Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 67
Filter
1.
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
2.
PLoS Negl Trop Dis ; 16(1): e0010171, 2022 Jan.
Article in English | MEDLINE | ID: mdl-35073317

ABSTRACT

Aedes simpsoni complex has a wide distribution in Africa and comprises at least three described sub-species including the yellow fever virus (YFV) vector Ae. bromeliae. To date, the distribution and relative contributions of the sub-species and/or subpopulations including bionomic characteristics in relation to YF transmission dynamics remain poorly studied. In this study conducted in two areas with divergent ecosystems: peri-urban (coastal Rabai) and rural (Rift Valley Kerio Valley) in Kenya, survival rate was estimated by parity in Ae. simpsoni s.l. mosquitoes sampled using CO2-baited BG Sentinel traps. We then applied PCR targeting the nuclear internal transcribed spacer 2 (ITS2), region followed by sequencing and phylogenetic analytics to identify the sibling species in the Ae. simpsoni complex among parous and blood fed cohorts. Our results show that Ae. bromeliae was the most dominant sub-species in both areas, exhibiting high survival rates, human blood-feeding, and potentially, high vectorial capacity for pathogen transmission. We document for the first time the presence of Ae. lilii in Kenya and potentially yet-to-be described species in the complex displaying human feeding tendencies. We also infer a wide host feeding range on rodents, reptile, and domestic livestock besides humans especially for Ae. bromeliae. This feeding trend could likely expose humans to various zoonotic pathogens. Taken together, we highlight the utility of genotype-based analyses to generate precision surveillance data of vector populations for enhanced disease risk prediction and to guide cost-effective interventions (e.g. YF vaccinations).


Subject(s)
Aedes/classification , Aedes/virology , Arbovirus Infections/transmission , Arboviruses/isolation & purification , Mosquito Vectors/virology , Yellow Fever/transmission , Aedes/physiology , Africa, Eastern/epidemiology , Animals , Arbovirus Infections/epidemiology , Arboviruses/classification , Ecosystem , Environment , Feeding Behavior , Female , Host Specificity , Yellow Fever/epidemiology , Yellow fever virus/classification , Yellow fever virus/isolation & purification
3.
Viruses ; 13(7)2021 07 16.
Article in English | MEDLINE | ID: mdl-34372589

ABSTRACT

The yellow fever virus vaccine, 17D, was derived through the serial passage of the wild-type (WT) strain Asibi virus in mouse and chicken tissue. Since its derivation, the mechanism of attenuation of 17D virus has been investigated using three 17D substrains and WT Asibi virus. Although all three substrains of 17D have been sequenced, only one isolate of Asibi has been examined genetically and all interpretation of attenuation is based on this one isolate. Here, we sequenced the genome of Asibi virus from three different laboratories and show that the WT strain is genetically homogenous at the amino acids that distinguish Asibi from 17D vaccine virus.


Subject(s)
Genome, Viral , Viral Envelope Proteins/genetics , Yellow Fever Vaccine/immunology , Yellow fever virus/genetics , Antigens, Viral/immunology , Genetic Variation , Vaccines, Attenuated , Viral Envelope Proteins/immunology , Whole Genome Sequencing , Yellow fever virus/classification , Yellow fever virus/immunology
4.
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
5.
PLoS Negl Trop Dis ; 14(8): e0008405, 2020 08.
Article in English | MEDLINE | ID: mdl-32780745

ABSTRACT

Yellow fever virus (YFV) causes a clinical syndrome of acute hemorrhagic hepatitis. YFV transmission involves non-human primates (NHP), mosquitoes and humans. By late 2016, Brazil experienced the largest YFV outbreak of the last 100 years, with 2050 human confirmed cases, with 681 cases ending in death and 764 confirmed epizootic cases in NHP. Among affected areas, Bahia state in Northeastern was the only region with no autochthonous human cases. By using next generation sequence approach, we investigated the molecular epidemiology of YFV in NHP in Bahia and discuss what factors might have prevented human cases. We investigated 47 YFV positive tissue samples from NHP cases to generate 8 novel YFV genomes. ML phylogenetic tree reconstructions and automated subtyping tools placed the newly generated genomes within the South American genotype I (SA I). Our analysis revealed that the YFV genomes from Bahia formed two distinct well-supported phylogenetic clusters that emerged most likely of an introduction from Minas Gerais and Espírito Santo states. Vegetation coverage analysis performed shows predominantly low to medium vegetation coverage in Bahia state. Together, our findings support the hypothesis of two independent YFV SA-I introductions. We also highlighted the effectiveness of the actions taken by epidemiological surveillance team of the state to prevented human cases.


Subject(s)
Primate Diseases/virology , Yellow Fever/veterinary , Yellow fever virus/genetics , Alouatta , Animals , Brazil/epidemiology , Callithrix , Ecosystem , Genome, Viral , Humans , Phylogeny , Yellow Fever/epidemiology , Yellow Fever/prevention & control , Yellow Fever/transmission , Yellow fever virus/classification
6.
PLoS Pathog ; 16(8): e1008699, 2020 08.
Article in English | MEDLINE | ID: mdl-32764827

ABSTRACT

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.


Subject(s)
Genome, Viral , Primate Diseases/virology , Yellow Fever/veterinary , Yellow Fever/virology , Yellow fever virus/genetics , Zoonoses/virology , Animals , Brazil/epidemiology , Disease Outbreaks , Genomics , Humans , Phylogeny , Phylogeography , Primate Diseases/epidemiology , Primate Diseases/transmission , Primates/virology , Yellow Fever/epidemiology , Yellow Fever/transmission , Yellow fever virus/classification , Yellow fever virus/isolation & purification , Zoonoses/epidemiology , Zoonoses/transmission
7.
Viruses ; 12(2)2020 02 17.
Article in English | MEDLINE | ID: mdl-32079143

ABSTRACT

One patient presented hyporexia, asthenia, adynamia, and jaundice two months after acute yellow fever (YF) onset; plus laboratory tests indicating hepatic cytolysis and a rebound of alanine and aspartate transaminases, and total and direct bilirubin levels. Laboratory tests discarded autoimmune hepatitis, inflammatory or metabolic liver disease, and new infections caused by hepatotropic agents. Anti-YFV IgM, IgG and neutralizing antibodies were detected in different times, but no viremia. A liver biopsy was collected three months after YF onset and tested positive for YFV antigens and wild-type YFV-RNA (364 RNA-copies/gram/liver). Transaminases and bilirubin levels remained elevated for five months, and the arresting of symptoms persisted for six months after the acute YF onset. Several serum chemokines, cytokines, and growth factors were measured. A similar immune response profile was observed in the earlier phases of the disease, followed by more pronounced changes in the later stages, when transaminases levels returned to normal. The results indicated viral persistence in the liver and continual liver cell damage three months after YF onset and reinforced the need for extended follow-ups of YF patients. Further studies to investigate the role of possible viral persistence and the immune response causing relapsing hepatitis following YF are also necessary.


Subject(s)
Antibodies, Viral/blood , Hepatitis A/diagnosis , Liver/virology , Yellow Fever/complications , Acute Disease , Antibodies, Neutralizing/blood , Biopsy , Cytokines/blood , Hepatitis A/immunology , Humans , Jaundice/virology , Liver/pathology , Liver Function Tests , Male , Middle Aged , Recurrence , Time Factors , Yellow fever virus/classification , Yellow fever virus/immunology
8.
J Virol ; 94(1)2019 12 12.
Article in English | MEDLINE | ID: mdl-31597773

ABSTRACT

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.


Subject(s)
Disease Outbreaks , Genome, Viral , Yellow Fever/epidemiology , Yellow Fever/transmission , Yellow fever virus/genetics , Aedes/virology , Alouatta/virology , Animals , Brazil/epidemiology , Callithrix/virology , Cebus/virology , Female , Genetic Variation , Humans , Incidence , Leontopithecus/virology , Male , Mosquito Vectors/virology , Phylogeny , Phylogeography , Whole Genome Sequencing , Yellow Fever/virology , Yellow fever virus/classification , Yellow fever virus/isolation & purification , Yellow fever virus/pathogenicity
9.
Int J Infect Dis ; 87: 143-150, 2019 Oct.
Article in English | MEDLINE | ID: mdl-31382047

ABSTRACT

OBJECTIVES: Yellow fever virus historically was a frequent threat to American and European coasts. Medical milestones such as the discovery of mosquitoes as vectors and subsequently an effective vaccine significantly reduced its incidence, in spite of which, thousands of cases of this deathly disease still occur regularly in Sub-Saharan Africa and the Amazonian basin in South America, which are usually not reported. An urban outbreak in Angola, consecutive years of increasing incidence near major Brazilian cities, and imported cases in China, South America and Europe, have brought this virus back to the global spotlight. The aim of this article is to underline that the preventive YFV measures, such as vaccination, need to be carefully revised in order to minimize the risks of new YFV outbreaks, especially in urban or immunologically vulnerable places. Furthermore, this article highlights the diverse factors that have favored the spread of other Aedes spp.-associated arboviral diseases like Dengue, Chikungunya and Zika, to northern latitudes causing epidemics in the United States and Europe, emphasizing the possibility that YFV might follow the path of these viruses unless enhanced surveillance and efficient control systems are urgently initiated.


Subject(s)
Yellow Fever/epidemiology , Yellow fever virus/isolation & purification , Animals , Humans , Mosquito Vectors/physiology , Mosquito Vectors/virology , North America/epidemiology , Yellow Fever/transmission , Yellow Fever/virology , Yellow fever virus/classification , Yellow fever virus/genetics
10.
Emerg Infect Dis ; 25(8): 1567-1570, 2019 08.
Article in English | MEDLINE | ID: mdl-31310221

ABSTRACT

We report a 3-year-old child who was hospitalized because of severe manifestations of the central nervous system. The child died after 6 days of hospitalization. Analysis of postmortem cerebrospinal fluid showed the presence of yellow fever virus RNA. Nucleotide sequencing confirmed that the virus was wild-type yellow fever virus.


Subject(s)
RNA, Viral/genetics , Yellow Fever/cerebrospinal fluid , Yellow Fever/virology , Yellow fever virus/genetics , Antiviral Agents/pharmacology , Antiviral Agents/therapeutic use , Autopsy , Biomarkers , Brazil , Child, Preschool , Disease Progression , Fatal Outcome , Female , Humans , Phylogeny , Sequence Analysis, DNA , Symptom Assessment , Tomography, X-Ray Computed , Yellow Fever/diagnosis , Yellow Fever/drug therapy , Yellow fever virus/classification , Yellow fever virus/isolation & purification
11.
Sci Rep ; 9(1): 5474, 2019 04 02.
Article in English | MEDLINE | ID: mdl-30940867

ABSTRACT

Beginning in late 2016 Brazil faced the worst outbreak of Yellow Fever in recent decades, mainly located in southeastern rural regions of the country. In the present study we characterize the Yellow Fever Virus (YFV) associated with this outbreak in São Paulo State, Brazil. Blood or tissues collected from 430 dead monkeys and 1030 pools containing a total of 5,518 mosquitoes were tested for YFV by quantitative RT-PCR, immunohistochemistry (IHC) and indirect immunofluorescence. A total of 67 monkeys were YFV-positive and 3 pools yielded YFV following culture in a C6/36 cell line. Analysis of five nearly full length genomes of YFV from collected samples was consistent with evidence that the virus associated with the São Paulo outbreak originated in Minas Gerais. The phylogenetic analysis also showed that strains involved in the 2016-2017 outbreak in distinct Brazilian states (i.e., Minas Gerais, Rio de Janeiro, Espirito Santo) intermingled in maximum-likelihood and Bayesian trees. Conversely, the strains detected in São Paulo formed a monophyletic cluster, suggesting that they were local-adapted. The finding of YFV by RT-PCR in five Callithrix monkeys who were all YFV-negative by histopathology or immunohistochemistry suggests that this YFV lineage circulating in Sao Paulo is associated with different outcomes in Callithrix when compared to other monkeys.


Subject(s)
Culicidae/virology , Disease Outbreaks/classification , Haplorhini/virology , Yellow Fever/epidemiology , Yellow fever virus/classification , Animals , Antigens, Viral/analysis , Bayes Theorem , Brazil/epidemiology , Cell Line , Humans , Phylogeny , Phylogeography , Whole Genome Sequencing , Yellow Fever/immunology , Yellow Fever/virology , Yellow fever virus/genetics , Yellow fever virus/immunology , Yellow fever virus/isolation & purification , Zoonoses/virology
12.
PLoS Negl Trop Dis ; 12(9): e0006738, 2018 09.
Article in English | MEDLINE | ID: mdl-30188905

ABSTRACT

The yellow fever virus (YFV) recently reemerged in the large outbreaks in Africa and Brazil, and the first imported patients into Asia have recalled the concerns of YFV evolution. Here we show phylogenomics of YFV with serial clinical samples of the 2016 YFV infections. Phylogenetics exhibited that the 2016 strains were close to Angola 1971 strains and only three amino acid changes presented new to other lineages. Deep sequencing of viral genomes discovered 101 intrahost single nucleotide variations (iSNVs) and 234 single nucleotide polymorphisms (SNPs). Analysis of iSNV distribution and mutated allele frequency revealed that the coding regions were under purifying selection. Comparison of the evolutionary rates estimated by iSNV and SNP showed that the intrahost rate was ~2.25 times higher than the epidemic rate, and both rates were higher than the long-term YFV substitution rate, as expected. In addition, the result also hinted that short viremia duration of YFV might further hinder the evolution of YFV.


Subject(s)
Evolution, Molecular , Phylogeny , Polymorphism, Single Nucleotide , Yellow fever virus/classification , Yellow fever virus/genetics , Genome, Viral , High-Throughput Nucleotide Sequencing , Humans , Mutation Rate , Yellow Fever/virology , Yellow fever virus/isolation & purification
13.
Science ; 361(6405): 894-899, 2018 08 31.
Article in English | MEDLINE | ID: mdl-30139911

ABSTRACT

The yellow fever virus (YFV) epidemic in Brazil is the largest in decades. The recent discovery of YFV in Brazilian Aedes species mosquitos highlights a need to monitor the risk of reestablishment of urban YFV transmission in the Americas. We use a suite of epidemiological, spatial, and genomic approaches to characterize YFV transmission. We show that the age and sex distribution of human cases is characteristic of sylvatic transmission. Analysis of YFV cases combined with genomes generated locally reveals an early phase of sylvatic YFV transmission and spatial expansion toward previously YFV-free areas, followed by a rise in viral spillover to humans in late 2016. Our results establish a framework for monitoring YFV transmission in real time that will contribute to a global strategy to eliminate future YFV epidemics.


Subject(s)
Disease Outbreaks/prevention & control , Epidemiological Monitoring , Genomics/methods , Yellow Fever/prevention & control , Yellow Fever/transmission , Yellow fever virus/isolation & purification , Aedes/virology , Age Factors , Animals , Brazil/epidemiology , Disease Outbreaks/statistics & numerical data , Evolution, Molecular , Humans , Phylogeny , Polymerase Chain Reaction , Risk , Sex Factors , Spatio-Temporal Analysis , Yellow Fever/epidemiology , Yellow Fever/virology , Yellow fever virus/classification , Yellow fever virus/genetics
14.
Sci Adv ; 4(7): eaar4297, 2018 07.
Article in English | MEDLINE | ID: mdl-29978039

ABSTRACT

How previous immunity influences immune memory recall and protection against related flaviviruses is largely unknown, yet encounter with multiple flaviviruses in a lifetime is increasingly likely. Using sequential challenges with dengue virus (DENV), yellow fever virus (YFV), and Japanese encephalitis virus (JEV), we induced cross-reactive cellular and humoral immunity among flaviviruses from differing serocomplexes. Antibodies against JEV enhanced DENV replication; however, JEV immunity was protective in vivo during secondary DENV1 infection, promoting rapid gains in antibody avidity. Mechanistically, JEV immunity activated dendritic cells and effector memory T cells, which developed a T follicular helper cell phenotype in draining lymph nodes upon secondary DENV1 infection. We identified cross-reactive epitopes that promote recall from a pool of flavivirus serocomplex cross-reactive memory CD4 T cells and confirmed that a similar serocomplex cross-reactive immunity occurs in humans. These results show that sequential immunizations for flaviviruses sharing CD4 epitopes should promote protection during a subsequent heterologous infection.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Cross Reactions/immunology , Flavivirus/immunology , Amino Acid Sequence , Animals , Antibodies, Neutralizing/immunology , CD4-Positive T-Lymphocytes/cytology , CD4-Positive T-Lymphocytes/metabolism , Dengue Virus/classification , Dengue Virus/immunology , Encephalitis Virus, Japanese/classification , Encephalitis Virus, Japanese/immunology , Epitopes/chemistry , Epitopes/immunology , Flavivirus/classification , Humans , Immunity, Cellular , Immunity, Humoral , Immunologic Memory , Leukocytes, Mononuclear/cytology , Leukocytes, Mononuclear/immunology , Leukocytes, Mononuclear/metabolism , Mice , Mice, Inbred C57BL , Phylogeny , Yellow fever virus/classification , Yellow fever virus/immunology
16.
J Gen Virol ; 99(4): 536-548, 2018 04.
Article in English | MEDLINE | ID: mdl-29469689

ABSTRACT

Southeastern Brazil has been suffering a rapid expansion of a severe sylvatic yellow fever virus (YFV) outbreak since late 2016, which has reached one of the most populated zones in Brazil and South America, heretofore a yellow fever-free zone for more than 70 years. In the current study, we describe the complete genome of 12 YFV samples from mosquitoes, humans and non-human primates from the Brazilian 2017 epidemic. All of the YFV sequences belong to the modern lineage (sub-lineage 1E) of South American genotype I, having been circulating for several months prior to the December 2016 detection. Our data confirm that viral strains associated with the most severe YF epidemic in South America in the last 70 years display unique amino acid substitutions that are mainly located in highly conserved positions in non-structural proteins. Our data also corroborate that YFV has spread southward into Rio de Janeiro state following two main sylvatic dispersion routes that converged at the border of the great metropolitan area comprising nearly 12 million unvaccinated inhabitants. Our original results can help public health authorities to guide the surveillance, prophylaxis and control measures required to face such a severe epidemiological problem. Finally, it will also inspire other workers to further investigate the epidemiological and biological significance of the amino acid polymorphisms detected in the Brazilian 2017 YFV strains.


Subject(s)
Yellow Fever/virology , Yellow fever virus/genetics , Brazil/epidemiology , Disease Outbreaks , Genome, Viral , Genomics , Genotype , Humans , Models, Molecular , Phylogeny , Viral Proteins/chemistry , Viral Proteins/genetics , Viral Proteins/metabolism , Yellow Fever/epidemiology , Yellow fever virus/chemistry , Yellow fever virus/classification , Yellow fever virus/isolation & purification
18.
Sci Rep ; 7(1): 7385, 2017 08 07.
Article in English | MEDLINE | ID: mdl-28785067

ABSTRACT

Yellow fever virus (YFV) strains circulating in the Americas belong to two distinct genotypes (I and II) that have diversified into several concurrent enzootic lineages. Since 1999, YFV genotype I has spread outside endemic regions and its recent (2017) reemergence in non-endemic Southeastern Brazilian states fuels one of the largest epizootic of jungle Yellow Fever registered in the country. To better understand this phenomenon, we reconstructed the phylodynamics of YFV American genotypes using sequences from nine countries sampled along 60 years, including strains from Brazilian 2017 outbreak. Our analyses reveals that YFV genotypes I and II follow roughly similar evolutionary and demographic dynamics until the early 1990s, when a dramatic change in the diversification process of the genotype I occurred associated with the emergence and dissemination of a new lineage (here called modern). Trinidad and Tobago was the most likely source of the YFV modern-lineage that spread to Brazil and Venezuela around the late 1980s, where it replaced all lineages previously circulating. The modern-lineage caused all major YFV outbreaks detected in non-endemic South American regions since 2000, including the 2017 Brazilian outbreak, and its dissemination was coupled to the accumulation of several amino acid substitutions particularly within non-structural viral proteins.


Subject(s)
Disease Outbreaks , Yellow Fever/epidemiology , Yellow fever virus/genetics , Brazil/epidemiology , Evolution, Molecular , Genotype , Humans , Phylogeny , Phylogeography , Trinidad and Tobago , Viral Proteins/genetics , Yellow Fever/virology , Yellow fever virus/classification
19.
Int J Infect Dis ; 60: 93-95, 2017 Jul.
Article in English | MEDLINE | ID: mdl-28623054

ABSTRACT

Yellow fever virus (YFV), as the first proven human-pathogenic virus, is still a major public health problem with a dramatic upsurge in recent years. This is a report on four imported cases of yellow fever virus into China identified by whole genome sequencing. Phylogenetic analysis was performed and the results showed that these four viruses were highly homologous with Angola 71 strains (AY968064). In addition, effective mutations of amino acids were not observed in the E protein domain of four viruses, thus confirming the effectiveness of the YFV-17D vaccine (X03700). Although there is low risk of local transmission in most part of China, the increasing public health risk of YF caused by international exchange should not be ignored.


Subject(s)
Genome, Viral , Yellow Fever/virology , Yellow fever virus/genetics , Adult , Amino Acid Sequence , Angola , China , Humans , Male , Middle Aged , Mutation , Phylogeny , RNA, Viral/blood , RNA, Viral/isolation & purification , RNA, Viral/urine , Travel , Viral Proteins/chemistry , Viral Proteins/genetics , Whole Genome Sequencing , Yellow Fever/blood , Yellow Fever/urine , Yellow Fever Vaccine/genetics , Yellow Fever Vaccine/immunology , Yellow fever virus/classification , Yellow fever virus/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...