Open access peer-reviewed chapter

The Enigma of Identifying New Cattle Tick Vaccine Antigens

Written By

Ala E. Tabor

Submitted: 04 May 2018 Reviewed: 25 August 2018 Published: 05 November 2018

DOI: 10.5772/intechopen.81145

From the Edited Volume

Ticks and Tick-Borne Pathogens

Edited by Muhammad Abubakar and Piyumali K. Perera

Chapter metrics overview

1,334 Chapter Downloads

View Full Metrics

Abstract

Several reviews have summarised cattle tick Rhipicephalus (Boophilus) microplus vaccine candidate discoveries by comparing efficacies and localisation characteristics. However, few have re-analysed all the reported proteins using modern bioinformatics tools. Bm86 was developed as a successful vaccine in the 1980s; however, global efficacies vary from 45 to 100%. Subsequent vaccines, including four published patents, were discovered by targeting enzymes important for blood digestion and/or metabolism or by targeting genes shown to disrupt tick survival following RNA interference experiments. This chapter analyses published vaccine candidates using InterPro, BLASTP, SignalP, TMHMM and PredGPI tools to confirm whether each reported protein is likely to be secreted, membrane associated or intracellular. Conversely, these proteins are considered as ‘exposed’, ‘exposed’ and ‘concealed’ or ‘concealed’, respectively. Bm86 was always described as a ‘concealed’ antigen; however, the protein has a confirmed signal peptide and GPI anchor which suggests it is anchored to the cell membrane and exposed on the surface of gut cells. It is the only tick vaccine with a GPI anchor. Secreted vaccine candidates appear to have promise and exhibit higher efficacies if delivered with an ‘intracellular’/‘concealed’ antigen. Improvements in tick genomics and bovine immunomic resources will assist to identify robust new cattle tick vaccines.

Keywords

  • cattle tick
  • vaccines
  • bioinformatics
  • Bm86
  • review
  • Rhipicephalus microplus

1. Introduction

Cattle ticks (Rhipicephalus (Boophilus) microplus) and the diseases they carry affect almost 80% of the world’s population of domestic cattle at an economic burden approximately $US 25–30 billion per annum [1]. The R. (B.) microplus taxonomic status is based upon Cytochrome c oxidase I (COX1) mitochondrial gene sequencing. There are three clades of R. (B.) microplus, plus R. (B.) australis and R. (B.) annulatus which are monophyletic with a different R. (B.) microplus clade [2, 3]. A recent study expanded this analysis and showed that R. (B.) australis is most similar to a large R. (B.) microplus clade (A) which has worldwide distribution, whereas R. (B.) annulatus is similar to R. (B.) microplus clade B predominantly from China [4]. An additional R. (B.) microplus clade C consists of Malaysian and Indian isolates [3]. Separation of species from several continents using morphological characters was not consistent with the above COI sequence clades and suggested that in some regions there exists a mixture of both R. (B.) microplus and R. (B.) australis [4]. It has been noted that more crossing studies need to be undertaken using geographically diverse wild strains and preferably not ‘inbred’ colony isolates of R. (B.) microplus before conclusions on clades and species relationships can be confirmed. Publications and sequences reviewed here are most likely to be from different R. (B). microplus clades and R. (B.) australis but will be referred to collectively as Rhipicephalus microplus.

Regardless of the above seemingly complicated taxonomic status, the treatment of cattle tick infestations is either addressed by vaccination using Bm86-based vaccines: TickGARDPLUS (now discontinued) or GAVAC™ and most commonly through the application of chemical acaricides [5]. Bm86 vaccines have diverse efficacies reported worldwide (45–100%), but in a few isolated countries, the vaccines have worked well apart from the need for multiple annual boosts to achieve adequate efficacies [1, 5, 6]. Ticks are also quite capable of developing resistance to acaricides; thus vaccine research continues globally [7] to identify conserved and immunogenic alternatives to Bm86.

The first notion that tick guts could be the source of viable tick vaccines was reported in 1979 [8] where native tick gut and organ extracts protected guinea pigs and cattle from Dermacentor andersoni ticks. The authors also suggested that this vaccine would affect tick feeding and reproduction and would be ideal for ‘Boophilus microplus’ as all tick stages feed on the same host [8]. A gut protein named Bm86 was discovered in the 1980s as a protective antigen isolated from R. microplus in Australia [9]. The most notable characteristics at this time was the presence of epidermal growth factor (EGF) domains which are highly conserved extracellular domains associated with membrane-bound or secreted proteins (https://www.ebi.ac.uk/interpro/entry/IPR000742).

Bm86 is also a glycosylphosphatidylinositol (GPI)-anchored protein and as such is modified post-translationally [10]. It has been proposed that Bm86 is secreted and anchored to gut digestive cells through its C terminus [11]. Using immunogold labelling Bm86 was found to be located on the microvilli of gut digest cells [12]. The immune response induced by Bm86 was hypothesised to be mediated through host complement and anti-Bm86 antibodies which damage the tick gut surface affecting egg viability [13, 14]. However, the actual function of this tick protein has never been determined. Nonetheless, the early successes of Bm86 vaccines such as TickGARDPLUS in Australia and GAVAC™ in Cuba provided researchers with the necessary fervour to identify alternative vaccine candidates to potentially be either ‘broad spectrum’ (i.e. cross protective for different tick species) or with a longer duration of immunity compared to Bm86-based vaccines.

Advertisement

2. Methods

Previously reviewed antigen types were summarised as ‘secreted’, ‘intracellular’ or ‘membrane associated’ [1]. In this review, each antigen was analysed in silico to confirm previously described localisations. Each ORF was submitted to InterPro to determine if the candidate antigen had domains or motifs representative of conserved protein families including the predicted GO Terms associated with ‘biological process’, ‘molecular function’ and ‘cellular component’ (https://www.ebi.ac.uk/interpro/) [15]. InterPro also predicts the presence of signal peptides and transmembrane helices; however these were examined separately using the SignalP 4.1 server (http://www.cbs.dtu.dk/services/SignalP/) [16, 17] and the TMHMM server v. 2 (http://www.cbs.dtu.dk/services/TMHMM/). GPI anchor predictions were undertaken using PredGPI (http://gpcr.biocomp.unibo.it/predgpi/pred.htm) [18]. The BLASTP server was employed to confirm published sequence identities (https://blast.ncbi.nlm.nih.gov/Blast.cgi). This analysis was limited to vaccine candidates reported as screened against R. microplus ticks in cattle challenge trials.

Advertisement

3. Results and discussion

Table 1 summarises BLASTP and InterPro analyses of published R. microplus recombinant vaccines which have cattle (or other ruminant) trial data. Table 2 summarises localisations of these vaccine candidates analysed through SignalP, TMHMM and PredGPI and provides known trial data, references and patents (if applicable).

Antigen descriptionGenBank accession/BLASTP hitInterPro analysisGO term predictions (InterPro)3
Biological processMolecular functionCellular component
Secreted
Angiotensin converting enzyme-like (Bm91)AAB04998.1Family peptidase M2, peptidyl-dipeptidase AGO:0006508 proteolysisGO:0008237 GO:0008241 metallopeptidase and peptidyl-dipeptidase activityGO:0016020 membrane
1Extra-cellular matrix protein (Rm39)No significant hitNo significant hit
Ferritin-2CK1905282Ferritin homologous superfamilyGO:0006826 iron ion transport
GO:0006879 cellular iron ion homeostasis
GO:0008199 ferric iron binding
1Immunoglobulin G-binding protein C (Rm76)AAB68803.1GM2-AP, lipid-recognition domain superfamilyGO:0006689 ganglioside catabolic processGO:0008047 enzyme activator activity
Metalloprotease Bmi-MP4AAZ39660.1Metallopeptidase homologous superfamilyGO:0006508 proteolysisGO:0008237 metallopeptidase
1Metalloprotease (Rm239)BAF43574.1GO:0008237 metallopeptidase
5’ NucleotidaseAAB38963.15’-Nucleotidase/apyraseGO:0009166 nucleotide catabolic processGO:0000166 nucleotide binding
GO:0016787 hydrolase activity
GO:0016788 hydrolase activity-ester bonds
GO:0046872 metal ion binding
1Proteinase inhibitor domain (Rm180)XM_011553087.1Pancreatic trypsin inhibitor Kunitz domain superfamilyGO:0004867 serine-type endopeptidase inhibitor activity
‘SILK’No significant hitNo significant hit
Membrane associated
AquaporinAIT69684.1Aquaporin-likeGO:0055085 transmembrane transportGO:0015267 channel activityGO:0016020 membrane
Bm86/Bm95M29321EGF-like domains
Intracellular
60S acidic ribosomal protein P0AGQ49465.1Ribosomal protein L10PGO:0042254 ribosome biogenesisGO:0005622 intracellular
Glutathione S-transferase Haemaphysalis longicornisAAQ74441.1GST, Mu class homologous superfamilyGO:0008152 metabolic processGO:0004364 GST activity
GO:0005515 protein binding
Subolesin and akirin chimerasABZ89745.1 AGI44632.1Akirin protein family
Trypsin inhibitor 1-BmTI-6P83606.2 CK1867262Pancreatic trypsin inhibitor Kunitz domain superfamilyGO:0004867 serine-type endopeptidase inhibitor activity
VitellinAAA92143.1Lipovitellin-phosvitin complex, lipid transport proteinGO:0006869 lipid transportGO:0005319 lipid transporter activity

Table 1.

Reported Rhipicephalus (Boophilus) microplus antigens with published vaccine challenge efficacies1 analysed using InterPro (GO terms, domains, protein family identification) including relevant GenBank accessions2.

Four proteins conform a cocktail vaccination [30]; see Table 2 for vaccine efficacies.


Sourced from the BmGI database [23].


Nil predictions denoted by a dash.


Antigen descriptionPublished efficacy2Signal P3TMHMM3PredGPI3References and patents4
Secreted
Angiotensin converting enzyme-like (Bm91)7% reduction egg viabilitySecreted[20, 21]
‘Extracellular matrix protein’ Rm39/Sequence811~73% in mix of four proteinsUnknown[30, 37]4
Ferritin-264%Secreted[22, 23, 24]4
Immunoglobulin G-binding protein C Rm76/Sequence761~73% in mix of four proteins1Incomplete ORF (likely secreted)[30, 37]4
Metalloprotease Bmi-MP460%Secreted[29, 74]
Metalloprotease Rm239/Sequence821~73% in mix of four proteins1Incomplete ORF (likely secreted)[30, 37]4
5’ NucleotidaseNo protectionSecretedWeakly probable[19]
Proteinase inhibitor domain Rm180/Sequence791~73% in mix of four proteins1Incomplete ORF (likely secreted)[30, 37]4
‘SILK’62%Secreted[38, 39]
Membrane associated
Aquaporin73%Four transmembrane helices[40]4, [41]
Bm86/Bm9545–100%SecretedHighly probable[53]4, [75, 76]
Intracellular
60S acidic ribosomal protein P0—peptide96%[55, 56]4, [77]
Glutathione S-transferase Haemaphysalis longicornis57%[60]
Subolesin and akirin chimeras83% (deer) 60–75%[39, 53]4, [76]
Trypsin inhibitor 1-BmTI-632%[23, 67]
VitellinNative protein 68%, recombinant 0% (sheep)[70]

Table 2.

Reported Rhipicephalus (Boophilus) microplus antigens with published vaccine challenge efficacies analysed using SignalP (secretion), TMHMM (transmembrane helices) and PredGPI (GPI anchor) including relevant references and patents.

Four proteins conform a cocktail vaccination with ‘Rm’ names [30] and ‘sequence’ names from corresponding patent [37].


Efficacy from cattle tick challenge trial unless otherwise stated in parentheses.


Nil predictions denoted by a dash.


Denotes published patent record.


3.1 Secreted antigens

Most tested antigens are predicted to be secreted with no membrane-associated moieties (transmembrane helices or GPI anchors) (Table 2). The idea of selecting secreted proteins may have been cultivated to identify putative antigens that are more immunogenic in comparison to Bm86 and therefore boosted by natural tick challenge. The latter is usually associated with the injection of proteins by tick salivary glands. Studies have also shown that tick gut proteins also elicit host antibody responses; however perhaps gut protein-based vaccines are less immunogenic, that is, Bm86, which requires multiple annual boosts.

Two secreted proteins were also isolated from salivary gland and gut fractions similarly to how Bm86 was originally derived: 5′ nucleotidase [19] and Bm91 angiotensin converting enzyme-like protein [20, 21]. However, neither demonstrated notable vaccine efficacies to warrant further development (Table 2).

In other studies, successful vaccine candidates were identified in other tick species, that is, Ixodes ricinus (sheep tick) Ferritin-2 at 96% efficacy [22]. The researchers subsequently mined the R. microplus (BmGI) database for a R. microplus IrFerritin-2 homologue [22, 23], and RmFerritin-2 was patented at 64% vaccination efficacy [24]. Ferritin-2 was discovered in the sheep tick when studying iron homeostasis and it was found to be required for optimal tick feeding. In addition, unlike other tick ferritins, it was found to be unique without functional orthologs in vertebrate hosts [25].

Metalloproteases were targeted as vaccine candidates as these proteins were considered crucial for the maintenance of blood meal-related functions in other tick species [26, 27]. After an examination of five R. microplus metalloprotease GenBank sequences (AAZ39657.1-AAZ39661.1; Untulan et al., 2005, unpublished), it was found that Bmi-MP4 (AAZ39660.1) was expressed in female organs and male ticks and exhibited potential antigenic properties in comparison to other R. microplus metalloproteases [28]. A Bmi-MP4 metalloprotease vaccination study in Brazil yielded 60% efficacy as reported in 2015 [29], with no patent published (Table 2). A different Brazil-based study identified an unrelated metalloprotease Rm239/Sequence 82 (31% identity with Bmi-MP4, data not shown) as a component of a cocktail vaccine of four proteins achieving 73% protection in a tick challenge trial [30]. These proteins were identified through a salivary gland transcriptome study; thus in this instance the researchers were targeting secreted salivary proteins. Interestingly, the proteins selected were highly up-regulated in male ticks found on tick susceptible cattle which were not known to induce antibodies in naturally infected bovines [30]. Note that these two metalloproteases (Bmi-MP4 and Rm239/Sequence 82) and the Bm91 angiotensin converting enzyme-like protein described above all possess the GO:0008237 pertaining to ‘metallopeptidase activity’ (Table 1). As metalloproteases are members of a large protein family [31], this may lead to differences between strains or clades of R. microplus causing variable vaccination responses. Metalloproteases have been considered as vaccine candidates for other parasite species such as hookworm and human amebiasis, but no commercial products have emerged [32, 33].

The second protein in the above-described cocktail with Rm239/Sequence 82 metalloproteinase was Rm180/Sequence 79 which has a proteinase inhibitor domain (IPR002223: pancreatic trypsin inhibitor Kunitz domain) similar to a trypsin inhibitor on the ‘intracellular’ list (Tables 1 and 2), also tested in Brazil. Rm180/Sequence 79 in contrast is likely to have a signal peptide based on its top BLAST hit, and this new proteinase inhibitor does not appear to have any homology with known tick proteins (data not shown). Trypsin inhibitors are serine protease inhibitors potentially involved with tick blood meal digestion through the inhibition of trypsin (a serine protease which hydrolyses proteins).

The third protein within the cocktail was Rm76/Sequence 76 (also secreted) which is an immunoglobulin G (IgG)-binding protein C possessing domain IPR036846 ganglioside GM2 activator associated with lipid recognition function (Table 1). The top BLASTP hit for this protein is AAB68803.1 Rhipicephalus appendiculatus IgG-binding protein C at 88% identity. Tick immunoglobulin-binding proteins have been examined previously in several other tick species including R. appendiculatus, Rhipicephalus haemaphysaloides and Ixodes scapularis [34, 35, 36] and are thought to function as tick defences against host antibodies. Rm 239/Sequence 82 (metalloprotease) and Rm76/Sequence 76 (IgG-binding C) were shown to be the most immunogenic proteins in the cocktail vaccine based on antibody titres, predicted T cell epitopes and antibody boosting during tick challenge [30]. The fourth protein in this cocktail Rm39/Sequence 81 did not return any significant hits using BLAST or InterPro thus could not be examined using the parameters in the tables. The vaccine cocktail consisting of the trypsin inhibitor (Sequence 79), IgG-binding protein C (Sequence 76), metalloprotease (Sequence 82) and the unknown protein (Sequence 81) has been patented [37]. All sequences were published in the associated patent [37] without signal peptide regions.

SILK’ protein was predicted from an expressed sequence tag (EST) library prepared from male R. microplus ticks in response to Anaplasma marginale infection, and it was thought to be similar to arachnid flagelliform silk proteins [38]. However, no significant hits of the R. microplus EST to a ‘SILK protein’ sequence could be confirmed in this study. The protein has not been exploited further as an anti-tick or anti-Anaplasma transmission vaccine; however, at 62% efficacy [39] perhaps further study is warranted. No patent has been published.

3.2 Membrane-bound antigens

Apart from Bm86, the only other published antigen with a membrane association was aquaporin. Aquaporin does not have a GPI anchor as Bm86 but has four transmembrane helices predicted by TMHMM (Table 2). A reported 73% trial efficacy has been published and the data patented [40, 41]. The protein was identified in tick gut transcriptome studies and predictably functions as a water-conducting channel. An aquaporin was previously suggested as vaccine candidate for the human blood fluke Schistosoma japonicum with six predicted immunogenic epitopes and an integral membrane structure [42]. No further testing has been reported which is common for many human vaccine candidates. Perhaps the tick aquaporin vaccine will inspire further investigations of similar orthologs in human parasite infections.

Bm86 is thus the only protein with a confirmed GPI anchor that has been examined as a tick vaccine candidate. GPI-anchored proteins are conserved in eukaryotes and are luminal secretory cargo proteins with several functions across mammals and parasites [10, 43]. Notably, the R. microplus5′ nucleotidase (listed as a ‘secreted protein’) was predicted to have a ‘weakly probable GPI anchor’, and it is known that mammalian 5′ nucleotidases possess GPI anchors [10]. In terms of vaccine candidates, GPI-anchored proteins have been investigated in several parasite species such as Leishmania amazonensis [44], Plasmodium falciparum [45], Schistosoma mansoni [46], Theileria annulata [47] and Babesia bovis [48] and have appeared to be associated with host invasion. In mammals, certain GPI-anchored proteins are cytokines with complement regulation functions [10]. Further studies pertaining to the discovery of tick salivary or gut proteins with GPI anchors have not been reported.

3.3 Intracellular antigens

Although Bm86 is cited as a ‘concealed antigen’ [49, 50], it appears to be a combination of ‘exposed’ and ‘concealed’ based on localisation predictions including a signal peptide (Table 2). Antigens in the ‘intracellular’ category do not have predicted signal peptides, GPI anchors or transmembrane helices and thus perhaps should be considered as truly ‘concealed’. Several intracellular antigens have been investigated as tick vaccine antigens; however, results have been variable and seemingly dependent on delivery mechanisms as host antibodies need to target the protein that resides intracellularly.

Subolesin from the akirin protein family (Table 1) has been investigated in several tick species as a putative vaccine candidate [51] with the first R. microplus ORF described in GenBank as accession ABZ89745.1 (Shao et al. 2008, unpublished). Studies have confirmed that subolesin is involved in blood ingestion and reproduction in 2006 [52]; however, no predicted GO terms or other localisation predictions were identified in this study to confirm any of these putative functions (Tables 1 and 2). Subolesin was recently patented with Bm86 as a dual vaccine emulsion at a reported patented efficacy of 100% [53]. This dual vaccine is currently being testing by the CATVAC consortium in Morocco [7]. It is unknown if the varied efficacies of Bm86 will affect the activity of this dual vaccine or whether the short duration of immunity will continue to be an issue as for Bm86-based vaccines. Previously, a strong phenotypic knockdown of Rhipicephalus sanguineus ticks was observed using RNA interference through the silencing of subolesin and Rs86 (R. sanguineus Bm86 homologue) [54].

The 60S acidic ribosomal protein P0 has demonstrated 96% efficacy using a peptide fragment in cattle tick challenge trials in Cuba [55]. This is otherwise a conserved ribosomal protein, and the peptide region selected had significant sequence differences from the host ortholog. This vaccine has been patented and is under further trial testing also through CATVAC [7, 56]. Previously, gene silencing of this intracellular protein was found to be lethal to Haemaphysalis longicornis ticks [57]. Ubiquitin (also an intracellular protein) when silenced is also found to be lethal to R. microplus ticks [58] but was not found to be an effective vaccine candidate [59].

Haemaphysalis longicornisglutathione S-transferase (GST) showed some cross protection against R. microplus in a cattle trial [60]; however, further investigation as a tick vaccine candidate has not been reported. GSTs have been examined by several researchers as candidate parasite vaccines, for example, for hookworm, schistosomiasis and trichinellosis [61, 62, 63], at varying degrees of efficacy. GST proteins are considered as common ‘housekeeping’ genes forming a large protein superfamily present in eukaryotes and prokaryotes [64]. They function as detoxifying enzymes and thus in ticks may function in response to acaricides or in response to tick-borne pathogens and or stress [65, 66].

Trypsin inhibitors are serine protease inhibitors potentially involved with blood meal digestion as described above. A BmTI-6 sequence was identified in the BmGI database [23] and while native protein vaccine efficacies were high (73%), the corresponding recombinant protein efficacy was poor at 32% [67, 68] (Table 2). This particular trypsin inhibitor is not predicted to be secreted (Table 2) thus may have a function different from gut digestion. The protein sequence reported by Andreotti et al. [67] is identical to BmTI-6 P83606.2 [69]. Alternatively, a ‘secreted’ trypsin Inhibitor showed promise within the cocktail vaccine described above [37]. As stated for metalloproteases, trypsin inhibitors are also members of large dynamic protein families which may circumvent host immune responses if administered as vaccines.

Vitellin was investigated as a native vaccine candidate showing some promise in sheep trials through a reduction in female ticks and their weights and a reduction in tick oviposition [70]. However, the recombinant form had no vaccine effect (Table 2), and no further studies were conducted. Vitellin is a high molecular weight yolk lipoglycoprotein, and in ticks and insects, it is synthesised in female fat bodies as a large precursor polypeptide—vitellogenin [70]. In insects, vitellogenin is processed into vitellin polypeptides by specific proteolytic cleavages during passage into haemolymph and/or upon receptor-mediated endocytosis by the developing oocyte [71, 72]. Tick vitellogenins are crucial for egg development and oviposition as demonstrated when silencing of three H. longicornis vitellogenin genes [73]. There are no reports of vitellin or vitellogenin as vaccine candidates in other species to date; however, this could be because they exist in arthropods (ticks and insects) rather than other ‘pathogenic’ species of parasites.

The investigation of intracellular vaccine candidates appears to less likely lead to a successful outcome. Perhaps some of these proteins could be delivered in dual emulsions as shown above for Bm86 and subolesin for a strong vaccination effect. It seems prudent to suggest that an intracellularly localised vaccine candidate requires a mechanism whereby host antibodies are able to access cells internally in order to be active against feeding ticks.

3.4 Other potential protein features

G protein-coupled receptors (GPCRs), also known as ‘seven-(pass)-transmembrane domain receptors’ are associated with many diseases and as such are the targets of several treatments. They are receptors for pheromones, hormones and neurotransmitters and could potentially be targeted as tick vaccine candidates [78]. Most literature associated with GPCR studies in ticks to date are acaricide-related and not associated with vaccines.

3.5 Protective immune response

The identification of tick vaccine candidates since the discovery of Bm86 appears to be haphazard in that selection has involved either targeting an enzyme involved with feeding or metabolism or to target a gene that showed diminished tick survival following RNA interference silencing. Neither of these approaches is directly linked to the development of a protective immune response which is fundamental for a protective vaccine. Many different experiments have been undertaken describing effective tick immune responses in different breeds of cattle including different mixtures of Bos indicus (naturally tick resistant) and Bos taurus (innately tick susceptible) cattle. These studies have also been undertaken in many different geographic regions with the use of highly divergent tick infestation protocols. The latter is particularly problematic where in some instances tick-naïve cattle cannot be sourced, and researchers treat the cattle for ticks prior to artificial tick infestations and subsequent immune studies. This topic has been reviewed in detail elsewhere and will not be repeated here [79]. The latter review summarised that there are different immune responses in tick-susceptible and tick-resistant breeds of cattle. Perhaps different R. microplus tick vaccine candidates will need to be developed for different cattle breeds and crosses? Is the tick host response in tick-resistant breeds of cattle a result of superior immune function or genetic differences or a combination of both? One theory is that naïve tick-resistant breeds are readily primed with epithelial γδ T cells able to respond to larval ticks, whereas susceptible breeds need to recruit these T cells to the larval bite sites [80, 81]. This immune cell recruitment phase seems to manifest in an inefficient immune response in susceptible breeds. It has been a challenge to demonstrate this phenomenon in all immune studies due to the common practise of studying previously exposed cattle in several published experiments, reviewed previously [79].

3.6 Further research

Reverse vaccinology or genome-based approaches have been reviewed elsewhere, and promise in this approach has been reported [1]. Studies have used EST and transcriptome sequence databases to mine for potential tick antigens using a variety of approaches [1, 30]. Tick genomics has only recently become possible due to the availability of new ‘long read’ sequencing technologies and a dramatic decrease in the cost of sequencing large repetitive genomes [82, 83]. Bovine-specific immunology resources are also increasing [84, 85] with earlier research relying on human models for the major histocompatibility complex predictions. In combination with new genome sequences and bovine immunomic resources, a modern approach to identify robust tick candidates could perhaps finally be developed.

Advertisement

4. Conclusions

Although several approaches have been examined, one way to determine the true significance of a particular antigen or protein is to examine the current-published patents associated with cattle tick (R. microplus) vaccines. Upon examination of all patents and publications with cattle trial data to date, there are mixed features for R. microplus vaccine candidates with either secreted, membrane-bound or intracellular localisations which can also be described as ‘exposed’, ‘a combination of exposed and concealed’ and ‘concealed, respectively. Intracellularly localised antigens are truly ‘concealed’ and in comparison to ‘secreted’ antigen types have highly variable outcomes. The key to identifying efficacious vaccine candidate(s) is to determine how best to stimulate a long-term protective immune response. This may also be feasible through new vaccine delivery options such as nanotechnologies or liposomes which may enhance the immunity to previously identified vaccine candidates.

References

  1. 1. Lew-Tabor AE, Rodriguez VM. A review of reverse vaccinology approaches for the development of vaccines against ticks and tick-borne diseases. Ticks and Tick-Borne Diseases. 2016;7(4):573-585
  2. 2. Burger TD, Shao R, Barker SC. Phylogenetic analysis of mitochondrial genome sequences indicates that the cattle tick, Rhipicephalus (Boophilus) microplus, contains a cryptic species. Molecular Phylogenetics and Evolution. 2014;76:241-253
  3. 3. Low VL, Tay ST, Kho KL, Koh FX, Tan TK, Lim YAL, et al. Molecular characterisation of the tick Rhipicephalus microplus in Malaysia: New insights into the cryptic diversity and distinct genetic assemblages throughout the world. Parasites & Vectors. 2015;8(1):341
  4. 4. Berry CM. Resolution of the taxonomic status of Rhipicephalus (Boophilus) microplus [PhD thesis]. University of Glasgow; 2017
  5. 5. de la Fuente J, Almazan C, Canales M, Perez de la Lastra JM, Kocan KM, Willadsen P. A ten-year review of commercial vaccine performance for control of tick infestations on cattle. Animal Health Research Reviews. 2007;8(1):23-28
  6. 6. Suarez M, Rubi J, Pérez D, Cordova V, Salazar Y, Vielma A, et al. High impact and effectiveness of Gavac™ vaccine in the national program for control of bovine ticks Rhipicephalus microplus in Venezuela. Livestock Science. 2016;187:48-52
  7. 7. Schetters T, Bishop R, Crampton M, Kopacek P, Lew-Tabor A, Maritz-Olivier C, et al. Cattle tick vaccine researchers join forces in CATVAC. Parasites & Vectors. 2016;9:105
  8. 8. Allen JR, Humphreys SJ. Immunisation of Guinea pigs and cattle against ticks. Nature. 1979;280(5722):491-493
  9. 9. Willadsen P, Riding GA, McKenna RV, Kemp DH, Tellam RL, Nielsen JN, et al. Immunologic control of a parasitic arthropod. Identification of a protective antigen from Boophilus microplus. Journal of Immunology. 1989;143(4):1346-1351
  10. 10. Paulick MG, Bertozzi CR. The glycosylphosphatidylinositol anchor: A complex membrane-anchoring structure for proteins. Biochemistry. 2008;47(27):6991-7000
  11. 11. González LJ, Cremata JA, Guanche Y, Ramos Y, Triguero A, Cabrera G, et al. The cattle tick antigen, Bm95, expressed in Pichia pastoris contains short chains of N- and O-glycans. Archives of Biochemistry and Biophysics. 2004;432(2):205-211
  12. 12. Gough JM, Kemp DH. Localization of a low abundance membrane protein (Bm86) on the gut cells of the cattle tick Boophilus microplus by immunogold labeling. The Journal of Parasitology. 1993;79(6):900-907
  13. 13. Kemp DH, Pearson RD, Gough JM, Willadsen P. Vaccination against Boophilus microplus: Localization of antigens on tick gut cells and their interaction with the host immune system. Experimental and Appled Acarology. 1989;7(1):43-58
  14. 14. Tellam RL, Smith D, Kemp DH, Willadsen P. Vaccination against ticks. In: Yong WK, editor. Animal Parasite Control Utilizing Biotechnology. Boca Raton, Florida, USA: CRC Press Inc.; 1992. pp. 303-331
  15. 15. Finn RD, Attwood TK, Babbitt PC, Bateman A, Bork P, Bridge AJ, et al. InterPro in 2017—Beyond protein family and domain annotations. Nucleic Acids Research. 2017;45(D1):D190-D199
  16. 16. Nielsen H. Predicting secretory proteins with SignalP. In: Kihara D, editor. Protein Function Prediction: Methods and Protocols. New York, NY: Springer New York; 2017. pp. 59-73
  17. 17. Petersen T, Brunak S, von Heijne G, Nielsen H. SignalP 4.0: Discriminating signal peptides from transmembrane regions. Nature Methods. 2011;8:785-786
  18. 18. Pierleoni A, Martelli PL, Casadio R. PredGPI: A GPI-anchor predictor. BMC Bioinformatics. 2008;9(1):392
  19. 19. Hope M, Jiang X, Gough L, Cadogan L, Josh P, Jonsson N, et al. Experimental vaccination of sheep and cattle against tick infestation using recombinant 5′-nucleotidase. Parasite Immunology. 2010;32:135-142
  20. 20. Lambertz C, Chongkasikit N, Jittapalapong S, Gauly M. Immune response of Bos indicus cattle against the anti-tick antigen Bm91 derived from local Rhipicephalus (Boophilus) microplus ticks and its effect on tick reproduction under natural infestation. Journal of Parasitology Research. 2012;2012:907607
  21. 21. Riding GA, Jarmey J, McKenna RV, Pearson R, Cobon GS, Willadsen P. A protective "concealed" antigen from Boophilus microplus. Purification, localization, and possible function. Journal of Immunology. 1994;153(11):5158-5166
  22. 22. Hajdusek O, Almazan C, Loosova G, Villar M, Canales M, Grubhoffer L, et al. Characterization of ferritin 2 for the control of tick infestations. Vaccine. 2010;28(17):2993-2998
  23. 23. Guerrero FD, Miller RJ, Rousseau M-E, Sunkara S, Quackenbush J, Lee Y, et al. BmGI: A database of cDNAs expressed in Boophilus microplus, the tropical/southern cattle tick. Insect Biochemistry and Molecular Biology. 2005;35:585-595
  24. 24. Kopacek P, Hajdusek O. Ferritin 2 for the Host Immunization against Ticks. Czech Republic; 2009. WO2009155886 A2
  25. 25. Hajdusek O, Sojka D, Kopacek P, Buresova V, Franta Z, Sauman I, et al. Knockdown of proteins involved in iron metabolism limits tick reproduction and development. Proceedings of the National Academy of Sciences of the United States of America. 2009;106(4):1033
  26. 26. Decrem Y, Mariller M, Lahaye K, Blasioli V, Beaufays J, Zouaoui Boudjeltia K, et al. The impact of gene knock-down and vaccination against salivary metalloproteases on blood feeding and egg laying by Ixodes ricinus. International Journal for Parasitology. 2008;38(5):549-560
  27. 27. Harnnoi T, Sakaguchi T, Nishikawa Y, Xuan X, Fujisaki K. Molecular characterization and comparative study of 6 salivary gland metalloproteases from the hard tick, Haemaphysalis longicornis. Comparative Biochemistry and Physiology Part B: Biochemistry and Molecular Biology. 2007;147(1):93-101
  28. 28. Ali A, Tirloni L, Isezaki M, Seixas A, Konnai S, Ohashi K, et al. Reprolysin metalloproteases from Ixodes persulcatus, Rhipicephalus sanguineus and Rhipicephalus microplus ticks. Experimental and Applied Acarology. 2014;63(4):559-578
  29. 29. Ali A, Parizi LF, Guizzo MG, Tirloni L, Seixas A, Vaz Ida S Jr, et al. Immunoprotective potential of a Rhipicephalus (Boophilus) microplus metalloprotease. Veterinary Parasitology. 2015;207(1-2):107-114
  30. 30. Maruyama SR, Garcia GR, Teixeira FR, Brandao LG, Anderson JM, Ribeiro JMC, et al. Mining a differential sialotranscriptome of Rhipicephalus microplus guides antigen discovery to formulate a vaccine that reduces tick infestations. Parasites & Vectors. 2017;10(1):206
  31. 31. Pulkoski-Gross AE. Historical perspective of matrix metalloproteases. Frontiers in Bioscience (Scholar Edition). 2015;7:125-149
  32. 32. Wiśniewski M, Jaros S, Bąska P, Cappello M, Wędrychowicz H. Ancylostoma ceylanicum metalloprotease 6 DNA vaccination induces partial protection against hookworm challenge infection. Acta Parasitologica. 2013;58:376-383
  33. 33. Roncolato EC, Teixeira JE, Barbosa JE, Zambelli Ramalho LN, Huston CD. Immunization with the Entamoeba histolytica surface metalloprotease EhMSP-1 protects hamsters from amebic liver abscess. Infection and Immunity. 2015;83:713-720
  34. 34. Wang H, Nuttall PA. Immunoglobulin-binding proteins in ticks: New target for vaccine development against a blood-feeding parasite. Cellular and Molecular Life Sciences. 1999;56:286-295
  35. 35. Packila M, Guilfoile PG. Mating, male Ixodes scapularis express several genes including those with sequence similarity to immunoglobulin-binding proteins and metalloproteases. Experimental and Applied Acarology. 2002;27:151-160
  36. 36. Gong H, Qin S, Wan X, Zhang H, Zhou Y, Cao J, et al. Immunoglobulin G binding protein (IGBP) from Rhipicephalus haemaphysaloides: Identification, expression, and binding specificity. Parasitology Research. 2014;113(12):4387-4395
  37. 37. de Miranda Santos IKF, Ribeiro JMC, Ferreira BR, Veríssimo CJ, Brandao LG, Maruyama SRC, et al. Method for Selecting Tick Antigens and Antigenic Compositions against Ticks. Brazil; 2015. WO2015077855 A1
  38. 38. Zivkovic Z, Esteves E, Almazan C, Daffre S, Nijhof AM, Kocan KM, et al. Differential expression of genes in salivary glands of male Rhipicephalus (Boophilus) microplus in response to infection with Anaplasma marginale. BMC Genomics. 2010;11:186
  39. 39. Merino O, Antunes S, Mosqueda J, Moreno-Cid JA, Pérez de la Lastra JM, Rosario-Cruz R, et al. Vaccination with proteins involved in tick–pathogen interactions reduces vector infestations and pathogen infection. Vaccine. 2013;31(49):5889-5896
  40. 40. Guerrero F, Perez de Leon AA. Vaccination of Animals to Elicit a Protective Immune Response against Tick Infestations and Tick-Borne Pathogen Transmission. USA; 2013. WO2013177142 A1
  41. 41. Guerrero FD, Andreotti R, Bendele KG, Cunha RC, Miller RJ, Yeater K, et al. Rhipicephalus (Boophilus) microplus aquaporin as an effective vaccine antigen to protect against cattle tick infestations. Parasites & Vectors. 2014;7:475
  42. 42. Song J, He Q-F. Bioinformatics analysis of the structure and linear B–cell epitopes of aquaporin–3 from Schistosoma japonicum. Asian Pacific Journal of Tropical Medicine. 2012;5(2):107-109
  43. 43. Muñiz M, Zurzolo C. Sorting of GPI-anchored proteins from yeast to mammals—Common pathways at different sites? Journal of Cell Science. 2014;127(13):2793-2801
  44. 44. Colhone MC, Silva-Jardim I, Stabeli RG, Ciancaglini P. Nanobiotechnologic approach to a promising vaccine prototype for immunisation against leishmaniasis: A fast and effective method to incorporate GPI-anchored proteins of Leishmania amazonensis into liposomes. Journal of Microencapsulation. 2015;32(2):143-150
  45. 45. Reddy KS, Amlabu E, Pandey AK, Mitra P, Chauhan VS, Gaur D. Multiprotein complex between the GPI-anchored CyRPA with PfRH5 and PfRipr is crucial for Plasmodium falciparum erythrocyte invasion. Proceedings of the National Academy of Sciences of the United States of America. 2015;112(4):1179-1184
  46. 46. Martins VP, Pinheiro CS, Figueiredo BCP, Assis NRG, Morais SB, Caliari MV, et al. Vaccination with enzymatically cleaved GPI-anchored proteins from Schistosoma mansoni induces protection against challenge infection. Clinical and Developmental Immunology. 2012;2012:11
  47. 47. Abbasnia T, Asoodeh A, Habibi G, Haghparast A. Isolation and purification of glycosylphosphatidylinositols (GPIs) in the schizont stage of Theileria annulata and determination of antibody response to GPI anchors in vaccinated and infected animals. Parasites & Vectors. 2018;11(1):82
  48. 48. Rodriguez AE, Florin-Christensen M, Flores DA, Echaide I, Suarez CE, Schnittger L. The glycosylphosphatidylinositol-anchored protein repertoire of Babesia bovis and its significance for erythrocyte invasion. Ticks and Tick-borne Diseases. 2014;5(3):343-348
  49. 49. Willadsen P, McKenna RV. Vaccination with ‘concealed’ antigens: Myth or reality? Parasite Immunology. 1991;13(6):605-616
  50. 50. Nuttall PA, Trimnell AR, Kazimirova M, Labuda M. Exposed and concealed antigens as vaccine targets for controlling ticks and tick-borne diseases. Parasite Immunology. 2006;28:155-163
  51. 51. de la Fuente J, Moreno-Cid JA, Galindo RC, Almazan C, Kocan KM, Merino O, et al. Subolesin/Akirin vaccines for the control of arthropod vectors and vectorborne pathogens. Transboundary and Emerging Diseases. 2013;60(Suppl 2):172-178
  52. 52. de la Fuente J, Almazan C, Blas-Machado U, Naranjo V, Mangold AJ, Blouin EF, et al. The tick protective antigen, 4D8, is a conserved protein involved in modulation of tick blood ingestion and reproduction. Vaccine. 2006;24(19):4082-4095
  53. 53. Schetters TPM, Jansen T. Vaccine against Rhipicephalus Ticks. Intervet International BV; 2017. WO2014154847 A1
  54. 54. de la Fuente J, Almazan C, Naranjo V, Blouin EF, Kocan KM. Synergistic effect of silencing the expression of tick protective antigens 4D8 and Rs86 in Rhipicephalus sanguineus by RNA interference. Parasitology Resesearch. 2006;99(2):108-113
  55. 55. Rodriguez-Mallon A, Encinosa PE, Mendez-Perez L, Bello Y, Rodriguez Fernandez R, Garay H, et al. High efficacy of a 20 amino acid peptide of the acidic ribosomal protein P0 against the cattle tick, Rhipicephalus microplus. Ticks and Tick-borne Diseases. 2015;6(4):530-537
  56. 56. Rodriguez Mallon A, Fernandez Diaz E, Estrada Garcia MP, Carpio Gonzalez Y. Vaccine Composition for Controlling Ectoparasite Infestations. Cuba; 2012. WO2012041260 A1
  57. 57. Gong H, Liao M, Zhou J, Hatta T, Huang P, Zhang G, et al. Gene silencing of ribosomal protein P0 is lethal to the tick Haemaphysalis longicornis. Veterinary Parasitology. 2008;151(2-4):268-278
  58. 58. Kurscheid S, Lew-Tabor AE, Rodriguez Valle M, Bruyeres AG, Doogan VJ, Munderloh UG, et al. Evidence of a tick RNAi pathway by comparative genomics and reverse genetics screen of targets with known loss-of-function phenotypes in Drosophila. BMC Molecular Biology. 2009;10:26
  59. 59. Almazan C, Moreno-Cantu O, Moreno-Cid A, Galindo RC, Canales M, Villar M, et al. Control of tick infestations in cattle vaccinated with bacterial membranes containing surface-exposed tick protective antigens. Vaccine. 2012;30:265-272
  60. 60. Parizi LF, Utiumi KU, Imamura S, Onuma M, Ohashi K, Masuda A, et al. Cross immunity with Haemaphysalis longicornis glutathione S-transferase reduces an experimental Rhipicephalus (Boophilus) microplus infestation. Experimental Parasitology. 2011;127(1):113-118
  61. 61. Curti E, Seid CA, Hudspeth E, Center L, Rezende W, Pollet J, et al. Optimization and revision of the production process of the Necator americanus glutathione S-transferase 1 (Na-GST-1), the lead hookworm vaccine recombinant protein candidate. Human Vaccines & Immunotherapeutics. 2014;10(7):1914-1925
  62. 62. Mbanefo EC, Kumagai T, Kodama Y, Kurosaki T, Furushima-Shimogawara R, Cherif MS, et al. Immunogenicity and anti-fecundity effect of nanoparticle coated glutathione S-transferase (SjGST) DNA vaccine against murine Schistosoma japonicum infection. Parasitology International. 2015;64(4):24-31
  63. 63. Li LG, Wang ZQ, Liu RD, Yang X, Liu LN, Sun GG, et al. Trichinella spiralis: Low vaccine potential of glutathione S-transferase against infections in mice. Acta Tropica. 2015;146:25-32
  64. 64. Sheehan D, Meade G, Foley VM, Dowd CA. Structure, function and evolution of glutathione transferases: Implications for classification of non-mammalian members of an ancient enzyme superfamily. Biochemical Journal. 2001;360(1):1-16
  65. 65. Duscher GG, Galindo RC, Tichy A, Hummel K, Kocan KM, de la Fuente J. Glutathione S-transferase affects permethrin detoxification in the brown dog tick, Rhipicephalus sanguineus. Ticks and Tick-borne Diseases. 2014;5(3):225-233
  66. 66. Sunyakumthorn P, Petchampai N, Grasperge BJ, Kearney MT, Sonenshine DE, Macaluso KR. Gene expression of tissue-specific molecules in ex vivo Dermacentor variabilis (Acari: Ixodidae) during Rickettsial exposure. Journal of Medical Entomology. 2013;50(5):1089-1096
  67. 67. Andreotti R, Cunha RC, Soares MA, Guerrero FD, Leite FP, de Leon AA. Protective immunity against tick infestation in cattle vaccinated with recombinant trypsin inhibitor of Rhipicephalus microplus. Vaccine. 2012;30(47):6678-6685
  68. 68. Andreotti R, Gomes A, Malavazi-Piza KC, Sasaki SD, Sampaio CAM, Tanaka AS. BmTI antigens induce a bovine protective immune response against Boophilus microplus tick. International Immunopharmacology. 2002;2(4):557-563
  69. 69. Sasaki SD, Tanaka AS. rBmTI-6, a Kunitz-BPTI domain protease inhibitor from the tick Boophilus microplus, its cloning, expression and biochemical characterization. Veterinary Parasitology. 2008;155(1-2):133-141
  70. 70. Tellam RL, Kemp D, Riding G, Briscoe S, Smith D, Sharp P, et al. Reduced oviposition of Boophilus microplus feeding on sheep vaccinated with vitellin. Veterinary Parasitology. 2002;103(1-2):141-156
  71. 71. Chen TT, Hillen LJ. Expression of the vitellogenin genes in insects. Gamete Research. 1983;7:179-196
  72. 72. Raikhel AS, Dhadialla TS. Accumulation of yolk proteins in insect oocytes. Annual Review of Entomology. 1992;37:217-251
  73. 73. Boldbaatar D, Umemiya-Shirafuji R, Liao M, Tanaka T, Xuan X, Fujisaki K. Multiple vitellogenins from the Haemaphysalis longicornis tick are crucial for ovarian development. Journal of Insect Physiology. 2010;56(11):1587-1598
  74. 74. Untalan PM, Pruett JH, Davey RB. Molecular Characterization of Five Salivary Gland Metalloproteases from the Southern Cattle Tick, Boophilus microplus. AAZ39657.1-39661.1. GenBank; 2005
  75. 75. Rand KN, Moore T, Sriskantha A, Spring K, Tellam R, Willadsen P, et al. Cloning and expression of a protective antigen from the cattle tick Boophilus microplus. Proceedings of the National Academy of Sciences of the United States of America. 1989;86(24):9657-9661
  76. 76. Carreon D, de la Lastra JM, Almazan C, Canales M, Ruiz-Fons F, Boadella M, et al. Vaccination with BM86, subolesin and akirin protective antigens for the control of tick infestations in white tailed deer and red deer. Vaccine. 2012;30(2):273-279
  77. 77. Rodriguez-Mallon A, Fernandez E, Encinosa PE, Bello Y, Mendez-Perez L, Ruiz LC, et al. A novel tick antigen shows high vaccine efficacy against the dog tick, Rhipicephalus sanguineus. Vaccine. 2012;30(10):1782-1789
  78. 78. Guerrero FD, Kellogg A, Ogrey AN, Heekin AM, Barrero R, Bellgard MI, et al. Prediction of G protein-coupled receptor encoding sequences from the synganglion transcriptome of the cattle tick, Rhipicephalus microplus. Ticks and Tick-borne Diseases. 2016;7(5):670-677
  79. 79. Tabor AE, Ali A, Rehman G, Rocha Garcia G, Zangirolamo AF, Malardo T, et al. Cattle tick Rhipicephalus microplus-host interface: A review of resistant and susceptible host responses. Frontiers in Cellular and Infection Microbiology. 2017;7:506
  80. 80. Piper EK, Jackson LA, Bagnall N, Kongsuwan K, Lew AE, Jonsson NN. Gene expression in skin of Bos taurus and Bos indicus cattle infested with the cattle tick. Veterinary Immunology and Immunopathology. 2008;126:110-119
  81. 81. Constantinoiu CC, Jackson LA, Jorgensen WK, Lew-Tabor AE, Piper EK, Mayer DG, et al. Local immune response against larvae of Rhipicephalus (Boophilus) microplus in Bos taurus indicus and Bos taurus taurus cattle. International Journal for Parasitology. 2010;40(7):865-875
  82. 82. Barrero RA, Guerrero FD, Black M, McCooke J, Chapman B, Schilkey F, et al. Gene-enriched draft genome of the cattle tick Rhipicephalus microplus: Assembly by the hybrid Pacific biosciences/illumina approach enabled analysis of the highly repetitive genome. International Journal for Parasitology. 2017;47(9):569-583
  83. 83. Cramaro W, Revets D, Hunewald O, Sinner R, Reye A, Muller C. Integration of Ixodes ricinus genome sequencing with transcriptome and proteome annotation of the naive midgut. BMC Genomics. 2015;16(1):871
  84. 84. Pandya M, Rasmussen M, Hansen A, Nielsen M, Buus S, Golde W, et al. A modern approach for epitope prediction: Identification of foot-and-mouth disease virus peptides binding bovine leukocyte antigen (BoLA) class I molecules. Immunogenetics. 2015;67(11):691-703
  85. 85. Carignano HA, Beribe MJ, Caffaro ME, Amadio A, Nani JP, Gutierrez G, et al. BOLA-DRB3 gene polymorphisms influence bovine leukaemia virus infection levels in Holstein and Holstein × Jersey crossbreed dairy cattle. Animal Genetics. 2017;48(4):420-430

Written By

Ala E. Tabor

Submitted: 04 May 2018 Reviewed: 25 August 2018 Published: 05 November 2018