Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 55
Filter
Add more filters

Publication year range
1.
Proc Natl Acad Sci U S A ; 119(5)2022 02 01.
Article in English | MEDLINE | ID: mdl-35091472

ABSTRACT

Microbes have been coevolving with their host for millions of years, exploiting host resources to their own benefit. We show that viral and bacterial pathogens convergently evolved to hijack cellular mitogen-activated protein kinase (MAPK) p90-ribosomal S6-kinases (RSKs). Theiler's virus leader (L) protein binds RSKs and prevents their dephosphorylation, thus maintaining the kinases active. Recruitment of RSKs enables L-protein-mediated inhibition of eukaryotic translation initiation factor 2 alpha kinase 2 (EIF2AK2 or PKR) and stress granule formation. Strikingly, ORF45 protein of Kaposi's sarcoma-associated herpesvirus (KSHV) and YopM protein of Yersinia use the same peptide motif as L to recruit and activate RSKs. All three proteins interact with a conserved surface-located loop of RSKs, likely acting as an allosteric regulation site. Some unrelated viruses and bacteria thus evolved to harness RSKs in a common fashion, yet to target distinct aspects of innate immunity. As documented for Varicella zoster virus ORF11, additional pathogens likely evolved to hijack RSKs, using a similar short linear motif.


Subject(s)
Host Microbial Interactions/physiology , Ribosomal Protein S6 Kinases, 90-kDa/genetics , Bacteria/pathogenicity , Bacterial Infections/genetics , Bacterial Infections/metabolism , Biological Evolution , Cell Line , Gene Expression Regulation, Viral/genetics , Host Microbial Interactions/genetics , Humans , Immediate-Early Proteins/genetics , MAP Kinase Signaling System/physiology , Mitogen-Activated Protein Kinase Kinases/metabolism , Ribosomal Protein S6 Kinases, 90-kDa/metabolism , Virus Diseases/genetics , Virus Diseases/metabolism , Virus Replication/physiology , Viruses/pathogenicity
2.
Nat Immunol ; 11(5): 449-54, 2010 May.
Article in English | MEDLINE | ID: mdl-20364150

ABSTRACT

Most antigenic peptides presented by major histocompatibility complex (MHC) class I molecules are produced by the proteasome. Here we show that a proteasome-independent peptide derived from the human tumor protein MAGE-A3 is produced directly by insulin-degrading enzyme (IDE), a cytosolic metallopeptidase. Cytotoxic T lymphocyte recognition of tumor cells was reduced after metallopeptidase inhibition or IDE silencing. Separate inhibition of the metallopeptidase and the proteasome impaired degradation of MAGE-A3 proteins, and simultaneous inhibition of both further stabilized MAGE-A3 proteins. These results suggest that MAGE-A3 proteins are degraded along two parallel pathways that involve either the proteasome or IDE and produce different sets of antigenic peptides presented by MHC class I molecules.


Subject(s)
Antigen Presentation , Antigens, Neoplasm/metabolism , Insulysin/metabolism , Neoplasm Proteins/metabolism , Peptide Fragments/metabolism , T-Lymphocytes, Cytotoxic/metabolism , Antibodies, Blocking/pharmacology , Antigen Presentation/drug effects , Antigen Presentation/genetics , Antigens, Neoplasm/immunology , Cell Fractionation , Cell Line, Tumor , Chromatography, High Pressure Liquid , Clone Cells , Cytosol , Glycopeptides/pharmacology , HLA-A1 Antigen/metabolism , Humans , Insulysin/genetics , Insulysin/immunology , Interferon-gamma/metabolism , Mass Spectrometry , Metalloendopeptidases/antagonists & inhibitors , Neoplasm Proteins/immunology , Oligopeptides/pharmacology , Peptide Fragments/immunology , Phenanthrolines/pharmacology , Proteasome Inhibitors , RNA, Small Interfering/genetics , T-Lymphocytes, Cytotoxic/immunology , T-Lymphocytes, Cytotoxic/pathology
3.
Blood ; 133(25): 2669-2681, 2019 06 20.
Article in English | MEDLINE | ID: mdl-30902807

ABSTRACT

Calreticulin (CALR) +1 frameshift mutations in exon 9 are prevalent in myeloproliferative neoplasms. Mutant CALRs possess a new C-terminal sequence rich in positively charged amino acids, leading to activation of the thrombopoietin receptor (TpoR/MPL). We show that the new sequence endows the mutant CALR with rogue chaperone activity, stabilizing a dimeric state and transporting TpoR and mutants thereof to the cell surface in states that would not pass quality control; this function is absolutely required for oncogenic transformation. Mutant CALRs determine traffic via the secretory pathway of partially immature TpoR, as they protect N117-linked glycans from further processing in the Golgi apparatus. A number of engineered or disease-associated TpoRs such as TpoR/MPL R102P, which causes congenital thrombocytopenia, are rescued for traffic and function by mutant CALRs, which can also overcome endoplasmic reticulum retention signals on TpoR. In addition to requiring N-glycosylation of TpoR, mutant CALRs require a hydrophobic patch located in the extracellular domain of TpoR to induce TpoR thermal stability and initial intracellular activation, whereas full activation requires cell surface localization of TpoR. Thus, mutant CALRs are rogue chaperones for TpoR and traffic-defective TpoR mutants, a function required for the oncogenic effects.


Subject(s)
Calreticulin/genetics , Calreticulin/metabolism , Hematologic Neoplasms/genetics , Hematologic Neoplasms/metabolism , Receptors, Thrombopoietin/metabolism , Animals , Humans , Mice , Molecular Chaperones/genetics , Molecular Chaperones/metabolism , Mutation , Protein Transport/physiology
4.
Proc Natl Acad Sci U S A ; 114(47): E10161-E10168, 2017 11 21.
Article in English | MEDLINE | ID: mdl-29109269

ABSTRACT

Human regulatory T cells (Tregs) suppress other T cells by converting the latent, inactive form of TGF-ß1 into active TGF-ß1. In Tregs, TGF-ß1 activation requires GARP, a transmembrane protein that binds and presents latent TGF-ß1 on the surface of Tregs stimulated through their T cell receptor. However, GARP is not sufficient because transduction of GARP in non-Treg T cells does not induce active TGF-ß1 production. RGD-binding integrins were shown to activate TGF-ß1 in several non-T cell types. Here we show that αVß8 dimers are present on stimulated human Tregs but not in other T cells, and that antibodies against αV or ß8 subunits block TGF-ß1 activation in vitro. We also show that αV and ß8 interact with GARP/latent TGF-ß1 complexes in human Tregs. Finally, a blocking antibody against ß8 inhibited immunosuppression by human Tregs in a model of xenogeneic graft-vs.-host disease induced by the transfer of human T cells in immunodeficient mice. These results show that TGF-ß1 activation on the surface of human Tregs implies an interaction between the integrin αVß8 and GARP/latent TGF-ß1 complexes. Immunosuppression by human Tregs can be inhibited by antibodies against GARP or against the integrin ß8 subunit. Such antibodies may prove beneficial against cancer or chronic infections.


Subject(s)
Graft vs Host Disease/immunology , Immune Tolerance/drug effects , Integrins/immunology , T-Lymphocytes, Regulatory/immunology , Transforming Growth Factor beta1/immunology , Animals , Antibodies, Monoclonal/pharmacology , Antibodies, Monoclonal/therapeutic use , Cells, Cultured , Disease Models, Animal , Humans , Integrins/antagonists & inhibitors , Membrane Proteins/immunology , Membrane Proteins/metabolism , Mice , Mice, SCID , Neoplasms/immunology , Neoplasms/therapy , T-Lymphocytes, Regulatory/transplantation , Transforming Growth Factor beta1/metabolism , Transplantation, Heterologous
5.
J Allergy Clin Immunol ; 144(1): 224-235, 2019 07.
Article in English | MEDLINE | ID: mdl-30707971

ABSTRACT

BACKGROUND: Janus kinase (JAK) 2 plays pivotal roles in signaling by several cytokine receptors. The mutant JAK2 V617F is the most common molecular event associated with myeloproliferative neoplasms. Selective targeting of the mutant would be ideal for treating these pathologies by sparing essential JAK2 functions. OBJECTIVE: We characterize inhibitory strategies for JAK2 V617F and assess their effect on physiologic signaling by distinct cytokine receptors. METHODS: Through structure-guided mutagenesis, we assessed the role of key residues around F617 and used a combination of cellular and biochemical assays to measure the activity of JAKs in reconstituted cells. We also assessed the effect of several specific JAK2 V617F inhibitory mutations on receptor dimerization using the NanoBiT protein complementation approach. RESULTS: We identified a novel Janus kinase homology 2 (JH2) αC mutation, A598F, which is suggested to inhibit the aromatic stacking between F617 with F594 and F595. Like other JAK2 V617F inhibitory mutations, A598F decreased oncogenic activation and spared cytokine activation while preventing JAK2 V617F-promoted erythropoietin receptor dimerization. Surprisingly, A598F and other V617F-inhibiting mutations (F595A, E596R, and F537A) significantly impaired IFN-γ signaling. This was specific for IFN-γ because the inhibitory mutations preserved responses to ligands of a series of receptor complexes. Similarly, homologous mutations in JAK1 prevented signaling by IFN-γ. CONCLUSIONS: The JH2 αC region, which is required for JAK2 V617F hyperactivation, is crucial for relaying cytokine-induced signaling of the IFN-γ receptor. We discuss how strategies aiming to inhibit JAK2 V617F could be used for identifying inhibitors of IFN-γ signaling.


Subject(s)
Janus Kinase 2/genetics , Receptors, Cytokine/metabolism , Animals , Cell Line , Humans , Janus Kinase 2/metabolism , Mice , Mutation , Signal Transduction
6.
Immunity ; 28(3): 414-24, 2008 Mar.
Article in English | MEDLINE | ID: mdl-18342010

ABSTRACT

For several days after antigenic stimulation, human cytolytic T lymphocyte (CTL) clones exhibit a decrease in their effector activity and in their binding to human leukocyte antigen (HLA)-peptide tetramers. We observed that, when in this state, CTLs lose the colocalization of the T cell receptor (TCR) and CD8. Effector function and TCR-CD8 colocalization were restored with galectin disaccharide ligands, suggesting that the binding of TCR to galectin plays a role in the distancing of TCR from CD8. These findings appear to be applicable in vivo, as TCR was observed to be distant from CD8 on human tumor-infiltrating lymphocytes, which were anergic. These lymphocytes recovered effector functions and TCR-CD8 colocalization after ex vivo treatment with galectin disaccharide ligands. The separation of TCR and CD8 molecules could be one major mechanism of anergy in tumors and other chronic stimulation conditions.


Subject(s)
CD8 Antigens/metabolism , Clonal Anergy/immunology , Lymphocytes, Tumor-Infiltrating/metabolism , Receptors, Antigen, T-Cell/metabolism , T-Lymphocytes, Cytotoxic/metabolism , CD8 Antigens/immunology , Cell Line, Tumor , Flow Cytometry , Galectins/metabolism , HLA Antigens/immunology , Humans , Lymphocyte Activation/immunology , Lymphocytes, Tumor-Infiltrating/immunology , Microscopy, Confocal , Microscopy, Electron, Scanning , Peptides/immunology , Receptors, Antigen, T-Cell/immunology , T-Lymphocytes, Cytotoxic/immunology
7.
Biochem J ; 473(11): 1579-91, 2016 06 01.
Article in English | MEDLINE | ID: mdl-27029346

ABSTRACT

The mechanisms by which JAK2 is activated by the prevalent pseudokinase (JH2) V617F mutation in blood cancers remain elusive. Via structure-guided mutagenesis and transcriptional and functional assays, we identify a community of residues from the JH2 helix αC, SH2-JH2 linker and JH1 kinase domain that mediate V617F-induced activation. This circuit is broken by altering the charge of residues along the solvent-exposed face of the JH2 αC, which is predicted to interact with the SH2-JH2 linker and JH1. Mutations that remove negative charges or add positive charges, such as E596A/R, do not alter the JH2 V617F fold, as shown by the crystal structure of JH2 V617F E596A. Instead, they prevent kinase domain activation via modulation of the C-terminal residues of the SH2-JH2 linker. These results suggest strategies for selective V617F JAK2 inhibition, with preservation of wild-type function.


Subject(s)
Cytokines/pharmacology , Janus Kinase 2/chemistry , Janus Kinase 2/metabolism , Blotting, Western , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Proliferation/genetics , Flow Cytometry , Humans , Janus Kinase 2/genetics , Mutation/genetics , Phosphorylation/drug effects , Protein Domains/genetics , Protein Domains/physiology
8.
Immunology ; 149(2): 172-85, 2016 10.
Article in English | MEDLINE | ID: mdl-27317384

ABSTRACT

Immunity against Theileria parva is associated with CD8 T-cell responses that exhibit immunodominance, focusing the response against limited numbers of epitopes. As candidates for inclusion in vaccines, characterization of responses against immunodominant epitopes is a key component in novel vaccine development. We have previously demonstrated that the Tp249-59 and Tp1214-224 epitopes dominate CD8 T-cell responses in BoLA-A10 and BoLA-18 MHC I homozygous animals, respectively. In this study, peptide-MHC I tetramers for these epitopes, and a subdominant BoLA-A10-restricted epitope (Tp298-106 ), were generated to facilitate accurate and rapid enumeration of epitope-specific CD8 T cells. During validation of these tetramers a substantial proportion of Tp249-59 -reactive T cells failed to bind the tetramer, suggesting that this population was heterogeneous with respect to the recognized epitope. We demonstrate that Tp250-59 represents a distinct epitope and that tetramers produced with Tp50-59 and Tp49-59 show no cross-reactivity. The Tp249-59 and Tp250-59 epitopes use different serine residues as the N-terminal anchor for binding to the presenting MHC I molecule. Molecular dynamic modelling predicts that the two peptide-MHC I complexes adopt structurally different conformations and Tcell receptor ß sequence analysis showed that Tp249-59 and Tp250-59 are recognized by non-overlapping T-cell receptor repertoires. Together these data demonstrate that although differing by only a single residue, Tp249-59 and Tp250-59 epitopes form distinct ligands for T-cell receptor recognition. Tetramer analysis of T. parva-specific CD8 T-cell lines confirmed the immunodominance of Tp1214-224 in BoLA-A18 animals and showed in BoLA-A10 animals that the Tp249-59 epitope response was generally more dominant than the Tp250-59 response and confirmed that the Tp298-106 response was subdominant.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Protozoan Vaccines/immunology , T-Lymphocyte Subsets/immunology , Theileria parva/immunology , Theileriasis/immunology , Animals , Antigens, Protozoan/metabolism , Cattle , Cell Line , Computer Simulation , Epitope Mapping , Epitopes, T-Lymphocyte/metabolism , Histocompatibility Antigens Class I/metabolism , Immunodominant Epitopes/metabolism , Lymphocyte Activation , Peptide Fragments/metabolism , Protein Binding
9.
Cancer Immunol Immunother ; 65(10): 1177-88, 2016 10.
Article in English | MEDLINE | ID: mdl-27514672

ABSTRACT

Nectin-4 is a tumor antigen present on the surface of breast, ovarian and lung carcinoma cells. It is rarely present in normal adult tissues and is therefore a candidate target for cancer immunotherapy. Here, we identified a Nectin-4 antigenic peptide that is naturally presented to T cells by HLA-A2 molecules. We first screened the 502 nonamer peptides of Nectin-4 (510 amino acids) for binding to and off-rate from eight different HLA class I molecules. We then combined biochemical, cellular and algorithmic assays to select 5 Nectin-4 peptides that bound to HLA-A*02:01 molecules. Cytolytic T lymphocytes were obtained from healthy donors, that specifically lyzed HLA-A2(+) cells pulsed with 2 out of the 5 peptides, indicating the presence of anti-Nectin-4 CD8(+) T lymphocytes in the human T cell repertoire. Finally, an HLA-A2-restricted cytolytic T cell clone derived from a breast cancer patient recognized peptide Nectin-4145-153 (VLVPPLPSL) and lyzed HLA-A2(+) Nectin-4(+) breast carcinoma cells. These results indicate that peptide Nectin-4145-153 is naturally processed for recognition by T cells on HLA-A2 molecules. It could be used to monitor antitumor T cell responses or to immunize breast cancer patients.


Subject(s)
Adenocarcinoma/immunology , Breast Neoplasms/immunology , Cancer Vaccines/immunology , Cell Adhesion Molecules/metabolism , Epitopes, T-Lymphocyte/metabolism , Immunoglobulins/metabolism , Peptides/metabolism , T-Lymphocytes, Cytotoxic/immunology , Antigens, Neoplasm/immunology , Cell Line, Tumor , Clone Cells , Epitope Mapping , Female , HLA-A2 Antigen/metabolism , Humans , Protein Binding
10.
Proc Natl Acad Sci U S A ; 109(7): 2497-502, 2012 Feb 14.
Article in English | MEDLINE | ID: mdl-22308364

ABSTRACT

Tryptophan catabolism mediated by indoleamine 2,3-dioxygenase (IDO1) is an important mechanism of peripheral immune tolerance contributing to tumoral immune resistance, and IDO1 inhibition is an active area of drug development. Tryptophan 2,3-dioxygenase (TDO) is an unrelated hepatic enzyme that also degrades tryptophan along the kynurenine pathway. Here, we show that enzymatically active TDO is expressed in a significant proportion of human tumors. In a preclinical model, TDO expression by tumors prevented their rejection by immunized mice. We developed a TDO inhibitor, which, upon systemic treatment, restored the ability of mice to reject TDO-expressing tumors. Our results describe a mechanism of tumoral immune resistance based on TDO expression and establish proof-of-concept for the use of TDO inhibitors in cancer therapy.


Subject(s)
Neoplasms/immunology , Tryptophan Oxygenase/antagonists & inhibitors , Animals , Cell Line, Tumor , Humans , Mice , Tryptophan Oxygenase/metabolism
11.
Int J Cancer ; 134(12): 2841-52, 2014 Jun 15.
Article in English | MEDLINE | ID: mdl-24249003

ABSTRACT

There is increasing evidence that the effect of chemotherapy on tumor growth is not cell autonomous but relies on the immune system. The objective of this study was therefore to decipher the cellular and molecular mechanisms underlying the role of innate and adaptive immunity in chemotherapy-induced tumor rejection. Treatment of DBA/2 mice bearing P815 mastocytoma with cyclophosphamide induced rejection and long-term protection in a CD4- and CD8-dependent manner. A population of inflammatory-type dendritic cells was dramatically expanded in the lymph nodes of mice that rejected the tumor and correlated with CD4-dependent infiltration, in tumor bed, of tumor-specific CD8+ T lymphocytes. Our data point to a major role of CD4+ T cells in inducing chemokine expression in the tumor, provoking migration of tumor-specific CXCR3+ CD8+ T lymphocytes. Importantly, the analysis of CD8+ T cells specific to P1A/H-2L(d) and P1E/H-2K(d) revealed that cyclophosphamide altered the P815-specific CD8 T repertoire by amplifying the response specific to the mutated P1E antigen.


Subject(s)
Antineoplastic Agents, Alkylating/pharmacology , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Cyclophosphamide/therapeutic use , Mastocytoma/drug therapy , Animals , Cell Line, Tumor , Cell Movement/immunology , Cell Proliferation , Dendritic Cells/immunology , H-2 Antigens/immunology , Integrin beta3/immunology , Lymph Nodes/cytology , Lymphocyte Activation/immunology , Mastocytoma/immunology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Inbred DBA , Receptors, CXCR3/metabolism
12.
Mol Vis ; 20: 956-69, 2014.
Article in English | MEDLINE | ID: mdl-24991188

ABSTRACT

PURPOSE: Vogt-Koyanagi-Harada (VKH) syndrome is an autoimmune disease characterized by inaugural uveomeningitidis and hearing loss and at late stages a depigmentation in eyes and skin. Melanocytes are the cells common to the four affected tissues, namely eye, brain, inner ear, and skin. Melanocytes are therefore considered as the source of self-antigens. The melanocytic proteins tyrosinase-related protein-1 (TRP1), TRP2, tyrosinase, and gp100 have been proposed as the proteins targeted by autoreactive T cells from VKH patients bearing human leukocyte antigen (HLA)-DRB1*04:05, the HLA allele classically associated with VKH disease. The objective of this work was to determine the antigens recognized by a large number of potentially autoreactive CD4 T lymphocytes obtained from the cerebrospinal fluid of one VKH patient who did not express HLA-DRB1*04:05. METHODS: T cells were isolated from the cerebrospinal fluid of a newly diagnosed HLA-DRB1*14:01,*15:03;-DPB1*01:01,*04:02 patient in the acute phase of the VKH disease and cloned by limiting dilution. Each of the 107 T cell clones, of which 90% were CD4(+), was tested for its ability to secrete cytokines upon contact with autologous antigen-presenting cells loaded with either of the melanocytic proteins TRP1, TRP2, tyrosinase, gp100, Melan-A and KU-MEL-1. The sensitivity of our recombinant bacteria-based approach was validated with a CD4 T cell clone with known antigen specificity. The ability of each of the 107 clones to secrete cytokines upon nonspecific stimulation was verified. RESULTS: None of the 107 T cell clones was able to secrete tumor necrosis factor-α, interferon-γ, interleukin (IL)-5, or IL-17 upon contact with autologous B cells loaded with any of the six common melanocytic proteins. Nine clones secreted high-level IL-17 upon stimulation with beads coated with antibodies. CONCLUSIONS: The self-antigens that triggered the VKH disease in this patient probably derive from proteins other than the six melanocytic proteins mentioned above. Further study of antigens that are recognized by potential autoreactive T cells from VKH patients is likely to benefit from testing a broader set of melanocytic proteins.


Subject(s)
Autoantigens/immunology , Melanocytes/immunology , T-Lymphocytes/immunology , Uveomeningoencephalitic Syndrome/cerebrospinal fluid , Uveomeningoencephalitic Syndrome/immunology , Adult , Antigens, Neoplasm/metabolism , B-Lymphocytes/virology , Bacteria/metabolism , Blotting, Western , CD4-Positive T-Lymphocytes/immunology , Cell Separation , Clone Cells , Cytokines/metabolism , Epitopes/immunology , Herpesvirus 4, Human/immunology , Humans , Interleukin-17/metabolism , Male , Neoplasm Proteins/metabolism , Recombinant Proteins/metabolism , Uveomeningoencephalitic Syndrome/pathology
13.
J Immunol ; 189(7): 3538-47, 2012 Oct 01.
Article in English | MEDLINE | ID: mdl-22925930

ABSTRACT

We recently described two proteasome subtypes that are intermediate between the standard proteasome and the immunoproteasome. They contain only one (ß5i) or two (ß1i and ß5i) of the three inducible catalytic subunits of the immunoproteasome. They are present in tumor cells and abundant in normal human tissues. We described two tumor antigenic peptides that are uniquely produced by these intermediate proteasomes. In this work, we studied the production by intermediate proteasomes of tumor antigenic peptides known to be produced exclusively by the immunoproteasome (MAGE-A3(114-122), MAGE-C2(42-50), MAGE-C2(336-344)) or the standard proteasome (Melan-A(26-35), tyrosinase(369-377), gp100(209-217)). We observed that intermediate proteasomes efficiently produced the former peptides, but not the latter. Two peptides from the first group were equally produced by both intermediate proteasomes, whereas MAGE-C2(336-344) was only produced by intermediate proteasome ß1i-ß5i. Those results explain the recognition of tumor cells devoid of immunoproteasome by CTL recognizing peptides not produced by the standard proteasome. We also describe a third antigenic peptide that is produced exclusively by an intermediate proteasome: peptide MAGE-C2(191-200) is produced only by intermediate proteasome ß1i-ß5i. Analyzing in vitro digests, we observed that the lack of production by a given proteasome usually results from destruction of the antigenic peptide by internal cleavage. Interestingly, we observed that the immunoproteasome and the intermediate proteasomes fail to cleave between hydrophobic residues, despite a higher chymotrypsin-like activity measured on fluorogenic substrates. Altogether, our results indicate that the repertoire of peptides produced by intermediate proteasomes largely matches the repertoire produced by the immunoproteasome, but also contains additional peptides.


Subject(s)
Antigens, Neoplasm/metabolism , MART-1 Antigen/metabolism , Monophenol Monooxygenase/metabolism , Neoplasm Proteins/metabolism , Peptide Fragments/metabolism , Proteasome Endopeptidase Complex/metabolism , Protein Processing, Post-Translational/immunology , gp100 Melanoma Antigen/metabolism , Amino Acid Sequence , Antigen Presentation/immunology , Antigens, Neoplasm/biosynthesis , Antigens, Neoplasm/genetics , Cell Line, Tumor , Clone Cells , Epitopes, T-Lymphocyte/biosynthesis , Epitopes, T-Lymphocyte/metabolism , HEK293 Cells , Humans , Hydrophobic and Hydrophilic Interactions , MART-1 Antigen/biosynthesis , Melanoma/enzymology , Melanoma/genetics , Melanoma/immunology , Monophenol Monooxygenase/biosynthesis , Neoplasm Proteins/biosynthesis , Peptide Fragments/biosynthesis , Proteasome Endopeptidase Complex/genetics , gp100 Melanoma Antigen/biosynthesis
14.
J Immunol ; 188(1): 111-21, 2012 Jan 01.
Article in English | MEDLINE | ID: mdl-22140254

ABSTRACT

Central tolerance toward tissue-restricted Ags is considered to rely on ectopic expression in the thymus, which was also observed for tumor Ags encoded by cancer-germline genes. It is unknown whether endogenous expression shapes the T cell repertoire against the latter Ags and explains their weak immunogenicity. We addressed this question using mouse cancer-germline gene P1A, which encodes antigenic peptide P1A(35-43) presented by H-2L(d). We made P1A-knockout (P1A-KO) mice and asked whether their anti-P1A(35-43) immune responses were stronger than those of wild-type mice and whether P1A-KO mice responded to other P1A epitopes, against which wild-type mice were tolerized. We observed that both types of mice mounted similar P1A(35-43)-specific CD8 T cell responses, although the frequency of P1A(35-43)-specific CD8 T cells generated in response to P1A-expressing tumors was slightly higher in P1A-KO mice. This higher reactivity allowed naive P1A-KO mice to reject spontaneously P1A-expressing tumors, which progressed in wild-type mice. TCR-Vß usage of P1A(35-43)-specific CD8 cells was slightly modified in P1A-KO mice. Peptide P1A(35-43) remained the only P1A epitope recognized by CD8 T cells in both types of mice, which also displayed similar thymic selection of a transgenic TCR recognizing P1A(35-43). These results indicate the existence of a minimal tolerance to an Ag encoded by a cancer-germline gene and suggest that its endogenous expression only slightly affects diversification of the T cell repertoire against this Ag.


Subject(s)
Antigens, Neoplasm/immunology , CD8-Positive T-Lymphocytes/immunology , Epitopes/immunology , Immune Tolerance , Neoplasms/immunology , Peptides/immunology , Animals , Antigens, Neoplasm/genetics , Cell Line, Tumor , Epitopes/genetics , Mice , Mice, Knockout , Neoplasms/genetics , Peptides/genetics , Receptors, Antigen, T-Cell, alpha-beta/genetics , Receptors, Antigen, T-Cell, alpha-beta/immunology
15.
Proc Natl Acad Sci U S A ; 107(43): 18599-604, 2010 Oct 26.
Article in English | MEDLINE | ID: mdl-20937868

ABSTRACT

Most antigenic peptides presented by MHC class I molecules result from the degradation of intracellular proteins by the proteasome. In lymphoid tissues and cells exposed to IFNγ, the standard proteasome is replaced by the immunoproteasome, in which all of the standard catalytic subunits ß1, ß2, and ß5 are replaced by their inducible counterparts ß1i, ß2i, and ß5i, which have different cleavage specificities. The immunoproteasome thereby shapes the repertoire of antigenic peptides. The existence of additional forms of proteasomes bearing a mixed assortment of standard and inducible catalytic subunits has been suggested. Using a new set of unique subunit-specific antibodies, we have now isolated, quantified, and characterized human proteasomes that are intermediate between the standard proteasome and the immunoproteasome. They contain only one (ß5i) or two (ß1i and ß5i) of the three inducible catalytic subunits of the immunoproteasome. These intermediate proteasomes represent between one-third and one-half of the proteasome content of human liver, colon, small intestine, and kidney. They are also present in human tumor cells and dendritic cells. We identified two tumor antigens of clinical interest that are processed exclusively either by intermediate proteasomes ß5i (MAGE-A3(271-279)) or by intermediate proteasomes ß1i-ß5i (MAGE-A10(254-262)). The existence of these intermediate proteasomes broadens the repertoire of antigens presented to CD8 T cells and implies that the antigens presented by a given cell depend on their proteasome content.


Subject(s)
Antigen Presentation , Histocompatibility Antigens Class I/metabolism , Proteasome Endopeptidase Complex/classification , Proteasome Endopeptidase Complex/metabolism , Amino Acid Sequence , Animals , Antigens, Neoplasm/genetics , Antigens, Neoplasm/metabolism , Cell Line, Tumor , Humans , Mice , Mice, Knockout , Molecular Sequence Data , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Proteasome Endopeptidase Complex/chemistry , Proteasome Endopeptidase Complex/genetics , Protein Subunits , Recombinant Proteins/genetics , Recombinant Proteins/immunology , Recombinant Proteins/metabolism , Sequence Homology, Amino Acid
16.
Methods Mol Biol ; 2548: 169-178, 2022.
Article in English | MEDLINE | ID: mdl-36151498

ABSTRACT

The envelope of Gram-negative bacteria is an essential compartment which is in direct contact with the environment; the envelope maintains cellular integrity and functions as a permeability barrier protecting the cell from toxic compounds. The outer layer of the envelope is an asymmetric membrane whose external leaflet is mainly composed of lipopolysaccharide molecules. Recently, there has been growing evidence that lipoproteins (i.e., soluble proteins anchored to a membrane by a lipid moiety) decorate the lipopolysaccharide leaflet in the model bacterium Escherichia coli, challenging the current paradigm that lipoproteins remain in the periplasm in this organism. However, assessing the surface exposure of lipoproteins is challenging. Here, we describe an optimized and reproducible dotblot protocol to assess the presence of lipoproteins at the surface of E. coli and other bacterial models. We added all necessary controls to reduce the possibility of artifacts giving rise to false-positive results. We selected the stress sensor RcsF as a model lipoprotein to illustrate the method, which can be used for any other lipoprotein.


Subject(s)
Escherichia coli Proteins , Escherichia coli , Bacterial Outer Membrane Proteins/metabolism , Cell Membrane/metabolism , Escherichia coli/metabolism , Escherichia coli Proteins/metabolism , Lipopolysaccharides/metabolism , Lipoproteins/metabolism
17.
Blood ; 113(8): 1651-60, 2009 Feb 19.
Article in English | MEDLINE | ID: mdl-19074732

ABSTRACT

Dendritic cell (DC) targeting in vivo has recently been shown to confer strong and protective cytotoxic T lymphocyte (CTL)-based immunity in tumor murine models. Our group has recently demonstrated in preclinical models that the infusion of genetically modified lymphocytes (GMLs) expressing the self/tumor antigen TRP-2 is able to elicit functional TRP-2-specific effectors with antitumor activity by targeting DCs in vivo. Here we have analyzed vaccine- and tumor-specific immune responses of 10 melanoma patients treated with autologous GMLs expressing the cancer germline gene MAGE-A3. Three of 10 patients treated with MAGE-A3-GML showed an increase of circulating anti-MAGE-A3 T cells, and developed skin delayed-type hypersensitivity to MAGE-A3. Interestingly, in 2 of these patients, with progressive and measurable tumors at study entry, anti-MAGE-A3 T cells were detected not only in the blood but also within tumors resected after vaccination. These results demonstrate that the infusion of MAGE-A3-GML elicits antitumor T cells, which are capable of trafficking to inflamed tissues and of infiltrating tumors. Clinical studies on a larger group of patients are needed to evaluate the clinical efficacy of such a strategy.


Subject(s)
Antigens, Neoplasm/genetics , Antigens, Neoplasm/immunology , Cancer Vaccines/administration & dosage , Genetic Therapy/methods , Melanoma/therapy , Neoplasm Proteins/genetics , Neoplasm Proteins/immunology , Skin Neoplasms/therapy , Adoptive Transfer , Animals , COS Cells , Cancer Vaccines/adverse effects , Cell Line, Tumor , Chlorocebus aethiops , Humans , Hypersensitivity, Delayed/immunology , Melanoma/immunology , Melanoma/pathology , Neoplasm Staging , Pilot Projects , Skin Neoplasms/immunology , Skin Neoplasms/pathology , T-Lymphocytes/cytology , T-Lymphocytes/physiology , T-Lymphocytes/transplantation , Thymidine Kinase/genetics , Thymidine Kinase/immunology , Transfection
18.
Bioorg Med Chem ; 19(4): 1550-61, 2011 Feb 15.
Article in English | MEDLINE | ID: mdl-21269836

ABSTRACT

Indoleamine 2,3-dioxygenase (IDO) is a heme dioxygenase which has been shown to be involved in the pathological immune escape of diseases such as cancer. The synthesis and structure-activity relationships (SAR) of a novel series of IDO inhibitors based on the indol-2-yl ethanone scaffold is described. In vitro and in vivo biological activities have been evaluated, leading to compounds with IC(50) values in the micromolar range in both tests. Introduction of small substituents in the 5- and 6-positions of the indole ring, indole N-methylation and variations of the aromatic side chain are all well tolerated. An iron coordinating group on the linker is a prerequisite for biological activity, thus corroborating the virtual screening results.


Subject(s)
Ethane/chemistry , Indoleamine-Pyrrole 2,3,-Dioxygenase/antagonists & inhibitors , Indoles/chemistry , Indoles/pharmacology , Catalytic Domain , Cell Line , Humans , Indoleamine-Pyrrole 2,3,-Dioxygenase/chemistry , Models, Molecular , Protein Interaction Domains and Motifs , Structure-Activity Relationship
19.
Nat Med ; 9(10): 1269-74, 2003 Oct.
Article in English | MEDLINE | ID: mdl-14502282

ABSTRACT

T lymphocytes undergo proliferation arrest when exposed to tryptophan shortage, which can be provoked by indoleamine 2,3-dioxygenase (IDO), an enzyme that is expressed in placenta and catalyzes tryptophan degradation. Here we show that most human tumors constitutively express IDO. We also observed that expression of IDO by immunogenic mouse tumor cells prevents their rejection by preimmunized mice. This effect is accompanied by a lack of accumulation of specific T cells at the tumor site and can be partly reverted by systemic treatment of mice with an inhibitor of IDO, in the absence of noticeable toxicity. These results suggest that the efficacy of therapeutic vaccination of cancer patients might be improved by concomitant administration of an IDO inhibitor.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Neoplasms/metabolism , Tryptophan Oxygenase/metabolism , Tryptophan/analogs & derivatives , Tryptophan/metabolism , Tumor Escape , Animals , Cell Line, Tumor , Female , Humans , Indoleamine-Pyrrole 2,3,-Dioxygenase , Mice , Neoplasm Transplantation , Neoplasms/immunology , Neoplasms/pathology , Placenta/enzymology , Pregnancy , RNA, Messenger/metabolism , Tryptophan/pharmacology , Tryptophan Oxygenase/antagonists & inhibitors , Tryptophan Oxygenase/genetics
20.
Eur J Immunol ; 39(12): 3315-22, 2009 Dec.
Article in English | MEDLINE | ID: mdl-19750484

ABSTRACT

Human Treg and Th clones secrete the latent form of TGF-beta, in which the mature TGF-beta protein is bound to the latency-associated peptide (LAP), and is thereby prevented from binding to the TGF-beta receptor. We previously showed that upon TCR stimulation, human Treg clones but not Th clones produce active TGF-beta and bear LAP on their surface. Here, we show that latent TGF-beta, i.e. both LAP and mature TGF-beta, binds to glycoprotein A repetitions predominant (GARP), a transmembrane protein containing leucine rich repeats, which is present on the surface of stimulated Treg clones but not on Th clones. Membrane localization of latent TGF-beta mediated by binding to GARP may be necessary for the ability of Treg to activate TGF-beta upon TCR stimulation. However, it is not sufficient as lentiviral-mediated expression of GARP in human Th cells induces binding of latent TGF-beta to the cell surface, but does not result in the production of active TGF-beta upon stimulation of these Th cells.


Subject(s)
Cell Membrane/metabolism , Membrane Proteins/metabolism , T-Lymphocytes, Regulatory/metabolism , Transforming Growth Factor beta/metabolism , Blotting, Western , CD4 Antigens/metabolism , Cells, Cultured , Flow Cytometry , Humans , Interleukin-2 Receptor alpha Subunit/metabolism , Interleukin-7 Receptor alpha Subunit/metabolism , Jurkat Cells , Latent TGF-beta Binding Proteins/genetics , Latent TGF-beta Binding Proteins/metabolism , Lymphocyte Activation/immunology , Membrane Proteins/genetics , Models, Biological , Muromonab-CD3/immunology , Protein Binding , Reverse Transcriptase Polymerase Chain Reaction , T-Lymphocytes/cytology , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , T-Lymphocytes, Regulatory/cytology , T-Lymphocytes, Regulatory/immunology , Transforming Growth Factor beta/genetics
SELECTION OF CITATIONS
SEARCH DETAIL