1
|
Mbani Mpega Ntigui CN, Oyegue-Liabagui SL, Mouloungui-Mavoungou J, Ndjangangoye NK, Madoungou Idoumi DL, Kouna LC, Kassa Kassa RF, Moukodoum ND, Ontoua SS, Imboumy Limoukou RK, Biteghe Bi Essone JC, Okouga AP, Bagueboussa F, Lekana-Douki JB. Cytokine pattern during asymptomatic Anaplasma spp. infections and effect of co-infections by malaria and helminths in schoolchildren of Franceville, southeastern Gabon. Parasit Vectors 2025; 18:118. [PMID: 40148890 PMCID: PMC11948865 DOI: 10.1186/s13071-025-06714-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2024] [Accepted: 02/04/2025] [Indexed: 03/29/2025] Open
Abstract
BACKGROUND Asymptomatic infections by Anaplasma spp. and the basis of the immune response during these infections have not yet been established. This study investigated the inflammatory cytokine responses during Anaplasma spp. infection in school children and the effect of co-infection with Plasmodium spp. and helminths. METHODS Blood and stool samples were taken from children aged 5 to 17 years. Parasitological diagnosis was carried out by RDT and microscopy, while microscopy and PCR were used to diagnose infection by Anaplasma spp. Plasma was used for cytokine assays using the ELISA technique. RESULTS A total of 219 children were included in the present study, of whom 205 were infected with Anaplasma spp. and 14 were uninfected. Levels of IL-6, IL-22 and TGF-β were lower not only in children mono-infected with Anaplasma spp. but also in those co-infected with Anaplasma spp. and Plasmodium spp., Anaplasma spp. and helminths, and Anaplasma spp., Plasmodium spp. and helminths compared to controls. However, higher levels of IL-6 and IL-22 were observed in children mono-infected with Anaplasma spp. compared to those co-infected with Anaplasma spp. and helminths. The latter group also had lower levels of IL-6, IL-22, TGF-β and IL-10 than children co-infected with Anaplasma spp. and Plasmodium spp. In addition, children co-infected with Anaplasma spp. and helminths had also lower TGF-β and IL-10 levels than children co-infected with Anaplasma spp., Plasmodium spp. and helminths. An increase of IFN-γ and IL-10 were observed in children co-infected with Anaplasma spp. and Plasmodium spp. compared to those mono-infected with Anaplasma spp. Finally, the results showed that febrile children infected with Anaplasma spp. had higher levels of IFN-γ and lower levels of TGF-β than afebrile children. CONCLUSIONS These results suggest that infection with Anaplasma spp. downregulates cytokines including IL-6, IL-22 and TGF-β and that co-infection with Plasmodium spp. might have a protective effect on the host, while co-infection with helminths might have a negative effect.
Collapse
Affiliation(s)
- Chérone Nancy Mbani Mpega Ntigui
- Unité d'Evolution Epidémiologie et Résistances Parasitaires (UNEEREP), Centre Interdisciplinaire de Recherches Médicales de Franceville (CIRMF), BP 769, Franceville, Gabon.
- Ecole Doctorale Régionale d'Afrique Centrale en Infectiologie Tropicale (ECODRAC), Université des Sciences et Techniques de Masuku, BP 876, Franceville, Gabon.
| | - Sandrine Lydie Oyegue-Liabagui
- Unité d'Evolution Epidémiologie et Résistances Parasitaires (UNEEREP), Centre Interdisciplinaire de Recherches Médicales de Franceville (CIRMF), BP 769, Franceville, Gabon
- Ecole Doctorale Régionale d'Afrique Centrale en Infectiologie Tropicale (ECODRAC), Université des Sciences et Techniques de Masuku, BP 876, Franceville, Gabon
- Département de Biologie, Université des Sciences et Techniques de Masuku (USTM), BP 914, Franceville, Gabon
| | - Jenny Mouloungui-Mavoungou
- Unité d'Evolution Epidémiologie et Résistances Parasitaires (UNEEREP), Centre Interdisciplinaire de Recherches Médicales de Franceville (CIRMF), BP 769, Franceville, Gabon
| | - Nal Kennedy Ndjangangoye
- Unité d'Evolution Epidémiologie et Résistances Parasitaires (UNEEREP), Centre Interdisciplinaire de Recherches Médicales de Franceville (CIRMF), BP 769, Franceville, Gabon
| | - Desly Luide Madoungou Idoumi
- Unité d'Evolution Epidémiologie et Résistances Parasitaires (UNEEREP), Centre Interdisciplinaire de Recherches Médicales de Franceville (CIRMF), BP 769, Franceville, Gabon
| | - Lady Charlene Kouna
- Unité d'Evolution Epidémiologie et Résistances Parasitaires (UNEEREP), Centre Interdisciplinaire de Recherches Médicales de Franceville (CIRMF), BP 769, Franceville, Gabon
| | - Roland Fabrice Kassa Kassa
- Unité de Recherches d'Analyses Médicales (URAM), Centre Interdisciplinaire de Recherches Médicales de Franceville (CIRMF), BP 769, Franceville, Gabon
| | - Nancy Diamella Moukodoum
- Unité d'Evolution Epidémiologie et Résistances Parasitaires (UNEEREP), Centre Interdisciplinaire de Recherches Médicales de Franceville (CIRMF), BP 769, Franceville, Gabon
| | - Steede Seinnat Ontoua
- Unité d'Evolution Epidémiologie et Résistances Parasitaires (UNEEREP), Centre Interdisciplinaire de Recherches Médicales de Franceville (CIRMF), BP 769, Franceville, Gabon
| | - Roméo Karl Imboumy Limoukou
- Unité d'Evolution Epidémiologie et Résistances Parasitaires (UNEEREP), Centre Interdisciplinaire de Recherches Médicales de Franceville (CIRMF), BP 769, Franceville, Gabon
| | - Jean-Claude Biteghe Bi Essone
- Unité d'Evolution Epidémiologie et Résistances Parasitaires (UNEEREP), Centre Interdisciplinaire de Recherches Médicales de Franceville (CIRMF), BP 769, Franceville, Gabon
| | - Alain Prince Okouga
- Unité d'Evolution Epidémiologie et Résistances Parasitaires (UNEEREP), Centre Interdisciplinaire de Recherches Médicales de Franceville (CIRMF), BP 769, Franceville, Gabon
| | - Félicien Bagueboussa
- Unité d'Evolution Epidémiologie et Résistances Parasitaires (UNEEREP), Centre Interdisciplinaire de Recherches Médicales de Franceville (CIRMF), BP 769, Franceville, Gabon
| | - Jean-Bernard Lekana-Douki
- Unité d'Evolution Epidémiologie et Résistances Parasitaires (UNEEREP), Centre Interdisciplinaire de Recherches Médicales de Franceville (CIRMF), BP 769, Franceville, Gabon
- Département de Parasitologie-Mycologie, Université des Sciences de la Santé (USS), BP 4009, Libreville, Gabon
| |
Collapse
|
2
|
Woo GE, Kim MJ, Ahn DH. In vitro and in vivo anti-inflammatory activities of ethanolic extract of Sargassum confusum. Food Sci Biotechnol 2024; 33:181-190. [PMID: 38186611 PMCID: PMC10767179 DOI: 10.1007/s10068-023-01439-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Revised: 09/05/2023] [Accepted: 09/15/2023] [Indexed: 01/09/2024] Open
Abstract
This study was to investigate the anti-inflammatory activity of ethanolic extract of Sargassum confusum (SCEE). LPS is recognized by TLR4 on the macrophage surface, which activates the NF-κB and MAPK signaling pathways, increasing the expression of inflammatory cytokine like interleukin-6 (IL-6), IL-1β, and tumor necrosis factor (TNF)-α and inflammatory mediators such as nitric oxide (NO) and cyclooxygenase-2 (COX-2). The anti-inflammatory effect of SCEE (0, 1, 10, 50 μg/mL) was identified using LPS stimulated RAW 264.7 macrophages. As a result, it was confirmed that inflammatory factors were suppressed by inhibiting expression of COX-2 and iNOS without any cytotoxicity. In addition, phosphorylation of ERK, JNK and NF-κB p65, transcription factors, were inhibited by SCEE does-dependent manner. These results suggest that SCEE has an anti-inflammatory effect and can be used as a material for health functional foods to prevent inflammatory diseases.
Collapse
Affiliation(s)
- Ga Eun Woo
- Department of Food Science & Technology, Pukyong National University, Busan, 48513 Korea
| | - Min Ji Kim
- Department of Food Science & Technology, Pukyong National University, Busan, 48513 Korea
| | - Dong-Hyun Ahn
- Department of Food Science & Technology, Pukyong National University, Busan, 48513 Korea
- Institute of Food Science, Pukyong National University, Busan, 48513 Korea
| |
Collapse
|
3
|
Trouillas P, Franck M. Complete Remission in Paralytic Late Tick-Borne Neurological Disease Comprising Mixed Involvement of Borrelia, Babesia, Anaplasma, and Bartonella: Use of Long-Term Treatments with Antibiotics and Antiparasitics in a Series of 10 Cases. Antibiotics (Basel) 2023; 12:1021. [PMID: 37370340 DOI: 10.3390/antibiotics12061021] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Revised: 04/27/2023] [Accepted: 05/25/2023] [Indexed: 06/29/2023] Open
Abstract
This study aimed to demonstrate that severe neurological motor deficits in the context of late tick-borne disease with mixed microorganism involvement are eligible for long-term combined antibiotic/antiparasitic treatments. The inclusion criteria were: 1. neurological limb paralysis with a disability score >4 according to the EDSS Kurtzke disability scale; 2. serological tests pointing to an involvement of the main tick-borne microorganisms Borrelia burgdorferi s.l., Babesia, Anaplasma, and Bartonella; 3. a general disease for more than 6 months with fatigue, pain and subjective cognitive deficit. The patients were administered long-term treatments with repeated cycles (at least three) of 35-day IV ceftriaxone and repeated oral regimens of azithromycin-doxycycline and azithromycin-doxycycline-rifampicin. For Babesia, repeated courses of atovaquone-azithromycin were administered. Ten patients had intractable or severe motor deficits before treatment in the context of Borrelia (two cases) Borrelia-Babesia (four cases), Borrelia-Babesia-Anaplasma (two cases), Borrelia-Babesia-Anaplasma-Bartonella (one case) and Babesia-Anaplasma (one case). For several months, five had been in wheelchairs, and four had been walking with sticks. Seven patients out of 10 (70%) showed complete remission after a mean active treatment duration of 20.1 + 6.6 months, with a mean number of 4 ceftriaxone cycles. Three patients showed an initial remission but suffered secondary antibiotic/antiparasitic-resistant motor recurrences. Among the nine patients with Borrelia serologic positivity, treatments obtained complete remission in seven cases (77%). The findings of this ten-case series suggest the usefulness of long-term antibiotic/antiparasitic treatments in patients with severe late tick-borne neurological deficits with highly significant elements of tick-borne involvement.
Collapse
Affiliation(s)
| | - Michel Franck
- ADNucleis Biological Laboratory, 69290 Grézieu la Varenne, France
| |
Collapse
|
4
|
Fujisawa S, Murata S, Isezaki M, Win SY, Sato T, Oishi E, Taneno A, Maekawa N, Okagawa T, Konnai S, Ohashi K. Suppressive modulation of host immune responses by Dermanyssus gallinae infestation. Poult Sci 2023; 102:102532. [PMID: 36796246 PMCID: PMC9958498 DOI: 10.1016/j.psj.2023.102532] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2022] [Revised: 12/22/2022] [Accepted: 01/19/2023] [Indexed: 01/27/2023] Open
Abstract
The poultry red mite (Dermanyssus gallinae, PRM) is a blood-sucking ectoparasite in chickens and is one of the most serious threats to poultry farms. Mass infestation with PRMs causes various health problems in chickens, resulting in significant productivity reduction in the poultry industry. Infestation with hematophagous ectoparasites, such as ticks, induces host inflammatory and hemostatic reactions. On the other hand, several studies have reported that hematophagous ectoparasites secrete various immunosuppressants from their saliva to suppress host immune responses to maintain blood sucking. Here, we examined the expression of cytokines in peripheral blood cells to investigate whether PRM infestation affects immunological states in chickens. In PRM-infested chickens, anti-inflammatory cytokines, IL-10 and TGF-β1, and immune checkpoint molecules, CTLA-4 and PD-1, were highly expressed compared to noninfested chickens. PRM-derived soluble mite extracts (SME) upregulated the gene expression of IL-10 in peripheral blood cells and HD-11 chicken macrophages. In addition, SME suppressed the expression of interferons and inflammatory cytokines in HD-11 chicken macrophages. Moreover, SME induces the polarization of macrophages into anti-inflammatory phenotypes. Collectively, PRM infestation could affect host immune responses, especially suppress the inflammatory responses. Further studies are warranted to fully understand the influence of PRM infestation on host immunity.
Collapse
Affiliation(s)
- Sotaro Fujisawa
- Department of Disease Control, Faculty of Veterinary Medicine, Hokkaido University, Sapporo, Japan
| | - Shiro Murata
- Department of Disease Control, Faculty of Veterinary Medicine, Hokkaido University, Sapporo, Japan; Department of Advanced Pharmaceutics, Faculty of Veterinary Medicine, Hokkaido University, Sapporo, Japan.
| | - Masayoshi Isezaki
- Department of Disease Control, Faculty of Veterinary Medicine, Hokkaido University, Sapporo, Japan
| | - Shwe Yee Win
- Department of Disease Control, Faculty of Veterinary Medicine, Hokkaido University, Sapporo, Japan
| | - Takumi Sato
- Vaxxinova Japan K.K., Minato-ku, Tokyo, Japan
| | - Eiji Oishi
- Vaxxinova Japan K.K., Minato-ku, Tokyo, Japan
| | | | - Naoya Maekawa
- Department of Advanced Pharmaceutics, Faculty of Veterinary Medicine, Hokkaido University, Sapporo, Japan
| | - Tomohiro Okagawa
- Department of Advanced Pharmaceutics, Faculty of Veterinary Medicine, Hokkaido University, Sapporo, Japan
| | - Satoru Konnai
- Department of Disease Control, Faculty of Veterinary Medicine, Hokkaido University, Sapporo, Japan,Department of Advanced Pharmaceutics, Faculty of Veterinary Medicine, Hokkaido University, Sapporo, Japan
| | - Kazuhiko Ohashi
- Department of Disease Control, Faculty of Veterinary Medicine, Hokkaido University, Sapporo, Japan,Department of Advanced Pharmaceutics, Faculty of Veterinary Medicine, Hokkaido University, Sapporo, Japan,International Affairs Office, Faculty of Veterinary Medicine, Hokkaido University, Sapporo, Japan
| |
Collapse
|
5
|
Fasae KD, Neelakanta G, Sultana H. Alterations in arthropod and neuronal exosomes reduce virus transmission and replication in recipient cells. EXTRACELLULAR VESICLES AND CIRCULATING NUCLEIC ACIDS 2022; 3:247-279. [PMID: 36939419 PMCID: PMC10018778 DOI: 10.20517/evcna.2022.30] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
Aim Targeting the modes of pathogen shedding/transmission via exosomes or extracellular vesicles has been envisioned as the best approach to control vector-borne diseases. This study is focused on altering exosomes stability to affect the pathogen transmission from infected to naïve recipient cells. Methods In this study, neuronal or arthropod exosomes were treated at different temperatures or with different salts or pH conditions to analyze their ability and efficiency in the transmission of tick-borne Langat virus (LGTV) from infected to naïve recipient cells. Results Quantitative real-time PCR (qRT-PCR) and immunoblotting analyses revealed that treatment of neuronal or tick exosomes at warmer temperatures of 37 °C or 23 °C, respectively, or with sulfate salts such as Magnesium or Ammonium sulfates or with highly alkaline pH of 9 or 11.5, dramatically reduced transmission of LGTV via infectious exosomes (human or tick cells-derived) to human neuronal (SH-SY5Y) cells or skin keratinocytes (HaCaT cells), respectively. Conclusion Overall, this study suggests that exosome-mediated viral transmission of vector-borne pathogens to the vertebrate host or the viral dissemination and replication within or between the mammalian host can be reduced by altering the ability of exosomes with basic changes in temperatures, salts or pH conditions.
Collapse
Affiliation(s)
- Kehinde Damilare Fasae
- Department of Biomedical and Diagnostic Sciences, College of Veterinary Medicine, University of Tennessee, Knoxville, TN 37996, USA
| | - Girish Neelakanta
- Department of Biomedical and Diagnostic Sciences, College of Veterinary Medicine, University of Tennessee, Knoxville, TN 37996, USA
| | - Hameeda Sultana
- Department of Biomedical and Diagnostic Sciences, College of Veterinary Medicine, University of Tennessee, Knoxville, TN 37996, USA
| |
Collapse
|
6
|
O'Neal AJ, Singh N, Mendes MT, Pedra JHF. The genus Anaplasma: drawing back the curtain on tick-pathogen interactions. Pathog Dis 2021; 79:ftab022. [PMID: 33792663 PMCID: PMC8062235 DOI: 10.1093/femspd/ftab022] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2021] [Accepted: 03/30/2021] [Indexed: 12/11/2022] Open
Abstract
Tick-borne illnesses pose a serious concern to human and veterinary health and their prevalence is on the rise. The interactions between ticks and the pathogens they carry are largely undefined. However, the genus Anaplasma, a group of tick-borne bacteria, has been instrumental in uncovering novel paradigms in tick biology. The emergence of sophisticated technologies and the convergence of entomology with microbiology, immunology, metabolism and systems biology has brought tick-Anaplasma interactions to the forefront of vector biology with broader implications for the infectious disease community. Here, we discuss the use of Anaplasma as an instrument for the elucidation of novel principles in arthropod-microbe interactions. We offer an outlook of the primary areas of study, outstanding questions and future research directions.
Collapse
Affiliation(s)
- Anya J O'Neal
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Nisha Singh
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Maria Tays Mendes
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Joao H F Pedra
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, MD, USA
| |
Collapse
|
7
|
Fisher JR, Chroust ZD, Onyoni F, Soong L. Pattern Recognition Receptors in Innate Immunity to Obligate Intracellular Bacteria. ZOONOSES (BURLINGTON, MASS.) 2021; 1:10. [PMID: 35282331 PMCID: PMC8909792 DOI: 10.15212/zoonoses-2021-0011] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Host pattern recognition receptors (PRRs) are crucial for sensing pathogenic microorganisms, launching innate responses, and shaping pathogen-specific adaptive immunity during infection. Rickettsia spp., Orientia tsutsugamushi, Anaplasma spp., Ehrlichia spp., and Coxiella burnetii are obligate intracellular bacteria, which can only replicate within host cells and must evade immune detection to successfully propagate. These five bacterial species are zoonotic pathogens of clinical or agricultural importance, yet, uncovering how immune recognition occurs has remained challenging. Recent evidence from in-vitro studies and animal models has offered new insights into the types and kinetics of PRR activation during infection with Rickettsia spp., A. phagocytophilum, E. chaffeensis, and C. burnetii, respectively. However, much less is known in these regards for O. tsutsugamushi infection, until the recent discovery for the role of the C-type lectin receptor Mincle during lethal infection in mice and in primary macrophage cultures. This review gives a brief summary for clinical and epidemiologic features of these five bacterial infections, focuses on fundamental biologic facets of infection, and recent advances in host recognition. In addition, we discuss knowledge gaps for innate recognition of these bacteria in the context of disease pathogenesis.
Collapse
Affiliation(s)
- James R. Fisher
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, Texas, USA
- School of Medicine, University of Texas Medical Branch, Galveston, Texas, USA
| | - Zachary D. Chroust
- School of Medicine, University of Texas Medical Branch, Galveston, Texas, USA
| | - Florence Onyoni
- Graduate School of Biomedical Sciences, University of Texas Medical Branch, Galveston, Texas, USA
| | - Lynn Soong
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, Texas, USA
- Department of Pathology, University of Texas Medical Branch, Galveston, Texas, USA
- Institute for Human Infections and Immunity, University of Texas Medical Branch, Galveston, Texas, USA
- Corresponding author: Lynn Soong, Department of Microbiology and Immunology, University of Texas Medical Branch, 301 University Blvd. MRB 3.142, Galveston, Texas 77555-1070,
| |
Collapse
|
8
|
Bakshi M, Kim TK, Porter L, Mwangi W, Mulenga A. Amblyomma americanum ticks utilizes countervailing pro and anti-inflammatory proteins to evade host defense. PLoS Pathog 2019; 15:e1008128. [PMID: 31756216 PMCID: PMC6897422 DOI: 10.1371/journal.ppat.1008128] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2019] [Revised: 12/06/2019] [Accepted: 10/05/2019] [Indexed: 02/07/2023] Open
Abstract
Feeding and transmission of tick-borne disease (TBD) agents by ticks are facilitated by tick saliva proteins (TSP). Thus, defining functional roles of TSPs in tick evasion is expected to reveal potential targets in tick-antigen based vaccines to prevent TBD infections. This study describes two types of Amblyomma americanum TSPs: those that are similar to LPS activate macrophage (MΦ) to express pro-inflammation (PI) markers and another set that suppresses PI marker expression by activated MΦ. We show that similar to LPS, three recombinant (r) A. americanum insulin-like growth factor binding-related proteins (rAamIGFBP-rP1, rAamIGFBP-rP6S, and rAamIGFBP-rP6L), hereafter designated as PI-rTSPs, stimulated both PBMC -derived MΦ and mice RAW 267.4 MΦ to express PI co-stimulatory markers, CD40, CD80, and CD86 and cytokines, TNFα, IL-1, and IL-6. In contrast, two A. americanum tick saliva serine protease inhibitors (serpins), AAS27 and AAS41, hereafter designated as anti-inflammatory (AI) rTSPs, on their own did not affect MΦ function or suppress expression of PI markers, but enhanced expression of AI cytokines (IL-10 and TGFβ) in MΦ that were pre-activated by LPS or PI-rTSPs. Mice paw edema test demonstrated that in vitro validated PI- and AI-rTSPs are functional in vivo since injection of HEK293-expressed PI-rTSPs (individually or as a cocktail) induced edema comparable to carrageenan-induced edema and was characterized by upregulation of CD40, CD80, CD86, TNF-α, IL-1, IL-6, and chemokines: CXCL1, CCL2, CCL3, CCL5, and CCL11, whereas the AI-rTSPs (individually and cocktail) were suppressive. We propose that the tick may utilize countervailing PI and AI TSPs to regulate evasion of host immune defenses whereby TSPs such as rAamIGFBP-rPs activate host immune cells and proteins such as AAS27 and AAS41 suppress the activated immune cells. Several studies have documented immuno-suppressive activities in whole tick saliva and salivary gland protein extracts. We have made contribution toward understanding the molecular basis of tick feeding, as we have described functions of defined tick saliva immuno-modulatory proteins. We have shown that A. americanum injects two groups of functionally opposed tick saliva proteins: those that could counter-intuitively be characterized as pro-host defense, and those that are expected to have anti-host immune defense functions. Based on our data, we propose that the tick evades host defense using countervailing pro- and anti- inflammatory proteins in which the pro-host defense tick saliva proteins stimulate host immune cells such as macrophages, and the anti-host defense tick saliva proteins suppress functions of the activated immune cells.
Collapse
Affiliation(s)
- Mariam Bakshi
- Department of Veterinary Pathobiology, College of Veterinary Medicine, TAMU, College Station, Texas, United States of America
| | - Tae Kwon Kim
- Department of Veterinary Pathobiology, College of Veterinary Medicine, TAMU, College Station, Texas, United States of America
| | - Lindsay Porter
- Department of Veterinary Pathobiology, College of Veterinary Medicine, TAMU, College Station, Texas, United States of America
| | - Waithaka Mwangi
- Department of Veterinary Pathobiology, College of Veterinary Medicine, TAMU, College Station, Texas, United States of America
- Department of Diagnostic Medicine/Pathobiology, College of Veterinary Medicine, Kansas State University, Manhattan, Kansas, United States of America
| | - Albert Mulenga
- Department of Veterinary Pathobiology, College of Veterinary Medicine, TAMU, College Station, Texas, United States of America
- * E-mail:
| |
Collapse
|
9
|
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
|
10
|
Islam A, Emran TB, Yamamoto DS, Iyori M, Amelia F, Yusuf Y, Yamaguchi R, Alam MS, Silveira H, Yoshida S. Anopheline antiplatelet protein from mosquito saliva regulates blood feeding behavior. Sci Rep 2019; 9:3129. [PMID: 30816309 PMCID: PMC6395645 DOI: 10.1038/s41598-019-39960-2] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2018] [Accepted: 02/01/2019] [Indexed: 01/23/2023] Open
Abstract
The saliva of hematophagous arthropods is enriched with a complex mixture of antihemostatic molecules, the biological functions of which are largely unknown. Anopheline antiplatelet protein (AAPP) from malaria vector mosquito exhibits strong antiplatelet activity when bound directly to host collagen by its C-terminus and through its N-terminus with Ca2+-binding activity. To investigate the biological functions of AAPP in blood feeding behavior and malaria transmission, we generated transgenic Anopheles stephensi mosquito lines expressing anti-AAPP antibody single-chain fragment (scFv) in their salivary glands. The AAPP-specific collagen-binding activity was completely abolished by AAPP-scFv complex formation in the saliva. Probing and prediuresis time, feeding success, blood meal size, and fecundity, which are all fitness characteristics, were significantly reduced in the transgenic mosquitoes. However, oocysts number in these mosquitoes were not significantly reduced following blood meal intake from Plasmodium berghei-infected mice. These results show that although AAPP plays an important role in mosquito blood feeding, its neutralizing activity did not affect sporogonic development in our laboratory model, but its high fitness cost would pose a survival risk for parasite-infected mosquitoes in nature.
Collapse
Affiliation(s)
- Ashekul Islam
- Laboratory of Vaccinology and Applied Immunology, Kanazawa University School of Pharmacy, Kakuma-machi, Kanazawa, 920-1192, Japan
| | - Talha Bin Emran
- Laboratory of Vaccinology and Applied Immunology, Kanazawa University School of Pharmacy, Kakuma-machi, Kanazawa, 920-1192, Japan
| | - Daisuke S Yamamoto
- Division of Medical Zoology, Department of Infection and Immunity, Jichi Medical University, 3311-1 Yakushiji, Shimotsuke, 329-0431, Japan
| | - Mitsuhiro Iyori
- Laboratory of Vaccinology and Applied Immunology, Kanazawa University School of Pharmacy, Kakuma-machi, Kanazawa, 920-1192, Japan
| | - Fitri Amelia
- Laboratory of Vaccinology and Applied Immunology, Kanazawa University School of Pharmacy, Kakuma-machi, Kanazawa, 920-1192, Japan
| | - Yenni Yusuf
- Laboratory of Vaccinology and Applied Immunology, Kanazawa University School of Pharmacy, Kakuma-machi, Kanazawa, 920-1192, Japan
| | - Ririka Yamaguchi
- Laboratory of Vaccinology and Applied Immunology, Kanazawa University School of Pharmacy, Kakuma-machi, Kanazawa, 920-1192, Japan
| | - Md Shah Alam
- Laboratory of Ecology, Graduate School of Natural Science and Technology, Kanazawa University, Kanazawa, 920-1192, Japan
| | - Henrique Silveira
- Laboratory of Vector-borne diseases and Pathogens, Global Health and Tropical Medicine, Instituto de Higiene e Medicina Tropical, Universidade Nova de Lisboa, Lisbon, 1099-085, Portugal
| | - Shigeto Yoshida
- Laboratory of Vaccinology and Applied Immunology, Kanazawa University School of Pharmacy, Kakuma-machi, Kanazawa, 920-1192, Japan.
| |
Collapse
|
11
|
Inward rectifier potassium (Kir) channels mediate salivary gland function and blood feeding in the lone star tick, Amblyomma americanum. PLoS Negl Trop Dis 2019; 13:e0007153. [PMID: 30730880 PMCID: PMC6382211 DOI: 10.1371/journal.pntd.0007153] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2018] [Revised: 02/20/2019] [Accepted: 01/13/2019] [Indexed: 02/06/2023] Open
Abstract
Background Tick feeding causes extreme morbidity and mortality to humans through transmission of pathogens and causes severe economic losses to the agricultural industry by reducing livestock yield. Salivary gland secretions are essential for tick feeding and thus, reducing or preventing saliva secretions into the vertebrate host is likely to reduce feeding and hinder pathogen life cycles. Unfortunately, the membrane physiology of tick salivary glands is underexplored and this gap in knowledge limits the development of novel therapeutics for inducing cessation of tick feeding. Methodology We studied the influence of inward rectifier potassium (Kir) channel subtypes to the functional capacity of the isolated tick salivary gland through the use of a modified Ramsay assay. The secreted saliva was subsequently used for quantification of the elemental composition of the secreted saliva after the glands were exposed to K+ channel modulators as a measure of osmoregulatory capacity. Lastly, changes to blood feeding behavior and mortality were measured with the use of a membrane feeding system. Principal findings In this study, we characterized the fundamental role of Kir channel subtypes in tick salivary gland function and provide evidence that pharmacological inhibition of these ion channels reduces the secretory activity of the Amblyomma americanum salivary gland. The reduced secretory capacity of the salivary gland was directly correlated with a dramatic reduction of blood ingestion during feeding. Further, exposure to small-molecule modulators of Kir channel subtypes induced mortality to ticks that is likely resultant from an altered osmoregulatory capacity. Conclusions Our data contribute to understanding of tick salivary gland function and could guide future campaigns aiming to develop chemical or reverse vaccinology technologies to reduce the worldwide burden of tick feeding and tick-vectored pathogens. Tick feeding results in negative health and economic consequences worldwide and there has been continued interest in the development of products with novel mechanisms of action for control of tick populations. Kir channels have been shown to be a significant ion conductance pathway in arthropods and are critical for proper functioning of multiple biological processes. Previous work on insect Kir channels has focused on their physiological roles in renal system of mosquitoes and the data suggest that these channels represent a viable pathway to induce renal failure that leads to mortality. Based on the functional and cellular similarities of arthropod salivary glands and Malpighian tubules, we hypothesized that Kir channels constitute a critical conductance pathway within arthropod salivary glands and inhibition of this pathway will preclude feeding. Data presented in this study show that pharmacological modulators of Kir channels elicited a significant reduction in the fluid and ion secretory activity of tick salivary glands that resulted in reduced feeding, altered osmoregulation, and lead to mortality. These data could guide the future development of novel acaricides, RNAi, or genetically modified ticks to mitigate health and economic damages resulting from their feeding. Further, these data indicate a conserved function of Kir channels within multiple tissues of taxonomically diverse organisms, such as ticks and humans.
Collapse
|
12
|
Manning JE, Cantaert T. Time to Micromanage the Pathogen-Host-Vector Interface: Considerations for Vaccine Development. Vaccines (Basel) 2019; 7:E10. [PMID: 30669682 PMCID: PMC6466432 DOI: 10.3390/vaccines7010010] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2018] [Revised: 01/10/2019] [Accepted: 01/16/2019] [Indexed: 12/18/2022] Open
Abstract
The current increase in vector-borne disease worldwide necessitates novel approaches to vaccine development targeted to pathogens delivered by blood-feeding arthropod vectors into the host skin. A concept that is gaining traction in recent years is the contribution of the vector or vector-derived components, like salivary proteins, to host-pathogen interactions. Indeed, the triad of vector-host-pathogen interactions in the skin microenvironment can influence host innate and adaptive responses alike, providing an advantage to the pathogen to establish infection. A better understanding of this "bite site" microenvironment, along with how host and vector local microbiomes immunomodulate responses to pathogens, is required for future vaccines for vector-borne diseases. Microneedle administration of such vaccines may more closely mimic vector deposition of pathogen and saliva into the skin with the added benefit of near painless vaccine delivery. Focusing on the 'micro'⁻from microenvironments to microbiomes to microneedles⁻may yield an improved generation of vector-borne disease vaccines in today's increasingly complex world.
Collapse
Affiliation(s)
- Jessica E Manning
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Phnom Penh 12201, Cambodia.
| | - Tineke Cantaert
- Immunology Unit, Institut Pasteur du Cambodge, Institut Pasteur International Network, Phnom Penh 12201, Cambodia.
| |
Collapse
|
13
|
Abreu MRD, Pereira MC, Simioni PU, Nodari EF, Paiatto LN, Camargo-Mathias MI. Immunomodulatory and morphophysiological effects of Rhipicephalus sanguineus s. l. (Acari: Ixodidae) salivary gland extracts. Vet Immunol Immunopathol 2018; 207:36-45. [PMID: 30593349 DOI: 10.1016/j.vetimm.2018.11.017] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2018] [Revised: 10/03/2018] [Accepted: 11/23/2018] [Indexed: 02/02/2023]
Abstract
Rhipicephalus sanguineus s. l. is popularly known as the "brown dog tick" since dogs are its preferential hosts, but the species has been reported to parasitize other mammals, including humans, with significant medical-veterinary importance since it transmits several important pathogenic agents during the feeding period. The tick saliva is a complex mixture that has several functions, including the capability to modulate the hemostatic, inflammatory and immunologic systems of the host, allowing pathogens to settle. Despite knowledge about the immunosuppressive action of tick saliva, little is known about the mechanisms involved in this process and the morphophysiological effects caused by exposure to the salivary gland extract, taking into consideration the different periods of the glandular cycle. Thus, the objective of this study was to analyze the in vitro effects of salivary gland extracts obtained from R. sanguineus s. l. females fed on host rabbits for two (SGE2 - Salivary Gland Extracts of 2 days) and four days (SGE4 - Salivary Gland Extracts of 4 days) on J774 cells (monocyte macrophage cell line) and verify the occurrence of morphological and immunomodulatory alterations in these cells when exposed to different concentrations of these extracts. The results showed that: (i) SGE2 and SGE4 at the concentration of 4 μg/mL presented cytotoxicity to the J774 cells exposed for 24 and 48 hours; (ii) SGE2 at the concentrations of 2 μg/mL(48-hour exposure) and 1 μg/mL (24-hour exposure) and SGE4 at the concentrations of 2 and 1 μg/mL (48-hour exposure) showed proinflammatory activity, confirmed by the increased secretion of NO and proinflammatory cytokine (IL-2), and the presence of morphological characteristics detected by microscopy; and (iii) SGE2 and SGE4 at the concentrations of 0.5 and 0.1 μg/mL had immunomodulatory activity, demonstrated by decreases in the secretion of NO and proinflammatory cytokines (IL2, IL-6 and TNF-α) and increase in the synthesis of IL-10, confirmed by the morphophysiological analysis. These unprecedented data are extremely relevant for future research to identify the processes involved in the ectoparasite-host relationship, as well to develop more efficient tick control strategies.
Collapse
Affiliation(s)
- Marina Rodrigues de Abreu
- Biology Department, Biosciences Institute, São Paulo State University (UNESP), Avenida 24 A, 1515, 13506-900 Rio Claro, SP, Brazil
| | - Melissa Carolina Pereira
- Biology Department, Biosciences Institute, São Paulo State University (UNESP), Avenida 24 A, 1515, 13506-900 Rio Claro, SP, Brazil
| | - Patrícia Ucelli Simioni
- Biology Department, Biosciences Institute, São Paulo State University (UNESP), Avenida 24 A, 1515, 13506-900 Rio Claro, SP, Brazil; Department of Biomedical Science, Faculty of Americana, FAM, Americana, SP, Brazil
| | - Elen Fernanda Nodari
- Biology Department, Biosciences Institute, São Paulo State University (UNESP), Avenida 24 A, 1515, 13506-900 Rio Claro, SP, Brazil
| | | | - Maria Izabel Camargo-Mathias
- Biology Department, Biosciences Institute, São Paulo State University (UNESP), Avenida 24 A, 1515, 13506-900 Rio Claro, SP, Brazil.
| |
Collapse
|
14
|
Kannangara DW, Patel P. Report of Non-Lyme, Erythema Migrans Rashes from New Jersey with a Review of Possible Role of Tick Salivary Toxins. Vector Borne Zoonotic Dis 2018; 18:641-652. [PMID: 30129909 DOI: 10.1089/vbz.2018.2278] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Erythema migrans (EM) rashes once considered pathognomonic of Lyme disease (LD) have been reported following bites of arthropods that do not transmit LD and in areas with no LD. Also, EM rashes have been reported in association with organisms other than members of Borrelia burgdorferi sensu lato complex. Arthropod saliva has chemicals that have effects on the host and pathogen transmission. Tick saliva has protein families similar to spiders and scorpions and even substances homologous to those found in snakes and other venomous animals. Ticks "invertebrate pharmacologists" have a sophisticated arsenal of chemicals that assist in blood feeding, pathogen transmission, and suppressing host defenses. No organisms have been isolated from many EM rashes. We propose that tick salivary toxins may play a role in the causation of rashes and laboratory abnormalities in tick-borne diseases. The role of tick salivary toxins needs further exploration. Cases of Lyme-like EM rashes referred to as STARI (Southern Tick-Associated Rash Illness) following bites of the lone star tick, Amblyomma americanum, in the United States have been reported predominantly in Southeastern Missouri and a few in South Carolina, North Carolina, Georgia, and one case each in Mississippi and Long Island, New York. Although there is one report of Borrelia lonestari in a patient with a rash, biopsies of 31 cases of STARI, with cultures and PCR, failed to show a relationship. Distribution of A. americanum, whose bites are associated with STARI, now extends along the East Coast of the United States, including New Jersey, up to the Canadian border. As far as we are aware, there have been no prior reports of Lyme-like rashes in New Jersey. In this study, we present case examples of 2 Lyme-like rashes, variations of EM rashes, and a brief review of studies that suggest a role of tick salivary toxins in tick-borne diseases.
Collapse
Affiliation(s)
| | - Pritiben Patel
- St Luke's Health NetWork , Warren Campus, Phillipsburg, New Jersey
| |
Collapse
|
15
|
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
|
16
|
Abstract
Alphaviruses are transmitted to humans via bites of infected mosquitoes. Although alphaviruses have caused a wide range of outbreaks and crippling disease, the availability of licensed vaccines or antiviral therapies remains limited. Mosquito vectors such as Aedes and Culex are the main culprits in the transmission of alphaviruses. This review explores how mosquito saliva may promote alphavirus infection. Identifying the roles of mosquito-derived factors in alphavirus pathogenesis will generate novel tools to circumvent and control mosquito-borne alphavirus infections in humans.
Collapse
|
17
|
Šimo L, Kazimirova M, Richardson J, Bonnet SI. The Essential Role of Tick Salivary Glands and Saliva in Tick Feeding and Pathogen Transmission. Front Cell Infect Microbiol 2017; 7:281. [PMID: 28690983 PMCID: PMC5479950 DOI: 10.3389/fcimb.2017.00281] [Citation(s) in RCA: 206] [Impact Index Per Article: 25.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2017] [Accepted: 06/08/2017] [Indexed: 12/30/2022] Open
Abstract
As long-term pool feeders, ticks have developed myriad strategies to remain discreetly but solidly attached to their hosts for the duration of their blood meal. The critical biological material that dampens host defenses and facilitates the flow of blood-thus assuring adequate feeding-is tick saliva. Saliva exhibits cytolytic, vasodilator, anticoagulant, anti-inflammatory, and immunosuppressive activity. This essential fluid is secreted by the salivary glands, which also mediate several other biological functions, including secretion of cement and hygroscopic components, as well as the watery component of blood as regards hard ticks. When salivary glands are invaded by tick-borne pathogens, pathogens may be transmitted via saliva, which is injected alternately with blood uptake during the tick bite. Both salivary glands and saliva thus play a key role in transmission of pathogenic microorganisms to vertebrate hosts. During their long co-evolution with ticks and vertebrate hosts, microorganisms have indeed developed various strategies to exploit tick salivary molecules to ensure both acquisition by ticks and transmission, local infection and systemic dissemination within the vertebrate host.
Collapse
Affiliation(s)
- Ladislav Šimo
- UMR BIPAR, INRA, Ecole Nationale Vétérinaire d'Alfort, ANSES, Université Paris-EstMaisons-Alfort, France
| | - Maria Kazimirova
- Institute of Zoology, Slovak Academy of SciencesBratislava, Slovakia
| | - Jennifer Richardson
- UMR Virologie, INRA, Ecole Nationale Vétérinaire d'Alfort, ANSES, Université Paris-EstMaisons-Alfort, France
| | - Sarah I. Bonnet
- UMR BIPAR, INRA, Ecole Nationale Vétérinaire d'Alfort, ANSES, Université Paris-EstMaisons-Alfort, France
| |
Collapse
|
18
|
Deviant Behavior: Tick-Borne Pathogens and Inflammasome Signaling. Vet Sci 2016; 3:vetsci3040027. [PMID: 29056735 PMCID: PMC5606592 DOI: 10.3390/vetsci3040027] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2016] [Revised: 09/22/2016] [Accepted: 09/23/2016] [Indexed: 12/11/2022] Open
Abstract
In the face of an assault, host cells mount an immediate response orchestrated by innate immunity. Two of the best described innate immune signaling networks are the Toll- and the Nod-like receptor pathways. Extensive work has been done characterizing both signaling cascades with several recent advances on the forefront of inflammasome biology. In this review, we will discuss how more commonly-studied pathogens differ from tick-transmitted microbes in the context of Nod-like receptor signaling and inflammasome formation. Because pathogens transmitted by ticks have unique characteristics, we offer the opinion that these microbes can be used to uncover novel principles of Nod-like receptor biology.
Collapse
|
19
|
Shaw DK, Kotsyfakis M, Pedra JHF. For Whom the Bell Tolls (and Nods): Spit-acular Saliva. CURRENT TROPICAL MEDICINE REPORTS 2016; 3:40-50. [PMID: 27547699 DOI: 10.1007/s40475-016-0072-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Having emerged during the early part of the Cretaceous period, ticks are an ancient group of hematophagous ectoparasites with significant veterinary and public health importance worldwide. The success of their life strategy can be attributed, in part, to saliva. As we enter into a scientific era where the collection of massive data sets and structures for biological application is possible, we suggest that understanding the molecular mechanisms that govern the life cycle of ticks is within grasp. With this in mind, we discuss what is currently known regarding the manipulation of Toll-like (TLR) and Nod-like (NLR) receptor signaling pathways by tick salivary proteins, and how these molecules impact pathogen transmission.
Collapse
Affiliation(s)
- Dana K Shaw
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, Maryland, USA.
| | - Michail Kotsyfakis
- Institute of Parasitology, Biology Centre, Czech Academy of Sciences, Budweis, Czech Republic
| | - Joao H F Pedra
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, Maryland, USA.
| |
Collapse
|
20
|
Human Coinfection with Borrelia burgdorferi and Babesia microti in the United States. J Parasitol Res 2015; 2015:587131. [PMID: 26697208 PMCID: PMC4677215 DOI: 10.1155/2015/587131] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2015] [Accepted: 11/08/2015] [Indexed: 11/18/2022] Open
Abstract
Borrelia burgdorferi, the causative agent of Lyme disease, and Babesia microti, a causative agent of babesiosis, are increasingly implicated in the growing tick-borne disease burden in the northeastern United States. These pathogens are transmitted via the bite of an infected tick vector, Ixodes scapularis, which is capable of harboring and inoculating a host with multiple pathogens simultaneously. Clinical presentation of the diseases is heterogeneous and ranges from mild flu-like symptoms to near-fatal cardiac arrhythmias. While the reason for the variability is not known, the possibility exists that concomitant infection with both B. burgdorferi and B. microti may synergistically increase disease severity. In an effort to clarify the current state of understanding regarding coinfection with B. burgdorferi and B. microti, in this review, we discuss the geographical distribution and pathogenesis of Lyme disease and babesiosis in the United States, the immunological response of humans to B. burgdorferi or B. microti infection, the existing knowledge regarding coinfection disease pathology, and critical factors that have led to ambiguity in the literature regarding coinfection, in order to eliminate confusion in future experimental design and investigation.
Collapse
|
21
|
Bernard Q, Gallo RL, Jaulhac B, Nakatsuji T, Luft B, Yang X, Boulanger N. Ixodes tick saliva suppresses the keratinocyte cytokine response to TLR2/TLR3 ligands during early exposure to Lyme borreliosis. Exp Dermatol 2015; 25:26-31. [PMID: 26307945 DOI: 10.1111/exd.12853] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/20/2015] [Indexed: 12/28/2022]
Abstract
Ixodes hard tick induces skin injury by its sophisticated biting process. Its saliva plays a key role to enable an efficient blood meal that lasts for several days. We hypothesized that this feeding process may also be exploited by pathogens to facilitate their transmission, especially in the context of arthropod-borne diseases. To test this, we used Lyme borreliosis as a model. This bacterial infection is caused by Borrelia burgdorferi sensu lato transmitted by Ixodes. We co-incubated Borrelia with human keratinocytes in the presence of poly (I: C), a dsRNA TLR3 agonist generated by skin injury. This induced a strong cytokine response from human primary keratinocytes that was much greater than that induced by Borrelia alone. OspC, a TLR2/1 agonist and a major surface lipoprotein of Borrelia also amplified the process. Interestingly, tick saliva inhibited cytokine responses by keratinocytes to these TLR agonists. We propose that Borrelia uses the immunoprivileged site produced by tick saliva to facilitate its transmission.
Collapse
Affiliation(s)
- Quentin Bernard
- EA7290 Virulence bactérienne précoce: groupe borréliose de Lyme, Fédération de médecine Translationnelle et Faculté de Pharmacie de Strasbourg, Université de Strasbourg, Strasbourg, France
| | - Richard L Gallo
- Department of Dermatology, University of California, San Diego, CA, USA
| | - Benoît Jaulhac
- EA7290 Virulence bactérienne précoce: groupe borréliose de Lyme, Fédération de médecine Translationnelle et Faculté de Pharmacie de Strasbourg, Université de Strasbourg, Strasbourg, France.,Centre National de Reference Borrelia, Centre Hospitalier Universitaire, Strasbourg, France
| | - Teruaki Nakatsuji
- Department of Dermatology, University of California, San Diego, CA, USA
| | - Benjamin Luft
- Department of Medicine, State University of New York, Stony Brook, NY, USA
| | - Xiahoua Yang
- Department of Medicine, State University of New York, Stony Brook, NY, USA
| | - Nathalie Boulanger
- EA7290 Virulence bactérienne précoce: groupe borréliose de Lyme, Fédération de médecine Translationnelle et Faculté de Pharmacie de Strasbourg, Université de Strasbourg, Strasbourg, France.,Centre National de Reference Borrelia, Centre Hospitalier Universitaire, Strasbourg, France
| |
Collapse
|
22
|
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
|
23
|
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
|
24
|
|
25
|
Proteomics informed by transcriptomics identifies novel secreted proteins in Dermacentor andersoni saliva. Int J Parasitol 2014; 44:1029-37. [DOI: 10.1016/j.ijpara.2014.07.003] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2014] [Revised: 07/13/2014] [Accepted: 07/14/2014] [Indexed: 12/19/2022]
|
26
|
Ockenfels B, Michael E, McDowell MA. Meta-analysis of the effects of insect vector saliva on host immune responses and infection of vector-transmitted pathogens: a focus on leishmaniasis. PLoS Negl Trop Dis 2014; 8:e3197. [PMID: 25275509 PMCID: PMC4183472 DOI: 10.1371/journal.pntd.0003197] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2014] [Accepted: 08/18/2014] [Indexed: 11/18/2022] Open
Abstract
A meta-analysis of the effects of vector saliva on the immune response and progression of vector-transmitted disease, specifically with regard to pathology, infection level, and host cytokine levels was conducted. Infection in the absence or presence of saliva in naïve mice was compared. In addition, infection in mice pre-exposed to uninfected vector saliva was compared to infection in unexposed mice. To control for differences in vector and pathogen species, mouse strain, and experimental design, a random effects model was used to compare the ratio of the natural log of the experimental to the control means of the studies. Saliva was demonstrated to enhance pathology, infection level, and the production of Th2 cytokines (IL-4 and IL-10) in naïve mice. This effect was observed across vector/pathogen pairings, whether natural or unnatural, and with single salivary proteins used as a proxy for whole saliva. Saliva pre-exposure was determined to result in less severe leishmaniasis pathology when compared with unexposed mice infected either in the presence or absence of sand fly saliva. The results of further analyses were not significant, but demonstrated trends toward protection and IFN-γ elevation for pre-exposed mice. Arthropod vectors transmit a wide variety of diseases resulting in substantial human morbidity and economic costs worldwide. When hematophagous arthropods blood feed, they release saliva into the host. This saliva elicits a strong immune response and has recently been a focus for vaccine research. There is evidence that the saliva enhances infection in naïve hosts, but that prior exposure to saliva results in less severe infection. This analysis endeavored to determine whether there was a statistically significant enhancement or protective effect with regard to saliva exposure and the progression of disease, and to determine the underlying immune mechanism driving these effects. We found that saliva does indeed enhance infection levels of vector-transmitted pathogens and leishmaniasis pathology in naïve mice and elevates Th2 cytokine levels (IL-4 and IL-10). We also determined that pre-exposure to saliva results in less severe pathology of experimental leishmaniasis in mice. These results are important for vaccine trials and vector control programs, though more studies are needed with regard to pre-exposure.
Collapse
Affiliation(s)
- Brittany Ockenfels
- Eck Institute for Global Health, Department of Biological Sciences, University of Notre Dame, Notre Dame, Indiana, United States of America
| | - Edwin Michael
- Eck Institute for Global Health, Department of Biological Sciences, University of Notre Dame, Notre Dame, Indiana, United States of America
| | - Mary Ann McDowell
- Eck Institute for Global Health, Department of Biological Sciences, University of Notre Dame, Notre Dame, Indiana, United States of America
- * E-mail:
| |
Collapse
|
27
|
Bernard Q, Jaulhac B, Boulanger N. Smuggling across the Border: How Arthropod-Borne Pathogens Evade and Exploit the Host Defense System of the Skin. J Invest Dermatol 2014; 134:1211-1219. [DOI: 10.1038/jid.2014.36] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2013] [Revised: 12/04/2013] [Accepted: 12/28/2013] [Indexed: 12/20/2022]
|
28
|
The tick salivary protein sialostatin L2 inhibits caspase-1-mediated inflammation during Anaplasma phagocytophilum infection. Infect Immun 2014; 82:2553-64. [PMID: 24686067 DOI: 10.1128/iai.01679-14] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Saliva from arthropod vectors facilitates blood feeding by altering host inflammation. Whether arthropod saliva counters inflammasome signaling, a protein scaffold that regulates the activity of caspase-1 and cleavage of interleukin-1β (IL-1β) and IL-18 into mature molecules, remains elusive. In this study, we provide evidence that a tick salivary protein, sialostatin L2, inhibits inflammasome formation during pathogen infection. We show that sialostatin L2 targets caspase-1 activity during host stimulation with the rickettsial agent Anaplasma phagocytophilum. A. phagocytophilum causes macrophage activation and hemophagocytic syndrome features. The effect of sialostatin L2 in macrophages was not due to direct caspase-1 enzymatic inhibition, and it did not rely on nuclear factor κB or cathepsin L signaling. Reactive oxygen species from NADPH oxidase and the Loop2 domain of sialostatin L2 were important for the regulatory process. Altogether, our data expand the knowledge of immunoregulatory pathways of tick salivary proteins and unveil an important finding in inflammasome biology.
Collapse
|
29
|
Liu J, Renneker S, Beyer D, Kullmann B, Seitzer U, Ahmed J, Bakheit MA. Identification and partial characterization of a Salp15 homolog from Ixodes ricinus. Ticks Tick Borne Dis 2014; 5:318-22. [PMID: 24572608 DOI: 10.1016/j.ttbdis.2013.12.004] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2013] [Revised: 12/17/2013] [Accepted: 12/18/2013] [Indexed: 02/03/2023]
Abstract
The immunomodulatory molecule Salp15 is originally described in Ixodes scapularis and has been shown to inhibit CD4 T cell activation. Many Salp15 homologs have been described from Ixodes species, and all were well conserved at C-terminal residues that seem to be essential for the function of the protein. In this study, a gene sequence was amplified from cDNA isolated from engorged female I. ricinus ticks, which was predicted to generate a protein of 12.3 kDa. The protein displayed distinct amino acid differences from previously described I. ricinus Salp15 homologs, with amino acid identity ranging between 46.6% and 93.9%. It was referred to as I. ricinus Salp15-like protein. The protein showed 48.1% sequence identity to I. scapularis Salp15. We analyzed the effect of the recombinant I. ricinus Salp15-like protein on the production of cytokines from human peripheral blood mononuclear cells stimulated with LPS. The recombinant protein exerted no effect on the production of TNF-α and IL-6, but the production of IL-10 was dose-dependently reduced. It can be concluded that I. ricinus Salp15-like protein exerts an immunomodulatory effect on the host. The inhibition of IL-10 production may possibly lead to a retardation of B cell activity.
Collapse
Affiliation(s)
- J Liu
- Division of Veterinary Infection Biology and Immunology, Department of Molecular Infection Biology, Research Center Borstel, Germany
| | - S Renneker
- Division of Veterinary Infection Biology and Immunology, Department of Molecular Infection Biology, Research Center Borstel, Germany
| | - D Beyer
- Division of Veterinary Infection Biology and Immunology, Department of Molecular Infection Biology, Research Center Borstel, Germany
| | - B Kullmann
- Division of Veterinary Infection Biology and Immunology, Department of Molecular Infection Biology, Research Center Borstel, Germany
| | - U Seitzer
- Division of Veterinary Infection Biology and Immunology, Department of Molecular Infection Biology, Research Center Borstel, Germany
| | - J Ahmed
- Division of Veterinary Infection Biology and Immunology, Department of Molecular Infection Biology, Research Center Borstel, Germany
| | - M A Bakheit
- Division of Veterinary Infection Biology and Immunology, Department of Molecular Infection Biology, Research Center Borstel, Germany.
| |
Collapse
|
30
|
Wikel S. Ticks and tick-borne pathogens at the cutaneous interface: host defenses, tick countermeasures, and a suitable environment for pathogen establishment. Front Microbiol 2013; 4:337. [PMID: 24312085 PMCID: PMC3833115 DOI: 10.3389/fmicb.2013.00337] [Citation(s) in RCA: 150] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2013] [Accepted: 10/25/2013] [Indexed: 11/21/2022] Open
Abstract
Ticks are unique among hematophagous arthropods by continuous attachment to host skin and blood feeding for days; complexity and diversity of biologically active molecules differentially expressed in saliva of tick species; their ability to modulate the host defenses of pain and itch, hemostasis, inflammation, innate and adaptive immunity, and wound healing; and, the diverse array of infectious agents they transmit. All of these interactions occur at the cutaneous interface in a complex sequence of carefully choreographed host defense responses and tick countermeasures resulting in an environment that facilitates successful blood feeding and establishment of tick-borne infectious agents within the host. Here, we examine diverse patterns of tick attachment to host skin, blood feeding mechanisms, salivary gland transcriptomes, bioactive molecules in tick saliva, timing of pathogen transmission, and host responses to tick bite. Ticks engage and modulate cutaneous and systemic immune defenses involving keratinocytes, natural killer cells, dendritic cells, T cell subpopulations (Th1, Th2, Th17, Treg), B cells, neutrophils, mast cells, basophils, endothelial cells, cytokines, chemokines, complement, and extracellular matrix. A framework is proposed that integrates tick induced changes of skin immune effectors with their ability to respond to tick-borne pathogens. Implications of these changes are addressed. What are the consequences of tick modulation of host cutaneous defenses? Does diversity of salivary gland transcriptomes determine differential modulation of host inflammation and immune defenses and therefore, in part, the clades of pathogens effectively transmitted by different tick species? Do ticks create an immunologically modified cutaneous environment that enhances specific pathogen establishment? Can tick saliva molecules be used to develop vaccines that block pathogen transmission?
Collapse
Affiliation(s)
- Stephen Wikel
- Department of Medical Sciences, Frank H. Netter MD School of Medicine, Quinnipiac University Hamden, CT, USA
| |
Collapse
|
31
|
Bizzarro B, Barros MS, Maciel C, Gueroni DI, Lino CN, Campopiano J, Kotsyfakis M, Amarante-Mendes GP, Calvo E, Capurro ML, Sá-Nunes A. Effects of Aedes aegypti salivary components on dendritic cell and lymphocyte biology. Parasit Vectors 2013; 6:329. [PMID: 24238038 PMCID: PMC3843549 DOI: 10.1186/1756-3305-6-329] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2013] [Accepted: 11/05/2013] [Indexed: 12/12/2022] Open
Abstract
Background Saliva is a key element of interaction between hematophagous mosquitoes and their vertebrate hosts. In addition to allowing a successful blood meal by neutralizing or delaying hemostatic responses, the salivary cocktail is also able to modulate the effector mechanisms of host immune responses facilitating, in turn, the transmission of several types of microorganisms. Understanding how the mosquito uses its salivary components to circumvent host immunity might help to clarify the mechanisms of transmission of such pathogens and disease establishment. Methods Flow cytometry was used to evaluate if increasing concentrations of A. aegypti salivary gland extract (SGE) affects bone marrow-derived DC differentiation and maturation. Lymphocyte proliferation in the presence of SGE was estimated by a colorimetric assay. Western blot and Annexin V staining assays were used to assess apoptosis in these cells. Naïve and memory cells from mosquito-bite exposed mice or OVA-immunized mice and their respective controls were analyzed by flow cytometry. Results Concentration-response curves were employed to evaluate A. aegypti SGE effects on DC and lymphocyte biology. DCs differentiation from bone marrow precursors, their maturation and function were not directly affected by A. aegypti SGE (concentrations ranging from 2.5 to 40 μg/mL). On the other hand, lymphocytes were very sensitive to the salivary components and died in the presence of A. aegypti SGE, even at concentrations as low as 0.1 μg/mL. In addition, A. aegypti SGE was shown to induce apoptosis in all lymphocyte populations evaluated (CD4+ and CD8+ T cells, and B cells) through a mechanism involving caspase-3 and caspase-8, but not Bim. By using different approaches to generate memory cells, we were able to verify that these cells are resistant to SGE effects. Conclusion Our results show that lymphocytes, and not DCs, are the primary target of A. aegypti salivary components. In the presence of A. aegypti SGE, naïve lymphocyte populations die by apoptosis in a caspase-3- and caspase-8-dependent pathway, while memory cells are selectively more resistant to its effects. The present work contributes to elucidate the activities of A. aegypti salivary molecules on the antigen presenting cell-lymphocyte axis and in the biology of these cells.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | - Anderson Sá-Nunes
- Laboratório de Imunologia Experimental, Departamento de Imunologia, Instituto de Ciências Biomédicas, Universidade de São Paulo, São Paulo, 05508-900, SP, Brazil.
| |
Collapse
|
32
|
Sakhon OS, Severo MS, Kotsyfakis M, Pedra JHF. A Nod to disease vectors: mitigation of pathogen sensing by arthropod saliva. Front Microbiol 2013; 4:308. [PMID: 24155744 PMCID: PMC3801108 DOI: 10.3389/fmicb.2013.00308] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2013] [Accepted: 09/26/2013] [Indexed: 01/29/2023] Open
Abstract
Arthropod saliva possesses anti-hemostatic, anesthetic, and anti-inflammatory properties that facilitate feeding and, inadvertently, dissemination of pathogens. Vector-borne diseases caused by these pathogens affect millions of people each year. Many studies address the impact of arthropod salivary proteins on various immunological components. However, whether and how arthropod saliva counters Nod-like (NLR) sensing remains elusive. NLRs are innate immune pattern recognition molecules involved in detecting microbial molecules and danger signals. Nod1/2 signaling results in activation of the nuclear factor-κB and the mitogen-activated protein kinase pathways. Caspase-1 NLRs regulate the inflammasome~– a protein scaffold that governs the maturation of interleukin (IL)-1β and IL-18. Recently, several vector-borne pathogens have been shown to induce NLR activation in immune cells. Here, we provide a brief overview of NLR signaling and discuss clinically relevant vector-borne pathogens recognized by NLR pathways. We also elaborate on possible anti-inflammatory effects of arthropod saliva on NLR signaling and microbial pathogenesis for the purpose of exchanging research perspectives.
Collapse
Affiliation(s)
- Olivia S Sakhon
- Department of Microbiology and Immunology, University of Maryland School of Medicine Baltimore, MD, USA
| | | | | | | |
Collapse
|
33
|
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
|
34
|
Kazimírová M, Štibrániová I. Tick salivary compounds: their role in modulation of host defences and pathogen transmission. Front Cell Infect Microbiol 2013; 3:43. [PMID: 23971008 PMCID: PMC3747359 DOI: 10.3389/fcimb.2013.00043] [Citation(s) in RCA: 205] [Impact Index Per Article: 17.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2013] [Accepted: 07/26/2013] [Indexed: 01/24/2023] Open
Abstract
Ticks require blood meal to complete development and reproduction. Multifunctional tick salivary glands play a pivotal role in tick feeding and transmission of pathogens. Tick salivary molecules injected into the host modulate host defence responses to the benefit of the feeding ticks. To colonize tick organs, tick-borne microorganisms must overcome several barriers, i.e., tick gut membrane, tick immunity, and moulting. Tick-borne pathogens co-evolved with their vectors and hosts and developed molecular adaptations to avoid adverse effects of tick and host defences. Large gaps exist in the knowledge of survival strategies of tick-borne microorganisms and on the molecular mechanisms of tick-host-pathogen interactions. Prior to transmission to a host, the microorganisms penetrate and multiply in tick salivary glands. As soon as the tick is attached to a host, gene expression and production of salivary molecules is upregulated, primarily to facilitate feeding and avoid tick rejection by the host. Pathogens exploit tick salivary molecules for their survival and multiplication in the vector and transmission to and establishment in the hosts. Promotion of pathogen transmission by bioactive molecules in tick saliva was described as saliva-assisted transmission (SAT). SAT candidates comprise compounds with anti-haemostatic, anti-inflammatory and immunomodulatory functions, but the molecular mechanisms by which they mediate pathogen transmission are largely unknown. To date only a few tick salivary molecules associated with specific pathogen transmission have been identified and their functions partially elucidated. Advanced molecular techniques are applied in studying tick-host-pathogen interactions and provide information on expression of vector and pathogen genes during pathogen acquisition, establishment and transmission. Understanding the molecular events on the tick-host-pathogen interface may lead to development of new strategies to control tick-borne diseases.
Collapse
Affiliation(s)
- Mária Kazimírová
- Institute of Zoology, Slovak Academy of Sciences Bratislava, Slovakia.
| | | |
Collapse
|
35
|
Stuen S, Granquist EG, Silaghi C. Anaplasma phagocytophilum--a widespread multi-host pathogen with highly adaptive strategies. Front Cell Infect Microbiol 2013; 3:31. [PMID: 23885337 PMCID: PMC3717505 DOI: 10.3389/fcimb.2013.00031] [Citation(s) in RCA: 393] [Impact Index Per Article: 32.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2013] [Accepted: 06/30/2013] [Indexed: 11/21/2022] Open
Abstract
The bacterium Anaplasma phagocytophilum has for decades been known to cause the disease tick-borne fever (TBF) in domestic ruminants in Ixodes ricinus-infested areas in northern Europe. In recent years, the bacterium has been found associated with Ixodes-tick species more or less worldwide on the northern hemisphere. A. phagocytophilum has a broad host range and may cause severe disease in several mammalian species, including humans. However, the clinical symptoms vary from subclinical to fatal conditions, and considerable underreporting of clinical incidents is suspected in both human and veterinary medicine. Several variants of A. phagocytophilum have been genetically characterized. Identification and stratification into phylogenetic subfamilies has been based on cell culturing, experimental infections, PCR, and sequencing techniques. However, few genome sequences have been completed so far, thus observations on biological, ecological, and pathological differences between genotypes of the bacterium, have yet to be elucidated by molecular and experimental infection studies. The natural transmission cycles of various A. phagocytophilum variants, the involvement of their respective hosts and vectors involved, in particular the zoonotic potential, have to be unraveled. A. phagocytophilum is able to persist between seasons of tick activity in several mammalian species and movement of hosts and infected ticks on migrating animals or birds may spread the bacterium. In the present review, we focus on the ecology and epidemiology of A. phagocytophilum, especially the role of wildlife in contribution to the spread and sustainability of the infection in domestic livestock and humans.
Collapse
Affiliation(s)
- Snorre Stuen
- Department of Production Animal Clinical Sciences, Norwegian School of Veterinary Science Sandnes, Norway.
| | | | | |
Collapse
|