Elsevier

Journal of Proteomics

Volume 128, 14 October 2015, Pages 58-68
Journal of Proteomics

Review Article
Modulation of host immunity by tick saliva

https://doi.org/10.1016/j.jprot.2015.07.005Get rights and content

Highlights

  • Tick salivary secretion is important in the interaction of ticks with the vertebrate hosts and the pathogens they transmit.

  • Recent systems biology approaches have characterized massively gene expression in tick salivary glands.

  • An important function of tick salivary secretion is to modulate vertebrate innate and adaptive immunity.

  • We overview the available literature about the immunomodulatory properties of tick saliva.

  • Our review may serve as a guideline for the discovery of the genes that mediate the specific function of tick saliva.

Abstract

Next generation sequencing and proteomics have helped to comprehensively characterize gene expression in tick salivary glands at both the transcriptome and the proteome level. Functional data are, however, lacking. Given that tick salivary secretions are critical to the success of the tick transmission lifecycle and, as a consequence, for host colonization by the pathogens they spread, we thoroughly review here the literature on the known interactions between tick saliva (or tick salivary gland extracts) and the innate and adaptive vertebrate immune system. The information is intended to serve as a reference for functional characterization of the numerous genes and proteins expressed in tick salivary glands with an ultimate goal to develop novel vector and pathogen control strategies.

Significance

We overview all the known interactions of tick saliva with the vertebrate immune system. The provided information is important, given the recent developments in high-throughput transcriptomic and proteomic analysis of gene expression in tick salivary glands, since it may serve as a guideline for the functional characterization of the numerous newly-discovered genes expressed in tick salivary glands.

Introduction

Ticks are obligatory blood-feeding arthropods that belong to the subclass Acari, order Ixodida, and three families: Ixodidae (hard ticks), Argasidae (soft ticks), and Nuttallielidae. Soft ticks feed repeatedly for minutes to hours, while hard ticks usually stay attached to their hosts and feed for several days or even weeks, but only once in each life stage [1], [2]. The amount of blood ingested is species and life-stage specific, with females of some tick species increasing their volume up to 200 times by the end of blood feeding [3].

Ticks are important vectors that transmit a wide range of pathogens. The most common tick-borne pathogens are viruses and bacteria, but fungi, protozoa, and helminths can also be transmitted [4]. Clinically and epidemiologically, the most important tick-borne diseases are: tick-borne encephalitis (TBE), caused by the TBE virus; Lyme disease, caused by spirochetes belonging to the Borrelia burgdorferi sensu lato complex in Europe and B. burgdorferi sensu stricto in the USA; tick-borne spotted fever, caused by Rickettsia spp.; anaplasmosis, caused by Anaplasma spp.; and babesiosis, caused by Babesia spp. protozoa [5], [6]. Pathogens have different life cycles, but the transmission usually begins with a tick biting an infected vertebrate host and pathogen uptake by the tick in the blood meal. Pathogens, e.g. Borrelia spp. spirochetes then stay in the midgut and wait until next feeding, which triggers their proliferation and migration through the midgut wall to hemocoel and, ultimately, to the salivary glands. Moreover, spirochetes interact with some midgut and salivary components that induce Borrelia proliferation or increase their infectious potential [7]. When the tick bites its next vertebrate host, pathogens are transmitted via tick saliva. In some tick species the pathogens are transmitted transovarially from the female to laid eggs, thus keeping the level of prevalence in the tick population [8]. Tick saliva has been shown to facilitate pathogen transfer to the vertebrate host by virtue of its pharmacological properties, including modulation of the vertebrate immune system [9], [10], [11]. Moreover, tick saliva contains toxins belonging to families also found in venomous animals, such as spiders or snakes, and that can induce paralysis and other toxicoses [12].

To secure uninterrupted blood uptake, ticks suppress and evade the complex physiological host immune and homeostatic responses that are raised against them. Hemostasis, which includes coagulation, vasoconstriction, and platelet aggregation, is the first innate host defense mechanism against the mechanical injury caused by intrusion of tick mouthparts into the host skin. This early vertebrate host response further includes complement activation and inflammation, with the host inflammatory response including, among other factors, rapid leukocyte infiltration after skin injury [13]. Keratinocytes, endothelial cells, and resident leukocytes such as mast cells, dendritic cells, and macrophages make immediate contact with tick saliva or the tick hypostome and are activated. Pro-inflammatory chemokines and cytokines including interleukin-8 (IL-8), tumor necrosis factor (TNF), and IL-1β are released to recruit neutrophils and other inflammatory cells to the area of tick infestation [14]. Following tick feeding, there is activation of both the cellular and humoral branches of vertebrate adaptive immunity [15]. Activated memory T and B cells (in the case of secondary infestation) amplify the host inflammatory response to ticks by releasing specific cytokines and producing antibodies that target tick salivary or mouthpart-derived antigens to activate complement or sensitize mast cells and basophils [9], [14], [15]. The strength and specificity of the host immune response and its effect on tick physiology depend on the host and tick species, the host's health, and its genotype [16]. The same is true for tick defense mechanisms, since both tick salivary components and host immune mechanisms have been co-evolving. As a result, the tick–host interaction can be considered an “arms race” between the new defense mechanisms developed by the host and the evasion strategies developed by ticks [17]. As an adaptation to blood feeding, ticks secrete a complex mixture of immunomodulatory substances in their saliva that suppress both innate and adaptive host immune responses that can cause pain, itch, blood flow disruption in the tick feeding cavity, or even direct damage to the tick, thereby subverting tick rejection and death [18], [19], [20]. Despite the specificity of tick salivary component targets, there is also redundancy at the molecular, cellular, and functional level [9], [13]. The richness and diversity of tick salivary compounds have been established in several transcriptomic studies over the last 15 years and, more recently, by next generation sequencing (NGS) studies.

The rapid developments in NGS and proteomics are reflected in the recent progress made in tick research, in which several transcriptomic and proteomic studies have been published over the last few years. These studies represent a rich data source that provides the basis for functional studies and investigation of gene expression dynamics during tick feeding and different physiological states. For instance, significant differences in the salivary proteome of partially and fully engorged female Rhipicephalus (Boophilus) microplus ticks have been described [21]. More recently, a transcriptomic study described over 800 immuno-proteins in Amblyomma americanum saliva during 24–48 h of feeding [22]. A transcriptomic analysis of Dermacentor andersoni salivary glands resulted in over 500 singletons and 200 clusters in which a number of sequences with similarity to mammalian genes associated with immune response regulation, tumor suppression, and wound healing were identified [23]. By combining transcriptomic and proteomic approaches, nearly 700 proteins were identified in D. andersoni saliva after 2 and 5 days of feeding, from which 157 were postulated to be involved in immunomodulation and blood feeding [24]. Schwarz and colleagues performed a comprehensive study of Ixodes ricinus salivary and midgut transcriptomes and proteomes and found that the transcriptomic and proteomic dynamics did not 100% overlap in different tick tissues [25]. A recent study by Kotsyfakis and colleagues characterized transcriptional dynamics in the I. ricinus female and nymph salivary glands and midguts at various feeding time points [26], and established that some gene families show stage- and time-specific expression, possibly via epigenetic control. In addition, the genes encoding secreted proteins exhibited a high mutation rate, possibly representing a mechanism of antigenic variation, and analysis of the midgut transcriptome revealed several novel enzymes, transporters, and antimicrobial peptides [26]. A transcriptomic analysis of Amblyomma maculatum salivary glands revealed almost 3500 contigs with a secretory function [27]. Another sialome (salivary gland transcriptome) of Amblyomma ticks was published by Garcia and colleagues [28]: the authors analyzed samples from Amblyomma triste nymphs and females, Amblyomma cajennese females, and Amblyomma parvum females and focused on putative transcripts encoding anticoagulants, immunosuppressants, and anti-inflammatory molecules. A further study characterized A. americanum nymph and adult proteomes and compared the data with other Amblyomma species [29]. A Rhipicephalus pulchellus tick sialome study revealed differences between males and females [30], with the sequences identified used for a preliminary proteomic study to identify 460 male and over 2000 female proteins. A sialomic study was also performed in Haemaphysalis flava that revealed tens of thousands of genes, some of which were putative secreted salivary proteins thought to be involved with blood feeding and ingestion [31]. A Rhipicephalus sanguineus salivary proteome showed recycling of host proteins and their secretion back into the host [32]. Lewis and colleagues used a transcriptomic approach to characterize immunogenic Ixodes scapularis salivary proteins present after 24 h of feeding [33]; these appeared to be involved in tick feeding even before the majority of pathogens could be transmitted.

In addition to the analysis of secreted tick salivary proteins, tick-feeding lesions on the host have been analyzed by high-throughput and histological methods. Recently, the feeding lesion of D. andersoni was described in detail together with microarray analysis of host gene expression dynamics, thereby characterizing the inflammatory infiltrate at the feeding site and the changes occurring in the epidermal and dermal compartments near the tick [34], [35]. The skin lesions examined from rats infested by Ornithodoros brasiliensis showed edema, muscle degeneration, and hemorrhage [36], with the rats themselves presenting with a bleeding tendency and signs of toxicosis. O. brasiliensis salivary gland homogenates delayed wound healing and had anti-proliferative or even cytotoxic activity on cultured epithelial cells [37]. An analysis of skin-draining lymph nodes in goats repeatedly infested with A. cajennese nymphs revealed an increased number of antigen presenting cells (APCs) such as B lymphocytes, macrophages, and dendritic cells [38]. A skin lesion from a human infested with female Amblyomma testudinarium was characterized by an inflammatory infiltrate and an eosinophilic cement in the center of the lesion [39]. Feeding lesions from rabbits injected with salivary gland extract (SGE) collected from R. sanguineus ticks after 2, 4, and 6 days of feeding showed signs of inflammation, especially at day four [40], suggesting that molecules present in R. sanguineus SGE have high immunogenicity and that immune reaction raised against SGE is stronger than the immunomodulatory action of R. sanguineus salivary effectors.

Such high-throughput studies in both ticks and hosts and complemented with histological information and detailed characterization of salivary components have made a valuable contribution to our knowledge of the dynamic processes occurring at the tick–host interface. However, experiments with saliva or SGE highlight the complexity of host modulation by the tick in vivo. Characterizing individual salivary components can help link specific pathophysiological events to particular molecules to provide a complete picture of tick–host interactions. In this review, we focus on the immunomodulatory actions of whole tick saliva or salivary gland extracts (SGE) rather than the effects of the individual salivary components, since these are reviewed elsewhere [13], [41], [42].

Section snippets

The role of tick saliva in modulating host hemostasis and complement

Ticks have developed various mechanisms to counteract the hemostatic responses of the host so that they can successfully feed on blood for many days [13], [19]. Serine proteases are key players in host hemostasis and, therefore, are specifically targeted by the wide range of serine protease inhibitors present in tick saliva. The net result is that the physiological balance between host proteases and endogenous anti-proteases is impaired. Tick salivary secretions also contain vasodilators,

Innate immunity and tick saliva

Innate immune responses against tick feeding involve the activation of resident immune cells that initiate and promote the local inflammatory response as a reaction to skin damage. The resident leukocytes are macrophages, Langerhans cells (LCs), mast cells, or innate lymphoid cells, and pro-inflammatory mediators are also released by endothelial cells and keratinocytes [51]. These mediators and complement components are chemotactic for circulating inflammatory cells including neutrophils and

Interaction of macrophages and monocytes with tick saliva

Macrophages are APCs as well as cytokine and chemokine producers [52]. They can be further divided into two different subpopulations: (i) bone marrow-derived hematopoietic macrophages, which circulate as monocytes and, after extravasation at the site of inflammation, differentiate into pro-inflammatory [53] or alternatively-activated macrophages [54] and (ii) tissue-resident macrophages of yolk sac origin that are found in many organs including the skin; the latter tend to be more

Dendritic cells and tick saliva

Dendritic cells (DCs) are APCs and are part of the innate immune system. After immature (unstimulated) DCs recognize and phagocytose pathogens, they mature and migrate to draining lymph nodes where they present antigens derived from the processed pathogen to CD4 + T cells, which subsequently launch an adaptive immune response. Thus, DCs initiate host adaptive immunity via presentation of pathogenic antigens. Two DC states exist: an immature form present in skin or mucosae and a mature form in

Mast cells and tick saliva

Mast cells serve as sentinel cells and reside in many tissues. They are divided into two main types based on the presence of mast cell-specific proteases: connective tissue mast cells, which produce both tryptase and chymase (MCTC), and mucosal mast cells, which produce only tryptase (MCT) [84]; skin mast cells are of the first type. Upon exposure to pathogens or other stimuli, activated mast cells degranulate and release a variety of pre-stored mediators including vasoactive compounds, serine

Granulocytes and tick saliva

Granulocytes are bone marrow-derived myeloid leukocytes that contain granules in their cytoplasm. The granulocyte group consists of three major cell types: basophils, eosinophils, and neutrophils [96].

Basophils and tick saliva

Basophils are IgE-activated granulocytes that, unlike tissue-resident mast cells, circulate in the blood. They play a critical role in the IgE-mediated development of chronic allergic reactions and inflammation [97], [98], and they can also promote polarization towards Th2 responses by IgE-independent antigen presentation in mice [99], [100]. Basophils are recruited to a tick-feeding site and accumulate in the host skin during second and consequent (but not primary) tick infestation, where they

Eosinophils and tick saliva

Eosinophils are mainly present in mucosal areas in contact with the external environment such as the gut or lung mucosae. Their circulating levels are relatively low in healthy organisms, but increase during allergic reactions or parasitic infections [46]. Eosinophils produce cytokines, chemokines, and other mediators, some of which (e.g., indoleamine 2,3 deoxygenase; IDO) induce apoptosis and inhibit T cell proliferation [110], [111]. Eosinophils are also rich in granules that contain

Neutrophils and tick saliva

Neutrophils are granulocytes with both phagocytosis and degranulation roles. They are highly motile cells and they have a relatively short lifespan. Neutrophils play an important role in the early stages of vertebrate immune homeostasis, such as during acute inflammation, but they also play a role in some chronic inflammatory diseases. Neutrophils are generally activated by pathogens and secrete effectors and mediators that promote inflammation by recruiting other leukocytes, and they also

T and B lymphocytes and tick saliva

Adaptive immunity relies on a wide range of antigen receptors (with varying antigen recognition specificities), which are clonally distributed in two types of lymphocytes: T cells and B cells. The induction of a specific immune response is only possible when a foreign antigen is recognized by the corresponding receptor. This first recognition signal is consolidated by the interaction of co-stimulatory molecules on T or B cells with those on APCs — such as dendritic cells or macrophages — that

Natural killer cells and tick saliva

Despite their lymphoid origin, natural killer (NK) cells are part of the innate immune system [46]. Their main function is microbial or tumor cell killing and the regulation of endothelial cell, dendritic cell, and macrophage interactions with T lymphocytes [166]. SGE from female Dermatocentor reticulatus ticks that fed for 3–6 days on mice decreased human NK cell activity, while SGE from unfed or 1 day-fed ticks had no effect. Weaker activity was reported for SGE from A. variegatum and

Conclusions

Tick saliva clearly contains numerous different pharmacologically-active molecules that affect various immune cell populations and facilitate tick feeding. In this “systems biology” era, the effects of tick saliva described in this review can help in the design of experiments to discover specific salivary molecules that account for those effects. Although molecular biology and biochemical methods such as transcriptome and proteome analyses have provided excellent information about the genes

Acknowledgments

We thank Nextgenediting (www.nextgenediting.com) for providing editorial assistance and the anonymous reviewers for their constructive comments. This work was supported by the Grant Agency of the Czech Republic (GACR grant P502/12/2409 to MK), the Academy of Sciences of the Czech Republic (grant no. RVO60077344 to the Biology Center—Institute of Parasitology), and the 7th Framework Program of the European Union (EU FP7; Marie Curie Reintegration grant PIRG07-GA-2010-268177 to MK). This

References (175)

  • R.C. Brusca et al.

    Invertebrates

    (2003)
  • D.E. Sonenshine et al.

    Biology of Ticks

    (2013)
  • J.L. Capinera

    Encyclopedia of Entomology

    (2008)
  • H. Mehlhorn

    Encyclopedia of Parasitology

    (2008)
  • D.E. Sonenshine et al.

    Biology of Ticks

    (2014)
  • P. Parola et al.

    Ticks and tickborne bacterial diseases in humans: an emerging infectious threat

    Clin. Infect. Dis.

    (2001)
  • K. Tilly et al.

    Biology of infection with Borrelia burgdorferi

    Infect. Dis. Clin. N. Am.

    (2008)
  • J.M. Howell et al.

    Transovarial transmission efficiency of Babesia bovis tick stages acquired by Rhipicephalus (Boophilus) microplus during acute infection

    J. Clin. Microbiol.

    (2007)
  • A.S. Bowman et al.

    Ticks: Biology, Disease and Control. Cambridge, UK

    (2008)
  • N. Ramamoorthi et al.

    The Lyme disease agent exploits a tick protein to infect the mammalian host

    Nature

    (2005)
  • X.Y. Liu et al.

    Hard tick factors implicated in pathogen transmission

    PLoS Negl. Trop. Dis.

    (2014)
  • A. Cabezas-Cruz et al.

    Are ticks venomous animals?

    Front. Zool.

    (2014)
  • I.M. Francischetti et al.

    The role of saliva in tick feeding

    Front. Biosci. (Landmark Ed)

    (2009)
  • S. Wikel

    Ticks and tick-borne pathogens at the cutaneous interface: host defenses, tick countermeasures, and a suitable environment for pathogen establishment

    Front. Microbiol.

    (2013)
  • M. Brossard et al.

    Tick immunobiology

    Parasitology

    (2004)
  • B.R. Ferreira et al.

    Antigens from Rhipicephalus sanguineus ticks elicit potent cell-mediated immune responses in resistant but not in susceptible animals

    Vet. Parasitol.

    (2003)
  • B.B. Andrade et al.

    Haematophagous arthropod saliva and host defense system: a tale of tear and blood

    An. Acad. Bras. Cienc.

    (2005)
  • J.W. Hovius

    Spitting image: tick saliva assists the causative agent of Lyme disease in evading host skin's innate immune response

    J. Invest. Dermatol.

    (2009)
  • J.M. Ribeiro et al.

    Role of arthropod saliva in blood feeding: sialome and post-sialome perspectives

    Annu. Rev. Entomol.

    (2003)
  • J. Chmelar et al.

    A tick salivary protein targets cathepsin G and chymase and inhibits host inflammation and platelet aggregation

    Blood

    (2011)
  • L. Tirloni et al.

    Proteomic analysis of cattle tick Rhipicephalus (Boophilus) microplus saliva: a comparison between partially and fully engorged females

    PLoS One

    (2014)
  • Z.M. Radulovic et al.

    A 24–48 h fed Amblyomma americanum tick saliva immuno-proteome

    BMC Genomics

    (2014)
  • F.J. Alarcon-Chaidez et al.

    Transcriptome analysis of the salivary glands of Dermacentor andersoni Stiles (Acari: Ixodidae)

    Insect Biochem. Mol. Biol.

    (2007)
  • L. Mudenda et al.

    Proteomics informed by transcriptomics identifies novel secreted proteins in Dermacentor andersoni saliva

    Int. J. Parasitol.

    (2014)
  • A. Schwarz et al.

    A systems level analysis reveals transcriptomic and proteomic complexity in Ixodes ricinus midgut and salivary glands during early attachment and feeding

    Mol. Cell. Proteomics MCP

    (2014)
  • M. Kotsyfakis et al.

    Tissue- and time-dependent transcription in Ixodes ricinus salivary glands and midguts when blood feeding on the vertebrate host

    Sci. Rep.

    (2015)
  • S. Karim et al.

    A deep insight into the sialotranscriptome of the gulf coast tick, Amblyomma maculatum

    PLoS One

    (2011)
  • G.R. Garcia et al.

    The sialotranscriptome of Amblyomma triste, Amblyomma parvum and Amblyomma cajennense ticks, uncovered by 454-based RNA-seq

    Parasit. Vectors.

    (2014)
  • M. Villar et al.

    Comparative proteomics for the characterization of the most relevant Amblyomma tick species as vectors of zoonotic pathogens worldwide

    J. Proteomics

    (2014)
  • A.W. Tan et al.

    Sexual differences in the sialomes of the zebra tick, Rhipicephalus pulchellus

    J. Proteomics

    (2015)
  • X.L. Xu et al.

    De novo sequencing, assembly and analysis of salivary gland transcriptome of Haemaphysalis flava and identification of sialoprotein genes

    Infect. Genet. Evol.

    (2015)
  • C.J. Oliveira et al.

    Proteome of Rhipicephalus sanguineus tick saliva induced by the secretagogues pilocarpine and dopamine

    Ticks Tick Borne Dis.

    (2013)
  • L.A. Lewis et al.

    Identification of 24 h Ixodes scapularis immunogenic tick saliva proteins

    Ticks Tick Borne Dis.

    (Apr 2015)
  • D.M. Heinze et al.

    Murine cutaneous responses to the rocky mountain spotted fever vector, Dermacentor andersoni, feeding

    Front. Microbiol.

    (2014)
  • D.M. Heinze et al.

    Transcriptional profiling of the murine cutaneous response during initial and subsequent infestations with Ixodes scapularis nymphs

    Parasit. Vectors.

    (2012)
  • J. Reck et al.

    Experimentally induced tick toxicosis in rats bitten by Ornithodoros brasiliensis (Chelicerata: Argasidae): a clinico-pathological characterization

    Toxicon

    (2014)
  • J. Reck et al.

    Ornithodoros brasiliensis (mouro tick) salivary gland homogenates inhibit in vivo wound healing and in vitro endothelial cell proliferation

    Parasitol. Res.

    (2013)
  • G.E. Monteiro et al.

    Antigen-presenting cells in draining lymph nodes of goats repeatedly infested by the Cayenne tick Amblyomma cajennense nymphs

    Exp. Appl. Acarol.

    (2011)
  • J. Kim et al.

    Perianal tick-bite lesion caused by a fully engorged female Amblyomma testudinarium

    Korean J. Parasitol.

    (2014)
  • L.M. Hebling et al.

    Inoculation of salivary gland extracts obtained from female of Rhipicephalus sanguineus (Latreille, 1806) (Acari, Ixodidae) with 2, 4, and 6 days of feeding in rabbit: I—histopathology of the feeding lesion

    Parasitol. Res.

    (2013)
  • M. Kazimirova et al.

    Tick salivary compounds: their role in modulation of host defences and pathogen transmission

    Front. Cell. Infect. Microbiol.

    (2013)
  • I. Stibraniova et al.

    Immunomodulators in tick saliva and their benefits

    Acta Virol.

    (2013)
  • I.M. Francischetti

    Platelet aggregation inhibitors from hematophagous animals

    Toxicon

    (2010)
  • J. Chmelar et al.

    Tick salivary secretion as a source of antihemostatics

    J. Proteomics

    (2012)
  • K. Rother et al.

    The Complement System

    (1998)
  • K. Murphy et al.

    Janeway's Immunobiology

    (2012)
  • J.M. Ribeiro

    Role of saliva in blood-feeding by arthropods

    Annu. Rev. Entomol.

    (1987)
  • C.H. Lawrie et al.

    Investigation of the mechanisms of anti-complement activity in Ixodes ricinus ticks

    Mol. Immunol.

    (2005)
  • C.H. Lawrie et al.

    Ixodes ticks: serum species sensitivity of anticomplement activity

    Exp. Parasitol.

    (1999)
  • H. Schroeder et al.

    Subversion of complement by hematophagous parasites

    Dev. Comp. Immunol.

    (2009)
  • Cited by (0)

    1

    Both authors contributed equally to this work.

    View full text