Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 1.380
Filter
1.
Nat Commun ; 15(1): 8221, 2024 Sep 19.
Article in English | MEDLINE | ID: mdl-39300135

ABSTRACT

The main vectors of Zika virus (ZIKV) and dengue virus (DENV) are Aedes aegypti and Ae. albopictus, with Ae. aegypti being more competent. However, the underlying mechanisms remain unclear. Here, we find Ae. albopictus shows comparable vector competence to ZIKV/DENV with Ae. aegypti by blood-feeding after antibiotic treatment or intrathoracic injection. This suggests that midgut microbiota can influence vector competence. Enterobacter hormaechei_B17 (Eh_B17) is isolated from field-collected Ae. albopictus and conferred resistance to ZIKV/DENV infection in Ae. aegypti after gut-transplantation. Sphingosine, a metabolite secreted by Eh_B17, effectively suppresses ZIKV infection in both Ae. aegypti and cell cultures by blocking viral entry during the fusion step, with an IC50 of approximately 10 µM. A field survey reveals that Eh_B17 preferentially colonizes Ae. albopictus compared to Ae. aegypti. And field Ae. albopictus positive for Eh_B17 are more resistant to ZIKV infection. These findings underscore the potential of gut symbiotic bacteria, such as Eh_B17, to modulate the arbovirus vector competence of Aedes mosquitoes. As a natural antiviral agent, Eh_B17 holds promise as a potential candidate for blocking ZIKV/DENV transmission.


Subject(s)
Aedes , Dengue Virus , Enterobacter , Gastrointestinal Microbiome , Mosquito Vectors , Sphingosine , Symbiosis , Zika Virus , Aedes/virology , Aedes/microbiology , Aedes/drug effects , Animals , Mosquito Vectors/microbiology , Mosquito Vectors/virology , Mosquito Vectors/drug effects , Zika Virus/physiology , Zika Virus/drug effects , Dengue Virus/drug effects , Dengue Virus/physiology , Gastrointestinal Microbiome/drug effects , Sphingosine/analogs & derivatives , Sphingosine/metabolism , Sphingosine/pharmacology , Enterobacter/drug effects , Enterobacter/physiology , Zika Virus Infection/transmission , Zika Virus Infection/virology , Dengue/transmission , Dengue/virology , Dengue/prevention & control , Female , Virus Internalization/drug effects , Humans
2.
mSphere ; 9(8): e0040124, 2024 Aug 28.
Article in English | MEDLINE | ID: mdl-39092912

ABSTRACT

As climate change alters Earth's biomes, it is expected the transmission dynamics of mosquito-borne viruses will change. While the effects of temperature changes on mosquito-virus interactions and the spread of the pathogens have been elucidated over the last decade, the impact of relative humidity changes is still relatively unknown. To overcome this knowledge gap, we exposed Aedes aegypti females to various humidity conditions. We measured different components of vectorial capacity such as survival, blood-feeding rates, and changes in infection and dissemination of Zika virus. Survival decreased as the humidity level decreased, while infection rates increased as the humidity level decreased. Alternatively, blood feeding rates and disseminated infection rates peaked at the intermediate 50% relative humidity treatment but were the same in the 30% and 80% relative humidity treatments. These results provide empirical evidence that Ae. aegypti exposure to low humidity can enhance Zika virus infection in the mosquito, which has important implications in predicting how climate change will impact mosquito-borne viruses.IMPORTANCEViruses transmitted by mosquitoes to humans are a major public health burden and are expected to increase under climate change. While we know that temperature is an important driver of variation in arbovirus replication in the mosquito, very little is known about how other relevant climate variables such as humidity will influence the interaction between mosquitoes and the viruses they transmit. Given the variability in humidity across environments, and the predicted changes in humidity under climate change, it is imperative that we also study the impact that it has on mosquito infection and transmission of arboviruses.


Subject(s)
Aedes , Climate Change , Humidity , Mosquito Vectors , Zika Virus Infection , Zika Virus , Aedes/virology , Aedes/physiology , Animals , Zika Virus Infection/transmission , Zika Virus Infection/virology , Mosquito Vectors/virology , Zika Virus/physiology , Female , Temperature , Feeding Behavior
3.
Med Trop Sante Int ; 4(2)2024 06 30.
Article in French | MEDLINE | ID: mdl-39099710

ABSTRACT

Zika virus infection, most oft n responsible for a benign arboviral disease or an asymptomatic infection, rarely Guillain-Barré syndrome, can become problematic in pregnant women, due to a risk of fetal malformations, in particular microcephaly linked to its neurotropism. The most recent large-scale epidemic was observed throughout Latin America between 2015 and 2017, causing several hundred thousand cases. Transmission is predominantly vector-borne, but sexual transmission has been described, mainly among travelers, although it undoubtedly accounts for a significant proportion of transmission in epidemic areas. The aim of this review is to describe this sexual transmission, mainly through examples linked to this large-scale epidemic in Latin America, to describe the link with prolonged excretion of infectious viral particles in genital secretions, especially semen but also vaginal secretions, and to highlight possible preventive measures apart from vector transmission, in particular the need for pregnant women or women wishing to become pregnant to avoid visiting countries where circulation of Zika virus is described.


Subject(s)
Sexually Transmitted Diseases, Viral , Zika Virus Infection , Zika Virus Infection/transmission , Zika Virus Infection/prevention & control , Zika Virus Infection/epidemiology , Humans , Female , Pregnancy , Sexually Transmitted Diseases, Viral/transmission , Sexually Transmitted Diseases, Viral/prevention & control , Sexually Transmitted Diseases, Viral/epidemiology , Pregnancy Complications, Infectious/prevention & control , Pregnancy Complications, Infectious/virology , Pregnancy Complications, Infectious/epidemiology , Male , Latin America/epidemiology
4.
Sci Rep ; 14(1): 18112, 2024 08 05.
Article in English | MEDLINE | ID: mdl-39103482

ABSTRACT

This study presents a computational investigation of a stochastic Zika virus along with optimal control model using the Legendre spectral collocation method (LSCM). By accumulation of stochasticity into the model through the proposed stochastic differential equations, we appropriating the random fluctuations essential in the progression and disease transmission. The stability, convergence and accuracy properties of the LSCM are conscientiously analyzed and also demonstrating its strength for solving the complex epidemiological models. Moreover, the study evaluates the various control strategies, such as treatment, prevention and treatment pesticide control, and identifies optimal combinations that the intervention costs and also minimize the proposed infection rates. The basic properties of the given model, such as the reproduction number, were determined with and without the presence of the control strategies. For R 0 < 0 , the model satisfies the disease-free equilibrium, in this case the disease die out after some time, while for R 0 > 1 , then endemic equilibrium is satisfied, in this case the disease spread in the population at higher scale. The fundamental findings acknowledge the significant impact of stochastic phonemes on the robustness and effectiveness of control strategies that accelerating the need for cost-effective and multi-faceted approaches. In last the results provide the valuable insights for public health department to enabling more impressive mitigation of Zika virus outbreaks and management in real-world scenarios.


Subject(s)
Stochastic Processes , Zika Virus Infection , Zika Virus , Zika Virus Infection/epidemiology , Zika Virus Infection/prevention & control , Zika Virus Infection/transmission , Humans , Zika Virus/physiology , Computer Simulation , Epidemiological Models
5.
Indian J Public Health ; 68(2): 163-166, 2024 Apr 01.
Article in English | MEDLINE | ID: mdl-38953800

ABSTRACT

BACKGROUND: Several sporadic cases and outbreaks of Zika virus disease have been reported from different states of India. OBJECTIVES: This paper explored the possibility of any ongoing transmission of Zika virus (ZIKV) in the Bhopal region of Central India, where the last outbreak of this disease was reported in 2018. MATERIALS AND METHODS: We screened a group of 75 febrile patients who had already tested negative for the locally endemic causes of fever like dengue, chikungunya, enteric fever, malaria, and scrub typhus and two groups of asymptomatic healthy individuals represented by blood donors (n = 75) and antenatal mothers (n = 75). We tested blood samples of febrile patients for ZIKV RNA using real-time polymerase chain reaction (PCR), and for the healthy individuals, we determined anti-zika immunoglobulin G (IgG) antibodies using enzyme-linked immunosorbent assay. RESULTS: ZIKV RNA was not detected in any of the 75 samples tested by real-time PCR assay. Among the voluntary blood donors and antenatal mothers, a total of 10 (15.38%) and 5 (6.66%) individuals were found to be seropositive for anti-ZIKV IgG antibodies, respectively. The seropositive group was found to have higher age 33.06 (±10.83) years as compared to seronegative individuals 26.60 (±5.12) years (P = 0.037). CONCLUSION: This study, which is the first survey of seroprevalence of anti-Zika antibodies from India, reports an overall seropositivity rate of 10% for anti-Zika antibodies among the healthy population, suggesting an ongoing, low level, silent transmission of ZIKV in the local community.


Subject(s)
Zika Virus Infection , Zika Virus , Humans , India/epidemiology , Zika Virus Infection/epidemiology , Zika Virus Infection/transmission , Seroepidemiologic Studies , Adult , Female , Pilot Projects , Male , Zika Virus/immunology , Zika Virus/isolation & purification , Immunoglobulin G/blood , Young Adult , Antibodies, Viral/blood , Middle Aged , RNA, Viral , Adolescent , Enzyme-Linked Immunosorbent Assay , Real-Time Polymerase Chain Reaction
6.
Travel Med Infect Dis ; 60: 102737, 2024.
Article in English | MEDLINE | ID: mdl-38996856

ABSTRACT

BACKGROUND: The spread of vector-borne infectious diseases is determined, among other things, by temperature. Thus, climate change will have an influence on their global distribution. In the future, Europe will approach the temperature optimum for the transmission of ZIKV and CHIKV. Climate scenarios and climate models can be used to depict future climatic changes and to draw conclusions about future risk areas for vector-borne infectious diseases. METHODS: Based on the RCP 4.5 and RCP 8.5 climate scenarios, a geospatial analysis was carried out for the future temperature suitability of ZIKV and CHIKV in Europe. The results were presented in maps and the percentage of the affected areas calculated. RESULTS: Due to rising temperatures, the risk areas for transmission of ZIKV and CHIKV spread in both RCP scenarios. For CHIKV transmission, Spain, Portugal, the Mediterranean coast and areas near the Black Sea are mainly affected. Due to high temperatures, large areas throughout Europe are at risk for ZIKV and CHIKV transmission. CONCLUSION: Temperature is only one of many factors influencing the spread of vector-borne infectious diseases. Nevertheless, the representation of risk areas on the basis of climate scenarios allows an assessment of future risk development. Monitoring and adaptation strategies are indispensable for coping with and containing possible future autochthonous transmissions and epidemics in Europe.


Subject(s)
Chikungunya Fever , Climate Change , Geographic Information Systems , Zika Virus Infection , Zika Virus Infection/epidemiology , Zika Virus Infection/transmission , Humans , Chikungunya Fever/epidemiology , Chikungunya Fever/transmission , Europe/epidemiology , Zika Virus , Animals , Risk Assessment , Chikungunya virus , Temperature
7.
Biometrics ; 80(3)2024 Jul 01.
Article in English | MEDLINE | ID: mdl-39036985

ABSTRACT

The dynamics that govern disease spread are hard to model because infections are functions of both the underlying pathogen as well as human or animal behavior. This challenge is increased when modeling how diseases spread between different spatial locations. Many proposed spatial epidemiological models require trade-offs to fit, either by abstracting away theoretical spread dynamics, fitting a deterministic model, or by requiring large computational resources for many simulations. We propose an approach that approximates the complex spatial spread dynamics with a Gaussian process. We first propose a flexible spatial extension to the well-known SIR stochastic process, and then we derive a moment-closure approximation to this stochastic process. This moment-closure approximation yields ordinary differential equations for the evolution of the means and covariances of the susceptibles and infectious through time. Because these ODEs are a bottleneck to fitting our model by MCMC, we approximate them using a low-rank emulator. This approximation serves as the basis for our hierarchical model for noisy, underreported counts of new infections by spatial location and time. We demonstrate using our model to conduct inference on simulated infections from the underlying, true spatial SIR jump process. We then apply our method to model counts of new Zika infections in Brazil from late 2015 through early 2016.


Subject(s)
Computer Simulation , Stochastic Processes , Zika Virus Infection , Humans , Normal Distribution , Zika Virus Infection/epidemiology , Zika Virus Infection/transmission , Epidemiological Models , Models, Statistical , Markov Chains
8.
J Community Health ; 49(6): 1044-1053, 2024 Dec.
Article in English | MEDLINE | ID: mdl-38824473

ABSTRACT

The 2016 outbreak of Zika virus (ZIKV) infected millions and resulted in thousands of infants born with malformations. Though the clusters of severe birth defects resulting from this outbreak have subsided, ZIKV continues to be a concern throughout much of Latin America and the Caribbean. Travel and sexual intercourse remain the dominant transmission risk factors for women of reproductive age and their partners. This is particularly true for communities in Brooklyn, New York, that comprise large immigrant and foreign-born populations. Practitioners of public health understand little about how women at risk for ZIKV are most likely to receive information about the virus or who they trust most to provide that information. In the context of five focus group discussions, this study explored the knowledge and communication preferences of 20 women of reproductive age in Central Brooklyn. Results derived from a thematic analysis suggest that while most women are familiar with mosquitos as ZIKV vectors, knowledge of sexual transmission is considerably lower. Many respondents believe that only women who are pregnant or trying to become pregnant are at risk, and public health agencies, such as the U.S. Centers for Disease Control and Prevention, remain the most trusted sources of information. These findings can support more effective communication about the risks of ZIKV infection and other vector-borne diseases to women in New York City and similar urban communities.


Subject(s)
Focus Groups , Health Knowledge, Attitudes, Practice , Zika Virus Infection , Humans , Zika Virus Infection/epidemiology , Zika Virus Infection/transmission , Zika Virus Infection/prevention & control , Female , New York City/epidemiology , Adult , Young Adult , Adolescent , Communication , Pregnancy , Middle Aged , Zika Virus
9.
Parasit Vectors ; 17(1): 267, 2024 Jun 25.
Article in English | MEDLINE | ID: mdl-38918848

ABSTRACT

BACKGROUND: Past findings demonstrate that arthropods can egest midgut microbiota into the host skin leading to dual colonization of the vertebrate host with pathogens and saliva microbiome. A knowledge gap exists on how the saliva microbiome interacts with the pathogen in the saliva. To fill this gap, we need to first define the microbial composition of mosquito saliva. METHODS: The current study aimed at analyzing and comparing the microbial profile of Aedes albopictus saliva and midgut as well as assessing the impact of Zika virus (ZIKV) infection on the midgut and saliva microbial composition. Colony-reared Ae. albopictus strains were either exposed to ZIKV infectious or noninfectious bloodmeal. At 14 ays postinfection, the 16S V3-V4 hypervariable rRNA region was amplified from midgut and saliva samples and sequenced on an Illumina MiSeq platform. The relative abundance and diversity of midgut and saliva microbial taxa were assessed. RESULTS: We observed a richer microbial community in the saliva compared with the midgut, yet some of the microbial taxa were common in the midgut and saliva. ZIKV infection did not impact the microbial diversity of midgut or saliva. Further, we identified Elizabethkingia spp. in the Ae. albopictus saliva. CONCLUSIONS: This study provides insights into the microbial community of the Ae. albopictus saliva as well as the influence of ZIKV infection on the microbial composition of its midgut and saliva. The identification of Elizabethkingia spp., an emerging pathogen of global health significance, in Ae. albopictus saliva is of medical importance. Future studies to assess the interactions between Ae. albopictus saliva microbiome and ZIKV could lead to novel strategies for developing transmission barrier tools.


Subject(s)
Aedes , Microbiota , Mosquito Vectors , Saliva , Zika Virus , Animals , Saliva/microbiology , Saliva/virology , Aedes/microbiology , Aedes/virology , Zika Virus/genetics , Zika Virus/isolation & purification , Mosquito Vectors/microbiology , Mosquito Vectors/virology , Gastrointestinal Microbiome , RNA, Ribosomal, 16S/genetics , Female , Zika Virus Infection/transmission , Zika Virus Infection/virology , Gastrointestinal Tract/microbiology , Gastrointestinal Tract/virology
10.
Curr Opin Infect Dis ; 37(4): 238-244, 2024 Aug 01.
Article in English | MEDLINE | ID: mdl-38842472

ABSTRACT

PURPOSE OF REVIEW: Arbovirus infections are a challenge for immunocompromised hosts who travel to or live in endemic regions or who receive organs or tissues from donors who travel or live in such areas. This review addresses Dengue (DENV), Chikungunya (CHIKV), and Zika (ZIKV) infections in hematological patients, hematopoietic cell or solid organ transplant recipients, and people with HIV (PWH). RECENT FINDINGS: Transmission is mainly due through Aedes mosquito bite. DENV and ZIKV may also be transmitted through blood, tissues or donor grafts. Clinical manifestations are quite similar and diagnosis requires laboratory confirmation to provide appropriate management. The best diagnostic method is PCR since serology may present false negative results in immunocompromised patients, or cross-reactivity as in the case of DENV and ZIKV. There is no specific treatment for any of these infections. SUMMARY: Educational and preventive measures are the best strategy: vector control, knowledge of the vector's habits, protection against mosquito bites, avoiding travel to endemic areas or with a current epidemic, and avoiding nonvector transmission according to local recommendations for donor deferral. Vaccination, currently only available for DENV, has not yet been studied in immunocompromised patients and is not currently recommended.


Subject(s)
Chikungunya Fever , Dengue , Immunocompromised Host , Zika Virus Infection , Humans , Dengue/immunology , Dengue/epidemiology , Dengue/transmission , Chikungunya Fever/immunology , Chikungunya Fever/epidemiology , Chikungunya Fever/transmission , Zika Virus Infection/immunology , Zika Virus Infection/transmission , Zika Virus Infection/epidemiology , Endemic Diseases , Animals
11.
Infect Genet Evol ; 123: 105627, 2024 Sep.
Article in English | MEDLINE | ID: mdl-38909667

ABSTRACT

BACKGROUND: Hainan Island and the Leizhou Peninsula, the southernmost part of mainland China, are areas where Aedes aegypti and Ae. albopictus are sympatric and are also high-incidence areas of dengue outbreaks in China. Many studies have suggested that Aedes endogenous viral components (EVEs) are enriched in piRNA clusters which can silence incoming viral genomes. Investigation the EVEs present in the piRNA clusters associated with viral infection of Aedes mosquitoes in these regions may provide a theoretical basis for novel transmission-blocking vector control strategies. METHODS: In this study, specific primers for endogenous Flaviviridae elements (EFVEs) and endogenous Rhabdoviridae elements (ERVEs) were used to detect the distribution of Zika virus infection associated EVEs in the genomes of individuals of the two Aedes mosquitoes. Genetic diversity of EVEs with a high detection rate was also analyzed. RESULTS: The results showed that many EVEs associated with Zika virus infection were detected in both Aedes species, with the detection rates were 47.68% to 100% in Ae. aegypti and 36.15% to 92.31% in sympatric Ae. albopictus populations. EVEs detection rates in another 17 Ae. albopictus populations ranged from 29.39% to 89.85%. Genetic diversity analyses of the four EVEs (AaFlavi53, AaRha61, AaRha91 and AaRha100) of Ae. aegypti showed that each had high haplotype diversity and low nucleotide diversity. The number of haplotypes in AaFlavi53 was 8, with the dominant haplotype being Hap_1 and the other 7 haplotypes being further mutated from Hap_1 in a lineage direction. In contrast, the haplotype diversity of the other three ERVEs (AaRha61, AaRha91 and AaRha100) was more diverse and richer, with the haplotype numbers were 9, 15 and 19 respectively. In addition, these EVEs all showed inconsistent patterns of both population differentiation and dispersal compared to neutral evolutionary genes such as the Mitochondrial COI gene. CONCLUSION: The EFVEs and ERVEs tested were present at high frequencies in the field Aedes mosquito populations. The haplotype diversity of the EFVE AaFlavi53 was relatively lower and the three ERVEs (AaRha61, AaRha91, AaRha100) were higher. None of the four EVEs could be indicative of the genetic diversity of the Ae. aegypti population. This study provided theoretical support for the use of EVEs to block arbovirus transmission, but further research is needed into the mechanisms by which these EVEs are antiviral to Aedes mosquitoes.


Subject(s)
Aedes , Genetic Variation , Rhabdoviridae , Aedes/virology , Aedes/genetics , Animals , China/epidemiology , Rhabdoviridae/genetics , Flaviviridae/genetics , Flaviviridae/classification , Mosquito Vectors/virology , Mosquito Vectors/genetics , Phylogeny , RNA, Small Interfering/genetics , Zika Virus/genetics , Zika Virus Infection/virology , Zika Virus Infection/transmission , Zika Virus Infection/epidemiology
12.
PLoS Negl Trop Dis ; 18(6): e0011811, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38829905

ABSTRACT

BACKGROUND: Dengue, Zika, and chikungunya, whose viruses are transmitted mainly by Aedes aegypti, significantly impact human health worldwide. Despite the recent development of promising vaccines against the dengue virus, controlling these arbovirus diseases still depends on mosquito surveillance and control. Nonetheless, several studies have shown that these measures are not sufficiently effective or ineffective. Identifying higher-risk areas in a municipality and directing control efforts towards them could improve it. One tool for this is the premise condition index (PCI); however, its measure requires visiting all buildings. We propose a novel approach capable of predicting the PCI based on facade street-level images, which we call PCINet. METHODOLOGY: Our study was conducted in Campinas, a one million-inhabitant city in São Paulo, Brazil. We surveyed 200 blocks, visited their buildings, and measured the three traditional PCI components (building and backyard conditions and shading), the facade conditions (taking pictures of them), and other characteristics. We trained a deep neural network with the pictures taken, creating a computational model that can predict buildings' conditions based on the view of their facades. We evaluated PCINet in a scenario emulating a real large-scale situation, where the model could be deployed to automatically monitor four regions of Campinas to identify risk areas. PRINCIPAL FINDINGS: PCINet produced reasonable results in differentiating the facade condition into three levels, and it is a scalable strategy to triage large areas. The entire process can be automated through data collection from facade data sources and inferences through PCINet. The facade conditions correlated highly with the building and backyard conditions and reasonably well with shading and backyard conditions. The use of street-level images and PCINet could help to optimize Ae. aegypti surveillance and control, reducing the number of in-person visits necessary to identify buildings, blocks, and neighborhoods at higher risk from mosquito and arbovirus diseases.


Subject(s)
Aedes , Dengue , Mosquito Vectors , Aedes/virology , Aedes/physiology , Animals , Brazil/epidemiology , Humans , Mosquito Vectors/virology , Mosquito Vectors/physiology , Dengue/prevention & control , Dengue/epidemiology , Dengue/transmission , Cities , Mosquito Control/methods , Image Processing, Computer-Assisted/methods , Zika Virus Infection/prevention & control , Zika Virus Infection/epidemiology , Zika Virus Infection/transmission
13.
Sci Rep ; 14(1): 10003, 2024 05 01.
Article in English | MEDLINE | ID: mdl-38693192

ABSTRACT

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


Subject(s)
Aedes , Markov Chains , Zika Virus Infection , Zika Virus , Zika Virus Infection/transmission , Zika Virus Infection/epidemiology , Colombia/epidemiology , Humans , Animals , Aedes/virology , Bayes Theorem , Mosquito Vectors/virology , Disease Outbreaks
14.
PLoS One ; 19(5): e0281851, 2024.
Article in English | MEDLINE | ID: mdl-38748732

ABSTRACT

Zika (ZIKV) and chikungunya (CHIKV) are arboviruses that cause infections in humans and can cause clinical complications, representing a worldwide public health problem. Aedes aegypti is the primary vector of these pathogens and Culex quinquefasciatus may be a potential ZIKV vector. This study aimed to evaluate fecundity, fertility, survival, longevity, and blood feeding activity in Ae. aegypti after exposure to ZIKV and CHIKV and, in Cx. quinquefasciatus exposed to ZIKV. Three colonies were evaluated: AeCamp (Ae. aegypti-field), RecL (Ae. aegypti-laboratory) and CqSLab (Cx. quinquefasciatus-laboratory). Seven to 10 days-old females from these colonies were exposed to artificial blood feeding with CHIKV or ZIKV. CHIKV caused reduction in fecundity and fertility in AeCamp and reduction in survival and fertility in RecL. ZIKV impacted survival in RecL, fertility in AeCamp and, fecundity and fertility in CqSLab. Both viruses had no effect on blood feeding activity. These results show that CHIKV produces a higher biological cost in Ae. aegypti, compared to ZIKV, and ZIKV differently alters the biological performance in colonies of Ae. aegypti and Cx. quinquefasciatus. These results provide a better understanding over the processes of virus-vector interaction and can shed light on the complexity of arbovirus transmission.


Subject(s)
Aedes , Chikungunya virus , Culex , Fertility , Mosquito Vectors , Zika Virus Infection , Zika Virus , Animals , Aedes/virology , Aedes/physiology , Chikungunya virus/physiology , Chikungunya virus/pathogenicity , Zika Virus/physiology , Zika Virus/pathogenicity , Culex/virology , Culex/physiology , Mosquito Vectors/virology , Mosquito Vectors/physiology , Female , Zika Virus Infection/transmission , Zika Virus Infection/virology , Chikungunya Fever/transmission , Chikungunya Fever/virology , Feeding Behavior/physiology , Humans , Longevity
15.
Euro Surveill ; 29(20)2024 05.
Article in English | MEDLINE | ID: mdl-38757289

ABSTRACT

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.


Subject(s)
Aedes , Chikungunya virus , Dengue Virus , Zika Virus , Animals , Aedes/virology , Humans , Zika Virus/isolation & purification , Dengue Virus/isolation & purification , Chikungunya virus/isolation & purification , Paris , Mosquito Vectors/virology , West Nile virus/isolation & purification , Arboviruses/isolation & purification , Arbovirus Infections/transmission , Flavivirus/isolation & purification , France , Dengue/transmission , Dengue/epidemiology , Zika Virus Infection/transmission
16.
Med Clin (Barc) ; 163(3): 134-142, 2024 08 16.
Article in English, Spanish | MEDLINE | ID: mdl-38643027

ABSTRACT

Arboviruses represent a threat to transfusion safety for several reasons: the presence of vectors and the notification of autochthonous cases in our region, the recent increase in the number of cases transmitted through blood and/or blood component transfusion, the high prevalence rates of RNA of the main arboviruses in asymptomatic blood donors, and their ability to survive processing and storage in the different blood components. In an epidemic outbreak caused by an arbovirus in our region, transfusion centres can apply different measures: reactive measures, related to donor selection or arbovirus screening, and proactive measures, such as pathogen inactivation methods. The study of the epidemiology of the main arboviruses and understanding the effectiveness of the different measures that we can adopt are essential to ensure that our blood components remain safe.


Subject(s)
Arbovirus Infections , Arboviruses , Blood Donors , Blood Safety , Transfusion Reaction , Humans , Spain/epidemiology , Arbovirus Infections/transmission , Arbovirus Infections/epidemiology , Arbovirus Infections/prevention & control , Transfusion Reaction/epidemiology , Donor Selection/standards , Disease Outbreaks/prevention & control , Blood Transfusion/standards , Zika Virus Infection/transmission , Zika Virus Infection/epidemiology , Zika Virus Infection/prevention & control , West Nile Fever/transmission , West Nile Fever/epidemiology , West Nile Fever/prevention & control , West Nile Fever/diagnosis
17.
Science ; 384(6693): eadn9524, 2024 Apr 19.
Article in English | MEDLINE | ID: mdl-38669573

ABSTRACT

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


Subject(s)
Aedes , Dengue Virus , Mosquito Vectors , Symbiosis , Zika Virus , Animals , Aedes/microbiology , Aedes/virology , Dengue Virus/physiology , Mosquito Vectors/virology , Mosquito Vectors/microbiology , Zika Virus/physiology , Dengue/transmission , Dengue/virology , Dengue/prevention & control , Gastrointestinal Microbiome , Acetobacteraceae/physiology , Female , Viral Envelope Proteins/metabolism , Viral Envelope Proteins/genetics , Flavivirus/physiology , Flavivirus/genetics , Zika Virus Infection/transmission , Zika Virus Infection/virology
18.
J Travel Med ; 31(4)2024 Jun 03.
Article in English | MEDLINE | ID: mdl-38630887

ABSTRACT

BACKGROUND: The international flight network creates multiple routes by which pathogens can quickly spread across the globe. In the early stages of infectious disease outbreaks, analyses using flight passenger data to identify countries at risk of importing the pathogen are common and can help inform disease control efforts. A challenge faced in this modelling is that the latest aviation statistics (referred to as contemporary data) are typically not immediately available. Therefore, flight patterns from a previous year are often used (referred to as historical data). We explored the suitability of historical data for predicting the spatial spread of emerging epidemics. METHODS: We analysed monthly flight passenger data from the International Air Transport Association to assess how baseline air travel patterns were affected by outbreaks of Middle East respiratory syndrome (MERS), Zika and severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) over the past decade. We then used a stochastic discrete time susceptible-exposed-infected-recovered (SEIR) metapopulation model to simulate the global spread of different pathogens, comparing how epidemic dynamics differed in simulations based on historical and contemporary data. RESULTS: We observed local, short-term disruptions to air travel from South Korea and Brazil for the MERS and Zika outbreaks we studied, whereas global and longer-term flight disruptions occurred during the SARS-CoV-2 pandemic. For outbreak events that were accompanied by local, small and short-term changes in air travel, epidemic models using historical flight data gave similar projections of the timing and locations of disease spread as when using contemporary flight data. However, historical data were less reliable to model the spread of an atypical outbreak such as SARS-CoV-2, in which there were durable and extensive levels of global travel disruption. CONCLUSION: The use of historical flight data as a proxy in epidemic models is an acceptable practice, except in rare, large epidemics that lead to substantial disruptions to international travel.


Subject(s)
Air Travel , COVID-19 , Disease Outbreaks , SARS-CoV-2 , Zika Virus Infection , Humans , Air Travel/statistics & numerical data , COVID-19/epidemiology , COVID-19/transmission , COVID-19/prevention & control , Zika Virus Infection/epidemiology , Zika Virus Infection/transmission , Coronavirus Infections/epidemiology , Coronavirus Infections/transmission , Coronavirus Infections/prevention & control , Communicable Diseases/epidemiology , Communicable Diseases/transmission , Travel/statistics & numerical data , Aircraft , Global Health
19.
Stem Cells Transl Med ; 13(5): 448-453, 2024 May 14.
Article in English | MEDLINE | ID: mdl-38521608

ABSTRACT

BACKGROUND: Cord blood units (CBUs) that are ineligible for licensure due to incomplete compliance with FDA recommendations may be used for hematopoietic stem cell transplantation under urgent medical need and an Investigational Drug Application. The largest reason for CBU donor ineligibility is Zika virus (ZIKV) risk. The study's objective was to analyze the impact of current FDA recommendations for ZIKA risk on a large public cord blood bank and propose updated recommendations. METHODS: We performed a retrospective analysis of Carolinas Cord Blood Bank (CCBB), an FDA licensed public CBB, using data from January 1, 2016 to November 21, 2023 and compared FDA recommendations for transfusion transmitted infections (TTI) for blood products and relevant communicable disease agents or diseases for human cell, tissue, or cellular or tissue-based products (HCT/Ps). RESULTS: CCBB: 9057 (84.3% licensed) CBUs were banked. 984/1682 (58.5%) of unlicensed CBUs had ZIKV risk. 22.0% of CBUs with ZIKV risk were from Hispanic parents, compared to 16.1% of all units. 31 of IND CBUs (11 due to ZIKV risk without reported ZIKV transmission) were safely infused. FDA Guidance: HCT/P ZIKV, HIV, and vCJD recommendations have not been updated since 2018 in contrast to FDA removal of ZIKV as a relevant TTI in 2021 and updating HIV and vCJD guidance related to TTI in 2023 and 2022, respectively. DISCUSSION: The FDA should consider new data to revise the HCT/P donor eligibility recommendations, which will increase the number of eligible HCT/P donors, and potentially improve access to therapies for a more diverse patient population.


Subject(s)
Blood Banks , Fetal Blood , United States Food and Drug Administration , Zika Virus Infection , Humans , United States , Zika Virus Infection/transmission , Fetal Blood/virology , Blood Banks/standards , Zika Virus , Retrospective Studies , Female , Male
20.
Trop Med Int Health ; 29(5): 414-423, 2024 May.
Article in English | MEDLINE | ID: mdl-38469931

ABSTRACT

OBJECTIVES: Arboviruses, such as dengue (DENV), zika (ZIKV), and chikungunya (CHIKV), constitute a growing urban public health threat. Focusing on Aedes aegypti mosquitoes, their primary vectors, is crucial for mitigation. While traditional immature-stage mosquito surveillance has limitations, capturing adult mosquitoes through traps yields more accurate data on disease transmission. However, deploying traps presents logistical and financial challenges, demonstrating effective temporal predictions but lacking spatial accuracy. Our goal is to identify smaller representative areas within cities to enhance the early warning system for DENV outbreaks. METHODS: We created Sentinel Geographic Units (SGUs), smaller areas of 1 km2 within each stratum, larger areas, with the aim of aligning the Trap Positivity Index (TPI) and Adult Density Index (ADI) with their respective strata. We conducted a two-step evaluation of SGUs. First, we examined the equivalence of TPI and ADI between SGUs and strata from January 2017 to July 2022. Second, we assessed the ability of SGU's TPI and ADI to predict DENV outbreaks in comparison to Foz do Iguaçu's Early-Warning System, which forecasts outbreaks up to 4 weeks ahead. Spatial and temporal analyses were carried out, including data interpolation and model selection based on Akaike information criteria (AIC). RESULTS: Entomological indicators produced in small SGUs can effectively replace larger sentinel areas to access dengue outbreaks. Based on historical data, the best predictive capability is achieved 2 weeks after infestation verification. Implementing the SGU strategy with more frequent sampling can provide more precise space-time estimates and enhance dengue control. CONCLUSIONS: The implementation of SGUs offers an efficient way to monitor mosquito populations, reducing the need for extensive resources. This approach has the potential to improve dengue transmission management and enhance the public health response in endemic cities.


Subject(s)
Aedes , Cities , Dengue , Mosquito Vectors , Animals , Aedes/virology , Brazil/epidemiology , Dengue/epidemiology , Dengue/transmission , Dengue/prevention & control , Humans , Disease Outbreaks/prevention & control , Zika Virus Infection/epidemiology , Zika Virus Infection/prevention & control , Zika Virus Infection/transmission , Mosquito Control/methods
SELECTION OF CITATIONS
SEARCH DETAIL