Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 984
Filter
Add more filters

Publication year range
1.
Mol Psychiatry ; 27(6): 2777-2786, 2022 06.
Article in English | MEDLINE | ID: mdl-35365808

ABSTRACT

ARID4A plays an important role in regulating gene expression and cell proliferation. ARID4A belongs to the AT-rich interaction domain (ARID)-containing family, and a PWWP domain immediately precedes its ARID region. The molecular mechanism and structural basis of ARID4A are largely unknown. Whole-exome sequencing (WES) revealed that a novel heterozygous missense variant, ARID4A c.1231 C > G (p.His411Asp), was associated with schizophrenia (SCZ) in this study. We determined the crystal structure of the PWWP-ARID tandem at 2.05 Å, revealing an unexpected mode in which ARID4A assembles with its PWWP and ARID from a structural and functional supramodule. Our results further showed that compared with the wild type, the p.His411Asp ARID mutant protein adopts a less compact conformation and exhibits a weaker dsDNA-binding ability. The p.His411Asp mutation decreased the number of cells that were arrested in the G0-G1 phase and caused more cells to progress to the G2-M phase. In addition, the missense mutation promoted the proliferation of HEK293T cells. In conclusion, our data provide evidence that ARID4A p.His411Asp could cause a conformational change in the ARID4A ARID domain, influence the DNA binding function, and subsequently disturb the cell cycle arrest in the G1 phase. ARID4A is likely a susceptibility gene for SCZ; thus, these findings provide new insight into the role of ARID4A in psychiatric disorders.


Subject(s)
Mutation, Missense , Retinoblastoma-Binding Protein 1 , Schizophrenia , China , DNA , HEK293 Cells , Humans , Retinoblastoma-Binding Protein 1/genetics , Retinoblastoma-Binding Protein 1/metabolism , Schizophrenia/genetics , Schizophrenia/metabolism , Siblings
2.
J Biol Chem ; 296: 100506, 2021.
Article in English | MEDLINE | ID: mdl-33675746

ABSTRACT

Human ARID4A and ARID4B are homologous proteins that are important in controlling gene expression and epigenetic regulation but have distinct functions. Previous studies have shown that the N-terminal domain of ARID4A is an unusual interdigitated double Tudor domain with DNA-binding activity. However, how the Tudor domain of ARID4B differs from that of ARID4A remains unknown. Here, we found that the ARID4B Tudor domain has significantly weaker DNA affinity than the ARID4A Tudor domain despite sharing more than 80% sequence identity. Structure determination and DNA titration analysis indicated that the ARID4B Tudor domain is also an interdigitated double Tudor domain with a DNA-binding surface similar to ARID4A. We identified a residue close to the DNA-binding site of the Tudor domain that differs between ARID4A and ARID4B. The Leu50 in ARID4A is Glu50 in ARID4B, and the latter forms salt bridges with two lysine residues at the DNA-binding surface. This causes a decrease in the strength of positive charge, thus reducing DNA-binding affinity while significantly increasing protein stability. We also found that a C-terminal extension region enhances the DNA-binding affinity of the ARID4B Tudor domain. This C-terminal extension is disordered and contains a positively charged RGR motif, providing an additional DNA-binding site. Finally, sequence and phylogenetic analyses indicated that the residue differences and the presence of the RGR extension region are conserved. These results provide new insight into the functional differences between ARID4A and ARID4B proteins, as well as elucidating the function of the disordered regions in these proteins.


Subject(s)
Antigens, Neoplasm/chemistry , Antigens, Neoplasm/metabolism , DNA/metabolism , Neoplasm Proteins/chemistry , Neoplasm Proteins/metabolism , Tudor Domain , Amino Acid Sequence , DNA-Binding Proteins/chemistry , DNA-Binding Proteins/metabolism , Humans , Protein Binding , Protein Conformation , Retinoblastoma-Binding Protein 1/chemistry , Retinoblastoma-Binding Protein 1/metabolism , Sequence Homology
3.
J Cell Biochem ; 119(9): 7245-7255, 2018 09.
Article in English | MEDLINE | ID: mdl-29797600

ABSTRACT

AT-rich interaction domain 4A (ARID4A) and AT-rich interaction domain 4B (ARID4B), which are both the AT-rich interaction domain (ARID) family, have been reported to be oncogene or tumor suppressor gene in various human malignances, but there is no involvement about their functions in prostate cancer (PCa). Our previous study has reported that microRNA-30d (miR-30d) expression can predicted poor clinical prognosis in PCa, however, the underlying mechanisms of miR-30d have not been fully described. The aim of our study is to investigate the expression relevance between miR-30d and ARID4A or ARID4B, and examine the clinical significance and biological function of ARID4A and AIRD4B in PCa. In this study, both ARID4A and ARID4B were identified as the target genes of miR-30d. In addition, the mRNA expression of miR-30d in PCa tissues were significantly negative correlated with ARID4A (Pearson correlation coefficient = -0.313, P = 0.001) and ARID4B (Pearson correlation coefficient = -0.349, P < 0.001), while there was a positive correlation between ARID4A and ARID4B (Pearson correlation coefficient = 0.865, P < 0.001). Moreover, both ARID4A and ARID4B were significantly downregulated in PCa tissues with high Gleason scores (P = 0.005, P = 0.033), PSA failure (P = 0.012, P = 0.05) and short biochemical recurrent-free survival (P = 0.033, P = 0.031). Furthermore, the knockout expression of ARID4A and ARID4B promoted PCa cell proliferation, migration and invasion in vitro. In conclusion, our results indicated that ARID4A and ARID4B may serve as tumor suppressor in PCa progression, suggesting that they might be the potential therapeutic targets in prostate cancer.


Subject(s)
Antigens, Neoplasm/genetics , Antigens, Neoplasm/metabolism , MicroRNAs/genetics , MicroRNAs/metabolism , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Prostatic Neoplasms/genetics , Prostatic Neoplasms/metabolism , Retinoblastoma-Binding Protein 1/genetics , Retinoblastoma-Binding Protein 1/metabolism , Aged , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Cell Line, Tumor , Cell Movement , Cell Proliferation , Cohort Studies , Disease Progression , Down-Regulation , Gene Expression Regulation, Neoplastic , Gene Knockout Techniques , Genes, Tumor Suppressor , Humans , Kaplan-Meier Estimate , Male , Neoplasm Invasiveness , Statistics, Nonparametric
4.
Biochem Biophys Res Commun ; 496(4): 1344-1348, 2018 02 19.
Article in English | MEDLINE | ID: mdl-29408527

ABSTRACT

RBBP1 is a retinoblastoma protein (pRb) binding protein acting as a repressor of gene transcription. RBBP1 is a multidomain protein including a chromo barrel domain, and its chromo barrel domain has been reported to recognize histone H4K20me3 weakly, and this binding is enhanced by the simultaneous binding of DNA. However, the molecular basis of this DNA-mediated histone binding by the chromo barrel domain of RBBP1 is unclear. Here we attempted to co-crystallize the chromo barrel domain of RBBP1 with either a histone H4K20me3 peptide alone or with both a histone H4K20me3 peptide and DNA, but only solved the peptide/DNA unbound crystal structure. Our structural analysis indicates that RBBP1 could interact with histone H4K20me3 similar to other histone binding chromo barrel domains, and the surface charge representation analysis of the chromo barrel domain of RBBP1 suggests that the chromo barrel domain of RBBP1 does not have a typical DNA binding surface, indicating that it might not bind to DNA. Consistently, our ITC assays also showed that DNA does not significantly enhance the histone binding ability of the chromo barrel domain of RBBP1.


Subject(s)
DNA/chemistry , DNA/ultrastructure , Histones/chemistry , Histones/ultrastructure , Molecular Docking Simulation , Retinoblastoma-Binding Protein 1/chemistry , Retinoblastoma-Binding Protein 1/ultrastructure , Binding Sites , Models, Chemical , Protein Binding , Protein Conformation , Protein Domains
5.
Proc Natl Acad Sci U S A ; 110(12): 4616-21, 2013 Mar 19.
Article in English | MEDLINE | ID: mdl-23487765

ABSTRACT

ARID4A and ARID4B are homologous members of the ARID (AT-rich interaction domain) gene family. ARID4A and ARID4B physically interact with each other. ARID4A is a retinoblastoma (RB)-binding protein. Biological function of these interactions is still unknown. Here, we report that mice with complete deficiency of Arid4a, combined with haploinsufficiency of Arid4b (Arid4a(-/-)Arid4b(+/-)), showed progressive loss of male fertility, accompanied by hypogonadism and seminal vesicle agenesis/hypodysplasia. Arid4a and Arid4b are expressed mainly in Sertoli cells of testes, which implies that their roles in Sertoli cell function are to support spermatogenesis and create the impermeable blood-testis barrier. In fact, evaluation of germ cell development in the Arid4a(-/-)Arid4b(+/-) mice showed spermatogenic arrest at the stages of meiotic spermatocytes and postmeiotic haploid spermatids. Analysis of the integrity of the blood-testis barrier showed increased permeability of seminiferous tubules in the Arid4a(-/-)Arid4b(+/-) testes. Interestingly, phenotypic Sertoli cell dysfunction in the Arid4a(-/-)Arid4b(+/-) mice, including spermatogenic failures and the impaired blood-testis barrier, recapitulated the defects found in the Sertoli cell-specific androgen receptor (AR) knockout mice and the Sertoli cell-specific RB knockout mice. Investigation of the molecular mechanism identified several AR- and RB-responsive genes as downstream targets of ARID4A and ARID4B. Our results thus indicate that ARID4A and ARID4B function as transcriptional coactivators for AR and RB and play an integral part in the AR and RB regulatory pathways involved in the regulation of Sertoli cell function and male fertility.


Subject(s)
DNA-Binding Proteins/metabolism , Fertility/physiology , Receptors, Androgen/metabolism , Retinoblastoma Protein/metabolism , Retinoblastoma-Binding Protein 1/metabolism , Sertoli Cells/metabolism , Spermatogenesis/physiology , Animals , Blood-Testis Barrier/physiology , DNA-Binding Proteins/genetics , Female , Hypogonadism/genetics , Hypogonadism/metabolism , Hypogonadism/pathology , Infertility, Male/genetics , Infertility, Male/metabolism , Infertility, Male/pathology , Male , Meiosis/physiology , Mice , Mice, Knockout , Receptors, Androgen/genetics , Retinoblastoma Protein/genetics , Retinoblastoma-Binding Protein 1/genetics , Spermatids/metabolism
6.
Int J Mol Sci ; 17(7)2016 Jul 08.
Article in English | MEDLINE | ID: mdl-27399693

ABSTRACT

Oral squamous cell carcinoma (OSCC) is the most common malignancy of head and neck. Although radiotherapy is used for OSCC treatment, the occurrence of radioresistant cancer cells limits its efficiency. MicroRNAs (miRNAs) are non-coding RNAs with lengths of 18-25 base pairs and known to be involved in carcinogenesis. We previously demonstrated that by targeting B lymphoma Mo-MLV insertion region 1 homolog (Bmi1), miR-494-3p functions as a putative tumor suppressor miRNA in OSCC. In this study, we further discovered that miR-494-3p could enhance the radiosensitivity of SAS OSCC cells and induce cellular senescence. The overexpression of miR-494-3p in SAS cells increased the population of senescence-associated ß-galactosidase positive cells, the expression of p16(INK4a) and retinoblastoma 1 (RB1), as well as downregulated Bmi1. The knockdown of Bmi1 by lentiviral-mediated delivery of specific short hairpin RNAs (shRNAs) also enhanced the radiosensitivity of SAS cells and the activation of the senescence pathway. Furthermore, the inverse correlation between Bmi1 and miR-494-3p expression was observed among OSCC tissues. Results suggest that miR-494-3p could increase the radiosensitivity of OSCC cells through the induction of cellular senescence caused by the downregulation of Bmi1.


Subject(s)
Carcinoma, Squamous Cell/pathology , Cellular Senescence , MicroRNAs/metabolism , Mouth Neoplasms/pathology , 3' Untranslated Regions , Antagomirs/metabolism , Base Sequence , Carcinoma, Squamous Cell/metabolism , Cell Line, Tumor , Cell Survival/radiation effects , Cyclin-Dependent Kinase Inhibitor p16/metabolism , Down-Regulation , Gamma Rays , Humans , MicroRNAs/antagonists & inhibitors , MicroRNAs/genetics , Mouth Neoplasms/metabolism , Polycomb Repressive Complex 1/antagonists & inhibitors , Polycomb Repressive Complex 1/genetics , Polycomb Repressive Complex 1/metabolism , RNA Interference , RNA, Small Interfering/metabolism , Radiation Tolerance , Retinoblastoma-Binding Protein 1/metabolism , Sequence Alignment , beta-Galactosidase/metabolism
7.
J Biol Chem ; 289(8): 4882-95, 2014 Feb 21.
Article in English | MEDLINE | ID: mdl-24379399

ABSTRACT

Retinoblastoma-binding protein 1 (RBBP1) is a tumor and leukemia suppressor that binds both methylated histone tails and DNA. Our previous studies indicated that RBBP1 possesses a Tudor domain, which cannot bind histone marks. In order to clarify the function of the Tudor domain, the solution structure of the RBBP1 Tudor domain was determined by NMR and is presented here. Although the proteins are unrelated, the RBBP1 Tudor domain forms an interdigitated double Tudor structure similar to the Tudor domain of JMJD2A, which is an epigenetic mark reader. This indicates the functional diversity of Tudor domains. The RBBP1 Tudor domain structure has a significant area of positively charged surface, which reveals a capability of the RBBP1 Tudor domain to bind nucleic acids. NMR titration and isothermal titration calorimetry experiments indicate that the RBBP1 Tudor domain binds both double- and single-stranded DNA with an affinity of 10-100 µM; no apparent DNA sequence specificity was detected. The DNA binding mode and key interaction residues were analyzed in detail based on a model structure of the Tudor domain-dsDNA complex, built by HADDOCK docking using the NMR data. Electrostatic interactions mediate the binding of the Tudor domain with DNA, which is consistent with NMR experiments performed at high salt concentration. The DNA-binding residues are conserved in Tudor domains of the RBBP1 protein family, resulting in conservation of the DNA-binding function in the RBBP1 Tudor domains. Our results provide further insights into the structure and function of RBBP1.


Subject(s)
DNA/metabolism , Retinoblastoma-Binding Protein 1/chemistry , Retinoblastoma-Binding Protein 1/metabolism , Amino Acid Sequence , Base Sequence , Binding Sites , Calorimetry , Humans , Jumonji Domain-Containing Histone Demethylases/chemistry , Kinetics , Magnetic Resonance Spectroscopy , Models, Molecular , Molecular Sequence Data , Protein Binding , Protein Structure, Secondary , Protein Structure, Tertiary , Sequence Alignment , Sequence Homology, Amino Acid , Solutions , Titrimetry
8.
Int J Cancer ; 135(3): 611-23, 2014 Aug 01.
Article in English | MEDLINE | ID: mdl-24382590

ABSTRACT

ARID1A has been identified as a novel tumor suppressor gene in ovarian cancer and subsequently in various other tumor types. ARID1A belongs to the ARID domain containing gene family, which comprises of 15 genes involved, for example, in transcriptional regulation, proliferation and chromatin remodeling. In this study, we used exome sequencing data to analyze the mutation frequency of all the ARID domain containing genes in 25 microsatellite unstable (MSI) colorectal cancers (CRCs) as a first systematic effort to characterize the mutation pattern of the whole ARID gene family. Genes which fulfilled the selection criteria in this discovery set (mutations in at least 4/25 [16%] samples, including at least one nonsense or splice site mutation) were chosen for further analysis in an independent validation set of 21 MSI CRCs. We found that in addition to ARID1A, which was mutated in 39% of the tumors (18/46), also ARID1B (13%, 6/46), ARID2 (13%, 6/46) and ARID4A (20%, 9/46) were frequently mutated. In all these genes, the mutations were distributed along the entire length of the gene, thus distinguishing them from typical MSI target genes previously described. Our results indicate that in addition to ARID1A, other members of the ARID gene family may play a role in MSI CRC.


Subject(s)
Colorectal Neoplasms/genetics , DNA-Binding Proteins/genetics , Exome/genetics , Microsatellite Repeats/genetics , Mutation/genetics , Nuclear Proteins/genetics , Retinoblastoma-Binding Protein 1/genetics , Transcription Factors/genetics , Adenocarcinoma/genetics , Adenocarcinoma/pathology , Adult , Aged , Aged, 80 and over , Cohort Studies , Colorectal Neoplasms/pathology , Female , Follow-Up Studies , Humans , Male , Microsatellite Instability , Middle Aged , Neoplasm Staging , Prognosis
9.
J Biol Chem ; 287(11): 8531-40, 2012 Mar 09.
Article in English | MEDLINE | ID: mdl-22247551

ABSTRACT

Retinoblastoma-binding protein 1 (RBBP1), also named AT-rich interaction domain containing 4A (ARID4A), is a tumor and leukemia suppressor involved in epigenetic regulation in leukemia and Prader-Willi/Angelman syndromes. Although the involvement in epigenetic regulation is proposed to involve its chromobarrel and/or Tudor domains because of their potential binding to methylated histone tails, the structures of these domains and their interactions with methylated histone tails are still uncharacterized. In this work, we first found that RBBP1 contains five domains by bioinformatics analysis. Three of the five domains, i.e. chromobarrel, Tudor, and PWWP domains, are Royal Family domains, which potentially bind to methylated histone tails. We further purified these domains and characterized their interaction with methylated histone tails by NMR titration experiments. Among the three Royal Family domains, only the chromobarrel domain could recognize trimethylated H4K20 (with an affinity of ∼3 mm), as well as recognizing trimethylated H3K9, H3K27, and H3K36 (with lower affinities). The affinity could be further enhanced up to 15-fold by the presence of DNA. The structure of the chromobarrel domain of RBBP1 determined by NMR spectroscopy has an aromatic cage. Mutagenesis analysis identified four aromatic residues of the cage as the key residues for methylated lysine recognition. Our studies indicate that the chromobarrel domain of RBBP1 is responsible for recognizing methylated histone tails in chromatin remodeling and epigenetic regulation, which presents a significant advance in our understanding of the mechanism and relationship between RBBP1-related gene suppression and epigenetic regulation.


Subject(s)
Histones/chemistry , Retinoblastoma-Binding Protein 1/chemistry , Angelman Syndrome/genetics , Angelman Syndrome/metabolism , Chromatin Assembly and Disassembly/physiology , Epigenesis, Genetic/physiology , Histones/genetics , Histones/metabolism , Humans , Methylation , Mutagenesis , Nuclear Magnetic Resonance, Biomolecular , Prader-Willi Syndrome/genetics , Prader-Willi Syndrome/metabolism , Protein Structure, Tertiary , Retinoblastoma-Binding Protein 1/genetics , Retinoblastoma-Binding Protein 1/metabolism , Structure-Activity Relationship
10.
Nat Genet ; 18(4): 360-4, 1998 Apr.
Article in English | MEDLINE | ID: mdl-9537419

ABSTRACT

Mutation of the retinoblastoma tumour-suppressor gene (RB) leads to the deregulation of many proteins and transcription factors that interact with the retinoblastoma gene product (pRB), including members of the E2F transcription factor family. As pRB is known to repress E2F transcriptional activity and overexpression of E2F is sufficient for cell cycle progression, it is thought that pRB suppresses growth in part by repressing E2F-mediated transcription. Previously, we reported that loss of E2f1 in mice results in tissue-specific tumour induction and tissue atrophy, demonstrating that E2F-1 normally controls growth both positively and negatively in a tissue-specific fashion. To determine whether E2F-1 deregulation--as a result of loss of pRB--promotes proliferation in vivo, we have tested whether loss of E2f1 interferes with the pituitary and thyroid tumorigenesis that occurs in Rb1(+/-) mice. We have found that loss of E2f1 reduces the frequency of pituitary and thyroid tumours, and greatly lengthens the lifespan of Rb1(+/-); E2f1(-/-) animals, demonstrating that E2F-1 is an important downstream target of pRB during tumorigenesis. Furthermore, loss of E2f1 reduces a previously reported strain-dependent difference in Rb1(+/-) lifespan, suggesting that E2f1 or an E2F-1-regulated gene acts as a genetic modifier between the 129/Sv and C57BL/6 strains.


Subject(s)
Carrier Proteins , Cell Cycle Proteins , Cell Transformation, Neoplastic/genetics , DNA-Binding Proteins , Longevity/physiology , Retinoblastoma Protein/genetics , Transcription Factors/genetics , Animals , E2F Transcription Factors , E2F1 Transcription Factor , Female , Longevity/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Inbred Strains , Mice, Mutant Strains , Mutation/genetics , Mutation/physiology , Pituitary Neoplasms/genetics , Pituitary Neoplasms/physiopathology , Retinoblastoma Protein/physiology , Retinoblastoma-Binding Protein 1 , Species Specificity , Thyroid Neoplasms/genetics , Thyroid Neoplasms/physiopathology , Transcription Factor DP1 , Transcription Factors/physiology
11.
Nat Genet ; 25(3): 338-42, 2000 Jul.
Article in English | MEDLINE | ID: mdl-10888886

ABSTRACT

Methylation of CpG islands is associated with transcriptional silencing and the formation of nuclease-resistant chromatin structures enriched in hypoacetylated histones. Methyl-CpG-binding proteins, such as MeCP2, provide a link between methylated DNA and hypoacetylated histones by recruiting histone deacetylase, but the mechanisms establishing the methylation patterns themselves are unknown. Whether DNA methylation is always causal for the assembly of repressive chromatin or whether features of transcriptionally silent chromatin might target methyltransferase remains unresolved. Mammalian DNA methyltransferases show little sequence specificity in vitro, yet methylation can be targeted in vivo within chromosomes to repetitive elements, centromeres and imprinted loci. This targeting is frequently disrupted in tumour cells, resulting in the improper silencing of tumour-suppressor genes associated with CpG islands. Here we show that the predominant mammalian DNA methyltransferase, DNMT1, co-purifies with the retinoblastoma (Rb) tumour suppressor gene product, E2F1, and HDAC1 and that DNMT1 cooperates with Rb to repress transcription from promoters containing E2F-binding sites. These results establish a link between DNA methylation, histone deacetylase and sequence-specific DNA binding activity, as well as a growth-regulatory pathway that is disrupted in nearly all cancer cells.


Subject(s)
Carrier Proteins , Cell Cycle Proteins , DNA (Cytosine-5-)-Methyltransferases/metabolism , DNA-Binding Proteins , Histone Deacetylases/metabolism , Promoter Regions, Genetic , Retinoblastoma Protein/metabolism , Transcription Factors/metabolism , Transcription, Genetic , 3T3 Cells , Animals , Cyclin-Dependent Kinase Inhibitor p16/genetics , DNA (Cytosine-5-)-Methyltransferase 1 , DNA (Cytosine-5-)-Methyltransferases/genetics , E2F Transcription Factors , E2F1 Transcription Factor , HeLa Cells , Histone Deacetylase 1 , Histone Deacetylases/genetics , Humans , Mice , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Response Elements , Retinoblastoma Protein/genetics , Retinoblastoma-Binding Protein 1 , Transcription Factor DP1 , Transcription Factors/genetics
12.
Nat Genet ; 19(4): 371-4, 1998 Aug.
Article in English | MEDLINE | ID: mdl-9697699

ABSTRACT

The retinoblastoma (RB) gene is one of the most extensively studied tumour-suppressor genes. Deletion or inactivation of both RB alleles is an essential, rate-limiting step in the formation of retinoblastoma and osteosarcoma that arise in families that carry mutant RB (ref. 2). RB inactivation is also found in other human tumours. Whereas loss of RB function is associated with the loss of cellular proliferative control, introduction of a wild-type RB can suppress cell growth and tumorigenicity. Thus, identification of factors that interfere with and/or control the function of the RB protein is critical for understanding both cell-cycle control and oncogenesis. Here we describe a new gene, Bog (for B5T over-expressed gene), which was identified and shown to be overexpressed in several transformed rat liver epithelial (RLE) cell lines resistant to the growth-inhibitory effect of TGF-beta1, as well as in primary human liver tumours. The Bog protein shares homology with other retinoblastoma-binding proteins and contains the Rb-binding motif LXCXE. Using the yeast two-hybrid system and co-immunoprecipitation, we demonstrated that Bog binds to Rb. In vivo, Bog/Rb complexes do not contain E2F-1, and Bog can displace E2F-1 from E2F-1/Rb complexes in vitro. Overexpression of Bog in normal RLE cells conferred resistance to the growth-inhibitory effect of TGF-beta1. Furthermore, normal RLE cells are rapidly transformed when Bog is continuously overexpressed and form hepatoblastoma-like tumours when transplanted into nude mice. These data suggest that Bog may be important in the transformation process, in part due to its capacity to confer resistance to the growth-inhibitory effects of TGF-beta1 through interaction with Rb and the subsequent displacement of E2F-1.


Subject(s)
Carrier Proteins/metabolism , Cell Cycle Proteins , Cell Cycle/physiology , Cell Transformation, Neoplastic , DNA-Binding Proteins/metabolism , Intracellular Signaling Peptides and Proteins , Neoplasm Proteins , Retinoblastoma Protein/metabolism , Xenopus Proteins , Amino Acid Sequence , Animals , Carcinoma, Hepatocellular/chemistry , Carcinoma, Hepatocellular/pathology , Carrier Proteins/genetics , Cell Division , Cell Line , Cloning, Molecular , DNA-Binding Proteins/genetics , DNA-Binding Proteins/isolation & purification , E2F Transcription Factors , E2F1 Transcription Factor , Epithelial Cells , Humans , Hydrolases , Liver/cytology , Liver/metabolism , Mice , Mice, Nude , Molecular Sequence Data , Organ Specificity , RNA, Messenger/analysis , RNA, Neoplasm/analysis , Rats , Retinoblastoma-Binding Protein 1 , Sequence Homology, Amino Acid , Serine Proteases , Transcription Factor DP1 , Transcription Factors/metabolism , Transforming Growth Factor beta/pharmacology , Tumor Cells, Cultured , Werner Syndrome Helicase
13.
Nat Genet ; 26(4): 464-9, 2000 Dec.
Article in English | MEDLINE | ID: mdl-11101847

ABSTRACT

Most human cancers harbour aberrations of cell-cycle control, which result in deregulated activity of the E2F transcription factors with concomitant enhanced cell-cycle progression. Oncogenic signalling by E2F1 has recently been linked to stabilization and activation of the tumour suppressor p53 (refs 1,3,4). The p73 protein shares substantial sequence homology and functional similarity with p53 (refs 5-7 ). Hence, several previously considered p53-independent cellular activities may be attributable to p73. Here we provide evidence that E2F1 directly activates transcription of TP73, leading to activation of p53-responsive target genes and apoptosis. Disruption of p73 function by a tumour-derived p53 mutant reduced E2F1-mediated apoptosis. Thus, p73 activation by deregulated E2F1 activity might constitute a p53-independent, anti-tumorigenic safeguard mechanism.


Subject(s)
Apoptosis/drug effects , Apoptosis/physiology , Carrier Proteins , Cell Cycle Proteins , DNA-Binding Proteins/genetics , DNA-Binding Proteins/physiology , Nuclear Proteins/genetics , Nuclear Proteins/physiology , Transcription Factors/pharmacology , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/physiology , Animals , Apoptosis/genetics , Base Sequence , Binding Sites/genetics , COS Cells , Cell Line , DNA Primers/genetics , E2F Transcription Factors , E2F1 Transcription Factor , Gene Expression Regulation/drug effects , Genes, Tumor Suppressor , Humans , Mice , Molecular Sequence Data , Mutation , Promoter Regions, Genetic , Retinoblastoma-Binding Protein 1 , Transcription Factor DP1 , Transcription Factors/metabolism , Transcriptional Activation , Transfection , Tumor Protein p73 , Tumor Suppressor Proteins , U937 Cells
14.
Nat Genet ; 26(3): 291-9, 2000 Nov.
Article in English | MEDLINE | ID: mdl-11062467

ABSTRACT

To identify new immortalizing genes with potential roles in tumorigenesis, we performed a genetic screen aimed to bypass the rapid and tight senescence arrest of primary fibroblasts deficient for the oncogene Bmi1. We identified the T-box member TBX2 as a potent immortalizing gene that acts by downregulating Cdkn2a (p19(ARF)). TBX2 represses the Cdkn2a (p19(ARF)) promoter and attenuates E2F1, Myc or HRAS-mediated induction of Cdkn2a (p19(ARF)). We found TBX2 to be amplified in a subset of primary human breast cancers, indicating that it might contribute to breast cancer development.


Subject(s)
Adenocarcinoma/genetics , Breast Neoplasms/genetics , Cell Cycle Proteins/physiology , Cellular Senescence/genetics , Chromosomes, Human, Pair 17/genetics , DNA-Binding Proteins , Gene Amplification , Gene Expression Regulation, Neoplastic , Neoplasm Proteins/physiology , Protein Biosynthesis , Repressor Proteins/physiology , T-Box Domain Proteins/physiology , Adenocarcinoma/metabolism , Animals , Breast Neoplasms/metabolism , COS Cells , Carrier Proteins/genetics , Cell Cycle Proteins/genetics , Cell Cycle Proteins/isolation & purification , Cell Transformation, Neoplastic/genetics , Cells, Cultured , Cyclin-Dependent Kinase Inhibitor p16 , E2F Transcription Factors , E2F1 Transcription Factor , Female , Fibroblasts/cytology , Fibroblasts/metabolism , Gene Deletion , Genes, BRCA1 , Humans , Mice , Neoplasm Proteins/biosynthesis , Neoplasm Proteins/genetics , Neoplasm Proteins/isolation & purification , Neoplastic Syndromes, Hereditary/genetics , Nuclear Proteins/genetics , Oncogenes , Polycomb Repressive Complex 1 , Promoter Regions, Genetic , Proteins/genetics , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins c-myc/antagonists & inhibitors , Proto-Oncogene Proteins p21(ras)/antagonists & inhibitors , Repressor Proteins/genetics , Repressor Proteins/isolation & purification , Retinoblastoma-Binding Protein 1 , T-Box Domain Proteins/genetics , T-Box Domain Proteins/isolation & purification , Transcription Factor DP1 , Transcription Factors/antagonists & inhibitors , Transfection , Tumor Cells, Cultured , Tumor Suppressor Protein p14ARF
15.
Curr Opin Cell Biol ; 6(3): 443-50, 1994 Jun.
Article in English | MEDLINE | ID: mdl-7917337

ABSTRACT

In mammalian cells, DRTF1/E2F is a transcription factor widely believed to integrate cell-cycle progression with the transcription apparatus through its cyclical interactions with important regulators of the cell cycle, such as the retinoblastoma tumour-suppressor gene product, cyclins and cyclin-dependent kinases. Recently, a number of exciting developments have uncovered the heterodimeric nature of DRTF1/E2F by defining two distinct families of proteins, DP and E2F, which comprise its activity; efficient DNA-binding activity arises when a DP protein interacts with an E2F protein. Combinatorial interactions generate a surprising array of sequence-specific heterodimers, a diversity that is probably necessary to enable different cell cycle regulating proteins to integrate their activities with transcription.


Subject(s)
Carrier Proteins , Cell Cycle Proteins , Cell Cycle/physiology , DNA-Binding Proteins , Transcription Factors/physiology , Transcription, Genetic/physiology , Amino Acid Sequence , Animals , E2F Transcription Factors , Humans , Molecular Sequence Data , Retinoblastoma-Binding Protein 1 , Transcription Factor DP1 , Transcription Factors/analysis , Transcription Factors/genetics
16.
Curr Opin Cell Biol ; 6(6): 859-66, 1994 Dec.
Article in English | MEDLINE | ID: mdl-7880534

ABSTRACT

Transcriptional control during the G1/S transition is important in regulating cell cycle progression. The transcription factor DRTF1/E2F is believed to play a crucial role in this process by integrating the activity of the machinery that drives the cell cycle with the transcription apparatus. Being the point of convergence for growth-promoting and growth-inhibitory signals, it is a pivotal cellular target for molecules which subvert normal cell cycle control, such as oncoviral proteins. Recent studies have indicated that members of two distinct families of proteins, DP and E2F, interact combinatorially as DP/E2F heterodimers in DRTF1/E2F. The activities of both DP and E2F proteins are under cell cycle control, being influenced by the level of phosphorylation imparted through the cell cycle regulated activity of cyclin-dependent kinases. Both DP and E2F proteins are endowed with proto-oncogenic activity and, conversely, have been implicated in regulating apoptosis. Current evidence suggests therefore that the activity of DRTF1/E2F is instrumental in regulating progression through the cell cycle.


Subject(s)
Carrier Proteins , Cell Cycle Proteins , Cell Cycle/physiology , DNA-Binding Proteins , Transcription Factors/physiology , Transcription, Genetic/physiology , Animals , Apoptosis , Cell Cycle/genetics , Cell Division/physiology , Cell Survival , E2F Transcription Factors , Homeostasis/physiology , Humans , Neoplasms/physiopathology , Retinoblastoma-Binding Protein 1 , Transcription Factor DP1
17.
Curr Opin Cell Biol ; 11(6): 745-52, 1999 Dec.
Article in English | MEDLINE | ID: mdl-10600713

ABSTRACT

Cell proliferation and cell death are essential yet opposing cellular processes. Crosstalk between these processes promotes a balance between proliferation and death, and it limits the growth and survival of cells with oncogenic mutations. New insights into the mechanisms by which strong signals to proliferate and activation of cyclin-dependent kinases promote apoptosis have recently been published, and a novel cell cycle regulated caspase inhibitor, Survivin, has been described.


Subject(s)
Apoptosis , Carrier Proteins , Cell Cycle Proteins , Cell Division , DNA-Binding Proteins , Microtubule-Associated Proteins , Animals , Cell Cycle/physiology , Cyclin-Dependent Kinases/antagonists & inhibitors , Cyclin-Dependent Kinases/physiology , E2F Transcription Factors , Growth Substances/physiology , Inhibitor of Apoptosis Proteins , Neoplasm Proteins , Oncogenes/physiology , Proteins/metabolism , Proteins/physiology , Retinoblastoma Protein/physiology , Retinoblastoma-Binding Protein 1 , Signal Transduction , Survivin , Transcription Factors/physiology
18.
Nat Cell Biol ; 2(4): 232-9, 2000 Apr.
Article in English | MEDLINE | ID: mdl-10783242

ABSTRACT

The E2F proteins form a family of transcription factors that regulate the transition from the G1 to the S phase in the cell cycle. E2F activity is regulated by members of the retinoblastoma protein (pRb) family, ensuring the tight control of E2F-responsive genes. During the G1 phase, phosphorylation of pRb by cyclin-dependent kinases (CDKs), most notably cyclin D-CDK complexes, releases pRb from E2F, facilitating cell-cycle progression by the timely induction of E2F-targeted genes such as cyclin E. However, it is not known whether E2F proteins are directly targeted by CDKs. Here we show that E2F-5 is phosphorylated by the cyclin E-Cdk2 complex, which functions in the late G1 phase, but not by the early-G1-phase-acting cyclin D-CDK complex. A phosphorylation site in the trans-activation domain of E2F-5 stimulates transcription and cell-cycle progression by the recruitment of the p300/CBP family of co-activators, whose binding to E2F-5 is stabilized upon phosphorylation by cyclin E-Cdk2. These results indicate that E2F activity may be directly regulated by cyclin E-Cdk2, and imply an autoregulatory mechanism for cell-cycle-dependent transcription through the CDK-stimulated interaction of E2F with p300/CBP co-activators.


Subject(s)
CDC2-CDC28 Kinases , Carrier Proteins , Cell Cycle Proteins , Cyclin E/metabolism , Cyclin-Dependent Kinases/metabolism , DNA-Binding Proteins , Nuclear Proteins/metabolism , Protein Serine-Threonine Kinases/metabolism , Trans-Activators/metabolism , Transcription Factors/genetics , Transcriptional Activation/physiology , Animals , CREB-Binding Protein , Cell Cycle/genetics , Cells, Cultured , Cyclin-Dependent Kinase 2 , E2F Transcription Factors , E2F5 Transcription Factor , G1 Phase/genetics , Gene Expression Regulation, Enzymologic/physiology , Homeostasis/genetics , Phosphorylation , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Retinoblastoma-Binding Protein 1 , S Phase/genetics , Threonine
19.
Nat Cell Biol ; 1(1): 14-9, 1999 May.
Article in English | MEDLINE | ID: mdl-10559858

ABSTRACT

The transcription factor E2F-1 is important in the control of cell proliferation. Its activity must be tightly regulated in a cell-cycle-dependent manner to enable programs of gene expression to be coupled closely with cell-cycle position. Here we show that, following its accumulation in the late G1 phase of the cell cycle, E2F-1 is rapidly degraded in S/G2 phase. This event is linked to a specific interaction of E2F-1 with the F-box-containing protein p45SKP2, which is the cell-cycle-regulated component of the ubiquitin-protein ligase SCFSKP2 that recognizes substrates for this ligase. Disruption of the interaction between E2F-1 and p45SKP2 results in a reduction in ubiquitination of E2F-1 and the stabilization and accumulation of transcriptionally active E2F-1 protein. These results indicate that an SCFSKP2-dependent ubiquitination pathway may be involved in the downregulation of E2F-1 activity in the S/G2 phase of the cell cycle, and suggest a link between SCFSKP2 and cell-cycle-dependent gene control.


Subject(s)
Carrier Proteins , Cell Cycle Proteins/metabolism , Cell Cycle/physiology , DNA-Binding Proteins , Ligases/metabolism , Transcription Factors/metabolism , 3T3 Cells , Animals , Cell Line , E2F Transcription Factors , E2F1 Transcription Factor , G1 Phase , G2 Phase , HeLa Cells , Homeostasis , Humans , Kinetics , Mice , Retinoblastoma-Binding Protein 1 , S Phase , S-Phase Kinase-Associated Proteins , Substrate Specificity , Transcription Factor DP1 , Ubiquitin-Protein Ligases , Ubiquitins/metabolism
20.
Nat Cell Biol ; 1(2): 88-93, 1999 Jun.
Article in English | MEDLINE | ID: mdl-10559879

ABSTRACT

Centrosome duplication is a key requirement for bipolar spindle formation and correct segregation of chromosomes during cell division. In a manner highly reminiscent of DNA replication, the centrosome must be duplicated once, and only once, in each cell cycle. How centrosome duplication is regulated and coordinated with other cell-cycle functions remains poorly understood. Here, we have established a centrosome duplication assay using mammalian somatic cells. We show that centrosome duplication requires the activation of E2F transcription factors and Cdk2-cyclin A activity.


Subject(s)
CDC2-CDC28 Kinases , Carrier Proteins , Cell Cycle/physiology , Centrosome/physiology , Centrosome/ultrastructure , Cyclin A/metabolism , Cyclin-Dependent Kinases/metabolism , DNA Replication , DNA-Binding Proteins , Protein Serine-Threonine Kinases/metabolism , Transcription Factors/metabolism , Animals , CHO Cells , Cell Cycle Proteins/metabolism , Cricetinae , Cyclin-Dependent Kinase 2 , E2F Transcription Factors , Mammals , Recombinant Proteins/metabolism , Retinoblastoma-Binding Protein 1 , Transfection
SELECTION OF CITATIONS
SEARCH DETAIL