Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21.788
Filtrar
Más filtros

Intervalo de año de publicación
1.
Cell ; 185(17): 3104-3123.e28, 2022 08 18.
Artículo en Inglés | MEDLINE | ID: mdl-35985288

RESUMEN

Aedes aegypti mosquitoes are a persistent human foe, transmitting arboviruses including dengue when they feed on human blood. Mosquitoes are intensely attracted to body odor and carbon dioxide, which they detect using ionotropic chemosensory receptors encoded by three large multi-gene families. Genetic mutations that disrupt the olfactory system have modest effects on human attraction, suggesting redundancy in odor coding. The canonical view is that olfactory sensory neurons each express a single chemosensory receptor that defines its ligand selectivity. We discovered that Ae. aegypti uses a different organizational principle, with many neurons co-expressing multiple chemosensory receptor genes. In vivo electrophysiology demonstrates that the broad ligand-sensitivity of mosquito olfactory neurons depends on this non-canonical co-expression. The redundancy afforded by an olfactory system in which neurons co-express multiple chemosensory receptors may increase the robustness of the mosquito olfactory system and explain our long-standing inability to disrupt the detection of humans by mosquitoes.


Asunto(s)
Aedes , Neuronas Receptoras Olfatorias , Aedes/genética , Animales , Humanos , Ligandos , Odorantes
2.
Cell ; 185(17): 3079-3081, 2022 08 18.
Artículo en Inglés | MEDLINE | ID: mdl-35985284

RESUMEN

Mosquitoes are strongly attracted to humans, and their bites not only cause intense itch but can beget severe diseases. In this issue of Cell, Herre et al. reveal that non-canonical olfactory circuit organization and coding likely endow mosquitoes with a robust ability to locate human hosts.


Asunto(s)
Aedes , Anopheles , Animales , Humanos , Odorantes , Feromonas
3.
Cell ; 185(22): 4099-4116.e13, 2022 10 27.
Artículo en Inglés | MEDLINE | ID: mdl-36261039

RESUMEN

Some people are more attractive to mosquitoes than others, but the mechanistic basis of this phenomenon is poorly understood. We tested mosquito attraction to human skin odor and identified people who are exceptionally attractive or unattractive to mosquitoes. These differences were stable over several years. Chemical analysis revealed that highly attractive people produce significantly more carboxylic acids in their skin emanations. Mutant mosquitoes lacking the chemosensory co-receptors Ir8a, Ir25a, or Ir76b were severely impaired in attraction to human scent, but retained the ability to differentiate highly and weakly attractive people. The link between elevated carboxylic acids in "mosquito-magnet" human skin odor and phenotypes of genetic mutations in carboxylic acid receptors suggests that such compounds contribute to differential mosquito attraction. Understanding why some humans are more attractive than others provides insights into what skin odorants are most important to the mosquito and could inform the development of more effective repellents.


Asunto(s)
Aedes , Anopheles , Repelentes de Insectos , Animales , Humanos , Ácidos Carboxílicos/farmacología , Odorantes/análisis , Repelentes de Insectos/farmacología , Repelentes de Insectos/análisis
4.
Cell ; 176(4): 679-681, 2019 02 07.
Artículo en Inglés | MEDLINE | ID: mdl-30735629

RESUMEN

New possibilities for vector-borne disease control are revealed by Duvall et al. (2019), who link host-seeking behavioral modulation in Aedes aegypti to neuropeptide Y (NPY)-like receptor 7. Small-molecule screening yields agonist compounds able to activate NPYLR7 and suppress attraction to hosts.


Asunto(s)
Aedes , Mordeduras y Picaduras de Insectos , Animales , Mosquitos Vectores , Receptores de Neuropéptido Y
5.
Cell ; 178(5): 1057-1071.e11, 2019 08 22.
Artículo en Inglés | MEDLINE | ID: mdl-31442400

RESUMEN

The Zika epidemic in the Americas has challenged surveillance and control. As the epidemic appears to be waning, it is unclear whether transmission is still ongoing, which is exacerbated by discrepancies in reporting. To uncover locations with lingering outbreaks, we investigated travel-associated Zika cases to identify transmission not captured by reporting. We uncovered an unreported outbreak in Cuba during 2017, a year after peak transmission in neighboring islands. By sequencing Zika virus, we show that the establishment of the virus was delayed by a year and that the ensuing outbreak was sparked by long-lived lineages of Zika virus from other Caribbean islands. Our data suggest that, although mosquito control in Cuba may initially have been effective at mitigating Zika virus transmission, such measures need to be maintained to be effective. Our study highlights how Zika virus may still be "silently" spreading and provides a framework for understanding outbreak dynamics. VIDEO ABSTRACT.


Asunto(s)
Epidemias , Genómica/métodos , Infección por el Virus Zika/epidemiología , Aedes/virología , Animales , Cuba/epidemiología , Humanos , Incidencia , Control de Mosquitos , Filogenia , ARN Viral/química , ARN Viral/metabolismo , Análisis de Secuencia de ARN , Viaje , Indias Occidentales/epidemiología , Virus Zika/clasificación , Virus Zika/genética , Virus Zika/aislamiento & purificación , Infección por el Virus Zika/transmisión , Infección por el Virus Zika/virología
6.
Cell ; 176(4): 687-701.e5, 2019 02 07.
Artículo en Inglés | MEDLINE | ID: mdl-30735632

RESUMEN

Female Aedes aegypti mosquitoes bite humans to obtain blood to develop their eggs. Remarkably, their strong attraction to humans is suppressed for days after the blood meal by an unknown mechanism. We investigated a role for neuropeptide Y (NPY)-related signaling in long-term behavioral suppression and discovered that drugs targeting human NPY receptors modulate mosquito host-seeking. In a screen of all 49 predicted Ae. aegypti peptide receptors, we identified NPY-like receptor 7 (NPYLR7) as the sole target of these drugs. To obtain small-molecule agonists selective for NPYLR7, we performed a high-throughput cell-based assay of 265,211 compounds and isolated six highly selective NPYLR7 agonists that inhibit mosquito attraction to humans. NPYLR7 CRISPR-Cas9 null mutants are defective in behavioral suppression and resistant to these drugs. Finally, we show that these drugs can inhibit biting and blood-feeding on a live host, suggesting a novel approach to control infectious disease transmission by controlling mosquito behavior. VIDEO ABSTRACT.


Asunto(s)
Conducta de Búsqueda de Hospedador/efectos de los fármacos , Mosquitos Vectores/efectos de los fármacos , Receptores de Neuropéptido Y/agonistas , Aedes/metabolismo , Animales , Conducta Alimentaria/efectos de los fármacos , Femenino , Células HEK293 , Humanos , Mordeduras y Picaduras de Insectos , Receptores de Neuropéptido Y/metabolismo , Bibliotecas de Moléculas Pequeñas/análisis
7.
Nat Immunol ; 22(8): 958-968, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-34267374

RESUMEN

Antibody-dependent enhancement (ADE) is an important safety concern for vaccine development against dengue virus (DENV) and its antigenically related Zika virus (ZIKV) because vaccine may prime deleterious antibodies to enhance natural infections. Cross-reactive antibodies targeting the conserved fusion loop epitope (FLE) are known as the main sources of ADE. We design ZIKV immunogens engineered to change the FLE conformation but preserve neutralizing epitopes. Single vaccination conferred sterilizing immunity against ZIKV without ADE of DENV-serotype 1-4 infections and abrogated maternal-neonatal transmission in mice. Unlike the wild-type-based vaccine inducing predominately cross-reactive ADE-prone antibodies, B cell profiling revealed that the engineered vaccines switched immunodominance to dispersed patterns without DENV enhancement. The crystal structure of the engineered immunogen showed the dimeric conformation of the envelope protein with FLE disruption. We provide vaccine candidates that will prevent both ZIKV infection and infection-/vaccination-induced DENV ADE.


Asunto(s)
Acrecentamiento Dependiente de Anticuerpo/inmunología , Antígenos Virales/inmunología , Reacciones Cruzadas/inmunología , Vacunas contra el Dengue/inmunología , Dengue/prevención & control , Virus Zika/inmunología , Aedes , Animales , Anticuerpos Neutralizantes/inmunología , Anticuerpos Antivirales/inmunología , Linfocitos B/inmunología , Chlorocebus aethiops , Cricetinae , Virus del Dengue/inmunología , Células HEK293 , Humanos , Ratones , Ratones Endogámicos BALB C , Ratones Noqueados , Receptor de Interferón alfa y beta/genética , Vacunación , Células Vero , Infección por el Virus Zika/inmunología , Infección por el Virus Zika/prevención & control
8.
Cell ; 170(2): 273-283.e12, 2017 Jul 13.
Artículo en Inglés | MEDLINE | ID: mdl-28708997

RESUMEN

The emergence of Zika virus (ZIKV) and its association with congenital malformations has prompted the rapid development of vaccines. Although efficacy with multiple viral vaccine platforms has been established in animals, no study has addressed protection during pregnancy. We tested in mice two vaccine platforms, a lipid nanoparticle-encapsulated modified mRNA vaccine encoding ZIKV prM and E genes and a live-attenuated ZIKV strain encoding an NS1 protein without glycosylation, for their ability to protect against transmission to the fetus. Vaccinated dams challenged with a heterologous ZIKV strain at embryo day 6 (E6) and evaluated at E13 showed markedly diminished levels of viral RNA in maternal, placental, and fetal tissues, which resulted in protection against placental damage and fetal demise. As modified mRNA and live-attenuated vaccine platforms can restrict in utero transmission of ZIKV in mice, their further development in humans to prevent congenital ZIKV syndrome is warranted.


Asunto(s)
Vacunas Virales/administración & dosificación , Infección por el Virus Zika/inmunología , Infección por el Virus Zika/prevención & control , Virus Zika/fisiología , Aedes/virología , Animales , Anticuerpos Neutralizantes/sangre , Anticuerpos Antivirales/sangre , Células Sanguíneas/virología , Embrión de Mamíferos/virología , Femenino , Feto/virología , Humanos , Lípidos/administración & dosificación , Masculino , Ratones , Ratones Endogámicos C57BL , Mutación , ARN Mensajero/genética , ARN Mensajero/inmunología , Organismos Libres de Patógenos Específicos , Vacunas de Subunidad/administración & dosificación , Vacunas de Subunidad/inmunología , Proteínas no Estructurales Virales/genética , Proteínas no Estructurales Virales/inmunología , Vacunas Virales/inmunología , Infección por el Virus Zika/virología
9.
Nat Immunol ; 19(4): 342-353, 2018 04.
Artículo en Inglés | MEDLINE | ID: mdl-29507355

RESUMEN

Pathogens have co-evolved with mosquitoes to optimize transmission to hosts. Mosquito salivary-gland extract is known to modulate host immune responses and facilitate pathogen transmission, but the underlying molecular mechanisms of this have remained unknown. In this study, we identified and characterized a prominent 15-kilodalton protein, LTRIN, obtained from the salivary glands of the mosquito Aedes aegypti. LTRIN expression was upregulated in blood-fed mosquitoes, and LTRIN facilitated the transmission of Zika virus (ZIKV) and exacerbated its pathogenicity by interfering with signaling through the lymphotoxin-ß receptor (LTßR). Mechanically, LTRIN bound to LTßR and 'preferentially' inhibited signaling via the transcription factor NF-κB and the production of inflammatory cytokines by interfering with the dimerization of LTßR during infection with ZIKV. Furthermore, treatment with antibody to LTRIN inhibited mosquito-mediated infection with ZIKV, and abolishing LTßR potentiated the infectivity of ZIKV both in vitro and in vivo. This study provides deeper insight into the transmission of mosquito-borne diseases in nature and supports the therapeutic potential of inhibiting the action of LTRIN to disrupt ZIKV transmission.


Asunto(s)
Aedes/virología , Proteínas de Insectos/metabolismo , Saliva/metabolismo , Infección por el Virus Zika/transmisión , Virus Zika/patogenicidad , Animales , Humanos , Receptor beta de Linfotoxina/inmunología , Receptor beta de Linfotoxina/metabolismo , Ratones , Mosquitos Vectores/química , Mosquitos Vectores/inmunología , Mosquitos Vectores/metabolismo , Saliva/química
10.
Nature ; 633(8030): 615-623, 2024 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-39169183

RESUMEN

Mosquito-borne diseases affect hundreds of millions of people annually and disproportionately impact the developing world1,2. One mosquito species, Aedes aegypti, is a primary vector of viruses that cause dengue, yellow fever and Zika. The attraction of Ae. aegypti female mosquitos to humans requires integrating multiple cues, including CO2 from breath, organic odours from skin and visual cues, all sensed at mid and long ranges, and other cues sensed at very close range3-6. Here we identify a cue that Ae. aegypti use as part of their sensory arsenal to find humans. We demonstrate that Ae. aegypti sense the infrared (IR) radiation emanating from their targets and use this information in combination with other cues for highly effective mid-range navigation. Detection of thermal IR requires the heat-activated channel TRPA1, which is expressed in neurons at the tip of the antenna. Two opsins are co-expressed with TRPA1 in these neurons and promote the detection of lower IR intensities. We propose that radiant energy causes local heating at the end of the antenna, thereby activating temperature-sensitive receptors in thermosensory neurons. The realization that thermal IR radiation is an outstanding mid-range directional cue expands our understanding as to how mosquitoes are exquisitely effective in locating hosts.


Asunto(s)
Aedes , Señales (Psicología) , Conducta de Búsqueda de Hospedador , Calor , Rayos Infrarrojos , Navegación Espacial , Sensación Térmica , Animales , Femenino , Humanos , Aedes/citología , Aedes/fisiología , Aedes/efectos de la radiación , Antenas de Artrópodos/citología , Antenas de Artrópodos/inervación , Antenas de Artrópodos/fisiología , Conducta de Búsqueda de Hospedador/fisiología , Conducta de Búsqueda de Hospedador/efectos de la radiación , Mosquitos Vectores/citología , Mosquitos Vectores/fisiología , Mosquitos Vectores/efectos de la radiación , Neuronas/efectos de la radiación , Neuronas/metabolismo , Neuronas/fisiología , Opsinas/metabolismo , Sensación Térmica/fisiología , Sensación Térmica/efectos de la radiación , Canal Catiónico TRPA1/metabolismo , Dióxido de Carbono/metabolismo , Olor Corporal , Navegación Espacial/fisiología , Navegación Espacial/efectos de la radiación
11.
Cell ; 156(5): 1060-71, 2014 Feb 27.
Artículo en Inglés | MEDLINE | ID: mdl-24581501

RESUMEN

Multiple sensory cues emanating from humans are thought to guide blood-feeding female mosquitoes to a host. To determine the relative contribution of carbon dioxide (CO2) detection to mosquito host-seeking behavior, we mutated the AaegGr3 gene, a subunit of the heteromeric CO2 receptor in Aedes aegypti mosquitoes. Gr3 mutants lack electrophysiological and behavioral responses to CO2. These mutants also fail to show CO2-evoked responses to heat and lactic acid, a human-derived attractant, suggesting that CO2 can gate responses to other sensory stimuli. Whereas attraction of Gr3 mutants to live humans in a large semi-field environment was only slightly impaired, responses to an animal host were greatly reduced in a spatial-scale-dependent manner. Synergistic integration of heat and odor cues likely drive host-seeking behavior in the absence of CO2 detection. We reveal a networked series of interactions by which multimodal integration of CO2, human odor, and heat orchestrates mosquito attraction to humans.


Asunto(s)
Aedes/fisiología , Dióxido de Carbono , Animales , Sangre , Humanos , Proteínas de Insectos/genética , Proteínas de Insectos/metabolismo , Insectos Vectores/fisiología , Ácido Láctico/metabolismo , Odorantes , Receptores de Superficie Celular/genética , Receptores de Superficie Celular/metabolismo
12.
EMBO J ; 43(9): 1690-1721, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38378891

RESUMEN

Mosquitoes transmit many disease-relevant flaviviruses. Efficient viral transmission to mammalian hosts requires mosquito salivary factors. However, the specific salivary components facilitating viral transmission and their mechanisms of action remain largely unknown. Here, we show that a female mosquito salivary gland-specific protein, here named A. aegypti Neutrophil Recruitment Protein (AaNRP), facilitates the transmission of Zika and dengue viruses. AaNRP promotes a rapid influx of neutrophils, followed by virus-susceptible myeloid cells toward mosquito bite sites, which facilitates establishment of local infection and systemic dissemination. Mechanistically, AaNRP engages TLR1 and TLR4 of skin-resident macrophages and activates MyD88-dependent NF-κB signaling to induce the expression of neutrophil chemoattractants. Inhibition of MyD88-NF-κB signaling with the dietary phytochemical resveratrol reduces AaNRP-mediated enhancement of flavivirus transmission by mosquitoes. These findings exemplify how salivary components can aid viral transmission, and suggest a potential prophylactic target.


Asunto(s)
Aedes , Virus Zika , Animales , Aedes/virología , Aedes/metabolismo , Femenino , Virus Zika/fisiología , Ratones , Virus del Dengue/fisiología , Proteínas y Péptidos Salivales/metabolismo , Mosquitos Vectores/virología , Proteínas de Insectos/metabolismo , Células Mieloides/virología , Células Mieloides/metabolismo , Infección por el Virus Zika/transmisión , Infección por el Virus Zika/virología , Infección por el Virus Zika/metabolismo , Dengue/transmisión , Dengue/virología , Dengue/metabolismo , FN-kappa B/metabolismo , Transducción de Señal , Factor 88 de Diferenciación Mieloide/metabolismo , Factor 88 de Diferenciación Mieloide/genética
13.
Cell ; 155(6): 1365-79, 2013 Dec 05.
Artículo en Inglés | MEDLINE | ID: mdl-24315103

RESUMEN

Female mosquitoes that transmit deadly diseases locate human hosts by detecting exhaled CO2 and skin odor. The identities of olfactory neurons and receptors required for attraction to skin odor remain a mystery. Here, we show that the CO2-sensitive olfactory neuron is also a sensitive detector of human skin odorants in both Aedes aegypti and Anopheles gambiae. We demonstrate that activity of this neuron is important for attraction to skin odor, establishing it as a key target for intervention. We screen ~0.5 million compounds in silico and identify several CO2 receptor ligands, including an antagonist that reduces attraction to skin and an agonist that lures mosquitoes to traps as effectively as CO2. Analysis of the CO2 receptor ligand space provides a foundation for understanding mosquito host-seeking behavior and identifies odors that are potentially safe, pleasant, and affordable for use in a new generation of mosquito control strategies worldwide.


Asunto(s)
Aedes/fisiología , Anopheles/fisiología , Dióxido de Carbono/metabolismo , Proteínas de Insectos/metabolismo , Odorantes , Receptores de Superficie Celular/metabolismo , Animales , Femenino , Humanos , Proteínas de Insectos/genética , Control de Mosquitos , Neuronas/fisiología , Receptores de Superficie Celular/genética , Piel/metabolismo
14.
Nature ; 605(7911): 706-712, 2022 05.
Artículo en Inglés | MEDLINE | ID: mdl-35508661

RESUMEN

A globally invasive form of the mosquito Aedes aegypti specializes in biting humans, making it an efficient disease vector1. Host-seeking female mosquitoes strongly prefer human odour over the odour of animals2,3, but exactly how they distinguish between the two is not known. Vertebrate odours are complex blends of volatile chemicals with many shared components4-7, making discrimination an interesting sensory coding challenge. Here we show that human and animal odours evoke activity in distinct combinations of olfactory glomeruli within the Ae. aegypti antennal lobe. One glomerulus in particular is strongly activated by human odour but responds weakly, or not at all, to animal odour. This human-sensitive glomerulus is selectively tuned to the long-chain aldehydes decanal and undecanal, which we show are consistently enriched in human odour and which probably originate from unique human skin lipids. Using synthetic blends, we further demonstrate that signalling in the human-sensitive glomerulus significantly enhances long-range host-seeking behaviour in a wind tunnel, recapitulating preference for human over animal odours. Our research suggests that animal brains may distil complex odour stimuli of innate biological relevance into simple neural codes and reveals targets for the design of next-generation mosquito-control strategies.


Asunto(s)
Aedes , Encéfalo , Conducta de Búsqueda de Hospedador , Odorantes , Aedes/fisiología , Animales , Encéfalo/fisiología , Femenino , Humanos , Control de Mosquitos , Mosquitos Vectores/fisiología
15.
PLoS Biol ; 22(3): e3002573, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-38547237

RESUMEN

The rising interest and success in deploying inherited microorganisms and cytoplasmic incompatibility (CI) for vector control strategies necessitate an explanation of the CI mechanism. Wolbachia-induced CI manifests in the form of embryonic lethality when sperm from Wolbachia-bearing testes fertilize eggs from uninfected females. Embryos from infected females however survive to sustain the maternally inherited symbiont. Previously in Drosophila melanogaster flies, we demonstrated that CI modifies chromatin integrity in developing sperm to bestow the embryonic lethality. Here, we validate these findings using wMel-transinfected Aedes aegypti mosquitoes released to control vector-borne diseases. Once again, the prophage WO CI proteins, CifA and CifB, target male gametic nuclei to modify chromatin integrity via an aberrant histone-to-protamine transition. Cifs are not detected in the embryo, and thus elicit CI via the nucleoprotein modifications established pre-fertilization. The rescue protein CifA in oogenesis localizes to stem cell, nurse cell, and oocyte nuclei, as well as embryonic DNA during embryogenesis. Discovery of the nuclear targeting Cifs and altered histone-to-protamine transition in both Aedes aegypti mosquitoes and D. melanogaster flies affirm the Host Modification Model of CI is conserved across these host species. The study also newly uncovers the cell biology of Cif proteins in the ovaries, CifA localization in the embryos, and an impaired histone-to-protamine transition during spermiogenesis of any mosquito species. Overall, these sperm modification findings may enable future optimization of CI efficacy in vectors or pests that are refractory to Wolbachia transinfections.


Asunto(s)
Aedes , Arbovirus , Wolbachia , Animales , Femenino , Masculino , Drosophila melanogaster/genética , Histonas/genética , Mosquitos Vectores , Semen , Drosophila/genética , Cromatina , Protaminas/genética
16.
PLoS Genet ; 20(4): e1010891, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38683842

RESUMEN

Transcriptional cis-regulatory modules, e.g., enhancers, control the time and location of metazoan gene expression. While changes in enhancers can provide a powerful force for evolution, there is also significant deep conservation of enhancers for developmentally important genes, with function and sequence characteristics maintained over hundreds of millions of years of divergence. Not well understood, however, is how the overall regulatory composition of a locus evolves, with important outstanding questions such as how many enhancers are conserved vs. novel, and to what extent are the locations of conserved enhancers within a locus maintained? We begin here to address these questions with a comparison of the respective single-minded (sim) loci in the two dipteran species Drosophila melanogaster (fruit fly) and Aedes aegypti (mosquito). sim encodes a highly conserved transcription factor that mediates development of the arthropod embryonic ventral midline. We identify two enhancers in the A. aegypti sim locus and demonstrate that they function equivalently in both transgenic flies and transgenic mosquitoes. One A. aegypti enhancer is highly similar to known Drosophila counterparts in its activity, location, and autoregulatory capability. The other differs from any known Drosophila sim enhancers with a novel location, failure to autoregulate, and regulation of expression in a unique subset of midline cells. Our results suggest that the conserved pattern of sim expression in the two species is the result of both conserved and novel regulatory sequences. Further examination of this locus will help to illuminate how the overall regulatory landscape of a conserved developmental gene evolves.


Asunto(s)
Aedes , Drosophila melanogaster , Elementos de Facilitación Genéticos , Regulación del Desarrollo de la Expresión Génica , Animales , Aedes/genética , Aedes/embriología , Drosophila melanogaster/genética , Drosophila melanogaster/embriología , Secuencia Conservada , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Animales Modificados Genéticamente , Evolución Molecular , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo
17.
Proc Natl Acad Sci U S A ; 121(35): e2407394121, 2024 Aug 27.
Artículo en Inglés | MEDLINE | ID: mdl-39159375

RESUMEN

Aedes aegypti mosquitoes are major vectors of dengue, chikungunya, and other arboviral diseases. Ae. aegypti's capacity to reproduce and to spread disease depends on the female mosquitoes' ability to obtain blood meals and find water-filled containers in which to lay eggs (oviposit). While humidity sensation (hygrosensation) has been implicated in these behaviors, the specific hygrosensory pathways involved have been unclear. Here, we establish the distinct molecular requirements and anatomical locations of Ae. aegypti Dry Cells and Moist Cells and examine their contributions to behavior. We show that Dry Cell and Moist Cell responses to humidity involve different ionotropic receptor (IR) family sensory receptors, with dry air-activated Dry Cells reliant upon the IR Ir40a, and humid air-activated Moist Cells upon Ir68a. Both classes of hygrosensors innervate multiple antennal sensilla, including sensilla ampullacea near the antennal base as well as two classes of coeloconic sensilla near the tip. Dry Cells and Moist Cells each support behaviors linked to mosquito reproduction but contribute differently: Ir40a-dependent Dry Cells act in parallel with Ir68a-dependent Moist Cells to promote blood feeding, while oviposition site seeking is driven specifically by Ir68a-dependent Moist Cells. Together these findings reveal the importance of distinct hygrosensory pathways in blood feeding and oviposition site seeking and suggest Ir40a-dependent Dry Cells and Ir68a-dependent Moist Cells as potential targets for vector control strategies.


Asunto(s)
Aedes , Conducta Alimentaria , Humedad , Mosquitos Vectores , Oviposición , Animales , Aedes/fisiología , Oviposición/fisiología , Femenino , Conducta Alimentaria/fisiología , Mosquitos Vectores/fisiología , Sensilos/fisiología , Receptores Ionotrópicos de Glutamato/metabolismo , Antenas de Artrópodos/fisiología
18.
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
19.
Proc Natl Acad Sci U S A ; 121(28): e2408072121, 2024 Jul 09.
Artículo en Inglés | MEDLINE | ID: mdl-38950363

RESUMEN

Female mosquitoes produce eggs in gonadotrophic cycles that are divided between a previtellogenic and vitellogenic phase. Previtellogenic females consume water and sugar sources like nectar while also being attracted to hosts for blood feeding. Consumption of a blood meal activates the vitellogenic phase, which produces mature eggs and suppresses host attraction. In this study, we tested the hypothesis that neuropeptide Y-like hormones differentially modulate host attraction behavior in the mosquito Aedes aegypti. A series of experiments collectively indicated that enteroendocrine cells (EECs) in the posterior midgut produce and release neuropeptide F (NPF) into the hemolymph during the previtellogenic phase which stimulates attraction to humans and biting behavior. Consumption of a blood meal, which primarily consists of protein by dry weight, down-regulated NPF in EECs until mature eggs developed, which was associated with a decline in hemolymph titer. NPF depletion depended on protein digestion but was not associated with EEC loss. Other experiments showed that neurons in the terminal ganglion extend axons to the posterior midgut and produce RYamide, which showed evidence of increased secretion into circulation after a blood meal. Injection of RYamide-1 and -2 into previtellogenic females suppressed host attraction, while coinjection of RYamides with or without short NPF-2 also inhibited the host attraction activity of NPF. Overall, our results identify NPF and RYamide as gut-associated hormones in A. aegypti that link host attraction behavior to shifts in diet during sequential gonadotrophic cycles.


Asunto(s)
Aedes , Neuropéptidos , Animales , Aedes/metabolismo , Aedes/fisiología , Neuropéptidos/metabolismo , Femenino , Conducta Alimentaria/fisiología , Hemolinfa/metabolismo , Células Enteroendocrinas/metabolismo , Proteínas de Insectos/metabolismo , Humanos , Vitelogénesis/fisiología
20.
PLoS Genet ; 20(3): e1011196, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-38466721

RESUMEN

Hematophagous mosquitoes require vertebrate blood for their reproductive cycles, making them effective vectors for transmitting dangerous human diseases. Thus, high-intensity metabolism is needed to support reproductive events of female mosquitoes. However, the regulatory mechanism linking metabolism and reproduction in mosquitoes remains largely unclear. In this study, we found that the expression of estrogen-related receptor (ERR), a nuclear receptor, is activated by the direct binding of 20-hydroxyecdysone (20E) and ecdysone receptor (EcR) to the ecdysone response element (EcRE) in the ERR promoter region during the gonadotropic cycle of Aedes aegypti (named AaERR). RNA interference (RNAi) of AaERR in female mosquitoes led to delayed development of ovaries. mRNA abundance of genes encoding key enzymes involved in carbohydrate metabolism (CM)-glucose-6-phosphate isomerase (GPI) and pyruvate kinase (PYK)-was significantly decreased in AaERR knockdown mosquitoes, while the levels of metabolites, such as glycogen, glucose, and trehalose, were elevated. The expression of fatty acid synthase (FAS) was notably downregulated, and lipid accumulation was reduced in response to AaERR depletion. Dual luciferase reporter assays and electrophoretic mobility shift assays (EMSA) determined that AaERR directly activated the expression of metabolic genes, such as GPI, PYK, and FAS, by binding to the corresponding AaERR-responsive motif in the promoter region of these genes. Our results have revealed an important role of AaERR in the regulation of metabolism during mosquito reproduction and offer a novel target for mosquito control.


Asunto(s)
Aedes , Receptores de Esteroides , Animales , Femenino , Humanos , Aedes/genética , Aedes/metabolismo , Ecdisona/metabolismo , Mosquitos Vectores/genética , Receptores Citoplasmáticos y Nucleares/genética , Receptores Citoplasmáticos y Nucleares/metabolismo , Homeostasis/genética , Proteínas de Insectos/genética , Proteínas de Insectos/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA