Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 75
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Cell ; 177(2): 315-325.e14, 2019 04 04.
Artículo en Inglés | MEDLINE | ID: mdl-30929905

RESUMEN

Transmission of malaria parasites occurs when a female Anopheles mosquito feeds on an infected host to acquire nutrients for egg development. How parasites are affected by oogenetic processes, principally orchestrated by the steroid hormone 20-hydroxyecdysone (20E), remains largely unknown. Here we show that Plasmodium falciparum development is intimately but not competitively linked to processes shaping Anopheles gambiae reproduction. We unveil a 20E-mediated positive correlation between egg and oocyst numbers; impairing oogenesis by multiple 20E manipulations decreases parasite intensities. These manipulations, however, accelerate Plasmodium growth rates, allowing sporozoites to become infectious sooner. Parasites exploit mosquito lipids for faster growth, but they do so without further affecting egg development. These results suggest that P. falciparum has adopted a non-competitive evolutionary strategy of resource exploitation to optimize transmission while minimizing fitness costs to its mosquito vector. Our findings have profound implications for currently proposed control strategies aimed at suppressing mosquito populations.


Asunto(s)
Ecdisterona/metabolismo , Interacciones Huésped-Parásitos/fisiología , Malaria Falciparum/parasitología , Animales , Anopheles/parasitología , Culicidae , Ecdisterona/fisiología , Femenino , Células HEK293 , Humanos , Insectos Vectores , Malaria/parasitología , Ratones , Mosquitos Vectores , Células 3T3 NIH , Oogénesis/fisiología , Plasmodium/metabolismo , Plasmodium falciparum , Esporozoítos , Esteroides/metabolismo
2.
Nature ; 608(7921): 93-97, 2022 08.
Artículo en Inglés | MEDLINE | ID: mdl-35794471

RESUMEN

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.


Asunto(s)
Anopheles , Ecdisteroides , Malaria , Conducta Sexual Animal , Animales , Anopheles/enzimología , Anopheles/parasitología , Anopheles/fisiología , Ecdisteroides/biosíntesis , Ecdisteroides/metabolismo , Femenino , Fertilidad , Humanos , Malaria/parasitología , Malaria/prevención & control , Malaria/transmisión , Masculino , Mosquitos Vectores/parasitología , Oviposición , Fosforilación , Plasmodium
3.
Nature ; 602(7897): 475-480, 2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-34929721

RESUMEN

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.


Asunto(s)
Mosquitos Vectores , Virus de los Bosques Semliki , Animales , Proteínas Relacionadas con Receptor de LDL , Ligandos , Ratones , Receptores de LDL , Virus de los Bosques Semliki/metabolismo , Virus Sindbis/fisiología
4.
PLoS Genet ; 20(1): e1011145, 2024 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-38285728

RESUMEN

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.


Asunto(s)
Aedes , Anopheles , Malaria , Femenino , Animales , Anopheles/genética , Mosquitos Vectores/genética , Vitelogeninas/genética , Vitelogeninas/metabolismo , Proteínas del Huevo/metabolismo , Proteínas de Transporte de Membrana/genética , Proteínas de Transporte de Membrana/metabolismo , Fertilidad/genética , Lípidos , Aedes/genética , Aedes/metabolismo
5.
PLoS Pathog ; 19(6): e1011448, 2023 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-37339122

RESUMEN

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.


Asunto(s)
Anopheles , Insecticidas , Malaria Falciparum , Malaria , Animales , Femenino , Insecticidas/farmacología , Plasmodium falciparum/fisiología , Resistencia a los Insecticidas/genética , Anopheles/fisiología , Mosquitos Vectores/genética , Lípidos , Control de Mosquitos
6.
Nature ; 567(7747): 239-243, 2019 03.
Artículo en Inglés | MEDLINE | ID: mdl-30814727

RESUMEN

Bites of Anopheles mosquitoes transmit Plasmodium falciparum parasites that cause malaria, which kills hundreds of thousands of people every year. Since the turn of this century, efforts to prevent the transmission of these parasites via the mass distribution of insecticide-treated bed nets have been extremely successful, and have led to an unprecedented reduction in deaths from malaria1. However, resistance to insecticides has become widespread in Anopheles populations2-4, which has led to the threat of a global resurgence of malaria and makes the generation of effective tools for controlling this disease an urgent public health priority. Here we show that the development of P. falciparum can be rapidly and completely blocked when female Anopheles gambiae mosquitoes take up low concentrations of specific antimalarials from treated surfaces-conditions that simulate contact with a bed net. Mosquito exposure to atovaquone before, or shortly after, P. falciparum infection causes full parasite arrest in the midgut, and prevents transmission of infection. Similar transmission-blocking effects are achieved using other cytochrome b inhibitors, which demonstrates that parasite mitochondrial function is a suitable target for killing parasites. Incorporating these effects into a model of malaria transmission dynamics predicts that impregnating mosquito nets with Plasmodium inhibitors would substantially mitigate the global health effects of insecticide resistance. This study identifies a powerful strategy for blocking Plasmodium transmission by female Anopheles mosquitoes, which has promising implications for efforts to eradicate malaria.


Asunto(s)
Anopheles/efectos de los fármacos , Anopheles/parasitología , Antimaláricos/farmacología , Malaria Falciparum/prevención & control , Malaria Falciparum/transmisión , Control de Mosquitos/métodos , Mosquitos Vectores/efectos de los fármacos , Plasmodium falciparum , África/epidemiología , Animales , Anopheles/crecimiento & desarrollo , Antimaláricos/administración & dosificación , Atovacuona/administración & dosificación , Atovacuona/farmacología , Citocromos b/antagonistas & inhibidores , Femenino , Mosquiteros Tratados con Insecticida , Malaria Falciparum/epidemiología , Modelos Biológicos , Mosquitos Vectores/parasitología , Plasmodium falciparum/efectos de los fármacos , Plasmodium falciparum/patogenicidad , Factores de Tiempo
7.
PLoS Pathog ; 18(6): e1010609, 2022 06.
Artículo en Inglés | MEDLINE | ID: mdl-35687594

RESUMEN

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.


Asunto(s)
Anopheles , Antimaláricos , Malaria Falciparum , Malaria , Plasmodium , Animales , Anopheles/parasitología , Antimaláricos/farmacología , Atovacuona/farmacología , Femenino , Malaria/parasitología , Malaria/prevención & control , Malaria Falciparum/tratamiento farmacológico , Malaria Falciparum/parasitología , Malaria Falciparum/prevención & control , Plasmodium falciparum/genética
8.
PLoS Pathog ; 16(12): e1009131, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-33382824

RESUMEN

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.


Asunto(s)
Malaria Falciparum/transmisión , Mosquitos Vectores/parasitología , Plasmodium falciparum/crecimiento & desarrollo , Animales , Anopheles/parasitología , Conducta Alimentaria , Femenino , Periodo de Incubación de Enfermedades Infecciosas
9.
PLoS Pathog ; 16(12): e1008908, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-33347501

RESUMEN

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.


Asunto(s)
Anopheles/genética , Interacciones Huésped-Parásitos/genética , Plasmodium falciparum/genética , Animales , Anopheles/parasitología , Ecdisterona/genética , Ecdisterona/metabolismo , Femenino , Fertilidad/genética , Expresión Génica , Hormonas/fisiología , Malaria/parasitología , Malaria Falciparum/parasitología , Masculino , Mosquitos Vectores/genética , Oogénesis , Plasmodium falciparum/patogenicidad , Reproducción/fisiología
10.
Nature ; 538(7625): 344-349, 2016 Oct 20.
Artículo en Inglés | MEDLINE | ID: mdl-27602946

RESUMEN

Antimalarial drugs have thus far been chiefly derived from two sources-natural products and synthetic drug-like compounds. Here we investigate whether antimalarial agents with novel mechanisms of action could be discovered using a diverse collection of synthetic compounds that have three-dimensional features reminiscent of natural products and are underrepresented in typical screening collections. We report the identification of such compounds with both previously reported and undescribed mechanisms of action, including a series of bicyclic azetidines that inhibit a new antimalarial target, phenylalanyl-tRNA synthetase. These molecules are curative in mice at a single, low dose and show activity against all parasite life stages in multiple in vivo efficacy models. Our findings identify bicyclic azetidines with the potential to both cure and prevent transmission of the disease as well as protect at-risk populations with a single oral dose, highlighting the strength of diversity-oriented synthesis in revealing promising therapeutic targets.


Asunto(s)
Antimaláricos/síntesis química , Antimaláricos/farmacología , Azetidinas/uso terapéutico , Descubrimiento de Drogas , Estadios del Ciclo de Vida/efectos de los fármacos , Malaria Falciparum/tratamiento farmacológico , Plasmodium falciparum/efectos de los fármacos , Plasmodium falciparum/crecimiento & desarrollo , Animales , Antimaláricos/administración & dosificación , Antimaláricos/uso terapéutico , Compuestos de Azabiciclo/administración & dosificación , Compuestos de Azabiciclo/síntesis química , Compuestos de Azabiciclo/farmacología , Compuestos de Azabiciclo/uso terapéutico , Azetidinas/administración & dosificación , Azetidinas/efectos adversos , Azetidinas/farmacología , Citosol/enzimología , Modelos Animales de Enfermedad , Femenino , Hígado/efectos de los fármacos , Hígado/parasitología , Macaca mulatta/parasitología , Malaria Falciparum/prevención & control , Malaria Falciparum/transmisión , Masculino , Ratones , Fenilalanina-ARNt Ligasa/antagonistas & inhibidores , Compuestos de Fenilurea/administración & dosificación , Compuestos de Fenilurea/síntesis química , Compuestos de Fenilurea/farmacología , Compuestos de Fenilurea/uso terapéutico , Plasmodium falciparum/citología , Plasmodium falciparum/enzimología , Seguridad
13.
PLoS Pathog ; 12(12): e1006060, 2016 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-27977810

RESUMEN

The control of mosquito populations with insecticide treated bed nets and indoor residual sprays remains the cornerstone of malaria reduction and elimination programs. In light of widespread insecticide resistance in mosquitoes, however, alternative strategies for reducing transmission by the mosquito vector are urgently needed, including the identification of safe compounds that affect vectorial capacity via mechanisms that differ from fast-acting insecticides. Here, we show that compounds targeting steroid hormone signaling disrupt multiple biological processes that are key to the ability of mosquitoes to transmit malaria. When an agonist of the steroid hormone 20-hydroxyecdysone (20E) is applied to Anopheles gambiae females, which are the dominant malaria mosquito vector in Sub Saharan Africa, it substantially shortens lifespan, prevents insemination and egg production, and significantly blocks Plasmodium falciparum development, three components that are crucial to malaria transmission. Modeling the impact of these effects on Anopheles population dynamics and Plasmodium transmission predicts that disrupting steroid hormone signaling using 20E agonists would affect malaria transmission to a similar extent as insecticides. Manipulating 20E pathways therefore provides a powerful new approach to tackle malaria transmission by the mosquito vector, particularly in areas affected by the spread of insecticide resistance.


Asunto(s)
Anopheles/efectos de los fármacos , Anopheles/parasitología , Hidrazinas/farmacología , Insecticidas/farmacología , Hormonas Juveniles/farmacología , Malaria/transmisión , Control de Mosquitos/métodos , Animales , Ecdisterona/agonistas , Femenino , Etiquetado Corte-Fin in Situ , Insectos Vectores/efectos de los fármacos , Insectos Vectores/parasitología , Mosquiteros Tratados con Insecticida , Estadios del Ciclo de Vida/efectos de los fármacos , Modelos Teóricos , Dinámica Poblacional
14.
Proc Natl Acad Sci U S A ; 111(16): 5854-9, 2014 Apr 22.
Artículo en Inglés | MEDLINE | ID: mdl-24711401

RESUMEN

Anopheles gambiae mosquitoes are major African vectors of malaria, a disease that kills more than 600,000 people every year. Given the spread of insecticide resistance in natural mosquito populations, alternative vector control strategies aimed at reducing the reproductive success of mosquitoes are being promoted. Unlike many other insects, An. gambiae females mate a single time in their lives and must use sperm stored in the sperm storage organ, the spermatheca, to fertilize a lifetime's supply of eggs. Maintenance of sperm viability during storage is therefore crucial to the reproductive capacity of these mosquitoes. However, to date, no information is available on the factors and mechanisms ensuring sperm functionality in the spermatheca. Here we identify cellular components and molecular mechanisms used by An. gambiae females to maximize their fertility. Pathways of energy metabolism, cellular transport, and oxidative stress are strongly regulated by mating in the spermatheca. We identify the mating-induced heme peroxidase (HPX) 15 as an important factor in long-term fertility, and demonstrate that its function is required during multiple gonotrophic cycles. We find that HPX15 induction is regulated by sexually transferred 20-hydroxy-ecdysone (20E), a steroid hormone that is produced by the male accessory glands and transferred during copulation, and that expression of this peroxidase is mediated via the 20E nuclear receptor. To our knowledge, our findings provide the first evidence of the mechanisms regulating fertility in Anopheles, and identify HPX15 as a target for vector control.


Asunto(s)
Estructuras Animales/enzimología , Anopheles/enzimología , Proteínas de Insectos/metabolismo , Peroxidasa/metabolismo , Conducta Sexual Animal , Espermatozoides/enzimología , Estructuras Animales/citología , Estructuras Animales/efectos de los fármacos , Estructuras Animales/ultraestructura , Animales , Anopheles/efectos de los fármacos , Anopheles/genética , Ecdisona/farmacología , Activación Enzimática/efectos de los fármacos , Femenino , Fertilidad/efectos de los fármacos , Fertilidad/genética , Regulación de la Expresión Génica/efectos de los fármacos , Hemo/metabolismo , Proteínas de Insectos/genética , Masculino , Peroxidasa/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Conducta Sexual Animal/efectos de los fármacos , Espermatozoides/citología , Espermatozoides/efectos de los fármacos , Espermatozoides/ultraestructura , Transcripción Genética/efectos de los fármacos
15.
Proc Natl Acad Sci U S A ; 111(46): 16353-8, 2014 Nov 18.
Artículo en Inglés | MEDLINE | ID: mdl-25368171

RESUMEN

Female insects generally mate multiple times during their lives. A notable exception is the female malaria mosquito Anopheles gambiae, which after sex loses her susceptibility to further copulation. Sex in this species also renders females competent to lay eggs developed after blood feeding. Despite intense research efforts, the identity of the molecular triggers that cause the postmating switch in females, inducing a permanent refractoriness to further mating and triggering egg-laying, remains elusive. Here we show that the male-transferred steroid hormone 20-hydroxyecdysone (20E) is a key regulator of monandry and oviposition in An. gambiae. When sexual transfer of 20E is impaired by partial inactivation of the hormone and inhibition of its biosynthesis in males, oviposition and refractoriness to further mating in the female are strongly reduced. Conversely, mimicking sexual delivery by injecting 20E into virgin females switches them to an artificial mated status, triggering egg-laying and reducing susceptibility to copulation. Sexual transfer of 20E appears to incapacitate females physically from receiving seminal fluids by a second male. Comparative analysis of microarray data from females after mating and after 20E treatment indicates that 20E-regulated molecular pathways likely are implicated in the postmating switch, including cytoskeleton and musculature-associated genes that may render the atrium impenetrable to additional mates. By revealing signals and pathways shaping key processes in the An. gambiae reproductive biology, our data offer new opportunities for the control of natural populations of malaria vectors.


Asunto(s)
Anopheles/fisiología , Ecdisterona/fisiología , Conducta Sexual Animal/fisiología , Animales , Copulación , Ecdisterona/farmacología , Femenino , Perfilación de la Expresión Génica , Genes de Insecto , Inyecciones , Insectos Vectores/fisiología , Malaria/transmisión , Masculino , Análisis de Secuencia por Matrices de Oligonucleótidos , Oviposición/fisiología , Factores de Tiempo , Transcripción Genética
16.
PLoS Biol ; 11(10): e1001695, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-24204210

RESUMEN

Molecular interactions between male and female factors during mating profoundly affect the reproductive behavior and physiology of female insects. In natural populations of the malaria mosquito Anopheles gambiae, blood-fed females direct nutritional resources towards oogenesis only when inseminated. Here we show that the mating-dependent pathway of egg development in these mosquitoes is regulated by the interaction between the steroid hormone 20-hydroxy-ecdysone (20E) transferred by males during copulation and a female Mating-Induced Stimulator of Oogenesis (MISO) protein. RNAi silencing of MISO abolishes the increase in oogenesis caused by mating in blood-fed females, causes a delay in oocyte development, and impairs the function of male-transferred 20E. Co-immunoprecipitation experiments show that MISO and 20E interact in the female reproductive tract. Moreover MISO expression after mating is induced by 20E via the Ecdysone Receptor, demonstrating a close cooperation between the two factors. Male-transferred 20E therefore acts as a mating signal that females translate into an increased investment in egg development via a MISO-dependent pathway. The identification of this male-female reproductive interaction offers novel opportunities for the control of mosquito populations that transmit malaria.


Asunto(s)
Anopheles/metabolismo , Ecdisona/análogos & derivados , Hormonas/metabolismo , Proteínas de Insectos/metabolismo , Malaria/parasitología , Oogénesis , Esteroides/metabolismo , Animales , Anopheles/efectos de los fármacos , Anopheles/genética , Ecdisona/metabolismo , Ecdisona/farmacología , Conducta Alimentaria/efectos de los fármacos , Femenino , Regulación de la Expresión Génica/efectos de los fármacos , Técnicas de Silenciamiento del Gen , Silenciador del Gen/efectos de los fármacos , Genes de Insecto/genética , Proteínas de Insectos/genética , Metabolismo de los Lípidos/efectos de los fármacos , Lipoproteínas/metabolismo , Masculino , Oocitos/efectos de los fármacos , Oocitos/metabolismo , Oogénesis/efectos de los fármacos , Oogénesis/genética , Óvulo/metabolismo , Conducta Sexual Animal/efectos de los fármacos
17.
J Biol Chem ; 288(7): 4844-53, 2013 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-23288850

RESUMEN

Male Anopheles mosquitoes coagulate their seminal fluids via cross-linking of a substrate, called Plugin, by the seminal transglutaminase AgTG3. Formation of the "mating plug" by cross-linking Plugin is necessary for efficient sperm storage by females. AgTG3 has a similar degree of sequence identity (~30%) to both human Factor XIII (FXIII) and tissue transglutaminase 2 (hTG2). Here we report the solution structure and in vitro activity for the cross-linking reaction of AgTG3 and Plugin. AgTG3 is a dimer in solution and exhibits Ca(2+)-dependent nonproteolytic activation analogous to cytoplasmic FXIII. The C-terminal domain of Plugin is predominantly α-helical with extended tertiary structure and oligomerizes in solution. The specific activity of AgTG3 was measured as 4.25 × 10(-2) units mg(-1). AgTG3 is less active than hTG2 assayed using the general substrate TVQQEL but has 8-10× higher relative activity when Plugin is the substrate. Mass spectrometric analysis of cross-linked Plugin detects specific peptides including a predicted consensus motif for cross-linking by AgTG3. These results support the development of AgTG3 inhibitors as specific and effective chemosterilants for A. gambiae.


Asunto(s)
Anopheles/enzimología , Transglutaminasas/química , Secuencia de Aminoácidos , Animales , Calcio/química , Reactivos de Enlaces Cruzados/química , Citoplasma/metabolismo , Dimerización , Femenino , Masculino , Espectrometría de Masas/métodos , Modelos Químicos , Datos de Secuencia Molecular , Péptidos/química , Unión Proteica , Proteína Glutamina Gamma Glutamiltransferasa 2 , Estructura Terciaria de Proteína , Homología de Secuencia de Aminoácido , Relación Estructura-Actividad , Transglutaminasas/metabolismo
18.
Malar J ; 13: 210, 2014 Jun 02.
Artículo en Inglés | MEDLINE | ID: mdl-24888439

RESUMEN

BACKGROUND: Current vector-based malaria control strategies are threatened by the rise of biochemical and behavioural resistance in mosquitoes. Researching mosquito traits of immunity and fertility is required to find potential targets for new vector control strategies. The seminal transglutaminase AgTG3 coagulates male Anopheles gambiae seminal fluids, forming a 'mating plug' that is required for male reproductive success. Inhibitors of AgTG3 can be useful both as chemical probes of A. gambiae reproductive biology and may further the development of new chemosterilants for mosquito population control. METHODS: A targeted library of 3-bromo-4,5-dihydroxoisoxazole inhibitors were synthesized and screened for inhibition of AgTG3 in a fluorescent, plate-based assay. Positive hits were tested for in vitro activity using cross-linking and mass spectrometry, and in vivo efficacy in laboratory mating assays. RESULTS: A targeted chemical library was screened for inhibition of AgTG3 in a fluorescent plate-based assay using its native substrate, plugin. Several inhibitors were identified with IC50 < 10 µM. Preliminary structure-activity relationships within the library support the stereo-specificity and preference for aromatic substituents in the chemical scaffold. Both inhibition of plugin cross-linking and covalent modification of the active site cysteine of AgTG3 were verified. Administration of an AgTG3 inhibitor to A. gambiae males by intrathoracic injection led to a 15% reduction in mating plug transfer in laboratory mating assays. CONCLUSIONS: A targeted screen has identified chemical inhibitors of A. gambiae transglutaminase 3 (AgTG3). The most potent inhibitors are known inhibitors of human transglutaminase 2, suggesting a common binding pose may exist within the active site of both enzymes. Future efforts to develop additional inhibitors will provide chemical tools to address important biological questions regarding the role of the A. gambiae mating plug. A second use for transglutaminase inhibitors exists for the study of haemolymph coagulation and immune responses to wound healing in insects.


Asunto(s)
Anopheles/enzimología , Esterilizantes Químicos/farmacología , Proteínas de Insectos/antagonistas & inhibidores , Isoxazoles/farmacología , Control de Mosquitos/métodos , Semen/enzimología , Transglutaminasas/antagonistas & inhibidores , Animales , Dominio Catalítico , Esterilizantes Químicos/síntesis química , Esterilizantes Químicos/química , Reactivos de Enlaces Cruzados/química , Reactivos de Enlaces Cruzados/farmacología , Inhibidores de Cisteína Proteinasa/farmacología , Evaluación Preclínica de Medicamentos , Femenino , Humanos , Concentración 50 Inhibidora , Isoxazoles/síntesis química , Isoxazoles/química , Masculino , Modelos Moleculares , Estructura Molecular , Conformación Proteica , Proteínas Recombinantes/efectos de los fármacos , Bibliotecas de Moléculas Pequeñas , Especificidad de la Especie , Relación Estructura-Actividad , Especificidad por Sustrato
19.
Proc Natl Acad Sci U S A ; 108(33): 13677-81, 2011 Aug 16.
Artículo en Inglés | MEDLINE | ID: mdl-21825136

RESUMEN

Anopheles gambiae sensu stricto is the major vector of malaria, a disease with devastating consequences for human health. Given the constant spread of the disease, alternative approaches to the use of insecticides are urgently needed to control vector populations. Females of this species undergo large behavioral changes after mating, which include a life-long refractoriness to further insemination and the induction of egg laying in blood-fed individuals. Genetic control strategies aimed at impacting Anopheles fertility through the release of sterile males are being advocated to reduce the size of mosquito field populations. Such strategies depend on the ability of the released sterile males to mate successfully with wild females and to switch off the female receptivity to further copulation. Here we evaluate the role of sperm in regulating female behavioral responses after mating in An. gambiae. We developed spermless males by RNAi silencing of a germ cell differentiation gene. These males mated successfully and preserved standard accessory gland functions. Females mated to spermless males exhibited normal postcopulatory responses, which included laying large numbers of eggs upon blood feeding and becoming refractory to subsequent insemination. Moreover, spermless males induced transcriptional changes in female reproductive genes comparable to those elicited by fertile males. Our data demonstrate that, in contrast to Drosophila, targeting sperm in An. gambiae preserves normal male and female reproductive behavior for the traits and time frame analyzed and validate the use of approaches based on incapacitation or elimination of sperm for genetic control of vector populations to block malaria transmission.


Asunto(s)
Anopheles/fisiología , Infertilidad Masculina/genética , ARN Interferente Pequeño/farmacología , Conducta Sexual Animal , Animales , Anopheles/efectos de los fármacos , Femenino , Silenciador del Gen/efectos de los fármacos , Inseminación , Masculino , Reproducción , Espermatozoides/efectos de los fármacos
20.
Sci Rep ; 14(1): 4057, 2024 02 19.
Artículo en Inglés | MEDLINE | ID: mdl-38374393

RESUMEN

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.


Asunto(s)
Anopheles , Infertilidad Masculina , Malaria , Humanos , Animales , Masculino , Femenino , Anopheles/genética , Control de Mosquitos/métodos , Mosquitos Vectores/genética , Semen , ARN Guía de Sistemas CRISPR-Cas , Infertilidad Masculina/genética , Mutagénesis , Células Germinativas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA