Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 31
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Nature ; 514(7520): 47-53, 2014 Oct 02.
Artigo em Inglês | MEDLINE | ID: mdl-25171469

RESUMO

Without an approved vaccine or treatments, Ebola outbreak management has been limited to palliative care and barrier methods to prevent transmission. These approaches, however, have yet to end the 2014 outbreak of Ebola after its prolonged presence in West Africa. Here we show that a combination of monoclonal antibodies (ZMapp), optimized from two previous antibody cocktails, is able to rescue 100% of rhesus macaques when treatment is initiated up to 5 days post-challenge. High fever, viraemia and abnormalities in blood count and blood chemistry were evident in many animals before ZMapp intervention. Advanced disease, as indicated by elevated liver enzymes, mucosal haemorrhages and generalized petechia could be reversed, leading to full recovery. ELISA and neutralizing antibody assays indicate that ZMapp is cross-reactive with the Guinean variant of Ebola. ZMapp exceeds the efficacy of any other therapeutics described so far, and results warrant further development of this cocktail for clinical use.


Assuntos
Anticorpos Monoclonais/uso terapêutico , Anticorpos Antivirais/uso terapêutico , Doença pelo Vírus Ebola/tratamento farmacológico , Imunização Passiva , Sequência de Aminoácidos , Animais , Anticorpos Monoclonais/imunologia , Anticorpos Neutralizantes/imunologia , Anticorpos Neutralizantes/uso terapêutico , Anticorpos Antivirais/imunologia , Reações Cruzadas/imunologia , Ebolavirus/imunologia , Ensaio de Imunoadsorção Enzimática , Feminino , Guiné , Cobaias , Doença pelo Vírus Ebola/sangue , Doença pelo Vírus Ebola/imunologia , Doença pelo Vírus Ebola/virologia , Macaca mulatta/imunologia , Macaca mulatta/virologia , Masculino , Dados de Sequência Molecular , Alinhamento de Sequência , Proteínas do Envelope Viral/química , Proteínas do Envelope Viral/imunologia , Viremia/tratamento farmacológico , Viremia/imunologia , Viremia/virologia
2.
J Infect Dis ; 218(suppl_5): S471-S474, 2018 11 22.
Artigo em Inglês | MEDLINE | ID: mdl-29889278

RESUMO

Ferrets are used for studying infections with wild-type Ebola virus isolates. Here, we investigated whether these animals are also susceptible to wild-type isolates of Marburg virus (MARV). Ferrets were challenged intramuscularly or intranasally with MARV strain Angola and monitored for 3 weeks. Unexpectedly, the animals neither showed observable signs of disease nor died of infection, and viremia was not detected after challenge. All animals were seropositive for MARV-specific immunoglobulin antibodies. Confirmatory studies with MARV strain Musoke and Ravn virus yielded the same outcomes. Therefore, ferrets may be of limited usefulness for studying the pathogenesis of MARV and Ravn virus infections.


Assuntos
Doença do Vírus de Marburg/imunologia , Animais , Modelos Animais de Doenças , Suscetibilidade a Doenças , Feminino , Furões , Masculino
3.
J Infect Dis ; 214(suppl 3): S326-S332, 2016 10 15.
Artigo em Inglês | MEDLINE | ID: mdl-27493239

RESUMO

A licensed vaccine against Ebola virus (EBOV) remains unavailable, despite >11 000 deaths from the 2014-2016 outbreak of EBOV disease in West Africa. Past studies have shown that recombinant vaccine viruses expressing EBOV glycoprotein (GP) are able to protect nonhuman primates (NHPs) from a lethal EBOV challenge. However, these vaccines express the viral GP-based EBOV variants found in Central Africa, which has 97.3% amino acid homology to the Makona variant found in West Africa. Our previous study showed that a recombinant adenovirus serotype 5 (Ad5)-vectored vaccine expressing the Makona EBOV GP (MakGP) was safe and immunogenic during clinical trials in China, but it is unknown whether the vaccine protects against EBOV infection. Here, we demonstrate that guinea pigs immunized with Ad5-MakGP developed robust humoral responses and were protected against exposure to guinea pig-adapted EBOV. Ad5-MakGP also elicited specific B- and T-cell immunity in NHPs and conferred 100% protection when animals were challenged 4 weeks after immunization. These results support further clinical development of this candidate and highlight the utility of Ad5-MakGP as a prophylactic measure in future outbreaks of EBOV disease.


Assuntos
Vacinas contra Adenovirus/imunologia , Vacinas contra Ebola/imunologia , Ebolavirus/imunologia , Doença pelo Vírus Ebola/prevenção & controle , Imunização , Proteínas Virais/imunologia , África Central , África Ocidental , Animais , China , Vetores Genéticos , Glicoproteínas/imunologia , Cobaias , Doença pelo Vírus Ebola/virologia , Humanos , Primatas
4.
J Infect Dis ; 214(suppl 3): S281-S289, 2016 10 15.
Artigo em Inglês | MEDLINE | ID: mdl-27651412

RESUMO

Enhanced virulence and/or transmission of West African Ebola virus (EBOV) variants, which are divergent from their Central African counterparts, are suspected to have contributed to the sizable toll of the recent Ebola virus disease (EVD) outbreak. This study evaluated the pathogenicity and shedding in rhesus macaques infected with 1 of 2 West African isolates (EBOV-C05 or EBOV-C07) or a Central African isolate (EBOV-K). All animals infected with EBOV-C05 or EBOV-C07 died of EVD, whereas 2 of 3 EBOV-K-infected animals died. The viremia level was elevated 10-fold in EBOV-C05-infected animals, compared with EBOV-C07- or EBOV-K-infected animals. More-severe lung pathology was observed in 2 of 6 EBOV-C05/C07-infected macaques. This is the first detailed analysis of the recently circulating EBOV-C05/C07 in direct comparison to EBOV-K with 6 animals per group, and it showed that EBOV-C05 but not EBOV-C07 can replicate at higher levels and cause more tissue damage in some animals. Increased virus shedding from individuals who are especially susceptible to EBOV replication is possibly one of the many challenges facing the community of healthcare and policy-making responders since the beginning of the outbreak.


Assuntos
Ebolavirus/patogenicidade , Doença pelo Vírus Ebola/virologia , Animais , Ebolavirus/fisiologia , Doença pelo Vírus Ebola/patologia , Humanos , Macaca mulatta , Especificidade da Espécie , Viremia , Virulência , Eliminação de Partículas Virais
5.
Genome Res ; 23(10): 1749-62, 2013 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-23800452

RESUMO

We have created a library of 2007 mutagenized Caenorhabditis elegans strains, each sequenced to a target depth of 15-fold coverage, to provide the research community with mutant alleles for each of the worm's more than 20,000 genes. The library contains over 800,000 unique single nucleotide variants (SNVs) with an average of eight nonsynonymous changes per gene and more than 16,000 insertion/deletion (indel) and copy number changes, providing an unprecedented genetic resource for this multicellular organism. To supplement this collection, we also sequenced 40 wild isolates, identifying more than 630,000 unique SNVs and 220,000 indels. Comparison of the two sets demonstrates that the mutant collection has a much richer array of both nonsense and missense mutations than the wild isolate set. We also find a wide range of rDNA and telomere repeat copy number in both sets. Scanning the mutant collection for molecular phenotypes reveals a nonsense suppressor as well as strains with higher levels of indels that harbor mutations in DNA repair genes and strains with abundant males associated with him mutations. All the strains are available through the Caenorhabditis Genetics Center and all the sequence changes have been deposited in WormBase and are available through an interactive website.


Assuntos
Caenorhabditis elegans/genética , Genes de Helmintos , Mutação , Alelos , Animais , Caenorhabditis elegans/classificação , Códon sem Sentido , Variações do Número de Cópias de DNA , DNA Ribossômico , Bases de Dados de Ácidos Nucleicos , Genes Essenciais , Genes Supressores , Variação Genética , Genoma Helmíntico , Genoma Mitocondrial , Heterozigoto , Mutação INDEL , Masculino , Mutação de Sentido Incorreto , Fenótipo , Polimorfismo de Nucleotídeo Único , Sequências de Repetição em Tandem
6.
J Infect Dis ; 212 Suppl 2: S234-41, 2015 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-25957966

RESUMO

BACKGROUND: The 2005 outbreak of Marburg virus (MARV) infection in Angola was the most lethal MARV infection outbreak in history, with a case-fatality rate (90%) similar to that for Zaire ebolavirus (EBOV) infection. However, very little is known about the pathogenicity of MARV Angola, as few studies have been conducted to date. Therefore, the immune response was examined in MARV Angola-infected nonhuman primates. METHODS: Cynomolgus macaques were infected with MARV Angola and monitored for survival. The effect of MARV Angola on the immune system was examined by immunophenotyping whole-blood and by analyzing cytokine and chemokine levels in plasma and spleen specimens, using flow cytometry. RESULTS: The prominent clinical findings were rapid onset of disease and death (mean time after infection, 6.7 days), fever, depression, anorexia, petechial rash, and lymphopenia. Specifically, T, B, and natural killer cells were severely depleted in the blood by day 6. The typical cytokine storm was present, with levels of interferon γ, tumor necrosis factor, interleukin 6, and CCL2 rising in the blood early during infection. CONCLUSIONS: MARV Angola displayed the same virulence and disease pathology as EBOV. MARV Angola appears to cause a more rapid onset and severe outcome of infection than other MARV strains.


Assuntos
Doença do Vírus de Marburg/imunologia , Marburgvirus/imunologia , Primatas/imunologia , Angola , Animais , Quimiocina CCL2/imunologia , Modelos Animais de Doenças , Ebolavirus/imunologia , Feminino , Interferon gama/imunologia , Interleucina-6/imunologia , Linfócitos/imunologia , Linfócitos/virologia , Macaca/imunologia , Macaca/virologia , Doença do Vírus de Marburg/virologia , Primatas/virologia , Baço/imunologia , Baço/virologia , Fator de Necrose Tumoral alfa/imunologia , Virulência/imunologia
7.
Mol Pharm ; 12(8): 2712-31, 2015 Aug 03.
Artigo em Inglês | MEDLINE | ID: mdl-25363619

RESUMO

As the Ebola outbreak in West Africa continues and cases appear in the United States and other countries, the need for long-lasting vaccines to preserve global health is imminent. Here, we evaluate the long-term efficacy of a respiratory and sublingual (SL) adenovirus-based vaccine in non-human primates in two phases. In the first, a single respiratory dose of 1.4×10(9) infectious virus particles (ivp)/kg of Ad-CAGoptZGP induced strong Ebola glycoprotein (GP) specific CD8+ and CD4+ T cell responses and Ebola GP-specific antibodies in systemic and mucosal compartments and was partially (67%) protective from challenge 62 days after immunization. The same dose given by the SL route induced Ebola GP-specific CD8+ T cell responses similar to that of intramuscular (IM) injection, however, the Ebola GP-specific antibody response was low. All primates succumbed to infection. Three primates were then given the vaccine in a formulation that improved the immune response to Ebola in rodents. Three primates were immunized with 2.0×10(10) ivp/kg of vaccine by the SL route. Diverse populations of polyfunctional Ebola GP-specific CD4+ and CD8+ T cells and significant anti-Ebola GP antibodies were present in samples collected 150 days after respiratory immunization. The formulated vaccine was fully protective against challenge 21 weeks after immunization. While diverse populations of Ebola GP-specific CD4+ T cells were produced after SL immunization, antibodies were not neutralizing and the vaccine was unprotective. To our knowledge, this is the first time that durable protection from a single dose respiratory adenovirus-based Ebola vaccine has been demonstrated in primates.


Assuntos
Adenoviridae/imunologia , Vacinas contra Ebola/administração & dosagem , Ebolavirus/imunologia , Doença pelo Vírus Ebola/prevenção & controle , Vacinas Sintéticas/administração & dosagem , Adenoviridae/genética , Animais , Células Cultivadas , Chlorocebus aethiops , Células HEK293 , Doença pelo Vírus Ebola/imunologia , Humanos , Macaca fascicularis , Masculino , Vacinação/métodos , Vacinas Sintéticas/genética , Células Vero
8.
J Virol ; 87(13): 7754-7, 2013 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-23616649

RESUMO

Monoclonal antibodies (MAbs) are currently a promising treatment strategy against Ebola virus infection. This study combined MAbs with an adenovirus-vectored interferon (DEF201) to evaluate the efficacy in guinea pigs and extend the treatment window obtained with MAbs alone. Initiating the combination therapy at 3 days postinfection (d.p.i.) provided 100% survival, a significant improvement over survival with either treatment alone. The administration of DEF201 within 2 d.p.i. permits later MAb use, with protective efficacy observed up to 8 d.p.i.


Assuntos
Anticorpos Monoclonais/imunologia , Anticorpos Monoclonais/uso terapêutico , Ebolavirus/imunologia , Doença pelo Vírus Ebola/tratamento farmacológico , Doença pelo Vírus Ebola/imunologia , Interferon-alfa/uso terapêutico , Adenoviridae , Animais , Vetores Genéticos/genética , Cobaias , Reação em Cadeia da Polimerase em Tempo Real , Reação em Cadeia da Polimerase Via Transcriptase Reversa
9.
Nature ; 445(7125): 319-23, 2007 Jan 18.
Artigo em Inglês | MEDLINE | ID: mdl-17230189

RESUMO

The 1918 influenza pandemic was unusually severe, resulting in about 50 million deaths worldwide. The 1918 virus is also highly pathogenic in mice, and studies have identified a multigenic origin of this virulent phenotype in mice. However, these initial characterizations of the 1918 virus did not address the question of its pathogenic potential in primates. Here we demonstrate that the 1918 virus caused a highly pathogenic respiratory infection in a cynomolgus macaque model that culminated in acute respiratory distress and a fatal outcome. Furthermore, infected animals mounted an immune response, characterized by dysregulation of the antiviral response, that was insufficient for protection, indicating that atypical host innate immune responses may contribute to lethality. The ability of influenza viruses to modulate host immune responses, such as that demonstrated for the avian H5N1 influenza viruses, may be a feature shared by the virulent influenza viruses.


Assuntos
Imunidade Inata/imunologia , Vírus da Influenza A Subtipo H1N1/imunologia , Vírus da Influenza A Subtipo H1N1/patogenicidade , Influenza Humana/imunologia , Influenza Humana/virologia , Macaca fascicularis/imunologia , Macaca fascicularis/virologia , Animais , Quimiocinas/sangue , Citocinas/sangue , Modelos Animais de Doenças , Feminino , Perfilação da Expressão Gênica , Regulação da Expressão Gênica , Humanos , Vírus da Influenza A Subtipo H1N1/genética , Influenza Humana/sangue , Pulmão/metabolismo , Pulmão/patologia , Pulmão/virologia , Camundongos , Camundongos Endogâmicos BALB C , Análise de Sequência com Séries de Oligonucleotídeos , Taxa de Sobrevida , Fatores de Tempo , Replicação Viral
10.
Nat Med ; 11(7): 786-90, 2005 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-15937495

RESUMO

Vaccines and therapies are urgently needed to address public health needs stemming from emerging pathogens and biological threat agents such as the filoviruses Ebola virus (EBOV) and Marburg virus (MARV). Here, we developed replication-competent vaccines against EBOV and MARV based on attenuated recombinant vesicular stomatitis virus vectors expressing either the EBOV glycoprotein or MARV glycoprotein. A single intramuscular injection of the EBOV or MARV vaccine elicited completely protective immune responses in nonhuman primates against lethal EBOV or MARV challenges. Notably, vaccine vector shedding was not detectable in the monkeys and none of the animals developed fever or other symptoms of illness associated with vaccination. The EBOV vaccine induced humoral and apparent cellular immune responses in all vaccinated monkeys, whereas the MARV vaccine induced a stronger humoral than cellular immune response. No evidence of EBOV or MARV replication was detected in any of the protected animals after challenge. Our data suggest that these vaccine candidates are safe and highly efficacious in a relevant animal model.


Assuntos
Ebolavirus/imunologia , Marburgvirus/imunologia , Vacinas Atenuadas/imunologia , Vacinas Combinadas/imunologia , Vacinas Virais/imunologia , Animais , Formação de Anticorpos , Reações Cruzadas , Vacinas contra Ebola/imunologia , Vacinas contra Ebola/farmacologia , Primatas , Vacinas Atenuadas/genética , Vacinas Atenuadas/farmacologia , Vacinas Combinadas/genética , Vacinas Combinadas/farmacologia , Vacinas Sintéticas/genética , Vacinas Sintéticas/imunologia , Vacinas Sintéticas/farmacologia , Vírus da Estomatite Vesicular Indiana/genética , Vacinas Virais/genética , Vacinas Virais/farmacologia , Viremia/imunologia , Viremia/virologia , Replicação Viral
11.
Clin Immunol ; 141(2): 218-27, 2011 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-21925951

RESUMO

Zaire ebolavirus (ZEBOV) can be transmitted by human-to-human contact and causes acute haemorrhagic fever with case fatality rates up to 90%. There are no effective therapeutic or prophylactic treatments available. The sole transmembrane glycoprotein (GP) is the key target for developing neutralizing antibodies. In this study, recombinant VSVΔG/ZEBOVGP was used to generate monoclonal antibodies (MAbs) against the ZEBOV GP. A total of 8 MAbs were produced using traditional hybridoma cell fusion technology, and then characterized by ELISA using ZEBOV VLPs, Western blotting, an immunofluorescence assay, and immunoprecipitation. All 8 MAbs worked in IFA and IP, suggesting that they are all conformational MAbs, however six of them recognized linearized epitopes by WB. ELISA results demonstrated that one MAb bound to a secreted GP (sGP 1-295aa); three bind to a part of the mucin domain (333-458aa); three MAbs recognized epitopes on the C-terminal domain of GP1 (296-501aa); and one bound to full length GP (VLPs/GP1,2 ΔTm). Using a mouse model these MAbs were evaluated for their therapeutic capacity during a lethal infection. All 8 MAb improved survival rates by 33%-100% against a high dose lethal challenge with mouse-adapted ZEBOV. This work has important implications for further development of vaccines and immunotherapies for ZEBOV infection.


Assuntos
Anticorpos Monoclonais Murinos/imunologia , Anticorpos Antivirais/imunologia , Antígenos Virais/imunologia , Ebolavirus/imunologia , Imunoglobulina G/imunologia , Proteínas do Envelope Viral/imunologia , Animais , Anticorpos Monoclonais Murinos/biossíntese , Anticorpos Antivirais/biossíntese , Especificidade de Anticorpos , Western Blotting , Relação Dose-Resposta Imunológica , Ensaio de Imunoadsorção Enzimática , Epitopos/imunologia , Feminino , Técnica Indireta de Fluorescência para Anticorpo , Doença pelo Vírus Ebola/terapia , Humanos , Hibridomas/imunologia , Imunização Passiva , Imunoglobulina G/biossíntese , Imunoprecipitação , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Estrutura Terciária de Proteína , Proteínas do Envelope Viral/química
12.
Nat Commun ; 12(1): 3612, 2021 06 14.
Artigo em Inglês | MEDLINE | ID: mdl-34127676

RESUMO

Widespread circulation of SARS-CoV-2 in humans raises the theoretical risk of reverse zoonosis events with wildlife, reintroductions of SARS-CoV-2 into permissive nondomesticated animals. Here we report that North American deer mice (Peromyscus maniculatus) are susceptible to SARS-CoV-2 infection following intranasal exposure to a human isolate, resulting in viral replication in the upper and lower respiratory tract with little or no signs of disease. Further, shed infectious virus is detectable in nasal washes, oropharyngeal and rectal swabs, and viral RNA is detectable in feces and occasionally urine. We further show that deer mice are capable of transmitting SARS-CoV-2 to naïve deer mice through direct contact. The extent to which these observations may translate to wild deer mouse populations remains unclear, and the risk of reverse zoonosis and/or the potential for the establishment of Peromyscus rodents as a North American reservoir for SARS-CoV-2 remains unknown.


Assuntos
COVID-19/veterinária , Peromyscus/virologia , Zoonoses/transmissão , Animais , Animais Selvagens , Anticorpos Neutralizantes/imunologia , COVID-19/patologia , COVID-19/transmissão , Suscetibilidade a Doenças , Fezes/virologia , Feminino , Histiócitos/patologia , Humanos , Masculino , Neutrófilos/imunologia , Neutrófilos/patologia , RNA Viral/isolamento & purificação , SARS-CoV-2/classificação , SARS-CoV-2/genética , Estados Unidos , Zoonoses/virologia
13.
PLoS Negl Trop Dis ; 14(4): e0008105, 2020 04.
Artigo em Inglês | MEDLINE | ID: mdl-32251473

RESUMO

Crimean-Congo hemorrhagic fever virus (CCHFV) is a tick-borne virus that can cause a hemorrhagic fever in humans, with a case fatality rate of up to 40%. Cases of CCHFV have been reported in Africa, Asia, and southern Europe; and recently, due to the expanding range of its vector, autochthonous cases have been reported in Spain. Although it was discovered over 70 years ago, our understanding of the pathogenesis of this virus remains limited. We used RNA-Seq in two human liver cell lines (HepG2 and Huh7) infected with CCHFV (strain IbAr10200), to examine kinetic changes in host expression and viral replication simultaneously at 1 and 3 days post infection. Through this, numerous host pathways were identified that were modulated by the virus including: antiviral response and endothelial cell leakage. Notably, the genes encoding DDX60, a cytosolic component of the RIG-I signalling pathway and OAS2 were both shown to be dysregulated. Interestingly, PTPRR was induced in Huh7 cells but not HepG2 cells. This has been associated with the TLR9 signalling cascade, and polymorphisms in TLR9 have been associated with poor outcomes in patients. Additionally, we performed whole-genome sequencing on CCHFV to assess viral diversity over time, and its relationship to the host response. As a result, we have demonstrated that through next-generation mRNA deep-sequencing it is possible to not only examine mRNA gene expression, but also to examine viral quasispecies and typing of the infecting strain. This demonstrates a proof-of-principle that CCHFV specimens can be analyzed to identify both the virus and host biomarkers that may have implications for prognosis.


Assuntos
Expressão Gênica , Vírus da Febre Hemorrágica da Crimeia-Congo/genética , Febre Hemorrágica da Crimeia/genética , Interações Hospedeiro-Patógeno/genética , Fígado/metabolismo , RNA-Seq/métodos , 2',5'-Oligoadenilato Sintetase/genética , Linhagem Celular , Proteína DEAD-box 58 , RNA Helicases DEAD-box/genética , Redes Reguladoras de Genes , Febre Hemorrágica da Crimeia/metabolismo , Febre Hemorrágica da Crimeia/virologia , Células Hep G2 , Interações Hospedeiro-Patógeno/fisiologia , Humanos , RNA Mensageiro , Receptores Imunológicos , Transdução de Sinais , Receptor Toll-Like 9 , Replicação Viral , Sequenciamento do Exoma
14.
PLoS Pathog ; 3(1): e2, 2007 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-17238284

RESUMO

Ebola viruses are highly lethal human pathogens that have received considerable attention in recent years due to an increasing re-emergence in Central Africa and a potential for use as a biological weapon. There is no vaccine or treatment licensed for human use. In the past, however, important advances have been made in developing preventive vaccines that are protective in animal models. In this regard, we showed that a single injection of a live-attenuated recombinant vesicular stomatitis virus vector expressing the Ebola virus glycoprotein completely protected rodents and nonhuman primates from lethal Ebola challenge. In contrast, progress in developing therapeutic interventions against Ebola virus infections has been much slower and there is clearly an urgent need to develop effective post-exposure strategies to respond to future outbreaks and acts of bioterrorism, as well as to treat laboratory exposures. Here we tested the efficacy of the vesicular stomatitis virus-based Ebola vaccine vector in post-exposure treatment in three relevant animal models. In the guinea pig and mouse models it was possible to protect 50% and 100% of the animals, respectively, following treatment as late as 24 h after lethal challenge. More important, four out of eight rhesus macaques were protected if treated 20 to 30 min following an otherwise uniformly lethal infection. Currently, this approach provides the most effective post-exposure treatment strategy for Ebola infections and is particularly suited for use in accidentally exposed individuals and in the control of secondary transmission during naturally occurring outbreaks or deliberate release.


Assuntos
Vacinas contra Ebola/uso terapêutico , Ebolavirus/imunologia , Doença pelo Vírus Ebola/prevenção & controle , Doença pelo Vírus Ebola/terapia , Animais , Surtos de Doenças , Cobaias , Humanos , Macaca mulatta , Camundongos , Dados de Sequência Molecular , Resultado do Tratamento
15.
G3 (Bethesda) ; 9(1): 135-144, 2019 01 09.
Artigo em Inglês | MEDLINE | ID: mdl-30420468

RESUMO

The Caenorhabditis elegans Gene Knockout Consortium is tasked with obtaining null mutations in each of the more than 20,000 open reading frames (ORFs) of this organism. To date, approximately 15,000 ORFs have associated putative null alleles. As there has been substantial success in using CRISPR/Cas9 in C. elegans, this appears to be the most promising technique to complete the task. To enhance the efficiency of using CRISPR/Cas9 to generate gene deletions in C. elegans we provide a web-based interface to access our database of guide RNAs (http://genome.sfu.ca/crispr). When coupled with previously developed selection vectors, optimization for homology arm length, and the use of purified Cas9 protein, we demonstrate a robust and effective protocol for generating deletions for this large-scale project. Debate and speculation in the larger scientific community concerning off-target effects due to non-specific Cas9 cutting has prompted us to investigate through whole genome sequencing the occurrence of single nucleotide variants and indels accompanying targeted deletions. We did not detect any off-site variants above the natural spontaneous mutation rate and therefore conclude that this modified protocol does not generate off-target events to any significant degree in C. elegans We did, however, observe a number of non-specific alterations at the target site itself following the Cas9-induced double-strand break and offer a protocol for best practice quality control for such events.


Assuntos
Sistemas CRISPR-Cas/genética , Caenorhabditis elegans/genética , Edição de Genes , Recombinação Homóloga/genética , Animais , Deleção de Genes , Técnicas de Inativação de Genes , Marcação de Genes , Mutagênese/genética
16.
Lancet ; 367(9520): 1399-404, 2006 Apr 29.
Artigo em Inglês | MEDLINE | ID: mdl-16650649

RESUMO

BACKGROUND: Effective countermeasures are urgently needed to prevent and treat infections caused by highly pathogenic and biological threat agents such as Marburg virus (MARV). We aimed to test the efficacy of a replication-competent vaccine based on attenuated recombinant vesicular stomatitis virus (rVSV), as a postexposure treatment for MARV haemorrhagic fever. METHODS: We used a rhesus macaque model of MARV haemorrhagic fever that produced 100% lethality. We administered rVSV vectors expressing the MARV Musoke strain glycoprotein to five macaques 20-30 min after a high-dose lethal injection of homologous MARV. Three animals were MARV-positive controls and received non-specific rVSV vectors. We tested for viraemia, undertook analyses for haematology and serum biochemistry, and measured humoral and cellular immune responses. FINDINGS: All five rhesus monkeys that were treated with the rVSV MARV vectors as a postexposure treatment survived a high-dose lethal challenge of MARV for at least 80 days. None of these five animals developed clinical symptoms consistent with MARV haemorrhagic fever. All the control animals developed fulminant disease and succumbed to the MARV challenge by day 12. MARV disease in the controls was indicated by: high titres of MARV (10(3)-10(5) plaque-forming units per mL); development of leucocytosis with concurrent neutrophilia at end-stage disease; and possible damage to the liver, kidney, and pancreas. INTERPRETATION: Postexposure protection against MARV in non-human primates provides a paradigm for the treatment of MARV haemorrhagic fever. Indeed, these data suggest that rVSV-based filoviral vaccines might not only have potential as preventive vaccines, but also could be equally useful for postexposure treatment of filoviral infections.


Assuntos
Doença do Vírus de Marburg/prevenção & controle , Marburgvirus/imunologia , Vírus da Estomatite Vesicular Indiana/imunologia , Vacinas Virais , Animais , Humanos , Imunoglobulina G/sangue , Imunoglobulina M/sangue , Macaca mulatta , Doença do Vírus de Marburg/imunologia
17.
Sci Transl Med ; 8(329): 329ra33, 2016 Mar 09.
Artigo em Inglês | MEDLINE | ID: mdl-26962157

RESUMO

The 2014-2015 Ebola virus (EBOV) outbreak in West Africa highlighted the urgent need for specific therapeutic interventions for infected patients. The human-mouse chimeric monoclonal antibody (mAb) cocktail ZMapp, previously shown to be efficacious in EBOV (variant Kikwit) lethally infected nonhuman primates (NHPs) when administration was initiated up to 5 days, was used in some patients during the outbreak. We show that a two-antibody cocktail, MIL77E, is fully protective in NHPs when administered at 50 mg/kg 3 days after challenge with a lethal dose of EBOV variant Makona, the virus responsible for the ongoing 2014-2015 outbreak, whereas a similar formulation of ZMapp protected two of three NHPs. The chimeric MIL77E mAb cocktail is produced in engineered Chinese hamster ovary cells and is based on mAbs c13C6 and c2G4 from ZMapp. The use of only two antibodies in MIL77E opens the door to a pan-ebolavirus cocktail.


Assuntos
Anticorpos Monoclonais/uso terapêutico , Ebolavirus/imunologia , Doença pelo Vírus Ebola/tratamento farmacológico , Doença pelo Vírus Ebola/prevenção & controle , Animais , Células CHO , Cromatografia Líquida , Cricetinae , Cricetulus , Feminino , Cobaias , Macaca , Peptídeo-N4-(N-acetil-beta-glucosaminil) Asparagina Amidase/metabolismo
18.
PLoS Med ; 2(6): e183, 2005 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-15971954

RESUMO

BACKGROUND: Recent importation of Lassa fever into Germany, the Netherlands, the United Kingdom, and the United States by travelers on commercial airlines from Africa underscores the public health challenge of emerging viruses. Currently, there are no licensed vaccines for Lassa fever, and no experimental vaccine has completely protected nonhuman primates against a lethal challenge. METHODS AND FINDINGS: We developed a replication-competent vaccine against Lassa virus based on attenuated recombinant vesicular stomatitis virus vectors expressing the Lassa viral glycoprotein. A single intramuscular vaccination of the Lassa vaccine elicited a protective immune response in nonhuman primates against a lethal Lassa virus challenge. Vaccine shedding was not detected in the monkeys, and none of the animals developed fever or other symptoms of illness associated with vaccination. The Lassa vaccine induced strong humoral and cellular immune responses in the four vaccinated and challenged monkeys. Despite a transient Lassa viremia in vaccinated animals 7 d after challenge, the vaccinated animals showed no evidence of clinical disease. In contrast, the two control animals developed severe symptoms including rashes, facial edema, and elevated liver enzymes, and ultimately succumbed to the Lassa infection. CONCLUSION: Our data suggest that the Lassa vaccine candidate based on recombinant vesicular stomatitis virus is safe and highly efficacious in a relevant animal model that faithfully reproduces human disease.


Assuntos
Febre Lassa/prevenção & controle , Vírus Lassa/imunologia , Vacinas Virais , Animais , Modelos Animais de Doenças , Avaliação Pré-Clínica de Medicamentos , Febre Lassa/imunologia , Macaca fascicularis , Vacinação , Vacinas Sintéticas , Vacinas Virais/imunologia , Viremia/prevenção & controle
19.
Virol J ; 2: 42, 2005 Apr 25.
Artigo em Inglês | MEDLINE | ID: mdl-15850490

RESUMO

BACKGROUND: Crimean-Congo Hemorrhagic Fever virus (CCHFV), a member of the genus Nairovirus, family Bunyaviridae, is a tick-borne pathogen causing severe disease in humans. To better understand the CCHFV life cycle and explore potential intervention strategies, we studied the biosynthesis and intracellular targeting of the glycoproteins, which are encoded by the M genome segment. RESULTS: Following determination of the complete genome sequence of the CCHFV reference strain IbAr10200, we generated expression plasmids for the individual expression of the glycoproteins GN and GC, using CMV- and chicken beta-actin-driven promoters. The cellular localization of recombinantly expressed CCHFV glycoproteins was compared to authentic glycoproteins expressed during virus infection using indirect immunofluorescence assays, subcellular fractionation/western blot assays and confocal microscopy. To further elucidate potential intracellular targeting/retention signals of the two glycoproteins, GFP-fusion proteins containing different parts of the CCHFV glycoprotein were analyzed for their intracellular targeting. The N-terminal glycoprotein GN localized to the Golgi complex, a process mediated by retention/targeting signal(s) in the cytoplasmic domain and ectodomain of this protein. In contrast, the C-terminal glycoprotein GC remained in the endoplasmic reticulum but could be rescued into the Golgi complex by co-expression of GN. CONCLUSION: The data are consistent with the intracellular targeting of most bunyavirus glycoproteins and support the general model for assembly and budding of bunyavirus particles in the Golgi compartment.


Assuntos
Glicoproteínas/metabolismo , Vírus da Febre Hemorrágica da Crimeia-Congo/metabolismo , Proteínas não Estruturais Virais/metabolismo , Animais , Linhagem Celular , Chlorocebus aethiops , Cricetinae , Citoplasma , Retículo Endoplasmático , Regulação Viral da Expressão Gênica , Glicoproteínas/genética , Complexo de Golgi , Humanos , Imuno-Histoquímica , Proteínas não Estruturais Virais/genética
20.
Viral Immunol ; 28(1): 51-61, 2015 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-25494457

RESUMO

Members of the species Zaire ebolavirus cause severe hemorrhagic fever with up to a 90% mortality rate in humans. The VSVΔG/EBOV GP vaccine has provided 100% protection in the mouse, guinea pig, and nonhuman primate (NHP) models, and has also been utilized as a post-exposure therapeutic to protect mice, guinea pigs, and NHPs from a lethal challenge of Ebola virus (EBOV). EBOV infection causes rapid mortality in human and animal models, with death occurring as early as 6 days after infection, suggesting a vital role for the innate immune system to control the infection before cells of the adaptive immune system can assume control. Natural killer (NK) cells are the predominant cell of the innate immune response, which has been shown to expand with VSVΔG/EBOV GP treatment. In the current study, an in vivo mouse model of the VSVΔG/EBOV GP post-exposure treatment was used for a mouse adapted (MA)-EBOV infection, to determine the putative VSVΔG/EBOV GP-induced protective mechanism of NK cells. NK depletion studies demonstrated that mice with NK cells survive longer in a MA-EBOV infection, which is further enhanced with VSVΔG/EBOV GP treatment. NK cell mediated cytotoxicity and IFN-γ secretion was significantly higher with VSVΔG/EBOV GP treatment. Cell mediated cytotoxicity assays and perforin knockout mice experiments suggest that there are perforin-dependent and -independent mechanisms involved. Together, these data suggest that NK cells play an important role in VSVΔG/EBOV GP-induced protection of EBOV by increasing NK cytotoxicity, and IFN-γ secretion.


Assuntos
Portadores de Fármacos , Vacinas contra Ebola/imunologia , Ebolavirus/imunologia , Doença pelo Vírus Ebola/prevenção & controle , Células Matadoras Naturais/imunologia , Vesiculovirus/genética , Proteínas do Envelope Viral/imunologia , Animais , Antígenos Virais/genética , Antígenos Virais/imunologia , Testes Imunológicos de Citotoxicidade , Modelos Animais de Doenças , Vacinas contra Ebola/administração & dosagem , Vacinas contra Ebola/genética , Ebolavirus/genética , Feminino , Vetores Genéticos , Interferon gama/metabolismo , Masculino , Camundongos Endogâmicos BALB C , Camundongos Knockout , Análise de Sobrevida , Proteínas do Envelope Viral/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA