Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
Add more filters










Publication year range
1.
Biology (Basel) ; 12(12)2023 Dec 11.
Article in English | MEDLINE | ID: mdl-38132337

ABSTRACT

The transcription factor E2F links the RB pathway to the p53 pathway upon loss of function of pRB, thereby playing a pivotal role in the suppression of tumorigenesis. E2F fulfills a major role in cell proliferation by controlling a variety of growth-associated genes. The activity of E2F is controlled by the tumor suppressor pRB, which binds to E2F and actively suppresses target gene expression, thereby restraining cell proliferation. Signaling pathways originating from growth stimulative and growth suppressive signals converge on pRB (the RB pathway) to regulate E2F activity. In most cancers, the function of pRB is compromised by oncogenic mutations, and E2F activity is enhanced, thereby facilitating cell proliferation to promote tumorigenesis. Upon such events, E2F activates the Arf tumor suppressor gene, leading to activation of the tumor suppressor p53 to protect cells from tumorigenesis. ARF inactivates MDM2, which facilitates degradation of p53 through proteasome by ubiquitination (the p53 pathway). P53 suppresses tumorigenesis by inducing cellular senescence or apoptosis. Hence, in almost all cancers, the p53 pathway is also disabled. Here we will introduce the canonical functions of the RB-E2F-p53 pathway first and then the non-classical functions of each component, which may be relevant to cancer biology.

2.
Biochem Biophys Res Commun ; 663: 154-162, 2023 06 30.
Article in English | MEDLINE | ID: mdl-37141667

ABSTRACT

The TFDP1 gene codes for the heterodimeric partner DP1 of the transcription factor E2F. E2F, principal target of the tumor suppressor pRB, plays central roles in cell proliferation by activating a group of growth-related genes. E2F also mediates tumor suppression by activating tumor suppressor genes such as ARF, an upstream activator of the tumor suppressor p53, when deregulated from pRB upon oncogenic changes. Among 8 E2F family members (E2F1∼E2F8), expression of activator E2Fs (E2F1∼E2F3a) is induced at the G1/S boundary of the cell cycle after growth stimulation by E2F itself. However, mechanisms regulating DP1 expression are not known. We show here that over-expression of E2F1 and forced inactivation of pRB, by adenovirus E1a, induced TFDP1 gene expression in human normal fibroblast HFFs, suggesting that the TFDP1 gene is a target of E2F. Serum stimulation of HFFs also induced TFDP1 gene expression, but with different kinetics from that of the CDC6 gene, a typical growth-related E2F target. Both over-expression of E2F1 and serum stimulation activated the TFDP1 promoter. We searched for E2F1-responsive regions by 5' and 3' deletion of the TFDP1 promoter and by introducing point mutations in putative E2F1-responsive elements. Promoter analysis identified several GC-rich elements, mutation of which reduced E2F1-responsiveness but not serum-responsiveness. ChIP assays showed that the GC-rich elements bound deregulated E2F1 but not physiological E2F1 induced by serum stimulation. These results suggest that the TFDP1 gene is a target of deregulated E2F. In addition, knockdown of DP1 expression by shRNA enhanced ARF gene expression, which is specifically induced by deregulated E2F activity, suggesting that activation of the TFDP1 gene by deregulated E2F may function as a failsafe feedback mechanism to suppress deregulated E2F and maintain normal cell growth in the event that DP1 expression is insufficient relative to that of its partner activator E2Fs. a maximum of 6 keywords: E2F, DP1, TFDP1 gene, pRB, gene expression.


Subject(s)
E2F1 Transcription Factor , Gene Expression Regulation , Humans , E2F Transcription Factors/genetics , E2F Transcription Factors/metabolism , E2F1 Transcription Factor/genetics , E2F1 Transcription Factor/metabolism , Genes, Tumor Suppressor , Cell Cycle Proteins/metabolism , E2F3 Transcription Factor/metabolism , Transcription Factor DP1/genetics , Transcription Factor DP1/metabolism
3.
Genes (Basel) ; 14(2)2023 02 02.
Article in English | MEDLINE | ID: mdl-36833320

ABSTRACT

The transcription factor E2F, the principal target of the tumor suppressor pRB, plays crucial roles in cell proliferation and tumor suppression. In almost all cancers, pRB function is disabled, and E2F activity is enhanced. To specifically target cancer cells, trials have been undertaken to suppress enhanced E2F activity to restrain cell proliferation or selectively kill cancer cells, utilizing enhanced E2F activity. However, these approaches may also impact normal growing cells, since growth stimulation also inactivates pRB and enhances E2F activity. E2F activated upon the loss of pRB control (deregulated E2F) activates tumor suppressor genes, which are not activated by E2F induced by growth stimulation, inducing cellular senescence or apoptosis to protect cells from tumorigenesis. Deregulated E2F activity is tolerated in cancer cells due to inactivation of the ARF-p53 pathway, thus representing a feature unique to cancer cells. Deregulated E2F activity, which activates tumor suppressor genes, is distinct from enhanced E2F activity, which activates growth-related genes, in that deregulated E2F activity does not depend on the heterodimeric partner DP. Indeed, the ARF promoter, which is specifically activated by deregulated E2F, showed higher cancer-cell specific activity, compared to the E2F1 promoter, which is also activated by E2F induced by growth stimulation. Thus, deregulated E2F activity is an attractive potential therapeutic tool to specifically target cancer cells.


Subject(s)
E2F1 Transcription Factor , Neoplasms , E2F1 Transcription Factor/genetics , E2F1 Transcription Factor/metabolism , E2F Transcription Factors/metabolism , Genes, Tumor Suppressor , Apoptosis , Promoter Regions, Genetic , Neoplasms/genetics
4.
Commun Biol ; 2: 3, 2019.
Article in English | MEDLINE | ID: mdl-30740539

ABSTRACT

Mitochondrial damage is caused by changes in the micro-environmental conditions during tumor progression. Cancer cells require mechanisms for mitochondrial quality control during this process; however, how mitochondrial integrity is maintained is unclear. Here we show that E2F3d, a previously unidentified E2F3 isoform, mediates hypoxia-induced mitophagy in cancer cells. Aberrant activity and expression of the E2F3 transcription factor is frequently observed in many cancer cells. Loss of retinoblastoma (Rb) protein family function increases the expression of E2F3d and E2F3a. E2F3d localizes to the outer mitochondrial membrane and its cytosolic domain contains an LC3-interacting region motif. Overexpression of E2F3d induces mitochondrial fragmentation and mitophagy, suggesting that E2F3d plays an important role in mitophagy. Furthermore, depletion of E2F3s attenuates hypoxia-induced mitophagy and increases intracellular levels of reactive oxygen species, which is reversed by the reintroduction of E2F3d. This study presents another key player that regulates mitochondrial quality control in cancer cells.


Subject(s)
Cell Hypoxia , E2F3 Transcription Factor/metabolism , Mitochondria/metabolism , Mitochondrial Proteins/metabolism , Mitophagy , Neoplasms/metabolism , Dynamins/genetics , E2F3 Transcription Factor/genetics , Gene Knockdown Techniques , HEK293 Cells , HeLa Cells , Humans , Microtubule-Associated Proteins/metabolism , Mitochondrial Membranes/metabolism , Protein Isoforms/metabolism , Reactive Oxygen Species/metabolism
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.
Biomed Res Int ; 2017: 5158961, 2017.
Article in English | MEDLINE | ID: mdl-28191463

ABSTRACT

The physical properties of the extracellular matrix (ECM), such as stiffness, are involved in the determination of the characteristics of cancer cells, including chemotherapy sensitivity. Resistance to chemotherapy is often linked to dysfunction of tumor suppressor p53; however, it remains elusive whether the ECM microenvironment interferes with p53 activation in cancer cells. Here, we show that, in MCF-7 breast cancer cells, extracellular stiffness influences p53 activation induced by the antitumor drug doxorubicin. Cell growth inhibition by doxorubicin was increased in response to ECM rigidity in a p53-dependent manner. The expression of Rho-associated coiled coil-containing protein kinase (ROCK) 2, which induces the activation of myosin II, was significantly higher when cells were cultured on stiffer ECM substrates. Knockdown of ROCK2 expression or pharmacological inhibition of ROCK decreased doxorubicin-induced p53 activation. Our results suggest that a soft ECM causes downregulation of ROCK2 expression, which drives resistance to chemotherapy by repressing p53 activation.


Subject(s)
Doxorubicin/pharmacology , Elasticity , Extracellular Matrix/chemistry , Tumor Suppressor Protein p53/metabolism , rho-Associated Kinases/metabolism , Biomechanical Phenomena/drug effects , Cell Proliferation/drug effects , Extracellular Matrix/drug effects , Humans , MCF-7 Cells
7.
Biochem Biophys Res Commun ; 483(1): 107-114, 2017 01 29.
Article in English | MEDLINE | ID: mdl-28042030

ABSTRACT

In cancer treatment, specifically targeting cancer cells is important for optimal therapeutic efficacy. One strategy is to utilize a cancer specific promoter to express a cytotoxic gene or a viral gene required for replication. In this approach, the therapeutic window is dependent on the relative promoter activity in cancer cells versus normal cells. Therefore, a promoter with optimal cancer cell-specificity should be used. The tumor suppressor ARF promoter, which specifically responds to deregulated E2F activity, is a potent candidate. Defects in the RB pathway resulting in deregulated E2F activity are observed in almost all cancers. Furthermore, the ARF promoter exhibits greater cancer cell specificity than the E2F1 promoter and consequently, adenovirus expressing HSV-TK under the control of the ARF promoter (Ad-ARF-TK) has more selective cytotoxicity in cancer cells than the analogous E2F1 construct. Ideally, cancer specific gene expression driven by the ARF promoter could be enhanced for optimal therapeutic efficacy, with minimal side effects. We show here that ectopic expression of the CDK inhibitor p21Cip1 enhanced deregulated E2F activity and pro-apoptotic E2F target gene expression in cancer cells. Moreover, ectopic expression of p21Cip1 augmented cancer specific cytotoxicity of Ad-ARF-TK, and apoptosis induced by p21Cip1 was dependent on deregulated E2F activity. These results suggest that p21Cip1 specifically enhances deregulated E2F activity and that a combination of the CDK inhibitor with Ad-ARF-TK could be effectively employed for cancer therapy.


Subject(s)
Cyclin-Dependent Kinase Inhibitor p21/genetics , Cyclin-Dependent Kinase Inhibitor p21/metabolism , E2F Transcription Factors/genetics , E2F Transcription Factors/metabolism , Genes, p16 , Apoptosis , Cell Line, Tumor , Cyclin-Dependent Kinase Inhibitor p16 , Cyclin-Dependent Kinase Inhibitor p18/genetics , Cyclin-Dependent Kinase Inhibitor p18/metabolism , E2F1 Transcription Factor/genetics , E2F1 Transcription Factor/metabolism , Ectopic Gene Expression , Gene Expression Regulation, Neoplastic , Humans , Neoplasms/genetics , Neoplasms/metabolism , Neoplasms/pathology , Promoter Regions, Genetic
8.
Biochem Biophys Res Commun ; 482(4): 784-790, 2017 Jan 22.
Article in English | MEDLINE | ID: mdl-27888102

ABSTRACT

The transcription factor E2F plays crucial roles in tumor suppression by activating pro-apoptotic genes such as the tumor suppressor ARF. The regulation of the ARF gene is distinct from that of growth-related E2F targets, in that it is specifically activated by deregulated E2F activity, induced by over-expression of E2F or forced inactivation of pRB, but not by physiological E2F activity induced by growth stimulation. The phosphatidyl inositol 3 kinase (PI3K) pathway was reported to suppress expression of some atypical pro-apoptotic genes by over-expressed E2F1. However, the effects of the PI3K pathway on the distinct regulation of typical pro-apoptotic E2F targets have not been elucidated. We examined whether the PI3K pathway suppressed activation of the typical pro-apoptotic E2F targets ARF and BIM. Activation of the PI3K pathway by growth stimulation or introduction of a constitutively active Akt/PKB did not reduce induction of ARF or BIM gene expression or activation of their promoters by over-expressed E2F1. These results suggest that the PI3K pathway does not suppress induction of typical pro-apoptotic genes that are selectively activated by deregulated E2F1.


Subject(s)
Bcl-2-Like Protein 11/metabolism , Cyclin-Dependent Kinase Inhibitor p18/metabolism , E2F1 Transcription Factor/metabolism , Gene Expression Regulation , Phosphatidylinositol 3-Kinases/metabolism , Apoptosis , Cyclin-Dependent Kinase Inhibitor p16 , Fibroblasts/metabolism , Humans , Retinoblastoma Protein/metabolism , Signal Transduction
9.
J Mol Biol ; 428(24 Pt B): 4993-5006, 2016 12 04.
Article in English | MEDLINE | ID: mdl-27825926

ABSTRACT

The heterodimeric transcription factor E2F1-DP1 plays crucial roles in coordinating gene expression during G1/S cell cycle progression. For transcriptional activation, the transactivation domain (TAD) of E2F1 is known to interact with the TATA-binding protein of TFIID and the p62 subunit of TFIIH. It is generally believed that DP1 facilitates E2F1 binding to target DNA and does not possess a TAD. Here, we show that an acidic region of DP1, whose function has remained elusive, binds to the plekstrin homology (PH) domain of p62 with higher affinity than that of E2F1 and contributes to transcriptional activation. The structure of the complex revealed that DP1 forms a twisted U-shaped, string-like conformation and binds to the surface of the PH domain by anchoring Phe403 into a pocket in the PH domain. The transcriptional activity of E2F1-DP1 was reduced when Phe403 of DP1 was mutated. These findings indicate that the acidic region of DP1 acts as a TAD by contacting TFIIH.


Subject(s)
Protein Interaction Mapping , Transcription Factor DP1/chemistry , Transcription Factor DP1/metabolism , Transcription Factor TFIIH/chemistry , Transcription Factor TFIIH/metabolism , Cell Cycle , Crystallography, X-Ray , Gene Expression Regulation , Humans , Models, Molecular , Protein Binding , Protein Conformation
10.
Biol Open ; 5(4): 499-506, 2016 Apr 15.
Article in English | MEDLINE | ID: mdl-27029899

ABSTRACT

Cell migration is a highly dynamic process that plays pivotal roles in both physiological and pathological processes. We have previously reported that p130Cas supports cell migration through the binding to Src as well as phosphorylation-dependent association with actin retrograde flow at focal adhesions. However, it remains elusive how phosphorylated Cas interacts with actin cytoskeletons. We observe that the actin-binding protein, tensin 1, co-localizes with Cas, but not with its phosphorylation-defective mutant, at focal adhesions in leading regions of migrating cells. While a truncation mutant of tensin 1 that lacks the phosphotyrosine-binding PTB and SH2 domains (tensin 1-SH2PTB) poorly co-localizes or co-immunoprecitates with Cas, bacterially expressed recombinant tensin 1-SH2PTB protein binds to Casin vitroin a Cas phosphorylation-dependent manner. Furthermore, exogenous expression of tensin 1-SH2PTB, which is devoid of the actin-interacting motifs, interferes with the Cas-driven cell migration, slows down the inward flux of Cas molecules, and impedes the displacement of Cas molecules from focal adhesions. Taken together, our results show that tensin 1 links inwardly moving actin cytoskeletons to phosphorylated Cas at focal adhesions, thereby driving cell migration.

11.
Cell Mol Life Sci ; 72(21): 4077-94, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26206378

ABSTRACT

Cancer cells possess unique characteristics such as invasiveness, the ability to undergo epithelial-mesenchymal transition, and an inherent stemness. Cell morphology is altered during these processes and this is highly dependent on actin cytoskeleton remodeling. Regulation of the actin cytoskeleton is, therefore, important for determination of cell fate. Mutations within the TP53 (tumor suppressor p53) gene leading to loss or gain of function (GOF) of the protein are often observed in aggressive cancer cells. Here, we highlight the roles of p53 and its GOF mutants in cancer cell invasion from the perspective of the actin cytoskeleton; in particular its reorganization and regulation by cell adhesion molecules such as integrins and cadherins. We emphasize the multiple functions of p53 in the regulation of actin cytoskeleton remodeling in response to the extracellular microenvironment, and oncogene activation. Such an approach provides a new perspective in the consideration of novel targets for anti-cancer therapy.


Subject(s)
Cytoskeleton/metabolism , Neoplasms/pathology , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism , Actin Cytoskeleton/metabolism , Actin Cytoskeleton/pathology , Animals , Cadherins/metabolism , Cell Adhesion Molecules/metabolism , Cell Movement , Humans , Integrins/metabolism , Mutation , Neoplasms/genetics , Neoplasms/metabolism , rho GTP-Binding Proteins/metabolism
12.
Elife ; 2: e01228, 2013 Dec 03.
Article in English | MEDLINE | ID: mdl-24302570

ABSTRACT

Skeletal muscle degeneration is a complication arising from a variety of chronic diseases including advanced cancer. Pro-inflammatory cytokine TNF-α plays a pivotal role in mediating cancer-related skeletal muscle degeneration. Here, we show a novel function for retinoblastoma protein (Rb), where Rb causes sarcomeric disorganization. In human skeletal muscle myotubes (HSMMs), up-regulation of cyclin-dependent kinase 4 (CDK4) and concomitant phosphorylation of Rb was induced by TNF-α treatment, resulting in the translocation of phosphorylated Rb to the cytoplasm. Moreover, induced expression of the nuclear exporting signal (NES)-fused form of Rb caused disruption of sarcomeric organization. We identified mammalian diaphanous-related formin 1 (mDia1), a potent actin nucleation factor, as a binding partner of cytoplasmic Rb and found that mDia1 helps maintain the structural integrity of the sarcomere. These results reveal a novel non-nuclear function for Rb and suggest a potential mechanism of TNF-α-induced disruption of sarcomeric organization. DOI: http://dx.doi.org/10.7554/eLife.01228.001.


Subject(s)
Cytoplasm/metabolism , Retinoblastoma Protein/metabolism , Sarcomeres , Humans , Muscle, Skeletal/metabolism , Phosphorylation , Protein Transport , Tumor Necrosis Factor-alpha/pharmacology
13.
Biochem J ; 445(3): 323-32, 2012 Aug 01.
Article in English | MEDLINE | ID: mdl-22587391

ABSTRACT

Actin dynamics are implicated in various cellular processes, not only through the regulation of cytoskeletal organization, but also via the control of gene expression. In the present study we show that the Src family kinase substrate p130Cas (Cas is Crk-associated substrate) influences actin remodelling and concomitant muscle-specific gene expression, thereby regulating myogenic differentiation. In C2C12 myoblasts, silencing of p130Cas expression by RNA interference impaired F-actin (filamentous actin) formation and nuclear localization of the SRF (serum-response factor) co-activator MAL (megakaryocytic acute leukaemia) following the induction of myogenic differentiation. Consequently, formation of multinucleated myotubes was abolished. Re-introduction of wild-type p130Cas, but not its phosphorylation-defective mutant, into p130Cas-knockdown myoblasts restored F-actin assembly, MAL nuclear localization and myotube formation. Depletion of the adhesion molecule integrin ß3, a key regulator of myogenic differentiation as well as actin cytoskeletal organization, attenuated p130Cas phosphorylation and MAL nuclear localization during C2C12 differentiation. Moreover, knockdown of p130Cas led to the activation of the F-actin-severing protein cofilin. The introduction of a dominant-negative mutant of cofilin into p130Cas-knockdown myoblasts restored muscle-specific gene expression and myotube formation. The results of the present study suggest that p130Cas phosphorylation, mediated by integrin ß3, facilitates cofilin inactivation and promotes myogenic differentiation through modulating actin cytoskeleton remodelling.


Subject(s)
Actins/metabolism , Crk-Associated Substrate Protein/metabolism , Muscle Development/physiology , Myoblasts, Skeletal/cytology , Myoblasts, Skeletal/metabolism , Actin Cytoskeleton/metabolism , Animals , Base Sequence , Cell Differentiation/genetics , Cell Differentiation/physiology , Cell Line , Cell Nucleus/metabolism , Cofilin 2/antagonists & inhibitors , Cofilin 2/genetics , Cofilin 2/metabolism , Crk-Associated Substrate Protein/antagonists & inhibitors , Crk-Associated Substrate Protein/genetics , DNA Primers/genetics , Gene Expression Regulation, Developmental , Gene Knockdown Techniques , Integrin beta3/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Mice , Models, Biological , Muscle Development/genetics , Mutagenesis , Phosphorylation , Protein Serine-Threonine Kinases/antagonists & inhibitors , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , RNA Interference
14.
Proc Natl Acad Sci U S A ; 106(9): 3431-6, 2009 Mar 03.
Article in English | MEDLINE | ID: mdl-19202066

ABSTRACT

The IkappaB kinase (IKK)-NF-kappaB pathway plays a critical role in oncogenesis. Recently, we have shown that p53 regulates glucose metabolism through the IKK-NF-kappaB pathway and that, in the absence of p53, the positive feedback loop between IKK-NF-kappaB and glycolysis has an integral role in oncogene-induced cell transformation. Here, we demonstrate that IKKbeta, a component of the IKK complex, was constitutively modified with O-linked beta-N-acetyl glucosamine (O-GlcNAc) in both p53-deficient mouse embryonic fibroblasts (MEFs) and transformed human fibroblasts. In p53-deficient cells, the O-GlcNAcylated IKKbeta and the activating phosphorylation of IKK were decreased by p65/NF-kappaB knockdown or glucose depletion. We also found that high glucose induced the O-GlcNAcylation of IKKbeta and sustained the TNFalpha-dependent IKKbeta activity. Moreover, the O-GlcNAcase inhibitor streptozotocin intensified O-GlcNAcylation and concomitant activating phosphorylation of IKKbeta. Mutational analysis revealed that O-GlcNAcylation of IKKbeta occurred at Ser 733 in the C-terminal domain, which was identified as an inactivating phosphorylation site, suggesting that IKKbeta O-GlcNAcylation regulates its catalytic activity. Taken together, we propose a novel mechanism for the enhancement of NF-kappaB activity by loss of p53, which evokes positive feedback regulation from enhanced glucose metabolism to IKK in oncogenesis.


Subject(s)
Acetylglucosamine/metabolism , Biocatalysis , I-kappa B Kinase/metabolism , Tumor Suppressor Protein p53/deficiency , Tumor Suppressor Protein p53/metabolism , Acetylation/drug effects , Animals , Cell Line , Glucose/pharmacology , Glycolysis , Humans , I-kappa B Kinase/genetics , Mice , Mice, Knockout , Tumor Suppressor Protein p53/genetics
15.
Biochem Biophys Res Commun ; 372(1): 137-41, 2008 Jul 18.
Article in English | MEDLINE | ID: mdl-18477470

ABSTRACT

NF-kappaB plays an important role in oncogenesis. Recently, we have demonstrated that loss of p53 function enhances DNA binding and transcriptional activities of NF-kappaB via IKKalpha and IKKbeta, and that glycolysis, activated by NF-kappaB, has an integral role in oncogene-induced cell transformation. Here, we show that ectopically expressed p53 induces acetylation and phosphorylation at Ser 536 of p65, an NF-kappaB component, and enhances DNA-binding activity of NF-kappaB. However, activated p53 suppresses transcriptional activity of NF-kappaB. Under non-stimulating conditions, p65 formed a complex with IKKalpha and IKKbeta. Activated p53 bound to p65 on DNA and disrupted binding of p65 to IKKbeta. Moreover, histone H3 kinase activity, which requires transcriptional activation of NF-kappaB, was diminished by p53. Thus, activated p53 may suppress transcriptional activity of NF-kappaB through inhibition of IKK and histone H3 kinase on DNA, suggesting a novel p53-mediated suppression system for tumorigenesis.


Subject(s)
DNA/metabolism , Transcription Factor RelA/antagonists & inhibitors , Transcription Factor RelA/metabolism , Transcriptional Activation , Tumor Suppressor Protein p53/metabolism , Acetylation , Animals , Humans , I-kappa B Kinase/antagonists & inhibitors , I-kappa B Kinase/metabolism , Mice , Phosphorylation , Protamine Kinase/antagonists & inhibitors , Protamine Kinase/metabolism , Protein Binding , Serine/metabolism
16.
Nat Cell Biol ; 10(5): 611-8, 2008 May.
Article in English | MEDLINE | ID: mdl-18391940

ABSTRACT

Cancer cells use aerobic glycolysis preferentially for energy provision and this metabolic change is important for tumour growth. Here, we have found a link between the tumour suppressor p53, the transcription factor NF-kappaB and glycolysis. In p53-deficient primary cultured cells, kinase activities of IKKalpha and IKKbeta and subsequent NF-kappaB activity were enhanced. Activation of NF-kappaB, by loss of p53, caused an increase in the rate of aerobic glycolysis and upregulation of Glut3. Oncogenic Ras-induced cell transformation and acceleration of aerobic glycolysis in p53-deficient cells were suppressed in the absence of p65/NF-kappaB expression, and were restored by GLUT3 expression. It was also shown that a glycolytic inhibitor diminished the enhanced IKK activity in p53-deficient cells. Moreover, in Ras-expressing p53-deficient cells, IKK activity was suppressed by p65 deficiency and restored by GLUT3 expression. Taken together, these data indicate that p53 restricts activation of the IKK-NF-kappaB pathway through suppression of glycolysis. These results suggest that a positive-feedback loop exists, whereby glycolysis drives IKK-NF-kappaB activation, and that hyperactivation of this loop by loss of p53 is important in oncogene-induced cell transformation.


Subject(s)
Cell Transformation, Neoplastic , Glucose/metabolism , I-kappa B Kinase/metabolism , Transcription Factor RelA/metabolism , Tumor Suppressor Protein p53/metabolism , Animals , Cells, Cultured , Fibroblasts/cytology , Fibroblasts/physiology , Genes, ras , Glucose Transporter Type 3/genetics , Glucose Transporter Type 3/metabolism , Glycolysis/physiology , I-kappa B Kinase/genetics , Lactic Acid/metabolism , Mice , Mice, Knockout , RNA Interference , Signal Transduction/physiology , Transcription Factor RelA/genetics , Tumor Suppressor Protein p53/genetics , ras Proteins/genetics , ras Proteins/metabolism
17.
Oncogene ; 22(48): 7632-41, 2003 Oct 23.
Article in English | MEDLINE | ID: mdl-14576826

ABSTRACT

The activity of E2F transcription factors plays a crucial role in mammalian cell-cycle progression and is controlled by physical association with the pocket proteins (pRb and its related p107 and p130). The E2F1 promoter, which contains two overlapping E2F-binding sites, is activated at the G1/S transition in an E2F-dependent manner. Mutational experiments have shown that the distal E2F-binding site on the E2F1 promoter is required for transcriptional repression in the G0 phase, whereas the proximal E2F-binding site contributes to transcriptional activation at the G1/S boundary. Consistent with these results, chromatin immunoprecipitation assays have revealed that the E2F4/p130 repressor complex specifically binds to the distal E2F-binding site, whereas E2F1 and E2F3 activators preferentially bind to the proximal E2F-binding site. The assays also showed that the specific binding of E2F4/p130 complex to the distal site was dramatically impaired by a mutation introduced into the contiguous repression site (cell Cycle gene Homology Region; CHR). Taken together, these findings indicate that the two E2F-binding sites play distinct roles in the regulation of E2F1 transcription by interacting with different sets of E2F members and cooperating with the contiguous repressor element.


Subject(s)
Cell Cycle Proteins , DNA-Binding Proteins , Gene Expression Regulation , Promoter Regions, Genetic/genetics , Proteins , Response Elements/genetics , Transcription Factors/genetics , Transcription Factors/metabolism , Base Sequence , Binding Sites , Cell Cycle/genetics , Cell Line , E2F Transcription Factors , E2F1 Transcription Factor , E2F3 Transcription Factor , E2F4 Transcription Factor , Electrophoretic Mobility Shift Assay , Humans , Mutation , Phosphoproteins/metabolism , Retinoblastoma-Like Protein p130 , Transcription, Genetic
18.
Mol Cancer Res ; 1(6): 438-44, 2003 Apr.
Article in English | MEDLINE | ID: mdl-12692263

ABSTRACT

E2FBP1/DRIL1 is an AT-rich interaction domain DNA-binding protein and is ubiquitously expressed in various tissues. It has been shown that Bright, the mouse orthologue of E2FBP1/DRIL1, exhibits sequence-specific DNA binding and regulates immunoglobulin transcription. Here we show a novel connection between E2FBP1/DRIL1 and the p53 tumor suppressor, a key regulator of growth arrest or apoptosis in response to cellular stress. We found a putative p53-binding site, which specifically responded to p53, in the second intron of the E2FBP1/DRIL1 gene. E2FBP1/DRIL1was induced by p53 and up-regulated following DNA damage caused by UV radiation or doxorubicin treatment in a manner dependent on endogenous p53. The ectopic expression of E2FBP1/DRIL1 induced growth arrest in U2OS cells expressing normal p53, but not Saos-2 cells lacking p53. These results suggest that E2FBP1/DRIL1 may play a role in growth suppression mediated by p53.


Subject(s)
AT Rich Sequence/genetics , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Oncogenes/genetics , Trans-Activators , Tumor Suppressor Protein p53/metabolism , Base Sequence , Cell Division , Cell Line, Tumor , Cyclin D1/metabolism , DNA Damage , DNA-Binding Proteins/chemistry , Humans , Introns/genetics , Molecular Sequence Data , Protein Binding , Protein Structure, Tertiary , RNA, Messenger/genetics , RNA, Messenger/metabolism , Transcription Factors , Transcription, Genetic/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...