Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 269
Filter
1.
Curr Opin Insect Sci ; 63: 101195, 2024 06.
Article in English | MEDLINE | ID: mdl-38552792

ABSTRACT

Mosquitoes encounter diverse microbes during their lifetime, including symbiotic bacteria, shaping their midgut ecosystem. The organization of the midgut supports microbiota persistence while defending against potential pathogens. The influx of nutrients during blood feeding triggers bacterial proliferation, challenging host homeostasis. Immune responses, aimed at controlling bacterial overgrowth, impact blood-borne pathogens such as malaria parasites. However, parasites deploy evasion strategies against mosquito immunity. Leveraging these mechanisms could help engineer malaria-resistant mosquitoes, offering a transformative tool for malaria elimination.


Subject(s)
Culicidae , Gastrointestinal Microbiome , Animals , Culicidae/microbiology , Culicidae/physiology , Culicidae/immunology , Symbiosis , Gastrointestinal Tract/microbiology , Gastrointestinal Tract/immunology
2.
EBioMedicine ; 86: 104375, 2022 Dec.
Article in English | MEDLINE | ID: mdl-36436281

ABSTRACT

BACKGROUND: Immunity to mosquito salivary proteins could provide protection against multiple mosquito-borne diseases and significantly impact public health. We evaluated the safety and immunogenicity of AGS-v PLUS, a mosquito salivary peptide vaccine, in healthy adults 18-50 years old. METHODS: We conducted a randomized, double-blind, placebo-controlled Phase 1 study of AGS-v PLUS administered subcutaneously on Days 1 and 22 at the Center for Vaccine Development and Global Health, Baltimore, MD, USA. Participants were block randomized 1:1:1:1:1 to two doses saline placebo, two doses AGS-v PLUS, AGS-v PLUS/ISA-51 and saline placebo, two doses AGS-v PLUS/ISA-51, or two doses AGS-v PLUS/Alhydrogel. Primary endpoints were safety (all participants receiving ≥1 injection) and antibody and cytokine responses (all participants with day 43 samples), analysed by intention to treat. FINDINGS: Between 26 August 2019 and 25 February 2020, 51 participants were enrolled and randomized, 11 into the single dose AGS-v PLUS/ISA-51 group and ten in other groups. Due to COVID-19, 15 participants did not return for day 43 samplings. Participants experienced no treatment-emergent or serious adverse events. All solicited symptoms in 2/10 placebo recipients and 22/41 AGS-v PLUS recipients after dose one and 1/10 placebo recipients and 22/41 AGS-v PLUS recipients after dose two were mild/moderate except for one severe fever the day after vaccination (placebo group). Only injection site pain was more common in vaccine groups (15/51 after dose 1 and 11/51 after dose 2) versus placebo. Compared to placebo, all vaccine groups had significantly greater fold change in anti-AGS-v PLUS IgG and IFN-É£ from baseline. INTERPRETATION: AGS-v PLUS had favourable safety profile and induced robust immune responses. Next steps will determine if findings translate into clinical efficacy against mosquito-borne diseases. FUNDING: UK Department of Health and Social Care.


Subject(s)
Arbovirus Infections , Culicidae , Salivary Proteins and Peptides , Vaccines, Subunit , Adolescent , Adult , Animals , Humans , Middle Aged , Young Adult , Culicidae/immunology , Culicidae/virology , Double-Blind Method , Vaccination , Vaccines, Subunit/immunology , Arbovirus Infections/prevention & control , Salivary Proteins and Peptides/immunology
3.
Proc Natl Acad Sci U S A ; 119(35): e2209729119, 2022 08 30.
Article in English | MEDLINE | ID: mdl-35994647

ABSTRACT

Glutaminyl cyclase (QC) modifies N-terminal glutamine or glutamic acid residues of target proteins into cyclic pyroglutamic acid (pGlu). Here, we report the biochemical and functional analysis of Plasmodium QC. We show that sporozoites of QC-null mutants of rodent and human malaria parasites are recognized by the mosquito immune system and melanized when they reach the hemocoel. Detailed analyses of rodent malaria QC-null mutants showed that sporozoite numbers in salivary glands are reduced in mosquitoes infected with QC-null or QC catalytically dead mutants. This phenotype can be rescued by genetic complementation or by disrupting mosquito melanization or phagocytosis by hemocytes. Mutation of a single QC-target glutamine of the major sporozoite surface protein (circumsporozoite protein; CSP) of the rodent parasite Plasmodium berghei also results in melanization of sporozoites. These findings indicate that QC-mediated posttranslational modification of surface proteins underlies evasion of killing of sporozoites by the mosquito immune system.


Subject(s)
Aminoacyltransferases , Culicidae , Malaria , Protein Processing, Post-Translational , Sporozoites , Aminoacyltransferases/immunology , Animals , Culicidae/immunology , Glutamic Acid/metabolism , Glutamine/metabolism , Humans , Malaria/genetics , Malaria/immunology , Malaria/parasitology , Plasmodium berghei/genetics , Plasmodium berghei/immunology , Protein Processing, Post-Translational/immunology , Protozoan Proteins/immunology , Sporozoites/immunology
4.
Viruses ; 14(2)2022 02 21.
Article in English | MEDLINE | ID: mdl-35216028

ABSTRACT

Emerging and re-emerging mosquito-borne viral diseases impose a significant burden on global public health. The most common mosquito-borne viruses causing recent epidemics include flaviviruses in the family Flaviviridae, including Dengue virus (DENV), Zika virus (ZIKV), Japanese encephalitis virus (JEV) and West Nile virus (WNV) and Togaviridae viruses, such as chikungunya virus (CHIKV). Several factors may have contributed to the recent re-emergence and spread of mosquito-borne viral diseases. Among these important causes are the evolution of mosquito-borne viruses and the genetic mutations that make them more adaptive and virulent, leading to widespread epidemics. RNA viruses tend to acquire genetic diversity due to error-prone RNA-dependent RNA polymerases, thus promoting high mutation rates that support adaptation to environmental changes or host immunity. In this review, we discuss recent findings on the adaptive evolution of mosquito-borne viruses and their impact on viral infectivity, pathogenicity, vector fitness, transmissibility, epidemic potential and disease emergence.


Subject(s)
Culicidae/virology , Flavivirus/physiology , Mosquito Vectors/virology , Vector Borne Diseases/virology , Animals , Biological Evolution , Culicidae/immunology , Epidemics , Flavivirus/classification , Flavivirus/immunology , Humans , Virus Replication
5.
Infect Immun ; 90(1): e0035921, 2022 01 25.
Article in English | MEDLINE | ID: mdl-34724388

ABSTRACT

Malaria begins when an infected mosquito injects saliva containing Plasmodium sporozoites into the skin of a vertebrate host. Passive immunization of mice with antiserum against the Anopheles gambiae mosquito saliva protein TRIO (AgTRIO) offers significant protection against Plasmodium infection of mice. Furthermore, passive transfer of both AgTRIO antiserum and an anti-circumsporozoite protein monoclonal antibody provides synergistic protection. In this study, we generated monoclonal antibodies against AgTRIO to delineate the regions of AgTRIO associated with protective immunity. Monoclonal antibody 13F-1 markedly reduced Plasmodium infection in mice and recognized a region (VDDLMAKFN) in the carboxyl terminus of AgTRIO. 13F-1 is an IgG2a isotype monoclonal antibody, and the Fc region is required for protection. These data will aid in the generation of future malaria vaccines that may include both pathogen and vector antigens.


Subject(s)
Anopheles/immunology , Antibodies, Monoclonal/immunology , Culicidae/immunology , Malaria/immunology , Malaria/prevention & control , Amino Acid Sequence , Animals , Disease Models, Animal , Immunization, Passive , Immunoglobulin Fc Fragments , Insect Proteins/chemistry , Insect Proteins/immunology , Malaria/parasitology , Mice , Plasmodium berghei/immunology , Protein Binding/immunology , Protein Interaction Domains and Motifs/immunology
6.
Parasit Vectors ; 14(1): 566, 2021 Nov 03.
Article in English | MEDLINE | ID: mdl-34732254

ABSTRACT

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.


Subject(s)
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
7.
Viruses ; 13(11)2021 10 20.
Article in English | MEDLINE | ID: mdl-34834923

ABSTRACT

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.


Subject(s)
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.
Proc Natl Acad Sci U S A ; 118(44)2021 11 02.
Article in English | MEDLINE | ID: mdl-34711682

ABSTRACT

Immune priming in Anopheles gambiae is mediated by the systemic release of a hemocyte differentiation factor (HDF), a complex of lipoxin A4 bound to Evokin, a lipid carrier. HDF increases the proportion of circulating granulocytes and enhances mosquito cellular immunity. Here, we show that Evokin is present in hemocytes and fat-body cells, and messenger RNA (mRNA) expression increases significantly after immune priming. The double peroxidase (DBLOX) enzyme, present in insects but not in vertebrates, is essential for HDF synthesis. DBLOX is highly expressed in oenocytes in the fat-body tissue, and these cells increase in number in primed mosquitoes. We provide direct evidence that the histone acetyltransferase AgTip60 (AGAP001539) is also essential for a sustained increase in oenocyte numbers, HDF synthesis, and immune priming. We propose that oenocytes may function as a population of cells that are reprogrammed, and orchestrate and maintain a broad, systemic, and long-lasting state of enhanced immune surveillance in primed mosquitoes.


Subject(s)
Culicidae/immunology , Histone Acetyltransferases/metabolism , Immunologic Memory/immunology , Animals , Anopheles/immunology , Anopheles/metabolism , Culicidae/metabolism , Female , Granulocytes/metabolism , Hemocytes/immunology , Immunity, Innate/immunology , Insect Proteins/genetics , Insecta/metabolism , Lipoxins/metabolism , Malaria/immunology , Male , Peroxidase/metabolism , Plasmodium/metabolism , Plasmodium berghei/metabolism
9.
Future Microbiol ; 16: 657-670, 2021 06.
Article in English | MEDLINE | ID: mdl-34100305

ABSTRACT

Drawing of host blood is a natural phenomenon during the bite of blood-probing insect vectors. Along with the blood meal, the vectors introduce salivary components and a trail of microbiota. In the case of infected vectors, the related pathogen accompanies the aforementioned biological components. In addition to Anopheles gambiae or Anopheles stephensi, the bites of other nonmalarial vectors cannot be ignored in malaria-endemic regions. Similarly, the bite incidence of Phlebotomus papatasi cannot be ignored in visceral leishmaniasis-endemic regions. Even the chances of getting bitten by uninfected vectors are higher than the infected vectors. We have discussed the probability or possibility of uninfected, infected, and/or nonvector's saliva and gut microbiota as a therapeutic option leading to the initial deterrent to pathogen establishment.


Subject(s)
Gastrointestinal Microbiome/immunology , Insect Vectors , Saliva/immunology , Animals , Culicidae/immunology , Humans , Immunomodulation , Insect Bites and Stings/immunology , Insect Bites and Stings/prevention & control , Insect Vectors/immunology , Psychodidae/immunology , Vector Borne Diseases/immunology , Vector Borne Diseases/prevention & control
10.
Viruses ; 13(5)2021 04 24.
Article in English | MEDLINE | ID: mdl-33923307

ABSTRACT

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.


Subject(s)
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
11.
Dev Comp Immunol ; 119: 104010, 2021 06.
Article in English | MEDLINE | ID: mdl-33476667

ABSTRACT

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.


Subject(s)
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
12.
Dev Comp Immunol ; 116: 103964, 2021 03.
Article in English | MEDLINE | ID: mdl-33301792

ABSTRACT

Mosquitoes are vectors of a large number of viral pathogens. In recent years, increased urbanization and climate change has expanded the range of many vector mosquitoes. The lack of effective medical interventions has made the control of mosquito-borne viral diseases very difficult. Understanding the interactions between the mosquito immune system and viruses is critical if we are to develop effective control strategies against these diseases. Mosquitoes harbor multiple conserved immune pathways that curb invading viral pathogens. Despite the conservation of these pathways, the activation and intensity of the mosquito immune response varies with the mosquito species, tissue, and the infecting virus. This article reviews major conserved antiviral immune pathways in vector mosquitoes, their interactions with invading viral pathogens, and how these interactions restrict or promote infection of these medically important viruses.


Subject(s)
Culicidae/immunology , Mosquito Vectors/immunology , Signal Transduction/immunology , Animals , Animals, Genetically Modified/genetics , Animals, Genetically Modified/immunology , Animals, Genetically Modified/virology , Antiviral Agents/immunology , Arboviruses/physiology , Carrier Proteins/immunology , Culicidae/genetics , Culicidae/virology , Janus Kinases/immunology , Mitogen-Activated Protein Kinases , Mosquito Vectors/genetics , Mosquito Vectors/virology , RNA Interference/immunology , STAT Transcription Factors/immunology , Toll-Like Receptors/immunology
14.
Trends Parasitol ; 36(11): 880-883, 2020 11.
Article in English | MEDLINE | ID: mdl-33036937

ABSTRACT

Malaria eradication is a global priority but requires innovative strategies. Humoral immune responses attack different parasite stages, and antibody-based therapy may prevent malaria infection or transmission. Here, we discuss targets of monoclonal antibodies in mosquito sexual stages of Plasmodium.


Subject(s)
Antibodies, Monoclonal/immunology , Culicidae/parasitology , Life Cycle Stages/immunology , Malaria/prevention & control , Malaria/transmission , Plasmodium falciparum/immunology , Animals , Culicidae/immunology , Disease Eradication , Humans , Malaria/parasitology
15.
Cell Host Microbe ; 27(3): 329-344, 2020 Mar 11.
Article in English | MEDLINE | ID: mdl-32164844

ABSTRACT

The intestine is an essential physical and immunological barrier comprised of a monolayer of diverse and specialized epithelial cells that perform functions ranging from nutrient absorption to pathogen sensing and intestinal homeostasis. The intestinal barrier prevents translocation of intestinal microbes into internal compartments. The microbiota is comprised of a complex community largely populated by diverse bacterial species that provide metabolites, nutrients, and immune stimuli that promote intestinal and organismal health. Although commensal organisms promote health, enteric pathogens, including a diverse plethora of enteric viruses, cause acute and chronic diseases. The barrier epithelium plays fundamental roles in immune defenses against enteric viral infections by integrating diverse signals, including those from the microbiota, to prevent disease. Importantly, many model systems have contributed to our understanding of this complex interface. This review will focus on the antiviral mechanisms at play within the intestinal epithelium and how these responses are shaped by the microbiota.


Subject(s)
Gastrointestinal Microbiome , Intestinal Diseases/virology , Intestinal Mucosa/microbiology , Virus Diseases/pathology , Animals , Caenorhabditis elegans/microbiology , Caenorhabditis elegans/virology , Culicidae/immunology , Culicidae/virology , Drosophila melanogaster/immunology , Drosophila melanogaster/virology , Humans , Immunity, Innate , Mice , Virus Diseases/microbiology
16.
Yi Chuan ; 42(2): 153-160, 2020 Feb 20.
Article in English | MEDLINE | ID: mdl-32102772

ABSTRACT

Mosquito-borne diseases have become an important public health issue of global concern because of their high incidence and transmission rate. As a vector for mosquito-borne diseases, studying the interaction mechanism between mosquitoes and mosquito-borne viruses will help control mosquito-borne diseases. The impaired innate immunity and immune barriers evasion caused by mosquito-borne viruses in mosquitoes pose a potential risk for the persistent infection of the virus in mosquitoes and the outbreak of mosquito-borne diseases. The RNA interference (RNAi) pathway, as a powerful antiviral defense barrier in mosquitoes, can inhibit viral replication and transmission by producing a variety of small RNAs to degrade viral RNA. In this review, we summarize the related studies on the innate immune mechanism against mosquito- borne virus infection in mosquitoes about small interfering RNA (siRNA), microRNA (miRNA), and Piwi-interacting RNA (piRNA), aiming to provide a theoretical reference for the prevention and control of mosquito-borne diseases.


Subject(s)
Culicidae/virology , RNA Interference , Virus Diseases , Animals , Culicidae/immunology , Immunity, Innate , Mosquito Vectors/immunology , Mosquito Vectors/virology , RNA, Small Interfering , Virus Diseases/prevention & control , Virus Diseases/transmission
17.
Curr Protein Pept Sci ; 21(2): 153-158, 2020.
Article in English | MEDLINE | ID: mdl-31362670

ABSTRACT

An allergic reaction to mosquitoes can result in severe or abnormal local or systemic reactions such as anaphylaxis, angioedema, and general urticarial or wheezing. The aim of this review is to provide information on mosquito saliva allergens that can support the production of highly specific recombinant saliva allergens. In particular, candidate allergens of mosquitoes that are well suited to the ecology of mosquitoes that occur mainly in East Asia will be identified and introduced. By doing so, the diagnosis and treatment of patients with severe sensitivity to mosquito allergy will be improved by predicting the characteristics of East Asian mosquito allergy, presenting the future direction of production of recombinant allergens, and understanding the difference between East and West.


Subject(s)
Allergens/immunology , Culicidae/immunology , Saliva/immunology , Animals , Asia, Eastern , Humans
19.
Dev Comp Immunol ; 103: 103458, 2020 02.
Article in English | MEDLINE | ID: mdl-31377103

ABSTRACT

Diseases spread by mosquitoes have killed more people than those spread by any other group of arthropod vectors and remain an important factor in determining global health and economic stability. The mosquito innate immune system can act to either modulate infection with human pathogens or fight off entomopathogens and increase the fitness and longevity of infected mosquitoes. While work remains towards understanding the larval immune system and the development of the mosquito immune system, it has recently become clearer that environmental factors heavily shape the developing mosquito immune system and continue to influence the adult immune system as well. The adult immune system has been well-studied and is known to involve multiple tissues and diverse molecular mechanisms. This review summarizes and synthesizes what is currently understood about the development of the mosquito immune system and includes comparisons of immune components unique to mosquitoes among the blood-feeding arthropods as well as important distinguishing factors between the anopheline and culicine mosquitoes. An explanation is included for how mosquito immunity factors into vector competence and vectorial capacity is presented along with a model for the interrelationships between nutrition, microbiome, pathogen interactions and behavior as they relate to mosquito development, immune status, adult female fitness and ultimately, vectorial capacity. Novel discoveries in the fields of mosquito ecoimmunology, neuroimmunology, and intracellular antiviral responses are highlighted.


Subject(s)
Culicidae/immunology , Animals , Host-Pathogen Interactions/immunology , Humans , Mosquito Vectors/immunology
20.
Insect Biochem Mol Biol ; 111: 103182, 2019 08.
Article in English | MEDLINE | ID: mdl-31265904

ABSTRACT

The mosquito immune system has evolved in the presence of continuous encounters with fungi that range from food to foes. Herein, we review the field of mosquito-fungal interactions, providing an overview of current knowledge and topics of interest. Mosquitoes encounter fungi in their aquatic and terrestrial habitats. Mosquito larvae are exposed to fungi on plant detritus, within the water column, and at the water surface. Adult mosquitoes are exposed to fungi during indoor and outdoor resting, blood and sugar feeding, mating, and oviposition. Fungi enter the mosquito body through different routes, including ingestion and through active or passive breaches in the cuticle. Oral uptake of fungi can be beneficial to mosquitoes, as yeasts hold nutritional value and support larval development. However, ingestion of or surface contact with fungal entomopathogens leads to colonization of the mosquito with often lethal consequences to the host. The mosquito immune system recognizes fungi and mounts cellular and humoral immune responses in the hemocoel, and possibly epithelial immune responses in the gut. These responses are regulated transcriptionally through multiple signal transduction pathways. Proteolytic protease cascades provide additional regulation of antifungal immunity. Together, these immune responses provide an efficient barrier to fungal infections, which need to be overcome by entomopathogens. Therefore, fungi constitute an excellent tool to examine the molecular underpinnings of mosquito immunity and to identify novel antifungal peptides. In addition, recent advances in mycobiome analyses can now be used to examine the contribution of fungi to various mosquito traits, including vector competence.


Subject(s)
Culicidae/immunology , Culicidae/microbiology , Fungi/physiology , Animals , Ecosystem , Fungi/pathogenicity , Host-Pathogen Interactions , Mycobiome
SELECTION OF CITATIONS
SEARCH DETAIL
...