Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 243
Filter
Add more filters

Publication year range
1.
Cell ; 167(4): 1079-1087.e5, 2016 11 03.
Article in English | MEDLINE | ID: mdl-27814505

ABSTRACT

The 2013-2016 outbreak of Ebola virus (EBOV) in West Africa was the largest recorded. It began following the cross-species transmission of EBOV from an animal reservoir, most likely bats, into humans, with phylogenetic analysis revealing the co-circulation of several viral lineages. We hypothesized that this prolonged human circulation led to genomic changes that increased viral transmissibility in humans. We generated a synthetic glycoprotein (GP) construct based on the earliest reported isolate and introduced amino acid substitutions that defined viral lineages. Mutant GPs were used to generate a panel of pseudoviruses, which were used to infect different human and bat cell lines. These data revealed that specific amino acid substitutions in the EBOV GP have increased tropism for human cells, while reducing tropism for bat cells. Such increased infectivity may have enhanced the ability of EBOV to transmit among humans and contributed to the wide geographic distribution of some viral lineages.


Subject(s)
Biological Evolution , Ebolavirus/physiology , Hemorrhagic Fever, Ebola/virology , Host Specificity , Africa, Western/epidemiology , Animals , Chiroptera/virology , Disease Outbreaks , Ebolavirus/classification , Ebolavirus/genetics , Ebolavirus/pathogenicity , Hemorrhagic Fever, Ebola/epidemiology , Hemorrhagic Fever, Ebola/transmission , Humans , Mutation , Phylogeny , Viral Envelope Proteins/genetics , Viral Envelope Proteins/metabolism , Zoonoses
2.
PLoS Biol ; 20(10): e3001864, 2022 10.
Article in English | MEDLINE | ID: mdl-36288328

ABSTRACT

The explosive emergence of Zika virus (ZIKV) across the Pacific and Americas since 2007 was associated with hundreds of thousands of human cases and severe outcomes, including congenital microcephaly caused by ZIKV infection during pregnancy. Although ZIKV was first isolated in Uganda, Africa has so far been exempt from large-scale ZIKV epidemics, despite widespread susceptibility among African human populations. A possible explanation for this pattern is natural variation among populations of the primary vector of ZIKV, the mosquito Aedes aegypti. Globally invasive populations of Ae. aegypti outside of Africa are considered effective ZIKV vectors because they are human specialists with high intrinsic ZIKV susceptibility, whereas African populations of Ae. aegypti across the species' native range are predominantly generalists with low intrinsic ZIKV susceptibility, making them less likely to spread viruses in the human population. We test this idea by studying a notable exception to the patterns observed across most of Africa: Cape Verde experienced a large ZIKV outbreak in 2015 to 2016. We find that local Ae. aegypti in Cape Verde have substantial human-specialist ancestry, show a robust behavioral preference for human hosts, and exhibit increased susceptibility to ZIKV infection, consistent with a key role for variation among mosquito populations in ZIKV epidemiology. These findings suggest that similar human-specialist populations of Ae. aegypti in the nearby Sahel region of West Africa, which may be expanding in response to rapid urbanization, could serve as effective vectors for ZIKV in the future.


Subject(s)
Aedes , Epidemics , Zika Virus Infection , Zika Virus , Animals , Humans , Zika Virus/physiology , Cabo Verde , Saliva , Mosquito Vectors
3.
Emerg Infect Dis ; 30(4): 770-774, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38526209

ABSTRACT

In 2020, a sylvatic dengue virus serotype 2 infection outbreak resulted in 59 confirmed dengue cases in Kedougou, Senegal, suggesting those strains might not require adaptation to reemerge into urban transmission cycles. Large-scale genomic surveillance and updated molecular diagnostic tools are needed to effectively prevent dengue virus infections in Senegal.


Subject(s)
Dengue Virus , Dengue , Humans , Dengue Virus/genetics , Senegal/epidemiology , Serogroup , Environment , Dengue/epidemiology
4.
N Engl J Med ; 384(13): 1240-1247, 2021 04 01.
Article in English | MEDLINE | ID: mdl-33789012

ABSTRACT

During the 2018-2020 Ebola virus disease (EVD) outbreak in North Kivu province in the Democratic Republic of Congo, EVD was diagnosed in a patient who had received the recombinant vesicular stomatitis virus-based vaccine expressing a ZEBOV glycoprotein (rVSV-ZEBOV) (Merck). His treatment included an Ebola virus (EBOV)-specific monoclonal antibody (mAb114), and he recovered within 14 days. However, 6 months later, he presented again with severe EVD-like illness and EBOV viremia, and he died. We initiated epidemiologic and genomic investigations that showed that the patient had had a relapse of acute EVD that led to a transmission chain resulting in 91 cases across six health zones over 4 months. (Funded by the Bill and Melinda Gates Foundation and others.).


Subject(s)
Ebolavirus/genetics , Hemorrhagic Fever, Ebola/transmission , Adult , Bayes Theorem , Democratic Republic of the Congo/epidemiology , Ebola Vaccines/immunology , Ebolavirus/isolation & purification , Fatal Outcome , Genome, Viral , Hemorrhagic Fever, Ebola/diagnosis , Hemorrhagic Fever, Ebola/epidemiology , Hemorrhagic Fever, Ebola/therapy , Humans , Male , Mutation , Phylogeny , RNA, Viral/blood , Recurrence
5.
Emerg Infect Dis ; 29(9): 1808-1817, 2023 09.
Article in English | MEDLINE | ID: mdl-37610149

ABSTRACT

Historically low levels of seasonal influenza circulation were reported during the first years of the COVID-19 pandemic and were mainly attributed to implementation of nonpharmaceutical interventions. In tropical regions, influenza's seasonality differs largely, and data on this topic are scarce. We analyzed data from Senegal's sentinel syndromic surveillance network before and after the start of the COVID-19 pandemic to assess changes in influenza circulation. We found that influenza shows year-round circulation in Senegal and has 2 distinct epidemic peaks: during January-March and during the rainy season in August-October. During 2021-2022, the expected January-March influenza peak completely disappeared, corresponding to periods of active SARS-CoV-2 circulation. We noted an unexpected influenza epidemic peak during May-July 2022. The observed reciprocal circulation of SARS-CoV-2 and influenza suggests that factors such as viral interference might be at play and should be further investigated in tropical settings.


Subject(s)
COVID-19 , Influenza, Human , Humans , COVID-19/epidemiology , SARS-CoV-2 , Senegal/epidemiology , Influenza, Human/epidemiology , Pandemics
6.
J Med Virol ; 95(1): e28347, 2023 01.
Article in English | MEDLINE | ID: mdl-36424699

ABSTRACT

Globally, 390 million people are at risk of dengue infection and over the past 50 years, the virus incidence increased thirty-fold. In Senegal, an unprecedented occurrence of outbreaks and sporadic cases have been noticed since 2017. In October 2018, an outbreak of Dengue virus 2 (DENV-2) was reported in the north of Senegal affecting multiple areas including Saint-Louis, Richard Toll, and Rosso which are located at the border with Mauritania. Of these 173 blood specimen samples collected from patients, 27 were positive for dengue by quantitative reverse transcription PCR (qRT-PCR), and eight were serologically confirmed to be positive for DENV immunoglobulin M (IgM). Serotyping using qRT-PCR reveals that isolates were positive for DENV-2. A subset of DENV-2 positive samples was selected and subjected to whole-genome sequencing followed by phylogenetic analysis. Analysis of six nearly complete genome sequences revealed that the isolates belong to the cosmopolitan genotype and are closely related to the Mauritanian strains detected between 2017 and 2018 and those detected in many West African countries such as Burkina Faso or Cote d'Ivoire. Our results suggest a transboundary circulation of the DENV-2 cosmopolitan genotype between Senegal and Mauritania and call for a need for coordinated surveillance of arboviruses between these two countries. Interestingly, a high level of homology between West African isolates highlights endemicity and calls for the set-up of subregional viral genomic surveillance which will lead to a better understanding of viral dynamics, transmission, and spread across Africa.


Subject(s)
Dengue Virus , Dengue , Humans , Dengue/epidemiology , Senegal/epidemiology , Phylogeny , Disease Outbreaks , Genotype , Burkina Faso , Serogroup
7.
J Med Virol ; 95(4): e28700, 2023 04.
Article in English | MEDLINE | ID: mdl-36951314

ABSTRACT

Yellow fever (YF) virus is a mosquito-borne virus belonging to the Flaviviridae family that circulates in tropical and subtropical areas of Africa and South America. Despite the availability of an effective vaccine, YF remains a threat to travelers, residents of endemic areas, and unvaccinated populations. YF vaccination and natural infection both induce the production of neutralizing antibodies. Serological diagnostic methods detecting YF virus-specific antibodies demonstrate high levels of cross-reactivities with other flaviviruses. To date, the plaque reduction neutralization test (PRNT) is the most specific serological test for the differentiation of flavivirus infections and is considered the reference method for detecting YF neutralizing antibodies and assessing the protective immune response following vaccination. In this study, we developed and validated a YF PRNT. We optimized different parameters including cell concentration and virus-serum neutralization time period and then assessed the intra- and inter-assay precisions, dilutability, specificity, and lower limit of quantification (LLOQ) using international standard YF serum, sera from vaccinees and human specimens collected through YF surveillance. The YF PRNT has shown good robustness and 100% of intra-assay precision, 95.6% of inter-assay precision, 100% of specificity, 100% of LLOQ, and 95.3% of dilutability. The test is, therefore, suitable for use in the YF diagnostic as well as evaluation of the YF vaccine neutralizing antibody response and risk assessment studies.


Subject(s)
Vaccines , Yellow Fever Vaccine , Yellow Fever , Humans , Yellow Fever/diagnosis , Yellow Fever/prevention & control , Neutralization Tests , Yellow fever virus , Antibodies, Neutralizing , Antibodies, Viral
8.
Emerg Infect Dis ; 28(10): 2027-2034, 2022 10.
Article in English | MEDLINE | ID: mdl-36148906

ABSTRACT

Environmental surveillance for poliovirus is increasingly used in poliovirus eradication efforts as a supplement to acute flaccid paralysis (AFP) surveillance. Environmental surveillance was officially established in 2017 in Senegal, where no poliovirus had been detected since 2010. We tested sewage samples from 2 sites in Dakar monthly for polioviruses. We identified a vaccine-derived poliovirus serotype 2 on January 19, 2021, from a sample collected on December 24, 2020; by December 31, 2021, we had detected 70 vaccine-derived poliovirus serotype 2 isolates circulating in 7 of 14 regions in Senegal. Sources included 18 AFP cases, 20 direct contacts, 17 contacts in the community, and 15 sewage samples. Phylogenetic analysis revealed the circulation of 2 clusters and provided evidence on the virus introduction from Guinea. Because novel oral polio vaccine serotype 2 was used for response activities throughout Senegal, we recommend expanding environmental surveillance into other regions.


Subject(s)
Poliomyelitis , Poliovirus , Humans , Environmental Monitoring , Phylogeny , Poliomyelitis/epidemiology , Poliomyelitis/prevention & control , Poliovirus/genetics , Poliovirus Vaccine, Oral/adverse effects , Senegal/epidemiology , Serogroup , Sewage
9.
J Gen Virol ; 103(4)2022 04.
Article in English | MEDLINE | ID: mdl-35412967

ABSTRACT

Crimean-Congo haemorrhagic fever virus (CCHFV) is the medically most important member of the rapidly expanding bunyaviral family Nairoviridae. Traditionally, CCHFV isolates have been assigned to six distinct genotypes. Here, the International Committee on Taxonomy of Viruses (ICTV) Nairoviridae Study Group outlines the reasons for the recent decision to re-classify genogroup VI (aka Europe-2 or AP-92-like) as a distinct virus, Aigai virus (AIGV).


Subject(s)
Hemorrhagic Fever Virus, Crimean-Congo , Hemorrhagic Fever, Crimean , Genotype , Hemorrhagic Fever Virus, Crimean-Congo/genetics , Humans
10.
J Med Virol ; 94(11): 5593-5600, 2022 11.
Article in English | MEDLINE | ID: mdl-35879861

ABSTRACT

To assess the genetic diversity of circulating dengue virus 2 (DENV-2) in Senegal, we analyzed nine newly generated complete genomes of strains isolated during the 2018 outbreaks and 06 sequences obtained in 2018 and 2019 from Thiès and Rosso, respectively. Phylogenetic analyses revealed that Senegalese strains belonged to the cosmopolitan genotype of DENV-2, but we observed intragenotype variability leading to a divergence in two clades associated with specific geographic distribution. We report two DENV-2 variants belonging to two distinct clades. Isolates from the "Northern clade" (n = 8) harbored three nonsynonymous mutations (V1183M, R1405K, P2266T) located respectively on NS2A, NS2B, and NS4A, while isolates from the "Western clade" (n = 7) had two nonsynonymous mutations (V1185E, V3214E) located respectively in the NS2A and NS5 genes. These findings call for phylogeographic analysis to investigate routes of introductions, dispersal patterns, and in-depth in vitro and functional study to elucidate the impact of observed mutations on viral fitness, spread, epidemiology, and pathology.


Subject(s)
Dengue Virus , Dengue , Dengue/epidemiology , Genotype , Humans , Phylogeny , Phylogeography , Senegal/epidemiology
11.
Nature ; 534(7606): 267-71, 2016 06 09.
Article in English | MEDLINE | ID: mdl-27279226

ABSTRACT

Zika virus (ZIKV) is an arbovirus belonging to the genus Flavivirus (family Flaviviridae) and was first described in 1947 in Uganda following blood analyses of sentinel Rhesus monkeys. Until the twentieth century, the African and Asian lineages of the virus did not cause meaningful infections in humans. However, in 2007, vectored by Aedes aegypti mosquitoes, ZIKV caused the first noteworthy epidemic on the Yap Island in Micronesia. Patients experienced fever, skin rash, arthralgia and conjunctivitis. From 2013 to 2015, the Asian lineage of the virus caused further massive outbreaks in New Caledonia and French Polynesia. In 2013, ZIKV reached Brazil, later spreading to other countries in South and Central America. In Brazil, the virus has been linked to congenital malformations, including microcephaly and other severe neurological diseases, such as Guillain-Barré syndrome. Despite clinical evidence, direct experimental proof showing that the Brazilian ZIKV (ZIKV(BR)) strain causes birth defects remains absent. Here we demonstrate that ZIKV(BR) infects fetuses, causing intrauterine growth restriction, including signs of microcephaly, in mice. Moreover, the virus infects human cortical progenitor cells, leading to an increase in cell death. We also report that the infection of human brain organoids results in a reduction of proliferative zones and disrupted cortical layers. These results indicate that ZIKV(BR) crosses the placenta and causes microcephaly by targeting cortical progenitor cells, inducing cell death by apoptosis and autophagy, and impairing neurodevelopment. Our data reinforce the growing body of evidence linking the ZIKV(BR) outbreak to the alarming number of cases of congenital brain malformations. Our model can be used to determine the efficiency of therapeutic approaches to counteracting the harmful impact of ZIKV(BR) in human neurodevelopment.


Subject(s)
Disease Models, Animal , Microcephaly/virology , Zika Virus/pathogenicity , Animals , Apoptosis , Autophagy , Brain/pathology , Brain/virology , Brazil/epidemiology , Cell Proliferation , Female , Fetal Growth Retardation/pathology , Fetal Growth Retardation/virology , Fetus/virology , Mice , Microcephaly/epidemiology , Microcephaly/etiology , Microcephaly/pathology , Neural Stem Cells/pathology , Neural Stem Cells/virology , Organoids/pathology , Organoids/virology , Placenta/virology , Pregnancy , Zika Virus Infection/complications , Zika Virus Infection/epidemiology , Zika Virus Infection/pathology , Zika Virus Infection/virology
12.
Nature ; 530(7589): 228-232, 2016 Feb 11.
Article in English | MEDLINE | ID: mdl-26840485

ABSTRACT

The Ebola virus disease epidemic in West Africa is the largest on record, responsible for over 28,599 cases and more than 11,299 deaths. Genome sequencing in viral outbreaks is desirable to characterize the infectious agent and determine its evolutionary rate. Genome sequencing also allows the identification of signatures of host adaptation, identification and monitoring of diagnostic targets, and characterization of responses to vaccines and treatments. The Ebola virus (EBOV) genome substitution rate in the Makona strain has been estimated at between 0.87 × 10(-3) and 1.42 × 10(-3) mutations per site per year. This is equivalent to 16-27 mutations in each genome, meaning that sequences diverge rapidly enough to identify distinct sub-lineages during a prolonged epidemic. Genome sequencing provides a high-resolution view of pathogen evolution and is increasingly sought after for outbreak surveillance. Sequence data may be used to guide control measures, but only if the results are generated quickly enough to inform interventions. Genomic surveillance during the epidemic has been sporadic owing to a lack of local sequencing capacity coupled with practical difficulties transporting samples to remote sequencing facilities. To address this problem, here we devise a genomic surveillance system that utilizes a novel nanopore DNA sequencing instrument. In April 2015 this system was transported in standard airline luggage to Guinea and used for real-time genomic surveillance of the ongoing epidemic. We present sequence data and analysis of 142 EBOV samples collected during the period March to October 2015. We were able to generate results less than 24 h after receiving an Ebola-positive sample, with the sequencing process taking as little as 15-60 min. We show that real-time genomic surveillance is possible in resource-limited settings and can be established rapidly to monitor outbreaks.


Subject(s)
Ebolavirus/genetics , Epidemiological Monitoring , Genome, Viral/genetics , Hemorrhagic Fever, Ebola/epidemiology , Hemorrhagic Fever, Ebola/virology , Sequence Analysis, DNA/instrumentation , Sequence Analysis, DNA/methods , Aircraft , Disease Outbreaks/statistics & numerical data , Ebolavirus/classification , Ebolavirus/pathogenicity , Guinea/epidemiology , Humans , Mutagenesis/genetics , Mutation Rate , Time Factors
13.
Emerg Infect Dis ; 27(1)2021 01.
Article in English | MEDLINE | ID: mdl-33256890

ABSTRACT

We investigated the dynamics of seroconversion in severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection. During March 29-May 22, 2020, we collected serum samples and associated clinical data from 177 persons in London, UK, who had SARS-CoV-2 infection. We measured IgG against SARS-CoV-2 and compared antibody levels with patient outcomes, demographic information, and laboratory characteristics. We found that 2.0%-8.5% of persons did not seroconvert 3-6 weeks after infection. Persons who seroconverted were older, were more likely to have concurrent conditions, and had higher levels of inflammatory markers. Non-White persons had higher antibody concentrations than those who identified as White; these concentrations did not decline during follow-up. Serologic assay results correlated with disease outcome, race, and other risk factors for severe SARS-CoV-2 infection. Serologic assays can be used in surveillance to clarify the duration and protective nature of humoral responses to SARS-CoV-2 infection.


Subject(s)
COVID-19/blood , COVID-19/immunology , Immunoglobulin G/blood , SARS-CoV-2 , Seroconversion , Adult , Aged , Antibodies, Viral/blood , COVID-19/physiopathology , Enzyme-Linked Immunosorbent Assay , Female , Humans , Male , Middle Aged , Reverse Transcriptase Polymerase Chain Reaction
15.
Anal Chem ; 93(4): 2627-2634, 2021 02 02.
Article in English | MEDLINE | ID: mdl-33471510

ABSTRACT

In March 2020, the SARS-CoV-2 virus outbreak was declared as a world pandemic by the World Health Organization (WHO). The only measures for controlling the outbreak are testing and isolation of infected cases. Molecular real-time polymerase chain reaction (PCR) assays are very sensitive but require highly equipped laboratories and well-trained personnel. In this study, a rapid point-of-need detection method was developed to detect the RNA-dependent RNA polymerase (RdRP), envelope protein (E), and nucleocapsid protein (N) genes of SARS-CoV-2 based on the reverse transcription recombinase polymerase amplification (RT-RPA) assay. RdRP, E, and N RT-RPA assays required approximately 15 min to amplify 2, 15, and 15 RNA molecules of molecular standard/reaction, respectively. RdRP and E RT-RPA assays detected SARS-CoV-1 and 2 genomic RNA, whereas the N RT-RPA assay identified only SARS-CoV-2 RNA. All established assays did not cross-react with nucleic acids of other respiratory pathogens. The RT-RPA assay's clinical sensitivity and specificity in comparison to real-time RT-PCR (n = 36) were 94 and 100% for RdRP; 65 and 77% for E; and 83 and 94% for the N RT-RPA assay. The assays were deployed to the field, where the RdRP RT-RPA assays confirmed to produce the most accurate results in three different laboratories in Africa (n = 89). The RPA assays were run in a mobile suitcase laboratory to facilitate the deployment at point of need. The assays can contribute to speed up the control measures as well as assist in the detection of COVID-19 cases in low-resource settings.


Subject(s)
COVID-19/diagnosis , Real-Time Polymerase Chain Reaction/methods , Recombinases/metabolism , SARS-CoV-2/isolation & purification , COVID-19/virology , Humans , Sensitivity and Specificity
16.
BMC Infect Dis ; 21(1): 867, 2021 Aug 24.
Article in English | MEDLINE | ID: mdl-34429064

ABSTRACT

BACKGROUND: Dengue fever is a mosquito born disease associated with self-limited to life threatening illness. First detected in Senegal in the nineteenth century, and despite its growing incidence this last decade, significant knowledge gaps exist in our knowledge of genetic diversity of circulating strains. This study highlights the circulating serotypes and genotypes between January 2017 and December 2018 and their spatial and temporal distribution throughout all regions of Senegal. METHODS: We used 56 dengue virus (DENV) strains for the analysis collected from 11 sampling areas: 39 from all regions of Senegal, and 17 isolates from Thiès, a particular area of the country. Two real time RT-qPCR systems were used to confirm dengue infection and corresponding serotypes. For molecular characterization, CprM gene was sequenced and submitted to phylogenetic analysis for serotypes and genotypes assignment. RESULTS: Three dengue virus serotypes (DENV-1-3) were detected by all used methods. DENV-3 was detected in 50% (28/56) of the isolates, followed by DENV-1 and DENV-2, each representing 25% (14/56) of the isolates. DENV-3 belongs to genotype III, DENV-1 to genotype V and DENV-2 to Cosmopolitan genotype. Serotype 3 was detected in 7 sampling locations and a co-circulation of different serotypes was observed in Thiès, Fatick and Richard-toll. CONCLUSIONS: These results emphasize the need of continuous DENV surveillance in Senegal to detect DENV cases, to define circulating serotypes/genotypes and to prevent the spread and the occurrence of severe cases.


Subject(s)
Dengue Virus/genetics , Dengue/epidemiology , Dengue/diagnosis , Dengue Virus/isolation & purification , Humans , Phylogeny , Public Health Surveillance , Senegal/epidemiology , Serogroup , Spatial Analysis
17.
Nature ; 524(7563): 102-4, 2015 Aug 06.
Article in English | MEDLINE | ID: mdl-26106863

ABSTRACT

An epidemic of Ebola virus disease of unprecedented scale has been ongoing for more than a year in West Africa. As of 29 April 2015, there have been 26,277 reported total cases (of which 14,895 have been laboratory confirmed) resulting in 10,899 deaths. The source of the outbreak was traced to the prefecture of Guéckédou in the forested region of southeastern Guinea. The virus later spread to the capital, Conakry, and to the neighbouring countries of Sierra Leone, Liberia, Nigeria, Senegal and Mali. In March 2014, when the first cases were detected in Conakry, the Institut Pasteur of Dakar, Senegal, deployed a mobile laboratory in Donka hospital to provide diagnostic services to the greater Conakry urban area and other regions of Guinea. Through this process we sampled 85 Ebola viruses (EBOV) from patients infected from July to November 2014, and report their full genome sequences here. Phylogenetic analysis reveals the sustained transmission of three distinct viral lineages co-circulating in Guinea, including the urban setting of Conakry and its surroundings. One lineage is unique to Guinea and closely related to the earliest sampled viruses of the epidemic. A second lineage contains viruses probably reintroduced from neighbouring Sierra Leone on multiple occasions, while a third lineage later spread from Guinea to Mali. Each lineage is defined by multiple mutations, including non-synonymous changes in the virion protein 35 (VP35), glycoprotein (GP) and RNA-dependent RNA polymerase (L) proteins. The viral GP is characterized by a glycosylation site modification and mutations in the mucin-like domain that could modify the outer shape of the virion. These data illustrate the ongoing ability of EBOV to develop lineage-specific and potentially phenotypically important variation.


Subject(s)
Ebolavirus/genetics , Genetic Variation/genetics , Hemorrhagic Fever, Ebola/epidemiology , Hemorrhagic Fever, Ebola/virology , Mutation/genetics , Phylogeny , Ebolavirus/isolation & purification , Evolution, Molecular , Genome, Viral/genetics , Glycoproteins/genetics , Glycoproteins/metabolism , Glycosylation , Guinea/epidemiology , Hemorrhagic Fever, Ebola/transmission , Humans , Mali/epidemiology , Molecular Sequence Data , Mucins/chemistry , Nucleocapsid Proteins , Nucleoproteins/genetics , Protein Structure, Tertiary/genetics , RNA-Dependent RNA Polymerase/genetics , Sierra Leone/epidemiology , Viral Core Proteins/genetics
18.
Emerg Infect Dis ; 26(10): 2460-2464, 2020 10.
Article in English | MEDLINE | ID: mdl-32946728

ABSTRACT

Hantaviruses cause hemorrhagic fever in humans worldwide. However, few hantavirus surveillance campaigns occur in Africa. We detected Seoul orthohantavirus in black rats in Senegal, although we did not find serologic evidence of this disease in humans. These findings highlight the need for increased surveillance of hantaviruses in this region.


Subject(s)
Hantavirus Infections , Hemorrhagic Fever with Renal Syndrome , Orthohantavirus , Seoul virus , Orthohantavirus/genetics , Hantavirus Infections/epidemiology , Hantavirus Infections/veterinary , Hemorrhagic Fever with Renal Syndrome/epidemiology , Hemorrhagic Fever with Renal Syndrome/veterinary , Humans , Rats , Senegal/epidemiology , Seoul , Seoul virus/genetics
20.
Emerg Infect Dis ; 26(6): 1084-1090, 2020 06.
Article in English | MEDLINE | ID: mdl-32441631

ABSTRACT

During 2015-2016, Cape Verde, an island nation off the coast of West Africa, experienced a Zika virus (ZIKV) outbreak involving 7,580 suspected Zika cases and 18 microcephaly cases. Analysis of the complete genomes of 3 ZIKV isolates from the outbreak indicated the strain was of the Asian (not African) lineage. The Cape Verde ZIKV sequences formed a distinct monophylogenetic group and possessed 1-2 (T659A, I756V) unique amino acid changes in the envelope protein. Phylogeographic and serologic evidence support earlier introduction of this lineage into Cape Verde, possibly from northeast Brazil, between June 2014 and August 2015, suggesting cryptic circulation of the virus before the initial wave of cases were detected in October 2015. These findings underscore the utility of genomic-scale epidemiology for outbreak investigations.


Subject(s)
Microcephaly , Zika Virus Infection , Zika Virus , Africa, Western , Brazil/epidemiology , Cabo Verde , Disease Outbreaks , Genomics , Humans , Microcephaly/epidemiology , Zika Virus/genetics , Zika Virus Infection/epidemiology
SELECTION OF CITATIONS
SEARCH DETAIL