Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 31
Filter
Add more filters










Publication year range
1.
Cancers (Basel) ; 14(24)2022 Dec 15.
Article in English | MEDLINE | ID: mdl-36551676

ABSTRACT

The Farnesoid X Receptor (FXR) belongs to the nuclear receptor superfamily and is an essential bile acid (BA) receptor that regulates the expression of genes involved in the metabolism of BAs. FXR protects the liver from BA overload, which is a major etiology of hepatocellular carcinoma. Herein, we investigated the changes in gene expression and chromatin accessibility in hepatocytes by performing RNA-seq in combination with the Assay for Transposase-Accessible Chromatin with high-throughput sequencing (ATAC-seq) using a novel FXR knockout mouse model (Fxrex5Δ: Nr1h4ex5Δ/ex5Δ) generated through CRISPR/Cas9. Consistent with previous Fxr knockout models, we found that Fxrex5Δ mice develop late-onset HCC associated with increased serum and hepatic BAs. FXR deletion was associated with a dramatic loss of chromatin accessibility, primarily at promoter-associated transcription factor binding sites. Importantly, several genes involved in BA biosynthesis and circadian rhythm were downregulated following loss of FXR, also displayed reduced chromatin accessibility at their promoter regions. Altogether, these findings suggest that FXR helps to maintain a transcriptionally active state by regulating chromatin accessibility through its binding and recruitment of transcription factors and coactivators.

2.
Cancers (Basel) ; 14(3)2022 Feb 01.
Article in English | MEDLINE | ID: mdl-35159032

ABSTRACT

DNA methylation is an essential covalent modification that is required for growth and development. Once considered to be a relatively stable epigenetic mark, many studies have established that DNA methylation is dynamic. The 5-methylcytosine (5-mC) mark can be removed through active DNA demethylation in which 5-mC is converted to an unmodified cytosine through an oxidative pathway coupled to base excision repair (BER). The BER enzyme Thymine DNA Glycosylase (TDG) plays a key role in active DNA demethylation by excising intermediates of 5-mC generated by this process. TDG acts as a key player in transcriptional regulation through its interactions with various nuclear receptors and transcription factors, in addition to its involvement in classical BER and active DNA demethylation, which serve to protect the stability of the genome and epigenome, respectively. Recent animal studies have identified a connection between the loss of Tdg and the onset of tumorigenesis. In this review, we summarize the recent findings on TDG's function as a transcriptional regulator as well as the physiological relevance of TDG and active DNA demethylation in cancer.

3.
J Clin Invest ; 131(4)2021 02 15.
Article in English | MEDLINE | ID: mdl-33444292

ABSTRACT

DREAM (Dp, Rb-like, E2F, and MuvB) is a transcriptional repressor complex that regulates cell proliferation, and its loss causes neonatal lethality in mice. To investigate DREAM function in adult mice, we used an assembly-defective p107 protein and conditional deletion of its redundant family member p130. In the absence of DREAM assembly, mice displayed shortened survival characterized by systemic amyloidosis but no evidence of excessive cellular proliferation. Amyloid deposits were found in the heart, liver, spleen, and kidneys but not the brain or bone marrow. Using laser-capture microdissection followed by mass spectrometry, we identified apolipoproteins as the most abundant components of amyloids. Intriguingly, apoA-IV was the most detected amyloidogenic protein in amyloid deposits, suggesting apoA-IV amyloidosis (AApoAIV). AApoAIV is a recently described form, whereby WT apoA-IV has been shown to predominate in amyloid plaques. We determined by ChIP that DREAM directly regulated Apoa4 and that the histone variant H2AZ was reduced from the Apoa4 gene body in DREAM's absence, leading to overexpression. Collectively, we describe a mechanism by which epigenetic misregulation causes apolipoprotein overexpression and amyloidosis, potentially explaining the origins of nongenetic amyloid subtypes.


Subject(s)
Amyloid/metabolism , Apolipoproteins A/metabolism , Immunoglobulin Light-chain Amyloidosis/metabolism , Multiprotein Complexes/immunology , Retinoblastoma-Like Protein p107/deficiency , Amyloid/genetics , Animals , Apolipoproteins A/genetics , Immunoglobulin Light-chain Amyloidosis/genetics , Immunoglobulin Light-chain Amyloidosis/pathology , Mice , Mice, Knockout , Multiprotein Complexes/genetics , Organ Specificity/genetics , Retinoblastoma-Like Protein p107/metabolism
4.
Mol Cell Oncol ; 7(4): 1768819, 2020.
Article in English | MEDLINE | ID: mdl-32944627

ABSTRACT

In a recent publication, we demonstrated that conditional deletion of the gene encoding thymine DNA glycosylase (TDG) leads to a late onset of hepatocellular carcinoma (HCC). TDG loss causes disruption in active DNA demethylation in the liver and dysregulation of the farnesoid X receptor and small heterodimer partner (FXR-SHP) regulatory cascade. This leads to a loss of bile acid and glucose homeostasis, which predisposes mice to HCC.

5.
Cell Rep ; 31(1): 107475, 2020 04 07.
Article in English | MEDLINE | ID: mdl-32268085

ABSTRACT

Thymine DNA glycosylase (TDG) is a nuclear receptor coactivator that plays an essential role in the maintenance of epigenetic stability in cells. Here, we demonstrate that the conditional deletion of TDG in adult mice results in a male-predominant onset of hepatocellular carcinoma (HCC). TDG loss leads to a prediabetic state, as well as bile acid (BA) accumulation in the liver and serum of male mice. Consistent with these data, TDG deletion led to dysregulation of the farnesoid X receptor (FXR) and small heterodimer partner (SHP) regulatory cascade in the liver. FXR and SHP are tumor suppressors of HCC and play an essential role in BA and glucose homeostasis. These results indicate that TDG functions as a tumor suppressor of HCC by regulating a transcriptional program that protects against the development of glucose intolerance and BA accumulation in the liver.


Subject(s)
Bile Acids and Salts/metabolism , Carcinoma, Hepatocellular/physiopathology , Thymine DNA Glycosylase/metabolism , Animals , Bile Acids and Salts/genetics , Carcinoma, Hepatocellular/metabolism , Female , Glucose/metabolism , Hep G2 Cells , Homeostasis , Humans , Liver/pathology , Liver Neoplasms/metabolism , Liver Neoplasms/physiopathology , Male , Mice , Mice, Inbred C57BL , Receptors, Cytoplasmic and Nuclear/metabolism , Thymine DNA Glycosylase/physiology
6.
Lab Invest ; 100(3): 400-413, 2020 03.
Article in English | MEDLINE | ID: mdl-31570773

ABSTRACT

TBX3 is a member of the highly conserved family of T-box transcription factors involved in embryogenesis, organogenesis and tumor progression. While the functional role of TBX3 in tumorigenesis has been widely studied, less is known about the specific functions of the different isoforms (TBX3iso1 and TBX3iso2) which differ in their DNA-binding domain. We therefore sought to investigate the functional consequence of this highly conserved splice event as it relates to TBX3-induced tumorigenesis. By utilizing a nude mouse xenograft model, we have identified differential tumorigenic potential between TBX3 isoforms, with TBX3iso1 overexpression more commonly associated with invasive carcinoma and high tumor vascularity. Transcriptional analysis of signaling pathways altered by TBX3iso1 and TBX3iso2 overexpression revealed significant differences in angiogenesis-related genes. Importantly, osteopontin (OPN), a cancer-associated secreted phosphoprotein, was significantly up-regulated with TBX3iso1 (but not TBX3iso2) overexpression. This pattern was observed across three non/weakly-tumorigenic breast cancer cell lines (21PT, 21NT, and MCF7). Up-regulation of OPN in TBX3iso1 overexpressing cells was associated with induction of hyaluronan synthase 2 (HAS2) expression and increased retention of hyaluronan in pericellular matrices. These transcriptional changes were accompanied by the ability to induce endothelial cell vascular channel formation by conditioned media in vitro, which could be inhibited through addition of an OPN neutralizing antibody. Within the TCGA breast cancer cohort, we identified an 8.1-fold higher TBX3iso1 to TBX3iso2 transcript ratio in tumors relative to control, and this ratio was positively associated with high-tumor grade and an aggressive molecular subtype. Collectively, the described changes involving TBX3iso1-dependent promotion of angiogenesis may thus serve as an adaptive mechanism within breast cancer cells, potentially explaining differences in tumor formation rates between TBX3 isoforms in vivo. This study is the first of its kind to report significant functional differences between the two TBX3 isoforms, both in vitro and in vivo.


Subject(s)
Breast Neoplasms/metabolism , Neovascularization, Pathologic/metabolism , Protein Isoforms , T-Box Domain Proteins , Animals , Breast Neoplasms/pathology , Cell Line, Tumor , Female , Humans , Mice , Mice, Nude , Neovascularization, Pathologic/pathology , Osteopontin/metabolism , Protein Isoforms/chemistry , Protein Isoforms/genetics , Protein Isoforms/metabolism , T-Box Domain Proteins/chemistry , T-Box Domain Proteins/genetics , T-Box Domain Proteins/metabolism
7.
Methods Mol Biol ; 2019: 129-141, 2019.
Article in English | MEDLINE | ID: mdl-31359394

ABSTRACT

Methylase-assisted bisulfite sequencing (MAB-seq) is a derivatization technique to evaluate the presence of 5-formylcytosine (5-fC) and 5-carboxylcytosine (5-caC) at base-pair resolution. Although MAB-seq was originally designed to study these metabolites under steady-state conditions, we have developed an alternative protocol to evaluate the dynamics of 5-fC/5-caC accumulation in response to agonists, such as all-trans retinoic acid (ATRA). In addition, this protocol utilizes a lower quantity of the M.SssI enzyme without compromising methylation efficiency and requires less bench time. Herein, we describe the use of MAB-seq assay to evaluate the generation of 5-fC/5-caC in response to ATRA in mouse embryonic fibroblasts, using the hypermethylated in cancer 1 (Hic1) locus as a model system.


Subject(s)
5-Methylcytosine/metabolism , Fibroblasts/metabolism , Kruppel-Like Transcription Factors/genetics , Sequence Analysis, DNA/methods , Tretinoin/pharmacology , Animals , Cytosine/analogs & derivatives , Cytosine/metabolism , DNA Methylation , Fibroblasts/physiology , Kruppel-Like Transcription Factors/metabolism , Methyltransferases/metabolism , Mice , Molecular Structure
8.
J Pathol ; 248(2): 191-203, 2019 06.
Article in English | MEDLINE | ID: mdl-30697731

ABSTRACT

The acquisition of cellular invasiveness by breast epithelial cells and subsequent transition from ductal carcinoma in situ (DCIS) to invasive breast cancer is a critical step in breast cancer progression. Little is known about the molecular dynamics governing this transition. We have previously shown that overexpression of the transcriptional regulator TBX3 in DCIS-like cells increases survival, growth, and invasiveness. To explore this mechanism further and assess direct transcriptional targets of TBX3 in a high-resolution, isoform-specific context, we conducted genome-wide chromatin-immunoprecipitation (ChIP) arrays coupled with transcriptomic analysis. We show that TBX3 regulates several epithelial-mesenchymal transition (EMT)-related genes, including SLUG and TWIST1. Importantly, we demonstrate that TBX3 is a direct regulator of SLUG expression, and SLUG expression is required for TBX3-induced migration and invasion. Assessing TBX3 by immunohistochemistry in early-stage (stage 0 and stage I) breast cancers revealed high expression in low-grade lesions. Within a second independent early-stage non-high-grade cohort, we observed an association between TBX3 level in the DCIS and size of the invasive focus. Additionally, there was a positive correlation between TBX3 and SLUG, and TBX3 and TWIST1 in the invasive carcinoma. Pathway analysis revealed altered expression of several proteases and their inhibitors, consistent with the ability to degrade basement membrane in vivo. These findings strongly suggest the involvement of TBX3 in the promotion of invasiveness and progression of early-stage pre-invasive breast cancer to invasive carcinoma through the low-grade molecular pathway. © 2019 The Authors. The Journal of Pathology published by John Wiley & Sons Ltd on behalf of Pathological Society of Great Britain and Ireland.


Subject(s)
Breast Neoplasms/metabolism , Carcinoma, Intraductal, Noninfiltrating/metabolism , Epithelial-Mesenchymal Transition , Snail Family Transcription Factors/metabolism , T-Box Domain Proteins/metabolism , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Carcinoma, Intraductal, Noninfiltrating/genetics , Carcinoma, Intraductal, Noninfiltrating/pathology , Cell Line, Tumor , Cell Movement , Disease Progression , Female , Gene Expression Regulation, Neoplastic , Humans , Middle Aged , Neoplasm Grading , Neoplasm Invasiveness , Neoplasm Staging , Signal Transduction , Snail Family Transcription Factors/genetics , T-Box Domain Proteins/genetics , Up-Regulation
9.
Epigenetics Chromatin ; 11(1): 5, 2018 01 29.
Article in English | MEDLINE | ID: mdl-29378668

ABSTRACT

BACKGROUND: The estrogen receptor (ER) is a ligand-dependant transcription factor expressed in many breast cancers and is the target of many endocrine-based cancer therapies. Genome-wide studies have shown that the ER binds to gene-specific enhancer regions in response to ß-estradiol (E2) which undergo transcription producing noncoding enhancer RNA (eRNA). While eRNAs are important for transcriptional activation of neighboring genes, the mechanism remains poorly understood. RESULTS: Using ChIP-Seq we generate a global profile of thymine DNA glycosylase (TDG), an ER coactivator that plays an essential role in DNA demethylation, in response to E2 in the MCF7 breast cancer cell line. Remarkably, we found that in response to E2 TDG localized to enhancers which also recruit ERα, RNA Pol II and other coregulators and which are marked by histone modifications indicative of active enhancers. Importantly, depletion of TDG inhibits E2-mediated transcription of eRNAs and transcription of ER-target genes. Functionally, we find that TDG both sensitizes MCF7 cells to tamoxifen-mediated cytostasis and increases migration and invasion of MCF7 cells. CONCLUSIONS: Taken together we find that TDG plays a central role in mediating transcription at a subset of enhancers and governs how MCF7 cells respond to both estrogenic and anti-estrogenic compounds and may be an effective therapeutic target.


Subject(s)
Breast Neoplasms/genetics , Enhancer Elements, Genetic , Estradiol/pharmacology , Receptors, Estrogen/metabolism , Sequence Analysis, RNA/methods , Tamoxifen/pharmacology , Breast Neoplasms/drug therapy , Breast Neoplasms/metabolism , Cell Movement/drug effects , Cell Proliferation/drug effects , Cell Survival/drug effects , Chromatin Immunoprecipitation , DNA Methylation , Drug Synergism , Female , Humans , MCF-7 Cells , RNA Polymerase II/genetics , Thymine DNA Glycosylase/genetics , Whole Genome Sequencing/methods
10.
Cell Rep ; 19(8): 1685-1697, 2017 05 23.
Article in English | MEDLINE | ID: mdl-28538185

ABSTRACT

Retinoic acid (RA) plays important roles in development, growth, and homeostasis through regulation of the nuclear receptors for RA (RARs). Herein, we identify Hypermethylated in Cancer 1 (Hic1) as an RA-inducible gene. HIC1 encodes a tumor suppressor, which is often silenced by promoter hypermethylation in cancer. Treatment of cells with an RAR agonist causes a rapid recruitment of an RAR/RXR complex consisting of TDG, the lysine acetyltransferase CBP, and TET 1/2 to the Hic1 promoter. Complex binding coincides with a transient accumulation of 5fC/5caC and concomitant upregulation of Hic1 expression, both of which are TDG dependent. Furthermore, conditional deletion of Tdg in vivo is associated with Hic1 silencing and DNA hypermethylation of the Hic1 promoter. These findings suggest that the catalytic and scaffolding activities of TDG are essential for RA-dependent gene expression and provide important insights into the mechanisms underlying targeting of TET-TDG complexes.


Subject(s)
DNA Demethylation , DNA-Binding Proteins/metabolism , Proto-Oncogene Proteins/metabolism , Receptors, Retinoic Acid/metabolism , Thymine DNA Glycosylase/metabolism , Animals , DNA Demethylation/drug effects , Dioxygenases , Gene Deletion , Gene Silencing/drug effects , Kruppel-Like Transcription Factors , Membrane Proteins/metabolism , Mice, Transgenic , Phosphoproteins/metabolism , Tretinoin/pharmacology
11.
J Immunol ; 189(7): 3347-54, 2012 Oct 01.
Article in English | MEDLINE | ID: mdl-22956576

ABSTRACT

B cell acute lymphoblastic leukemia (B-ALL) is frequently associated with mutations or chromosomal translocations of genes encoding transcription factors. Conditional deletion of genes encoding the E26-transformation-specific transcription factors, PU.1 and Spi-B, in B cells (ΔPB mice) leads to B-ALL in mice at 100% incidence rate and with a median survival of 21 wk. We hypothesized that PU.1 and Spi-B may redundantly activate transcription of genes encoding tumor suppressors in the B cell lineage. Characterization of aging ΔPB mice showed that leukemia cells expressing IL-7R were found in enlarged thymuses. IL-7R-expressing B-ALL cells grew in culture in response to IL-7 and could be maintained as cell lines. Cultured ΔPB cells expressed reduced levels of B cell linker protein (BLNK), a known tumor suppressor gene, compared with controls. The Blnk promoter contained a predicted PU.1 and/or Spi-B binding site that was required for promoter activity and occupied by PU.1 and/or Spi-B as determined by chromatin immunoprecipitation. Restoration of BLNK expression in cultured ΔPB cells opposed IL-7-dependent proliferation and induced early apoptosis. We conclude that the tumor suppressor BLNK is a target of transcriptional activation by PU.1 and Spi-B in the B cell lineage.


Subject(s)
Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , B-Lymphocytes/immunology , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/genetics , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/metabolism , Proto-Oncogene Proteins c-ets/physiology , Proto-Oncogene Proteins/physiology , Trans-Activators/physiology , Transcriptional Activation/immunology , Adaptor Proteins, Signal Transducing/immunology , Animals , B-Lymphocytes/metabolism , B-Lymphocytes/pathology , Cell Line, Tumor , Cell Lineage/genetics , Cell Lineage/immunology , Female , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , NIH 3T3 Cells , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/immunology , Promoter Regions, Genetic/immunology , Protein Binding/genetics , Protein Binding/immunology , Receptors, Antigen, B-Cell/physiology
12.
Mol Cell ; 46(5): 636-49, 2012 Jun 08.
Article in English | MEDLINE | ID: mdl-22560925

ABSTRACT

In this study we examine the mechanisms of dynamic DNA methylation of the p15(ink4b) tumor suppressor gene. Using conventional ChIP and ChiPseq, we identify the p15(ink4b) promoter as a target for the ZNF217 oncogene, the CoREST complex, and DNMT3A. Treatment of cells with TGF-ß triggers active demethylation involving loss of ZNF217/CoREST/DNMT3A and the corecruitment of SMAD2/3, CBP, and the DNA glycosylase TDG. Knockdown of TDG, or its functional homolog MBD4, prevents TGF-ß-dependent demethylation of p15(ink4b). DNA immunoprecipitation of 5mC and 5hmC indicates that 5mC undergoes conversion to 5hmC prior to activation of p15(ink4b). Remarkably, overexpression of ZNF217 inhibits active demethylation and expression of the p15(ink4b) gene by preventing recruitment of SMAD2/3 and TDG. These findings suggest that active demethylation is essential for regulating a subset of TGF-ß-dependent genes. Importantly, disruption of active demethylation by the ZNF217 oncogene may be a paradigm for other oncogenic signals on DNA methylation dynamics.


Subject(s)
Cyclin-Dependent Kinase Inhibitor p15/metabolism , DNA Methylation , Nerve Tissue Proteins/physiology , Repressor Proteins/physiology , Trans-Activators/physiology , Transforming Growth Factor beta/physiology , Cell Cycle/genetics , Cell Line, Tumor , Co-Repressor Proteins , Cyclin-Dependent Kinase Inhibitor p15/genetics , Gene Expression Regulation, Neoplastic , Humans , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Promoter Regions, Genetic , Repressor Proteins/genetics , Repressor Proteins/metabolism , Trans-Activators/genetics , Trans-Activators/metabolism , Transforming Growth Factor beta/metabolism
13.
BMC Biochem ; 12: 50, 2011 Sep 13.
Article in English | MEDLINE | ID: mdl-21914189

ABSTRACT

BACKGROUND: Steroid Receptor coactivator 3(SRC3) is an oncogene and a member of the SRC family of nuclear receptor coactivator proteins that mediate the transcriptional effects of nuclear hormone receptors as well as other transcription factors. RESULTS: We have used protein purification and mass spectrometry to identify the 53BP1 tumour suppressor as a novel SRC3-associated protein. Copurification was demonstrated using multiple antibodies, and was not dependent on DNA damage suggesting that SRC3 is not directly involved in the DNA damage response. However using chromatin immunoprecipitation(ChIP) and siRNA knockdown, we have demonstrated that both SRC3 and 53BP1 co-occupy the same region of the BRCA1 promoter and both are required for BRCA1 expression in HeLa cells. CONCLUSIONS: Our results suggest that both 53BP1 and SRC3 have a common function that converge at the BRCA1 promoter and possibly other genes important for DNA repair and genomic stability.


Subject(s)
Genes, BRCA1 , Intracellular Signaling Peptides and Proteins/metabolism , Nuclear Receptor Coactivator 3/metabolism , CREB-Binding Protein/metabolism , Chromatin Immunoprecipitation , DNA Damage , Gene Knockdown Techniques , HeLa Cells , Humans , Intracellular Signaling Peptides and Proteins/deficiency , Intracellular Signaling Peptides and Proteins/genetics , Mass Spectrometry , Nuclear Receptor Coactivator 3/deficiency , Nuclear Receptor Coactivator 3/genetics , Protein Transport , RNA, Small Interfering/genetics , Tumor Suppressor p53-Binding Protein 1
14.
J Immunol ; 185(12): 7374-84, 2010 Dec 15.
Article in English | MEDLINE | ID: mdl-21057087

ABSTRACT

Splenic B-2 cells can be divided into two major subsets: follicular (FO) and marginal zone (MZ) B cells. FO and MZ B cells are generated from immature transitional B cells. Few transcription factors have been identified that regulate FO B cell differentiation. The highly related proteins PU.1, Spi-B, and Spi-C are transcription factors of the E26-transformation-specific family and are important for B cell differentiation and function. To determine whether these proteins play a role in the differentiation of FO B cells, we performed a detailed analysis of splenic B cells in mice with inactivating mutations in the genes encoding PU.1 (Sfpi1) or Spi-B (Spib). Sfpi1(+/-) Spib(-/-) (PUB) mice had a 9-fold reduction in the frequency of CD23(+) FO B cells compared with that of wild-type mice. In contrast, PUB mice had a 2-fold increase in the frequency of MZ B cells that was confirmed by immunofluorescence staining. Expression of Spi-C in Eµ-Spi-C transgenic PUB mice partially rescued frequencies of CD23(+) B cells. Gene expression analysis, in vitro reporter assays, and chromatin immunoprecipitation experiments showed that transcription of the Fcer2a gene encoding CD23 is activated by PU.1, Spi-B, and Spi-C. These results demonstrate that FO B cell differentiation is regulated by the E26-transformation-specific transcription factors PU.1, Spi-B, and Spi-C.


Subject(s)
B-Lymphocytes/immunology , Cell Differentiation/immunology , DNA-Binding Proteins/immunology , Proto-Oncogene Proteins c-ets/immunology , Proto-Oncogene Proteins/immunology , Trans-Activators/immunology , Animals , B-Lymphocytes/metabolism , Cell Differentiation/genetics , Cell Line, Tumor , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Female , Male , Mice , Mice, Knockout , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-ets/genetics , Proto-Oncogene Proteins c-ets/metabolism , Receptors, IgE/genetics , Receptors, IgE/immunology , Receptors, IgE/metabolism , Trans-Activators/genetics , Trans-Activators/metabolism
15.
Cell Stem Cell ; 6(2): 153-66, 2010 Feb 05.
Article in English | MEDLINE | ID: mdl-20144788

ABSTRACT

Polycomb group (PcG) proteins are conserved epigenetic transcriptional repressors that control numerous developmental gene expression programs and have recently been implicated in modulating embryonic stem cell (ESC) fate. We identified the PcG protein PCL2 (polycomb-like 2) in a genome-wide screen for regulators of self-renewal and pluripotency and predicted that it would play an important role in mouse ESC-fate determination. Using multiple biochemical strategies, we provide evidence that PCL2 is a Polycomb Repressive Complex 2 (PRC2)-associated protein in mouse ESCs. Knockdown of Pcl2 in ESCs resulted in heightened self-renewal characteristics, defects in differentiation, and altered patterns of histone methylation. Integration of global gene expression and promoter occupancy analyses allowed us to identify PCL2 and PRC2 transcriptional targets and draft regulatory networks. We describe the role of PCL2 in both modulating transcription of ESC self-renewal genes in undifferentiated ESCs as well as developmental regulators during early commitment and differentiation.


Subject(s)
Cell Differentiation , Cellular Reprogramming , Embryonic Stem Cells/cytology , Embryonic Stem Cells/metabolism , Gene Regulatory Networks , Repressor Proteins/metabolism , Animals , Cell Line , Down-Regulation , Gene Expression Profiling , Genome-Wide Association Study , Histones/metabolism , Methylation , Mice , Pluripotent Stem Cells/cytology , Pluripotent Stem Cells/metabolism , Polycomb Repressive Complex 2 , Polycomb-Group Proteins , Protein Binding , Repressor Proteins/genetics
16.
Nucleic Acids Res ; 38(4): 1135-48, 2010 Mar.
Article in English | MEDLINE | ID: mdl-19966277

ABSTRACT

CpG dinucleotides are mutational hotspots associated with cancer and genetic diseases. Thymine DNA glycosylase (TDG) plays an integral role in CpG maintenance by excising mispaired thymine and uracil in a CpG context and also participates in transcriptional regulation via gene-specific CpG demethylation and functional interactions with the transcription machinery. Here, we report that protein kinase C alpha (PKCalpha) interacts with TDG and phosphorylates amino-terminal serine residues adjacent to lysines acetylated by CREB-binding protein (CBP) and p300 (CBP/p300). We establish that acetylation and phosphorylation are mutually exclusive, and their interplay dramatically alters the DNA mispair-processing functions of TDG. Remarkably, acetylation of the amino-terminal region abrogates high-affinity DNA binding and selectively prevents processing of G:T mispairs. In contrast, phosphorylation does not markedly alter DNA interactions, but may preserve G:T processing in vivo by preventing CBP-mediated acetylation. Mutational analysis suggests that the acetyl-acceptor lysines are not directly involved in contacting DNA, but may constitute a conformationally sensitive interface that modulates DNA interactions. These findings reveal opposing roles of CBP/p300 and PKCalpha in regulating the DNA repair functions of TDG and suggest that the interplay of these modifications in vivo may be critically important in the maintenance of CpG dinucleotides and epigenetic regulation.


Subject(s)
DNA Repair , Thymine DNA Glycosylase/metabolism , Acetylation , Animals , Cell Line , Humans , Mice , NIH 3T3 Cells , Phosphorylation , Protein Kinase C-alpha/metabolism , Tetradecanoylphorbol Acetate/pharmacology , Thymine DNA Glycosylase/chemistry
17.
Methods Mol Biol ; 523: 235-47, 2009.
Article in English | MEDLINE | ID: mdl-19381935

ABSTRACT

Protein arginine methylation has emerged as an important mechanism for regulating the functions of proteins involved in diverse aspects of gene regulation such as transcriptional activation and repression, mRNA processing and nuclear-cytoplasmic shuttling. This modification is catalyzed by the PRMT family of enzymes which utilize intracellular S-adenosyl methionine as a cofactor to dimethylate-specific arginines found within many target proteins.The establishment of in vitro biochemical assays as well as the development of modification-specific antibodies, and more recently mass spectrometry, have increased our understanding of the mechanism of catalysis of the PRMT family of enzymes. In the following discussion, we present some of the more commonly used in vivo and in vitro techniques which can be utilized to study the mechanism of arginine methylation and its role in transcription.


Subject(s)
Arginine/metabolism , Biochemistry/methods , Transcription, Genetic , Animals , Antibody Specificity , Baculoviridae , Biological Assay , Biotinylation , Cell Extracts , Chromatin Immunoprecipitation , Histone Acetyltransferases/chemistry , Histone Acetyltransferases/metabolism , Humans , Mass Spectrometry , Methylation , Methyltransferases/metabolism , Nuclear Receptor Coactivator 3 , Peptides/metabolism , Protein-Arginine N-Methyltransferases/chemistry , Protein-Arginine N-Methyltransferases/metabolism , Recombinant Fusion Proteins/isolation & purification , Recombinant Fusion Proteins/metabolism , Staining and Labeling , Trans-Activators/chemistry , Trans-Activators/metabolism
18.
Nucleic Acids Res ; 37(4): 1095-106, 2009 Mar.
Article in English | MEDLINE | ID: mdl-19129215

ABSTRACT

The human adenovirus type 5 (HAdV-5) E1A 13S oncoprotein is a potent regulator of gene expression and is used extensively as a model for transcriptional activation. It possesses two independent transcriptional activation domains located in the N-terminus/conserved region (CR) 1 and CR3. The protein acetyltransferase p300 was previously identified by its association with the N-terminus/CR1 portion of E1A and this association is required for oncogenic transformation by E1A. We report here that transcriptional activation by 13S E1A is inhibited by co-expression of sub-stoichiometric amounts of the smaller 12S E1A isoform, which lacks CR3. Transcriptional inhibition by E1A 12S maps to the N-terminus and correlates with the ability to bind p300/CBP, suggesting that E1A 12S is sequestering this limiting factor from 13S E1A. This is supported by the observation that the repressive effect of E1A 12S is reversed by expression of exogenous p300 or CBP, but not by a CBP mutant lacking actyltransferase activity. Furthermore, we show that transcriptional activation by 13S E1A is greatly reduced by siRNA knockdown of p300 and that CR3 binds p300 independently of the well-characterized N-terminal/CR1-binding site. Importantly, CR3 is also required to recruit p300 to the adenovirus E4 promoter during infection. These results identify a new functionally significant interaction between E1A CR3 and the p300/CBP acetyltransferases, expanding our understanding of the mechanism by which this potent transcriptional activator functions.


Subject(s)
Adenovirus E1A Proteins/metabolism , Transcriptional Activation , p300-CBP Transcription Factors/metabolism , Adenovirus E1A Proteins/chemistry , Adenovirus E1A Proteins/genetics , Adenovirus E4 Proteins/genetics , Amino Acid Sequence , Cell Line , Conserved Sequence , E1A-Associated p300 Protein/antagonists & inhibitors , E1A-Associated p300 Protein/genetics , Humans , Mutation , Promoter Regions, Genetic , Recombinant Fusion Proteins/metabolism
19.
Mol Cell Biol ; 28(19): 6066-77, 2008 Oct.
Article in English | MEDLINE | ID: mdl-18625718

ABSTRACT

The ZNF217 oncoprotein is a constituent of a core transcriptional complex that includes CoREST, histone deacetylase 1/2, lysine demethylase 1, and the C-terminal binding protein 1/2. We have combined genome-wide expression profiling and chromatin immunoprecipitation with directed selection and ligation (ChIP-DSL) to identify a subset of genes directly regulated by ZNF217. Our results establish p15(ink4b) as a direct target of the ZNF217 complex. Downregulation of ZNF217 in MCF-7 breast cancer cells resulted in a dramatic increase in p15(ink4b) expression and coincided with increases in dimethylation of H3-K4 and, surprisingly, a decrease in K9/K14-H3 acetylation. Stimulation of HaCaT cells with transforming growth factor beta (TGF-beta) resulted in a release of ZNF217 and a concomitant binding of SMAD2 to the proximal promoter, which preceded increases in ink4b protein expression. Furthermore, the changes in chromatin marks at the p15(ink4b) promoter following TGF-beta stimulation were similar to those observed following ZNF217 downregulation. Collectively, these results establish the ZNF217 complex as a novel negative regulator of the p15(ink4b) gene and may constitute an important link between amplification of ZNF217 and the loss of TGF-beta responsiveness in breast cancer.


Subject(s)
Cyclin-Dependent Kinase Inhibitor p15/genetics , Trans-Activators/metabolism , Breast Neoplasms/metabolism , Cell Line, Tumor , Co-Repressor Proteins , Cyclin-Dependent Kinase Inhibitor p15/metabolism , DNA-Binding Proteins/metabolism , Genomics , Humans , Nerve Tissue Proteins/metabolism , Promoter Regions, Genetic , Repressor Proteins/metabolism , Trans-Activators/isolation & purification , Transforming Growth Factor beta/metabolism
20.
J Virol ; 82(17): 8476-86, 2008 Sep.
Article in English | MEDLINE | ID: mdl-18524818

ABSTRACT

C-terminal binding protein (CtBP) binds to adenovirus early region 1A (AdE1A) through a highly conserved PXDLS motif close to the C terminus. We now have demonstrated that CtBP1 also interacts directly with the transcriptional activation domain (conserved region 3 [CR3]) of adenovirus type 5 E1A (Ad5E1A) and requires the integrity of the entire CR3 region for optimal binding. The interaction appears to be at least partially mediated through a sequence ((161)RRNTGDP(167)) very similar to a recently characterized novel CtBP binding motif in ZNF217 as well as other regions of CR3. Using reporter assays, we further demonstrated that CtBP1 represses Ad5E1A CR3-dependent transcriptional activation. Ad5E1A also appears to be recruited to the E-cadherin promoter through its interaction with CtBP. Significantly, Ad5E1A, CtBP1, and ZNF217 form a stable complex which requires CR3 and the PLDLS motif. It has been shown that Ad513SE1A, containing the CR3 region, is able to overcome the transcriptional repressor activity of a ZNF217 polypeptide fragment in a GAL4 reporter assay through recruitment of CtBP1. These results suggest a hitherto-unsuspected complexity in the association of Ad5E1A with CtBP, with the interaction resulting in transcriptional activation by recruitment of CR3-bound factors to CtBP1-containing complexes.


Subject(s)
Adenovirus E1A Proteins/chemistry , Adenovirus E1A Proteins/metabolism , Adenovirus E1A Proteins/genetics , Alcohol Oxidoreductases/metabolism , Amino Acid Motifs , Amino Acid Sequence , Antibodies, Monoclonal/metabolism , Binding Sites , Carcinoma, Small Cell/metabolism , Cell Line, Tumor , Conserved Sequence , DNA-Binding Proteins/metabolism , Genes, Reporter , Glutathione Transferase/metabolism , Green Fluorescent Proteins/metabolism , HCT116 Cells , HT29 Cells , HeLa Cells , Humans , Luciferases/analysis , Lung Neoplasms/metabolism , Molecular Sequence Data , Mutation , Osteosarcoma/metabolism , Plasmids , Protein Binding , Protein Structure, Tertiary , RNA, Small Interfering/metabolism , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/classification , Recombinant Fusion Proteins/metabolism , Time Factors , Transfection
SELECTION OF CITATIONS
SEARCH DETAIL
...