Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
1.
J Immunother Cancer ; 9(1)2021 01.
Article in English | MEDLINE | ID: mdl-33500260

ABSTRACT

BACKGROUND: In contrast to immune checkpoint inhibitors, the use of antibodies as agonists of immune costimulatory receptors as cancer therapeutics has largely failed. We sought to address this problem using a new class of modular synthetic drugs, termed tumor-targeted immune cell agonists (TICAs), based on constrained bicyclic peptides (Bicycles). METHODS: Phage libraries displaying Bicycles were panned for binders against tumor necrosis factor (TNF) superfamily receptors CD137 and OX40, and tumor antigens EphA2, Nectin-4 and programmed death ligand 1. The CD137 and OX40 Bicycles were chemically conjugated to tumor antigen Bicycles with different linkers and stoichiometric ratios of binders to obtain a library of low molecular weight TICAs (MW <8 kDa). The TICAs were evaluated in a suite of in vitro and in vivo assays to characterize their pharmacology and mechanism of action. RESULTS: Linking Bicycles against costimulatory receptors (e.g., CD137) to Bicycles against tumor antigens (e.g., EphA2) created potent agonists that activated the receptors selectively in the presence of tumor cells expressing these antigens. An EphA2/CD137 TICA (BCY12491) efficiently costimulated human peripheral blood mononuclear cells in vitro in the presence of EphA2 expressing tumor cell lines as measured by the increased secretion of interferon γ and interleukin-2. Treatment of C57/Bl6 mice transgenic for the human CD137 extracellular domain (huCD137) bearing EphA2-expressing MC38 tumors with BCY12491 resulted in the infiltration of CD8+ T cells, elimination of tumors and generation of immunological memory. BCY12491 was cleared quickly from the circulation (plasma t1/2 in mice of 1-2 hr), yet intermittent dosing proved effective. CONCLUSION: Tumor target-dependent CD137 agonism using a novel chemical approach (TICAs) afforded elimination of tumors with only intermittent dosing suggesting potential for a wide therapeutic index in humans. This work unlocks a new path to effective cancer immunotherapy via agonism of TNF superfamily receptors.


Subject(s)
Neoplasms/drug therapy , Peptides, Cyclic/administration & dosage , Receptor, EphA2/agonists , Tumor Necrosis Factor Receptor Superfamily, Member 9/agonists , A549 Cells , Animals , Antigens, Neoplasm/metabolism , CD8-Positive T-Lymphocytes/metabolism , Cell Line, Tumor , Female , HT29 Cells , Humans , Jurkat Cells , Mice , Mice, Transgenic , Neoplasms/genetics , Neoplasms/immunology , PC-3 Cells , Peptide Library , Peptides, Cyclic/chemistry , Peptides, Cyclic/pharmacology , Receptors, OX40/metabolism , Xenograft Model Antitumor Assays
2.
Immunity ; 46(1): 51-64, 2017 01 17.
Article in English | MEDLINE | ID: mdl-28099864

ABSTRACT

Despite the importance of programmed cell death-1 (PD-1) in inhibiting T cell effector activity, the mechanisms regulating its expression remain poorly defined. We found that the chromatin organizer special AT-rich sequence-binding protein-1 (Satb1) restrains PD-1 expression induced upon T cell activation by recruiting a nucleosome remodeling deacetylase (NuRD) complex to Pdcd1 regulatory regions. Satb1 deficienct T cells exhibited a 40-fold increase in PD-1 expression. Tumor-derived transforming growth factor ß (Tgf-ß) decreased Satb1 expression through binding of Smad proteins to the Satb1 promoter. Smad proteins also competed with the Satb1-NuRD complex for binding to Pdcd1 enhancers, releasing Pdcd1 expression from Satb1-mediated repression, Satb1-deficient tumor-reactive T cells lost effector activity more rapidly than wild-type lymphocytes at tumor beds expressing PD-1 ligand (CD274), and these differences were abrogated by sustained CD274 blockade. Our findings suggest that Satb1 functions to prevent premature T cell exhaustion by regulating Pdcd1 expression upon T cell activation. Dysregulation of this pathway in tumor-infiltrating T cells results in diminished anti-tumor immunity.


Subject(s)
Epigenetic Repression/immunology , Gene Expression Regulation/immunology , Lymphocytes, Tumor-Infiltrating/immunology , Matrix Attachment Region Binding Proteins/biosynthesis , Programmed Cell Death 1 Receptor/biosynthesis , Animals , Enzyme-Linked Immunospot Assay , Humans , Immunoprecipitation , Lymphocyte Activation/immunology , Matrix Attachment Region Binding Proteins/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Neoplasms/immunology , Neoplasms/metabolism
3.
Cancer Res ; 76(21): 6253-6265, 2016 11 01.
Article in English | MEDLINE | ID: mdl-27803104

ABSTRACT

Targeted therapies elicit seemingly paradoxical and poorly understood effects on tumor immunity. Here, we show that the MEK inhibitor trametinib abrogates cytokine-driven expansion of monocytic myeloid-derived suppressor cells (mMDSC) from human or mouse myeloid progenitors. MEK inhibition also reduced the production of the mMDSC chemotactic factor osteopontin by tumor cells. Together, these effects reduced mMDSC accumulation in tumor-bearing hosts, limiting the outgrowth of KRas-driven breast tumors, even though trametinib largely failed to directly inhibit tumor cell proliferation. Accordingly, trametinib impeded tumor progression in vivo through a mechanism requiring CD8+ T cells, which was paradoxical given the drug's reported ability to inhibit effector lymphocytes. Confirming our observations, adoptive transfer of tumor-derived mMDSC reversed the ability of trametinib to control tumor growth. Overall, our work showed how the effects of trametinib on immune cells could partly explain its effectiveness, distinct from its activity on tumor cells themselves. More broadly, by providing a more incisive view into how MEK inhibitors may act against tumors, our findings expand their potential uses to generally block mMDSC expansion, which occurs widely in cancers to drive their growth and progression. Cancer Res; 76(21); 6253-65. ©2016 AACR.


Subject(s)
Antineoplastic Agents/pharmacology , Mutation , Myelopoiesis/drug effects , Neoplasms/drug therapy , Proto-Oncogene Proteins p21(ras)/genetics , Pyridones/pharmacology , Pyrimidinones/pharmacology , T-Lymphocytes/physiology , Animals , Cell Line, Tumor , Female , Humans , MAP Kinase Signaling System/drug effects , Mice , Mice, Inbred C57BL , Myeloid Cells/drug effects , Myeloid Cells/physiology , Neoplasms/genetics , Neoplasms/physiopathology , Osteopontin/biosynthesis
4.
Cancer Cell ; 30(1): 120-135, 2016 07 11.
Article in English | MEDLINE | ID: mdl-27374224

ABSTRACT

Based on studies in mouse tumor models, granulocytes appear to play a tumor-promoting role. However, there are limited data about the phenotype and function of tumor-associated neutrophils (TANs) in humans. Here, we identify a subset of TANs that exhibited characteristics of both neutrophils and antigen-presenting cells (APCs) in early-stage human lung cancer. These APC-like "hybrid neutrophils," which originate from CD11b(+)CD15(hi)CD10(-)CD16(low) immature progenitors, are able to cross-present antigens, as well as trigger and augment anti-tumor T cell responses. Interferon-γ and granulocyte-macrophage colony-stimulating factor are requisite factors in the tumor that, working through the Ikaros transcription factor, synergistically exert their APC-promoting effects on the progenitors. Overall, these data demonstrate the existence of a specialized TAN subset with anti-tumor capabilities in human cancer.


Subject(s)
Antigen-Presenting Cells/immunology , Lung Neoplasms/immunology , Neutrophils/immunology , Gene Expression Regulation, Neoplastic , Granulocyte-Macrophage Colony-Stimulating Factor/immunology , Humans , Ikaros Transcription Factor/metabolism , Interferon-gamma/immunology , Neoplasm Staging , Neutrophils/cytology , T-Lymphocytes, Cytotoxic/immunology
5.
Cancer Res ; 76(9): 2561-72, 2016 05 01.
Article in English | MEDLINE | ID: mdl-26980764

ABSTRACT

Many signal transduction inhibitors are being developed for cancer therapy target pathways that are also important for the proper function of antitumor lymphocytes, possibly weakening their therapeutic effects. Here we show that most inhibitors targeting multiple signaling pathways have especially strong negative effects on T-cell activation at their active doses on cancer cells. In particular, we found that recently approved MEK inhibitors displayed potent suppressive effects on T cells in vitro However, these effects could be attenuated by certain cytokines that can be administered to cancer patients. Among them, clinically available IL15 superagonists, which can activate PI3K selectively in T lymphocytes, synergized with MEK inhibitors in vivo to elicit potent and durable antitumor responses, including by a vaccine-like effect that generated resistance to tumor rechallenge. Our work identifies a clinically actionable approach to overcome the T-cell-suppressive effects of MEK inhibitors and illustrates how to reconcile the deficiencies of signal transduction inhibitors, which impede desired immunologic effects in vivo Cancer Res; 76(9); 2561-72. ©2016 AACR.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , CD8-Positive T-Lymphocytes/drug effects , Lymphocyte Activation/drug effects , Neoplasms, Experimental/pathology , Proteins/pharmacology , Animals , Blotting, Western , Cell Line, Tumor , Flow Cytometry , High-Throughput Screening Assays , Humans , Interleukin-15 , Mice , Mice, Inbred C57BL , Mice, Transgenic , Protein Kinase Inhibitors/pharmacology , Pyridones/pharmacology , Pyrimidinones/pharmacology , Recombinant Fusion Proteins , Signal Transduction/drug effects , Xenograft Model Antitumor Assays
6.
Cell Rep ; 14(7): 1774-1786, 2016 Feb 23.
Article in English | MEDLINE | ID: mdl-26876172

ABSTRACT

Special AT-rich sequence-binding protein 1 (Satb1) governs genome-wide transcriptional programs. Using a conditional knockout mouse, we find that Satb1 is required for normal differentiation of conventional dendritic cells (DCs). Furthermore, Satb1 governs the differentiation of inflammatory DCs by regulating major histocompatibility complex class II (MHC II) expression through Notch1 signaling. Mechanistically, Satb1 binds to the Notch1 promoter, activating Notch expression and driving RBPJ occupancy of the H2-Ab1 promoter, which activates MHC II transcription. However, tumor-driven, unremitting expression of Satb1 in activated Zbtb46(+) inflammatory DCs that infiltrate ovarian tumors results in an immunosuppressive phenotype characterized by increased secretion of tumor-promoting Galectin-1 and IL-6. In vivo silencing of Satb1 in tumor-associated DCs reverses their tumorigenic activity and boosts protective immunity. Therefore, dynamic fluctuations in Satb1 expression govern the generation and immunostimulatory activity of steady-state and inflammatory DCs, but continuous Satb1 overexpression in differentiated DCs converts them into tolerogenic/pro-inflammatory cells that contribute to malignant progression.


Subject(s)
Dendritic Cells/immunology , Gene Expression Regulation, Neoplastic , Histocompatibility Antigens Class II/immunology , Matrix Attachment Region Binding Proteins/immunology , Ovarian Neoplasms/immunology , Animals , Cell Differentiation , Cell Proliferation , Cell Transformation, Neoplastic , Dendritic Cells/pathology , Female , Galectin 1/genetics , Galectin 1/immunology , Histocompatibility Antigens Class II/genetics , Histones/genetics , Histones/immunology , Humans , Immune Tolerance , Immunoglobulin J Recombination Signal Sequence-Binding Protein/genetics , Immunoglobulin J Recombination Signal Sequence-Binding Protein/immunology , Interleukin-6/genetics , Interleukin-6/immunology , Matrix Attachment Region Binding Proteins/antagonists & inhibitors , Matrix Attachment Region Binding Proteins/genetics , Mice , Mice, Knockout , Neoplasm Transplantation , Ovarian Neoplasms/genetics , Ovarian Neoplasms/pathology , Promoter Regions, Genetic , Protein Binding , RNA, Small Interfering/genetics , RNA, Small Interfering/immunology , Receptor, Notch1/genetics , Receptor, Notch1/immunology , Signal Transduction , Transcription Factors/genetics , Transcription Factors/immunology
7.
Cancer Cell ; 27(1): 27-40, 2015 Jan 12.
Article in English | MEDLINE | ID: mdl-25533336

ABSTRACT

The dominant TLR5(R392X) polymorphism abrogates flagellin responses in >7% of humans. We report that TLR5-dependent commensal bacteria drive malignant progression at extramucosal locations by increasing systemic IL-6, which drives mobilization of myeloid-derived suppressor cells (MDSCs). Mechanistically, expanded granulocytic MDSCs cause γδ lymphocytes in TLR5-responsive tumors to secrete galectin-1, dampening antitumor immunity and accelerating malignant progression. In contrast, IL-17 is consistently upregulated in TLR5-unresponsive tumor-bearing mice but only accelerates malignant progression in IL-6-unresponsive tumors. Importantly, depletion of commensal bacteria abrogates TLR5-dependent differences in tumor growth. Contrasting differences in inflammatory cytokines and malignant evolution are recapitulated in TLR5-responsive/unresponsive ovarian and breast cancer patients. Therefore, inflammation, antitumor immunity, and the clinical outcome of cancer patients are influenced by a common TLR5 polymorphism.


Subject(s)
Interleukin-17/metabolism , Interleukin-6/metabolism , Microbiota , Neoplasms/immunology , Neoplasms/pathology , Toll-Like Receptor 5/genetics , Toll-Like Receptor 5/metabolism , Animals , Cell Line, Tumor , Cells, Cultured , Galectin 1/metabolism , Gene Expression Regulation, Neoplastic , Humans , Mice , Mice, Inbred C57BL , Mice, Transgenic , Molecular Sequence Data , Neoplasm Transplantation , Polymorphism, Single Nucleotide , Signal Transduction
8.
J Clin Invest ; 124(12): 5466-80, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25384214

ABSTRACT

Infiltrating inflammatory cells are highly prevalent within the tumor microenvironment and mediate many processes associated with tumor progression; however, the contribution of specific populations remains unclear. For example, the nature and function of tumor-associated neutrophils (TANs) in the cancer microenvironment is largely unknown. The goal of this study was to provide a phenotypic and functional characterization of TANs in surgically resected lung cancer patients. We found that TANs constituted 5%-25% of cells isolated from the digested human lung tumors. Compared with blood neutrophils, TANs displayed an activated phenotype (CD62L(lo)CD54(hi)) with a distinct repertoire of chemokine receptors that included CCR5, CCR7, CXCR3, and CXCR4. TANs produced substantial quantities of the proinflammatory factors MCP-1, IL-8, MIP-1α, and IL-6, as well as the antiinflammatory IL-1R antagonist. Functionally, both TANs and neutrophils isolated from distant nonmalignant lung tissue were able to stimulate T cell proliferation and IFN-γ release. Cross-talk between TANs and activated T cells led to substantial upregulation of CD54, CD86, OX40L, and 4-1BBL costimulatory molecules on the neutrophil surface, which bolstered T cell proliferation in a positive-feedback loop. Together our results demonstrate that in the earliest stages of lung cancer, TANs are not immunosuppressive, but rather stimulate T cell responses.


Subject(s)
Cell Proliferation , Lung Neoplasms/immunology , Neutrophil Activation , Neutrophils/immunology , T-Lymphocytes/immunology , Antigens, CD/immunology , Cytokines/immunology , Humans , Lung Neoplasms/pathology , Male , Neoplasm Staging , Neutrophils/pathology , Receptors, Chemokine/immunology , T-Lymphocytes/pathology
9.
Immunity ; 41(3): 427-439, 2014 Sep 18.
Article in English | MEDLINE | ID: mdl-25238097

ABSTRACT

Tumor-reactive T cells become unresponsive in advanced tumors. Here we have characterized a common mechanism of T cell unresponsiveness in cancer driven by the upregulation of the transcription factor Forkhead box protein P1 (Foxp1), which prevents CD8⁺ T cells from proliferating and upregulating Granzyme-B and interferon-γ in response to tumor antigens. Accordingly, Foxp1-deficient lymphocytes induced rejection of incurable tumors and promoted protection against tumor rechallenge. Mechanistically, Foxp1 interacted with the transcription factors Smad2 and Smad3 in preactivated CD8⁺ T cells in response to microenvironmental transforming growth factor-ß (TGF-ß), and was essential for its suppressive activity. Therefore, Smad2 and Smad3-mediated c-Myc repression requires Foxp1 expression in T cells. Furthermore, Foxp1 directly mediated TGF-ß-induced c-Jun transcriptional repression, which abrogated T cell activity. Our results unveil a fundamental mechanism of T cell unresponsiveness different from anergy or exhaustion, driven by TGF-ß signaling on tumor-associated lymphocytes undergoing Foxp1-dependent transcriptional regulation.


Subject(s)
Forkhead Transcription Factors/immunology , Neoplasms/immunology , Repressor Proteins/immunology , T-Lymphocytes, Cytotoxic/immunology , Transforming Growth Factor beta/immunology , Tumor Escape/immunology , Adoptive Transfer , Animals , Antigens, Neoplasm/immunology , CD4-Positive T-Lymphocytes/immunology , Cell Proliferation , Female , Forkhead Transcription Factors/biosynthesis , Forkhead Transcription Factors/genetics , Gene Expression Regulation , Granzymes/biosynthesis , Interferon-gamma/biosynthesis , JNK Mitogen-Activated Protein Kinases/biosynthesis , JNK Mitogen-Activated Protein Kinases/genetics , Lymphocyte Activation/immunology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Proto-Oncogene Proteins c-myc/biosynthesis , Proto-Oncogene Proteins c-myc/genetics , Repressor Proteins/biosynthesis , Repressor Proteins/genetics , Signal Transduction/immunology , Smad2 Protein/immunology , Smad3 Protein/immunology , T-Lymphocytes, Cytotoxic/transplantation , Transcription, Genetic , Transcriptional Activation , Tumor Microenvironment/immunology
10.
J Vis Exp ; (85)2014 Mar 26.
Article in English | MEDLINE | ID: mdl-24748051

ABSTRACT

Breast cancer is a heterogeneous disease involving complex cellular interactions between the developing tumor and immune system, eventually resulting in exponential tumor growth and metastasis to distal tissues and the collapse of anti-tumor immunity. Many useful animal models exist to study breast cancer, but none completely recapitulate the disease progression that occurs in humans. In order to gain a better understanding of the cellular interactions that result in the formation of latent metastasis and decreased survival, we have generated an inducible transgenic mouse model of YFP-expressing ductal carcinoma that develops after sexual maturity in immune-competent mice and is driven by consistent, endocrine-independent oncogene expression. Activation of YFP, ablation of p53, and expression of an oncogenic form of K-ras was achieved by the delivery of an adenovirus expressing Cre-recombinase into the mammary duct of sexually mature, virgin female mice. Tumors begin to appear 6 weeks after the initiation of oncogenic events. After tumors become apparent, they progress slowly for approximately two weeks before they begin to grow exponentially. After 7-8 weeks post-adenovirus injection, vasculature is observed connecting the tumor mass to distal lymph nodes, with eventual lymphovascular invasion of YFP+ tumor cells to the distal axillary lymph nodes. Infiltrating leukocyte populations are similar to those found in human breast carcinomas, including the presence of αß and γδ T cells, macrophages and MDSCs. This unique model will facilitate the study of cellular and immunological mechanisms involved in latent metastasis and dormancy in addition to being useful for designing novel immunotherapeutic interventions to treat invasive breast cancer.


Subject(s)
Disease Models, Animal , Mammary Neoplasms, Experimental/pathology , Adenoviridae/genetics , Alleles , Animals , Female , Genes, p53 , Genes, ras , Mammary Neoplasms, Experimental/genetics , Mammary Neoplasms, Experimental/immunology , Mammary Neoplasms, Experimental/virology , Mice , Mice, Inbred C57BL , Mice, Transgenic
11.
Hum Pathol ; 44(11): 2549-57, 2013 Nov.
Article in English | MEDLINE | ID: mdl-24074532

ABSTRACT

Fibroblast activation protein (FAP) has long been known to be expressed in the stroma of breast cancer. However, very little is known if the magnitude of FAP expression within the stroma may have a prognostic value and reflect the heterogeneous biology of the tumor cell. An earlier study had suggested that stromal FAP expression in breast cancer was inversely proportional to prognosis. We, therefore, hypothesized that stromal FAP expression may correlate with clinicopathologic variables and may serve as an adjunct prognostic factor in breast cancer. We evaluated the expression of FAP in a panel of breast cancer tissues (n = 52) using a combination of immunostain analyses at the tissue and single-cell level using freshly frozen or freshly digested human breast tumor samples, respectively. Our results showed that FAP expression was abundantly expressed in the stroma across all breast cancer subtypes without significant correlation with clinicopathologic factors. We further identified a subset of FAP-positive (or FAP(+)) stromal cells that also expressed CD45, a pan-leukocyte marker. Using freshly dissociated human breast tumor specimens (n = 5), we demonstrated that some of these FAP(+)CD45(+) cells were CD11b(+)CD14(+)MHC-II(+), indicating that they were likely tumor-associated macrophages (TAMs). Although FAP(+)CD45(+) cells have been demonstrated in the mouse tumor stroma, our results demonstrating that human breast TAMs expressed FAP were novel and suggested that existing and future FAP-directed therapy may have dual-therapeutic benefits targeting both stromal mesenchymal cells and immune cells such as TAMs. More work is needed to explore the role of FAP as a potential targetable molecule in breast cancer treatment.


Subject(s)
Biomarkers, Tumor/metabolism , Breast Neoplasms/metabolism , Gelatinases/metabolism , Membrane Proteins/metabolism , Serine Endopeptidases/metabolism , Actins/metabolism , Adult , Aged , Breast Neoplasms/pathology , Breast Neoplasms/therapy , Cohort Studies , Endopeptidases , Female , Flow Cytometry , Humans , Immunohistochemistry , In Situ Hybridization, Fluorescence , Leukocyte Common Antigens/metabolism , Macrophages/metabolism , Middle Aged , Prognosis , Stromal Cells/metabolism
13.
Cell Immunol ; 278(1-2): 21-6, 2012.
Article in English | MEDLINE | ID: mdl-23121972

ABSTRACT

The immune surveillance hypothesis proposed over 50 years ago that many precancerous lesions are eliminated without a histological trace due to immunological pressure. Since then, it has become apparent that both the tumor and the anti-cancer immune response evolve over a long period to allow the eventual escape of nascent precancerous lesions into full-blown tumors. Although primarily focusing on loss of antigenicity, the immunoediting hypothesis has gradually evolved to appreciate the role of active immunosuppression in tumor progression, where myeloid leukocytes are increasingly recognized as the major driving force. This review highlights recent studies implicating how myeloid cells with antigen-presenting capabilities are co-opted by tumors to promote malignant progression. Because at least some advanced tumors remain significantly immunogenic, these new studies add a tweak to the immunoediting hypothesis as well as a rationale to block immunosuppressive mechanisms as a first-line intervention in cancer patients.


Subject(s)
Dendritic Cells/immunology , Immunologic Surveillance , Myeloid Cells/immunology , Neoplasms/immunology , T-Lymphocytes/immunology , Adaptive Immunity , Antigens, Neoplasm/immunology , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/immunology , Cytokines/biosynthesis , Cytokines/immunology , Dendritic Cells/pathology , Disease Progression , Humans , Immune Tolerance , Immunity, Innate , Myeloid Cells/pathology , T-Lymphocytes/pathology , Tumor Escape
14.
Proc Natl Acad Sci U S A ; 109(19): 7415-20, 2012 May 08.
Article in English | MEDLINE | ID: mdl-22529380

ABSTRACT

Mature peripheral T cells respond to foreign but not to self-antigens. During development in the thymus, deletion of high-affinity self-reactive immature thymocytes contributes to tolerance of mature T cells. However, double-positive thymocytes are positively selected to survive if they respond to self-peptide-MHC complexes; thus, there must be mechanisms to prevent overt reactivity to those same complexes in the periphery. "Developmental tuning" is the active process through which T-cell receptor (TCR)-associated signaling pathways of single-positive (SP) thymocytes are attenuated to respond appropriately to self-peptide-MHC complexes in the periphery. We previously showed that MHC class II expression in the thymic medulla was necessary to tune CD4(+) SP (CD4 SP) thymocytes. CD4 SP thymocytes from mice lacking medullary MHC class II expression had inappropriately enhanced proximal TCR signaling to low-affinity self-ligands that was associated with altered cellular distribution of the tyrosine kinase Lck. Now, we report that activation of both tuned and untuned CD4 SP thymocytes is Lck-dependent. Untuned CD4 SP cells contain a pool of Lck with increased basal phosphorylation that is not associated with the CD4 coreceptor. Phosphorylation of this pool of Lck decreases with tuning. Immunogold transmission electron microscopy of membrane sheets permitted direct visualization of Lck. In the absence of tuning, a significant proportion of Lck and the TCR subunit CD3ζ are expressed on the same protein island; this close association of Lck and the TCR probably explains the enhanced activation of untuned CD4 SP cells. Thus, changes in membrane topography during thymic maturation determine the set point for TCR responsiveness.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Lymphocyte Activation/immunology , Lymphocyte Specific Protein Tyrosine Kinase p56(lck)/immunology , Thymus Gland/immunology , Animals , Blotting, Western , CD3 Complex/immunology , CD3 Complex/metabolism , CD4-Positive T-Lymphocytes/metabolism , Cell Differentiation/immunology , Cell Membrane/immunology , Cell Membrane/metabolism , Cell Membrane/ultrastructure , Cells, Cultured , Dasatinib , Histocompatibility Antigens Class II/genetics , Histocompatibility Antigens Class II/immunology , Histocompatibility Antigens Class II/metabolism , Intracellular Space/enzymology , Lymphocyte Activation/drug effects , Lymphocyte Specific Protein Tyrosine Kinase p56(lck)/antagonists & inhibitors , Lymphocyte Specific Protein Tyrosine Kinase p56(lck)/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Microscopy, Immunoelectron , Phosphorylation , Protein Binding , Protein Kinase Inhibitors/pharmacology , Pyrimidines/pharmacology , Receptors, Antigen, T-Cell/genetics , Receptors, Antigen, T-Cell/immunology , Receptors, Antigen, T-Cell/metabolism , Signal Transduction/drug effects , Signal Transduction/immunology , Thiazoles/pharmacology , Thymocytes/immunology , Thymocytes/metabolism , Thymus Gland/cytology , Thymus Gland/metabolism
15.
J Autoimmun ; 36(1): 56-64, 2011 Feb.
Article in English | MEDLINE | ID: mdl-21095100

ABSTRACT

Experimental autoimmune encephalomyelitis (EAE), a model for the human disease multiple sclerosis (MS), is dependent upon the activation and effector functions of autoreactive CD4 T cells. Multiple interactions between CD4 T cells and major histocompatibility class II (MHCII)+ antigen presenting cells (APCs) must occur in both the periphery and central nervous system (CNS) to elicit autoimmunity. The identity of the MHCII+ APCs involved throughout this process remains in question. We investigated which APC in the periphery and CNS mediates disease using transgenic mice with MHCII expression restricted to dendritic cells (DCs). MHCII expression restricted to DCs results in normal susceptibility to peptide-mediated EAE. Indeed, radiation-sensitive bone marrow-derived DCs were sufficient for all APC functions during peptide-induced disease. However, DCs alone were inefficient at promoting disease after immunization with the myelin protein myelin oligodendrocyte glycoprotein (MOG), even in the presence of MHCII-deficient B cells. Consistent with a defect in disease induction following protein immunization, antigen presentation by DCs alone was incapable of mediating spontaneous optic neuritis. These results indicate that DCs are capable of perpetuating CNS-targeted autoimmunity when antigens are readily available, but other APCs are required to efficiently initiate pathogenic cognate CD4 T cell responses.


Subject(s)
Antigen Presentation/immunology , Dendritic Cells/immunology , Encephalomyelitis, Autoimmune, Experimental/immunology , Animals , CD4-Positive T-Lymphocytes/immunology , Cell Separation , Encephalomyelitis, Autoimmune, Experimental/pathology , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , Histocompatibility Antigens Class II/immunology , Lymphocyte Activation/immunology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Optic Nerve/pathology , Spinal Cord/pathology
16.
Int J Microbiol ; 2010: 917075, 2010.
Article in English | MEDLINE | ID: mdl-21234388

ABSTRACT

The detection of pathogen-derived molecules as foreign particles by adaptive immune cells triggers T and B lymphocytes to mount protective cellular and humoral responses, respectively. Recent immunological advances elucidated that proteins and some lipids are the principle biological molecules that induce protective T cell responses during microbial infections. Polysaccharides are important components of microbial pathogens and many vaccines. However, research concerning the activation of the adaptive immune system by polysaccharides gained interest only recently. Traditionally, polysaccharides were considered to be T cell-independent antigens that did not directly activate T cells or induce protective immune responses. Here, we review several recent advances in "carbohydrate immunobiology". A group of bacterial polysaccharides that are known as "zwitterionic polysaccharides (ZPSs)" were recently identified as potent immune modulators. The immunomodulatory effect of ZPSs required antigen processing and presentation by antigen presenting cells, the activation of CD4 T cells and subpopulations of CD8 T cells and the modulation of host cytokine responses. In this review, we also discuss the potential use of these unique immunomodulatory ZPSs in new vaccination strategies against chronic inflammatory conditions, autoimmunity, infectious diseases, allergies and asthmatic conditions.

17.
J Immunol ; 183(9): 5554-62, 2009 Nov 01.
Article in English | MEDLINE | ID: mdl-19843939

ABSTRACT

Immature thymocytes that are positively selected based upon their response to self-peptide-MHC complexes develop into mature T cells that are not overtly reactive to those same complexes. Developmental tuning is the active process through which TCR-associated signaling pathways of single-positive thymocytes are attenuated to respond appropriately to the peptide-MHC molecules that will be encountered in the periphery. In this study, we explore the mechanisms that regulate the tuning of CD4(+) single-positive T cells to MHC class II encountered in the thymic medulla. Experiments with murine BM chimeras demonstrate that tuning can be mediated by MHC class II expressed by either thymic medullary epithelial cells or thymic dendritic cells. Tuning does not require the engagement of CD4 by MHC class II on stromal cells. Rather, it is mediated by interactions between MHC class II and the TCR. To understand the molecular changes that distinguish immature hyperactive T cells from tuned mature CD4(+) T cells, we compared their responses to TCR stimulation. The altered response of mature CD4 single-positive thymocytes is characterized by the inhibition of ERK activation by low-affinity self-ligands and increased expression of the inhibitory tyrosine phosphatase SHP-1. Thus, persistent TCR engagement by peptide-MHC class II on thymic medullary stroma inhibits reactivity to self-Ags and prevents autoreactivity in the mature repertoire.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Histocompatibility Antigens Class II/metabolism , Lymphocyte Activation/immunology , Peptide Fragments/immunology , Receptors, Antigen, T-Cell/metabolism , Thymus Gland/immunology , Thymus Gland/metabolism , Animals , CD4-Positive T-Lymphocytes/enzymology , CD4-Positive T-Lymphocytes/metabolism , Cell Differentiation/genetics , Cell Differentiation/immunology , Down-Regulation/genetics , Down-Regulation/immunology , Epithelial Cells/immunology , Epithelial Cells/metabolism , Extracellular Signal-Regulated MAP Kinases/metabolism , Histocompatibility Antigens Class II/biosynthesis , Histocompatibility Antigens Class II/genetics , Ligands , Lymphocyte Activation/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Peptide Fragments/metabolism , Protein Tyrosine Phosphatase, Non-Receptor Type 6/biosynthesis , Protein Tyrosine Phosphatase, Non-Receptor Type 6/genetics , Receptors, Antigen, T-Cell/antagonists & inhibitors , Receptors, Antigen, T-Cell/genetics , Thymus Gland/cytology , Thymus Gland/growth & development
18.
Infect Immun ; 77(9): 3705-12, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19546196

ABSTRACT

Zwitterionic polysaccharides of the normal flora bacteria represent a novel class of antigens in that they correct systemic CD4(+) T-cell deficiencies and direct lymphoid organogenesis during colonization of the host. Presentation of these polysaccharides to CD4(+) T cells depends on major histocompatibility complex class II- and DM-dependent retrograde transport from lysosomes to the cell surface. Yet the phenotype and clonality of the immune response to the polysaccharide in the mature host immune system have not been studied. Using the zwitterionic capsular polysaccharide Sp1 of Streptococcus pneumoniae, a transient member of the bacterial flora, in an experimental mouse model of cellular immunity, we demonstrated the accumulation of TH1- and TH17-polarized CD4(+) CD44(high) CD62(low) CD25(-) memory T cells. Subcutaneous immunization with Sp1 resulted in an increase of serum immunoglobulin G (IgG), predominantly of the IgG1 subclass, and suggested the presence of a humoral memory response to the polysaccharide. CD4(+) T cells stimulated with polysaccharide in vitro and in vivo showed a nonrestricted pattern for the T-cell receptor (TCR) beta-chain variable region, as demonstrated by semiquantitative reverse transcription-PCR and flow cytometry. Clonotype mapping of in vivo and in vitro polysaccharide-activated CD4(+) T cells revealed clonotypic TCR transcripts. Taken together, the data show the induction of clonal expansion of CD4(+) T cells by polysaccharides of commensal bacteria. Cellular and humoral memory host responses imply the ability of these polysaccharides to mediate the expansion of T cells via recognition within the CDR3 region of the TCR.


Subject(s)
CD4-Positive T-Lymphocytes/physiology , Immunologic Memory , Polysaccharides, Bacterial/immunology , Streptococcus pneumoniae/immunology , Abscess/etiology , Animals , Immunoglobulin G/blood , Interleukin-17/immunology , Lymphocyte Activation , Mice , Mice, Inbred C57BL , Th1 Cells/immunology
19.
Cell Biol Int ; 33(7): 778-84, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19427914

ABSTRACT

Dendritic cells (DCs) link the innate and adaptive immune system. Currently, murine DCs for cell biology investigations are developed from MHC class II-negative bone marrow (BM) precursor cells, non-depleted BM cells or BM monocytes in the presence of granulocyte-macrophage colony-stimulating factor (GM-CSF). Here we demonstrate an isolation procedure of functionally intact myeloid CD11c(+) CD11b(+) DCs derived from murine CD34-positive precursors. DCs derived from CD34(+) cells show functional internalization, maturation, cytokine secretion, MHC-restricted antigen presentation, and MHCII retrograde transport of antigens from the lysosomes to the cell surface. In comparison to the established method, the advantages of this isolation procedure are a shorter cultivation period, a superior transfection efficiency, the yield of a purer and more homogeneous population of immature DCs, and less consumption of cell culture medium and GM-CSF. The new isolation procedure and the functional quality of CD34(+) cell-derived murine myeloid DCs make them ideally suited for immunology and cell biology studies.


Subject(s)
Antigens, CD34/metabolism , Dendritic Cells/cytology , Granulocyte-Macrophage Colony-Stimulating Factor/pharmacology , Animals , Bone Marrow Cells/cytology , Bone Marrow Cells/drug effects , CD11b Antigen/metabolism , CD11c Antigen/metabolism , Cell Culture Techniques , Dendritic Cells/drug effects , Dendritic Cells/immunology , Mice , Mice, Inbred C57BL , Phenotype , Recombinant Proteins , Transfection
20.
PLoS Pathog ; 3(3): e32, 2007 Mar.
Article in English | MEDLINE | ID: mdl-17367207

ABSTRACT

Bacterial capsular polysaccharides are virulence factors and are considered T cell-independent antigens. However, the capsular polysaccharide Sp1 from Streptococcus pneumoniae serotype 1 has been shown to activate CD4(+) T cells in a major histocompatibility complex (MHC) class II-dependent manner. The mechanism of carbohydrate presentation to CD4(+) T cells is unknown. We show in live murine dendritic cells (DCs) that Sp1 translocates from lysosomal compartments to the plasma membrane in MHCII-positive tubules. Sp1 cell surface presentation results in reduction of self-peptide presentation without alteration of the MHCII self peptide repertoire. In DM-deficient mice, retrograde transport of Sp1/MHCII complexes resulting in T cell-dependent immune responses to the polysaccharide in vitro and in vivo is significantly reduced. The results demonstrate the capacity of a bacterial capsular polysaccharide antigen to use DC tubules as a vehicle for its transport as an MHCII/saccharide complex to the cell surface for the induction of T cell activation. Furthermore, retrograde transport requires the functional role of DM in self peptide-carbohydrate exchange. These observations open new opportunities for the design of vaccines against microbial encapsulated pathogens.


Subject(s)
Antigens, Bacterial/metabolism , Bacterial Capsules/metabolism , Histocompatibility Antigens Class II/physiology , Streptococcus pneumoniae/immunology , Animals , Antigen Presentation , Antigens, Differentiation, B-Lymphocyte/metabolism , Autoantigens/metabolism , Biological Transport , CD4-Positive T-Lymphocytes/immunology , Dendritic Cells/physiology , Endosomes/metabolism , HLA-D Antigens/physiology , Histocompatibility Antigens Class II/metabolism , Lymphocyte Activation , Mice , Mice, Inbred C57BL
SELECTION OF CITATIONS
SEARCH DETAIL
...