Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 1.358
Filtrar
2.
J Virol ; 97(10): e0116223, 2023 10 31.
Artigo em Inglês | MEDLINE | ID: mdl-37800949

RESUMO

IMPORTANCE: Previously, we modeled direct transmission chains of Zika virus (ZIKV) by serially passaging ZIKV in mice and mosquitoes and found that direct mouse transmission chains selected for viruses with increased virulence in mice and the acquisition of non-synonymous amino acid substitutions. Here, we show that these same mouse-passaged viruses also maintain fitness and transmission capacity in mosquitoes. We used infectious clone-derived viruses to demonstrate that the substitution in nonstructural protein 4A contributes to increased virulence in mice.


Assuntos
Culicidae , Aptidão Genética , Mosquitos Vetores , Virulência , Zika virus , Animais , Camundongos , Culicidae/virologia , Mosquitos Vetores/virologia , Virulência/genética , Zika virus/química , Zika virus/genética , Zika virus/patogenicidade , Infecção por Zika virus/transmissão , Infecção por Zika virus/virologia , Inoculações Seriadas , Substituição de Aminoácidos , Aptidão Genética/genética
4.
Chaos ; 32(4): 041105, 2022 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-35489839

RESUMO

Over the last decade, the release of Wolbachia-infected Aedes aegypti into the natural habitat of this mosquito species has become the most sustainable and long-lasting technique to prevent and control vector-borne diseases, such as dengue, zika, or chikungunya. However, the limited resources to generate such mosquitoes and their effective distribution in large areas dominated by the Aedes aegypti vector represent a challenge for policymakers. Here, we introduce a mathematical framework for the spread of dengue in which competition between wild and Wolbachia-infected mosquitoes, the cross-contagion patterns between humans and vectors, the heterogeneous distribution of the human population in different areas, and the mobility flows between them are combined. Our framework allows us to identify the most effective areas for the release of Wolbachia-infected mosquitoes to achieve a large decrease in the global dengue prevalence.


Assuntos
Aedes/microbiologia , Febre de Chikungunya/prevenção & controle , Dengue/prevenção & controle , Mosquitos Vetores/microbiologia , Wolbachia/fisiologia , Infecção por Zika virus/prevenção & controle , Animais , Febre de Chikungunya/epidemiologia , Febre de Chikungunya/transmissão , Dengue/epidemiologia , Dengue/transmissão , Humanos , Controle de Mosquitos/economia , Wolbachia/crescimento & desenvolvimento , Infecção por Zika virus/epidemiologia , Infecção por Zika virus/transmissão
5.
Rev. enferm. Inst. Mex. Seguro Soc ; 30(1): 1-3, 04-abr-2022.
Artigo em Espanhol | LILACS, BDENF - Enfermagem | ID: biblio-1378881

RESUMO

La enfermedad por virus del Zika ha sufrido una importante disminución en la notificación de casos. A nivel mundial se observa un descenso aproximado del 58%, comparado con el mismo periodo en 2020. En México ocurre una situación similar, pues en la semana epidemiológica 48 solamente se han confirmado 34 casos. Dicha situación coincide con la pandemia por SARS-CoV-2, la cual se vive desde el 2019; sin embargo, es de suma importancia reestablecer las acciones de vigilancia epidemiológica enfocadas en el Zika para así continuar con las medidas de prevención y control dirigidas a minimizar el impacto de la enfermedad.


Zika virus disease has suffered a significant decrease in case reporting. Worldwide, an approximate decrease of 58% is observed, compared to the same period in 2020. Mexico is experiencing a similar situation, given that at epidemiological week 48 only 34 cases have been confirmed. This situation coincides with the SARS-CoV-2 pandemic, which has been experienced since 2019; however, it is of the utmost importance to reestablish epidemiological surveillance actions aimed at Zika to continue with prevention and control measures focused on minimizing the impact of the illness.


Assuntos
Humanos , Masculino , Feminino , Pandemias/história , Zika virus/crescimento & desenvolvimento , Infecção por Zika virus/transmissão , SARS-CoV-2/crescimento & desenvolvimento , Monitoramento Epidemiológico , México/epidemiologia
7.
Viruses ; 14(2)2022 02 11.
Artigo em Inglês | MEDLINE | ID: mdl-35215967

RESUMO

Zika virus (ZIKV), a re-emerging virus, causes congenital brain abnormalities and Guillain-Barré syndrome. It is mainly transmitted by Aedes mosquitoes, but infections are also linked to sexual transmissions. Infectious ZIKV has been isolated, and viral RNA has been detected in semen over a year after the onset of initial symptoms, but the mode of long-term persistence is not yet understood. ZIKV can proliferate in human Sertoli cells (HSerC) for several weeks in vitro, suggesting that it might be a reservoir for persistent ZIKV infection. This study determined proteomic changes in HSerC during ZIKV infections by TMT-mass spectrometry analysis. Levels of 4416 unique Sertoli cell proteins were significantly altered at 3, 5, and 7 days after ZIKV infection. The significantly altered proteins include enzymes, transcription regulators, transporters, kinases, peptidases, transmembrane receptors, cytokines, ion channels, and growth factors. Many of these proteins are involved in pathways associated with antiviral response, antigen presentation, and immune cell activation. Several immune response pathway proteins were significantly activated during infection, e.g., interferon signaling, T cell receptor signaling, IL-8 signaling, and Th1 signaling. The altered protein levels were linked to predicted activation of immune response in HSerC, which was predicted to suppress ZIKV infection. ZIKV infection also affected the levels of critical regulators of gluconeogenesis and glycolysis pathways such as phosphoglycerate mutase, phosphoglycerate kinase, and enolase. Interestingly, many significantly altered proteins were associated with cardiac hypertrophy, which may induce heart failure in infected patients. In summary, our research contributes to a better understanding of ZIKV replication dynamics and infection in Sertoli cells.


Assuntos
Sêmen/virologia , Células de Sertoli/imunologia , Replicação Viral , Infecção por Zika virus/imunologia , Metabolismo dos Carboidratos/imunologia , Doenças Cardiovasculares/imunologia , Transmissão de Doença Infecciosa , Humanos , Masculino , Processamento de Proteína Pós-Traducional , Proteômica , RNA Viral/genética , Células de Sertoli/virologia , Zika virus/isolamento & purificação , Infecção por Zika virus/transmissão
8.
PLoS One ; 17(1): e0261602, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35085257

RESUMO

BACKGROUND: On 1 February 2016, the World Health Organization declared Zika transmission a public health emergency of international concern. Monitoring and responding to community awareness, concern, and possible knowledge gaps are critical during public health emergencies. Here, we describe the review and analysis of micro-blogs posted on Sina-Weibo, China's largest social media platform, to develop and disseminate a Zika virus education campaign. METHODS: We used CYYUN Voice Express' Weibo Spider tool and the search terms of "Zhaika" OR "Zika" OR "Zikv" to capture microblogs about Zika virus retrospectively from February 1 to December 31, 2016 and prospectively from June 1 to November 15, 2017. We described microblogs meeting our inclusion criteria by month and Zika virus outbreaks in Asia and by source (e.g., government agency, individual, or other). We identified common misleading or inaccurate content authored by individual micro-bloggers (i.e., not supported by available scientific evidence) through a qualitative review. We used this information to develop and disseminate health awareness material about the Zika virus through China CDC's 12320 Health Hotline Weibo account. An online survey was conducted to obtain feedback on the material. RESULTS: We captured 15,888 microblogs meeting our inclusion criteria. Zika-related microblogs peaked in September 2016, corresponding to news reports about the Zika outbreak in Singapore (August to November 2016). Most microblogs (12,994 [82%]) were authored by individual users, followed by media agencies (842 [5%]), businesses (829 [5%]), international organizations (370 [2%]), and Chinese government agencies (235 [1%]). Relevant microblogs primarily focused on clinical symptoms and health risks, modes of transmission, and actions taken by individuals to prevent infection and seek health care. Incorrect and/or mis-leading information from individual users concentrated on modes of transmission and possible treatments. The microblog "#Zika is that far and this close" health campaign was posted on Sina-Weibo and Baidu (Internet search engine in China) on September 18, 2016. Younger respondents (p-value = 0.01), and those with at least a college education (p-value = 0.03), were more likely than other respondents to consider the online campaign reliable and trustworthy. CONCLUSION: Routine review of Sina-Weibo and other social media platforms could enhance the ability of public health staff to effectively respond to community concerns and awareness during public health emergencies. Advancements of social media monitoring tools and staff training could help to promote health awareness during emergencies by directly addressing public perceptions and concerns. Various approaches may be needed to reach different at-risk populations, particularly older and less educated populations who may prefer more traditional modes of communication.


Assuntos
Promoção da Saúde/métodos , Infecção por Zika virus/epidemiologia , Infecção por Zika virus/transmissão , Blogging , China/epidemiologia , Conhecimentos, Atitudes e Prática em Saúde , Humanos , Projetos Piloto , Saúde Pública , Pesquisa Qualitativa , Estudos Retrospectivos , Mídias Sociais
9.
J Gen Virol ; 103(1)2022 01.
Artigo em Inglês | MEDLINE | ID: mdl-35077341

RESUMO

Decades after its discovery in East Africa, Zika virus (ZIKV) emerged in Brazil in 2013 and infected millions of people during intense urban transmission. Whether vertebrates other than humans are involved in ZIKV transmission cycles remained unclear. Here, we investigate the role of different animals as ZIKV reservoirs by testing 1723 sera of pets, peri-domestic animals and African non-human primates (NHP) sampled during 2013-2018 in Brazil and 2006-2016 in Côte d'Ivoire. Exhaustive neutralization testing substantiated co-circulation of multiple flaviviruses and failed to confirm ZIKV infection in pets or peri-domestic animals in Côte d'Ivoire (n=259) and Brazil (n=1416). In contrast, ZIKV seroprevalence was 22.2% (2/9, 95% CI, 2.8-60.1) in West African chimpanzees (Pan troglodytes verus) and 11.1% (1/9, 95% CI, 0.3-48.3) in king colobus (Colobus polycomos). Our results indicate that while NHP may represent ZIKV reservoirs in Africa, pets or peri-domestic animals likely do not play a role in ZIKV transmission cycles.


Assuntos
Animais Domésticos/virologia , Primatas/virologia , Infecção por Zika virus/epidemiologia , Infecção por Zika virus/virologia , Zika virus , África , Animais , Brasil , Côte d'Ivoire , Humanos , Testes de Neutralização , Estudos Soroepidemiológicos , Infecção por Zika virus/transmissão
10.
Parasit Vectors ; 15(1): 36, 2022 Jan 24.
Artigo em Inglês | MEDLINE | ID: mdl-35073977

RESUMO

BACKGROUND: Aedes albopictus and Aedes japonicus, two invasive mosquito species in the United States, are implicated in the transmission of arboviruses. Studies have shown interactions of these two mosquito species with a variety of vertebrate hosts; however, regional differences exist and may influence their contribution to arbovirus transmission. METHODS: We investigated the distribution, abundance, host interactions, and West Nile virus infection prevalence of Ae. albopictus and Ae. japonicus by examining Pennsylvania mosquito and arbovirus surveillance data for the period between 2010 and 2018. Mosquitoes were primarily collected using gravid traps and BG-Sentinel traps, and sources of blood meals were determined by analyzing mitochondrial cytochrome b gene sequences amplified in PCR assays. RESULTS: A total of 10,878,727 female mosquitoes representing 51 species were collected in Pennsylvania over the 9-year study period, with Ae. albopictus and Ae. japonicus representing 4.06% and 3.02% of all collected mosquitoes, respectively. Aedes albopictus was distributed in 39 counties and Ae. japonicus in all 67 counties, and the abundance of these species increased between 2010 and 2018. Models suggested an increase in the spatial extent of Ae. albopictus during the study period, while that of Ae. japonicus remained unchanged. We found a differential association between the abundance of the two mosquito species and environmental conditions, percent development, and median household income. Of 110 Ae. albopictus and 97 Ae. japonicus blood meals successfully identified to species level, 98% and 100% were derived from mammalian hosts, respectively. Among 12 mammalian species, domestic cats, humans, and white-tailed deer served as the most frequent hosts for the two mosquito species. A limited number of Ae. albopictus acquired blood meals from avian hosts solely or in mixed blood meals. West Nile virus was detected in 31 pools (n = 3582 total number of pools) of Ae. albopictus and 12 pools (n = 977 total pools) of Ae. japonicus. CONCLUSIONS: Extensive distribution, high abundance, and frequent interactions with mammalian hosts suggest potential involvement of Ae. albopictus and Ae. japonicus in the transmission of human arboviruses including Cache Valley, Jamestown Canyon, La Crosse, dengue, chikungunya, and Zika should any of these viruses become prevalent in Pennsylvania. Limited interaction with avian hosts suggests that Ae. albopictus might occasionally be involved in transmission of arboviruses such as West Nile in the region.


Assuntos
Aedes , Infecções por Arbovirus/transmissão , Comportamento Alimentar , Mosquitos Vetores , Análise Espaço-Temporal , Aedes/fisiologia , Aedes/virologia , Animais , Arbovírus , Aves/virologia , Febre de Chikungunya/transmissão , Cervos/virologia , Reservatórios de Doenças/virologia , Humanos , Espécies Introduzidas , Mamíferos/virologia , Mosquitos Vetores/fisiologia , Mosquitos Vetores/virologia , Pennsylvania , Densidade Demográfica , Especificidade da Espécie , Vírus do Nilo Ocidental , Zika virus , Infecção por Zika virus/transmissão , Zoonoses/virologia
11.
Biochim Biophys Acta Mol Basis Dis ; 1868(1): 166270, 2022 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-34582966

RESUMO

Zika virus (ZIKV) infection has caused severe unexpected clinical outcomes in neonates and adults during the recent outbreak in Latin America, particularly in Brazil. Congenital malformations associated with ZIKV have been frequently reported; nevertheless, the mechanism of vertical transmission and the involvement of placental cells remains unclear. In this study, we applied quantitative proteomics analysis in a floating explant model of chorionic villi of human placental tissues incubated with ZIKV and with ZIKV pre-adsorbed with anti-ZIKV envelope protein. Proteomic data are available via ProteomeXchange with identifier PXD025764. Altered levels of proteins were involved in cell proliferation, apoptosis, inflammatory processes, and the integrin-cytoskeleton complex. Antibody-opsonized ZIKV particles differentially modulated the pattern of protein expression in placental cells; this phenomenon may play a pivotal role in determining the course of infection and the role of mixed infections. The expression of specific proteins was also evaluated by immunoperoxidase assays. These data fill gaps in our understanding of early events after ZIKV placental exposure and help identify infection control targets.


Assuntos
Placenta/metabolismo , Proteínas do Envelope Viral/genética , Infecção por Zika virus/genética , Zika virus/genética , Adulto , Apoptose/genética , Brasil/epidemiologia , Anormalidades Congênitas/epidemiologia , Anormalidades Congênitas/genética , Anormalidades Congênitas/virologia , Feminino , Humanos , Recém-Nascido , Transmissão Vertical de Doenças Infecciosas/prevenção & controle , Placenta/patologia , Placenta/virologia , Gravidez , Proteômica , Zika virus/patogenicidade , Infecção por Zika virus/epidemiologia , Infecção por Zika virus/transmissão , Infecção por Zika virus/virologia
12.
Arch Dis Child ; 107(3): 244-250, 2022 03.
Artigo em Inglês | MEDLINE | ID: mdl-34479857

RESUMO

OBJECTIVE: Zika virus (ZIKV) targets neural stem cells in the developing brain. However, the majority of ZIKV-exposed children are born without apparent neurological manifestations. It remains unclear if these children were protected from ZIKV neurotropism or if they harbour subtle pathology that is disruptive to brain development. We assess this by comparing neurodevelopmental outcomes in normocephalic ZIKV-exposed children relative to a parallel control group of unexposed controls. DESIGN: Cohort study. SETTING: Public health centres in Grenada, West Indies. PATIENTS: 384 mother-child pairs were enrolled during a period of active ZIKV transmission (April 2016-March 2017) and prospectively followed up to 30 months. Child exposure status was based on laboratory assessment of prenatal and postnatal maternal serum. MAIN OUTCOME MEASURES: The INTERGROWTH-21st Neurodevelopment Assessment (INTER-NDA) package and Cardiff Vision Tests, administered and scored by research staff masked to child's exposure status. RESULTS: A total of 131 normocephalic ZIKV exposed (n=68) and unexposed (n=63) children were assessed between 22 and 30 months of age. Approximately half of these children completed vision testing. There were no group differences in sociodemographics. Deficits in visual acuity (31%) and contrast sensitivity (23%) were apparent in the ZIKV-exposed infants in the absence of cognitive, motor, language or behavioural delays. CONCLUSIONS: Overall neurodevelopment is likely to be unaffected in ZIKV-exposed children with normal head circumference at birth and normal head growth in the first 2 years of life. However, the visual system may be selectively vulnerable, which indicates the need for vision testing by 3 years of age.


Assuntos
Encéfalo/crescimento & desenvolvimento , Transmissão Vertical de Doenças Infecciosas , Complicações Infecciosas na Gravidez/virologia , Efeitos Tardios da Exposição Pré-Natal/virologia , Infecção por Zika virus/transmissão , Adulto , Desenvolvimento Infantil , Pré-Escolar , Estudos de Coortes , Feminino , Humanos , Lactente , Masculino , Microcefalia/epidemiologia , Gravidez , Estudos Prospectivos , Índias Ocidentais , Zika virus
13.
J Med Entomol ; 59(1): 372-375, 2022 01 12.
Artigo em Inglês | MEDLINE | ID: mdl-34617566

RESUMO

Severe human arboviral diseases can be transmitted by the mosquito Aedes aegypti (Linnaeus), including dengue, chikungunya, Zika, and yellow fever. Adult control using spatial sprays with adulticides is recommended only when dengue outbreaks occur. In Argentina, mainly pyrethroids, like cis-permethrin, have been used as an adulticide, especially since 2008. The evolution and spread of resistance to insecticides is a major concern for vector control. This study reports for the first time pyrethroid resistance in Ae. aegypti adults from Argentina, in the city of Salvador Mazza (Salta). WHO discriminating doses of 0.75% were used for permethrin, 0.05% for deltamethrin, and 5% for malathion. Also the discriminating dose for cis-permethrin (0.6%) was calculated and evaluated for the first time. We found a resistance ratio 50 (RR50) of 10.3 (9.7-10.4) for cis-permethrin, which is considered as high resistance. Our results also indicated resistance to deltamethrin (22.6% mortality) and permethrin (53.6% mortality), and a total susceptibility to malathion (100% mortality). Results from this study highlight the importance of the correct use of insecticides within an Integrated Vector Management (IVM) approach and of early detection of resistance to enable Ae. aegypti control in Argentina. More studies are needed to determine the spread of mosquito resistance to pyrethroids.


Assuntos
Aedes/efeitos dos fármacos , Resistência a Inseticidas , Piretrinas/farmacologia , Animais , Infecções por Arbovirus/transmissão , Argentina/epidemiologia , Vetores de Doenças , Humanos , Inseticidas/farmacologia , Malation/farmacologia , Mosquitos Vetores/efeitos dos fármacos , Infecção por Zika virus/transmissão
14.
Biochim Biophys Acta Mol Basis Dis ; 1868(1): 166285, 2022 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-34624499

RESUMO

During pregnancy, a series of physiological changes are determined at the molecular, cellular and macroscopic level that make the mother and fetus more susceptible to certain viral and bacterial infections, especially the infections in this and the companion review. Particular situations increase susceptibility to infection in neonates. The enhanced susceptibility to certain infections increases the risk of developing particular diseases that can progress to become morbidly severe. For example, during the current pandemic caused by the SARS-CoV-2 virus, epidemiological studies have established that pregnant women with COVID-19 disease are more likely to be hospitalized. However, the risk for intensive care unit admission and mechanical ventilation is not increased compared with nonpregnant women. Although much remains unknown with this particular infection, the elevated risk of progression during pregnancy towards more severe manifestations of COVID-19 disease is not associated with an increased risk of death. In addition, the epidemiological data available in neonates suggest that their risk of acquiring COVID-19 is low compared with infants (<12 months of age). However, they might be at higher risk for progression to severe COVID-19 disease compared with older children. The data on clinical presentation and disease severity among neonates are limited and based on case reports and small case series. It is well documented the importance of the Zika virus infection as the main cause of several congenital anomalies and birth defects such as microcephaly, and also adverse pregnancy outcomes. Mycoplasma infections also increase adverse pregnancy outcomes. This review will focus on the molecular, pathophysiological and biophysical characteristics of the mother/placental-fetal/neonatal interactions and the possible mechanisms of these pathogens (SARS-CoV-2, ZIKV, and Mycoplasmas) for promoting disease at this level.


Assuntos
COVID-19/etiologia , COVID-19/transmissão , Infecções por Mycoplasma/etiologia , Infecções por Mycoplasma/transmissão , Complicações Infecciosas na Gravidez , Infecção por Zika virus/etiologia , Infecção por Zika virus/transmissão , Biomarcadores , Aleitamento Materno/efeitos adversos , Suscetibilidade a Doenças , Feminino , Interações Hospedeiro-Patógeno/imunologia , Humanos , Recém-Nascido , Transmissão Vertical de Doenças Infecciosas , Troca Materno-Fetal , Mycoplasma , Placenta/imunologia , Placenta/metabolismo , Placenta/microbiologia , Placenta/virologia , Gravidez , SARS-CoV-2 , Zika virus
15.
Am J Trop Med Hyg ; 106(2): 585-592, 2021 12 20.
Artigo em Inglês | MEDLINE | ID: mdl-34929668

RESUMO

Serological cross-reactivity has proved to be a challenge to diagnose Zika virus (ZIKV) infections in dengue virus (DENV) endemic countries. Confirmatory testing of ZIKV IgM positive results by plaque reduction neutralization tests (PRNTs) provides clarification in only a minority of cases because most individuals infected with ZIKV were previously exposed to DENV. The goal of this study was to evaluate the performance of a ZIKV/DENV DUO IgM antibody capture ELISA (MAC-ELISA) for discriminating between DENV and ZIKV infections in endemic regions. Our performance evaluation included acute and convalescent specimens from patients with real-time reverse transcription polymerase chain reaction (RT-PCR)-confirmed DENV or ZIKV from the Sentinel Enhanced Dengue Surveillance System in Ponce, Puerto Rico. The ZIKV/DENV DUO MAC-ELISA specificity was 100% for DENV (N = 127) and 98.4% for ZIKV (N = 275) when specimens were tested during the optimal testing window (days post-onset of illness [DPO] 6-120). The ZIKV/DENV DUO MAC-ELISA sensitivity of RT-PCR confirmed specimens reached 100% for DENV by DPO 6 and for ZIKV by DPO 9. Our new ZIKV/DENV DUO MAC-ELISA was also able to distinguish ZIKV and DENV regardless of previous DENV exposure. We conclude this novel serologic diagnostic assay can accurately discriminate ZIKV and DENV infections. This can potentially be useful considering that the more labor-intensive and expensive PRNT assay may not be an option for confirmatory diagnosis in areas that lack PRNT capacity, but experience circulation of both DENV and ZIKV.


Assuntos
Anticorpos Antivirais/imunologia , Dengue/diagnóstico , Ensaio de Imunoadsorção Enzimática/métodos , Imunoglobulina M/imunologia , Infecção por Zika virus/diagnóstico , Reações Cruzadas , Dengue/imunologia , Dengue/transmissão , Vírus da Dengue/imunologia , Doenças Endêmicas , Feminino , Humanos , Masculino , Testes Sorológicos/métodos , Proteínas não Estruturais Virais , Zika virus/imunologia , Infecção por Zika virus/imunologia , Infecção por Zika virus/transmissão
16.
mSphere ; 6(6): e0068721, 2021 12 22.
Artigo em Inglês | MEDLINE | ID: mdl-34878293

RESUMO

Arthropod-borne viruses comprise a significant global disease burden. Surveillance and mitigation of arboviruses like Zika virus (ZIKV) require accurate estimates of transmissibility by vector mosquitoes. Although Aedes species mosquitoes are established as competent ZIKV vectors, differences in experimental protocols across studies prevent direct comparisons of relative transmissibility. An understudied factor complicating these comparisons is differential environmental microbiota exposures, where most vector competence studies use mosquitoes reared in laboratory tap water, which does not represent the microbial complexity of environmental water where wild larvae develop. We simulated natural larval development by rearing Californian Aedes aegypti larvae with microbes obtained from cemetery headstone water compared to conventional tap water. A. aegypti larvae reared in environmental cemetery water pupated 3 days faster and at higher rates. Mosquitoes reared in environmental water were less competent vectors of ZIKV than laboratory water-reared A. aegypti, as evidenced by significantly reduced infection and transmission rates. Microbiome comparisons of laboratory water- and environment water-reared mosquitoes and their rearing water showed significantly higher bacterial diversity in environment water. Despite this pattern, corresponding differences in bacterial diversity were not consistently observed between the respective adult mosquitoes. We also observed that the microbial compositions of adult mosquitoes differed more by whether they ingested a bloodmeal than by larval water type. Together, these results highlight the role of transient microbes in the larval environment in modulating A. aegypti vector competence for ZIKV. Laboratory vector competence likely overestimates the true transmissibility of arboviruses like ZIKV when conventional laboratory water is used for rearing. IMPORTANCE We observed that A. aegypti mosquitoes reared in water from cemetery headstones instead of the laboratory tap exhibited a reduced capacity to become infected with and transmit Zika virus. Water from the environment contained more bacterial species than tap water, but these bacteria were not consistently detected in adult mosquitoes. Our results suggest that rearing mosquito larvae in water collected from local environments as opposed to laboratory tap water, as is conventional, could provide a more realistic assessment of ZIKV vector competence since it better recapitulates the natural environment in which larvae develop. Given that laboratory vector competence is used to define the species to target for control, the use of environmental water to rear larvae could better approximate the microbial exposures of wild mosquitoes, lessening the potential for overestimating ZIKV transmission risk. These studies raise the question of whether rearing larvae in natural water sources also reduces vector competence for other mosquito-borne viruses.


Assuntos
Aedes/virologia , Mosquitos Vetores/virologia , Infecção por Zika virus/transmissão , Zika virus/crescimento & desenvolvimento , Animais , Chlorocebus aethiops , Humanos , Larva/virologia , Saliva/virologia , Glândulas Salivares/virologia , Células Vero , Carga Viral , Água , Microbiologia da Água , Zika virus/isolamento & purificação
17.
Viruses ; 13(12)2021 11 25.
Artigo em Inglês | MEDLINE | ID: mdl-34960632

RESUMO

To date, no vaccines or antivirals are available against Zika virus (ZIKV). In addition, the mechanisms underlying ZIKV-associated pathogenesis of the central nervous system (CNS) are largely unexplored. Getting more insight into the cellular pathways that ZIKV recruits to facilitate infection of susceptible cells will be crucial for establishing an effective treatment strategy. In general, cells secrete a number of vesicles, known as extracellular vesicles (EVs), in response to viral infections. These EVs serve as intercellular communicators. Here, we investigated the role of EVs derived from ZIKV-infected human brain microvascular endothelial cells on the blood-brain barrier (BBB) system. We demonstrated that ZIKV-infected EVs (IEVs) can incorporate viral components, including ZIKV RNA, NS1, and E-protein, and further transfer them to several types of CNS cells. Using label-free impedance-based biosensing, we observed that ZIKV and IEVs can temporally disturb the monolayer integrity of BBB-mimicking cells, possibly by inducing structural rearrangements of the adherent protein VE-cadherin (immunofluorescence staining). Finally, differences in the lipidomic profile between EVs and their parental cells possibly suggest a preferential sorting mechanism of specific lipid species into the vesicles. To conclude, these data suggest that IEVs could be postulated as vehicles (Trojan horse) for ZIKV transmission via the BBB.


Assuntos
Barreira Hematoencefálica/metabolismo , Vesículas Extracelulares/metabolismo , Infecção por Zika virus/transmissão , Zika virus/fisiologia , Barreira Hematoencefálica/virologia , Células Cultivadas , Sistema Nervoso Central/virologia , Células Endoteliais/virologia , Vesículas Extracelulares/virologia , Humanos , Lipidômica , RNA Viral/metabolismo , Proteínas não Estruturais Virais/metabolismo , Proteínas Virais/genética , Proteínas Virais/metabolismo , Infecção por Zika virus/virologia
18.
Viruses ; 13(12)2021 12 14.
Artigo em Inglês | MEDLINE | ID: mdl-34960776

RESUMO

The global spread of invasive mosquito species increases arbovirus infections. In addition to the invasive species Aedes albopictus and Aedes japonicus, Aedes koreicus has spread within Central Europe. Extensive information on its vector competence is missing. Ae. koreicus from Germany were investigated for their vector competence for chikungunya virus (CHIKV), Zika virus (ZIKV) and West Nile virus (WNV). Experiments were performed under different climate conditions (27 ± 5 °C; 24 ± 5 °C) for fourteen days. Ae. koreicus had the potential to transmit CHIKV and ZIKV but not WNV. Transmission was exclusively observed at the higher temperature, and transmission efficiency was rather low, at 4.6% (CHIKV) or 4.7% (ZIKV). Using a whole virome analysis, a novel mosquito-associated virus, designated Wiesbaden virus (WBDV), was identified in Ae. koreicus. Linking the WBDV infection status of single specimens to their transmission capability for the arboviruses revealed no influence on ZIKV transmission. In contrast, a coinfection of WBDV and CHIKV likely has a boost effect on CHIKV transmission. Due to its current distribution, the risk of arbovirus transmission by Ae. koreicus in Europe is rather low but might gain importance, especially in regions with higher temperatures. The impact of WBDV on arbovirus transmission should be analyzed in more detail.


Assuntos
Aedes/virologia , Infecções por Arbovirus/transmissão , Mosquitos Vetores/virologia , Interferência Viral , Animais , Febre de Chikungunya/transmissão , Infecção por Zika virus/transmissão
19.
Front Immunol ; 12: 750365, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34745123

RESUMO

Zika virus (ZIKV) received worldwide attention over the past decade when outbreaks of the disease were found to be associated with severe neurological syndromes and congenital abnormalities. Unlike most other flaviviruses, ZIKV can spread through sexual and transplacental transmission, adding to the complexity of Zika pathogenesis and clinical outcomes. In addition, the spread of ZIKV in flavivirus-endemic regions, and the high degree of structural and sequence homology between Zika and its close cousin Dengue have raised questions on the interplay between ZIKV and the pre-existing immunity to other flaviviruses and the potential immunopathogenesis. The Zika epidemic peaked in 2016 and has affected over 80 countries worldwide. The re-emergence of large-scale outbreaks in the future is certainly a possibility. To date, there has been no approved antiviral or vaccine against the ZIKV. Therefore, continuing Zika research and developing an effective antiviral and vaccine is essential to prepare the world for a future Zika epidemic. For this purpose, an in-depth understanding of ZIKV interaction with many different pathways in the human host and how it exploits the host immune response is required. For successful infection, the virus has developed elaborate mechanisms to escape the host response, including blocking host interferon response and shutdown of certain host cell translation. This review provides a summary on the key host factors that facilitate ZIKV entry and replication and the mechanisms by which ZIKV antagonizes antiviral innate immune response and involvement of adaptive immune response leading to immunopathology. We also discuss how ZIKV modulates the host immune response during sexual transmission and pregnancy to induce infection, how the cross-reactive immunity from other flaviviruses impacts ZIKV infection, and provide an update on the current status of ZIKV vaccine development.


Assuntos
Infecção por Zika virus , Imunidade Adaptativa , Animais , Autoimunidade , Reações Cruzadas , Feminino , Síndrome de Guillain-Barré/etiologia , Síndrome de Guillain-Barré/imunologia , Interações Hospedeiro-Patógeno , Humanos , Imunidade Inata , Gravidez , Doenças Virais Sexualmente Transmissíveis/imunologia , Doenças Virais Sexualmente Transmissíveis/transmissão , Zika virus/fisiologia , Infecção por Zika virus/complicações , Infecção por Zika virus/imunologia , Infecção por Zika virus/transmissão
20.
PLoS Negl Trop Dis ; 15(11): e0009839, 2021 11.
Artigo em Inglês | MEDLINE | ID: mdl-34727099

RESUMO

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.


Assuntos
Aedes/virologia , Mosquitos Vetores/virologia , Zika virus/isolamento & purificação , Aedes/fisiologia , Animais , Brasil/epidemiologia , Doenças Endêmicas , Feminino , Humanos , Masculino , Mosquitos Vetores/fisiologia , Glândulas Salivares/virologia , Carga Viral , Zika virus/genética , Zika virus/fisiologia , Infecção por Zika virus/epidemiologia , Infecção por Zika virus/transmissão , Infecção por Zika virus/virologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...