Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 36
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Nature ; 563(7732): 501-507, 2018 11.
Artículo en Inglés | MEDLINE | ID: mdl-30429615

RESUMEN

Female Aedes aegypti mosquitoes infect more than 400 million people each year with dangerous viral pathogens including dengue, yellow fever, Zika and chikungunya. Progress in understanding the biology of mosquitoes and developing the tools to fight them has been slowed by the lack of a high-quality genome assembly. Here we combine diverse technologies to produce the markedly improved, fully re-annotated AaegL5 genome assembly, and demonstrate how it accelerates mosquito science. We anchored physical and cytogenetic maps, doubled the number of known chemosensory ionotropic receptors that guide mosquitoes to human hosts and egg-laying sites, provided further insight into the size and composition of the sex-determining M locus, and revealed copy-number variation among glutathione S-transferase genes that are important for insecticide resistance. Using high-resolution quantitative trait locus and population genomic analyses, we mapped new candidates for dengue vector competence and insecticide resistance. AaegL5 will catalyse new biological insights and intervention strategies to fight this deadly disease vector.


Asunto(s)
Aedes/genética , Infecciones por Arbovirus/virología , Arbovirus , Genoma de los Insectos/genética , Genómica/normas , Control de Insectos , Mosquitos Vectores/genética , Mosquitos Vectores/virología , Aedes/virología , Animales , Infecciones por Arbovirus/transmisión , Arbovirus/aislamiento & purificación , Variaciones en el Número de Copia de ADN/genética , Virus del Dengue/aislamiento & purificación , Femenino , Variación Genética/genética , Genética de Población , Glutatión Transferasa/genética , Resistencia a los Insecticidas/efectos de los fármacos , Masculino , Anotación de Secuencia Molecular , Familia de Multigenes/genética , Piretrinas/farmacología , Estándares de Referencia , Procesos de Determinación del Sexo/genética
2.
Euro Surveill ; 29(13)2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-38551097

RESUMEN

In 2023, dengue virus serotype 2 (DENV2) affected most French overseas territories. In the French Caribbean Islands, viral circulation continues with > 30,000 suspected infections by March 2024. Genome sequence analysis reveals that the epidemic lineage in the French Caribbean islands has also become established in French Guiana but not Réunion. It has moreover seeded autochthonous circulation events in mainland France. To guide prevention of further inter-territorial spread and DENV introduction in non-endemic settings, continued molecular surveillance and mosquito control are essential.


Asunto(s)
Epidemias , Humanos , Guyana Francesa/epidemiología , Epidemiología Molecular , Indias Occidentales/epidemiología , Francia/epidemiología
3.
PLoS Genet ; 16(5): e1008794, 2020 05.
Artículo en Inglés | MEDLINE | ID: mdl-32463828

RESUMEN

Although specific interactions between host and pathogen genotypes have been well documented in invertebrates, the identification of host genes involved in discriminating pathogen genotypes remains a challenge. In the mosquito Aedes aegypti, the main dengue virus (DENV) vector worldwide, statistical associations between host genetic markers and DENV types or strains were previously detected, but the host genes underlying this genetic specificity have not been identified. In particular, it is unknown whether DENV type- or strain-specific resistance relies on allelic variants of the same genes or on distinct gene sets. Here, we investigated the genetic architecture of DENV resistance in a population of Ae. aegypti from Bakoumba, Gabon, which displays a stronger resistance phenotype to DENV type 1 (DENV-1) than to DENV type 3 (DENV-3) infection. Following experimental exposure to either DENV-1 or DENV-3, we sequenced the exomes of large phenotypic pools of mosquitoes that are either resistant or susceptible to each DENV type. Using variation in single-nucleotide polymorphism (SNP) frequencies among the pools, we computed empirical p values based on average gene scores adjusted for the differences in SNP counts, to identify genes associated with infection in a DENV type-specific manner. Among the top 5% most significant genes, 263 genes were significantly associated with resistance to both DENV-1 and DENV-3, 287 genes were only associated with DENV-1 resistance and 290 were only associated with DENV-3 resistance. The shared significant genes were enriched in genes with ATP binding activity and sulfur compound transmembrane transporter activity, whereas the genes uniquely associated with DENV-3 resistance were enriched in genes with zinc ion binding activity. Together, these results indicate that specific resistance to different DENV types relies on largely non-overlapping sets of genes in this Ae. aegypti population and pave the way for further mechanistic studies.


Asunto(s)
Aedes/genética , Virus del Dengue/clasificación , Resistencia a la Enfermedad , Secuenciación del Exoma/métodos , Proteínas de Insectos/genética , Aedes/virología , Animales , Células Cultivadas , Virus del Dengue/patogenicidad , Femenino , Gabón , Genotipo , Fenotipo , Polimorfismo de Nucleótido Simple , ARN Viral/genética , Especificidad de la Especie
4.
PLoS Pathog ; 16(12): e1009068, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-33382858

RESUMEN

Originating from African forests, Zika virus (ZIKV) has now emerged worldwide in urbanized areas, mainly transmitted by Aedes aegypti mosquitoes. Although Aedes albopictus can transmit ZIKV experimentally and was suspected to be a ZIKV vector in Central Africa, the potential of this species to sustain virus transmission was yet to be uncovered until the end of 2019, when several autochthonous transmissions of the virus vectored by Ae. albopictus occurred in France. Aside from these few locally acquired ZIKV infections, most territories colonized by Ae. albopictus have been spared so far. The risk level of ZIKV emergence in these areas remains however an open question. To assess Ae. albopictus' vector potential for ZIKV and identify key virus outbreak predictors, we built a complete framework using the complementary combination of (i) dose-dependent experimental Ae. albopictus exposure to ZIKV followed by time-dependent assessment of infection and systemic infection rates, (ii) modeling of intra-human ZIKV viremia dynamics, and (iii) in silico epidemiological simulations using an Agent-Based Model. The highest risk of transmission occurred during the pre-symptomatic stage of the disease, at the peak of viremia. At this dose, mosquito infection probability was estimated to be 20%, and 21 days were required to reach the median systemic infection rates. Mosquito population origin, either temperate or tropical, had no impact on infection rates or intra-host virus dynamic. Despite these unfavorable characteristics for transmission, Ae. albopictus was still able to trigger and yield large outbreaks in a simulated environment in the presence of sufficiently high mosquito biting rates. Our results reveal a low but existing epidemic potential of Ae. albopictus for ZIKV, that might explain the absence of large scale ZIKV epidemics so far in territories occupied only by Ae. albopictus. They nevertheless support active surveillance and eradication programs in these territories to maintain the risk of emergence to a low level.


Asunto(s)
Mosquitos Vectores/metabolismo , Mosquitos Vectores/virología , Infección por el Virus Zika/transmisión , Aedes/metabolismo , Aedes/virología , Animales , Brotes de Enfermedades , Vectores de Enfermedades , Epidemias , Humanos , Modelos Teóricos , Saliva/virología , Carga Viral , Viremia/transmisión , Virus Zika/patogenicidad , Infección por el Virus Zika/epidemiología , Infección por el Virus Zika/virología
5.
Med Vet Entomol ; 36(4): 486-495, 2022 12.
Artículo en Inglés | MEDLINE | ID: mdl-35762523

RESUMEN

The urban mosquito species Aedes aegypti is the main vector of arboviruses worldwide. Mosquito control with insecticides is the most prevalent method for preventing transmission in the absence of effective vaccines and available treatments; however, the extensive use of insecticides has led to the development of resistance in mosquito populations throughout the world, and the number of epidemics caused by arboviruses has increased. Three mosquito lines with different resistance profiles to deltamethrin were isolated in French Guiana, including one with the I1016 knock-down resistant allele. Significant differences were observed in the cumulative proportion of mosquitoes with a disseminated chikungunya virus infection over time across these lines. In addition, some genes related to resistance (CYP6BB2, CYP6N12, GST2, trypsin) were variably overexpressed in the midgut at 7 days after an infectious bloodmeal in these three lines. Our work shows that vector competence for chikungunya virus varied between Ae. aegypti laboratory lines with different deltamethrin resistance profiles. More accurate verification of the functional association between insecticide resistance and vector competence remains to be demonstrated.


Asunto(s)
Aedes , Arbovirus , Virus Chikungunya , Insecticidas , Animales , Insecticidas/farmacología , Mosquitos Vectores , Resistencia a los Insecticidas/genética
6.
PLoS Pathog ; 14(7): e1007187, 2018 07.
Artículo en Inglés | MEDLINE | ID: mdl-30005085

RESUMEN

The kinetics of arthropod-borne virus (arbovirus) transmission by their vectors have long been recognized as a powerful determinant of arbovirus epidemiology. The time interval between virus acquisition and transmission by the vector, termed extrinsic incubation period (EIP), combines with vector mortality rate and vector competence to determine the proportion of infected vectors that eventually become infectious. However, the dynamic nature of this process, and the amount of natural variation in transmission kinetics among arbovirus strains, are poorly documented empirically and are rarely considered in epidemiological models. Here, we combine newly generated empirical measurements in vivo and outbreak simulations in silico to assess the epidemiological significance of genetic variation in dengue virus (DENV) transmission kinetics by Aedes aegypti mosquitoes. We found significant variation in the dynamics of systemic mosquito infection, a proxy for EIP, among eight field-derived DENV isolates representing the worldwide diversity of recently circulating type 1 strains. Using a stochastic agent-based model to compute time-dependent individual transmission probabilities, we predict that the observed variation in systemic mosquito infection kinetics may drive significant differences in the probability of dengue outbreak and the number of human infections. Our results demonstrate that infection dynamics in mosquitoes vary among wild-type DENV isolates and that this variation potentially affects the risk and magnitude of dengue outbreaks. Our quantitative assessment of DENV genetic variation in transmission kinetics contributes to improve our understanding of heterogeneities in arbovirus epidemiological dynamics.


Asunto(s)
Culicidae/virología , Virus del Dengue/genética , Dengue/genética , Dengue/transmisión , Mosquitos Vectores/virología , Animales , Variación Genética
7.
PLoS Genet ; 12(6): e1006111, 2016 06.
Artículo en Inglés | MEDLINE | ID: mdl-27304978

RESUMEN

Due to their error-prone replication, RNA viruses typically exist as a diverse population of closely related genomes, which is considered critical for their fitness and adaptive potential. Intra-host demographic fluctuations that stochastically reduce the effective size of viral populations are a challenge to maintaining genetic diversity during systemic host infection. Arthropod-borne viruses (arboviruses) traverse several anatomical barriers during infection of their arthropod vectors that are believed to impose population bottlenecks. These anatomical barriers have been associated with both maintenance of arboviral genetic diversity and alteration of the variant repertoire. Whether these patterns result from stochastic sampling (genetic drift) rather than natural selection, and/or from the influence of vector genetic heterogeneity has not been elucidated. Here, we used deep sequencing of full-length viral genomes to monitor the intra-host evolution of a wild-type dengue virus isolate during infection of several mosquito genetic backgrounds. We estimated a bottleneck size ranging from 5 to 42 founding viral genomes at initial midgut infection, irrespective of mosquito genotype, resulting in stochastic reshuffling of the variant repertoire. The observed level of genetic diversity increased following initial midgut infection but significantly differed between mosquito genetic backgrounds despite a similar initial bottleneck size. Natural selection was predominantly negative (purifying) during viral population expansion. Taken together, our results indicate that dengue virus intra-host genetic diversity in the mosquito vector is shaped by genetic drift and purifying selection, and point to a novel role for vector genetic factors in the genetic breadth of virus populations during infection. Identifying the evolutionary forces acting on arboviral populations within their arthropod vector provides novel insights into arbovirus evolution.


Asunto(s)
Aedes/virología , Virus del Dengue/genética , Dengue/transmisión , Flujo Genético , Genoma Viral/genética , Interacciones Huésped-Patógeno , Aedes/genética , Animales , Secuencia de Bases , Dengue/virología , Evolución Molecular , Femenino , Variación Genética/genética , Secuenciación de Nucleótidos de Alto Rendimiento , Insectos Vectores/genética , Masculino , ARN Viral/genética , Análisis de Secuencia de ARN , Replicación Viral
8.
PLoS Genet ; 9(8): e1003621, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23935524

RESUMEN

Specific interactions between host genotypes and pathogen genotypes (G×G interactions) are commonly observed in invertebrate systems. Such specificity challenges our current understanding of invertebrate defenses against pathogens because it contrasts the limited discriminatory power of known invertebrate immune responses. Lack of a mechanistic explanation, however, has questioned the nature of host factors underlying G×G interactions. In this study, we aimed to determine whether G×G interactions observed between dengue viruses and their Aedes aegypti vectors in nature can be mapped to discrete loci in the mosquito genome and to document their genetic architecture. We developed an innovative genetic mapping strategy to survey G×G interactions using outbred mosquito families that were experimentally exposed to genetically distinct isolates of two dengue virus serotypes derived from human patients. Genetic loci associated with vector competence indices were detected in multiple regions of the mosquito genome. Importantly, correlation between genotype and phenotype was virus isolate-specific at several of these loci, indicating G×G interactions. The relatively high percentage of phenotypic variation explained by the markers associated with G×G interactions (ranging from 7.8% to 16.5%) is consistent with large-effect host genetic factors. Our data demonstrate that G×G interactions between dengue viruses and mosquito vectors can be assigned to physical regions of the mosquito genome, some of which have a large effect on the phenotype. This finding establishes the existence of tangible host genetic factors underlying specific interactions between invertebrates and their pathogens in a natural system. Fine mapping of the uncovered genetic loci will elucidate the molecular mechanisms of mosquito-virus specificity.


Asunto(s)
Aedes/genética , Virus del Dengue/genética , Dengue/genética , Insectos Vectores/genética , Aedes/virología , Animales , Mapeo Cromosómico , Dengue/patología , Virus del Dengue/patogenicidad , Genotipo , Interacciones Huésped-Patógeno/genética , Humanos , Insectos Vectores/virología , Sitios de Carácter Cuantitativo/genética
9.
Proc Biol Sci ; 281(1792)2014 Oct 07.
Artículo en Inglés | MEDLINE | ID: mdl-25122228

RESUMEN

Interactions between pathogens and their insect vectors in nature are under the control of both genetic and non-genetic factors, yet most studies on mosquito vector competence for human pathogens are conducted in laboratory systems that do not consider genetic and/or environmental variability. Evaluating the risk of emergence of arthropod-borne viruses (arboviruses) of public health importance such as chikungunya virus (CHIKV) requires a more realistic appraisal of genetic and environmental contributions to vector competence. In particular, sources of variation do not necessarily act independently and may combine in the form of interactions. Here, we measured CHIKV transmission potential by the mosquito Aedes albopictus in all combinations of six worldwide vector populations, two virus strains and two ambient temperatures (20°C and 28°C). Overall, CHIKV transmission potential by Ae. albopictus strongly depended on the three-way combination of mosquito population, virus strain and temperature. Such genotype-by-genotype-by-environment (G × G × E) interactions question the relevance of vector competence studies conducted with a simpler set of conditions. Our results highlight the need to account for the complex interplay between vectors, pathogens and environmental factors to accurately assess the potential of vector-borne diseases to emerge.


Asunto(s)
Aedes/genética , Aedes/virología , Infecciones por Alphavirus/transmisión , Virus Chikungunya/genética , Insectos Vectores/genética , Insectos Vectores/virología , Temperatura , Animales , Genotipo , Ratones
10.
Antiviral Res ; 225: 105858, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38490342

RESUMEN

Chikungunya virus (CHIKV) is a mosquito-borne virus transmitted by Aedes mosquitoes. While there are no antiviral therapies currently available to treat CHIKV infections, several licensed oral drugs have shown significant anti-CHIKV activity in cells and in mouse models. However, the efficacy in mosquitoes has not yet been assessed. Such cross-species antiviral activity could be favorable, since virus inhibition in the mosquito vector might prevent further transmission to vertebrate hosts. Here, we explored the antiviral effect of ß-d-N4-hydroxycytidine (NHC, EIDD-1931), the active metabolite of molnupiravir, on CHIKV replication in Aedes aegypti mosquitoes. Antiviral assays in mosquito cells and in ex vivo cultured mosquito guts showed that NHC had significant antiviral activity against CHIKV. Exposure to a clinically relevant concentration of NHC did not affect Ae. aegypti lifespan when delivered via a bloodmeal, but it slightly reduced the number of eggs developed in the ovaries. When mosquitoes were exposed to a blood meal containing both CHIKV and NHC, the compound did not significantly reduce virus infection and dissemination in the mosquitoes. This was confirmed by modelling and could be explained by pharmacokinetic analysis, which revealed that by 6 h post-blood-feeding, 90% of NHC had been cleared from the mosquito bodies. Our data show that NHC inhibited CHIKV replication in mosquito cells and gut tissue, but not in vivo when mosquitoes were provided with a CHIKV-infectious bloodmeal spiked with NHC. The pipeline presented in this study offers a suitable approach to identify anti-arboviral drugs that may impede replication in mosquitoes.


Asunto(s)
Aedes , Fiebre Chikungunya , Virus Chikungunya , Citidina/análogos & derivados , Animales , Ratones , Virus Chikungunya/fisiología , Replicación Viral , Antivirales
11.
J Med Entomol ; 61(3): 818-823, 2024 May 13.
Artículo en Inglés | MEDLINE | ID: mdl-38408180

RESUMEN

Arboviruses can be difficult to detect in the field due to relatively low prevalence in mosquito populations. The discovery that infected mosquitoes can release viruses in both their saliva and excreta gave rise to low-cost methods for the detection of arboviruses during entomological surveillance. We implemented both saliva and excreta-based entomological surveillance during the emergence of Zika virus (ZIKV) in French Guiana in 2016 by trapping mosquitoes around households of symptomatic cases with confirmed ZIKV infection. ZIKV was detected in mosquito excreta and not in mosquito saliva in 1 trap collection out of 85 (1.2%). One female Ae. aegypti L. (Diptera: Culicidae) was found with a ZIKV systemic infection in the corresponding trap. The lag time between symptom onset in a ZIKV-infected individual living near the trap site and ZIKV detection in this mosquito was 1 wk. These results highlight the potential of detection in excreta from trapped mosquitoes as a sensitive and cost-effective method to non invasively detect arbovirus circulation.


Asunto(s)
Aedes , Heces , Saliva , Virus Zika , Animales , Guyana Francesa , Virus Zika/aislamiento & purificación , Heces/virología , Femenino , Aedes/virología , Saliva/virología , Mosquitos Vectores/virología , Masculino , Infección por el Virus Zika/transmisión
13.
Commun Biol ; 6(1): 646, 2023 06 16.
Artículo en Inglés | MEDLINE | ID: mdl-37328568

RESUMEN

Chemical control of disease vectoring mosquitoes Aedes albopictus and Aedes aegypti is costly, unsustainable, and increasingly ineffective due to the spread of insecticide resistance. The Sterile Insect Technique is a valuable alternative but is limited by slow, error-prone, and wasteful sex-separation methods. Here, we present four Genetic Sexing Strains (two for each Aedes species) based on fluorescence markers linked to the m and M sex loci, allowing for the isolation of transgenic males. Furthermore, we demonstrate how combining these sexing strains enables the production of non-transgenic males. In a mass-rearing facility, 100,000 first instar male larvae could be sorted in under 1.5 h with an estimated 0.01-0.1% female contamination on a single machine. Cost-efficiency analyses revealed that using these strains could result in important savings while setting up and running a mass-rearing facility. Altogether, these Genetic Sexing Strains should enable a major upscaling in control programmes against these important vectors.


Asunto(s)
Aedes , Animales , Masculino , Femenino , Aedes/genética , Animales Modificados Genéticamente , Larva/genética , Mosquitos Vectores/genética , Resistencia a los Insecticidas
14.
PLoS Negl Trop Dis ; 17(6): e0011415, 2023 06.
Artículo en Inglés | MEDLINE | ID: mdl-37315094

RESUMEN

BACKGROUND: The cutaneous leishmaniasis (CL) incubation period (IP) is defined as the time between parasite inoculation by sandfly bite and the onset of the first CL lesion. IP distribution is difficult to assess for CL because the date of exposure to an infectious bite cannot be accurately determined in endemic areas. IP current estimates for CL range from 14 days to several months with a median around 30-60 days, as established by a few previous studies in both New and Old Worlds. METHODOLOGY: We estimated CL incubation period distribution using time-to-event models adapted to interval-censored data based on declared date of travels from symptomatic military personnel living in non-endemic areas that were exposed during their short stays in French Guiana (FG) between January 2001 and December 2021. PRINCIPAL FINDINGS: A total of 180 patients were included, of which 176 were men (97.8%), with a median age of 26 years. When recorded, the parasite species was always Leishmania guyanensis (31/180, 17.2%). The main periods of CL diagnosis spread from November to January (84/180, 46.7%) and over March-April (54/180, 30.0%). The median IP was estimated at 26.2 days (95% Credible Level, 23.8-28.7 days) using a Bayesian accelerated failure-time regression model. Estimated IP did not exceed 62.1 days (95% CI, 56-69.8 days) in 95% of cases (95th percentile). Age, gender, lesion number, lesion evolution and infection date did not significantly modify the IP. However, disseminated CL was significantly associated with a 2.8-fold shortening of IP. CONCLUSIONS: This work suggests that the CL IP distribution in French Guiana is shorter and more restricted than anticipated. As the incidence of CL in FG usually peaks in January and March, these findings suggest that patients are contaminated at the start of the rainy season.


Asunto(s)
Leishmania guyanensis , Leishmaniasis Cutánea , Masculino , Humanos , Adulto , Femenino , Guyana Francesa/epidemiología , Teorema de Bayes , Periodo de Incubación de Enfermedades Infecciosas , Leishmaniasis Cutánea/parasitología
15.
Mol Ecol Resour ; 23(2): 410-423, 2023 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-36161270

RESUMEN

Emerging and endemic mosquito-borne viruses can be difficult to detect and monitor because they often cause asymptomatic infections in human or vertebrate animals or cause nonspecific febrile illness with a short recovery waiting period. Some of these pathogens circulate into complex cryptic cycles involving several animal species as reservoir or amplifying hosts. Detection of cases in vertebrate hosts can be complemented by entomological surveillance, but this method is not adapted to low infection rates in mosquito populations that typically occur in low or nonendemic areas. We identified West Nile virus circulation in Camargue, a wetland area in South of France, using a cost-effective xenomonitoring method based on the molecular detection of virus in excreta from trapped mosquitoes. We also succeeded at identifying the mosquito species community on several sampling sites, together with the vertebrate hosts on which they fed prior to being captured using amplicon-based metabarcoding on mosquito excreta without processing any mosquitoes. Mosquito excreta-based virus surveillance can complement standard surveillance methods because it is cost-effective and does not require personnel with a strong background in entomology. This strategy can also be used to noninvasively explore the ecological network underlying arbovirus circulation.


Asunto(s)
Arbovirus , Culicidae , Flavivirus , Virus del Nilo Occidental , Animales , Humanos , Arbovirus/genética , Biodiversidad
16.
BMC Genomics ; 13: 614, 2012 Nov 13.
Artículo en Inglés | MEDLINE | ID: mdl-23148599

RESUMEN

BACKGROUND: Antibody responses against Anopheles salivary proteins can indicate individual exposure to bites of malaria vectors. The extent to which these salivary proteins are species-specific is not entirely resolved. Thus, a better knowledge of the diversity among salivary protein repertoires from various malaria vector species is necessary to select relevant genus-, subgenus- and/or species-specific salivary antigens. Such antigens could be used for quantitative (mosquito density) and qualitative (mosquito species) immunological evaluation of malaria vectors/host contact. In this study, salivary gland protein repertoires (sialomes) from several Anopheles species were compared using in silico analysis and proteomics. The antigenic diversity of salivary gland proteins among different Anopheles species was also examined. RESULTS: In silico analysis of secreted salivary gland protein sequences retrieved from an NCBInr database of six Anopheles species belonging to the Cellia subgenus (An. gambiae, An. arabiensis, An. stephensi and An. funestus) and Nyssorhynchus subgenus (An. albimanus and An. darlingi) displayed a higher degree of similarity compared to salivary proteins from closely related Anopheles species. Additionally, computational hierarchical clustering allowed identification of genus-, subgenus- and species-specific salivary proteins. Proteomic and immunoblot analyses performed on salivary gland extracts from four Anopheles species (An. gambiae, An. arabiensis, An. stephensi and An. albimanus) indicated that heterogeneity of the salivary proteome and antigenic proteins was lower among closely related anopheline species and increased with phylogenetic distance. CONCLUSION: This is the first report on the diversity of the salivary protein repertoire among species from the Anopheles genus at the protein level. This work demonstrates that a molecular diversity is exhibited among salivary proteins from closely related species despite their common pharmacological activities. The involvement of these proteins as antigenic candidates for genus-, subgenus- or species-specific immunological evaluation of individual exposure to Anopheles bites is discussed.


Asunto(s)
Anopheles/genética , Insectos Vectores/genética , Filogenia , Proteoma/genética , Glándulas Salivales/metabolismo , Animales , Anopheles/metabolismo , Secuencia de Bases , Análisis por Conglomerados , Biología Computacional , Electroforesis en Gel de Poliacrilamida , Immunoblotting , Insectos Vectores/metabolismo , Espectrometría de Masas , Proteómica , Alineación de Secuencia , Especificidad de la Especie
17.
Malar J ; 11: 439, 2012 Dec 31.
Artículo en Inglés | MEDLINE | ID: mdl-23276246

RESUMEN

BACKGROUND: Malaria transmission occurs during the blood feeding of infected anopheline mosquitoes concomitant with a saliva injection into the vertebrate host. In sub-Saharan Africa, most malaria transmission is due to Anopheles funestus s.s and to Anopheles gambiae s.l. (mainly Anopheles gambiae s.s. and Anopheles arabiensis). Several studies have demonstrated that the immune response against salivary antigens could be used to evaluate individual exposure to mosquito bites. The aim of this study was to assess the use of secreted salivary proteins as specific biomarkers of exposure to An. gambiae and/or An. funestus bites. METHODS: For this purpose, salivary gland proteins 6 (SG6) and 5'nucleotidases (5'nuc) from An. gambiae (gSG6 and g-5'nuc) and An. funestus (fSG6 and f-5'nuc) were selected and produced in recombinant form. The specificity of the IgG response against these salivary proteins was tested using an ELISA with sera from individuals living in three Senegalese villages (NDiop, n = 50; Dielmo, n = 38; and Diama, n = 46) that had been exposed to distinct densities and proportions of the Anopheles species. Individuals who had not been exposed to these tropical mosquitoes were used as controls (Marseille, n = 45). RESULTS: The IgG responses against SG6 recombinant proteins from these two Anopheles species and against g-5'nucleotidase from An. gambiae, were significantly higher in Senegalese individuals compared with controls who were not exposed to specific Anopheles species. Conversely, an association was observed between the level of An. funestus exposure and the serological immune response levels against the f-5'nucleotidase protein. CONCLUSION: This study revealed an Anopheles salivary antigenic protein that could be considered to be a promising antigenic marker to distinguish malaria vector exposure at the species level. The epidemiological interest of such species-specific antigenic markers is discussed.


Asunto(s)
Anopheles/inmunología , Antígenos/inmunología , Mordeduras y Picaduras de Insectos/inmunología , Mordeduras y Picaduras de Insectos/parasitología , Proteínas de Insectos/inmunología , Malaria/inmunología , Malaria/transmisión , Proteínas y Péptidos Salivales/inmunología , 5'-Nucleotidasa/genética , 5'-Nucleotidasa/inmunología , Adulto , Secuencia de Aminoácidos , Animales , Anopheles/genética , Anopheles/parasitología , Antígenos/genética , Biomarcadores , Estudios de Casos y Controles , Reacciones Cruzadas , Femenino , Interacciones Huésped-Parásitos/inmunología , Humanos , Inmunoglobulina G/sangre , Proteínas de Insectos/genética , Masculino , Persona de Mediana Edad , Datos de Secuencia Molecular , Proteínas Recombinantes/genética , Proteínas Recombinantes/inmunología , Proteínas y Péptidos Salivales/genética , Homología de Secuencia de Aminoácido , Especificidad de la Especie
18.
Parasitol Res ; 110(2): 545-56, 2012 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-21744020

RESUMEN

Over the past decade, advances in proteomic and mass spectrometry techniques and the sequencing of the Plasmodium falciparum genome have led to an increasing number of studies regarding the parasite proteome. However, these studies have focused principally on parasite protein expression, neglecting parasite-induced variations in the host proteome. Here, we investigated P. falciparum-induced modifications of the infected red blood cell (iRBC) membrane proteome, taking into account both host and parasite proteome alterations. Furthermore, we also determined if some protein changes were associated with genotypically distinct P. falciparum strains. Comparison of host membrane proteomes between iRBCs and uninfected red blood cells using fluorescence-based proteomic approaches, such as 2D difference gel electrophoresis revealed that more than 100 protein spots were highly up-represented (fold change increase greater than five) following P. falciparum infection for both strains (i.e. RP8 and Institut Pasteur Pregnancy Associated Malaria). The majority of spots identified by mass spectrometry corresponded to Homo sapiens proteins. However, infection-induced changes in host proteins did not appear to affect molecules located at the outer surface of the plasma membrane. The under-representation of parasite proteins could not be attributed to deficient parasite protein expression. Thus, this study describes for the first time that considerable host protein modifications were detected following P. falciparum infection at the erythrocyte membrane level. Further analysis of infection-induced host protein modifications will improve our knowledge of malaria pathogenesis.


Asunto(s)
Membrana Eritrocítica/química , Eritrocitos/química , Eritrocitos/parasitología , Interacciones Huésped-Patógeno , Proteínas de la Membrana/análisis , Plasmodium falciparum/patogenicidad , Electroforesis en Gel Bidimensional , Humanos , Espectrometría de Masas , Proteoma/análisis
19.
Nat Commun ; 12(1): 916, 2021 02 10.
Artículo en Inglés | MEDLINE | ID: mdl-33568638

RESUMEN

The global emergence of Zika virus (ZIKV) revealed the unprecedented ability for a mosquito-borne virus to cause congenital birth defects. A puzzling aspect of ZIKV emergence is that all human outbreaks and birth defects to date have been exclusively associated with the Asian ZIKV lineage, despite a growing body of laboratory evidence pointing towards higher transmissibility and pathogenicity of the African ZIKV lineage. Whether this apparent paradox reflects the use of relatively old African ZIKV strains in most laboratory studies is unclear. Here, we experimentally compare seven low-passage ZIKV strains representing the recently circulating viral genetic diversity. We find that recent African ZIKV strains display higher transmissibility in mosquitoes and higher lethality in both adult and fetal mice than their Asian counterparts. We emphasize the high epidemic potential of African ZIKV strains and suggest that they could more easily go unnoticed by public health surveillance systems than Asian strains due to their propensity to cause fetal loss rather than birth defects.


Asunto(s)
Infección por el Virus Zika/mortalidad , Infección por el Virus Zika/virología , Virus Zika/fisiología , Virus Zika/patogenicidad , Aedes/fisiología , Aedes/virología , África , Animales , Asia , Femenino , Humanos , Masculino , Ratones , Filogenia , Virulencia , Virus Zika/clasificación , Virus Zika/genética , Infección por el Virus Zika/transmisión
20.
FASEB J ; 23(10): 3449-58, 2009 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-19535685

RESUMEN

Cerebral malaria (CM) is characterized by accumulation of circulating cells within brain microvessels, among which platelets play an important role. In vitro, platelets modulate the cytoadherence of Plasmodium falciparum-parasitized red blood cells (PRBCs) to brain endothelial cells. Here we show for the first time that platelet microparticles (PMPs) are able to bind to PRBCs, thereby transferring platelet antigens to the PRBC surface. This binding is largely specific to PRBCs, because PMPs show little adherence to normal red blood cells. PMP adherence is also dependent on the P. falciparum erythrocyte membrane protein 1 variant expressed by PRBCs. PMP binding to PRBCs decreases after neutralization of PRBC surface proteins by trypsin or after treatment of PMPs with a mAb to platelet-endothelial cell adhesion molecule-1 (CD31) and glycoprotein IV (CD36). Furthermore, PMP uptake is a dynamic process that can be achieved by human brain endothelial cells (HBECs), inducing changes in the endothelial phenotype. Lastly, PMPs dramatically increase PRBC cytoadherence to HBECs. In conclusion, our study identifies several mechanisms by which PMPs may participate in CM pathogenesis while interacting with both PRBCs and HBECs. PMPs thereby provide a novel target for antagonizing interactions between vascular cells that promote microvascular sludging and blood brain barrier alteration during CM.


Asunto(s)
Plaquetas/parasitología , Encéfalo/parasitología , Eritrocitos/parasitología , Malaria Cerebral/sangre , Malaria Falciparum/sangre , Plasmodium falciparum , Animales , Plaquetas/metabolismo , Plaquetas/fisiología , Encéfalo/irrigación sanguínea , Antígenos CD36/metabolismo , Adhesión Celular , Endotelio/parasitología , Eritrocitos/fisiología , Humanos , Molécula-1 de Adhesión Celular Endotelial de Plaqueta/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA