1
|
Tsankov BK, Luchak A, Carr C, Philpott DJ. The effects of NOD-like receptors on adaptive immune responses. Biomed J 2024; 47:100637. [PMID: 37541620 PMCID: PMC10796267 DOI: 10.1016/j.bj.2023.100637] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2023] [Revised: 07/19/2023] [Accepted: 07/20/2023] [Indexed: 08/06/2023] Open
Abstract
It has long been appreciated that cues from the innate immune system orchestrate downstream adaptive immune responses. Although previous work has focused on the roles of Toll-like receptors in this regard, relatively little is known about how Nod-like receptors instruct adaptive immunity. Here we review the functions of different members of the Nod-like receptor family in orchestrating effector and anamnestic adaptive immune responses. In particular, we address the ways in which inflammasome and non-inflammasome members of this family affect adaptive immunity under various infectious and environmental contexts. Furthermore, we identify several key mechanistic questions that studies in this field have left unaddressed. Our aim is to provide a framework through which immunologists in the adaptive immune field may view their questions through an innate-immune lens and vice-versa.
Collapse
Affiliation(s)
- Boyan K Tsankov
- Department of Immunology, University of Toronto, 1 King's College Circle, Toronto, Ontario, Canada
| | - Alexander Luchak
- Department of Immunology, University of Toronto, 1 King's College Circle, Toronto, Ontario, Canada
| | - Charles Carr
- Department of Immunology, University of Toronto, 1 King's College Circle, Toronto, Ontario, Canada
| | - Dana J Philpott
- Department of Immunology, University of Toronto, 1 King's College Circle, Toronto, Ontario, Canada; Department of Laboratory Medicine and Pathobiology, University of Toronto, 1 King's College Circle, Toronto, Ontario, Canada.
| |
Collapse
|
2
|
EBNA2 driven enhancer switching at the CIITA-DEXI locus suppresses HLA class II gene expression during EBV infection of B-lymphocytes. PLoS Pathog 2021; 17:e1009834. [PMID: 34352044 PMCID: PMC8370649 DOI: 10.1371/journal.ppat.1009834] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2021] [Revised: 08/17/2021] [Accepted: 07/23/2021] [Indexed: 11/18/2022] Open
Abstract
Viruses suppress immune recognition through diverse mechanisms. Epstein-Barr Virus (EBV) establishes latent infection in memory B-lymphocytes and B-cell malignancies where it impacts B-cell immune function. We show here that EBV primary infection of naïve B-cells results in a robust down-regulation of HLA genes. We found that the viral encoded transcriptional regulatory factor EBNA2 bound to multiple regulatory regions in the HLA locus. Conditional expression of EBNA2 correlated with the down regulation of HLA class II transcription. EBNA2 down-regulation of HLA transcription was found to be dependent on CIITA, the major transcriptional activator of HLA class II gene transcription. We identified a major EBNA2 binding site downstream of the CIITA gene and upstream of DEXI, a dexamethasone inducible gene that is oriented head-to-head with CIITA gene transcripts. CRISPR/Cas9 deletion of the EBNA2 site upstream of DEXI attenuated CIITA transcriptional repression. EBNA2 caused an increase in DEXI transcription and a graded change in histone modifications with activation mark H3K27ac near the DEXI locus, and a loss of activation marks at the CIITA locus. A prominent CTCF binding site between CIITA and DEXI enhancers was mutated and further diminished the effects of EBNA2 on CIITA. Analysis of HiC data indicate that DEXI and CIITA enhancers are situated in different chromosome topological associated domains (TADs). These findings suggest that EBNA2 down regulates HLA-II genes through the down regulation of CIITA, and that this down regulation is an indirect consequence of EBNA2 enhancer formation at a neighboring TAD. We propose that enhancer competition between these neighboring chromosome domains represents a novel mechanism for gene regulation demonstrated by EBNA2.
Collapse
|
3
|
Heuberger C, Pott J, Maloy KJ. Why do intestinal epithelial cells express MHC class II? Immunology 2021; 162:357-367. [PMID: 32966619 PMCID: PMC7968399 DOI: 10.1111/imm.13270] [Citation(s) in RCA: 47] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Revised: 08/19/2020] [Accepted: 08/20/2020] [Indexed: 12/23/2022] Open
Abstract
Intestinal epithelial cells (IECs) constitute the border between the vast antigen load present in the intestinal lumen and the mucosal immune compartment. Their ability to express antigen processing and presentation machinery evokes the question whether IECs function as non-conventional antigen-presenting cells. Major histocompatibility complex (MHC) class II expression by non-haematopoietic cells, such as IECs, is tightly regulated by the class II transactivator (CIITA) and is classically induced by IFN-γ. As MHC class II expression by IECs is upregulated under inflammatory conditions, it has been proposed to activate effector CD4+ T (Teff) cells. However, other studies have reported contradictory results and instead suggested a suppressive role of antigen presentation by IECs, through regulatory T (Treg)-cell activation. Recent studies investigating the role of MHC class II + exosomes released by IECs also reported conflicting findings of either immune enhancing or immunosuppressive activities. Moreover, in addition to modulating inflammatory responses, recent findings suggest that MHC class II expression by intestinal stem cells may elicit crosstalk that promotes epithelial renewal. A more complete understanding of the different consequences of IEC MHC class II antigen presentation will guide future efforts to modulate this pathway to selectively invoke protective immunity while maintaining tolerance to beneficial antigens.
Collapse
Affiliation(s)
- Cornelia Heuberger
- Sir William Dunn School of PathologyUniversity of OxfordOxfordUK
- Kennedy Institute of RheumatologyUniversity of OxfordOxfordUK
| | - Johanna Pott
- Sir William Dunn School of PathologyUniversity of OxfordOxfordUK
- Hubrecht Organoid TechnologyUtrechtNetherlands
| | - Kevin Joseph Maloy
- Sir William Dunn School of PathologyUniversity of OxfordOxfordUK
- Institute of Infection, Immunity and InflammationUniversity of GlasgowGlasgowUK
| |
Collapse
|
4
|
Le-Trilling VTK, Becker T, Nachshon A, Stern-Ginossar N, Schöler L, Voigt S, Hengel H, Trilling M. The Human Cytomegalovirus pUL145 Isoforms Act as Viral DDB1-Cullin-Associated Factors to Instruct Host Protein Degradation to Impede Innate Immunity. Cell Rep 2021; 30:2248-2260.e5. [PMID: 32075763 DOI: 10.1016/j.celrep.2020.01.070] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2019] [Revised: 11/10/2019] [Accepted: 01/21/2020] [Indexed: 01/02/2023] Open
Abstract
Human cytomegalovirus (HCMV) causes diseases in individuals with immature or compromised immunity. To evade immune control, HCMV evolved numerous antagonists targeting the interferon system at multiple levels. By comparative analysis of naturally arising variants of the most widely studied HCMV strain, AD169, and a panel of targeted mutants, we uncover the UL145 gene as indispensable for STAT2 downregulation. Ribosome profiling confirms the translation of the canonical pUL145 protein (pUL145-Long) and newly identifies a shorter isoform (pUL145-Short). Both isoforms recruit DDB1-containing ubiquitin ligases to induce proteasomal degradation of STAT2. An alanine-scanning mutagenesis discloses the DDB1 interaction motif of pUL145 that resembles the DDB1-binding interface of cellular substrate receptors of DDB1-containing ubiquitin ligases. Thus, pUL145 constitutes a viral DDB1-cullin-associated factor (vDCAF), which mimics cellular DCAFs to exploit the ubiquitin-proteasome system to impede antiviral immunity. Notably, the viral exploitation of the cullins can be targeted to restore the efficacy of the host immune response.
Collapse
Affiliation(s)
| | - Tanja Becker
- Institute for Virology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Aharon Nachshon
- The Department of Molecular Genetics, Weizmann Institute of Science, Rehovot, Israel
| | - Noam Stern-Ginossar
- The Department of Molecular Genetics, Weizmann Institute of Science, Rehovot, Israel
| | - Lara Schöler
- Institute for Virology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Sebastian Voigt
- Department of Infectious Diseases, Robert Koch Institute, Berlin, Germany; Department of Pediatric Oncology/Hematology/SCT, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Hartmut Hengel
- Institute of Virology, Medical Center and Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Mirko Trilling
- Institute for Virology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany.
| |
Collapse
|
5
|
El-Meguid MA, Dawood RM, Ibrahim MK, Salum GM, Ahmed Abd Alla MD, El Awady MK. Reactivation of human cytomegalovirus inhibits expression of liver fibrosis related cytokines in patients chronically infected with hepatitis C virus genotype 4a. Microb Pathog 2021; 152:104596. [PMID: 33127535 DOI: 10.1016/j.micpath.2020.104596] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2020] [Revised: 10/22/2020] [Accepted: 10/23/2020] [Indexed: 12/12/2022]
Abstract
BACKGROUND The impact of human cytomegalovirus (HCMV) reactivation on the expression pattern of matrix metalloproteinases, their inhibitors and related cytokines during HCV infection poorly understood. METHODS Reactivation of CMV in 95 subjects (75 chronically infected HCV patients and 20 healthy subjects) was examined. All studied subjects had detectable IgG antibodies for CMV, but only 35/75 of HCV patients (46.7%) had detectable CMV DNA. The expressions of 11 fibrosis related genes by quantitative real-time PCR were analyzed in subjects' PBMCs. The serum levels of TGFβ2 and PDGFα have been measured by ELISA. RESULTS Chronically infected HCV patients with reactivated CMV had less expression of TGF-β1, TGF-β2, PDGFα and STAT1 transcripts than HCV patients with latent CMV (p = 0.037, 0.006, 0.001 and 0.009; respectively) and normal controls (TGF-β2, p = 0.008). Moreover the expression of (TGFβ2 and PDGFα) genes decreased significantly in CMV-reactivated patients during the early stage of fibrosis relative to the comparable stage of HCV infection (p = 0.004 and 0.008; respectively). Besides, the mRNA abundance of STAT1 gene in CMV-reactivated patients decreased dramatically as compared to HCV infections during the late stage of fibrosis (p = 0.014). The TGFβ2 protein level has been declined dramatically in CMV-reactivated patients compared to HCV infected patients and control group (p = 0.001 and 0.033; respectively). Our results suggest that CMV reactivation disrupts the expression of several cytokines as compared to solitary infection with HCV. Noticeably, the expressions of matrix metalloproteinases genes and their inhibitors have not been significantly influenced by reactivation of CMV. CONCLUSION The current data reveal that reactivation of CMV partially blocks the upregulation of 2 important pro-inflammatory cytokines i.e. TGFβ 2 and PDGFα at early stages of fibrosis, moreover this CMV mediated blockage of the STAT1 shows statistical significance at late stage of fibrosis.
Collapse
Affiliation(s)
- Mai Abd El-Meguid
- Microbial Biotechnology Department, Genetic Engineering Division, National Research Centre, 33 EL Bohouth St. (former El Tahrir St.), Dokki, Giza, 12622, Egypt
| | - Reham M Dawood
- Microbial Biotechnology Department, Genetic Engineering Division, National Research Centre, 33 EL Bohouth St. (former El Tahrir St.), Dokki, Giza, 12622, Egypt
| | - Marwa K Ibrahim
- Microbial Biotechnology Department, Genetic Engineering Division, National Research Centre, 33 EL Bohouth St. (former El Tahrir St.), Dokki, Giza, 12622, Egypt
| | - Ghada M Salum
- Microbial Biotechnology Department, Genetic Engineering Division, National Research Centre, 33 EL Bohouth St. (former El Tahrir St.), Dokki, Giza, 12622, Egypt
| | - Mohamed Darwish Ahmed Abd Alla
- Tropical Medicine Department, Faculty of Medicine, Al-Azhar University, Gouhar Al-Kaed Street, El-Hussein University Hospital, Al-Darasah, Cairo, 11675, Egypt
| | - Mostafa K El Awady
- Microbial Biotechnology Department, Genetic Engineering Division, National Research Centre, 33 EL Bohouth St. (former El Tahrir St.), Dokki, Giza, 12622, Egypt.
| |
Collapse
|
6
|
León Machado JA, Steimle V. The MHC Class II Transactivator CIITA: Not (Quite) the Odd-One-Out Anymore among NLR Proteins. Int J Mol Sci 2021; 22:1074. [PMID: 33499042 PMCID: PMC7866136 DOI: 10.3390/ijms22031074] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Revised: 01/15/2021] [Accepted: 01/19/2021] [Indexed: 12/14/2022] Open
Abstract
In this review, we discuss the major histocompatibility complex (MHC) class II transactivator (CIITA), which is the master regulator of MHC class II gene expression. CIITA is the founding member of the mammalian nucleotide-binding and leucine-rich-repeat (NLR) protein family but stood apart for a long time as the only transcriptional regulator. More recently, it was found that its closest homolog, NLRC5 (NLR protein caspase activation and recruitment domain (CARD)-containing 5), is a regulator of MHC-I gene expression. Both act as non-DNA-binding activators through multiple protein-protein interactions with an MHC enhanceosome complex that binds cooperatively to a highly conserved combinatorial cis-acting module. Thus, the regulation of MHC-II expression is regulated largely through the differential expression of CIITA. In addition to the well-defined role of CIITA in MHC-II GENE regulation, we will discuss several other aspects of CIITA functions, such as its role in cancer, its role as a viral restriction element contributing to intrinsic immunity, and lastly, its very recently discovered role as an inhibitor of Ebola and SARS-Cov-2 virus replication. We will briefly touch upon the recently discovered role of NLRP3 as a transcriptional regulator, which suggests that transcriptional regulation is, after all, not such an unusual feature for NLR proteins.
Collapse
Affiliation(s)
| | - Viktor Steimle
- Département de Biologie, Université de Sherbrooke, 2500 Boul., Sherbrooke, QC J1K 2R1, Canada;
| |
Collapse
|
7
|
Davignon JL, Combe B, Cantagrel A. Cytomegalovirus infection: friend or foe in rheumatoid arthritis? Arthritis Res Ther 2021; 23:16. [PMID: 33413603 PMCID: PMC7792325 DOI: 10.1186/s13075-020-02398-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Accepted: 12/13/2020] [Indexed: 12/27/2022] Open
Abstract
Human cytomegalovirus (HCMV) is a β-herpesvirus that causes inflammation and remains for life in a latent state in their host. HCMV has been at the center of many hypotheses regarding RA. We have recently shown that HCMV infection impairs bone erosion through the induction of the mRNA-binding protein QKI5. Latently infected RA patients display a slower progression of bone erosion in patients from a national cohort. Our observations question the possible association between HCMV and the pathophysiology of RA. In this review, we examine the possibility that HCMV may be an aggravating factor of inflammation in RA while protecting from bone erosion. We also assess its relationship with other pathogens such as bacteria causing periodontitis and responsible for ACPA production. This review thus considers whether HCMV can be regarded as a friend or a foe in the pathogenesis and the course of RA.
Collapse
Affiliation(s)
- Jean-Luc Davignon
- Centre de Physiopathologie Toulouse Purpan, U.1043 INSERM, CNRS, CHU Purpan, BP 3028, 31024, Toulouse cedex 3, France. .,Centre de Rhumatologie, CHU de Toulouse, Toulouse, France.
| | - Bernard Combe
- Lapeyronie Hospital, Montpellier I University, UMR, 5535, Montpellier, France
| | - Alain Cantagrel
- Centre de Physiopathologie Toulouse Purpan, U.1043 INSERM, CNRS, CHU Purpan, BP 3028, 31024, Toulouse cedex 3, France.,Centre de Rhumatologie, CHU de Toulouse, Toulouse, France.,Faculté de Médecine, Université Paul Sabatier Toulouse, Toulouse, France
| |
Collapse
|
8
|
Human Cytomegalovirus Decreases Major Histocompatibility Complex Class II by Regulating Class II Transactivator Transcript Levels in a Myeloid Cell Line. J Virol 2020; 94:JVI.01901-19. [PMID: 31915281 DOI: 10.1128/jvi.01901-19] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2019] [Accepted: 01/02/2020] [Indexed: 12/17/2022] Open
Abstract
Human cytomegalovirus (HCMV) is a ubiquitous pathogen that encodes many proteins to modulate the host immune response. Extensive efforts have led to the elucidation of multiple strategies employed by HCMV to effectively block NK cell targeting of virus-infected cells and the major histocompatibility complex (MHC) class I-primed CD8+ T cell response. However, viral regulation of the MHC class II-mediated CD4+ T cell response is understudied in endogenous MHC class II-expressing cells, largely because the popular cell culture systems utilized for studying HCMV do not endogenously express MHC class II. Of the many cell types infected by HCMV in the host, myeloid cells, such as monocytes, are of particular importance due to their role in latency and subsequent dissemination throughout the host. We investigated the impact of HCMV infection on MHC class II in Kasumi-3 cells, a myeloid-progenitor cell line that endogenously expresses the MHC class II gene, HLA-DR. We observed a significant reduction in the expression of surface and total HLA-DR at 72 h postinfection (hpi) and 120 hpi in infected cells. The decrease in HLA-DR expression was independent of the expression of previously described viral genes that regulate the MHC class II complex or the unique short (US) region of HCMV, a region expressing many immunomodulatory genes. The altered surface level of HLA-DR was not a result of increased endocytosis and degradation but was a result of a reduction in HLA-DR transcripts due to a decrease in the expression of the class II transactivator (CIITA).IMPORTANCE Human cytomegalovirus (HCMV) is an opportunistic herpesvirus that is asymptomatic for healthy individuals but that can lead to severe pathology in patients with congenital infections and immunosuppressed patients. Thus, it is important to understand the modulation of the immune response by HCMV, which is understudied in the context of endogenous MHC class II regulation. Using Kasumi-3 cells as a myeloid progenitor cell model endogenously expressing MHC class II (HLA-DR), this study shows that HCMV decreases the expression of HLA-DR in infected cells by reducing the transcription of HLA-DR transcripts early during infection independently of the expression of previously implicated genes. This is an important finding, as it highlights a mechanism of immune evasion utilized by HCMV to decrease the expression of MHC class II in a relevant cell system that endogenously expresses the MHC class II complex.
Collapse
|
9
|
Feng L, Sheng J, Vu GP, Liu Y, Foo C, Wu S, Trang P, Paliza-Carre M, Ran Y, Yang X, Sun X, Deng Z, Zhou T, Lu S, Li H, Liu F. Human cytomegalovirus UL23 inhibits transcription of interferon-γ stimulated genes and blocks antiviral interferon-γ responses by interacting with human N-myc interactor protein. PLoS Pathog 2018; 14:e1006867. [PMID: 29377960 PMCID: PMC5805366 DOI: 10.1371/journal.ppat.1006867] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2017] [Revised: 02/08/2018] [Accepted: 01/09/2018] [Indexed: 12/24/2022] Open
Abstract
Interferon-γ (IFN-γ) represents one of the most important innate immunity responses in a host to combat infections of many human viruses including human herpesviruses. Human N-myc interactor (Nmi) protein, which has been shown to interact with signal transducer and activator of transcription (STAT) proteins including STAT1, is important for the activation of IFN-γ induced STAT1-dependent transcription of many genes responsible for IFN-γ immune responses. However, no proteins encoded by herpesviruses have been reported to interact with Nmi and inhibit Nmi-mediated activation of IFN-γ immune responses to achieve immune evasion from IFN-γ responses. In this study, we show strong evidence that the UL23 protein of human cytomegalovirus (HCMV), a human herpesvirus, specifically interacts with Nmi. This interaction was identified through a yeast two-hybrid screen and co-immunoprecipitation in human cells. We observed that Nmi, when bound to UL23, was not associated with STAT1, suggesting that UL23 binding of Nmi disrupts the interaction of Nmi with STAT1. In cells overexpressing UL23, we observed (a) significantly reduced levels of Nmi and STAT1 in the nuclei, the sites where these proteins act to induce transcription of IFN-γ stimulated genes, and (b) decreased levels of the induction of the transcription of IFN-γ stimulated genes. UL23-deficient HCMV mutants induced higher transcription of IFN-γ stimulated genes and exhibited lower titers than parental and control revertant viruses expressing functional UL23 in IFN-γ treated cells. Thus, UL23 appears to interact directly with Nmi and inhibit nuclear translocation of Nmi and its associated protein STAT1, leading to a decrease of IFN-γ induced responses and an increase of viral resistance to IFN-γ. Our results further highlight the roles of UL23-Nmi interactions in facilitating viral immune escape from IFN-γ responses and enhancing viral resistance to IFN antiviral effects. Interferon-γ (IFN-γ) responses are vital for a host to combat infections of many human viruses including human herpesviruses. Upon treatment of IFN-γ, transcription of many genes responsible for IFN-γ immune responses is activated primarily by the signal transducer and activator of transcription (STAT) proteins such as STAT1 protein. Human N-myc interactor (Nmi) protein has been shown to interact with STAT proteins including STAT1 and activate IFN-γ induced STAT-dependent transcription. However, no proteins encoded by herpesviruses have been reported to interact with Nmi and inhibit Nmi-mediated activation of IFN-γ immune responses to achieve immune evasion from IFN-γ responses. In this study, we show strong evidence that the UL23 protein of human cytomegalovirus (HCMV), a human herpesvirus, specifically interacts with Nmi protein. UL23 appears to interact directly with Nmi and inhibit nuclear translocation of Nmi and its associated protein STAT1, leading to a decrease of IFN-γ responses and an increase of viral resistance to IFN-γ. Blocking UL23 expression led to higher transcription of IFN-γ stimulated genes and significant inhibition of viral growth in infected cells. These results suggest that interfering with Nmi function may represent an effective mechanism for a herpesvirus to block Nmi-mediated IFN-γ responses and increase viral resistance to IFN-γ. This also provides a potentially new therapeutic strategy to treat HCMV infection by modulating Nmi activity with blocking the expression of a viral protein.
Collapse
Affiliation(s)
- Linyuan Feng
- Department of Biotechnology, College of Life Science and Technology, Jinan University, Guangzhou, Guangdong, China
| | - Jingxue Sheng
- School of Public Health, University of California, Berkeley, Berkeley, California, United States of America
| | - Gia-Phong Vu
- School of Public Health, University of California, Berkeley, Berkeley, California, United States of America
| | - Yujun Liu
- Department of Biotechnology, College of Life Science and Technology, Jinan University, Guangzhou, Guangdong, China
- School of Medicine, St. George’s University, Grenada, West Indies
- Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China
| | - Chingman Foo
- Department of Biotechnology, College of Life Science and Technology, Jinan University, Guangzhou, Guangdong, China
| | - Songbin Wu
- Department of Biotechnology, College of Life Science and Technology, Jinan University, Guangzhou, Guangdong, China
| | - Phong Trang
- Department of Biotechnology, College of Life Science and Technology, Jinan University, Guangzhou, Guangdong, China
- School of Public Health, University of California, Berkeley, Berkeley, California, United States of America
| | - Marco Paliza-Carre
- School of Public Health, University of California, Berkeley, Berkeley, California, United States of America
| | - Yanhong Ran
- Department of Biotechnology, College of Life Science and Technology, Jinan University, Guangzhou, Guangdong, China
| | - Xiaoping Yang
- Department of Biotechnology, College of Life Science and Technology, Jinan University, Guangzhou, Guangdong, China
| | - Xu Sun
- Department of Biotechnology, College of Life Science and Technology, Jinan University, Guangzhou, Guangdong, China
| | - Zemin Deng
- Department of Biotechnology, College of Life Science and Technology, Jinan University, Guangzhou, Guangdong, China
- Keck School of Medicine, University of Southern California, Los Angeles, California, United States of America
| | - Tianhong Zhou
- Department of Biotechnology, College of Life Science and Technology, Jinan University, Guangzhou, Guangdong, China
| | - Sangwei Lu
- School of Public Health, University of California, Berkeley, Berkeley, California, United States of America
| | - Hongjian Li
- Department of Biotechnology, College of Life Science and Technology, Jinan University, Guangzhou, Guangdong, China
- * E-mail: (FL); (HL)
| | - Fenyong Liu
- Department of Biotechnology, College of Life Science and Technology, Jinan University, Guangzhou, Guangdong, China
- School of Public Health, University of California, Berkeley, Berkeley, California, United States of America
- * E-mail: (FL); (HL)
| |
Collapse
|
10
|
Kim JE, Kim YE, Stinski MF, Ahn JH, Song YJ. Human Cytomegalovirus IE2 86 kDa Protein Induces STING Degradation and Inhibits cGAMP-Mediated IFN-β Induction. Front Microbiol 2017; 8:1854. [PMID: 29018427 PMCID: PMC5622937 DOI: 10.3389/fmicb.2017.01854] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2017] [Accepted: 09/11/2017] [Indexed: 12/20/2022] Open
Abstract
Stimulator of interferon genes (STING) is a critical signaling molecule in the innate immune response against DNA viruses by either directly sensing intracellular DNA or functioning as an adaptor molecule to activate the type I interferon (IFN) signaling pathway. We determined the functional interaction between STING and human cytomegalovirus (HCMV). A cDNA library containing 133 HCMV ORFs was screened to identify viral genes that inhibit STING-induced IFN-β promoter activation. Among the screened ORFs, UL122, which encodes the immediate-early 2 86 kDa (IE86) protein, strongly abolished STING-induced IFN-β promoter activation. Interestingly, IE86 protein facilitated the proteasome-dependent degradation of STING and inhibited 2′3′-cGAMP-mediated induction of IFNB1 and CXCL10. Taken together, this study demonstrates the existence of a post-translational regulation of STING by HCMV IE86 protein.
Collapse
Affiliation(s)
- Jung-Eun Kim
- Department of Life Science, Gachon University, Seongnam, South Korea
| | - Young-Eui Kim
- Department of Microbiology, Carver College of Medicine, University of Iowa, Iowa City, IA, United States
| | - Mark F Stinski
- Department of Molecular Cell Biology, Samsung Biomedical Research Institute, Sungkyunkwan University School of Medicine, Suwon, South Korea
| | - Jin-Hyun Ahn
- Department of Microbiology, Carver College of Medicine, University of Iowa, Iowa City, IA, United States
| | - Yoon-Jae Song
- Department of Life Science, Gachon University, Seongnam, South Korea
| |
Collapse
|
11
|
Ibrahim MK, Khedr A, Bader El Din NG, Khairy A, El Awady MK. Increased incidence of cytomegalovirus coinfection in HCV-infected patients with late liver fibrosis is associated with dysregulation of JAK-STAT pathway. Sci Rep 2017; 7:10364. [PMID: 28871140 PMCID: PMC5583286 DOI: 10.1038/s41598-017-10604-7] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2017] [Accepted: 08/10/2017] [Indexed: 12/12/2022] Open
Abstract
Herein, we examined the association between cytomegalovirus (CMV) coinfection and the progression of liver fibrosis in hepatitis C virus (HCV) infection, and investigated the effect of CMV coinfection on JAK-STAT pathway. CMV DNAemia was detected by PCR in DNA from controls (n = 120), and HCV patients with early (F0-F1, n = 131) and late (F2-F4, n = 179) liver fibrosis. By quantitative real time PCR (qRT-PCR), we examined the profile of 8 JAK-STAT transcripts in PBMCs RNA from 90 HCV patients (39 CMV positive and 51 CMV negative), 4 CMV mono-infected patients, and 15 controls. Our results demonstrated higher incidence of CMV in F2-F4 group than in control (OR 5.479, 95% CI 3.033-9.895, p < 0.0001) or F0-F1 groups (OR 2, 95% CI 1.238-3.181, p = 0.005). qRT-PCR showed downregulation of STAT2 (p = 0.006) and IRF7 (p = 0.02) in CMV positive group compared to CMV negative one. The downregulation of STAT2 and IRF7 was mainly in CMV positive patients with late fibrosis compared to CMV negative patients (p = 0.0007 for IRF7 and p = 0.01 for STAT2). Our results are the first to report that CMV coinfection is a possible risk factor for the progression of HCV-induced liver fibrosis, and thereby CMV screening and treatment are important for HCV patients.
Collapse
Affiliation(s)
- Marwa K Ibrahim
- Department of Microbial Biotechnology, Genetic Engineering Division, National Research Centre, 33 EL Bohouth St.(former El Tahrir St.), Dokki, Giza, P.O. 12622, Egypt.
| | - Ahmed Khedr
- Department of Microbial Biotechnology, Genetic Engineering Division, National Research Centre, 33 EL Bohouth St.(former El Tahrir St.), Dokki, Giza, P.O. 12622, Egypt
| | - Noha G Bader El Din
- Department of Microbial Biotechnology, Genetic Engineering Division, National Research Centre, 33 EL Bohouth St.(former El Tahrir St.), Dokki, Giza, P.O. 12622, Egypt
| | - Ahmed Khairy
- Endemic Medicine Department, Faculty of Medicine, Cairo University, Giza, Egypt
| | - Mostafa K El Awady
- Department of Microbial Biotechnology, Genetic Engineering Division, National Research Centre, 33 EL Bohouth St.(former El Tahrir St.), Dokki, Giza, P.O. 12622, Egypt
| |
Collapse
|
12
|
Fierz W. Multiple sclerosis: an example of pathogenic viral interaction? Virol J 2017; 14:42. [PMID: 28241767 PMCID: PMC5330019 DOI: 10.1186/s12985-017-0719-3] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2016] [Accepted: 02/25/2017] [Indexed: 12/21/2022] Open
Abstract
A hypothesis is formulated on viral interaction between HHV-6A and EBV as a pathogenic mechanism in Multiple Sclerosis (MS). Evidence of molecular and genetic mechanisms suggests a link between HHV-6A infection and EBV activation in the brain of MS patients leading to intrathecal B-cell transformation. Consequent T-cell immune response against the EBV-infected cells is postulated as a pathogenic basis for inflammatory lesion formation in the brain of susceptible individuals. A further link between HHV-6A and EBV involves their induction of expression of the human endogenous retrovirus HERV-K18-encoded superantigen. Such virally induced T-cell responses might secondarily also lead to local autoimmune phenomena. Finally, research recommendations are formulated for substantiating the hypothesis on several levels: epidemiologically, genetically, and viral expression in the brain.
Collapse
Affiliation(s)
- Walter Fierz
- labormedizinisches zentrum Dr Risch, Landstr. 157, 9494, Schaan, Fürstentum, Liechtenstein.
| |
Collapse
|
13
|
Forsyth KS, Eisenlohr LC. Giving CD4+ T cells the slip: viral interference with MHC class II-restricted antigen processing and presentation. Curr Opin Immunol 2016; 40:123-9. [PMID: 27115617 PMCID: PMC4894315 DOI: 10.1016/j.coi.2016.03.003] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2016] [Accepted: 03/08/2016] [Indexed: 01/18/2023]
Abstract
Activation of CD4+ T cells through interactions with peptides bound to Major Histocompatibility Complex Class II (MHC-II) molecules is a crucial step in clearance of most pathogens. Consequently, many viruses have evolved ways of blocking this aspect of adaptive immunity, from specific targeting of processing and presentation components to modulation of signaling pathways that regulate peptide presentation in addition to many other host defense mechanisms. Such cases of interference are far less common compared to what has been elucidated in MHC-I processing and presentation. This may be attributable in part to the complexity of MHC-II antigen processing, the scope of which is only now coming to light.
Collapse
Affiliation(s)
- Katherine S Forsyth
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Laurence C Eisenlohr
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States; Department of Pathology and Laboratory Medicine at the Children's Hospital of Philadelphia Research Institute, Philadelphia, PA, United States.
| |
Collapse
|
14
|
Abstract
UNLABELLED Bluetongue virus (BTV) is a double-stranded RNA (dsRNA) virus that causes an economically important disease in ruminants. BTV infection is a strong inducer of type I interferon (IFN-I) in multiple cell types. It has been shown recently that BTV and, more specifically, the nonstructural protein NS3 of BTV are able to modulate the IFN-I synthesis pathway. However, nothing is known about the ability of BTV to counteract IFN-I signaling. Here, we investigated the effect of BTV on the IFN-I response pathway and, more particularly, the Janus tyrosine kinase (JAK)/signal transducer and activator of transcription protein (STAT) signaling pathway. We found that BTV infection triggered the expression of IFN-stimulated genes (ISGs) in A549 cells. However, when BTV-infected cells were stimulated with external IFN-I, we showed that activation of the IFN-stimulated response element (ISRE) promoter and expression of ISGs were inhibited. We found that this inhibition involved two different mechanisms that were dependent on the time of infection. After overnight infection, BTV blocked specifically the phosphorylation and nuclear translocation of STAT1. This inhibition correlated with the redistribution of STAT1 in regions adjacent to the nucleus. At a later time point of infection, BTV was found to interfere with the activation of other key components of the JAK/STAT pathway and to induce the downregulation of JAK1 and TYK2 protein expression. Overall, our study indicates for the first time that BTV is able to interfere with the JAK/STAT pathway to modulate the IFN-I response. IMPORTANCE Bluetongue virus (BTV) causes a severe disease in ruminants and has an important impact on the livestock economy in areas of endemicity such as Africa. The emergence of strains, such as serotype 8 in Europe in 2006, can lead to important economic losses due to commercial restrictions and prophylactic measures. It has been known for many years that BTV is a strong inducer of type I interferon (IFN-I) in vitro and in vivo in multiple cell types. However, the ability of BTV to interact with the IFN-I system remains unclear. Here, we report that BTV is able to modulate the IFN-I response by interfering with the Janus tyrosine kinase (JAK)/signal transducer and activator of transcription protein (STAT) signaling pathway. These findings contribute to knowledge of how BTV infection interferes with the host's innate immune response and becomes pathogenic. This will also be important for the design of efficacious vaccine candidates.
Collapse
|
15
|
Hanley PJ, Bollard CM. Controlling cytomegalovirus: helping the immune system take the lead. Viruses 2014; 6:2242-58. [PMID: 24872114 PMCID: PMC4074926 DOI: 10.3390/v6062242] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2014] [Revised: 05/09/2014] [Accepted: 05/13/2014] [Indexed: 02/07/2023] Open
Abstract
Cytomegalovirus, of the Herpesviridae family, has evolved alongside humans for thousands of years with an intricate balance of latency, immune evasion, and transmission. While upwards of 70% of humans have evidence of CMV infection, the majority of healthy people show little to no clinical symptoms of primary infection and CMV disease is rarely observed during persistent infection in immunocompetent hosts. Despite the fact that the majority of infected individuals are asymptomatic, immunologically, CMV hijacks the immune system by infecting and remaining latent in antigen-presenting cells that occasionally reactivate subclinically and present antigen to T cells, eventually causing the inflation of CMV-specific T cells until they can compromise up to 10% of the entire T cell repertoire. Because of this impact on the immune system, as well as its importance in fields such as stem cell and organ transplant, the relationship between CMV and the immune response has been studied in depth. Here we provide a review of many of these studies and insights into how CMV-specific T cells are currently being used therapeutically.
Collapse
Affiliation(s)
- Patrick J Hanley
- Program for Cell Enhancement and Technologies for Immunotherapy, Sheikh Zayed Institute for Pediatric Surgical Innovation, and Center for Cancer and Immunology Research, Children's National Health System, Washington, DC 20010, USA.
| | - Catherine M Bollard
- Program for Cell Enhancement and Technologies for Immunotherapy, Sheikh Zayed Institute for Pediatric Surgical Innovation, and Center for Cancer and Immunology Research, Children's National Health System, Washington, DC 20010, USA.
| |
Collapse
|
16
|
The carboxy terminal region of the human cytomegalovirus immediate early 1 (IE1) protein disrupts type II inteferon signaling. Viruses 2014; 6:1502-24. [PMID: 24699362 PMCID: PMC4014707 DOI: 10.3390/v6041502] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2013] [Revised: 03/07/2014] [Accepted: 03/07/2014] [Indexed: 12/21/2022] Open
Abstract
Interferons (IFNs) activate the first lines of defense against viruses, and promote innate and adaptive immune responses to viruses. We report that the immediate early 1 (IE1) protein of human cytomegalovirus (HCMV) disrupts signaling by IFNγ. The carboxyl-terminal region of IE1 is required for this function. We found no defect in the initial events in IFNγ signaling or in nuclear accumulation of signal transducer and activator of transcription 1 (STAT1) in IE1-expressing cells. Moreover, we did not observe an association between disruption of IFNγ signaling and nuclear domain 10 (ND10) disruption. However, there is reduced binding of STAT1 homodimers to target gamma activated sequence (GAS) elements in the presence of IE1. Co-immunoprecipitation studies failed to support a direct interaction between IE1 and STAT1, although these studies revealed that the C-terminal region of IE1 was required for interaction with STAT2. Together, these results indicate that IE1 disrupts IFNγ signaling by interfering with signaling events in the nucleus through a novel mechanism.
Collapse
|
17
|
Mason GM, Jackson S, Okecha G, Poole E, Sissons JGP, Sinclair J, Wills MR. Human cytomegalovirus latency-associated proteins elicit immune-suppressive IL-10 producing CD4⁺ T cells. PLoS Pathog 2013; 9:e1003635. [PMID: 24130479 PMCID: PMC3795018 DOI: 10.1371/journal.ppat.1003635] [Citation(s) in RCA: 62] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2013] [Accepted: 08/01/2013] [Indexed: 01/09/2023] Open
Abstract
Human cytomegalovirus (HCMV) is a widely prevalent human herpesvirus, which, after primary infection, persists in the host for life. In healthy individuals, the virus is well controlled by the HCMV-specific T cell response. A key feature of this persistence, in the face of a normally robust host immune response, is the establishment of viral latency. In contrast to lytic infection, which is characterised by extensive viral gene expression and virus production, long-term latency in cells of the myeloid lineage is characterised by highly restricted expression of viral genes, including UL138 and LUNA. Here we report that both UL138 and LUNA-specific T cells were detectable directly ex vivo in healthy HCMV seropositive subjects and that this response is principally CD4⁺ T cell mediated. These UL138-specific CD4⁺ T cells are able to mediate MHC class II restricted cytotoxicity and, importantly, show IFNγ effector function in the context of both lytic and latent infection. Furthermore, in contrast to CDCD4⁺ T cells specific to antigens expressed solely during lytic infection, both the UL138 and LUNA-specific CD4⁺ T cell responses included CD4⁺ T cells that secreted the immunosuppressive cytokine cIL-10. We also show that cIL-10 expressing CD4⁺ T-cells are directed against latently expressed US28 and UL111A. Taken together, our data show that latency-associated gene products of HCMV generate CD4⁺ T cell responses in vivo, which are able to elicit effector function in response to both lytic and latently infected cells. Importantly and in contrast to CD4⁺ T cell populations, which recognise antigens solely expressed during lytic infection, include a subset of cells that secrete the immunosuppressive cytokine cIL-10. This suggests that HCMV skews the T cell responses to latency-associated antigens to one that is overall suppressive in order to sustain latent carriage in vivo.
Collapse
Affiliation(s)
- Gavin M. Mason
- University of Cambridge, Department of Medicine, Cambridge, Cambridgeshire, United Kingdom
| | - Sarah Jackson
- University of Cambridge, Department of Medicine, Cambridge, Cambridgeshire, United Kingdom
| | - Georgina Okecha
- University of Cambridge, Department of Medicine, Cambridge, Cambridgeshire, United Kingdom
| | - Emma Poole
- University of Cambridge, Department of Medicine, Cambridge, Cambridgeshire, United Kingdom
| | - J. G. Patrick Sissons
- University of Cambridge, Department of Medicine, Cambridge, Cambridgeshire, United Kingdom
| | - John Sinclair
- University of Cambridge, Department of Medicine, Cambridge, Cambridgeshire, United Kingdom
| | - Mark R. Wills
- University of Cambridge, Department of Medicine, Cambridge, Cambridgeshire, United Kingdom
| |
Collapse
|
18
|
Kaposi's sarcoma-associated herpesvirus-encoded viral IRF3 modulates major histocompatibility complex class II (MHC-II) antigen presentation through MHC-II transactivator-dependent and -independent mechanisms: implications for oncogenesis. J Virol 2013; 87:5340-50. [PMID: 23449805 DOI: 10.1128/jvi.00250-13] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
Evasion of immune T cell responses is crucial for persistent viruses to establish a normal carrier state. Most studies on active immune modulation mechanisms have focused on the stage of virus production in infected cells, when large numbers of viral antigens and potential immune modulators are expressed. For oncogenic viruses such as Kaposi's sarcoma-associated herpesvirus (KSHV), which is carried as a lifelong infection, usually with little harmful effect, but can cause various tumors, the immune evasion strategies can also be relevant in the context of tumorigenesis. Here we report that the virus-encoded interferon regulatory factor 3 (vIRF3) latent viral gene expressed in KSHV-related tumors functions as a potent immunevasin. Expression of vIRF3 downregulates surface major histocompatibility complex class II (MHC-II) DR expression with slow kinetics but, more importantly, can substantially inhibit recognition by KSHV-specific CD4 T cells prior to its effects on MHC-II DR downregulation in model cell systems. This property of vIRF3 is only partly due to its ability to inhibit the transcription of CIITA and, thus, MHC-II expression; CIITA-independent inhibition of MHC-II transcripts and another as yet unidentified posttranscriptional mechanism are also involved in qualitatively modulating the availability of specific peptide/MHC-II complexes at the cell surface. Consistent with these observations, the vIRF3-expressing KSHV-associated primary effusion lymphoma (PEL) lines are generally resistant to recognition by KSHV-specific CD4 T cells. Interestingly, some PEL lines exhibit small subpopulations with lower vIRF3 expression that can be recognized. These data implicate vIRF3 as a critical determinant of the MHC-II antigen presentation function in KSHV-associated PELs that is likely to be important in the pathogenesis of these tumors.
Collapse
|
19
|
Fornara O, Odeberg J, Khan Z, Stragliotto G, Peredo I, Butler L, Söderberg-Nauclér C. Human cytomegalovirus particles directly suppress CD4 T-lymphocyte activation and proliferation. Immunobiology 2013; 218:1034-40. [PMID: 23434301 DOI: 10.1016/j.imbio.2013.01.002] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2012] [Revised: 01/01/2013] [Accepted: 01/02/2013] [Indexed: 10/27/2022]
Abstract
CD4 T cells are important regulators of the immune system and are vital for mounting a strong immune response against viral infections. Human cytomegalovirus (HCMV) is known to be a strong modulator of the innate as well as adaptive immune responses. In this study, we found that HCMV directly inhibited proliferation of CD4 T cells and rendered them unresponsive to immunological stimuli. This effect was not observed when CD4 T cells were treated with herpes simplex virus-1/2 or measles virus. When stimulated with phytohemagglutinin, concanavalin A, or phorbol myristate acetate, HCMV-treated T cells were unable to proliferate, revealing an ability of HCMV to inhibit CD4 T cell response. Furthermore, HCMV also prevented proliferation of leukemic T-cell lines. HCMV-treated CD4 T cells expressed the activation markers CD45RO and CD69, were not apoptotic and produced decreased levels of the cytokines IL-4, IFN-γ and TNF-α, compared to untreated controls. The inhibitory effect of HCMV on CD4 T cell proliferation was not mediated by HCMV gH, gB or other immunogenic glycoproteins, since intravenous immunoglobulins or gB- or gH-specific neutralizing antibodies did not prevent the suppression of T-cell proliferation. Our observations show that HCMV inhibits CD4 T cell function with potential clinical consequences for both humoral and cell-mediated immune responses.
Collapse
Affiliation(s)
- Olesja Fornara
- Center for Molecular Medicine, Department of Medicine, Karolinska University Hospital, Karolinska Institutet, 17176 Stockholm, Sweden
| | | | | | | | | | | | | |
Collapse
|
20
|
MHC2TA mRNA levels and human herpesvirus 6 in multiple sclerosis patients treated with interferon beta along two-year follow-up. BMC Neurol 2012; 12:107. [PMID: 23009575 PMCID: PMC3585729 DOI: 10.1186/1471-2377-12-107] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2012] [Accepted: 09/24/2012] [Indexed: 01/10/2023] Open
Abstract
Background In previous studies we found that MHC2TA +1614 genotype frequency was very different when MS patients with and without human herpesvirus 6 (HHV-6) in serum samples were compared; a different clinical behavior was also described. The purpose of the study was: 1. To evaluate if MHC2TA expression in MS patients was influenced by interferon beta (IFN-beta) treatment. 2. To study MHC2TA expression in MS patients with and without minor allele C. 3. To analyze the relation between MHC2TA mRNA levels and HHV-6 active infection in MS patients. Methods Blood and serum samples of 154 MS patients were collected in five programmed visits: basal (prior to beginning IFN-beta treatment), six, twelve, eighteen and twenty-four months later. HHV-6 in serum and MHC2TA mRNA levels were evaluated by PCR and RT-PCR, respectively. Neutralizing antibodies (NAbs) against IFN-beta were analyzed by the cytopathic effect assay. Results We found that MHC2TA mRNA levels were significantly lower among MS patients with HHV-6 active infection at the basal visit (without treatment) than in those MS patients without HHV-6 active infection at the basal visit (p = 0.012); in all the positive samples we only found variant A. Furthermore, 58/99 (58.6%) MS patients without HHV-6 along the five programmed visits and an increase of MHC2TA expression after two-years of IFN-beta treatment were clinical responders vs. 5/21 (23.8%) among those MS patients with HHV-6 and a decrease of MHC2TA mRNA levels along the two-years with IFN-beta treatment (p = 0.004); no differences were found between patients with and without NAbs. Conclusions MHC2TA mRNA levels could be decreased by the active replication of HHV-6; the absence of HHV-6 in serum and the increase of MHC2TA expression could be further studied as markers of good clinical response to IFN-beta treatment.
Collapse
|
21
|
Zuo J, Rowe M. Herpesviruses placating the unwilling host: manipulation of the MHC class II antigen presentation pathway. Viruses 2012; 4:1335-53. [PMID: 23012630 PMCID: PMC3446767 DOI: 10.3390/v4081335] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2012] [Revised: 08/14/2012] [Accepted: 08/15/2012] [Indexed: 12/18/2022] Open
Abstract
Lifelong persistent infection by herpesviruses depends on the balance between host immune responses and viral immune evasion. CD4 T cells responding to antigens presented on major histocompatibility complex class II (MHC-II) molecules are known to play an important role in controlling herpesvirus infections. Here we review, with emphasis on human herpesvirus infections, the strategies evolved to evade CD4 T cell immunity. These viruses target multiple points on the MHC class II antigen presentation pathway. The mechanisms include: suppression of CIITA to inhibit the synthesis of MHC class II molecules, diversion or degradation of HLA-DR molecules during membrane transport, and direct targeting of the invariant chain chaperone of HLA-DR.
Collapse
Affiliation(s)
- Jianmin Zuo
- Cancer Research UK Birmingham Cancer Centre, University of Birmingham, Birmingham B15 2TT, UK.
| | | |
Collapse
|
22
|
Garcia-Montojo M, Martinez A, De Las Heras V, Dominguez-Mozo MI, Cenit MDC, López-Cavanillas M, Garcia-Martinez A, Arias-Leal AM, Gomez de la Concha E, Urcelay E, Arroyo R, Alvarez-Lafuente R. Herpesvirus active replication in multiple sclerosis: a genetic control? J Neurol Sci 2011; 311:98-102. [PMID: 21962857 DOI: 10.1016/j.jns.2011.09.001] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2011] [Revised: 09/01/2011] [Accepted: 09/05/2011] [Indexed: 11/30/2022]
Abstract
Although the etiology of multiple sclerosis (MS) is unknown, it is generally believed that genetic, immunologic, and environmental factors are involved. The objectives of this study were: 1. to analyze if a genetic control could explain why HHV-6 would be able to actively replicate in a subset of MS patients but not in controls; 2. to study if MS patients with HHV-6 active replication are clinically different from those without HHV-6 active replication. A total of 195 MS patients and 195 controls were analyzed for two SNPs at the MHC2TA locus and two SNPs at the CD46 locus. Furthermore, the MS cohort was analyzed by PCR for the detection of HHV-6 genomes in five serum samples collected every six months along two-year follow-up. We found that 59/195 (30.2%) MS patients had at least one HHV-6 positive serum sample. No statistical significant difference was found for the two genes when the comparison was made between MS patients and controls; however, a statistical significance was found for the two polymorphisms of MHC2TA when we compared MS patients with active replication and controls (p=0.0000004 for rs4774C and p=0.011 for rs3087456G). Furthermore, increased significant differences were found for MHC2TA and CD46 when we compared interferon beta responders and non-responders within MS patients. In conclusion, we describe a gene-environment interaction in MS patients between HHV-6 and MHC2TA and CD46 that should be further studied to clarify if that interaction could be a genetic control. The results show that MS patients without HHV-6 active replication are better responders to interferon beta treatment than those with HHV-6 active replication.
Collapse
|
23
|
Lee AW, Wang N, Hornell TM, Harding JJ, Deshpande C, Hertel L, Lacaille V, Pashine A, Macaubas C, Mocarski ES, Mellins ED. Human cytomegalovirus decreases constitutive transcription of MHC class II genes in mature Langerhans cells by reducing CIITA transcript levels. Mol Immunol 2011; 48:1160-7. [PMID: 21458073 PMCID: PMC3086682 DOI: 10.1016/j.molimm.2011.02.010] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2010] [Revised: 02/20/2011] [Accepted: 02/21/2011] [Indexed: 11/29/2022]
Abstract
Human cytomegalovirus (HCMV) productively infects CD34(+) progenitor-derived, mature Langerhans-type dendritic cells (matLC) and reduces surface expression of MHC class II complexes (MHC II) by increasing intracellular retention of these molecules. To determine whether HCMV also inhibits MHC II expression by other mechanisms, we assessed mRNA levels of the class II transcriptional regulator, CIITA, and several of its target genes in infected matLC. Levels of CIITA, HLA-DRA (DRA) and DRB transcripts, and new DR protein synthesis were compared in mock-infected and HCMV-infected cells by quantitative PCR and pulse-chase immunoprecipitation analyses, respectively. CIITA mRNA levels were significantly lower in HCMV-infected matLC as compared to mock-infected cells. When assessed in the presence of Actinomycin D, the stability of CIITA transcripts was not diminished by HCMV. Analysis of promoter-specific CIITA isoforms revealed that types I, III and IV all were decreased by HCMV, a result that differs from changes after incubation of these cells with lipopolysaccharide (LPS). Exposure to UV-inactivated virus failed to reduce CIITA mRNA levels, implicating de novo viral gene expression in this effect. HCMV-infected matLC also expressed lower levels of DR transcripts and reduced DR protein synthesis rates compared to mock-infected matLC. In summary, we demonstrate that HCMV infection of a human dendritic cell subset inhibits constitutive CIITA expression, most likely at the transcriptional level, resulting in reduced MHC II biosynthesis. We suggest this represents a new mechanism of modulation of mature LC by HCMV.
Collapse
Affiliation(s)
- Andrew W. Lee
- Department of Pediatrics, Stanford University School of Medicine, Stanford, CA 94305, USA
- Department of Medicine (Division of Infectious Diseases), Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Nan Wang
- Department of Pediatrics, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Tara M.C. Hornell
- Department of Pediatrics, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - James J. Harding
- Department of Pediatrics, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Chetan Deshpande
- Department of Pediatrics, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Laura Hertel
- Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Vashti Lacaille
- Department of Pediatrics, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Achal Pashine
- Department of Pediatrics, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Claudia Macaubas
- Department of Pediatrics, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Edward S. Mocarski
- Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Elizabeth D. Mellins
- Department of Pediatrics, Stanford University School of Medicine, Stanford, CA 94305, USA
| |
Collapse
|
24
|
Lian L, Qu L, Zheng J, Liu C, Zhang Y, Chen Y, Xu G, Yang N. Expression profiles of genes within a subregion of chicken major histocompatibility complex B in spleen after Marek’s disease virus infection. Poult Sci 2010; 89:2123-9. [DOI: 10.3382/ps.2010-00919] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
|
25
|
Alvarez-Lafuente R, Martinez A, Garcia-Montojo M, Mas A, De Las Heras V, Dominguez-Mozo MI, Maria Del Carmen C, López-Cavanillas M, Bartolome M, Gomez de la Concha E, Urcelay E, Arroyo R. MHC2TA rs4774C and HHV-6A active replication in multiple sclerosis patients. Eur J Neurol 2009; 17:129-35. [PMID: 19659749 DOI: 10.1111/j.1468-1331.2009.02758.x] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
BACKGROUND AND PURPOSE In a previous report, a strong gene-environment interaction between human herpesvirus 6A (HHV6A) active replication and MHC2TA rs4774C was demonstrated. The objectives of this study were: (i) to reappraise the association that was found in the previous study; (ii) to evaluate if MS patients with minor allele C and HHV-6A active infection had different clinical behavior; and (iii) to analyze the possible association of MHC2TA rs4774C with Epstein-Barr virus (EBV). METHODS A total of 149 MS patients were analyzed both at the MHC2TA locus and by HHV-6A status in serum. We studied a G/C polymorphism (rs4774) by a TaqMan Assay-on-Demand. HHV-6A genomes in serum were evaluated by quantitative PCR. A control group of 562 healthy Spanish individuals was included for comparative purposes in the genetic analyses. A battery of clinical data was collected for all the MS patients included in the study. RESULTS (i) MHC2TA/HHV-6A interaction: we found the same strong association of the rs4774C allele with HHV-6A active replication than in the previous study (P = 0.0001). (ii) CLINICAL DATA the two main statistical significant differences for MS patients with HHV-6A active infection and minor allele C were: (a) a significant number of them were not free of progression (EDSS = 0) 2 years after the diagnosis (P = 0.01); (b) only a third of them responded to interferon beta treatment (P = 0.05). CONCLUSIONS This study has verified previous results about the strong gene-environment interaction between HHV6A active replication and MHC2TA rs4774C. Furthermore, a different clinical behavior for MS patients with HHV-6A active infection and minor allele C was found.
Collapse
|
26
|
Le VTK, Trilling M, Wilborn M, Hengel H, Zimmermann A. Human cytomegalovirus interferes with signal transducer and activator of transcription (STAT) 2 protein stability and tyrosine phosphorylation. J Gen Virol 2008; 89:2416-2426. [PMID: 18796709 DOI: 10.1099/vir.0.2008/001669-0] [Citation(s) in RCA: 73] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
We have investigated the role of signal transducer and activator of transcription (STAT) 2 during human cytomegalovirus (HCMV) replication and found that protein levels of STAT2 are downregulated. STAT2 downregulation was observed in HCMV clinical isolates and laboratory strains with the exception of strain Towne. The HCMV-induced loss of STAT2 protein occurred despite an increased accumulation of STAT2 mRNA; it required HCMV early gene expression. The decrease in STAT2 was sensitive to proteasome inhibition, suggesting degradation of STAT2 via the ubiquitin proteasome pathway. Notably, pUL27, the HCMV homologue of the mouse CMV pM27 protein, which mediates the selective proteolysis of STAT2, did not induce STAT2 downregulation. Moreover, preceding STAT2 degradation, alpha/beta interferon (IFN)-receptor-mediated tyrosine phosphorylation of STAT2 was inhibited in HCMV-infected cells. This effect was paralleled by impaired tyrosine activation of STAT1 and STAT3. Accordingly, IFNs affected the replication efficiency of STAT2 degrading and non-degrading HCMV strains to a similar degree. In summary, HCMV abrogates IFN receptor signalling at multiple checkpoints by independent mechanisms including UL27-independent degradation of STAT2 and a preceding blockade of STAT2 phosphorylation.
Collapse
Affiliation(s)
- Vu Thuy Khanh Le
- Heinrich-Heine-Universität Düsseldorf, Institut für Virologie, 40225 Düsseldorf, Germany
| | - Mirko Trilling
- Heinrich-Heine-Universität Düsseldorf, Institut für Virologie, 40225 Düsseldorf, Germany
| | - Manuel Wilborn
- Heinrich-Heine-Universität Düsseldorf, Institut für Virologie, 40225 Düsseldorf, Germany
| | - Hartmut Hengel
- Heinrich-Heine-Universität Düsseldorf, Institut für Virologie, 40225 Düsseldorf, Germany
| | - Albert Zimmermann
- Heinrich-Heine-Universität Düsseldorf, Institut für Virologie, 40225 Düsseldorf, Germany
| |
Collapse
|
27
|
Baron M, Davignon JL. Inhibition of IFN-gamma-induced STAT1 tyrosine phosphorylation by human CMV is mediated by SHP2. THE JOURNAL OF IMMUNOLOGY 2008; 181:5530-6. [PMID: 18832710 DOI: 10.4049/jimmunol.181.8.5530] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Human CMV (HCMV) is a ubiquitous beta-herpesvirus which has developed several mechanisms of escape from the immune system. IFN-gamma-induced signaling relies on the integrity of the JAK/STAT pathway which is regulated by phosphorylation steps and leads to nuclear translocation of tyrosine-phosphorylated STAT1 (STAT1-P-Tyr), and its binding to IFN-gamma activation site sequences of IFN-gamma-inducible promoters. Activation of those promoters leads to the expression of genes involved in the immune response and in the antiviral effects of IFN-gamma. Src homology region 2 domain-containing phosphatase 2 (SHP2) is a ubiquitous phosphatase involved in the regulation of IFN-gamma-mediated tyrosine phosphorylation. Several mechanisms account for the inhibition IFN-gamma signaling pathway by HCMV. In this study, we have identified a new mechanism that involved the inhibition of STAT1 tyrosine phosphorylation within 12-24 h postinfection. This defect was dependent on HCMV transcription. Consequences were impaired nuclear translocation of STAT1-P-Tyr, inhibition of IFN-gamma activation site-STAT1 interaction, and inhibition of HLA-DR expression. Expression of indoleamine-2,3-dioxygenase which is involved in the antiviral effects of IFN-gamma was also inhibited. Treatment of cells with sodium orthovanadate rescued STAT1 tyrosine phosphorylation, suggesting that a tyrosine phosphatase was involved in this inhibition. Coimmunoprecipitation of STAT1 and SHP2 was induced by HCMV infection, and SHP2 small interfering RNA restored the expression of STAT1-P-Tyr. Our data suggest that SHP2 activation induced by HCMV infection is responsible for the down-regulation of IFN-gamma-induced STAT1 tyrosine phosphorylation.
Collapse
Affiliation(s)
- Michel Baron
- Institut National de la Santé et de la Recherche Médicale Unité 563, Centre de Physiopathologie de Toulouse Purpan, Paul Sabatier University, Toulouse, France
| | | |
Collapse
|
28
|
Human immunodeficiency virus type 1 Vpu protein interacts with CD74 and modulates major histocompatibility complex class II presentation. J Virol 2007; 82:893-902. [PMID: 17959659 DOI: 10.1128/jvi.01373-07] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The human immunodeficiency virus type 1 (HIV-1) Vpu accessory protein is a transmembrane protein that down regulates CD4 expression and promotes the release of new virions. We screened a human leukocyte-specific yeast two-hybrid expression library to discover novel Vpu-interacting cellular proteins. The major histocompatibility complex class II (MHC II) invariant chain, also called Ii or CD74, was found to be one such protein. We show direct binding of Vpu and CD74 by using a yeast two-hybrid assay and coimmunoprecipitation from HIV-1-infected cells. The cytoplasmic region of Vpu was found to interact with the 30-amino-acid cytoplasmic tail of CD74. Human monocytic U937 cells infected with wild-type or Vpu-defective HIV-1 and transfected cells showed that Vpu down modulated the surface expression of mature MHC II molecules. The reduction in cell surface mature MHC II molecules correlated with decreased antigen presentation to T cells in culture. Thus, the Vpu protein also contributes to viral persistence by attenuating immune responses during HIV infection. This report further exemplifies the rich diversity and redundancy shown by HIV in immune evasion.
Collapse
|
29
|
Delmas S, Brousset P, Clément D, Le Roy E, Davignon JL. Anti-IE1 CD4+ T-cell clones kill peptide-pulsed, but not human cytomegalovirus-infected, target cells. J Gen Virol 2007; 88:2441-2449. [PMID: 17698653 DOI: 10.1099/vir.0.82958-0] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Cellular immunity plays a major role in the control of human cytomegalovirus (HCMV) infection. CD4(+) T lymphocytes have been shown to contribute to this function but their precise role is a matter of debate. Although CD4(+) T cells have been shown to kill target cells through the perforin/granzyme pathway, whether HCMV-specific CD4(+) T cells are capable of killing HCMV-infected targets has not yet been documented. In the present paper, we have taken advantage of well established cellular reagents to address this issue. Human CD4(+) T-cell clones specific for the major immediate-early protein IE1 were shown to perform perforin-based cytotoxicity against peptide-pulsed targets. However, when tested on infected anitgen presenting cell targets, cytotoxicity was not detectable, although gamma interferon (IFN-gamma) production was significant. Furthermore, cytotoxicity against peptide-pulsed targets was inhibited by HCMV infection, whereas IFN-gamma production was not modified, suggesting that antigen processing was not altered. Remarkably, degranulation of CD4(+) T cells in the presence of infected targets was significant. Together, our data suggest that impaired cytotoxicity is not due to failure to recognize infected targets but rather to a mechanism specifically related to cytotoxicity.
Collapse
Affiliation(s)
- Sandra Delmas
- INSERM U563, Centre de Physiopathologie de Toulouse Purpan, IFR 30, Centre Hospitalier Purpan, 31024 Toulouse Cedex, France
| | - Pierre Brousset
- INSERM U563, Centre de Physiopathologie de Toulouse Purpan, IFR 30, Centre Hospitalier Purpan, 31024 Toulouse Cedex, France
| | - Danièle Clément
- INSERM U563, Centre de Physiopathologie de Toulouse Purpan, IFR 30, Centre Hospitalier Purpan, 31024 Toulouse Cedex, France
| | - Emmanuelle Le Roy
- INSERM U563, Centre de Physiopathologie de Toulouse Purpan, IFR 30, Centre Hospitalier Purpan, 31024 Toulouse Cedex, France
| | - Jean-Luc Davignon
- INSERM U563, Centre de Physiopathologie de Toulouse Purpan, IFR 30, Centre Hospitalier Purpan, 31024 Toulouse Cedex, France
| |
Collapse
|
30
|
Martínez A, Alvarez-Lafuente R, Mas A, Bartolomé M, García-Montojo M, de Las Heras V, de la Concha EG, Arroyo R, Urcelay E. Environment-gene interaction in multiple sclerosis: human herpesvirus 6 and MHC2TA. Hum Immunol 2007; 68:685-9. [PMID: 17678724 DOI: 10.1016/j.humimm.2007.05.005] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2007] [Revised: 04/27/2007] [Accepted: 05/24/2007] [Indexed: 11/28/2022]
Abstract
Multiple sclerosis (MS) is an inflammatory disorder affecting the central nervous system, in which both genetic and environmental factors interact. Among these environmental contributors, herpesvirus has been proposed as an important etiologic factor. CIITA is a transcription factor controlling the expression of MHC class II genes, the main genetic determinants of MS susceptibility. This gene has been described as a target of the immunoevasive strategies, and it is therefore an attractive candidate gene to be at the genetic-viral crossroads. Two polymorphisms in MHC2TA gene (rs4,774G/C and rs3,087,456A/G) were studied in two groups: one in 22 multiple sclerosis patients with active human herpes virus 6 (HHV-6A) replication (HHV-6A-positive), and the other of 77 patients with no detectable HHV-6A active infection (HHV-6A-negative); a Spanish healthy control group (n = 520) was also included as external control. An association of the rs4,774C allele with the HHV-6A-positive group was found when compared with the HHV-6A-negative (47.7% vs 18.8%, p = 0.0001; odds ratio = 3.94) and also with the control group (47.7% vs 25.5%, p = 0.001, odds ratio = 2.67). No significant differences were observed between HHV-6A-negative subjects and healthy controls. Our data suggest that a strong gene-environment interaction occurs between HHV-6A active replication and MHC2TA rs4,774C or another polymorphism in tight linkage disequilibrium with it. Besides, this report indicates that when patients are grouped based upon a well-defined molecular event, complex diseases may reveal themselves as being constituted by distinct entities in which some genes may have a strong influence.
Collapse
Affiliation(s)
- Alfonso Martínez
- Clinical Immunology Department, Hospital Clínico San Carlos, Madrid, Spain.
| | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Wiertz EJ, Devlin R, Collins HL, Ressing ME. Herpesvirus interference with major histocompatibility complex class II-restricted T-cell activation. J Virol 2006; 81:4389-96. [PMID: 17079308 PMCID: PMC1900142 DOI: 10.1128/jvi.01525-06] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Affiliation(s)
- Emmanuel J Wiertz
- Department of Medical Microbiology, Leiden University Medical Center, P.O. Box 9600, 2300 RC Leiden, The Netherlands.
| | | | | | | |
Collapse
|
32
|
Niikura M, Kim T, Hunt HD, Burnside J, Morgan RW, Dodgson JB, Cheng HH. Marek's disease virus up-regulates major histocompatibility complex class II cell surface expression in infected cells. Virology 2006; 359:212-9. [PMID: 17028059 DOI: 10.1016/j.virol.2006.09.010] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2006] [Revised: 08/25/2006] [Accepted: 09/06/2006] [Indexed: 01/16/2023]
Abstract
Many herpesviruses modulate major histocompatibility complex (MHC) expression on the cell surface as an immune evasion mechanism. We report here that Marek's disease virus (MDV), a lymphotrophic avian alphaherpesvirus, up-regulates MHC class II cell surface expression in infected cells, contrary to all other herpesviruses examined to date. This MDV-induced class II up-regulation was detected both in vitro and in vivo. This effect was not solely an indirect effect of interferon, which is a highly potent natural inducer of MHC class II expression, since MHC class II up-regulation in cultured primary fibroblast cells was confined to the infected cells only. MHC class II up-regulation was also observed in infected cells of the bursa of Fabricius during the lytic phase of MDV infection in birds and upon reactivation of MDV from latency in an MDV-transformed cell line. As MDV is a strictly cell-associated virus and requires activated T cells for its life cycle, this up-regulation of MHC class II in infected cells may contribute to virus spread within the infected host by increasing the chance of contact between productively infected cells and susceptible activated T cells.
Collapse
Affiliation(s)
- Masahiro Niikura
- Department of Microbiology and Molecular Genetics, Michigan State University, East Lansing, MI 48824, USA
| | | | | | | | | | | | | |
Collapse
|
33
|
Reith W, LeibundGut-Landmann S, Waldburger JM. Regulation of MHC class II gene expression by the class II transactivator. Nat Rev Immunol 2005; 5:793-806. [PMID: 16200082 DOI: 10.1038/nri1708] [Citation(s) in RCA: 354] [Impact Index Per Article: 17.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
MHC class II molecules are pivotal for the adaptive immune system, because they guide the development and activation of CD4+ T helper cells. Fulfilling these functions requires that the genes encoding MHC class II molecules are transcribed according to a strict cell-type-specific and quantitatively modulated pattern. This complex gene-expression profile is controlled almost exclusively by a single master regulatory factor, which is known as the class II transactivator. As we discuss here, differential activation of the three independent promoters that drive expression of the gene encoding the class II transactivator ultimately determines the exquisitely regulated pattern of MHC class II gene expression.
Collapse
Affiliation(s)
- Walter Reith
- Department of Pathology and Immunology, University of Geneva Medical School, Centre Médical Universitaire, 1 Rue Michel-Servet, CH-1211, Geneva, Switzerland.
| | | | | |
Collapse
|
34
|
Cinatl J, Scholz M, Doerr HW. Role of tumor cell immune escape mechanisms in cytomegalovirus-mediated oncomodulation. Med Res Rev 2005; 25:167-85. [PMID: 15389728 DOI: 10.1002/med.20018] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
It has been known for a long time that cytomegalovirus (CMV) has evolved mechanisms that allow the escape from the host immune surveillance. In the past, many efforts have been done to elucidate the molecular mechanisms underlying this virus-mediated immune escape and thus virus persistence. However, it is unknown, whether CMV may also impair immune responses directed against tumor cells. This might have severe consequences on tumor progression and may explain the growing evidence for CMV-mediated oncomodulation. This review summarizes recent work on CMV-mediated immune escape mechanisms of tumor cells and oncomodulation and proposes novel aspects that may be important for understanding the CMV-associated tumor progression.
Collapse
Affiliation(s)
- Jindrich Cinatl
- Interdisziplinäres Labor für Tumor- und Virusforschung, Klinikum der Johann Wolfgang Goethe-Universität, Frankfurt am Main, Germany.
| | | | | |
Collapse
|
35
|
Rowshani AT, Bemelman FJ, van Leeuwen EMM, van Lier RAW, ten Berge IJM. Clinical and immunologic aspects of cytomegalovirus infection in solid organ transplant recipients. Transplantation 2005; 79:381-6. [PMID: 15729162 DOI: 10.1097/01.tp.0000148239.00384.f0] [Citation(s) in RCA: 128] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Primary cytomegalovirus (CMV) infection is a major cause of morbidity and mortality in recipients after solid organ transplantation (SOT). Widespread and prolonged use of antiviral drugs has changed the natural course of CMV disease by delaying its onset and causing drug resistance. CMV induces a strong cellular immune response, even in immunosuppressed patients, and has developed strategies to evade this immune surveillance. This review summarizes challenges in managing CMV infection in transplant recipients and highlights current insights in the cellular immune response against CMV.
Collapse
Affiliation(s)
- Ajda T Rowshani
- Department of Internal Medicine, Division of Clinical Immunology and Rheumatology, Academic Medical Center, University of Amsterdam, 1100 DE Amsterdam, the Netherlands.
| | | | | | | | | |
Collapse
|
36
|
Abstract
More than a half a century ago, interferons (IFN) were identified as antiviral cytokines. Since that discovery, IFN have been in the forefront of basic and clinical cytokine research. The pleiotropic nature of these cytokines continues to engage a large number of investigators to define their actions further. IFN paved the way for discovery of Janus tyrosine kinase (JAK)-signal transducing activators of transcription (STAT) pathways. A number of important tumor suppressive pathways are controlled by IFN. Several infectious pathogens counteract IFN-induced signaling pathways. Recent studies indicate that IFN activate several new protein kinases, including the MAP kinase family, and downstream transcription factors. This review not only details the established IFN signaling paradigms but also provides insights into emerging alternate signaling pathways and mechanisms of pathogen-induced signaling interference.
Collapse
Affiliation(s)
- Dhananjaya V Kalvakolanu
- Molecular and Cellular Biology Graduate Program, Greenebaum Cancer Center, Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, MD 21201, USA.
| |
Collapse
|
37
|
Popkin DL, Watson MA, Karaskov E, Dunn GP, Bremner R, Virgin HW. Murine cytomegalovirus paralyzes macrophages by blocking IFN gamma-induced promoter assembly. Proc Natl Acad Sci U S A 2003; 100:14309-14. [PMID: 14614150 PMCID: PMC283588 DOI: 10.1073/pnas.1835673100] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023] Open
Abstract
Macrophages (M phi) are activated by IFN gamma and are important cellular targets for infection by human and murine cytomegalovirus (MCMV), making it advantageous for CMVs to block IFN gamma-induced M phi differentiation. We found that MCMV infection inhibited IFN gamma regulation of many genes in M phi. MCMV infection blocked IFN gamma responses at the level of transcription without blocking Janus kinase/signal transducer and activator of transcription pathway activation and targeted IFN response factor 1- and class II transactivator-dependent and independent promoters. MCMV did not alter basal transcription from IFN gamma-responsive promoters and left the majority of cellular transcripts unchanged even after 48 h of infection. The effects of MCMV infection were specific to chromosomal rather than transiently transfected promoters. Characterization of the IFN gamma-responsive chromosomal class II transactivator promoter revealed that MCMV infection blocked IFN gamma-induced promoter assembly, allowing the virus to transcriptionally paralyze infected M phi responses while allowing basal transcription to proceed.
Collapse
Affiliation(s)
- Daniel L Popkin
- Department of Pathology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | | | | | | | | | | |
Collapse
|
38
|
Radosevich M, Ono SJ. Novel mechanisms of class II major histocompatibility complex gene regulation. Immunol Res 2003; 27:85-106. [PMID: 12637770 DOI: 10.1385/ir:27:1:85] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Class II MHC molecules present processed peptides from exogenous antigens to CD4+ helper T lymphocytes. In so doing, they are central to immunity, driving both the humoral and cell mediated arms of the immune response. Class II MHC molecules, and the genes encoding them, are expressed primarily in cells of the immune system (B cells, thymic epithelial cells, activated T cells and professional antigen presenting cells). The expression is also under developmental control. Research over the past 20 years have provided a clear understanding of the cis-elements and transcription factors that regulate the expression of Class II MHC genes. Perhaps the most critical advance has been the discovery of CIITA, a non- DNA binding activator of transcription that is a master control gene for class II gene expression. Current research is focused on understanding the situations where class II MHC gene expression occurs in a CIITA-independent pathway, and the molecular basis for this expression. Finally, significant emphasis is being placed on targeting class II MHC transcription factors to either inhibit or stimulate the immune response to transplanted tissue or in cell based vaccines. This communication outlines recent advances in this field and discusses likely areas for future research.
Collapse
Affiliation(s)
- Michael Radosevich
- Department of Immunology, Institute of Ophthalmology, University College London, University of London, London, UK
| | | |
Collapse
|
39
|
Hegde NR, Johnson DC. Human cytomegalovirus US2 causes similar effects on both major histocompatibility complex class I and II proteins in epithelial and glial cells. J Virol 2003; 77:9287-94. [PMID: 12915544 PMCID: PMC187418 DOI: 10.1128/jvi.77.17.9287-9294.2003] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The human cytomegalovirus (HCMV) glycoprotein US2 specifically binds to major histocompatibility complex (MHC) class I heavy chain (HC) and class II proteins DRalpha and DMalpha, triggering their degradation by proteasomes. Effects of US2 on class II proteins were originally characterized in HCMV- or adenovirus vector-infected U373 astroglioma cells. Here, we have extended characterization of US2-mediated degradation of class II DRalpha to two other cell lines, including biologically relevant epithelial cells. Comparison of the effects of US2 in cells expressing both class I and II proteins demonstrated only a slight preference for class I HC. Moreover, US2 caused degradation of DRalpha and DMalpha when these proteins were expressed by transfection without DRbeta, invariant chain (Ii), or DMbeta. Therefore, US2 binds to alpha chains of DR and DM and triggers endoplasmic reticulum degradation without formation of class II DR alphabeta/Ii or DM alphabeta complexes. Similar levels of degradation of class II alpha were observed in cells expressing vastly different amounts of class II, suggesting that cellular factors, other than class II, were limiting. We concluded that US2 has broad effects in a variety of cells that express both class I and II proteins and is relevant to HCMV infection in vivo.
Collapse
Affiliation(s)
- Nagendra R Hegde
- Department of Molecular Microbiology and Immunology, Oregon Health and Science University, Portland, Oregon 97239-3098, USA
| | | |
Collapse
|
40
|
Repique CJ, Li A, Brickey WJ, Ting JPY, Collins FM, Morris SL. Susceptibility of mice deficient in the MHC class II transactivator to infection with Mycobacterium tuberculosis. Scand J Immunol 2003; 58:15-22. [PMID: 12828554 DOI: 10.1046/j.1365-3083.2003.01266.x] [Citation(s) in RCA: 20] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Major histocompatibility complex (MHC) class II antigen presentation and subsequent CD4+ T-cell activation are critical for acquired immunity to Mycobacterium tuberculosis infection. MHC class II gene expression is primarily controlled by the master transactivator CIITA protein. Without functional CIITA protein, MHC class II expression is lost, impairing immune responses and increasing susceptibility to infection. In this study, we compared protective immune responses of CIITA-deficient mice and wild-type C57BL/6 controls with low dose aerosol M. tuberculosis infection. After aerogenic challenge, CIITA-/- mice failed to limit mycobacterial growth (2.5 and 2.0 log10 > WT lung and spleen CFUs, respectively, at day 58). Lung histopathology involved extensive necrosis, severe pneumonitis and overwhelming inflammation in the gene knockout mice. Mean survival time for CIITA-/- mice was significantly reduced (57 versus >300 days for WT). This extreme sensitivity to tuberculous infection was largely attributed to the absence of CD4+ cells. Flow cytometric studies detected virtually no CD4+ cells in CIITA-/- mouse spleens after infection versus elevated numbers in WT spleens. Failed CD4+ T-cell expansion markedly reduced interferon-gamma (IFN-gamma production in CIITA-/- mice versus WT controls. These results suggest the necessity of a functional CIITA pathway for controlling tuberculous infections and that interventions targeting CIITA expression may be useful antimycobacterial therapeutics.
Collapse
Affiliation(s)
- C J Repique
- Laboratory of Mycobacterial Diseases and Cellular Immunology, Center for Biologics Evaluation and Research, Food and Drug Administration, Bethesda, MD 20892, USA
| | | | | | | | | | | |
Collapse
|
41
|
Odeberg J, Plachter B, Brandén L, Söderberg-Nauclér C. Human cytomegalovirus protein pp65 mediates accumulation of HLA-DR in lysosomes and destruction of the HLA-DR alpha-chain. Blood 2003; 101:4870-7. [PMID: 12609847 DOI: 10.1182/blood-2002-05-1504] [Citation(s) in RCA: 67] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Human cytomegalovirus (HCMV) has developed multiple strategies to escape immune recognition. Here, we demonstrate that HCMV down-regulates HLA-DR expression in infected interferon gamma (IFN-gamma)-stimulated fibroblasts at 1 day after infection. Decreased HLA-DR expression was not observed on cells infected with an HCMV strain lacking the pp65 gene (RVAD65), but was observed on cells transfected with the pp65 gene. HLA-DR expression accumulated in vacuoles near the nucleus in HCMV-infected, but not in uninfected or RVAD65-infected cells. In addition, the HLA-DR alpha-chain, but not the beta-chain or HLA-DM, was degraded in HCMV-infected but not in RVAD65-infected cells. Thus, the HCMV protein pp65 mediates decreased expression of HLA-DR, by mediating an accumulation of HLA class II molecules in lysosomes that results in degradation of the HLA-DR alpha-chain.
Collapse
Affiliation(s)
- Jenny Odeberg
- Department of Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
| | | | | | | |
Collapse
|
42
|
Odeberg J, Browne H, Metkar S, Froelich CJ, Brandén L, Cosman D, Söderberg-Nauclér C. The human cytomegalovirus protein UL16 mediates increased resistance to natural killer cell cytotoxicity through resistance to cytolytic proteins. J Virol 2003; 77:4539-45. [PMID: 12663760 PMCID: PMC152133 DOI: 10.1128/jvi.77.8.4539-4545.2003] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Several reports have shown that human cytomegalovirus (HCMV)-infected cells are resistant to NK lysis. These studies have focused on receptor-ligand interactions, and different HCMV proteins have been indicated to mediate inhibitory NK signals. Here, we report that the HCMV protein UL16 is of major importance for the ability of HCMV-infected cells to resist NK cell-mediated cytotoxicity. Fibroblasts infected with the UL16 deletion mutant HCMV strain exhibited a 70% increased sensitivity to NK killing at 7 days postinfection compared to AD169-infected cells. Interestingly, HCMV-infected cells did not appear to engage inhibitory molecules on NK cells, since the levels of granzyme B were not reduced in supernatants obtained from NK cell cocultures with infected target cells compared to uninfected target cells. Furthermore, HCMV-infected cells, but not cells infected with the UL16 deletion mutant HCMV strain, exhibited a significantly increased resistance to the action of cytolytic proteins, including perforin, granzyme B, streptolysin O, and porcine NK lysin. In addition, fluorescence-activated cell sorting for UL16-positive transfected cells resulted in protection levels of 90% compared to control cells carrying the green fluorescent protein vector. Thus, the UL16 protein mediates an increased protection against the action of cytolytic proteins released by activated NK cells, possibly by a membrane-stabilizing mechanisms, rather than by delivering negative signals to NK cells.
Collapse
Affiliation(s)
- Jenny Odeberg
- Department of Medicine, Center for Molecular Medicine, Karolinska Institute, Stockholm, Sweden
| | | | | | | | | | | | | |
Collapse
|
43
|
Ilangumaran S, Finan D, La Rose J, Raine J, Silverstein A, De Sepulveda P, Rottapel R. A positive regulatory role for suppressor of cytokine signaling 1 in IFN-gamma-induced MHC class II expression in fibroblasts. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2002; 169:5010-20. [PMID: 12391216 DOI: 10.4049/jimmunol.169.9.5010] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Suppressor of cytokine signaling 1 (SOCS1) is rapidly induced following stimulation by several cytokines. SOCS1 negatively regulates cytokine receptor signal transduction by inhibiting Janus family tyrosine kinases. Lack of such feedback regulation underlies the premature death of SOCS1(-/-) mice due to unbridled IFN-gamma signaling. We used mouse embryo fibroblasts derived from SOCS1(-/-) mice to investigate the role of SOCS1 in IFN-gamma signaling pathways. SOCS1(-/-) fibroblasts were exquisitely sensitive to the IFN-gamma-mediated growth arrest and showed sustained STAT1 phosphorylation. However, SOCS1(-/-) fibroblasts were inefficient in MHC class II surface expression following IFN-gamma stimulation, despite a marked induction of the MHC class II transactivator and MHC class II gene expression. Retroviral transduction of wild-type SOCS1 relieved the growth-inhibitory effects of IFN-gamma in SOCS1(-/-) fibroblasts by inhibiting STAT1 activation. SOCS1R105K, carrying a mutation within the phosphotyrosine-binding pocket of the Src homology 2 domain, did not inhibit STAT1 phosphorylation, yet considerably inhibited IFN-gamma-mediated growth arrest. Strikingly, expression of SOCS1R105K restored the IFN-gamma-induced MHC class II expression in SOCS1(-/-) cells, indicating that expression of SOCS1 facilitates MHC class II expression in fibroblasts. Our results show that SOCS1, in addition to its negative regulatory role of inhibiting Janus kinases, has an unanticipated positive regulatory function in retarding the degradation of IFN-gamma-induced MHC class II proteins in fibroblasts.
Collapse
|
44
|
Hegde NR, Tomazin RA, Wisner TW, Dunn C, Boname JM, Lewinsohn DM, Johnson DC. Inhibition of HLA-DR assembly, transport, and loading by human cytomegalovirus glycoprotein US3: a novel mechanism for evading major histocompatibility complex class II antigen presentation. J Virol 2002; 76:10929-41. [PMID: 12368336 PMCID: PMC136637 DOI: 10.1128/jvi.76.21.10929-10941.2002] [Citation(s) in RCA: 88] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
Human cytomegalovirus (HCMV) establishes persistent lifelong infections and replicates slowly. To withstand robust immunity, HCMV utilizes numerous immune evasion strategies. The HCMV gene cassette encoding US2 to US11 encodes four homologous glycoproteins, US2, US3, US6, and US11, that inhibit the major histocompatibility complex class I (MHC-I) antigen presentation pathway, probably inhibiting recognition by CD8(+) T lymphocytes. US2 also inhibits the MHC-II antigen presentation pathway, causing degradation of human leukocyte antigen (HLA)-DR-alpha and -DM-alpha and preventing recognition by CD4(+) T cells. We investigated the effects of seven of the US2 to US11 glycoproteins on the MHC-II pathway. Each of the glycoproteins was expressed by using replication-defective adenovirus vectors. In addition to US2, US3 inhibited recognition of antigen by CD4(+) T cells by a novel mechanism. US3 bound to class II alpha/beta complexes in the endoplasmic reticulum (ER), reducing their association with Ii. Class II molecules moved normally from the ER to the Golgi apparatus in US3-expressing cells but were not sorted efficiently to the class II loading compartment. As a consequence, formation of peptide-loaded class II complexes was reduced. We concluded that US3 and US2 can collaborate to inhibit class II-mediated presentation of endogenous HCMV antigens to CD4(+) T cells, allowing virus-infected cells to resist recognition by CD4(+) T cells.
Collapse
Affiliation(s)
- Nagendra R Hegde
- Department of Molecular Microbiology and Immunology, Oregon Health and Science University, Portland, Oregon 97239, USA
| | | | | | | | | | | | | |
Collapse
|
45
|
Slobedman B, Mocarski ES, Arvin AM, Mellins ED, Abendroth A. Latent cytomegalovirus down-regulates major histocompatibility complex class II expression on myeloid progenitors. Blood 2002; 100:2867-73. [PMID: 12351397 DOI: 10.1182/blood.v100.8.2867] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Following primary infection, human cytomegalovirus (CMV) establishes a lifelong latent infection in bone marrow-derived myeloid lineage cells. Although downmodulation of major histocompatibility complex (MHC) class I and class II protein levels occurs during active viral replication, little is known about the modulation of these proteins during latent infection. When analyzed by flow cytometry, latently infected adherent cells collected from granulocyte macrophage progenitor (GM-P) cultures exhibited a striking reduction in MHC class II antigen present on the cell surface starting very early after exposure to virus that continued for more than 2 weeks. In comparison, cell surface levels of the monocyte cell surface marker CD14 remained unaltered in these cells. A recombinant virus (RV798) lacking the virus genes US2-US11 retained the ability to downmodulate MHC class II levels during latent infection. Immunoblot and immunofluorescent antibody staining analyses showed that the reduction in MHC class II surface levels during latency was associated with a block in protein trafficking. HLA-DR was retained within cytoplasmic vesicles that also contained HLA-DM. Thus, downmodulation remained independent of all previously characterized MHC class I and class II immunomodulatory viral gene products and involved a mechanism not previously ascribed to any viral function. These data show that latent infection is accompanied by reduced cell surface expression of MHC class II proteins, a strategy that would afford the virus escape from immunosurveillance and increase the chances for lifelong latent infection.
Collapse
Affiliation(s)
- Barry Slobedman
- Center for Virus Research, Westmead Millennium Institute and University of Sydney, Westmead, Australia.
| | | | | | | | | |
Collapse
|
46
|
Vaz-Santiago J, Lulé J, Rohrlich P, Kravtzoff R, Le Roy E, Davignon JL, Betbeder D, Davrinche C. IE1-pp65 recombinant protein from human CMV combined with a nanoparticulate carrier, SMBV, as a potential source for the development of anti-human CMV adoptive immunotherapy. Cytotherapy 2002; 4:11-9. [PMID: 11953037 DOI: 10.1080/146532402317251482] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/16/2022]
Abstract
BACKGROUND Human cytomegalovirus (HCMV) infection and reactivation following allogeneic bone marrow transplantation is a major source of complications in grafted patients including pneumonitis, graft rejection and even death. Adoptive immunotherapy consisting in transfer of CD4(+) and CD8(+) T cells directed against HCMV has proved its worth. Nevertheless, established procedures have to be improved in terms of safety and waiting period required to obtain specific T cells. METHODS As an alternative to infectious virus used in current strategies, we purified a recombinant protein IE1-pp65 resulting from the fusion of the regulatory IE1 and matrix pp65 proteins, both known as the major targets of the overall anti-HCMV T cell response. Based on our previous data demonstrating its use for in vitro stimulation and expansion of anti-HCMV CD4(+) and CD8(+) T cells (Vaz-Santiago et al, 2001, J.Virol, 75:7840-47) from peripheral blood mononuclear cells (PBMC) of seropositive donors, we planned to improve its in vitro immunogenicity through association with a nanoparticulate carrier, SMBV. RESULTS We demonstrated that using of SMBV/IE1-pp65 formulation allowed to potentiate in vitro activation of T cells and to expand more CD8(+) T cells than with soluble IE1-pp65, following stimulation of PBMC. DISCUSSION These data suggest the use of SMBV/IE1-pp65 formulation as a potential source of antigen for efficient T cells expansion in the development of safe anti-HCMV immunotherapy.
Collapse
|
47
|
Le Roy E, Baron M, Faigle W, Clément D, Lewinsohn DM, Streblow DN, Nelson JA, Amigorena S, Davignon JL. Infection of APC by human cytomegalovirus controlled through recognition of endogenous nuclear immediate early protein 1 by specific CD4(+) T lymphocytes. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2002; 169:1293-301. [PMID: 12133951 DOI: 10.4049/jimmunol.169.3.1293] [Citation(s) in RCA: 19] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Infections by human CMV are controlled by cellular immune responses. Professional APC such as monocytes and macrophages can be infected in vivo and are considered as a reservoir of virus. However, CMV-specific CD4(+) responses against infected APC have not been reported. To develop a model of CD4-infected APC interaction, we have transfected the U373MG astrocytoma cell line with the class II transactivator (CIITA). Confocal microscopy experiments showed that U373MG-CIITA cells expressed markers characteristic of APC. Functional assays demonstrated that infected U373MG-CIITA APC processed and presented both exogenous and endogenously neosynthesized nuclear immediate early (IE) protein 1 through the MHC class II pathway. More importantly, endogenous presentation of IE1 by infected APC lead to efficient control of CMV infection as revealed by decreased viral titer. Thus, these results describe the endogenous presentation of a nuclear viral protein by the MHC class II pathway and suggest that IE1-specific CD4(+) T cells may play an important role in CMV infection by directly acting against infected APC.
Collapse
Affiliation(s)
- Emmanuelle Le Roy
- Institut National de la Santé et de la Recherche Médicale, Toulouse, France
| | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Odeberg J, Cerboni C, Browne H, Kärre K, Möller E, Carbone E, Söderberg-Nauclér C. Human cytomegalovirus (HCMV)-infected endothelial cells and macrophages are less susceptible to natural killer lysis independent of the downregulation of classical HLA class I molecules or expression of the HCMV class I homologue, UL18. Scand J Immunol 2002; 55:149-61. [PMID: 11896931 DOI: 10.1046/j.1365-3083.2002.01025.x] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
A number of reports have suggested that human cytomegalovirus (HCMV)-infected fibroblasts are resistant to natural killer (NK) lysis, and that the HCMV-encoded human leucocyte antigen (HLA) class I homologue UL18 may be responsible for this effect. While fibroblasts are easy to infect in vitro, their role in HCMV pathogenesis in vivo is unclear. Here, we have established systems to address NK recognition of infected endothelial cells and macrophages, two important HCMV cellular reservoirs in vivo. The HCMV-infected endothelial cells exhibited increased resistance to NK killing, and, in most experiments, infected macrophages demonstrated a decreased susceptibility to NK lysis. Infection with the mutant HCMV strain RV670, lacking the genes US1-9 and US11 that are responsible for downregulation of HLA class I molecules, also led to decreased NK susceptibility. Furthermore, reduced NK susceptibility was independent of the expression of the HLA class I homologue UL18, since cells infected with the UL18Delta HCMV strain were also less susceptible to NK killing. These results suggest that HCMV-induced resistance to NK cytotoxicity in endothelial cells and macrophages is independent of known pathways that interfere with the expression of cellular HLA class I A, B and C surface antigens and the HCMV encoded class I homologue UL18.
Collapse
Affiliation(s)
- J Odeberg
- Division of Immunology, Karolinska Institute, Huddinge Hospital, Sweden
| | | | | | | | | | | | | |
Collapse
|
49
|
Abstract
The dynamics of the relationship between the immune system and latent viruses are highly complex. Latent viruses not only avoid elimination by the host's primary immune response, they also remain with the host for life in the presence of strong acquired immunity, often exhibiting periodic reactivation and recurrence from the latent state. The continual battle between reemergent infectious virus and immunological memory cells provides an essential virus-host regulatory loop in latency. In this review, we speculate on the critical importance of immune interference mechanisms by viruses contributing to the regulatory loop in viral homeostasis of latency. Central to the notion of viral homeostasis, we further invoke the concept of threshold limits in naive and memory states of immunity to account for the failure of the host to completely eradicate these intracellular parasites.
Collapse
Affiliation(s)
- S Redpath
- Department of Immunology, The Scripps Research Institute, La Jolla, California 92037, USA.
| | | | | | | |
Collapse
|
50
|
Miller DM, Cebulla CM, Sedmak DD. Human cytomegalovirus inhibition of major histocompatibility complex transcription and interferon signal transduction. Curr Top Microbiol Immunol 2002; 269:153-70. [PMID: 12224507 DOI: 10.1007/978-3-642-59421-2_10] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Pathogens have evolved diverse mechanisms for escaping host innate and adaptive immunity. Viruses that maintain a persistent infection are particularly effective at disabling key arms of the host immune response. For example, the herpesviruses establish a persistent infection in human and animal hosts, in part through critical immunoevasive strategies. Cytomegalovirus, a beta-herpesvirus, impairs major histocompatibility complex (MHC) class I and class II antigen presentation by decreasing MHC expression on the surface of the infected cell, thus enabling infected cells to escape CD8+ and CD4+ T lymphocyte immunosurveillance. Moreover, cytomegalovirus blocks the interferon signal transduction pathway, thereby limiting the direct and indirect antiviral effects of the interferons. In this review, we focus on an emerging paradigm in which the effectiveness of viruses, particularly human cytomegalovirus, to escape antiviral immune responses is significantly enhanced by their ability to inhibit MHC transcription and interferon (IFN)-stimulated (JAK/STAT) signal transduction.
Collapse
Affiliation(s)
- D M Miller
- Department of Pathology, Ohio State University College of Medicine, 1645 Neil Avenue, Room 129, Columbus, OH 43210, USA
| | | | | |
Collapse
|