1
|
Gonzalez J, Harvey C, Ribeiro-Silva CDS, Leal-Galvan B, Persinger KA, Olafson PU, Johnson TL, Oliva Chavez A. Evaluation of tick salivary and midgut extracellular vesicles as anti-tick vaccines in White-tailed deer (Odocoileus virginianus). Ticks Tick Borne Dis 2025; 16:102420. [PMID: 39667072 DOI: 10.1016/j.ttbdis.2024.102420] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2024] [Revised: 11/14/2024] [Accepted: 11/15/2024] [Indexed: 12/14/2024]
Abstract
Current tick control measures are focused on the use of synthetic acaricides and personal protective measures. However, the emergence of acaricide resistance and the maintenance of tick populations in wildlife has precluded the efficient management of ticks. Thus, host-targeted, non-chemical control measures are needed to reliably reduce ticks parasitizing sylvatic reservoirs. This project aimed to evaluate extracellular vesicles (EVs) from Amblyomma americanum as vaccine candidates for white-tailed deer (Odocoileus virginianus; WTD). Salivary gland (SG) and midgut (MG) EVs were isolated by ultracentrifugation. Three deer were vaccinated with SG and MG EVs and received two boosters at days 28 and 50. Two control deer were injected with adjuvant and PBS only. On day 58, WTD were infested with 100 A. americanum nymphs, 50 females, and 50 males that were allowed to feed to repletion. On-host and off-host mortality, tick engorgement weight, nymph molting, time to oviposition, and egg hatchability were evaluated. Serum samples were recovered every seven days until the last day of tick drop off, and then at one year (Y1) and 1-year and 1-month (Y1M1). Vaccination resulted in seroconversion and significant increases in total IgG levels that remained significantly higher than controls and pre-vaccination levels at Y1 and Y1M1. No negative effects were observed in nymphs, but on-host mortality of female A. americanum was significantly higher in vaccinated animals. No effects were observed on reproductive parameters. These results indicate that proteins within female tick SG and MG vesicles are not good candidates for vaccine design against nymphs; however, the on-host adult mortality suggests that tick EVs harbor protective antigens against A. americanum females.
Collapse
Affiliation(s)
- Julia Gonzalez
- Department of Entomology, Texas A&M University, College Station, TX 77843, USA
| | - Cristina Harvey
- Department of Entomology, Texas A&M University, College Station, TX 77843, USA
| | - Cárita de Souza Ribeiro-Silva
- Departamento de Biociências e Tecnologia, Instituto de Patologia Tropical e Saúde Pública, Universidade Federal de Goiás, Goiânia, GO 74690-900, Brazil
| | - Brenda Leal-Galvan
- Department of Entomology, Texas A&M University, College Station, TX 77843, USA
| | | | - Pia U Olafson
- USDA-ARS, Knipling-Bushland United States Livestock Insects Research Laboratory, Kerrville, TX 78028, USA
| | | | - Adela Oliva Chavez
- Department of Entomology, Texas A&M University, College Station, TX 77843, USA.
| |
Collapse
|
2
|
Dougherty MW, Russart NM, Gaultney RA, Gisi EM, Cooper HM, Kallis LR, Brissette CA, Vaughan JA. The role of southern red-backed voles, Myodes gapperi, and Peromyscus mice in the enzootic maintenance of Lyme disease spirochetes in North Dakota, USA. Ticks Tick Borne Dis 2024; 15:102385. [PMID: 39096783 PMCID: PMC11670892 DOI: 10.1016/j.ttbdis.2024.102385] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Revised: 07/18/2024] [Accepted: 07/22/2024] [Indexed: 08/05/2024]
Abstract
Lyme disease has expanded into the Great Plains of the USA. To investigate local enzootic transmission, small mammals were trapped in two forested tracts in northeastern North Dakota during 2012 and 2013. Peromyscus mice and southern red-backed voles, Myodes gapperi, comprised over 90% of all mammals captured. One site was dominated by Peromyscus (79% of 100 mammals captured). At the other site, M. gapperi (59% of 107 mammals captured) was more abundant than Peromyscus (36%). Immature stages of two tick species parasitized small mammals: Dermacentor variabilis and Ixodes scapularis. Larval I. scapularis ectoparasitism was significantly higher on Peromyscus (81% infested; 3.7 larvae per infested mouse) than M. gapperi (47% infested; 2.6 larvae per infested vole) whereas larval and nymphal D. variabilis ectoparasitism were highest on M. gapperi. Over 45% of infested rodents were concurrently infested with both tick species. Testing engorged I. scapularis larvae from Peromyscus (n = 66) and M. gapperi (n = 20) yielded xenopositivity prevalence for Borrelia burgdorferi sensu lato (s.l.) in these rodents of 6% and 5%, respectively. Progeny of field collected M. gapperi were used to determine host infectivity for a local isolate of B. burgdorferi sensu stricto (s.s.). Five M. gapperi were injected with spirochetes, infested with pathogen-free I. scapularis larvae on days 10, 20, and 40 after infection, and engorged larvae molted to nymphs. Subsamples of nymphs were tested by PCR for B. burgdorferi s. s. DNA and yielded infection rates of 56% (n = 100 nymphs tested), 75% (n = 8) and 64% (n = 31), respectively. The remaining infected nymphs were fed on BALB/c Mus musculus mice and 7 d later, mice were euthanized, and tissues were cultured for B. burgdorferi s.s. Nymphs successfully transmitted spirochetes to 13 of 18 (72%) mice that were exposed to 1-5 infected ticks. Theoretical reservoir potentials - i.e., ability to generate B. burgdorferi infected nymphs - were compared between Peromyscus and M. gapperi. At one site, Peromyscus accounted for nearly all Borrelia-infected nymphs produced (reservoir potential value of 0.935). At the other site, the reservoir potentials for Peromyscus (0.566) and M. gapperi (0.434) were comparable. The difference was attributed to differences in the relative abundance of voles versus mice between sites and the higher level of ectoparasitism by larval I. scapularis on Peromyscus versus M. gapperi at both sites. The southern red-backed vole, M. gapperi, contributes to the enzootic maintenance of Lyme disease spirochetes in North Dakota and possibly other areas where this rodent species is abundant.
Collapse
Affiliation(s)
- Michael W Dougherty
- Department of Biology, University of North Dakota, Grand Forks, ND, United States; Department of Medicine, University of Florida College of Medicine, University of Florida, Gainesville, FL, United States
| | - Nathan M Russart
- Department of Biology, University of North Dakota, Grand Forks, ND, United States; Aldeveron, Fargo, ND, United States
| | - Robert A Gaultney
- Department of Biomedical Sciences, North Dakota School of Medicine and Health Sciences, University of North Dakota, Grand Forks, ND, United States; Institute of Tissue Medicine and Pathology, University of Bern, Bern, Switzerland
| | - Emily M Gisi
- Department of Biology, University of North Dakota, Grand Forks, ND, United States
| | - Haley M Cooper
- Department of Biology, University of North Dakota, Grand Forks, ND, United States
| | - Lindsey R Kallis
- Department of Biology, University of North Dakota, Grand Forks, ND, United States
| | - Catherine A Brissette
- Department of Biomedical Sciences, North Dakota School of Medicine and Health Sciences, University of North Dakota, Grand Forks, ND, United States
| | - Jefferson A Vaughan
- Department of Biology, University of North Dakota, Grand Forks, ND, United States.
| |
Collapse
|
3
|
Bourgeois JS, Hu LT. Hitchhiker's Guide to Borrelia burgdorferi. J Bacteriol 2024; 206:e0011624. [PMID: 39140751 PMCID: PMC11411949 DOI: 10.1128/jb.00116-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/15/2024] Open
Abstract
Don't Panic. In the nearly 50 years since the discovery of Lyme disease, Borrelia burgdorferi has emerged as an unlikely workhorse of microbiology. Interest in studying host-pathogen interactions fueled significant progress in making the fastidious microbe approachable in laboratory settings, including the development of culture methods, animal models, and genetic tools. By developing these systems, insight has been gained into how the microbe is able to survive its enzootic cycle and cause human disease. Here, we discuss the discovery of B. burgdorferi and its development as a model organism before diving into the critical lessons we have learned about B. burgdorferi biology at pivotal stages of its lifecycle: gene expression changes during the tick blood meal, colonization of a new vertebrate host, and developing a long-lasting infection in that vertebrate until a new tick feeds. Our goal is to highlight the advancements that have facilitated B. burgdorferi research and identify gaps in our current understanding of the microbe.
Collapse
Affiliation(s)
- Jeffrey S. Bourgeois
- Department of Molecular Biology and Microbiology, Tufts University Lyme Disease Initiative, Tufts University School of Medicine, Boston, Massachusetts, USA
| | - Linden T. Hu
- Department of Molecular Biology and Microbiology, Tufts University Lyme Disease Initiative, Tufts University School of Medicine, Boston, Massachusetts, USA
| |
Collapse
|
4
|
Narasimhan S, Cibichakravarthy B, Wu MJ, Holter MM, Walsh CA, Goodrich JA. Laboratory Management of Mammalian Hosts for Ixodes scapularis -Host-Pathogen Interaction Studies. Comp Med 2024; 74:235-245. [PMID: 39289828 PMCID: PMC11373684 DOI: 10.30802/aalas-cm-24-036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Revised: 05/19/2024] [Accepted: 07/10/2024] [Indexed: 09/19/2024]
Abstract
Due to their hematophagous life cycle, hard-bodied ticks including the genus Ixodes are a potential vector for numerous pathogenic organisms including bacteria, protozoa, viruses, and infectious prions. The natural geographic range of several hard tick species, includig Ixodes scapularis, has expanded over recent decades. Consequently, there is an ongoing need to maintain, feed, and propagate ticks for host-pathogen interaction studies to better understand and mitigate their impact on human and animal health. Artificial membrane feeding of hard ticks has advanced in recent years, has study design advantages, and should be used, when possible, to reduce animal use, but it also has several limitations that require the continued use of mammalian hosts including mice, guinea pigs, and rabbits. In this overview, we discuss the best management practices for these relevant species with respect to biosafety, health, and optimal host comfort when used in studies that depend on tick feeding. The capsule-jacket method is preferred over the ear sock-E-collar method of tick feeding on rabbit hosts because of better host health, comfort, and increased study versatility.
Collapse
Affiliation(s)
- Sukanya Narasimhan
- Section of Infectious Diseases, Department of Internal Medicine, Yale School of Medicine, New Haven, Connecticut; and
| | | | - Ming-Jie Wu
- Section of Infectious Diseases, Department of Internal Medicine, Yale School of Medicine, New Haven, Connecticut; and
| | - Marlena M Holter
- Department of Comparative Medicine, Yale School of Medicine, New Haven, Connecticut
| | - Courtney A Walsh
- Department of Comparative Medicine, Yale School of Medicine, New Haven, Connecticut
| | - James A Goodrich
- Department of Comparative Medicine, Yale School of Medicine, New Haven, Connecticut
| |
Collapse
|
5
|
de Sousa-Paula LC, Berger M, Talyuli OAC, Schwartz CL, Saturday GA, Ribeiro JMC, Tirloni L. Exploring the transcriptome of immature stages of Ornithodoros hermsi, the soft-tick vector of tick-borne relapsing fever. Sci Rep 2024; 14:12466. [PMID: 38816418 PMCID: PMC11140000 DOI: 10.1038/s41598-024-62732-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2024] [Accepted: 05/21/2024] [Indexed: 06/01/2024] Open
Abstract
Blood-feeding behavior has independently evolved in arthropods multiple times. Unlike hard ticks, soft ticks employ a rapid-feeding strategy for hematophagy, and there are comparatively limited studies on the transcriptomes of these organisms. This study investigates the soft tick Ornithodoros hermsi, conducting histopathological examinations at bitten skin sites and tick whole-body transcriptomic analyses across various developmental and feeding stages, including larvae, 1st-nymphal, and 2nd-nymphal stages. The results revealed the ability of O. hermsi to induce skin hemorrhage at the bite sites. Transcriptomic analyses identified three consistent transcriptional profiles: unfed, early-fed (6 h, 12 h, 24 h), and late-fed (5 days). The unfed profile exhibited high transcriptional activity across most of the functional classes annotated. In contrast, early-fed stages exhibited decreased expression of most functional classes, except for the unknown, which is highly expressed. Finally, transcriptional expression of most functional classes increased in the late-fed groups, resembling the baseline expression observed in the unfed groups. These findings highlight intense pre-feeding transcriptional activity in O. hermsi ticks, aligning with their rapid-feeding strategy. Moreover, besides shedding light on the temporal dynamics of key pathways during blood meal processing and tick development, this study contributes significantly to the transcriptome repertoire of a medically relevant soft tick species with relatively limited prior knowledge.
Collapse
Affiliation(s)
- Lucas C de Sousa-Paula
- Tick-Pathogen Transmission Unit, Laboratory of Bacteriology, Rocky Mountain Laboratories, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, Hamilton, MT, USA
| | - Markus Berger
- Tick-Pathogen Transmission Unit, Laboratory of Bacteriology, Rocky Mountain Laboratories, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, Hamilton, MT, USA
- Grupo de Reprodução e Farmacologia Celular, Laboratório de Bioquímica Farmacológica, Centro de Pesquisa Experimental (CPE), Hospital de Clínicas de Porto Alegre (HCPA-UFRGS), Porto Alegre, RS, Brazil
| | - Octavio A C Talyuli
- Mosquito Immunity and Vector Competence Section, Laboratory of Malaria and Vector Research, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, Rockville, MD, USA
| | - Cindi L Schwartz
- Electron Microscopy Unit, Research Technologies Branch, Rocky Mountain Laboratories, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, Hamilton, MT, USA
| | - Greg A Saturday
- Rocky Mountain Veterinary Branch, Rocky Mountain Laboratories, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, Hamilton, MT, USA
| | - José M C Ribeiro
- Vector Biology Section, Laboratory of Malaria and Vector Research, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, Rockville, MD, USA
| | - Lucas Tirloni
- Tick-Pathogen Transmission Unit, Laboratory of Bacteriology, Rocky Mountain Laboratories, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, Hamilton, MT, USA.
| |
Collapse
|
6
|
Bourgeois JS, McCarthy JE, Turk SP, Bernard Q, Clendenen LH, Wormser GP, Marcos LA, Dardick K, Telford SR, Marques AR, Hu LT. Peromyscus leucopus , Mus musculus , and humans have distinct transcriptomic responses to larval Ixodes scapularis bites. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.02.592193. [PMID: 38746284 PMCID: PMC11092580 DOI: 10.1101/2024.05.02.592193] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/16/2024]
Abstract
Ixodes scapularis ticks are an important vector for at least six tick-borne human pathogens, including the predominant North American Lyme disease spirochete Borrelia burgdorferi . The ability for these ticks to survive in nature is credited, in part, to their ability to feed on a variety of hosts without excessive activation of the proinflammatory branch of the vertebrate immune system. While the ability for nymphal ticks to feed on a variety of hosts has been well-documented, the host-parasite interactions between larval I. scapularis and different vertebrate hosts is relatively unexplored. Here we report on the changes in the vertebrate transcriptome present at the larval tick bite site using the natural I. scapularis host Peromyscus leucopus deermouse, a non-natural rodent host Mus musculus (BALB/c), and humans. We note substantially less evidence of activation of canonical proinflammatory pathways in P. leucopus compared to BALB/c mice and pronounced evidence of inflammation in humans. Pathway enrichment analyses revealed a particularly strong signature of interferon gamma, tumor necrosis factor, and interleukin 1 signaling at the BALB/c and human tick bite site. We also note that bite sites on BALB/c mice and humans, but not deermice, show activation of wound-healing pathways. These data provide molecular evidence of the coevolution between larval I. scapularis and P. leucopus as well as expand our overall understanding of I. scapularis feeding. Significance Ixodes scapularis tick bites expose humans to numerous diseases in North America. While larval tick feeding enables pathogens to enter the tick population and eventually spread to humans, how larval ticks interact with mammals has been understudied compared to other tick stages. Here we examined the transcriptomic response of a natural I. scapularis rodent host ( Peromyscus leucopus ), a non-native I. scapularis rodent host ( Mus musculus ), and an incidental host (humans). We find that there are differences in how all three species respond to larval I. scapularis , with the natural host producing the smallest transcriptomic signature of a canonical proinflammatory immune response and the incidental human host producing the most robust signature of inflammation in response to the larval tick. These data expand our understanding of the pressures on ticks in the wild and inform our ability to model these interactions in laboratory settings.
Collapse
|
7
|
Hils M, Hoffard N, Iuliano C, Kreft L, Chakrapani N, Swiontek K, Fischer K, Eberlein B, Köberle M, Fischer J, Hilger C, Ohnmacht C, Kaesler S, Wölbing F, Biedermann T. IgE and anaphylaxis specific to the carbohydrate alpha-gal depend on IL-4. J Allergy Clin Immunol 2024; 153:1050-1062.e6. [PMID: 38135009 PMCID: PMC10997276 DOI: 10.1016/j.jaci.2023.12.003] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Revised: 11/23/2023] [Accepted: 12/01/2023] [Indexed: 12/24/2023]
Abstract
BACKGROUND Alpha-gal (Galα1-3Galβ1-4GlcNAc) is a carbohydrate with the potential to elicit fatal allergic reactions to mammalian meat and drugs of mammalian origin. This type of allergy is induced by tick bites, and therapeutic options for this skin-driven food allergy are limited to the avoidance of the allergen and treatment of symptoms. Thus, a better understanding of the immune mechanisms resulting in sensitization through the skin is crucial, especially in the case of a carbohydrate allergen for which underlying immune responses are poorly understood. OBJECTIVE We aimed to establish a mouse model of alpha-gal allergy for in-depth immunologic analyses. METHODS Alpha-galactosyltransferase 1-deficient mice devoid of alpha-gal glycosylations were sensitized with the alpha-gal-carrying self-protein mouse serum albumin by repetitive intracutaneous injections in combination with the adjuvant aluminum hydroxide. The role of basophils and IL-4 in sensitization was investigated by antibody-mediated depletion. RESULTS Alpha-gal-sensitized mice displayed increased levels of alpha-gal-specific IgE and IgG1 and developed systemic anaphylaxis on challenge with both alpha-gal-containing glycoproteins and glycolipids. In accordance with alpha-gal-allergic patients, we detected elevated numbers of basophils at the site of sensitization as well as increased numbers of alpha-gal-specific B cells, germinal center B cells, and B cells of IgE and IgG1 isotypes in skin-draining lymph nodes. By depleting IL-4 during sensitization, we demonstrated for the first time that sensitization and elicitation of allergy to alpha-gal and correspondingly to a carbohydrate allergen is dependent on IL-4. CONCLUSION These findings establish IL-4 as a potential target to interfere with alpha-gal allergy elicited by tick bites.
Collapse
Affiliation(s)
- Miriam Hils
- Department of Dermatology and Allergy Biederstein, School of Medicine, Technical University of Munich, Munich, Germany
| | - Nils Hoffard
- Department of Dermatology and Allergy Biederstein, School of Medicine, Technical University of Munich, Munich, Germany
| | - Caterina Iuliano
- Department of Dermatology and Allergy Biederstein, School of Medicine, Technical University of Munich, Munich, Germany
| | - Luisa Kreft
- Center of Allergy and Environment (ZAUM) and Institute of Allergy Research, Technical University of Munich, School of Medicine, and Helmholtz Center Munich, Research Center for Environmental Health, Neuherberg, Germany
| | - Neera Chakrapani
- Department of Infection and Immunity, Luxembourg Institute of Health (LIH), Esch-sur-Alzette, Luxembourg; Faculty of Science, Technology and Medicine, University of Luxembourg, Esch-sur-Alzette, Luxembourg
| | - Kyra Swiontek
- Department of Infection and Immunity, Luxembourg Institute of Health (LIH), Esch-sur-Alzette, Luxembourg
| | - Konrad Fischer
- Department of Livestock Biotechnology, School of Life Sciences Weihenstephan, Technical University of Munich, Freising, Germany
| | - Bernadette Eberlein
- Department of Dermatology and Allergy Biederstein, School of Medicine, Technical University of Munich, Munich, Germany
| | - Martin Köberle
- Department of Dermatology and Allergy Biederstein, School of Medicine, Technical University of Munich, Munich, Germany
| | - Jörg Fischer
- Department of Dermatology, Faculty of Medicine, Eberhard Karls University Tübingen, Tübingen, Germany; Department of Dermatology and Allergology, University Hospital Augsburg, Augsburg, Germany
| | - Christiane Hilger
- Department of Infection and Immunity, Luxembourg Institute of Health (LIH), Esch-sur-Alzette, Luxembourg
| | - Caspar Ohnmacht
- Center of Allergy and Environment (ZAUM) and Institute of Allergy Research, Technical University of Munich, School of Medicine, and Helmholtz Center Munich, Research Center for Environmental Health, Neuherberg, Germany
| | - Susanne Kaesler
- Department of Dermatology and Allergy Biederstein, School of Medicine, Technical University of Munich, Munich, Germany
| | - Florian Wölbing
- Department of Dermatology and Allergy Biederstein, School of Medicine, Technical University of Munich, Munich, Germany
| | - Tilo Biedermann
- Department of Dermatology and Allergy Biederstein, School of Medicine, Technical University of Munich, Munich, Germany.
| |
Collapse
|
8
|
Brown JE, Tiffin HS, Pagac A, Poh KC, Evans JR, Miller TM, Herrin BH, Tomlinson T, Sutherland C, Machtinger ET. Differential burdens of blacklegged ticks ( Ixodes scapularis) on sympatric rodent hosts. JOURNAL OF VECTOR ECOLOGY : JOURNAL OF THE SOCIETY FOR VECTOR ECOLOGY 2023; 49:44-52. [PMID: 38147300 DOI: 10.52707/1081-1710-49.1.44] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/14/2023] [Accepted: 10/05/2023] [Indexed: 12/27/2023]
Abstract
In the United States, there has been a steady increase in diagnosed cases of tick-borne diseases in people, most notably Lyme disease. The pathogen that causes Lyme disease, Borrelia burgdorferi, is transmitted by the blacklegged tick (Ixodes scapularis). Several small mammals are considered key reservoirs of this pathogen and are frequently-used hosts by blacklegged ticks. However, limited studies have evaluated between-species host use by ticks. This study compared I. scapularis burdens and tick-associated pathogen presence in wild-caught Clethrionomys gapperi (southern red-backed voles) and Peromyscus spp. (white-footed mice) in forested areas where the habitat of both species overlapped. Rodent trapping data collected over two summers showed a significant difference in the average tick burden between species. Adult Peromyscus spp. had an overall mean of 4.03 ticks per capture, while adult C. gapperi had a mean of 0.47 ticks per capture. There was a significant association between B. burgdorferi infection and host species with more Peromyscus spp. positive samples than C. gapperi (65.8% and 10.2%, respectively). This work confirms significant differences in tick-host use and pathogen presence between sympatric rodent species. It is critical to understand tick-host interactions and tick distributions to develop effective and efficient tick control methods.
Collapse
Affiliation(s)
- Jessica E Brown
- Department of Entomology, Pennsylvania State University, University Park, PA 16802, U.S.A.,
| | - Hannah S Tiffin
- Department of Entomology, Pennsylvania State University, University Park, PA 16802, U.S.A
| | - Alexandra Pagac
- Department of Entomology, Pennsylvania State University, University Park, PA 16802, U.S.A
| | - Karen C Poh
- Department of Entomology, Pennsylvania State University, University Park, PA 16802, U.S.A
| | - Jesse R Evans
- Department of Entomology, Pennsylvania State University, University Park, PA 16802, U.S.A
| | - Taylor M Miller
- Department of Entomology, Pennsylvania State University, University Park, PA 16802, U.S.A
| | - Brian H Herrin
- Department of Diagnostic Medicine and Pathobiology, Kansas State University, Manhattan, KS 66506, U.S.A
| | - Trey Tomlinson
- Department of Diagnostic Medicine and Pathobiology, Kansas State University, Manhattan, KS 66506, U.S.A
| | - Cameron Sutherland
- Department of Diagnostic Medicine and Pathobiology, Kansas State University, Manhattan, KS 66506, U.S.A
| | - Erika T Machtinger
- Department of Entomology, Pennsylvania State University, University Park, PA 16802, U.S.A
| |
Collapse
|
9
|
Karim S, Leyva-Castillo JM, Narasimhan S. Tick salivary glycans - a sugar-coated tick bite. Trends Parasitol 2023; 39:1100-1113. [PMID: 37838514 DOI: 10.1016/j.pt.2023.09.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2023] [Revised: 09/22/2023] [Accepted: 09/22/2023] [Indexed: 10/16/2023]
Abstract
Ticks are hematophagous arthropods that transmit disease-causing pathogens worldwide. Tick saliva deposited into the tick-bite site is composed of an array of immunomodulatory proteins that ensure successful feeding and pathogen transmission. These salivary proteins are often glycosylated, and glycosylation is potentially critical for the function of these proteins. Some salivary glycans are linked to the phenomenon of red meat allergy - an allergic response to red meat consumption in humans exposed to certain tick species. Tick salivary glycans are also invoked in the phenomenon of acquired tick resistance wherein non-natural host species exposed to tick bites develop an immune response that thwarts subsequent tick feeding. This review dwells on our current knowledge of these two phenomena, thematically linked by salivary glycans.
Collapse
Affiliation(s)
- Shahid Karim
- University of Southern Mississippi, Hattiesburg, MS, USA
| | - Juan Manuel Leyva-Castillo
- Division of Immunology, Boston Children's Hospital, Department of Pediatrics, Harvard Medical School, Boston, MA, USA
| | - Sukanya Narasimhan
- Section of Infectious Diseases, Department of Internal Medicine, Yale School of Medicine, New Haven-06520, CT, USA.
| |
Collapse
|
10
|
Blubaugh CK, Jones CR, Josefson C, Scoles GA, Snyder WE, Owen JP. Omnivore diet composition alters parasite resistance and host condition. J Anim Ecol 2023; 92:2175-2188. [PMID: 37732627 DOI: 10.1111/1365-2656.14004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2022] [Accepted: 08/25/2023] [Indexed: 09/22/2023]
Abstract
Diet composition modulates animals' ability to resist parasites and recover from stress. Broader diet breadths enable omnivores to mount dynamic responses to parasite attack, but little is known about how plant/prey mixing might influence responses to infection. Using omnivorous deer mice (Peromyscus maniculatus) as a model, we examine how varying plant and prey concentrations in blended diets influence resistance and body condition following infestation by Rocky Mountain wood ticks (Dermacentor andersoni). In two repeated experiments, deer mice fed for 4 weeks on controlled diets that varied in proportions of seeds and insects were then challenged with 50 tick larvae in two sequential infestations. The numbers of ticks successfully feeding on mice declined by 25% and 66% after the first infestation (in the first and second experiments, respectively), reflecting a pattern of acquired resistance, and resistance was strongest when plant/prey ratios were more equally balanced in mouse diets, relative to seed-dominated diets. Diet also dramatically impacted the capacity of mice to cope with tick infestations. Mice fed insect-rich diets lost 15% of their body weight when parasitized by ticks, while mice fed seed-rich diets lost no weight at all. While mounting/maintaining an immune response may be energetically demanding, mice may compensate for parasitism with fat and carbohydrate-rich diets. Altogether, these results suggest that a diverse nutritional landscape may be key in enabling omnivores' resistance and resilience to infection and immune stressors in their environments.
Collapse
Affiliation(s)
- Carmen K Blubaugh
- Department of Crop Sciences, University of Illinois Urbana-Champaign, Urbana, Illinois, USA
| | - Cami R Jones
- Department of Entomology, Washington State University, Pullman, Washington, USA
| | - Chloe Josefson
- Department of Animal, Veterinary and Food Sciences, University of Idaho, Moscow, Idaho, USA
| | - Glen A Scoles
- Invasive Insect Biocontrol & Behavior Laboratory, USDA-ARS, Beltsville, Maryland, USA
| | - William E Snyder
- Department of Crop Sciences, University of Illinois Urbana-Champaign, Urbana, Illinois, USA
| | - Jeb P Owen
- Department of Entomology, Washington State University, Pullman, Washington, USA
| |
Collapse
|
11
|
Obaid MK, Almutairi MM, Alouffi A, Safi SZ, Tanaka T, Ali A. Assessment of cypermethrin and amitraz resistance and molecular profiling of voltage-gated sodium channel and octopamine tyramine genes of Rhipicephalus microplus. Front Cell Infect Microbiol 2023; 13:1176013. [PMID: 37305408 PMCID: PMC10248163 DOI: 10.3389/fcimb.2023.1176013] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2023] [Accepted: 04/14/2023] [Indexed: 06/13/2023] Open
Abstract
Control of ticks and tick-borne pathogens is a priority for human and animal health. Livestock-holders extensively rely on acaricide applications for tick control. Different groups of acaricides including cypermethrin and amitraz have been consistently used in Pakistan. There has been a gap in understanding the susceptibility or resistance of Rhipicephalus microplus, the most prevalent tick in Pakistan, to acaricides. The present study aimed to molecularly characterize cypermethrin and amitraz targeted genes such as voltage-gated sodium channel (VGSC) and octopamine tyramine (OCT/Tyr) of R. microplus ticks in Khyber Pakhtunkhwa (KP), Pakistan to monitor the acaricides resistance. Tick specimens were collected from cattle and buffaloes in northern (Chitral, Shangla, Swat, Dir, and Buner), central (Peshawar, Mardan, Charsadda, Swabi, and Nowshera), and southern districts (Kohat, Karak, Lakki Marwat, Tank, and Dera Ismail Khan) of KP, Pakistan. Different concentrations of commercially available cypermethrin (10%) and amitraz (12.5%) were prepared for in vitro larval immersion tests (LIT). In LIT, the average mortality rate of immersed larvae was recorded that was increased gradually with an increase in the concentration of specific acaricide. The larvae's highest mortality rates (94.5% and 79.5%) were observed at 100-ppm of cypermethrin and amitraz, respectively. A subset of 82 R. microplus ticks was subjected to extract genomic DNA, followed by PCR to amplify partial fragments of VGSC (domain-II) and OCT/Tyr genes. The BLAST results of the consensus sequence of VGSC gene (domain-II) showed 100% identity with the acaricides susceptible tick sequence from the United States (reference sequence). Obtained identical sequences of OCT/Tyr genes showed maximum identity (94-100%) with the identical sequences reported from Australia (reference sequence), India, Brazil, Philippines, USA, South Africa, and China. Thirteen single nucleotide polymorphisms (10 synonymous and three non-synonymous) were observed at various positions of partial OCT/Tyr gene fragments. The SNP at position A-22-C (T-8-P) in OCT/Tyr gene has been linked to amitraz resistance in R. microplus ticks. Molecular analysis and LIT bioassay's findings indicate the availability of resistant R. microplus ticks in the KP region. To our understanding, this is the first preliminary study to monitor cypermethrin and amitraz resistance via molecular profiling of cypermethrin and amitraz targeted genes (VGSC and OCT/Tyr) in combination with in vitro bioassays (LIT) in R. microplus ticks from Pakistan.
Collapse
Affiliation(s)
| | - Mashal M. Almutairi
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Abdulaziz Alouffi
- King Abdulaziz City for Science and Technology, Riyadh, Saudi Arabia
| | - Sher Zaman Safi
- Faculty of Medicine, Bioscience and Nursing, MAHSA University, Jenjarom, Selangor, Malaysia
| | - Tetsuya Tanaka
- Laboratory of Infectious Diseases, Joint Faculty of Veterinary Medicine, Kagoshima University, Kagoshima, Japan
| | - Abid Ali
- Department of Zoology, Abdul Wali Khan University Mardan, Mardan, Pakistan
| |
Collapse
|
12
|
Ribeiro JMC, Bayona-Vásquez NJ, Budachetri K, Kumar D, Frederick JC, Tahir F, Faircloth BC, Glenn TC, Karim S. A draft of the genome of the Gulf Coast tick, Amblyomma maculatum. Ticks Tick Borne Dis 2023; 14:102090. [PMID: 36446165 PMCID: PMC9898150 DOI: 10.1016/j.ttbdis.2022.102090] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2022] [Revised: 10/17/2022] [Accepted: 11/20/2022] [Indexed: 11/24/2022]
Abstract
The Gulf Coast tick, Amblyomma maculatum, inhabits the Southeastern states of the USA bordering the Gulf of Mexico, Mexico, and other Central and South American countries. More recently, its U.S. range has extended West to Arizona and Northeast to New York state and Connecticut. It is a vector of Rickettsia parkeri and Hepatozoon americanum. This tick species has become a model to study tick/Rickettsia interactions. To increase our knowledge of the basic biology of A. maculatum we report here a draft genome of this tick and an extensive functional classification of its proteome. The DNA from a single male tick was used as a genomic source, and a 10X genomics protocol determined 28,460 scaffolds having equal or more than 10 Kb, totaling 1.98 Gb. The N50 scaffold size was 19,849 Kb. The BRAKER pipeline was used to find the protein-coding gene boundaries on the assembled A. maculatum genome, discovering 237,921 CDS. After trimming and classifying the transposable elements, bacterial contaminants, and truncated genes, a set of 25,702 were annotated and classified as the core gene products. A BUSCO analysis revealed 83.4% complete BUSCOs. A hyperlinked spreadsheet is provided, allowing browsing of the individual gene products and their matches to several databases.
Collapse
Affiliation(s)
- Jose M C Ribeiro
- NIAID NIH Laboratory of Malaria and Vector Research, Bethesda, MD 20892-8132, USA.
| | - Natalia J Bayona-Vásquez
- Department of Environmental Health Science and Georgia Genomics Facility, Environmental Health Science Building, University of Georgia, Athens, GA 30602, USA
| | - Khemraj Budachetri
- Center for Molecular and Cellular Biology, School of Biological, Environmental, and Earth Sciences, 118 College Drive, 5018, University of Southern Mississippi, Hattiesburg, MS 39406, USA; The Ohio State University, Columbus, OH 43210, USA
| | - Deepak Kumar
- Department of Environmental Health Science and Georgia Genomics Facility, Environmental Health Science Building, University of Georgia, Athens, GA 30602, USA
| | - Julia Catherine Frederick
- Department of Environmental Health Science and Georgia Genomics Facility, Environmental Health Science Building, University of Georgia, Athens, GA 30602, USA
| | - Faizan Tahir
- Center for Molecular and Cellular Biology, School of Biological, Environmental, and Earth Sciences, 118 College Drive, 5018, University of Southern Mississippi, Hattiesburg, MS 39406, USA
| | - Brant C Faircloth
- Department of Biological Sciences, Louisiana State University, 202 Life Sciences Building, Baton Rouge, LA 70803, USA
| | - Travis C Glenn
- Department of Environmental Health Science and Georgia Genomics Facility, Environmental Health Science Building, University of Georgia, Athens, GA 30602, USA
| | - Shahid Karim
- Center for Molecular and Cellular Biology, School of Biological, Environmental, and Earth Sciences, 118 College Drive, 5018, University of Southern Mississippi, Hattiesburg, MS 39406, USA
| |
Collapse
|
13
|
Narasimhan S, Booth CJ, Philipp MT, Fikrig E, Embers ME. Repeated Tick Infestations Impair Borrelia burgdorferi Transmission in a Non-Human Primate Model of Tick Feeding. Pathogens 2023; 12:132. [PMID: 36678479 PMCID: PMC9861725 DOI: 10.3390/pathogens12010132] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Revised: 01/03/2023] [Accepted: 01/10/2023] [Indexed: 01/15/2023] Open
Abstract
The blacklegged tick, Ixodes scapularis, is the predominant vector of Borrelia burgdorferi, the agent of Lyme disease in the USA. Natural hosts of I. scapularis such as Peromyscus leucopus are repeatedly infested by these ticks without acquiring tick resistance. However, upon repeated tick infestations, non-natural hosts such as guinea pigs, mount a robust immune response against critical tick salivary antigens and acquire tick resistance able to thwart tick feeding and Borrelia burgdorferi transmission. The salivary targets of acquired tick resistance could serve as vaccine targets to prevent tick feeding and the tick transmission of human pathogens. Currently, there is no animal model able to demonstrate both tick resistance and diverse clinical manifestations of Lyme disease. Non-human primates serve as robust models of human Lyme disease. By evaluating the responses to repeated tick infestation, this animal model could accelerate our ability to define the tick salivary targets of acquired resistance that may serve as vaccines to prevent the tick transmission of human pathogens. Towards this goal, we assessed the development of acquired tick resistance in non-human primates upon repeated tick infestations. We report that following repeated tick infestations, non-human primates do not develop the hallmarks of acquired tick resistance observed in guinea pigs. However, repeated tick infestations elicit immune responses able to impair the tick transmission of B. burgdorferi. A mechanistic understanding of the protective immune responses will provide insights into B. burgdorferi-tick-host interactions and additionally contribute to anti-tick vaccine discovery.
Collapse
Affiliation(s)
- Sukanya Narasimhan
- Section of Infectious Diseases, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT 06510, USA
| | - Carmen J. Booth
- Department of Comparative Medicine, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Mario T. Philipp
- Division of Bacteriology & Parasitology, Tulane School of Medicine, New Orleans, LA 70112, USA
| | - Erol Fikrig
- Section of Infectious Diseases, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT 06510, USA
| | - Monica E. Embers
- Division of Bacteriology & Parasitology, Tulane School of Medicine, New Orleans, LA 70112, USA
| |
Collapse
|
14
|
Trimèche M, Boussoffara T, Chelbi I, Cherni S, Zhioua S, Msallem N, Labidi I, Zhioua E. Effects of multiple feedings on sensitized rabbits on the fitness of Phlebotomus papatasi (Diptera: Psychodidae). Acta Trop 2022; 228:106303. [PMID: 35021103 DOI: 10.1016/j.actatropica.2022.106303] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2021] [Revised: 12/18/2021] [Accepted: 01/03/2022] [Indexed: 11/01/2022]
Abstract
In the present study, we evaluated the effects of antibodies anti-sandfly saliva on the fecundity of Phlebotomus papatasi, vector of zoonotic cutaneous leishmaniasis in the Old World. Rabbits were repeatedly exposed to sandfly bites. Immune sera showed increased levels of anti-sandfly saliva antibody compared to the pre-exposition period. The analysis of biological parameters revealed no decline on the feeding success of females P. papatasi fed on rabbits repeatedly exposed to sandfly bites. Our results showed that anti-sandfly saliva antibodies of rabbits are not detrimental to the fitness of females P. papatasi. Thus, rabbits did not acquire resistance to sandflies following repeated exposures, and that contribute in maintaining a high density of P. papatasi. To control sandfly infestations and Leishmania transmission, more studies are needed for a better understanding of the mechanisms governing the resistance of hosts to bites of sandflies.
Collapse
|
15
|
Fellin E, Schulte-Hostedde A. Tick infestation effects on haemoglobin levels of deer mice ( Peromyscus maniculatus). Parasitology 2022; 149:209-217. [PMID: 35234604 PMCID: PMC11010517 DOI: 10.1017/s0031182021001700] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2021] [Revised: 09/20/2021] [Accepted: 09/25/2021] [Indexed: 11/06/2022]
Abstract
Deer mice (Peromyscus maniculatus) are hosts to ixodid ticks as well as the associated tick-borne pathogens they can spread. As the ranges of black-legged ticks (Ixodes scapularis) and American dog ticks (Dermacentor variabilis) expand northwards, naïve host populations of deer mice are likely to become infested by ticks and experience the physiological effects that ticks can have on them via blood-feeding. The prevalence of these haematophagous ticks can affect the haemoglobin levels of the mice they infest. Haemoglobin levels were compared and analysed in deer mice populations at three different sites with varying tick exposure. These results suggested that without confounding effects, the abundance of black-legged and American dog ticks on individual mice had a significant negative effect on the hosts' haemoglobin levels, but only in an area with high tick infestation. This was seen across the average haemoglobin levels between populations, where there was a significant difference between the source population with the longest established tick populations and the source population where neither black-legged nor American dog ticks were prevalent. As the ticks' ranges expand and they become more abundant, it is important to understand how their prevalence and intensity can alter host physiology, potentially affecting their own range expansion and the spread of the diseases they may carry.
Collapse
Affiliation(s)
- Erica Fellin
- Department of Biology, Laurentian University, Sudbury, ON, Canada
| | | |
Collapse
|
16
|
Agwunobi DO, Wang N, Huang L, Zhang Y, Chang G, Wang K, Li M, Wang H, Liu J. Phosphoproteomic Analysis of Haemaphysalis longicornis Saliva Reveals the Influential Contributions of Phosphoproteins to Blood-Feeding Success. Front Cell Infect Microbiol 2022; 11:769026. [PMID: 35118006 PMCID: PMC8804221 DOI: 10.3389/fcimb.2021.769026] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2021] [Accepted: 12/23/2021] [Indexed: 11/13/2022] Open
Abstract
Tick saliva, an essential chemical secretion of the tick salivary gland, is indispensable for tick survival owing to the physiological influence it exerts on the host defence mechanisms via the instrumentality of its cocktail of pharmacologically active molecules (proteins and peptides). Much research about tick salivary proteome has been performed, but how most of the individual salivary proteins are utilized by ticks to facilitate blood acquisition and pathogen transmission is not yet fully understood. In addition, the phosphorylation of some proteins plays a decisive role in their function. However, due to the low phosphorylation level of protein, especially for a small amount of protein, it is more difficult to study phosphorylation. Maybe, for this reason, the scarcity of works on the phosphorylated tick salivary proteomes still abound. Here, we performed a phosphoproteomic analysis of Haemaphysalis longicornis tick saliva via TiO2 enrichment and the most advanced Thermo Fisher Orbitrap Exploris 480 mass spectrometer for identification. A total of 262 phosphorylated tick saliva proteins were identified and were subjected to functional annotation/enrichment analysis. Cellular and metabolic process terms accounted for the largest proportion of the saliva proteins, with the participation of these proteins in vital intracellular and extracellular transport-oriented processes such as vesicle-mediated transport, exocytic process, cell adhesion, and movement of cell/subcellular component. “Endocytosis”, “Protein processing in endoplasmic reticulum”, and “Purine metabolism” were the most significantly enriched pathways. The knockdown (RNAi) of Tudor domain-containing protein (TCP), actin-depolymerizing factors (ADF), programmed cell death protein (PD), and serine/threonine-protein kinase (SPK) resulted in the dissociation of collagen fibers and the pilosebaceous unit, increased inflammatory infiltrates/granulocytes (possibly heterophiles), and the depletion of the epithelium. Ticks injected with SPK dsRNA engorged normally but with a change in skin colour (possibly an autoimmune reaction) and the failure to produce eggs pointing to a possible role of SPK in reproduction and host immune modulation. Ticks injected with ADF dsRNA failed to acquire blood, underscoring the role of ADF in facilitating tick feeding. The results of this study showed the presence of phosphorylation in tick saliva and highlight the roles of salivary phosphoproteins in facilitating tick feeding.
Collapse
Affiliation(s)
- Desmond O. Agwunobi
- Ministry of Education Key Laboratory of Molecular and Cellular Biology, Hebei Key Laboratory of Animal Physiology, Biochemistry and Molecular Biology, College of Life Sciences, Hebei Normal University, Shijiazhuang, China
| | - Ningmei Wang
- Ministry of Education Key Laboratory of Molecular and Cellular Biology, Hebei Key Laboratory of Animal Physiology, Biochemistry and Molecular Biology, College of Life Sciences, Hebei Normal University, Shijiazhuang, China
| | - Lei Huang
- Hebei Xiaowutai Mountain National Nature Reserve Management Center, Zhangjiakou, China
| | - Yefei Zhang
- Hebei Xiaowutai Mountain National Nature Reserve Management Center, Zhangjiakou, China
| | - Guomin Chang
- Ministry of Education Key Laboratory of Molecular and Cellular Biology, Hebei Key Laboratory of Animal Physiology, Biochemistry and Molecular Biology, College of Life Sciences, Hebei Normal University, Shijiazhuang, China
| | - Kuang Wang
- Ministry of Education Key Laboratory of Molecular and Cellular Biology, Hebei Key Laboratory of Animal Physiology, Biochemistry and Molecular Biology, College of Life Sciences, Hebei Normal University, Shijiazhuang, China
| | - Mengxue Li
- Ministry of Education Key Laboratory of Molecular and Cellular Biology, Hebei Key Laboratory of Animal Physiology, Biochemistry and Molecular Biology, College of Life Sciences, Hebei Normal University, Shijiazhuang, China
| | - Hui Wang
- Ministry of Education Key Laboratory of Molecular and Cellular Biology, Hebei Key Laboratory of Animal Physiology, Biochemistry and Molecular Biology, College of Life Sciences, Hebei Normal University, Shijiazhuang, China
- *Correspondence: Jingze Liu, ; Hui Wang,
| | - Jingze Liu
- Ministry of Education Key Laboratory of Molecular and Cellular Biology, Hebei Key Laboratory of Animal Physiology, Biochemistry and Molecular Biology, College of Life Sciences, Hebei Normal University, Shijiazhuang, China
- *Correspondence: Jingze Liu, ; Hui Wang,
| |
Collapse
|
17
|
Matias J, Kurokawa C, Sajid A, Narasimhan S, Arora G, Diktas H, Lynn GE, DePonte K, Pardi N, Valenzuela JG, Weissman D, Fikrig E. Tick immunity using mRNA, DNA and protein-based Salp14 delivery strategies. Vaccine 2021; 39:7661-7668. [PMID: 34862075 PMCID: PMC8671329 DOI: 10.1016/j.vaccine.2021.11.003] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2021] [Revised: 09/29/2021] [Accepted: 11/01/2021] [Indexed: 11/28/2022]
Abstract
Guinea pigs exposed to multiple infestations with Ixodes scapularis ticks develop acquired resistance to ticks, which is also known as tick immunity. The I. scapularis salivary components that contribute to tick immunity are likely multifactorial. An anticoagulant that inhibits factor Xa, named Salp14, is present in tick saliva and is associated with partial tick immunity. A tick bite naturally releases tick saliva proteins into the vertebrate host for several days, which suggests that the mode of antigen delivery may influence the genesis of tick immunity. We therefore utilized Salp14 as a model antigen to examine tick immunity using mRNA lipid nanoparticles (LNPs), plasmid DNA, or recombinant protein platforms. salp14 containing mRNA-LNPs vaccination elicited erythema at the tick bite site after tick challenge that occurred earlier, and that was more pronounced, compared with DNA or protein immunizations. Humoral and cellular responses associated with tick immunity were directed towards a 25 amino acid region of Salp14 at the carboxy terminus of the protein, as determined by antibody responses and skin-testing assays. This study demonstrates that the model of antigen delivery, also known as the vaccine platform, can influence the genesis of tick immunity in guinea pigs. mRNA-LNPs may be useful in helping to elicit erythema at the tick bite site, one of the most important early hallmarks of acquired tick resistance. mRNA-LNPs containing tick genes is a useful platform for the development of vaccines that can potentially prevent selected tick-borne diseases.
Collapse
Affiliation(s)
- Jaqueline Matias
- Section of Infectious Diseases, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Cheyne Kurokawa
- Section of Infectious Diseases, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Andaleeb Sajid
- Section of Infectious Diseases, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Sukanya Narasimhan
- Section of Infectious Diseases, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Gunjan Arora
- Section of Infectious Diseases, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Husrev Diktas
- Section of Infectious Diseases, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Geoffrey E Lynn
- Section of Infectious Diseases, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Kathleen DePonte
- Section of Infectious Diseases, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Norbert Pardi
- Department of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Jesus G Valenzuela
- Vector Molecular Biology Section, Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Disease, National Institutes of Health, 12735 Twinbrook Parkway, Rockville, MD 20852, USA
| | - Drew Weissman
- Department of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Erol Fikrig
- Section of Infectious Diseases, Yale University School of Medicine, New Haven, CT 06520, USA.
| |
Collapse
|
18
|
Sajid A, Matias J, Arora G, Kurokawa C, DePonte K, Tang X, Lynn G, Wu MJ, Pal U, Strank NO, Pardi N, Narasimhan S, Weissman D, Fikrig E. mRNA vaccination induces tick resistance and prevents transmission of the Lyme disease agent. Sci Transl Med 2021; 13:eabj9827. [PMID: 34788080 DOI: 10.1126/scitranslmed.abj9827] [Citation(s) in RCA: 71] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Ixodes scapularis ticks transmit many pathogens that cause human disease, including Borrelia burgdorferi. Acquired resistance to I. scapularis due to repeated tick exposure has the potential to prevent tick-borne infectious diseases, and salivary proteins have been postulated to contribute to this process. We examined the ability of lipid nanoparticle–containing nucleoside-modified mRNAs encoding 19 I. scapularis salivary proteins (19ISP) to enhance the recognition of a tick bite and diminish I. scapularis engorgement on a host and thereby prevent B. burgdorferi infection. Guinea pigs were immunized with a 19ISP mRNA vaccine and subsequently challenged with I. scapularis. Animals administered 19ISP developed erythema at the bite site shortly after ticks began to attach, and these ticks fed poorly, marked by early detachment and decreased engorgement weights. 19ISP immunization also impeded B. burgdorferi transmission in the guinea pigs. The effective induction of local redness early after I. scapularis attachment and the inability of the ticks to take a normal blood meal suggest that 19ISP may be used either alone or in conjunction with traditional pathogen-based vaccines for the prevention of Lyme disease, and potentially other tick-borne infections.
Collapse
Affiliation(s)
- Andaleeb Sajid
- Section of Infectious Diseases, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Jaqueline Matias
- Section of Infectious Diseases, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Gunjan Arora
- Section of Infectious Diseases, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Cheyne Kurokawa
- Section of Infectious Diseases, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Kathleen DePonte
- Section of Infectious Diseases, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Xiaotian Tang
- Section of Infectious Diseases, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Geoffrey Lynn
- Section of Infectious Diseases, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Ming-Jie Wu
- Section of Infectious Diseases, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Utpal Pal
- Department of Veterinary Medicine, University of Maryland, College Park, MD 20472, USA
- Virginia-Maryland Regional College of Veterinary Medicine, College Park, MD 20472, USA
| | - Norma Olivares Strank
- Section of Infectious Diseases, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Norbert Pardi
- Department of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Sukanya Narasimhan
- Section of Infectious Diseases, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Drew Weissman
- Department of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Erol Fikrig
- Section of Infectious Diseases, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT 06520, USA
| |
Collapse
|
19
|
Ndawula C. From Bench to Field: A Guide to Formulating and Evaluating Anti-Tick Vaccines Delving beyond Efficacy to Effectiveness. Vaccines (Basel) 2021; 9:vaccines9101185. [PMID: 34696291 PMCID: PMC8539545 DOI: 10.3390/vaccines9101185] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2021] [Revised: 08/25/2021] [Accepted: 08/26/2021] [Indexed: 01/04/2023] Open
Abstract
Ticks are ubiquitous blood-sucking ectoparasites capable of transmitting a wide range of pathogens such as bacteria, viruses, protozoa, and fungi to animals and humans. Although the use of chemicals (acaricides) is the predominant method of tick-control, there are increasing incidents of acaricide tick resistance. Furthermore, there are concerns over accumulation of acaricide residues in meat, milk and in the environment. Therefore, alternative methods of tick-control have been proposed, of which anti-tick cattle vaccination is regarded as sustainable and user-friendly. Over the years, tremendous progress has been made in identifying and evaluating novel candidate tick vaccines, yet none of them have reached the global market. Until now, Bm86-based vaccines (Gavac™ in Cuba and TickGARDPLUS™ Australia-ceased in 2010) are still the only globally commercialized anti-tick vaccines. In contrast to Bm86, often, the novel candidate anti-tick vaccines show a lower protection efficacy. Why is this so? In response, herein, the potential bottlenecks to formulating efficacious anti-tick vaccines are examined. Aside from Bm86, the effectiveness of other anti-tick vaccines is rarely assessed. So, how can the researchers assess anti-tick vaccine effectiveness before field application? The approaches that are currently used to determine anti-tick vaccine efficacy are re-examined in this review. In addition, a model is proposed to aid in assessing anti-tick vaccine effectiveness. Finally, based on the principles for the development of general veterinary vaccines, a pipeline is proposed to guide in the development of anti-tick vaccines.
Collapse
Affiliation(s)
- Charles Ndawula
- National Agricultural Research Organization, P.O. Box 295, Entebbe, Wakiso 256, Uganda;
- National Livestock Resources Research Institute, Vaccinology Research Programme, P.O. Box 5704, Nakyesasa, Wakiso 256, Uganda
| |
Collapse
|
20
|
Kitsou C, Fikrig E, Pal U. Tick host immunity: vector immunomodulation and acquired tick resistance. Trends Immunol 2021; 42:554-574. [PMID: 34074602 PMCID: PMC10089699 DOI: 10.1016/j.it.2021.05.005] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Revised: 05/04/2021] [Accepted: 05/06/2021] [Indexed: 12/25/2022]
Abstract
Ticks have an unparalleled ability to parasitize diverse land vertebrates. Their natural persistence and vector competence are supported by the evolution of sophisticated hematophagy and remarkable host immune-evasion activities. We analyze the immunomodulatory roles of tick saliva which facilitates their acquisition of a blood meal from natural hosts and allows pathogen transmission. We also discuss the contrasting immunological events of tick-host associations in non-reservoir or incidental hosts, in which the development of acquired tick resistance can deter tick attachment. A critical appraisal of the intricate immunobiology of tick-host associations can plant new seeds of innovative research and contribute to the development of novel preventive strategies against ticks and tick-transmitted infections.
Collapse
Affiliation(s)
- Chrysoula Kitsou
- Department of Veterinary Medicine, University of Maryland, College Park, MD, USA
| | - Erol Fikrig
- Section of Infectious Diseases, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT, USA; Howard Hughes Medical Institute, Chevy Chase, MD, USA
| | - Utpal Pal
- Department of Veterinary Medicine, University of Maryland, College Park, MD, USA; Virginia-Maryland College of Veterinary Medicine, College Park, MD, USA.
| |
Collapse
|
21
|
Cattle ticks and tick-borne diseases: a review of Uganda's situation. Ticks Tick Borne Dis 2021; 12:101756. [PMID: 34134062 DOI: 10.1016/j.ttbdis.2021.101756] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2021] [Revised: 04/14/2021] [Accepted: 05/24/2021] [Indexed: 02/06/2023]
Abstract
Herein we review the epidemiology of ticks and tick-borne diseases (TTBDs), their impact on livestock health and on the economy, control and associated challenges in Uganda. Ticks are leading vectors of economically important pathogens and are widespread in Uganda due to suitable climatic conditions. Besides the physical injury inflicted on the animal host, ticks transmit a number of pathogens that can cause morbidity and mortality of livestock if untreated, resulting in economic losses. Uganda suffers an aggregated annual loss (direct and indirect) of over USD 1.1 billion in the TTBDs complex. East Coast fever (ECF) caused by a protozoan haemoparasite, Theileria parva, is the most prevalent and economically important tick-borne disease (TBD) in Uganda and its vector, the brown ear tick (Rhipicephalus appendiculatus) widely distributed. Other prevalent TBDs in Uganda include anaplasmosis, babesiosis and heartwater. We highlight the role of agro-ecological zones (AEZs) and livestock management system in the distribution of TTBDs, citing warm and humid lowlands as being ideal habitats for ticks and endemic for TBDs. Control of TTBDs is a matter of great importance as far as animal health is concerned in Uganda. Indigenous cattle, which make up over 90% of the national herd are known to be more tolerant to TTBDs and most farms rely on endemic stability to TBDs for control. However, exotic cattle breeds are more capital intensive than indigenous breeds, but the increasing adoption of tick-susceptible exotic cattle breeds (especially dairy) in western and central Uganda demands intensive use of acaricides for tick control and prevention of TBDs. Such acaricide pressure has unfortunately led to selection of acaricide-resistant tick populations and the consequent acaricide resistance observed in the field. Vaccination against ECF, selective breeding for tick resistance and integrated tick control approaches that limit tick exposure, could be adopted to interrupt spread of acaricide resistance. We recommend increasing monitoring and surveillance for TTBDs and for emerging acaricide resistance, improved extension services and sensitization of farmers on tick control measures, appropriate acaricide use and the development and implementation of vaccines for the control of TTBDs as more sustainable and effective interventions. A tick control policy should be developed, taking into account variations of agro-ecological zones, farm circumstances and indigenous technical knowledge, and this should be incorporated into the overall animal health program.
Collapse
|
22
|
Butler RA, Kennedy ML, Houston AE, Bowers EK, Coons LB, Paulsen D, Trout Fryxell RT. No Evidence of Competition Between the Blacklegged Tick (Acari: Ixodidae) and American Dog Tick on the Rodent Host White-Footed Deermouse (Rodentia: Cricetidae) in Southwestern Tennessee. JOURNAL OF MEDICAL ENTOMOLOGY 2021; 58:1470-1475. [PMID: 33629730 DOI: 10.1093/jme/tjab012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Indexed: 06/12/2023]
Abstract
Investigations that analyze interspecific associations of vectors on their hosts are important for understanding community structure and implementing ways to comprehend mechanisms of pathogen transmission. We assessed the interspecific association of two tick species (Ixodes scapularis Say (Ixodida: Ixodidae) and Dermacentor variabilis Say (Ixodida: Ixodidae)) on the rodent host Peromyscus leucopus Rafinesque (Rodentia: Cricetidae) at the Hobart Ames Research and Education Center in southwestern Tennessee. Of the rodents captured, 95 (63%) had neither species of tick, 6 (4%) had both tick species, 25 (16%) had I. scapularis only, and 26 (17%) had D. variabilis only. A coefficient of association (C7 = -0.08) was calculated, which suggested there was competition between the two species of ectoparasites, but this value was not significant, indicating that there was a neutral relationship between the tick species on P. leucopus. The co-occurrence of both tick species on their host at the same time suggested that the two tick species can occupy the same host and use the same resources without competing.
Collapse
Affiliation(s)
- R A Butler
- Department of Biological Sciences, The University of Memphis, Memphis, TN, USA
- Department of Entomology and Plant Pathology, The University of Tennessee, Knoxville, TN, USA
| | - M L Kennedy
- Department of Biological Sciences, The University of Memphis, Memphis, TN, USA
| | - A E Houston
- Department of Forestry, Wildlife and Fisheries, The University of Tennessee, Knoxville, TN, USA
| | - E K Bowers
- Department of Biological Sciences, The University of Memphis, Memphis, TN, USA
| | - L B Coons
- Department of Biological Sciences, The University of Memphis, Memphis, TN, USA
| | - D Paulsen
- Department of Entomology and Plant Pathology, The University of Tennessee, Knoxville, TN, USA
| | - R T Trout Fryxell
- Department of Entomology and Plant Pathology, The University of Tennessee, Knoxville, TN, USA
| |
Collapse
|
23
|
Tick-human interactions: from allergic klendusity to the α-Gal syndrome. Biochem J 2021; 478:1783-1794. [PMID: 33988703 DOI: 10.1042/bcj20200915] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2021] [Revised: 04/06/2021] [Accepted: 04/21/2021] [Indexed: 11/17/2022]
Abstract
Ticks and the pathogens they transmit, including bacteria, viruses, protozoa, and helminths, constitute a growing burden for human and animal health worldwide. The ability of some animal species to acquire resistance to blood-feeding by ticks after a single or repeated infestation is known as acquired tick resistance (ATR). This resistance has been associated to tick-specific IgE response, the generation of skin-resident memory CD4+ T cells, basophil recruitment, histamine release, and epidermal hyperplasia. ATR has also been associated with protection to tick-borne tularemia through allergic klendusity, a disease-escaping ability produced by the development of hypersensitivity to an allergen. In addition to pathogen transmission, tick infestation in humans is associated with the α-Gal syndrome (AGS), a type of allergy characterized by an IgE response against the carbohydrate Galα1-3Gal (α-Gal). This glycan is present in tick salivary proteins and on the surface of tick-borne pathogens such as Borrelia burgdorferi and Anaplasma phagocytophilum, the causative agents of Lyme disease and granulocytic anaplasmosis. Most α-Gal-sensitized individuals develop IgE specific against this glycan, but only a small fraction develop the AGS. This review summarizes our current understanding of ATR and its impact on the continuum α-Gal sensitization, allergy, and the AGS. We propose that the α-Gal-specific IgE response in humans is an evolutionary adaptation associated with ATR and allergic klendusity with the trade-off of developing AGS.
Collapse
|
24
|
Ng YQ, Gupte TP, Krause PJ. Tick hypersensitivity and human tick-borne diseases. Parasite Immunol 2021; 43:e12819. [PMID: 33428244 DOI: 10.1111/pim.12819] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2020] [Revised: 12/22/2020] [Accepted: 01/08/2021] [Indexed: 12/15/2022]
Abstract
Immune-mediated hypersensitivity reactions to ticks and other arthropods are well documented. Hypersensitivity to ixodid (hard bodied) ticks is especially important because they transmit infection to humans throughout the world and are responsible for most vector-borne diseases in the United States. The causative pathogens of these diseases are transmitted in tick saliva that is secreted into the host while taking a blood meal. Tick salivary proteins inhibit blood coagulation, block the local itch response and impair host anti-tick immune responses, which allows completion of the blood meal. Anti-tick host immune responses are heightened upon repeated tick exposure and have the potential to abrogate tick salivary protein function, interfere with the blood meal and prevent pathogen transmission. Although there have been relatively few tick bite hypersensitivity studies in humans compared with those in domestic animals and laboratory animal models, areas of human investigation have included local hypersensitivity reactions at the site of tick attachment and generalized hypersensitivity reactions. Progress in the development of anti-tick vaccines for humans has been slow due to the complexities of such vaccines but has recently accelerated. This approach holds great promise for future prevention of tick-borne diseases.
Collapse
Affiliation(s)
- Yu Quan Ng
- Yale School of Public Health and Yale School of Medicine, New Haven, CT, USA
| | - Trisha P Gupte
- Yale School of Public Health and Yale School of Medicine, New Haven, CT, USA
| | - Peter J Krause
- Yale School of Public Health and Yale School of Medicine, New Haven, CT, USA
| |
Collapse
|
25
|
Reif KE, Backus EA. AC-DC electropenetrography unmasks fine temporal details of feeding behaviors for two tick species on unsedated hosts. Sci Rep 2021; 11:2040. [PMID: 33479263 PMCID: PMC7820320 DOI: 10.1038/s41598-020-80257-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2020] [Accepted: 12/18/2020] [Indexed: 11/09/2022] Open
Abstract
Ticks are significant nuisance pests and vectors of pathogens for humans, companion animals, and livestock. Limited information on tick feeding behaviors hampers development and rigorous evaluation of tick and tick-borne pathogen control measures. To address this obstacle, the present study examined the utility of AC–DC electropenetrography (EPG) to monitor feeding behaviors of adult Dermacentor variabilis and Amblyomma americanum in real-time. EPG recording was performed during early stages of slow-phase tick feeding using an awake calf host. Both tick species exhibited discernable and stereotypical waveforms of low-, medium-, and high-frequencies. Similar waveform families and types were observed for both tick species; however, species-specific waveform structural differences were also observed. Tick waveforms were hierarchically categorized into three families containing seven types. Some waveform types were conserved by both species (e.g., Types 1b, 1c, 2b, 2c) while others were variably performed among species and individually recorded ticks (e.g., Types 1a, 2a, 2d). This study provides a proof-of-principle demonstration of the feasibility for using EPG to monitor, evaluate, and compare tick feeding behaviors, providing a foundation for future studies aimed at correlating specific feeding behaviors with waveforms, and ultimately the influence of control measures and pathogens on tick feeding behaviors.
Collapse
Affiliation(s)
- Kathryn E Reif
- Department of Diagnostic Medicine/Pathobiology, College of Veterinary Medicine, Kansas State University, Manhattan, KS, 66506-5802, USA.
| | - Elaine A Backus
- USDA Agricultural Research Service, San Joaquin Valley Agricultural Sciences Center, 9611 South Riverbend Ave., Parlier, CA, 93648, USA
| |
Collapse
|
26
|
Vaccine approaches applied to controlling dog ticks. Ticks Tick Borne Dis 2021; 12:101631. [PMID: 33494026 DOI: 10.1016/j.ttbdis.2020.101631] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Revised: 11/17/2020] [Accepted: 11/18/2020] [Indexed: 11/20/2022]
Abstract
Ticks are considered the most important vectors in veterinary medicine with a profound impact on animal health worldwide, as well as being key vectors of diseases affecting household pets. The leading strategy applied to dog tick control is the continued use of acaricides. However, this approach is not sustainable due to surging tick resistance, growing public concern over pesticide residues in food and in the environment, and the rising costs associated with their development. In contrast, tick vaccines are a cost-effective and environmentally friendly alternative against tick-borne diseases by controlling vector infestations and reducing pathogen transmission. These premises have encouraged researchers to develop an effective vaccine against ticks, with several proteins having been characterized and used in native, synthetic, and recombinant forms as antigens in immunizations. The growing interaction between domestic pets and people underscores the importance of developing new tick control measures that require effective screening platforms applied to vaccine development. However, as reviewed in this paper, very little progress has been made in controlling ectoparasite infestations in pets using the vaccine approach. The control of tick infestations and pathogen transmission could be obtained through immunization programs aimed at reducing the tick population and interfering in the pathogenic transmission that affects human and animal health on a global scale.
Collapse
|
27
|
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
|
28
|
Karasuyama H, Miyake K, Yoshikawa S. Immunobiology of Acquired Resistance to Ticks. Front Immunol 2020; 11:601504. [PMID: 33154758 PMCID: PMC7591762 DOI: 10.3389/fimmu.2020.601504] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2020] [Accepted: 09/30/2020] [Indexed: 11/13/2022] Open
Abstract
Ticks are blood-sucking arthropods of great importance in the medical and veterinary fields worldwide. They are considered second only to mosquitos as vectors of pathogenic microorganisms that can cause serious infectious disorders, such as Lyme borreliosis and tick-borne encephalitis. Hard (Ixodid) ticks feed on host animals for several days and inject saliva together with pathogens to hosts during blood feeding. Some animal species can acquire resistance to blood-feeding by ticks after a single or repeated tick infestation, resulting in decreased weights and numbers of engorged ticks or the death of ticks in subsequent infestations. Importantly, this acquired tick resistance (ATR) can reduce the risk of pathogen transmission from pathogen-infected ticks to hosts. This is the basis for the development of tick antigen-targeted vaccines to forestall tick infestation and tick-borne diseases. Accumulation of basophils is detected in the tick re-infested skin lesion of animals showing ATR, and the ablation of basophils abolishes ATR in mice and guinea pigs, illustrating the critical role for basophils in the expression of ATR. In this review article, we provide a comprehensive overview of recent advances in our understanding of the cellular and molecular mechanisms responsible for the development and manifestation of ATR, with a particular focus on the role of basophils.
Collapse
Affiliation(s)
- Hajime Karasuyama
- Inflammation, Infection and Immunity Laboratory, TMDU Advanced Research Institute, Tokyo Medical and Dental University (TMDU), Tokyo, Japan
| | - Kensuke Miyake
- Inflammation, Infection and Immunity Laboratory, TMDU Advanced Research Institute, Tokyo Medical and Dental University (TMDU), Tokyo, Japan
| | - Soichiro Yoshikawa
- Department of Cellular Physiology, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| |
Collapse
|
29
|
Kurokawa C, Narasimhan S, Vidyarthi A, Booth CJ, Mehta S, Meister L, Diktas H, Strank N, Lynn GE, DePonte K, Craft J, Fikrig E. Repeat tick exposure elicits distinct immune responses in guinea pigs and mice. Ticks Tick Borne Dis 2020; 11:101529. [PMID: 32993942 DOI: 10.1016/j.ttbdis.2020.101529] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2020] [Revised: 07/23/2020] [Accepted: 07/24/2020] [Indexed: 12/19/2022]
Abstract
Ticks deposit salivary proteins into the skin during a bite to mediate acquisition of a blood meal. Acquired resistance to tick bites has been demonstrated to prevent Borrelia burgdorferi sensu lato (s.l.) transmission. However, the mechanism of resistance, as well as the protective antigens, have remained elusive. To address these unknowns, we utilized a guinea pig model of tick resistance and a mouse model of permissiveness. Guinea pigs developed immunity after multiple Ixodes scapularis tick infestations, characterized by rapid tick detachment and impaired feeding. In comparison, mice tolerated at least 6 infestations with no significant impact on feeding. We analyzed the bite sites by RNA-sequencing and histology, identifying several inflammatory pathways in tick immune animals, such as FcεRI signaling and complement activation, and activation of coagulation pathways that could impair local blood flow. Together, these results identify important pathways altered during tick rejection and potential tick proteins that could serve as vaccine candidates.
Collapse
Affiliation(s)
- Cheyne Kurokawa
- Section of Infectious Diseases, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Sukanya Narasimhan
- Section of Infectious Diseases, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Aurobind Vidyarthi
- Department of Immunobiology, Yale School of Medicine, New Haven, CT 06520, USA
| | - Carmen J Booth
- Department of Comparative Medicine, Yale School of Medicine, New Haven, CT 06520, USA
| | - Sameet Mehta
- Yale Center for Genome Analysis, Yale University, New Haven, CT 06520, USA
| | - Lea Meister
- Clermont Auvergne University School of Engineering Polytech, Clermont-Ferrand, France
| | - Husrev Diktas
- Section of Infectious Diseases, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Norma Strank
- Section of Infectious Diseases, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Geoffrey E Lynn
- Section of Infectious Diseases, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Kathy DePonte
- Section of Infectious Diseases, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Joseph Craft
- Department of Immunobiology, Yale School of Medicine, New Haven, CT 06520, USA; Section of Rheumatology, Department of Internal Medicine, Yale School of Medicine, New Haven, CT 06520, USA.
| | - Erol Fikrig
- Section of Infectious Diseases, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT 06520, USA; Howard Hughes Medical Institute. Chevy Chase, MD 20815, USA.
| |
Collapse
|
30
|
Abstract
Borrelia burgdorferi is the causative agent of Lyme disease and is transmitted to vertebrate hosts by Ixodes spp. ticks. The spirochaete relies heavily on its arthropod host for basic metabolic functions and has developed complex interactions with ticks to successfully colonize, persist and, at the optimal time, exit the tick. For example, proteins shield spirochaetes from immune factors in the bloodmeal and facilitate the transition between vertebrate and arthropod environments. On infection, B. burgdorferi induces selected tick proteins that modulate the vector gut microbiota towards an environment that favours colonization by the spirochaete. Additionally, the recent sequencing of the Ixodes scapularis genome and characterization of tick immune defence pathways, such as the JAK–STAT, immune deficiency and cross-species interferon-γ pathways, have advanced our understanding of factors that are important for B. burgdorferi persistence in the tick. In this Review, we summarize interactions between B. burgdorferi and I. scapularis during infection, as well as interactions with tick gut and salivary gland proteins important for establishing infection and transmission to the vertebrate host. Borrelia burgdorferi has a complex life cycle with several different hosts, causing Lyme disease when it infects humans. In this Review, Fikrig and colleagues discuss how B. burgdorferi infects and interacts with its tick vector to ensure onward transmission.
Collapse
|
31
|
Ye T, Wu J, Xu Z, Chai J, Zeng Q, Zeng B, Gao Y, Guo R, Chen X, Xu X. Esc-1GN shows therapeutic potentials for acne vulgaris and inflammatory pain. J Pept Sci 2020; 26:e3269. [PMID: 32558003 DOI: 10.1002/psc.3269] [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: 03/26/2020] [Revised: 05/16/2020] [Accepted: 05/25/2020] [Indexed: 02/03/2023]
Abstract
The inflammatory response plays important roles in acne vulgaris and pain pathogenesis. In previous study, Esc-1GN with anti-inflammatory, antimicrobial, and lipopolysacchride (LPS) binding activity was identified from the skin of the frog Hylarana guentheri. Here, we report its therapeutic potentials for acne vulgaris and inflammatory pain. Esc-1GN destroyed the cell membrane of Propionibacteria acnes in the membrane permeability assays. In addition, bacterial agglutination test suggested that Esc-1GN triggered the agglutination of P. acnes, which was affected by LPS and Ca2+ . Meanwhile, in vivo anti-P. acnes and anti-inflammatory effects of Esc-1GN were confirmed by reducing the counts of P. acnes in mice ear, relieving P. acnes-induced mice ear swelling, decreasing mRNA expression and the production of pro-inflammatory cytokines, and attenuating the infiltration of inflammatory cells. Moreover, Esc-1GN also displayed antinociceptive effect in mice induced by acetic acid and formalin. Therefore, Esc-1GN is a promising candidate drug for treatment of acne vulgaris and inflammatory pain.
Collapse
Affiliation(s)
- Tiaofei Ye
- Department of Pulmonary and Critical Care Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, 510282, China.,Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, 510060, China
| | - Jiena Wu
- Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, 510060, China
| | - Zhengnan Xu
- Guangzhou Zhixin High School, Guangzhou, 510515, China
| | - Jinwei Chai
- Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, 510060, China
| | - Qingye Zeng
- Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, 510060, China
| | - Baishuang Zeng
- Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, 510060, China
| | - Yahua Gao
- Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, 510060, China
| | - Ruiyin Guo
- Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, 510060, China
| | - Xin Chen
- Department of Pulmonary and Critical Care Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, 510282, China
| | - Xueqing Xu
- Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, 510060, China
| |
Collapse
|
32
|
Villar M, Pacheco I, Merino O, Contreras M, Mateos-Hernández L, Prado E, Barros-Picanço DK, Lima-Barbero JF, Artigas-Jerónimo S, Alberdi P, Fernández de Mera IG, Estrada-Peña A, Cabezas-Cruz A, de la Fuente J. Tick and Host Derived Compounds Detected in the Cement Complex Substance. Biomolecules 2020; 10:E555. [PMID: 32260542 PMCID: PMC7226240 DOI: 10.3390/biom10040555] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2020] [Revised: 04/02/2020] [Accepted: 04/03/2020] [Indexed: 02/07/2023] Open
Abstract
Ticks are obligate hematophagous arthropods and vectors of pathogens affecting human and animal health worldwide. Cement is a complex protein polymerization substance secreted by ticks with antimicrobial properties and a possible role in host attachment, sealing the feeding lesion, facilitating feeding and pathogen transmission, and protection from host immune and inflammatory responses. The biochemical properties of tick cement during feeding have not been fully characterized. In this study, we characterized the proteome of Rhipicephalus microplus salivary glands (sialome) and cement (cementome) together with their physicochemical properties at different adult female parasitic stages. The results showed the combination of tick and host derived proteins and other biomolecules such as α-Gal in cement composition, which varied during the feeding process. We propose that these compounds may synergize in cement formation, solidification and maintenance to facilitate attachment, feeding, interference with host immune response and detachment. These results advanced our knowledge of the complex tick cement composition and suggested that tick and host derived compounds modulate cement properties throughout tick feeding.
Collapse
Affiliation(s)
- Margarita Villar
- SaBio, Instituto de Investigación en Recursos Cinegéticos (IREC-CSIC-UCLM-JCCM), Ronda de Toledo s/n, 13005 Ciudad Real, Spain; (M.V.); (I.P.); (M.C.); (L.M.-H.); (D.K.B.-P.); (J.F.L.-B.); (S.A.-J.); (P.A.); (I.G.F.d.M.)
- Biochemistry Section, Faculty of Science and Chemical Technologies, and Regional Centre for Biomedical Research (CRIB), University of Castilla-La Mancha, 13071 Ciudad Real, Spain
| | - Iván Pacheco
- SaBio, Instituto de Investigación en Recursos Cinegéticos (IREC-CSIC-UCLM-JCCM), Ronda de Toledo s/n, 13005 Ciudad Real, Spain; (M.V.); (I.P.); (M.C.); (L.M.-H.); (D.K.B.-P.); (J.F.L.-B.); (S.A.-J.); (P.A.); (I.G.F.d.M.)
| | - Octavio Merino
- Facultad de Medicina Veterinaria y Zootecnia, Universidad Autónoma de Tamaulipas, Km 5, Carretera Victoria-Mante, CP 87000 Ciudad Victoria, Tamaulipas, Mexico;
| | - Marinela Contreras
- SaBio, Instituto de Investigación en Recursos Cinegéticos (IREC-CSIC-UCLM-JCCM), Ronda de Toledo s/n, 13005 Ciudad Real, Spain; (M.V.); (I.P.); (M.C.); (L.M.-H.); (D.K.B.-P.); (J.F.L.-B.); (S.A.-J.); (P.A.); (I.G.F.d.M.)
| | - Lourdes Mateos-Hernández
- SaBio, Instituto de Investigación en Recursos Cinegéticos (IREC-CSIC-UCLM-JCCM), Ronda de Toledo s/n, 13005 Ciudad Real, Spain; (M.V.); (I.P.); (M.C.); (L.M.-H.); (D.K.B.-P.); (J.F.L.-B.); (S.A.-J.); (P.A.); (I.G.F.d.M.)
- UMR BIPAR, INRAE, ANSES, Ecole Nationale Vétérinaire d’Alfort, Université Paris-Est, 94700 Maisons-Alfort, France;
| | - Eduardo Prado
- Department of Applied Physics, Faculty of Chemical Sciences and Technologies, Universidad de Castilla-La Mancha, Avda. Camilo José Cela 10, 13071 Ciudad Real, Spain;
| | - Dina Karen Barros-Picanço
- SaBio, Instituto de Investigación en Recursos Cinegéticos (IREC-CSIC-UCLM-JCCM), Ronda de Toledo s/n, 13005 Ciudad Real, Spain; (M.V.); (I.P.); (M.C.); (L.M.-H.); (D.K.B.-P.); (J.F.L.-B.); (S.A.-J.); (P.A.); (I.G.F.d.M.)
| | - José Francisco Lima-Barbero
- SaBio, Instituto de Investigación en Recursos Cinegéticos (IREC-CSIC-UCLM-JCCM), Ronda de Toledo s/n, 13005 Ciudad Real, Spain; (M.V.); (I.P.); (M.C.); (L.M.-H.); (D.K.B.-P.); (J.F.L.-B.); (S.A.-J.); (P.A.); (I.G.F.d.M.)
- Sabiotec, Camino de Moledores s/n. 13003, 13071 Ciudad Real, Spain
| | - Sara Artigas-Jerónimo
- SaBio, Instituto de Investigación en Recursos Cinegéticos (IREC-CSIC-UCLM-JCCM), Ronda de Toledo s/n, 13005 Ciudad Real, Spain; (M.V.); (I.P.); (M.C.); (L.M.-H.); (D.K.B.-P.); (J.F.L.-B.); (S.A.-J.); (P.A.); (I.G.F.d.M.)
| | - Pilar Alberdi
- SaBio, Instituto de Investigación en Recursos Cinegéticos (IREC-CSIC-UCLM-JCCM), Ronda de Toledo s/n, 13005 Ciudad Real, Spain; (M.V.); (I.P.); (M.C.); (L.M.-H.); (D.K.B.-P.); (J.F.L.-B.); (S.A.-J.); (P.A.); (I.G.F.d.M.)
| | - Isabel G. Fernández de Mera
- SaBio, Instituto de Investigación en Recursos Cinegéticos (IREC-CSIC-UCLM-JCCM), Ronda de Toledo s/n, 13005 Ciudad Real, Spain; (M.V.); (I.P.); (M.C.); (L.M.-H.); (D.K.B.-P.); (J.F.L.-B.); (S.A.-J.); (P.A.); (I.G.F.d.M.)
| | | | - Alejandro Cabezas-Cruz
- UMR BIPAR, INRAE, ANSES, Ecole Nationale Vétérinaire d’Alfort, Université Paris-Est, 94700 Maisons-Alfort, France;
| | - José de la Fuente
- SaBio, Instituto de Investigación en Recursos Cinegéticos (IREC-CSIC-UCLM-JCCM), Ronda de Toledo s/n, 13005 Ciudad Real, Spain; (M.V.); (I.P.); (M.C.); (L.M.-H.); (D.K.B.-P.); (J.F.L.-B.); (S.A.-J.); (P.A.); (I.G.F.d.M.)
- Department of Veterinary Pathobiology, Center for Veterinary Health Sciences, Oklahoma State University, Stillwater, OK 74078, USA
| |
Collapse
|
33
|
Ixodes scapularis saliva components that elicit responses associated with acquired tick-resistance. Ticks Tick Borne Dis 2020; 11:101369. [PMID: 31924502 DOI: 10.1016/j.ttbdis.2019.101369] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2019] [Revised: 12/20/2019] [Accepted: 12/23/2019] [Indexed: 02/07/2023]
Abstract
Ticks and tick-borne diseases are on the rise world-wide and vaccines to prevent transmission of tick-borne diseases is an urgent public health need. Tick transmission of pathogens to the mammalian host occurs during tick feeding. Therefore, it is reasoned that vaccine targeting of tick proteins essential for feeding would thwart tick feeding and consequently prevent pathogen transmission. The phenomenon of acquired tick-immunity, wherein, repeated tick infestations of non-natural hosts results in the development of host immune responses detrimental to tick feeding has served as a robust paradigm in the pursuit of tick salivary antigens that may be vaccine targeted. While several salivary antigens have been identified, immunity elicited against these antigens have only provided modest tick rejection. This has raised the possibility that acquired tick-immunity is directed against tick components other than tick salivary antigens. Using Ixodes scapularis, the blacklegged tick, that vectors several human pathogens, we demonstrate that immunity directed against tick salivary glycoproteins is indeed sufficient to recapitulate the phenomenon of tick-resistance. These observations emphasize the utility of tick salivary glycoproteins as viable vaccine targets to thwart tick feeding and direct our search for anti-tick vaccine candidates.
Collapse
|
34
|
Bakshi M, Kim TK, Porter L, Mwangi W, Mulenga A. Amblyomma americanum ticks utilizes countervailing pro and anti-inflammatory proteins to evade host defense. PLoS Pathog 2019; 15:e1008128. [PMID: 31756216 PMCID: PMC6897422 DOI: 10.1371/journal.ppat.1008128] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2019] [Revised: 12/06/2019] [Accepted: 10/05/2019] [Indexed: 02/07/2023] Open
Abstract
Feeding and transmission of tick-borne disease (TBD) agents by ticks are facilitated by tick saliva proteins (TSP). Thus, defining functional roles of TSPs in tick evasion is expected to reveal potential targets in tick-antigen based vaccines to prevent TBD infections. This study describes two types of Amblyomma americanum TSPs: those that are similar to LPS activate macrophage (MΦ) to express pro-inflammation (PI) markers and another set that suppresses PI marker expression by activated MΦ. We show that similar to LPS, three recombinant (r) A. americanum insulin-like growth factor binding-related proteins (rAamIGFBP-rP1, rAamIGFBP-rP6S, and rAamIGFBP-rP6L), hereafter designated as PI-rTSPs, stimulated both PBMC -derived MΦ and mice RAW 267.4 MΦ to express PI co-stimulatory markers, CD40, CD80, and CD86 and cytokines, TNFα, IL-1, and IL-6. In contrast, two A. americanum tick saliva serine protease inhibitors (serpins), AAS27 and AAS41, hereafter designated as anti-inflammatory (AI) rTSPs, on their own did not affect MΦ function or suppress expression of PI markers, but enhanced expression of AI cytokines (IL-10 and TGFβ) in MΦ that were pre-activated by LPS or PI-rTSPs. Mice paw edema test demonstrated that in vitro validated PI- and AI-rTSPs are functional in vivo since injection of HEK293-expressed PI-rTSPs (individually or as a cocktail) induced edema comparable to carrageenan-induced edema and was characterized by upregulation of CD40, CD80, CD86, TNF-α, IL-1, IL-6, and chemokines: CXCL1, CCL2, CCL3, CCL5, and CCL11, whereas the AI-rTSPs (individually and cocktail) were suppressive. We propose that the tick may utilize countervailing PI and AI TSPs to regulate evasion of host immune defenses whereby TSPs such as rAamIGFBP-rPs activate host immune cells and proteins such as AAS27 and AAS41 suppress the activated immune cells. Several studies have documented immuno-suppressive activities in whole tick saliva and salivary gland protein extracts. We have made contribution toward understanding the molecular basis of tick feeding, as we have described functions of defined tick saliva immuno-modulatory proteins. We have shown that A. americanum injects two groups of functionally opposed tick saliva proteins: those that could counter-intuitively be characterized as pro-host defense, and those that are expected to have anti-host immune defense functions. Based on our data, we propose that the tick evades host defense using countervailing pro- and anti- inflammatory proteins in which the pro-host defense tick saliva proteins stimulate host immune cells such as macrophages, and the anti-host defense tick saliva proteins suppress functions of the activated immune cells.
Collapse
Affiliation(s)
- Mariam Bakshi
- Department of Veterinary Pathobiology, College of Veterinary Medicine, TAMU, College Station, Texas, United States of America
| | - Tae Kwon Kim
- Department of Veterinary Pathobiology, College of Veterinary Medicine, TAMU, College Station, Texas, United States of America
| | - Lindsay Porter
- Department of Veterinary Pathobiology, College of Veterinary Medicine, TAMU, College Station, Texas, United States of America
| | - Waithaka Mwangi
- Department of Veterinary Pathobiology, College of Veterinary Medicine, TAMU, College Station, Texas, United States of America
- Department of Diagnostic Medicine/Pathobiology, College of Veterinary Medicine, Kansas State University, Manhattan, Kansas, United States of America
| | - Albert Mulenga
- Department of Veterinary Pathobiology, College of Veterinary Medicine, TAMU, College Station, Texas, United States of America
- * E-mail:
| |
Collapse
|
35
|
Amblyomma americanum serpin 27 (AAS27) is a tick salivary anti-inflammatory protein secreted into the host during feeding. PLoS Negl Trop Dis 2019; 13:e0007660. [PMID: 31449524 PMCID: PMC6730956 DOI: 10.1371/journal.pntd.0007660] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2019] [Revised: 09/06/2019] [Accepted: 07/24/2019] [Indexed: 11/20/2022] Open
Abstract
Ticks successfully feed and transmit pathogens by injecting pharmacological compounds in saliva to thwart host defenses. We have previously used LC-MS/MS to identify proteins that are present in saliva of unfed Amblyomma americanum ticks that were exposed to different hosts. Here we show that A. americanum serine protease inhibitor (serpin) 27 (AAS27) is an immunogenic saliva protein that is injected into the host within the first day of tick feeding and is an anti-inflammatory protein that might act by blocking plasmin and trypsin functions. Although AAS27 is injected into the host throughout tick feeding, qRT-PCR and western blotting analyses indicate that the respective transcript and protein are present in high amounts within the first 24 h of tick feeding. Biochemical screening of Pichia pastoris-expressed recombinant (r) AAS27 against mammalian proteases related to host defense shows it is an inhibitor of trypsin and plasmin, with stoichiometry of inhibition indices of 3.5 and 3.8, respectively. Consistent with typical inhibitory serpins, rAAS27 formed heat- and SDS-stable irreversible complexes with both proteases. We further demonstrate that rAAS27 inhibits trypsin with ka of 6.46 ± 1.24 x 104 M-1 s-1, comparable to serpins of other tick species. We show that native AAS27 is part of the repertoire of proteins responsible for the inhibitory activity against trypsin in crude tick saliva. AAS27 is likely utilized by the tick to evade the hosts inflammation defense since rAAS27 blocks both formalin and compound 48/80-induced inflammation in rats. Tick immune sera of rabbits that had acquired resistance against tick feeding following repeated infestations with A. americanum or Ixodes scapularis ticks reacts with rAAS27. Of significant interest, antibody to rAAS27 blocks this serpin inhibitory functions. Taken together, we conclude that AAS27 is an anti-inflammatory protein secreted into the host during feeding and may represent a potential candidate for development of an anti-tick vaccine.
Collapse
|
36
|
Henrique MO, Neto LS, Assis JB, Barros MS, Capurro ML, Lepique AP, Fonseca DM, Sá-Nunes A. Evaluation of inflammatory skin infiltrate following Aedes aegypti bites in sensitized and non-sensitized mice reveals saliva-dependent and immune-dependent phenotypes. Immunology 2019; 158:47-59. [PMID: 31315156 DOI: 10.1111/imm.13096] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2018] [Revised: 05/12/2019] [Accepted: 06/12/2019] [Indexed: 12/19/2022] Open
Abstract
During probing and blood feeding, haematophagous mosquitoes inoculate a mixture of salivary molecules into their vertebrate hosts' skin. In addition to the anti-haemostatic and immunomodulatory activities, mosquito saliva also triggers acute inflammatory reactions, especially in sensitized hosts. Here, we characterize the oedema and the cellular infiltrate following Aedes aegypti mosquito bites in the skin of sensitized and non-sensitized BALB/c mice by flow cytometry. Ae. aegypti bites induced an increased oedema in the ears of both non-sensitized and salivary gland extract- (SGE-)sensitized mice, peaking at 6 hr and 24 hr after exposure, respectively. The quantification of the total cell number in the ears revealed that the cellular recruitment was more robust in SGE-sensitized mice than in non-sensitized mice, and the histological evaluation confirmed these findings. The immunophenotyping performed by flow cytometry revealed that mosquito bites were able to produce complex changes in cell populations present in the ears of non-sensitized and SGE-sensitized mice. When compared with steady-state ears, the leucocyte populations significantly recruited to the skin after mosquito bites in non-sensitized and sensitized mice were eosinophils, neutrophils, monocytes, inflammatory monocytes, mast cells, B-cells and CD4+ T-cells, each one with its specific kinetics. The changes in the absolute number of cells suggested two cell recruitment profiles: (i) a saliva-dependent migration; and (ii) a migration dependent on the immune status of the host. These findings suggest that mosquito bites influence the skin microenvironment by inducing differential cell migration, which is dependent on the degree of host sensitization to salivary molecules.
Collapse
Affiliation(s)
- Maressa O Henrique
- Laboratório de Imunologia Experimental, Departamento de Imunologia, Instituto de Ciências Biomédicas, Universidade de São Paulo, São Paulo, SP, Brazil
| | - Leila S Neto
- Laboratório de Imunologia Experimental, Departamento de Imunologia, Instituto de Ciências Biomédicas, Universidade de São Paulo, São Paulo, SP, Brazil
| | - Josiane B Assis
- Laboratório de Imunologia Experimental, Departamento de Imunologia, Instituto de Ciências Biomédicas, Universidade de São Paulo, São Paulo, SP, Brazil
| | - Michele S Barros
- Laboratório de Imunologia Experimental, Departamento de Imunologia, Instituto de Ciências Biomédicas, Universidade de São Paulo, São Paulo, SP, Brazil
| | - Margareth L Capurro
- Laboratório de Mosquitos Geneticamente Modificados, Departamento de Parasitologia, Instituto de Ciências Biomédicas, Universidade de São Paulo, São Paulo, SP, Brazil.,Instituto Nacional de Ciência e Tecnologia em Entomologia Molecular, Conselho Nacional de Desenvolvimento Científico e Tecnológico (INCT-EM/CNPq), Rio de Janeiro, RJ, Brazil
| | - Ana P Lepique
- Laboratório de Imunomodulação, Departamento de Imunologia, Instituto de Ciências Biomédicas, Universidade de São Paulo, São Paulo, SP, Brazil
| | - Denise M Fonseca
- Laboratório de Imunologia de Mucosas, Departamento de Imunologia, Instituto de Ciências Biomédicas, Universidade de São Paulo, São Paulo, SP, Brazil
| | - Anderson Sá-Nunes
- Laboratório de Imunologia Experimental, Departamento de Imunologia, Instituto de Ciências Biomédicas, Universidade de São Paulo, São Paulo, SP, Brazil.,Instituto Nacional de Ciência e Tecnologia em Entomologia Molecular, Conselho Nacional de Desenvolvimento Científico e Tecnológico (INCT-EM/CNPq), Rio de Janeiro, RJ, Brazil
| |
Collapse
|
37
|
Rego ROM, Trentelman JJA, Anguita J, Nijhof AM, Sprong H, Klempa B, Hajdusek O, Tomás-Cortázar J, Azagi T, Strnad M, Knorr S, Sima R, Jalovecka M, Fumačová Havlíková S, Ličková M, Sláviková M, Kopacek P, Grubhoffer L, Hovius JW. Counterattacking the tick bite: towards a rational design of anti-tick vaccines targeting pathogen transmission. Parasit Vectors 2019; 12:229. [PMID: 31088506 PMCID: PMC6518728 DOI: 10.1186/s13071-019-3468-x] [Citation(s) in RCA: 72] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2019] [Accepted: 04/29/2019] [Indexed: 02/07/2023] Open
Abstract
Hematophagous arthropods are responsible for the transmission of a variety of pathogens that cause disease in humans and animals. Ticks of the Ixodes ricinus complex are vectors for some of the most frequently occurring human tick-borne diseases, particularly Lyme borreliosis and tick-borne encephalitis virus (TBEV). The search for vaccines against these diseases is ongoing. Efforts during the last few decades have primarily focused on understanding the biology of the transmitted viruses, bacteria and protozoans, with the goal of identifying targets for intervention. Successful vaccines have been developed against TBEV and Lyme borreliosis, although the latter is no longer available for humans. More recently, the focus of intervention has shifted back to where it was initially being studied which is the vector. State of the art technologies are being used for the identification of potential vaccine candidates for anti-tick vaccines that could be used either in humans or animals. The study of the interrelationship between ticks and the pathogens they transmit, including mechanisms of acquisition, persistence and transmission have come to the fore, as this knowledge may lead to the identification of critical elements of the pathogens' life-cycle that could be targeted by vaccines. Here, we review the status of our current knowledge on the triangular relationships between ticks, the pathogens they carry and the mammalian hosts, as well as methods that are being used to identify anti-tick vaccine candidates that can prevent the transmission of tick-borne pathogens.
Collapse
Affiliation(s)
- Ryan O. M. Rego
- Biology Centre, Institute of Parasitology, Czech Academy of Sciences, Branišovská 31, 37005 Ceske Budejovice, Czech Republic
| | - Jos J. A. Trentelman
- Amsterdam UMC, Location AMC, Center for Experimental and Molecular Medicine, Amsterdam, The Netherlands
| | - Juan Anguita
- CIC bioGUNE, 48160 Derio, Spain
- Ikerbasque, Basque Foundation for Science, 48012 Bilbao, Spain
| | - Ard M. Nijhof
- Institute for Parasitology and Tropical Veterinary Medicine, Freie Universität Berlin, Berlin, Germany
| | - Hein Sprong
- Centre for Zoonoses and Environmental Microbiology, Centre for Infectious Disease Control, National Institute for Public Health and the Environment (RIVM), Bilthoven, The Netherlands
| | - Boris Klempa
- Institute of Virology, Biomedical Research Center of the Slovak Academy of Sciences, Bratislava, Slovakia
| | - Ondrej Hajdusek
- Biology Centre, Institute of Parasitology, Czech Academy of Sciences, Branišovská 31, 37005 Ceske Budejovice, Czech Republic
| | | | - Tal Azagi
- Centre for Zoonoses and Environmental Microbiology, Centre for Infectious Disease Control, National Institute for Public Health and the Environment (RIVM), Bilthoven, The Netherlands
| | - Martin Strnad
- Biology Centre, Institute of Parasitology, Czech Academy of Sciences, Branišovská 31, 37005 Ceske Budejovice, Czech Republic
- Faculty of Science, University of South Bohemia, Branišovská 31, 37005 Ceske Budejovice, Czech Republic
| | - Sarah Knorr
- Institute for Parasitology and Tropical Veterinary Medicine, Freie Universität Berlin, Berlin, Germany
| | - Radek Sima
- Biology Centre, Institute of Parasitology, Czech Academy of Sciences, Branišovská 31, 37005 Ceske Budejovice, Czech Republic
| | - Marie Jalovecka
- Biology Centre, Institute of Parasitology, Czech Academy of Sciences, Branišovská 31, 37005 Ceske Budejovice, Czech Republic
- Faculty of Science, University of South Bohemia, Branišovská 31, 37005 Ceske Budejovice, Czech Republic
| | - Sabína Fumačová Havlíková
- Institute of Virology, Biomedical Research Center of the Slovak Academy of Sciences, Bratislava, Slovakia
| | - Martina Ličková
- Institute of Virology, Biomedical Research Center of the Slovak Academy of Sciences, Bratislava, Slovakia
| | - Monika Sláviková
- Institute of Virology, Biomedical Research Center of the Slovak Academy of Sciences, Bratislava, Slovakia
| | - Petr Kopacek
- Biology Centre, Institute of Parasitology, Czech Academy of Sciences, Branišovská 31, 37005 Ceske Budejovice, Czech Republic
- Faculty of Science, University of South Bohemia, Branišovská 31, 37005 Ceske Budejovice, Czech Republic
| | - Libor Grubhoffer
- Biology Centre, Institute of Parasitology, Czech Academy of Sciences, Branišovská 31, 37005 Ceske Budejovice, Czech Republic
- Faculty of Science, University of South Bohemia, Branišovská 31, 37005 Ceske Budejovice, Czech Republic
| | - Joppe W. Hovius
- Amsterdam UMC, Location AMC, Center for Experimental and Molecular Medicine, Amsterdam, The Netherlands
| |
Collapse
|
38
|
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
|
39
|
Host-specific expression of Ixodes scapularis salivary genes. Ticks Tick Borne Dis 2018; 10:386-397. [PMID: 30545615 DOI: 10.1016/j.ttbdis.2018.12.001] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2018] [Revised: 09/20/2018] [Accepted: 12/02/2018] [Indexed: 11/22/2022]
Abstract
Ixodes scapularis vectors several pathogens including Borrelia burgdorferi, the agent of Lyme disease. Nymphal and larval stages, and the pathogens transmitted by I. scapularis are maintained in a zoonotic cycle involving rodent reservoir hosts, predominantly Peromyscus leucopus. Humans are not reservoir hosts, however, accidental encounters of infected ticks with humans, results in pathogen transmission to the human host. Laboratory models of non-reservoir hosts such as guinea pigs develop a strong immune response to tick salivary proteins and reject ticks upon repeated tick infestations. Anecdotal and scientific evidence suggests that humans that get frequent tick bites might also develop resistance to ticks. Mus musculus, the laboratory model of natural host, does not develop resistance to I. scapularis upon repeated tick infestations. Addressing this dichotomy in vector-host interaction, we present data that suggest that the salivary transcriptome and proteome composition is different in mouse and guinea pig-fed I. scapularis, and that these differences might contribute to differences in host immune responses. These findings reveal a new insight into vector-host interactions and offer a functional paradigm to better understand the phenomenon of acquired tick-resistance.
Collapse
|
40
|
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
|
41
|
de la Fuente J. Controlling ticks and tick-borne diseases…looking forward. Ticks Tick Borne Dis 2018; 9:1354-1357. [PMID: 29656834 DOI: 10.1016/j.ttbdis.2018.04.001] [Citation(s) in RCA: 59] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2018] [Revised: 04/02/2018] [Accepted: 04/04/2018] [Indexed: 01/05/2023]
Abstract
Tick-borne diseases (TBDs) represent a growing burden for human and animal health worldwide. Several approaches including the use of chemicals with repellency and parasiticidal activity, habitat management, genetic selection of hosts with higher resistance to ticks, and vaccines have been implemented for reducing the risk of TBDs. However, the application of latest gene editing technologies in combination with vaccines likely combining tick and pathogen derived antigens and other control measures should result in the development of effective, safe, and environmentally sound integrated control programs for the prevention and control of TBDs. This paper is not a review of current approaches for the control of ticks and TBDs, but an opinion about future directions in this area.
Collapse
Affiliation(s)
- José de la Fuente
- SaBio, Instituto de Investigación en Recursos Cinegéticos IREC (CSIC-UCLM-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, USA.
| |
Collapse
|