêutica (N-Biofar), Belo Horizonte MG, Brazil.
| | - Bruna Barbosa de Sousa
- Instituto de Genética e Bioquímica, Universidade Federal de Uberlândia, Uberlândia MG, Brazil; Instituto Nacional de Ciência e Tecnologia em Nano-Biofarmacêutica (N-Biofar), Belo Horizonte MG, Brazil
| | - Carla Cristine Neves Mamede
- Instituto de Ciências Biomédicas, Universidade Federal de Uberlândia, Uberlândia MG, Brazil; Instituto Nacional de Ciência e Tecnologia em Nano-Biofarmacêutica (N-Biofar), Belo Horizonte MG, Brazil
| | - Nadia Cristina Gomes de Morais
- Instituto de Genética e Bioquímica, Universidade Federal de Uberlândia, Uberlândia MG, Brazil; Instituto Nacional de Ciência e Tecnologia em Nano-Biofarmacêutica (N-Biofar), Belo Horizonte MG, Brazil
| | - Mayara Ribeiro de Queiroz
- Instituto de Genética e Bioquímica, Universidade Federal de Uberlândia, Uberlândia MG, Brazil; Instituto Nacional de Ciência e Tecnologia em Nano-Biofarmacêutica (N-Biofar), Belo Horizonte MG, Brazil
| | | | - Mariana S Matias
- Instituto de Genética e Bioquímica, Universidade Federal de Uberlândia, Uberlândia MG, Brazil
| | | |
Collapse
|
48
|
Silica Nanoparticles Effects on Blood Coagulation Proteins and Platelets. Biochem Res Int 2016; 2016:2959414. [PMID: 26881078 PMCID: PMC4736757 DOI: 10.1155/2016/2959414] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2015] [Accepted: 12/20/2015] [Indexed: 01/09/2023] Open
Abstract
Interaction of nanoparticles with the blood coagulation is important prior to their using as the drug carriers or therapeutic agents. The aim of present work was studying of the primary effects of silica nanoparticles (SiNPs) on haemostasis in vitro. We studied the effect of SiNPs on blood coagulation directly estimating the activation of prothrombin and factor X and to verify any possible effect of SiNPs on human platelets. It was shown that SiNPs shortened coagulation time in APTT and PT tests and increased the activation of factor X induced by RVV possibly due to the sorption of intrinsic pathway factors on their surface. SiNPs inhibited the aggregation of platelet rich plasma induced by ADP but in the same time partially activated platelets as it was shown using flow cytometry. The possibility of SiNPs usage in nanomedicine is strongly dependant on their final concentration in bloodstream and the size of the particles that are used. However SiNPs are extremely promising as the haemostatic agents for preventing the blood loss after damage.
Collapse
|
49
|
Engineering of Harobin for enhanced fibrinolytic activity obtained by random and site-directed mutagenesis. Protein Expr Purif 2015; 129:162-172. [PMID: 26363113 DOI: 10.1016/j.pep.2015.09.010] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2015] [Revised: 09/06/2015] [Accepted: 09/08/2015] [Indexed: 11/21/2022]
Abstract
We have previously published a report on the cloning and characterization of Harobin, a fibrinolytic serine protease. However, the broad application of this fibrinolytic enzyme is limited by its low expression level that was achieved in Pichia pastoris. To counteract this shortcoming, random and site-directed mutagenesis have been combined in order to improve functional expression and activity of Harobin. By screening 400 clones from random mutant libraries for enhanced fibrinolytic activity, two mutants were obtained: N111R, R230G. By performing site-directed mutagenesis, a Harobin double mutant, N111R/R230G, was constructed and can be functionally expressed at higher level than the wild type enzyme. In addition, it possessed much higher fibrinolytic and amidolytic activity than the wild type enzyme and other single mutants. The N111R/R230G expressed in basal salts medium was purified by a three step purification procedure. At pH of 6.0-9.0, and the temperature range of 40-90 °C, N111R/R230G was more active and more heat resistant. The fibrinolytic activities of Harobin mutants were completely inhibited by PMSF and SBTI, but not by EDTA, EGTA, DTT, indicating that Harobin is a serine protease. N111R/R230G showed much better anti-thrombosis effect than wild type Harobin and single mutants, and could significantly increase bleeding and clotting time. Intravenous injection of N111R/R230G in spontaneous hypertensive rats (SHR) led to a significant reduction in systolic blood pressure (SBP), diastolic blood pressure (DBP) and mean arterial pressure (MAP) (p < 0.01), while heart rate (HR) was not affected. The in vitro and in vivo results of the present study revealed that Harobin double mutant N111R/R230G is an appropriate candidate for biotechnological applications due to its high expression level and high activity in area of thrombosis and hypertension.
Collapse
|
50
|
Amel KS, Fatima LD. Purification and Characterization of a New Serine Protease (VLCII) Isolated fromVipera lebetinaVenom: Its Role in Hemostasis. J Biochem Mol Toxicol 2015; 29:388-97. [DOI: 10.1002/jbt.21709] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2015] [Revised: 03/12/2015] [Accepted: 03/25/2015] [Indexed: 11/08/2022]
Affiliation(s)
- Kadi-Saci Amel
- USTHB, Faculty of Biological Sciences; Laboratory of Cellular and Molecular Biology; BP32 El-Alia Bab Ezzouar Algiers Algeria
| | - Laraba-Djebari Fatima
- USTHB, Faculty of Biological Sciences; Laboratory of Cellular and Molecular Biology; BP32 El-Alia Bab Ezzouar Algiers Algeria
| |
Collapse
|