1
|
Wang L, Liu Y, Pang R, Guo Y, Ren Y, Wu Y, Cao Z. The Tick Saliva Peptide HIDfsin2 TLR4-Dependently Inhibits the Tick-Borne Severe Fever with Thrombocytopenia Syndrome Virus in Mouse Macrophages. Antibiotics (Basel) 2024; 13:449. [PMID: 38786177 PMCID: PMC11117380 DOI: 10.3390/antibiotics13050449] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2024] [Revised: 04/25/2024] [Accepted: 05/06/2024] [Indexed: 05/25/2024] Open
Abstract
Ticks transmit a variety of pathogens to their hosts by feeding on blood. The interactions and struggle between tick pathogens and hosts have evolved bilaterally. The components of tick saliva can directly or indirectly trigger host biological responses in a manner that promotes pathogen transmission; however, host cells continuously develop strategies to combat pathogen infection and transmission. Moreover, it is still unknown how host cells develop their defense strategies against tick-borne viruses during tick sucking. Here, we found that the tick saliva peptide HIDfsin2 enhanced the antiviral innate immunity of mouse macrophages by activating the Toll-like receptor 4 (TLR4) signaling pathway, thereby restricting tick-borne severe fever with thrombocytopenia syndrome virus (SFTSV) replication. HIDfsin2 was identified to interact with lipopolysaccharide (LPS), a ligand of TLR4, and then depolymerize LPS micelles into smaller particles, effectively enhancing the activation of the nuclear factor kappa-B (NF-κB) and type I interferon (IFN-I) signaling pathways, which are downstream of TLR4. Expectedly, TLR4 knockout completely eliminated the promotion effect of HIDfsin2 on NF-κB and type I interferon activation. Moreover, HIDfsin2 enhanced SFTSV replication in TLR4-knockout mouse macrophages, which is consistent with our recent report that HIDfsin2 hijacked p38 mitogen-activated protein kinase (MAPK) to promote the replication of tick-borne SFTSV in A549 and Huh7 cells (human cell lines) with low expression of TLR4. Together, these results provide new insights into the innate immune mechanism of host cells following tick bites. Our study also shows a rare molecular event relating to the mutual antagonism between tick-borne SFTSV and host cells mediated by the tick saliva peptide HIDfsin2 at the tick-host-virus interface.
Collapse
Affiliation(s)
- Luyao Wang
- National “111” Center for Cellular Regulation and Molecular Pharmaceutics, Key Laboratory of Fermentation Engineering (Ministry of Education), Hubei University of Technology, Wuhan 430068, China;
- State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan 430072, China; (Y.L.); (R.P.); (Y.G.); (Y.R.); (Y.W.)
- Shenzhen Research Institute, Wuhan University, Shenzhen 518057, China
| | - Yishuo Liu
- State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan 430072, China; (Y.L.); (R.P.); (Y.G.); (Y.R.); (Y.W.)
| | - Rui Pang
- State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan 430072, China; (Y.L.); (R.P.); (Y.G.); (Y.R.); (Y.W.)
| | - Yiyuan Guo
- State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan 430072, China; (Y.L.); (R.P.); (Y.G.); (Y.R.); (Y.W.)
| | - Yingying Ren
- State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan 430072, China; (Y.L.); (R.P.); (Y.G.); (Y.R.); (Y.W.)
| | - Yingliang Wu
- State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan 430072, China; (Y.L.); (R.P.); (Y.G.); (Y.R.); (Y.W.)
| | - Zhijian Cao
- National “111” Center for Cellular Regulation and Molecular Pharmaceutics, Key Laboratory of Fermentation Engineering (Ministry of Education), Hubei University of Technology, Wuhan 430068, China;
- State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan 430072, China; (Y.L.); (R.P.); (Y.G.); (Y.R.); (Y.W.)
- Shenzhen Research Institute, Wuhan University, Shenzhen 518057, China
| |
Collapse
|
2
|
A Deeper Insight into the Tick Salivary Protein Families under the Light of Alphafold2 and Dali: Introducing the TickSialoFam 2.0 Database. Int J Mol Sci 2022; 23:ijms232415613. [PMID: 36555254 PMCID: PMC9779611 DOI: 10.3390/ijms232415613] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2022] [Revised: 12/05/2022] [Accepted: 12/06/2022] [Indexed: 12/14/2022] Open
Abstract
Hard ticks feed for several days or weeks on their hosts and their saliva contains thousands of polypeptides belonging to dozens of families, as identified by salivary transcriptomes. Comparison of the coding sequences to protein databases helps to identify putative secreted proteins and their potential functions, directing and focusing future studies, usually done with recombinant proteins that are tested in different bioassays. However, many families of putative secreted peptides have a unique character, not providing significant matches to known sequences. The availability of the Alphafold2 program, which provides in silico predictions of the 3D polypeptide structure, coupled with the Dali program which uses the atomic coordinates of a structural model to search the Protein Data Bank (PDB) allows another layer of investigation to annotate and ascribe a functional role to proteins having so far being characterized as "unique". In this study, we analyzed the classification of tick salivary proteins under the light of the Alphafold2/Dali programs, detecting novel protein families and gaining new insights relating the structure and function of tick salivary proteins.
Collapse
|
3
|
Identification of Aedes aegypti salivary gland proteins interacting with human immune receptor proteins. PLoS Negl Trop Dis 2022; 16:e0010743. [PMID: 36070318 PMCID: PMC9484696 DOI: 10.1371/journal.pntd.0010743] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2022] [Revised: 09/19/2022] [Accepted: 08/15/2022] [Indexed: 11/19/2022] Open
Abstract
Mosquito saliva proteins modulate the human immune and hemostatic systems and control mosquito-borne pathogenic infections. One mechanism through which mosquito proteins may influence host immunity and hemostasis is their interactions with key human receptor proteins that may act as receptors for or coordinate attacks against invading pathogens. Here, using pull-down assays and proteomics-based mass spectrometry, we identified 11 Ae. aegypti salivary gland proteins (SGPs) (e.g., apyrase, Ae. aegypti venom allergen-1 [AaVA-1], neutrophil stimulating protein 1 [NeSt1], and D7 proteins), that interact with one or more of five human receptor proteins (cluster of differentiation 4 [CD4], CD14, CD86, dendritic cell-specific intercellular adhesion molecule-3-grabbing non-integrin [DC-SIGN], and Toll-like receptor 4 [TLR4]). We focused on CD4- and DC-SIGN-interacting proteins and confirmed that CD4 directly interacts with AaVA-1, D7, and NeST1 recombinant proteins and that AaVA-1 showed a moderate interaction with DC-SIGN using ELISA. Bacteria responsive protein 1 (AgBR1), an Ae. aegypti saliva protein reported to enhance ZIKV infection in humans but that was not identified in our pull-down assay moderately interacts with CD4 in the ELISA assay. Functionally, we showed that AaVA-1 and NeST1 proteins promoted activation of CD4+ T cells. We propose the possible impact of these interactions and effects on mosquito-borne viral infections such as dengue, Zika, and chikungunya viruses. Overall, this study provides key insight into the vector-host (protein-protein) interaction network and suggests roles for these interactions in mosquito-borne viral infections. Here, we report our results from a pull-down assay and ELISA, which identified Ae. aegypti salivary gland proteins that interact with one or more of five human receptor proteins. Some of these interactions could affect the expression of costimulatory molecules involved in host defense against pathogens. This underscores the potential proviral or antiviral roles of these interactions on mosquito-borne viral infections. Our study provides a preliminary enquiry into the vector (mosquito)-host (human) interaction networks and how this interaction could be further investigated and harnessed as a strategy to augment existing vector-borne diseases control approaches.
Collapse
|
4
|
Maqbool M, Sajid MS, Saqib M, Anjum FR, Tayyab MH, Rizwan HM, Rashid MI, Rashid I, Iqbal A, Siddique RM, Shamim A, Hassan MA, Atif FA, Razzaq A, Zeeshan M, Hussain K, Nisar RHA, Tanveer A, Younas S, Kamran K, Rahman SU. Potential Mechanisms of Transmission of Tick-Borne Viruses at the Virus-Tick Interface. Front Microbiol 2022; 13:846884. [PMID: 35602013 PMCID: PMC9121816 DOI: 10.3389/fmicb.2022.846884] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2021] [Accepted: 03/18/2022] [Indexed: 11/13/2022] Open
Abstract
Ticks (Acari; Ixodidae) are the second most important vector for transmission of pathogens to humans, livestock, and wildlife. Ticks as vectors for viruses have been reported many times over the last 100 years. Tick-borne viruses (TBVs) belong to two orders (Bunyavirales and Mononegavirales) containing nine families (Bunyaviridae, Rhabdoviridae, Asfarviridae, Orthomyxovirida, Reoviridae, Flaviviridae, Phenuviridae, Nyamiviridae, and Nairoviridae). Among these TBVs, some are very pathogenic, causing huge mortality, and hence, deserve to be covered under the umbrella of one health. About 38 viral species are being transmitted by <10% of the tick species of the families Ixodidae and Argasidae. All TBVs are RNA viruses except for the African swine fever virus from the family Asfarviridae. Tick-borne viral diseases have also been classified as an emerging threat to public health and animals, especially in resource-poor communities of the developing world. Tick-host interaction plays an important role in the successful transmission of pathogens. The ticks' salivary glands are the main cellular machinery involved in the uptake, settlement, and multiplication of viruses, which are required for successful transmission into the final host. Furthermore, tick saliva also participates as an augmenting tool during the physiological process of transmission. Tick saliva is an important key element in the successful transmission of pathogens and contains different antimicrobial proteins, e.g., defensin, serine, proteases, and cement protein, which are key players in tick-virus interaction. While tick-virus interaction is a crucial factor in the propagation of tick-borne viral diseases, other factors (physiological, immunological, and gut flora) are also involved. Some immunological factors, e.g., toll-like receptors, scavenger receptors, Janus-kinase (JAK-STAT) pathway, and immunodeficiency (IMD) pathway are involved in tick-virus interaction by helping in virus assembly and acting to increase transmission. Ticks also harbor some endogenous viruses as internal microbial faunas, which also play a significant role in tick-virus interaction. Studies focusing on tick saliva and its role in pathogen transmission, tick feeding, and control of ticks using functional genomics all point toward solutions to this emerging threat. Information regarding tick-virus interaction is somewhat lacking; however, this information is necessary for a complete understanding of transmission TBVs and their persistence in nature. This review encompasses insight into the ecology and vectorial capacity of tick vectors, as well as our current understanding of the predisposing, enabling, precipitating, and reinforcing factors that influence TBV epidemics. The review explores the cellular, biochemical, and immunological tools which ensure and augment successful evading of the ticks' defense systems and transmission of the viruses to the final hosts at the virus-vector interface. The role of functional genomics, proteomics, and metabolomics in profiling tick-virus interaction is also discussed. This review is an initial attempt to comprehensively elaborate on the epidemiological determinants of TBVs with a focus on intra-vector physiological processes involved in the successful execution of the docking, uptake, settlement, replication, and transmission processes of arboviruses. This adds valuable data to the existing bank of knowledge for global stakeholders, policymakers, and the scientific community working to devise appropriate strategies to control ticks and TBVs.
Collapse
Affiliation(s)
- Mahvish Maqbool
- Department of Parasitology, University of Agriculture, Faisalabad, Pakistan
| | - Muhammad Sohail Sajid
- Department of Parasitology, University of Agriculture, Faisalabad, Pakistan
- Department of Epidemiology and Public Health, University of Agriculture, Faisalabad, Pakistan
| | - Muhammad Saqib
- Department of Clinical Medicine and Surgery, University of Agriculture, Faisalabad, Pakistan
| | - Faisal Rasheed Anjum
- Department of Epidemiology and Public Health, University of Agriculture, Faisalabad, Pakistan
- Institute of Microbiology, University of Agriculture, Faisalabad, Pakistan
| | - Muhammad Haleem Tayyab
- Department of Clinical Medicine and Surgery, University of Agriculture, Faisalabad, Pakistan
| | - Hafiz Muhammad Rizwan
- Section of Parasitology, Department of Pathobiology, KBCMA College of Veterinary and Animal Sciences Narowal, Lahore, Pakistan
| | - Muhammad Imran Rashid
- Department of Parasitology, University of Veterinary and Animal Sciences, Lahore, Pakistan
| | - Imaad Rashid
- Department of Clinical Medicine and Surgery, University of Agriculture, Faisalabad, Pakistan
| | - Asif Iqbal
- Section of Parasitology, Department of Pathobiology, Riphah College of Veterinary Sciences, Riphah International University, Lahore, Pakistan
| | - Rao Muhammad Siddique
- Section of Parasitology, Department of Pathobiology, Riphah College of Veterinary Sciences, Riphah International University, Lahore, Pakistan
| | - Asim Shamim
- Department of Pathobiology, University of the Poonch Rawalakot, Rawalakot, Pakistan
| | - Muhammad Adeel Hassan
- Department of Parasitology, Cholistan University of Veterinary and Animal Sciences, Bahawalpur, Pakistan
| | - Farhan Ahmad Atif
- Medicine Section, Department of Clinical Sciences, Collège of Veterinary and Animal Sciences, Jhang, Pakistan
- University of Veterinary and Animal Sciences, Lahore, Pakistan
| | - Abdul Razzaq
- Agricultural Linkages Program, Pakistan Agriculture Research Council, Islamabad, Pakistan
| | - Muhammad Zeeshan
- Department of Parasitology, University of Agriculture, Faisalabad, Pakistan
| | - Kashif Hussain
- Department of Parasitology, University of Agriculture, Faisalabad, Pakistan
| | | | - Akasha Tanveer
- Department of Parasitology, University of Agriculture, Faisalabad, Pakistan
| | - Sahar Younas
- Department of Parasitology, University of Agriculture, Faisalabad, Pakistan
| | - Kashif Kamran
- Department of Zoology, University of Balochistan, Quetta, Pakistan
| | - Sajjad ur Rahman
- Institute of Microbiology, University of Agriculture, Faisalabad, Pakistan
| |
Collapse
|
5
|
Ali A, Zeb I, Alouffi A, Zahid H, Almutairi MM, Ayed Alshammari F, Alrouji M, Termignoni C, Vaz IDS, Tanaka T. Host Immune Responses to Salivary Components - A Critical Facet of Tick-Host Interactions. Front Cell Infect Microbiol 2022; 12:809052. [PMID: 35372098 PMCID: PMC8966233 DOI: 10.3389/fcimb.2022.809052] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2021] [Accepted: 02/04/2022] [Indexed: 12/15/2022] Open
Abstract
Tick sialome is comprised of a rich cocktail of bioactive molecules that function as a tool to disarm host immunity, assist blood-feeding, and play a vibrant role in pathogen transmission. The adaptation of the tick's blood-feeding behavior has lead to the evolution of bioactive molecules in its saliva to assist them to overwhelm hosts' defense mechanisms. During a blood meal, a tick secretes different salivary molecules including vasodilators, platelet aggregation inhibitors, anticoagulants, anti-inflammatory proteins, and inhibitors of complement activation; the salivary repertoire changes to meet various needs such as tick attachment, feeding, and modulation or impairment of the local dynamic and vigorous host responses. For instance, the tick's salivary immunomodulatory and cement proteins facilitate the tick's attachment to the host to enhance prolonged blood-feeding and to modulate the host's innate and adaptive immune responses. Recent advances implemented in the field of "omics" have substantially assisted our understanding of host immune modulation and immune inhibition against the molecular dynamics of tick salivary molecules in a crosstalk between the tick-host interface. A deep understanding of the tick salivary molecules, their substantial roles in multifactorial immunological cascades, variations in secretion, and host immune responses against these molecules is necessary to control these parasites. In this article, we reviewed updated knowledge about the molecular mechanisms underlying host responses to diverse elements in tick saliva throughout tick invasion, as well as host defense strategies. In conclusion, understanding the mechanisms involved in the complex interactions between the tick salivary components and host responses is essential to decipher the host defense mechanisms against the tick evasion strategies at tick-host interface which is promising in the development of effective anti-tick vaccines and drug therapeutics.
Collapse
Affiliation(s)
- Abid Ali
- Department of Zoology, Abdul Wali Khan University Mardan, Mardan, Pakistan
| | - Ismail Zeb
- Department of Zoology, Abdul Wali Khan University Mardan, Mardan, Pakistan
| | - Abdulaziz Alouffi
- King Abdulaziz City for Science and Technology, Riyadh, Saudi Arabia
| | - Hafsa Zahid
- Department of Zoology, Abdul Wali Khan University Mardan, Mardan, Pakistan
| | - Mashal M. Almutairi
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Fahdah Ayed Alshammari
- College of Sciences and Literature Microbiology, Nothern Border University, Rafha, Saudi Arabia
| | - Mohammed Alrouji
- College of Applied Medical Sciences, Shaqra University, Shaqra, Saudi Arabia
| | - Carlos Termignoni
- Centro de Biotecnologia, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Itabajara da Silva Vaz
- Centro de Biotecnologia, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Tetsuya Tanaka
- Laboratory of Infectious Diseases, Joint Faculty of Veterinary Medicine, Kagoshima University, Kagoshima, Japan
| |
Collapse
|
6
|
Broeckel RM, Feldmann F, McNally KL, Chiramel AI, Sturdevant GL, Leung JM, Hanley PW, Lovaglio J, Rosenke R, Scott DP, Saturday G, Bouamr F, Rasmussen AL, Robertson SJ, Best SM. A pigtailed macaque model of Kyasanur Forest disease virus and Alkhurma hemorrhagic disease virus pathogenesis. PLoS Pathog 2021; 17:e1009678. [PMID: 34855915 PMCID: PMC8638978 DOI: 10.1371/journal.ppat.1009678] [Citation(s) in RCA: 4] [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: 05/26/2021] [Accepted: 10/15/2021] [Indexed: 11/18/2022] Open
Abstract
Kyasanur Forest disease virus (KFDV) and the closely related Alkhurma hemorrhagic disease virus (AHFV) are emerging flaviviruses that cause severe viral hemorrhagic fevers in humans. Increasing geographical expansion and case numbers, particularly of KFDV in southwest India, class these viruses as a public health threat. Viral pathogenesis is not well understood and additional vaccines and antivirals are needed to effectively counter the impact of these viruses. However, current animal models of KFDV pathogenesis do not accurately reproduce viral tissue tropism or clinical outcomes observed in humans. Here, we show that pigtailed macaques (Macaca nemestrina) infected with KFDV or AHFV develop viremia that peaks 2 to 4 days following inoculation. Over the course of infection, animals developed lymphocytopenia, thrombocytopenia, and elevated liver enzymes. Infected animals exhibited hallmark signs of human disease characterized by a flushed appearance, piloerection, dehydration, loss of appetite, weakness, and hemorrhagic signs including epistaxis. Virus was commonly present in the gastrointestinal tract, consistent with human disease caused by KFDV and AHFV where gastrointestinal symptoms (hemorrhage, vomiting, diarrhea) are common. Importantly, RNAseq of whole blood revealed that KFDV downregulated gene expression of key clotting factors that was not observed during AHFV infection, consistent with increased severity of KFDV disease observed in this model. This work characterizes a nonhuman primate model for KFDV and AHFV that closely resembles human disease for further utilization in understanding host immunity and development of antiviral countermeasures.
Collapse
MESH Headings
- Animals
- Chlorocebus aethiops
- Cytokines/blood
- Disease Models, Animal
- Encephalitis Viruses, Tick-Borne/genetics
- Encephalitis Viruses, Tick-Borne/immunology
- Encephalitis Viruses, Tick-Borne/pathogenicity
- Encephalitis, Tick-Borne/immunology
- Encephalitis, Tick-Borne/pathology
- Encephalitis, Tick-Borne/virology
- Female
- HEK293 Cells
- Hemorrhagic Fevers, Viral/immunology
- Hemorrhagic Fevers, Viral/pathology
- Hemorrhagic Fevers, Viral/virology
- Humans
- Lymph Nodes/virology
- Macaca nemestrina
- Vero Cells
- Viremia
Collapse
Affiliation(s)
- Rebecca M. Broeckel
- Laboratory of Virology, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, Montana, United States of America
| | - Friederike Feldmann
- Rocky Mountain Veterinary Branch, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, Montana, United States of America
| | - Kristin L. McNally
- Laboratory of Virology, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, Montana, United States of America
| | - Abhilash I. Chiramel
- Laboratory of Virology, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, Montana, United States of America
| | - Gail L. Sturdevant
- Laboratory of Virology, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, Montana, United States of America
| | - Jacqueline M. Leung
- Research Technologies Branch, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, Montana, United States of America
| | - Patrick W. Hanley
- Rocky Mountain Veterinary Branch, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, Montana, United States of America
| | - Jamie Lovaglio
- Rocky Mountain Veterinary Branch, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, Montana, United States of America
| | - Rebecca Rosenke
- Rocky Mountain Veterinary Branch, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, Montana, United States of America
| | - Dana P. Scott
- Rocky Mountain Veterinary Branch, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, Montana, United States of America
| | - Greg Saturday
- Rocky Mountain Veterinary Branch, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, Montana, United States of America
| | - Fadila Bouamr
- Laboratory of Molecular Microbiology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Angela L. Rasmussen
- Vaccine and Infectious Disease Organization, University of Saskatchewan, Saskatoon, Saskatchewan, Canada
- Center for Global Health Science and Security, Georgetown University, Washington, District of Columbia, United States of America
| | - Shelly J. Robertson
- Laboratory of Virology, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, Montana, United States of America
| | - Sonja M. Best
- Laboratory of Virology, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, Montana, United States of America
- * E-mail:
| |
Collapse
|
7
|
Denisov SS, Dijkgraaf I. Immunomodulatory Proteins in Tick Saliva From a Structural Perspective. Front Cell Infect Microbiol 2021; 11:769574. [PMID: 34722347 PMCID: PMC8548845 DOI: 10.3389/fcimb.2021.769574] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2021] [Accepted: 09/28/2021] [Indexed: 12/25/2022] Open
Abstract
To feed successfully, ticks must bypass or suppress the host’s defense mechanisms, particularly the immune system. To accomplish this, ticks secrete specialized immunomodulatory proteins into their saliva, just like many other blood-sucking parasites. However, the strategy of ticks is rather unique compared to their counterparts. Ticks’ tendency for gene duplication has led to a diverse arsenal of dozens of closely related proteins from several classes to modulate the immune system’s response. Among these are chemokine-binding proteins, complement pathways inhibitors, ion channels modulators, and numerous poorly characterized proteins whose functions are yet to be uncovered. Studying tick immunomodulatory proteins would not only help to elucidate tick-host relationships but would also provide a rich pool of potential candidates for the development of immunomodulatory intervention drugs and potentially new vaccines. In the present review, we will attempt to summarize novel findings on the salivary immunomodulatory proteins of ticks, focusing on biomolecular targets, structure-activity relationships, and the perspective of their development into therapeutics.
Collapse
Affiliation(s)
- Stepan S Denisov
- Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), University of Maastricht, Maastricht, Netherlands
| | - Ingrid Dijkgraaf
- Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), University of Maastricht, Maastricht, Netherlands
| |
Collapse
|
8
|
Dinkel KD, Herndon DR, Noh SM, Lahmers KK, Todd SM, Ueti MW, Scoles GA, Mason KL, Fry LM. A U.S. isolate of Theileria orientalis, Ikeda genotype, is transmitted to cattle by the invasive Asian longhorned tick, Haemaphysalis longicornis. Parasit Vectors 2021; 14:157. [PMID: 33726815 PMCID: PMC7962341 DOI: 10.1186/s13071-021-04659-9] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2020] [Accepted: 02/25/2021] [Indexed: 12/16/2022] Open
Abstract
Background Theileria orientalis is a tick-borne hemoparasite that causes anemia, ill thrift, and death in cattle globally. The Ikeda strain of T.orientalis is more virulent than other strains, leading to severe clinical signs and death of up to 5% of affected animals. Within the Asia–Pacific region, where it affects 25% of Australian cattle, T.orientalis Ikeda has a significant economic impact on the cattle industry. In 2017, T.orientalis Ikeda was detected in a cattle herd in Albermarle County, Virginia, United States. Months earlier, the U.S. was alerted to the invasion of the Asian longhorned tick, Haemaphysalis longicornis, throughout the eastern U.S. Abundant H.longicornis ticks were identified on cattle in the T.orientalis-affected herd in VA, and a subset of ticks from the environment were PCR-positive for T.orientalis Ikeda. A strain of T.orientalis from a previous U.S. outbreak was not transmissible by H.longicornis; however, H.longicornis is the primary tick vector of T.orientalis Ikeda in other regions of the world. Thus, the objective of this study was to determine whether invasive H.longicornis ticks in the U.S. are competent vectors of T.orientalis Ikeda. Methods Nymphal H.longicornis ticks were fed on a splenectomized calf infected with the VA-U.S.-T.orientalis Ikeda strain. After molting, a subset of adult ticks from this cohort were dissected, and salivary glands assayed for T.orientalis Ikeda via qPCR. The remaining adult ticks from the group were allowed to feed on three calves. Calves were subsequently monitored for T.orientalis Ikeda infection via blood smear cytology and PCR. Results After acquisition feeding on a VA-U.S.-T.orientalis Ikeda-infected calf as nymphs, a subset of molted adult tick salivary glands tested positive by qPCR for T.orientalis Ikeda. Adult ticks from the same cohort successfully transmitted T.orientalis Ikeda to 3/3 naïve calves, each of which developed parasitemia reaching 0.4–0.9%. Conclusions Our findings demonstrate that U.S. H.longicornis ticks are competent vectors of the VA-U.S.-T.orientalis Ikeda strain. This data provides important information for the U.S. cattle industry regarding the potential spread of this parasite and the necessity of enhanced surveillance and control measures.![]() Supplementary Information The online version contains supplementary material available at 10.1186/s13071-021-04659-9.
Collapse
Affiliation(s)
- Kelcey D Dinkel
- Department of Veterinary Microbiology & Pathology, Washington State University, Pullman, WA, USA
| | - David R Herndon
- United States, Department of Agriculture, Agricultural Research Service, Animal Disease Research Unit, Pullman, WA, USA
| | - Susan M Noh
- Department of Veterinary Microbiology & Pathology, Washington State University, Pullman, WA, USA.,United States, Department of Agriculture, Agricultural Research Service, Animal Disease Research Unit, Pullman, WA, USA
| | - Kevin K Lahmers
- Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Blacksburg, VA, USA
| | - S Michelle Todd
- Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Blacksburg, VA, USA
| | - Massaro W Ueti
- Department of Veterinary Microbiology & Pathology, Washington State University, Pullman, WA, USA.,United States, Department of Agriculture, Agricultural Research Service, Animal Disease Research Unit, Pullman, WA, USA
| | - Glen A Scoles
- United States, Department of Agriculture, Agricultural Research Service, Animal Disease Research Unit, Pullman, WA, USA.,United States Department of Agriculture, Agricultural Research Service, Invasive Insect Biocontrol and Behavior Laboratory, Beltsville, MD, USA
| | - Kathleen L Mason
- United States, Department of Agriculture, Agricultural Research Service, Animal Disease Research Unit, Pullman, WA, USA
| | - Lindsay M Fry
- Department of Veterinary Microbiology & Pathology, Washington State University, Pullman, WA, USA. .,United States, Department of Agriculture, Agricultural Research Service, Animal Disease Research Unit, Pullman, WA, USA.
| |
Collapse
|
9
|
Changing the Recipe: Pathogen Directed Changes in Tick Saliva Components. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2021; 18:ijerph18041806. [PMID: 33673273 PMCID: PMC7918122 DOI: 10.3390/ijerph18041806] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/15/2021] [Revised: 02/07/2021] [Accepted: 02/08/2021] [Indexed: 12/27/2022]
Abstract
Ticks are obligate hematophagous parasites and are important vectors of a wide variety of pathogens. These pathogens include spirochetes in the genus Borrelia that cause Lyme disease, rickettsial pathogens, and tick-borne encephalitis virus, among others. Due to their prolonged feeding period of up to two weeks, hard ticks must counteract vertebrate host defense reactions in order to survive and reproduce. To overcome host defense mechanisms, ticks have evolved a large number of pharmacologically active molecules that are secreted in their saliva, which inhibits or modulates host immune defenses and wound healing responses upon injection into the bite site. These bioactive molecules in tick saliva can create a privileged environment in the host’s skin that tick-borne pathogens take advantage of. In fact, evidence is accumulating that tick-transmitted pathogens manipulate tick saliva composition to enhance their own survival, transmission, and evasion of host defenses. We review what is known about specific and functionally characterized tick saliva molecules in the context of tick infection with the genus Borrelia, the intracellular pathogen Anaplasma phagocytophilum, and tick-borne encephalitis virus. Additionally, we review studies analyzing sialome-level responses to pathogen challenge.
Collapse
|
10
|
Boulanger N, Wikel S. Induced Transient Immune Tolerance in Ticks and Vertebrate Host: A Keystone of Tick-Borne Diseases? Front Immunol 2021; 12:625993. [PMID: 33643313 PMCID: PMC7907174 DOI: 10.3389/fimmu.2021.625993] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2020] [Accepted: 01/22/2021] [Indexed: 12/23/2022] Open
Abstract
Ticks and tick transmitted infectious agents are increasing global public health threats due to increasing abundance, expanding geographic ranges of vectors and pathogens, and emerging tick-borne infectious agents. Greater understanding of tick, host, and pathogen interactions will contribute to development of novel tick control and disease prevention strategies. Tick-borne pathogens adapt in multiple ways to very different tick and vertebrate host environments and defenses. Ticks effectively pharmacomodulate by its saliva host innate and adaptive immune defenses. In this review, we examine the idea that successful synergy between tick and tick-borne pathogen results in host immune tolerance that facilitates successful tick infection and feeding, creates a favorable site for pathogen introduction, modulates cutaneous and systemic immune defenses to establish infection, and contributes to successful long-term infection. Tick, host, and pathogen elements examined here include interaction of tick innate immunity and microbiome with tick-borne pathogens; tick modulation of host cutaneous defenses prior to pathogen transmission; how tick and pathogen target vertebrate host defenses that lead to different modes of interaction and host infection status (reservoir, incompetent, resistant, clinically ill); tick saliva bioactive molecules as important factors in determining those pathogens for which the tick is a competent vector; and, the need for translational studies to advance this field of study. Gaps in our understanding of these relationships are identified, that if successfully addressed, can advance the development of strategies to successfully disrupt both tick feeding and pathogen transmission.
Collapse
Affiliation(s)
- Nathalie Boulanger
- Fédération de Médecine Translationnelle - UR7290, Early Bacterial Virulence, Group Borrelia, Université de Strasbourg, Strasbourg, France.,Centre National de Référence Borrelia, Centre Hospitalier Universitaire, Strasbourg, France
| | - Stephen Wikel
- Department of Medical Sciences, Frank H. Netter, M.D., School of Medicine, Quinnipiac University, Hamden, CT, United States
| |
Collapse
|
11
|
Sajiki Y, Konnai S, Ikenaka Y, Gulay KCM, Kobayashi A, Parizi LF, João BC, Watari K, Fujisawa S, Okagawa T, Maekawa N, Logullo C, da Silva Vaz I, Murata S, Ohashi K. Tick saliva-induced programmed death-1 and PD-ligand 1 and its related host immunosuppression. Sci Rep 2021; 11:1063. [PMID: 33441793 PMCID: PMC7806669 DOI: 10.1038/s41598-020-80251-y] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2020] [Accepted: 12/18/2020] [Indexed: 11/22/2022] Open
Abstract
The tick Rhipicephalus microplus is a harmful parasite of cattle that causes considerable economic losses to the cattle breeding industry. Although R. microplus saliva (Rm-saliva) contains several immunosuppressants, any association between Rm-saliva and the expression of immunoinhibitory molecules, such as programmed death (PD)-1 and PD-ligand 1 (PD-L1), has not been described. In this study, flow cytometric analyses revealed that Rm-saliva upregulated PD-1 expression in T cells and PD-L1 expression in CD14+ and CD11c+ cells in cattle. Additionally, Rm-saliva decreased CD69 expression in T cells and Th1 cytokine production from peripheral blood mononuclear cells. Furthermore, PD-L1 blockade increased IFN-γ production in the presence of Rm-saliva, suggesting that Rm-saliva suppresses Th1 responses via the PD-1/PD-L1 pathway. To reveal the upregulation mechanism of PD-1/PD-L1 by Rm-saliva, we analyzed the function of prostaglandin E2 (PGE2), which is known as an inducer of PD-L1 expression, in Rm-saliva. We found that Rm-saliva contained a high concentration of PGE2, and PGE2 treatment induced PD-L1 expression in CD14+ cells in vitro. Immunohistochemical analyses revealed that PGE2 and PD-L1 expression was upregulated in tick-attached skin in cattle. These data suggest that PGE2 in Rm-saliva has the potential to induce the expression of immunoinhibitory molecules in host immune cells.
Collapse
Affiliation(s)
- Yamato Sajiki
- Department of Disease Control, Faculty of Veterinary Medicine, Hokkaido University, Kita 18, Nishi 9, Kita-ku, Sapporo, 060-0818, Japan
| | - Satoru Konnai
- Department of Disease Control, Faculty of Veterinary Medicine, Hokkaido University, Kita 18, Nishi 9, Kita-ku, Sapporo, 060-0818, Japan. .,Department of Advanced Pharmaceutics, Faculty of Veterinary Medicine, Hokkaido University, Sapporo, 060-0818, Japan.
| | - Yoshinori Ikenaka
- Department of Environmental Veterinary Sciences, Faculty of Veterinary Medicine, Hokkaido University, Sapporo, 060-0818, Japan
| | | | - Atsushi Kobayashi
- Department of Veterinary Clinical Medicine, Faculty of Veterinary Medicine, Hokkaido University, Sapporo, 060-0818, Japan
| | - Luís Fernando Parizi
- Centro de Biotecnologia, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, 91501-970, Brazil
| | - Benvindo Capela João
- Centro de Biotecnologia, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, 91501-970, Brazil
| | - Kei Watari
- Department of Disease Control, Faculty of Veterinary Medicine, Hokkaido University, Kita 18, Nishi 9, Kita-ku, Sapporo, 060-0818, Japan
| | - Sotaro Fujisawa
- Department of Disease Control, Faculty of Veterinary Medicine, Hokkaido University, Kita 18, Nishi 9, Kita-ku, Sapporo, 060-0818, Japan
| | - Tomohiro Okagawa
- Department of Advanced Pharmaceutics, Faculty of Veterinary Medicine, Hokkaido University, Sapporo, 060-0818, Japan
| | - Naoya Maekawa
- Department of Advanced Pharmaceutics, Faculty of Veterinary Medicine, Hokkaido University, Sapporo, 060-0818, Japan
| | - Carlos Logullo
- Laboratório Integrado de Bioquímica Hatisaburo Masuda and Laboratório Integrado de Morfologia, NUPEM-UFRJ, Macaé, RJ, Brazil
| | - Itabajara da Silva Vaz
- Centro de Biotecnologia, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, 91501-970, Brazil
| | - Shiro Murata
- Department of Disease Control, Faculty of Veterinary Medicine, Hokkaido University, Kita 18, Nishi 9, Kita-ku, Sapporo, 060-0818, Japan.,Department of Advanced Pharmaceutics, Faculty of Veterinary Medicine, Hokkaido University, Sapporo, 060-0818, Japan
| | - Kazuhiko Ohashi
- Department of Disease Control, Faculty of Veterinary Medicine, Hokkaido University, Kita 18, Nishi 9, Kita-ku, Sapporo, 060-0818, Japan.,Department of Advanced Pharmaceutics, Faculty of Veterinary Medicine, Hokkaido University, Sapporo, 060-0818, Japan
| |
Collapse
|
12
|
Narasimhan S, Kurokawa C, DeBlasio M, Matias J, Sajid A, Pal U, Lynn G, Fikrig E. Acquired tick resistance: The trail is hot. Parasite Immunol 2020; 43:e12808. [PMID: 33187012 DOI: 10.1111/pim.12808] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2020] [Accepted: 11/09/2020] [Indexed: 12/17/2022]
Abstract
Acquired tick resistance is a phenomenon wherein the host elicits an immune response against tick salivary components upon repeated tick infestations. The immune responses, potentially directed against critical salivary components, thwart tick feeding, and the animal becomes resistant to subsequent tick infestations. The development of tick resistance is frequently observed when ticks feed on non-natural hosts, but not on natural hosts. The molecular mechanisms that lead to the development of tick resistance are not fully understood, and both host and tick factors are invoked in this phenomenon. Advances in molecular tools to address the host and the tick are beginning to reveal new insights into this phenomenon and to uncover a deeper understanding of the fundamental biology of tick-host interactions. This review will focus on the expanding understanding of acquired tick resistance and highlight the impact of this understanding on anti-tick vaccine development efforts.
Collapse
Affiliation(s)
- Sukanya Narasimhan
- Section of Infectious Diseases, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT, USA
| | - Cheyne Kurokawa
- Section of Infectious Diseases, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT, USA
| | - Melody DeBlasio
- Section of Infectious Diseases, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT, USA
| | - Jaqueline Matias
- Section of Infectious Diseases, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT, USA
| | - Andaleeb Sajid
- Section of Infectious Diseases, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT, USA
| | - Utpal Pal
- Department of Veterinary Medicine, University of Maryland, College Park, MD, USA
| | - Geoffrey Lynn
- Section of Infectious Diseases, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT, USA
| | - Erol Fikrig
- Section of Infectious Diseases, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT, USA
| |
Collapse
|
13
|
Dendritic Cells as a Disputed Fortress on the Tick-Host Battlefield. Trends Parasitol 2020; 37:340-354. [PMID: 33303363 DOI: 10.1016/j.pt.2020.11.004] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2020] [Revised: 10/23/2020] [Accepted: 11/09/2020] [Indexed: 12/11/2022]
Abstract
From seminal publications in the early 1970s, the world learned that dendritic cells (DCs) are powerful and versatile antigen-presenting cells. It took a few years until the first studies expanded our understanding of the pivotal role of these immune 'soldiers' against ticks. Advances in biochemistry, molecular biology, and bioinformatics have shed light on the identification of key salivary molecules that modulate the biology of DCs in favor of tick parasitism. Here, we present a critical overview of the discoveries accumulated on the tick-host battlefield from a DC perspective. Moreover, the clinical significance of DC-targeted tick salivary components is discussed, not only as facilitators of the transmission of tick-borne pathogens or vaccine candidates, but also as potential immunobiologics to treat immune-mediated diseases.
Collapse
|
14
|
Aounallah H, Bensaoud C, M'ghirbi Y, Faria F, Chmelar JI, Kotsyfakis M. Tick Salivary Compounds for Targeted Immunomodulatory Therapy. Front Immunol 2020; 11:583845. [PMID: 33072132 PMCID: PMC7538779 DOI: 10.3389/fimmu.2020.583845] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Accepted: 09/02/2020] [Indexed: 12/11/2022] Open
Abstract
Immunodeficiency disorders and autoimmune diseases are common, but a lack of effective targeted drugs and the side-effects of existing drugs have stimulated interest in finding therapeutic alternatives. Naturally derived substances are a recognized source of novel drugs, and tick saliva is increasingly recognized as a rich source of bioactive molecules with specific functions. Ticks use their saliva to overcome the innate and adaptive host immune systems. Their saliva is a rich cocktail of molecules including proteins, peptides, lipid derivatives, and recently discovered non-coding RNAs that inhibit or modulate vertebrate immune reactions. A number of tick saliva and/or salivary gland molecules have been characterized and shown to be promising candidates for drug development for vertebrate immune diseases. However, further validation of these molecules at the molecular, cellular, and organism levels is now required to progress lead candidates to clinical testing. In this paper, we review the data on the immuno-pharmacological aspects of tick salivary compounds characterized in vitro and/or in vivo and present recent findings on non-coding RNAs that might be exploitable as immunomodulatory therapies.
Collapse
Affiliation(s)
- Hajer Aounallah
- Institut Pasteur de Tunis, LR19IPTX, Service d'Entomologie Médicale, Université de Tunis El Manar, Tunis, Tunisia.,Innovation and Development Laboratory, Innovation and Development Center, Instituto Butantan, São Paulo, Brazil
| | - Chaima Bensaoud
- Institute of Parasitology, Biology Centre, Czech Academy of Sciences, České Budějovice, Czechia
| | - Youmna M'ghirbi
- Institut Pasteur de Tunis, LR19IPTX, Service d'Entomologie Médicale, Université de Tunis El Manar, Tunis, Tunisia
| | - Fernanda Faria
- Innovation and Development Laboratory, Innovation and Development Center, Instituto Butantan, São Paulo, Brazil
| | - Jindr Ich Chmelar
- Department of Medical Biology, Faculty of Science, University of South Bohemia in České Budějovice, České Budějovice, Czechia
| | - Michail Kotsyfakis
- Institute of Parasitology, Biology Centre, Czech Academy of Sciences, České Budějovice, Czechia.,Department of Medical Biology, Faculty of Science, University of South Bohemia in České Budějovice, České Budějovice, Czechia
| |
Collapse
|
15
|
Fernando DD, Fischer K. Proteases and pseudoproteases in parasitic arthropods of clinical importance. FEBS J 2020; 287:4284-4299. [PMID: 32893448 DOI: 10.1111/febs.15546] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2020] [Revised: 08/18/2020] [Accepted: 08/28/2020] [Indexed: 12/19/2022]
Abstract
Parasitic arthropods feed on blood or skin tissue and share comparable repertoires of proteases involved in haematophagy, digestion, egg development and immunity. While proteolytically active proteases of multiple classes dominate, an increasing number of pseudoproteases have been discovered that have no proteolytic function but are pharmacologically active biomolecules, evolved to carry out alternative functions as regulatory, antihaemostatic, anti-inflammatory or immunomodulatory compounds. In this review, we provide an overview of proteases and pseudoproteases from clinically important arthropod parasites. Many of these act in central biological pathways of parasite survival and host-parasite interaction and may be potential targets for therapeutic interventions.
Collapse
Affiliation(s)
- Deepani Darshika Fernando
- Cell and Molecular Biology Department, Infectious Diseases Program, QIMR Berghofer Medical Research Institute, Brisbane, Qld, Australia
| | - Katja Fischer
- Cell and Molecular Biology Department, Infectious Diseases Program, QIMR Berghofer Medical Research Institute, Brisbane, Qld, Australia
| |
Collapse
|
16
|
Ayala EV, Rodrigues da Cunha G, Azevedo MA, Calderon M, Jimenez J, Venuto AP, Gazzinelli R, Lavalle RJY, Riva AGV, Hincapie R, Finn MG, Marques AF. C57BL/6 α-1,3-Galactosyltransferase Knockout Mouse as an Animal Model for Experimental Chagas Disease. ACS Infect Dis 2020; 6:1807-1815. [PMID: 32374586 DOI: 10.1021/acsinfecdis.0c00061] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The leading animal model of experimental Chagas disease, the mouse, plays a significant role in studies for vaccine development, diagnosis, and human therapies. Humans, along with Old World primates, alone among mammals, cannot make the terminal carbohydrate linkage of the α-Gal trisaccharide. It has been established that the anti-α-Gal immune response is likely to be a critical factor for protection against Trypanosoma cruzi (T. cruzi) infection in humans. However, the mice customarily employed for the study of T. cruzi infection naturally express the α-Gal epitope and therefore do not produce anti-α-Gal antibodies. Here, we used the C57BL/6 α-1,3-galactosyltransferase knockout (α-GalT-KO) mouse, which does not express the α-Gal epitope as a model for experimental Chagas disease. We found the anti-α-Gal IgG antibody response to an increase in α-GalT-KO mice infected with Arequipa and Colombiana strains of T. cruzi, leading to fewer parasite nests, lower parasitemia, and an increase of INF-γ, TNF-α, and IL-12 cytokines in the heart of α-GalT-KO mice compared with α-GalT-WT mice on days 60 and 120 postinfection. We therefore agree that the C57BL/6 α-GalT-KO mouse represents a useful model for initial testing of therapeutic and immunological approaches against different strains of T. cruzi.
Collapse
Affiliation(s)
- Edward Valencia Ayala
- Laboratório de Imunologia e Genômica de Parasitos, Departamento de Parasitologia, Instituto de Ciências Biológicas/ICB, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais 31270901, Brazil
- Instituto de Investigación, Centro de Investigación en Inmunología e Infectología, Facultad de Medicina Humana, Universidad de San Martin de Porres, Lima 15000, Perú
| | - Gisele Rodrigues da Cunha
- Laboratório de Imuno-Proteômica e Biologia de Parasitos, Departamento de Parasitologia, Instituto de Ciências Biológicas/ICB, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais 31270901, Brazil
| | - Maira Araujo Azevedo
- Laboratório de Imuno-Proteômica e Biologia de Parasitos, Departamento de Parasitologia, Instituto de Ciências Biológicas/ICB, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais 31270901, Brazil
| | - Maritza Calderon
- Laboratorio de Investigación en Enfermedades Infecciosas and Laboratorio de Biología Molecular, Facultad de Ciencias y Filosofía, Universidad Peruana Cayetano Heredia, Lima 15000, Perú
| | - Juan Jimenez
- Laboratorio de Parasitología en Fauna Silvestre y Zoonosis, Facultad de Ciencias Biológicas, Universidad Nacional Mayor de San Marcos, Lima 15000, Perú
| | - Ana Paula Venuto
- Laboratório de Imuno-Proteômica e Biologia de Parasitos, Departamento de Parasitologia, Instituto de Ciências Biológicas/ICB, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais 31270901, Brazil
| | - Ricardo Gazzinelli
- Instituto de Pesquisa Rene Rachou, Fundacao Oswaldo Cruz, Belo Horizonte, Minas Gerais 30190-009, Brazil
- Plataforma de Medicina Translacional, Fundacao Oswaldo Cruz, Belo Horizonte, Minas Gerais 30190-009, Brazil
| | - Raúl Jesus Ynocente Lavalle
- Laboratorio de Parasitología en Fauna Silvestre y Zoonosis, Facultad de Ciencias Biológicas, Universidad Nacional Mayor de San Marcos, Lima 15000, Perú
| | - Angela Giovana Vidal Riva
- Instituto de Investigación, Centro de Investigación en Inmunología e Infectología, Facultad de Medicina Humana, Universidad de San Martin de Porres, Lima 15000, Perú
- Laboratorio de Investigación en Enfermedades Infecciosas and Laboratorio de Biología Molecular, Facultad de Ciencias y Filosofía, Universidad Peruana Cayetano Heredia, Lima 15000, Perú
| | - Robert Hincapie
- School of Chemistry and Biochemistry, School of Biological Sciences, Georgia Institute of Technology, Atlanta, Georgia 30332 United States
| | - M. G. Finn
- School of Chemistry and Biochemistry, School of Biological Sciences, Georgia Institute of Technology, Atlanta, Georgia 30332 United States
| | - Alexandre F. Marques
- Laboratório de Imuno-Proteômica e Biologia de Parasitos, Departamento de Parasitologia, Instituto de Ciências Biológicas/ICB, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais 31270901, Brazil
| |
Collapse
|
17
|
Hart CE, Ribeiro JM, Kazimirova M, Thangamani S. Tick-Borne Encephalitis Virus Infection Alters the Sialome of Ixodes ricinus Ticks During the Earliest Stages of Feeding. Front Cell Infect Microbiol 2020; 10:41. [PMID: 32133301 PMCID: PMC7041427 DOI: 10.3389/fcimb.2020.00041] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2019] [Accepted: 01/21/2020] [Indexed: 01/14/2023] Open
Abstract
Ticks are hematophagous arthropods that transmit a number of pathogens while feeding. Among these is tick-borne encephalitis virus (TBEV), a flavivirus transmitted by Ixodes ricinus ticks in the temperate zone of Europe. The infection results in febrile illness progressing to encephalitis and meningitis with a possibility of fatality or long-term neurological sequelae. The composition of tick saliva plays an essential role in the initial virus transmission during tick feeding. Ticks secrete a diverse range of salivary proteins to modulate the host response, such as lipocalins to control the itch and inflammatory response, and both proteases and protease inhibitors to prevent blood coagulation. Here, the effect of viral infection of adult females of Ixodes ricinus was studied with the goal of determining how the virus alters the tick sialome to modulate host tissue response at the site of infection. Uninfected ticks or those infected with TBEV were fed on mice and removed and dissected one- and 3-h post-attachment. RNA from the salivary glands of these ticks, as well as from unfed ticks, was extracted and subjected to next-generation sequencing to determine the expression of key secreted proteins at each timepoint. Genes showing statistically significant up- or down-regulation between infected and control ticks were selected and compared to published literature to ascertain their function. From this, the effect of tick viral infection on the modulation of the tick-host interface was determined. Infected ticks were found to differentially express a number of uncategorized genes, proteases, Kunitz-type serine protease inhibitors, cytotoxins, and lipocalins at different timepoints. These virus-induced changes to the tick sialome may play a significant role in facilitating virus transmission during the early stages of tick feeding.
Collapse
Affiliation(s)
- Charles E. Hart
- SUNY Center for Environmental Health and Medicine, SUNY Upstate Medical University, Syracuse, NY, United States
- Institute for Global Health and Translational Science, SUNY Upstate Medical University, Syracuse, NY, United States
- The Institute for Translational Science, University of Texas Medical Branch, Galveston, TX, United States
| | - Jose M. Ribeiro
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, Bethesda, MD, United States
| | - Maria Kazimirova
- Institute of Zoology, Slovak Academy of Sciences, Bratislava, Slovakia
| | - Saravanan Thangamani
- SUNY Center for Environmental Health and Medicine, SUNY Upstate Medical University, Syracuse, NY, United States
- Institute for Global Health and Translational Science, SUNY Upstate Medical University, Syracuse, NY, United States
- Department of Microbiology and Immunology, SUNY Upstate Medical University, Syracuse, NY, United States
| |
Collapse
|
18
|
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
|
19
|
Xu Z, Lin Z, Wei N, Di Q, Cao J, Zhou Y, Gong H, Zhang H, Zhou J. Immunomodulatory effects of Rhipicephalus haemaphysaloides serpin RHS2 on host immune responses. Parasit Vectors 2019; 12:341. [PMID: 31296257 PMCID: PMC6624921 DOI: 10.1186/s13071-019-3607-4] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2018] [Accepted: 07/08/2019] [Indexed: 02/06/2023] Open
Abstract
Background Rhipicephalus haemaphysaloides is a widespread tick species in China and other South East Asian countries, where it is the vector of many pathogens. The objective of this study was to study the role of serpin (serine protease inhibitor) during the tick-host interaction. Methods The differentiation of bone marrow-derived dendritic cells (BMDC) was induced in vitro, and the effect of RHS2 on the maturation of DCs was evaluated. The effects of RHS2 on T cell activation and cytotoxic T lymphocytes’ (CTLs) activity were analyzed by flow cytometry. Antibody subtypes after immunization of mice with RHS2 and OVA were determined. Results RHS2 can inhibit the differentiation of bone marrow-derived cells into DCs and promote their differentiation into macrophages. RHS2 can inhibit the maturation of DCs and the expression of CD80, CD86 and MHCII. The number of CD3+CD4+ and CD3+CD8+ T cells secreting IFN-γ, IL-2 and TNF-α was decreased, and the number of CD3+CD4+ T cells secreting IL-4 was increased, indicating that RHS2 can inhibit the activation of CD4 T cells and CD8 T cells, leading to inhibition of Th1 immune response. RHS2 inhibits the elimination of target cells by cytotoxic T lymphocytes. After immunization of mice with RHS2 and OVA, serum IgG2b was significantly reduced and IgM was increased. Conclusions The results show that RHS2 has an inhibitory effect on the host immune response. Ticks have evolved various ways to circumvent adaptive immunity. Their serpin inhibits BMDC differentiation to reduce immune responses.
Collapse
Affiliation(s)
- Zhengmao Xu
- Key Laboratory of Animal Parasitology of Ministry of Agriculture, Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai, China
| | - Zhibing Lin
- Key Laboratory of Animal Parasitology of Ministry of Agriculture, Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai, China
| | - Nana Wei
- Key Laboratory of Animal Parasitology of Ministry of Agriculture, Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai, China
| | - Qing Di
- Key Laboratory of Animal Parasitology of Ministry of Agriculture, Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai, China
| | - Jie Cao
- Key Laboratory of Animal Parasitology of Ministry of Agriculture, Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai, China
| | - Yongzhi Zhou
- Key Laboratory of Animal Parasitology of Ministry of Agriculture, Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai, China
| | - Haiyan Gong
- Key Laboratory of Animal Parasitology of Ministry of Agriculture, Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai, China
| | - Houshuang Zhang
- Key Laboratory of Animal Parasitology of Ministry of Agriculture, Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai, China
| | - Jinlin Zhou
- Key Laboratory of Animal Parasitology of Ministry of Agriculture, Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai, China.
| |
Collapse
|
20
|
Štibrániová I, Bartíková P, Holíková V, Kazimírová M. Deciphering Biological Processes at the Tick-Host Interface Opens New Strategies for Treatment of Human Diseases. Front Physiol 2019; 10:830. [PMID: 31333488 PMCID: PMC6617849 DOI: 10.3389/fphys.2019.00830] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2019] [Accepted: 06/17/2019] [Indexed: 12/14/2022] Open
Abstract
Ticks are obligatory blood-feeding ectoparasites, causing blood loss and skin damage in their hosts. In addition, ticks also transmit a number of various pathogenic microorganisms that cause serious diseases in humans and animals. Ticks evolved a wide array of salivary bioactive compounds that, upon injection into the host skin, inhibit or modulate host reactions such as hemostasis, inflammation and wound healing. Modulation of the tick attachment site in the host skin involves mainly molecules which affect physiological processes orchestrated by cytokines, chemokines and growth factors. Suppressing host defense reactions is crucial for tick survival and reproduction. Furthermore, pharmacologically active compounds in tick saliva have a promising therapeutic potential for treatment of some human diseases connected with disorders in hemostasis and immune system. These disorders are often associated to alterations in signaling pathways and dysregulation or overexpression of specific cytokines which, in turn, affect mechanisms of angiogenesis, cell motility and cytoskeletal regulation. Moreover, tick salivary molecules were found to exert cytotoxic and cytolytic effects on various tumor cells and have anti-angiogenic properties. Elucidation of the mode of action of tick bioactive molecules on the regulation of cell processes in their mammalian hosts could provide new tools for understanding the complex changes leading to immune disorders and cancer. Tick bioactive molecules may also be exploited as new pharmacological inhibitors of the signaling pathways of cytokines and thus help alleviate patient discomfort and increase patient survival. We review the current knowledge about tick salivary peptides and proteins that have been identified and functionally characterized in in vitro and/or in vivo models and their therapeutic perspective.
Collapse
Affiliation(s)
- Iveta Štibrániová
- Biomedical Research Center, Institute of Virology, Slovak Academy of Sciences, Bratislava, Slovakia
| | - Pavlína Bartíková
- Biomedical Research Center, Institute of Virology, Slovak Academy of Sciences, Bratislava, Slovakia
| | - Viera Holíková
- Biomedical Research Center, Institute of Virology, Slovak Academy of Sciences, Bratislava, Slovakia
| | - Mária Kazimírová
- Institute of Zoology, Slovak Academy of Sciences, Bratislava, Slovakia
| |
Collapse
|
21
|
Chmelař J, Kotál J, Kovaříková A, Kotsyfakis M. The Use of Tick Salivary Proteins as Novel Therapeutics. Front Physiol 2019; 10:812. [PMID: 31297067 PMCID: PMC6607933 DOI: 10.3389/fphys.2019.00812] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2019] [Accepted: 06/11/2019] [Indexed: 12/12/2022] Open
Abstract
The last three decades of research into tick salivary components have revealed several proteins with important pharmacological and immunological activities. Two primary interests have driven research into tick salivary secretions: the search for suitable pathogen transmission blocking or “anti-tick” vaccine candidates and the search for novel therapeutics derived from tick salivary components. Intensive basic research in the field of tick salivary gland transcriptomics and proteomics has identified several major protein families that play important roles in tick feeding and overcoming vertebrate anti-tick responses. Moreover, these families contain members with unrealized therapeutic potential. Here we review the major tick salivary protein families exploitable in medical applications such as immunomodulation, inhibition of hemostasis and inflammation. Moreover, we discuss the potential, opportunities, and challenges in searching for novel tick-derived drugs.
Collapse
Affiliation(s)
- Jindřich Chmelař
- Department of Medical Biology, Faculty of Science, University of South Bohemia in České Budějovice, České Budějovice, Czechia
| | - Jan Kotál
- Department of Medical Biology, Faculty of Science, University of South Bohemia in České Budějovice, České Budějovice, Czechia.,Laboratory of Genomics and Proteomics of Disease Vectors, Biology Centre CAS, Institute of Parasitology, České Budějovice, Czechia
| | - Anna Kovaříková
- Department of Medical Biology, Faculty of Science, University of South Bohemia in České Budějovice, České Budějovice, Czechia
| | - Michail Kotsyfakis
- Department of Medical Biology, Faculty of Science, University of South Bohemia in České Budějovice, České Budějovice, Czechia.,Laboratory of Genomics and Proteomics of Disease Vectors, Biology Centre CAS, Institute of Parasitology, České Budějovice, Czechia
| |
Collapse
|
22
|
Mans BJ. Chemical Equilibrium at the Tick-Host Feeding Interface:A Critical Examination of Biological Relevance in Hematophagous Behavior. Front Physiol 2019; 10:530. [PMID: 31118903 PMCID: PMC6504839 DOI: 10.3389/fphys.2019.00530] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2019] [Accepted: 04/15/2019] [Indexed: 12/14/2022] Open
Abstract
Ticks secrete hundreds to thousands of proteins into the feeding site, that presumably all play important functions in the modulation of host defense mechanisms. The current review considers the assumption that tick proteins have functional relevance during feeding. The feeding site may be described as a closed system and could be treated as an ideal equilibrium system, thereby allowing modeling of tick-host interactions in an equilibrium state. In this equilibrium state, the concentration of host and tick proteins and their affinities will determine functional relevance at the tick-host interface. Using this approach, many characterized tick proteins may have functional relevant concentrations and affinities at the feeding site. Conversely, the feeding site is not an ideal closed system, but is dynamic and changing, leading to possible overestimation of tick protein concentration at the feeding site and consequently an overestimation of functional relevance. Ticks have evolved different possible strategies to deal with this dynamic environment and overcome the barrier that equilibrium kinetics poses to tick feeding. Even so, cognisance of the limitations that equilibrium binding place on deductions of functional relevance should serve as an important incentive to determine both the concentration and affinity of tick proteins proposed to be functional at the feeding site.
Collapse
Affiliation(s)
- Ben J. Mans
- Epidemiology, Parasites and Vectors, Agricultural Research Council-Onderstepoort Veterinary Research, Pretoria, South Africa
- Department of Veterinary Tropical Diseases, University of Pretoria, Pretoria, South Africa
- Department of Life and Consumer Sciences, University of South Africa, Pretoria, South Africa
| |
Collapse
|
23
|
de la Fuente J, Pacheco I, Villar M, Cabezas-Cruz A. The alpha-Gal syndrome: new insights into the tick-host conflict and cooperation. Parasit Vectors 2019; 12:154. [PMID: 30944017 PMCID: PMC6448316 DOI: 10.1186/s13071-019-3413-z] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2019] [Accepted: 03/27/2019] [Indexed: 02/04/2023] Open
Abstract
This primer focuses on a recently diagnosed tick-borne allergic disease known as the alpha-Gal syndrome (AGS). Tick bites induce in humans high levels of IgE antibodies against the carbohydrate Galα1-3Galβ1-(3)4GlcNAc-R (α-Gal) present on tick salivary glycoproteins and tissues of non-catarrhine mammals, leading to the AGS in some individuals. This immune response evolved as a conflict and cooperation between ticks and human hosts including their gut microbiota. The conflict is characterized by the AGS that mediate delayed anaphylaxis to red meat consumption and certain drugs such as cetuximab, and immediate anaphylaxis to tick bites. The cooperation is supported by the capacity of anti-α-Gal IgM and IgG antibody response to protect against pathogens with α-Gal on their surface. Despite the growing diagnosis of AGS in all world continents, many questions remain to be elucidated on the tick proteins and immune mechanisms triggering this syndrome, and the protective response against pathogen infection elicited by anti-α-Gal antibodies. The answer to these questions will provide information for the evaluation of risks, diagnosis and prevention of the AGS, and the possibility of using the carbohydrate α-Gal to develop vaccines for the control of major infectious diseases.
Collapse
Affiliation(s)
- José de la Fuente
- SaBio, Instituto de Investigación en Recursos Cinegéticos (IREC), Consejo Superior de Investigaciones Científicas (CSIC), Universidad de Castilla-La Mancha (UCLM)-Junta de Comunidades de Castilla-La Mancha (JCCM), Ronda de Toledo s/n, 13005 Ciudad Real, Spain
- Department of Veterinary Pathobiology, Center for Veterinary Health Sciences, Oklahoma State University, Stillwater, OK 74078 USA
| | - Iván Pacheco
- SaBio, Instituto de Investigación en Recursos Cinegéticos (IREC), Consejo Superior de Investigaciones Científicas (CSIC), Universidad de Castilla-La Mancha (UCLM)-Junta de Comunidades de Castilla-La Mancha (JCCM), Ronda de Toledo s/n, 13005 Ciudad Real, Spain
| | - Margarita Villar
- SaBio, Instituto de Investigación en Recursos Cinegéticos (IREC), Consejo Superior de Investigaciones Científicas (CSIC), Universidad de Castilla-La Mancha (UCLM)-Junta de Comunidades de Castilla-La Mancha (JCCM), Ronda de Toledo s/n, 13005 Ciudad Real, Spain
| | - Alejandro Cabezas-Cruz
- UMR BIPAR, INRA, ANSES, Ecole Nationale Vétérinaire d’Alfort, Université Paris-Est, Maisons-Alfort, 94700 France
| |
Collapse
|
24
|
Esteves E, Bizzarro B, Costa FB, Ramírez-Hernández A, Peti APF, Cataneo AHD, Wowk PF, Timóteo RP, Labruna MB, Silva Junior PI, Silva CL, Faccioli LH, Fogaça AC, Sorgi CA, Sá-Nunes A. Amblyomma sculptum Salivary PGE 2 Modulates the Dendritic Cell- Rickettsia rickettsii Interactions in vitro and in vivo. Front Immunol 2019; 10:118. [PMID: 30778355 PMCID: PMC6369204 DOI: 10.3389/fimmu.2019.00118] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2018] [Accepted: 01/15/2019] [Indexed: 01/06/2023] Open
Abstract
Amblyomma sculptum is an important vector of Rickettsia rickettsii, causative agent of Rocky Mountain spotted fever and the most lethal tick-borne pathogen affecting humans. To feed on the vertebrate host's blood, A. sculptum secretes a salivary mixture, which may interact with skin resident dendritic cells (DCs) and modulate their function. The present work was aimed at depicting the A. sculptum saliva-host DC network and the biochemical nature of the immunomodulatory component(s) involved in this interface. A. sculptum saliva inhibits the production of inflammatory cytokines by murine DCs stimulated with LPS. The fractionation of the low molecular weight salivary content by reversed-phase chromatography revealed active fractions eluting from 49 to 55% of the acetonitrile gradient. Previous studies suggested that this pattern of elution matches with that observed for prostaglandin E2 (PGE2) and the molecular identity of this lipid mediator was unambiguously confirmed by a new high-resolution mass spectrometry methodology. A productive infection of murine DCs by R. rickettsii was demonstrated for the first time leading to proinflammatory cytokine production that was inhibited by both A. sculptum saliva and PGE2, a result also achieved with human DCs. The adoptive transfer of murine DCs incubated with R. rickettsii followed by treatment with A. sculptum saliva or PGE2 did not change the cytokine profile associated to cellular recall responses while IgG2a-specific antibodies were decreased in the serum of these mice. Together, these findings emphasize the role of PGE2 as a universal immunomodulator of tick saliva. In addition, it contributes to new approaches to explore R. rickettsii-DC interactions both in vitro and in vivo.
Collapse
Affiliation(s)
- Eliane Esteves
- Department of Immunology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Bruna Bizzarro
- Department of Immunology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Francisco Borges Costa
- Department of Preventive Veterinary Medicine and Animal Health, School of Veterinary Medicine and Animal Science, University of São Paulo, São Paulo, Brazil
| | - Alejandro Ramírez-Hernández
- Department of Preventive Veterinary Medicine and Animal Health, School of Veterinary Medicine and Animal Science, University of São Paulo, São Paulo, Brazil
| | - Ana Paula Ferranti Peti
- Department of Clinical Analysis, Toxicology and Food Science, School of Pharmaceutical Sciences of Ribeirao Preto, University of São Paulo, Ribeirao Preto, Brazil
| | | | - Pryscilla Fanini Wowk
- Laboratory of Molecular Virology, Carlos Chagas Institute, Fundação Oswaldo Cruz, Curitiba, Brazil
| | - Rodolfo Pessato Timóteo
- Institute of Natural and Biological Sciences, Federal University of Triângulo Mineiro, Uberaba, Brazil
| | - Marcelo Bahia Labruna
- Department of Preventive Veterinary Medicine and Animal Health, School of Veterinary Medicine and Animal Science, University of São Paulo, São Paulo, Brazil
| | | | - Célio Lopes Silva
- Department of Biochemistry and Immunology, School of Medicine of Ribeirao Preto, University of São Paulo, Ribeirao Preto, Brazil
| | - Lúcia Helena Faccioli
- Department of Clinical Analysis, Toxicology and Food Science, School of Pharmaceutical Sciences of Ribeirao Preto, University of São Paulo, Ribeirao Preto, Brazil
| | - Andréa Cristina Fogaça
- Department de Parasitology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil.,National Institute of Science and Technology in Molecular Entomology, National Council of Scientific and Technological Development (INCT-EM/CNPq), Rio de Janeiro, Brazil
| | - Carlos Arterio Sorgi
- Department of Clinical Analysis, Toxicology and Food Science, School of Pharmaceutical Sciences of Ribeirao Preto, University of São Paulo, Ribeirao Preto, Brazil
| | - Anderson Sá-Nunes
- Department of Immunology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil.,National Institute of Science and Technology in Molecular Entomology, National Council of Scientific and Technological Development (INCT-EM/CNPq), Rio de Janeiro, Brazil
| |
Collapse
|
25
|
Nuttall PA. Wonders of tick saliva. Ticks Tick Borne Dis 2018; 10:470-481. [PMID: 30459085 DOI: 10.1016/j.ttbdis.2018.11.005] [Citation(s) in RCA: 69] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2018] [Revised: 10/31/2018] [Accepted: 11/09/2018] [Indexed: 12/16/2022]
Abstract
Saliva of ticks is arguably the most complex saliva of any animal. This is particularly the case for ixodid species that feed for many days firmly attached to the same skin site of their obliging host. Sequencing and spectrometry technologies combined with bioinformatics are enumerating ingredients in the saliva cocktail. The dynamic and expanding saliva recipe is helping decipher the wonderous activities of tick saliva, revealing how ticks stealthily hide from their hosts while satisfying their gluttony and sharing their individual resources. This review takes a tick perspective on the composition and functions of tick saliva, covering water balance, gasket and holdfast, control of host responses, dynamics, individuality, mate guarding, saliva-assisted transmission, and redundancy. It highlights areas sometimes overlooked - feeding aggregation and sharing of sialomes, and the contribution of salivary gland storage granules - and questions whether the huge diversity of tick saliva molecules is 'redundant' or more a reflection on the enormous adaptability wonderous saliva confers on ticks.
Collapse
Affiliation(s)
- Patricia A Nuttall
- Department of Zoology, University of Oxford, UK and Centre for Ecology & Hydrology, Wallingford, Oxfordshire, UK.
| |
Collapse
|
26
|
Ticks and Tick-Borne Infections: Complex Ecology, Agents, and Host Interactions. Vet Sci 2018; 5:vetsci5020060. [PMID: 29925800 PMCID: PMC6024845 DOI: 10.3390/vetsci5020060] [Citation(s) in RCA: 90] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2018] [Revised: 06/11/2018] [Accepted: 06/15/2018] [Indexed: 12/21/2022] Open
Abstract
Ticks transmit the most diverse array of infectious agents of any arthropod vector. Both ticks and the microbes they transmit are recognized as significant threats to human and veterinary public health. This article examines the potential impacts of climate change on the distribution of ticks and the infections they transmit; the emergence of novel tick-borne pathogens, increasing geographic range and incidence of tick-borne infections; and advances in the characterization of tick saliva mediated modulation of host defenses and the implications of those interactions for transmission, establishment, and control of tick infestation and tick-borne infectious agents.
Collapse
|
27
|
Santiago PB, de Araújo CN, Charneau S, Bastos IMD, Assumpção TCF, Queiroz RML, Praça YR, Cordeiro TDM, Garcia CHS, da Silva IG, Raiol T, Motta FN, de Araújo Oliveira JV, de Sousa MV, Ribeiro JMC, de Santana JM. Exploring the molecular complexity of Triatoma dimidiata sialome. J Proteomics 2017; 174:47-60. [PMID: 29288089 DOI: 10.1016/j.jprot.2017.12.016] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2017] [Revised: 12/20/2017] [Accepted: 12/22/2017] [Indexed: 01/19/2023]
Abstract
Triatoma dimidiata, a Chagas disease vector widely distributed along Central America, has great capability for domestic adaptation as the majority of specimens caught inside human dwellings or in peridomestic areas fed human blood. Exploring the salivary compounds that overcome host haemostatic and immune responses is of great scientific interest. Here, we provide a deeper insight into its salivary gland molecules. We used high-throughput RNA sequencing to examine in depth the T. dimidiata salivary gland transcriptome. From >51 million reads assembled, 92.21% are related to putative secreted proteins. Lipocalin is the most abundant gene family, confirming it is an expanded family in Triatoma genus salivary repertoire. Other putatively secreted members include phosphatases, odorant binding protein, hemolysin, proteases, protease inhibitors, antigen-5 and antimicrobial peptides. This work expands the previous set of functionally annotated sequences from T. dimidiata salivary glands available in NCBI from 388 to 3815. Additionally, we complemented the salivary analysis through proteomics (available data via ProteomeXchange with identifier PXD008510), disclosing the set complexity of 119 secreted proteins and validating the transcriptomic results. Our large-scale approach enriches the pharmacologically active molecules database and improves our knowledge about the complexity of salivary compounds from haematophagous vectors and their biological interactions. SIGNIFICANCE Several haematophagous triatomine species can transmit Trypanosoma cruzi, the etiological agent of Chagas disease. Due to the reemergence of this disease, new drugs for its prevention and treatment are considered priorities. For this reason, the knowledge of vector saliva emerges as relevant biological finding, contributing to the design of different strategies for vector control and disease transmission. Here we report the transcriptomic and proteomic compositions of the salivary glands (sialome) of the reduviid bug Triatoma dimidiata, a relevant Chagas disease vector in Central America. Our results are robust and disclosed unprecedented insights into the notable diversity of its salivary glands content, revealing relevant anti-haemostatic salivary gene families. Our work expands almost ten times the previous set of functionally annotated sequences from T. dimidiata salivary glands available in NCBI. Moreover, using an integrated transcriptomic and proteomic approach, we showed a correlation pattern of transcription and translation processes for the main gene families found, an important contribution to the research of triatomine sialomes. Furthermore, data generated here reinforces the secreted proteins encountered can greatly contribute for haematophagic habit, Trypanosoma cruzi transmission and development of therapeutic agent studies.
Collapse
Affiliation(s)
- Paula Beatriz Santiago
- Programa Pós-Graduação em Ciências Médicas, Faculty of Medicine, The University of Brasília, Brasília, Brazil
| | - Carla Nunes de Araújo
- Programa Pós-Graduação em Ciências Médicas, Faculty of Medicine, The University of Brasília, Brasília, Brazil; Faculty of Ceilândia, The University of Brasília, Brasília, Brazil.
| | - Sébastien Charneau
- Department of Cell Biology, The University of Brasília, Brasília, Brazil
| | | | - Teresa Cristina F Assumpção
- Vector Biology Section, Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Disease, Rockville, United States
| | | | - Yanna Reis Praça
- Programa Pós-Graduação em Ciências Médicas, Faculty of Medicine, The University of Brasília, Brasília, Brazil
| | | | | | | | - Tainá Raiol
- Department of Cell Biology, The University of Brasília, Brasília, Brazil; Instituto Leônidas e Maria Deane - Fiocruz Amazônia, Manaus, AM, Brazil
| | | | | | | | - José Marcos C Ribeiro
- Vector Biology Section, Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Disease, Rockville, United States
| | - Jaime Martins de Santana
- Programa Pós-Graduação em Ciências Médicas, Faculty of Medicine, The University of Brasília, Brasília, Brazil; Department of Cell Biology, The University of Brasília, Brasília, Brazil
| |
Collapse
|
28
|
Roversi P, Johnson S, Preston SG, Nunn MA, Paesen GC, Austyn JM, Nuttall PA, Lea SM. Structural basis of cholesterol binding by a novel clade of dendritic cell modulators from ticks. Sci Rep 2017; 7:16057. [PMID: 29167574 PMCID: PMC5700055 DOI: 10.1038/s41598-017-16413-2] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2017] [Accepted: 11/13/2017] [Indexed: 12/13/2022] Open
Abstract
Two crystal structures of Japanin, an 18 kDa immune-modulatory lipocalin from the Brown Ear Tick (Rhipicephalus appendiculatus), have been determined at 2.2 and 2.4 Å resolution. In both crystal forms the protein is in complex with cholesterol, which sits in a closed pocket at the centre of the lipocalin barrel. Both crystal forms are dimers, which are also observed in solution. Molecular modelling suggests that previously-described members of a tick protein family bearing high sequence homology to Japanin are also likely to bind cholesterol or cholesterol derivatives.
Collapse
Affiliation(s)
- Pietro Roversi
- Biochemistry Department, University of Oxford, Oxford, OX1 3QU, England, United Kingdom. .,Leicester Institute of Structural and Chemical Biology, Department of Molecular and Cell Biology, University of Leicester, Henry Wellcome Building, Lancaster Road, Leicester, LE1 7RH, England, United Kingdom.
| | - Steven Johnson
- Sir William Dunn School of Pathology, University of Oxford, Oxford, OX1 3RE, England, United Kingdom
| | - Stephen G Preston
- Department of Zoology, University of Oxford, Oxford, OX1 3PS, England, United Kingdom
| | - Miles A Nunn
- Akari Therapeutics, Plc, 75/76 Wimpole Street, London, W1G 9RT, England, United Kingdom
| | - Guido C Paesen
- Division of Structural Biology, Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, OX3 7BN, United Kingdom
| | - Jonathan M Austyn
- Nuffield Department of Surgical Sciences, John Radcliffe Hospital, University of Oxford, Oxford, OX3 9DU, England, United Kingdom
| | - Patricia A Nuttall
- Department of Zoology, University of Oxford, Oxford, OX1 3PS, England, United Kingdom
| | - Susan M Lea
- Sir William Dunn School of Pathology, University of Oxford, Oxford, OX1 3RE, England, United Kingdom.
| |
Collapse
|
29
|
Tick-host conflict: immunoglobulin E antibodies to tick proteins in patients with anaphylaxis to tick bite. Oncotarget 2017; 8:20630-20644. [PMID: 28423486 PMCID: PMC5400532 DOI: 10.18632/oncotarget.15243] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2016] [Accepted: 01/28/2017] [Indexed: 11/25/2022] Open
Abstract
Tick-borne infectious diseases and allergies are a growing problem worldwide. Tick bite allergy has been associated with the direct effect of immunoglobulin E (IgE) response to tick salivary antigens, or secondary to the induction of allergy to red meat consumption through IgE antibodies against the carbohydrate α-Gal (Gal α 1-3Gal β 1-(3)4GlcNAc-R). However, despite the growing burden of this pathology, the proteins associated with anaphylaxis to tick bite have not been characterized. To address this question, a comparative proteomics approach was used to characterize tick proteins producing an IgE antibody response in a healthy individual with record of tick bites, which had not resulted in any allergic reactions, and two patients with anaphylactic reactions to Rhipicephalus bursa or Hyalomma marginatum tick bites. Both patients and the healthy individual were red meat tolerant. The results supported a patient-specific IgE antibody response to tick species responsible for the anaphylaxis to tick bite. Both patients and the healthy individual serologically recognized tick proteins with and without α-Gal modifications, with proteins differentially recognized by patients but not control sera. These proteins could be used as potential antigens for diagnostics, treatment and prevention of tick bite-induced allergies.
Collapse
|
30
|
Mans BJ, Featherston J, de Castro MH, Pienaar R. Gene Duplication and Protein Evolution in Tick-Host Interactions. Front Cell Infect Microbiol 2017; 7:413. [PMID: 28993800 PMCID: PMC5622192 DOI: 10.3389/fcimb.2017.00413] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2017] [Accepted: 09/06/2017] [Indexed: 01/01/2023] Open
Abstract
Ticks modulate their hosts' defense responses by secreting a biopharmacopiea of hundreds to thousands of proteins and bioactive chemicals into the feeding site (tick-host interface). These molecules and their functions evolved over millions of years as ticks adapted to blood-feeding, tick lineages diverged, and host-shifts occurred. The evolution of new proteins with new functions is mainly dependent on gene duplication events. Central questions around this are the rates of gene duplication, when they occurred and how new functions evolve after gene duplication. The current review investigates these questions in the light of tick biology and considers the possibilities of ancient genome duplication, lineage specific expansion events, and the role that positive selection played in the evolution of tick protein function. It contrasts current views in tick biology regarding adaptive evolution with the more general view that neutral evolution may account for the majority of biological innovations observed in ticks.
Collapse
Affiliation(s)
- Ben J Mans
- Epidemiology, Parasites and Vectors, Agricultural Research Council-Onderstepoort Veterinary ResearchOnderstepoort, South Africa.,Department of Veterinary Tropical Diseases, University of PretoriaPretoria, South Africa.,Department of Life and Consumer Sciences, University of South AfricaPretoria, South Africa
| | - Jonathan Featherston
- Agricultural Research Council-The Biotechnology PlatformOnderstepoort, South Africa
| | - Minique H de Castro
- Epidemiology, Parasites and Vectors, Agricultural Research Council-Onderstepoort Veterinary ResearchOnderstepoort, South Africa.,Department of Life and Consumer Sciences, University of South AfricaPretoria, South Africa.,Agricultural Research Council-The Biotechnology PlatformOnderstepoort, South Africa
| | - Ronel Pienaar
- Epidemiology, Parasites and Vectors, Agricultural Research Council-Onderstepoort Veterinary ResearchOnderstepoort, South Africa
| |
Collapse
|
31
|
de Castro MH, de Klerk D, Pienaar R, Rees DJG, Mans BJ. Sialotranscriptomics of Rhipicephalus zambeziensis reveals intricate expression profiles of secretory proteins and suggests tight temporal transcriptional regulation during blood-feeding. Parasit Vectors 2017; 10:384. [PMID: 28797301 PMCID: PMC5553602 DOI: 10.1186/s13071-017-2312-4] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2017] [Accepted: 07/27/2017] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Ticks secrete a diverse mixture of secretory proteins into the host to evade its immune response and facilitate blood-feeding, making secretory proteins attractive targets for the production of recombinant anti-tick vaccines. The largely neglected tick species, Rhipicephalus zambeziensis, is an efficient vector of Theileria parva in southern Africa but its available sequence information is limited. Next generation sequencing has advanced sequence availability for ticks in recent years and has assisted the characterisation of secretory proteins. This study focused on the de novo assembly and annotation of the salivary gland transcriptome of R. zambeziensis and the temporal expression of secretory protein transcripts in female and male ticks, before the onset of feeding and during early and late feeding. RESULTS The sialotranscriptome of R. zambeziensis yielded 23,631 transcripts from which 13,584 non-redundant proteins were predicted. Eighty-six percent of these contained a predicted start and stop codon and were estimated to be putatively full-length proteins. A fifth (2569) of the predicted proteins were annotated as putative secretory proteins and explained 52% of the expression in the transcriptome. Expression analyses revealed that 2832 transcripts were differentially expressed among feeding time points and 1209 between the tick sexes. The expression analyses further indicated that 57% of the annotated secretory protein transcripts were differentially expressed. Dynamic expression profiles of secretory protein transcripts were observed during feeding of female ticks. Whereby a number of transcripts were upregulated during early feeding, presumably for feeding site establishment and then during late feeding, 52% of these were downregulated, indicating that transcripts were required at specific feeding stages. This suggested that secretory proteins are under stringent transcriptional regulation that fine-tunes their expression in salivary glands during feeding. No open reading frames were predicted for 7947 transcripts. This class represented 17% of the differentially expressed transcripts, suggesting a potential transcriptional regulatory function of long non-coding RNA in tick blood-feeding. CONCLUSIONS The assembled sialotranscriptome greatly expands the sequence availability of R. zambeziensis, assists in our understanding of the transcription of secretory proteins during blood-feeding and will be a valuable resource for future vaccine candidate selection.
Collapse
Affiliation(s)
- Minique Hilda de Castro
- Epidemiology, Parasites and Vectors, Onderstepoort Veterinary Research, Agricultural Research Council, Onderstepoort, South Africa.,Biotechnology Platform, Agricultural Research Council, Onderstepoort, South Africa.,College of Agriculture and Environmental Sciences, University of South Africa, Johannesburg, South Africa
| | - Daniel de Klerk
- Epidemiology, Parasites and Vectors, Onderstepoort Veterinary Research, Agricultural Research Council, Onderstepoort, South Africa
| | - Ronel Pienaar
- Epidemiology, Parasites and Vectors, Onderstepoort Veterinary Research, Agricultural Research Council, Onderstepoort, South Africa.,Department of Veterinary Tropical Diseases, Faculty of Veterinary Science, University of Pretoria, Pretoria, South Africa
| | - D Jasper G Rees
- Biotechnology Platform, Agricultural Research Council, Onderstepoort, South Africa.,College of Agriculture and Environmental Sciences, University of South Africa, Johannesburg, South Africa
| | - Ben J Mans
- Epidemiology, Parasites and Vectors, Onderstepoort Veterinary Research, Agricultural Research Council, Onderstepoort, South Africa. .,College of Agriculture and Environmental Sciences, University of South Africa, Johannesburg, South Africa. .,Department of Veterinary Tropical Diseases, Faculty of Veterinary Science, University of Pretoria, Pretoria, South Africa.
| |
Collapse
|
32
|
Kazimírová M, Thangamani S, Bartíková P, Hermance M, Holíková V, Štibrániová I, Nuttall PA. Tick-Borne Viruses and Biological Processes at the Tick-Host-Virus Interface. Front Cell Infect Microbiol 2017; 7:339. [PMID: 28798904 PMCID: PMC5526847 DOI: 10.3389/fcimb.2017.00339] [Citation(s) in RCA: 80] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2017] [Accepted: 07/11/2017] [Indexed: 01/08/2023] Open
Abstract
Ticks are efficient vectors of arboviruses, although less than 10% of tick species are known to be virus vectors. Most tick-borne viruses (TBV) are RNA viruses some of which cause serious diseases in humans and animals world-wide. Several TBV impacting human or domesticated animal health have been found to emerge or re-emerge recently. In order to survive in nature, TBV must infect and replicate in both vertebrate and tick cells, representing very different physiological environments. Information on molecular mechanisms that allow TBV to switch between infecting and replicating in tick and vertebrate cells is scarce. In general, ticks succeed in completing their blood meal thanks to a plethora of biologically active molecules in their saliva that counteract and modulate different arms of the host defense responses (haemostasis, inflammation, innate and acquired immunity, and wound healing). The transmission of TBV occurs primarily during tick feeding and is a complex process, known to be promoted by tick saliva constituents. However, the underlying molecular mechanisms of TBV transmission are poorly understood. Immunomodulatory properties of tick saliva helping overcome the first line of defense to injury and early interactions at the tick-host skin interface appear to be essential in successful TBV transmission and infection of susceptible vertebrate hosts. The local host skin site of tick attachment, modulated by tick saliva, is an important focus of virus replication. Immunomodulation of the tick attachment site also promotes co-feeding transmission of viruses from infected to non-infected ticks in the absence of host viraemia (non-viraemic transmission). Future research should be aimed at identification of the key tick salivary molecules promoting virus transmission, and a molecular description of tick-host-virus interactions and of tick-mediated skin immunomodulation. Such insights will enable the rationale design of anti-tick vaccines that protect against disease caused by tick-borne viruses.
Collapse
Affiliation(s)
- Mária Kazimírová
- Department of Medical Zoology, Institute of Zoology, Slovak Academy of SciencesBratislava, Slovakia
| | - Saravanan Thangamani
- Department of Pathology, University of Texas Medical BranchGalveston, TX, United States
- Institute for Human Infections and Immunity, University of Texas Medical BranchGalveston, TX, United States
- Center for Tropical Diseases, University of Texas Medical BranchGalveston, TX, United States
| | - Pavlína Bartíková
- Biomedical Research Center, Institute of Virology, Slovak Academy of SciencesBratislava, Slovakia
| | - Meghan Hermance
- Department of Pathology, University of Texas Medical BranchGalveston, TX, United States
- Institute for Human Infections and Immunity, University of Texas Medical BranchGalveston, TX, United States
- Center for Tropical Diseases, University of Texas Medical BranchGalveston, TX, United States
| | - Viera Holíková
- Biomedical Research Center, Institute of Virology, Slovak Academy of SciencesBratislava, Slovakia
| | - Iveta Štibrániová
- Biomedical Research Center, Institute of Virology, Slovak Academy of SciencesBratislava, Slovakia
| | - Patricia A. Nuttall
- Department of Zoology, University of OxfordOxford, United Kingdom
- Centre for Ecology and HydrologyWallingford, United Kingdom
| |
Collapse
|
33
|
Wilson JM, Schuyler AJ, Schroeder N, Platts-Mills TAE. Galactose-α-1,3-Galactose: Atypical Food Allergen or Model IgE Hypersensitivity? Curr Allergy Asthma Rep 2017; 17:8. [PMID: 28224342 DOI: 10.1007/s11882-017-0672-7] [Citation(s) in RCA: 57] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
PURPOSE OF REVIEW Galactose-α-1,3-galactose (α-gal) is a carbohydrate allergen with several unique characteristics. In this article, we discuss some recent advances in our understanding of the 'alpha-gal syndrome,' highlight data supporting the role of ticks in pathogenesis, and speculate on immune mechanisms that lead to sensitization. RECENT FINDINGS First described as the target of IgE in individuals suffering immediate hypersensitivity reactions to the novel anti-EGF monoclonal antibody cetuximab, it is now clear that α-gal sensitization is associated with mammalian meat allergy as well as reactions to other mammalian products. Unlike traditional IgE-mediated food allergies, reactions to α-gal often do not manifest until several hours following an exposure, although co-factors can influence the presentation. Multiple pieces of evidence, including recent work with a mouse model, point to the fact that sensitization is mediated by exposure to certain hard ticks and increasingly we are aware of its globally widespread impact. The oligosaccharide α-gal represents a novel allergen with several unusual clinical features. It has been recognized now on multiple continents and its clinical presentation can be quite variable. Moreover, efforts to delineate the mechanisms leading to α-gal sensitization may have ramifications for our broader understanding of type 2 immunity.
Collapse
Affiliation(s)
- Jeffrey M Wilson
- Division of Allergy, Asthma, and Immunology, University of Virginia Health System, PO Box 801355, Charlottesville, VA, 22908, USA.
| | - Alexander J Schuyler
- Division of Allergy, Asthma, and Immunology, University of Virginia Health System, PO Box 801355, Charlottesville, VA, 22908, USA
| | - Nikhila Schroeder
- Division of Allergy, Asthma, and Immunology, University of Virginia Health System, PO Box 801355, Charlottesville, VA, 22908, USA
| | - Thomas A E Platts-Mills
- Division of Allergy, Asthma, and Immunology, University of Virginia Health System, PO Box 801355, Charlottesville, VA, 22908, USA
| |
Collapse
|
34
|
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
|
35
|
Zavašnik-Bergant T, Vidmar R, Sekirnik A, Fonović M, Salát J, Grunclová L, Kopáček P, Turk B. Salivary Tick Cystatin OmC2 Targets Lysosomal Cathepsins S and C in Human Dendritic Cells. Front Cell Infect Microbiol 2017; 7:288. [PMID: 28713775 PMCID: PMC5492865 DOI: 10.3389/fcimb.2017.00288] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2017] [Accepted: 06/14/2017] [Indexed: 11/14/2022] Open
Abstract
To ensure successful feeding tick saliva contains a number of inhibitory proteins that interfere with the host immune response and help to create a permissive environment for pathogen transmission. Among the potential targets of the salivary cystatins are two host cysteine proteases, cathepsin S, which is essential for antigen- and invariant chain-processing, and cathepsin C (dipeptidyl peptidase 1, DPP1), which plays a critical role in processing and activation of the granule serine proteases. Here, the effect of salivary cystatin OmC2 from Ornithodoros moubata was studied using differentiated MUTZ-3 cells as a model of immature dendritic cells of the host skin. Following internalization, cystatin OmC2 was initially found to inhibit the activity of several cysteine cathepsins, as indicated by the decreased rates of degradation of fluorogenic peptide substrates. To identify targets, affinity chromatography was used to isolate His-tagged cystatin OmC2 together with the bound proteins from MUTZ-3 cells. Cathepsins S and C were identified in these complexes by mass spectrometry and confirmed by immunoblotting. Furthermore, reduced increase in the surface expression of MHC II and CD86, which are associated with the maturation of dendritic cells, was observed. In contrast, human inhibitor cystatin C, which is normally expressed and secreted by dendritic cells, did not affect the expression of CD86. It is proposed that internalization of salivary cystatin OmC2 by the host dendritic cells targets cathepsins S and C, thereby affecting their maturation.
Collapse
Affiliation(s)
- Tina Zavašnik-Bergant
- Department of Biochemistry, Molecular and Structural Biology, Jožef Stefan InstituteLjubljana, Slovenia
| | - Robert Vidmar
- Department of Biochemistry, Molecular and Structural Biology, Jožef Stefan InstituteLjubljana, Slovenia
| | - Andreja Sekirnik
- Department of Biochemistry, Molecular and Structural Biology, Jožef Stefan InstituteLjubljana, Slovenia
| | - Marko Fonović
- Department of Biochemistry, Molecular and Structural Biology, Jožef Stefan InstituteLjubljana, Slovenia
| | - Jiří Salát
- Institute of Parasitology, Biology Centre of the Czech Academy of SciencesČeské Budějovice, Czechia
| | - Lenka Grunclová
- Institute of Parasitology, Biology Centre of the Czech Academy of SciencesČeské Budějovice, Czechia
| | - Petr Kopáček
- Institute of Parasitology, Biology Centre of the Czech Academy of SciencesČeské Budějovice, Czechia
| | - Boris Turk
- Department of Biochemistry, Molecular and Structural Biology, Jožef Stefan InstituteLjubljana, Slovenia.,Centre of Excellence for Integrated Approaches in Chemistry and Biology of ProteinsLjubljana, Slovenia.,Faculty of Chemistry and Chemical Technology, University of LjubljanaLjubljana, Slovenia
| |
Collapse
|
36
|
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: 191] [Impact Index Per Article: 23.9] [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
|
37
|
Bukiya AN, Dopico AM. Common structural features of cholesterol binding sites in crystallized soluble proteins. J Lipid Res 2017; 58:1044-1054. [PMID: 28420706 DOI: 10.1194/jlr.r073452] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2016] [Revised: 04/12/2017] [Indexed: 01/24/2023] Open
Abstract
Cholesterol-protein interactions are essential for the architectural organization of cell membranes and for lipid metabolism. While cholesterol-sensing motifs in transmembrane proteins have been identified, little is known about cholesterol recognition by soluble proteins. We reviewed the structural characteristics of binding sites for cholesterol and cholesterol sulfate from crystallographic structures available in the Protein Data Bank. This analysis unveiled key features of cholesterol-binding sites that are present in either all or the majority of sites: i) the cholesterol molecule is generally positioned between protein domains that have an organized secondary structure; ii) the cholesterol hydroxyl/sulfo group is often partnered by Asn, Gln, and/or Tyr, while the hydrophobic part of cholesterol interacts with Leu, Ile, Val, and/or Phe; iii) cholesterol hydrogen-bonding partners are often found on α-helices, while amino acids that interact with cholesterol's hydrophobic core have a slight preference for β-strands and secondary structure-lacking protein areas; iv) the steroid's C21 and C26 constitute the "hot spots" most often seen for steroid-protein hydrophobic interactions; v) common "cold spots" are C8-C10, C13, and C17, at which contacts with the proteins were not detected. Several common features we identified for soluble protein-steroid interaction appear evolutionarily conserved.
Collapse
Affiliation(s)
- Anna N Bukiya
- Department of Pharmacology, College of Medicine, University of Tennessee Health Science Center, Memphis, TN 38103
| | - Alejandro M Dopico
- Department of Pharmacology, College of Medicine, University of Tennessee Health Science Center, Memphis, TN 38103
| |
Collapse
|
38
|
Wang F, Lu X, Guo F, Gong H, Zhang H, Zhou Y, Cao J, Zhou J. The immunomodulatory protein RH36 is relating to blood-feeding success and oviposition in hard ticks. Vet Parasitol 2017; 240:49-59. [PMID: 28449954 DOI: 10.1016/j.vetpar.2017.03.017] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2017] [Revised: 03/20/2017] [Accepted: 03/22/2017] [Indexed: 11/30/2022]
Abstract
An immunomodulatory protein designated RH36 was identified in the tick Rhipicephalus haemaphysaloides. The cDNA sequence of RH36 has 844bp and encodes a deduced protein with a predicted molecular weight of 24kDa. Bioinformatics analysis indicated that RH36 presented a degree of similarity of 34.36% with the immunomodulatory protein p36 from the tick Dermacentor andersoni. The recombinant RH36 (rRH36) expressed in Sf9 insect cells suppressed the T-lymphocyte mitogen-driven in vitro proliferation of splenocytes and the expression of several cytokines such as IL-2, IL-12, and TNF-α. Furthermore, the proliferation of splenocytes isolated from rRH36-inoculated mice was significantly lower than that in control mice, suggesting that rRH36 could directly suppress immune responses in vivo. In addition, microarray analysis of splenocytes indicated that the expression of several immunomodulatory genes was downregulated by rRH36. The silencing of the RH36 gene by RNAi led to a 37.5% decrease in the tick attachment rate 24h after placement into the rabbit ears, whereas vaccination with RH36 caused a 53.06% decrease in the tick engorgement rate. Unexpectedly, RNAi induced a significant decrease in the oviposition rate, ovary weight at day 12 after engorgement, and egg-hatching rate. The effects of RH36 on blood feeding and oviposition were further confirmed by vaccination tests using the recombinant protein. These results indicate that RH36 is a novel member of immunosuppressant proteins and affects tick blood feeding and oviposition.
Collapse
Affiliation(s)
- Fangfang Wang
- Key Laboratory of Animal Parasitology of Ministry of Agriculture, Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai 200241, China
| | - Xiaojuan Lu
- Key Laboratory of Animal Parasitology of Ministry of Agriculture, Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai 200241, China
| | - Fengxun Guo
- Key Laboratory of Animal Parasitology of Ministry of Agriculture, Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai 200241, China
| | - Haiyan Gong
- Key Laboratory of Animal Parasitology of Ministry of Agriculture, Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai 200241, China
| | - Houshuang Zhang
- Key Laboratory of Animal Parasitology of Ministry of Agriculture, Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai 200241, China
| | - Yongzhi Zhou
- Key Laboratory of Animal Parasitology of Ministry of Agriculture, Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai 200241, China
| | - Jie Cao
- Key Laboratory of Animal Parasitology of Ministry of Agriculture, Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai 200241, China
| | - Jinlin Zhou
- Key Laboratory of Animal Parasitology of Ministry of Agriculture, Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai 200241, China; Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou 225009, China.
| |
Collapse
|
39
|
Franzin AM, Maruyama SR, Garcia GR, Oliveira RP, Ribeiro JMC, Bishop R, Maia AAM, Moré DD, Ferreira BR, Santos IKFDM. Immune and biochemical responses in skin differ between bovine hosts genetically susceptible and resistant to the cattle tick Rhipicephalus microplus. Parasit Vectors 2017; 10:51. [PMID: 28143523 PMCID: PMC5282843 DOI: 10.1186/s13071-016-1945-z] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2016] [Accepted: 12/16/2016] [Indexed: 11/17/2022] Open
Abstract
Background Ticks attach to and penetrate their hosts’ skin and inactivate multiple components of host responses in order to acquire a blood meal. Infestation loads with the cattle tick, Rhipicephalus microplus, are heritable: some breeds carry high loads of reproductively successful ticks, whereas in others, few ticks feed and reproduce efficiently. Methods In order to elucidate the mechanisms that result in the different outcomes of infestations with cattle ticks, we examined global gene expression and inflammation induced by tick bites in skins from one resistant and one susceptible breed of cattle that underwent primary infestations with larvae and nymphs of R. microplus. We also examined the expression profiles of genes encoding secreted tick proteins that mediate parasitism in larvae and nymphs feeding on these breeds. Results Functional analyses of differentially expressed genes in the skin suggest that allergic contact-like dermatitis develops with ensuing production of IL-6, CXCL-8 and CCL-2 and is sustained by HMGB1, ISG15 and PKR, leading to expression of pro-inflammatory chemokines and cytokines that recruit granulocytes and T lymphocytes. Importantly, this response is delayed in susceptible hosts. Histopathological analyses of infested skins showed inflammatory reactions surrounding tick cement cones that enable attachment in both breeds, but in genetically tick-resistant bovines they destabilized the cone. The transcription data provided insights into tick-mediated activation of basophils, which have previously been shown to be a key to host resistance in model systems. Skin from tick-susceptible bovines expressed more transcripts encoding enzymes that detoxify tissues. Interestingly, these enzymes also produce volatile odoriferous compounds and, accordingly, skin rubbings from tick-susceptible bovines attracted significantly more tick larvae than rubbings from resistant hosts. Moreover, transcripts encoding secreted modulatory molecules by the tick were significantly more abundant in larval and in nymphal salivary glands from ticks feeding on susceptible bovines. Conclusions Compared with tick-susceptible hosts, genes encoding enzymes producing volatile compounds exhibit significantly lower expression in resistant hosts, which may render them less attractive to larvae; resistant hosts expose ticks to an earlier inflammatory response, which in ticks is associated with significantly lower expression of genes encoding salivary proteins that suppress host immunity, inflammation and coagulation. Electronic supplementary material The online version of this article (doi:10.1186/s13071-016-1945-z) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Alessandra Mara Franzin
- Departament of Biochemistry and Immunology, Ribeirão Preto School of Medicine, University of São Paulo, Ribeirão Preto, SP, 14049-900, Brazil
| | - Sandra Regina Maruyama
- Departament of Biochemistry and Immunology, Ribeirão Preto School of Medicine, University of São Paulo, Ribeirão Preto, SP, 14049-900, Brazil.,Department of Genetics and Evolution, Federal University of São Carlos, São Carlos, SP, 13565-905, Brazil
| | - Gustavo Rocha Garcia
- Departament of Biochemistry and Immunology, Ribeirão Preto School of Medicine, University of São Paulo, Ribeirão Preto, SP, 14049-900, Brazil
| | - Rosane Pereira Oliveira
- Department of Animal Sciences, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA.,Integrative Medicine Program, School of Medicine, University of California Davis, Sacramento, CA, 95817, USA
| | - José Marcos Chaves Ribeiro
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Richard Bishop
- International Livestock Research Institute, Nairobi, Kenya.,Embrapa Pecuária Sudeste, São Carlos, SP, 13560-970, Brazil
| | - Antônio Augusto Mendes Maia
- Department of Basic Sciences, School of Animal Science and Food Technology, University of São Paulo, Pirassununga, SP, 13635-900, Brazil
| | - Daniela Dantas Moré
- Departament of Biochemistry and Immunology, Ribeirão Preto School of Medicine, University of São Paulo, Ribeirão Preto, SP, 14049-900, Brazil.,Department of Veterinary Microbiology & Pathology, Washington State University, Pullman, WA, 99164-7040, USA
| | | | | |
Collapse
|
40
|
Mendes MT, Carvalho-Costa TM, da Silva MV, Anhê ACBM, Guimarães RM, da Costa TA, Ramirez LE, Rodrigues V, Oliveira CJF. Effect of the saliva from different triatomine species on the biology and immunity of TLR-4 ligand and Trypanosoma cruzi-stimulated dendritic cells. Parasit Vectors 2016; 9:634. [PMID: 27938380 PMCID: PMC5148907 DOI: 10.1186/s13071-016-1890-x] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2016] [Accepted: 11/16/2016] [Indexed: 02/07/2023] Open
Abstract
Background Triatomines are blood-sucking vectors of Trypanosoma cruzi, the causative agent of Chagas disease. During feeding, triatomines surpass the skin host response through biomolecules present in their saliva. Dendritic cells (DCs) play a crucial role in the induction of the protection to aggressive agents, including blood-sucking arthropods. Here, we evaluated if salivary components of triatomines from different genera evade the host immunity by modulating the biology and the function of LPS- or T. cruzi-stimulated DCs. Methods Saliva of Panstrongylus lignarius, Meccus pallidipennis, Triatoma lecticularia and Rhodnius prolixus were obtained by dissection of salivary glands and the DCs were obtained from the differentiation of mouse bone marrow precursors. Results The differentiation of DCs was inhibited by saliva of all species tested. Saliva differentially inhibited the expression of MHC-II, CD40, CD80 and CD86 in LPS-matured DCs. Except for the saliva of R. prolixus, which induced IL-6 cytokine production, TNF-α, IL-12 and IL-6 were inhibited by the saliva of the other three tested species and IL-10 was increased in all of them. Saliva per se, also induced the production of IL-12, IL-6 and IL-10. Only the saliva of R. prolixus induced DCs apoptosis. The presence of PGE2 was not detected in the saliva of the four triatomines studied. Finally, T. cruzi invasion on DCs is enhanced by the presence of the triatomine saliva. Conclusions These results demonstrate that saliva from different triatomine species exhibit immunomodulatory effects on LPS and T. cruzi-stimulated DCs. These effects could be related to hematophagy and transmission of T. cruzi during feeding. Electronic supplementary material The online version of this article (doi:10.1186/s13071-016-1890-x) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Maria Tays Mendes
- Department of Biological Sciences, University of Texas at El Paso, El Paso, TX, USA.,Laboratory of Immunology, Federal University of Triângulo Mineiro, Uberaba, Minas Gerais, Brazil
| | | | - Marcos Vinicius da Silva
- Laboratory of Immunology, Federal University of Triângulo Mineiro, Uberaba, Minas Gerais, Brazil.
| | | | - Rafaela Mano Guimarães
- Laboratory of Immunology, Federal University of Triângulo Mineiro, Uberaba, Minas Gerais, Brazil
| | - Thiago Alvares da Costa
- Laboratory of Immunology, Federal University of Triângulo Mineiro, Uberaba, Minas Gerais, Brazil
| | - Luis Eduardo Ramirez
- Laboratory of Immunology, Federal University of Triângulo Mineiro, Uberaba, Minas Gerais, Brazil
| | - Virmondes Rodrigues
- Laboratory of Immunology, Federal University of Triângulo Mineiro, Uberaba, Minas Gerais, Brazil
| | | |
Collapse
|
41
|
Wang Y, Li Z, Zhou Y, Cao J, Zhang H, Gong H, Zhou J. Specific histamine binding activity of a new lipocalin from Hyalomma asiaticum (Ixodidae) and therapeutic effects on allergic asthma in mice. Parasit Vectors 2016; 9:506. [PMID: 27639693 PMCID: PMC5027092 DOI: 10.1186/s13071-016-1790-0] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2016] [Accepted: 09/02/2016] [Indexed: 11/23/2022] Open
Abstract
Background Lipocalin proteins are secreted by tick salivary glands as an important strategy to interfere with the immune response of hosts. A large number of lipocalins are secreted, but the functions of most of these proteins are unclear. Here, we report a new lipocalin protein with particular histamine binding capacity, which was isolated from the salivary glands of the tick Hyalomma asiaticum. Methods The full length cDNA of the Ha24 gene was obtained by RACE, and Ha24 gene was expressed in E. coli; after protein purification and mice immunizations, specific Polyclonal antibodies (PcAb) were created in response to the recombinant protein. Reverse transcription PCR (RT-PCR), Quantitative PCR (Q-PCR), indirect immunofluorescence antibody (IFA) assay and western blot were used to detect the existence of native Ha24 in ticks. To confirm the histamine-binding capacity of rHa24, a histamine-binding assay was completed in vitro (ELISA) and in vivo by inhibition of allergic asthma in mice. Results Ha24 is coded by 681 bases, contains 227 amino acids, and has a molecular weight of 23.3 kDa. Abundant expression in the salivary glands of feeding ticks was confirmed by the identification of native Ha24 in ticks. The results of a histamine binding assay both in vitro and in vivo demonstrated that rHa24 binds specifically with histamine in a dose-dependent manner, and can provide relief from allergic asthma in mice. Conclusions Ha24 is a new tick lipocalin with specific histamine binding activity that can provide relief from host inflammation response.
Collapse
Affiliation(s)
- Yanan Wang
- Key Laboratory of Animal Parasitology of Ministry of Agriculture, Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai, 200241, China
| | - Zhuang Li
- Chizhou Vocational and Technique Colleague, Chizhou, 247000, China
| | - Yongzhi Zhou
- Key Laboratory of Animal Parasitology of Ministry of Agriculture, Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai, 200241, China
| | - Jie Cao
- Key Laboratory of Animal Parasitology of Ministry of Agriculture, Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai, 200241, China
| | - Houshuang Zhang
- Key Laboratory of Animal Parasitology of Ministry of Agriculture, Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai, 200241, China
| | - Haiyan Gong
- Key Laboratory of Animal Parasitology of Ministry of Agriculture, Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai, 200241, China
| | - Jinlin Zhou
- Key Laboratory of Animal Parasitology of Ministry of Agriculture, Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai, 200241, China. .,Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, 225009, China.
| |
Collapse
|
42
|
Valdés JJ, Cabezas-Cruz A, Sima R, Butterill PT, Růžek D, Nuttall PA. Substrate prediction of Ixodes ricinus salivary lipocalins differentially expressed during Borrelia afzelii infection. Sci Rep 2016; 6:32372. [PMID: 27584086 PMCID: PMC5008119 DOI: 10.1038/srep32372] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2016] [Accepted: 08/05/2016] [Indexed: 11/13/2022] Open
Abstract
Evolution has provided ticks with an arsenal of bioactive saliva molecules that counteract host defense mechanisms. This salivary pharmacopoeia enables blood-feeding while enabling pathogen transmission. High-throughput sequencing of tick salivary glands has thus become a major focus, revealing large expansion within protein encoding gene families. Among these are lipocalins, ubiquitous barrel-shaped proteins that sequester small, typically hydrophobic molecules. This study was initiated by mining the Ixodes ricinus salivary gland transcriptome for specific, uncharacterized lipocalins: three were identified. Differential expression of these I. ricinus lipocalins during feeding at distinct developmental stages and in response to Borrelia afzelii infection suggests a role in transmission of this Lyme disease spirochete. A phylogenetic analysis using 803 sequences places the three I. ricinus lipocalins with tick lipocalins that sequester monoamines, leukotrienes and fatty acids. Both structural analysis and biophysical simulations generated robust predictions showing these I. ricinus lipocalins have the potential to bind monoamines similar to other tick species previously reported. The multidisciplinary approach employed in this study characterized unique lipocalins that play a role in tick blood-feeding and transmission of the most important tick-borne pathogen in North America and Eurasia.
Collapse
Affiliation(s)
- James J Valdés
- Institute of Parasitology, The Czech Academy of Sciences, Branišovská 31, CZ-37005 České Budějovice, Czech Republic.,Department of Virology, Veterinary Research Institute, Hudcova 70, CZ-62100 Brno, Czech Republic
| | - Alejandro Cabezas-Cruz
- Center for Infection and Immunity of Lille (CIIL), INSERM U1019 - CNRS UMR 8204, Université Lille Nord de France, Institut Pasteur de Lille, Lille, France
| | - Radek Sima
- Institute of Parasitology, The Czech Academy of Sciences, Branišovská 31, CZ-37005 České Budějovice, Czech Republic
| | - Philip T Butterill
- Biology Center, The Czech Academy of Sciences, University of South Bohemia, Branišovská 31, CZ-37005 České Budějovice, Czech Republic
| | - Daniel Růžek
- Institute of Parasitology, The Czech Academy of Sciences, Branišovská 31, CZ-37005 České Budějovice, Czech Republic.,Department of Virology, Veterinary Research Institute, Hudcova 70, CZ-62100 Brno, Czech Republic.,Biology Center, The Czech Academy of Sciences, University of South Bohemia, Branišovská 31, CZ-37005 České Budějovice, Czech Republic
| | | |
Collapse
|
43
|
de Castro MH, de Klerk D, Pienaar R, Latif AA, Rees DJG, Mans BJ. De novo assembly and annotation of the salivary gland transcriptome of Rhipicephalus appendiculatus male and female ticks during blood feeding. Ticks Tick Borne Dis 2016; 7:536-48. [DOI: 10.1016/j.ttbdis.2016.01.014] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2015] [Revised: 12/23/2015] [Accepted: 01/20/2016] [Indexed: 01/19/2023]
|
44
|
Mans BJ, de Castro MH, Pienaar R, de Klerk D, Gaven P, Genu S, Latif AA. Ancestral reconstruction of tick lineages. Ticks Tick Borne Dis 2016; 7:509-35. [DOI: 10.1016/j.ttbdis.2016.02.002] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2015] [Revised: 01/26/2016] [Accepted: 02/02/2016] [Indexed: 01/15/2023]
|
45
|
Tian Y, Chen W, Mo G, Chen R, Fang M, Yedid G, Yan X. An Immunosuppressant Peptide from the Hard Tick Amblyomma variegatum. Toxins (Basel) 2016; 8:133. [PMID: 27153086 PMCID: PMC4885048 DOI: 10.3390/toxins8050133] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2016] [Revised: 04/23/2016] [Accepted: 04/26/2016] [Indexed: 12/15/2022] Open
Abstract
Ixodid ticks are well known for spreading transmitted tick-borne pathogens while being attached to their hosts for almost 1-2 weeks to obtain blood meals. Thus, they must secrete many immunosuppressant factors to combat the hosts' immune system. In the present work, we investigated an immunosuppressant peptide of the hard tick Amblyomma variegatum. This peptide, named amregulin, is composed of 40 residues with an amino acid sequence of HLHMHGNGATQVFKPRLVLKCPNAAQLIQPGKLQRQLLLQ. A cDNA of the precursor peptide was obtained from the National Center for Biotechnology Information (NCBI, Bethesda, MD, USA). In rat splenocytes, amregulin exerts significant anti-inflammatory effects by inhibiting the secretion of inflammatory factors in vitro, such as tumor necrosis factor-alpha (TNF-α), interleukin-1 (IL-1), interleukin-8 (IL-8) and interferon-gamma (IFN-γ). In rat splenocytes, treated with amregulin, compared to lipopolysaccharide (LPS) alone, the inhibition of the above inflammatory factors was significant at all tested concentrations (2, 4 and 8 µg/mL). Amregulin shows strong free radical scavenging and antioxidant activities (5, 10 and 20 µg/mL) in vitro. Amregulin also significantly inhibits adjuvant-induced paw inflammation in mouse models in vivo. This peptide may facilitate the ticks' successful blood feeding and may lead to host immunotolerance of the tick. These findings have important implications for the understanding of tick-host interactions and the co-evolution between ticks and the viruses that they bear.
Collapse
Affiliation(s)
- Yufeng Tian
- Clinical Laboratory, People's Hospital of Rizhao, 126th Taian Road, Rizhao 276826, Shandong, China.
- College of Life Sciences, Nanjing Agricultural University, Weigang #1, Nanjing 210095, Jiangsu, China.
| | - Wenlin Chen
- Yunnan Clinical Research Center of Breast Cancer, The Third Affiliated Hospital of Kunming Medical College, Kunming 650032, China.
| | - Guoxiang Mo
- College of Life Sciences, Nanjing Agricultural University, Weigang #1, Nanjing 210095, Jiangsu, China.
| | - Ran Chen
- College of Life Sciences, Nanjing Agricultural University, Weigang #1, Nanjing 210095, Jiangsu, China.
| | - Mingqian Fang
- College of Life Sciences, Nanjing Agricultural University, Weigang #1, Nanjing 210095, Jiangsu, China.
| | - Gabriel Yedid
- College of Life Sciences, Nanjing Agricultural University, Weigang #1, Nanjing 210095, Jiangsu, China.
| | - Xiuwen Yan
- College of Life Sciences, Nanjing Agricultural University, Weigang #1, Nanjing 210095, Jiangsu, China.
| |
Collapse
|
46
|
Cabezas-Cruz A, Mateos-Hernández L, Pérez-Cruz M, Valdés JJ, Mera IGFD, Villar M, de la Fuente J. Regulation of the Immune Response to α-Gal and Vector-borne Diseases. Trends Parasitol 2015; 31:470-476. [DOI: 10.1016/j.pt.2015.06.016] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2015] [Revised: 05/18/2015] [Accepted: 06/24/2015] [Indexed: 11/26/2022]
|
47
|
Vachiery N, Puech C, Cavelier P, Rodrigues V, Aprelon R, Lefrançois T, Martinez D, Epardaud M. An in vitro model to assess the immunosuppressive effect of tick saliva on the mobilization of inflammatory monocyte-derived cells. Vet Res 2015; 46:117. [PMID: 26412247 PMCID: PMC4586012 DOI: 10.1186/s13567-015-0229-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2015] [Accepted: 07/27/2015] [Indexed: 11/10/2022] Open
Abstract
Tick-borne pathogens cause potent infections. These pathogens benefit from molecules contained in tick saliva that have evolved to modulate host innate and adaptive immune responses. This is called “saliva-activated transmission” and enables tick-borne pathogens to evade host immune responses. Ticks feed on their host for relatively long periods; thus, mechanisms counteracting the inflammation-driven recruitment and activation of innate effector cells at the bite site, are an effective strategy to escape the immune response. Here, we developed an original in vitro model to evaluate and to characterize the immunomodulatory effects of tick saliva that prevent the establishment of a local inflammatory immune response. This model mimics the tick bite and enables the assessment of the effect of saliva on the inflammatory-associated dynamic recruitment of cells from the mononuclear phagocyte system. Using this model, we were able to recapitulate the dual effect of tick saliva on the mobilization of inflammatory monocyte-derived cells, i.e. (i) impaired recruitment of monocytes from the blood to the bite wound; and (ii) poor mobilization of monocyte-derived cells from the skin to the draining lymph node. This simple tool reconstitutes the effect of tick saliva in vivo, which we characterized in the mouse, and should enable the identification of important factors facilitating pathogen infection. Furthermore, this model may be applied to the characterization of any pathogen-derived immunosuppressive molecule affecting the establishment of the inflammatory immune response.
Collapse
Affiliation(s)
- Nathalie Vachiery
- INRA-CIRAD, UMR 1309 Contrôle des maladies animales, exotiques et émergentes, F-97170, Petit-Bourg, Guadeloupe, France.
| | - Carinne Puech
- INRA-CIRAD, UMR 1309 Contrôle des maladies animales, exotiques et émergentes, F-34398, Montpellier, France.
| | - Patricia Cavelier
- UMR C5535 Institut de Génétique Moléculaire de Montpellier, Montpellier, France.
| | - Valérie Rodrigues
- INRA-CIRAD, UMR 1309 Contrôle des maladies animales, exotiques et émergentes, F-34398, Montpellier, France.
| | - Rosalie Aprelon
- INRA-CIRAD, UMR 1309 Contrôle des maladies animales, exotiques et émergentes, F-97170, Petit-Bourg, Guadeloupe, France.
| | - Thierry Lefrançois
- INRA-CIRAD, UMR 1309 Contrôle des maladies animales, exotiques et émergentes, F-34398, Montpellier, France.
| | - Dominique Martinez
- INRA-CIRAD, UMR 1309 Contrôle des maladies animales, exotiques et émergentes, F-34398, Montpellier, France.
| | - Mathieu Epardaud
- INRA-CIRAD, UMR 1309 Contrôle des maladies animales, exotiques et émergentes, F-34398, Montpellier, France. .,INRA, UMR 1282 Infectiologie et Santé Publique, 37380, Nouzilly, France.
| |
Collapse
|
48
|
Posch B, Irsara C, Gamper FS, Herrmann M, Bindreither D, Fuchs D, Reider N, Redl B, Heufler C. Allergenic Can f 1 and its human homologue Lcn-1 direct dendritic cells to induce divergent immune responses. J Cell Mol Med 2015. [PMID: 26218644 PMCID: PMC4594679 DOI: 10.1111/jcmm.12616] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Why and when the immune system skews to Th2 mediated allergic immune responses is still poorly characterized. With two homologous lipocalins, the major respiratory dog allergen Can f 1 and the human endogenous, non-allergenic Lipocalin-1, we investigated their impact on human monocyte-derived dendritic cells (DC). The two lipocalins had differential effects on DC according to their allergenic potential. Compared to Lipocalin-1, Can f 1 persistently induced lower levels of the Th1 skewing maturation marker expression, tryptophan breakdown and interleukin (IL)-12 production in DC. As a consequence, T cells stimulated by DC treated with Can f 1 produced more of the Th2 signature cytokine IL-13 and lower levels of the Th1 signature cytokine interferon-γ than T cells stimulated by Lipocalin-1 treated DC. These data were partially verified by a second pair of homologous lipocalins, the cat allergen Fel d 4 and its putative human homologue major urinary protein. Our data indicate that the crosstalk of DC with lipocalins alone has the potential to direct the type of immune response to these particular antigens. A global gene expression analysis further supported these results and indicated significant differences in intracellular trafficking, sorting and antigen presentation pathways when comparing Can f 1 and Lipocalin-1 stimulated DC. With this study we contribute to a better understanding of the induction phase of a Th2 immune response.
Collapse
Affiliation(s)
- Beate Posch
- Department of Dermatology, Medical University Innsbruck, Innsbruck, Austria
| | - Christian Irsara
- Department of Dermatology, Medical University Innsbruck, Innsbruck, Austria
| | - Fabian S Gamper
- Department of Dermatology, Medical University Innsbruck, Innsbruck, Austria
| | - Martin Herrmann
- Department of Anaesthesiology and Critical Care Medicine, Medical University of Innsbruck, Innsbruck, Austria
| | - Daniel Bindreither
- Division of Molecular Pathophysiology, Biocenter, Medical University Innsbruck, Innsbruck, Austria
| | - Dietmar Fuchs
- Division of Medical Biochemistry, Biocenter, Medical University Innsbruck, Innsbruck, Austria
| | - Norbert Reider
- Department of Dermatology, Medical University Innsbruck, Innsbruck, Austria
| | - Bernhard Redl
- Division of Molecular Biology, Biocenter, Medical University Innsbruck, Innsbruck, Austria
| | - Christine Heufler
- Department of Dermatology, Medical University Innsbruck, Innsbruck, Austria
| |
Collapse
|
49
|
Saito TB, Walker DH. A Tick Vector Transmission Model of Monocytotropic Ehrlichiosis. J Infect Dis 2015; 212:968-77. [PMID: 25737562 DOI: 10.1093/infdis/jiv134] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2014] [Accepted: 02/13/2015] [Indexed: 01/13/2023] Open
Abstract
BACKGROUND Ehrlichioses are emerging, tick-borne diseases distributed worldwide. Previously established animal models use needle inoculation as a mode of infection; however, there is limited representation of natural transmission in artificially inoculated models compared with transmission by the tick vector. The objective of this study was to develop a tick vector transmission animal model of ehrlichial infection using a human pathogen, Ehrlichia muris-like agent (EMLA). METHODS Ixodes scapularis larvae were fed on EMLA-infected mice, and after molting, infected nymphs were used to infest naive animals. RESULTS Ehrlichiae were acquired by 90%-100% of feeding larvae. The majority of animals fed upon by infected nymphs developed sublethal infection with 27% lethality. Bacteria disseminated to all tissues tested with greatest bacterial loads in lungs, but also spleen, lymph nodes, liver, kidneys, brain, and bone marrow. Numerous foci of cellular infiltration, mitoses, and hepatocellular death were observed in liver. Mice infected by tick transmission developed higher antiehrlichial antibody levels than needle-inoculated animals. Tick-feeding-site reactions were observed, but there was no observed difference between animals infested with infected or uninfected ticks. CONCLUSIONS For the first time we were able to develop a tick transmission model with an Ehrlichia that is pathogenic for humans.
Collapse
Affiliation(s)
- Tais Berelli Saito
- Department of Pathology, University of Texas Medical Branch at Galveston
| | - David H Walker
- Department of Pathology, University of Texas Medical Branch at Galveston
| |
Collapse
|
50
|
Ixodes ricinus salivary serpin IRS-2 affects Th17 differentiation via inhibition of the interleukin-6/STAT-3 signaling pathway. Infect Immun 2015; 83:1949-56. [PMID: 25712932 DOI: 10.1128/iai.03065-14] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2014] [Accepted: 02/20/2015] [Indexed: 01/10/2023] Open
Abstract
Th17 cells constitute a subset of CD4(+) T lymphocytes that play a crucial role in protection against extracellular bacteria and fungi. They are also associated with tissue injury in autoimmune and inflammatory diseases. Here, we report that serpin from the tick Ixodes ricinus, IRS-2, inhibits Th17 differentiation by impairment of the interleukin-6 (IL-6)/STAT-3 signaling pathway. Following activation, mature dendritic cells produce an array of cytokines, including the pleiotropic cytokine IL-6, which triggers the IL-6 signaling pathway. The major transcription factor activated by IL-6 is STAT-3. We show that IRS-2 selectively inhibits production of IL-6 in dendritic cells stimulated with Borrelia spirochetes, which leads to attenuated STAT-3 phosphorylation and finally to impaired Th17 differentiation. The results presented extend the knowledge about the effect of tick salivary serpins on innate immunity cells and their function in driving adaptive immune responses.
Collapse
|