Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Publication year range
1.
J Med Chem ; 63(18): 10433-10459, 2020 09 24.
Article in English | MEDLINE | ID: mdl-32865411

ABSTRACT

The adenosinergic pathway represents an attractive new therapeutic approach in cancer immunotherapy. In this pathway, ecto-5-nucleotidase CD73 has the unique function of regulating production of immunosuppressive adenosine (ADO) through the hydrolysis of AMP. CD73 is overexpressed in many cancers, resulting in elevated levels of ADO that correspond to poor patient prognosis. Therefore, reducing the level of ADO via inhibition of CD73 is a potential strategy for treating cancers. Based on the binding mode of adenosine 5'-(α,ß-methylene)diphosphate (AOPCP) with human CD73, we designed a series of novel monophosphonate small-molecule CD73 inhibitors. Among them, OP-5244 (35) proved to be a highly potent and orally bioavailable CD73 inhibitor. In preclinical studies, 35 completely inhibited ADO production in both human cancer cells and CD8+ T cells. Furthermore, 35 lowered the ratio of ADO/AMP significantly and reversed immunosuppression in mouse models, indicating its potential as an in vivo tool compound for further development.


Subject(s)
5'-Nucleotidase/antagonists & inhibitors , Adenosine/antagonists & inhibitors , Enzyme Inhibitors/pharmacology , Immunologic Factors/pharmacology , Nucleosides/pharmacology , Organophosphonates/pharmacology , Administration, Oral , Animals , CD8-Positive T-Lymphocytes/drug effects , Cell Line, Tumor , Dogs , Enzyme Inhibitors/administration & dosage , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/pharmacokinetics , GPI-Linked Proteins/antagonists & inhibitors , Humans , Immunologic Factors/administration & dosage , Immunologic Factors/chemical synthesis , Immunologic Factors/pharmacokinetics , Macaca fascicularis , Mice, Inbred BALB C , Molecular Structure , Nucleosides/administration & dosage , Nucleosides/chemical synthesis , Nucleosides/pharmacokinetics , Organophosphonates/administration & dosage , Organophosphonates/chemical synthesis , Organophosphonates/pharmacokinetics , Rats , Stereoisomerism , Structure-Activity Relationship
2.
J Immunol ; 204(11): 2877-2886, 2020 06 01.
Article in English | MEDLINE | ID: mdl-32269095

ABSTRACT

Central tolerance prevents autoimmunity, but also limits T cell responses to potentially immunodominant tumor epitopes with limited expression in healthy tissues. In peripheral APCs, γ-IFN-inducible lysosomal thiol reductase (GILT) is critical for MHC class II-restricted presentation of disulfide bond-containing proteins, including the self-antigen and melanoma Ag tyrosinase-related protein 1 (TRP1). The role of GILT in thymic Ag processing and generation of central tolerance has not been investigated. We found that GILT enhanced the negative selection of TRP1-specific thymocytes in mice. GILT expression was enriched in thymic APCs capable of mediating deletion, namely medullary thymic epithelial cells (mTECs) and dendritic cells, whereas TRP1 expression was restricted solely to mTECs. GILT facilitated MHC class II-restricted presentation of endogenous TRP1 by pooled thymic APCs. Using bone marrow chimeras, GILT expression in thymic epithelial cells (TECs), but not hematopoietic cells, was sufficient for complete deletion of TRP1-specific thymocytes. An increased frequency of TRP1-specific regulatory T (Treg) cells was present in chimeras with increased deletion of TRP1-specific thymocytes. Only chimeras that lacked GILT in both TECs and hematopoietic cells had a high conventional T/Treg cell ratio and were protected from melanoma challenge. Thus, GILT expression in thymic APCs, and mTECs in particular, preferentially facilitates MHC class II-restricted presentation, negative selection, and increased Treg cells, resulting in a diminished antitumor response to a tissue-restricted, melanoma-associated self-antigen.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Epithelial Cells/metabolism , Membrane Glycoproteins/metabolism , Neoplasms/immunology , Oxidoreductases Acting on Sulfur Group Donors/metabolism , Oxidoreductases/metabolism , T-Lymphocytes, Regulatory/immunology , Thymocytes/immunology , Thymus Gland/immunology , Animals , Antigen Presentation , Autoantigens/metabolism , Cells, Cultured , Central Tolerance , Clonal Selection, Antigen-Mediated , Epithelial Cells/immunology , Histocompatibility Antigens Class II/metabolism , Humans , Mice , Mice, Inbred C57BL , Mice, Knockout , Organ Specificity , Oxidoreductases Acting on Sulfur Group Donors/genetics
3.
Nature ; 559(7715): 627-631, 2018 07.
Article in English | MEDLINE | ID: mdl-30022164

ABSTRACT

The thymus is responsible for generating a diverse yet self-tolerant pool of T cells1. Although the thymic medulla consists mostly of developing and mature AIRE+ epithelial cells, recent evidence has suggested that there is far greater heterogeneity among medullary thymic epithelial cells than was previously thought2. Here we describe in detail an epithelial subset that is remarkably similar to peripheral tuft cells that are found at mucosal barriers3. Similar to the periphery, thymic tuft cells express the canonical taste transduction pathway and IL-25. However, they are unique in their spatial association with cornified aggregates, ability to present antigens and expression of a broad diversity of taste receptors. Some thymic tuft cells pass through an Aire-expressing stage and depend on a known AIRE-binding partner, HIPK2, for their development. Notably, the taste chemosensory protein TRPM5 is required for their thymic function through which they support the development and polarization of thymic invariant natural killer T cells and act to establish a medullary microenvironment that is enriched in the type 2 cytokine, IL-4. These findings indicate that there is a compartmentalized medullary environment in which differentiation of a minor and highly specialized epithelial subset has a non-redundant role in shaping thymic function.


Subject(s)
Epithelial Cells/cytology , Epithelial Cells/metabolism , Interleukin-4/metabolism , Thymocytes/cytology , Thymus Gland/cytology , Thymus Gland/metabolism , Animals , Cellular Microenvironment , Doublecortin-Like Kinases , Female , Humans , Immune Tolerance/immunology , Interleukin-4/biosynthesis , Interleukins/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Protein Serine-Threonine Kinases/metabolism , TRPM Cation Channels/metabolism , Thymocytes/metabolism , Thymus Gland/anatomy & histology , Transcription Factors/deficiency , Transcription Factors/genetics , AIRE Protein
4.
Cancer Res ; 76(13): 3684-9, 2016 07 01.
Article in English | MEDLINE | ID: mdl-27197182

ABSTRACT

ICOS is a T-cell coregulatory receptor that provides a costimulatory signal to T cells during antigen-mediated activation. Antitumor immunity can be improved by ICOS-targeting therapies, but their mechanism of action remains unclear. Here, we define the role of ICOS signaling in antitumor immunity using a blocking, nondepleting antibody against ICOS ligand (ICOS-L). ICOS signaling provided critical support for the effector function of CD4(+) Foxp3(-) T cells during anti-OX40-driven tumor immune responses. By itself, ICOS-L blockade reduced accumulation of intratumoral T regulatory cells (Treg), but it was insufficient to substantially inhibit tumor growth. Furthermore, it did not impede antitumor responses mediated by anti-4-1BB-driven CD8(+) T cells. We found that anti-OX40 efficacy, which is based on Treg depletion and to a large degree on CD4(+) effector T cell (Teff) responses, was impaired with ICOS-L blockade. In contrast, the provision of additional ICOS signaling through direct ICOS-L expression by tumor cells enhanced tumor rejection and survival when administered along with anti-OX40 therapy. Taken together, our results showed that ICOS signaling during antitumor responses acts on both Teff and Treg cells, which have opposing roles in promoting immune activation. Thus, effective therapies targeting the ICOS pathway should seek to promote ICOS signaling specifically in effector CD4(+) T cells by combining ICOS agonism and Treg depletion. Cancer Res; 76(13); 3684-9. ©2016 AACR.


Subject(s)
Adenocarcinoma/immunology , CD8-Positive T-Lymphocytes/immunology , Colorectal Neoplasms/immunology , Receptors, OX40/metabolism , T-Lymphocytes, Regulatory/immunology , Adenocarcinoma/metabolism , Adenocarcinoma/prevention & control , Animals , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/prevention & control , Cytokines/metabolism , Female , Flow Cytometry , Humans , Inducible T-Cell Co-Stimulator Ligand/metabolism , Lymphocyte Activation , Mice , Mice, Inbred BALB C , Mice, Nude , Receptors, Antigen, T-Cell , Signal Transduction , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
5.
Am J Pathol ; 184(6): 1695-705, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24736166

ABSTRACT

Development of novel strategies to treat noninfectious posterior uveitis is an ongoing challenge, in part because of limited availability of animal models that mimic the naturally occurring disease in humans. Mice deficient in the autoimmune regulatory gene Aire develop a spontaneous T-cell and macrophage-mediated autoimmune uveitis that closely recapitulates human endogenous uveitis and thus provide a useful model for mechanistic and therapeutic investigations. Lymphocytic and mononuclear infiltration of the retina in Aire knockout (KO) mice triggers the onset of uveitis from initial retinal inflammation to eventual destruction of the neuroretina with loss of photoreceptors. The C-C chemokine receptor type 2 protein (CCR2) functions in directing monocyte and macrophage migration to inflamed tissues via interaction with monocyte chemotactic proteins. Using the Aire KO mouse model, we demonstrated an essential role for CCR2 in the pathogenesis of autoimmune-mediated uveitis. Loss of functional CCR2 effectively reduced immune cell infiltration and rescued the retina from destruction. CCR2-dependent migration of bone marrow-derived cells provided the driving force for retinal inflammation, with CCR2-expressing mononuclear cells contributing to retinal damage via recruitment of CD4(+) T cells. These studies identify the CCR2 pathway as a promising therapeutic target that may prove an effective approach to treat uveitis associated with autoimmunity.


Subject(s)
Autoimmune Diseases/immunology , CD4-Positive T-Lymphocytes/immunology , Cell Movement/immunology , Receptors, CCR2/immunology , Retina/immunology , Uveitis/immunology , Animals , Autoimmune Diseases/genetics , Autoimmune Diseases/pathology , CD4-Positive T-Lymphocytes/pathology , Cell Movement/genetics , Humans , Macrophages/immunology , Macrophages/pathology , Mice , Mice, Inbred BALB C , Mice, Knockout , Monocytes/immunology , Monocytes/pathology , Receptors, CCR2/genetics , Retina/pathology , Uveitis/genetics , Uveitis/pathology
6.
Nat Immunol ; 15(3): 258-65, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24464130

ABSTRACT

The maintenance of immunological tolerance requires the deletion of self-reactive T cells in the thymus. The expression of genes encoding tissue-specific antigens (TSAs) by thymic epithelial cells is critical for this process and depends on activity of the transcriptional regulator Aire; however, the molecular mechanisms Aire uses to target loci encoding TSAs are unknown. Here we identified two Aire-interacting proteins known to be involved in gene repression, ATF7ip and MBD1, that were required for Aire's targeting of loci encoding TSAs. Moreover, Mbd1(-/-) mice developed pathological autoimmunity and had a defect in Aire-dependent thymic expression of genes encoding TSAs, which underscores the importance of Aire's interaction with the ATF7ip-MBD1 protein complex in maintaining central tolerance.


Subject(s)
Central Tolerance/immunology , DNA-Binding Proteins/immunology , Gene Expression Regulation/immunology , Immune Tolerance , Repressor Proteins/immunology , Transcription Factors/immunology , Animals , Autoantigens/immunology , Central Tolerance/genetics , DNA-Binding Proteins/genetics , Flow Cytometry , HEK293 Cells , Humans , Immunoblotting , Immunoprecipitation , Mice , Mice, Inbred C57BL , Mice, Knockout , Oligonucleotide Array Sequence Analysis , Protein Binding , Repressor Proteins/genetics , Reverse Transcriptase Polymerase Chain Reaction , Transcription Factors/genetics , Transfection , Two-Hybrid System Techniques , AIRE Protein
7.
Cell Rep ; 5(1): 166-79, 2013 Oct 17.
Article in English | MEDLINE | ID: mdl-24095736

ABSTRACT

Thymic epithelial cells in the medulla (mTECs) play a critical role in enforcing central tolerance through expression and presentation of tissue-specific antigens (TSAs) and deletion of autoreactive thymocytes. TSA expression requires autoimmune regulator (Aire), a transcriptional activator present in a subset of mTECs characterized by high CD80 and major histocompatibility complex II expression and a lack of potential for differentiation or proliferation. Here, using an Aire-DTR transgenic line, we show that short-term ablation specifically targets Aire(+) mTECs, which quickly undergo RANK-dependent recovery. Repeated ablation also affects Aire(-) mTECs, and using an inducible Aire-Cre fate-mapping system, we find that this results from the loss of a subset of mTECs that showed prior expression of Aire, maintains intermediate TSA expression, and preferentially migrates toward the center of the medulla. These results clearly identify a distinct stage of mTEC development and underscore the diversity of mTECs that play a key role in maintaining tolerance.


Subject(s)
Epithelial Cells/cytology , Epithelial Cells/metabolism , Thymocytes/cytology , Thymocytes/metabolism , Thymus Gland/cytology , Thymus Gland/metabolism , Animals , Cell Differentiation/physiology , Female , Humans , Mice , Mice, Transgenic , Signal Transduction
8.
Sci Transl Med ; 5(206): 206ra139, 2013 Oct 09.
Article in English | MEDLINE | ID: mdl-24107778

ABSTRACT

Interstitial lung disease (ILD) is a complex and heterogeneous disorder that is often associated with autoimmune syndromes. Despite the connection between ILD and autoimmunity, it remains unclear whether ILD can develop from an autoimmune response that specifically targets the lung parenchyma. We examined a severe form of autoimmune disease, autoimmune polyglandular syndrome type 1 (APS1), and established a strong link between an autoimmune response to the lung-specific protein BPIFB1 (bactericidal/permeability-increasing fold-containing B1) and clinical ILD. Screening of a large cohort of APS1 patients revealed autoantibodies to BPIFB1 in 9.6% of APS1 subjects overall and in 100% of APS1 subjects with ILD. Further investigation of ILD outside the APS1 disorder revealed BPIFB1 autoantibodies present in 14.6% of patients with connective tissue disease-associated ILD and in 12.0% of patients with idiopathic ILD. The animal model for APS1, Aire⁻/⁻ mice, harbors autoantibodies to a similar lung antigen (BPIFB9); these autoantibodies are a marker for ILD. We found that a defect in thymic tolerance was responsible for the production of BPIFB9 autoantibodies and the development of ILD. We also found that immunoreactivity targeting BPIFB1 independent of a defect in Aire also led to ILD, consistent with our discovery of BPIFB1 autoantibodies in non-APS1 patients. Overall, our results demonstrate that autoimmunity targeting the lung-specific antigen BPIFB1 may contribute to the pathogenesis of ILD in patients with APS1 and in subsets of patients with non-APS1 ILD, demonstrating the role of lung-specific autoimmunity in the genesis of ILD.


Subject(s)
Autoantigens/immunology , Carrier Proteins/metabolism , Glycoproteins/metabolism , Lung Diseases, Interstitial/immunology , Lung Diseases, Interstitial/pathology , Lung/immunology , Lung/pathology , Proteins/metabolism , Adoptive Transfer , Animals , Autoantibodies/immunology , Autoantigens/metabolism , Autoimmunity/immunology , Biomarkers/metabolism , CD4-Positive T-Lymphocytes/immunology , Fatty Acid-Binding Proteins , Genotype , Humans , Immune Tolerance/immunology , Mice , Organ Specificity , Polyendocrinopathies, Autoimmune/immunology , Radioligand Assay , Reproducibility of Results , Thymus Gland/immunology , Thymus Gland/transplantation , Transcription Factors/deficiency , Transcription Factors/genetics , Transcription Factors/metabolism , AIRE Protein
9.
Immunity ; 39(3): 560-72, 2013 Sep 19.
Article in English | MEDLINE | ID: mdl-23993652

ABSTRACT

The autoimmune regulator (Aire) is essential for prevention of autoimmunity; its role is best understood in the thymus, where it promotes self-tolerance through tissue-specific antigen (TSA) expression. Recently, extrathymic Aire-expressing cells (eTACs) have been described in murine secondary lymphoid organs, but the identity of such cells and their role in immune tolerance remains unclear. Here we have shown that eTACs are a discrete major histocompatibility complex class II (MHC II)(hi), CD80(lo), CD86(lo), epithelial cell adhesion molecule (EpCAM)(hi), CD45(lo) bone marrow-derived peripheral antigen-presenting cell (APC) population. We also have demonstrated that eTACs can functionally inactivate CD4⁺ T cells through a mechanism that does not require regulatory T cells (Treg) and is resistant to innate inflammatory stimuli. Together, these findings further define eTACs as a distinct tolerogenic cell population in secondary lymphoid organs.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Self Tolerance , Transcription Factors/metabolism , Adoptive Transfer , Animals , Antigen Presentation , Antigen-Presenting Cells/metabolism , Antigens, Neoplasm/metabolism , Autoimmunity , B7-1 Antigen/metabolism , B7-2 Antigen/metabolism , Bone Marrow Cells , Cell Adhesion Molecules/metabolism , Epithelial Cell Adhesion Molecule , Histocompatibility Antigens Class II/immunology , Leukocyte Common Antigens/metabolism , Mice , Mice, Inbred NOD , Transcription Factors/biosynthesis , AIRE Protein
10.
Cell Stem Cell ; 13(2): 219-29, 2013 Aug 01.
Article in English | MEDLINE | ID: mdl-23684540

ABSTRACT

Inducing immune tolerance to prevent rejection is a key step toward successful engraftment of stem-cell-derived tissue in a clinical setting. Using human pluripotent stem cells to generate thymic epithelial cells (TECs) capable of supporting T cell development represents a promising approach to reach this goal; however, progress toward generating functional TECs has been limited. Here, we describe a robust in vitro method to direct differentiation of human embryonic stem cells (hESCs) into thymic epithelial progenitors (TEPs) by precise regulation of TGFß, BMP4, RA, Wnt, Shh, and FGF signaling. The hESC-derived TEPs further mature into functional TECs that support T cell development upon transplantation into thymus-deficient mice. Importantly, the engrafted TEPs produce T cells capable of in vitro proliferation as well as in vivo immune responses. Thus, hESC-derived TEP grafts may have broad applications for enhancing engraftment in cell-based therapies as well as restoring age- and stress-related thymic decline.


Subject(s)
Embryonic Stem Cells/cytology , Epithelium/growth & development , T-Lymphocytes/cytology , Thymus Gland/growth & development , Animals , Cell Differentiation/immunology , Epithelial Cells/cytology , Epithelial Cells/immunology , Epithelium/metabolism , Epithelium/transplantation , Humans , Immunity , Mice , Mice, Inbred C57BL , Mice, Nude , Models, Biological , Stem Cell Transplantation , T-Lymphocytes/immunology , Thymus Gland/cytology , Thymus Gland/immunology
11.
Immunol Rev ; 241(1): 89-103, 2011 May.
Article in English | MEDLINE | ID: mdl-21488892

ABSTRACT

The negative selection of self-reactive thymocytes depends on the expression of tissue-specific antigens by medullary thymic epithelial cells. The autoimmune regulator (Aire) protein plays an important role in turning on these antigens, and the absence of even one Aire-induced tissue-specific antigen in the thymus can lead to autoimmunity in the antigen-expressing target organ. Recently, Aire protein has been detected in peripheral lymphoid organs, suggesting that peripheral Aire plays a complementary role here. In these peripheral sites, Aire was found to regulate the expression of a group of tissue-specific antigens that is distinct from those expressed in the thymus. Furthermore, transgenic antigen expression in extrathymic Aire-expressing cells (eTACs) can mediate deletional tolerance, but the immunological relevance of Aire-dependent, endogenous tissue-specific antigens remains to be determined.


Subject(s)
Autoantigens/immunology , Immune System/immunology , Immune Tolerance , Thymus Gland/immunology , Transcription Factors/immunology , Animals , Autoimmunity , Clonal Deletion , Humans , Immune System/embryology , Immune System/growth & development , Mice , Mice, Transgenic , Organ Specificity , AIRE Protein
12.
J Clin Invest ; 121(4): 1251-3, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21436581

ABSTRACT

Myocarditis, or inflammation of the heart, is a potentially devastating disease that can result from both viral infection and autoimmune attack of self antigens in the heart. In the current issue of the JCI, Lv and colleagues use a genetically susceptible mouse model to show that myocarditis is a T cell-mediated autoimmune disease that occurs due to insufficient thymic negative selection of α-myosin-reactive T cells.


Subject(s)
Immune Tolerance , Myocarditis/immunology , Animals , Disease Models, Animal , HLA-DQ Antigens/genetics , Mice , Mice, Inbred NOD , Mice, Transgenic , Myocarditis/etiology , Myocarditis/prevention & control , T-Lymphocytes/immunology , Ventricular Myosins/immunology
SELECTION OF CITATIONS
SEARCH DETAIL