Next Article in Journal
Monitoring the Bioprotective Potential of Lactiplantibacillus pentosus Culture on Pathogen Survival in and the Shelf-Life of Fresh Ready-to-Eat Salads Stored under Modified Atmosphere Packaging
Previous Article in Journal
Zika Virus Neuropathogenesis—Research and Understanding
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Hard Ticks as Vectors: The Emerging Threat of Tick-Borne Diseases in India

by
Nandhini Perumalsamy
,
Rohit Sharma
,
Muthukumaravel Subramanian
and
Shriram Ananganallur Nagarajan
*
Division of Vector Biology and Control, Indian Council of Medical Research—Vector Control Research Centre (ICMR-VCRC), Puducherry 605006, India
*
Author to whom correspondence should be addressed.
Pathogens 2024, 13(7), 556; https://doi.org/10.3390/pathogens13070556
Submission received: 3 April 2024 / Revised: 6 May 2024 / Accepted: 7 May 2024 / Published: 2 July 2024
(This article belongs to the Section Ticks)

Abstract

:
Hard ticks (Ixodidae) play a critical role in transmitting various tick-borne diseases (TBDs), posing significant global threats to human and animal health. Climatic factors influence the abundance, diversity, and vectorial capacity of tick vectors. It is imperative to have a comprehensive understanding of hard ticks, pathogens, eco-epidemiology, and the impact of climatic changes on the transmission dynamics of TBDs. The distribution and life cycle patterns of hard ticks are influenced by diverse ecological factors that, in turn, can be impacted by changes in climate, leading to the expansion of the tick vector’s range and geographical distribution. Vector competence, a pivotal aspect of vectorial capacity, involves the tick’s ability to acquire, maintain, and transmit pathogens. Hard ticks, by efficiently feeding on diverse hosts and manipulating their immunity through their saliva, emerge as competent vectors for various pathogens, such as viruses, parasites and bacteria. This ability significantly influences the success of pathogen transmission. Further exploration of genetic diversity, population structure, and hybrid tick vectors is crucial, as they play a substantial role in influencing vector competence and complicating the dynamics of TBDs. This comprehensive review deals with important TBDs in India and delves into a profound understanding of hard ticks as vectors, their biology, and the factors influencing their vector competence. Given that TBDs continue to pose a substantial threat to global health, the review emphasizes the urgency of investigating tick control strategies and advancing vaccine development. Special attention is given to the pivotal role of population genetics in comprehending the genetic diversity of tick populations and providing essential insights into their adaptability to environmental changes.

Graphical Abstract

1. Introduction

Arthropoda stands out as the largest and most diverse phylum within the animal kingdom, encompassing crustaceans, arachnids, and insects. Insects, a remarkably varied group, play vital roles in ecosystems by contributing to pollination and honey production, and serving as a food source [1,2]. However, hematophagous insects like mosquitoes, sandflies, and triatomine bugs take on a different role, transmitting pathogens and acting as vectors for several human diseases such as malaria, dengue, zika, chikungunya, leishmaniasis, sleeping sickness, and Chagas disease [3,4]. Within blood-sucking arthropods, “Ticks” hold the distinction of being the second most significant vectors of human diseases [5,6,7,8]. Ticks, belonging to the class Arachnida, which also includes spiders, scorpions, and mites, [9,10,11,12] differ from insects with their non-segmented and spherical bodies [13,14]. The two main types of ticks are hard ticks (Ixodidae) and soft ticks (Argasidae), with an additional monotypic family known as Nutalliellidae [15,16,17].
Hard ticks, as obligate blood feeders, stand out as primary vectors capable of transmitting a broader array of pathogens than any other arthropod vector globally [18,19]. Infesting various vertebrates, they play a pivotal role in transmitting infections to both humans and animals, leading to a spectrum of tick-borne diseases (TBDs) [6,17,20]. Ixodid ticks, during a single feeding, can consume substantial amounts of host blood, thereby increasing the likelihood of acquiring pathogens from infected hosts [8,21,22]. Notably, during blood feeding, ticks inject a significant amount of saliva containing immunomodulatory proteins to impair the host’s anti-tick response [23,24]. Factors such as a long lifespan, high reproductive capacity, a broad host range, and the ability to survive in harsh conditions contribute to making ticks efficient vectors for various infections [6,25,26,27]. Despite extensive research on mosquitoes, mosquito-borne diseases, and mosquito control, there is a noticeable dearth of studies focused on ticks, underscoring their equal significance as disease vectors [28,29].
Tick vectors, with ubiquitous distribution, are expanding globally, posing significant public health risks linked to emerging TBDs [30,31,32]. Notable hard ticks that commonly bite humans include Ixodes scapularis, Amblyomma americanum, Dermacentor variabilis, Ixodes pacificus, Amblyomma hebraeum, Hyalomma anatolicum, Hyalomma marginatum, Haemaphysalis spinigera, Ixodes ricinus, Ixodes persulcatus, Ixodes holocyclus, and Dermacentor andersoni, while soft ticks comprise Otobius megnini and Ornithodoros coriaceus [33,34,35]. These tick species are commonly encountered in areas where people come into contact with ticks [35]. They are crucial as vectors for certain diseases.
There are records of at least five hard tick species (Amblyomma integrum, Haemaphysalis spinigera, Dermacentor auratus, Rhipicephalus haemaphysaloides, and Hyalomma isaaci) and one soft tick species (Otobius megnini) infesting people in India [36,37]. Researchers have sporadically observed zooanthrophilic Hyalomma species, such as H. marginatum and H. truncatum, feeding on humans [38]. Typically, these ticks target individuals exposed to vegetation and wooded areas, underscoring the importance of personal protection, routine tick checks, and prompt removal for effective prevention of TBDs [29].
In India, notable TBDs encompass Kyasanur Forest Disease (KFD), Crimean–Congo hemorrhagic fever (CCHF), Lyme disease (LD), Q fever (coxiellosis), and Rickettsial infections [39,40,41,42]. The first reported outbreaks of KFD occurred in Karnataka in 1957, with subsequent positive sero-surveillance findings documented in various states across India, despite the absence of major outbreaks [39,43,44,45,46]. Concerns regarding the prevalence of CCHF exist in Gujarat, Rajasthan, and Uttar Pradesh, with positive cases identified in both human and animal samples across the country [47,48,49,50]. Given these circumstances, a critical focus on TBDs is essential, necessitating the implementation of effective tick control measures [51]. Ongoing studies predominantly center on tick control methods, incorporating chemical, biological, and immunological approaches, owing to the transmission of various diseases to both humans and animals [26,52,53].
The occurrence and outbreak of TBDs exhibit variability depending on climatic conditions, including the presence of virulent pathogens, abundant competent vectors, diverse animal hosts, suitable environmental settings, and vulnerable human populations [25,54,55,56,57]. Given that tick populations are highly localized, factors such as global warming contributing to climate change may escalate the threat of TBDs, heightening the potential for outbreaks [16,58]. The recent outbreaks of severe viral infections, notably CCHF in India [49,50], are concerning, especially due to the presence of a diverse range of hard ticks serving as competent vectors. Insufficient research exists on competent vectors, their geographical distribution, and the potential expansion of known vectors in India. The growing apprehension about tick-borne infections in the country, coupled with the expanding host range of tick vectors, has prompted an analysis of available data on TBDs. This review article delves into the distribution and diversity of hard tick vectors, exploring their biology, expansion patterns, vector competence, and control strategies. Additionally, the review highlights the epidemiology, transmission dynamics, and potential future risks associated with TBDs in India.

1.1. Tick Vectors: An Introduction

Ticks, originating 225 million years ago as parasites of reptiles during the pre-mid cretaceous period, have evolved from their ancient origins [6,59,60]. Taxonomically, ticks are closely related to mites and fall within the Arachnida class, which also includes spiders and scorpions. Three families classify them: Argasidae (soft ticks), Ixodidae (hard ticks), and the monotypic Nutalliellidae [61,62,63] (Figure 1). There are approximately 109 tick species distributed across 12 genera in India [33,64,65].
The Ixodidae family, or hard ticks, is the dominant family in terms of species diversity and medical and veterinary significance. With over 650 species organized into two major groups (Prostriata and Metastriata), five subfamilies, and 13 genera, hard ticks play a crucial role in disease transmission [67,68,69,70]. As a result of taxonomic re-evaluation, the family Argasidae, ranked second to Ixodidae, has been divided into two subfamilies, Ornithodorinae and Argasinae [71,72,73]. Argasidae, or soft ticks, consist of around 170 species, predominantly found in sheltered environments such as caves, animal nests, burrows, and human habitats. They are characterized by their leathery integument and lack of a dorsal shield (scutum) [73,74]. Some of these ticks serve as vectors for relapsing fevers caused by Borrelia spp. in humans and can transmit arboviruses and other bacterial pathogens [75,76]. The third tick family, Nuttalliella namaqua (only species of Nuttaliellidae), is a unique species bridging the characteristics of hard and soft ticks [62]. N. namaqua, considered the “evolutionary missing link” between the two tick families, was previously classified within the hard tick family, specifically related to the genus Ixodes [62,63,77,78]. Hard ticks serve as the main carriers of viruses and bacteria significant to the medical and veterinary field [30,79].

1.2. Structure and Physiology of Hard Ticks

Ixodidae, commonly known as hard ticks, are categorized into two groups, Prostriata (e.g., Ixodes spp.) and Metastriata (e.g., Hyalomma excavatum and Rhipicephalus sanguineus), based on morphological distinctions [67,80,81]. Prostriate ticks are identified by a prominent anterior anal groove, while metastriate ticks feature a small posterior slit-like anal groove [70,82].
The life cycle of Ixodid ticks comprises four stages: egg, larva, nymph, and adult [70]. Females possess a rigid cuticular plate, or scutum, on the front part of the dorsal body surface, whereas in males, the scutum covers nearly the entire dorsal surface [70,82]. Metastriata hard ticks have ornate patterns on their scutum and festoons that help identify them at the species level, but it is still not clear what their biological purpose is [83]. Ticks have a distinct body structure characterized by the anterior capitulum, housing their mouthparts, and the posterior idiosoma, where the legs attach, along with the genital pore, spiracles, and anal aperture [84] (Figure 2). In the adult stage, key features include the genital aperture situated between coxa III, spiracles positioned adjacent to coxa IV, and the presence or absence of eyes in immature and adult ticks. Larvae have six legs, while nymphs and adults each have eight legs [70,82]. Adult ticks have well-developed pulvilli, four separate plates (ventral, anal, and two lateral), and possibly festoons with a scalloped pattern along the back edge. Larvae show an anterior scutal shield, and the entire dorsal capitulum is visible from above [5,85]. The historical understanding of tick biology dates back to texts like Pliny the Elder’s “Historia Naturalis” and Dr. Thomas Moufet’s “Insectorum sive Minimorum Animalium Theatrum”, which noted the absence of a waste elimination mechanism in engorged ticks [86]. The synganglion is the tick’s central nervous system. It is located anteroventrally above the genital pore and controls many bodily functions. It is the fused central nervous system that controls organ activity and coordinates responses [87,88]. Respiration in ticks occurs through numerous tiny air tubes connected to paired spiracles, enabling essential gas exchange for their survival [89]. A simple heart, mid-dorsally located, filters and circulates hemolymph, a vital circulating fluid containing salts, amino acids, and soluble proteins that nourish organs and transport essential substances throughout the tick’s body [90,91]. Following the heart, paired salivary glands, resembling grape clusters, are antero-laterally situated. Through salivary ducts that connect the chelicerae and the hypostome [86], these glands release substances into the salivarium, a small space inside the mouth. The primary internal organ, the midgut, resembles a sac with numerous lateral diverticula. In unfed ticks, these tube-like diverticula expand during feeding and fill with blood, aiding digestion and influencing tick growth, reproduction, and fitness [92].
Malpighian tubules manage excretion by releasing nitrogenous wastes into the rectal sac, maintaining the tick’s internal balance by eliminating metabolic by-products, mainly in the form of guanine. The reproductive system includes male structures like testes, vasa deferentia, seminal vesicle, and ejaculatory duct, while females have an ovary, oviducts, uterus, vagina, and seminal receptacles. The ovary expands significantly during feeding and mating, containing large, amber-colored eggs in gravid females [70].

1.3. Life Cycle of Hard Ticks

The tick life cycle encompasses distinct phases, including host-seeking, feeding, development, metamorphosis, and reproduction, interspersed with diapause (dormancy) periods aligned with seasonal changes [93]. Hard ticks have a single nymphal instar, while soft ticks have two or more nymphal instars. Throughout their life cycle, ticks, as obligate ectoparasites, rely on blood feeding [70], characterized by short feeding phases and extended non-feeding intervals [94]. Sexual dimorphism is evident only in the adult stage [95], and host selection may differ between juvenile and adult ticks of the same species [10]. Over 90% of Ixodid species exhibit a three-host life cycle, with larvae and nymphs feeding on small to medium-sized hosts and adults targeting larger host species [70].
Ixodes scapularis, for example, feeds on various host species, showcasing preferences in host selection [82]. The number and variety of hosts for hard-bodied ticks vary, with the completion of the typical three-host life cycle ranging from less than a year to 5 or 6 years under favorable conditions [70]. Most arthropods display a photo-periodically controlled life cycle to synchronize development stages with favorable climatic conditions [95]. Host-seeking diapause commonly occurs during periods of decreasing photoperiod [70], enabling ticks to endure harsh environments and conserve energy. Diapause is crucial for ticks to survive unfavorable conditions, during which they reduce metabolic rates and abstain from feeding [96]. Over 90% of the life cycle occurs off the host, with molting typically taking place in sheltered microhabitats such as soil, leaf litter, or host nests [70,97].
Ticks regulate body water levels by absorbing water vapor from the air, a critical aspect of their survival [98]. The critical equilibrium activity (CEA) represents the minimum moisture level needed for water balance [99]. Ticks have adaptations to minimize water loss, such as a modest metabolic rate, a relatively impermeable cuticle, and the excretion of solid urine in the form of guanine [100]. The careful regulation of water balance is crucial for ticks, influencing their distribution, survival, and activity and playing a critical role in their capacity to transmit disease-causing pathogens [101]. Some Ixodids have evolved as two-host species (e.g., Hyalomma isaaci), where larvae attach to a host, ecdyse on the host upon completing feeding, and the resulting nymphs reattach and complete feeding. Single-host tick species, like Rhipicephalus (B) microplus, undergo the larval, nymphal, and adult stages on a single-host animal, feeding and molting during each phase. Upon becoming engorged, the female tick detaches from the host to lay eggs [67,94] (Figure 3).

2. Hard Ticks: An Efficient Vector for TBDs

The role of ticks and the pathogens they carry has become an increasingly prominent concern for global human and animal health [56]. Certain ticks employ an ambush strategy, residing in open environments and crawling onto vegetation to await passing hosts, a behavior known as questing [102]. While questing, ticks sway their forelegs, exposing ‘Haller’s organs’, located at the end of the first pair of legs. These organs detect various host odors, humidity, body heat, vibrations, and carbon dioxide [70,102]. Genera like Rhipicephalus, Haemaphysalis, and Ixodes have larvae, nymphs, and adult stages that engage in questing on vegetation. Ticks attach to hosts by utilizing their front legs and then traverse the skin to locate an appropriate site for attachment and feeding. Conversely, adult ticks belonging to the Amblyomma and Hyalomma genera are proactive hunters, running across the ground after nearby hosts [103].
Ticks play a crucial role in host range evolution, impacting host reproductive success and population dynamics, especially at high infestation intensities [94]. The efficient transmission of pathogens depends on vector competence, which encompasses the ability to acquire, maintain, and transmit pathogens, with factors specific to tick species and associated pathogens [104]. Experiments have shown that different tick species can carry and spread different pathogens. This highlights the need for studies that focus on specific regions and use different tick species [105,106,107,108]. For example, Ixodes scapularis is the main tick that spreads POWV II (Powassan (POW) virus lineage II), and Dermacentor variabilis and Amblyomma americanum ticks in the eastern USA can also get infected and transmit POWV II because they live in the same places and feed on the same hosts [105]. Researchers observed variations in the vector competence of Rhipicephalus appendiculatus and Rhipicephalus zambeziensis ticks in transmitting Theileria parva, which were attributed to differences in parasite stock and tick population [106]. Comparisons between I. scapularis and I. pacificus, the vectors for LD, highlighted that I. pacificus had higher attachment rates and engorgement, while I. scapularis transmitted the pathogen more efficiently [104].
Ticks can acquire multiple pathogenic microorganisms, such as bacteria, viruses and protozoans. This occurs through two main mechanisms: systemic transmission and co-feeding [109,110]. The main way that tick borne pathogens (TBPs) are spread is through horizontal (oral) transmission, which means that the pathogen moves from an infected tick to a definitive host that is not infected and back again [111]. Co-feeding transmission enables co-feeding ticks, whether infected or susceptible and potentially at different life stages, to transfer TBPs. This occurs directly, even in the absence of established pathogens within the reservoir host [112] (Figure 4). For example, LD, Thogoto virus, and TBE virus are diseases that can be transmitted from infected ticks to non-infected ticks during close-proximity feeding [113]. In systemic transmission, ticks can maintain pathogens through transstadial, sexual, and transovarial routes [114]. In the systemic transmission pathway, questing ticks obtain viruses by feeding on infected hosts, and during subsequent feeding, infected ticks can transmit the virus to susceptible hosts [115]. The transstadial transmission of pathogens from one developmental stage to the next [116] occurs in Borrelia, Rickettsia, and Anaplasma phagocytophilum [113]. Sexual transmission is observed in Rickettsia and some relapsing fevers during copulation [117].
Hosts, vectors, and pathogens engage in an ongoing antagonistic arms race, co-evolving to enhance their respective performance and fitness [118]. The route of transmission is a crucial factor shaping the co-evolutionary dynamics between hosts and parasites [118]. The tick microbiome, which comprises commensal and symbiotic obligate endosymbionts, affects the tick’s ability to grow, reproduce, adapt to feeding sources, and protect itself from environmental stresses [119]. Non-pathogenic microorganisms in ticks can influence TBP replication, either inhibiting or enhancing it, and play a crucial role in determining the outcome of TBP transmission in tick populations [120].

2.1. Blood-Feeding Mechanism of Hard Ticks

The tick-feeding process involves intricate mechanisms, including the regulation of specialized structures such as the chelicerae and the pre-oral canal, with labrum elevation, pharyngeal valve action, muscle contractions, and neural signaling [121]. During feeding, the size of larvae, nymphs, and females significantly increases, resulting in engorged individuals adopting a lenticular or egg-shaped form resembling a blood-filled sac. In a non-fed state, hard-bodied ticks exhibit a flat and elongated form, transitioning to an oval or nearly circular shape when engorged [82] (Figure 5).
Female hard ticks need a blood meal to lay eggs and can mate either while on their host (metastriate ixodids) or on or off the host (prostriate ixodids) [122]. Interestingly, adult males show minimal change in body length following feeding [81,123]. Unlike females, adult males rarely engorge over twice their body weight [124]. Male ticks of most Ixodid species display intermittent feeding behavior, with prostriate ticks experiencing spermatogenesis before adult feeding, while metastriate ticks generally require blood feeding to initiate sperm development. However, once stimulated, metastriate males can detach from hosts and actively search for female ticks to mate [8].
Male mouthparts play an active role in reproduction by penetrating the female genital pore and delivering the spermatophore. The relationship between salivary gland components and sexual variances in tick feeding or behavior remains unclear. Tick feeding studies have demonstrated that ticks in different life stages, including larvae, nymphs, and adults, possess the ability to reattach and resume feeding even if their blood meal is interrupted, a phenomenon known as “interrupted feeding” [8].
Ticks exhibit distinctive characteristics compared to other blood-feeding arthropods [125]. They rely on blood for nutrition and reproduction, employing their chelicerae to create blood pools for feeding [126]. Ticks possess a unique feeding mechanism, penetrating the host epidermis and dermal capillaries with their chelicerae and drawing in fluids exuded into the wound. Ticks have varied mouthpart adaptations for different skin penetration depths, with ixodid ticks having longer mouthparts, while argasid ticks possess well-developed chelicerae for swift penetration [5].
Ticks’ mouthparts have a hypostome with backward-pointing barbs, and many ixodid species secrete cement around the hypostome for secure anchoring [127]. The origin of cement appears to be from the cells of both type II acini and type III acini within the salivary glands [128]. The Ixodidae family exhibits attachment cement production as a distinctive trait, in contrast to its apparent absence or rarity in Argasidae. Notably, there are no reports of cement production in N. namaqua [128]. Ticks of different genera secrete varying amounts of cement, which affects their attachment to hosts [127]. Ticks in different genera produce varying amounts of cement, impacting their attachment to hosts [127]. The cement, besides its adhesive properties, is proposed to have antimicrobial functions, including sealing lesions during feeding, facilitating feeding and pathogen transmission, and protecting ticks from host immune and inflammatory responses [129].
Feeding duration varies among tick species, with ixodid ticks taking several days to a couple of weeks, while argasid ticks are notably faster, often feeding for less than an hour [23,127]. During a single feeding session, an ixodid tick can consume over 200 times its unfed body weight, significantly increasing the risk of pathogen acquisition from an infected host [27,130]. Ticks have a unique cuticle with properties distinct from other arthropods, using resilin to enhance cuticle flexibility. When ticks get swollen, big changes happen to the epithelial cells in the midgut diverticula, which makes it easier for the ticks to handle changes in volume [22]. Ticks exhibit intracellular digestion, known as heterophagy, unlike many other blood-feeding arthropods. Ticks digest their meal entirely within the epithelial cells of the midgut, allowing them to survive extended periods without feeding and harbor pathogens over time, contributing to their role as disease reservoirs [131,132]. Ticks, with their unique feeding mechanisms, digestion processes, and disease transmission capabilities, are captivating subjects in the fields of parasitology and vector-borne diseases [133].

2.2. Specialized Function of Salivary Glands in Hard Ticks

It is important for ticks to have multifunctional salivary glands (SGs) that help them stay alive and spread diseases. These glands are also used to study development and transmission routes, which has led to possible interventions that target these glands because they are so important to tick survival and vector competency [56,86,97,134,135,136]. The salivary glands of female argasid and ixodid ticks consist of numerous acini, categorized into two types in argasid ticks (types I and II) and three types in ixodid ticks (types I, II, and III). In ixodid males, there is an additional type (type IV) of acini in their salivary glands. In both male and female ticks, Type I acini primarily connect to the anterior portion of the main salivary duct, whereas Type II and III acini are linked to secondary and tertiary ducts located more distally. This organization reflects the specialized functions of these acini in tick salivary gland activity [134,136]. Distinct acini types, such as type I for hydration and type II/III for saliva production, serve specific functions within these glands [137]. Newly hatched larvae exhibit minute salivary glands with only discernible ducts. In a mature larval stage, some alveoli start to form, such as types 1, 2, and 3 alveoli observed in H. spinigera larvae. Type 4 alveoli are not distinguishable during the larval stages [109].
Tick saliva serves various functions, including water balance, holdfast and gasket formation, regulation of host responses, dynamics, individuality, mate guarding, and saliva-assisted transmission (SAT) [23,27]. SAT, which facilitates pathogen transmission through arthropod saliva, is well-documented in blood-feeding arthropods, particularly ticks [134,138,139]. SAT greatly enhances co-feeding transmission, an efficient disease transmission strategy employed by ticks [111]. Co-feeding depends on how easily pathogens can be transmitted and how well each host species can support this route. It is affected by the fact that infecting and infection-acquiring instars feed on the same host at the same time, which is linked to tick activity seasons [140]. Host reactions, such as hemostatic plug formation (initial response to vessel wall injury), involve platelets promptly engaging with any breaks in the vascular endothelium [141], and inflammatory responses aim to reject feeding ticks [23,126,134]. However, ticks counteract these defenses using biologically active molecules in their salivary glands [142]. These molecules evolved through host–parasite co-evolution and exhibit anticoagulant, antiplatelet, vasodilatory, anti-inflammatory, and immunomodulatory properties. Tick proteins, such as Prostacyclin 51 and IxscS-1E1 52, disrupt platelet aggregation. Ticks called I. scapularis and Haemaphysalis release different substances that stop and start the coagulation cascade, which changes how blood clots [13]. Ticks employ these bioactive compounds to facilitate successful feeding, ensuring their survival and reproductive success [130,132,134].
Tick saliva contains an arsenal of bioactive molecules that modulate host hemostasis and immune reactions, thus enabling blood acquisition [143]. A protein with anti-complement properties has been identified, cloned, and expressed in I. scapularis. It inhibits C3b binding and expedites the dissociation of factor Bb from the alternative pathway of C3 convertase in the host. The tick saliva and the recombinant protein exhibit comparable inhibitory effects on the complement system [144]. Hard ticks like R. sanguineus, R. appendiculatus, Dermacentor reticulatus, and Amblyomma variegatum produce proteins in their salivary glands. These proteins include evasins, which bind to chemokines [133,145]. These evasins bind to host chemokines, hindering their ability to activate chemokine receptors [145], and they serve as chemoattractants for immune cells, influencing leukocyte recruitment and host inflammation. For instance, Evasin-1 binds to CCL3, CCL4, and CCL18, while Evasin-3 binds to CXCL8 and CXCL1. Researchers have used in vitro tests and given recombinant Evasin proteins to BALB/c and C57BL/6 mice to show that they can stop inflammation [133]. The macrophage inhibitory factor (MIF) found in the saliva of A. americanum and H. spinigera ticks strongly inhibits the recruitment of neutrophils and monocytes to the attachment site [146,147]. Apyrase (ATP-diphosphohydrolase) is an enzyme that plays a role in inhibiting platelet aggregation by converting the active forms of ATP and ADP into the inactive form AMP [148] (Figure 6).
Interestingly, the innate immune responses in arthropods are predominantly coordinated through the Toll, immune deficiency (Imd), and Janus kinase (JAK)-signal transducer and activator of transcription (STAT) pathways [149]. The tick JAK/STAT pathway responds to host-derived interferon gamma during feeding, activating antimicrobial effectors like Dae2 and the 5.3-kDa AMP, which limit the proliferation of pathogens such as Borrelia burgdorferi and Anaplasma phagocytophilum. Peritrophin-1 is a tick JAK/STAT effector that plays two roles; namely, it changes the integrity of the peritrophic matrix to help B. burgdorferi survive in the gut and stop A. phagocytophilum from colonizing. The JAK/STAT pathway in ticks complicates the balance between positive and negative regulation of infection, highlighting its complex role in responding to TBPs [140].

3. Distribution and Diversity of Tick Vectors

The hard ticks exhibit diverse distribution and significance across continents. In America, prevalent hard tick genera infesting pets include Amblyomma, Dermacentor, Ixodes, Rhipicephalus, and the recently established Haemaphysalis longicornis [123,127]. Australia faces economic losses and disease transmission primarily from Ixodes, Haemaphysalis, and Rhipicephalus [150,151]. Europe, Northern Africa, and Southern Africa collectively report a total of 67 tick species, which includes the genera: Amblyomma, Dermacentor, Haemaphysalis, Hyalomma, Ixodes, and Rhipicephalus [152,153]. Avian hosts in Europe harbor 37 hard tick species, exhibiting preferences for specific hosts, such as seabirds in Western–Northern Europe (Ixodes rothschildi, I. unicavatus, and I. uriae) or associations with tortoises (Hyalomma aegyptium) and birds of prey (Rhipicephalus turanicus, R. sanguineus) [154]. In China, the Ixodidae family comprises 111 species distributed across seven genera: Amblyomma (8 species), Anomalohimalaya (2 species), Dermacentor (14 species), Haemaphysalis (43 species), Hyalomma (7 species), Ixodes (29 species), and Rhipicephalus (8 species) [155].

3.1. Distribution of Hard Ticks in India

Over the years, our knowledge of ticks in India has evolved through various contributions, including those of [156,157,158,159]. The compilation by the National Institute of Virology and other institutes includes 106 valid tick species collected from different hosts across the country [160].
Between 1997 and 2023, studies in India significantly advanced our knowledge of tick populations, revealing their diverse presence and distribution. Researchers in India have identified nine genera of hard ticks, which include Amblyomma, Aponomma, Haemaphysalis, Hyalomma, Ixodes, Rhipicephalus, Boophilus, Dermacentor, and Nosomma [160,161]. Kerala, for instance, has reported various tick species on domestic animals [162,163,164,165]. The species of Haemaphysalis, Amblyomma, Ixodes, and Rhipicephalus were documented in the Western Ghats [166].
Researchers have identified several tick species in Tamil Nadu, including Amblyomma integrum, Haemaphysalis bispinosa, and R. turanicus, among others [65,167,168]. Andhra Pradesh reports eight tick types, including Rhipicephalus (B) microplus and Hyalomma marginatum [169,170]. Other states like Chhattisgarh, Odisha, Maharashtra, Gujarat, Rajasthan, and Uttar Pradesh also document diverse tick genera and species [171,172,173,174,175,176,177]. This extensive literature underscores the critical importance of studying ticks and TBDs in India, offering valuable insights into their distribution and diversity (Figure 7). Table 1 provides an overview of important tick-borne diseases and their respective vector distributions in India.

3.2. Genetic Diversity of Ticks

Studies in population genetics related to wingless arthropod vectors, like ticks, have provided valuable insights into their dispersal patterns. These patterns are closely tied to host mobility and behaviors, significantly influencing genetic structure and disease transmission dynamics [178]. Researchers utilized molecular markers to address questions about genetic variability, population genetic structure, gene flow, and potential genetic isolation by distance in one or more tick species [59,74,178]. Additionally, newer studies have employed whole-genome sequencing and functional genomics, recognizing the potential of next-generation approaches in advancing our understanding of tick biology and evolution [178,179].
Understanding genetic diversity, stemming from factors like DNA recombination, mutations, gene flow, and genetic drift, is crucial [180]. Researchers commonly use mitochondrial genomes in molecular systematics because they undergo faster evolutionary changes [181]. In ticks, the genomes of 66 species from 18 genera have been sequenced, offering consistent genus-level classification [182].
The impact of host mobility on the genetic composition of tick populations is variable. Studies on I. scapularis and Ornithodoros coriaceus reported limited gene flow despite their hosts’ high mobility [178]. In contrast, ticks associated with less mobile hosts exhibit reduced gene flow, while those connected to highly mobile hosts show higher gene flow. For example, A. americanum and A. triste displayed increased gene flow attributed to the dispersal capabilities of their hosts in Arkansas ecoregions, Georgia, and Argentina [183]. The genetic diversity of Rhipicephalus sanguineus s.l. ticks in Colombia, which are known to be relevant in TBD transmission, is important for a complete study of tick-borne disease epidemiology [184].
High genetic diversity and low differentiation observed in Rhicephalus (B) microplus populations suggest an infinite island model in migration-drift equilibrium in the provinces of Zimbabwe [185]. Gene flow through cattle movement was noted, leading to allele loss and founder effects (refers to the decrease in genomic variability that arises when a small group of individuals becomes isolated or separated from a larger population during migration), potentially impacting tick adaptation in diverse habitats [186]. Host species influence the genetic structure of A. dissimile ticks in a 500 km area, resulting in the formation of small breeding groups [187]. Host mobility during immature stages contributes to the distinct genetic structures observed in two Ixodid ticks, Haemaphysalis flava and Ixodes ovatus, in Niigata Prefecture, Japan. I. ovatus has a stronger genetic structure because its small mammal host does not move around much, while H. flava does not have any genetic structure because its host probably moves around a lot with birds [188].
A phylogenetic tree generated by the 16S mitochondrial gene from Rhipicephalus sanguineus available in the database showed mixed genetic patterns. We observed two prominent groups. Group I consisted of ticks from South America, Africa, and Asia, and Group II predominantly represented ticks from European regions (Figure 8). However, population structure insights on important tick vectors across different geographic locations are essential for assessing disease transmission dynamics, host adaptation, and overall tick ecology.
Hybridization, a natural phenomenon, plays a pivotal role in the emergence of new species on Earth [189]. In nature, ticks engage in hybridization activities, supported by both observational evidence and experimental studies. Researchers have documented natural hybridization between Ixodes ricinus and I. persulcatus ticks in Estonia [190]. Furthermore, laboratory studies have reported hybridization in ticks of the genus Ixodes for several species [191]. A recent study that looked at how the tick-borne encephalitis virus (TBEV) is spread in I. ricinus and I. persulcatus ticks, including hybrids, found that many complicated factors affect how TBEV is spread [192].

4. Impacts of Climate Change on the Expansion of Hard Ticks

Climate change has an impact on tick populations since it can modify their habitat and areas where ticks can persist over extreme weather conditions. Ticks exhibit diverse distribution patterns globally, which are influenced by climatic factors like temperature, humidity, altitude, vegetation types, and host availability [193,194,195,196]. Climate change, especially the rapid rise in global temperatures, has played a significant role in the expanded range of arthropod vectors, including ticks [140,197,198,199]. Recently, a warmer climate in the northeastern USA has been linked to the geographical expansion of an aggressive human-biting tick A. americanum, commonly known as the lone star tick, impacting its range and population dynamics, and presenting a growing public health challenge [200]. Also, warmer climates are associated with accelerating the maturation rates of I. scapularis ticks which may likely elevate the prevalence of LD in the northeastern United States by extending the transmission season [201]. Projected future climatic conditions in Europe are expected to increase the suitability of habitats for Ixodes ricinus, D. reticulatus, and Dermacentor marginatus, resulting in expanded geographical ranges, especially towards Eastern Europe [202]. In Russia, climate changes are responsible for the expansion of I. persulcatus toward newer areas like Russia’s Arkhangelsk Oblast (AO) and Komi Republic [203]. In China, I. ovatus is distributed mainly in southwest and northwest China, but changes in the regional temperature profile and annual precipitation may influence its habitat distribution, with a predicted expansion toward northeast China in the future [204]. Climate change, particularly rising temperatures, may adversely affect tick species in tropical zones, forcing some to colonize new areas and leading to changes in habitat suitability, such as the dispersion of ticks like Amblyomma variegatum into regions beyond those with prolonged dry periods, as observed in Zimbabwe [197].
Ticks have evolved sophisticated life-history strategies, including behavioral and physiological adaptations, to maximize host exploitation [197,198]. Climate change’s indirect impacts on host communities might promote the increased transmission of TBPs through alterations in tick abundance [201,205]. Understanding factors like host specificity, habitat suitability, and climate tolerance is crucial for comprehending tick range expansion [200]. Climate and host availability play a crucial role in shaping tick life cycles and feeding physiology, leading to their shift from predatory to ectoparasitic lifestyles [5]. An increase in the availability of small mammal host density correlates with heightened questing nymph and adult tick densities, reflecting accelerated progression through larval and nymphal stages [198,199,206] (Figure 9).
Host–parasite associations, particularly host-specificity relationships, play a role in constraining many tick species [207]. A variety of vertebrates act as reservoirs for tick-borne pathogens, contributing to the persistent nature of these diseases and posing risks to domestic animals and humans [201,208]. Understanding the relationship between host diversity and disease risk is complex, with global trends suggesting that higher host biodiversity can increase tick exposure. However, local effects may vary, and introducing highly competent hosts can raise transmission risk [94]. Wildlife vertebrate hosts play a crucial role in the ongoing cycles of TBPs. In some instances, they have been significant factors in the recent surge of ticks and tick-borne illnesses [30,209]. Various vertebrates, including lizards, birds, hedgehogs, snakes, and mammals, serve as reservoirs for pathogens; for example, wild boars are known reservoirs for many tick-borne viruses, bacteria, and parasites, which play a crucial role in the sylvatic cycle [210,211,212]. Rodents from various families serve as zoonotic reservoirs for ectoparasites like mites and ticks, playing a vital role in the transmission of various vector-borne diseases such as scrub typhus, and TBDs [210]. Wild animals, including species like wild boars, lizards, and snakes, commonly act as extensive and frequently undiscovered reservoirs of hosts for zoonotic diseases, particularly those transmitted by ticks [212,213].
Both large-wildlife loss and climatic changes can independently influence the prevalence and distribution of zoonotic disease. The decline of large-wildlife populations globally disrupts ecological functions like disease control, potentially impacting tick abundance and tick-borne disease risk due to altered host dynamics and vegetation structure. Understanding the interplay between host loss, vegetation changes, rainfall and tick survival is crucial for assessing the overall impact on vector-borne disease transmission [214,215]. In India, studies on the impact of climate change on the expansion of Ixodid ticks have not been documented.

5. Important TBDs in India Transmitted by Hard Ticks

In India, TBDs pose a significant public health concern, presenting unique scenarios such as KFD [216,217], CCHF [47,218,219], rickettsial infections like Indian Tick Typhus (ITT) [220], LD [221], Q-Fever [222], and anaplasmosis [223].

5.1. Kyasanur Forest Disease (KFD)

In 1957, researchers first identified KFD in the Kyasanur forest of India. Caused by the KFDV, a member of the Flavivirus family, it can induce hemorrhagic illness in various animals [224]. The primary mode of transmission is through ticks, particularly H. spinigera [225,226,227]. Common symptoms include chills, headaches, muscle aches, vomiting, bleeding, and high fever, with a mortality rate ranging from 3–5% [41,46,216,217].
Ticks, prevalent in regions characterized by drier and warmer climates, exhibit a 58.66% prevalence in Karnataka (Shivamogga, Chamrajnagar, and Chikmagalur districts), 12.02% in Kerala (Kozhikode and Wayanad districts), 18.15% in Maharashtra (Raigad and Ratnagiri districts), and in the Nilgiris districts of Tamil Nadu and North Goa district in Goa. Nymphal activity occurs during the non-rainy season [226].
Ticks, mammals, and birds maintain the KFD virus through cycles. Monkeys, particularly Macaca radiata (bonnet macaque) and langurs in the genus Semnopithecus, are susceptible and often fatal hosts, yet they serve as important reservoirs. Cattle, the primary hosts for the ticks, do not amplify the virus, and their role in KFD transmission requires further investigation [227,228]. Tick species such as Haemaphysalis turturis, Haemaphysalis kinneari, Haemaphysalis kyasanurensis, Haemaphysalis wellingtoni, Haemaphysalis minuta, Haemaphysalis cuspidata, Ixodes petauristae, Ixodes sceylonensis, Dermacentor auratus, and Rhipicephalus haemaphysaloides have been reported in eleven out of 29 states and 7 union territories in India, and they are capable of carrying the virus [45,229].

5.2. Crimean–Congo Hemorrhagic Fever (CCHF)

CCHF virus, belonging to the Nairovirus genus within the Bunyaviridae family, is responsible for causing severe illness in humans [230]. Since its initial identification during a 1944 outbreak in the West Crimean region of the former Soviet Union and official isolation in 1956, CCHF has been documented in various regions, including parts of Africa, Asia, Eastern Europe, and the Middle East [47]. CCHF is a tick-borne viral disease with an average mortality rate ranging from 30% to 50% [47,49,231].
During a nosocomial outbreak in 2011, CCHF was first confirmed in India, specifically in Gujarat State. Subsequently, Gujarat has experienced numerous outbreaks and sporadic cases of CCHF [49]. In 2014 and 2015, a nosocomial outbreak was recorded in a private hospital in the neighboring state of Rajasthan. The virus was detected in the southern Indian state of Kerala in 2016, associated with the travel history of a slaughterhouse worker who came from Oman [218]. In 2019, there was an outbreak in Gujarat that reportedly had a mortality rate exceeding 50% [219].
Domestic animals and Hyalomma ticks are involved in amplifying and transmitting the CCHF virus, although the virus has also been isolated from Rhipicephalus, Boophilus, and Dermacentor species, which may serve as additional vectors [232]. Various factors such as land use changes and climate have been identified as crucial drivers of CCHF infections [218]. The major routes of CCHF transmission to humans include nosocomial infections, tick bites, crushing ticks with bare hands, and contact with the blood or tissue fluids of infected animals or humans. Available information suggests that CCHF cases mainly result from occupational exposure among abattoir workers, farmers, veterinarians, and healthcare workers [219]. A wide range of vertebrates, such as cattle, goats, donkeys, and horses, along with smaller wildlife species like hares and hedgehogs, act as reservoirs for the CCHF virus [233,234].

5.3. Lyme Disease (LD)

In 1976, LD garnered attention when a cluster of juvenile arthritis cases in Old Lyme, Connecticut, accompanied by skin lesions resembling those from European tick bites, raised suspicions of a shared infectious agent [235]. LD is primarily caused by the B. burgdorferi spirochete. The increasing risk of LD in the United States is mainly attributed to the expanding range of the blacklegged tick, I. scapularis, the principal vector for the spirochetal pathogen B. burgdorferi [236]. It predominantly affects the skin, nervous system, heart, and joints [221,236]. In Lyme borreliosis, the infection process involves adaptations for survival and replication in the mammalian host, evading host immune mechanisms [237,238]. A literature search revealed a rare cluster of five LD cases over three months in Rohtak, Haryana, a non-endemic region of India. The patients, including a farmer, a boy, and housewives, presented with distinct skin manifestations following tick or insect bites, with serological tests confirming B. burgdorferi infection. These cases highlight an unusual occurrence in a region not traditionally associated with LD [40]. A study in North India involving 252 individuals found 18 cases of LD. They used a standard two-tiered testing algorithm (STTA) with ELISA and immunoblot assays. Intrathecal IgG synthesis indicated a 7.14% antibody response to B. burgdorferi infection. Molecular evidence showed B. burgdorferi sensu lato and Anaplasma phagocytophilum in neuroborreliosis patients [221].

5.4. Tick-Associated Rickettsial Pathogens

Rickettsiae are intracellular endosymbionts. They are spread by blood-sucking ectoparasites like ticks, fleas, and lice [239]. Approximately 24% of terrestrial arthropods carry Rickettsia endosymbionts [240]. Infections caused by Gram-negative bacteria like Rickettsia, Orientia, Ehrlichia, Neorickettsia, Neoehrlichia, and Anaplasma are the common pathogens involved in typhus and spotted fever groups [241,242].
The dog tick R. sanguineus is the primary vector for ITT (Indian Tick Typhus) in India, although certain species of Haemaphysalis and Hyalomma ticks can also transmit the infection. Other than scrub typhus (non-scrub typhus rickettsial diseases), rickettsial diseases, including spotted fever group (SFG) rickettsioses and typhus group (TG) rickettsioses, are not uncommon in India [243]. Delhi, Himachal Pradesh, Jammu and Kashmir, Uttarakhand, Rajasthan, Tamil Nadu, and Maharashtra have reported several cases. In the early years, cases were mainly reported from soldiers on the Assam and Burma fronts during the Second World War, when it was a problem second only to malaria [244]. Globally, diverse SFG rickettsioses occur, highlighting the need for regional understanding and control measures [245].

5.5. Eco-Epidemiology of TBDs

The dynamics of TBPs, encompassing their occurrence and abundance, are influenced by ecological interactions between tick species and their vertebrate hosts [246]. The expansion of tick species intricately links to the complex dynamics of TBPs, which rely on the abundance and distribution of hosts that sustain tick populations and act as reservoirs for these pathogens through intricate demographic processes and movement dynamics [247] (Figure 8).
Ecological epidemiology explores the interactions between hosts and their pathogens at the population and community levels, encompassing infectious disease studies in both human and wildlife populations [248,249]. Many human diseases transmitted by arthropods, insects, or other invertebrates or vertebrates originate in animals [21]. Alterations to ecosystems through infrastructure development, such as deforestation and converting forestlands for urbanization, contribute to changes in disease epidemiology [250]. Diseases often emerge from pathogens sustained in complex natural transmission cycles involving interacting host and vector communities. The composition of these communities significantly influences pathogen amplification and the risk of spillover transmission [251]. The complicated eco-epidemiology of TBP transmission cycles is mostly because tick vectors have a life cycle that changes and adapts. Climate change also affects arthropod populations and the dynamics of reservoir hosts [252] (Figure 9).
Dynamic interactions among biotic and abiotic elements shape the epidemiology and ecology of TBDs [253]. The pathogens that they carry or the disease they transmit may affect their distribution areas and their population densities. These changes can be caused by climate change, human activities, land use, economic and political factors, and changes that happen naturally in ticks and the diseases they carry [254].
KFD virus was limited to Karnataka’s Shimoga, Chikmagalur, Uttara Kannada, Dakshina Kannada, and Udupi districts in India [255]. Since 2012, the virus has extended to Nilgiris in Tamil Nadu, and cases were reported in Kerala, Goa, and Maharashtra from 2013 to 2017, raising concerns due to the shared Western Ghats ecology in these regions [44]. The potential transmission of KFD poses a significant threat to residents and workers in forested areas in these regions. The movement of monkeys and rodents further compounds the risk, serving as hosts for vector ticks perpetuating the KFD virus in the natural environment [44,255]. The intricate eco-epidemiology of TBP transmission, exemplified by the CCHF virus, demands a nuanced understanding of tick vectors, climate influences, and reservoir host dynamics.

5.6. Molecular Diagnostics and Emerging TBDs

The effective management of TBDs is very important and requires efficient diagnostic tools for correct treatment [256]. Serology-based diagnostics have been used for decades, but initial non-specific clinical presentations and the delayed appearance of specific antibodies (15–26 days) for serological diagnosis can mask the identification of TBDs, hindering timely and impactful confirmation by specific laboratory testing [257]. The Weil–Felix Test (WFT), Latex Agglutination Assays (LAA), and Western blots commonly used for the diagnosis of TBDs are low in specificity and sensitivity. Furthermore, ELISA, a widely used diagnostic assay, struggles with varying specificity and sensitivity; for example, an FDA-licensed ELISA for anti-IgM Rickettsia typhi exhibits 45% sensitivity and 98.3% specificity [258,259]. The cross-reactivity of antibodies between different pathogens or strains can lead to false-positive test results, further complicating the situation [260].
In recent times, the use of molecular diagnostics for TBPs has shown many advantages as they are technically straightforward, more specific, and sensitive [260,261]. Different types of PCRs (polymerase chain reactions) have been used for the clinical diagnosis of TBPs [262]. Conventional PCR involves amplifying target genes and visualizing it on an agarose gel; it can be further sequenced for phylogenetic analysis of the pathogens [261,262]. Recent real-time (RT) PCR methods, employing chemistries like SYBR Green (binds specifically to double-stranded DNA) and TaqMan (fluorogenic probe for specific PCR product detection during amplification cycles), offer advantages such as the quantification of pathogens, high sensitivity, and specificity [263,264]. TaqMan-based multiplex RT PCRs are another notable development that allows for the detection of multiple pathogens in single-tube reactions [265]. For example, the newly developed Tick Path Layerplex assay employs RTPCR technology to simultaneously detect and characterize 11 pathogens responsible for these diseases in domestic dogs, and it shows high sensitivity and compatibility with standard diagnostic instruments [266]. When used with serologic testing, whole blood real-time polymerase chain reaction (WB-RTPCR) improves the ability to diagnose early LD (ELD) [267]. Multi-locus sequence typing (MLST) enhances genotypic characterization for population studies [268]. For example, MLST identified nine Rickettsia species, including six human pathogens, from tick DNA samples, highlighting diverse rickettsial species diversity [269]. Next-generation sequencing (NGS) has greatly advanced our knowledge of tick-borne pathogens and their impact on human health, uncovering new microbes and providing insights into tick microbiome diversity. These studies also contribute to understanding evolutionary relationships, identifying diagnostic targets, and innovating approaches to tackle tick-borne diseases (TBDs) [270].

6. Challenges Associated with the TBDs

Managing ticks and TBDs poses a significant challenge due to variations in diagnostics, treatment, disease surveillance, control, and management strategies [271,272,273]. Ticks, acting as vectors and reservoirs of pathogens, cause various diseases in humans and animals and are responsible for substantial harm to livestock [26]. Apart from causing diseases in humans and animals, tick bites can trigger severe allergic reactions in humans [28,274]. For example, alpha-gal syndrome is a rare allergic reaction to a sugar molecule called alpha-gal, injected through tick bites, leading to the delayed onset of allergic symptoms triggered by the consumption of red meat [275]. Additionally, these vectors have indirect repercussions on livestock, particularly affecting the dairy industry. The costly and singular focus of diagnostic methods discourages cattle herders from screening for these infections [276]. Controlling ticks in domestic animals to prevent disease transmission, avoid tick paralysis or toxicosis, and mitigate the physical damage caused by ticks is emphasized in India, which has the world’s largest population of cattle at 192.52 million and buffaloes at 109.85 million [52,277].

6.1. Strategies to Control TBDs

With the rise in TBD cases and the spread of pathogens to new regions, the demand for effective tick management strategies becomes crucial [277]. Integrated tick management encompasses components such as public education, routine inspection and surveillance, disease diagnostics, and the application of control measures [29]. It combines various methods to minimize the reliance on acaricide treatments. Alternatives include employing anti-tick vaccines, implementing pasture management (which is the practice of growing healthy forage grasses and legumes that ensure a lasting food source for livestock) [278], and utilizing tick-resistant cattle breeds [279]. To enhance control efforts, there is a necessity for Integrated Tick Management (ITM) on larger scales, employing diverse strategies to impact tick populations in residential areas, greenspaces, and public lands [280,281]. Currently, tick population suppression relies primarily on individual homeowners, lacking coordinated efforts at neighborhood or community scales [29]. The effective management of ticks and tick-borne diseases requires the integration of various rational tactics, encompassing biological, chemical, physical, and vaccine technologies on and off hosts [282]. Crucial to this is the safer application of acaricides, considering environmental concerns. Area-wide tick management research should prioritize understanding tick biology, ecology, and behavior, adopting a One Health approach that encompasses human, animal, and environmental health [283].

6.2. Acaricides and Biological Control

Different methods exist for tick control, each with its own limitations [52]. Acaricides are chemicals that are used to get rid of acari. These chemicals are grouped by where they come from or what they are composed of, and they include plant extracts, organochlorines, organophosphates, carbamates, formamidines, synthetic pyrethroids, and macrocyclic lactones [52,284]. Effective tick control primarily occurs on the host, especially for multi-host ticks that spend the majority of their time off the host, with brief feeding periods lasting between four and ten days [285]. Active compounds such as azadirachtin, carvacrol, linalool, geraniol, and citronellal suggest that plant-based compounds could be useful for farmers in rural areas who raise animals for food [286]. In the 20th century, scientists studied tick biocontrol agents like pathogens, parasitoids, and predators. This led to a switch from chemical acaricides to biological control because of health and environmental concerns, cost, and rising pesticide resistance [287]. Classical biocontrol introduces non-native agents to control pests, while augmentative biocontrol uses native predators or parasites. Potential natural enemies for tick control include insectivorous birds, parasitoid wasps, nematodes, bacteria, and fungi, with entomopathogenic fungi being a promising option [286,287,288]. An often-overlooked method involves the careful examination of pets for attached ticks and the removal and destruction of each tick, providing a simple yet effective means of tick control [289]. In summary, a comprehensive approach to tick control involves balancing the efficacy of chemical interventions with the environmentally friendly and sustainable aspects of biological control, recognizing the limitations of each method.

6.3. Acaricide Resistance and Mechanism

There are more reports of resistant tick populations around the world [284,290,291]. This is a big problem because it means that tick populations that are exposed to acaricides are passing on traits that make them resistant. Common acaricides target the tick nervous system through various mechanisms, including antagonism, inhibition, modulation, and activation [284,290,291]. Notably, there is a big rise in populations that are not killed by acaricides, mainly ticks like R. (B) microplus, which shows that more research is needed [290]. The study of acaricide resistance mechanisms focuses on target-site insensitivity. It has been found that changes in important nerve function channels, like voltage-gated Na+ and K+ channels, cause resistance [291]. Metabolic detoxification, which includes more esterases, cytochrome P450, and glutathione S-transferase, seems to be the main way that acaricide-resistant ticks stay healthy [292,293]. Also, ABC transporters, especially P-glycoproteins, are linked to ticks’ multidrug resistance, which shows how complicated acaricide resistance mechanisms are [291].
From a historical point of view, acaricide resistance is a trait that is passed through the vertical route of transmission and changes over time, causing phenotypic resistance, tolerance, and cross-resistance between acaricides that are chemically similar [284]. The study stresses how important it is to keep an eye on field tick populations to see if they are becoming resistant. It suggests a number of tests, such as the larval packet test (LPT), the larval immersion test (LIT), the adult immersion test (AIT), and the larval tarsal test (LTT), which can be used to perform a correct diagnosis [284,291,294]. A case study along the Texas–Mexico border underscores the significance of vigilant monitoring and rapid detection of acaricide resistance. The systematic program, employing the LPT, identified resistance to coumaphos and growing concerns about permethrin resistance. Genetic studies showed different types of fever ticks that came from different places and could infest again. This shows how important genetic tools are for making quick decisions about tick control programs [295].

6.4. Drugs/Vaccines against TBDs

The global incidence of tick-borne diseases is increasing each year [26]. Beta-lactam and tetracycline antibiotics, such as doxycycline, have proven effective in treating Lyme disease, Rocky Mountain spotted fever (RMSF), and human ehrlichiosis [296]. In endemic areas, doxycycline is preferred for its reduced frequency of administration and lower adverse effects due to the rising prevalence of coinfections involving B. burgdorferi and the human granulocytic ehrlichiosis agent [297]. Ivermectin is a macrocyclic lactone endectocide that works very well against both endo- and ecto-parasites and has antiparasitic effects that last for a long time. Its use, particularly in long-acting formulations like 3.15%, is widespread for controlling R. (B) microplus infestations in cattle, and oxytetracycline was the predominant drug used on farms in various regions [298]. In Brazil, research on the long-term and therapeutic effects of doramectin showed that it effectively got rid of R. (B) microplus infestations in cattle and kept them from coming back [299].
Vaccines, using the “isolate-inactivate-inject” method, offer a cost-effective strategy against tick-borne diseases [300,301]. Initially using live or killed pathogens, second-generation vaccines with purified components have emerged with technological advancements [301]. In Europe, widely utilized TBE vaccines (formalin-inactivated) include FSME-IMMUN and Encepur, while Russian alternatives such as IPVE and EnceVir are also available for protection against TBEV [302,303,304].
In India, formalin-inactivated KFDV vaccines have been used for decades [305,306]. Currently, there is no definitive treatment for KFD, underscoring the importance of vaccination as a crucial public health measure to manage the disease [305]. The live-attenuated VSV-based KFDV vaccine shows promise, demonstrating safety, eliciting robust immune responses, and providing effective protection in both mouse and macaque models, but it is not yet available for humans [307].
In cattle, anti-tick vaccine (immunological formulations designed to stimulate the host’s immune system, providing protection against tick infestations and the transmission of associated pathogens [308]) success primarily centers around Bm86-based vaccines like GavacTM; however, challenges are posed by tick genetic diversity [308,309]. Subolesin-based vaccines offer promising protection against multiple tick species with efficacy of 80–97%, comparable to or surpassing other tick antigens like Bm86/Bm95, metalloprotease, ribosomal protein P0, ferritin 2, and aquaporin [310].
Table 1. Important TBDs and distribution of vectors in India.
Table 1. Important TBDs and distribution of vectors in India.
DiseasesTick SpeciesDistributionAuthor
Kyasanur Forest Disease (KFD)Haemaphysalis spinigeraKarnataka, Tamil Nadu, Kerala, Andhra Pradesh, Orissa, Madhya Pradesh, Meghalaya, Goa, Maharashtra, Gujarat, West Bengal, Andaman and Nicobar Islands[45,217,224,229,311]
Haemaphysalis turturisKarnataka, Tamil Nadu, Kerala and Uttar Pradesh[45,217,224,229,311]
Haemaphysalis wellingtoniKarnataka, Orissa, Andaman and Nicobar Islands and West Bengal[45,224,311]
Haemaphysalis cuspidataSri Lanka and Karnataka[224,229]
Haemaphysalis aculeataKarnataka[229,311]
Haemaphysalis bispinosaAndhra Pradesh, Arunachal Pradesh, Assam, Bihar, Goa, Gujarat, Himachal Pradesh, Jammu and Kashmir, Karnataka, Madhya Pradesh, Maharashtra, Mizoram, Orissa, Punja, Sikkim, Tamil Nadu, West Bengal[160,224,311]
Haemaphysalis kyasanurensisKarnataka[45,224,311]
Haemaphysalis minutaHimachal Pradesh, Karnataka, Orissa and Uttar Pradesh[45,311]
Haemaphysalis kinneriKarnataka and West Bengal[45,311]
Haemaphysalis papuanakinneariKarnataka and other parts of South East Asia[311]
Ixodes petauristaeKarnataka[45,225]
IxodesceylonensisKarnataka and Tamil Nadu[45,225]
Rhipicephalus haemaphysaloidesThroughout India[45,225]
Dermacentor auratusArunachal Pradesh, Assam, Bihar, Himachal Pradesh, Jammu and Kashmir, Orissa, Karnataka, Madhya Pradesh, Maharashtra, Punjab, Uttar Pradesh and West Bengal[45,225]
Hyalomma marginatum isaaciArunachal Pradesh, Bihar, Delhi, Gujarat, Himachal Pradesh, Jammu and Kashmir, Karnataka, Maharashtra, Madhya Pradesh, Orissa, Punjab and Uttar Pradesh[225]
OrnithodorusAndra Pradesh, Jammu and Kashmir, Himachal Pradesh, Uttar Pradesh, Karnataka, Maharashtra, Madhya Pradesh, Tamil Nadu and Pondicherry[225,312]
Crimean-Congo haemorrhagic fever (CCHF)Hyalomma anatolicumAndhra Pradesh, Assam, Delhi, Gujarat, Haryana, Himachal Pradesh, Jammu and Kashmir, Karnataka, Maharashtra, Madhya Pradesh, Orissa, Punjab and Rajasthan[17,233,312,313]
H. marginatumArunachal Pradesh, Bihar, Delhi, Gujarat, Himachal Pradesh, Jammu and Kashmir, Karnataka, Maharashtra, Madhya Pradesh, Orissa, Punjab and Uttar Pradesh[17,38,160,312,313]
Hyalomma asiaticum_[17,38,160,233]
Hyalomma truncatum_[38,160,233]
Hyalomma rufipes_[38,160,233]
Hyalomma lusitanicum_[233]
Hyalomma excavatum_[233]
Hyalomma dromedariiAndhra Pradesh, Gujarat, Himachal Pradesh, Jammu and kahmir, Maharashtra, Orissa, Punjab and Uttar Pradesh[17,38,313]
Rhipicephalus sanguineusThroughout India[17,160,233,313]
Ixodes ricinusNortheastern States of India[31,163]
Indian Tick Typhus (ITT)R. sanguineusThroughout India[38,61,242,314]
Ixodes ricinusNortheastern States of India[38,314]
Haemaphysalis indicaBihar, Gujarat, Himachal Pradesh, Jammu and Kashmir, Karnataka, Maharashtra, Orissa, Rajasthan, Uttar Pradesh and West Bengal[244]
H. kinneriKarnataka and West Bengal[244]
H. spinigeraKarnataka, Tamil Nadu, Kerala, Andhra Pradesh, Orissa, Madhya Pradesh, Meghalaya, Goa, Maharashtra, Gujarat, West Bengal, Andaman and Nicobar Islands[244]
H. turturisKarnataka, Tamil Nadu, Kerala and Uttar Pradesh[244]
Haemaphysalis leachiEastern West Pakistan, India, and Ceylon, and the terai of southern Nepal[244]

7. Conclusions

This comprehensive review explores various facets of tick biology, delving into morphology, blood-feeding mechanisms, and the profound impact of salivary glands on host immunity. By dissecting distribution patterns and life cycles, the review provides valuable insights into the intricate ecological dynamics governing tick populations. Given that ticks serve as vectors for a range of diseases, understanding their biology is crucial for public health reasons. The discussion on important tick-borne diseases emphasizes the urgent need for effective control measures. The multifaceted approach advocated for in this review goes beyond tick control, encompassing advancements in vaccine development and population genetics. It calls for progress in vaccine development, recognizing immunization as a pivotal strategy against tick-borne diseases. Simultaneously, a nuanced understanding of population genetics is crucial for predicting adaptability to environmental changes and devising targeted interventions. The review underscores the significance of population genetics not only as an academic pursuit but as a practical tool for predicting and influencing tick population behavior. Geographical and range expansions, influenced by factors like climatic variations, highlight the urgency of anticipating and mitigating the spread of ticks and associated diseases. In essence, this review underscores the interdependence of biological, ecological, and genetic factors in the intricate tapestry of tick-related dynamics. The call for a holistic and interdisciplinary approach reflects the acknowledgment that addressing the challenges posed by ticks and tick-borne diseases requires a comprehensive understanding. This approach not only tackles current issues but also lays the groundwork for anticipating and adapting to the evolving landscape of ticks and associated diseases. The synthesis of these diverse elements encapsulates the complexity of the task at hand and emphasizes the need for collaborative efforts across various scientific disciplines to effectively address this public health concern. This review serves as a valuable resource for researchers and individuals seeking comprehensive information about tick-borne diseases and their evolutionary nature in the context of a constantly changing climate environment, often linked to human actions.

Author Contributions

Conception: S.A.N., N.P. and R.S.; Data collection and curation: N.P.; Illustrations: N.P.; First draft preparation: N.P., R.S. and M.S.; Critical revision of the article: S.A.N. and M.S.; Resources: S.A.N.; Final edits: S.A.N. and R.S. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Acknowledgments

The authors express their deep sense of gratitude to the Director, ICMR—VCRC for extending all the facilities to undertake this review.

Conflicts of Interest

The authors declare that they have no competing interests.

Abbreviations

TBDTick-borne diseases
KFDKyasanur Forest Disease
CCHFCrimean–Congo haemorrhagic fever
LDLyme disease
TBPTick-Borne Pathogen
POWV IIPowassan (POW) virus lineage II
SATSaliva-Assisted Transmission
MIFMacrophage migration Inhibitory Factor
JAK/STATJanus kinase (JAK)–signal transducer and activator of transcription (STAT) pathways
TBETick-Borne Encephalitis
ITTIndian Tick Typhus
PCRPolymerase Chain Reaction

References

  1. Jankielsohn, A. The Importance of Insects in Agricultural Ecosystems. Adv. Entomol. 2018, 6, 62–73. [Google Scholar] [CrossRef]
  2. Redak, R. Introduction to and Importance of Insects. In Forest Entomology and Pathology; Allison, J.D., Paine, T.D., Slippers, B., Wingfield, M.J., Eds.; Springer: Cham, Switzerland, 2023; Volume 1, pp. 1–17. [Google Scholar] [CrossRef]
  3. Baxter, R.H.G.; Contet, A.; Krueger, K. Arthropod Innate Immune Systems and Vector-Borne Diseases. Biochemistry 2017, 56, 907–918. [Google Scholar] [CrossRef]
  4. Khan, A.; Yasin, M.; Aqueel, M.A.; Farooqi, M.A.; Akram, M.I.; Yousuf, H.M.B.; Noor, M.; Maqsood, A. Vector-Borne Disease and Climate Change; IntechOpen eBooks; IntechOpen: London, UK, 2023. [Google Scholar]
  5. Cupp, E.W. Biology of Ticks. Vet. Clin. N. Am. Small Anim. Pract. 1991, 21, 1–26. [Google Scholar] [CrossRef]
  6. Brites-Neto, J.; Duarte, K.M.R.; Martins, T.F. Tick-borne infections in human and animal population worldwide. Vet. World 2015, 8, 301–315. [Google Scholar] [CrossRef] [PubMed]
  7. Yu, Z.; Wang, H.; Wang, T.; Sun, W.; Yang, X.; Liu, J. Tick-borne pathogens and the vector potential of ticks in China. Parasites Vectors 2015, 8, 24. [Google Scholar] [CrossRef] [PubMed]
  8. Tahir, D.; Meyer, L.; Fourie, J.; Jongejan, F.; Mather, T.; Choumet, V.; Blagburn, B.; Straubinger, R.K.; Varloud, M. Interrupted Blood Feeding in Ticks: Causes and Consequences. Microorganisms 2020, 8, 910. [Google Scholar] [CrossRef]
  9. Varma, M.R.G. Ticks and mites (Acari). In Medical Insects and Arachnids; Springer: Dordrecht, The Netherlands, 1993; pp. 597–658. [Google Scholar] [CrossRef]
  10. Cordeiro, F.A.; Amorim, F.G.; Anjolette, F.A.P.; Arantes, E.C. Arachnids of medical importance in Brazil: Main active compounds present in scorpion and spider venoms and tick saliva. J. Venom. Anim. Toxins Incl. Trop. Dis. 2015, 21, 24. [Google Scholar] [CrossRef]
  11. Francis, S.; Frank, C.; Buchanan, L.; Green, S.; Stennett-Brown, R.; Gordon-Strachan, G.; Rubio-Palis, Y.; Grant, C.; Alexander-Lindo, R.L.; Nwokocha, C.; et al. Challenges in the control of neglected insect vector diseases of human importance in the Anglo-Caribbean. One Health 2021, 13, 100316. [Google Scholar] [CrossRef]
  12. Kader, S.; Arriaza, R.H.; Khatri, K.; O’malley, A.; Grbic, V.; Grbic, M.; Chruszcz, M. Current status of structural studies of proteins originating from Arachnida. Syst. Appl. Acarol. 2023, 28, 298–308. [Google Scholar] [CrossRef]
  13. Snodgrass, R.E. Principles of Insect Morphology; Comstock Publishing Associates: Ithaca, NY, USA, 1993. [Google Scholar]
  14. Randolph, S. Ticks are not Insects: Consequences of Contrasting Vector Biology for Transmission Potential. Parasitol. Today 1998, 14, 186–192. [Google Scholar] [CrossRef] [PubMed]
  15. Charrier, N.P.; Hermouet, A.; Hervet, C.; Agoulon, A.; Barker, S.C.; Heylen, D.; Toty, C.; McCoy, K.D.; Plantard, O.; Rispe, C. A transcriptome-based phylogenetic study of hard ticks (Ixodidae). Sci. Rep. 2019, 9, 12923. [Google Scholar] [CrossRef] [PubMed]
  16. Gilbert, L. The Impacts of Climate Change on Ticks and Tick-Borne Disease Risk. Annu. Rev. Entomol. 2021, 66, 373–388. [Google Scholar] [CrossRef]
  17. Telmadarraiy, Z.; Kooshki, H.; Edalat, H.; Vatandoost, H.; Bakhshi, H.; Faghihi, F.; Hosseini-Chegeni, A.; Oshaghi, M.A. Study on Hard and Soft Ticks of Domestic and Wild Animals in Western Iran. J. Arthropod-Borne Dis. 2023, 16, 225–232. [Google Scholar] [CrossRef]
  18. Paddock, C.D.; Lane, R.S.; Staples, J.E.; Labruna, M.B. Changing Paradigms for Tick-Borne Diseases in the Americas; National Academies Press (US): Washington, DC, USA, 2016. Available online: www.ncbi.nlm.nih.gov (accessed on 12 December 2023).
  19. Diarra, A.Z.; Kelly, P.; Davoust, B.; Parola, P. Tick-Borne Diseases of Humans and Animals in West Africa. Pathogens 2023, 12, 1276. [Google Scholar] [CrossRef]
  20. de la Fuente, J.; Estrada-Pena, A.; Venzal, J.M.; Kocan, K.M.; Sonenshine, D.E. Overview: Ticks as vectors of pathogens that cause disease in humans and animals. Front. Biosci. 2008, 13, 6938–6946. [Google Scholar] [CrossRef]
  21. Franta, Z.; Frantová, H.; Konvičková, J.; Horn, M.; Sojka, D.; Mareš, M.; Kopáček, P. Dynamics of digestive proteolytic system during blood feeding of the hard tick Ixodes ricinus. Parasites Vectors 2010, 3, 119. [Google Scholar] [CrossRef]
  22. Starck, J.M.; Mehnert, L.; Biging, A.; Bjarsch, J.; Franz-Guess, S.; Kleeberger, D.; Hörnig, M. Morphological responses to feeding in ticks (Ixodes ricinus). Zool. Lett. 2018, 4, 20. [Google Scholar] [CrossRef]
  23. Francischetti, I.M.B.; Sa-Nunes, A.; Mans, B.J.; Santos, I.M.; Ribeiro, J.M.C. The role of saliva in tick feeding. Front. Biosci. 2009, 14, 2051–2088. [Google Scholar] [CrossRef] [PubMed]
  24. Šimo, L.; Kazimirova, M.; Richardson, J.; Bonnet, S.I. The Essential Role of Tick Salivary Glands and Saliva in Tick Feeding and Pathogen Transmission. Front. Cell. Infect. Microbiol. 2017, 7, 281. [Google Scholar] [CrossRef]
  25. Bouchard, C.; Dibernardo, A.; Koffi, J.; Wood, H.; Leighton, P.A.; Lindsay, L.R. Increased risk of tick-borne diseases with climate and environmental changes. Can. Commun. Dis. Rep. 2019, 45, 83–89. [Google Scholar] [CrossRef]
  26. Hromníková, D.; Furka, D.; Furka, S.; Santana, J.A.D.; Ravingerová, T.; Klöcklerová, V.; Žitňan, D. Prevention of tick-borne diseases: Challenge to recent medicine. Biologia 2022, 77, 1533–1554. [Google Scholar] [CrossRef] [PubMed]
  27. Nuttall, P.A. Climate change impacts on ticks and tick-borne infections. Biologia 2021, 77, 1503–1512. [Google Scholar] [CrossRef]
  28. Stafford, I.K.C.; Williams, S.C.; Molaei, G. Integrated Pest Management in Controlling Ticks and Tick-Associated Diseases. J. Integr. Pest Manag. 2017, 8, 28. [Google Scholar] [CrossRef]
  29. Eisen, L.; Stafford, K.C. Barriers to Effective Tick Management and Tick-Bite Prevention in the United States (Acari: Ixodidae). J. Med. Entomol. 2021, 58, 1588–1600. [Google Scholar] [CrossRef] [PubMed]
  30. Rochlin, I.; Toledo, A. Emerging tick-borne pathogens of public health importance: A mini-review. J. Med. Microbiol. 2020, 69, 781–791. [Google Scholar] [CrossRef] [PubMed]
  31. Zhao, G.-P.; Wang, Y.-X.; Fan, Z.-W.; Ji, Y.; Liu, M.-J.; Zhang, W.-H.; Li, X.-L.; Zhou, S.-X.; Li, H.; Liang, S.; et al. Mapping ticks and tick-borne pathogens in China. Nat. Commun. 2021, 12, 1075. [Google Scholar] [CrossRef] [PubMed]
  32. Bai, Y.; Li, Y.; Liu, W.; Li, J.; Tian, F.; Liu, L.; Han, X.; Tong, Y. Analysis of the diversity of tick-borne viruses at the border areas in Liaoning Province, China. Front. Microbiol. 2023, 14, 1179156. [Google Scholar] [CrossRef] [PubMed]
  33. de la Fuente, J.; Estrada-Peña, A.; Rafael, M.; Almazán, C.; Bermúdez, S.; Abdelbaset, A.E.; Kasaija, P.D.; Kabi, F.; Akande, F.A.; Ajagbe, D.O.; et al. Perception of Ticks and Tick-Borne Diseases Worldwide. Pathogens 2023, 12, 1258. [Google Scholar] [CrossRef] [PubMed]
  34. Chao, L.-L.; Shih, C.-M. First report of human biting activity of Ixodes acutitarsus (Acari: Ixodidae) collected in Taiwan. Exp. Appl. Acarol. 2012, 56, 159–164. [Google Scholar] [CrossRef]
  35. Estrada-Peña, A.; Jongejan, F. Ticks feeding on humans: A review of records on human-biting Ixodoidea with special reference to pathogen transmission. Exp. Appl. Acarol. 1999, 23, 685–715. [Google Scholar] [CrossRef]
  36. Ajithkumar, K.G.; Ravindran, R.; Ghosh, S. Dermacentor auratus Supino, 1897 (Acarina, Ixodidae) reported from Wayanad, Kerala. Indian J. Med. Res. 2012, 135, 435–436. [Google Scholar] [PubMed]
  37. Soundararajan, C.; Nagarajan, K.; Prakash, M.A. Tick infestation in human beings in the Nilgiris and Kancheepuram district of Tamil Nadu, India. J. Parasit. Dis. 2017, 42, 50–54. [Google Scholar] [CrossRef] [PubMed]
  38. Kumar, B.; Manjunathachar, H.V.; Ghosh, S. A review on Hyalomma species infestations on human and animals and progress on management strategies. Heliyon 2020, 6, e05675. [Google Scholar] [CrossRef] [PubMed]
  39. Stephen, S.; Sangeetha, B.; Antony, P.X. Seroprevalence of coxiellosis (Q fever) in sheep & goat in Puducherry & neighbouring Tamil Nadu. Indian J. Med. Res. 2014, 140, 785–787. [Google Scholar] [PubMed]
  40. Jairath, V.; Sehrawat, M.; Jindal, N.; Jain, V.K.; Aggarwal, P. Lyme disease in Haryana, India. Indian J. Dermatol. Venereol. Leprol. 2014, 80, 320–323. [Google Scholar] [CrossRef]
  41. Negi, T.; Kandari, L.S.; Arunachalam, K. Update on prevalence and distribution pattern of tick-borne diseases among humans in India: A review. Parasitol. Res. 2021, 120, 1523–1539. [Google Scholar] [CrossRef]
  42. Mourya, D.T.; Yadav, P.D.; Patil, D.Y.; Sahay, R.R.; Rahi, M. Experiences of Indian Council of Medical Research with tick-borne zoonotic infections: Kyasanur Forest disease & Crimean-Congo haemorrhagic fever in India with One Health focus. Indian J. Med. Res. 2021, 153, 339–347. [Google Scholar] [CrossRef] [PubMed]
  43. Gurav, Y.K.; Yadav, P.D.; Gokhale, M.D.; Chiplunkar, T.R.; Vishwanathan, R.; Patil, D.Y.; Jain, R.; Shete, A.M.; Patil, S.L.; Sarang, G.; et al. Kyasanur Forest Disease Prevalence in Western Ghats Proven and Confirmed by Recent Outbreak in Maharashtra, India, 2016. Vector-Borne Zoonotic Dis. 2018, 18, 164–172. [Google Scholar] [CrossRef]
  44. Munivenkatappa, A.; Sahay, R.R.; Yadav, P.D.; Viswanathan, R.; Mourya, D.T. Clinical & epidemiological significance of Kyasanur forest disease. Indian J. Med. Res. 2018, 148, 145–150. [Google Scholar] [CrossRef]
  45. Chakraborty, S.; Andrade, F.C.D.; Ghosh, S.; Uelmen, J.; Ruiz, M.O. Historical Expansion of Kyasanur Forest Disease in India from 1957 to 2017: A Retrospective Analysis. GeoHealth 2019, 3, 44–55. [Google Scholar] [CrossRef]
  46. Pattnaik, S.; Agrawal, R.; Murmu, J.; Kanungo, S.; Pati, S. Does the rise in cases of Kyasanur forest disease call for the implementation of One Health in India? IJID Reg. 2023, 7, 18–21. [Google Scholar] [CrossRef] [PubMed]
  47. Yadav, P.D.; Raut, C.G.; Patil, D.Y.; Majumdar, T.D.; Mourya, D.T. Crimean-Congo Hemorrhagic Fever: Current Scenario in India. Proc. Natl. Acad. Sci. India Sect. B Biol. Sci. 2014, 84, 9–18. [Google Scholar] [CrossRef] [PubMed]
  48. Mourya, D.T.; Yadav, P.D.; Shete, A.M.; Sathe, P.S.; Sarkale, P.C.; Pattnaik, B.; Sharma, G.; Upadhyay, K.J.; Gosavi, S.; Patil, D.Y.; et al. Cross-sectional Serosurvey of Crimean-Congo Hemorrhagic Fever Virus IgG in Livestock, India, 2013–2014. Emerg. Infect. Dis. 2015, 21, 1837–1839. [Google Scholar] [CrossRef] [PubMed]
  49. Mourya, D.T.; Makwana, D.; Yadav, P.D.; Kelaiya, A. First confirmed case of Crimean-Congo haemorrhagic fever from Sirohi district in Rajasthan State, India. Indian J. Med. Res. 2015, 142, 489–491. [Google Scholar] [CrossRef] [PubMed]
  50. Yadav, P.D.; Patil, D.Y.; Shete, A.M.; Kokate, P.; Goyal, P.; Jadhav, S.; Sinha, S.; Zawar, D.; Sharma, S.K.; Kapil, A.; et al. Nosocomial infection of CCHF among health care workers in Rajasthan, India. BMC Infect. Dis. 2016, 16, 624. [Google Scholar] [CrossRef]
  51. Larcombe, S.D.; Kolte, S.W.; Ponnudurai, G.; Kurkure, N.; Magar, S.; Velusamy, R.; Rani, N.; Rubinibala, B.; Rekha, B.; Alagesan, A.; et al. The impact of tick-borne pathogen infection in Indian bovines is determined by host type but not the genotype of Theileria annulata. Infect. Genet. Evol. 2019, 75, 103972. [Google Scholar] [CrossRef] [PubMed]
  52. Rajput, Z.I.; Hu, S.-H.; Chen, W.-J.; Arijo, A.G.; Xiao, C.-W. Importance of ticks and their chemical and immunological control in livestock. J. Zhejiang Univ. B 2006, 7, 912–921. [Google Scholar] [CrossRef]
  53. Bhowmick, B.; Han, Q. Understanding Tick Biology and Its Implications in Anti-tick and Transmission Blocking Vaccines against Tick-Borne Pathogens. Front. Vet. Sci. 2020, 7, 319. [Google Scholar] [CrossRef]
  54. Hofmeester, T.R.; Rowcliffe, J.M.; Jansen, P.A. Quantifying the Availability of Vertebrate Hosts to Ticks: A Camera-Trapping Approach. Front. Vet. Sci. 2017, 4, 115. [Google Scholar] [CrossRef]
  55. De La Fuente, J.; Antunes, S.; Bonnet, S.; Cabezas-Cruz, A.; Domingos, A.G.; Estrada-Peña, A.; Johnson, N.; Kocan, K.M.; Mansfield, K.L.; Nijhof, A.M.; et al. Tick-Pathogen Interactions and Vector Competence: Identification of Molecular Drivers for Tick-Borne Diseases. Front. Cell. Infect. Microbiol. 2017, 7, 114. [Google Scholar] [CrossRef]
  56. Gomez-Chamorro, A.; Hodžić, A.; King, K.C.; Cabezas-Cruz, A. Ecological and evolutionary perspectives on tick-borne pathogen co-infections. Curr. Res. Parasitol. Vector-Borne Dis. 2021, 1, 100049. [Google Scholar] [CrossRef]
  57. Hoffman, T.; Olsen, B.; Lundkvist, Å. The Biological and Ecological Features of Northbound Migratory Birds, Ticks, and Tick-Borne Microorganisms in the African–Western Palearctic. Microorganisms 2023, 11, 158. [Google Scholar] [CrossRef] [PubMed]
  58. Voyiatzaki, C.; Papailia, S.I.; Venetikou, M.S.; Pouris, J.; Tsoumani, M.E.; Papageorgiou, E.G. Climate Changes Exacerbate the Spread of Ixodes ricinus and the Occurrence of Lyme Borreliosis and Tick-Borne Encephalitis in Europe—How Climate Models Are Used as a Risk Assessment Approach for Tick-Borne Diseases. Int. J. Environ. Res. Public Health 2022, 19, 6516. [Google Scholar] [CrossRef] [PubMed]
  59. Nava, S.; Guglielmone, A.A.; Mangold, A.J. An overview of systematics and evolution of ticks. Front. Biosci. 2009, 14, 2857–2877. [Google Scholar] [CrossRef] [PubMed]
  60. Leung, T.L.F. Fossils of parasites: What can the fossil record tell us about the evolution of parasitism? Biol. Rev. 2017, 92, 410–430. [Google Scholar] [CrossRef] [PubMed]
  61. Parola, P.; Fenollar, F.; Badiaga, S.; Brouqui, P.; Raoult, D. First Documentation of Rickettsia conorii Infection (Strain Indian Tick Typhus) in a Traveler. Emerg. Infect. Dis. 2001, 7, 909–910. [Google Scholar] [CrossRef] [PubMed]
  62. Latif, A.A.; Putterill, J.F.; de Klerk, D.G.; Pienaar, R.; Mans, B.J. Nuttalliella namaqua (Ixodoidea: Nuttalliellidae): First Description of the Male, Immature Stages and Re-Description of the Female. PLoS ONE 2012, 7, e41651. [Google Scholar] [CrossRef] [PubMed]
  63. Mans, B.J.; de Klerk, D.; Pienaar, R.; de Castro, M.H.; Latif, A.A. The Mitochondrial Genomes of Nuttalliella namaqua (Ixodoidea: Nuttalliellidae) and Argas africolumbae (Ixodoidae: Argasidae): Estimation of Divergence Dates for the Major Tick Lineages and Reconstruction of Ancestral Blood-Feeding Characters. PLoS ONE 2012, 7, e49461. [Google Scholar] [CrossRef] [PubMed]
  64. Chhillar, S.; Chhilar, J.S.; Kaur, H. Investigations on Some Hard Ticks (Acari: Ixodidae) Infesting Domestic Buffalo and Cattle from Haryana, India. J. Entomol. Zool. Stud. 2014, 2, 99–104. [Google Scholar]
  65. Ranganathan, K.; Renu, G.; Ayyanar, E.; Veeramanoharan, R.; Paulraj, P.S. Species composition of hard ticks (Acari: Ixodidae) on domestic animals and their public health importance in Tamil Nadu, South India. Acarol. Stud. 2021, 3, 16–21. [Google Scholar] [CrossRef]
  66. Wikimedia Commons Ornithodoros Adult Soft-Tick.jpg. 2020. Available online: https://commons.wikimedia.org/w/index.php?title=File:Ornithodoros_adult_soft-tick.jpg&oldid=493571749 (accessed on 17 December 2023).
  67. Oliver, J.H. Biology and Systematics of Ticks (Acari:Ixodida). Annu. Rev. Ecol. Syst. 1989, 20, 397–430. [Google Scholar] [CrossRef]
  68. Couvreur, B.; Beaufays, J.; Charon, C.; Lahaye, K.; Gensale, F.; Denis, V.; Charloteaux, B.; Decrem, Y.; Prévôt, P.-P.; Brossard, M.; et al. Variability and Action Mechanism of a Family of Anticomplement Proteins in Ixodes ricinus. PLoS ONE 2008, 3, e1400. [Google Scholar] [CrossRef] [PubMed]
  69. Vargas-Sandoval, M.; Priego-Santander, A.G.; Larrazábal, A.; Sosa-Gutiérrez, C.G.; Lara-Chávez, B.; Avila-Val, T. Potential Species Distribution and Richness of Ixodidae Ticks Associated with Wild Vertebrates from Michoacán, Mexico. J. Geogr. Inf. Syst. 2014, 6, 467–477. [Google Scholar] [CrossRef]
  70. Nicholson, W.L.; Sonenshine, D.E.; Noden, B.H.; Brown, R.N. Ticks (Ixodida). Med. Vet. Entomol. 2019, 2019, 603–672. [Google Scholar] [CrossRef]
  71. Pospelova-Shtrom, M.V. On the system of classification of ticks of the family Argasidae. Acarologia 1969, 11, 1–22. [Google Scholar] [PubMed]
  72. Sándor, A.D.; Mihalca, A.D.; Domşa, C.; Péter, Á.; Hornok, S. Argasid Ticks of Palearctic Bats: Distribution, Host Selection, and Zoonotic Importance. Front. Vet. Sci. 2021, 8, 684737. [Google Scholar] [CrossRef] [PubMed]
  73. Chen, Z.; Liu, J. A review of argasid ticks and associated pathogens of China. Front. Vet. Sci. 2022, 9, 865664. [Google Scholar] [CrossRef] [PubMed]
  74. Estrada-Pena, A.; Mangold, A.; Nava, S.; Venzal, J.; Labruna, M.; Guglielmone, A. A review of the systematics of the tick family Argasidae (Ixodida). Acarologia 2010, 50, 317–333. [Google Scholar] [CrossRef]
  75. Lafri, I.; Benredjem, W.; Neffah-Baaziz, F.; Lalout, R.; Abdelouahed, K.; Gassen, B.; Bakhouch, S.; Chergui, M.; Karakellah, M.; Adjmi-Hamoudi, H.; et al. Inventory and update on argasid ticks and associated pathogens in Algeria. New Microbes New Infect. 2018, 23, 110–114. [Google Scholar] [CrossRef]
  76. Trevisan, G.; Cinco, M.; Trevisini, S.; di Meo, N.; Ruscio, M.; Forgione, P.; Bonin, S. Borreliae Part 2: Borrelia Relapsing Fever Group and Unclassified Borrelia. Biology 2021, 10, 1117. [Google Scholar] [CrossRef]
  77. Keirans, J.E.; Clifford, C.M.; Hoogstraal, H.; Easton, E.R. Discovery of Nuttalliella namaqua Bedford (Acarina: Ixodoidea: Nuttalliellidae) in Tanzania and Redescription of the Female Based on Scanning Electron Microcopy. Ann. Entomol. Soc. Am. 1976, 69, 926–932. [Google Scholar] [CrossRef]
  78. Roshdy, M.A.; Hoogstraal, H.; Banaja, A.A.; Shoura, S.M. Nuttalliella namaqua (Ixodoidea: Nuttalliellidae): Spiracle structure and surface morphology. Z. Parasitenkd. 1983, 69, 817–821. [Google Scholar] [CrossRef]
  79. Jin, K.; Koh, Y.-J.; Ahn, S.K.; Cho, J.; Lim, J.; Song, J.; Lee, J.; Gong, Y.W.; Kwon, M.J.; Kwon, H.W.; et al. Hard Ticks as Vectors Tested Negative for Severe Fever with Thrombocytopenia Syndrome in Ganghwa-do, Korea during 2019–2020. Korean J. Parasitol. 2021, 59, 281–289. [Google Scholar] [CrossRef]
  80. Arthur, D.R. The morphology of the British Prostriata, with particular reference to Ixodes hexagonus Leach. III. Parasitology 1956, 46, 261–307. [Google Scholar] [CrossRef]
  81. Feldman-Muhsam, B.; Borut, S. Copulation in Ixodid Ticks. J. Parasitol. 1971, 57, 630. [Google Scholar] [CrossRef]
  82. Anderson, J.F. The natural history of ticks. Med. Clin. N. Am. 2002, 86, 205–218. [Google Scholar] [CrossRef]
  83. Chitimia-Dobler, L.; Dunlop, J.A.; Pfeffer, T.; Würzinger, F.; Handschuh, S.; Mans, B.J. Hard ticks in Burmese amber with Australasian affinities. Parasitology 2023, 150, 157–171. [Google Scholar] [CrossRef] [PubMed]
  84. Sonenshine, D.E.; Šimo, L. Biology and molecular biology of Ixodes scapularis. In Lyme Disease and Relapsing Fever Spirochetes: Genomics, Molecular Biology, Host Interactions and Disease Pathogenesis; Radolf, J.D., Samuels, D.S., Eds.; Caister Academic Press: Poole, UK, 2021; pp. 339–366. [Google Scholar] [CrossRef]
  85. Yamaguti, N.; Tipton, V.J.; Keegan, H.L.; Toshioka, S. Ticks of Japan, Korea, and the Ryukyu Islands. Brigh. Young Univ. Sci. Bull. Biol. Ser. 1971, 15, 1. [Google Scholar]
  86. Bowman, A.S.; Sauer, J.R. Tick salivary glands: Function, physiology and future. Parasitology 2005, 129, S67–S81. [Google Scholar] [CrossRef]
  87. Szlendak, E.; Oliver, J.H. Anatomy of synganglia, including their neurosecretory regions, in unfed, virgin female Ixodes scapularis say (Acari: Ixodidae). J. Morphol. 1992, 213, 349–364. [Google Scholar] [CrossRef]
  88. Borges, L.M.F.; Li, A.Y.; Olafson, P.U.; Renthal, R.; Bauchan, G.R.; Lohmeyer, K.H.; de León, A.A.P. Neuronal projections from the Haller’s organ and palp sensilla to the synganglion of Amblyomma americanum. Rev. Bras. Parasitol. Veterinária 2016, 25, 217–224. [Google Scholar] [CrossRef] [PubMed]
  89. Fielden, L.J.; Duncan, F.D.; Rechav, Y.; Crewe, R.M. Respiratory Gas Exchange in the Tick Amblyomma hebraeum (Acari: Ixodidae). J. Med. Entomol. 1994, 31, 30–35. [Google Scholar] [CrossRef] [PubMed]
  90. Sonenshine, D.E.; Roe, R.M. Biology of Ticks; Oxford University Press: New York, NY, USA, 2014. [Google Scholar]
  91. Feitosa, A.P.S.; Alves, L.C.; Chaves, M.M.; Veras, D.L.; Silva, E.M.; Aliança, A.S.S.; França, I.R.S.; Gonçalves, G.G.A.; Lima-Filho, J.L.; Brayner, F.A. Hemocytes of Rhipicephalus sanguineus (Acari: Ixodidae): Characterization, Population Abundance, and Ultrastructural Changes Following Challenge with Leishmania infantum. J. Med. Entomol. 2015, 52, 1193–1202. [Google Scholar] [CrossRef]
  92. Patton, T.G.; Dietrich, G.; Brandt, K.; Dolan, M.C.; Piesman, J.; Gilmore, R.D., Jr. Saliva, Salivary Gland, and Hemolymph Collection from Ixodes scapularis Ticks. J. Vis. Exp. JoVE 2012, 60, e3894. [Google Scholar] [CrossRef] [PubMed]
  93. Kubiak, K.; Dziekońska-Rynko, J. Seasonal activity of the common European tick, Ixodes ricinus (Linnaeus, 1758), in the forested areas of the city of Olsztyn and its surroundings. Wiad Parazytol. 2006, 52, 59–64. [Google Scholar]
  94. McCoy, K.D.; Léger, E.; Dietrich, M. Host specialization in ticks and transmission of tick-borne diseases: A review. Front. Cell. Infect. Microbiol. 2013, 3, 57. [Google Scholar] [CrossRef] [PubMed]
  95. Maxime Madder, M.; Horak, I.; Stoltsz, H. Ticks: Tick Identification; AfriVIP: Pretoria, South Africa, 2018. [Google Scholar]
  96. Gray, J.S.; Dautel, H.; Estrada-Peña, A.; Kahl, O.; Lindgren, E. Effects of climate change on ticks and tick-borne diseases in Europe. Interdiscip. Perspect. Infect. Dis. 2009, 2009, 593232. [Google Scholar] [CrossRef] [PubMed]
  97. Sauer, J.R.; McSwain, J.L.; Bowman, A.S.; Essenberg, R.C. Tick Salivary Gland Physiology. Annu. Rev. Entomol. 1995, 40, 245–267. [Google Scholar] [CrossRef]
  98. Estrada-Peña, A.; Gray, J.S.; Kahl, O.; Lane, R.S.; Nijhof, A.M. Research on the ecology of ticks and tick-borne pathogens—Methodological principles and caveats. Front. Cell. Infect. Microbiol. 2013, 3, 29. [Google Scholar] [CrossRef]
  99. Maldonado-Ruiz, L.P.; Park, Y.; Zurek, L. Liquid water intake of the lone star tick, Amblyomma americanum: Implications for tick survival and management. Sci. Rep. 2020, 10, 6000. [Google Scholar] [CrossRef]
  100. Yoder, J.A.; Benoit, J.B.; Rellinger, E.J.; Tank, J.L. Developmental profiles in tick water balance with a focus on the new Rocky Mountain spotted fever vector, Rhipicephalus sanguineus. Med. Vet. Entomol. 2006, 20, 365–372. [Google Scholar] [CrossRef] [PubMed]
  101. Knülle, W.; Rudolph, D. Humidity Relationships and Water Balance of Ticks. In Physiology of Ticks; Pergamon: Oxford, UK, 1982; pp. 43–70. [Google Scholar] [CrossRef]
  102. Rahlenbeck, S.; Fingerle, V.; Doggett, S. Prevention of tick-borne diseases: An overview. Br. J. Gen. Pract. 2016, 66, 492–494. [Google Scholar] [CrossRef] [PubMed]
  103. Nejash, A. Review of Important Cattle Tick and Its Control in Ethiopia. Open Access Libr. J. 2016, 3, 69073. [Google Scholar] [CrossRef]
  104. Couper, L.I.; Yang, Y.; Yang, X.F.; Swei, A. Comparative vector competence of North American Lyme disease vectors. Parasites Vectors 2020, 13, 29. [Google Scholar] [CrossRef] [PubMed]
  105. Sharma, R.; Cozens, D.W.; Armstrong, P.M.; Brackney, D.E. Vector competence of human-biting ticks Ixodes scapularis, Amblyomma americanum and Dermacentor variabilis for Powassan virus. Parasites Vectors 2021, 14, 466. [Google Scholar] [CrossRef] [PubMed]
  106. Ochanda, H.; Young, A.S.; Medley, G.F.; Perry, B.D. Vector competence of 7 Rhipicephalid tick stocks in transmitting 2 Theileria parva parasite stocks from Kenya and Zimbabwe. Parasitology 1998, 116, 539–545. [Google Scholar] [CrossRef] [PubMed]
  107. Migné, C.V.; de Seixas, H.B.; Heckmann, A.; Galon, C.; Jaafar, F.M.; Monsion, B.; Attoui, H.; Moutailler, S. Evaluation of Vector Competence of Ixodes Ticks for Kemerovo Virus. Viruses 2022, 14, 1102. [Google Scholar] [CrossRef] [PubMed]
  108. Eisen, L. Vector competence studies with hard ticks and Borrelia burgdorferi sensu lato spirochetes: A review. Ticks Tick-Borne Dis. 2020, 11, 101359. [Google Scholar] [CrossRef] [PubMed]
  109. Madison-Antenucci, S.; Kramer, L.D.; Gebhardt, L.L.; Kauffman, E. Emerging Tick-Borne Diseases. Clin. Microbiol. Rev. 2020, 33, e00083-18. [Google Scholar] [CrossRef]
  110. Cutler, S.J.; Vayssier-Taussat, M.; Estrada-Peña, A.; Potkonjak, A.; Mihalca, A.D.; Zeller, H. Tick-borne diseases and co-infection: Current considerations. Ticks Tick-Borne Dis. 2021, 12, 101607. [Google Scholar] [CrossRef]
  111. Maqbool, M.; Sajid, M.S.; Saqib, M.; Anjum, F.R.; Tayyab, M.H.; Rizwan, H.M.; Rashid, M.I.; Rashid, I.; Iqbal, A.; Siddique, R.M.; et al. Potential Mechanisms of Transmission of Tick-Borne Viruses at the Virus-Tick Interface. Front. Microbiol. 2022, 13, 846884. [Google Scholar] [CrossRef] [PubMed]
  112. Richter, D.; Allgöwer, R.; Matuschka, F.-R. Co-feeding Transmission and Its Contribution to the Perpetuation of the Lyme Disease Spirochete Borrelia afzelii. Emerg. Infect. Dis. 2002, 8, 1421–1425. [Google Scholar] [CrossRef] [PubMed]
  113. Nah, K.; Magpantay, F.M.G.; Bede-Fazekas, Á.; Röst, G.; Trájer, A.J.; Wu, X.; Zhang, X.; Wu, J. Assessing systemic and non-systemic transmission risk of tick-borne encephalitis virus in Hungary. PLoS ONE 2019, 14, e0217206. [Google Scholar] [CrossRef] [PubMed]
  114. Wu, X.; Gao, D.; Song, Z.; Wu, J. Modelling triatomine bug population and Trypanosoma rangeli transmission dynamics: Co-feeding, pathogenic effect and linkage with chagas disease. Math. Biosci. 2020, 324, 108326. [Google Scholar] [CrossRef]
  115. Voordouw, M.J. Co-feeding transmission in Lyme disease pathogens. Parasitology 2015, 142, 290–302. [Google Scholar] [CrossRef] [PubMed]
  116. González, J.; González, M.G.; Valcárcel, F.; Sánchez, M.; Martín-Hernández, R.; Tercero, J.M.; Olmeda, A.S. Transstadial Transmission from Nymph to Adult of Coxiella burnetii by Naturally Infected Hyalomma lusitanicum. Pathogens 2020, 9, 884. [Google Scholar] [CrossRef] [PubMed]
  117. Hauck, D.; Jordan, D.; Springer, A.; Schunack, B.; Pachnicke, S.; Fingerle, V.; Strube, C. Transovarial transmission of Borrelia spp., Rickettsia spp. And Anaplasma phagocytophilum in Ixodes ricinus under field conditions extrapolated from DNA detection in questing larvae. Parasites Vectors 2020, 13, 176. [Google Scholar] [CrossRef] [PubMed]
  118. Dutra, D.d.A.; Poulin, R.; Ferreira, F.C. Evolutionary consequences of vector-borne transmission: How using vectors shapes host, vector and pathogen evolution. Parasitology 2022, 149, 1667–1678. [Google Scholar] [CrossRef] [PubMed]
  119. Beard, D.; Stannard, H.J.; Old, J.M. Morphological identification of ticks and molecular detection of tick-borne pathogens from bare-nosed wombats (Vombatus ursinus). Parasites Vectors 2021, 14, 60. [Google Scholar] [CrossRef]
  120. Bonnet, S.I.; Binetruy, F.; Hernández-Jarguín, A.M.; Duron, O. The Tick Microbiome: Why Non-pathogenic Microorganisms Matter in Tick Biology and Pathogen Transmission. Front. Cell. Infect. Microbiol. 2017, 7, 236. [Google Scholar] [CrossRef]
  121. Vancová, M.; Bílý, T.; Šimo, L.; Touš, J.; Horodyský, P.; Růžek, D.; Novobilský, A.; Salát, J.; Strnad, M.; Sonenshine, D.E.; et al. Three-dimensional reconstruction of the feeding apparatus of the tick Ixodes ricinus (Acari: Ixodidae): A new insight into the mechanism of blood-feeding. Sci. Rep. 2020, 10, 165. [Google Scholar] [CrossRef]
  122. Shepherd, J.G. Mating, Sperm Transfer and Oviposition in Soft Ticks (Acari: Argasidae), a Review. Pathogens 2023, 12, 582. [Google Scholar] [CrossRef] [PubMed]
  123. Carter, C.; Yambem, O.; Carlson, T.; Hickling, G.J.; Collins, K.; Jacewicz, M.; Tsao, J.W. Male tick bite: A rare cause of adult tick paralysis. Neurol. Neuroimmunol. Neuroinflamm. 2016, 3, e243. [Google Scholar] [CrossRef] [PubMed]
  124. Tan, A.W.; Francischetti, I.M.; Slovak, M.; Kini, R.M.; Ribeiro, J.M. Sexual differences in the sialomes of the zebra tick, Rhipicephalus pulchellus. J. Proteom. 2015, 117, 120–144. [Google Scholar] [CrossRef] [PubMed]
  125. Lu, S.; Martins, L.A.; Kotál, J.; Ribeiro, J.M.C.; Tirloni, L. A longitudinal transcriptomic analysis from unfed to post-engorgement midguts of adult female Ixodes scapularis. Sci. Rep. 2023, 13, 11360. [Google Scholar] [CrossRef]
  126. Bartíková, P.; Kazimírová, M.; Štibrániová, I. Ticks and the effects of their saliva on growth factors involved in skin wound healing. J. Venom Res. 2020, 10, 45–52. [Google Scholar]
  127. Saleh, M.N.; Allen, K.E.; Lineberry, M.W.; Little, S.E.; Reichard, M.V. Ticks infesting dogs and cats in North America: Biology, geographic distribution, and pathogen transmission. Vet. Parasitol. 2021, 294, 109392. [Google Scholar] [CrossRef]
  128. Suppan, J.; Engel, B.; Marchetti-Deschmann, M.; Nürnberger, S. Tick attachment cement—Reviewing the mysteries of a biological skin plug system. Biol. Rev. 2018, 93, 1056–1076. [Google Scholar] [CrossRef]
  129. Villar, M.; Pacheco, I.; Merino, O.; Contreras, M.; Mateos-Hernández, L.; Prado, E.; Barros-Picanço, D.K.; Lima-Barbero, J.F.; Artigas-Jerónimo, S.; Alberdi, P.; et al. Tick and Host Derived Compounds Detected in the Cement Complex Substance. Biomolecules 2020, 10, 555. [Google Scholar] [CrossRef]
  130. Waladde, S.; Young, A.; Morzaria, S. Artificial feeding of ixodid ticks. Parasitol. Today 1996, 12, 272–278. [Google Scholar] [CrossRef]
  131. Nava, S.; Venzal, J.; González-Acuña, D.; Martins, T.F.; Guglielmone, A.A. Ticks of the Southern Cone of America: Diagnosis, Distribution, and Hosts with Taxonomy, Ecology and Sanitary Importance; Academic Press: Cambridge, MA, USA, 2017. [Google Scholar]
  132. Kitsou, C.; Foor, S.D.; Dutta, S.; Bista, S.; Pal, U. Tick gut barriers impacting tick–microbe interactions and pathogen persistence. Mol. Microbiol. 2021, 116, 1241–1248. [Google Scholar] [CrossRef]
  133. Kitsou, C.; Fikrig, E.; Pal, U. Tick host immunity: Vector immunomodulation and acquired tick resistance. Trends Immunol. 2021, 42, 554–574. [Google Scholar] [CrossRef] [PubMed]
  134. Mateos-Hernandéz, L.; Defaye, B.; Vancová, M.; Hajdusek, O.; Sima, R.; Park, Y.; Attoui, H.; Šimo, L. Cholinergic axons regulate type I acini in salivary glands of Ixodes ricinus and Ixodes scapularis ticks. Sci. Rep. 2020, 10, 16054. [Google Scholar] [CrossRef] [PubMed]
  135. Neelakanta, G.; Sultana, H. Tick Saliva and Salivary Glands: What Do We Know So Far on Their Role in Arthropod Blood Feeding and Pathogen Transmission. Front. Cell. Infect. Microbiol. 2022, 11, 816547. [Google Scholar] [CrossRef] [PubMed]
  136. Ghannam, M.G.; Singh, P. Anatomy, Head and Neck, Salivary Glands. [Updated 2023 May 29]. In StatPearls; StatPearls Publishing: Treasure Island, FL, USA, 2023. Available online: https://www.ncbi.nlm.nih.gov/books/NBK538325/ (accessed on 20 December 2023).
  137. Kazimírová, M.; Štibrániová, I. Tick salivary compounds: Their role in modulation of host defences and pathogen transmission. Front. Cell. Infect. Microbiol. 2013, 3, 43. [Google Scholar] [CrossRef] [PubMed]
  138. Bensaoud, C.; Aounallah, H.; Sciani, J.M.; Faria, F.; Chudzinski-Tavassi, A.M.; Bouattour, A.; M’ghirbi, Y. Proteomic informed by transcriptomic for salivary glands components of the camel tick Hyalomma dromedarii. BMC Genom. 2019, 20, 675. [Google Scholar] [CrossRef] [PubMed]
  139. Ogden, N.H.; Mechai, S.; Margos, G. Changing geographic ranges of ticks and tick-borne pathogens: Drivers, mechanisms and consequences for pathogen diversity. Front. Cell. Infect. Microbiol. 2013, 3, 46. [Google Scholar] [CrossRef] [PubMed]
  140. Hawiger, J. Formation and regulation of platelet and fibrin hemostatic plug. Hum. Pathol. 1987, 18, 111–122. [Google Scholar] [CrossRef]
  141. Chmelar, J.; Calvo, E.; Pedra, J.H.; Francischetti, I.M.; Kotsyfakis, M. Tick salivary secretion as a source of antihemostatics. J. Proteom. 2012, 75, 3842–3854. [Google Scholar] [CrossRef]
  142. Narasimhan, S.; Kurokawa, C.; DeBlasio, M.; Matias, J.; Sajid, A.; Pal, U.; Lynn, G.; Fikrig, E. Acquired tick resistance: The trail is hot. Parasite Immunol. 2021, 43, e12808. [Google Scholar] [CrossRef]
  143. Fogaça, A.C.; Sousa, G.; Pavanelo, D.B.; Esteves, E.; Martins, L.A.; Urbanová, V.; Kopáček, P.; Daffre, S. Tick Immune System: What Is Known, the Interconnections, the Gaps, and the Challenges. Front. Immunol. 2021, 12, 628054. [Google Scholar] [CrossRef] [PubMed]
  144. Brossard, M.; Wikel, S.K. Tick immunobiology. Parasitology 2004, 129, S161–S176. [Google Scholar] [CrossRef]
  145. Bhusal, R.P.; Eaton, J.R.; Chowdhury, S.T.; Power, C.A.; Proudfoot, A.E.; Stone, M.J.; Bhattacharya, S. Evasins: Tick Salivary Proteins that Inhibit Mammalian Chemokines. Trends Biochem. Sci. 2020, 45, 108–122. [Google Scholar] [CrossRef] [PubMed]
  146. Bowen, C.J.; Jaworski, D.C.; Wasala, N.B.; Coons, L.B. Macrophage migration inhibitory factor expression and protein localization in Amblyomma americanum (Ixodidae). Exp. Appl. Acarol. 2010, 50, 343–352. [Google Scholar] [CrossRef] [PubMed]
  147. Aounallah, H.; Bensaoud, C.; M’ghirbi, Y.; Faria, F.; Chmelar, J.; Kotsyfakis, M. Tick Salivary Compounds for Targeted Immunomodulatory Therapy. Front. Immunol. 2020, 11, 583845. [Google Scholar] [CrossRef]
  148. Mans, B.J.; Gaspar, A.R.; Louw, A.I.; Neitz, A.W. Apyrase activity and platelet aggregation inhibitors in the tick Ornithodoros savignyi (Acari: Argasidae). Exp. Appl. Acarol. 1998, 22, 353–366. [Google Scholar] [CrossRef]
  149. Liu, L.; Dai, J.; Zhao, Y.O.; Narasimhan, S.; Yang, Y.; Zhang, L.; Fikrig, E. Ixodes scapularis JAK-STAT pathway regulates tick antimicrobial peptides, thereby controlling the agent of human granulocytic anaplasmosis. J. Infect. Dis. 2012, 206, 1233–1241. [Google Scholar] [CrossRef]
  150. Greay, T.L.; Oskam, C.L.; Gofton, A.W.; Rees, R.L.; Ryan, U.M.; Irwin, P.J. A survey of ticks (Acari: Ixodidae) of companion animals in Australia. Parasites Vectors 2016, 9, 207. [Google Scholar] [CrossRef]
  151. Dehhaghi, M.; Panahi, H.K.S.; Holmes, E.C.; Hudson, B.J.; Schloeffel, R.; Guillemin, G.J. Human Tick-Borne Diseases in Australia. Front. Cell. Infect. Microbiol. 2019, 9, 3. [Google Scholar] [CrossRef]
  152. Ledwaba, M.B.; Nozipho, K.; Tembe, D.; Onyiche, T.E.; Chaisi, M.E. Distribution and prevalence of ticks and tick-borne pathogens of wild animals in South Africa: A systematic review. Curr. Res. Parasitol. Vector-Borne Dis. 2022, 2, 100088. [Google Scholar] [CrossRef]
  153. Makwarela, T.G.; Nyangiwe, N.; Masebe, T.; Mbizeni, S.; Nesengani, L.T.; Djikeng, A.; Mapholi, N.O. Tick Diversity and Distribution of Hard (Ixodidae) Cattle Ticks in South Africa. Microbiol. Res. 2023, 14, 42–59. [Google Scholar] [CrossRef]
  154. Keve, G.; Sándor, A.D.; Hornok, S. Hard ticks (Acari: Ixodidae) associated with birds in Europe: Review of literature data. Front. Vet. Sci. 2022, 9, 928756. [Google Scholar] [CrossRef] [PubMed]
  155. Zhang, Y.; Zhang, X.; Liu, J. Ticks (Acari: Ixodoidea) in China: Geographical distribution, host diversity, and specificity. Arch. Insect Biochem. Physiol. 2019, 102, e21544. [Google Scholar] [CrossRef] [PubMed]
  156. Sharif, M. A Revision of the Indian Ixodidae with Special Reference to the Collection in the Indian Museum. Rec. Zool. Surv. India 1928, 30, 217–344. [Google Scholar] [CrossRef]
  157. Sen, P. A checklist and host list of Ixodidae (ticks) occurring in India. Indian J. Vet. Sci. Anim. Husbandary 1938, 8, 133–149. [Google Scholar]
  158. Jagannath, M.S.; Alwar, V.S.; Lalitha, C.M. Ixodid ticks of domestic stock in Tamil Nadu. Indian J. Anim. Sci. 1973, 43, 119–124. [Google Scholar]
  159. Miranpuri, G.S.; Naithani, R.C. A check list of Indian ticks (Ixodoidea: Acarina); Indian Veterinary Research Institute: Izatnagar, India, 1978; 50p. [Google Scholar]
  160. Geevarghese, G.; Fernandes, S.; Kulkarni, S.M. A checklist of Indian ticks (acari: Ixodoidea). Indian J. Anim. Sci. 1997, 67, 566–574. [Google Scholar]
  161. Gupta, S. Tick diversity in India and its impact on livestock production system. In Faunal Diversity in India; Narendra Publishing House: Delhi, India, 2015. [Google Scholar]
  162. Prakasan, K.; Ramani, N. Tick parasites of domestic animals of Kerala, South India. Asian J. Anim. Vet. Adv. 2007, 2, 74–80. [Google Scholar] [CrossRef]
  163. Shyma, K.P.; Stanley, B.; Ray, D.; Ghosh, S. Prevalence of ticks in northern region of Kerala. J. Vet. Parasitol. 2007, 27, 55–56. [Google Scholar]
  164. Nimisha, M.; Devassy, J.K.; Pradeep, R.K.; Pakideery, V.; Sruthi, M.K.; Pious, A.; Kurbet, P.S.; Amrutha, B.M.; Chandrasekhar, L.; Deepa, C.K.; et al. Ticks and accompanying pathogens of domestic and wild animals of Kerala, South India. Exp. Appl. Acarol. 2019, 79, 137–155. [Google Scholar] [CrossRef]
  165. Balasubramanian, R.; Yadav, P.D.; Sahina, S.; Nadh, V.A. Distribution and prevalence of ticks on livestock population in endemic area of Kyasanur forest disease in Western Ghats of Kerala, South India. J. Parasit. Dis. 2019, 43, 256–262. [Google Scholar] [CrossRef] [PubMed]
  166. Sadanandane, C.; Gokhale, M.D.; Elango, A.; Yadav, P.; Mourya, D.T.; Jambulingam, P. Prevalence and spatial distribution of Ixodid tick populations in the forest fringes of Western Ghats reported with human cases of Kyasanur forest disease and monkey deaths in South India. Exp. Appl. Acarol. 2018, 75, 135–142. [Google Scholar] [CrossRef] [PubMed]
  167. Vathsala, M.; Mohan, P.; Sacikumar; Ramessh, S. Survey of tick species distribution in sheep and goats in Tamil Nadu, India. Small Rumin. Res. 2008, 74, 238–242. [Google Scholar] [CrossRef]
  168. Kumar, K.; Balakrishnan, N.; Sharma, A.K. Studies on the Vertical Distribution of Ticks of Domestic Animals and Their Public Health Importance in Nilgiri Hills and Adjoining Areas of Tamil Nadu State (India). Int. J. Zool. 2014, 2014, 359812. [Google Scholar] [CrossRef]
  169. Anish, R.; Venu, R.; Rayulu, V.; Jacob, S.S.; Srilatha, C.; Surya, U.; Pradeep, B.; Prasad, T. Prevalence and diversity of ixodid tick fauna in domestic animals of Andhra Pradesh state, India. J. Entomol. Zool. Stud. 2020, 8, 2346–2351. [Google Scholar] [CrossRef]
  170. Kandi, S.; Chennuru, S.; Chitichoti, J.; Metta, M.; Krovvidi, S. Morphological and molecular characterization of ticks infesting cattle and buffaloes in different agro-climatic zones in Andhra Pradesh, India, and factors associated with high tick prevalence. Int. J. Acarol. 2022, 48, 192–200. [Google Scholar] [CrossRef]
  171. Jadhao, S.G.; Sanyal, P.K.; Borkar, S.D.; Chigure, G.M.; Jadhav, N.D.; Shirsikar, P.M.; Kumar, S. Prevalence of ixodid ticks infesting in cattle of Chhattisgarh state, an east-central part of India. Int. J. Trop. Insect Sci. 2020, 40, 951–954. [Google Scholar] [CrossRef]
  172. Dehuri, M.; Panda, M.R.; Mohanty, B.; Hembram, A.; Mahapatra, T.; Sahu, A. Ixodid ticks infesting cattle and associated risk factors in coastal districts of Odisha. J. Entomol. Zool. Stud. 2017, 5, 129–132. [Google Scholar]
  173. Thakur, P.S.; Ia, R. Genetic Analysis of Ticks from Livestock of Akola District Maharashtra India. Biosci. Biotechnol. Res. Commun. 2019, 12, 1110–1114. [Google Scholar] [CrossRef]
  174. Khan, V.; Zala, D.B.; Joshi, K.M. Occurrence of Hyalomma, (Acari: Ixodidae) Koch, 1844 on domestic animal in the Union Territory of Dadra & Nagar Haveli, Indian. J. Parasit. Dis. 2016, 40, 543–545. [Google Scholar] [CrossRef]
  175. Oza, J.; Bhatt, D.; Patel, K.; Trivedi, J. Study of Prevalence of tick Hyalomma excavatum (Acari: Ixodidae) on Bubalus bubalis in Patan District, Gujarat state, India. J. Biol. Stud. 2020, 3, 69–78. [Google Scholar] [CrossRef]
  176. Sanyal, A.K.; De, S.K. Status of Ticks (Acari: Metastigmata) of Rajasthan. Rec. Zool. Surv. India 2005, 104, 129. [Google Scholar] [CrossRef]
  177. Kumar, S.; Singh, A.; Cossio-Bayugar, R.; Moradi-Asl, E.; Singh, D.; Chaubey, A.K. Diversity and Seasonal Distribution of Hard Ticks in Livestock Animal Population from Western part of Uttar Pradesh in India. Acta Sci. Vet. Sci. 2023, 5, 73–84. [Google Scholar] [CrossRef]
  178. Araya-Anchetta, A.; Busch, J.D.; Scoles, G.A.; Wagner, D.M. Thirty years of tick population genetics: A comprehensive review. Infect. Genet. Evol. 2015, 29, 164–179. [Google Scholar] [CrossRef] [PubMed]
  179. Andreotti, R.; De León, A.A.P.; Dowd, S.E.; Guerrero, F.D.; Bendele, K.G.; Scoles, G.A. Assessment of bacterial diversity in the cattle tick Rhipicephalus (Boophilus) microplus through tag-encoded pyrosequencing. BMC Microbiol. 2011, 11, 6. [Google Scholar] [CrossRef] [PubMed]
  180. Salgotra, R.K.; Chauhan, B.S. Genetic Diversity, Conservation, and Utilization of Plant Genetic Resources. Genes 2023, 14, 174. [Google Scholar] [CrossRef] [PubMed]
  181. Dowling, D.K.; Wolff, J.N. Evolutionary genetics of the mitochondrial genome: Insights from Drosophila. Genetics 2023, 224, iyad036. [Google Scholar] [CrossRef] [PubMed]
  182. Tian, J.; Hou, X.; Ge, M.; Xu, H.; Yu, B.; Liu, J.; Shao, R.; Holmes, E.C.; Lei, C.; Shi, M. The diversity and evolutionary relationships of ticks and tick-borne bacteria collected in China. Parasites Vectors 2022, 15, 352. [Google Scholar] [CrossRef] [PubMed]
  183. Guglielmone, A.A.; Nava, S.; Mastropaolo, M.; Mangold, A.J. Distribution and genetic variation of Amblyomma triste (Acari: Ixodidae) in Argentina. Ticks Tick-Borne Dis. 2013, 4, 386–390. [Google Scholar] [CrossRef]
  184. Páez-Triana, L.; Muñoz, M.; Herrera, G.; Moreno-Pérez, D.A.; Tafur-Gómez, G.A.; Montenegro, D.; Patarroyo, M.A.; Paniz-Mondolfi, A.; Ramírez, J.D. Genetic diversity and population structure of Rhipicephalus sanguineus sensu lato across different regions of Colombia. Parasites Vectors 2021, 14, 424. [Google Scholar] [CrossRef]
  185. Sungirai, M.; Baron, S.; Van der Merwe, N.A.; Moyo, D.Z.; De Clercq, P.; Maritz-Olivier, C.; Madder, M. Population structure and genetic diversity of Rhipicephalus microplus in Zimbabwe. Acta Trop. 2018, 180, 42–46. [Google Scholar] [CrossRef]
  186. Weaver, S.C.; Forrester, N.L.; Liu, J.; Vasilakis, N. Population bottlenecks and founder effects: Implications for mosquito-borne arboviral emergence. Nat. Rev. Microbiol. 2021, 19, 184–195. [Google Scholar] [CrossRef]
  187. Lampo, M.; Rangel, Y.; Mata, A. Population genetic structure of a three-host tick, Amblyomma dissimile, in eastern Venezuela. J. Parasitol. 1998, 84, 1137. [Google Scholar] [CrossRef]
  188. Regilme, M.A.F.; Sato, M.; Tamura, T.; Arai, R.; Sato, M.O.; Ikeda, S.; Gamboa, M.; Monaghan, M.T.; Watanabe, K. Comparative population genetic structure of two ixodid tick species (Acari:Ixodidae) (Ixodes ovatus and Haemaphysalis flava) in Niigata prefecture, Japan. Infect. Genet. Evol. 2021, 94, 104999. [Google Scholar] [CrossRef]
  189. Wei, S.; Zhang, Q.; Tang, S.; Liao, W. Genetic and ecophysiological evidence that hybridization facilitated lineage diversification in yellow Camellia (Theaceae) species: A case study of natural hybridization between C. micrantha and C. flavida. BMC Plant Biol. 2023, 23, 154. [Google Scholar] [CrossRef]
  190. Kovalev, S.; Golovljova, I.; Mukhacheva, T. Natural hybridization between Ixodes ricinus and Ixodes persulcatus ticks evidenced by molecular genetics methods. Ticks Tick-Borne Dis. 2016, 7, 113–118. [Google Scholar] [CrossRef]
  191. Kovalev, S.Y.; Mikhaylishcheva, M.S.; Mukhacheva, T.A. Natural hybridization of the ticks Ixodes persulcatus and Ixodes pavlovskyi in their sympatric populations in Western Siberia. Infect. Genet. Evol. 2015, 32, 388–395. [Google Scholar] [CrossRef]
  192. Belova, O.A.; Polienko, A.E.; Averianova, A.D.; Karganova, G.G. Hybrids of Ixodes ricinus and Ixodes persulcatus ticks effectively acquire and transmit tick-borne encephalitis virus. Front. Cell. Infect. Microbiol. 2023, 13, 1104484. [Google Scholar] [CrossRef]
  193. Kaba, T. Geographical distribution of ixodid ticks and tick-borne pathogens of domestic animals in Ethiopia: A systematic review. Parasites Vectors 2022, 15, 108. [Google Scholar] [CrossRef]
  194. Brennan, R.N.; Boychuck, S.; Washkwich, A.J.; John-Alder, H.; Fonseca, D.M. Tick abundance and diversity are substantially lower in thinned vs. unthinned forests in the New Jersey Pinelands National Reserve, USA. Ticks Tick-Borne Dis. 2023, 14, 102106. [Google Scholar] [CrossRef] [PubMed]
  195. Babayani, N.D.; Makati, A. Predictive Analytics of Cattle Host and Environmental and Micro-Climate Factors for Tick Distribution and Abundance at the Livestock–Wildlife Interface in the Lower Okavango Delta of Botswana. Front. Vet. Sci. 2021, 8, 698395. [Google Scholar] [CrossRef]
  196. Paul, R.E.L.; Cote, M.; Le Naour, E.; Bonnet, S.I. Environmental factors influencing tick densities over seven years in a French suburban forest. Parasites Vectors 2016, 9, 309. [Google Scholar] [CrossRef]
  197. Dantas-Torres, F. Climate change, biodiversity, ticks and tick-borne diseases: The butterfly effect. Int. J. Parasitol. Parasites Wildl. 2015, 4, 452–461. [Google Scholar] [CrossRef]
  198. Institute of Medicine (US) Committee on Lyme Disease and Other Tick-Borne Diseases: The State of the Science. 4, Emerging Infections, Tick Biology, and Host–Vector Interactions. In Critical Needs and Gaps in Understanding Prevention, Amelioration, and Resolution of Lyme and Other Tick-Borne Diseases: The Short-Term and Long-Term Outcomes: Workshop Report; National Academies Press (US): Washington, DC, USA, 2011. Available online: https://www.ncbi.nlm.nih.gov/books/NBK57022/ (accessed on 23 November 2023).
  199. O’neill, X.; White, A.; Gortázar, C.; Ruiz-Fons, F. The Impact of Host Abundance on the Epidemiology of Tick-Borne Infection. Bull. Math. Biol. 2023, 85, 30. [Google Scholar] [CrossRef]
  200. Fecchio, A.; Martins, T.F.; Bell, J.A.; De La Torre, G.M.; Pinho, J.B.; Weckstein, J.D.; Tkach, V.V.; Labruna, M.B.; Dias, R.I. Low host specificity and lack of parasite avoidance by immature ticks in Brazilian birds. Parasitol. Res. 2020, 119, 2039–2045. [Google Scholar] [CrossRef]
  201. Sonenshine, D.E. Range Expansion of Tick Disease Vectors in North America: Implications for Spread of Tick-Borne Disease. Int. J. Environ. Res. Public Health 2018, 15, 478. [Google Scholar] [CrossRef]
  202. Molaei, G.; Little, E.A.; Williams, S.C.; Stafford, K.C. Bracing for the Worst—Range Expansion of the Lone Star Tick in the Northeastern United States. N. Engl. J. Med. 2019, 381, 2189–2192. [Google Scholar] [CrossRef]
  203. Tokarevich, N.K.; Tronin, A.A.; Blinova, O.V.; Buzinov, R.V.; Boltenkov, V.P.; Yurasova, E.D.; Nurse, J. The impact of climate change on the expansion of Ixodes persulcatus habitat and the incidence of tick-borne encephalitis in the north of European Russia. Glob. Health Action 2011, 4, 8448. [Google Scholar] [CrossRef]
  204. Ma, B.; Ma, X.Y.; Chen, H.B.; Zhang, Y.; Li, L.H. Effects of environmental factors on the distribution of suitable habitats of Ixodes ovatus in China. Chin. J. Schistosomiasis Control 2021, 33, 281–286. [Google Scholar] [CrossRef]
  205. Wallace, D.; Ratti, V.; Kodali, A.; Winter, J.M.; Ayres, M.P.; Chipman, J.W.; Aoki, C.F.; Osterberg, E.C.; Silvanic, C.; Partridge, T.F.; et al. Effect of Rising Temperature on Lyme Disease: Ixodes scapularis Population Dynamics and Borrelia burgdorferi Transmission and Prevalence. Can. J. Infect. Dis. Med. Microbiol. 2019, 2019, 9817930. [Google Scholar] [CrossRef]
  206. Cunze, S.; Glock, G.; Kochmann, J.; Klimpel, S. Ticks on the move—Climate change-induced range shifts of three tick species in Europe: Current and future habitat suitability for Ixodes ricinus in comparison with Dermacentor reticulatus and Dermacentor marginatus. Parasitol. Res. 2022, 121, 2241–2252. [Google Scholar] [CrossRef]
  207. Ogden, N.H.; Ben Beard, C.; Ginsberg, H.S.; Tsao, J.I. Possible Effects of Climate Change on Ixodid Ticks and the Pathogens They Transmit: Predictions and Observations. J. Med. Entomol. 2021, 58, 1536–1545. [Google Scholar] [CrossRef]
  208. Strona, G. Past, present and future of host–parasite co-extinctions. Int. J. Parasitol. Parasites Wildl. 2015, 4, 431–441. [Google Scholar] [CrossRef]
  209. Nabi, G.; Wang, Y.; Lü, L.; Jiang, C.; Ahmad, S.; Wu, Y.; Li, D. Bats and birds as viral reservoirs: A physiological and ecological perspective. Sci. Total Environ. 2021, 754, 142372. [Google Scholar] [CrossRef]
  210. Rahman, T.; Sobur, A.; Islam, S.; Ievy, S.; Hossain, J.; El Zowalaty, M.E.; Rahman, A.T.; Ashour, H.M. Zoonotic Diseases: Etiology, Impact, and Control. Microorganisms 2020, 8, 1405. [Google Scholar] [CrossRef]
  211. Egan, S.L.; Egan, S.L.; Taylor, C.L.; Taylor, C.L.; Banks, P.B.; Banks, P.B.; Northover, A.S.; Northover, A.S.; Ahlstrom, L.A.; Ahlstrom, L.A.; et al. The bacterial biome of ticks and their wildlife hosts at the urban–wildland interface. Microb. Genom. 2021, 7, 000730. [Google Scholar] [CrossRef]
  212. Paulraj, P.S.; Renu, G.; Ranganathan, K.; Veeramanoharan, R.; Kumar, A. Ectoparasites Diversity on Rodents and Shrews at Scrub Typhus Endemic Vellore District of Tamil Nadu, India. J. Arthropod-Borne Dis. 2022, 16, 51–60. [Google Scholar] [CrossRef]
  213. Kruse, H.; Kirkemo, A.-M.; Handeland, K. Wildlife as Source of Zoonotic Infections. Emerg. Infect. Dis. 2004, 10, 2067–2072. [Google Scholar] [CrossRef]
  214. Titcomb, G.; Allan, B.F.; Ainsworth, T.; Henson, L.; Hedlund, T.; Pringle, R.M.; Palmer, T.M.; Njoroge, L.; Campana, M.G.; Fleischer, R.C.; et al. Interacting effects of wildlife loss and climate on ticks and tick-borne disease. Proc. R. Soc. B Biol. Sci. 2017, 284, 20170475. [Google Scholar] [CrossRef] [PubMed]
  215. Chepkwony, R.; Castagna, C.; Heitkönig, I.; van Bommel, S.; van Langevelde, F. Associations between monthly rainfall and mortality in cattle due to East Coast fever, anaplasmosis and babesiosis. Parasitology 2020, 147, 1743–1751. [Google Scholar] [CrossRef] [PubMed]
  216. Pattnaik, P. Kyasanur forest disease: An epidemiological view in India. Rev. Med. Virol. 2006, 16, 151–165. [Google Scholar] [CrossRef]
  217. Rajaiah, P. Kyasanur Forest Disease in India: Innovative options for intervention. Hum. Vaccines Immunother. 2019, 15, 2243–2248. [Google Scholar] [CrossRef]
  218. Chanda, M.M.; Kharkwal, P.; Dhuria, M.; Prajapathi, A.; Yogisharadhya, R.; Shome, B.R.; Shivachandra, S.B. Quantifying the influence of climate, host and change in land-use patterns on occurrence of Crimean Congo Hemorrhagic Fever (CCHF) and development of spatial risk map for India. One Health 2023, 17, 100609. [Google Scholar] [CrossRef]
  219. Mourya, D.T.; Yadav, P.D.; Gurav, Y.K.; Pardeshi, P.G.; Shete, A.M.; Jain, R.; Raval, D.D.; Upadhyay, K.J.; Patil, D.Y. Crimean Congo hemorrhagic fever serosurvey in humans for identifying high-risk populations and high-risk areas in the endemic state of Gujarat, India. BMC Infect. Dis. 2019, 19, 104. [Google Scholar] [CrossRef] [PubMed]
  220. Dash, N.; Gonttumukkula, V.; Samyanathan, P.; Rajangam, M.; Biswal, M.; Verma, S. Indian Tick Typhus Presenting as Gangrene: A Case Report. Pediatr. Infect. Dis. J. 2023, 42, e249–e250. [Google Scholar] [CrossRef]
  221. Vinayaraj, E.; Gupta, N.; Sreenath, K.; Thakur, C.K.; Gulati, S.; Anand, V.; Tripathi, M.; Bhatia, R.; Vibha, D.; Dash, D.; et al. Clinical and laboratory evidence of Lyme disease in North India, 2016–2019. Travel Med. Infect. Dis. 2021, 43, 102134. [Google Scholar] [CrossRef]
  222. Balasubramanian, R.; Fournier, P.-E.; Ganesan, P.S.; Menon, T. Q fever endocarditis in India: A report of two cases. Indian J. Med. Microbiol. 2022, 40, 315–316. [Google Scholar] [CrossRef]
  223. Paramanandham, K.; Mohankumar, A.; Suresh, K.P.; Jacob, S.S.; Roy, P. Prevalence of Anaplasma species in India and the World in dairy animals: A systematic review and meta-analysis. Res. Vet. Sci. 2019, 123, 159–170. [Google Scholar] [CrossRef]
  224. Shah, S.Z.; Jabbar, B.; Ahmed, N.; Rehman, A.; Nasir, H.; Nadeem, S.; Jabbar, I.; Rahman, Z.U.; Azam, S. Epidemiology, Pathogenesis, and Control of a Tick-Borne Disease-Kyasanur Forest Disease: Current Status and Future Directions. Front. Cell. Infect. Microbiol. 2018, 8, 149. [Google Scholar] [CrossRef] [PubMed]
  225. Holbrook, M.R. Kyasanur forest disease. Antivir. Res. 2012, 96, 353–362. [Google Scholar] [CrossRef] [PubMed]
  226. Yadav, P.D.; Patil, S.; Jadhav, S.M.; Nyayanit, D.A.; Kumar, V.; Jain, S.; Sampath, J.; Mourya, D.T.; Cherian, S.S. Phylogeography of Kyasanur Forest Disease virus in India (1957–2017) reveals evolution and spread in the Western Ghats region. Sci. Rep. 2020, 10, 1966. [Google Scholar] [CrossRef]
  227. Pramanik, M.; Singh, P.; Dhiman, R.C. Identification of bio-climatic determinants and potential risk areas for Kyasanur forest disease in Southern India using MaxEnt modelling approach. BMC Infect. Dis. 2021, 21, 1226. [Google Scholar] [CrossRef]
  228. Bhatia, B.; Feldmann, H.; Marzi, A. Kyasanur Forest Disease and Alkhurma Hemorrhagic Fever Virus—Two Neglected Zoonotic Pathogens. Microorganisms 2020, 8, 1406. [Google Scholar] [CrossRef]
  229. Balasubramanian, R.; Yadav, P.D.; Sahina, S.; Nadh, V.A. The species distribution of ticks & the prevalence of Kyasanur forest disease virus in questing nymphal ticks from Western Ghats of Kerala, South India. Indian J. Med. Res. 2021, 154, 743–749. [Google Scholar] [CrossRef]
  230. Papa, A.; Sidira, P.; Larichev, V.; Gavrilova, L.; Kuzmina, K.; Mousavi-Jazi, M.; Mirazimi, A.; Ströher, U.; Nichol, S. Crimean-Congo hemorrhagic fever virus, Greece. Emerg. Infect. Dis. 2014, 20, 288–290. [Google Scholar] [CrossRef]
  231. Hawman, D.W.; Feldmann, H. Crimean–Congo haemorrhagic fever virus. Nat. Rev. Microbiol. 2023, 21, 463–477. [Google Scholar] [CrossRef]
  232. Patel, A.A.; Dalal, Y.D.; Parikh, A.; Gandhi, R.; Shah, A.; Patel, A.A. Crimean-Congo Hemorrhagic Fever: An Emerging Viral Infection in India, Revisited and Lessons Learned. Cureus 2023, 15, e43315. [Google Scholar] [CrossRef]
  233. Sharma, S.N.; Singh, R.; Balakrishnan, N.; Kumawat, R.; Singh, S.K. Vectors of Crimean-Congo Hemorrhagic Fever (CCHF): Prevention and its Control. J. Commun. Dis. 2020, 52, 22–26. [Google Scholar] [CrossRef]
  234. Ergönül, Ö. Crimean-Congo haemorrhagic fever. Lancet Infect. Dis. 2006, 6, 203–214. [Google Scholar] [CrossRef] [PubMed]
  235. Coburn, J.; Garcia, B.; Hu, L.T.; Jewett, M.W.; Kraiczy, P.; Norris, S.J.; Skare, J.; Garcia, O. Lyme Disease Pathogenesis. Curr. Issues Mol. Biol. 2021, 42, 473–518. [Google Scholar] [CrossRef] [PubMed]
  236. Hamer, S.A.; Tsao, J.I.; Walker, E.D.; Hickling, G.J. Invasion of the Lyme Disease Vector Ixodes scapularis: Implications for Borrelia burgdorferi Endemicity. Ecohealth 2010, 7, 47–63. [Google Scholar] [CrossRef]
  237. Shapiro, E.D. Borrelia burgdorferi (Lyme Disease). Pediatr. Rev. 2014, 35, 500–509. [Google Scholar] [CrossRef]
  238. Anguita, J.; Hedrick, M.N.; Fikrig, E. Adaptation of Borrelia burgdorferi in the tick and the mammalian host. FEMS Microbiol. Rev. 2003, 27, 493–504. [Google Scholar] [CrossRef] [PubMed]
  239. Nováková, M.; Šmajs, D. Rickettsial Endosymbionts of Ticks; IntechOpen: London, UK, 2019. [Google Scholar] [CrossRef]
  240. Tomassone, L.; Portillo, A.; Nováková, M.; de Sousa, R.; Oteo, J.A. Neglected aspects of tick-borne rickettsioses. Parasites Vectors 2018, 11, 263. [Google Scholar] [CrossRef]
  241. Rahi, M.; Gupte, M.D.; Bhargava, A.; Varghese, G.M.; Arora, R. DHR-ICMR Guidelines for diagnosis & management of Rickettsial diseases in India. Indian J. Med. Res. 2015, 141, 417–422. [Google Scholar] [CrossRef]
  242. Sahni, A.; Fang, R.; Sahni, S.K.; Walker, D.H. Pathogenesis of Rickettsial Diseases: Pathogenic and Immune Mechanisms of an Endotheliotropic Infection. Annu. Rev. Pathol. Mech. Dis. 2019, 14, 127–152. [Google Scholar] [CrossRef] [PubMed]
  243. Hulmani, M.; Alekya, P.; Kumar, V. Indian Tick Typhus Presenting as Purpura Fulminans with Review on Rickettsial Infections. Indian J. Dermatol. 2017, 62, 1–6. [Google Scholar] [CrossRef] [PubMed]
  244. Sharma, A.; Mishra, B. Rickettsial disease existence in India: Resurgence in outbreaks with the advent of 20th century. Indian J. Health Sci. Biomed. Res. (KLEU) 2020, 13, 5. [Google Scholar] [CrossRef]
  245. Krishnamoorthi, S.; Goel, S.; Kaur, J.; Bisht, K.; Biswal, M. A Review of Rickettsial Diseases Other Than Scrub Typhus in India. Trop. Med. Infect. Dis. 2023, 8, 280. [Google Scholar] [CrossRef]
  246. Matei, I.A.; Estrada-Peña, A.; Cutler, S.J.; Vayssier-Taussat, M.; Varela-Castro, L.; Potkonjak, A.; Zeller, H.; Mihalca, A.D. A review on the eco-epidemiology and clinical management of human granulocytic anaplasmosis and its agent in Europe. Parasites Vectors 2019, 12, 599. [Google Scholar] [CrossRef]
  247. Diuk-Wasser, M.A.; VanAcker, M.C.; Fernandez, M.P. Impact of Land Use Changes and Habitat Fragmentation on the Eco-epidemiology of Tick-Borne Diseases. J. Med. Entomol. 2021, 58, 1546–1564. [Google Scholar] [CrossRef]
  248. March, D.; Susser, E. The eco- in eco-epidemiology. Leuk. Res. 2006, 35, 1379–1383. [Google Scholar] [CrossRef] [PubMed]
  249. Bain, L.E.; Awah, P.K. Eco–epidemiology: Challenges and opportunities for tomorrow’s epidemiologists. Pan Afr. Med. J. 2014, 17, 317. [Google Scholar] [CrossRef] [PubMed]
  250. Dhiman, R.C. Emerging Vector-Borne Zoonoses: Eco-Epidemiology and Public Health Implications in India. Front. Public Health 2014, 2, 168. [Google Scholar] [CrossRef] [PubMed]
  251. MacDonald, A.J.; McComb, S.; Sambado, S. Linking Lyme disease ecology and epidemiology: Reservoir host identity, not richness, determines tick infection and human disease in California. Environ. Res. Lett. 2022, 17, 114041. [Google Scholar] [CrossRef]
  252. Estrada-Peña, A.; Ayllón, N.; de la Fuente, J. Impact of Climate Trends on Tick-Borne Pathogen Transmission. Front. Physiol. 2012, 3, 64. [Google Scholar] [CrossRef]
  253. Pfäffle, M.; Littwin, N.; Muders, S.V.; Petney, T.N. The ecology of tick-borne diseases. Int. J. Parasitol. 2013, 43, 1059–1077. [Google Scholar] [CrossRef]
  254. Estrada-Peña, A.; Jameson, L.; Medlock, J.; Vatansever, Z.; Tishkova, F. Unraveling the Ecological Complexities of Tick-Associated Crimean-Congo Hemorrhagic Fever Virus Transmission: A Gap Analysis for the Western Palearctic. Vector-Borne Zoonotic Dis. 2012, 12, 743–752. [Google Scholar] [CrossRef]
  255. Murhekar, M.V.; Kasabi, G.S.; Mehendale, S.M.; Mourya, D.T.; Yadav, P.D.; Tandale, B.V. On the transmission pattern of Kyasanur Forest disease (KFD) in India. Infect. Dis. Poverty 2015, 4, 37. [Google Scholar] [CrossRef]
  256. Alcon-Chino, M.E.T.; De-Simone, S.G. Recent Advances in the Immunologic Method Applied to Tick-Borne Diseases in Brazil. Pathogens 2022, 11, 870. [Google Scholar] [CrossRef]
  257. Kularatne, S.A.; Gawarammana, I.B. Validity of the Weil-Felix test in the diagnosis of acute rickettsial infections in Sri Lanka. Trans. R. Soc. Trop. Med. Hyg. 2009, 103, 423–424. [Google Scholar] [CrossRef]
  258. Shriyan, A.S.A. An Atypical Presentation of Rocky Mountain spotted fever (RMSF)—A Case Report. J. Clin. Diagn. Res. 2010, 4, 2546–2549. [Google Scholar]
  259. Garcia, K.; Weakley, M.; Do, T.; Mir, S. Current and Future Molecular Diagnostics of Tick-Borne Diseases in Cattle. Vet. Sci. 2022, 9, 241. [Google Scholar] [CrossRef]
  260. Guillemi, E.C.; Tomassone, L.; Farber, M.D. Tick-borne Rickettsiales: Molecular tools for the study of an emergent group of pathogens. J. Microbiol. Methods 2015, 119, 87–97. [Google Scholar] [CrossRef]
  261. Liu, Q.; Jin, X.; Cheng, J.; Zhou, H.; Zhang, Y.; Dai, Y. Advances in the application of molecular diagnostic techniques for the detection of infectious disease pathogens (Review). Mol. Med. Rep. 2023, 27, 104. [Google Scholar] [CrossRef]
  262. Amin, I.; Idrees, M.; Awan, Z.; Shahid, M.; Afzal, S.; Hussain, A. PCR could be a method of choice for identification of both pulmonary and extra-pulmonary tuberculosis. BMC Res. Notes 2011, 4, 332. [Google Scholar] [CrossRef]
  263. Navarro, E.; Serrano-Heras, G.; Castaño, M.; Solera, J. Real-time PCR detection chemistry. Clin. Chim. Acta 2015, 439, 231–250. [Google Scholar] [CrossRef]
  264. Tokarz, R.; Lipkin, W.I. Discovery and Surveillance of Tick-Borne Pathogens. J. Med. Entomol. 2021, 58, 1525–1535. [Google Scholar] [CrossRef]
  265. Radzi, S.F.M.; Rückert, C.; Sam, S.-S.; Teoh, B.-T.; Jee, P.-F.; Phoon, W.-H.; Abubakar, S.; Zandi, K. Detection of Langat virus by TaqMan real-time one-step qRT-PCR method. Sci. Rep. 2015, 5, 14007. [Google Scholar] [CrossRef]
  266. Modarelli, J.J.; Ferro, P.J.; de León, A.A.P.; Esteve-Gasent, M.D. TickPath Layerplex: Adaptation of a real-time PCR methodology for the simultaneous detection and molecular surveillance of tick-borne pathogens. Sci. Rep. 2019, 9, 6950. [Google Scholar] [CrossRef]
  267. Zakham, F.; Korhonen, E.M.; Puonti, P.T.; Castrén, R.S.; Uusitalo, R.; Smura, T.; Kant, R.; Vapalahti, O.; Sironen, T.; Kinnunen, P.M. Molecular detection of pathogens from ticks collected from dogs and cats at veterinary clinics in Finland. Parasites Vectors 2023, 16, 327. [Google Scholar] [CrossRef]
  268. Pratt, G.W.; Platt, M.; Velez, A.; Rao, L.V. Utility of Whole Blood Real-Time PCR Testing for the Diagnosis of Early Lyme Disease. Am. J. Clin. Pathol. 2022, 158, 327–330. [Google Scholar] [CrossRef]
  269. Sukhiashvili, R.; Zhgenti, E.; Khmaladze, E.; Burjanadze, I.; Imnadze, P.; Jiang, J.; St John, H.; Farris, C.M.; Gallagher, T.; Obiso, R.J.; et al. Identification and distribution of nine tick-borne spotted fever group Rickettsiae in the Country of Georgia. Ticks Tick-Borne Dis. 2020, 11, 101470. [Google Scholar] [CrossRef]
  270. Radolf, J.D.; Strle, K.; Lemieux, J.E.; Strle, F. Lyme Disease in Humans. Curr. Issues Mol. Biol. 2021, 42, 333–384. [Google Scholar] [CrossRef]
  271. Tiffin, H.S.; Rajotte, E.G.; Sakamoto, J.M.; Machtinger, E.T. Tick Control in a Connected World: Challenges, Solutions, and Public Policy from a United States Border Perspective. Trop. Med. Infect. Dis. 2022, 7, 388. [Google Scholar] [CrossRef]
  272. Sparagano, O.; Földvári, G.; Derdáková, M.; Kazimírová, M. New challenges posed by ticks and tick-borne diseases. Biologia 2022, 77, 1497–1501. [Google Scholar] [CrossRef]
  273. Narurkar, R.; Mamorska-Dyga, A.; Nelson, J.C.; Liu, D. Autoimmune hemolytic anemia associated with babesiosis. Biomark. Res. 2017, 5, 14. [Google Scholar] [CrossRef]
  274. Singh, K.; Kumar, S.; Sharma, A.K.; Jacob, S.; RamVerma, M.; Singh, N.K.; Shakya, M.; Sankar, M.; Ghosh, S. Economic impact of predominant ticks and tick-borne diseases on Indian dairy production systems. Exp. Parasitol. 2022, 243, 108408. [Google Scholar] [CrossRef]
  275. Sharma, S.R.; Karim, S. Tick Saliva and the Alpha-Gal Syndrome: Finding a Needle in a Haystack. Front. Cell. Infect. Microbiol. 2021, 11, 680264. [Google Scholar] [CrossRef]
  276. Barkema, H.W.; Von Keyserlingk, M.A.G.; Kastelic, J.P.; Lam, T.J.G.M.; Luby, C.; Roy, J.-P.; Leblanc, S.J.; Keefe, G.P.; Kelton, D.F. Invited review: Changes in the dairy industry affecting dairy cattle health and welfare. J. Dairy Sci. 2015, 98, 7426–7445. [Google Scholar] [CrossRef]
  277. Mandli, J.T.; Lee, X.; Bron, G.M.; Paskewitz, S.M. Integrated Tick Management in South Central Wisconsin: Impact of Invasive Vegetation Removal and Host-Targeted Acaricides on the Density of Questing Ixodes scapularis (Acari: Ixodidae) Nymphs. J. Med. Entomol. 2021, 58, 2358–2367. [Google Scholar] [CrossRef]
  278. Wróbel, B.; Zielewicz, W.; Staniak, M. Challenges of Pasture Feeding Systems—Opportunities and Constraints. Agriculture 2023, 13, 974. [Google Scholar] [CrossRef]
  279. Hue, T.; Wang, H.-H.; Grant, W.E.; Teel, P.D.; de Leon, A.A.P. Implementation Research for Integrated Tick Control of Rhipicephalus australis (Acari: Ixodidae) Through the Pasture and Cattle Management Method in New Caledonia. J. Integr. Pest Manag. 2022, 13, 26. [Google Scholar] [CrossRef]
  280. Schulze, T.L.; Eisen, L.; Russell, K.; Jordan, R.A. Community-based integrated tick management programs: Cost and feasibility scenarios. J. Med. Entomol. 2023, 60, 1048–1060. [Google Scholar] [CrossRef]
  281. Stafford, K.C. Tick Management Handbook: An Integrated Guide for Homeowners, Pest Control Operators, and Public Health Officials for the Prevention of Tick-Associated Disease; The Connecticut Agricultural Experiment Station: New Heaven, CT, USA, 2007.
  282. de León, A.A.P.; Teel, P.D.; Auclair, A.N.; Messenger, M.T.; Guerrero, F.D.; Schuster, G.; Miller, R.J. Integrated Strategy for Sustainable Cattle Fever Tick Eradication in USA is Required to Mitigate the Impact of Global Change. Front. Physiol. 2012, 3, 195. [Google Scholar] [CrossRef]
  283. Obaid, M.K.; Islam, N.; Alouffi, A.; Khan, A.Z.; Vaz, I.d.S.; Tanaka, T.; Ali, A. Acaricides Resistance in Ticks: Selection, Diagnosis, Mechanisms, and Mitigation. Front. Cell. Infect. Microbiol. 2022, 12, 941831. [Google Scholar] [CrossRef]
  284. Chizyuka, H.G.; Mulilo, J.B. Methods currently used for the control of multi-host ticks: Their validity and proposals for future control strategies. Parassitologia 1990, 32, 127–132. [Google Scholar]
  285. Adenubi, O.; Fasina, F.; McGaw, L.; Eloff, J.; Naidoo, V. Plant extracts to control ticks of veterinary and medical importance: A review. S. Afr. J. Bot. 2016, 105, 178–193. [Google Scholar] [CrossRef]
  286. Kwenti, T.E. Biological Control of Parasites; InTech: London, UK, 2017. [Google Scholar] [CrossRef]
  287. Ostfeld, R.S.; Brunner, J.L. Climate change and Ixodes tick-borne diseases of humans. Philos. Trans. R. Soc. Lond. Ser. B Biol. Sci. 2015, 370, 20140051. [Google Scholar] [CrossRef] [PubMed]
  288. Garris, G.I. Control of Ticks. Vet. Clin. N. Am. Small Anim. Pract. 1991, 21, 173–183. [Google Scholar] [CrossRef] [PubMed]
  289. Alonso-Díaz, M.A.; Fernández-Salas, A. Entomopathogenic Fungi for Tick Control in Cattle Livestock from Mexico. Front. Fungal Biol. 2021, 2, 657694. [Google Scholar] [CrossRef]
  290. Dzemo, W.D.; Thekisoe, O.; Vudriko, P. Development of acaricide resistance in tick populations of cattle: A systematic review and meta-analysis. Heliyon 2022, 8, e08718. [Google Scholar] [CrossRef]
  291. Waldman, J.; Klafke, G.M.; Tirloni, L.; Logullo, C.; da Silva Vaz, I., Jr. Putative target sites in synganglion for novel ixodid tick control strategies. Ticks Tick-Borne Dis. 2023, 14, 102123. [Google Scholar] [CrossRef]
  292. Nagar, G.; Upadhaya, D.; Sharma, A.K.; Kumar, R.; Fular, A.; Ghosh, S. Association between overexpression of cytochrome P450 genes and deltamethrin resistance in Rhipicephalus microplus. Ticks Tick-Borne Dis. 2021, 12, 101610. [Google Scholar] [CrossRef]
  293. Gupta, S.; Sangwan, N.; Sangwan, A.K.; Mann, S.; Gupta, S.; Kumar, A.; Kumar, S. Understanding the resistance mechanisms of Rhipicephalus microplus ticks to synthetic pyrethroids and organophosphates in south-west regions of Haryana, North India. Pestic. Biochem. Physiol. 2023, 196, 105634. [Google Scholar] [CrossRef]
  294. Chitombo, L.; Lebani, K.; Sungirai, M. Acaricide resistance in Rhipicephalus appendiculatus ticks collected from different farming systems in Zimbabwe. Trop. Anim. Health Prod. 2021, 53, 431. [Google Scholar] [CrossRef]
  295. Li, A.Y.; Chen, A.C.; Miller, R.J.; Davey, R.B.; George, J.E. Acaricide resistance and synergism between permethrin and amitraz against susceptible and resistant strains of Boophilus microplus (Acari: Ixodidae). Pest Manag. Sci. 2007, 63, 882–889. [Google Scholar] [CrossRef]
  296. Donovan, B.J.; Weber, D.J.; Rublein, J.C.; Raasch, R.H. Treatment of Tick-Borne Diseases. Ann. Pharmacother. 2002, 36, 1590–1597. [Google Scholar] [CrossRef]
  297. Sanchez-Vicente, S.; Tokarz, R. Tick-Borne Co-Infections: Challenges in Molecular and Serologic Diagnoses. Pathogens 2023, 12, 1371. [Google Scholar] [CrossRef] [PubMed]
  298. Sarli, M.; Miró, M.V.; Rossner, M.V.; Nava, S.; Lifschitz, A. Successive treatments with ivermectin (3.15%) to control the tick Rhipicephalus (Boophilus) microplus in cattle: Pharmacokinetic and efficacy assessment. Ticks Tick-Borne Dis. 2021, 13, 101848. [Google Scholar] [CrossRef] [PubMed]
  299. Gonzales, J.; Muniz, R.; Farias, A.; Goncalves, L.; Rew, R. Therapeutic and persistent efficacy of doramectin against Boophilus microplus in cattle. Vet. Parasitol. 1993, 49, 107–119. [Google Scholar] [CrossRef]
  300. Abbas, M.N.; Jmel, M.A.; Mekki, I.; Dijkgraaf, I.; Kotsyfakis, M. Recent Advances in Tick Antigen Discovery and Anti-Tick Vaccine Development. Int. J. Mol. Sci. 2023, 24, 4969. [Google Scholar] [CrossRef]
  301. Bragazzi, N.L.; Gianfredi, V.; Villarini, M.; Rosselli, R.; Nasr, A.; Hussein, A.; Martini, M.; Behzadifar, M. Vaccines Meet Big Data: State-of-the-Art and Future Prospects. From the Classical 3Is (“Isolate–Inactivate–Inject”) Vaccinology 1.0 to Vaccinology 3.0, Vaccinomics, and Beyond: A Historical Overview. Front. Public Health 2018, 6, 62. [Google Scholar] [CrossRef]
  302. Domnich, A.; Panatto, D.; Arbuzova, E.K.; Signori, A.; Avio, U.; Gasparini, R.; Amicizia, D. Immunogenicity against Far Eastern and Siberian subtypes of tick-borne encephalitis (TBE) virus elicited by the currently available vaccines based on the European subtype: Systematic review and meta-analysis. Hum. Vaccines Immunother. 2014, 10, 2819–2833. [Google Scholar] [CrossRef]
  303. Zavadska, D.; Anca, I.; Andre, F.; Bakir, M.; Chlibek, R.; Čižman, M.; Ivaskeviciene, I.; Mangarov, A.; Mészner, Z.; Pokorn, M.; et al. Recommendations for tick-borne encephalitis vaccination from the Central European Vaccination Awareness Group (CEVAG). Hum. Vaccines Immunother. 2013, 9, 362–374. [Google Scholar] [CrossRef]
  304. Angulo, F.J.; Zhang, P.; Halsby, K.; Kelly, P.; Pilz, A.; Madhava, H.; Moïsi, J.C.; Jodar, L. A systematic literature review of the effectiveness of tick-borne encephalitis vaccines in Europe. Vaccine 2023, 41, 6914–6921. [Google Scholar] [CrossRef]
  305. Oliveira, A.; Selvaraj, K.; Tripathy, J.P.; Betodkar, U.; Cacodcar, J.; Wadkar, A. Kyasanur Forest Disease vaccination coverage and its perceived barriers in Goa, India—A mixed methods operational research. PLoS ONE 2019, 14, e0226141. [Google Scholar] [CrossRef]
  306. Kasabi, G.S.; Murhekar, M.V.; Sandhya, V.K.; Raghunandan, R.; Kiran, S.K.; Channabasappa, G.H.; Mehendale, S.M. Coverage and Effectiveness of Kyasanur Forest Disease (KFD) Vaccine in Karnataka, South India, 2005–2010. PLoS Neglected Trop. Dis. 2013, 7, e2025. [Google Scholar] [CrossRef]
  307. Bhatia, B.; Tang-Huau, T.-L.; Feldmann, F.; Hanley, P.W.; Rosenke, R.; Shaia, C.; Marzi, A.; Feldmann, H. Single-dose VSV-based vaccine protects against Kyasanur Forest disease in nonhuman primates. Sci. Adv. 2023, 9, eadj1428. [Google Scholar] [CrossRef]
  308. Valle, M.R.; Mèndez, L.; Valdez, M.; Redondo, M.; Espinosa, C.M.; Vargas, M.; Cruz, R.L.; Barrios, H.P.; Seoane, G.; Ramirez, E.S.; et al. Integrated control of Boophilus microplus ticks in Cuba based on vaccination with the anti-tick vaccine GavacTM. Exp. Appl. Acarol. 2004, 34, 375–382. [Google Scholar] [CrossRef]
  309. Ndawula, C. From Bench to Field: A Guide to Formulating and Evaluating Anti-Tick Vaccines Delving beyond Efficacy to Effectiveness. Vaccines 2021, 9, 1185. [Google Scholar] [CrossRef]
  310. Kasaija, P.D.; Contreras, M.; Kirunda, H.; Nanteza, A.; Kabi, F.; Mugerwa, S.; de la Fuente, J. Inspiring Anti-Tick Vaccine Research, Development and Deployment in Tropical Africa for the Control of Cattle Ticks: Review and Insights. Vaccines 2022, 11, 99. [Google Scholar] [CrossRef]
  311. Sharma, S.N.; Kumawat, R.; Singh, S.K. Kyasanur Forest Disease: Vector Surveillance and its Control. J. Commun. Dis. 2019, 51, 38–44. [Google Scholar] [CrossRef]
  312. Mostafavizadeh, K.; Ataei, B.; Rostami, M.; Salehi, H.; Karimi, I.; Javadi, A.A.; Sherkat, R.; Emami, A.R.; Khademi, M.R.; Chinikar, M.; et al. Crime congo hemorhagic fever epidemy: A preliminary report of Isfahan province in Iran. J. Res. Med. Sci. 2002, 7, 78–79. [Google Scholar]
  313. Bonnet, S.I.; Vourc’h, G.; Raffetin, A.; Falchi, A.; Figoni, J.; Fite, J.; Hoch, T.; Moutailler, S.; Quillery, E. The control of Hyalomma ticks, vectors of the Crimean–Congo hemorrhagic fever virus: Where are we now and where are we going? PLoS Neglected Trop. Dis. 2022, 16, e0010846. [Google Scholar] [CrossRef]
  314. Kumar, K.; Jain, S.; Kimar, A.; Sharma, A. Outbreak Indian Tick Typhus amongst residents of Deol village, District, Kangra, Himachal Pradesh (INDIA). Int. J. Med. Public Health 2011, 1, 67–71. [Google Scholar] [CrossRef]
Figure 1. Images of characteristic ticks: (A) Ornithodoros spp. (Soft tick), (B) Amblyomma integrum (Hard tick) and (C) Nuttaliella namaqua (Monotypic tick), (Images (A,C) adapted from [63,66]), respectively.
Figure 1. Images of characteristic ticks: (A) Ornithodoros spp. (Soft tick), (B) Amblyomma integrum (Hard tick) and (C) Nuttaliella namaqua (Monotypic tick), (Images (A,C) adapted from [63,66]), respectively.
Pathogens 13 00556 g001
Figure 2. The figure depicts the morphology of the male tick Rhipicephalus sanguineus, commonly known as the brown dog tick. In the right panel, a dorsal view highlights the distinct shape of the tick, showcasing a well-defined scutum and festoons. On the left side of the illustration, a ventral view is presented with detailed annotations for the genital aperture, coxa, anal region, and anal groove. Notably, the adanal plate’s shape is emphasized as a key characteristic feature for species identification.
Figure 2. The figure depicts the morphology of the male tick Rhipicephalus sanguineus, commonly known as the brown dog tick. In the right panel, a dorsal view highlights the distinct shape of the tick, showcasing a well-defined scutum and festoons. On the left side of the illustration, a ventral view is presented with detailed annotations for the genital aperture, coxa, anal region, and anal groove. Notably, the adanal plate’s shape is emphasized as a key characteristic feature for species identification.
Pathogens 13 00556 g002
Figure 3. The figure depicts the life cycle of one-host tick (e.g., Rhipicephalus (B) microplus) and two-host tick (Hyalomma spp.) [67,94] (illustration created using by BioRender.com, Toronto, Canada).
Figure 3. The figure depicts the life cycle of one-host tick (e.g., Rhipicephalus (B) microplus) and two-host tick (Hyalomma spp.) [67,94] (illustration created using by BioRender.com, Toronto, Canada).
Pathogens 13 00556 g003
Figure 4. A diagram illustrates the life cycle of the three-host tick Haemaphysalis spinigera and its interactions with hosts, highlighting the transmission of the Kyasanur Forest Disease Virus (KFDV). The figure depicts questing ticks and their infestation process involving humans and domestic animals. Special attention is given to the co-feeding phenomenon [14], demonstrating the simultaneous presence of infected and non-infected ticks on the same host (illustration created by using BioRender.com).
Figure 4. A diagram illustrates the life cycle of the three-host tick Haemaphysalis spinigera and its interactions with hosts, highlighting the transmission of the Kyasanur Forest Disease Virus (KFDV). The figure depicts questing ticks and their infestation process involving humans and domestic animals. Special attention is given to the co-feeding phenomenon [14], demonstrating the simultaneous presence of infected and non-infected ticks on the same host (illustration created by using BioRender.com).
Pathogens 13 00556 g004
Figure 5. Illustration of dorsal (A) and ventral (B) view of partially fed and dorsal (C) and ventral (D) view of engorged female ticks of Rhipicephalus spp.
Figure 5. Illustration of dorsal (A) and ventral (B) view of partially fed and dorsal (C) and ventral (D) view of engorged female ticks of Rhipicephalus spp.
Pathogens 13 00556 g005
Figure 6. A comprehensive depiction of a tick bite is presented, elucidating the intricate process through which the tick injects various components into its host. The visual narrative highlights the molecular and biochemical aspects of the tick–host interaction, unveiling the complexity inherent in this parasitic relationship. (Illustration created by using BioRender.com).
Figure 6. A comprehensive depiction of a tick bite is presented, elucidating the intricate process through which the tick injects various components into its host. The visual narrative highlights the molecular and biochemical aspects of the tick–host interaction, unveiling the complexity inherent in this parasitic relationship. (Illustration created by using BioRender.com).
Pathogens 13 00556 g006
Figure 7. Map of important tick vectors responsible for major TBDs in India.
Figure 7. Map of important tick vectors responsible for major TBDs in India.
Pathogens 13 00556 g007
Figure 8. A phylogenetic tree utilizing the Neighbour-Joining method with a Bootstrap value of 1000 was generated from 35 nucleotide sequences of the 16S mitochondrial gene of Rhipicephalus sanguineus collected from diverse geographical regions. The analysis excluded any ambiguous positions for each sequence pair through pairwise deletion, resulting in a final dataset of 347 positions. The evolutionary analyses were conducted using MEGA11, and consisted of 347 positions.
Figure 8. A phylogenetic tree utilizing the Neighbour-Joining method with a Bootstrap value of 1000 was generated from 35 nucleotide sequences of the 16S mitochondrial gene of Rhipicephalus sanguineus collected from diverse geographical regions. The analysis excluded any ambiguous positions for each sequence pair through pairwise deletion, resulting in a final dataset of 347 positions. The evolutionary analyses were conducted using MEGA11, and consisted of 347 positions.
Pathogens 13 00556 g008
Figure 9. A schematic representation highlights the geographical expansion of ticks, underscoring the significant role played by global warming and climate change in facilitating this spread. It illustrates the impact of climate change on tick habitats and distribution, emphasizing the connection between the loss of floral and faunal diversity and the subsequent increase in tick populations. The figure elucidates the interrelationship between climate change and human activities, emphasizing how anthropogenic factors contribute to the proliferation of ticks. This visual representation offers a comprehensive overview of the intricate dynamics among environmental changes, human activities, and the escalating prevalence of ticks in various regions. (Illustration created by using BioRender.com).
Figure 9. A schematic representation highlights the geographical expansion of ticks, underscoring the significant role played by global warming and climate change in facilitating this spread. It illustrates the impact of climate change on tick habitats and distribution, emphasizing the connection between the loss of floral and faunal diversity and the subsequent increase in tick populations. The figure elucidates the interrelationship between climate change and human activities, emphasizing how anthropogenic factors contribute to the proliferation of ticks. This visual representation offers a comprehensive overview of the intricate dynamics among environmental changes, human activities, and the escalating prevalence of ticks in various regions. (Illustration created by using BioRender.com).
Pathogens 13 00556 g009
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Perumalsamy, N.; Sharma, R.; Subramanian, M.; Nagarajan, S.A. Hard Ticks as Vectors: The Emerging Threat of Tick-Borne Diseases in India. Pathogens 2024, 13, 556. https://doi.org/10.3390/pathogens13070556

AMA Style

Perumalsamy N, Sharma R, Subramanian M, Nagarajan SA. Hard Ticks as Vectors: The Emerging Threat of Tick-Borne Diseases in India. Pathogens. 2024; 13(7):556. https://doi.org/10.3390/pathogens13070556

Chicago/Turabian Style

Perumalsamy, Nandhini, Rohit Sharma, Muthukumaravel Subramanian, and Shriram Ananganallur Nagarajan. 2024. "Hard Ticks as Vectors: The Emerging Threat of Tick-Borne Diseases in India" Pathogens 13, no. 7: 556. https://doi.org/10.3390/pathogens13070556

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop