Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 42
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Cell ; 183(7): 1901-1912.e9, 2020 12 23.
Artículo en Inglés | MEDLINE | ID: mdl-33248470

RESUMEN

Long-term severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) shedding was observed from the upper respiratory tract of a female immunocompromised individual with chronic lymphocytic leukemia and acquired hypogammaglobulinemia. Shedding of infectious SARS-CoV-2 was observed up to 70 days, and of genomic and subgenomic RNA up to 105 days, after initial diagnosis. The infection was not cleared after the first treatment with convalescent plasma, suggesting a limited effect on SARS-CoV-2 in the upper respiratory tract of this individual. Several weeks after a second convalescent plasma transfusion, SARS-CoV-2 RNA was no longer detected. We observed marked within-host genomic evolution of SARS-CoV-2 with continuous turnover of dominant viral variants. However, replication kinetics in Vero E6 cells and primary human alveolar epithelial tissues were not affected. Our data indicate that certain immunocompromised individuals may shed infectious virus longer than previously recognized. Detection of subgenomic RNA is recommended in persistently SARS-CoV-2-positive individuals as a proxy for shedding of infectious virus.


Asunto(s)
COVID-19/inmunología , Inmunodeficiencia Variable Común/inmunología , Leucemia Linfocítica Crónica de Células B/inmunología , SARS-CoV-2/aislamiento & purificación , Anciano , Anticuerpos Antivirales/sangre , Anticuerpos Antivirales/inmunología , COVID-19/complicaciones , COVID-19/virología , Inmunodeficiencia Variable Común/sangre , Inmunodeficiencia Variable Común/complicaciones , Inmunodeficiencia Variable Común/virología , Femenino , Humanos , Leucemia Linfocítica Crónica de Células B/sangre , Leucemia Linfocítica Crónica de Células B/complicaciones , Leucemia Linfocítica Crónica de Células B/virología , Infecciones del Sistema Respiratorio/sangre , Infecciones del Sistema Respiratorio/complicaciones , Infecciones del Sistema Respiratorio/inmunología , Infecciones del Sistema Respiratorio/virología , SARS-CoV-2/inmunología , SARS-CoV-2/patogenicidad
2.
PLoS Pathog ; 18(9): e1010828, 2022 09.
Artículo en Inglés | MEDLINE | ID: mdl-36136995

RESUMEN

Spillover of sarbecoviruses from animals to humans has resulted in outbreaks of severe acute respiratory syndrome SARS-CoVs and the ongoing COVID-19 pandemic. Efforts to identify the origins of SARS-CoV-1 and -2 has resulted in the discovery of numerous animal sarbecoviruses-the majority of which are only distantly related to known human pathogens and do not infect human cells. The receptor binding domain (RBD) on sarbecoviruses engages receptor molecules on the host cell and mediates cell invasion. Here, we tested the receptor tropism and serological cross reactivity for RBDs from two sarbecoviruses found in Russian horseshoe bats. While these two viruses are in a viral lineage distinct from SARS-CoV-1 and -2, the RBD from one virus, Khosta 2, was capable of using human ACE2 to facilitate cell entry. Viral pseudotypes with a recombinant, SARS-CoV-2 spike encoding for the Khosta 2 RBD were resistant to both SARS-CoV-2 monoclonal antibodies and serum from individuals vaccinated for SARS-CoV-2. Our findings further demonstrate that sarbecoviruses circulating in wildlife outside of Asia also pose a threat to global health and ongoing vaccine campaigns against SARS-CoV-2.


Asunto(s)
COVID-19 , Quirópteros , Enzima Convertidora de Angiotensina 2 , Animales , Anticuerpos Monoclonales , Anticuerpos Antivirales , COVID-19/prevención & control , Vacunas contra la COVID-19 , Humanos , Pandemias/prevención & control , SARS-CoV-2 , Glicoproteína de la Espiga del Coronavirus
3.
Biol Lett ; 18(1): 20210427, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-34982955

RESUMEN

Host-virus association data underpin research into the distribution and eco-evolutionary correlates of viral diversity and zoonotic risk across host species. However, current knowledge of the wildlife virome is inherently constrained by historical discovery effort, and there are concerns that the reliability of ecological inference from host-virus data may be undermined by taxonomic and geographical sampling biases. Here, we evaluate whether current estimates of host-level viral diversity in wild mammals are stable enough to be considered biologically meaningful, by analysing a comprehensive dataset of discovery dates of 6571 unique mammal host-virus associations between 1930 and 2018. We show that virus discovery rates in mammal hosts are either constant or accelerating, with little evidence of declines towards viral richness asymptotes, even in highly sampled hosts. Consequently, inference of relative viral richness across host species has been unstable over time, particularly in bats, where intensified surveillance since the early 2000s caused a rapid rearrangement of species' ranked viral richness. Our results illustrate that comparative inference of host-level virus diversity across mammals is highly sensitive to even short-term changes in sampling effort. We advise caution to avoid overinterpreting patterns in current data, since it is feasible that an analysis conducted today could draw quite different conclusions than one conducted only a decade ago.


Asunto(s)
Quirópteros , Virus , Animales , Evolución Biológica , Mamíferos , Reproducibilidad de los Resultados
4.
Emerg Infect Dis ; 27(12): 3052-3062, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34808078

RESUMEN

Middle East respiratory syndrome coronavirus (MERS-CoV) infects humans and dromedary camels and is responsible for an ongoing outbreak of severe respiratory illness in humans in the Middle East. Although some mutations found in camel-derived MERS-CoV strains have been characterized, most natural variation found across MERS-CoV isolates remains unstudied. We report on the environmental stability, replication kinetics, and pathogenicity of several diverse isolates of MERS-CoV, as well as isolates of severe acute respiratory syndrome coronavirus 2, to serve as a basis of comparison with other stability studies. Although most MERS-CoV isolates had similar stability and pathogenicity in our experiments, the camel-derived isolate C/KSA/13 had reduced surface stability, and another camel isolate, C/BF/15, had reduced pathogenicity in a small animal model. These results suggest that although betacoronaviruses might have similar environmental stability profiles, individual variation can influence this phenotype, underscoring the need for continual global viral surveillance.


Asunto(s)
COVID-19 , Coronavirus del Síndrome Respiratorio de Oriente Medio , Aerosoles , Animales , Camelus , Humanos , Coronavirus del Síndrome Respiratorio de Oriente Medio/genética , SARS-CoV-2 , Virulencia , Zoonosis
5.
J Infect Dis ; 221(Suppl 4): S383-S388, 2020 05 11.
Artículo en Inglés | MEDLINE | ID: mdl-31784761

RESUMEN

Viruses in the genus Henipavirus encompass 2 highly pathogenic emerging zoonotic pathogens, Hendra virus (HeV) and Nipah virus (NiV). Despite the impact on human health, there is currently limited full-genome sequence information available for henipaviruses. This lack of full-length genomes hampers our ability to understand the molecular drivers of henipavirus emergence. Furthermore, rapidly deployable viral genome sequencing can be an integral part of outbreak response and epidemiological investigations to study transmission chains. In this study, we describe the development of a reverse-transcription, long-range polymerase chain reaction (LRPCR) assay for efficient genome amplification of NiV, HeV, and a related non-pathogenic henipavirus, Cedar virus (CedPV). We then demonstrated the utility of our method by amplifying partial viral genomes from 6 HeV-infected tissue samples from Syrian hamsters and 4 tissue samples from a NiV-infected African green monkey with viral loads as low as 52 genome copies/mg. We subsequently sequenced the amplified genomes on the portable Oxford Nanopore MinION platform and analyzed the data using a newly developed field-deployable bioinformatic pipeline. Our LRPCR assay allows amplification and sequencing of 2 or 4 amplicons in semi-nested reactions. Coupled with an easy-to-use bioinformatics pipeline, this method is particularly useful in the field during outbreaks in resource-poor environments.


Asunto(s)
Henipavirus/genética , Secuenciación de Nucleótidos de Alto Rendimiento/métodos , Reacción en Cadena de la Polimerasa/métodos , Genoma Viral , ARN Viral
6.
J Infect Dis ; 221(Suppl 4): S407-S413, 2020 05 11.
Artículo en Inglés | MEDLINE | ID: mdl-31682727

RESUMEN

Nipah virus (NiV) is a bat-borne zoonotic pathogen that can cause severe respiratory distress and encephalitis upon spillover into humans. NiV is capable of infecting a broad range of hosts including humans, pigs, ferrets, dogs, cats, hamsters, and at least 2 genera of bats. Little is known about the biology of NiV in the bat reservoir. In this study, we evaluate the potential for the Egyptian fruit bat (EFB), Rousettus aegyptiacus, to serve as a model organism for studying NiV in bats. Our data suggest that NiV does not efficiently replicate in EFBs in vivo. Furthermore, we show no seroconversion against NiV glycoprotein and a lack of viral replication in primary and immortalized EFB-derived cell lines. Our data show that despite using a conserved target for viral entry, NiV replication is limited in some bat species. We conclude that EFBs are not an appropriate organism to model NiV infection or transmission in bats.


Asunto(s)
Quirópteros/clasificación , Quirópteros/virología , Infecciones por Henipavirus/veterinaria , Virus Nipah/fisiología , Replicación Viral/fisiología , Animales , Infecciones por Henipavirus/virología , Especificidad de la Especie
7.
J Infect Dis ; 221(Suppl 4): S375-S382, 2020 05 11.
Artículo en Inglés | MEDLINE | ID: mdl-32034942

RESUMEN

Bat-borne zoonotic pathogens belonging to the family Paramxyoviridae, including Nipah and Hendra viruses, and the family Filoviridae, including Ebola and Marburg viruses, can cause severe disease and high mortality rates on spillover into human populations. Surveillance efforts for henipaviruses and filoviruses have been largely restricted to the Old World; however, recent studies suggest a potentially broader distribution for henipaviruses and filoviruses than previously recognized. In the current study, we screened for henipaviruses and filoviruses in New World bats collected across 4 locations in Trinidad near the coast of Venezuela. Bat tissue samples were screened using previously established reverse-transcription polymerase chain reaction assays. Serum were screened using a multiplex immunoassay to detect antibodies reactive with the envelope glycoprotein of viruses in the genus Henipavirus and the family Filoviridae. Serum samples were also screened by means of enzyme-linked immunosorbent assay for antibodies reactive with Nipah G and F glycoproteins. Of 84 serum samples, 28 were reactive with ≥1 henipavirus glycoprotein by ≥1 serological method, and 6 serum samples were reactive against ≥1 filovirus glycoproteins. These data provide evidence of potential circulation of viruses related to the henipaviruses and filoviruses in New World bats.


Asunto(s)
Quirópteros/virología , Infecciones por Filoviridae/veterinaria , Filoviridae , Infecciones por Henipavirus/veterinaria , Henipavirus , Animales , Quirópteros/sangre , Quirópteros/clasificación , Infecciones por Filoviridae/epidemiología , Infecciones por Filoviridae/virología , Infecciones por Henipavirus/epidemiología , Infecciones por Henipavirus/virología , Pruebas Serológicas , Trinidad y Tobago/epidemiología
8.
Emerg Infect Dis ; 26(9)2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32511089

RESUMEN

We found that environmental conditions affect the stability of severe acute respiratory syndrome coronavirus 2 in nasal mucus and sputum. The virus is more stable at low-temperature and low-humidity conditions, whereas warmer temperature and higher humidity shortened half-life. Although infectious virus was undetectable after 48 hours, viral RNA remained detectable for 7 days.


Asunto(s)
Betacoronavirus/genética , Infecciones por Coronavirus/virología , Moco/virología , Neumonía Viral/virología , ARN Viral/análisis , Esputo/virología , COVID-19 , Calor , Humanos , Humedad , Cavidad Nasal/virología , Pandemias , Estabilidad del ARN , SARS-CoV-2
9.
Emerg Infect Dis ; 26(9)2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32491983

RESUMEN

The coronavirus pandemic has created worldwide shortages of N95 respirators. We analyzed 4 decontamination methods for effectiveness in deactivating severe acute respiratory syndrome coronavirus 2 virus and effect on respirator function. Our results indicate that N95 respirators can be decontaminated and reused, but the integrity of respirator fit and seal must be maintained.


Asunto(s)
Betacoronavirus , Infecciones por Coronavirus/prevención & control , Descontaminación/métodos , Equipo Reutilizado , Pandemias/prevención & control , Neumonía Viral/prevención & control , Ventiladores Mecánicos/virología , COVID-19 , Infecciones por Coronavirus/virología , Humanos , Neumonía Viral/virología , SARS-CoV-2
10.
Emerg Infect Dis ; 26(9): 2205-2209, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32818404

RESUMEN

After the 2017 Ebola virus (EBOV) outbreak in Likati, a district in northern Democratic Republic of the Congo, we sampled small mammals from the location where the primary case-patient presumably acquired the infection. None tested positive for EBOV RNA or antibodies against EBOV, highlighting the ongoing challenge in detecting animal reservoirs for EBOV.


Asunto(s)
Ebolavirus , Fiebre Hemorrágica Ebola , Animales , Animales Salvajes , República Democrática del Congo/epidemiología , Brotes de Enfermedades , Ebolavirus/genética , Fiebre Hemorrágica Ebola/epidemiología , Humanos
11.
J Infect Dis ; 218(suppl_5): S301-S304, 2018 11 22.
Artículo en Inglés | MEDLINE | ID: mdl-30085166

RESUMEN

Sequencing viral genomes during an outbreak can facilitate response and containment efforts. In this study, we describe a reverse transcription long-range polymerase chain reaction for efficient amplification and sequencing of the Ebola virus (EBOV) genome in 2 seminested reactions. We demonstrate that our method remains robust with complex biological samples by amplifying and sequencing the EBOV genome from EBOV-infected nonhuman primates (NHPs). We further demonstrate that we are able to recover viral genomes from starting concentrations as low as 103 50% tissue culture infective dose (TCID50)/mL, suggesting that this method can be employed to sequence EBOV genomes from ecologically or clinically derived samples.


Asunto(s)
Ebolavirus/genética , Genoma Viral/genética , Fiebre Hemorrágica Ebola/virología , Reacción en Cadena de la Polimerasa/métodos , Animales , Brotes de Enfermedades , Humanos , Macaca fascicularis/virología , Análisis de Secuencia de ARN/métodos
12.
Trends Genet ; 31(4): 201-7, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25765920

RESUMEN

Population genomic studies have the potential to address many unresolved questions about microbial pathogens by facilitating the identification of genes underlying ecologically important traits, such as novel virulence factors and adaptations to humans or other host species. Additionally, this framework improves estimations of population demography and evolutionary history to accurately reconstruct recent epidemics and identify the molecular and environmental factors that resulted in the outbreak. The Lyme disease bacterium, Borrelia burgdorferi, exemplifies the power and promise of the application of population genomics to microbial pathogens. We discuss here the future of evolutionary studies in B. burgdorferi, focusing on the primary evolutionary forces of horizontal gene transfer, natural selection, and migration, as investigations transition from analyses of single genes to genomes.


Asunto(s)
Borrelia burgdorferi/genética , Evolución Molecular , Genética de Población , Genómica , Enfermedad de Lyme/microbiología , Borrelia burgdorferi/clasificación , Humanos , Filogenia , Filogeografía , Investigación , Selección Genética
13.
Bioinformatics ; 33(14): 2071-2077, 2017 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-28334194

RESUMEN

MOTIVATION: Population genomic analyses are often hindered by difficulties in obtaining sufficient numbers of genomes for analysis by DNA sequencing. Selective whole-genome amplification (SWGA) provides an efficient approach to amplify microbial genomes from complex backgrounds for sequence acquisition. However, the process of designing sets of primers for this method has many degrees of freedom and would benefit from an automated process to evaluate the vast number of potential primer sets. RESULTS: Here, we present swga , a program that identifies primer sets for SWGA and evaluates them for efficiency and selectivity. We used swga to design and test primer sets for the selective amplification of Wolbachia pipientis genomic DNA from infected Drosophila melanogaster and Mycobacterium tuberculosis from human blood. We identify primer sets that successfully amplify each against their backgrounds and describe a general method for using swga for arbitrary targets. In addition, we describe characteristics of primer sets that correlate with successful amplification, and present guidelines for implementation of SWGA to detect new targets. AVAILABILITY AND IMPLEMENTATION: Source code and documentation are freely available on https://www.github.com/eclarke/swga . The program is implemented in Python and C and licensed under the GNU Public License. CONTACT: ecl@mail.med.upenn.edu. SUPPLEMENTARY INFORMATION: Supplementary data are available at Bioinformatics online.


Asunto(s)
Genética de Población/métodos , Análisis de Secuencia de ADN/métodos , Programas Informáticos , Animales , Cartilla de ADN , Drosophila melanogaster/microbiología , Genoma Bacteriano , Genómica/métodos , Humanos , Mycobacterium tuberculosis/genética , Wolbachia/genética
15.
Virus Evol ; 10(1): vead079, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38361817

RESUMEN

Pathogen evolution is one of the least predictable components of disease emergence, particularly in nature. Here, building on principles established by the geographic mosaic theory of coevolution, we develop a quantitative, spatially explicit framework for mapping the evolutionary risk of viral emergence. Driven by interest in diseases like Severe Acute Respiratory Syndrome (SARS), Middle East Respiratory Syndrome (MERS), and Coronavirus disease 2019 (COVID-19), we examine the global biogeography of bat-origin betacoronaviruses, and find that coevolutionary principles suggest geographies of risk that are distinct from the hotspots and coldspots of host richness. Further, our framework helps explain patterns like a unique pool of merbecoviruses in the Neotropics, a recently discovered lineage of divergent nobecoviruses in Madagascar, and-most importantly-hotspots of diversification in southeast Asia, sub-Saharan Africa, and the Middle East that correspond to the site of previous zoonotic emergence events. Our framework may help identify hotspots of future risk that have also been previously overlooked, like West Africa and the Indian subcontinent, and may more broadly help researchers understand how host ecology shapes the evolution and diversity of pandemic threats.

16.
J Med Entomol ; 50(1): 43-51, 2013 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-23427651

RESUMEN

Seasonal variation in dengue virus transmission in northwestern Thailand is inversely related to the magnitude of diurnal temperature fluctuations, although mean temperature does not vary significantly across seasons. We tested the hypothesis that diurnal temperature fluctuations negatively influence epidemiologically important life-history traits of the primary dengue vector, Aedes aegypti (L.), compared with a constant 26 degrees C temperature. A large diurnal temperature range (DTR) (approximately equals 18 degrees C daily swing) extended immature development time (>1 d), lowered larval survival (approximately equals 6%), and reduced adult female reproductive output by 25% 14 d after blood feeding, relative to the constant 26 degreesC temperature. A small DTR (approximately equal 8 degrees C daily swing) led to a negligible or slightly positive effect on the life history traits tested. Our results indicate that there is a negative impact of large DTR on mosquito biology and are consistent with the hypothesis that, in at least some locations, large temperature fluctuations contribute to seasonal reduction in dengue virus transmission.


Asunto(s)
Aedes/crecimiento & desarrollo , Temperatura , Animales , Dengue/transmisión , Femenino , Fertilidad , Insectos Vectores , Masculino , Periodicidad
17.
Trends Microbiol ; 31(11): 1102-1110, 2023 11.
Artículo en Inglés | MEDLINE | ID: mdl-37277284

RESUMEN

Orthohantaviruses present a global public health threat; there are 58 distinct viruses currently recognized and case fatality of pathogenic orthohantaviruses ranges from <0.1% to 50%. An Old World versus New World dichotomy is frequently applied to distinguish human diseases caused by orthohantaviruses. However, this geographic grouping masks the importance of phylogeny and virus-host ecology in shaping orthohantavirus traits, especially since related arvicoline rodents and their orthohantaviruses are found in both regions. We argue that orthohantaviruses can be separated into three phylogenetically based rodent host groups with differences in key functional traits, including human disease, transmission route, and virus-host fidelity. This framework can help understand and predict traits of under-studied and newly discovered orthohantaviruses and guide public health and biosafety policy.


Asunto(s)
Infecciones por Hantavirus , Orthohantavirus , Virus ARN , Humanos , Filogenia , Reservorios de Enfermedades
18.
bioRxiv ; 2023 Oct 27.
Artículo en Inglés | MEDLINE | ID: mdl-37961540

RESUMEN

Orthopoxviruses (OPVs), including the causative agents of smallpox and mpox have led to devastating outbreaks in human populations worldwide. However, the discontinuation of smallpox vaccination, which also provides cross-protection against related OPVs, has diminished global immunity to OPVs more broadly. We apply machine learning models incorporating both host ecological and viral genomic features to predict likely reservoirs of OPVs. We demonstrate that incorporating viral genomic features in addition to host ecological traits enhanced the accuracy of potential OPV host predictions, highlighting the importance of host-virus molecular interactions in predicting potential host species. We identify hotspots for geographic regions rich with potential OPV hosts in parts of southeast Asia, equatorial Africa, and the Amazon, revealing high overlap between regions predicted to have a high number of potential OPV host species and those with the lowest smallpox vaccination coverage, indicating a heightened risk for the emergence or establishment of zoonotic OPVs. Our findings can be used to target wildlife surveillance, particularly related to concerns about mpox establishment beyond its historical range.

19.
medRxiv ; 2023 Dec 19.
Artículo en Inglés | MEDLINE | ID: mdl-38196653

RESUMEN

Human movement drives the transmission and spread of communicable pathogens. It is especially influential for emerging pathogens when population immunity is low and spillover events are rare. We digitized serial printed maps to measure transportation networks (roads and rivers) in Central and West Africa as proxies for population mobility to assess relationships between movement and Ebola transmission. We find that the lengths of roads and rivers in close proximity to spillover sites at or near the time of spillover events are significantly correlated with the number of EVD cases, particularly in the first 100 days of each outbreak. Early management and containment efforts along transportation networks may be beneficial in mitigation during the early days of transmission and spatial spread for Ebola outbreaks.

20.
Patterns (N Y) ; 4(6): 100738, 2023 Jun 09.
Artículo en Inglés | MEDLINE | ID: mdl-37409053

RESUMEN

Predicting host-virus interactions is fundamentally a network science problem. We develop a method for bipartite network prediction that combines a recommender system (linear filtering) with an imputation algorithm based on low-rank graph embedding. We test this method by applying it to a global database of mammal-virus interactions and thus show that it makes biologically plausible predictions that are robust to data biases. We find that the mammalian virome is under-characterized anywhere in the world. We suggest that future virus discovery efforts could prioritize the Amazon Basin (for its unique coevolutionary assemblages) and sub-Saharan Africa (for its poorly characterized zoonotic reservoirs). Graph embedding of the imputed network improves predictions of human infection from viral genome features, providing a shortlist of priorities for laboratory studies and surveillance. Overall, our study indicates that the global structure of the mammal-virus network contains a large amount of information that is recoverable, and this provides new insights into fundamental biology and disease emergence.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA