1
|
Tsai TS, Tsai IH. Full sequencing and comparison of five venom metalloproteases of Trimeresurus gracilis: The PI-enzyme is most similar to okinalysin but the PIII-enzyme is most similar to Crotalus venom enzymes. Toxicon 2023; 225:107053. [PMID: 36758773 DOI: 10.1016/j.toxicon.2023.107053] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2022] [Revised: 02/06/2023] [Accepted: 02/07/2023] [Indexed: 02/11/2023]
Abstract
The cDNAs encoding the Zn+2-metalloproteases (SVMPs) of Trimeresurus gracilis (abbreviated as Tgc), a pitviper endemic to Taiwan, were cloned from venom glands and sequenced. The amino-acid sequences of five novel SVMPs, including one P-III, three P-II and one P-I class enzymes, were thus deduced and subjected to BLAST-analyses. The P-III enzyme (designated as Tgc-PIII) is structurally most similar to the PIII-SVMPs of New World pitvipers but not similar to the PIII-SVMP of Ovophis okinavensis. Sequence-similarity analysis of 22 homologous PIII-SVMPs reveal three major structural subtypes of the pitviper PIII-SVMPs, which possibly have different substrate specificities. In addition, Tgc-PIII and the PI-class SVMP (named Tgc-MP) were isolated from the venom and verified by mass spectrometry. All the three deduced sequences of PII-SVMPs (Tgc-PIIs) contain an abnormal Zn+2-binding-site in their catalytic-domain, and an identical "long-disintegrin" domain. The predicted 85-residues disintegrin, gracilisin, bears high similarities to some long-disintegrins of the New-World pitvipers and salmosin3. By BLAST search and comparison, Tgc-MP is 96% similar to okinalysin, the hemorrhagic PI-SVMP of O. okinavensis, rather than any other PI-SVMPs in the databanks. Our results confirm the fast evolution of Tgc-SVMPs as well as their structural similarities to different SVMP-classes of the New-World pitvipers and of O. okinavensis, respectively. The implications of our findings are discussed along with our previous sequence comparisons of venom phospholipases A2 and ten venom serine proteases of Tgc.
Collapse
Affiliation(s)
- Tein-Shun Tsai
- Department of Biological Science and Technology, National Pingtung University of Science and Technology, Pingtung, Taiwan
| | - Inn-Ho Tsai
- Institute of Biological Chemistry, Academia Sinica, Taipei, Taiwan; Institute of Biochemical Sciences, National Taiwan University, Taipei, Taiwan.
| |
Collapse
|
2
|
Nowlan JP, Sies AN, Britney SR, Cameron ADS, Siah A, Lumsden JS, Russell S. Genomics of Tenacibaculum Species in British Columbia, Canada. Pathogens 2023; 12:pathogens12010101. [PMID: 36678448 PMCID: PMC9864904 DOI: 10.3390/pathogens12010101] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2022] [Revised: 12/30/2022] [Accepted: 01/04/2023] [Indexed: 01/11/2023] Open
Abstract
Tenacibaculum is a genus of Gram-negative filamentous bacteria with a cosmopolitan distribution. The research describing Tenacibaculum genomes stems primarily from Norway and Chile due to their impacts on salmon aquaculture. Canadian salmon aquaculture also experiences mortality events related to the presence of Tenacibaculum spp., yet no Canadian Tenacibaculum genomes are publicly available. Ribosomal DNA sequencing of 16S and four species-specific 16S quantitative-PCR assays were used to select isolates cultured from Atlantic salmon with mouthrot in British Columbia (BC), Canada. Ten isolates representing four known and two unknown species of Tenacibaculum were selected for shotgun whole genome sequencing using the Oxford Nanopore's MinION platform. The genome assemblies achieved closed circular chromosomes for seven isolates and long contigs for the remaining three isolates. Average nucleotide identity analysis identified T. ovolyticum, T. maritimum, T. dicentrarchi, two genomovars of T. finnmarkense, and two proposed novel species T. pacificus sp. nov. type strain 18-2881-AT and T. retecalamus sp. nov. type strain 18-3228-7BT. Annotation in most of the isolates predicted putative virulence and antimicrobial resistance genes, most-notably toxins (i.e., hemolysins), type-IX secretion systems, and oxytetracycline resistance. Comparative analysis with the T. maritimum type-strain predicted additional toxins and numerous C-terminal secretion proteins, including an M12B family metalloprotease in the T. maritimum isolates from BC. The genomic prediction of virulence-associated genes provides important targets for studies of mouthrot disease, and the annotation of the antimicrobial resistance genes provides targets for surveillance and diagnosis in veterinary medicine.
Collapse
Affiliation(s)
- Joseph P. Nowlan
- Center for Innovation in Fish Health, Vancouver Island University, Nanaimo, BC V9R 5S5, Canada
- Department of Pathobiology, University of Guelph, Guelph, ON N1G 2W1, Canada
- Correspondence:
| | - Ashton N. Sies
- Department of Biology, University of Regina, Regina, SK S4S 0A2, Canada
- Institute for Microbial Systems and Society, Faculty of Science, University of Regina, Regina, SK S4S 0A2, Canada
| | - Scott R. Britney
- Center for Innovation in Fish Health, Vancouver Island University, Nanaimo, BC V9R 5S5, Canada
- Department of Pathobiology, University of Guelph, Guelph, ON N1G 2W1, Canada
| | - Andrew D. S. Cameron
- Department of Biology, University of Regina, Regina, SK S4S 0A2, Canada
- Institute for Microbial Systems and Society, Faculty of Science, University of Regina, Regina, SK S4S 0A2, Canada
| | - Ahmed Siah
- BC Center for Aquatic Health Sciences, Campbell River, BC V9W 2C2, Canada
| | - John S. Lumsden
- Department of Pathobiology, University of Guelph, Guelph, ON N1G 2W1, Canada
| | - Spencer Russell
- Center for Innovation in Fish Health, Vancouver Island University, Nanaimo, BC V9R 5S5, Canada
| |
Collapse
|
3
|
Cavalcante JS, Borges da Silva WRG, de Oliveira LA, Brito IMC, Muller KS, J Vidal IS, Dos Santos LD, Jorge RJB, Almeida C, de Lima Bicho C. Blood plasma proteome alteration after local tissue damage induced by Bothrops erythromelas snake venom in mice. J Proteomics 2022; 269:104742. [PMID: 36174952 DOI: 10.1016/j.jprot.2022.104742] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2022] [Revised: 09/03/2022] [Accepted: 09/22/2022] [Indexed: 11/26/2022]
Abstract
Snakes of the genus Bothrops are responsible the most snakebites in the Brazil, causing a diverse and complex pathophysiological condition. Bothrops erythromelas is the main specie of medical relevance found in the Caatinga from the Brazilian Northeast region. The pathophysiological effects involving B. erythromelas snakebite as well as the organism reaction in response to this envenomation are not so explored. Thus, edema was induced in mice paws using 2.5 μg or 5.0 μg of B. erythromelas venom, and the percentage of edema was measured. Plasma was collected 30 minutes after the envenomation-induced in mice and analyzed by mass spectrometry. It was identified a total of 112 common plasma proteins differentially abundant among experimental groups, which are involved with the complement system and coagulation cascades, oxidative stress, neutrophil degranulation, platelets degranulation and inflammatory response. Apolipoprotein A1 (Apoa), serum amyloid protein A-4 (Saa4), adiponectin (Adipoq) showed up-regulated in mice plasma after injection of venom, while fibulin (Fbln1), factor XII (F12) and vitamin K-dependent protein Z (Proz) showed down-regulated. The results indicate a protein pattern of thrombo-inflammation to the B. erythromelas snakebite, evidencing potential biomarkers for monitoring this snakebite, new therapeutic targets and its correlations with the degree of envenomation once showed modulations in the abundance among the different groups according to the amount of venom injected into the mice.
Collapse
Affiliation(s)
- Joeliton S Cavalcante
- Graduate Program in Tropical Diseases, Botucatu Medical School (FMB), São Paulo University (UNESP - Univ Estadual Paulista), Botucatu, São Paulo, Brazil.
| | - Weslley Ruan G Borges da Silva
- Department of Biology, Center of Biological and Health Sciences, Paraíba State University (UEPB), Campina Grande, Paraíba, Brazil
| | - Laudicéia Alves de Oliveira
- Graduate Program in Tropical Diseases, Botucatu Medical School (FMB), São Paulo University (UNESP - Univ Estadual Paulista), Botucatu, São Paulo, Brazil
| | - Ingrid Mayara C Brito
- Graduate Program in Tropical Diseases, Botucatu Medical School (FMB), São Paulo University (UNESP - Univ Estadual Paulista), Botucatu, São Paulo, Brazil
| | - Kevin S Muller
- Institute of Biosciences, São Paulo University (UNESP - Univ Estadual Paulista), Botucatu, São Paulo, Brazil
| | - Ivynna Suellen J Vidal
- Graduate Program in Translational Medicine, Drug Research and Development Center, Federal University of Ceará (UFC), Fortaleza, Ceará, Brazil
| | - Lucilene Delazari Dos Santos
- Graduate Program in Tropical Diseases, Botucatu Medical School (FMB), São Paulo University (UNESP - Univ Estadual Paulista), Botucatu, São Paulo, Brazil; Biotechnology Institute (IBTEC), São Paulo University (UNESP - Univ Estadual Paulista), Botucatu, São Paulo, Brazil
| | - Roberta Jeane Bezerra Jorge
- Drug Research and Development Center, Federal University of Ceará (UFC), Fortaleza, Ceará, Brazil; Department of Physiology and Pharmacology, School of Medicine, Federal University of Ceará (UFC), Fortaleza, Ceará, Brazil
| | - Cayo Almeida
- Center of Mathematics, Computing Sciences and Cognition, Federal University of ABC, São Paulo, SP, Brazil
| | - Carla de Lima Bicho
- Department of Biology, Center of Biological and Health Sciences, Paraíba State University (UEPB), Campina Grande, Paraíba, Brazil
| |
Collapse
|
4
|
Chiang LC, Chien KY, Su HY, Chen YC, Mao YC, Wu WG. Comparison of Protein Variation in Protobothrops mucrosquamatus Venom between Northern and Southeast Taiwan and Association with Human Envenoming Effects. Toxins (Basel) 2022; 14:toxins14090643. [PMID: 36136582 PMCID: PMC9501293 DOI: 10.3390/toxins14090643] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Revised: 09/13/2022] [Accepted: 09/16/2022] [Indexed: 12/03/2022] Open
Abstract
Reports of bite from Protobothrops mucrosquamatus (Pmu) are frequent in Taiwan, and its wide-spread distribution and diverse habitats drove us to investigate its envenoming effects and relevant venom variations. We used reversed-phase high-performance liquid chromatography and mass spectrometry to analyze 163 Pmu venom samples collected from northern and southeastern Taiwan. Twenty-two major protein fractions were separated and analyzed, and their contents were determined semi-quantitatively. The results showed that despite the trivial differences in the protein family, there is an existing variation in acidic phospholipases A2s, serine proteinases, metalloproteinases, C-type lectin-like proteins, and other less abundant components in the Pmu venoms. Moreover, clinical manifestations of 209 Pmu envenomed patients hospitalized in northern or southeastern Taiwan revealed significant differences in local symptoms, such as ecchymosis and blistering. The mechanism of these local effects and possibly relevant venom components were examined. Further analysis showed that certain venom components with inter-population variation might work alone or synergistically with others to aggravate the local effects. Therefore, our findings of the venom variation may help one to improve antivenom production and better understand and manage Pmu bites.
Collapse
Affiliation(s)
- Liao-Chun Chiang
- College of Life Sciences, National Tsing Hua University, Hsinchu City 300, Taiwan
| | - Kun-Yi Chien
- Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Taoyuan County 333, Taiwan
- Clinical Proteomics Core Laboratory, Chang Gung Memorial Hospital, Taoyuan County 333, Taiwan
- Department of Biochemistry and Molecular Biology, College of Medicine, Chang Gung University, Taoyuan County 333, Taiwan
| | - Hung-Yuan Su
- Department of Emergency Medicine, E-Da Hospital, Kaohsiung County 824, Taiwan
- The School of Chinese Medicine for Post Baccalaureate, I-Shou University, Kaohsiung County 840, Taiwan
- Department of Safety, Health and Environmental Engineering, National Kaohsiung University of Science and Technology, Kaohsiung County 811, Taiwan
| | - Yen-Chia Chen
- Department of Emergency Medicine, Taipei Veterans General Hospital, Taipei City 112, Taiwan
- School of Medicine, National Yang Ming Chiao Tung University, Taipei City 112, Taiwan
- Department of Emergency Medicine, National Defense Medical Center, Taipei City 114, Taiwan
| | - Yan-Chiao Mao
- Department of Emergency Medicine, National Defense Medical Center, Taipei City 114, Taiwan
- Division of Clinical Toxicology, Department of Emergency Medicine, Taichung Veterans General Hospital, Taichung City 407, Taiwan
- College of Medicine, National Chung Hsing University, Taichung City 402, Taiwan
- Correspondence: (Y.-C.M.); (W.-G.W.)
| | - Wen-Guey Wu
- College of Life Sciences, National Tsing Hua University, Hsinchu City 300, Taiwan
- Correspondence: (Y.-C.M.); (W.-G.W.)
| |
Collapse
|
5
|
Leonel TB, Gabrili JJM, Squaiella-Baptistão CC, Woodruff TM, Lambris JD, Tambourgi DV. Bothrops jararaca Snake Venom Inflammation Induced in Human Whole Blood: Role of the Complement System. Front Immunol 2022; 13:885223. [PMID: 35720304 PMCID: PMC9201114 DOI: 10.3389/fimmu.2022.885223] [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: 02/27/2022] [Accepted: 03/21/2022] [Indexed: 11/13/2022] Open
Abstract
The clinical manifestations of envenomation by Bothrops species are complex and characterized by prominent local effects that can progress to tissue loss, physical disability, or amputation. Systemic signs can also occur, such as hemorrhage, coagulopathy, shock, and acute kidney failure. The rapid development of local clinical manifestations is accompanied by the presence of mediators of the inflammatory process originating from tissues damaged by the bothropic venom. Considering the important role that the complement system plays in the inflammatory response, in this study, we analyzed the action of Bothrops jararaca snake venom on the complement system and cell surface receptors involved in innate immunity using an ex vivo human whole blood model. B. jararaca venom was able to induce activation of the complement system in the human whole blood model and promoted a significant increase in the production of anaphylatoxins C3a/C3a-desArg, C4a/C4a-desArg, C5a/C5a-desArg and sTCC. In leukocytes, the venom of B. jararaca reduced the expression of CD11b, CD14 and C5aR1. Inhibition of the C3 component by Cp40, an inhibitor of C3, resulted in a reduction of C3a/C3a-desArg, C5a/C5a-desArg and sTCC to basal levels in samples stimulated with the venom. Exposure to B. jararaca venom induced the production of inflammatory cytokines and chemokines such as TNF-α, IL-8/CXCL8, MCP-1/CCL2 and MIG/CXCL9 in the human whole blood model. Treatment with Cp40 promoted a significant reduction in the production of TNF-α, IL-8/CXCL8 and MCP-1/CCL2. C5aR1 inhibition with PMX205 also promoted a reduction of TNF-α and IL-8/CXCL8 to basal levels in the samples stimulated with venom. In conclusion, the data presented here suggest that the activation of the complement system promoted by the venom of the snake B. jararaca in the human whole blood model significantly contributes to the inflammatory process. The control of several inflammatory parameters using Cp40, an inhibitor of the C3 component, and PMX205, a C5aR1 antagonist, indicates that complement inhibition may represent a potential therapeutic tool in B. jararaca envenoming.
Collapse
Affiliation(s)
| | | | | | - Trent M. Woodruff
- School of Biomedical Sciences, Faculty of Medicine, The University of Queensland, St Lucia, QLD, Australia
| | - John D. Lambris
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | | |
Collapse
|
6
|
Cavalcante JDS, de Almeida CAS, Clasen MA, da Silva EL, de Barros LC, Marinho AD, Rossini BC, Marino CL, Carvalho PC, Jorge RJB, Dos Santos LD. A fingerprint of plasma proteome alteration after local tissue damage induced by Bothrops leucurus snake venom in mice. J Proteomics 2022; 253:104464. [PMID: 34954398 DOI: 10.1016/j.jprot.2021.104464] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2021] [Revised: 11/30/2021] [Accepted: 12/19/2021] [Indexed: 12/21/2022]
Abstract
Bothrops spp. is responsible for about 70% of snakebites in Brazil, causing a diverse and complex pathophysiological condition. Bothrops leucurus is the main species of medical relevance found in the Atlantic coast in the Brazilian Northeast region. The pathophysiological effects involved B. leucurus snakebite as well as the organism's reaction in response to this envenoming, it has not been explored yet. Thus, edema was induced in mice paw using 1.2, 2.5, and 5.0 μg of B. leucurus venom, the percentage of edema was measured 30 min after injection and the blood plasma was collected and analyzed by shotgun proteomic strategy. We identified 80 common plasma proteins with differential abundance among the experimental groups and we can understand the early aspects of this snake envenomation, regardless of the suggestive severity of an ophidian accident. The results showed B. leucurus venom triggers a thromboinflammation scenario where family's proteins of the Serpins, Apolipoproteins, Complement factors and Component subunits, Cathepsins, Kinases, Oxidoreductases, Proteases inhibitors, Proteases, Collagens, Growth factors are related to inflammation, complement and coagulation systems, modulators platelets and neutrophils, lipid and retinoid metabolism, oxidative stress and tissue repair. Our findings set precedents for future studies in the area of early diagnosis and/or treatment of snakebites. SIGNIFICANCE: The physiopathological effects that the snake venoms can cause have been investigated through classical and reductionist tools, which allowed, so far, the identification of action mechanisms of individual components associated with specific tissue damage. The currently incomplete limitations of this knowledge must be expanded through new approaches, such as proteomics, which may represent a big leap in understanding the venom-modulated pathological process. The exploration of the complete protein set that suffer modifications by the simultaneous action of multiple toxins, provides a map of the establishment of physiopathological phenotypes, which favors the identification of multiple toxin targets, that may or may not act in synergy, as well as favoring the discovery of biomarkers and therapeutic targets for manifestations that are not neutralized by the antivenom.
Collapse
Affiliation(s)
- Joeliton Dos Santos Cavalcante
- Graduate Program in Tropical Diseases, Botucatu Medical School (FMB), São Paulo State University (UNESP), Botucatu, SP, Brazil
| | | | - Milan Avila Clasen
- Laboratory for Structural and Computational Proteomics, ICC, Oswaldo Cruz Foundation (FIOCRUZ), Curitiba, PR, Brazil
| | - Emerson Lucena da Silva
- Drug Research and Development Center, Federal University of Ceará (UFC), Fortaleza, CE, Brazil; Department of Physiology and Pharmacology, School of Medicine, Federal University of Ceará (UFC), Fortaleza, CE, Brazil
| | - Luciana Curtolo de Barros
- Center for the Study of Venoms and Venomous Animals (CEVAP), São Paulo State University (UNESP), Botucatu, SP, Brazil
| | - Aline Diogo Marinho
- Drug Research and Development Center, Federal University of Ceará (UFC), Fortaleza, CE, Brazil; Department of Physiology and Pharmacology, School of Medicine, Federal University of Ceará (UFC), Fortaleza, CE, Brazil
| | - Bruno Cesar Rossini
- Biotechnology Institute (IBTEC), São Paulo State University (UNESP), Botucatu, SP, Brazil; Department of Chemical and Biological Sciences, São Paulo State University (UNESP), Botucatu, SP, Brazil
| | - Celso Luís Marino
- Biotechnology Institute (IBTEC), São Paulo State University (UNESP), Botucatu, SP, Brazil; Department of Chemical and Biological Sciences, São Paulo State University (UNESP), Botucatu, SP, Brazil
| | - Paulo Costa Carvalho
- Laboratory for Structural and Computational Proteomics, ICC, Oswaldo Cruz Foundation (FIOCRUZ), Curitiba, PR, Brazil
| | - Roberta Jeane Bezerra Jorge
- Drug Research and Development Center, Federal University of Ceará (UFC), Fortaleza, CE, Brazil; Department of Physiology and Pharmacology, School of Medicine, Federal University of Ceará (UFC), Fortaleza, CE, Brazil
| | - Lucilene Delazari Dos Santos
- Graduate Program in Tropical Diseases, Botucatu Medical School (FMB), São Paulo State University (UNESP), Botucatu, SP, Brazil; Biotechnology Institute (IBTEC), São Paulo State University (UNESP), Botucatu, SP, Brazil.
| |
Collapse
|
7
|
Janovits PM, Leiguez E, Portas V, Teixeira C. A Metalloproteinase Induces an Inflammatory Response in Preadipocytes with the Activation of COX Signalling Pathways and Participation of Endogenous Phospholipases A 2. Biomolecules 2021; 11:921. [PMID: 34206390 PMCID: PMC8301905 DOI: 10.3390/biom11070921] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2021] [Revised: 06/10/2021] [Accepted: 06/11/2021] [Indexed: 12/12/2022] Open
Abstract
Matrix metalloproteinases (MMPs) are proteolytic enzymes that have been associated with the pathogenesis of inflammatory diseases and obesity. Adipose tissue in turn is an active endocrine organ capable of secreting a range of proinflammatory mediators with autocrine and paracrine properties, which contribute to the inflammation of adipose tissue and adjacent tissues. However, the potential inflammatory effects of MMPs in adipose tissue cells are still unknown. This study investigates the effects of BmooMPα-I, a single-domain snake venom metalloproteinase (SVMP), in activating an inflammatory response by 3T3-L1 preadipocytes in culture, focusing on prostaglandins (PGs), cytokines, and adipocytokines biosynthesis and mechanisms involved in prostaglandin E2 (PGE2) release. The results show that BmooMPα-I induced the release of PGE2, prostaglandin I2 (PGI2), monocyte chemoattractant protein-1 (MCP-1), and adiponectin by preadipocytes. BmooMPα-I-induced PGE2 biosynthesis was dependent on group-IIA-secreted phospholipase A2 (sPLA2-IIA), cytosolic phospholipase A2-α (cPLA2-α), and cyclooxygenase (COX)-1 and -2 pathways. Moreover, BmooMPα-I upregulated COX-2 protein expression but not microsomal prostaglandin E synthase-1 (mPGES-1) expression. In addition, we demonstrate that the enzymatic activity of BmooMPα-I is essential for the activation of prostanoid synthesis pathways in preadipocytes. These data highlight preadipocytes as important targets for metalloproteinases and provide new insights into the contribution of these enzymes to the inflammation of adipose tissue and tissues adjacent to it.
Collapse
Affiliation(s)
- Priscila Motta Janovits
- Laboratório de Farmacologia, Instituto Butantan, São Paulo 05503-900, Brazil;
- Centre of Excellence in New Target Discovery (CENTD), Instituto Butantan, São Paulo 05503-900, Brazil;
| | - Elbio Leiguez
- Laboratório de Farmacologia, Instituto Butantan, São Paulo 05503-900, Brazil;
- Centre of Excellence in New Target Discovery (CENTD), Instituto Butantan, São Paulo 05503-900, Brazil;
| | - Viviane Portas
- Centre of Excellence in New Target Discovery (CENTD), Instituto Butantan, São Paulo 05503-900, Brazil;
- Laboratório de Desenvolvimento e Inovação, Instituto Butantan, São Paulo 05503-900, Brazil
| | - Catarina Teixeira
- Laboratório de Farmacologia, Instituto Butantan, São Paulo 05503-900, Brazil;
- Centre of Excellence in New Target Discovery (CENTD), Instituto Butantan, São Paulo 05503-900, Brazil;
| |
Collapse
|
8
|
Silva de França F, Villas-Boas IM, Cogliati B, Woodruff TM, Reis EDS, Lambris JD, Tambourgi DV. C5a-C5aR1 Axis Activation Drives Envenomation Immunopathology by the Snake Naja annulifera. Front Immunol 2021; 12:652242. [PMID: 33936074 PMCID: PMC8082402 DOI: 10.3389/fimmu.2021.652242] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2021] [Accepted: 03/22/2021] [Indexed: 12/03/2022] Open
Abstract
Systemic complement activation drives a plethora of pathological conditions, but its role in snake envenoming remains obscure. Here, we explored complement's contribution to the physiopathogenesis of Naja annulifera envenomation. We found that N. annulifera venom promoted the generation of C3a, C4a, C5a, and the soluble Terminal Complement Complex (sTCC) mediated by the action of snake venom metalloproteinases. N. annulifera venom also induced the release of lipid mediators and chemokines in a human whole-blood model. This release was complement-mediated, since C3/C3b and C5a Receptor 1 (C5aR1) inhibition mitigated the effects. In an experimental BALB/c mouse model of envenomation, N. annulifera venom promoted lipid mediator and chemokine production, neutrophil influx, and swelling at the injection site in a C5a-C5aR1 axis-dependent manner. N. annulifera venom induced systemic complementopathy and increased interleukin and chemokine production, leukocytosis, and acute lung injury (ALI). Inhibition of C5aR1 with the cyclic peptide antagonist PMX205 rescued mice from these systemic reactions and abrogated ALI development. These data reveal hitherto unrecognized roles for complement in envenomation physiopathogenesis, making complement an interesting therapeutic target in envenomation by N. annulifera and possibly by other snake venoms.
Collapse
Affiliation(s)
| | | | - Bruno Cogliati
- Department of Pathology, School of Veterinary Medicine and Animal Science, University of São Paulo, São Paulo, Brazil
| | - Trent M. Woodruff
- Neuroinflammation Laboratory, School of Biomedical Sciences, The University of Queensland, St Lucia, QLD, Australia
| | - Edimara da Silva Reis
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - John D. Lambris
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | | |
Collapse
|
9
|
Minutti-Zanella C, Gil-Leyva EJ, Vergara I. Immunomodulatory properties of molecules from animal venoms. Toxicon 2021; 191:54-68. [PMID: 33417946 DOI: 10.1016/j.toxicon.2020.12.018] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2020] [Revised: 12/02/2020] [Accepted: 12/30/2020] [Indexed: 10/22/2022]
Abstract
The immune system can amplify or decrease the strength of its response when it is stimulated by chemical or biological substances that act as immunostimulators, immunosuppressants, or immunoadjuvants. Immunomodulation is a progressive approach to treat a diversity of pathologies with promising results, including autoimmune disorders and cancer. Animal venoms are a mixture of chemical compounds that include proteins, peptides, amines, salts, polypeptides, enzymes, among others, which produce the toxic effect. Since the discovery of captopril in the early 1980s, other components from snakes, spiders, scorpions, and marine animal venoms have been demonstrated to be useful for treating several human diseases. The valuable progress in fields such as venomics, molecular biology, biotechnology, immunology, and others has been crucial to understanding the interaction of toxins with the immune system and its application on immune pathologies. More in-depth knowledge of venoms' components and multi-disciplinary studies could facilitate their transformation into effective novel immunotherapies. This review addresses advances and research of molecules from venoms that have immunomodulatory properties.
Collapse
Affiliation(s)
- C Minutti-Zanella
- Departamento de Ciencias Químico-Biológicas, Universidad de Las Américas Puebla, ExHda. Sta. Catarina Mártir s/n, San Andrés Cholula, 72820, Puebla, Mexico
| | - E J Gil-Leyva
- Departamento de Ciencias Químico-Biológicas, Universidad de Las Américas Puebla, ExHda. Sta. Catarina Mártir s/n, San Andrés Cholula, 72820, Puebla, Mexico
| | - I Vergara
- Departamento de Ciencias Químico-Biológicas, Universidad de Las Américas Puebla, ExHda. Sta. Catarina Mártir s/n, San Andrés Cholula, 72820, Puebla, Mexico.
| |
Collapse
|
10
|
Cleavage of proteoglycans, plasma proteins and the platelet-derived growth factor receptor in the hemorrhagic process induced by snake venom metalloproteinases. Sci Rep 2020; 10:12912. [PMID: 32737331 PMCID: PMC7395112 DOI: 10.1038/s41598-020-69396-y] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2020] [Accepted: 07/07/2020] [Indexed: 12/13/2022] Open
Abstract
Envenoming by viperid snakes results in a complex pattern of tissue damage, including hemorrhage, which in severe cases may lead to permanent sequelae. Snake venom metalloproteinases (SVMPs) are main players in this pathogenesis, acting synergistically upon different mammalian proteomes. Hemorrhagic Factor 3 (HF3), a P-III class SVMP from Bothrops jararaca, induces severe local hemorrhage at pmol doses in a murine model. Our hypothesis is that in a complex scenario of tissue damage, HF3 triggers proteolytic cascades by acting on a partially known substrate repertoire. Here, we focused on the hypothesis that different proteoglycans, plasma proteins, and the platelet derived growth factor receptor (PDGFR) could be involved in the HF3-induced hemorrhagic process. In surface plasmon resonance assays, various proteoglycans were demonstrated to interact with HF3, and their incubation with HF3 showed degradation or limited proteolysis. Likewise, Western blot analysis showed in vivo degradation of biglycan, decorin, glypican, lumican and syndecan in the HF3-induced hemorrhagic process. Moreover, antithrombin III, complement components C3 and C4, factor II and plasminogen were cleaved in vitro by HF3. Notably, HF3 cleaved PDGFR (alpha and beta) and PDGF in vitro, while both receptor forms were detected as cleaved in vivo in the hemorrhagic process induced by HF3. These findings outline the multifactorial character of SVMP-induced tissue damage, including the transient activation of tissue proteinases, and underscore for the first time that endothelial glycocalyx proteoglycans and PDGFR are targets of SVMPs in the disruption of microvasculature integrity and generation of hemorrhage.
Collapse
|
11
|
Teixeira C, Fernandes CM, Leiguez E, Chudzinski-Tavassi AM. Inflammation Induced by Platelet-Activating Viperid Snake Venoms: Perspectives on Thromboinflammation. Front Immunol 2019; 10:2082. [PMID: 31572356 PMCID: PMC6737392 DOI: 10.3389/fimmu.2019.02082] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2019] [Accepted: 08/16/2019] [Indexed: 01/01/2023] Open
Abstract
Envenomation by viperid snakes is characterized by systemic thrombotic syndrome and prominent local inflammation. To date, the mechanisms underlying inflammation and blood coagulation induced by Viperidae venoms have been viewed as distinct processes. However, studies on the mechanisms involved in these processes have revealed several factors and signaling molecules that simultaneously act in both the innate immune and hemostatic systems, suggesting an overlap between both systems during viper envenomation. Moreover, distinct classes of venom toxins involved in these effects have also been identified. However, the interplay between inflammation and hemostatic alterations, referred as to thromboinflammation, has never been addressed in the investigation of viper envenomation. Considering that platelets are important targets of viper snake venoms and are critical for the process of thromboinflammation, in this review, we summarize the inflammatory effects and mechanisms induced by viper snake venoms, particularly from the Bothrops genus, which strongly activate platelet functions and highlight selected venom components (metalloproteases and C-type lectins) that both stimulate platelet functions and exhibit pro-inflammatory activities, thus providing insights into the possible role(s) of thromboinflammation in viper envenomation.
Collapse
Affiliation(s)
- Catarina Teixeira
- Laboratory of Pharmacology, Butantan Institute, São Paulo, Brazil.,Centre of Excellence in New Target Discovery, Butantan Institute, São Paulo, Brazil
| | - Cristina Maria Fernandes
- Laboratory of Pharmacology, Butantan Institute, São Paulo, Brazil.,Centre of Excellence in New Target Discovery, Butantan Institute, São Paulo, Brazil
| | - Elbio Leiguez
- Laboratory of Pharmacology, Butantan Institute, São Paulo, Brazil.,Centre of Excellence in New Target Discovery, Butantan Institute, São Paulo, Brazil
| | - Ana Marisa Chudzinski-Tavassi
- Centre of Excellence in New Target Discovery, Butantan Institute, São Paulo, Brazil.,Laboratory of Molecular Biology, Butantan Institute, São Paulo, Brazil
| |
Collapse
|
12
|
Ibiapina HNS, Costa AG, Sachett JAG, Silva IM, Tarragô AM, Neves JCF, Kerr MWA, Santana MF, Martins-Filho OA, Lacerda MVG, Ferreira LCL, Malheiro A, Monteiro WM. An Immunological Stairway to Severe Tissue Complication Assembly in Bothrops atrox Snakebites. Front Immunol 2019; 10:1882. [PMID: 31474982 PMCID: PMC6705225 DOI: 10.3389/fimmu.2019.01882] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2019] [Accepted: 07/24/2019] [Indexed: 01/05/2023] Open
Abstract
Snakebites are a serious public health problem and, in the Amazon, the Bothrops atrox snake is the most frequent cause of envenomation. B. atrox venom (BaV) causes pathophysiological changes with intense, local inflammatory processes, such as severe tissue complication (STC). However, mechanisms associated with the inflammatory process in humans are still poorly understood. Thus, in this study, we sought to describe the profile of local and systemic immunological soluble molecules in Bothrops envenomation patients treated at a specialist tertiary healthcare unit in the Brazilian Amazon. An analytical and prospective study was performed with patients who had snakebites with different clinical outcomes (STC and Mild Tissue Complication-MTC) using venous blood and blister exudate in order to measure immunological soluble molecules present in the response process. Twenty STC patients and 20 MTC patients were eligible for the study. In addition, 20 healthy donors (HD) who had never been bitten by a snake were used as controls. The biomarkers CXCL-8, CCL-5, CXCL-9, CCL-2 and CXCL-10; C3a, C4a, and C5a; IL-1, IL-2, IL-4, IL-5, IL-6, IL-10, TNF, IFN-γ and IL-17A were quantified using flow cytometry and ELISA. The circulating response profile differs between the studied groups, with MTC patients presenting a mixed profile and STC patients presenting a more polarized profile for Th1 response. In addition, individuals who develop STC have a more intense local immune response, because the tissue response differs from the circulating immunological soluble molecules and presents Th1/Th2/Th17 response polarization. Furthermore, these results suggest that CCL-2 and CXCL-10 are biomarkers for STC and the response profile they assume against Bothrops snakebite should reflect in the clinical practice for the patient.
Collapse
Affiliation(s)
- Hiochelson Najibe Santos Ibiapina
- Programa de Pós-Graduação em Medicina Tropical, Universidade do Estado do Amazonas (UEA), Manaus, Brazil.,Instituto de Pesquisa Clínica Carlos Borborema, Fundação de Medicina Tropical Doutor Heitor Vieira Dourado (FMT-HVD), Manaus, Brazil
| | - Allyson Guimarães Costa
- Programa de Pós-Graduação em Medicina Tropical, Universidade do Estado do Amazonas (UEA), Manaus, Brazil.,Instituto de Pesquisa Clínica Carlos Borborema, Fundação de Medicina Tropical Doutor Heitor Vieira Dourado (FMT-HVD), Manaus, Brazil.,Programa de Pós-Graduação em Imunologia Básica e Aplicada, Instituto de Ciências Biológicas, Universidade Federal do Amazonas (UFAM), Manaus, Brazil.,Diretoria de Ensino e Pesquisa, Fundação Hospitalar de Hematologia e Hemoterapia do Amazonas (HEMOAM), Manaus, Brazil.,Programa de Pós-Graduação em Ciências Aplicadas à Hematologia, Universidade do Estado do Amazonas (UEA), Manaus, Brazil
| | - Jacqueline Almeida Gonçalves Sachett
- Programa de Pós-Graduação em Medicina Tropical, Universidade do Estado do Amazonas (UEA), Manaus, Brazil.,Instituto de Pesquisa Clínica Carlos Borborema, Fundação de Medicina Tropical Doutor Heitor Vieira Dourado (FMT-HVD), Manaus, Brazil
| | - Iran Mendonça Silva
- Instituto de Pesquisa Clínica Carlos Borborema, Fundação de Medicina Tropical Doutor Heitor Vieira Dourado (FMT-HVD), Manaus, Brazil
| | - Andréa Monteiro Tarragô
- Programa de Pós-Graduação em Imunologia Básica e Aplicada, Instituto de Ciências Biológicas, Universidade Federal do Amazonas (UFAM), Manaus, Brazil.,Diretoria de Ensino e Pesquisa, Fundação Hospitalar de Hematologia e Hemoterapia do Amazonas (HEMOAM), Manaus, Brazil
| | - Juliana Costa Ferreira Neves
- Programa de Pós-Graduação em Medicina Tropical, Universidade do Estado do Amazonas (UEA), Manaus, Brazil.,Instituto de Pesquisa Clínica Carlos Borborema, Fundação de Medicina Tropical Doutor Heitor Vieira Dourado (FMT-HVD), Manaus, Brazil
| | - Marllon Wendell Athaydes Kerr
- Diretoria de Ensino e Pesquisa, Fundação Hospitalar de Hematologia e Hemoterapia do Amazonas (HEMOAM), Manaus, Brazil.,Programa de Pós-Graduação em Ciências Aplicadas à Hematologia, Universidade do Estado do Amazonas (UEA), Manaus, Brazil
| | - Monique Freire Santana
- Programa de Pós-Graduação em Medicina Tropical, Universidade do Estado do Amazonas (UEA), Manaus, Brazil.,Instituto de Pesquisa Clínica Carlos Borborema, Fundação de Medicina Tropical Doutor Heitor Vieira Dourado (FMT-HVD), Manaus, Brazil
| | - Olindo Assis Martins-Filho
- Grupo Integrado de Pesquisas em Biomarcadores de Diagnóstico e Monitoração, Centro de Pesquisas René Rachou, Fundação Oswaldo Cruz (FIOCRUZ), Belo Horizonte, Brazil
| | - Marcus Vinícius Guimarães Lacerda
- Programa de Pós-Graduação em Medicina Tropical, Universidade do Estado do Amazonas (UEA), Manaus, Brazil.,Instituto de Pesquisa Clínica Carlos Borborema, Fundação de Medicina Tropical Doutor Heitor Vieira Dourado (FMT-HVD), Manaus, Brazil.,Instituto de Pesquisas Leônidas & Maria Deane, FIOCRUZ-Amazônia, Manaus, Brazil
| | - Luiz Carlos Lima Ferreira
- Programa de Pós-Graduação em Medicina Tropical, Universidade do Estado do Amazonas (UEA), Manaus, Brazil.,Instituto de Pesquisa Clínica Carlos Borborema, Fundação de Medicina Tropical Doutor Heitor Vieira Dourado (FMT-HVD), Manaus, Brazil
| | - Adriana Malheiro
- Programa de Pós-Graduação em Medicina Tropical, Universidade do Estado do Amazonas (UEA), Manaus, Brazil.,Programa de Pós-Graduação em Imunologia Básica e Aplicada, Instituto de Ciências Biológicas, Universidade Federal do Amazonas (UFAM), Manaus, Brazil.,Diretoria de Ensino e Pesquisa, Fundação Hospitalar de Hematologia e Hemoterapia do Amazonas (HEMOAM), Manaus, Brazil.,Programa de Pós-Graduação em Ciências Aplicadas à Hematologia, Universidade do Estado do Amazonas (UEA), Manaus, Brazil
| | - Wuelton Marcelo Monteiro
- Programa de Pós-Graduação em Medicina Tropical, Universidade do Estado do Amazonas (UEA), Manaus, Brazil.,Instituto de Pesquisa Clínica Carlos Borborema, Fundação de Medicina Tropical Doutor Heitor Vieira Dourado (FMT-HVD), Manaus, Brazil
| |
Collapse
|
13
|
Luchini LSG, Pidde G, Squaiella-Baptistão CC, Tambourgi DV. Complement System Inhibition Modulates the Pro-Inflammatory Effects of a Snake Venom Metalloproteinase. Front Immunol 2019; 10:1137. [PMID: 31231362 PMCID: PMC6558526 DOI: 10.3389/fimmu.2019.01137] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2019] [Accepted: 05/07/2019] [Indexed: 12/20/2022] Open
Abstract
Envenomation by Bothrops snakes causes prominent local effects, including pain, oedema, local bleeding, blistering and necrosis, and systemic manifestations, such as hemorrhage, hypotension, shock and acute renal failure. These snake venoms are able to activate the complement system and induce the generation of anaphylatoxins, whose mechanisms include the direct cleavage of complement components by snake venom metalloproteinases and serine proteinases present in the venoms. A metalloproteinase able to activate the three complement pathways and generate active anaphylatoxins, named C-SVMP, was purified from the venom of Bothrops pirajai. Considering the inflammatory nature of Bothrops venoms and the complement-activation property of C-SVMP, in the present work, we investigated the inflammatory effects of C-SVMP in a human whole blood model. The role of the complement system in the inflammatory process and its modulation by the use of compstatin were also investigated. C-SVMP was able to activate the complement system in the whole blood model, generating C3a/C3a desArg, C5a/C5a desArg and SC5b-9. This protein was able to promote an increase in the expression of CD11b, CD14, C3aR, C5aR1, TLR2, and TLR4 markers in leukocytes. Inhibition of component C3 by compstatin significantly reduced the production of anaphylatoxins and the Terminal Complement Complex (TCC) in blood plasma treated with the toxin, as well as the expression of CD11b, C3aR, and C5aR on leukocytes. C-SVMP was able to induce increased production of the cytokines IL-1β and IL-6 and the chemokines CXCL8/IL-8, CCL2/MCP-1, and CXCL9/MIG in the human whole blood model. The addition of compstatin to the reactions caused a significant reduction in the production of IL-1β, CXCL8/IL-8, and CCL2/MCP-1 in cells treated with C-SVMP. We therefore conclude that C-SVMP is able to activate the complement system, which leads to an increase in the inflammatory process. The data obtained with the use of compstatin indicate that complement inhibition may significantly control the inflammatory process initiated by Bothrops snake venom toxins.
Collapse
Affiliation(s)
| | - Giselle Pidde
- Immunochemistry Laboratory, Butantan Institute, São Paulo, Brazil
| | | | | |
Collapse
|
14
|
Lin Z, Cheng Y, Wang RJ, Du J, Volovych O, Li JC, Hu Y, Lu ZY, Lu Z, Zou Z. A Metalloprotease Homolog Venom Protein From a Parasitoid Wasp Suppresses the Toll Pathway in Host Hemocytes. Front Immunol 2018; 9:2301. [PMID: 30405599 PMCID: PMC6206080 DOI: 10.3389/fimmu.2018.02301] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2018] [Accepted: 09/17/2018] [Indexed: 12/18/2022] Open
Abstract
Parasitoid wasps depend on a variety of maternal virulence factors to ensure successful parasitism. Encapsulation response carried out by host hemocytes is one of the major host immune responses toward limiting endoparasitoid wasp offspring production. We found that VRF1, a metalloprotease homolog venom protein identified from the endoparasitoid wasp, Microplitis mediator, could modulate egg encapsulation in its host, the cotton bollworm, Helicoverpa armigera. Here, we show that the VRF1 proenzyme is cleaved after parasitism, and that the C-terminal fragment containing the catalytic domain enters host hemocytes 6 h post-parasitism. Furthermore, using yeast two-hybrid and pull-down assays, VRF1 is shown to interact with the H. armigera NF-κB factor, Dorsal. We also show that overexpressed of VRF1 in an H. armigera cell line cleaved Dorsal in vivo. Taken together, our results have revealed a novel mechanism by which a component of endoparasitoid wasp venom interferes with the Toll signaling pathway in the host hemocytes.
Collapse
Affiliation(s)
- Zhe Lin
- State Key Laboratory of Integrated Management of Pest Insects and Rodents, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
| | - Yang Cheng
- State Key Laboratory of Integrated Management of Pest Insects and Rodents, Institute of Zoology, Chinese Academy of Sciences, Beijing, China.,College of Life Sciences, University of Chinese Academy of Sciences, Beijing, China
| | - Rui-Juan Wang
- Department of Entomology, College of Plant Protection, Northwest A&F University, Yangling, China
| | - Jie Du
- State Key Laboratory of Integrated Management of Pest Insects and Rodents, Institute of Zoology, Chinese Academy of Sciences, Beijing, China.,College of Life Sciences, University of Chinese Academy of Sciences, Beijing, China
| | - Olga Volovych
- State Key Laboratory of Integrated Management of Pest Insects and Rodents, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
| | - Jian-Cheng Li
- Institute of Plant Protection of Hebei Academy of Agriculture and Forestry Sciences, Baoding, China
| | - Yang Hu
- State Key Laboratory of Integrated Management of Pest Insects and Rodents, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
| | - Zi-Yun Lu
- Institute of Plant Protection of Hebei Academy of Agriculture and Forestry Sciences, Baoding, China
| | - Zhiqiang Lu
- Department of Entomology, College of Plant Protection, Northwest A&F University, Yangling, China
| | - Zhen Zou
- State Key Laboratory of Integrated Management of Pest Insects and Rodents, Institute of Zoology, Chinese Academy of Sciences, Beijing, China.,College of Life Sciences, University of Chinese Academy of Sciences, Beijing, China
| |
Collapse
|
15
|
Shende R, Wong SSW, Rapole S, Beau R, Ibrahim-Granet O, Monod M, Gührs KH, Pal JK, Latgé JP, Madan T, Aimanianda V, Sahu A. Aspergillus fumigatus conidial metalloprotease Mep1p cleaves host complement proteins. J Biol Chem 2018; 293:15538-15555. [PMID: 30139746 PMCID: PMC6177592 DOI: 10.1074/jbc.ra117.001476] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2017] [Revised: 08/02/2018] [Indexed: 12/30/2022] Open
Abstract
Innate immunity in animals including humans encompasses the complement system, which is considered an important host defense mechanism against Aspergillus fumigatus, one of the most ubiquitous opportunistic human fungal pathogens. Previously, it has been shown that the alkaline protease Alp1p secreted from A. fumigatus mycelia degrades the complement components C3, C4, and C5. However, it remains unclear how the fungal spores (i.e. conidia) defend themselves against the activities of the complement system immediately after inhalation into the lung. Here, we show that A. fumigatus conidia contain a metalloprotease Mep1p, which is released upon conidial contact with collagen and inactivates all three complement pathways. In particular, Mep1p efficiently inactivated the major complement components C3, C4, and C5 and their activation products (C3a, C4a, and C5a) as well as the pattern-recognition molecules MBL and ficolin-1, either by directly cleaving them or by cleaving them to a form that is further broken down by other proteases of the complement system. Moreover, incubation of Mep1p with human serum significantly inhibited the complement hemolytic activity and conidial opsonization by C3b and their subsequent phagocytosis by macrophages. Together, these results indicate that Mep1p associated with and released from A. fumigatus conidia likely facilitates early immune evasion by disarming the complement defense in the human host.
Collapse
Affiliation(s)
- Rajashri Shende
- From the Complement Biology Laboratory and
- the Dr. D. Y. Patil Biotechnology and Bioinformatics Institute, Dr. D. Y. Patil Vidyapeeth, Tathawade, Pune-411033, India
| | | | - Srikanth Rapole
- Proteomics Laboratory, National Centre for Cell Science, S. P. Pune University Campus, Ganeshkhind, Pune-411007, India
| | | | | | - Michel Monod
- the Service de Dermatologie, Laboratoire de Mycologie, Centre Hospitalier Universitaire Vaudois, 1011 Lausanne, Switzerland
| | - Karl-Heinz Gührs
- the Leibniz Institute on Aging-Fritz Lipmann Institute, Jena-07745, Germany, and
| | - Jayanta Kumar Pal
- the Dr. D. Y. Patil Biotechnology and Bioinformatics Institute, Dr. D. Y. Patil Vidyapeeth, Tathawade, Pune-411033, India
| | | | - Taruna Madan
- the ICMR-National Institute for Research in Reproductive Health, Parel, Mumbai-400012, India
| | | | | |
Collapse
|
16
|
Venom from Bothrops lanceolatus, a Snake Species Native to Martinique, Potently Activates the Complement System. J Immunol Res 2018; 2018:3462136. [PMID: 30116749 PMCID: PMC6079423 DOI: 10.1155/2018/3462136] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2017] [Accepted: 05/30/2018] [Indexed: 12/22/2022] Open
Abstract
Bothrops lanceolatus snake venom causes systemic thrombotic syndrome but also local inflammation involving extensive oedema, pain, and haemorrhage. Systemic thrombotic syndrome may lead to fatal pulmonary embolism and myocardial and cerebral infarction. Here, we investigated the ability of B. lanceolatus venom to activate the Complement system (C) in order to improve the understanding of venom-induced local inflammation. Data presented show that B. lanceolatus venom is able to activate all C-pathways. In human serum, the venom strongly induced the generation of anaphylatoxins, such as C5a and C4a, and the Terminal Complement complex. The venom also induced cleavage of purified human components C3, C4, and C5, with the production of biologically active C5a. Furthermore, the venom enzymatically inactivated the soluble C-regulator and the C1-inhibitor (C1-INH), and significantly increased the expression of bound C-regulators, such as MCP and CD59, on the endothelial cell membrane. Our observations that B. lanceolatus venom activates the three Complement activation pathways, resulting in anaphylatoxins generation, may suggest that this could play an important role in local inflammatory reaction and systemic thrombosis caused by the venom. Inactivation of C1-INH, which is also an important inhibitor of several coagulation proteins, may also contribute to inflammation and thrombosis. Thus, further in vivo studies may support the idea that therapeutic management of systemic B. lanceolatus envenomation could include the use of Complement inhibitors as adjunct therapy.
Collapse
|
17
|
Jimenez R, Ikonomopoulou MP, Lopez JA, Miles JJ. Immune drug discovery from venoms. Toxicon 2017; 141:18-24. [PMID: 29170055 DOI: 10.1016/j.toxicon.2017.11.006] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2017] [Revised: 11/14/2017] [Accepted: 11/18/2017] [Indexed: 01/22/2023]
Abstract
This review catalogues recent advances in knowledge on venoms as standalone therapeutic agents or as blueprints for drug design, with an emphasis on venom-derived compounds that affects the immune system. We discuss venoms and venom-derived compounds that affect total immune cell numbers, immune cell proliferation, immune cell migration, immune cell phenotype and cytokine secretion. Identifying novel compounds that 'tune' the system, up-regulating the immune response during infectious disease and cancer and down-regulating the immune response during autoimmunity, will greatly expand the tool kit of human immunotherapeutics. Targeting these pathways may also open therapeutic options that alleviate symptoms of envenomation. Finally, combining recent advances in venomics with progress in low cost, high-throughput screening platforms will no doubt yield hundreds of prototype immune modulating compounds in the coming years.
Collapse
Affiliation(s)
- Rocio Jimenez
- Griffith University, School of Natural Sciences, Brisbane, Queensland, Australia; QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
| | - Maria P Ikonomopoulou
- QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia; School of Medicine, The University of Queensland, Brisbane, Australia; Madrid Institute for Advanced Studies (IMDEA) in Food, CEI UAM+CSIC, Madrid, Spain
| | - J Alejandro Lopez
- Griffith University, School of Natural Sciences, Brisbane, Queensland, Australia; QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
| | - John J Miles
- Griffith University, School of Natural Sciences, Brisbane, Queensland, Australia; QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia; School of Medicine, The University of Queensland, Brisbane, Australia; Centre for Biodiscovery and Molecular Development of Therapeutics, AITHM, James Cook University, Cairns, Queensland, Australia; Institute of Infection and Immunity, Cardiff University School of Medicine, Heath Park, Cardiff, United Kingdom.
| |
Collapse
|
18
|
Lodovicho ME, Costa TR, Bernardes CP, Menaldo DL, Zoccal KF, Carone SE, Rosa JC, Pucca MB, Cerni FA, Arantes EC, Tytgat J, Faccioli LH, Pereira-Crott LS, Sampaio SV. Investigating possible biological targets of Bj-CRP, the first cysteine-rich secretory protein (CRISP) isolated from Bothrops jararaca snake venom. Toxicol Lett 2017; 265:156-169. [DOI: 10.1016/j.toxlet.2016.12.003] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2016] [Revised: 11/22/2016] [Accepted: 12/04/2016] [Indexed: 12/13/2022]
|
19
|
Menaldo DL, Bernardes CP, Jacob-Ferreira AL, Nogueira-Santos CG, Casare-Ogasawara TM, Pereira-Crott LS, Sampaio SV. Effects of Bothrops atrox venom and two isolated toxins on the human complement system: Modulation of pathways and generation of anaphylatoxins. Mol Immunol 2016; 80:91-100. [DOI: 10.1016/j.molimm.2016.10.015] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2016] [Revised: 10/18/2016] [Accepted: 10/25/2016] [Indexed: 10/20/2022]
|
20
|
Haemostasis disorders caused by envenomation by Cerastes cerastes and Macrovipera mauritanica vipers. Toxicon 2016; 116:43-8. [PMID: 26724273 DOI: 10.1016/j.toxicon.2015.12.012] [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: 07/27/2015] [Revised: 12/11/2015] [Accepted: 12/22/2015] [Indexed: 11/23/2022]
Abstract
Viper venoms are a real source of proteolytic enzymes causing clotting, bleeding, edema, necrosis, hemorrhage, pain at the bite site and systemic changes. This study was conducted to evaluate the changes induced in hematological and haemostatic parameters in rabbits after 1, 3, 6 and 24 h post-venom of subcutaneously administration of a sublethal dose of Cerastes cerastes and Macrovipera mauritanica venoms. Our results indicated that most hematological and haemostatic parameters showed significant changes 3 and 6 h after envenomation. The hemoglobin, hematocrit, red blood cells, platelets and prothrombin time were reduced significantly 3 h after envenomation. A very significant increase in the levels of white blood cells, lymphocytes, monocytes, activated thromboplastin time and fibrinogen were recorded 6 h following envenomation. However, no significant difference was found for the mean corpuscular volume, corpuscular hemoglobin content and mean corpuscular hemoglobin concentration throughout the whole duration of the experiment. These results suggest that severe hematological and haemostatic changes may be initiated during the early stages of envenomation leading to local and systemic hemorrhages and coagulopathies which are the main cause of death in case of vipers envenomation.
Collapse
|
21
|
Quantitative proteomic analysis of Vietnamese krait venoms: Neurotoxins are the major components in Bungarus multicinctus and phospholipases A2 in Bungarus fasciatus. Toxicon 2015; 107:197-209. [DOI: 10.1016/j.toxicon.2015.08.026] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2015] [Revised: 08/21/2015] [Accepted: 08/27/2015] [Indexed: 11/18/2022]
|
22
|
Tsetse GmmSRPN10 has anti-complement activity and is important for successful establishment of trypanosome infections in the fly midgut. PLoS Negl Trop Dis 2015; 9:e3448. [PMID: 25569180 PMCID: PMC4287558 DOI: 10.1371/journal.pntd.0003448] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2014] [Accepted: 12/01/2014] [Indexed: 11/19/2022] Open
Abstract
The complement cascade in mammalian blood can damage the alimentary tract of haematophagous arthropods. As such, these animals have evolved their own repertoire of complement-inactivating factors, which are inadvertently exploited by blood-borne pathogens to escape complement lysis. Unlike the bloodstream stages, the procyclic (insect) stage of Trypanosoma brucei is highly susceptible to complement killing, which is puzzling considering that a tsetse takes a bloodmeal every 2–4 days. In this study, we identified four tsetse (Glossina morsitans morsitans) serine protease inhibitors (serpins) from a midgut expressed sequence tag (EST) library (GmmSRPN3, GmmSRPN5, GmmSRPN9 and GmmSRPN10) and investigated their role in modulating the establishment of a T. brucei infection in the midgut. Although not having evolved in a common blood-feeding ancestor, all four serpins have an active site sharing remarkable homology with the human complement C1-inhibitor serpin, SerpinG1. RNAi knockdown of individual GmmSRPN9 and GmmSRPN10 genes resulted in a significant decreased rate of infection by procyclic form T. brucei. Furthermore, recombinant GmmSRPN10 was both able to inhibit the activity of human complement-cascade serine proteases, C1s and Factor D, and to protect the in vitro killing of procyclic trypanosomes when incubated with complement-activated human serum. Thus, the secretion of serpins, which may be part of a bloodmeal complement inactivation system in tsetse, is used by procyclic trypanosomes to evade an influx of fresh trypanolytic complement with each bloodmeal. This highlights another facet of the complicated relationship between T. brucei and its tsetse vector, where the parasite takes advantage of tsetse physiology to further its chances of propagation and transmission. Blood feeding arthropods are exploited by blood borne parasites as vectors of transmission. Trypanosoma brucei, a salivarian trypanosome species, must survive, migrate and differentiate in the tsetse until they become mature, mammalian-infective forms within the fly salivary glands. This constitutes a significant challenge to trypanosomes as the major parasite form colonising the tsetse midgut is sensitive to lysis by blood complement, which is introduced into the tsetse gut whenever the fly feeds. In this study, we show that T. brucei may avoid being eliminated by bloodmeal complement by benefitting from a complement-inhibiting enzyme secreted by the fly itself. We showed that this serine protease inhibitor (serpin) enzyme, Serpin10, can inactivate triggers of the complement cascade, protect tsetse-infective trypanosomes from complement lysis, and is important for trypanosome establishment in the tsetse midgut. Taken together, we propose that GmmSRPN10 may be part of a repertoire of complement-inhibiting proteins secreted by tsetse that are utilized by T. brucei to evade complement lysis in the tsetse midgut.
Collapse
|
23
|
Abstract
Complement is traditionally known to be a system of serum proteins that provide protection against pathogens through direct cell lysis and the mobilization of innate and adaptive immunity. However, recent work indicates that the complement system has additional physiological roles beyond those in host defence. In this Opinion article, we describe the new modes and locations of complement activation that enable it to interact with other cell effector systems, such as growth factor receptors, inflammasomes and metabolic pathways. We propose that the location of complement activation dictates its function.
Collapse
|
24
|
Tambourgi DV, van den Berg CW. Animal venoms/toxins and the complement system. Mol Immunol 2014; 61:153-62. [PMID: 24998802 DOI: 10.1016/j.molimm.2014.06.020] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2014] [Revised: 06/10/2014] [Accepted: 06/10/2014] [Indexed: 12/22/2022]
Abstract
Nature is a wealthy source of agents that have been shown to be beneficial to human health, but nature is also a rich source of potential dangerous health damaging compounds. This review will summarise and discuss the agents from the animal kingdom that have been shown to interact with the human complement (C) system. Most of these agents are toxins found in animal venoms and animal secretions. In addition to the mechanism of action of these toxins, their contribution to the field of complement, their role in human pathology and the potential benefit to the venomous animal itself will be discussed. Potential therapeutic applications will also be discussed.
Collapse
Affiliation(s)
| | - Carmen W van den Berg
- Institute of Molecular and Experimental Medicine, School of Medicine, Cardiff University, Cardiff, UK.
| |
Collapse
|