Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 5 de 5
Filter
Add more filters










Database
Language
Publication year range
1.
J Gen Virol ; 91(Pt 6): 1524-34, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20147515

ABSTRACT

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.


Subject(s)
Endoplasmic Reticulum/chemistry , Glycoproteins/metabolism , Membrane Proteins/metabolism , Muromegalovirus/physiology , Protein Sorting Signals , Viral Proteins/metabolism , Animals , COS Cells , Chlorocebus aethiops , Glycoproteins/chemistry , Glycoproteins/genetics , Mass Spectrometry , Membrane Proteins/chemistry , Membrane Proteins/genetics , Molecular Weight , Muromegalovirus/chemistry , Muromegalovirus/genetics , Open Reading Frames , Protein Transport , Viral Proteins/chemistry , Viral Proteins/genetics
2.
J Virol ; 84(3): 1221-36, 2010 Feb.
Article in English | MEDLINE | ID: mdl-19906905

ABSTRACT

For recognition of infected cells by CD8 T cells, antigenic peptides are presented at the cell surface, bound to major histocompatibility complex class I (MHC-I) molecules. Downmodulation of cell surface MHC-I molecules is regarded as a hallmark function of cytomegalovirus-encoded immunoevasins. The molecular mechanisms by which immunoevasins interfere with the MHC-I pathway suggest, however, that this downmodulation may be secondary to an interruption of turnover replenishment and that hindrance of the vesicular transport of recently generated peptide-MHC (pMHC) complexes to the cell surface is the actual function of immunoevasins. Here we have used the model of murine cytomegalovirus (mCMV) infection to provide experimental evidence for this hypothesis. To quantitate pMHC complexes at the cell surface after infection in the presence and absence of immunoevasins, we generated the recombinant viruses mCMV-SIINFEKL and mCMV-Deltam06m152-SIINFEKL, respectively, expressing the K(b)-presented peptide SIINFEKL with early-phase kinetics in place of an immunodominant peptide of the viral carrier protein gp36.5/m164. The data revealed approximately 10,000 K(b) molecules presenting SIINFEKL in the absence of immunoevasins, which is an occupancy of approximately 10% of all cell surface K(b) molecules, whereas immunoevasins reduced this number to almost the detection limit. To selectively evaluate their effect on preexisting pMHC complexes, cells were exogenously loaded with SIINFEKL peptide shortly after infection with mCMV-SIINFEKA, in which endogenous presentation is prevented by an L174A mutation of the C-terminal MHC-I anchor residue. The data suggest that pMHC complexes present at the cell surface in advance of immunoevasin gene expression are downmodulated due to constitutive turnover in the absence of resupply.


Subject(s)
Immune Evasion , Muromegalovirus/physiology , Peptides/metabolism , Viral Proteins/physiology , Animals , Base Sequence , Cells, Cultured , Chromosomes, Artificial, Bacterial , DNA Primers , Histocompatibility Antigens Class I/immunology , Mice , Mice, Inbred C57BL , Muromegalovirus/immunology , Muromegalovirus/metabolism , Mutagenesis , Viral Proteins/chemistry , Viral Proteins/metabolism
3.
J Virol ; 82(20): 9900-16, 2008 Oct.
Article in English | MEDLINE | ID: mdl-18684825

ABSTRACT

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.


Subject(s)
Gene Expression Regulation , Immediate-Early Proteins/metabolism , Muromegalovirus/physiology , Nucleotides/metabolism , Transcriptional Activation , Virus Replication , Amino Acid Sequence , Animals , Base Sequence , Cells, Cultured , Fibroblasts/cytology , Fibroblasts/physiology , Immediate-Early Proteins/genetics , Liver/cytology , Liver/virology , Mice , Mice, Inbred BALB C , Molecular Sequence Data , Muromegalovirus/genetics , NIH 3T3 Cells , Peptides/genetics , Peptides/metabolism , Point Mutation , Promoter Regions, Genetic , Sequence Alignment
4.
J Virol ; 82(12): 5781-96, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18367531

ABSTRACT

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.


Subject(s)
Adoptive Transfer , CD8-Positive T-Lymphocytes/immunology , Epitopes, T-Lymphocyte/immunology , Herpesviridae Infections/immunology , Muromegalovirus/immunology , Animals , CD8-Positive T-Lymphocytes/virology , Cells, Cultured , Disease Models, Animal , Female , Fibroblasts/virology , Immunodominant Epitopes/genetics , Kinetics , Mice , Mice, Inbred BALB C , Viral Proteins/genetics , Viral Proteins/metabolism
5.
J Virol ; 80(21): 10436-56, 2006 Nov.
Article in English | MEDLINE | ID: mdl-16928768

ABSTRACT

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.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/virology , Lung/immunology , Lung/virology , Muromegalovirus/immunology , Amino Acid Sequence , Amino Acid Substitution , Animals , Antigens, Viral/genetics , Base Sequence , Bone Marrow Transplantation , DNA, Viral/genetics , Epitopes/genetics , Female , Herpesviridae Infections/immunology , Herpesviridae Infections/virology , Immediate-Early Proteins/genetics , Immediate-Early Proteins/immunology , Immunocompromised Host , Mice , Mice, Inbred BALB C , Models, Biological , Muromegalovirus/genetics , Muromegalovirus/pathogenicity , Muromegalovirus/physiology , Mutagenesis, Site-Directed , Phenotype , Trans-Activators/genetics , Trans-Activators/immunology , Transcriptional Activation , Virus Latency , Virus Replication
SELECTION OF CITATIONS
SEARCH DETAIL
...