Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
1.
Front Immunol ; 14: 1161901, 2023.
Article in English | MEDLINE | ID: mdl-37600767

ABSTRACT

Introduction: The imbalance between Th17 and regulatory T cells in inflammatory bowel diseases (IBD) promotes intestinal epithelial cell damage. In this scenario, T helper cell lineage commitment is accompanied by dynamic changes to the chromatin that facilitate or repress gene expression. Methods: Here, we characterized the chromatin landscape and heterogeneity of intestinal and peripheral CD4 T cellsfrom IBD patients using in house ATAC-Seq and single cell RNA-Seq libraries. Results: We show that chromatin accessibility profiles of CD4 T cells from inflamed intestinal biopsies relate to genes associated with a network of inflammatory processes. After integrating the chromatin profiles of tissue-derived CD4 T cells and in-vitro polarized CD4 T cell subpopulations, we found that the chromatin accessibility changes of CD4 T cells were associated with a higher predominance of pathogenic Th17 cells (pTh17 cells) in inflamed biopsies. In addition, IBD risk loci in CD4 T cells were colocalized with accessible chromatin changes near pTh17-related genes, as shown in intronic STAT3 and IL23R regions enriched in areas of active intestinal inflammation. Moreover, single cell RNA-Seq analysis revealed a population of pTh17 cells that co-expresses Th1 and cytotoxic transcriptional programs associated with IBD severity. Discussion: Altogether, we show that cytotoxic pTh17 cells were specifically associated with IBD genetic variants and linked to intestinal inflammation of IBD patients.


Subject(s)
Chromatin , Inflammatory Bowel Diseases , Humans , Chromatin/genetics , CD4-Positive T-Lymphocytes , Th17 Cells , Inflammatory Bowel Diseases/genetics , Inflammation
2.
J Crohns Colitis ; 14(1): 96-109, 2020 Jan 01.
Article in English | MEDLINE | ID: mdl-31158273

ABSTRACT

BACKGROUND: The aetiology of Crohn's disease [CD] involves immune dysregulation in a genetically susceptible individual. Genome-wide association studies [GWAS] have identified 200 loci associated with CD, ulcerative colitis, or both, most of which fall within non-coding DNA regions. Long non-coding RNAs [lncRNAs] regulate gene expression by diverse mechanisms and have been associated with disease activity in inflammatory bowel disease. However, disease-associated lncRNAs have not been characterised in pathogenic immune cell populations. METHODS: Terminal ileal samples were obtained from 22 CD patients and 13 controls. RNA from lamina propria CD4+ T cells was sequenced and long intergenic non-coding RNAs [lincRNAs] were detected. Overall expression patterns, differential expression [DE], and pathway and gene enrichment analyses were performed. Knockdown of novel lincRNAs XLOC_000261 and XLOC_000014 was performed. Expression of Th1 or Th17-associated transcription factors, T-bet and RORγt, respectively, was assessed by flow cytometry. RESULTS: A total of 6402 lincRNAs were expressed, 960 of which were novel. Unsupervised clustering and principal component analysis showed that the lincRNA expression discriminated patients from controls. A total of 1792 lincRNAs were DE, and 295 [79 novel; 216 known] mapped to 267 of 5727 DE protein-coding genes. The novel lincRNAs were enriched in inflammatory and Notch signalling pathways [p <0.05]. Furthermore, DE lincRNAs in CD patients were more frequently found in DNA regions with known inflammatory bowel disease [IBD]-associated loci. The novel lincRNA XLOC_000261 negatively regulated RORγt expression in Th17 cells. CONCLUSIONS: We describe a novel set of DE lincRNAs in CD-associated CD4+ cells and demonstrate that novel lincRNA XLOC_000261 appears to negatively regulate RORγt protein expression in Th17 cells.


Subject(s)
CD4-Positive T-Lymphocytes/physiology , Crohn Disease/etiology , RNA, Long Noncoding/metabolism , Aged , Case-Control Studies , Crohn Disease/metabolism , Crohn Disease/pathology , Female , Gene Expression Profiling , Humans , Male , Middle Aged , Mucous Membrane/metabolism
3.
Article in English | MEDLINE | ID: mdl-30510991

ABSTRACT

Background & Aims: Forkhead box protein 3 (FOXP3)+ regulatory T cell (Treg) dysfunction is associated with autoimmune diseases; however, the mechanisms responsible for inflammatory bowel disease pathophysiology are poorly understood. Here, we tested the hypothesis that a physical interaction between transcription factor FOXP3 and the epigenetic enzyme enhancer of zeste homolog 2 (EZH2) is essential for gene co-repressive function. Methods: Human FOXP3 mutations clinically relevant to intestinal inflammation were generated by site-directed mutagenesis. T lymphocytes were isolated from mice, human blood, and lamina propria of Crohn's disease (CD) patients and non-CD controls. We performed proximity ligation or a co-immunoprecipitation assay in FOXP3-mutant+, interleukin 6 (IL6)-treated or CD-CD4+ T cells to assess FOXP3-EZH2 protein interaction. We studied IL2 promoter activity and chromatin state of the interferon γ locus via luciferase reporter and chromatin-immunoprecipitation assays, respectively, in cells expressing FOXP3 mutants. Results: EZH2 binding was abrogated by inflammatory bowel disease-associated FOXP3 cysteine 232 (C232) mutation. The C232 mutant showed impaired repression of IL2 and diminished EZH2-mediated trimethylation of histone 3 at lysine 27 on interferon γ, indicative of compromised Treg physiologic function. Generalizing this mechanism, IL6 impaired FOXP3-EZH2 interaction. IL6-induced effects were reversed by Janus kinase 1/2 inhibition. In lamina propria-derived CD4+T cells from CD patients, we observed decreased FOXP3-EZH2 interaction. Conclusions: FOXP3-C232 mutation disrupts EZH2 recruitment and gene co-repressive function. The proinflammatory cytokine IL6 abrogates FOXP3-EZH2 interaction. Studies in lesion-derived CD4+ T cells have shown that reduced FOXP3-EZH2 interaction is a molecular feature of CD patients. Destabilized FOXP3-EZH2 protein interaction via diverse mechanisms and consequent Treg abnormality may drive gastrointestinal inflammation.


Subject(s)
Enhancer of Zeste Homolog 2 Protein/metabolism , Forkhead Transcription Factors/metabolism , Inflammation/metabolism , Inflammation/pathology , Intestines/pathology , Adult , Animals , Cell Nucleus/metabolism , Cell Separation , Co-Repressor Proteins/metabolism , Female , Humans , Inflammation/immunology , Inflammatory Bowel Diseases/immunology , Inflammatory Bowel Diseases/pathology , Interleukin-6/metabolism , Janus Kinases/metabolism , Jurkat Cells , Male , Mice, Inbred C57BL , Middle Aged , Mutation/genetics , Phosphorylation , Phosphotyrosine/metabolism , Polycomb Repressive Complex 2/metabolism , Protein Binding , STAT3 Transcription Factor/metabolism , Signal Transduction , T-Lymphocytes, Regulatory/metabolism
4.
Front Immunol ; 9: 2955, 2018.
Article in English | MEDLINE | ID: mdl-30619315

ABSTRACT

T cell lineage decisions are critical for the development of proper immune responses to pathogens as well as important for the resolution of inflammatory responses. This differentiation process relies on a combination of intrinsic and extrinsic factors converging upon epigenetic regulation of transcriptional networks relevant to specific T cell lineages. As these biochemical modifications represent therapeutic opportunities in cancer biology and autoimmunity, implications of writers and readers of epigenetic marks to immune cell differentiation and function are highly relevant. Given the ready adoption of histone methyltransferase inhibitors in the clinic, we focus this review on the role of three histone modifying complexes: PRC-1, PRC-2, and G9A in modulating T cell fate decisions. Furthermore, we explore the role of long non-coding RNAs in regulating these processes, and discuss recent advances and challenges of implementing epigenetic therapies into clinical practice.


Subject(s)
Cell Differentiation/genetics , Epigenesis, Genetic/immunology , Histone Methyltransferases/metabolism , RNA, Long Noncoding/metabolism , T-Lymphocytes/immunology , Cell Differentiation/immunology , Clinical Trials as Topic , DNA Methylation/immunology , Gene Expression Regulation/immunology , Genetic Therapy/methods , Histone Code , Histones/genetics , Histones/immunology , Humans , Immune System Diseases/immunology , Immune System Diseases/surgery , Neoplasms/immunology , Neoplasms/therapy
5.
J Biol Chem ; 292(2): 706-722, 2017 Jan 13.
Article in English | MEDLINE | ID: mdl-27909059

ABSTRACT

Regulatory T (Treg) cells expressing the transcription factor FOXP3 play a pivotal role in maintaining immunologic self-tolerance. We and others have shown previously that EZH2 is recruited to the FOXP3 promoter and its targets in Treg cells. To further address the role for EZH2 in Treg cellular function, we have now generated mice that lack EZH2 specifically in Treg cells (EZH2Δ/ΔFOXP3+). We find that EZH2 deficiency in FOXP3+ T cells results in lethal multiorgan autoimmunity. We further demonstrate that EZH2Δ/ΔFOXP3+ T cells lack a regulatory phenotype in vitro and secrete proinflammatory cytokines. Of special interest, EZH2Δ/ΔFOXP3+ mice develop spontaneous inflammatory bowel disease. Guided by these results, we assessed the FOXP3 and EZH2 gene networks by RNA sequencing in isolated intestinal CD4+ T cells from patients with Crohn's disease. Gene network analysis demonstrates that these CD4+ T cells display a Th1/Th17-like phenotype with an enrichment of gene targets shared by FOXP3 and EZH2. Combined, these results suggest that the inflammatory milieu found in Crohn's disease could lead to or result from deregulation of FOXP3/EZH2-enforced T cell gene networks contributing to the underlying intestinal inflammation.


Subject(s)
Crohn Disease/immunology , Enhancer of Zeste Homolog 2 Protein/immunology , Gene Regulatory Networks/immunology , T-Lymphocytes, Regulatory/immunology , Th17 Cells/immunology , Animals , Crohn Disease/pathology , Cytokines/genetics , Cytokines/immunology , Enhancer of Zeste Homolog 2 Protein/genetics , Forkhead Transcription Factors/genetics , Forkhead Transcription Factors/immunology , Humans , Mice , Mice, Transgenic , T-Lymphocytes, Regulatory/pathology , Th17 Cells/pathology
6.
Nat Med ; 22(6): 657-65, 2016 06.
Article in English | MEDLINE | ID: mdl-27183216

ABSTRACT

Although somatic cell activation of the embryonic stem cell (ESC) pluripotency factor OCT4 has been reported, this previous work has been controversial and has not demonstrated a functional role for OCT4 in somatic cells. Here we demonstrate that smooth muscle cell (SMC)-specific conditional knockout of Oct4 in Apoe(-/-) mice resulted in increased lesion size and changes in lesion composition that are consistent with decreased plaque stability, including a thinner fibrous cap, increased necrotic core area, and increased intraplaque hemorrhage. Results of SMC-lineage-tracing studies showed that these effects were probably the result of marked reductions in SMC numbers within lesions and SMC investment within the fibrous cap, which may result from impaired SMC migration. The reactivation of Oct4 within SMCs was associated with hydroxymethylation of the Oct4 promoter and was hypoxia inducible factor-1α (HIF-1α, encoded by HIF1A) and Krüppel-like factor-4 (KLF4)-dependent. These results provide the first direct evidence that OCT4 has a functional role in somatic cells, and they highlight the potential role of OCT4 in normal and diseased somatic cells.


Subject(s)
Atherosclerosis/genetics , Cell Movement/genetics , Myocytes, Smooth Muscle/metabolism , Octamer Transcription Factor-3/genetics , Plaque, Atherosclerotic/genetics , Animals , Aorta/metabolism , Apolipoproteins E/genetics , Blotting, Western , Cell Lineage , Cell Survival , Chromatin Immunoprecipitation , Coronary Artery Disease/metabolism , Diet, Western , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Kruppel-Like Factor 4 , Kruppel-Like Transcription Factors/metabolism , Male , Mice , Mice, Knockout , Middle Aged , Mutagenesis, Site-Directed , Myocytes, Smooth Muscle/cytology , Octamer Transcription Factor-3/metabolism , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction
7.
Am J Physiol Gastrointest Liver Physiol ; 309(11): G900-9, 2015 Dec 01.
Article in English | MEDLINE | ID: mdl-26472224

ABSTRACT

Krüppel-like factor (KLF)-10 is an important transcriptional regulator of TGF-ß1 signaling in both CD8(+) and CD4(+) T lymphocytes. In the present study, we demonstrate a novel role for KLF10 in the regulation of TGFßRII expression with functional relevance in macrophage differentiation and activation. We first show that transfer of KLF10(-/-) bone marrow-derived macrophages into wild-type (WT) mice leads to exacerbation of experimental colitis. At the cell biological level, using two phenotypic strategies, we show that KLF10-deficient mice have an altered colonic macrophage phenotype with higher frequency of proinflammatory LyC6(+)MHCII(+) cells and a reciprocal decrease of the anti-inflammatory LyC6(-)MHCII(+) subset. Additionally, the anti-inflammatory CD11b(+)CX3CR1(hi) subset of colonic macrophages is significantly decreased in KLF10(-/-) compared with WT mice under inflammatory conditions. Molecularly, CD11b(+) colonic macrophages from KLF10(-/-) mice exhibit a proinflammatory cytokine profile with increased production of TNF-α and lower production of IL-10 in response to LPS stimulation. Because KLF10 is a transcription factor, we explored how this protein may regulate macrophage function. Consequently, we analyzed the expression of TGFßRII expression in colonic macrophages and found that, in the absence of KLF10, macrophages express lower levels of TGFßRII and display an attenuated Smad-2 phosphorylation following TGF-ß1 stimulation. We further show that KLF10 directly binds to the TGFßRII promoter in macrophages, leading to enhanced gene expression through histone H3 acetylation. Collectively, our data reveal a critical role for KLF10 in the epigenetic regulation of TGFßRII expression in macrophages and the acquisition of a "regulatory" phenotype that contributes to intestinal mucosal homeostasis.


Subject(s)
Colitis/metabolism , Colon/metabolism , Early Growth Response Transcription Factors/deficiency , Intestinal Mucosa/metabolism , Kruppel-Like Transcription Factors/deficiency , Macrophages/metabolism , Acetylation , Animals , Base Sequence , Binding Sites , CD11b Antigen/metabolism , CX3C Chemokine Receptor 1 , Colitis/chemically induced , Colitis/genetics , Colitis/pathology , Colon/pathology , Dextran Sulfate , Disease Models, Animal , Early Growth Response Transcription Factors/genetics , Genetic Predisposition to Disease , Histocompatibility Antigens Class II/metabolism , Histones/metabolism , Inflammation Mediators/metabolism , Interleukin-10/metabolism , Intestinal Mucosa/pathology , Kruppel-Like Transcription Factors/genetics , Macrophages/transplantation , Mice, Knockout , Molecular Sequence Data , Phenotype , Phosphorylation , Promoter Regions, Genetic , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Receptor, Transforming Growth Factor-beta Type II , Receptors, Chemokine/metabolism , Receptors, Transforming Growth Factor beta/genetics , Receptors, Transforming Growth Factor beta/metabolism , Signal Transduction , Smad2 Protein/metabolism , Tumor Necrosis Factor-alpha/metabolism
8.
Cell Mol Gastroenterol Hepatol ; 1(2): 188-202.e4, 2015 Mar 01.
Article in English | MEDLINE | ID: mdl-25750932

ABSTRACT

BACKGROUND AND AIMS: KLF proteins function as epigenetic reprogramming factors during cell differentiation in many cell populations and in engineered iPS cells. In this study, we determined KLF14 function in the regulation of FOXP3, a transcription factor critical for Treg cell differentiation. METHODS: We studied the effects of KLF14 on FOXP3 expression at the level of the protein and mRNA. We evaluated the functional relevance of KLF14 to FOXP3+ Treg cells in vitro and in vivo through suppression assays and two colitis models. Finally, we analyzed the effect of KLF14 on the epigenetic landscape of the FOXP3 promoter locus through chromatin immuno-precipitation. RESULTS: KLF14, induced upon activation of naïve CD4+ T cells, segregates to the FOXP3- population and is inversely associated with FOXP3 expression and Treg function. KLF14 KO CD4+ cells differentiated into adaptive Tregs more readily in vitro and in vivo. KLF14 KO cells demonstrated enhanced Treg suppressor function in vitro and in vivo. KLF14 repressed FOXP3 at the level of the mRNA and protein, and by ChIP assay KLF14 was found to bind to the TSDR enhancer region of FOXP3. Furthermore, loss of KLF14 reduced the levels of H3K9me3, HP1 and Suv39H1at the TSDR. CONCLUSIONS: These results outline a novel mechanism by which KLF14 regulates Treg cell differentiation via chromatin remodeling at the FOXP3 TSDR. To our knowledge, this is the first evidence supporting a role for KLF14 in maintaining the differentiated state of Treg cells and outlines a potential mechanism to modify the expression of immune genes, such as FOXP3, which are critical to T cell fate.

9.
Am J Physiol Cell Physiol ; 308(5): C362-71, 2015 Mar 01.
Article in English | MEDLINE | ID: mdl-25472963

ABSTRACT

KLF10 has recently elicited significant attention as a transcriptional regulator of transforming growth factor-ß1 (TGF-ß1) signaling in CD4(+) T cells. In the current study, we demonstrate a novel role for KLF10 in the regulation of TGF-ß receptor II (TGF-ßRII) expression with functional relevance in antiviral immune response. Specifically, we show that KLF10-deficient mice have an increased number of effector/memory CD8(+) T cells, display higher levels of the T helper type 1 cell-associated transcription factor T-bet, and produce more IFN-γ following in vitro stimulation. In addition, KLF10(-/-) CD8(+) T cells show enhanced proliferation in vitro and homeostatic proliferation in vivo. Freshly isolated CD8(+) T cells from the spleen of adult mice express lower levels of surface TGF-ßRII (TßRII). Congruently, in vitro activation of KLF10-deficient CD8(+) T cells upregulate TGF-ßRII to a lesser extent compared with wild-type (WT) CD8(+) T cells, which results in attenuated Smad2 phosphorylation following TGF-ß1 stimulation compared with WT CD8(+) T cells. Moreover, we demonstrate that KLF10 directly binds to the TGF-ßRII promoter in T cells, leading to enhanced gene expression. In vivo viral infection with Daniel's strain Theiler's murine encephalomyelitis virus (TMEV) also led to lower expression of TGF-ßRII among viral-specific KLF10(-/-) CD8(+) T cells and a higher percentage of IFN-γ-producing CD8(+) T cells in the spleen. Collectively, our data reveal a critical role for KLF10 in the transcriptional activation of TGF-ßRII in CD8(+) T cells. Thus, KLF10 regulation of TGF-ßRII in this cell subset may likely play a critical role in viral and tumor immune responses for which the integrity of the TGF-ß1/TGF-ßRII signaling pathway is crucial.


Subject(s)
CD8-Positive T-Lymphocytes/metabolism , Early Growth Response Transcription Factors/physiology , Kruppel-Like Transcription Factors/physiology , Protein Serine-Threonine Kinases/biosynthesis , Receptors, Transforming Growth Factor beta/biosynthesis , Transforming Growth Factor beta/biosynthesis , Animals , Cells, Cultured , Early Growth Response Transcription Factors/deficiency , Gene Expression Regulation , Humans , Jurkat Cells , Kruppel-Like Transcription Factors/deficiency , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Receptor, Transforming Growth Factor-beta Type II
10.
J Biol Chem ; 287(41): 34372-85, 2012 Oct 05.
Article in English | MEDLINE | ID: mdl-22896699

ABSTRACT

Inducible gene expression underlies the epigenetically inherited differentiation program of most immune cells. We report that the promoter of the FOXP3 gene possesses two distinct functional states: an "off state" mediated by the polycomb histone methyltransferase complex and a histone acetyltransferase-dependent "on state." Regulating these states is the presence of a Kruppel-like factor (KLF)-containing Polycomb response element. In the KLF10(-/-) mouse, the FOXP3 promoter is epigenetically silenced by EZH2 (Enhancer of Zeste 2)-mediated trimethylation of Histone 3 K27; thus, impaired FOXP3 induction and inappropriate adaptive T regulatory cell differentiation results in vitro and in vivo. The epigenetic transmittance of adaptive T regulatory cell deficiency is demonstrated throughout more than 40 generations of mice. These results provide insight into chromatin remodeling events key to phenotypic features of distinct T cell populations.


Subject(s)
Early Growth Response Transcription Factors/biosynthesis , Forkhead Transcription Factors/biosynthesis , Gene Silencing/physiology , Kruppel-Like Transcription Factors/biosynthesis , Polycomb-Group Proteins/metabolism , Response Elements/physiology , T-Lymphocytes, Regulatory/metabolism , p300-CBP Transcription Factors/metabolism , Animals , Chromatin Assembly and Disassembly/physiology , Early Growth Response Transcription Factors/genetics , Early Growth Response Transcription Factors/immunology , Enhancer of Zeste Homolog 2 Protein , Forkhead Transcription Factors/genetics , Forkhead Transcription Factors/immunology , Kruppel-Like Transcription Factors/genetics , Kruppel-Like Transcription Factors/immunology , Male , Mice , Mice, Knockout , Polycomb Repressive Complex 2/genetics , Polycomb Repressive Complex 2/immunology , Polycomb Repressive Complex 2/metabolism , Polycomb-Group Proteins/genetics , Polycomb-Group Proteins/immunology , T-Lymphocytes, Regulatory/cytology , T-Lymphocytes, Regulatory/immunology , p300-CBP Transcription Factors/antagonists & inhibitors , p300-CBP Transcription Factors/genetics , p300-CBP Transcription Factors/immunology
11.
Circ Res ; 104(5): 609-18, 2009 Mar 13.
Article in English | MEDLINE | ID: mdl-19168440

ABSTRACT

Phenotypic switching of vascular smooth muscle cells (VSMCs) is known to play a critical role in the development of atherosclerosis. However, the factors present within lesions that mediate VSMC phenotypic switching are unclear. Oxidized phospholipids (OxPLs), including 1-palmitoyl-2-(5-oxovaleroyl)-sn-glycero-3-phosphorylcholine (POVPC), are active components of minimally modified low density lipoprotein and have been previously shown to induce multiple proatherogenic events in endothelial cells and macrophages, but their effects on VSMCs have been largely unexplored until recently. We previously showed that OxPLs induced phenotypic switching of VSMCs, including suppression of SMC differentiation marker genes. The goal of the present studies was to test the hypothesis that OxPLs alter extracellular matrix production and VSMC migration. Results showed that POVPC activated expression of several extracellular matrix proteins in VSMC. POVPC increased expression of type VIII collagen alpha1 chain (Col8a1) mRNA in cultured VSMCs and in vivo in rat carotid arteries by 9-fold and 4-fold, respectively. POVPC-induced activation of Col8a1 gene expression was reduced by small interfering RNA-mediated suppression of Krüppel-like factor 4 (Klf4) and Sp1, and was abolished in Klf4-knockout VSMCs. POVPC increased Klf4 binding to the Col8a1 gene promoter both in vivo in rat carotid arteries and in cultured VSMCs based on chromatin immunoprecipitation assays. Moreover, POVPC-induced VSMC migration was markedly reduced in Klf4- or type VIII collagen-knockout VSMCs. Given evidence that OxPLs are present within atherosclerotic lesions, it is interesting to suggest that OxPL-induced changes in VSMC phenotype may contribute to the pathogenesis of atherosclerosis at least in part through changes in extracellular matrix composition.


Subject(s)
Cell Movement , Collagen Type VIII/metabolism , Muscle, Smooth, Vascular/metabolism , Myocytes, Smooth Muscle/metabolism , Phospholipids/metabolism , Animals , Aorta/metabolism , Apolipoproteins E/genetics , Apolipoproteins E/metabolism , Carotid Arteries/metabolism , Cells, Cultured , Collagen Type VIII/genetics , Kruppel-Like Factor 4 , Kruppel-Like Transcription Factors/genetics , Kruppel-Like Transcription Factors/metabolism , Lipoproteins, LDL/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Oxidation-Reduction , Phenotype , Phosphatidylcholines/metabolism , Phospholipid Ethers/metabolism , Promoter Regions, Genetic , RNA Interference , RNA, Messenger/metabolism , RNA, Small Interfering/metabolism , Rats , Sp1 Transcription Factor/metabolism , Time Factors , Transfection , Up-Regulation
12.
Mol Cell Biol ; 28(1): 140-53, 2008 Jan.
Article in English | MEDLINE | ID: mdl-17954561

ABSTRACT

Hbo1 is a histone acetyltransferase (HAT) that is required for global histone H4 acetylation, steroid-dependent transcription, and chromatin loading of MCM2-7 during DNA replication licensing. It is the catalytic subunit of protein complexes that include ING and JADE proteins, growth regulatory factors and candidate tumor suppressors. These complexes are thought to act via tumor suppressor p53, but the molecular mechanisms and links between stress signaling and chromatin, are currently unknown. Here, we show that p53 physically interacts with Hbo1 and negatively regulates its HAT activity in vitro and in cells. Two physiological stresses that stabilize p53, hyperosmotic shock and DNA replication fork arrest, also inhibit Hbo1 HAT activity in a p53-dependent manner. Hyperosmotic stress during G(1) phase specifically inhibits the loading of the MCM2-7 complex, providing an example of the chromatin output of this pathway. These results reveal a direct regulatory connection between p53-responsive stress signaling and Hbo1-dependent chromatin pathways.


Subject(s)
DNA Replication/genetics , DNA/genetics , Histone Acetyltransferases/metabolism , Signal Transduction , Tumor Suppressor Protein p53/metabolism , Animals , Cell Cycle , Cell Cycle Proteins/metabolism , Cell Line , Enzyme Activation , Histone Acetyltransferases/genetics , Homeodomain Proteins/metabolism , Humans , Mice , Mice, Knockout , Osmotic Pressure , Protein Binding , Transcription Factors/metabolism , Tumor Suppressor Protein p53/deficiency , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Proteins/metabolism
13.
J Cell Sci ; 117(Pt 19): 4449-59, 2004 Sep 01.
Article in English | MEDLINE | ID: mdl-15316069

ABSTRACT

In order to investigate whether covalent histone modifications may be involved in early embryonic reprogramming events, changes in global levels of a series of histone tail modifications were studied during oocyte maturation and pre-implantation mouse development using indirect immunofluorescence and scanning confocal microscopy. Results showed that histone modifications could be classified into two strikingly distinct categories. The first contains stable 'epigenetic' marks such as histone H3 lysine 9 methylation [Me(Lys9)H3], histone H3 lysine 4 methylation [Me(Lys4)H3] and histone H4/H2A serine 1 phosphorylation [Ph(Ser1)H4/H2A]. The second group contains dynamic and reversible marks and includes hyperacetylated histone H4, histone H3 arginine 17 methylation [Me(Arg17)H3] and histone H4 arginine 3 methylation [Me(Arg3)H4]). Our results also showed that removal of these marks in eggs and early embryos occurs during metaphase suggesting that the enzymes responsible for the loss of these modifications are probably cytoplasmic in nature. Finally, we provide data demonstrating that treatment of cellular histones with peptidylarginine deiminase (PAD) results in loss of staining for the histone H4 arginine 3 methyl mark, suggesting that PADs can reverse histone arginine methyl modifications.


Subject(s)
Histones/metabolism , Hydrolases/metabolism , Metaphase/physiology , Oocytes/metabolism , Acetylation , Animals , Arginine/metabolism , Embryo, Mammalian/metabolism , Female , Fluorescent Antibody Technique, Indirect , Lysine/metabolism , Methylation , Mice , Microscopy, Confocal , Oocytes/growth & development , Phosphorylation , Protein-Arginine Deiminases , Serine/metabolism
14.
Dev Biol ; 256(1): 73-88, 2003 Apr 01.
Article in English | MEDLINE | ID: mdl-12654293

ABSTRACT

Selected for its high relative abundance, a protein spot of MW approximately 75 kDa, pI 5.5 was cored from a Coomassie-stained two-dimensional gel of proteins from 2850 zona-free metaphase II mouse eggs and analyzed by tandem mass spectrometry (TMS), and novel microsequences were identified that indicated a previously uncharacterized egg protein. A 2.4-kb cDNA was then amplified from a mouse ovarian adapter-ligated cDNA library by RACE-PCR, and a unique 2043-bp open reading frame was defined encoding a 681-amino-acid protein. Comparison of the deduced amino acid sequence with the nonredundant database demonstrated that the protein was approximately 40% identical to the calcium-dependent peptidylarginine deiminase (PAD) enzyme family. Northern blotting, RT-PCR, and in situ hybridization analyses indicated that the protein was abundantly expressed in the ovary, weakly expressed in the testis, and absent from other tissues. Based on the homology with PADs and its oocyte-abundant expression pattern, the protein was designated ePAD, for egg and embryo-abundant peptidylarginine deiminase-like protein. Anti-recombinant ePAD monospecific antibodies localized the molecule to the cytoplasm of oocytes in primordial, primary, secondary, and Graafian follicles in ovarian sections, while no other ovarian cell type was stained. ePAD was also expressed in the immature oocyte, mature egg, and through the blastocyst stage of embryonic development, where expression levels began to decrease. Immunoelectron microscopy localized ePAD to egg cytoplasmic sheets, a unique keratin-containing intermediate filament structure found only in mammalian eggs and in early embryos, and known to undergo reorganization at critical stages of development. Previous reports that PAD-mediated deimination of epithelial cell keratin results in cytoskeletal remodeling suggest a possible role for ePAD in cytoskeletal reorganization in the egg and early embryo.


Subject(s)
Cleavage Stage, Ovum/enzymology , Hydrolases/metabolism , Oocytes/enzymology , Amino Acid Sequence , Animals , Antibody Specificity , Base Sequence , Cloning, Molecular , DNA, Complementary/genetics , Female , Gene Expression Regulation, Developmental , Hydrolases/genetics , Hydrolases/immunology , In Situ Hybridization , Male , Mice , Mice, Inbred ICR , Microscopy, Immunoelectron , Molecular Sequence Data , Oocytes/ultrastructure , Ovary/enzymology , Pregnancy , Protein-Arginine Deiminase Type 4 , Protein-Arginine Deiminases , Recombinant Proteins/genetics , Recombinant Proteins/immunology , Recombinant Proteins/metabolism , Sequence Homology, Amino Acid , Solubility
SELECTION OF CITATIONS
SEARCH DETAIL
...