Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 58
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Front Immunol ; 14: 1093381, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-36911702

RESUMO

Natural killer (NK) cells have an established role in controlling poxvirus infection and there is a growing interest to exploit their capabilities in the context of poxvirus-based oncolytic therapy and vaccination. How NK cells respond to poxvirus-infected cells to become activated is not well established. To address this knowledge gap, we studied the NK cell response to vaccinia virus (VACV) in vivo, using a systemic infection murine model. We found broad alterations in NK cells transcriptional activity in VACV-infected mice, consistent with both direct target cell recognition and cytokine exposure. There were also alterations in the expression levels of specific NK surface receptors (NKRs), including the Ly49 family and SLAM receptors, as well as upregulation of memory-associated NK markers. Despite the latter observation, adoptive transfer of VACV-expercienced NK populations did not confer protection from infection. Comparison with the NK cell response to murine cytomegalovirus (MCMV) infection highlighted common features, but also distinct NK transcriptional programmes initiated by VACV. Finally, there was a clear overlap between the NK transcriptional response in humans vaccinated with an attenuated VACV, modified vaccinia Ankara (MVA), demonstrating conservation between the NK response in these different host species. Overall, this study provides new data about NK cell activation, function, and homeostasis during VACV infection, and may have implication for the design of VACV-based therapeutics.


Assuntos
Poxviridae , Vacínia , Camundongos , Humanos , Animais , Vaccinia virus/fisiologia , Células Matadoras Naturais/metabolismo , Citocinas/metabolismo
2.
Front Microbiol ; 13: 1027015, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36478862

RESUMO

Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), the causative agent of the current coronavirus disease 2019 (COVID-19) pandemic, induces an unbalanced immune response in the host. For instance, the production of type I interferon (IFN) and the response to it, which act as a front-line defense against virus invasion, are inhibited during SARS-CoV-2 infection. In addition, tumor necrosis factor alpha (TNF-α), a proinflammatory cytokine, is upregulated in COVID-19 patients with severe symptoms. Studies on the closely related betacoronavirus, SARS-CoV, showed that viral proteins such as Nsp1, Orf6 and nucleocapsid protein inhibit IFN-ß production and responses at multiple steps. Given the conservation of these proteins between SARS-CoV and SARS-CoV-2, it is not surprising that SARS-CoV-2 deploys similar immune evasion strategies. Here, we carried out a screen to examine the role of individual SARS-CoV-2 proteins in regulating innate immune signaling, such as the activation of transcription factors IRF3 and NF-κB and the response to type I and type II IFN. In addition to established roles of SARS-CoV-2 proteins, we report that SARS-CoV-2 proteins Nsp6 and Orf8 inhibit the type I IFN response but at different stages. Orf6 blocks the translocation of STAT1 and STAT2 into the nucleus, whereas ORF8 inhibits the pathway in the nucleus after STAT1/2 translocation. SARS-CoV-2 Orf6 also suppresses IRF3 activation and TNF-α-induced NF-κB activation.

3.
PLoS Pathog ; 18(6): e1010612, 2022 06.
Artigo em Inglês | MEDLINE | ID: mdl-35727847

RESUMO

The interaction between immune cells and virus-infected targets involves multiple plasma membrane (PM) proteins. A systematic study of PM protein modulation by vaccinia virus (VACV), the paradigm of host regulation, has the potential to reveal not only novel viral immune evasion mechanisms, but also novel factors critical in host immunity. Here, >1000 PM proteins were quantified throughout VACV infection, revealing selective downregulation of known T and NK cell ligands including HLA-C, downregulation of cytokine receptors including IFNAR2, IL-6ST and IL-10RB, and rapid inhibition of expression of certain protocadherins and ephrins, candidate activating immune ligands. Downregulation of most PM proteins occurred via a proteasome-independent mechanism. Upregulated proteins included a decoy receptor for TRAIL. Twenty VACV-encoded PM proteins were identified, of which five were not recognised previously as such. Collectively, this dataset constitutes a valuable resource for future studies on antiviral immunity, host-pathogen interaction, poxvirus biology, vector-based vaccine design and oncolytic therapy.


Assuntos
Doenças Transmissíveis , Poxviridae , Vacínia , Humanos , Evasão da Resposta Imune , Proteínas de Membrana/metabolismo , Vaccinia virus
4.
J Gen Virol ; 101(5): 533-541, 2020 05.
Artigo em Inglês | MEDLINE | ID: mdl-32100702

RESUMO

Vaccinia virus (VACV) strain Western Reserve gene A49L encodes a small intracellular protein with a Bcl-2 fold that is expressed early during infection and has multiple functions. A49 co-precipitates with the E3 ubiquitin ligase ß-TrCP and thereby prevents ubiquitylation and proteasomal degradation of IκBα, and consequently blocks activation of NF-κB. In a similar way, A49 stabilizes ß-catenin, leading to activation of the wnt signalling pathway. However, a VACV strain expressing a mutant A49 that neither co-precipitates with ß-TrCP nor inhibits NF-κB activation, is more virulent than a virus lacking A49, indicating that A49 has another function that also contributes to virulence. Here we demonstrate that gene A49L encodes a second, smaller polypeptide that is expressed via leaky scanning translation from methionine 20 and is unable to block NF-κB activation. Viruses engineered to express either only the large protein or only the small A49 protein both have lower virulence than wild-type virus and greater virulence than an A49L deletion mutant. This demonstrates that the small protein contributes to virulence by an unknown mechanism that is independent of NF-κB inhibition. Despite having a large genome with about 200 genes, this study illustrates how VACV makes efficient use of its coding potential and from gene A49L expresses a protein with multiple functions and multiple proteins with different functions.


Assuntos
Vaccinia virus/genética , Proteínas Virais/genética , Virulência/genética , Animais , Feminino , Células HEK293 , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Inibidor de NF-kappaB alfa/genética , NF-kappa B/genética , Transdução de Sinais/genética , Ubiquitina-Proteína Ligases/genética , Ubiquitinação/genética , Vacínia/virologia , Proteínas Contendo Repetições de beta-Transducina/genética
5.
Redox Biol ; 28: 101361, 2020 01.
Artigo em Inglês | MEDLINE | ID: mdl-31693977

RESUMO

The mechanisms by which the Golgi apparatus (GA) impacts on cell invasion are poorly understood. The human Golgi Anti-Apoptotic Protein (hGAAP, also known as TMBIM4) is a highly conserved Golgi cation channel that modulates intracellular Ca2+ fluxes. Human GAAP is expressed in all human tissues, is essential for cell viability and provides resistance against a range of apoptotic stresses. Furthermore, hGAAP enhances adhesion and cell migration by increasing the turnover of focal adhesions due to activation of store-operated Ca2+ entry. Here, we describe a GA-derived mechanism that controls cell invasion. The overexpression of hGAAP stimulates 3-dimensional proteolytic cell invasion by a mechanism that is dependent on the accumulation of intracellular hydrogen peroxide, which might be produced by the hGAAP-dependent stimulation of mitochondrial respiration. These findings provide new insight into the complex mechanisms by which Ca2+ and reactive oxygen species signaling contribute to cell invasion and to the role of the GA in these processes.


Assuntos
Peróxido de Hidrogênio/metabolismo , Proteínas de Membrana/genética , Mitocôndrias/metabolismo , Neoplasias/genética , Animais , Sinalização do Cálcio , Adesão Celular , Linhagem Celular Tumoral , Movimento Celular , Sobrevivência Celular , Humanos , Células MCF-7 , Camundongos , Invasividade Neoplásica , Transplante de Neoplasias , Neoplasias/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Regulação para Cima
6.
Proc Natl Acad Sci U S A ; 116(24): 11997-12006, 2019 06 11.
Artigo em Inglês | MEDLINE | ID: mdl-31127039

RESUMO

Interferons (IFNs) represent an important host defense against viruses. Type I IFNs induce JAK-STAT signaling and expression of IFN-stimulated genes (ISGs), which mediate antiviral activity. Histone deacetylases (HDACs) perform multiple functions in regulating gene expression and some class I HDACs and the class IV HDAC, HDAC11, influence type I IFN signaling. Here, HDAC4, a class II HDAC, is shown to promote type I IFN signaling and coprecipitate with STAT2. Pharmacological inhibition of class II HDAC activity, or knockout of HDAC4 from HEK-293T and HeLa cells, caused a defective response to IFN-α. This defect in HDAC4-/- cells was rescued by reintroduction of HDAC4 or catalytically inactive HDAC4, but not HDAC1 or HDAC5. ChIP analysis showed HDAC4 was recruited to ISG promoters following IFN stimulation and was needed for binding of STAT2 to these promoters. The biological importance of HDAC4 as a virus restriction factor was illustrated by the observations that (i) the replication and spread of vaccinia virus (VACV) and herpes simplex virus type 1 (HSV-1) were enhanced in HDAC4-/- cells and inhibited by overexpression of HDAC4; and (ii) HDAC4 is targeted for proteasomal degradation during VACV infection by VACV protein C6, a multifunctional IFN antagonist that coprecipitates with HDAC4 and is necessary and sufficient for HDAC4 degradation.


Assuntos
Vírus de DNA/metabolismo , Histona Desacetilases/metabolismo , Interferon Tipo I/metabolismo , Proteínas Repressoras/metabolismo , Transdução de Sinais/fisiologia , Vaccinia virus/metabolismo , Vacínia/metabolismo , Proteínas Virais/metabolismo , Linhagem Celular , Linhagem Celular Tumoral , Células HEK293 , Células HeLa , Herpesvirus Humano 1/metabolismo , Humanos , Vacínia/virologia , Replicação Viral/fisiologia
7.
J Gen Virol ; 99(6): 790-804, 2018 06.
Artigo em Inglês | MEDLINE | ID: mdl-29676720

RESUMO

Poxviruses comprise a group of large dsDNA viruses that include members relevant to human and animal health, such as variola virus, monkeypox virus, cowpox virus and vaccinia virus (VACV). Poxviruses are remarkable for their unique replication cycle, which is restricted to the cytoplasm of infected cells. The independence from the host nucleus requires poxviruses to encode most of the enzymes involved in DNA replication, transcription and processing. Here, we use the CRISPR/Cas9 genome engineering system to induce DNA damage to VACV (strain Western Reserve) genomes. We show that targeting CRISPR/Cas9 to essential viral genes limits virus replication efficiently. Although VACV is a strictly cytoplasmic pathogen, we observed extensive viral genome editing at the target site; this is reminiscent of a non-homologous end-joining DNA repair mechanism. This pathway was not dependent on the viral DNA ligase, but critically involved the cellular DNA ligase IV. Our data show that DNA ligase IV can act outside of the nucleus to allow repair of dsDNA breaks in poxvirus genomes. This pathway might contribute to the introduction of mutations within the genome of poxviruses and may thereby promote the evolution of these viruses.


Assuntos
Quebras de DNA de Cadeia Dupla , DNA Ligase Dependente de ATP/metabolismo , Reparo do DNA , Genoma Viral , Interações entre Hospedeiro e Microrganismos/genética , Vaccinia virus/genética , Sistemas CRISPR-Cas , Linhagem Celular Tumoral , Citosol/metabolismo , Citosol/virologia , DNA Ligase Dependente de ATP/genética , Replicação do DNA , DNA Viral/genética , Células HEK293 , Humanos , Mutagênese , Vaccinia virus/fisiologia , Replicação Viral/genética
8.
J Gen Virol ; 99(2): 246-252, 2018 02.
Artigo em Inglês | MEDLINE | ID: mdl-29393023

RESUMO

Molluscum contagiosum virus (MCV) causes persistent, benign skin neoplasm in children and adults. MCV is refractive to growth in standard tissue culture and there is no relevant animal model of infection. Here we investigated whether another poxvirus (vaccinia virus; VACV) could be used to examine MCV immunoevasion protein properties in vivo. The MCV MC159L or MC160L genes, which encode NF-κB antagonists, were inserted into an attenuated VACV lacking an NF-κB antagonist (vΔA49), creating vMC159 and vMC160. vMC160 slightly increased vΔA49 virulence in the intranasal and intradermal routes of inoculation. vMC159 infection was less virulent than vΔA49 in both inoculation routes. vMC159-infected ear pinnae did not form lesions, but virus replication still occurred. Thus, the lack of lesions was not due to abortive virus replication. This system provides a new approach to examine MCV immunoevasion proteins within the context of a complete and complex immune system.


Assuntos
Vírus do Molusco Contagioso/imunologia , NF-kappa B/antagonistas & inibidores , Vaccinia virus/patogenicidade , Proteínas Virais/administração & dosagem , Administração Intranasal , Animais , Criança , Feminino , Humanos , Injeções Intradérmicas , Camundongos Endogâmicos BALB C , Vírus do Molusco Contagioso/genética , Proteínas Virais/imunologia , Virulência
9.
Open Biol ; 7(5)2017 05.
Artigo em Inglês | MEDLINE | ID: mdl-28469007

RESUMO

Golgi anti-apoptotic protein (GAAP), also known as transmembrane Bax inhibitor-1 motif-containing 4 (TMBIM4) or Lifeguard 4 (Lfg4), shares remarkable amino acid conservation with orthologues throughout eukaryotes, prokaryotes and some orthopoxviruses, suggesting a highly conserved function. GAAPs regulate Ca2+ levels and fluxes from the Golgi and endoplasmic reticulum, confer resistance to a broad range of apoptotic stimuli, promote cell adhesion and migration via the activation of store-operated Ca2+ entry, are essential for the viability of human cells, and affect orthopoxvirus virulence. GAAPs are oligomeric, multi-transmembrane proteins that are resident in Golgi membranes and form cation-selective ion channels that may explain the multiple functions of these proteins. Residues contributing to the ion-conducting pore have been defined and provide the first clues about the mechanistic link between these very different functions of GAAP. Although GAAPs are naturally oligomeric, they can also function as monomers, a feature that distinguishes them from other virus-encoded ion channels that must oligomerize for function. This review summarizes the known functions of GAAPs and discusses their potential importance in disease.


Assuntos
Cálcio/metabolismo , Canais Iônicos/metabolismo , Proteínas de Membrana/metabolismo , Neoplasias/metabolismo , Animais , Apoptose , Camelus , Retículo Endoplasmático/metabolismo , Complexo de Golgi/metabolismo , Humanos , Proteínas de Membrana/química , Proteínas de Membrana/genética , Neoplasias/patologia
10.
Immunol Lett ; 186: 68-80, 2017 06.
Artigo em Inglês | MEDLINE | ID: mdl-28366525

RESUMO

Regulated cell death is a powerful anti-viral mechanism capable of aborting the virus replicative cycle and alerting neighbouring cells to the threat of infection. The biological importance of regulated cell death is illustrated by the rich repertoire of host signalling cascades causing cell death and by the multiple strategies exhibited by viruses to block death signal transduction and preserve cell viability. Vaccinia virus (VACV), a poxvirus and the vaccine used to eradicate smallpox, encodes multiple proteins that interfere with apoptotic, necroptotic and pyroptotic signalling. Here the current knowledge on cell death pathways and how VACV proteins interact with them is reviewed. Studying the mechanisms evolved by VACV to counteract host programmed cell death has implications for its successful use as a vector for vaccination and as an oncolytic agent against cancer.


Assuntos
Evasão da Resposta Imune , Vaccinia virus/fisiologia , Vacínia/imunologia , Animais , Evolução Biológica , Morte Celular , Vetores Genéticos , Interações Hospedeiro-Patógeno , Humanos , Transdução de Sinais
11.
PLoS Pathog ; 11(9): e1005151, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-26334635

RESUMO

Vaccinia virus (VACV) is the prototypic orthopoxvirus and the vaccine used to eradicate smallpox. Here we show that VACV strain Western Reserve protein 169 is a cytoplasmic polypeptide expressed early during infection that is excluded from virus factories and inhibits the initiation of cap-dependent and cap-independent translation. Ectopic expression of protein 169 causes the accumulation of 80S ribosomes, a reduction of polysomes, and inhibition of protein expression deriving from activation of multiple innate immune signaling pathways. A virus lacking 169 (vΔ169) replicates and spreads normally in cell culture but is more virulent than parental and revertant control viruses in intranasal and intradermal murine models of infection. Intranasal infection by vΔ169 caused increased pro-inflammatory cytokines and chemokines, infiltration of pulmonary leukocytes, and lung weight. These alterations in innate immunity resulted in a stronger CD8+ T-cell memory response and better protection against virus challenge. This work illustrates how inhibition of host protein synthesis can be a strategy for virus suppression of innate and adaptive immunity.


Assuntos
Imunidade Adaptativa , Interações Hospedeiro-Patógeno , Imunidade Inata , Iniciação Traducional da Cadeia Peptídica , Vaccinia virus/fisiologia , Vacínia/virologia , Proteínas Virais/metabolismo , Animais , Linfócitos T CD8-Positivos/imunologia , Linfócitos T CD8-Positivos/metabolismo , Linfócitos T CD8-Positivos/patologia , Linfócitos T CD8-Positivos/virologia , Linhagem Celular , Feminino , Deleção de Genes , Regulação da Expressão Gênica , Humanos , Memória Imunológica , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Proteínas Recombinantes de Fusão/metabolismo , Proteínas Recombinantes/metabolismo , Subunidades Ribossômicas Maiores de Eucariotos/metabolismo , Subunidades Ribossômicas Menores de Eucariotos/metabolismo , Vacínia/imunologia , Vacínia/metabolismo , Vacínia/patologia , Vaccinia virus/imunologia , Vaccinia virus/patogenicidade , Proteínas Virais/genética , Virulência
12.
J Biol Chem ; 290(10): 5991-6002, 2015 Mar 06.
Artigo em Inglês | MEDLINE | ID: mdl-25605733

RESUMO

Vaccinia virus (VACV) encodes several proteins that inhibit activation of the proinflammatory transcription factor nuclear factor κB (NF-κB). VACV protein A49 prevents translocation of NF-κB to the nucleus by sequestering cellular ß-TrCP, a protein required for the degradation of the inhibitor of κB. A49 does not share overall sequence similarity with any protein of known structure or function. We solved the crystal structure of A49 from VACV Western Reserve to 1.8 Å resolution and showed, surprisingly, that A49 has the same three-dimensional fold as Bcl-2 family proteins despite lacking identifiable sequence similarity. Whereas Bcl-2 family members characteristically modulate cellular apoptosis, A49 lacks a surface groove suitable for binding BH3 peptides and does not bind proapoptotic Bcl-2 family proteins Bax or Bak. The N-terminal 17 residues of A49 do not adopt a single well ordered conformation, consistent with their proposed role in binding ß-TrCP. Whereas pairs of A49 molecules interact symmetrically via a large hydrophobic surface in crystallo, A49 does not dimerize in solution or in cells, and we propose that this hydrophobic interaction surface may mediate binding to a yet undefined cellular partner. A49 represents the eleventh VACV Bcl-2 family protein and, despite these proteins sharing very low sequence identity, structure-based phylogenetic analysis shows that all poxvirus Bcl-2 proteins are structurally more similar to each other than they are to any cellular or herpesvirus Bcl-2 proteins. This is consistent with duplication and diversification of a single BCL2 family gene acquired by an ancestral poxvirus.


Assuntos
Imunidade Inata/genética , Filogenia , Proteínas Proto-Oncogênicas c-bcl-2/genética , Proteínas Virais/química , Apoptose/genética , Cristalografia por Raios X , Células HEK293 , Humanos , NF-kappa B/genética , NF-kappa B/metabolismo , Conformação Proteica , Dobramento de Proteína , Vacínia/genética , Vacínia/virologia , Vaccinia virus/química , Vaccinia virus/genética , Vaccinia virus/patogenicidade , Proteínas Virais/genética
13.
J Gen Virol ; 95(Pt 12): 2757-2768, 2014 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-25090990

RESUMO

Vaccinia virus (VACV) is a large dsDNA virus encoding ~200 proteins, several of which inhibit apoptosis. Here, a comparative study of anti-apoptotic proteins N1, F1, B13 and Golgi anti-apoptotic protein (GAAP) in isolation and during viral infection is presented. VACVs strains engineered to lack each gene separately still blocked apoptosis to some degree because of functional redundancy provided by the other anti-apoptotic proteins. To overcome this redundancy, we inserted each gene separately into a VACV strain (vv811) that lacked all these anti-apoptotic proteins and that induced apoptosis efficiently during infection. Each protein was also expressed in cells using lentivirus vectors. In isolation, each VACV protein showed anti-apoptotic activity in response to specific stimuli, as measured by immunoblotting for cleaved poly(ADP ribose) polymerase-1 and caspase-3 activation. Of the proteins tested, B13 was the most potent inhibitor, blocking both intrinsic and extrinsic stimuli, whilst the activity of the other proteins was largely restricted to inhibition of intrinsic stimuli. In addition, B13 and F1 were effective blockers of apoptosis induced by vv811 infection. Finally, whilst differences in induction of apoptosis were barely detectable during infection with VACV strain Western Reserve compared with derivative viruses lacking individual anti-apoptotic genes, several of these proteins reduced activation of caspase-3 during infection by vv811 strains expressing these proteins. These results illustrated that vv811 was a useful tool to determine the role of VACV proteins during infection and that whilst all of these proteins have some anti-apoptotic activity, B13 was the most potent.


Assuntos
Vaccinia virus/metabolismo , Proteínas Virais/metabolismo , Apoptose , Linhagem Celular , Linhagem Celular Tumoral , Regulação Viral da Expressão Gênica , Humanos , Osteossarcoma , Proteínas Virais/genética
14.
J Gen Virol ; 95(Pt 9): 2038-2049, 2014 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-24914067

RESUMO

Ubiquitylation is a covalent post-translational modification that regulates protein stability and is involved in many biological functions. Proteins may be modified with mono-ubiquitin or ubiquitin chains. Viruses have evolved multiple mechanisms to perturb the cell ubiquitin system and manipulate it to their own benefit. Here, we report ubiquitylation of vaccinia virus (VACV) protein N1. N1 is an inhibitor of the nuclear factor NF-κB and apoptosis that contributes to virulence, has a Bcl-2-like fold, and is highly conserved amongst orthopoxviruses. The interaction between N1 and ubiquitin occurs at endogenous protein levels during VACV infection and following ectopic expression of N1. Biochemical analysis demonstrated that N1 is covalently ubiquitylated, and heterodimers of ubiquitylated and non-ubiquitylated N1 monomers were identified, suggesting that ubiquitylation does not inhibit N1 dimerization. Studies with other VACV Bcl-2 proteins, such as C6 or B14, revealed that although these proteins also interact with ubiquitin, these interactions are non-covalent. Finally, mutagenesis of N1 showed that ubiquitylation occurs in a conventional lysine-dependent manner at multiple acceptor sites because only an N1 allele devoid of lysine residues remained unmodified. Taken together, we described a previously uncharacterized modification of the VACV protein N1 that provided a new layer of complexity to the biology of this virulence factor, and provided another example of the intricate interplay between poxviruses and the host ubiquitin system.


Assuntos
Ubiquitinação/genética , Vaccinia virus/patogenicidade , Proteínas Virais/genética , Proteínas Virais/metabolismo , Animais , Apoptose , Linhagem Celular , Chlorocebus aethiops , Dimerização , Células HEK293 , Humanos , Lisina/química , Macrófagos/virologia , Camundongos , Mutação , NF-kappa B/antagonistas & inibidores , Ubiquitinação/fisiologia , Vacínia , Fatores de Virulência/genética
15.
Adv Exp Med Biol ; 808: 65-76, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24595611

RESUMO

Vaccinia virus (VACV) is the live vaccine that was used to eradicate smallpox, a feat achieved in 1977 and certified by the World Health Organization in 1980. Since 1980, research with VACV has continued in part because of the development of techniques to genetically manipulate VACV and create live VACV strains expressing foreign genes. These recombinant VACVs can be used as live vaccines against other infectious diseases and cancers, and as a powerful tool to study virus pathogenesis, immunology, cell biology, and virus-host interactions. This short article describes two examples of how enduring interest in VACV has revealed new features of VACV biology and the immune system.


Assuntos
Interações Hospedeiro-Patógeno , Evasão da Resposta Imune , Vacina Antivariólica/imunologia , Varíola/prevenção & controle , Vacinação , Vaccinia virus/imunologia , Imunidade Adaptativa , Erradicação de Doenças , Engenharia Genética , Humanos , Imunidade Inata , Varíola/imunologia , Varíola/virologia , Vacina Antivariólica/administração & dosagem , Transgenes , Vacinas Atenuadas , Replicação Viral
16.
J Virol ; 88(6): 3092-102, 2014 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-24371075

RESUMO

The transcription factor nuclear factor kappa light-chain enhancer of activated B cells (NF-κB) plays a critical role in host defense against viral infection by inducing the production of proinflammatory mediators and type I interferon. Consequently, viruses have evolved many mechanisms to block its activation. The poxvirus vaccinia virus (VACV) encodes numerous inhibitors of NF-κB activation that target multiple points in the signaling pathway. A derivative of VACV strain Copenhagen, called vv811, lacking 55 open reading frames in the left and right terminal regions of the genome was reported to still inhibit NF-κB activation downstream of tumor necrosis factor alpha (TNF-α) and interleukin-1ß (IL-1ß), suggesting the presence of one or more additional inhibitors. In this study, we constructed a recombinant vv811 lacking the recently described NF-κB inhibitor A49 (vv811ΔA49), yielding a virus that lacked all currently described inhibitors downstream of TNF-α and IL-1ß. Unlike vv811, vv811ΔA49 no longer inhibited degradation of the phosphorylated inhibitor of κBα and p65 translocated into the nucleus. However, despite this translocation, vv811ΔA49 still inhibited TNF-α- and IL-1ß-induced NF-κB-dependent reporter gene expression and the transcription and production of cytokines induced by these agonists. This inhibition did not require late viral gene expression. These findings indicate the presence of another inhibitor of NF-κB that is expressed early during infection and acts by a novel mechanism downstream of p65 translocation into the nucleus.


Assuntos
NF-kappa B/metabolismo , Fator de Transcrição RelA/metabolismo , Vaccinia virus/fisiologia , Vacínia/genética , Vacínia/metabolismo , Núcleo Celular/genética , Núcleo Celular/metabolismo , Regulação para Baixo , Regulação da Expressão Gênica , Humanos , Interleucina-1/genética , Interleucina-1/metabolismo , NF-kappa B/genética , Transporte Proteico , Fator de Transcrição RelA/genética , Fator de Necrose Tumoral alfa/genética , Fator de Necrose Tumoral alfa/metabolismo , Vacínia/virologia , Vaccinia virus/genética
17.
Proc Natl Acad Sci U S A ; 110(30): 12444-9, 2013 Jul 23.
Artigo em Inglês | MEDLINE | ID: mdl-23836663

RESUMO

Viruses have evolved sophisticated strategies to exploit host cell function for their benefit. Here we show that under physiologically normal oxygen levels (normoxia) vaccinia virus (VACV) infection leads to a rapid stabilization of hypoxia-inducible factor (HIF)-1α, its translocation into the nucleus and the activation of HIF-responsive genes, such as vascular endothelial growth factor (VEGF), glucose transporter-1, and pyruvate dehydrogenase kinase-1. HIF-1α stabilization is mediated by VACV protein C16 that binds the human oxygen sensing enzyme prolyl-hydroxylase domain containing protein (PHD)2 and thereby inhibits PHD2-dependent hydroxylation of HIF-1α. The binding between C16 and PHD2 is direct and specific, and ectopic expression of C16 alone induces transcription of HIF-1α responsive genes. Conversely, a VACV strain lacking the gene for C16, C16L, is unable to induce HIF-1α stabilization. Interestingly, the N-terminal region of C16 is predicted to have a PHD2-like structural fold but lacks the catalytic active site residues of PHDs. The induction of a hypoxic response by VACV is reminiscent of the biochemical consequences of solid tumor formation, and illustrates a poxvirus strategy for manipulation of cellular gene expression and biochemistry.


Assuntos
Hipóxia Celular/fisiologia , Vaccinia virus/fisiologia , Sequência de Aminoácidos , Células HEK293 , Humanos , Hidroxilação , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Prolina Dioxigenases do Fator Induzível por Hipóxia , Dados de Sequência Molecular , Pró-Colágeno-Prolina Dioxigenase/metabolismo , Homologia de Sequência de Aminoácidos , Proteínas Virais/química , Proteínas Virais/metabolismo
18.
J Gen Virol ; 94(Pt 9): 2070-2081, 2013 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-23761407

RESUMO

Vaccinia virus (VACV) expresses many proteins that are non-essential for virus replication but promote virulence by inhibiting components of the host immune response to infection. These immunomodulators include a family of proteins that have, or are predicted to have, a structure related to the B-cell lymphoma (Bcl)-2 protein. Five members of the VACV Bcl-2 family (N1, B14, A52, F1 and K7) have had their crystal structure solved, others have been characterized and a function assigned (C6, A46), and others are predicted to be Bcl-2 proteins but are uncharacterized hitherto (N2, B22, C1). Data presented here show that N2 is a nuclear protein that is expressed early during infection and inhibits the activation of interferon regulatory factor (IRF)3. Consistent with its nuclear localization, N2 inhibits IRF3 downstream of the TANK-binding kinase (TBK)-1 and after IRF3 translocation into the nucleus. A mutant VACV strain Western Reserve lacking the N2L gene (vΔN2) showed normal replication and spread in cultured cells compared to wild-type parental (vN2) and revertant (vN2-rev) viruses, but was attenuated in two murine models of infection. After intranasal infection, the vΔN2 mutant induced lower weight loss and signs of illness, and virus was cleared more rapidly from the infected tissue. In the intradermal model of infection, vΔN2 induced smaller lesions that were resolved more rapidly. In summary, the N2 protein is an intracellular virulence factor that inhibits IRF3 activity in the nucleus.


Assuntos
Interações Hospedeiro-Patógeno , Fator Regulador 3 de Interferon/antagonistas & inibidores , Vaccinia virus/patogenicidade , Proteínas Virais/metabolismo , Fatores de Virulência/metabolismo , Animais , Modelos Animais de Doenças , Feminino , Deleção de Genes , Camundongos , Camundongos Endogâmicos BALB C , Vacínia/patologia , Vacínia/virologia , Vaccinia virus/genética , Vaccinia virus/fisiologia , Virulência , Replicação Viral
19.
J Gen Virol ; 94(Pt 7): 1647-1657, 2013 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-23580427

RESUMO

Vaccinia virus (VACV) encodes many proteins that antagonize the innate immune system including a family of intracellular proteins with a B-cell lymphoma (Bcl)-2-like structure. One of these Bcl-2 proteins called K7 binds Toll-like receptor-adaptor proteins and the DEAD-box RNA helicase DDX3 and thereby inhibits the activation of NF-κB and interferon regulatory factor 3. However, the contribution of K7 to virus virulence is not known. Here a VACV lacking the K7R gene (vΔK7) was constructed and compared with control viruses that included a plaque purified wt (vK7), a revertant with the K7R gene reinserted (vK7-rev) and a frame-shifted virus in which the translational initiation codon was mutated to prevent K7 protein expression (vK7-fs). Data presented show that loss of K7 does not affect virus replication in cell culture or in vivo; however, viruses lacking the K7 protein were less virulent than controls in murine intradermal (i.d.) and intranasal (i.n.) infection models and there was an altered acute immune response to infection. In the i.d. model, vΔK7 induced smaller lesions than controls, and after i.n. infection vΔK7 induced a reduced weight loss and signs of illness, and more rapid clearance of virus from infected tissue. Concomitantly, the intrapulmonary innate immune response to infection with vΔK7 showed increased infiltration of NK cells and CD8⁺ T-cells, enhanced MHC class II expression by macrophages, and enhanced cytolysis of target cells by NK cells and VACV-specific CD8⁺ T-cells. Thus protein K7 is a virulence factor that affects the acute immune response to infection.


Assuntos
Imunidade Inata/efeitos dos fármacos , Vaccinia virus/patogenicidade , Vacínia/imunologia , Vacínia/patologia , Proteínas Virais/metabolismo , Fatores de Virulência/metabolismo , Animais , Linhagem Celular , Derme/imunologia , Derme/patologia , Derme/virologia , Feminino , Células HeLa , Humanos , Células L , Pulmão/imunologia , Pulmão/patologia , Pulmão/virologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Vacínia/virologia , Vaccinia virus/imunologia , Proteínas Virais/genética , Proteínas Virais/farmacologia , Virulência , Fatores de Virulência/genética , Fatores de Virulência/farmacologia
20.
J Biol Chem ; 288(18): 13057-67, 2013 May 03.
Artigo em Inglês | MEDLINE | ID: mdl-23508950

RESUMO

Golgi anti-apoptotic proteins (GAAPs) are hydrophobic proteins resident in membranes of the Golgi complex. They protect cells from a range of apoptotic stimuli, reduce the Ca(2+) content of intracellular stores, and regulate Ca(2+) fluxes. GAAP was discovered in camelpox virus, but it is highly conserved throughout evolution and encoded by all eukaryote genomes examined. GAAPs are part of the transmembrane Bax inhibitor-containing motif (TMBIM) family that also includes other anti-apoptotic and Ca(2+)-modulating membrane proteins. Most TMBIM members show multiple bands when analyzed by SDS-PAGE, suggesting that they may be oligomeric. However, the molecular mechanisms of oligomerization, the native state of GAAPs in living cells and the functional significance of oligomerization have not been addressed. TMBIM members are thought to have evolved from an ancestral GAAP. Two different GAAPs, human (h) and viral (v)GAAP were therefore selected as models to examine oligomerization of TMBIM family members. We show that both hGAAP and vGAAP in their native states form oligomers and that oligomerization is pH-dependent. Surprisingly, hGAAP and vGAAP do not share the same oligomerization mechanism. Oligomerization of hGAAP is independent of cysteines, but oligomerization of vGAAP depends on cysteines 9 and 60. A mutant vGAAP that is unable to oligomerize revealed that monomeric vGAAP retains both its anti-apoptotic function and its effect on intracellular Ca(2+) stores. In conclusion, GAAP can oligomerize in a pH-regulated manner, and monomeric GAAP is functional.


Assuntos
Apoptose , Cálcio/metabolismo , Proteínas Inibidoras de Apoptose/metabolismo , Proteínas de Membrana/metabolismo , Orthopoxvirus/metabolismo , Multimerização Proteica , Proteínas Virais/metabolismo , Substituição de Aminoácidos , Células HeLa , Humanos , Concentração de Íons de Hidrogênio , Proteínas Inibidoras de Apoptose/genética , Proteínas de Membrana/genética , Mutação de Sentido Incorreto , Orthopoxvirus/genética , Proteínas Virais/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA