Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 38
Filtrar
Más filtros










Intervalo de año de publicación
1.
Sci Rep ; 13(1): 20472, 2023 11 22.
Artículo en Inglés | MEDLINE | ID: mdl-37993652

RESUMEN

Malaria is a severe public health problem in several developing tropical and subtropical countries. Anopheles aquasalis is the primary coastal malaria vector in Central and South America and the Caribbean Islands, and it has the peculiar feature of living in water with large changes in salinity. Recent research has recognised An. aquasalis as an important model for studying the interactions of murine and human Plasmodium parasites. This study presents the complete genome of An. aquasalis and offers insights into its evolution and physiology. The genome is similar in size and gene content to other Neotropical anophelines, with 162 Mb and 12,446 protein-coding genes. There are 1387 single-copy orthologs at the Diptera level (eg. An. gambiae, An. darlingi and Drosophila melanogaster). An. aquasalis diverged from An. darlingi, the primary malaria vector in inland South America, nearly 20 million years ago. Proteins related to ion transport and metabolism belong to the most abundant gene families with 660 genes. We identified gene families relevant to osmosis control (e.g., aquaporins, vacuolar-ATPases, Na+/K+-ATPases, and carbonic anhydrases). Evolutionary analysis suggests that all osmotic regulation genes are under strong purifying selection. We also observed low copy number variation in insecticide resistance and immunity-related genes for all known classical pathways. The data provided by this study offers candidate genes for further studies of parasite-vector interactions and for studies on how anophelines of brackish water deal with the high fluctuation in water salinity. We also established data and insights supporting An. aquasalis as an emerging Neotropical malaria vector model for genetic and molecular studies.


Asunto(s)
Anopheles , Malaria , Humanos , Animales , Ratones , Malaria/parasitología , Anopheles/genética , Anopheles/parasitología , Variaciones en el Número de Copia de ADN/genética , Drosophila melanogaster , Mosquitos Vectores/genética , Agua , Adenosina Trifosfatasas/genética
2.
Malar J ; 22(1): 337, 2023 Nov 07.
Artículo en Inglés | MEDLINE | ID: mdl-37936198

RESUMEN

BACKGROUND: Reducing mosquito abundance or interfering with its ability to support the parasite cycle can help to interrupt malaria in areas of significant risk of malaria transmission. Fluralaner is a safe and effective drug for veterinary use indicated for the treatment against fleas and ticks which acts as an antagonist of chloride ion channels mediated by γ-aminobutyric acid (GABA), preventing the entry of these ions into the postsynaptic neuron, leading to hyperexcitability of the postsynaptic neuron of the central nervous system of arthropods. Fluralaner demonstrated insecticidal activity against different insect species. METHODS: The study aimed to evaluate the effects of fluralaner on the biology, survival, and reproductive fitness of Anopheles aquasalis. The following lethal concentrations (LC) were determined for An. aquasalis: LC5 = 0.511 µM; LC25 = 1.625 µM; LC50 = 3.237 µM. RESULTS: A significant decrease (P < 0.001) was evident in the number of eggs, larvae, and pupae in the group exposed to a sublethal dose of fluralaner when compared to a control group (without the drug). Using blood from dogs after administration of fluralaner, it was observed that the drug causes 100% mortality in An. aquasalis in less than 24 h after feeding; this effect remains even after 90 days in all samples. DISCUSSION: Fluralaner showed the same result for up to 60 days, and after that, there was a slight reduction in its effect, evidenced by a decrease in the percentage of dead females; however, still significant when compared to the control group. CONCLUSION: Fluralaner affects the biology and reduction of survival in An. aquasalis in a lasting and prolonged period, and its fecundity with lower dosages, is a strong candidate for controlling disease vectors.


Asunto(s)
Anopheles , Insecticidas , Malaria , Femenino , Animales , Perros , Anopheles/fisiología , Malaria/prevención & control , Aptitud Genética , Mosquitos Vectores , Insecticidas/farmacología , Biología
3.
Open Biol ; 13(7): 230061, 2023 07.
Artículo en Inglés | MEDLINE | ID: mdl-37433331

RESUMEN

Anophelines are vectors of malaria, the deadliest disease worldwide transmitted by mosquitoes. The availability of genomic data from various Anopheles species allowed evolutionary comparisons of the immune response genes in search of alternative vector control of the malarial parasites. Now, with the Anopheles aquasalis genome, it was possible to obtain more information about the evolution of the immune response genes. Anopheles aquasalis has 278 immune genes in 24 families or groups. Comparatively, the American anophelines possess fewer genes than Anopheles gambiae s. s., the most dangerous African vector. The most remarkable differences were found in the pathogen recognition and modulation families like FREPs, CLIP and C-type lectins. Even so, genes related to the modulation of the expression of effectors in response to pathogens and gene families that control the production of reactive oxygen species were more conserved. Overall, the results show a variable pattern of evolution in the immune response genes in the anopheline species. Environmental factors, such as exposure to different pathogens and differences in the microbiota composition, could shape the expression of this group of genes. The results presented here will contribute to a better knowledge of the Neotropical vector and open opportunities for malaria control in the endemic-affected areas of the New World.


Asunto(s)
Anopheles , Malaria , Animales , Anopheles/genética , Mosquitos Vectores/genética , América del Sur , Indias Occidentales
4.
Cell Tissue Res ; 393(2): 297-320, 2023 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-37272999

RESUMEN

The mosquito larval midgut is responsible for acquiring and storing most of the nutrients that will sustain the events of metamorphosis and the insect's adult life. Despite its importance, the basic biology of this larval organ is poorly understood. To help fill this gap, we carried out a comparative morphophysiological investigation of three larval midgut regions (gastric caeca, anterior midgut, and posterior midgut) of phylogenetically distant mosquitoes: Anopheles gambiae (Anopheles albimanus was occasionally used as an alternate), Aedes aegypti, and Toxorhynchites theobaldi. Larvae of Toxorhynchites mosquitoes are predacious, in contrast to the other two species, that are detritivorous. In this work, we show that the larval gut of the three species shares basic histological characteristics, but differ in other aspects. The lipid and carbohydrate metabolism of the An. gambiae larval midgut is different compared with that of Ae. aegypti and Tx. theobaldi. The gastric caecum is the most variable region, with differences probably related to the chemical composition of the diet. The peritrophic matrix is morphologically similar in the three species, and processes involved in the post-embryonic development of the organ, such as cell differentiation and proliferation, were also similar. FMRF-positive enteroendocrine cells are grouped in the posterior midgut of Tx. theobaldi, but individualized in An. gambiae and Ae. aegypti. We hypothesize that Tx. theobaldi larval predation is an ancestral condition in mosquito evolution.


Asunto(s)
Aedes , Anopheles , Animales , Anopheles/fisiología , Larva/metabolismo , Sistema Digestivo , Células Enteroendocrinas
5.
Viruses ; 15(3)2023 03 17.
Artículo en Inglés | MEDLINE | ID: mdl-36992479

RESUMEN

Zika virus (ZIKV) is transmitted to humans by the infectious bite of mosquitoes such as Aedes aegypti. In a city, the population control of mosquitoes is carried out according to alerts generated by different districts via the analysis of the mosquito index. However, we do not know whether, besides mosquito abundance, the susceptibility of mosquitoes could also diverge among districts and thus impact the dissemination and transmission of arboviruses. After a viremic blood meal, the virus must infect the midgut, disseminate to tissues, and reach the salivary gland to be transmitted to a vertebrate host. This study evaluated the patterns of ZIKV infection in the Ae. aegypti field populations of a city. The disseminated infection rate, viral transmission rate, and transmission efficiency were measured using quantitative PCR at 14 days post-infection. The results showed that all Ae. aegypti populations had individuals susceptible to ZIKV infection and able to transmit the virus. The infection parameters showed that the geographical area of origin of the Ae. aegypti influences its vector competence for ZIKV transmission.


Asunto(s)
Aedes , Infección por el Virus Zika , Virus Zika , Animales , Humanos , Virus Zika/genética , Brasil/epidemiología , Saliva , Mosquitos Vectores
6.
Rev Soc Bras Med Trop ; 55: e0427, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36000618

RESUMEN

BACKGROUND: Arthropod-borne viruses have recently emerged and are pathogens of various human diseases, including dengue, zika, and chikungunya viruses. METHODS: We collectedAedes aegyptilarvae (N = 20) from Brumado, Bahia, Brazil, and treated and individually preserved the specimens. We analyzed the samples for dengue, zika, and chikungunya viruses using molecular biology methods. RESULTS: We found that 25% (N = 5) and 15% (N = 3) were positive exclusively for dengue and chikungunya viruses, respectively; 15% (N = 3) were coinfected with both. CONCLUSIONS: This is the first report of dengue and chikungunya virus coinfection in A. aegypti larvae.


Asunto(s)
Aedes , Fiebre Chikungunya , Virus Chikungunya , Virus del Dengue , Dengue , Infección por el Virus Zika , Virus Zika , Animales , Brasil , Virus Chikungunya/genética , Virus del Dengue/genética , Humanos , Mosquitos Vectores
7.
Parasit Vectors ; 15(1): 57, 2022 Feb 17.
Artículo en Inglés | MEDLINE | ID: mdl-35177110

RESUMEN

BACKGROUND: Emerging and re-emerging vector-borne diseases (VBDs) pose a recurring threat to tropical countries, mainly due to the abundance and distribution of the Aedes aegypti mosquito, which is a vector of the Zika, dengue, chikungunya, and yellow fever arboviruses. METHODS: Female 3-5 day-old Ae. aegypti were distributed into two experimental groups: group I-survey of cultivable bacteria; sucrose group: fed only on sucrose, i.e., non-blood-fed (UF); blood-fed group: (i) fed with non-infected blood (BF); (ii) fed with blood infected with the Zika virus (BZIKV); (iii) pretreated with penicillin/streptomycin (pen/strep), and fed with non-infected blood (TBF); (iv) pretreated with pen/strep and fed blood infected with ZIKV, i.e., gravid with developed ovaries, (TGZIKV); group II-experimental co-infections: bacteria genera isolated from the group fed on sucrose, i.e., non-blood-fed (UF). RESULTS: Using the cultivable method and the same mosquito colony and ZIKV strain described by in a previous work, our results reveled 11 isolates (Acinetobacter, Aeromonas, Cedecea, Cellulosimicrobium, Elizabethkingia, Enterobacter, Lysinibacillus, Pantoea, Pseudomonas, Serratia, and Staphylococcus). Enterobacter was present in all evaluated groups (i.e., UF, BF, BZIKV, TBF, and TGZIKV), whereas Elizabethkingia was present in the UF, BZIKV, and TBF groups. Pseudomonas was present in the BZIKV and TBF groups, whereas Staphylococcus was present in the TBF and TGZIKV groups. The only genera of bacteria that were found to be present in only one group were Aeromonas, Lysinibacillus, and Serratia (UF); Cedacea, Pantoea and Acinetobacter (BF); and Cellulosimicrobium (BZIKV). The mosquitoes co-infected with ZIKV plus the isolates group fed on sucrose (UF) showed interference in the outcome of infection. CONCLUSIONS: We demonstrate that the distinct feeding aspects assessed herein influence the composition of bacterial diversity. In the co-infection, among ZIKV, Ae. aegypti and the bacterial isolates, the ZIKV/Lysinibacillus-Ae. aegypti had the lowest number of viral copies in the head-SG, which means that it negatively affects vector competence. However, when the saliva was analyzed after forced feeding, no virus was detected in the mosquito groups ZIKV/Lysinibacillus-Lu. longipalpis and Ae. aegypti; the combination of ZIKV/Serratia may interfere in salivation. This indicates that the combinations do not produce viable viruses and may have great potential as a method of biological control.


Asunto(s)
Aedes , Microbiota , Infección por el Virus Zika , Virus Zika , Animales , Femenino , Mosquitos Vectores
9.
Rev. Soc. Bras. Med. Trop ; 55: e0427, 2022. tab, graf
Artículo en Inglés | LILACS-Express | LILACS | ID: biblio-1394692

RESUMEN

ABSTRACT Background: Arthropod-borne viruses have recently emerged and are pathogens of various human diseases, including dengue, zika, and chikungunya viruses. Methods: We collectedAedes aegyptilarvae (N = 20) from Brumado, Bahia, Brazil, and treated and individually preserved the specimens. We analyzed the samples for dengue, zika, and chikungunya viruses using molecular biology methods. Results: We found that 25% (N = 5) and 15% (N = 3) were positive exclusively for dengue and chikungunya viruses, respectively; 15% (N = 3) were coinfected with both. Conclusions: This is the first report of dengue and chikungunya virus coinfection in A. aegypti larvae.

10.
PLoS Negl Trop Dis ; 15(11): e0009839, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34727099

RESUMEN

Dengue virus (DENV) and Zika virus (ZIKV) belong to the same viral family, the Flaviviridae. They cause recurring threats to the public health systems of tropical countries such as Brazil. The primary Brazilian vector of both viruses is the mosquito Aedes aegypti. After the mosquito ingests a blood meal from an infected person, the viruses infect and replicate in the midgut, disseminate to secondary tissues and reach the salivary gland (SG), where they are ready to be transmitted to a vertebrate host. It is thought that the intrinsic discrepancies among mosquitoes could affect their ability to deal with viral infections. This study confirms that the DENV and ZIKV infection patterns of nine Ae. aegypti field populations found in geographically separate health districts of an endemic Brazilian city vary. We analyzed the infection rate, disseminated infection, vector competence, and viral load through quantitative PCR. Mosquitoes were challenged using the membrane-feeding assay technique and were tested seven and fourteen days post-infection (early and late infection phases, respectively). The infection responses varied among the Ae. aegypti populations for both flaviviruses in the two infection phases. There was no similarity between DENV and ZIKV vector competencies or viral loads. According to the results of our study, the risk of viral transmission overtime after infection either increases or remains unaltered in ZIKV infected vectors. However, the risk may increase, decrease, or remain unaltered in DENV-infected vectors depending on the mosquito population. For both flaviviruses, the viral load persisted in the body even until the late infection phase. In contrast to DENV, the ZIKV accumulated in the SG over time in all the mosquito populations. These findings are novel and may help direct the development of control strategies to fight dengue and Zika outbreaks in endemic regions, and provide a warning about the importance of understanding mosquito responses to arboviral infections.


Asunto(s)
Aedes/virología , Mosquitos Vectores/virología , Virus Zika/aislamiento & purificación , Aedes/fisiología , Animales , Brasil/epidemiología , Enfermedades Endémicas , Femenino , Humanos , Masculino , Mosquitos Vectores/fisiología , Glándulas Salivales/virología , Carga Viral , Virus Zika/genética , Virus Zika/fisiología , Infección por el Virus Zika/epidemiología , Infección por el Virus Zika/transmisión , Infección por el Virus Zika/virología
11.
Acta Trop ; 224: 106129, 2021 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-34509458

RESUMEN

Anopheles aquasalis is an important malaria vector in coastal regions of South America and islands of the Caribbean. In its original description, the species was divided into two varieties, based on the scaling patterns of their hind-tarsomere 2. Specimens from our 25-year established colony, used for Plasmodium experimental infections, still exhibit both scaling tarsomere patterns. This study examined the DNA sequence of the nuclear Internal Transcribed Spacer 2 (ITS2) and susceptibility to Plasmodium, looking for differences among the phenotypes 30BS and 50BS. One hundred mosquitoes, 25 males and 25 females of each sex, and phenotype were analyzed. Twenty-seven novel haplotypes were identified. Three were found in both phenotypes (30BS and 50BS) regardless of gender. Among the other 27 genotypes, we observed a male-oriented bias in both phenotypic categories. Evaluation of Plasmodium yoelii N67 infections, based on oocyst counts, showed a higher susceptibility of 30BS compared with 50BS. Future studies need to be conducted to evaluate if these genotype assortments among the phenotypic groups reflect differences in fitness, mating, and their susceptibility to infection by Plasmodium parasites.


Asunto(s)
Anopheles , Malaria , Plasmodium , Animales , Anopheles/genética , Femenino , Humanos , Masculino , Mosquitos Vectores/genética , Fenotipo , Plasmodium/genética
12.
J Med Entomol ; 58(2): 634-645, 2021 03 12.
Artículo en Inglés | MEDLINE | ID: mdl-33710316

RESUMEN

We investigated by scanning electron microscopy the morphology, distribution, and abundance of antennal sensilla of females Phlebotomus duboscqi sand fly, an important vector of zoonotic cutaneous leishmaniasis at Afrotropical region. Thirteen well-differentiated sensilla were identified, among six types of cuticular sensilla. The probable function of these sensillary types is discussed in relation to their external structure and distribution. Five sensillary types were classified as olfactory sensilla, as they have specific morphological characters of sensilla with this function. Number and distribution of sensilla significantly differed between antennal segments. The results of the present work, besides corroborating in the expansion of the morphological and ultrastructural knowledge of P. duboscqi, can foment future electrophysiological studies for the development of volatile semiochemicals, to be used as attractants in traps for monitoring and selective vector control of this sand fly.


Asunto(s)
Phlebotomus/ultraestructura , Sensilos/ultraestructura , Animales , Femenino , Phlebotomus/fisiología , Sensilos/fisiología
13.
J Infect Dis ; 224(1): 101-108, 2021 07 02.
Artículo en Inglés | MEDLINE | ID: mdl-33544850

RESUMEN

BACKGROUND: Aedes aegypti is a highly competent vector in the transmission of arboviruses, such as chikungunya, dengue, Zika, and yellow fever viruses, and causes single and coinfections in the populations of tropical countries. METHODS: The infection rate, viral abundance (VA), vector competence (VC), disseminated infection, and survival rate were recorded after single and multiple infections of the vector with 15 combinations of chikungunya, dengue, Zika, and yellow fever arboviruses. RESULTS: Infection rates were 100% in all single and multiple infection experiments, except in 1 triple coinfection that presented a rate of 50%. The VC and disseminated infection rate varied from 100% (in single and quadruple infections) to 40% (in dual and triple infections). The dual and triple coinfections altered the VC and/or VA of ≥1 arbovirus. The highest viral VAs were detected for a single infection with chikungunya. The VAs in quadruple infections were similar when compared with each respective single infection. A decrease in survival rates was observed in a few combinations. CONCLUSIONS: A. aegypti was able to host all single and multiple arboviral coinfections. The interference of the chikungunya virus suggests that distinct arbovirus families may have a significant role in complex coinfections.


Asunto(s)
Aedes/virología , Infecciones por Arbovirus/transmisión , Coinfección/transmisión , Mosquitos Vectores/virología , Animales , Arbovirus/aislamiento & purificación , Femenino
14.
Acta Trop ; 215: 105819, 2021 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-33406443

RESUMEN

The outbreaks caused by the Aedes aegypti-transmitted dengue virus (DENV), zakat virus (ZIKV), and chikungunya virus (CHIKV) result in a significant impact to the health systems of tropical countries. Furthermore, the occurrence of patients coinfected by at least two of these arboviruses is an aggravating factor in that scenario. On this basis, surveillance tools such as the Rapid Index Survey for Aedes aegypti (LIRAa) are used to estimate vector infestation in order to improve the prediction of human outbreaks. Ae. aegypti eggs were collected in the city of Vitória da Conquista, in Bahia State, Brazil, and subsequently hatched into larvae, which were analyzed in pools or individually for the presence of DENV, ZIKV, and CHIKV by molecular biology methods. The detection data for arboviruses were crossed with the LIRAa obtained in each region of the study city. Thirty larvae pools were analyzed, and fourteen (46.6%) of them were detected positive for DENV, ZIKV, and/or CHIKV. Among the individually analyzed larvae (n = 30), nine (30%) were positive for any of these arboviruses, and four (13.3%) were simultaneously coinfected by DENV and ZIKV. Furthermore, there was a positive correlation between the detection of circulating arboviruses and LIRAa. The simultaneous Ae. aegypti larvae infection by two different arboviruses is an unprecedented finding. This result suggests the occurrence of a vertical arboviruses co-transmission from the female mosquito to its offspring in nature. The occurrence of concomitant circulation of DENV, ZIKV, and CHIKV in Ae. aegypti from a single study region is another finding of this article. Finally, LIRAa seems to not only estimate vector infestation but also to predict circulation of arboviruses.


Asunto(s)
Aedes/virología , Virus Chikungunya/aislamiento & purificación , Coinfección/transmisión , Virus del Dengue/aislamiento & purificación , Transmisión Vertical de Enfermedad Infecciosa , Mosquitos Vectores/virología , Virus Zika/aislamiento & purificación , Animales , Femenino , Humanos , Larva/virología
15.
Viruses ; 14(1)2021 12 23.
Artículo en Inglés | MEDLINE | ID: mdl-35062224

RESUMEN

The successful spread and maintenance of the dengue virus (DENV) in mosquito vectors depends on their viral infection susceptibility, and parameters related to vector competence are the most valuable for measuring the risk of viral transmission by mosquitoes. These parameters may vary according to the viral serotype in circulation and in accordance with the geographic origin of the mosquito population that is being assessed. In this study, we investigated the effect of DENV serotypes (1-4) with regards to the infection susceptibility of five Brazilian Ae. aegypti populations from Manaus, the capital of the state of Amazonas, Brazil. Mosquitoes were challenged by oral infection with the DENV serotypes and then tested for the presence of the arbovirus using quantitative PCR at 14 days post-infection, which is the time point that corresponds to the extrinsic incubation period of Ae. aegypti when reared at 28 °C. Thus, we were able to determine the infection patterns for DENV-1, -2, -3 and -4 in the mosquito populations. The mosquitoes had both interpopulation and inter-serotype variation in their viral susceptibilities. All DENV serotypes showed a similar tendency to accumulate in the body in a greater amount than in the head/salivary gland (head/SG), which does not occur with other flaviviruses. For DENV-1, DENV-3, and DENV-4, the body viral load varied among populations, but the head/SG viral loads were similar. Differently for DENV-2, both body and head/SG viral loads varied among populations. As the lack of phenotypic homogeneity represents one of the most important reasons for the long-term fight against dengue incidence, we expect that this study will help us to understand the dynamics of the infection patterns that are triggered by the distinct serotypes of DENV in mosquitoes.


Asunto(s)
Aedes/virología , Virus del Dengue/fisiología , Mosquitos Vectores/virología , Animales , Brasil , Virus del Dengue/genética , Femenino , Serogrupo , Carga Viral
16.
PLoS Negl Trop Dis ; 14(10): e0008666, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-33052941

RESUMEN

The microbial consortium associated with sandflies has gained relevance, with its composition shifting throughout distinct developmental stages, being strongly influenced by the surroundings and food sources. The bacterial components of the microbiota can interfere with Leishmania development inside the sandfly vector. Microbiota diversity and host-microbiota-pathogen interactions regarding New World sandfly species have yet to be thoroughly studied, particularly in Lutzomyia longipalpis, the primary vector of visceral leishmaniasis in Brazil.The native microbiota of different developmental stages and physiological conditions of Lu. longipalpis (Lapinha Cave), was described by culturing and 16s rRNA gene sequencing. The 16s rRNA sequencing of culture-dependent revealed 13 distinct bacterial genera (Bacillus, Enterococcus, Erwinia, Enterobacter, Escherichia, Klebsiella, Lysinibacillus, Pseudocitrobacter, Providencia, Pseudomonas, Serratia, Staphylococcus and Solibacillus). The in vitro and in vivo effects of each one of the 13 native bacteria from the Lu. longipalpis were analyzed by co-cultivation with promastigotes of L.i. chagasi, L. major, L. amazonensis, and L. braziliensis. After 24 h of co-cultivation, a growth reduction observed in all parasite species. When the parasites were co-cultivated with Lysinibacillus, all parasites of L. infantum chagasi and L. amazonensis died within 24 hours. In the in vivo co-infection of L.chagasi, L. major and L. amazonensis with the genera Lysinibacillus, Pseudocitrobacter and Serratia it was possible to observe a significant difference between the groups co-infected with the bacterial genera and the control group.These findings suggest that symbiont bacteria (Lysinibacillus, Serratia, and Pseudocitrobacter) are potential candidates for paratransgenic or biological control. Further studies are needed to identify the nature of the effector molecules involved in reducing the vector competence for Leishmania.


Asunto(s)
Bacterias/clasificación , Leishmania/fisiología , Psychodidae/microbiología , Psychodidae/parasitología , Animales , Microbiota , ARN Bacteriano/genética , ARN Ribosómico 16S/genética
17.
J Med Entomol ; 57(5): 1447-1458, 2020 09 07.
Artículo en Inglés | MEDLINE | ID: mdl-32424423

RESUMEN

The ultrastructure of the mouthparts of Haematobia irritans (L.) was investigated by scanning electron microscopy. The morphological characteristics of the maxillary palps, labium (prementum and postmentum), labrum, hypopharynx, haustellum, and labellar lobes are described, as well as of the sensilla evidenced on all the surface of the mouthparts, and the set of different positions assumed by the mouth apparatus of this fly. Based on their morphology, 12 well-differentiated sensilla were identified, among three types of cuticular sensilla: trichoidea, coeloconica, and campaniformia. A slight sexual dimorphism in the sensilla patterns found in the mouthparts of H. irritans was evidenced. These observations are discussed with reference to the current literature on the functional morphology of sense organs of Insecta. These results could facilitate the recognition of the chemosensory sensilla by electrophysiological techniques, and foment future taxonomic and phylogenetic studies to better elucidate the evolution of Diptera, Muscomorpha.


Asunto(s)
Muscidae/ultraestructura , Animales , Brasil , Femenino , Masculino , Microscopía Electrónica de Rastreo , Boca/ultraestructura , Sensilos/ultraestructura , Factores Sexuales
18.
J Med Entomol ; 56(6): 1739-1744, 2019 10 28.
Artículo en Inglés | MEDLINE | ID: mdl-31278737

RESUMEN

Zika virus (ZIKV) has emerged as a globally important arbovirus and has been reported from all states of Brazil. The virus is primarily transmitted to humans through the bite of an infective Aedes aegypti (Linnaeus, 1762) or Aedes albopictus (Skuse, 1895). However, it is important to know if ZIKV transmission also occurs from Ae. aegypti through infected eggs to her offspring. Therefore, a ZIKV and dengue virus (DENV) free colony was established from eggs collected in Manaus and maintained until the third-fourth generation in order to conduct ZIKV vertical transmission (VT) experiments which used an infectious bloodmeal as the route of virus exposure. The eggs from ZIKV-infected females were allowed to hatch. The resulting F1 progeny (larvae, pupae, and adults) were quantitative polymerase chain reaction (qPCR) assayed for ZIKV. The viability of ZIKV vertically transmitted to F1 progeny was evaluated by cultivation in C6/36 cells. The effects of ZIKV on immature development of Ae. aegypti was assessed and compared with noninfected mosquitoes. AmazonianAe. aegypti were highly susceptible to ZIKV infection (96.7%), and viable virus passed to their progeny via VT. Moreover, eggs from the ZIKV-infected mosquitoes had a significantly lower hatch rate and the slowest hatching. In addition, the larval development period was slower when compared to noninfected, control mosquitoes. This is the first study to illustrate VT initiated by oral infection of the parental population by using mosquitoes, which originated from the field and a ZIKV strain that is naturally circulating in-country. Additionally, this study suggests that ZIKV present in the Ae. aegypti can modify the mosquito life cycle. The data reported here suggest that VT of ZIKV to progeny from naturally infected females may have a critical epidemiological role in the dissemination and maintenance of the virus circulating in the vector.


Asunto(s)
Aedes/crecimiento & desarrollo , Aedes/virología , Mosquitos Vectores/crecimiento & desarrollo , Mosquitos Vectores/virología , Virus Zika/fisiología , Animales , Brasil , Femenino , Larva/crecimiento & desarrollo , Larva/virología , Óvulo/crecimiento & desarrollo , Óvulo/virología
19.
J Infect Dis ; 218(4): 563-571, 2018 07 13.
Artículo en Inglés | MEDLINE | ID: mdl-29659904

RESUMEN

Background: Several tropical cities are permissive to Aedes aegypti and dengue virus (DENV) endemicity and have allowed for invasion and circulation of Zika virus (ZIKV) in the same areas. People living in arbovirus-endemic regions have been simultaneously infected with ≥2 arboviruses. Methods: A. aegypti mosquitoes from Manaus, the capital city of Amazonas State in Brazil, were coinfected with circulating strains of DENV and ZIKV. The coinfected vectors were allowed to bite BALB/c mice. Results: A. aegypti from Manaus is highly permissive to monoinfection and coinfection with DENV and ZIKV and is capable of cotransmitting both pathogens by bite. Coinfection strongly influences vector competence, favoring transmission of ZIKV to the vertebrate host. Conclusions: This finding suggests that A. aegypti is an efficient vector of ZIKV and that ZIKV would be preferentially transmitted by coinfected A. aegypti. Coinfection in the vector population should be considered a new critical epidemiological factor and may represent a major public health challenge.


Asunto(s)
Aedes/virología , Coinfección/transmisión , Dengue/transmisión , Transmisión de Enfermedad Infecciosa , Mosquitos Vectores/virología , Infección por el Virus Zika/transmisión , Aedes/crecimiento & desarrollo , Animales , Brasil , Ciudades , Virus del Dengue/crecimiento & desarrollo , Modelos Animales de Enfermedad , Femenino , Ratones Endogámicos BALB C , Mosquitos Vectores/crecimiento & desarrollo , Virus Zika/crecimiento & desarrollo
20.
Parasit Vectors ; 11(1): 148, 2018 03 06.
Artículo en Inglés | MEDLINE | ID: mdl-29510729

RESUMEN

BACKGROUND: Plasmodium vivax is predominant in the Amazon region, and enhanced knowledge of its development inside a natural vector, Anopheles aquasalis, is critical for future strategies aimed at blocking parasite development. The peritrophic matrix (PM), a chitinous layer produced by the mosquito midgut in response to blood ingestion, is a protective barrier against pathogens. Plasmodium can only complete its life-cycle, and consequently be transmitted to a new host, after successfully passing this barrier. Interestingly, fully engorged mosquitoes that had a complete blood meal form a thicker, well-developed PM than ones that feed in small amounts. The amount of red blood cells (RBC) in the blood meal directly influences the production of digestive enzymes and can protect parasites from being killed during the meal digestion. A specific study interrupting the development of the PM associated with the proteolytic activity inhibition, and distinct RBC concentrations, during the P. vivax infection of the New World malaria vector An. aquasalis is expected to clarify whether these factors affect the parasite development. RESULTS: Absence of PM in the vector caused a significant reduction in P. vivax infection. However, the association of chitinase with trypsin inhibitor restored infection rates to those of mosquitoes with a structured PM. Also, only the ingestion of trypsin inhibitor by non-chitinase treated mosquitoes increased the infection intensity. Moreover, the RBC concentration in the infected P. vivax blood meal directly influenced the infection rate and its intensity. A straight correlation was observed between RBC concentrations and infection intensity. CONCLUSIONS: This study established that there is a balance between the PM role, RBC concentration and digestive enzyme activity influencing the establishment and development of P. vivax infection inside An. aquasalis. Our results indicate that the absence of PM in the midgut facilitates digestive enzyme dispersion throughout the blood meal, causing direct damage to P. vivax. On the other hand, high RBC concentrations support a better and thick, well-developed PM and protect P. vivax from being killed. Further studies of this complex system may provide insights into other details of the malaria vector response to P. vivax infection.


Asunto(s)
Anopheles/parasitología , Sangre , Sistema Digestivo/enzimología , Eritrocitos/metabolismo , Plasmodium vivax/fisiología , Animales , Sistema Digestivo/anatomía & histología , Fenómenos Fisiológicos del Sistema Digestivo , Hematócrito , Interacciones Huésped-Parásitos , Estadios del Ciclo de Vida , Malaria/transmisión , Malaria Vivax , Comidas , Mosquitos Vectores/parasitología , Tripsina/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...