Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 2.698
Filtrar
1.
Biochem Biophys Res Commun ; 711: 149914, 2024 Apr 07.
Artículo en Inglés | MEDLINE | ID: mdl-38608434

RESUMEN

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.

2.
Proc Natl Acad Sci U S A ; 121(16): e2317978121, 2024 Apr 16.
Artículo en Inglés | MEDLINE | ID: mdl-38593069

RESUMEN

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.


Asunto(s)
Aedes , Infecciones por Flavivirus , Flavivirus , Infección por el Virus Zika , Virus Zika , Animales , Ratones , Flavivirus/genética , Virus Zika/genética , Ubiquitina/metabolismo , Ligasas/metabolismo , Proteínas Virales/metabolismo , Mamíferos
3.
Parasit Vectors ; 17(1): 177, 2024 Apr 04.
Artículo en Inglés | MEDLINE | ID: mdl-38575981

RESUMEN

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.


Asunto(s)
Aedes , Arbovirus , Virus de Insectos , Infección por el Virus Zika , Virus Zika , Femenino , Animales , Mosquitos Vectores
4.
PLoS Negl Trop Dis ; 18(4): e0012053, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38557981

RESUMEN

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.


Asunto(s)
Aedes , Arbovirus , Coinfección , Virus del Dengue , Dengue , Infección por el Virus Zika , Virus Zika , Animales , Coinfección/epidemiología , Mosquitos Vectores , Dengue/epidemiología
5.
Exp Biol Med (Maywood) ; 249: 10114, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38510492

RESUMEN

Bunyamwera virus (BUNV) (Bunyamwera orthobunyavirus) has been found in Sub-Saharan Africa and demonstrated recently as cocirculating with Rift Valley Fever Virus (RVFV). Little is known regarding the breadth of transmission modalities of Bunyamwera. Given its co-occurence with RVFV, we hypothesized the transmission system of BUNV shared similarities to the RVFV system including transmission by Ae. aegypti mosquitoes and environmentally mediated transmission through fomites and environmental contamination. We exposed Ae. aegypti mosquitoes to BUNV and evaluated their ability to transmit both vertically and horizontally. Further, we investigated the potential for a novel transmission modality via environmental contamination. We found that the LSU colony of Ae. aegypti was not competent for the virus for either horizontal or vertical transmission; but, 20% of larva exposed to virus via contaminated aquatic habitat were positive. However, transstadial clearance of the virus was absolute. Finally, under simulated temperature conditions that matched peak transmission in Rwanda, we found that BUNV was stable in both whole blood and serum for up to 28 days at higher total volume in tubes at moderate quantities (103-5 genome copies/mL). In addition, infectiousness of these samples was demonstrated in 80% of the replicates. At lower volume samples (in plates), infectiousness was retained out to 6-8 days with a maximum infectious titer of 104 PFU/mL. Thus, the potential for contamination of the environment and/or transmission via contaminated fomites exists. Our findings have implications for biosafety and infection control, especially in the context of food animal production.


Asunto(s)
Aedes , Virus Bunyamwera , Virus de la Fiebre del Valle del Rift , Animales , Virus de la Fiebre del Valle del Rift/genética
6.
bioRxiv ; 2024 Mar 03.
Artículo en Inglés | MEDLINE | ID: mdl-38463973

RESUMEN

During major, recent yellow fever (YF) epidemics in Brazil, human cases were attributed only to spillover infections from sylvatic transmission with no evidence of human amplification. Furthermore, the historic absence of YF in Asia, despite abundant peridomestic Aedes aegypti and naive human populations, represents a longstanding enigma. We tested the hypothesis that immunity from dengue (DENV) and Zika (ZIKV) flaviviruses limits YF virus (YFV) viremia and transmission by Ae. aegypti . Prior DENV and ZIKV immunity consistently suppressed YFV viremia in experimentally infected macaques, leading to reductions in Ae. aegypti infection when mosquitoes were fed on infected animals. These results indicate that, in DENV- and ZIKV-endemic regions such as South America and Asia, flavivirus immunity suppresses YFV human amplification potential, reducing the risk of urban outbreaks. One-Sentence Summary: Immunity from dengue and Zika viruses suppresses yellow fever viremia, preventing infection of mosquitoes and reducing the risk of epidemics.

7.
Nat Commun ; 15(1): 2682, 2024 Mar 27.
Artículo en Inglés | MEDLINE | ID: mdl-38538621

RESUMEN

Mosquito-borne dengue (DENV) and Zika (ZIKV) viruses originated in Old World sylvatic (forest) cycles involving monkeys and canopy-living Aedes mosquitoes. Both viruses spilled over into human transmission and were translocated to the Americas, opening a path for spillback into Neotropical sylvatic cycles. Studies of the trade-offs that shape within-host dynamics and transmission of these viruses are lacking, hampering efforts to predict spillover and spillback. We infected a native, Asian host species (cynomolgus macaque) and a novel, American host species (squirrel monkey) with sylvatic strains of DENV-2 or ZIKV via mosquito bite. We then monitored aspects of viral replication (viremia), innate and adaptive immune response (natural killer (NK) cells and neutralizing antibodies, respectively), and transmission to mosquitoes. In both hosts, ZIKV reached high titers that translated into high transmission to mosquitoes; in contrast DENV-2 replicated to low levels and, unexpectedly, transmission occurred only when serum viremia was below or near the limit of detection. Our data reveal evidence of an immunologically-mediated trade-off between duration and magnitude of virus replication, as higher peak ZIKV titers are associated with shorter durations of viremia, and higher NK cell levels are associated with lower peak ZIKV titers and lower anti-DENV-2 antibody levels. Furthermore, patterns of transmission of each virus from a Neotropical monkey suggest that ZIKV has greater potential than DENV-2 to establish a sylvatic transmission cycle in the Americas.


Asunto(s)
Aedes , Virus del Dengue , Dengue , Infección por el Virus Zika , Virus Zika , Animales , Humanos , Viremia
8.
Sci Rep ; 14(1): 7424, 2024 03 28.
Artículo en Inglés | MEDLINE | ID: mdl-38548897

RESUMEN

The Zika virus (ZIKV) is a serious global public health crisis. A major control challenge is its multiple transmission modes. This paper aims to simulate the transmission patterns of ZIKV using a dynamic process-based epidemiological model written in ordinary differential equations, which incorporates the human-to-mosquito infection by bites and sewage, mosquito-to-human infection by bites, and human-to-human infection by sex. Mathematical analyses are carried out to calculate the basic reproduction number and backward bifurcation, and prove the existence and stability of the equilibria. The model is validated with infection data by applying it to the 2015-2016 ZIKV epidemic in Brazil. The results indicate that the reproduction number is estimated to be 2.13, in which the contributions by mosquito bite, sex and sewage account for 85.7%, 3.5% and 10.8%, respectively. This number and the morbidity rate are most sensitive to parameters related to mosquito ecology, rather than asymptomatic or human-to-human transmission. Multiple transmission routes and suitable temperature exacerbate ZIKV infection in Brazil, and the vast majority of human infection cases were prevented by the intervention implemented. These findings may provide new insights to improve the risk assessment of ZIKV infection.


Asunto(s)
Aedes , Epidemias , Infección por el Virus Zika , Virus Zika , Animales , Humanos , Brasil/epidemiología , Aguas del Alcantarillado
9.
Viruses ; 16(2)2024 Jan 24.
Artículo en Inglés | MEDLINE | ID: mdl-38399952

RESUMEN

The frequency of locally transmitted dengue virus (DENV) infections has increased in Europe in recent years, facilitated by the invasive mosquito species Aedes albopictus, which is well established in a large area of Europe. In Italy, the first indigenous dengue outbreak was reported in August 2020 with 11 locally acquired cases in the Veneto region (northeast Italy), caused by a DENV-1 viral strain closely related to a previously described strain circulating in Singapore and China. In this study, we evaluated the vector competence of two Italian populations of Ae. albopictus compared to an Ae. aegypti lab colony. We performed experimental infections using a DENV-1 strain that is phylogenetically close to the strain responsible for the 2020 Italian autochthonous outbreak. Our results showed that local Ae. albopictus is susceptible to infection and is able to transmit the virus, confirming the relevant risk of possible outbreaks starting from an imported case.


Asunto(s)
Aedes , Virus del Dengue , Dengue , Animales , Humanos , Dengue/epidemiología , Brotes de Enfermedades
10.
Viruses ; 16(2)2024 Jan 31.
Artículo en Inglés | MEDLINE | ID: mdl-38399996

RESUMEN

Snowshoe hare virus (SSHV) is a zoonotic arthropod-borne virus (arbovirus) circulating in colder areas of the Northern Hemisphere. SSHV is maintained in an enzootic cycle between small mammals and mosquitoes, assumably of the genera Aedes and Culiseta. Symptoms of SSHV human infection can range from asymptomatic to severe neuroinvasive disease. Studies on SSHV transmission are limited, and there is no information available on whether mosquitoes of the genus Culex are able to transmit SSHV. Therefore, we investigated six mosquito species via salivation assay for their vector competence. We demonstrated that SSHV can be transmitted by the abundant European Culex species Cx. pipiens biotype pipiens, Cx. pipiens biotype molestus, and Cx. torrentium with low transmission efficiency between 3.33% and 6.67%. Additionally, the invasive species Ae. albopictus can also transmit SSHV with a low transmission efficiency of 3.33%. Our results suggest that local transmission of SSHV after introduction to Europe seems to be possible from a vector perspective.


Asunto(s)
Aedes , Culex , Virus de la Encefalitis de California , Animales , Humanos , Mosquitos Vectores , Europa (Continente) , Mamíferos
11.
Viruses ; 16(2)2024 Feb 02.
Artículo en Inglés | MEDLINE | ID: mdl-38400018

RESUMEN

Noncoding RNAs (ncRNAs) constitute a class of RNA molecules that lack protein-coding capacity. ncRNAs frequently modulate gene expression through specific interactions with target proteins or messenger RNAs, thereby playing integral roles in a wide array of cellular processes. The Flavivirus genus comprises several significant members, such as dengue virus (DENV), Zika virus (ZIKV), and yellow fever virus (YFV), which have caused global outbreaks, resulting in high morbidity and mortality in human populations. The life cycle of arthropod-borne flaviviruses encompasses their transmission between hematophagous insect vectors and mammalian hosts. During this process, a complex three-way interplay occurs among the pathogen, vector, and host, with ncRNAs exerting a critical regulatory influence. ncRNAs not only constitute a crucial regulatory mechanism that has emerged from the coevolution of viruses and their hosts but also hold potential as antiviral targets for controlling flavivirus epidemics. This review introduces the biogenesis of flavivirus-derived ncRNAs and summarizes the regulatory roles of ncRNAs in viral replication, vector-mediated viral transmission, antiviral innate immunity, and viral pathogenicity. A profound comprehension of the interplay between ncRNAs and flaviviruses will help formulate efficacious prophylactic and therapeutic strategies against flavivirus-related diseases.


Asunto(s)
Flavivirus , Infección por el Virus Zika , Virus Zika , Animales , Humanos , Flavivirus/genética , Virus Zika/genética , Virus Zika/metabolismo , Virulencia , Replicación Viral , Proteínas/metabolismo , ARN no Traducido/genética , ARN no Traducido/metabolismo , Antivirales/metabolismo , Mamíferos
12.
Am J Trop Med Hyg ; 110(4): 724-730, 2024 Apr 03.
Artículo en Inglés | MEDLINE | ID: mdl-38377614

RESUMEN

Since the Zika virus (ZIKV) pandemic in 2015-2017, there has been a near absence of reported cases in the Americas outside of Brazil. However, the conditions for Aedes-borne transmission persist in Latin America, and the threat of ZIKV transmission is increasing as population immunity wanes. Mexico has reported only 70 cases of laboratory-confirmed ZIKV infection since 2020, with no cases recorded in the Yucatán peninsula. Here, we provide evidence of active ZIKV transmission, despite the absence of official case reports, in the city of Mérida, Mexico, the capital of the state of Yucatán. Capitalizing on an existing cohort, we detected cases in participants with symptoms consistent with flavivirus infection from 2021 to 2022. Serum samples from suspected cases were tested for ZIKV RNA by polymerase chain reaction or ZIKV-reactive IgM by ELISA. To provide more specific evidence of exposure, focus reduction neutralization tests were performed on ELISA-positive samples. Overall, we observed 25 suspected ZIKV infections for an estimated incidence of 2.8 symptomatic cases per 1,000 persons per year. Our findings emphasize the continuing threat of ZIKV transmission in the setting of decreased surveillance and reporting.


Asunto(s)
Aedes , Infección por el Virus Zika , Virus Zika , Animales , Humanos , México/epidemiología , Américas/epidemiología
13.
Acta Trop ; 253: 107159, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38412904

RESUMEN

Widespread resurgence of dengue outbreaks has seriously threatened the global health. Due to lack of treatments and vaccines, one key strategy in dengue control is to reduce the vector population size. As an environment-friendly mosquito control approach, releasing male mosquitoes transinfected with specific Wolbachia strain into the field to suppress the wild mosquito population size has become wildly accepted. The current study evaluates the effectiveness of this suppression strategy on dengue control under changing temperature and precipitation profiles. We formulate a mathematical model which includes larval intra-specific competition, the maturation period for mosquitoes, the extrinsic incubation period (EIP) and intrinsic incubation period (IIP). The persistence of mosquitoes and disease is discussed in terms of two basic reproduction numbers (RM and R0) and the release ratio pw. Further numerical simulations are carried out to not only validate theoretical results, but also provide interesting quantitative observations. Sensitivity analysis on the reproduction numbers, peak size, peak time and the final epidemic size is performed with respect to model parameters, which highlights effective control measures against dengue transmission. Moreover, by assuming temperature and precipitation dependent mosquito-related parameters, the model can be used to project the effectiveness of releasing Wolbachia-carrying males under climatic variations. It is shown that the effectiveness of various control strategies is highly dependent on the changing temperature and precipitation profiles. In particular, the model projects that it is most challenging to control the disease at the favorable temperature (around 27∼30∘C) and precipitation (5∼8mm/day) range, during which the basic reproduction number R0 is very high and more Wolbachia-infected males should be released.


Asunto(s)
Aedes , Dengue , Wolbachia , Animales , Masculino , Dengue/epidemiología , Temperatura , Control de Mosquitos , Modelos Teóricos , Mosquitos Vectores
14.
Viruses ; 16(1)2024 Jan 04.
Artículo en Inglés | MEDLINE | ID: mdl-38257784

RESUMEN

In viral disease research, few diseases can compete with yellow fever for the volume of literature, historical significance, richness of the topics and the amount of strong interest among both scientists and laypersons. While the major foci of viral disease research shifted to other more pressing new diseases in recent decades, many critically important basic tasks still remain unfinished for yellow fever. Some of the examples include the mechanisms of transmission, the process leading to outbreak occurrence, environmental factors, dispersal, and viral persistence in nature. In this review, these subjects are analyzed in depth, based on information not only in old but in modern literatures, to fill in blanks and to update the current understanding on these topics. As a result, many valuable facts, ideas, and other types of information that complement the present knowledge were discovered. Very serious questions about the validity of the arbovirus concept and some research practices were also identified. The characteristics of YFV and its pattern of transmission that make this virus unique among viruses transmitted by Ae. aegypti were also explored. Another emphasis was identification of research questions. The discovery of a few historical surprises was an unexpected benefit.


Asunto(s)
Médicos , Fiebre Amarilla , Humanos , Fiebre Amarilla/epidemiología , Brotes de Enfermedades
15.
PLoS One ; 19(1): e0295317, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38271346

RESUMEN

The high infestation of Aedes aegypti populations in Urabá, Antioquia, Colombia represents a risk factor for increased dengue morbidity and mortality. This study aimed to determine the risk of dengue transmission by estimating the population of Ae. aegypti using entomological indices, density of females per dwelling and inhabitant, and virological surveillance in two municipalities in Colombia. A cross-sectional study was conducted with quarterly entomological surveys in three neighborhoods of Apartadó and Turbo between 2021 and 2022. Aedes indices and vector density per dwelling and per inhabitant were calculated. The Kernel method was used for spatial analysis, and correlations between climatic variables and mosquito density were examined. Virus detection and serotyping in mosquitoes was performed using single-step reverse transcription polymerase chain reaction. The housing, reservoir, and Breteau indices were 48.9%, 29.5%, and 70.2%, respectively. The mean density of Ae. aegypti was 1.47 females / dwelling and 0.51 females / inhabitant. The overall visual analysis showed several critical points in the neighborhoods studied. There was significant correlation of vector density and relative humidity and precipitation in the neighborhoods 29 de noviembre and 24 de diciembre. Additionally, serotypes DENV-1 and DENV-2 were found. The overall indices for dwellings, reservoirs, and Breteau were lower than those recorded in 2014 in Urabá. The vector density results in this study were similar to those reported in other studies conducted in Latin America, and vector infection was detected. The Aedes and density indices are complementary, emphasizing the importance of continuous surveillance of Ae. aegypti to inform appropriate control strategies and prevent future dengue outbreaks in these municipalities.


Asunto(s)
Aedes , Virus del Dengue , Dengue , Animales , Femenino , Ciudades , Colombia/epidemiología , Estudios Transversales , Mosquitos Vectores
16.
Parasitol Res ; 123(1): 102, 2024 Jan 18.
Artículo en Inglés | MEDLINE | ID: mdl-38233721

RESUMEN

Unplanned human population shifts in urban areas are expected to increase the prevalence of vector-borne diseases. This study aimed to investigate mosquito species composition, blood meal sources, and malaria vectors in an urban area. Indoor-resting adult mosquitoes were collected using Prokopack and host-seeking mosquitoes using Centers for Disease Control and Prevention light traps in Arba Minch town. Larval collection from artificial containers was done in those houses selected for adult mosquito collection. Anopheles adults collected and emerged from larvae were identified morphologically using a taxonomic key. ELISA was used to identify blood meal sources in freshly fed Anopheles and Culex mosquitoes, and CSP of Anopheles mosquitoes. A total of 16,756 female mosquitoes were collected. Of these, 93% (15,571) were Culex, 6% (1016) were Anopheles, and 1% (169) were Aedes mosquitoes. Out of the 130 adult mosquitoes that were raised from larvae collected from the containers, 20% were An. rhodesiensis, while the remaining 80% were Aedes mosquitoes. Out of 823 mosquitoes tested for blood meal origins, 86.3% (710/823) tested positive for human blood, 2.2% (18/823) tested positive for bovine blood, and 11.5% (95/823) were negative for human and bovine antibodies. Anopheles gambiae complex had a human blood meal index (HBI) of 50% (90/180; CI 42.3-57.5%) and a bovine blood meal index (BBI) of only 0.5% (95% CI 0.01-3.1%). Culex HBI was 96.7% (620/641), and its BBI index was 2.4% (15/641). While it was low (0.8%) in Culex, the proportion of An. gambiae complex with unidentified blood meal sources was 49.5% (95 CI% 41.9-56.9%). Among the 1016 Anopheles mosquitoes tested, a single An. gambiae complex (0.1%; 1/1016) was positive for P. vivax CSP. The high HBI indicates frequent contact between humans and vectors. To reduce human exposure, personal protection tools should be implemented.


Asunto(s)
Aedes , Anopheles , Culex , Malaria Vivax , Malaria , 60509 , Humanos , Animales , Femenino , Bovinos , Etiopía/epidemiología , Mosquitos Vectores , Malaria/epidemiología , Conducta Alimentaria
17.
bioRxiv ; 2024 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-38293180

RESUMEN

Background: Since its reemergence in 2017, yellow fever (YF) has been active in Nigeria. The Nigeria Centre for Disease Control (NCDC) has coordinated responses to the outbreaks with the support of the World Health Organization (WHO). The National Arbovirus and Vectors Research Centre (NAVRC) handles the vector component of these responses. This study sought to identify the vectors driving YF transmission and any of the targeted arboviruses and their distribution across states. Methods: Eggs, larvae and pupae as well as adult mosquitoes were collected in observational, analytical, and cross-sectional surveys conducted in sixteen YF outbreak states between 2017 and 2020. Adult mosquitoes (field-collected or reared from immature stages) were morphologically identified, and arboviruses were detected using RT-qPCR at the African Centre of Excellence for Genomics of Infectious Diseases (ACEGID). Results: Aedes mosquitoes were collected in eleven of the sixteen states surveyed and the mosquitoes in nine states were found infected with arboviruses. A total of seven Aedes species were collected from different parts of the country. Aedes aegypti was the most dominant (51%) species, whereas Aedes africanus was the least (0.2%). Yellow fever virus (YFV) was discovered in 33 (~26%) out of the 127 Aedes mosquito pools. In addition to YFV, the Chikungunya virus (CHIKV) was found in nine pools. Except for Ae. africanus, all the Aedes species tested positive for at least one arbovirus. YFV-positive pools were found in six (6) Aedes species while CHIKV-positive pools were only recorded in two Aedes species. Edo State had the most positive pools (16), while Nasarawa, Imo, and Anambra states had the least (1 positive pool). Breteau and house indices were higher than normal transmission thresholds in all but one state. Conclusion: In Nigeria, there is a substantial risk of arbovirus transmission by Aedes mosquitoes, with YFV posing the largest threat at the moment. This risk is heightened by the fact that YFV and CHIKV have been detected in vectors across outbreak locations. Hence, there is an urgent need to step up arbovirus surveillance and control activities in the country.

18.
Lancet Planet Health ; 8(1): e30-e40, 2024 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-38199719

RESUMEN

BACKGROUND: Estimates of the spatiotemporal distribution of different mosquito vector species and the associated risk of transmission of arboviruses are key to design adequate policies for preventing local outbreaks and reducing the number of human infections in endemic areas. In this study, we quantified the abundance of Aedes albopictus and Aedes aegypti and the local transmission potential for three arboviral infections at an unprecedented spatiotemporal resolution in areas where no entomological surveillance is available. METHODS: We developed a computational model to quantify the daily abundance of Aedes mosquitoes, leveraging temperature and precipitation records. The model was calibrated on mosquito surveillance data collected in 115 locations in Europe and the Americas between 2007 and 2018. Model estimates were used to quantify the reproduction number of dengue virus, Zika virus, and chikungunya in Europe and the Americas, at a high spatial resolution. FINDINGS: In areas colonised by both Aedes species, A aegypti was estimated to be the main vector for the transmission of dengue virus, Zika virus, and chikungunya, being associated with a higher estimate of R0 when compared with A albopictus. Our estimates highlighted that these arboviruses were endemic in tropical and subtropical countries, with the highest risks of transmission found in central America, Venezuela, Colombia, and central-east Brazil. A non-negligible potential risk of transmission was also estimated for Florida, Texas, and Arizona (USA). The broader ecological niche of A albopictus could contribute to the emergence of chikungunya outbreaks and clusters of dengue autochthonous cases in temperate areas of the Americas, as well as in mediterranean Europe (in particular, in Italy, southern France, and Spain). INTERPRETATION: Our results provide a comprehensive overview of the transmission potential of arboviral diseases in Europe and the Americas, highlighting areas where surveillance and mosquito control capacities should be prioritised. FUNDING: EU and Ministero dell'Università e della Ricerca, Italy (Piano Nazionale di Ripresa e Resilienza Extended Partnership initiative on Emerging Infectious Diseases); EU (Horizon 2020); Ministero dell'Università e della Ricerca, Italy (Progetti di ricerca di Rilevante Interesse Nazionale programme); Brazilian National Council of Science, Technology and Innovation; Ministry of Health, Brazil; and Foundation of Research for Minas Gerais, Brazil.


Asunto(s)
Aedes , Arbovirus , Fiebre Chikungunya , Infección por el Virus Zika , Virus Zika , Humanos , Animales , Fiebre Chikungunya/epidemiología , Europa (Continente)/epidemiología , Infección por el Virus Zika/epidemiología
19.
Nat Microbiol ; 9(2): 377-389, 2024 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-38263454

RESUMEN

Buruli ulcer, a chronic subcutaneous infection caused by Mycobacterium ulcerans, is increasing in prevalence in southeastern Australia. Possums are a local wildlife reservoir for M. ulcerans and, although mosquitoes have been implicated in transmission, it remains unclear how humans acquire infection. We conducted extensive field survey analyses of M. ulcerans prevalence among mosquitoes in the Mornington Peninsula region of southeastern Australia. PCR screening of trapped mosquitoes revealed a significant association between M. ulcerans and Aedes notoscriptus. Spatial scanning statistics revealed overlap between clusters of M. ulcerans-positive Ae. notoscriptus, M. ulcerans-positive possum excreta and Buruli ulcer cases, and metabarcoding analyses showed individual mosquitoes had fed on humans and possums. Bacterial genomic analysis confirmed shared single-nucleotide-polymorphism profiles for M. ulcerans detected in mosquitoes, possum excreta and humans. These findings indicate Ae. notoscriptus probably transmit M. ulcerans in southeastern Australia and highlight mosquito control as a Buruli ulcer prevention measure.


Asunto(s)
Aedes , Úlcera de Buruli , Mycobacterium ulcerans , Animales , Humanos , Úlcera de Buruli/epidemiología , Úlcera de Buruli/genética , Úlcera de Buruli/microbiología , Mycobacterium ulcerans/genética , Australia , Genoma Bacteriano , Aedes/genética
20.
PLoS Negl Trop Dis ; 18(1): e0011408, 2024 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-38295108

RESUMEN

The distribution and intensity of viral diseases transmitted by Aedes aegypti mosquitoes, including dengue, have rapidly increased over the last century. Here, we study dengue virus (DENV) transmission across the ecologically and demographically distinct regions or Ecuador. We analyzed province-level age-stratified dengue incidence data from 2000-2019 using catalytic models to estimate the force of infection of DENV over eight decades. We found that provinces established endemic DENV transmission at different time periods. Coastal provinces with the largest and most connected cities had the earliest and highest increase in DENV transmission, starting around 1980 and continuing to the present. In contrast, remote and rural areas with reduced access, like the northern coast and the Amazon regions, experienced a rise in DENV transmission and endemicity only in the last 10 to 20 years. The newly introduced chikungunya and Zika viruses have age-specific distributions of hospital-seeking cases consistent with recent emergence across all provinces. To evaluate factors associated with geographic differences in DENV transmission potential, we modeled DENV vector risk using 11,693 Aedes aegypti presence points to the resolution of 1 hectare. In total, 56% of the population of Ecuador, including in provinces identified as having increasing DENV transmission in our models, live in areas with high risk of Aedes aegypti, with population size, trash collection, elevation, and access to water as important determinants. Our investigation serves as a case study of the changes driving the expansion of DENV and other arboviruses globally and suggest that control efforts should be expanded to semi-urban and rural areas and to historically isolated regions to counteract increasing dengue outbreaks.


Asunto(s)
Aedes , Virus del Dengue , Dengue , Infección por el Virus Zika , Virus Zika , Animales , Humanos , Ecuador/epidemiología , Mosquitos Vectores , Factores de Riesgo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...