Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 3.155
Filtrar
Mais filtros

Intervalo de ano de publicação
1.
PLoS Pathog ; 20(5): e1012010, 2024 May.
Artigo em Inglês | MEDLINE | ID: mdl-38753575

RESUMO

Arboviruses are a diverse group of insect-transmitted pathogens that pose global public health challenges. Identifying evolutionarily conserved host factors that combat arbovirus replication in disparate eukaryotic hosts is important as they may tip the balance between productive and abortive viral replication, and thus determine virus host range. Here, we exploit naturally abortive arbovirus infections that we identified in lepidopteran cells and use bacterial effector proteins to uncover host factors restricting arbovirus replication. Bacterial effectors are proteins secreted by pathogenic bacteria into eukaryotic hosts cells that can inhibit antimicrobial defenses. Since bacteria and viruses can encounter common host defenses, we hypothesized that some bacterial effectors may inhibit host factors that restrict arbovirus replication in lepidopteran cells. Thus, we used bacterial effectors as molecular tools to identify host factors that restrict four distinct arboviruses in lepidopteran cells. By screening 210 effectors encoded by seven different bacterial pathogens, we identify several effectors that individually rescue the replication of all four arboviruses. We show that these effectors encode diverse enzymatic activities that are required to break arbovirus restriction. We further characterize Shigella flexneri-encoded IpaH4 as an E3 ubiquitin ligase that directly ubiquitinates two evolutionarily conserved proteins, SHOC2 and PSMC1, promoting their degradation in insect and human cells. We show that depletion of either SHOC2 or PSMC1 in insect or human cells promotes arbovirus replication, indicating that these are ancient virus restriction factors conserved across invertebrate and vertebrate hosts. Collectively, our study reveals a novel pathogen-guided approach to identify conserved antimicrobial machinery, new effector functions, and conserved roles for SHOC2 and PSMC1 in virus restriction.


Assuntos
Proteínas de Bactérias , Interações Hospedeiro-Patógeno , Replicação Viral , Animais , Replicação Viral/fisiologia , Proteínas de Bactérias/metabolismo , Proteínas de Bactérias/genética , Humanos , Arbovírus , Shigella flexneri/patogenicidade , Infecções por Arbovirus/virologia , Linhagem Celular
2.
Immunity ; 44(6): 1455-69, 2016 06 21.
Artigo em Inglês | MEDLINE | ID: mdl-27332734

RESUMO

Aedes aegypti mosquitoes are responsible for transmitting many medically important viruses such as those that cause Zika and dengue. The inoculation of viruses into mosquito bite sites is an important and common stage of all mosquito-borne virus infections. We show, using Semliki Forest virus and Bunyamwera virus, that these viruses use this inflammatory niche to aid their replication and dissemination in vivo. Mosquito bites were characterized by an edema that retained virus at the inoculation site and an inflammatory influx of neutrophils that coordinated a localized innate immune program that inadvertently facilitated virus infection by encouraging the entry and infection of virus-permissive myeloid cells. Neutrophil depletion and therapeutic blockade of inflammasome activity suppressed inflammation and abrogated the ability of the bite to promote infection. This study identifies facets of mosquito bite inflammation that are important determinants of the subsequent systemic course and clinical outcome of virus infection.


Assuntos
Infecções por Arbovirus/imunologia , Vírus Bunyamwera/fisiologia , Inflamação/imunologia , Mordeduras e Picadas de Insetos/imunologia , Neutrófilos/imunologia , Vírus da Floresta de Semliki/fisiologia , Replicação Viral , Animais , Movimento Celular , Células Cultivadas , Culicidae/imunologia , Humanos , Imunidade Inata , Inflamassomos/metabolismo , Inflamação/virologia , Mordeduras e Picadas de Insetos/virologia , Camundongos , Neutrófilos/virologia
3.
Proc Natl Acad Sci U S A ; 119(24): e2114309119, 2022 06 14.
Artigo em Inglês | MEDLINE | ID: mdl-35675424

RESUMO

Viruses transmitted by Aedes mosquitoes are an increasingly important global cause of disease. Defining common determinants of host susceptibility to this large group of heterogenous pathogens is key for informing the rational design of panviral medicines. Infection of the vertebrate host with these viruses is enhanced by mosquito saliva, a complex mixture of salivary-gland-derived factors and microbiota. We show that the enhancement of infection by saliva was dependent on vascular function and was independent of most antisaliva immune responses, including salivary microbiota. Instead, the Aedes gene product sialokinin mediated the enhancement of virus infection through a rapid reduction in endothelial barrier integrity. Sialokinin is unique within the insect world as having a vertebrate-like tachykinin sequence and is absent from Anopheles mosquitoes, which are incompetent for most arthropod-borne viruses, whose saliva was not proviral and did not induce similar vascular permeability. Therapeutic strategies targeting sialokinin have the potential to limit disease severity following infection with Aedes-mosquito-borne viruses.


Assuntos
Aedes , Infecções por Arbovirus , Arbovírus , Saliva , Taquicininas , Viroses , Aedes/genética , Aedes/virologia , Animais , Infecções por Arbovirus/transmissão , Arbovírus/genética , Arbovírus/metabolismo , Saliva/virologia , Taquicininas/genética , Taquicininas/metabolismo , Viroses/transmissão
4.
Insect Mol Biol ; 33(4): 362-371, 2024 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-38450861

RESUMO

Multiple Wolbachia strains can block pathogen infection, replication and/or transmission in Aedes aegypti mosquitoes under both laboratory and field conditions. However, Wolbachia effects on pathogens can be highly variable across systems and the factors governing this variability are not well understood. It is increasingly clear that the mosquito host is not a passive player in which Wolbachia governs pathogen transmission phenotypes; rather, the genetics of the host can significantly modulate Wolbachia-mediated pathogen blocking. Specifically, previous work linked variation in Wolbachia pathogen blocking to polymorphisms in the mosquito alpha-mannosidase-2 (αMan2) gene. Here we use CRISPR-Cas9 mutagenesis to functionally test this association. We developed αMan2 knockouts and examined effects on both Wolbachia and virus levels, using dengue virus (DENV; Flaviviridae) and Mayaro virus (MAYV; Togaviridae). Wolbachia titres were significantly elevated in αMan2 knockout (KO) mosquitoes, but there were complex interactions with virus infection and replication. In Wolbachia-uninfected mosquitoes, the αMan2 KO mutation was associated with decreased DENV titres, but in a Wolbachia-infected background, the αMan2 KO mutation significantly increased virus titres. In contrast, the αMan2 KO mutation significantly increased MAYV replication in Wolbachia-uninfected mosquitoes and did not affect Wolbachia-mediated virus blocking. These results demonstrate that αMan2 modulates arbovirus infection in A. aegypti mosquitoes in a pathogen- and Wolbachia-specific manner, and that Wolbachia-mediated pathogen blocking is a complex phenotype dependent on the mosquito host genotype and the pathogen. These results have a significant impact for the design and use of Wolbachia-based strategies to control vector-borne pathogens.


Assuntos
Aedes , Wolbachia , alfa-Manosidase , Animais , Aedes/microbiologia , Aedes/virologia , Aedes/genética , Wolbachia/fisiologia , alfa-Manosidase/metabolismo , alfa-Manosidase/genética , Vírus da Dengue/fisiologia , Arbovírus/fisiologia , Mosquitos Vetores/microbiologia , Mosquitos Vetores/virologia , Mosquitos Vetores/genética , Feminino , Infecções por Arbovirus/transmissão , Proteínas de Insetos/metabolismo , Proteínas de Insetos/genética , Sistemas CRISPR-Cas
5.
Nature ; 563(7732): 501-507, 2018 11.
Artigo em Inglês | MEDLINE | ID: mdl-30429615

RESUMO

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


Assuntos
Aedes/genética , Infecções por Arbovirus/virologia , Arbovírus , Genoma de Inseto/genética , Genômica/normas , Controle de Insetos , Mosquitos Vetores/genética , Mosquitos Vetores/virologia , Aedes/virologia , Animais , Infecções por Arbovirus/transmissão , Arbovírus/isolamento & purificação , Variações do Número de Cópias de DNA/genética , Vírus da Dengue/isolamento & purificação , Feminino , Variação Genética/genética , Genética Populacional , Glutationa Transferase/genética , Resistência a Inseticidas/efeitos dos fármacos , Masculino , Anotação de Sequência Molecular , Família Multigênica/genética , Piretrinas/farmacologia , Padrões de Referência , Processos de Determinação Sexual/genética
6.
Proc Natl Acad Sci U S A ; 118(41)2021 10 12.
Artigo em Inglês | MEDLINE | ID: mdl-34607949

RESUMO

Releasing sterile or incompatible male insects is a proven method of population management in agricultural systems with the potential to revolutionize mosquito control. Through a collaborative venture with the "Debug" Verily Life Sciences team, we assessed the incompatible insect technique (IIT) with the mosquito vector Aedes aegypti in northern Australia in a replicated treatment control field trial. Backcrossing a US strain of Ae. aegypti carrying Wolbachia wAlbB from Aedes albopictus with a local strain, we generated a wAlbB2-F4 strain incompatible with both the wild-type (no Wolbachia) and wMel-Wolbachia Ae. aegypti now extant in North Queensland. The wAlbB2-F4 strain was manually mass reared with males separated from females using Verily sex-sorting technologies to obtain no detectable female contamination in the field. With community consent, we delivered a total of three million IIT males into three isolated landscapes of over 200 houses each, releasing ∼50 males per house three times a week over 20 wk. Detecting initial overflooding ratios of between 5:1 and 10:1, strong population declines well beyond 80% were detected across all treatment landscapes when compared to controls. Monitoring through the following season to observe the ongoing effect saw one treatment landscape devoid of adult Ae. aegypti early in the season. A second landscape showed reduced adults, and the third recovered fully. These encouraging results in suppressing both wild-type and wMel-Ae. aegypti confirms the utility of bidirectional incompatibility in the field setting, show the IIT to be robust, and indicate that the removal of this arbovirus vector from human-occupied landscapes may be achievable.


Assuntos
Aedes/microbiologia , Infecções por Arbovirus/prevenção & controle , Infertilidade Masculina , Controle de Mosquitos/métodos , Wolbachia/metabolismo , Aedes/fisiologia , Animais , Infecções por Arbovirus/transmissão , Arbovírus , Austrália , Agentes de Controle Biológico , Feminino , Humanos , Masculino , Mosquitos Vetores/microbiologia , Queensland
7.
Mem Inst Oswaldo Cruz ; 119: e240015, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38922217

RESUMO

The coinfection between malaria (ML) and arboviral diseases represents a major global public health problem, particularly in tropical and subtropical countries. Despite its relevance, this topic is still insufficiently discussed in the current literature. Here, we aimed to investigate the worldwide distribution, symptoms, and diagnosis during coinfection between ML and arboviral diseases. We conducted a systematic review following the Preferred reporting items for systematic reviews and meta-analyses (PRISMA) statement and assessed the selection and eligibility criteria, created and diagrammed maps, and analysed major symptoms with 95% confidence intervals (CI) using prevalence ratio and effect size, also performing latent class analysis. A total of 85,485 studies were retrieved, of which 56 were included: 57.14% in Asia, 25% in Africa, 14.30% in South America, and 3.56% in Europe. A total of 746 individuals were reported to be coinfected with Plasmodium and arbovirus. Concurrent ML, Dengue (DEN), Chikungunya (CHIK), and Zika (ZIK) patients are more likely to present headache and skin rash. Regarding diagnosis, 58,253 were made, of which 38,176 were positive (ML and at least one arboviral disease). The magnitude of these pathogens' coexistence points out the pressing need for improvements in public health policies towards diagnosis and prevention of both diseases, especially in endemic areas.


Assuntos
Infecções por Arbovirus , Coinfecção , Malária , Humanos , Coinfecção/epidemiologia , Malária/epidemiologia , Malária/complicações , Malária/diagnóstico , Infecções por Arbovirus/epidemiologia , Infecções por Arbovirus/diagnóstico , Saúde Global , Prevalência
8.
Euro Surveill ; 29(20)2024 05.
Artigo em Inglês | MEDLINE | ID: mdl-38757289

RESUMO

Aedes albopictus collected in 2023 in the greater Paris area (Île-de-France) were experimentally able to transmit five arboviruses: West Nile virus from 3 days post-infection (dpi), chikungunya virus and Usutu virus from 7 dpi, dengue virus and Zika virus from 21 dpi. Given the growing number of imported dengue cases reported in early 2024 in France, surveillance of Ae. albopictus should be reinforced during the Paris Olympic Games in July, when many international visitors including from endemic countries are expected.


Assuntos
Aedes , Vírus Chikungunya , Vírus da Dengue , Zika virus , Animais , Aedes/virologia , Humanos , Zika virus/isolamento & purificação , Vírus da Dengue/isolamento & purificação , Vírus Chikungunya/isolamento & purificação , Paris , Mosquitos Vetores/virologia , Vírus do Nilo Ocidental/isolamento & purificação , Arbovírus/isolamento & purificação , Infecções por Arbovirus/transmissão , Flavivirus/isolamento & purificação , França , Dengue/transmissão , Dengue/epidemiologia , Infecção por Zika virus/transmissão
9.
Clin Infect Dis ; 76(6): 1142-1148, 2023 03 21.
Artigo em Inglês | MEDLINE | ID: mdl-36103602

RESUMO

With increasing use of rituximab and other B-cell depleting monoclonal antibodies for multiple indications, infectious complications are being recognized. We summarize clinical findings of patients on rituximab with arboviral diseases identified through literature review or consultation with the Centers for Disease Control and Prevention. We identified 21 patients on recent rituximab therapy who were diagnosed with an arboviral disease caused by West Nile, tick-borne encephalitis, eastern equine encephalitis, Cache Valley, Jamestown Canyon, and Powassan viruses. All reported patients had neuroinvasive disease. The diagnosis of arboviral infection required molecular testing in 20 (95%) patients. Median illness duration was 36 days (range, 12 days to 1 year), and 15/19 (79%) patients died from their illness. Patients on rituximab with arboviral disease can have a severe or prolonged course with an absence of serologic response. Patients should be counseled about mosquito and tick bite prevention when receiving rituximab and other B-cell depleting therapies.


Assuntos
Infecções por Arbovirus , Encefalite Transmitida por Carrapatos , Febre do Nilo Ocidental , Animais , Rituximab/uso terapêutico , Febre do Nilo Ocidental/tratamento farmacológico , Febre do Nilo Ocidental/complicações , Febre do Nilo Ocidental/epidemiologia , Surtos de Doenças , Encefalite Transmitida por Carrapatos/epidemiologia
10.
PLoS Pathog ; 17(9): e1009870, 2021 09.
Artigo em Inglês | MEDLINE | ID: mdl-34473801

RESUMO

As mosquito females require a blood meal to reproduce, they can act as vectors of numerous pathogens, such as arboviruses (e.g. Zika, dengue and chikungunya viruses), which constitute a substantial worldwide public health burden. In addition to blood meals, mosquito females can also take sugar meals to get carbohydrates for their energy reserves. It is now recognised that diet is a key regulator of health and disease outcome through interactions with the immune system. However, this has been mostly studied in humans and model organisms. So far, the impact of sugar feeding on mosquito immunity and in turn, how this could affect vector competence for arboviruses has not been explored. Here, we show that sugar feeding increases and maintains antiviral immunity in the digestive tract of the main arbovirus vector Aedes aegypti. Our data demonstrate that the gut microbiota does not mediate the sugar-induced immunity but partly inhibits it. Importantly, sugar intake prior to an arbovirus-infected blood meal further protects females against infection with arboviruses from different families. Sugar feeding blocks arbovirus initial infection and dissemination from the gut and lowers infection prevalence and intensity, thereby decreasing the transmission potential of female mosquitoes. Finally, we show that the antiviral role of sugar is mediated by sugar-induced immunity. Overall, our findings uncover a crucial role of sugar feeding in mosquito antiviral immunity which in turn decreases vector competence for arboviruses. Since Ae. aegypti almost exclusively feed on blood in some natural settings, our findings suggest that this lack of sugar intake could increase the spread of mosquito-borne arboviral diseases.


Assuntos
Aedes/virologia , Infecções por Arbovirus , Dieta , Insetos Vetores/virologia , Intestinos/imunologia , Aedes/imunologia , Animais , Arbovírus , Insetos Vetores/imunologia , Açúcares
11.
MMWR Morb Mortal Wkly Rep ; 72(34): 901-906, 2023 Aug 25.
Artigo em Inglês | MEDLINE | ID: mdl-37616182

RESUMO

Arthropod-borne viruses (arboviruses) are transmitted to humans primarily through the bites of infected mosquitoes or ticks, and in the continental United States, West Nile virus (WNV) is the leading cause of domestically acquired arboviral disease. Other arboviruses cause sporadic cases of disease as well as occasional outbreaks. This report summarizes 2021 surveillance data reported to CDC by U.S. jurisdictions for nationally notifiable arboviruses; the report excludes chikungunya, dengue, yellow fever, and Zika virus disease cases, because these infections were acquired primarily through travel during 2021. Forty-nine states and the District of Columbia reported 3,035 cases of domestic arboviral disease, including those caused by West Nile (2,911), La Crosse (40), Jamestown Canyon (32), Powassan (24), St. Louis encephalitis (17), unspecified California serogroup (six), and eastern equine encephalitis (five) viruses. Among the WNV disease cases, 2,008 (69%) were classified as neuroinvasive disease, for a national incidence of 0.61 cases per 100,000 population. Because arboviral diseases continue to cause serious illness, maintaining surveillance programs to monitor their transmission and prevalence is important to the direction and promotion of prevention activities. Health care providers should consider arboviral infections in the differential diagnosis of aseptic meningitis and encephalitis, obtain appropriate specimens for laboratory testing, and promptly report cases to public health authorities. Prevention depends on community and household efforts to reduce vector populations and personal protective measures to prevent mosquito and tick bites, such as use of Environmental Protection Agency-registered insect repellent and wearing protective clothing.


Assuntos
Infecções por Arbovirus , Culicidae , Vírus do Nilo Ocidental , Febre Amarela , Infecção por Zika virus , Zika virus , Cavalos , Animais , Humanos , Mosquitos Vetores , Infecções por Arbovirus/epidemiologia , District of Columbia
12.
Curr Opin Pediatr ; 35(2): 155-165, 2023 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-36801979

RESUMO

PURPOSE OF REVIEW: Dengue, chikungunya and zika have caused significant epidemics in the Caribbean in recent years. This review highlights their impact in Caribbean children. RECENT FINDINGS: Dengue has been increasingly intense and severe, seroprevalence is 80-100% in the Caribbean, children have increased attributable morbidity and mortality. Severe dengue, especially dengue with haemorrhage was significantly associated with haemoglobin SC disease and multiple organ-systems involved. These included the gastrointestinal and haematologic systems with extremely high lactate dehydrogenases and creatinine phosphokinases and severely abnormal bleeding indices. Despite appropriate interventions, mortality was highest within the first 48 h of admission. Chikungunya, a togavirus, affected 80% of some Caribbean populations. Paediatric presentations included high fever, skin, joint and neurological manifestations. Children less than 5 years of age had the highest morbidity and mortality. This maiden chikungunya epidemic was explosive and overwhelmed public health systems. Zika, another flavivirus, has a seroprevalence of 15% in pregnancy, so the Caribbean remains susceptible. Paediatric complications include pregnancy losses, stillbirths, Congenital Zika syndrome, Guillain-Barre syndrome, acute disseminated encephalomyelitis and transverse myelitis. Neurodevelopment stimulation programs for zika-exposed infants have been effective in improving language and positive behaviour scores. SUMMARY: Caribbean children remain at risk for dengue, chikungunya and zika, with high attributable morbidity and mortality.


Assuntos
Infecções por Arbovirus , Febre de Chikungunya , Dengue , Infecção por Zika virus , Zika virus , Criança , Humanos , Infecção por Zika virus/complicações , Infecção por Zika virus/diagnóstico , Infecção por Zika virus/epidemiologia , Febre de Chikungunya/complicações , Febre de Chikungunya/diagnóstico , Febre de Chikungunya/epidemiologia , Dengue/complicações , Dengue/diagnóstico , Dengue/epidemiologia , Estudos Soroepidemiológicos , Infecções por Arbovirus/diagnóstico , Infecções por Arbovirus/epidemiologia , Infecções por Arbovirus/complicações , Região do Caribe/epidemiologia
13.
Epidemiol Infect ; 151: e181, 2023 10 12.
Artigo em Inglês | MEDLINE | ID: mdl-37823310

RESUMO

Dengue, chikungunya, and Zika are arboviruses that cause 390 million infections annually. Risk factors for hospitalization are poorly understood. Communities affected by these diseases have an escalating prevalence of allergies and obesity, which are linked to immune dysfunction. We assessed the association of allergies or body mass with hospitalization for an arbovirus infection. From 2014 to 2017, we recruited participants with a clinical diagnosis of arbovirus infection. Arbovirus infections were laboratory-confirmed and allergies were self-reported. Mid-upper arm circumference (MUAC), weight, and height were measured. We used two logistic regression models to assess the relationships between hospitalization and allergies and between hospitalization and body mass (MUAC for participants <20 years old and body mass index (BMI) for adults ≥20 years old). Models were stratified by age group and adjusted for confounders. For allergies, 41 of 265 were hospitalized. There was no association between allergies and hospitalization. For body mass, 34 of 251 were hospitalized. There was a 43% decrease in hospitalization odds for each additional centimetre MUAC among children (aOR 0.566, 95% CI 0.252-1.019) and a 12% decrease in hospitalization odds for each additional BMI unit among adults (aOR 0.877, 95% CI 0.752-0.998). Our work encourages the exploration of the underlying mechanisms.


Assuntos
Infecções por Arbovirus , Hipersensibilidade , Infecção por Zika virus , Zika virus , Adulto , Criança , Humanos , Adulto Jovem , Estudos Prospectivos , Equador/epidemiologia , Índice de Massa Corporal , Hospitalização
14.
BMC Infect Dis ; 23(1): 708, 2023 Oct 20.
Artigo em Inglês | MEDLINE | ID: mdl-37864153

RESUMO

BACKGROUND: Aedes (Stegomyia)-borne diseases are an expanding global threat, but gaps in surveillance make comprehensive and comparable risk assessments challenging. Geostatistical models combine data from multiple locations and use links with environmental and socioeconomic factors to make predictive risk maps. Here we systematically review past approaches to map risk for different Aedes-borne arboviruses from local to global scales, identifying differences and similarities in the data types, covariates, and modelling approaches used. METHODS: We searched on-line databases for predictive risk mapping studies for dengue, Zika, chikungunya, and yellow fever with no geographical or date restrictions. We included studies that needed to parameterise or fit their model to real-world epidemiological data and make predictions to new spatial locations of some measure of population-level risk of viral transmission (e.g. incidence, occurrence, suitability, etc.). RESULTS: We found a growing number of arbovirus risk mapping studies across all endemic regions and arboviral diseases, with a total of 176 papers published 2002-2022 with the largest increases shortly following major epidemics. Three dominant use cases emerged: (i) global maps to identify limits of transmission, estimate burden and assess impacts of future global change, (ii) regional models used to predict the spread of major epidemics between countries and (iii) national and sub-national models that use local datasets to better understand transmission dynamics to improve outbreak detection and response. Temperature and rainfall were the most popular choice of covariates (included in 50% and 40% of studies respectively) but variables such as human mobility are increasingly being included. Surprisingly, few studies (22%, 31/144) robustly tested combinations of covariates from different domains (e.g. climatic, sociodemographic, ecological, etc.) and only 49% of studies assessed predictive performance via out-of-sample validation procedures. CONCLUSIONS: Here we show that approaches to map risk for different arboviruses have diversified in response to changing use cases, epidemiology and data availability. We identify key differences in mapping approaches between different arboviral diseases, discuss future research needs and outline specific recommendations for future arbovirus mapping.


Assuntos
Aedes , Infecções por Arbovirus , Arbovírus , Febre de Chikungunya , Dengue , Febre Amarela , Infecção por Zika virus , Zika virus , Animais , Humanos , Infecções por Arbovirus/epidemiologia , Febre Amarela/epidemiologia , Mosquitos Vetores , Dengue/epidemiologia
15.
BMC Infect Dis ; 23(1): 371, 2023 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-37264307

RESUMO

BACKGROUND: Dengue, Chikungunya, and Zika are co-endemic in Honduras and are often misdiagnosed due to similar clinical and epidemiological behavior. Most arboviral infections reported in primary care are based on clinical diagnoses without laboratory confirmation. Therefore, the accuracy of physicians' diagnoses and the factors that affect them needs to be evaluated. METHODS: A cross-sectional study with convenience sampling at primary healthcare centers was conducted from June to September 2016 and 2017. Clinical data and dried blood spots on Whatman 903 filter paper from 415 arboviral cases and 248 non-arboviral febrile cases were collected. Viral RNA was extracted from a 6-mm DBS paper disc and confirmed by RT-qPCR and sequencing. RESULTS: Only 30.84% of diagnostic accuracy was observed in physicians in primary care when comparing arboviral clinical diagnosis with RT-qPCR detection. Moreover, in Dengue and Zika clinical cases, only 8.23% and 27.08% were RT-qPCR confirmed, respectively. No Chikungunya cases were confirmed. In 2017, 20.96% of febrile cases were RT-qPCR confirmed arboviral infections. The symptoms of 45.5% of arboviral cases can fit more than one case definition for arboviruses. The "symptom compliance" and "patient with suspected close contact" were the criteria most utilized by physicians for arboviral diagnosis. The pattern of the epidemiological curves of the arboviral clinical cases didn't match the one of the RT-qPCR confirmed cases. CONCLUSIONS: Low diagnostic accuracy for overall and individual arboviral infections was observed in physicians. Unspecific symptomatology, overlapping case definitions, and reported close contact to an arboviral patient might contribute to misdiagnosis. Without laboratory confirmation, surveillance data may not reflect the real behavior of these diseases and could impact health interventions.


Assuntos
Infecções por Arbovirus , Arbovírus , Febre de Chikungunya , Dengue , Infecção por Zika virus , Zika virus , Humanos , Dengue/diagnóstico , Dengue/epidemiologia , Honduras/epidemiologia , Estudos Transversais , Infecção por Zika virus/diagnóstico , Infecção por Zika virus/epidemiologia , Febre de Chikungunya/diagnóstico , Febre de Chikungunya/epidemiologia , Zika virus/genética , Infecções por Arbovirus/epidemiologia , Febre/etiologia , Atenção Primária à Saúde
16.
Postgrad Med J ; 99(1174): 826-833, 2023 Jul 21.
Artigo em Inglês | MEDLINE | ID: mdl-37130817

RESUMO

The rapid spread of arboviral infections in recent years has continually established arthropod-borne encephalitis to be a pressing global health concern. Causing a wide range of clinical presentations ranging from asymptomatic infection to fulminant neurological disease, the hallmark features of arboviral infection are important to clinically recognise. Arboviral infections may cause severe neurological presentations such as meningoencephalitis, epilepsy, acute flaccid paralysis and stroke. While the pathogenesis of arboviral infections is still being investigated, shared neuroanatomical pathways among these viruses may give insight into future therapeutic targets. The shifting infection transmission patterns and evolving distribution of arboviral vectors are heavily influenced by global climate change and human environmental disruption, therefore it is of utmost importance to consider this potential aetiology when assessing patients with encephalitic presentations.


Assuntos
Infecções por Arbovirus , Arbovírus , Encefalite por Arbovirus , Humanos
17.
J Gen Virol ; 103(2)2022 02.
Artigo em Inglês | MEDLINE | ID: mdl-35191823

RESUMO

Arboviruses are medically important arthropod-borne viruses that cause a range of diseases in humans from febrile illness to arthritis, encephalitis and hemorrhagic fever. Given their transmission cycles, these viruses face the challenge of replicating in evolutionarily divergent organisms that can include ticks, flies, mosquitoes, birds, rodents, reptiles and primates. Furthermore, their cell attachment receptor utilization may be affected by the opposing needs for generating high and sustained serum viremia in vertebrates such that virus particles are efficiently collected during a hematophagous arthropod blood meal but they must also bind sufficiently to cellular structures on divergent organisms such that productive infection can be initiated and viremia generated. Sulfated polysaccharides of the glycosaminoglycan (GAG) groups, primarily heparan sulfate (HS), have been identified as cell attachment moieties for many arboviruses. Original identification of GAG binding as a phenotype of arboviruses appeared to involve this attribute arising solely as a consequence of adaptation of virus isolates to growth in cell culture. However, more recently, naturally circulating strains of at least one arbovirus, eastern equine encephalitis, have been shown to bind HS efficiently and the GAG binding phenotype continues to be associated with arbovirus infection in published studies. If GAGs are attachment receptors for many naturally circulating arboviruses, this could lead to development of broad-spectrum antiviral therapies through blocking of the virus-GAG interaction. This review summarizes the available data for GAG/HS binding as a phenotype of naturally circulating arbovirus strains emphasizing the importance of avoiding tissue culture amplification and artifactual phenotypes during their isolation.


Assuntos
Infecções por Arbovirus/virologia , Arbovírus/imunologia , Heparitina Sulfato/imunologia , Animais , Humanos
18.
PLoS Pathog ; 16(12): e1009134, 2020 12.
Artigo em Inglês | MEDLINE | ID: mdl-33351855

RESUMO

Mosquitoes are responsible for the transmission of many clinically important arboviruses that cause significant levels of annual mortality and socioeconomic health burden worldwide. Deciphering the mechanisms by which mosquitoes modulate arbovirus infection is crucial to understand how viral-host interactions promote vector transmission and human disease. SUMOylation is a post-translational modification that leads to the covalent attachment of the Small Ubiquitin-like MOdifier (SUMO) protein to host factors, which in turn can modulate their stability, interaction networks, sub-cellular localisation, and biochemical function. While the SUMOylation pathway is known to play a key role in the regulation of host immune defences to virus infection in humans, the importance of this pathway during arbovirus infection in mosquito vectors, such as Aedes aegypti (Ae. aegypti), remains unknown. Here we characterise the sequence, structure, biochemical properties, and tissue-specific expression profiles of component proteins of the Ae. aegypti SUMOylation pathway. We demonstrate significant biochemical differences between Ae. aegypti and Homo sapiens SUMOylation pathways and identify cell-type specific patterns of SUMO expression in Ae. aegypti tissues known to support arbovirus replication. Importantly, depletion of core SUMOylation effector proteins (SUMO, Ubc9 and PIAS) in Ae. aegypti cells led to enhanced levels of arbovirus replication from three different families; Zika (Flaviviridae), Semliki Forest (Togaviridae), and Bunyamwera (Bunyaviridae) viruses. Our findings identify an important role for mosquito SUMOylation in the cellular restriction of arboviruses that may directly influence vector competence and transmission of clinically important arboviruses.


Assuntos
Aedes/virologia , Arbovírus/fisiologia , Interações Hospedeiro-Patógeno/fisiologia , Mosquitos Vetores/virologia , Replicação Viral/fisiologia , Animais , Infecções por Arbovirus/transmissão , Humanos , Sumoilação
19.
MMWR Morb Mortal Wkly Rep ; 71(18): 628-632, 2022 May 06.
Artigo em Inglês | MEDLINE | ID: mdl-35511710

RESUMO

Arthropod-borne viruses (arboviruses) are transmitted to humans primarily through the bite of infected mosquitoes and ticks. West Nile virus (WNV), mainly transmitted by Culex species mosquitos, is the leading cause of domestically acquired arboviral disease in the United States (1). Other arboviruses cause sporadic cases of disease and occasional outbreaks. This report summarizes passive data for nationally notifiable domestic arboviruses in the United States reported to CDC for 2020. Forty-four states reported 884 cases of domestic arboviral disease, including those caused by West Nile (731), La Crosse (88), Powassan (21), St. Louis encephalitis (16), eastern equine encephalitis (13), Jamestown Canyon (13), and unspecified California serogroup (2) viruses. A total of 559 cases of neuroinvasive WNV disease were reported, for a national incidence of 0.17 cases per 100,000 population. Because arboviral diseases continue to cause serious illness and the locations of outbreaks vary annually, health care providers should consider arboviral infections in patients with aseptic meningitis or encephalitis that occur during periods when ticks and mosquitoes are active, perform recommended diagnostic testing, and promptly report cases to public health authorities to guide prevention strategies and messaging.


Assuntos
Infecções por Arbovirus , Culicidae , Febre do Nilo Ocidental , Vírus do Nilo Ocidental , Animais , Infecções por Arbovirus/epidemiologia , Surtos de Doenças , Humanos , Vigilância da População , Estados Unidos/epidemiologia , Febre do Nilo Ocidental/epidemiologia
20.
Arch Virol ; 167(11): 2151-2162, 2022 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-35841448

RESUMO

Arthropod-borne viruses (arboviruses) are a significant public health threat, especially in tropical and subtropical regions. More than 150 arboviruses can cause febrile illness following infection in humans. The Brazilian Amazon region has the highest number of arboviruses detected worldwide. In addition to arboviruses, malaria, caused by Plasmodium vivax, is endemic in the Amazon. Patients with malaria and arboviral disease frequently show similar clinical presentation and laboratory findings, making the diagnosis of the cause of the infection challenging. The aim of this study was to evaluate the potential for viral infections in patients with suspected malaria but without Plasmodium infection in the Brazilian Amazon. We recruited 200 subjects with suspected malaria in Manaus, Brazil. First, we tested for arboviruses in serum samples from 124 of the 200 participants using an arbovirus DNA microarray platform, which did not detect any virus. Then, we mixed the serum samples of the other 76 participants in 10 pools and subjected them to next-generation sequencing. Analysis of the sequencing data revealed the presence of only one arbovirus (Zika virus) in one sample pool. This analysis also detected the presence of primate erythroparvovirus 1 and pegivirus C. These results suggest that arboviruses are not the most frequent viral infections in patients with suspected malaria but without Plasmodium infection in the metropolitan region of Manaus. Implementation of specific viral surveillance tests will help in the early detection of viruses with epidemic potential.


Assuntos
Infecções por Arbovirus , Arbovírus , Malária , Infecção por Zika virus , Zika virus , Animais , Infecções por Arbovirus/diagnóstico , Infecções por Arbovirus/epidemiologia , Arbovírus/genética , Brasil/epidemiologia , Febre , Humanos , Malária/diagnóstico , Malária/epidemiologia , Zika virus/genética
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa