1
|
Kumar D, Downs LP, Embers M, Flynt AS, Karim S. Identification of microRNAs in the Lyme Disease Vector Ixodes scapularis. Int J Mol Sci 2022; 23:5565. [PMID: 35628370 PMCID: PMC9141961 DOI: 10.3390/ijms23105565] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2022] [Revised: 05/05/2022] [Accepted: 05/10/2022] [Indexed: 02/01/2023] Open
Abstract
MicroRNAs (miRNAs) are a class of small non-coding RNAs involved in many biological processes, including the immune pathways that control bacterial, parasitic, and viral infections. Pathogens probably modify host miRNAs to facilitate successful infection, so they might be useful targets for vaccination strategies. There are few data on differentially expressed miRNAs in the black-legged tick Ixodes scapularis after infection with Borrelia burgdorferi, the causative agent of Lyme disease in the United States. Small RNA sequencing and qRT-PCR analysis were used to identify and validate differentially expressed I. scapularis salivary miRNAs. Small RNA-seq yielded 133,465,828 (≥18 nucleotides) and 163,852,135 (≥18 nucleotides) small RNA reads from Borrelia-infected and uninfected salivary glands for downstream analysis using the miRDeep2 algorithm. As such, 254 miRNAs were identified across all datasets, 25 of which were high confidence and 51 low confidence known miRNAs. Further, 23 miRNAs were differentially expressed in uninfected and infected salivary glands: 11 were upregulated and 12 were downregulated upon pathogen infection. Gene ontology and network analysis of target genes of differentially expressed miRNAs predicted roles in metabolic, cellular, development, cellular component biogenesis, and biological regulation processes. Several Kyoto Encyclopedia of Genes and Genomes (KEGG) pathways, including sphingolipid metabolism; valine, leucine and isoleucine degradation; lipid transport and metabolism; exosome biogenesis and secretion; and phosphate-containing compound metabolic processes, were predicted as targets of differentially expressed miRNAs. A qRT-PCR assay was utilized to validate the differential expression of miRNAs. This study provides new insights into the miRNAs expressed in I. scapularis salivary glands and paves the way for their functional manipulation to prevent or treat B. burgdorferi infection.
Collapse
Affiliation(s)
- Deepak Kumar
- Center for Molecular and Cellular Biosciences, University of Southern Mississippi, Hattiesburg, MS 39406, USA; (D.K.); (A.S.F.)
- School of Biological, Environmental, and Earth Sciences, University of Southern Mississippi, Hattiesburg, MS 39406, USA;
| | - Latoyia P. Downs
- School of Biological, Environmental, and Earth Sciences, University of Southern Mississippi, Hattiesburg, MS 39406, USA;
| | - Monica Embers
- Division of Immunology, Tulane National Primate Research Center, Covington, LA 70433, USA;
| | - Alex Sutton Flynt
- Center for Molecular and Cellular Biosciences, University of Southern Mississippi, Hattiesburg, MS 39406, USA; (D.K.); (A.S.F.)
- School of Biological, Environmental, and Earth Sciences, University of Southern Mississippi, Hattiesburg, MS 39406, USA;
| | - Shahid Karim
- Center for Molecular and Cellular Biosciences, University of Southern Mississippi, Hattiesburg, MS 39406, USA; (D.K.); (A.S.F.)
- School of Biological, Environmental, and Earth Sciences, University of Southern Mississippi, Hattiesburg, MS 39406, USA;
| |
Collapse
|
2
|
Pereira MC, Nodari EF, de Abreu MR, Paiatto LN, Simioni PU, Camargo-Mathias MI. Rhipicephalus sanguineus salivary gland extract as a source of immunomodulatory molecules. EXPERIMENTAL & APPLIED ACAROLOGY 2021; 83:387-398. [PMID: 33590358 DOI: 10.1007/s10493-021-00591-w] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/04/2019] [Accepted: 01/22/2021] [Indexed: 06/12/2023]
Abstract
Rhipicephalus sanguineus sensu lato (s.l.), popularly known as 'brown dog tick', is the primary vector of pathogens affecting dogs worldwide. To enter the host's organism, these pathogens utilise the anticoagulant, antiplatelet, anti-inflammatory and immunomodulatory actions of compounds present in the tick's saliva; such compounds are released by the ectoparasite in order to attach and feed on dogs. Nitric oxide (NO) is one of the regulatory factors in inflammation, apoptosis and immunomodulation. Here, we evaluated the in vitro activity of salivary gland extract of female dog ticks on the macrophage-derived J774 cell line, with and without lipopolysaccharide (LPS) stimulation. Cultures were evaluated for possible morphological alterations caused by exposure to the extract. There was no apparent in vitro cytotoxicity of the extract. Also, the NO secretory response in the non-LPS-stimulated cells was not inhibited. On the other hand, the extract presented modulatory action in the cultures of LPS-stimulated cells at a concentration of 0.1 μg/mL, possibly through macrophage activation, and induced a significant decrease in NO secretion. These results confirm the modulatory potential of bioactive molecules in the salivary glands of R. sanguineus ticks.
Collapse
Affiliation(s)
- Melissa Carolina Pereira
- Department of Biology, Institute of Biosciences, São Paulo State University - UNESP, Rio Claro, SP, 13506-900, Brazil
| | - Elen Fernanda Nodari
- Department of Biology, Institute of Biosciences, São Paulo State University - UNESP, Rio Claro, SP, 13506-900, Brazil
| | - Marina Rodrigues de Abreu
- Department of Biology, Institute of Biosciences, São Paulo State University - UNESP, Rio Claro, SP, 13506-900, Brazil
| | - Lisiery Negrini Paiatto
- Department of Genetics, Evolution and Bioagents, Institute of Biology, University of Campinas - UNICAMP, Campinas, SP, 13083-970, Brazil
| | - Patrícia Ucelli Simioni
- Department of Genetics, Evolution and Bioagents, Institute of Biology, University of Campinas - UNICAMP, Campinas, SP, 13083-970, Brazil
| | - Maria Izabel Camargo-Mathias
- Department of Biology, Institute of Biosciences, São Paulo State University - UNESP, Rio Claro, SP, 13506-900, Brazil.
| |
Collapse
|
3
|
Abstract
Borrelia burgdorferi is the causative agent of Lyme disease and is transmitted to vertebrate hosts by Ixodes spp. ticks. The spirochaete relies heavily on its arthropod host for basic metabolic functions and has developed complex interactions with ticks to successfully colonize, persist and, at the optimal time, exit the tick. For example, proteins shield spirochaetes from immune factors in the bloodmeal and facilitate the transition between vertebrate and arthropod environments. On infection, B. burgdorferi induces selected tick proteins that modulate the vector gut microbiota towards an environment that favours colonization by the spirochaete. Additionally, the recent sequencing of the Ixodes scapularis genome and characterization of tick immune defence pathways, such as the JAK–STAT, immune deficiency and cross-species interferon-γ pathways, have advanced our understanding of factors that are important for B. burgdorferi persistence in the tick. In this Review, we summarize interactions between B. burgdorferi and I. scapularis during infection, as well as interactions with tick gut and salivary gland proteins important for establishing infection and transmission to the vertebrate host. Borrelia burgdorferi has a complex life cycle with several different hosts, causing Lyme disease when it infects humans. In this Review, Fikrig and colleagues discuss how B. burgdorferi infects and interacts with its tick vector to ensure onward transmission.
Collapse
|
4
|
Coutinho ML, Bizzarro B, Tirloni L, Berger M, Freire Oliveira CJ, Sá-Nunes A, Silva Vaz I. Rhipicephalus microplus serpins interfere with host immune responses by specifically modulating mast cells and lymphocytes. Ticks Tick Borne Dis 2020; 11:101425. [PMID: 32335011 PMCID: PMC11000276 DOI: 10.1016/j.ttbdis.2020.101425] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2019] [Revised: 03/10/2020] [Accepted: 03/22/2020] [Indexed: 01/09/2023]
Abstract
Rhipicephalus microplus ticks feed on a bovine host for three weeks. At the attachment site, inflammatory and immune responses are triggered resulting in the recruitment of cells and production of a set of immunological mediators. To oppose the host's immune responses, ticks inoculate bioactive salivary molecules capable of interfering with these defense mechanisms. Serpins are among the most frequent molecules present in tick saliva and have been shown to negatively affect the host's anti-tick immunity. R. microplus has at least eighteen full-length serpins (RmS) and eleven are transcribed during blood feeding. Among them, RmS-3, RmS-6, and RmS-17 are present in the saliva of engorged females. Here, the effect of these serpins on the immune responses was evaluated in cells involved in innate/inflammatory (mast cells and macrophages) and adaptive (T cells) immunity. RmS-3 modulated mast cells due to its inhibitory activity on peritoneal rat chymase and on vascular permeability in acute inflammation. In addition, both RmS-6 and RmS-17 inhibited vascular permeability. Of the three serpins studied, neither affected activation nor inflammatory cytokine production by murine macrophages. On the other hand, RmS-3 and RmS-17 presented an inhibitory effect on the metabolic activity of lymphocytes, with the latter being the most potent, while RmS-6 had no effect on it. This activity was associated with a decrease in lymphocyte proliferation, but not with induction of cell death. The present study highlights the powerful modulatory role of tick salivary serpins in the host's immune system and inspire the discovery of targets for the treatment of inflammatory/immune disorders.
Collapse
Affiliation(s)
- Mariana L Coutinho
- Centro de Biotecnologia, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, 91501-970, Brazil.
| | - Bruna Bizzarro
- Departamento de Imunologia, Instituto de Ciências Biomédicas, Universidade de São Paulo, São Paulo, SP, 05508-000, Brazil.
| | - Lucas Tirloni
- Centro de Biotecnologia, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, 91501-970, Brazil.
| | - Markus Berger
- Laboratório de Bioquímica Farmacológica, Centro de Pesquisa Experimental, Hospital de Clínicas de Porto Alegre, Porto Alegre, RS, 90035-007, Brazil.
| | - Carlo Jose Freire Oliveira
- Instituto de Ciências Biológicas e Naturais, Universidade Federal do Triângulo Mineiro, Uberaba, MG, 38025-180, Brazil; Instituto Nacional de Ciência e Tecnologia em Entomologia Molecular (INCT-EM), Rio de Janeiro, RJ, 21941-902, Brazil.
| | - Anderson Sá-Nunes
- Departamento de Imunologia, Instituto de Ciências Biomédicas, Universidade de São Paulo, São Paulo, SP, 05508-000, Brazil; Instituto Nacional de Ciência e Tecnologia em Entomologia Molecular (INCT-EM), Rio de Janeiro, RJ, 21941-902, Brazil.
| | - Itabajara Silva Vaz
- Centro de Biotecnologia, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, 91501-970, Brazil; Instituto Nacional de Ciência e Tecnologia em Entomologia Molecular (INCT-EM), Rio de Janeiro, RJ, 21941-902, Brazil; Faculdade de Veterinária, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, 91501-970, Brazil.
| |
Collapse
|
5
|
Blisnick AA, Šimo L, Grillon C, Fasani F, Brûlé S, Le Bonniec B, Prina E, Marsot M, Relmy A, Blaise-Boisseau S, Richardson J, Bonnet SI. The Immunomodulatory Effect of IrSPI, a Tick Salivary Gland Serine Protease Inhibitor Involved in Ixodes ricinus Tick Feeding. Vaccines (Basel) 2019; 7:vaccines7040148. [PMID: 31614804 PMCID: PMC6963187 DOI: 10.3390/vaccines7040148] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2019] [Revised: 10/02/2019] [Accepted: 10/04/2019] [Indexed: 12/31/2022] Open
Abstract
Ticks are the most important vectors of pathogens affecting both domestic and wild animals worldwide. Hard tick feeding is a slow process—taking up to several days—and necessitates extended control over the host response. The success of the feeding process depends upon injection of tick saliva, which not only controls host hemostasis and wound healing, but also subverts the host immune response to avoid tick rejection that creates a favorable niche for the survival and propagation of diverse tick-borne pathogens. Here, we report on the molecular and biochemical features and functions of an Ixodes ricinus serine protease inhibitor (IrSPI). We characterize IrSPI as a Kunitz elastase inhibitor that is overexpressed in several tick organs—especially salivary glands—during blood-feeding. We also demonstrated that when IrSPI is injected into the host through saliva, it had no impact on tissue factor pathway-induced coagulation, fibrinolysis, endothelial cell angiogenesis or apoptosis, but the protein exhibits immunomodulatory activity. In particular, IrSPI represses proliferation of CD4+ T lymphocytes and proinflammatory cytokine secretion from both splenocytes and macrophages. Our study contributes valuable knowledge to tick-host interactions and provides insights that could be further exploited to design anti-tick vaccines targeting this immunomodulator implicated in I. ricinus feeding.
Collapse
Affiliation(s)
- Adrien A Blisnick
- UMR BIPAR, Animal Health Laboratory, INRA, ANSES, Ecole Nationale Vétérinaire d'Alfort, Université Paris-Est, 94701 Maisons-Alfort CEDEX, France.
| | - Ladislav Šimo
- UMR BIPAR, Animal Health Laboratory, INRA, ANSES, Ecole Nationale Vétérinaire d'Alfort, Université Paris-Est, 94701 Maisons-Alfort CEDEX, France.
| | - Catherine Grillon
- Centre de Biophysique Moléculaire-UPR 4301 CNRS, 45000 Orléans, France.
| | - Fabienne Fasani
- Centre de Biophysique Moléculaire-UPR 4301 CNRS, 45000 Orléans, France.
| | - Sébastien Brûlé
- Plateforme de Biophysique moléculaire, Institut Pasteur, UMR 3528 CNRS, 75015 Paris, France.
| | - Bernard Le Bonniec
- INSERM UMR-S1140, Faculté de Pharmacie Université Paris Descartes, Sorbonne Paris Cité, 75270 Paris CEDEX 06, France.
| | - Eric Prina
- Unité de Parasitologie moléculaire et Signalisation-INSERM U1201, Institut Pasteur, 75015 Paris, France.
| | - Maud Marsot
- Unité EPI, Animal Health Laboratory, INRA, ANSES, Ecole Nationale Vétérinaire d'Alfort, Université Paris-Est, 94701 Maisons-Alfort CEDEX, France.
| | - Anthony Relmy
- UMR Virologie 1161, Animal Health Laboratory, INRA, ANSES, Ecole Nationale Vétérinaire d'Alfort, Université Paris-Est, 94701 Maisons-Alfort CEDEX, France.
| | - Sandra Blaise-Boisseau
- UMR Virologie 1161, Animal Health Laboratory, INRA, ANSES, Ecole Nationale Vétérinaire d'Alfort, Université Paris-Est, 94701 Maisons-Alfort CEDEX, France.
| | - Jennifer Richardson
- UMR Virologie 1161, Animal Health Laboratory, INRA, ANSES, Ecole Nationale Vétérinaire d'Alfort, Université Paris-Est, 94701 Maisons-Alfort CEDEX, France.
| | - Sarah I Bonnet
- UMR BIPAR, Animal Health Laboratory, INRA, ANSES, Ecole Nationale Vétérinaire d'Alfort, Université Paris-Est, 94701 Maisons-Alfort CEDEX, France.
| |
Collapse
|
6
|
Barros MS, Lara PG, Fonseca MT, Moretti EH, Filgueiras LR, Martins JO, Capurro ML, Steiner AA, Sá-Nunes A. Aedes aegypti saliva impairs M1-associated proinflammatory phenotype without promoting or affecting M2 polarization of murine macrophages. Parasit Vectors 2019; 12:239. [PMID: 31097013 PMCID: PMC6524299 DOI: 10.1186/s13071-019-3487-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2018] [Accepted: 05/06/2019] [Indexed: 02/01/2023] Open
Abstract
BACKGROUND During the feeding process, the mouthparts of hematophagous mosquitoes break the skin barrier and probe the host tissue to find the blood. The saliva inoculated in this microenvironment modulates host hemostasis, inflammation and adaptive immune responses. However, the mechanisms involved in these biological activities remain poorly understood and few studies explored the potential roles of mosquito saliva on the individual cellular components of the immune system. Here, we report the immunomodulatory activities of Aedes aegypti salivary cocktail on murine peritoneal macrophages. RESULTS The salivary gland extract (SGE) of Ae. aegypti inhibited the production of nitric oxide and inflammatory cytokines such as interleukin-6 (IL-6) and IL-12, as well as the expression of inducible nitric oxide synthase and NF-κB by murine macrophages stimulated by lipopolysaccharide (LPS) plus interferon-γ (IFN-γ). The spare respiratory capacity, the phagocytic and microbicidal activities of these macrophages were also reduced by Ae. aegypti SGE. These phenotypic changes are consistent with SGE suppressing the proinflammatory program of M1 macrophages. On the other hand, Ae. aegypti SGE did not influence M2-associated markers (urea production, arginase-1 and mannose receptor-1 expression), either in macrophages alternatively activated by IL-4 or in those classically activated by LPS plus IFN-γ. In addition, Ae. aegypti SGE did not display any cytokine-binding activity, nor did it affect macrophage viability, thus excluding supposed experimental artifacts. CONCLUSIONS Given the importance of macrophages in a number of biological processes, our findings help to enlighten how vector saliva modulates vertebrate host immunity.
Collapse
Affiliation(s)
- Michele S Barros
- Laboratory of Experimental Immunology, Department of Immunology, Institute of Biomedical Sciences, University of Sao Paulo, Sao Paulo, SP, 05508-000, Brazil
| | - Priscila G Lara
- Laboratory of Experimental Immunology, Department of Immunology, Institute of Biomedical Sciences, University of Sao Paulo, Sao Paulo, SP, 05508-000, Brazil
| | - Monique T Fonseca
- Laboratory of Sepsis Neurobiology, Department of Immunology, Institute of Biomedical Sciences, University of Sao Paulo, Sao Paulo, SP, 05508-000, Brazil
| | - Eduardo H Moretti
- Laboratory of Sepsis Neurobiology, Department of Immunology, Institute of Biomedical Sciences, University of Sao Paulo, Sao Paulo, SP, 05508-000, Brazil
| | - Luciano R Filgueiras
- Laboratory of Immunopharmacology, Department of Immunology, Institute of Biomedical Sciences, University of Sao Paulo, Sao Paulo, SP, 05508-000, Brazil
| | - Joilson O Martins
- Laboratory of Immunoendocrinology, Department of Clinical and Toxicological Analyses, School of Pharmaceutical Sciences, University of Sao Paulo, Sao Paulo, SP, 05508-000, Brazil
| | - Margareth L Capurro
- Laboratory of Genetically Modified Mosquitoes, Department of Parasitology, Institute of Biomedical Sciences, University of Sao Paulo, Sao Paulo, SP, 05508-000, Brazil
- National Institute of Science and Technology on Molecular Entomology, National Council for Scientific and Technological Development (INCT-EM/CNPq), Rio de Janeiro, RJ, Brazil
| | - Alexandre A Steiner
- Laboratory of Sepsis Neurobiology, Department of Immunology, Institute of Biomedical Sciences, University of Sao Paulo, Sao Paulo, SP, 05508-000, Brazil
| | - Anderson Sá-Nunes
- Laboratory of Experimental Immunology, Department of Immunology, Institute of Biomedical Sciences, University of Sao Paulo, Sao Paulo, SP, 05508-000, Brazil.
- National Institute of Science and Technology on Molecular Entomology, National Council for Scientific and Technological Development (INCT-EM/CNPq), Rio de Janeiro, RJ, Brazil.
| |
Collapse
|
7
|
Breijo M, Esteves E, Bizzarro B, Lara PG, Assis JB, Rocha S, Pastro L, Fernández C, Meikle A, Sá-Nunes A. Hematobin is a novel immunomodulatory protein from the saliva of the horn fly Haematobia irritans that inhibits the inflammatory response in murine macrophages. Parasit Vectors 2018; 11:435. [PMID: 30053916 PMCID: PMC6064106 DOI: 10.1186/s13071-018-3017-z] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2018] [Accepted: 07/16/2018] [Indexed: 12/05/2022] Open
Abstract
Background The horn fly Haematobia irritans is a blood-sucking ectoparasite responsible for substantial economic loss of livestock. Like other hematophagous arthropods species, the successful blood-feeding of H. irritans is highly dependent on the modulation of the host’s hemostasis and immune system. Here, we evaluated the biological activity of hematobin (HTB), a protein recently identified in the H. irritans saliva, on macrophage biology. The goal was to understand the putative interactions between the components of H. irritans saliva and the early host immune responses. Results Thioglycolate-elicited peritoneal macrophages from BALB/c mice were stimulated by lipopolysaccharide (LPS) plus interferon-γ (IFN-γ) in the presence or absence of recombinant HTB. The presence of the salivary protein in the cultures inhibited nitric oxide production and decreased the inducible nitric oxide synthase (iNOS) expression induced by LPS plus IFN-γ. The tumor necrosis factor-α (TNF-α) and interleukin-12p40 (IL-12p40) levels were also reduced in the macrophages pre-incubated with HTB; these findings correlated to the decreased NF-κB expression. The biological activities described here were not associated with changes in annexin V binding to macrophages suggesting that HTB does not induce cell death. In addition, the activity of HTB seems to be specific to macrophages because no changes were observed in lymphocyte proliferation or cytokine production. Conclusions We describe here the first bioactive salivary protein of H. irritans. We characterized its ability to modulate macrophage inflammatory response, and the results can help explain how horn flies modulate the host immune system to feed on blood. Electronic supplementary material The online version of this article (10.1186/s13071-018-3017-z) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Martin Breijo
- Unidad de Reactivos y Biomodelos de Experimentación, Facultad de Medicina, Universidad de la República, Gral. Flores, 2125, Montevideo, Uruguay.
| | - Eliane Esteves
- Department of Immunology, Laboratory of Experimental Immunology, Institute of Biomedical Sciences, University of Sao Paulo, Sao Paulo, SP, 05508-000, Brazil
| | - Bruna Bizzarro
- Department of Immunology, Laboratory of Experimental Immunology, Institute of Biomedical Sciences, University of Sao Paulo, Sao Paulo, SP, 05508-000, Brazil
| | - Priscila G Lara
- Department of Immunology, Laboratory of Experimental Immunology, Institute of Biomedical Sciences, University of Sao Paulo, Sao Paulo, SP, 05508-000, Brazil
| | - Josiane B Assis
- Department of Immunology, Laboratory of Experimental Immunology, Institute of Biomedical Sciences, University of Sao Paulo, Sao Paulo, SP, 05508-000, Brazil
| | - Sergio Rocha
- Unidad de Reactivos y Biomodelos de Experimentación, Facultad de Medicina, Universidad de la República, Gral. Flores, 2125, Montevideo, Uruguay
| | - Lucía Pastro
- Laboratorio de Interacciones Moleculares, Facultad de Ciencias, Universidad de la República, Iguá, 4225, Montevideo, Uruguay
| | - Cecilia Fernández
- Cátedra de Inmunología, Facultad de Química, Universidad de la República, Av. Alfredo Navarro, 3051, Montevideo, Uruguay
| | - Ana Meikle
- Laboratorio de Técnicas Nucleares, Facultad de Veterinaria, Universidad de la República, Lasplaces, 1550, Montevideo, Uruguay
| | - Anderson Sá-Nunes
- Department of Immunology, Laboratory of Experimental Immunology, Institute of Biomedical Sciences, University of Sao Paulo, Sao Paulo, SP, 05508-000, Brazil. .,National Institute of Science and Technology in Molecular Entomology, National Council of Scientific and Technological Development (INCT-EM/CNPq), Rio de Janeiro, RJ, Brazil.
| |
Collapse
|
8
|
Terry AI, Benitez-Kruidenier S, DeKrey GK. Effects of endosulfan isomers on cytokine and nitric oxide production by differentially activated RAW 264.7 cells. Toxicol Rep 2018; 5:396-400. [PMID: 29765866 PMCID: PMC5949890 DOI: 10.1016/j.toxrep.2018.03.006] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2017] [Revised: 02/22/2018] [Accepted: 03/05/2018] [Indexed: 01/08/2023] Open
Abstract
Non-cytotoxic concentrations of endosulfan suppressed NO production. Suppression of NO was a more sensitive endpoint than suppression of TNF. Endosulfan alpha had greater cytotoxic potency than endosulfan beta.
Endosulfan is an organochlorine insecticide comprised of two isomers: endosulfan-α and endosulfan-β. Endosulfan exposure has been shown to elevate some inflammatory factors, such as nitric oxide (NO) and tumor necrosis factor (TNF), in animals or cultures of animal cells. Because the two endosulfan isomers can vary in their biological activities, the goal of this study was to determine if individual endosulfan isomers differentially impact production of NO or TNF by the mouse macrophage cell RAW 264.7 at non-cytotoxic levels. We found elevated TNF with exposure to endosulfan-α (not endosulfan-β), but only at concentrations that were cytotoxic (≥100 μM), whereas neither endosulfan isomer altered baseline levels of NO at any concentration up to 300 μM. In interferon (IFN)-γ-activated cultures, NO levels were significantly suppressed by either endosulfan isomer at 10 μM (the lowest concentration examined), whereas only endosulfan-β significantly lowered TNF levels at non-cytotoxic concentrations. In lipopolysaccharide (LPS)-activated cultures, both endosulfan isomers significantly reduced NO, but not TNF, at non-cytotoxic concentrations. These results suggest that the endosulfan isomers have some capacity to alter inflammatory responses differentially, particularly with IFN-γ stimulation.
Collapse
Key Words
- AP-1, activator protein 1
- DMSO, dimethylsulfoxide
- ER, estrogen receptor
- Endosulfan
- IFN, interferon
- IL, interleukin
- Inflammation
- LPS, lipopolysaccharide
- Macrophage
- NF-κB, nuclear factor kappa-light-chain-enhancer of activated B cells
- NO, nitric oxide
- NOS2, nitric oxide synthase type 2
- Nitric oxide
- PAMP, pathogen-associated molecular pattern
- STAT, signal transducer and activator of transcription
- TNF
- TNF, tumor necrosis factor
Collapse
Affiliation(s)
- Alexander I Terry
- School of Biological Sciences, University of Northern Colorado, 501 20th Street, Greeley, CO 80639, USA
| | - Sandra Benitez-Kruidenier
- School of Biological Sciences, University of Northern Colorado, 501 20th Street, Greeley, CO 80639, USA
| | - Gregory K DeKrey
- School of Biological Sciences, University of Northern Colorado, 501 20th Street, Greeley, CO 80639, USA
| |
Collapse
|
9
|
Interaction between saliva's adenosine and tick parasitism: effects on feeding and reproduction. Parasit Vectors 2017; 10:326. [PMID: 28693553 PMCID: PMC5502490 DOI: 10.1186/s13071-017-2248-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2017] [Accepted: 06/15/2017] [Indexed: 02/02/2023] Open
Abstract
Background It has recently been demonstrated that saliva from Rhipicephalus sanguineus ticks contains adenosine (ADO) and prostaglandin E2 (PGE2), two non-protein molecules that have significant immunomodulatory properties. These molecules can inhibit cytokine production by dendritic cells (DCs), while also reducing the expression of CD40 in these cells. However, more studies are needed for a better understanding of their participation in the feeding of ticks in vivo. This work, therefore, evaluated the importance of ADO during tick infestations. Mice were infested with adult ticks (3 couples/mouse), and their skin was collected at the tick-infested site (3rd and 7th day), and mRNA for receptors of ADO was quantified by real-time PCR. Results Tick infestation increased by four and two times the expression of the A2b and A3v1 receptors on day 3, respectively, while expression of other ADO receptors was unaltered. In addition, we treated mice (n = 10/group) daily with 8-(p-Sulfophenyl)theophylline, 8-pSPT, 20 mg/kg, i.p.), a non-selective antagonist of ADO receptors, and evaluated the performance of ticks during infestations. Female ticks fed on 8-pSPT-treated mice presented a reduction in their engorgement, weight and hatching rates of egg masses, and survival times of larvae compared to the same parameters presented by ticks in the control group. To investigate if these 8-pSPT-treated mice presented altered immune responses, we performed three tick infestations and collected their lymph node cells to determine the percentages and activation state of DCs and cytokine production by lymphocytes by flow cytometry (Cytometric Bead Array technique, CBA). Our data showed that 8-pSPT-treated mice presented an increase in the percentage of DCs as well as of their stimulatory and co-stimulatory molecules (CD40, CD80 and MHCII). Regarding production of T cell cytokines, we observed a significant increase in the levels of IL-2 and a significant decrease in IL-10, IL-17, TNF-α and IFN-γ cytokines. Conclusions These results suggest that ADO produced by ticks helps them feed and reproduce and that this effect may be due to modulation of host DCs and T cells.
Collapse
|
10
|
Schoeler GB, Wikel SK. Modulation of host immunity by haematophagous arthropods. ANNALS OF TROPICAL MEDICINE AND PARASITOLOGY 2016. [DOI: 10.1080/00034983.2001.11813695] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
|
11
|
Wei L, Huang C, Yang H, Li M, Yang J, Qiao X, Mu L, Xiong F, Wu J, Xu W. A potent anti-inflammatory peptide from the salivary glands of horsefly. Parasit Vectors 2015; 8:556. [PMID: 26496724 PMCID: PMC4619319 DOI: 10.1186/s13071-015-1149-y] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2015] [Accepted: 10/07/2015] [Indexed: 01/11/2023] Open
Abstract
Background A diverse group of physiologically active peptides/proteins are present in the salivary glands of horsefly Tabanus yao (Diptera, Tabanidae) that facilitate acquisition of blood meal. However, their roles in the regulation of local inflammation remains poorly understood. Methods Induction expression profiles of immune-related molecules in the salivary glands of T. yao was analyzed by quantitative PCR (qPCR) after bacterial feeding. A significantly up-regulated molecule (cecropin-TY1) was selected for anti-inflammatory assay in lipopolysaccharide (LPS)-stimulated mouse peritoneal macrophages. The transcription levels of inducible NO synthase (iNOS) and pro-inflammatory cytokines were quantified by qPCR. Nitric oxide (NO) production was determined by Griess reagent. Pro-inflammatory cytokine production was determined by an enzyme-linked immunosorbent assay (ELISA). The inflammatory signals were assayed by Western blotting analysis. The secondary structure of cecropin-TY1 was measured by Circular dichroism (CD) spectroscopy. Interaction of cecropin-TY1 with LPS was evaluated by the dissociation of fluorescein isothiocyanate (FITC)-conjugated LPS aggregates and neutralization of LPS determined by a quantitative Chromogenic End-point Tachypleus amebocyte lysate (TAL) assay kit. Homology modeled structure analysis and mutation of key residues/structures were performed to understand its structure-activity relationship. Results Cecropin-TY1 was demonstrated to possess high anti-inflammatory activity and low cytotoxicity toward mouse macrophages. In LPS-stimulated mouse peritoneal macrophage, addition of cecropin-TY1 significantly inhibited the production of nitric oxide (NO) and pro-inflammatory cytokines. Further study revealed that cecropin-TY1 inhibited inflammatory cytokine production by blocking activation of mitogen-activated protein kinases (MAPKs) and transcriptional nuclear factor-κB (NF-κB) signals. Cecropin-TY1 even interacted with LPS and neutralized LPS. The secondary structure analysis revealed that cecropin-TY1 adopted unordered structures in hydrophobic environment but converted to α-helical confirmation in membrane mimetic environments. Homology modeled structure analysis demonstrated that cecropin-TY1 adopted two α-helices (Leu3-Thr24, Ile27-Leu38) linked by a hinge (Leu25-Pro26) and the structure surface was partly positively charged. Structure-activity relationship analysis indicated that several key residues/structures are crucial for its anti-inflammatory activity including α-helices, aromatic residue Trp2, positively charged residues Lys and Arg, hinge residue Pro26 and N-terminal amidation. Conclusions We found a novel anti-inflammatory function of horsefly-derived cecropin-TY1 peptide, laying groundwork for better understanding the ectoparasite-host interaction of horsefly with host and highlighting its potency in anti-inflammatory therapy for sepsis and endotoxin shock caused by Gram-negative bacterial infections. Electronic supplementary material The online version of this article (doi:10.1186/s13071-015-1149-y) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Lin Wei
- Jiangsu Key Laboratory of Infection and Immunity, Institutes of Biology and Medical Sciences, Soochow University, 199 Ren-Ai Road, Suzhou Industrial Park, Suzhou, 215123, Jiangsu Province, China.
| | - Chunjing Huang
- Jiangsu Key Laboratory of Infection and Immunity, Institutes of Biology and Medical Sciences, Soochow University, 199 Ren-Ai Road, Suzhou Industrial Park, Suzhou, 215123, Jiangsu Province, China.
| | - Hailong Yang
- School of Basic Medical Sciences, Kunming Medical University, 1168 West Chunrong Road, Yuhua Avenue, Chenggong District, Kunming, 650500, Yunnan Province, China.
| | - Min Li
- Jiangsu Key Laboratory of Infection and Immunity, Institutes of Biology and Medical Sciences, Soochow University, 199 Ren-Ai Road, Suzhou Industrial Park, Suzhou, 215123, Jiangsu Province, China.
| | - Juanjuan Yang
- College of Biological Science and Technology, Fuzhou University, Fuzhou, 350108, Fujian, China.
| | - Xue Qiao
- Institute of Marine Biological Technology, School of Life Science and Biotechnology, Dalian University of Technology, Dalian, 116024, Liaoning, China.
| | - Lixian Mu
- School of Basic Medical Sciences, Kunming Medical University, 1168 West Chunrong Road, Yuhua Avenue, Chenggong District, Kunming, 650500, Yunnan Province, China.
| | - Fei Xiong
- Jiangsu Key Laboratory of Infection and Immunity, Institutes of Biology and Medical Sciences, Soochow University, 199 Ren-Ai Road, Suzhou Industrial Park, Suzhou, 215123, Jiangsu Province, China.
| | - Jing Wu
- School of Basic Medical Sciences, Kunming Medical University, 1168 West Chunrong Road, Yuhua Avenue, Chenggong District, Kunming, 650500, Yunnan Province, China.
| | - Wei Xu
- Jiangsu Key Laboratory of Infection and Immunity, Institutes of Biology and Medical Sciences, Soochow University, 199 Ren-Ai Road, Suzhou Industrial Park, Suzhou, 215123, Jiangsu Province, China.
| |
Collapse
|
12
|
Kotál J, Langhansová H, Lieskovská J, Andersen JF, Francischetti IMB, Chavakis T, Kopecký J, Pedra JHF, Kotsyfakis M, Chmelař J. Modulation of host immunity by tick saliva. J Proteomics 2015; 128:58-68. [PMID: 26189360 PMCID: PMC4619117 DOI: 10.1016/j.jprot.2015.07.005] [Citation(s) in RCA: 147] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2015] [Revised: 06/30/2015] [Accepted: 07/12/2015] [Indexed: 12/14/2022]
Abstract
UNLABELLED Next generation sequencing and proteomics have helped to comprehensively characterize gene expression in tick salivary glands at both the transcriptome and the proteome level. Functional data are, however, lacking. Given that tick salivary secretions are critical to the success of the tick transmission lifecycle and, as a consequence, for host colonization by the pathogens they spread, we thoroughly review here the literature on the known interactions between tick saliva (or tick salivary gland extracts) and the innate and adaptive vertebrate immune system. The information is intended to serve as a reference for functional characterization of the numerous genes and proteins expressed in tick salivary glands with an ultimate goal to develop novel vector and pathogen control strategies. SIGNIFICANCE We overview all the known interactions of tick saliva with the vertebrate immune system. The provided information is important, given the recent developments in high-throughput transcriptomic and proteomic analysis of gene expression in tick salivary glands, since it may serve as a guideline for the functional characterization of the numerous newly-discovered genes expressed in tick salivary glands.
Collapse
Affiliation(s)
- Jan Kotál
- Faculty of Science, University of South Bohemia in České Budějovice, Budweis, Czech Republic; Laboratory of Genomics and Proteomics of Disease Vectors, Institute of Parasitology, Biology Centre, Czech Academy of Sciences, Budweis, Czech Republic
| | - Helena Langhansová
- Faculty of Science, University of South Bohemia in České Budějovice, Budweis, Czech Republic
| | - Jaroslava Lieskovská
- Faculty of Science, University of South Bohemia in České Budějovice, Budweis, Czech Republic
| | - John F Andersen
- Section of Vector Biology, Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Ivo M B Francischetti
- Section of Vector Biology, Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Triantafyllos Chavakis
- Department of Clinical Pathobiochemistry, Technische Universität Dresden, Dresden, Germany
| | - Jan Kopecký
- Faculty of Science, University of South Bohemia in České Budějovice, Budweis, Czech Republic
| | - Joao H F Pedra
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Michail Kotsyfakis
- Laboratory of Genomics and Proteomics of Disease Vectors, Institute of Parasitology, Biology Centre, Czech Academy of Sciences, Budweis, Czech Republic.
| | - Jindřich Chmelař
- Faculty of Science, University of South Bohemia in České Budějovice, Budweis, Czech Republic; Department of Clinical Pathobiochemistry, Technische Universität Dresden, Dresden, Germany
| |
Collapse
|
13
|
Milhano N, Saito TB, Bechelli J, Fang R, Vilhena M, DE Sousa R, Walker DH. The role of Rhipicephalus sanguineus sensu lato saliva in the dissemination of Rickettsia conorii in C3H/HeJ mice. MEDICAL AND VETERINARY ENTOMOLOGY 2015; 29:225-229. [PMID: 26011701 DOI: 10.1111/mve.12118] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/08/2014] [Revised: 12/24/2014] [Accepted: 01/05/2015] [Indexed: 06/04/2023]
Abstract
Animal models have been developed for the study of rickettsial pathogenesis. However, to understand what occurs during the natural route of rickettsial transmission via the tick bite, the role of tick saliva should be considered in these models. To address this, we analysed the role of tick saliva in the transmission of Rickettsia conorii (Rickettsiales: Rickettsiaceae) in a murine host by intradermally (i.d.) inoculating two groups of susceptible C3H/HeJ mice with this Rickettsia, and infesting one group with nymphal Rhipicephalus sanguineus sensu lato (Ixodida: Ixodidae) ticks. Quantification of bacterial loads and mRNA levels of interleukin-1β (IL-1β), IL-10 and NF-κB was performed in C3H/HeJ lung samples by real-time quantitative polymerase chain reaction (PCR) and real-time reverse transcriptase PCR, respectively. Lung histology was examined to evaluate the pathological manifestations of infection. No statistically significant difference in bacterial load in the lungs of mice was observed between these two groups; however, a statistically significant difference was observed in levels of IL-1β and NF-κB, both of which were higher in the group inoculated with rickettsiae but not infected with ticks. Lung histology in both groups of animals revealed infiltration of inflammatory cells. Overall, this study showed that i.d. inoculation of R. conorii caused infection in the lungs of C3H/HeJ mice and tick saliva inhibited proinflammatory effects.
Collapse
Affiliation(s)
- N Milhano
- Centre for the Study of Vectors and Infectious Diseases Dr Francisco Cambournac, National Institute of Health Dr Ricardo Jorge, Águas de Moura, Portugal
| | - T B Saito
- Department of Pathology, Center for Biodefense and Emerging Infectious Diseases, University of Texas Medical Branch, Galveston, TX, U.S.A
| | - J Bechelli
- Department of Pathology, Center for Biodefense and Emerging Infectious Diseases, University of Texas Medical Branch, Galveston, TX, U.S.A
| | - R Fang
- Department of Pathology, Center for Biodefense and Emerging Infectious Diseases, University of Texas Medical Branch, Galveston, TX, U.S.A
| | - M Vilhena
- Department of Veterinary Medicine, University of Évora, Évora, Portugal
| | - R DE Sousa
- Centre for the Study of Vectors and Infectious Diseases Dr Francisco Cambournac, National Institute of Health Dr Ricardo Jorge, Águas de Moura, Portugal
| | - D H Walker
- Department of Pathology, Center for Biodefense and Emerging Infectious Diseases, University of Texas Medical Branch, Galveston, TX, U.S.A
| |
Collapse
|
14
|
Carregaro V, Ribeiro JM, Valenzuela JG, Souza-Júnior DL, Costa DL, Oliveira CJF, Sacramento LA, Nascimento MSL, Milanezi CM, Cunha FQ, Silva JS. Nucleosides present on phlebotomine saliva induce immunossuppression and promote the infection establishment. PLoS Negl Trop Dis 2015; 9:e0003600. [PMID: 25849562 PMCID: PMC4388340 DOI: 10.1371/journal.pntd.0003600] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2014] [Accepted: 02/05/2015] [Indexed: 01/30/2023] Open
Abstract
Background Sand fly saliva plays a crucial role in establishing Leishmania infection. We identified adenosine (ADO) and adenosine monophosphate (AMP) as active pharmacologic compounds present in Phlebotomus papatasi saliva that inhibit dendritic cell (DC) functions through a PGE2/IL 10-dependent mechanism. Methodology/Principal Findings Herein, we prepared a mixture of ADO and AMP in equimolar amounts similar to those present in the salivary-gland extract (SGE) form one pair of salivary glands of P. papatasi and co-injected it with Leishmania amazonensis or L. major into mouse ears. ADO+AMP mimicked exacerbative effects of P. papatasi saliva in leishmaniasis, increasing parasite burden and cutaneous lesions. Enzymatic catabolism of salivary nucleosides reversed the SGE-induced immunosuppressive effect associated with IL-10 enhancement. Immunosuppressive factors COX2 and IL-10 were upregulated and failed to enhance ear lesion and parasite burden in IL 10-/- infected mice. Furthermore, nucleosides increased regulatory T cell (Treg) marker expression on CD4+CD25- cells, suggesting induction of Tregs on effector T cells (T eff). Treg induction (iTreg) was associated with nucleoside-induced tolerogenic dendritic cells (tDCs) expressing higher levels of COX2 and IL-10. In vitro generation of Tregs was more efficient in DCs treated with nucleosides. Suppressive effects of nucleosides during cutaneous leishmaniasis were mediated through an A2AR-dependent mechanism. Using BALB/c mice deficient in A2A ADO receptor (A2AR–/–), we showed that co-inoculated mice controlled infection, displaying lower parasite numbers at infection sites and reduced iTreg generation. Conclusion/Significance We have demonstrated that ADO and AMP in P. papatasi saliva mediate exacerbative effects of Leishmania infection by acting preferentially on DCs promoting a tolerogenic profile in DCs and by generating iTregs in inflammatory foci through an A2AR mechanism. Leishmania parasites are transmitted to their vertebrate hosts by infected Phlebotomine sand flies during the blood meal of the flies. During the Leishmania transmission, the saliva is inoculated together with parasites and exhibit several pharmacological compounds that facilitate blood feeding, interfering on homeostasis and avoiding inflammation. Thus, these compounds allow the establishment of pathogen infection. We recently identified adenosine (ADO) and adenosine monophosphate (AMP) as major immunomodulatory compounds present within the Old World sand fly species Phlebotomus papatasii, which protected mice from extreme inflammatory insults. ADO limits the magnitude of immune response by displaying a potent anti-inflammatory activity. Here, we demonstrated that ADO and AMP present in Phlebotomus papatasi saliva are involved in the establishment of parasite infection. Such nucleosides act through adenosine A2A receptor (A2AR), inducing a tolerogenic profile on dendritic cells (tDC) that may generate regulatory T cells differentiation, thus leading to suppression of the immune response and parasite survival. The identification of the active salivary constituents could serve as a strategy for the development of new vaccines to control pathogen transmission.
Collapse
Affiliation(s)
- Vanessa Carregaro
- Department of Biochemistry and Immunology, School of Medicine of Ribeirão Preto, University of São Paulo, Ribeirão Preto, Brazil
| | - José M. Ribeiro
- Section of Vector Biology, Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, United States of America
- Vector Molecular Biology Unit, Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Jesus G. Valenzuela
- Section of Vector Biology, Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, United States of America
- Vector Molecular Biology Unit, Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Djalma L. Souza-Júnior
- Department of Biochemistry and Immunology, School of Medicine of Ribeirão Preto, University of São Paulo, Ribeirão Preto, Brazil
| | - Diego L. Costa
- Department of Biochemistry and Immunology, School of Medicine of Ribeirão Preto, University of São Paulo, Ribeirão Preto, Brazil
| | - Carlo J. F. Oliveira
- Institute of Biological and Natural Sciences, Federal University of Triângulo Mineiro, Uberaba, Brazil
| | - Laís A. Sacramento
- Department of Biochemistry and Immunology, School of Medicine of Ribeirão Preto, University of São Paulo, Ribeirão Preto, Brazil
| | - Manuela S. L. Nascimento
- Department of Biochemistry and Immunology, School of Medicine of Ribeirão Preto, University of São Paulo, Ribeirão Preto, Brazil
| | - Cristiane M. Milanezi
- Department of Biochemistry and Immunology, School of Medicine of Ribeirão Preto, University of São Paulo, Ribeirão Preto, Brazil
| | - Fernando Q. Cunha
- Department of Biochemistry and Immunology, School of Medicine of Ribeirão Preto, University of São Paulo, Ribeirão Preto, Brazil
- Department of Pharmacology, School of Medicine of Ribeirão Preto, University of São Paulo, Ribeirão Preto, Brazil
| | - João S. Silva
- Department of Biochemistry and Immunology, School of Medicine of Ribeirão Preto, University of São Paulo, Ribeirão Preto, Brazil
- * E-mail:
| |
Collapse
|
15
|
Poole NM, Mamidanna G, Smith RA, Coons LB, Cole JA. Prostaglandin E(2) in tick saliva regulates macrophage cell migration and cytokine profile. Parasit Vectors 2013; 6:261. [PMID: 24025197 PMCID: PMC3846740 DOI: 10.1186/1756-3305-6-261] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2013] [Accepted: 09/04/2013] [Indexed: 12/29/2022] Open
Abstract
Background Ticks are obligate hematophagous ectoparasites that suppress the host’s immune and inflammatory responses by secreting immuno-modulatory and anti-inflammatory molecules in their saliva. In previous studies we have shown that tick salivary gland extract (SGE) and saliva from Dermacentor variabilis have distinct effects on platelet-derived growth factor (PDGF)-stimulated IC-21 macrophage and NIH3T3-L1 fibroblast migration. Since tick saliva contains a high concentration of prostaglandin E2 (PGE2), a potent modulator of inflammation, we used a PGE2 receptor antagonist to evaluate the role of PGE2 in the different migratory responses induced by saliva and its impact on macrophage cytokine profile. Methods Adult ticks were fed on female New Zealand white rabbits for 5-8 days. Female ticks were stimulated with dopamine/theophylline to induce salivation and saliva was pooled. Competitive enzyme immunoassays (EIA) were used to measure saliva PGE2 content and the changes in macrophage intracellular cyclic adenosine monophosphate (cAMP) levels. The effects of tick saliva on macrophage and fibroblast migration were assessed in the absence and presence of the PGE2 receptor antagonist, AH 6809, using blind well chamber assays. A cytokine antibody array was used to examine the effects of tick saliva on macrophage cytokine secretion. Statistical significance was determined by one-way ANOVA; Student Newman-Kuels post-test was used for multiple comparisons. Results The saliva-induced increase in PDGF-stimulated macrophage migration was reversed by AH 6809. The inhibition of PDGF-stimulated fibroblast migration by saliva was also antagonist-sensitive. Tick saliva induced macrophages to secrete copious amounts of PGE2, and conditioned medium from these cells caused an AH 6809-sensitive inhibition of stimulated fibroblast migration, showing that macrophages can regulate fibroblast activity. We show that tick saliva decreased the secretion of the pro-inflammatory cytokines regulated and normal T cell expressed and secreted (RANTES/CCL5), tumor necrosis factor-alpha (TNF-α), and soluble TNF receptor I (sTNFRI) through a PGE2-dependent mechanism mediated by cAMP. Saliva had similar effects on lipopolysaccharide (LPS) stimulated macrophages. Conclusions Our data show that ticks utilize salivary PGE2 to subvert the ability of macrophages to secrete pro-inflammatory mediators and recruit fibroblasts to the feeding lesion, therefore inhibiting wound healing.
Collapse
Affiliation(s)
- Nina M Poole
- Department of Biological Sciences, The University of Memphis, 239 Ellington Hall, 3700 Walker Avenue, Memphis TN, 38152, USA.
| | | | | | | | | |
Collapse
|
16
|
Rhipicephalus microplus lipocalins (LRMs): Genomic identification and analysis of the bovine immune response using in silico predicted B and T cell epitopes. Int J Parasitol 2013; 43:739-52. [DOI: 10.1016/j.ijpara.2013.04.005] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2013] [Revised: 04/23/2013] [Accepted: 04/25/2013] [Indexed: 11/17/2022]
|
17
|
Furne M, Holen E, Araujo P, Lie KK, Moren M. Cytokine gene expression and prostaglandin production in head kidney leukocytes isolated from Atlantic cod (Gadus morhua) added different levels of arachidonic acid and eicosapentaenoic acid. FISH & SHELLFISH IMMUNOLOGY 2013; 34:770-777. [PMID: 23291252 DOI: 10.1016/j.fsi.2012.11.044] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/06/2012] [Revised: 11/01/2012] [Accepted: 11/30/2012] [Indexed: 05/28/2023]
Abstract
Primary head kidney leukocytes from Atlantic cod were isolated to evaluate the use of arachidonic acid and eicosapentaenoic acid by cyclooxygenases and the production of prostaglandins E₂ and E₃. The expression of cyclooxygenase genes and selected interleukin genes like Interleukin 1β, Interleukin 6, interleukin 8 and interleukin 10 were monitored. Increasing concentrations of eicosapentaenoic acid and arachidonic acid in equal amounts increased cyclooxygenase2 transcription as well as cell secretion of prostaglandin E₂. Even though the ratio of the two fatty acids was 1:1, the ratio between prostaglandin E₂ and E₃ was 50:1. The addition of arachidonic acid alone increased prostaglandin E₂ secretion but did not induce cyclooxygenase2 transcription. However, when the concentration of eicosapentaenoic acid was increased, maintaining arachidonic acid constant, both prostaglandin E₃ and prostaglandin E₂ production was induced and the prostaglandin E₂ production was higher than in cell cultures only added arachidonic acid. An up-regulation of cyclooxygenase2 transcription was also observed. The addition of the two fatty acids also affected the immune response by alteration of leukocytic cytokines gene expression. According to our results the Cyclooxygenase in cod seem to prefer arachidonic acid as substrate. Therefore, we suggest that the shift from marine oils (rich in n-3 fatty acids) to plant oils (higher in n-6 fatty acids) in the diet of commercially reared Atlantic cod could have negative effects on the whole organism through the increase in the production of prostaglandins belonging to those derived from n-6 fatty acids.
Collapse
Affiliation(s)
- Miriam Furne
- National Institute of Nutrition and Seafood Research-NIFES, P.B. 2029 Nordnes, 5817 Bergen, Norway
| | | | | | | | | |
Collapse
|
18
|
Abstract
L-Arginine is converted to the highly reactive and unstable nitric oxide (NO) and L-citrulline by an enzyme named nitric oxide synthase (NOS). NO decomposes into other nitrogen oxides such as nitrite
(NO2-) and nitrate (NO2-), and in the presence of superoxide anion to the potent oxidizing agent peroxynitrite (ONOO−). Activated rodent macrophages are capable of expressing an inducible form of this enzyme (iNOS) in response to appropriate stimuli, i.e., lipopolysaccharide (LPS) and interferon-γ (IFNγ). Other cytokines can modulate the induction of NO biosynthesis in macrophages. NO is a major effector molecule of the anti-microbial and cytotoxic activity of rodent macrophages against certain micro-organisms and tumour cells, respectively. The NO synthesizing pathway has been demonstrated in human monocytes and other cells, but its role in host defence seems to be accessory. A delicate functional balance between microbial stimuli, host-derived cytokines and hormones in the microenvironment regulates iNOS expression. This review will focus mainly on the known and proposed mechanisms of the regulation of iNOS induction, and on agents that can modulate NO release once the active enzyme has been expressed in the macrophage.
Collapse
|
19
|
LIESKOVSKA J, KOPECKY J. Effect of tick saliva on signalling pathways activated by TLR-2 ligand and Borrelia afzelii in dendritic cells. Parasite Immunol 2012; 34:421-9. [DOI: 10.1111/j.1365-3024.2012.01375.x] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
|
20
|
Chmelar J, Calvo E, Pedra JHF, Francischetti IMB, Kotsyfakis M. Tick salivary secretion as a source of antihemostatics. J Proteomics 2012; 75:3842-54. [PMID: 22564820 DOI: 10.1016/j.jprot.2012.04.026] [Citation(s) in RCA: 88] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2012] [Revised: 04/13/2012] [Accepted: 04/18/2012] [Indexed: 01/04/2023]
Abstract
Ticks are mostly obligatory blood feeding ectoparasites that have an impact on human and animal health. In addition to direct damage due to feeding, some tick species serve as the vectors for the causative agents of several diseases, such as the spirochetes of the genus Borrelia causing Lyme disease, the virus of tick-borne encephalitis, various Rickettsial pathogens or even protozoan parasites like Babesia spp. Hard ticks are unique among bloodfeeders because of their prolonged feeding period that may last up to two weeks. During such a long period of blood uptake, the host develops a wide range of mechanisms to prevent blood loss. The arthropod ectoparasite, in turn, secretes saliva in the sites of bite that assists blood feeding. Indeed, tick saliva represents a rich source of proteins with potent pharmacologic action that target different mechanisms of coagulation, platelet aggregation and vasoconstriction. Tick adaptation to their vertebrate hosts led to the inclusion of a powerful protein armamentarium in their salivary secretion that has been investigated by high-throughput methods. The resulting knowledge can be exploited for the isolation of novel antihemostatic agents. Here we review the tick salivary antihemostatics and their characterized functions at the molecular and cellular levels.
Collapse
Affiliation(s)
- Jindrich Chmelar
- Division of Vascular Inflammation, Diabetes and Kidney, Department of Medicine and Institute of Physiology, Technical University Dresden, 01307 Dresden, Germany
| | | | | | | | | |
Collapse
|
21
|
LIESKOVSKÁ J, KOPECKÝ J. Tick saliva suppresses IFN signalling in dendritic cells upon Borrelia afzelii infection. Parasite Immunol 2011; 34:32-9. [DOI: 10.1111/j.1365-3024.2011.01345.x] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|
22
|
Fontaine A, Diouf I, Bakkali N, Missé D, Pagès F, Fusai T, Rogier C, Almeras L. Implication of haematophagous arthropod salivary proteins in host-vector interactions. Parasit Vectors 2011; 4:187. [PMID: 21951834 PMCID: PMC3197560 DOI: 10.1186/1756-3305-4-187] [Citation(s) in RCA: 133] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2011] [Accepted: 09/28/2011] [Indexed: 01/25/2023] Open
Abstract
The saliva of haematophagous arthropods contains an array of anti-haemostatic, anti-inflammatory and immunomodulatory molecules that contribute to the success of the blood meal. The saliva of haematophagous arthropods is also involved in the transmission and the establishment of pathogens in the host and in allergic responses. This survey provides a comprehensive overview of the pharmacological activity and immunogenic properties of the main salivary proteins characterised in various haematophagous arthropod species. The potential biological and epidemiological applications of these immunogenic salivary molecules will be discussed with an emphasis on their use as biomarkers of exposure to haematophagous arthropod bites or vaccine candidates that are liable to improve host protection against vector-borne diseases.
Collapse
Affiliation(s)
- Albin Fontaine
- Unité de Parasitologie - UMR6236 - IFR48, Antenne Marseille de l'Institut de Recherche Biomédicale des Armées (IRBA), Le Pharo, BP 60109, 13 262 Marseille Cedex 07, France
| | - Ibrahima Diouf
- Unité de Parasitologie - UMR6236 - IFR48, Antenne Marseille de l'Institut de Recherche Biomédicale des Armées (IRBA), Le Pharo, BP 60109, 13 262 Marseille Cedex 07, France
| | - Nawal Bakkali
- Unité de Parasitologie - UMR6236 - IFR48, Antenne Marseille de l'Institut de Recherche Biomédicale des Armées (IRBA), Le Pharo, BP 60109, 13 262 Marseille Cedex 07, France
| | - Dorothée Missé
- Laboratoire de Génétique et Evolution des Maladies infectieuses, UMR 2724 CNRS/IRD, Montpellier, France
| | - Frédéric Pagès
- Unité d'Entomologie Médicale, Antenne Marseille de l'Institut de Recherche Biomédicale des Armées (IRBA), Le Pharo, BP 60109, 13 262 Marseille Cedex 07, France
| | - Thierry Fusai
- Unité de Parasitologie - UMR6236 - IFR48, Antenne Marseille de l'Institut de Recherche Biomédicale des Armées (IRBA), Le Pharo, BP 60109, 13 262 Marseille Cedex 07, France
| | - Christophe Rogier
- Unité de Parasitologie - UMR6236 - IFR48, Antenne Marseille de l'Institut de Recherche Biomédicale des Armées (IRBA), Le Pharo, BP 60109, 13 262 Marseille Cedex 07, France
- Institut Pasteur de Madagascar, B.P. 1274, Ambohitrakely, 101 Antananarivo, Madagascar
| | - Lionel Almeras
- Unité de Parasitologie - UMR6236 - IFR48, Antenne Marseille de l'Institut de Recherche Biomédicale des Armées (IRBA), Le Pharo, BP 60109, 13 262 Marseille Cedex 07, France
| |
Collapse
|
23
|
Carregaro V, Sá-Nunes A, Cunha TM, Grespan R, Oliveira CJF, Lima-Junior DS, Costa DL, Verri WA, Milanezi CM, Pham VM, Brand DD, Valenzuela JG, Silva JS, Ribeiro JMC, Cunha FQ. Nucleosides from Phlebotomus papatasi salivary gland ameliorate murine collagen-induced arthritis by impairing dendritic cell functions. THE JOURNAL OF IMMUNOLOGY 2011; 187:4347-59. [PMID: 21930966 DOI: 10.4049/jimmunol.1003404] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Among several pharmacological compounds, Phlebotomine saliva contains substances with anti-inflammatory properties. In this article, we demonstrated the therapeutic activity of salivary gland extract (SGE) of Phlebotomus papatasi in an experimental model of arthritis (collagen-induced arthritis [CIA]) and identified the constituents responsible for such activity. Daily administration of SGE, initiated at disease onset, attenuated the severity of CIA, reducing the joint lesion and proinflammatory cytokine release. In vitro incubation of dendritic cells (DCs) with SGE limited specific CD4(+) Th17 cell response. We identified adenosine (ADO) and 5'AMP as the major salivary molecules responsible for anti-inflammatory activities. Pharmacologic inhibition of ADO A2(A) receptor or enzymatic catabolism of salivary nucleosides reversed the SGE-induced immunosuppressive effect. Importantly, CD73 (ecto-5'-nucleotidase enzyme) is expressed on DC surface during stage of activation, suggesting that ADO is also generated by 5'AMP metabolism. Moreover, both nucleosides mimicked SGE-induced anti-inflammatory activity upon DC function in vitro and attenuated establishment of CIA in vivo. We reveal that ADO and 5'AMP are present in pharmacological amounts in P. papatasi saliva and act preferentially on DC function, consequently reducing Th17 subset activation and suppressing the autoimmune response. Thus, it is plausible that these constituents might be promising therapeutic molecules to target immune inflammatory diseases.
Collapse
Affiliation(s)
- Vanessa Carregaro
- Department of Biochemistry and Immunology, School of Medicine of Ribeirão Preto, University of São Paulo, 14049-900 São Paulo, Brazil
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
24
|
Ma D, Li Y, Dong J, An S, Wang Y, Liu C, Yang X, Yang H, Xu X, Lin D, Lai R. Purification and characterization of two new allergens from the salivary glands of the horsefly, Tabanus yao. Allergy 2011; 66:101-9. [PMID: 20608917 DOI: 10.1111/j.1398-9995.2010.02435.x] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
BACKGROUND Horsefly bite can cause allergic reactions in humans. There is no information about allergenic horsefly proteins. OBJECTIVES The current work aims to purify and characterize IgE-binding proteins from horsefly salivary glands. METHODS Two IgE-binding proteins, Tab a 1 and Tab a 2 with molecular weight of 26 and 35 kd, respectively, were purified and characterized from 60,000 pairs of horsefly salivary glands of Tabanus yao, respectively. Their primary sequences were determined by Edman degradation and cDNA cloning. Their allergenicity was examined using enzyme-linked immunosorbent assay (ELISA), ELISA inhibition tests, and immunoblots. RESULTS Immunoblotting demonstrated IgE binding by 32 and 34 of 37 (86.5% and 91.8%) subjects' sera to Tab a 1 and Tab a 2, respectively. They were identified as an antigen 5-related (Ag 5) protein and hyaluronidase, respectively. ELISA inhibitions of serum IgE reactivity to the horsefly salivary gland extract (SGE) using purified Tab a 1 and Tab a 2 were significant (about 45%). In addition, these proteins showed some IgE-binding capacity to sera of subjects with wasp sting allergy. CONCLUSIONS We have first identified and characterized two IgE-binding proteins, Tab a 1, an Ag 5-like protein and Tab a 2, a hyaluronidase, from the horsefly salivary glands. They appear to be of importance for the allergic reactions induced by horsefly bite. These allergens are thus not only found in stinging but also found in hematophagous insects. These results also provided support for the presence of the so-called wasp-horsefly syndrome (WHS).
Collapse
Affiliation(s)
- D Ma
- Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, China
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Krause PJ, Grant-Kels JM, Tahan SR, Dardick KR, Alarcon-Chaidez F, Bouchard K, Visini C, Deriso C, Foppa IM, Wikel S. Dermatologic changes induced by repeated Ixodes scapularis bites and implications for prevention of tick-borne infection. Vector Borne Zoonotic Dis 2010; 9:603-10. [PMID: 19196014 DOI: 10.1089/vbz.2008.0091] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Previous studies in rodents and people have demonstrated that repeated tick exposure is associated with reduced Borrelia burgdorferi transmission but the mechanism of prevention remains unclear. We examined the acute histopathologic reactions to initial and repeated Ixodes scapularis bites in BALB/c mice and in people. Skin biopsies of BALB/c mice infested for the first time by I. scapularis nymphs revealed vascular dilatation and an accumulation of inflammatory cells adjacent to the bite site but absent at the site of tick attachment. Such changes would enhance tick-borne pathogen transmission. Mice reexposed to I. scapularis nymphs experienced a decrease in vascular dilatation and a marked increase in inflammatory cells at the site of tick attachment. Skin biopsies of people with attached I. scapularis nymphs revealed similar histologic patterns. These results indicate that cellular changes at the tick-dermal interface following I. scapularis attachment are likely to allow for successful transmission of tick-borne pathogens in non-tick-immune hosts and to inhibit tick-borne pathogen transmission in hosts that have developed tick immunity.
Collapse
Affiliation(s)
- Peter J Krause
- University of Connecticut School of Medicine, Farmington, Connecticut, USA.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
26
|
Oliveira CJF, Carvalho WA, Garcia GR, Gutierrez FRS, de Miranda Santos IKF, Silva JS, Ferreira BR. Tick saliva induces regulatory dendritic cells: MAP-kinases and Toll-like receptor-2 expression as potential targets. Vet Parasitol 2009; 167:288-97. [PMID: 19836139 DOI: 10.1016/j.vetpar.2009.09.031] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
Ticks (Acari: Ixodidae) are bloodsucking ectoparasitic arthropods of human and veterinary medical importance. Tick saliva has been shown to contain a wide range of bioactive molecules with vasodilatory, antihemostatic, and immunomodulatory activities. We have previously demonstrated that saliva from Rhipicephalus sanguineus ticks inhibits the maturation of dendritic cells (DCs) stimulated with LPS. Here we examined the mechanism of this immune subversion, evaluating the effect of tick saliva on Toll-like receptor (TLR)-4 signalling pathway in bone marrow-derived DCs. We demonstrated that R. sanguineus tick saliva impairs maturation of DCs stimulated with LPS, a TLR-4 ligand, leading to increased production of interleukin (IL)-10 and reduced synthesis of IL-12p70 and TNF-alpha. The immunomodulatory effect of the tick saliva on the production of pro-inflammatory cytokines by DCs stimulated with LPS was associated with the observation that tick saliva inhibits the activation of the ERK 1/2 and p38 MAP kinases. These effects were independent of the expression of TLR-4 on the surface of DCs. Additionally, saliva-treated DCs also presented a similar pattern of cytokine modulation in response to other TLR ligands. Since the recent literature reports that several parasites evade immune responses through TLR-2-mediated production of IL-10, we evaluated the effect of tick saliva on the percentage of TLR-2(+) DCs stimulated with the TLR-2 ligand lipoteicoic acid (LTA). The data showed that the population of DCs expressing TLR-2 was significantly increased in DCs treated with LTA plus saliva. In addition, tick saliva alone increased the expression of TLR-2 in a dose- and time-dependent manner. Our data suggest that tick saliva induces regulatory DCs, which secrete IL-10 and low levels of IL-12 and TNF-alpha when stimulated by TLR ligands. Such regulatory DCs are associated with expression of TLR-2 and inhibition of ERK and p38, which promotes the production of IL-10 and thus down-modulates the host's immune response, possibly favouring susceptibility to tick infestations.
Collapse
Affiliation(s)
- Carlo José F Oliveira
- Department of Biochemistry and Immunology, School of Medicine of Ribeirão Preto, University of São Paulo, SP, Brazil
| | | | | | | | | | | | | |
Collapse
|
27
|
Zhao R, Yu X, Yu H, Han W, Zhai L, Han J, Liu J. Immunoregulatory peptides from salivary glands of the horsefly, Tabanus pleskei. Comp Biochem Physiol B Biochem Mol Biol 2009; 154:1-5. [PMID: 19332144 DOI: 10.1016/j.cbpb.2009.03.009] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2009] [Revised: 03/11/2009] [Accepted: 03/12/2009] [Indexed: 10/21/2022]
|
28
|
Ma D, Wang Y, Yang H, Wu J, An S, Gao L, Xu X, Lai R. Anti-thrombosis repertoire of blood-feeding horsefly salivary glands. Mol Cell Proteomics 2009; 8:2071-9. [PMID: 19531497 PMCID: PMC2742439 DOI: 10.1074/mcp.m900186-mcp200] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2009] [Revised: 06/01/2009] [Indexed: 11/06/2022] Open
Abstract
Blood-feeding arthropods rely heavily on the pharmacological properties of their saliva to get a blood meal and suppress immune reactions of hosts. Little information is available on antihemostatic substances in horsefly salivary glands although their saliva has been thought to contain wide range of physiologically active molecules. In traditional Eastern medicine, horseflies are used as anti-thrombosis material for hundreds of years. By proteomics coupling transcriptome analysis with pharmacological testing, several families of proteins or peptides, which exert mainly on anti-thrombosis functions, were identified and characterized from 60,000 pairs of salivary glands of the horsefly Tabanus yao Macquart (Diptera, Tabanidae). They are: (I) ten fibrin(ogen)olytic enzymes, which hydrolyze specially alpha chain of fibrin(ogen) and are the first family of fibrin(ogen)olytic enzymes purified and characterized from arthropods; (II) another fibrin(ogen)olytic enzyme, which hydrolyzes both alpha and beta chain of fibrin(ogen); (III) ten Arg-Gly-Asp-motif containing proteins acting as platelet aggregation inhibitors; (IV) five thrombin inhibitor peptides; (V) three vasodilator peptides; (VI) one apyrase acting as platelet aggregation inhibitor; (VII) one peroxidase with both platelet aggregation inhibitory and vasodilator activities. The first three families are belonging to antigen five proteins, which show obvious similarity with insect allergens. They are the first members of the antigen 5 family found in salivary glands of blood sucking arthropods to have anti-thromobosis function. The current results imply a possible evolution from allergens of blood-sucking insects to anti-thrombosis agents. The extreme diversity of horsefly anti-thrombosis components also reveals the anti-thrombosis molecular mechanisms of the traditional Eastern medicine insect material.
Collapse
Affiliation(s)
- Dongying Ma
- From the ‡Biotoxin Units of Key Laboratory of Animal Models and Human Disease Mechanisms, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming 650223, Yunnan, China
- §Graduate School of the Chinese Academy of Sciences, Beijing 100009, China
| | - Yipeng Wang
- From the ‡Biotoxin Units of Key Laboratory of Animal Models and Human Disease Mechanisms, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming 650223, Yunnan, China
- §Graduate School of the Chinese Academy of Sciences, Beijing 100009, China
| | - Hailong Yang
- From the ‡Biotoxin Units of Key Laboratory of Animal Models and Human Disease Mechanisms, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming 650223, Yunnan, China
- §Graduate School of the Chinese Academy of Sciences, Beijing 100009, China
| | - Jing Wu
- From the ‡Biotoxin Units of Key Laboratory of Animal Models and Human Disease Mechanisms, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming 650223, Yunnan, China
- §Graduate School of the Chinese Academy of Sciences, Beijing 100009, China
| | - Shu An
- ‖School of Life Sciences, University of Science and Technology of China, Hefei, Anhui, 230026, China, and
- §Graduate School of the Chinese Academy of Sciences, Beijing 100009, China
| | - Li Gao
- From the ‡Biotoxin Units of Key Laboratory of Animal Models and Human Disease Mechanisms, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming 650223, Yunnan, China
- §Graduate School of the Chinese Academy of Sciences, Beijing 100009, China
| | - Xueqing Xu
- From the ‡Biotoxin Units of Key Laboratory of Animal Models and Human Disease Mechanisms, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming 650223, Yunnan, China
- §Graduate School of the Chinese Academy of Sciences, Beijing 100009, China
| | - Ren Lai
- From the ‡Biotoxin Units of Key Laboratory of Animal Models and Human Disease Mechanisms, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming 650223, Yunnan, China
- **Life Sciences College of Nanjing Agricultural University, Nanjing 210095, Jiangsu, China
| |
Collapse
|
29
|
Ma D, Gao L, An S, Song Y, Wu J, Xu X, Lai R. A horsefly saliva antigen 5-like protein containing RTS motif is an angiogenesis inhibitor. Toxicon 2009; 55:45-51. [PMID: 19635491 DOI: 10.1016/j.toxicon.2009.06.038] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2009] [Revised: 06/25/2009] [Accepted: 06/26/2009] [Indexed: 11/28/2022]
Abstract
The Ag5 proteins are the most abundant and immunogenic proteins in the venom secretory ducts of stinging insects. An antigen 5-like protein (named tabRTS) composed of 221 amino acid residues was purified and characterized from the salivary glands of the horsefly, Tabanus yao (Diptera, Tabanidae). Its cDNA was cloned from the cDNA library of the horsefly's salivary gland. TabRTS containing the SCP domain (Sc7 family of extracellular protein domain) was found in insect antigen 5 proteins. More interestingly, there is an Arg-Thr-Ser (RTS) disintegrin motif at the C-terminus of tabRTS. The RTS motif is positioned in a loop bracketed by cysteine residues as those found in RTS-disintegrins of Crotalidae and Viperidae snake venoms, which act as angiogenesis inhibitors. Endothelial Cell Tube formation assay in vitro and chicken chorioallantoic membrane (CAM) angiogenesis assay in vivo were performed as to investigate the effect of tabRTS on angiogenesis. It was found that tabRTS could significantly inhibit angiogenesis in vitro and in vivo. Anti-alpha(1)beta(1) monoclonal antibody could dose-dependently inhibit the anti-angiogenic activity of tabRTS. This result indicated that tabRTS possibly targets the alpha(1)beta(1) integrin to exert the anti-angiogenic activity as snake venom RTS-/KTS-disintegrins do. The current work revealed the first angiogenesis inhibitor protein containing RTS motif from invertebrates, a possible novel type of RTS-disintegrin.
Collapse
Affiliation(s)
- Dongying Ma
- Biotoxin Units of Key Laboratory of Animal Models and Human Disease Mechanisms, Kunming Institute of Zoology, Chinese Academy of Sciences, 32# Jiaochangdonglu, Kunming 650223, Yunnan, China
| | | | | | | | | | | | | |
Collapse
|
30
|
Sá-Nunes A, Bafica A, Antonelli LR, Choi EY, Francischetti IMB, Andersen JF, Shi GP, Chavakis T, Ribeiro JM, Kotsyfakis M. The immunomodulatory action of sialostatin L on dendritic cells reveals its potential to interfere with autoimmunity. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2009; 182:7422-9. [PMID: 19494265 PMCID: PMC2694955 DOI: 10.4049/jimmunol.0900075] [Citation(s) in RCA: 74] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
Sialostatin L (SialoL) is a secreted cysteine protease inhibitor identified in the salivary glands of the Lyme disease vector Ixodes scapularis. In this study, we reveal the mechanisms of SialoL immunomodulatory actions on the vertebrate host. LPS-induced maturation of dendritic cells from C57BL/6 mice was significantly reduced in the presence of SialoL. Although OVA degradation was not affected by the presence of SialoL in dendritic cell cultures, cathepsin S activity was partially inhibited, leading to an accumulation of a 10-kDa invariant chain intermediate in these cells. As a consequence, in vitro Ag-specific CD4(+) T cell proliferation was inhibited in a time-dependent manner by SialoL, and further studies engaging cathepsin S(-/-) or cathepsin L(-/-) dendritic cells confirmed that the immunomodulatory actions of SialoL are mediated by inhibition of cathepsin S. Moreover, mice treated with SialoL displayed decreased early T cell expansion and recall response upon antigenic stimulation. Finally, SialoL administration during the immunization phase of experimental autoimmune encephalomyelitis in mice significantly prevented disease symptoms, which was associated with impaired IFN-gamma and IL-17 production and specific T cell proliferation. These results illuminate the dual mechanism by which a human disease vector protein modulates vertebrate host immunity and reveals its potential in prevention of an autoimmune disease.
Collapse
MESH Headings
- Animals
- Antigens/immunology
- Autoimmunity/drug effects
- Autoimmunity/immunology
- Cathepsins/antagonists & inhibitors
- Cathepsins/metabolism
- Cell Differentiation/drug effects
- Cell Differentiation/immunology
- Cells, Cultured
- Cystatins/pharmacology
- Cytokines/biosynthesis
- Cytokines/immunology
- Dendritic Cells/cytology
- Dendritic Cells/drug effects
- Dendritic Cells/immunology
- Dendritic Cells/metabolism
- Encephalomyelitis, Autoimmune, Experimental/genetics
- Encephalomyelitis, Autoimmune, Experimental/immunology
- Encephalomyelitis, Autoimmune, Experimental/metabolism
- Encephalomyelitis, Autoimmune, Experimental/prevention & control
- Enzyme Inhibitors/pharmacology
- Female
- Ixodes/chemistry
- Lipopolysaccharides/pharmacology
- Lymphocyte Culture Test, Mixed
- Mice
- Mice, Knockout
- Protein Binding
- T-Lymphocytes/cytology
- T-Lymphocytes/drug effects
- T-Lymphocytes/immunology
- T-Lymphocytes/metabolism
Collapse
Affiliation(s)
- Anderson Sá-Nunes
- Laboratory of Experimental Immunology, Departament of Immunology, Institute of Biomedical Sciences, University of São Paulo, SP 055908, Brazil
- Section of Vector Biology, Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD 20852
| | - André Bafica
- Division of Immunology, Microbiology and Parasitology Department, Federal University of Santa Catarina, Florianópolis, SC 88040, Brazil
| | - Lis R. Antonelli
- Immunobiology Section, Laboratory of Parasitic Diseases, NIAID, NIH, Bethesda, MD 20892
| | - Eun Young Choi
- Experimental Immunology Branch, National Cancer Institute, NIH, Bethesda, MD 20892
| | - Ivo M. B. Francischetti
- Section of Vector Biology, Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD 20852
| | - John F. Andersen
- Section of Vector Biology, Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD 20852
| | - Guo-Ping Shi
- Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115
| | | | - José M. Ribeiro
- Section of Vector Biology, Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD 20852
| | - Michalis Kotsyfakis
- Section of Vector Biology, Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD 20852
| |
Collapse
|
31
|
Grigorieva LA, Amosova LI. Morphofunctional changes of salivary glands of female ixodid ticks of subfamilies Ixodinae and Amblyomminae (Acari: Ixodidae) during feeding and their significance. J EVOL BIOCHEM PHYS+ 2009. [DOI: 10.1134/s0022093008060100] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
32
|
Tsao JI. Reviewing molecular adaptations of Lyme borreliosis spirochetes in the context of reproductive fitness in natural transmission cycles. Vet Res 2009; 40:36. [PMID: 19368764 PMCID: PMC2701186 DOI: 10.1051/vetres/2009019] [Citation(s) in RCA: 73] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2008] [Accepted: 04/15/2009] [Indexed: 02/04/2023] Open
Abstract
Lyme borreliosis (LB) is caused by a group of pathogenic spirochetes – most often Borrelia burgdorferi, B. afzelii, and B. garinii – that are vectored by hard ticks in the Ixodes ricinus-persulcatus complex, which feed on a variety of mammals, birds, and lizards. Although LB is one of the best-studied vector-borne zoonoses, the annual incidence in North America and Europe leads other vector-borne diseases and continues to increase. What factors make the LB system so successful, and how can researchers hope to reduce disease risk – either through vaccinating humans or reducing the risk of contacting infected ticks in nature? Discoveries of molecular interactions involved in the transmission of LB spirochetes have accelerated recently, revealing complex interactions among the spirochete-tick-vertebrate triad. These interactions involve multiple, and often redundant, pathways that reflect the evolution of general and specific mechanisms by which the spirochetes survive and reproduce. Previous reviews have focused on the molecular interactions or population biology of the system. Here molecular interactions among the LB spirochete, its vector, and vertebrate hosts are reviewed in the context of natural maintenance cycles, which represent the ecological and evolutionary contexts that shape these interactions. This holistic system approach may help researchers develop additional testable hypotheses about transmission processes, interpret laboratory results, and guide development of future LB control measures and management.
Collapse
Affiliation(s)
- Jean I Tsao
- Department of Fisheries and Wildlife, Michigan State University, East Lansing, MI 48864, USA.
| |
Collapse
|
33
|
Alarcon-Chaidez FJ, Boppana VD, Hagymasi AT, Adler AJ, Wikel SK. A novel sphingomyelinase-like enzyme in Ixodes scapularis tick saliva drives host CD4 T cells to express IL-4. Parasite Immunol 2009; 31:210-9. [PMID: 19292772 PMCID: PMC2748904 DOI: 10.1111/j.1365-3024.2009.01095.x] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Tick feeding modulates host immune responses. Tick-induced skewing of host CD4(+) T cells towards a Th2 cytokine profile facilitates transmission of tick-borne pathogens that would otherwise be neutralized by Th1 cytokines. Tick-derived factors that drive this Th2 response have not previously been characterized. In the current study, we examined an I. scapularis cDNA library prepared at 18-24 h of feeding and identified and expressed a tick gene with homology to Loxosceles spider venom proteins with sphingomyelinase activity. This I. scapularis sphingomyelinase-like (IsSMase) protein is a Mg(2+)-dependent, neutral (pH 7.4) form of sphingomyelinase. Significantly, in an in vivo TCR transgenic adoptive transfer assay IsSMase programmed host CD4(+) T cells to express the hallmark Th2 effector cytokine IL-4. IsSMase appears to directly programme host CD4 T cell IL-4 expression (as opposed to its metabolic by-products) because induced IL-4 expression was not altered when enzymatic activity was neutralized. TCR transgenic CD4 T cell proliferation (CFSE-dilution) was also significantly increased by IsSMase. Furthermore, a Th2 response is superimposed onto a virally primed Th1 response by IsSMase. Thus, IsSMase is the first identified tick molecule capable of programming host CD4(+) T cells to express IL-4.
Collapse
Affiliation(s)
- F J Alarcon-Chaidez
- Department of Immunology, University of Connecticut Health Center, Farmington, 06030 USA
| | | | | | | | | |
Collapse
|
34
|
Juncadella IJ, Anguita J. The immunosuppresive tick salivary protein, Salp15. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2009; 666:121-31. [PMID: 20054980 DOI: 10.1007/978-1-4419-1601-3_10] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
The interaction between Ixodid ticks and their mammalian hosts is a complex relationship. While the mammalian host tries to avoid the completion of the feeding process, the tick has devised strategies to counteract these attempts. Tick saliva contains a vast array of pharmacological activities that presumably aid the tick to evade host responses, including anticomplement, oxidative and innate and adaptive immune responses. The characterization of these activities has gained momentum in the last several years. One of the best studied activities present in tick saliva corresponds to the antigen known as Salp15, which binds specifically to the T-cell coreceptor CD4 resulting in the specific repression of CD4+ T-cell activation. We discuss here the current state of our knowledge of the mode of action of this salivary protein.
Collapse
Affiliation(s)
- Ignacio J Juncadella
- Department of Veterinary and Animal Sciences, University of Massachusetts Amherst, Amherst, MA 01003, USA
| | | |
Collapse
|
35
|
Skallová A, Iezzi G, Ampenberger F, Kopf M, Kopecky J. Tick saliva inhibits dendritic cell migration, maturation, and function while promoting development of Th2 responses. THE JOURNAL OF IMMUNOLOGY 2008; 180:6186-92. [PMID: 18424740 DOI: 10.4049/jimmunol.180.9.6186] [Citation(s) in RCA: 65] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Similarly to other blood-feeding arthropods, ticks have evolved immunosuppressive mechanisms enabling them to overcome the host immune system. Although the immunomodulatory effect of tick saliva on several cell populations of the immune system has been extensively studied, little is known about its impact on dendritic cells (DCs). We have examined the effect of Ixodes ricinus tick saliva on DC function in vitro and in vivo. Exposure of DCs to tick saliva in vitro resulted in impaired maturation, upon CD40 or TLR9, TLR3 and TLR7 ligation, as well as reduced Ag presentation capacity. Administration of tick saliva in vivo significantly inhibited maturation and early migration of DCs from inflamed skin to draining lymph nodes, and decreased the capacity of lymph node DCs to present soluble Ag to specific T cells. Moreover, saliva-exposed DCs failed to induce efficient Th1 and Th17 polarization and promoted development of Th2 responses. Our data reveal a complex inhibitory effect exerted by tick saliva on DC function. Given the role of DCs as the key instigators of adaptive immune responses, alteration of their function might represent a major mechanism of tick-mediated immune evasion.
Collapse
Affiliation(s)
- Anna Skallová
- Institute of Parasitology, Biology Centre AS CR, Branisovska 31, Ceské Budejovice, Czech Republic
| | | | | | | | | |
Collapse
|
36
|
Yan X, Feng H, Yu H, Yang X, Liu J, Lai R. An immunoregulatory peptide from salivary glands of the horsefly, Hybomitra atriperoides. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2008; 32:1242-1247. [PMID: 18502504 DOI: 10.1016/j.dci.2008.04.003] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/25/2008] [Revised: 04/15/2008] [Accepted: 04/15/2008] [Indexed: 05/26/2023]
Abstract
Horseflies are economically important blood-feeding arthropods and also a nuisance for humans, and vectors for filariasis. They rely heavily on the pharmacological propriety of their saliva to get blood meal and suppress immune reactions of hosts. Little information is available on horsefly immune suppressants. By high-performance liquid chromatography (HPLC) purification coupling with pharmacological testing, an immunoregulatory peptide named immunoregulin HA has been identified and characterized from salivary glands of the horsefly of Hybomitra atriperoides (Diptera, Tabanidae). Immunoregulin HA could inhibit the secretion of interferon-gamma (IFN-gamma) and monocyte chemoattractant protein (MCP-1) and increase the secretion of interleukin-10 (IL-10) induced by lipopolysaccharide (LPS) in rat splenocytes. IL-10 is a suppressor cytokine of T-cell proliferative and cytokine responses. IL-10 can inhibit the elaboration of pro-inflammatory cytokines. Immunoregulin HA possibly unregulated the IL-10 production to inhibit IFN-gamma and MCP-1 secretion in the current experiments. This immunosuppression may facilitate the blood feeding of this horsefly. The current works will facilitate to understand the molecular mechanisms of the ectoparasite-host relationship.
Collapse
Affiliation(s)
- Xiuwen Yan
- College of Life Sciences School of Heibei Normal University, Shijiazhuang, Hebei 050016, China
| | | | | | | | | | | |
Collapse
|
37
|
Carregaro V, Valenzuela JG, Cunha TM, Verri WA, Grespan R, Matsumura G, Ribeiro JMC, Elnaiem DE, Silva JS, Cunha FQ. Phlebotomine salivas inhibit immune inflammation-induced neutrophil migration via an autocrine DC-derived PGE2/IL-10 sequential pathway. J Leukoc Biol 2008; 84:104-14. [PMID: 18390928 DOI: 10.1189/jlb.1107797] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
In the present study, we investigated whether saliva from Phlebotomus papatasi and Phlebotomus duboscqi inhibited antigen-induced neutrophil migration and the mechanisms involved in these effects. The pretreatment of immunized mice with salivary gland extracts (SGE) of both phlebotomines inhibited OVA challenge-induced neutrophil migration and release of the neutrophil chemotactic mediators, MIP-1alpha, TNF-alpha, and leukotriene B4 (LTB4). Furthermore, SGE treatment enhanced the production of anti-inflammatory mediators, IL-10 and PGE2. SGE treatments failed to inhibit neutrophil migration and MIP-1alpha and LTB4 production in IL-10-/- mice, also failing in mice treated with nonselective (indomethacin) or selective (rofecoxibe) cyclooxygenase (COX) inhibitors. COX inhibition resulted in diminished SGE-induced IL-10 production, and PGE2 release triggered by SGE remained increased in IL-10-/- mice, suggesting that prostanoids are acting through an IL-10-dependent mechanism. SGE treatments in vivo reduced the OVA-induced lymphoproliferation of spleen-derived cells. Further, the in vitro incubation of bone marrow-derived dendritic cells (DC) with SGE inhibited the proliferation of CD4+T cells from OVA-immunized mice, which was reversed by indomethacin and anti-IL-10 antibody treatments. Supporting these results, SGE induced the production of PGE2 and IL-10 by DC, which were blocked by COX inhibition. These effects were associated with the reduction of DC-membrane expression of MHC-II and CD86 by SGE treatment. Altogether, the results showed that Phlebotomine saliva inhibits immune inflammation-induced neutrophil migration by an autocrine DC sequential production of PGE2/IL-10, suggesting that the saliva constituents might be promising therapeutic molecules to target immune inflammatory diseases.
Collapse
Affiliation(s)
- Vanessa Carregaro
- Department of Biochemistry and Immunology, School of Medicine of Ribeirão Preto, University of Sao Paulo, Av Bandeirantes, 3900, Ribeirão Preto, SP, Brazil, 14049-900
| | | | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Xu X, Yang H, Ma D, Wu J, Wang Y, Song Y, Wang X, Lu Y, Yang J, Lai R. Toward an understanding of the molecular mechanism for successful blood feeding by coupling proteomics analysis with pharmacological testing of horsefly salivary glands. Mol Cell Proteomics 2007; 7:582-90. [PMID: 18087067 DOI: 10.1074/mcp.m700497-mcp200] [Citation(s) in RCA: 72] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Horseflies are economically important blood-feeding arthropods and also a nuisance for humans and vectors for filariasis. They rely heavily on the pharmacological properties of their saliva to get a blood meal and suppress immune reactions of hosts. Little information is available on antihemostatic substances in horsefly salivary glands; especially no horsefly immune suppressants have been reported. By proteomics or peptidomics and coupling transcriptome analysis with pharmacological testing, several families of proteins or peptides, which act mainly on the hemostatic system or immune system of the host, were identified and characterized from 30,000 pairs salivary glands of the horsefly Tabanus yao (Diptera, Tabanidae). They are: (i) a novel family of inhibitors of platelet aggregation including two members, which possibly inhibit platelet aggregation by a novel mechanism and act on platelet membrane, (ii) a novel family of immunosuppressant peptides including 12 members, which can inhibit interferon-gamma production and increase interleukin-10 secretion, (iii) a serine protease inhibitor with 56 amino acid residues containing anticoagulant activity, (iv) a serine protease with anticoagulant activity, (v) a protease with fibrinogenolytic activity, (vi) three families of antimicrobial peptides including six members, (vii) a hyaluronidase, (viii) a vasodilator peptide, which is an isoform of vasotab identified from Hybomitra bimaculata, and interestingly (ix) two metallothioneins, which are the first metallothioneins reported from invertebrate salivary glands. The current work will facilitate the understanding of the molecular mechanisms of the ectoparasite-host relationship and help in identifying novel vaccine targets and novel leading pharmacological compounds.
Collapse
Affiliation(s)
- Xueqing Xu
- Biotoxin Units of Key Laboratory of Animal Models and Human Disease Mechanisms, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming 650223, Yunnan, China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Müller-Doblies UU, Maxwell SS, Boppana VD, Mihalyo MA, McSorley SJ, Vella AT, Adler AJ, Wikel SK. Feeding by the tick, Ixodes scapularis, causes CD4+T cells responding to cognate antigen to develop the capacity to express IL-4. Parasite Immunol 2007; 29:485-99. [PMID: 17883452 DOI: 10.1111/j.1365-3024.2007.00966.x] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Effects of tick feeding on an early antigen-specific T cell response were studied by monitoring a clonotypic population of adoptively transferred T cell receptor (TCR) transgenic CD4 cells responding to a tick-associated antigen. When recipient mice were infested with pathogen-free Ixodes scapularis nymphs several days prior to T cell transfer and intradermal injection of soluble cognate antigen at the feeding site, the clonotypic CD4 cells gained the ability to express the Th2 effector cytokine IL-4. Notably, this effect was not only observed in BALB/c mice predisposed towards developing Th2 responses but also in B10.D2 mice predisposed towards Th1 responsiveness. Furthermore, tick feeding was able to superimpose IL-4 expression potential onto a strong Th1 response (indicated by robust IFN-gamma expression potential) elicited by immunization with a vaccinia virus expressing the cognate antigen. The magnitude to which tick feeding was able to programme IL-4 expression potential in CD4 cells was partially reduced in mice that had been previously exposed to pathogen-free tick nymphs 6 weeks earlier, as well as when the nymphs were infected with Borrelia burgdorferi. Intradermal injection of salivary gland extract programmed IL-4 expression potential similar to that of tick infestation, suggesting that IL-4 programming activity is contained within tick saliva.
Collapse
|
40
|
Sá-Nunes A, Bafica A, Lucas DA, Conrads TP, Veenstra TD, Andersen JF, Mather TN, Ribeiro JMC, Francischetti IMB. Prostaglandin E2 is a major inhibitor of dendritic cell maturation and function in Ixodes scapularis saliva. THE JOURNAL OF IMMUNOLOGY 2007; 179:1497-505. [PMID: 17641015 DOI: 10.4049/jimmunol.179.3.1497] [Citation(s) in RCA: 100] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Tick saliva is thought to contain a number of molecules that prevent host immune and inflammatory responses. In this study, the effects of Ixodes scapularis saliva on cytokine production by bone marrow-derived dendritic cells (DCs) from C57BL/6 mice stimulated by TLR-2, TLR-4, and TLR-9 ligands were studied. Saliva at remarkably diluted concentrations (<1/2000) promotes a dose-dependent inhibition of IL-12 and TNF-alpha production induced by all TLR ligands used. Using a combination of fractionation techniques (microcon filtration, molecular sieving, and reversed-phase chromatography), we unambiguously identified PGE(2) as the salivary inhibitor of IL-12 and TNF-alpha production by DCs. Moreover, we have found that I. scapularis saliva (dilution 1/200; approximately 10 nM PGE(2)) marginally inhibited LPS-induced CD40, but not CD80, CD86, or MHC class II expression. In addition, saliva significantly suppressed the ability of DCs to stimulate Ag-specific CD4(+) T cell proliferation and IL-2 production. Notably, the effect of saliva on DC maturation and function was reproduced by comparable concentrations of standard PGE(2). These findings indicate that PGE(2) accounts for most inhibition of DC function observed with saliva in vitro. The role of salivary PGE(2) in vector-host interaction and host immune modulation and inflammation in vivo is also discussed. This study is the first to identify molecularly a DC inhibitor from blood-sucking arthropods.
Collapse
Affiliation(s)
- Anderson Sá-Nunes
- Section of Vector Biology, Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases/National Institutes of Health, Rockville, MD 20852, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Fonseca L, Seixas E, Butcher G, Langhorne J. Cytokine responses of CD4+ T cells during a Plasmodium chabaudi chabaudi (ER) blood-stage infection in mice initiated by the natural route of infection. Malar J 2007; 6:77. [PMID: 17555592 PMCID: PMC1904224 DOI: 10.1186/1475-2875-6-77] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2007] [Accepted: 06/07/2007] [Indexed: 12/23/2022] Open
Abstract
BACKGROUND Investigation of host responses to blood stages of Plasmodium spp, and the immunopathology associated with this phase of the life cycle are often performed on mice infected directly with infected red blood cells. Thus, the effects of mosquito bites and the pre-erythrocytic stages of the parasite, which would be present in natural infection, are ignored In this paper, Plasmodium chabaudi chabaudi infections of mice injected directly with infected red blood cells were compared with those of mice infected by the bites of infected mosquitoes, in order to determine whether the courses of primary infection and splenic CD4 T cell responses are similar. METHODS C57Bl/6 mice were injected with red blood cells infected with P. chabaudi (ER) or infected via the bite of Anopheles stephensi mosquitoes. Parasitaemia were monitored by Giemsa-stained thin blood films. Total spleen cells, CD4+ T cells, and cytokine production (IFN-gamma, IL-2, IL-4, IL-10) were analysed by flow cytometry. In some experiments, mice were subjected to bites of uninfected mosquitoes prior to infectious bites in order to determine whether mosquito bites per se could affect a subsequent P. chabaudi infection. RESULTS P. chabaudi (ER) infections initiated by mosquito bite were characterized by lower parasitaemia of shorter duration than those observed after direct blood challenge. However, splenomegaly was comparable suggesting that parasitaemia alone does not account for the increase in spleen size. Total numbers of CD4 T cells and those producing IFN-gamma, IL-10 and IL-2 were reduced in comparison to direct blood challenge. By contrast, the reduction in IL-4 producing cells was less marked suggesting that there is a proportionally lower Th1-like response in mice infected via infectious mosquitoes. Strikingly, pre-exposure to bites of uninfected mosquitoes reduced the magnitude and duration of the subsequent mosquito-transmitted infection still further, but enhanced the response of CD4 T cells producing IFN-gamma and IL-4. CONCLUSION The data in this paper suggest that studying early host responses in blood stage malaria infections measured after direct blood challenge of mice may not completely reflect the natural situation, and more detailed investigations of blood-stage immunity after mosquito transmission in experimental models should be considered.
Collapse
Affiliation(s)
- Luis Fonseca
- Division of Parasitology, National Institute for Medical Research, London NW7 1AA, UK
| | - Elsa Seixas
- Division of Parasitology, National Institute for Medical Research, London NW7 1AA, UK
- Instituto Gulbenkian de Ciência, Portugal
| | - Geoffrey Butcher
- Faculty of Natural Sciences, Division of Cell and Molecular Biology, Imperial College, London SW7 2AZ, UK
| | - Jean Langhorne
- Division of Parasitology, National Institute for Medical Research, London NW7 1AA, UK
| |
Collapse
|
42
|
Fast MD, Johnson SC, Eddy TD, Pinto D, Ross NW. Lepeophtheirus salmonis secretory/excretory products and their effects on Atlantic salmon immune gene regulation. Parasite Immunol 2007; 29:179-89. [PMID: 17371455 DOI: 10.1111/j.1365-3024.2007.00932.x] [Citation(s) in RCA: 61] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
We have previously shown that Lepeophtheirus salmonis produces trypsin and prostaglandin E(2) (PGE(2)) that are most likely responsible for the limited inflammatory response of Atlantic salmon to infection. After removal of the dopamine and PGE(2), the immunomodulatory activity of unfractionated and pools of the fractionated secretions was determined by examining the effects of the secretions on Atlantic salmon immune gene expression. Incubation of macrophage-enriched isolates of Atlantic salmon head kidney cells with the unfractionated secretion + PGE(2) revealed a significant inhibition of interleukin-1beta (IL-1beta) and major histocompatibility class I gene expression. Inhibition of lipopolysaccharide-induced IL-1beta expression in the Atlantic salmon head kidney cell line (SHK-1) was observed when three pools of the secretory/excretory products were tested. Further purification of products within these pools revealed that fraction 1-2 could account fully for the inhibition of IL-1beta expression in SHK-1 cells observed in pooled fraction 1. This study demonstrates that there are other immunomodulatory compounds produced by L. salmonis, in addition to PGE(2) and trypsin, that can inhibit the expression of Atlantic salmon immune-related genes in vitro.
Collapse
Affiliation(s)
- M D Fast
- Institute for Marine Biosciences, National Research Council, Halifax, NS, Canada.
| | | | | | | | | |
Collapse
|
43
|
Brodie TM, Smith MC, Morris RV, Titus RG. Immunomodulatory effects of the Lutzomyia longipalpis salivary gland protein maxadilan on mouse macrophages. Infect Immun 2007; 75:2359-65. [PMID: 17339357 PMCID: PMC1865772 DOI: 10.1128/iai.01812-06] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Infection with Leishmania major is enhanced when the sand fly Lutzomyia longipalpis salivary peptide maxadilan (MAX) is injected along with the parasite. Here we determined the effect that MAX has on the secretion of cytokines and nitric oxide (NO) and on parasite survival in macrophages (MPhis). The cytokines produced by MPhis can enhance a type 1 response, which will increase NO and the killing of intracellular pathogens such as L. major, or a type 2 response, leading to antibody production that is ineffective against intracellular pathogens such as L. major. A mouse macrophage cell line (RAW 264.7) was stimulated with various concentrations of MAX and lipopolysaccharide (LPS), and the supernatants were collected after 1, 2, and 3 days. Supernatants were assayed for interleukin-12p70 (IL-12p70), IL-10, IL-6, transforming growth factor beta (TGF-beta), NO, and tumor necrosis factor alpha (TNF-alpha). Our results indicate that the addition of MAX upregulates the cytokines associated with a type 2 response (IL-10, IL-6, and TGF-beta) but downregulates type 1 cytokines (IL-12p70 and TNF-alpha) and NO. MAX was also added to L. major-infected mouse peritoneal exudate cells (PECs), and the parasite load increased significantly. The enhanced parasite load correlated with decreased NO production by PECs that were stimulated with LPS and gamma interferon in the presence of MAX. The ability of MAX to foster a type 2 response, to enhance parasite survival, and to decrease NO argues that MAX may be crucial for the early survival of Leishmania in the vertebrate host, and therefore, MAX holds considerable promise as an antigenic component for a vaccine against Leishmania.
Collapse
Affiliation(s)
- Tess M Brodie
- Department of Microbiology, Immunology and Pathology, CVMBS, Colorado State University, 1619 Campus Delivery, Fort Collins, CO 80523-1619, USA.
| | | | | | | |
Collapse
|
44
|
Alarcon-Chaidez FJ, Sun J, Wikel SK. Transcriptome analysis of the salivary glands of Dermacentor andersoni Stiles (Acari: Ixodidae). INSECT BIOCHEMISTRY AND MOLECULAR BIOLOGY 2007; 37:48-71. [PMID: 17175446 DOI: 10.1016/j.ibmb.2006.10.002] [Citation(s) in RCA: 59] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/07/2006] [Revised: 10/10/2006] [Accepted: 10/10/2006] [Indexed: 05/13/2023]
Abstract
Amongst blood-feeding arthropods, ticks of the family Ixodidae (hard ticks) are vectors and reservoirs of a greater variety of infectious agents than any other ectoparasite. Salivary glands of ixodid ticks secrete a large number of pharmacologically active molecules that not only facilitate feeding but also promote establishment of infectious agents. Genomic, proteomic and immunologic characterization of bioactive salivary gland molecules are, therefore, important as they offer new insights into molecular events occurring at the tick-host interface and they have implications for development of novel control strategies. The present work uses complementary DNA (cDNA) sequence analysis to identify salivary gland transcripts expressed by the Rocky Mountain wood tick, Dermacentor andersoni, a vector of the human pathogens causing Rocky Mountain spotted fever, Colorado tick fever, tularemia, and Powassan encephalitis as well as the veterinary pathogen Anaplasma marginale. Dermacentor andersoni is also capable of inducing tick paralysis. Automated single-pass DNA sequencing was conducted on 1440 randomly selected cDNA clones from the salivary glands of adult female D. andersoni collected during the early stages of feeding (18-24h). Analysis of the expressed sequence tags (ESTs) resulted in 544 singletons and 218 clusters with more than one quality read and attempts were made to assign putative functions to tick genes based on amino acid identity to published protein databases. Approximately 25.6% (195) of the sequences showed limited or no homology to previously identified gene products. A number of novel sequences were identified which presented significant sequence similarity to mammalian genes normally associated with extracellular matrix (ECM), regulation of immune responses, tumor suppression, and wound healing. Several coding sequences possessed various degrees of homology to previously described proteins from other tick species. Preliminary nucleotide variation analysis of these and other tick sequences suggests extensive nucleotide diversity, which has implications for evolution of tick feeding. Intra-species diversity studies can be a promising tool for identifying sequence variations potentially associated with phenotypic traits affecting vector-host-pathogen interactions.
Collapse
Affiliation(s)
- Francisco J Alarcon-Chaidez
- School of Medicine, Department of Immunology, University of Connecticut Health Center, 263 Farmington Avenue, MC3710, Farmington, CT 06030, USA.
| | | | | |
Collapse
|
45
|
Koník P, Slavíková V, Salát J, Reznícková J, Dvoroznáková E, Kopecký J. Anti-tumour necrosis factor-? activity in Ixodes ricinus saliva. Parasite Immunol 2006; 28:649-56. [PMID: 17096644 DOI: 10.1111/j.1365-3024.2006.00899.x] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Tumour necrosis factor-alpha (TNF-alpha) is one of the most prominent inflammatory mediators playing a central role in starting off the inflammatory reactions of the innate immune system. We identified a TNF-alpha-inhibitory activity in the saliva and salivary gland extract (SGE) from partially fed Ixodes ricinus ticks. Using mouse and human TNF-alpha specific ELISA, we showed that tick saliva or SGE markedly reduced the level of detectable cytokine. Both saliva and SGE inhibited the cytotoxic effect of TNF-alpha in a bioassay. Elimination of the TNF-alpha-inhibitory activity in SGE by trypsin digestion demonstrated that the anti-TNF-alpha factor is a protein. Fast protein liquid chromatography fractionation of SGE showed one peak of TNF-alpha-inhibitory activity corresponding to a protein with estimated molecular mass 23 kDa. The likely mechanism of the inhibitory effect is a direct binding of the cytokine. The TNF-alpha-inhibitory molecule seems to play an important role in the anti-inflammatory effect of tick saliva at the tick feeding site, providing a gateway to the host for tick-borne pathogens.
Collapse
Affiliation(s)
- P Koník
- Faculty of Biological Sciences, University of South Bohemia, Ceské Budejovice, Czech Republic
| | | | | | | | | | | |
Collapse
|
46
|
Garg R, Juncadella IJ, Ramamoorthi N, Ashish, Ananthanarayanan SK, Thomas V, Rincón M, Krueger JK, Fikrig E, Yengo CM, Anguita J. Cutting edge: CD4 is the receptor for the tick saliva immunosuppressor, Salp15. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2006; 177:6579-83. [PMID: 17082567 PMCID: PMC4302324 DOI: 10.4049/jimmunol.177.10.6579] [Citation(s) in RCA: 94] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Salp15 is an Ixodes scapularis salivary protein that inhibits CD4+ T cell activation through the repression of TCR ligation-triggered calcium fluxes and IL-2 production. We show in this study that Salp15 binds specifically to the CD4 coreceptor on mammalian host T cells. Salp15 specifically associates through its C-terminal residues with the outermost two extracellular domains of CD4. Upon binding to CD4, Salp15 inhibits the subsequent TCR ligation-induced T cell signaling at the earliest steps including tyrosine phosphorylation of the Src kinase Lck, downstream effector proteins, and lipid raft reorganization. These results provide a molecular basis to understanding the immunosuppressive activity of Salp15 and its specificity for CD4+ T cells.
Collapse
Affiliation(s)
- Renu Garg
- Department of Biology, University of North Carolina at Charlotte, Charlotte, NC 28223
| | - Ignacio J. Juncadella
- Department of Biology, University of North Carolina at Charlotte, Charlotte, NC 28223
- Department of Veterinary and Animal Sciences, University of Massachusetts Amherst, Amherst, MA 01003
| | - Nandhini Ramamoorthi
- Department of Internal Medicine, Section of Rheumatology, Yale University School of Medicine, New Haven, CT 06520
| | - Ashish
- Department of Chemistry, University of North Carolina at Charlotte, Charlotte, NC 28223
| | | | - Venetta Thomas
- Department of Internal Medicine, Section of Rheumatology, Yale University School of Medicine, New Haven, CT 06520
| | - Mercedes Rincón
- Department of Internal Medicine, Section of Immunobiology, University of Vermont, Burlington, VT 05405
| | - Joanna K. Krueger
- Department of Chemistry, University of North Carolina at Charlotte, Charlotte, NC 28223
| | - Erol Fikrig
- Department of Internal Medicine, Section of Rheumatology, Yale University School of Medicine, New Haven, CT 06520
| | - Christopher M. Yengo
- Department of Biology, University of North Carolina at Charlotte, Charlotte, NC 28223
| | - Juan Anguita
- Department of Biology, University of North Carolina at Charlotte, Charlotte, NC 28223
- Department of Veterinary and Animal Sciences, University of Massachusetts Amherst, Amherst, MA 01003
| |
Collapse
|
47
|
Caljon G, Van Den Abbeele J, Stijlemans B, Coosemans M, De Baetselier P, Magez S. Tsetse fly saliva accelerates the onset of Trypanosoma brucei infection in a mouse model associated with a reduced host inflammatory response. Infect Immun 2006; 74:6324-30. [PMID: 16954393 PMCID: PMC1695494 DOI: 10.1128/iai.01046-06] [Citation(s) in RCA: 49] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Tsetse flies (Glossina sp.) are the vectors that transmit African trypanosomes, protozoan parasites that cause human sleeping sickness and veterinary infections in the African continent. These blood-feeding dipteran insects deposit saliva at the feeding site that enables the blood-feeding process. Here we demonstrate that tsetse fly saliva also accelerates the onset of a Trypanosoma brucei infection. This effect was associated with a reduced inflammatory reaction at the site of infection initiation (reflected by a decrease of interleukin-6 [IL-6] and IL-12 mRNA) as well as lower serum concentrations of the trypanocidal cytokine tumor necrosis factor. Variant-specific surface glycoprotein-specific antibody isotypes immunoglobulin M (IgM) and IgG2a, implicated in trypanosome clearance, were not suppressed. We propose that tsetse fly saliva accelerates the onset of trypanosome infection by inhibiting local and systemic inflammatory responses involved in parasite control.
Collapse
Affiliation(s)
- Guy Caljon
- Unit of Cellular and Molecular Immunology, Flanders Interuniversity Institute for Biotechnology (VIB), Vrije Universiteit Brussel (VUB), Pleinlaan 2, B-1050 Brussels, Belgium.
| | | | | | | | | | | |
Collapse
|
48
|
Bishop JV, Mejia JS, Pérez de León AA, Tabachnick WJ, Titus RG. Salivary gland extracts of Culicoides sonorensis inhibit murine lymphocyte proliferation and no production by macrophages. Am J Trop Med Hyg 2006; 75:532-6. [PMID: 16968936 PMCID: PMC1624860] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/11/2023] Open
Abstract
Culicoides biting midges serve as vectors of pathogens affecting humans and domestic animals. Culicoides sonorensis is a vector of several arboviruses in North American that cause substantial economic losses to the US livestock industry. Previous studies showed that C. sonorensis saliva, like the saliva of many hematophagous arthropods, contains numerous pharmacological agents that affect hemostasis and early events in the inflammatory response, which may enhance the infectivity of Culicoides-borne pathogens. This paper reports on the immunomodulatory properties of C. sonorensis salivary gland extracts on murine immune cells and discusses the possible immunomodulatory role of C. sonorensis saliva in vesicular stomatitis virus infection of vertebrate hosts. Splenocytes treated with C. sonorensis mitogens were significantly affected in their proliferative response, and peritoneal macrophages secreted significantly less NO. A 66-kDa glycoprotein was purified from C. sonorensis salivary gland extract, which may be in part responsible for these observations and may be considered as a vaccine candidate.
Collapse
Affiliation(s)
- Jeanette V Bishop
- Department of Microbiology, Immunology and Pathology, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, Colorado 80523, USA.
| | | | | | | | | |
Collapse
|
49
|
Abstract
Ticks are of vast medical and veterinary public health importance due to direct damage caused by feeding and their roles in transmitting well known and emerging infectious agents. Ticks and tick-borne pathogens stimulate the immune system of the host. Those immune interactions are of importance in tick biology, pathogen transmission and control of ticks and tick-borne diseases. Both innate and specific acquired immune defenses are involved in the responses of vertebrate hosts to infestation. Ticks have evolved countermeasures to circumvent host immune defenses. This review addresses the immunobiology of the tick-host interface from the perspectives of the pharmacology of tick saliva; relationship of tick saliva to pathogen transmission; host immune responses to infestation; tick modulation of host immune defences; and genomic/proteomic strategies for studying tick salivary gland molecules.
Collapse
Affiliation(s)
- M Brossard
- Institute of Zoology, University of Neuchatel, Emile-Argand 11, CH-2007, Neuchatel, Switzerland
| | | |
Collapse
|
50
|
Macháčková M, Oborník M, Kopecký J. Effect of salivary gland extract from Ixodes ricinus ticks on the proliferation of Borrelia burgdorferi sensu stricto in vivo. Folia Parasitol (Praha) 2006. [DOI: 10.14411/fp.2006.020] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
|