Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 43
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
PLoS Pathog ; 19(8): e1011588, 2023 08.
Artículo en Inglés | MEDLINE | ID: mdl-37651317

RESUMEN

Several aspects of mosquito ecology that are important for vectored disease transmission and control have been difficult to measure at epidemiologically important scales in the field. In particular, the ability to describe mosquito population structure and movement rates has been hindered by difficulty in quantifying fine-scale genetic variation among populations. The mosquito virome represents a possible avenue for quantifying population structure and movement rates across multiple spatial scales. Mosquito viromes contain a diversity of viruses, including several insect-specific viruses (ISVs) and "core" viruses that have high prevalence across populations. To date, virome studies have focused on viral discovery and have only recently begun examining viral ecology. While nonpathogenic ISVs may be of little public health relevance themselves, they provide a possible route for quantifying mosquito population structure and dynamics. For example, vertically transmitted viruses could behave as a rapidly evolving extension of the host's genome. It should be possible to apply established analytical methods to appropriate viral phylogenies and incidence data to generate novel approaches for estimating mosquito population structure and dispersal over epidemiologically relevant timescales. By studying the virome through the lens of spatial and genomic epidemiology, it may be possible to investigate otherwise cryptic aspects of mosquito ecology. A better understanding of mosquito population structure and dynamics are key for understanding mosquito-borne disease ecology and methods based on ISVs could provide a powerful tool for informing mosquito control programs.


Asunto(s)
Virus de Insectos , Animales , Ecología , Vectores Genéticos , Genómica , Insectos
2.
Ecol Lett ; 26(7): 1029-1049, 2023 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-37349261

RESUMEN

Vector-borne diseases cause significant financial and human loss, with billions of dollars spent on control. Arthropod vectors experience a complex suite of environmental factors that affect fitness, population growth and species interactions across multiple spatial and temporal scales. Temperature and water availability are two of the most important abiotic variables influencing their distributions and abundances. While extensive research on temperature exists, the influence of humidity on vector and pathogen parameters affecting disease dynamics are less understood. Humidity is often underemphasized, and when considered, is often treated as independent of temperature even though desiccation likely contributes to declines in trait performance at warmer temperatures. This Perspectives explores how humidity shapes the thermal performance of mosquito-borne pathogen transmission. We summarize what is known about its effects and propose a conceptual model for how temperature and humidity interact to shape the range of temperatures across which mosquitoes persist and achieve high transmission potential. We discuss how failing to account for these interactions hinders efforts to forecast transmission dynamics and respond to epidemics of mosquito-borne infections. We outline future research areas that will ground the effects of humidity on the thermal biology of pathogen transmission in a theoretical and empirical framework to improve spatial and temporal prediction of vector-borne pathogen transmission.


Asunto(s)
Culicidae , Enfermedades Transmitidas por Vectores , Humanos , Animales , Humedad , Mosquitos Vectores , Temperatura , Biología
3.
Malar J ; 22(1): 104, 2023 Mar 21.
Artículo en Inglés | MEDLINE | ID: mdl-36945014

RESUMEN

BACKGROUND: Anopheles stephensi is a malaria-transmitting mosquito that has recently expanded from its primary range in Asia and the Middle East, to locations in Africa. This species is a competent vector of both Plasmodium falciparum and Plasmodium vivax malaria. Perhaps most alarming, the characteristics of An. stephensi, such as container breeding and anthropophily, make it particularly adept at exploiting built environments in areas with no prior history of malaria risk. METHODS: In this paper, global maps of thermal transmission suitability and people at risk (PAR) for malaria transmission by An. stephensi were created, under current and future climate. Temperature-dependent transmission suitability thresholds derived from recently published species-specific thermal curves were used to threshold gridded, monthly mean temperatures under current and future climatic conditions. These temperature driven transmission models were coupled with gridded population data for 2020 and 2050, under climate-matched scenarios for future outcomes, to compare with baseline predictions for 2020 populations. RESULTS: Using the Global Burden of Disease regions approach revealed that heterogenous regional increases and decreases in risk did not mask the overall pattern of massive increases of PAR for malaria transmission suitability with An. stephensi presence. General patterns of poleward expansion for thermal suitability were seen for both P. falciparum and P. vivax transmission potential. CONCLUSIONS: Understanding the potential suitability for An. stephensi transmission in a changing climate provides a key tool for planning, given an ongoing invasion and expansion of the vector. Anticipating the potential impact of onward expansion to transmission suitable areas, and the size of population at risk under future climate scenarios, and where they occur, can serve as a large-scale call for attention, planning, and monitoring.


Asunto(s)
Anopheles , Malaria Falciparum , Malaria Vivax , Malaria , Humanos , Animales , Malaria/epidemiología , Malaria Falciparum/epidemiología , Plasmodium falciparum , África/epidemiología , Mosquitos Vectores
4.
Malar J ; 21(1): 264, 2022 Sep 13.
Artículo en Inglés | MEDLINE | ID: mdl-36100902

RESUMEN

BACKGROUND: Sporozoites isolated from the salivary glands of Plasmodium-infected mosquitoes are a prerequisite for several basic and pre-clinical applications. Although salivary glands are pooled to maximize sporozoite recovery, insufficient yields pose logistical and analytical hurdles; thus, predicting yields prior to isolation would be valuable. Preceding oocyst densities in the midgut is an obvious candidate. However, it is unclear whether current understanding of its relationship with sporozoite densities can be used to maximize yields, or whether it can capture the potential density-dependence in rates of sporozoite invasion of the salivary glands. METHODS: This study presents a retrospective analysis of Anopheles stephensi mosquitoes infected with two strains of the rodent-specific Plasmodium berghei. Mean oocyst densities were estimated in the midguts earlier in the infection (11-15 days post-blood meal), with sporozoites pooled from the salivary glands later in the infection (17-29 days). Generalized linear mixed effects models were used to determine if (1) mean oocyst densities can predict sporozoite yields from pooled salivary glands, (2) whether these densities can capture differences in rates of sporozoite invasion of salivary glands, and (3), if the interaction between oocyst densities and time could be leveraged to boost overall yields. RESULTS: The non-linear effect of mean oocyst densities confirmed the role of density-dependent constraints in limiting yields beyond certain oocyst densities. Irrespective of oocyst densities however, the continued invasion of salivary glands by the sporozoites boosted recoveries over time (17-29 days post-blood meal) for either parasite strain. CONCLUSIONS: Sporozoite invasion of the salivary glands over time can be leveraged to maximize yields for P. berghei. In general, however, invasion of the salivary glands over time is a critical fitness determinant for all Plasmodium species (extrinsic incubation period, EIP). Thus, delaying sporozoite collection could, in principle, substantially reduce dissection effort for any parasite within the genus, with the results also alluding to the potential for changes in sporozoites densities over time to modify infectivity for the next host.


Asunto(s)
Anopheles , Esporozoítos , Animales , Anopheles/parasitología , Plasmodium berghei , Estudios Retrospectivos , Glándulas Salivales/parasitología
5.
Proc Biol Sci ; 288(1946): 20202501, 2021 03 10.
Artículo en Inglés | MEDLINE | ID: mdl-33653145

RESUMEN

Precision health mapping is a technique that uses spatial relationships between socio-ecological variables and disease to map the spatial distribution of disease, particularly for diseases with strong environmental signatures, such as diarrhoeal disease (DD). While some studies use GPS-tagged location data, other precision health mapping efforts rely heavily on data collected at coarse-spatial scales and may not produce operationally relevant predictions at fine enough spatio-temporal scales to inform local health programmes. We use two fine-scale health datasets collected in a rural district of Madagascar to identify socio-ecological covariates associated with childhood DD. We constructed generalized linear mixed models including socio-demographic, climatic and landcover variables and estimated variable importance via multi-model inference. We find that socio-demographic variables, and not environmental variables, are strong predictors of the spatial distribution of disease risk at both individual and commune-level (cluster of villages) spatial scales. Climatic variables predicted strong seasonality in DD, with the highest incidence in colder, drier months, but did not explain spatial patterns. Interestingly, the occurrence of a national holiday was highly predictive of increased DD incidence, highlighting the need for including cultural factors in modelling efforts. Our findings suggest that precision health mapping efforts that do not include socio-demographic covariates may have reduced explanatory power at the local scale. More research is needed to better define the set of conditions under which the application of precision health mapping can be operationally useful to local public health professionals.


Asunto(s)
Diarrea , Niño , Diarrea/epidemiología , Humanos , Incidencia , Modelos Lineales , Madagascar/epidemiología , Factores de Riesgo
6.
Glob Chang Biol ; 27(1): 84-93, 2021 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-33037740

RESUMEN

In the aftermath of the 2015 pandemic of Zika virus (ZIKV), concerns over links between climate change and emerging arboviruses have become more pressing. Given the potential that much of the world might remain at risk from the virus, we used a previously established temperature-dependent transmission model for ZIKV to project climate change impacts on transmission suitability risk by mid-century (a generation into the future). Based on these model predictions, in the worst-case scenario, over 1.3 billion new people could face suitable transmission temperatures for ZIKV by 2050. The next generation will face substantially increased ZIKV transmission temperature suitability in North America and Europe, where naïve populations might be particularly vulnerable. Mitigating climate change even to moderate emissions scenarios could significantly reduce global expansion of climates suitable for ZIKV transmission, potentially protecting around 200 million people. Given these suitability risk projections, we suggest an increased priority on research establishing the immune history of vulnerable populations, modeling when and where the next ZIKV outbreak might occur, evaluating the efficacy of conventional and novel intervention measures, and increasing surveillance efforts to prevent further expansion of ZIKV.


Asunto(s)
Aedes , Infección por el Virus Zika , Virus Zika , Animales , Europa (Continente) , Humanos , Mosquitos Vectores , América del Norte , Temperatura , Infección por el Virus Zika/epidemiología
7.
Ecol Appl ; 31(5): e02334, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-33772946

RESUMEN

Invasive mosquitoes are expanding their ranges into new geographic areas and interacting with resident mosquito species. Understanding how novel interactions can affect mosquito population dynamics is necessary to predict transmission risk at invasion fronts. Mosquito life-history traits are extremely sensitive to temperature, and this can lead to temperature-dependent competition between competing invasive mosquito species. We explored temperature-dependent competition between Aedes aegypti and Anopheles stephensi, two invasive mosquito species whose distributions overlap in India, the Middle East, and North Africa, where An. stephensi is currently expanding into the endemic range of Ae. aegypti. We followed mosquito cohorts raised at different intraspecific and interspecific densities across five temperatures (16-32°C) to measure traits relevant for population growth and to estimate species' per capita growth rates. We then used these growth rates to derive each species' competitive ability at each temperature. We find strong evidence for asymmetric competition at all temperatures, with Ae. aegypti emerging as the dominant competitor. This was primarily because of differences in larval survival and development times across all temperatures that resulted in a higher estimated intrinsic growth rate and competitive tolerance estimate for Ae. aegypti compared to An. stephensi. The spread of An. stephensi into the African continent could lead to urban transmission of malaria, an otherwise rural disease, increasing the human population at risk and complicating malaria elimination efforts. Competition has resulted in habitat segregation of other invasive mosquito species, and our results suggest that it may play a role in determining the distribution of An. stephensi across its invasive range.


Asunto(s)
Aedes , Anopheles , Animales , Humanos , Especies Introducidas , Larva , Temperatura
8.
Proc Biol Sci ; 285(1884)2018 08 15.
Artículo en Inglés | MEDLINE | ID: mdl-30111605

RESUMEN

Temperature is a strong driver of vector-borne disease transmission. Yet, for emerging arboviruses we lack fundamental knowledge on the relationship between transmission and temperature. Current models rely on the untested assumption that Zika virus responds similarly to dengue virus, potentially limiting our ability to accurately predict the spread of Zika. We conducted experiments to estimate the thermal performance of Zika virus (ZIKV) in field-derived Aedes aegypti across eight constant temperatures. We observed strong, unimodal effects of temperature on vector competence, extrinsic incubation period and mosquito survival. We used thermal responses of these traits to update an existing temperature-dependent model to infer temperature effects on ZIKV transmission. ZIKV transmission was optimized at 29°C, and had a thermal range of 22.7°C-34.7°C. Thus, as temperatures move towards the predicted thermal optimum (29°C) owing to climate change, urbanization or seasonality, Zika could expand north and into longer seasons. By contrast, areas that are near the thermal optimum were predicted to experience a decrease in overall environmental suitability. We also demonstrate that the predicted thermal minimum for Zika transmission is 5°C warmer than that of dengue, and current global estimates on the environmental suitability for Zika are greatly over-predicting its possible range.


Asunto(s)
Aedes/fisiología , Cambio Climático , Mosquitos Vectores/fisiología , Temperatura , Infección por el Virus Zika/transmisión , Virus Zika/fisiología , Aedes/virología , Animales , Modelos Biológicos , Mosquitos Vectores/virología , Estaciones del Año , Urbanización
9.
Malar J ; 17(1): 457, 2018 Dec 06.
Artículo en Inglés | MEDLINE | ID: mdl-30522507

RESUMEN

BACKGROUND: The malaria Eradication Research Agenda (malERA) has identified human-to-mosquito transmission of Plasmodium falciparum as a major target for eradication. The cornerstone for identifying and evaluating transmission in the laboratory is standard membrane feeding assays (SMFAs) where mature gametocytes of P. falciparum generated in vitro are offered to mosquitoes as part of a blood-meal. However, propagation of "infectious" gametocytes requires 10-12 days with considerable physico-chemical demands imposed on host RBCs and thus, "fresh" RBCs that are ≤ 1-week old post-collection are generally recommended. However, in addition to the costs, physico-chemical characteristics unique to RBC donors may confound reproducibility and interpretation of SMFAs. Cryogenic storage of RBCs ("cryo-preserved RBCs") is accepted by European and US FDAs as an alternative to refrigeration (4 °C) for preserving RBC "quality" and while cryo-preserved RBCs have been used for in vitro cultures of other Plasmodia and the asexual stages of P. falciparum, none of the studies required RBCs to support parasite development for > 4 days. RESULTS: Using the standard laboratory strain, P. falciparum NF54, 11 SMFAs were performed with RBCs from four separate donors to demonstrate that RBCs cryo-preserved in the gaseous phase of liquid nitrogen (- 196 °C) supported gametocytogenesis in vitro and subsequent gametogenesis in Anopheles stephensi mosquitoes. Overall levels of sporogony in the mosquito, as measured by oocyst and sporozoite prevalence, as well as oocyst burden, from each of the four donors thawed after varying intervals of cryopreservation (1, 4, 8, and 12 weeks) were comparable to using ≤ 1-week old refrigerated RBCs. Lastly, the potential for cryo-preserved RBCs to serve as a suitable alternative substrate is demonstrated for a Cambodian isolate of P. falciparum across two independent SMFAs. CONCLUSIONS: Basic guidelines are presented for integrating cryo-preserved RBCs into an existing laboratory/insectary framework for P. falciparum SMFAs with significant potential for reducing running costs while achieving greater reliability. Lastly, scenarios are discussed where cryo-preserved RBCs may be especially useful in enhancing the understanding and/or providing novel insights into the patterns and processes underlying human-to-mosquito transmission.


Asunto(s)
Criopreservación/métodos , Eritrocitos/parasitología , Gametogénesis/fisiología , Mosquitos Vectores/parasitología , Plasmodium falciparum/fisiología , Animales , Anopheles/parasitología , Investigación Biomédica/métodos , Humanos , Malaria Falciparum/parasitología , Malaria Falciparum/prevención & control , Malaria Falciparum/transmisión , Prevalencia
10.
Proc Natl Acad Sci U S A ; 111(34): 12498-503, 2014 Aug 26.
Artículo en Inglés | MEDLINE | ID: mdl-25114252

RESUMEN

Over evolutionary time, Wolbachia has been repeatedly transferred between host species contributing to the widespread distribution of the symbiont in arthropods. For novel infections to be maintained, Wolbachia must infect the female germ line after being acquired by horizontal transfer. Although mechanistic examples of horizontal transfer exist, there is a poor understanding of factors that lead to successful vertical maintenance of the acquired infection. Using Anopheles mosquitoes (which are naturally uninfected by Wolbachia) we demonstrate that the native mosquito microbiota is a major barrier to vertical transmission of a horizontally acquired Wolbachia infection. After injection into adult Anopheles gambiae, some strains of Wolbachia invade the germ line, but are poorly transmitted to the next generation. In Anopheles stephensi, Wolbachia infection elicited massive blood meal-induced mortality, preventing development of progeny. Manipulation of the mosquito microbiota by antibiotic treatment resulted in perfect maternal transmission at significantly elevated titers of the wAlbB Wolbachia strain in A. gambiae, and alleviated blood meal-induced mortality in A. stephensi enabling production of Wolbachia-infected offspring. Microbiome analysis using high-throughput sequencing identified that the bacterium Asaia was significantly reduced by antibiotic treatment in both mosquito species. Supplementation of an antibiotic-resistant mutant of Asaia to antibiotic-treated mosquitoes completely inhibited Wolbachia transmission and partly contributed to blood meal-induced mortality. These data suggest that the components of the native mosquito microbiota can impede Wolbachia transmission in Anopheles. Incompatibility between the microbiota and Wolbachia may in part explain why some hosts are uninfected by this endosymbiont in nature.


Asunto(s)
Anopheles/microbiología , Wolbachia/crecimiento & desarrollo , Acetobacteraceae/efectos de los fármacos , Acetobacteraceae/crecimiento & desarrollo , Animales , Antibacterianos/farmacología , Evolución Biológica , Transmisión de Enfermedad Infecciosa , Femenino , Transmisión Vertical de Enfermedad Infecciosa , Microbiota/efectos de los fármacos , Óvulo/microbiología , Simbiosis
11.
Proc Biol Sci ; 283(1834)2016 Jul 13.
Artículo en Inglés | MEDLINE | ID: mdl-27412284

RESUMEN

Adult traits of holometabolous insects are shaped by conditions experienced during larval development, which might impact interactions between adult insect hosts and parasites. However, the ecology of larval insects that vector disease remains poorly understood. Here, we used Anopheles stephensi mosquitoes and the human malaria parasite Plasmodium falciparum, to investigate whether larval conditions affect the capacity of adult mosquitoes to transmit malaria. We reared larvae in two groups; one group received a standard laboratory rearing diet, whereas the other received a reduced diet. Emerging adult females were then provided an infectious blood meal. We assessed mosquito longevity, parasite development rate and prevalence of infectious mosquitoes over time. Reduced larval food led to increased adult mortality and caused a delay in parasite development and a slowing in the rate at which parasites invaded the mosquito salivary glands, extending the time it took for mosquitoes to become infectious. Together, these effects increased transmission potential of mosquitoes in the high food regime by 260-330%. Such effects have not, to our knowledge, been shown previously for human malaria and highlight the importance of improving knowledge of larval ecology to better understand vector-borne disease transmission dynamics.


Asunto(s)
Anopheles/parasitología , Dieta , Insectos Vectores/parasitología , Malaria/transmisión , Animales , Anopheles/fisiología , Femenino , Humanos , Insectos Vectores/fisiología , Larva/parasitología , Larva/fisiología , Plasmodium falciparum
12.
bioRxiv ; 2024 Jun 10.
Artículo en Inglés | MEDLINE | ID: mdl-38915528

RESUMEN

Understanding how variation in key abiotic and biotic factors interact at spatial scales relevant for mosquito fitness and population dynamics is crucial for predicting current and future mosquito distributions and abundances, and the transmission potential for human pathogens. However, studies investigating the effects of environmental variation on mosquito traits have investigated environmental factors in isolation or in laboratory experiments that examine constant environmental conditions that often do not occur in the field. To address these limitations, we conducted a semi-field experiment in Athens, Georgia using the invasive Asian tiger mosquito (Aedes albopictus). We selected nine sites that spanned natural variation in impervious surface and vegetation cover to explore effects of the microclimate (temperature and humidity) on mosquitoes. On these sites, we manipulated conspecific larval density at each site. We repeated the experiment in the summer and fall. We then evaluated the effects of land cover, larval density, and time of season, as well as interactive effects, on the mean proportion of females emerging, juvenile development time, size upon emergence, and predicted per capita population growth (i.e., fitness). We found significant effects of larval density, land cover, and season on all response variables. Of most note, we saw strong interactive effects of season and intra-specific density on each response variable, including a non-intuitive decrease in development time with increasing intra-specific competition in the fall. Our study demonstrates that ignoring the interaction between variation in biotic and abiotic variables could reduce the accuracy and precision of models used to predict mosquito population and pathogen transmission dynamics, especially those inferring dynamics at finer-spatial scales across which transmission and control occur.


Para poder predecir la distribución y abundancia de las poblaciones de mosquitos y la transmisión potencial de patógenos a humanos, es crucial comprender cómo factores abióticos y bióticos clave para el éxito reproductivo y la dinámica poblacional de los mosquitos interactúan a escalas relevantes. Sin embargo, los estudios que han investigado los efectos de variables ambientales en las características demográficas de los mosquitos han considerado su efecto de forma aislada o en experimentos de laboratorio bajo condiciones ambientales constantes que, a menudo, no reflejan lo que ocurre en el campo. Para abordar estas limitaciones, llevamos a cabo un experimento de semi-campo en Athens, Georgia, utilizando el mosquito invasor tigre asiático (Aedes albopictus). Seleccionamos nueve sitios que abarcaban variaciones naturales en la superficie impermeable y cobertura vegetal para explorar los efectos del microclima (temperatura y humedad) en los mosquitos. También manipulamos la densidad de larvas de tigre asiático en dos experimentos que fueron realizados en el verano y otoño. Evaluamos los efectos de la cobertura vegetal, la densidad de larvas, la temporada climática, y la interacción entre estas variables en la proporción de hembras que emergieron, el tiempo de desarrollo de las larvas, el tamaño al momento de la emergencia, y el crecimiento demográfico per cápita previsto (éxito reproductivo). Encontramos efectos significativos de la densidad de larvas, la variación de la cobertura vegetal y la estación del año en todas las variables de respuesta. Más notablemente, observamos un fuerte efecto de la interacción entre la temporada climática y la densidad de larvas en todas las variables de respuesta, incluyendo una disminución no intuitiva en el tiempo de desarrollo con el aumento de la competencia intraespecífica en el otoño. Nuestro estudio evidencia que ignorar la interacción entre variables abióticas y bióticas podría reducir la exactitud y precisión de los modelos utilizados para predecir las dinámicas de las poblaciones de mosquitos, y por tanto, de la transmisión de patógenos. Esto, especialmente en modelos que infieren estas dinámicas a escalas espaciales más finas, en las cuales ocurre la transmisión y el control.

13.
Proc Biol Sci ; 280(1770): 20132030, 2013 Nov 07.
Artículo en Inglés | MEDLINE | ID: mdl-24048159

RESUMEN

Considerable research effort has been directed at understanding the genetic and molecular basis of mosquito innate immune mechanisms. Whether environmental factors interact with these mechanisms to shape overall resistance remains largely unexplored. Here, we examine how changes in mean ambient temperature, diurnal temperature fluctuation and time of day of infection affected the immunity and resistance of Anopheles stephensi to infection with Escherichia coli. We used quantitative PCR to estimate the gene expression of three immune genes in response to challenge with heat-killed E. coli. We also infected mosquitoes with live E. coli and ran bacterial growth assays to quantify host resistance. Both mosquito immune parameters and resistance were directly affected by mean temperature, diurnal temperature fluctuation and time of day of infection. Furthermore, there was a suite of complex two- and three-way interactions yielding idiosyncratic phenotypic variation under different environmental conditions. The results demonstrate mosquito immunity and resistance to be strongly influenced by a complex interplay of environmental variables, challenging the interpretation of the very many mosquito immune studies conducted under standard laboratory conditions.


Asunto(s)
Anopheles/genética , Anopheles/inmunología , Regulación de la Expresión Génica , Proteínas de Insectos/genética , Animales , Anopheles/microbiología , Cecropinas/genética , Cecropinas/metabolismo , Defensinas/genética , Defensinas/metabolismo , Escherichia coli/fisiología , Femenino , Proteínas de Insectos/metabolismo , Óxido Nítrico Sintasa/genética , Óxido Nítrico Sintasa/metabolismo , Reacción en Cadena de la Polimerasa , Temperatura , Factores de Tiempo
14.
Proc Biol Sci ; 280(1763): 20130711, 2013 Jul 22.
Artículo en Inglés | MEDLINE | ID: mdl-23698008

RESUMEN

Previous studies have suggested that Plasmodium parasites can manipulate mosquito feeding behaviours such as probing, persistence and engorgement rate in order to enhance transmission success. Here, we broaden analysis of this 'manipulation phenotype' to consider proximate foraging behaviours, including responsiveness to host odours and host location. Using Anopheles stephensi and Plasmodium yoelii as a model system, we demonstrate that mosquitoes with early stage infections (i.e. non-infectious oocysts) exhibit reduced attraction to a human host, whereas those with late-stage infections (i.e. infectious sporozoites) exhibit increased attraction. These stage-specific changes in behaviour were paralleled by changes in the responsiveness of mosquito odourant receptors, providing a possible neurophysiological mechanism for the responses. However, we also found that both the behavioural and neurophysiological changes could be generated by immune challenge with heat-killed Escherichia coli and were thus not tied explicitly to the presence of malaria parasites. Our results support the hypothesis that the feeding behaviour of female mosquitoes is altered by Plasmodium, but question the extent to which this is owing to active manipulation by malaria parasites of host behaviour.


Asunto(s)
Anopheles/parasitología , Conducta Alimentaria/fisiología , Interacciones Huésped-Parásitos , Insectos Vectores/parasitología , Plasmodium yoelii/fisiología , Animales , Anopheles/fisiología , Femenino , Humanos , Insectos Vectores/fisiología , Malaria/parasitología , Malaria/transmisión , Masculino , Plasmodium yoelii/patogenicidad , Esporozoítos/fisiología
15.
Ecology ; 103(8): e3685, 2022 08.
Artículo en Inglés | MEDLINE | ID: mdl-35315521

RESUMEN

Extrinsic environmental factors influence the spatiotemporal dynamics of many organisms, including insects that transmit the pathogens responsible for vector-borne diseases (VBDs). Temperature is an especially important constraint on the fitness of a wide variety of ectothermic insects. A mechanistic understanding of how temperature impacts traits of ectotherms, and thus the distribution of ectotherms and vector-borne infections, is key to predicting the consequences of climate change on transmission of VBDs like malaria. However, the response of transmission to temperature and other drivers is complex, as thermal traits of ectotherms are typically nonlinear, and they interact to determine transmission constraints. In this study, we assess and compare the effect of temperature on the transmission of two malaria parasites, Plasmodium falciparum and Plasmodium vivax, by two malaria vector species, Anopheles gambiae and Anopheles stephensi. We model the nonlinear responses of temperature dependent mosquito and parasite traits (mosquito development rate, bite rate, fecundity, proportion of eggs surviving to adulthood, vector competence, mortality rate, and parasite development rate) and incorporate these traits into a suitability metric based on a model for the basic reproductive number across temperatures. Our model predicts that the optimum temperature for transmission suitability is similar for the four mosquito-parasite combinations assessed in this study, but may differ at the thermal limits. More specifically, we found significant differences in the upper thermal limit between parasites spread by the same mosquito (A. stephensi) and between mosquitoes carrying P. falciparum. In contrast, at the lower thermal limit the significant differences were primarily between the mosquito species that both carried the same pathogen (e.g., A. stephensi and A. gambiae both with P. falciparum). Using prevalence data, we show that the transmission suitability metric ST$$ S(T) $$ calculated from our mechanistic model is consistent with observed P. falciparum prevalence in Africa and Asia but is equivocal for P. vivax prevalence in Asia, and inconsistent with P. vivax prevalence in Africa. We mapped risk to illustrate the number of months various areas in Africa and Asia predicted to be suitable for malaria transmission based on this suitability metric. This mapping provides spatially explicit predictions for suitability and transmission risk.


Asunto(s)
Anopheles , Malaria Falciparum , Malaria Vivax , Temperatura , Animales , Anopheles/parasitología , Anopheles/fisiología , Malaria Falciparum/epidemiología , Malaria Falciparum/transmisión , Malaria Vivax/epidemiología , Malaria Vivax/transmisión , Mosquitos Vectores
16.
Anal Sci Adv ; 3(1-2): 47-53, 2022 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-38716056

RESUMEN

Mosquito-borne pathogens, including malaria, Zika, dengue, and chikungunya continue to be a major public health concern globally. Based on the understanding that only older female mosquitoes are infectious and represent a risk to human health, scientists have sought to age-grade mosquitoes for decades. To date, however, no reliable, cost-effective and practical methods exist to age older mosquitoes despite the tremendous epidemiological value of this approach. This study is the first attempt to develop a surface-enhanced Raman spectroscopic (SERS) method to age mosquitoes. The water extracts of Aedes aegypti mosquitoes aged 0-22 days were mixed with silver nanoparticles. The SERS spectra, which were analysed by principal component analysis and partial least square (PLS), demonstrated the capability of this approach to predict the calendar age of mosquitoes between 0 and 22 days with the coefficient of correlation (R) = 0.994 and 0.990 for PLS model calibration and validation, respectively. Spectral analysis with both SERS and infrared spectroscopy revealed the key biological sources leading to changes in spectra allowing mosquito age-grading is adenine-containing compounds and proteins. In addition, we evaluated the impact of two arthropod-borne pathogen deactivating pre-treatments (bleach and ethanol) on the discrimination capability of the SERS approach. The result shows the ethanol treatment has the potential to enhance the discrimination capability and the safety of the approach. This study represents the first step towards developing the SERS approach as a quick, reliable and field-deployable method for mosquito age-grading, which would significantly improve the effectiveness of vector-borne disease monitoring and prevention.

18.
Commun Biol ; 4(1): 723, 2021 06 11.
Artículo en Inglés | MEDLINE | ID: mdl-34117363

RESUMEN

Harmonic convergence is a potential cue, female mosquitoes use to choose male mates. However, very little is known about the benefits this choice confers to offspring performance. Using Aedes aegypti (an important vector of human disease), we investigated whether offspring of converging parental pairs showed differences in immune competence compared to offspring derived from non-converging parental pairs. Here we show that harmonic convergence, along with several other interacting factors (sex, age, reproductive, and physiological status), significantly shaped offspring immune responses (melanization and response to a bacterial challenge). Harmonic convergence had a stronger effect on the immune response of male offspring than on female offspring. Further, female offspring from converging parental pairs disseminated dengue virus more quickly than offspring derived from non-converging parental pairs. Our results provide insight into a wide range of selective pressures shaping mosquito immune function and could have important implications for disease transmission and control.


Asunto(s)
Aedes/fisiología , Acústica , Aedes/inmunología , Aedes/virología , Factores de Edad , Animales , Virus del Dengue/fisiología , Femenino , Masculino , Reproducción/fisiología , Factores Sexuales , Conducta Sexual Animal/fisiología
19.
J R Soc Interface ; 18(178): 20210165, 2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-33947225

RESUMEN

When a rare pathogen emerges to cause a pandemic, it is critical to understand its dynamics and the impact of mitigation measures. We use experimental data to parametrize a temperature-dependent model of Zika virus (ZIKV) transmission dynamics and analyse the effects of temperature variability and control-related parameters on the basic reproduction number (R0) and the final epidemic size of ZIKV. Sensitivity analyses show that these two metrics are largely driven by different parameters, with the exception of temperature, which is the dominant driver of epidemic dynamics in the models. Our R0 estimate has a single optimum temperature (≈30°C), comparable to other published results (≈29°C). However, the final epidemic size is maximized across a wider temperature range, from 24 to 36°C. The models indicate that ZIKV is highly sensitive to seasonal temperature variation. For example, although the model predicts that ZIKV transmission cannot occur at a constant temperature below 23°C (≈ average annual temperature of Rio de Janeiro, Brazil), the model predicts substantial epidemics for areas with a mean temperature of 20°C if there is seasonal variation of 10°C (≈ average annual temperature of Tampa, Florida). This suggests that the geographical range of ZIKV is wider than indicated from static R0 models, underscoring the importance of climate dynamics and variation in the context of broader climate change on emerging infectious diseases.


Asunto(s)
Infección por el Virus Zika , Virus Zika , Brasil , Florida , Humanos , Mosquitos Vectores , Temperatura , Infección por el Virus Zika/epidemiología
20.
PLoS Negl Trop Dis ; 15(7): e0009540, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-34214096

RESUMEN

BACKGROUND: The mosquito Aedes aegypti is a medically important, globally distributed vector of the viruses that cause dengue, yellow fever, chikungunya, and Zika. Although reproduction and mate choice are key components of vector population dynamics and control, our understanding of the mechanisms of sexual selection in mosquitoes remains poor. In "good genes" models of sexual selection, females use male cues as an indicator of both mate and offspring genetic quality. Recent studies in Ae. aegypti provide evidence that male wingbeats may signal aspects of offspring quality and performance during mate selection in a process known as harmonic convergence. However, the extent to which harmonic convergence may signal overall inherent quality of mates and their offspring remains unknown. METHODOLOGY/PRINCIPAL FINDINGS: To examine this, we measured the relationship between acoustic signaling and a broad panel of parent and offspring fitness traits in two generations of field-derived Ae. aegypti originating from dengue-endemic field sites in Thailand. Our data show that in this population of mosquitoes, harmonic convergence does not signal male fertility, female fecundity, or male flight performance traits, which despite displaying robust variability in both parents and their offspring were only weakly heritable. CONCLUSIONS/SIGNIFICANCE: Together, our findings suggest that vector reproductive control programs should treat harmonic convergence as an indicator of some, but not all aspects of inherent quality, and that sexual selection likely affects Ae. aegypti in a trait-, population-, and environment-dependent manner.


Asunto(s)
Aedes/fisiología , Mosquitos Vectores/fisiología , Aedes/genética , Animales , Femenino , Masculino , Control de Mosquitos , Dinámica Poblacional , Reproducción , Conducta Sexual Animal , Tailandia
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA