Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 29
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Vaccine ; 40(33): 4845-4855, 2022 08 05.
Artigo em Inglês | MEDLINE | ID: mdl-35803846

RESUMO

BACKGROUND: COVID-19 vaccination reduces SARS-CoV-2 infection and transmission. However, evidence is emerging on the degree of protection across variants and in high-transmission settings. To better understand the protection afforded by vaccination specifically in a high-transmission setting, we examined household transmission of SARS-CoV-2 during a period of high community incidence with predominant SARS-CoV-2 B.1.1.7 (Alpha) variant, among vaccinated and unvaccinated contacts. METHODS: We conducted a household transmission investigation in San Diego County, California, and Denver, Colorado, during January-April 2021. Households were enrolled if they had at least one person with documented SARS-CoV-2 infection. We collected nasopharyngeal swabs, blood, demographic information, and vaccination history from all consenting household members. We compared infection risks (IRs), RT-PCR cycle threshold values, SARS-CoV-2 culture results, and antibody statuses among vaccinated and unvaccinated household contacts. RESULTS: We enrolled 493 individuals from 138 households. The SARS-CoV-2 variant was identified from 121/138 households (88%). The most common variants were Alpha (75/121, 62%) and Epsilon (19/121, 16%). There were no households with discordant lineages among household members. One fully vaccinated secondary case was symptomatic (13%); the other 5 were asymptomatic (87%). Among unvaccinated secondary cases, 105/108 (97%) were symptomatic. Among 127 households with a single primary case, the IR for household contacts was 45% (146/322; 95% Confidence Interval [CI] 40-51%). The observed IR was higher in unvaccinated (130/257, 49%, 95% CI 45-57%) than fully vaccinated contacts (6/26, 23%, 95% CI 11-42%). A lower proportion of households with a fully vaccinated primary case had secondary cases (1/5, 20%) than households with an unvaccinated primary case (66/108, 62%). CONCLUSIONS: Although SARS-CoV-2 infections in vaccinated household contacts were reported in this high transmission setting, full vaccination protected against SARS-CoV-2 infection. These findings further support the protective effect of COVID-19 vaccination and highlight the need for ongoing vaccination among eligible persons.


Assuntos
COVID-19 , SARS-CoV-2 , COVID-19/epidemiologia , COVID-19/prevenção & controle , Vacinas contra COVID-19 , California/epidemiologia , Colorado/epidemiologia , Humanos
2.
J Infect Dis ; 219(11): 1716-1721, 2019 05 05.
Artigo em Inglês | MEDLINE | ID: mdl-30590775

RESUMO

Although bats are increasingly being recognized as natural reservoir hosts of emerging zoonotic viruses, little is known about how they control and clear virus infection in the absence of clinical disease. Here, we test >50 convalescent sera from Egyptian rousette bats (ERBs) experimentally primed or prime-boosted with Marburg virus, Ebola virus, or Sosuga virus for the presence of virus-specific neutralizing antibodies, using infectious reporter viruses. After serum neutralization testing, we conclude that antibody-mediated virus neutralization does not contribute significantly to the control and clearance of Marburg virus, Ebola virus, or Sosuga virus infection in ERBs.


Assuntos
Quirópteros/virologia , Ebolavirus/imunologia , Doença pelo Vírus Ebola/imunologia , Doença do Vírus de Marburg/imunologia , Marburgvirus/imunologia , Paramyxoviridae/imunologia , Animais , Anticorpos Antivirais/imunologia , Convalescença , Reservatórios de Doenças/virologia , Egito/epidemiologia , Doença pelo Vírus Ebola/virologia , Humanos , Imunidade Humoral , Doença do Vírus de Marburg/virologia , Testes de Neutralização
3.
Artigo em Inglês | MEDLINE | ID: mdl-30533642

RESUMO

Monongahela hantavirus was first identified in deer mice and was later found responsible for hantavirus pulmonary syndrome cases in Pennsylvania and West Virginia in the United States. Here, we report the complete sequences of Monongahela virus S, M, and L genomic segments obtained from a fatal clinical case reported in 1997.

4.
PLoS One ; 13(8): e0201827, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30071116

RESUMO

Filoviruses are notorious viral pathogens responsible for high-consequence diseases in humans and non-human primates. Transcription of filovirus mRNA shares several common features with transcription in other non-segmented negative-strand viruses, including differential expression of genes located across the viral genome. Transcriptional patterns of Ebola virus (EBOV) and Marburg virus (MARV) have been previously described using traditional, laborious methods, such as northern blots and in vivo labeling of viral mRNAs. More recently, however, the availability of the next generation sequencing (NGS) technology has offered a more straightforward approach to assess transcriptional patterns. In this report, we analyzed the transcription patterns of four ebolaviruses-EBOV, Sudan (SUDV), Bundibugyo (BDBV), and Reston (RESTV) viruses-in two different cell lines using standard NGS library preparation and sequencing protocols. In agreement with previous reports mainly focused on EBOV and MARV, the remaining filoviruses used in this study also showed a consistent transcription pattern, with only minor variations between the different viruses. We have also analyzed the proportions of the three mRNAs transcribed from the GP gene, which are characteristic of the genus Ebolavirus and encode the glycoprotein (GP), the soluble GP (sGP), and the small soluble GP (ssGP). In addition, we used NGS methodology to analyze the transcription pattern of two previously described recombinant MARV. This analysis allowed us to correct our construction design, and to make an improved version of the original MARV expressing reporter genes.


Assuntos
Infecções por Filoviridae/metabolismo , Filoviridae/metabolismo , RNA Mensageiro/metabolismo , RNA Viral/metabolismo , Transcrição Gênica , Animais , Técnicas de Cultura de Células , Células Cultivadas , Chlorocebus aethiops , Cricetinae , Humanos , Fígado/metabolismo , Fígado/virologia , Macrófagos/metabolismo , Macrófagos/virologia , Temperatura
5.
Artigo em Inglês | MEDLINE | ID: mdl-30050872

RESUMO

Genome reassortment in Lassa virus (LASV) has been reported in nature, but phenotypic consequences of this phenomenon are not well described. Here we characterize, both in vitro and in vivo, reassortment between 2 LASV strains: the prototypic 1976 Josiah strain and a more recently isolated 2015 Liberian strain. In vitro analysis showed that although cis- and trans-acting elements of viral RNA synthesis were compatible between strains, reassortants demonstrated different levels of viral replication. These differences were also apparent in vivo, as reassortants varied in pathogenicity in the guinea pig model of LASV infection. The reassortant variant containing the Josiah S segment retained the virulence of the parental Josiah strain, but the reassortant variant containing the S segment of the Liberian isolate was highly attenuated compared to both parental strains. Contrary to observations in reassortants between LASV and other arenavirus species, which suggest that L segment-encoded factors are responsible for virulence, these studies highlight a role for S segment-encoded virulence factors in disease, and also suggest that inefficient interactions between proteins of heterologous strains may limit the prevalence of reassortant LASV variants in nature.


Assuntos
Febre Lassa/patologia , Febre Lassa/virologia , Vírus Lassa/patogenicidade , Vírus Reordenados/patogenicidade , Fatores de Virulência/genética , Animais , Modelos Animais de Doenças , Cobaias , Vírus Lassa/genética , Vírus Reordenados/genética , Vírus Reordenados/fisiologia , Virulência , Replicação Viral
6.
PLoS Negl Trop Dis ; 12(3): e0006326, 2018 03.
Artigo em Inglês | MEDLINE | ID: mdl-29522528

RESUMO

Sosuga virus (SOSV) is a recently discovered zoonotic paramyxovirus isolated from a single human case in 2012; it has been ecologically and epidemiologically associated with transmission by the Egyptian rousette bat (Rousettus aegyptiacus). Bats have long been recognized as sources of novel zoonotic pathogens, including highly lethal paramyxoviruses like Nipah virus (NiV) and Hendra virus (HeV). The ability of SOSV to cause severe human disease supports the need for studies on SOSV pathogenesis to better understand the potential impact of this virus and to identify effective treatments. Here we describe a reverse genetics system for SOSV comprising a minigenome-based assay and a replication-competent infectious recombinant reporter SOSV that expresses the fluorescent protein ZsGreen1 in infected cells. First, we used the minigenome assay to rapidly screen for compounds inhibiting SOSV replication at biosafety level 2 (BSL-2). The antiviral activity of candidate compounds was then tested against authentic viral replication using the reporter SOSV at BSL-3. We identified several compounds with anti-SOSV activity, several of which also inhibit NiV and HeV. Alongside its utility in screening for potential SOSV therapeutics, the reverse genetics system described here is a powerful tool for analyzing mechanisms of SOSV pathogenesis, which will facilitate our understanding of how to combat the potential public health threats posed by emerging bat-borne paramyxoviruses.


Assuntos
Antivirais/farmacologia , Paramyxoviridae/genética , Genética Reversa/métodos , Replicação Viral/efeitos dos fármacos , Animais , Linhagem Celular , Quirópteros/virologia , Humanos , Paramyxoviridae/fisiologia , Infecções por Paramyxoviridae/virologia
7.
PLoS One ; 12(5): e0178224, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28542463

RESUMO

Reston virus (family Filoviridae) is unique among the viruses of the Ebolavirus genus in that it is considered non-pathogenic in humans, in contrast to the other members which are highly virulent. The virus has however, been associated with several outbreaks of highly lethal hemorrhagic fever in non-human primates (NHPs), specifically cynomolgus monkeys (Macaca fascicularis) originating in the Philippines. In addition, Reston virus has been isolated from domestic pigs in the Philippines. To better understand virus spillover events and potential adaption to new hosts, the whole genome sequences of representative Reston virus isolates were obtained using a next generation sequencing (NGS) approach and comparative genomic analysis and virus fitness analyses were performed. Nine virus genome sequences were completed for novel and previously described isolates obtained from a variety of hosts including a human case, non-human primates and pigs. Results of phylogenetic analysis of the sequence differences are consistent with multiple independent introductions of RESTV from a still unknown natural reservoir into non-human primates and swine farming operations. No consistent virus genetic markers were found specific for viruses associated with primate or pig infections, but similar to what had been seen with some Ebola viruses detected in the large Western Africa outbreak in 2014-2016, a truncated version of VP30 was identified in a subgroup of Reston viruses obtained from an outbreak in pigs 2008-2009. Finally, the genetic comparison of two closely related viruses, one isolated from a human case and one from an NHP, showed amino acid differences in the viral polymerase and detectable differences were found in competitive growth assays on human and NHP cell lines.


Assuntos
Filoviridae/genética , Genoma Viral/genética , Animais , Surtos de Doenças/veterinária , Ebolavirus/genética , Ebolavirus/patogenicidade , Filoviridae/patogenicidade , Infecções por Filoviridae/veterinária , Infecções por Filoviridae/virologia , Marcadores Genéticos/genética , Doença pelo Vírus Ebola/veterinária , Doença pelo Vírus Ebola/virologia , Sequenciamento de Nucleotídeos em Larga Escala , Humanos , Macaca fascicularis/virologia , Suínos/virologia
8.
Virology ; 501: 147-165, 2017 01 15.
Artigo em Inglês | MEDLINE | ID: mdl-27930961

RESUMO

Filoviruses are highly lethal in humans and nonhuman primates, likely due to potent antagonism of host interferon (IFN) responses early in infection. Filoviral protein VP35 is implicated as the major IFN induction antagonist, while Ebola virus (EBOV) VP24 or Marburg virus (MARV) VP40 are known to block downstream IFN signaling. Despite progress elucidating EBOV and MARV antagonist function, those for most other filoviruses, including Reston (RESTV), Sudan (SUDV), Taï Forest (TAFV), Bundibugyo (BDBV) and Ravn (RAVV) viruses, remain largely neglected. Thus, using standardized vectors and reporter assays, we characterized activities by each IFN antagonist from all known ebolavirus and marburgvirus species side-by-side. We uncover noncanonical suppression of IFN induction by ebolavirus VP24, differing potencies by MARV and RAVV proteins, and intriguingly, weaker antagonism by VP24 of RESTV. These underlying molecular explanations for differential virulence in humans could guide future investigations of more-neglected filoviruses as well as treatment and vaccine studies.


Assuntos
Ebolavirus/metabolismo , Doença pelo Vírus Ebola/virologia , Interferons/antagonistas & inibidores , Doença do Vírus de Marburg/virologia , Marburgvirus/metabolismo , Proteínas Virais/metabolismo , Animais , Ebolavirus/genética , Genes Reporter , Interações Hospedeiro-Patógeno , Humanos , Interferons/metabolismo , Marburgvirus/genética , Proteínas Virais/genética
9.
mBio ; 7(6)2016 12 20.
Artigo em Inglês | MEDLINE | ID: mdl-27999160

RESUMO

Lassa virus (LASV) infection is a major public health concern due to high fatality rates and limited effective treatment. The interferon-stimulated gene cholesterol 25-hydroxylase (CH25H) encodes an enzyme that catalyzes the production of 25-hydroxycholesterol (25HC). 25HC is involved in regulating cholesterol biosynthesis and has recently been identified as a potent antiviral targeting enveloped virus entry. Here, we show a previously unrecognized role of CH25H in inhibiting LASV glycoprotein glycosylation and the production of infectious virus. Overexpression of CH25H or treatment with 25HC decreased LASV G1 glycoprotein N-glycan maturation and reduced the production of infectious LASV. Depletion of endogenous CH25H using small interfering RNA (siRNA) enhanced the levels of fully glycosylated G1 and increased infectious LASV production. Finally, LASV particles produced from 25HC-treated cells were found to be less infectious, to incorporate aberrantly glycosylated GP1 species, and to be defective in binding alpha-dystroglycan, an attachment and entry receptor. Our findings identify a novel role for CH25H in controlling LASV propagation and indicate that manipulation of the expression of CH25H or the administration of 25HC may be a useful anti-LASV therapy. IMPORTANCE: Lassa fever is an acute viral hemorrhagic fever in humans caused by Lassa virus (LASV). No vaccine for LASV is currently available. Treatment is limited to the administration of ribavirin, which is only effective when given early in the course of illness. Cholesterol 25-hydroxylase (CH25H) is a recently identified interferon-stimulated gene (ISG); it encodes an enzyme that catalyzes the production of 25-hydroxycholesterol (25HC), which inhibits several viruses. Here, we identify a novel antiviral mechanism of 25HC that is dependent on inhibiting the glycosylation of Lassa virus (LASV) glycoprotein and reducing the infectivity of LASV as a means of suppressing viral replication. Since N-linked glycosylation is a critical feature of other enveloped-virus glycoproteins, 25HC may be a broad inhibitor of virus infectivity.


Assuntos
Antivirais/farmacologia , Hidroxicolesteróis/farmacologia , Vírus Lassa/efeitos dos fármacos , Vírus Lassa/metabolismo , Animais , Linhagem Celular , Glicosilação/efeitos dos fármacos , Humanos , Vírus Lassa/química , RNA Interferente Pequeno , Esteroide Hidroxilases/genética , Esteroide Hidroxilases/metabolismo , Proteínas do Envelope Viral , Internalização do Vírus/efeitos dos fármacos , Replicação Viral/efeitos dos fármacos
10.
Antiviral Res ; 136: 9-18, 2016 12.
Artigo em Inglês | MEDLINE | ID: mdl-27771389

RESUMO

Lassa virus (LASV) and Ebola virus (EBOV) infections are important global health issues resulting in significant morbidity and mortality. While several promising drug and vaccine trials for EBOV are ongoing, options for LASV infection are currently limited to ribavirin treatment. A major factor impeding the development of antiviral compounds to treat these infections is the need to manipulate the virus under BSL-4 containment, limiting research to a few institutes worldwide. Here we describe the development of a novel LASV minigenome assay based on the ambisense LASV S segment genome, with authentic terminal untranslated regions flanking a ZsGreen (ZsG) fluorescent reporter protein and a Gaussia princeps luciferase (gLuc) reporter gene. This assay, along with a similar previously established EBOV minigenome, was optimized for high-throughput screening (HTS) of potential antiviral compounds under BSL-2 containment. In addition, we rescued a recombinant LASV expressing ZsG, which, in conjunction with a recombinant EBOV reporter virus, was used to confirm any potential antiviral hits in vitro. Combining an initial screen to identify potential antiviral compounds at BSL-2 containment before progressing to HTS with infectious virus will reduce the amount of expensive and technically challenging BSL-4 containment research. Using these assays, we identified 6-azauridine as having anti-LASV activity, and demonstrated its anti-EBOV activity in human cells. We further identified 2'-deoxy-2'-fluorocytidine as having potent anti-LASV activity, with an EC50 value 10 times lower than that of ribavirin.


Assuntos
Antivirais/farmacologia , Ebolavirus/efeitos dos fármacos , Ebolavirus/genética , Vírus Lassa/efeitos dos fármacos , Vírus Lassa/genética , Antivirais/química , Azauridina/farmacologia , Desoxicitidina/análogos & derivados , Desoxicitidina/farmacologia , Descoberta de Drogas/métodos , Genes Reporter , Genoma Viral , Proteínas de Fluorescência Verde/genética , Doença pelo Vírus Ebola , Ensaios de Triagem em Larga Escala/métodos , Humanos , Febre Lassa , Luciferases/genética
11.
J Infect Dis ; 214(suppl 3): S258-S262, 2016 10 15.
Artigo em Inglês | MEDLINE | ID: mdl-27587631

RESUMO

During the Ebola virus outbreak of 2013-2016, the Viral Special Pathogens Branch field laboratory in Sierra Leone tested approximately 26 000 specimens between August 2014 and October 2015. Analysis of the B2M endogenous control Ct values showed its utility in monitoring specimen quality, comparing results with different specimen types, and interpretation of results. For live patients, blood is the most sensitive specimen type and oral swabs have little diagnostic utility. However, swabs are highly sensitive for diagnostic testing of corpses.


Assuntos
Surtos de Doenças , Ebolavirus/isolamento & purificação , Doença pelo Vírus Ebola/diagnóstico , RNA Viral/análise , Reação em Cadeia da Polimerase Via Transcriptase Reversa/métodos , Serviços de Laboratório Clínico , Ebolavirus/genética , Doença pelo Vírus Ebola/epidemiologia , Doença pelo Vírus Ebola/virologia , Humanos , Laboratórios , Reação em Cadeia da Polimerase em Tempo Real/métodos , Sensibilidade e Especificidade , Serra Leoa/epidemiologia
12.
Virology ; 496: 237-243, 2016 09.
Artigo em Inglês | MEDLINE | ID: mdl-27366976

RESUMO

During the large outbreak of Ebola virus disease that occurred in Western Africa from late 2013 to early 2016, several hundred Ebola virus (EBOV) genomes have been sequenced and the virus genetic drift analyzed. In a previous report, we described an efficient reverse genetics system designed to generate recombinant EBOV based on a Makona variant isolate obtained in 2014. Using this system, we characterized the replication and fitness of 2 isolates of the Makona variant. These virus isolates are nearly identical at the genetic level, but have single amino acid differences in the VP30 and L proteins. The potential effects of these differences were tested using minigenomes and recombinant viruses. The results obtained with this approach are consistent with the role of VP30 and L as components of the EBOV RNA replication machinery. Moreover, the 2 isolates exhibited clear fitness differences in competitive growth assays.


Assuntos
Ebolavirus/fisiologia , Aptidão Genética , Genoma Viral , Doença pelo Vírus Ebola/virologia , Genética Reversa , Evolução Molecular , Ordem dos Genes , Recombinação Genética
13.
J Infect Dis ; 213(5): 703-11, 2016 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-26582961

RESUMO

Animal models recapitulating human Ebola virus disease (EVD) are critical for insights into virus pathogenesis. Ebola virus (EBOV) isolates derived directly from human specimens do not, without adaptation, cause disease in immunocompetent adult rodents. Here, we describe EVD in mice engrafted with human immune cells (hu-BLT). hu-BLT mice developed EVD following wild-type EBOV infection. Infection with high-dose EBOV resulted in rapid, lethal EVD with high viral loads, alterations in key human antiviral immune cytokines and chemokines, and severe histopathologic findings similar to those shown in the limited human postmortem data available. A dose- and donor-dependent clinical course was observed in hu-BLT mice infected with lower doses of either Mayinga (1976) or Makona (2014) isolates derived from human EBOV cases. Engraftment of the human cellular immune system appeared to be essential for the observed virulence, as nonengrafted mice did not support productive EBOV replication or develop lethal disease. hu-BLT mice offer a unique model for investigating the human immune response in EVD and an alternative animal model for EVD pathogenesis studies and therapeutic screening.


Assuntos
Ebolavirus/fisiologia , Regulação da Expressão Gênica/imunologia , Doença pelo Vírus Ebola/imunologia , Animais , Encéfalo/virologia , Citocinas/genética , Citocinas/metabolismo , Doença pelo Vírus Ebola/urina , Doença pelo Vírus Ebola/virologia , Humanos , Rim/virologia , Fígado/virologia , Pulmão/virologia , Masculino , Camundongos , Camundongos Transgênicos , RNA Viral/isolamento & purificação , Baço/virologia , Testículo/virologia , Replicação Viral
14.
Emerg Infect Dis ; 21(10): 1816-9, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-26401603

RESUMO

To determine the utility of oral swabs for diagnosing infection with Ebola virus, we used a guinea pig model and obtained daily antemortem and postmortem swab samples. According to quantitative reverse transcription PCR analysis, the diagnostic value was poor for antemortem swab samples but excellent for postmortem samples.


Assuntos
Diagnóstico Precoce , Ebolavirus , Doença pelo Vírus Ebola/diagnóstico , Boca/virologia , Manejo de Espécimes/métodos , Animais , Cobaias , Doença pelo Vírus Ebola/virologia , Humanos
15.
J Infect Dis ; 212 Suppl 2: S350-8, 2015 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-26232439

RESUMO

In August 2014, the Viral Special Pathogens Branch of the US Centers for Disease Control and Prevention established a field laboratory in Sierra Leone in response to the ongoing Ebola virus outbreak. Through March 2015, this laboratory tested >12 000 specimens from throughout Sierra Leone. We describe the organization and procedures of the laboratory located in Bo, Sierra Leone.


Assuntos
Ebolavirus/isolamento & purificação , Doença pelo Vírus Ebola/diagnóstico , Doença pelo Vírus Ebola/virologia , Centers for Disease Control and Prevention, U.S. , Surtos de Doenças , Epidemias , Humanos , Laboratórios , Serra Leoa/epidemiologia , Estados Unidos
16.
J Virol ; 89(20): 10219-29, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-26223644

RESUMO

UNLABELLED: In the cytoplasm, the retinoic acid-inducible gene I (RIG-I) senses the RNA genomes of several RNA viruses. RIG-I binds to viral RNA, eliciting an antiviral response via the cellular adaptor MAVS. Crimean-Congo hemorrhagic fever virus (CCHFV), a negative-sense RNA virus with a 5'-monophosphorylated genome, is a highly pathogenic zoonotic agent with significant public health implications. We found that, during CCHFV infection, RIG-I mediated a type I interferon (IFN) response via MAVS. Interfering with RIG-I signaling reduced IFN production and IFN-stimulated gene expression and increased viral replication. Immunostimulatory RNA was isolated from CCHFV-infected cells and from virion preparations, and RIG-I coimmunoprecipitation of infected cell lysates isolated immunostimulatory CCHFV RNA. This report serves as the first description of a pattern recognition receptor for CCHFV and highlights a critical signaling pathway in the antiviral response to CCHFV. IMPORTANCE: CCHFV is a tick-borne virus with a significant public health impact. In order for cells to respond to virus infection, they must recognize the virus as foreign and initiate antiviral signaling. To date, the receptors involved in immune recognition of CCHFV are not known. Here, we investigate and identify RIG-I as a receptor involved in initiating an antiviral response to CCHFV. This receptor initially was not expected to play a role in CCHFV recognition because of characteristics of the viral genome. These findings are important in understanding the antiviral response to CCHFV and support continued investigation into the spectrum of potential viruses recognized by RIG-I.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/imunologia , RNA Helicases DEAD-box/imunologia , Genoma Viral , Vírus da Febre Hemorrágica da Crimeia-Congo/imunologia , Interferon Tipo I/imunologia , RNA Viral/imunologia , Proteínas Adaptadoras de Transdução de Sinal/genética , Animais , Linhagem Celular , Linhagem Celular Tumoral , Chlorocebus aethiops , Proteína DEAD-box 58 , RNA Helicases DEAD-box/genética , Células Epiteliais , Fibroblastos/imunologia , Fibroblastos/virologia , Regulação da Expressão Gênica , Células HEK293 , Vírus da Febre Hemorrágica da Crimeia-Congo/genética , Interações Hospedeiro-Patógeno , Humanos , Interferon Tipo I/genética , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/metabolismo , RNA Viral/genética , Receptores Imunológicos , Receptores Virais/genética , Receptores Virais/imunologia , Transdução de Sinais , Células Vero , Replicação Viral
17.
PLoS Pathog ; 11(5): e1004879, 2015 May.
Artigo em Inglês | MEDLINE | ID: mdl-25933376

RESUMO

Crimean Congo hemorrhagic fever virus (CCHFV) is a negative-strand RNA virus of the family Bunyaviridae (genus: Nairovirus). In humans, CCHFV causes fever, hemorrhage, severe thrombocytopenia, and high fatality. A major impediment in precisely determining the basis of CCHFV's high pathogenicity has been the lack of methodology to produce recombinant CCHFV. We developed a reverse genetics system based on transfecting plasmids into BSR-T7/5 and Huh7 cells. In our system, bacteriophage T7 RNA polymerase produced complementary RNA copies of the viral S, M, and L segments that were encapsidated with the support, in trans, of CCHFV nucleoprotein and L polymerase. The system was optimized to systematically recover high yields of infectious CCHFV. Additionally, we tested the ability of the system to produce specifically designed CCHFV mutants. The M segment encodes a polyprotein that is processed by host proprotein convertases (PCs), including the site-1 protease (S1P) and furin-like PCs. S1P and furin cleavages are necessary for producing the non-structural glycoprotein GP38, while S1P cleavage yields structural Gn. We studied the role of furin cleavage by rescuing a recombinant CCHFV encoding a virus glycoprotein precursor lacking a functional furin cleavage motif (RSKR mutated to ASKA). The ASKA mutation blocked glycoprotein precursor's maturation to GP38, and Gn precursor's maturation to Gn was slightly diminished. Furin cleavage was not essential for replication, as blocking furin cleavage resulted only in transient reduction of CCHFV titers, suggesting that either GP38 and/or decreased Gn maturation accounted for the reduced virion production. Our data demonstrate that nairoviruses can be produced by reverse genetics, and the utility of our system uncovered a function for furin cleavage. This viral rescue system could be further used to study the CCHFV replication cycle and facilitate the development of efficacious vaccines to counter this biological and public health threat.


Assuntos
Furina/metabolismo , Glicoproteínas/metabolismo , Vírus da Febre Hemorrágica da Crimeia-Congo/metabolismo , RNA/metabolismo , Proteínas não Estruturais Virais/metabolismo , Motivos de Aminoácidos , Substituição de Aminoácidos , Animais , Linhagem Celular , Chlorocebus aethiops , Células Clonais , Cricetulus , Furina/genética , Glicoproteínas/química , Glicoproteínas/genética , Vírus da Febre Hemorrágica da Crimeia-Congo/genética , Hepatócitos/enzimologia , Hepatócitos/metabolismo , Hepatócitos/virologia , Humanos , Mesocricetus , Mutação , Fragmentos de Peptídeos/genética , Fragmentos de Peptídeos/metabolismo , Pró-Proteína Convertases/metabolismo , Proteólise , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo , Serina Endopeptidases/metabolismo , Especificidade por Substrato , Proteínas não Estruturais Virais/química , Proteínas não Estruturais Virais/genética
18.
Virology ; 476: 85-91, 2015 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-25531184

RESUMO

Previous in vitro studies have demonstrated that Ebola and Marburg virus (EBOV and MARV) VP35 antagonize the host cell immune response. Moreover, specific mutations in the IFN inhibitory domain (IID) of EBOV and MARV VP35 that abrogate their interaction with virus-derived dsRNA, lack the ability to inhibit the host immune response. To investigate the role of MARV VP35 in the context of infectious virus, we used our reverse genetics system to generate two recombinant MARVs carrying specific mutations in the IID region of VP35. Our data show that wild-type and mutant viruses grow to similar titers in interferon deficient cells, but exhibit attenuated growth in interferon-competent cells. Furthermore, in contrast to wild-type virus, both MARV mutants were unable to inhibit expression of various antiviral genes. The MARV VP35 mutants exhibit similar phenotypes to those previously described for EBOV, suggesting the existence of a shared immune-modulatory strategy between filoviruses.


Assuntos
Doença do Vírus de Marburg/imunologia , Marburgvirus/genética , Proteínas do Core Viral/química , Proteínas do Core Viral/genética , Animais , Linhagem Celular , Interações Hospedeiro-Patógeno , Interferon beta/genética , Interferon beta/imunologia , Doença do Vírus de Marburg/virologia , Marburgvirus/química , Marburgvirus/imunologia , Mutação , Estrutura Terciária de Proteína , Proteínas do Core Viral/imunologia
19.
Antiviral Res ; 106: 86-94, 2014 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-24713118

RESUMO

Marburg virus (MARV) and Ebola virus (EBOV), members of the family Filoviridae, represent a significant challenge to global public health. Currently, no licensed therapies exist to treat filovirus infections, which cause up to 90% mortality in human cases. To facilitate development of antivirals against these viruses, we established two distinct screening platforms based on MARV and EBOV reverse genetics systems that express secreted Gaussia luciferase (gLuc). The first platform is a mini-genome replicon to screen viral replication inhibitors using gLuc quantification in a BSL-2 setting. The second platform is complementary to the first and expresses gLuc as a reporter gene product encoded in recombinant infectious MARV and EBOV, thereby allowing for rapid quantification of viral growth during treatment with antiviral compounds. We characterized these viruses by comparing luciferase activity to virus production, and validated luciferase activity as an authentic real-time measure of viral growth. As proof of concept, we adapt both mini-genome and infectious virus platforms to high-throughput formats, and demonstrate efficacy of several antiviral compounds. We anticipate that both approaches will prove highly useful in the development of anti-filovirus therapies, as well as in basic research on the filovirus life cycle.


Assuntos
Antivirais/isolamento & purificação , Avaliação Pré-Clínica de Medicamentos/métodos , Ebolavirus/efeitos dos fármacos , Marburgvirus/efeitos dos fármacos , Genética Reversa/métodos , Animais , Antivirais/farmacologia , Linhagem Celular , Ebolavirus/genética , Ebolavirus/fisiologia , Genes Reporter , Luciferases/análise , Luciferases/genética , Marburgvirus/genética , Marburgvirus/fisiologia , Replicação Viral/efeitos dos fármacos
20.
Virology ; 446(1-2): 230-7, 2013 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-24074586

RESUMO

Recent investigations have shown the Egyptian fruit bat (Rousettus aegyptiacus) to be a natural reservoir for marburgviruses. To better understand the life cycle of these viruses in the natural host, a new reverse genetics system was developed for the reliable rescue of a Marburg virus (MARV) originally isolated directly from a R. aegyptiacus bat (371Bat). To develop this system, the exact terminal sequences were first determined by 5' and 3' RACE, followed by the cloning of viral proteins NP, VP35, VP30 and L into expression plasmids. Novel conditions were then developed to efficiently replicate virus mini-genomes followed by the construction of full-length genomic clones from which recombinant wild type and GFP-containing MARVs were rescued. Surprisingly, when these recombinant MARVs were propagated in primary human macrophages, a dramatic difference was found in their ability to grow and to elicit anti-viral cytokine responses.


Assuntos
Quirópteros/virologia , Marburgvirus/genética , Recombinação Genética , Genética Reversa/métodos , Virologia/métodos , Animais , Células Cultivadas , Clonagem Molecular , Vetores Genéticos , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Humanos , Macrófagos/virologia , Marburgvirus/isolamento & purificação , Plasmídeos , Coloração e Rotulagem/métodos , Proteínas Virais/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...