Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Type of study
Publication year range
1.
Sci Adv ; 10(19): eadm7515, 2024 May 10.
Article in English | MEDLINE | ID: mdl-38728394

ABSTRACT

The nonpolymorphic major histocompatibility complex E (MHC-E) molecule is up-regulated on many cancer cells, thus contributing to immune evasion by engaging inhibitory NKG2A/CD94 receptors on NK cells and tumor-infiltrating T cells. To investigate whether MHC-E expression by cancer cells can be targeted for MHC-E-restricted T cell control, we immunized rhesus macaques (RM) with rhesus cytomegalovirus (RhCMV) vectors genetically programmed to elicit MHC-E-restricted CD8+ T cells and to express established tumor-associated antigens (TAAs) including prostatic acidic phosphatase (PAP), Wilms tumor-1 protein, or Mesothelin. T cell responses to all three tumor antigens were comparable to viral antigen-specific responses with respect to frequency, duration, phenotype, epitope density, and MHC restriction. Thus, CMV-vectored cancer vaccines can bypass central tolerance by eliciting T cells to noncanonical epitopes. We further demonstrate that PAP-specific, MHC-E-restricted CD8+ T cells from RhCMV/PAP-immunized RM respond to PAP-expressing HLA-E+ prostate cancer cells, suggesting that the HLA-E/NKG2A immune checkpoint can be exploited for CD8+ T cell-based immunotherapies.


Subject(s)
Antigens, Neoplasm , CD8-Positive T-Lymphocytes , HLA-E Antigens , Histocompatibility Antigens Class I , Macaca mulatta , Animals , CD8-Positive T-Lymphocytes/immunology , Histocompatibility Antigens Class I/immunology , Histocompatibility Antigens Class I/metabolism , Antigens, Neoplasm/immunology , Humans , Cancer Vaccines/immunology , Antigen Presentation/immunology , Cell Line, Tumor , Male , Cytomegalovirus/immunology , Mesothelin , Acid Phosphatase
2.
J Immunol ; 204(8): 2169-2176, 2020 04 15.
Article in English | MEDLINE | ID: mdl-32161099

ABSTRACT

Currently 247 million people are living with chronic hepatitis B virus infection (CHB), and the development of novel curative treatments is urgently needed. Immunotherapy is an attractive approach to treat CHB, yet therapeutic approaches to augment the endogenous hepatitis B virus (HBV)-specific T cell response in CHB patients have demonstrated little success. In this study, we show that strain 68-1 rhesus macaque (RM) CMV vaccine vectors expressing HBV Ags engender HBV-specific CD8+ T cells unconventionally restricted by MHC class II and the nonclassical MHC-E molecule in RM. Surface staining of human donor and RM primary hepatocytes (PH) ex vivo revealed the majority of PH expressed MHC-E but not MHC class II. HBV-specific, MHC-E-restricted CD8+ T cells from RM vaccinated with RM CMV vaccine vectors expressing HBV Ags recognized HBV-infected PH from both human donor and RM. These results provide proof-of-concept that MHC-E-restricted CD8+ T cells could be harnessed for the treatment of CHB, either through therapeutic vaccination or adoptive immunotherapy.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Hepatitis B virus/immunology , Hepatitis B, Chronic/immunology , Hepatocytes/immunology , Histocompatibility Antigens Class I/immunology , Animals , Hepatitis B, Chronic/virology , Hepatocytes/virology , Macaca mulatta
3.
PLoS Pathog ; 12(12): e1006072, 2016 Dec.
Article in English | MEDLINE | ID: mdl-27977791

ABSTRACT

Cytomegalovirus (CMV) elicits long-term T-cell immunity of unparalleled strength, which has allowed the development of highly protective CMV-based vaccine vectors. Counterintuitively, experimental vaccines encoding a single MHC-I restricted epitope offered better immune protection than those expressing entire proteins, including the same epitope. To clarify this conundrum, we generated recombinant murine CMVs (MCMVs) encoding well-characterized MHC-I epitopes at different positions within viral genes and observed strong immune responses and protection against viruses and tumor growth when the epitopes were expressed at the protein C-terminus. We used the M45-encoded conventional epitope HGIRNASFI to dissect this phenomenon at the molecular level. A recombinant MCMV expressing HGIRNASFI on the C-terminus of M45, in contrast to wild-type MCMV, enabled peptide processing by the constitutive proteasome, direct antigen presentation, and an inflation of antigen-specific effector memory cells. Consequently, our results indicate that constitutive proteasome processing of antigenic epitopes in latently infected cells is required for robust inflationary responses. This insight allows utilizing the epitope positioning in the design of CMV-based vectors as a novel strategy for enhancing their efficacy.


Subject(s)
Antigens, Viral/immunology , CD8-Positive T-Lymphocytes/immunology , Herpesviridae Infections/immunology , Immunodominant Epitopes/immunology , Viral Vaccines/immunology , Animals , Antigens, Viral/metabolism , Chromatography, Liquid , Epitopes, T-Lymphocyte/immunology , Flow Cytometry , Immunologic Memory/immunology , Lymphocyte Activation/immunology , Mass Spectrometry , Mice , Muromegalovirus/immunology , Mutagenesis, Site-Directed , Peptides , Vaccines, Synthetic/immunology , Viral Proteins/immunology , Viral Proteins/metabolism
4.
PLoS Pathog ; 7(11): e1002332, 2011 Nov.
Article in English | MEDLINE | ID: mdl-22072961

ABSTRACT

Interferon-induced BST2/Tetherin prevents budding of vpu-deficient HIV-1 by tethering mature viral particles to the plasma membrane. BST2 also inhibits release of other enveloped viruses including Ebola virus and Kaposi's sarcoma associated herpesvirus (KSHV), indicating that BST2 is a broadly acting antiviral host protein. Unexpectedly however, recovery of human cytomegalovirus (HCMV) from supernatants of BST2-expressing human fibroblasts was increased rather than decreased. Furthermore, BST2 seemed to enhance viral entry into cells since more virion proteins were released into BST2-expressing cells and subsequent viral gene expression was elevated. A significant increase in viral entry was also observed upon induction of endogenous BST2 during differentiation of the pro-monocytic cell line THP-1. Moreover, treatment of primary human monocytes with siRNA to BST2 reduced HCMV infection, suggesting that BST2 facilitates entry of HCMV into cells expressing high levels of BST2 either constitutively or in response to exogenous stimuli. Since BST2 is present in HCMV particles we propose that HCMV entry is enhanced via a reverse-tethering mechanism with BST2 in the viral envelope interacting with BST2 in the target cell membrane. Our data suggest that HCMV not only counteracts the well-established function of BST2 as inhibitor of viral egress but also employs this anti-viral protein to gain entry into BST2-expressing hematopoietic cells, a process that might play a role in hematogenous dissemination of HCMV.


Subject(s)
Antigens, CD/metabolism , Cytomegalovirus/physiology , Virus Internalization , Virus Release , Antigens, CD/genetics , Cell Line , Ebolavirus/physiology , GPI-Linked Proteins/genetics , GPI-Linked Proteins/metabolism , Gene Expression Regulation, Viral , HEK293 Cells , HIV-1/physiology , Herpesvirus 8, Human/physiology , Humans , Monocytes/virology , RNA Interference , RNA, Small Interfering
5.
PLoS One ; 5(12): e15132, 2010 Dec 02.
Article in English | MEDLINE | ID: mdl-21151997

ABSTRACT

Membrane-associated RING-CH (MARCH) proteins represent a family of transmembrane ubiquitin ligases modulating intracellular trafficking and turnover of transmembrane protein targets. While homologous proteins encoded by gamma-2 herpesviruses and leporipoxviruses have been studied extensively, limited information is available regarding the physiological targets of cellular MARCH proteins. To identify host cell proteins targeted by the human MARCH-VIII ubiquitin ligase we used stable isotope labeling of amino-acids in cell culture (SILAC) to monitor MARCH-dependent changes in the membrane proteomes of human fibroblasts. Unexpectedly, we observed that MARCH-VIII reduced the surface expression of Bap31, a chaperone that predominantly resides in the endoplasmic reticulum (ER). We demonstrate that Bap31 associates with the transmembrane domains of several MARCH proteins and controls intracellular transport of MARCH proteins. In addition, we observed that MARCH-VIII reduced the surface expression of the hyaluronic acid-receptor CD44 and both MARCH-VIII and MARCH-IV sequestered the tetraspanin CD81 in endo-lysosomal vesicles. Moreover, gene knockdown of MARCH-IV increased surface levels of endogenous CD81 suggesting a constitutive involvement of this family of ubiquitin ligases in the turnover of tetraspanins. Our data thus suggest a role of MARCH-VIII and MARCH-IV in the regulated turnover of CD81 and CD44, two ubiquitously expressed, multifunctional proteins.


Subject(s)
Antigens, CD/metabolism , Hyaluronan Receptors/metabolism , Lysosomes/metabolism , Membrane Proteins/metabolism , Cell Membrane/metabolism , Endosomes/metabolism , Fibroblasts/metabolism , HeLa Cells , Humans , Hyaluronic Acid/chemistry , Membrane Glycoproteins/metabolism , Models, Biological , Proteome , Proteomics/methods , Tandem Mass Spectrometry/methods , Tetraspanin 28 , Tetraspanin 29
6.
PLoS Pathog ; 6(5): e1000913, 2010 May 13.
Article in English | MEDLINE | ID: mdl-20485522

ABSTRACT

The interferon-induced BST-2 protein has the unique ability to restrict the egress of HIV-1, Kaposi's sarcoma-associated herpesvirus (KSHV), Ebola virus, and other enveloped viruses. The observation that virions remain attached to the surface of BST-2-expressing cells led to the renaming of BST-2 as "tetherin". However, viral proteins such as HIV-1 Vpu, simian immunodeficiency virus Nef, and KSHV K5 counteract BST-2, thereby allowing mature virions to readily escape from infected cells. Since the anti-viral function of BST-2 was discovered, there has been an explosion of research into several aspects of this intriguing interplay between host and virus. This review focuses on recent work addressing the molecular mechanisms involved in BST-2 restriction of viral egress and the species-specific countermeasures employed by various viruses.


Subject(s)
Antigens, CD/metabolism , HIV Infections/metabolism , HIV Infections/virology , HIV-1/growth & development , Membrane Glycoproteins/metabolism , Antigens, CD/genetics , GPI-Linked Proteins , Humans , Membrane Glycoproteins/genetics , Virion/metabolism , Virus Diseases/metabolism , Virus Diseases/virology
7.
J Virol ; 83(19): 9672-81, 2009 Oct.
Article in English | MEDLINE | ID: mdl-19605472

ABSTRACT

K3/MIR1 and K5/MIR2 of Kaposi's sarcoma-associated herpesvirus (KSHV) are viral members of the membrane-associated RING-CH (MARCH) ubiquitin ligase family and contribute to viral immune evasion by directing the conjugation of ubiquitin to immunostimulatory transmembrane proteins. In a quantitative proteomic screen for novel host cell proteins downregulated by viral immunomodulators, we previously observed that K5, as well as the human immunodeficiency virus type 1 (HIV-1) immunomodulator VPU, reduced steady-state levels of bone marrow stromal cell antigen 2 (BST2; also called CD317 or tetherin), suggesting that BST2 might be a novel substrate of K5 and VPU. Recent work revealed that in the absence of VPU, HIV-1 virions are tethered to the plasma membrane in BST2-expressing HeLa cells. By targeting BST2, K5 might thus similarly overcome an innate antiviral host defense mechanism. Here we establish that despite its type II transmembrane topology and carboxy-terminal glycosylphosphatidylinositol (GPI) anchor, BST2 represents a bona fide target of K5 that is downregulated during primary infection by and reactivation of KSHV. Upon exit of the protein from the endoplasmic reticulum, lysines in the short amino-terminal domain of BST2 are ubiquitinated by K5, resulting in rapid degradation of BST2. Ubiquitination of BST2 is required for degradation, since BST2 lacking cytosolic lysines was K5 resistant and ubiquitin depletion by proteasome inhibitors restored BST2 surface expression. Thus, BST2 represents the first type II transmembrane protein targeted by K5 and the first example of a protein that is both ubiquitinated and GPI linked. We further demonstrate that KSHV release is decreased in the absence of K5 in a BST2-dependent manner, suggesting that K5 contributes to the evasion of intracellular antiviral defense programs.


Subject(s)
Antigens, CD/biosynthesis , Gene Expression Regulation, Viral , Herpesvirus 8, Human/metabolism , Immediate-Early Proteins/biosynthesis , Membrane Glycoproteins/biosynthesis , Viral Proteins/biosynthesis , Biotinylation , Cells, Cultured , Endothelial Cells/virology , GPI-Linked Proteins , HeLa Cells , Humans , Microcirculation , Models, Biological , Polymerase Chain Reaction , Proteasome Endopeptidase Complex/metabolism
8.
J Virol ; 82(19): 9615-28, 2008 Oct.
Article in English | MEDLINE | ID: mdl-18667499

ABSTRACT

Vascular endothelial cadherin (VE-cadherin) connects neighboring endothelial cells (ECs) via interendothelial junctions and regulates EC proliferation and adhesion during vasculogenesis and angiogenesis. The cytoplasmic domain of VE-cadherin recruits alpha- and beta-catenins and gamma-catenin, which interact with the actin cytoskeleton, thus modulating cell morphology. Dysregulation of the adherens junction/cytoskeletal axis is a hallmark of invasive tumors. We now demonstrate that the transmembrane ubiquitin ligase K5/MIR-2 of Kaposi's sarcoma-associated herpesvirus targets VE-cadherin for ubiquitin-mediated destruction, thus disturbing EC adhesion. In contrast, N-cadherin levels in K5-expressing cells were increased compared to those in control cells. Steady-state levels of alpha- and beta-catenins and gamma-catenin in K5-expressing ECs were drastically reduced due to proteasomal destruction. Moreover, the actin cytoskeleton was rearranged, resulting in the dysregulation of EC barrier function as measured by electric cell-substrate impedance sensing. Our data represent the first example of a viral protein targeting adherens junction proteins and suggest that K5 contributes to EC proliferation, vascular leakage, and the reprogramming of the EC proteome during Kaposi's sarcoma tumorigenesis.


Subject(s)
Adherens Junctions/metabolism , Gene Expression Regulation, Viral , Herpesvirus 8, Human/metabolism , Actins/metabolism , Biotinylation , Catenins/metabolism , Cell Adhesion , Cell Membrane/metabolism , Cell Proliferation , Cell Survival , Cytoskeleton/metabolism , Electric Impedance , Humans , Skin/metabolism , Ubiquitin/metabolism
9.
Blood ; 108(6): 1932-40, 2006 Sep 15.
Article in English | MEDLINE | ID: mdl-16601245

ABSTRACT

The transmembrane ubiquitin ligase K5/MIR2 of Kaposi sarcoma herpesvirus (KSHV) mediates internalization and lysosomal degradation of glycoproteins involved in antigen presentation and co-stimulation. In endothelial cells (ECs), K5 additionally reduced expression of CD31/platelet-endothelial cell adhesion molecule (PECAM), an adhesion molecule regulating cell-cell interactions of ECs, platelets, monocytes, and T cells. K5 also reduced EC migration, a CD31-dependent process. Unlike other K5 substrates, both newly synthesized and pre-existing CD31 molecules were targeted by K5. K5 was transported to the cell surface and ubiquitinated pre-existing CD31, resulting in endocytosis and lysosomal degradation. In the endoplasmic reticulum, newly synthesized CD31 was degraded by proteasomes, which required binding of phosphofurin acidic cluster sorting protein-2 (PACS-2) to acidic residues in the carboxyterminal tail of K5. Thus, CD31, a novel target of K5, is efficiently removed from ECs by a dual degradation mechanism that is regulated by the subcellular sorting of the ubiquitin ligase. K5-mediated degradation of CD31 is likely to affect EC function in KS tumors.


Subject(s)
Endothelial Cells/immunology , Endothelial Cells/virology , Herpesvirus 8, Human/enzymology , Herpesvirus 8, Human/pathogenicity , Platelet Endothelial Cell Adhesion Molecule-1/metabolism , Ubiquitin-Protein Ligases/metabolism , Adaptor Proteins, Signal Transducing/metabolism , Cell Movement , Cells, Cultured , Down-Regulation , Endothelial Cells/cytology , Humans , Proteasome Endopeptidase Complex/metabolism , Sarcoma, Kaposi/enzymology , Sarcoma, Kaposi/immunology , Sarcoma, Kaposi/virology , Substrate Specificity , Vesicular Transport Proteins , Viral Proteins/metabolism
10.
J Virol ; 78(3): 1109-20, 2004 Feb.
Article in English | MEDLINE | ID: mdl-14722266

ABSTRACT

Poxviruses and gamma-2 herpesviruses share the K3 family of viral immune evasion proteins that inhibit the surface expression of glycoproteins such as major histocompatibility complex class I (MHC-I), B7.2, ICAM-1, and CD95(Fas). K3 family proteins contain an amino-terminal PHD/LAP or RING-CH domain followed by two transmembrane domains. To examine whether human homologues are functionally related to the viral immunoevasins, we studied seven membrane-associated RING-CH (MARCH) proteins. All MARCH proteins located to subcellular membranes, and several MARCH proteins reduced surface levels of known substrates of the viral K3 family. Two closely related proteins, MARCH-IV and MARCH-IX, reduced surface expression of MHC-I molecules. In the presence of MARCH-IV or MARCH-IX, MHC-I was ubiquitinated and rapidly internalized by endocytosis, whereas MHC-I molecules lacking lysines in their cytoplasmic tail were resistant to downregulation. The amino-terminal regions containing the RING-CH domain of several MARCH proteins examined catalyzed multiubiquitin formation in vitro, suggesting that MARCH proteins are ubiquitin ligases. The functional similarity of the MARCH family and the K3 family suggests that the viral immune evasion proteins were derived from MARCH proteins, a novel family of transmembrane ubiquitin ligases that seems to target glycoproteins for lysosomal destruction via ubiquitination of the cytoplasmic tail.


Subject(s)
Down-Regulation , Histocompatibility Antigens Class I/metabolism , Immediate-Early Proteins/metabolism , Ligases/metabolism , Ubiquitin/metabolism , Viral Proteins/metabolism , Amino Acid Sequence , Animals , Cell Membrane/metabolism , HeLa Cells , Humans , Immediate-Early Proteins/genetics , Molecular Sequence Data , Sequence Homology, Amino Acid , Substrate Specificity , Transfection , Viral Proteins/genetics
11.
J Virol ; 77(2): 1427-40, 2003 Jan.
Article in English | MEDLINE | ID: mdl-12502858

ABSTRACT

The genomes of several poxviruses contain open reading frames with homology to the K3 and K5 genes of Kaposi's sarcoma-associated herpesvirus (KSHV) and the K3 gene of murine gammaherpesvirus 68, which target major histocompatibility complex class I (MHC-I) as well as costimulatory molecules for proteasomal or lysosomal degradation. The homologous gene product of myxomavirus (MV), M153R, was recently shown to reduce the cell surface expression of MHC-I. In addition, normal MHC-I surface expression was observed in cells infected with MV lacking M153R (J. L. Guerin, J. Gelfi, S. Boullier, M. Delverdier, F. A. Bellanger, S. Bertagnoli, I. Drexler, G. Sutter, and F. Messud-Petit, J. Virol. 76:2912-2923, 2002). Here, we show that M153R also downregulates the T-cell coreceptor CD4 and we study the molecular mechanism by which M153R achieves the downregulation of CD4 and MHC-I. Upon M153R expression, CD4 was rapidly internalized and degraded in lysosomes, whereas deletion of M153R from the genome of MV restored CD4 expression. The downregulation of both CD4 and MHC-I was dependent on the presence of lysine residues in their cytoplasmic tails. Increased ubiquitination of CD4 was observed upon coexpression with M153R in the presence of inhibitors of lysosomal acidification. Surface expression of CD4 was restored upon overexpression of Hrs, a ubiquitin interaction motif-containing protein that sorts ubiquitinated proteins into endosomes. Moreover, the purified PHD/LAP zinc finger of M153R catalyzed the formation of multiubiquitin adducts in vitro. Our data suggest that M153R acts as a membrane-bound ubiquitin ligase that conjugates ubiquitin to the cytoplasmic domain of substrate glycoproteins, with ubiquitin serving as a lysosomal targeting signal. Since a similar mechanism was recently proposed for KSHV K5, it seems that members of the unrelated families of gamma-2 herpesviruses and poxviruses share a common immune evasion mechanism that targets host cell immune receptors.


Subject(s)
CD4 Antigens/metabolism , Endocytosis , Ligases/metabolism , Poxviridae/enzymology , Ubiquitin/metabolism , Amino Acid Sequence , Base Sequence , Cell Line , DNA, Viral , Down-Regulation , Endosomes/metabolism , Golgi Apparatus/metabolism , Humans , Molecular Sequence Data
12.
Virus Res ; 88(1-2): 55-69, 2002 Sep.
Article in English | MEDLINE | ID: mdl-12297327

ABSTRACT

Many viruses have developed mechanisms to escape the cellular immune response by inhibiting antigen presentation from major histocompatibility complex (MHC) molecules. Most of these immune escape mechanisms are highly host adapted and specific to a given virus species or family. Recent observations however, suggest that a conserved family of viral proteins is used by both gamma-2 herpesviruses and by poxviruses to downregulate MHC class I. In addition, other cell surface molecules involved in immune recognition by T cells and NK cells are also downregulated. Two open reading frames (ORFs), K3 and K5, of Kaposi's sarcoma associated virus (KSHV) and one ORFs, K3, of murine gamma herpesvirus 68 (MHV 68) inhibit surface expression of MHC I molecules. In cells transfected with KSHV-K3 and KSHV-K5, MHC I is rapidly endocytosed and degraded in lysosomes whereas in MHV 68-K3 transfected cells, MHC I is targeted for proteasomal degradation. The K3 and K5 genes display a characteristic conserved domain structure of an amino-terminal plant homeo domain/leukemia associated protein-zinc finger domain followed by two carboxyterminal transmembrane domains. Related proteins are not only found in other gamma-2 herpesviruses, but also in several poxviruses. Moreover, recent data suggest that the K3-related protein of myxoma virus also downregulates MHC I. The presence of similar genes in eukaryotic genomes further indicates that the viral ORFs were originally derived from host genes of as yet unknown function. The molecular mechanism of MHC I downregulation by this novel gene family is only poorly understood at present. However, several lines of evidence suggest that they might function as ubiquitin ligases that regulate the intracellular transport of transmembrane proteins through ubiquitination.


Subject(s)
Gammaherpesvirinae/pathogenicity , Herpesviridae Infections/immunology , Poxviridae Infections/immunology , Poxviridae/pathogenicity , Viral Proteins/metabolism , Amino Acid Sequence , Animals , Down-Regulation , Histocompatibility Antigens Class I/metabolism , Mice , Molecular Sequence Data , Rabbits , Ubiquitin/metabolism , Viral Proteins/chemistry , Zinc Fingers
SELECTION OF CITATIONS
SEARCH DETAIL
...