Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Viruses ; 16(7)2024 Jul 16.
Artigo em Inglês | MEDLINE | ID: mdl-39066299

RESUMO

Influenza A viruses (IAV) utilize host proteins throughout their life cycle to infect and replicate in their hosts. We previously showed that host adaptive mutations in avian IAV PA help recruit host protein G-Rich RNA Sequence Binding Factor 1 (GRSF1) to the nucleoprotein (NP) 5' untranslated region (UTR), leading to the enhanced nuclear export and translation of NP mRNA. In this study, we evaluated the impact of GRSF1 in the viral life cycle. We rescued and characterized a 2009 pH1N1 virus with a mutated GRSF1 binding site in the 5' UTR of NP mRNA. Mutant viral growth was attenuated relative to pH1N1 wild-type (WT) in mammalian cells. We observed a specific reduction in the NP protein production and cytosolic accumulation of NP mRNAs, indicating a critical role of GRSF1 in the nuclear export of IAV NP mRNAs. Further, in vitro-transcribed mutated NP mRNA was translated less efficiently than WT NP mRNA in transfected cells. Together, these findings show that GRSF1 binding is important for both mRNA nuclear export and translation and affects overall IAV growth. Enhanced association of GRSF1 to NP mRNA by PA mutations leads to rapid virus growth, which could be a key process of mammalian host adaptation of IAV.


Assuntos
Transporte Ativo do Núcleo Celular , Biossíntese de Proteínas , RNA Mensageiro , RNA Viral , Humanos , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , RNA Viral/genética , RNA Viral/metabolismo , Animais , Vírus da Influenza A/genética , Vírus da Influenza A/fisiologia , Vírus da Influenza A/metabolismo , Replicação Viral , Vírus da Influenza A Subtipo H1N1/genética , Vírus da Influenza A Subtipo H1N1/metabolismo , Vírus da Influenza A Subtipo H1N1/fisiologia , Núcleo Celular/metabolismo , Núcleo Celular/virologia , Regiões 5' não Traduzidas/genética , Proteínas do Nucleocapsídeo/metabolismo , Proteínas do Nucleocapsídeo/genética , Células Madin Darby de Rim Canino , Células HEK293 , Proteínas de Ligação a RNA/metabolismo , Proteínas de Ligação a RNA/genética , Cães , Influenza Humana/virologia , Influenza Humana/metabolismo , Influenza Humana/genética , Mutação , Interações Hospedeiro-Patógeno/genética , Proteínas do Core Viral/metabolismo , Proteínas do Core Viral/genética
2.
Hum Vaccin Immunother ; 17(2): 554-559, 2021 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-32750273

RESUMO

SeVRSV is a replication-competent Sendai virus (SeV)-based vaccine carrying the respiratory syncytial virus (RSV) fusion protein (F) gene. Unmanipulated, non-recombinant SeV is a murine parainfluenza virus type 1 (PIV-1) and serves as a Jennerian vaccine for human PIV-1 (hPIV-1). SeV protects African green monkeys (AGM) from infection after hPIV-1 challenge. The recombinant SeVRSV additionally targets RSV and protects AGM from lower respiratory infections after RSV challenge. The present study is the first to report on the safety, viral genome detection, and immunogenicity following SeVRSV vaccination of healthy adults. Seventeen and four healthy adults received intranasal SeVRSV and PBS, respectively, followed by six months of safety monitoring. Virus genome (in nasal wash) and vaccine-specific antibodies (in sera) were monitored for two and four weeks, respectively, post-vaccination. The vaccine was well-tolerated with only mild to moderate reactions that were also present in the placebo group. No severe reactions occurred. As expected, due to preexisting immunity toward hPIV-1 and RSV in adults, vaccine genome detection was transient. There were minimal antibody responses to SeV and negligible responses to RSV F. Results encourage further studies of SeVRSV with progression toward a clinical trial in seronegative children. Abbreviations: AE-adverse event; SAE-serious adverse event; SeV-Sendai virus; RSV-respiratory syncytial virus; PIV-1-parainfluenza virus-type 1; hPIV-1-human parainfluenza virus-type 1; F-RSV fusion protein; SeVRSV-recombinant SeV carrying the RSV F gene; Ab-antibody; MSW-medically significant wheezing; NOCMC-new onset chronic medical condition, mITT-modified Intent to Treat; ALRI-acute lower respiratory tract infection.


Assuntos
Infecções por Vírus Respiratório Sincicial , Vacinas contra Vírus Sincicial Respiratório , Vírus Sincicial Respiratório Humano , Adulto , Animais , Anticorpos Antivirais , Chlorocebus aethiops , Humanos , Imunogenicidade da Vacina , Vírus da Parainfluenza 1 Humana/genética , Infecções por Vírus Respiratório Sincicial/prevenção & controle , Vacinas contra Vírus Sincicial Respiratório/efeitos adversos , Vacinas contra Vírus Sincicial Respiratório/genética , Vírus Sincicial Respiratório Humano/genética , Vírus Sendai/genética , Proteínas Virais de Fusão/genética
3.
Virology ; 509: 60-66, 2017 09.
Artigo em Inglês | MEDLINE | ID: mdl-28605636

RESUMO

Human metapneumovirus (hMPV) infections pose a serious health risk to young children, particularly in cases of premature birth. No licensed vaccine exists and there is no standard treatment for hMPV infections apart from supportive hospital care. We describe the production of a Sendai virus (SeV) recombinant that carries a gene for a truncated hMPV fusion (F) protein (SeV-MPV-Ft). The vaccine induces binding and neutralizing antibody responses toward hMPV and protection against challenge with hMPV in a cotton rat system. Results encourage advanced development of SeV-MPV-Ft to prevent the morbidity and mortality caused by hMPV infections in young children.


Assuntos
Antígenos Virais/imunologia , Portadores de Fármacos , Metapneumovirus/imunologia , Infecções por Paramyxoviridae/prevenção & controle , Vírus Sendai/genética , Proteínas Virais de Fusão/imunologia , Vacinas Virais/imunologia , Animais , Anticorpos Neutralizantes/sangue , Anticorpos Antivirais/sangue , Antígenos Virais/genética , Modelos Animais de Doenças , Metapneumovirus/genética , Infecções por Paramyxoviridae/imunologia , Sigmodontinae , Resultado do Tratamento , Vacinas Sintéticas/administração & dosagem , Vacinas Sintéticas/genética , Vacinas Sintéticas/imunologia , Proteínas Virais de Fusão/genética , Vacinas Virais/administração & dosagem , Vacinas Virais/genética
4.
Virology ; 505: 23-32, 2017 05.
Artigo em Inglês | MEDLINE | ID: mdl-28219018

RESUMO

Influenza A viruses contain eight single-stranded, negative-sense RNA segments as viral genomes in the form of viral ribonucleoproteins (vRNPs). During genome replication in the nucleus, positive-sense complementary RNPs (cRNPs) are produced as replicative intermediates, which are not incorporated into progeny virions. To analyze the mechanism of selective vRNP incorporation into progeny virions, we quantified vRNPs and cRNPs in the nuclear and cytosolic fractions of infected cells, using a strand-specific qRT-PCR. Unexpectedly, we found that cRNPs were also exported to the cytoplasm. This export was chromosome region maintenance 1 (CRM1)-independent unlike that of vRNPs. Although both vRNPs and cRNPs were present in the cytosol, viral matrix (M1) protein, a key regulator for viral assembly, preferentially bound vRNPs over cRNPs. These results indicate that influenza A viruses selectively uptake cytosolic vRNPs through a specific interaction with M1 during viral assembly.


Assuntos
Vírus da Influenza A/genética , RNA Viral/genética , Ribonucleoproteínas/metabolismo , Proteínas da Matriz Viral/metabolismo , Montagem de Vírus/fisiologia , Replicação Viral/fisiologia , Células A549 , Transporte Ativo do Núcleo Celular , Animais , Linhagem Celular Tumoral , Núcleo Celular/virologia , Citoplasma/virologia , Cães , Genoma Viral/genética , Células HEK293 , Humanos , Carioferinas/metabolismo , Células Madin Darby de Rim Canino , RNA Viral/metabolismo , Receptores Citoplasmáticos e Nucleares/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Proteínas da Matriz Viral/genética , Montagem de Vírus/genética , Replicação Viral/genética , Proteína Exportina 1
5.
Int Immunol ; 27(5): 229-36, 2015 May.
Artigo em Inglês | MEDLINE | ID: mdl-25477211

RESUMO

The respiratory syncytial virus (RSV) is responsible for as many as 199000 annual deaths worldwide. Currently, there is no standard treatment for RSV disease and no vaccine. Sendai virus (SeV) is an attractive pediatric vaccine candidate because it elicits robust and long-lasting virus-specific B cell and T cell activities in systemic and mucosal tissues. The virus serves as a gene delivery system as well as a Jennerian vaccine against its close cousin, human parainfluenza virus type 1. Here we describe the testing of a recombinant SeV (SeVRSV-Fs) that expresses an unconstrained, secreted RSV-F protein as a vaccine against RSV in cotton rats. After a single intranasal immunization of cotton rats with SeVRSV-Fs, RSV-specific binding and neutralizing antibodies were generated. These antibodies exhibited cross-reactivity with both RSV A and B isolates. RSV-F-specific IFN-γ-producing T cells were also activated. The SeVRSV-Fs vaccine conferred protection against RSV challenge without enhanced immunopathology. In total, results showed that an SeV recombinant that expresses RSV F in an unconstrained, soluble form can induce humoral and cellular immunity that protects against infection with RSV.


Assuntos
Infecções por Vírus Respiratório Sincicial/prevenção & controle , Vírus Sinciciais Respiratórios/metabolismo , Vírus Sendai/imunologia , Linfócitos T/imunologia , Vacinas Virais/administração & dosagem , Administração Intranasal , Animais , Anticorpos Antivirais/metabolismo , Modelos Animais de Doenças , Técnicas de Transferência de Genes , Humanos , Imunização , Interferon gama/metabolismo , Ativação Linfocitária , Ratos , Proteínas Recombinantes de Fusão/genética , Infecções por Vírus Respiratório Sincicial/imunologia , Vírus Sinciciais Respiratórios/genética , Vírus Sendai/genética , Sigmodontinae , Linfócitos T/virologia , Proteínas Virais de Fusão/genética , Vacinas Virais/genética
6.
PLoS One ; 8(4): e61281, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23593451

RESUMO

Interactions between viral glycoproteins, matrix protein and nucleocapsid sustain assembly of parainfluenza viruses at the plasma membrane. Although the protein interactions required for virion formation are considered to be highly specific, virions lacking envelope glycoprotein(s) can be produced, thus the molecular interactions driving viral assembly and production are still unclear. Sendai virus (SeV) and human parainfluenza virus type 1 (hPIV1) are highly similar in structure, however, the cytoplasmic tail sequences of the envelope glycoproteins (HN and F) are relatively less conserved. To unveil the specific role of the envelope glycoproteins in viral assembly, we created chimeric SeVs whose HN (rSeVhHN) or HN and F (rSeVh(HN+F)) were replaced with those of hPIV1. rSeVhHN grew as efficiently as wt SeV or hPIV1, suggesting that the sequence difference in HN does not have a significant impact on SeV replication and virion production. In sharp contrast, the growth of rSeVh(HN+F) was significantly impaired compared to rSeVhHN. rSeVh(HN+Fstail) which expresses a chimeric hPIV1 F with the SeV cytoplasmic tail sequence grew similar to wt SeV or rSeVhHN. Further analysis indicated that the F cytoplasmic tail plays a critical role in cell surface expression/accumulation of HN and F, as well as NP and M association at the plasma membrane. Trafficking of nucelocapsids in infected cells was not significantly affected by the origin of F, suggesting that F cytoplasmic tail is not involved in intracellular movement. These results demonstrate the role of the F cytoplasmic tail in accumulation of structural components at the plasma membrane assembly sites.


Assuntos
Citoplasma/metabolismo , Vírus da Parainfluenza 1 Humana/fisiologia , Proteínas Virais de Fusão/química , Proteínas Virais de Fusão/metabolismo , Montagem de Vírus/fisiologia , Sequência de Aminoácidos , Linhagem Celular , Membrana Celular/metabolismo , Proteínas de Fluorescência Verde/metabolismo , Humanos , Cinética , Dados de Sequência Molecular , Vírus da Parainfluenza 1 Humana/crescimento & desenvolvimento , Transporte Proteico , Ribonucleoproteínas/metabolismo , Relação Estrutura-Atividade , Frações Subcelulares/metabolismo
7.
Vaccine ; 30(5): 959-68, 2012 Jan 20.
Artigo em Inglês | MEDLINE | ID: mdl-22119594

RESUMO

Respiratory syncytial virus (RSV) is a serious disease of children, responsible for an estimated 160,000 deaths per year worldwide. Despite the ongoing need for global prevention of RSV and decades of research, there remains no licensed vaccine. Sendai virus (SeV) is a mouse parainfluenza virus-type 1 which has been previously shown to confer protection against its human cousin, human parainfluenza virus-type 1 in African green monkeys (AGM). Here is described the study of a RSV vaccine (SeVRSV), produced by reverse genetics technology using SeV as a backbone to carry the full-length gene for RSV F. To test for immunogenicity, efficacy and safety, the vaccine was administered to AGM by intratracheal (i.t.) and intranasal (i.n.) routes. Control animals received the empty SeV vector or PBS. There were no booster immunizations. SeV and SeVRSV were cleared from the URT and LRT of vaccinated animals by day 10. Antibodies with specificities toward SeV and RSV were detected in SeVRSV primed animals as early as day ten after immunizations in both sera and nasal wash samples. One month after immunization all test and control AGM received an i.n. challenge with RSV-A2. SeVRSV-vaccinated animals exhibited reduced RSV in the URT compared to controls, and complete protection against RSV in the LRT. There were no clinically relevant adverse events associated with vaccination either before or after challenge. These data encourage advanced testing of the SeVRSV vaccine candidate in clinical trials for protection against RSV.


Assuntos
Portadores de Fármacos , Vetores Genéticos , Infecções por Vírus Respiratório Sincicial/prevenção & controle , Vacinas contra Vírus Sincicial Respiratório/imunologia , Vírus Sendai/genética , Proteínas Virais de Fusão/genética , Proteínas Virais de Fusão/imunologia , Administração por Inalação , Administração Intranasal , Animais , Anticorpos Antivirais/sangue , Chlorocebus aethiops , Modelos Animais de Doenças , Vacinas contra Vírus Sincicial Respiratório/administração & dosagem , Vacinas contra Vírus Sincicial Respiratório/efeitos adversos , Vacinas contra Vírus Sincicial Respiratório/genética , Vacinas Sintéticas/administração & dosagem , Vacinas Sintéticas/efeitos adversos , Vacinas Sintéticas/genética , Vacinas Sintéticas/imunologia
8.
Biol Pharm Bull ; 34(7): 996-1000, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21720003

RESUMO

Human parainfluenza virus type 1 (hPIV1) generally does not show visible plaques in common cell lines, including Lewis lung carcinoma-monkey kidney (LLC-MK(2)) cells, by plaque formation assays for human parainfluenza virus type 3 (hPIV3) and Sendai virus. In several conditions of the plaque formation assay, complete elimination of serum proteins in the overlay medium was necessary for visualization of hPIV1-induced plaque formation in LLC-MK(2) cells. We developed a plaque formation assay for hPIV1 isolation and titration in LLC-MK(2) cells using an initial overlay medium of bovine serum albumin-free Eagle's minimum essential medium containing agarose and acetylated trypsin for 4-6 d followed by a second overlay staining medium containing agarose and neutral red. The assay allowed both laboratory and clinical hPIV1 strains to form large plaques. The plaque reduction assay was also performed with rabbit anti-hPIV1 antibody as a general evaluation model of viral inhibitors to decrease both the plaque number and size. The results indicate that the plaque formation assay is useful for hPIV1 isolation, titration, evaluation of antiviral reagents and epidemiologic research.


Assuntos
Vírus da Parainfluenza 1 Humana/crescimento & desenvolvimento , Ensaio de Placa Viral , Animais , Linhagem Celular , Meios de Cultura
9.
PLoS One ; 5(4): e10372, 2010 Apr 29.
Artigo em Inglês | MEDLINE | ID: mdl-20454455

RESUMO

BACKGROUND: It is widely accepted that the highly error prone replication process of influenza A virus (IAV), together with viral genome assortment, facilitates the efficient evolutionary capacity of IAV. Therefore, it has been logically assumed that the enzyme responsible for viral RNA replication process, influenza virus type A RNA polymerase (IAV Pol), is a highly error-prone polymerase which provides the genomic mutations necessary for viral evolution and host adaptation. Importantly, however, the actual enzyme fidelity of IAV RNA polymerase has never been characterized. PRINCIPAL FINDINGS: Here we established new biochemical assay conditions that enabled us to assess both polymerase activity with physiological NTP pools and enzyme fidelity of IAV Pol. We report that IAV Pol displays highly active RNA-dependent RNA polymerase activity at unbiased physiological NTP substrate concentrations. With this robust enzyme activity, for the first time, we were able to compare the enzyme fidelity of IAV Pol complex with that of bacterial phage T7 RNA polymerase and the reverse transcriptases (RT) of human immunodeficiency virus (HIV-1) and murine leukemia virus (MuLV), which are known to be low and high fidelity enzymes, respectively. We observed that IAV Pol displayed significantly higher fidelity than HIV-1 RT and T7 RNA polymerase and equivalent or higher fidelity than MuLV RT. In addition, the IAV Pol complex showed increased fidelity at lower temperatures. Moreover, upon replacement of Mg(++) with Mn(++), IAV Pol displayed increased polymerase activity, but with significantly reduced processivity, and misincorporation was slightly elevated in the presence of Mn(++). Finally, when the IAV nucleoprotein (NP) was included in the reactions, the IAV Pol complex exhibited enhanced polymerase activity with increased fidelity. SIGNIFICANCE: Our study indicates that IAV Pol is a high fidelity enzyme. We envision that the high fidelity nature of IAV Pol may be important to counter-balance the multiple rounds of IAV genome amplification per infection cycle, which provides IAV Pol with ample opportunities to generate and amplify genomic founder mutations, and thus achieve optimal viral mutagenesis for its evolution.


Assuntos
RNA Polimerases Dirigidas por DNA/metabolismo , Vírus da Influenza A/enzimologia , RNA Polimerases Dirigidas por DNA/química , Transcriptase Reversa do HIV/química , Transcriptase Reversa do HIV/metabolismo , Humanos , Vírus da Leucemia Murina/enzimologia , DNA Polimerase Dirigida por RNA/química , DNA Polimerase Dirigida por RNA/metabolismo , Especificidade por Substrato , Proteínas Virais/química , Proteínas Virais/metabolismo
10.
Virology ; 400(2): 224-32, 2010 May 10.
Artigo em Inglês | MEDLINE | ID: mdl-20185156

RESUMO

Influenza virus leads to acute respiratory disease resulting in seasonal epidemics and periodic pandemics. Little is known about the signaling events that regulate host defense to influenza. One particular pathway, the c-Jun amino-terminal kinase (JNK) cascade is activated following influenza infection and blocking JNK leads to enhanced viral replication. We hypothesize that Mixed Lineage Kinase 3 (MLK3), an upstream regulator of JNK, is involved in the host response to influenza. To test this, wild-type and MLK3-/- mice were infected with pathogenic strain of influenza A virus, A/PR/8/34 (PR8). Although, cellular and humoral immune responses were similar between wild-type and MLK3-/- hosts, the viral load in the lungs was comparatively higher in MLK3-/- mice at day 8 post-infection. Consistent with this, MLK3-/- murine lung fibroblast and epithelial cells had prolonged survival and increased virion production following infection compared to wild-type. These findings support a role for MLK3 in viral production during influenza infection.


Assuntos
Efeito Citopatogênico Viral , Vírus da Influenza A/imunologia , Pulmão/virologia , MAP Quinase Quinase Quinases/metabolismo , Infecções por Orthomyxoviridae/imunologia , Infecções por Orthomyxoviridae/patologia , Animais , Anticorpos Antivirais/imunologia , Sobrevivência Celular , Células Epiteliais/virologia , Fibroblastos/virologia , MAP Quinase Quinase Quinases/deficiência , Camundongos , Camundongos Knockout , Infecções por Orthomyxoviridae/virologia , Linfócitos T/imunologia , Carga Viral , MAP Quinase Quinase Quinase 11 Ativada por Mitógeno
11.
J Gen Virol ; 89(Pt 12): 2923-2932, 2008 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-19008377

RESUMO

Influenza A virus (IAV) replicates in the upper respiratory tract of humans at 33 degrees C and in the intestinal tract of birds at close to 41 degrees C. The viral RNA polymerase complex comprises three subunits (PA, PB1 and PB2) and plays an important role in host adaptation. We therefore developed an in vitro system to examine the temperature sensitivity of IAV RNA polymerase complexes from different origins. Complexes were prepared from human lung epithelial cells (A549) using a novel adenoviral expression system. Affinity-purified complexes were generated that contained either all three subunits (PA/PB1/PB2) from the A/Viet/1203/04 H5N1 virus (H/H/H) or the A/WSN/33 H1N1 strain (W/W/W). We also prepared chimeric complexes in which the PB2 subunit was exchanged (H/H/W, W/W/H) or substituted with an avian PB2 from the A/chicken/Nanchang/3-120/01 H3N2 strain (W/W/N). All complexes were functional in transcription, cap-binding and endonucleolytic activity. Complexes containing the H5N1 or Nanchang PB2 protein retained transcriptional activity over a broad temperature range (30-42 degrees C). In contrast, complexes containing the WSN PB2 protein lost activity at elevated temperatures (39 degrees C or higher). The E627K mutation in the avian PB2 was not required for this effect. Finally, the avian PB2 subunit was shown to confer enhanced stability to the WSN 3P complex. These results show that PB2 plays an important role in regulating the temperature optimum for IAV RNA polymerase activity, possibly due to effects on the functional stability of the 3P complex.


Assuntos
Vírus da Influenza A Subtipo H1N1/enzimologia , Virus da Influenza A Subtipo H5N1/enzimologia , Complexos Multienzimáticos/metabolismo , RNA Polimerase Dependente de RNA/metabolismo , Temperatura , Proteínas Virais/metabolismo , Animais , Linhagem Celular , Células Endoteliais , Estabilidade Enzimática , Humanos , Rim/citologia , Pulmão/citologia , Camundongos , Complexos Multienzimáticos/isolamento & purificação , RNA Polimerase Dependente de RNA/isolamento & purificação , Proteínas Virais/isolamento & purificação
12.
Vaccine ; 26(27-28): 3480-8, 2008 Jun 25.
Artigo em Inglês | MEDLINE | ID: mdl-18499307

RESUMO

The human parainfluenza viruses (hPIVs) and respiratory syncytial virus (RSV) are the leading causes of serious respiratory illness in the human pediatric population. Despite decades of research, there are currently no licensed vaccines for either the hPIV or RSV pathogens. Here we describe the testing of hPIV-3 and RSV candidate vaccines using Sendai virus (SeV, murine PIV-1) as a vector. SeV was selected as the vaccine backbone, because it has been shown to elicit robust and durable immune activities in animal studies, and has already advanced to human safety trials as a xenogenic vaccine for hPIV-1. Two new SeV-based hPIV-3 vaccine candidates were first generated by inserting either the fusion (F) gene or hemagglutinin-neuraminidase (HN) gene from hPIV-3 into SeV. The resultant rSeV-hPIV3-F and rSeV-hPIV3-HN vaccines expressed their inserted hPIV-3 genes upon infection. The inoculation of either vaccine into cotton rats elicited binding and neutralizing antibody activities, as well as interferon-gamma-producing T cells. Vaccination of cotton rats resulted in protection against subsequent challenges with either homologous or heterologous hPIV-3. Furthermore, vaccination of cotton rats with a mixture of rSeV-hPIV3-HN and a previously described recombinant SeV expressing the F protein of RSV resulted in protection against three different challenge viruses: hPIV-3, hPIV-1 and RSV. Results encourage the continued development of the candidate recombinant SeV vaccines to combat serious respiratory infections of children.


Assuntos
Proteína HN/imunologia , Vacinas contra Parainfluenza/imunologia , Infecções por Vírus Respiratório Sincicial/prevenção & controle , Vacinas contra Vírus Sincicial Respiratório/imunologia , Infecções por Respirovirus/prevenção & controle , Vírus Sendai/genética , Proteínas Virais de Fusão/imunologia , Sequência de Aminoácidos , Animais , Anticorpos Antivirais/sangue , Feminino , Vetores Genéticos , Proteína HN/genética , Pulmão/virologia , Dados de Sequência Molecular , Vacinas contra Parainfluenza/genética , Ratos , Vacinas contra Vírus Sincicial Respiratório/genética , Sigmodontinae , Linfócitos T/imunologia , Vacinas Sintéticas/genética , Vacinas Sintéticas/imunologia , Proteínas Virais de Fusão/genética
13.
Vaccine ; 25(52): 8782-93, 2007 Dec 17.
Artigo em Inglês | MEDLINE | ID: mdl-18037543

RESUMO

The respiratory syncytial virus (RSV) is a serious pediatric pathogen for which there is currently no clinically approved vaccine. This report describes the design and testing of a new RSV vaccine construct (rSV-RSV-F), created by the recombination of an RSV F sequence with the murine parainfluenza virus-type 1 (Sendai virus, SV) genome. SV was selected as the vaccine backbone for this study, because it has previously been shown to elicit high-magnitude, durable immune activities in animal studies and has advanced to human safety trials as a xenogenic vaccine for human parainfluenza virus-type 1 (hPIV-1). Cells infected with the recombinant SV expressed RSV F protein, but F was not incorporated into progeny SV virions. When cotton rats were inoculated with the vaccine, high-titer RSV-binding and neutralizing antibodies as well as interferon-gamma-producing T-cells were induced. Most striking was the protection against intra-nasal RSV challenge conferred by the vaccine. The rSV-RSV-F construct was also tested as a mixture with a second SV construct expressing the RSV G protein, but no clear advantage was demonstrated by combining the two vaccines. As a final analysis, the efficacy of the rSV-RSV-F vaccine was tested against an array of RSV isolates. Results showed that neutralizing and protective responses were effective against RSV isolates of both A and B subtypes. Together, experimental results encourage promotion of this recombinant SV construct as a vaccine candidate for the prevention of RSV in humans.


Assuntos
Infecções por Vírus Respiratório Sincicial/prevenção & controle , Vírus Sinciciais Respiratórios/imunologia , Vírus Sendai/imunologia , Proteínas Virais de Fusão/imunologia , Vacinas Virais/imunologia , Sequência de Aminoácidos , Animais , Anticorpos Antivirais/sangue , Ensaio de Imunoadsorção Enzimática , Feminino , Humanos , Interferon gama/biossíntese , Pulmão/patologia , Pulmão/virologia , Dados de Sequência Molecular , Testes de Neutralização , Infecções por Vírus Respiratório Sincicial/imunologia , Infecções por Vírus Respiratório Sincicial/patologia , Vírus Sinciciais Respiratórios/genética , Vírus Sendai/genética , Sigmodontinae , Linfócitos T/imunologia , Vacinas Sintéticas/genética , Vacinas Sintéticas/imunologia , Proteínas do Envelope Viral/genética , Proteínas do Envelope Viral/imunologia , Proteínas Virais de Fusão/genética , Ensaio de Placa Viral , Vacinas Virais/genética
14.
J Virol ; 80(11): 5145-55, 2006 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-16698995

RESUMO

Newcastle disease virus (NDV) is a negative-strand RNA virus with oncolytic activity against human tumors. Its effectiveness against tumors and safety in normal tissue have been demonstrated in several clinical studies. Here we show that the spread of NDV infection is drastically different in normal cell lines than in tumor cell lines and that the two cell types respond differently to beta interferon (IFN-beta) treatment. NDV rapidly replicated and killed HT-1080 human fibrosarcoma cells but spread poorly in CCD-1122Sk human skin fibroblast cells. Pretreatment with endogenous or exogenous IFN-beta completely inhibited NDV replication in normal cells but had little or no effect in tumor cells. Thus, the outcome of NDV infection appeared to depend on the response of uninfected cells to IFN-beta. To investigate their differences in IFN responsiveness, we analyzed and compared the expression and activation of components of the IFN signal transduction pathway in these two types of cells. The levels of phosphorylated STAT1 and STAT2 and that of the ISGF3 complex were markedly reduced in IFN-beta-treated tumor cells. Moreover, cDNA microarray analysis revealed significantly fewer IFN-regulated genes in the HT-1080 cells than in the CDD-1122Sk cells. This finding suggests that tumor cells demonstrate a less-than-optimum antiviral response because of a lesion in their IFN signal transduction pathway. The rapid spread of NDV in HT-1080 cells appears to be caused by their deficient expression of anti-NDV proteins upon exposure to IFN-beta.


Assuntos
Interferons/biossíntese , Doença de Newcastle/fisiopatologia , Vírus da Doença de Newcastle/fisiologia , Proteínas Virais/fisiologia , Animais , Linhagem Celular , Regulação Viral da Expressão Gênica , Interferons/imunologia , Células Tumorais Cultivadas/metabolismo , Células Tumorais Cultivadas/virologia , Proteínas Virais/genética
15.
Antimicrob Agents Chemother ; 48(5): 1495-502, 2004 May.
Artigo em Inglês | MEDLINE | ID: mdl-15105096

RESUMO

Human parainfluenza viruses are important respiratory tract pathogens, especially of children. However, no vaccines or specific therapies for infections caused by these viruses are currently available. In the present study we characterized the efficacy of the novel parainfluenza virus inhibitors BCX 2798 and BCX 2855, which were designed based on the three-dimensional structure of the hemagglutinin-neuraminidase (HN) protein. The compounds were highly effective in inhibiting hemagglutinin (HA) and neuraminidase (NA) activities and the growth of hPIV-1, hPIV-2, and hPIV-3 in LLC-MK(2) cells. The concentrations required to reduce the activity to 50% of that of a control ranged from 0.1 to 6.0 micro M in HA inhibition assays and from 0.02 to 20 micro M in NA inhibition assays. The concentrations required to inhibit virus replication to 50% of the level of the control ranged from 0.7 to 11.5 micro M. BCX 2798 and BCX 2855 were inactive against influenza virus HA and NA and bacterial NA. In mice infected with a recombinant Sendai virus whose HN gene was replaced with that of hPIV-1 [rSV(hHN)], intranasal administration of BCX 2798 (10 mg/kg per day) and of BCX 2855 (50 mg/kg per day) 4 h before the start of infection resulted in a significant reduction in titers of virus in the lungs and protection from death. Treatment beginning 24 h after the start of infection did not prevent death. Together, our results indicate that BCX 2798 and BCX 2855 are effective inhibitors of parainfluenza virus HN and may limit parainfluenza virus infections in humans.


Assuntos
Antivirais/farmacologia , Azidas/farmacologia , Inibidores Enzimáticos/farmacologia , Hemaglutininas/efeitos dos fármacos , Ácidos Hexurônicos/farmacologia , Neuraminidase/antagonistas & inibidores , Vírus da Parainfluenza 1 Humana/efeitos dos fármacos , Vírus da Parainfluenza 2 Humana/efeitos dos fármacos , Vírus da Parainfluenza 3 Humana/efeitos dos fármacos , Sulfonamidas/farmacologia , Animais , Feminino , Humanos , Células LLC-PK1 , Pulmão/patologia , Pulmão/virologia , Camundongos , Infecções por Paramyxoviridae/tratamento farmacológico , Infecções por Paramyxoviridae/patologia , Infecções por Paramyxoviridae/virologia , Proteínas Recombinantes , Vírus Sendai/efeitos dos fármacos , Vírus Sendai/genética , Suínos , Proteínas Virais de Fusão/efeitos dos fármacos , Proteínas Virais de Fusão/genética , Replicação Viral/efeitos dos fármacos
16.
J Virol ; 76(24): 13028-33, 2002 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-12438628

RESUMO

Paramyxovirus infects cells by initially attaching to a sialic acid-containing cellular receptor and subsequently fusing with the plasma membrane of the cells. Hemagglutinin-neuraminidase (HN) protein, which is responsible for virus attachment, interacts with the fusion protein in a virus type-specific manner to induce efficient membrane fusion. To elucidate the mechanism of HN-promoted membrane fusion, we characterized a series of Newcastle disease virus HN proteins whose surface residues were mutated. Fusion promotion activity was substantially altered in only the HN proteins with a mutation in the first or sixth beta sheet. These regions overlap the large hydrophobic surface of HN; thus, the hydrophobic surface may contain the fusion promotion domain. Furthermore, a comparison of the HN structure crystallized alone or in complex with 2-deoxy-2,3-dehydro-N-acetylneuraminic acid revealed substantial conformational changes in several loops within or near the hydrophobic surface. Our results suggest that the binding of HN protein to the receptor induces the conformational change of residues near the hydrophobic surface of HN protein and that this change triggers the activation of the F protein, which initiates membrane fusion.


Assuntos
Proteína HN/fisiologia , Fusão de Membrana/fisiologia , Vírus da Doença de Newcastle/fisiologia , Dimerização , Proteína HN/química , Células HeLa , Humanos , Conformação Proteica , Proteínas Virais de Fusão/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA