Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
2.
Sci Rep ; 12(1): 22175, 2022 12 22.
Artículo en Inglés | MEDLINE | ID: mdl-36550362

RESUMEN

Sero-surveillance can monitor and project disease burden and risk. However, SARS-CoV-2 antibody test results can produce false positive results, limiting their efficacy as a sero-surveillance tool. False positive SARS-CoV-2 antibody results are associated with malaria exposure, and understanding this association is essential to interpret sero-surveillance results from malaria-endemic countries. Here, pre-pandemic samples from eight malaria endemic and non-endemic countries and four continents were tested by ELISA to measure SARS-CoV-2 Spike S1 subunit reactivity. Individuals with acute malaria infection generated substantial SARS-CoV-2 reactivity. Cross-reactivity was not associated with reactivity to other human coronaviruses or other SARS-CoV-2 proteins, as measured by peptide and protein arrays. ELISAs with deglycosylated and desialated Spike S1 subunits revealed that cross-reactive antibodies target sialic acid on N-linked glycans of the Spike protein. The functional activity of cross-reactive antibodies measured by neutralization assays showed that cross-reactive antibodies did not neutralize SARS-CoV-2 in vitro. Since routine use of glycosylated or sialated assays could result in false positive SARS-CoV-2 antibody results in malaria endemic regions, which could overestimate exposure and population-level immunity, we explored methods to increase specificity by reducing cross-reactivity. Overestimating population-level exposure to SARS-CoV-2 could lead to underestimates of risk of continued COVID-19 transmission in sub-Saharan Africa.


Asunto(s)
COVID-19 , Malaria , Humanos , Glicoproteína de la Espiga del Coronavirus , SARS-CoV-2 , Anticuerpos Antivirales , Reacciones Cruzadas , Ácido N-Acetilneuramínico , Epítopos
3.
Science ; 378(6615): eabq5358, 2022 10 07.
Artículo en Inglés | MEDLINE | ID: mdl-36108049

RESUMEN

Investment in severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) sequencing in Africa over the past year has led to a major increase in the number of sequences that have been generated and used to track the pandemic on the continent, a number that now exceeds 100,000 genomes. Our results show an increase in the number of African countries that are able to sequence domestically and highlight that local sequencing enables faster turnaround times and more-regular routine surveillance. Despite limitations of low testing proportions, findings from this genomic surveillance study underscore the heterogeneous nature of the pandemic and illuminate the distinct dispersal dynamics of variants of concern-particularly Alpha, Beta, Delta, and Omicron-on the continent. Sustained investment for diagnostics and genomic surveillance in Africa is needed as the virus continues to evolve while the continent faces many emerging and reemerging infectious disease threats. These investments are crucial for pandemic preparedness and response and will serve the health of the continent well into the 21st century.


Asunto(s)
COVID-19 , Monitoreo Epidemiológico , Pandemias , SARS-CoV-2 , África/epidemiología , COVID-19/epidemiología , COVID-19/virología , Genómica , Humanos , SARS-CoV-2/genética
5.
Lancet Infect Dis ; 22(6): 891-900, 2022 06.
Artículo en Inglés | MEDLINE | ID: mdl-35298901

RESUMEN

BACKGROUND: The Democratic Republic of the Congo has confronted 13 outbreaks of Ebola virus disease since 1976. Rapid diagnostic tests (RDTs) detecting viral antigens have been developed to circumvent difficulties encountered with RT-PCR for diagnosis in remote low-resource settings, but there is still uncertainty about their performance characteristics and usability during outbreaks. We aimed to assess the field performance of three antigen detection RDTs compared with the gold-standard Cepheid GeneXpert Ebola assay results. METHODS: We conducted a retrospective, multicentre observational study using complete and de-identified databases of five mobile laboratories (managed by the Institut National de Recherche Biomédicale) to assess the performance of three Ebola virus disease RDTs (QuickNavi-Ebola, OraQuick Ebola Rapid Antigen Test, and Coris EBOLA Ag K-SeT rapid test) run on blood samples of patients with suspected Ebola virus disease in direct comparison with the Cepheid GeneXpert Ebola assay reference test during the 2018-20 outbreak in the eastern Democratic Republic of the Congo. We estimated the sensitivity and specificity of each test through generalised linear mixed models against the GeneXpert Ebola assay reference test and corrected for cycle threshold value and random site effects. FINDINGS: 719 (7·9%) of 9157 samples had a positive GeneXpert Ebola assay result. The QuickNavi-Ebola RDT had a sensitivity of 87·4% (95% CI 63·6-96·8) around the mean cycle threshold value and a specificity of 99·6% (99·3-99·8). The OraQuick Ebola Rapid Antigen Test had a sensitivity of 57·4% (95% CI 38·8-75·8) and specificity of 98·3% (97·5-99·0), and the Coris EBOLA Ag K-SeT rapid test had a sensitivity of 38·9% (23·0-63·6) against the GeneXpert Ebola assay reference and specificity of 97·4% (85·3-99·6). The QuickNavi-Ebola RDT showed a robust performance with good sensitivity, particularly with increasing viral loads (ie, low cycle threshold values), and specificity. INTERPRETATION: The three RDTs evaluated did not achieve the desired sensitivity and specificity of the WHO target product profile. Although the RDTs cannot triage and rule out Ebola virus infection among clinical suspects, they can still help to sort people with suspected Ebola virus disease into high-risk and low-risk groups while waiting for GeneXpert Ebola assay reference testing. FUNDING: None. TRANSLATION: For the French translation of the abstract see Supplementary Materials section.


Asunto(s)
Ebolavirus , Fiebre Hemorrágica Ebola , República Democrática del Congo/epidemiología , Pruebas Diagnósticas de Rutina , Brotes de Enfermedades , Ebolavirus/genética , Fiebre Hemorrágica Ebola/diagnóstico , Fiebre Hemorrágica Ebola/epidemiología , Humanos , Estudios Retrospectivos , Sensibilidad y Especificidad
6.
Nature ; 597(7877): 539-543, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-34526718

RESUMEN

Seven years after the declaration of the first epidemic of Ebola virus disease in Guinea, the country faced a new outbreak-between 14 February and 19 June 2021-near the epicentre of the previous epidemic1,2. Here we use next-generation sequencing to generate complete or near-complete genomes of Zaire ebolavirus from samples obtained from 12 different patients. These genomes form a well-supported phylogenetic cluster with genomes from the previous outbreak, which indicates that the new outbreak was not the result of a new spillover event from an animal reservoir. The 2021 lineage shows considerably lower divergence than would be expected during sustained human-to-human transmission, which suggests a persistent infection with reduced replication or a period of latency. The resurgence of Zaire ebolavirus from humans five years after the end of the previous outbreak of Ebola virus disease reinforces the need for long-term medical and social care for patients who survive the disease, to reduce the risk of re-emergence and to prevent further stigmatization.


Asunto(s)
Brotes de Enfermedades , Ebolavirus/genética , Ebolavirus/aislamiento & purificación , Fiebre Hemorrágica Ebola/epidemiología , Fiebre Hemorrágica Ebola/virología , Modelos Biológicos , Animales , República Democrática del Congo/epidemiología , Brotes de Enfermedades/estadística & datos numéricos , Ebolavirus/clasificación , Femenino , Guinea/epidemiología , Fiebre Hemorrágica Ebola/transmisión , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Masculino , Infección Persistente/virología , Filogenia , Sobrevivientes , Factores de Tiempo , Zoonosis Virales/transmisión , Zoonosis Virales/virología
7.
medRxiv ; 2021 May 12.
Artículo en Inglés | MEDLINE | ID: mdl-34013301

RESUMEN

Individuals with acute malaria infection generated high levels of antibodies that cross-react with the SARS-CoV-2 Spike protein. Cross-reactive antibodies specifically recognized the sialic acid moiety on N-linked glycans of the Spike protein and do not neutralize in vitro SARS-CoV-2. Sero-surveillance is critical for monitoring and projecting disease burden and risk during the pandemic; however, routine use of Spike protein-based assays may overestimate SARS-CoV-2 exposure and population-level immunity in malaria-endemic countries.

8.
Nat Med ; 27(4): 710-716, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-33846610

RESUMEN

On 1 August 2018, the Democratic Republic of the Congo (DRC) declared its tenth Ebola virus disease (EVD) outbreak. To aid the epidemiologic response, the Institut National de Recherche Biomédicale (INRB) implemented an end-to-end genomic surveillance system, including sequencing, bioinformatic analysis and dissemination of genomic epidemiologic results to frontline public health workers. We report 744 new genomes sampled between 27 July 2018 and 27 April 2020 generated by this surveillance effort. Together with previously available sequence data (n = 48 genomes), these data represent almost 24% of all laboratory-confirmed Ebola virus (EBOV) infections in DRC in the period analyzed. We inferred spatiotemporal transmission dynamics from the genomic data as new sequences were generated, and disseminated the results to support epidemiologic response efforts. Here we provide an overview of how this genomic surveillance system functioned, present a full phylodynamic analysis of 792 Ebola genomes from the Nord Kivu outbreak and discuss how the genomic surveillance data informed response efforts and public health decision making.


Asunto(s)
Brotes de Enfermedades , Ebolavirus/genética , Genómica , Fiebre Hemorrágica Ebola/epidemiología , Fiebre Hemorrágica Ebola/genética , Análisis de Secuencia de ADN , Congo/epidemiología , Vacunas contra el Virus del Ébola/inmunología , Genoma Viral , Fiebre Hemorrágica Ebola/transmisión , Fiebre Hemorrágica Ebola/virología , Filogenia , Recurrencia , Reinfección/virología , Análisis Espacio-Temporal
9.
N Engl J Med ; 384(13): 1240-1247, 2021 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-33789012

RESUMEN

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.).


Asunto(s)
Ebolavirus/genética , Fiebre Hemorrágica Ebola/transmisión , Adulto , Teorema de Bayes , República Democrática del Congo/epidemiología , Vacunas contra el Virus del Ébola/inmunología , Ebolavirus/aislamiento & purificación , Resultado Fatal , Genoma Viral , Fiebre Hemorrágica Ebola/diagnóstico , Fiebre Hemorrágica Ebola/epidemiología , Fiebre Hemorrágica Ebola/terapia , Humanos , Masculino , Mutación , Filogenia , ARN Viral/sangre , Recurrencia
10.
Afr J Lab Med ; 9(2): 1041, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32934915

RESUMEN

BACKGROUND: Past and recent outbreaks have highlighted the vulnerability of humans to infectious diseases, which represent serious economic and health security threats. A paradigm shift in the management of sanitary crises is urgently needed. Based on lessons from the 2014 Ebola outbreak, the Praesens Foundation has developed an all-terrain mobile biosafety laboratory (MBS-Lab) for effective field diagnostics capabilities. OBJECTIVE: The aim of the study was to train African teams and run a field evaluation of the MBS-Lab, including robustness, technical and operational sustainability, biosafety, connectivity, turn-around times for testing and result delivery. METHODS: The MBS-Lab was deployed in Senegal in October 2017 for a six-month field assessment under various ecological conditions and was mobilised during the dengue outbreaks in 2017 and 2018. RESULTS: The MBS-Lab can be considered an off-grid solution that addresses field challenges with regard to working conditions, mobility, deployment, environment and personnel safety. Blood (n = 398) and nasal swab (n = 113) samples were collected from 460 study participants for molecular screening for acute febrile illnesses and respiratory infections. The results showed that malaria (particularly in Kédougou) and upper respiratory tract infections remain problematic. Suspected dengue samples were tested on board during the dengue outbreaks in 2017 (882 tests; 128 confirmed cases) and 2018 (1736 tests; 202 confirmed cases). CONCLUSION: The MBS-Lab is an innovative solution for outbreak response, even in remote areas. The study demonstrated successful local ownership and community engagement. The MBS-Lab can also be considered an open mobile healthcare platform that offers various opportunities for field-deployable, point-of-care technologies for surveillance programmes.

11.
Emerg Infect Dis ; 26(10): 2460-2464, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-32946728

RESUMEN

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.


Asunto(s)
Infecciones por Hantavirus , Fiebre Hemorrágica con Síndrome Renal , Orthohantavirus , Virus Seoul , Orthohantavirus/genética , Infecciones por Hantavirus/epidemiología , Infecciones por Hantavirus/veterinaria , Fiebre Hemorrágica con Síndrome Renal/epidemiología , Fiebre Hemorrágica con Síndrome Renal/veterinaria , Humanos , Ratas , Senegal/epidemiología , Seúl , Virus Seoul/genética
12.
Virus Res ; 275: 197802, 2020 01 02.
Artículo en Inglés | MEDLINE | ID: mdl-31697989

RESUMEN

An increasing number of insect-specific viruses are found around the world. Very recently, a new group of insect-specific viruses, the Mesoniviridae family, was discovered in Africa, Asia, North America and Australia. Here we report the first detection and isolation of a new virus belonging to Mesonivirus genus in Senegal, West Africa. The so-called Dianke virus was detected in 21 species of arthropods trapped in the eastern part of the country. Male individuals were also infected, supporting vertical transmission assertion of insect specific viruses. As described for other mesoniviruses, no viral replication was observed after inoculation of mammalian cells. Viral replication in mosquito cells was blocked at a temperature of 37 °C, highlighting the importance of thermal conditions in Mesonivirus host restriction. Similar to our study, where a diverse range of arthropod vectors were found infected by the new virus, several studies have detected mesonivirus infection in mosquitoes with concerns for human health. It has been shown that dual infections in mosquito can alter viral infectivity. Due to their extensive geographic distribution and host range, as well as their use as potential disease control agents in vector populations, more studies should be done for a better knowledge of arthropod-restricted viruses prevalence and diversity.


Asunto(s)
Aedes/virología , Nidovirales/clasificación , Filogenia , Animales , Vectores Artrópodos/virología , Virus de Insectos/clasificación , Virus de Insectos/aislamiento & purificación , Masculino , Mosquitos Vectores/virología , Nidovirales/aislamiento & purificación , ARN Viral/genética , Senegal , Temperatura , Replicación Viral
13.
Lancet Infect Dis ; 19(6): 648-657, 2019 06.
Artículo en Inglés | MEDLINE | ID: mdl-31000464

RESUMEN

BACKGROUND: The real-time generation of information about pathogen genomes has become a vital goal for transmission analysis and characterisation in rapid outbreak responses. In response to the recently established genomic capacity in the Democratic Republic of the Congo, we explored the real-time generation of genomic information at the start of the 2018 Ebola virus disease (EVD) outbreak in North Kivu Province. METHODS: We used targeted-enrichment sequencing to produce two coding-complete Ebola virus genomes 5 days after declaration of the EVD outbreak in North Kivu. Subsequent sequencing efforts yielded an additional 46 genomes. Genomic information was used to assess early transmission, medical countermeasures, and evolution of Ebola virus. FINDINGS: The genomic information demonstrated that the EVD outbreak in the North Kivu and Ituri Provinces was distinct from the 2018 EVD outbreak in Équateur Province of the Democratic Republic of the Congo. Primer and probe mismatches to Ebola virus were identified in silico for all deployed diagnostic PCR assays, with the exception of the Cepheid GeneXpert GP assay. INTERPRETATION: The first two coding-complete genomes provided actionable information in real-time for the deployment of the rVSVΔG-ZEBOV-GP Ebola virus envelope glycoprotein vaccine, available therapeutics, and sequence-based diagnostic assays. Based on the mutations identified in the Ebola virus surface glycoprotein (GP12) observed in all 48 genomes, deployed monoclonal antibody therapeutics (mAb114 and ZMapp) should be efficacious against the circulating Ebola virus variant. Rapid Ebola virus genomic characterisation should be included in routine EVD outbreak response procedures to ascertain efficacy of medical countermeasures. FUNDING: Defense Biological Product Assurance Office.


Asunto(s)
Anticuerpos Monoclonales/genética , Antivirales/uso terapéutico , Vacunas contra el Virus del Ébola/uso terapéutico , Ebolavirus/genética , Genómica , Fiebre Hemorrágica Ebola/tratamiento farmacológico , Fiebre Hemorrágica Ebola/epidemiología , República Democrática del Congo/epidemiología , Brotes de Enfermedades , Humanos , Contramedidas Médicas , Estudios Retrospectivos
14.
Lancet Infect Dis ; 19(6): 641-647, 2019 06.
Artículo en Inglés | MEDLINE | ID: mdl-31000465

RESUMEN

BACKGROUND: The 2018 Ebola virus disease (EVD) outbreak in Équateur Province, Democratic Republic of the Congo, began on May 8, and was declared over on July 24; it resulted in 54 documented cases and 33 deaths. We did a retrospective genomic characterisation of the outbreak and assessed potential therapeutic agents and vaccine (medical countermeasures). METHODS: We used target-enrichment sequencing to produce Ebola virus genomes from samples obtained in the 2018 Équateur Province outbreak. Combining these genomes with genomes associated with known outbreaks from GenBank, we constructed a maximum-likelihood phylogenetic tree. In-silico analyses were used to assess potential mismatches between the outbreak strain and the probes and primers of diagnostic assays and the antigenic sites of the experimental rVSVΔG-ZEBOV-GP vaccine and therapeutics. An in-vitro flow cytometry assay was used to assess the binding capability of the individual components of the monoclonal antibody cocktail ZMapp. FINDINGS: A targeted sequencing approach produced 16 near-complete genomes. Phylogenetic analysis of these genomes and 1011 genomes from GenBank revealed a distinct cluster, confirming a new Ebola virus variant, for which we propose the name "Tumba". This new variant appears to have evolved at a slower rate than other Ebola virus variants (0·69 × 10-3 substitutions per site per year with "Tumba" vs 1·06 × 10-3 substitutions per site per year without "Tumba"). We found few sequence mismatches in the assessed assay target regions and antigenic sites. We identified nine amino acid changes in the Ebola virus surface glycoprotein, of which one resulted in reduced binding of the 13C6 antibody within the ZMapp cocktail. INTERPRETATION: Retrospectively, we show the feasibility of using genomics to rapidly characterise a new Ebola virus variant within the timeframe of an outbreak. Phylogenetic analysis provides further indications that these variants are evolving at differing rates. Rapid in-silico analyses can direct in-vitro experiments to quickly assess medical countermeasures. FUNDING: Defense Biological Product Assurance Office.


Asunto(s)
Antivirales/uso terapéutico , Brotes de Enfermedades , Vacunas contra el Virus del Ébola/uso terapéutico , Ebolavirus/genética , Genómica , Fiebre Hemorrágica Ebola/tratamiento farmacológico , Fiebre Hemorrágica Ebola/epidemiología , República Democrática del Congo/epidemiología , Humanos , Estudios Retrospectivos
16.
One Health ; 3: 23-28, 2017 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-28616499

RESUMEN

Wesselsbron disease is a neglected mosquito transmitted Flavivirus infection that causes abortions and has teratogenic effects on sheep and cattle in Africa. Human can also be infected. The detection of human or animal cases is complicated by the non-specific symptoms close to Rift Valley Fever (RVF) in domestic livestock species or Dengue like syndrome in humans. Then, these detections are usually made during RVF investigations in sheep. These domestic animals should take a role in the life cycle of the virus but some evidences of Wesselsbron virus (WSLV) presence in wild animals suggest that the latter may be involved in the virus maintenance in nature. However, the reservoir status of wild vertebrate in general and rodents particularly for WSLV is only based on an isolation from a Cape short-eared gerbil in southern Africa. Most of WSLV isolations are from southern parts of Africa even if it has been found in western and central Africa or Madagascar. In Senegal, there are serological evidences of WSLV circulation in human since the 1970s and some isolations, the last one of which dates back in 1992. Despite the detection of the virus on mosquitoes until the 2000s in different parts of the country, no new human case has been noted. In this paper, we report the WSLV re-emergence in eastern Senegal in 2013 with 2 human cases and its first isolation from a black rat Rattus rattus. Sequencing analyses show the circulation of the same strain between these humans and the commensal rodent. The putative impact on WSLV transmission to human populations could be more important if the reservoir status of the black rat is confirmed. Focused survey in human populations, specific entomological and mammalogical investigations would permit a better understanding of the life cycle of the virus and its impact on public health.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...