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











Base de dados
Intervalo de ano de publicação
1.
PLoS Pathog ; 9(7): e1003493, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23935483

RESUMO

Human cytomegalovirus (HCMV) forms two gH/gL glycoprotein complexes, gH/gL/gO and gH/gL/pUL(128,130,131A), which determine the tropism, the entry pathways and the mode of spread of the virus. For murine cytomegalovirus (MCMV), which serves as a model for HCMV, a gH/gL/gO complex functionally homologous to the HCMV gH/gL/gO complex has been described. Knock-out of MCMV gO does impair, but not abolish, virus spread indicating that also MCMV might form an alternative gH/gL complex. Here, we show that the MCMV CC chemokine MCK-2 forms a complex with the glycoprotein gH, a complex which is incorporated into the virion. We could additionally show that mutants lacking both, gO and MCK-2 are not able to produce infectious virus. Trans-complementation of these double mutants with either gO or MCK-2 showed that both proteins can promote infection of host cells, although through different entry pathways. MCK-2 has been extensively studied in vivo by others. It has been shown to be involved in attracting cells for virus dissemination and in regulating antiviral host responses. We now show that MCK-2, by forming a complex with gH, strongly promotes infection of macrophages in vitro and in vivo. Thus, MCK-2 may play a dual role in MCMV infection, as a chemokine regulating the host response and attracting specific target cells and as part of a glycoprotein complex promoting entry into cells crucial for virus dissemination.


Assuntos
Quimiocinas CC/metabolismo , Infecções por Herpesviridae/imunologia , Imunidade Inata , Macrófagos/imunologia , Muromegalovirus/fisiologia , Proteínas do Envelope Viral/metabolismo , Proteínas Virais/metabolismo , Internalização do Vírus , Animais , Linhagem Celular , Células Cultivadas , Quimiocinas CC/química , Quimiocinas CC/genética , Feminino , Infecções por Herpesviridae/metabolismo , Infecções por Herpesviridae/patologia , Infecções por Herpesviridae/virologia , Fígado/imunologia , Fígado/patologia , Fígado/virologia , Macrófagos/patologia , Macrófagos/virologia , Macrófagos Peritoneais/imunologia , Macrófagos Peritoneais/patologia , Macrófagos Peritoneais/virologia , Camundongos , Camundongos Endogâmicos BALB C , Muromegalovirus/imunologia , Mutação , Multimerização Proteica , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo , Organismos Livres de Patógenos Específicos , Proteínas do Envelope Viral/química , Proteínas do Envelope Viral/genética , Proteínas Virais/química , Proteínas Virais/genética , Vírion/imunologia , Vírion/fisiologia
2.
Cell Host Microbe ; 13(5): 535-545, 2013 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-23684305

RESUMO

The immune response against a variety of pathogens can lead to activation of blood formation at ectopic sites, a process termed extramedullary hematopoiesis (EMH). The underlying mechanisms of EMH have been enigmatic. Investigating splenic EMH in mice infected with murine cytomegalovirus (MCMV), we find that, while cells of the adaptive immune system were dispensable for EMH, natural killer (NK) cells were essential. EMH required recognition of infected cells via activating NK cell receptors Ly49H or NKG2D, and correspondingly, viral interference with NK cell recognition abolished EMH. Surprisingly, development of EMH was not induced by NK cell-derived cytokines but was dependent on perforin-mediated cytotoxicity in order to control virus spread. Spreading virus reduced the numbers of F4/80(+) macrophages that were crucial for inflammatory EMH. Hence, whereas MCMV suppresses inflammation-induced EMH, NK cells confine virus spread, thereby protecting extramedullary hematopoietic niches and facilitating EMH.


Assuntos
Hematopoese Extramedular , Infecções por Herpesviridae/imunologia , Infecções por Herpesviridae/patologia , Células Matadoras Naturais/imunologia , Muromegalovirus/imunologia , Muromegalovirus/patogenicidade , Animais , Modelos Animais de Doenças , Infecções por Herpesviridae/virologia , Células Matadoras Naturais/virologia , Camundongos , Baço/imunologia , Baço/patologia , Baço/virologia
3.
J Virol ; 87(8): 4596-608, 2013 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-23408606

RESUMO

Human gammaherpesviruses cause morbidity and mortality associated with infection and transformation of lymphoid and endothelial cells. Knowledge of cell types involved in virus dissemination from primary virus entry to virus latency is fundamental for the understanding of gammaherpesvirus pathogenesis. However, the inability to directly trace cell types with respect to virus dissemination pathways has prevented definitive conclusions regarding the relative contribution of individual cell types. Here, we describe that the route of infection affects gammaherpesvirus dissemination pathways. We constructed a recombinant murine gammaherpesvirus 68 (MHV-68) variant harboring a cassette which switches fluorescent markers in a Cre-dependent manner. Since the recombinant virus which was constructed on the wild-type background was attenuated, in this study we used an M1-deleted version, which infected mice with normal kinetics. Infection of Cre-transgenic mice with this convertible virus was used to estimate the quantitative contribution of defined cell types to virus productivity and dissemination during the acute phase of MHV-68 infection. In systemic infection, we found splenic vascular endothelial cells (EC) among the first and main cells to produce virus. After local infection, the contribution of EC to splenic virus production did not represent such early kinetics. However, at later time points, B cell-derived viruses dominated splenic productivity independently of systemic or local infection. Systemic versus local infection also governed the cell types involved in loading peritoneal exudate cells, leading to latency in F4/80- and CD11b-positive target cells. Systemic infection supported EC-driven dissemination, whereas local infection supported B cell-driven dissemination.


Assuntos
Infecções por Herpesviridae/virologia , Rhadinovirus/patogenicidade , Infecções Tumorais por Vírus/virologia , Tropismo Viral , Replicação Viral , Animais , Linfócitos B/virologia , Linhagem Celular , Células Endoteliais/virologia , Genes Reporter , Infecções por Herpesviridae/patologia , Estudos Longitudinais , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Transgênicos , Rhadinovirus/genética , Rhadinovirus/crescimento & desenvolvimento , Rhadinovirus/fisiologia , Baço/virologia , Coloração e Rotulagem/métodos , Infecções Tumorais por Vírus/patologia
4.
Virol J ; 9: 209, 2012 Sep 18.
Artigo em Inglês | MEDLINE | ID: mdl-22988938

RESUMO

BACKGROUND: The polyomaviruses WUPyV and KIPyV have been detected in various sample types including feces indicating pathogenicity in the gastrointestinal (GI) system. However, quantitative viral load data from other simultaneously collected sample types are missing. As a consequence, primary replication in the GI system cannot be differentiated from swallowed virus from the respiratory tract. Here we present a retrospective quantitative longitudinal analysis in simultaneously harvested specimens from different organ sites of patients undergoing hematopoietic stem cell transplantation (HSCT). This allows the definition of sample types where deoxyribonucleic acid (DNA) detection can be expected and, as a consequence, the identification of their primary replication site. FINDINGS: Viral DNA loads from 37 patients undergoing HSCT were quantified in respiratory tract secretions (RTS), stool and urine samples as well as in leukocytes (n = 449). Leukocyte-associated virus could not be found. WUPyV was found in feces, RTS and urine samples of an infant, while KIPyV was repeatedly detected in RTS and stool samples of 4 adult patients.RTS and stool samples were matched to determine the viral load difference showing a mean difference of 2.3 log copies/ml (p < 0.001). CONCLUSIONS: The data collected in this study suggest that virus detection in the GI tract results from swallowed virus from the respiratory tract (RT). We conclude that shedding from the RT should be ruled out before viral DNA detection in the feces can be correlated to GI symptoms.


Assuntos
Transplante de Células-Tronco Hematopoéticas/efeitos adversos , Infecções por Polyomavirus/virologia , Polyomavirus/classificação , Polyomavirus/isolamento & purificação , Adulto , Fezes/virologia , Feminino , Gastroenteropatias/virologia , Humanos , Lactente , Estudos Longitudinais , Masculino , Infecções Respiratórias/virologia , Estudos Retrospectivos , Escarro/virologia , Urina/virologia
5.
PLoS Pathog ; 8(2): e1002510, 2012 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-22346748

RESUMO

Cytomegaloviruses express large amounts of viral miRNAs during lytic infection, yet, they only modestly alter the cellular miRNA profile. The most prominent alteration upon lytic murine cytomegalovirus (MCMV) infection is the rapid degradation of the cellular miR-27a and miR-27b. Here, we report that this regulation is mediated by the ∼1.7 kb spliced and highly abundant MCMV m169 transcript. Specificity to miR-27a/b is mediated by a single, apparently optimized, miRNA binding site located in its 3'-UTR. This site is easily and efficiently retargeted to other cellular and viral miRNAs by target site replacement. Expression of the 3'-UTR of m169 by an adenoviral vector was sufficient to mediate its function, indicating that no other viral factors are essential in this process. Degradation of miR-27a/b was found to be accompanied by 3'-tailing and -trimming. Despite its dramatic effect on miRNA stability, we found this interaction to be mutual, indicating potential regulation of m169 by miR-27a/b. Most interestingly, three mutant viruses no longer able to target miR-27a/b, either due to miRNA target site disruption or target site replacement, showed significant attenuation in multiple organs as early as 4 days post infection, indicating that degradation of miR-27a/b is important for efficient MCMV replication in vivo.


Assuntos
Regiões 3' não Traduzidas/genética , Infecções por Citomegalovirus/virologia , MicroRNAs/metabolismo , Muromegalovirus/fisiologia , RNA Viral/metabolismo , Replicação Viral/genética , Animais , Proteínas Argonautas/genética , Proteínas Argonautas/metabolismo , Sítios de Ligação , Linhagem Celular , Regulação para Baixo/genética , Regulação da Expressão Gênica/genética , Sequenciamento de Nucleotídeos em Larga Escala , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Camundongos SCID , MicroRNAs/genética , Muromegalovirus/genética , Mutação , Processamento Pós-Transcricional do RNA , Estabilidade de RNA/genética , RNA Viral/genética , Análise de Sequência de RNA
6.
J Immunol ; 185(1): 157-65, 2010 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-20530257

RESUMO

Human tumors frequently express membrane-bound or soluble NK group 2, member D (NKG2D) ligands. This results in chronic engagement of NKG2D on the surfaces of NK and CD8(+) T cells and rapid internalization of the receptor. Although it is well appreciated that this phenomenon impairs NKG2D-dependent function, careful analysis of NKG2D-independent functions in cells chronically stimulated through NKG2D is lacking. Using a mouse model of chronic NKG2D ligand expression, we show that constant exposure to NKG2D ligands does not functionally impair NK cells and CD8(+) T cells in the context of viral infection.


Assuntos
Células Matadoras Naturais/imunologia , Células Matadoras Naturais/metabolismo , Ativação Linfocitária/imunologia , Subfamília K de Receptores Semelhantes a Lectina de Células NK/fisiologia , Proteínas Associadas à Matriz Nuclear/fisiologia , Proteínas de Transporte Nucleocitoplasmático/fisiologia , Animais , Linfócitos T CD8-Positivos/imunologia , Linfócitos T CD8-Positivos/metabolismo , Linfócitos T CD8-Positivos/virologia , Diferenciação Celular/imunologia , Células Cultivadas , Testes Imunológicos de Citotoxicidade , Células Matadoras Naturais/citologia , Células Matadoras Naturais/virologia , Ligantes , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Muromegalovirus/imunologia , Subfamília K de Receptores Semelhantes a Lectina de Células NK/antagonistas & inibidores , Proteínas Associadas à Matriz Nuclear/genética , Proteínas Associadas à Matriz Nuclear/metabolismo , Proteínas de Transporte Nucleocitoplasmático/genética , Proteínas de Transporte Nucleocitoplasmático/metabolismo , Fatores de Tempo
7.
Cell Host Microbe ; 7(4): 324-334, 2010 Apr 22.
Artigo em Inglês | MEDLINE | ID: mdl-20413099

RESUMO

The mRNA targets of microRNAs (miRNAs) can be identified by immunoprecipitation of Argonaute (Ago) protein-containing RNA-induced silencing complexes (RISCs) followed by microarray analysis (RIP-Chip). Here we used Ago2-based RIP-Chip to identify transcripts targeted by Kaposi's sarcoma-associated herpesvirus (KSHV) miRNAs (n = 114), Epstein-Barr virus (EBV) miRNAs (n = 44), and cellular miRNAs (n = 2337) in six latently infected or stably transduced human B cell lines. Of the six KSHV miRNA targets chosen for validation, four showed regulation via their 3'UTR, while two showed regulation via binding sites within coding sequences. Two genes governing cellular transport processes (TOMM22 and IPO7) were confirmed to be targeted by EBV miRNAs. A significant number of viral miRNA targets were upregulated in infected cells, suggesting that viral miRNAs preferentially target cellular genes induced upon infection. Transcript half-life both of cellular and viral miRNA targets negatively correlated with recruitment to RISC complexes, indicating that RIP-Chip offers a quantitative estimate of miRNA function.


Assuntos
Imunoprecipitação da Cromatina/métodos , Herpesvirus Humano 4/genética , Herpesvirus Humano 8/genética , Interações Hospedeiro-Patógeno , MicroRNAs/metabolismo , Virologia/métodos , Linhagem Celular , Células Cultivadas , Regulação da Expressão Gênica , Regulação Viral da Expressão Gênica , Humanos , MicroRNAs/genética , Análise em Microsséries , RNA Viral/metabolismo
8.
J Virol ; 81(24): 13825-34, 2007 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-17913824

RESUMO

Human cytomegalovirus (CMV), a ubiquitous human pathogen, is a leading cause of congenital infections and represents a serious health risk for the immunosuppressed patient. A vaccine against CMV is currently not available. CMV is characterized by its large genome and by multiple genes modulating the immunity of the host, which cluster predominantly at genome termini. Here, we tested whether the deletion of gene blocks rich in immunomodulatory genes could be used as a novel concept in the generation of immunogenic but avirulent, herpesvirus vaccines. To generate an experimental CMV vaccine, we selectively deleted 32 genes from the mouse cytomegalovirus (MCMV) genome. The resulting mutant grew to titers similar to that of wild-type MCMV in vitro. In vivo, the mutant was 10,000-fold attenuated and well tolerated, even by highly susceptible mice deficient for B, T, and NK cells or for the interferon type I receptor. Equally relevant for safety concerns, immune suppression did not lead to the mutant's reactivation from latency. Immunization with the replication-competent mutant, but not with inactivated virus, resulted in protective immunity, which increased over time. Vaccination induced MCMV-specific antibodies and a strong T-cell response. We propose that a targeted and rational approach can improve future herpesvirus vaccines and vaccine vectors.


Assuntos
Vacinas contra Citomegalovirus/imunologia , Deleção de Genes , Infecções por Herpesviridae/prevenção & controle , Muromegalovirus/genética , Muromegalovirus/imunologia , Vacinas Atenuadas/imunologia , Animais , Anticorpos Antivirais/sangue , Vacinas contra Citomegalovirus/administração & dosagem , Vacinas contra Citomegalovirus/genética , Fibroblastos/virologia , Genoma Viral , Infecções por Herpesviridae/imunologia , Infecções por Herpesviridae/virologia , Imunização , Macrófagos/virologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos SCID , Muromegalovirus/crescimento & desenvolvimento , Células NIH 3T3 , Linfócitos T/imunologia , Vacinas Atenuadas/administração & dosagem , Vacinas Atenuadas/genética , Latência Viral
9.
PLoS Pathog ; 3(8): e123, 2007 Aug 24.
Artigo em Inglês | MEDLINE | ID: mdl-17722980

RESUMO

Understanding the mechanisms that help promote protective immune responses to pathogens is a major challenge in biomedical research and an important goal for the design of innovative therapeutic or vaccination strategies. While natural killer (NK) cells can directly contribute to the control of viral replication, whether, and how, they may help orchestrate global antiviral defense is largely unknown. To address this question, we took advantage of the well-defined molecular interactions involved in the recognition of mouse cytomegalovirus (MCMV) by NK cells. By using congenic or mutant mice and wild-type versus genetically engineered viruses, we examined the consequences on antiviral CD8 T cell responses of specific defects in the ability of the NK cells to control MCMV. This system allowed us to demonstrate, to our knowledge for the first time, that NK cells accelerate CD8 T cell responses against a viral infection in vivo. Moreover, we identify the underlying mechanism as the ability of NK cells to limit IFN-alpha/beta production to levels not immunosuppressive to the host. This is achieved through the early control of cytomegalovirus, which dramatically reduces the activation of plasmacytoid dendritic cells (pDCs) for cytokine production, preserves the conventional dendritic cell (cDC) compartment, and accelerates antiviral CD8 T cell responses. Conversely, exogenous IFN-alpha administration in resistant animals ablates cDCs and delays CD8 T cell activation in the face of NK cell control of viral replication. Collectively, our data demonstrate that the ability of NK cells to respond very early to cytomegalovirus infection critically contributes to balance the intensity of other innate immune responses, which dampens early immunopathology and promotes optimal initiation of antiviral CD8 T cell responses. Thus, the extent to which NK cell responses benefit the host goes beyond their direct antiviral effects and extends to the prevention of innate cytokine shock and to the promotion of adaptive immunity.


Assuntos
Linfócitos T CD8-Positivos/imunologia , Infecções por Herpesviridae/imunologia , Células Matadoras Naturais/imunologia , Muromegalovirus/imunologia , Animais , Linfócitos T CD8-Positivos/citologia , Citotoxicidade Imunológica , Células Dendríticas/citologia , Células Dendríticas/imunologia , Modelos Animais de Doenças , Infecções por Herpesviridae/metabolismo , Imunidade Inata , Interferon-alfa/metabolismo , Interferon beta/metabolismo , Células Matadoras Naturais/citologia , Ativação Linfocitária , Camundongos , Camundongos Congênicos , Camundongos Endogâmicos BALB C , Organismos Livres de Patógenos Específicos , Baço/citologia , Baço/virologia , Replicação Viral/imunologia
10.
J Virol ; 80(23): 11658-66, 2006 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-17005637

RESUMO

The proteins encoded by the UL34 and UL31 genes of herpes simplex virus are conserved among herpesviruses. They form a complex that is essential for the egress of the herpesvirus nucleocapsids from the nucleus. In previous work on the homologous protein complex in murine cytomegalovirus (MCMV), we defined their mutual binding domains. Here, we started to map binding domains within the UL34/UL31 proteins of alpha-, beta-, and gammaherpesviruses and to locate other functional properties. A protein complementation assay (PCA) using the TEM-1 beta-lactamase fragments fused to UL31 and UL34 protein homologues was used to study protein-protein interactions in cells. Wild-type MCMV M50 and M53 provided a strong reaction in the PCA, whereas mutants unable to form a complex did not. The homologous pairs of herpes simplex virus type 1, pseudorabies virus, human cytomegalovirus (HCMV), Epstein-Barr virus (EBV), and murine herpes virus 68 proteins also reacted, with the exception of the EBV proteins. Cross-complementation was found to be positive only within the same herpesvirus subfamily. Moreover, the HCMV homologues rescued replication-defective MCMV genomes lacking one or the other gene. We identified the binding site of M53 for M50 in the first conserved region (CR1) (M. Loetzerich, Z. Ruzsics, and U. H. Koszinowski, J. Virol. 80:73-84). Here we show that the CR1 of all tested UL31 proteins contains the UL34 binding site, and chimeric proteins carrying the subfamily-specific CR1 rescued the ability to cross-complement in the PCA.


Assuntos
Herpesvirus Humano 1/fisiologia , Proteínas Nucleares/metabolismo , Mapeamento de Interação de Proteínas , Proteínas Virais/metabolismo , Animais , Linhagem Celular Tumoral , Herpesvirus Humano 1/genética , Proteínas Nucleares/química , Proteínas Nucleares/genética , Ligação Proteica , Estrutura Terciária de Proteína , Proteínas Virais/química , Proteínas Virais/genética
11.
J Immunol ; 177(5): 3225-34, 2006 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-16920962

RESUMO

Murine CMV (MCMV) encodes three viral genes that interfere with Ag presentation (VIPRs) to CD8 T cells, m04, m06, and m152. Because the functional impact of these genes during normal infection of C57BL/6 mice is surprisingly modest, we wanted to determine whether the VIPRs are equally effective against the entire spectrum of H-2(b)-restricted CD8 T cell epitopes. We also wanted to understand how the VIPRs interact at a functional level. To address these questions, we used a panel of MCMV mutants lacking each VIPR in all possible combinations, and CTL specific for 15 H-2(b)-restricted MCMV epitopes. Only expression of all three MCMV VIPRs completely inhibited killing by CTL specific for all 15 epitopes, but removal of any one VIPR enabled lysis by at least some CTL. The dominant interaction between the VIPRs was cooperation: m06 increased the inhibition of lysis achieved by either m152 or m04. However, for 1 of 15 epitopes m04 functionally antagonized m152. There was little differential impact of any of the VIPRs on K(b) vs D(b), but a surprising degree of differential impact of the three VIPRs for different epitopes. These epitope-specific differences did not correlate with functional avidity, or with timing of VIPR expression in relation to Ag expression in the virus replication cycle. Although questions remain about the molecular mechanism and in vivo role of these genes, we conclude that the coordinated function of MCMV's three VIPRs results in a powerful inhibition of lysis of infected cells by CD8 T cells.


Assuntos
Muromegalovirus/genética , Linfócitos T Citotóxicos/citologia , Linfócitos T Citotóxicos/metabolismo , Animais , Apresentação de Antígeno/imunologia , Antígenos Virais/imunologia , Células Cultivadas , Epitopos/imunologia , Feminino , Fibroblastos , Antígenos de Histocompatibilidade Classe I/metabolismo , Macrófagos/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Muromegalovirus/imunologia , Mutação/genética , Linfócitos T Citotóxicos/imunologia , Transcrição Gênica/genética
12.
Proc Natl Acad Sci U S A ; 101(7): 2017-22, 2004 Feb 17.
Artigo em Inglês | MEDLINE | ID: mdl-14764895

RESUMO

Can CD4(+) and CD8(+) "memory" T cells that are generated and maintained in the context of low-level virus persistence protect, in the absence of antibody, against a repeat challenge with the same pathogen? Although immune T cells exert effective, long-term control of a persistent gamma-herpesvirus (gammaHV68) in Ig(-/-) microMT mice, subsequent exposure to a high dose of the same virus leads to further low-level replication in the lung. This lytic phase in the respiratory tract is dealt with effectively by the recall of memory T cells induced by a gammaHV68 recombinant (M3LacZ) that does not express the viral M3 chemokine binding protein. At least for the CD8(+) response, greater numbers of memory T cells confer enhanced protection in the M3LacZ-immune mice. However, neither WT gammaHV68 nor the minimally persistent M3LacZ primes the T cell response to the extent that a WT gammaHV68 challenge fails to establish latency in the microMT mice. Memory CD4(+) and CD8(+) T cells thus act together to limit gammaHV68 infection but are unable to provide absolute protection against a high-dose, homologous challenge.


Assuntos
Gammaherpesvirinae/imunologia , Imunoglobulinas/deficiência , Memória Imunológica/imunologia , Superinfecção/imunologia , Superinfecção/virologia , Linfócitos T/imunologia , Linfócitos T/virologia , Animais , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD4-Positivos/virologia , Linfócitos T CD8-Positivos/imunologia , Linfócitos T CD8-Positivos/virologia , Imunização Secundária , Imunoglobulinas/genética , Pulmão/virologia , Camundongos , Baço/imunologia , Baço/virologia , Latência Viral/imunologia
13.
J Virol ; 77(10): 5557-70, 2003 May.
Artigo em Inglês | MEDLINE | ID: mdl-12719548

RESUMO

The large cytomegalovirus (CMV) US22 gene family, found in all betaherpesviruses, comprises 12 members in both human cytomegalovirus (HCMV) and murine cytomegalovirus (MCMV). Conserved sequence motifs suggested a common ancestry and related functions for these gene products. Two members of this family, m140 and m141, were recently shown to affect MCMV replication on macrophages. To test the role of all US22 members in cell tropism, we analyzed the growth properties in different cell types of MCMV mutants carrying transposon insertions in all 12 US22 gene family members. When necessary, additional targeted mutants with gene deletions, ATG deletions, and ectopic gene revertants were constructed. Mutants with disruption of genes M23, M24, m25.1, m25.2, and m128 (ie2) showed no obvious growth phenotype, whereas growth of M43 mutants was reduced in a number of cell lines. Genes m142 and m143 were shown to be essential for virus replication. Growth of mutants with insertions into genes M36, m139, m140, and m141 in macrophages was severely affected. The common phenotype of the m139, m140, and m141 mutants was explained by an interaction at the protein level. The M36-dependent macrophage growth phenotype could be explained by the antiapoptotic function of the gene that was required for growth on macrophages but not for growth on other cell types. Together, the comprehensive set of mutants of the US22 gene family suggests that individual family members have diverged through evolution to serve a variety of functions for the virus.


Assuntos
Proteínas Imediatamente Precoces/genética , Proteínas Imediatamente Precoces/metabolismo , Macrófagos/virologia , Muromegalovirus/fisiologia , Replicação Viral , Células 3T3 , Animais , Apoptose , Linhagem Celular , Células Cultivadas , Elementos de DNA Transponíveis , Regulação Viral da Expressão Gênica , Infecções por Herpesviridae/virologia , Macrófagos Peritoneais/virologia , Camundongos , Camundongos Endogâmicos BALB C , Muromegalovirus/genética , Mutagênese Insercional , Mutação
14.
Rev Med Virol ; 13(2): 111-21, 2003.
Artigo em Inglês | MEDLINE | ID: mdl-12627394

RESUMO

Herpesviruses, which are important pathogens for both animals and humans, have large and complex genomes with a coding capacity for up to 225 open reading frames (ORFs). Due to the large genome size and the slow replication kinetics in vitro of some herpesviruses, mutagenesis of viral genes in the context of the viral genome by conventional recombination methods in cell culture has been difficult. Given that mutagenesis of viral genes is the basic strategy to investigate function, many of the herpesvirus ORFs could not be defined functionally. Recently, a completely new approach for the construction of herpesvirus mutants has been developed, based on cloning of the virus genome as a bacterial artificial chromosome (BAC) in E. coli. This technique allows the maintenance of viral genomes as a plasmid in E. coli and the reconstitution of viral progeny by transfection of the BAC plasmid into eukaryotic cells. Any genetic modification of the viral genome in E. coli using prokaryotic recombination proteins is possible, thereby allowing the generation of mutant viruses and facilitating the analysis of herpesvirus genomes cloned as infectious BACs. In this review, we describe the principle of cloning a viral genome as a BAC using murine gammaherpesvirus 68 (MHV-68), a mouse model for gammaherpesvirus infections, as an example.


Assuntos
Cromossomos Artificiais Bacterianos , Clonagem Molecular/métodos , Gammaherpesvirinae/genética , Genoma Viral , Animais , Escherichia coli/genética , Células Eucarióticas/virologia , Gammaherpesvirinae/fisiologia , Camundongos , Mutagênese
15.
J Infect Dis ; 187(6): 988-99, 2003 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-12660946

RESUMO

The aim of this study is to analyze the dynamics of the mouse cytomegalovirus (MCMV)-dendritic cell (DC) interaction. Immature and mature DCs derived from the mouse stem cell line factor-dependent cell Paterson mixed potential were infected with a recombinant MCMV expressing green fluorescent protein. Infection of immature DCs resulted in DC activation and virus production, both of which may contribute to viral dissemination. The infection of mature DCs was nonproductive and was restricted to immediate-early and early viral protein expression. During early stages of MCMV infection, mature DCs up-regulated major histocompatibility complex (MHC) and costimulatory molecules and activated autologous, but not allogeneic, naive T cells. At later times of MCMV infection, DCs prevented T cell activation by down-regulation of MHC and costimulatory molecules. Thus, DCs under the influence of MCMV have a physiologic dual role: to initiate and to restrict T cell activation. The lack of immunostimulation in allogeneic settings may explain the increased risk of MCMV morbidity after allogeneic transplantation.


Assuntos
Células Dendríticas/virologia , Infecções por Herpesviridae/imunologia , Muromegalovirus/fisiologia , Linfócitos T/imunologia , Animais , Diferenciação Celular , Células Dendríticas/imunologia , Proteínas de Fluorescência Verde , Infecções por Herpesviridae/virologia , Proteínas Luminescentes/genética , Ativação Linfocitária , Complexo Principal de Histocompatibilidade/imunologia , Camundongos , Muromegalovirus/genética , Muromegalovirus/crescimento & desenvolvimento , Recombinação Genética , Regulação para Cima , Replicação Viral
16.
J Virol ; 77(1): 301-8, 2003 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-12477835

RESUMO

Macrophages play an important role in murine cytomegalovirus (MCMV) infection in vivo, both in disseminating infection and in harboring latent virus. MCMV encodes three immune evasion genes (m4, m6, and m152) that interfere with the ability of cytotoxic T cells (CTL) to detect virus-infected fibroblasts, but the efficacy of immune evasion in macrophages has been controversial. Here we show that MCMV immune evasion genes function in H-2(b) primary bone marrow macrophages (BMMphi) in the same way that they do in fibroblasts. Metabolic labeling experiments showed that class I is retained in the endoplasmic reticulum by MCMV infection and associates with m4/gp34 to a similar extent in fibroblasts and BMMphi. We tested a series of K(b)- and D(b)-restricted CTL clones specific for MCMV early genes against a panel of MCMV wild-type virus and mutants lacking m152, m4, or m6. MCMV immune evasion genes effectively inhibited antigen presentation. m152 appeared sufficient to abolish D(b)-restricted presentation in infected macrophages, as has been previously observed in infected fibroblasts. However, for inhibition of recognition of infected macrophages by K(b)-restricted CTL, m4, m6, and m152 were all required. The contribution of m4 to inhibition of recognition appeared much more important in macrophages than in fibroblasts. Thus, MCMV immune evasion genes function effectively in primary macrophages to prevent CTL recognition of early antigens and show the same pattern of major histocompatibility complex class I allele discrimination as is seen in fibroblasts. Furthermore, for inhibition of K(b)-restricted presentation, a strong synergistic effect was noted among m152, m4, and m6.


Assuntos
Apresentação de Antígeno , Antígenos H-2/fisiologia , Macrófagos/imunologia , Muromegalovirus/fisiologia , Animais , Antígeno de Histocompatibilidade H-2D , Interferon gama/farmacologia , Camundongos , Camundongos Endogâmicos C57BL , Muromegalovirus/genética , Vírus 40 dos Símios/genética , Linfócitos T Citotóxicos/imunologia
17.
Nat Immunol ; 3(6): 529-35, 2002 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-12021778

RESUMO

The susceptibility of certain inbred mouse strains to murine cytomegalovirus (MCMV) is related to their inability to generate a strong natural killer (NK) cell response. We addressed here whether the MCMV susceptibility of the BALB/c strain is due to viral functions that control NK cell activation in a strain-specific manner. MCMV expresses two proteins, gp48 and gp40, that are encoded by the genes m06 and m152, respectively; they down-regulate major histocompatibility complex (MHC) class I expression at the plasma membrane. Using MCMV deletion mutants and revertants, we found that gp40 but not gp48 controls NK cell activation. Absence of gp40 improved antiviral NK cell control in BALB/c, but not C57BL/6, mice. Down-regulation of H-60, the high-affinity ligand for the NKG2D receptor, was the mechanism by which gp40 modulates NK cell activation. Thus, a single herpesvirus protein has a dual function in inhibiting both the adaptive as well as the innate immune response.


Assuntos
Linfócitos T CD8-Positivos/imunologia , Citomegalovirus/imunologia , Células Matadoras Naturais/imunologia , Proteínas Virais/imunologia , Animais , Animais Congênicos , Citomegalovirus/genética , Citomegalovirus/patogenicidade , Citotoxicidade Imunológica , Feminino , Deleção de Genes , Genes Virais , Antígenos de Histocompatibilidade Classe I/metabolismo , Ligantes , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Nus , Subfamília K de Receptores Semelhantes a Lectina de Células NK , Receptores Imunológicos/metabolismo , Receptores de Células Matadoras Naturais , Proteínas do Envelope Viral/genética , Proteínas do Envelope Viral/metabolismo , Proteínas Virais/genética
18.
J Biol Chem ; 277(3): 2216-24, 2002 Jan 18.
Artigo em Inglês | MEDLINE | ID: mdl-11707434

RESUMO

Degradation of misfolded or unassembled proteins that are co-translationally inserted into the endoplasmic reticulum involves the cytosolic proteasome system. Different principles may exist for the export of proteins into the cytosol for proteasomal degradation. Here we studied the degradation pathway of the viral glycoprotein gp48, a type I transmembrane protein, encoded by the m06 gene of murine cytomegalovirus. In cells stably transfected with the cytomegalovirus m06 gene or infected with the virus itself, two populations of gp48 can be distinguished that have different fates. Complexes of gp48 and the major histocompatibility complex (MHC) class I molecule, are transported to the lysosome for degradation. Unassembled gp48 is degraded by the cytosolic proteasome. Proteasomal inhibitors stabilize the unassembled gp48 in its core-glycosylated and membrane-associated form in the endoplasmic reticulum (ER)-Golgi intermediate compartment. This implicates that both endoplasmic reticulum and ER-Golgi intermediate compartment export gp48 and that degradation is coupled to a functional proteasome. Analysis of gp48 mutants revealed that the cytosolic part of gp48 was not responsible for the proteasome-dependent substrate transport out of the ER-Golgi intermediate compartment. Thus an indirect interaction between the proteasome and its substrate has to be discussed.


Assuntos
Acetilcisteína/análogos & derivados , Cisteína Endopeptidases/metabolismo , Citosol/metabolismo , Complexos Multienzimáticos/metabolismo , Proteínas do Envelope Viral/metabolismo , Células 3T3 , Acetilcisteína/farmacologia , Sequência de Aminoácidos , Animais , Sequência de Bases , Inibidores de Cisteína Proteinase/farmacologia , Infecções por Citomegalovirus/metabolismo , Primers do DNA , Hidrólise , Camundongos , Camundongos Endogâmicos BALB C , Dados de Sequência Molecular , Complexo de Endopeptidases do Proteassoma , Transporte Proteico
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA