Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 238
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Annu Rev Immunol ; 42(1): 455-488, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38360546

ABSTRACT

Ten-eleven translocation (TET) proteins are iron-dependent and α-ketoglutarate-dependent dioxygenases that sequentially oxidize the methyl group of 5-methylcytosine (5mC) to 5-hydroxymethylcytosine (5hmC), 5-formylcytosine (5fC) and 5-carboxylcytosine (5caC). All three epigenetic modifications are intermediates in DNA demethylation. TET proteins are recruited by transcription factors and by RNA polymerase II to modify 5mC at enhancers and gene bodies, thereby regulating gene expression during development, cell lineage specification, and cell activation. It is not yet clear, however, how the established biochemical activities of TET enzymes in oxidizing 5mC and mediating DNA demethylation relate to the known association of TET deficiency with inflammation, clonal hematopoiesis, and cancer. There are hints that the ability of TET deficiency to promote cell proliferation in a signal-dependent manner may be harnessed for cancer immunotherapy. In this review, we draw upon recent findings in cells of the immune system to illustrate established as well as emerging ideas of how TET proteins influence cellular function.


Subject(s)
DNA Demethylation , Dioxygenases , Immunotherapy , Inflammation , Neoplasms , Humans , Neoplasms/therapy , Neoplasms/immunology , Neoplasms/etiology , Neoplasms/metabolism , Animals , Inflammation/metabolism , Inflammation/immunology , Immunotherapy/methods , Dioxygenases/metabolism , Immune System/metabolism , Immune System/immunology , Epigenesis, Genetic , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins/genetics , DNA Methylation , DNA-Binding Proteins/metabolism , DNA-Binding Proteins/genetics , Mixed Function Oxygenases/metabolism , Mixed Function Oxygenases/genetics
2.
Nat Immunol ; 23(1): 99-108, 2022 01.
Article in English | MEDLINE | ID: mdl-34937926

ABSTRACT

Enzymes of the TET family are methylcytosine dioxygenases that undergo frequent mutational or functional inactivation in human cancers. Recurrent loss-of-function mutations in TET proteins are frequent in human diffuse large B cell lymphoma (DLBCL). Here, we investigate the role of TET proteins in B cell homeostasis and development of B cell lymphomas with features of DLBCL. We show that deletion of Tet2 and Tet3 genes in mature B cells in mice perturbs B cell homeostasis and results in spontaneous development of germinal center (GC)-derived B cell lymphomas with increased G-quadruplexes and R-loops. At a genome-wide level, G-quadruplexes and R-loops were associated with increased DNA double-strand breaks (DSBs) at immunoglobulin switch regions. Deletion of the DNA methyltransferase DNMT1 in TET-deficient B cells prevented expansion of GC B cells, diminished the accumulation of G-quadruplexes and R-loops and delayed B lymphoma development, consistent with the opposing functions of DNMT and TET enzymes in DNA methylation and demethylation. Clustered regularly interspaced short palindromic repeats (CRISPR)-mediated depletion of nucleases and helicases that regulate G-quadruplexes and R-loops decreased the viability of TET-deficient B cells. Our studies suggest a molecular mechanism by which TET loss of function might predispose to the development of B cell malignancies.


Subject(s)
B-Lymphocytes/immunology , Carcinogenesis/immunology , DNA-Binding Proteins/deficiency , DNA-Binding Proteins/immunology , Dioxygenases/immunology , Homeostasis/immunology , R-Loop Structures/immunology , Animals , Cell Differentiation/immunology , DNA Methylation/immunology , G-Quadruplexes , Germinal Center/immunology , Mice , Mice, Inbred C57BL
3.
Nat Immunol ; 22(8): 983-995, 2021 08.
Article in English | MEDLINE | ID: mdl-34282330

ABSTRACT

The transcription factors nuclear factor of activated T cells (NFAT) and activator protein 1 (AP-1; Fos-Jun) cooperate to promote the effector functions of T cells, but NFAT in the absence of AP-1 imposes a negative feedback program of T cell hyporesponsiveness (exhaustion). Here, we show that basic leucine zipper ATF-like transcription factor (BATF) and interferon regulatory factor 4 (IRF4) cooperate to counter T cell exhaustion in mouse tumor models. Overexpression of BATF in CD8+ T cells expressing a chimeric antigen receptor (CAR) promoted the survival and expansion of tumor-infiltrating CAR T cells, increased the production of effector cytokines, decreased the expression of inhibitory receptors and the exhaustion-associated transcription factor TOX and supported the generation of long-lived memory T cells that controlled tumor recurrence. These responses were dependent on BATF-IRF interaction, since cells expressing a BATF variant unable to interact with IRF4 did not survive in tumors and did not effectively delay tumor growth. BATF may improve the antitumor responses of CAR T cells by skewing their phenotypes and transcriptional profiles away from exhaustion and towards increased effector function.


Subject(s)
Basic-Leucine Zipper Transcription Factors/metabolism , CD8-Positive T-Lymphocytes/immunology , Interferon Regulatory Factors/metabolism , Lymphocytes, Tumor-Infiltrating/immunology , Neoplasms/immunology , Receptors, Chimeric Antigen/immunology , Animals , Cell Line, Tumor , Disease Models, Animal , Female , Humans , Male , Melanoma, Experimental , Mice , Mice, Inbred C57BL , Mice, Transgenic , NFATC Transcription Factors/metabolism , Neoplasm Recurrence, Local/immunology , Transcription Factor AP-1/metabolism
4.
Cell ; 175(6): 1701-1715.e16, 2018 11 29.
Article in English | MEDLINE | ID: mdl-30449622

ABSTRACT

While many genetic variants have been associated with risk for human diseases, how these variants affect gene expression in various cell types remains largely unknown. To address this gap, the DICE (database of immune cell expression, expression quantitative trait loci [eQTLs], and epigenomics) project was established. Considering all human immune cell types and conditions studied, we identified cis-eQTLs for a total of 12,254 unique genes, which represent 61% of all protein-coding genes expressed in these cell types. Strikingly, a large fraction (41%) of these genes showed a strong cis-association with genotype only in a single cell type. We also found that biological sex is associated with major differences in immune cell gene expression in a highly cell-specific manner. These datasets will help reveal the effects of disease risk-associated genetic polymorphisms on specific immune cell types, providing mechanistic insights into how they might influence pathogenesis (https://dice-database.org).


Subject(s)
Gene Expression Regulation/immunology , Genotype , Polymorphism, Single Nucleotide/immunology , Quantitative Trait Loci/immunology , Sex Characteristics , Adolescent , Adult , Female , Gene Expression Profiling , Genome-Wide Association Study , Humans , Male , Middle Aged
5.
Annu Rev Immunol ; 28: 491-533, 2010.
Article in English | MEDLINE | ID: mdl-20307213

ABSTRACT

Ca(2+) entry into cells of the peripheral immune system occurs through highly Ca(2+)-selective channels known as CRAC (calcium release-activated calcium) channels. CRAC channels are a very well-characterized example of store-operated Ca(2+) channels, so designated because they open when the endoplasmic reticulum (ER) Ca(2+) store becomes depleted. Physiologically, Ca(2+) is released from the ER lumen into the cytoplasm when activated receptors couple to phospholipase C and trigger production of the second messenger inositol 1,4,5-trisphosphate (IP(3)). IP(3) binds to IP(3) receptors in the ER membrane and activates Ca(2+) release. The proteins STIM and ORAI were discovered through limited and genome-wide RNAi screens, respectively, performed in Drosophila cells and focused on identifying modulators of store-operated Ca(2+) entry. STIM1 and STIM2 sense the depletion of ER Ca(2+) stores, whereas ORAI1 is a pore subunit of the CRAC channel. In this review, we discuss selected aspects of Ca(2+) signaling in cells of the immune system, focusing on the roles of STIM and ORAI proteins in store-operated Ca(2+) entry.


Subject(s)
Calcium Channels/immunology , Calcium Channels/metabolism , Calcium Signaling , Lymphocytes/immunology , Lymphocytes/metabolism , Membrane Proteins/immunology , Membrane Proteins/metabolism , Animals , Calcium Channels/chemistry , Humans , Lymphocytes/chemistry , Membrane Proteins/chemistry , Protein Transport
6.
Genes Dev ; 36(7-8): 433-450, 2022 04 01.
Article in English | MEDLINE | ID: mdl-35450882

ABSTRACT

Somatic hypermutation (SHM) produces point mutations in immunoglobulin (Ig) genes in B cells when uracils created by the activation-induced deaminase are processed in a mutagenic manner by enzymes of the base excision repair (BER) and mismatch repair (MMR) pathways. Such uracil processing creates DNA strand breaks and is susceptible to the generation of deleterious deletions. Here, we demonstrate that the DNA repair factor HMCES strongly suppresses deletions without significantly affecting other parameters of SHM in mouse and human B cells, thereby facilitating the production of antigen-specific antibodies. The deletion-prone repair pathway suppressed by HMCES operates downstream from the uracil glycosylase UNG and is mediated by the combined action of BER factor APE2 and MMR factors MSH2, MSH6, and EXO1. HMCES's ability to shield against deletions during SHM requires its capacity to form covalent cross-links with abasic sites, in sharp contrast to its DNA end-joining role in class switch recombination but analogous to its genome-stabilizing role during DNA replication. Our findings lead to a novel model for the protection of Ig gene integrity during SHM in which abasic site cross-linking by HMCES intercedes at a critical juncture during processing of vulnerable gapped DNA intermediates by BER and MMR enzymes.


Subject(s)
Genes, Immunoglobulin , Somatic Hypermutation, Immunoglobulin , Animals , Cytidine Deaminase/genetics , Cytidine Deaminase/metabolism , DNA/genetics , DNA-Binding Proteins , Genes, Immunoglobulin/genetics , Immunoglobulin Class Switching/genetics , Mice , Somatic Hypermutation, Immunoglobulin/genetics , Uracil
7.
Nat Immunol ; 18(1): 45-53, 2017 01.
Article in English | MEDLINE | ID: mdl-27869820

ABSTRACT

TET proteins oxidize 5-methylcytosine in DNA to 5-hydroxymethylcytosine and other oxidation products. We found that simultaneous deletion of Tet2 and Tet3 in mouse CD4+CD8+ double-positive thymocytes resulted in dysregulated development and proliferation of invariant natural killer T cells (iNKT cells). Tet2-Tet3 double-knockout (DKO) iNKT cells displayed pronounced skewing toward the NKT17 lineage, with increased DNA methylation and impaired expression of genes encoding the key lineage-specifying factors T-bet and ThPOK. Transfer of purified Tet2-Tet3 DKO iNKT cells into immunocompetent recipient mice resulted in an uncontrolled expansion that was dependent on the nonclassical major histocompatibility complex (MHC) protein CD1d, which presents lipid antigens to iNKT cells. Our data indicate that TET proteins regulate iNKT cell fate by ensuring their proper development and maturation and by suppressing aberrant proliferation mediated by the T cell antigen receptor (TCR).


Subject(s)
Cell Differentiation , DNA-Binding Proteins/metabolism , Natural Killer T-Cells/physiology , Precursor Cells, T-Lymphoid/physiology , Proto-Oncogene Proteins/metabolism , Animals , Antigens, CD1d/genetics , Antigens, CD1d/metabolism , CD4 Antigens/metabolism , CD8 Antigens/metabolism , Cell Lineage , Cell Proliferation , Cells, Cultured , DNA Methylation/genetics , DNA-Binding Proteins/genetics , Dioxygenases , Mice , Mice, Inbred C57BL , Mice, Knockout , Proto-Oncogene Proteins/genetics , Receptors, Antigen, T-Cell/metabolism , T-Box Domain Proteins/genetics , T-Box Domain Proteins/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism
8.
Nat Immunol ; 18(7): 791-799, 2017 Jul.
Article in English | MEDLINE | ID: mdl-28530712

ABSTRACT

During infection, antigen-specific T cells undergo tightly regulated developmental transitions controlled by transcriptional and post-transcriptional regulation of gene expression. We found that the microRNA miR-31 was strongly induced by activation of the T cell antigen receptor (TCR) in a pathway involving calcium and activation of the transcription factor NFAT. During chronic infection with lymphocytic choriomeningitis virus (LCMV) clone 13, miR-31-deficent mice recovered from clinical disease, while wild-type mice continued to show signs of disease. This disease phenotype was explained by the presence of larger numbers of cytokine-secreting LCMV-specific CD8+ T cells in miR-31-deficent mice than in wild-type mice. Mechanistically, miR-31 increased the sensitivity of T cells to type I interferons, which interfered with effector T cell function and increased the expression of several proteins related to T cell dysfunction during chronic infection. These studies identify miR-31 as an important regulator of T cell exhaustion in chronic infection.


Subject(s)
Arenaviridae Infections/immunology , CD8-Positive T-Lymphocytes/immunology , Cytokines/immunology , MicroRNAs/immunology , Receptors, Antigen, T-Cell/immunology , Animals , Antibodies, Viral/immunology , Arenaviridae Infections/genetics , CD8-Positive T-Lymphocytes/drug effects , Calcium/metabolism , Chromatin Immunoprecipitation , Dendritic Cells/immunology , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , Gene Expression Profiling , Immunoblotting , Interferon Type I/pharmacology , Lymphocytic choriomeningitis virus/immunology , Mice , Mice, Knockout , MicroRNAs/genetics , NFATC Transcription Factors/metabolism , Real-Time Polymerase Chain Reaction
9.
Mol Cell ; 77(2): 384-394.e4, 2020 01 16.
Article in English | MEDLINE | ID: mdl-31806351

ABSTRACT

HMCES (5hmC binding, embryonic stem cell-specific-protein), originally identified as a protein capable of binding 5-hydroxymethylcytosine (5hmC), an epigenetic modification generated by TET proteins, was previously reported to covalently crosslink to DNA at abasic sites via a conserved cysteine. We show here that Hmces-deficient mice display normal hematopoiesis without global alterations in 5hmC. HMCES specifically enables DNA double-strand break repair through the microhomology-mediated alternative-end-joining (Alt-EJ) pathway during class switch recombination (CSR) in B cells, and HMCES deficiency leads to a significant defect in CSR. HMCES mediates Alt-EJ through its SOS-response-associated-peptidase domain (SRAPd), a function that requires DNA binding but is independent of its autopeptidase and DNA-crosslinking activities. We show that HMCES is recruited to switch regions of the immunoglobulin locus and provide a potential structural basis for the interaction of HMCES with long DNA overhangs generated by Alt-EJ during CSR. Our studies provide further evidence for a specialized role for HMCES in DNA repair.


Subject(s)
B-Lymphocytes/physiology , DNA End-Joining Repair/genetics , DNA-Binding Proteins/genetics , DNA/genetics , Immunoglobulin Class Switching/genetics , Animals , Cell Line, Tumor , DNA Breaks, Double-Stranded , DNA Damage/genetics , Mice , Mice, Inbred C57BL , Translocation, Genetic/genetics
11.
Nat Immunol ; 15(8): 777-88, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24997565

ABSTRACT

A characteristic feature of asthma is the aberrant accumulation, differentiation or function of memory CD4(+) T cells that produce type 2 cytokines (TH2 cells). By mapping genome-wide histone modification profiles for subsets of T cells isolated from peripheral blood of healthy and asthmatic individuals, we identified enhancers with known and potential roles in the normal differentiation of human TH1 cells and TH2 cells. We discovered disease-specific enhancers in T cells that differ between healthy and asthmatic individuals. Enhancers that gained the histone H3 Lys4 dimethyl (H3K4me2) mark during TH2 cell development showed the highest enrichment for asthma-associated single nucleotide polymorphisms (SNPs), which supported a pathogenic role for TH2 cells in asthma. In silico analysis of cell-specific enhancers revealed transcription factors, microRNAs and genes potentially linked to human TH2 cell differentiation. Our results establish the feasibility and utility of enhancer profiling in well-defined populations of specialized cell types involved in disease pathogenesis.


Subject(s)
Asthma/genetics , Asthma/immunology , Genetic Predisposition to Disease , Th1 Cells/immunology , Th2 Cells/immunology , Adolescent , Adult , Aged , Binding Sites/genetics , Binding Sites/immunology , Cell Differentiation/immunology , Cells, Cultured , Core Binding Factor Alpha 3 Subunit/genetics , DNA Methylation/genetics , Epigenomics , Female , GATA3 Transcription Factor/genetics , Genome-Wide Association Study , Histones/genetics , Histones/immunology , Humans , Immunologic Memory/immunology , Male , MicroRNAs/genetics , Middle Aged , Polymorphism, Single Nucleotide , Promoter Regions, Genetic/genetics , Protein Binding/genetics , Protein Binding/immunology , Sequence Analysis, RNA , T-Box Domain Proteins/genetics , Young Adult
12.
Proc Natl Acad Sci U S A ; 120(37): e2221405120, 2023 09 12.
Article in English | MEDLINE | ID: mdl-37669386

ABSTRACT

DNA methylation functions as a repressive epigenetic mark that can be reversed by the Ten-eleven translocation (TET) family of DNA dioxygenases that sequentially oxidize 5-methylcytosine into 5-hydroxymethylcytosine (5hmC), 5-formylcytosine (5fC), and 5-carboxylcytosine (5caC). Both 5fC and 5caC can be excised by DNA base-excision repair factors leading to unmodified cytosines. TET enzymes were recently implicated as potential risk factors for inflammatory bowel disease (IBD), but the contribution of TET-mediated DNA oxidation to intestinal homeostasis and response to environmental stressors are unknown. Here, we show prominent roles of TET3 in regulating mouse intestinal epithelial differentiation and response to luminal stressors. Compared with wild-type littermates, mice with intestinal epithelial cell-specific ablation of Tet3 (Tet3ΔIEC) demonstrated a decreased transcriptome involved in innate immune response, Paneth cell differentiation, and epithelial regeneration. Tet3IEC mice exhibited an elevated susceptibility to enteric pathogen infection that is correlated with a decreased epithelial 5hmC abundance. Infection of human enterocytes or mice with the pathogenic bacteria acutely increased 5hmC abundance. Genome-wide 5hmC profiling revealed a shift of genomic enrichment of 5hmC toward genes involved in activating Notch, Wnt, and autophagy pathways. Furthermore, chemical stressor dextran sulfate sodium (DSS) represses epithelial 5hmC abundance in a temporal fashion, and Tet3IEC mice exhibited increased susceptibility to DSS experimental colitis with reduced regenerative capacity. TET3 is a critical regulator of gut epithelial DNA methylome and transcriptome, especially in response to luminal stressors, for the maintenance of tissue homeostasis.


Subject(s)
Colitis , Dioxygenases , Animals , Humans , Mice , DNA , Enterocytes , Oxidation-Reduction , Paneth Cells
13.
Proc Natl Acad Sci U S A ; 120(6): e2214824120, 2023 02 07.
Article in English | MEDLINE | ID: mdl-37406303

ABSTRACT

The three mammalian TET dioxygenases oxidize the methyl group of 5-methylcytosine in DNA, and the oxidized methylcytosines are essential intermediates in all known pathways of DNA demethylation. To define the in vivo consequences of complete TET deficiency, we inducibly deleted all three Tet genes in the mouse genome. Tet1/2/3-inducible TKO (iTKO) mice succumbed to acute myeloid leukemia (AML) by 4 to 5 wk. Single-cell RNA sequencing of Tet iTKO bone marrow cells revealed the appearance of new myeloid cell populations characterized by a striking increase in expression of all members of the stefin/cystatin gene cluster on mouse chromosome 16. In patients with AML, high stefin/cystatin gene expression correlates with poor clinical outcomes. Increased expression of the clustered stefin/cystatin genes was associated with a heterochromatin-to-euchromatin compartment switch with readthrough transcription downstream of the clustered stefin/cystatin genes as well as other highly expressed genes, but only minor changes in DNA methylation. Our data highlight roles for TET enzymes that are distinct from their established function in DNA demethylation and instead involve increased transcriptional readthrough and changes in three-dimensional genome organization.


Subject(s)
Dioxygenases , Leukemia, Myeloid, Acute , Animals , Mice , Heterochromatin/genetics , Euchromatin , DNA Methylation , 5-Methylcytosine/metabolism , Leukemia, Myeloid, Acute/genetics , Dioxygenases/genetics , Dioxygenases/metabolism , Mammals/genetics
14.
Proc Natl Acad Sci U S A ; 120(3): e2218332120, 2023 01 17.
Article in English | MEDLINE | ID: mdl-36626549

ABSTRACT

O-GlcNAc transferase (OGT) modifies serine and threonine residues on nuclear and cytosolic proteins with O-linked N-acetylglucosamine (GlcNAc). OGT is essential for mammalian cell viability, but the underlying mechanisms are still enigmatic. We performed a genome-wide CRISPR-Cas9 screen in mouse embryonic stem cells (mESCs) to identify candidates whose depletion rescued the block in cell proliferation induced by OGT deficiency. We show that the block in cell proliferation in OGT-deficient cells stems from mitochondrial dysfunction secondary to mTOR (mechanistic target of rapamycin) hyperactivation. In normal cells, OGT maintains low mTOR activity and mitochondrial fitness through suppression of proteasome activity; in the absence of OGT, increased proteasome activity results in increased steady-state amino acid levels, which in turn promote mTOR lysosomal translocation and activation, and increased oxidative phosphorylation. mTOR activation in OGT-deficient mESCs was confirmed by an independent phospho-proteomic screen. Our study highlights a unique series of events whereby OGT regulates the proteasome/ mTOR/ mitochondrial axis in a manner that maintains homeostasis of intracellular amino acid levels, mitochondrial fitness, and cell viability. A similar mechanism operates in CD8+ T cells, indicating its generality across mammalian cell types. Manipulating OGT activity may have therapeutic potential in diseases in which this signaling pathway is impaired.


Subject(s)
CD8-Positive T-Lymphocytes , Proteasome Endopeptidase Complex , Animals , Mice , Acetylglucosamine/metabolism , CD8-Positive T-Lymphocytes/metabolism , Cell Survival , Mitochondria/metabolism , N-Acetylglucosaminyltransferases/metabolism , Proteasome Endopeptidase Complex/metabolism , Proteomics , TOR Serine-Threonine Kinases/genetics , TOR Serine-Threonine Kinases/metabolism
15.
Immunity ; 45(6): 1327-1340, 2016 12 20.
Article in English | MEDLINE | ID: mdl-27939672

ABSTRACT

In response to acute infection, naive CD8+ T cells expand, differentiate into effector cells, and then contract to a long-lived pool of memory cells after pathogen clearance. During chronic infections or in tumors, CD8+ T cells acquire an "exhausted" phenotype. Here we present genome-wide comparisons of chromatin accessibility and gene expression from endogenous CD8+ T cells responding to acute and chronic viral infection using ATAC-seq and RNA-seq techniques. Acquisition of effector, memory, or exhausted phenotypes was associated with stable changes in chromatin accessibility away from the naive T cell state. Regions differentially accessible between functional subsets in vivo were enriched for binding sites of transcription factors known to regulate these subsets, including E2A, BATF, IRF4, T-bet, and TCF1. Exhaustion-specific accessible regions were enriched for consensus binding sites for NFAT and Nr4a family members, indicating that chronic stimulation confers a unique accessibility profile on exhausted cells.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Chromatin Assembly and Disassembly/immunology , Gene Expression/immunology , Immunologic Memory/immunology , Lymphocyte Activation/immunology , Animals , Arenaviridae Infections/immunology , Chromatin , Disease Models, Animal , Gene Expression Profiling , Lymphocytic choriomeningitis virus , Mice , Mice, Inbred C57BL , Mice, Transgenic
16.
Nat Rev Mol Cell Biol ; 14(6): 341-56, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23698584

ABSTRACT

In many organisms, the methylation of cytosine in DNA has a key role in silencing 'parasitic' DNA elements, regulating transcription and establishing cellular identity. The recent discovery that ten-eleven translocation (TET) proteins are 5-methylcytosine oxidases has provided several chemically plausible pathways for the reversal of DNA methylation, thus triggering a paradigm shift in our understanding of how changes in DNA methylation are coupled to cell differentiation, embryonic development and cancer.


Subject(s)
Cell Differentiation , DNA Methylation , DNA-Binding Proteins/metabolism , Embryonic Development , Neoplasms/metabolism , Proto-Oncogene Proteins/metabolism , Animals , DNA-Binding Proteins/genetics , Humans , Neoplasms/genetics , Proto-Oncogene Proteins/genetics
17.
Nature ; 567(7749): 530-534, 2019 03.
Article in English | MEDLINE | ID: mdl-30814732

ABSTRACT

T cells expressing chimeric antigen receptors (CAR T cells) targeting human CD19 (hCD19) have shown clinical efficacy against B cell malignancies1,2. CAR T cells have been less effective against solid tumours3-5, in part because they enter a hyporesponsive ('exhausted' or 'dysfunctional') state6-9 triggered by chronic antigen stimulation and characterized by upregulation of inhibitory receptors and loss of effector function. To investigate the function of CAR T cells in solid tumours, we transferred hCD19-reactive CAR T cells into hCD19+ tumour-bearing mice. CD8+CAR+ tumour-infiltrating lymphocytes and CD8+ endogenous tumour-infiltrating lymphocytes expressing the inhibitory receptors PD-1 and TIM3 exhibited similar profiles of gene expression and chromatin accessibility, associated with secondary activation of nuclear receptor transcription factors NR4A1 (also known as NUR77), NR4A2 (NURR1) and NR4A3 (NOR1) by the initiating transcription factor NFAT (nuclear factor of activated T cells)10-12. CD8+ T cells from humans with cancer or chronic viral infections13-15 expressed high levels of NR4A transcription factors and displayed enrichment of NR4A-binding motifs in accessible chromatin regions. CAR T cells lacking all three NR4A transcription factors (Nr4a triple knockout) promoted tumour regression and prolonged the survival of tumour-bearing mice. Nr4a triple knockout CAR tumour-infiltrating lymphocytes displayed phenotypes and gene expression profiles characteristic of CD8+ effector T cells, and chromatin regions uniquely accessible in Nr4a triple knockout CAR tumour-infiltrating lymphocytes compared to wild type were enriched for binding motifs for NF-κB and AP-1, transcription factors involved in activation of T cells. We identify NR4A transcription factors as having an important role in the cell-intrinsic program of T cell hyporesponsiveness and point to NR4A inhibition as a promising strategy for cancer immunotherapy.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Neoplasms/genetics , Neoplasms/immunology , Nuclear Receptor Subfamily 4, Group A, Member 1/metabolism , Receptors, Chimeric Antigen/immunology , Transcription Factors/metabolism , Adoptive Transfer , Animals , Antigens, CD19/immunology , CD8-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/pathology , Cell Line, Tumor , Chromatin/genetics , Chromatin/metabolism , DNA-Binding Proteins/deficiency , DNA-Binding Proteins/metabolism , Female , Gene Expression Profiling , Humans , Lymphocytes, Tumor-Infiltrating/immunology , Lymphocytes, Tumor-Infiltrating/metabolism , Lymphocytes, Tumor-Infiltrating/pathology , Male , Melanoma, Experimental/genetics , Melanoma, Experimental/immunology , Melanoma, Experimental/pathology , Mice , Mice, Inbred C57BL , NF-kappa B/metabolism , Neoplasms/pathology , Nuclear Receptor Subfamily 4, Group A, Member 1/deficiency , Nuclear Receptor Subfamily 4, Group A, Member 2/deficiency , Nuclear Receptor Subfamily 4, Group A, Member 2/metabolism , Receptors, Steroid/deficiency , Receptors, Steroid/metabolism , Receptors, Thyroid Hormone/deficiency , Receptors, Thyroid Hormone/metabolism , Survival Rate , Transcription Factor AP-1/metabolism , Transcription Factors/deficiency
19.
Immunity ; 42(2): 265-278, 2015 Feb 17.
Article in English | MEDLINE | ID: mdl-25680272

ABSTRACT

During persistent antigen stimulation, CD8(+) T cells show a gradual decrease in effector function, referred to as exhaustion, which impairs responses in the setting of tumors and infections. Here we demonstrate that the transcription factor NFAT controls the program of T cell exhaustion. When expressed in cells, an engineered form of NFAT1 unable to interact with AP-1 transcription factors diminished T cell receptor (TCR) signaling, increased the expression of inhibitory cell surface receptors, and interfered with the ability of CD8(+) T cells to protect against Listeria infection and attenuate tumor growth in vivo. We defined the genomic regions occupied by endogenous and engineered NFAT1 in primary CD8(+) T cells and showed that genes directly induced by the engineered NFAT1 overlapped with genes expressed in exhausted CD8(+) T cells in vivo. Our data show that NFAT promotes T cell anergy and exhaustion by binding at sites that do not require cooperation with AP-1.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Clonal Anergy/genetics , NFATC Transcription Factors/physiology , Recombinant Proteins/pharmacology , Transcription Factor AP-1/metabolism , Animals , Cells, Cultured , Clonal Anergy/drug effects , Gene Expression Regulation/genetics , Listeria monocytogenes/immunology , Listeriosis/immunology , Listeriosis/microbiology , Lymphocyte Activation/immunology , Mice , Mice, Transgenic , NFATC Transcription Factors/genetics , Neoplasms/immunology , Promoter Regions, Genetic/genetics , Receptors, Antigen, T-Cell/immunology , Recombinant Proteins/genetics
20.
Matern Child Health J ; 28(1): 38-43, 2024 Jan.
Article in English | MEDLINE | ID: mdl-37938443

ABSTRACT

PURPOSE: Perinatal substance use disorders (SUDs) remain an urgent public health concern in the United States and are associated with increased maternal and infant morbidity and mortality. Establishing holistic prenatal care among this population allows for engaging or re-engaging the pregnant population in appropriate medical care, including treatment for SUD. DESCRIPTION: The Florida Department of Health in Citrus County (DOH-Citrus) noticed an increase in SUD among their pregnant population and developed a pilot program that incorporates Medication for Opioid Use Disorder (MOUD) and mental health services during routine prenatal care appointments. ASSESSMENT: Since the launch of the pilot program, DOH-Citrus has provided prenatal programs with buprenorphine assistance to 23 members of the community. CONCLUSION: A growing number of local health departments (LHDs) provide harm reduction supplies, overdose prevention education, and local resources for treatment and other life-saving services. In many communities, LHDs are typically the most accessible sources of public health information and health care services. By framing the pilot program as a prenatal care center that incorporates SUD treatment as the prescribed standard of care, DOH-Citrus has implemented a holistic model for treating SUD and reducing barriers while improving continuity of care. LHDs are uniquely positioned to implement harm reduction strategies that address perinatal SUDs, treatment, and recovery within maternal and child health populations. As a health department located in a state without expanded Medicaid and with high rates of uninsured people, this pilot program has the potential to be replicated in other states facing similar challenges.


Subject(s)
Mental Health Services , Opioid-Related Disorders , Pregnancy , Infant , Child , Female , United States , Humans , Florida/epidemiology , Analgesics, Opioid , Prenatal Care , Opioid-Related Disorders/epidemiology , Opioid-Related Disorders/prevention & control
SELECTION OF CITATIONS
SEARCH DETAIL