Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 100
Filter
1.
Sci Rep ; 10(1): 14156, 2020 08 25.
Article in English | MEDLINE | ID: mdl-32843651

ABSTRACT

Recent data suggest that the transcription factor Zfp148 represses activation of the tumor suppressor p53 in mice and that therapeutic targeting of the human orthologue ZNF148 could activate the p53 pathway without causing detrimental side effects. We have previously shown that Zfp148 deficiency promotes p53-dependent proliferation arrest of mouse embryonic fibroblasts (MEFs), but the underlying mechanism is not clear. Here, we showed that Zfp148 deficiency downregulated cell cycle genes in MEFs in a p53-dependent manner. Proliferation arrest of Zfp148-deficient cells required increased expression of ARF, a potent activator of the p53 pathway. Chromatin immunoprecipitation showed that Zfp148 bound to the ARF promoter, suggesting that Zfp148 represses ARF transcription. However, Zfp148 preferentially bound to promoters of other transcription factors, indicating that deletion of Zfp148 may have pleiotropic effects that activate ARF and p53 indirectly. In line with this, we found no evidence of genetic interaction between TP53 and ZNF148 in CRISPR and siRNA screen data from hundreds of human cancer cell lines. We conclude that Zfp148 deficiency, by increasing ARF transcription, downregulates cell cycle genes and cell proliferation in a p53-dependent manner. However, the lack of genetic interaction between ZNF148 and TP53 in human cancer cells suggests that therapeutic targeting of ZNF148 may not increase p53 activity in humans.


Subject(s)
DNA-Binding Proteins/genetics , Gene Expression Regulation/genetics , Signal Transduction/genetics , Transcription Factors/genetics , Tumor Suppressor Protein p53/physiology , Animals , CRISPR-Cas Systems , Cell Cycle Checkpoints/genetics , Cell Cycle Proteins/biosynthesis , Cell Cycle Proteins/genetics , Cell Division , Cell Line , Chromatin Immunoprecipitation , Cisplatin/toxicity , Cyclin-Dependent Kinase Inhibitor p16/metabolism , DNA Damage , DNA-Binding Proteins/deficiency , DNA-Binding Proteins/physiology , Down-Regulation , E2F Transcription Factors/physiology , Etoposide/toxicity , Fibroblasts , Gene Ontology , Mice , RNA Interference , RNA, Small Interfering/genetics , Transcription Factors/deficiency , Transcription Factors/physiology
2.
Dev Cell ; 52(3): 335-349.e7, 2020 02 10.
Article in English | MEDLINE | ID: mdl-31983631

ABSTRACT

E2F transcription factors control the cytokinesis machinery and thereby ploidy in hepatocytes. If or how these proteins limit proliferation of polyploid cells with extra centrosomes remains unknown. Here, we show that the PIDDosome, a signaling platform essential for caspase-2-activation, limits hepatocyte ploidy and is instructed by the E2F network to control p53 in the developing as well as regenerating liver. Casp2 and Pidd1 act as direct transcriptional targets of E2F1 and its antagonists, E2F7 and E2F8, that together co-regulate PIDDosome expression during juvenile liver growth and regeneration. Of note, whereas hepatocyte aneuploidy correlates with the basal ploidy state, the degree of aneuploidy itself is not limited by PIDDosome-dependent p53 activation. Finally, we provide evidence that the same signaling network is engaged to control ploidy in the human liver after resection. Our study defines the PIDDosome as a primary target to manipulate hepatocyte ploidy and proliferation rates in the regenerating liver.


Subject(s)
Caspase 2/physiology , Death Domain Receptor Signaling Adaptor Proteins/physiology , E2F Transcription Factors/physiology , Hepatocytes/cytology , Liver Regeneration , Polyploidy , Tumor Suppressor Protein p53/physiology , Aneuploidy , Animals , CRADD Signaling Adaptor Protein/physiology , Centrosome , Cyclin-Dependent Kinase Inhibitor p21/physiology , Cytokinesis , Female , Hepatocytes/metabolism , Humans , Male , Mice , Mice, Knockout
4.
Acta Biochim Biophys Sin (Shanghai) ; 51(3): 313-322, 2019 Mar 01.
Article in English | MEDLINE | ID: mdl-30726872

ABSTRACT

Activation of transcription factor STAT3 is involved in cell proliferation, differentiation, and cell survival. Constitutive activation of STAT3 pathway has been associated with the oncogenesis of various types of cancers. It has been reported that STAT3 plays a key role in the G1 to S phase cell cycle transition induced by the cytokine receptor subunit gp130, through the upregulation of cyclins D1, D2, D3, A, and Cdc25A and the concomitant downregulation of p21 and p27. However, its role in mediating G2-M phase transition has not been studied. The cyclin B1/Cdc2 complex is widely accepted as the trigger of mitosis in all organisms and is believed to be necessary for progression through S phase and keep active during the G2-M transition and progression. In the present study, we found that activation of STAT3 stimulates cyclin B1 and Cdc2 expressions. Deletion and site-directed mutations on cyclin B1 and Cdc2 promoters indicated that E2F element mediates the upregulation of these two promoters in a STAT3-dependent manner. The findings reported here demonstrated that STAT3 participates in modulating G2-M phase checkpoint by regulating gene expressions of cyclin B1 and Cdc2 via E2F.


Subject(s)
CDC2 Protein Kinase/genetics , Cyclin B1/genetics , E2F Transcription Factors/physiology , STAT3 Transcription Factor/physiology , Cell Division , Cells, Cultured , G2 Phase , Gene Expression Regulation , Humans , Promoter Regions, Genetic , Up-Regulation
5.
Circ Res ; 124(6): 856-873, 2019 03 15.
Article in English | MEDLINE | ID: mdl-30696354

ABSTRACT

RATIONALE: Mutations in the LMNA gene, encoding LMNA (lamin A/C), are responsible for laminopathies. Dilated cardiomyopathy (DCM) is a major cause of mortality and morbidity in laminopathies. OBJECTIVE: To gain insights into the molecular pathogenesis of DCM in laminopathies. METHODS AND RESULTS: We generated a tet-off bigenic mice expressing either a WT (wild type) or a mutant LMNA (D300N) protein in cardiac myocytes. LMNAD300N mutation is associated with DCM in progeroid syndromes. Expression of LMNAD300N led to severe myocardial fibrosis, apoptosis, cardiac dysfunction, and premature death. Administration of doxycycline suppressed LMNAD300N expression and prevented the phenotype. Whole-heart RNA sequencing in 2-week-old WT and LMNAD300N mice led to identification of ≈6000 differentially expressed genes. Gene Set Enrichment and Hallmark Pathway analyses predicted activation of E2F (E2F transcription factor), DNA damage response, TP53 (tumor protein 53), NFκB (nuclear factor κB), and TGFß (transforming growth factor-ß) pathways, which were validated by Western blotting, quantitative polymerase chain reaction of selected targets, and immunofluorescence staining. Differentially expressed genes involved cell death, cell cycle regulation, inflammation, and epithelial-mesenchymal differentiation. RNA sequencing of human hearts with DCM associated with defined LMNA pathogenic variants corroborated activation of the DNA damage response/TP53 pathway in the heart. Increased expression of CDKN2A (cyclin-dependent kinase inhibitor 2A)-a downstream target of E2F pathway and an activator of TP53-provided a plausible mechanism for activation of the TP53 pathway. To determine pathogenic role of TP53 pathway in DCM, Tp53 gene was conditionally deleted in cardiac myocytes in mice expressing the LMNAD300N protein. Deletion of Tp53 partially rescued myocardial fibrosis, apoptosis, proliferation of nonmyocyte cells, left ventricular dilatation and dysfunction, and slightly improved survival. CONCLUSIONS: Cardiac myocyte-specific expression of LMNAD300N, associated with DCM, led to pathogenic activation of the E2F/DNA damage response/TP53 pathway in the heart and induction of myocardial fibrosis, apoptosis, cardiac dysfunction, and premature death. The findings denote the E2F/DNA damage response/TP53 axis as a responsible mechanism for DCM in laminopathies and as a potential intervention target.


Subject(s)
Cardiomyopathy, Dilated/etiology , DNA Damage , Lamin Type A/genetics , Mutation , Tumor Suppressor Protein p53/physiology , Animals , Apoptosis , Cell Proliferation , E2F Transcription Factors/physiology , Female , Fibrosis , Male , Mice , Myocardium/pathology , Myocytes, Cardiac/metabolism , Signal Transduction
6.
Proc Natl Acad Sci U S A ; 115(16): E3837-E3845, 2018 04 17.
Article in English | MEDLINE | ID: mdl-29610335

ABSTRACT

DNA damage poses a serious threat to genome integrity and greatly affects growth and development. To maintain genome stability, all organisms have evolved elaborate DNA damage response mechanisms including activation of cell cycle checkpoints and DNA repair. Here, we show that the DNA repair protein SNI1, a subunit of the evolutionally conserved SMC5/6 complex, directly links these two processes in Arabidopsis SNI1 binds to the activation domains of E2F transcription factors, the key regulators of cell cycle progression, and represses their transcriptional activities. In turn, E2Fs activate the expression of SNI1, suggesting that E2Fs and SNI1 form a negative feedback loop. Genetically, overexpression of SNI1 suppresses the phenotypes of E2F-overexpressing plants, and loss of E2F function fully suppresses the sni1 mutant, indicating that SNI1 is necessary and sufficient to inhibit E2Fs. Altogether, our study revealed that SNI1 is a negative regulator of E2Fs and plays dual roles in DNA damage responses by linking cell cycle checkpoint and DNA repair.


Subject(s)
Arabidopsis Proteins/physiology , Arabidopsis/genetics , Cell Cycle Checkpoints/genetics , DNA Repair/genetics , E2F Transcription Factors/physiology , Gene Expression Regulation, Plant , Nuclear Proteins/physiology , Arabidopsis/cytology , Arabidopsis/metabolism , Arabidopsis Proteins/genetics , Arabidopsis Proteins/metabolism , DNA Damage , E2F Transcription Factors/chemistry , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Plant Roots/genetics , Plant Roots/growth & development , Plant Roots/metabolism , Protein Domains
7.
PLoS One ; 13(1): e0190899, 2018.
Article in English | MEDLINE | ID: mdl-29329320

ABSTRACT

The presence of polyploid cells in the endocrine and exocrine pancreas has been reported for four decades. In rodents, pancreatic polyploidization is initiated after weaning and the number of polyploid cells increases with age. Surprisingly the molecular regulators and biological functions of polyploidization in the pancreas are still unknown. We discovered that atypical E2f activity is essential for polyploidization in the pancreas, using an inducible Cre/LoxP approach in new-born mice to delete ubiquitously the atypical E2f transcription factors, E2f7 and E2f8. In contrast to its critical role in embryonic survival, conditional deletion of both of both atypical E2fs in newborn mice had no impact on postnatal survival and mice lived until old age. However, deficiency of E2f7 or E2f8 alone was sufficient to suppress polyploidization in the pancreas and associated with only a minor decrease in blood serum levels of glucose, insulin, amylase and lipase under 4 hours starvation condition compared to wildtype littermates. In mice with fewer pancreatic polyploid cells that were fed ad libitum, no major impact on hormones or enzymes levels was observed. In summary, we identified atypical E2fs to be essential for polyploidization in the pancreas and discovered that postnatal induced loss of both atypical E2fs in many organs is compatible with life until old age.


Subject(s)
E2F Transcription Factors/physiology , Pancreas/cytology , Polyploidy , Amylases/blood , Animals , Blood Glucose/metabolism , Growth , Insulin/blood , Lipase/blood , Mice , Survival Analysis
8.
Gastroenterology ; 154(6): 1625-1629.e8, 2018 05.
Article in English | MEDLINE | ID: mdl-29366840

ABSTRACT

Lamins have important roles in nuclear structure and cell signaling. Several diseases are associated with mutations in the lamin A/C gene (LMNA in humans). Patients with familial partial lipodystrophy caused by LMNA mutations develop pancreatitis, but lamin function in the pancreas and how these mutations affect pancreatic regulation are unknown. We generated mice with inducible exocrine pancreas-specific disruption of Lmna and showed that LMNA is lost from most exocrine pancreas cells. LMNA-knockout pancreata develop endoplasmic reticulum stress with loss of acinar cell markers, increased autophagy, apoptosis, and cell proliferation, compared to CreERT2- mice (littermate controls). Disruption of Lmna led to a phenotype that resembled chronic pancreatitis, with increased Sirius Red staining and α-smooth muscle actin in male LMNA-knockout mice compared to littermate males, but not in female mice. LMNA-knockout pancreata have reduced levels of RB and activation of E2F, based on increased expression of E2F target genes. Therefore, lamins maintain pancreatic homeostasis by regulating RB stability and E2F activity.


Subject(s)
E2F Transcription Factors/physiology , Homeostasis/genetics , Lamin Type A/physiology , Pancreas, Exocrine/metabolism , Retinoblastoma Protein/physiology , Animals , Female , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Signal Transduction/genetics
9.
PLoS One ; 12(1): e0170036, 2017.
Article in English | MEDLINE | ID: mdl-28076433

ABSTRACT

Platelet-derived growth factor (PDGF) plays an essential role in proliferation of vascular smooth muscle cells (VSMCs). The Rb/E2F and TSC/mTOR pathways contribute to the proliferation of VSMCs, but its exact roles in PDGF-induced proliferation are unclear. In this study, we demonstrated the roles of Rb/E2F and TSC/mTOR pathways in PDGF-induced proliferation in VSMCs. We found that PDGF stimulates the activity of E2F and mTOR pathways, and knockdown of either Rb or TSC2 increases PDGF-induced proliferation in VSMCs. More interestingly, we revealed that enhancing both E2F and mTOR activity leads to synergistic inhibition of PDGF-induced proliferation in VSMCs. We further identified that the synergistic inhibition effect is caused by the induced oxidative stress. Summarily, these data suggest the important regulations of Rb/E2F and TSC/mTOR pathways in PDGF-induced proliferation in VSMCs, and also present a promising way to limit deregulated proliferation by PDGF induction in VSMCs.


Subject(s)
Cell Proliferation/drug effects , E2F Transcription Factors/physiology , Muscle, Smooth, Vascular/drug effects , Myocytes, Smooth Muscle/drug effects , Platelet-Derived Growth Factor/pharmacology , Retinoblastoma Protein/physiology , Calcium-Binding Proteins/metabolism , Calcium-Binding Proteins/physiology , Cells, Cultured , E2F Transcription Factors/metabolism , Humans , Muscle, Smooth, Vascular/physiology , Myocytes, Smooth Muscle/physiology , Oxidative Stress/drug effects , Oxidative Stress/physiology , Retinoblastoma Protein/metabolism , Signal Transduction/drug effects , TOR Serine-Threonine Kinases/metabolism , TOR Serine-Threonine Kinases/physiology , Up-Regulation/drug effects
10.
Cancer Res ; 77(4): 982-995, 2017 02 15.
Article in English | MEDLINE | ID: mdl-27923835

ABSTRACT

RB loss occurs commonly in neoplasia but its contributions to advanced cancer have not been assessed directly. Here we show that RB loss in multiple murine models of cancer produces a prometastatic phenotype. Gene expression analyses showed that regulation of the cell motility receptor RHAMM by the RB/E2F pathway was critical for epithelial-mesenchymal transition, motility, and invasion by cancer cells. Genetic modulation or pharmacologic inhibition of RHAMM activity was sufficient and necessary for metastatic phenotypes induced by RB loss in prostate cancer. Mechanistic studies in this setting established that RHAMM stabilized F-actin polymerization by controlling ROCK signaling. Collectively, our findings show how RB loss drives metastatic capacity and highlight RHAMM as a candidate therapeutic target for treating advanced prostate cancer. Cancer Res; 77(4); 982-95. ©2016 AACR.


Subject(s)
Prostatic Neoplasms/pathology , Retinoblastoma Protein/physiology , Actins/metabolism , Animals , Cell Line, Tumor , Cyclin-Dependent Kinase 4/antagonists & inhibitors , Cyclin-Dependent Kinase 6/antagonists & inhibitors , E2F Transcription Factors/physiology , Extracellular Matrix Proteins/antagonists & inhibitors , Extracellular Matrix Proteins/genetics , Extracellular Matrix Proteins/physiology , Humans , Hyaluronan Receptors/genetics , Hyaluronan Receptors/physiology , Male , Mice , Neoplasm Metastasis , Signal Transduction/physiology , rho-Associated Kinases/physiology
11.
Biochem J ; 473(15): 2331-43, 2016 08 01.
Article in English | MEDLINE | ID: mdl-27274086

ABSTRACT

WDR77 (WD repeat domain 77) is expressed during earlier lung development when cells are rapidly proliferating, but is absent from adult lung. It is re-activated during lung tumorigenesis and is essential for lung cancer cell proliferation. Signalling pathways/molecules that control WDR77 gene expression are unknown. Promoter mapping, gel shift assay and ChIP revealed that the WDR77 promoter contains bona fide response elements for E2F and GATA transcriptional factors as demonstrated in prostate cancer, lung cancer and erythroid cells, as well as in mouse lung tissues. The WDR77 promoter is transactivated by E2F1, E2F3, GATA1 and GATA6, but suppressed by E2F6, GATA1 and GATA3 in prostate cancer PC3 cells. WDR77 expression is associated with E2F1, E2F3, GATA2 and GATA6 occupancy on the WDR77 gene, whereas, in contrast, E2F6, GATA1 and GATA3 occupancy is associated with the loss of WDR77 expression during erythroid maturation and lung development. More importantly, the loss of WDR77 expression that results from E2F and GATA switches is required for cellular differentiation of erythroid and lung epithelial cells. In contrast, lung cancer cells avoid post-mitotic differentiation by sustaining WDR77 expression. Altogether, the present study provides a novel molecular mechanism by which WDR77 is regulated during erythroid and lung development and lung tumorigenesis.


Subject(s)
Cell Differentiation , E2F Transcription Factors/physiology , GATA Transcription Factors/physiology , Gene Expression Regulation/physiology , Transcription Factors/genetics , Animals , Base Sequence , Cell Line, Tumor , Cell Proliferation/genetics , Humans , Mice , Mice, Transgenic , Promoter Regions, Genetic , Regulatory Sequences, Nucleic Acid
12.
Oncogene ; 35(50): 6429-6438, 2016 12 15.
Article in English | MEDLINE | ID: mdl-27270425

ABSTRACT

The retinoblastoma tumor suppressor protein (RB) plays a critical role in cell proliferation and differentiation and its inactivation is a frequent underlying factor in tumorigenesis. While the regulation of RB function by phosphorylation is well studied, proteasome-mediated RB protein degradation is emerging as an important regulatory mechanism. Although our understanding of RB turnover is currently limited, there is evidence that the nuclear lamina filament protein Lamin A/C protects RB from proteasomal degradation. Here we show that SUMO1 conjugation of RB and Lamin A/C is modulated by the SUMO protease SENP1 and that sumoylation of both proteins is required for their interaction. Importantly, this SUMO1-dependent complex protects both RB and Lamin A/C from proteasomal turnover.


Subject(s)
Endopeptidases/physiology , Lamin Type A/physiology , Retinoblastoma Protein/physiology , Sumoylation , Animals , Cells, Cultured , Cysteine Endopeptidases , E2F Transcription Factors/physiology , Mice , Proteasome Endopeptidase Complex/physiology , Protein Stability
13.
Dev Biol ; 411(2): 195-206, 2016 Mar 15.
Article in English | MEDLINE | ID: mdl-26859702

ABSTRACT

The E2F family of transcription factors are evolutionarily conserved regulators of the cell cycle that can be divided into two groups based on their ability to either activate or repress transcription. In Drosophila, there is only one "activator" E2F, dE2F1, which provides all of the pro-proliferative activity of E2F during development. Interestingly, the de2f1 gene can be transcribed from multiple promoters resulting in six alternate transcripts. In this study, we sought to investigate the biological significance of the alternate transcriptional start sites. We focused on the de2f1 promoter region where tissue and cell-type specific enhancer activities were observed at the larval stage. While a genomic deletion of this region, de2f1(ΔRA), decreased the overall expression level of dE2F1, flies developed normally with no obvious proliferation defects. However, a detailed analysis of the de2f1(ΔRA) mutant eye imaginal discs revealed that dE2F1 is needed for proper cell cycle exit. We discovered that dE2F1 expression during G1 arrest prior to the differentiation process of the developing eye is important for maintaining cell cycle arrest at a later stage of the eye development. Overall, our study suggests that specific alternate transcripts of "activator" E2F, dE2F1, may have a dual function on cell cycle progression and cannot simply be viewed as a pro-proliferative transcription factor.


Subject(s)
Drosophila Proteins/genetics , Drosophila Proteins/physiology , Drosophila melanogaster/genetics , E2F Transcription Factors/genetics , E2F Transcription Factors/physiology , Gene Expression Regulation, Developmental , Animals , Cell Cycle , Cell Differentiation , Cell Division , Crosses, Genetic , Eye/embryology , G1 Phase , In Situ Hybridization , Mutation , Promoter Regions, Genetic , Reverse Transcriptase Polymerase Chain Reaction , Transcription Factors , Transcription, Genetic
14.
J Biol Chem ; 290(47): 28070-28083, 2015 Nov 20.
Article in English | MEDLINE | ID: mdl-26378234

ABSTRACT

We previously identified two distinct molecular subtypes of osteosarcoma through gene expression profiling. These subtypes are associated with distinct tumor behavior and clinical outcomes. Here, we describe mechanisms that give rise to these molecular subtypes. Using bioinformatic analyses, we identified a significant association between deregulation of the retinoblastoma (RB)-E2F pathway and the molecular subtype with worse clinical outcomes. Xenotransplantation models recapitulated the corresponding behavior for each osteosarcoma subtype; thus, we used cell lines to validate the role of the RB-E2F pathway in regulating the prognostic gene signature. Ectopic RB resets the patterns of E2F regulated gene expression in cells derived from tumors with worse clinical outcomes (molecular phenotype 2) to those comparable with those observed in cells derived from tumors with less aggressive outcomes (molecular phenotype 1), providing a functional association between RB-E2F dysfunction and altered gene expression in osteosarcoma. DNA methyltransferase and histone deacetylase inhibitors similarly reset the transcriptional state of the molecular phenotype 2 cells from a state associated with RB deficiency to one seen with RB sufficiency. Our data indicate that deregulation of RB-E2F pathway alters the epigenetic landscape and biological behavior of osteosarcoma.


Subject(s)
E2F Transcription Factors/physiology , Gene Expression Regulation/physiology , Osteosarcoma/genetics , Retinoblastoma Protein/physiology , Transcription, Genetic/physiology , Animals , Cell Line, Tumor , Dogs , Humans , Jurkat Cells , Osteosarcoma/pathology , Prognosis
15.
Oncogene ; 34(11): 1432-41, 2015 Mar 12.
Article in English | MEDLINE | ID: mdl-24704824

ABSTRACT

Aberrant splicing of the cyclin-dependent kinase-associated phosphatase, KAP, promotes glioblastoma invasion in a Cdc2-dependent manner. However, the mechanism by which this occurs is unknown. Here we show that miR-26a, which is often amplified in glioblastoma, promotes invasion in phosphatase and tensin homolog (PTEN)-competent and PTEN-deficient glioblastoma cells by directly downregulating KAP expression. Mechanistically, we find that KAP binds and activates ROCK2. Thus, RNA-mediated downregulation of KAP leads to decreased ROCK2 activity and this, in turn, increases Rac1-mediated invasion. In addition, the decrease in KAP expression activates the cyclin-dependent kinase, Cdk2, and this directly promotes invasion by increasing retinoblastoma phosphorylation, E2F-dependent Cdc2 expression and Cdc2-mediated inactivation of the actomyosin inhibitor, caldesmon. Importantly, glioblastoma cell invasion mediated by this pathway can be antagonized by Cdk2/Cdc2 inhibitors in vitro and in vivo. Thus, two distinct RNA-based mechanisms activate this novel KAP/ROCK2/Cdk2-dependent invasion pathway in glioblastoma.


Subject(s)
Cyclin-Dependent Kinase 2/metabolism , Cyclin-Dependent Kinase Inhibitor Proteins/metabolism , Dual-Specificity Phosphatases/metabolism , Glioblastoma/pathology , MicroRNAs/physiology , rho-Associated Kinases/metabolism , Actomyosin/antagonists & inhibitors , Brain Neoplasms/pathology , CDC2 Protein Kinase , Calmodulin-Binding Proteins/antagonists & inhibitors , Cell Line, Tumor , Cyclin-Dependent Kinase 2/antagonists & inhibitors , Cyclin-Dependent Kinase 2/genetics , Cyclin-Dependent Kinase Inhibitor Proteins/biosynthesis , Cyclin-Dependent Kinase Inhibitor Proteins/genetics , Cyclin-Dependent Kinases/antagonists & inhibitors , Cyclin-Dependent Kinases/biosynthesis , Dual-Specificity Phosphatases/biosynthesis , Dual-Specificity Phosphatases/genetics , E2F Transcription Factors/physiology , Enzyme Activation , Humans , MicroRNAs/antagonists & inhibitors , MicroRNAs/genetics , Neoplasm Invasiveness , PTEN Phosphohydrolase/metabolism , Phosphorylation , Protein Binding , RNA Interference , RNA, Small Interfering , Retinoblastoma Protein/metabolism , rac1 GTP-Binding Protein/physiology
16.
Cancer Res ; 74(22): 6565-6577, 2014 Nov 15.
Article in English | MEDLINE | ID: mdl-25252918

ABSTRACT

Bladder cancer is a highly prevalent human disease in which retinoblastoma (Rb) pathway inactivation and epigenetic alterations are common events. However, the connection between these two processes is still poorly understood. Here, we show that the in vivo inactivation of all Rb family genes in the mouse urothelium is sufficient to initiate bladder cancer development. The characterization of the mouse tumors revealed multiple molecular features of human bladder cancer, including the activation of E2F transcription factor and subsequent Ezh2 expression and the activation of several signaling pathways previously identified as highly relevant in urothelial tumors. These mice represent a genetically defined model for human high-grade superficial bladder cancer. Whole transcriptional characterizations of mouse and human bladder tumors revealed a significant overlap and confirmed the predominant role for Ezh2 in the downregulation of gene expression programs. Importantly, the increased tumor recurrence and progression in human patients with superficial bladder cancer is associated with increased E2F and Ezh2 expression and Ezh2-mediated gene expression repression. Collectively, our studies provide a genetically defined model for human high-grade superficial bladder cancer and demonstrate the existence of an Rb-E2F-Ezh2 axis in bladder whose disruption can promote tumor development.


Subject(s)
E2F Transcription Factors/physiology , Polycomb Repressive Complex 2/physiology , Retinoblastoma Protein/physiology , Signal Transduction/physiology , Urinary Bladder Neoplasms/etiology , Animals , Disease Progression , Enhancer of Zeste Homolog 2 Protein , Gene Expression Regulation, Neoplastic , Humans , Mice , Neoplasm Recurrence, Local/etiology , Polycomb Repressive Complex 2/genetics , Transcriptome
17.
Adv Cancer Res ; 121: 147-182, 2014.
Article in English | MEDLINE | ID: mdl-24889531

ABSTRACT

The retinoblastoma tumor suppressor protein Rb plays a major role in regulating G1/S transition and is a critical regulator of cell proliferation. Rb protein exerts its growth regulatory properties mainly by physically interacting with the transcriptionally active members of the E2F transcription factor family, especially E2Fs 1, 2, and 3. Given its critical role in regulating cell proliferation, it is not surprising that Rb is inactivated in almost all tumors, either through the mutation of Rb gene itself or through the mutations of its upstream regulators including K-Ras and INK4. Recent studies have revealed a significant role for Rb and its downstream effectors, especially E2Fs, in regulating various aspects of tumor progression, angiogenesis, and metastasis. Thus, components of the Rb-E2F pathway have been shown to regulate the expression of genes involved in angiogenesis, including VEGF and VEGFR, genes involved in epithelial-mesenchymal transition including E-cadherin and ZEB proteins, and genes involved in invasion and migration like matrix metalloproteinases. Rb has also been shown to play a major role in the functioning of normal and cancer stem cells; further, Rb and E2F appear to play a regulatory role in the energy metabolism of cancer cells. These findings raise the possibility that mutational events that initiate tumorigenesis by inducing uncontrolled cell proliferation might also contribute to the progression and metastasis of cancers through the mediation of the Rb-E2F transcriptional regulatory pathway. This review highlights these recent studies on tumor promoting functions of the Rb-E2F pathway.


Subject(s)
E2F Transcription Factors/physiology , Gene Expression Regulation, Neoplastic , Neoplasms/blood supply , Neoplasms/pathology , Neovascularization, Pathologic/genetics , Retinoblastoma Protein/physiology , Animals , Humans , Neoplasm Metastasis/genetics , Neoplasms/genetics , Transcription, Genetic
18.
Mol Cell Biol ; 34(17): 3229-43, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24934442

ABSTRACT

While the E2F transcription factors (E2Fs) have a clearly defined role in cell cycle control, recent work has uncovered new functions. Using genomic signature methods, we predicted a role for the activator E2F transcription factors in the mouse mammary tumor virus (MMTV)-polyomavirus middle T oncoprotein (PyMT) mouse model of metastatic breast cancer. To genetically test the hypothesis that the E2Fs function to regulate tumor development and metastasis, we interbred MMTV-PyMT mice with E2F1, E2F2, or E2F3 knockout mice. With the ablation of individual E2Fs, we noted alterations of tumor latency, histology, and vasculature. Interestingly, we noted striking reductions in metastatic capacity and in the number of circulating tumor cells in both the E2F1 and E2F2 knockout backgrounds. Investigating E2F target genes that mediate metastasis, we found that E2F loss led to decreased levels of vascular endothelial growth factor (Vegfa), Bmp4, Cyr61, Nupr1, Plod 2, P4ha1, Adamts1, Lgals3, and Angpt2. These gene expression changes indicate that the E2Fs control the expression of genes critical to angiogenesis, the remodeling of the extracellular matrix, tumor cell survival, and tumor cell interactions with vascular endothelial cells that facilitate metastasis to the lungs. Taken together, these results reveal that the E2F transcription factors play key roles in mediating tumor development and metastasis in addition to their well-characterized roles in cell cycle control.


Subject(s)
E2F Transcription Factors/physiology , Mammary Neoplasms, Experimental/etiology , Animals , Antigens, Polyomavirus Transforming , E2F Transcription Factors/deficiency , E2F Transcription Factors/genetics , E2F1 Transcription Factor/deficiency , E2F1 Transcription Factor/genetics , E2F1 Transcription Factor/physiology , E2F2 Transcription Factor/deficiency , E2F2 Transcription Factor/genetics , E2F2 Transcription Factor/physiology , E2F3 Transcription Factor/deficiency , E2F3 Transcription Factor/genetics , E2F3 Transcription Factor/physiology , Female , Humans , Lung Neoplasms/genetics , Lung Neoplasms/pathology , Lung Neoplasms/secondary , Mammary Neoplasms, Experimental/pathology , Mammary Neoplasms, Experimental/secondary , Mammary Tumor Virus, Mouse , Mice , Mice, Knockout , Neoplastic Cells, Circulating/pathology , Neovascularization, Pathologic/genetics , Retroviridae Infections/etiology , Retroviridae Infections/pathology , Signal Transduction , Tumor Microenvironment , Tumor Virus Infections/etiology , Tumor Virus Infections/pathology
19.
Biochim Biophys Acta ; 1839(9): 800-12, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24863159

ABSTRACT

Drosophila lin52 (dlin52) is a member of Myb transcription regulator complex and it shows a dynamic pattern of expression in all Drosophila tissues. Myb complex functions to activate or repress transcription in a site-specific manner; however, the detailed mechanism is yet to be clearly understood. Members of the Drosophila melanogaster Myb-MuvB/dREAM complex have been known to regulate expression of a wide range of genes including those involved in regulating apoptosis. E2F and its corepressor RBF also belong to this complex and together they regulate expression of genes involved in cell cycle progression, apoptosis, differentiation, and development. In the present study, we examined whether the depletion of dlin52 in developing photoreceptor neurons results in enhanced apoptosis and disorganisation of the ommatidia. Strikingly, we found that dLin52 is essential for transcriptional repression of the pro-apoptotic gene, hid; decrease in dlin52 levels led to dramatic induction of hid and apoptosis in eye-antennal discs. Reduction of Rpd3 (HDAC1), another member of the dREAM complex, also led to marginal upregulation of Hid. In addition, we also demonstrated that an optimum level of dLin52 is needed for dE2F1/2 activity on the hid promoter. dlin52 cooperates with dRBF and dE2F1/2 for recruitment of repressor complex on the hid promoter. Preliminary data indicate that Rpd3/HDAC1 also contributes to hid repression. Based on the findings, we conclude that dLin52 functions as a co-factor and modulates activity of members of dMyb/dREAM complex at hid promoter, thus regulating apoptosis by repressing this pro-apoptotic gene in the developing Drosophila eye.


Subject(s)
Carrier Proteins/physiology , Drosophila Proteins/genetics , Drosophila Proteins/physiology , Drosophila melanogaster/physiology , E2F Transcription Factors/physiology , Neuropeptides/genetics , Retinoblastoma Protein/physiology , Transcription Factors/physiology , Transcription, Genetic , Animals , Apoptosis , Promoter Regions, Genetic
20.
Proc Natl Acad Sci U S A ; 111(14): 5248-53, 2014 Apr 08.
Article in English | MEDLINE | ID: mdl-24706837

ABSTRACT

Glioblastoma is the most common adult primary brain tumor and has a dismal median survival. Radiation is a mainstay of treatment and significantly improves survival, yet recurrence is nearly inevitable. Better understanding the radiation response of glioblastoma will help improve strategies to treat this devastating disease. Here, we present a comprehensive study of the in vivo radiation response of glioma cells in a mouse model of proneural glioblastoma. These tumors are a heterogeneous mix of cell types with differing radiation sensitivities. To explicitly study the gene expression changes comprising the radiation response of the Olig2(+) tumor bulk cells, we used translating ribosome affinity purification (TRAP) from Olig2-TRAP transgenic mice. Comparing both ribosome-associated and total pools of mRNA isolated from Olig2(+) cells indicated that the in vivo gene expression response to radiation occurs primarily at the total transcript level. Genes related to apoptosis and cell growth were significantly altered. p53 and E2F were implicated as major regulators of the radiation response, with p53 activity needed for the largest gene expression changes after radiation. Additionally, radiation induced a marked shift away from a proneural expression pattern toward a mesenchymal one. This shift occurs in Olig2(+) cells within hours and in multiple genetic backgrounds. Targets for Stat3 and CEBPB, which have been suggested to be master regulators of a mesenchymal shift, were also up-regulated by radiation. These data provide a systematic description of the events following radiation and may be of use in identifying biological processes that promote glioma radioresistance.


Subject(s)
Brain Neoplasms/pathology , Glioblastoma/pathology , Mesoderm/metabolism , Neurons/metabolism , Radiation Tolerance/genetics , Transcription, Genetic , Tumor Suppressor Protein p53/physiology , Animals , Animals, Newborn , E2F Transcription Factors/physiology , Mesoderm/cytology , Mice , Mice, Transgenic , Neurons/cytology , RNA, Messenger/genetics , Tumor Suppressor Protein p53/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...