Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters










Database
Language
Publication year range
1.
Proc Natl Acad Sci U S A ; 120(49): e2309077120, 2023 Dec 05.
Article in English | MEDLINE | ID: mdl-38011551

ABSTRACT

Human cytomegalovirus (HCMV) is a paradigm of pathogen immune evasion and sustains lifelong persistent infection in the face of exceptionally powerful host immune responses through the concerted action of multiple immune-evasins. These reduce NK cell activation by inhibiting ligands for activating receptors, expressing ligands for inhibitory receptors, or inhibiting synapse formation. However, these functions only inhibit direct interactions with the infected cell. To determine whether the virus also expresses soluble factors that could modulate NK function at a distance, we systematically screened all 170 HCMV canonical protein-coding genes. This revealed that UL4 encodes a secreted and heavily glycosylated protein (gpUL4) that is expressed with late-phase kinetics and is capable of inhibiting NK cell degranulation. Analyses of gpUL4 binding partners by mass spectrometry identified an interaction with TRAIL. gpUL4 bound TRAIL with picomolar affinity and prevented TRAIL from binding its receptor, thus acting as a TRAIL decoy receptor. TRAIL is found in both soluble and membrane-bound forms, with expression of the membrane-bound form strongly up-regulated on NK cells in response to interferon. gpUL4 inhibited apoptosis induced by soluble TRAIL, while also binding to the NK cell surface in a TRAIL-dependent manner, where it blocked NK cell degranulation and cytokine secretion. gpUL4 therefore acts as an immune-evasin by inhibiting both soluble and membrane-bound TRAIL and is a viral-encoded TRAIL decoy receptor. Interestingly, gpUL4 could also suppress NK responses to heterologous viruses, suggesting that it may act as a systemic virally encoded immunosuppressive agent.


Subject(s)
Cytomegalovirus , Killer Cells, Natural , Humans , Cytomegalovirus/physiology , Immune Evasion , Glycoproteins/metabolism , Apoptosis
2.
J Gen Virol ; 101(8): 863-872, 2020 08.
Article in English | MEDLINE | ID: mdl-32510303

ABSTRACT

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


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Down-Regulation/immunology , Histocompatibility Antigens Class I/immunology , Immune Evasion/immunology , Killer Cells, Natural/immunology , Molluscum contagiosum virus/immunology , Antigen Presentation/immunology , Cell Line , Endoplasmic Reticulum/immunology , Host-Pathogen Interactions/immunology , Humans , Keratinocytes/immunology , T-Lymphocytes, Cytotoxic/immunology , Viral Proteins/immunology
3.
Front Immunol ; 9: 2214, 2018.
Article in English | MEDLINE | ID: mdl-30327650

ABSTRACT

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


Subject(s)
Cytomegalovirus Infections/metabolism , Cytomegalovirus/genetics , Cytomegalovirus/metabolism , Killer Cells, Natural/metabolism , Antigens, Differentiation, T-Lymphocyte/metabolism , Chromosomes, Artificial, Bacterial/genetics , Cytomegalovirus Infections/prevention & control , Genetic Variation , Genomic Instability , Histocompatibility Antigens Class I/metabolism , Humans , Immune Evasion , Lymphocyte Activation , Membrane Glycoproteins/genetics , Membrane Glycoproteins/metabolism , Receptor Activator of Nuclear Factor-kappa B/metabolism , Receptors, TNF-Related Apoptosis-Inducing Ligand/metabolism , Viral Proteins/genetics , Viral Proteins/metabolism
4.
Proc Natl Acad Sci U S A ; 115(19): 4998-5003, 2018 05 08.
Article in English | MEDLINE | ID: mdl-29691324

ABSTRACT

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


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Cytomegalovirus Infections/immunology , Cytomegalovirus/immunology , Immune Evasion , Immunity, Cellular , Killer Cells, Natural/immunology , Viral Fusion Proteins/immunology , CD8-Positive T-Lymphocytes/pathology , Cell Line, Transformed , Cytomegalovirus/genetics , Cytomegalovirus Infections/genetics , Cytomegalovirus Infections/pathology , Humans , Killer Cells, Natural/pathology , Viral Fusion Proteins/genetics
5.
J Virol ; 89(2): 1479-83, 2015 Jan 15.
Article in English | MEDLINE | ID: mdl-25392213

ABSTRACT

The effect of abrogating the interferon (IFN) response on human cytomegalovirus (HCMV) replication was investigated using primary human cells engineered to block either the production of or the response to type I IFNs. In IFN-deficient cells, HCMV produced larger plaques and spread and replicated more rapidly than in parental cells. These cells demonstrate the vital role of IFNs in controlling HCMV replication and provide useful tools to investigate the IFN response to HCMV.


Subject(s)
Cytomegalovirus/immunology , Cytomegalovirus/physiology , Interferon Type I/immunology , Virus Replication , Cells, Cultured , Humans , Interferon Type I/deficiency , Viral Plaque Assay
6.
J Virol ; 88(18): 10990-4, 2014 Sep.
Article in English | MEDLINE | ID: mdl-25008927

ABSTRACT

Regulation of the lectin galectin 9 (Gal-9) was investigated for the first time during human cytomegalovirus (HCMV) infection. Gal-9 transcription was significantly upregulated in transplant recipients with reactivated HCMV in vivo. In vitro, Gal-9 was potently upregulated by HCMV independently of viral gene expression, with interferon beta (IFN-ß) identified as the mediator of this effect. This study defines an immunoregulatory protein potently increased by HCMV infection and a novel mechanism to control Gal-9 through IFN-ß induction.


Subject(s)
Cytomegalovirus Infections/genetics , Cytomegalovirus/physiology , Galectins/genetics , Interferon-beta/metabolism , Up-Regulation , Cytomegalovirus Infections/metabolism , Cytomegalovirus Infections/virology , Galectins/metabolism , Host-Pathogen Interactions , Humans , Interferon-beta/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...