Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 100
1.
J Math Biol ; 89(1): 7, 2024 May 21.
Article En | MEDLINE | ID: mdl-38772937

Malaria is a vector-borne disease that exacts a grave toll in the Global South. The epidemiology of Plasmodium vivax, the most geographically expansive agent of human malaria, is characterised by the accrual of a reservoir of dormant parasites known as hypnozoites. Relapses, arising from hypnozoite activation events, comprise the majority of the blood-stage infection burden, with implications for the acquisition of immunity and the distribution of superinfection. Here, we construct a novel model for the transmission of P. vivax that concurrently accounts for the accrual of the hypnozoite reservoir, (blood-stage) superinfection and the acquisition of immunity. We begin by using an infinite-server queueing network model to characterise the within-host dynamics as a function of mosquito-to-human transmission intensity, extending our previous model to capture a discretised immunity level. To model transmission-blocking and antidisease immunity, we allow for geometric decay in the respective probabilities of successful human-to-mosquito transmission and symptomatic blood-stage infection as a function of this immunity level. Under a hybrid approximation-whereby probabilistic within-host distributions are cast as expected population-level proportions-we couple host and vector dynamics to recover a deterministic compartmental model in line with Ross-Macdonald theory. We then perform a steady-state analysis for this compartmental model, informed by the (analytic) distributions derived at the within-host level. To characterise transient dynamics, we derive a reduced system of integrodifferential equations, likewise informed by our within-host queueing network, allowing us to recover population-level distributions for various quantities of epidemiological interest. In capturing the interplay between hypnozoite accrual, superinfection and acquired immunity-and providing, to the best of our knowledge, the most complete population-level distributions for a range of epidemiological values-our model provides insights into important, but poorly understood, epidemiological features of P. vivax.


Malaria, Vivax , Mathematical Concepts , Mosquito Vectors , Plasmodium vivax , Superinfection , Humans , Plasmodium vivax/immunology , Plasmodium vivax/physiology , Superinfection/immunology , Superinfection/transmission , Superinfection/parasitology , Malaria, Vivax/transmission , Malaria, Vivax/immunology , Malaria, Vivax/epidemiology , Malaria, Vivax/parasitology , Animals , Mosquito Vectors/parasitology , Mosquito Vectors/immunology , Disease Reservoirs/parasitology , Models, Biological , Computer Simulation , Anopheles/parasitology , Anopheles/immunology
2.
Front Immunol ; 15: 1368066, 2024.
Article En | MEDLINE | ID: mdl-38751433

Introduction: Aedes spp. are the most prolific mosquito vectors in the world. Found on every continent, they can effectively transmit various arboviruses, including the dengue virus which continues to cause outbreaks worldwide and is spreading into previously non-endemic areas. The lack of widely available dengue vaccines accentuates the importance of targeted vector control strategies to reduce the dengue burden. High-throughput tools to estimate human-mosquito contact and evaluate vector control interventions are lacking. We propose a novel serological tool that allows rapid screening of human cohorts for exposure to potentially infectious mosquitoes. Methods: We tested 563 serum samples from a longitudinal pediatric cohort study previously conducted in Cambodia. Children enrolled in the study were dengue-naive at baseline and were followed biannually for dengue incidence for two years. We used Western blotting and enzyme-linked immunosorbent assays to identify immunogenic Aedes aegypti salivary proteins and measure total anti-Ae. aegypti IgG. Results: We found a correlation (rs=0.86) between IgG responses against AeD7L1 and AeD7L2 recombinant proteins and those to whole salivary gland homogenate. We observed seasonal fluctuations of AeD7L1+2 IgG responses and no cross-reactivity with Culex quinquefasciatus and Anopheles dirus mosquitoes. The baseline median AeD7L1+2 IgG responses for young children were higher in those who developed asymptomatic versus symptomatic dengue. Discussion: The IgG response against AeD7L1+2 recombinant proteins is a highly sensitive and Aedes specific marker of human exposure to Aedes bites that can facilitate standardization of future serosurveys and epidemiological studies by its ability to provide a robust estimation of human-mosquito contact in a high-throughput fashion.


Aedes , Dengue , Insect Proteins , Mosquito Vectors , Salivary Proteins and Peptides , Humans , Aedes/immunology , Aedes/virology , Animals , Salivary Proteins and Peptides/immunology , Child , Mosquito Vectors/immunology , Mosquito Vectors/virology , Dengue/immunology , Dengue/transmission , Insect Proteins/immunology , Female , Child, Preschool , Immunoglobulin G/immunology , Immunoglobulin G/blood , Male , Cambodia , Longitudinal Studies , Dengue Virus/immunology , Adolescent , Insect Bites and Stings/immunology
3.
PLoS Pathog ; 20(4): e1012145, 2024 Apr.
Article En | MEDLINE | ID: mdl-38598552

Wolbachia, a maternally transmitted symbiotic bacterium of insects, can suppress a variety of human pathogens in mosquitoes, including malaria-causing Plasmodium in the Anopheles vector. However, the mechanistic basis of Wolbachia-mediated Plasmodium suppression in mosquitoes is not well understood. In this study, we compared the midgut and carcass transcriptomes of stably infected Anopheles stephensi with Wolbachia wAlbB to uninfected mosquitoes in order to discover Wolbachia infection-responsive immune genes that may play a role in Wolbachia-mediated anti-Plasmodium activity. We show that wAlbB infection upregulates 10 putative immune genes and downregulates 14 in midguts, while it upregulates 31 putative immune genes and downregulates 15 in carcasses at 24 h after blood-fed feeding, the time at which the Plasmodium ookinetes are traversing the midgut tissue. Only a few of these regulated immune genes were also significantly differentially expressed between Wolbachia-infected and non-infected midguts and carcasses of sugar-fed mosquitoes. Silencing of the Wolbachia infection-responsive immune genes TEP 4, TEP 15, lysozyme C2, CLIPB2, CLIPB4, PGRP-LD and two novel genes (a peritrophin-44-like gene and a macro domain-encoding gene) resulted in a significantly greater permissiveness to P. falciparum infection. These results indicate that Wolbachia infection modulates mosquito immunity and other processes that are likely to decrease Anopheles permissiveness to Plasmodium infection.


Anopheles , Malaria, Falciparum , Plasmodium falciparum , Wolbachia , Animals , Anopheles/parasitology , Anopheles/microbiology , Anopheles/immunology , Wolbachia/immunology , Plasmodium falciparum/immunology , Malaria, Falciparum/immunology , Malaria, Falciparum/parasitology , Mosquito Vectors/parasitology , Mosquito Vectors/microbiology , Mosquito Vectors/immunology , Insect Proteins/genetics , Insect Proteins/metabolism , Insect Proteins/immunology , Transcriptome , Female
4.
mBio ; 12(6): e0309121, 2021 12 21.
Article En | MEDLINE | ID: mdl-34903042

Malaria is caused when Plasmodium sporozoites are injected along with saliva by an anopheline mosquito into the dermis of a vertebrate host. Arthropod saliva has pleiotropic effects that can influence local host responses, pathogen transmission, and exacerbation of the disease. A mass spectrometry screen identified mosquito salivary proteins that are associated with Plasmodium sporozoites during saliva secretions. In this study, we demonstrate that one of these salivary antigens, Anopheles gambiae sporozoite-associated protein (AgSAP), interacts directly with Plasmodium falciparum and Plasmodium berghei sporozoites. AgSAP binds to heparan sulfate and inhibits local inflammatory responses in the skin. The silencing of AgSAP in mosquitoes reduces their ability to effectively transmit sporozoites to mice. Moreover, immunization with AgSAP decreases the Plasmodium burden in mice that are bitten by Plasmodium-infected mosquitoes. These data suggest that AgSAP facilitates early Plasmodium infection in the vertebrate host and serves as a target for the prevention of malaria. IMPORTANCE Malaria is a vector-borne disease caused by Plasmodium sporozoites. When an anopheline mosquito bites its host, it releases Plasmodium sporozoites as well as saliva components. Mosquito proteins have the potential to serve as antigens to prevent or influence malaria without directly targeting the pathogen. This may help set a new paradigm for vaccine development. In this study, we have elucidated the role of a novel salivary antigen, named Anopheles gambiae sporozoite-associated protein (AgSAP). The results presented here show that AgSAP interacts with Plasmodium falciparum and Plasmodium berghei sporozoites and modulates local inflammatory responses in the skin. Furthermore, our results show that AgSAP is a novel mosquito salivary antigen that influences the early stages of Plasmodium infection in the vertebrate host. Individuals living in countries where malaria is endemic generate antibodies against AgSAP, which indicates that AgSAP can serve as a biomarker for disease prevalence and epidemiological analysis.


Anopheles/immunology , Insect Proteins/immunology , Malaria/parasitology , Mosquito Vectors/immunology , Plasmodium berghei/physiology , Plasmodium falciparum/physiology , Salivary Proteins and Peptides/immunology , Animals , Anopheles/genetics , Anopheles/parasitology , Female , Humans , Insect Proteins/genetics , Malaria/immunology , Malaria/transmission , Mice , Mice, Inbred C57BL , Mosquito Vectors/genetics , Mosquito Vectors/parasitology , Plasmodium berghei/genetics , Plasmodium falciparum/genetics , Salivary Proteins and Peptides/genetics , Sporozoites/genetics , Sporozoites/physiology
5.
Elife ; 102021 12 23.
Article En | MEDLINE | ID: mdl-34939933

Background: Entomological surveillance for malaria is inherently resource-intensive and produces crude population-level measures of vector exposure which are insensitive in low-transmission settings. Antibodies against Anopheles salivary proteins measured at the individual level may serve as proxy biomarkers for vector exposure and malaria transmission, but their relationship is yet to be quantified. Methods: A systematic review of studies measuring antibodies against Anopheles salivary antigens (PROSPERO: CRD42020185449). Multilevel modelling (to account for multiple study-specific observations [level 1], nested within study [level 2], and study nested within country [level 3]) estimated associations between seroprevalence with Anopheles human biting rate (HBR) and malaria transmission measures. Results: From 3981 studies identified in literature searches, 42 studies across 16 countries were included contributing 393 study-specific observations of anti-Anopheles salivary antibodies determined in 42,764 samples. A positive association between HBR (log transformed) and seroprevalence was found; overall a twofold (100% relative) increase in HBR was associated with a 23% increase in odds of seropositivity (OR: 1.23, 95% CI: 1.10-1.37; p<0.001). The association between HBR and Anopheles salivary antibodies was strongest with concordant, rather than discordant, Anopheles species. Seroprevalence was also significantly positively associated with established epidemiological measures of malaria transmission: entomological inoculation rate, Plasmodium spp. prevalence, and malarial endemicity class. Conclusions: Anopheles salivary antibody biomarkers can serve as a proxy measure for HBR and malaria transmission, and could monitor malaria receptivity of a population to sustain malaria transmission. Validation of Anopheles species-specific biomarkers is important given the global heterogeneity in the distribution of Anopheles species. Salivary biomarkers have the potential to transform surveillance by replacing impractical, inaccurate entomological investigations, especially in areas progressing towards malaria elimination. Funding: Australian National Health and Medical Research Council, Wellcome Trust.


Anopheles/immunology , Antigens, Protozoan/immunology , Insect Proteins/immunology , Malaria/transmission , Salivary Proteins and Peptides/immunology , Animals , Antibodies, Protozoan/immunology , Australia , Biomarkers , Humans , Immunoglobulin G/immunology , Insect Bites and Stings , Malaria/epidemiology , Malaria/immunology , Models, Theoretical , Mosquito Vectors/immunology , Plasmodium falciparum/immunology , Seroepidemiologic Studies
6.
Sci Rep ; 11(1): 23699, 2021 12 08.
Article En | MEDLINE | ID: mdl-34880307

Dengue, caused by the dengue virus (DENV) is a significant vector-borne disease. In absence of a specific treatment and vaccine, dengue is becoming a rising threat to public health. Currently, control of dengue mainly focuses on the surveillance of the mosquito vectors. Improved surveillance methods for DENV in mosquito populations would be highly beneficial to the public health. However, current methods of DENV detection in mosquitoes requires specialized equipment and expensive reagents and highly trained personnel. As an alternative, commercially available dengue NS1 antigen ELISA kits could be used for detection of DENV infection in Aedes aegypti mosquitoes. In this study, we explored the utility of commercially available Dengue NS1 antigen kit (J. Mitra & Co. Pvt. Ltd) for the detection of recombinant dengue virus-2 (rDENV-2) NS1 protein and serum of dengue infected patient spiked with Ae. aegypti mosquito pools. The kit was found to be highly sensitive and specific towards detection of all serotypes of DENV. Further, it could detect as low as 750 femto gram rDENV-2 NS1 protein. It was also observed that rDENV-2 NS1 antigen spiked with blood-fed and unfed mosquito pools could be detected. In addition, the kit also detected dengue infected patient serum spiked with Ae. aegypti mosquito pools. Overall, the Dengue NS1 antigen kit displayed high sensitivity towards detection of recombinant as well as serum NS1 protein spiked with Ae. aegypti mosquito pools and could be considered for the dengue virus surveillance after a field evaluation in Ae. aegypti mosquitoes.


Aedes/virology , Antigens, Viral/immunology , Dengue Virus/immunology , Enzyme-Linked Immunosorbent Assay/methods , Mosquito Vectors/virology , Viral Nonstructural Proteins/immunology , Aedes/immunology , Animals , Dengue/diagnosis , Dengue/immunology , Dengue/transmission , Dengue/virology , Dengue Virus/classification , Enzyme-Linked Immunosorbent Assay/standards , Humans , Mosquito Vectors/immunology , Reagent Kits, Diagnostic , Sensitivity and Specificity , Serogroup
7.
Viruses ; 13(11)2021 10 20.
Article En | MEDLINE | ID: mdl-34834923

Mosquito-borne viruses of the Flavivirus genus (Flaviviridae family) pose an ongoing threat to global public health. For example, dengue, Japanese encephalitis, West Nile, yellow fever, and Zika viruses are transmitted by infected mosquitoes and cause severe and fatal diseases in humans. The means by which mosquito-borne flaviviruses establish persistent infection in mosquitoes and cause disease in humans are complex and depend upon a myriad of virus-host interactions, such as those of the innate immune system, which are the main focus of our review. This review also covers the different strategies utilized by mosquito-borne flaviviruses to antagonize the innate immune response in humans and mosquitoes. Given the lack of antiviral therapeutics for mosquito-borne flaviviruses, improving our understanding of these virus-immune interactions could lead to new antiviral therapies and strategies for developing refractory vectors incapable of transmitting these viruses, and can also provide insights into determinants of viral tropism that influence virus emergence into new species.


Culicidae/immunology , Flavivirus Infections/immunology , Flavivirus Infections/veterinary , Flavivirus/immunology , Persistent Infection/immunology , Persistent Infection/veterinary , Animals , Culicidae/physiology , Culicidae/virology , Flavivirus/genetics , Flavivirus/physiology , Flavivirus Infections/transmission , Flavivirus Infections/virology , Humans , Immunity, Innate , Mosquito Vectors/immunology , Mosquito Vectors/physiology , Mosquito Vectors/virology , Persistent Infection/virology
8.
PLoS Negl Trop Dis ; 15(11): e0009984, 2021 11.
Article En | MEDLINE | ID: mdl-34843477

Microbial control of mosquitoes via the use of symbiotic or pathogenic microbes, such as Wolbachia and entomopathogenic fungi, are promising alternatives to synthetic insecticides to tackle the rapid increase in insecticide resistance and vector-borne disease outbreaks. This study evaluated the susceptibility and host responses of two important mosquito vectors, Ae. albopictus and Cx. pipiens, that naturally carry Wolbachia, to infections by entomopathogenic fungi. Our study indicated that while Wolbachia presence did not provide a protective advantage against entomopathogenic fungal infection, it nevertheless influenced the bacterial / fungal load and the expression of select anti-microbial effectors and phenoloxidase cascade genes in mosquitoes. Furthermore, although host responses from Ae. albopictus and Cx. pipiens were mostly similar, we observed contrasting phenotypes with regards to susceptibility and immune responses to fungal entomopathogenic infection in these two mosquitoes. This study provides new insights into the intricate multipartite interaction between the mosquito host, its native symbiont and pathogenic microbes that might be employed to control mosquito populations.


Aedes/immunology , Culex/immunology , Immunity/genetics , Mosquito Vectors/immunology , Wolbachia/genetics , Aedes/drug effects , Aedes/genetics , Aedes/microbiology , Animals , Culex/drug effects , Culex/genetics , Culex/microbiology , Fungi , Gene Expression , Insecticide Resistance , Insecticides , Monophenol Monooxygenase/genetics , Monophenol Monooxygenase/metabolism , Mosquito Vectors/microbiology , Symbiosis , Vector Borne Diseases
9.
Parasit Vectors ; 14(1): 566, 2021 Nov 03.
Article En | MEDLINE | ID: mdl-34732254

BACKGROUND: A number of zoonotic mosquito-borne viruses have emerged in Europe in recent decades. Batai virus (BATV), a member of the genus Orthobunyavirus, is one example of a relatively newly emerged mosquito-borne virus, having been detected in mosquitoes and livestock. We conducted vector competency studies on three mosquito species at a low temperature to assess whether Aedes and Culex mosquito species are susceptible to infection with BATV. METHODS: Colonised lines of Aedes aegypti and Culex pipiens and a wild-caught species, Aedes detritus, were orally inoculated with BATV strain 53.2, originally isolated from mosquitoes trapped in Germany in 2009. Groups of blood-fed female mosquitoes were maintained at 20 °C for 7 or 14 days. Individual mosquitoes were screened for the presence of BATV in body, leg and saliva samples for evidence of infection, dissemination and transmission, respectively. BATV RNA was detected by reverse transcription-PCR, and positive results confirmed by virus isolation in Vero cells. RESULTS: Aedes detritus was highly susceptible to BATV, with an infection prevalence of ≥ 80% at both measurement time points. Disseminated infections were recorded in 30.7-41.6% of Ae. detritus, and evidence of virus transmission with BATV in saliva samples (n = 1, days post-infection: 14) was observed. Relatively lower rates of infection for Ae. aegypti and Cx. pipiens were observed, with no evidence of virus dissemination or transmission at either time point. CONCLUSIONS: This study shows that Ae. detritus may be a competent vector for BATV at 20 °C, whereas Ae. aegypti and Cx. pipiens were not competent. Critically, the extrinsic incubation period appears to be ≤ 7 days for Ae. detritus, which may increase the onward transmissibility potential of BATV in these populations.


Bunyamwera virus/physiology , Culicidae/virology , Mosquito Vectors/virology , Animals , Bunyamwera virus/genetics , Bunyaviridae Infections/transmission , Bunyaviridae Infections/virology , Culicidae/immunology , Europe , Female , Humans , Male , Mosquito Vectors/immunology , Saliva/virology
10.
Front Immunol ; 12: 680020, 2021.
Article En | MEDLINE | ID: mdl-34484178

Lipid-derived signaling molecules known as eicosanoids have integral roles in mediating immune and inflammatory processes across metazoans. This includes the function of prostaglandins and their cognate G protein-coupled receptors (GPCRs) to employ their immunological actions. In insects, prostaglandins have been implicated in the regulation of both cellular and humoral immune responses, yet in arthropods of medical importance, studies have been limited. Here, we describe a prostaglandin E2 receptor (AgPGE2R) in the mosquito Anopheles gambiae and demonstrate that its expression is most abundant in oenocytoid immune cell populations. Through the administration of prostaglandin E2 (PGE2) and AgPGE2R-silencing, we demonstrate that prostaglandin E2 signaling regulates a subset of prophenoloxidases (PPOs) and antimicrobial peptides (AMPs) that are strongly expressed in populations of oenocytoids. We demonstrate that PGE2 signaling via the AgPGE2R significantly limits both bacterial replication and Plasmodium oocyst survival. Additional experiments establish that PGE2 treatment increases phenoloxidase (PO) activity through the increased expression of PPO1 and PPO3, genes essential to anti-Plasmodium immune responses that promote oocyst killing. We also provide evidence that the mechanisms of PGE2 signaling are concentration-dependent, where high concentrations of PGE2 promote oenocytoid lysis, negating the protective effects of lower concentrations of PGE2 on anti-Plasmodium immunity. Taken together, our results provide new insights into the role of PGE2 signaling on immune cell function and its contributions to mosquito innate immunity that promote pathogen killing.


Anopheles/immunology , Anopheles/microbiology , Anopheles/parasitology , Dinoprostone/metabolism , Oocysts/immunology , Plasmodium/immunology , Signal Transduction , Animals , Anopheles/classification , Hemocytes/metabolism , Host-Pathogen Interactions/immunology , Immunity, Innate , Microbial Viability , Mosquito Vectors/immunology , Mosquito Vectors/microbiology , Mosquito Vectors/parasitology , Phylogeny , Plasmodium/growth & development , Pore Forming Cytotoxic Proteins/metabolism , Receptors, Prostaglandin E/genetics , Receptors, Prostaglandin E/metabolism
11.
Elife ; 102021 07 28.
Article En | MEDLINE | ID: mdl-34318744

Mosquito immune cells, known as hemocytes, are integral to cellular and humoral responses that limit pathogen survival and mediate immune priming. However, without reliable cell markers and genetic tools, studies of mosquito immune cells have been limited to morphological observations, leaving several aspects of their biology uncharacterized. Here, we use single-cell RNA sequencing (scRNA-seq) to characterize mosquito immune cells, demonstrating an increased complexity to previously defined prohemocyte, oenocytoid, and granulocyte subtypes. Through functional assays relying on phagocytosis, phagocyte depletion, and RNA-FISH experiments, we define markers to accurately distinguish immune cell subtypes and provide evidence for immune cell maturation and differentiation. In addition, gene-silencing experiments demonstrate the importance of lozenge in defining the mosquito oenocytoid cell fate. Together, our scRNA-seq analysis provides an important foundation for future studies of mosquito immune cell biology and a valuable resource for comparative invertebrate immunology.


Anopheles/immunology , Cell Differentiation/immunology , Hemocytes/immunology , Mosquito Vectors/immunology , Phagocytes/immunology , Animals , Anopheles/genetics , Drosophila , Female , Gene Silencing , Hemocytes/metabolism , Immunity, Innate , Malaria/immunology , Malaria/parasitology , Mosquito Vectors/genetics , Phagocytes/metabolism , Sequence Analysis, RNA , Single-Cell Analysis
12.
Nat Microbiol ; 6(6): 806-817, 2021 06.
Article En | MEDLINE | ID: mdl-33958765

The stalling global progress in the fight against malaria prompts the urgent need to develop new intervention strategies. Whilst engineered symbiotic bacteria have been shown to confer mosquito resistance to parasite infection, a major challenge for field implementation is to address regulatory concerns. Here, we report the identification of a Plasmodium-blocking symbiotic bacterium, Serratia ureilytica Su_YN1, isolated from the midgut of wild Anopheles sinensis in China that inhibits malaria parasites via secretion of an antimalarial lipase. Analysis of Plasmodium vivax epidemic data indicates that local malaria cases in Tengchong (Yunnan province, China) are significantly lower than imported cases and importantly, that the local vector A. sinensis is more resistant to infection by P. vivax than A. sinensis from other regions. Analysis of the gut symbiotic bacteria of mosquitoes from Yunnan province led to the identification of S. ureilytica Su_YN1. This bacterium renders mosquitoes resistant to infection by the human parasite Plasmodium falciparum or the rodent parasite Plasmodium berghei via secretion of a lipase that selectively kills parasites at various stages. Importantly, Su_YN1 rapidly disseminates through mosquito populations by vertical and horizontal transmission, providing a potential tool for blocking malaria transmission in the field.


Anopheles/microbiology , Bacterial Proteins/immunology , Lipase/immunology , Mosquito Vectors/microbiology , Serratia/enzymology , Serratia/isolation & purification , Animals , Anopheles/immunology , Anopheles/parasitology , Anopheles/physiology , Bacterial Proteins/genetics , China , Female , Gastrointestinal Tract/microbiology , Humans , Lipase/genetics , Malaria, Vivax/transmission , Male , Mosquito Vectors/immunology , Mosquito Vectors/parasitology , Mosquito Vectors/physiology , Plasmodium falciparum/physiology , Plasmodium vivax/physiology , Serratia/genetics , Serratia/physiology , Symbiosis
13.
Viruses ; 13(5)2021 04 24.
Article En | MEDLINE | ID: mdl-33923307

Mosquito-borne viral infections are responsible for a significant degree of morbidity and mortality across the globe due to the severe diseases these infections cause, and they continue to increase each year. These viruses are dependent on the mosquito vector as the primary means of transmission to new vertebrate hosts including avian, livestock, and human populations. Due to the dynamic host environments that mosquito-borne viruses pass through as they are transmitted between vector and vertebrate hosts, there are various host factors that control the response to infection over the course of the pathogen's life cycle. In this review, we discuss these host factors that are present in either vector or vertebrate models during infection, how they vary or are conserved between hosts, and their implications in future research pertaining to disease prevention and treatment.


Arbovirus Infections/transmission , Arboviruses/pathogenicity , Culicidae/virology , Host Microbial Interactions , Mosquito Vectors/virology , Animals , Arbovirus Infections/virology , Culicidae/immunology , Humans , Life Cycle Stages , Livestock/virology , Mosquito Vectors/immunology
14.
Front Immunol ; 12: 640367, 2021.
Article En | MEDLINE | ID: mdl-33767710

The C-type lectins, one family of lectins featuring carbohydrate binding domains which participate in a variety of bioprocesses in both humans and mosquitoes, including immune response, are known to target DENV. A human C-type lectin protein CLEC18A in particular shows extensive glycan binding abilities and correlates with type-I interferon expression, making CLEC18A a potential player in innate immune responses to DENV infection; this potential may provide additional regulatory point in improving mosquito immunity. Here, we established for the first time a transgenic Aedes aegypti line that expresses human CLEC18A. This expression enhanced the Toll immune pathway responses to DENV infection. Furthermore, viral genome and virus titers were reduced by 70% in the midgut of transgenic mosquitoes. We found significant changes in the composition of the midgut microbiome in CLEC18A expressing mosquitoes, which may result from the Toll pathway enhancement and contribute to DENV inhibition. Transgenic mosquito lines offer a compelling option for studying DENV pathogenesis, and our analyses indicate that modifying the mosquito immune system via expression of a human immune gene can significantly reduce DENV infection.


Aedes/immunology , Aedes/virology , Animals, Genetically Modified , Dengue/immunology , Lectins, C-Type/immunology , Aedes/genetics , Animals , Animals, Genetically Modified/genetics , Animals, Genetically Modified/immunology , Dengue Virus , Disease Models, Animal , Humans , Mosquito Vectors/genetics , Mosquito Vectors/immunology , Mosquito Vectors/virology
15.
PLoS Pathog ; 17(2): e1009353, 2021 02.
Article En | MEDLINE | ID: mdl-33626094

Repeated blood meals provide essential nutrients for mosquito egg development and routes for pathogen transmission. The target of rapamycin, the TOR pathway, is essential for vitellogenesis. However, its influence on pathogen transmission remains to be elucidated. Here, we show that rapamycin, an inhibitor of the TOR pathway, effectively suppresses Plasmodium berghei infection in Anopheles stephensi. An. stephensi injected with rapamycin or feeding on rapamycin-treated mice showed increased resistance to P. berghei infection. Exposing An. stephensi to a rapamycin-coated surface not only decreased the numbers of both oocysts and sporozoites but also impaired mosquito survival and fecundity. Transcriptome analysis revealed that the inhibitory effect of rapamycin on parasite infection was through the enhanced activation of immune responses, especially the NF-κB transcription factor REL2, a regulator of the immune pathway and complement system. Knockdown of REL2 in rapamycin-treated mosquitoes abrogated the induction of the complement-like proteins TEP1 and SPCLIP1 and abolished rapamycin-mediated refractoriness to Plasmodium infection. Together, these findings demonstrate a key role of the TOR pathway in regulating mosquito immune responses, thereby influencing vector competence.


Anopheles/drug effects , Immunity, Innate/immunology , Malaria/drug therapy , Mosquito Vectors/drug effects , Plasmodium berghei/pathogenicity , Sirolimus/pharmacology , Animals , Anopheles/immunology , Anopheles/parasitology , Female , Gene Expression Profiling , Immunity, Innate/drug effects , Immunosuppressive Agents/pharmacology , Malaria/immunology , Malaria/parasitology , Malaria/transmission , Mice , Mice, Inbred BALB C , Mosquito Vectors/immunology , Mosquito Vectors/parasitology , Oocysts/drug effects , Oocysts/growth & development , Oocysts/immunology , Sporozoites/drug effects , Sporozoites/growth & development , Sporozoites/immunology
16.
Commun Biol ; 4(1): 69, 2021 01 15.
Article En | MEDLINE | ID: mdl-33452408

Inter-host transmission of pathogenic arboviruses such as dengue virus (DENV) and Zika virus (ZIKV) requires systemic infection of the mosquito vector. Successful systemic infection requires initial viral entry and proliferation in the midgut cells of the mosquito followed by dissemination to secondary tissues and eventual entry into salivary glands1. Lack of arbovirus proliferation in midgut cells has been observed in several Aedes aegypti strains2, but the midgut antiviral responses underlying this phenomenon are not yet fully understood. We report here that there is a rapid induction of apoptosis (RIA) in the Aedes aegypti midgut epithelium within 2 hours of infection with DENV-2 or ZIKV in both in vivo blood-feeding and ex vivo midgut infection models. Inhibition of RIA led to increased virus proliferation in the midgut, implicating RIA as an innate immune mechanism mediating midgut infection in this mosquito vector.


Aedes/virology , Apoptosis , Dengue Virus/physiology , Host-Pathogen Interactions/immunology , Mosquito Vectors/virology , Zika Virus/physiology , Aedes/immunology , Animals , Female , Intestinal Mucosa/immunology , Intestinal Mucosa/virology , Mosquito Vectors/immunology
17.
Expert Rev Vaccines ; 20(2): 199-214, 2021 02.
Article En | MEDLINE | ID: mdl-33430656

Introduction: The development of effective vaccines remains a major health priority to combat the global burden of malaria, a life-threatening disease caused by Plasmodium parasites. Transmission-blocking vaccines (TBVs) elicit antibodies that neutralize the sexual stages of the parasite in blood meals ingested by the Anopheles mosquito, interrupting parasite development in the vector host and preventing disease spread to other individuals.Areas covered: The P. falciparum gametocyte surface antigens Pfs230, Pfs48/45, and Pfs47, the parasite ookinete surface protein Pfs25, and the male gametocyte specific protein PfHAP2 are leading TBV candidates, some of which are in clinical development. The recent expansion of methodology to study monoclonal antibodies isolated directly from humans and animal models, coupled with effective measures for parasite neutralization, has provided unprecedented insight into TBV efficacy and development.Expert opinion: Available structural and functional data on antigen-monoclonal antibody (Ag-mAb) complexes, as well as epitope classification studies, have identified neutralizing epitopes that may aid vaccine development and improve protection. Here, we review the clinical prospects of TBV candidates, progress in the development of novel vaccine strategies for TBVs, and the impact of structural vaccinology in TBV design.


Malaria Vaccines/administration & dosage , Malaria, Falciparum/prevention & control , Plasmodium falciparum/immunology , Animals , Anopheles/parasitology , Antibodies, Monoclonal/immunology , Antibodies, Protozoan/immunology , Antigens, Protozoan/immunology , Humans , Malaria Vaccines/immunology , Malaria, Falciparum/immunology , Malaria, Falciparum/transmission , Mosquito Vectors/immunology , Mosquito Vectors/parasitology , Plasmodium falciparum/parasitology , Vaccinology
18.
Expert Rev Vaccines ; 20(2): 185-198, 2021 02.
Article En | MEDLINE | ID: mdl-33478283

INTRODUCTION: Transmission-blocking vaccines (TBV) prevent community spread of malaria by targeting mosquito sexual stage parasites, a life-cycle bottleneck, and will be used in elimination programs. TBV rely on herd immunity to reduce mosquito infections and thereby new infections in both vaccine recipients and non-recipients, but do not provide protection once an individual receives an infectious mosquito bite which complicates clinical development. AREAS COVERED: Here, we describe the concept and biology behind TBV, and we provide an update on clinical development of the leading vaccine candidate antigens. Search terms 'malaria vaccine,' 'sexual stages,' 'transmission blocking vaccine,' 'VIMT' and 'SSM-VIMT' were used for PubMed queries to identify relevant literature. EXPERT OPINION: Candidates targeting P. falciparum zygote surface antigen Pfs25, and its P. vivax orthologue Pvs25, induced functional activity in humans that reduced mosquito infection in surrogate assays, but require increased durability to be useful in the field. Candidates targeting gamete surface antigens Pfs230 and Pfs48/45, respectively, are in or nearing clinical trials. Nanoparticle platforms and adjuvants are being explored to enhance immunogenicity. Efficacy trials require special considerations, such as cluster-randomized designs to measure herd immunity that reduces human and mosquito infection rates, while addressing human and mosquito movements as confounding factors.


Malaria Vaccines/administration & dosage , Malaria, Falciparum/prevention & control , Malaria, Vivax/prevention & control , Animals , Antibodies, Protozoan/immunology , Antigens, Surface/immunology , Humans , Immunity, Herd/immunology , Malaria Vaccines/immunology , Malaria, Falciparum/immunology , Malaria, Falciparum/transmission , Malaria, Vivax/immunology , Malaria, Vivax/transmission , Mosquito Control/methods , Mosquito Vectors/immunology , Mosquito Vectors/parasitology , Plasmodium falciparum/immunology , Plasmodium vivax/immunology
19.
Dev Comp Immunol ; 119: 104010, 2021 06.
Article En | MEDLINE | ID: mdl-33476667

The genetic basis of antiviral immunity in dipteran insects is extensively studied in Drosophila melanogaster and advanced technologies for genetic manipulation allow a better characterization of immune responses also in non-model insect species. Especially, immunity in vector mosquitoes is recently in the spotlight, due to the medical impact that these insects have by transmitting viruses and other pathogens. Here, we review the current state of experimental evidence that supports antiviral functions for immune genes acting in different cellular pathways. We discuss the well-characterized RNA interference mechanism along with the less well-defined JAK-STAT, Toll, and IMD signaling pathways. Furthermore, we highlight the initial evidence for antiviral activity observed for the autophagy pathway, transcriptional pausing, as well as piRNA production from endogenous viral elements. We focus our review on studies from Drosophila and mosquito species from the lineages Aedes, Culex, and Anopheles, which contain major vector species responsible for virus transmission.


Diptera/immunology , Genes, Insect/immunology , Immunity, Innate/immunology , Insect Viruses/immunology , Signal Transduction/immunology , Animals , Culicidae/genetics , Culicidae/immunology , Culicidae/virology , Diptera/genetics , Diptera/virology , Drosophila melanogaster/genetics , Drosophila melanogaster/immunology , Drosophila melanogaster/virology , Genes, Insect/genetics , Host-Pathogen Interactions/immunology , Immunity, Innate/genetics , Insect Viruses/physiology , Mosquito Vectors/genetics , Mosquito Vectors/immunology , Mosquito Vectors/virology , RNA, Small Interfering/genetics , RNA, Small Interfering/immunology , Signal Transduction/genetics
20.
Open Biol ; 11(1): 200246, 2021 01.
Article En | MEDLINE | ID: mdl-33401993

The principal vector of dengue, Zika and chikungunya viruses is the mosquito Aedes aegypti, with its ability to transmit pathogens influenced by ambient temperature. We use chikungunya virus (CHIKV) to understand how the mosquito transcriptome responds to arbovirus infection at different ambient temperatures. We exposed CHIKV-infected mosquitoes to 18, 28 and 32°C, and found that higher temperature correlated with higher virus levels, particularly at 3 days post infection, but lower temperature resulted in reduced virus levels. RNAseq analysis indicated significantly altered gene expression levels in CHIKV infection. The highest number of significantly differentially expressed genes was observed at 28°C, with a more muted effect at the other temperatures. At the higher temperature, the expression of many classical immune genes, including Dicer-2, was not substantially altered in response to CHIKV. The upregulation of Toll, IMD and JAK-STAT pathways was only observed at 28°C. Functional annotations suggested that genes in immune response and metabolic pathways related to energy supply and DNA replication were involved in temperature-dependent changes. Time post infection also led to substantially different gene expression profiles, and this varied with temperature. In conclusion, temperature significantly modulates mosquito gene expression in response to infection, potentially leading to impairment of immune defences at higher temperatures.


Aedes/metabolism , Chikungunya virus/physiology , Immunity/genetics , Mosquito Vectors/immunology , Aedes/virology , Animals , Down-Regulation , Gene Ontology , Mosquito Vectors/virology , RNA, Long Noncoding/metabolism , Signal Transduction/genetics , Temperature , Up-Regulation
...