8 research outputs found

    Regulation of cellular immunity by human cytomegalovirus

    Get PDF
    The success of HCMV as a lifelong pathogen is attributed at least in part to the broad range of encoded immune evasion molecules that inhibit the host cellular immune response. Indeed, HCMV has become a paradigm for immune evasion, the study of which has revealed a number of basic immunological processes. To screen for novel immune evasion genes, HCMV-specific CD8+ T-cell lines were grown from seropositive donors and used against a series of block deletion viruses, each missing a region of genes non-essential for replication in vitro. UL13-UL20 was flagged as important for inhibition of CD8+ T-cells. Further screening with individual gene knockout HCMVs showed that the published NK-cell inhibitor UL16 could inhibit CD8+ T-cells, but also revealed UL19 as a previously unrecognised strong immune evasin, inhibiting 3 separate CD8+ T-cell lines. UL19 had no effect on HLA-I downregulation indicating that it may affect other pathways involved with T-cell activation. Proteomic data showed that surface TNFR2 was increased by HCMV infection. This is important as this would influence the response to TNF, a major inflammatory cytokine and soluble effector molecule released by T and NK cells. Screening using different HCMV strains and knockout viruses identified UL148 and UL148D as responsible for the increase in surface TNFR2 but prevented the release of soluble TNFR2, indicating that UL148 and UL148D were influencing the ability of TNFR2 to be retained at the cell surface. Infection with HCMV Merlin profoundly downregulated surface ADAM17, the metalloproteinase responsible for cleaving TNFR2 from the cell surface. Deleting UL148 and UL148D recovered ADAM17 expression, blocking the function of which returned surface and soluble TNFR2 levels to those observed with Merlin. This was also true of TNFR1. HCMV infected cell lysates showed that UL148 and UL148D interfered with the maturation of ADAM17. Thus, UL148 and UL148D allow upregulation of TNFR2 and maintain TNFR1 expression during and HCMV infection by impairing surface ADAM17 expression through impairment of ADAM17 maturation. Given that ADAM17 is involved with the cleaving of multiple cytokines, cytokine receptors, adhesion molecules and immune cell receptors, this work identifies a novel mechanism through which HCMV can alter the surface and soluble proteome by preventing the shedding of inflammatory/immune receptors and mediators. More detailed studies will be required to define the global impact of this on the immune system

    HCMV-encoded NK modulators: Lessons from in vitro and in vivo genetic variation

    Get PDF
    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

    Downregulation of HLA-I by the molluscum contagiosum virus mc080 impacts NK-cell recognition and promotes CD8+ T-cell evasion.

    Get PDF
    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

    Suppression of costimulation by human cytomegalovirus promotes evasion of cellular immune defenses.

    Get PDF
    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

    ADAM17 targeting by human cytomegalovirus remodels the cell surface proteome to simultaneously regulate multiple immune pathways

    Get PDF
    Human cytomegalovirus (HCMV) is a major human pathogen whose life-long persistence is enabled by its remarkable capacity to systematically subvert host immune defenses. In exploring the finding that HCMV infection up-regulates tumor necrosis factor receptor 2 (TNFR2), a ligand for the pro-inflammatory antiviral cytokine TNFα, we found that the underlying mechanism was due to targeting of the protease, A Disintegrin And Metalloproteinase 17 (ADAM17). ADAM17 is the prototype ‘sheddase’, a family of proteases that cleaves other membrane-bound proteins to release biologically active ectodomains into the supernatant. HCMV impaired ADAM17 surface expression through the action of two virally-encoded proteins in its UL/b’ region, UL148 and UL148D. Proteomic plasma membrane profiling of cells infected with an HCMV double-deletion mutant for UL148 and UL148D with restored ADAM17 expression, combined with ADAM17 functional blockade, showed that HCMV stabilized the surface expression of 114 proteins (P < 0.05) in an ADAM17-dependent fashion. These included reported substrates of ADAM17 with established immunological functions such as TNFR2 and jagged1, but also numerous unreported host and viral targets, such as nectin1, UL8, and UL144. Regulation of TNFα-induced cytokine responses and NK inhibition during HCMV infection were dependent on this impairment of ADAM17. We therefore identify a viral immunoregulatory mechanism in which targeting a single sheddase enables broad regulation of multiple critical surface receptors, revealing a paradigm for viral-encoded immunomodulation
    corecore