Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 29
Filtrar
1.
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
2.
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
3.
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
4.
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
5.
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
6.
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
7.
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
8.
J Virol ; 86(19): 10759-65, 2012 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-22837210

RESUMO

Arenaviruses are rodent-borne viruses with a bisegmented RNA genome. A genetically unique arenavirus, Lujo virus, was recently discovered as the causal agent of a nosocomial outbreak of acute febrile illness with hemorrhagic manifestations in Zambia and South Africa. The outbreak had a case fatality rate of 80%. A reverse genetics system to rescue infectious Lujo virus from cDNA was established to investigate the biological properties of this virus. Sequencing the genomic termini showed unique nucleotides at the 3' terminus of the S segment promoter element. While developing this system, we discovered that reconstructing infectious Lujo virus using the previously reported L segment intergenic region (IGR), comprising the arenaviral transcription termination signal, yielded an attenuated Lujo virus. Resequencing revealed that the correct L segment IGR was 36 nucleotides longer, and incorporating it into the reconstructed Lujo virus restored the growth rate to that of the authentic clinical virus isolate. These additional nucleotides were predicted to more than double the free energy of the IGR main stem-loop structure. In addition, incorporating the newly determined L-IGR into a replicon reporter system enhanced the expression of a luciferase reporter L segment. Overall, these results imply that an extremely stable secondary structure within the L-IGR is critical for Lujo virus propagation and viral protein production. The technology for producing recombinant Lujo virus now provides a method to precisely investigate the molecular determinants of virulence of this newly identified pathogen.


Assuntos
Arenavirus/genética , RNA Viral/genética , Regiões 3' não Traduzidas , Animais , Arenavirus/fisiologia , Sequência de Bases , Cricetinae , DNA Complementar/metabolismo , Genes Reporter , Modelos Genéticos , Dados de Sequência Molecular , Conformação de Ácido Nucleico , Nucleotídeos/genética , Plasmídeos/metabolismo , Vírus de RNA/genética , África do Sul , Fatores de Tempo , Virulência , Zâmbia
9.
J Virol ; 85(1): 112-22, 2011 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-20980515

RESUMO

The Arenaviridae are a diverse and globally distributed collection of viruses that are maintained primarily by rodent reservoirs. Junin virus (JUNV) and Lassa virus (LASV) can both cause significant outbreaks of severe and often fatal human disease throughout their respective areas of endemicity. In an effort to improve upon the existing live attenuated JUNV Candid1 vaccine, we generated a genetically homogenous stock of this virus from cDNA copies of the virus S and L segments by using a reverse genetics system. Further, these cDNAs were used in combination with LASV cDNAs to successfully generate two recombinant Candid1 JUNV/LASV chimeric viruses (via envelope glycoprotein [GPC] exchange). It was found that while the GPC extravirion domains were readily exchangeable, homologous stable signal peptide (SSP) and G2 transmembrane and cytoplasmic tail domains were essential for correct GPC maturation and production of infectious chimeric viruses. The switching of the JUNV and LASV G1/G2 ectodomains within the Candid1 vaccine background did not alter the attenuated phenotype of the vaccine strain in a lethal mouse model. These recombinant chimeric viruses shed light on the fundamental requirements of arenavirus GPC maturation and may serve as a strategy for the development of bivalent JUNV and LASV vaccine candidates.


Assuntos
Glicoproteínas/genética , Vírus Junin/genética , Vírus Lassa/genética , Recombinação Genética , Proteínas do Envelope Viral/genética , Vacinas Virais , Animais , Infecções por Arenaviridae/mortalidade , Infecções por Arenaviridae/prevenção & controle , Infecções por Arenaviridae/virologia , Chlorocebus aethiops , Glicoproteínas/química , Glicoproteínas/metabolismo , Humanos , Vírus Junin/metabolismo , Vírus Junin/patogenicidade , Vírus Lassa/metabolismo , Vírus Lassa/patogenicidade , Camundongos , Sinais Direcionadores de Proteínas/genética , Sinais Direcionadores de Proteínas/fisiologia , Estrutura Terciária de Proteína/genética , Estrutura Terciária de Proteína/fisiologia , Células Vero , Proteínas do Envelope Viral/química , Proteínas do Envelope Viral/metabolismo , Vacinas Virais/administração & dosagem , Vacinas Virais/genética
10.
J Virol ; 85(8): 4020-4, 2011 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-21307206

RESUMO

Lassa virus (LASV), is a significant cause of severe, often fatal, hemorrhagic fever in humans throughout western Africa, with an estimated 100,000 infections each year. No vaccines are commercially available. We report the development of an efficient reverse genetics system to rescue recombinant LASV and to investigate the contributions of the long 5' and 3' noncoding regions (NCRs) of the S genomic segment to in vitro growth and in vivo virulence. This work demonstrates that deletions of large portions of these NCRs confer an attenuated phenotype and are a first step toward further insights into the high virulence of LASV.


Assuntos
Vírus Lassa/genética , Vírus Lassa/patogenicidade , RNA Viral/genética , Replicação Viral , Regiões 3' não Traduzidas , Regiões 5' não Traduzidas , Animais , Linhagem Celular , Dados de Sequência Molecular , Análise de Sequência de DNA , Deleção de Sequência , Virulência
11.
J Virol ; 85(19): 10404-8, 2011 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-21795336

RESUMO

Candid1, a live-attenuated Junin virus vaccine strain, was developed during the early 1980s to control Argentine hemorrhagic fever, a severe and frequently fatal human disease. Six amino acid substitutions were found to be unique to this vaccine strain, and their role in virulence attenuation in mice was analyzed using a series of recombinant viruses. Our results indicate that Candid1 is attenuated in mice through a single amino acid substitution in the transmembrane domain of the G2 glycoprotein. This work provides insight into the molecular mechanisms of attenuation of the only arenavirus vaccine currently available.


Assuntos
Vírus Junin/imunologia , Vírus Junin/patogenicidade , Mutação de Sentido Incorreto , Proteínas do Envelope Viral/genética , Vacinas Virais/genética , Fatores de Virulência/genética , Substituição de Aminoácidos/genética , Animais , Infecções por Arenaviridae/patologia , Infecções por Arenaviridae/virologia , Análise Mutacional de DNA , Modelos Animais de Doenças , Camundongos , Dados de Sequência Molecular , Mutação Puntual , Doenças dos Roedores/patologia , Doenças dos Roedores/virologia , Análise de Sequência de DNA , Vacinas Atenuadas/genética , Vacinas Atenuadas/imunologia , Proteínas do Envelope Viral/metabolismo , Vacinas Virais/imunologia , Fatores de Virulência/metabolismo
12.
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
13.
Proc Natl Acad Sci U S A ; 105(8): 2981-6, 2008 Feb 26.
Artigo em Inglês | MEDLINE | ID: mdl-18299564

RESUMO

Cytotoxic T lymphocytes (CTL) play an important role in the control and elimination of infection by West Nile virus (WNV), yet the class I human leukocyte antigen (HLA)-presented peptide epitopes that enable CTL recognition of WNV-infected cells remain uncharacterized. The goals of this work were first to discover the peptide epitopes that distinguish the class I HLA of WNV-infected cells and then to test the T cell reactivity of newly discovered WNV epitopes. To discover WNV-immune epitopes, class I HLA was harvested from WNV (NY99 strain)-infected and uninfected HeLa cells. Then peptide epitopes were eluted from affinity-purified HLA, and peptide epitopes from infected and uninfected cells were comparatively mapped by mass spectroscopy. Six virus-derived peptides from five different viral proteins (E, NS2b, NS3, NS4b, and NS5) were discovered as unique to HLA-A*0201 of infected cells, demonstrating that the peptides sampled by class I HLA are distributed widely throughout the WNV proteome. When tested with CTL from infected individuals, one dominant WNV target was apparent, two epitopes were subdominant, and three demonstrated little CTL reactivity. Finally, a sequence comparison of these epitopes with the hundreds of viral isolates shows that HLA-A*0201 presents epitopes derived from conserved regions of the virus. Detection and recovery from WNV infection are therefore functions of the ability of class I HLA molecules to reveal conserved WNV epitopes to an intact cellular immune system that subsequently recognizes infected cells.


Assuntos
Epitopos de Linfócito T/genética , Antígenos HLA/metabolismo , Febre do Nilo Ocidental/imunologia , Vírus do Nilo Ocidental/imunologia , Animais , Sequência de Bases , Chlorocebus aethiops , Primers do DNA/genética , Epitopos de Linfócito T/imunologia , Epitopos de Linfócito T/metabolismo , Antígenos HLA/imunologia , Humanos , Espectrometria de Massas , Dados de Sequência Molecular , Análise de Sequência de DNA , Homologia de Sequência , Células Vero , Vírus do Nilo Ocidental/genética
14.
AIDS Res Ther ; 6: 20, 2009 Oct 02.
Artigo em Inglês | MEDLINE | ID: mdl-19799780

RESUMO

HIV-1 infects gut associated lymphoid tissues (GALT) very early after transmission by multiple routes. The infected GALT consequently serves as the major reservoir for HIV-1 infection and could constantly shed HIV-1 and CD4+ T cells into the intestinal lumen. To examine this hypothesis, we monitored HIV-1 RNA/DNA and CD4 mRNA in fecal samples of chronically infected subjects with and without antiretroviral therapy (ART). We compared this to levels of HIV-1 RNA/DNA in urine and blood from the same subjects. Our results show that HIV-1 DNA, RNA and CD4 mRNA were detected in 8%, 19% and 31% respectively, of feces samples from infected subjects with detectable plasma viral load, and were not detected in any of subjects on ART with undetectable plasma viral load. In urine samples, HIV-1 DNA was detected in 24% of infected subjects with detectable plasma viral load and 23% of subjects on ART with undetectable plasma viral load. Phylogenetic analysis of the envelope sequences of HIV-1 revealed distinct virus populations in concurrently collected serum, feces and urine samples from one subject. In addition, our study demonstrated for the first time the presence of CD4 mRNA in fecal specimens of HIV-1 infected subjects, which could be used to assess GALT pathogenesis in HIV-1 infection.

15.
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
16.
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.

17.
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
18.
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
19.
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
20.
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
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA