Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 30
Filter
Add more filters










Publication year range
1.
J Mol Biol ; 433(18): 167119, 2021 09 03.
Article in English | MEDLINE | ID: mdl-34181981

ABSTRACT

The E2F1 transcription factor is a master regulator of cell-cycle progression whose uncontrolled activation contributes to tumor cells growth. E2F1 binds DNA as a heterodimer with DP partners, resulting in a multi-domain quaternary-structure complex composed of DNA binding domains, a coiled coil domain and a marked box domain separated by short linkers. Building on the 3D knowledge of the single domains of E2F and DPs, we characterized the structure and dynamics of the complete E2F1/DP1/DNA complex by a combination of small-angle X-ray scattering and molecular dynamics simulations. It shows an asymmetric contribution of the dynamics of the two proteins. Namely, the coiled-coil domain leans toward the DP1 side of the complex; the DP1 loop between α2 and α3 of the DBD partially populates a helical structure leaning far from the DNA and in the same direction of the coiled-coil domain; and the N-terminal disordered region of DP1, rich in basic residues, contributes to DNA binding stabilization. Intriguingly, tumor mutations in the flexible regions of the complex suggest that perturbation of protein dynamics could affect protein function in a context-dependent way. Our data suggest fundamental contributions of DP proteins in distinct aspects of E2F biology.


Subject(s)
DNA/chemistry , DNA/metabolism , E2F1 Transcription Factor/chemistry , E2F1 Transcription Factor/metabolism , Transcription Factor DP1/chemistry , Transcription Factor DP1/metabolism , Cell Cycle , Humans , Models, Molecular , Molecular Dynamics Simulation , Multiprotein Complexes/chemistry , Multiprotein Complexes/metabolism , Nucleic Acid Conformation , Phosphorylation , Protein Binding , Protein Conformation
2.
Biochemistry ; 60(9): 648-662, 2021 03 09.
Article in English | MEDLINE | ID: mdl-33620209

ABSTRACT

Almost all eukaryotic proteins receive diverse post-translational modifications (PTMs) that modulate protein activity. Many histone PTMs are well characterized, heavily influence gene regulation, and are often predictors of distinct transcriptional programs. Although our understanding of the histone PTM network has matured, much is yet to be understood about the roles of transcription factor (TF) PTMs, which might well represent a similarly complex and dynamic network of functional regulation. Members of the bromodomain and extra-terminal domain (BET) family of proteins recognize acetyllysine residues and relay the signals encoded by these modifications. Here, we have investigated the acetylation dependence of several functionally relevant BET-TF interactions in vitro using surface plasmon resonance, nuclear magnetic resonance, and X-ray crystallography. We show that motifs known to be acetylated in TFs E2F1 and MyoD1 can interact with all bromodomains of BRD2, BRD3, and BRD4. The interactions are dependent on diacetylation of the motifs and show a preference for the first BET bromodomain. Structural mapping of the interactions confirms a conserved mode of binding for the two TFs to the acetyllysine binding pocket of the BET bromodomains, mimicking that of other already established functionally important histone- and TF-BET interactions. We also examined a motif from the TF RelA that is known to be acetylated but were unable to observe any interaction, regardless of the acetylation state of the sequence. Our findings overall advance our understanding of BET-TF interactions and suggest a physical link between the important diacetylated motifs found in E2F1 and MyoD1 and the BET-family proteins.


Subject(s)
Cell Cycle Proteins/metabolism , E2F1 Transcription Factor/metabolism , MyoD Protein/metabolism , Transcription Factors/metabolism , Acetylation , Cell Cycle Proteins/chemistry , Crystallography, X-Ray , E2F1 Transcription Factor/chemistry , Histones/chemistry , Humans , Lysine/chemistry , Models, Molecular , MyoD Protein/chemistry , Protein Conformation , Protein Domains , Protein Processing, Post-Translational , Transcription Factors/chemistry
3.
Proc Natl Acad Sci U S A ; 117(26): 14958-14969, 2020 06 30.
Article in English | MEDLINE | ID: mdl-32541040

ABSTRACT

Oxidative stress is a ubiquitous threat to all aerobic organisms and has been implicated in numerous pathological conditions such as cancer. Here we demonstrate a pivotal role for E2F1, a cell cycle regulatory transcription factor, in cell tolerance of oxidative stress. Cells lacking E2F1 are hypersensitive to oxidative stress due to the defects in cell cycle arrest. Oxidative stress inhibits E2F1 transcriptional activity, independent of changes in association with Rb and without decreasing its DNA-binding activity. Upon oxidative insult, SUMO2 is extensively conjugated to E2F1 mainly at lysine 266 residue, which specifically modulates E2F1 transcriptional activity to enhance cell cycle arrest for cell survival. We identify SENP3, a desumoylating enzyme, as an E2F1-interacting partner. Oxidative stress inhibits the interaction between E2F1 and SENP3, which leads to accumulation of sumoylated E2F1. SENP3-deficient cells exhibit hypersumoylation of E2F1 and are resistant to oxidative insult. High levels of SENP3 in breast cancer are associated with elevated levels of E2F targets, high tumor grade, and poor survival. Given the prevalence of elevated levels of SENP3 across numerous cancer types, the SENP3-E2F1 axis may serve as an avenue for therapeutic intervention in cancer.


Subject(s)
E2F1 Transcription Factor/metabolism , Oxidative Stress , Amino Acid Motifs , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Cysteine Endopeptidases/genetics , Cysteine Endopeptidases/metabolism , E2F1 Transcription Factor/chemistry , E2F1 Transcription Factor/genetics , Female , Humans , Protein Binding , Small Ubiquitin-Related Modifier Proteins/genetics , Small Ubiquitin-Related Modifier Proteins/metabolism , Sumoylation
4.
PLoS One ; 13(10): e0206253, 2018.
Article in English | MEDLINE | ID: mdl-30359437

ABSTRACT

The cellular inhibitor of apoptosis 1 (cIAP1) is an E3-ubiquitin ligase that regulates cell signaling pathways involved in fundamental cellular processes including cell death, cell proliferation, cell differentiation and inflammation. It recruits ubiquitination substrates thanks to the presence of three baculoviral IAP repeat (BIR) domains at its N-terminal extremity. We previously demonstrated that cIAP1 promoted the ubiquitination of the E2 factor 1 (E2F1) transcription factor. Moreover, we showed that cIAP1 was required for E2F1 stabilization during the S phase of cell cycle and in response to DNA damage. Here, we report that E2F1 binds within the cIAP1 BIR3 domain. The BIR3 contains a surface hydrophobic groove that specifically anchors a conserved IAP binding motif (IBM) found in a number of intracellular proteins including Smac. The Smac N-7 peptide that includes the IBM, as well as a Smac mimetic, competed with E2F1 for interaction with cIAP1 demonstrating the importance of the BIR surface hydrophobic groove. We demonstrated that the first alpha-helix of BIR3 was required for E2F1 binding, as well as for the binding of Smac and Smac mimetics. Overexpression of cIAP1 modified the ubiquitination profile of E2F1, increasing the ratio of E2F1 conjugated with K11- and K63-linked ubiquitin chains, and decreasing the proportion of E2F1 modified by K48-linked ubiquitin chains. ChIP-seq analysis demonstrated that cIAP1 was required for the recruitment of E2F1 onto chromatin. Lastly, we identified an E2F-binding site on the cIAP1-encoding birc2 gene promoter, suggesting a retro-control regulation loop.


Subject(s)
Chromatin/metabolism , E2F1 Transcription Factor/metabolism , Inhibitor of Apoptosis Proteins/metabolism , Binding Sites , Cell Communication/genetics , Cell Line , E2F1 Transcription Factor/chemistry , HeLa Cells , Humans , Inhibitor of Apoptosis Proteins/chemistry , Inhibitor of Apoptosis Proteins/genetics , Protein Binding , Protein Domains , Signal Transduction , Ubiquitination
5.
Sci Rep ; 8(1): 8438, 2018 05 31.
Article in English | MEDLINE | ID: mdl-29855511

ABSTRACT

The transcription factor E2F plays crucial roles in cell proliferation and tumor suppression by activating growth-related genes and pro-apoptotic tumor suppressor genes, respectively. It is generally accepted that E2F binds to target sequences with its heterodimeric partner DP. Here we show that, while knockdown of DP1 expression inhibited ectopic E2F1- or adenovirus E1a-induced expression of the CDC6 gene and cell proliferation, knockdown of DP1 and DP2 expression did not affect ectopic E2F1- or E1a-induced expression of the tumor suppressor ARF gene, an upstream activator of the tumor suppressor p53, activation of p53 or apoptosis. These observations suggest that growth related and pro-apoptotic E2F targets are regulated by distinct molecular mechanisms and contradict the threshold model, which postulates that E2F activation of pro-apoptotic genes requires a higher total activity of activator E2Fs, above that necessary for E2F-dependent activation of growth-related genes.


Subject(s)
E2F1 Transcription Factor/metabolism , Transcription Factor DP1/metabolism , Tumor Suppressor Protein p53/metabolism , ADP-Ribosylation Factors/genetics , ADP-Ribosylation Factors/metabolism , Apoptosis , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Cell Line , DNA-Binding Proteins/antagonists & inhibitors , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Dimerization , E2F1 Transcription Factor/chemistry , E2F1 Transcription Factor/genetics , Fibroblasts/cytology , Fibroblasts/metabolism , Humans , Promoter Regions, Genetic , RNA Interference , RNA, Small Interfering/metabolism , Transcription Factor DP1/antagonists & inhibitors , Transcription Factor DP1/genetics , Transcription Factors/antagonists & inhibitors , Transcription Factors/genetics , Transcription Factors/metabolism
6.
Neuro Oncol ; 20(9): 1173-1184, 2018 08 02.
Article in English | MEDLINE | ID: mdl-29547938

ABSTRACT

Background: The cancer-testis specific gene Opa interacting protein 5 (OIP5) is reactivated in many human cancers, but its functions in glioblastoma remain unclear. Here, we assessed the significance of OIP5 in the tumorigenesis and metastasis of glioblastoma for the first time. Methods: An immunohistochemistry assay was performed to detect OIP5 expression changes in glioblastoma patients. Overall survival analysis was performed to evaluate the prognostic significance of OIP5. Growth curve, colony formation, and transwell assays were used to analyze cell proliferation and metastasis. Tumorigenicity potential was investigated in orthotopic tumor models, and immunoprecipitation, chromatin immunoprecipitation, and luciferase assays were employed to explore the mechanisms underlying the activation of OIP5 expression by E2F transcription factor 1 (E2F1) to stabilize and maintain E2F1 signaling. Results: OIP5 was found to be upregulated in glioblastoma patients and to impair patient survival, and the increased expression of OIP5 was positively correlated with tumor stage. Compared with short hairpin green fluorescent protein cells, cells in which OIP5 was knocked down exhibited significantly reduced proliferation, metastasis, colony formation, and tumorigenicity abilities, whereas OIP5 recovery enhanced these abilities. OIP5 was highly correlated with cell cycle progression but had no obvious effects on apoptosis. Notably, we demonstrated a feedback loop in which E2F1 activates the expression of OIP5 to stabilize and maintain E2F1 signaling and promote the E2F1-regulated gene expression that is required for aggressive tumor biology. Conclusions: Collectively, our findings demonstrate that OIP5 promotes glioblastoma progression and metastasis, suggesting that OIP5 is a potential target for anticancer therapy.


Subject(s)
Biomarkers, Tumor/metabolism , Carcinogenesis/pathology , Chromosomal Proteins, Non-Histone/metabolism , E2F1 Transcription Factor/chemistry , E2F1 Transcription Factor/metabolism , Gene Expression Regulation, Neoplastic , Glioblastoma/secondary , Animals , Apoptosis , Biomarkers, Tumor/genetics , Carcinogenesis/metabolism , Case-Control Studies , Cell Cycle , Cell Cycle Proteins , Cell Movement , Cell Proliferation , Chromosomal Proteins, Non-Histone/genetics , E2F1 Transcription Factor/genetics , Glioblastoma/metabolism , Glioblastoma/pathology , Humans , Mice , Mice, Nude , Neoplasm Invasiveness , Neoplasm Metastasis , Prognosis , Protein Stability , Survival Rate , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
7.
Eur Rev Med Pharmacol Sci ; 22(2): 299-306, 2018 Jan.
Article in English | MEDLINE | ID: mdl-29424887

ABSTRACT

OBJECTIVE: miR-205 has been previously identified as a diagnostic and prognostic factor in glioma. However, its exact functions in glioma remain unclear. The current research aimed to decipher the role of miR-205 in the development of cisplatin resistance in glioma cells. MATERIALS AND METHODS: miR-205 expressions in both cisplatin sensitive and resistant cell lines were compared by the Real-time PCR method. The dose-response to cisplatin of U87/DDP cells was determined by MTT assay. Cell cycle and apoptosis were determined by flow cytometry, caspase 3/7 activity assay and Western blot assay. The direct repression of E2F1 by miR-205 was confirmed by luciferase assay and Western blot assay. RESULTS: miR-205 expression was decreased in cisplatin resistant glioma cell lines, and cisplatin treatment led to a decrease of miR-205 in glioma cells. Overexpression of miR-205 in U87/DDP restored its cisplatin sensitivity by enhancing apoptosis and G1/S cell cycle arrest; notably, all these effects were then partially abrogated by E2F1 overexpression. Luciferase assay and Western blot assay confirmed E2F1 as the direct target of miR-205 in U87/DDP cells. CONCLUSIONS: These findings suggest that down-regulation of miR-205 confers the cisplatin resistance in glioma cells via upregulation of E2F1. It might serve as a candidate for glioma therapy.


Subject(s)
Drug Resistance, Neoplasm/genetics , E2F1 Transcription Factor/metabolism , MicroRNAs/metabolism , 3' Untranslated Regions , Apoptosis/drug effects , Base Sequence , Cell Line, Tumor , Cisplatin/pharmacology , Down-Regulation/drug effects , E2F1 Transcription Factor/chemistry , E2F1 Transcription Factor/genetics , G1 Phase Cell Cycle Checkpoints , Genetic Vectors/genetics , Genetic Vectors/metabolism , Glioma/genetics , Glioma/pathology , Humans , MicroRNAs/genetics , Sequence Alignment , Up-Regulation/drug effects
8.
PLoS Genet ; 14(2): e1007204, 2018 02.
Article in English | MEDLINE | ID: mdl-29420631

ABSTRACT

Across metazoans, cell cycle progression is regulated by E2F family transcription factors that can function as either transcriptional activators or repressors. For decades, the Drosophila E2F family has been viewed as a streamlined RB/E2F network, consisting of one activator (dE2F1) and one repressor (dE2F2). Here, we report that an uncharacterized isoform of dE2F1, hereon called dE2F1b, plays an important function during development and is functionally distinct from the widely-studied dE2F1 isoform, dE2F1a. dE2F1b contains an additional exon that inserts 16 amino acids to the evolutionarily conserved Marked Box domain. Analysis of de2f1b-specific mutants generated via CRISPR/Cas9 indicates that dE2F1b is a critical regulator of the cell cycle during development. This is particularly evident in endocycling salivary glands in which a tight control of dE2F1 activity is required. Interestingly, close examination of mitotic tissues such as eye and wing imaginal discs suggests that dE2F1b plays a repressive function as cells exit from the cell cycle. We also provide evidence demonstrating that dE2F1b differentially interacts with RBF1 and alters the recruitment of RBF1 and dE2F1 to promoters. Collectively, our data suggest that dE2F1b is a novel member of the E2F family, revealing a previously unappreciated complexity in the Drosophila RB/E2F network.


Subject(s)
Alternative Splicing/genetics , Cell Cycle/genetics , E2F1 Transcription Factor/genetics , Animals , Animals, Genetically Modified , Cell Division/genetics , Cells, Cultured , Drosophila melanogaster/embryology , Drosophila melanogaster/genetics , E2F1 Transcription Factor/chemistry , Embryo, Nonmammalian , Eye/embryology , Eye/metabolism , Gene Expression Regulation, Developmental , Gene Regulatory Networks , Organogenesis/genetics , Protein Domains/genetics , Retinoblastoma Protein/physiology
10.
EMBO Rep ; 19(2): 234-243, 2018 02.
Article in English | MEDLINE | ID: mdl-29233828

ABSTRACT

E2F1 is the main pro-apoptotic effector of the pRB-regulated tumor suppressor pathway by promoting the transcription of various pro-apoptotic proteins. We report here that E2F1 partly localizes to mitochondria, where it favors mitochondrial outer membrane permeabilization. E2F1 interacts with BCL-xL independently from its BH3 binding interface and induces a stabilization of BCL-xL at mitochondrial membranes. This prevents efficient control of BCL-xL over its binding partners, in particular over BAK resulting in the induction of cell death. We thus identify a new, non-BH3-binding regulator of BCL-xL localization dynamics that influences its anti-apoptotic activity.


Subject(s)
Cell Death , E2F1 Transcription Factor/metabolism , bcl-X Protein/metabolism , Apoptosis , Cell Line, Tumor , E2F1 Transcription Factor/chemistry , Extracellular Space/metabolism , Gene Expression Regulation/drug effects , Humans , Mitochondria/metabolism , Protein Binding , Protein Transport , Proto-Oncogene Proteins c-bcl-2/genetics , Proto-Oncogene Proteins c-bcl-2/metabolism , Transcription, Genetic , bcl-2 Homologous Antagonist-Killer Protein/metabolism , bcl-X Protein/chemistry
11.
PLoS One ; 12(7): e0181153, 2017.
Article in English | MEDLINE | ID: mdl-28704519

ABSTRACT

BACKGROUND: Genetic studies have largely concentrated on the impact of somatic mutations found in coding regions, and have neglected mutations outside of these. However, 3' untranslated regions (3' UTR) mutations can also disrupt or create miRNA target sites, and trigger oncogene activation or tumor suppressor inactivation. METHODS: We used next-generation sequencing to widely screen for genetic alterations within predicted miRNA target sites of oncogenes associated with colorectal cancer, and evaluated the functional impact of a new somatic mutation. Target sequencing of 47 genes was performed for 29 primary colorectal tumor samples. For 71 independent samples, Sanger methodology was used to screen for E2F1 mutations in miRNA predicted target sites, and the functional impact of these mutations was evaluated by luciferase reporter assays. RESULTS: We identified germline and somatic alterations in E2F1. Of the 100 samples evaluated, 3 had germline alterations at the MIR205-5p target site, while one had a somatic mutation at MIR136-5p target site. E2F1 gene expression was similar between normal and tumor tissues bearing the germline alteration; however, expression was increased 4-fold in tumor tissue that harbored a somatic mutation compared to that in normal tissue. Luciferase reporter assays revealed both germline and somatic alterations increased E2F1 activity relative to wild-type E2F1. CONCLUSIONS: We demonstrated that somatic mutation within E2F1:MIR136-5p target site impairs miRNA-mediated regulation and leads to increased gene activity. We conclude that somatic mutations that disrupt miRNA target sites have the potential to impact gene regulation, highlighting an important mechanism of oncogene activation.


Subject(s)
Colorectal Neoplasms/genetics , E2F1 Transcription Factor/genetics , High-Throughput Nucleotide Sequencing/methods , Mutation , Sequence Analysis, DNA/methods , 3' Untranslated Regions , Aged , Binding Sites , E2F1 Transcription Factor/chemistry , Female , Gene Expression Regulation, Neoplastic , Genetic Predisposition to Disease , Humans , Male , MicroRNAs/metabolism , Middle Aged , Up-Regulation
12.
Oncotarget ; 8(3): 4977-4993, 2017 Jan 17.
Article in English | MEDLINE | ID: mdl-27903963

ABSTRACT

The E2F1 transcription factor plays key roles in skin homeostasis. In the epidermis, E2F1 expression is essential for normal proliferation of undifferentiated keratinocytes, regeneration after injury and DNA repair following UV radiation-induced photodamage. Abnormal E2F1 expression promotes nonmelanoma skin carcinoma. In addition, E2F1 must be downregulated for proper keratinocyte differentiation, but the relevant mechanisms involved remain poorly understood. We show that differentiation signals induce a series of post-translational modifications in E2F1 that are jointly required for its downregulation. Analysis of the structural determinants that govern these processes revealed a central role for S403 and T433. In particular, substitution of these two amino acid residues with non-phosphorylatable alanine (E2F1 ST/A) interferes with E2F1 nuclear export, K11- and K48-linked polyubiquitylation and degradation in differentiated keratinocytes. In contrast, replacement of S403 and T433 with phosphomimetic aspartic acid to generate a pseudophosphorylated E2F1 mutant protein (E2F1 ST/D) generates a protein that is regulated in a manner indistinguishable from that of wild type E2F1. Cdh1 is an activating cofactor that interacts with the anaphase-promoting complex/cyclosome (APC/C) ubiquitin E3 ligase, promoting proteasomal degradation of various substrates. We found that Cdh1 associates with E2F1 in keratinocytes. Inhibition or RNAi-mediated silencing of Cdh1 prevents E2F1 degradation in response to differentiation signals. Our results reveal novel regulatory mechanisms that jointly modulate post-translational modifications and downregulation of E2F1, which are necessary for proper epidermal keratinocyte differentiation.


Subject(s)
Cdh1 Proteins/metabolism , Cell Nucleus/metabolism , E2F1 Transcription Factor/chemistry , E2F1 Transcription Factor/metabolism , Keratinocytes/cytology , Active Transport, Cell Nucleus , Amino Acid Substitution , Animals , Cell Differentiation , Cells, Cultured , DNA Damage , Down-Regulation , E2F1 Transcription Factor/genetics , Keratinocytes/metabolism , Lysine/metabolism , Mice , Protein Processing, Post-Translational , Proteolysis , Serine/metabolism , Threonine/metabolism
13.
Sci Adv ; 2(2): e1501257, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26989780

ABSTRACT

Peptidyl arginine deiminase 4 (PAD4) is a nuclear enzyme that converts arginine residues to citrulline. Although increasingly implicated in inflammatory disease and cancer, the mechanism of action of PAD4 and its functionally relevant pathways remains unclear. E2F transcription factors are a family of master regulators that coordinate gene expression during cellular proliferation and diverse cell fates. We show that E2F-1 is citrullinated by PAD4 in inflammatory cells. Citrullination of E2F-1 assists its chromatin association, specifically to cytokine genes in granulocyte cells. Mechanistically, citrullination augments binding of the BET (bromodomain and extra-terminal domain) family bromodomain reader BRD4 (bromodomain-containing protein 4) to an acetylated domain in E2F-1, and PAD4 and BRD4 coexist with E2F-1 on cytokine gene promoters. Accordingly, the combined inhibition of PAD4 and BRD4 disrupts the chromatin-bound complex and suppresses cytokine gene expression. In the murine collagen-induced arthritis model, chromatin-bound E2F-1 in inflammatory cells and consequent cytokine expression are diminished upon small-molecule inhibition of PAD4 and BRD4, and the combined treatment is clinically efficacious in preventing disease progression. Our results shed light on a new transcription-based mechanism that mediates the inflammatory effect of PAD4 and establish the interplay between citrullination and acetylation in the control of E2F-1 as a regulatory interface for driving inflammatory gene expression.


Subject(s)
Citrulline/metabolism , E2F1 Transcription Factor/chemistry , E2F1 Transcription Factor/metabolism , Inflammation/metabolism , Acetylation , Animals , Arthritis, Experimental/genetics , Arthritis, Experimental/immunology , Arthritis, Experimental/metabolism , Cell Cycle Proteins , Cell Line , Cytokines/genetics , E2F1 Transcription Factor/genetics , Gene Expression Regulation , HL-60 Cells , Humans , Hydrolases/antagonists & inhibitors , Hydrolases/genetics , Hydrolases/metabolism , Inflammation/genetics , Inflammation/immunology , Male , Mice , Mice, Inbred DBA , Nuclear Proteins/metabolism , Promoter Regions, Genetic , Protein-Arginine Deiminase Type 4 , Protein-Arginine Deiminases , RNA, Small Interfering/genetics , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Transcription Factors/metabolism
14.
Int J Oncol ; 48(4): 1650-8, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26891801

ABSTRACT

Increased expression of E2F1 has been reported to be associated with tumor growth and cell survival of prostate cancer (PCa). However, its roles and mechanisms on PCa have not been fully elucidated. The present study found that E2F1 overexpression in PCa tissues was significantly associated with high Gleason score (P=0.01) and advanced pathological stage (P=0.02). In addition, PCa patients with high E2F1 expression more frequently had shorter biochemical recurrence-free survival (P=0.047) than those with low E2F1 expression. Then, we confirmed that the knock-down of E2F1 expression was able to inhibit cell cycle progression, invasion and migration of PCa cell lines in vitro, along with tumor xenograft growth and epithelial-to-mesenchymal transition (EMT) in vivo. Moreover, we identified CD147 as a novel interaction partner for E2F1 through bio-informatic binding site prediction, combined with chromatin immunoprecipitation-PCR (ChIP-PCR) and western blot analysis. Taken together, our data delineate an as yet unrecognized function of E2F1 as enhancer of tumor invasion and migration of PCa via regulating the expression of CD147 in PCa. Importantly, E2F1 may function as a biomarker that can differentiate patients with biochemical recurrent and non-biochemical recurrent disease following radical prostatectomy, highlighting its potential as a therapeutic target.


Subject(s)
Basigin/metabolism , E2F1 Transcription Factor/metabolism , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , Animals , Basigin/genetics , Binding Sites , Cell Line, Tumor , Cell Movement , E2F1 Transcription Factor/chemistry , Epithelial-Mesenchymal Transition , Gene Expression Regulation, Neoplastic , Humans , Male , Mice , Neoplasm Grading , Neoplasm Invasiveness , Prognosis , Prostatic Neoplasms/genetics , Survival Analysis
15.
Tumour Biol ; 37(3): 2823-31, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26749284

ABSTRACT

E2F is a family of transcription factors that recognized to regulate the expression of genes essential for a wide range of cellular functions, including cell cycle progression, DNA repair, DNA replication, differentiation, proliferation, and apoptosis. E2F1, the most classic member of the E2F family, exhibits a complex role in tumor development regulation. In recent years, a growing body of data suggested an intimate relationship between E2F1 and ovarian carcinoma. And E2F1 was well identified to play dual functions and serve as a useful prognostic indicator in ovarian carcinoma. However, the mechanism underlying E2F1 associated with ovarian carcinoma remains elusive. It is necessary to clarify the fundamental role of E2F1 in ovarian carcinoma. In this review, we tried to sum up the knowledge of E2F1, including its structure and related mechanism. We also attempt to absorb the research achievements and collect the mechanism of E2F1 in ovarian carcinoma.


Subject(s)
E2F1 Transcription Factor/physiology , Ovarian Neoplasms/etiology , E2F1 Transcription Factor/chemistry , Female , Genes, BRCA1 , Humans , Tumor Suppressor Protein p53/physiology
16.
Nat Commun ; 6: 8704, 2015 Oct 29.
Article in English | MEDLINE | ID: mdl-26510456

ABSTRACT

Hyperactivation of the transcriptional factor E2F1 occurs frequently in human cancers and contributes to malignant progression. E2F1 activity is regulated by proteolysis mediated by the ubiquitin-proteasome system. However, the deubiquitylase that controls E2F1 ubiquitylation and stability remains undefined. Here we demonstrate that the deubiquitylase POH1 stabilizes E2F1 protein through binding to and deubiquitylating E2F1. Conditional knockout of Poh1 alleles results in reduced E2F1 expression in primary mouse liver cells. The POH1-mediated regulation of E2F1 expression strengthens E2F1-downstream prosurvival signals, including upregulation of Survivin and FOXM1 protein levels, and efficiently facilitates tumour growth of liver cancer cells in nude mice. Importantly, human hepatocellular carcinomas (HCCs) recapitulate POH1 regulation of E2F1 expression, as nuclear abundance of POH1 is increased in HCCs and correlates with E2F1 overexpression and tumour growth. Thus, our study suggests that the hyperactivated POH1-E2F1 regulation may contribute to the development of liver cancer.


Subject(s)
Cell Proliferation , E2F1 Transcription Factor/metabolism , Liver Neoplasms/metabolism , Proteasome Endopeptidase Complex/metabolism , Trans-Activators/metabolism , Ubiquitin/metabolism , Animals , Carcinogenesis , E2F1 Transcription Factor/chemistry , E2F1 Transcription Factor/genetics , Forkhead Box Protein M1 , Forkhead Transcription Factors/genetics , Forkhead Transcription Factors/metabolism , Humans , Liver Neoplasms/genetics , Liver Neoplasms/physiopathology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Mice, Nude , Proteasome Endopeptidase Complex/genetics , Protein Binding , Protein Stability , Trans-Activators/genetics , Ubiquitination
17.
Oncogene ; 33(27): 3594-603, 2014 Jul 03.
Article in English | MEDLINE | ID: mdl-23934193

ABSTRACT

Transcription factor E2F-1 and its interaction with pRb provide a key point of control in cell proliferation. E2F-1 participates in both cell cycle progression and apoptosis, and in cells exists with a DP dimerization partner protein, the most prominent being DP-1. By mining the tumor tissue and cancer cell line encyclopedia genomic databases, we identified the first somatic mutations in the DP-1 gene and describe 53 distinct mutation events here. The mutations are mostly missense mutations, but also include nonsense and frame-shift mutations that result in truncated DP-1 derivatives. Mutation occurs throughout the DP-1 gene but generally leaves protein dimerization activity intact. This allows the mutant derivatives to affect the properties of the E2F-1/DP-1 heterodimer through a transdominant mechanism, which changes the DNA binding, transcriptional activation and pRb-binding properties of the heterodimer. In particular, many DP-1 mutants were found to impair E2F-1-dependent apoptosis. Our results establish that somatic mutations in DP-1 uncouple normal control of the E2F pathway, and thus define a new mechanism that could contribute to aberrant proliferation in tumor cells.


Subject(s)
E2F1 Transcription Factor/genetics , Genetic Pleiotropy , Mutation , Protein Subunits/genetics , Transcription Factor DP1/genetics , Amino Acid Sequence , Apoptosis , Cell Line, Tumor , E2F1 Transcription Factor/chemistry , E2F1 Transcription Factor/metabolism , Humans , Models, Molecular , Molecular Sequence Data , Phosphoproteins/metabolism , Protein Multimerization , Protein Structure, Quaternary , Protein Subunits/chemistry , Protein Subunits/metabolism , Retinoblastoma Protein/metabolism , Transcription Factor DP1/chemistry , Transcription Factor DP1/metabolism
18.
Cancer Biol Ther ; 14(8): 742-51, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23792570

ABSTRACT

E2F-1, a key transcription factor necessary for cell growth, DNA repair, and differentiation, is an attractive target for development of anticancer drugs in tumors that are E2F "oncogene addicted". We identified a peptide isolated from phage clones that bound tightly to the E2F-1 promoter consensus sequence. The peptide was coupled to penetratin to enhance cellular uptake. Modeling of the penetratin-peptide (PEP) binding to the DNA E2F-1 promoter demonstrated favorable interactions that also involved the participation of most of the penetratin sequence. The penetratin-peptide (PEP) demonstrated potent in vitro cytotoxic effects against a range of cancer cell lines, particularly against Burkitt lymphoma cells and small cell lung cancer (SCLC) cells. Further studies in the H-69 SCLC cell line showed that the PEP inhibited transcription of E2F-1 and also several important E2F-regulated enzymes involved in DNA synthesis, namely, thymidylate synthase, thymidine kinase, and ribonucleotide reductase. As the PEP was found to be relatively unstable in serum, it was encapsulated in PEGylated liposomes for in vivo studies. Treatment of mice bearing the human small cell lung carcinoma H-69 with the PEP encapsulated in PEGylated liposomes (PL-PEP) caused tumor regression without significant toxicity. The liposome encapsulated PEP has promise as an antitumor agent, alone or in combination with inhibitors of DNA synthesis.


Subject(s)
Carrier Proteins/metabolism , E2F1 Transcription Factor/metabolism , Lung Neoplasms/drug therapy , Peptide Fragments/pharmacology , Small Cell Lung Carcinoma/drug therapy , Amino Acid Sequence , Animals , Apoptosis/drug effects , Carrier Proteins/chemistry , Carrier Proteins/genetics , Cell-Penetrating Peptides , Down-Regulation , Drug Screening Assays, Antitumor , E2F1 Transcription Factor/biosynthesis , E2F1 Transcription Factor/chemistry , E2F1 Transcription Factor/genetics , Female , Humans , Lung Neoplasms/genetics , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Mice , Models, Molecular , Molecular Sequence Data , Molecular Targeted Therapy , Peptide Fragments/chemistry , Peptide Fragments/pharmacokinetics , Promoter Regions, Genetic , RNA, Messenger/genetics , RNA, Messenger/metabolism , Random Allocation , Small Cell Lung Carcinoma/genetics , Small Cell Lung Carcinoma/metabolism , Small Cell Lung Carcinoma/pathology , Xenograft Model Antitumor Assays
19.
Oncogene ; 32(30): 3520-30, 2013 Jul 25.
Article in English | MEDLINE | ID: mdl-22945643

ABSTRACT

Activation of the Wnt/ß-catenin pathway is a critical step in the development of colorectal cancers. A key mediator of this activation is the recently described oncogene CDK8, which is amplified in a large number of colorectal tumors. CDK8 affects ß-catenin activation by interaction of the CDK8 submodule of the mediator complex with ß-catenin/TCF transcriptional complex, and by CDK8 interacting with and phosphorylating E2F1, which acts as a repressor of ß-catenin/TCF transcriptional activity. The amino-acid residue in E2F1 that CDK8 phosphorylates and how this phosphorylation impacts E2F1 activity in general is not known. Here, we describe that CDK8 phosphorylates serine 375 in E2F1 both in vitro and in cells, and that phosphorylation of this residue is required for E2F1 interaction with CDK8, and that the phosphorylation is dependent on CDK8 kinase activity. The phosphorylation of S375 by CDK8 regulates E2F1 ability to repress transcription of ß-catenin/TCF-dependent genes, as well as activation of E2F1-dependent genes. This regulation is due to inactivation of E2F1 transcriptional activation, and not to the interference of E2F1's ability to bind to E2F1-binding sites in various promoters or to interact with DP1.


Subject(s)
Cyclin-Dependent Kinase 8/physiology , E2F1 Transcription Factor/metabolism , E2F1 Transcription Factor/physiology , Cyclin-Dependent Kinase 8/genetics , Cyclin-Dependent Kinase 8/metabolism , E2F1 Transcription Factor/chemistry , Humans , Phosphorylation/genetics , Serine/metabolism , Transcriptional Activation/genetics
20.
Exp Cell Res ; 318(10): 1146-59, 2012 Jun 10.
Article in English | MEDLINE | ID: mdl-22484375

ABSTRACT

Melanoma, the most aggressive form of skin cancer, is notoriously resistant to all current modalities of cancer therapy, including to the drug methotrexate. Melanosomal sequestration and cellular exportation of methotrexate have been proposed to be important melanoma-specific mechanisms that contribute to the resistance of melanoma to methotrexate. In addition, other mechanisms of resistance that are present in most epithelial cancer cells are also operative in melanoma. This report elucidates how melanoma orchestrates these mechanisms to become extremely resistant to methotrexate, where both E2F1 and checkpoint kinase 1 (Chk1), two molecules with dual roles in survival/apoptosis, play prominent roles. The results indicated that MTX induced the depletion of dihydrofolate in melanoma cells, which stimulated the transcriptional activity of E2F1. The elevate expression of dihydrofolate reductase and thymidylate synthase, two E2F1-target genes involved in folate metabolism and required for G(1) progression, favored dTTP accumulation, which promoted DNA single strand breaks and the subsequent activation of Chk1. Under these conditions, melanoma cells are protected from apoptosis by arresting their cell cycle in S phase. Excess of dTTP could also inhibit E2F1-mediated apoptosis in melanoma cells.


Subject(s)
Antimetabolites, Antineoplastic/pharmacology , Cell Survival/drug effects , Drug Resistance, Neoplasm , Melanoma/metabolism , Methotrexate/pharmacology , Skin Neoplasms/metabolism , Amino Acid Sequence , Animals , Apoptosis/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Checkpoint Kinase 1 , E2F1 Transcription Factor/chemistry , E2F1 Transcription Factor/metabolism , Enzyme Activation/drug effects , Folic Acid/metabolism , Humans , Melanoma/pathology , Mice , Molecular Sequence Data , Protein Kinases/metabolism , Protein Processing, Post-Translational , S Phase Cell Cycle Checkpoints , Skin Neoplasms/pathology , Tetrahydrofolate Dehydrogenase/genetics , Tetrahydrofolate Dehydrogenase/metabolism , Thymidylate Synthase/genetics , Thymidylate Synthase/metabolism , Thymine Nucleotides/metabolism , Transcription, Genetic
SELECTION OF CITATIONS
SEARCH DETAIL
...