Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 314
Filter
1.
Pan Afr Med J ; 47: 120, 2024.
Article in English | MEDLINE | ID: mdl-38828420

ABSTRACT

Introduction: Aedes albopictus, like Aedes aegypti, is a virulent vector of arboviruses especially the well-documented spread of yellow fever around the world. Although yellow fever is prevalent in Nigeria, there is a paucity of information in the Niger Delta region on the distribution of Aedes mosquito vectors and molecular detection of the virus in infected mosquitoes. This study sampled Aedes mosquitoes around houses associated with farms from four communities (Otolokpo, Ute-Okpu, Umunede, and Ute Alohen) in Ika North-East Local Government Area of Delta State, Nigeria. Methods: various sampling methods were used in Aedes mosquito collection to test their efficacy in the survey. Mosquitoes in holding cages were killed by freezing and morphologically identified. A pool of 15 mosquitoes per Eppendorf tube was preserved in RNAi later for yellow fever virus screening. Two samples were molecularly screened for each location. Results: seven hundred and twenty-five (725) mosquitoes were obtained from the various traps. The mean abundance of the mosquitoes was highest in m-HLC (42.9) compared to the mosquitoes sampled using other techniques (p<0.0001). The mean abundance of mosquitoes was lowest in Center for Disease Control (CDC) light traps without attractant (0.29). No yellow fever virus strain was detected in all the mosquitoes sampled at the four locations. Conclusion: this study suggests that Aedes albopictus are the mosquitoes commonly biting around houses associated with farms. More so, yellow fever virus was not detected in the mosquitoes probably due to the mass vaccination exercise that was carried out the previous year in the study area. More studies are required using the m-HLC to determine the infection rate in this endemic area.


Subject(s)
Aedes , Mosquito Vectors , Yellow Fever , Yellow fever virus , Animals , Aedes/virology , Nigeria , Yellow fever virus/isolation & purification , Mosquito Vectors/virology , Yellow Fever/transmission , Yellow Fever/epidemiology , Yellow Fever/virology , Humans
2.
Int J Biol Macromol ; 269(Pt 2): 132169, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38723801

ABSTRACT

In our study, we developed a point of care electrochemical biosensing platform based on the functionalized cysteine-positioned gold electrode to diagnose yellow fever disease from human plasma samples. The developed platform underwent characterization through diverse methods encompassing cyclic voltammetry, electrochemical impedance spectroscopy, scanning electron microscopy, energy dispersive X-ray spectroscopy, and density-functional theory. The capacitive interaction between yellow fever virus non-structural antigen and antibody gave a cathodic signal at approximately -260 mV, and increased in proportion to the amount of non-structural antibody. The created electrochemical biosensor has an ability to detect 96 ag/mL of the yellow fever non-structural antibody with an extensive analytical range varied from 0.1 fg/mL to 1 µg/mL. The interference effects of various substances that could be found in human plasma, and the performance of the method were examined from the point of recovery and relative standard deviation for human plasma samples; hereby, the results confirmed the unprecedented selectivity and accuracy of the proposed method.


Subject(s)
Biosensing Techniques , Electrochemical Techniques , Viral Nonstructural Proteins , Yellow Fever , Humans , Biosensing Techniques/methods , Yellow Fever/diagnosis , Yellow Fever/blood , Yellow Fever/immunology , Yellow Fever/virology , Viral Nonstructural Proteins/immunology , Viral Nonstructural Proteins/blood , Electrochemical Techniques/methods , Point-of-Care Systems , Yellow fever virus/immunology , Density Functional Theory , Electrodes , Antibodies, Viral/blood , Antibodies, Viral/immunology , Gold/chemistry
3.
J Virol ; 98(5): e0151623, 2024 May 14.
Article in English | MEDLINE | ID: mdl-38567951

ABSTRACT

The non-human primate (NHP) model (specifically rhesus and cynomolgus macaques) has facilitated our understanding of the pathogenic mechanisms of yellow fever (YF) disease and allowed the evaluation of the safety and efficacy of YF-17D vaccines. However, the accuracy of this model in mimicking vaccine-induced immunity in humans remains to be fully determined. We used a systems biology approach to compare hematological, biochemical, transcriptomic, and innate and antibody-mediated immune responses in cynomolgus macaques and human participants following YF-17D vaccination. Immune response progression in cynomolgus macaques followed a similar course as in adult humans but with a slightly earlier onset. Yellow fever virus neutralizing antibody responses occurred earlier in cynomolgus macaques [by Day 7[(D7)], but titers > 10 were reached in both species by D14 post-vaccination and were not significantly different by D28 [plaque reduction neutralization assay (PRNT)50 titers 3.6 Log vs 3.5 Log in cynomolgus macaques and human participants, respectively; P = 0.821]. Changes in neutrophils, NK cells, monocytes, and T- and B-cell frequencies were higher in cynomolgus macaques and persisted for 4 weeks versus less than 2 weeks in humans. Low levels of systemic inflammatory cytokines (IL-1RA, IL-8, MIP-1α, IP-10, MCP-1, or VEGF) were detected in either or both species but with no or only slight changes versus baseline. Similar changes in gene expression profiles were elicited in both species. These included enriched and up-regulated type I IFN-associated viral sensing, antiviral innate response, and dendritic cell activation pathways D3-D7 post-vaccination in both species. Hematological and blood biochemical parameters remained relatively unchanged versus baseline in both species. Low-level YF-17D viremia (RNAemia) was transiently detected in some cynomolgus macaques [28% (5/18)] but generally absent in humans [except one participant (5%; 1/20)].IMPORTANCECynomolgus macaques were confirmed as a valid surrogate model for replicating YF-17D vaccine-induced responses in humans and suggest a key role for type I IFN.


Subject(s)
Macaca fascicularis , Models, Animal , Yellow Fever Vaccine , Animals , Female , Humans , Male , Antibodies, Neutralizing/blood , Antibodies, Neutralizing/immunology , Antibodies, Viral/blood , Antibodies, Viral/immunology , Immunity, Innate , Systems Biology/methods , Vaccination , Yellow Fever/prevention & control , Yellow Fever/immunology , Yellow Fever/virology , Yellow Fever Vaccine/immunology , Yellow fever virus/immunology
4.
Microbiol Spectr ; 12(5): e0370323, 2024 May 02.
Article in English | MEDLINE | ID: mdl-38511952

ABSTRACT

Between 2016 and 2018, Brazil experienced major sylvatic yellow fever (YF) outbreaks that caused hundreds of casualties, with Minas Gerais (MG) being the most affected state. These outbreaks provided a unique opportunity to assess the immune response triggered by the wild-type (WT) yellow fever virus (YFV) in humans. The plaque reduction neutralization test (PRNT) is currently the standard method to assess the humoral immune response to YFV by measuring neutralizing antibodies (nAbs). The present study aimed to evaluate the humoral immune response of patients from the 2017-2018 sylvatic YF outbreak in MG with different disease outcomes by using PRNTs with a WT YFV strain, isolated from the 2017-2018 outbreak, and a vaccine YFV strain. Samples from naturally infected YF patients were tested, in comparison with healthy vaccinees. Results showed that both groups presented different levels of nAb against the WT and vaccine strains, and the levels of neutralization against the strains varied homotypically and heterotypically. Results based on the geometric mean titers (GMTs) suggest that the humoral immune response after a natural infection of YFV can reach higher levels than that induced by vaccination (GMT of patients against WT YFV compared to GMT of vaccinees, P < 0.0001). These findings suggest that the humoral immune responses triggered by the vaccine and WT strains of YFV are different, possibly due to genetic and antigenic differences between these viruses. Therefore, current means of assessing the immune response in naturally infected YF individuals and immunological surveillance methods in areas with intense viral circulation may need to be updated.IMPORTANCEYellow fever is a deadly febrile disease caused by the YFV. Despite the existence of effective vaccines, this disease still represents a public health concern worldwide. Much is known about the immune response against the vaccine strains of the YFV, but recent studies have shown that it differs from that induced by WT strains. The extent of this difference and the mechanisms behind it are still unclear. Thus, studies aimed to better understand the immune response against this virus are relevant and necessary. The present study evaluated levels of neutralizing antibodies of yellow fever patients from recent outbreaks in Brazil, in comparison with healthy vaccinees, using plaque reduction neutralization tests with WT and vaccine YFV strains. Results showed that the humoral immune response in naturally infected patients was higher than that induced by vaccination, thus providing new insights into the immune response triggered against these viruses.


Subject(s)
Antibodies, Neutralizing , Antibodies, Viral , Disease Outbreaks , Immunity, Humoral , Yellow Fever Vaccine , Yellow Fever , Yellow fever virus , Yellow Fever/immunology , Yellow Fever/epidemiology , Yellow Fever/virology , Humans , Brazil/epidemiology , Yellow fever virus/immunology , Yellow fever virus/genetics , Antibodies, Neutralizing/blood , Antibodies, Neutralizing/immunology , Antibodies, Viral/blood , Antibodies, Viral/immunology , Male , Yellow Fever Vaccine/immunology , Female , Adult , Middle Aged , Vaccination , Neutralization Tests , Young Adult , Aged , Adolescent
5.
Emerg Infect Dis ; 29(9): 1818-1826, 2023 09.
Article in English | MEDLINE | ID: mdl-37610174

ABSTRACT

Yellow fever virus, transmitted by infected Aedes spp. mosquitoes, causes an acute viral hemorrhagic disease. During October 2021-February 2022, a yellow fever outbreak in some communities in Ghana resulted in 70 confirmed cases with 35 deaths (case-fatality rate 50%). The outbreak started in a predominantly unvaccinated nomadic community in the Savannah region, from which 65% of the cases came. The molecular amplification methods we used for diagnosis produced full-length DNA sequences from 3 confirmed cases. Phylogenetic analysis characterized the 3 sequences within West Africa genotype II; strains shared a close homology with sequences from Cote d'Ivoire and Senegal. We deployed more sensitive advanced molecular diagnostic techniques, which enabled earlier detection, helped control spread, and improved case management. We urge increased efforts from health authorities to vaccinate vulnerable groups in difficult-to-access areas and to educate the population about potential risks for yellow fever infections.


Subject(s)
Yellow Fever , Yellow fever virus , Yellow fever virus/classification , Yellow fever virus/isolation & purification , Yellow Fever/virology , Disease Outbreaks , Ghana/epidemiology , Humans , Phylogeny , Sequence Analysis, RNA , RNA, Viral/analysis
6.
J Virol ; 97(4): e0194922, 2023 04 27.
Article in English | MEDLINE | ID: mdl-37017533

ABSTRACT

Genome cyclization is essential for viral RNA (vRNA) replication of the vertebrate-infecting flaviviruses, and yet its regulatory mechanisms are not fully understood. Yellow fever virus (YFV) is a notorious pathogenic flavivirus. Here, we demonstrated that a group of cis-acting RNA elements in YFV balance genome cyclization to govern efficient vRNA replication. It was shown that the downstream of the 5'-cyclization sequence hairpin (DCS-HP) is conserved in the YFV clade and is important for efficient YFV propagation. By using two different replicon systems, we found that the function of the DCS-HP is determined primarily by its secondary structure and, to a lesser extent, by its base-pair composition. By combining in vitro RNA binding and chemical probing assays, we found that the DCS-HP orchestrates the balance of genome cyclization through two different mechanisms, as follows: the DCS-HP assists the correct folding of the 5' end in a linear vRNA to promote genome cyclization, and it also limits the overstabilization of the circular form through a potential crowding effect, which is influenced by the size and shape of the DCS-HP structure. We also provided evidence that an A-rich sequence downstream of the DCS-HP enhances vRNA replication and contributes to the regulation of genome cyclization. Interestingly, diversified regulatory mechanisms of genome cyclization, involving both the downstream of the 5'-cyclization sequence (CS) and the upstream of the 3'-CS elements, were identified among different subgroups of the mosquito-borne flaviviruses. In summary, our work highlighted how YFV precisely controls the balance of genome cyclization to ensure viral replication. IMPORTANCE Yellow fever virus (YFV), the prototype of the Flavivirus genus, can cause devastating yellow fever disease. Although it is preventable by vaccination, there are still tens of thousands of yellow fever cases per year, and no approved antiviral medicine is available. However, the understandings about the regulatory mechanisms of YFV replication are obscure. In this study, by a combination of bioinformatics, reverse genetics, and biochemical approaches, it was shown that the downstream of the 5'-cyclization sequence hairpin (DCS-HP) promotes efficient YFV replication by modulating the conformational balance of viral RNA. Interestingly, we found specialized combinations for the downstream of the 5'-cyclization sequence (CS) and upstream of the 3'-CS elements in different groups of the mosquito-borne flaviviruses. Moreover, possible evolutionary relationships among the various downstream of the 5'-CS elements were implied. This work highlighted the complexity of RNA-based regulatory mechanisms in the flaviviruses and will facilitate the design of RNA structure-targeted antiviral therapies.


Subject(s)
Virus Replication , Yellow fever virus , Animals , Humans , Cyclization , RNA, Viral/metabolism , Virus Replication/genetics , Yellow Fever/virology , Yellow fever virus/metabolism , Genome, Viral/genetics , Cell Line , Cricetinae , Mesocricetus , A549 Cells
7.
PLoS One ; 17(2): e0262149, 2022.
Article in English | MEDLINE | ID: mdl-35139078

ABSTRACT

There is an urgent need for better diagnostic and analytical methods for vaccine research and infection control in virology. This has been highlighted by recently emerging viral epidemics and pandemics (Zika, SARS-CoV-2), and recurring viral outbreaks like the yellow fever outbreaks in Angola and the Democratic Republic of Congo (2016) and in Brazil (2016-2018). Current assays to determine neutralising activity against viral infections in sera are costly in time and equipment and suffer from high variability. Therefore, both basic infection research and diagnostic population screenings would benefit from improved methods to determine virus-neutralising activity in patient samples. Here we describe a robust, objective, and scalable Fluorescence Reduction Neutralisation Test (FluoRNT) for yellow fever virus, relying on flow cytometric detection of cells infected with a fluorescent Venus reporter containing variant of the yellow fever vaccine strain 17D (YF-17D-Venus). It accurately measures neutralising antibody titres in human serum samples within as little as 24 h. Samples from 32 vaccinees immunised with YF-17D were tested for neutralising activity by both a conventional focus reduction neutralisation test (FRNT) and FluoRNT. Both types of tests proved to be equally reliable for the detection of neutralising activity, however, FluoRNT is significantly more precise and reproducible with a greater dynamic range than conventional FRNT. The FluoRNT assay protocol is substantially faster, easier to control, and cheaper in per-assay costs. FluoRNT additionally reduces handling time minimising exposure of personnel to patient samples. FluoRNT thus brings a range of desirable features that can accelerate and standardise the measurement of neutralising anti-yellow fever virus antibodies. It could be used in applications ranging from vaccine testing to large cohort studies in systems virology and vaccinology. We also anticipate the potential to translate the methodology and analysis of FluoRNT to other flaviviruses such as West Nile, Dengue and Zika or to RNA viruses more generally.


Subject(s)
Antibodies, Neutralizing/immunology , Yellow Fever/immunology , Yellow fever virus/immunology , Animals , Antibodies, Neutralizing/blood , Antibodies, Viral/blood , Antibodies, Viral/immunology , Chlorocebus aethiops , Fluorescence , Humans , Neutralization Tests/economics , Neutralization Tests/methods , Vero Cells , Yellow Fever/blood , Yellow Fever/virology
8.
PLoS Negl Trop Dis ; 16(1): e0010019, 2022 Jan.
Article in English | MEDLINE | ID: mdl-34995277

ABSTRACT

BACKGROUND: Yellow fever (YF) is an arboviral disease which is endemic to Brazil due to a sylvatic transmission cycle maintained by infected mosquito vectors, non-human primate (NHP) hosts, and humans. Despite the existence of an effective vaccine, recent sporadic YF epidemics have underscored concerns about sylvatic vector surveillance, as very little is known about their spatial distribution. Here, we model and map the environmental suitability of YF's main vectors in Brazil, Haemagogus spp. and Sabethes spp., and use human population and NHP data to identify locations prone to transmission and spillover risk. METHODOLOGY/PRINCIPAL FINDINGS: We compiled a comprehensive set of occurrence records on Hg. janthinomys, Hg. leucocelaenus, and Sabethes spp. from 1991-2019 using primary and secondary data sources. Linking these data with selected environmental and land-cover variables, we adopted a stacked regression ensemble modelling approach (elastic-net regularized GLM, extreme gradient boosted regression trees, and random forest) to predict the environmental suitability of these species across Brazil at a 1 km x 1 km resolution. We show that while suitability for each species varies spatially, high suitability for all species was predicted in the Southeastern region where recent outbreaks have occurred. By integrating data on NHP host reservoirs and human populations, our risk maps further highlight municipalities within the region that are prone to transmission and spillover. CONCLUSIONS/SIGNIFICANCE: Our maps of sylvatic vector suitability can help elucidate potential locations of sylvatic reservoirs and be used as a tool to help mitigate risk of future YF outbreaks and assist in vector surveillance. Furthermore, at-risk regions identified from our work could help disease control and elucidate gaps in vaccination coverage and NHP host surveillance.


Subject(s)
Culicidae/virology , Mosquito Vectors/virology , Yellow Fever/transmission , Yellow fever virus/physiology , Animals , Brazil/epidemiology , Host-Pathogen Interactions , Species Specificity , Yellow Fever/epidemiology , Yellow Fever/virology
9.
PLoS Negl Trop Dis ; 16(1): e0009937, 2022 Jan.
Article in English | MEDLINE | ID: mdl-35073319

ABSTRACT

Yellow fever virus (YFV) is a zoonotic pathogen re-emerging in parts of the world, causing a viral hemorrhagic fever associated with high mortality rates. While an effective vaccine is available, having an effective antiviral against YFV is critical against unexpected outbreaks, or when vaccination is not recommended. We have previously identified AT-281, the free base of AT-752, an orally available double prodrug of a guanosine nucleotide analog, as a potent inhibitor of YFV in vitro, with a 50% effective concentration (EC50) of 0.31 µM. In hamsters infected with YFV (Jimenez strain), viremia rose about 4 log10-fold and serum alanine aminotransferase (ALT) 2-fold compared to sham-infected animals. Treatment with 1000 mg/kg AT-752 for 7 days, initiated 4 h prior to viral challenge, reduced viremia to below the limit of detection by day 4 post infection (pi) and returned ALT to normal levels by day 6 pi. When treatment with AT-752 was initiated 2 days pi, the virus titer and ALT dropped >2 log10 and 53% by day 4 and 6 pi, respectively. In addition, at 21 days pi, 70-100% of the infected animals in the treatment groups survived compared to 0% of the untreated group (p<0.001). Moreover, in vivo formation of the active triphosphate metabolite AT-9010 was measured in the animal tissues, with the highest concentrations in liver and kidney, organs that are vulnerable to the virus. The demonstrated in vivo activity of AT-752 suggests that it is a promising compound for clinical development in the treatment of YFV infection.


Subject(s)
Antiviral Agents/pharmacology , Guanosine/analogs & derivatives , Prodrugs/pharmacology , Yellow Fever/drug therapy , Yellow fever virus/drug effects , Animals , Antiviral Agents/chemistry , Antiviral Agents/pharmacokinetics , Chlorocebus aethiops , Cricetinae , Female , Male , Mesocricetus , Prodrugs/chemistry , Prodrugs/pharmacokinetics , Vero Cells , Viremia , Yellow Fever/virology
10.
PLoS Negl Trop Dis ; 15(11): e0009907, 2021 11.
Article in English | MEDLINE | ID: mdl-34735450

ABSTRACT

Zika virus (ZIKV) emerged as an important infectious disease agent in Brazil in 2016. Infection usually leads to mild symptoms, but severe congenital neurological disorders and Guillain-Barré syndrome have been reported following ZIKV exposure. Creating an effective vaccine against ZIKV is a public health priority. We describe the protective effect of an already licensed attenuated yellow fever vaccine (YFV, 17DD) in type-I interferon receptor knockout mice (A129) and immunocompetent BALB/c and SV-129 (A129 background) mice infected with ZIKV. YFV vaccination provided protection against ZIKV, with decreased mortality in A129 mice, a reduction in the cerebral viral load in all mice, and weight loss prevention in BALB/c mice. The A129 mice that were challenged two and three weeks after the first dose of the vaccine were fully protected, whereas partial protection was observed five weeks after vaccination. In all cases, the YFV vaccine provoked a substantial decrease in the cerebral viral load. YFV immunization also prevented hippocampal synapse loss and microgliosis in ZIKV-infected mice. Our vaccine model is T cell-dependent, with AG129 mice being unable to tolerate immunization (vaccination is lethal in this mouse model), indicating the importance of IFN-γ in immunogenicity. To confirm the role of T cells, we immunized nude mice that we demonstrated to be very susceptible to infection. Immunization with YFV and challenge 7 days after booster did not protect nude mice in terms of weight loss and showed partial protection in the survival curve. When we evaluated the humoral response, the vaccine elicited significant antibody titers against ZIKV; however, it showed no neutralizing activity in vitro and in vivo. The data indicate that a cell-mediated response promotes protection against cerebral infection, which is crucial to vaccine protection, and it appears to not necessarily require a humoral response. This protective effect can also be attributed to innate factors, but more studies are needed to strengthen this hypothesis. Our findings open the way to using an available and inexpensive vaccine for large-scale immunization in the event of a ZIKV outbreak.


Subject(s)
Yellow Fever Vaccine/administration & dosage , Zika Virus Infection/prevention & control , Zika Virus/physiology , Animals , Antibodies, Viral/immunology , Chlorocebus aethiops , Disease Models, Animal , Female , Humans , Immunity, Cellular , Interferon-gamma/immunology , Mice , Mice, Inbred BALB C , T-Lymphocytes/immunology , Vaccination , Vero Cells , Yellow Fever/virology , Yellow fever virus/genetics , Yellow fever virus/immunology , Zika Virus/genetics , Zika Virus/immunology , Zika Virus Infection/immunology , Zika Virus Infection/virology
11.
Emerg Microbes Infect ; 10(1): 2279-2290, 2021 Dec.
Article in English | MEDLINE | ID: mdl-34792431

ABSTRACT

The live-attenuated yellow fever 17D (YF17D) vaccine is one of the most efficacious human vaccines and also employed as a vector for novel vaccines. However, in the lack of appropriate immunocompetent small animal models, mechanistic insight in YF17D-induced protective immunity remains limited. To better understand YF17D vaccination and to identify a suitable mouse model, we evaluated the immunogenicity and protective efficacy of YF17D in five complementary mouse models, i.e. wild-type (WT) BALB/c, C57BL/6, IFN-α/ß receptor (IFNAR-/-) deficient mice, and in WT mice in which type I IFN signalling was temporally ablated by an IFNAR blocking (MAR-1) antibody. Alike in IFNAR-/- mice, YF17D induced in either WT mice strong humoral immune responses dominated by IgG2a/c isotype (Th1 type) antibodies, yet only when IFNAR was blocked. Vigorous cellular immunity characterized by CD4+ T-cells producing IFN-γ and TNF-α were mounted in MAR-1 treated C57BL/6 and in IFNAR-/- mice. Surprisingly, vaccine-induced protection was largely mouse model dependent. Full protection against lethal intracranial challenge and a massive reduction of virus loads was conferred already by a minimal dose of 2 PFU YF17D in BALB/c and IFNAR-/- mice, but not in C57BL/6 mice. Correlation analysis of infection outcome with pre-challenge immunological markers indicates that YFV-specific IgG might suffice for protection, even in the absence of detectable levels of neutralizing antibodies. Finally, we propose that, in addition to IFNAR-/- mice, C57BL/6 mice with temporally blocked IFN-α/ß receptors represent a promising immunocompetent mouse model for the study of YF17D-induced immunity and evaluation of YF17D-derived vaccines.


Subject(s)
Yellow Fever Vaccine/administration & dosage , Yellow Fever Vaccine/immunology , Yellow Fever/prevention & control , Yellow fever virus/immunology , Animals , Antibodies, Neutralizing/immunology , Antibodies, Viral/immunology , Disease Models, Animal , Female , Humans , Immunity, Cellular , Immunity, Humoral , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , T-Lymphocytes/immunology , Vaccines, Attenuated/administration & dosage , Vaccines, Attenuated/genetics , Vaccines, Attenuated/immunology , Yellow Fever/immunology , Yellow Fever/virology , Yellow Fever Vaccine/genetics , Yellow fever virus/genetics
12.
Nat Commun ; 12(1): 5374, 2021 09 10.
Article in English | MEDLINE | ID: mdl-34508072

ABSTRACT

The mosquito Aedes aegypti is the principal vector for arboviruses including dengue/yellow fever, chikungunya, and Zika virus, infecting hundreds of millions of people annually. Unfortunately, traditional control methodologies are insufficient, so innovative control methods are needed. To complement existing measures, here we develop a molecular genetic control system termed precision-guided sterile insect technique (pgSIT) in Aedes aegypti. PgSIT uses a simple CRISPR-based approach to generate flightless females and sterile males that are deployable at any life stage. Supported by mathematical models, we empirically demonstrate that released pgSIT males can compete, suppress, and even eliminate mosquito populations. This platform technology could be used in the field, and adapted to many vectors, for controlling wild populations to curtail disease in a safe, confinable, and reversible manner.


Subject(s)
Aedes/virology , Infertility, Male/veterinary , Mosquito Control/methods , Mosquito Vectors/virology , Aedes/genetics , Animals , Animals, Genetically Modified , Arboviruses , Chikungunya Fever/prevention & control , Chikungunya Fever/transmission , Chikungunya Fever/virology , Dengue/prevention & control , Dengue/transmission , Dengue/virology , Female , Humans , Infertility, Male/genetics , Male , Models, Biological , Mosquito Vectors/genetics , Yellow Fever/prevention & control , Yellow Fever/transmission , Yellow Fever/virology , Zika Virus , Zika Virus Infection/prevention & control , Zika Virus Infection/transmission , Zika Virus Infection/virology
13.
Sci Rep ; 11(1): 16277, 2021 08 11.
Article in English | MEDLINE | ID: mdl-34381111

ABSTRACT

We investigated the sylvatic yellow fever (SYF) diffusion process in São Paulo (SP) between 2016 and 2019. We developed an ecological study of SYF through autochthonous human cases and epizootics of non-human primates (NHPs) that were spatiotemporally evaluated. We used kriging to obtain maps with isochrones representative of the evolution of the outbreak and characterized its diffusion pattern. We confirmed 648 human cases of SYF in SP, with 230 deaths and 843 NHP epizootics. Two outbreak waves were identified: one from West to East (2016 and 2017), and another from the Campinas region to the municipalities bordering Rio de Janeiro, Minas Gerais, and Paraná and those of the SP coast (2017-2019). The SYF outbreak diffusion process was by contagion. The disease did not exhibit jumps between municipalities, indicating that the mosquitoes and NHPs were responsible for transmitting the virus. There were not enough vaccines to meet the population at risk; hence, health authorities used information about the epizootic occurrence in NHPs in forest fragments to identify priority populations for vaccination.


Subject(s)
Yellow Fever/epidemiology , Animals , Brazil/epidemiology , Disease Outbreaks/prevention & control , Humans , Primates/virology , Vaccination/methods , Yellow Fever/immunology , Yellow Fever/virology , Yellow fever virus/immunology , Zoonoses/epidemiology , Zoonoses/immunology , Zoonoses/virology
14.
Emerg Microbes Infect ; 10(1): 1739-1750, 2021 Dec.
Article in English | MEDLINE | ID: mdl-34379047

ABSTRACT

Yellow fever virus (YFV) is a re-emerging flavivirus, which can lead to severe clinical manifestations and high mortality, with no specific antiviral therapies available. The live-attenuated yellow fever vaccine 17D (YF17D) has been widely used for over eighty years. However, the emergence of yellow fever vaccine-associated viscerotropic disease (YFL-AVD) and yellow fever vaccine-associated neurotropic disease (YFL-AND) raised non-negligible concerns. Additionally, the attenuation mechanism of YF17D is still unclear. Thus, the development of convenient models is crucial to understand the mechanisms behind YF17D attenuation and its adverse effects. In this work, we generated a reporter YF17D expressing nano-luciferase (NLuc). In vitro and in vivo characterization demonstrated that the NLuc-YF17D shared similar biological properties with its parental strain and the NLuc activity can reflect viral infectivity reliably. Combined with in vivo bioluminescence imaging, a series of mice models of YF17D infection was established, which will be useful for the evaluation of antiviral medicines and novel vaccine candidates. Especially, we demonstrated that intraperitoneally (i.p.) infection of NLuc-YF17D in type I interferon receptor-deficient mice A129 resulted in outcomes resembling YEL-AVD and YEL-AND, evidenced by viral replication in multiple organs and invasion of the central neuronal system. Finally, in vitro and in vivo assays based on this reporter virus were established to evaluate the antiviral activities of validated antiviral agents. In conclusion, the bioluminescent reporter virus described herein provides a powerful platform to study YF17D attenuation and vaccine-associated diseases as well as to develop novel countermeasures against YFV.


Subject(s)
Luminescent Measurements/methods , Yellow Fever/virology , Yellow fever virus/metabolism , Animals , Cell Line , Imaging, Three-Dimensional/methods , Luciferases/genetics , Luciferases/metabolism , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Virus Replication , Yellow fever virus/genetics
15.
Viruses ; 13(8)2021 07 28.
Article in English | MEDLINE | ID: mdl-34452343

ABSTRACT

Yellow fever virus remains a major threat in low resource countries in South America and Africa despite the existence of an effective vaccine. In Senegal and particularly in the eastern part of the country, periodic sylvatic circulation has been demonstrated with varying degrees of impact on populations in perpetual renewal. We report an outbreak that occurred from October 2020 to February 2021 in eastern Senegal, notified and managed through the synergistic effort yellow fever national surveillance implemented by the Senegalese Ministry of Health in collaboration with the World Health Organization, the countrywide 4S network set up by the Ministry of Health, the Institut Pasteur de Dakar, and the surveillance of arboviruses and hemorrhagic fever viruses in human and vector populations implemented since mid 2020 in eastern Senegal. Virological analyses highlighted the implication of sylvatic mosquito species in virus transmission. Genomic analysis showed a close relationship between the circulating strain in eastern Senegal, 2020, and another one from the West African lineage previously detected and sequenced two years ago from an unvaccinated Dutch traveler who visited the Gambia and Senegal before developing signs after returning to Europe. Moreover, genome analysis identified a 6-nucleotide deletion in the variable domain of the 3'UTR with potential impact on the biology of the viral strain that merits further investigations. Integrated surveillance of yellow fever virus but also of other arboviruses of public health interest is crucial in an ecosystem such as eastern Senegal.


Subject(s)
Yellow Fever/epidemiology , Yellow Fever/virology , Yellow fever virus/physiology , Adolescent , Adult , Aedes/classification , Aedes/physiology , Aedes/virology , Amino Acid Sequence , Animals , Child , Disease Outbreaks , Female , Humans , Male , Mosquito Vectors/classification , Mosquito Vectors/physiology , Mosquito Vectors/virology , Phylogeny , Senegal/epidemiology , Sequence Alignment , Viral Proteins/chemistry , Viral Proteins/genetics , Yellow Fever/transmission , Yellow fever virus/classification , Yellow fever virus/genetics , Yellow fever virus/isolation & purification , Young Adult
16.
Pan Afr Med J ; 38: 248, 2021.
Article in French | MEDLINE | ID: mdl-34104296

ABSTRACT

Yellow fever (YF) is a viral haemorrhagic fever caused by yellow fever virus transmitted by Aedes mosquitoes. Since 2013, in Chad, four cases of yellow fever have been detected and confirmed as part of the national fever surveillance program. We here report the last clinical case confirmed in the health district of Lai. The patient was a 57-year-old man with no significant medical and surgical history and unknown immunisation status. He consulted on April 21st, 2020 for fever, moderate to low abundance jaundice and epistaxis (nosebleed) and painful hepatomegaly. Paraclinical examinations, such as RT-PCR, objectified yellow fever virus in post-mortem tissue sample. Thus, confirmed yellow fever cases in this district, the low level of vaccination coverage, the circulation of the virus and the presence of vector in the country should warn of a real threat of reemergence of yellow fever in Chad.


Subject(s)
Yellow Fever/diagnosis , Yellow fever virus/isolation & purification , Aedes/virology , Animals , Chad , Humans , Male , Middle Aged , Mosquito Vectors/virology , Recurrence , Yellow Fever/transmission , Yellow Fever/virology , Yellow Fever Vaccine/administration & dosage
17.
Immunity ; 54(6): 1245-1256.e5, 2021 06 08.
Article in English | MEDLINE | ID: mdl-34004140

ABSTRACT

We examined how baseline CD4+ T cell repertoire and precursor states impact responses to pathogen infection in humans using primary immunization with yellow fever virus (YFV) vaccine. YFV-specific T cells in unexposed individuals were identified by peptide-MHC tetramer staining and tracked pre- and post-vaccination by tetramers and TCR sequencing. A substantial number of YFV-reactive T cells expressed memory phenotype markers and contained expanded clones in the absence of exposure to YFV. After vaccination, pre-existing YFV-specific T cell populations with low clonal diversity underwent limited expansion, but rare populations with a reservoir of unexpanded TCRs generated robust responses. These altered dynamics reorganized the immunodominance hierarchy and resulted in an overall increase in higher avidity T cells. Thus, instead of further increasing the representation of dominant clones, YFV vaccination recruits rare and more responsive T cells. Our findings illustrate the impact of vaccines in prioritizing T cell responses and reveal repertoire reorganization as a key component of effective vaccination.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Yellow Fever Vaccine/immunology , Yellow Fever/immunology , Yellow fever virus/immunology , Adult , Animals , Antibodies, Neutralizing/immunology , Antibodies, Viral/immunology , Antigens, Viral/immunology , Cells, Cultured , Chlorocebus aethiops , Humans , Receptors, Antigen, T-Cell/immunology , Vaccination/methods , Vero Cells , Yellow Fever/virology
18.
Cell Rep ; 35(8): 109174, 2021 05 25.
Article in English | MEDLINE | ID: mdl-34038736

ABSTRACT

The CD8+ T cell response to an antigen is composed of many T cell clones with unique T cell receptors, together forming a heterogeneous repertoire of effector and memory cells. How individual T cell clones contribute to this heterogeneity throughout immune responses remains largely unknown. In this study, we longitudinally track human CD8+ T cell clones expanding in response to yellow fever virus (YFV) vaccination at the single-cell level. We observed a drop in clonal diversity in blood from the acute to memory phase, suggesting that clonal selection shapes the circulating memory repertoire. Clones in the memory phase display biased differentiation trajectories along a gradient from stem cell to terminally differentiated effector memory fates. In secondary responses, YFV- and influenza-specific CD8+ T cell clones are poised to recapitulate skewed differentiation trajectories. Collectively, we show that the sum of distinct clonal phenotypes results in the multifaceted human T cell response to acute viral infections.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Virus Diseases/virology , Yellow Fever/virology , Acute Disease , Cell Differentiation , Cells, Cultured , Humans
19.
Sci Rep ; 11(1): 10431, 2021 05 17.
Article in English | MEDLINE | ID: mdl-34001945

ABSTRACT

The present study aimed to investigate whether the serum biomarkers of immune response orchestrate the seroconversion status in patients with autoimmune diseases (AID) upon planned primary 17DD-YF vaccination. For this purpose a total of 161 individuals were enrolled in a prospective study, including patients with Rheumatoid Arthritis (RA = 38), Spondyloarthritis (SpA = 51), Systemic Lupus Erythematosus (SLE = 21) and Sjögren's Syndrome (SS = 30) along with a group of healthy controls (HC = 21). Analysis of plaque reduction neutralization test (PRNT) titers and seropositivity rates along with the 17DD-YF viremia and serum biomarkers were carried out at distinct time points (D0/D3-4/D5-6/D7/D14-28). The results demonstrated an overall lower PRNT titer and seropositivity rate (170 vs. 448; 77 vs. 95%) in AID as compared to HC, especially in SpA and SLE subgroups. No significant differences were observed in the viremia levels amongst groups. In general, a more prominent serum biomarker response was observed in AID as compared to HC, throughout the timeline kinetics. Remarkably, AID/PRNT(-) exhibited higher levels of several biomarkers at baseline as compared to AID/PRNT+. Moreover, while AID/PRNT(+) exhibited earlier increase in serum biomarkers at D3-4/D5-6, the AID/PRNT(-) displayed higher response at later time points (D7/D14-D28). Of note, a synchronic increase of IFN-γ at the peak of viremia (D5-6) was observed in HC and AID/PRNT(+) groups, whereas a later asynchronous IFN-γ response was reported for AID/PRNT(-) at D7. The biomarker profile tends to deflate at post-vaccination timeline, highlighting a putative immunomodulatory effect of live attenuated 17DD-YF vaccine in AID/PRNT(+), but not in AID/PRNT(-). Altogether these data suggested that inflammatory status prior vaccination, low IFN-γ at viremia peak and the occurrence of asynchronous biomarker storm after 17DD-YF vaccination may orchestrate the lack of neutralizing antibody response γ.


Subject(s)
Autoimmune Diseases/immunology , Yellow Fever Vaccine/immunology , Yellow Fever/prevention & control , Yellow fever virus/immunology , Adolescent , Adult , Aged , Aged, 80 and over , Antibodies, Neutralizing/blood , Antibodies, Neutralizing/immunology , Antibodies, Viral/blood , Antibodies, Viral/immunology , Autoimmune Diseases/blood , Case-Control Studies , Female , Healthy Volunteers , Humans , Immunogenicity, Vaccine , Male , Middle Aged , Prospective Studies , Seroconversion , Vaccination , Vaccines, Attenuated/administration & dosage , Vaccines, Attenuated/immunology , Yellow Fever/immunology , Yellow Fever/virology , Yellow Fever Vaccine/administration & dosage , Young Adult
20.
Pathog Dis ; 79(4)2021 04 09.
Article in English | MEDLINE | ID: mdl-33739369

ABSTRACT

The 2019 and 2020 sporadic outbreaks of yellow fever (YF) in Sub-Saharan African countries had raised a lot of global health concerns. This article aims to narratively review the vector biology, YF vaccination program, environmental factors and climatic changes, and to understand how they could facilitate the reemergence of YF. This study comprehensively reviewed articles that focused on the interplay and complexity of YF virus (YFV) vector diversity/competence, YF vaccine immunodynamics and climatic change impacts on YFV transmission as they influence the 2019/2020 sporadic outbreaks in Sub-Saharan Africa (SSA). Based on available reports, vectorial migration, climatic changes and YF immunization level could be reasons for the re-mergence of YF at the community and national levels. Essentially, the drivers of YFV infection due to spillover are moderately constant. However, changes in land use and landscape have been shown to influence sylvan-to-urban spillover. Furthermore, increased precipitation and warmer temperatures due to climate change are likely to broaden the range of mosquitoes' habitat. The 2019/2020 YF outbreaks in SSA is basically a result of inadequate vaccination campaigns, YF surveillance and vector control. Consequently, and most importantly, adequate immunization coverage must be implemented and properly achieved under the responsibility of the public health stakeholders.


Subject(s)
Disease Outbreaks , Yellow Fever Vaccine/administration & dosage , Yellow Fever/epidemiology , Yellow Fever/prevention & control , Yellow fever virus/pathogenicity , Aedes/virology , Africa South of the Sahara/epidemiology , Animals , Climate Change , Global Health/trends , Humans , Incidence , Mosquito Vectors/virology , Rain , Vaccination/methods , Yellow Fever/transmission , Yellow Fever/virology , Yellow fever virus/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...