Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
1.
PLoS Genet ; 17(1): e1009277, 2021 01.
Article in English | MEDLINE | ID: mdl-33411704

ABSTRACT

The nuclear protein CCCTC-binding factor (CTCF) has diverse roles in chromatin architecture and gene regulation. Functionally, CTCF associates with thousands of genomic sites and interacts with proteins, such as cohesin, or non-coding RNAs to facilitate specific transcriptional programming. In this study, we examined CTCF during the cellular stress response in human primary cells using immune-blotting, quantitative real time-PCR, chromatin immunoprecipitation-sequence (ChIP-seq) analysis, mass spectrometry, RNA immunoprecipitation-sequence analysis (RIP-seq), and Airyscan confocal microscopy. Unexpectedly, we found that CTCF is exquisitely sensitive to diverse forms of stress in normal patient-derived human mammary epithelial cells (HMECs). In HMECs, a subset of CTCF protein forms complexes that localize to Serine/arginine-rich splicing factor (SC-35)-containing nuclear speckles. Upon stress, this species of CTCF protein is rapidly downregulated by changes in protein stability, resulting in loss of CTCF from SC-35 nuclear speckles and changes in CTCF-RNA interactions. Our ChIP-seq analysis indicated that CTCF binding to genomic DNA is largely unchanged. Restoration of the stress-sensitive pool of CTCF protein abundance and re-localization to nuclear speckles can be achieved by inhibition of proteasome-mediated degradation. Surprisingly, we observed the same characteristics of the stress response during neuronal differentiation of human pluripotent stem cells (hPSCs). CTCF forms stress-sensitive complexes that localize to SC-35 nuclear speckles during a specific stage of neuronal commitment/development but not in differentiated neurons. We speculate that these particular CTCF complexes serve a role in RNA processing that may be intimately linked with specific genes in the vicinity of nuclear speckles, potentially to maintain cells in a certain differentiation state, that is dynamically regulated by environmental signals. The stress-regulated activity of CTCF is uncoupled in persistently stressed, epigenetically re-programmed "variant" HMECs and certain cancer cell lines. These results reveal new insights into CTCF function in cell differentiation and the stress-response with implications for oxidative damage-induced cancer initiation and neuro-degenerative diseases.


Subject(s)
CCCTC-Binding Factor/genetics , DNA-Binding Proteins/genetics , Neoplasms/genetics , Neurodegenerative Diseases/genetics , Serine-Arginine Splicing Factors/genetics , Binding Sites , Cell Differentiation , Cell Line, Tumor , Chromatin , Chromosomes , Epigenesis, Genetic/genetics , Gene Expression Regulation , Genomics , Humans , Mammary Glands, Human/cytology , Mammary Glands, Human/metabolism , Neoplasms/pathology , Neurodegenerative Diseases/pathology , Neurons/metabolism , Neurons/pathology , Oxidative Stress/genetics , Pluripotent Stem Cells/metabolism , Pluripotent Stem Cells/pathology , Protein Binding , RNA Processing, Post-Transcriptional/genetics , Stress, Physiological/genetics
2.
Oncotarget ; 8(39): 64698-64713, 2017 Sep 12.
Article in English | MEDLINE | ID: mdl-29029387

ABSTRACT

Poly(ADP)ribosylation (PARylation) of the chromatin architectural protein CTCF is critical for CTCF-dependent regulation of chromatin boundary and insulator elements. Loss of CTCF PARylation results in epigenetic silencing of certain tumor suppressor genes through destabilization of nearby chromatin boundaries. We investigated the metabolic and mechanistic processes that regulate PARP-1-mediated CTCF PARylation in human cancer cell lines and discovered a key role for the expression and activity of ß-NAD+ salvage enzymes, NAMPT and NMNAT-1. These enzymes are downregulated in cells that exhibit reduced CTCF PARylation, resulting in a decreased concentration of nuclear ß-NAD+. In these cells, decreased NMNAT-1 expression is enforced by a proteasome-mediated feedback loop resulting in degradation of NMNAT-1, transcriptional repression of NAMPT, and suppression of PARP-1 activity. Interestingly, dePARylated CTCF is associated in a stable protein complex with PARP-1 and NMNAT-1 in cancer cells harboring silenced tumor suppressor genes. Although the metabolic context in these cells favors suppression of PARP-1 activity, CTCF PARylation can be restored by Protein Kinase C (PKC) signaling. PKC induces dissociation of the catalytically inactive PARP-1/NMNAT-1/CTCF protein complex and phosphorylation of NMNAT-1, which stimulates its proteasome-mediated degradation. Our findings suggest that CTCF PARylation is underpinned by a cellular metabolic context engendered by regulation of the ß-NAD+ salvage pathway in which NMNAT-1 acts as a rheostat to control localized ß-NAD+ synthesis at CTCF/PARP-1 complexes.

3.
Proc Natl Acad Sci U S A ; 112(7): E677-86, 2015 Feb 17.
Article in English | MEDLINE | ID: mdl-25646466

ABSTRACT

CCCTC-binding factor (CTCF) is a key regulator of nuclear chromatin structure and gene regulation. The impact of CTCF on transcriptional output is highly varied, ranging from repression to transcriptional pausing and transactivation. The multifunctional nature of CTCF may be directed solely through remodeling chromatin architecture. However, another hypothesis is that the multifunctional nature of CTCF is mediated, in part, through differential association with protein partners having unique functions. Consistent with this hypothesis, our mass spectrometry analyses of CTCF interacting partners reveal a previously undefined association with the transcription factor general transcription factor II-I (TFII-I). Biochemical fractionation of CTCF indicates that a distinct CTCF complex incorporating TFII-I is assembled on DNA. Unexpectedly, we found that the interaction between CTCF and TFII-I is essential for directing CTCF to the promoter proximal regulatory regions of target genes across the genome, particularly at genes involved in metabolism. At genes coregulated by CTCF and TFII-I, we find knockdown of TFII-I results in diminished CTCF binding, lack of cyclin-dependent kinase 8 (CDK8) recruitment, and an attenuation of RNA polymerase II phosphorylation at serine 5. Phenotypically, knockdown of TFII-I alters the cellular response to metabolic stress. Our data indicate that TFII-I directs CTCF binding to target genes, and in turn the two proteins cooperate to recruit CDK8 and enhance transcription initiation.


Subject(s)
Epigenesis, Genetic , Genome, Human , Promoter Regions, Genetic , Repressor Proteins/genetics , Transcription Factors/physiology , CCCTC-Binding Factor , Cell Line, Tumor , Gene Knockdown Techniques , Humans , Phosphorylation
4.
Proc Natl Acad Sci U S A ; 111(44): E4726-35, 2014 Nov 04.
Article in English | MEDLINE | ID: mdl-25339441

ABSTRACT

The acute cellular response to stress generates a subpopulation of reversibly stress-tolerant cells under conditions that are lethal to the majority of the population. Stress tolerance is attributed to heterogeneity of gene expression within the population to ensure survival of a minority. We performed whole transcriptome sequencing analyses of metastatic human breast cancer cells subjected to the chemotherapeutic agent paclitaxel at the single-cell and population levels. Here we show that specific transcriptional programs are enacted within untreated, stressed, and drug-tolerant cell groups while generating high heterogeneity between single cells within and between groups. We further demonstrate that drug-tolerant cells contain specific RNA variants residing in genes involved in microtubule organization and stabilization, as well as cell adhesion and cell surface signaling. In addition, the gene expression profile of drug-tolerant cells is similar to that of untreated cells within a few doublings. Thus, single-cell analyses reveal the dynamics of the stress response in terms of cell-specific RNA variants driving heterogeneity, the survival of a minority population through generation of specific RNA variants, and the efficient reconversion of stress-tolerant cells back to normalcy.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Breast Neoplasms , Drug Resistance, Neoplasm , Paclitaxel/pharmacology , RNA, Neoplasm , Sequence Analysis, RNA , Transcription, Genetic , Breast Neoplasms/drug therapy , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Line, Tumor , Drug Resistance, Neoplasm/drug effects , Drug Resistance, Neoplasm/genetics , Female , Humans , RNA, Neoplasm/biosynthesis , RNA, Neoplasm/genetics , Transcription, Genetic/drug effects , Transcription, Genetic/genetics
5.
Elife ; 3: e01776, 2014 Apr 29.
Article in English | MEDLINE | ID: mdl-24843008

ABSTRACT

Deregulated expression of COX-2 has been causally linked to development, progression, and outcome of several types of human cancer. We describe a novel fundamental level of transcriptional control of COX-2 expression. Using primary human mammary epithelial cells and monocyte/macrophage cell lines, we show that the chromatin boundary/insulator factor CTCF establishes an open chromatin domain and induces expression of a long non-coding RNA within the upstream promoter region of COX-2. Upon induction of COX-2 expression, the lncRNA associates with p50, a repressive subunit of NF-κB, and occludes it from the COX-2 promoter, potentially facilitating interaction with activation-competent NF-κB p65/p50 dimers. This enables recruitment of the p300 histone acetyltransferase, a domain-wide increase in histone acetylation and assembly of RNA Polymerase II initiation complexes. Our findings reveal an unexpected mechanism of gene control by lncRNA-mediated repressor occlusion and identify the COX-2-lncRNA, PACER, as a new potential target for COX-2-modulation in inflammation and cancer.DOI: http://dx.doi.org/10.7554/eLife.01776.001.


Subject(s)
Cyclooxygenase 2/biosynthesis , Epithelial Cells/physiology , Gene Expression Regulation , Monocytes/physiology , NF-kappa B p50 Subunit/antagonists & inhibitors , RNA, Untranslated/metabolism , Transcription, Genetic , Cells, Cultured , Cyclooxygenase 2/genetics , Humans
6.
Mol Cell ; 50(4): 552-64, 2013 May 23.
Article in English | MEDLINE | ID: mdl-23706820

ABSTRACT

Cellular stress results in profound changes in RNA and protein synthesis. How cells integrate this intrinsic, p53-centered program with extracellular signals is largely unknown. We demonstrate that TGF-ß1 signaling interferes with the stress response through coordinate transcriptional and translational repression of p53 levels, which reduces p53-activated transcription, and apoptosis in precancerous cells. Mechanistically, E2F-4 binds constitutively to the TP53 gene and induces transcription. TGF-ß1-activated Smads are recruited to a composite Smad/E2F-4 element by an E2F-4/p107 complex that switches to a Smad corepressor, which represses TP53 transcription. TGF-ß1 also causes dissociation of ribosomal protein RPL26 and elongation factor eEF1A from p53 mRNA, thereby reducing p53 mRNA association with polyribosomes and p53 translation. TGF-ß1 signaling is dominant over stress-induced transcription and translation of p53 and prevents stress-imposed downregulation of Smad proteins. Thus, crosstalk between the TGF-ß and p53 pathways defines a major node of regulation in the cellular stress response, enhancing drug resistance.


Subject(s)
Gene Expression Regulation/drug effects , Stress, Physiological/drug effects , Transforming Growth Factor beta1/pharmacology , Tumor Suppressor Protein p53/genetics , Apoptosis/drug effects , Apoptosis/genetics , Base Sequence , Blotting, Western , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Cycle/drug effects , Cell Cycle/genetics , Cell Line , Cell Survival/drug effects , Cell Survival/genetics , Cells, Cultured , E2F4 Transcription Factor/genetics , E2F4 Transcription Factor/metabolism , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Humans , Immunohistochemistry , Mammary Glands, Human/cytology , Molecular Sequence Data , Promoter Regions, Genetic/genetics , Protein Binding/drug effects , RNA Interference , Reverse Transcriptase Polymerase Chain Reaction , Ribosomal Proteins/genetics , Ribosomal Proteins/metabolism , Signal Transduction/drug effects , Signal Transduction/genetics , Smad Proteins/genetics , Smad Proteins/metabolism , Stress, Physiological/genetics , Tumor Suppressor Protein p53/metabolism
7.
PLoS One ; 7(8): e42424, 2012.
Article in English | MEDLINE | ID: mdl-22879976

ABSTRACT

The CCCTC-binding factor CTCF is the only known vertebrate insulator protein and has been shown to regulate important developmental processes such as imprinting, X-chromosome inactivation and genomic architecture. In this study, we examined the role of CTCF in human embryonic stem cell (hESC) biology. We demonstrate that CTCF associates with several important pluripotency genes, including NANOG, SOX2, cMYC and LIN28 and is critical for hESC proliferation. CTCF depletion impacts expression of pluripotency genes and accelerates loss of pluripotency upon BMP4 induced differentiation, but does not result in spontaneous differentiation. We find that CTCF associates with the distal ends and internal sites of the co-regulated 160 kb NANOG-DPPA3-GDF3 locus. Each of these sites can function as a CTCF-dependent enhancer-blocking insulator in heterologous assays. In hESCs, CTCF exists in multisubunit protein complexes and can be poly(ADP)ribosylated. Known CTCF cofactors, such as Cohesin, differentially co-localize in the vicinity of specific CTCF binding sites within the NANOG locus. Importantly, the association of some cofactors and protein PARlation selectively changes upon differentiation although CTCF binding remains constant. Understanding how unique cofactors may impart specialized functions to CTCF at specific genomic locations will further illuminate its role in stem cell biology.


Subject(s)
Embryonic Stem Cells/metabolism , Repressor Proteins/genetics , Repressor Proteins/metabolism , Binding Sites , Biomarkers/metabolism , CCCTC-Binding Factor , Cell Differentiation/genetics , Cell Line , Chromosomal Proteins, Non-Histone , Embryonic Stem Cells/cytology , Enhancer Elements, Genetic/genetics , Gene Expression Regulation , Genetic Loci/genetics , Growth Differentiation Factor 3/metabolism , Homeodomain Proteins/metabolism , Humans , Models, Biological , Nanog Homeobox Protein , Pluripotent Stem Cells/cytology , Pluripotent Stem Cells/metabolism , Poly Adenosine Diphosphate Ribose/metabolism , Protein Binding/genetics , Proteins/metabolism
8.
Nat Rev Cancer ; 11(5): 375-82, 2011 05.
Article in English | MEDLINE | ID: mdl-21508974

ABSTRACT

Cancer cells rapidly evolve drug resistance through somatic evolution and, in order to continue growth in the metastatic phase, violate the organism-wide consensus of regulated growth and beneficial communal interactions. We suggest that there is a fundamental mechanistic connection between the rapid evolution of resistance to chemotherapy in cellular communities within malignant tissues and the rapid evolution of antibiotic resistance in bacterial communities. We propose that this evolution is the result of a programmed and collective stress response performed by interacting cells, and that, given this fundamental connection, studying bacterial communities can provide deeper insights into the dynamics of adaptation and the evolution of cells within tumours.


Subject(s)
Antineoplastic Agents/therapeutic use , Drug Resistance, Bacterial/physiology , Neoplasms/drug therapy , Neoplasms/pathology , Humans , Models, Biological , Neoplasms/etiology
9.
Oncotarget ; 2(1-2): 18-28, 2011.
Article in English | MEDLINE | ID: mdl-21378408

ABSTRACT

Our current understanding of eukaryotic transcription has greatly benefited from use of small molecule inhibitors that have delineated multiple regulatory steps in site-specific initiation and elongation of RNA synthesis by multiple forms of RNA polymerase (RNAP). This class of "transcription" drugs is also of therapeutic interest and under evaluation in clinical trials. However, to date very few small molecules that directly abolish transcription have been identified, particularly those that act at the level of RNAP II initiation. Using a biochemical assay that measures transcription from recombinant, natural p53-responsive promoters and an artificial "super" promoter, we have identified three distinct small molecules that inhibit mRNA synthesis in vitro. Unexpectedly, these are kinase inhibitors, Hypericin, Rottlerin, and SP600125, with known substrates, which we find also strongly impair transcriptional initiation (IC50s = µM range) by targeting specific components of the RNAP II pre-initiation complex. When measured before and during transcription in vitro, one common target of inhibition by all three compounds is modification of the TATA Binding Protein (TBP) within the RNAP II holocomplex as it converts to an active transcribing enzyme. On this basis, by blocking the critical step of TBP modification, transcriptional initiation is effectively abolished even on structurally distinct core promoters.


Subject(s)
Protein Kinase Inhibitors/pharmacology , RNA Polymerase II/antagonists & inhibitors , Drug Evaluation, Preclinical , HeLa Cells , Humans , RNA Polymerase II/genetics , RNA Polymerase II/metabolism , TATA-Box Binding Protein/antagonists & inhibitors , TATA-Box Binding Protein/genetics , TATA-Box Binding Protein/metabolism , Transcription, Genetic/drug effects
10.
Genes Dev ; 24(2): 135-47, 2010 Jan 15.
Article in English | MEDLINE | ID: mdl-20040571

ABSTRACT

p53 target promoters are structurally diverse and display pronounced differences in RNA polymerase II (RNAP II) occupancy even in unstressed cells, with higher levels observed on cell cycle arrest genes (p21) compared with apoptotic genes (Fas/APO1). This occupancy correlates well with their ability to undergo rapid or delayed stress induction. To understand the basis for such distinct temporal assembly of transcription complexes, we examined the role of core promoter structures in this process. We find that the p21 core promoter directs rapid, TATA box-dependent assembly of RNAP II preinitiation complexes (PICs), but permits few rounds of RNAP II reinitiation. In contrast, PIC formation at the Fas/APO1 core promoter is very inefficient but supports multiple rounds of transcription. We define a downstream element within the Fas/APO1 core promoter that is essential for its activation, and identify nuclear transcription factor Y (NF-Y) as its binding partner. NF-Y acts as a bifunctional transcription factor that regulates basal expression of Fas/APO1 in vivo. Thus, two critical parameters of the stress-induced p53 transcriptional response are the kinetics of gene induction and duration of expression through frequent reinitiation. These features are intrinsic, DNA-encoded properties of diverse core promoters that may be fundamental to anticipatory programming of p53 response genes upon stress.


Subject(s)
Gene Expression Regulation , Promoter Regions, Genetic/genetics , RNA Polymerase II/metabolism , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism , Amino Acid Sequence , CCAAT-Binding Factor/metabolism , HeLa Cells , Humans , Kinetics , Molecular Sequence Data , Protein Binding , Recombinant Fusion Proteins , Stress, Physiological/genetics , TATA Box/genetics , fas Receptor/genetics , fas Receptor/metabolism
11.
Mol Cell ; 34(3): 271-84, 2009 May 15.
Article in English | MEDLINE | ID: mdl-19450526

ABSTRACT

The p16(INK4a) tumor suppressor gene is a frequent target of epigenetic inactivation in human cancers, which is an early event in breast carcinogenesis. We describe the existence of a chromatin boundary upstream of the p16 gene that is lost when this gene is aberrantly silenced. We show that the multifunctional protein CTCF associates in the vicinity of this boundary and absence of binding strongly coincides with p16 silencing in multiple types of cancer cells. CTCF binding also correlates with RASSF1A and CDH1 gene activation, and CTCF interaction is absent when these genes are methylated and silenced. Interestingly, defective poly(ADP-ribosyl)ation of CTCF and dissociation from the molecular chaperone Nucleolin occur in p16-silenced cells, abrogating its proper function. Thus, destabilization of specific chromosomal boundaries through aberrant crosstalk between CTCF, poly(ADP-ribosyl)ation, and DNA methylation may be a general mechanism to inactivate tumor suppressor genes and initiate tumorigenesis in numerous forms of human cancers.


Subject(s)
Breast Neoplasms/metabolism , Chromatin/metabolism , Cyclin-Dependent Kinase Inhibitor p16/genetics , DNA-Binding Proteins/metabolism , Epigenesis, Genetic , Gene Silencing , Repressor Proteins/metabolism , Antigens, CD , Breast Neoplasms/genetics , CCCTC-Binding Factor , Cadherins/genetics , Cadherins/metabolism , Cell Line, Tumor , Cyclin-Dependent Kinase Inhibitor p16/metabolism , DNA-Binding Proteins/genetics , Female , Gene Expression Regulation , Humans , Models, Genetic , Promoter Regions, Genetic , Repressor Proteins/genetics , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/metabolism
12.
J Biol Chem ; 283(47): 32254-63, 2008 Nov 21.
Article in English | MEDLINE | ID: mdl-18809673

ABSTRACT

The composition of chromatin-remodeling complexes dictates how these enzymes control transcriptional programs and cellular identity. In the present study we investigated the composition of SWI/SNF complexes in embryonic stem cells (ESCs). In contrast to differentiated cells, ESCs have a biased incorporation of certain paralogous SWI/SNF subunits with low levels of BRM, BAF170, and ARID1B. Upon differentiation, the expression of these subunits increases, resulting in a higher diversity of compositionally distinct SWI/SNF enzymes. We also identified BRD7 as a novel component of the Polybromo-associated BRG1-associated factor (PBAF) complex in both ESCs and differentiated cells. Using short hairpin RNA-mediated depletion of BRG1, we showed that SWI/SNF can function as both a repressor and an activator in pluripotent cells, regulating expression of developmental modifiers and signaling components such as Nodal, ADAMTS1, BMI-1, CRABP1, and thyroid releasing hormone. Knockdown studies of PBAF-specific BRD7 and of a signature subunit within the BAF complex, ARID1A, showed that these two subcomplexes affect SWI/SNF target genes differentially, in some cases even antagonistically. This may be due to their different biochemical properties. Finally we examined the role of SWI/SNF in regulating its target genes during differentiation. We found that SWI/SNF affects recruitment of components of the preinitiation complex in a promoter-specific manner to modulate transcription positively or negatively. Taken together, our results provide insight into the function of compositionally diverse SWI/SNF enzymes that underlie their inherent gene-specific mode of action.


Subject(s)
Chromosomal Proteins, Non-Histone/metabolism , Chromosomal Proteins, Non-Histone/physiology , Embryonic Stem Cells/cytology , Gene Expression Regulation, Developmental , Transcription Factors/metabolism , Animals , Cell Cycle , Cell Differentiation , DNA Helicases/metabolism , Humans , Mice , Models, Biological , Multiprotein Complexes/chemistry , Nuclear Proteins/metabolism , Transcription, Genetic
13.
Curr Opin Genet Dev ; 16(5): 508-12, 2006 Oct.
Article in English | MEDLINE | ID: mdl-16905306

ABSTRACT

Chromatin-remodeling complexes are biochemically diverse, functionally selective machines that regulate crucial aspects of DNA metabolism, including transcription and chromatin assembly. These complexes modulate histone-DNA interactions to affect nucleosome repositioning and disassembly, and histone variant exchange, thereby generating compositionally specialized chromatin. Recent studies have revealed precise mechanisms by which specific remodelers control the transition from proliferating progenitors to committed cells through a highly synchronized switch in transcriptional programs. This involves temporal and, often, signal-dependent gene-targeted interactions between individual remodelers and tissue-specific master proteins that regulate myogenesis, neurogenesis and lymphogenesis. Distinct remodelers have also been shown to direct self-renewal of different types of stem cells in response to particular microenvironments.


Subject(s)
Cell Differentiation , Chromatin Assembly and Disassembly/genetics , Stem Cells/cytology , Stem Cells/metabolism , Animals , Muscle Development , Neurons/cytology , Neurons/metabolism , Signal Transduction
14.
Genes Dev ; 20(5): 601-12, 2006 Mar 01.
Article in English | MEDLINE | ID: mdl-16510875

ABSTRACT

Activation of the p53 pathway mediates cellular responses to diverse forms of stress. Here we report that the p53 target gene p21(CIP1) is regulated by stress at post-initiation steps through conversion of paused RNA polymerase II (RNAP II) into an elongating form. High-resolution chromatin immunoprecipitation assays (ChIP) demonstrate that p53-dependent activation of p21(CIP1) transcription after DNA damage occurs concomitantly with changes in RNAP II phosphorylation status and recruitment of the elongation factors DSIF (DRB Sensitivity-Inducing Factor), P-TEFb (Positive Transcription Elongation Factor b), TFIIH, TFIIF, and FACT (Facilitates Chromatin Transcription) to distinct regions of the p21(CIP1) locus. Paradoxically, pharmacological inhibition of P-TEFb leads to global inhibition of mRNA synthesis but activation of the p53 pathway through p53 accumulation, expression of specific p53 target genes, and p53-dependent apoptosis. ChIP analyses of p21(CIP1) activation in the absence of functional P-TEFb reveals the existence of two distinct kinases that phosphorylate Ser5 of the RNAP II C-terminal domain (CTD). Importantly, CTD phosphorylation at Ser2 is not required for p21(CIP1) transcription, mRNA cleavage, or polyadenylation. Furthermore, recruitment of FACT requires CTD kinases, yet FACT is dispensable for p21(CIP1) expression. Thus, select genes within the p53 pathway bypass the requirement for P-TEFb and RNAP II phosphorylation to trigger a cellular response to inhibition of global mRNA synthesis.


Subject(s)
Positive Transcriptional Elongation Factor B/metabolism , RNA Polymerase II/metabolism , Transcription, Genetic , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism , Apoptosis , Chromatin Immunoprecipitation , Cyclin-Dependent Kinase Inhibitor p21/chemistry , Cyclin-Dependent Kinase Inhibitor p21/genetics , Cyclin-Dependent Kinase Inhibitor p21/metabolism , DNA/analysis , DNA/metabolism , DNA-Binding Proteins/metabolism , Doxorubicin/pharmacology , Flow Cytometry , HCT116 Cells , High Mobility Group Proteins/metabolism , Humans , Kinetics , Nuclear Proteins/metabolism , Phosphorylation , Positive Transcriptional Elongation Factor B/antagonists & inhibitors , Positive Transcriptional Elongation Factor B/genetics , Protein Isoforms/metabolism , Protein Structure, Tertiary , RNA Polymerase II/chemistry , Reverse Transcriptase Polymerase Chain Reaction , Serine/chemistry , Transcription Factor TFIIH/metabolism , Transcription Factors/metabolism , Transcription Factors, TFII/metabolism , Transcriptional Elongation Factors/metabolism
15.
J Mol Biol ; 350(5): 883-96, 2005 Jul 29.
Article in English | MEDLINE | ID: mdl-15979088

ABSTRACT

The HS2 enhancer in the beta-globin locus control region (LCR) regulates transcription of the globin genes 10-50 kb away. Earlier studies show that a transcription mechanism initiated by the HS2 enhancer through the intervening DNA in the direction of the cis-linked promoter and gene mediates long-range enhancer function. Here, we further analyzed the enhancer-initiated RNAs and their mode of transcription from the HS2 enhancer in the endogenous genome of erythroid K562 cells, in plasmids integrated into K562 cells and in purified DNA used as template in in vitro transcription reactions. We found that the HS2 enhancer was able to initiate transcription autonomously in the absence of a cis-linked globin promoter. The enhancer-initiated, intergenic RNAs were different from the mRNA synthesized at the promoter in several aspects. The enhancer RNAs were synthesized not from a defined site but from multiple sites both within and as far as 1 kb downstream of the enhancer. The enhancer RNAs did not appear to contain a normal cap structure at the 5' ends. They were polyadenylated at multiple sites within 3 kb downstream of their initiation sites and were therefore shorter than 3 kb in lengths. The enhancer RNAs remained in discrete spots within the nucleus and were not processed into mRNA or translated into proteins. These particular features of enhancer-initiated transcription indicate that the transcriptional complex assembled by the enhancer was different from the basal transcription complex assembled at the promoter. The results suggest that in synthesizing non-coding, intergenic RNAs, the enhancer-assembled transcription complex could track through the intervening DNA to reach the basal promoter complex and activate efficient mRNA synthesis from the promoter.


Subject(s)
Enhancer Elements, Genetic/physiology , Globins/genetics , RNA, Messenger/biosynthesis , Transcription, Genetic/genetics , DNA, Intergenic , Gene Expression Regulation/genetics , Humans , Macromolecular Substances , Promoter Regions, Genetic/genetics
16.
Proc Natl Acad Sci U S A ; 102(12): 4459-64, 2005 Mar 22.
Article in English | MEDLINE | ID: mdl-15753290

ABSTRACT

Hormones and nutrients often induce genetic programs via signaling pathways that interface with gene-specific activators. Activation of the cAMP pathway, for example, stimulates cellular gene expression by means of the PKA-mediated phosphorylation of cAMP-response element binding protein (CREB) at Ser-133. Here, we use genome-wide approaches to characterize target genes that are regulated by CREB in different cellular contexts. CREB was found to occupy approximately 4,000 promoter sites in vivo, depending on the presence and methylation state of consensus cAMP response elements near the promoter. The profiles for CREB occupancy were very similar in different human tissues, and exposure to a cAMP agonist stimulated CREB phosphorylation over a majority of these sites. Only a small proportion of CREB target genes was induced by cAMP in any cell type, however, due in part to the preferential recruitment of the coactivator CREB-binding protein to those promoters. These results indicate that CREB phosphorylation alone is not a reliable predictor of target gene activation and that additional CREB regulatory partners are required for recruitment of the transcriptional apparatus to the promoter.


Subject(s)
Transcription Factors/genetics , Transcription Factors/metabolism , Base Sequence , Binding Sites/genetics , Cell Line , Cyclic AMP/metabolism , Cyclic AMP Response Element-Binding Protein , DNA/genetics , DNA/metabolism , DNA Methylation , Gene Expression Regulation , Genome, Human , Hepatocytes/metabolism , Humans , Islets of Langerhans/metabolism , Phosphorylation , Promoter Regions, Genetic , Tissue Distribution , Transcription Factors/chemistry , Transcriptional Activation
17.
Proc Natl Acad Sci U S A ; 101(51): 17622-7, 2004 Dec 21.
Article in English | MEDLINE | ID: mdl-15585582

ABSTRACT

Phosphorylation of the cAMP response element binding protein (CREB) at Ser-133 in response to hormonal stimuli triggers cellular gene expression via the recruitment of the histone acetylase coactivator paralogs CREB binding protein (CBP) and p300 to the promoter. The NMR structure of the CREB:CBP complex, using relevant interaction domains called KID and KIX, respectively, reveals a shallow hydrophobic groove on the surface of KIX that accommodates an amphipathic helix in phospho (Ser-133) KID. Using an NMR-based screening approach on a preselected small-molecule library, we identified several compounds that bind to different surfaces on KIX. One of these, KG-501 (2-naphthol-AS-E-phosphate), targeted a surface distal to the CREB binding groove that includes Arg-600, a residue that is required for the CREB:CBP interaction. When added to live cells, KG-501 disrupted the CREB: CBP complex and attenuated target gene induction in response to cAMP agonist. These results demonstrate the ability of small molecules to interfere with second-messenger signaling cascades by inhibiting specific protein-protein interactions in the nucleus.


Subject(s)
Cyclic AMP Response Element-Binding Protein/antagonists & inhibitors , Cyclic AMP Response Element-Binding Protein/metabolism , Naphthols/pharmacology , Nuclear Proteins/antagonists & inhibitors , Nuclear Proteins/metabolism , Organophosphates/pharmacology , Trans-Activators/antagonists & inhibitors , Trans-Activators/metabolism , CREB-Binding Protein , Cell Line , Cyclic AMP Response Element-Binding Protein/chemistry , Humans , Models, Molecular , Naphthols/chemistry , Nuclear Magnetic Resonance, Biomolecular , Nuclear Proteins/chemistry , Organophosphates/chemistry , Phosphoserine/metabolism , Protein Binding/drug effects , Protein Conformation , Signal Transduction/drug effects , Trans-Activators/chemistry
18.
Genes Dev ; 18(2): 144-56, 2004 Jan 15.
Article in English | MEDLINE | ID: mdl-14729568

ABSTRACT

The recruitment of coactivators by nuclear hormone receptors (NRs) promotes transcription by subverting chromatin-mediated repression. Although the histone methylation enzyme CARM1 and an ATP-remodeling complex have been individually implicated in nuclear receptor-dependent transcription, neither a functional nor mechanistic linkage between these systems has been identified. In the process of purifying endogenous CARM1-interacting proteins, we identified an associated complex, nucleosomal methylation activator complex (NUMAC), which includes at least eight components of SWI/SNF, including the ATPase BRG1. In the NUMAC complex, the methylase, CARM1, acquires the ability to covalently modify nucleosomal histones, and the directed nucleosome versus free core histone methylation-specificity change is increased dramatically. Reciprocally, CARM1 stimulates the ATPase activity of BRG1, a key component in nucleosome remodeling. In vivo, CARM1 and BRG1 coassemble on an estrogen receptor (ER)-target gene to cooperatively activate ER-dependent transcription. This association of ATP-remodeling factors with HMT CARM1 defines a new component of regulation in the nuclear hormone-signaling pathway.


Subject(s)
Chromosomal Proteins, Non-Histone/metabolism , Protein-Arginine N-Methyltransferases/metabolism , Receptors, Cytoplasmic and Nuclear/metabolism , Signal Transduction/physiology , Transcription Factors/metabolism , Adenosine Triphosphate/metabolism , Animals , Humans , Macromolecular Substances , Methylation , Methyltransferases/metabolism , Protein-Arginine N-Methyltransferases/isolation & purification , Substrate Specificity
19.
Mol Cell ; 12(4): 1015-27, 2003 Oct.
Article in English | MEDLINE | ID: mdl-14580351

ABSTRACT

The tumor suppressor protein p53 regulates transcriptional programs that control the response to cellular stress. We show that distinct mechanisms exist to activate p53 target genes as revealed by marked differences in affinities and damage-specific recruitment of transcription initiation components. p53 functions in a temporal manner to regulate promoter activity both before and after stress. Before DNA damage, basal levels of p53 are required to assemble a poised RNA polymerase II initiation complex on the p21 promoter. RNA pol II is converted into an elongating form shortly after stress but before p53 stabilization. Proapoptotic promoters, such as Fas/APO1, have low levels of bound RNA pol II but undergo damage-induced activation through efficient reinitiation. Surprisingly, in a p53-dependent process key basal factors TAFII250 and TFIIB assemble into the transcription machinery in a stress- and promoter-specific manner, behaving as differential cofactors for p53 action after distinct types of DNA damage.


Subject(s)
DNA Damage/genetics , Promoter Regions, Genetic/genetics , Stress, Physiological/genetics , Transcriptional Activation/genetics , Tumor Suppressor Protein p53/genetics , Apoptosis/genetics , Cell Line, Tumor , Cyclin-Dependent Kinase Inhibitor p21 , Cyclins/genetics , Histone Acetyltransferases , Humans , RNA Polymerase II/genetics , TATA-Binding Protein Associated Factors/metabolism , Transcription Factor TFIIB/metabolism , Transcription Factor TFIID/metabolism , Transcription Initiation Site/physiology , fas Receptor/metabolism
20.
Mol Cell ; 11(2): 377-89, 2003 Feb.
Article in English | MEDLINE | ID: mdl-12620226

ABSTRACT

Mammalian SWI/SNF chromatin remodeling complexes are involved in critical aspects of cellular growth and genomic stability. Each complex contains one of two highly homologous ATPases, BRG1 and BRM, yet little is known about their specialized functions. We show that BRG1and BRM associate with different promoters during cellular proliferation and differentiation, and in response to specific signaling pathways by preferential interaction with certain classes of transcription factors. BRG1 binds to zinc finger proteins through a unique N-terminal domain that is not present in BRM. BRM interacts with two ankyrin repeat proteins that are critical components of Notch signal transduction. Thus, BRG1 and BRM complexes may direct distinct cellular processes by recruitment to specific promoters through protein-protein interactions that are unique to each ATPase.


Subject(s)
Chromosomal Proteins, Non-Histone/metabolism , Nuclear Proteins/metabolism , Transcription Factors/metabolism , Adenosine Triphosphatases/metabolism , Animals , Cell Differentiation , Cell Division , Cell Line , Chromatin/metabolism , Chromosomal Proteins, Non-Histone/genetics , DNA/genetics , DNA Helicases , Humans , In Vitro Techniques , Mice , Nuclear Proteins/genetics , Promoter Regions, Genetic , Protein Binding , Protein Structure, Tertiary , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Signal Transduction , Transcription Factors/genetics , Transcription, Genetic , Zinc Fingers
SELECTION OF CITATIONS
SEARCH DETAIL
...