Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 316
Filtrar
Mais filtros

Eixos temáticos
Intervalo de ano de publicação
1.
Nat Immunol ; 21(6): 684-694, 2020 06.
Artigo em Inglês | MEDLINE | ID: mdl-32231301

RESUMO

Aging is associated with remodeling of the immune system to enable the maintenance of life-long immunity. In the CD8+ T cell compartment, aging results in the expansion of highly differentiated cells that exhibit characteristics of cellular senescence. Here we found that CD27-CD28-CD8+ T cells lost the signaling activity of the T cell antigen receptor (TCR) and expressed a protein complex containing the agonistic natural killer (NK) receptor NKG2D and the NK adaptor molecule DAP12, which promoted cytotoxicity against cells that expressed NKG2D ligands. Immunoprecipitation and imaging cytometry indicated that the NKG2D-DAP12 complex was associated with sestrin 2. The genetic inhibition of sestrin 2 resulted in decreased expression of NKG2D and DAP12 and restored TCR signaling in senescent-like CD27-CD28-CD8+ T cells. Therefore, during aging, sestrins induce the reprogramming of non-proliferative senescent-like CD27-CD28-CD8+ T cells to acquire a broad-spectrum, innate-like killing activity.


Assuntos
Linfócitos T CD8-Positivos/imunologia , Linfócitos T CD8-Positivos/metabolismo , Senescência Celular/imunologia , Células Matadoras Naturais/imunologia , Células Matadoras Naturais/metabolismo , Proteínas Nucleares/genética , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Citotoxicidade Imunológica , Perfilação da Expressão Gênica , Humanos , Proteínas de Membrana/metabolismo , Subfamília K de Receptores Semelhantes a Lectina de Células NK/metabolismo , Proteínas Nucleares/metabolismo , Receptores de Antígenos de Linfócitos T/metabolismo , Receptores de Células Matadoras Naturais/metabolismo , Transdução de Sinais , Febre Amarela/genética , Febre Amarela/imunologia , Febre Amarela/metabolismo , Febre Amarela/virologia , Vírus da Febre Amarela/imunologia
2.
Immunity ; 54(6): 1245-1256.e5, 2021 06 08.
Artigo em Inglês | MEDLINE | ID: mdl-34004140

RESUMO

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.


Assuntos
Linfócitos T CD4-Positivos/imunologia , Vacina contra Febre Amarela/imunologia , Febre Amarela/imunologia , Vírus da Febre Amarela/imunologia , Adulto , Animais , Anticorpos Neutralizantes/imunologia , Anticorpos Antivirais/imunologia , Antígenos Virais/imunologia , Células Cultivadas , Chlorocebus aethiops , Humanos , Receptores de Antígenos de Linfócitos T/imunologia , Vacinação/métodos , Células Vero , Febre Amarela/virologia
3.
J Virol ; 98(5): e0151623, 2024 May 14.
Artigo em Inglês | MEDLINE | ID: mdl-38567951

RESUMO

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.


Assuntos
Macaca fascicularis , Modelos Animais , Vacina contra Febre Amarela , Animais , Feminino , Humanos , Masculino , Anticorpos Neutralizantes/sangue , Anticorpos Neutralizantes/imunologia , Anticorpos Antivirais/sangue , Anticorpos Antivirais/imunologia , Imunidade Inata , Biologia de Sistemas/métodos , Vacinação , Febre Amarela/prevenção & controle , Febre Amarela/imunologia , Febre Amarela/virologia , Vacina contra Febre Amarela/imunologia , Vírus da Febre Amarela/imunologia
4.
J Virol ; 97(4): e0194922, 2023 04 27.
Artigo em Inglês | MEDLINE | ID: mdl-37017533

RESUMO

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.


Assuntos
Replicação Viral , Vírus da Febre Amarela , Animais , Humanos , Ciclização , RNA Viral/metabolismo , Replicação Viral/genética , Febre Amarela/virologia , Vírus da Febre Amarela/metabolismo , Genoma Viral/genética , Linhagem Celular , Cricetinae , Mesocricetus , Células A549
5.
Biologicals ; 86: 101765, 2024 May.
Artigo em Inglês | MEDLINE | ID: mdl-38593685

RESUMO

Yellow fever (YF) is one of the most acute viral hemorrhagic diseases of the 18th and 19th centuries, which continues to cause severe morbidity and mortality in Africa. After 21 years of no reported cases of yellow fever in Nigeria, till 2017 where a case was confirmed in Kwara State, also in November 2018,WHO was informed of a cluster of suspected yellow fever cases and deaths in Edo state, Nigeria. The study was among all age group attending health centres in Benin City, Edo state. A total of 280 blood samples were collected from consented febrile patients and were screened for antibodies to Zika virus using rapid diagnostic test (RDT) kits. Blood samples positive to Zika virus (IgM/IgG RDT), were subjected to molecular characterization. Using the flavividae family primers, six (6) samples where confirmed positive by Hemi-nested reverse transcription PCR (hnRT-PCR) sequencing. Nucleotide sequence blast revealed the sequenceswere similar to Yellow fever virus strains. Phylogenetic analysis revealed that the yellow fever virus sequences are closely related to the African strains. Despite the safe and effective yellow fever vaccine, yellow fever virus is seen to be in circulation, hence the need for continues mass vaccination.


Assuntos
Filogenia , Febre Amarela , Vírus da Febre Amarela , Humanos , Nigéria/epidemiologia , Vírus da Febre Amarela/genética , Vírus da Febre Amarela/imunologia , Febre Amarela/epidemiologia , Febre Amarela/virologia , Febre Amarela/sangue , Adulto , Feminino , Masculino , Adolescente , Pessoa de Meia-Idade , Criança , Pré-Escolar , Adulto Jovem , Anticorpos Antivirais/sangue , Anticorpos Antivirais/imunologia , Lactente , Zika virus/genética , Zika virus/imunologia , Zika virus/isolamento & purificação
6.
Emerg Infect Dis ; 29(9): 1818-1826, 2023 09.
Artigo em Inglês | MEDLINE | ID: mdl-37610174

RESUMO

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.


Assuntos
Febre Amarela , Vírus da Febre Amarela , Vírus da Febre Amarela/classificação , Vírus da Febre Amarela/isolamento & purificação , Febre Amarela/virologia , Surtos de Doenças , Gana/epidemiologia , Humanos , Filogenia , Análise de Sequência de RNA , RNA Viral/análise
7.
N Engl J Med ; 383(5): 452-459, 2020 07 30.
Artigo em Inglês | MEDLINE | ID: mdl-32726531

RESUMO

BACKGROUND: Insufficient vaccine doses and the lack of therapeutic agents for yellow fever put global health at risk, should this virus emerge from sub-Saharan Africa and South America. METHODS: In phase 1a of this clinical trial, we assessed the safety, side-effect profile, and pharmacokinetics of TY014, a fully human IgG1 anti-yellow fever virus monoclonal antibody. In a double-blind, phase 1b clinical trial, we assessed the efficacy of TY014, as compared with placebo, in abrogating viremia related to the administration of live yellow fever vaccine (YF17D-204; Stamaril). The primary safety outcomes were adverse events reported 1 hour after the infusion and throughout the trial. The primary efficacy outcome was the dose of TY014 at which 100% of the participants tested negative for viremia within 48 hours after infusion. RESULTS: A total of 27 healthy participants were enrolled in phase 1a, and 10 participants in phase 1b. During phase 1a, TY014 dose escalation to a maximum of 20 mg per kilogram of body weight occurred in 22 participants. During phases 1a and 1b, adverse events within 1 hour after infusion occurred in 1 of 27 participants who received TY014 and in none of the 10 participants who received placebo. At least one adverse event occurred during the trial in 22 participants who received TY014 and in 8 who received placebo. The mean half-life of TY014 was approximately 12.8 days. At 48 hours after the infusion, none of the 5 participants who received the starting dose of TY014 of 2 mg per kilogram had detectable YF17D-204 viremia; these participants remained aviremic throughout the trial. Viremia was observed at 48 hours after the infusion in 2 of 5 participants who received placebo and at 72 hours in 2 more placebo recipients. Symptoms associated with yellow fever vaccine were less frequent in the TY014 group than in the placebo group. CONCLUSIONS: This phase 1 trial of TY014 did not identify worrisome safety signals and suggested potential clinical benefit, which requires further assessment in a phase 2 trial. (Funded by Tysana; ClinicalTrials.gov number, NCT03776786.).


Assuntos
Anticorpos Monoclonais Humanizados/administração & dosagem , Vacina contra Febre Amarela , Febre Amarela/tratamento farmacológico , Vírus da Febre Amarela/imunologia , Adulto , Anticorpos Monoclonais Humanizados/efeitos adversos , Anticorpos Monoclonais Humanizados/farmacocinética , Relação Dose-Resposta a Droga , Método Duplo-Cego , Meia-Vida , Humanos , Estimativa de Kaplan-Meier , Viremia/tratamento farmacológico , Febre Amarela/virologia , Vírus da Febre Amarela/efeitos dos fármacos
8.
Nature ; 552(7685): 362-367, 2017 12 21.
Artigo em Inglês | MEDLINE | ID: mdl-29236685

RESUMO

The differentiation of human memory CD8 T cells is not well understood. Here we address this issue using the live yellow fever virus (YFV) vaccine, which induces long-term immunity in humans. We used in vivo deuterium labelling to mark CD8 T cells that proliferated in response to the virus and then assessed cellular turnover and longevity by quantifying deuterium dilution kinetics in YFV-specific CD8 T cells using mass spectrometry. This longitudinal analysis showed that the memory pool originates from CD8 T cells that divided extensively during the first two weeks after infection and is maintained by quiescent cells that divide less than once every year (doubling time of over 450 days). Although these long-lived YFV-specific memory CD8 T cells did not express effector molecules, their epigenetic landscape resembled that of effector CD8 T cells. This open chromatin profile at effector genes was maintained in memory CD8 T cells isolated even a decade after vaccination, indicating that these cells retain an epigenetic fingerprint of their effector history and remain poised to respond rapidly upon re-exposure to the pathogen.


Assuntos
Linfócitos T CD8-Positivos/citologia , Linfócitos T CD8-Positivos/imunologia , Diferenciação Celular/imunologia , Epigênese Genética , Memória Imunológica/imunologia , Vacina contra Febre Amarela/imunologia , Animais , Linfócitos T CD8-Positivos/metabolismo , Diferenciação Celular/genética , Proliferação de Células , Cromatina/genética , Cromatina/metabolismo , Metilação de DNA , Deutério , Perfilação da Expressão Gênica , Meia-Vida , Humanos , Memória Imunológica/genética , Contagem de Linfócitos , Camundongos , Técnica de Diluição de Radioisótopos , Transcrição Gênica , Febre Amarela/imunologia , Febre Amarela/virologia , Vírus da Febre Amarela/imunologia
9.
Proc Natl Acad Sci U S A ; 117(12): 6675-6685, 2020 03 24.
Artigo em Inglês | MEDLINE | ID: mdl-32152119

RESUMO

A comprehensive understanding of the development and evolution of human B cell responses induced by pathogen exposure will facilitate the design of next-generation vaccines. Here, we utilized a high-throughput single B cell cloning technology to longitudinally track the human B cell response to the yellow fever virus 17D (YFV-17D) vaccine. The early memory B cell (MBC) response was mediated by both classical immunoglobulin M (IgM) (IgM+CD27+) and switched immunoglobulin (swIg+) MBC populations; however, classical IgM MBCs waned rapidly, whereas swIg+ and atypical IgM+ and IgD+ MBCs were stable over time. Affinity maturation continued for 6 to 9 mo following vaccination, providing evidence for the persistence of germinal center activity long after the period of active viral replication in peripheral blood. Finally, a substantial fraction of the neutralizing antibody response was mediated by public clones that recognize a fusion loop-proximal antigenic site within domain II of the viral envelope glycoprotein. Overall, our findings provide a framework for understanding the dynamics and complexity of human B cell responses elicited by infection and vaccination.


Assuntos
Anticorpos Antivirais/imunologia , Antígenos Virais/imunologia , Linfócitos B/imunologia , Memória Imunológica/imunologia , Vacina contra Febre Amarela/imunologia , Febre Amarela/prevenção & controle , Vírus da Febre Amarela/imunologia , Adulto , Humanos , Vacinação , Vacinas Atenuadas/imunologia , Proteínas do Envelope Viral/imunologia , Replicação Viral , Febre Amarela/imunologia , Febre Amarela/virologia , Vacina contra Febre Amarela/administração & dosagem
10.
Proc Natl Acad Sci U S A ; 117(51): 32648-32656, 2020 12 22.
Artigo em Inglês | MEDLINE | ID: mdl-33268494

RESUMO

Yellow fever (YF) is a mosquito-transmitted viral disease that causes tens of thousands of deaths each year despite the long-standing deployment of an effective vaccine. In its most severe form, YF manifests as a hemorrhagic fever that causes severe damage to visceral organs. Although coagulopathy is a defining feature of severe YF in humans, the mechanism by which it develops remains uncertain. Hepatocytes are a major target of yellow fever virus (YFV) infection, and the coagulopathy in severe YF has long been attributed to massive hepatocyte infection and destruction that results in a defect in clotting factor synthesis. However, when we analyzed blood from Brazilian patients with severe YF, we found high concentrations of plasma D-dimer, a fibrin split product, suggestive of a concurrent consumptive process. To define the relationship between coagulopathy and hepatocellular tropism, we compared infection and disease in Fah-/-, Rag2-/-, and Il2rɣ-/- mice engrafted with human hepatocytes (hFRG mice) and rhesus macaques using a highly pathogenic African YFV strain. YFV infection of macaques and hFRG mice caused substantial hepatocyte infection, liver damage, and coagulopathy as defined by virological, clinical, and pathological criteria. However, only macaques developed a consumptive coagulopathy whereas YFV-infected hFRG mice did not. Thus, infection of cell types other than hepatocytes likely contributes to the consumptive coagulopathy associated with severe YF in primates and humans. These findings expand our understanding of viral hemorrhagic disease and associated coagulopathy and suggest directions for clinical management of severe YF cases.


Assuntos
Coagulação Intravascular Disseminada/virologia , Hepatopatias/virologia , Tropismo Viral/fisiologia , Febre Amarela/fisiopatologia , Vírus da Febre Amarela/fisiologia , Animais , Modelos Animais de Doenças , Coagulação Intravascular Disseminada/sangue , Feminino , Produtos de Degradação da Fibrina e do Fibrinogênio/análise , Hepatócitos/transplante , Hepatócitos/virologia , Humanos , Hepatopatias/fisiopatologia , Macaca mulatta , Masculino , Camundongos Endogâmicos C57BL , Camundongos Knockout , Febre Amarela/complicações , Febre Amarela/virologia
11.
PLoS Pathog ; 16(8): e1008699, 2020 08.
Artigo em Inglês | MEDLINE | ID: mdl-32764827

RESUMO

São Paulo, a densely inhabited state in southeast Brazil that contains the fourth most populated city in the world, recently experienced its largest yellow fever virus (YFV) outbreak in decades. YFV does not normally circulate extensively in São Paulo, so most people were unvaccinated when the outbreak began. Surveillance in non-human primates (NHPs) is important for determining the magnitude and geographic extent of an epizootic, thereby helping to evaluate the risk of YFV spillover to humans. Data from infected NHPs can give more accurate insights into YFV spread than when using data from human cases alone. To contextualise human cases, identify epizootic foci and uncover the rate and direction of YFV spread in São Paulo, we generated and analysed virus genomic data and epizootic case data from NHPs in São Paulo. We report the occurrence of three spatiotemporally distinct phases of the outbreak in São Paulo prior to February 2018. We generated 51 new virus genomes from YFV positive cases identified in 23 different municipalities in São Paulo, mostly sampled from NHPs between October 2016 and January 2018. Although we observe substantial heterogeneity in lineage dispersal velocities between phylogenetic branches, continuous phylogeographic analyses of generated YFV genomes suggest that YFV lineages spread in São Paulo at a mean rate of approximately 1km per day during all phases of the outbreak. Viral lineages from the first epizootic phase in northern São Paulo subsequently dispersed towards the south of the state to cause the second and third epizootic phases there. This alters our understanding of how YFV was introduced into the densely populated south of São Paulo state. Our results shed light on the sylvatic transmission of YFV in highly fragmented forested regions in São Paulo state and highlight the importance of continued surveillance of zoonotic pathogens in sentinel species.


Assuntos
Genoma Viral , Doenças dos Primatas/virologia , Febre Amarela/veterinária , Febre Amarela/virologia , Vírus da Febre Amarela/genética , Zoonoses/virologia , Animais , Brasil/epidemiologia , Surtos de Doenças , Genômica , Humanos , Filogenia , Filogeografia , Doenças dos Primatas/epidemiologia , Doenças dos Primatas/transmissão , Primatas/virologia , Febre Amarela/epidemiologia , Febre Amarela/transmissão , Vírus da Febre Amarela/classificação , Vírus da Febre Amarela/isolamento & purificação , Zoonoses/epidemiologia , Zoonoses/transmissão
12.
J Infect Dis ; 221(12): 2018-2025, 2020 06 11.
Artigo em Inglês | MEDLINE | ID: mdl-31545367

RESUMO

BACKGROUND: The once-in-a-lifetime recommendation for vaccination against yellow fever virus (YFV) has been controversial, leading to increased scrutiny of the durability of immunity after 17D vaccination. METHODS: This is a cross-sectional analysis of 17D vaccinees living in nonendemic Portland, Oregon. Neutralization assays were used to determine YFV immunity. The relationships between 17D immunity and vaccination history, demographics, and travel were evaluated using nominal logistic regression. RESULTS: Seventy-one of 92 (77.2%) subjects were YFV seropositive (90 percent plaque reduction neutralization test ≥1:10) at all timepoints, and 24 of 38 (63.8%) were YFV seropositive at ≥10 years after single-dose vaccination. No relationship was found between YFV immunity and time in endemic countries, other flavivirus immunity, or demographics. Subjects were most likely to become seronegative between 3 and 12 years postvaccination (logistic regression, odds ratio [OR] = 1.75; 95% confidence interval [CI], 1.12-2.73). A comparison of our results and 4 previous studies of YFV nonendemic vaccinees found that overall, 79% (95% CI, 70%-86%) of vaccinees are likely to be seropositive ≥10 years postvaccination. CONCLUSIONS: These results suggest that 1 in 5 17D vaccinees will lack neutralizing antibodies at ~10 years postvaccination, and a booster vaccination should be considered for nonendemic vaccinees before travel to regions where there is a high risk of YFV transmission.


Assuntos
Anticorpos Neutralizantes/sangue , Anticorpos Antivirais/sangue , Imunogenicidade da Vacina , Vacina contra Febre Amarela/imunologia , Febre Amarela/prevenção & controle , Adulto , Idoso , Idoso de 80 Anos ou mais , Anticorpos Neutralizantes/imunologia , Anticorpos Antivirais/imunologia , Estudos Transversais , Feminino , Humanos , Imunização Secundária , Masculino , Pessoa de Meia-Idade , Testes de Neutralização , Oregon , Fatores de Tempo , Doença Relacionada a Viagens , Febre Amarela/imunologia , Febre Amarela/transmissão , Febre Amarela/virologia , Vacina contra Febre Amarela/administração & dosagem , Vírus da Febre Amarela/imunologia , Adulto Jovem
13.
J Infect Dis ; 221(12): 2026-2034, 2020 06 11.
Artigo em Inglês | MEDLINE | ID: mdl-31545372

RESUMO

BACKGROUND: Yellow fever (YF) is a vector-borne viral hemorrhagic disease endemic in Africa and Latin America. In 2016, the World Health Organization (WHO) developed the Eliminate YF Epidemics strategy aiming at eliminating YF epidemics by 2026. METHODS: We developed a spatiotemporal model of YF, accounting for the impact of temperature, vector distribution, and socioeconomic factors on disease transmission. We validated our model against previous estimates of YF basic reproductive number (R0). We used the model to estimate global risk of YF outbreaks and vaccination efforts needed to achieve elimination of YF epidemics. RESULTS: We showed that the global risk of YF outbreaks is highly heterogeneous. High-risk transmission areas (R0 > 6) are mainly found in West Africa and the Equatorial region of Latin America. We showed that vaccination coverage needed to eliminate YF epidemics in an endemic country varies substantially between districts. In many endemic countries, a 90% vaccination coverage is needed to achieve elimination. However, in some high-risk districts in Africa, a 95% coverage may be required. CONCLUSIONS: Global elimination of YF epidemics requires higher population-level immunity than the 80% coverage recommended by the WHO. Optimal YF vaccination strategy should be tailored to the risk profile of each endemic country.


Assuntos
Erradicação de Doenças , Doenças Endêmicas/prevenção & controle , Epidemias/prevenção & controle , Vacina contra Febre Amarela/administração & dosagem , Febre Amarela/epidemiologia , África , América , Humanos , América Latina , Modelos Estatísticos , Mosquitos Vetores/virologia , Medição de Risco , Estações do Ano , Análise Espaço-Temporal , Cobertura Vacinal/normas , Organização Mundial da Saúde , Febre Amarela/prevenção & controle , Febre Amarela/transmissão , Febre Amarela/virologia , Vírus da Febre Amarela/imunologia , Vírus da Febre Amarela/isolamento & purificação
14.
J Infect Dis ; 221(Suppl 3): S308-S318, 2020 03 28.
Artigo em Inglês | MEDLINE | ID: mdl-31711190

RESUMO

Next-generation sequencing technologies, exponential increases in the availability of virus genomic data, and ongoing advances in phylogenomic methods have made genomic epidemiology an increasingly powerful tool for public health response to a range of mosquito-borne virus outbreaks. In this review, we offer a brief primer on the scope and methods of phylogenomic analyses that can answer key epidemiological questions during mosquito-borne virus public health emergencies. We then focus on case examples of outbreaks, including those caused by dengue, Zika, yellow fever, West Nile, and chikungunya viruses, to demonstrate the utility of genomic epidemiology to support the prevention and control of mosquito-borne virus threats. We extend these case studies with operational perspectives on how to best incorporate genomic epidemiology into structured surveillance and response programs for mosquito-borne virus control. Many tools for genomic epidemiology already exist, but so do technical and nontechnical challenges to advancing their use. Frameworks to support the rapid sharing of multidimensional data and increased cross-sector partnerships, networks, and collaborations can support advancement on all scales, from research and development to implementation by public health agencies.


Assuntos
Culicidae/virologia , Surtos de Doenças/prevenção & controle , Genômica , Controle de Mosquitos , Saúde Pública , Doenças Transmitidas por Vetores/prevenção & controle , Animais , Febre de Chikungunya/epidemiologia , Febre de Chikungunya/prevenção & controle , Febre de Chikungunya/virologia , Vírus Chikungunya/genética , Dengue/epidemiologia , Dengue/prevenção & controle , Dengue/virologia , Humanos , Mosquitos Vetores/virologia , Doenças Transmitidas por Vetores/epidemiologia , Doenças Transmitidas por Vetores/virologia , Febre do Nilo Ocidental/epidemiologia , Febre do Nilo Ocidental/prevenção & controle , Febre do Nilo Ocidental/virologia , Febre Amarela/epidemiologia , Febre Amarela/prevenção & controle , Febre Amarela/virologia , Zika virus/genética , Infecção por Zika virus/epidemiologia , Infecção por Zika virus/prevenção & controle , Infecção por Zika virus/virologia
15.
J Virol ; 94(1)2019 12 12.
Artigo em Inglês | MEDLINE | ID: mdl-31597773

RESUMO

The recent reemergence of yellow fever virus (YFV) in Brazil has raised serious concerns due to the rapid dissemination of the virus in the southeastern region. To better understand YFV genetic diversity and dynamics during the recent outbreak in southeastern Brazil, we generated 18 complete and nearly complete genomes from the peak of the epidemic curve from nonhuman primates (NHPs) and human infected cases across the Espírito Santo and Rio de Janeiro states. Genomic sequencing of 18 YFV genomes revealed the estimated timing, source, and likely routes of yellow fever virus transmission and dispersion during one of the largest outbreaks ever registered in Brazil. We showed that during the recent epidemic, YFV was reintroduced from Minas Gerais to the Espírito Santo and Rio de Janeiro states multiple times between 2016 and 2019. The analysis of data from portable sequencing could identify the corridor of spread of YFV. These findings reinforce the idea that continued genomic surveillance strategies can provide information on virus genetic diversity and transmission dynamics that might assist in understanding arbovirus epidemics.IMPORTANCE Arbovirus infections in Brazil, including yellow fever, dengue, zika, and chikungunya, result in considerable morbidity and mortality and are pressing public health concerns. However, our understanding of these outbreaks is hampered by the limited availability of genomic data. In this study, we investigated the genetic diversity and spatial distribution of YFV during the current outbreak by analyzing genomic data from areas in southeastern Brazil not covered by other previous studies. To gain insights into the routes of YFV introduction and dispersion, we tracked the virus by sequencing YFV genomes sampled from nonhuman primates and infected patients from the southeastern region. Our study provides an understanding of how YFV initiates transmission in new Brazilian regions and illustrates that genomics in the field can augment traditional approaches to infectious disease surveillance and control.


Assuntos
Surtos de Doenças , Genoma Viral , Febre Amarela/epidemiologia , Febre Amarela/transmissão , Vírus da Febre Amarela/genética , Aedes/virologia , Alouatta/virologia , Animais , Brasil/epidemiologia , Callithrix/virologia , Cebus/virologia , Feminino , Variação Genética , Humanos , Incidência , Leontopithecus/virologia , Masculino , Mosquitos Vetores/virologia , Filogenia , Filogeografia , Sequenciamento Completo do Genoma , Febre Amarela/virologia , Vírus da Febre Amarela/classificação , Vírus da Febre Amarela/isolamento & purificação , Vírus da Febre Amarela/patogenicidade
16.
Vet Pathol ; 57(5): 681-686, 2020 09.
Artigo em Inglês | MEDLINE | ID: mdl-32783517

RESUMO

Yellow fever is an important zoonotic viral disease that can be fatal for both human and nonhuman primates. We evaluated histopathologic changes in free-ranging neotropical primates naturally infected with yellow fever virus (YFV) compared with uninfected cohorts. The most frequent lesions in primates infected with YFV were hepatic changes characterized by midzonal necrosis with lipidosis and mild inflammation including lymphocytes, macrophages, plasma cells, and infrequently neutrophils. Importantly, severe necrotizing hepatic lesions were often observed in Alouatta sp. (howler monkeys), whereas Callithrix sp. (common marmosets) had nearly no hepatic changes. Moderate to severe hepatic necrosis was present in 21/23 (91%) of the YFV-positive Alouatta sp. compared with 10/29 (34%) of the YFV-positive Callithrix sp. (P < .0001; odds ratio = 20). Similarly, hepatitis was more intense in Alouatta sp. compared with Callithrix sp. Furthermore, the frequency of YFV infection was significantly higher in Alouatta sp. compared with Callithrix sp. or Sapajus sp. (capuchin monkeys). Therefore, these data support the notion that Alouatta sp. is highly susceptible to infection and YFV-induced lesions, whereas Callithrix sp. is susceptible to infection but has a lower frequency of YFV-induced lesions.


Assuntos
Alouatta/virologia , Callithrix/virologia , Doenças dos Macacos/patologia , Febre Amarela/veterinária , Animais , Suscetibilidade a Doenças , Feminino , Fígado/patologia , Fígado/virologia , Masculino , Doenças dos Macacos/virologia , Febre Amarela/patologia , Febre Amarela/virologia
17.
Bull Hist Med ; 94(2): 215-243, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-33416552

RESUMO

This article explores the entangled histories of dengue and yellow fever. It traces how historical conflations of these diseases deepened at the start of the twentieth century in the context of rising fears that yellow fever might spread to Asia. Advances in biomedicine, I suggest, reinforced notions of their kinship and generated competing theories that dengue either foreshadowed yellow fever in Asia or inoculated the region against it. This history in which the language and science of dengue and yellow fever shadowed one another offers a nonlinear narrative of scientific progress. Furthermore, as the so-called neglected tropical diseases resurge in the present, it elucidates how disease threats are read against one another. Thus, the article offers a historical context to ongoing discussions on disease emergence and pandemic preparedness.


Assuntos
Dengue/história , Febre Amarela/história , Ásia , Dengue/virologia , História do Século XIX , História do Século XX , Humanos , Febre Amarela/virologia
18.
Emerg Infect Dis ; 25(6): 1248-1249, 2019 06.
Artigo em Inglês | MEDLINE | ID: mdl-30870138

RESUMO

In 2018, yellow fever with hepatitis was diagnosed for 2 unvaccinated travelers returning to France from Brazil. Hepatitis persisted for >6 months; liver enzyme levels again increased 2 months after disease onset with no detection of yellow fever virus RNA or other pathogens. Persistent hepatitis with hepatic cytolysis rebound probably resulted from immune response.


Assuntos
Hepatite/epidemiologia , Febre Amarela/epidemiologia , Vírus da Febre Amarela , Biópsia , Brasil/epidemiologia , Comorbidade , Hepatite/diagnóstico , Hepatite/etiologia , Humanos , Testes de Função Hepática , Vigilância em Saúde Pública , Febre Amarela/diagnóstico , Febre Amarela/virologia
20.
Artigo em Inglês | MEDLINE | ID: mdl-31262759

RESUMO

Yellow fever virus (YFV) is a human Flavivirus reemerging in parts of the world. While a vaccine is available, large outbreaks have recently occurred in Brazil and certain African countries. Development of an effective antiviral against YFV is crucial, as there is no available effective drug against YFV. We have identified several novel nucleoside analogs with potent antiviral activity against YFV with 50% effective concentration (EC50) values between 0.25 and 1 µM with selectivity indices over 100 in culture.


Assuntos
Antivirais/uso terapêutico , Nucleosídeos/análogos & derivados , Nucleosídeos/uso terapêutico , Febre Amarela/tratamento farmacológico , Vírus da Febre Amarela/efeitos dos fármacos , Vírus da Febre Amarela/patogenicidade , África , Animais , Brasil , Linhagem Celular Tumoral , Chlorocebus aethiops , Humanos , Estrutura Molecular , Células Vero , Febre Amarela/virologia
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa