Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Med Microbiol Immunol ; 201(4): 551-66, 2012 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-22991040

RESUMO

Low public awareness of cytomegalovirus (CMV) results from the only mild and transient symptoms that it causes in the healthy immunocompetent host, so that primary infection usually goes unnoticed. The virus is not cleared, however, but stays for the lifetime of the host in a non-infectious, replicatively dormant state known as 'viral latency'. Medical interest in CMV results from the fact that latent virus can reactivate to cytopathogenic, tissue-destructive infection causing life-threatening end-organ disease in immunocompromised recipients of solid organ transplantation (SOT) or hematopoietic cell transplantation (HCT). It is becoming increasingly clear that CMV latency is not a static state in which the viral genome is silenced at all its genetic loci making the latent virus immunologically invisible, but rather is a dynamic state characterized by stochastic episodes of transient viral gene desilencing. This gene expression can lead to the presentation of antigenic peptides encoded by 'antigenicity-determining transcripts expressed in latency (ADTELs)' sensed by tissue-patrolling effector-memory CD8 T cells for immune surveillance of latency [In Reddehase et al., Murine model of cytomegalovirus latency and reactivation, Current Topics in Microbiology and Immunology, vol 325. Springer, Berlin, pp 315-331, 2008]. A hallmark of the CD8 T cell response to CMV is the observation that with increasing time during latency, CD8 T cells specific for certain viral epitopes increase in numbers, a phenomenon that has gained much attention in recent years and is known under the catchphrase 'memory inflation.' Here, we provide a unifying hypothesis linking stochastic viral gene desilencing during latency to 'memory inflation.'


Assuntos
Infecções por Citomegalovirus/imunologia , Citomegalovirus/imunologia , Citomegalovirus/patogenicidade , Memória Imunológica , Latência Viral/imunologia , Animais , Modelos Animais de Doenças , Humanos , Camundongos
2.
J Biomed Biotechnol ; 2011: 812742, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21253509

RESUMO

The advent of cloning herpesviral genomes as bacterial artificial chromosomes (BACs) has made herpesviruses accessible to bacterial genetics and has thus revolutionised their mutagenesis. This opened all possibilities of reverse genetics to ask scientific questions by introducing precisely accurate mutations into the viral genome for testing their influence on the phenotype under study or to create phenotypes of interest. Here, we report on our experience with using BAC technology for a designed modulation of viral antigenicity and immunogenicity with focus on the CD8 T-cell response. One approach is replacing an intrinsic antigenic peptide in a viral carrier protein with a foreign antigenic sequence, a strategy that we have termed "orthotopic peptide swap". Another approach is the functional deletion of an antigenic peptide by point mutation of its C-terminal MHC class-I anchor residue. We discuss the concepts and summarize recently published major scientific results obtained with immunological mutants of murine cytomegalovirus.


Assuntos
Antígenos Virais/imunologia , Linfócitos T CD8-Positivos/imunologia , Citomegalovirus/genética , Citomegalovirus/imunologia , Epitopos de Linfócito T/genética , Mutagênese Insercional/métodos , Deleção de Sequência/genética , Animais , Humanos
3.
J Gen Virol ; 91(Pt 6): 1524-34, 2010 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-20147515

RESUMO

Gene m164 of murine cytomegalovirus belongs to the large group of 'private' genes that show no homology to those of other cytomegalovirus species and are thought to represent 'host adaptation' genes involved in virus-host interaction. Previous interest in the m164 gene product was based on the presence of an immunodominant CD8 T-cell epitope presented at the surface of infected cells, despite interference by viral immune-evasion proteins. Here, we provide data to reveal that the m164 gene product shows unusual features in its cell biology. A novel strategy of mass-spectrometric analysis was employed to map the N terminus of the mature protein, 107 aa downstream of the start site of the predicted open reading frame. The resulting 36.5 kDa m164 gene product is identified here as an integral type-I membrane glycoprotein with exceptional intracellular trafficking dynamics, moving within the endoplasmic reticulum (ER) and outer nuclear membrane with an outstandingly high lateral membrane motility, actually 100 times higher than those published for cellular ER-resident proteins. Notably, gp36.5/m164 does not contain any typical ER-retention/retrieval signals, such as the C-terminal motifs KKXX or KXKXX, and does not pass the Golgi apparatus. Instead, it belongs to the rare group of viral glycoproteins in which the transmembrane domain (TMD) itself mediates direct ER retention. This is the first report relating TMD usage of an ER-resident transmembrane protein to its lateral membrane motility as a paradigm in cell biology. We propose that TMD usage for ER retention facilitates free and fast floating in ER-related membranes and between ER subdomains.


Assuntos
Retículo Endoplasmático/química , Glicoproteínas/metabolismo , Proteínas de Membrana/metabolismo , Muromegalovirus/fisiologia , Sinais Direcionadores de Proteínas , Proteínas Virais/metabolismo , Animais , Células COS , Chlorocebus aethiops , Glicoproteínas/química , Glicoproteínas/genética , Espectrometria de Massas , Proteínas de Membrana/química , Proteínas de Membrana/genética , Peso Molecular , Muromegalovirus/química , Muromegalovirus/genética , Fases de Leitura Aberta , Transporte Proteico , Proteínas Virais/química , Proteínas Virais/genética
4.
J Gen Virol ; 90(Pt 10): 2395-2401, 2009 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-19553390

RESUMO

Major immediate-early (MIE) transcriptional enhancers of cytomegaloviruses are key regulators that are regarded as determinants of virus replicative fitness and pathogenicity. The MIE locus of murine cytomegalovirus (mCMV) shows bidirectional gene-pair architecture, with a bipartite enhancer flanked by divergent core promoters. Here, we have constructed recombinant viruses mCMV-DeltaEnh1 and mCMV-DeltaEnh2 to study the impact of either enhancer component on bidirectional MIE gene transcription and on virus replication in cell culture and various host tissues that are relevant to CMV disease. The data revealed that the two unipartite enhancers can operate independently, but synergize in enhancing MIE gene expression early after infection. Kick-start transcription facilitated by the bipartite enhancer configuration, however, did not ultimately result in accelerated virus replication. We conclude that virus replication, once triggered, proceeds with a fixed speed and we propose that synergism between the components of the bipartite enhancer may rather increase the probability for transcription initiation.


Assuntos
Antígenos Virais/metabolismo , Elementos Facilitadores Genéticos/fisiologia , Regulação Viral da Expressão Gênica/fisiologia , Proteínas Imediatamente Precoces/metabolismo , Replicação Viral/fisiologia , Animais , Antígenos Virais/genética , Células Cultivadas , Replicação do DNA/fisiologia , DNA Viral/genética , DNA Viral/fisiologia , Fibroblastos/virologia , Proteínas Imediatamente Precoces/genética , Hospedeiro Imunocomprometido , Camundongos , Transcrição Gênica
5.
J Virol ; 83(17): 8869-84, 2009 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-19535440

RESUMO

Latent cytomegalovirus (CMV) is frequently transmitted by organ transplantation, and its reactivation under conditions of immunosuppressive prophylaxis against graft rejection by host-versus-graft disease bears a risk of graft failure due to viral pathogenesis. CMV is the most common cause of infection following liver transplantation. Although hematopoietic cells of the myeloid lineage are a recognized source of latent CMV, the cellular sites of latency in the liver are not comprehensively typed. Here we have used the BALB/c mouse model of murine CMV infection to identify latently infected hepatic cell types. We performed sex-mismatched bone marrow transplantation with male donors and female recipients to generate latently infected sex chromosome chimeras, allowing us to distinguish between Y-chromosome (gene sry or tdy)-positive donor-derived hematopoietic descendants and Y-chromosome-negative cells of recipients' tissues. The viral genome was found to localize primarily to sry-negative CD11b(-) CD11c(-) CD31(+) CD146(+) cells lacking major histocompatibility complex class II antigen (MHC-II) but expressing murine L-SIGN. This cell surface phenotype is typical of liver sinusoidal endothelial cells (LSECs). Notably, sry-positive CD146(+) cells were distinguished by the expression of MHC-II and did not harbor latent viral DNA. In this model, the frequency of latently infected cells was found to be 1 to 2 per 10(4) LSECs, with an average copy number of 9 (range, 4 to 17) viral genomes. Ex vivo-isolated, latently infected LSECs expressed the viral genes m123/ie1 and M122/ie3 but not M112-M113/e1, M55/gB, or M86/MCP. Importantly, in an LSEC transfer model, infectious virus reactivated from recipients' tissue explants with an incidence of one reactivation per 1,000 viral-genome-carrying LSECs. These findings identified LSECs as the main cellular site of murine CMV latency and reactivation in the liver.


Assuntos
Células Endoteliais/virologia , Fígado/virologia , Muromegalovirus/fisiologia , Ativação Viral , Latência Viral , Animais , Feminino , Perfilação da Expressão Gênica , Genes Virais , Masculino , Camundongos , Camundongos Endogâmicos BALB C
6.
J Virol ; 82(20): 9900-16, 2008 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-18684825

RESUMO

Despite its high coding capacity, murine CMV (mCMV) does not encode functional enzymes for nucleotide biosynthesis. It thus depends on cellular enzymes, such as ribonucleotide reductase (RNR) and thymidylate synthase (TS), to be supplied with deoxynucleoside triphosphates (dNTPs) for its DNA replication. Viral transactivation of these cellular genes in quiescent cells of host tissues is therefore a parameter of viral fitness relevant to pathogenicity. Previous work has shown that the IE1, but not the IE3, protein of mCMV transactivates RNR and TS gene promoters and has revealed an in vivo attenuation of the mutant virus mCMV-DeltaIE1. It was attractive to propose the hypothesis that lack of transactivation by IE1 and a resulting deficiency in the supply of dNTPs are the reasons for growth attenuation. Here, we have tested this hypothesis with the mutant virus mCMV-IE1-Y165C expressing an IE1 protein that selectively fails to transactivate RNR and TS in quiescent cells upon transfection while maintaining the capacity to disperse repressive nuclear domains (ND10). Our results confirm in vivo attenuation of mCMV-DeltaIE1, as indicated by a longer doubling time in host organs, whereas mCMV-IE1-Y165C replicated like mCMV-WT and the revertant virus mCMV-IE1-C165Y. Notably, the mutant virus transactivated RNR and TS upon infection of quiescent cells, thus indicating that IE1 is not the only viral transactivator involved. We conclude that transactivation of cellular genes of dNTP biosynthesis is ensured by redundancy and that attenuation of mCMV-DeltaIE1 results from the loss of other critical functions of IE1, with its function in the dispersal of ND10 being a promising candidate.


Assuntos
Regulação da Expressão Gênica , Proteínas Imediatamente Precoces/metabolismo , Muromegalovirus/fisiologia , Nucleotídeos/metabolismo , Ativação Transcricional , Replicação Viral , Sequência de Aminoácidos , Animais , Sequência de Bases , Células Cultivadas , Fibroblastos/citologia , Fibroblastos/fisiologia , Proteínas Imediatamente Precoces/genética , Fígado/citologia , Fígado/virologia , Camundongos , Camundongos Endogâmicos BALB C , Dados de Sequência Molecular , Muromegalovirus/genética , Células NIH 3T3 , Peptídeos/genética , Peptídeos/metabolismo , Mutação Puntual , Regiões Promotoras Genéticas , Alinhamento de Sequência
7.
J Virol ; 82(12): 5781-96, 2008 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-18367531

RESUMO

Cytomegalovirus (CMV) infection continues to be a complication in recipients of hematopoietic stem cell transplantation (HSCT). Preexisting donor immunity is recognized as a favorable prognostic factor for the reconstitution of protective antiviral immunity mediated primarily by CD8 T cells. Furthermore, adoptive transfer of CMV-specific memory CD8 T (CD8-T(M)) cells is a therapeutic option for preventing CMV disease in HSCT recipients. Given the different CMV infection histories of donor and recipient, a problem may arise from an antigenic mismatch between the CMV variant that has primed donor immunity and the CMV variant acquired by the recipient. Here, we have used the BALB/c mouse model of CMV infection in the immunocompromised host to evaluate the importance of donor-recipient CMV matching in immundominant epitopes (IDEs). For this, we generated the murine CMV (mCMV) recombinant virus mCMV-DeltaIDE, in which the two memory repertoire IDEs, the IE1-derived peptide 168-YPHFMPTNL-176 presented by the major histocompatibility complex class I (MHC-I) molecule L(d) and the m164-derived peptide 257-AGPPRYSRI-265 presented by the MHC-I molecule D(d), are both functionally deleted. Upon adoptive transfer, polyclonal donor CD8-T(M) cells primed by mCMV-DeltaIDE and the corresponding revertant virus mCMV-revDeltaIDE controlled infection of immunocompromised recipients with comparable efficacy and regardless of whether or not IDEs were presented in the recipients. Importantly, CD8-T(M) cells primed under conditions of immunodomination by IDEs protected recipients in which IDEs were absent. This shows that protection does not depend on compensatory expansion of non-IDE-specific CD8-T(M) cells liberated from immunodomination by the deletion of IDEs. We conclude that protection is, rather, based on the collective antiviral potential of non-IDEs independent of the presence or absence of IDE-mediated immunodomination.


Assuntos
Transferência Adotiva , Linfócitos T CD8-Positivos/imunologia , Epitopos de Linfócito T/imunologia , Infecções por Herpesviridae/imunologia , Muromegalovirus/imunologia , Animais , Linfócitos T CD8-Positivos/virologia , Células Cultivadas , Modelos Animais de Doenças , Feminino , Fibroblastos/virologia , Epitopos Imunodominantes/genética , Cinética , Camundongos , Camundongos Endogâmicos BALB C , Proteínas Virais/genética , Proteínas Virais/metabolismo
8.
Med Microbiol Immunol ; 197(2): 251-9, 2008 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-18365252

RESUMO

Hematopoietic stem cell transplantation (HSCT) bears a risk of reactivating latent cytomegalovirus (CMV) in either the transplanted hematopoietic donor cells or in parenchymal and stromal tissue cells of the immunocompromised recipient, or in both. While reactivated human CMV in recipients of organ transplantations is frequently the virus variant of the donor, this is not usually the case in HSCT recipients. Here we have used experimental sex-mismatched HSCT in the BALB/c mouse model to test if latent murine CMV from CMV-immune donors is transmitted with bone marrow cells to naive immunocompromised recipients.


Assuntos
Infecções por Citomegalovirus/transmissão , Citomegalovirus/isolamento & purificação , Transplante de Células-Tronco Hematopoéticas , Animais , Feminino , Hospedeiro Imunocomprometido , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Doadores de Tecidos , Latência Viral
9.
Med Microbiol Immunol ; 197(2): 135-44, 2008 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-18340461

RESUMO

Preclinical research in murine models as well as subsequent clinical trials have concordantly revealed a high protective potential of antiviral CD8 T cells, of donor-derived ex vivo memory CD8 T cells in particular, in the immunotherapy of cytomegalovirus (CMV) infection in immunocompromised recipients. Although it is generally held view that the observed beneficial effect of the transferred cells is viral epitope-specific, involving the recognition of MHC class-I presented peptides by cognate T cell receptors, this assumption awaits formal proof, at least with regard to the in vivo function of the CD8 T cells. This question is particularly evident for CMV, since the function of viral immune evasion proteins interferes with the MHC class-I pathway of peptide presentation. Alternatively, therefore, one has to consider the possibility that the requirement for epitope recognition may be bypassed by other ligand-receptor interactions between CD8 T cells and infected cells, which may trigger the signaling for effector functions. Clearly, such a mechanism might explain why CD8 T cells are so efficient in controlling CMV infection despite the expression of viral immune evasion proteins. Here we provide direct evidence for epitope-specificity of antiviral protection by employing a recombinant murine CMV (mCMV), namely the mutant virus mCMV-IE1-L176A, in which an immunodominant viral epitope of the regulatory immediate-early protein IE1 is functionally deleted by a point mutation replacing leucine with alanine at the C-terminal MHC anchor position of the antigenic peptide.


Assuntos
Linfócitos T CD8-Positivos/imunologia , Infecções por Citomegalovirus/prevenção & controle , Citomegalovirus/imunologia , Epitopos de Linfócito T/imunologia , Imunoterapia Adotiva/métodos , Animais , Citomegalovirus/genética , Citometria de Fluxo , Antígenos de Histocompatibilidade Classe I/biossíntese , Hospedeiro Imunocomprometido , Interferon gama/biossíntese , Fígado/imunologia , Fígado/patologia , Camundongos , Camundongos Endogâmicos BALB C , Modelos Biológicos
10.
J Virol ; 81(14): 7805-10, 2007 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-17494084

RESUMO

Enhancers are defined as DNA elements that increase transcription when placed in any orientation relative to a promoter. The major immediate-early (MIE) enhancer region of murine cytomegalovirus is flanked by transcription units ie1/3 and ie2, which are transcribed in opposite directions. We have addressed the fundamental mechanistic question of whether the enhancer synchronizes transcription of the bidirectional gene pair (synchronizer model) or whether it operates as a genetic switch, enhancing transcription of either gene in a stochastic alternation (switch model). Clonal analysis of cytokine-triggered, transcription factor-mediated MIE gene expression from latent viral genomes provided evidence in support of the switch model.


Assuntos
Citomegalovirus/genética , Elementos Facilitadores Genéticos , Genes Precoces , Transcrição Gênica , Animais , Sequência de Bases , Primers do DNA , Camundongos , Camundongos Endogâmicos BALB C
11.
J Virol ; 80(21): 10436-56, 2006 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-16928768

RESUMO

During murine cytomegalovirus (mCMV) latency in the lungs, most of the viral genomes are transcriptionally silent at the major immediate-early locus, but rare and stochastic episodes of desilencing lead to the expression of IE1 transcripts. This low-frequency but perpetual expression is accompanied by an activation of lung-resident effector-memory CD8 T cells specific for the antigenic peptide 168-YPHFMPTNL-176, which is derived from the IE1 protein. These molecular and immunological findings were combined in the "silencing/desilencing and immune sensing hypothesis" of cytomegalovirus latency and reactivation. This hypothesis proposes that IE1 gene expression proceeds to cell surface presentation of the IE1 peptide by the major histocompatibility complex (MHC) class I molecule L(d) and that its recognition by CD8 T cells terminates virus reactivation. Here we provide experimental evidence in support of this hypothesis. We generated mutant virus mCMV-IE1-L176A, in which the antigenic IE1 peptide is functionally deleted by a point mutation of the C-terminal MHC class I anchor residue Leu into Ala. Two revertant viruses, mCMV-IE1-A176L and the wobble nucleotide-marked mCMV-IE1-A176L*, in which Leu is restored by back-mutation of Ala codon GCA into Leu codons CTA and CTT, respectively, were constructed. Pulmonary latency of the mutant virus was found to be associated with an increased prevalence of IE1 transcription and with events of IE3 transactivator splicing. In conclusion, IE1-specific CD8 T cells recognize and terminate virus reactivation in vivo at the first opportunity in the reactivated gene expression program. The perpetual gene expression and antigen presentation might represent the driving molecular force in CMV-associated immunosenescence.


Assuntos
Linfócitos T CD8-Positivos/imunologia , Linfócitos T CD8-Positivos/virologia , Pulmão/imunologia , Pulmão/virologia , Muromegalovirus/imunologia , Sequência de Aminoácidos , Substituição de Aminoácidos , Animais , Antígenos Virais/genética , Sequência de Bases , Transplante de Medula Óssea , DNA Viral/genética , Epitopos/genética , Feminino , Infecções por Herpesviridae/imunologia , Infecções por Herpesviridae/virologia , Proteínas Imediatamente Precoces/genética , Proteínas Imediatamente Precoces/imunologia , Hospedeiro Imunocomprometido , Camundongos , Camundongos Endogâmicos BALB C , Modelos Biológicos , Muromegalovirus/genética , Muromegalovirus/patogenicidade , Muromegalovirus/fisiologia , Mutagênese Sítio-Dirigida , Fenótipo , Transativadores/genética , Transativadores/imunologia , Ativação Transcricional , Latência Viral , Replicação Viral
12.
J Virol ; 79(1): 326-40, 2005 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-15596827

RESUMO

Interstitial pneumonia is a major clinical manifestation of primary or recurrent cytomegalovirus (CMV) infection in immunocompromised recipients of a bone marrow transplant. In a murine model, lungs were identified as a prominent site of CMV latency and recurrence. Pulmonary latency of murine CMV is characterized by high viral genome burden and a low incidence of variegated immediate-early (IE) gene expression, reflecting a sporadic activity of the major IE promoters (MIEPs) and enhancer. The enhancer-flanking promoters MIEP1/3 and MIEP2 are switched on and off during latency in a ratio of approximately 2:1. MIEP1/3 latency-associated activity generates the IE1 transcript of the ie1/3 transcription unit but not the alternative splicing product IE3 that encodes the essential transactivator of early gene expression. Splicing thus appeared to be an important checkpoint for maintenance of latency. In accordance with previous work of others, we show here that signaling by the proinflammatory cytokine tumor necrosis factor alpha (TNF-alpha) activates IE1/3 transcription in vivo. As an addition to current knowledge, Poisson distribution analysis revealed an increased incidence of IE1/3 transcriptional events as well as a higher amount of transcripts per event. Notably, TNF-alpha promoted the splicing to IE3 transcripts, but transcription did not proceed to the M55/gB early gene. Moreover, the activated transcriptional state induced by TNF-alpha did not predispose latently infected mice to a higher incidence of virus recurrence after hematoablative treatment. In conclusion, TNF-alpha is an important inductor of IE gene transcriptional reactivation, whereas early genes downstream in the viral replicative cycle appear to be the rate-limiting checkpoint(s) for virus recurrence.


Assuntos
Regulação Viral da Expressão Gênica , Muromegalovirus/fisiologia , Transcrição Gênica , Fator de Necrose Tumoral alfa/metabolismo , Ativação Viral , Latência Viral , Animais , Transplante de Medula Óssea/efeitos adversos , Modelos Animais de Doenças , Feminino , Infecções por Herpesviridae/virologia , Humanos , Proteínas Imediatamente Precoces/genética , Proteínas Imediatamente Precoces/metabolismo , Pulmão/virologia , Camundongos , Camundongos Endogâmicos BALB C , Muromegalovirus/genética , Muromegalovirus/metabolismo , Transplante Isogênico/efeitos adversos , Proteínas Virais/genética , Proteínas Virais/metabolismo
13.
J Clin Virol ; 25 Suppl 2: S23-36, 2002 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-12361754

RESUMO

BACKGROUND: The molecular regulation of viral latency and reactivation is a central unsolved issue in the understanding of cytomegalovirus (CMV) biology. Like human CMV (hCMV), murine CMV (mCMV) can establish a latent infection in cells of the myeloid lineage. Since mCMV genome remains present in various organs after its clearance from hematopoietic cells first in bone marrow and much later in blood, there must exist one or more widely distributed cell type(s) representing the cellular site(s) of enduring mCMV latency in host tissues. Endothelial cells and histiocytes are candidates, but the question is not yet settled. Another long debated problem appears to be solved: mCMV establishes true molecular latency rather than a low-level persistence of productive infection. This conclusion is based on two recent advances. First, on a highly improved assay of infectivity, and second, on very sensitive RT PCRs for detecting viral transcripts during latency. In essence, infectious virus and productive cycle transcripts, such as transcripts of early-phase gene M55 (gB) and ie3 transcripts specifying the essential transactivator protein IE3, were found to be absent during mCMV latency in the lungs. OBJECTIVES: We will here review recent data on the variegated expression of IE-phase genes ie1 and ie2 during mCMV latency in the lungs, and on the expression patterns found in transcriptional foci during induced reactivation. We will discuss immunological implications of ie1 gene expression during latency and will speculate a bit on how CD8 T cells might trigger latency-associated ie1 gene expression in a regulatory circuit.


Assuntos
Citomegalovirus/fisiologia , Modelos Animais de Doenças , Transativadores , Proteínas Virais , Latência Viral , Animais , Infecções por Citomegalovirus/virologia , Humanos , Proteínas Imediatamente Precoces/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Ativação Viral
14.
J Virol ; 76(12): 6044-53, 2002 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-12021337

RESUMO

CD8 T cells are the principal effector cells in the resolution of acute murine cytomegalovirus (mCMV) infection in host organs. This undoubted antiviral and protective in vivo function of CD8 T cells appeared to be inconsistent with immunosubversive strategies of the virus effected by early (E)-phase genes m04, m06, and m152. The so-called immune evasion proteins gp34, gp48, and gp37/40, respectively, were found to interfere with peptide presentation at different steps in the major histocompatibility complex (MHC) class I pathway of antigen processing and presentation in fibroblasts. Accordingly, they were proposed to prevent recognition and lysis of infected fibroblasts by cytolytic T lymphocytes (CTL) during the E phase of viral gene expression. We document here that the previously identified MHC class I D(d)-restricted antigenic peptide (257)AGPPRYSRI(265) encoded by gene m164 is processed as well as presented for recognition by m164-specific CTL during the E and late phases of viral replication in the very same cells in which the immunosubversive viral proteins are effectual in preventing the presentation of processed immediate-early 1 (m123-exon 4) peptide (168)YPHFMPTNL(176). Thus, while immunosubversion is a reality, these mechanisms are apparently not as efficient as the term immune evasion implies. The pORFm164-derived peptide is the first noted peptide that constitutively escapes the immunosubversive viral functions. The most important consequence is that even the concerted action of all immunosubversive E-phase proteins eventually fails to prevent immune recognition in the E phase. The bottom-line message is that there exists no immune evasion of mCMV in fibroblasts.


Assuntos
Apresentação de Antígeno , Antígenos Virais , Fibroblastos/imunologia , Glicoproteínas de Membrana , Muromegalovirus/imunologia , Fragmentos de Peptídeos , Proteínas Virais , Animais , Antígenos Virais/genética , Antígenos Virais/imunologia , Células Cultivadas , Feminino , Fibroblastos/metabolismo , Genes Precoces/genética , Proteínas Imediatamente Precoces/genética , Proteínas Imediatamente Precoces/imunologia , Proteínas Imediatamente Precoces/metabolismo , Glicoproteínas de Membrana/química , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/imunologia , Camundongos , Camundongos Endogâmicos BALB C , Muromegalovirus/química , Fases de Leitura Aberta/genética , Fragmentos de Peptídeos/química , Fragmentos de Peptídeos/genética , Fragmentos de Peptídeos/imunologia , Linfócitos T Citotóxicos/imunologia
15.
J Gen Virol ; 83(Pt 2): 311-316, 2002 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-11807223

RESUMO

The identification of all antigenic peptides encoded by a pathogen, its T cell 'immunome', is a research aim for rational vaccine design. Screening of proteome-spanning peptide libraries or computational prediction is used to identify antigenic peptides recognized by CD8 T cells. Based on their high coding capacity, cytomegaloviruses (CMVs) could specify numerous antigenic peptides. Yet, current evidence indicates that the memory CD8 T cell response in a given haplotype is actually focused on a few viral proteins. CMVs actively interfere with antigen processing and presentation by the expression of immune evasion proteins. In the case of murine CMV (mCMV), these proteins are effectual in the early (E) phase of the virus replication cycle and should thus preclude the presentation of peptides derived from E proteins. Notably, the m18 gene is here added to a growing list of mCMV E genes that encode antigenic peptides in spite of the E phase immune evasion strategies of the virus.


Assuntos
Antígenos Virais/genética , Antígenos Virais/imunologia , Linfócitos T CD8-Positivos/imunologia , Proteínas Imediatamente Precoces/genética , Proteínas Imediatamente Precoces/imunologia , Muromegalovirus/patogenicidade , Peptídeos/imunologia , Animais , Antígenos Virais/química , Sequência de Bases , Células Cultivadas , Fibroblastos , Proteínas Imediatamente Precoces/química , Memória Imunológica , Camundongos , Dados de Sequência Molecular , Muromegalovirus/imunologia , Muromegalovirus/fisiologia , Peptídeos/genética , Replicação Viral
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...