Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 28
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
J Gen Virol ; 105(4)2024 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-38687323

RESUMO

The human cytomegalovirus (HCMV) pUS2 glycoprotein exploits the host's endoplasmic reticulum (ER)-associated degradation (ERAD) pathway to degrade major histocompatibility complex class I (MHC-I) and prevent antigen presentation. Beyond MHC-I, pUS2 has been shown to target a range of cellular proteins for degradation, preventing their cell surface expression. Here we have identified a novel pUS2 target, ER-resident protein lectin mannose binding 2 like (LMAN2L). pUS2 expression was both necessary and sufficient for the downregulation of LMAN2L, which was dependent on the cellular E3 ligase TRC8. Given the hypothesized role of LMAN2L in the trafficking of glycoproteins, we employed proteomic plasma membrane profiling to measure LMAN2L-dependent changes at the cell surface. A known pUS2 target, integrin alpha-6 (ITGA6), was downregulated from the surface of LMAN2L-deficient cells, but not other integrins. Overall, these results suggest a novel strategy of pUS2-mediated protein degradation whereby pUS2 targets LMAN2L to impair trafficking of ITGA6. Given that pUS2 can directly target other integrins, we propose that this single viral protein may exhibit both direct and indirect mechanisms to downregulate key cell surface molecules.


Assuntos
Citomegalovirus , Retículo Endoplasmático , Proteínas do Envelope Viral , Proteínas Virais , Humanos , Citomegalovirus/genética , Citomegalovirus/metabolismo , Retículo Endoplasmático/metabolismo , Retículo Endoplasmático/virologia , Proteínas Virais/metabolismo , Proteínas Virais/genética , Ubiquitina-Proteína Ligases/metabolismo , Ubiquitina-Proteína Ligases/genética , Proteólise , Proteínas de Membrana/metabolismo , Proteínas de Membrana/genética , Lectinas de Ligação a Manose/metabolismo , Lectinas de Ligação a Manose/genética , Degradação Associada com o Retículo Endoplasmático , Interações Hospedeiro-Patógeno , Membrana Celular/metabolismo , Membrana Celular/virologia
2.
Proc Natl Acad Sci U S A ; 120(33): e2303155120, 2023 08 15.
Artigo em Inglês | MEDLINE | ID: mdl-37561786

RESUMO

Human cytomegalovirus (HCMV) is a major human pathogen whose life-long persistence is enabled by its remarkable capacity to systematically subvert host immune defenses. In exploring the finding that HCMV infection up-regulates tumor necrosis factor receptor 2 (TNFR2), a ligand for the pro-inflammatory antiviral cytokine TNFα, we found that the underlying mechanism was due to targeting of the protease, A Disintegrin And Metalloproteinase 17 (ADAM17). ADAM17 is the prototype 'sheddase', a family of proteases that cleaves other membrane-bound proteins to release biologically active ectodomains into the supernatant. HCMV impaired ADAM17 surface expression through the action of two virally-encoded proteins in its UL/b' region, UL148 and UL148D. Proteomic plasma membrane profiling of cells infected with an HCMV double-deletion mutant for UL148 and UL148D with restored ADAM17 expression, combined with ADAM17 functional blockade, showed that HCMV stabilized the surface expression of 114 proteins (P < 0.05) in an ADAM17-dependent fashion. These included reported substrates of ADAM17 with established immunological functions such as TNFR2 and jagged1, but also numerous unreported host and viral targets, such as nectin1, UL8, and UL144. Regulation of TNFα-induced cytokine responses and NK inhibition during HCMV infection were dependent on this impairment of ADAM17. We therefore identify a viral immunoregulatory mechanism in which targeting a single sheddase enables broad regulation of multiple critical surface receptors, revealing a paradigm for viral-encoded immunomodulation.


Assuntos
Citomegalovirus , Fator de Necrose Tumoral alfa , Humanos , Citomegalovirus/fisiologia , Fator de Necrose Tumoral alfa/metabolismo , Proteoma/metabolismo , Receptores Tipo II do Fator de Necrose Tumoral/metabolismo , Proteômica , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Citocinas/metabolismo , Membrana Celular/metabolismo , Metaloproteases/metabolismo , Proteína ADAM17/genética , Proteína ADAM17/metabolismo , Glicoproteínas de Membrana/metabolismo , Proteínas Virais/metabolismo
3.
J Virol ; 97(3): e0184622, 2023 03 30.
Artigo em Inglês | MEDLINE | ID: mdl-36916924

RESUMO

Cellular antiviral factors that recognize viral nucleic acid can inhibit virus replication. These include the zinc finger antiviral protein (ZAP), which recognizes high CpG dinucleotide content in viral RNA. Here, we investigated the ability of ZAP to inhibit the replication of human cytomegalovirus (HCMV). Depletion of ZAP or its cofactor KHNYN increased the titer of the high-passage HCMV strain AD169 but had little effect on the titer of the low-passage strain Merlin. We found no obvious difference in expression of several viral proteins between AD169 and Merlin in ZAP knockdown cells, but observed a larger increase in infectious virus in AD169 compared to Merlin in the absence of ZAP, suggesting that ZAP inhibited events late in AD169 replication. In addition, there was no clear difference in the CpG abundance of AD169 and Merlin RNAs, indicating that genomic content of the two virus strains was unlikely to be responsible for differences in their sensitivity to ZAP. Instead, we observed less ZAP expression in Merlin-infected cells late in replication compared to AD169-infected cells, which may be related to different abilities of the two virus strains to regulate interferon signaling. Therefore, there are strain-dependent differences in the sensitivity of HCMV to ZAP, and the ability of low-passage HCMV strain Merlin to evade inhibition by ZAP is likely related to its ability to regulate interferon signaling, not the CpG content of RNAs produced from its genome. IMPORTANCE Determining the function of cellular antiviral factors can inform our understanding of virus replication. The zinc finger antiviral protein (ZAP) can inhibit the replication of diverse viruses. Here, we examined ZAP interaction with the DNA virus human cytomegalovirus (HCMV). We found HCMV strain-dependent differences in the ability of ZAP to influence HCMV replication, which may be related to the interaction of HCMV strains with the type I interferon system. These observations affect our current understanding of how ZAP restricts HCMV and how HCMV interacts with the type I interferon system.


Assuntos
Citomegalovirus , Interferon Tipo I , Humanos , Citomegalovirus/metabolismo , Neurofibromina 2/metabolismo , Neurofibromina 2/farmacologia , Proteínas de Ligação a RNA/metabolismo , Replicação Viral/fisiologia , Antivirais/farmacologia , Interferon Tipo I/metabolismo , Dedos de Zinco
4.
Front Immunol ; 14: 1107497, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-36845106

RESUMO

Introduction: The antigen presentation molecule MHC class I related protein-1 (MR1) is best characterized by its ability to present bacterially derived metabolites of vitamin B2 biosynthesis to mucosal-associated invariant T-cells (MAIT cells). Methods: Through in vitro human cytomegalovirus (HCMV) infection in the presence of MR1 ligand we investigate the modulation of MR1 expression. Using coimmunoprecipitation, mass spectrometry, expression by recombinant adenovirus and HCMV deletion mutants we investigate HCMV gpUS9 and its family members as potential regulators of MR1 expression. The functional consequences of MR1 modulation by HCMV infection are explored in coculture activation assays with either Jurkat cells engineered to express the MAIT cell TCR or primary MAIT cells. MR1 dependence in these activation assays is established by addition of MR1 neutralizing antibody and CRISPR/Cas-9 mediated MR1 knockout. Results: Here we demonstrate that HCMV infection efficiently suppresses MR1 surface expression and reduces total MR1 protein levels. Expression of the viral glycoprotein gpUS9 in isolation could reduce both cell surface and total MR1 levels, with analysis of a specific US9 HCMV deletion mutant suggesting that the virus can target MR1 using multiple mechanisms. Functional assays with primary MAIT cells demonstrated the ability of HCMV infection to inhibit bacterially driven, MR1-dependent activation using both neutralizing antibodies and engineered MR1 knockout cells. Discussion: This study identifies a strategy encoded by HCMV to disrupt the MR1:MAIT cell axis. This immune axis is less well characterized in the context of viral infection. HCMV encodes hundreds of proteins, some of which regulate the expression of antigen presentation molecules. However the ability of this virus to regulate the MR1:MAIT TCR axis has not been studied in detail.


Assuntos
Células T Invariantes Associadas à Mucosa , Humanos , Antígenos de Histocompatibilidade Classe I , Citomegalovirus/metabolismo , Antígenos de Histocompatibilidade Menor , Receptores de Antígenos de Linfócitos T/metabolismo
5.
Proc Natl Acad Sci U S A ; 119(6)2022 02 08.
Artigo em Inglês | MEDLINE | ID: mdl-35105802

RESUMO

Human cytomegalovirus (HCMV) is an important human pathogen and a paradigm of viral immune evasion, targeting intrinsic, innate, and adaptive immunity. We have employed two orthogonal multiplexed tandem mass tag-based proteomic screens to identify host proteins down-regulated by viral factors expressed during the latest phases of viral infection. This approach revealed that the HIV-1 restriction factor Schlafen-11 (SLFN11) was degraded by the poorly characterized, late-expressed HCMV protein RL1, via recruitment of the Cullin4-RING E3 Ubiquitin Ligase (CRL4) complex. SLFN11 potently restricted HCMV infection, inhibiting the formation and spread of viral plaques. Overall, we show that a restriction factor previously thought only to inhibit RNA viruses additionally restricts HCMV. We define the mechanism of viral antagonism and also describe an important resource for revealing additional molecules of importance in antiviral innate immunity and viral immune evasion.


Assuntos
Infecções por Citomegalovirus/imunologia , Citomegalovirus/imunologia , Evasão da Resposta Imune , Proteínas Nucleares/imunologia , Proteólise , Proteínas do Envelope Viral/imunologia , Citomegalovirus/genética , Infecções por Citomegalovirus/genética , Humanos , Proteínas Nucleares/genética , Complexos Ubiquitina-Proteína Ligase/genética , Complexos Ubiquitina-Proteína Ligase/imunologia , Proteínas do Envelope Viral/genética
6.
Hum Gene Ther ; 33(17-18): 990-1001, 2022 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-35196879

RESUMO

Recombinant adenovirus vectors enable highly efficient gene delivery in vitro and in vivo. As a result, they are widely used in gene therapy, vaccination, and anticancer applications. We have previously developed the AdZ vector system, which uses recombineering to permit high-throughput cloning of transgenes into Adenovirus vectors, simplifies alteration of the vector backbone, and enables rapid recovery of infectious virus, even if a transgene is incompatible with vector replication. In this study, we adapt this vector system to enable high-throughput cloning of sequences for CRISPR/Cas9 editing. Vectors were optimized to ensure efficient cloning, and high editing efficiency using spCas9 and single guide RNA (sgRNA) sequences in a single vector. Using a multiplicity of infection of 50, knockout efficiencies of up to 80% could be achieved with a single sgRNA. Vectors were further enhanced by altering the spCas9 sequence to match that of SniperCas9, which has reduced off-target activity, but maintains on-target efficiency, and by applying modifications to the sgRNA sequence that significantly enhance editing efficiency. Thus, the AdZ-CRISPR vectors offer highly efficient knockout, even in hard to transfect cells, and enables large-scale CRISPR/Cas9 projects to be undertaken easily and quickly.


Assuntos
Sistemas CRISPR-Cas , RNA Guia de Cinetoplastídeos , Adenoviridae/genética , Clonagem Molecular , Edição de Genes , Vetores Genéticos/genética , RNA Guia de Cinetoplastídeos/genética
7.
Artigo em Inglês | MEDLINE | ID: mdl-32793512

RESUMO

Long non-coding RNAs (lncRNAs) are transcripts of >200 nucleotides that are not translated into functional proteins. Cellular lncRNAs have been shown to act as regulators by interacting with target nucleic acids or proteins and modulating their activities. We investigated the role of RNA1.2, which is one of four major lncRNAs expressed by human cytomegalovirus (HCMV), by comparing the properties of parental virus in vitro with those of deletion mutants lacking either most of the RNA1.2 gene or only the TATA element of the promoter. In comparison with parental virus, these mutants exhibited no growth defects and minimal differences in viral gene expression in human fibroblasts. In contrast, 76 cellular genes were consistently up- or down-regulated by the mutants at both the RNA and protein levels at 72 h after infection. Differential expression of the gene most highly upregulated by the mutants (Tumor protein p63-regulated gene 1-like protein; TPRG1L) was confirmed at both levels by RT-PCR and immunoblotting. Consistent with the known ability of TPRG1L to upregulate IL-6 expression via NF-κB stimulation, RNA1.2 mutant-infected fibroblasts were observed to upregulate IL-6 in addition to TPRG1L. Comparable surface expression of TNF receptors and responsiveness to TNF-α in cells infected by the parental and mutant viruses indicated that activation of signaling by TNF-α is not involved in upregulation of IL-6 by the mutants. In contrast, inhibition of NF-κB activity and knockdown of TPRG1L expression reduced the extracellular release of IL-6 by RNA1.2 mutant-infected cells, thus demonstrating that upregulation of TPRG1L activates NF-κB. The levels of MCP-1 and CXCL1 transcripts were also increased in RNA1.2 mutant-infected cells, further demonstrating the presence of active NF-κB signaling. These results suggest that RNA1.2 plays a role in manipulating intrinsic NF-κB-dependent cytokine and chemokine release during HCMV infection, thereby impacting downstream immune responses.


Assuntos
Citomegalovirus , Interleucina-6 , NF-kappa B , RNA Longo não Codificante/genética , Células Cultivadas , Citocinas , Citomegalovirus/genética , Fibroblastos/virologia , Humanos , Interleucina-6/genética , RNA Viral/genética
8.
J Gen Virol ; 101(8): 863-872, 2020 08.
Artigo em Inglês | MEDLINE | ID: mdl-32510303

RESUMO

Molluscum contagiosum virus (MCV) is a common cause of benign skin lesions in young children and currently the only endemic human poxvirus. Following the infection of primary keratinocytes in the epidermis, MCV induces the proliferation of infected cells and this results in the production of wart-like growths. Full productive infection is observed only after the infected cells differentiate. During this prolonged replication cycle the virus must avoid elimination by the host immune system. We therefore sought to investigate the function of the two major histocompatibility complex class-I-related genes encoded by the MCV genes mc033 and mc080. Following insertion into a replication-deficient adenovirus vector, codon-optimized versions of mc033 and mc080 were expressed as endoglycosidase-sensitive glycoproteins that localized primarily in the endoplasmic reticulum. MC080, but not MC033, downregulated cell-surface expression of endogenous classical human leucocyte antigen (HLA) class I and non-classical HLA-E by a transporter associated with antigen processing (TAP)-independent mechanism. MC080 exhibited a capacity to inhibit or activate NK cells in autologous assays in a donor-specific manner. MC080 consistently inhibited antigen-specific T cells being activated by peptide-pulsed targets. We therefore propose that MC080 acts to promote evasion of HLA-I-restricted cytotoxic T cells.


Assuntos
Linfócitos T CD8-Positivos/imunologia , Regulação para Baixo/imunologia , Antígenos de Histocompatibilidade Classe I/imunologia , Evasão da Resposta Imune/imunologia , Células Matadoras Naturais/imunologia , Vírus do Molusco Contagioso/imunologia , Apresentação de Antígeno/imunologia , Linhagem Celular , Retículo Endoplasmático/imunologia , Interações Hospedeiro-Patógeno/imunologia , Humanos , Queratinócitos/imunologia , Linfócitos T Citotóxicos/imunologia , Proteínas Virais/imunologia
9.
Clin Cancer Res ; 26(13): 3360-3370, 2020 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-32122920

RESUMO

PURPOSE: Broadly expressed, highly differentiated tumor-associated antigens (TAA) can elicit antitumor immunity. However, vaccines targeting TAAs have demonstrated disappointing clinical results, reflecting poor antigen selection and/or immunosuppressive mechanisms. EXPERIMENTAL DESIGN: Here, a panel of widely expressed, novel colorectal TAAs were identified by performing RNA sequencing of highly purified colorectal tumor cells in comparison with patient-matched colonic epithelial cells; tumor cell purification was essential to reveal these genes. Candidate TAA protein expression was confirmed by IHC, and preexisting T-cell immunogenicity toward these antigens tested. RESULTS: The most promising candidate for further development is DNAJB7 [DnaJ heat shock protein family (Hsp40) member B7], identified here as a novel cancer-testis antigen. It is expressed in many tumors and is strongly immunogenic in patients with cancers originating from a variety of sites. DNAJB7-specific T cells were capable of killing colorectal tumor lines in vitro, and the IFNγ+ response was markedly magnified by control of immunosuppression with cyclophosphamide in patients with cancer. CONCLUSIONS: This study highlights how prior methods that sequence whole tumor fractions (i.e., inclusive of alive/dead stromal cells) for antigen identification may have limitations. Through tumor cell purification and sequencing, novel candidate TAAs have been identified for future immunotherapeutic targeting.


Assuntos
Antígenos de Neoplasias/imunologia , Sequenciamento de Nucleotídeos em Larga Escala , Neoplasias/imunologia , Análise de Sequência de RNA , Antígenos de Neoplasias/genética , Antineoplásicos/farmacologia , Linhagem Celular Tumoral , Citotoxicidade Imunológica , Perfilação da Expressão Gênica/métodos , Regulação Neoplásica da Expressão Gênica , Humanos , Imunofenotipagem , Leucócitos Mononucleares/imunologia , Leucócitos Mononucleares/metabolismo , Neoplasias/genética , Linfócitos T/imunologia , Linfócitos T/metabolismo , Células Tumorais Cultivadas
10.
Elife ; 82019 12 24.
Artigo em Inglês | MEDLINE | ID: mdl-31873071

RESUMO

Human cytomegalovirus (HCMV) extensively modulates host cells, downregulating >900 human proteins during viral replication and degrading ≥133 proteins shortly after infection. The mechanism of degradation of most host proteins remains unresolved, and the functions of many viral proteins are incompletely characterised. We performed a mass spectrometry-based interactome analysis of 169 tagged, stably-expressed canonical strain Merlin HCMV proteins, and two non-canonical HCMV proteins, in infected cells. This identified a network of >3400 virus-host and >150 virus-virus protein interactions, providing insights into functions for multiple viral genes. Domain analysis predicted binding of the viral UL25 protein to SH3 domains of NCK Adaptor Protein-1. Viral interacting proteins were identified for 31/133 degraded host targets. Finally, the uncharacterised, non-canonical ORFL147C protein was found to interact with elements of the mRNA splicing machinery, and a mutational study suggested its importance in viral replication. The interactome data will be important for future studies of herpesvirus infection.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/genética , Infecções por Citomegalovirus/genética , Citomegalovirus/genética , Proteínas Oncogênicas/genética , Proteômica , Citomegalovirus/patogenicidade , Infecções por Citomegalovirus/virologia , Regulação Viral da Expressão Gênica/genética , Interações Hospedeiro-Patógeno/genética , Humanos , Splicing de RNA/genética , RNA Mensageiro/genética , Proteínas Virais/genética , Replicação Viral/genética
11.
Front Immunol ; 9: 2214, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30327650

RESUMO

Human cytomegalovirus (HCMV) is under constant selective pressure from the immune system in vivo. Study of HCMV genes that have been lost in the absence of, or genetically altered by, such selection can focus research toward findings of in vivo significance. We have been particularly interested in the most pronounced change in the highly passaged laboratory strains AD169 and Towne-the deletion of 13-15 kb of sequence (designated the UL/b' region) that encodes up to 22 canonical genes, UL133-UL150. At least 5 genes have been identified in UL/b' that inhibit NK cell function. UL135 suppresses formation of the immunological synapse (IS) by remodeling the actin cytoskeleton, thereby illustrating target cell cooperation in IS formation. UL141 inhibits expression of two activating ligands (CD155, CD112) for the activating receptor CD226 (DNAM-1), and two receptors (TRAIL-R1, R2) for the apoptosis-inducing TRAIL. UL142, ectopically expressed in isolation, and UL148A, target specific MICA allotypes that are ligands for NKG2D. UL148 impairs expression of CD58 (LFA-3), the co-stimulatory cell adhesion molecule for CD2 found on T and NK cells. Outside UL/b', studies on natural variants have shown UL18 mutants change affinity for their inhibitory ligand LIR-1, while mutations in UL40's HLA-E binding peptide differentially drive NKG2C+ NK expansions. Research into HCMV genomic stability and its effect on NK function has provided important insights into virus:host interactions, but future studies will require consideration of genetic variability and the effect of genes expressed in the context of infection to fully understand their in vivo impact.


Assuntos
Infecções por Citomegalovirus/metabolismo , Citomegalovirus/genética , Citomegalovirus/metabolismo , Células Matadoras Naturais/metabolismo , Antígenos de Diferenciação de Linfócitos T/metabolismo , Cromossomos Artificiais Bacterianos/genética , Infecções por Citomegalovirus/prevenção & controle , Variação Genética , Instabilidade Genômica , Antígenos de Histocompatibilidade Classe I/metabolismo , Humanos , Evasão da Resposta Imune , Ativação Linfocitária , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/metabolismo , Receptor Ativador de Fator Nuclear kappa-B/metabolismo , Receptores do Ligante Indutor de Apoptose Relacionado a TNF/metabolismo , Proteínas Virais/genética , Proteínas Virais/metabolismo
12.
Proc Natl Acad Sci U S A ; 115(19): 4998-5003, 2018 05 08.
Artigo em Inglês | MEDLINE | ID: mdl-29691324

RESUMO

CD58 is an adhesion molecule that is known to play a critical role in costimulation of effector cells and is intrinsic to immune synapse structure. Herein, we describe a virally encoded gene that inhibits CD58 surface expression. Human cytomegalovirus (HCMV) UL148 was necessary and sufficient to promote intracellular retention of CD58 during HCMV infection. Blocking studies with antagonistic anti-CD58 mAb and an HCMV UL148 deletion mutant (HCMV∆UL148) with restored CD58 expression demonstrated that the CD2/CD58 axis was essential for the recognition of HCMV-infected targets by CD8+ HCMV-specific cytotoxic T lymphocytes (CTLs). Further, challenge of peripheral blood mononuclear cells ex vivo with HCMV∆UL148 increased both CTL and natural killer (NK) cell degranulation against HCMV-infected cells, including NK-driven antibody-dependent cellular cytotoxicity, showing that UL148 is a modulator of the function of multiple effector cell subsets. Our data stress the effect of HCMV immune evasion functions on shaping the immune response, highlighting the capacity for their potential use in modulating immunity during the development of anti-HCMV vaccines and HCMV-based vaccine vectors.


Assuntos
Linfócitos T CD8-Positivos/imunologia , Infecções por Citomegalovirus/imunologia , Citomegalovirus/imunologia , Evasão da Resposta Imune , Imunidade Celular , Células Matadoras Naturais/imunologia , Proteínas Virais de Fusão/imunologia , Linfócitos T CD8-Positivos/patologia , Linhagem Celular Transformada , Citomegalovirus/genética , Infecções por Citomegalovirus/genética , Infecções por Citomegalovirus/patologia , Humanos , Células Matadoras Naturais/patologia , Proteínas Virais de Fusão/genética
13.
Proc Natl Acad Sci U S A ; 114(23): 6104-6109, 2017 06 06.
Artigo em Inglês | MEDLINE | ID: mdl-28533400

RESUMO

Human cytomegalovirus (HCMV) strains that have been passaged in vitro rapidly acquire mutations that impact viral growth. These laboratory-adapted strains of HCMV generally exhibit restricted tropism, produce high levels of cell-free virus, and develop susceptibility to natural killer cells. To permit experimentation with a virus that retained a clinically relevant phenotype, we reconstructed a wild-type (WT) HCMV genome using bacterial artificial chromosome technology. Like clinical virus, this genome proved to be unstable in cell culture; however, propagation of intact virus was achieved by placing the RL13 and UL128 genes under conditional expression. In this study, we show that WT-HCMV produces extremely low titers of cell-free virus but can efficiently infect fibroblasts, epithelial, monocyte-derived dendritic, and Langerhans cells via direct cell-cell transmission. This process of cell-cell transfer required the UL128 locus, but not the RL13 gene, and was significantly less vulnerable to the disruptive effects of IFN, cellular restriction factors, and neutralizing antibodies compared with cell-free entry. Resistance to neutralizing antibodies was dependent on high-level expression of the pentameric gH/gL/gpUL128-131A complex, a feature of WT but not passaged strains of HCMV.


Assuntos
Técnicas de Cultura de Células/métodos , Citomegalovirus/genética , Citomegalovirus/metabolismo , Anticorpos Neutralizantes , Linhagem Celular , Células Cultivadas , Cromossomos Artificiais Bacterianos/metabolismo , Citomegalovirus/patogenicidade , Infecções por Citomegalovirus/imunologia , Infecções por Citomegalovirus/metabolismo , Fibroblastos/metabolismo , Humanos , Glicoproteínas de Membrana/metabolismo , Mutação , Fenótipo , Tropismo/imunologia , Proteínas do Envelope Viral/metabolismo , Internalização do Vírus , Replicação Viral/imunologia
14.
J Gen Virol ; 97(8): 1917-1927, 2016 08.
Artigo em Inglês | MEDLINE | ID: mdl-27050420

RESUMO

The glycoproteins gH and gL of human cytomegalovirus (HCMV) form a complex either with pUL74 (trimeric complex) or with proteins of the UL128 locus (pentameric complex). While the pentameric complex is dispensable for viral growth in fibroblasts, deletion of pUL74 causes a small plaque phenotype in HCMV lab strains, accompanied by greatly reduced cell-free infectivity. As HCMV isolates, shortly after cultivation from clinical specimens, do not release cell-free infectious viruses, we wondered whether deletion of pUL74 would also affect virus growth in this background. To address this question, we took advantage of the bacterial artificial chromosome (BAC)-cloned virus Merlin-RL13tetO, which grows cell associated due to the inducible expression of the viral RL13 gene, thereby resembling clinical isolates. Stop codons were introduced by seamless mutagenesis into UL74 and/or the UL128 locus to prevent expression of the trimeric or pentameric complex, respectively. Virus mutants were reconstituted by transfection of the respective genomes into cultured cells and analysed with respect to focal growth. When the UL128 locus was intact, deletion of pUL74 did not notably affect focal growth of Merlin, irrespective of RL13 expression. In the absence of UL128 expression, foci were increased compared with wild-type, and infectious cell-free virus was produced. Under these conditions, disruption of UL74 completely prevented virus spread from initially transfected cells to surrounding cells. In conclusion the contribution of pUL74 is masked when the UL128 locus is expressed at high levels, and its role in cell-free virus spread is only revealed when expression of the pentameric complex is inhibited.


Assuntos
Citomegalovirus/crescimento & desenvolvimento , Glicoproteínas de Membrana/metabolismo , Proteínas do Envelope Viral/metabolismo , Células Cultivadas , Citomegalovirus/genética , Fibroblastos/virologia , Deleção de Genes , Humanos , Glicoproteínas de Membrana/genética , Proteínas do Envelope Viral/genética
15.
J Virol ; 90(8): 3929-43, 2016 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-26842472

RESUMO

UNLABELLED: Clinical human cytomegalovirus (HCMV) strains invariably mutate when propagatedin vitro Mutations in gene RL13 are selected in all cell types, whereas in fibroblasts mutants in the UL128 locus (UL128L; genes UL128, UL130, and UL131A) are also selected. In addition, sporadic mutations are selected elsewhere in the genome in all cell types. We sought to investigate conditions under which HCMV can be propagated without incurring genetic defects. Bacterial artificial chromosomes (BACs) provide a stable, genetically defined source of viral genome. Viruses were generated from BACs containing the genomes of strains TR, TB40, FIX, and Merlin, as well as from Merlin-BAC recombinants containing variant nucleotides in UL128L from TB40-BAC4 or FIX-BAC. Propagation of viruses derived from TR-BAC, TB40-BAC4, and FIX-BAC in either fibroblast or epithelial cells was associated with the generation of defects around the prokaryotic vector, which is retained in the unique short (US) region of viruses. This was not observed for Merlin-BAC, from which the vector is excised in derived viruses; however, propagation in epithelial cells was consistently associated with mutations in the unique longb' (UL/b') region, all impacting on gene UL141. Viruses derived from Merlin-BAC in fibroblasts had mutations in UL128L, but mutations occurred less frequently with recombinants containing UL128L nucleotides from TB40-BAC4 or FIX-BAC. Viruses derived from a Merlin-BAC derivative in which RL13 and UL128L were either mutated or repressed were remarkably stable in fibroblasts. Thus, HCMV containing a wild-type gene complement can be generatedin vitroby deriving virus from a self-excising BAC in fibroblasts and repressing RL13 and UL128L. IMPORTANCE: Researchers should aim to study viruses that accurately represent the causative agents of disease. This is problematic for HCMV because clinical strains mutate rapidly when propagatedin vitro, becoming less cell associated, altered in tropism, more susceptible to natural killer cells, and less pathogenic. Following isolation from clinical material, HCMV genomes can be stabilized by cloning into bacterial artificial chromosomes (BACs), and then virus is regenerated by DNA transfection. However, mutations can occur not only during isolation prior to BAC cloning but also when virus is regenerated. We have identified conditions under which BAC-derived viruses containing an intact, wild-type genome can be propagatedin vitrowith minimal risk of mutants being selected, enabling studies of viruses expressing the gene complement of a clinical strain. However, even under these optimized conditions, sporadic mutations can occur, highlighting the advisability of sequencing the HCMV stocks used in experiments.


Assuntos
Cromossomos Artificiais Bacterianos , Citomegalovirus/crescimento & desenvolvimento , Cultura de Vírus/métodos , Linhagem Celular , Citomegalovirus/genética , Células Epiteliais , Fibroblastos , Genes Virais , Genoma Viral , Instabilidade Genômica , Humanos , Técnicas In Vitro , Glicoproteínas de Membrana/genética , Proteínas do Envelope Viral/genética
16.
Hum Gene Ther ; 26(5): 320-9, 2015 May.
Artigo em Inglês | MEDLINE | ID: mdl-25919378

RESUMO

Oncolytic virotherapies based on adenovirus 5 (Ad5) hold promise as adjunctive cancer therapies; however, their efficacy when delivered systemically is hampered by poor target cell specificity and preexisting anti-Ad5 immunity. Ovarian cancer represents a promising target for virotherapy, since the virus can be delivered locally into the peritoneal cavity. Both epidermal growth factor receptor (EGFR) and fibroblast growth factor receptor 1 (FGFR1) are overexpressed in the majority of human tumors, including ovarian cancer. To generate adenoviral vectors with improved tumor specificity, we generated a panel of Ad5 vectors with altered tropism for EGFR and FGFR, rather than the natural Ad5 receptor, hCAR. We have included mutations within AB loop of the viral fiber knob (KO1 mutation) to preclude interaction with hCAR, combined with insertions in the HI loop to incorporate peptides that bind either EGFR (peptide YHWYGYTPQNVI, GE11) or FGFR1 (peptides MQLPLAT, M*, and LSPPRYP, LS). Viruses were produced to high titers, and the integrity of the fiber protein was validated by Western blotting. The KO1 mutation efficiently ablated hCAR interactions, and significantly increased transduction was observed in hCAR(low)/EGFR(high) cell lines using Ad5.GE11, while transduction levels using Ad5.M* or Ad5.LS were not increased. In the presence of physiological concentrations of human blood clotting factor X (hFX), significantly increased levels of transduction via the hFX-mediated pathway were observed in cell lines, but not in primary tumor cells derived from epithelial ovarian cancer (EOC) ascites samples. Ad5-mediated transduction of EOC cells was completely abolished by the presence of 2.5% serum from patients, while, surprisingly, incorporation of the GE11 peptide resulted in significant evasion of neutralization in the same samples. We thus speculate that incorporation of the YHWYGYTPQNVI dodecapeptide within the fiber knob domain may provide a novel means of circumventing preexisting Ad5 immunity that warrants further investigation.


Assuntos
Adenoviridae/genética , Proteínas do Capsídeo/genética , Receptores ErbB/genética , Vetores Genéticos/genética , Peptídeos/genética , Receptor Tipo 1 de Fator de Crescimento de Fibroblastos/genética , Proteínas Recombinantes de Fusão , Adenoviridae/imunologia , Animais , Anticorpos Neutralizantes/imunologia , Proteínas do Capsídeo/química , Linhagem Celular , Receptores ErbB/química , Feminino , Expressão Gênica , Técnicas de Transferência de Genes , Terapia Genética , Vetores Genéticos/imunologia , Humanos , Neoplasias/genética , Neoplasias/terapia , Testes de Neutralização , Neoplasias Ovarianas/genética , Neoplasias Ovarianas/imunologia , Neoplasias Ovarianas/terapia , Peptídeos/química , Receptor Tipo 1 de Fator de Crescimento de Fibroblastos/química , Receptores Virais/genética , Receptores Virais/metabolismo , Transdução Genética , Transgenes
17.
PLoS Pathog ; 11(4): e1004811, 2015 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-25875600

RESUMO

Human cytomegalovirus (HCMV) US2, US3, US6 and US11 act in concert to prevent immune recognition of virally infected cells by CD8+ T-lymphocytes through downregulation of MHC class I molecules (MHC-I). Here we show that US2 function goes far beyond MHC-I degradation. A systematic proteomic study using Plasma Membrane Profiling revealed US2 was unique in downregulating additional cellular targets, including: five distinct integrin α-chains, CD112, the interleukin-12 receptor, PTPRJ and thrombomodulin. US2 recruited the cellular E3 ligase TRC8 to direct the proteasomal degradation of all its targets, reminiscent of its degradation of MHC-I. Whereas integrin α-chains were selectively degraded, their integrin ß1 binding partner accumulated in the ER. Consequently integrin signaling, cell adhesion and migration were strongly suppressed. US2 was necessary and sufficient for degradation of the majority of its substrates, but remarkably, the HCMV NK cell evasion function UL141 requisitioned US2 to enhance downregulation of the NK cell ligand CD112. UL141 retained CD112 in the ER from where US2 promoted its TRC8-dependent retrotranslocation and degradation. These findings redefine US2 as a multifunctional degradation hub which, through recruitment of the cellular E3 ligase TRC8, modulates diverse immune pathways involved in antigen presentation, NK cell activation, migration and coagulation; and highlight US2's impact on HCMV pathogenesis.


Assuntos
Evasão da Resposta Imune/imunologia , Glicoproteínas de Membrana/metabolismo , Proteínas do Envelope Viral/metabolismo , Proteínas Virais/metabolismo , Linhagem Celular Tumoral , Membrana Celular/metabolismo , Cromatografia Líquida de Alta Pressão , Citomegalovirus/imunologia , Citometria de Fluxo , Humanos , Immunoblotting , Imunoprecipitação , Células Matadoras Naturais/imunologia , Ativação Linfocitária/imunologia , Espectrometria de Massas , Proteínas de Membrana/metabolismo , Proteômica/métodos , RNA Interferente Pequeno , Transdução Genética
18.
PLoS Pathog ; 11(2): e1004641, 2015 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-25654642

RESUMO

CD200 receptor (CD200R) negatively regulates peripheral and mucosal innate immune responses. Viruses, including herpesviruses, have acquired functional CD200 orthologs, implying that viral exploitation of this pathway is evolutionary advantageous. However, the role that CD200R signaling plays during herpesvirus infection in vivo requires clarification. Utilizing the murine cytomegalovirus (MCMV) model, we demonstrate that CD200R facilitates virus persistence within mucosal tissue. Specifically, MCMV infection of CD200R-deficient mice (CD200R(-/-)) elicited heightened mucosal virus-specific CD4 T cell responses that restricted virus persistence in the salivary glands. CD200R did not directly inhibit lymphocyte effector function. Instead, CD200R(-/-) mice exhibited enhanced APC accumulation that in the mucosa was a consequence of elevated cellular proliferation. Although MCMV does not encode an obvious CD200 homolog, productive replication in macrophages induced expression of cellular CD200. CD200 from hematopoietic and non-hematopoietic cells contributed independently to suppression of antiviral control in vivo. These results highlight the CD200-CD200R pathway as an important regulator of antiviral immunity during cytomegalovirus infection that is exploited by MCMV to establish chronicity within mucosal tissue.


Assuntos
Antígenos CD/imunologia , Infecções por Citomegalovirus/imunologia , Macrófagos/imunologia , Mucosa/imunologia , Mucosa/virologia , Animais , Citomegalovirus/imunologia , Infecções por Citomegalovirus/metabolismo , Modelos Animais de Doenças , Citometria de Fluxo , Imunofluorescência , Macrófagos/metabolismo , Macrófagos/virologia , Glicoproteínas de Membrana/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Análise de Sequência com Séries de Oligonucleotídeos
19.
PLoS Pathog ; 10(5): e1004058, 2014 May.
Artigo em Inglês | MEDLINE | ID: mdl-24787765

RESUMO

NKG2D plays a major role in controlling immune responses through the regulation of natural killer (NK) cells, αß and γδ T-cell function. This activating receptor recognizes eight distinct ligands (the MHC Class I polypeptide-related sequences (MIC) A andB, and UL16-binding proteins (ULBP)1-6) induced by cellular stress to promote recognition cells perturbed by malignant transformation or microbial infection. Studies into human cytomegalovirus (HCMV) have aided both the identification and characterization of NKG2D ligands (NKG2DLs). HCMV immediate early (IE) gene up regulates NKGDLs, and we now describe the differential activation of ULBP2 and MICA/B by IE1 and IE2 respectively. Despite activation by IE functions, HCMV effectively suppressed cell surface expression of NKGDLs through both the early and late phases of infection. The immune evasion functions UL16, UL142, and microRNA(miR)-UL112 are known to target NKG2DLs. While infection with a UL16 deletion mutant caused the expected increase in MICB and ULBP2 cell surface expression, deletion of UL142 did not have a similar impact on its target, MICA. We therefore performed a systematic screen of the viral genome to search of addition functions that targeted MICA. US18 and US20 were identified as novel NK cell evasion functions capable of acting independently to promote MICA degradation by lysosomal degradation. The most dramatic effect on MICA expression was achieved when US18 and US20 acted in concert. US18 and US20 are the first members of the US12 gene family to have been assigned a function. The US12 family has 10 members encoded sequentially through US12-US21; a genetic arrangement, which is suggestive of an 'accordion' expansion of an ancestral gene in response to a selective pressure. This expansion must have be an ancient event as the whole family is conserved across simian cytomegaloviruses from old world monkeys. The evolutionary benefit bestowed by the combinatorial effect of US18 and US20 on MICA may have contributed to sustaining the US12 gene family.


Assuntos
Citomegalovirus , Antígenos de Histocompatibilidade Classe I/metabolismo , Evasão da Resposta Imune , Células Matadoras Naturais/imunologia , Lisossomos/metabolismo , Proteólise , Proteínas Virais/fisiologia , Adulto , Proteínas de Bactérias/metabolismo , Células Cultivadas , Citomegalovirus/imunologia , Citomegalovirus/patogenicidade , Inibidores Enzimáticos/farmacologia , Humanos , Evasão da Resposta Imune/efeitos dos fármacos , Células Matadoras Naturais/efeitos dos fármacos , Leupeptinas/farmacologia , Proteínas Luminescentes/metabolismo , Lisossomos/efeitos dos fármacos , Macrolídeos/farmacologia , Subfamília K de Receptores Semelhantes a Lectina de Células NK/fisiologia , Proteólise/efeitos dos fármacos , Proteínas Recombinantes/metabolismo
20.
Methods Mol Biol ; 1064: 275-87, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23996265

RESUMO

Natural Killer (NK) cells are crucial to the control of many viral infections. They are able to kill infected cells directly through the secretion of cytotoxic granules or through binding to death receptors on target cells. They also secrete cytokines and chemokines and, through interactions with dendritic cells, can shape adaptive immunity. The activity of NK cells can be controlled by a balance of activating and inhibitory signals conveyed through ligands on target cells binding to receptors on the NK cell. As a result viruses have devised mechanisms to modulate the expression of NK ligands on target cells, interfering with NK cell recognition and prolonging the life of infected cells. An understanding of how viruses modulate the NK response can lead to an understanding both of NK cell function, and of virus pathogenesis. Measuring the ability of NK cells to kill target cells infected with different viruses, or expressing different viral proteins, is an invaluable technique to identify the proteins and mechanisms by which viruses modulate the NK response. Here we describe two methods to measure this; one method measures sodium dichromate (51)Cr that is released from target cells as they are killed, and the other uses 7-amino-actinomycin D (7-AAD) to measure apoptosis and death of target cells following incubation with NK cells.


Assuntos
Radioisótopos de Cromo , Citotoxicidade Imunológica , Células Matadoras Naturais/imunologia , Contagem de Cintilação/métodos , Separação Celular/métodos , Dactinomicina/análogos & derivados , Fibroblastos/imunologia , Fibroblastos/metabolismo , Fibroblastos/virologia , Humanos , Leucócitos Mononucleares
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA