You are on page 1of 13

|

Received: 6 October 2020    Accepted: 9 November 2020

DOI: 10.1111/pim.12808

COMMISSIONED REVIEW ARTICLE

Acquired tick resistance: The trail is hot

Sukanya Narasimhan1  | Cheyne Kurokawa1 | Melody DeBlasio1 | Jaqueline Matias1 |


Andaleeb Sajid1 | Utpal Pal2 | Geoffrey Lynn1 | Erol Fikrig1

1
Section of Infectious Diseases, Department
of Internal Medicine, Yale University School Abstract
of Medicine, New Haven, CT, USA Acquired tick resistance is a phenomenon wherein the host elicits an immune re-
2
Department of Veterinary Medicine,
sponse against tick salivary components upon repeated tick infestations. The immune
University of Maryland, College Park, MD,
USA responses, potentially directed against critical salivary components, thwart tick feed-
ing, and the animal becomes resistant to subsequent tick infestations. The develop-
Correspondence
Sukanya Narasimhan, Section of Infectious ment of tick resistance is frequently observed when ticks feed on non-natural hosts,
Diseases, Department of Internal Medicine,
but not on natural hosts. The molecular mechanisms that lead to the development of
Yale University School of Medicine, New
Haven, CT 06420, USA. tick resistance are not fully understood, and both host and tick factors are invoked
Email: sukanya.narasimhan@yale.edu
in this phenomenon. Advances in molecular tools to address the host and the tick
Funding information are beginning to reveal new insights into this phenomenon and to uncover a deeper
NIH/NIAID, Grant/Award Number:
understanding of the fundamental biology of tick-host interactions. This review will
AI126033, AI128182 and AI138949; Steven
and Alexandra Cohen Foundation focus on the expanding understanding of acquired tick resistance and highlight the
impact of this understanding on anti-tick vaccine development efforts.

KEYWORDS

acquired tick resistance, anti-tick vaccine, basophils, immune modulation, Ixodes

1 |  I NTRO D U C TI O N weight loss and anaemia, and this impacts milk and meat production
resulting in significant economic losses to the animal industry.8 From
Ticks are obligate haematophagous ectoparasites distributed world- the veterinary disease perspective, controlling tick densities at spe-
wide and serve as vectors of human and animal diseases.1 The focus cific localities is, therefore, highly valuable. This is exemplified by the
of this review is on Ixodid or hard ticks, one of three families within success of the Bm86 protein subunit vaccine, the only commercially
the suborder Ixodida that includes about 700 species. 2 Ixodid ticks available anti-tick vaccine, based on a gut protein of Rhipicephalus
are vectors of numerous human and livestock diseases.3 Since the microplus that impairs tick feeding and significantly reduces R. micro-
bite of a tick is the only route of natural transmission of tick-borne plus infestations on cattle.9
pathogens, several strategies have been explored over the last few While reducing tick populations in endemic areas also impacts
decades to prevent getting bitten by ticks. Acaricides, a first-line human disease prevalence by reducing the probability of tick en-
strategy used to control tick populations in endemic areas, may be counters, vaccines that can efficiently prevent disease transmission
detrimental to the environment, and ticks rapidly develop resistance are also desired.10 Traditionally, transmission-blocking vaccines have
4,5
to acaricides making them ineffective. Biocontrol measures using largely relied on targeting the pathogen, and not the vector. A vac-
entomopathogenic fungi have also shown promise in controlling di- cine to prevent tick-transmitted tick-borne encephalitis virus infec-
verse Ixodid tick populations.6 However, the logistical difficulties in tion of humans is available for human use in Europe.11 The vaccine
the implementation of biocontrol strategies are hampered by envi- is an antigen extract derived from in vitro grown TBE virus that is
ronmental variables including temperature and humidity. Vaccines filtered and inactivated in formaldehyde.12 In the USA, a vaccine to
that can target and impair tick feeding or fecundity have been shown prevent Lyme disease based on OspA, a surface antigen of the Lyme
to be effective at controlling tick populations.7 Tick infestations of disease agent, was approved for human use in 1998,13,14 but was
livestock animals result not just in disease transmission, but also in withdrawn from the market in 2002 by the manufacturer.15 Global

Parasite Immunology. 2021;43:e12808. wileyonlinelibrary.com/journal/pim © 2020 John Wiley & Sons Ltd |
    1 of 13
https://doi.org/10.1111/pim.12808
|
2 of 13       NARASIMHAN et al.

warming is affecting the spread of diverse vectors of human and saliva is secreted at continuous intervals over the course of the 4-
16-18
livestock pathogens, including ticks and tick-borne pathogens. to 10-day feeding period typical for Ixodes complex ticks. In order
In the past two decades alone, several newly recognized tick-borne to remain stably tethered to the host over the prolonged feeding
pathogens have been recognized in the Western hemisphere.19 period, the tick secretes an adhesive cement within hours of at-
Therefore, there is an urgent need to develop vaccine strategies tachment to the host.36 Tick cement is shown to be composed of
that would simultaneously reduce tick populations and also prevent a mixture of proteins, lipids, amino acids such as glycine, serine and
transmission of tick-borne pathogens to mammalian hosts. Towards tyrosine, and carbohydrates37 that together form a viscous gel-like
this goal, several research efforts are focused on identifying tick cone composed of a core cement that fits snugly around the hypos-
antigens that may serve as effective vaccine targets to thwart tick tome and a cortical cement secreted outside the core cement.37
feeding and consequently also prevent pathogen transmission. The Providing an impenetrable physical barrier to the tick mouth parts
sequencing of transcriptomes and genomes of multiple tick species is thought to be one of the prime functions of tick cement.38 Until
20-24
in the last decade coupled with technological advancements a decade or so ago, only a descriptive understanding of tick cement
have added critical molecular tools25-27 and collectively accelerated was available.39,40 Advances in molecular techniques, and the avail-
our efforts to gain functional insights into the tick genome. ability of an artificial feeding system for ticks have helped circum-
While we have overcome the paucity of genomic data that vent qualitative and quantitative limitations in cement research.37
plagued tick research a decade ago, the task of sifting through these Cement-specific proteins have been identified predominantly from
meta-datasets to identify critical antigens that may serve as vaccine Rhipicephalus species41-44 and from Amblyomma species45,46 with
targets remains a daunting challenge. Providing a functional para- potential antimicrobial and antihaemostatic activities. A cement an-
digm to address this challenge is the phenomenon of acquired tick tigen from Rhipicephalus appendiculatus named 64P41 was shown to
28
resistance or ATR. Seminal observations by William Trager in 1939 be represented in multiple Ixodid tick species47 with the potential to
showed that nonpermissive hosts mount a robust immune response serve as a broad-spectrum anti-tick vaccine. Further progress in the
to critical tick salivary antigens, and thwart tick feeding. Research molecular understanding of tick cement may reveal novel strategies
efforts to identify salivary proteins targeted by ATR have yielded a to thwart tick feeding.
diverse list of salivary antigens with functions relevant to tick feed- Tick saliva contains an array of immunomodulators, anticoagu-
ing. 29-32 However, immunity elicited against these antigens individ- lants and haemostatic compounds, which allow the tick to manipulate
ually or as subset of cocktails have only partially recapitulated ATR. and maintain the feeding site and subvert host defences,48-50 and is
It is clear that the molecular basis of this phenomenon that has re- pivotal for the tick to feed to repletion. An overview of the predom-
mained a puzzle33,34 poses a bottleneck in our ability to fully exploit inant functions elaborated in the saliva of hard ticks is provided in
this robust paradigm towards defining tick salivary vaccine targets. Figure 1. The first step in haemostasis is the development of a plate-
This review will examine our current and expanding understand- let plug, and salivary proteins that interfere with this process are
ing of ATR, and highlight how this understanding might reveal key secreted by multiple hard tick species. Prostacyclin51 and the serine
events at the tick-host interface that enable or disable tick feeding. protease inhibitor (serpin) IxscS-1E152 from Ixodes scapularis, IRS-2
This understanding will educate the prioritization of salivary anti- from Ixodes ricinus,53 R. microplus serpins, RmS-3 and RmS-17,54 and
gens that may be vaccine targeted and guide the development of an Variabilin from Dermacentor variabilis55 disrupt platelet aggregation.
anti-tick vaccine. In addition to platelet aggregation, multiple pro-coagulation factors
are activated by the host to prevent blood loss. Several I. scapularis
salivary proteins that inhibit various steps of coagulation have been
2 |  TI C K SA LI VA RY PROTEO M E: identified including Salp14,56 Innonexin,57 TIX-5,58 Ixolaris59 and
PA R A M O U NT FO R TI C K FE E D I N G Penthalaris.60 Other anticoagulants from Ixodes ticks include metal-
loproteases,61-64 various serpins,52,65,66 Ir-CPI,67 Iris68 and Rhippilin-1
Hard ticks (Ixodidae) are obligate haematophagous arthropods and and -2 from Rhipicephalus hemaphysaloides.69 Haemaphysalis ticks
obtain their blood meal by attaching to the skin of vertebrate hosts, have a particularly large number of demonstrated and putative
tearing the skin and feeding from the pool of blood formed at the thrombin inhibitors, including: madanin-1 and -2,70,71 chimadanin,72
bite site. Successful haematophagy is central to the completion of and the serpin HLS173 from Haemaphysalis longicornis; haemathrin
the life cycle of the hard tick. Since the life cycles of the pathogens from Haemaphysalis bispinosa74; and serpins HDS1 and HDS2 from H.
transmitted by hard ticks are entwined with that of the tick, there is doenitzi75 Iris from I. ricinus68 has also demonstrated anti-thrombin
significant impetus to gain a molecular understanding of how ticks activity, in addition to BmGTI,76 the serpin RmS-15,77 BmAP,78 and
acquire a bloodmeal. Tick attachment to the host involves tearing microphilin79 from R. microplus. Haemaphysalis proteins longistatin80
the skin, which is accomplished by a pair of barbed, articulated cheli- and enolase81 stimulate this pathway by activating plasminogen that
cerae at the tip of the mouthparts. The chelicerae flex and retract, negatively regulates the coagulation cascade.
inserting the hypostome and pushing the tick further into the host Histamine released at the bite site by platelets, and mast cells
until the mouthparts are completely embedded into the dermis of can be detrimental to tick attachment by inducing pain and itch re-
the skin.35 Slicing mouthparts create a feeding lesion into which sponses at the bite site that eventually result in the host grooming
NARASIMHAN et al. |
      3 of 13

F I G U R E 1   Hard tick attached to host skin and secreting pharmacologically active salivary components into the feeding lesion. A, Ixodid
ticks attach to the host skin by inserting their hypostome into the dermis of the host skin and adhere firmly with the help of salivary cement
that is deposited at the bite site and around the hypostome. Saliva secreted into the feeding lesion thwarts defence responses of the host.
SG, salivary glands; Mc, macrophage; N, neutrophils; Lc, Langerhans cells; Dc, dendritic cells; E, eosinophils; Mt, mast cells; B, B cells; L,
lymphocytes; Cg, procoagulants; C, complement factors. B, Brief overview of predominant salivary functions characterized in tick saliva.
Percent calculations based on the literature surveyed in this review. Illustration created using Biorender.com and Adobe Photoshop CC

the tick off.33,82 Inhibiting histamine-related inflammation is there- proteins Salp20 and Isac113-116; I. ricinus proteins IRAC I and II117,118
83
fore essential for the tick to remain attached to the host. Multiple and R. pulchellus protein Cirp-T119 interrupt the alternative pathway.
histamine binding proteins have been identified in the saliva of R. ap- Tick Salivary Lectin Pathway Inhibitors in I. scapularis and I. ricinus
pendiculatus,83 D. reticulatis,84 Ixodes persulcatus85 I. ricinus86 and interfere with the lectin pathway.120,121 Recent reports suggest that
I. scapularis.61 Ticks also maintain their feeding lesion by interfering Amblyomma species may encode functions that also impair the clas-
with host immune responses. Pro-inflammatory cytokines such as sical pathway.50 Additionally, antioxidant salivary proteins have been
IL-1β, IL-6 and TNF-ɑ are down-regulated by salivary proteins such described in Ixodid ticks and function to quench reactive oxygen and
as sialostatins (L, L2, Ip-sL1 and Ip-sL2),87-89 Salp15,90 Iris,91,92 Isac,93 nitrogen species generated by immune cells such as neutrophils and
Ir-SPI,94 IRS-295 and Iristatin.96 Other anti-inflammatory salivary pro- macrophages that migrate to the tick bite site. 29,122-124
97 98 99
teins include serpins, Hl-p36 and Longistatin ( from H. longicor- It is important to note that not all salivary proteins are expressed
nis; PGE2 from D. variabilis,100 and evasins from R. sanguineas.101,102 and secreted throughout the course of feeding. The tick sialome is
Interestingly, Japanin from R. appendiculatus and PGE2 from D. vari- dynamic, and the composition changes over time,125,126 potentially
abilis and I. scapularis100,103,104 upregulate the anti-inflammatory orchestrated by the different phases of tick feeding.127 Tick salivary
cytokine IL-10. A sphingomylenase enzyme activity in I. scapularis protein functions are also exploited by pathogens as they transit to
saliva was shown to promote IL-4 production by CD4-T cells.105 and from the host skin.106,128,129 Therefore, it is logical to envision
These functions of the tick sialome are suggested to skew the host that salivary molecules may serve as viable vaccine targets to ab-
immune response towards a Th2 profile that may be advantageous rogate tick feeding and prevent pathogen transmission. Temporal
to the survival of tick-borne pathogens during transmission and is changes in the salivary composition, in conjunction with the func-
a key element of saliva-assisted pathogen transmission.106 Tick sia- tional complexity, and redundancy of the tick sialome have rendered
lome also encodes functions that prevent immune cell activation and the search for salivary vaccine targets a task akin to searching for the
proliferation91,94,96-98,104,107-112 proverbial “needle in the haystack”. An opportunity to circumvent
Salivary proteins also impair the host complement system that this challenge has come from the phenomenon of ATR, originally de-
represents a major innate immune response triggered by microbes, scribed by William Trager in 1939. 28 Since ATR results in thwarting
cell damage, antigen-antibody complex, and glycans and lead to tick feeding and pathogen transmission, expanding a molecular and
increased inflammation detrimental to tick feeding. I. scapularis mechanistic understanding of ATR may offer a robust paradigm to
|
4 of 13       NARASIMHAN et al.

define the critical subset of salivary proteins that may be vaccine Acquired tick resistance has been observed in C57BL/6 mice re-
targeted. peatedly infested with H. longicornis larvae.137 Larger mammals such
as deer, horse, and cattle, and birds are the natural hosts for H. longi-
cornis, whereas mice are not natural hosts. Cattle have been shown
3 |  A N I M A L M O D E L S O F ACQ U I R E D TI C K to develop tick resistance following repeated exposures to R. micro-
R E S I S TA N C E plus. Importantly, ATR impaired Babesia transmission to cattle.146
Unlike guinea pigs, mice and rabbits, generation of tick immunity in
Trager, in 1939, 28 showed that upon repeated tick infestations of cattle is dependent on the breed of cattle.147 Bos taurus indicus cat-
guinea pigs with D. variabilis nymphs, animals developed an im- tle develop immunity to R. microplus, whereas Bos taurus taurus do
mune response that was potent enough to derail subsequent tick not develop immunity.147 Several studies have attempted to identify
challenges. Tick-immune animals rapidly rejected ticks within the the genetic markers that could account for the differences, but, no
first 24 hours of tick attachment. This phenomenon of ATR is char- clear markers have emerged.148 Histological analysis of the bite site
acterized by visible erythema at the tick bite site due to the rapid revealed robust infiltration of basophils, mast cells and eosinophils
recruitment of immune cells, predominantly composed of basophils in the skin of the resistant Bos taurus indicus cattle. In comparison,
and eosinophils and hence termed cutaneous basophilic hypersensi- the bite site of permissive Bos taurus taurus cattle was primarily infil-
tivity in guinea pigs,130 also known as Jones-Mote hypersensitivity trated by neutrophils.39,149,150
in humans.131 The immune responses recruited to the tick bite site Evidence for tick immunity in humans is primarily anecdotal.
on tick-resistant animals are thought to be detrimental to tick feed- There have been reported cases of individuals developing hyper-
ing resulting in early tick detachment and decreased engorgement sensitivity reactions at the tick bite site, similar to the observation
weights. Similar observations with additional tick species, as well as in tick-immune animals.151,152 Additionally, people with frequent
the generation of ATR in rabbits, mice and cattle132-135 have dem- exposures to tick bites have been shown to develop antibodies to
onstrated that the phenomenon of ATR is widespread in tick-host tick proteins.153,154 Importantly, individuals that report itching at
interactions. the tick bite site may have a decreased chance of acquiring Borrelia
Ticks deposit a diverse set of salivary proteins into the host skin burgdorferi.155 Collectively, these results suggest that prior expo-
in order to facilitate acquisition of a blood meal, as outlined above. It sure to ticks and the development of immunity towards tick pro-
is generally believed that when ticks feed on non-natural hosts, the teins could potentially trigger itching at the bite site and grooming
host mounts a robust immune response towards the deposited sal- to remove ticks early. Since B. burgdorferi is transmitted only after
ivary proteins.33 Attesting to this hypothesis, experiments wherein 24-36 hours of tick attachment,156,157 provoking the immunologi-
animals were immunized with salivary gland extract or saliva have cal parameters of ATR upon tick attachment may offer an effective
shown that immunity against salivary components is indeed capable strategy for preventing tick transmission of Lyme disease.125,141
of eliciting several parameters of tick resistance including erythema Pathogens such as Babesia microti, Anaplasma phagocytophilum
at the bite site, and impaired feeding.136 It is, however, important to and Powassan virus that reside primarily in the salivary glands may
note that none of the effects were as robust as natural ATR. These be transmitted within 24 hours or earlier.158,159 Whether ATR may
immune responses potentially neutralize salivary functions essential impact the transmission of these tick-borne pathogens remains to
for feeding, and salivary functions essential for keeping detrimental be seen.
cells at bay. Activation and degranulation of basophils are thought
to result in the release of basophil components including histamine
that is noxious to the tick during the early stages of feeding.82,137 4 | M O LECU L A R BA S I S O F ACQ U I R E D
Whether basophilic components released upon degranulation im- TI C K R E S I S TA N C E
pairs tick feeding due to host responses such as vasodilation, pain
and itching or whether it enters the tick gut and causes damage to The mechanisms mediating tick rejection are complex and involve
the organ is not clear. multiple components of the host's adaptive immune system. As de-
Guinea pigs have been shown to acquire tick immunity follow- scribed above, cattle and guinea pigs readily acquire tick immunity
ing repeated tick infestations with Amblyomma americanum,138,139 following multiple tick infestations and have been used extensively
Rhipicephalus species,140 I. scapularis125,141 and Dermacentor ander- to examine the tick bite site.143,160,161 Although the reagents re-
142
soni. Resistance has been observed at the larval, nymphal and quired to characterize the immune response in guinea pigs and cat-
adult stages. Guinea pigs and rabbits support I. scapularis feeding, tle are not fully available, several studies have identified that this is
but they are not the natural host species. Peromyscus leucopus are dependent on both cellular and humoral responses. Repeated tick
the natural host for I. scapularis larvae and nymphs and do not de- infestation results in robust immune cell infiltration to the bite site.
143
velop tick immunity following repeated tick infestations. Similarly, The bite site of I. scapularis immune guinea pigs is heavily infiltrated
laboratory mouse models also do not acquire tick resistance after by heterophils and macrophages by day 2, followed by an intense in-
repeated I. scapularis tick infestations by mechanisms that are not filtration of leucocytes on days 3 and 4.143 Infiltrating basophils have
understood.144,145 also been observed at the tick bite site in guinea pigs repeatedly
NARASIMHAN et al. |
      5 of 13

infested with D. andersoni and A. americanum,160,162,163 and at the tick feeding. In guinea pigs, repeat tick infestation with I. scapularis
82,161
bite site of tick-immune cattle and mice. results in epicutaneous erythema, severe epidermal hyperplasia, oe-
Basophils are a major source for histamine upon activation and dema and hyperkeratosis at the tick bite site.143 Similar results to
have an important role during tick rejection. Additionally, degranu- dermal tissue have been observed in guinea pigs exposed to multiple
lation of basophils plays an important role in the recruitment of eo- infestations with Rhipicephalus sanguineus and A. americanum.160,171
sinophils to the bite site, although the role of eosinophils requires Unlike guinea pigs, Peromyscus leucopus fail to acquire tick immunity
further investigation.139 In a study to define the role of basophils, following repeat exposures to I. scapularis nymphs.143 Examination
the authors demonstrated that tick rejection was abolished when of the bite site in P. leucopus revealed minimal disruption to the der-
immune guinea pigs were administered anti-basophil sera prior to mal tissue following repeat tick infestation.143 The observed changes
139
challenge with A. americanum. However, anti-histamines admin- to dermal tissue are speculated to contribute towards tick rejection
istered at the time of challenge with A. americanum demonstrated in guinea pigs, whereas the lack of dermal changes in P. leucopus
that the critical role of basophils occurs through a histamine-in- could support repeat tick feeding.
dependent mechanism.130 This is in contrast to tick rejection with Basophils were mistakenly thought to be absent in mice,172 until
other tick species that demonstrated that anti-histamines delivered mouse basophils were described by Dvorak et al.173 Over the last
to tick-immune guinea pigs, mice and cattle ablate tick immunity to- two decades, an understanding of the enigmatic role of basophils in
wards R. microplus, D. andersoni and H. longicornis.82,164,165 Overall, Th2-immunity has started to unravel174-176 and these studies have
these results suggest that basophils are important for immunity also provided insights into the possible mechanisms of ATR. While
against multiple tick species; however, the mechanism of restriction the lack of immunological reagents has hampered our ability to
can be species-specific. dissect the molecular basis of ATR using the guinea pig and cattle
Although degranulation of basophils has been demonstrated to model, this has been circumvented using the mouse model of ATR
be critical for tick rejection, the mechanism of degranulation is not that is elicited by repeated infestations with H. longicornis.137 Using
understood. Salivary proteins deposited into the skin during feed- mice expressing the diphtheria toxin receptor under the control of
ing generate homocytotropic antibodies, that is antibodies that basophil-specific mast cell protease 8 promoter, Wada et al137 se-
only engage with cells of the same or closely related species, bind lectively ablated basophils by administering diphtheria toxin to the
predominantly to mast cells and basophils, resulting in degranula- animals after the first tick infestation and showed that basophils play
166
tion. Cutaneous basophil hypersensitivity responses have been a nonredundant role in eliciting ATR to H. longicornis nymphs. Ohta
proposed to be mediated by IgG1 antibodies in guinea pigs resis- et al177 then demonstrated that basophil recruitment is promoted by
166 137
tant to A. americanum, although Wada et al show that IgE an- IL-3 secreted by CD4+ memory T cells that arrive at the skin after a
tibodies play a role in rejection of H. longicornis ticks. Clearly, the tick challenge. Dissecting the mechanism of ATR, Tabakawa et al82
role of antibodies in mediating tick rejection is complex and varies showed that histamine released by skin-infiltrating basophils, but
depending on the life stage of the ticks, host species and the tick not by skin-resident mast cells, promotes H. longicornis rejection.
species.142 Passive transfer of lymph node cells, but not serum, from The exact mechanism/s by which histamine promotes tick rejection
guinea pigs resistant to D. andersoni larvae conferred resistance to remains to be deciphered. While the molecular understanding of
167
naïve guinea pigs. Similar results have also been observed with H. longicornis-provoked ATR in mice is beginning to clarify, given the
passive transfer of lymph node cells from guinea pigs resistant to heterogeneity in the immunological responses of host species to tick
A. americanum larvae.162 However, passive transfer with sera from species and stages, a single unifying mechanism may not emerge.
guinea pigs infested with adult D. andersoni was able to confer pro-
tection.142 These results suggest that the life stage of the tick may
also be important for the induction of protective antibody titres.142 5 | D I C H OTO M O U S I M M U N E R E S P O N S E S
The role of complement has also been demonstrated to play an TO TI C K B ITE S
important role during tick rejection. Resistance to D. andersoni lar-
vae in guinea pigs was ablated when the animals were administered Although ticks can feed on diverse hosts, each tick species appears
cobra venom factor, a protein which depletes serum complement, to have a host preference in nature that is likely determined by a
prior to tick challenge.168,169 This was further evaluated using C4- combination of host, tick and ecological factors. In general, pre-
deficient guinea pigs, which are deficient in the classical complement ferred hosts serve as reservoir hosts for the specific tick and are
170
activation pathway. In this study, C4-deficient guinea pigs were able to host multiple infestations of the tick species without devel-
able to acquire resistance to D. andersoni larvae, suggesting that the oping any immune responses detrimental to tick feeding. This char-
alternate pathway of complement activation is important for tick acteristic is essential in order to ensure the successful completion
rejection.170 This is also consistent with the observations that tick of the tick's life cycle. For example, I. scapularis nymphs and larvae
salivary proteomes, in general, do not contain proteins to inhibit the predominantly feed on P. leucopus, the reservoir host.178 Expectedly,
classical pathway of complement. in summer and fall when nymphs and larvae are active, P. leucopus
The immune reaction at the tick bite site of immune animals can are fed upon multiple times by these ticks without any evidence of
induce significant changes in the skin, which may play a role to limit ATR. Mus musculus has served as a laboratory model of reservoir
|
6 of 13       NARASIMHAN et al.

host for I. scapularis studies despite the fact that P. leucopus and exposing them to specific host skin semiochemicals of rabbit, dog
M. musculus belong to different genera and M. musculus is not the or human was indeed different. This iterated that the tick salivary
natural reservoir host.178 The genetic traits that allow both P. leuco- composition is not just temporally modulated, but also modulated
pus and M. musculus species to host I. scapularis ticks without devel- by host skin components. Narasimhan et al's study145 showed that
oping ATR are not known. Guinea pigs and rabbits are non-natural mouse splenocytes incubated with mouse-fed salivary gland ex-
hosts for I. scapularis that readily develop ATR. This dichotomy in tracts elicited significantly less IL-4, a Th2-defining cytokine, when
immune response to tick feeding is thought to be multifactorial as compared to the amounts elicited by guinea pig-fed salivary gland
summarized in Figure 2. Proposing a lock and key hypothesis to ex- extracts. Consistent with this, Franzin et al181 showed that R. mi-
plain this dichotomy, Ribeiro suggested that tick salivary molecules croplus fed on tick-susceptible Bos taurus taurus expressed signifi-
have co-evolved with reservoir hosts like P. leucopus and geared to cantly increased amounts of salivary transcripts for genes encoding
efficiently engage with and diffuse adaptive immune responses of immune modulators such evasins, and metalloproteases, when
P. leucopus.179 Conversely, I. scapularis salivary components engage compared to that in ticks fed on tick-resistant Bos taurus indicus.
poorly with immune components of non-natural hosts, hence un- This differential expression of secreted salivary immunomodulators
able to thwart host adaptive immune responses. In essence, natural/ could account for differential evasion of host defence responses.
permissive hosts do not have an immunological memory of tick bites, Therefore, host-modulated differences in the tick sialome composi-
while non-natural/nonpermissive hosts do. tion could inadvertently account, in part, for functional differences
An explanation that does not exclude the lock and key hypoth- critical to dampen host immune responses. A detailed characteri-
esis, but adds another facet to the dichotomous immune response zation of the qualitative differences between tick-susceptible host-
on natural and non-natural host is that the tick sialome is differ- fed and tick-resistant host-fed tick sialomes would offer molecular
ent when it feeds on different hosts. Narasimhan et al145 compared insights into ATR.
the salivary composition of I. scapularis nymphs fed on M. muscu- The dichotomous immune response to tick feeding could also be
lus, the laboratory model of natural host, and on guinea pigs, the driven by the inherent structural and immunological differences be-
model non-natural host and observed that the sialome composi- tween the guinea pig and mouse skin. While the epidermis, dermis
tion is host-specific. Tirloni et al180 also observed that the protein and hypodermis are well defined and thick in the guinea pig, these
composition of adult I. scapularis and A. americanum stimulated by layers are thin in mice.182,183 Since the mouse skin is covered by fur,
the fat-laden hypodermis that normally provides thermal homoeo-
stasis in guinea pig and human skin184 is significantly reduced in the
murine skin.183 Mice have an additional subcutaneous layer called
panniculus carnosus, an extra layer of muscle that is thought to allow
wound healing via contraction resulting in little or no scarring.185
While guinea pigs also have the panniculus carnosus layer, wound
healing may occur via re-epithelization leading to scar formation186
and could account for the epidermal hyperplasia and hyperkerato-
sis observed at the I. scapularis bite site on guinea pigs, but not on
mice.143 While the immunology of the guinea pig skin is not fully
understood, presumably it behaves more like the human skin than
the murine skin. The Langerhans cells, the dendritic cell subset of the
epidermis, in mice are not critical for priming CD8+ T cells, unlike in
humans.187,188 Lymphocytes in the murine epidermis are predomi-
nantly populated by γδT cells, while in humans it is predominantly
αβT cells.189
Although paucity of immunological tools to examine traditional
animal models of ATR such as the guinea pig and cattle has sty-
mied progress in this research, Franzin et al,181 More et al190 and
F I G U R E 2   Potential mechanisms of acquired tick resistance.
Kurokawa et al191 have harnessed the power of rapidly evolving
Factors that may drive the dichotomous immune responses to tick
bites on permissive or resistant host species include the following: next-generation RNA sequencing tools to examine host compo-
optimal (A) or suboptimal (A’) engagement of salivary proteins nents at the tick bite site that may serve as molecular drivers of
with host defence responses; genetic predisposition to decreased ATR. Franzin et al181 underscore the genetic predisposition to
(B) or increased (B’) inflammatory responses to salivary proteins; resistance or susceptibility and show that R. microplus-resistant
structural and immunological differences in the skin (C, C’); host-
B. taurus indicus have a higher baseline expression of genes encod-
specific salivary proteome that is sufficient (D) or deficient (D’)
ing pro-inflammatory cytokines. Consistent with histological ex-
in modulating host defence responses; and differences in wound
healing without scar (E) or with scar (E’) formation. Illustration amination of tick bite sites of resistant animals, the tick-resistant
generated using Biorender.com breed of B. taurus demonstrated increased expression of basophil
NARASIMHAN et al. |
      7 of 13

and T lymphocyte recruiting cytokine CCL2 when compared to Coast region of the United States, while Lyme spirochetes are
tick-sensitive animals. More interestingly, their study showed transmitted by I. scapularis in other geographic parts of the United
that the skin of the susceptible breed expresses higher levels of States. In Europe and Eurasia, the castor bean tick, I. ricinus is the
enzymes involved in detoxification and this also generates vola- primary vector for Lyme spirochetes. Cross-protective immunity
tile metabolites that serve as semiochemicals that are attractive could also potentially be useful in areas where multiple medically
to ticks. More et al190 showed that upon repeated infestations of important tick species occur in close proximity and parasitize the
tick-resistant breed of B. taurus with R. microplus expression of same hosts. This could include the eastern United States, where
genes involved in skin remodelling and in basophil activation were I. scapularis, D. variabilis, and A. americanum frequently co-exist
increased at the bite site when compared to that in tick-suscep- and utilize some of the same host species, including white-tail deer
tible breed. Further, transcripts encoding for CCL13, a cytokine (Odocoileus virginianus). Therefore, a molecular understanding of
invoked in eosinophil recruitment was upregulated at the tick bite salivary proteins that are involved in cross-protective immunity
site. This was consistent with the findings of Kurokawa et al191 would help define and prioritize vaccine targets that may serve to
that eosinophils are increased at the tick bite sites on tick-resistant simultaneously impair feeding and fecundity of multiple tick spe-
192
guinea pigs. Further, Robbertse et al addressed lymphocyte cies in endemic regions. It is conceivable that a broad-spectrum
populations from the skin and draining lymph nodes of resistant anti-tick vaccine applied to wild deer could limit the density of
and susceptible B. taurus after challenge with R. microplus and tick populations, potentially reducing enzootic transmission of
showed that the numbers of B lymphocytes and T-helper lympho- Borrelia, Anaplasma, Ehrlichia, Rickettsia and Babesia species trans-
cytes were decreased in the lymph nodes of tick-susceptible ani- mitted by these ticks. Such an approach would also be operation-
mals. Increased variability in WC1 + γδ T lymphocytes populations ally cost-effective.
was also associated with increased susceptibility to R. microplus
ticks. Kurokawa et al191 demonstrated that FCεRI-signalling, com-
plement pathways and pro-coagulation pathways are activated in 7 | CO N C LU S I O N S
the guinea pig skin, but not in mice skin upon repeated infestations
with I. scapularis nymphs. The phenomenon of ATR to ticks remains a puzzling facet of tick-
These genetic, transcriptomic and immunological studies of host interactions, since its first description almost 80 years ago. 28
host responses to tick bites have begun to reveal new insights into Technological advancements in molecular tools to address tick
the differential responses to tick bites on susceptible and resistant and host functional genomes, transcriptomes and immunomes
hosts. However, an understanding of what components of the tick have provided the much-needed momentum to unravel a new un-
saliva drive these differential responses on different host species derstanding of this phenomenon. A molecular understanding of
will be essential to develop strategies to prevent tick feeding and this phenomenon will enhance approaches to define the subset of
tick transmission of pathogens. An interesting facet of ATR that may antigens that may serve as potential vaccine targets. Translating
additionally help to shed light on the molecular mechanisms of ATR insights from ATR to tick vaccine development will also be ac-
is the observation that ATR against one tick genus may be cross-pro- celerated by recent advancements in vaccine delivery platforms.
tective against another related tick genus. Although protein-based vaccines have been the mainstay of
vaccinology for the last century,196 the rapidly evolving science
of vaccinomics has highlighted the exciting possibility of using
6 | ATR- M E D I ATE D C ROS S - PROTEC TI O N DNA197-199 and mRNA 200 -based vaccines to deliver nucleotide se-
quences encoding target antigens into the host. This enables the in
Cross-protective immunity is the phenomenon wherein ATR vivo generation of recombinant antigens by the host translational
against a tick species of primary exposure confers host resistance machinery and immunological presentation of these antigens
against a secondary tick species for which the host has no prior elicit B and T cell-mediated responses critical for effective and
28,193,194
exposure. Numerous tick antigens are conserved among long-lasting immunity. 200-202 These approaches may circumvent
195
hard tick species including many salivary proteins and could, in the cumbersome process of optimization of protein production
part, explain the acquisition of cross-protective immunity. This is and formulation and spur progress in tick vaccine development.
also likely the reason why immunity against truncated protein con- Importantly, deciphering a mechanistic understanding of ATR will
structs (64TRP) of the 64P cement antigen from R. appendiculatus reveal interesting facets of mammalian immunology, and of tick
was shown to induce immunity in rabbits and hamsters against biology that will transcend the field of tick research.
Ixodes ricinus, and R. sanguineus that impacted tick feeding and
mortality.47 Cross-species protection is important for several rea- AC K N OW L E D G E M E N T S
sons related to vaccine development against ticks. Closely related We are grateful to Ms Aine Kaminski for excellent technical assis-
tick species are frequently competent vectors of similar patho- tance during literature collation. This research was supported by
gens. For example, the western blacklegged tick, Ixodes pacificus, grants from the Steven and Alexandra Cohen Foundation, and the
transmits Lyme disease spirochetes (B. burgdorferi) in the West NIH (AI138949, AI126033 and AI128182).
|
8 of 13       NARASIMHAN et al.

AU T H O R C O N T R I B U T I O N S 17. Estrada-Pena A, Ostfeld RS, Peterson AT, Poulin R, de la Fuente J.


Effects of environmental change on zoonotic disease risk: an eco-
SN, CK, MD, JM, AS and GL surveyed the literature and drafted sec-
logical primer. Trends Parasitol. 2014;30(4):205-214.
tions of the manuscript. SN wrote the manuscript. CK, UP, AS and 18. Estrada-Pena A. Tick-borne pathogens, transmission rates and cli-
EF provided critical input. SN and EF reviewed and assembled the mate change. Front Biosci. 2009;14:2674-2687.
manuscript. 19. Rochlin I, Toledo A. Emerging tick-borne pathogens of public health
importance: a mini-review. J Med Microbiol. 2020;69(6):781-791.
20. Chmelar J, Kotál J, Karim S, et al. Sialomes and mialomes: a sys-
PEER REVIEW tems-biology view of tick tissues and tick-host interactions. Trends
The peer review history for this article is available at https://publo​ Parasitol. 2016;32(3):242-254.
ns.com/publo​n/10.1111/pim.12808. 21. Artigas-Jeronimo S, De La Fuente J, Villar M. Interactomics and
tick vaccine development: new directions for the control of tick-
borne diseases. Expert Rev Proteomics. 2018;15(8):627-635.
DATA AVA I L A B I L I T Y S TAT E M E N T
22. Nene V. Tick genomics–coming of age. Front Biosci.
Data cited in this review are published and available online or upon 2009;14:2666-2673.
request from the authors of the respective publications. No unpub- 23. Kotsyfakis M, Kopáček P, Franta Z, Pedra JHF, Ribeiro JMC. Deep
lished data are included in this manuscript. sequencing analysis of the Ixodes ricinus Haemocytome. PLoS Negl
Trop Dis. 2015;9(5):e0003754.
24. Jia N, Wang J, Shi W, et al. Large-scale comparative analyses of tick
ORCID genomes elucidate their genetic diversity and vector capacities.
Sukanya Narasimhan  https://orcid.org/0000-0003-0396-8757 Cell. 2020;182(5):1328-1340 e13.
25. Ramakrishnan VG, Aljamali MN, Sauer JR, Essenberg RC.
Application of RNA interference in tick salivary gland research. J
REFERENCES
Biomol Tech. 2005;16(4):297-305.
1. Sonenshine DE. The Biology of Tick Vectors of Human Disease. In: 26. Kurtti TJ, Mattila JT, Herron MJ, et al. Transgene expression and
Goodman JL, Dennis DT, Sonenshine DE, eds. Tick-Borne Diseases silencing in a tick cell line: a model system for functional tick ge-
of Humans. Washington, DC: ASM Press; 2005:401. nomics. Insect Biochem Mol Biol. 2008;38(10):963-968.
2. Anderson JM, Ammerman NC, Norris DE. Molecular differen- 27. Oliver JD, Chávez ASO, Felsheim RF, Kurtti TJ, Munderloh UG. An
tiation of metastriate tick immatures. Vector Borne Zoonotic Dis. Ixodes scapularis cell line with a predominantly neuron-like pheno-
2004;4(4):334-342. type. Exp Appl Acarol. 2015;66(3):427-442.
3. Jongejan F, Uilenberg G. The global importance of ticks. 28. Trager W. Accquired immunity to ticks. J. Parasitology.
Parasitology. 2004;129(Suppl):S3-S14. 1939;25:57-81.
4. George JE, Pound JM, Davey RB. Chemical control of ticks on cat- 29. Das S, Banerjee G, DePonte K, Marcantonio N, Kantor FS, Fikrig
tle and the resistance of these parasites to acaricides. Parasitology. E. Salp25D, an Ixodes scapularis antioxidant, is 1 of 14 immuno-
2004;129(Suppl):S353-S366. dominant antigens in engorged tick salivary glands. J Infect Dis.
5. Graf JF, Gogolewski R, Leach-bing N, et al. Tick control: an indus- 2001;184(8):1056-1064.
try point of view. Parasitology. 2004;129(Suppl):S427-S442. 30. Schuijt TJ, Narasimhan S, Daffre S, et al. Identification and char-
6. Perinotto WM, Angelo IC, Golo PS, et al. Susceptibility of differ- acterization of Ixodes scapularis antigens that elicit tick immunity
ent populations of ticks to entomopathogenic fungi. Exp Parasitol. using yeast surface display. PLoS One. 2011;6(1):e15926.
2012;130(3):257-260. 31. Shapiro SZ, Voigt WP, Fujisaki K. Tick antigens recognized by
7. de la Fuente J, Kocan KM. Strategies for development of vaccines for serum from a guinea pig resistant to infestation with the tick
control of ixodid tick species. Parasite Immunol. 2006;28(7):275-283. Rhipicephalus appendiculatus. J Parasitol. 1986;72(3):454-463.
8. Jonsson NN, Davis R, De Witt M. An estimate of the economic ef- 32. Rego ROM, Trentelman JJA, Anguita J, et al. Counterattacking the
fects of cattle tick (Boophilus microplus) infestation on Queensland tick bite: towards a rational design of anti-tick vaccines targeting
dairy farms. Aust Vet J. 2001;79(12):826-831. pathogen transmission. Parasit Vectors. 2019;12(1):229.
9. de la Fuente J, Almazán C, Canales M, et al. A ten-year review of 33. Wikel SK. Host immunity to ticks. Annu Rev Entomol. 1996;41:1-22.
commercial vaccine performance for control of tick infestations on 34. Willadsen P, Jongejan F. Immunology of the tick-host interaction
cattle. Anim Health Res Rev. 2007;8(1):23-28. and the control of ticks and tick-borne diseases. Parasitol Today.
10. Willadsen P.Anti-tickvaccines. Parasitology. 2004;129(Suppl):S367-S387. 1999;15(7):258-262.
11. Heinz FX, Stiasny K, Holzmann H, et al. Vaccination and tick-borne 35. Richter D, Matuschka F-R, Spielman A, Mahadevan L. How ticks
encephalitis, central Europe. Emerg Infect Dis. 2013;19(1):69-76. get under your skin: insertion mechanics of the feeding apparatus
12. Bogovic P, Strle F. Tick-borne encephalitis: a review of epidemiol- of Ixodes ricinus ticks. Proc Biol Sci. 2013;280(1773):20131758.
ogy, clinical characteristics, and management. World J Clin Cases. 36. Roe RM, Sonenshine ED, eds. Biology of Ticks. 1. 2nd Edition. New
2015;3(5):430-441. York, NY: Oxford University Press; 2014.
13. Embers ME, Narasimhan S. Vaccination against Lyme disease: past, 37. Suppan J, Engel B, Marchetti-Deschmann M, Nürnberger S. Tick
present, and future. Front Cell Infect Microbiol. 2013;3:6. attachment cement - reviewing the mysteries of a biological skin
14. Plotkin SA. Need for a new lyme disease vaccine. N Engl J Med. plug system. Biol Rev Camb Philos Soc. 2018;93(2):1056-1076.
2016;375(10):911-913. 38. Moorhouse DE, Tatchell RJ. The feeding processes of the cat-
15. Nigrovic LE, Thompson KM. The Lyme vaccine: a cautionary tale. tle-tick Boophilus microplus (Canestrini): a study in host-par-
Epidemiol Infect. 2007;135(1):1-8. asite relations. I. Attachment to the host. Parasitology.
16. Campbell-Lendrum D, Manga L, Bagayoko M, Sommerfeld J. 1966;56(4):623-632.
Climate change and vector-borne diseases: what are the implica- 39. Tatchell RJ, Moorhouse DE. The feeding processes of the cattle
tions for public health research and policy? Philos Trans R Soc Lond tick Boophilus microplus (Canestrini). II. The sequence of host-tis-
B Biol Sci. 2015;370(1665):20130552. sue changes. Parasitology. 1968;58(2):441-459.
NARASIMHAN et al. |
      9 of 13

40. Cowdry EV, Danks WBC. Studies on East Coast Fever. II. Behaviour scapularis: identification of factor X and factor Xa as scaffolds
of the parasite and the development of distinctive lesions in sus- for the inhibition of factor VIIa/tissue factor complex. Blood.
ceptible animals. Parasitology. 1933;25(1):1-63. 2002;99(10):3602-3612.
41. Havlikova S, Roller L, Koci J, et al. Functional role of 64P, 60. Francischetti IM, Mather TN, Ribeiro JM. Penthalaris, a novel re-
the candidate transmission-blocking vaccine antigen combinant five-Kunitz tissue factor pathway inhibitor (TFPI) from
from the tick, Rhipicephalus appendiculatus. Int J Parasitol. the salivary gland of the tick vector of Lyme disease, Ixodes scapu-
2009;39(13):1485-1494. laris. Thromb Haemost. 2004;91(5):886-898.
42. Mulenga A, Sugimoto C, Sako Y, et al. Molecular characteriza- 61. Valenzuela JG, Francischetti IM, Pham VM, Garfield MK, Mather
tion of a Haemaphysalis longicornis tick salivary gland-associated TN, Ribeiro JM. Exploring the sialome of the tick Ixodes scapularis.
29-kilodalton protein and its effect as a vaccine against tick infes- J Exp Biol. 2002;205(Pt 18):2843-2864.
tation in rabbits. Infect Immun. 1999;67(4):1652-1658. 62. Decrem Y, Beaufays J, Blasioli V, et al. A family of putative metal-
43. Bishop R, Lambson B, Wells C, et al. A cement protein of the tick loproteases in the salivary glands of the tick Ixodes ricinus. FEBS J.
Rhipicephalus appendiculatus, located in the secretory e cell gran- 2008;275(7):1485-1499.
ules of the type III salivary gland acini, induces strong antibody 63. Decrem Y, Mariller M, Lahaye K, et al. The impact of gene
responses in cattle. Int J Parasitol. 2002;32(7):833-842. knock-down and vaccination against salivary metalloproteases
44. Zhou J, Gong H, Zhou Y, Xuan X, Fujisaki K. Identification of a gly- on blood feeding and egg laying by Ixodes ricinus. Int J Parasitol.
cine-rich protein from the tick Rhipicephalus haemaphysaloides and 2008;38(5):549-560.
evaluation of its vaccine potential against tick feeding. Parasitol 64. Ali A, Tirloni L, Isezaki M, et al. Reprolysin metalloproteases from
Res. 2006;100(1):77-84. Ixodes persulcatus, Rhipicephalus sanguineus and Rhipicephalus mi-
45. Karim S, Ribeiro JM. An insight into the sialome of the lone star croplus ticks. Exp Appl Acarol. 2014;63(4):559-578.
tick, Amblyomma americanum, with a glimpse on its time depen- 65. Mulenga A, Khumthong R, Chalaire KC Ixodes scapularis tick serine
dent gene expression. PLoS One. 2015;10(7):e0131292. proteinase inhibitor (serpin) gene family; annotation and transcrip-
46. Bullard R, Allen P, Chao C-C, et al. Structural characterization of tional analysis. BMC Genom. 2009;10:217.
tick cement cones collected from in vivo and artificial membrane 66. Pichu S, Ribeiro JMC, Mather TN, Francischetti IMB. Purification
blood-fed Lone Star ticks (Amblyomma americanum). Ticks Tick of a serine protease and evidence for a protein C activator from
Borne Dis. 2016;7(5):880-892. the saliva of the tick, Ixodes scapularis. Toxicon. 2014;77:32-39.
47. Trimnell AR, Davies GM, Lissina O, Hails RS, Nuttall PA. A cross-re- 67. Decrem Y, Rath G, Blasioli V, et al. Ir-CPI, a coagulation con-
active tick cement antigen is a candidate broad-spectrum tick vac- tact phase inhibitor from the tick Ixodes ricinus, inhibits throm-
cine. Vaccine. 2005;23(34):4329-4341. bus formation without impairing hemostasis. J Exp Med.
48. Brossard M, Wikel SK. Tick immunobiology. Parasitology. 2009;206(11):2381-2395.
2004;129(Suppl):S161-S176. 68. Prevot PP, Adam B, Boudjeltia KZ, et al. Anti-hemostatic effects
49. Francischetti IM, Sá-Nunes A, Mans BJ, Santos IM, Ribeiro JMC. of a serpin from the saliva of the tick Ixodes ricinus. J Biol Chem.
The role of saliva in tick feeding. Front Biosci. 2009;14:2051-2088. 2006;281(36):26361-26369.
50. Kotal J, Langhansová H, Lieskovská J, et al. Modulation of host im- 69. Cao J, Shi L, Zhou Y, et al. Characterization of a new Kunitz-type
munity by tick saliva. J Proteomics. 2015;128:58-68. serine protease inhibitor from the hard tick Rhipicephalus hemaph-
51. Ribeiro JM, Makoul GT, Robinson DR. Ixodes dammini: evidence for ysaloides. Arch Insect Biochem Physiol. 2013;84(2):104-113.
salivary prostacyclin secretion. J Parasitol. 1988;74(6):1068-1069. 70. Figueiredo AC, de Sanctis D, Pereira PJ. The tick-derived anticoag-
52. Ibelli AM, Kim TK, Hill CC, et al. A blood meal-induced Ixodes ulant madanin is processed by thrombin and factor Xa. PLoS One.
scapularis tick saliva serpin inhibits trypsin and thrombin, and in- 2013;8(8):e71866.
terferes with platelet aggregation and blood clotting. Int J Parasitol. 71. Iwanaga S, Okada M, Isawa H, Morita A, Yuda M, Chinzei Y.
2014;44(6):369-379. Identification and characterization of novel salivary thrombin
53. Chmelar J, Oliveira CJ, Rezacova P, et al. A tick salivary protein inhibitors from the ixodidae tick, Haemaphysalis longicornis. Eur J
targets cathepsin G and chymase and inhibits host inflammation Biochem. 2003;270(9):1926-1934.
and platelet aggregation. Blood. 2011;117(2):736-744. 72. Nakajima C, Imamura S, Konnai S, et al. A novel gene encoding a
54. Tirloni L, Kim TK, Coutinho ML, et al. The putative role of thrombin inhibitory protein in a cDNA library from Haemaphysalis
Rhipicephalus microplus salivary serpins in the tick-host relation- longicornis salivary gland. J Vet Med Sci. 2006;68(5):447-452.
ship. Insect Biochem Mol Biol. 2016;71:12-28. 73. Imamura S, Junior ISV, Sugino M, Ohashi K, Onuma M. A serine
55. Wang X, Coons LB, Taylor DB, Stevens SE Jr, Gartner TK Variabilin, protease inhibitor (serpin) from Haemaphysalis longicornis as an an-
a novel RGD-containing antagonist of glycoprotein IIb-IIIa and ti-tick vaccine. Vaccine. 2005;23(10):1301-1311.
platelet aggregation inhibitor from the hard tick Dermacentor vari- 74. Brahma RK, Blanchet G, Kaur S, Manjunatha Kini R, Doley
abilis. J Biol Chem. 1996;271(30):17785-17790. R. Expression and characterization of haemathrins, mad-
56. Narasimhan S, Montgomery RR, DePonte K, et al. Disruption of anin-like thrombin inhibitors, isolated from the salivary gland
Ixodes scapularis anticoagulation by using RNA interference. Proc of tick Haemaphysalis bispinosa (Acari: Ixodidae). Thromb Res.
Natl Acad Sci USA. 2004;101(5):1141-1146. 2017;152:20-29.
57. Assumpcao TC, Mizurini DM, Ma D, et al. Ixonnexin from tick sa- 75. Du W, Gao Z, Wang K, et al. Expression and function assessment
liva promotes fibrinolysis by interacting with plasminogen and tis- of two serpin-type serine protease inhibitors from Haemaphysalis
sue-type plasminogen activator, and prevents arterial thrombosis. doenitzi. Res Vet Sci. 2020;132:1-9.
Sci Rep. 2018;8(1):4806. 76. Ricci CG, Pinto AFM, Berger M, Termignoni C. A thrombin in-
58. Schuijt TJ, Bakhtiari K, Daffre S, et al. Factor Xa activation of factor V hibitor from the gut of Boophilus microplus ticks. Exp Appl Acarol.
is of paramount importance in initiating the coagulation system: les- 2007;42(4):291-300.
sons from a tick salivary protein. Circulation. 2013;128(3):254-266. 77. Xu T, Lew-Tabor A, Rodriguez-Valle M. Effective inhibition
59. Francischetti IM, Valenzuela JG, Andersen JF, Mather TN, of thrombin by Rhipicephalus microplus serpin-15 (RmS-15)
Ribeiro JMC. Ixolaris, a novel recombinant tissue factor path- obtained in the yeast Pichia pastoris. Ticks Tick Borne Dis.
way inhibitor (TFPI) from the salivary gland of the tick, Ixodes 2016;7(1):180-187.
|
10 of 13       NARASIMHAN et al.

78. Horn F, Coutinho dos Santos P, Termignoni C. Boophilus microplus 98. Konnai S, Nakajima C, Imamura S, et al. Suppression of cell pro-
anticoagulant protein: an antithrombin inhibitor isolated from the liferation and cytokine expression by HL-p36, a tick salivary
cattle tick saliva. Archiv Biochem Biophys. 2000;384(1):68-73. gland-derived protein of Haemaphysalis longicornis. Immunology.
79. Ciprandi A, de Oliveira SK, Masuda A, Horn F, Termignoni C 2009;126(2):209-219.
Boophilus microplus: its saliva contains microphilin, a small throm- 99. Anisuzzaman  , Alim MA, Tsuji NN. Longistatin in tick-saliva targets
bin inhibitor. Exp Parasitol. 2006;114(1):40-46. RAGE. Oncotarget. 2015;6(34):35133-35134.
80. Anisuzzaman  , Islam MK, Alim MA, et al. Longistatin, a plasmin- 100. Poole NM, Mamidanna G, Smith RA, Coons LB, Cole JA.
ogen activator, is key to the availability of blood-meals for ixodid Prostaglandin E(2) in tick saliva regulates macrophage cell migra-
ticks. PLoS Pathog. 2011;7(3):e1001312. tion and cytokine profile. Parasit Vectors. 2013;6(1):261.
81. Xu XL, Cheng TY, Yang H. Enolase, a plasminogen receptor isolated 101. Deruaz M, Frauenschuh A, Alessandri AL, et al. Ticks produce
from salivary gland transcriptome of the ixodid tick Haemaphysalis highly selective chemokine binding proteins with antiinflamma-
flava. Parasitol Res. 2016;115(5):1955-1964. tory activity. J Exp Med. 2008;205(9):2019-2031.
82. Tabakawa Y, Ohta T, Yoshikawa S, et al. Histamine released from 102. Denisov SS, Ippel JH, Heinzmann ACA, et al. Tick saliva pro-
skin-infiltrating basophils but not mast cells is crucial for acquired tein Evasin-3 modulates chemotaxis by disrupting CXCL8 in-
tick resistance in mice. Front Immunol. 2018;9:1540. teractions with glycosaminoglycans and CXCR2. J Biol Chem.
83. Paesen GC, Adams PL, Harlos K, Nuttall PA, Stuart DI. Tick his- 2019;294(33):12370-12379.
tamine-binding proteins: isolation, cloning, and three-dimensional 103. Preston SG, Majtán J, Kouremenou C, et al. Novel immunomodula-
structure. Mol Cell. 1999;3(5):661-671. tors from hard ticks selectively reprogramme human dendritic cell
84. Sangamnatdej S, Paesen GC, Slovak M, Nuttall PA. A high affinity responses. PLoS Pathog. 2013;9(6):e1003450.
serotonin- and histamine-binding lipocalin from tick saliva. Insect 104. Sa-Nunes A, Bafica A, Lucas DA, et al. Prostaglandin E2 is a major
Mol Biol. 2002;11(1):79-86. inhibitor of dendritic cell maturation and function in Ixodes scapu-
85. Konnai S, Nishikado H, Yamada S, et al. Molecular identification laris saliva. J Immunol. 2007;179(3):1497-1505.
and expression analysis of lipocalins from blood feeding taiga tick, 105. Alarcon-Chaidez FJ, Boppana VD, Hagymasi AT, Adler AJ, Wikel
Ixodes persulcatus Schulze. Exp Parasitol. 2011;127(2):467-474. SK. A novel sphingomyelinase-like enzyme in Ixodes scapularis tick
86. Valdes JJ, Cabezas-Cruz A, Sima R, Butterill PT, Růžek D, Nuttall saliva drives host CD4 T cells to express IL-4. Parasite Immunol.
PA. Substrate prediction of Ixodes ricinus salivary lipocalins dif- 2009;31(4):210-219.
ferentially expressed during Borrelia afzelii infection. Sci Rep. 106. Nuttall PA, Labuda M. Tick-host interactions: saliva-activated
2016;6:32372. transmission. Parasitology. 2004;129(Suppl):S177-S189.
87. Sajiki Y, Konnai S, Ochi A, et al. Immunosuppressive effects of si- 107. Kotsyfakis M, Horka H, Salat J, Andersen JF. The crystal structures
alostatin L1 and L2 isolated from the taiga tick Ixodes persulcatus of two salivary cystatins from the tick Ixodes scapularis and the ef-
Schulze. Ticks Tick Borne Dis. 2020;11(2):101332. fect of these inhibitors on the establishment of Borrelia burgdorferi
88. Lieskovska J, Páleníková J, Langhansová H, et al. Tick sialostatins L infection in a murine model. Mol Microbiol. 2010;77(2):456-470.
and L2 differentially influence dendritic cell responses to Borrelia 108. Anguita J, Ramamoorthi N, Hovius JWR, et al. Salp15, an Ixodes
spirochetes. Parasit Vectors. 2015;8:275. scapularis salivary protein, inhibits CD4(+) T cell activation.
89. Kotsyfakis M, Karim S, Andersen JF, Mather TN, Ribeiro Immunity. 2002;16(6):849-859.
JM. Selective cysteine protease inhibition contributes to 109. Gillespie RD, Dolan MC, Piesman J, Titus RG. Identification of an
blood-feeding success of the tick Ixodes scapularis. J Biol Chem. IL-2 binding protein in the saliva of the Lyme disease vector tick,
2007;282(40):29256-29263. Ixodes scapularis. J Immunol. 2001;166(7):4319-4326.
90. Hovius JW, de Jong MAWP, den Dunnen J, et al. Salp15 bind- 110. Beaufays J, Adam B, Menten-Dedoyart C, et al. Ir-LBP, an ixodes
ing to DC-SIGN inhibits cytokine expression by impairing both ricinus tick salivary LTB4-binding lipocalin, interferes with host
nucleosome remodeling and mRNA stabilization. PLoS Pathog. neutrophil function. PLoS One. 2008;3(12):e3987.
2008;4(2):e31. 111. Toyomane K, Konnai S, Niwa A, et al. Identification and the pre-
91. Leboulle G, Crippa M, Decrem Y, et al. Characterization of a novel liminary in vitro characterization of IRIS homologue from sali-
salivary immunosuppressive protein from Ixodes ricinus ticks. J Biol vary glands of Ixodes persulcatus Schulze. Ticks Tick Borne Dis.
Chem. 2002;277(12):10083-10089. 2016;7(1):119-125.
92. Prevot PP, Beschin A, Lins L, et al. Exosites mediate the anti-in- 112. Coutinho ML, Bizzarro B, Tirloni L, et al. Rhipicephalus microplus
flammatory effects of a multifunctional serpin from the saliva of serpins interfere with host immune responses by specifically
the tick Ixodes ricinus. FEBS J. 2009;276(12):3235-3246. modulating mast cells and lymphocytes. Ticks Tick Borne Dis.
93. Ullmann AJ, Dolan MC, Sackal CA, Fikrig E, Piesman J, Zeidner 2020;11(4):101425.
NS. Immunization with adenoviral-vectored tick salivary gland 113. Tyson K, Elkins C, Patterson H, Fikrig E, de Silva A. Biochemical
proteins (SALPs) in a murine model of Lyme borreliosis. Ticks Tick and functional characterization of Salp20, an Ixodes scapularis tick
Borne Dis. 2013;4(1–2):160-163. salivary protein that inhibits the complement pathway. Insect Mol
94. Blisnick AA, Šimo L, Grillon C, et al. The immunomodulatory effect Biol. 2007;16(4):469-479.
of IrSPI, a tick salivary gland serine protease inhibitor involved in 114. Hourcade DE, Akk AM, Mitchell LM, et al. Anti-complement ac-
Ixodes ricinus tick feeding. Vaccines. 2019;7(4):148. tivity of the Ixodes scapularis salivary protein Salp20. Mol Immunol.
95. Palenikova J, Lieskovská J, Langhansová H, Kotsyfakis M, Chmelař 2016;69:62-69.
J, Kopecký J. Ixodes ricinus salivary serpin IRS-2 affects Th17 dif- 115. Tyson KR, Elkins C, de Silva AM. A novel mechanism of comple-
ferentiation via inhibition of the interleukin-6/STAT-3 signaling ment inhibition unmasked by a tick salivary protein that binds to
pathway. Infect Immun. 2015;83(5):1949-1956. properdin. J Immunol. 2008;180(6):3964-3968.
96. Kotal J, Stergiou N, Buša M, et al. The structure and function of 116. Valenzuela JG, Charlab R, Mather TN, Ribeiro JM. Purification, clon-
Iristatin, a novel immunosuppressive tick salivary cystatin. Cell Mol ing, and expression of a novel salivary anticomplement protein from
Life Sci. 2019;76(10):2003-2013. the tick, Ixodes scapularis. J Biol Chem. 2000;275(25):18717-18723.
97. Wang F, Song Z, Chen J, et al. The immunosuppressive func- 117. Daix V, Schroeder H, Praet N, et al. Ixodes ticks belonging to the
tions of two novel tick serpins, HlSerpin-a and HlSerpin-b, from Ixodes ricinus complex encode a family of anticomplement pro-
Haemaphysalis longicornis. Immunology. 2020;159(1):109-120. teins. Insect Mol Biol. 2007;16(2):155-166.
NARASIMHAN et al. |
      11 of 13

118. Schroeder H, Daix V, Gillet L, Renauld J-C, Vanderplasschen A. The 138. Brown SJ, Askenase PW. Amblyomma americanum: physiochemical
paralogous salivary anti-complement proteins IRAC I and IRAC II isolation of a protein derived from the tick salivary gland that is ca-
encoded by Ixodes ricinus ticks have broad and complementary in- pable of inducing immune resistance in guinea pigs. Exp Parasitol.
hibitory activities against the complement of different host spe- 1986;62(1):40-50.
cies. Microbes Infect. 2007;9(2):247-250. 139. Brown SJ, Galli SJ, Gleich GJ, Askenase PW. Ablation of immu-
119. Reichhardt MP, Johnson S, Tang T, et al. An inhibitor of comple- nity to Amblyomma americanum by anti-basophil serum: co-
ment C5 provides structural insightss into activation. Proc Natl operation between basophils and eosinophils in expression
Acad Sci USA. 2020;117(1):362-370. of immunity to ectoparasites (ticks) in guinea pigs. J Immunol.
120. Schuijt TJ, Coumou J, Narasimhan S, et al. A tick mannose-binding 1982;129(2):790-796.
lectin inhibitor interferes with the vertebrate complement cas- 140. Brown SJ, Worms MJ, Askenase PW. Rhipicephalus appendiculatus:
cade to enhance transmission of the lyme disease agent. Cell Host larval feeding sites in guinea pigs actively sensitized and receiving
Microbe. 2011;10(2):136-146. immune serum. Exp Parasitol. 1983;55(1):111-120.
121. Wagemakers A, Coumou J, Schuijt TJ, et al. An Ixodes ricinus tick 141. Nazario S, de Silva AM, Kantor FS, et al. Prevention of Borrelia
salivary lectin pathway inhibitor protects Borrelia burgdorferi burgdorferi transmission in guinea pigs by tick immunity. Am J Trop
sensu lato from human complement. Vector Borne Zoonotic Dis. Med Hyg. 1998;58(6):780-785.
2016;16(4):223-228. 142. Whelen AC, Wikel SK. Acquired resistance of guinea pigs to
122. Tsuji N, Kamio T, Isobe T, Fujisaki K. Molecular characterization of Dermacentor andersonimediated by humoral factors. J Parasitol.
a peroxiredoxin from the hard tick Haemaphysalis longicornis. Insect 1993;79(6):908-912.
Mol Biol. 2001;10(2):121-129. 143. Anderson JM, Moore IN, Nagata BM, Ribeiro JMC, Valenzuela JG,
123. Kusakisako K, Galay RL, Umemiya-Shirafuji R, et al. 2-Cys perox- Sonenshine DE. Ticks, Ixodes scapularis, feed repeatedly on white-
iredoxin is required in successful blood-feeding, reproduction, and footed mice despite strong inflammatory response: an expanding
antioxidant response in the hard tick Haemaphysalis longicornis. paradigm for understanding tick-host interactions. Front Immunol.
Parasit Vectors. 2016;9:457. 2017;8:1784.
124. Hernandez EP, Kusakisako K, Talactac MR, et al. Characterization 144. Wikel SK, Ramachandra RN, Bergman DK, Burkot TR, Piesman J.
and expression analysis of a newly identified glutathione Infestation with pathogen-free nymphs of the tick Ixodes scapularis
S-transferase of the hard tick Haemaphysalis longicornis during induces host resistance to transmission of Borrelia burgdorferi by
blood-feeding. Parasit Vectors. 2018;11(1):91. ticks. Infect Immun. 1997;65(1):335-338.
125. Narasimhan S, DePonte K, Marcantonio N, et al. Immunity against 145. Narasimhan S, Booth CJ, DePonte K, et al. Host-specific ex-
Ixodes scapularis salivary proteins expressed within 24 hours of at- pression of Ixodes scapularis salivary genes. Ticks Tick Borne Dis.
tachment thwarts tick feeding and impairs Borrelia transmission. 2019;10(2):386-397.
PLoS One. 2007;2(5):e451. 146. Francis J, Little DA. Resistance of droughtmaster cattle to tick in-
126. Kim TK, Tirloni L, Pinto AFM, et al. Ixodes scapularis tick saliva pro- festation and babesiosis. Australian Vet J. 1964;40:247-253.
teins sequentially secreted every 24 h during blood feeding. PLoS 147. Wambura PN, Gwakisa PS, Silayo RS, Rugaimukamu EA. Breed-
Negl Trop Dis. 2016;10(1):e0004323. associated resistance to tick infestation in Bos indicus and their
127. Anderson JF, Magnarelli LA. Biology of ticks. Infect Dis Clin North crosses with Bos taurus. Vet Parasitol. 1998;77(1):63-70.
Am. 2008;22(2):195-215, v. 148. Porto Neto LR, Jonsson NN, D’Occhio MJ, Barendse W.
128. Wikel SK. Tick-host-pathogen systems immunobiology: an inter- Molecular genetic approaches for identifying the basis of vari-
active trio. Front Biosci. 2018;23:265-283. ation in resistance to tick infestation in cattle. Vet Parasitol.
129. Kurokawa C, Lynn GE, Pedra JHF, Pal U, Narasimhan S, Fikrig 2011;180(3–4):165-172.
E. Interactions between Borrelia burgdorferi and ticks. Nat Rev 149. Schleger AV, Lincoln DT, McKenna RV, Kemp DH, Roberts
Microbiol. 2020. JA. Boophilus microplus: cellular responses to larval attach-
130. Brown SJ, Askenase PW. Rejection of ticks from guinea pigs by ment and their relationship to host resistance. Aust J Biol Sci.
anti-hapten-antibody-mediated degranulation of basophils at cu- 1976;29(5–6):499-512.
taneous basophil hypersensitivity sites: role of mediators other 150. Tabor AE, Ali A, Rehman G, et al. Cattle tick Rhipicephalus micro-
than histamine. J Immunol. 1985;134(2):1160-1165. plus-host interface: a review of resistant and susceptible host re-
131. Askenase PW, Atwood JE. Basophils in tuberculin and "Jones-Mote" sponses. Front Cell Infect Microbiol. 2017;7:506.
delayed reactions of humans. J Clin Invest. 1976;58(5):1145-1154. 151. Beaudouin E, Kanny G, Guerin B, Guerin L, Plenat F, Moneret-
132. Hewetson RW. The inheritance of resistance by cattle to cattle Vautrin DA. Unusual manifestations of hypersensitivity after
tick. Aust Vet J. 1972;48(5):299-303. a tick bite: report of two cases. Ann Allergy Asthma Immunol.
133. Allen JR, Kemp DH. Observations on the behaviour of Dermacentor 1997;79(1):43-46.
andersoni larvae infesting normal and tick resistant guinea-pigs. 152. Garcia MV, Matias J, Aguirre ADAR, Csordas BG, SzabO MJ,
Parasitology. 1982;84(Pt 2):195-204. Andreotti R. Successful feeding of Amblyomma coelebs (Acari:
134. Fivaz BH, Tucker S, Petney T. Cross-resistance between instars Ixodidae) nymphs on humans in Brazil: skin reactions to parasitism.
of the brown ear-tick Rhipicephalus appendiculatus (Acarina: J Med Entomol. 2015;52(2):117-119.
Ixodidae). Exp Appl Acarol. 1991;11(4):323-326. 153. Schwartz BS, Ribeiro JM, Goldstein MD. Anti-tick antibodies:

135. Gebbia JA, Bosler EM, Evans RD, Schneider EM. Acquired resis- an epidemiologic tool in Lyme disease research. Am J Epidemiol.
tance in dogs to repeated infestation with Ixodes scapularis (Acari: 1990;132(1):58-66.
Ixodidae) reduces tick viability and reproductive success. Exp Appl 154. Schwartz BS, Goldstein MD, Childs JE. Antibodies to Borrelia
Acarol. 1995;19(10):593-605. burgdorferi and tick salivary gland proteins in New Jersey outdoor
136. Narasimhan S, Kurokawa C, Diktas H, et al. Ixodes scapularis saliva workers. Am J Public Health. 1993;83(12):1746-1748.
components that elicit responses associated with acquired tick-re- 155. Burke G, Wikel SK, Spielman A, et al. Hypersensitivity to ticks and
sistance. Ticks Tick Borne Dis. 2020;11(3):101369. Lyme disease risk. Emerg Infect Dis. 2005;11(1):36-41.
137. Wada T, Ishiwata K, Koseki H, et al. Selective ablation of baso- 156. Piesman J, Mather TN, Sinsky RJ, Spielman A. Duration of tick
phils in mice reveals their nonredundant role in acquired immunity attachment and Borrelia burgdorferi transmission. J Clin Microbiol.
against ticks. J Clin Invest. 2010;120(8):2867-2875. 1987;25(3):557-558.
|
12 of 13       NARASIMHAN et al.

157. Crippa M, Rais O, Gern L. Investigations on the mode and dynamics 180. Tirloni L, Kim TK, Pinto AFM, Yates JR 3rd, da Silva Vaz I Jr,
of transmission and infectivity of Borrelia burgdorferi sensu stricto Mulenga A. Tick-host range adaptation: changes in protein profiles
and Borrelia afzelii in Ixodes ricinus ticks. Vector Borne Zoonotic Dis. in unfed Adult Ixodes scapularis and Amblyomma americanum saliva
2002;2(1):3-9. stimulated to feed on different hosts. Front Cell Infect Microbiol.
158. Hodzic E, Fish D, Maretzki CM, De Silva AM, Feng S, Barthold 2017;7:517.
SW. Acquisition and transmission of the agent of human gran- 181. Franzin AM, Maruyama SR, Garcia GR, et al. Immune and biochem-
ulocytic ehrlichiosis by Ixodes scapularis ticks. J Clin Microbiol. ical responses in skin differ between bovine hosts genetically sus-
1998;36(12):3574-3578. ceptible and resistant to the cattle tick Rhipicephalus microplus.
159. Hermance ME, Thangamani S. Tick saliva enhances powassan Parasit Vectors. 2017;10(1):51.
virus transmission to the host, influencing its dissemination and 182. Sueki H, Gammal C, Kudoh K, Kligman AM. Hairless guinea pig
the course of disease. J Virol. 2015;89(15):7852-7860. skin: anatomical basis for studies of cutaneous biology. Eur J
160. Allen JR. Tick resistance: basophils in skin reactions of resistant Dermatol. 2000;10(5):357-364.
guinea pigs. Int J Parasitol. 1973;3(2):195-200. 183. Gudjonsson JE, Johnston A, Dyson M, Valdimarsson H,

161. Allen JR, Doube BM, Kemp DH. Histology of bovine skin reactions Elder JT. Mouse models of psoriasis. J Invest Dermatol.
to Ixodes holocyclus Neumann. Can J Comp Med. 1977;41(1):26-35. 2007;127(6):1292-1308.
162. Brown SJ, Askenase PW. Cutaneous basophil responses and 184. Savoji H, Godau B, Hassani MS, Akbari M. Skin tissue substi-

immune resistance of guinea pigs to ticks: passive trans- tutes and biomaterial risk assessment and testing. Front Bioeng
fer with peritoneal exudate cells or serum. J Immunol. Biotechnol. 2018;6:86.
1981;127(5):2163-2167. 185. Naldaiz-Gastesi N, Bahri OA, López de Munain A, McCullagh
163. Brown SJ, Knapp FW. Response of hypersensitized guinea pigs KJA, Izeta A. The panniculus carnosus muscle: an evolution-
to the feeding of Amblyomma americanum ticks. Parasitology. ary enigma at the intersection of distinct research fields. J Anat.
1981;83(Pt 1):213-223. 2018:233;275-288.
164. Tatchell RJ, Bennett GF Boophilus microplus: antihistaminic
186. Kassem R, Liberty Z, Babaev M, Trau H, Cohen O. Harnessing
and tranquilizing drugs and cattle resistance. Exp Parasitol. the skin-thyroid connection for wound healing: a pro-
1969;26(3):369-377. spective controlled trial in guinea pigs. Clin Exp Dermatol.
165. Wikel SK. Histamine content of tick attachment sites and the ef- 2012;37(8):850-856.
fects of H1 and H2 histamine antagonists on the expression of re- 187. Flacher V, Tripp CH, Mairhofer DG, et al. Murine Langerin+ dermal
sistance. Ann Trop Med Parasitol. 1982;76(2):179-185. dendritic cells prime CD8+ T cells while Langerhans cells induce
166. Brown SJ, Graziano FM, Askenase PW. Immune serum transfer of cross-tolerance. EMBO Mol Med. 2014;6(9):1191-1204.
cutaneous basophil-associated resistance to ticks: mediation by 188. Mutyambizi K, Berger CL, Edelson RL. The balance between im-
7SIgG1 antibodies. J Immunol. 1982;129(6):2407-2412. munity and tolerance: the role of Langerhans cells. Cell Mol Life Sci.
167. Wikel SK, Allen JR. Acquired resistance to ticks. I. Passive transfer 2009;66(5):831-840.
of resistance. Immunology. 1976;30(3):311-316. 189. Summerfield A, Meurens F, Ricklin ME. The immunology of the
168. Wikel SK, Allen JR. Acquired resistance to ticks. III. Cobra porcine skin and its value as a model for human skin. Mol Immunol.
venom factor and the resistance response. Immunology. 2015;66(1):14-21.
1978;32(4):457-465. 190. More DD, Cardoso FF, Mudadu MA, et al. Network analysis un-
169. Kock MA, Hew BE, Bammert H, Fritzinger DC, Vogel C-W. covers putative genes affecting resistance to tick infestation in
Structure and function of recombinant cobra venom factor. J Biol Braford cattle skin. BMC Genom. 2019;20(1):998.
Chem. 2004;279(29):30836-30843. 191. Kurokawa C, Narasimhan S, Vidyarthi A, et al. Repeat tick expo-
170. Wikel SK. Acquired resistance to ticks: expression of resistance by sure elicits distinct immune responses in guinea pigs and mice.
C4-deficient guinea pigs. Am J Trop Med Hyg. 1979;28(3):586-590. Ticks Tick Borne Dis. 2020;11(6):101529.s
171. Szabo MP, Bechara GH. Sequential histopathology at the 192. Robbertse L, Richards SA, Maritz-Olivier C. Bovine immune fac-
Rhipicephalus sanguineus tick feeding site on dogs and guinea pigs. tors underlying tick resistance: integration and future directions.
Exp Appl Acarol. 1999;23(11):915-928. Front Cell Infect Microbiol. 2017;7:522.
172. Urbina C, Ortiz C, Hurtado I. A new look at basophils in mice. Int 193. Heller-Haupt A, Kagaruki LK, Varma MG. Resistance and cross-re-
Arch Allergy Appl Immunol. 1981;66(2):158-160. sistance in rabbits to adults of three species of African ticks (Acari:
173. Dvorak AM, Galli SJ, Morgan E, Galli AS, Hammond ME, Dvorak Ixodidae). Exp Appl Acarol. 1996;20(3):155-165.
HF. Anaphylactic degranulation of guinea pig basophilic leuko- 194. Parizi LF, Githaka NW, Logullo C, et al. The quest for a uni-
cytes. II. Evidence for regranulation of mature basophils during re- versal vaccine against ticks: cross-immunity insights. Vet J.
covery from degranulation in vitro. Lab Invest. 1982;46(5):461-475. 2012;194(2):158-165.
174. Cromheecke JL, Nguyen KT, Huston DP. Emerging role of human 195. Kim TK, Tirloni L, Pinto AFM, et al. Time-resolved proteomic pro-
basophil biology in health and disease. Curr Allergy Asthma Rep. file of Amblyomma americanum tick saliva during feeding. PLoS Negl
2014;14(1):408. Trop Dis. 2020;14(2):e0007758.
175. Sokol CL, Medzhitov R. Role of basophils in the initiation of Th2 196. Plotkin SA. Vaccines: the fourth century. Clin Vaccine Immunol.
responses. Curr Opin Immunol. 2010;22(1):73-77. 2009;16(12):1709-1719.
176. Min B. Basophils induce Th2 immunity: is this final answer? 197. Suschak JJ, Williams JA, Schmaljohn CS. Advancements in DNA
Virulence. 2010;1(5):399-401. vaccine vectors, non-mechanical delivery methods, and molecu-
177. Ohta T, Yoshikawa S, Tabakawa Y, et al. Skin CD4(+) memory T lar adjuvants to increase immunogenicity. Hum Vaccin Immunother.
cells play an essential role in acquired anti-tick immunity through 2017;13(12):2837-2848.
interleukin-3-mediated basophil recruitment to tick-feeding sites. 198. Ruiz LM, Orduz S, López ED, Guzmán F, Patarroyo ME, Armengol
Front Immunol. 2017;8:1348. G. Immune response in mice and cattle after immunization with
178. Barbour AG, Fish D. The biological and social phenomenon of a Boophilus microplus DNA vaccine containing bm86 gene. Vet
Lyme disease. Science. 1993;260(5114):1610-1616. Parasitol. 2007;144(1–2):138-145.
179. Ribeiro JM. Role of saliva in tick/host interactions. Exp Appl Acarol. 199. Hassan IA, Wang Y, Zhou Y, Cao J, Zhang H, Zhou J. Cross pro-
1989;7(1):15-20. tection induced by combined Subolesin-based DNA and protein
NARASIMHAN et al. |
      13 of 13

immunizations against adult Haemaphysalis longicornis. Vaccine. flavivirus replicons derived from tick-borne encephalitis virus.
2020;38(4):907-915. J Virol. 2005;79(24):15107-15113.
200. Pardi N, Hogan MJ, Porter FW, Weissman D. mRNA vaccines - a
new era in vaccinology. Nat Rev Drug Discov. 2018;17(4):261-279.
01. VanBlargan LA, Himansu S, Foreman BM, Ebel GD, Pierson TC,
2
How to cite this article: Narasimhan S, Kurokawa C, DeBlasio
Diamond MS. An mRNA vaccine protects mice against multiple
M, et al. Acquired tick resistance: The trail is hot. Parasite
tick-transmitted flavivirus infections. Cell Rep. 2018;25(12):3382-
3392 e3. Immunol. 2021;43:e12808. https://doi.org/10.1111/pim.12808
202. Aberle JH, Aberle SW, Kofler RM, Mandl CW. Humoral
and cellular immune response to RNA immunization with

You might also like