Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 51
Filter
1.
J Mol Biol ; 431(24): 4941-4958, 2019 12 06.
Article in English | MEDLINE | ID: mdl-31704286

ABSTRACT

The coreceptor CD8αß can greatly promote activation of T cells by strengthening T-cell receptor (TCR) binding to cognate peptide-MHC complexes (pMHC) on antigen presenting cells and by bringing p56Lck to TCR/CD3. Here, we demonstrate that CD8 can also bind to pMHC on the T cell (in cis) and that this inhibits their activation. Using molecular modeling, fluorescence resonance energy transfer experiments on living cells, biochemical and mutational analysis, we show that CD8 binding to pMHC in cis involves a different docking mode and is regulated by posttranslational modifications including a membrane-distal interchain disulfide bond and negatively charged O-linked glycans near positively charged sequences on the CD8ß stalk. These modifications distort the stalk, thus favoring CD8 binding to pMHC in cis. Differential binding of CD8 to pMHC in cis or trans is a means to regulate CD8+ T-cell responses and provides new translational opportunities.


Subject(s)
CD8 Antigens/metabolism , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Histocompatibility Antigens/chemistry , Histocompatibility Antigens/immunology , Multiprotein Complexes/metabolism , Peptides/metabolism , Amino Acid Sequence , Animals , CD8 Antigens/chemistry , CD8 Antigens/genetics , Histocompatibility Antigens/genetics , Lymphocyte Activation/immunology , Mice , Mice, Knockout , Models, Biological , Models, Molecular , Multiprotein Complexes/chemistry , Multiprotein Complexes/immunology , Mutation , Peptides/chemistry , Peptides/immunology , Protein Binding , Protein Conformation , Protein Interaction Domains and Motifs , Structure-Activity Relationship
2.
Biotechnol Lett ; 40(1): 85-92, 2018 Jan.
Article in English | MEDLINE | ID: mdl-28993910

ABSTRACT

OBJECTIVES: To investigate the effects of operational process conditions on expression of MHC class II protein from a stable Drosophila S2 cell line. RESULTS: When the Drosophila S2 cells were grown in vented orbitally shaken TubeSpin bioreactor 600 containers, cell growth was improved three-fold and the yield of recombinant major histocompatibility (MHC) class II protein (HLA-DR12xHis) increased four-fold over the levels observed for the same cells cultivated in roller bottles (RB) without vented caps. Culturing in RB with vented caps while increasing the rotation speed from 6 rpm to 18 rpm also improved cell growth five-fold and protein productivity three-fold which is comparable to the levels observed in the orbitally shaken containers. Protein activity was found to be almost identical between the two vessel systems tested. CONCLUSIONS: Optimized cell culture conditions and a more efficient vessel type can enhance gas transfer and mixing and lead to substantial improvement of recombinant product yields from S2 cells.


Subject(s)
Cell Culture Techniques/methods , HLA-DR1 Antigen/biosynthesis , Recombinant Proteins/biosynthesis , Animals , Bioreactors , Biotechnology/methods , Cell Line , Cell Proliferation , Drosophila , HLA-DR1 Antigen/genetics , Recombinant Proteins/genetics
3.
Oncoimmunology ; 5(7): e1177691, 2016 Jul.
Article in English | MEDLINE | ID: mdl-27622028

ABSTRACT

Tumor exome and RNA sequencing data provide a systematic and unbiased view on cancer-specific expression, over-expression, and mutations of genes, which can be mined for personalized cancer vaccines and other immunotherapies. Of key interest are tumor-specific mutations, because T cells recognizing neoepitopes have the potential to be highly tumoricidal. Here, we review recent developments and technical advances in identifying MHC class I and class II-restricted tumor antigens, especially neoantigen derived MHC ligands, including in silico predictions, immune-peptidome analysis by mass spectrometry, and MHC ligand validation by biochemical methods on T cells.

4.
Oncoimmunology ; 5(3): e1086861, 2016 Mar.
Article in English | MEDLINE | ID: mdl-27141342

ABSTRACT

Cytotoxic T lymphocytes (CTL) from CD8ß-deficient mice have powerful FasL-mediated cytotoxicity and IFNγ responses, but ablated Ca2+ and NFAT signaling, which can be restored by transduction with CD8ß. Upon infection with lymphocytic choriomeningitis virus (LCMV), these cells yielded GP33-specific CTL (CD8ßR) that exhibited high FasL/Fas-mediated cytotoxicity, IFNγ CXCL9 and 10 chemokine responses. Transfer of these cells in B16-GP33 tumor bearing mice resulted in (i) massive T cell tumor infiltration, (ii) strong reduction of myeloid-derived suppressor cells (MDSCs), regulatory T cells (Treg) and IL-17-expressing T helper cells, (iii) maturation of tumor-associated antigen-presenting cells and (iv) production of endogenous, B16 melanoma-specific CTL that eradicated the tumor long after the transferred CD8ßR CTL perished. Our study demonstrates that the synergistic combination of strong Fas/FasL mediated cytotoxicity, IFNγ and CXCL9 and 10 responses endows adoptively transferred CTL to reprogram the tumor environment and to thus enable the generation of endogenous, tumoricidal immunity.

5.
Sci Rep ; 5: 14896, 2015 Oct 08.
Article in English | MEDLINE | ID: mdl-26447332

ABSTRACT

Tumor antigen-specific CD4(+) T cells generally orchestrate and regulate immune cells to provide immune surveillance against malignancy. However, activation of antigen-specific CD4(+) T cells is restricted at local tumor sites where antigen-presenting cells (APCs) are frequently dysfunctional, which can cause rapid exhaustion of anti-tumor immune responses. Herein, we characterize anti-tumor effects of a unique human CD4(+) helper T-cell subset that directly recognizes the cytoplasmic tumor antigen, NY-ESO-1, presented by MHC class II on cancer cells. Upon direct recognition of cancer cells, tumor-recognizing CD4(+) T cells (TR-CD4) potently induced IFN-γ-dependent growth arrest in cancer cells. In addition, direct recognition of cancer cells triggers TR-CD4 to provide help to NY-ESO-1-specific CD8(+) T cells by enhancing cytotoxic activity, and improving viability and proliferation in the absence of APCs. Notably, the TR-CD4 either alone or in collaboration with CD8(+) T cells significantly inhibited tumor growth in vivo in a xenograft model. Finally, retroviral gene-engineering with T cell receptor (TCR) derived from TR-CD4 produced large numbers of functional TR-CD4. These observations provide mechanistic insights into the role of TR-CD4 in tumor immunity, and suggest that approaches to utilize TR-CD4 will augment anti-tumor immune responses for durable therapeutic efficacy in cancer patients.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Cytotoxicity, Immunologic/immunology , Ovarian Neoplasms/immunology , T-Lymphocytes, Helper-Inducer/immunology , Animals , Antigens, Neoplasm/immunology , Antigens, Neoplasm/metabolism , CD4-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Cell Survival/immunology , Cells, Cultured , Coculture Techniques , Female , Humans , Interferon-gamma/immunology , Interferon-gamma/metabolism , Interferon-gamma/pharmacology , Jurkat Cells , Membrane Proteins/immunology , Membrane Proteins/metabolism , Mice, SCID , Ovarian Neoplasms/pathology , T-Lymphocytes, Helper-Inducer/metabolism , Transplantation, Heterologous , Tumor Burden/immunology , Tumor Necrosis Factor-alpha/immunology , Tumor Necrosis Factor-alpha/metabolism , Tumor Necrosis Factor-alpha/pharmacology
6.
J Immunol ; 195(9): 4210-7, 2015 Nov 01.
Article in English | MEDLINE | ID: mdl-26416277

ABSTRACT

Abs bind to unprocessed Ags, whereas cytotoxic CD8(+) T cells recognize peptides derived from endogenously processed Ags presented in the context of class I MHC complexes. We screened, by ELISA, human sera for Abs reacting specifically with the influenza matrix protein (IMP)-derived peptide(58-66) displayed by HLA-A*0201 complexes. Among 653 healthy volunteers, blood donors, and women on delivery, high-titered HLA-A*0201/IMP(58-66) complex-specific IgG Abs were detected in 11 females with a history of pregnancies and in 1 male, all HLA-A*0201(-). These Abs had the same specificity as HLA-A*0201/IMP(58-66)-specific cytotoxic T cells and bound neither to HLA-A*0201 nor the peptide alone. No such Abs were detected in HLA-A*0201(+) volunteers. These Abs were not cross-reactive to other self-MHC class I alleles displaying IMP(58-66), but bound to MHC class I complexes of an HLA nonidentical offspring. HLA-A*0201/IMP(58-66) Abs were also detected in the cord blood of newborns, indicating that HLA-A*0201/IMP(58-66) Abs are produced in HLA-A*0201(-) mothers and enter the fetal blood system. That Abs can bind to peptides derived from endogenous Ags presented by MHC complexes opens new perspectives on interactions between the cellular and humoral immune system.


Subject(s)
HLA-A2 Antigen/immunology , Isoantibodies/immunology , Peptide Fragments/immunology , Viral Matrix Proteins/immunology , Antibody Specificity , Female , Fetal Blood/immunology , Humans , Immunity, Humoral , MCF-7 Cells , Male
7.
Proc Natl Acad Sci U S A ; 111(11): E1007-15, 2014 Mar 18.
Article in English | MEDLINE | ID: mdl-24594598

ABSTRACT

CD8αß plays crucial roles in the thymic selection, differentiation, and activation of some, but not all, CD8(+) T cells, whereas CD8αα does not. To investigate these roles, we produced mice that expressed transgene P14 T-cell receptor ß (TCRß) chain and CD8ß or did not (WT and KO mice, respectively). The primary CD8(+) T-cell response to acute lymphocytic choriomeningitis virus (LCMV) infection was predominantly D(b)/GP33 specific and CD8 independent in KO mice and was mostly CD8 dependent in WT mice. Cytotoxic T lymphocytes (CTL) from KO mice failed to mobilize intracellular Ca(2+) and to kill via perforin/granzyme. Their strong Fas/FasL-mediated cytotoxicity and IFN-γ response were signaled via a Ca(2+)-independent, PI3K-dependent pathway. This was also true for 15-20% of CD8-independent CTL found in WT mice. Conversely, the perforin/granzyme-mediated killing and IFN-γ response of CD8-dependent CTL were signaled via a Ca(2+), p56(lck), and nuclear factor of activated T cells-dependent pathway. Deep sequencing of millions of TCRα chain transcripts revealed that the TCR repertoires of preimmune CD8(+) T cells were highly diverse, but those of LCMV D(b)/GP33-specific CTL, especially from KO mice, were narrow. The immune repertoires exhibited biased use of Vα segments that encoded different complementary-determining region 1α (CDR1α) and CDR2α sequences. We suggest that TCR from WT CD8-independent T cells may engage MHC-peptide complexes in a manner unfavorable for efficient CD8 engagement and Ca(2+) signaling but permissive for Ca(2+)-independent, PI3K-dependent signaling. This duality of the CD8 compartment may provide organisms with broader protective immunity.


Subject(s)
CD8 Antigens/metabolism , Cell Differentiation/immunology , Immunity, Cellular/immunology , NFATC Transcription Factors/metabolism , Signal Transduction/immunology , T-Lymphocytes, Cytotoxic/metabolism , Animals , Base Sequence , Calcium/metabolism , Cell Line, Tumor , High-Throughput Nucleotide Sequencing , Mice , Mice, Knockout , Molecular Sequence Data , Phosphatidylinositol 3-Kinases/metabolism , Receptors, Antigen, T-Cell/genetics , Receptors, Antigen, T-Cell/metabolism , T-Lymphocytes, Cytotoxic/physiology
8.
Cancer Res ; 74(4): 1045-55, 2014 Feb 15.
Article in English | MEDLINE | ID: mdl-24343228

ABSTRACT

Although melanoma vaccines stimulate tumor antigen-specific CD8(+) T cells, objective clinical responses are rarely observed. To investigate this discrepancy, we evaluated the character of vaccine-induced CD8(+) T cells with regard to the inhibitory T-cell coreceptors PD-1 and Tim-3 in patients with metastatic melanoma who were administered tumor vaccines. The vaccines included incomplete Freund's adjuvant, CpG oligodeoxynucleotide (CpG), and the HLA-A2-restricted analog peptide NY-ESO-1 157-165V, either by itself or in combination with the pan-DR epitope NY-ESO-1 119-143. Both vaccines stimulated rapid tumor antigen-specific CD8(+) T-cell responses detected ex vivo, however, tumor antigen-specific CD8(+) T cells produced more IFN-γ and exhibited higher lytic function upon immunization with MHC class I and class II epitopes. Notably, the vast majority of vaccine-induced CD8(+) T cells upregulated PD-1 and a minority also upregulated Tim-3. Levels of PD-1 and Tim-3 expression by vaccine-induced CD8(+) T cells at the time of vaccine administration correlated inversely with their expansion in vivo. Dual blockade of PD-1 and Tim-3 enhanced the expansion and cytokine production of vaccine-induced CD8(+) T cells in vitro. Collectively, our findings support the use of PD-1 and Tim-3 blockades with cancer vaccines to stimulate potent antitumor T-cell responses and increase the likelihood of clinical responses in patients with advanced melanoma.


Subject(s)
CD8-Positive T-Lymphocytes/physiology , Cancer Vaccines/therapeutic use , Cell Proliferation , Lymphocyte Activation/genetics , Melanoma/therapy , Membrane Proteins/physiology , Programmed Cell Death 1 Receptor/physiology , CD4-Positive T-Lymphocytes/physiology , CD8-Positive T-Lymphocytes/pathology , Cancer Vaccines/immunology , Freund's Adjuvant/therapeutic use , Hepatitis A Virus Cellular Receptor 2 , Histocompatibility Antigens Class I/immunology , Histocompatibility Antigens Class II/immunology , Humans , Interferon-gamma/metabolism , Lipids/therapeutic use , Melanoma/genetics , Melanoma/immunology , Oligodeoxyribonucleotides/therapeutic use , T-Cell Antigen Receptor Specificity , Treatment Outcome
9.
Proc Natl Acad Sci U S A ; 110(3): E221-30, 2013 Jan 15.
Article in English | MEDLINE | ID: mdl-23267099

ABSTRACT

Cytotoxic T lymphocyte-associated antigen-4 (CTLA-4; CD152) is of pivotal importance for self-tolerance, with deficiency or unfavorable polymorphisms leading to autoimmune disease. Tolerance to self-antigens is achieved through thymic deletion of highly autoreactive conventional T (Tconv) cells and generation of FoxP3(+) regulatory T (Treg) cells. The main costimulatory molecule, CD28, augments the negative selection of Tconv cells and promotes the generation of FoxP3(+) Treg cells. The role of its antagonistic homolog CTLA-4, however, remains a topic of debate. To address this topic, we investigated the thymic development of T cells in the presence and absence of CTLA-4 in a T-cell receptor (TCR) transgenic mouse model specific for the myelin basic protein peptide Ac1-9. We reveal that CTLA-4 is expressed in the corticomedullary region of the thymus. Its absence alters the response of CD4(+)CD8(-) thymocytes to self-antigen recognition, which affects the quantity of the Treg cells generated and broadens the repertoire of peripheral Tconv cells. T-cell repertoire alteration after deletion of CTLA-4 results from changes in TCR Vα and Jα segment selection as well as CDR3α composition in Tconv and Treg cells. CTLA-4, therefore, regulates the early development of self-reactive T cells in the thymus and plays a key role in central tolerance.


Subject(s)
CTLA-4 Antigen/immunology , Gene Rearrangement, T-Lymphocyte , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes/immunology , Amino Acid Sequence , Animals , Antigenic Variation , CTLA-4 Antigen/deficiency , CTLA-4 Antigen/genetics , Cell Differentiation , Complementarity Determining Regions , Cytokines/biosynthesis , Dendritic Cells/cytology , Dendritic Cells/immunology , Encephalomyelitis, Autoimmune, Experimental/genetics , Encephalomyelitis, Autoimmune, Experimental/immunology , Female , Male , Mice , Mice, Knockout , Mice, Transgenic , Molecular Sequence Data , Receptors, Antigen, T-Cell, alpha-beta/genetics , Self Tolerance , T-Lymphocytes/cytology , T-Lymphocytes, Regulatory/cytology , Thymus Gland/cytology , Thymus Gland/growth & development , Thymus Gland/immunology
10.
J Immunol Methods ; 387(1-2): 173-80, 2013 Jan 31.
Article in English | MEDLINE | ID: mdl-23098837

ABSTRACT

Establishing CD8(+) T cell cultures has been empirical and the published methods have been largely individual laboratory based. In this study, we optimized culturing conditions and show that IL-2 concentration is the most critical factor for the success of establishing CD8(+) T cell cultures. High IL-2 concentration encouraged T cells to non-specifically proliferate, express a B cell marker, B220, and undergo apoptosis. These cells also lose typical irregular T cell morphology and are incapable of sustaining long-term cultures. Using tetramer and intracellular cytokine assessments, we further demonstrated that many antigen-specific T cells have been rendered nonfunctional when expanded under high IL-2 concentration. When IL-2 is used in the correct range, B220-mediated cell depletion greatly enhanced the success rate of such T cell cultures.


Subject(s)
Apoptosis/immunology , CD8-Positive T-Lymphocytes/immunology , Cell Culture Techniques/methods , Leukocyte Common Antigens/immunology , Amino Acid Sequence , Animals , Apoptosis/drug effects , CD8-Positive T-Lymphocytes/drug effects , CD8-Positive T-Lymphocytes/metabolism , Cell Proliferation/drug effects , Cells, Cultured , Dose-Response Relationship, Drug , Interferon-gamma/immunology , Interferon-gamma/metabolism , Interleukin-2/pharmacology , Leukocyte Common Antigens/metabolism , Lymphocyte Count , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL
12.
J Biol Chem ; 287(51): 42664-74, 2012 Dec 14.
Article in English | MEDLINE | ID: mdl-23091059

ABSTRACT

The T-cell antigen receptor (TCR) exists in monomeric and nanoclustered forms independently of antigen binding. Although the clustering is involved in the regulation of T-cell sensitivity, it is unknown how the TCR nanoclusters form. We show that cholesterol is required for TCR nanoclustering in T cells and that this clustering enhances the avidity but not the affinity of the TCR-antigen interaction. Investigating the mechanism of the nanoclustering, we found that radioactive photocholesterol specifically binds to the TCRß chain in vivo. In order to reduce the complexity of cellular membranes, we used a synthetic biology approach and reconstituted the TCR in liposomes of defined lipid composition. Both cholesterol and sphingomyelin were required for the formation of TCR dimers in phosphatidylcholine-containing large unilamellar vesicles. Further, the TCR was localized in the liquid disordered phase in giant unilamellar vesicles. We propose a model in which cholesterol and sphingomyelin binding to the TCRß chain causes TCR dimerization. The lipid-induced TCR nanoclustering enhances the avidity to antigen and thus might be involved in enhanced sensitivity of memory compared with naive T cells. Our work contributes to the understanding of the function of specific nonannular lipid-membrane protein interactions.


Subject(s)
Cholesterol/metabolism , Nanoparticles/chemistry , Receptors, Antigen, T-Cell, alpha-beta/metabolism , Receptors, Antigen, T-Cell/metabolism , Sphingomyelins/metabolism , Animals , Cell Membrane/metabolism , Cell Survival , Cluster Analysis , Cross-Linking Reagents/metabolism , Histocompatibility Antigens/metabolism , Humans , Ligands , Mice , Peptides/metabolism , Phosphatidylcholines/metabolism , Protein Binding , Protein Multimerization , Protein Subunits/metabolism , Receptors, Antigen, B-Cell/metabolism , Receptors, Transferrin , Unilamellar Liposomes/metabolism
13.
Proc Natl Acad Sci U S A ; 109(45): E3111-8, 2012 Nov 06.
Article in English | MEDLINE | ID: mdl-23077253

ABSTRACT

The bias of αß T cells for MHC ligands has been proposed to be intrinsic to the T-cell receptor (TCR). Equally, the CD4 and CD8 coreceptors contribute to ligand restriction by colocalizing Lck with the TCR when MHC ligands are engaged. To determine the importance of intrinsic ligand bias, the germ-line TCR complementarity determining regions were extensively diversified in vivo. We show that engagement with MHC ligands during thymocyte selection and peripheral T-cell activation imposes remarkably little constraint over TCR structure. Such versatility is more consistent with an opportunist, rather than a predetermined, mode of interface formation. This hypothesis was experimentally confirmed by expressing a hybrid TCR containing TCR-γ chain germ-line complementarity determining regions, which engaged efficiently with MHC ligands.


Subject(s)
Major Histocompatibility Complex/immunology , Receptors, Antigen, T-Cell, alpha-beta/immunology , Amino Acid Sequence , Animals , Base Sequence , Cell Lineage/immunology , Complementarity Determining Regions/genetics , Complementarity Determining Regions/immunology , Germ Cells/immunology , Ligands , Mice , Mice, Inbred C57BL , Molecular Sequence Data , Mutation/genetics , Receptors, Antigen, T-Cell, alpha-beta/chemistry , Receptors, Antigen, T-Cell, alpha-beta/genetics , Receptors, Antigen, T-Cell, gamma-delta/immunology , Recombination, Genetic/genetics , Selection, Genetic , Thymus Gland/immunology
14.
PLoS One ; 7(9): e44707, 2012.
Article in English | MEDLINE | ID: mdl-22970293

ABSTRACT

NY-ESO-1 has been a major target of many immunotherapy trials because it is expressed by various cancers and is highly immunogenic. In this study, we have identified a novel HLA-B*1801-restricted CD8(+) T cell epitope, NY-ESO-1(88-96) (LEFYLAMPF) and compared its direct- and cross-presentation to that of the reported NY-ESO-1(157-165) epitope restricted to HLA-A*0201. Although both epitopes were readily cross-presented by DCs exposed to various forms of full-length NY-ESO-1 antigen, remarkably NY-ESO-1(88-96) is much more efficiently cross-presented from the soluble form, than NY-ESO-1(157-165). On the other hand, NY-ESO-1(157-165) is efficiently presented by NY-ESO-1-expressing tumor cells and its presentation was not enhanced by IFN-γ treatment, which induced immunoproteasome as demonstrated by Western blots and functionally a decreased presentation of Melan A(26-35); whereas NY-ESO-1(88-96) was very inefficiently presented by the same tumor cell lines, except for one that expressed high level of immunoproteasome. It was only presented when the tumor cells were first IFN-γ treated, followed by infection with recombinant vaccinia virus encoding NY-ESO-1, which dramatically increased NY-ESO-1 expression. These data indicate that the presentation of NY-ESO-1(88-96) is immunoproteasome dependent. Furthermore, a survey was conducted on multiple samples collected from HLA-B18(+) melanoma patients. Surprisingly, all the detectable responses to NY-ESO-1(88-96) from patients, including those who received NY-ESO-1 ISCOMATRIX™ vaccine were induced spontaneously. Taken together, these results imply that some epitopes can be inefficiently presented by tumor cells although the corresponding CD8(+) T cell responses are efficiently primed in vivo by DCs cross-presenting these epitopes. The potential implications for cancer vaccine strategies are further discussed.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Dendritic Cells/immunology , Epitopes/immunology , HLA-B18 Antigen/immunology , Blotting, Western , Cell Line, Tumor , Humans , Melanoma/immunology , Melanoma/pathology
15.
FASEB J ; 26(11): 4561-75, 2012 Nov.
Article in English | MEDLINE | ID: mdl-22872677

ABSTRACT

Chromatin remodeling at specific genomic loci controls lymphoid differentiation. Here, we investigated the role played in this process by Kruppel-associated box (KRAB)-associated protein 1 (KAP1), the universal cofactor of KRAB-zinc finger proteins (ZFPs), a tetrapod-restricted family of transcriptional repressors. T-cell-specific Kap1-deleted mice displayed a significant expansion of immature thymocytes, imbalances in CD4(+)/CD8(+) cell ratios, and altered responses to TCR and TGFß stimulation when compared to littermate KAP1 control mice. Transcriptome and chromatin studies revealed that KAP1 binds T-cell-specific cis-acting regulatory elements marked by the H3K9me3 repressive mark and enriched in Ikaros/NuRD complexes. Also, KAP1 directly controls the expression of several genes involved in TCR and cytokine signaling. Among these, regulation of FoxO1 seems to play a major role in this system. Likely responsible for tethering KAP1 to at least part of its genomic targets, a small number of KRAB-ZFPs are selectively expressed in T-lymphoid cells. These results reveal the so far unsuspected yet important role of KAP1-mediated epigenetic regulation in T-lymphocyte differentiation and activation.


Subject(s)
Gene Expression Regulation/physiology , Nuclear Proteins/metabolism , Repressor Proteins/metabolism , T-Lymphocytes/physiology , Animals , Binding Sites , CD4-Positive T-Lymphocytes/cytology , CD4-Positive T-Lymphocytes/physiology , CD8-Positive T-Lymphocytes/cytology , CD8-Positive T-Lymphocytes/physiology , DNA/genetics , DNA/metabolism , Epigenesis, Genetic , Mice , Mice, Knockout , Nuclear Proteins/genetics , Oligonucleotide Array Sequence Analysis , Phenotype , Phylogeny , Protein Binding , RNA/genetics , RNA/metabolism , Repressor Proteins/genetics , T-Lymphocytes/cytology , Transcriptome , Tripartite Motif-Containing Protein 28
16.
EMBO J ; 31(7): 1666-78, 2012 Apr 04.
Article in English | MEDLINE | ID: mdl-22373576

ABSTRACT

Although the T-cell receptor αδ (TCRαδ) locus harbours large libraries of variable (TRAV) and junctional (TRAJ) gene segments, according to previous studies the TCRα chain repertoire is of limited diversity due to restrictions imposed by sequential coordinate TRAV-TRAJ recombinations. By sequencing tens of millions of TCRα chain transcripts from naive mouse CD8(+) T cells, we observed a hugely diverse repertoire, comprising nearly all possible TRAV-TRAJ combinations. Our findings are not compatible with sequential coordinate gene recombination, but rather with a model in which contraction and DNA looping in the TCRαδ locus provide equal access to TRAV and TRAJ gene segments, similarly to that demonstrated for IgH gene recombination. Generation of the observed highly diverse TCRα chain repertoire necessitates deletion of failed attempts by thymic-positive selection and is essential for the formation of highly diverse TCRαß repertoires, capable of providing good protective immunity.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Gene Rearrangement, alpha-Chain T-Cell Antigen Receptor , Genes, T-Cell Receptor alpha , Recombination, Genetic/immunology , Animals , Base Sequence , Mice , Mice, Inbred C57BL , Molecular Sequence Data
17.
Int J Cancer ; 130(11): 2607-17, 2012 Jun 01.
Article in English | MEDLINE | ID: mdl-21796616

ABSTRACT

T-cells specific for foreign (e.g., viral) antigens can give rise to strong protective immune responses, whereas self/tumor antigen-specific T-cells are thought to be less powerful. However, synthetic T-cell vaccines composed of Melan-A/MART-1 peptide, CpG and IFA can induce high frequencies of tumor-specific CD8 T-cells in PBMC of melanoma patients. Here we analyzed the functionality of these T-cells directly ex vivo, by multiparameter flow cytometry. The production of multiple cytokines (IFNγ, TNFα, IL-2) and upregulation of LAMP-1 (CD107a) by tumor (Melan-A/MART-1) specific T-cells was comparable to virus (EBV-BMLF1) specific CD8 T-cells. Furthermore, phosphorylation of STAT1, STAT5 and ERK1/2, and expression of CD3 zeta chain were similar in tumor- and virus-specific T-cells, demonstrating functional signaling pathways. Interestingly, high frequencies of functionally competent T-cells were induced irrespective of patient's age or gender. Finally, CD8 T-cell function correlated with disease-free survival. However, this result is preliminary since the study was a Phase I clinical trial. We conclude that human tumor-specific CD8 T-cells can reach functional competence in vivo, encouraging further development and Phase III trials assessing the clinical efficacy of robust vaccination strategies.


Subject(s)
Antigens, Neoplasm/immunology , CD8-Positive T-Lymphocytes/immunology , Vaccination , Adult , Aged , CD3 Complex/analysis , Extracellular Signal-Regulated MAP Kinases/metabolism , Female , Humans , Immunocompetence , MART-1 Antigen/immunology , Male , Middle Aged , Phosphorylation , STAT1 Transcription Factor/metabolism , STAT5 Transcription Factor/metabolism
18.
Cancer Res ; 72(4): 887-96, 2012 Feb 15.
Article in English | MEDLINE | ID: mdl-22205715

ABSTRACT

Cytotoxic T cells that are present in tumors and capable of recognizing tumor epitopes are nevertheless generally impotent in eliciting tumor rejection. Thus, identifying the immune escape mechanisms responsible for inducing tumor-specific CD8(+) T-cell dysfunction may reveal effective strategies for immune therapy. The inhibitory receptors PD-1 and Tim-3 are known to negatively regulate CD8(+) T-cell responses directed against the well-characterized tumor antigen NY-ESO-1. Here, we report that the upregulation of the inhibitory molecule BTLA also plays a critical role in restricting NY-ESO-1-specific CD8(+) T-cell expansion and function in melanoma. BTLA-expressing PD-1(+)Tim-3(-) CD8(+) T cells represented the largest subset of NY-ESO-1-specific CD8(+) T cells in patients with melanoma. These cells were partially dysfunctional, producing less IFN-γ than BTLA(-) T cells but more IFN-γ, TNF, and interleukin-2 than the highly dysfunctional subset expressing all three receptors. Expression of BTLA did not increase with higher T-cell dysfunction or upon cognate antigen stimulation, as it does with PD-1, suggesting that BTLA upregulation occurs independently of functional exhaustion driven by high antigen load. Added with PD-1 and Tim-3 blockades, BTLA blockade enhanced the expansion, proliferation, and cytokine production of NY-ESO-1-specific CD8(+) T cells. Collectively, our findings indicate that targeting BTLA along with the PD-1 and Tim-3 pathways is critical to reverse an important mechanism of immune escape in patients with advanced melanoma.


Subject(s)
Antigens, Neoplasm/immunology , Melanoma/immunology , Programmed Cell Death 1 Receptor/metabolism , Receptors, Immunologic/metabolism , T-Lymphocytes, Cytotoxic/immunology , Antibodies, Blocking/pharmacology , Hepatitis A Virus Cellular Receptor 2 , Humans , Membrane Proteins , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Receptors, Immunologic/antagonists & inhibitors , Tumor Microenvironment , Up-Regulation
19.
J Biol Chem ; 286(48): 41723-41735, 2011 Dec 02.
Article in English | MEDLINE | ID: mdl-21990358

ABSTRACT

MHC-peptide multimers containing biotinylated MHC-peptide complexes bound to phycoerythrin (PE) streptavidin (SA) are widely used for analyzing and sorting antigen-specific T cells. Here we describe alternative T cell-staining reagents that are superior to conventional reagents. They are built on reversible chelate complexes of Ni(2+)-nitrilotriacetic acid (NTA) with oligohistidines. We synthesized biotinylated linear mono-, di-, and tetra-NTA compounds using conventional solid phase peptide chemistry and studied their interaction with HLA-A*0201-peptide complexes containing a His(6), His(12), or 2×His(6) tag by surface plasmon resonance on SA-coated sensor chips and equilibrium dialysis. The binding avidity increased in the order His(6) < His(12) < 2×His(6) and NTA(1) < NTA(2) < NTA(4), respectively, depending on the configuration of the NTA moieties and increased to picomolar K(D) for the combination of a 2×His(6) tag and a 2×Ni(2+)-NTA(2). We demonstrate that HLA-A2-2×His(6)-peptide multimers containing either Ni(2+)-NTA(4)-biotin and PE-SA- or PE-NTA(4)-stained influenza and Melan A-specific CD8+ T cells equal or better than conventional multimers. Although these complexes were highly stable, they very rapidly dissociated in the presence of imidazole, which allowed sorting of bona fide antigen-specific CD8+ T cells without inducing T cell death as well as assessment of HLA-A2-peptide monomer dissociation kinetics on CD8+ T cells.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , HLA-A2 Antigen , Histidine , Nickel/chemistry , Nitrilotriacetic Acid/chemistry , Peptides , HLA-A2 Antigen/chemistry , HLA-A2 Antigen/immunology , Histidine/chemistry , Histidine/immunology , Humans , MART-1 Antigen/chemistry , MART-1 Antigen/immunology , Nitrilotriacetic Acid/immunology , Peptides/chemical synthesis , Peptides/chemistry , Peptides/immunology , Staining and Labeling/methods
20.
J Immunol ; 186(4): 2282-90, 2011 Feb 15.
Article in English | MEDLINE | ID: mdl-21228347

ABSTRACT

The CD3ε cytoplasmic tail contains a conserved proline-rich sequence (PRS) that influences TCR-CD3 expression and signaling. Although the PRS can bind the SH3.1 domain of the cytosolic adapter Nck, whether the PRS is constitutively available for Nck binding or instead represents a cryptic motif that is exposed via conformational change upon TCR-CD3 engagement (CD3Δc) is currently unresolved. Furthermore, the extent to which a cis-acting CD3ε basic amino acid-rich stretch (BRS), with its unique phosphoinositide-binding capability, might impact PRS accessibility is not clear. In this study, we found that freshly harvested primary thymocytes expressed low to moderate basal levels of Nck-accessible PRS ("open-CD3"), although most TCR-CD3 complexes were inaccessible to Nck ("closed-CD3"). Ag presentation in vivo induced open-CD3, accounting for half of the basal level found in thymocytes from MHC(+) mice. Additional stimulation with either anti-CD3 Abs or peptide-MHC ligands further elevated open-CD3 above basal levels, consistent with a model wherein antigenic engagement induces maximum PRS exposure. We also found that the open-CD3 conformation induced by APCs outlasted the time of ligand occupancy, marking receptors that had been engaged. Finally, CD3ε BRS-phosphoinositide interactions played no role in either adoption of the initial closed-CD3 conformation or induction of open-CD3 by Ab stimulation. Thus, a basal level of open-CD3 is succeeded by a higher, induced level upon TCR-CD3 engagement, involving CD3Δc and prolonged accessibility of the CD3ε PRS to Nck.


Subject(s)
Antigen-Presenting Cells/immunology , Antigen-Presenting Cells/metabolism , CD3 Complex/metabolism , Proline/metabolism , Amino Acid Motifs/immunology , Animals , CD3 Complex/genetics , CD3 Complex/immunology , Cell Line, Tumor , Epitopes, T-Lymphocyte/physiology , Hybridomas , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Peptide Fragments/genetics , Peptide Fragments/immunology , Peptide Fragments/metabolism , Proline/immunology , Receptor-CD3 Complex, Antigen, T-Cell/genetics , Receptor-CD3 Complex, Antigen, T-Cell/immunology , Receptor-CD3 Complex, Antigen, T-Cell/metabolism , T-Lymphocytes/immunology , T-Lymphocytes/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...