Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 75
Filtrar
1.
Sci Rep ; 14(1): 4057, 2024 02 19.
Artigo em Inglês | MEDLINE | ID: mdl-38374393

RESUMO

Rapid spread of insecticide resistance among anopheline mosquitoes threatens malaria elimination efforts, necessitating development of alternative vector control technologies. Sterile insect technique (SIT) has been successfully implemented in multiple insect pests to suppress field populations by the release of large numbers of sterile males, yet it has proven difficult to adapt to Anopheles vectors. Here we outline adaptation of a CRISPR-based genetic sterilization system to selectively ablate male sperm cells in the malaria mosquito Anopheles gambiae. We achieve robust mosaic biallelic mutagenesis of zero population growth (zpg, a gene essential for differentiation of germ cells) in F1 individuals after intercrossing a germline-expressing Cas9 transgenic line to a line expressing zpg-targeting gRNAs. Approximately 95% of mutagenized males display complete genetic sterilization, and cause similarly high levels of infertility in their female mates. Using a fluorescence reporter that allows detection of the germline leads to a 100% accurate selection of spermless males, improving the system. These males cause a striking reduction in mosquito population size when released at field-like frequencies in competition cages against wild type males. These findings demonstrate that such a genetic system could be adopted for SIT against important malaria vectors.


Assuntos
Anopheles , Infertilidade Masculina , Malária , Humanos , Animais , Masculino , Feminino , Anopheles/genética , Controle de Mosquitos/métodos , Mosquitos Vetores/genética , Sêmen , RNA Guia de Sistemas CRISPR-Cas , Infertilidade Masculina/genética , Mutagênese , Células Germinativas
2.
PLoS Negl Trop Dis ; 18(1): e0011890, 2024 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-38206958

RESUMO

Anopheles gambiae and its sibling species Anopheles coluzzii are the most efficient vectors of the malaria parasite Plasmodium falciparum. When females of these species feed on an infected human host, oogenesis and parasite development proceed concurrently, but interactions between these processes are not fully understood. Using multiple natural P. falciparum isolates from Burkina Faso, we show that in both vectors, impairing steroid hormone signaling to disrupt oogenesis leads to accelerated oocyst growth and in a manner that appears to depend on both parasite and mosquito genotype. Consistently, we find that egg numbers are negatively linked to oocyst size, a metric for the rate of oocyst development. Oocyst growth rates are also strongly accelerated in females that are in a pre-gravid state, i.e. that fail to develop eggs after an initial blood meal. Overall, these findings advance our understanding of mosquito-parasite interactions that influence P. falciparum development in malaria-endemic regions.


Assuntos
Anopheles , Malária Falciparum , Malária , Animais , Feminino , Humanos , Plasmodium falciparum , Anopheles/parasitologia , Mosquitos Vetores , Interações Hospedeiro-Parasita , Malária Falciparum/parasitologia , Malária/parasitologia , Oocistos
3.
PLoS Genet ; 20(1): e1011145, 2024 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-38285728

RESUMO

Females from many mosquito species feed on blood to acquire nutrients for egg development. The oogenetic cycle has been characterized in the arboviral vector Aedes aegypti, where after a bloodmeal, the lipid transporter lipophorin (Lp) shuttles lipids from the midgut and fat body to the ovaries, and a yolk precursor protein, vitellogenin (Vg), is deposited into the oocyte by receptor-mediated endocytosis. Our understanding of how the roles of these two nutrient transporters are mutually coordinated is however limited in this and other mosquito species. Here, we demonstrate that in the malaria mosquito Anopheles gambiae, Lp and Vg are reciprocally regulated in a timely manner to optimize egg development and ensure fertility. Defective lipid transport via Lp knockdown triggers abortive ovarian follicle development, leading to misregulation of Vg and aberrant yolk granules. Conversely, depletion of Vg causes an upregulation of Lp in the fat body in a manner that appears to be at least partially dependent on target of rapamycin (TOR) signaling, resulting in excess lipid accumulation in the developing follicles. Embryos deposited by Vg-depleted mothers are completely inviable, and are arrested early during development, likely due to severely reduced amino acid levels and protein synthesis. Our findings demonstrate that the mutual regulation of these two nutrient transporters is essential to safeguard fertility by ensuring correct nutrient balance in the developing oocyte, and validate Vg and Lp as two potential candidates for mosquito control.


Assuntos
Aedes , Anopheles , Malária , Feminino , Animais , Anopheles/genética , Mosquitos Vetores/genética , Vitelogeninas/genética , Vitelogeninas/metabolismo , Proteínas do Ovo/metabolismo , Proteínas de Membrana Transportadoras/genética , Proteínas de Membrana Transportadoras/metabolismo , Fertilidade/genética , Lipídeos , Aedes/genética , Aedes/metabolismo
4.
bioRxiv ; 2023 Jun 03.
Artigo em Inglês | MEDLINE | ID: mdl-37398018

RESUMO

Females from many mosquito species feed on blood to acquire nutrients for egg development. The oogenetic cycle has been characterized in the arboviral vector Aedes aegypti, where after a bloodmeal, the lipid transporter lipophorin (Lp) shuttles lipids from the midgut and fat body to the ovaries, and a yolk precursor protein, vitellogenin (Vg), is deposited into the oocyte by receptor-mediated endocytosis. Our understanding of how the roles of these two nutrient transporters are mutually coordinated is however limited in this and other mosquito species. Here, we demonstrate that in the malaria mosquito Anopheles gambiae, Lp and Vg are reciprocally regulated in a timely manner to optimize egg development and ensure fertility. Defective lipid transport via Lp silencing triggers abortive ovarian follicle development, leading to misregulation of Vg and aberrant yolk granules. Conversely, depletion of Vg causes an upregulation of Lp in the fat body in a manner that appears to be at least partially dependent on target of rapamycin (TOR) signaling, resulting in excess lipid accumulation in the developing follicles. Embryos deposited by Vg-depleted mothers are completely infertile, and are arrested early during development, likely due to severely reduced amino acid levels and protein synthesis. Our findings demonstrate that the mutual regulation of these two nutrient transporters is essential to safeguard fertility by ensuring correct nutrient balance in the developing oocyte, and validate Vg and Lp as two potential candidates for mosquito control.

5.
bioRxiv ; 2023 Jun 13.
Artigo em Inglês | MEDLINE | ID: mdl-37398131

RESUMO

Rapid spread of insecticide resistance among anopheline mosquitoes threatens malaria elimination efforts, necessitating development of alternative vector control technologies. Sterile Insect Technique (SIT) has been successfully implemented in multiple insect pests to suppress field populations by the release of large numbers of sterile males, yet it has proven difficult to adapt to Anopheles vectors. Here we outline adaptation of a CRISPR-based genetic sterilization system to selectively ablate male sperm cells in the malaria mosquito Anopheles gambiae. We achieve robust mosaic biallelic mutagenesis of zero population growth (zpg, a gene essential for differentiation of germ cells) in F1 individuals after intercrossing a germline-expressing Cas9 transgenic line to a line expressing zpg-targeting gRNAs. Approximately 95% of mutagenized males display complete genetic sterilization, and cause similarly high levels of infertility in their female mates. Using a fluorescence reporter that allows detection of the germline leads to a 100% accurate selection of spermless males, improving the system. These males cause a striking reduction in mosquito population size when released at field-like frequencies in competition cages against wild type males. These findings demonstrate that such a genetic system could be adopted for SIT against important malaria vectors.

6.
PLoS Pathog ; 19(6): e1011448, 2023 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-37339122

RESUMO

Insecticide resistance is under strong selective pressure in Anopheles mosquitoes due to widespread usage of insecticides in vector control strategies. Resistance mechanisms likely cause changes that profoundly affect mosquito physiology, yet it remains poorly understood how selective pressures imposed by insecticides may alter the ability of the mosquito to host and transmit a Plasmodium infection. From pyrethroid-resistant field-derived Anopheles gambiae s.l. mosquitoes, we established resistant (RES) and susceptible (SUS) colonies by either selection for, or loss of insecticide resistance. We show increased oocyst intensity and growth rate as well as increased sporozoite prevalence and intensity in RES compared to SUS females infected with Plasmodium falciparum. The increase in infection intensity in RES females was not associated with the presence of the kdrL1014F mutation and was not impacted by inhibition of Cytochrome P450s. The lipid transporter lipophorin (Lp), which was upregulated in RES compared to SUS, was at least partly implicated in the increased intensity of P. falciparum but not directly involved in the insecticide resistance phenotype. Interestingly, we observed that although P. falciparum infections were not affected when RES females were exposed to permethrin, these females had decreased lipid abundance in the fat body following exposure, pointing to a possible role for lipid mobilization in response to damage caused by insecticide challenge. The finding that selection for insecticide resistance can increase P. falciparum infection intensities and growth rate reinforces the need to assess the overall impact on malaria transmission dynamics caused by selective pressures mosquitoes experience during repeated insecticide challenge.


Assuntos
Anopheles , Inseticidas , Malária Falciparum , Malária , Animais , Feminino , Inseticidas/farmacologia , Plasmodium falciparum/fisiologia , Resistência a Inseticidas/genética , Anopheles/fisiologia , Mosquitos Vetores/genética , Lipídeos , Controle de Mosquitos
8.
Trends Parasitol ; 38(12): 1031-1040, 2022 12.
Artigo em Inglês | MEDLINE | ID: mdl-36209032

RESUMO

Proof-of-concept studies demonstrate that antimalarial drugs designed for human treatment can also be applied to mosquitoes to interrupt malaria transmission. Deploying a new control tool is ideally undertaken within a stewardship programme that maximises a drug's lifespan by minimising the risk of resistance evolution and slowing its spread once emerged. We ask: what are the epidemiological and evolutionary consequences of targeting parasites within mosquitoes? Our synthesis argues that targeting parasites inside mosquitoes (i) can be modelled by readily expanding existing epidemiological frameworks; (ii) provides a functionally novel control method that has potential to be more robust to resistance evolution than targeting parasites in humans; and (iii) could extend the lifespan and clinical benefit of antimalarials used exclusively to treat humans.


Assuntos
Antimaláricos , Culicidae , Malária , Parasitos , Animais , Humanos , Culicidae/parasitologia , Antimaláricos/uso terapêutico , Malária/parasitologia
9.
Nature ; 608(7921): 93-97, 2022 08.
Artigo em Inglês | MEDLINE | ID: mdl-35794471

RESUMO

Insects, unlike vertebrates, are widely believed to lack male-biased sex steroid hormones1. In the malaria mosquito Anopheles gambiae, the ecdysteroid 20-hydroxyecdysone (20E) appears to have evolved to both control egg development when synthesized by females2 and to induce mating refractoriness when sexually transferred by males3. Because egg development and mating are essential reproductive traits, understanding how Anopheles females integrate these hormonal signals can spur the design of new malaria control programs. Here we reveal that these reproductive functions are regulated by distinct sex steroids through a sophisticated network of ecdysteroid-activating/inactivating enzymes. We identify a male-specific oxidized ecdysteroid, 3-dehydro-20E (3D20E), which safeguards paternity by turning off female sexual receptivity following its sexual transfer and activation by dephosphorylation. Notably, 3D20E transfer also induces expression of a reproductive gene that preserves egg development during Plasmodium infection, ensuring fitness of infected females. Female-derived 20E does not trigger sexual refractoriness but instead licenses oviposition in mated individuals once a 20E-inhibiting kinase is repressed. Identifying this male-specific insect steroid hormone and its roles in regulating female sexual receptivity, fertility and interactions with Plasmodium parasites suggests the possibility for reducing the reproductive success of malaria-transmitting mosquitoes.


Assuntos
Anopheles , Ecdisteroides , Malária , Comportamento Sexual Animal , Animais , Anopheles/enzimologia , Anopheles/parasitologia , Anopheles/fisiologia , Ecdisteroides/biossíntese , Ecdisteroides/metabolismo , Feminino , Fertilidade , Humanos , Malária/parasitologia , Malária/prevenção & controle , Malária/transmissão , Masculino , Mosquitos Vetores/parasitologia , Oviposição , Fosforilação , Plasmodium
10.
PLoS Pathog ; 18(6): e1010609, 2022 06.
Artigo em Inglês | MEDLINE | ID: mdl-35687594

RESUMO

The spread of insecticide resistance in Anopheles mosquitoes and drug resistance in Plasmodium parasites is contributing to a global resurgence of malaria, making the generation of control tools that can overcome these roadblocks an urgent public health priority. We recently showed that the transmission of Plasmodium falciparum parasites can be efficiently blocked when exposing Anopheles gambiae females to antimalarials deposited on a treated surface, with no negative consequences on major components of mosquito fitness. Here, we demonstrate this approach can overcome the hurdles of insecticide resistance in mosquitoes and drug resistant in parasites. We show that the transmission-blocking efficacy of mosquito-targeted antimalarials is maintained when field-derived, insecticide resistant Anopheles are exposed to the potent cytochrome b inhibitor atovaquone, demonstrating that this drug escapes insecticide resistance mechanisms that could potentially interfere with its function. Moreover, this approach prevents transmission of field-derived, artemisinin resistant P. falciparum parasites (Kelch13 C580Y mutant), proving that this strategy could be used to prevent the spread of parasite mutations that induce resistance to front-line antimalarials. Atovaquone is also highly effective at limiting parasite development when ingested by mosquitoes in sugar solutions, including in ongoing infections. These data support the use of mosquito-targeted antimalarials as a promising tool to complement and extend the efficacy of current malaria control interventions.


Assuntos
Anopheles , Antimaláricos , Malária Falciparum , Malária , Plasmodium , Animais , Anopheles/parasitologia , Antimaláricos/farmacologia , Atovaquona/farmacologia , Feminino , Malária/parasitologia , Malária/prevenção & controle , Malária Falciparum/tratamento farmacológico , Malária Falciparum/parasitologia , Malária Falciparum/prevenção & controle , Plasmodium falciparum/genética
12.
Nature ; 602(7897): 475-480, 2022 02.
Artigo em Inglês | MEDLINE | ID: mdl-34929721

RESUMO

Alphaviruses, like many other arthropod-borne viruses, infect vertebrate species and insect vectors separated by hundreds of millions of years of evolutionary history. Entry into evolutionarily divergent host cells can be accomplished by recognition of different cellular receptors in different species, or by binding to receptors that are highly conserved across species. Although multiple alphavirus receptors have been described1-3, most are not shared among vertebrate and invertebrate hosts. Here we identify the very low-density lipoprotein receptor (VLDLR) as a receptor for the prototypic alphavirus Semliki forest virus. We show that the E2 and E1 glycoproteins (E2-E1) of Semliki forest virus, eastern equine encephalitis virus and Sindbis virus interact with the ligand-binding domains (LBDs) of VLDLR and apolipoprotein E receptor 2 (ApoER2), two closely related receptors. Ectopic expression of either protein facilitates cellular attachment, and internalization of virus-like particles, a VLDLR LBD-Fc fusion protein or a ligand-binding antagonist block Semliki forest virus E2-E1-mediated infection of human and mouse neurons in culture. The administration of a VLDLR LBD-Fc fusion protein has protective activity against rapidly fatal Semliki forest virus infection in mouse neonates. We further show that invertebrate receptor orthologues from mosquitoes and worms can serve as functional alphavirus receptors. We propose that the ability of some alphaviruses to infect a wide range of hosts is a result of their engagement of evolutionarily conserved lipoprotein receptors and contributes to their pathogenesis.


Assuntos
Mosquitos Vetores , Vírus da Floresta de Semliki , Animais , Proteínas Relacionadas a Receptor de LDL , Ligantes , Camundongos , Receptores de LDL , Vírus da Floresta de Semliki/metabolismo , Vírus Sindbis/fisiologia
13.
Trends Parasitol ; 38(2): 124-135, 2022 02.
Artigo em Inglês | MEDLINE | ID: mdl-34548252

RESUMO

Interactions between the Anopheles mosquito vector and Plasmodium parasites shape how malaria is transmitted in endemic regions. The long association of these two organisms has led to evolutionary processes that minimize fitness costs of infection and benefit both players through shared nutrient resources, parasite immune suppression, and mosquito tolerance to infection. In this review we explore recent data describing how Plasmodium falciparum, the deadliest malaria parasite, associates with one of its most important natural mosquito hosts, Anopheles gambiae, and we discuss the implications of these findings for parasite transmission and vector control strategies currently in development.


Assuntos
Anopheles , Malária Falciparum , Malária , Plasmodium , Animais , Anopheles/parasitologia , Interações Hospedeiro-Parasita , Malária/parasitologia , Malária Falciparum/parasitologia , Mosquitos Vetores/parasitologia , Plasmodium falciparum
14.
Nat Microbiol ; 6(12): 1575-1582, 2021 12.
Artigo em Inglês | MEDLINE | ID: mdl-34819638

RESUMO

Wolbachia, a maternally inherited intracellular bacterial species, can manipulate host insect reproduction by cytoplasmic incompatibility (CI), which results in embryo lethality in crosses between infected males and uninfected females. CI is encoded by two prophage genes, cifA and cifB. Wolbachia, coupled with the sterile insect technique, has been used in field trials to control populations of the dengue vector Aedes albopictus, but CI-inducing strains are not known to infect the malaria vector Anopheles gambiae. Here we show that cifA and cifB can induce conditional sterility in the malaria vector An. gambiae. We used transgenic expression of these Wolbachia-derived genes in the An. gambiae germline to show that cifB is sufficient to cause embryonic lethality and that cifB-induced sterility is rescued by cifA expression in females. When we co-expressed cifA and cifB in male mosquitoes, the CI phenotype was attenuated. In female mosquitoes, cifB impaired fertility, which was overcome by co-expression of cifA. Our findings pave the way towards using CI to control malaria mosquito vectors.


Assuntos
Anopheles/microbiologia , Anopheles/fisiologia , Proteínas de Bactérias/metabolismo , Herança Extracromossômica , Mosquitos Vetores/microbiologia , Mosquitos Vetores/fisiologia , Wolbachia/metabolismo , Aedes/genética , Aedes/microbiologia , Aedes/fisiologia , Animais , Anopheles/genética , Proteínas de Bactérias/genética , Feminino , Infertilidade Masculina , Malária/transmissão , Masculino , Mosquitos Vetores/genética , Wolbachia/genética
15.
Commun Biol ; 4(1): 911, 2021 07 26.
Artigo em Inglês | MEDLINE | ID: mdl-34312484

RESUMO

Anopheles coluzzii females, important malaria vectors in Africa, mate only once in their lifetime. Mating occurs in aerial swarms with a high male-to-female ratio, where traits underlying male mating success are largely unknown. Here, we investigated whether cuticular hydrocarbons (CHCs) influence mating success in natural mating swarms in Burkina Faso. As insecticides are widely used in this area for malaria control, we also determined whether CHCs affect insecticide resistance levels. We find that mated males have higher CHC abundance than unmated controls, suggesting CHCs could be determinants of mating success. Additionally, mated males have higher insecticide resistance under pyrethroid challenge, and we show a link between resistance intensity and CHC abundance. Taken together, our results suggest that CHC abundance may be subject to sexual selection in addition to selection by insecticide pressure. This has implications for insecticide resistance management, as these traits may be sustained in the population due to their benefits in mating even in the absence of insecticides.


Assuntos
Anopheles/fisiologia , Hidrocarbonetos/farmacologia , Resistência a Inseticidas , Mosquitos Vetores/fisiologia , Feromônios/farmacologia , Comportamento Sexual Animal , Animais , Anopheles/efeitos dos fármacos , Burkina Faso , Epiderme/química , Inseticidas/efeitos adversos , Malária , Mosquitos Vetores/efeitos dos fármacos , Piretrinas/efeitos adversos , Reprodução
16.
PLoS Pathog ; 16(12): e1008908, 2020 12.
Artigo em Inglês | MEDLINE | ID: mdl-33347501

RESUMO

Anopheles mosquitoes have transmitted Plasmodium parasites for millions of years, yet it remains unclear whether they suffer fitness costs to infection. Here we report that the fecundity of virgin and mated females of two important vectors-Anopheles gambiae and Anopheles stephensi-is not affected by infection with Plasmodium falciparum, demonstrating that these human malaria parasites do not inflict this reproductive cost on their natural mosquito hosts. Additionally, parasite development is not impacted by mating status. However, in field studies using different P. falciparum isolates in Anopheles coluzzii, we find that Mating-Induced Stimulator of Oogenesis (MISO), a female reproductive gene strongly induced after mating by the sexual transfer of the steroid hormone 20-hydroxyecdysone (20E), protects females from incurring fecundity costs to infection. MISO-silenced females produce fewer eggs as they become increasingly infected with P. falciparum, while parasite development is not impacted by this gene silencing. Interestingly, previous work had shown that sexual transfer of 20E has specifically evolved in Cellia species of the Anopheles genus, driving the co-adaptation of MISO. Our data therefore suggest that evolution of male-female sexual interactions may have promoted Anopheles tolerance to P. falciparum infection in the Cellia subgenus, which comprises the most important malaria vectors.


Assuntos
Anopheles/genética , Interações Hospedeiro-Parasita/genética , Plasmodium falciparum/genética , Animais , Anopheles/parasitologia , Ecdisterona/genética , Ecdisterona/metabolismo , Feminino , Fertilidade/genética , Expressão Gênica , Hormônios/fisiologia , Malária/parasitologia , Malária Falciparum/parasitologia , Masculino , Mosquitos Vetores/genética , Oogênese , Plasmodium falciparum/patogenicidade , Reprodução/fisiologia
17.
Sci Rep ; 10(1): 21974, 2020 12 15.
Artigo em Inglês | MEDLINE | ID: mdl-33319823

RESUMO

Anopheles gambiae mosquitoes are the most important vectors of human malaria. The reproductive success of these mosquitoes relies on a single copulation event after which the majority of females become permanently refractory to further mating. This refractory behavior is at least partially mediated by the male-synthetized steroid hormone 20-hydroxyecdysone (20E), which is packaged together with other seminal secretions into a gelatinous mating plug and transferred to the female atrium during mating. In this study, we show that two 20E-regulated chymotrypsin-like serine proteases specifically expressed in the reproductive tract of An. gambiae females play an important role in modulating the female susceptibility to mating. Silencing these proteases by RNA interference impairs correct plug processing and slows down the release of the steroid hormone 20E from the mating plug. In turn, depleting one of these proteases, the Mating Regulated Atrial Protease 1 (MatRAP1), reduces female refractoriness to further copulation, so that a significant proportion of females mate again. Microscopy analysis reveals that MatRAP1 is localized on a previously undetected peritrophic matrix-like structure surrounding the mating plug. These data provide novel insight into the molecular mechanisms shaping the post-mating biology of these important malaria vectors.


Assuntos
Anopheles/anatomia & histologia , Anopheles/enzimologia , Peptídeo Hidrolases/metabolismo , Comportamento Sexual Animal , Animais , Anopheles/ultraestrutura , Regulação para Baixo , Ecdisterona/metabolismo , Feminino , Inseminação , Modelos Biológicos
18.
PLoS Pathog ; 16(12): e1009131, 2020 12.
Artigo em Inglês | MEDLINE | ID: mdl-33382824

RESUMO

Many mosquito species, including the major malaria vector Anopheles gambiae, naturally undergo multiple reproductive cycles of blood feeding, egg development and egg laying in their lifespan. Such complex mosquito behavior is regularly overlooked when mosquitoes are experimentally infected with malaria parasites, limiting our ability to accurately describe potential effects on transmission. Here, we examine how Plasmodium falciparum development and transmission potential is impacted when infected mosquitoes feed an additional time. We measured P. falciparum oocyst size and performed sporozoite time course analyses to determine the parasite's extrinsic incubation period (EIP), i.e. the time required by parasites to reach infectious sporozoite stages, in An. gambiae females blood fed either once or twice. An additional blood feed at 3 days post infection drastically accelerates oocyst growth rates, causing earlier sporozoite accumulation in the salivary glands, thereby shortening the EIP (reduction of 2.3 ± 0.4 days). Moreover, parasite growth is further accelerated in transgenic mosquitoes with reduced reproductive capacity, which mimic genetic modifications currently proposed in population suppression gene drives. We incorporate our shortened EIP values into a measure of transmission potential, the basic reproduction number R0, and find the average R0 is higher (range: 10.1%-12.1% increase) across sub-Saharan Africa than when using traditional EIP measurements. These data suggest that malaria elimination may be substantially more challenging and that younger mosquitoes or those with reduced reproductive ability may provide a larger contribution to infection than currently believed. Our findings have profound implications for current and future mosquito control interventions.


Assuntos
Malária Falciparum/transmissão , Mosquitos Vetores/parasitologia , Plasmodium falciparum/crescimento & desenvolvimento , Animais , Anopheles/parasitologia , Comportamento Alimentar , Feminino , Período de Incubação de Doenças Infecciosas
19.
Sci Rep ; 10(1): 14344, 2020 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-32873857

RESUMO

The reproductive fitness of the Anopheles gambiae mosquito represents a promising target to prevent malaria transmission. The ecdysteroid hormone 20-hydroxyecdysone (20E), transferred from male to female during copulation, is key to An. gambiae reproductive success as it licenses females to oviposit eggs developed after blood feeding. Here we show that 20E-triggered oviposition in these mosquitoes is regulated by the stress- and immune-responsive c-Jun N-terminal kinase (JNK). The heads of mated females exhibit a transcriptional signature reminiscent of a JNK-dependent wounding response, while mating-or injection of virgins with exogenous 20E-selectively activates JNK in the same tissue. RNAi-mediated depletion of JNK pathway components inhibits oviposition in mated females, whereas JNK activation by silencing the JNK phosphatase puckered induces egg laying in virgins. Together, these data identify JNK as a potential conduit linking stress responses and reproductive success in the most important vector of malaria.


Assuntos
Anopheles/fisiologia , Sistema de Sinalização das MAP Quinases/genética , Proteína Quinase 8 Ativada por Mitógeno/metabolismo , Mosquitos Vetores/fisiologia , Oviposição/genética , Animais , Copulação/efeitos dos fármacos , Ecdisterona/farmacologia , Feminino , Malária/parasitologia , Malária/transmissão , Masculino , Proteína Quinase 8 Ativada por Mitógeno/genética , Fosfatases da Proteína Quinase Ativada por Mitógeno/genética , Fosfatases da Proteína Quinase Ativada por Mitógeno/metabolismo , Plasmodium , Interferência de RNA
20.
Sci Rep ; 10(1): 13847, 2020 08 14.
Artigo em Inglês | MEDLINE | ID: mdl-32796890

RESUMO

Wolbachia, an endosymbiotic alpha-proteobacterium commonly found in insects, can inhibit the transmission of human pathogens by mosquitoes. Biocontrol programs are underway using Aedes aegypti mosquitoes trans-infected with a non-natural Wolbachia strain to reduce dengue virus transmission. Less is known about the impact of Wolbachia on the biology and vectorial capacity of Anopheles mosquitoes, the vectors of malaria parasites. A naturally occurring strain of Wolbachia, wAnga, infects populations of the major malaria vectors Anopheles gambiae and Anopheles coluzzii in Burkina Faso. Previous studies found wAnga infection was negatively correlated with Plasmodium infection in the mosquito and wAnga influenced mosquito egg-laying behavior. Here, we investigate wAnga in natural populations of An. coluzzii and its interactions with other resident microbiota using targeted 16S sequencing. Though we find no major differences in microbiota composition associated with wAnga infection, we do find several taxa that correlate with the presence or absence of wAnga in female mosquitoes following oviposition, with the caveat that we could not rule out batch effects due to the unanticipated impact of wAnga on oviposition timing. These data suggest wAnga may influence or interact with the Anopheles microbiota, which may contribute to the impact of wAnga on Anopheles biology and vectorial capacity.


Assuntos
Anopheles/microbiologia , Anopheles/fisiologia , Interações entre Hospedeiro e Microrganismos/fisiologia , Insetos Vetores , Wolbachia , Animais , Burkina Faso , Vetores de Doenças , Feminino , Malária/transmissão , Controle de Mosquitos/métodos , Oviposição , Doenças Transmitidas por Vetores/transmissão
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...