Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 8.351
Filtrer
1.
Article de Anglais | MEDLINE | ID: mdl-38594795

RÉSUMÉ

Abstract: Timor-Leste is a mountainous, half-island nation with a population of 1.3 million, which shares a land border with Indonesia and is 550 km from Darwin, Australia. Since independence in 2002, Timor-Leste has achieved significant development; however, high levels of poverty remain. Chikungunya virus (CHIKV) is endemic in over 100 countries in Africa, Asia, Europe and in the Americas. It is transmitted by the bite of infected Aedes aegypti or Ae. albopictus mosquitoes, which are present in Timor-Leste and which contribute to annual rainy-season dengue virus (DENV) outbreaks. Symptomatic people typically suffer from acute onset of fever, usually accompanied by severe arthritis or arthralgia. Joint pain can be debilitating for several days, and may sometimes last for weeks, months or years. Unlike DENV infection which has significant mortality, most people recover completely. Between 2002 and 2023, there were 26 cases of CHIKV notified in Australia who acquired their infection in Timor-Leste; however, laboratory testing capability for CHIKV in Timor-Leste only became available in 2021 using polymerase chain reaction (PCR). The first locally diagnosed case was notified in November 2023. In January 2024, an outbreak of CHIKV was recognised in Timor-Leste for the first time, with 195 outbreak cases reported during 1-31 January 2024; all were PCR positive. There were no cases hospitalised, and no deaths. The median age of cases was 17 years (range 1-76 years); 51% were males. Cases were reported across the country; most (88/195) were from Dili, although the highest incidence was seen in the neighbouring municipality of Ermera (monthly incidence rate of 58.8 cases per 100,000 population). This first reported outbreak of CHIKV in Timor-Leste highlights the need for improved mosquito-borne illness control and response strategies, including minimising breeding sites and promoting early presentation for treatment and differential diagnosis from DENV, and consideration of the deployment of Wolbachia-infected mosquitoes, particularly as they have shown to reduce the transmission of CHIKV, DENV and Zika virus, all of which pose threats in Timor-Leste.


Sujet(s)
Fièvre chikungunya , Virus du chikungunya , Infection par le virus Zika , Virus Zika , Mâle , Animaux , Humains , Nourrisson , Enfant d'âge préscolaire , Enfant , Adolescent , Jeune adulte , Adulte , Adulte d'âge moyen , Sujet âgé , Femelle , Fièvre chikungunya/épidémiologie , Timor oriental/épidémiologie , Australie/épidémiologie , Virus du chikungunya/génétique , Épidémies de maladies , Infection par le virus Zika/épidémiologie , Infection par le virus Zika/prévention et contrôle
2.
Med Educ Online ; 29(1): 2336331, 2024 Dec 31.
Article de Anglais | MEDLINE | ID: mdl-38577972

RÉSUMÉ

PURPOSE: Medical school educators face challenges determining which new and emerging topics to incorporate into medical school curricula, and how to do so. A study was conducted to gain a better understanding of the integration of emerging topics related to microbiology and immunology in the undergraduate medical curriculum (UME). METHODS: An anonymous survey with 17 questions was emailed to medical school faculty who teach immunology and/or microbiology through the DR-Ed listserv, the American Society for Microbiology (ASM) Connect listserv, and attendees of the Association of Medical School Microbiology and Immunology Chairs (AMSMIC) Educational Strategies Workshop. Participants were asked about experiences, perceptions, and the decision-making process regarding integrating emerging topics into UME. RESULTS: The top emerging topics that were added to the curriculum or considered for addition in the last 10 years included COVID-19, Zika virus, mRNA vaccines, and Mpox (formerly known as monkeypox). Most respondents reported lectures and active learning as the major methods for topic delivery, with most faculty indicating that formative assessment was the best way to assess emerging topics. Content experts and course directors were the most cited individuals making these decisions. Top reasons for incorporating emerging topics into curricula included preparing students for clinical treatment of cases, followed by demonstrating the importance of basic science, and opportunities to integrate basic science into other disciplines. Challenges for incorporating these topics included making room in an already crowded curriculum, followed by content overload for students. CONCLUSIONS: This study describes the rationale for integrating emerging topics related to microbiology and immunology into UME, and identifies the current new and emerging topics, as well as the main methods of integration and assessment. These results may be used by medical educators to inform curricular decisions at their institutions. Future studies will include developing innovative learning modules that overcome barriers to integration.


Sujet(s)
Enseignement médical premier cycle , Infection par le virus Zika , Virus Zika , Humains , États-Unis , Programme d'études , Apprentissage par problèmes
3.
Curr Microbiol ; 81(5): 133, 2024 Apr 09.
Article de Anglais | MEDLINE | ID: mdl-38592489

RÉSUMÉ

Zika virus (ZIKV) infections have been associated with severe clinical outcomes, which may include neurological manifestations, especially in newborns with intrauterine infection. However, licensed vaccines and specific antiviral agents are not yet available. Therefore, a safe and low-cost therapy is required, especially for pregnant women. In this regard, metformin, an FDA-approved drug used to treat gestational diabetes, has previously exhibited an anti-ZIKA effect in vitro in HUVEC cells by activating AMPK. In this study, we evaluated metformin treatment during ZIKV infection in vitro in a JEG3-permissive trophoblast cell line. Our results demonstrate that metformin affects viral replication and protein synthesis and reverses cytoskeletal changes promoted by ZIKV infection. In addition, it reduces lipid droplet formation, which is associated with lipogenic activation of infection. Taken together, our results indicate that metformin has potential as an antiviral agent against ZIKV infection in vitro in trophoblast cells.


Sujet(s)
Metformine , Infection par le virus Zika , Virus Zika , Nouveau-né , Grossesse , Femelle , Humains , Infection par le virus Zika/traitement médicamenteux , Lignée cellulaire tumorale , Trophoblastes , Antiviraux/pharmacologie , Metformine/pharmacologie
4.
Science ; 384(6693): 260, 2024 Apr 19.
Article de Anglais | MEDLINE | ID: mdl-38635700

RÉSUMÉ

Naturally occurring bacterium could offer an additional way to control mosquito-borne diseases.


Sujet(s)
Aedes , Virus de la dengue , Dengue , Microbiome gastro-intestinal , Infection par le virus Zika , Virus Zika , Animaux , Humains , Vecteurs moustiques
5.
Parasit Vectors ; 17(1): 177, 2024 Apr 04.
Article de Anglais | MEDLINE | ID: mdl-38575981

RÉSUMÉ

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.


Sujet(s)
Aedes , Arbovirus , Virus des insectes , Infection par le virus Zika , Virus Zika , Femelle , Animaux , Vecteurs moustiques
6.
Cells ; 13(7)2024 Mar 29.
Article de Anglais | MEDLINE | ID: mdl-38607037

RÉSUMÉ

Zika virus (ZIKV) infection and pathogenesis are linked to the disruption of neurogenesis, congenital Zika syndrome and microcephaly by affecting neural progenitor cells. Nonstructural protein 5 (NS5) is the largest product encoded by ZIKV-RNA and is important for replication and immune evasion. Here, we studied the potential effects of NS5 on microtubules (MTs) and autophagy flux, together with the interplay of NS5 with histone deacetylase 6 (HDAC6). Fluorescence microscopy, biochemical cell-fractionation combined with the use of HDAC6 mutants, chemical inhibitors and RNA interference indicated that NS5 accumulates in nuclear structures and strongly promotes the acetylation of MTs that aberrantly reorganize in nested structures. Similarly, NS5 accumulates the p62 protein, an autophagic-flux marker. Therefore, NS5 alters events that are under the control of the autophagic tubulin-deacetylase HDAC6. HDAC6 appears to degrade NS5 by autophagy in a deacetylase- and BUZ domain-dependent manner and to control the cytoplasmic expression of NS5. Moreover, NS5 inhibits RNA-mediated RIG-I interferon (IFN) production, resulting in greater activity when autophagy is inhibited (i.e., effect correlated with NS5 stability). Therefore, it is conceivable that NS5 contributes to cell toxicity and pathogenesis, evading the IFN-immune response by overcoming HDAC6 functions. HDAC6 has emerged as an anti-ZIKV factor by targeting NS5.


Sujet(s)
Infection par le virus Zika , Virus Zika , Humains , Virus Zika/physiologie , Histone deacetylase 6 , Tubuline , Microtubules , ARN , Autophagie
7.
Viral Immunol ; 37(3): 167-175, 2024 Apr.
Article de Anglais | MEDLINE | ID: mdl-38574259

RÉSUMÉ

Zika virus (ZIKV) is an emerging flavivirus associated with several neurological diseases such as Guillain-Barré syndrome in adults and microcephaly in newborn children. Its distribution and mode of transmission (via Aedes aegypti and Aedes albopictus mosquitoes) collectively cause ZIKV to be a serious concern for global health. High genetic homology of flaviviruses and shared ecology is a hurdle for accurate detection. Distinguishing infections caused by different viruses based on serological recognition can be misleading as many anti-flavivirus monoclonal antibodies (mAbs) discovered to date are highly cross-reactive, especially those against the envelope (E) protein. To provide more specific research tools, we produced ZIKV E directed hybridoma cell lines and characterized two highly ZIKV-specific mAb clones (mAbs A11 and A42) against several members of the Flavivirus genus. Epitope mapping of mAb A11 revealed glycan loop specificity in Domain I of the ZIKV E protein. The development of two highly specific mAbs targeting the surface fusion protein of ZIKV presents a significant advancement in research capabilities as these can be employed as essential tools to enhance our understanding of ZIKV identification on infected cells ex vivo or in culture.


Sujet(s)
Aedes , Flavivirus , Infection par le virus Zika , Virus Zika , Animaux , Nouveau-né , Humains , Protéines de l'enveloppe virale , Anticorps monoclonaux , Anticorps neutralisants , Anticorps antiviraux
8.
PLoS Pathog ; 20(4): e1011975, 2024 Apr.
Article de Anglais | MEDLINE | ID: mdl-38557892

RÉSUMÉ

Arboviruses can emerge rapidly and cause explosive epidemics of severe disease. Some of the most epidemiologically important arboviruses, including dengue virus (DENV), Zika virus (ZIKV), Chikungunya (CHIKV) and yellow fever virus (YFV), are transmitted by Aedes mosquitoes, most notably Aedes aegypti and Aedes albopictus. After a mosquito blood feeds on an infected host, virus enters the midgut and infects the midgut epithelium. The virus must then overcome a series of barriers before reaching the mosquito saliva and being transmitted to a new host. The virus must escape from the midgut (known as the midgut escape barrier; MEB), which is thought to be mediated by transient changes in the permeability of the midgut-surrounding basal lamina layer (BL) following blood feeding. Here, we present a mathematical model of the within-mosquito population dynamics of DENV (as a model system for mosquito-borne viruses more generally) that includes the interaction of the midgut and BL which can account for the MEB. Our results indicate a dose-dependency of midgut establishment of infection as well as rate of escape from the midgut: collectively, these suggest that the extrinsic incubation period (EIP)-the time taken for DENV virus to be transmissible after infection-is shortened when mosquitoes imbibe more virus. Additionally, our experimental data indicate that multiple blood feeding events, which more closely mimic mosquito-feeding behavior in the wild, can hasten the course of infections, and our model predicts that this effect is sensitive to the amount of virus imbibed. Our model indicates that mutations to the virus which impact its replication rate in the midgut could lead to even shorter EIPs when double-feeding occurs. Mechanistic models of within-vector viral infection dynamics provide a quantitative understanding of infection dynamics and could be used to evaluate novel interventions that target the mosquito stages of the infection.


Sujet(s)
Aedes , Virus de la dengue , Dengue , Infection par le virus Zika , Virus Zika , Animaux , Tube digestif , Vecteurs moustiques
9.
Rev Med Virol ; 34(3): e2535, 2024 May.
Article de Anglais | MEDLINE | ID: mdl-38610091

RÉSUMÉ

Arthropod-borne viruses (arboviruses) pose significant threats to global public health by causing a spectrum of diseases ranging from mild febrile illnesses to severe neurological complications. Understanding the intricate interplay between arboviruses and the immune system within the central nervous system is crucial for developing effective strategies to combat these infections and mitigate their neurological sequelae. This review comprehensively explores the mechanisms by which arboviruses such as Zika virus, West Nile virus, and Dengue virus manipulate immune responses within the CNS, leading to diverse clinical manifestations.


Sujet(s)
Virus de la dengue , Virus du Nil occidental , Infection par le virus Zika , Virus Zika , Humains , Système nerveux central , Immunité , Infection par le virus Zika/complications
10.
Proc Natl Acad Sci U S A ; 121(16): e2317978121, 2024 Apr 16.
Article de Anglais | MEDLINE | ID: mdl-38593069

RÉSUMÉ

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.


Sujet(s)
Aedes , Infections à flavivirus , Flavivirus , Infection par le virus Zika , Virus Zika , Animaux , Souris , Flavivirus/génétique , Virus Zika/génétique , Ubiquitine/métabolisme , Ligases/métabolisme , Protéines virales/métabolisme , Mammifères
11.
Proc Natl Acad Sci U S A ; 121(16): e2400203121, 2024 Apr 16.
Article de Anglais | MEDLINE | ID: mdl-38598338

RÉSUMÉ

Viral outbreaks can cause widespread disruption, creating the need for diagnostic tools that provide high performance and sample versatility at the point of use with moderate complexity. Current gold standards such as PCR and rapid antigen tests fall short in one or more of these aspects. Here, we report a label-free and amplification-free nanopore sensor platform that overcomes these challenges via direct detection and quantification of viral RNA in clinical samples from a variety of biological fluids. The assay uses an optofluidic chip that combines optical waveguides with a fluidic channel and integrates a solid-state nanopore for sensing of individual biomolecules upon translocation through the pore. High specificity and low limit of detection are ensured by capturing RNA targets on microbeads and collecting them by optical trapping at the nanopore location where targets are released and rapidly detected. We use this device for longitudinal studies of the viral load progression for Zika and Severe Acute Respiratory Syndrome Coronavirus-2 (SARS-CoV-2) infections in marmoset and baboon animal models, respectively. The up to million-fold trapping-based target concentration enhancement enables amplification-free RNA quantification across the clinically relevant concentration range down to the assay limit of RT-qPCR as well as cases in which PCR failed. The assay operates across all relevant biofluids, including semen, urine, and whole blood for Zika and nasopharyngeal and throat swab, rectal swab, and bronchoalveolar lavage for SARS-CoV-2. The versatility, performance, simplicity, and potential for full microfluidic integration of the amplification-free nanopore assay points toward a unique approach to molecular diagnostics for nucleic acids, proteins, and other targets.


Sujet(s)
Nanopores , Infection par le virus Zika , Virus Zika , Animaux , ARN viral/génétique , ARN viral/métabolisme , SARS-CoV-2/génétique , SARS-CoV-2/métabolisme , Primates/génétique , Virus Zika/génétique , Sensibilité et spécificité , Techniques d'amplification d'acides nucléiques
12.
PLoS Negl Trop Dis ; 18(4): e0012053, 2024 Apr.
Article de Anglais | MEDLINE | ID: mdl-38557981

RÉSUMÉ

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.


Sujet(s)
Aedes , Arbovirus , Co-infection , Virus de la dengue , Dengue , Infection par le virus Zika , Virus Zika , Animaux , Co-infection/épidémiologie , Vecteurs moustiques , Dengue/épidémiologie
13.
PLoS Negl Trop Dis ; 18(4): e0012100, 2024 Apr.
Article de Anglais | MEDLINE | ID: mdl-38635656

RÉSUMÉ

Zika virus (ZIKV), an arbovirus from the Flaviviridae family, is the causative agent of Zika fever, a mild and frequent oligosymptomatic disease in humans. Nonetheless, on rare occasions, ZIKV infection can be associated with Guillain-Barré Syndrome (GBS), and severe congenital complications, such as microcephaly. The oligosymptomatic disease, however, presents symptoms that are quite similar to those observed in infections caused by other frequent co-circulating arboviruses, including dengue virus (DENV). Moreover, the antigenic similarity between ZIKV and DENV, and even with other members of the Flaviviridae family, complicates serological testing due to the high cross-reactivity of antibodies. Here, we designed, produced in a prokaryotic expression system, and purified three multiepitope proteins (ZIKV-1, ZIKV-2, and ZIKV-3) for differential diagnosis of Zika. The proteins were evaluated as antigens in ELISA tests for the detection of anti-ZIKV IgG using ZIKV- and DENV-positive human sera. The recombinant proteins were able to bind and detect anti-ZIKV antibodies without cross-reactivity with DENV-positive sera and showed no reactivity with Chikungunya virus (CHIKV)- positive sera. ZIKV-1, ZIKV-2, and ZIKV-3 proteins presented 81.6%, 95%, and 66% sensitivity and 97%, 96%, and 84% specificity, respectively. Our results demonstrate the potential of the designed and expressed antigens in the development of specific diagnostic tests for the detection of IgG antibodies against ZIKV, especially in regions with the circulation of multiple arboviruses.


Sujet(s)
Arbovirus , Fièvre chikungunya , Virus de la dengue , Dengue , Infection par le virus Zika , Virus Zika , Humains , Infection par le virus Zika/diagnostic , Virus Zika/génétique , Épitopes , Anticorps antiviraux , Immunoglobuline G
14.
PeerJ ; 12: e17038, 2024.
Article de Anglais | MEDLINE | ID: mdl-38529314

RÉSUMÉ

The mosquito Aedes aegypti, known to transmit important arboviral diseases, including dengue, chikungunya, Zika and yellow fever. Given the importance of this disease vector, a number of control programs have been proposed involving the use of the sterile insect technique (SIT). However, the success of this technique hinges on having a good understanding of the biology and behavior of the male mosquito. Behavioral responses of Ae. aegypti male populations developed for SIT technology were tested under laboratory conditions against chemical and natural irritants and repellents using an excito-repellency (ER) chamber. The results showed that there were no significant behavioral escape responses in any of the radiation-sterilized male Ae. aegypti test populations when exposed to citronella, DEET, transfluthrin, and deltamethrin, suggesting that SIT did not suppress the expected irritancy and repellency (avoidance) behaviors. The type of information reported in the current study is vital in defining the effects of SIT on vector behavior and understanding how such behavior may influence the success of SIT technology with regard to other vector control interventions.


Sujet(s)
Aedes , Infertilité masculine , Insectifuges , Infection par le virus Zika , Virus Zika , Mâle , Humains , Animaux , Irritants/pharmacologie , Vecteurs moustiques/physiologie , Insectifuges/pharmacologie , Infertilité masculine/prévention et contrôle
15.
Rev Soc Bras Med Trop ; 57: e004032024, 2024.
Article de Anglais | MEDLINE | ID: mdl-38536999

RÉSUMÉ

BACKGROUND: The riverine communities of the Amazon comprise different social groups that inhabit the rural areas on the banks of rivers and lakes. Residents usually travel by river to rural and urban areas and are then exposed to urbanized diseases such as those caused by arbovirus infection. In Brazil, emerging diseases such as dengue, Zika, chikungunya, and those caused by infection with Oropouche and Mayaro viruses necessitate epidemiological surveillance. This study was aimed at determining the frequency of positivity for immunoglobulin (Ig)G and IgM antibodies against Zika, chikungunya, and dengue viruses and performing molecular analyses to detect viral RNA for the Zika, chikungunya, dengue virus, Oropouche, and Mayaro viruses, in the same serum samples obtained from riverside populations. METHODS: This cross-sectional study was conducted in a riverside population in the Humaitá municipality of the Brazilian Amazon. More than 80% of the local population participated in this study. Entomological samples were collected to identify local mosquito vectors. RESULTS: Analysis of 205 human serological samples revealed IgG antibodies against the dengue virus in 85 individuals. No molecular positivity was observed in human samples. Entomological analyses revealed 3,187 Diptera species, with Mansonia being the most frequent genus. Aedes aegypti and Aedes albopictus were not detected in the two collections. CONCLUSIONS: IgG antibodies against the dengue virus were highly prevalent, suggesting previous exposure. The absence of the arbovirus vectors Aedes aegypti and Aedes albopictus in the samples supports the hypothesis that the infections recorded likely occurred outside the riverside communities investigated.


Sujet(s)
Aedes , Alphavirus , Infections à arbovirus , Fièvre chikungunya , Dengue , Infection par le virus Zika , Virus Zika , Animaux , Humains , Fièvre chikungunya/épidémiologie , Brésil/épidémiologie , Études transversales , Infections à arbovirus/épidémiologie , Vecteurs moustiques , Immunoglobuline G
16.
Viruses ; 16(3)2024 Feb 24.
Article de Anglais | MEDLINE | ID: mdl-38543716

RÉSUMÉ

We investigated the interaction between the insect-specific virus, Piura virus (PIUV), and the arbovirus Zika virus (ZIKV) in Aedes albopictus cells. We performed coinfection experiments in C6/36 cells. Piura virus (Cor 33 strain, Colombia) and ZIKV (PRVABC58 strain, Puerto Rico) were co-inoculated into C6/36 cells using two multiplicity of infection (MOI) combinations: 0.1 for both viruses and 1.0 for ZIKV, 0.1 for PIUV. Wells were infected in triplicate with either PIUV and ZIKV coinfection, ZIKV-only, or PIUV-only. Mock infected cells served as control wells. The cell suspension was collected daily 7 days post-infection. Zika virus load was titrated by TCID50 on Vero 76 cells. The ZIKV-only infection and PIUV and ZIKV coinfection experiments were also quantified by RT-qPCR. We also investigated whether ZIKV interfered in the PIUV replication. PIUV suppressed the replication of ZIKV, resulting in a 10,000-fold reduction in ZIKV titers within 3 days post-infection. PIUV viral loads were not reduced in the presence of ZIKV. We conclude that, when concurrently infected, PIUV suppresses ZIKV in C6/36 cells while ZIKV does not interfere in PIUV replication.


Sujet(s)
Aedes , Co-infection , Virus des insectes , Infection par le virus Zika , Virus Zika , Animaux , Réplication virale
17.
Lancet Neurol ; 23(4): 344-381, 2024 Apr.
Article de Anglais | MEDLINE | ID: mdl-38493795

RÉSUMÉ

BACKGROUND: Disorders affecting the nervous system are diverse and include neurodevelopmental disorders, late-life neurodegeneration, and newly emergent conditions, such as cognitive impairment following COVID-19. Previous publications from the Global Burden of Disease, Injuries, and Risk Factor Study estimated the burden of 15 neurological conditions in 2015 and 2016, but these analyses did not include neurodevelopmental disorders, as defined by the International Classification of Diseases (ICD)-11, or a subset of cases of congenital, neonatal, and infectious conditions that cause neurological damage. Here, we estimate nervous system health loss caused by 37 unique conditions and their associated risk factors globally, regionally, and nationally from 1990 to 2021. METHODS: We estimated mortality, prevalence, years lived with disability (YLDs), years of life lost (YLLs), and disability-adjusted life-years (DALYs), with corresponding 95% uncertainty intervals (UIs), by age and sex in 204 countries and territories, from 1990 to 2021. We included morbidity and deaths due to neurological conditions, for which health loss is directly due to damage to the CNS or peripheral nervous system. We also isolated neurological health loss from conditions for which nervous system morbidity is a consequence, but not the primary feature, including a subset of congenital conditions (ie, chromosomal anomalies and congenital birth defects), neonatal conditions (ie, jaundice, preterm birth, and sepsis), infectious diseases (ie, COVID-19, cystic echinococcosis, malaria, syphilis, and Zika virus disease), and diabetic neuropathy. By conducting a sequela-level analysis of the health outcomes for these conditions, only cases where nervous system damage occurred were included, and YLDs were recalculated to isolate the non-fatal burden directly attributable to nervous system health loss. A comorbidity correction was used to calculate total prevalence of all conditions that affect the nervous system combined. FINDINGS: Globally, the 37 conditions affecting the nervous system were collectively ranked as the leading group cause of DALYs in 2021 (443 million, 95% UI 378-521), affecting 3·40 billion (3·20-3·62) individuals (43·1%, 40·5-45·9 of the global population); global DALY counts attributed to these conditions increased by 18·2% (8·7-26·7) between 1990 and 2021. Age-standardised rates of deaths per 100 000 people attributed to these conditions decreased from 1990 to 2021 by 33·6% (27·6-38·8), and age-standardised rates of DALYs attributed to these conditions decreased by 27·0% (21·5-32·4). Age-standardised prevalence was almost stable, with a change of 1·5% (0·7-2·4). The ten conditions with the highest age-standardised DALYs in 2021 were stroke, neonatal encephalopathy, migraine, Alzheimer's disease and other dementias, diabetic neuropathy, meningitis, epilepsy, neurological complications due to preterm birth, autism spectrum disorder, and nervous system cancer. INTERPRETATION: As the leading cause of overall disease burden in the world, with increasing global DALY counts, effective prevention, treatment, and rehabilitation strategies for disorders affecting the nervous system are needed. FUNDING: Bill & Melinda Gates Foundation.


Sujet(s)
Trouble du spectre autistique , COVID-19 , Maladies transmissibles , Neuropathies diabétiques , Naissance prématurée , Infection par le virus Zika , Virus Zika , Femelle , Humains , Nouveau-né , Charge mondiale de morbidité , Maladies transmissibles/épidémiologie , Facteurs de risque , Évolution de la maladie , Santé mondiale , COVID-19/épidémiologie , Années de vie ajustées sur la qualité , Espérance de vie
18.
Sci Rep ; 14(1): 7424, 2024 03 28.
Article de Anglais | MEDLINE | ID: mdl-38548897

RÉSUMÉ

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.


Sujet(s)
Aedes , Épidémies , Infection par le virus Zika , Virus Zika , Animaux , Humains , Brésil/épidémiologie , Eaux d'égout
19.
J Gen Virol ; 105(3)2024 03.
Article de Anglais | MEDLINE | ID: mdl-38488850

RÉSUMÉ

Arboviruses such as chikungunya, dengue and zika viruses cause debilitating diseases in humans. The principal vector species that transmits these viruses is the Aedes mosquito. Lack of substantial knowledge of the vector species hinders the advancement of strategies for controlling the spread of arboviruses. To supplement our information on mosquitoes' responses to virus infection, we utilized Aedes aegypti-derived Aag2 cells to study changes at the transcriptional level during infection with chikungunya virus (CHIKV). We observed that genes belonging to the redox pathway were significantly differentially regulated. Upon quantifying reactive oxygen species (ROS) in the cells during viral infection, we further discovered that ROS levels are considerably higher during the early hours of infection; however, as the infection progresses, an increase in antioxidant gene expression suppresses the oxidative stress in cells. Our study also suggests that ROS is a critical regulator of viral replication in cells and inhibits intracellular and extracellular viral replication by promoting the Rel2-mediated Imd immune signalling pathway. In conclusion, our study provides evidence for a regulatory role of oxidative stress in infected Aedes-derived cells.


Sujet(s)
Aedes , Arbovirus , Fièvre chikungunya , Infection par le virus Zika , Virus Zika , Humains , Animaux , Espèces réactives de l'oxygène , Vecteurs moustiques , Stress oxydatif , Immunité innée
20.
PLoS Negl Trop Dis ; 18(3): e0012017, 2024 Mar.
Article de Anglais | MEDLINE | ID: mdl-38517912

RÉSUMÉ

The 2015-17 Zika virus (ZIKV) epidemic in the Americas subsided faster than expected and evolving population immunity was postulated to be the main reason. Herd immunization is suggested to occur around 60-70% seroprevalence, depending on demographic density and climate suitability. However, herd immunity was only documented for a few cities in South America, meaning a substantial portion of the population might still be vulnerable to a future Zika virus outbreak. The aim of our study was to determine the vulnerability of populations to ZIKV by comparing the environmental suitability of ZIKV transmission to the observed seroprevalence, based on published studies. Using a systematic search, we collected seroprevalence and geospatial data for 119 unique locations from 37 studies. Extracting the environmental suitability at each location and converting to a hypothetical expected seroprevalence, we were able to determine the discrepancy between observed and expected. This discrepancy is an indicator of vulnerability and divided into three categories: high risk, low risk, and very low risk. The vulnerability was used to evaluate the level of risk that each location still has for a ZIKV outbreak to occur. Of the 119 unique locations, 69 locations (58%) fell within the high risk category, 47 locations (39%) fell within the low risk category, and 3 locations (3%) fell within the very low risk category. The considerable heterogeneity between environmental suitability and seroprevalence potentially leaves a large population vulnerable to future infection. Vulnerability seems to be especially pronounced at the fringes of the environmental suitability for ZIKV (e.g. Sao Paulo, Brazil). The discrepancies between observed and expected seroprevalence raise the question: "why did the ZIKV epidemic stop with large populations unaffected?". This lack of understanding also highlights that future ZIKV outbreaks currently cannot be predicted with confidence.


Sujet(s)
Infection par le virus Zika , Virus Zika , Humains , Études séroépidémiologiques , Brésil/épidémiologie , Épidémies de maladies
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...