Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 28
Filter
Add more filters










Publication year range
1.
Cancer Discov ; 12(6): 1449-1461, 2022 06 02.
Article in English | MEDLINE | ID: mdl-35255120

ABSTRACT

Immune escape represents a major driver of acute myeloid leukemia (AML) reemergence after allogeneic hematopoietic cell transplantation (allo-HCT), with up to 40% of relapses prompted by nongenomic loss of HLA class II expression in leukemia cells. By integrative analysis of gene expression, DNA methylation, and chromatin accessibility in paired diagnosis/relapse primary samples and in the respective patient-derived xenografts (PDX), we identify the polycomb repressive complex 2 (PRC2) as a key epigenetic driver of this immune escape modality. We report that loss of expression of HLA class II molecules is accompanied by a PRC2-dependent reduction in chromatin accessibility. Pharmacologic inhibition of PRC2 subunits rescues HLA class II expression in AML relapses in vitro and in vivo, with consequent recovery of leukemia recognition by CD4+ T cells. Our results uncover a novel link between epigenetics and leukemia immune escape, which may rapidly translate into innovative strategies to cure or prevent AML posttransplantation relapse. SIGNIFICANCE: Loss of HLA class II expression represents a frequent mechanism of leukemia posttransplantation relapse. Here we identify PRC2 as the main epigenetic driver of this immune escape modality and show that its chemical inhibition can reinstate a proficient graft-versus-leukemia effect, providing an innovative rationale for personalized epigenetic immunotherapies. See related commentary by Köhler and Zeiser, p. 1410. This article is highlighted in the In This Issue feature, p. 1397.


Subject(s)
Leukemia, Myeloid, Acute , Polycomb Repressive Complex 2 , Chromatin/genetics , Chromatin/immunology , Epigenesis, Genetic , Hematopoietic Stem Cell Transplantation , Histocompatibility Antigens Class II/biosynthesis , Histocompatibility Antigens Class II/genetics , Histocompatibility Antigens Class II/immunology , Humans , Leukemia, Myeloid, Acute/immunology , Leukemia, Myeloid, Acute/therapy , Polycomb Repressive Complex 2/genetics , Polycomb Repressive Complex 2/immunology , Recurrence , Tumor Escape/genetics
2.
Sci Immunol ; 6(63): eabf7268, 2021 Sep 17.
Article in English | MEDLINE | ID: mdl-34533976

ABSTRACT

Dendritic cells (DCs) and macrophages are at the forefront of immune responses, modifying their transcriptional programs in response to their tissue environment or immunological challenge. Posttranslational modifications of histones, such as histone H3 lysine-27 trimethylation (H3K27me3) by the Polycomb repressive complex 2 (PRC2), are tightly associated with epigenetic regulation of gene expression. To explore whether H3K27me3 is involved in either the establishment or function of the mononuclear phagocyte system, we selectively deleted core components of PRC2, either EZH2 or SUZ12, in CD11c-expressing myeloid cells. Unexpectedly, EZH2 deficiency neither prevented the deposition and maintenance of H3K27me3 in DCs nor hindered DC/macrophage homeostasis. In contrast, SUZ12 deficiency markedly impaired the capacity of DCs and macrophages to maintain H3K27me3. SUZ12 ablation induced a rapid loss of the alveolar macrophage and Langerhans cell networks under both steady state and inflammatory conditions because these cells could no longer proliferate to facilitate their self-renewal. Despite the reduced H3K27me3, DC development and function were unaffected by SUZ12 ablation, suggesting that PRC2-mediated gene repression was dispensable for DC homeostasis. Thus, the role of SUZ12 highlights the fundamentally different homeostatic mechanisms used by tissue-resident myeloid cells versus DCs.


Subject(s)
Dendritic Cells/immunology , Homeostasis/immunology , Myeloid Cells/immunology , Polycomb Repressive Complex 2/immunology , Animals , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Polycomb Repressive Complex 2/deficiency
3.
Cancer Res ; 79(21): 5587-5596, 2019 Nov 01.
Article in English | MEDLINE | ID: mdl-31395608

ABSTRACT

Aberrant activity of polycomb repressive complex 2 (PRC2) is involved in a wide range of human cancer progression. The WD40 repeat-containing protein EED is a core component of PRC2 and enhances PRC2 activity through interaction with H3K27me3. In this study, we report the discovery of a class of pyrimidone compounds, represented by BR-001, as potent allosteric inhibitors of PRC2. X-ray co-crystallography showed that BR-001 directly binds EED in the H3K27me3-binding pocket. BR-001 displayed antitumor potency in vitro and in vivo. In Karpas422 and Pfeiffer xenograft mouse models, twice daily oral dosing with BR-001 resulted in robust antitumor activity. BR-001 was also efficacious in syngeneic CT26 colon tumor-bearing mice; oral dosing of 30 mg/kg of BR-001 led to 59.3% tumor growth suppression and increased frequency of effector CD8+ T-cell infiltrates in tumors. Pharmacodynamic analysis revealed that CXCL10 was highly upregulated, suggesting that CXCL10 triggers the trafficking of CD8+ T cells toward tumor sites. Our results demonstrate for the first time that inhibition of EED modulates the tumor immune microenvironment to induce regression of colon tumors and therefore has the potential to be used in combination with immune-oncology therapy. SIGNIFICANCE: BR-001, a potent inhibitor of the EED subunit of the PRC2 complex, suppresses tumor progression by modulating the tumor microenvironment.


Subject(s)
Antineoplastic Agents/pharmacology , Colonic Neoplasms/immunology , Colonic Neoplasms/therapy , Polycomb Repressive Complex 2/antagonists & inhibitors , Animals , CD8-Positive T-Lymphocytes/drug effects , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Chemokine CXCL10/immunology , Chemokine CXCL10/metabolism , Colonic Neoplasms/metabolism , Disease Models, Animal , Disease Progression , Female , Heterografts/immunology , Heterografts/metabolism , Histones/immunology , Histones/metabolism , Humans , Mice , Mice, Inbred NOD , Mice, SCID , Polycomb Repressive Complex 2/immunology , Polycomb Repressive Complex 2/metabolism , Pyrimidinones/pharmacology , Tumor Microenvironment/drug effects , Tumor Microenvironment/immunology , Up-Regulation/drug effects , Up-Regulation/immunology
4.
JCI Insight ; 4(10)2019 05 16.
Article in English | MEDLINE | ID: mdl-31092733

ABSTRACT

Strategies that intervene with the development of immune-mediated diseases are urgently needed, as current treatments mostly focus on alleviating symptoms rather than reversing the disease. Targeting enzymes involved in epigenetic modifications to chromatin represents an alternative strategy that has the potential to perturb the function of the lymphocytes that drive the immune response. Here, we report that 2 major epigenetic silencing pathways are increased after T cell activation. By specific inactivation of these molecules in the T cell compartment in vivo, we demonstrate that the polycomb repressive complex 2 (PRC2) is essential for the generation of allergic responses. Furthermore, we show that small-molecule inhibition of the PRC2 methyltransferase, enhancer of zeste homolog 2 (Ezh2), reduces allergic inflammation in mice. Therefore, by systematically surveying the pathways involved in epigenetic gene silencing we have identified Ezh2 as a target for the suppression of allergic disease.


Subject(s)
Inflammation/immunology , Polycomb Repressive Complex 2/immunology , Animals , Enhancer of Zeste Homolog 2 Protein/genetics , Enhancer of Zeste Homolog 2 Protein/immunology , Epigenesis, Genetic , Gene Silencing , Inflammation/genetics , Lung/immunology , Lung/pathology , Lymphocyte Activation , Mice , Mice, Inbred C57BL , T-Lymphocytes/immunology
5.
Nat Commun ; 10(1): 2157, 2019 05 14.
Article in English | MEDLINE | ID: mdl-31089138

ABSTRACT

T cell senescence and exhaustion are major barriers to successful cancer immunotherapy. Here we show that miR-155 increases CD8+ T cell antitumor function by restraining T cell senescence and functional exhaustion through epigenetic silencing of drivers of terminal differentiation. miR-155 enhances Polycomb repressor complex 2 (PRC2) activity indirectly by promoting the expression of the PRC2-associated factor Phf19 through downregulation of the Akt inhibitor, Ship1. Phf19 orchestrates a transcriptional program extensively shared with miR-155 to restrain T cell senescence and sustain CD8+ T cell antitumor responses. These effects rely on Phf19 histone-binding capacity, which is critical for the recruitment of PRC2 to the target chromatin. These findings establish the miR-155-Phf19-PRC2 as a pivotal axis regulating CD8+ T cell differentiation, thereby paving new ways for potentiating cancer immunotherapy through epigenetic reprogramming of CD8+ T cell fate.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Melanoma, Experimental/immunology , MicroRNAs/metabolism , Skin Neoplasms/immunology , Transcription Factors/metabolism , Adoptive Transfer/methods , Animals , CD8-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/transplantation , Cell Differentiation/genetics , Cell Differentiation/immunology , Cellular Senescence/genetics , Cellular Senescence/immunology , Epigenesis, Genetic/immunology , Female , Gene Expression Regulation, Neoplastic , Humans , Melanoma, Experimental/genetics , Melanoma, Experimental/therapy , Mice , Mice, Inbred C57BL , Mice, Knockout , Phosphatidylinositol-3,4,5-Trisphosphate 5-Phosphatases/genetics , Phosphatidylinositol-3,4,5-Trisphosphate 5-Phosphatases/metabolism , Polycomb Repressive Complex 2/immunology , Polycomb Repressive Complex 2/metabolism , Skin Neoplasms/genetics , Skin Neoplasms/therapy , Transcription Factors/genetics , Transcription Factors/immunology
6.
J Mol Biol ; 431(9): 1792-1804, 2019 04 19.
Article in English | MEDLINE | ID: mdl-30660620

ABSTRACT

Microglia, the resident macrophages of the central nervous system, critically influence neural function during development and in adulthood. Microglia are also profoundly sensitive to insults to the brain to which they respond with process of activation that includes spectrum of changes in morphology, function, and gene expression. Ataxias are a class of neurodegenerative diseases characterized by motor discoordination and predominant cerebellar involvement. In case of inherited forms of ataxia, mutant proteins are expressed throughout the brain and it is unclear why cerebellum is particularly vulnerable. Recent studies demonstrated that cerebellar microglia have a uniquely hyper-vigilant immune phenotype compared to microglia from other brain regions. These findings may indicate that microglia actively contribute to cerebellar vulnerability in ataxias. Here we review current knowledge about cerebellar microglia, their activation, and their role in the pathogenesis of ataxias. In addition, we briefly review advantages and disadvantages of several experimental approaches available to study microglia.


Subject(s)
Ataxia/pathology , Cerebellum/pathology , Microglia/pathology , Phagocytes/pathology , Adult , Animals , Ataxia/genetics , Ataxia/immunology , Cerebellum/immunology , Disease Models, Animal , Gene Expression Regulation , Humans , Induced Pluripotent Stem Cells/immunology , Induced Pluripotent Stem Cells/pathology , Microglia/immunology , NF-kappa B/genetics , NF-kappa B/immunology , Organ Specificity , Phagocytes/immunology , Polycomb Repressive Complex 2/genetics , Polycomb Repressive Complex 2/immunology , Potassium Channels, Tandem Pore Domain/genetics , Potassium Channels, Tandem Pore Domain/immunology , Signal Transduction
7.
Immunity ; 46(4): 596-608, 2017 04 18.
Article in English | MEDLINE | ID: mdl-28410989

ABSTRACT

Understanding immunological memory formation depends on elucidating how multipotent memory precursor (MP) cells maintain developmental plasticity and longevity to provide long-term immunity while other effector cells develop into terminally differentiated effector (TE) cells with limited survival. Profiling active (H3K27ac) and repressed (H3K27me3) chromatin in naive, MP, and TE CD8+ T cells during viral infection revealed increased H3K27me3 deposition at numerous pro-memory and pro-survival genes in TE relative to MP cells, indicative of fate restriction, but permissive chromatin at both pro-memory and pro-effector genes in MP cells, indicative of multipotency. Polycomb repressive complex 2 deficiency impaired clonal expansion and TE cell differentiation, but minimally impacted CD8+ memory T cell maturation. Abundant H3K27me3 deposition at pro-memory genes occurred late during TE cell development, probably from diminished transcription factor FOXO1 expression. These results outline a temporal model for loss of memory cell potential through selective epigenetic silencing of pro-memory genes in effector T cells.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Cell Differentiation/immunology , Chromatin/immunology , Polycomb Repressive Complex 2/immunology , Animals , CD8-Positive T-Lymphocytes/metabolism , Cell Differentiation/genetics , Chromatin/genetics , Chromatin/metabolism , Enhancer of Zeste Homolog 2 Protein/genetics , Enhancer of Zeste Homolog 2 Protein/immunology , Enhancer of Zeste Homolog 2 Protein/metabolism , Flow Cytometry , Forkhead Box Protein O1/genetics , Forkhead Box Protein O1/immunology , Forkhead Box Protein O1/metabolism , Gene Expression/immunology , Histones/immunology , Histones/metabolism , Immunoblotting , Immunologic Memory/genetics , Immunologic Memory/immunology , Lysine/immunology , Lysine/metabolism , Methylation , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Models, Immunological , Multipotent Stem Cells/immunology , Multipotent Stem Cells/metabolism , Polycomb Repressive Complex 2/genetics , Polycomb Repressive Complex 2/metabolism , Reverse Transcriptase Polymerase Chain Reaction
8.
Nat Immunol ; 17(1): 95-103, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26523864

ABSTRACT

Aerobic glycolysis regulates T cell function. However, whether and how primary cancer alters T cell glycolytic metabolism and affects tumor immunity in cancer patients remains a question. Here we found that ovarian cancers imposed glucose restriction on T cells and dampened their function via maintaining high expression of microRNAs miR-101 and miR-26a, which constrained expression of the methyltransferase EZH2. EZH2 activated the Notch pathway by suppressing Notch repressors Numb and Fbxw7 via trimethylation of histone H3 at Lys27 and, consequently, stimulated T cell polyfunctional cytokine expression and promoted their survival via Bcl-2 signaling. Moreover, small hairpin RNA-mediated knockdown of human EZH2 in T cells elicited poor antitumor immunity. EZH2(+)CD8(+) T cells were associated with improved survival in patients. Together, these data unveil a metabolic target and mechanism of cancer immune evasion.


Subject(s)
Gene Expression Regulation, Neoplastic/immunology , MicroRNAs , Neoplasms/immunology , Polycomb Repressive Complex 2/immunology , T-Lymphocytes/immunology , Tumor Escape/immunology , Animals , Cell Separation , Chromatin Immunoprecipitation , Enhancer of Zeste Homolog 2 Protein , Female , Flow Cytometry , Fluorescent Antibody Technique , Glycolysis , Humans , Immunoblotting , Melanoma, Experimental/immunology , Mice, Inbred C57BL , Ovarian Neoplasms/immunology , Real-Time Polymerase Chain Reaction , Tissue Array Analysis , Transfection
9.
Cancer Res ; 76(2): 275-82, 2016 Jan 15.
Article in English | MEDLINE | ID: mdl-26567139

ABSTRACT

Infiltration of tumors with effector T cells is positively associated with therapeutic efficacy and patient survival. However, the mechanisms underlying effector T-cell trafficking to the tumor microenvironment remain poorly understood in patients with colon cancer. The polycomb repressive complex 2 (PRC2) is involved in cancer progression, but the regulation of tumor immunity by epigenetic mechanisms has yet to be investigated. In this study, we examined the relationship between the repressive PRC2 machinery and effector T-cell trafficking. We found that PRC2 components and demethylase JMJD3-mediated histone H3 lysine 27 trimethylation (H3K27me3) repress the expression and subsequent production of Th1-type chemokines CXCL9 and CXCL10, mediators of effector T-cell trafficking. Moreover, the expression levels of PRC2 components, including EZH2, SUZ12, and EED, were inversely associated with those of CD4, CD8, and Th1-type chemokines in human colon cancer tissue, and this expression pattern was significantly associated with patient survival. Collectively, our findings reveal that PRC2-mediated epigenetic silencing is not only a crucial oncogenic mechanism, but also a key circuit controlling tumor immunosuppression. Therefore, targeting epigenetic programs may have significant implications for improving the efficacy of current cancer immunotherapies relying on effective T-cell-mediated immunity at the tumor site.


Subject(s)
Chemokines/immunology , Colonic Neoplasms/genetics , Polycomb Repressive Complex 2/genetics , T-Lymphocytes/immunology , Th1 Cells/immunology , Cell Movement/genetics , Cell Movement/immunology , Cell Proliferation/genetics , Colonic Neoplasms/immunology , Colonic Neoplasms/pathology , Epigenesis, Genetic , Humans , Polycomb Repressive Complex 2/immunology , T-Lymphocytes/metabolism , Th1 Cells/metabolism , Transfection
11.
Nat Commun ; 6: 8823, 2015 Nov 09.
Article in English | MEDLINE | ID: mdl-26549758

ABSTRACT

T-cell development is accompanied by epigenetic changes that ensure the silencing of stem cell-related genes and the activation of lymphocyte-specific programmes. How transcription factors influence these changes remains unclear. We show that the Ikaros transcription factor forms a complex with Polycomb repressive complex 2 (PRC2) in CD4(-)CD8(-) thymocytes and allows its binding to more than 500 developmentally regulated loci, including those normally activated in haematopoietic stem cells and others induced by the Notch pathway. Loss of Ikaros in CD4(-)CD8(-) cells leads to reduced histone H3 lysine 27 trimethylation and ectopic gene expression. Furthermore, Ikaros binding triggers PRC2 recruitment and Ikaros interacts with PRC2 independently of the nucleosome remodelling and deacetylation complex. Our results identify Ikaros as a fundamental regulator of PRC2 function in developing T cells.


Subject(s)
Gene Expression Regulation, Developmental , Ikaros Transcription Factor/genetics , Polycomb Repressive Complex 2/genetics , T-Lymphocytes/immunology , Thymocytes/immunology , Animals , Blotting, Western , Cell Differentiation/genetics , Cell Differentiation/immunology , Chromatin Immunoprecipitation , Ectopic Gene Expression , Epigenesis, Genetic , Gene Expression Profiling , Gene Silencing , Histone Code/genetics , Histones/metabolism , Ikaros Transcription Factor/immunology , Methylation , Mice , Nucleosomes , Polycomb Repressive Complex 2/immunology
12.
Int J Biochem Cell Biol ; 67: 148-57, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26193001

ABSTRACT

Gene expression is epigenetically regulated through DNA methylation and covalent chromatin modifications, such as acetylation, phosphorylation, ubiquitination, sumoylation, and methylation of histones. Histone methylation state is dynamically regulated by different groups of histone methyltransferases and demethylases. The trimethylation of histone 3 (H3K4) at lysine 4 is usually associated with the activation of gene expression, whereas trimethylation of histone 3 at lysine 27 (H3K27) is associated with the repression of gene expression. The polycomb repressive complex contains the H3K27 methyltransferase Ezh2 and controls dimethylation and trimethylation of H3K27 (H3K27me2/3). The Jumonji domain containing-3 (Jmjd3, KDM6B) and ubiquitously transcribed X-chromosome tetratricopeptide repeat protein (UTX, KDM6A) have been identified as H3K27 demethylases that catalyze the demethylation of H3K27me2/3. The role and mechanisms of both JMJD3 and UTX have been extensively studied for their involvement in development, cell plasticity, immune system, neurodegenerative disease, and cancer. In this review, we will focus on recent progresses made on understanding JMJD3 in the regulation of gene expression in development and diseases. This article is part of a Directed Issue entitled: Epigenetics dynamics in development and disease.


Subject(s)
Epigenesis, Genetic , Histone Demethylases/genetics , Jumonji Domain-Containing Histone Demethylases/genetics , Neoplasms/genetics , Neurodegenerative Diseases/genetics , Nuclear Proteins/genetics , Polycomb Repressive Complex 2/genetics , Animals , Chromatin/chemistry , Chromatin/metabolism , Enhancer of Zeste Homolog 2 Protein , Gene Expression Regulation, Developmental , Germ Layers/cytology , Germ Layers/growth & development , Germ Layers/metabolism , Histone Demethylases/immunology , Histones/genetics , Histones/metabolism , Humans , Immunity, Innate , Jumonji Domain-Containing Histone Demethylases/immunology , Lysine/metabolism , Mice , Neoplasms/immunology , Neoplasms/pathology , Neurodegenerative Diseases/immunology , Neurodegenerative Diseases/pathology , Nuclear Proteins/immunology , Polycomb Repressive Complex 2/immunology
13.
Sci Rep ; 5: 10643, 2015 Jun 19.
Article in English | MEDLINE | ID: mdl-26090605

ABSTRACT

The roles of EZH2 in various subsets of CD4(+) T cells are controversial and its mechanisms of action are incompletely understood. FOXP3-positive Treg cells are a critical helper T cell subset, and dysregulation of Treg generation or function results in systemic autoimmunity. FOXP3 associates with EZH2 to mediate gene repression and suppressive function. Herein, we demonstrate that deletion of Ezh2 in CD4 T cells resulted in reduced numbers of Treg cells in vivo and differentiation in vitro and an increased proportion of memory CD4 T cells in part due to exaggerated production of effector cytokines. Furthermore, we found that both Ezh2-deficient Treg cells and T effector cells were functionally impaired in vivo: Tregs failed to constrain autoimmune colitis and T effector cells neither provided a protective response to T. gondii infection nor mediated autoimmune colitis. The dichotomous function of EZH2 in regulating differentiation and senescence in effector and regulatory T cells helps to explain the apparent existing contradictions in literature.


Subject(s)
Cell Differentiation/immunology , Cytokines/metabolism , Gene Expression Regulation/immunology , Polycomb Repressive Complex 2/immunology , T-Lymphocytes, Regulatory/immunology , Animals , Autoimmune Diseases/genetics , Autoimmune Diseases/immunology , Cell Differentiation/genetics , Colitis/genetics , Colitis/immunology , Cytokines/genetics , Enhancer of Zeste Homolog 2 Protein , Mice , Mice, Knockout , Polycomb Repressive Complex 2/genetics
14.
Int Immunopharmacol ; 26(1): 133-8, 2015 May.
Article in English | MEDLINE | ID: mdl-25819666

ABSTRACT

Analyses on reactivity of anti-cancer cytotoxic T lymphocytes (CTLs) and clinical application of peptide-based anti-cancer vaccine have been mainly focused on patients with HLA-A2 or -A24 alleles. In this study, we identified an enhancer of zeste homolog (EZH) 2-derived peptide applicable for anti-cancer vaccine for prostate cancer patients with HLA-A3 supertype alleles. Five EZH2-derived peptides that were prepared based on the binding motif to the HLA-A3 supertype alleles (HLA-A11, -A31, and -A33) were functionally screened for their potential to induce peptide-specific CTLs from peripheral blood mononuclear cells (PBMCs) of HLA-A3 supertype allele(+) prostate cancer patients. As a result, EZH2733-741 peptide was found to efficiently induce peptide-specific CTLs. The EZH2733-741 peptide-stimulated and purified CD8(+) T cells from PBMCs of HLA-A3 supertype allele(+) prostate cancer patients showed higher cytotoxicity against HLA-A3 supertype allele-expressing LNCaP prostate cancer cells than against parental LNCaP cells. This cytotoxicity against HLA-A3 supertype allele-expressing LNCaP cells was partially but significantly inhibited by the addition of EZH2733-741 peptide-pulsed competitive cells. These results indicate that the EZH2733-741 peptide could be a promising candidate for peptide-based immunotherapy for HLA-A3 supertype allele(+) prostate cancer patients.


Subject(s)
Cancer Vaccines/therapeutic use , HLA-A3 Antigen/immunology , Peptide Fragments/therapeutic use , Polycomb Repressive Complex 2/immunology , Prostatic Neoplasms/immunology , T-Lymphocytes, Cytotoxic/immunology , Alleles , Cancer Vaccines/chemistry , Cancer Vaccines/immunology , Cell Line, Tumor , Cell Survival/drug effects , Enhancer of Zeste Homolog 2 Protein , Flow Cytometry , HLA-A3 Antigen/genetics , Humans , Leukocytes, Mononuclear/immunology , Male , Peptide Fragments/chemistry , Peptide Fragments/immunology , Prostatic Neoplasms/pathology , T-Lymphocytes, Cytotoxic/drug effects , Transfection
15.
J Immunol ; 194(6): 2838-46, 2015 Mar 15.
Article in English | MEDLINE | ID: mdl-25687760

ABSTRACT

Histone modifications play critical roles in the regulation of gene expression; however, their roles in the regulation of the innate response remain to be fully investigated. Using transcriptome analysis of mouse immature dendritic cells (DCs) and LPS-induced mature DCs, we identified that Ezh1 was the most upregulated histone methyltransferase during DC maturation. In this study, we investigated the role of Ezh1 in regulating the innate immune response. We found that silencing of Ezh1 significantly suppressed TLR-triggered production of cytokines, including IL-6, TNF-α, and IFN-ß, in DCs and macrophages. Accordingly, TLR-activated signaling pathways were impaired in Ezh1-silenced macrophages. By transcriptome analysis of Ezh1-silenced macrophages, we found that Toll-interacting protein (Tollip), one well-known negative regulator of TLR signaling, was upregulated. Silencing of Tollip rescued TLR-triggered cytokine production in Ezh1-silenced macrophages. The SET domain of Ezh1 is essential for its enhancing effect on the TLR-triggered innate immune response and downstream signaling, indicating that Ezh1 promotes a TLR-triggered innate response through its lysine methyltransferase activity. Finally, Ezh1 was found to suppress the transcription of Tollip by directly targeting the proximal promoter of tollip and maintaining the high level of trimethylation of histone H3 lysine 27 there. Therefore, Ezh1 promotes TLR-triggered inflammatory cytokine production by suppressing the TLR negative regulator Tollip, contributing to full activation of the innate immune response against invading pathogens.


Subject(s)
Cytokines/immunology , Intracellular Signaling Peptides and Proteins/immunology , Polycomb Repressive Complex 2/immunology , Toll-Like Receptors/immunology , Animals , Blotting, Western , Cells, Cultured , Cytokines/genetics , Cytokines/metabolism , Dendritic Cells/immunology , Dendritic Cells/metabolism , Histones/immunology , Histones/metabolism , Immunity, Innate/genetics , Immunity, Innate/immunology , Inflammation Mediators/immunology , Inflammation Mediators/metabolism , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Lipopolysaccharides/immunology , Lipopolysaccharides/pharmacology , Lysine/immunology , Lysine/metabolism , Macrophages/immunology , Macrophages/metabolism , Methylation , Mice, Inbred C57BL , Oligodeoxyribonucleotides/immunology , Oligodeoxyribonucleotides/pharmacology , Poly I-C/immunology , Poly I-C/pharmacology , Polycomb Repressive Complex 2/genetics , Polycomb Repressive Complex 2/metabolism , Promoter Regions, Genetic/genetics , Promoter Regions, Genetic/immunology , Protein Binding/immunology , RNA Interference , Reverse Transcriptase Polymerase Chain Reaction , Toll-Like Receptors/agonists , Toll-Like Receptors/metabolism , Transcriptome/immunology
16.
Immunity ; 42(2): 201-203, 2015 Feb 17.
Article in English | MEDLINE | ID: mdl-25692693

ABSTRACT

Epigenetic modulation is critical for regulating the development and function of T cells. In this issue of Immunity, DuPage et al. (2015) show that the chromatin-modifying enzyme Ezh2 induced by CD28 costimulation is essential for regulatory T (Treg) cell maintenance during activation and differentiation.


Subject(s)
CD28 Antigens/immunology , Lymphocyte Activation/immunology , Polycomb Repressive Complex 2/immunology , T-Lymphocytes, Regulatory/immunology , Animals , Enhancer of Zeste Homolog 2 Protein , Female
17.
Immunity ; 42(2): 227-238, 2015 Feb 17.
Article in English | MEDLINE | ID: mdl-25680271

ABSTRACT

Regulatory T cells (Treg cells) are required for immune homeostasis. Chromatin remodeling is essential for establishing diverse cellular identities, but how the epigenetic program in Treg cells is maintained throughout the dynamic activation process remains unclear. Here we have shown that CD28 co-stimulation, an extracellular cue intrinsically required for Treg cell maintenance, induced the chromatin-modifying enzyme, Ezh2. Treg-specific ablation of Ezh2 resulted in spontaneous autoimmunity with reduced Foxp3(+) cells in non-lymphoid tissues and impaired resolution of experimental autoimmune encephalomyelitis. Utilizing a model designed to selectively deplete wild-type Treg cells in adult mice co-populated with Ezh2-deficient Treg cells, Ezh2-deficient cells were destabilized and failed to prevent autoimmunity. After activation, the transcriptome of Ezh2-deficient Treg cells was disrupted, with altered expression of Treg cell lineage genes in a pattern similar to Foxp3-deficient Treg cells. These studies reveal a critical role for Ezh2 in the maintenance of Treg cell identity during cellular activation.


Subject(s)
CD28 Antigens/immunology , Lymphocyte Activation/immunology , Polycomb Repressive Complex 2/immunology , T-Lymphocytes, Regulatory/immunology , Animals , Autoimmunity/genetics , Autoimmunity/immunology , CD8-Positive T-Lymphocytes/immunology , Chromatin Assembly and Disassembly , Encephalomyelitis, Autoimmune, Experimental/immunology , Enhancer of Zeste Homolog 2 Protein , Female , Forkhead Transcription Factors/biosynthesis , Forkhead Transcription Factors/genetics , Gene Expression Regulation , Heparin-binding EGF-like Growth Factor/genetics , Immune Tolerance/genetics , Immune Tolerance/immunology , Lymphocyte Depletion , Mice , Mice, Inbred C57BL , Mice, Transgenic , Polycomb Repressive Complex 2/genetics , Promoter Regions, Genetic/genetics , T-Lymphocytes, Regulatory/cytology
18.
Immunity ; 40(6): 855-6, 2014 Jun 19.
Article in English | MEDLINE | ID: mdl-24950205

ABSTRACT

In this issue of Immunity, Escobar et al. (2014) bring microRNAs and chromatin together by showing how activation-induced miR-155 targets the chromatin protein Jarid2 to regulate proinflammatory cytokine production in T helper 17 cells.


Subject(s)
Cytokines/genetics , Gene Expression Regulation , MicroRNAs/metabolism , Polycomb Repressive Complex 2/immunology , Th17 Cells/immunology , Animals , Humans , Polycomb Repressive Complex 2/metabolism
19.
Blood ; 124(5): 737-49, 2014 Jul 31.
Article in English | MEDLINE | ID: mdl-24951427

ABSTRACT

Differentiation of naïve CD4(+) T cells into effector (Th1, Th2, and Th17) and induced regulatory (iTreg) T cells requires lineage-specifying transcription factors and epigenetic modifications that allow appropriate repression or activation of gene transcription. The epigenetic silencing of cytokine genes is associated with the repressive H3K27 trimethylation mark, mediated by the Ezh2 or Ezh1 methyltransferase components of the polycomb repressive complex 2 (PRC2). Here we show that silencing of the Ifng, Gata3, and Il10 loci in naïve CD4(+) T cells is dependent on Ezh2. Naïve CD4(+) T cells lacking Ezh2 were epigenetically primed for overproduction of IFN-γ in Th2 and iTreg and IL-10 in Th2 cells. In addition, deficiency of Ezh2 accelerated effector Th cell death via death receptor-mediated extrinsic and intrinsic apoptotic pathways, confirmed in vivo for Ezh2-null IFN-γ-producing CD4(+) and CD8(+) T cells responding to Listeria monocytogenes infection. These findings demonstrate the key role of PRC2/Ezh2 in differentiation and survival of peripheral T cells and reveal potential immunotherapeutic targets.


Subject(s)
Apoptosis/immunology , Cell Differentiation/immunology , Gene Silencing/immunology , Polycomb Repressive Complex 2/immunology , T-Lymphocytes, Helper-Inducer/immunology , Animals , Cell Survival/immunology , Enhancer of Zeste Homolog 2 Protein , Female , Humans , Interferon-gamma/immunology , Interleukin-10/immunology , Listeria monocytogenes/immunology , Listeriosis/immunology , Listeriosis/pathology , Male , Mice , T-Lymphocytes, Helper-Inducer/cytology
20.
Immunity ; 40(6): 865-79, 2014 Jun 19.
Article in English | MEDLINE | ID: mdl-24856900

ABSTRACT

Specification of the T helper 17 (Th17) cell lineage requires a well-defined set of transcription factors, but how these integrate with posttranscriptional and epigenetic programs to regulate gene expression is poorly understood. Here we found defective Th17 cell cytokine expression in miR-155-deficient CD4+ T cells in vitro and in vivo. Mir155 was bound by Th17 cell transcription factors and was highly expressed during Th17 cell differentiation. miR-155-deficient Th17 and T regulatory (Treg) cells expressed increased amounts of Jarid2, a DNA-binding protein that recruits the Polycomb Repressive Complex 2 (PRC2) to chromatin. PRC2 binding to chromatin and H3K27 histone methylation was increased in miR-155-deficient cells, coinciding with failure to express Il22, Il10, Il9, and Atf3. Defects in Th17 cell cytokine expression and Treg cell homeostasis in the absence of Mir155 could be partially suppressed by Jarid2 deletion. Thus, miR-155 contributes to Th17 cell function by suppressing the inhibitory effects of Jarid2.


Subject(s)
Cytokines/genetics , Gene Expression Regulation , MicroRNAs/metabolism , Polycomb Repressive Complex 2/immunology , Th17 Cells/immunology , Activating Transcription Factor 3/genetics , Activating Transcription Factor 3/metabolism , Animals , Cell Differentiation/immunology , Cells, Cultured , Chromatin/genetics , Humans , Mice , Mice, Inbred C57BL , Mice, Knockout , MicroRNAs/genetics , Polycomb Repressive Complex 2/genetics , Polycomb Repressive Complex 2/metabolism , Protein Binding , Signal Transduction/genetics , Signal Transduction/immunology , T-Lymphocytes, Regulatory/immunology , Th1 Cells/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...