Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 2.725
Filtrar
1.
PLoS One ; 19(5): e0303137, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38722911

RESUMO

The Asian tiger mosquito, Aedes albopictus, is a significant public health concern owing to its expanding habitat and vector competence. Disease outbreaks attributed to this species have been reported in areas under its invasion, and its northward expansion in Japan has caused concern because of the potential for dengue virus infection in newly populated areas. Accurate prediction of Ae. albopictus distribution is crucial to prevent the spread of the disease. However, limited studies have focused on the prediction of Ae. albopictus distribution in Japan. Herein, we used the random forest model, a machine learning approach, to predict the current and potential future habitat ranges of Ae. albopictus in Japan. The model revealed that these mosquitoes prefer urban areas over forests in Japan on the current map. Under predictions for the future, the species will expand its range to the surrounding areas and eventually reach many areas of northeastern Kanto, Tohoku District, and Hokkaido, with a few variations in different scenarios. However, the affected human population is predicted to decrease owing to the declining birth rate. Anthropogenic and climatic factors contribute to range expansion, and urban size and population have profound impacts. This prediction map can guide responses to the introduction of this species in new areas, advance the spatial knowledge of diseases vectored by it, and mitigate the possible disease burden. To our knowledge, this is the first distribution-modelling prediction for Ae. albopictus with a focus on Japan.


Assuntos
Aedes , Mosquitos Vetores , Animais , Aedes/virologia , Aedes/fisiologia , Japão , Mosquitos Vetores/virologia , Ecossistema , Humanos , Distribuição Animal , Dengue/transmissão , Dengue/epidemiologia , Aprendizado de Máquina , Modelos Biológicos
2.
Parasit Vectors ; 17(1): 204, 2024 May 07.
Artigo em Inglês | MEDLINE | ID: mdl-38715075

RESUMO

BACKGROUND: Mosquito-borne viruses cause various infectious diseases in humans and animals. Oya virus (OYAV) and Ebinur Lake virus (EBIV), belonging to the genus Orthobunyavirus within the family Peribunyaviridae, are recognized as neglected viruses with the potential to pose threats to animal or public health. The evaluation of vector competence is essential for predicting the arbovirus transmission risk. METHODS: To investigate the range of mosquito vectors for OYAV (strain SZC50) and EBIV (strain Cu20-XJ), the susceptibility of four mosquito species (Culex pipiens pallens, Cx. quinquefasciatus, Aedes albopictus, and Ae. aegypti) was measured through artificial oral infection. Then, mosquito species with a high infection rate (IR) were chosen to further evaluate the dissemination rate (DR), transmission rate (TR), and transmission efficiency. The viral RNA in each mosquito sample was determined by RT-qPCR. RESULTS: The results revealed that for OYAV, Cx. pipiens pallens had the highest IR (up to 40.0%) among the four species, but the DR and TR were 4.8% and 0.0%, respectively. For EBIV, Cx. pipiens pallens and Cx. quinquefasciatus had higher IR compared to Ae. albopictus (1.7%). However, the EBIV RNA and infectious virus were detected in Cx. pipiens pallens, with a TR of up to 15.4% and a transmission efficiency of 3.3%. CONCLUSIONS: The findings indicate that Cx. pipiens pallens was susceptible to OYAV but had an extremely low risk of transmitting the virus. Culex pipiens pallens and Cx. quinquefasciatus were susceptible to EBIV, and Cx. pipiens pallens had a higher transmission risk to EBIV than Cx. quinquefasciatus.


Assuntos
Aedes , Culex , Mosquitos Vetores , Orthobunyavirus , Animais , Mosquitos Vetores/virologia , Aedes/virologia , Culex/virologia , Orthobunyavirus/genética , Orthobunyavirus/classificação , Orthobunyavirus/isolamento & purificação , RNA Viral/genética , Infecções por Bunyaviridae/transmissão , Infecções por Bunyaviridae/virologia
3.
Res Sq ; 2024 Apr 19.
Artigo em Inglês | MEDLINE | ID: mdl-38699327

RESUMO

Background: In late 2021, Ghana was hit by a Yellow Fever outbreak that started in two (2) districts in the Savannah region and spread to several other Districts in (3) regions (Oti, Bono and Upper West).Yellow fever is endemic in Ghana. However, there is currently no structured vector control programme for the yellow vector, Aedes mosquitoes in Ghana. Knowledge of Aedes bionomics and insecticide susceptibility status is important to control the vectors. This study therefore sought todetermine Aedes vector bionomics and their insecticide resistance status during a yellow fever outbreak. Methods: The study was performed in two yellow fever outbreak sites (Wenchi, Larabanga) and two non-outbreak sites (Kpalsogu, Pagaza) in Ghana. Immature Aedes mosquitoes were sampled from water-holding containers in and around human habitations. The risk of disease transmission was determined in each site using stegomyia indices. Adult Aedes mosquitoes were sampled using Biogents Sentinel (BG) traps, Human Landing Catch (HLC), and Prokopack (PPK) aspirators. Phenotypic resistance was determined with WHO susceptibility tests using Aedes mosquitoes collected as larvae and reared into adults. Knockdown resistance (kdr) mutations were detected using allele-specific multiplex PCR. Results: Of the 2,664 immature Aedes sampled, more than 60% were found in car tyres. Larabanga, an outbreak site, was classified as a high-risk zone for the Yellow Fever outbreak (BI: 84%, CI: 26.4%). Out of 1,507 adult Aedes mosquitoes collected, Aedes aegypti was the predominant vector species (92%). A significantly high abundance of Aedes mosquitoes was observed during the dry season (61.2%) and outdoors (60.6%) (P < 0.001). Moderate to high resistance to deltamethrin was observed in all sites (33.75% to 70%). Moderate resistance to pirimiphos-methyl (65%) was observed in Kpalsogu. Aedesmosquitoes from Larabanga were susceptible (98%) to permethrin. The F1534C kdr, V1016I kdr and V410 kdr alleles were present in all the sites with frequencies between (0.05-0.92). The outbreak sites had significantly higher allele frequencies of F1534C and V1016I respectively compared to non-outbreak sites (P < 0.001). Conclusion: This study indicates that Aedes mosquitoes in Ghana pose a significant risk to public health, and there is a need for continuous surveillance to inform effective vector control strategies.

4.
PLoS One ; 19(5): e0302496, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38709760

RESUMO

Adult mosquitoes require regular sugar meals, including nectar, to survive in natural habitats. Both males and females locate potential sugar sources using sensory proteins called odorant receptors (ORs) activated by plant volatiles to orient toward flowers or honeydew. The yellow fever mosquito, Aedes aegypti (Linnaeus, 1762), possesses a large gene family of ORs, many of which are likely to detect floral odors. In this study, we have uncovered ligand-receptor pairings for a suite of Aedes aegypti ORs using a panel of environmentally relevant, plant-derived volatile chemicals and a heterologous expression system. Our results support the hypothesis that these odors mediate sensory responses to floral odors in the mosquito's central nervous system, thereby influencing appetitive or aversive behaviors. Further, these ORs are well conserved in other mosquitoes, suggesting they function similarly in diverse species. This information can be used to assess mosquito foraging behavior and develop novel control strategies, especially those that incorporate mosquito bait-and-kill technologies.


Assuntos
Aedes , Flores , Receptores Odorantes , Compostos Orgânicos Voláteis , Animais , Aedes/fisiologia , Aedes/metabolismo , Receptores Odorantes/metabolismo , Receptores Odorantes/genética , Compostos Orgânicos Voláteis/metabolismo , Compostos Orgânicos Voláteis/análise , Feminino , Masculino , Febre Amarela/transmissão , Odorantes/análise , Plantas/metabolismo , Plantas/química
5.
Sci Rep ; 14(1): 10003, 2024 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-38693192

RESUMO

Zika, a viral disease transmitted to humans by Aedes mosquitoes, emerged in the Americas in 2015, causing large-scale epidemics. Colombia alone reported over 72,000 Zika cases between 2015 and 2016. Using national surveillance data from 1121 municipalities over 70 weeks, we identified sociodemographic and environmental factors associated with Zika's emergence, re-emergence, persistence, and transmission intensity in Colombia. We fitted a zero-state Markov-switching model under the Bayesian framework, assuming Zika switched between periods of presence and absence according to spatially and temporally varying probabilities of emergence/re-emergence (from absence to presence) and persistence (from presence to presence). These probabilities were assumed to follow a series of mixed multiple logistic regressions. When Zika was present, assuming that the cases follow a negative binomial distribution, we estimated the transmission intensity rate. Our results indicate that Zika emerged/re-emerged sooner and that transmission was intensified in municipalities that were more densely populated, at lower altitudes and/or with less vegetation cover. Warmer temperatures and less weekly-accumulated rain were also associated with Zika emergence. Zika cases persisted for longer in more densely populated areas with more cases reported in the previous week. Overall, population density, elevation, and temperature were identified as the main contributors to the first Zika epidemic in Colombia. We also estimated the probability of Zika presence by municipality and week, and the results suggest that the disease circulated undetected by the surveillance system on many occasions. Our results offer insights into priority areas for public health interventions against emerging and re-emerging Aedes-borne diseases.


Assuntos
Aedes , Cadeias de Markov , Infecção por Zika virus , Zika virus , Infecção por Zika virus/transmissão , Infecção por Zika virus/epidemiologia , Colômbia/epidemiologia , Humanos , Animais , Aedes/virologia , Teorema de Bayes , Mosquitos Vetores/virologia , Surtos de Doenças
6.
Parasit Vectors ; 17(1): 200, 2024 May 04.
Artigo em Inglês | MEDLINE | ID: mdl-38704595

RESUMO

BACKGROUND: Mayaro virus (MAYV) is an emerging alphavirus, primarily transmitted by the mosquito Haemagogus janthinomys in Central and South America. However, recent studies have shown that Aedes aegypti, Aedes albopictus and various Anopheles mosquitoes can also transmit the virus under laboratory conditions. MAYV causes sporadic outbreaks across the South American region, particularly in areas near forests. Recently, cases have been reported in European and North American travelers returning from endemic areas, raising concerns about potential introductions into new regions. This study aims to assess the vector competence of three potential vectors for MAYV present in Europe. METHODS: Aedes albopictus from Italy, Anopheles atroparvus from Spain and Culex pipiens biotype molestus from Belgium were exposed to MAYV and maintained under controlled environmental conditions. Saliva was collected through a salivation assay at 7 and 14 days post-infection (dpi), followed by vector dissection. Viral titers were determined using focus forming assays, and infection rates, dissemination rates, and transmission efficiency were calculated. RESULTS: Results indicate that Ae. albopictus and An. atroparvus from Italy and Spain, respectively, are competent vectors for MAYV, with transmission possible starting from 7 dpi under laboratory conditions. In contrast, Cx. pipiens bioform molestus was unable to support MAYV infection, indicating its inability to contribute to the transmission cycle. CONCLUSIONS: In the event of accidental MAYV introduction in European territories, autochthonous outbreaks could potentially be sustained by two European species: Ae. albopictus and An. atroparvus. Entomological surveillance should also consider certain Anopheles species when monitoring MAYV transmission.


Assuntos
Aedes , Infecções por Alphavirus , Alphavirus , Culex , Mosquitos Vetores , Animais , Aedes/virologia , Mosquitos Vetores/virologia , Alphavirus/fisiologia , Alphavirus/isolamento & purificação , Culex/virologia , Europa (Continente) , Infecções por Alphavirus/transmissão , Infecções por Alphavirus/virologia , Saliva/virologia , Anopheles/virologia , Espanha , Itália , Feminino , Bélgica
7.
BMC Infect Dis ; 24(1): 463, 2024 May 02.
Artigo em Inglês | MEDLINE | ID: mdl-38698345

RESUMO

BACKGROUND: The use of temephos, the most common intervention for the chemical control of Aedes aegypti over the last half century, has disappointing results in control of the infection. The footprint of Aedes and the diseases it carries have spread relentlessly despite massive volumes of temephos. Recent advances in community participation show this might be more effective and sustainable for the control of the dengue vector. METHODS: Using data from the Camino Verde cluster randomized controlled trial, a compartmental mathematical model examines the dynamics of dengue infection with different levels of community participation, taking account of gender of respondent and exposure to temephos. RESULTS: Simulation of dengue endemicity showed community participation affected the basic reproductive number of infected people. The greatest short-term effect, in terms of people infected with the virus, was the combination of temephos intervention and community participation. There was no evidence of a protective effect of temephos 220 days after the onset of the spread of dengue. CONCLUSIONS: Male responses about community participation did not significantly affect modelled numbers of infected people and infectious mosquitoes. Our model suggests that, in the long term, community participation alone may have the best results. Adding temephos to community participation does not improve the effect of community participation alone.


Assuntos
Aedes , Participação da Comunidade , Dengue , Inseticidas , Temefós , Dengue/prevenção & controle , Dengue/transmissão , Humanos , Masculino , Feminino , Animais , Aedes/virologia , Adulto , Modelos Teóricos , Fatores Sexuais , Adulto Jovem , Adolescente , Controle de Mosquitos/métodos , Pessoa de Meia-Idade
8.
Sci Rep ; 14(1): 10814, 2024 05 11.
Artigo em Inglês | MEDLINE | ID: mdl-38734695

RESUMO

Chikungunya virus (CHIKV) poses a significant global health threat, re-emerging as a mosquito-transmitted pathogen that caused high fever, rash, and severe arthralgia. In Thailand, a notable CHIKV outbreak in 2019-2020 affected approximately 20,000 cases across 60 provinces, underscoring the need for effective mosquito control protocols. Previous studies have highlighted the role of midgut bacteria in the interaction between mosquito vectors and pathogen infections, demonstrating their ability to protect the insect from invading pathogens. However, research on the midgut bacteria of Aedes (Ae.) aegypti, the primary vector for CHIKV in Thailand remains limited. This study aims to characterize the bacterial communities in laboratory strains of Ae. aegypti, both infected and non-infected with CHIKV. Female mosquitoes from a laboratory strain of Ae. aegypti were exposed to a CHIKV-infected blood meal through membrane feeding, while the control group received a non-infected blood meal. At 7 days post-infection (dpi), mosquito midguts were dissected for 16S rRNA gene sequencing to identify midgut bacteria, and CHIKV presence was confirmed by E1-nested RT-PCR using mosquito carcasses. The study aimed to compare the bacterial communities between CHIKV-infected and non-infected groups. The analysis included 12 midgut bacterial samples, divided into three groups: CHIKV-infected (exposed and infected), non-infected (exposed but not infected), and non-exposed (negative control). Alpha diversity indices and Bray-Curtis dissimilarity matrix revealed significant differences in bacterial profiles among the three groups. The infected group exhibited an increased abundance of bacteria genus Gluconobacter, while Asaia was prevalent in both non-infected and negative control groups. Chryseobacterium was prominent in the negative control group. These findings highlight potential alterations in the distribution and abundance of gut microbiomes in response to CHIKV infection status. This study provides valuable insights into the dynamic relationship between midgut bacteria and CHIKV, underscoring the potential for alterations in bacterial composition depending on infection status. Understanding the relationships between mosquitoes and their microbiota holds promise for developing new methods and tools to enhance existing strategies for disease prevention and control. This research advances our understanding of the circulating bacterial composition, opening possibilities for new approaches in combating mosquito-borne diseases.


Assuntos
Aedes , Vírus Chikungunya , Microbioma Gastrointestinal , Mosquitos Vetores , Animais , Feminino , Aedes/microbiologia , Aedes/virologia , Bactérias/genética , Bactérias/classificação , Bactérias/isolamento & purificação , Febre de Chikungunya/transmissão , Febre de Chikungunya/virologia , Vírus Chikungunya/genética , Vírus Chikungunya/isolamento & purificação , Vírus Chikungunya/fisiologia , Mosquitos Vetores/microbiologia , Mosquitos Vetores/virologia , RNA Ribossômico 16S/genética , Tailândia
9.
Sci Rep ; 14(1): 8930, 2024 04 18.
Artigo em Inglês | MEDLINE | ID: mdl-38637572

RESUMO

In the last decades, dengue has become one of the most widespread mosquito-borne arboviruses in the world, with an increasing incidence in tropical and temperate regions. The mosquito Aedes aegypti is the dengue primary vector and is more abundant in highly urbanized areas. Traditional vector control methods have showing limited efficacy in sustaining mosquito population at low levels to prevent dengue virus outbreaks. Considering disease transmission is not evenly distributed in the territory, one perspective to enhance vector control efficacy relies on identifying the areas that concentrate arbovirus transmission within an endemic city, i.e., the hotspots. Herein, we used a 13-month timescale during the SARS-Cov-2 pandemic and its forced reduction in human mobility and social isolation to investigate the spatiotemporal association between dengue transmission in children and entomological indexes based on adult Ae. aegypti trapping. Dengue cases and the indexes Trap Positive Index (TPI) and Adult Density Index (ADI) varied seasonally, as expected: more than 51% of cases were notified on the first 2 months of the study, and higher infestation was observed in warmer months. The Moran's Eigenvector Maps (MEM) and Generalized Linear Models (GLM) revealed a strong large-scale spatial structuring in the positive dengue cases, with an unexpected negative correlation between dengue transmission and ADI. Overall, the global model and the purely spatial model presented a better fit to data. Our results show high spatial structure and low correlation between entomological and epidemiological data in Foz do Iguaçu dengue transmission dynamics, suggesting the role of human mobility might be overestimated and that other factors not evaluated herein could be playing a significant role in governing dengue transmission.


Assuntos
Aedes , Dengue , Animais , Adulto , Criança , Humanos , Brasil/epidemiologia , Mosquitos Vetores , Análise Espacial , Cidades/epidemiologia
10.
Biochem Biophys Res Commun ; 711: 149914, 2024 Jun 04.
Artigo em Inglês | MEDLINE | ID: mdl-38608434

RESUMO

The steroid hormone ecdysone is essential for the reproduction and survival of insects. The hormone is synthesized from dietary sterols such as cholesterol, yielding ecdysone in a series of consecutive enzymatic reactions. In the insect orders Lepidoptera and Diptera a glutathione transferase called Noppera-bo (Nobo) plays an essential, but biochemically uncharacterized, role in ecdysteroid biosynthesis. The Nobo enzyme is consequently a possible target in harmful dipterans, such as disease-carrying mosquitoes. Flavonoid compounds inhibit Nobo and have larvicidal effects in the yellow-fever transmitting mosquito Aedes aegypti, but the enzyme is functionally incompletely characterized. We here report that within a set of glutathione transferase substrates the double-bond isomerase activity with 5-androsten-3,17-dione stands out with an extraordinary specific activity of 4000 µmol min-1 mg-1. We suggest that the authentic function of Nobo is catalysis of a chemically analogous ketosteroid isomerization in ecdysone biosynthesis.


Assuntos
Aedes , Aedes/enzimologia , Aedes/metabolismo , Animais , Glutationa Transferase/metabolismo , Glutationa/metabolismo , Ecdisona/metabolismo , Proteínas de Insetos/metabolismo , Especificidade por Substrato , Esteroide Isomerases/metabolismo , Esteroide Isomerases/genética , Mosquitos Vetores/metabolismo , Cetosteroides/metabolismo , Cetosteroides/química
11.
Proc Natl Acad Sci U S A ; 121(16): e2317978121, 2024 Apr 16.
Artigo em Inglês | MEDLINE | ID: mdl-38593069

RESUMO

Mosquito-borne flaviviruses such as dengue (DENV) and Zika (ZIKV) cause hundreds of millions of infections annually. The single-stranded RNA genome of flaviviruses is translated into a polyprotein, which is cleaved equally into individual functional proteins. While structural proteins are packaged into progeny virions and released, most of the nonstructural proteins remain intracellular and could become cytotoxic if accumulated over time. However, the mechanism by which nonstructural proteins are maintained at the levels optimal for cellular fitness and viral replication remains unknown. Here, we identified that the ubiquitin E3 ligase HRD1 is essential for flaviviruses infections in both mammalian hosts and mosquitoes. HRD1 directly interacts with flavivirus NS4A and ubiquitylates a conserved lysine residue for ER-associated degradation. This mechanism avoids excessive accumulation of NS4A, which otherwise interrupts the expression of processed flavivirus proteins in the ER. Furthermore, a small-molecule inhibitor of HRD1 named LS-102 effectively interrupts DENV2 infection in both mice and Aedes aegypti mosquitoes, and significantly disturbs DENV transmission from the infected hosts to mosquitoes owing to reduced viremia. Taken together, this study demonstrates that flaviviruses have evolved a sophisticated mechanism to exploit the ubiquitination system to balance the homeostasis of viral proteins for their own advantage and provides a potential therapeutic target to interrupt flavivirus infection and transmission.


Assuntos
Aedes , Infecções por Flavivirus , Flavivirus , Infecção por Zika virus , Zika virus , Animais , Camundongos , Flavivirus/genética , Zika virus/genética , Ubiquitina/metabolismo , Ligases/metabolismo , Proteínas Virais/metabolismo , Mamíferos
12.
Science ; 384(6693): eadn9524, 2024 Apr 19.
Artigo em Inglês | MEDLINE | ID: mdl-38669573

RESUMO

The commensal microbiota of the mosquito gut plays a complex role in determining the vector competence for arboviruses. In this study, we identified a bacterium from the gut of field Aedes albopictus mosquitoes named Rosenbergiella sp. YN46 (Rosenbergiella_YN46) that rendered mosquitoes refractory to infection with dengue and Zika viruses. Inoculation of 1.6 × 103 colony forming units (CFUs) of Rosenbergiella_YN46 into A. albopictus mosquitoes effectively prevents viral infection. Mechanistically, this bacterium secretes glucose dehydrogenase (RyGDH), which acidifies the gut lumen of fed mosquitoes, causing irreversible conformational changes in the flavivirus envelope protein that prevent viral entry into cells. In semifield conditions, Rosenbergiella_YN46 exhibits effective transstadial transmission in field mosquitoes, which blocks transmission of dengue virus by newly emerged adult mosquitoes. The prevalence of Rosenbergiella_YN46 is greater in mosquitoes from low-dengue areas (52.9 to ~91.7%) than in those from dengue-endemic regions (0 to ~6.7%). Rosenbergiella_YN46 may offer an effective and safe lead for flavivirus biocontrol.


Assuntos
Aedes , Vírus da Dengue , Mosquitos Vetores , Simbiose , Zika virus , Animais , Aedes/microbiologia , Aedes/virologia , Vírus da Dengue/fisiologia , Mosquitos Vetores/virologia , Mosquitos Vetores/microbiologia , Zika virus/fisiologia , Dengue/transmissão , Dengue/virologia , Dengue/prevenção & controle , Microbioma Gastrointestinal , Acetobacteraceae/fisiologia , Feminino , Proteínas do Envelope Viral/metabolismo , Proteínas do Envelope Viral/genética , Flavivirus/fisiologia , Flavivirus/genética , Infecção por Zika virus/transmissão , Infecção por Zika virus/virologia
13.
PLoS Negl Trop Dis ; 18(4): e0012053, 2024 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-38557981

RESUMO

BACKGROUND: Mosquito-borne arboviruses are expanding their territory and elevating their infection prevalence due to the rapid climate change, urbanization, and increased international travel and global trade. Various significant arboviruses, including the dengue virus, Zika virus, Chikungunya virus, and yellow fever virus, are all reliant on the same primary vector, Aedes aegypti. Consequently, the occurrence of arbovirus coinfection in mosquitoes is anticipated. Arbovirus coinfection in mosquitoes has two patterns: simultaneous and sequential. Numerous studies have demonstrated that simultaneous coinfection of arboviruses in mosquitoes is unlikely to exert mutual developmental influence on these viruses. However, the viruses' interplay within a mosquito after the sequential coinfection seems intricated and not well understood. METHODOLOGY/PRINCIPAL FINDINGS: We conducted experiments aimed at examining the phenomenon of arbovirus sequential coinfection in both mosquito cell line (C6/36) and A. aegypti, specifically focusing on dengue virus (DENV, serotype 2) and Zika virus (ZIKV). We firstly observed that DENV and ZIKV can sequentially infect mosquito C6/36 cell line, but the replication level of the subsequently infected ZIKV was significantly suppressed. Similarly, A. aegypti mosquitoes can be sequentially coinfected by these two arboviruses, regardless of the order of virus exposure. However, the replication, dissemination, and the transmission potential of the secondary virus were significantly inhibited. We preliminarily explored the underlying mechanisms, revealing that arbovirus-infected mosquitoes exhibited activated innate immunity, disrupted lipid metabolism, and enhanced RNAi pathway, leading to reduced susceptibility to the secondary arbovirus infections. CONCLUSIONS/SIGNIFICANCE: Our findings suggest that, in contrast to simultaneous arbovirus coinfection in mosquitoes that can promote the transmission and co-circulation of these viruses, sequential coinfection appears to have limited influence on arbovirus transmission dynamics. However, it is important to note that more experimental investigations are needed to refine and expand upon this conclusion.


Assuntos
Aedes , Arbovírus , Coinfecção , Vírus da Dengue , Dengue , Infecção por Zika virus , Zika virus , Animais , Coinfecção/epidemiologia , Mosquitos Vetores , Dengue/epidemiologia
14.
Medicina (B Aires) ; 84(2): 189-195, 2024.
Artigo em Espanhol | MEDLINE | ID: mdl-38683503

RESUMO

OBJECTIVES: To monitor the oviposition activity of the mosquito Aedes aegypti and of dengue and chikungunya cases in four localities of temperate Argentina, during the 2023 epidemic. METHODS: During the summer and autumn of 2023, the oviposition activity of the mosquito vector was monitored weekly using ovitraps, and the arrival of cases with dengue or chikungunya in Tandil, Olavarría, Bahía Blanca and Laprida were registered. RESULTS: Monthly variations of the percentage of positive traps were similar in the first three locations; in Laprida the mosquito was not detected. On the contrary, a significant difference was observed in the percentage of total traps that ever tested positive in each locality, being higher in Olavarría (83.3%) than in Bahía Blanca (68.6%) and Tandil (48.7%). Regarding diseases, 18 imported cases of dengue and 3 of chikungunya were registered. In addition, the first autochthonous case of dengue in the region was recorded, being the southernmost until known. CONCLUSION: It is essential to raise awareness and train the members of the health systems of the new regions exposed to Ae. aegypti for early detection of cases, and to the general population to enhance prevention actions.


OBJETIVOS: Monitorear la actividad de oviposición del mosquito Aedes aegypti y de casos de dengue y chikungunya en cuatro localidades de Argentina templada, durante la epidemia del 2023. Métodos: Durante el verano y otoño del 2023, se monitoreó semanalmente mediante ovitrampas la actividad de oviposición del mosquito vector, y se registró el arribo de casos con dengue o chikungunya a Tandil, Olavarría, Bahía Blanca y Laprida. RESULTADOS: La variación mensual del porcentaje de trampas positivas fue similar en las tres primeras localidades; en Laprida no se detectó el mosquito. Por el contrario, se observó una diferencia significativa del porcentaje de trampas que alguna vez resultó positiva en cada localidad, siendo mayor en Olavarría (83%), que en Bahía Blanca (67%) y Tandil (49%). Respecto a las enfermedades, se registraron 18 casos importados de dengue y 3 de chikungunya. Además, se registró el primer caso autóctono de dengue en la región, siendo el más austral hasta el momento. Conclusión: Es imprescindible sensibilizar y capacitar a los integrantes de los sistemas de salud de las nuevas regiones expuestas al Ae. aegypti para la detección temprana de casos, y a la población en general para potenciar las acciones de prevención.


Assuntos
Aedes , Febre de Chikungunya , Dengue , Mosquitos Vetores , Estações do Ano , Argentina/epidemiologia , Dengue/epidemiologia , Dengue/transmissão , Dengue/prevenção & controle , Febre de Chikungunya/epidemiologia , Febre de Chikungunya/transmissão , Febre de Chikungunya/prevenção & controle , Animais , Aedes/virologia , Aedes/fisiologia , Mosquitos Vetores/fisiologia , Humanos , Epidemias , Feminino , Oviposição/fisiologia
15.
Viruses ; 16(4)2024 Mar 28.
Artigo em Inglês | MEDLINE | ID: mdl-38675870

RESUMO

In the last few years, there has been a dramatic increase in the number of discovered viruses that are transmitted by arthropods. Some of them are pathogenic for humans and mammals, and the pathogenic potential of others is unknown. The genus Orthoflavivirus belongs to the family Flaviviridae and includes arboviruses that cause severe human diseases with damage to the central nervous system and hemorrhagic fevers, as well as viruses with unknown vectors and viruses specific only to insects. The latter group includes Lammi virus, first isolated from a mosquito pool in Finland. It is known that Lammi virus successfully replicates in mosquito cell lines but not in mammalian cell cultures or mice. Lammi virus reduces the reproduction of West Nile virus during superinfection and thus has the potential to reduce the spread of West Nile virus in areas where Lammi virus is already circulating. In this work, we isolated Lammi virus from a pool of adult Aedes cinereus mosquitoes that hatched from larvae/pupae collected in Saint Petersburg, Russia. This fact may indicate transovarial transmission and trans-stadial survival of the virus.


Assuntos
Aedes , Mosquitos Vetores , Animais , Aedes/virologia , Federação Russa , Feminino , Mosquitos Vetores/virologia , Flaviviridae/fisiologia , Flaviviridae/isolamento & purificação , Flaviviridae/classificação , Flaviviridae/genética , Larva/virologia
16.
J Vector Borne Dis ; 61(1): 101-106, 2024 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-38648411

RESUMO

BACKGROUND OBJECTIVES: Dengue is a major vector-borne disease having public health importance. It is caused by Dengue Virus (DENV) and is transmitted by mosquitoes of Aedes species. With the unavailability of a vaccine, vector control remains the only preventive measure for dengue. Studies have already been conducted to establish the presence of dengue vectors in the north-eastern states of India. However, limited studies have been conducted in Tripura state. In the present study we aimed to identify the preferred breeding habitats of dengue vectors in the state. METHODS: Clinical case data of dengue since the last five years was studied and the areas with the highest case numbers were identified. Entomological investigation was carried out in areas reporting the highest number of cases. Larvae were collected from the breeding habitats using standard protocol followed by morphological and molecular identification. Further, House index (HI), Container index (CI) and Pupal index (PI) were determined. The positive pools were then processed for incrimination for the presence of dengue virus. Calculation of entomological indices was done. RESULTS: Of the total 815 containers searched, 36.80% containers were positive for mosquito larvae. Among the immature mosquito collection, 836 adults emerged and were identified as Aedes albopictus using standard taxonomic keys followed by molecular methods. HI, CI and PI, varied from 15.38% to 100%, 21% to 31.04 %, and 2.93% to 110.53% respectively. However, none of the pools was positive for dengue virus. INTERPRETATION CONCLUSION: The present study identified Ae. albopictus as a potential vector of dengue in Tripura. The study gave important insights on the preferred larval habitats and provides information on the indication of displacement of Ae. albopictus from rural to urban and semi-urban areas. However, longitudinal studies for longer time frame are necessary for any conclusive remarks.


Assuntos
Aedes , Vírus da Dengue , Dengue , Ecossistema , Larva , Mosquitos Vetores , Pupa , Animais , Índia , Larva/virologia , Larva/crescimento & desenvolvimento , Larva/fisiologia , Mosquitos Vetores/virologia , Mosquitos Vetores/fisiologia , Mosquitos Vetores/crescimento & desenvolvimento , Aedes/virologia , Aedes/fisiologia , Aedes/crescimento & desenvolvimento , Pupa/virologia , Pupa/crescimento & desenvolvimento , Dengue/transmissão , Humanos , Feminino
17.
J Vector Borne Dis ; 61(1): 136-142, 2024 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-38648416

RESUMO

BACKGROUND OBJECTIVES: Annual mass drug administration (MDA) is the main strategy to interrupt the transmission of lymphatic filariasis (LF) in the community. The main aim of monitoring the MDA program, for its effectiveness and interruption of LF is the post-MDA surveillance using antigen survey in children born after MDA. The latest technique of new research suggests that xenomonitoring is an effective tool for monitoring LF intervention. The objective of this study was to assess the W. bancrofti infection/or infectivity in vector mosquitoes by xenomonitoring during post-MDA surveillance. METHODS: A descriptive cross-sectional study was conducted in the hotspots of selected four districts of Central Nepal. A gravid trap technique was used for sampling mosquitoes. Infection/or infectivity was determined via the dissection of vector mosquitoes. Anopheles, Aedes, Armigerus and Culex species were collected from hotspots of four endemic districts, two from the hilly region (Lalitpur and Dhading) and two from Terai region (Bara and Mahottari) of Central Nepal. RESULTS: A total of 4450 mosquitoes belonging to four genera, Anopheles, Culex, Armigeres, and Aedes were collected from four hotspots. The distribution of Culex quinquefasciatus was found to be the highest, 88.9% (n=3955/4450) followed by Cx. vishnui (4.5%), Armigeres sp (5.8%), An. culicifascies (0.2%), Aedes spp (0.8%). The proportion of female mosquitoes trapped is significantly higher. A total of 3344 parous Cx. quinquefasciatus mosquitoes were dissected for any larval stage of W. bancrofti. We could not find any filarial infection in dissected mosquito samples. INTERPRETATION CONCLUSION: We conclude that the gravid trap is an efficient tool for the collection of gravid Cx. quinquefasciatus mosquitoes for xenomonitoring studies of filariasis endemic regions. Vector composition indicated a maximum number of vector mosquitoes of lymphatic filariasis were trapped compared with the other three species. Distribution and density of Cx. quinquefasciatus was found highest in four hotspots of endemic districts. None of the Cx. quinquefasciatus dissected were found to be infected by larval forms of filaria. Since the low levels of infection persistence in the human population in these hot spots, vector infection and infectivity can't be ignored. Microscopic xenomonitoring at a low level of infection persistent is less likely to be efficient so molecular xenomonitoring along with a large sample should be required in each of the hot spots of the districts. Additionally, area is receptive so further vector control intervention should be required to reduce the risk of resurgence of infection.


Assuntos
Aedes , Culex , Filariose Linfática , Administração Massiva de Medicamentos , Mosquitos Vetores , Wuchereria bancrofti , Filariose Linfática/epidemiologia , Filariose Linfática/transmissão , Filariose Linfática/prevenção & controle , Animais , Nepal/epidemiologia , Estudos Transversais , Mosquitos Vetores/parasitologia , Mosquitos Vetores/fisiologia , Humanos , Wuchereria bancrofti/isolamento & purificação , Culex/parasitologia , Culex/fisiologia , Aedes/fisiologia , Aedes/parasitologia , Feminino , Anopheles/parasitologia , Anopheles/fisiologia , Monitoramento Epidemiológico , Masculino , Doenças Endêmicas
18.
Acta Trop ; 254: 107205, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38579960

RESUMO

Lumpy skin disease virus (LSDV) is a transboundary viral disease in cattle and water buffaloes. Although this Poxvirus is supposedly transmitted by mechanical vectors, only a few studies have investigated the role of local vectors in the transmission of LSDV. This study examined the infection, dissemination, and transmission rates of LSDV in Aedes aegypti, Culex tritaeniorhynchus, and Culex quinquefasciatus following artificial membrane feeding of 102.7, 103.7, 104.7 TCID50/mL LSDV in sheep blood. The results demonstrated that these mosquito species were susceptible to LSDV, with Cx tritaeniorhynchus exhibiting significantly different characteristics from Ae. aegypti and Cx. quinquefasciatus. These three mosquito species were susceptible to LSDV. Ae. aegypti showed it as early as 2 days post-infection (dpi), indicating swift dissemination in this particular species. The extrinsic incubation period (EIP) of LSDV in Cx. tritaeniorhynchus and Cx. quinquefasciatus was 8 and 14 dpi, respectively. Ingestion of different viral titers in blood did not affect the infection, dissemination, or transmission rates of Cx. tritaeniorhynchus and Cx. quinquefasciatus. All rates remained consistently high at 8-14 dpi for Cx. tritaeniorhynchus. In all three species, LSDV remained detectable until 14 dpi. The present findings indicate that, Ae. aegypti, Cx. tritaeniorhynchus, and Cx. quinquefasciatus may act as vectors during the LSDV outbreak; their involvement may extend beyond being solely mechanical vectors.


Assuntos
Aedes , Culex , Vírus da Doença Nodular Cutânea , Animais , Culex/virologia , Aedes/virologia , Vírus da Doença Nodular Cutânea/isolamento & purificação , Vírus da Doença Nodular Cutânea/fisiologia , Ovinos , Doença Nodular Cutânea/transmissão , Doença Nodular Cutânea/virologia , Mosquitos Vetores/virologia , Feminino
19.
Parasit Vectors ; 17(1): 177, 2024 Apr 04.
Artigo em Inglês | MEDLINE | ID: mdl-38575981

RESUMO

BACKGROUND: Vertical transmission (VT) of arboviruses (arthropod-borne viruses) can serve as an essential link in the transmission cycle during adverse environmental conditions. The extent of VT among mosquito-borne arboviruses can vary significantly among different virus families and even among different viruses within the same genus. For example, orthobunyaviruses exhibit a higher VT rate than orthoflaviviruses and alphaviruses. Mosquitoes are also the natural hosts of a large number of insect-specific viruses (ISV) that belong to several virus families, including Bunyaviridae, Flaviviridae, and Togaviridae. Cell fusing agent virus (CFAV), an insect-specific orthoflavivirus, displays higher VT rates than other dual-host orthoflaviviruses, such as Zika and dengue viruses. High VT rates require establishment of stabilized infections in the germinal tissues of female vectors. To delve deeper into understanding the mechanisms governing these differences in VT rates and the establishment of stabilized infections, the ovary infection patterns and VT of Zika virus (ZIKV) and CFAV were compared. METHODS: Laboratory colonized Aedes aegypti females were infected with either ZIKV or CFAV by intrathoracic injection. Ovary infection patterns were monitored by in situ hybridization using virus-specific probes, and VT was determined by detecting the presence of the virus among the progeny, using a reverse-transcription quantitative polymerase chain reaction (PCR) assay. RESULTS: Both ZIKV and CFAV infect mosquito ovaries after intrathoracic injection. Infections then become widespread following a non-infectious blood meal. VT rates of ZIKV are similar to previously reported results (3.33%). CFAV, on the contrary transmits vertically very rarely. VT was not observed in the first gonotrophic cycle following intrathoracic injection, and only rarely in the second gonotrophic cycle. VT of CFAV is mosquito population independent, since similar results were obtained with Aedes aegypti collected from two different geographic locations. CONCLUSIONS: Although CFAV infects mosquito ovaries, the occurrence of VT remains infrequent in artificially infected Ae. aegypti, despite the observation of high VT rates in field-collected mosquitoes. These results suggest that infections of insect-specific viruses are stabilized in mosquitoes by some as yet unidentified mechanisms.


Assuntos
Aedes , Arbovírus , Vírus de Insetos , Infecção por Zika virus , Zika virus , Feminino , Animais , Mosquitos Vetores
20.
J Math Biol ; 88(6): 74, 2024 Apr 29.
Artigo em Inglês | MEDLINE | ID: mdl-38684552

RESUMO

In this paper, we propose a reaction-advection-diffusion dengue fever model with seasonal developmental durations and intrinsic incubation periods. Firstly, we establish the well-posedness of the model. Secondly, we define the basic reproduction number ℜ 0 for this model and show that ℜ 0 is a threshold parameter: if ℜ 0 < 1 , then the disease-free periodic solution is globally attractive; if ℜ 0 > 1 , the system is uniformly persistent. Thirdly, we study the global attractivity of the positive steady state when the spatial environment is homogeneous and the advection of mosquitoes is ignored. As an example, we use the model to investigate the dengue fever transmission case in Guangdong Province, China, and explore the impact of model parameters on ℜ 0 . Our findings indicate that ignoring seasonality may underestimate ℜ 0 . Additionally, the spatial heterogeneity of transmission may increase the risk of disease transmission, while the increase of seasonal developmental durations, intrinsic incubation periods and advection rates can all reduce the risk of disease transmission.


Assuntos
Número Básico de Reprodução , Dengue , Período de Incubação de Doenças Infecciosas , Conceitos Matemáticos , Modelos Biológicos , Mosquitos Vetores , Estações do Ano , Dengue/transmissão , Número Básico de Reprodução/estatística & dados numéricos , Animais , Humanos , China/epidemiologia , Mosquitos Vetores/crescimento & desenvolvimento , Mosquitos Vetores/virologia , Aedes/virologia , Aedes/crescimento & desenvolvimento , Modelos Epidemiológicos , Vírus da Dengue/crescimento & desenvolvimento , Simulação por Computador
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...