Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 20
Filtrar
1.
bioRxiv ; 2024 May 05.
Artigo em Inglês | MEDLINE | ID: mdl-38746363

RESUMO

Tumor Necrosis Factor-α (TNF-α) is a proinflammatory cytokine and a master regulator of immune cell function in vertebrates. While previous studies have implicated TNF signaling in invertebrate immunity, the roles of TNF in mosquito innate immunity and vector competence have yet to be explored. Herein, we confirm the identification of a conserved TNF-α pathway in Anopheles gambiae consisting of the TNF-α ligand, Eiger, and its cognate receptors Wengen and Grindelwald. Through gene expression analysis, RNAi, and in vivo injection of recombinant TNF-α, we provide direct evidence for the requirement of TNF signaling in regulating mosquito immune cell function by promoting granulocyte midgut attachment, increased granulocyte abundance, and oenocytoid rupture. Moreover, our data demonstrate that TNF signaling is an integral component of anti-Plasmodium immunity that limits malaria parasite survival. Together, our data support the existence of a highly conserved TNF signaling pathway in mosquitoes that mediates cellular immunity and influences Plasmodium infection outcomes, offering potential new approaches to interfere with malaria transmission by targeting the mosquito host.

2.
bioRxiv ; 2024 Apr 04.
Artigo em Inglês | MEDLINE | ID: mdl-38617257

RESUMO

Mosquito-borne viruses cause more than 400 million annual infections and place over half of the world's population at risk. Despite this importance, the mechanisms by which arboviruses infect the mosquito host and disseminate to tissues required for transmission are not well understood. Here, we provide evidence that mosquito immune cells, known as hemocytes, play an integral role in the dissemination of dengue virus (DENV) and Zika virus (ZIKV) in the mosquito Aedes aegypti. We establish that phagocytic hemocytes are a focal point for virus infection and demonstrate that these immune cell populations facilitate virus dissemination to the ovaries and salivary glands. Additional transfer experiments confirm that virus-infected hemocytes confer a virus infection to non-infected mosquitoes more efficiently than free virus in acellular hemolymph, revealing that hemocytes are an important tropism to enhance virus dissemination in the mosquito host. These data support a "trojan horse" model of virus dissemination where infected hemocytes transport virus through the hemolymph to deliver virus to mosquito tissues required for transmission and parallels vertebrate systems where immune cell populations promote virus dissemination to secondary sites of infection. In summary, this study significantly advances our understanding of virus infection dynamics in mosquitoes and highlights conserved roles of immune cells in virus dissemination across vertebrate and invertebrate systems.

3.
Sci Rep ; 13(1): 8778, 2023 05 31.
Artigo em Inglês | MEDLINE | ID: mdl-37258694

RESUMO

Vector-borne, filarial nematode diseases cause significant disease burdens in humans and domestic animals worldwide. Although there is strong direct evidence of parasite-driven immunomodulation of mammalian host responses, there is less evidence of parasite immunomodulation of the vector host. We have previously reported that all life stages of Brugia malayi, a filarial nematode and causative agent of Lymphatic filariasis, secrete extracellular vesicles (EVs). Here we investigate the immunomodulatory effects of microfilariae-derived EVs on the vector host Aedes aegypti. RNA-seq analysis of an Ae. aegypti cell line treated with B. malayi microfilariae EVs showed differential expression of both mRNAs and miRNAs. AAEL002590, an Ae. aegypti gene encoding a serine protease, was shown to be downregulated when cells were treated with biologically relevant EV concentrations in vitro. Injection of adult female mosquitoes with biologically relevant concentrations of EVs validated these results in vivo, recapitulating the downregulation of AAEL002590 transcript. This gene was predicted to be involved in the mosquito phenoloxidase (PO) cascade leading to the canonical melanization response and correspondingly, both suppression of this gene using RNAi and parasite EV treatment reduced PO activity in vivo. Our data indicate that parasite-derived EVs interfere with critical immune responses in the vector host, including melanization.


Assuntos
Aedes , Brugia Malayi , Vesículas Extracelulares , Animais , Humanos , Feminino , Brugia Malayi/genética , Microfilárias/genética , Mosquitos Vetores , Mamíferos
5.
Front Immunol ; 12: 680020, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34484178

RESUMO

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.


Assuntos
Anopheles/imunologia , Anopheles/microbiologia , Anopheles/parasitologia , Dinoprostona/metabolismo , Oocistos/imunologia , Plasmodium/imunologia , Transdução de Sinais , Animais , Anopheles/classificação , Hemócitos/metabolismo , Interações Hospedeiro-Patógeno/imunologia , Imunidade Inata , Viabilidade Microbiana , Mosquitos Vetores/imunologia , Mosquitos Vetores/microbiologia , Mosquitos Vetores/parasitologia , Filogenia , Plasmodium/crescimento & desenvolvimento , Proteínas Citotóxicas Formadoras de Poros/metabolismo , Receptores de Prostaglandina E/genética , Receptores de Prostaglandina E/metabolismo
6.
Elife ; 102021 07 28.
Artigo em Inglês | MEDLINE | ID: mdl-34318744

RESUMO

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.


Assuntos
Anopheles/imunologia , Diferenciação Celular/imunologia , Hemócitos/imunologia , Mosquitos Vetores/imunologia , Fagócitos/imunologia , Animais , Anopheles/genética , Drosophila , Feminino , Inativação Gênica , Hemócitos/metabolismo , Imunidade Inata , Malária/imunologia , Malária/parasitologia , Mosquitos Vetores/genética , Fagócitos/metabolismo , Análise de Sequência de RNA , Análise de Célula Única
7.
mSphere ; 6(2)2021 03 31.
Artigo em Inglês | MEDLINE | ID: mdl-33789941

RESUMO

Mosquitoes may feed multiple times during their life span in addition to those times needed to acquire and transmit malaria. To determine the impact of subsequent blood feeding on parasite development in Anopheles gambiae, we examined Plasmodium parasite infection with or without an additional noninfected blood meal. We found that an additional blood meal significantly reduced Plasmodium berghei immature oocyst numbers, yet had no effect on the human parasite Plasmodium falciparum These observations were reproduced when mosquitoes were fed an artificial protein meal, suggesting that parasite losses are independent of blood ingestion. We found that feeding with either a blood or protein meal compromises midgut basal lamina integrity as a result of the physical distention of the midgut, enabling the recognition and lysis of immature P. berghei oocysts by mosquito complement. Moreover, we demonstrate that additional feeding promotes P. falciparum oocyst growth, suggesting that human malaria parasites exploit host resources provided with blood feeding to accelerate their growth. This is in contrast to experiments with P. berghei, where the size of surviving oocysts is independent of an additional blood meal. Together, these data demonstrate distinct differences in Plasmodium species in evading immune detection and utilizing host resources at the oocyst stage, representing an additional, yet unexplored component of vectorial capacity that has important implications for the transmission of malaria.IMPORTANCE Mosquitoes must blood feed multiple times to acquire and transmit malaria. However, the impact of an additional mosquito blood meal following malaria parasite infection has not been closely examined. Here, we demonstrate that additional feeding affects mosquito vector competence; namely, additional feeding significantly limits Plasmodium berghei infection, yet has no effect on infection of the human parasite P. falciparum Our experiments support that these killing responses are mediated by the physical distension of the midgut and by temporary damage to the midgut basal lamina that exposes immature P. berghei oocysts to mosquito complement, while human malaria parasites are able to evade these killing mechanisms. In addition, we provide evidence that additional feeding promotes P. falciparum oocyst growth. This is in contrast to P. berghei, where oocyst size is independent of an additional blood meal. This suggests that human malaria parasites are able to exploit host resources provided by an additional feeding to accelerate their growth. In summary, our data highlight distinct differences in malaria parasite species in evading immune recognition and adapting to mosquito blood feeding. These observations have important, yet previously unexplored, implications for the impact of multiple blood meals on the transmission of malaria.


Assuntos
Anopheles/parasitologia , Comportamento Alimentar , Interações Hospedeiro-Parasita , Plasmodium/crescimento & desenvolvimento , Plasmodium/imunologia , Animais , Anopheles/fisiologia , Sangue , Feminino , Evasão da Resposta Imune , Malária/parasitologia , Malária/transmissão , Refeições , Camundongos , Mosquitos Vetores/parasitologia , Oocistos/crescimento & desenvolvimento , Oocistos/imunologia , Plasmodium/classificação , Plasmodium berghei/crescimento & desenvolvimento , Plasmodium berghei/imunologia , Plasmodium falciparum/crescimento & desenvolvimento , Plasmodium falciparum/imunologia
8.
Front Cell Dev Biol ; 9: 627976, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33604338

RESUMO

The innate immune system is the primary defense response to limit invading pathogens for all invertebrate species. In insects, immune cells are central to both cellular and humoral immune responses, however few genetic resources exist beyond Drosophila to study immune cell function. Therefore, the development of innovative tools that can be widely applied to a variety of insect systems is of importance to advance the study of insect immunity. Here, we have adapted the use of clodronate liposomes (CLD) to deplete phagocytic immune cells in the vinegar fly, Drosophila melanogaster, and the yellow fever mosquito, Aedes aegypti. Through microscopy and molecular techniques, we validate the depletion of phagocytic cell populations in both insect species and demonstrate the integral role of phagocytes in combating bacterial pathogens. Together, these data demonstrate the wide utility of CLD in insect systems to advance the study of phagocyte function in insect innate immunity.

9.
Front Physiol ; 12: 821869, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-35140633

RESUMO

Characterizing the physiological changes that accompany malaria parasite infection of the mosquito host is crucial to our understanding of vectorial capacity in Anopheles mosquitoes, yet has not fully been explored. In this study, we examine the role of uric acid metabolism in the mosquito, Anopheles gambiae, following malaria parasite infection. We demonstrate that levels of uric acid are significantly decreased in the excreta and the mosquito at 24 and 48 h post-Plasmodium infection when compared to controls fed on naïve mouse blood. When we examine the expression of well-known enzymes responsible for uric acid metabolism, we see a significant increase in both urate oxidase (UO) and allatoicase (ALLC) expression following Plasmodium infection. Targeting the essential first step in uric acid metabolism by silencing UO resulted in elevated levels of uric acid, enhancing malaria parasite survival. With implications from other insect systems that bacteria can modulate UO expression, we examined the possibility that the mosquito microbiota and its expansion following blood-feeding may contribute to increased UO levels. However, there was no difference in uric acid metabolism between septic and aseptic mosquitoes, indicating that the mosquito microbiome is not associated with the manipulation of UO expression. Together, our study provides new evidence that Plasmodium infection causes the mosquito host to actively metabolize uric acid by increasing UO expression to limit Plasmodium oocyst survival, suggesting that nitrogen metabolism is an essential pathway in defining mosquito vector competence.

10.
Sci Rep ; 10(1): 21084, 2020 12 03.
Artigo em Inglês | MEDLINE | ID: mdl-33273588

RESUMO

Mosquito physiology and immunity are integral determinants of malaria vector competence. This includes the principal role of hormonal signaling in Anopheles gambiae initiated shortly after blood-feeding, which stimulates immune induction and promotes vitellogenesis through the function of 20-hydroxyecdysone (20E). Previous studies demonstrated that manipulating 20E signaling through the direct injection of 20E or the application of a 20E agonist can significantly impact Plasmodium infection outcomes, reducing oocyst numbers and the potential for malaria transmission. In support of these findings, we demonstrate that a 20E agonist, halofenozide, is able to induce anti-Plasmodium immune responses that limit Plasmodium ookinetes. We demonstrate that halofenozide requires the function of ultraspiracle (USP), a component of the canonical heterodimeric ecdysone receptor, to induce malaria parasite killing responses. Additional experiments suggest that the effects of halofenozide treatment are temporal, such that its application only limits malaria parasites when applied prior to infection. Unlike 20E, halofenozide does not influence cellular immune function or AMP production. Together, our results further demonstrate the potential of targeting 20E signaling pathways to reduce malaria parasite infection in the mosquito vector and provide new insight into the mechanisms of halofenozide-mediated immune activation that differ from 20E.


Assuntos
Anopheles/efeitos dos fármacos , Benzoatos/farmacologia , Interações Hospedeiro-Parasita , Hidrazinas/farmacologia , Inseticidas/farmacologia , Plasmodium berghei/patogenicidade , Animais , Anopheles/imunologia , Anopheles/parasitologia , Células Cultivadas , Ecdisterona/agonistas , Feminino , Masculino , Fagocitose , Receptores de Esteroides/metabolismo
11.
mSphere ; 5(2)2020 04 15.
Artigo em Inglês | MEDLINE | ID: mdl-32295874

RESUMO

Blood feeding is an integral behavior of mosquitoes to acquire nutritional resources needed for reproduction. This requirement also enables mosquitoes to serve as efficient vectors to acquire and potentially transmit a multitude of mosquito-borne diseases, most notably malaria. Recent studies suggest that mosquito immunity is stimulated following a blood meal, independent of infection status. Since blood feeding promotes production of the hormone 20-hydroxyecdysone (20E), we hypothesized that 20E plays an important role in priming the immune response for pathogen challenge. Here, we examine the immunological effects of priming Anopheles gambiae with 20E prior to pathogen infection, demonstrating a significant reduction in bacteria and Plasmodium berghei survival in the mosquito host. Transcriptome sequencing (RNA-seq) analysis following 20E treatment identifies several known 20E-regulated genes, as well as several immune genes with previously reported function in antipathogen defense. Together, these data demonstrate that 20E influences cellular immune function and antipathogen immunity following mosquito blood feeding, arguing the importance of hormones in the regulation of mosquito innate immune function.IMPORTANCE Blood feeding is required to provide nutrients for mosquito egg production and serves as a mechanism to acquire and transmit pathogens. Shortly after a blood meal is taken, there is a peak in the production of 20-hydroxyecdysone (20E), a mosquito hormone that initiates physiological changes, including yolk protein production and mating refractoriness. Here, we examine additional roles of 20E in the regulation of mosquito immunity, demonstrating that priming the immune system with 20E increases mosquito resistance to pathogens. We identify differentially expressed genes in response to 20E treatment, including several involved in innate immune function as well as lipid metabolism and transport. Together, these data argue that 20E stimulates mosquito cellular immune function and innate immunity shortly after blood feeding.


Assuntos
Anopheles/efeitos dos fármacos , Anopheles/imunologia , Bactérias , Ecdisterona/imunologia , Imunidade Inata , Plasmodium berghei/fisiologia , Animais , Anopheles/microbiologia , Anopheles/parasitologia , Ecdisterona/administração & dosagem , Comportamento Alimentar , Feminino , Imunidade Celular , Mosquitos Vetores
12.
Insect Biochem Mol Biol ; 117: 103290, 2020 02.
Artigo em Inglês | MEDLINE | ID: mdl-31790798

RESUMO

In arthropods, eicosanoids derived from the oxygenated metabolism of arachidonic acid are significant in mediating immune responses. However, the lack of information about insect eicosanoid receptors is an obstacle to completely decipher immune mechanisms underlying both eicosanoid downstream signal cascades and their relationship to immune pathogen-associated molecular patterns (PAMPs). Here, we cloned and sequenced a G protein-coupled receptor (MW 46.16 kDa) from the model lepidopteran, Manduca sexta (Sphingidae). The receptor shares similarity of amino acid motifs to human prostaglandin E2 (PGE2) receptors, and phylogenetic analysis supports its classification as a prostaglandin receptor. In agreement, the recombinant receptor was activated by PGE2 resulting in intracellular cAMP increase, and therefore designated MansePGE2R. Expression of MansePGE2R in Sf9 cells in which the endogenous orthologous receptor had been silenced showed similar cAMP increase upon PGE2 challenge. Receptor transcript expression was identified in various tissues in larvae and female adults, including Malpighian tubules, fat body, gut and hemocytes, and in female ovaries. In addition to the cDNA cloned that encodes the functional receptor, an mRNA was found featuring the poly-A tail but lacking the predicted transmembrane (TM) regions 2 and 3, suggesting the possibility that internally deleted receptor proteins exist in insects. Immunocytochemistry and in situ hybridization revealed that among hemocytes, the receptor was exclusively localized in the oenocytoids. Larval immune challenges injecting bacterial components showed that lipoteichoic acid (LTA) increased MansePGE2R expression in hemocytes. In contrast, injection of LPS or peptidoglycan did not increase MansePGE2R transcript levels in hemocytes, suggesting the LTA-associated increase in receptor transcript is regulated through a distinct pathway. This study provides the first characterization of an eicosanoid receptor in insects, and paves the way for establishing the hierarchy in signaling steps required for establishing insect immune responses to infections.


Assuntos
Expressão Gênica , Proteínas de Insetos/genética , Lipopolissacarídeos/metabolismo , Manduca/genética , Receptores de Prostaglandina E Subtipo EP2/genética , Ácidos Teicoicos/metabolismo , Sequência de Aminoácidos , Animais , Sequência de Bases , Feminino , Regulação da Expressão Gênica , Hemócitos/metabolismo , Proteínas de Insetos/química , Proteínas de Insetos/metabolismo , Larva/genética , Larva/metabolismo , Manduca/metabolismo , Filogenia , Receptores de Prostaglandina E Subtipo EP2/química , Receptores de Prostaglandina E Subtipo EP2/metabolismo , Alinhamento de Sequência
13.
Insects ; 10(10)2019 Sep 20.
Artigo em Inglês | MEDLINE | ID: mdl-31547026

RESUMO

Eicosanoids are bioactive signaling lipids derived from the oxidation of fatty acids that act as important regulators of immune homeostasis and inflammation. As a result, effective anti-inflammatory drugs have been widely used to reduce pain and inflammation which target key eicosanoid biosynthesis enzymes. Conserved from vertebrates to insects, the use of these eicosanoid pathway inhibitors offer opportunities to evaluate the roles of eicosanoids in less-characterized insect systems. In this study, we examine the potential roles of eicosanoids on malaria parasite survival in the mosquito Anopheles gambiae. Using Plasmodium oocyst numbers to evaluate parasite infection, general or specific inhibitors of eicosanoid biosynthesis pathways were evaluated. Following the administration of dexamethasone and indomethacin, respective inhibitors of phospholipid A2 (PLA2) and cyclooxygenase (COX), oocyst numbers were unaffected. However, inhibition of lipoxygenase (LOX) activity through the use of esculetin significantly increased oocyst survival. In contrast, 12-[[(tricyclo[3.3.1.13,7]dec-1-ylamino)carbonyl]amino]-dodecanoic acid (AUDA), an inhibitor of epoxide hydroxylase (EH), decreased oocyst numbers. These experiments were further validated through RNAi experiments to silence candidate genes homologous to EH in An. gambiae to confirm their contributions to Plasmodium development. Similar to the results of AUDA treatment, the silencing of EH significantly reduced oocyst numbers. These results imply that specific eicosanoids in An. gambiae can have either agonist or antagonistic roles on malaria parasite survival in the mosquito host.

14.
Proc Natl Acad Sci U S A ; 116(28): 14119-14128, 2019 07 09.
Artigo em Inglês | MEDLINE | ID: mdl-31235594

RESUMO

Mosquito immunity is composed of both cellular and humoral factors that provide protection from invading pathogens. Immune cells known as hemocytes, have been intricately associated with phagocytosis and innate immune signaling. However, the lack of genetic tools has limited hemocyte study despite their importance in mosquito anti-Plasmodium immunity. To address these limitations, we employ the use of a chemical-based treatment to deplete phagocytic immune cells in Anopheles gambiae, demonstrating the role of phagocytes in complement recognition and prophenoloxidase production that limit the ookinete and oocyst stages of malaria parasite development, respectively. Through these experiments, we also define specific subtypes of phagocytic immune cells in An. gambiae, providing insights beyond the morphological characteristics that traditionally define mosquito hemocyte populations. Together, this study represents a significant advancement in our understanding of the roles of mosquito phagocytes in mosquito vector competence and demonstrates the utility of clodronate liposomes as an important tool in the study of invertebrate immunity.


Assuntos
Anopheles/imunologia , Imunidade Inata , Malária Falciparum/imunologia , Fagocitose/imunologia , Animais , Anopheles/genética , Anopheles/parasitologia , Catecol Oxidase/genética , Ácido Clodrônico/farmacologia , Proteínas do Sistema Complemento/imunologia , Precursores Enzimáticos/genética , Hemócitos/efeitos dos fármacos , Hemócitos/imunologia , Hemócitos/parasitologia , Humanos , Lipossomos/farmacologia , Malária Falciparum/tratamento farmacológico , Malária Falciparum/parasitologia , Mosquitos Vetores/imunologia , Mosquitos Vetores/parasitologia , Oocistos/imunologia , Fagócitos/efeitos dos fármacos , Fagócitos/imunologia , Fagócitos/parasitologia , Fagocitose/efeitos dos fármacos
15.
Parasit Vectors ; 10(1): 369, 2017 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-28764765

RESUMO

BACKGROUND: There is emerging evidence that mosquito anti-Plasmodium immunity is multimodal with distinct mechanisms for killing malaria parasites at either the ookinete or oocyst stages. Early-phase responses targeting the ookinete require complement-like components circulating in the mosquito hemolymph that result in TEP1-mediated lysis or melanization. Additional responses mediated by the LL3 and STAT pathways limit oocyst survival through unknown mechanisms that require mosquito hemocyte function. While previous experiments argue that these mechanisms of parasite killing are independent, the transient nature of gene-silencing has rendered these experiments inconclusive. To address this issue, we outline experiments using a TALEN-derived TEP1 mutant line to examine the role of TEP1 in the Anopheles gambiae late-phase immune response. RESULTS: Despite higher early oocyst numbers in the TEP1 mutant line, no differences in oocyst survival were observed when compared to control mosquitoes, suggesting that TEP1 function is independent of the late-phase immune response. To further validate this phenotype in the TEP1 mutant, oocyst survival was evaluated in the TEP1 mutant background by silencing either LL3 or STAT-A. Surprisingly, only STAT-A silenced mosquitoes were able to reconstitute the late-phase immune phenotype increasing oocyst survival in the TEP1 mutant line. Additional experiments highlight significant differences in LL3 expression in the M/S hybrid genetic background of the TEP1 mutant line compared to that of the Keele strain (M form) of An. gambiae, and demonstrate that LL3 is not required for granulocyte differentiation in the M/S hybrid G3 genetic background in response to malaria parasite infection. CONCLUSIONS: Through the combination of genetic experiments utilizing genetic mutants and reverse genetic approaches, new information has emerged regarding the mechanisms of mosquito late-phase immunity. When combined with previously published experiments, the body of evidence argues that Plasmodium oocyst survival is TEP1 independent, thus establishing that the mechanisms of early- and late-phase immunity are distinct. Moreover, we identify that the known components that mediate oocyst survival are susceptible to strain-specific differences depending on their genetic background and provide further evidence that the signals that promote hemocyte differentiation are required to limit oocyst survival. Together, this study provides new insights into the mechanisms of oocyst killing and the importance of genetics in shaping mosquito vector competence.


Assuntos
Anopheles/imunologia , Anopheles/parasitologia , Proteínas de Insetos/metabolismo , Mosquitos Vetores/imunologia , Oocistos/fisiologia , Animais , Anopheles/genética , Antígenos de Protozoários/imunologia , Proteínas do Sistema Complemento , Inativação Gênica , Hemócitos/fisiologia , Hemolinfa/imunologia , Hemolinfa/parasitologia , Imunidade Inata , Proteínas de Insetos/genética , Malária/parasitologia , Malária/transmissão , Mosquitos Vetores/genética , Mosquitos Vetores/parasitologia , Mutação , Oocistos/imunologia , Proteínas de Protozoários/imunologia , Interferência de RNA , Genética Reversa , Fatores de Transcrição STAT/metabolismo
16.
Proc Natl Acad Sci U S A ; 113(25): 6880-5, 2016 06 21.
Artigo em Inglês | MEDLINE | ID: mdl-27274056

RESUMO

Insect kinins (leucokinins) are multifunctional peptides acting as neurohormones and neurotransmitters. In females of the mosquito vector Aedes aegypti (L.), aedeskinins are known to stimulate fluid secretion from the renal organs (Malpighian tubules) and hindgut contractions by activating a G protein-coupled kinin receptor designated "Aedae-KR." We used protease-resistant kinin analogs 1728, 1729, and 1460 to evaluate their effects on sucrose perception and feeding behavior. In no-choice feeding bioassays (capillary feeder and plate assays), the analog 1728, which contains α-amino isobutyric acid, inhibited females from feeding on sucrose. It further induced quick fly-away or walk-away behavior following contact with the tarsi and the mouthparts. Electrophysiological recordings from single long labellar sensilla of the proboscis demonstrated that mixing the analog 1728 at 1 mM with sucrose almost completely inhibited the detection of sucrose. Aedae-KR was immunolocalized in contact chemosensory neurons in prothoracic tarsi and in sensory neurons and accessory cells of long labellar sensilla in the distal labellum. Silencing Aedae-KR by RNAi significantly reduced gene expression and eliminated the feeding-aversion behavior resulting from contact with the analog 1728, thus directly implicating the Aedae-KR in the aversion response. To our knowledge, this is the first report that kinin analogs modulate sucrose perception in any insect. The aversion to feeding elicited by analog 1728 suggests that synthetic molecules targeting the mosquito Aedae-KR in the labellum and tarsi should be investigated for the potential to discover novel feeding deterrents of mosquito vectors.


Assuntos
Aedes/fisiologia , Cininas/farmacologia , Mimetismo Molecular , Neurônios/fisiologia , Sacarose , Paladar , Animais , Clonagem Molecular , DNA Complementar , Feminino , Humanos , Cininas/química , Masculino , Microscopia Confocal
17.
Nat Commun ; 7: 10507, 2016 Feb 09.
Artigo em Inglês | MEDLINE | ID: mdl-26856261

RESUMO

Ticks transmit more pathogens to humans and animals than any other arthropod. We describe the 2.1 Gbp nuclear genome of the tick, Ixodes scapularis (Say), which vectors pathogens that cause Lyme disease, human granulocytic anaplasmosis, babesiosis and other diseases. The large genome reflects accumulation of repetitive DNA, new lineages of retro-transposons, and gene architecture patterns resembling ancient metazoans rather than pancrustaceans. Annotation of scaffolds representing ∼57% of the genome, reveals 20,486 protein-coding genes and expansions of gene families associated with tick-host interactions. We report insights from genome analyses into parasitic processes unique to ticks, including host 'questing', prolonged feeding, cuticle synthesis, blood meal concentration, novel methods of haemoglobin digestion, haem detoxification, vitellogenesis and prolonged off-host survival. We identify proteins associated with the agent of human granulocytic anaplasmosis, an emerging disease, and the encephalitis-causing Langat virus, and a population structure correlated to life-history traits and transmission of the Lyme disease agent.


Assuntos
Anaplasma phagocytophilum , Vetores Aracnídeos/genética , Genoma/genética , Ixodes/genética , Canais Iônicos de Abertura Ativada por Ligante/genética , Animais , Perfilação da Expressão Gênica , Genômica , Doença de Lyme/transmissão , Oócitos , Xenopus laevis
18.
Insect Biochem Mol Biol ; 43(7): 588-93, 2013 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-23523607

RESUMO

In anautogenous mosquitoes such as Aedes aegypti females the calcitonin-like diuretic hormone 31 (DH31) stimulates natriuretic fluid excretion from the Malpighian tubules (MTs) after a blood meal. We previously cloned and functionally characterized AedaeGPCRcal1 from A. aegypti, the ortholog of the Drosophila melanogaster DH31 receptor and immunolocalized it in the MTs. However, localization of the calcitonin receptor-like receptor 1 (GPCRCAL1) in the hindgut of any insect is unknown, and specifically, knowledge on its role in hindgut contraction in response to Aedae-DH31 peptide is lacking. We analyzed the expression of AedaeGPCRCAL1 in hindgut by western blot and immunohistochemistry, and evaluated its role in hindgut contractility by application of Aedae-DH31 before and after receptor RNA interference (RNAi). The receptor was detected as a 73 kDa band in western blots of hindgut and immunofluorescence revealed the receptor was expressed in hindgut circular and longitudinal muscles but not in the hindgut epithelial cells. In vitro, incubation in 1 µM solution of Aedae-DH31 peptide significantly increased the hindgut contraction frequency in normal mosquitoes. Hindguts from females treated with AedaeGPCRcal1 dsRNA and incubated with DH31 showed a reduction of 50% percent in their contraction frequency with respect to controls. These results suggest that DH31 hormone released from the brain post-blood meal has a direct and coordinative action on the excretory system, MTs and hindgut, by which AedaeGPCRCAL1 signaling stimulates MT primary urine secretion and hindgut contraction resulting in rapid postprandial fluid excretion.


Assuntos
Aedes/metabolismo , Proteína Semelhante a Receptor de Calcitonina/genética , Proteína Semelhante a Receptor de Calcitonina/metabolismo , Proteínas de Insetos/genética , Proteínas de Insetos/metabolismo , Aedes/genética , Animais , Transporte Biológico , Sistema Digestório/metabolismo , Feminino , Regulação da Expressão Gênica , Hormônios de Inseto/metabolismo , Masculino , Músculos/metabolismo , Transporte Proteico
19.
PLoS One ; 7(11): e50374, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-23209727

RESUMO

Evolution of anthropophilic hematophagy in insects resulted in the coordination of various physiological processes for survival. In female mosquitoes, a large blood meal provides proteins for egg production and as a trade-off, rapid elimination of the excess water and solutes (Na(+), Cl(-)) is critical for maintaining homeostasis and removing excess weight to resume flight and avoid predation. This post-prandial excretion is achieved by the concerted action of multiple hormones. Diuresis and natriuresis elicited by the calcitonin-like diuretic hormone 31 (DH(31)) are believed to be mediated by a yet uncharacterized calcitonin receptor (GPRCAL) in the mosquito Malpighian tubules (MTs), the renal organs. To contribute knowledge on endocrinology of mosquito diuresis we cloned GPRCAL1 from MT cDNA. This receptor is the ortholog of the DH(31) receptor from Drosophila melanogaster that is expressed in principal cells of the fruit fly MT. Immunofluorescence similarly showed AaegGPRCAL1 is present in MT principal cells in A. aegypti, however, exhibiting an overall gradient-like pattern along the tubule novel for a GPCR in insects. Variegated, cell-specific receptor expression revealed a subpopulation of otherwise phenotypically similar principal cells. To investigate the receptor contribution to fluid elimination, RNAi was followed by urine measurement assays. In vitro, MTs from females that underwent AaegGPRcal1 knock-down exhibited up to 57% decrease in the rate of fluid secretion in response to DH(31). Live females treated with AaegGPRcal1 dsRNA exhibited 30% reduction in fluid excreted after a blood meal. The RNAi-induced phenotype demonstrates the critical contribution of this single secretin-like family B GPCR to fluid excretion in invertebrates and highlights its relevance for the blood feeding adaptation. Our results with the mosquito AaegGPRCAL1 imply that the regulatory function of calcitonin-like receptors for ion and fluid transport in renal organs arose early in evolution.


Assuntos
Aedes/metabolismo , Diurese , Regulação da Expressão Gênica , Proteínas de Insetos/fisiologia , Rim/metabolismo , Receptores da Calcitonina/metabolismo , Animais , Calcitonina/metabolismo , Clonagem Molecular , DNA Complementar/metabolismo , Feminino , Proteínas de Insetos/biossíntese , Íons , Ligantes , Túbulos de Malpighi/metabolismo , Microscopia de Fluorescência/métodos , Fenótipo , Filogenia , Probabilidade , Interferência de RNA , RNA de Cadeia Dupla/metabolismo , RNA Mensageiro/metabolismo
20.
ACS Appl Mater Interfaces ; 3(10): 3779-87, 2011 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-21957003

RESUMO

Every day, people and animals contract debilitating and life threatening diseases due to bites from infected flies, ticks, and mosquitoes. The current methods utilized to fight against these diseases are only partially effective or safe for humans and animals. When it comes to insect vector control, a conceptual paradigm shift is urgently needed. This work proposes a novel synthetic scheme to produce a nanoparticle-pesticide core-shell conjugate to be used as an active agent against arthropod vectors, such as mosquitoes. As a proof of concept, we conjugated nanosilver to the pyrethroid pesticide deltamethrin. First, electron microscopy and Fourier transform infrared spectroscopy verified the presence of a 15 nm nanosilver core surrounded by deltamethrin. Second, when the conjugate was exposed to mosquitoes for a 24 h bioassay, mortality was observed at 9 × 10(-4) M. Silver was detected in the hemolymph of mosquitoes exposed to the conjugate. We concluded that the newly developed nanoconjugate did not inactivate the primary function of the pesticide and was effective in killing mosquitoes at low concentrations. These results demonstrate the potential to use nanoparticle surfaces to kill insects, specifically vectors of human pathogens.


Assuntos
Anopheles/efeitos dos fármacos , Controle de Insetos/métodos , Insetos Vetores/efeitos dos fármacos , Inseticidas/farmacologia , Nanopartículas Metálicas/química , Animais , Anopheles/fisiologia , Controle de Insetos/instrumentação , Insetos Vetores/fisiologia , Inseticidas/química , Nitrilas/química , Nitrilas/farmacologia , Piretrinas/química , Piretrinas/farmacologia , Prata/química
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA