Open access peer-reviewed chapter

Molecular Dynamics of Mosquito-Plasmodium vivax Interaction: A Smart Strategy of Parasitism

Written By

Charu Chauhan, Sanjay Tevatiya, Seena Kumari, Punita Sharma, Jyoti Rani and Rajnikant Dixit

Submitted: 05 August 2020 Reviewed: 14 January 2021 Published: 23 March 2021

DOI: 10.5772/intechopen.96008

From the Edited Volume

Cell Interaction - Molecular and Immunological Basis for Disease Management

Edited by Bhawana Singh

Chapter metrics overview

429 Chapter Downloads

View Full Metrics

Abstract

Parallel to Plasmodium falciparum, P. vivax is a fast emerging challenge to control malaria in South-East Asia regions. Owing to unique biological differences such as the preference for invading reticulocytes, early maturation of sexual stages during the infection, the formation of hypnozoites, unavailability of in-vitro culture, the molecular relation of P. vivax development inside the mosquito host is poorly known. In this chapter, we briefly provide a basic overview of Mosquito-Plasmodium interaction and update current knowledge of tissue-specific viz. midgut, hemocyte, and salivary glands- molecular dynamics of Plasmodium vivax interaction during its developmental transformation inside the mosquito host, in specific.

Keywords

  • malaria
  • mosquito
  • Anopheles
  • Plasmodium vivax
  • host–parasite interaction

1. Introduction

The apicomplexan parasite Plasmodium, which is accountable for malaria, has a complex life cycle that includes both vertebrate hosts and invertebrate mosquitoes. Adaptation to blood-feeding in mosquitoes has made it inadvertently a carrier of various diseases. A blood meal is indispensable for adult female mosquitoes to nourish its egg, and maintain the gonotrophic cycle. But during blood feeding, ingestion of Plasmodium gametocyte from an infected person’s blood results in the onset of 18-20 days long sporogonic cycle that culminates in the production of infectious sporozoites in the mosquito host [1]. These infectious sporozoites are then delivered into the human body through salivary discharge, which initiates the intricate stages of the asexual process causing malaria. In humans, malaria is caused by five Plasmodium species i.e., Plasmodium falciparum, P. vivax, P. malariae, P. ovale, and P. knowlesi [2].

P. falciparum and P. vivax, vectored by the adult female Anopheline mosquitoes, are two principal parasites of human malaria [3]. Of the five Plasmodium species that cause human malaria, Plasmodium vivax is the most geographically widespread [4]. The parasite could survive quiescent for extended periods when circumstances are not conducive to its ongoing transmission [5]. According to the current report by WHO, in the year 2019 around 75% of malaria cases were caused by P. vivax in the WHO Region of the Americas. An approximated 52% of the global burden of P. vivax emerged from the WHO South-East Asia Region, among which 47% were contributed from India [6].

P. vivax is considered as a less fatal parasite, but the recent emergence of more P. vivax infected cases in P. falciparum endemic areas, and increased mortality, morbidity rates are drawing our attention to this least studied parasite. It is more difficult to monitor and eradicate the P. vivax than P. falciparum, because of limited information, and associated biological complexities of its development in the mosquito as well as the human host [7, 8].

P. vivax normally circulates at low peripheral parasite densities, but still, they are transmissible by the mosquito vectors, and hence presents major challenges for the diagnosis of infected peoples. P. vivax has adapted to live with varying Anopheles vectors in different ecological conditions. Unlike other Plasmodium species, P. vivax has the potential to form dormant hypnozoites inside the host liver, and these liver-stage parasites are accountable for malaria relapses for weeks or months after initial infection [5]. Lastly, the lack of long-term in-vitro culture further restricts our understanding of the biological consequences of P. vivax development and transmission [9]. Nevertheless, for the last two decades, the integration and utilization of high-throughput molecular technologies such as genomics, RNA-Seq/transcriptomics, proteomics, have been valuable to decode and trace the genetic variation and diversity in the P. vivax population collected from different geographical origins [10, 11]. Efforts are continuing to uncover molecular and functional correlation of tissue/stage-specific P. vivax biology in the vertebrate host, identify genetic signatures to develop new diagnosis tools, anti-P. vivax drugs, or vaccine development. However, the biological complexity of the P. vivax development cycle in the mosquito vector-host is too limited, and therefore in this article, we highlight the current progress made so far in the understanding of the Mosquito-P. vivax interaction biology.

Advertisement

2. A general overview of the sporogonic cycle in mosquito host

The transmission of the parasite from human host to mosquito transpire when a female mosquito acquires gametocyte containing blood meal from the infected vertebrate host. When the parasite enters the midgut lumen it faces the dynamically changing environment, where both male and female gametocytes get differentiated into male and female gametes [12, 13, 14, 15]. Ingested gametocytes also encounter proteolytic enzymes released by midgut epithelium in the midgut environment to digest the blood meal, which may have an agonistic or antagonistic effect on parasite growth. Fertilization of male and female gametes results in zygote formation, which rapidly transforms into motile ookinetes [16]. After exiting from the blood bolus, ookinete traverses the midgut epithelium either through intracellular or intercellular route and then rests beneath the epithelial cell at basal lamina. Later ookinetes transform into replicative oocyst stage which undergoes an umpteen round of nuclear division to produce thousands of sporozoites within a time period of one to two weeks. Once in the hemolymph circulation, the free circulatory sporozoites (fcSPZ) target to invade salivary glands, but most of them are rapidly cleared off by hemocytes, the immune blood cells of the mosquitoes [17]. Thus tracking of molecular, biochemical, and cellular events during Plasmodium developmental transition from one stage to another stage, is of particular interest. Several laboratory studies on mosquito-parasite interaction involving P. berghei or P. falciparum, demonstrate that the developmental kinetics of the Plasmodium population is significantly altered, though the mechanism is not fully understood [18, 19, 20]. The last two decades of research highlights the crucial role of the tissue-specific mosquito immune system to control the parasite load, though the physiological relevance is yet to be investigated [21, 22, 23, 24].

Advertisement

3. Plasmodium population dynamics and their immune regulation in the mosquito host

During Plasmodium development inside the mosquito host, the parasite population undergoes various bottlenecks. Previous investigations demonstrated that if a female mosquito takes ~1000 gametocytes through its infected blood meal, ~100 can be transformed into ookinetes, and among them, only 1–5 can successfully form oocysts. Furthermore, these survived oocysts will form millions of sporozoites, but only 19-20% can successfully invade the salivary glands for further transmission [25]. In refractory strains, not a single ookinete could transform into oocysts [26]. In general, a substantial loss of parasite population occurs at each developmental stage of the parasite, and this major parasite loss can be attributed to both human as well as mosquito components, which are harmful to Plasmodium.

The human component includes cytokines, complement protein, and reactive nitrogen species that are ingested along with the gametocytes during blood meal intake, and detrimental to the parasite within the midgut lumen of vector [26]. During the parasite transition through midgut epithelium, the mosquito mounts early immune response by increasing midgut nitration and activation of the signaling pathway. The nitration process modifies the ookinetes surface, and mark them to be recognized by the mosquito complement system when they emerge toward the basal side of the midgut [27]. Signaling pathways provide varying responses to various species of Plasmodium, such as the IMD pathway acts more efficiently against P. falciparum than P. berghei, and the Toll pathway is more responsive against P. berghei, and P. gallinaceum [28]. The proliferation of microbiota following blood meal also exacerbated the mosquito immune response, which in turn is detrimental to parasite development. Plasmodium parasite faces population bottlenecks throughout their development (in vertebrate as well as invertebrate host) but the mosquito midgut serves as the major site of extermination, where the number of parasites is minimal during the oocyst stage which makes it the most susceptible stage to identify molecular targets to disrupt the transmission [26]. Parallel to gut-immune interaction, several factors have been identified from mosquito hemocyte and salivary glands that interact with Plasmodium sporozoites; a bulk of literature is available on the mosquito innate immune system against P. berghei and P. falciparum, and therefore readers may refer to many excellent reviews [29, 30, 31]. Here we update the reports on the Mosquito-P. vivax interactions, and highlight their relevance for future implications.

Advertisement

4. Mosquito-P. vivax interaction

Undoubtedly, advanced omics technologies, especially genome sequencing and transcriptome analysis, has now become a basic method in living organisms for the assessment of genome-scale gene identification. The expression of large scale identified genes is currently being explored to decode the molecular complexity of P. vivax development in the vertebrate host. Earlier, a high-density tiling microarray-based study showed the gene expression variation of P. vivax from human and mosquito stages such as sporozoites, gametes, zygotes, ookinetes, and in-vivo asexual blood stages. Their comparison to P. falciparum and P. yoelii further reveals conserved and species-specific patterns highlighting the metabolic state of parasites growing within humans and identifies many orthologs of P. falciparum transcripts that are needed for exoerythrocytic development, which may also likely help in hypnozoite formation in the P. vivax [32].

4.1 Plasmodium vivax strategy to adapt in the mosquito Anopheles stephensi

The successful development of P. vivax within the midgut of a susceptible strain of Anopheles stephensi can be divided into two phases: pre-invasion (within midgut lumen) and post-invasion strategy i.e. development of oocyst stage which depends upon the nutrient availability within the host. During the pre-midgut invasion phase, P. vivax imparts an intricate mechanism to evade the mosquito immune response. It indirectly attenuates the mosquito immune response by dramatically suppressing the bacterial population, and whereas in the post- midgut invasion phase i.e. during the development of oocyst it modulates the expression of genes that are directly or indirectly involved with nutrition physiology to fulfill their nutritional requirement. We have limited information about the phases beyond oocysts maturation and their strategies to evade the mosquito immune system and promote their transmission.

4.1.1 Pre- invasion strategy of P. vivax

The midgut of Anopheles mosquitoes is housed by a complex and diverse community of bacteria, protozoa, fungi, etc. collectively referred to as the microbiota, and this microbiota is believed to shape the vector competency of mosquito. The gut bacteria of Anopheles mosquitoes adversely affect the Plasmodium infection [33, 34]. These tripartite interactions have been studied between the mosquito, its microbiota, and the Plasmodium parasites, but the precise relationship between the three remains unknown.

Numerous research reports have revealed that microbiota of specific bacterial species, particularly gram-negative bacteria, in many Anopheles species have an inhibitory effect on various Plasmodium species. The elimination of midgut bacteria through antibiotic treatment enhances oocyst load and parasite prevalence in different species of Anopheles. There are two mechanisms by which microbiota interfere with the Plasmodium development in the midgut lumen:-(i) indirectly by triggering the immune response of the mosquito (Imd Pathway) that guides the synthesis of AMP and other immune effectors that interferes with the development of parasites, and (ii) directly by certain bacterial species producing the metabolites that interfere with Plasmodium development and survival [33]. Recently, we have demonstrated that P. vivax plays a unique strategy to steer clear off the mosquito immune response during its pre-invasive phase, by dramatic suppression of the gut-bacterial population [35] (Figure 1). This study hypothesizes that the parasites outcompete the midgut microbiota presumably by scavenging the iron from the blood meal which is necessary for bacterial growth [35].

Figure 1.

Alteration of midgut microbiota proliferation by P. vivax. Blood meal induces midgut microbiota proliferation within 24 hours. But during P. vivax infection, somehow this parasite restricts this microbiota proliferation after blood-meal, to avoid nutritional competition and immune defense exerted by the microbiota. This smart strategy of restriction helps the parasite to survive and proliferate better.

4.1.2 Post- invasion strategy of P. vivax (development of oocyst)

During the Plasmodium transit through midgut epithelium within the susceptible strain of Anopheles, some of the ookinetes successfully manage to escape the mosquito immune response [36], and reach the basal lamina of midgut to further differentiate into oocyst, and rests there nearly for two weeks. The sessile oocyst stage is metabolically active, and follows an umpteen rounds of the nuclear division to transform into sporozoites. A single oocyst is capable of producing thousands of haploid sporozoites [37, 38]. Limited research has been undertaken on the underlying mechanism of P. vivax oocyst development (transition from a small oocyst of 7-8 μm to a large oocyst of 35-40 μm) in the mosquitoes. A few recent RNA-Seq analyses of P. vivax infected mosquitoes have been valuable to understand the ookinete and oocyst stage of P. vivax which reveals the alteration of several transcripts in the gut after 18 hours and 7 days post-infection in mosquito Anopheles dirus [39]. Notably, the authors identified several genes such as Anoctamin 6 (ANO6; ADIR005670) and Fibroblast Growth Factor (FGF; ADIR008464), which may likely have immune regulation of P. vivax growth in the gut of the mosquito.

The parasite scavenges the nutrients from the host, and thus one of the main deciding factors of the infection outcome is likely dependent on the availability of nutritional resources of the host [40]. Our ongoing tissue-specific RNA-Seq analysis of An. stephensi infected with P. vivax oocyst identifies several unique sets of transcripts/genes, which have not yet find associated with any other Plasmodium infection. This study revealed the expression of genes involved in maintaining glucose homeostasis (Trehalase), nutrient transport (Sterol Carrier protein), energy, and nutrient homeostasis (Folliculin) during P. vivax infection [24]. We noticed that P. vivax infection modulates the Trehalase and Sterol Carrier protein expression in the midgut and salivary gland (SCP) for its own development and maturation. Trehalase, a glucosidase enzyme, catalyze the hydrolysis of disaccharide trehalose sugar into glucose units. Glucose is the main source of energy for the extensive proliferation of malarial parasites during both the blood and liver stages of malaria infection [41, 42, 43, 44]. Plasmodium obtains the host glucose via hexose transporter. However, the role of sugar metabolism on Plasmodium infection in the mosquito vector remains poorly known. A multifold enriched expression of Trehalase transcript during early to late-stage oocysts in the gut as well as salivary glands, in addition to retrieval of Plasmodium hexose transcript in the midgut during oocyst stage, suggests that Trehalase may significantly contribute to hydrolyze the trehalose to provide glucose for the rapid proliferation of parasites, and also affect the reproductive capacity of adult female mosquito An. stephensi [45].

Similar to sugar requirement, Plasmodium also relies heavily on the host’s cholesterol for its growth when maturing from small oocysts to large oocysts in the gut. Since Plasmodium is incapable to synthesize de-novo cholesterol [46], and P. vivax infection induces a multifold expression of SCP after seven days of infection in the gut, likely indicates its role in cholesterol transport. Currently, there is no functional correlation exists between SCP and Plasmodium infection, however, with the current observation of SCP enrichment in the midgut as well as salivary gland, we propose that besides a possible role of supplying cholesterol to developing oocyst, it is possible that As-SCP may impart an anti-Plasmodium immune response, as increased lipid droplets have been shown in the midgut of Ae. aegypti during bacterial and viral infection [47]. Folliculin (FLCN) is a tumor suppressor protein associated with Birt-Hogg-Dube(BHD) syndrome [48, 49]. It is involved in many biological processes including vesicular trafficking, energy, and nutrient homeostasis, and monitors E-cadherin protein level [50, 51]. Late induction of FLCN in response to P. vivax infection (unpublished) suggests that it might also play an important role in maintaining the integrity of midgut epithelial cells during oocyst bursting or acquisition of nutrients by developing oocyst, though further studies needed to support this hypothesis.

4.2 P. vivax infection and immune strategy of the Anopheles stephensi

As described earlier, the parasite population undergoes several bottlenecks throughout their development inside the mosquito host. These bottlenecks are achieved because of the mosquito immune system [26]. Once the Plasmodium parasite transforms in the ookinete, midgut nitration modifies the parasite surface, which is then recognized by the hemocyte encoded pattern recognition receptors (like TEP1) circulating in the hemolymph [52]. Studies in the mosquito An. gambiae suggest that the complex of LRIM1/APL1C and TEP1 bind to the parasite surface and activate the complement system, and in turn, the circulating hemocytes kill the parasite through cell lysis, phagocytosis, melanization, etc [53, 54, 55]. This whole phase is completed within 24 hours after infective blood meal uptake and is known as the “early phase” immune response (Figure 2). Once the ookinetes reach the midgut epithelium, they get transformed into oocysts, and the immune system working against these transformed parasites is known as “late phase” immune response [56, 57]. Although very little is known about this phase but recent literature suggests that LL3 mediated hemocyte differentiation, and STAT pathway activation, together helps in the restriction of the oocysts development [58]. Post oocysts maturation, millions of sporozoites evade the midgut lamina and circulate in the hemolymph, in order to reach and invade salivary glands for their successful transmission. Current literature suggests that among thousands of sporozoites only 19% can successfully invade the salivary gland, the rest are eliminated by the hemocyte mediated mosquito immune system [25]. But we have very limited information about this direct cell (hemocytes)-cell (free circulating sporozoites) interaction and elimination mechanism [29].

Figure 2.

Systematic representation of events occurring during early and late phase immunity in malaria parasite-infected mosquito: Once the ookinete invade the midgut epithelium, PRRs (pattern recognition receptors) like TEP1 recognize the pathogen and activate the complement system, which further triggers the hemocytes for phagocytosis, melanization, etc.

Altogether this information is restricted to the model organisms, and due to problems in culturing of P. vivax and extraction of hemocytes the exact species-specific interaction biology of this neglected parasite is still unknown [29]. As hemocytes play a crucial role in immune regulation, decoding the direct or indirect immune interactions between hemocytes and P. vivax parasite, will help us to figure out the parasite population control strategies of the mosquito hosts.

4.2.1 Hemocytes: the cellular immune army of the mosquito host

Mosquitoes have an open circulatory system, and hemocytes are the tiny blood cells circulating across the body reaching every mosquito tissue. These are the major immune elicitors working against a diverse range of pathogens [29]. Hemocytes are the core of the mosquito immune system which can induce both cellular as well as humoral immune responses [30, 59, 60]. Mosquito hemocytes population can be discriminated on the basis of their anatomical location (circulatory and sessile), DNA content (euploid and polyploid), morphology, and functions (granulocytes, oenocytoids, and prohemocytes) [61, 62, 63]. Granulocytes are the phagocytic cells, which engulf the invaded parasite and kill them by lysozyme activity [64, 65]. Oenocytoids are the producers of the Pro-phenoloxidases, the rate-limiting enzyme of the melanization pathway [66]. Melanization is the systematic enzymatic process, which ultimately produces the melanin protein. When a foreign invader infects the mosquito, hemocytes cover the parasite in the melanin envelop, which will cut-off the parasite from the outside environment, nutrition, and also induces oxidative stress which results in the killing of the parasite. Prohemocytes are considered as the progenitor cells, which produce granulocytes and oenocytoids, although the actual function is not known yet about these tiny cells [64, 67]. Previous literature illustrated various hemocyte encoded molecules, like TEP1, FBN30, LRR3, etc. are vital for the early and late phase immune responses [55, 68, 69, 70, 71]. Researchers have also successfully tracked the involvement of phagocytosis and melanization events for the removal of parasites [17]. But we do not have much information about the direct cell–cell interaction of the hemocytes and P. vivax free-circulating sporozoites (fcSPZ).

Recently we conducted a transcriptome based study, to understand that how hemocytes control the P. vivax free circulatory sporozoites (fcSPZ) population before salivary invasion [24]. Here we found that hemocyte encoded transcripts undergo a major shift during P. vivax infection. A detailed comparison of the P. vivax infected and uninfected hemocyte transcriptomes revealed that transcripts of organelle organization and riboprotein complex biogenesis have exclusively emerged during P. vivax fcSPZ infection. Altogether these findings suggested that the hemocyte population undergoes dynamic changes i.e., differentiate and increase the population in response to the fcSPZ. Through the immune database comparison, we found that AMPs like Defensin and Gambicin were exclusively induced when fcSPZ were circulating in the hemolymph. These findings were further validated by the real-time based experiments and depicted that Defensin3 and Gambicin may likely play a crucial role during P. vivax late-phase immune function against fcSPZ infection. Hence, conclusively current findings illustrate that hemocytes rapidly proliferate and impart humoral immune responses against the parasite to limit the fcSPZ population before salivary gland invasion (Figure 3).

Figure 3.

Direct interaction of free-circulatory sporozoites and hemocytes. Post oocyst maturation, sporozoites circulate freely in the hemolymph, in order to reach salivary glands for further transmission. But oocyst rupture triggers the mosquito immune system and activates the hemocyte proliferation, which leads to the sporozoite clearance by phagocytosis, melanization, and AMP (Defensin and Gambicin) production. Although the role of AMPs in hemocyte activation is still unclear.

Apart from global transcriptomic changes undergone by the hemocyte population to manage P. vivax infection, we also found the species-specific molecular differences among the hemocyte encoded immune transcripts. FBN9 which was previously considered as the potent anti-Plasmodium molecule and showed multifold upregulation during P. berghei/ P. falciparum infection [71, 72] was found to be downregulated during P. vivax infection. Novel molecules like FREP12 and FREP50 were predicted to be involved in the clearance of P. vivax sporozoites. Furthermore, storage proteins like ApolipophorinIII, Hexamerin were also found to be highly induced during P. vivax oocysts development, which further supported the previous evidence of host nutrient scavenging by the maturing oocysts [73, 74, 75].

4.2.2 Mosquito salivary glands: Gatekeeper of entry and exit for the parasite

The salivary glands are the crucial organ for the development and transmission of the Plasmodium to a vertebrate host. Salivary glands are paired epithelial organs that are located in the thorax, and consist of three lobes namely, two lateral and one median, where each lateral lobe is comprised of proximal and distal lobes [76, 77]. The proximal portion of the female glands produces enzymes involved in sugar metabolism, where distal lobes are shown to play roles in blood meal acquisition, Plasmodium invasion, and transmission. Although, studies suggest that only 10–20% hemolymph circulating sporozoites, manage to invade the salivary glands, however, the mechanism of this drastic reduction of 80–90% sporozoites is poorly known [25, 78]. Accumulating evidence highlights that sporozoite invasion into the glands is mediated by salivary specific receptor-ligand interactions [18, 79].

The sporozoites must leave (egress) the oocyst after maturation to invade the salivary gland and to be transmitted to the next vertebrate host. The egress of sporozoite is mediated by a protease named Cysteine protease (ECP1) which ruptures the oocyst [80, 81]. The sporozoites are released into the hemolymph and carried to the salivary glands by the circulation of hemolymph in an anterior direction from the abdomen to head, and facilitate the sporozoite invasion to the salivary gland [82]. The salivary gland epithelium forms a physical barrier that pathogens must cross, and Plasmodium parasites are evolved with unique proteins that drive invasion by first binding to the salivary gland specific surface receptors [83]. The salivary invasion process completion occurs in two stages, where first, sporozoite binds to invade the salivary gland basal lamina; and second, then interacts with the plasma membrane of the epithelial cells favoring sporozoite internalization. During the invasion, sporozoites attach and invade the distal and medial lobe of the salivary glands, and this attachment and invasion are highly specific to the nature of Plasmodium species [84, 85].

Empirical evidence showing that the salivary glands serve as an active immune organ is largely lacking, except some studies highlighting that a Serine Protease Inhibitor (SRPN6) produced in the salivary epithelium limits gland invasion by Plasmodium sporozoites, and thus SRPN6 serves as an important salivary invasion immune-marker [86]. Several putative salivary encoded factors such as Saglin, CSP binding proteins which effectively binds with sporozoites surface antigens such as TRAP, CSP are well known salivary receptors for sporozoites invasion [87, 88, 89, 90, 91]. However, several other salivary factors such as Plasmodium Responsive Salivary1 (PRS1), ESP, Peptide-O-xylosyl Transferase 1 (OXT1) have also been identified to play a crucial role in parasite invasion of both midgut and salivary glands [92, 93, 94, 95]. Once inside the salivary glands, the parasite undergoes transcriptional reprogramming before its transmission to the next mammalian host.

Transmission of many viral and protozoan parasites to a vertebrate host requires their salivary injection with the mosquito saliva during blood-feeding, and thus the migration of sporozoites needs duct for the continuation of the life cycle. Mosquito saliva has a pleiotropic property such as anti-hemostatic, vasodilator, or anti-inflammatory properties and immune modulators, and basic function to facilitate blood-feeding [96, 97, 98]. However, saliva proteins can also have an indirect impact on pathogen development and transmission. For example, a recent study in mosquito An. gambiae shows that mosquito saliva proteins such as AgTRIO and mosGILT serve as an important mediator of the transmission of P. falciparum, and inhibition of this protein can reduce the parasite burden in the human host [99, 100]. Although, a major study on salivary-sporozoites interaction is restricted to P. berghei and/or P. falciparum, however, very limited information is available on the salivary-P. vivax interaction.

A comparative RNA-Seq analysis of uninfected and P. vivax-infected mosquito salivary glands suggests that salivary transcripts undergo substantial changes during P. vivax infection. The maturation of sporozoite seems to coincide with the change in gene expression essential for invasion and transmission. Findings of several classes of immune proteins such as Heme-peroxidase, FADD, Gambicin, GNBP, and multiple family proteins of Serine proteases, and SCRC in the P. vivax sporozoites invaded salivary glands highlighted their anti-Plasmodium immune role of salivary glands. The transcriptome of the infected salivary glands also revealed that P. vivax infection decreased the expression of apyrase significantly which suggests that P. vivax interferes with salivary secretion before probing and feeding to ensure their delivery into the next human host. These findings offer valuable new insights into the biology of malaria parasites. Manipulating tissue-specific immuno-physiology of the mosquitoes may halt the Plasmodium vivax development and hence the transmission (Figure 4).

Figure 4.

Proposed hypothesis of salivary gland-Plasmodium interaction and transmission: Free circulating sporozoite in the hemolymph recognize and attach to the basal lamina of the salivary gland receptor-ligand interaction;(1) initial attachment of the sporozoite mediated by interactions of carbohydrate residues on the basal lamina with a parasite CS, SGS1, MABEL. CSP binding protein and Saglin bind with CSP and TRAP respectively are an important component of salivary gland invasion. (2) Sporozoite internalization: After invasion sporozoite passes into the secretory cavity and sporozoites begin to assembly there as a large bundle form. Within the salivary duct component of the mosquito immune responses Gambicin, Cecropin, GNBP, and SCR family members presumably act upon sporozoite and limit the number.

Advertisement

5. Conclusion

Plasmodium and mosquito host both are involved in the dynamic molecular relationship, where parasite tries to dodge the host immune system and utilize its nutrients for their successful proliferation/ transmission. On the contrary, the mosquito host immune system tries to restrict the parasite development and eliminate the remnants. During this ultimate battle, some host species defeat the parasite through its active immune system and become resistant but in others, the parasite smartly manipulates the host system and defends itself for successful transmission.

P. vivax is one of the neglected parasites which successfully manipulated the host system for its efficient transmission. P. vivax suppresses the microbiota proliferation to avoid nutritional competition as well as early immune responses. Different nutrient transport proteins like Trehalase, Sterol Carrier, ApolipophorinIII, etc. were modulated by the parasite for fulfilling its nutritional requirements. But still, mosquito hosts also developed species-specific immune effector molecules like FREP50, FREP12, LRIM17, etc. to block the parasite development. Likewise finding salivary-specific factors such as Heme-peroxidase, SP24D that are crucial to sporozoite invasion and survival, may further help to halt the progression of Plasmodium development and malaria transmission.

In summary, future functional exploration of the novel P. vivax specific host factors, will help in the development of transmission-blocking vaccines and the generation of new intervention techniques or modify current ones.

Advertisement

Acknowledgments

CC, ST, SK, RKD conceptualize the idea and drafted MS. CC, ST, SK, PS, JR and RKD reviewed, edited, corrected and revised the MS. We would like to thank ICMR-NIMR and funding agencies, CSIR/UGC/DST for infrastructural and financial support to conduct the research. CC, ST, and SK are recipients of DST (DST/INSPIRE/03/2014/003463), UGC (22/12/2013(II)EU-V), and CSIR (09/905(0015)/2015-EMR-1), fellowships, respectively.

Advertisement

Conflict of interest

No competing interests were disclosed.

References

  1. 1. Antinori, S., Galimberti, L., Milazzo, L. & Corbellino, M. Biology of human malaria plasmodia including Plasmodium knowlesi. Mediterranean Journal of Hematology and Infectious Diseases (2012). doi:10.4084/MJHID.2012.013
  2. 2. Singh, B. & Daneshvar, C. Human infections and detection of Plasmodium knowlesi. Clinical Microbiology Reviews (2013). doi:10.1128/CMR.00079-12
  3. 3. White, M. T. et al. Plasmodium vivax and Plasmodium falciparum infection dynamics: Re-infections, recrudescences and relapses. Malar. J. (2018). doi:10.1186/s12936-018-2318-1
  4. 4. Howes, R. E. et al. Global epidemiology of Plasmodium vivax. American Journal of Tropical Medicine and Hygiene (2016). doi:10.4269/ajtmh.16-0141
  5. 5. White, N. J. Determinants of relapse periodicity in Plasmodium vivax malaria. Malaria Journal (2011). doi:10.1186/1475-2875-10-297
  6. 6. Global Malaria Programme: WHO Global. World malaria report 2019. WHO Regional Office for Africa (2019)
  7. 7. Mueller, I. et al. Key gaps in the knowledge of Plasmodium vivax, a neglected human malaria parasite. The Lancet Infectious Diseases (2009). doi:10.1016/S1473-3099(09)70177-X
  8. 8. Mendis, K., Sina, B. J., Marchesini, P. & Carter, R. The neglected burden of Plasmodium vivax malaria. in American Journal of Tropical Medicine and Hygiene (2001). doi:10.4269/ajtmh.2001.64.97
  9. 9. Bermúdez, M., Moreno-Pérez, D. A., Arévalo-Pinzón, G., Curtidor, H. & Patarroyo, M. A. Plasmodium vivax in vitro continuous culture: The spoke in the wheel. Malaria Journal (2018). doi:10.1186/s12936-018-2456-5
  10. 10. Garrido-Cardenas, J. A., González-Cerón, L., Manzano-Agugliaro, F. & Mesa-Valle, C. Plasmodium genomics: an approach for learning about and ending human malaria. Parasitol. Res. (2019). doi:10.1007/s00436-018-6127-9
  11. 11. Bourgard, C., Albrecht, L., Kayano, A. C. A. V., Sunnerhagen, P. & Costa, F. T. M. Plasmodium vivax biology: Insights provided by genomics, transcriptomics and proteomics. Frontiers in Cellular and Infection Microbiology (2018). doi:10.3389/fcimb.2018.00034
  12. 12. Aly, A. S. I., Vaughan, A. M. & Kappe, S. H. I. Malaria Parasite Development in the Mosquito and Infection of the Mammalian Host. Annu. Rev. Microbiol. (2009). doi:10.1146/annurev.micro.091208.073403
  13. 13. Tuteja, R. Malaria - An overview. FEBS Journal (2007). doi:10.1111/j.1742-4658.2007.05997.x
  14. 14. Wiser, M. Plasmodium Life Cycle. Tulane Univ.(5) 1-4. (2009)
  15. 15. Delves, M. J. et al. Male and female Plasmodium falciparum mature gametocytes show different responses to antimalarial drugs. Antimicrob. Agents Chemother. (2013). doi:10.1128/AAC.00325-13
  16. 16. Bennink, S., Kiesow, M. J. & Pradel, G. The development of malaria parasites in the mosquito midgut. Cellular Microbiology (2016). doi:10.1111/cmi.12604
  17. 17. Hillyer, J. F., Schmidt, S. L. & Christensen, B. M. Rapid phagocytosis and melanization of bacteria and Plasmodium sporozoites by hemocytes of the mosquito Aedes aegypti. J. Parasitol. (2003). doi:10.1645/0022-3395(2003)089[0062:rpamob]2.0.co;2
  18. 18. Mueller, A. K., Kohlhepp, F., Hammerschmidt, C. & Michel, K. Invasion of mosquito salivary glands by malaria parasites: Prerequisites and defense strategies. International Journal for Parasitology (2010). doi:10.1016/j.ijpara.2010.05.005
  19. 19. Volohonsky, G. et al. Transgenic Expression of the Anti-parasitic Factor TEP1 in the Malaria Mosquito Anopheles gambiae. PLoS Pathog. (2017). doi:10.1371/journal.ppat.1006113
  20. 20. Garcia, J. E., Puentes, A. & Patarroyo, M. E. Developmental biology of sporozoite-host interactions in Plasmodium falciparum malaria: Implications for vaccine design. Clinical Microbiology Reviews (2006). doi:10.1128/CMR.00063-05
  21. 21. Smith, R. C., Barillas-Mury, C. & Jacobs-Lorena, M. Hemocyte differentiation mediates the mosquito late-phase immune response against Plasmodium in Anopheles gambiae. Proc. Natl. Acad. Sci.112, E3412–E3420 (2015)
  22. 22. Dixit, R. et al. Salivary gland transcriptome analysis during Plasmodium infection in malaria vector Anopheles stephensi. Int. J. Infect. Dis. (2009). doi:10.1016/j.ijid.2008.07.027
  23. 23. Qayum, A. A. & Telang, A. A protocol for collecting and staining hemocytes from the yellow fever mosquito Aedes aegypti. J. Vis. Exp. 2-5 (2011). doi:10.3791/2772
  24. 24. Kumari S, Chauhan C, Tevatiya S, et al (2021) Current Research in Immunology Genetics changes of Plasmodium vivax tempers host tissue-specific responses in Anopheles stephensi. Curr Res Immunol 2:12-22. https://doi.org/10.1016/j.crimmu.2021.02.002
  25. 25. Hillyer, J. F., Barreau, C. & Vernick, K. D. Efficiency of salivary gland invasion by malaria sporozoites is controlled by rapid sporozoite destruction in the mosquito haemocoel. Int. J. Parasitol.37, 673-681 (2007)
  26. 26. Smith, R. C., Vega-Rodríguez, J. & Jacobs-Lorena, M. The Plasmodium bottleneck: Malaria parasite losses in the mosquito vector. Mem. Inst. Oswaldo Cruz109, 644-661 (2014)
  27. 27. Oliveira, G. D. A., Lieberman, J. & Barillas-Mury, C. Epithelial nitration by a peroxidase/NOX5 system mediates mosquito antiplasmodial immunity. Science (80). (2012). doi:10.1126/science.1209678
  28. 28. Garver, L. S., Dong, Y. & Dimopoulos, G. Caspar controls resistance to Plasmodium falciparum in diverse anopheline species. PLoS Pathog. (2009). doi:10.1371/journal.ppat.1000335
  29. 29. Hillyer, J. F. & Strand, M. R. Mosquito hemocyte-mediated immune responses. Curr. Opin. Insect Sci.3, 14-21 (2014)
  30. 30. Strand, M. R. The insect cellular immune response. Insect Science (2008). doi:10.1111/j.1744-7917.2008.00183.x
  31. 31. Peterson, T. M. L., Gow, A. J. & Luckhart, S. Nitric oxide metabolites induced in Anopheles stephensi control malaria parasite infection. Free Radic. Biol. Med. (2007). doi:10.1016/j.freeradbiomed.2006.10.037
  32. 32. Westenberger, S. J. et al. A systems-based analysis of Plasmodium vivax lifecycle transcription from human to mosquito. PLoS Negl. Trop. Dis. (2010). doi:10.1371/journal.pntd.0000653
  33. 33. Romoli, O. & Gendrin, M. The tripartite interactions between the mosquito, its microbiota and Plasmodium. Parasites and Vectors11, 1-8 (2018)
  34. 34. Mukherjee, D., Chora, Â. F. & Mota, M. M. Microbiota, a Third Player in the Host–Plasmodium Affair. Trends in Parasitology (2020). doi:10.1016/j.pt.2019.11.001
  35. 35. Sharma, P. et al. Altered Gut Microbiota and Immunity Defines Plasmodium vivax Survival in Anopheles stephensi. Front. Immunol. (2020). doi:10.3389/fimmu.2020.00609
  36. 36. Molina-Cruz, A. et al. Plasmodium falciparum evades immunity of anopheline mosquitoes by interacting with a Pfs47 midgut receptor. Proc. Natl. Acad. Sci. U. S. A. (2020). doi:10.1073/pnas.1917042117
  37. 37. Volohonsky, G. et al. Kinetics of Plasmodium midgut invasion in Anopheles mosquitoes. PLoS Pathog.16, 1-19 (2020)
  38. 38. Smith, R. C. & Barillas-Mury, C. Plasmodium Oocysts: Overlooked Targets of Mosquito Immunity. Trends Parasitol.32, 979-990 (2016)
  39. 39. Boonkaew, T. et al. Transcriptome analysis of Anopheles dirus and Plasmodium vivax at ookinete and oocyst stages. Acta Trop. (2020). doi:10.1016/j.actatropica.2020.105502
  40. 40. Zuzarte-Luís, V. & Mota, M. M. Parasite Sensing of Host Nutrients and Environmental Cues. Cell Host and Microbe (2018). doi:10.1016/j.chom.2018.05.018
  41. 41. Meireles, P. et al. GLUT1-mediated glucose uptake plays a crucial role during Plasmodium hepatic infection. Cell. Microbiol. (2017). doi:10.1111/cmi.12646
  42. 42. Olszewski, K. L. & Llinás, M. Central carbon metabolism of Plasmodium parasites. Molecular and Biochemical Parasitology (2011). doi:10.1016/j.molbiopara.2010.09.001
  43. 43. Stanway, R. R. et al. Genome-Scale Identification of Essential Metabolic Processes for Targeting the Plasmodium Liver Stage. Cell (2019). doi:10.1016/j.cell.2019.10.030
  44. 44. Wang, A. et al. Glucose metabolism mediates disease tolerance in cerebral malaria. Proc. Natl. Acad. Sci. U. S. A. (2018). doi:10.1073/pnas.1806376115
  45. 45. Tevatiya, S. et al. Molecular and Functional Characterization of Trehalase in the Mosquito Anopheles stephensi. Front. Physiol. (2020). doi:10.3389/fphys.2020.575718
  46. 46. Labaied, M. et al. Plasmodium salvages cholesterol internalized by LDL and synthesized de novo in the liver. Cell. Microbiol. (2011). doi:10.1111/j.1462-5822.2010.01555.x
  47. 47. Barletta, A. B. F. et al. Emerging role of lipid droplets in Aedes aegypti immune response against bacteria and Dengue virus. Sci. Rep. (2016). doi:10.1038/srep19928
  48. 48. Zong, D., Li, J., Liu, X., Guo, T. & Ouyang, R. Identification of a Novel Pathogenic Folliculin Variant in a Chinese Family With Birt–Hogg–Dubé Syndrome (Hornstein-Knickenberg Syndrome). Front. Genet. (2020). doi:10.3389/fgene.2020.565566
  49. 49. Nickerson, M. L. et al. Mutations in a novel gene lead to kidney tumors, lung wall defects, and benign tumors of the hair follicle in patients with the Birt-Hogg-Dubé syndrome. Cancer Cell (2002). doi:10.1016/S1535-6108(02)00104-6
  50. 50. Schmidt, L. S. & Linehan, W. M. FLCN: The causative gene for Birt-Hogg-Dubé syndrome. Gene (2018). doi:10.1016/j.gene.2017.09.044
  51. 51. Yang, T. et al. Folliculin Controls the Intracellular Survival and Trans-Epithelial Passage of Neisseria gonorrhoeae. Front. Cell. Infect. Microbiol. (2020). doi:10.3389/fcimb.2020.00422
  52. 52. Jaramillo-Gutierrez, G. et al. Mosquito immune responses and compatibility between Plasmodium parasites and anopheline mosquitoes. BMC Microbiol. (2009). doi:10.1186/1471-2180-9-154
  53. 53. Baxter, R. H. G. et al. A heterodimeric complex of the LRR proteins LRIM1 and APL1C regulates complement-like immunity in Anopheles gambiae. Proc. Natl. Acad. Sci. U. S. A. (2010). doi:10.1073/pnas.1010575107
  54. 54. Ruiz, V. M. R. et al. Stimulation of a protease targeting the LRIM1/APL1C complex reveals specificity in complement-like pathway activation in Anopheles gambiae. PLoS One14, 1-14 (2019)
  55. 55. Fraiture, M. et al. Two Mosquito LRR Proteins Function as Complement Control Factors in the TEP1-Mediated Killing of Plasmodium. Cell Host Microbe (2009). doi:10.1016/j.chom.2009.01.005
  56. 56. Kwon, H., Arends, B. R. & Smith, R. C. Late-phase immune responses limiting oocyst survival are independent of TEP1 function yet display strain specific differences in Anopheles gambiae. Parasites and Vectors (2017). doi:10.1186/s13071-017-2308-0
  57. 57. Subbarao, S. K., Nanda, N., Rahi, M. & Raghavendra, K. Biology and bionomics of malaria vectors in India: Existing information and what more needs to be known for strategizing elimination of malaria. Malaria Journal (2019). doi:10.1186/s12936-019-3011-8
  58. 58. Gupta, L. et al. The STAT Pathway Mediates Late-Phase Immunity against Plasmodium in the Mosquito Anopheles gambiae. Cell Host Microbe5, 498-507 (2009)
  59. 59. Hillyer, J. F. Insect immunology and hematopoiesis. Dev. Comp. Immunol.58, 102-118 (2016)
  60. 60. De, T. Das et al. Interorgan molecular communication strategies of ‘Local’ and ‘Systemic’ innate immune responses in mosquito Anopheles stephensi. Front. Immunol. (2018). doi:10.3389/fimmu.2018.00148
  61. 61. Castillo, J. C., Robertson, A. E. & Strand, M. R. Characterization of hemocytes from the mosquitoes Anopheles gambiae and Aedes aegypti. Insect Biochem. Mol. Biol.36, 891-903 (2006)
  62. 62. Bryant, W. B. & Michel, K. Anopheles gambiae hemocytes exhibit transient states of activation. Dev. Comp. Immunol. (2016). doi:10.1016/j.dci.2015.10.020
  63. 63. Honti, V., Csordás, G., Kurucz, É., Márkus, R. & Andó, I. The cell-mediated immunity of Drosophila melanogaster: Hemocyte lineages, immune compartments, microanatomy and regulation. Dev. Comp. Immunol. (2014). doi:10.1016/j.dci.2013.06.005
  64. 64. Smith, R. C. et al. Molecular profiling of phagocytic immune cells in Anopheles gambiae reveals integral roles for hemocytes in mosquito innate immunity. Mol. Cell. Proteomics15, 3373-3387 (2016)
  65. 65. Kwon, H. & Smith, R. C. Chemical depletion of phagocytic immune cells reveals dual roles of mosquito hemocytes in Anopheles gambiae anti-Plasmodium 2 immunity 3. (2018). doi:10.1101/422543
  66. 66. Hillyer, J. F., Schmidt, S. L. & Christensen, B. M. Hemocyte-mediated phagocytosis and melanization in the mosquito Armigeres subalbatus following immune challenge by bacteria. Cell Tissue Res.313, 117-127 (2003)
  67. 67. King, J. G. & Hillyer, J. F. Spatial and temporal in vivo analysis of circulating and sessile immune cells in mosquitoes: Hemocyte mitosis following infection. BMC Biol.11, (2013)
  68. 68. Blandin, S. et al. Complement-like protein TEP1 is a determinant of vectorial capacity in the malaria vector Anopheles gambiae. Cell116, 661-670 (2004)
  69. 69. Yan, Y. & Hillyer, J. F. Complement-like proteins TEP1, TEP3 and TEP4 are positive regulators of periostial hemocyte aggregation in the mosquito Anopheles gambiae. Insect Biochem. Mol. Biol. (2019). doi:10.1016/j.ibmb.2019.01.007
  70. 70. Niu, G. et al. FBN30 in wild Anopheles gambiae functions as a pathogen recognition molecule against clinically circulating Plasmodium falciparum in malaria endemic areas in Kenya. Sci. Rep.7, (2017)
  71. 71. Simões, M. L. et al. The Anopheles FBN9 immune factor mediates Plasmodium species-specific defense through transgenic fat body expression. Dev. Comp. Immunol.67, 257-265 (2017)
  72. 72. Oliveira, S. B. et al. Identification of a fibrinogen-related protein (FBN9) gene in neotropical anopheline mosquitoes. Malar. J.10, 1-8 (2011)
  73. 73. Whitten, M. M. A., Tew, I. F., Lee, B. L. & Ratcliffe, N. A. A Novel Role for an Insect Apolipoprotein (Apolipophorin III) in β-1,3-Glucan Pattern Recognition and Cellular Encapsulation Reactions. J. Immunol. (2004). doi:10.4049/jimmunol.172.4.2177
  74. 74. Gupta, L. et al. Apolipophorin-III mediates antiplasmodial epithelial responses in Anopheles gambiae (G3) mosquitoes. PLoS One (2010). doi:10.1371/journal.pone.0015410
  75. 75. Chauhan C, Das De T, Kumari S, Rani J, Sharma P, Tevatiya S, Pandey KC, Pande V, Dixit R Hemocyte-specific FREP 13 abrogates the exogenous bactorial population in the hemolymph and promotes midgut endosymbionts in Anopheles stephensi. Immunol Cell Biol. 2020 oct;98 (9):757-769. doi: 10.1111/imcb.12374
  76. 76. Wells, M. B. & Andrew, D. J. Salivary gland cellular architecture in the Asian malaria vector mosquito Anopheles stephensi. Parasites and Vectors (2015). doi:10.1186/s13071-015-1229-z
  77. 77. Dhar, R. & Kumar, N. Role of mosquito salivary glands. Current Science85(9):1308- 1313 (2003)
  78. 78. Akaki, M. & Dvorak, J. A. A chemotactic response facilitates mosquito salivary gland infection by malaria sporozoites. J. Exp. Biol. (2005). doi:10.1242/jeb.01756
  79. 79. Sreenivasamurthy, S. K. et al. A compendium of molecules involved in vector-pathogen interactions pertaining to malaria. Malar. J. (2013). doi:10.1186/1475-2875-12-216
  80. 80. Aly, A. S. I. & Matuschewski, K. A malarial cysteine protease is necessary for Plasmodium sporozoite egress from oocysts. J. Exp. Med. (2005). doi:10.1084/jem.20050545
  81. 81. Boysen, K. E. & Matuschewski, K. Inhibitor of cysteine proteases is critical for motility and infectivity of Plasmodium sporozoites. MBio (2013). doi:10.1128/mBio.00874-13
  82. 82. Rodriguez, M. H. & Hernández-Hernández, F. D. L. C. Insect-malaria parasites interactions: The salivary gland. in Insect Biochemistry and Molecular Biology (2004). doi:10.1016/j.ibmb.2004.03.014
  83. 83. Brennan, J. D. G., Kent, M., Dhar, R., Fujioka, H. & Kumar, N. Anopheles gambiae salivary gland proteins as putative targets for blocking the transmission of malaria parasites. Proc. Natl. Acad. Sci. U. S. A. (2000). doi:10.1073/pnas.250472597
  84. 84. Sterling, C. R., Aikawa, M. & Vanderberg, J. P. The passage of Plasmodium berghei sporozoites through the salivary glands of Anopheles stephensi: An electron microscope study. J. Parasitol. (1973). doi:10.2307/3278847
  85. 85. Rosenberg, R. Inability of Plasmodium knowlesi sporozoites to invade Anopheles freeborni salivary glands. Am. J. Trop. Med. Hyg. (1985). doi:10.4269/ajtmh.1985.34.687
  86. 86. Pinto, S. B., Kafatos, F. C. & Michel, K. The parasite invasion marker SRPN6 reduces sporozoite numbers in salivary glands of Anopheles gambiae. Cell. Microbiol. (2008). doi:10.1111/j.1462-5822.2007.01091.x
  87. 87. Ghosh, A. K. et al. Malaria parasite invasion of the mosquito salivary gland requires interaction between the Plasmodium TRAP and the Anopheles saglin proteins. PLoS Pathog. (2009). doi:10.1371/journal.ppat.1000265
  88. 88. Okulate, M. A. et al. Identification and molecular characterization of a novel protein Saglin as a target of monoclonal antibodies affecting salivary gland infectivity of Plasmodium sporozoites. Insect Mol. Biol. (2007). doi:10.1111/j.1365-2583.2007.00765.x
  89. 89. Sultan, A. A. et al. TRAP is necessary for gliding motility and infectivity of Plasmodium sporozoites. Cell (1997). doi:10.1016/S0092-8674(00)80511-5
  90. 90. Sidjanski, S. P., Vanderberg, J. P. & Sinnis, P. Anopheles stephensi salivary glands bear receptors for region I of the circumsporozoite protein of Plasmodium falciparum. Mol. Biochem. Parasitol. (1997). doi:10.1016/S0166-6851(97)00124-2
  91. 91. Myung, J. M., Marshall, P. & Sinnis, P. The Plasmodium circumsporozoite protein is involved in mosquito salivary gland invasion by sporozoites. Mol. Biochem. Parasitol. (2004). doi:10.1016/j.molbiopara.2003.09.002
  92. 92. Rodrigues, J. et al. An epithelial serine protease, AgESP, is required for plasmodium invasion in the mosquito Anopheles gambiae. PLoS One (2012). doi:10.1371/journal.pone.0035210
  93. 93. Chertemps, T. et al. Anopheles gambiae PRS1 modulates Plasmodium development at both midgut and salivary gland steps. PLoS One (2010). doi:10.1371/journal.pone.0011538
  94. 94. Armistead, J. S., Wilson, I. B. H., Van Kuppevelt, T. H. & Dinglasan, R. R. A role for heparan sulfate proteoglycans in Plasmodium falciparum sporozoite invasion of anopheline mosquito salivary glands. Biochem. J. (2011). doi:10.1042/BJ20110694
  95. 95. Ramakrishnan, C. et al. Salivary gland-specific P. berghei reporter lines enable rapid evaluation of tissue-specific sporozoite loads in mosquitoes. PLoS One (2012). doi:10.1371/journal.pone.0036376
  96. 96. Ribeiro, J. M. Role of saliva in blood-feeding by arthropods. Annual review of entomology (1987). doi:10.1146/annurev.en.32.010187.002335
  97. 97. Ribeiro, J. M., Rossignol, P. A. & Spielman, A. Role of mosquito saliva in blood vessel location. J. Exp. Biol. 108: 1-7; (1984)
  98. 98. Fontaine, A. et al. Implication of haematophagous arthropod salivary proteins in host-vector interactions. Parasites and Vectors (2011). doi:10.1186/1756-3305-4-187
  99. 99. Schleicher, T. R. et al. A mosquito salivary gland protein partially inhibits Plasmodium sporozoite cell traversal and transmission. Nat. Commun. (2018). doi:10.1038/s41467-018-05374-3
  100. 100. Dragovic, S. M. et al. Immunization with AgTRIO, a Protein in Anopheles Saliva, Contributes to Protection against Plasmodium Infection in Mice. Cell Host Microbe (2018). doi:10.1016/j.chom.2018.03.008

Written By

Charu Chauhan, Sanjay Tevatiya, Seena Kumari, Punita Sharma, Jyoti Rani and Rajnikant Dixit

Submitted: 05 August 2020 Reviewed: 14 January 2021 Published: 23 March 2021