Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
mBio ; 9(5)2018 10 16.
Artigo em Inglês | MEDLINE | ID: mdl-30327438

RESUMO

Enterovirus D68 (EV-D68) has historically been associated with respiratory illnesses. However, in the summers of 2014 and 2016, EV-D68 outbreaks coincided with a spike in polio-like acute flaccid myelitis/paralysis (AFM/AFP) cases. This raised concerns that EV-D68 could be the causative agent of AFM during these recent outbreaks. To assess the potential neurotropism of EV-D68, we utilized the neuroblastoma-derived neuronal cell line SH-SY5Y as a cell culture model to determine if differential infection is observed for different EV-D68 strains. In contrast to HeLa and A549 cells, which support viral infection of all EV-D68 strains tested, SH-SY5Y cells only supported infection by a subset of contemporary EV-D68 strains, including isolates from the 2014 outbreak. Viral replication and infectivity in SH-SY5Y were assessed using multiple assays: virus production, cytopathic effects, cellular ATP release, and VP1 capsid protein production. Similar differential neurotropism was also observed in differentiated SH-SY5Y cells, primary human neuron cultures, and a mouse paralysis model. Using the SH-SY5Y cell culture model, we determined that barriers to viral binding and entry were at least partly responsible for the differential infectivity phenotype. Transfection of genomic RNA into SH-SY5Y generated virions for all EV-D68 isolates, but only a single round of replication was observed from strains that could not directly infect SH-SY5Y. In addition to supporting virus replication and other functional studies, this cell culture model may help identify the signatures of virulence to confirm epidemiological associations between EV-D68 strains and AFM and allow for the rapid identification and characterization of emerging neurotropic strains.IMPORTANCE Since the EV-D68 outbreak during the summer of 2014, evidence of a causal link to a type of limb paralysis (AFM) has been mounting. In this article, we describe a neuronal cell culture model (SH-SY5Y cells) in which a subset of contemporary 2014 outbreak strains of EV-D68 show infectivity in neuronal cells, or neurotropism. We confirmed the difference in neurotropism in vitro using primary human neuron cell cultures and in vivo with a mouse paralysis model. Using the SH-SY5Y cell model, we determined that a barrier to viral entry is at least partly responsible for neurotropism. SH-SY5Y cells may be useful in determining if specific EV-D68 genetic determinants are associated with neuropathogenesis, and replication in this cell line could be used as rapid screening tool for identification of neurotropic EV-D68 strains. This may assist with better understanding of pathogenesis and epidemiology and with the development of potential therapies.


Assuntos
Enterovirus Humano D/fisiologia , Neurônios/virologia , Tropismo Viral , Internalização do Vírus , Replicação Viral , Células A549 , Animais , Técnicas de Cultura de Células , Linhagem Celular , Viroses do Sistema Nervoso Central/virologia , Enterovirus Humano D/genética , Enterovirus Humano D/patogenicidade , Infecções por Enterovirus/virologia , Feminino , Células HeLa , Interações entre Hospedeiro e Microrganismos , Humanos , Camundongos , Mielite/virologia , Doenças Neuromusculares/virologia , Neurônios/citologia , Ligação Viral
2.
Virology ; 515: 250-260, 2018 02.
Artigo em Inglês | MEDLINE | ID: mdl-29324290

RESUMO

Mannose binding lectin (MBL) generally plays a protective role during viral infection, yet MBL-mediated complement activation promotes Ross River virus (RRV)-induced inflammatory tissue destruction, contributing to arthritis and myositis. As MBL binds to carbohydrates, we hypothesized that N-linked glycans on the RRV envelope glycoproteins act as ligands for MBL. Using a panel of RRV mutants lacking the envelope N-linked glycans, we found that MBL deposition onto infected cells was dependent on the E2 glycans. Moreover, the glycan-deficient viruses exhibited reduced disease and tissue damage in a mouse model of RRV-induced myositis compared to wild-type RRV, despite similar viral load and inflammatory infiltrates within the skeletal muscle. Instead, the reduced disease induced by glycan-deficient viruses was linked to decreased MBL deposition and complement activation within inflamed tissues. These results demonstrate that the viral N-linked glycans promote MBL deposition and complement activation onto RRV-infected cells, contributing to the development of RRV-induced myositis.


Assuntos
Infecções por Alphavirus/imunologia , Proteínas do Sistema Complemento/imunologia , Polissacarídeos/imunologia , Ross River virus/imunologia , Proteínas do Envelope Viral/imunologia , Infecções por Alphavirus/virologia , Animais , Ativação do Complemento , Modelos Animais de Doenças , Humanos , Lectina de Ligação a Manose/imunologia , Camundongos Endogâmicos C57BL , Polissacarídeos/química , Ross River virus/genética , Proteínas do Envelope Viral/química , Proteínas do Envelope Viral/genética
3.
J Immunol ; 199(11): 3808-3820, 2017 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-29070670

RESUMO

Type III IFNs are important mediators of antiviral immunity. IFN-λ4 is a unique type III IFN because it is produced only in individuals who carry a dG allele of a genetic variant rs368234815-dG/TT. Counterintuitively, those individuals who can produce IFN-λ4, an antiviral cytokine, are also less likely to clear hepatitis C virus infection. In this study, we searched for unique functional properties of IFN-λ4 that might explain its negative effect on hepatitis C virus clearance. We used fresh primary human hepatocytes (PHHs) treated with recombinant type III IFNs or infected with Sendai virus to model acute viral infection and subsequently validated our findings in HepG2 cell line models. Endogenous IFN-λ4 protein was detectable only in Sendai virus-infected PHHs from individuals with the dG allele, where it was poorly secreted but highly functional, even at concentrations < 50 pg/ml. IFN-λ4 acted faster than other type III IFNs in inducing antiviral genes, as well as negative regulators of the IFN response, such as USP18 and SOCS1 Transient treatment of PHHs with IFN-λ4, but not IFN-λ3, caused a strong and sustained induction of SOCS1 and refractoriness to further stimulation with IFN-λ3. Our results suggest unique functional properties of IFN-λ4 that can be important in viral clearance and other clinical conditions.


Assuntos
Alelos , Hepatócitos/imunologia , Interferons/genética , Interleucinas/genética , Infecções por Respirovirus/imunologia , Vírus Sendai/imunologia , Adolescente , Adulto , Idoso , Endopeptidases/genética , Feminino , Células Hep G2 , Hepacivirus/imunologia , Hepatite C/genética , Hepatite C/imunologia , Hepatócitos/virologia , Humanos , Imunidade , Interferons/metabolismo , Interleucinas/metabolismo , Masculino , Pessoa de Meia-Idade , Polimorfismo de Nucleotídeo Único , Proteína 1 Supressora da Sinalização de Citocina/genética , Ubiquitina Tiolesterase , Regulação para Cima , Carga Viral , Adulto Jovem
4.
PLoS Pathog ; 13(2): e1006164, 2017 02.
Artigo em Inglês | MEDLINE | ID: mdl-28152048

RESUMO

Zika virus (ZIKV) is an emerging mosquito-borne flavivirus that is causally linked to severe neonatal birth defects, including microcephaly, and is associated with Guillain-Barre syndrome in adults. Dendritic cells (DCs) are an important cell type during infection by multiple mosquito-borne flaviviruses, including dengue virus, West Nile virus, Japanese encephalitis virus, and yellow fever virus. Despite this, the interplay between ZIKV and DCs remains poorly defined. Here, we found human DCs supported productive infection by a contemporary Puerto Rican isolate with considerable variability in viral replication, but not viral binding, between DCs from different donors. Historic isolates from Africa and Asia also infected DCs with distinct viral replication kinetics between strains. African lineage viruses displayed more rapid replication kinetics and infection magnitude as compared to Asian lineage viruses, and uniquely induced cell death. Infection of DCs with both contemporary and historic ZIKV isolates led to minimal up-regulation of T cell co-stimulatory and MHC molecules, along with limited secretion of inflammatory cytokines. Inhibition of type I interferon (IFN) protein translation was observed during ZIKV infection, despite strong induction at the RNA transcript level and up-regulation of other host antiviral proteins. Treatment of human DCs with RIG-I agonist potently restricted ZIKV replication, while type I IFN had only modest effects. Mechanistically, we found all strains of ZIKV antagonized type I IFN-mediated phosphorylation of STAT1 and STAT2. Combined, our findings show that ZIKV subverts DC immunogenicity during infection, in part through evasion of type I IFN responses, but that the RLR signaling pathway is still capable of inducing an antiviral state, and therefore may serve as an antiviral therapeutic target.


Assuntos
Proteína DEAD-box 58/imunologia , Células Dendríticas/virologia , Evasão da Resposta Imune/imunologia , Interferon Tipo I/imunologia , Infecção por Zika virus/imunologia , Western Blotting , Células Dendríticas/imunologia , Citometria de Fluxo , Humanos , Reação em Cadeia da Polimerase , Receptores Imunológicos , Zika virus/imunologia
5.
Cell Rep ; 14(7): 1632-1640, 2016 Feb 23.
Artigo em Inglês | MEDLINE | ID: mdl-26876165

RESUMO

Suppression of innate immune responses during filoviral infection contributes to disease severity. Ebola (EBOV) and Marburg (MARV) viruses each encode a VP35 protein that suppresses RIG-I-like receptor signaling and interferon-α/ß (IFN-α/ß) production by several mechanisms, including direct binding to double stranded RNA (dsRNA). Here, we demonstrate that in cell culture, MARV infection results in a greater upregulation of IFN responses as compared to EBOV infection. This correlates with differences in the efficiencies by which EBOV and MARV VP35s antagonize RIG-I signaling. Furthermore, structural and biochemical studies suggest that differential recognition of RNA elements by the respective VP35 C-terminal IFN inhibitory domain (IID) rather than affinity for RNA by the respective VP35s is critical for this observation. Our studies reveal functional differences in EBOV versus MARV VP35 RNA binding that result in unexpected differences in the host response to deadly viral pathogens.


Assuntos
RNA Helicases DEAD-box/genética , Ebolavirus/genética , Interferon-alfa/imunologia , Interferon beta/imunologia , Marburgvirus/genética , RNA de Cadeia Dupla/genética , Proteínas Virais Reguladoras e Acessórias/genética , Sequência de Aminoácidos , Linhagem Celular , Proteína DEAD-box 58 , RNA Helicases DEAD-box/química , RNA Helicases DEAD-box/imunologia , Ebolavirus/imunologia , Regulação da Expressão Gênica , Interações Hospedeiro-Patógeno , Humanos , Interferon-alfa/antagonistas & inibidores , Interferon-alfa/biossíntese , Interferon beta/antagonistas & inibidores , Interferon beta/biossíntese , Marburgvirus/imunologia , Modelos Moleculares , Dados de Sequência Molecular , Monócitos , Ligação Proteica , Estrutura Secundária de Proteína , Estrutura Terciária de Proteína , RNA de Cadeia Dupla/química , RNA de Cadeia Dupla/imunologia , Receptores Imunológicos , Alinhamento de Sequência , Transdução de Sinais , Especificidade da Espécie , Proteínas Virais Reguladoras e Acessórias/química , Proteínas Virais Reguladoras e Acessórias/imunologia
6.
Cell Rep ; 11(3): 376-89, 2015 Apr 21.
Artigo em Inglês | MEDLINE | ID: mdl-25865894

RESUMO

During viral RNA synthesis, Ebola virus (EBOV) nucleoprotein (NP) alternates between an RNA-template-bound form and a template-free form to provide the viral polymerase access to the RNA template. In addition, newly synthesized NP must be prevented from indiscriminately binding to noncognate RNAs. Here, we investigate the molecular bases for these critical processes. We identify an intrinsically disordered peptide derived from EBOV VP35 (NPBP, residues 20-48) that binds NP with high affinity and specificity, inhibits NP oligomerization, and releases RNA from NP-RNA complexes in vitro. The structure of the NPBP/ΔNPNTD complex, solved to 3.7 Å resolution, reveals how NPBP peptide occludes a large surface area that is important for NP-NP and NP-RNA interactions and for viral RNA synthesis. Together, our results identify a highly conserved viral interface that is important for EBOV replication and can be targeted for therapeutic development.


Assuntos
Ebolavirus/fisiologia , Nucleoproteínas/metabolismo , RNA Viral/metabolismo , Proteínas do Core Viral/metabolismo , Calorimetria , Microscopia Crioeletrônica , Cristalografia por Raios X , Células HeLa , Doença pelo Vírus Ebola/metabolismo , Humanos , Proteínas do Nucleocapsídeo , Estrutura Quaternária de Proteína , Replicação Viral
7.
mBio ; 5(6): e02011, 2014 Nov 04.
Artigo em Inglês | MEDLINE | ID: mdl-25370495

RESUMO

UNLABELLED: Deep sequencing of RNAs produced by Zaire ebolavirus (EBOV) or the Angola strain of Marburgvirus (MARV-Ang) identified novel viral and cellular mechanisms that diversify the coding and noncoding sequences of viral mRNAs and genomic RNAs. We identified previously undescribed sites within the EBOV and MARV-Ang mRNAs where apparent cotranscriptional editing has resulted in the addition of non-template-encoded residues within the EBOV glycoprotein (GP) mRNA, the MARV-Ang nucleoprotein (NP) mRNA, and the MARV-Ang polymerase (L) mRNA, such that novel viral translation products could be produced. Further, we found that the well-characterized EBOV GP mRNA editing site is modified at a high frequency during viral genome RNA replication. Additionally, editing hot spots representing sites of apparent adenosine deaminase activity were found in the MARV-Ang NP 3'-untranslated region. These studies identify novel filovirus-host interactions and reveal production of a greater diversity of filoviral gene products than was previously appreciated. IMPORTANCE: This study identifies novel mechanisms that alter the protein coding capacities of Ebola and Marburg virus mRNAs. Therefore, filovirus gene expression is more complex and diverse than previously recognized. These observations suggest new directions in understanding the regulation of filovirus gene expression.


Assuntos
Ebolavirus/genética , Marburgvirus/genética , Edição de RNA , RNA Mensageiro/metabolismo , Replicação Viral , Animais , Linhagem Celular , Chlorocebus aethiops , Ebolavirus/fisiologia , Sequenciamento de Nucleotídeos em Larga Escala , Interações Hospedeiro-Patógeno , Humanos , Marburgvirus/fisiologia , Dados de Sequência Molecular , RNA Mensageiro/genética , RNA Viral/genética , RNA Viral/metabolismo , Análise de Sequência de DNA
8.
J Mol Biol ; 426(10): 2045-58, 2014 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-24495995

RESUMO

The Ebola virus (EBOV) genome only encodes a single viral polypeptide with enzymatic activity, the viral large (L) RNA-dependent RNA polymerase protein. However, currently, there is limited information about the L protein, which has hampered the development of antivirals. Therefore, antifiloviral therapeutic efforts must include additional targets such as protein-protein interfaces. Viral protein 35 (VP35) is multifunctional and plays important roles in viral pathogenesis, including viral mRNA synthesis and replication of the negative-sense RNA viral genome. Previous studies revealed that mutation of key basic residues within the VP35 interferon inhibitory domain (IID) results in significant EBOV attenuation, both in vitro and in vivo. In the current study, we use an experimental pipeline that includes structure-based in silico screening and biochemical and structural characterization, along with medicinal chemistry, to identify and characterize small molecules that target a binding pocket within VP35. NMR mapping experiments and high-resolution x-ray crystal structures show that select small molecules bind to a region of VP35 IID that is important for replication complex formation through interactions with the viral nucleoprotein (NP). We also tested select compounds for their ability to inhibit VP35 IID-NP interactions in vitro as well as VP35 function in a minigenome assay and EBOV replication. These results confirm the ability of compounds identified in this study to inhibit VP35-NP interactions in vitro and to impair viral replication in cell-based assays. These studies provide an initial framework to guide development of antifiloviral compounds against filoviral VP35 proteins.


Assuntos
Antivirais/química , Coenzimas/antagonistas & inibidores , Ebolavirus/efeitos dos fármacos , Bibliotecas de Moléculas Pequenas/química , Proteínas Virais Reguladoras e Acessórias/antagonistas & inibidores , Antivirais/farmacologia , Coenzimas/química , Simulação por Computador , Cristalografia por Raios X , RNA Polimerases Dirigidas por DNA/antagonistas & inibidores , RNA Polimerases Dirigidas por DNA/metabolismo , Ebolavirus/enzimologia , Modelos Moleculares , Ressonância Magnética Nuclear Biomolecular , Ligação Proteica , Domínios e Motivos de Interação entre Proteínas/fisiologia , Pirróis/química , Pirróis/metabolismo , Pirróis/farmacologia , Bibliotecas de Moléculas Pequenas/farmacologia , Proteínas Virais Reguladoras e Acessórias/química
9.
J Virol ; 87(7): 3801-14, 2013 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-23345511

RESUMO

Antigen-presenting cells (APCs) are critical targets of Ebola virus (EBOV) infection in vivo. However, the susceptibility of monocytes to infection is controversial. Studies indicate productive monocyte infection, and yet monocytes are also reported to be resistant to EBOV GP-mediated entry. In contrast, monocyte-derived macrophages and dendritic cells are permissive for both EBOV entry and replication. Here, freshly isolated monocytes are demonstrated to indeed be refractory to EBOV entry. However, EBOV binds monocytes, and delayed entry occurs during monocyte differentiation. Cultured monocytes spontaneously downregulate the expression of viral entry restriction factors such as interferon-inducible transmembrane proteins, while upregulating the expression of critical EBOV entry factors cathepsin B and NPC1. Moreover, these processes are accelerated by EBOV infection. Finally, ectopic expression of NPC1 is sufficient to rescue entry into an undifferentiated, normally nonpermissive monocytic cell line. These results define the molecular basis for infection of APCs and suggest means to limit APC infection.


Assuntos
Diferenciação Celular/fisiologia , Ebolavirus/fisiologia , Monócitos/virologia , Ligação Viral , Internalização do Vírus , Proteínas de Transporte/metabolismo , Catepsina B/metabolismo , Primers do DNA/genética , Células Dendríticas/virologia , Citometria de Fluxo , Células HEK293 , Humanos , Peptídeos e Proteínas de Sinalização Intracelular , Macrófagos/virologia , Glicoproteínas de Membrana/metabolismo , Monócitos/fisiologia , Proteína C1 de Niemann-Pick , Reação em Cadeia da Polimerase Via Transcriptase Reversa
10.
Proc Natl Acad Sci U S A ; 109(50): 20661-6, 2012 Dec 11.
Artigo em Inglês | MEDLINE | ID: mdl-23185024

RESUMO

Filoviruses, marburgvirus (MARV) and ebolavirus (EBOV), are causative agents of highly lethal hemorrhagic fever in humans. MARV and EBOV share a common genome organization but show important differences in replication complex formation, cell entry, host tropism, transcriptional regulation, and immune evasion. Multifunctional filoviral viral protein (VP) 35 proteins inhibit innate immune responses. Recent studies suggest double-stranded (ds)RNA sequestration is a potential mechanism that allows EBOV VP35 to antagonize retinoic-acid inducible gene-I (RIG-I) like receptors (RLRs) that are activated by viral pathogen-associated molecular patterns (PAMPs), such as double-strandedness and dsRNA blunt ends. Here, we show that MARV VP35 can inhibit IFN production at multiple steps in the signaling pathways downstream of RLRs. The crystal structure of MARV VP35 IID in complex with 18-bp dsRNA reveals that despite the similar protein fold as EBOV VP35 IID, MARV VP35 IID interacts with the dsRNA backbone and not with blunt ends. Functional studies show that MARV VP35 can inhibit dsRNA-dependent RLR activation and interferon (IFN) regulatory factor 3 (IRF3) phosphorylation by IFN kinases TRAF family member-associated NFkb activator (TANK) binding kinase-1 (TBK-1) and IFN kB kinase e (IKKe) in cell-based studies. We also show that MARV VP35 can only inhibit RIG-I and melanoma differentiation associated gene 5 (MDA5) activation by double strandedness of RNA PAMPs (coating backbone) but is unable to inhibit activation of RLRs by dsRNA blunt ends (end capping). In contrast, EBOV VP35 can inhibit activation by both PAMPs. Insights on differential PAMP recognition and inhibition of IFN induction by a similar filoviral VP35 fold, as shown here, reveal the structural and functional plasticity of a highly conserved virulence factor.


Assuntos
Marburgvirus/imunologia , Marburgvirus/patogenicidade , Proteínas Virais Reguladoras e Acessórias/química , Proteínas Virais Reguladoras e Acessórias/imunologia , Sequência de Aminoácidos , Animais , Sequência de Bases , Cristalografia por Raios X , Células HEK293 , Interações Hospedeiro-Patógeno , Humanos , Quinase I-kappa B/antagonistas & inibidores , Imunidade Inata , Interferon Tipo I/antagonistas & inibidores , Doença do Vírus de Marburg/etiologia , Doença do Vírus de Marburg/imunologia , Doença do Vírus de Marburg/virologia , Marburgvirus/química , Modelos Biológicos , Modelos Moleculares , Dados de Sequência Molecular , Proteínas Serina-Treonina Quinases/antagonistas & inibidores , Estrutura Terciária de Proteína , RNA/química , RNA/genética , RNA/metabolismo , Homologia de Sequência de Aminoácidos , Virulência/imunologia
11.
PLoS Pathog ; 8(10): e1002934, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-23055924

RESUMO

Dengue virus (DENV) is a pathogen with a high impact on human health. It replicates in a wide range of cells involved in the immune response. To efficiently infect humans, DENV must evade or inhibit fundamental elements of the innate immune system, namely the type I interferon response. DENV circumvents the host immune response by expressing proteins that antagonize the cellular innate immunity. We have recently documented the inhibition of type I IFN production by the proteolytic activity of DENV NS2B3 protease complex in human monocyte derived dendritic cells (MDDCs). In the present report we identify the human adaptor molecule STING as a target of the NS2B3 protease complex. We characterize the mechanism of inhibition of type I IFN production in primary human MDDCs by this viral factor. Using different human and mouse primary cells lacking STING, we show enhanced DENV replication. Conversely, mutated versions of STING that cannot be cleaved by the DENV NS2B3 protease induced higher levels of type I IFN after infection with DENV. Additionally, we show that DENV NS2B3 is not able to degrade the mouse version of STING, a phenomenon that severely restricts the replication of DENV in mouse cells, suggesting that STING plays a key role in the inhibition of DENV infection and spread in mice.


Assuntos
Células Dendríticas/metabolismo , Vírus da Dengue/imunologia , Vírus da Dengue/patogenicidade , Interferon Tipo I/biossíntese , Proteínas de Membrana/metabolismo , Proteínas não Estruturais Virais/metabolismo , Aedes , Animais , Células Cultivadas , Chlorocebus aethiops , Cricetinae , Células Dendríticas/virologia , Vírus da Dengue/metabolismo , Células HEK293 , Humanos , Evasão da Resposta Imune , Macrófagos/metabolismo , Macrófagos/virologia , Proteínas de Membrana/genética , Camundongos , Camundongos Endogâmicos C57BL , Interferência de RNA , RNA Interferente Pequeno , Transdução de Sinais , Células Vero , Replicação Viral
12.
J Virol ; 84(20): 10581-91, 2010 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-20686031

RESUMO

The ebolavirus (EBOV) VP35 protein binds to double-stranded RNA (dsRNA), inhibits host alpha/beta interferon (IFN-α/ß) production, and is an essential component of the viral polymerase complex. Structural studies of the VP35 C-terminal IFN inhibitory domain (IID) identified specific structural features, including a central basic patch and a hydrophobic pocket, that are important for dsRNA binding and IFN inhibition. Several other conserved basic residues bordering the central basic patch and a separate cluster of basic residues, called the first basic patch, were also identified. Functional analysis of alanine substitution mutants indicates that basic residues outside the central basic patch are not required for dsRNA binding or for IFN inhibition. However, minigenome assays, which assess viral RNA polymerase complex function, identified these other basic residues to be critical for viral RNA synthesis. Of these, a subset located within the first basic patch is important for VP35-nucleoprotein (NP) interaction, as evidenced by the inability of alanine substitution mutants to coimmunoprecipitate with NP. Therefore, first basic patch residues are likely critical for replication complex formation through interactions with NP. Coimmunoprecipitation studies further demonstrate that the VP35 IID is sufficient to interact with NP and that dsRNA can modulate VP35 IID interactions with NP. Other basic residue mutations that disrupt the VP35 polymerase cofactor function do not affect interaction with NP or with the amino terminus of the viral polymerase. Collectively, these results highlight the importance of conserved basic residues from the EBOV VP35 C-terminal IID and validate the VP35 IID as a potential therapeutic target.


Assuntos
Ebolavirus/fisiologia , Proteínas Virais Reguladoras e Acessórias/química , Proteínas Virais Reguladoras e Acessórias/fisiologia , Substituição de Aminoácidos , Animais , Linhagem Celular , Chlorocebus aethiops , Ebolavirus/genética , Ebolavirus/patogenicidade , Interações Hospedeiro-Patógeno/genética , Interações Hospedeiro-Patógeno/fisiologia , Humanos , Modelos Moleculares , Mutagênese Sítio-Dirigida , Proteínas Mutantes/química , Proteínas Mutantes/genética , Proteínas Mutantes/metabolismo , Proteínas do Nucleocapsídeo , Nucleoproteínas/química , Nucleoproteínas/genética , Nucleoproteínas/fisiologia , Domínios e Motivos de Interação entre Proteínas , Estrutura Terciária de Proteína , RNA/genética , RNA/metabolismo , Proteínas Recombinantes de Fusão/química , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , Eletricidade Estática , Células Vero , Proteínas do Core Viral/química , Proteínas do Core Viral/genética , Proteínas do Core Viral/fisiologia , Proteínas Virais Reguladoras e Acessórias/genética , Virulência/genética , Virulência/fisiologia
13.
J Virol ; 82(24): 12374-83, 2008 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-18922878

RESUMO

Alphaviruses are mosquito-transmitted viruses that cause significant human disease, and understanding how these pathogens successfully transition from the mosquito vector to the vertebrate host is an important area of research. Previous studies demonstrated that mosquito and mammalian-cell-derived alphaviruses differentially induce type I interferons (alpha/beta interferon [IFN-alpha/beta]) in myeloid dendritic cells (mDCs), where the mosquito cell-derived virus is a poor inducer of IFN-alpha/beta compared to the mammalian-cell-derived virus. Furthermore, the reduced IFN-alpha/beta induction by the mosquito cell-derived virus is attributed to differential N-linked glycosylation. To further evaluate the role of viral envelope glycans in regulating the IFN-alpha/beta response, studies were performed to assess whether the mosquito cell-derived virus actively inhibits IFN-alpha/beta induction or is simply a poor inducer of IFN-alpha/beta. Coinfection studies using mammalian- and mosquito cell-derived Ross River virus (mam-RRV and mos-RRV, respectively) indicated that mos-RRV was unable to suppress IFN-alpha/beta induction by mam-RRV in mDC cultures. Additionally, a panel of mutant viruses lacking either individual or multiple N-linked glycosylation sites was used to demonstrate that N-linked glycans were essential for high-level IFN-alpha/beta induction by the mammalian-cell-derived virus. These results suggest that the failure of the mosquito cell-derived virus to induce IFN-alpha/beta is due to a lack of complex carbohydrates on the virion rather than the active suppression of the DC antiviral response.


Assuntos
Células Dendríticas/metabolismo , Interferon Tipo I/biossíntese , Células Mieloides/metabolismo , Polissacarídeos/metabolismo , Ross River virus/metabolismo , Proteínas do Envelope Viral/metabolismo , Aedes , Animais , Linhagem Celular , Cricetinae , Genoma Viral/genética , Mutação/genética , Ross River virus/genética , Proteínas do Envelope Viral/genética
14.
J Virol ; 80(2): 737-49, 2006 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-16378976

RESUMO

Mosquito-borne alphaviruses are a significant cause of both encephalitic and arthritic disease in humans worldwide. In contrast to the encephalitic alphaviruses, the pathogenesis of alphavirus-induced arthritic disease is not well understood. Utilizing a mouse model of Ross River virus (RRV) disease, we found that the primary targets of RRV infection are bone, joint, and skeletal muscle tissues of the hind limbs in both outbred CD-1 mice and adult C57BL/6J mice. Moreover, histological analyses demonstrated that RRV infection resulted in severe inflammation of these tissues. Characterization of the inflammatory infiltrate within the skeletal muscle tissue identified inflammatory macrophages, NK cells, and CD4+ and CD8+ T lymphocytes. To determine the contribution of the adaptive immune system, the outcome of RRV-induced disease was examined in C57BL/6J RAG-1(-/-) mice, which lack functional T and B lymphocytes. RAG-1(-/-) and wild-type mice developed similar disease signs, infiltration of inflammatory macrophages and NK cells, and muscle pathology, suggesting that the adaptive immune response does not play a critical role in the development of disease. These results establish the mouse model of RRV disease as a useful system for the identification of viral and host factors that contribute to alphavirus-induced arthritis and myositis.


Assuntos
Infecções por Alphavirus/patologia , Infecções por Alphavirus/virologia , Artrite Infecciosa/patologia , Artrite Infecciosa/virologia , Inflamação/patologia , Miosite/patologia , Miosite/virologia , Ross River virus , Animais , Osso e Ossos/patologia , Osso e Ossos/virologia , Linfócitos T CD4-Positivos , Linfócitos T CD8-Positivos , Modelos Animais de Doenças , Feminino , Membro Posterior/patologia , Membro Posterior/virologia , Proteínas de Homeodomínio/genética , Inflamação/imunologia , Articulações/patologia , Articulações/virologia , Células Matadoras Naturais , Macrófagos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Músculo Esquelético/patologia , Músculo Esquelético/virologia , Especificidade de Órgãos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA