Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Emerg Microbes Infect ; 12(1): 2192821, 2023 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-36927227

RESUMO

Influenza and Respiratory Syncytial virus (RSV) infections together contribute significantly to the burden of acute lower respiratory tract infections. Despite the disease burden, no approved RSV vaccine is available. While approved vaccines are available for influenza, seasonal vaccination is required to maintain protection. In addition to both being respiratory viruses, they follow a common seasonality, which warrants the necessity for a concerted vaccination approach. Here, we designed bivalent vaccines by utilizing highly conserved sequences, targeting both influenza A and RSV, as either a chimeric antigen or individual antigens separated by a ribosome skipping sequence. These vaccines were found to be effective in protecting the animals from challenge by either virus, with mechanisms of protection being substantially interrogated in this communication.


Assuntos
Vacinas contra Influenza , Influenza Humana , Infecções por Vírus Respiratório Sincicial , Camundongos , Animais , Humanos , Vírus Sinciciais Respiratórios/genética , Vacinas Combinadas , Anticorpos Antivirais , Infecções por Vírus Respiratório Sincicial/prevenção & controle , Vacinas contra Influenza/genética , Anticorpos Neutralizantes
2.
iScience ; 24(11): 103328, 2021 Nov 19.
Artigo em Inglês | MEDLINE | ID: mdl-34805790

RESUMO

A quarter of all seasonal influenza cases are caused by type B influenza virus (IBV) that also dominates periodically. Here, we investigated a recombinant adenovirus vaccine carrying a synthetic HA2 representing the consensus sequence of all IBV hemagglutinins. The vaccine fully protected mice from lethal challenges by IBV of both genetic lineages, demonstrating its breadth of protection. The protection was not mediated by neutralizing antibodies but robust antibody-dependent cellular cytotoxicity and cell-mediated immune responses. Complete protection of the animals required the entire codon-optimized HA2 sequence that elicited a balanced immune response, whereas truncated vaccines without either the fusion peptide or the transmembrane domain reduced the efficacy of protection. Finally, the vaccines did not demonstrate any sign of disease exacerbation following lung pathology and morbidity monitoring. Collectively, these data suggest that it could be worth further exploring this prototype universal vaccine because of its considerable efficacy, safety, and breadth of protection.

3.
Front Immunol ; 12: 747866, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34603336

RESUMO

Respiratory syncytial virus (RSV) is a leading cause of respiratory infections worldwide and disease management measures are hampered by the lack of a safe and effective vaccine against the infection. We constructed a novel recombinant RSV vaccine candidate based on a deletion mutant vaccinia virus platform, in that the host range genes E3L and K3L were deleted (designated as VACVΔE3LΔK3L) and a poxvirus K3L ortholog gene was used as a marker for the rapid and efficient selection of recombinant viruses. The safety of the modified vaccinia virus was investigated by intranasal administration of BALB/c mice with the modified vaccinia vector using a dose known to be lethal in the wild-type Western Reserve. Only a minor loss of body weight by less than 5% and mild pulmonary inflammation were observed, both of which were transient in nature following nasal administration of the high-dose modified vaccinia virus. In addition, the viruses were cleared from the lung in 2 days with no viral invasions of the brain and other vital organs. These results suggest that the virulence of the virus has been essentially abolished. We then investigated the efficiency of the vector for the delivery of vaccines against RSV through comparison with another RSV vaccine delivered by the widely used Modified Vaccinia virus Ankara (MVA) backbone. In the cotton rats, we found a single intramuscular administration of VACVΔE3LΔK3L-vectored vaccine elicited immune responses and protection at a level comparable to the MVA-vectored vaccine against RSV infection. The distinct features of this novel VACV vector, such as an E3L deletion for attenuation and a K3L ortholog for positive selection and high efficiency for vaccine delivery, could provide unique advantages to the application of VACV as a platform for vaccine development.


Assuntos
Infecções por Vírus Respiratório Sincicial/prevenção & controle , Vacinas contra Vírus Sincicial Respiratório/imunologia , Vacinas Sintéticas/imunologia , Animais , Feminino , Vetores Genéticos , Camundongos , Camundongos Endogâmicos BALB C , Vírus Sinciciais Respiratórios , Sigmodontinae , Desenvolvimento de Vacinas , Proteínas Virais de Fusão/imunologia
4.
Front Immunol ; 12: 785349, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-35095861

RESUMO

SARS-CoV-2 infections present a tremendous threat to public health. Safe and efficacious vaccines are the most effective means in preventing the infections. A variety of vaccines have demonstrated excellent efficacy and safety around the globe. Yet, development of alternative forms of vaccines remains beneficial, particularly those with simpler production processes, less stringent storage conditions, and the capability of being used in heterologous prime/boost regimens which have shown improved efficacy against many diseases. Here we reported a novel DNA vaccine comprised of the SARS-CoV-2 spike protein fused with CD40 ligand (CD40L) serving as both a targeting ligand and molecular adjuvant. A single intramuscular injection in Syrian hamsters induced significant neutralizing antibodies 3-weeks after vaccination, with a boost substantially improving immune responses. Moreover, the vaccine also reduced weight loss and suppressed viral replication in the lungs and nasal turbinates of challenged animals. Finally, the incorporation of CD40L into the DNA vaccine was shown to reduce lung pathology more effectively than the DNA vaccine devoid of CD40L. These results collectively indicate that this DNA vaccine candidate could be further explored because of its efficacy and known safety profile.


Assuntos
Ligante de CD40/imunologia , COVID-19/imunologia , Mesocricetus/imunologia , SARS-CoV-2/imunologia , Glicoproteína da Espícula de Coronavírus/imunologia , Vacinas de DNA/imunologia , Adjuvantes Imunológicos/farmacologia , Animais , Anticorpos Neutralizantes/imunologia , Anticorpos Antivirais/imunologia , COVID-19/virologia , Linhagem Celular , Feminino , Células HEK293 , Humanos , Pulmão/imunologia , Pulmão/virologia , Mesocricetus/virologia , Modelos Animais , Vacinação/métodos , Vacinas de Produtos Inativados/imunologia
5.
Virology ; 541: 101-112, 2020 02.
Artigo em Inglês | MEDLINE | ID: mdl-32056708

RESUMO

Protein kinase R plays a key role in innate antiviral immune responses of vertebrate animals. Most mammalian poxviruses encode two PKR antagonists, E3 (dsRNA binding) and K3 (eIF2α homolog) proteins. In this study, the role of K3 family proteins from poxviruses with distinct host tropisms in determining the virus host range was examined in a vaccinia E3L deletion mutant virus. It was found that K3 orthologs from the species-specific poxviruses (taterapox virus, sheeppox virus, myxoma virus, swinepox virus and yaba monkey tumor virus) restored the virus replication competency in cells derived from their natural hosts or related animal species. Further, it was found that the residues located in the helix insert region of the protein, K45 of vaccinia K3 and Y47 of the sheep poxvirus ortholog 011, are critical for the virus host species specificity. These observations demonstrate that poxvirus K3 proteins are major determinants of the virus host specificity.


Assuntos
Fator de Iniciação 2 em Eucariotos/fisiologia , Especificidade de Hospedeiro , Poxviridae/fisiologia , Proteínas Virais/fisiologia , Animais , Células Cultivadas , Humanos , Fosforilação , Especificidade da Espécie , Tropismo Viral , Replicação Viral
6.
PLoS One ; 13(7): e0199067, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30052641

RESUMO

Cotton rats are an important animal model to study infectious diseases. They have demonstrated higher susceptibility to a wider variety of human pathogens than other rodents and are also the animal model of choice for pre-clinical evaluations of some vaccine candidates. However, the genome of cotton rats remains to be fully sequenced, with much fewer genes cloned and characterised compared to other rodent species. Here we report the cloning and characterization of CD40 ligand, whose human and murine counterparts are known to be expressed on a range of cell types including activated T cells and B cells, dendritic cells, granulocytes, macrophages and platelets and exerts a broad array of immune responses. The cDNA for cotton rat CD40L we isolated is comprised of 1104 nucleotides with an open reading frame (ORF) of 783bp coding for a 260 amino acid protein. The recombinant cotton rat CD40L protein was recognized by an antibody against mouse CD40L. Moreover, it demonstrated functional activities on immature bone marrow dendritic cells by upregulating surface maturation markers (CD40, CD54, CD80, and CD86), and increasing IL-6 gene and protein expression. The availability of CD40L gene identity could greatly facilitate mechanistic research on pathogen-induced-immunopathogenesis and vaccine-elicited immune responses.


Assuntos
Ligante de CD40/química , Ligante de CD40/farmacologia , Células Dendríticas/efeitos dos fármacos , Sigmodontinae/imunologia , Sequência de Aminoácidos , Animais , Antígenos CD/genética , Antígenos CD/imunologia , Linfócitos B/citologia , Linfócitos B/imunologia , Sequência de Bases , Plaquetas/citologia , Plaquetas/imunologia , Células da Medula Óssea/citologia , Células da Medula Óssea/imunologia , Ligante de CD40/genética , Ligante de CD40/imunologia , Clonagem Molecular , Células Dendríticas/citologia , Células Dendríticas/imunologia , Expressão Gênica , Vetores Genéticos/química , Vetores Genéticos/metabolismo , Granulócitos/citologia , Granulócitos/imunologia , Células HeLa , Humanos , Interleucina-6/genética , Interleucina-6/imunologia , Macrófagos/citologia , Macrófagos/imunologia , Mesocricetus , Camundongos , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/imunologia , Proteínas Recombinantes/farmacologia , Alinhamento de Sequência , Homologia de Sequência de Aminoácidos , Linfócitos T/citologia , Linfócitos T/imunologia
7.
J Virol ; 85(23): 12280-91, 2011 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-21917954

RESUMO

Poxviruses are important human and animal pathogens that have evolved elaborate strategies for antagonizing host innate and adaptive immunity. The E3 protein of vaccinia virus, the prototypic member of the orthopoxviruses, functions as an inhibitor of innate immune signaling and is essential for vaccinia virus replication in vivo and in many human cell culture systems. However, the function of orthologues of E3 expressed by poxviruses of other genera with different host specificity remains largely unknown. In the present study, we characterized the E3 orthologues from sheeppox virus, yaba monkey tumor virus, swinepox virus, and myxoma virus for their ability to modulate protein kinase R (PKR) function, cytokine responses and virus pathogenicity. We found that the E3 orthologues of myxoma virus and swinepox virus could suppress PKR activation and interferon (IFN)-induced antiviral activities and restore the host range function of E3 in HeLa cells. In contrast, the E3 orthologues from sheeppox virus and yaba monkey tumor virus were unable to inhibit PKR activation. While the sheeppox orthologue was unable to restore the host range function of E3, the yaba monkey tumor virus orthologue partially restored E3-deficient vaccinia virus replication in HeLa cells, correlated with its ability to suppress IFN-induced antiviral activities. Moreover, poxvirus E3 orthologues show varying ability to inhibit the induction of antiviral and proinflammatory cytokines. Despite these in vitro results, none of the E3 orthologues tested was capable of restoring pathogenicity to E3-deficient vaccinia virus in vivo.


Assuntos
Citocinas/metabolismo , Proteínas de Ligação a RNA/metabolismo , Vaccinia virus/metabolismo , Vaccinia virus/patogenicidade , Vacínia/imunologia , Vacínia/virologia , Proteínas Virais/metabolismo , eIF-2 Quinase/metabolismo , Sequência de Aminoácidos , Animais , Western Blotting , Cricetinae , Citocinas/genética , Feminino , Células HeLa , Humanos , Imunoprecipitação , Camundongos , Camundongos Endogâmicos BALB C , Dados de Sequência Molecular , Fosforilação , RNA Mensageiro/genética , RNA Interferente Pequeno/genética , Proteínas de Ligação a RNA/genética , Reação em Cadeia da Polimerase em Tempo Real , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Homologia de Sequência de Aminoácidos , Transdução de Sinais , Vacínia/metabolismo , Vaccinia virus/genética , Proteínas Virais/genética , Replicação Viral , eIF-2 Quinase/antagonistas & inibidores , eIF-2 Quinase/genética
8.
Virology ; 413(2): 183-93, 2011 May 10.
Artigo em Inglês | MEDLINE | ID: mdl-21354589

RESUMO

RNA species produced during virus replication are pathogen-associated molecular patterns (PAMPs) triggering cellular innate immune responses including induction of type I interferon expression and apoptosis. Pattern recognition receptors (PRRs) for these RNAs include the retinoic acid-inducible gene I (RIG-I) like receptors (RLRs) RIG-I and melanoma differentiation associated gene 5 (MDA5) and the dsRNA dependent protein kinase (PKR). Currently, poxvirus PAMPs and their associated PRRs are not well characterized. We report that RNA species generated in vaccinia infected cells can activate MDA5 or RIG-I dependent interferon-ß (IFN-ß) gene transcription in a cell type-specific manner. These RNA species also induce the activation of apoptosis in a PKR dependent, but MDA5 and RIG-I independent, manner. Collectively our results demonstrate that RNA species generated during vaccinia virus replication are major PAMPs activating apoptosis and IFN-ß gene transcription. Moreover, our results delineate the signaling pathways involved in the recognition of RNA-based poxvirus PAMPs.


Assuntos
RNA Helicases DEAD-box/metabolismo , Interferon beta/metabolismo , Proteínas Quinases/metabolismo , RNA Viral/genética , Vaccinia virus/metabolismo , Apoptose/fisiologia , Citocinas/genética , Citocinas/metabolismo , Proteína DEAD-box 58 , RNA Helicases DEAD-box/genética , Regulação da Expressão Gênica/fisiologia , Células HeLa , Humanos , Imunidade Inata , Helicase IFIH1 Induzida por Interferon , Interferon beta/genética , Proteínas Quinases/genética , Receptores Imunológicos , Vaccinia virus/genética
9.
Mol Cell Biochem ; 333(1-2): 221-32, 2010 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-19652917

RESUMO

The T:G mismatch specific DNA glycosylase (TDG) is known as an important enzyme in repairing damaged DNA. Recent studies also showed that TDG interacts with a p160 protein, steroid receptor coactivator 1 or nuclear receptor coactivator 1 (SRC1), and is involved in transcriptional activation of the estrogen receptor. However, whether other members of the p160 family are also involved in TDG-interaction and signal transduction regulation remains to be seen. In this study, we employed the mammalian two-hybrid system to investigate the interaction between TDG and another member of the p160 family, nuclear receptor coactivator 3 (NCoA-3). We found that a DXXD motif from aa 294-297 within TDG was responsible for the TDG-NCoA-3 interaction, we also found that a LLXXXL motif (X means any amino acid) from aa 1029-1037 (LLRNSL) and a merged LLXXL motif (LLDQLHTLL) from aa 1053-1061 in NCoA-3 were important for the TDG-NCoA-3 interactions. Mutation of the two aspartic acids (aa 294 and 297) into two alanines in TDG significantly affected the interaction and subsequent transcriptional activation of several steroid hormone receptors including, estrogen-, androgen- and progesterone- receptors in Huh7 cells. We also identified that mutations of NCoA-3 at either leucines 1029-1030 or 1053-1054 (replaced by alanines) also reduced the interaction activity between TDG and NCoA1. These data indicated that the TDG-NCoA-3 interaction is important for broad range activation of steroid hormone nuclear receptors, and may also contribute significantly to further understanding of TDG-related nuclear receptor regulation.


Assuntos
Coativador 3 de Receptor Nuclear/metabolismo , Receptores Citoplasmáticos e Nucleares/genética , Timina DNA Glicosilase/metabolismo , Ativação Transcricional , Motivos de Aminoácidos , Linhagem Celular Tumoral , Humanos , Mutagênese Sítio-Dirigida , Coativador 3 de Receptor Nuclear/fisiologia , Mapeamento de Interação de Proteínas , Timina DNA Glicosilase/fisiologia , Técnicas do Sistema de Duplo-Híbrido
10.
J Virol ; 83(20): 10627-36, 2009 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-19656868

RESUMO

Cellular tropism of vaccinia virus (VACV) is regulated by host range genes, including K1L, C7L, and E3L. While E3L is known to support viral replication by antagonizing interferon (IFN) effectors, including PKR, the exact functions of K1L and C7L are unclear. Here, we show that K1L and C7L can also inhibit antiviral effectors induced by type I IFN. In human Huh7 and MCF-7 cells, a VACV mutant lacking both K1L and C7L (vK1L-C7L-) replicated as efficiently as wild-type (WT) VACV, even in the presence of IFN. However, pretreating the cells with type I IFN, while having very little effect on WT VACV, blocked the replication of vK1L-C7L- at the step of intermediate viral gene translation. Restoring either K1L or C7L to vK1L(-)C7L(-) fully restored the IFN resistance phenotype. The deletion of K1L and C7L from VACV did not affect the ability of the virus to inhibit IFN signaling or its ability to inhibit the phosphorylation of PKR and the alpha subunit of eukaryotic initiation factor 2, indicating that K1L and C7L function by antagonizing an IFN effector(s) but with a mechanism that is different from those of IFN antagonists previously identified for VACV. Mutations of K1L that inactivate the host range function also rendered K1L unable to antagonize IFN, suggesting that K1L supports VACV replication in mammalian cells by antagonizing the same antiviral factor(s) that is induced by IFN in Huh7 cells.


Assuntos
Antivirais , Interferon Tipo I/antagonistas & inibidores , Vaccinia virus/metabolismo , Proteínas Virais/efeitos dos fármacos , Animais , Antivirais/metabolismo , Antivirais/farmacologia , Linhagem Celular , Linhagem Celular Tumoral , Cricetinae , Humanos , Interferon Tipo I/imunologia , Vaccinia virus/efeitos dos fármacos , Vaccinia virus/genética , Proteínas Virais/genética , Proteínas Virais/metabolismo , Replicação Viral
11.
J Virol ; 83(13): 6757-68, 2009 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-19369349

RESUMO

The vaccinia virus double-stranded RNA binding protein E3 has been demonstrated to inhibit the expression of cytokines, including beta interferon (IFN-beta) and tumor necrosis factor alpha (TNF-alpha). However, few details regarding the molecular mechanisms of this inhibition have been described. Using real-time PCR arrays, we found that E3 suppressed the induction of a diverse array of cytokines representing members of the IFN, interleukin (IL), TNF, and transforming growth factor cytokine families. We discovered that the factor(s) responsible for the induction of IL-6, TNF-alpha, and inhibin beta A (INHBA) was associated with the early and late phases of virus infection. In contrast, the factor(s) which regulates IFN-beta induction was associated with the late phase of replication. We have found that expression of these cytokines can be induced by transfection of cells with RNA isolated from vaccinia virus-infected cells. Moreover, we provide evidence that E3 antagonizes both PKR-dependent and PKR-independent pathways to regulate cytokine expression. PKR-dependent activation of p38 and NF-kappaB was required for vaccinia virus-induced INHBA expression, whereas induction of TNF-alpha required only PKR-dependent NF-kappaB activation. In contrast, induction of IL-6 and IFN-beta was largely PKR independent. IL-6 induction is regulated by NF-kappaB, while IFN-beta induction is mediated by IFN-beta promoter stimulator 1 and IFN regulatory factor 3/NF-kappaB. Collectively, these results indicate that E3 suppresses distinct but interlinked host signaling pathways to inhibit the expression of a diverse array of cytokines.


Assuntos
Fator Regulador 3 de Interferon/metabolismo , NF-kappa B/metabolismo , Proteínas de Ligação a RNA/metabolismo , Vaccinia virus/metabolismo , Proteínas Virais/metabolismo , eIF-2 Quinase/metabolismo , Animais , Cricetinae , Regulação da Expressão Gênica , Células HeLa , Humanos , Subunidades beta de Inibinas/metabolismo , Interleucina-6/metabolismo , Fosforilação , Transdução de Sinais , Transcrição Gênica , Fator de Necrose Tumoral alfa/metabolismo , Vaccinia virus/genética , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
12.
PLoS Pathog ; 4(7): e1000099, 2008 Jul 11.
Artigo em Inglês | MEDLINE | ID: mdl-18617992

RESUMO

The sensing of pathogen infection and subsequent triggering of innate immunity are key to controlling zoonotic infections. Myxoma virus (MV) is a cytoplasmic DNA poxvirus that in nature infects only rabbits. Our previous studies have shown that MV infection of primary mouse cells is restricted by virus-induced type I interferon (IFN). However, little is known about the innate sensor(s) involved in activating signaling pathways leading to cellular defense responses in primary human immune cells. Here, we show that the complete restriction of MV infection in the primary human fibroblasts requires both tumor necrosis factor (TNF) and type I IFN. We also demonstrate that MV infection of primary human macrophages (pHMs) activates the cytoplasmic RNA sensor called retinoic acid inducible gene I (RIG-I), which coordinately induces the production of both TNF and type I IFN. Of note, RIG-I sensing of MV infection in pHMs initiates a sustained TNF induction through the sequential involvement of the downstream IFN-regulatory factors 3 and 7 (IRF3 and IRF7). Thus, RIG-I-mediated co-induction of TNF and type I IFN by virus-infected pHMs represents a novel innate defense mechanism to restrict viral infection in human cells. These results also reveal a new regulatory mechanism for TNF induction following viral infection.


Assuntos
RNA Helicases DEAD-box/metabolismo , Interferon Tipo I/biossíntese , Macrófagos/virologia , Myxoma virus/fisiologia , Fator de Necrose Tumoral alfa/biossíntese , Células Cultivadas , Proteína DEAD-box 58 , RNA Helicases DEAD-box/biossíntese , RNA Helicases DEAD-box/genética , Combinação de Medicamentos , Fibroblastos/metabolismo , Fibroblastos/virologia , Regulação Viral da Expressão Gênica , Inativação Gênica , Interações Hospedeiro-Patógeno , Humanos , Fator Regulador 3 de Interferon/metabolismo , Fator Regulador 7 de Interferon/metabolismo , Interferon Tipo I/genética , Helicase IFIH1 Induzida por Interferon , Interferon beta/farmacologia , Linfócitos/metabolismo , Linfócitos/virologia , Macrófagos/metabolismo , RNA Interferente Pequeno/farmacologia , Receptores Imunológicos , Fator de Necrose Tumoral alfa/genética , Fator de Necrose Tumoral alfa/farmacologia
13.
Virology ; 377(1): 124-32, 2008 Jul 20.
Artigo em Inglês | MEDLINE | ID: mdl-18502465

RESUMO

The E3L protein of vaccinia virus (VV) is well known for its capacity to evade cellular innate antiviral immunity related to interferon (IFN), for example PKR and RNaseL mediated antiviral activities. However, due to the limited range of cells that support VV E3L deletion mutant replication, the full capacity of E3L inhibiting the innate immune response induced by IFNs remains to be examined. In this report, the inhibition activity of VV E3L against a wide spectrum of human IFNs, including type I IFNs (12 IFN-alpha subtypes, IFN-beta, and IFN-omega), and type II IFN (gamma), was comparatively examined using the Copenhagen strain E3L deletion mutant and its revertant control virus in a human hepatoma cell line, Huh7. Deletion of the E3L open reading frame rendered the mutant VV sensitive to all types of IFNs, while the revertant VV was strongly resistant to these treatments. Furthermore, we show that the inhibition of VV E3L deletion mutant by IFN occurs at the stage of intermediate gene translation, while the expression of early genes and transcription of intermediate genes are largely unaffected. Using specific siRNAs to suppress the classical IFN-induced antiviral pathways, we found that PKR is the key factor modulated by E3L, while the RNaseL and MxA pathways play limited roles in this Huh7 cell system. Thus, our data demonstrates that VV E3L can mediate strong inhibition activity against all human type I and type II IFNs, mainly through modulation of the PKR pathway in Huh7 cells.


Assuntos
Interferons/antagonistas & inibidores , Proteínas de Ligação a RNA/fisiologia , Proteínas de Ligação a RNA/toxicidade , Vaccinia virus/fisiologia , Vaccinia virus/patogenicidade , Proteínas Virais/fisiologia , Proteínas Virais/toxicidade , Sequência de Bases , Linhagem Celular , Primers do DNA/genética , DNA Viral/genética , Endorribonucleases/fisiologia , Proteínas de Ligação ao GTP/antagonistas & inibidores , Proteínas de Ligação ao GTP/genética , Proteínas de Ligação ao GTP/fisiologia , Deleção de Genes , Genes Virais , Humanos , Imunidade Inata , Proteínas de Resistência a Myxovirus , Biossíntese de Proteínas , RNA Interferente Pequeno/genética , Proteínas de Ligação a RNA/genética , Proteínas de Ligação a RNA/imunologia , Transdução de Sinais/efeitos dos fármacos , Vaccinia virus/genética , Vaccinia virus/imunologia , Proteínas Virais/genética , Proteínas Virais/imunologia , Replicação Viral , eIF-2 Quinase/fisiologia
14.
J Virol ; 81(6): 3027-32, 2007 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-17192307

RESUMO

Aurintricarboxylic acid (ATA) has been shown to inhibit the replication of viruses from several different families, including human immunodeficiency virus, vesicular stomatitis virus, and the coronavirus causing severe acute respiratory syndrome. This study characterizes the inhibitory effect of ATA on vaccinia virus replication in HeLa, Huh7, and AD293 cells. Vaccinia virus replication is significantly abrogated upon ATA treatment, which is associated with the inhibition of early viral gene transcription. This inhibitory effect may be attributed to two findings. First, ATA blocks the phosphorylation of extracellular signal-regulated kinase 1/2, an event shown to be essential for vaccinia virus replication. Second, ATA inhibits the phosphatase activity of the viral enzyme H1L, which is required to initiate viral transcription. Thus, ATA inhibits vaccinia virus replication by targeting both cellular and viral factors essential for the early stage of replication.


Assuntos
Antivirais/farmacologia , Ácido Aurintricarboxílico/farmacologia , Fosfoproteínas Fosfatases/antagonistas & inibidores , Vaccinia virus/efeitos dos fármacos , Replicação Viral/efeitos dos fármacos , Carcinoma Hepatocelular/patologia , Linhagem Celular , Linhagem Celular Tumoral , Relação Dose-Resposta a Droga , Ativação Enzimática/efeitos dos fármacos , Células HeLa , Humanos , Concentração Inibidora 50 , Neoplasias Hepáticas/patologia , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , Fosfoproteínas Fosfatases/genética , Fosfoproteínas Fosfatases/metabolismo , Fosforilação/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos , Transcrição Gênica/efeitos dos fármacos , Transfecção , Vaccinia virus/genética , Vaccinia virus/fisiologia , Ensaio de Placa Viral
15.
Vaccine ; 23(17-18): 2273-9, 2005 Mar 18.
Artigo em Inglês | MEDLINE | ID: mdl-15755610

RESUMO

Severe acute respiratory syndrome (SARS) caused by a newly identified coronavirus (SARS-CoV) remains a threat to cause epidemics as evidenced by recent sporadic cases in China. In this communication, we evaluated the efficacy and safety of two SARS vaccine candidates based on the recombinant modified vaccinia Ankara (MVA) expressing SARS-CoV spike or nucleocapsid proteins in ferrets. No clinical signs were observed in all the ferrets challenged with SARS-CoV. On the other hand, vaccination did not prevent SARS-CoV infection in ferrets. In contrast, immunized ferrets (particularly those immunized with rMVA-spike) exhibited significantly stronger inflammatory responses and focal necrosis in liver tissue after SARS-CoV challenge than control animals. Thus, our data suggest that enhanced hepatitis is linked to vaccination with rMVA expressing SARS-CoV antigens.


Assuntos
Coronavírus Relacionado à Síndrome Respiratória Aguda Grave/imunologia , Vacinas Virais/farmacologia , Animais , Antígenos Virais/genética , Sequência de Bases , Proteínas do Nucleocapsídeo de Coronavírus , DNA Viral/genética , Furões , Hepatite Viral Animal/patologia , Hepatite Viral Animal/prevenção & controle , Masculino , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/imunologia , Proteínas do Nucleocapsídeo/genética , Proteínas do Nucleocapsídeo/imunologia , RNA Viral/análise , RNA Viral/genética , Coronavírus Relacionado à Síndrome Respiratória Aguda Grave/genética , Coronavírus Relacionado à Síndrome Respiratória Aguda Grave/isolamento & purificação , Síndrome Respiratória Aguda Grave/patologia , Síndrome Respiratória Aguda Grave/prevenção & controle , Glicoproteína da Espícula de Coronavírus , Vacinas Sintéticas/genética , Vacinas Sintéticas/farmacologia , Proteínas do Envelope Viral/genética , Proteínas do Envelope Viral/imunologia , Vacinas Virais/genética
16.
J Virol ; 78(22): 12672-6, 2004 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-15507655

RESUMO

Severe acute respiratory syndrome (SARS) caused by a newly identified coronavirus (SARS-CoV) is a serious emerging human infectious disease. In this report, we immunized ferrets (Mustela putorius furo) with recombinant modified vaccinia virus Ankara (rMVA) expressing the SARS-CoV spike (S) protein. Immunized ferrets developed a more rapid and vigorous neutralizing antibody response than control animals after challenge with SARS-CoV; however, they also exhibited strong inflammatory responses in liver tissue. Inflammation in control animals exposed to SARS-CoV was relatively mild. Thus, our data suggest that vaccination with rMVA expressing SARS-CoV S protein is associated with enhanced hepatitis.


Assuntos
Hepatite/etiologia , Glicoproteínas de Membrana/imunologia , Coronavírus Relacionado à Síndrome Respiratória Aguda Grave/imunologia , Vacinas Sintéticas/efeitos adversos , Vaccinia virus/imunologia , Proteínas do Envelope Viral/imunologia , Vacinas Virais/efeitos adversos , Animais , Anticorpos Antivirais/sangue , Furões , Fígado/patologia , Masculino , Glicoproteínas de Membrana/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Glicoproteína da Espícula de Coronavírus , Vacinação , Proteínas do Envelope Viral/genética
17.
Biochem Biophys Res Commun ; 311(4): 870-6, 2003 Nov 28.
Artigo em Inglês | MEDLINE | ID: mdl-14623261

RESUMO

In March 2003, a novel coronavirus was isolated from patients exhibiting atypical pneumonia and subsequently proven to be the causative agent of the disease now referred to as severe acute respiratory syndrome (SARS). The complete genome of the SARS coronavirus (SARS-CoV) has since been sequenced. The SARS-CoV nucleocapsid (SARS-CoV N) shares little homology with other members of the coronavirus family. To determine if the N protein is involved in the regulation of cellular signal transduction, an ELISA-based assay on transcription factors was used. We found that the amount of transcription factors binding to promoter sequences of c-Fos, ATF2, CREB-1, and FosB was increased by the expression of SARS-CoV N. Since these factors are related to AP-1 signal transduction pathway, we investigated whether the AP-1 pathway was activated by SARS-CoV N protein using the PathDetect system. The results demonstrated that the expression of N protein, not the membrane protein (M), activated AP-1 pathway. We also found that SARS-CoV N protein does not activate NF-kappaB pathway, demonstrating that activation of important cellular pathways by SAS-CoV N protein is selective. Thus our data for the first time indicate that SARS-CoV has encoded a strategy to regulate cellular signaling process.


Assuntos
Carcinoma Hepatocelular/metabolismo , Proteínas do Nucleocapsídeo/metabolismo , Análise de Sequência com Séries de Oligonucleotídeos/métodos , Coronavírus Relacionado à Síndrome Respiratória Aguda Grave/metabolismo , Transdução de Sinais , Fator de Transcrição AP-1/metabolismo , Ativação Transcricional/fisiologia , Animais , Carcinoma Hepatocelular/genética , Linhagem Celular Tumoral , Chlorocebus aethiops , Humanos , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/metabolismo , Proteínas do Nucleocapsídeo/genética , Coronavírus Relacionado à Síndrome Respiratória Aguda Grave/genética , Especificidade da Espécie , Fator de Transcrição AP-1/genética , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo
18.
J Virol ; 77(10): 5877-88, 2003 May.
Artigo em Inglês | MEDLINE | ID: mdl-12719581

RESUMO

Subversion or appropriation of cellular signal transduction pathways is a common strategy employed by viruses to promote an environment within infected cells that supports the viral replicative cycle. Using subsets of 3T3 murine fibroblasts previously shown to differ in their ability to support myxoma virus (MV) replication, we investigated the role of host serine-threonine kinases (STKs) as potential mediators of the permissive phenotype. Both permissive and nonpermissive 3T3 cells supported equivalent levels of virion binding, entry, and early virus gene expression, indicating that MV tropism in 3T3 cells was not determined by receptor-mediated entry. In contrast, late virus gene expression and viral DNA replication were selectively compromised in restrictive 3T3 cells. Addition of specific protein kinase inhibitors, many of which shared the ability to influence the activity of the STKs p21-activated kinase 1 (PAK-1) and Raf-1 attenuated MV replication in permissive 3T3 cells. Western blot detection of the phosphorylated forms of PAK-1 (Thr423) and Raf-1 (Ser338) confirmed activation of these kinases in permissive cells after MV infection or gamma interferon treatment, but the activated forms of both kinases were greatly reduced or absent in restrictive 3T3 cells. The biological significance of these activations was demonstrated by using the autoinhibitory domain of PAK-1 (amino acids 83 to 149), expression of which reduced the efficiency of MV infection in permissive 3T3 cells concurrent with a decrease in PAK-1 activation. In comparison, overexpression of a constitutively active PAK-1 (T423E) mutant increased MV replication in restrictive 3T3 cells. These observations suggest that induced signaling via cellular STKs may play important roles in determining the permissiveness of host cells to poxvirus infection.


Assuntos
Myxoma virus/patogenicidade , Proteínas Serina-Treonina Quinases/metabolismo , Replicação Viral , Células 3T3 , Animais , Linhagem Celular , Ativação Enzimática , Inibidores Enzimáticos/farmacologia , Camundongos , Myxoma virus/fisiologia , Proteínas Serina-Treonina Quinases/antagonistas & inibidores , Proteínas Serina-Treonina Quinases/genética , Transdução de Sinais , Virulência , Quinases Ativadas por p21
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA