Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
J Virol ; 82(1): 522-8, 2008 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-17959666

RESUMO

Interleukin-18 (IL-18) is a critical proinflammatory cytokine whose extracellular bioactivity is regulated by a cellular IL-18 binding protein (IL-18BP). Many poxviruses have acquired variants of this IL-18BP gene, some of which have been shown to act as viral virulence factors. Yaba monkey tumor virus (YMTV) encodes a related family member, 14L, which is similar to the orthopoxvirus IL-18BPs. YMTV 14L was expressed from a baculovirus system and tested for its ability to bind and inhibit IL-18. We found that YMTV 14L bound both human IL-18 (hIL-18) and murine IL-18 with high affinity, at 4.1 nM and 6.5 nM, respectively. YMTV 14L was able to fully sequester hIL-18 but could only partially inhibit the biological activity of hIL-18 as measured by gamma interferon secretion from KG-1 cells. Additionally, 17 hIL-18 point mutants were tested by surface plasmon resonance for their ability to bind to YMTV 14L. Two clusters of hIL-18 surface residues were found to be important for the hIL-18-YMTV 14L interaction, in contrast to results for the Variola virus IL-18BP, which has been shown to primarily interact with a single cluster of three amino acids. The altered binding specificity of YMTV 14L most likely represents an adaptation resulting in increased fitness of the virus and affirms the plasticity of poxviral inhibitor domains that target cytokines like IL-18.


Assuntos
Peptídeos e Proteínas de Sinalização Intercelular/fisiologia , Interleucina-18/antagonistas & inibidores , Proteínas Virais/fisiologia , Vírus do Tumor do Macaco de Yaba/imunologia , Linhagem Celular , Humanos , Interferon gama/biossíntese , Mutagênese Sítio-Dirigida , Mutação Puntual , Ligação Proteica , Mapeamento de Interação de Proteínas , Ressonância de Plasmônio de Superfície
2.
Virology ; 368(1): 32-40, 2007 Nov 10.
Artigo em Inglês | MEDLINE | ID: mdl-17632198

RESUMO

Tanapox virus (TPV) belongs to the genus Yatapoxvirus and causes a relatively benign zoonotic disease in man, with symptoms that resemble a mild version of human monkeypox. In order to investigate the underlying mechanisms of TPV pathogenesis, the tropism and replication characteristics of TPV were examined in a variety of primary human cells. A GFP expressing TPV (TPV-GFP) was constructed and used to infect primary human dermal fibroblasts (pHDFs) and peripheral blood mononuclear cells (PBMCs), both of which are believed to be major in vivo targets of poxvirus infection. pHDFs fully supported productive replication and cell-cell spread of TPV-GFP. However, induction of cell cycle arrest in pHDFs by contact mediated inhibition or rapamycin treatment eliminated the ability of TPV to fully stimulate cell cycle progression and dramatically reduced viral replication. TPV-GFP-infected human PBMCs were screened for permissiveness by FACS analysis. CD14+ cells (monocytes) were the primary cellular target for TPV infection. A small proportion of CD3+ cells (T cells) were positive for GFP expression, yet TPV was not able to replicate and spread in cultured peripheral blood lymphocytes, regardless of their state of activation. Primary human monocytes, however, demonstrated robust TPV replication, yet these cells no longer supported replication of TPV once they differentiated into macrophages. This unique ex vivo tropism of TPV gives key insights into the basis for the self-limiting pathogenicity of TPV in man.


Assuntos
Fibroblastos/virologia , Monócitos/virologia , Yatapoxvirus/fisiologia , Complexo CD3/análise , Células Cultivadas , Genes Reporter , Proteínas de Fluorescência Verde/biossíntese , Proteínas de Fluorescência Verde/genética , Humanos , Receptores de Lipopolissacarídeos/análise , Linfócitos T/virologia , Replicação Viral/fisiologia , Yatapoxvirus/genética , Yatapoxvirus/crescimento & desenvolvimento
3.
Virus Res ; 129(1): 11-25, 2007 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-17574698

RESUMO

Members of the genus Yatapoxvirus, which include Tanapox virus (TPV) and Yaba monkey tumor virus, infect primates including humans. Two strains of TPV isolated 50 years apart from patients infected from the equatorial region of Africa have been sequenced. The original isolate from a human case in the Tana River Valley, Kenya, in 1957 (TPV-Kenya) and an isolate from an infected traveler in the Republic of Congo in 2004 (TPV-RoC). Although isolated 50 years apart the genomes were highly conserved. The genomes differed at only 35 of 144,565 nucleotide positions (99.98% identical). We predict that TPV-RoC encodes 155 ORFs, however a single transversion (at nucleotide 10241) in TPV-Kenya resulted in the coding capacity for two predicted ORFs (11.1L and 11.2L) in comparison to a single ORF (11L) in TPV-RoC. The genomes of TPV are A+T rich (73%) and 96% of the sequence encodes predicted ORFs. Comparative genomic analysis identified several features shared with other chordopoxviruses. A conserved sequence within the terminal inverted repeat region that is also present in the other members of the Yatapoxviruses as well as members of the Capripoxviruses, Swinepox virus and an unclassified Deerpox virus suggests the existence of a conserved near-terminal sequence secondary structure. Two previously unidentified gene families were annotated that are represented by ORF TPV28L, which matched homologues in certain other chordopoxviruses, and TPV42.5L, which is highly conserved among currently reported chordopoxvirus sequences.


Assuntos
DNA Viral/genética , Genoma Viral , Infecções por Poxviridae/virologia , Infecções Tumorais por Vírus/virologia , Yatapoxvirus/genética , África , Sequência de Aminoácidos , Humanos , Dados de Sequência Molecular , Fases de Leitura Aberta , Alinhamento de Sequência , Homologia de Sequência
4.
J Virol ; 81(3): 1251-60, 2007 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-17108021

RESUMO

Myxoma virus is a rabbit-specific poxvirus pathogen that also exhibits a unique tropism for human tumor cells and is dramatically oncolytic for human cancer xenografts. Most tumor cell lines tested are permissive for myxoma infection in a fashion intimately tied to the activation state of Akt kinase. A host range factor of myxoma virus, M-T5, directly interacts with Akt and mediates myxoma virus tumor cell tropism. mTOR is a regulator of cell growth and metabolism downstream of Akt and is specifically inhibited by rapamycin. We report that treatment of nonpermissive human tumor cell lines, which normally restrict myxoma virus replication, with rapamycin dramatically increased virus tropism and spread in vitro. This increased myxoma replication is concomitant with global effects on mTOR signaling, specifically, an increase in Akt kinase. In contrast to the effects on human cancer cells, rapamycin does not increase myxoma virus replication in rabbit cell lines or permissive human tumor cell lines with constitutively active Akt. This indicates that rapamycin increases the oncolytic capacity of myxoma virus for human cancer cells by reconfiguring the internal cell signaling environment to one that is optimal for productive virus replication and suggests the possibility of a potentially therapeutic synergism between kinase signaling inhibitors and oncolytic poxviruses for cancer treatment.


Assuntos
Myxoma virus/fisiologia , Neoplasias/terapia , Terapia Viral Oncolítica/métodos , Vírus Oncolíticos/metabolismo , Proteínas Virais/metabolismo , Linhagem Celular Tumoral , Humanos , Sirolimo
5.
Virus Genes ; 33(1): 15-26, 2006 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-16791414

RESUMO

Transient expression of viral genes from certain poxviruses in uninfected mammalian cells can sometimes be unexpectedly inefficient. The reasons for poor expression levels can be due to a number of features of the gene cassette, such as cryptic splice sites, polymerase II termination sequences or motifs that lead to mRNA instability. Here we suggest that in some cases the problem of low protein expression in transfected mammalian cells may be due to inefficient codon usage. We have observed that for many poxvirus genes from the yatapoxvirus genus this deficiency can be overcome by synthesis of the gene with codon sequences optimized for expression in primate cells. This led us to examine colon usage across 2-dozen sequenced members of the Poxviridae. We conclude that codon usage is surprisingly divergent across the different Poxviridae genera but is much more conserved within a single genus. Thus, Poxviridae genera can be divided into distinct groups based on their observed codon bias. When viewed in this context, successful transient expression of transfected poxvirus genes in uninfected mammalian cells can be more accurately predicted based on codon bias. As a corollary, for specific poxvirus genes with less favorable codon usage, codon optimization can result in profoundly increased transient expression levels following transfection of uninfected mammalian cell lines.


Assuntos
Códon , DNA Viral/genética , Regulação Viral da Expressão Gênica/fisiologia , Poxviridae/genética , Animais , Sequência de Bases , Células COS , Linhagem Celular , Chlorocebus aethiops , Humanos , Dados de Sequência Molecular , Poxviridae/classificação , Poxviridae/metabolismo , Transfecção
6.
J Biol Chem ; 281(12): 8041-50, 2006 Mar 24.
Artigo em Inglês | MEDLINE | ID: mdl-16407226

RESUMO

The thrombolytic serine protease cascade is intricately involved in activation of innate immune responses. The urokinase-type plasminogen activator and receptor form complexes that aid inflammatory cell invasion at sites of arterial injury. Plasminogen activator inhibitor-1 is a mammalian serpin that binds and regulates the urokinase receptor complex. Serp-1, a myxomaviral serpin, also targets the urokinase receptor, displaying profound anti-inflammatory and anti-atherogenic activity in a wide range of animal models. Serp-1 reactive center site mutations, mimicking known mammalian and viral serpins, were constructed in order to define sequences responsible for regulation of inflammation. Thrombosis, inflammation, and plaque growth were assessed after treatment with Serp-1, Serp-1 chimeras, plasminogen activator inhibitor-1, or unrelated viral serpins in plasminogen activator inhibitor or urokinase receptor-deficient mouse aortic transplants. Altering the P1-P1' Arg-Asn sequence compromised Serp-1 protease-inhibitory activity and anti-inflammatory activity in animal models; P1-P1' Ala-Ala mutants were inactive, P1 Met increased remodeling, and P1' Thr increased thrombosis. Substitution of Serp-1 P2-P7 with Ala6 allowed for inhibition of urokinase but lost plasmin inhibition, unexpectedly inducing a diametrically opposed, proinflammatory response with mononuclear cell activation, thrombosis, and aneurysm formation (p < 0.03). Other serpins did not reproduce Serp-1 activity; plasminogen activator inhibitor-1 increased thrombosis (p < 0.0001), and unrelated viral serpin, CrmA, increased inflammation. Deficiency of urokinase receptor in mouse transplants blocked Serp-1 and chimera activity, in some cases increasing inflammation. In summary, 1) Serp-1 anti-inflammatory activity is highly dependent upon the reactive center loop sequence, and 2) plasmin inhibition is central to anti-inflammatory activity.


Assuntos
Myxoma virus/metabolismo , Serpinas/química , Animais , Aorta/metabolismo , Aorta/transplante , Sequência de Bases , Sítios de Ligação , Coagulação Sanguínea , Células CHO , Adesão Celular , Movimento Celular , Separação Celular , Cricetinae , Modelos Animais de Doenças , Citometria de Fluxo , Sistema Imunitário , Inflamação , Cinética , Leucócitos Mononucleares/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Modelos Estatísticos , Dados de Sequência Molecular , Peritônio/citologia , Peritônio/metabolismo , Inibidor 1 de Ativador de Plasminogênio/metabolismo , Ativação Plaquetária , Ligação Proteica , Isoformas de Proteínas , Proteínas Recombinantes de Fusão/química , Proteínas Recombinantes de Fusão/metabolismo , Trombose , Fatores de Tempo , Tempo de Coagulação do Sangue Total
7.
Immunity ; 23(6): 587-98, 2005 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-16356857

RESUMO

Proinflammatory caspases play an essential role in innate immune responses to infection by regulating the cleavage and activation of proinflammatory cytokines. Activation of these enzymes requires the assembly of an intracellular molecular platform, termed the inflammasome, which is comprised of members of the pyrin domain (PYD)-containing superfamily of apoptosis and inflammation-regulatory proteins. We report here the identification and characterization of a poxvirus-encoded PYD-containing protein that interacts with the ASC-1 component of the inflammasome and inhibits caspase-1 activation and the processing of IL-1beta and IL-18 induced by diverse stimuli. Knockout viruses that do not express this protein are unable to productively infect monocytes and lymphocytes due to an abortive phenotype and are markedly attenuated in susceptible hosts due to decreased virus dissemination and enhanced inflammatory responses at sites of infection. Thus, modulation of inflammasome function constitutes an important immunomodulatory strategy employed by poxviruses to circumvent host antiviral responses.


Assuntos
Apoptose/imunologia , Caspase 1/metabolismo , Inflamação/imunologia , Modelos Imunológicos , Myxoma virus/imunologia , Proteínas Virais/imunologia , Proteínas Virais/metabolismo , Sequência de Aminoácidos , Animais , Proteínas Adaptadoras de Sinalização CARD , Biologia Computacional , Proteínas do Citoesqueleto/metabolismo , Ativação Enzimática/imunologia , Técnica Indireta de Fluorescência para Anticorpo , Humanos , Immunoblotting , Inflamação/virologia , Interleucina-1/metabolismo , Interleucina-18/metabolismo , Microscopia de Fluorescência , Dados de Sequência Molecular , Myxoma virus/genética , Coelhos , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Alinhamento de Sequência , Proteínas Virais/genética
8.
Virology ; 332(1): 235-48, 2005 Feb 05.
Artigo em Inglês | MEDLINE | ID: mdl-15661156

RESUMO

Recent studies have indicated a critical role for interferon (IFN)-mediated antiviral responses in the host range of myxoma virus (MV), a pathogenic poxvirus of rabbits. To investigate the contribution of IFN to MV tropism in nonleporine cells, primary human dermal fibroblasts (HDFs) were tested for permissiveness to MV infection. Low-passage HDFs that underwent fewer than 25 population doublings (PD) were fully permissive for MV infection, supporting productive virus replication and cell-to-cell spread. In contrast, early and late viral gene expression was detectable in high-passage HDF (>75 PD), but MV failed to generate infectious progeny and could not form foci in these cells. Vesicular stomatitis virus (VSV) plaque reduction assays confirmed that constitutive IFN production progressively increased as HDFs were passaged, concurrent with an increase in the expression of transcripts for type I IFN and IFN-responsive genes involved in antiviral responses. These findings correlated with the enhanced sensitivity of higher-passage HDF to inducers of type I IFN responses, such as dsRNA. Furthermore, pretreatment of low-passage HDF with type I IFN abrogated MV spread and replication while treatment of mature HDF with neutralizing antibodies to IFN-beta, but not IFN-alpha, restored the capacity to form foci. These findings emphasize the importance of post-entry events in determining the permissiveness of human cells to MV infection and support a critical role for innate type I IFN responses as key determinants of poxvirus host range and species restriction.


Assuntos
Fibroblastos/virologia , Interferon-alfa/biossíntese , Interferon beta/fisiologia , Myxoma virus/fisiologia , Linhagem Celular , Fibroblastos/imunologia , Fibroblastos/fisiologia , Humanos , Interferon beta/metabolismo , Ensaio de Placa Viral
9.
J Virol ; 78(13): 7097-111, 2004 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-15194786

RESUMO

M11L, a 166-amino-acid antiapoptotic protein of myxoma virus, was previously shown to bind to the peripheral benzodiazepine receptor by hydrophobic interactions at the outer mitochondrial membrane. Here we demonstrate that an additional property of M11L is the ability to constitutively form inhibitory complexes with the proapoptotic Bcl-2 family member Bak in human cells. This binding interaction was identified by both FLAG-tagged pull-down assays and tandem affinity purification from transfected and virus-infected human cells. M11L binds constitutively to human Bak and, under some inducible conditions, to human Bax as well, but not to the other Bcl-2 family members (Bad, Bid, Bcl-2). When stably expressed in human embryonic kidney (HEK293) cells, M11L effectively protects these cells from Fas ligand-induced apoptosis, thereby blocking release of cytochrome c, activation of caspase 9, and cleavage of poly(ADP-ribose) polymerase. We also demonstrate in coexpression studies that M11L can interact with Bak independently of any involvement with Bax. Furthermore, cells stably expressing M11L function to prevent apoptosis that is induced by overexpression of Bak. We conclude that M11L inhibits, in a species-independent fashion, apoptotic signals mediated by activation of Bak.


Assuntos
Apoptose , Proteínas de Membrana/metabolismo , Myxoma virus/patogenicidade , Proteínas Virais/metabolismo , Sequência de Aminoácidos , Animais , Linhagem Celular , Humanos , Proteínas de Membrana/química , Dados de Sequência Molecular , Proteínas Proto-Oncogênicas/química , Proteínas Proto-Oncogênicas/metabolismo , Proteínas Proto-Oncogênicas c-bcl-2/química , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Coelhos , Especificidade da Espécie , Transfecção , Proteína Killer-Antagonista Homóloga a bcl-2 , Proteína X Associada a bcl-2
10.
Mol Cell Biol ; 23(22): 8042-57, 2003 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-14585965

RESUMO

Stimulation of T cells through their antigen receptors (TCRs) causes a transient increase in the intracellular concentration of cyclic AMP (cAMP). However, sustained high levels of cAMP inhibit T-cell responses, suggesting that TCR signaling is coordinated with the activation of cyclic nucleotide phosphodiesterases (PDEs). The molecular basis of such a pathway is unknown. Here we show that TCR-dependent signaling activates PDE4B2 and that this enhances interleukin-2 production. Such an effect requires the regulatory N terminus of PDE4B2 and correlates with partitioning within lipid rafts, early targeting of this PDE to the immunological synapse, and subsequent accumulation in the antipodal pole of the T cell as activation proceeds.


Assuntos
3',5'-AMP Cíclico Fosfodiesterases/metabolismo , Linfócitos T/enzimologia , Linfócitos T/imunologia , 3',5'-AMP Cíclico Fosfodiesterases/química , 3',5'-AMP Cíclico Fosfodiesterases/genética , Compartimento Celular , Nucleotídeo Cíclico Fosfodiesterase do Tipo 4 , Ativação Enzimática , Humanos , Técnicas In Vitro , Interleucina-2/biossíntese , Células Jurkat , Ativação Linfocitária , Microdomínios da Membrana/enzimologia , Estrutura Terciária de Proteína , Receptores de Antígenos de Linfócitos T/metabolismo , Proteínas Recombinantes de Fusão/química , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , Deleção de Sequência , Transdução de Sinais
11.
Annu Rev Immunol ; 21: 377-423, 2003.
Artigo em Inglês | MEDLINE | ID: mdl-12543935

RESUMO

Large DNA viruses defend against hostile assault executed by the host immune system by producing an array of gene products that systematically sabotage key components of the inflammatory response. Poxviruses target many of the primary mediators of innate immunity including interferons, tumor necrosis factors, interleukins, complement, and chemokines. Poxviruses also manipulate a variety of intracellular signal transduction pathways such as the apoptotic response. Many of the poxvirus genes that disrupt these pathways have been hijacked directly from the host immune system, while others have demonstrated no clear resemblance to any known host genes. Nonetheless, the immunological targets and the diversity of strategies used by poxviruses to disrupt these host pathways have provided important insights into diverse aspects of immunology, virology, and inflammation. Furthermore, because of their anti-inflammatory nature, many of these poxvirus proteins hold promise as potential therapeutic agents for acute or chronic inflammatory conditions.


Assuntos
Poxviridae/imunologia , Poxviridae/patogenicidade , Animais , Apoptose , Quimiocinas/metabolismo , Proteínas do Sistema Complemento/metabolismo , Citocinas/metabolismo , Genoma Viral , Humanos , Imunidade Celular , Interferons/antagonistas & inibidores , Modelos Imunológicos , Poxviridae/genética , Infecções por Poxviridae/imunologia , Infecções por Poxviridae/virologia , Transdução de Sinais , Proteínas Virais/imunologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...