Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters










Database
Language
Publication year range
1.
Monoclon Antib Immunodiagn Immunother ; 38(6): 242-254, 2019 Dec.
Article in English | MEDLINE | ID: mdl-31825302

ABSTRACT

Although CD3 T cell redirecting antibodies have been successfully utilized for the treatment of hematological malignancies (blinatumomab), the T cell signaling pathways induced by these molecules are incompletely understood. To gain insight into the mechanism of action for T cell redirection antibodies, we created a novel murine CD3xEpCAM bispecific antibody that incorporates a silent Fc to dissect function and signaling of murine CD8 OT1 T cells upon stimulation. T cell-mediated cytotoxicity, cytokine secretion, expression of activation markers, and proliferation were directly induced in T cells treated with the novel CD3xEpCAM bispecific molecule in vitro in the presence of epithelial cell adhesion molecule (EpCAM) expressing tumor cells. Nanostring analysis showed that CD3xEpCAM induced a gene expression profile that resembled antigen-mediated activation, although the magnitude was lower than that of the antigen-induced response. In addition, this CD3xEpCAM bispecific antibody exhibited in vivo efficacy. This is the first study that investigates both in vitro and in vivo murine CD8 T cell function and signaling induced by a CD3xEpCAM antibody having a silent Fc to delineate differences between antigen-independent and antigen-specific T cell activation. These findings expand the understanding of T cell function and signaling induced by CD3 redirection bispecific antibodies and may help to develop more efficacious CD3 redirection therapeutics for cancer treatment, particularly for solid tumors.


Subject(s)
Antibodies, Bispecific/immunology , CD3 Complex/immunology , Neoplasms/immunology , T-Lymphocytes/immunology , Animals , CD3 Complex/genetics , CD8-Positive T-Lymphocytes/immunology , Cell Proliferation/genetics , Epithelial Cell Adhesion Molecule/genetics , Epithelial Cell Adhesion Molecule/immunology , Humans , Immunoglobulin Fc Fragments/immunology , Mice , Neoplasms/therapy , Signal Transduction/immunology
2.
J Immunol ; 199(12): 4091-4102, 2017 12 15.
Article in English | MEDLINE | ID: mdl-29127145

ABSTRACT

T cell expression of TIM-3 following Ag encounter has been associated with a continuum of functional states ranging from effector memory T cells to exhaustion. We have designed an in vitro culture system to specifically address the impact of anti-TIM-3/TIM-3 engagement on human Ag-specific CD8 T cells during a normal response to Ag and found that anti-TIM-3 treatment enhances T cell function. In our in vitro T cell culture system, MART1-specific CD8 T cells were expanded from healthy donors using artificial APCs. To ensure that the T cells were the only source of TIM-3, cells were rechallenged with peptide-loaded artificial APCs in the presence of anti-TIM-3 Ab. In these conditions, anti-TIM-3 treatment promotes generation of effector T cells as shown by acquisition of an activated phenotype, increased cytokine production, enhanced proliferation, and a transcription program associated with T cell differentiation. Activation of mTORC1 has been previously demonstrated to enhance CD8 T cell effector function and differentiation. Anti-TIM-3 drives CD8 T cell differentiation through activation of the mTORC1 as evidenced by increased levels of phosphorylated S6 protein and rhebl1 transcript. Altogether these findings suggest that anti-TIM-3, together with Ag, drives differentiation in favor of effector T cells via the activation of mTOR pathway. To our knowledge, this is the first report demonstrating that TIM-3 engagement during Ag stimulation directly influences T cell differentiation through mTORC1.


Subject(s)
Antigens/immunology , CD8-Positive T-Lymphocytes/immunology , Hepatitis A Virus Cellular Receptor 2/immunology , Immunologic Memory/immunology , Mechanistic Target of Rapamycin Complex 1/immunology , Antibodies, Monoclonal/pharmacology , Cell Division , Cells, Cultured , Gene Expression Profiling , Gene Expression Regulation/immunology , Humans , Lymphocyte Activation , Lymphokines/biosynthesis , Lymphokines/genetics , MART-1 Antigen/immunology , Phosphorylation , Protein Processing, Post-Translational , T-Cell Antigen Receptor Specificity , ras Proteins/biosynthesis , ras Proteins/genetics
3.
J Immunol ; 198(10): 3939-3948, 2017 05 15.
Article in English | MEDLINE | ID: mdl-28424242

ABSTRACT

CD4+ T cells lacking the mTORC1 activator Rheb fail to secrete IFN-γ under Th1 polarizing conditions. We hypothesized that this phenotype is due to defects in regulation of the canonical Th1 transcription factor T-bet at the level of protein phosphorylation downstream of mTORC1. To test this hypothesis, we employed targeted mass-spectrometry proteomic analysis-multiple reaction monitoring mass spectrometry. We used this method to detect and quantify predicted phosphopeptides derived from T-bet. By analyzing activated murine wild-type and Rheb-deficient CD4+ T cells, as well as murine CD4+ T cells activated in the presence of rapamycin, a pharmacologic inhibitor of mTORC1, we were able to identify six T-bet phosphorylation sites. Five of these are novel, and four sites are consistently dephosphorylated in both Rheb-deficient CD4+ T cells and T cells treated with rapamycin, suggesting mTORC1 signaling controls their phosphorylation. Alanine mutagenesis of each of the six phosphorylation sites was tested for the ability to impair IFN-γ expression. Single phosphorylation site mutants still support induction of IFN-γ expression; however, simultaneous mutation of three of the mTORC1-dependent sites results in significantly reduced IFN-γ expression. The reduced activity of the triple mutant T-bet is associated with its failure to recruit chromatin remodeling complexes to the Ifng gene promoter. These results establish a novel mechanism by which mTORC1 regulates Th1 differentiation, through control of T-bet phosphorylation.


Subject(s)
Multiprotein Complexes/metabolism , T-Box Domain Proteins/metabolism , TOR Serine-Threonine Kinases/metabolism , Th1 Cells/physiology , Animals , CD4-Positive T-Lymphocytes/drug effects , CD4-Positive T-Lymphocytes/immunology , Cell Differentiation , Chromatin Assembly and Disassembly , Gene Expression Regulation , Interferon-gamma/biosynthesis , Interferon-gamma/genetics , Interferon-gamma/immunology , Mass Spectrometry/methods , Mechanistic Target of Rapamycin Complex 1 , Mice , Multiprotein Complexes/antagonists & inhibitors , Multiprotein Complexes/genetics , Mutation , Phosphorylation , Proteomics/methods , Sirolimus/pharmacology , T-Box Domain Proteins/chemistry , T-Box Domain Proteins/genetics , T-Box Domain Proteins/immunology , TOR Serine-Threonine Kinases/antagonists & inhibitors , TOR Serine-Threonine Kinases/genetics , Th1 Cells/immunology , Th2 Cells/immunology
4.
Mol Cell Proteomics ; 11(11): 1457-67, 2012 Nov.
Article in English | MEDLINE | ID: mdl-22942358

ABSTRACT

The major histocompatibility complex (MHC) class II-associated Invariant chain (Ii) is present in professional antigen presenting cells where it regulates peptide loading onto MHC class II molecules and the peptidome presented to CD4+ T lymphocytes. Because Ii prevents peptide loading in neutral subcellular compartments, we reasoned that Ii- cells may present peptides not presented by Ii+ cells. Based on the hypothesis that patients are tolerant to MHC II-restricted tumor peptides presented by Ii+ cells, but will not be tolerant to novel peptides presented by Ii- cells, we generated MHC II vaccines to activate cancer patients' T cells. The vaccines are Ii- tumor cells expressing syngeneic HLA-DR and the costimulatory molecule CD80. We used liquid chromatography coupled with mass spectrometry to sequence MHC II-restricted peptides from Ii+ and Ii- MCF10 human breast cancer cells transfected with HLA-DR7 or the MHC Class II transactivator CIITA to determine if Ii- cells present novel peptides. Ii expression was induced in the HLA-DR7 transfectants by transfection of Ii, and inhibited in the CIITA transfectants by RNA interference. Peptides were analyzed and binding affinity predicted by artificial neural net analysis. HLA-DR7-restricted peptides from Ii- and Ii+ cells do not differ in size or in subcellular location of their source proteins; however, a subset of HLA-DR7-restricted peptides of Ii- cells are not presented by Ii+ cells, and are derived from source proteins not used by Ii+ cells. Peptides from Ii- cells with the highest predicted HLA-DR7 binding affinity were synthesized, and activated tumor-specific HLA-DR7+ human T cells from healthy donors and breast cancer patients, demonstrating that the MS-identified peptides are bonafide tumor antigens. These results demonstrate that Ii regulates the repertoire of tumor peptides presented by MHC class II+ breast cancer cells and identify novel immunogenic MHC II-restricted peptides that are potential therapeutic reagents for cancer patients.


Subject(s)
Antigen Presentation/immunology , Antigen-Presenting Cells/immunology , Antigens, Differentiation, B-Lymphocyte/immunology , Breast Neoplasms/immunology , Histocompatibility Antigens Class II/immunology , Lymphocyte Activation/immunology , Peptides/immunology , T-Lymphocytes/immunology , Amino Acid Motifs , Amino Acid Sequence , Antigens, Differentiation, B-Lymphocyte/chemistry , Breast Neoplasms/prevention & control , Cancer Vaccines/immunology , Cell Line, Tumor , Female , HLA-DR Antigens/immunology , Histocompatibility Antigens Class II/chemistry , Humans , Mass Spectrometry , Molecular Sequence Data , Peptides/chemistry , Phenotype , Protein Binding , Tissue Donors , Transduction, Genetic
5.
Cancer Immunol Immunother ; 61(8): 1319-25, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22546994

ABSTRACT

Immune suppressive myeloid-derived suppressor cells (MDSC) are present in most cancer patients where they inhibit innate anti-tumor immunity and are a significant obstacle to cancer immunotherapy. Inflammation is a known inducer of Gr1(+)CD11b(+) MDSC; however, the factors/conditions that regulate MDSC survival and half-life have not been identified. We have used mass spectrometry (MS) and proteomic analysis to identify proteins and pathways that regulate MDSC survival. This analysis revealed high expression of caspase family proteins and the Fas-FasL, p38 MAPK, and TGFß pathways, suggesting that Fas-FasL apoptosis regulates MDSC survival. Flow cytometry, confocal microscopy, and western blot analyses confirmed the MS findings and demonstrated that Fas(+) MDSC are susceptible to Fas-mediated killing in vitro. In vivo studies with FasL-deficient and Fas-deficient mice demonstrated that Fas-FasL interactions are essential for MDSC apoptosis and for rejection of established metastatic disease and survival and that FasL(+) T cells are the effector population mediating MDSC apoptosis. MS findings validated by biological experiments demonstrated that inflammation increases MDSC levels by protecting MDSC from Fas-mediated apoptosis, possibly by activating p38 MAPK. These results demonstrate that MDSC half-life in vivo is regulated by FasL(+) T cells and that inflammation increases MDSC levels by conferring resistance to Fas-mediated apoptosis and identifies T cells as the relevant effector cells causing MDSC apoptosis in vivo. This newly recognized mechanism for regulating MDSC levels identifies potential new targets for decreasing MDSC in cancer patients.


Subject(s)
Apoptosis/immunology , Inflammation/immunology , Myeloid Cells/immunology , Neoplasms/immunology , Animals , Blotting, Western , Cell Survival , Congresses as Topic , Fas Ligand Protein/immunology , Flow Cytometry , Mice , Microscopy, Confocal , T-Lymphocytes/immunology , fas Receptor/immunology
6.
Blood ; 117(20): 5381-90, 2011 May 19.
Article in English | MEDLINE | ID: mdl-21450901

ABSTRACT

Myeloid-derived suppressor cells (MDSCs) inhibit adaptive and innate immunity and accumulate in the blood of persons with cancer, chronic inflammation, trauma, infection, and stress. Some of the factors inducing their accumulation are known; however, mechanisms regulating their turnover have not been identified. Mass spectrometry showed prominent expression of apoptosis pathway proteins, suggesting that MDSC turnover may be regulated by Fas-FasL-mediated apoptosis. This hypothesis was confirmed by showing that blood MDSCs induced by 3 mouse tumors were Fas(+) and apoptosed in response to Fas agonist in vitro and to activated FasL(+) T cells in vivo. FasL-deficient mice contained significantly more blood MDSCs than FasL(+/+) mice, and after removal of primary tumors MDSCs regressed in STAT6(-/-) and CD1(-/-) mice but not in STAT6(-/-)FasL(-/-) or CD1(-/-)FasL(-/-) mice. Fas(+) macrophages and dendritic cells did not apoptose in response to activated T cells, indicating that Fas-FasL regulation of myeloid cells was restricted to MDSCs. These results identify a new mechanism regulating MDSC levels in vivo and show a retaliatory relationship between T cells and MDSCs in that MDSCs suppress T-cell activation; however, once activated, T cells mediate MDSC apoptosis.


Subject(s)
Fas Ligand Protein/metabolism , Myeloid Cells/cytology , Myeloid Cells/immunology , T-Lymphocytes/immunology , fas Receptor/metabolism , Adoptive Transfer , Animals , Apoptosis , Cell Line, Tumor , Fas Ligand Protein/deficiency , Fas Ligand Protein/genetics , Female , Lymphocyte Activation , Mammary Neoplasms, Experimental/immunology , Mammary Neoplasms, Experimental/metabolism , Mammary Neoplasms, Experimental/pathology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Myeloid Cells/metabolism , T-Lymphocytes/metabolism
7.
Mol Cell Proteomics ; 10(3): M110.002980, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21191032

ABSTRACT

Myeloid-derived suppressor cells (MDSC) accumulate in patients and animals with cancer where they mediate systemic immune suppression and obstruct immune-based cancer therapies. We have previously demonstrated that inflammation, which frequently accompanies tumor onset and progression, increases the rate of accumulation and the suppressive potency of MDSC. To determine how inflammation enhances MDSC levels and activity we used mass spectrometry to identify proteins produced by MDSC induced in highly inflammatory settings. Proteomic pathway analysis identified the Fas pathway and caspase network proteins, leading us to hypothesize that inflammation enhances MDSC accumulation by increasing MDSC resistance to Fas-mediated apoptosis. The MS findings were validated and extended by biological studies. Using activated caspase 3 and caspase 8 as indicators of apoptosis, flow cytometry, confocal microscopy, and Western blot analyses demonstrated that inflammation-induced MDSC treated with a Fas agonist contain lower levels of activated caspases, suggesting that inflammation enhances resistance to Fas-mediated apoptosis. Resistance to Fas-mediated apoptosis was confirmed by viability studies of MDSC treated with a Fas agonist. These results suggest that an inflammatory environment, which is frequently present in tumor-bearing individuals, protects MDSC against extrinsic-induced apoptosis resulting in MDSC with a longer in vivo half-life, and may explain why MDSC accumulate more rapidly and to higher levels in inflammatory settings.


Subject(s)
Apoptosis/immunology , Inflammation/immunology , Myeloid Cells/immunology , Proteomics/methods , Animals , Cell Separation , Inflammation/complications , Inflammation/pathology , Lymphocyte Activation/immunology , Mass Spectrometry , Mice , Mice, Inbred BALB C , Microscopy, Confocal , Neoplasms/complications , Neoplasms/immunology , Neoplasms/pathology , Proteome/chemistry , Proteome/metabolism , T-Lymphocytes/immunology , Toll-Like Receptor 4/deficiency , Toll-Like Receptor 4/metabolism , fas Receptor/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...