Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 796
Filter
1.
Immunity ; 54(3): 586-602.e8, 2021 03 09.
Article in English | MEDLINE | ID: mdl-33691136

ABSTRACT

To identify disease-relevant T cell receptors (TCRs) with shared antigen specificity, we analyzed 778,938 TCRß chain sequences from 178 non-small cell lung cancer patients using the GLIPH2 (grouping of lymphocyte interactions with paratope hotspots 2) algorithm. We identified over 66,000 shared specificity groups, of which 435 were clonally expanded and enriched in tumors compared to adjacent lung. The antigenic epitopes of one such tumor-enriched specificity group were identified using a yeast peptide-HLA A∗02:01 display library. These included a peptide from the epithelial protein TMEM161A, which is overexpressed in tumors and cross-reactive epitopes from Epstein-Barr virus and E. coli. Our findings suggest that this cross-reactivity may underlie the presence of virus-specific T cells in tumor infiltrates and that pathogen cross-reactivity may be a feature of multiple cancers. The approach and analytical pipelines generated in this work, as well as the specificity groups defined here, present a resource for understanding the T cell response in cancer.


Subject(s)
Carcinoma, Non-Small-Cell Lung/immunology , Epitope Mapping/methods , Epitopes, T-Lymphocyte/genetics , Lung Neoplasms/immunology , Receptors, Antigen, T-Cell, alpha-beta/genetics , T-Lymphocytes/immunology , Algorithms , Antigen Presentation , Antigens, Neoplasm/metabolism , Cells, Cultured , Cross Reactions , Epitopes, T-Lymphocyte/metabolism , HLA-A2 Antigen/metabolism , Humans , Protein Binding , T-Cell Antigen Receptor Specificity
2.
Mol Cell ; 73(5): 1015-1027.e7, 2019 03 07.
Article in English | MEDLINE | ID: mdl-30711376

ABSTRACT

TCRs recognize cognate pMHCs to initiate T cell signaling and adaptive immunity. Mechanical force strengthens TCR-pMHC interactions to elicit agonist-specific catch bonds to trigger TCR signaling, but the underlying dynamic structural mechanism is unclear. We combined steered molecular dynamics (SMD) simulation, single-molecule biophysical approaches, and functional assays to collectively demonstrate that mechanical force induces conformational changes in pMHCs to enhance pre-existing contacts and activates new interactions at the TCR-pMHC binding interface to resist bond dissociation under force, resulting in TCR-pMHC catch bonds and T cell activation. Intriguingly, cancer-associated somatic mutations in HLA-A2 that may restrict these conformational changes suppressed TCR-pMHC catch bonds. Structural analysis also indicated that HLA polymorphism might alter the equilibrium of these conformational changes. Our findings not only reveal critical roles of force-induced conformational changes in pMHCs for activating TCR-pMHC catch bonds but also have implications for T cell-based immunotherapy.


Subject(s)
Adaptive Immunity , HLA-A2 Antigen/immunology , Mechanotransduction, Cellular , Receptors, Antigen, T-Cell/immunology , T-Lymphocytes/immunology , Animals , HEK293 Cells , HLA-A2 Antigen/chemistry , HLA-A2 Antigen/genetics , HLA-A2 Antigen/metabolism , Humans , Hybridomas , Mice, Inbred C57BL , Mice, Transgenic , Molecular Dynamics Simulation , Mutation , Protein Binding , Protein Conformation , Receptors, Antigen, T-Cell/chemistry , Receptors, Antigen, T-Cell/genetics , Receptors, Antigen, T-Cell/metabolism , Single Molecule Imaging/methods , Structure-Activity Relationship , T-Lymphocytes/metabolism
3.
Brief Bioinform ; 25(1)2023 11 22.
Article in English | MEDLINE | ID: mdl-38233090

ABSTRACT

Immunologic recognition of peptide antigens bound to class I major histocompatibility complex (MHC) molecules is essential to both novel immunotherapeutic development and human health at large. Current methods for predicting antigen peptide immunogenicity rely primarily on simple sequence representations, which allow for some understanding of immunogenic features but provide inadequate consideration of the full scale of molecular mechanisms tied to peptide recognition. We here characterize contributions that unsupervised and supervised artificial intelligence (AI) methods can make toward understanding and predicting MHC(HLA-A2)-peptide complex immunogenicity when applied to large ensembles of molecular dynamics simulations. We first show that an unsupervised AI method allows us to identify subtle features that drive immunogenicity differences between a cancer neoantigen and its wild-type peptide counterpart. Next, we demonstrate that a supervised AI method for class I MHC(HLA-A2)-peptide complex classification significantly outperforms a sequence model on small datasets corrected for trivial sequence correlations. Furthermore, we show that both unsupervised and supervised approaches reveal determinants of immunogenicity based on time-dependent molecular fluctuations and anchor position dynamics outside the MHC binding groove. We discuss implications of these structural and dynamic immunogenicity correlates for the induction of T cell responses and therapeutic T cell receptor design.


Subject(s)
HLA-A2 Antigen , Molecular Dynamics Simulation , Humans , HLA-A2 Antigen/metabolism , Artificial Intelligence , Peptides/chemistry , Histocompatibility Antigens Class I/metabolism , Protein Binding
4.
J Chem Inf Model ; 64(13): 5016-5027, 2024 Jul 08.
Article in English | MEDLINE | ID: mdl-38920330

ABSTRACT

The intricate interaction between major histocompatibility complexes (MHCs) and antigen peptides with diverse amino acid sequences plays a pivotal role in immune responses and T cell activity. In recent years, deep learning (DL)-based models have emerged as promising tools for accelerating antigen peptide screening. However, most of these models solely rely on one-dimensional amino acid sequences, overlooking crucial information required for the three-dimensional (3-D) space binding process. In this study, we propose TransfIGN, a structure-based DL model that is inspired by our previously developed framework, Interaction Graph Network (IGN), and incorporates sequence information from transformers to predict the interactions between HLA-A*02:01 and antigen peptides. Our model, trained on a comprehensive data set containing 61,816 sequences with 9051 binding affinity labels and 56,848 eluted ligand labels, achieves an area under the curve (AUC) of 0.893 on the binary data set, better than state-of-the-art sequence-based models trained on larger data sets such as NetMHCpan4.1, ANN, and TransPHLA. Furthermore, when evaluated on the IEDB weekly benchmark data sets, our predictions (AUC = 0.816) are better than those of the recommended methods like the IEDB consensus (AUC = 0.795). Notably, the interaction weight matrices generated by our method highlight the strong interactions at specific positions within peptides, emphasizing the model's ability to provide physical interpretability. This capability to unveil binding mechanisms through intricate structural features holds promise for new immunotherapeutic avenues.


Subject(s)
Deep Learning , HLA-A2 Antigen , Peptides , HLA-A2 Antigen/chemistry , HLA-A2 Antigen/metabolism , Peptides/chemistry , Peptides/metabolism , Humans , Protein Binding , Models, Molecular , Amino Acid Sequence , Protein Conformation
5.
J Immunol ; 208(1): 169-180, 2022 01 01.
Article in English | MEDLINE | ID: mdl-34853077

ABSTRACT

Adoptive T cell therapy with T cells expressing affinity-enhanced TCRs has shown promising results in phase 1/2 clinical trials for solid and hematological tumors. However, depth and durability of responses to adoptive T cell therapy can suffer from an inhibitory tumor microenvironment. A common immune-suppressive agent is TGF-ß, which is secreted by tumor cells and cells recruited to the tumor. We investigated whether human T cells could be engineered to be resistant to inhibition by TGF-ß. Truncating the intracellular signaling domain from TGF-ß receptor (TGFßR) II produces a dominant-negative receptor (dnTGFßRII) that dimerizes with endogenous TGFßRI to form a receptor that can bind TGF-ß but cannot signal. We previously generated specific peptide enhanced affinity receptor TCRs recognizing the HLA-A*02-restricted peptides New York esophageal squamous cell carcinoma 1 (NY-ESO-1)157-165/l-Ag family member-1A (TCR: GSK3377794, formerly NY-ESO-1c259) and melanoma Ag gene A10254-262 (TCR: ADP-A2M10, formerly melanoma Ag gene A10c796). In this article, we show that exogenous TGF-ß inhibited in vitro proliferation and effector functions of human T cells expressing these first-generation high-affinity TCRs, whereas inhibition was reduced or abolished in the case of second-generation TCRs coexpressed with dnTGFßRII (e.g., GSK3845097). TGF-ß isoforms and a panel of TGF-ß-associated genes are overexpressed in a range of cancer indications in which NY-ESO-1 is commonly expressed, particularly in synovial sarcoma. As an example, immunohistochemistry/RNAscope identified TGF-ß-positive cells close to T cells in tumor nests and stroma, which had low frequencies of cells expressing IFN-γ in a non-small cell lung cancer setting. Coexpression of dnTGFßRII may therefore improve the efficacy of TCR-transduced T cells.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Carcinoma, Squamous Cell/therapy , Hematologic Neoplasms/therapy , Immunotherapy, Adoptive/methods , Melanoma/therapy , Receptor, Transforming Growth Factor-beta Type II/metabolism , Receptors, Antigen, T-Cell/metabolism , Receptors, Chimeric Antigen/metabolism , Sarcoma, Synovial/therapy , Transforming Growth Factor beta/metabolism , Antigens, Neoplasm/immunology , Carcinoma, Squamous Cell/immunology , Cell Line, Tumor , Genetic Engineering , HLA-A2 Antigen/metabolism , Hematologic Neoplasms/immunology , Humans , Immune Tolerance , Melanoma/immunology , Membrane Proteins/immunology , Neoplasm Proteins/immunology , Peptide Fragments/immunology , Receptor, Transforming Growth Factor-beta Type II/genetics , Receptors, Antigen, T-Cell/genetics , Receptors, Chimeric Antigen/genetics , Sarcoma, Synovial/immunology , T-Cell Antigen Receptor Specificity , Tumor Microenvironment
6.
Gut ; 72(11): 2123-2137, 2023 11.
Article in English | MEDLINE | ID: mdl-36717219

ABSTRACT

OBJECTIVE: Exhausted hepatitis B virus (HBV)-specific CD8 T cells in chronic HBV infection are broadly heterogeneous. Characterisation of their functional impairment may allow to distinguish patients with different capacity to control infection and reconstitute antiviral function. DESIGN: HBV dextramer+CD8 T cells were analysed ex vivo for coexpression of checkpoint/differentiation markers, transcription factors and cytokines in 35 patients with HLA-A2+chronic hepatitis B (CHB) and in 29 control HBsAg negative CHB patients who seroconverted after NUC treatment or spontaneously. Cytokine production was also evaluated in HBV peptide-stimulated T cell cultures, in the presence or absence of antioxidant, polyphenolic, PD-1/PD-L1 inhibitor and TLR-8 agonist compounds and the effect on HBV-specific responses was further validated on additional 24 HLA-A2 negative CHB patients. RESULTS: Severely exhausted HBV-specific CD8 T cell subsets with high expression of inhibitory receptors, such as PD-1, TOX and CD39, were detected only in a subgroup of chronic viraemic patients. Conversely, a large predominance of functionally more efficient HBV-specific CD8 T cell subsets with lower expression of coinhibitory molecules and better response to in vitro immune modulation, typically detected after resolution of infection, was also observed in a proportion of chronic viraemic HBV patients. Importantly, the same subset of patients who responded more efficiently to in vitro immune modulation identified by HBV-specific CD8 T cell analysis were also identified by staining total CD8 T cells with PD-1, TOX, CD127 and Bcl-2. CONCLUSIONS: The possibility to distinguish patient cohorts with different capacity to respond to immune modulatory compounds in vitro by a simple analysis of the phenotypic CD8 T cell exhaustion profile deserves evaluation of its clinical applicability.


Subject(s)
Hepatitis B, Chronic , Hepatitis B , Humans , Hepatitis B, Chronic/drug therapy , Hepatitis B virus , HLA-A2 Antigen/metabolism , HLA-A2 Antigen/pharmacology , HLA-A2 Antigen/therapeutic use , Programmed Cell Death 1 Receptor/metabolism , CD8-Positive T-Lymphocytes
7.
J Virol ; 96(18): e0116621, 2022 09 28.
Article in English | MEDLINE | ID: mdl-36069549

ABSTRACT

Studies on Ebola virus disease (EVD) survivors and clinical studies on Ebola virus (EBOV) vaccine candidates have pinpointed the importance of a strong antibody response in protection and survival from EBOV infection. However, little is known about the T cell responses to EBOV or EBOV vaccines. We used HLA-A*02:01 (HLA-A2) transgenic mice to study HLA-A2-specific T cell responses elicited following vaccination with EBOV glycoprotein (EBOV-GP) presented with three different systems: (i) recombinant protein (rEBOV-GP), (ii) vesicular stomatitis replication-competent recombinant virus (VSV-EBOV-GP), and (iii) modified vaccinia Ankara virus recombinant (MVA-EBOV-GP). T cells from immunized animals were analyzed using peptide pools representing the entire GP region and individual peptides. Regardless of the vaccine formulation, we identified a minimal 9mer epitope containing an HLA-A2 motif (FLDPATTS), which was confirmed through HLA-A2 binding affinity and immunization studies. Using binding prediction software, we identified substitutions surrounding position 9 (S9V, P10V, and Q11V) that predicted enhanced binding to the HLA-A2 molecule. This enhanced binding was confirmed through in vitro binding studies and enhanced potency was shown with in vivo immunization studies using the enhanced sequences and the wild-type sequence. Of note, in silico studies predicted the enhanced 9mer epitope carrying the S9V substitution as the best overall HLA-A2 epitope for the full-length EBOV-GP. These results suggest that EBOV-GP-S9V and EBOV-GP-P10V represent more potent in vivo immunogens. Identification and enhancement of EBOV-specific human HLA epitopes could lead to the development of tools and reagents to induce more robust T cell responses in human subjects. IMPORTANCE Vaccine efficacy and immunity to viral infection are often measured by neutralizing antibody titers. T cells are specialized subsets of immune cells with antiviral activity, but this response is variable and difficult to track. We showed that the HLA-A2-specific T cell response to the Ebola virus glycoprotein can be enhanced significantly by a single residue substitution designed to improve an epitope binding affinity to one of the most frequent MHC alleles in the human population. This strategy could be applied to improve T cell responses to Ebola vaccines designed to elicit antibodies and adapted to target MHC alleles of populations in regions where endemic infections, like Ebola virus disease, are still causing outbreaks with concerning pandemic potential.


Subject(s)
Amino Acids , Ebolavirus , Epitopes, T-Lymphocyte , Glycoproteins , Hemorrhagic Fever, Ebola , Amino Acids/metabolism , Animals , Antibodies, Neutralizing , Antibodies, Viral , Ebola Vaccines/genetics , Ebolavirus/genetics , Epitopes, T-Lymphocyte/metabolism , HLA-A2 Antigen/genetics , HLA-A2 Antigen/metabolism , Humans , Mice , Recombinant Proteins , Vaccinia virus , Vesiculovirus
8.
J Biol Chem ; 297(3): 101065, 2021 09.
Article in English | MEDLINE | ID: mdl-34384783

ABSTRACT

CD8+ T cells play an important role in vaccination and immunity against severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection. Although numerous SARS-CoV-2 CD8+ T cell epitopes have been identified, the molecular basis underpinning T cell receptor (TCR) recognition of SARS-CoV-2-specific T cells remains unknown. The T cell response directed toward SARS-CoV-2 spike protein-derived S269-277 peptide presented by the human leukocyte antigen (HLA)-A∗02:01 allomorph (hereafter the HLA-A2S269-277 epitope) is, to date, the most immunodominant SARS-CoV-2 epitope found in individuals bearing this allele. As HLA-A2S269-277-specific CD8+ T cells utilize biased TRAV12 gene usage within the TCR α-chain, we sought to understand the molecular basis underpinning this TRAV12 dominance. We expressed four TRAV12+ TCRs which bound the HLA-A2S269-277 complex with low micromolar affinity and determined the crystal structure of the HLA-A2S269-277 binary complex, and subsequently a ternary structure of the TRAV12+ TCR complexed to HLA-A2S269-277. We found that the TCR made extensive contacts along the entire length of the S269-277 peptide, suggesting that the TRAV12+ TCRs would be sensitive to sequence variation within this epitope. To examine this, we investigated cross-reactivity toward analogous peptides from existing SARS-CoV-2 variants and closely related coronaviruses. We show via surface plasmon resonance and tetramer studies that the TRAV12+ T cell repertoire cross-reacts poorly with these analogous epitopes. Overall, we defined the structural basis underpinning biased TCR recognition of CD8+ T cells directed at an immunodominant epitope and provide a framework for understanding TCR cross-reactivity toward viral variants within the S269-277 peptide.


Subject(s)
Epitopes, T-Lymphocyte/chemistry , HLA-A2 Antigen/metabolism , Immunodominant Epitopes/metabolism , Receptors, Antigen, T-Cell/metabolism , Spike Glycoprotein, Coronavirus/metabolism , Humans , Protein Conformation , Receptors, Antigen, T-Cell/chemistry
9.
Proteins ; 90(9): 1645-1654, 2022 09.
Article in English | MEDLINE | ID: mdl-35403257

ABSTRACT

The startling diversity in αß T-cell receptor (TCR) sequences and structures complicates molecular-level analyses of the specificity and sensitivity determining T-cell immunogenicity. A number of three-dimensional (3D) structures are now available of ternary complexes between TCRs and peptides: major histocompatibility complexes (pMHC). Here, to glean molecular-level insights we analyze structures of TCRs bound to human class I nonamer peptide-MHC complexes. Residues at peptide positions 4-8 are found to be particularly important for TCR binding. About 90% of the TCRs hydrogen bond with one or both of the peptide residues at positions 4 and 8 presented by MHC allele HLA-A2, and this number is still ~79% for peptides presented by other MHC alleles. Residue 8, which lies outside the previously-identified central peptide region, is crucial for TCR recognition of class I MHC-presented nonamer peptides. The statistics of the interactions also sheds light on the MHC residues important for TCR binding. The present analysis will aid in the structural modeling of TCR:pMHC complexes and has implications for the rational design of peptide-based vaccines and T-cell-based immunotherapies.


Subject(s)
Peptides , Receptors, Antigen, T-Cell , HLA-A2 Antigen/chemistry , HLA-A2 Antigen/genetics , HLA-A2 Antigen/metabolism , Humans , Major Histocompatibility Complex , Peptides/chemistry , Protein Binding , Receptors, Antigen, T-Cell/genetics
10.
Eur J Immunol ; 51(6): 1505-1518, 2021 06.
Article in English | MEDLINE | ID: mdl-33835499

ABSTRACT

A T-cell receptor (TCR) with optimal avidity to a tumor antigen can be used to redirect T cells to eradicate cancer cells via adoptive cell transfer. Cancer testis antigens (CTAs) are attractive targets because they are expressed in the testis, which is immune-privileged, and in the tumor. However, CTAs are self-antigens and natural TCRs to CTAs have low affinity/avidity due to central tolerance. We previously described a method of directed evolution of TCR avidity using somatic hypermutation. In this study, we made several improvements to this method and enhanced the avidity of the hT27 TCR, which is specific for the cancer testis antigen HLA-A2-MAGE-A1278-286 . We identified eight point mutations with varying degrees of improved avidity. Human T cells transduced with TCRs containing these mutations displayed enhanced tetramer binding, IFN-γ and IL2 production, and cytotoxicity. Most of the mutations have retained specificity, except for one mutant with extremely high avidity. We demonstrate that somatic hypermutation is capable of optimizing avidity of clinically relevant TCRs for immunotherapy.


Subject(s)
Cancer Vaccines/immunology , Immunotherapy, Adoptive/methods , Neoplasm Proteins/immunology , Peptide Fragments/immunology , Receptors, Antigen, T-Cell/genetics , T-Lymphocytes/immunology , Cells, Cultured , Central Tolerance , Cytotoxicity, Immunologic , HLA-A2 Antigen/metabolism , Humans , Interferon-gamma/metabolism , Lymphocyte Activation , Point Mutation/genetics , Protein Binding , Receptors, Antigen, T-Cell/metabolism , Somatic Hypermutation, Immunoglobulin , T-Lymphocytes/transplantation
11.
J Immunol ; 205(2): 539-549, 2020 07 15.
Article in English | MEDLINE | ID: mdl-32571843

ABSTRACT

The availability of MHC-binding prediction tools has been useful in guiding studies aimed at identifying candidate target Ags to generate reactive T cells and to characterize viral and tumor-reactive T cells. Nevertheless, prediction algorithms appear to function poorly for epitopes containing cysteine (Cys) residues, which can oxidize and form disulfide bonds with other Cys residues under oxidizing conditions, thus potentially interfering with their ability to bind to MHC molecules. Analysis of the results of HLA-A*02:01 class I binding assays carried out in the presence and absence of the reducing agent 2-ME indicated that the predicted affinity for 25% of Cys-containing epitopes was underestimated by a factor of 3 or more. Additional analyses were undertaken to evaluate the responses of human CD8+ tumor-reactive T cells against 10 Cys-containing HLA class I-restricted minimal determinants containing substitutions of α-aminobutyric acid (AABA), a cysteine analogue containing a methyl group in place of the sulfhydryl group present in Cys, for the native Cys residues. Substitutions of AABA for Cys at putative MHC anchor positions often significantly enhanced T cell recognition, whereas substitutions at non-MHC anchor positions were neutral, except for one epitope where this modification abolished T cell recognition. These findings demonstrate the need to evaluate MHC binding and T cell recognition of Cys-containing peptides under conditions that prevent Cys oxidation, and to adjust current prediction binding algorithms for HLA-A*02:01 and potentially additional class I alleles to more accurately rank peptides containing Cys anchor residues.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Cysteine/metabolism , Neoplasms/immunology , Peptides/metabolism , Receptors, Antigen, T-Cell, alpha-beta/metabolism , Amino Acid Sequence , Antigen Presentation , Antigens, Neoplasm/immunology , Cells, Cultured , Epitopes, T-Lymphocyte/immunology , HLA-A2 Antigen/metabolism , Humans , Lymphocyte Activation , Protein Binding , T-Cell Antigen Receptor Specificity
12.
J Immunol ; 205(2): 454-468, 2020 07 15.
Article in English | MEDLINE | ID: mdl-32540992

ABSTRACT

A large proportion of the world's population harbors latent HSV type 1 (HSV-1). Cross-talk between antiviral CD8+ T cells and HSV-1 appear to control latency/reactivation cycles. We found that compared with healthy asymptomatic individuals, in symptomatic (SYMP) patients, the CD8+ T cells with the same HLA-A*0201-restricted HSV-1 epitope specificities expressed multiple genes and proteins associated to major T cell exhaustion pathways and were dysfunctional. Blockade of immune checkpoints with anti-LAG-3 and anti-PD-1 antagonist mAbs synergistically restored the frequency and function of antiviral CD8+ T cells, both 1) ex vivo, in SYMP individuals and SYMP HLA-A*0201 transgenic mice; and 2) in vivo in HSV-1-infected SYMP HLA-A*0201 transgenic mice. This was associated with a significant reduction in virus reactivation and recurrent ocular herpetic disease. These findings confirm antiviral CD8+ T cell exhaustion during SYMP herpes infection and pave the way to targeting immune checkpoints to combat recurrent ocular herpes.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Eye/immunology , Herpes Simplex/immunology , Herpesvirus 1, Human/physiology , Adult , Animals , Antibodies, Blocking/metabolism , Asymptomatic Diseases , Cells, Cultured , Disease Progression , Eye/virology , Female , HLA-A2 Antigen/metabolism , Host-Pathogen Interactions , Humans , Male , Mice, Transgenic , Middle Aged , Programmed Cell Death 1 Receptor/immunology , Programmed Cell Death 1 Receptor/metabolism , Virus Activation , Virus Latency , Young Adult
13.
Cancer Sci ; 112(3): 1161-1172, 2021 Mar.
Article in English | MEDLINE | ID: mdl-33410215

ABSTRACT

Activation of CD8+ Tax-specific CTL is a new therapeutic concept for adult T-cell leukemia (ATL) caused by HTLV-1. A recent clinical study of the dendritic cell vaccine pulsed with Tax peptides corresponding to CTL epitopes showed promising outcomes in ATL patients possessing limited human leukocyte antigen (HLA) alleles. In this study, we aimed to develop another immunotherapy to activate Tax-specific CTL without HLA limitation by using patients' own HTLV-1-infected cells as a vaccine. To examine the potential of HTLV-1-infected T-cells to activate CTL via antigen presenting cells, we established a unique co-culture system. We demonstrated that mitomycin C-treated HLA-A2-negative HTLV-1-infected T-cell lines or short-term cultured peripheral blood mononuclear cells (PBMC) derived from ATL patients induced cross-presentation of Tax antigen in co-cultured HLA-A2-positive antigen presenting cells, resulting in activation of HLA-A2-restricted CD8+ Tax-specific CTL. This effect was not inhibited by a reverse transcriptase inhibitor. IL-12 production and CD86 expression were also induced in antigen presenting cells co-cultured with HTLV-1-infected cells at various levels, which were improved by pre-treatment of the infected cells with histone deacetylase inhibitors. Furthermore, monocyte-derived dendritic cells induced from PBMC of a chronic ATL patient produced IL-12 and expressed enhanced levels of CD86 when co-cultured with autologous lymphocytes that had been isolated from the same PBMC and cultured for several days. These findings suggest that short-term cultured autologous PBMC from ATL patients could potentially serve as a vaccine to evoke Tax-specific CTL responses.


Subject(s)
Cancer Vaccines/administration & dosage , Cell Culture Techniques , HTLV-I Infections/therapy , Immunotherapy/methods , Leukemia-Lymphoma, Adult T-Cell/therapy , Leukocytes, Mononuclear/transplantation , Antigens, Viral/immunology , Cancer Vaccines/immunology , Cell Line, Tumor , Coculture Techniques , Cross-Priming/immunology , Gene Products, tax/immunology , HLA-A2 Antigen/immunology , HLA-A2 Antigen/metabolism , HTLV-I Infections/blood , HTLV-I Infections/immunology , Histone Deacetylase Inhibitors/pharmacology , Human T-lymphotropic virus 1/immunology , Humans , Leukemia-Lymphoma, Adult T-Cell/blood , Leukemia-Lymphoma, Adult T-Cell/immunology , Leukocytes, Mononuclear/drug effects , Leukocytes, Mononuclear/immunology , Mitomycin/pharmacology , T-Lymphocytes, Cytotoxic/immunology , Time Factors , Transplantation, Autologous
14.
Cancer Immunol Immunother ; 70(5): 1189-1202, 2021 May.
Article in English | MEDLINE | ID: mdl-33123756

ABSTRACT

Identification of neoepitopes as tumor-specific targets remains challenging, especially for cancers with low mutational burden, such as ovarian cancer. To identify mutated human leukocyte antigen (HLA) ligands as potential targets for immunotherapy in ovarian cancer, we combined mass spectrometry analysis of the major histocompatibility complex (MHC) class I peptidomes of ovarian cancer cells with parallel sequencing of whole exome and RNA in a patient with high-grade serous ovarian cancer. Four of six predicted mutated epitopes capable of binding to HLA-A*02:01 induced peptide-specific T cell responses in blood from healthy donors. In contrast, all six peptides failed to induce autologous peptide-specific response by T cells in peripheral blood or tumor-infiltrating lymphocytes from ascites of the patient. Surprisingly, T cell responses against a low-affinity p53-mutant Y220C epitope were consistently detected in the patient with either unprimed or in vitro peptide-stimulated T cells even though the patient's primary tumor did not bear this mutation. Our results demonstrated that tumor heterogeneity and distinct immune microenvironments within a patient should be taken into consideration for identification of immunogenic neoantigens. T cell responses to a driver gene-derived p53 Y220C mutation in ovarian cancer warrant further study.


Subject(s)
Antigens, Neoplasm/metabolism , Epitopes, T-Lymphocyte/metabolism , HLA-A2 Antigen/metabolism , Immunotherapy, Adoptive/methods , Mutation/genetics , Ovarian Neoplasms/immunology , T-Lymphocytes/immunology , Tumor Suppressor Protein p53/metabolism , Antigens, Neoplasm/genetics , Cells, Cultured , Epitopes, T-Lymphocyte/genetics , Female , HLA-A2 Antigen/genetics , Humans , Middle Aged , Neoplasm Staging , Tumor Microenvironment , Tumor Suppressor Protein p53/genetics , Exome Sequencing
15.
J Virol ; 94(7)2020 03 17.
Article in English | MEDLINE | ID: mdl-31915283

ABSTRACT

The HIV-1 accessory protein Nef downregulates the cell surface expression of major histocompatibility complex class I (MHC-I) molecules to facilitate virus spreading. The Nef-induced downregulation of MHC-I molecules such as HLA-A requires the clathrin adaptor protein 1 (AP-1) complex. The cooperative interaction of Nef, AP-1, and the cytosolic tail (CT) of HLA-A leads to a redirection of HLA-A targeting from the trans-Golgi network (TGN) to lysosomes for degradation. Although the γ-adaptin subunit of AP-1 has two distinct isoforms (γ1 and γ2), which may form two AP-1 complex variants, so far, only the importance of AP-1γ1 in MHC-I downregulation by Nef has been investigated. Here, we report that the AP-1γ2 isoform also participates in this process. We found that AP-1γ2 forms a complex with Nef and HLA-A2_CT and that this interaction depends on the Y320 residue in HLA-A2_CT and Nef expression. Moreover, Nef targets AP-1γ1 and AP-1γ2 to different compartments in T cells, and the depletion of either AP-1 variant impairs the Nef-mediated reduction of total endogenous HLA-A levels and rescues HLA-A levels on the cell surface. Finally, immunofluorescence and immunoelectron microscopy analyses reveal that the depletion of γ2 in T cells compromises both the Nef-mediated retention of HLA-A molecules in the TGN and targeting to multivesicular bodies/late endosomes. Altogether, these results show that in addition to AP-1γ1, Nef also requires the AP-1γ2 variant for efficient MHC-I downregulation.IMPORTANCE HIV-1 Nef mediates evasion of the host immune system by inhibiting MHC-I surface presentation of viral antigens. To achieve this goal, Nef modifies the intracellular trafficking of MHC-I molecules in several ways. Despite being the subject of intense study, the molecular details underlying these modifications are not yet fully understood. Adaptor protein 1 (AP-1) plays an essential role in the Nef-mediated downregulation of MHC-I molecules such as HLA-A in different cell types. However, AP-1 has two functionally distinct variants composed of either γ1 or γ2 subunit isoforms. Because previous studies on the role of AP-1 in MHC-I downregulation by Nef focused on AP-1γ1, an important open question is the participation of AP-1γ2 in this process. Here, we show that AP-1γ2 is also essential for Nef-mediated depletion of surface HLA-A molecules in T cells. Our results indicate that Nef hijacks AP-1γ2 to modify HLA-A intracellular transport, redirecting these proteins to lysosomes for degradation.


Subject(s)
Down-Regulation , Gene Expression Regulation , HLA-A2 Antigen/metabolism , Transcription Factor AP-1/metabolism , nef Gene Products, Human Immunodeficiency Virus/metabolism , Adaptor Protein Complex gamma Subunits/metabolism , Cell Membrane/metabolism , Cytosol/metabolism , Endosomes/metabolism , HEK293 Cells , HeLa Cells , Humans , Lysosomes/metabolism , Microscopy, Immunoelectron , Protein Transport , T-Lymphocytes/immunology , T-Lymphocytes/virology , trans-Golgi Network/metabolism
16.
Transfusion ; 61(4): 1023-1028, 2021 04.
Article in English | MEDLINE | ID: mdl-33241556

ABSTRACT

BACKGROUND: Platelet (PLT) transfusions are an essential treatment for bleeding disorders. However, immunologic complications can occur, including alloantibody production against Class I HLA molecules. The principal source of HLA molecules in PLT concentrates (PCs) is the PLTs themselves. However, extracellular microparticles (MPs) present in PCs may express HLA molecules. STUDY DESIGN AND METHODS: We used nanoscale flow cytometry to explore the expression of HLA-A2, HLA-B7, and HLA-B57 on the surface of cells, PLT-derived MPs (PMPs), lymphocyte-derived MPs (LMPs), and monocyte-derived MPs (MMPs) present in PCs. Expression was studied during 7 days of storage. RESULTS: Platelets were not the only source of HLA molecules in PCs. HLA molecules were present on PMPs, LMPs, and MMPs. The level of HLA Class I molecule expression varied between haplotypes and MPs of different origins and during storage. CONCLUSION: Platelets or residual cells remaining after leukoreduction are not the only source of HLA Class I molecules in PCs, highlighting the contribution of MPs to alloimmunization mechanisms. These data may be relevant for the development of new transfusion guidelines.


Subject(s)
Hemorrhage/therapy , Isoantibodies/immunology , Phosphoproteins/immunology , Platelet Transfusion/adverse effects , Platelet Transfusion/methods , Blood Donors , Blood Platelets/immunology , Blood Platelets/metabolism , Cell-Derived Microparticles/metabolism , Flow Cytometry/methods , HLA-A2 Antigen/metabolism , HLA-B Antigens/metabolism , HLA-B7 Antigen/metabolism , Healthy Volunteers , Humans , Lymphocytes/immunology , Lymphocytes/metabolism , Monocytes/immunology , Monocytes/metabolism , Phosphoproteins/classification , Phosphoproteins/metabolism
17.
J Immunol ; 202(2): 451-459, 2019 01 15.
Article in English | MEDLINE | ID: mdl-30559321

ABSTRACT

The intracellular pathway of cross-presentation, which allows MHC class I-restricted presentation of peptides derived from exogenous Ags, remains poorly defined and may vary with the nature of the exogenous Ag and the type of APC. It can be cytosolic, characterized by proteasome and TAP dependency, or vacuolar, usually believed to be proteasome and TAP independent. Cross-presentation is particularly effective with long synthetic peptides, and we previously reported that the HLA-A2-restricted cross-presentation of a long peptide derived from melanoma Ag gp100 by human monocyte-derived immature dendritic cells occurred in a vacuolar pathway, making use of newly synthesized HLA-A2 molecules that follow a nonclassical secretion route. In this article, we show that the HLA-A1-restricted cross-presentation of a long peptide derived from tumor Ag MAGE-A3 by human monocyte-derived immature dendritic cells also follows a vacuolar pathway. However, as opposed to the HLA-A2-restricted peptide, cross-presentation of the HLA-A1-restricted peptide is TAP dependent. We show that this paradoxical TAP-dependency is indirect and reflects the need for TAP to load HLA-A1 molecules with peptides in the endoplasmic reticulum, to allow them to escape the endoplasmic reticulum and reach the vacuole, where peptide exchange with the cross-presented peptide likely occurs. Our results confirm and extend the involvement of the vacuolar pathway in the cross-presentation of long peptides, and indicate that TAP-dependency can no longer be used as a key criterion to distinguish the cytosolic from the vacuolar pathway of cross-presentation. They also stress the existence of an alternative secretory route for MHC class I, which will be worthy of further studies.


Subject(s)
ATP-Binding Cassette Transporters/metabolism , Antigens, Neoplasm/metabolism , Dendritic Cells/immunology , Endoplasmic Reticulum/metabolism , HLA-A1 Antigen/metabolism , Neoplasm Proteins/metabolism , T-Lymphocytes, Cytotoxic/immunology , Vacuoles/metabolism , Antigen Presentation , Cell Line , Cross-Priming , Cytosol/metabolism , HLA-A2 Antigen/metabolism , Humans , Peptides/metabolism , gp100 Melanoma Antigen/metabolism
18.
Phys Chem Chem Phys ; 23(4): 2836-2845, 2021 Feb 04.
Article in English | MEDLINE | ID: mdl-33470998

ABSTRACT

Melanoma is a type of skin cancer with increasing incidence worldwide and high lethality. Conventional forms of treatment are not effective in advanced cancer stages. Hence, immunotherapeutic approaches have been tested to modulate immune response against tumor cells. Some vaccine models using tumor-associated antigens (TAAs) such as glycoprotein 100 (gp100) have been studied, but their expected effectiveness has not been shown until now. Antigen immunogenicity is a crucial point to improve the immune response, and therefore mutations are inserted in peptide sequences. It is possible to understand the interactions which occur between peptides and immune system molecules through computer simulation, and this is essential in order to guide efficient vaccine models. In this work, we have calculated the interaction binding energies of crystallographic data based on modified gp100 peptides and HLA-A*0201 using density functional theory (DFT) and the molecular fractionation with conjugated caps (MFCC) approach. Our results show the most relevant residue-residue interactions, the impact of three mutations in their binding sites, and the main HLA-A*0201 amino acids for peptide-HLA binding.


Subject(s)
HLA-A2 Antigen/metabolism , gp100 Melanoma Antigen/metabolism , Computer Simulation , Density Functional Theory , HLA-A2 Antigen/chemistry , Humans , Models, Chemical , Mutation , Peptide Fragments , Protein Binding , Thermodynamics , gp100 Melanoma Antigen/chemistry , gp100 Melanoma Antigen/genetics
19.
J Biol Chem ; 294(7): 2470-2485, 2019 02 15.
Article in English | MEDLINE | ID: mdl-30554144

ABSTRACT

The membrane-associated RING-CH (MARCH) family of membrane-bound E3 ubiquitin ligases regulates the levels of cell-surface membrane proteins, many of which are involved in immune responses. Although their role in ubiquitin-dependent endocytosis and degradation of cell-surface proteins is extensively documented, the features of MARCH proteins and their substrates that drive the molecular recognition events leading to ubiquitin transfer remain poorly defined. In this study, we sought to determine the features of human MARCH9 that are required for regulating the surface levels of its substrate proteins. Consistent with previous studies of other MARCH proteins, we found that susceptibility to MARCH9 activity is encoded in the transmembrane (TM) domains of its substrates. Accordingly, substitutions at specific residues and motifs within MARCH9's TM domains resulted in varying degrees of functional impairment. Most notably, a single serine-to-alanine substitution in the first of its two TM domains rendered MARCH9 completely unable to alter the surface levels of two different substrates: the major histocompatibility class I molecule HLA-A2 and the T-cell co-receptor CD4. Solution NMR analysis of a MARCH9 fragment encompassing the two TM domains and extracellular connecting loop revealed that the residues contributing most to MARCH9 activity are located in the α-helical portions of TM1 and TM2 that are closest to the extracellular face of the lipid bilayer. This observation defines a key region required for substrate regulation. In summary, our biochemical and structural findings demonstrate that specific sequences in the α-helical MARCH9 TM domains make crucial contributions to its ability to down-regulate its protein substrates.


Subject(s)
Down-Regulation , Gene Expression Regulation, Enzymologic , Membrane Proteins/biosynthesis , Ubiquitin-Protein Ligases/biosynthesis , CD4 Antigens/chemistry , CD4 Antigens/genetics , CD4 Antigens/metabolism , HEK293 Cells , HLA-A2 Antigen/chemistry , HLA-A2 Antigen/genetics , HLA-A2 Antigen/metabolism , Humans , Membrane Proteins/chemistry , Membrane Proteins/genetics , Protein Domains , Protein Structure, Secondary , Serine/chemistry , Serine/genetics , Serine/metabolism , Ubiquitin-Protein Ligases/chemistry , Ubiquitin-Protein Ligases/genetics
20.
Eur J Immunol ; 49(8): 1278-1290, 2019 08.
Article in English | MEDLINE | ID: mdl-31054264

ABSTRACT

Introduction of Chimeric Antigen Receptors to NK cells has so far been the main practical method for targeting NK cells to specific surface antigens. In contrast, T cell receptor (TCR) gene delivery can supply large populations of cytotoxic T-lymphocytes (CTL) targeted against intracellular antigens. However, a major barrier in the development of safe CTL-TCR therapies exists, wherein the mispairing of endogenous and genetically transferred TCR subunits leads to formation of TCRs with off-target specificity. To overcome this and enable specific intracellular antigen targeting, we have tested the use of NK cells for TCR gene transfer to human cells. Our results show that ectopic expression of TCR α/ß chains, along with CD3 subunits, enables the functional expression of an antigen-specific TCR complex on NK cell lines NK-92 and YTS, demonstrated by using a TCR against the HLA-A2-restricted tyrosinase-derived melanoma epitope, Tyr368-377 . Most importantly, the introduction of a TCR complex to NK cell lines enables MHC-restricted, antigen-specific killing of tumor cells both in vitro and in vivo. Targeting of NK cells via TCR gene delivery stands out as a novel tool in the field of adoptive immunotherapy which can also overcome the major hurdle of "mispairing" in TCR gene therapy.


Subject(s)
Immunotherapy, Adoptive/methods , Killer Cells, Natural/physiology , Melanoma/therapy , Receptors, Antigen, T-Cell, alpha-beta/genetics , Receptors, Chimeric Antigen/genetics , Antigens, Neoplasm/immunology , Cell Line , Cytotoxicity, Immunologic , HLA-A2 Antigen/metabolism , Humans , Killer Cells, Natural/transplantation , Melanoma/immunology , Monophenol Monooxygenase/immunology , Peptides/immunology , Protein Engineering
SELECTION OF CITATIONS
SEARCH DETAIL