Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
1.
Immunity ; 53(6): 1182-1201.e8, 2020 12 15.
Article in English | MEDLINE | ID: mdl-33242395

ABSTRACT

αß lineage T cells, most of which are CD4+ or CD8+ and recognize MHC I- or MHC II-presented antigens, are essential for immune responses and develop from CD4+CD8+ thymocytes. The absence of in vitro models and the heterogeneity of αß thymocytes have hampered analyses of their intrathymic differentiation. Here, combining single-cell RNA and ATAC (chromatin accessibility) sequencing, we identified mouse and human αß thymocyte developmental trajectories. We demonstrated asymmetric emergence of CD4+ and CD8+ lineages, matched differentiation programs of agonist-signaled cells to their MHC specificity, and identified correspondences between mouse and human transcriptomic and epigenomic patterns. Through computational analysis of single-cell data and binding sites for the CD4+-lineage transcription factor Thpok, we inferred transcriptional networks associated with CD4+- or CD8+-lineage differentiation, and with expression of Thpok or of the CD8+-lineage factor Runx3. Our findings provide insight into the mechanisms of CD4+ and CD8+ T cell differentiation and a foundation for mechanistic investigations of αß T cell development.


Subject(s)
Cell Differentiation/immunology , Cell Lineage/immunology , T-Lymphocyte Subsets/immunology , Thymocytes/immunology , Animals , Antigen Presentation/immunology , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Cell Differentiation/genetics , Cell Lineage/genetics , Epigenome , Gene Expression Regulation , Gene Regulatory Networks , Histocompatibility Antigens/genetics , Histocompatibility Antigens/immunology , Histocompatibility Antigens/metabolism , Humans , Mice , T-Lymphocyte Subsets/metabolism , Thymocytes/metabolism , Thymus Gland/immunology , Transcription Factors/genetics , Transcription Factors/metabolism , Transcriptome
2.
Immunity ; 50(1): 91-105.e4, 2019 01 15.
Article in English | MEDLINE | ID: mdl-30638736

ABSTRACT

Memory CD4+ T cells mediate long-term immunity, and their generation is a key objective of vaccination strategies. However, the transcriptional circuitry controlling the emergence of memory cells from early CD4+ antigen-responders remains poorly understood. Here, using single-cell RNA-seq to study the transcriptome of virus-specific CD4+ T cells, we identified a gene signature that distinguishes potential memory precursors from effector cells. We found that both that signature and the emergence of memory CD4+ T cells required the transcription factor Thpok. We further demonstrated that Thpok cell-intrinsically protected memory cells from a dysfunctional, effector-like transcriptional program, similar to but distinct from the exhaustion pattern of cells responding to chronic infection. Mechanistically, Thpok- bound genes encoding the transcription factors Blimp1 and Runx3 and acted by antagonizing their expression. Thus, a Thpok-dependent circuitry promotes both memory CD4+ T cells' differentiation and functional fitness, two previously unconnected critical attributes of adaptive immunity.


Subject(s)
CD4-Positive T-Lymphocytes/physiology , T-Lymphocyte Subsets/physiology , Transcription Factors/metabolism , Animals , Antigens, Viral/immunology , Cell Differentiation , Cells, Cultured , Core Binding Factor Alpha 3 Subunit/metabolism , Humans , Immunologic Memory/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Positive Regulatory Domain I-Binding Factor 1/metabolism , Protein Binding , Sequence Analysis, RNA , Single-Cell Analysis , Transcription Factors/genetics , Transcriptome
3.
Immunity ; 51(3): 465-478.e6, 2019 09 17.
Article in English | MEDLINE | ID: mdl-31422869

ABSTRACT

The generation of high-affinity neutralizing antibodies, the objective of most vaccine strategies, occurs in B cells within germinal centers (GCs) and requires rate-limiting "help" from follicular helper CD4+ T (Tfh) cells. Although Tfh differentiation is an attribute of MHC II-restricted CD4+ T cells, the transcription factors driving Tfh differentiation, notably Bcl6, are not restricted to CD4+ T cells. Here, we identified a requirement for the CD4+-specific transcription factor Thpok during Tfh cell differentiation, GC formation, and antibody maturation. Thpok promoted Bcl6 expression and bound to a Thpok-responsive region in the first intron of Bcl6. Thpok also promoted the expression of Bcl6-independent genes, including the transcription factor Maf, which cooperated with Bcl6 to mediate the effect of Thpok on Tfh cell differentiation. Our findings identify a transcriptional program that links the CD4+ lineage with Tfh differentiation, a limiting factor for efficient B cell responses, and suggest avenues to optimize vaccine generation.


Subject(s)
Cell Differentiation/immunology , Proto-Oncogene Proteins c-bcl-6/immunology , Proto-Oncogene Proteins c-maf/immunology , T-Lymphocytes, Helper-Inducer/immunology , Transcription Factors/immunology , Transcription, Genetic/immunology , Animals , Antibodies, Neutralizing/immunology , B-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/immunology , Female , Gene Expression Regulation/immunology , Germinal Center/immunology , Lymphocyte Activation/immunology , Mice , Mice, Inbred C57BL
4.
Immunity ; 39(2): 272-85, 2013 Aug 22.
Article in English | MEDLINE | ID: mdl-23973223

ABSTRACT

Regulatory T (Treg) cells suppress inflammatory immune responses and autoimmunity caused by self-reactive T cells. The key Treg cell transcription factor Foxp3 is downregulated during inflammation to allow for the acquisition of effector T cell-like functions. Here, we demonstrate that stress signals elicited by proinflammatory cytokines and lipopolysaccharides lead to the degradation of Foxp3 through the action of the E3 ubiquitin ligase Stub1. Stub1 interacted with Foxp3 to promote its K48-linked polyubiquitination in an Hsp70-dependent manner. Knockdown of endogenous Stub1 or Hsp70 prevented Foxp3 degradation. Furthermore, the overexpression of Stub1 in Treg cells abrogated their ability to suppress inflammatory immune responses in vitro and in vivo and conferred a T-helper-1-cell-like phenotype. Our results demonstrate the critical role of the stress-activated Stub1-Hsp70 complex in promoting Treg cell inactivation, thus providing a potential therapeutic target for the intervention against autoimmune disease, infection, and cancer.


Subject(s)
Forkhead Transcription Factors/metabolism , HSP70 Heat-Shock Proteins/metabolism , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/metabolism , Ubiquitin-Protein Ligases/metabolism , Animals , Autoimmune Diseases/immunology , Autoimmune Diseases/metabolism , Cells, Cultured , Cytokines/metabolism , Enzyme Inhibitors , HEK293 Cells , HSP70 Heat-Shock Proteins/genetics , Humans , Imidazoles , Inflammation/genetics , Inflammation/immunology , Lipopolysaccharides/metabolism , Mice , Mice, Inbred BALB C , Phenotype , Pyridines , RNA Interference , RNA, Small Interfering , T-Lymphocytes, Helper-Inducer/immunology , Ubiquitin-Protein Ligases/genetics , Ubiquitination
5.
EMBO Rep ; 21(9): e50308, 2020 09 03.
Article in English | MEDLINE | ID: mdl-32644293

ABSTRACT

The transcription factor forkhead box P3 (FOXP3) is essential for the development of regulatory T cells (Tregs) and their function in immune homeostasis. Previous studies have shown that in natural Tregs (nTregs), FOXP3 can be regulated by polyubiquitination and deubiquitination. However, the molecular players active in this pathway, especially those modulating FOXP3 by deubiquitination in the distinct induced Treg (iTreg) lineage, remain unclear. Here, we identify the ubiquitin-specific peptidase 44 (USP44) as a novel deubiquitinase for FOXP3. USP44 interacts with and stabilizes FOXP3 by removing K48-linked ubiquitin modifications. Notably, TGF-ß induces USP44 expression during iTreg differentiation. USP44 co-operates with USP7 to stabilize and deubiquitinate FOXP3. Tregs genetically lacking USP44 are less effective than their wild-type counterparts, both in vitro and in multiple in vivo models of inflammatory disease and cancer. These findings suggest that USP44 plays an important role in the post-translational regulation of Treg function and is thus a potential therapeutic target for tolerance-breaking anti-cancer immunotherapy.


Subject(s)
Forkhead Transcription Factors , T-Lymphocytes, Regulatory , Forkhead Transcription Factors/genetics , Humans , Inflammation/genetics , Transforming Growth Factor beta , Ubiquitin Thiolesterase , Ubiquitin-Specific Peptidase 7
6.
Proc Natl Acad Sci U S A ; 112(25): E3246-54, 2015 Jun 23.
Article in English | MEDLINE | ID: mdl-26060310

ABSTRACT

Forkhead box P3 (FOXP3)-positive Treg cells are crucial for maintaining immune homeostasis. FOXP3 cooperates with its binding partners to elicit Treg cells' signature and function, but the molecular mechanisms underlying the modulation of the FOXP3 complex remain unclear. Here we report that Deleted in breast cancer 1 (DBC1) is a key subunit of the FOXP3 complex. We found that DBC1 interacts physically with FOXP3, and depletion of DBC1 attenuates FOXP3 degradation in inflammatory conditions. Treg cells from Dbc1-deficient mice were more resistant to inflammation-mediated abrogation of Foxp3 expression and function and delayed the onset and severity of experimental autoimmune encephalomyelitis and colitis in mice. These findings establish a previously unidentified mechanism regulating FOXP3 stability during inflammation and reveal a pathway for potential therapeutic modulation and intervention in inflammatory diseases.


Subject(s)
Adaptor Proteins, Signal Transducing/physiology , Forkhead Transcription Factors/physiology , T-Lymphocytes, Regulatory/immunology , Animals , Encephalomyelitis, Autoimmune, Experimental/immunology , Mice , Mice, Inbred C57BL
7.
Int J Mol Sci ; 16(11): 27956-66, 2015 Nov 24.
Article in English | MEDLINE | ID: mdl-26610488

ABSTRACT

IL-33 is a new member of the IL-1 family cytokines, which is expressed by different types of immune cells and non-immune cells. IL-33 is constitutively expressed in the nucleus, where it can act as a transcriptional regulator. So far, no direct target for nuclear IL-33 has been identified, and the regulation of IL-33 nuclear function remains largely unclear. Here, we report that the transcription of type 2 inflammatory cytokine IL-13 is positively regulated by nuclear IL-33. IL-33 can directly bind to the conserved non-coding sequence (CNS) before the translation initiation site in the IL13 gene locus. Moreover, IL-33 nuclear function and stability are regulated by the enzyme ubiquitin-specific protease 17 (USP17) through deubiquitination of IL-33 both at the K48 and at the K63 sites. Our data suggest that IL13 gene transcription can be directly activated by nuclear IL-33, which is negatively regulated by the deubiquitinase USP17.


Subject(s)
Cell Nucleus/metabolism , Endopeptidases/metabolism , Interleukin-33/metabolism , Chromatin/genetics , Chromatin/metabolism , Endopeptidases/genetics , Gene Expression Regulation , Genetic Loci , HEK293 Cells , Humans , Interleukin-13/genetics , Interleukin-33/genetics , Protein Binding , Protein Stability , Ubiquitination
8.
J Biol Chem ; 288(22): 15537-46, 2013 May 31.
Article in English | MEDLINE | ID: mdl-23609452

ABSTRACT

The abundant expression of IFNγ in Th-inducing POK (ThPOK)-deficient CD4(+) T cells requires the activation of Eomesodermin (Eomes); however, the underlying mechanism of this phenomenon remains unclear. Here we report that ThPOK binds directly to the promoter region of the Eomes gene to repress its expression in CD4(+) T cells. We identified the histone acetyltransferase TIP60 as a co-repressor of ThPOK-target genes, where ectopically expressed TIP60 increased ThPOK protein stability by promoting its acetylation at its Lys(360) residue to then augment the transcriptional repression of Eomes. Moreover, knockdown of endogenous TIP60 abolished the stabilization of ThPOK in CD4(+) T cells, which led to the transcriptional activation of Eomes and increased production of IFNγ. Our results reveal a novel pathway by which TIP60 and ThPOK synergistically suppresses Eomes function and IFNγ production, which could contribute to the regulation of inflammation.


Subject(s)
CD4-Positive T-Lymphocytes/metabolism , DNA-Binding Proteins/metabolism , Gene Expression Regulation/immunology , Histone Acetyltransferases/metabolism , Repressor Proteins/metabolism , T-Box Domain Proteins/metabolism , Transcription Factors/metabolism , Acetylation , CD4-Positive T-Lymphocytes/cytology , CD4-Positive T-Lymphocytes/immunology , DNA-Binding Proteins/genetics , DNA-Binding Proteins/immunology , Gene Expression Regulation/genetics , HEK293 Cells , Histone Acetyltransferases/genetics , Histone Acetyltransferases/immunology , Humans , Inflammation/genetics , Inflammation/immunology , Inflammation/metabolism , Interferon-gamma/biosynthesis , Interferon-gamma/genetics , Interferon-gamma/immunology , Lysine Acetyltransferase 5 , Promoter Regions, Genetic/genetics , Promoter Regions, Genetic/immunology , Protein Stability , Repressor Proteins/genetics , Repressor Proteins/immunology , T-Box Domain Proteins/genetics , T-Box Domain Proteins/immunology , Transcription Factors/genetics , Transcription Factors/immunology
9.
J Biol Chem ; 288(13): 9373-82, 2013 Mar 29.
Article in English | MEDLINE | ID: mdl-23395819

ABSTRACT

The expression of the transcription factor GATA3 in FOXP3(+) regulatory T (Treg) cells is crucial for their physiological function in limiting inflammatory responses. Although other studies have shown how T cell receptor (TcR) signals induce the up-regulation of GATA3 expression in Treg cells, the underlying mechanism that maintains GATA3 expression in Treg cells remains unclear. Here, we show how USP21 interacts with and stabilizes GATA3 by mediating its deubiquitination. In a T cell line model, we found that TcR stimulation promoted USP21 expression, which was further up-regulated in the presence of FOXP3. The USP21 mutant C221A reduced its capacity to stabilize GATA3 expression, and its knockdown led to the down-regulation of GATA3 protein expression in Treg cells. Furthermore, we found that FOXP3 could directly bind to the USP21 gene promoter and activated its transcription upon TcR stimulation. Finally, USP21, GATA3, and FOXP3 were found up-regulated in Treg cells that were isolated from asthmatic subjects. In summary, we have identified a USP21-mediated pathway that promotes GATA3 stabilization and expression at the post-translational level. We propose that this pathway forms an important signaling loop that stabilizes the expression of GATA3 in Treg cells.


Subject(s)
GATA3 Transcription Factor/metabolism , Gene Expression Regulation , Ubiquitin Thiolesterase/metabolism , Adolescent , Adult , Asthma/metabolism , Cell Line, Tumor , Forkhead Transcription Factors/biosynthesis , HEK293 Cells , Humans , Lymphocyte Activation , Middle Aged , Promoter Regions, Genetic , Protein Processing, Post-Translational , T-Lymphocytes/metabolism
10.
J Biol Chem ; 288(49): 35093-103, 2013 Dec 06.
Article in English | MEDLINE | ID: mdl-24129573

ABSTRACT

Although lysine methylation is classically known to regulate histone function, its role in modulating antiviral restriction factor activity remains uncharacterized. Interferon-induced transmembrane protein 3 (IFITM3) was found monomethylated on its lysine 88 residue (IFITM3-K88me1) to reduce its antiviral activity, mediated by the lysine methyltransferase SET7. Vesicular stomatitis virus and influenza A virus infection increased IFITM3-K88me1 levels by promoting the interaction between IFITM3 and SET7, suggesting that this pathway could be hijacked to support infection; conversely, IFN-α reduced IFITM3-K88me1 levels. These findings may have important implications in the design of therapeutics targeting protein methylation against infectious diseases.


Subject(s)
Histone-Lysine N-Methyltransferase/metabolism , Membrane Proteins/chemistry , Membrane Proteins/metabolism , RNA-Binding Proteins/chemistry , RNA-Binding Proteins/metabolism , Amino Acid Sequence , Animals , Cell Line , Chlorocebus aethiops , Gene Knockdown Techniques , HEK293 Cells , Histone-Lysine N-Methyltransferase/antagonists & inhibitors , Histone-Lysine N-Methyltransferase/genetics , Humans , Influenza A virus/immunology , Influenza A virus/pathogenicity , Interferon Type I/metabolism , Lysine/chemistry , Membrane Proteins/genetics , Methylation , Molecular Sequence Data , Protein Processing, Post-Translational , RNA-Binding Proteins/genetics , Signal Transduction , Vero Cells , Vesiculovirus/immunology , Vesiculovirus/pathogenicity , Virus Diseases/immunology , Virus Diseases/metabolism , Virus Diseases/prevention & control
11.
Biochim Biophys Acta ; 1820(12): 1886-92, 2012 Dec.
Article in English | MEDLINE | ID: mdl-22954804

ABSTRACT

BACKGROUND: Cyclophilins (CyPs) are cellular proteins that are essential to hepatitis C virus (HCV) replication. Since cyclosporine A was discovered to inhibit HCV infection, the CyP pathway contributing to HCV replication is a potential attractive stratagem for controlling HCV infection. Among them, CyPA is accepted to interact with HCV nonstructural protein (NS) 5A, although interaction of CyPB and NS5B, an RNA-dependent RNA polymerase (RdRp), was proposed first. METHODS: CyPA, CyPB, and HCV RdRp were expressed in bacteria and purified using combination column chromatography. HCV RdRp activity was analyzed in vitro with purified CyPA and CyPB. RESULTS: CyPA at a high concentration (50× higher than that of RdRp) but not at low concentration activated HCV RdRp. CyPB had an allosteric effect on genotype 1b RdRp activation. CyPB showed genotype specificity and activated genotype 1b and J6CF (2a) RdRps but not genotype 1a or JFH1 (2a) RdRps. CyPA activated RdRps of genotypes 1a, 1b, and 2a. CyPB may also support HCV genotype 1b replication within the infected cells, although its knockdown effect on HCV 1b replicon activity was controversial in earlier reports. CONCLUSIONS: CyPA activated HCV RdRp at the early stages of transcription, including template RNA binding. CyPB also activated genotype 1b RdRp. However, their activation mechanisms are different. GENERAL SIGNIFICANCE: These data suggest that both CyPA and CyPB are excellent targets for the treatment of HCV 1b, which shows the greatest resistance to interferon and ribavirin combination therapy.


Subject(s)
Cyclophilin A/metabolism , Cyclophilins/metabolism , Hepacivirus/genetics , Hepatitis C/metabolism , RNA-Dependent RNA Polymerase/genetics , RNA-Dependent RNA Polymerase/metabolism , Virus Replication , Cyclophilin A/genetics , Cyclophilin A/isolation & purification , Cyclophilins/genetics , Cyclophilins/isolation & purification , Cyclosporine/pharmacology , Gene Expression Regulation, Enzymologic , Genotype , Hepacivirus/enzymology , Hepatitis C/virology , Humans , Immunosuppressive Agents/pharmacology , In Vitro Techniques , Mutant Proteins/genetics , Mutant Proteins/metabolism , Plasmids , RNA, Viral/genetics , RNA, Viral/metabolism , Transcription, Genetic
12.
Methods Mol Biol ; 2580: 165-171, 2023.
Article in English | MEDLINE | ID: mdl-36374456

ABSTRACT

Bone marrow chimeras are widely used in immunological studies, to dissect the contributions of hematopoietic and non-hematopoietic cells in immune cell development or functions, to quantify the impact of a given mutation, or in preclinical studies for hematopoietic stem cell transplantation. Here we describe a set of procedures for the generation of bone marrow chimeras.


Subject(s)
Bone Marrow , Hematopoietic Stem Cell Transplantation , Bone Marrow Transplantation , Cell Differentiation
13.
Methods Mol Biol ; 2580: 199-209, 2023.
Article in English | MEDLINE | ID: mdl-36374459

ABSTRACT

T cells develop in the thymus from bone marrow precursors, and genetic manipulation is an indispensable tool to explore their development in vivo. Retroviral transduction of T cell precursors in the bone marrow can be used to specifically eliminate or enforce gene expression. Here, we describe a fast and efficient method to ectopically express a gene in T cell precursors through retroviral transduction and transplant into recipient mice, which will enable laboratories to evaluate gene function in T cell development in vivo.


Subject(s)
Receptors, Antigen, T-Cell , Retroviridae , Mice , Animals , Receptors, Antigen, T-Cell/genetics , Retroviridae/genetics , Retroviridae/metabolism , Thymus Gland/metabolism , Cell Differentiation/genetics , Bone Marrow/metabolism , Bone Marrow Cells/metabolism
14.
Sci Immunol ; 8(89): eadi9066, 2023 11 10.
Article in English | MEDLINE | ID: mdl-37948511

ABSTRACT

How CD4+ lineage gene expression is initiated in differentiating thymocytes remains poorly understood. Here, we show that the paralog transcription factors Zfp281 and Zfp148 control both this process and cytokine expression by T helper cell type 2 (TH2) effector cells. Genetic, single-cell, and spatial transcriptomic analyses showed that these factors promote the intrathymic CD4+ T cell differentiation of class II major histocompatibility complex (MHC II)-restricted thymocytes, including expression of the CD4+ lineage-committing factor Thpok. In peripheral T cells, Zfp281 and Zfp148 promoted chromatin opening at and expression of TH2 cytokine genes but not of the TH2 lineage-determining transcription factor Gata3. We found that Zfp281 interacts with Gata3 and is recruited to Gata3 genomic binding sites at loci encoding Thpok and TH2 cytokines. Thus, Zfp148 and Zfp281 collaborate with Gata3 to promote CD4+ T cell development and TH2 cell responses.


Subject(s)
CD4-Positive T-Lymphocytes , Transcription Factors , Animals , Mice , CD4-Positive T-Lymphocytes/metabolism , Cell Differentiation/genetics , Cytokines/metabolism , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism
15.
Sci Immunol ; 7(72): eabn5917, 2022 06 10.
Article in English | MEDLINE | ID: mdl-35687698

ABSTRACT

Although BTB-zinc finger (BTB-ZF) transcription factors control the differentiation of multiple hematopoietic and immune lineages, how they function is poorly understood. The BTB-ZF factor Thpok controls intrathymic CD4+ T cell development and the expression of most CD4+ and CD8+ lineage genes. Here, we identify the nucleosome remodeling and deacetylase (NuRD) complex as a critical Thpok cofactor. Using mass spectrometry and coimmunoprecipitation in primary T cells, we show that Thpok binds NuRD components independently of DNA association. We locate three amino acid residues within the Thpok BTB domain that are required for both NuRD binding and Thpok functions. Conversely, a chimeric protein merging the NuRD component Mta2 to a BTB-less version of Thpok supports CD4+ T cell development, indicating that NuRD recruitment recapitulates the functions of the Thpok BTB domain. We found that NuRD mediates Thpok repression of CD8+ lineage genes, including the transcription factor Runx3, but is dispensable for Cd4 expression. We show that these functions cannot be performed by the BTB domain of the Thpok-related factor Bcl6, which fails to bind NuRD. Thus, cofactor binding critically contributes to the functional specificity of BTB-ZF factors, which control the differentiation of most hematopoietic subsets.


Subject(s)
CD4-Positive T-Lymphocytes , Mi-2 Nucleosome Remodeling and Deacetylase Complex , Cell Differentiation , Cell Lineage , Mi-2 Nucleosome Remodeling and Deacetylase Complex/metabolism , Transcription Factors
17.
FEBS Lett ; 591(13): 1929-1939, 2017 07.
Article in English | MEDLINE | ID: mdl-28542810

ABSTRACT

Glucocorticoid-induced TNFR-related protein (GITR) is constitutively expressed in T regulatory (Treg) cells and regulates their suppressive function. We identified two methylated CpG islands in the Gitr locus. Using a ChIP assay, we demonstrate that both DNMT1 and methyl-CpG-binding domain Protein 4 (MBD4) bind to the Gitr promoter. Moreover, knockdown of DNMT1 decreases the binding activity of MBD4. We observed much higher levels of both DNMT1 and MBD4 in human CD4+ CD25- conventional T (Tconv) cells. Moreover, co-overexpression of DNMT1 and MBD4 in Treg cells significantly inhibits GITR expression and impairs their suppressive activity. Our results reveal a novel molecular mechanism by which MBD4 inhibits GITR expression in a DNMT1-dependent manner.


Subject(s)
DNA (Cytosine-5-)-Methyltransferases/metabolism , Endodeoxyribonucleases/metabolism , Gene Expression Regulation , Glucocorticoid-Induced TNFR-Related Protein/genetics , T-Lymphocytes, Regulatory/metabolism , CpG Islands/genetics , DNA (Cytosine-5-)-Methyltransferase 1 , DNA Methylation , Humans , Promoter Regions, Genetic/genetics
18.
Sci Rep ; 5: 16355, 2015 Nov 09.
Article in English | MEDLINE | ID: mdl-26549310

ABSTRACT

T helper 17 (Th17) cells not only play critical roles in protecting against bacterial and fungal infections but are also involved in the pathogenesis of autoimmune diseases. The retinoic acid-related orphan receptor (RORγt) is a key transcription factor involved in Th17 cell differentiation through direct transcriptional activation of interleukin 17(A) (IL-17). How RORγt itself is regulated remains unclear. Here, we report that p300, which has histone acetyltransferase (HAT) activity, interacts with and acetylates RORγt at its K81 residue. Knockdown of p300 downregulates RORγt protein and RORγt-mediated gene expression in Th17 cells. In addition, p300 can promote RORγt-mediated transcriptional activation. Interestingly, the histone deacetylase (HDAC) HDAC1 can also interact with RORγt and reduce its acetylation level. In summary, our data reveal previously unappreciated posttranslational regulation of RORγt, uncovering the underlying mechanism by which the histone acetyltransferase p300 and the histone deacetylase HDAC1 reciprocally regulate the RORγt-mediated transcriptional activation of IL-17.


Subject(s)
E1A-Associated p300 Protein/metabolism , Histone Deacetylase 1/metabolism , Nuclear Receptor Subfamily 1, Group F, Member 3/metabolism , Acetylation/drug effects , Gene Expression Regulation , Histone Deacetylase Inhibitors/pharmacology , Humans , Interleukin-17/genetics , Protein Binding , Protein Stability , Th17 Cells/drug effects , Th17 Cells/metabolism , Transcription, Genetic
19.
FEBS Lett ; 588(6): 878-83, 2014 Mar 18.
Article in English | MEDLINE | ID: mdl-24561192

ABSTRACT

Nuclear factor of activated T cells (NFAT) is an important regulator of T cell activation. However, the molecular mechanism whereby NFATc2 regulates IL2 transcription is not fully understood. In this study, we showed that ubiquitin-specific protease 22 (USP22), known as a cancer stem cell marker, specifically interacted with and deubiquitinated NFATc2. USP22 stabilized NFATc2 protein levels, which required its deubiquitinase activity. Consistent with these observations, depletion of USP22 in T cells reduced the expression of IL2, which is a cytokine that signifies T effector cell activation. Our findings thus unveil a previously uncharacterized positive regulator of NFATc2, suggesting that targeting the deubiquitinase activity of USP22 could have therapeutic benefit to control IL2 expression and T cell function.


Subject(s)
Interleukin-2/metabolism , NFATC Transcription Factors/metabolism , Thiolester Hydrolases/metabolism , Gene Knockdown Techniques , HEK293 Cells , Humans , Interleukin-2/genetics , Jurkat Cells , Protein Interaction Mapping , Protein Stability , RNA, Small Interfering/genetics , Thiolester Hydrolases/genetics , Transcription, Genetic , Transcriptional Activation , Ubiquitin Thiolesterase , Ubiquitination
20.
Int J Clin Exp Pathol ; 5(7): 626-33, 2012.
Article in English | MEDLINE | ID: mdl-22977658

ABSTRACT

The forkhead family transcription factor FOXP3 is critical for the differentiation and function of CD4(+) CD25(+) regulatory T cells (Treg). How FOXP3 protein level is negatively regulated under the inflammatory microenvironment is largely unknown. Here we report that the combination of transforming growth factor-beta (TGF-ß) and IL-6 treatment (IL-6/TGF-ß) can synergistically downregulate FOXP3 at the posttranslational level by promoting FOXP3 protein degradation. In our FOXP3 overexpression model, we found that IL-6/TGF-ß treatment upregulated IL-6R expression but did not affect the stability of FOXP3 mRNA. Moreover, we found that the proteasome inhibitor MG132 could inhibit IL-6/TGF-ß-mediated downregulation of FOXP3 protein, which reveals a potential pathway for modulating Treg activity by preventing FOXP3 degradation during inflammation.


Subject(s)
Forkhead Transcription Factors/metabolism , Interleukin-6/pharmacology , Signal Transduction/drug effects , Transforming Growth Factor beta/pharmacology , Cellular Microenvironment/physiology , Down-Regulation/drug effects , Drug Synergism , Humans , Inflammation/pathology , Interleukin-6/metabolism , Jurkat Cells , Leupeptins/pharmacology , Proteolysis/drug effects , RNA, Messenger/metabolism , Receptors, Interleukin-6/biosynthesis , Signal Transduction/physiology , T-Lymphocytes, Regulatory/drug effects , T-Lymphocytes, Regulatory/metabolism , Transforming Growth Factor beta/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL