Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 49
Filter
1.
Mol Cell ; 84(2): 202-220.e15, 2024 Jan 18.
Article in English | MEDLINE | ID: mdl-38103559

ABSTRACT

Compounds binding to the bromodomains of bromodomain and extra-terminal (BET) family proteins, particularly BRD4, are promising anticancer agents. Nevertheless, side effects and drug resistance pose significant obstacles in BET-based therapeutics development. Using high-throughput screening of a 200,000-compound library, we identified small molecules targeting a phosphorylated intrinsically disordered region (IDR) of BRD4 that inhibit phospho-BRD4 (pBRD4)-dependent human papillomavirus (HPV) genome replication in HPV-containing keratinocytes. Proteomic profiling identified two DNA damage response factors-53BP1 and BARD1-crucial for differentiation-associated HPV genome amplification. pBRD4-mediated recruitment of 53BP1 and BARD1 to the HPV origin of replication occurs in a spatiotemporal and BRD4 long (BRD4-L) and short (BRD4-S) isoform-specific manner. This recruitment is disrupted by phospho-IDR-targeting compounds with little perturbation of the global transcriptome and BRD4 chromatin landscape. The discovery of these protein-protein interaction inhibitors (PPIi) not only demonstrates the feasibility of developing PPIi against phospho-IDRs but also uncovers antiviral agents targeting an epigenetic regulator essential for virus-host interaction and cancer development.


Subject(s)
Papillomavirus Infections , Transcription Factors , Humans , Transcription Factors/genetics , Transcription Factors/metabolism , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Human Papillomavirus Viruses , Papillomavirus Infections/drug therapy , Papillomavirus Infections/genetics , Proteomics , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Papillomaviridae/genetics , Papillomaviridae/metabolism , Viral Proteins/genetics , Virus Replication/physiology , DNA Repair , Bromodomain Containing Proteins
2.
Mol Cell ; 78(6): 1133-1151.e14, 2020 06 18.
Article in English | MEDLINE | ID: mdl-32402252

ABSTRACT

Precise control of the RNA polymerase II (RNA Pol II) cycle, including pausing and pause release, maintains transcriptional homeostasis and organismal functions. Despite previous work to understand individual transcription steps, we reveal a mechanism that integrates RNA Pol II cycle transitions. Surprisingly, KAP1/TRIM28 uses a previously uncharacterized chromatin reader cassette to bind hypo-acetylated histone 4 tails at promoters, guaranteeing continuous progression of RNA Pol II entry to and exit from the pause state. Upon chromatin docking, KAP1 first associates with RNA Pol II and then recruits a pathway-specific transcription factor (SMAD2) in response to cognate ligands, enabling gene-selective CDK9-dependent pause release. This coupling mechanism is exploited by tumor cells to aberrantly sustain transcriptional programs commonly dysregulated in cancer patients. The discovery of a factor integrating transcription steps expands the functional repertoire by which chromatin readers operate and provides mechanistic understanding of transcription regulation, offering alternative therapeutic opportunities to target transcriptional dysregulation.


Subject(s)
RNA Polymerase II/metabolism , Tripartite Motif-Containing Protein 28/metabolism , Acetylation , Cell Line, Tumor , Chromatin/genetics , Chromatin/metabolism , Cyclin-Dependent Kinase 9/metabolism , Gene Expression Regulation/genetics , Histones/metabolism , Humans , Oncogenes/genetics , Promoter Regions, Genetic/genetics , Protein Processing, Post-Translational/genetics , RNA Polymerase II/genetics , Smad2 Protein/metabolism , Transcription Factors/metabolism , Transcription, Genetic , Tripartite Motif-Containing Protein 28/genetics
3.
Mol Cell ; 78(6): 1114-1132.e10, 2020 06 18.
Article in English | MEDLINE | ID: mdl-32446320

ABSTRACT

Bromodomain-containing protein 4 (BRD4) is a cancer therapeutic target in ongoing clinical trials disrupting primarily BRD4-regulated transcription programs. The role of BRD4 in cancer has been attributed mainly to the abundant long isoform (BRD4-L). Here we show, by isoform-specific knockdown and endogenous protein detection, along with transgene expression, the less abundant BRD4 short isoform (BRD4-S) is oncogenic while BRD4-L is tumor-suppressive in breast cancer cell proliferation and migration, as well as mammary tumor formation and metastasis. Through integrated RNA-seq, genome-wide ChIP-seq, and CUT&RUN association profiling, we identify the Engrailed-1 (EN1) homeobox transcription factor as a key BRD4-S coregulator, particularly in triple-negative breast cancer. BRD4-S and EN1 comodulate the extracellular matrix (ECM)-associated matrisome network, including type II cystatin gene cluster, mucin 5, and cathepsin loci, via enhancer regulation of cancer-associated genes and pathways. Our work highlights the importance of targeted therapies for the oncogenic, but not tumor-suppressive, activity of BRD4.


Subject(s)
Breast Neoplasms/metabolism , Cell Cycle Proteins/metabolism , Cell Cycle Proteins/physiology , Transcription Factors/metabolism , Transcription Factors/physiology , Animals , Breast Neoplasms/genetics , Cell Line, Tumor , Cell Movement , Cell Proliferation , Female , Gene Expression Regulation, Neoplastic/genetics , Genes, Homeobox , Homeodomain Proteins/metabolism , Humans , Mice , Neoplasm Invasiveness , Nuclear Proteins/metabolism , Protein Isoforms/metabolism , Proteins/antagonists & inhibitors , Proteins/metabolism , Transcription, Genetic/genetics , Triple Negative Breast Neoplasms/genetics
4.
J Virol ; 97(10): e0074723, 2023 10 31.
Article in English | MEDLINE | ID: mdl-37712706

ABSTRACT

IMPORTANCE: Respiratory syncytial virus (RSV) matrix (M) protein is indispensable for virion assembly and release. It is localized to the nucleus during early infection to perturb host transcription. However, the function of RSV M protein in other cellular activities remains poorly understood. In this study, several interferon response-associated host factors, including RACK1, were identified by proteomic analysis as RSV M interactors. Knockdown of RACK1 attenuates RSV-restricted IFN signaling leading to enhanced host defense against RSV infection, unraveling a role of M protein in antagonizing IFN response via association with RACK1. Our study uncovers a previously unrecognized mechanism of immune evasion by RSV M protein and identifies RACK1 as a novel host factor recruited by RSV, highlighting RACK1 as a potential new target for RSV therapeutics development.


Subject(s)
Receptors for Activated C Kinase , Respiratory Syncytial Virus Infections , Respiratory Syncytial Virus, Human , Viral Matrix Proteins , Humans , Interferons , Neoplasm Proteins/genetics , Proteins , Proteomics , Receptors for Activated C Kinase/metabolism , Respiratory Syncytial Virus Infections/metabolism , Respiratory Syncytial Virus Infections/virology , Respiratory Syncytial Virus, Human/genetics , Viral Matrix Proteins/metabolism
5.
J Virol ; 97(10): e0078223, 2023 10 31.
Article in English | MEDLINE | ID: mdl-37712702

ABSTRACT

IMPORTANCE: Human papillomavirus 16 (HPV16) is a causative agent in around 3%-4% of all human cancers, and currently, there are no anti-viral therapeutics available for combating this disease burden. In order to identify new therapeutic targets, we must increase our understanding of the HPV16 life cycle. Previously, we demonstrated that an interaction between E2 and the cellular protein TopBP1 mediates the plasmid segregation function of E2, allowing distribution of viral genomes into daughter nuclei following cell division. Here, we demonstrate that E2 interaction with an additional host protein, BRD4, is also essential for E2 segregation function, and that BRD4 exists in a complex with TopBP1. Overall, these results enhance our understanding of a critical part of the HPV16 life cycle and presents several therapeutic targets for disruption of the viral life cycle.


Subject(s)
Chromatin , Oncogene Proteins, Viral , Humans , Bromodomain Containing Proteins , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Chromatin/metabolism , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Human papillomavirus 16/genetics , Human papillomavirus 16/metabolism , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Oncogene Proteins, Viral/genetics , Oncogene Proteins, Viral/metabolism , Plasmids/genetics , Transcription Factors/genetics , Transcription Factors/metabolism
6.
Nature ; 529(7586): 413-417, 2016 Jan 21.
Article in English | MEDLINE | ID: mdl-26735014

ABSTRACT

Triple-negative breast cancer (TNBC) is a heterogeneous and clinically aggressive disease for which there is no targeted therapy. BET bromodomain inhibitors, which have shown efficacy in several models of cancer, have not been evaluated in TNBC. These inhibitors displace BET bromodomain proteins such as BRD4 from chromatin by competing with their acetyl-lysine recognition modules, leading to inhibition of oncogenic transcriptional programs. Here we report the preferential sensitivity of TNBCs to BET bromodomain inhibition in vitro and in vivo, establishing a rationale for clinical investigation and further motivation to understand mechanisms of resistance. In paired cell lines selected for acquired resistance to BET inhibition from previously sensitive TNBCs, we failed to identify gatekeeper mutations, new driver events or drug pump activation. BET-resistant TNBC cells remain dependent on wild-type BRD4, which supports transcription and cell proliferation in a bromodomain-independent manner. Proteomic studies of resistant TNBC identify strong association with MED1 and hyper-phosphorylation of BRD4 attributable to decreased activity of PP2A, identified here as a principal BRD4 serine phosphatase. Together, these studies provide a rationale for BET inhibition in TNBC and present mechanism-based combination strategies to anticipate clinical drug resistance.


Subject(s)
Azepines/pharmacology , Azepines/therapeutic use , Drug Resistance, Neoplasm/drug effects , Nuclear Proteins/antagonists & inhibitors , Protein Structure, Tertiary/drug effects , Transcription Factors/antagonists & inhibitors , Triazoles/pharmacology , Triazoles/therapeutic use , Triple Negative Breast Neoplasms/drug therapy , Animals , Binding, Competitive/drug effects , Casein Kinase II/metabolism , Cell Cycle Proteins , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Proliferation/genetics , Chromatin/genetics , Chromatin/metabolism , Drug Resistance, Neoplasm/genetics , Epigenesis, Genetic/drug effects , Epigenesis, Genetic/genetics , Female , Gene Expression Regulation, Neoplastic/drug effects , Genome, Human/drug effects , Genome, Human/genetics , Humans , Mediator Complex Subunit 1/metabolism , Mice , Nuclear Proteins/deficiency , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Phosphorylation/drug effects , Phosphoserine/metabolism , Protein Binding/drug effects , Protein Phosphatase 2/metabolism , Proteomics , Transcription Factors/deficiency , Transcription Factors/genetics , Transcription Factors/metabolism , Transcription, Genetic/drug effects , Triple Negative Breast Neoplasms/genetics , Triple Negative Breast Neoplasms/metabolism , Triple Negative Breast Neoplasms/pathology , Xenograft Model Antitumor Assays
7.
Cancer ; 127(6): 850-864, 2021 03 15.
Article in English | MEDLINE | ID: mdl-33270909

ABSTRACT

BACKGROUND: Despite the significant societal burden of human papillomavirus (HPV)-associated cancers, clinical screening interventions for HPV-associated noncervical cancers are not available. Blood-based biomarkers may help close this gap in care. METHODS: Five databases were searched, 5687 articles were identified, and 3631 unique candidate titles and abstracts were independently reviewed by 2 authors; 702 articles underwent a full-text review. Eligibility criteria included the assessment of a blood-based biomarker within a cohort or case-control study. RESULTS: One hundred thirty-seven studies were included. Among all biomarkers assessed, HPV-16 E seropositivity and circulating HPV DNA were most significantly correlated with HPV-associated cancers in comparison with cancer-free controls. In most scenarios, HPV-16 E6 seropositivity varied nonsignificantly according to tumor type, specimen collection timing, and anatomic site (crude odds ratio [cOR] for p16+ or HPV+ oropharyngeal cancer [OPC], 133.10; 95% confidence interval [CI], 59.40-298.21; cOR for HPV-unspecified OPC, 25.41; 95% CI, 8.71-74.06; cOR for prediagnostic HPV-unspecified OPC, 59.00; 95% CI, 15.39-226.25; cOR for HPV-unspecified cervical cancer, 12.05; 95% CI, 3.23-44.97; cOR for HPV-unspecified anal cancer, 73.60; 95% CI, 19.68-275.33; cOR for HPV-unspecified penile cancer, 16.25; 95% CI, 2.83-93.48). Circulating HPV-16 DNA was a valid biomarker for cervical cancer (cOR, 15.72; 95% CI, 3.41-72.57). In 3 cervical cancer case-control studies, cases exhibited unique microRNA expression profiles in comparison with controls. Other assessed biomarker candidates were not valid. CONCLUSIONS: HPV-16 E6 antibodies and circulating HPV-16 DNA are the most robustly analyzed and most promising blood-based biomarkers for HPV-associated cancers to date. Comparative validity analyses are warranted. Variations in tumor type-specific, high-risk HPV DNA prevalence according to anatomic site and world region highlight the need for biomarkers targeting more high-risk HPV types. Further investigation of blood-based microRNA expression profiling appears indicated.


Subject(s)
Antibodies, Viral/blood , Anus Neoplasms/virology , Biomarkers/blood , DNA, Viral/blood , Oropharyngeal Neoplasms/virology , Papillomavirus Infections/complications , Female , Human papillomavirus 16/isolation & purification , Humans , Uterine Cervical Neoplasms/virology
8.
Mol Cell ; 49(5): 843-57, 2013 Mar 07.
Article in English | MEDLINE | ID: mdl-23317504

ABSTRACT

Bromodomain-containing protein 4 (Brd4) is an epigenetic reader and transcriptional regulator recently identified as a cancer therapeutic target for acute myeloid leukemia, multiple myeloma, and Burkitt's lymphoma. Although chromatin targeting is a crucial function of Brd4, there is little understanding of how bromodomains that bind acetylated histones are regulated, nor how the gene-specific activity of Brd4 is determined. Via interaction screen and domain mapping, we identified p53 as a functional partner of Brd4. Interestingly, Brd4 association with p53 is modulated by casein kinase II (CK2)-mediated phosphorylation of a conserved acidic region in Brd4 that selectively contacts either a juxtaposed bromodomain or an adjacent basic region to dictate the ability of Brd4 binding to chromatin and also the recruitment of p53 to regulated promoters. The unmasking of bromodomains and activator recruitment, concurrently triggered by the CK2 phospho switch, provide an intriguing mechanism for gene-specific targeting by a universal epigenetic reader.


Subject(s)
Casein Kinase II/metabolism , Chromatin/metabolism , Gene Targeting , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism , Casein Kinase II/genetics , Cell Cycle Proteins , Chromatin/genetics , HCT116 Cells , HEK293 Cells , Histones/chemistry , Histones/metabolism , Humans , Leukemia, Myeloid, Acute/genetics , Transcription, Genetic , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism
9.
Nucleic Acids Res ; 46(13): 6576-6591, 2018 07 27.
Article in English | MEDLINE | ID: mdl-29860315

ABSTRACT

Hypoxia-inducible factor 1 (HIF-1) is a master transcriptional regulator in response to hypoxia and its transcriptional activity is crucial for cancer cell mobility. Here we present evidence for a novel epigenetic mechanism that regulates HIF-1 transcriptional activity and HIF-1-dependent migration of glioblastoma cells. The lysine methyltransferases G9a and GLP directly bound to the α subunit of HIF-1 (HIF-1α) and catalyzed mono- and di-methylation of HIF-1α at lysine (K) 674 in vitro and in vivo. K674 methylation suppressed HIF-1 transcriptional activity and expression of its downstream target genes PTGS1, NDNF, SLC6A3, and Linc01132 in human glioblastoma U251MG cells. Inhibition of HIF-1 by K674 methylation is due to reduced HIF-1α transactivation domain function but not increased HIF-1α protein degradation or impaired binding of HIF-1 to hypoxia response elements. K674 methylation significantly decreased HIF-1-dependent migration of U251MG cells under hypoxia. Importantly, we found that G9a was downregulated by hypoxia in glioblastoma, which was inversely correlated with PTGS1 expression and survival of patients with glioblastoma. Therefore, our findings uncover a hypoxia-induced negative feedback mechanism that maintains high activity of HIF-1 and cell mobility in human glioblastoma.


Subject(s)
Autoantigens/metabolism , Epigenesis, Genetic , Gene Expression Regulation, Neoplastic , Glioblastoma/genetics , Golgi Matrix Proteins/metabolism , Histocompatibility Antigens/metabolism , Histone-Lysine N-Methyltransferase/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Transcription, Genetic , Cell Hypoxia , Cell Line , Cell Movement , Glioblastoma/metabolism , Glioblastoma/physiopathology , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/antagonists & inhibitors , Hypoxia-Inducible Factor 1, alpha Subunit/chemistry , Lysine/metabolism , Methylation , Response Elements
10.
EMBO J ; 34(2): 184-99, 2015 Jan 13.
Article in English | MEDLINE | ID: mdl-25425577

ABSTRACT

Acetylation of transcriptional regulators is normally dynamically regulated by nutrient status but is often persistently elevated in nutrient-excessive obesity conditions. We investigated the functional consequences of such aberrantly elevated acetylation of the nuclear receptor FXR as a model. Proteomic studies identified K217 as the FXR acetylation site in diet-induced obese mice. In vivo studies utilizing acetylation-mimic and acetylation-defective K217 mutants and gene expression profiling revealed that FXR acetylation increased proinflammatory gene expression, macrophage infiltration, and liver cytokine and triglyceride levels, impaired insulin signaling, and increased glucose intolerance. Mechanistically, acetylation of FXR blocked its interaction with the SUMO ligase PIASy and inhibited SUMO2 modification at K277, resulting in activation of inflammatory genes. SUMOylation of agonist-activated FXR increased its interaction with NF-κB but blocked that with RXRα, so that SUMO2-modified FXR was selectively recruited to and trans-repressed inflammatory genes without affecting FXR/RXRα target genes. A dysregulated acetyl/SUMO switch of FXR in obesity may serve as a general mechanism for diminished anti-inflammatory response of other transcriptional regulators and provide potential therapeutic and diagnostic targets for obesity-related metabolic disorders.


Subject(s)
Gene Expression Regulation , Inflammation/pathology , Liver Diseases/pathology , Obesity/complications , Receptors, Cytoplasmic and Nuclear/metabolism , Small Ubiquitin-Related Modifier Proteins/metabolism , Acetylation , Amino Acid Sequence , Animals , Blotting, Western , Cytokines/genetics , Cytokines/metabolism , Electrophoretic Mobility Shift Assay , Gene Expression Profiling , Immunoenzyme Techniques , Immunoprecipitation , Inflammation/etiology , Inflammation/metabolism , Liver Diseases/etiology , Liver Diseases/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Obese , Molecular Sequence Data , NF-kappa B/genetics , NF-kappa B/metabolism , Obesity/physiopathology , Protein Conformation , Protein Processing, Post-Translational , Proteomics , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Receptors, Cytoplasmic and Nuclear/chemistry , Receptors, Cytoplasmic and Nuclear/genetics , Reverse Transcriptase Polymerase Chain Reaction , Sequence Homology, Amino Acid , Small Ubiquitin-Related Modifier Proteins/chemistry , Small Ubiquitin-Related Modifier Proteins/genetics , Sumoylation , Tandem Mass Spectrometry
11.
J Virol ; 91(2)2017 Jan 15.
Article in English | MEDLINE | ID: mdl-27807239

ABSTRACT

Papillomaviruses are small, double-stranded DNA viruses that encode the E2 protein, which controls transcription, replication, and genome maintenance in infected cells. Posttranslational modifications (PTMs) affecting E2 function and stability have been demonstrated for multiple types of papillomaviruses. Here we describe the first phosphorylation event involving a conserved tyrosine (Y) in the bovine papillomavirus 1 (BPV-1) E2 protein at amino acid 102. While its phosphodeficient phenylalanine (F) mutant activated both transcription and replication in luciferase reporter assays, a mutant that may act as a phosphomimetic, with a Y102-to-glutamate (E) mutation, lost both activities. The E2 Y102F protein interacted with cellular E2-binding factors and the viral helicase E1; however, in contrast, the Y102E mutant associated with only a subset and was unable to bind to E1. While the Y102F mutant fully supported transient viral DNA replication, BPV genomes encoding this mutation as well as Y102E were not maintained as stable episomes in murine C127 cells. These data imply that phosphorylation at Y102 disrupts the helical fold of the N-terminal region of E2 and its interaction with key cellular and viral proteins. We hypothesize that the resulting inhibition of viral transcription and replication in basal epithelial cells prevents the development of a lytic infection. IMPORTANCE: Papillomaviruses (PVs) are small, double-stranded DNA viruses that are responsible for cervical, oropharyngeal, and various genitourinary cancers. Although vaccines against the major oncogenic human PVs are available, there is no effective treatment for existing infections. One approach to better understand the viral replicative cycle, and potential therapies to target it, is to examine the posttranslational modification of viral proteins and its effect on function. Here we have discovered that the bovine papillomavirus 1 (BPV-1) transcription and replication regulator E2 is phosphorylated at residue Y102. While a phosphodeficient mutant at this site was fully functional, a phosphomimetic mutant displayed impaired transcription and replication activity as well as a lack of an association with certain E2-binding proteins. This study highlights the influence of posttranslational modifications on viral protein function and provides additional insight into the complex interplay between papillomaviruses and their hosts.


Subject(s)
Bovine papillomavirus 1/physiology , DNA-Binding Proteins/metabolism , Gene Expression Regulation, Viral , Transcription, Genetic , Tyrosine/metabolism , Viral Proteins/metabolism , Virus Replication , Alphapapillomavirus/physiology , Animals , Cattle , DNA-Binding Proteins/chemistry , DNA-Binding Proteins/genetics , Genome, Viral , Humans , Models, Molecular , Mutation , Phosphorylation , Plasmids/genetics , Protein Conformation , Protein Transport , Viral Proteins/chemistry , Viral Proteins/genetics
12.
PLoS Pathog ; 12(1): e1005366, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26727473

ABSTRACT

We investigated the mechanism of how the papillomavirus E2 transcription factor can activate promoters through activator protein (AP)1 binding sites. Using an unbiased approach with an inducible cell line expressing the viral transcription factor E2 and transcriptome analysis, we found that E2 induces the expression of the two AP1 components c-Fos and FosB in a Brd4-dependent manner. In vitro RNA interference confirmed that c-Fos is one of the AP1 members driving the expression of viral oncogenes E6/E7. Mutation analysis and in vivo RNA interference identified an essential role for c-Fos/AP1 and also for the bromodomain protein Brd4 for papillomavirus-induced tumorigenesis. Lastly, chromatin immunoprecipitation analysis demonstrated that E2 binds together with Brd4 to a canonical E2 binding site (E2BS) in the promoter of c-Fos, thus activating c-Fos expression. Thus, we identified a novel way how E2 activates the viral oncogene promoter and show that E2 may act as a viral oncogene by direct activation of c-Fos involved in skin tumorigenesis.


Subject(s)
Cell Transformation, Viral/physiology , DNA-Binding Proteins/metabolism , Gene Expression Regulation, Viral/physiology , Oncogene Proteins, Viral/metabolism , Papillomavirus Infections/metabolism , Proto-Oncogene Proteins c-fos/biosynthesis , Animals , Cell Line , Chromatin Immunoprecipitation , DNA Mutational Analysis , DNA-Binding Proteins/genetics , Disease Models, Animal , Electrophoretic Mobility Shift Assay , Genes, Viral , Immunoprecipitation , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Oncogene Proteins, Viral/genetics , Oncogenes , Papillomaviridae , Papillomavirus Infections/complications , Papillomavirus Infections/genetics , Proto-Oncogene Proteins c-fos/genetics , Rabbits , Transcription Factors/genetics , Transcription Factors/metabolism
13.
Nucleic Acids Res ; 44(3): 1080-94, 2016 Feb 18.
Article in English | MEDLINE | ID: mdl-26464433

ABSTRACT

The transcription start site (TSS) determines the length and composition of the 5' UTR and therefore can have a profound effect on translation. Yet, little is known about the mechanism underlying start site selection, particularly from promoters lacking conventional core elements such as TATA-box and Initiator. Here we report a novel mechanism of start site selection in the TATA- and Initiator-less promoter of miR-22, through a strictly localized downstream element termed DTIE and an upstream distal element. Changing the distance between them reduced promoter strength, altered TSS selection and diminished Pol II recruitment. Biochemical assays suggest that DTIE does not serve as a docking site for TFIID, the major core promoter-binding factor. TFIID is recruited to the promoter through DTIE but is dispensable for TSS selection. We determined DTIE consensus and found it to be remarkably prevalent, present at the same TSS downstream location in ≈20.8% of human promoters, the vast majority of which are TATA-less. Analysis of DTIE in the tumor suppressor p53 confirmed a similar function. Our findings reveal a novel mechanism of transcription initiation from TATA-less promoters.


Subject(s)
Promoter Regions, Genetic , TATA Box/genetics , Animals , Base Sequence , HEK293 Cells , Humans , MicroRNAs/genetics , Sequence Homology, Nucleic Acid , Transcription, Genetic
14.
Nucleic Acids Res ; 42(8): 4996-5006, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24589584

ABSTRACT

Functions of Kub5-Hera (In Greek Mythology Hera controlled Artemis) (K-H), the human homolog of the yeast transcription termination factor Rtt103, remain undefined. Here, we show that K-H has functions in both transcription termination and DNA double-strand break (DSB) repair. K-H forms distinct protein complexes with factors that repair DSBs (e.g. Ku70, Ku86, Artemis) and terminate transcription (e.g. RNA polymerase II). K-H loss resulted in increased basal R-loop levels, DSBs, activated DNA-damage responses and enhanced genomic instability. Significantly lowered Artemis protein levels were detected in K-H knockdown cells, which were restored with specific K-H cDNA re-expression. K-H deficient cells were hypersensitive to cytotoxic agents that induce DSBs, unable to reseal complex DSB ends, and showed significantly delayed γ-H2AX and 53BP1 repair-related foci regression. Artemis re-expression in K-H-deficient cells restored DNA-repair function and resistance to DSB-inducing agents. However, R loops persisted consistent with dual roles of K-H in transcription termination and DSB repair.


Subject(s)
Cell Cycle Proteins/metabolism , DNA Repair , Neoplasm Proteins/metabolism , Transcription Termination, Genetic , Animals , Antineoplastic Agents/toxicity , Cell Cycle Proteins/chemistry , Cell Cycle Proteins/genetics , Cell Cycle Proteins/physiology , Cells, Cultured , DNA Breaks, Double-Stranded , DNA End-Joining Repair , DNA-Binding Proteins , Endonucleases , Genomic Instability , Humans , Mice , Neoplasm Proteins/chemistry , Neoplasm Proteins/genetics , Neoplasm Proteins/physiology , Nuclear Proteins/metabolism , Protein Structure, Tertiary , Saccharomyces cerevisiae Proteins/genetics , Transcription Factors/genetics
15.
PLoS Pathog ; 9(11): e1003777, 2013.
Article in English | MEDLINE | ID: mdl-24278023

ABSTRACT

Replication foci are generated by many viruses to concentrate and localize viral DNA synthesis to specific regions of the cell. Expression of the HPV16 E1 and E2 replication proteins in keratinocytes results in nuclear foci that recruit proteins associated with the host DNA damage response. We show that the Brd4 protein localizes to these foci and is essential for their formation. However, when E1 and E2 begin amplifying viral DNA, Brd4 is displaced from the foci and cellular factors associated with DNA synthesis and homologous recombination are recruited. Differentiated HPV-infected keratinocytes form similar nuclear foci that contain amplifying viral DNA. We compare the different foci and show that, while they have many characteristics in common, there is a switch between early Brd4-dependent foci and mature Brd4-independent replication foci. However, HPV genomes encoding mutated E2 proteins that are unable to bind Brd4 can replicate and amplify the viral genome. We propose that, while E1, E2 and Brd4 might bind host chromatin at early stages of infection, there is a temporal and functional switch at later stages and increased E1 and E2 levels promote viral DNA amplification, displacement of Brd4 and growth of a replication factory. The concomitant DNA damage response recruits proteins required for DNA synthesis and repair, which could then be utilized for viral DNA replication. Hence, while Brd4 can enhance replication by concentrating viral processes in specific regions of the host nucleus, this interaction is not absolutely essential for HPV replication.


Subject(s)
DNA Replication/physiology , DNA, Viral/biosynthesis , Genome, Viral/physiology , Human papillomavirus 16/physiology , Nuclear Proteins/metabolism , Papillomavirus Infections/metabolism , Transcription Factors/metabolism , Virus Replication/physiology , Cell Cycle Proteins , Cell Line , Chromatin/genetics , Chromatin/metabolism , Chromatin/virology , DNA, Viral/genetics , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Humans , Nuclear Proteins/genetics , Oncogene Proteins, Viral/genetics , Oncogene Proteins, Viral/metabolism , Papillomavirus Infections/genetics , Transcription Factors/genetics
16.
J Virol ; 87(3): 1497-507, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23152516

ABSTRACT

The p300, CBP, and pCAF lysine acetyltransferase (KAT) proteins have been reported to physically interact with bovine (BPV) and human (HPV) papillomavirus E2 proteins. While overexpression of these KAT proteins enhances E2-dependent transcription, the mechanism has not been determined. Using RNA interference (RNAi) to deplete these factors, we demonstrated that E2 transcriptional activity requires physiological levels of p300, CBP, and pCAF. Each protein appears to have a unique function in E2-dependent transcription, since overexpression of one KAT failed to compensate for RNAi knockdown of another KAT. Using an in vitro acetylation assay, we identified highly conserved lysines that are targeted by p300 for acetylation. The conservative changes of lysines at positions 111 and 112 to arginine were of particular interest. The K111R and the K111R/K112R mutants showed reduced transcriptional activity that was not responsive to p300 overexpression, while the K112R mutant retained activity. p300 and CBP were detected at the viral promoter; however, pCAF was not. We propose a model by which E2 transcriptional activity is controlled by p300-mediated acetylation of lysine 111. This model represents a novel mechanism regulating papillomavirus gene expression.


Subject(s)
Bovine papillomavirus 1/genetics , DNA-Binding Proteins/metabolism , Gene Expression Regulation, Viral , Host-Pathogen Interactions , Lysine/metabolism , Transcription, Genetic , Viral Proteins/metabolism , p300-CBP Transcription Factors/metabolism , Acetylation , Animals , Cattle , Cell Line
17.
ACS Pharmacol Transl Sci ; 7(4): 1178-1190, 2024 Apr 12.
Article in English | MEDLINE | ID: mdl-38633583

ABSTRACT

A promising alternative for cancer treatment involves targeted inhibition of the epigenetic regulator bromodomain-containing protein 4 (BRD4); however, available BRD4 inhibitors are constrained by their potency, oral bioavailability, and cytotoxicity. Herein, to overcome the drawback of the translational BRD4 inhibitors, we describe a novel BRD4-p53 inhibitor, SDU-071, which suppresses BRD4 interaction with the p53 tumor suppressor and its biological activity in MDA-MB-231 triple-negative breast cancer (TNBC) cells in vitro and in vivo. This novel small-molecule BRD4-p53 inhibitor suppresses cell proliferation, migration, and invasion by downregulating the expression of BRD4-targeted genes, such as c-Myc and Mucin 5AC, and inducing cell cycle arrest and apoptosis, as demonstrated in cultured MDA-MB-231 TNBC cells. Its antitumor activity is illustrated in an orthotopic mouse xenograft mammary tumor model. Overall, our results show that SDU-071 is a viable option for potentially treating TNBC as a new BRD4-p53 inhibitor.

18.
EMBO J ; 28(9): 1246-59, 2009 May 06.
Article in English | MEDLINE | ID: mdl-19339993

ABSTRACT

Covalent modification by small ubiquitin-related modifiers (SUMO) regulates p53 transcription activity through an undefined mechanism. Using reconstituted sumoylation components, we purified SUMO-1-conjugated p53 (Su-p53) to near homogeneity. Su-p53 exists in solution as a tetramer and interacts with p300 histone acetyltransferase as efficiently as the unmodified protein. Nevertheless, it fails to activate p53-dependent chromatin transcription because of its inability to bind DNA. With sequential modification assays, we found that sumoylation of p53 at K386 blocks subsequent acetylation by p300, whereas p300-acetylated p53 remains permissive for ensuing sumoylation at K386 and alleviates sumoylation-inhibited DNA binding. While preventing the free form of p53 from accessing its cognate sites, sumoylation fails to disengage prebound p53 from DNA. The sumoylation-deficient K386R protein, when expressed in p53-null cells, exhibits higher transcription activity and binds better to the endogenous p21 gene compared with the wild-type protein. These studies unravel a molecular mechanism underlying sumoylation-regulated p53 function and further uncover a new role of acetylation in antagonizing the inhibitory effect of sumoylation on p53 binding to DNA.


Subject(s)
Chromatin/genetics , DNA/metabolism , Small Ubiquitin-Related Modifier Proteins/metabolism , Transcription, Genetic/genetics , Tumor Suppressor Protein p53/metabolism , Acetylation , Cell Line, Tumor , Chromatin Immunoprecipitation , Chromatography, Gel , Electrophoretic Mobility Shift Assay , Humans , Protein Binding , Reverse Transcriptase Polymerase Chain Reaction , p300-CBP Transcription Factors/metabolism
19.
bioRxiv ; 2023 May 26.
Article in English | MEDLINE | ID: mdl-37292798

ABSTRACT

During the human papillomavirus 16 life cycle, the E2 protein binds simultaneously to the viral genome and host chromatin throughout mitosis, ensuring viral genomes reside in daughter cell nuclei following cell division. Previously, we demonstrated that CK2 phosphorylation of E2 on serine 23 promotes interaction with TopBP1, and that this interaction is required for optimum E2 mitotic chromatin association and plasmid segregation function. Others have implicated BRD4 in mediating the plasmid segregation function of E2 and we have demonstrated that there is a TopBP1-BRD4 complex in the cell. We therefore further investigated the role of the E2-BRD4 interaction in mediating E2 association with mitotic chromatin and plasmid segregation function. Using a combination of immunofluorescence and our novel plasmid segregation assay in U2OS and N/Tert-1 cells stably expressing a variety of E2 mutants, we report that direct interaction with the BRD4 carboxyl-terminal motif (CTM) and TopBP1 is required for E2 association with mitotic chromatin and plasmid segregation. We also identify a novel TopBP1 mediated interaction between E2 and the BRD4 extra-terminal (ET) domain in vivo . Overall, the results demonstrate that direct interaction with TopBP1 and the BRD4 CTM are required for E2 mitotic chromatin association and plasmid segregation function. Disruption of this complex offers therapeutic options for targeting segregation of viral genomes into daughter cells, potentially combatting HPV16 infections, and cancers that retain episomal genomes. Importance: HPV16 is a causative agent in around 3-4% of all human cancers and currently there are no anti-viral therapeutics available for combating this disease burden. In order to identify new therapeutic targets, we must increase our understanding of the HPV16 life cycle. Previously, we demonstrated that an interaction between E2 and the cellular protein TopBP1 mediates the plasmid segregation function of E2, allowing distribution of viral genomes into daughter nuclei following cell division. Here, we demonstrate that E2 interaction with an additional host protein, BRD4, is also essential for E2 segregation function, and that BRD4 exists in a complex with TopBP1. Overall, these results enhance our understanding of a critical part of the HPV16 life cycle and presents several therapeutic targets for disruption of the viral life cycle.

20.
J Biol Chem ; 286(47): 40974-86, 2011 Nov 25.
Article in English | MEDLINE | ID: mdl-21937452

ABSTRACT

Activator protein-1 (AP-1) regulates diverse gene responses triggered by environmental cues and virus-induced cellular stress. Although many signaling events leading to AP-1 activation have been described, the fundamental features underlying binding site selection and factor recruitment of dimeric AP-1 complexes to their target genes remain mostly uncharacterized. Using recombinant full-length human AP-1 dimers formed between c-Jun and Fos family members (c-Fos, FosB, Fra-1, Fra-2) for DNA binding and transcriptional analysis, we found that each of these AP-1 complex exhibits differential activity for distinct non-consensus AP-1 sites present in human papillomavirus (HPV), and each AP-1 complex is capable of activating transcription from in vitro-reconstituted HPV chromatin in a p300- and acetyl-CoA-dependent manner. Transcription from HPV chromatin requires AP-1-dependent and contact-driven recruitment of p300. Acetylation of dimeric AP-1 complexes by p300 enhances AP-1 binding to DNA. Using a human C-33A cervical cancer-derived cell line harboring the episomal HPV type 11 genome, we illustrate binding site selectivity recognized by c-Jun, JunB, JunD, and various Fos family members in a combinatorial and unique pattern, highlighting the diversity and importance of non-canonical binding site recognition by various AP-1 family proteins.


Subject(s)
Chromatin/metabolism , Human papillomavirus 11/genetics , Transcription Factor AP-1/chemistry , Transcription Factor AP-1/metabolism , Transcription, Genetic , Base Sequence , Binding Sites , Cell Survival , Chromatin/genetics , Condylomata Acuminata/virology , Conserved Sequence , DNA, Viral/genetics , DNA, Viral/metabolism , E1A-Associated p300 Protein/metabolism , HCT116 Cells , HeLa Cells , Histones/metabolism , Humans , Nucleosome Assembly Protein 1/metabolism , Protein Multimerization , Protein Structure, Quaternary , Proto-Oncogene Proteins c-fos/metabolism , Proto-Oncogene Proteins c-jun/metabolism , RNA-Binding Proteins/metabolism , Regulatory Sequences, Nucleic Acid/genetics , Substrate Specificity
SELECTION OF CITATIONS
SEARCH DETAIL