Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 25
Filtrar
Más filtros










Intervalo de año de publicación
1.
bioRxiv ; 2024 Jun 17.
Artículo en Inglés | MEDLINE | ID: mdl-38948765

RESUMEN

Modification of RNA with N6-methyladenosine (m6A) has gained attention in recent years as a general mechanism of gene regulation. In the liver, m6A, along with its associated machinery, has been studied as a potential biomarker of disease and cancer, with impacts on metabolism, cell cycle regulation, and pro-cancer state signaling. However these observational data have yet to be causally examined in vivo. For example, neither perturbation of the key m6A writers Mettl3 and Mettl14, nor the m6A readers Ythdf1 and Ythdf2 have been thoroughly mechanistically characterized in vivo as they have been in vitro. To understand the functions of these machineries, we developed mouse models and found that deleting Mettl14 led to progressive liver injury characterized by nuclear heterotypia, with changes in mRNA splicing, processing and export leading to increases in mRNA surveillance and recycling.

2.
Sci Signal ; 16(806): eadf5494, 2023 10 10.
Artículo en Inglés | MEDLINE | ID: mdl-37816090

RESUMEN

Interferons (IFNs) play crucial roles in antiviral defenses. Despite using the same Janus-activated kinase (JAK)-signal transducer and activator of transcription (STAT) signaling cascade, type I and III IFN receptors differ in the magnitude and dynamics of their signaling in terms of STAT phosphorylation, gene transcription, and antiviral responses. These differences are not due to ligand-binding affinity and receptor abundance. Here, we investigated the ability of the intracellular domains (ICDs) of IFN receptors to differentiate between type I and III IFN signaling. We engineered synthetic, heterodimeric type I and III IFN receptors that were stably expressed at similar amounts in human cells and responded to a common ligand. We found that our synthetic type I IFN receptors stimulated STAT phosphorylation and gene expression to greater extents than did the corresponding type III IFN receptors. Furthermore, we identified short "box motifs" within ICDs that bind to JAK1 that were sufficient to encode differences between the type I and III IFN receptors. Together, our results indicate that specific regions within the ICDs of IFN receptor subunits encode different downstream signaling strengths that enable type I and III IFN receptors to produce distinct signaling outcomes.


Asunto(s)
Interferón Tipo I , Receptores de Interferón , Humanos , Receptores de Interferón/genética , Receptores de Interferón/metabolismo , Ligandos , Interferones/metabolismo , Transducción de Señal , Interferón Tipo I/genética , Interferón Tipo I/metabolismo , Quinasas Janus/metabolismo , Fosforilación , Antivirales/farmacología , Factor de Transcripción STAT1/genética , Factor de Transcripción STAT1/metabolismo
3.
Nat Microbiol ; 8(10): 1846-1862, 2023 10.
Artículo en Inglés | MEDLINE | ID: mdl-37653008

RESUMEN

Bacterial populations are highly adaptive. They can respond to stress and survive in shifting environments. How the behaviours of individual bacteria vary during stress, however, is poorly understood. To identify and characterize rare bacterial subpopulations, technologies for single-cell transcriptional profiling have been developed. Existing approaches show some degree of limitation, for example, in terms of number of cells or transcripts that can be profiled. Due in part to these limitations, few conditions have been studied with these tools. Here we develop massively-parallel, multiplexed, microbial sequencing (M3-seq)-a single-cell RNA-sequencing platform for bacteria that pairs combinatorial cell indexing with post hoc rRNA depletion. We show that M3-seq can profile bacterial cells from different species under a range of conditions in single experiments. We then apply M3-seq to hundreds of thousands of cells, revealing rare populations and insights into bet-hedging associated with stress responses and characterizing phage infection.


Asunto(s)
Bacteriófagos , Bacteriófagos/genética , Bacterias/genética , ARN Ribosómico/genética , Secuenciación de Nucleótidos de Alto Rendimiento
4.
Nat Commun ; 14(1): 3866, 2023 06 30.
Artículo en Inglés | MEDLINE | ID: mdl-37391481

RESUMEN

Long non-coding RNAs (lncRNAs) are involved in numerous biological processes and are pivotal mediators of the immune response, yet little is known about their properties at the single-cell level. Here, we generate a multi-tissue bulk RNAseq dataset from Ebola virus (EBOV) infected and not-infected rhesus macaques and identified 3979 novel lncRNAs. To profile lncRNA expression dynamics in immune circulating single-cells during EBOV infection, we design a metric, Upsilon, to estimate cell-type specificity. Our analysis reveals that lncRNAs are expressed in fewer cells than protein-coding genes, but they are not expressed at lower levels nor are they more cell-type specific when expressed in the same number of cells. In addition, we observe that lncRNAs exhibit similar changes in expression patterns to those of protein-coding genes during EBOV infection, and are often co-expressed with known immune regulators. A few lncRNAs change expression specifically upon EBOV entry in the cell. This study sheds light on the differential features of lncRNAs and protein-coding genes and paves the way for future single-cell lncRNA studies.


Asunto(s)
Ebolavirus , Fiebre Hemorrágica Ebola , ARN Largo no Codificante , Animales , Fiebre Hemorrágica Ebola/genética , ARN Largo no Codificante/genética , Macaca mulatta , Ebolavirus/genética , Internalización del Virus
5.
Cell Genom ; 3(12): 100440, 2023 Dec 13.
Artículo en Inglés | MEDLINE | ID: mdl-38169842

RESUMEN

Ebola virus (EBOV) causes Ebola virus disease (EVD), marked by severe hemorrhagic fever; however, the mechanisms underlying the disease remain unclear. To assess the molecular basis of EVD across time, we performed RNA sequencing on 17 tissues from a natural history study of 21 rhesus monkeys, developing new methods to characterize host-pathogen dynamics. We identified alterations in host gene expression with previously unknown tissue-specific changes, including downregulation of genes related to tissue connectivity. EBOV was widely disseminated throughout the body; using a new, broadly applicable deconvolution method, we found that viral load correlated with increased monocyte presence. Patterns of viral variation between tissues differentiated primary infections from compartmentalized infections, and several variants impacted viral fitness in a EBOV/Kikwit minigenome system, suggesting that functionally significant variants can emerge during early infection. This comprehensive portrait of host-pathogen dynamics in EVD illuminates new features of pathogenesis and establishes resources to study other emerging pathogens.


Asunto(s)
Ebolavirus , Fiebre Hemorrágica Ebola , Fiebres Hemorrágicas Virales , Animales , Fiebre Hemorrágica Ebola/patología , Macaca mulatta , Ebolavirus/genética
6.
Science ; 371(6529)2021 02 05.
Artículo en Inglés | MEDLINE | ID: mdl-33303686

RESUMEN

Analysis of 772 complete severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) genomes from early in the Boston-area epidemic revealed numerous introductions of the virus, a small number of which led to most cases. The data revealed two superspreading events. One, in a skilled nursing facility, led to rapid transmission and significant mortality in this vulnerable population but little broader spread, whereas other introductions into the facility had little effect. The second, at an international business conference, produced sustained community transmission and was exported, resulting in extensive regional, national, and international spread. The two events also differed substantially in the genetic variation they generated, suggesting varying transmission dynamics in superspreading events. Our results show how genomic epidemiology can help to understand the link between individual clusters and wider community spread.


Asunto(s)
COVID-19/epidemiología , Genoma Viral , Filogenia , SARS-CoV-2/genética , Boston/epidemiología , COVID-19/transmisión , Brotes de Enfermedades , Monitoreo Epidemiológico , Humanos
7.
Cell ; 183(5): 1383-1401.e19, 2020 11 25.
Artículo en Inglés | MEDLINE | ID: mdl-33159858

RESUMEN

Ebola virus (EBOV) causes epidemics with high mortality yet remains understudied due to the challenge of experimentation in high-containment and outbreak settings. Here, we used single-cell transcriptomics and CyTOF-based single-cell protein quantification to characterize peripheral immune cells during EBOV infection in rhesus monkeys. We obtained 100,000 transcriptomes and 15,000,000 protein profiles, finding that immature, proliferative monocyte-lineage cells with reduced antigen-presentation capacity replace conventional monocyte subsets, while lymphocytes upregulate apoptosis genes and decline in abundance. By quantifying intracellular viral RNA, we identify molecular determinants of tropism among circulating immune cells and examine temporal dynamics in viral and host gene expression. Within infected cells, EBOV downregulates STAT1 mRNA and interferon signaling, and it upregulates putative pro-viral genes (e.g., DYNLL1 and HSPA5), nominating pathways the virus manipulates for its replication. This study sheds light on EBOV tropism, replication dynamics, and elicited immune response and provides a framework for characterizing host-virus interactions under maximum containment.


Asunto(s)
Ebolavirus/fisiología , Fiebre Hemorrágica Ebola/genética , Fiebre Hemorrágica Ebola/virología , Interacciones Huésped-Patógeno/genética , Análisis de la Célula Individual , Animales , Antígenos CD/metabolismo , Biomarcadores/metabolismo , Efecto Espectador , Diferenciación Celular , Proliferación Celular , Citocinas/metabolismo , Ebolavirus/genética , Chaperón BiP del Retículo Endoplásmico , Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Regulación Viral de la Expresión Génica , Fiebre Hemorrágica Ebola/inmunología , Fiebre Hemorrágica Ebola/patología , Antígenos de Histocompatibilidad Clase II/metabolismo , Interferones/genética , Interferones/metabolismo , Macaca mulatta , Macrófagos/metabolismo , Monocitos/metabolismo , Mielopoyesis , ARN Mensajero/genética , ARN Mensajero/metabolismo , Factores de Tiempo , Transcriptoma/genética
8.
Cell ; 183(5): 1325-1339.e21, 2020 11 25.
Artículo en Inglés | MEDLINE | ID: mdl-33080218

RESUMEN

Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is a recently identified coronavirus that causes the respiratory disease known as coronavirus disease 2019 (COVID-19). Despite the urgent need, we still do not fully understand the molecular basis of SARS-CoV-2 pathogenesis. Here, we comprehensively define the interactions between SARS-CoV-2 proteins and human RNAs. NSP16 binds to the mRNA recognition domains of the U1 and U2 splicing RNAs and acts to suppress global mRNA splicing upon SARS-CoV-2 infection. NSP1 binds to 18S ribosomal RNA in the mRNA entry channel of the ribosome and leads to global inhibition of mRNA translation upon infection. Finally, NSP8 and NSP9 bind to the 7SL RNA in the signal recognition particle and interfere with protein trafficking to the cell membrane upon infection. Disruption of each of these essential cellular functions acts to suppress the interferon response to viral infection. Our results uncover a multipronged strategy utilized by SARS-CoV-2 to antagonize essential cellular processes to suppress host defenses.


Asunto(s)
COVID-19/metabolismo , Interacciones Huésped-Patógeno , Biosíntesis de Proteínas , Empalme del ARN , SARS-CoV-2/metabolismo , Proteínas no Estructurales Virales/metabolismo , Células A549 , Animales , COVID-19/virología , Chlorocebus aethiops , Células HEK293 , Humanos , Interferones/metabolismo , Transporte de Proteínas , ARN Mensajero/metabolismo , ARN Ribosómico 18S/metabolismo , ARN Citoplasmático Pequeño/química , ARN Citoplasmático Pequeño/metabolismo , Partícula de Reconocimiento de Señal/química , Partícula de Reconocimiento de Señal/metabolismo , Células Vero , Proteínas no Estructurales Virales/química
9.
medRxiv ; 2020 Aug 25.
Artículo en Inglés | MEDLINE | ID: mdl-32869040

RESUMEN

SARS-CoV-2 has caused a severe, ongoing outbreak of COVID-19 in Massachusetts with 111,070 confirmed cases and 8,433 deaths as of August 1, 2020. To investigate the introduction, spread, and epidemiology of COVID-19 in the Boston area, we sequenced and analyzed 772 complete SARS-CoV-2 genomes from the region, including nearly all confirmed cases within the first week of the epidemic and hundreds of cases from major outbreaks at a conference, a nursing facility, and among homeless shelter guests and staff. The data reveal over 80 introductions into the Boston area, predominantly from elsewhere in the United States and Europe. We studied two superspreading events covered by the data, events that led to very different outcomes because of the timing and populations involved. One produced rapid spread in a vulnerable population but little onward transmission, while the other was a major contributor to sustained community transmission, including outbreaks in homeless populations, and was exported to several other domestic and international sites. The same two events differed significantly in the number of new mutations seen, raising the possibility that SARS-CoV-2 superspreading might encompass disparate transmission dynamics. Our results highlight the failure of measures to prevent importation into MA early in the outbreak, underscore the role of superspreading in amplifying an outbreak in a major urban area, and lay a foundation for contact tracing informed by genetic data.

10.
Nat Commun ; 11(1): 4131, 2020 08 17.
Artículo en Inglés | MEDLINE | ID: mdl-32807807

RESUMEN

Recent outbreaks of viral hemorrhagic fevers (VHFs), including Ebola virus disease (EVD) and Lassa fever (LF), highlight the urgent need for sensitive, deployable tests to diagnose these devastating human diseases. Here we develop CRISPR-Cas13a-based (SHERLOCK) diagnostics targeting Ebola virus (EBOV) and Lassa virus (LASV), with both fluorescent and lateral flow readouts. We demonstrate on laboratory and clinical samples the sensitivity of these assays and the capacity of the SHERLOCK platform to handle virus-specific diagnostic challenges. We perform safety testing to demonstrate the efficacy of our HUDSON protocol in heat-inactivating VHF viruses before SHERLOCK testing, eliminating the need for an extraction. We develop a user-friendly protocol and mobile application (HandLens) to report results, facilitating SHERLOCK's use in endemic regions. Finally, we successfully deploy our tests in Sierra Leone and Nigeria in response to recent outbreaks.


Asunto(s)
Ebolavirus/patogenicidad , Fiebre Hemorrágica Ebola/diagnóstico , Fiebre de Lassa/diagnóstico , Virus Lassa/patogenicidad , Anticuerpos Antivirales , Repeticiones Palindrómicas Cortas Agrupadas y Regularmente Espaciadas/genética , Ebolavirus/genética , Fiebre Hemorrágica Ebola/virología , Fiebre de Lassa/virología , Virus Lassa/genética
11.
Viruses ; 12(1)2020 01 15.
Artículo en Inglés | MEDLINE | ID: mdl-31952352

RESUMEN

For highly pathogenic viruses, reporter assays that can be rapidly performed are critically needed to identify potentially functional mutations for further study under maximal containment (e.g., biosafety level 4 [BSL-4]). The Ebola virus nucleoprotein (NP) plays multiple essential roles during the viral life cycle, yet few tools exist to study the protein under BSL-2 or equivalent containment. Therefore, we adapted reporter assays to measure NP oligomerization and virion-like particle (VLP) production in live cells and further measured transcription and replication using established minigenome assays. As a proof-of-concept, we examined the NP-R111C substitution, which emerged during the 2013‒2016 Western African Ebola virus disease epidemic and rose to high frequency. NP-R111C slightly increased NP oligomerization and VLP budding but slightly decreased transcription and replication. By contrast, a synthetic charge-reversal mutant, NP-R111E, greatly increased oligomerization but abrogated transcription and replication. These results are intriguing in light of recent structures of NP oligomers, which reveal that the neighboring residue, K110, forms a salt bridge with E349 on adjacent NP molecules. By developing and utilizing multiple reporter assays, we find that the NP-111 position mediates a complex interplay between NP's roles in protein structure, virion budding, and transcription and replication.


Asunto(s)
Aminoácidos/química , Ebolavirus/genética , Genoma Viral , Proteínas de la Nucleocápside/química , Liberación del Virus , Aminoácidos/genética , Ebolavirus/química , Ebolavirus/fisiología , Células HEK293 , Humanos , Proteínas de la Nucleocápside/genética , Prueba de Estudio Conceptual , Virión/fisiología , Ensamble de Virus
12.
Mol Cell ; 76(5): 826-837.e11, 2019 12 05.
Artículo en Inglés | MEDLINE | ID: mdl-31607545

RESUMEN

The CRISPR effector Cas13 could be an effective antiviral for single-stranded RNA (ssRNA) viruses because it programmably cleaves RNAs complementary to its CRISPR RNA (crRNA). Here, we computationally identify thousands of potential Cas13 crRNA target sites in hundreds of ssRNA viral species that can potentially infect humans. We experimentally demonstrate Cas13's potent activity against three distinct ssRNA viruses: lymphocytic choriomeningitis virus (LCMV); influenza A virus (IAV); and vesicular stomatitis virus (VSV). Combining this antiviral activity with Cas13-based diagnostics, we develop Cas13-assisted restriction of viral expression and readout (CARVER), an end-to-end platform that uses Cas13 to detect and destroy viral RNA. We further screen hundreds of crRNAs along the LCMV genome to evaluate how conservation and target RNA nucleotide content influence Cas13's antiviral activity. Our results demonstrate that Cas13 can be harnessed to target a wide range of ssRNA viruses and CARVER's potential broad utility for rapid diagnostic and antiviral drug development.


Asunto(s)
Proteínas Asociadas a CRISPR/metabolismo , Sistemas CRISPR-Cas , Repeticiones Palindrómicas Cortas Agrupadas y Regularmente Espaciadas , Marcación de Gen/métodos , Estabilidad del ARN , Virus ARN/enzimología , ARN Viral/metabolismo , Células A549 , Animales , Proteínas Asociadas a CRISPR/genética , Chlorocebus aethiops , Perros , Escherichia coli/enzimología , Escherichia coli/genética , Células HEK293 , Humanos , Células de Riñón Canino Madin Darby , Virus ARN/genética , ARN Viral/genética , Células Vero
13.
Nat Biotechnol ; 37(2): 160-168, 2019 02.
Artículo en Inglés | MEDLINE | ID: mdl-30718881

RESUMEN

Metagenomic sequencing has the potential to transform microbial detection and characterization, but new tools are needed to improve its sensitivity. Here we present CATCH, a computational method to enhance nucleic acid capture for enrichment of diverse microbial taxa. CATCH designs optimal probe sets, with a specified number of oligonucleotides, that achieve full coverage of, and scale well with, known sequence diversity. We focus on applying CATCH to capture viral genomes in complex metagenomic samples. We design, synthesize, and validate multiple probe sets, including one that targets the whole genomes of the 356 viral species known to infect humans. Capture with these probe sets enriches unique viral content on average 18-fold, allowing us to assemble genomes that could not be recovered without enrichment, and accurately preserves within-sample diversity. We also use these probe sets to recover genomes from the 2018 Lassa fever outbreak in Nigeria and to improve detection of uncharacterized viral infections in human and mosquito samples. The results demonstrate that CATCH enables more sensitive and cost-effective metagenomic sequencing.


Asunto(s)
Biología Computacional/métodos , Genoma Viral , Metagenoma , Metagenómica , Animales , Culicidae/virología , Brotes de Enfermedades , Biblioteca de Genes , Variación Genética , Genómica , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Fiebre de Lassa/virología , Nigeria/epidemiología , Sondas de Oligonucleótidos , Oligonucleótidos/genética , Análisis de Secuencia de ADN , Virosis
14.
Sci Rep ; 8(1): 5939, 2018 04 12.
Artículo en Inglés | MEDLINE | ID: mdl-29651117

RESUMEN

Lassa fever, a hemorrhagic fever caused by Lassa virus (LASV), is endemic in West Africa. It is difficult to distinguish febrile illnesses that are common in West Africa from Lassa fever based solely on a patient's clinical presentation. The field performance of recombinant antigen-based Lassa fever immunoassays was compared to that of quantitative polymerase chain assays (qPCRs) using samples from subjects meeting the case definition of Lassa fever presenting to Kenema Government Hospital in Sierra Leone. The recombinant Lassa virus (ReLASV) enzyme-linked immunosorbant assay (ELISA) for detection of viral antigen in blood performed with 95% sensitivity and 97% specificity using a diagnostic standard that combined results of the immunoassays and qPCR. The ReLASV rapid diagnostic test (RDT), a lateral flow immunoassay based on paired monoclonal antibodies to the Josiah strain of LASV (lineage IV), performed with 90% sensitivity and 100% specificity. ReLASV immunoassays performed better than the most robust qPCR currently available, which had 82% sensitivity and 95% specificity. The performance characteristics of recombinant antigen-based Lassa virus immunoassays indicate that they can aid in the diagnosis of LASV Infection and inform the clinical management of Lassa fever patients.


Asunto(s)
Anticuerpos Antivirales/inmunología , Antígenos Virales/aislamiento & purificación , Fiebre de Lassa/diagnóstico , Virus Lassa/aislamiento & purificación , África Occidental , Anticuerpos Antivirales/genética , Antígenos Virales/genética , Humanos , Inmunoensayo/métodos , Inmunoglobulina M/inmunología , Fiebre de Lassa/inmunología , Fiebre de Lassa/virología , Virus Lassa/inmunología , Virus Lassa/patogenicidad , Proteínas Recombinantes/genética , Proteínas Recombinantes/inmunología , Sierra Leona , Estudios de Validación como Asunto
15.
Clin Infect Dis ; 65(8): 1400-1403, 2017 10 15.
Artículo en Inglés | MEDLINE | ID: mdl-28582513

RESUMEN

In one patient over time, we found that concentration of Ebola virus RNA in semen during recovery is remarkably higher than blood at peak illness. Virus in semen is replication-competent with no change in viral genome over time. Presence of sense RNA suggests replication in cells present in semen.


Asunto(s)
Ebolavirus/genética , Fiebre Hemorrágica Ebola/virología , Semen/virología , Adulto , Ebolavirus/clasificación , Genoma Viral/genética , Humanos , Masculino , ARN Viral/análisis , ARN Viral/genética , Carga Viral
16.
Nature ; 546(7658): 411-415, 2017 06 15.
Artículo en Inglés | MEDLINE | ID: mdl-28538734

RESUMEN

Although the recent Zika virus (ZIKV) epidemic in the Americas and its link to birth defects have attracted a great deal of attention, much remains unknown about ZIKV disease epidemiology and ZIKV evolution, in part owing to a lack of genomic data. Here we address this gap in knowledge by using multiple sequencing approaches to generate 110 ZIKV genomes from clinical and mosquito samples from 10 countries and territories, greatly expanding the observed viral genetic diversity from this outbreak. We analysed the timing and patterns of introductions into distinct geographic regions; our phylogenetic evidence suggests rapid expansion of the outbreak in Brazil and multiple introductions of outbreak strains into Puerto Rico, Honduras, Colombia, other Caribbean islands, and the continental United States. We find that ZIKV circulated undetected in multiple regions for many months before the first locally transmitted cases were confirmed, highlighting the importance of surveillance of viral infections. We identify mutations with possible functional implications for ZIKV biology and pathogenesis, as well as those that might be relevant to the effectiveness of diagnostic tests.


Asunto(s)
Filogenia , Infección por el Virus Zika/transmisión , Infección por el Virus Zika/virología , Virus Zika/genética , Virus Zika/aislamiento & purificación , Animales , Brasil/epidemiología , Colombia/epidemiología , Culicidae/virología , Brotes de Enfermedades/estadística & datos numéricos , Genoma Viral/genética , Mapeo Geográfico , Honduras/epidemiología , Humanos , Metagenoma/genética , Epidemiología Molecular , Mosquitos Vectores/virología , Mutación , Vigilancia en Salud Pública , Puerto Rico/epidemiología , Estados Unidos/epidemiología , Virus Zika/clasificación , Virus Zika/patogenicidad , Infección por el Virus Zika/diagnóstico , Infección por el Virus Zika/epidemiología
18.
Cell ; 167(4): 1088-1098.e6, 2016 11 03.
Artículo en Inglés | MEDLINE | ID: mdl-27814506

RESUMEN

The magnitude of the 2013-2016 Ebola virus disease (EVD) epidemic enabled an unprecedented number of viral mutations to occur over successive human-to-human transmission events, increasing the probability that adaptation to the human host occurred during the outbreak. We investigated one nonsynonymous mutation, Ebola virus (EBOV) glycoprotein (GP) mutant A82V, for its effect on viral infectivity. This mutation, located at the NPC1-binding site on EBOV GP, occurred early in the 2013-2016 outbreak and rose to high frequency. We found that GP-A82V had heightened ability to infect primate cells, including human dendritic cells. The increased infectivity was restricted to cells that have primate-specific NPC1 sequences at the EBOV interface, suggesting that this mutation was indeed an adaptation to the human host. GP-A82V was associated with increased mortality, consistent with the hypothesis that the heightened intrinsic infectivity of GP-A82V contributed to disease severity during the EVD epidemic.


Asunto(s)
Ebolavirus/genética , Ebolavirus/patogenicidad , Fiebre Hemorrágica Ebola/virología , Proteínas del Envoltorio Viral/química , Proteínas del Envoltorio Viral/genética , África Occidental/epidemiología , Sustitución de Aminoácidos , Animales , Callithrix , Proteínas Portadoras/química , Proteínas Portadoras/metabolismo , Cheirogaleidae , Citoplasma/virología , Ebolavirus/fisiología , Fiebre Hemorrágica Ebola/epidemiología , Humanos , Péptidos y Proteínas de Señalización Intracelular , Glicoproteínas de Membrana/química , Glicoproteínas de Membrana/metabolismo , Proteína Niemann-Pick C1 , Conformación Proteica en Hélice alfa , Proteínas del Envoltorio Viral/metabolismo , Virión/química , Virión/patogenicidad , Virulencia
19.
BMC Genomics ; 17: 707, 2016 09 05.
Artículo en Inglés | MEDLINE | ID: mdl-27595844

RESUMEN

BACKGROUND: Ebola virus is the causative agent of a severe syndrome in humans with a fatality rate that can approach 90 %. During infection, the host immune response is thought to become dysregulated, but the mechanisms through which this happens are not entirely understood. In this study, we analyze RNA sequencing data to determine the host response to Ebola virus infection in circulating immune cells. RESULTS: Approximately half of the 100 genes with the strongest early increases in expression were interferon-stimulated genes, such as ISG15, OAS1, IFIT2, HERC5, MX1 and DHX58. Other highly upregulated genes included cytokines CXCL11, CCL7, IL2RA, IL2R1, IL15RA, and CSF2RB, which have not been previously reported to change during Ebola virus infection. Comparing this response in two different models of exposure (intramuscular and aerosol) revealed a similar signature of infection. The strong innate response in the aerosol model was seen not only in circulating cells, but also in primary and secondary target tissues. Conversely, the innate immune response of vaccinated macaques was almost non-existent. This suggests that the innate response is a major aspect of the cellular response to Ebola virus infection in multiple tissues. CONCLUSIONS: Ebola virus causes a severe infection in humans that is associated with high mortality. The host immune response to virus infection is thought to be an important aspect leading to severe pathology, but the components of this overactive response are not well characterized. Here, we analyzed how circulating immune cells respond to the virus and found that there is a strong innate response dependent on active virus replication. This finding is in stark contrast to in vitro evidence showing a suppression of innate immune signaling, and it suggests that the strong innate response we observe in infected animals may be an important contributor to pathogenesis.


Asunto(s)
Ebolavirus/fisiología , Fiebre Hemorrágica Ebola/inmunología , Inmunidad Innata , Leucocitos Mononucleares/inmunología , Animales , Ebolavirus/patogenicidad , Perfilación de la Expresión Génica/métodos , Regulación de la Expresión Génica , Redes Reguladoras de Genes , Fiebre Hemorrágica Ebola/genética , Fiebre Hemorrágica Ebola/virología , Leucocitos Mononucleares/metabolismo , Macaca/virología , Ratones , Análisis de Secuencia de ARN/métodos , Replicación Viral
20.
Cell ; 162(4): 738-50, 2015 Aug 13.
Artículo en Inglés | MEDLINE | ID: mdl-26276630

RESUMEN

The 2013-2015 West African epidemic of Ebola virus disease (EVD) reminds us of how little is known about biosafety level 4 viruses. Like Ebola virus, Lassa virus (LASV) can cause hemorrhagic fever with high case fatality rates. We generated a genomic catalog of almost 200 LASV sequences from clinical and rodent reservoir samples. We show that whereas the 2013-2015 EVD epidemic is fueled by human-to-human transmissions, LASV infections mainly result from reservoir-to-human infections. We elucidated the spread of LASV across West Africa and show that this migration was accompanied by changes in LASV genome abundance, fatality rates, codon adaptation, and translational efficiency. By investigating intrahost evolution, we found that mutations accumulate in epitopes of viral surface proteins, suggesting selection for immune escape. This catalog will serve as a foundation for the development of vaccines and diagnostics. VIDEO ABSTRACT.


Asunto(s)
Genoma Viral , Fiebre de Lassa/virología , Virus Lassa/genética , ARN Viral/genética , África Occidental/epidemiología , Animales , Evolución Biológica , Reservorios de Enfermedades , Ebolavirus/genética , Variación Genética , Glicoproteínas/genética , Fiebre Hemorrágica Ebola/virología , Humanos , Fiebre de Lassa/epidemiología , Fiebre de Lassa/transmisión , Virus Lassa/clasificación , Virus Lassa/fisiología , Murinae/genética , Mutación , Nigeria/epidemiología , Proteínas Virales/genética , Zoonosis/epidemiología , Zoonosis/virología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...